Download as pdf or txt
Download as pdf or txt
You are on page 1of 114

VU Research Portal

High-resolution screening of metabolite-like lead libraries


Falck, D.

2013

document version
Publisher's PDF, also known as Version of record

Link to publication in VU Research Portal

citation for published version (APA)


Falck, D. (2013). High-resolution screening of metabolite-like lead libraries.

General rights
Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright owners
and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights.

• Users may download and print one copy of any publication from the public portal for the purpose of private study or research.
• You may not further distribute the material or use it for any profit-making activity or commercial gain
• You may freely distribute the URL identifying the publication in the public portal ?

Take down policy


If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately
and investigate your claim.

E-mail address:
vuresearchportal.ub@vu.nl

Download date: 25. jun. 2021


VRIJE UNIVERSITEIT

High-resolution screening
of metabolite-like lead libraries

ACADEMISCH PROEFSCHRIFT

ter verkrijging van de graad Doctor aan


de Vrije Universiteit Amsterdam,
op gezag van de rector magnificus
prof.dr. F.A. van der Duyn Schouten,
in het openbaar te verdedigen
ten overstaan van de promotiecommissie
van de Faculteit der Exacte Wetenschappen
op donderdag 14 november 2013 om 13.45 uur
in de aula van de universiteit,
De Boelelaan 1105

door

David Falck
geboren te Haldensleben, Duitsland
Reading comittee: dr. F. Cuyckens
prof.dr. U. Karst
prof. dr. R. Leurs
prof.dr. N. M. M. Nibbering
promotoren: prof.dr. H. Irth dr. J. Wieling
prof.dr. W.M.A. Niessen prof. dr. S. S. Wijmenga
prof.dr. M. Honing
copromotor: dr. J. Kool

The research described in this thesis was performed (partly) within the framework of the
project D2-102 ‘Metabolic stability assessment as new tool in the Hit-to-Lead selection pro-
cess and the generation of new lead compound libraries’ of the Dutch Top Institute Pharma.

Shimadzu Benelux B.V. (’s-Hertogenbosch) and Antec (Zoeterwoude) are thanked for fi-
nancial support for the printing of this thesis. Printing was excecuted by Wöhrmann Print
Services (Zutphen).

Copyright © 2013 David Falck, Amsterdam. All rights reserved. No part of this thesis may
be reproduced in any form or by any means without permission from the author.
Table of Contents Liquid chromatography – biochemical detection (LC–BCD) system
Determination of inhibitor potency
69
71
Metabolite identification using mass spectrometry 71
Buffer and compound solutions 71
Plate reader measurements 71
Microsomal incubations 72
1. Introduction 8 Results and Discussion 72
1.1 General Introduction 9 Development of an LC–BCD system for the detection
1.1.1 Discovery of new active pharmaceutical ingredients 10 of sEH inhibitors 72
a) Screening 10 Characterization of the LC–BCD system 73
b) Metabolite-like lead libraries 10 Application of LC-BCD on the analysis of metabolic mixtures
1.1.2 High-resolution screening 11 of sEHis 75
1.1.3 Modification methods in metabolism studies 15 Conclusion 77
a) Chemical oxidation 16 Supplementary Material 79
b) Electrochemistry 17
c) Biocatalytic approaches 17
d) Photochemistry 18
1.1.4 Structure elucidation in mixtures using mass spectrometry 3. Integration of modification methods and HRS work-flows
and nuclear magnetic resonance spectroscopy 18 for the study of metabolite-like lead libraries 80
a) Mass spectrometry 18 3.1 On-line electrochemistry–bioaffinity screening with parallel
b) Nuclear Magnetic resonance spectroscopy 20 HR-LC–MS for the generation and characterization of
1.2 Scope 29 modified p38α kinase inhibitors 81
Introduction 82
Materials and Methods 83
2. High-resolution screening platforms 34 Chemicals 83
Instrumentation 83
2.1 A practical guide to high-resolution screening 35
Optimization of electrochemical conversion 85
2.1.1 The high-resolution screening platform 36
a) Separation 36
Results and Discussion 85
b) Flow splitting 39 Conclusion 91
c) Bioassay 39 Supplementary Material 93
d) MS detection 42 3.2 Combination of biotransformation by P450 BM3 mutants with
2.1.2 Data analysis 43 on-line post-column bioaffinity and mass spectrometric
a) Differences between HTS and HRS 43 profiling as a novel strategy to diversify and characterize
b) Validation 46 p38α kinase inhibitors 97
2.1.3 Conclusions 48 Introduction 98
2.2 Development of an on-line p38α mitogen-activated protein Materials and Methods 99
kinase binding assay and integration of LC–HR-MS 51 Chemicals 99
Introduction 52 Expression and isolation of P450 BM3 mutants 99
Materials and Methods 53 Selection of the P450 BM3 mutant library 100
Materials 53 Biotransformation of TAK-715 by P450 BM3 mutants
Instrumentation 53 and human liver microsomes 100
Plate Reader Assay 54 HRS analysis of biotransformation products of TAK-715
On-line Assay 54 using the LC-p38α kinase affinity/MS platform 100
Results and Discussion 56 Large-scale production and isolation of biotransformation products
Assay conditions optimization 56 by preparative HPLC 101
Validation of the p38α binding assay 57 Results and Discussion 102
High resolution screening of mixtures 61 Biocatalysed synthesis of lead compounds from TAK-715
Semi-Quantitative affinity measurements 61 by P450 BM3 mutants; comparison to human liver metabolism 102
Applicability and binding modes 62 Parallel on-line bioaffinity assay and mass spectrometry of
TAK-715 biotransformation products produced
Conclusion 63
by P450 BM3 M11 and HLM 103
Supplementary Material 65 Preparative scale biosynthesis of TAK-715
2.3 Development of on-line liquid chromatography– biotransformation products 103
biochemical detection for soluble epoxide hydrolase Structure elucidation of TAK-715 biotransformation products 105
inhibitors in mixtures 67 Estimation of product affinity (IC50) 108
Introduction 68 Conclusion 109
Materials and Methods 69 Supplementary Material 112
Materials 69
3.3 Comparison of (bio-)transformation methods for the generation 5. High temperature liquid chromatography hyphenated with
of metabolite-like compound libraries of p38α MAP kinase ESI–MS and ICP–MS detection for the structural
inhibitors using high-resolution screening 117
3.3.1. Introduction 118 characterization and quantification of halogen containing
3.3.2. Materials and Methods 119 drug metabolites 188
1. Materials 119 Introduction 190
2. Chemical oxidation 119 Experimental 191
3. Photochemical modification 120 5.2.1 Materials 191
4. Microsomal incubations 120 5.2.2 Methods 191
5. HRS analysis 120 Results and Discussion 193
3.3.3 Results 121 5.3.1 Solvent effect on electrospray ionization 193
1. Oxidation with hydrogen peroxide 123 5.3.2 Hyphenation of HTLC to ICP-MS 194
2. Photochemical modification 126 5.3.3 Profiling bromine containing MAPK inhibitor VIII products 195
3.3.4 Discussion 128 5.3.4 Analysis of iodine containing SB-I and conversion products
1. Comparison of compound modification methods 128 195
2. Conversion products as metabolite standards 130 5.3.5 Response comparison of ICP-MS and ESI-MS 199
3.3.5 Conclusion 131 Conclusion 201
Supplementary Material 135 Supplementary Material 202

6. Summary 204
4. Integrated structure elucidation approaches 6.1 Conclusions and Perspectives 205
for high-resolution screening 144 6.2 Nederlandse Samenvatting 213
4.1 Tandem mass spectrometry study of p38α kinase inhibitors
and related substances 145 Appendix 218
Introduction 146 I. List of Publications 219
Materials and Methods 147 II. Curriculum Vitae 221
Chemicals 147 III. Acknowledgements 222
Generation of related substances of p38α kinase inhibitors 147
HRS platform: LC with parallel bioaffinity screening and
high-resolution MS 147
Results 149
Structure elucidation of DMPIP conversion products 150
Structure elucidation of SB203580 conversion products 153
Structure elucidation of BIRB796 conversion products 156
Structure elucidation of TAK-715 conversion products 158
Occurrence of homolytic cleavages 164
Conclusion 164
Supplementary Material 167
4.2 EC–SPE–stripline-NMR analysis of reactive products:
A feasibility study 171
Introduction 172
Materials and Methods 174
Chemicals 174
Electrochemical conversion 174
On-line Solid Phase Extraction 174
Stripline chip, probe and microfluidics 174
Acquisition and processing 175
EC–SPE–stripline-NMR setup 176
Results and Discussion 176
Optimization of EC and SPE steps 176
SPE–stripline-NMR 177
EC–SPE–stripline-NMR 180
Conclusions and Perspectives 181
Supplementary Material 184
Section 1 Chapter 1.1

Introduction General introduction


1.1.1 Discovery of new active pharmaceutical ingredients known as drug metabolism, because APIs are modified.
The metabolites of APIs are independent entities. In designing molecules, medicinal
1 1.1.1.a) Screening chemists often try to avoid metabolism as they fear unwanted side effects and a decreased
therapeutic efficacy [17,18]. However, a metabolite may possibly show better therapeutic
1
The vast majority of medicines work by modulating the activity of a target protein. Therefore,
properties [18]. Even more so, the intrinsic tendency towards increased hydrophilicity is a
the discovery of an active pharmaceutical ingredient (API) involves finding molecules which
real advantage. The molecules used in modern drug discovery often suffer from excessively
interact with the chosen target protein. This process is called screening. Traditionally, this
low hydrophilicity which prevents them from entering the circulation or allows them to enter
would be achieved by administering the molecules to diseased animals (animal models),
the brain [19,20]. Consequently, more polar compounds are more easily absorbed and
a so-called phenotypic screening [1]. The high cost, limited throughput and increasing
more effectively excluded from the brain. An indirect advantage of hydrophilicity is found
awareness of animal welfare, catalyzed by the emergence of biotechnology, have triggered
in the intended and unintended interactions with (target) proteins. Non-polar molecules
the search for alternative in vitro methods [2]. In addition, it is easier to link an in vitro
mostly draw their affinity from van der Waals interactions which are undirected induced-
assay response to a molecular mechanism of action. In in vitro assays, the target protein
dipole interactions. The unspecific nature of those interactions increases the likelihood of
is present either isolated or in individual living cells. The former is often the cheapest and
affinity with multiple proteins which may lead to side effects [19]. In more polar molecules,
simplest approach [3], which, in combination with the implementation of lab automation and
amine, hydroxyl or carboxylic acid groups form interactions like hydrogen bonds, dipole-
robotics, has led to the emergence of high-throughput screening (HTS) [2]. Conceptually,
dipole interactions or ionic interactions, requiring more specific partners in the protein.
HTS is based on the screening of large numbers, sometimes up to millions, of molecules
This may increase the specificity for the target, thus reducing side effects. In this context,
(compound libraries) with the idea that APIs are discovered by pure chance [4]. However,
specificity is defined as the relative affinity for the intended target versus other proteins.
even in HTS, there is a strong trend to reduce the chance element by designing focused
There are actually quite a number of examples where a metabolite ended up replacing the
compound libraries [5]. The technological platform consists of well plates, dispenser robots
original active ingredient due to improved pharmacological properties [18].
and specialized spectrophotometers, called plate readers [4]. With the extensive use of
Thus, the collection of metabolites derived from an active ingredient and a lead library
HTS within pharmaceutical companies, its limitations are becoming increasingly obvious
derived from a hit are quite comparable. The scaffold is randomly (at least before
[6]. A major issue is the quality of the hits (the compounds that show interaction with the
establishing a pharmacophore model) modified by simple reactions yielding distinct but
target protein) [7]. The statistical approach used results in false positives which requires
similar products. One aim of this thesis is, therefore, to investigate lead libraries which
all hits to be re-screened for confirmation [6]. While HTS is unmatched for large libraries of
incorporate the positive bias of metabolism towards selectivity and hydrophilicity. Though
compounds, it is not as useful for efficiently screening small numbers of compounds in the
the chemical space associated with metabolites is not easily accessed by classical
confirmation screening or at later stages like in lead optimization. Additionally, the purity of
synthetic approaches, there are a number of methods which are frequently employed for
the compounds screened can have a significant influence on hit quality [7]. Thus, achieving
the generation of metabolite standards, some of which are discussed later. As our interest
sufficient purity of these libraries as well as maintaining them over years in different
is not limited to actual metabolites, but extends to all products which show typical metabolic
screening campaigns is challenging. Therefore, impurities and degradation products can
modifications, we use the term metabolite-like lead libraries. However, we use this term to
lead to false assignment of active molecules as well. A significant effort has been put into
describe the structural similarity of the molecules to drug metabolites and not (necessarily)
improving, supplementing or even replacing HTS, for example with high-content screening
to actual endogenous metabolites [21].
[4], improved animal models [1] or in silico screening [8]. One of the key technologies in
this thesis is high-resolution screening (HRS) [9,10] which can address some shortcomings
of HTS in terms of flexibility and hit quality [Chapters  1.1.2 and 2.1]. Additionally, HRS 1.1.2 High-resolution screening
is uniquely suited to screen mixtures and thus not limited to the use of pure compound
libraries. Although, the term high-resolution screening (HRS) was originally introduced for on-line
post-column bioassays (see Figure 1A), we use HRS more generally to describe in vitro
approaches which achieve the integration of a separation technique and a biological assay
1.1.1.b) Metabolite-like lead libraries (bioassay) for screening purposes in drug discovery. The key benefit is that integrating
The large compound libraries that are used in the initial screening preferably have a high
separation and biological evaluation enables the HRS platform to handle mixtures of
chemical diversity [4], possibly within a predefined space [1]. After the hit confirmation
compounds and still be able to assess individual biological effects. Chapter 1.1.2 introduces
and hit-to-lead selection [6], new libraries are created around the selected hit scaffold,
important concepts and distinctions, and briefly discusses advantages and disadvantages
the so-called lead libraries. Lead libraries are much smaller and incorporate a much
of different HRS assay variants as well as of HRS over HTS assays. Examples are cited
narrower chemical space than the screening libraries [11]. However, both types of libraries
for the different concepts, preferably showing their use in HRS assays. A comprehensive
traditionally consist of (more or less) pure compounds. An exception is presented by
overview of HRS methods is not attempted. The reader may refer to several recent reviews
some combinatorial chemistry libraries [12] and natural extract libraries [13,14]. The latter
[24,25,10,9,23,26].
originate from a wide variety of sources, including animal venoms [15] and plant extracts
Although bioassays can also be set up to screen a single (or a limited number of)
[16]. Due to their huge chemical complexity, these libraries mostly consist of mixtures of
compound(s) for the interaction with multiple targets [27], we focus on the more common
compounds, even if fractionation of the original sample was included.
approach to develop a screening for a single target (or a small number of targets) [4].
Not only chemists can generate molecules, but also our body itself is a complicated
An important distinction lies in the nature of the biological effect which is probed. This can
assortment of chemical reactions known as metabolism. Amongst others, this includes
be either biological affinity (bioaffinity) or biological activity (bioactivity). Bioaffinity assays
dealing with potentially harmful, foreign substances in our diet, the so-called xenobiotic
measure the binding of an analyte to the target [Chapter 2.2]. Bioactivity assays can be
metabolism. This consists of different enzymatically catalyzed reactions which are intended
further divided into biochemical assays which follow an enzymatic reaction [Chapter 2.3]
to increase the hydrophilicity of the xenobiotics with the ultimate goal of (renal or fecal)
and functional assays which visualize an indirect response to the analyte-target interaction
excretion. The main impact thereof is in pharmacotherapy, where the phenomenon is
[28]. Bioaffinity of an analyte is measured either by the displacement of a tracer molecule
10 11
[Chapter  2.2] or by directly visualizing the affinity of the analyte [29]. Bioaffinity assays
either rely on affinity selection of the analyte using an immobilized protein [29-31], separate
1 free and bound tracer [32] or utilize the difference between free tracer (or analyte) and
tracer-target (or analyte-target) complex with regard to mass [33], spectroscopic properties
1
[Chapter 2.2] [34,35] or Gibbs free energy [36]. Advantages are that the inactive form of
a protein can be studied and no expensive co-factors are needed (see also Chapter 2.1)
[Chapter 2.2]. However, though without affinity there is no activity, binding may potentially
evoke different responses in the protein. Therefore, in bioaffinity assays the direction of
activity remains unknown, for example whether a binder is an agonist or an antagonist.
This is the domain of biochemical and functional assays. Biochemical assays measure a
biocatalytic reaction by detection of a substrate and/or the product(s) and are therefore
mostly applied to the study of enzymes [Chapter  2.3]. The difference in mass [16] or
spectroscopic properties [Chapter 2.3] between the substrate and the product(s) is the key
to this approach. Detection of multiple reaction partners (mainly substrates and products or
reactants in short) usually increases the robustness of the assay, for example by revealing
the influence of unexpected side reactions [37]. While, in principle, biochemical assays can
be used to screen different substrates for the same (detected) product, the more common
approach is to observe the effect of inhibitors, allosteric modulators, or competing substrates
on a particular reaction. Given the use of the same detection technique, biochemical assays
are usually more sensitive than bioaffinity assays, because of the signal amplification by
reiterative, biocatalytical reaction cycles. In proteins, which do not catalyze a reaction, for
example nuclear receptors or G-protein coupled receptors, functional assays are needed
to investigate the nature of the biological effect [28]. These highly complex assays are often
cell-based and mostly measure an effect of some signal cascade, like calcium release [38],
labeled secondary messengers [39] or protein expression [28]. Because several signal
amplification steps can be involved, functional assays are usually even more sensitive
than biochemical assays. An endpoint has to be chosen beforehand, meaning that effects
on the target leading to different signaling endpoints are not detected. However, in many
studies, the effect of the analyte on the endpoint is more interesting than the mere effect on
the target itself. This is both the advantage and the drawback of functional assays, because
any effect of the analyte on the endpoint detected is merely assumed to be caused by
the analyte-target interaction. In this respect, better certainty is achieved, if bioaffinity is
established in addition to the specific bioactivity yielded by the functional assay [28].
An almost uncountable number of assay principles is currently available. Nonetheless, a brief
overview is attempted referring to categories of principles rather than individual methods.
The most desirable situation for bioaffinity assays is the direct measurement of the analyte.
This is especially useful for novel targets with no or little known ligands [40]. However, it
requires a detection technique capable of measuring all envisioned analytes or analyte-
target complexes. Though mass spectrometry (MS) [29] and surface plasmon resonance
(SPR) [31] are well suited to achieve both the necessary selectivity for binders over non-
binders and the broad detection, even their application often requires at least additional
purification steps. For example, it is very difficult to reliably ionize the intact analyte-target
complex in MS which thus often requires prior separation of the unbound analytes [29].
Therefore, a tracer is often used for bioaffinity assays. This is a binder whose bound or
unbound fraction can be easily monitored. Affinity of an analyte induces competition for

Figure 1: Comparison of pre-column and post-column HRS setups. A) shows the specific setup used
in Chapter 2.2 including an on-line post-column HRS bioaffinity assay and parallel HR-MSn detection.
The setup is rather generic for on-line post-column assays and holds true for Chapter 2.3, by simply
changing tracer to substrate. Variations are discussed in detail in Chapter 2.1. B) represents a unified
schematic of pre-column affinity selection assays rather than a specific setup [12,22,23]. In the first
step, protein and analytes are mixed so the binders form protein-analyte complexes. The proteins
can be immobilized or can be free in solution during this step. Then, binders and non-binders are
separated in a selection step, for example by SEC or by washing the immobilized proteins. Afterwards,
the binders are released, trapped on an LC column and finally analyzed by LC–MS.

12 13
the binding site and is thus detected as the signal change caused by reduction or increase based on radioactively labeled tracers where bound and unbound fraction need to be
of the bound or unbound fraction of the tracer, respectively. Focusing on the tracer allows separated because they show no difference in radioactivity [28]. Due to the highly dynamic
1 a more selective detection method [Chapter 2.2] or an increased response [31]. Thus, it
often improves the assay, e.g., through better signal-to-noise ratios (S/N) or less reagent
nature of bioaffinity, washing away the unbound fraction, while retaining all bound tracer,
is not trivial. Additionally, heterogeneous assay are usually much more labor intensive and
1
consumption. In these respects, the detection of a tracer in bioaffinity assays has a similar error prone than their homogeneous counterparts [32]. Consequently, state-of-the-art post-
function to the monitoring of a reactant in biochemical assays. The selection of either the column HRS assays are usually homogeneous assays.
tracer or the reactant(s) is of utmost importance. The tracer only probes the binding sites Assays can also be distinguished on whether the separation step is operated in the same
to which it binds itself, and, as for the reactant, the influence of the analyte might vary flow system as the bioassay. If this is the case, we speak of an on-line or continuous-flow
with different substrates. In pharmacology, for example, this choice might influence the assay [9]. The (virtually) closed system and the retention of a peak shape for the effect signal
translatability of the assay results to the desired efficacy in human. Therefore, a (relevant) result in many advantages. Chapter 2.1 deals extensively with on-line assays. On the other
natural reactant [16] or ligand is often desirable. Spectroscopic methods, like fluorescence end of the spectrum stands the classical HTS assay were each separate step involves
[Chapters  2.2 and 2.3] or fluorescence polarization [35], promise excellent selectivity, liquid handling [4]. However, next to on-line approaches, HRS also utilizes hybrids called
sensitivity and robustness. Unfortunately, natural reactants or ligands rarely show the at-line assays [9]. These are based on post-column high-resolution micro-fractionation.
necessary optical properties or at least not at a wavelength allowing sufficient selectivity After separation, the effluent is mixed with the bioassay reagents and the mixture is then
[16]. Therefore, synthetic substrates or tracers are often used which are either labeled micro-fractionated into 96-, 384- or 1536-well plates [28,41,42]. Incubation and readout of
versions of a natural reactant/ligand [28] or de novo designed compounds with some label- the well plates are conducted like in HTS. If the fractionation is conducted at sufficiently high
like features [Chapter 2.2 and 2.3]. The label obviously depends mainly on the envisioned resolution, the chromatographic peak shape of the effect signal is retained. The greatest
detection method, but it is important to establish that the target structure and the binding advantage of the at-line approach is that long incubation times can be achieved with limited
mode are as close as possible to the natural state which the assay is meant to probe. For compromise on the chromatographic resolution in the effect signal. This advantage is
some labels, like radioactive 3H or 14C labeling of natural substrates/ligands, this is the case accompanied by the drawbacks of an open system and a major increase in time and effort.
almost by definition [28], while for others, like fluorescence labels, which can reach the size However, in cell-based assays, the at-line HRS approach may be preferred, because of the
of proteins (for example green fluorescence protein), this has to be carefully investigated long incubation times or because of the need to use adhesive cells. To avoid the physical
during assay development. Unfortunately, radioactive labeling is very costly, due to the stress on the cells or to enable solvent evaporation, the mixing is sometimes postponed
necessary safety, synthesis and waste disposal efforts. to after the fractionation. However, in this way, the efficient mixing of the on-line system
Perhaps, the most important distinction in HRS from a technical perspective is between pre- is lost and solvent evaporation is also not without risks. For example, we developed an
column and post-column assays, that is, in whether the biological effect is probed before HRS platform where parallel MS detection was coupled at-line via microfractionation to
or after the separation step (see Figure 1). Pre-column assays rely entirely on bioaffinity three parallel bioassays; a functional assay for the human histamine H4 receptor and two
measurements [23], while post-column assays are also open to biochemical [Chapter 2.3] radioligand binding assays which assessed selectivity of the H4 ligands over histamine H3
and even functional readouts [28]. In pre-column assays, the first step is an affinity selection receptor binding [28,43].
where the mixture of analytes is allowed to interact with the target which can be soluble or MS has become an integral part of state-of-the-art HRS platforms [9]. Next to exploiting the
immobilized [23]. The bound fraction of analytes is then selected and, subsequently, either excellent flexibility of MS between general and specific detection for the effect detection in
directly detected, for example with SPR [31], or released from the target, for example for bioassays, MS is used as a means of implementing rapid structure elucidation into HRS
MS [30] or liquid chromatography–MS (LC–MS) detection [12]. Because bioaffinity is not platforms [Chapter 4.1]. It additionally allows the detection of analytes without biological
assessed on individual compounds, but from the mixture, an excess of one high-affinity effect, though this ability is limited to certain HRS setups [Chapter 2.2]. For example, the
analyte compared to the target concentration prevents lower-affinity analytes from being necessary selection step between bound and unbound analytes in pre-column assays
detected if they compete for the same binding site. Affinity selection is not biased towards prevents detection of analytes without bioaffinity [30]. Though this can be advantageous
a certain binding mode or region. On the one hand, this can lead to issues with nonspecific with MS based bioassay readouts, because it significantly reduces the complexity of the
binding. On the other hand, compounds using new (allosteric) binding sites can be mixture prior to analysis, it limits the usefulness of these assays for mixtures of unknown
detected without prior knowledge of these sites. This, the relative ease of implementation analytes.
and the potentially high throughput have made pre-column bioaffinity assays commercially
attractive. Consequently, the Automated Ligand Identification System (ALIS), which is 1.1.3 Modification methods in metabolism studies
based on size exclusion chromatography–reversed phase LC–MS, has become the biggest
commercial success in HRS so far [12]. In post-column assays, the separation is the first For the study of the metabolism of an API, different model systems are used to predict the
step. Consequently, all following steps are applied to the pre-separated analytes meaning changes of an API after ingestion by patients or healthy volunteers. Most commonly, this is
that they affect each analyte individually. This means that, in principle, a separate signal is attempted by the use of animal models, cultured human hepatocytes or in vitro incubations
obtained for every compound with a (sufficiently large) biological effect. Due to the fact that, with (human) liver fractions [44,45]. However, none of these methods is particularly suitable
assuming sufficient separation, every analyte enters the post-column assay separately, to produce sufficient amounts of material to further investigate the chemical and biological
every HTS assay is theoretically convertible into a post-column assay. For more details on properties of the metabolites. Therefore, an increasing number of methods is proposed to
post-column assays, refer to Chapter 2.1. augment metabolic studies by producing metabolite standards. This is necessary, because
A distinction which only applies to post-column assays is whether the binder, tracer or the many reactions observed in metabolism are not easily reproduced by classic organic
detected reactant is separated from other molecules before the actual detection step. This synthesis approaches. Oxidative metabolism yields a broad variety of reactions, depending
is necessary when the assay principle does not allow to distinguish bound and unbound on the structure of the active ingredient. Mainly aromatic and aliphatic hydroxylation, N- and
tracer (or binders) or substrate and product(s) solely on basis of their detected properties. O-dealkylation, dehalogenation, epoxidation, carbonyl oxidation and N- and S-oxidation
If this is indeed necessary, the assay is called heterogeneous. A prime example are assays are observed [46]. Especially the introduction of hydroxyl groups to non-activated carbon
14 15
sodium hypochlorite [49]. In classic synthesis, the oxidation aims at an existing functional
group and the power of the oxidizing agent is tightly controlled in order to direct the
1 modification to this site. However, enzymatic catalysis, which underlies metabolism, is much
more controlled by proximity to the catalytic groups, e.g., the heme group in cytochrome
1
P450s (CYPs), than by redox potentials [50]. Therefore, for the generation of metabolite-
like lead libraries, the side reactions obtained with strong and thus less selective oxidizing
agents can be of great interest.
Though hydrogen peroxide by itself is not a strong oxidizing agent, it can be activated,
for example thermally, photochemically or by metal catalysis [51]. The reactive species is
the hydroxyl radical. Additionally, hydrogen peroxide can yield epoxidation of electrophilic
alkenes under basic conditions [52]. In the context of drug metabolism studies, hydrogen
peroxide has been used, amongst others, to achieve S-oxidation [47,53,54], aromatic and
aliphatic hydroxylation [55], formation of aliphatic ketones [55], N-oxdidation [56-58], N-
and O-dealkylation [57,58] and carboxylic acid formation [58].
Furthermore, metallo-organic catalysts have been employed for the generation of
metabolites [59]. These metalloporphyrins are meant to mimic the active state of the CYP
coenzyme Protoporphyrin IX. They can be operated with molecular oxygen and reducing
co-factors, but are often also used in combination with chemical oxidants like hydrogen
peroxide or sodium hypochlorite. This opens a grey area between catalytic activation
of hydrogen peroxide and catalytic, metallo-organic oxidation with hydrogen peroxide
regeneration.

1.1.3.b) Electrochemistry
Electrochemical conversion (EC) has been extensively explored [60] and in recent years
has found increasing use in metabolic studies. Due to the low matrix content of the reactions
in EC, it has become very popular in combination with MS and LC–MS, and has even lead
to hyphenated EC–MS and EC–LC–MS systems. The field has been extensively reviewed
by the groups of Karst [59,61-63], Bischoff/Bruins [64,65] and Jurva [66]. EC can mimic
a wide range of metabolic reactions, such as aromatic and aliphatic hydroxylation, N-, P-
and S-oxidation, N- and O-dealkylation, alcohol oxidation, dehydrogenation, epoxidation
and dehalogenation [61,64]. Additionally, it has been used for the generation and study of
reactive metabolites, such as quinones and quinoneimines. Prominent examples are N-(4-
Figure 2: Generation of metabolite-like lead libraries. A schematic representation of the four
modification methods (see Chapter  1.1.3), used to generate metabolite-like lead libraries (see hydroxyphenyl)acetamid (Paracetamol), amodiaquine and clozapine [67].
Chapter 1.1.1.b), is shown. As substrates, we employed p38 mitogen-activated protein kinase α (p38α) The EC reaction is driven by adding (reduction) or removing (oxidation) electrons to/from
inhibitors (either common molecular probes or clinical candidates) which were very hydrophobic but the substrate at the electrode surface. Two reaction classifications are distinguished in
had high affinity. We aimed at generating more hydrophilic conversion products which retain p38α EC as well as with CYP reactions: single electron transfer oxidation and hydrogen atom
affinity, as is visualized in the right column. Though HRS platforms have been reported that allow transfer/abstraction [64]. Additionally, ozonolysis might occur at high voltages [68].
selectivity determination, we did not include this feature in either of the two HRS platforms developed
in this thesis.
1.1.3.c) Biocatalytic approaches
moieties is a great synthetic challenge. Because the methods, used for metabolite standard Because in enzymes the catalytic group is presented in the context of a complex, three-
synthesis, offer access to a unique chemical space and are often scalable to preparative dimensional active site in which the substrate has to fit in one (or a limited number of)
dimensions, this thesis applies some of them to the generation of metabolite-like lead specific orientation(s) [50], biosynthetic approaches offer an unmatched product selectivity,
libraries (see Chapter 1.1.1.b). especially when it comes to chiral synthesis [69]. A variety of enzyme classes has been
As mentioned, an increasing number of these modification methods are available and this employed for biocatalytic purposes including aldolases, lyases, dehydrogenases, reductases,
section gives some examples, mainly focused on the methods actually used in this thesis transaminases, esterases, oxidases, proteases and lipases [69-74]. Viewed as “by far the
(see Figure 2). For a better overview, the methods are divided into chemical, electrochemical, most dominant and important” enzymes in drug metabolism, the CYP superfamily has
biocatalytic and photochemical approaches. Though the methods are logically separated received specific attention in drug metabolism studies [46]. CYPs are capable of catalyzing
by the nature of the (initial) redox agent, with respect to other aspects such as reactions, most of the reactions of oxidative metabolism (see above). Very promising in biocatalysis
driving forces or even the ultimate redox species, the division may be artificial. is a CYP from Bacillus megaterium, named BM3, which shows a wide substrate spectrum,
a high solubility, high expression levels in E. coli and a high catalytic efficiency [75,76].
The latter is especially important, because CYPs use the expensive co-factor nicotinamide
1.1.3.a) Chemical oxidation adenine dinucleotide phosphate (reduced form: NADPH). Although its consumption can
Organic synthesis knows many oxidizing agents and some of them have been used in be reduced from stoichiometric to catalytic amounts by employing a co-factor regenerating
metabolism studies such as hydrogen peroxide [47], potassium permanganate [48] or
16 17
system based on glucose or glucose-6-phospate and their corresponding dehydrogenases, instruments, like quadrupole–TOF, ion trap–TOF, linear ion trap–orbitrap, quadrupole–
this presents a significant hurdle in large-scale application [77]. orbitrap, and quadrupole– or linear ion trap–Fourier-transform ion cyclotron resonance
1 The versatility of employing CYP BM3 for the biocatalytic conversion of APIs is demonstrated
in a recent review [76].
instruments [92] [93]. Obviously, the combination of high mass accuracy and fragmentation
makes the HR tandem MS instruments ideally suited for structure elucidation. Nowadays,
1
a kaleidoscope of fragmentation techniques is being developed, but for small molecules
1.1.3.d) Photochemistry collision induced dissociation (CID) is still applied almost exclusively [94]. However,
Photochemistry, meaning the use of light to activate chemical reactions, mainly acts uncertainty in the fundamental understanding of the ESI process [95] and the reactions
through the formation of radicals by homolytic cleavage [78]. While visible (VIS) light with governing CID-fragmentation (especially for small molecules), as opposed to the thoroughly
between 167  kJ•mol-1 (red) and 293  kJ•mol-1 (blue) is limited to weak bonds, ultraviolet elucidated main fragmentation reactions of the radical ions, for example the α-cleavage
(UV) light at 586 kJ•mol-1 strongly decomposes organic molecules. For this reason, most and the McLafferty rearrangement [85], present a serious hurdle in modern MS structure
applications use visible light. Next to radical mechanisms, light can induce electrocyclic elucidation. Additionally, there is a large variation between different instruments in the
reactions. Light- and thermally-induced electrocyclic reactions start from an exited state or actual energy deposited to the ions by CID, which varies between 1 and 100 eV for ion trap
the ground state, respectively. The difference in orbital geometry between the two states and quadrupole based fragmentation [94], while in EI a standard setting of 70 eV is applied.
results in a different stereochemistry [79]. Classical, synthetic applications of photochemistry Therefore, almost every ESI instrument produces different MS/MS spectra of the same
include polymerization, halogenation of alkanes and carbon-carbon bond formation using analyte, especially with respect to the relative abundance of product ions. A fundamental
organo-tin reagents [78]. In the pharmaceutical industry, another important aspect of difference is found between ion trap geometry collision cells and those with quadrupole
photochemistry is known as photostability which refers to the light-induced degradation of geometry. Quadrupole geometries are limited to one collision experiment per cell while ion
active ingredients during production and storage of a drug [80,81]. Similar to metabolism, traps can perform iterative fragmentation cycles until the number of ions falls below the
degradation can have a huge influence on efficacy and side effects of a formulation. In detection limit. Therefore, ion trap instruments allow a much more reliable interpretation of
addition, light-induced degradation can also occur in vivo during the use of cosmetics [82] precursor-product ion relationships [96].
or pharmaceuticals [83]. The advent of tandem MS required the development of various data acquisition strategies
With respect to the radical mechanism, photochemistry [78] shares some common ground to fully exploit its possibilities. The most straightforward is the product-ion analysis mode.
with hydrogen peroxide [51], electrochemical and CYP oxidation [64]. Light and EC have Herein, a precursor ion is selected, fragmented and a mass spectrum of the fragments
the advantage that the energy input is tunable by changing the wavelength or the voltage, collected. Two other widespread strategies have greatly contributed to the success of
respectively. The radical and electrocyclic reaction mechanisms in photochemistry allow triple-quadrupole instruments. Selected reaction monitoring (SRM) follows one or multiple
rearrangements in the scaffold of the molecule which are not accessible by the other specific CID-reactions by selecting one precursor ion in Q1 and one fragment ion in Q3 for
methods [84]. each reaction. It is the most popular method for targeted quantitation, probably because
of the dramatically increased speed, selectivity, and data reduction compared to a full-
spectrum analysis. The neutral loss analysis mode searches for a specific m/z difference
1.1.4 Structure elucidation in mixtures using mass between the precursor and the product ion, thus for a neutral loss. It is often used to monitor
spectrometry and nuclear magnetic resonance classes of molecules which show the same specific neutral loss. Because neutral loss
spectroscopy analysis shows all class compounds regardless of the precursor m/z, as long as the neutral
loss is retained, it can also detect (some) unknown molecules. Nowadays, product-ion
For the structure elucidation of small, organic molecules a wide variety of methods is analysis is becoming increasingly fast and sensitive, e.g., in quadrupole–TOF instruments.
available [85,86]. This includes the classical spectrophotometric techniques like UV/VIS CID-reactions and neutral losses may also be monitored by data interpretation of product-
absorption, fluorescence and phosphorescence emission spectroscopy, and vibrational ion analysis spectra. Thus, combining the strength of HR-(tandem)MS in structure
approaches like infra-red and Raman spectroscopy. Additionally, scattering techniques like elucidation with the quantitative interpretation of CID-reactions or neutral losses is an
X-ray or neutron scattering are used. However, the focus is on mass spectrometry (MS) emerging approach [97]. A powerful tandem MS data acquisition strategy for monitoring
and nuclear magnetic resonance spectroscopy (NMR) which dominate today’s routine unknowns is data-dependent acquisition. First, a full spectrum without fragmentation is
structure elucidation of small molecules. Special attention is paid to structure elucidation in collected and processed in real-time. Based on pre-selected criteria, the software selects
mixtures which is one of the main challenges in this thesis. the most abundant ions to be fragmented and collects their MS/MS spectra. For ion trap
fragmentation this can be an iterative process (MSn). In this way, the process of identifying
1.1.4.a) Mass spectrometry unknown ions and collecting fragmentation data for structure elucidation can be greatly
For MS, mainly two developments have enabled structure elucidation. Firstly, the high automated.
mass accuracy of the high-resolution (HR) mass analyzers [87], i.e., sector field, time-of- The most important breakthrough with respect to MS analysis of mixtures was certainly the
flight (TOF), Orbitrap [88,89], and Fourier-transform ion cyclotron resonance, allows the hyphenation of MS to separation techniques [98,99], most importantly gas chromatography
determination of the elemental composition of ions [90,91] [92]. Secondly, fragmentation (GC) using EI and LC using ESI. This hyphenation does not only enable the detection of
reveals functional groups and residues by observing specific neutral losses and allows individual isobaric and isomeric compounds, but it also significantly reduces the occurrence
interpretation of their connectivity. Classically, fragmentation was achieved during electron of ionization suppression issues in ESI. Fortunately, modern (reversed phase (RP)) LC and
ionization (EI) with the risk of missing the molecular ion [87]. After the introduction of the ESI-MS are very well compatible [99]. In addition, LC–HR-MS resolves isobaric compounds
soft ionization techniques, especially electrospray ionization (ESI), fragmentation of the which have not been separated, but issues with isomers and ionization suppression cannot
protonated molecule had to be achieved in the mass analyzer. This fueled the development be elevated by HR-MS alone. In some cases, fragmentation can help in the discrimination of
of tandem MS and ion trap MSn. Tandem MS instruments can be divided into unit-resolution isomers. When standards are available, fragmentation patterns on the same MS instrument
instruments, like quadrupole–linear ion trap and triple-quadrupole, and (hybrid) HR can be compared to that of the unknown. Recently, the hyphenation of ion mobility

18 19
spectrometry and MS has received increasing attention accompanied by the introduction the backbone atom of organic molecules, is the second most commonly detected element
of new commercial instrumentation [100]. Because the drift-time is usually on the scale of [85,105,106]. However, its most abundant natural isotope 12C is not NMR active, leaving
1 the MS experiment or even shorter and separation is based on size and shape rather than
on the physicochemical properties employed in GC and LC, IMS might offer an interesting
only 13C with ca. 1% natural abundance. Consequentially, 13C-NMR is much less sensitive
than 1H-NMR. However, chemical shifts are more widely distributed, leading to higher
1
secondary separation dimension. However, in contrast to separation techniques, IMS does resolution, and quaternary carbon atoms are revealed which are obviously hidden in
not reduce ionization suppression as it is positioned after the ionization step. 1
H-NMR. Heteronuclear 13C-1H correlation NMR also suffers from the low 13C abundance
There are a number of data interpretation methods which were specifically developed for and is therefore even less sensitive than 2D-1H-NMR. Examples are heteronuclear single-
the analysis of unknown mixtures of related compounds such as they occur, for example, in quantum correlation (HSQC) and heteronuclear multi-bond correlation (HMBC) which
metabolism studies. The profile group approach [101,102] assumes that the identity of one reveal short-range or long-range 13C-1H J-coupling partners, respectively.
compound in the mixture is known and that this compound is related by simple chemical Heteroatoms are also used in NMR of small molecules, mainly 15N, 19F and 31P [85], and
modifications to the other compounds in the mixture, for example an active ingredient and sometimes employed in correlation experiments like 19F-1H [107], 19F-13C, 19F-19F [108], and
its metabolites [102] or a substrate and its multiple products from a shotgun synthesis 15
N-1H [109].
approach [42]. Firstly, the fragmentation of the standard (parent drug) is thoroughly LC–NMR is certainly an important development in mixture analysis by NMR though the less
analyzed on the MS instrument intended for the study, the data are interpreted, and a straightforward hyphenation has so far prevented the same widespread use which LC–MS
fragmentation tree is established. A number of structurally informative fragment ions are has seen [110]. The sensitivity issue usually also prevents real-time NMR detection of LC.
selected as profile groups. Then, the fragmentation trees of all analytes are measured LC–NMR systems often operate with a stopped-flow approach and/or with an intermediate
and compared to that of the standard, following mainly the profile groups. The difference solid-phase extraction (SPE) step, decoupling the chromatographic from the detection time
in elemental composition between the analyte and the standard is found back in some scale and (in SPE) additionally allowing solvent exchange and analyte pre-concentration
profile groups, which is evidence that the molecule is modified in the corresponding part [111,112]. The increasing resolution of NMR instruments has also enabled the direct
of the molecule. The difference depends on the modification, for example, oxygenation detection of mixtures by NMR. Today, it is possible to analyze even complex biological
and dehydrogenation result in +O (+16 Da) or –H2 (–2 Da), respectively. Thus, the site of samples [113]. Simplification of the complex spectra by specialized pulse methods can
modification can be narrowed down. In addition, changes in (relative) retention time in RPLC contribute to resolving overlapping signals. The effect is similar to the transition from EI to
can be used to distinguish the isomeric modifications of common metabolic reactions, like ESI in the way that information is sacrificed in order to strengthen the data that remains.
hydroxylation and N-oxidation [90]. Simplification is achieved by canceling the influence of J-coupling on the spectra which
MS is one of the most sensitive techniques for structure elucidation and fast enough for real- ideally yields singlet peaks and thus greatly simplifies the determination of chemical shifts
time collection of structural data in LC–MSn combinations. LC–MS or GC–MS are therefore in complex samples. It is routine to decouple different nuclei (heteronuclear decoupling)
often employed as the first line of structure elucidation/confirmation [103]. Unfortunately, [114], for example protons in 13C-NMR. However, the decoupling of the same nuclei
modifications can rarely be pinpointed with single atom accuracy. As a result, NMR is often (homonuclear decoupling), for example protons in 1H-NMR, is subject to ongoing study
employed at a later stage to elucidate the exact position of the modifications [Chapter 3.2] [115] and especially interesting for the NMR analysis of mixtures.
[104]. However, NMR requires much more material than MS. Miniaturization is a promising approach to enhance the sensitivity of NMR. By reducing
the detection volume without equally reducing the concentration sensitivity, an increase in
1.1.4.b) Nuclear magnetic resonance spectroscopy mass sensitivity is achieved. While sample pre-concentration, e.g., by SPE, becomes even
The basis of the structure elucidation of a small, organic molecule by NMR is usually a more crucial, the total amount of sample material used can be decreased by ca. 100-fold
proton spectrum (1H-NMR) [85,105,106]. This can already reveal a lot of information about in some cases. Different geometries have been proposed to achieve a well focus rf-field
the structure of the molecule. The chemical shift of a proton indicates the hybridization [116,117].
of the connected carbon atom and the proximity to a heteroatom. Integration reveals the Despite all the structural information that can be obtained from the various NMR
relative number of chemically equivalent protons which, combined with the hybridization experiments, the input of information obtained with MS experiments greatly complements
information from the chemical shift, establishes the connectivity of the associated carbon NMR data [111]. The simplest example is the calculation of the elemental composition
atom. Analysis of the J-coupling pattern hints to the number of neighboring protons while from HR-MS experiments which amongst many other advantages is a much faster way of
J-coupling strength indicates their distance and steric features, such as E/Z isomerism, and revealing heteroatoms than any method available to NMR [118]. Especially, sulfur, chlorine
can be used to match J-coupling partners. The signals of exchangeable protons are usually and bromine are readily followed by MS while their NMR detection has to be called exotic at
broader than the other signals, if they are not eliminated completely by efficient exchange the least. Even if all structural information can be obtained by NMR alone, MS can provide
with the deuterated solvent. a second independent confirmation and exclude symmetric dimeric structures. Additionally,
For the structural analysis of more complex molecules, homonuclear 1H correlation (2D-1H- because MS is generally faster and more sensitive than NMR, the same information can
NMR) spectra are often useful [85,105,106]. They can reveal more structural information, be obtained by MS earlier on and more economically in a structure elucidation procedure.
but are less sensitive or require longer measurement time. Correlation spectroscopy Due to the more straightforward hyphenation of LC and ESI-MS, this is especially true were
(COSY) reveals the interaction of J-coupling partners which is especially useful when separation is needed, thus for LC–MS vs. LC–NMR [110]. However, because NMR has
several J-couplings of similar strength were observed in the 1H spectrum. As a variation much more possibilities to elucidate structural details and even conformational questions,
of COSY, total correlation spectroscopy (TOCSY) reveals not only direct coupling, but LC–MS and NMR have become a dream team of structural analysis for small, organic
also interactions of couplings, thus revealing so-called spin systems. Instead J-coupling molecules [104,111,112].
interactions, nuclear Overhauser effect spectroscopy (NOESY) and rotating-frame nuclear
Overhauser effect spectroscopy (ROESY) reveal spacial proximity of protons by employing
dipole-dipole interactions.
Protons are not the only nuclei which can be followed by NMR. Not surprisingly, carbon,
20 21
References 24. Oosterkamp AJ, Irth H, Villaverde Herraiz MT, Tjaden UR, van der Greef J (1997) Theoretical
concepts of on-line liquid chromatographic-biochemical detection systems. I. Detection systems
1. Giacomotto J, Segalat L (2010) High-throughput screening and small animal models, where are
1 2.
we? Br J Pharmacol 160 (2):204-216
Erhardt PW, Proudfoot JR (2007) 1.02 - Drug Discovery: Historical Perspective, Current Status,
based on labelled ligands. J Chromatogr A 787 (1-2):27-35
25. Oosterkamp AJ, Irth H, Tjaden UR, van der Greef J (1997) Theoretical concepts of on-line 1
and Outlook. In: Taylor JB, Triggle DJ (eds) Comprehensive Medicinal Chemistry II, vol 1. liquid chromatographic- biochemical detection systems II. Detection systems based on labelled
Elsevier, pp 29-96 affinity proteins. Journal of Chromatography A 787 (1–2):37-46
3. Kool J, Lingeman H, Niessen W, Irth H (2010) High throughput screening methodologies 26. Kool J, Jonker N, Irth H, Niessen WM (2011) Studying protein-protein affinity and immobilized
classified for major drug target classes according to target signaling pathways. Comb Chem ligand-protein affinity interactions using MS-based methods. Anal Bioanal Chem 401 (4):1109-
High Throughput Screen 13 (6):548-561 1125
4. Cik M, Jurzak MR (2007) 3.32 - High-Throuput and High-Content Screening. In: Taylor JB, 27. Reinen J, Ferman S, Vottero E, Vermeulen NP, Commandeur JN (2011) Application of a
Triggle DJ (eds) Comprehensive Medicinal Chemistry II, vol 3. Elsevier, pp 679-696 fluorescence-based continuous-flow bioassay to screen for diversity of cytochrome P450 BM3
5. Harris CJ, Hill RD, Sheppard DW, Slater MJ, Stouten PF (2011) The design and application of mutant libraries. J Biomol Screen 16 (2):239-250
target-focused compound libraries. Comb Chem High Throughput Screen 14 (6):521-531 28. Kool J, Rudebeck AF, Fleurbaaij F, Nijmeijer S, Falck D, Smits RA, Vischer HF, Leurs R, Niessen
6. Keseru GM, Makara GM (2006) Hit discovery and hit-to-lead approaches. Drug Discov Today 11 WM (2012) High-resolution metabolic profiling towards G protein-coupled receptors: rapid and
(15-16):741-748 comprehensive screening of histamine H(4) receptor ligands. J Chromatogr A 1259:213-220
7. Janzen WP, Popa-Burke IG (2009) Advances in improving the quality and flexibility of compound 29. Jonker N, Lingeman H, Irth H (2010) Direct Dynamic Protein-Affinity Selection Mass-
management. J Biomol Screen 14 (5):444-451 Spectrometry. Chromatographia 72 (1-2):7-13
8. Davies JW, Glick M, Jenkins JL (2006) Streamlining lead discovery by aligning in silico and high- 30. Jonker N, Kretschmer A, Kool J, Fernandez A, Kloos D, Krabbe JG, Lingeman H, Irth H (2009)
throughput screening. Curr Opin Chem Biol 10 (4):343-351 Online magnetic bead dynamic protein-affinity selection coupled to LC-MS for the screening of
9. Kool J, Giera M, Irth H, Niessen WM (2011) Advances in mass spectrometry-based post-column pharmacologically active compounds. Anal Chem 81 (11):4263-4270
bioaffinity profiling of mixtures. Anal Bioanal Chem 399 (8):2655-2668 31. Retra K, Geitmann M, Kool J, Smit AB, de Esch IJ, Danielson UH, Irth H (2010) Development
10. Shi SY, Zhou HH, Zhang YP, Jiang XY, Chen XQ, Huang KL (2009) Coupling HPLC to on-line, of surface plasmon resonance biosensor assays for primary and secondary screening of
post-column (bio)chemical assays for high-resolution screening of bioactive compounds from acetylcholine binding protein ligands. Anal Biochem 407 (1):58-64
complex mixtures. Trac-Trends in Analytical Chemistry 28 (7):865-877 32. Oosterkamp AJ, Villaverde Herraiz MT, Irth H, Tjaden UR, van der Greef J (1996) Reversed-
11. Moos WH (2007) 2.01 - The Intersection of Strategy and Drug Research. In: Taylor JB, Triggle phase liquid chromatography coupled on-line to receptor affinity detection based on the human
DJ (eds) Comprehensive Medicinal Chemistry II, vol 2. Elsevier, pp 1-84 estrogen receptor. Anal Chem 68 (7):1201-1206
12. Annis DA, Athanasopoulos J, Curran PJ, Felsch JS, Kalghatgi K, Lee WH, Nash HM, Orminati 33. Derks RJ, Hogenboom AC, van der Zwan G, Irth H (2003) On-line continuous-flow, multi-
J-PA, Rosner KE, Shipps Jr GW, Thaddupathy GRA, Tyler AN, Vilenchik L, Wagner CR, protein biochemical assays for the characterization of bioaffinity compounds using electrospray
Wintner EA (2004) An affinity selection–mass spectrometry method for the identification of small quadrupole time-of-flight mass spectrometry. Anal Chem 75 (14):3376-3384
molecule ligands from self-encoded combinatorial libraries: Discovery of a novel antagonist of 34. Heus F, Giera M, de Kloe GE, van Iperen D, Buijs J, Nahar TT, Smit AB, Lingeman H, de Esch
E. coli dihydrofolate reductase. International Journal of Mass Spectrometry 238 (2):77-83 IJ, Niessen WM, Irth H, Kool J (2010) Development of a microfluidic confocal fluorescence
13. Wolfender JL, Queiroz EF (2012) New approaches for studying the chemical diversity of natural detection system for the hyphenation of nano-LC to on-line biochemical assays. Anal Bioanal
resources and the bioactivity of their constituents. Chimia (Aarau) 66 (5):324-329 Chem 398 (7-8):3023-3032
14. Potterat O, Hamburger M (2013) Concepts and technologies for tracking bioactive compounds 35. Reinen J, Kool J, Vermeulen NP (2008) Reversed-phase liquid chromatography coupled on-
in natural product extracts: generation of libraries, and hyphenation of analytical processes with line to estrogen receptor bioaffinity detection based on fluorescence polarization. Anal Bioanal
bioassays. Nat Prod Rep 30 (4):546-564 Chem 390 (8):1987-1998
15. Heus F, Vonk F, Otvos RA, Bruyneel B, Smit AB, Lingeman H, Richardson M, Niessen WM, Kool 36. Ladbury JE (2010) Calorimetry as a tool for understanding biomolecular interactions and an aid
J (2013) An efficient analytical platform for on-line microfluidic profiling of neuroactive snake to drug design. Biochem Soc Trans 38 (4):888-893
venoms towards nicotinic receptor affinity. Toxicon 61:112-124 37. Schebb NH, Falck D, Faber H, Hein EM, Karst U, Hayen H (2009) Fast method for monitoring
16. de Jong CF, Derks RJ, Bruyneel B, Niessen W, Irth H (2006) High-performance liquid phospholipase A2 activity by liquid chromatography-electrospray ionization mass spectrometry.
chromatography-mass spectrometry-based acetylcholinesterase assay for the screening of J Chromatogr A 1216 (27):5249-5255
inhibitors in natural extracts. J Chromatogr A 1112 (1-2):303-310 38. Hodder P, Mull R, Cassaday J, Berry K, Strulovici B (2004) Miniaturization of intracellular calcium
17. Guengerich FP (2008) Cytochrome p450 and chemical toxicology. Chem Res Toxicol 21 (1):70- functional assays to 1536-well plate format using a fluorometric imaging plate reader. J Biomol
83 Screen 9 (5):417-426
18. Fura A, Shu YZ, Zhu M, Hanson RL, Roongta V, Humphreys WG (2004) Discovering drugs 39. Prystay L, Gagne A, Kasila P, Yeh LA, Banks P (2001) Homogeneous cell-based fluorescence
through biological transformation: role of pharmacologically active metabolites in drug discovery. polarization assay for the direct detection of cAMP. J Biomol Screen 6 (2):75-82
J Med Chem 47 (18):4339-4351 40. Temporini C, Ceruti S, Calleri E, Ferrario S, Moaddel R, Abbracchio MP, Massolini G (2009)
19. Hann MM (2011) Molecular obesity, potency and other addictions in drug discovery. Development of an immobilized GPR17 receptor stationary phase for binding determination
Medchemcomm 2 (5):349-355 using frontal affinity chromatography coupled to mass spectrometry. Anal Biochem 384 (1):123-
20. Camenisch G, Folkers G, van de Waterbeemd H (1996) Review of theoretical passive drug 129
absorption models: historical background, recent developments and limitations. Pharm Acta 41. Giera M, Heus F, Janssen L, Kool J, Lingeman H, Irth H (2009) Microfractionation revisited: a
Helv 71 (5):309-327 1536 well high resolution screening assay. Anal Chem 81 (13):5460-5466
21. Dobson PD, Patel Y, Kell DB (2009) ‘Metabolite-likeness’ as a criterion in the design and 42. Giera M, de Vlieger JS, Lingeman H, Irth H, Niessen WM (2010) Structural elucidation of
selection of pharmaceutical drug libraries. Drug Discov Today 14 (1-2):31-40 biologically active neomycin N-octyl derivatives in a regioisomeric mixture by means of liquid
22. Jonker N, Kool J, Krabbe JG, Retra K, Lingeman H, Irth H (2008) Screening of protein-ligand chromatography/ion trap time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 24
interactions using dynamic protein-affinity chromatography solid-phase extraction-liquid (10):1439-1446
chromatography-mass spectrometry. J Chromatogr A 1205 (1-2):71-77 43. Nijmeijer S, Vischer HF, Rudebeck AF, Fleurbaaij F, Falck D, Leurs R, Niessen WM, Kool J
23. Jonker N, Kool J, Irth H, Niessen WM (2011) Recent developments in protein-ligand affinity (2012) Development of a profiling strategy for metabolic mixtures by combining chromatography
mass spectrometry. Anal Bioanal Chem 399 (8):2669-2681 and mass spectrometry with cell-based GPCR signaling. J Biomol Screen 17 (10):1329-1338

22 23
44. Laveé T, Funk C (2007) 5.03 - In Vivo Absorption, Metabolism, and Excretion Studies in Discovery using online electrochemistry/liquid chromatography/mass spectrometry. Anal Chem 80
and Development. In: Taylor JB, Triggle DJ (eds) Comprehensive Medicinal Chemistry II, vol 5. (24):9714-9719
1 Elsevier, pp 31-50
45. Parmentier Y, M.-J. B, M B, B W (2007) 5.10 - In Vitro Studies of Drug Metabolism. In: Taylor JB,
68. Ottinger SE, Mayura K, Lemke SL, McKenzie KS, Wang N, Kubena LF, Phillips TD (1999)
Utilization of electrochemically generated ozone in the degradation and detoxication of benzo[a] 1
Triggle DJ (eds) Comprehensive Medicinal Chemistry II, vol 5. Elsevier, pp 231-257 pyrene. J Toxicol Environ Health A 57 (8):565-583
46. Trager WF (2007) 5.05 - Principles of Drug Metabolism 1: Redox Reactions. Comprehensive 69. May O (2011) 16 - Sustainable Processes Based on Enzymes Enabling 100% Yield and 100%
Medicinal Chemistry II, vol 5. Elsevier, ee Concepts. In: Shioiri T, Izawa K, Konoike T (eds) Pharmaceutical Process Chemistry. Wiley-
47. Thevis M, Moller I, Thomas A, Beuck S, Rodchenkov G, Bornatsch W, Geyer H, Schanzer W VCH, pp 321-343
(2010) Characterization of two major urinary metabolites of the PPARdelta-agonist GW1516 70. Turner NJ, Truppo MD (2010) 14 - Biocatalytic Routes to Nonracemic Chiral Amines. In: Nugent
and implementation of the drug in routine doping controls. Anal Bioanal Chem 396 (7):2479- TC (ed) Chiral Amine Synthesis. Wiley-VCH, pp 431-459
2491 71. Yuryev R, Strompen S, Liese A (2011) Coupled chemo(enzymatic) reactions in continuous flow.
48. Meyer AH, Elsner M (2013) 13C/12C and 15N/14N Isotope Analysis to Characterize Degradation Beilstein J Org Chem 7:1449-1467
of Atrazine: Evidence from Parent and Daughter Compound Values. Environ Sci Technol 72. Turner NJ (2009) Directed evolution drives the next generation of biocatalysts. Nat Chem Biol 5
49. Lu W, Uetrecht JP (2007) Possible bioactivation pathways of lamotrigine. Drug Metab Dispos 35 (8):567-573
(7):1050-1056 73. Strohmeier GA, Pichler H, May O, Gruber-Khadjawi M (2011) Application of designed enzymes
50. Rea V, Kolkman AJ, Vottero E, Stronks EJ, Ampt KA, Honing M, Vermeulen NP, Wijmenga SS, in organic synthesis. Chem Rev 111 (7):4141-4164
Commandeur JN (2012) Active site substitution A82W improves the regioselectivity of steroid 74. Nestl BM, Nebel BA, Hauer B (2011) Recent progress in industrial biocatalysis. Curr Opin Chem
hydroxylation by cytochrome P450 BM3 mutants as rationalized by spin relaxation nuclear Biol 15 (2):187-193
magnetic resonance studies. Biochemistry 51 (3):750-760 75. Munro AW, Leys DG, McLean KJ, Marshall KR, Ost TW, Daff S, Miles CS, Chapman SK,
51. Jones CW (1999) 2.2 - Basic Chemistry of Hydrogen Peroxide. In: Applications of Hydrogen Lysek DA, Moser CC, Page CC, Dutton PL (2002) P450 BM3: the very model of a modern
Peroxide and Derivatives. The Royal Society of Chemistry, pp 37-78 flavocytochrome. Trends Biochem Sci 27 (5):250-257
52. Clayden J, Greeves N, Warren S, Wothers P (2001) 23 - Electrophilic alkenes - Nucleophilic 76. Whitehouse CJ, Bell SG, Wong LL (2012) P450(BM3) (CYP102A1): connecting the dots. Chem
epoxidation. In: Organic Chemistry. Oxford University Press, pp 588-589 Soc Rev 41 (3):1218-1260
53. Stariat J, Sestak V, Vavrova K, Nobilis M, Kollarova Z, Klimes J, Kalinowski DS, Richardson 77. Wichmann R, Vasic-Racki D (2005) Cofactor regeneration at the lab scale. Adv Biochem Eng
DR, Kovarikova P (2012) LC-MS/MS identification of the principal in vitro and in vivo phase I Biotechnol 92:225-260
metabolites of the novel thiosemicarbazone anti-cancer drug, Bp4eT. Anal Bioanal Chem 403 78. Clayden J, Greeves N, Warren S, Wothers P (2001) 39 - Radical reactions. In: Organic
(1):309-321 Chemistry. Oxford University Press, pp 1019-1052
54. Sarma D, Hajovsky H, Koen YM, Galeva NA, Williams TD, Staudinger JL, Hanzlik RP (2012) 79. Clayden J, Greeves N, Warren S, Wothers P (2001) 36 - Pericyclic Reaction 2: Sigmatropic and
Covalent modification of lipids and proteins in rat hepatocytes and in vitro by thioacetamide electrocyclic reactions - Electrocyclic reactions. In: Organic Chemistry. Oxford University Press,
metabolites. Chem Res Toxicol 25 (9):1868-1877 pp 956-965
55. Marques LA, Kool J, de Kanter F, Lingeman H, Niessen W, Irth H (2010) Production and on- 80. Baertschi SW, Alsante KM, Tonnesen HH (2010) A critical assessment of the ICH guideline
line acetylcholinesterase bioactivity profiling of chemical and biological degradation products of on photostability testing of new drug substances and products (Q1B): Recommendation for
tacrine. J Pharm Biomed Anal 53 (3):609-616 revision. J Pharm Sci 99 (7):2934-2940
56. Marques LA, Maada I, de Kanter FJ, Lingeman H, Irth H, Niessen WM, Giera M (2011) Stability- 81. Bakshi M, Singh S (2002) Development of validated stability-indicating assay methods--critical
indicating study of the anti-Alzheimer’s drug galantamine hydrobromide. J Pharm Biomed Anal review. J Pharm Biomed Anal 28 (6):1011-1040
55 (1):85-92 82. Tolleson WH, Cherng SH, Xia Q, Boudreau M, Yin JJ, Wamer WG, Howard PC, Yu H, Fu PP
57. Edlund PO, Baranczewski P (2004) Identification of BVT.2938 metabolites by LC/MS and LC/ (2005) Photodecomposition and phototoxicity of natural retinoids. Int J Environ Res Public
MS/MS after in vitro incubations with liver microsomes and hepatocytes. J Pharm Biomed Anal Health 2 (1):147-155
34 (5):1079-1090 83. Busker RW, Beijersbergen van Henegouwen GM, Menke RF, Vasbinder W (1988) Formation of
58. Jayamanne M, Granelli I, Tjernberg A, Edlund PO (2010) Development of a two-dimensional methemoglobin by photoactivation of nitrofurantoin or of 5-nitrofurfural in rats exposed to UV-A
liquid chromatography system for isolation of drug metabolites. J Pharm Biomed Anal 51 light. Toxicology 51 (2-3):255-266
(3):649-657 84. Marques LA, Giera M, de Kanter FJ, Niessen WM, Lingeman H, Irth H (2010) Photohuperzine
59. Lohmann W, Karst U (2008) Biomimetic modeling of oxidative drug metabolism : Strategies, A-A new photoisomer of huperzine A: structure elucidation, formation kinetics and activity
advantages and limitations. Anal Bioanal Chem 391 (1):79-96 assessment. J Pharm Biomed Anal 52 (2):190-194
60. Hammerich O, Lund H (2000) Organic Electrochemistry. Fourth edn. CRC Press, 85. Hesse M, Meier H, Zeeh B (2005) Spektroskopische Methoden in der organischen Chemie. 7.
61. Baumann A, Karst U (2010) Online electrochemistry/mass spectrometry in drug metabolism edn. Georg Thieme Verlag,
studies: principles and applications. Expert Opin Drug Metab Toxicol 6 (6):715-731 86. Clayden J, Greeves N, Warren S, Wothers P (2001) 3 - Determining organic structures. In:
62. Faber H, Jahn S, Kunnemeyer J, Simon H, Melles D, Vogel M, Karst U (2011) Electrochemistry/ Organic Chemistry. Oxford University Press, pp 47-79
liquid chromatography/mass spectrometry as a tool in metabolism studies. Angew Chem Int Ed 87. Niessen WMA (2006) 2 - Mass Spectrometry. In: Liquid Chromatography – Mass Spectrometry.
Engl 50 (37):A52-58 Chromatographic Science Series, Third edn. CRC Press, pp 23-50
63. Jahn S, Karst U (2012) Electrochemistry coupled to (liquid chromatography/) mass spectrometry- 88. Makarov A (2000) Electrostatic axially harmonic orbital trapping: a high-performance technique
-current state and future perspectives. J Chromatogr A 1259:16-49 of mass analysis. Anal Chem 72 (6):1156-1162
64. Permentier HP, Bruins AP, Bischoff R (2008) Electrochemistry-mass spectrometry in drug 89. Zubarev RA, Makarov A (2013) Orbitrap mass spectrometry. Anal Chem 85 (11):5288-5296
metabolism and protein research. Mini Rev Med Chem 8 (1):46-56 90. Holcapek M, Kolarova L, Nobilis M (2008) High-performance liquid chromatography-tandem
65. Nouri-Nigjeh E, Bischoff R, Bruins AP, Permentier HP (2011) Electrochemistry in the mimicry of mass spectrometry in the identification and determination of phase I and phase II drug
oxidative drug metabolism by cytochrome P450s. Curr Drug Metab 12 (4):359-371 metabolites. Anal Bioanal Chem 391 (1):59-78
66. Johansson T, Weidolf L, Jurva U (2007) Mimicry of phase I drug metabolism--novel methods for 91. Niessen WMA (2011) Fragmentation of toxicologically relevant drugs in positive-ion liquid
metabolite characterization and synthesis. Rapid Commun Mass Spectrom 21 (14):2323-2331 chromatography-tandem mass spectrometry. Mass Spectrom Rev 30 (4):626-663
67. Lohmann W, Hayen H, Karst U (2008) Covalent protein modification by reactive drug metabolites 92. Singh S, Handa T, Narayanam M, Sahu A, Junwal M, Shah RP (2012) A critical review on the use

24 25
of modern sophisticated hyphenated tools in the characterization of impurities and degradation 119. de Boer AR, Alcaide-Hidalgo JM, Krabbe JG, Kolkman J, van Emde Boas CN, Niessen WM,
products. J Pharm Biomed Anal 69:148-173 Lingeman H, Irth H (2005) High-temperature liquid chromatography coupled on-line to a
1 93. Glish GL, Burinsky DJ (2008) Hybrid mass spectrometers for tandem mass spectrometry. J Am
Soc Mass Spectrom 19 (2):161-172
continuous-flow biochemical screening assay with electrospray ionization mass spectrometric
detection. Anal Chem 77 (24):7894-7900 1
94. Sleno L, Volmer DA (2004) Ion activation methods for tandem mass spectrometry. J Mass
Spectrom 39 (10):1091-1112
95. Crotti S, Seraglia R, Traldi P (2011) Some thoughts on electrospray ionization mechanisms. Eur
J Mass Spectrom (Chichester, Eng) 17 (2):85-99
96. Kind T, Fiehn O (2010) Advances in structure elucidation of small molecules using mass
spectrometry. Bioanal Rev 2 (1-4):23-60
97. Campbell JL, Le Blanc JC (2012) Using high-resolution quadrupole TOF technology in DMPK
analyses. Bioanalysis 4 (5):487-500
98. Hübschmann H-J (2008) Handbook of GC/MS: Fundamentals and Applications. Wiley-VCH,
99. Niessen WMA (2006) 3 - Strategies in LC–MS interfacing. In: Liquid Chromatography – Mass
Spectrometry. Chromatographic Science Series, Third edn. CRC Press, pp 53-72
100. Kanu AB, Dwivedi P, Tam M, Matz L, Hill HH, Jr. (2008) Ion mobility-mass spectrometry. J Mass
Spectrom 43 (1):1-22
101. Kerns EH, Volk KJ, Hill SE, Lee MS (1995) Profiling New Taxanes Using LC/MS and LC/MS/MS
Substructural Analysis Techniques. Rapid Communications in Mass Spectrometry 9:1539-1545
102. Kerns EH, Rourick RA, Volk KJ, Lee MS (1997) Buspirone metabolite structure profile using a
standard liquid chromatographic-mass spectrometric protocol. J Chromatogr B Biomed Sci Appl
698 (1-2):133-145
103. Prakash C, Shaffer CL, Nedderman A (2007) Analytical strategies for identifying drug metabolites.
Mass Spectrom Rev 26 (3):340-369
104. de Vlieger JS, Kolkman AJ, Ampt KA, Commandeur JN, Vermeulen NP, Kool J, Wijmenga SS,
Niessen WM, Irth H, Honing M (2010) Determination and identification of estrogenic compounds
generated with biosynthetic enzymes using hyphenated screening assays, high resolution mass
spectrometry and off-line NMR. J Chromatogr B 878 (7-8):667-674
105. Günther H (1995) NMR Spectroscopy: basic principles, concepts, and applications in chemistry.
Wiley,
106. Keeler J (2007) Understanding NMR Spectroscopy. Wiley,
107. Ampt KA, Aspers RL, Jaeger M, Geutjes PE, Honing M, Wijmenga SS (2011) Application of
fluorine NMR for structure identification of steroids. Magn Reson Chem 49 (5):221-230
108. Aspers RL, Ampt KA, Dvortsak P, Jaeger M, Wijmenga SS (2013) Fluorine detected 2D NMR
experiments for the practical etermination of size and sign of homonuclear F-F and heteronuclear
C-F multiple bond J-coupling constants in multiple fluorinated compounds. J Magn Reson
231C:79-89
109. Martin GE, Hadden CE (2000) Long-range (1)H-(15)N heteronuclear shift correlation at natural
abundance. J Nat Prod 63 (4):543-585
110. Walker GS, O’Connell TN (2008) Comparison of LC-NMR and conventional NMR for structure
elucidation in drug metabolism studies. Expert Opin Drug Metab Toxicol 4 (10):1295-1305
111. Corcoran O, Spraul M (2003) LC-NMR-MS in drug discovery. Drug Discov Today 8 (14):624-631
112. Tang H, Xiao C, Wang Y (2009) Important roles of the hyphenated HPLC-DAD-MS-SPE-NMR
technique in metabonomics. Magn Reson Chem 47 Suppl 1:S157-162
113. Bart J, Kolkman AJ, Oosthoek-de Vries AJ, Koch K, Nieuwland PJ, Janssen JWG, van Bentum
JPM, Ampt KAM, Rutjes FPTJ, Wijmenga SS, Gardeniers JGE, Kentgens APM (2009) A
microfluidic high-resolution NMR flow probe. J Am Chem Soc 131 (14):5014-5015
114. Shaka AJ, Keeler J, Frenkiel T, Freeman R (1983) An Improved Sequence for Broadband
Decoupling: WALTZ-16. Journal of Magnetic Resonance 52:335-338
115. Kolkman AJ (2013) NMR spectroscopy in Drug Discovery – Application and Development
of NMR Based Methodologies for P450BM3 Drug Metabolism, Metabolite Identification and
Mixture Analysis. Radboud University Nijmegen, The Netherlands; ISBN 978-94-6191-514-6
116. van Bentum PJM, Janssen JWG, Kentgens APM, Bart J, Gardeniers JGE (2007) Stripline
probes for nuclear magnetic resonance. J Magn Reson 189 (1):104-113
117. Gokay O, Albert K (2012) From single to multiple microcoil flow probe NMR and related capillary
techniques: a review. Anal Bioanal Chem 402 (2):647-669
118. Zhu P, Tong W, Alton K, Chowdhury S (2009) An accurate-mass-based spectral-averaging
isotope-pattern-filtering algorithm for extraction of drug metabolites possessing a distinct isotope
pattern from LC-MS data. Anal Chem 81 (14):5910-5917

26 27
Chapter 1.2

Scope
Figure 3: The HRS workflow integrating modification, separation, affinity assessment and structure
determination. This approach is used in different forms throughout Chapter 3. The library generation
1 was explained in Chapter 1.1.3. Sample preparation beyond HPLC was only necessary for biocatalysis
[Chapter  3.2]. Different forms of pre-screening were applied to select only potentially interesting 1
libraries. The compromise between information content and sample throughput varied due to varying
sample numbers. The heart of the strategy is the HRS system allowing simultaneous separation,
affinity measurement and structural analysis [Chapter 2.2]. Also included are the example steps for
further evaluation of the new leads demonstrated for the biocatalysis libraries [Chapter 3.2]. Upscaling
and purification allow additional structural information from NMR and a quantitative comparison of
affinities, for example by IC50 values.

The aim of this thesis is to investigate the possibilities for the integration of innovative
synthetic methods into analytical platforms for drug discovery (see Figure  3). A strong
focus of this work is the advancement of existing and the development of novel analytical
methodology which would be demanded and inspired by this challenging objective. More
specifically, we studied workflows and developed platforms which integrate innovative,
analytical-scale analyte modification methods with HRS technology, MS structure
elucidation and (miniaturized) NMR. This will make neglected chemical space available
early on in lead optimization and might contribute to a more efficient drug discovery cycle,
if boldly implemented in novel screening paradigms.
Chapter 1.1 introduces important concepts starting with general aspects of the discovery
of API, for example screening. At the core, it introduces several approaches to HRS and
rationalizes the focus of this thesis on on-line post-column assay formats. As structure
elucidation has become an integral part of state-of-the-art HRS platforms, structure
elucidation by MS and NMR is introduced with a special attention to the analysis of
mixtures. Building on the introduction of these concepts, Chapter 2.1 describes how to set
up a state-of-the-art HRS platform and a successful HRS campaign. It gives practical tips
on optimization and validation of such platforms, warns of common pitfalls, and introduces
specialized concepts of data analysis.
Chapter 2 is dedicated to the development of HRS assays for the drug targets encountered
in this thesis, being p38 mitogen-activated protein kinase α (p38α) and soluble epoxide
hydrolase (sEH). The former is widely investigated as anti-inflammatory and analgesic
target, and the latter presents an emerging target for therapy of inflammation, pain and
cardiovascular disease. Chapter  2.2 describes the development of the first reported on-
line post-column bioaffinity assay for an enzyme target. Decreased stability as compared
to more robust protein targets presented a challenge and the generally high lipophilicity of
p38α inhibitors inspired novel solutions. While for sEH a more classical approach, based
on homogeneous detection of a fluorescent product, was chosen, the benefits of HRS in
the analysis of metabolic mixtures catalyzed the development of this assay [Chapter 2.3].
Chapter  3 deals with the integration of the innovative synthetic methods described in
Chapter 1.1.3. Chapter 3.1 reports on the direct integration of flow-through EC into the HRS
setup described in Chapter  2.2. This promises superior reproducibility of the combined
modification, separation, affinity assessment, and structure elucidation approach. As a
significant number of samples either lacking conversion or showing redundant product
profiles were expected, a suitable pre-screening preceded all HRS analysis in Chapter 3.
Implementation of LC–MS for pre-screening (Chapter 3.1) provides more information, but is
more resource intensive than (UP)LC–UV (Chapter 3.2 and 3.3). The compound modification
with BM3, described in Chapter  3.2, offered the largest potential sample variation due
to the number of tested BM3 mutants. Therefore, UPLC–UV pre-screening was applied.
After selection of some favorable biocatalysts by HRS, BM3 catalyzed production was
scaled up and the main products further probed for structural details and IC50-values by
NMR and HRS, respectively. Chapter 3.3 expands the combined modification, separation,
affinity assessment, and structure elucidation approach to the application of chemical
(hydrogen peroxide) and photochemical (visible light) conversion. It also compares the
different techniques, identifies strengths and weaknesses, and discusses the usefulness of
30 31
the approach in lead optimization and generation of metabolite standards.
Chapter  4 is dedicated to structure analysis in HRS. While Chapter  4.1 deals with MS
1 as an integral part of HRS platforms, Chapter 4.2 explores an interesting solution to the
sensitivity gap between HRS and the desirable structural refinement by NMR. Chapter 4.1
1
describes in detail the structure elucidation of the kinase inhibitor drug-like lead libraries
by MS. On the basis of examples, general aspects related to MS structure elucidation in
HRS are discussed as well as interesting observations of fragmentation pathways and gas-
phase ion and radical chemistry. Chapter 4.2 shows the evaluation of EC hyphenated via
SPE to stripline-NMR, the latter being a novel, miniaturized NMR approach with nanoliter
sample volume detection. Next to compatibility with analytical scale samples, the integration
achieves advantages in the detection of reactive products, as demonstrated in Chapter 3.1.
Finally, Chapter  5 describes the quantitation and structure elucidation of APIs and their
conversion products by high-temperature liquid chromatography (HTLC) – inductively
coupled plasma (ICP) MS (HTLC–ICP-MS) and HTLC–ESI-MS, respectively. Although,
ICP-MS has not been employed in HRS so far, the technique is an interesting candidate
for integration into novel HRS platforms. The integration of analyte quantitation into HRS,
for example by ICP-MS, would complete the information package desired in many types
of studies [1]. The low and constant organic modifier content of HTLC is beneficial for both
ICP-MS [Chapter 5] and HRS assays [2], promising an interesting synergy. We report the
sensitive quantitation of bromine or chlorine containing conversion products produced by
HLM, BM3 and EC from two kinase inhibitors, including a complete mass balance for EC
derived samples. Furthermore, ESI-MSn structure elucidation was achieved and correlated
to the quantitation results.

References
1. Campbell JL, Le Blanc JC (2012) Using high-resolution quadrupole TOF technology in DMPK
analyses. Bioanalysis 4 (5):487-500
2. de Boer AR, Alcaide-Hidalgo JM, Krabbe JG, Kolkman J, van Emde Boas CN, Niessen WM,
Lingeman H, Irth H (2005) High-temperature liquid chromatography coupled on-line to a
continuous-flow biochemical screening assay with electrospray ionization mass spectrometric
detection. Anal Chem 77 (24):7894-7900

32 33
Section 2 Chapter 2.1

High-resolution screening A practical guide to high-


platforms resolution screening
In this chapter, we want to shed light on the different aspects that are involved in a success-
ful high-resolution screening (HRS) campaign. We limit the discussion to homogeneous
on-line post-column assays, but many of the aspects are interesting to users of at-line as-
says as well. As discussed in Chapter 1.1.2, heterogeneous assays are very laborious and
should thus be avoided. We use certain terms like bioassay or biochemical assay in the
way defined in Chapter 1.1.2 despite their much broader meaning. The most obvious re-
2.1 quirement for HRS is a suitable, robust and sufficiently sensitive analytical platform. There-
fore, the setup of such a platform is the first topic [Chapter 2.1.1]. It includes practical tips 2.1
to design a state-of-the-art analytical system for HRS. The second part [Chapter 2.1.2] is
dedicated to data analysis which needs special attention due to the innate complexity of the
data obtained in HRS. Furthermore, certain concepts in HRS data analysis, some of which
are still under development, are just a bit different than in high-throughput screening (HTS)
or in conventional chromatographic detection. These concepts are needed to fully harness
the advantages of HRS platforms over HTS platforms, but awareness of sources of error is Figure 1: Example of an HRS setup. A typical single assay HRS platform is depicted like
critical, especially where the concepts are not fully developed yet [Chapter 2.1.2]. Screen- the ones developed in Chapters 2.2 and 2.3, where detailed descriptions can be found.
ing campaigns at the scale realized in HTS have rarely been realized for HRS (except [1]), Fluorescence is probably the most widely used bioassay detection in HRS, but other tech-
in fact to the best of our knowledge none including post-column HRS assays. Therefore, niques such as fluorescence polarization have been employed as well. When using MS
many lessons remain to be learned. for both structure and affinity measurements, these two detections are usually combined
in one instrument [2]. As discussed in Chapter 1.1.4.a), MS instruments offering both frag-
2.1.1. The high-resolution screening platform mentation as well as high mass accuracy provide the most useful data for structure eluci-
dation.
An HRS platform consists of four essential parts, as exemplified in Figure 1. At the core is
a separation setup which allows the different analytes of a mixture to be detected individ- the compatibility has to be achieved in the step in which the target protein is added, usually
ually by separating them in time. In order to allow two parallel detection techniques, one the first, as once unfolded protein molecules often remain denatured even after additional
for structure and one for effect analysis (affinity, activity or functional), the effluent of the dilution. For example, the sEH bioassay features a ca. 1:11 dilution in the first enzyme
separation step has to be divided. This is achieved by a flow split in the second part. The mixing step, but only a 1:0.15 dilution when the substrate is mixed in [Chapter 2.3]. In the
third and fourth part constitutes the two parallel detection techniques being the bioassay p38α bioassay, the influence of the gradient on the baseline is much more pronounced, be-
detection and mass spectrometry (MS), respectively. The bioassay detection reports the cause the first mixing step is 1:5 though the total mixing dilution (DM) is with 1:9 similar that
effect, by which we mean the bioactivity or bioaffinity towards the target protein. MS gives in the sEH assay (Attention: The baselines are straightened by background subtraction.)
information about the chemical composition and structure of the analytes. These four stag- [Chapter 2.2].
es are scrutinized subsequently. However, dilution is often an important challenge in HRS. Dilution reduces the final con-
centration at the point of detection and thereby reduces the sensitivity of the HRS platform.
2.1.1.a) Separation There are two dilution steps involved for the bioassay detection. The first one is chromato-
We identified the purpose of the separation step as being the separation of the different graphic dilution (DC). It is a result of diffusion and convection which transforms the initially
analytes in a mixture on the time scale. In other words, we seek to obtain a virtual chro- homogeneous injection plug into a concentration gradient by mixing with the solvent flow.
matogram, virtual because we do not (necessarily) subject the eluent to a detection method The result is the (nearly) Gaussian signal distribution so typical for chromatographic sepa-
yet. An efficient separation is desired meaning as many fully separated compounds in as rations. Depending on the interplay between injection volume (Vi), flow rate in the bioassay
little time as possible. Thus, standard state-of-the-art chromatographic or electrophoretic detector (uBio) and void volume, the maximum concentration of the analyte is diluted and
equipment should be utilized. While this point is of course true for any separation, there are the signal height is reduced. Though we can consider increasing the ratio of Vi to uBio,
significant differences to separations with more classical detectors like ultra-violet/visible this has a negative influence on the separation due to the consequential increase in peak
spectrophotometers (UV) which can mainly be attributed to the very specialized bioassay width. The second dilution DM was already mentioned above. It is exactly the same dilution
detection. We focus on high-performance liquid chromatography (LC) as other separation that appears when sample and reagents are mixed in a beaker or well plate, for example
techniques, e.g., incorporating electrophoretic separations, are still rare [3]. in HTS. DM exclusively occurs in the bioassay detection. Firstly, electrospray ionization
The most important stepping stone is the innate incompatibility of many classical LC mobile (ESI) MS does not usually need reagents or suffers as much from incompatibility with LC.
phase constituents (and flow rates) with the bioassay detection. The main reason for this Secondly, even if a makeup flow is desired, the reduced ionization efficiency at higher flow
incompatibility is the unfolding/denaturation of the target protein which is triggered by ex- rates can be more than compensated by suitable additives and/or an increased organic
cessive concentrations of organic modifiers after post-column mixing of effluent and protein modifier concentration. We have a more quantitative look at dilution in the respective data
[4]. Additionally, extreme acid, salt or base content or immiscibility of non-polar solvents analysis section (Chapter 2.1.2.a)). Suffice it to say for now that total dilution is usually in
with the aqueous buffers used in the bioassay detection could interfere when using for the order of 50 to 100 times, therefore thus far impairing the HRS measurements of low
example ion-exchange [5] or normal-phase LC, respectively. Fortunately, standard RP-LC concentration samples, for example from clinical or animal studies.
solvents like acetonitrile and especially methanol usually only produce unfolding at higher We have discussed compatibility of the separation with the biochemical detection, but
concentrations [6]. Thus, compatibility can often be achieved by suitable dilution of the compatibility with MS is equally important, though usually easier to achieve. Surely, it is
eluent with the bioassay reagent solutions. If several reagent addition steps are involved, superfluous to recap here the hyphenation of HPLC and MS as there are excellent books
and reviews on that topic [7,8]. However, one has to be aware of the many mobile phase
36 37
additives that are used to enhance ionization efficiency. For example, formic acid (FA) is 2.1.1.b) Flow splitting
often used in concentrations of 0.1% to 1% (v/v) to enhance ESI+ efficiency. FA, like any Flow splitting has two main functions: 1) to allow the parallel use of several detectors, and
other acid, salt or base, can result in two major interferences with the bioassay. Firstly, 2) to control the flow rate at which the eluent is provided to either detector. We first discuss
the pH of the bioassay which is crucial for the protein-ligand interaction can be affected. principles behind flow splitting and the attributes of a good flow split. Later, we go into detail
Secondly, interaction of any kind, for example ionic or hydrophobic, with the protein can on how to optimize the split ratio.
denature its structure. This adds to the influence of the organic modifier and might lead Flow splitting is achieved by providing more than one exit for the eluent after the separation
2.1 to more unfolding. In Chapter 2.2, for example, even 0.1% (v/v) FA in the mobile phase in
combination with methanol caused unacceptable protein unfolding. In this case, reducing
step. The total flow rate applied during the separation (uLC) then divides itself according to
the resistance encountered. That means that the flow rate towards each exit (e.g. uBio and 2.1
the mobile phase content of FA to 0.01% (v/v) resulted in a sufficient compromise between uMS) is such that the resistance or, in LC terms, the backpressure experienced as a conse-
bioassay compatibility and enhancement of ESI+ efficiency. quence of this flow rate is the same in all lines. One way to make a simple split is to take
The influences on the bioassay described above are less critical with isocratic elution LC, a connector with 1+N connections where one is connected to the LC column and N is the
because they are at least constant. When gradient chromatography is used in HRS, this number of desired exits. The exits are then fitted with tubing of various dimensions. When
leads to changing assay conditions during the chromatographic run, due to the changing calculating these dimensions, the first decision to be made is the operating pressure of
modifier content. Mainly, the assay window (the maximum signal) is reduced with increas- the split. On the one hand, it should be high enough to compensate for post-split pressure
ing protein unfolding. Furthermore, in assays based on signal reduction, the high signal differences and variations. For example, at an operating pressure of 50 bar, a difference
baseline decreases as a consequence of protein unfolding which complicates peak detec- of 1 bar in two lines already causes 2% change in split ratio while at an operating pressure
tion and integration. On the one hand, this can be prevented by increasing DM to the point of 5 bar the same interference renders the split effectively useless because of a 20% er-
where the change in influence is negligible compared to the noise and in return accepting ror. On the other hand, the pressure from the split adds to the demand on the LC system
the accompanying decrease in sensitivity. On the other hand, the changing assay window, and should therefore not be excessive. While especially with analytical and microbore LC
as long as it sufficiently large over the whole gradient, does not prevent effect detection columns the optimal flow rate for separation is often only reached at backpressures over
nor does it significantly change measured IC50 values. Additionally, the increased modifier 100 bar, standard LC equipment has an upper limit between 200 and 400 bar. Pressures of
concentration reduces tailing and precipitation of excessively hydrophobic compounds in 40 to 60 bar in the flow split have proven to be a sensible compromise.
the bioassay. The tradeoff here is the increased protein concentration which ensures a suf- Of course, the pressure is not only dependent on split dimensions but also on solvent
ficient assay window at higher modifier concentrations. Issues with (manual and automatic) viscosity and total flow rate. Solvent viscosity is especially tricky to handle as it changes
peak detection and integration can be minimized by subtracting a blank gradient run from during a chromatographic gradient. However, this change does not influence the split ratio.
every bioassay chromatogram [Chapter 2.2]. Another possible interference of the gradient, For ease and safety of calculating and testing a split, it is useful to employ water as solvent.
which has to be taken into account during assay development, is a change in detection However, it is always necessary to check that the pressure is still sufficiently high for a sta-
properties of the analyte or tracer (in the bioassay detection), such as fluorescence wave- ble split ratio at the lowest viscosity encountered in the gradient and that at the highest vis-
length, quantum yield or ionization efficiency. cosity the combined backpressure of column and split does not exceed the system limits.
Two solutions have been offered to combine gradient chromatography with bioassays The uLC is determined mainly by the separation and by MS compatibility, the latter because
aiming to eliminate the drawbacks, i.e., a counter gradient system and high-temperature the majority of the flow is usually directed towards this detector. Though it would be pos-
LC. The counter gradient system is an intricate setup which splits off half of the LC flow sible to add a waste line to the split in order to divert excess flow, the split is the part of
pre-column to a storage unit and releases it post-column in reverse order during the next the HRS system most liable to failure, the possibility of which increases with the number
run [6]. Alternatively, a second gradient LC system can be coupled post-column with which of exits. Additionally, splits with a larger number of exits are also increasingly complicated
the mirror image of the LC gradient is pumped [9]. As a result, the solvent entering the bio- to prepare. Therefore, it is often preferred to match the column dimensions to the MS ion
assay is always approximately a 50:50 mix of both LC solvents. The baseline shift during source to achieve compatibility without additional splitting. The influence of the split ratio on
gradient elution is reduced with only a 2-fold dilution. However, the system adds quite the decision for uLC is limited. The bioassay is usually carried out with a concentration-de-
some complexity to the HRS platform. Additionally, it is limited to symmetrical gradients pendent detector, for example a fluorescence detector. However, the split ratio has no
and switching between two gradient programs requires at least once the gradient run time. direct influence on the concentration of the analyte in the fraction of eluent that enters the
Alternatively, high-temperature liquid chromatography (HTLC) employs isocratic solvent bioassay. The fraction of total injected analyte mass entering the bioassay, which is equiv-
elution in combination with a temperature gradient. As solvents change their polarity with alent to the split ratio, is of interest merely in samples considered mass limited with respect
temperature, for example water reaches the polarity of methanol at around 200°C, HTLC to the high absolute sensitivity of both HRS detectors [12].
can very effectively replace organic modifier gradients in RP-LC. Because the effluent is There are also commercial solutions available for flow splitting, which operate by the same
cooled post column, there is no influence of the gradient on the bioassay. Though tempera- principles. In some variants, the split ratio is adjustable within a limited range. However,
ture stability of the analytes can be an issue, HTLC offers great advantages in combination when using a costly commercial flow splitter, the integration of a sufficiently fine particle
with bioassays [10]. In some cases, the solvent can be simplified down to pure water with- filter between column and split is advised as clogging of the split is a frequent problem es-
out compromising the separation allowing minimization of DM [11]. However, adsorption of pecially in matrix intensive samples.
hydrophobic analytes in the bioassay and MS sensitivity can become issues at these very
low modifier concentrations. 2.1.1.c) Bioassay
In conclusion, the composition of the mobile phase is a compromise between separa- The flow rate directed towards the bioassay (uE) is a result of the desired DM and the flow
tion efficiency, ionization efficiency and compatibility with the bioassay. The choice of the rates at which the bioassay reagents are mixed in (uR1+uR2) (see Equation 1). This is ex-
stationary phase is thus often limited by these restrictions. The flow rate and column di- emplified with values taken from the sEH HRS platform in Equation 1 [Chapter 2.3]. uBio
mensions are discussed in the following paragraph as this is, via DM, closely linked to the is preferred to be as low as possible to minimize reagent consumption, while the ratio of
concept of flow splitting. uR1 to uE should be large enough to prevent protein unfolding (see Chapter 2.1.1.a). Both
38 39
are important as the purified protein often makes up for the bulk of the running cost of the volume are also meticulously managed. Minimal void volume connectors and T-pieces
HRS platform. There are two factors which limit minimization of the reagent flow. The flow should be utilized and special attention has to be directed to avoiding ill-fitted connections.
rate needs to be exceptionally stable which becomes an increasing technical challenge the In addition, excessively large spectrophotometric detection flow cells compared to uBio are
lower the flow rate is, which also sets a limit to the ratio of uR1 to uR2. Additionally, uBio has a main cause of peak broadening [14]. From post-column derivatization, the coiled open
to match the dimensions of the optical detector used in the bioassay part to avoid extensive tubular reactor (coiled reactor) was adopted to reduce peak broadening. It is simply a spe-
peak broadening in the detector cell. Thus, miniaturization of the bioassay by employing cially ‘knitted’ PTFE tubing which induces secondary flow patterns thus increasing radial
2.1 micro- or even nano-fluidic instrumentation promises significant cost reduction [13]. mass transfer and reducing peak broadening [15]. By increasing the radial mass transfer,
the coiled reactor also contributes to the mixing of eluent and reagents. Other contribu- 2.1
tors to the deterioration of the separation are interaction between the reactor material and
Equation 1 the analytes. In PEEK and PTFE reactors, these are mainly hydrophobic interactions. We
therefore recently proposed to use chemically modified fused silica tubing for highly lipo-
philic analytes [Chapter 2.2]. Chemical modification is necessary, as the use of untreated
One strategy frequently employed to stabilize the reagent flow is the use of pulse dampen- fused silica results in strong dipole-dipole or ionic interactions of the free silanol groups
ers. A main contribution to flow instability is the sinus-shaped fluctuation resulting from the with nitrogen containing moieties often found in drug-like molecules. Instead, by chemical-
work cycle of the pump pistons. This is true provided the appropriate measures (solvent ly modifying the fused silica, its surface properties can be tuned to minimize interactions
degassing and regular seal maintenance) have been taken to prevent the formation of air with the respective analyte. For example, polyethylene glycol (PEG) modified fused silica
bubbles in the solvent delivery system. The pulses are smoothened by passing the solvent tubing has been successfully used for highly lipophilic drug-like molecules [Chapter 2.2].
over an oil-supported, impermeable membrane in the pulse dampener. For its function, it is A disadvantage of the fused silica tubing is that its low flexibility does not allow knitting it
necessary to apply high pressure to the solvent which is achieved by a restriction capillary in the same way as the PTFE tubing. Therefore, depending on whether void volume or
providing 100+ bar backpressure. adsorption is the main cause of peak broadening, a choice must be made between coiled
We can gain distinct advantages by delivering the reagents indirectly instead of pumping PTFE or straight fused-silica reactors until such time when fused-silica reactors with the
them through an LC pump. This can be achieved by running the pump on deionized water knitted-coil geometry become available. A third option, which however has been applied
which is passed through a pulse dampener and restriction capillary to generate a stable only in miniaturized assay formats so far, is the use of glass microfluidic chips [13,16]. Like
solvent flow which in turn operates a superloop. A superloop is a plastic covered glass cyl- fused silica tubing, they can be deactivated by chemical modification [13]. On top of that,
inder. Both ends are sealed watertight but provide a standard LC connection. The cylinder microfabrication of chips offers a lot of freedom in designing the flow paths. The introduc-
also hosts a moveable, water tight piston. On the inlet side, the water enters the superloop tion of hair-pin curves prevents a laminar flow profile [17], and thus enables efficient mixing
and pushes the piston. On the outlet side, the reagents are stored. By the movement of and avoids excessive peak broadening (see above).
the piston, the reagents are pushed out at the same flow rate as the water enters. The For optimal performance, the inner diameter (I.D.) of the tubing should match the flow rate
reagents are then mixed with the LC eluent. By using this setup, we save reagent solution applied. Additionally, this results in a large surface-to-volume ratio which is favorable for
as we need only fill the transfer tubing from superloop to mixer and not the significant void the heat transfer [15]. This tight and rapid temperature control is another advantage of the
volume of an LC pump with reagent. By keeping the void volume of the transfer tubing low, HRS over a classical HTS system and a second reason why shorter reaction times can be
the equilibration time, needed to achieve a stable assay baseline, is also shortened. In ad- achieved [Chapter  2.3]. Furthermore, incubation, especially for longer times at elevated
dition to saving expensive reagents, the setup reduces maintenance and increases lifetime temperatures, in HTS results in evaporation, which is a major source of errors in HTS. Due
of the pump as contact with chemical and/or biological reagents is avoided. The superloop to the (virtually) closed system, this is no issue in HRS. Of course, minimizing the I.D. is
is also easily cooled by placing it in an ice bucket. This allows to run an HRS system for limited practically by the pressure limits of the superloop and economically because the
days, depending on the stability of protein or reagents, without interruption. length increases with the square of the diameter reduction. An I.D. around 250 µm pres-
There are a number of measures we should consider to achieve an optimal performance ents the most common compromise, but longer incubation times might require 500 µm or
of the superloop. The piston should move as freely as possible within the superloop. Thus, 750 µm I.D. tubing.
regular greasing is essential. While filling the superloop with reagent solution and water, air We have now discussed the delivery and mixing of a reagent with the column effluent.
bubbles should be avoided. These increase fluctuations in the outlet flow as they compress However, usually, there are at least two different reagents needed. On one hand, the en-
and decompress every time the piston movement is slightly hindered. Because prevention zyme or receptor whose interaction with or manipulation by the analytes is studied, and
of air bubbles is not always possible, the superloop should be stored with the inlet slightly on the other hand, the substrate or tracer indicating inhibition of or affinity with the target
elevated. This at least prevents artifacts from release of the bubbles into the reactors during protein. These two reagents might be delivered in one [13] or two separate steps [Chap-
the analysis. The specific described superloop type has a pressure limit of around 10 bar. ter 2.2 and 2.3]. Delivering protein and tracer with the same superloop reduces the com-
Mixing of the eluent with the reagents is achieved with a T-piece. The mixing in an on-line plexity and cost of the system. Whether the reagents can be pre-mixed, depends on the
post-column bioassay is highly efficient, especially when coiled reactors are used which bioassay principle. For example, it is obviously not possible for bioactivity assays. Due to
is discussed later. This is one of the major advantages over well plate or test tube based the short incubation times typical in HRS (usually ≤5 min), affinity assays can be negatively
assays and the reason why shorter incubation times can be achieved at similar reagent influenced as well. While binding to the protein is usually quite fast, competition reaches
concentrations [Chapter 2.3]. equilibrium significantly slower [18]. Therefore, it is often favorable for the sensitivity of the
Although, being essentially only a simple piece of PEEK, PTFE or fused silica tubing, affinity/activity detection to make a first mixing and incubation step of the protein with the
much attention has to be devoted to the reactors in post-column bioassays. One reason eluent, thus allowing fast association with the analytes, and then to introduce the substrate
is the fact that we purposefully introduce additional void volume between separation and or tracer. Nonetheless, for binders with slow association kinetics (small kon), which do not
detection. This necessarily leads to reduced separation efficiency by peak broadening. reach equilibrium in the first incubation step, concentration sensitivity of the affinity mea-
The efforts described in the following are only justified if other prominent sources of void surement is lowered and IC50 values are underestimated. This issue may also be observed

40 41
in bioactivity assays. 2.1.2 Data analysis
Of course, as in any biological assay, the post-column bioassay should be buffered and
contain the appropriate co-factors, for example NADPH or Mg2+. Sometimes, reagents like
BSA are added to improve protein stability. On top of that, chemicals may be added which
2.1.2.a) Differences between HRS and HTS
help to prevent adsorption of analytes, proteins and substrate/tracer to the coiled reactors.
Influence of shorter incubation times
This presents a major challenge, especially due to the large surface-to-volume ratio in HRS
As the incubation time is generally limited to max. ~5 min by peak broadening, incubation
2.1 (see above). Often, reagents which are classically employed to counter unspecific binding
are used to this end. A few examples are ELISA blocking reagent, PEG or lactic acid. These
times are often much shorter in on-line bioassays than in HTS assays. Note that this is
not applicable to HRS assays based on high-resolution fractionation, as in such cases the
2.1
have the additional advantage of preventing unspecific binding to the target protein, but
bioassay is essentially decoupled from the LC, and thus does not have to take place within
their influence on protein stability has to be carefully monitored.
the LC timescale. One of the consequences is that competition between the analytes and
Detection of substrate conversion or tracer competition is most often achieved with a chro-
the tracer/substrate in on-line bioassays is more likely to be under kinetic rather than under
matographic fluorescence detector. When using a spectrophotometric LC detector, a main
thermodynamic control (compare Equation 2). This can have an influence on the detected
source of interference are air bubbles in the detector cell. These can be effectively prevent-
IC50 values and the sensitivity of the assay. In the two superloop setup, this creates a bias
ed by creating backpressure at the detector outlet, either by sufficiently narrow I.D. waste
towards binders/inhibitors with a slow dissociation kinetic (koff). However, as current opin-
tubing or by using commercially available backpressure regulators. Herein, the pressure
ion sees slow koff as favorable due to a link with better PK/PD properties, this bias seems
limits of the detector cell as well as of the superloops have to be carefully observed to
acceptable [21]. When looking to association kinetics (kon), most binders/inhibitors reach a
prevent damage to the system. However, usually a pressure excess of only 1–4 bar is
significant amount of binding in 30 s of pre-incubation. The sensitivity of the assay is there-
sufficient.
fore only negatively influenced in binders/inhibitors with a very slow kon.
In most cases, fluorescence detection (usually of a tracer) is preferred because it is superi-
or to other detectors in combining sensitivity, selectivity and linearity, while still being appli-
cable to a large number of molecules. Sensitivity is very important as it is directly related to
Equation 2
reducing reagent consumption and therefore the assay cost. Selectivity is a prerequisite for
any homogeneous assay, as the detection should not be disturbed by other bioassay con-
stituents present [19]. For example, in an activity assay with fluorescence product detec-
Slow assays might not be possible, but on-line assays usually allow quite a time reduction
tion, the substrate should not show similar fluorescence properties [Chapter 2.3]. Linearity
from HTS.
is also important, but rarely a hurdle. Two orders of magnitude linearity allow the detection
of 2–200% binding or activity which is more than sufficient to obtain good IC50 curves for
The assay signal
both inhibitors as well as agonists/ligands.
One difference in data analysis between HRS and HTS is the calculation of the signal which
is directly linked to the origin of the reference. In HTS, the reference is represented by the
2.1.1.d) MS detection control sample which gives the assay readout without any observable effect. Replicates
As elaborated in Chapter  1.1.4.a), structure elucidation by MS has become an integral produce a mean and standard deviation (SD) of the control, µC and σC, respectively. This is
part of HRS. HR-MS instruments are preferred as accurate analyte masses and superior essentially an external reference. In another experiment, mean and SD of the sample are
full-spectrum sensitivity can be obtained. The strength of HRS lies in the analysis of mix- obtained, µS and σS, respectively. The signal is then calculated by subtracting the means
tures of unknown compounds, and this application requires full-spectrum acquisition in- |µS–µC|. In HRS, no control sample needs to be measured. The baseline of the on-line bio-
stead of SRM or neutral loss methods. In addition, firstly, the limit of detection with respect assay readout provides an easily accessible and highly relevant internal reference for each
to the injected concentration is usually lower for drug-like molecules in the MS response individual sample. Thus, the signal in HRS is identified as the peak height (|µH|). Therefore,
than in the bioassay, partly due to DM. Secondly, the data acquisition speed of the MS contrary to HTS, changes in the background do not influence the signal which results in a
instrument is not a limiting factor. Therefore, the full-spectrum speed and sensitivity of lower SD of the peak height (σH) compared to σS. However, changes in the (theoretical) sig-
HR-MS instruments is sufficient, and the advantages of (triple) quadrupole MS in speed nal at full effect, the assay window, are equally uncorrected. The variation of the reference,
and sensitivity of targeted analysis are irrelevant to HRS. As for related compounds, much also called the noise, is usually calculated differently in HRS than in HTS (see σC). Instead
structure information is obtained from fragmentation analysis. HR tandem MS instruments, of calculating the standard deviation of the baseline, the maximum deviation of the baseline
like quadrupole–time-of-flight (Q-TOF) or ion-trap–time-of-flight (IT-TOF) combinations, are signal is calculated which is more easily accessible. This is halved to account for positive
ideally used in HRS setups. The differences in the spectral data obtained from unit-reso- and negative deviation from the baseline. Though the noise is slightly overestimated in this
lution MS instruments as compared to HR-MS instruments have been discussed in Chap- way, this method is more robust in dealing with baseline changes due to gradient elution.
ter 1.1.4.a). The resulting differences in the depth of structure elucidation are plainly visible In bioassays, there is always a (spectrometric) signal which indirectly visualizes the under-
when comparing earlier HRS systems featuring unit-resolution MS [20] with the data re- lying bioactivity/bioaffinity. For correct measurement of this signal, the chromatographic de-
ported in Chapter 4.1. tector has to be within its linear range. However, the resulting concentration-signal depen-
In case the bioassay is also based on MS detection, an interesting synergy is the combina- dence is not linear due to the underlying biochemical interaction. If the detector response
tion of structure and affinity measurements within one MS instrument [2]. is linear, the typical sigmoidal dose-response curves result when plotting |µH| against the
logarithm of the concentration. However, the influence is not limited to the peak height. As
in any chromatographic peak, the analyte molecules are distributed according to a Gauss-
ian distribution because of the longitudinal diffusion of the initially homogenous injection
plug (Vi), each data point has its own analyte concentration. This results in a distortion of

42 43
the ideal Gaussian shape in the bioassay detection. Fortunately, the sigmoidal curve has MS structure elucidation
a significant pseudo-linear part, usually between 10 and 90% of the assay window, thus Most often HRS is used for the analysis of mixtures of related molecules. These are mix-
allowing the calculation of important peak parameters like the full width at half maximum tures of molecules that (often) have the same or similar core structures. They can be de-
(FWHM) (see Figure  4). However, if |µH| is outside this pseudo-linear range, the conse- rived, for example, from chemical or biochemical diversification approaches [Chapter 3] or
quences are much the same as if a classical signal would be outside the linear range. For they could be different molecules from one class or type of natural products [12,22]. The
example, if µH is above 90% of the assay window, the FWHM increases as the flanks of the structure of individual compounds is identified by a combination of accurate-mass mea-
2.1 peak are still growing linear while the peak is not, the so-called overloading effect.
While the use of signal intensity (height) is natural to a pharmacologist, as HTS and other
surements and CID-fragmentation experiments (see also Chapter 1.1.4.a)). As HRS often
deals with unknown compounds, data-dependent acquisition (involving switching between 2.1
batch assays simply do not show peaks, separation scientists prefer working with peak ar- MS and MSn mode) is of key importance. MS instrument settings should be optimized us-
eas, as these are less sensitive to fluctuations in the separation than the peak heights are. ing an available model compound from within the compound class targeted at expected
The most prominent assumption is the detection of an underlying (pseudo-) linear phenom- concentrations. If data-dependent precursor selection fails, the afflicted experiments have
enon. However, in order to obtain reliable IC50 values, we would like to acquire sufficient to be repeated with manual precursor selection after data processing of the MS1 data. Pep-
data points in the non-linear parts of the dose response curve (this increases the quality of tides and small proteins might be elucidated (partly) by database comparison [12] or by top-
the data fitting), automatically violating that assumption. As a consequence, employing the down proteomics approaches. For small molecules, the first step is deducing the elemental
peak area of the bioassay response would create a relatively large, systematic y-error in composition of the parent ion. To this end, a range of elemental compositions pre-selected
the asymptotic region of the dose-response curve (see Figure 4). Due to the fact that the from prior knowledge of the analyte class, modification pathways, etc. is matched to the ac-
curve is asymptotic towards a y-value, the x-value error caused by distortion of the FWHM curate mass. Based on the stringency in matching accurate and exact mass and the range
calculation is far less influential. More importantly, while the y-value error in the peak area of elemental compositions allowed, one or more elemental compositions are yielded. For
increases continuously with increasing injected concentration, the x-value error in the cal- the fragmentation analysis, the elemental compositions can be limited to that of the precur-
culated concentration becomes increasingly meaningless with increasing x-values. In ad- sor ion. Specific neutral losses can then be used to identify functional groups and elucidate
dition, the FWHM of the pseudo-linear range might be used for the non-linear range thus their connectivity [Chapter 1.1.4.a)]. If several possibilities for the precursor ion exist, some
reducing the x-error for the most affected data points close to the pseudo-linear range. The wrong elemental compositions might be uncovered through proof by contradiction that is if
determination of IC50 values from the dose-response curve is discussed in more detail in no sensibly neutral losses connect the precursor ion with the product ions.
the next section.

Dilution calculations
As explained earlier, there are two types of dilution, DM and DC, whose product forms the
total dilution D. While DM represents essentially the same step as the mixing of the ana-
lyte solution with the assay reagents necessary in HTS, DC is unique to HRS. DM is sim-
ply calculated by dividing the total flow rate through the bioassay detector (uBio) by the
flow rate with which the analyte containing effluent enters the bioassay (uE). DC results
from the transformation of the injection volume (Vi) into the near-Gaussian distribution (see
Chapter 2.1.1a)) and can therefore be calculated by Equation 3. With DM and DC, we can
calculate the final concentration (cF) at µH from the injected concentration (ci), using Equa-
tion 5. It has to be remembered, however, that Equation 5 is derived for an ideal Gaussian
distribution which means that the results grow less accurate the more the shape of the
bioassay peak deviates from the ideal (see discussion in the previous section). Therefore,
this concept should be further developed in the future to include deviations from the ideal
distribution, for example, by including the peak tailing factor in a revised equation.

Equation 3

Equation 4

Figure 4: Theoretical dose response curve. The dots represent the curve which would be
Equation 5 expected when using peak height as readout. The triangles at high concentrations present
the systematic y-value error which would occur when using peak area instead of peak
height.
44 45
Structure-affinity matching results in complete inhibition or binding is a way to find µHmax which works in both activity
Matching the structure and the affinity of a molecule by matching its MS and its bioaffinity and affinity assays. To calculate the S/N of the assay window, we simply divide µHmax by
response starts by aligning the extracted ion currents (EIC) of the MS detection with the the noise. The Z’-factor as defined for HTS is given in Equation 6 [24]. Like the S/N, it re-
bioaffinity chromatogram (e.g. from the fluorescence detector). As both chromatograms flects the ratio between the noise and the assay window. However, in contrast to S/N, the
result from the same chromatographic run, one chromatogram is simply time-adjusted for Z’-factor accounts for the deviation in the signal as well as in the baseline, and thus for the
the stable retention time delay (Δr) which is a consequence of differences in post-column likelihood of both false negatives and false positives. To measure the Z’-factor, the mean
2.1 void volume. Variations in flow rate could cause Δr to vary. Therefore, the SD of the total
retention time could be used as a boundary [Chapter 3.1]. All MS peaks with retention times
(µ) and SD of positive (subscript C+; maximum signal) and negative control (subscript C–;
no signal) have to be determined. 2.1
(tr,MS) within this SD around the bioaffinity peak with tr,BIO are accepted as peaks potentially
having bioaffinity. This is evidently overcautious, especially considering all measures taken
to ensure flow stability in the bioassay, because the analytes usually spend significantly Equation 6
more time in the separation part than post-column. Additionally, variations in temperature
and solvent composition influence only the chromatographic retention time and not the
additional time spend post-column. However, other factors are usually more crucial for an With the adjustments discussed in Chapter 2.1.2.a), we derived the Z’Chrom-factor for HRS in
accurate peak matching (see below). In most HRS setups, the FWHM of the bioaffinity Equation 7 [Chapter 2.2]. μC+–μC- is identified with µHmax and consequentially, SDC+ changes
peaks is larger than the SD of tr let alone the deviation of Δr. Especially when dealing with to SDHmax. As discussed, the baseline represents the internal (negative) control and there-
overloading (see above) in either of the responses, peak maxima and consequently tr-val- fore SDC- becomes the noise.
ues can be difficult to identify. Therefore, we often mark both MS and bioaffinity peaks at
their onset rather than at their top, because this is better defined. Nonetheless, this may
result in different marks for the same compound depending on its concentration and thus Equation 7
in unstable retention time differences for different compounds, because the onset is further
away from tr in a broad peak than it is in a sharp peak. Consequently, the variation in peak
width (Δw) is another criterion for the accuracy in the peak matching and overloaded peaks HRS platforms have been successfully used to quantify the affinity/activity of analytes by
have to be even more carefully assigned. If peaks are assigned on the onset, it is more measuring their IC50 values. However, this is no core strength of HRS platforms as to
logical to use the width at 10% rather than FWHM. Δw can be deduced, for example, from this end they still rely on purified compounds as much as HTS platforms, due to the as
IC50 measurement curves during the validation and then 0.5•Δw can be used in the same yet missing capacity of HRS platforms to quantify unknown compounds (see Chapter 5).
way as described for the SD of tr above. The alignment of the two chromatograms and the Nonetheless, it is valuable to discuss the measurement of IC50 values in HRS platforms. In
matching via Δr can also be tested and possibly adjusted if a known affinity compound is the context of validation, good sigmoidal curves confirm that signal changes observed are
present. However, co-elution of other affinity compounds and overloading of either the MS indeed due to interaction of the analytes with the target and do not result from (non-spe-
or the bioaffinity response can influence this matching as well. cific) interferences or artifacts. If literature values measured with the same assay principle
are available, the actual IC50 values provide a validation. However, great caution is advised
2.1.2.b) Validation when comparing IC50 values as there are many pitfalls. Even seemingly identical assay for-
Validation is the final stage of every assay development, not excluding HRS systems. In mats can show major differences. A classic example is the use of different substrates in an
HRS, we usually start by validation of the bioassay and add the MS later for further vali- activity assay, but sometimes changes in buffer constituents might impact the interaction of
dation of the whole setup. A good first validation step is the measurement of a number of analyte and target as well. Additionally, kinetics might impact IC50 values when comparing
specific assay parameters which give a quick first quality assessment of the assay and if assays with largely different incubation times (see Chapter 2.1.2.a), Influence of shorter
necessary show potential for improvement. Simple measurements reveal all contributions incubation times) [Chapter 2.2] [18]. However, this is by no means an issue unique to HRS,
to the background signal as well as the assay window [Chapter  2.3]. An assay window although it appears more pronounced than in HTS. In terms of application, measuring IC50
(for calculation see below) with a signal-to-noise ratio (S/N) between 50 and 100 is an values with HRS sometimes offers an advantage as well. Due to the influences discussed
indication of a good-quality bioassay, because it allows detection (S/N≥3) of the flat part of in The assay signal [Chapter 2.1.2.a)], HRS assays are often either of higher quality at the
the sigmoidal curve between 5 and 10% signal and quantitation (S/N≥9) of (most of) the same protein concentration or necessitate a lower protein concentration to achieve the
pseudo-linear part of the sigmoidal curve between 10 and 90% signal [Chapter  2.2 and same quality. While the influence of the former on IC50 value quality is evident, the latter
2.3]. The former greatly enhances sensitivity of the bioassay, the latter is essential to ob- allows a better distinction of high-affinity binder/inhibitors [Chapter 2.2]. The reason is that
tain good-quality IC50 values. If sufficient S/N is not yet reached, the individual background half of the protein concentration (cP) is the theoretical lower limit of measurable IC50 values,
contributions provide a clue to which part still has to be optimized. For example, if by far the because below that limit the enzyme is simply titrated. For example, we have cp of 90 and
largest contribution comes from the tracer, decreasing the tracer concentration or increas- 450 nM in an HRS assay and in an HTS assay, respectively, and an analyte with an IC50
ing the enzyme concentration are obvious starting points. It should be mentioned here that of 45 nM. If we add the analyte at 45 nM, it interacts with 50% of the protein in the HRS
sometimes lower S/N for the assay window are preferred due to cost considerations or assay thus correctly giving an IC50 value of 45 nM. Note that also all analytes with higher
other limitations, excepting the loss in assay quality [13,23]. affinity yield this value. If we add the same analyte concentration to the HTS assay, it only
We quickly explain how two important benchmarks of bioassay quality can be calculated occupies 10% of the protein binding sites. Only with 225 nM of the analyte, we reach 50%
in an HRS setup, the assay window and the Z’ factor [24]. For both of them, we need µH at interaction thus the apparent IC50 in the HTS is 225 nM (0.5•cP). We should add that this
full inhibition or binding (µHmax) as well as the baseline noise. For activity assays, µHmax can advantage is more often observed in affinity assays as in activity assays the cp can be re-
often be found by comparing the signal at the incubation temperature and at 0°C (where duced by increasing the incubation time.
virtually no enzymatic reaction is observed). Injection of an analyte concentration which Similar to HTS, IC50 values are determined by measuring a series of concentrations and

46 47
fitting the resulting dose-response curve. To this end, the individual concentrations of the parking system for gradient liquid chromatography with online biochemical detection of serine
pure compound are injected into the HRS platform. This can be done either in flow injection protease inhibitors. Anal Chem 80 (17):6764-6772
analysis (FIA) mode accounting for the injection peak by subtraction of a blank injection 7. Niessen WMA (2006) Liquid Chromatography – Mass Spectrometry. Chromatographic Science
or by isocratic elution with minimal retention thus separating the injection peak. Then, the Series, Third edn. CRC Press,
8. Kostiainen R, Kauppila TJ (2009) Effect of eluent on the ionization process in liquid chromatog-
critical step is the calculation of cF by dividing the injected concentration ci by the two dilu-
raphy-mass spectrometry. J Chromatogr A 1216 (4):685-699
tion events (see Equation 5). The models and algorithms used for fitting the dose-response 9. Kool J, van Liempd SM, Ramautar R, Schenk T, Meerman JH, Irth H, Commandeur JN, Vermeu-
2.1 curve and calculating the IC50 are the same as in HTS. Theoretically, it is possible to de-
duce individual IC50 values by injecting a dilution series of a mixture. The repeatability of
len NP (2005) Development of a novel cytochrome p450 bioaffinity detection system coupled
online to gradient reversed-phase high-performance liquid chromatography. J Biomol Screen 10
2.1
IC50 values in HRS systems seems to be quite good, as can be judged by almost identical (5):427-436
values obtained for TAK-715 in different studies (81 nM (71 to 92 nM) [Chapter 2.2] and 10. de Boer AR, Alcaide-Hidalgo JM, Krabbe JG, Kolkman J, van Emde Boas CN, Niessen WM,
71 nM (36 to 140 nM) [Chapter 3.2]). Lingeman H, Irth H (2005) High-temperature liquid chromatography coupled on-line to a contin-
A good approach to test the complete HRS platform, and especially the structure-affinity uous-flow biochemical screening assay with electrospray ionization mass spectrometric detec-
correlation, is the analysis of mixtures of pure compounds, preferably containing both effec- tion. Anal Chem 77 (24):7894-7900
tors (inhibitors, ligands, etc.) and non-binders [Chapter 2.2] [13]. The first indication is that 11. Causon TJ, Shellie RA, Hilder EF (2009) High temperature liquid chromatography with monolith-
ic capillary columns and pure water eluent. Analyst 134 (3):440-442
the correct number of compounds is detected in the MS response and that only as many
12. Heus F, Vonk F, Otvos RA, Bruyneel B, Smit AB, Lingeman H, Richardson M, Niessen WM, Kool
signals as effectors are detected in the bioassay response. Then, the MS and bioassay J (2013) An efficient analytical platform for on-line microfluidic profiling of neuroactive snake
responses are matched blindly and the compounds are identified from the MS response. venoms towards nicotinic receptor affinity. Toxicon 61:112-124
Only the effectors should show bioaffinity. 13. Heus F, Giera M, de Kloe GE, van Iperen D, Buijs J, Nahar TT, Smit AB, Lingeman H, de Esch IJ,
Niessen WM, Irth H, Kool J (2010) Development of a microfluidic confocal fluorescence detec-
2.1.3 Conclusions tion system for the hyphenation of nano-LC to on-line biochemical assays. Anal Bioanal Chem
398 (7-8):3023-3032
14. Schebb NH, Falck D, Faber H, Hein EM, Karst U, Hayen H (2009) Fast method for monitoring
The strong link between molecular structure and individual biological effect, which is cre-
phospholipase A2 activity by liquid chromatography-electrospray ionization mass spectrometry.
ated by the integration of separation, bioassay and structure elucidation, is undoubtedly J Chromatogr A 1216 (27):5249-5255
the greatest potential of HRS. In addition, the (virtually) closed system, the efficient mixing 15. Engelhardt H, Neue UD (1982) Reaction Detector with Three Dimensional Coiled Open Tubes
and heat exchange, and the internal control offered by the baseline hold the possibility to in HPLC. Chromatographia 15 (7):403-408
achieve high-quality bioassays in the on-line post-column mode. Though throughput is 16. de Boer AR, Bruyneel B, Krabbe JG, Lingeman H, Niessen WM, Irth H (2005) A microfluid-
not a primary attribute of HRS, the data density intrinsic to the approach is so high that it ic-based enzymatic assay for bioactivity screening combined with capillary liquid chromatogra-
presents both a potential and a challenge. Similar to any hyphenated system, a challenge phy and mass spectrometry. Lab Chip 5 (11):1286-1292
is presented by finding a useful compromise between the complex requirements of each 17. Yamaguchi Y, Takagi F, Yamashita K, Nakamura H, Maeda H, Sotowa K, Kusakabe K, Yamasaki
individual analytical technique. However, such a compromise has been reached in various Y, Morooka S (2004) 3-D simulation and visualization of laminar flow in a microchannel with hair-
ways and sometimes even synergies become evident. Dilution is a key factor to compat- pin curves. AIChE Journal 50 (7):1530-1535
18. Pargellis C, Tong L, Churchill L, Cirillo PF, Gilmore T, Graham AG, Grob PM, Hickey ER, Moss
ibility, but at the same time one of the biggest hurdles for HRS sensitivity (with regard to
N, Pav S, Regan J (2002) Inhibition of p38 MAP kinase by utilizing a novel allosteric binding site.
amount and concentration LOD). We have shown that a great number of the (data analysis) Nat Struct Biol 9 (4):268-272
concepts, generally applied to bioassays, can be adapted to HRS, though some of these 19. Oosterkamp AJ, Villaverde Herraiz MT, Irth H, Tjaden UR, van der Greef J (1996) Reversed-phase
derived concepts would surely benefit from further refinement. Exploring the potentials of liquid chromatography coupled on-line to receptor affinity detection based on the human estro-
HRS can certainly lead to routine application in the future, especially if accompanied by gen receptor. Anal Chem 68 (7):1201-1206
continued technical development of the systems. 20. de Boer AR, Letzel T, van Elswijk DA, Lingeman H, Niessen WM, Irth H (2004) On-line coupling
of high-performance liquid chromatography to a continuous-flow enzyme assay based on elec-
trospray ionization mass spectrometry. Anal Chem 76 (11):3155-3161
References 21. Copeland RA, Pompliano DL, Meek TD (2006) Drug-target residence time and its implications
1. Annis DA, Athanasopoulos J, Curran PJ, Felsch JS, Kalghatgi K, Lee WH, Nash HM, Orminati for lead optimization. Nat Rev Drug Discov 5 (9):730-739
J-PA, Rosner KE, Shipps Jr GW, Thaddupathy GRA, Tyler AN, Vilenchik L, Wagner CR, Wintner 22. de Jong CF, Derks RJ, Bruyneel B, Niessen W, Irth H (2006) High-performance liquid chroma-
EA (2004) An affinity selection–mass spectrometry method for the identification of small mole- tography-mass spectrometry-based acetylcholinesterase assay for the screening of inhibitors in
cule ligands from self-encoded combinatorial libraries: Discovery of a novel antagonist of E. coli natural extracts. J Chromatogr A 1112 (1-2):303-310
dihydrofolate reductase. International Journal of Mass Spectrometry 238 (2):77-83 23. Retra K, Geitmann M, Kool J, Smit AB, de Esch IJ, Danielson UH, Irth H (2010) Development of
2. Derks RJ, Hogenboom AC, van der Zwan G, Irth H (2003) On-line continuous-flow, multi-protein surface plasmon resonance biosensor assays for primary and secondary screening of acetyl-
biochemical assays for the characterization of bioaffinity compounds using electrospray quadru- choline binding protein ligands. Anal Biochem 407 (1):58-64
pole time-of-flight mass spectrometry. Anal Chem 75 (14):3376-3384 24. Zhang JH, Chung TD, Oldenburg KR (1999) A Simple Statistical Parameter for Use in Evalua-
3. Chen J, Lee CS (2002) On-line post-capillary affinity detection of immunoglobulin G for capillary tion and Validation of High Throughput Screening Assays. J Biomol Screen 4 (2):67-73
zone electrophoresis. J Chromatogr B Analyt Technol Biomed Life Sci 768 (1):105-111
4. Kool J, Giera M, Irth H, Niessen WM (2011) Advances in mass spectrometry-based post-column
bioaffinity profiling of mixtures. Anal Bioanal Chem 399 (8):2655-2668
5. Schebb NH, Vielhaber T, Jousset A, Karst U (2009) Development of a liquid chromatog-
raphy-based screening methodology for proteolytic enzyme activity. J Chromatogr A 1216
(20):4407-4415
6. Schebb NH, Heus F, Saenger T, Karst U, Irth H, Kool J (2008) Development of a countergradient
48 49
Chapter 2.2

Development of an on-line p38α


mitogen-activated protein kinase
binding assay and
integration of LC–HR-MS

David Falck,1, Jon S. B. de Vlieger1, Wilfried M. A. Niessen1, Jeroen Kool1,


Maarten Honing2, Martin Giera1,*, Hubertus Irth1

1
LACDR-Division of Biomolecular Analysis, Department of Chemistry and Pharmaceutical
Sciences, VU University Amsterdam, The Netherlands
2
Department of Medicinal Chemistry, MSD Research Laboratories, Oss, The Netherlands
Abstract free technologies, like surface plasmon resonance [16] or isothermal titration calorimetry
[19], have been employed, which avoid additional reagents and the problems of molecular
A high resolution screening method was developed for the p38α mitogen-activated protein probes. Still, these techniques are complex and instrumental requirements and logistics
kinase to detect and identify small molecule binders. Its central role in inflammatory may limit their throughput. A relatively simple approach was used by Pargellis et al during
diseases makes this enzyme a very important drug target. The setup integrates separation the discovery of BIRB796 [10, 11]. The method is based on fluorescence enhancement
by high performance liquid chromatography with two parallel detection techniques. High and requires solely native p38 and the small molecule kinase inhibitor SKF. SKF shows
resolution-mass spectrometry gives structural information to identify small molecules fluorescence, which is strongly enhanced upon binding to the target enzyme. Therefore,
while an on-line enzyme binding detection method provides data on p38α binding. The high fluorescence intensity indicates a high concentration of the enzyme-tracer complex. A
separation step allows the individual assessment of compounds in a mixture and links compound showing affinity to the target enzyme will compete with the tracer thus reducing
affinity and structure information via the retention time. Enzyme binding detection was the concentration of enzyme-tracer complex and thereby reducing the fluorescence. This
achieved with a competitive binding assay based on fluorescence enhancement which has principle enables a competitive binding assay in a homogeneous setup, which is based on
2.2 a simple principle, is inexpensive and easy to interpret. The concentrations of p38α and the the strong difference in fluorescence intensity between the free tracer and the enzyme-tracer
complex. Because of its convenience and the possibility to use non-phosphorylated p38α
2.2
fluorescence tracer SK&F86002 were optimized as well as incubation temperature, formic
acid content of the LC eluents and the material of the incubation tubing. The latter notably as target, this technique presented a good basis for the development of a high resolution
improved the screening of highly lipophilic compounds. For optimization and validation screening (HRS) platform. HRS is defined by the combination of separation and screening
purposes, the known kinase inhibitors BIRB796, TAK715 and MAPKI1 were used among in an integrated system. Therefore, it enables the affinity assessment and the simultaneous
others. The result is a high quality assay with Z’-factors around 0.8, which is suitable structural characterization of individual compounds in mixtures [20, 21]. This feature makes
for semi-quantitative affinity measurements and applicable to various binding modes. HRS platforms especially attractive when complex mixtures such as metabolic incubations
Furthermore, the integrated approach gives affinity data on individual compounds instead [22], combinatorial mixtures [21] or natural extracts [23] are screened where dereplication,
of averaged ones for mixtures. affinity and structural information are crucial. In contrast to conventional high throughput
screening (HTS), isolation of individual compounds not showing affinity can be avoided.
Thus, the laborious development of high yield synthesis and isolation methods can be
Introduction limited to compounds showing the desired affinity. One common approach in HRS is the
use of on-line post-column assays, which have been recently reviewed by Shi et al [24].
Kinases are certainly among the most important target classes in contemporary drug
Colourimetry, fluorescence and mass spectrometry based readouts were developed in
discovery [1]. The mitogen-activated protein kinase p38 (p38) is one prominent example
both homogeneous and inhomogeneous formats. Homogeneous setups are easier to
of a drug-target kinase especially its most important isoform p38α [2, 3]. It is involved
apply, so their significance exceeds inhomogeneous setups in cases where background
in signal transduction pathways, transmitting signals through successive activation
fluorescence is of minor importance. However, HRS formats described so far are either
by phosphorylation [4]. Being one of the important mediators for a cell’s response to
activity based [21, 23] or target a receptor [20, 22]. We report the implementation of the
extracellular stimuli, this pathway presents a promising opportunity for drug intervention [5].
enzyme p38α as the first kinase to be targeted in an on-line post-column binding assay
Because p38α is heavily involved in inflammation processes, the envisaged diseases are
(HRS system). Furthermore, the screening of highly lipophilic molecules was greatly
mostly chronic inflammatory diseases like rheumatoid arthritis, psoriasis or Crohn’s disease
improved by the unprecedented use of covalently modified fused silica tubing as material
[6]. Several p38α inhibitors, which target these diseases, are currently in different stages of
for post-column incubation, resulting in improved affinity signals.
preclinical and clinical development up to phase II [7, 3]. Nearly all known kinase inhibitors
show interactions with the highly conserved ATP-binding pocket. Therefore, most early
kinase inhibitors, possessing only these interactions, exhibited poor kinase selectivity [5]. Materials and Methods
Although different systems of categorization exist, these will be called type I inhibitors. Type
I inhibitors used in this study are TAK715 [8], MAP kinase inhibitor 1 (MAPKI1) from Merck Materials
KGaA [9] and SK&F 86002 (SKF) [9]. The discovery of an allosteric binding pocket adjacent Human recombinant p38α, ORGX, BIRB796 and TAK715 were a kind gift of MSD research
to the ATP-binding site led to the discovery of inhibitors using both sites (type II). BIRB796 laboratories (Oss, The Netherlands). SKF and MAPKI1 were purchased from Merck KGaA
is a prominent example of this type of binding [10, 11]. Consequently, type  III binders (Darmstadt, Germany). ELISA blocking reagent was delivered by Roche Diagnostics
like 1-(3-tert-butyl-1-methyl-1H-pyrazol-5-yl)-3-(4-chlorophenyl)urea (pyrazolourea) are (Mannheim, Germany). Pyrazolourea was obtained from Ambinter (Paris, France). All other
targeting only this allosteric site [12]. The type IV inhibitors are not binding to either of the chemicals were from Sigma-Aldrich (Schnelldorf, Germany). Fused silica tubing (250 μm
two pockets, as has been discovered e.g. for GNF-2 binding to the Brc-abl kinase [13]. inner and 375  μm outer diameter) untreated or covalently coated with PEG, methyl or
The importance of p38α has led to a multitude of assay principles for the identification methyl/phenyl groups was also purchased from Sigma-Aldrich (Schnelldorf, Germany).
of small molecules targeting this kinase [14, 15]. Among the cell-free in vitro formats for 96 ‘flat’ bottom chimney well, black polypropylene microtiter plates from Greiner bio-one
p38α, those based on activity and those based on affinity can be distinguished. As readout (Alphen a/d Rijn, The Netherlands) were used. Methanol (LC-MS Grade) and formic acid
for activity based methods, scintillation [11] and fluorescence polarization [16] have been (ULC-MS Grade) were obtained from Biosolve (Valkenswaard, The Netherlands). Purified
used among others [14, 15]. These formats require expensive reagents (antibodies, etc.) water was generated with a Milli-Q academic from Millipore (Amsterdam, The Netherlands).
and rely on labels which can impair the usefulness of results, or they involve handling
radioactivity. In addition to these drawbacks, activity assays can, by definition, not employ Instrumentation
the non-phosphorylated kinase as target. This is the advantage of affinity based assays. 96 well plate experiments were performed on a Victor3 plate reader from Perkin-Elmer
However, scintillation [17], chemiluminescence [16] and fluorescence polarization [18] (Groningen, The Netherlands). The setup used for the on-line assay included two LC-20AD
suffer from similar reagent based drawbacks as mentioned above. Furthermore, some label and two LC-10AD isocratic pumps, a SIL-20AC autosampler, one CTO-20AC and one
CTO-10AC column oven, an RF-10AXL fluorescence detector, a CBM-20A controller and
52 53
an Ion Trap-Time-of-flight (IT-TOF) mass spectrometer equipped with an ESI source all
products of Shimadzu (’s Hertogenbosch, The Netherlands). HR-MS spectra were obtained
from m/z 200 to 650 switching positive- and negative-ion mode with 10 ms accumulation
time each. The curved desolvation line and the heating block were equilibrated at 200°C
and the voltages applied were 5 kV to the interface and 1.7 kV to the detector. Nitrogen
(99.9990%) was used at flows of 1.5 L/min and 10 L/min as nebulising gas and drying gas,
respectively. To allow accurate mass measurements, external calibration of the IT-TOF was
performed using TFA clusters.
Furthermore, two superloops and two pulse dampeners made in house were used.

Plate Reader Assay


2.2 A 20 mM TRIS-HCl buffer (pH 7.5), containing 10 mM MgCl2, 2 g/L PEG 6000 and 0.2 g/L
ELISA blocking reagent (TRIS buffer), was used in all experiments. All solutions of p38α
2.2
were prepared in TRIS buffer. Enzyme stock solutions were stored at –80°C until use.
During on-line experiments, the enzyme solutions were kept at 0°C. A stock solution of Figure 1: Scheme of the on-line setup. The system enables the separation of mixtures and
SKF (2 mM) was made in DMSO and further diluted with TRIS buffer. Solutions of inhibitors subsequent parallel detection of enzyme binding and accurate mass. The samples are injected (1)
were prepared in water:methanol 1:1 containing 0.01% formic acid from 2 mM or 5 mM into and separated (2) by an HPLC system. The eluent is split (3) between HR-MS and enzyme
stock solutions in methanol (DMSO in case of MAPKI1). Small molecules stock solutions binding detection. The fraction which enters the enzyme binding detection is mixed with the target (4)
and the tracer (6), and incubated after each mixing step (5 & 7). Finally, the fluorescence is measured
were stored at -20°C.
(8) as readout of affinity towards p38α. In parallel, the second part of the eluent is analyzed by HR-
For plate reader measurements, the wells were filled with 50 μL enzyme solution, 12.5 μL MS (9), delivering structural information to identify the small molecules tested.
inhibitor solution and 50 μL SKF solution in the given order. Fluorescence was measured
at 25°C with the wavelength of absorption and emission being 355±4 nm and 405±5 nm, consecutive measurements. To this end, a stepwise gradient was used starting at 100%
respectively. The sum of ten flashes was used as readout to improve performance. A and increasing the fraction of B by 15% approximately every 10  min. In the first run,
Saturation of a 90 nM solution of p38α with SKF was determined by using a concentration the baseline without inhibition was recorded. In the second run, MAPKI1 was added to
range from 40 nM to 2500 nM of SKF. The Z’-factor is a measure for assay quality. If not the SKF solution for full inhibition, reaching a final assay concentration of around 0.9 μM.
otherwise stated, it was calculated according to the original publication of Zhang et al [25]. By comparing these measurements, the assay window in relation to the different organic
A more detailed explanation is given in the results and discussion section. Measurements modifier concentrations can be estimated.
of the Z’-factor dependence on enzyme concentration were performed on three days with In order to assess the influence of the incubation tubing material, two measurements were
five repeats per day and a final concentration of 3  μM SKF. For full inhibition, TAK715 carried out, in which the volume and therefore the incubation time was kept constant: In
was added to a final concentration of 1  μM, while the control values contained only the the first experiment, polyether ether ketone (PEEK) of 250 μm and 170 μm inner diameter
corresponding amount (see above) of HPLC solvents. (ID), polytetrafluoroethylene (PTFE) of 250 μm ID and untreated fused silica of 250 μm ID
were compared by injecting a 2 μM solution of ORGX in FIA mode at 60% B. In a second
On-line Assay experiment, 250  μm ID fused silica tubing with covalent coatings of different polarity
HPLC solvents were water:methanol 100:1 (solvent A) and methanol:water 100:1 (solvent were compared. In these experiments, a 20  μM solution of ORGX was injected in FIA
B) both containing 0.01% formic acid. The instrumental setup described by de Vlieger et mode at 60% B. The coatings were PEG (polar), a mixture of phenyl and methyl groups
al [20] was used for the analysis with one enzyme binding detection line and the following (intermediate) or methyl groups only (non-polar).
modifications (cf Figure 1). HPLC was done either in flow injection analysis mode (FIA – The on-line Z’-factor was calculated from triplicate injections each of a control (blank)
where no separation column is present) or with a SunFire C18 column 30 x 2.1 mm, 3.5 μm experiment and a full inhibition sample on five different days. The full inhibition sample
particles (Waters, Milford, MA, USA) (Figure 1 - 2). 10  μL of the sample were injected was a mixture of 50  μM BIRB796, 20  μM ORGX and 20  μM TAK715 while the control
(Figure 1 - 1) into a flow of 113 μL/min. Elution was performed at 40°C either isocratic at sample was a 1:1 mixture of the solvents. The separation was carried out with the following
different methanol concentrations or using a binary gradient. Post-column, the flow was gradient: 0 to 1.0 min isocratic at 50% B, 1.0 to 12.5 min linear to 90% B, 12.5 to 14.5 min
split 1:9 (Figure 1 - 3), sending 100 μL/min to high resolution/mass spectrometry (HR-MS) isocratic at 90% B, 14.5 to 15.0 min linear to 50% B and 15.0 to 21.0 min isocratic at 50% B.
detection and 13 μL/min to the enzyme binding detection. To the 13 μL/min HPLC eluent, MAPKI1 and TAK715 were tested in a mixture with six drug molecules to simulate a
first 50 μL/min of a 200 nM p38α solution were added (Figure 1 - 4) and thereafter 50 μL/ screening situation. Carbamazepine, norethisterone, warfarin, phenylbutazone, celecoxib
min of a 1400 nM solution of SKF (Figure 1 - 6). These solutions were constantly released and diclofenac were used as test compounds without p38α affinity. TAK 715 was injected
by a linear setup of an LC-10AD pump, a pulse dampener with restriction capillary and a at 20 μM concentration, the others at 50 μM. The mixture was separated with the following
superloop each. Following each addition, the mixture was incubated in PEG deactivated gradient: 0 to 2.0 min isocratic at 50% B, 2.0 to 32.0 min linear to 70% B, 32.0 to 38.0 min
fused silica loops of 25 μL (Figure 1 - 5) and 20 μL (Figure 1 - 7), respectively. The incubation isocratic at 70% B, 38.0 to 38.5 min linear to 50% B and 38.5 to 50.0 min isocratic at 50% B.
loops were kept at 25°C in a second column oven. The resulting incubation times were For the measurements of IC50 values, isocratic elution was applied, taking care that the
24 s for the target-ligand interaction (Figure 1 - 5) and 11 s for the target-tracer interaction substance peak showed separation from the injection peak. TAK715 was eluted at 85% B,
(Figure 1 - 7). Finally, at concentrations of 90 nM p38α and 630 nM of SKF the fluorescence BIRB796 at 50% and MAPKI1 at 70%. Solutions of nine different concentrations for each
was measured with excitation at 355±7.5 nm and emission at 425±7.5 nm (Figure 1 - 8). inhibitor were prepared in 50% methanol, 50% water and 0.01% formic acid from 5 mM
The used detector settings were gain 4, recorder range 1 and sensitivity medium. stock solutions of TAK715 and BIRB796 in methanol or a 2 mM stock solution of MAPKI1
The influence of the methanol concentration on the maximum signal was assessed in two in DMSO. The following concentrations for each inhibitor were prepared: 50 nM, 100 nM,
54 55
200 nM, 500 nM, 1 μM, 2 μM, 5 μM, 10 μM and 20 μM for TAK715; 500 nM, 1 μM, 2 μM, concentration, depicted in Figure 2A, displays the asymptotic form of saturation behaviour.
5 μM, 10 μM, 20 μM 50 μM, 100 μM and 200 μM for BIRB796 and 200 nM, 500 nM, 1 μM, The final concentration of 630  nM SKF is selected to be slightly above the saturation
2 μM, 5 μM, 10 μM, 20 μM 50 μM and 100 μM for MAPKI1. These nine concentrations and value. This choice presents a compromise between avoiding deviations of the signal
a blank were injected in triplicate for every inhibitor. due to variability in the tracer concentration and maximising the sensitivity with regard to
To finalise the binding mode studies, the pyrazolourea compound was used as an example detection limits for binders. Namely, competition reduces this sensitivity with increasing
of a type III inhibitor. It was injected at a concentration of 100 μM and tested under isocratic tracer concentration.
conditions at 70% B. There are several factors mainly related to the HPLC separation system, which can have
a negative influence on the assay performance. Methanol, formic acid and the incubation
Results and Discussion temperature are the most prominent of these variables. Using typical HPLC conditions
such as 50% methanol and 0.1% formic acid at 37°C completely inhibited the fluorescence
Assay conditions optimization enhancement. As a result, these factors were carefully optimized and need to be controlled
2.2 Optimization was carried out in a 96 well format. The protocol of Regan et al [10] was cautiously. The influence of organic modifier at no and full inhibition is shown in Figure 2B.
The assay window decreased with increasing organic modifier (methanol) content. However,
2.2
used with some modifications. Since Mg2+ ions are known to be involved in ATP binding
and can potentially influence binding of inhibitors to the ATP binding site, MgCl2 was the Z’chrom-factors for the different compounds measured at different MeOH concentrations,
added. Furthermore, ELISA blocking reagent and PEG 6000 were added in order to avoid due to gradient elution, showed only minor differences thereby proving the robustness
non-specific binding and unwanted interactions with the material of the enzyme binding of the on-line setup even at high methanol concentrations. Increasing temperature also
detection [26]. Enzyme and tracer concentrations were optimized under these conditions. yielded a similar decrease in maximum signal as seen with methanol. In order to guarantee
The fluorescence signal showed a linear dependence on the enzyme concentration (cf robustness and enable the use of up to 90% methanol in the HPLC separation, the formic
Online Resource 1 Figure S1). The most suitable p38α concentration was therefore acid concentration in the eluent and the incubation temperature were restricted to 0.01%
chosen considering the Z’-factor measurements. The signal dependence on the tracer and 25°C, respectively. Hence, the temperatures of the HPLC separation step and the
enzyme binding detection were controlled separately.
The screening of highly lipophilic compounds is not only challenging in off-line screening
A due to adhesion and solubility issues [27] but also presents challenges in on-line screening
approaches [26]. In on-line screening systems, the amount of organic modifier is usually
kept low to prevent protein degradation. On the other hand, the tubing used for incubation
loops is mostly PEEK or PTFE. The favoured interaction of non-polar compounds with
these surfaces results in peak broadening and tailing, thus decreasing resolution and
sensitivity. By using polar material for the incubation loops, this effect was significantly
decreased. Figure 3A shows a two to threefold increase in signal height and reduced tailing
when untreated fused silica tubing is used instead of PEEK. Although, PTFE compared to
PEEK already yields decreased peak width, the width is additionally approximately halved
with untreated fused silica. The polarity of the incubation coil material plays a crucial role
explaining these findings. This can clearly be seen in Figure 3B where the influence of fused
silica tubing with covalent coatings of different polarities on the peak shape was compared.
B The polar (PEG coated) tubing gave the best results while peak height and shape degraded
towards intermediate (methyl/phenyl coated) and non-polar (methyl coated) surfaces. This
indicates the influence of tubing polarity on post-column band broadening and justifies a
preference of polar coated fused silica tubing over PEEK and PTFE in the presented study,
taking the structural features of the studied compounds into account.

Validation of the p38α binding assay


The quality of the p38α binding assay was assessed using the method of Zhang et al [25].
The Z’-factor is introduced as a single value to judge assay quality, because it incorporates
the SD of signal and background as well as the assay window. Therefore, it indicates the
ability to distinguish between signals and false positives or false negatives. According to
this method, the Z’-factor was calculated with equation 1 for several enzyme concentrations
in the plate reader based setup. μs and μc are defined as the average signal of the sample
Figure 2: Optimization of the p38α affinity assay. A) Saturation curve of the tracer for 90 nM p38α and the control, respectively. Consequently, σs and σc are the standard deviations (SD) of
(n=3). Full saturation was reached at 630 nM SKF, which was consequently the selected tracer these values.
concentration.
B) Influence of the organic modifier concentration on the enzyme binding detection. I) is the baseline
at different percentages of organic modifier in HPLC mobile phase, which shows the maximum signal
reached at these different MeOH concentrations. In opposition, line II) shows the fluorescence signal Equation 1
at maximum inhibition hence the difference between both lines describes the maximum assay window.
II) was measured by premixing SKF with MAPKI1 (900 nM final concentration) in the superloop.

56 57
Kool et al. already applied the Z’-factor to chromatography based on-line assays, adapting
the variables of equation 1 in order to take into account these discrepancies [28]. A main
advantage is the internal control value (μc) represented by the baseline, which is more
relevant than an external control, because it is available for each individual experiment.
For a complete consideration of the differences between the on-line and the plate reader
setup, all classical variables are systematically substituted with parameters more suitable
to the on-line approach. Doing this without compromising the comparability of this Z’-factor
is not trivial but possible. The assay window (μc-μs), as determined by the difference of the
mean of control (no inhibition) and signal (full inhibition), could more suitably be expressed
as the mean of the peak height of full inhibition (μH), thereby still describing the desired
parameter. Thus, the assay window is automatically corrected for changes in the baseline,
2.2 Figure 3: Influence of the reaction coil material on the peak shape. A) PEEK and PTFE were
representing the internal control. The SD of the sample (σs) would then consequentially
be identified with the SD of the peak heights at full inhibition. For the control, usually no
2.2
compared with untreated fused silica tubing. A 2  μM solution of ORGX was injected in FIA mode signals are obtained and changes of the baseline between samples are irrelevant as they
at 60% methanol. B) At the same conditions, a 20 μM solution of the same inhibitor was injected. have already been taken into account. The SD of the control (σc) can therefore logically be
Fused silica tubing with covalent coatings of either PEG (polar), a mixture of methyl and phenyl identified with the baseline deviation within one sample. This value is in chromatography
groups (intermediate) or only methyl groups (non-polar) was compared for their influence. In both better known as the noise. The measure of three times the noise is often used as limit
experiments, peak width and asymmetry increased with decreasing polarity while the peak height of detection. This is consistent with the ideas behind the Z’-factor as both interpretations
decreased as a consequence. Fused silica tubing, coated with PEG (polar), exhibited drastically
improved performance over the compared materials.
of 3σc are aimed at avoiding false positives. In summary, the transition from an external
control to an internal one strengthens the reproducibility and accuracy of the assay and
For the sake of comparability, the number of experiments (n=15) was kept the same in plate should therefore be appropriately reflected in the determination of Z’-factors. Therefore, an
reader and on-line based measurements. However, this is recognized as a relatively low adjusted Z’-factor for the use in on-line systems is proposed in equation 2 on basis of the
number for plate reader experiments. Therefore, an additional plate reader experiment with previous argumentation.
n=50 was performed, yielding a Z’-factor of -0.02. The conclusion from both experiments
was as follows: The plate reader based assay at 90  nM p38α gave insufficient results
with Z’-factors lower than 0. In the plate reader assay, a Z’-factor above 0.5, indicating Equation 2
an excellent assay, could only be achieved by raising the p38α concentration to 450 nM.
Table 1 compares this data with the results of the on-line setup, in which 90 nM p38α was
sufficient for an excellent assay. As a confirmation, the signal to noise ratios (S/N) were
also calculated (Table 1), resulting in the same conclusions.
Regardless of the findings of the last paragraph, the on-line setup has two disadvantages.
Reagent consumption is usually higher in an on-line setup than in a plate reader based assay.
Furthermore, the separation step introduces an additional dilution due to chromatographic
peak broadening, which requires higher analyte concentrations. There are additional
differences between the two setups, which should be adequately reflected in the Z’-factor.
Table 1: Z’-factors and signal to noise rations (S/N) for plate reader
and/or on-line based enzyme binding detection
Inhibitor a c (p38α) [nM] Z’ or Z’chrom b (n=15) S/N
Plate reader TAK715 90 -0.41 4
180 0.49 12
270 0.36 23
360 0.49 55
450 0.75 48
On-line BIRB796 90 0.77 70
ORGX 90 0.82 106
TAK715 90 0.80 97
Figure 4: Comparison of MS and enzyme binding detection chromatogram. The matching of the
a
Concentration of the inhibitor was 1 μM TAK715 (final concentration) in the plate reader experiments. fluorescence trace of the enzyme binding detection (above) and the total ion current (TIC) of the MS
In the on-line setup ORGX and TAK715 were injected at 20 μM concentration and BIRB796 at 50 μM (below) for BIRB796 (1), ORGX (2) and TAK715 (3) are shown. Depicted is a triplicate from the Z’chrom-
concentration for this purpose. factor determination. The excellent reproducibility in response, peak shape and retention time are
b
In the plate reader experiments the Z’-factor was calculated according to equation 1. For the on-line clearly visible for both the enzyme binding detection and the MS signal. The low variability enables
experiments, equation 2 was used, yielding the Z’chrom-factor. an easy and accurate matching of the two traces.

58 59
The new interpretation of the Z’-factor could also be used to omit the measurements of a High resolution screening of mixtures
control, significantly shortening the time of Z’chrom-factor determination. Because the noise In order to assess whether the presented HRS method was suitable for the identification of
is calculated from the baseline of the chromatogram, a prerequisite is to have an empty small molecule binders of p38α in mixtures, a solution of eight compounds was screened.
time frame to observe both required values. Therefore, the first 4 min of the enzyme binding Two of these were the kinase inhibitors MAPKI1 and TAK715 while the other six were
detection chromatogram were used to determine the noise. The mean noises calculated drug molecules from different classes. As illustrated in Figure 5, all eight compounds could
from signal and control runs were identical, thus proving the redundancy of the control be separated for individual identification and affinity assessment. Based on HR-MS data,
measurements for Z’chrom-factor determinations. the structure was linked to a retention time. Accurate mass measurements enabled the
The repeatability of triplicates is excellent, which is clearly visible in Figure 4, 5 and 6. This unambiguous distinction of all compounds as two of them differed by less than 0.05 u.
is true for the enzyme binding detection responses as well as for the chromatographic In case of more scarce knowledge of the sample, structure elucidation can be facilitated
parameters like peak width, measured as full width at half maximum (FWHM), and retention by HR-MSn experiments [21]. In parallel to the structure confirmation, the retention time
time (tr). For the enzyme binding detection, the relative SDs are ≤10% for FWHM and <3% is also linked with affinity information through the enzyme binding detection. However,
2.2 for tr. Low variability and noise compared to the total signal results in the high Z’-factor and
signal-to-noise ratio (S/N) shown in Table 1. The high Z’-factors indicate the excellent intra-
the readout of the enzyme binding detection is delayed compared to the HR-MS readout,
because of a higher void volume and a lower average flow rate after splitting. This delay
2.2
and interday reproducibility, However, the on-line setup showed comparable Z’-factors was measured to be 0.5 min by comparing both detection times in several experiments. It
at five times lower enzyme concentration than needed in the plate based assay. This is remains constant as long as void volume and flow rates are unchanged. The retention time
directly related to the minimal measurable IC50. While IC50 values down to 45 nM can be corrected for this delay is used to couple affinity to identity information. Two affinity peaks
distinguished in the on-line method, the plate reader method only allows differentiation were detected, which were linked to the elution of the kinase inhibitors. In conclusion,
of IC50 values of 230 nM and higher. Furthermore, the higher enzyme concentration the approach presents a fast method to match the structure and activity of compounds in
needed diminishes the advantage of the plate setup with regard to reagent consumption. mixtures.
Therefore, the on-line setup has advantages beside the additional possibilities associated
with separation. Semi-Quantitative affinity measurements
Several known p38α inhibitors were tested for their dose related responses in the enzyme
binding detection. Therefore, their injected concentrations had to be converted to final
assay concentrations by taking into account the dilution factors. On one hand, splitting of
Fluorescence response [mV]

0 the HPLC eluent and subsequent mixing with the reagents leads to dilution. This can be
easily calculated from the ratios of the flow in the beginning and the end of the enzyme
binding detection (113/13 = 9). On the other hand, the chromatographic dilution, meaning
-50 the transition from an injected plug to a series of peaks of (assumed) Gaussian shape, has
to be taken into account. The corresponding calculations have been described elsewhere
[29]. The necessary peak parameters were taken from the enzyme binding detection.
By dividing the injected concentration by the product of both dilution factors, the final
-100 concentration at maximum peak height can be calculated and used in the IC50 calculations

5 8
100
4

2 6 80
MS response (XIC)

Binding [%]
1 3
60
7
40

20
0 10 20 30 40 50
0
Retention time [min] 0 1 2 3 4
Figure 5: HRS of a mixture of kinase inhibitors and drug molecules. MAPKI1 (4) and TAK715 (8) were logc [nM]
mixed with carbamazepine (1), norethisterone (2), warfarin (3), phenylbutazone (5), celecoxib (6) and
diclofenac (7). The mixture was separated by HPLC and measured with HR-MS and enzyme binding Figure 6: Quantitative data of the tested p38α inhibitors. Binding curves are depicted for several
detection in parallel. Accurate mass measurements with low deviation (<8ppm) and well matching known p38α inhibitors. The inhibitors TAK715 (circles), BIRB796 (squares) and MAPKI1 (triangles)
isotope patterns allowed unambiguous identification. Structure and affinity were easily matched via are shown. Standard deviations of triplicate measurements were smaller than data point size.
the corrected retention times. Consequently, the two affinity compounds were detected in the mixture.
60 61
(for a brief description see Online Resource 1). Conclusion
Dose-response curves depicted in Figure 6 were measured in triplicate with excellent R2-
values of >0.998 and minimal standard deviation (SD) of the triplicates (<2% SD relative to An on-line binding assay for the separation and parallel assessment of identity and
assay window). This shows again the excellent reproducibility and accuracy of the assay. affinity of small molecules in complex mixtures towards non-phosphorylated p38α was
The IC50 values and, given in brackets, their 95% confidence intervals, calculated after developed. In terms of applicability, the scope of the presented HRS platform is different
correcting the concentrations for the dilution factors are 81 nM (71 to 92 nM) for TAK715, from conventional HTS assays. HTS is more suited for the screening of huge libraries
400 nM (360 to 440 nM) for BIRB796 and 760 nM (530 to 1100 nM) for MAPKI1. If the where speed and low sample consumption are essential. The value of HRS lies in the
inhibitors are ranked for their affinity, the obtained results are in agreement with other parallel determination of structural and biochemical data of potential binders. While an
methods, stating values of 7.1 nM for TAK715 [8], 18 nM for BIRB796 [10] and 35 nM for HTS assay will only give a sum of compound affinities, HRS gives individual information
MAPKI1 [9]. However, the absolute values of measurements are 10 to 20 times higher on all (separated) molecules. The aim was to make available the advantages of the on-line
than in literature. This can be explained by the profound differences in the assay formats. approach, mainly the measurement of mixtures and a comparison of structure and affinity,
2.2 While the reported method gives affinities, the reference values were obtained by either
activity assays (TAK715) or cell based functional assays (BIRB796, MAPKI1). Another
for the important drug target p38α. An on-line post-column enzyme binding detection 2.2
based on fluorescence enhancement was developed and its compatibility with modern
specific reason responsible for this deviation is the slow binding kinetics of BIRB796. This HPLC separation techniques, such as gradient elution and increased temperature, was
compound is known to show an up to 12-fold deviation of its IC50 value, depending on the demonstrated. HR-MS as parallel readout is an integral part of the developed screening
incubation time [11]. Despite the aforementioned issues, the method was proven to be platform, allowing the identification of the tested molecules. Additionally, the prospects of
suitable for the semi-quantitative assessment of small molecule relative affinities towards measuring compounds individually in mixtures were demonstrated by matching identity
p38α serving as a selection tool in the drug discovery process. and affinity. Regarding the presented assay validation data, it can be stated that a high
quality assay was achieved. Finally, the assay provided ranking of various p38α inhibitors
Applicability and binding modes (types I, II and III) allowing the use within a drug discovery process.
The type of binding to p38α as discussed in the introduction can have a significant influence
on the ability of a screening technology to detect this binding. SKF is a type I binder and
should therefore enable the detection of type I and type II binders as they compete for the
Acknowledgements
ATP-binding pocket. This has been proven by the detection of TAK715 and MAPKI1 (type
This work was performed within the framework of the Dutch Top Institute Pharma project D2-
I) as well as BIRB796 (type II). Furthermore, structural changes in p38α upon binding of
102. We thank MSD research laboratories for kindly providing p38α and other chemicals.
type III inhibitors may allow the detection of such compounds. The rearrangement of p38α
We thank Guido Zaman for important discussions. The work of the author M. Giera was
from the DFG-in to the DFG-out conformation results in a reorientation of Phe405 thereby
supported by the German Academic Exchange Service (DAAD).
affecting the ATP-binding pocket. Hence, we tested the pyrazolourea structure, which
was discussed by Simard et al as an example of a type III inhibitor [12]. Figure 7 shows
the enzyme binding detection of this compound to p38α, thereby proving the suitability of References
the method for the detection of type III inhibitors. Consequently, the assay is capable of 1. Cohen P (2002) Protein kinases-the major drug targets of the twenty-first century? Nat Rev Drug
detecting a wide range of p38α binders including type I, II and III binders. Discov 1 (4):309-315
2. Zhang J, Shen B, Lin A (2007) Novel strategies for inhibition of the p38 mapk pathway. Trends
Pharmacol Sci 28 (6):286-295
270
Fluorescence [mV]

3. Kumar S, Boehm J, Lee JC (2003) P38 map kinases: Key signalling molecules as therapeutic
targets for inflammatory diseases. Nat Rev Drug Discov 2 (9):717-726
4. Saklatvala J (2004) The p38 map kinase pathway as a therapeutic target in inflammatory
260 disease. Curr Opin Pharmacol 4 (4):372-377
5. Schindler JF, Monahan JB, Smith WG (2007) P38 pathway kinases as anti-inflammatory drug
targets. J Dent Res 86 (9):800-811
6. Mayer RJ, Callahan JF (2006) P38 map kinase inhibitors: A future therapy for inflammatory
250 diseases. Drug Discov Today 3 (1):49-54
7. Goldstein DM, Kuglstatter A, Lou Y, Soth MJ (2010) Selective p38alpha inhibitors clinically
evaluated for the treatment of chronic inflammatory disorders. J Med Chem 53 (6):2345-2353
8. Miwatashi S, Arikawa Y, Kotani E, Miyamoto M, Naruo K, Kimura H, Tanaka T, Asahi S, Ohkawa
TIC

S (2005) Novel inhibitor of p38 map kinase as an anti-tnf-alpha drug: Discovery of n-[4-[2-ethyl-
4-(3-methylphenyl)-1,3-thiazol-5-yl]-2-pyridyl]benzamide (tak-715) as a potent and orally active
anti-rheumatoid arthritis agent. J Med Chem 48 (19):5966-5979
0 5 10 9. de Laszlo SE, Visco D, Agarwal L, Chang L, Chin J, Croft G, Forsyth A, Fletcher D, Frantz B,
Hacker C, Hanlon W, Harper C, Kostura M, Li B, Luell S, MacCoss M, Mantlo N, O’Neill EA,
Retention time [min] Orevillo C, Pang M, Parsons J, Rolando A, Sahly Y, Sidler K, O’Keefe SJ (1998) Pyrroles and
other heterocycles as inhibitors of p38 kinase. Bioorg Med Chem Lett 8 (19):2689-2694
Figure 7: Detection of the binding of the pyrazolourea type III inhibitor to p38α. Triplicate injections 10. Regan J, Breitfelder S, Cirillo P, Gilmore T, Graham AG, Hickey E, Klaus B, Madwed J, Moriak
of the inhibitor (black) showed signals in the enzyme binding detection (top). In contrast, the control M, Moss N, Pargellis C, Pav S, Proto A, Swinamer A, Tong L, Torcellini C (2002) Pyrazole urea-
(grey) did not show binding to p38α. HR-MS (bottom) confirmed the pyrazolourea compound as the based inhibitors of p38 map kinase: From lead compound to clinical candidate. J Med Chem 45
binder. This clearly shows the ability of the assay to detect type III inhibitors. In combination with Fig. (14):2994-3008
4 it proves the applicability to several types of p38α inhibitors (types I, II and III).

62 63
11. Pargellis C, Tong L, Churchill L, Cirillo PF, Gilmore T, Graham AG, Grob PM, Hickey ER, Moss
N, Pav S, Regan J (2002) Inhibition of p38 map kinase by utilizing a novel allosteric binding site.
Supplementary Material
Nat Struct Biol 9 (4):268-272
12. Simard JR, Klüter S, Grütter C, Getlik M, Rabiller M, Rode HB, Rauh D (2009) A new screening Signal dependence on p38α concentration
assay for allosteric inhibitors of csrc. Nat Chem Biol 5 (6):394-396 3000
13. Adrian FJ, Ding Q, Sim T, Velentza A, Sloan C, Liu Y, Zhang G, Hur W, Ding S, Manley P, Mestan
J, Fabbro D, Gray NS (2006) Allosteric inhibitors of bcr-abl-dependent cell proliferation. Nat

Fluorescence [counts]
y = 6,226x + 195,77
Chem Biol 2 (2):95-102 R2 = 0,9995
2000
14. Klumpp M, Boettcher A, Becker D, Meder G, Blank J, Leder L, Forstner M, Ottl J, Mayr LM
(2006) Readout technologies for highly miniaturized kinase assays applicable to high-throughput
screening in a 1536-well format. J Biomol Screen 11 (6):617-633
15. Olive DM (2004) Quantitative methods for the analysis of protein phosphorylation in drug 1000

2.2 development. Expert Rev Proteomics 1 (3):327-341


16. Zaman GJ, van der Lee MM, Kok JJ, Nelissen RL, Loomans EE (2006) Enzyme fragment
2.2
complementation binding assay for p38alpha mitogen-activated protein kinase to study the 0
binding kinetics of enzyme inhibitors. Assay Drug Dev Technol 4 (4):411-420 0 100 200 300 400
17. Lee JC, Laydon JT, McDonnell PC, Gallagher TF, Kumar S, Green D, McNulty D, Blumenthal MJ,
Concentration p38α [nM]
Heys JR, Landvatter SW, Strickler JE, McLaughlin MM, Siemens IR, Fisher SM, Livi GP, White
JR, Adams JL, Young PR (1994) A protein kinase involved in the regulation of inflammatory Figure S1: Baseline signal dependence on enzyme concentration in the plate reader setup. The
cytokine biosynthesis. Nature 372 (6508):739-746 maximum fluorescence, equivalent to the maximum enzyme-tracer complex concentration, was
18. Munoz L, Selig R, Yeung YT, Peifer C, Hauser D, Laufer S (2010) Fluorescence polarization measured at different enzyme concentrations. 1250  nM of SKF were used. The data justifies the
binding assay to develop inhibitors of inactive p38alpha mitogen-activated protein kinase. Anal homogeneous setup as it proves the insignificance of the background fluorescence at the tested
Biochem 401 (1):125-133 concentration. Furthermore, it suggests that the assay is tuneable to a desired compromise between
19. Young PR, McLaughlin MM, Kumar S, Kassis S, Doyle ML, McNulty D, Gallagher TF, Fisher S,
quality and enzyme consumption.
McDonnell PC, Carr SA, Huddleston MJ, Seibel G, Porter TG, Livi GP, Adams JL, Lee JC (1997)
Pyridinyl imidazole inhibitors of p38 mitogen-activated protein kinase bind in the atp site. J Biol
Chem 272 (18):12116-12121 Calculation of final inhibitor concentration
20. de Vlieger JS, Kolkman AJ, Ampt KA, Commandeur JN, Vermeulen NP, Kool J, Wijmenga SS, There are two factors of dilution which diminish the concentration of the tested compound
Niessen WM, Irth H, Honing M (2010) Determination and identification of estrogenic compounds in the enzyme binding detection. The first arises from the mixing of the HPLC eluent with
generated with biosynthetic enzymes using hyphenated screening assays, high resolution mass the reagents of the enzyme binding detection. It can therefore be called mixing dilution
spectrometry and off-line nmr. J Chromatogr B 878 (7-8):667-674 (DM). DM is easily calculated by dividing the final flow rate in the fluorescence detector (uF)
21. Giera M, de Vlieger JS, Lingeman H, Irth H, Niessen WM (2010) Structural elucidation of with the flow rate of the HPLC eluent entering the enzyme binding detection (uE), because
biologically active neomycin n-octyl derivatives in a regioisomeric mixture by means of liquid this represents the ratio of volumes in any given time frame.
chromatography/ion trap time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 24
(10):1439-1446
Equation S1
22. Van Liempd SM, Kool J, Meerman JH, Irth H, Vermeulen NP (2007) Metabolic profiling of
endocrine-disrupting compounds by on-line cytochrome p450 bioreaction coupled to on-line
receptor affinity screening. Chem Res Toxicol 20 (12):1825-1832 The second factor is the chromatographic dilution (DC). It can be calculated assuming
23. Giera M, Heus F, Janssen L, Kool J, Lingeman H, Irth H (2009) Microfractionation revisited: A a symmetrical peak of Gaussian shape. Based on this assumption, the ratio between
1536 well high resolution screening assay. Anal Chem 81 (13):5460-5466 the height of a plug of the injection volume and the resulting Gaussian peak equals the
24. Shi SY, Zhou HH, Zhang YP, Jiang XY, Chen XQ, Huang KL (2009) Coupling hplc to on-line, dilution factor. Influences are the full width at half maximum (FWHM), the flowrate in the
post-column (bio)chemical assays for high-resolution screening of bioactive compounds from HPLC (uC) and the injection volume (Vi). It is imperative that the FWHM is taken from the
complex mixtures. Trac-Trends in Analytical Chemistry 28 (7):865-877 fluorescence and not from the MS chromatogram. The bigger void volume in the enzyme
25. Zhang JH, Chung TD, Oldenburg KR (1999) A simple statistical parameter for use in evaluation binding detection leads to a broader peak because of diffusion and surface interactions.
and validation of high throughput screening assays. J Biomol Screen 4 (2):67-73
26. Kool J, van Liempd SM, Ramautar R, Schenk T, Meerman JH, Irth H, Commandeur JN,
Equation S2 [1]
Vermeulen NP (2005) Development of a novel cytochrome p450 bioaffinity detection system
coupled online to gradient reversed-phase high-performance liquid chromatography. J Biomol
Screen 10 (5):427-436 By combining these two factors the final concentration (cF) of the binders can be determined
27. Schebb NH, Falck D, Faber H, Hein EM, Karst U, Hayen H (2009) Fast method for monitoring from the injected concentration (ci).
phospholipase a2 activity by liquid chromatography-electrospray ionization mass spectrometry.
J Chromatogr A 1216 (27):5249-5255 Equation S3
28. Kool J, van Marle A, Hulscher S, Selman M, van Iperen DJ, van Altena K, Gillard M, Bakker RA,
Irth H, Leurs R, Vermeulen NP (2007) A flow-through fluorescence polarization detection system
for measuring gpcr-mediated modulation of camp production. J Biomol Screen 12 (8):1074- 1. Schebb NH, Heus F, Saenger T, Karst U, Irth H, Kool J (2008) Development of a countergradient
1083 parking system for gradient liquid chromatography with online biochemical detection of serine
29. Schebb NH, Heus F, Saenger T, Karst U, Irth H, Kool J (2008) Development of a countergradient protease inhibitors. Anal Chem 80 (17):6764-6772
parking system for gradient liquid chromatography with online biochemical detection of serine
protease inhibitors. Anal Chem 80 (17):6764-6772

64 65
Chapter 2.3

Development of on-line liquid


chromatography-biochemical
detection for soluble epoxide
hydrolase inhibitors in mixtures

David Falck1, Nils Helge Schebb2,3, Setyo Prihatiningtyas1, Jiawen Zhang1,


Ferry Heus1, Christophe Morisseau2, Jeroen Kool1, Bruce D. Hammock2
and Wilfried M. A. Niessen1
1
Department of BioMolecular Analysis, VU University Amsterdam, Amsterdam, The
Netherlands.
2
Department of Entomology and UC Davis Comprehensive Cancer Center, University of
California, Davis, CA, USA.
3
Institute for Food Toxicology and Chemical Analysis, University of Veterinary Medicine
Hannover, Hannover, Germany.
Abstract methoxy-naphthalen-2-yl)-methyl ester (PHOME), which is converted to the fluorescent
product 6-methoxy-2-naphthaldehyde by sEH. The new analytical method was thoroughly
In this study, an end-point based fluorescence assay for soluble epoxide hydrolase optimized and validated. The obtained inhibition efficacy of known inhibitors compared well
(sEH) was transformed into an on-line continuous-flow format. The on-line biochemical to literature values. Finally, microsomal incubations of three sEHis were screened for active
detection system (BCD) was coupled on-line to liquid chromatography (LC) to allow metabolites.
mixture analysis. The on-line BCD was based on a flow system wherein sEH activity was
detected by competition of analytes with the substrate hydrolysis. The reaction product was Materials and Methods
measured by fluorescence detection. In parallel to the BCD data, UV and MS data were
obtained through post-column splitting of the LC effluent. The buffer system and reagent Materials
concentrations were optimized resulting in a stable on-line BCD with a good assay window Human recombinant sEH was expressed and purified as described [19]. The sEH inhibitors
and good sensitivity (S/N > 60). The potency of known sEH inhibitors (sEHis) obtained by and the substrate (3-phenyl-oxiranyl)-acetic acid cyano-(6-methoxy-naphthalen-2-yl)-
LC–BCD correlates well with published values. The LC–BCD system was applied to test methyl ester (PHOME) as well as its fluorescent product are shown in Figure  1. Their
how oxidative microsomal metabolism affects the potency of three sEHis. After incubation
2.3 2.3
synthesis was reported earlier: PHOME [8]; sEHi 1 [8]; sEHi 2 [20]; sEHi 3 [21]; sEHi 4 [22];
with pig liver microsomes, several metabolites of sEHis were characterized by MS, while sEHi 5 [23]; sEHi 6 and sEHi 7 [24]. ELISA blocking reagent (EBR) was purchased from
their individual potencies were measured by BCD. For all compounds tested, active Roche Diagnostics (Mannheim, Germany). All other chemicals were from Sigma-Aldrich
metabolites were observed. The developed method allows for the first time the detection (Schnelldorf, Germany). Methanol (LC–MS Grade) and formic acid (ULC–MS Grade) were
of sEHis in mixtures providing new opportunities in the development of drug candidates. obtained from Biosolve (Valkenswaard, The Netherlands). The water used in this study was
generated with a Milli-Q academic from Millipore (Amsterdam, The Netherlands).
Introduction
Liquid chromatography – biochemical detection (LC–BCD) system
Soluble epoxide hydrolase (sEH) plays an important role in regulation of blood pressure, The LC–BCD system shown in Figure 2 includes separation, on-line BCD and additional
pain and inflammation [1]. In mammals, sEH is expressed in various tissues. The parallel spectrometric detection. Separation and on-line BCD were done on an Agilent
endogenous substrates of sEH are among others epoxyeicosatrienoic acids (EETs), which Technologies (Amstelveen, The Netherlands) 1100 LC system including a binary and two
are hydrolyzed to dihydroxyeicosatrienoic acids (DHETs), thus leading to decreased blood isocratic pumps, an autosampler, a column oven and a fluorescence detector. Separation
levels of EETs. Various studies show, that EETs and epoxides of other unsaturated fatty
acids are anti-inflammatory, analgesic agents and lower blood pressure [2; 3; 4]. The A
biological levels of epoxy fatty acids can be increased by sEH inhibitors (sEHis), leading to
reduction in inflammation, pain, and cardiovascular diseases in various animal models [5;
6]. Thus, sEHis are a promising new class of pharmaceutical drug candidates.
During the lead development process, metabolism studies play an important role. Not
only the pharmacokinetic profile, but also the biological effects of metabolites are relevant
for the action of drugs. Metabolites can be inactive, reactive, but also pharmacologically
active towards the same pharmacological target or against off-targets. Screening metabolic
mixtures for individual bioactive metabolites is not possible with standard end-point plate-
reader based screening methodologies. For sEH, several end-point assays have been
developed, based on fluorescent detection, radiometry, and mass spectrometry [7; 8; 9].
However, these assays are only suited to screen pure compounds. The analysis of mixtures
would only yield the sum of the total bioactivity of the mixture [10]. In order to assess the
bioactivity of individual metabolites, fractionation has to precede the screening. As this is
time-consuming, costly, and has to be performed at low resolution to prevent too much
dilution, such approaches are inefficient for bioactivity profiling of metabolic mixtures [11].
One way of tackling this problem is the application of an on-line post-column screening B
approach, known as high-resolution screening (HRS) [12]. This technology continuously
mixes bioassay reagents with the eluent after an LC separation of a mixture of compounds
[13; 14; 15], such as metabolic mixtures [16]. For HRS screening with enzyme targets,
like in this LC–BCD system, inhibition can be measured by detecting a decrease in the
enzymatic formation of a fluorescent product. Splitting part of the LC eluent between the
BCD and mass spectrometry (MS) enables correlation of bioactivity with identity for all
individual metabolites [14; 17; 18].
This paper describes the development of an LC–BCD system for sEHis and its application
in the efficient profiling of active oxidative metabolites. For this purpose, a fluorescence
end-point plate-reader assay [7] was converted into an on-line BCD format. The on-
line BCD uses the non-fluorescent substrate (3-phenyl-oxiranyl)-acetic acid cyano-(6- Figure 1: Structures of A) the sEHis used in this study, and B) the reaction scheme of the substrate
PHOME to its fluorescent product.
68 69
was performed on a Waters (Milford, MA, USA) Atlantis dC18 column (100 x 2.1 mm, 3 μm Figure 3: Analysis of BCD
particles) at 40°C. The mobile phase consisted of water:methanol 100:1 (v:v) as solvent A parameters. Several sections of the
and methanol:water 100:1 (v:v) as solvent B, both containing 0.01% formic acid. A mixture graph are highlighted by horizontal
of known sEHis (125 µM each) was separated using the following gradient: 0 to 2  min double-headed arrows in contrast
to important time points which
isocratic at 50% B, then a linear gradient to 90% B in 43 min and isocratic at 90% B for
are highlighted by vertical arrows.
4 min; afterwards a linear decrease to 50% B in 2 min followed by 15 min re-equilibration. Sections 1, 3 and 8 show the buffer,
The LC gradient applied for the analysis of the metabolic incubations was as follows: 0 to the substrate related and the complete
2 min isocratic at 5% B, followed by a linear gradient to 95% B in 43 min, isocratic at 95% background levels of fluorescence,
B until 49 min then a linear decrease to 5% B in 6 min followed by 10 min re-equilibration. respectively. Furthermore, section
For analysis in flow injection analysis mode (FIA), the same set-up was used, but without 5 depicts the baseline due to the
the column. The LC flow-rate was 150 μL/min and the injection volume 10 μL in all cases. enzymatic reaction. The points 2
The flow was post-column split (see Figure 2), directing 135 μL/min to the MS detection and 4 mark the introduction of the
and 15 μL/min to the on-line BCD. In the BCD, the eluent was first mixed with 155 μL/min PHOME substrate solution and of the
2.3 of a 5 nM sEH solution and incubated for 30 s. This allowed an initial interaction between
analytes and enzyme. In a second step, a 30 μM solution of PHOME was added at 30 μL/
sEH enzyme solution, respectively.
Point 7 indicates the start of the 2.3
cooling of the incubation tubing,
min and incubated for 5 min. Here, the substrate conversion to the fluorescent product took which stops the enzymatic conversion in section 8. The difference between the fluorescence at full
place which allowed detection of the enzyme activity. The incubations were done in 1.73 m steady state reaction (section 5) and the combined fluorescence background at completely stopped
straight 250 µm i.d. and 1.59 mm o.d. PTFE tubing (Sigma-Aldrich, Schnelldorf, Germany) reaction (section 8), as indicated by a vertical double-headed arrow (number 6), indicates the assay
and 2.26 m of 750 µm i.d. and 1.59 mm o.d. coiled PTFE tubing, forming 85 μL and 1 mL window. The eight numbers are explained in more detail in the text.
reactors, respectively. Both reactors were kept at 37°C by a Grant Instruments (Shepreth,
UK) water bath. The enzyme and substrate solutions were delivered by in-house built Determination of inhibitor potency
superloops which were kept on ice [25]. The fluorescence was measured at excitation The potency of five known sEHis (Figure  1) was determined based on their apparent
of 320±10 nm and emission of 460±10 nm. In parallel to the on-line BCD, detection was IC50 values to characterize the performance of the LC-BCD system. These sEHis have
performed by UV at 210 nm and/or by mass spectrometry. been selected in such a way that their IC50 values ranged from low to high nanomolar,
Both techniques are able to visualize both the binders and the non-binders. Additionally, thus covered approximately three order of magnitude of inhibitory activity. For measuring
MS provides structural information. The on-line BCD and the parallel UV or MS detection the IC50 values, dose-response curves were obtained by injecting the inhibitors into the
have different void volumes after the splitting and thus the elution times differ. The UV or LC–BCD system under isocratic conditions at 50% methanol in FIA mode. The following
MS and BCD chromatograms were aligned using a known compound, e.g., the residual concentrations and one blank were injected in duplicate per inhibitor: 0.5, 1, 2, 5, 10, 20
parent compound in case of the metabolic incubations. and 50 μM for sEHi 1; 1, 2, 5, 10, 20, 50, 100 and 200 μM for sEHi 2; 0.5, 1, 2, 5, 10 and
20 μM for sEHi 3; 1, 2, 5, 10, 20, 100, 500 and 1000 μM for sEHi 4; 0.05, 0.1, 0.2, 0.5, 1,
2 and 10 μM for sEHi 5.

Metabolite identification using mass spectrometry


LC–MS for metabolite identification was done either on a Bruker Daltonik (Bremen,
Germany) micrOTOF-Q quadrupole time-of-flight hybrid MS, using the above described
conditions, or using an ion-trap time-of-flight mass spectrometer (IT-TOF, Shimadzu, ‘s
Hertogenbosch, The Netherlands). In the latter case, a 30-min gradient and a 100 × 2.1
mm Waters XBridge C18 column (3.5 μm particles) was used. Positive-ion electrospray
ionization (ESI) was applied in both instrument. Other relevant instrument settings are
summarized in the Supporting Information. The mass accuracy was better than 5 ppm on
both instruments. The accurate-mass data obtained were used to determine the elemental
composition of the metabolites and accordingly of the fragments.

Buffer and compound solutions


A 25 mM 2-bis(2-hydroxyethyl)amino-2-(hydroxymethyl)-1,3-propanediol (BIS-TRIS) buffer
containing 1 g/L EBR, 1 g/L bovine serum albumin (BSA) and 0.1 g/L Tween 80 was used
at pH 7.0. Stock solutions of the sEH inhibitors and PHOME were prepared at 20  mM
concentrations in DMSO. sEH stocks of 100  μM (6  mg/mL) concentration were kept at
-80°C until use and dilutions were handled on ice at all times. All PHOME and sEH dilutions
Figure 2: Setup of the LC–BCD system. The system combines separation, on-line BCD and additional were prepared in this BIS-TRIS buffer.
UV or MS detection in parallel. It includes (1) autoinjector, (2) reversed-phase LC column, (3) flow-
splitting between parallel (9) UV or ESI-MS detection and (4–8) the on-line BCD. The BCD comprises Plate reader measurements
of (4) mixing of LC effluent and an sEH solution, (5) incubation with the enzyme, followed by (6) Plate reader based measurements were performed to evaluate the reagent concentrations
mixing of PHOME solution, (7) incubation with PHOME, and finally (8) fluorescence detection. on a Victor3 plate reader from Perkin-Elmer (Groningen, The Netherlands). Black 96 ‘flat’
70 71
bottom chimney well, polypropylene microtiter plates from Greiner bio-one (Alphen a/d comprise a BSA concentration of 1.0 g/l rather than 0.1 g/l.
Rijn, The Netherlands) were used. The total sample volume was 200  μL and the plates The enzyme concentration was adjusted to yield about 20% substrate conversion in a
were incubated at 37°C. Product formation was followed by measuring the fluorescence at 5-min reaction time. This low percentage of conversion ensures that the reaction is still
355 ± 4 nm excitation and 460 ± 12.5 nm emission. The PHOME concentration was 50 μM in the initial conversion stage where no significant rate and concentration limiting effects
and the sEH concentration 40 nM. Product formation was measured in 30 s intervals for are observed. It was found that only 4 nM sEH in the reaction coil were sufficient, which is
20 min under the influence of two different BSA concentrations, 0.1 g/L and 1.0 g/L. End- almost identical to the concentration of 3 nM in the end-point method [7].
point measurements at 6 min were used to compare the activity of sEH under the influence Figure 3 shows the most important BCD parameters and demonstrates that the reaction
of several solubilising agents. In additional experiments, the solubility of PHOME under the causing the BCD baseline is of enzymatic nature. Section 1 of Figure 3 shows the background
influence of these solubilising agents was tested in transparent plates. This was done by level of fluorescence generated with buffer-filled superloops. When the PHOME substrate
measuring precipitation of a 45 μM PHOME solution by visible absorption at 595±10 nm. solution was introduced (point 2 in Figure 3), the fluorescence signal somewhat increased
due to fluorescence of the substrate and/or autohydrolysis and/or presence of trace amount
Microsomal incubations of the fluorescent product in the substrate (section 3 in Figure 3). After the enzyme is added
The LC–BCD/MS system was applied to investigate the metabolism of the three known to the other superloop (point  4 in Figure  3), a rapid increase in the signal is observed
2.3 sEH inhibitors sEHi 6, sEHi 1 and sEHi 7 and the bioactivity profile of their metabolites.
Oxidative metabolites were generated by pig liver microsomal incubations in the presence
resulting in a stable baseline due to the steady state of the enzymatic reaction (section 5 in
Figure 3). When cooling of the reaction coil on ice (point 7 in Figure 3), the signal returns to
2.3
of NADPH according to a modified version of a protocol described elsewhere [26]. In the baseline of substrate solution (section 8 in Figure 3). The absence of conversion under
brief, reaction mixtures were prepared in 50  mM potassium phosphate buffer (pH 7.4) these conditions clearly demonstrates that the increased fluorescence signal is caused by
including 5 mM magnesium chloride. The mixtures containing 6 mM NADPH, 2.6 mg/mL the enzymatic conversion of PHOME to its fluorescent product.
pig liver microsomes and 100 mM sEHi were incubated for 2 h at 37°C. To ensure continued The most important parameter of the BCD system is the difference between the steady
availability of NADPH, 5  mM glucose-6-phosphate and 5  U/mL glucose-6-phosphate state of the reaction serving as baseline of BCD and the background signal (number 6 in
dehydrogenase were used as regenerating system. Additionally, 10% (v/v) of a 10  mM Figure 3, indicating the difference in fluorescence signal between section 5 and section 8 in
NADPH solution in the above mentioned phosphate buffer was added after 30, 60 and Figure 3), also known as assay window. In this case, the assay window is nine times higher
90 min, respectively. The reactions were stopped by adding ice-cold acetonitrile 2:1 (v:v). than the substrate fluorescence. Together with the stability of the signal, a signal-to-noise
The samples were then centrifuged at 16,000 g for 5 min. The supernatants were taken, ratio of 70 results between full inhibition of sEH and the steady state, allowing a detection
freeze-dried and stored at –20°C. For the LC–BCD/MS analysis, the samples were re- of as little as 5% inhibition of the added sEH. In conclusion, the data quality of the on-line
dissolved in a 30% aqueous methanol solution, providing 20-fold higher concentrations. BCD is more than adequate to detect the significant part of the sigmoidal dose-response
behaviour of inhibitors between 10% and 90% inhibition.
Results and Discussion As the sEHis are introduced into the on-line BCD from an LC setup, their activity is
visualized as a negative chromatographic peak which is a result of the indirect enzymatic
Development of an LC–BCD system for the detection of sEH inhibitors activity measurement: higher inhibitor activity means less fluorescent product formation.
The aim of this study was to develop a system for the bioactivity assessment towards Furthermore, the dose-response behaviour in the on-line BCD changes to a sigmoidal one
sEH of individual compounds in complex mixtures. For this purpose, a homogeneous, due to the underlying competitive biophysical interactions between enzyme, substrate and
continuous-flow detection format was applied based on the enzymatic conversion of the inhibitor. All this is apparent from Figure 4A where the overlaid BCD chromatograms of a
substrate PHOME and fluorescence readout of the reaction product (Figure 1) [7]. dilution series of several sEHi 1 concentrations are shown.
Initial plate-reader experiments were used as starting conditions for the optimization of
the enzyme and substrate concentrations in the on-line setup. This was achieved by Characterization of the LC–BCD system
determining the signal-to-noise ratios (S/N) at full inhibition in the LC–BCD by injecting The performance of the LC–BCD system to quantitatively measure the potency of sEHis
sEHi  2 at a concentration of 100  µM while the on-line BCD was performed at several was tested by analysis of five known inhibitors at different concentrations in FIA mode,
enzyme/substrate concentration combinations. PHOME was applied at concentrations of which is a fast way to measure IC50 values if pure compounds are available. As shown in
4.5, 2.3 and 1.2 μM in combination with a sEH concentration of 4 nM. Concentrations of Figure 4A, the injection of sEHi 1 resulted in negative peaks in the BCD chromatogram with
40, 8 and 4 nM sEH were tested at a PHOME concentration of 4.5 μM. Finally, a mixture of increasing negative peak heights upon injecting increasing concentrations. The variance
4 nM sEH and 4.5 μM PHOME was incubated off-line for 30, 45, 60 or 90 min and directly in peak height between duplicate injections was generally lower than 10% (in 90% of 36
infused into the fluorescence detector in order to assess the fluorescence intensity at full samples). As previously shown for various enzymes the negative peak height in LC–BCD
substrate conversion. systems can be used to calculate the percentage of inhibition [18; 27]. Based on the
In contrast to the earlier developed end-point plate-reader assay, an on-line format resulting dose response curves (Figure 4B), it is possible to quantitatively rank the sEHis
requires a shorter incubation time. The BCD system was set up with a 5  min reaction by their potency. Among the compounds tested, sEHi 5 was the most active and sEHi 4 the
time in order to minimize band broadening in the LC–BCD system. In order to increase least active inhibitor. In order to deduce IC50 values for each compound, the dilution of the
the solubility of PHOME at room temperature, 0.1 g/L Tween 80 were added to the buffer injected amount of inhibitor in the LC–BCD system has to be taken into account [18; 28].
without measurable influence on the enzymatic conversion (data not shown). The substrate The dilution results from the mixing of LC eluent and BCD reagents (Dm) which depends
was used close to its limit of solubility (30  μM) in the superloop, thus with a substrate on the flow rates of eluent entering the on-line BCD (uE) and the total flow at detection (uF).
concentration of 4.5 µM in the reaction coil (Figure 2). This concentration is lower than the
Michaelis-Menten-constant (KM) of PHOME for sEH, which is above its limit of solubility [8]. uF
Thus, at a concentration of 4.5 µM of PHOME, the enzyme is not in saturating condition DM =
Equation 1
allowing the sensitive detection of inhibitors. Additional actions to shorten the reaction time uE
72 73
Table 1: IC50 values determined for sEHi 1-5 and comparison to literature values
Figure 4: Analysis of sEHi in FIA Compound LC–BCD system nMa Literature values Substrate
mode. A BCD signals of injections (10
µL) of eight different concentrations sEHi 1 19 (1) 29 ± 13 nM [7] PHOME
of sEHi  1 [blank (black), 0.5  μM sEHi 2 25 (1) 684 nM [22] CMNPCb
(blue), 1  μM (green), 2  μM (red),
sEHi 3 12 (2) 15 nM [23] CMNPCb
5  μM (violet), 10  μM (light blue),
20  μM (brown) and 50  μM (grey)]. sEHi 4 880 (90) 171 nM [22] CMNPCb
The percentage of sEH inhibition, sEHi 5 2.6 (0.6) 2 nM [23] CMNPCb
calculated from the negative peak
height, is plotted against the inhibitor a
Mean and difference (in brackets) from individual fitting of the duplicate curves.
concentration for five different b
cyano(2-methoxynaphthalen-6-yl)methyl trans-(3-phenyloxyran-2-yl)methylcarbonate
sEHis in panel B [sEHi 1 (●), sEHi 2
(■), sEHi  3 (▲), sEHi  4 (▼) and percentages of inhibition and the data fitted with GraphPadPrism (GraphPad Software,
2.3 sEHi  5 (♦)]. Mean and range of
a determination in duplicate are
La Jolla, US). This results in the sigmoidal dose-response curves shown in Figure  4B.
The reproducibility of the data points measured (see above) and the quality of the all fits,
2.3
shown. The concentration is given expressed by resulting R2-values of >0.975, are further indications of the high data quality.
as final concentration of the inhibitor The calculated IC50 values for the five compounds tested are given in Table 1. They covered
in the reaction coil, taking a dilution
factor into account.
a range of about three orders-of-magnitude. This demonstrates that the developed LC–
BCD method allows the measurement/detection of highly potent as well as weak sEHis.
The potency of sEHi 5 was remarkably high, with an apparent IC50 value of 2 nM. Given an
enzyme concentration of 4 nM in the reaction coil, this is the highest potency which can be
observed with the setup [9]. Most importantly, the newly developed LC–BCD method ranked
the potency of the tested inhibitors in the same order as commonly employed end-point
assays, except for sEHi 2 (Table 1). However, for few sEHis, the determined IC50 differed
significantly from literature values: For sEHi  2, the observed potency by LC–BCD was
about 20-fold higher compared to the value from an end-point assay, while the measured
IC50 value for sEHi 4 was about 4-fold higher than previously reported (Table 1). Similar
to our observations, up to 20-fold differences have been described between different sEH
assays, because the measured potency for individual sEHis is substrate-dependent [9].
This observation is substantiated with the results for sEHi 1. For this compound, using the
same substrate, a very good agreement was found between our data and the literature [7].
In addition, inhibitors injected are also diluted due to their residence time in the flow system The potencies for sEHi 3 and sEHi 5 are also consistent with literature values, despite use
(DC). In a test tube or well plate experiment, the inhibitor can be assumed to be evenly of different substrates (Table 1). Overall, the results from the analysis of individual sEHis in
distributed after mixing. The same is only true for the diagonal distribution in the on-line the LC–BCD system show, that the negative peak height is a suitable quantitative measure
BCDs. In the longitudinal dimension, which is reflected on the time axis, the inhibitor is for the potency of inhibitors, and that the data obtained are in good agreement with other
distributed according to a near-Gaussian distribution which is typical to chromatography. methods to characterize the potency of sEHis.
This phenomenon is a result of longitudinal diffusion of the initially homogeneous injection In contrast to end-point assays, the LC–BCD system combines identity and activity detection
plug (Vi). It further dilutes the injected concentration (ci). The full width at half maximum after chromatographic separation. It thus allows assessment of individual compounds in
(FWHM) and the flow rate (uC) have to be derived from the same chromatogram, preferably mixtures. This is demonstrated by analysing a mixture of sEHi 6, sEHi 1, sEHi 7, and two
from the BCD chromatogram which results in uC = uF. compounds without sEH activity, diclofenac and phenylbutazone. In this case, the BCD
signal shows only three major peaks, which corresponded well with the elution times to
the three sEHis, whereas in the corresponding LC–UV or LC–MS all five compounds are
FWHM π uC observed (data not shown). These experiments show that the developed LC–BCD system
DC =
• • Equation 2
2 ln 2 Vi allows assessing the bioactivity of individual compounds in mixtures. In only one analysis
step, it can distinguish between active and non-active compounds in mixtures.
Therefore, the final concentration at the maximum negative peak height (cF), which can be
calculated from equation 3, was used for the dose-response curves. Application of LC-BCD on the analysis of metabolic mixtures of sEHis
In order to demonstrate the applicability of the LC–BCD system to the analysis of unknown
mixtures, in vitro microsomal incubations of three sEHis were analysed. The LC–MS
ci data showed that each compound was metabolized to several metabolites (Figure  5).
cF =
Equation 3
DM • DC By combining BCD traces and MS extracted ion chromatograms, the peaks of active
compounds can be directly identified and structurally characterized.
Thus, the dilution factors are calculated individually for every measurement, and they For sEHi  6, three bioactivity peaks were observed (Figure  5A). The main peak eluting
range from 71 to 210, The final concentrations were plotted against the corresponding around 43.2 min in LC–MS, peak A4MS, corresponding to peak AIIIBCD in LC–BCD, is

74 75
the parent compound (protonated molecule [M+H]+ with m/z 277.228). The three peaks peaks CIIBCD and C3MS correspond to the parent compound ([M+H]+ with m/z 393.312).
A1MS, A2MS and A3MS, eluting at 35.0 min, 35.8 min and 38.0 min, respectively, in LC–MS, The two metabolites C1MS and C2MS are not well separated and result in only one peak in
were not observed in the control incubations (data not shown). These compounds could the LC–BCD chromatogram. The retention time of C1MS corresponds to the peak CIBCD,
be tentatively identified as hydroxylated metabolites because all contained an additional indicating the compound is bioactive, but the increased tailing of CIBIO suggests that C2 is
oxygen compared to the parent compound ([M+H]+ with m/z 293.223). While peak A1MS is bioactive as well.
not bioactive, peaks A2MS and A3MS correspond to the peaks AIBCD and AIIBCD, respectively. All three sEHis were metabolized in the aliphatic chains and rings at either side of the urea
Note that peak A3MS consists of three non-separated compounds thus three different mono- function. A more detailed structural analysis was not possible as MS fragmentation only
hydroxylated metabolites with m/z 293.209, which were not well separated. The peaks occurred in or next to the urea function. For all three sEHis tested, LC–BCD/MS analysis
A5MS and A6MS are present as contaminants; they shows the same nominal mass as the allowed the tentative identification of at least two inhibitory active metabolites. In only a
oxygenated metabolites and the parent sEHi 6, but different accurate mass (m/z 293.209 single step analysis of 60 minutes, active metabolites can be detected. Moreover, it is
and 277.217, respectively). possible to distinguish between active and non-active metabolites and to characterize
The LC–BCD chromatogram of the metabolic incubation trace of sEHi  1 showed four bioactive compounds by their inhibitory potency and MS spectra.
peaks (Figure 5B). The main peak BIVBCD corresponds to peak B9MS, the parent compound
2.3 ([M+H]+ with m/z 227.214). In this case, six mono-hydroxylated metabolites (B2MS through
B7MS, [M+H]+ with m/z 243.208) were observed (Figure 5B). These metabolites gave rise
Conclusion 2.3
to peaks BIBCD and BIIBCD. By careful evaluation of the peak shapes and retention times, it A new LC–BCD system for the detection of sEHis in complex mixtures has been developed.
may be concluded that B2MS and at least two of the metabolites B4MS to B7MS are bioactive. The detection principle is a continuous-flow enzyme activity assay coupled on-line to LC
The oxidated dehydrogenated metabolite B1MS ([M+H]+ with m/z 241.192) is clearly not with parallel MS detection. The substrate PHOME allowed sensitive and robust monitoring
bioactive, whereas the peak shape of the dehydrogenated metabolite B8MS ([M+H]+ with m/z of bioactivity by fluorescence. After thorough optimization of the assay conditions, the
225.197) matched the retention time of BIIIBCD. This is an excellent example of the added incubation time in the BCD was reduced from 60 to 5.5 min. With a sEH concentration
value of the LC–BCD approach: whereas the minor peak B8MS would be easily ignored in of only 4 nM in the reaction coil, the signal-to-noise ratio for complete sEH inhibition was
an MS-only approach, its strong corresponding peak BIIIBCD cannot be overlooked. still higher than 60. Analysis of several known sEHis demonstrated that the peak height,
Compared to sEHi 6 and sEHi 1, sEHi 7 showed less metabolic conversion: only two mono- observed in LC–BCD can be used as quantitative measure for sEH inhibition. Moreover,
hydroxylated metabolites were detected ([M+H]+ with m/z 409.306) (Figure 5C). The main the obtained potencies, measured as IC50 values, for sEHis are in good agreement with
previously reported values. The LC–BCD system is able to perform bioactivity analysis of
individual compounds in mixtures. This was successfully demonstrated by the analysis
of a standard mixture as well as of in vitro metabolic conversions of three known sEHi
containing both active and non-active metabolites towards sEH. Here, LC–BCD revealed
the formation of new active metabolites, which could be simultaneously characterized by
LC–MS.
With the developed LC–BCD system, inhibitors can be detected and characterized in a
single analysis. Given the increasing interest in sEH as drug-target for various diseases,
this new technique may pave the route for the detection of new classes of sEHis in natural
products or crude mixtures arising from organic synthesis. Moreover, metabolism studies
with LC–BCD as read out will allow the identification of active metabolites in early stages
of lead development and thus assist the identification of the best compounds as drug
candidates.

Acknowledgements
This study was supported by NIEHS (R01 ES002710, P42 ES004699), NIOSH (PHS
OH07550) and the German Academic Exchange Service. B.D.H is as senior fellow of
the American Asthma Society. We acknowledge Lionel Blanchet for his help with data
management.

References
1. Newman JW, Morisseau C, Hammock BD (2005) Epoxide hydrolases: their roles and interactions
with lipid metabolism, Prog Lipid Res 44: 1-51.
2. Certikova Chabova V, Walkowska A, Kompanowska-Jezierska E, Sadowski J, Kujal P, Vernerova
Z, Vanourkova Z, Kopkan L, Kramer HJ, Falck JR, Imig JD, Hammock BD, Vaneckova I,
Figure 5: Analysis of oxidative microsomal incubations of three sEH inhibitors by the LC–BCD/MS Cervenka L (2011) Combined inhibition of 20-hydroxyeicosatetraenoic acid formation and of
system. In each panel, the LC–BCD chromatogram (blue line) is combined with the MS trace (black, epoxyeicosatrienoic acids degradation attenuates hypertension and hypertension-induced end-
red, green). (A) sEHi 6, (B) sEHi 1, and (C) sEHi 7. The peaks are labelled in order of retention time organ damage in Ren-2 transgenic rats, Clin Sci (Lond) 118: 617-632.
with Arabic (MS) or Latin (BCD) numbers. 3. Schmelzer KR, Kubala L, Newman JW, Kim IH, Eiserich JP, Hammock BD (2005) Soluble
76 77
epoxide hydrolase is a therapeutic target for acute inflammation, Proc Natl Acad Sci USA 102: 24. Morisseau C, Goodrow MH, Newman JW, Wheelock CE, Dowdy DL, Hammock BD (2002)
9772-9777. Structural refinement of inhibitors of urea-based soluble epoxide hydrolases, Biochem
4. Inceoglu B, Wagner K, Schebb NH, Morisseau C, Jinks SL, Ulu A, Hegedus C, Rose T, Brosnan Pharmacol 63: 1599-1608.
R, Hammock BD (2011) Analgesia mediated by soluble epoxide hydrolase inhibitors is dependent 25. Falck D, de Vlieger JSB, Giera M, Honing M, Irth H, Niessen WMA, Kool J (2012) On-line
on cAMP, Proc Natl Acad Sci USA 108: 5093-5097. electrochemistry-bioaffinity screening with parallel HR-LC-MS for the generation and
5. Imig JD, Zhao X, Zaharis CZ, Olearczyk JJ, Pollock DM, Newman JW, Kim IH, Watanabe T, characterization of modified p38alpha kinase inhibitors, Anal Bioanal Chem 403: 367-375.
Hammock BD (2005) An orally active epoxide hydrolase inhibitor lowers blood pressure and [Chapter 3.1]
provides renal protection in salt-sensitive hypertension, Hypertension 46: 975-981. 26. Kool J, Ramautar R, van Liempd SM, Beckman J, de Kanter FJJ, Meerman JH, Schenk T, Irth H,
6. Imig JD, Hammock BD (2009) Soluble epoxide hydrolase as a therapeutic target for Commandeur JNM, Vermeulen NPE (2006) Rapid on-line profiling of estrogen receptor binding
cardiovascular diseases, Nat. Rev. Drug Discovery 8: 794-805. metabolites of tamoxifen, J Med Chem 49: 3287-3292.
7. Wolf NM, Morisseau C, Jones PD, Hock B, Hammock BD (2006) Development of a high- 27. Kool J, Eggink M, van Rossum H, van Liempd SM, van Elswijk DA, Irth H, Commandeur JNM,
throughput screen for soluble epoxide hydrolase inhibition, Anal Biochem 355: 71-80. Meerman JH, Vermeulen NPE (2007) Online biochemical detection of glutathione-S-transferase
8. Jones PD, Wolf NM, Morisseau C, Whetstone P, Hock B, Hammock BD (2005) Fluorescent P1-specific inhibitors in complex mixtures, J Biomol Screen 12: 396-405.
substrates for soluble epoxide hydrolase and application to inhibition studies, Anal Biochem 28. Schebb NH, Heus F, Saenger T, Karst U, Irth H, Kool J (2008) Development of a countergradient
2.3 9.
343: 66-75.
Schebb NH, Huby M, Morisseau C, Hwang SH, Hammock BD (2011) Development of an online
parking system for gradient liquid chromatography with online biochemical detection of serine
protease inhibitors, Anal Chem 80: 6764-6772. 2.3
SPE-LC-MS-based assay using endogenous substrate for investigation of soluble epoxide
hydrolase (sEH) inhibitors, Anal Bioanal Chem 400: 1359-1366.
10. Ulu A, Davis BB, Tsai HJ, Kim IH, Morisseau C, Inceoglu B, Fiehn O, Hammock BD, Weiss
Supplementary Material
RH (2008) Soluble epoxide hydrolase inhibitors reduce the development of atherosclerosis in
apolipoprotein e-knockout mouse model, J Cardiovasc Pharmacol 52: 314-323.
11. Ebada SS, Edrada RA, Lin W, Proksch P (2008) Methods for isolation, purification and structural LC–MS for metabolite identification was done either on a Bruker Daltonik (Bremen,
elucidation of bioactive secondary metabolites from marine invertebrates, Nat Protoc 3: 1820- Germany) micrOTOF-Q quadrupole time-of-flight hybrid MS, using the above described
1831. conditions, or using an ion-trap time-of-flight mass spectrometer (IT-TOF, Shimadzu, ‘s
12. Kool J, Giera M, Irth H, Niessen WMA (2011) Advances in mass spectrometry-based post- Hertogenbosch, the Netherlands). In the latter case, a 35-min gradient and a 100 × 2.1 mm
column bioaffinity profiling of mixtures, Anal Bioanal Chem 399: 2655-2668. Waters XBridge C18 column (3.5 μm particles) was used. In the Bruker quadrupole time-
13. Schebb NH, Faber H, Maul R, Heus F, Kool J, Irth H, Karst U (2009) Analysis of glutathione of-flight hybrid MS, an electrospray ionization (ESI) source operated in positive-ion mode
adducts of patulin by means of liquid chromatography (HPLC) with biochemical detection (BCD) at the following settings: capillary voltage 4500V; end plate offset -500V; nebulizer gas 1.8
and electrospray ionization tandem mass spectrometry (ESI-MS/MS), Anal Bioanal Chem 394: bar; drying gas 8.0 L/min at 200°C. For accurate mass measurements, the TOF settings
1361-1373. were automatically calibrated prior to the measurements by infusing a 5 mM sodium formate
14. Marques LA, Kool J, de Kanter FJJ, Lingeman H, Niessen WMA, Irth H (2010) Production and
solution. Mass spectra were collected between m/z 50 and 1000.
on-line acetylcholinesterase bioactivity profiling of chemical and biological degradation products
of tacrine, J Pharm Biomed Anal 53: 609-616. In the Shimadzu ion-trap time-of-flight mass spectrometer, the ESI source was operated
15. Kool J, van Liempd SM, Harmsen S, Beckman J, van Elswijk D, Commandeur JNM, Irth H, in positive-ion mode at a needle voltage of 4.5 kV, a temperature of 200°C on the source
Vermeulen NPE (2007) Cytochrome P450 bio-affinity detection coupled to gradient HPLC: on- heating block and the curved desolvation line, a drying gas pressure of 62 kPa and a
line screening of affinities to cytochrome P4501A2 and 2D6, J Chromatogr B 858: 49-58. nebulising gas flow-rate of 1.5 L/min. For accurate mass measurements, the IT-TOF
16. Van Liempd SM, Kool J, Meerman JH, Irth H, Vermeulen NPE (2007) Metabolic profiling of was externally calibrated on a weekly basis using sodium TFA clusters. MS spectra were
endocrine-disrupting compounds by on-line cytochrome p450 bioreaction coupled to on-line collected between m/z 150 and 650 and MS/MS spectra between m/z 100 and 650, both
receptor affinity screening, Chem Res Toxicol 20: 1825-1832. with 10 milliseconds ion accumulation. The precursors were selected data-dependent with
17. de Vlieger JSB, Kolkman AJ, Ampt KA, Commandeur JNM, Vermeulen NPE, Kool J, Wijmenga a width of 3 Da and fragmented at 50% CID energy.
SS, Niessen WMA, Irth H, Honing M (2010) Determination and identification of estrogenic The mass accuracy was better than 5 ppm on both instruments. The accurate-mass
compounds generated with biosynthetic enzymes using hyphenated screening assays, high
resolution mass spectrometry and off-line NMR, J Chromatogr B 878: 667-674.
data obtained were used to determine the elemental composition of the metabolites and
18. Falck D, de Vlieger JSB, Niessen WMA, Kool J, Honing M, Giera M, Irth H (2010) Development accordingly of the fragments. This was done with the software provided by the respective
of an online p38alpha mitogen-activated protein kinase binding assay and integration of LC–HR- instrument manufacturers, ESI Compass 1.3 from Bruker Daltonik and LCMSsolution
MS, Anal Bioanal Chem 398: 1771-1780. [Chapter 2.2] 3.50 from Shimadzu. The relevant elemental composition in each case was extracted by
19. Beetham JK, Tian T, Hammock BD (1993) cDNA cloning and expression of a soluble epoxide applying additional restrictions next to the mass accuracy. It should not contain more atoms
hydrolase from human liver, Arch Biochem Biophys 305: 197-201. of any kind than the original compound except for oxygens which are known to be added by
20. Kim IH, Morisseau C, Watanabe T, Hammock BD (2004) Design, synthesis, and biological activity phase I metabolism. For the fragments, the restriction followed the elemental composition
of 1,3-disubstituted ureas as potent inhibitors of the soluble epoxide hydrolase of increased of the parent ion. Both assumptions are reasonable because the metabolites are linked
water solubility, J Med Chem 47: 2110-2122. to the original inhibitor by well-known metabolic pathways while the fragments can only
21. Jones PD, Tsai HJ, Do ZN, Morisseau C, Hammock BD (2006) Synthesis and SAR of arise from the parent ion selected for fragmentation. This proved sufficient to arrive at one
conformationally restricted inhibitors of soluble epoxide hydrolase, Bioorg Med Chem Lett 16:
5212-5216.
possible elemental composition.
22. Olearczyk JJ, Field MB, Kim IH, Morisseau C, Hammock BD, Imig JD (2006) Substituted
adamantyl-urea inhibitors of the soluble epoxide hydrolase dilate mesenteric resistance vessels,
J Pharmacol Exp Ther 318: 1307-1314.
23. Hwang SH, Tsai HJ, Liu JY, Morisseau C, Hammock BD (2007) Orally bioavailable potent soluble
epoxide hydrolase inhibitors, J Med Chem 50: 3825-3840.

78 79
Section 3 Chapter 3.1

Integration of modification On-line electrochemistry–


methods and HRS workflows bioaffinity screening with parallel
for the study of HR-LC–MS for the generation and
metabolite-like lead libraries characterization of modified p38α
kinase inhibitors

David Falck1 #, Jon S.B. de Vlieger1 #, Martin Giera1, Maarten Honing2,


Hubertus Irth1, Wilfried M.A. Niessen1, and Jeroen Kool1.

# Both authors equally contributed to this work

1
Division of BioMolecular Analysis, Faculty of Sciences, VU University Amsterdam,
De Boelelaan 1083, 1081 HV Amsterdam, the Netherlands
2
DSM Resolve, PO Box 18, 6160 MD Geleen, the Netherlands
Abstract chromatography (LC) - on-line p38α mitogen-activated protein kinase binding assay
(p38α bioaffinity assay) with parallel high resolution MS [18]. EC provides relatively clean
In this study, an integrated approach is developed for the formation, identification and samples and has shown to facilitate the formation of interesting molecules for drug research
biological characterization of electrochemical conversion products of p38α mitogen-activated [19]. The p38α mitogen-activated protein kinase (p38α kinase) is a prominent example
protein kinase inhibitors. This work demonstrates the hyphenation of an electrochemical of a drug target kinase [20] and is heavily involved in inflammation processes [21]. The
reaction cell with a continuous-flow bioaffinity assay and parallel LC-HR-MS. Competition hyphenation of these techniques to develop a fully integrated platform can facilitate the
of the formed products with a tracer (SKF-86002) that shows fluorescence enhancement in hit-to-lead selection process in drug discovery. This complete hyphenation of EC with LC
the orthosteric binding site of the p38α kinase is the readout for bioaffinity. Parallel HR-MSn and ultimately with parallel detection by p38α bioaffinity assay and high resolution MS
experiments provided information on the identity of binders and non-binders. Finally, the combines modification with separation, bioaffinity determination and structure elucidation
data produced with this on-line system were compared to off-line generated electrochemical on a new level of integration.
conversion products. The electrochemical conversion of 1-(6-chloro-5-((2R,5S)-4-(4-
fluorobenzyl)-2,5-dimethylpiperazine-1-carbonyl)-3aH-indol-3-yl)-2-morpholinoethane- Materials and methods
1,2-dione (DMPIP) resulted in eight products, three of which showed bioaffinity in the
used continuous-flow p38α bioaffinity assay. Electrochemical conversion of BIRB796 Chemicals
resulted, amongst others, in the formation of the reactive quinoneimine structure and its Acetonitrile, methanol (LC–MS grade) and formic acid (ULC–MS grade) were obtained
corresponding hydroquinone. Both products were detected in the p38α bioaffinity assay, from Biosolve (Valkenswaard, The Netherlands). Water was produced by a Milli-Q device
which indicates binding to the p38α kinase. of Millipore (Amsterdam, The Netherlands). Nitrogen 5.0 was purchased from Praxair
3.1 Introduction
(Vlaardingen, The Netherlands) and used in all MS experiments. SKF-86002 (SKF) was
delivered by Merck KGaA (Darmstadt, Germany). Enzyme-linked immunosorbent assay
3.1
(ELISA) blocking reagent was purchased from Roche Diagnostics (Mannheim, Germany).
Electrochemistry (EC) is increasingly described as a tool to support the formation and Ammonium acetate and ammonium hydrogen carbonate were obtained from Mallinckrodt
identification of drug oxidation products [1]. Efforts are reported to mimic a human Baker (Deventer, the Netherlands). Fused silica tubing (250-mm inner and 375-mm outer
metabolite profile [2] or to produce specific reactive metabolite species [3]. The application diameter) covalently coated with polyethylene glycol was obtained from Sigma-Aldrich
of electrochemical techniques specifically focused at drug development is reviewed by (Schnelldorf, Germany). Human recombinant p38α kinase, BIRB796, TAK715, 1-(6-chloro-
Hillard et al [4]. These applications are mainly directed at the study of reactive oxygen 5-((2R,5S)-4-(4-fluorobenzyl)-2,5-dimethylpiperazine-1-carbonyl)-3aH-indol-3-yl)-2-
species, oxidation and reduction of prodrugs, and the alkylation of DNA. Bauman and Karst morpholinoethane-1,2-dione (DMPIP) and SB203580 were a kind gift of MSD Research
[5] recently described the principles and use of on-line electrochemistry-mass spectrometry Laboratories (Oss, the Netherlands).
(EC-MS) in drug metabolism studies. Jurva et al. systematically compared the oxidation Structures of the kinase inhibitor standards
of drugs by EC-MS and by cytochrome P450s [6]. This approach provides chemical used can be found in Figure  1. All other
information on the products formed by electrochemistry and facilitates the comparison with chemicals were from Sigma-Aldrich
in-vitro incubation models [7, 8]. The chemical information obtained can eventually be used (Schnelldorf, Germany).
to correlate biological action of already characterized metabolites to the products analyzed.
However, this is not feasible for newly formed products for which no biological data are Instrumentation
yet available. Still, few examples in EC related drug metabolism studies employ detection A schematic representation of the complete
of biological activity [9, 10] and, to the best of our knowledge, none tested for a specific on-line setup is shown in Figure  2. The
target enzyme or receptor. More importantly, in studies employing EC-MS or EC-LC-MS, system consists of four modules: (A) an
biological testing seems to be a completely new development. This is surprising because electrochemical reaction cell, (B) an LC
the biological activity of the metabolites towards the drug target is of utmost importance for system, (C) a continuous-flow bioaffinity
the efficacy of a drug. For many years, we have been developing hyphenated screening assay unit, equipped with a fluorescence
assays to obtain chemical and biological information in a combined manner [11]. This detector, and (D) a mass spectrometer.
resulted in several approaches to assess bioaffinity, e.g., on-line receptor binding [12], A Roxy electrochemical reaction cell (Antec
enzyme activity assessment [13], bacterial growth inhibition [14], as well as several other Leyden, Zoeterwoude, the Netherlands)
strategies which allowed us to identify and characterize bioactive compounds [15-17] in equipped with a glassy carbon electrode
(complex) mixtures. These mixtures included natural extracts, crude synthesis products, was controlled by the Decade II Potentiostat
medicinal chemistry compound libraries, degradation products by light or harsh chemical either manually or under Dialogue software
conditions as well as in vitro metabolic incubations. control (Antec Leyden). The 10  mM kinase
The implementation of a device for electrochemical oxidation in our on-line screening inhibitor standards, dissolved in 25% ACN
platform would lead to a fully automated process of formation of drug-related chemical and 75% 1 mM aqueous buffer, were infused
entities followed by their simultaneous chemical and biological characterization. This leads at a flow rate of 5  mL/min with a Harvard
to a quick feedback between the modifications of a lead compound and their consequences Apparatus (Hollister, USA) syringe pump. The
for binding to the drug target. Furthermore, instable and/or reactive products could be conversion products were collected either in
analyzed directly after their formation and as such have less chance of degradation. an autosampler vial (“off-line” mode) or in
In this paper, we describe the hyphenation of EC with our recently developed liquid 100 mL volume PEEK tubing (“on-line” mode) Figure 1: Structures of the kinase inhibitors used
serving as an injection loop mounted in a for electrochemical conversion experiments
82 83
the-art LC pump delivered a constant flow of water. The pulse dampener was based on an
impermeable membrane and had a comparably extensive water reservoir. The restriction
capillary was chosen to produce ca 70 bar backpressure at the applied flow rate of 50 mL/
min. As a result, a constant and stable flow entered the superloop. The superloop served
as a reagent reservoir of constant volume which released exactly the same flow rate as
it received. Compared to a delivery by the pump alone, reagents were also more easily
cooled and pump maintenance was reduced which further improved the stability of the
system.
Parallel to the p38α bioaffinity assay, the electrochemical conversion products were
analyzed using a Shimadzu ion-trap time-of-flight hybrid mass spectrometer (LC-IT-TOF-
MS), equipped with an ESI source and operated in positive-ion mode to obtain accurate
m/z values for [M+H]+ and fragment ions. The ESI needle voltage was set to 4.5 kV, while
the source heating block and curved desolvation line temperature were set to 200°C.
Drying gas pressure was set at 62 kPa and nebulizing gas was applied at a flow-rate of
1.5 L/min. MS2 and MS3 spectra were acquired in data-dependent mode with the following
settings: full-spectrum MS with m/z 200 – 650, MS2 with m/z 150 – 650 and MS3 acquisition
with m/z 100 – 650 with ion accumulation time of 10  ms, precursor isolation width of
3.1 Figure 2: Scheme of the on-line setup. (1) On-line electrochemical conversion of inhibitor. (2)
Gradient LC separation of products formed. (3) Split of 1:9 to MS and p38α bioaffinity assay. (4)
3 Da and collision energy set at 75% for TAK715 and 50% for all other compounds. The
accurate-mass fragmentation data obtained were used for structure identification. External 3.1
Reaction coil for enzyme binding. (5) Addition of tracer molecule. (6) Detection of enzyme tracer calibration of the instrument was based on sodium TFA clusters and allowed a mass
complex by fluorescence and parallel HR–MSn for structural information of binders. accuracy lower than 5 ppm. From the measured accurate mass, the elemental composition
remote controlled six port valve (VICI, Schenkon, Switzerland). of the products and their fragment ions were calculated. In order to extract the relevant
In the off-line mode, collected fractions were injected via the autosampler, whereas in the elemental compositions, restrictions were applied to exclude those which have more atoms
on-line mode the six port valve with the 100 mL PEEK tubing acted as the injection valve to of any kind than the substrate plus possible modifications. This is reasonable because the
a Shimadzu LC system (‘s Hertogenbosch, the Netherlands), consisting of two LC-10ADvp products are linked to the substrate by electrochemical modification. For fragments, the
pumps, a DGU-14A solvent degasser, a SIL-10ADvp auto injector, two CTO-10ASvp column elemental composition of the precursor ion was used as restriction. The few compositions
oven, an SPD-10Avp UV-detector and a SCL-10Avp system controller. The LC separation left were analyzed for electrochemical modifications possibly leading to the formation of
was performed on a Waters Xbridge C18 column (2.1 mm x 100 mm I.D., 3.5 µm particles). products with a specific composition. Where CID fragments were concerned, this analysis
The mobile phases consisted of 99% water with 1% methanol (solvent A) and 1% water was based on reasonable neutral losses. The procedure was sufficient to restrict the
with 99% methanol (solvent B), both containing 0.01% formic acid. A generic gradient was discussion to only one elemental composition.
applied for the separation: isocratic for 2 min at 20% B, gradient to 90% B until 18 min,
isocratic at 90% B until 22 min, a gradient back to 20% B until 23 min and isocratic re- Optimization of electrochemical conversion
equilibration at 20% B until the end of the run at 30 min. The flow-rate was 113 mL/min. The For flow-through electrochemical reaction cells, important parameters reported are pH,
column temperature was kept at 40°C. All standards eluted with significant retention, which electrode voltage, organic modifier content and flow-rate [5, 6]. Several techniques can
provided sufficient resolution for the separation of products with slightly differing polarities. be used to find the optimal conditions. Those include cyclic voltammetry, direct EC-MS
Initial off-line experiments were performed with the EC unit and an LC-MS system, without coupling, and off-line EC and LC-MS experiments [5, 22]. The latter method was applied
the p38 bioaffinity assay attached, in order to optimize the EC conditions. For this, the LC in this study. We evaluated per substrate 4 different potentials (0; 0.4; 0.8; 1.2 and 1.5 V)
system was coupled via an electrospray ionization (ESI) interface to a Thermo Finnigan and 4 different pH values (3.5; 5.0; 7.0; 10.0). The four different buffers used to vary the pH
LCQ Deca ion trap mass spectrometer (LRMS) (Breda, the Netherlands). Experimental were: an ammonium formate / formic acid buffer with pH 3.5, an ammonium acetate / acetic
conditions for the ion trap MS were as follows: Capillary temperature 200°C, sheath gas acid buffer with pH 5.0, and two ammonium hydrogen carbonate buffers with pH 7.0 and
flow 45 arbitrary units, aux gas flow 5 arbitrary units, source voltage 5  kV and capillary 10.0, adjusted with formic acid and sodium hydroxide, respectively. Each series resulted
voltage 6 V. The instrument was used in positive-ion ESI mode for full spectra acquisition in 20 samples. The series was injected in duplicate, which due to the 30 min LC run time
between m/z 150 and 650. resulted in a well defined 10 hrs difference between the two duplicate analyses of the same
In on-line experiments with the complete setup, a post-column split was applied (see sample. This provided an indication of the stability of the products formed.
Figure 2), with 13 mL/min of the LC mobile phase being directed to the p38α bioaffinity From the off-line optimization data, optimum conditions were selected to be applied in the
assay and 100  mL/min to the MS system. The post-column fluorescence enhancement on-line mode, using the complete setup. These conditions are summarized for the three
based bioaffinity assay towards the p38α kinase was identical to the system previously model compounds in Table 1.
described by us [18]. Important features of the system are shown in Figure 2. Competition
of the formed products with a tracer (SKF) that shows fluorescence enhancement in the Results and discussion
orthosteric binding site of the p38α kinase is the readout for bioaffinity. Stability of the flow
system is essential for the quality of the assay as small changes in the reaction parameters In this study, an integrated approach is developed for the formation, identification and
have relatively pronounced consequences in the signal. Therefore, a stable reagent flow biological characterization of conversion products derived from p38α kinase inhibitors. This
is important in order to keep the noise to a minimum. This was achieved by a linear setup work demonstrates the hyphenation of an electrochemical reaction cell with a continuous-
of an LC pump, a pulse dampener, a restriction capillary and a superloop. The state-of- flow bioaffinity assay and parallel LC-HR-MS. The recently developed p38α bioaffinity
84 85
assay, based on fluorescence enhancement, was used as a model assay to investigate the possibly responsible for the bioaffinity signal. Fortunately, this case did not occur in the
possibilities of this combination, but the approach is not limited to this specific continuous- investigation. Please note that bioaffinity chromatograms were correlated with EIC at the
flow assay. Parallel HR-MSn experiments provide information on the identity of binders and front of the peak. In this way, peaks are more easily assessed when overloading of either
non-binders. Finally, the data produced with this on-line system were compared to that of readout leads to broad peaks with poorly defined maxima.
off-line generated electrochemical conversion products. The major affinity peak is due to the unmodified kinase inhibitor, DMPIP. The seemingly
First of all, the electrochemical conversion of the kinase inhibitors had to be optimized large broadening of this affinity peak is due to the sigmoidal dose-response character of
for substrate conversion and product formation. Formation and identification of bioaffinity the bioassay. Based on the bioaffinity profile, the product with m/z 539.1902 (D –2.011 u)
products is the ultimate goal of the method. High substrate conversion simplifies the bioaffinity showed to be the main bioactive product in the oxidative mode. However, in the reductive
mode, this product was less abundant and a product with m/z 472.1461 (D –69.0551 u)
Table 1: Optimized electrochemical conversion conditions for DMPIP, BIRB796 and SB203580. also showed significant binding to the p38α kinase. In both the reductive and the oxidative
Inhibitor EC conditions mode, a minor affinity peak was observed corresponding to hydroxylated DMPIP, with m/z
557.1962 (D +15.9950 u).
  pH potential Identification of the (bioactive) products was done using the HR-MSn data obtained in parallel
3.5 + 1.2 V   with the bioaffinity measurements. Based on the fragmentation of the unmodified kinase
DMPIP inhibitor, the sites of modification were identified as far as possible. The fragmentation of
10.0 + 1.2 V – 1.5 V
DMPIP allowed the annotation of characteristic fragments to be used for the determination
3.5 + 0.8 V   of the modification sites. Figure  4 shows the structure of DMPIP with the characteristic
BIRB796
3.1 10.0 + 0.8 V   parts of the molecule involved in the fragmentation reactions.
Important fragments observed in the MS/MS spectra of DMPIP are C+D with m/z 319.0480 3.1
5.0 + 1.5 V   and C with m/z 206.0003. Table 2 summarizes the identification study.
SB203580
10.0 + 1.5 V   Below, the MS structure elucidation of two ions is discussed in detail. A full description of

assessment by reducing the interference of the unmodified inhibitor. The off-line and on- 3.75
pH 10 oxidative
line approaches employ the same flow-through EC instrument which is operated under 3.70
the same experimental conditions in both cases. Therefore, the off-line data generated for pH 10 reductive

Affinity response in mV
validation of the on-line setup can be used to extract the optimal electrochemical reaction 3.65
pH 3.5 oxidative
conditions for both setups. An excerpt of the off-line generated data can be found in the 3.60
Online Resource figure  S2. Although the EC and LC-MS approach can be considered 3.55
laborious for reaction optimization only, it has advantages over other methods. Cyclic
voltammetry will not give any information on product formation and direct EC-MS analysis 3.50

can be strongly hampered by ion suppression effects as well as by unwanted up front 3.45
fragmentation.
3.40
Figure  3 shows an overlay of the bioaffinity traces of on-line electrochemistry-LC-p38α
bioaffinity assay experiments and the relevant extracted ion chromatograms (EIC) from 3.35

the simultaneous LC-MS experiments for the compound DMPIP (C28H30ClFN4O4, [M+H]+ 3.30
with m/z 541.2012). Clearly, bioactive products were formed under all conditions applied,
3.25
although in different relative concentrations. The four negative peaks in the affinity
chromatogram indicate binding/affinity of three conversion products to the p38α kinase by 1.60

MS response x 107
displacing the tracer ligand upon elution from the LC column. The affinity is linked in the 1.40
same figure to the EIC for identification of the binders. As the products showing affinity are
1.20
the same under all conditions, the EICs are derived from one measurement. As reported
previously, the retention times in the bioaffinity detection as well as in the MS detection are 1.00
very stable (<3% relative standard deviation) [18]. This is expectable with state-of-the-art LC 0.80 m/z 501.1699 (1.00)
equipment, even more so with reagent delivery through pump, pulse dampener, restriction
m/z 472.1461 (1.00)
capillary and superloop which ensures stability of the bioaffinity trace. An increase in peak 0.60
m/z 557.1962 (x 5.00)
tailing is the only difference expected between bioaffinity and MS chromatograms in terms 0.40
of chromatographic parameters. Even this only applies to highly nonpolar compounds as a 0.20
m/z 541.2012 (1.00)

result of the low organic modifier content of the bioaffinity assay. Therefore, the bioaffinity m/z 539.1902 (x 5.00)

and MS chromatograms are matched by their stable retention time difference. The validity 0.0 5.0 10.0 15.0 20.0 25.0 min
of this retention time difference can be double checked if a known binder is present in the
mixture, in this case the substrate of the EC. The correlation of a bioaffinity peak with an
Retention time in minutes
EIC peak is achieved with high certainty because a margin of twice the maximum relative
standard deviation of the retention time, in this case ca 30 s, is applied. If more than two Figure 3: Correlation of bioaffinity profiles (top) and HR-MS traces (bottom) of electrochemical
EIC peaks would be matched within ±30  s to a bioaffinity peak, both compounds were conversion products related to DMPIP.
86 87
Figure 4: Structure of DMPIP and characteristic parts for MS/MS identification of electrochemical
conversion products.
Table 2: Identification of electrochemical conversion products related to DMPIP based on high
resolution MSn experiments (for explanation: see text). MS fragmentation is not to be confused with
EC modifications (e.g., hydrolysis). For conclusions on nature and site of the EC modifications drawn
from the MSn data, please refer to the text. Binding to p38α kinase is defined as an S/N ratio of > 3 in
the p38α bioaffinity assay (see also figure 3).
Tr in min. [M+H]+ Change Molecular MSn
3.1 in
formula
formula AB C CD other
fragments
3.1
16.1 541.2012 - C28H31 ClFN4O4+ √ √ √ -
9.3 431.1481 -C7H6F C21H24ClN4O4 +
- √ - D
11.9 501.1699 -C3H4 C25H27ClFN4O4+ - - √ BC-C3H4
Figure 5: MS and MS/MS spectra of the electrochemical conversion product of DMPIP, with
12.8 525.1699 -CH4 C27H27ClFN4O4 +
-CH4 - √ - m/z 472.1461. Annotations related to the parent structure (figure 4) are included in the spectra.
14.0 472.1461 -C4H7N C24H24ClFN3O4+ √ √ -C4H7N -CO2 modification has to be in part B. A modification ‘across’ parts B and C can also be excluded,
15.6 557.1962 +O C28H31ClFN4O5 +
√ √ +O - as the connecting amide has no hydrogen atoms to lose and as any other possibility would
have to involve the formation of a new bond. This would require proximity which is hindered
15.6 529.1649 -C2H4 +O C26H27ClFN4O5+ - √ A, by the rigid structure of the C part and the connecting amide. The only likely crosslink
between B and C would produce the loss of HCl instead of H2. All other B-C crosslinks would
-CO, -H2O
lead to new carbon-carbon bonds which would block the formation of the CD fragment.
17.5 571.2118 +CH2O C29H33ClFN4O5 +
- - √ D, -H2CO Other products were identified in a similar way. The products of oxidation in part D (m/z
472.1461 and m/z 557.1962) as well as dehydrogenation in part B (m/z 539.1902) of DMPIP
19.6 539.1902 -2H C28H29ClFN4O4+ - - √ BC-2H all retained the affinity for the p38α kinase, see table 2. Products not showing bioaffinity
MS fragmentation is not to be confused with EC modifications (e.g., hydrolysis). For conclusions on include methoxylation of part A (m/z 571.2110, C29H33ClFN4O5+; see also below), cleavage
nature and site of the EC modifications drawn from the MSn data, please refer to the text. Binding to of methane from part B (m/z 525.1720, C27H27ClFN4O4+) and the loss of C3H4 from part B
p38α kinase is defined as an S/N ratio of >3 in the p38α bioaffinity assay (see also Figure 3). (m/z 501.1715, C25H27ClFN4O4+). The electrochemically induced cleavage of part A resulted
The products showing bioaffinity are underlined in gray. in a product with m/z 431.1476 (C21H24ClN4O4+).
The bioaffinity assessment of the products based on the inhibitor BIRB796 (C31H37N5O3,
the structure elucidation for all ions observed is avoided in order to focus the discussion [M+H]+ with m/z 528.2975) showed that next to BIRB796 itself, two bioactive electrochemical
on the innovative combination of techniques. As an example, the MS and MS/MS spectra conversion products were formed. Figure 6 shows the extracted ion chromatograms of the
for the bioactive product with m/z 472.1461 are given in figure  5. The m/z difference three bioactive compounds aligned with the bioaffinity trace. The two additional bioactive
compared to the parent compound is D –69.0551 u, consistent with a net loss of C4H7N. compounds were the main conversion products, being identified as the quinoneimine (m/z
The data indicate that the molecule is modified in the D part, see figure  4. Parts A+B 413.1985, C25H25N4O2+, D 115.0990 u) and the hydroquinone (m/z 415.2134, C25H27N4O2+,
and C are detected unmodified, whilst the fragment of C+D shows the net loss of C4H7N. D 113.0841 u), both being present in all conversion samples.
Fragmentation of the [M+H]+ additionally results in de neutral loss of CO2, leading to an Interestingly, Regan et al. described the structure-activity relationships of BIRB796 and
unmodified B+C fragment. The characteristic losses of C4H7N and CO2 prove the hydrolysis some of its fragments [23]. The hydroquinone was found to bind to the kinase, while the
of the amide in part D to a carboxylic acid, which is further supported by the MS3 fragments reactive quinoneimine was addressed neither in this publication nor elsewhere in literature.
(data not shown). Therefore, full structure elucidation could be achieved by MSn analysis The fact that we can detect bioaffinity of the reactive quinoneimine underlines the importance
for this product. For the product with m/z 539.1902, attributed to a dehydrogenation of of a quick and clean sample handling technology between formation and analysis of the
part B, absolute identification is not possible. The attribution is made by exclusion. As conversion products. In this respect, our on-line approach can be of added value with respect
fragment CD is observed unchanged, modification in parts C and D is excluded. Since to the assessment of the affinity of reactive species. Other electrochemical conversion
the dehydrogenation is observed in a BC fragment and C has already been excluded, the products of BIRB796 were not bioactive. These included a product with m/z 230.1652
88 89
(x10,000,000)
area of the reactive quinoneimine significantly decreased in the duplicate measurement
after 10 hrs (see Online Resource figure S2), whilst in the pH 7 samples the hydroquinone
3.5
was only present after 10 hrs. The products with m/z 230 and m/z 273 had an increased
peak area in all duplicate samples and can therefore be either considered as secondary
3.0 conversion products or degradation products. This is a direct consequence of the storage
and handling steps and can be avoided in the on-line approach presented in this paper.
2.5

Conclusion
Detector Response

2.0

Electrochemical conversion of drug molecules has already been demonstrated to be a


1.5
valuable tool to assist in drug metabolite identification studies. As an additional valuable
1.0
feature, we showed that electrochemistry coupled on-line with LC-MS/bioassay can be
528.2969 (0.20) used for the generation of small focused libraries of bioactive structures closely resembling
0.5 413.1995 (1.00) the parent structures and assessment of their bioaffinity profile. The hit rate in these
focused libraries can be significant. With the integration of direct affinity assessment of
0.0
415.2156 (1.00) the electrochemical conversion products, one can quickly determine critical positions
of a molecule influencing its affinity towards the target protein. Moreover, the on-line
3.1 3.1
2.5 5.0 7.5 10.0 12.5 15.0 17.5 20.0 22.5 25.0
hyphenation of formation with identification and affinity determination strategies allows
Retention time in minutes
the affinity assessment of reactive species since sample handling steps and times are
significantly reduced. The latter was clearly demonstrated by the affinity determination of
Figure 6: Correlation of bioaffinity profiles (top) and HR–MS traces (bottom) of electrochemical
conversion products related to BIRB796, conversion at pH 3.5 with 0.8 V the reactive quinoneimine of the BIRB796 inhibitor. All of these results stress the importance
of employing biological characterization as an integral part of EC studies concerned with
corresponding to C14H20N3+ and a low abundant product with m/z 273.1710 corresponding drug metabolism or lead discovery/optimization.
to C15H21N4O+, co-eluting with the unmodified BIRB796. Structure proposals can be found In principle, the on-line integrated setup could be expanded to an automated system
in the Online Resource figure S1. Oxidation at pH 7.0 and 10.0 resulted in two additional applicable in routine analysis. Instead of by infusion, the parent compounds could be flow-
products with m/z 386.1863 (C24H24N3O2+) and 411.1821 (C25H23N4O2+). injected into the electrochemical reaction cell. Solvent select valves could be used for
For SB203580 and its conversion products, affinity determination was hampered by the convenient and automated changing of the solvent composition and pH. Evidently, the
auto-fluorescence of SB203580 and its products at the wavelengths used for the detection subsequent electrochemical conversion can also be fully automated and controlled via the
of the enzyme-tracer complex in the p38α bioaffinity assay. Electrochemical conversion of appropriate controlling software during the analytical runs. The complete setup should be
SB203580 (C21H17FN3OS+, [M+H]+ with m/z 378.1071) resulted in the formation of single capable of implementation in fully automated and integrated analytical workflows in drug
(m/z 394.1026) and double oxygenated analogues (m/z 410.0969). The main product discovery laboratories.
could be identified as the sulfone product based on its fragmentation pattern described
previously by Henklova et al [24]. The rather scarce fragmentation of SB203580 did not Acknowledgements
allow proposing structures of the other products generated by electrochemical conversion.
In order to compare the on-line formation with the conventional off-line generated This research was performed within the framework of project D2-102 ‘Metabolic Stability
products, a set of off-line electrochemical conversion experiments was conducted. This Assessment’ of the Dutch Top Institute Pharma. We thank Junella Antonie for technical
included variation of pH and electrochemical potential for the conversion of the inhibitors. assistance. The invaluable discussions with the analytical chemistry group of the Münster
The samples were measured twice, once immediately after generation and once with an University, especially Uwe Karst and Anne Baumann, are gratefully acknowledged. Further
additional storage time of 10 hours. In this way, a focus was set on the effects of storage as thanks go to Jean-Pierre Chervet and Agnieszka Kray of Antec Leiden B.V. for the initial
a major difference between the off-line and on-line approach. For DMPIP and SB203580, loan of the Roxy EC system and continued technical support.
no differences were observed in the conversion profiles between off-line and on-line
conversion and analysis. This means that product formation is comparable between the
two modes. The results of the off-line experiments also help to explain the origin of the References
methoxylation product for DMPIP. It may be suggested that the methoxylation does not 1. Nouri-Nigjeh E, Bischoff R, Bruins AP, Permentier HP (2011) Electrochemistry in the mimicry of
take place in the EC cell, but rather via a reaction of a reactive intermediate with methanol oxidative drug metabolism by cytochrome p450s. Curr Drug Metab 12 (4):359-371
2. Thevis M, Lohmann W, Schrader Y, Kohler M, Bornatsch W, Karst U, Schanzer W (2008) Use
in the LC mobile phase. However, the off-line experiments provide evidence against this. of an electrochemically synthesised metabolite of a selective androgen receptor modulator for
The off-line data, shown in figure S3, are not consistent with an intermediate reaction mass spectrometry-based sports drug testing. Eur J Mass Spectrom 14 (3):163-170
with the LC eluent, because, if it would be reacting, the amount of methoxylated product 3. Madsen KG, Gronberg G, Skonberg C, Jurva U, Hansen SH, Olsen J (2008) Electrochemical
should be the same or less after storage (depending on the stability of the intermediate). oxidation of troglitazone: Identification and characterization of the major reactive metabolite in
However, the data actually show a higher abundance of methoxylated product after storage liver microsomes. Chem Res Toxicol 21 (10):2035-2041
which indicates a reaction of an intermediate within the EC reaction mixture. Furthermore, 4. Hillard EA, de Abreu FC, Ferreira DCM, Jaouen G, Goulart MOF, Amatore C (2008)
if a reaction would take place during the chromatographic step, the peak shape of the Electrochemical parameters and techniques in drug development, with an emphasis on
methoxylated product would be seriously deteriorated. This is not observed. For BIRB796, quinones and related compounds. Chem Commun (Cambridge, U K) 23 (23):2612-2628
differences were observed between off-line and on-line conversion products. The peak 5. Baumann A, Karst U (2010) Online electrochemistry/mass spectrometry in drug metabolism

90 91
studies: Principles and applications. Expert Opin Drug Metab Toxicol 6 (6):715-731 J, Maurel P, Pavek P, Dvorak Z (2008) Sb203580, a pharmacological inhibitor of p38 map
6. Jurva U, Wikstrom HV, Weidolf L, Bruins AP (2003) Comparison between electrochemistry/ kinase transduction pathway activates erk and jnk map kinases in primary cultures of human
mass spectrometry and cytochrome p450 catalyzed oxidation reactions. Rapid Commun Mass hepatocytes. Eur J Pharmacol 593 (1-3):16-23
Spectrom 17 (8):800-810
7. Baumann A, Lohmann W, Schubert B, Oberacher H, Karst U (2009) Metabolic studies of
tetrazepam based on electrochemical simulation in comparison to in vivo and in vitro methods.
J Chromatogr A 1216 (15):3192-3198
8. Tong W, Chowdhury SK, Su AD, Alton KB (2010) Quantitation of parent drug and its unstable
metabolites by in situ coulometric oxidation and liquid chromatography-tandem mass
spectrometry. Anal Chem 82 (24):10251-10257
9. Goyal RN, Garg DK (1996) Electro-oxidation of the neurotoxin 5,6-dihydroxytryptamine and
effects of its oxidation products in mice. Bioelectrochem Bioenerg 39 (2):249-257 Supplementary Material
10. Felim A, Urios A, Neudorffer A, Herrera G, Blanco M, Largeron M (2007) Bacterial plate
assays and electrochemical methods: An efficient tandem for evaluating the ability of catechol-
thioether metabolites of mdma (“Ecstasy”) to induce toxic effects through redox-cycling. Chem
Res Toxicol 20 (4):685-693
11. Kool J, Giera M, Irth H, Niessen W (2011) Advances in mass spectrometry-based post-column
bioaffinity profiling of mixtures. Anal Bioanal Chem (399):2655-2668
12. de Vlieger JSB, Kolkman AJ, Ampt KAM, Commandeur JNM, Vermeulen NPE, Kool J,
3.1 Wijmenga SS, Niessen WMA, Irth H, Honing M (2010) Determination and identification of
estrogenic compounds generated with biosynthetic enzymes using hyphenated screening 3.1
assays, high resolution mass spectrometry and off-line nmr. J Chromatogr B 878 (7-8):667-674
13. Marques LA, Kool J, de Kanter F, Lingeman H, Niessen W, Irth H (2010) Production and on-
line acetylcholinesterase bioactivity profiling of chemical and biological degradation products of
tacrine. J Pharm Biomed Anal 53 (3):609-616
14. Giera M, de Vlieger JSB, Lingeman H, Irth H, Niessen WMA (2010) Structural elucidation of
biologically active neomycin n-octyl derivatives in a regioisomeric mixture by means of liquid
chromatography/ion trap time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 24
(10):1439-1446
15. Jonker N, Kool J, Krabbe JG, Retra K, Lingeman H, Irth H (2008) Screening of protein-ligand
interactions using dynamic protein-affinity chromatography solid-phase extraction-liquid
chromatography-mass spectrometry. J Chromatogr A 1205 (1-2):71-77
16. de Boer AR, Alcaide-Hidalgo JM, Krabbe JG, Kolkman J, Boas CNV, Niessen WMA, Lingeman
H, Irth H (2005) High-temperature liquid chromatography coupled on-line to a continuous-flow
biochemical screening assay with electrospray ionization mass spectrometric detection. Anal
Chem 77 (24):7894-7900
17. Heus F, Giera M, de Kloe GE, van Iperen D, Buijs J, Nahar TT, Smit AB, Lingeman H, de Esch
IJP, Niessen WMA, Irth H, Kool J (2010) Development of a microfluidic confocal fluorescence
detection system for the hyphenation of nano-lc to on-line biochemical assays. Anal Bioanal Figure S1: Structure proposals for the BIRB796 related conversion products. The main products
Chem 398 (7-8):3023-3032 are the hydroquinone and the quinoneimine. Both showed bioaffinity. The bioaffinity detection of the
18. Falck D, de Vlieger JSB, Niessen WMA, Kool J, Honing M, Giera M, Irth H (2010) reactive quinoneimine species was especially noteworthy. Furthermore, two deamination products
Development of an online p38 alpha mitogen-activated protein kinase binding assay and with m/z 273.1710 and 230.1652 were observed which did not show bioaffintity. In the case of
integration of LC–HR-MS. Anal Bioanal Chem 398 (4):1771-1780 [Chapter 2.2] BIRB796, the MS structure elucidation of the breakdown products is very precise. The elemental
19. Sperry JB, Whitehead CR, Ghiviriga I, Walczak RM, Wright DL (2004) Electrooxidative compositions explain only the modification to one specific isomer in contrast to for example the
coupling of furans and silyl enol ethers: Application to the synthesis of annulated furans. J Org hydroxylation product of DMPIP where multiple isomers are possible.
Chem 69 (11):3726-3734
20. Zhang JY, Shen BF, Lin AN (2007) Novel strategies for inhibition of the p38 mapk pathway.
Trends Pharmacol Sci 28 (6):286-295
21. Schindler JF, Monahan JB, Smith WG (2007) P38 pathway kinases as anti-inflammatory drug
targets. J Dent Res 86 (9):800-811
22. Tahara K, Nishikawa T, Hattori Y, Iijima S, Kouno Y, Abe Y (2009) Production of a reactive
metabolite of troglitazone by electrochemical oxidation performed in nonaqueous medium. J
Pharm Biomed Anal 50 (5):1030-1036
23. Regan J, Capolino A, Cirillo PF, Gilmore T, Graham AG, Hickey E, Kroe RR, Madwed JR,
Moriak M, Nelson R, Pargellis CA, Swinamer A, Torcellini C, Tsang M, Moss N (2003)
Structure-activity relationships of the p38 alpha map kinase inhibitor 1-(5-tert-butyl-2-p-tolyl-
2h-pyrazol-3-yl)-3-[4-(2-morpholin-4-yl-ethoxy)naphthalen-1-yl]urea (birb 796). J Med Chem 46
(22):4676-4686
24. Henklova P, Vrzal R, Papouskova B, Bednar P, Jancova P, Anzenbacherova E, Ulrichova

92 93
Figure S2: Product profiles of BIRB796 for the off-line Figure S3: Product profiles of DMPIP
conversion. The x-axis gives the varying EC reaction for the off-line conversion. The
conditions being different pH and voltage. For every x-axis gives the varying EC reaction
pH/voltage pair and every ion, there are two rows in conditions being different pH and
the x-dimension, the left one representing the first voltage. For every pH/voltage pair and
measurement and the right one the measurement with every ion, there are two rows in the
an additional storage time of 10h. The y-axis gives the x-dimension, the left one representing
area of MS response in arbitrary units calculated from the first measurement (black) and the
the total ion current (A) or the extracted ion currents right one the measurement with an
(B and C). additional storage time of 10h (blue).
The y-axis gives the area of MS
A) The area of BIRB796 is calculated from the total ion response in arbitrary units calculated
current because of the coexistence of the single- and from the extracted ion currents.
double-charge protonated molecule. This indicated
stability of the MS response over time as well as the A) The area calculated from the MS
chemical stability of BIRB796. response of DMPIP is stable over time.
Only the samples at pH 3.5 at 0 and
B) The highest signals at low pH are obtained from 800  mV show a large deviation. As
m/z 413.1 (black and dark blue) and 415.2 (grey figure B and C show no response at

3.1 and light blue) later identified


these conditions, this is not relevant for
the analysis. 3.1
as the quinoneimine and the
hydroquinone, respectively.
Their signals are lower at
every pH in the x+10h sample B) The product with m/z 539.1 which
(dark and light blue) than in the showed the highest affinity in the
immediate measurements (black later on-line analysis is presented
and grey). This hints to instability here. It was identified as the
or reactivity which is in line with product of a dehydrogenation in the
the structures proposed. dimethylpiperazine ring of DMPIP. It
seems to be stable at high pH, but
instable at low pH.

C) The opposite effect is


observed for the analytes with C) The product of methoxylation
m/z 273.2 (black and dark (m/z 571.0) has a higher response
blue) and 230.2 (grey and after storage. As explained in
light blue). Their response is the Discussion, this indicates the
always higher after additional reaction of an intermediate with
storage (dark and light blue) the EC mixture and excludes
which implies their formation interference of the eluent. It also
by a secondary reaction seems to be pH dependent as the
or degradation step. The response is more similar at higher
apparent changes in the pH.
profile under off-line conditions
emphasize the need to avoid
liquid handling and storage
steps.

94 95
Chapter 3.2
Combination of biotransformation
by P450 BM3 mutants with on-line
post-column bioaffinity and mass
spectrometric profiling as a novel
strategy to diversify and charac-
terize p38α kinase inhibitors

David Falck2 #, Vanina Rea1 #, Jeroen Kool2, Frans J.J. de Kanter3,


Jan N.M. Commandeur1, Nico P.E. Vermeulen1, Wilfried M.A. Niessen2 and
Maarten Honing2,4
# Both authors contributed equally to this work.

1
Division of Molecular Toxicology, LACDR, Leiden/Amsterdam Center for Drug Research,
Faculty of Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam,
The Netherlands.
2
Division of BioMolecular Analysis, Faculty of Sciences, VU University Amsterdam, De
Boelelaan 1083, 1081 HV Amsterdam, The Netherlands.
3
Division of Organic Chemistry, Faculty of Sciences, VU University Amsterdam, De
Boelelaan 1083, 1081 HV Amsterdam, The Netherlands.
4
DSM Resolve, Urmonderbaan 22, 6160 MD Geleen, the Netherlands.
Abstract such as CYPs and glutathione S-transferases [16]. The combination of P450 BM3-mutants
as biocatalyst and a HRS platform has been previously used to study activity of metabolites
Mutants of bacterial Cytochrome P450 BM3 from Bacillus megaterium have gained increasing of norethisterone and the mycotoxin zearalenone to the ligand binding domains of (human)
interest to support drug metabolism studies by producing human relevant biotransformation estrogen receptors (hER) as drug targets [17,18].
products and/or to support drug discovery by producing libraries of new chemical entities Recently, a HRS methodology has been developed for the on-line screening of inhibitors
based on initial lead compounds. In this study, a library of 33 P450 BM3 mutants was used of p38α mitogen-activated protein kinase (p38α) with simultaneous structure elucidation by
to diversify TAK-715, a representative (highly lipophilic) inhibitor of p38α mitogen-activated high-resolution MS (HR-MS) [19]. The p38α is an important drug target in contemporary
protein kinase (p38α). To simultaneously determine the individual bioaffinity and identity of drug discovery because it plays a central role in inflammatory cellular signalling processes
the different products, an analytical high-resolution screening approach was used, based [20]. Several companies have reported preliminary human clinical results for p38α inhibitors,
on post-column on-line bioaffinity profiling with parallel mass spectrometric identification. but until now no drug has reached the market [21].
The screening of the product mixtures produced demonstrated that introducing mutations In the present study, a library of P450 BM3 mutants was used to prepare mixtures of
in the active site of the P450 BM3 resulted in different product profiles. Several P450 BM3 biotransformation products of the p38α inhibitor TAK-715 (N-[4-[2-ethyl-4-(3-methylphenyl)-
mutants were mimicking the metabolic profile obtained by human liver microsomes. The 1,3-thiazol-5-yl]-2-pyridyl]benzamide, see Figure 3) and to characterize these using the
major biotransformation products of TAK-715 could be produced with the most active P450 p38α HRS platform. The high turnover rates of P450 BM3s and the convenient large-scale
BM3 mutant in sufficient amounts to enable further structure elucidation by 1H-NMR and production and purification protocols for these enzymes allow semi-preparative production
quantification of their p38α affinity. of human relevant metabolites or new chemical entities for further pharmacological
profiling [1]. TAK-715 was one of the most potent p38α inhibitors from a series of synthetic
4-phenyl-5-pyridyl-1,3-thiazoles [22,23]. TAK-715 was advanced to clinical Phase II trials
Introduction for rheumatoid arthritis, but was discontinued [24]. Recently Verkaar et al. showed that
TAK-715 is able to inhibit the Wnt-3α-stimulated β-catenin signalling pathway [25] whose
A relatively new development in drug discovery is the application of biocatalysts for the
aberrations are associated with several malignancies, most notably cancer [26]. Therefore,
generation of new chemical entities or even unique compound libraries [1,2]. When
lead diversification of TAK-715 is still relevant. Previously reported lead optimization
3.2 using monooxygenase systems, such as the cytochrome P450 enzymes (CYPs), the
biotransformation products are particularly interesting in lead optimization as they can
of typical p38α inhibitors was performed using a library encompassing rather lipophilic
compounds. Application of the P450 BM3 mutant mono-oxygenases is anticipated to
3.2
show improved physicochemical properties, such as solubility, and target selectivity
improve the physicochemical properties of the lead compounds, while maintaining their
without significant loss of their pharmacological activity compared to the parent itself.
bioaffinity.
CYPs are also involved in the majority of drug metabolism reactions: in particular CYP1A2,
CYP2D6, CYP2C9, CYP2C19 and CYP3A4 are responsible for the bulk of metabolism
of known drugs in humans [3]. Because of the potential pharmacological activity of drug Materials and methods
metabolites, safety studies on circulating metabolites are required [4]. As a consequence,
organic synthesis is needed to obtain sufficient amounts of pure compound. Biosynthetical Chemicals
approaches are upcoming alternatives to organic synthesis. Biocatalysts, i.e., enzymes, Human recombinant p38α and TAK-715 were a kind gift of MSD Research Laboratories (Oss,
can perform reactions with a high degree of regio- and stereo- selectivity that often cannot the Netherlands). SKF86002 was delivered by Merck KGaA (Darmstadt, Germany). Fused
be achieved by organic synthesis [5,6]. Obviously, CYPs are emerging as biocatalysts due silica tubing (250-μm inner and 375-μm outer diameter) covalently coated with polyethylene
to their catalytic versatility and extremely wide substrate diversity [7,8]. Here, bacterial glycol (PEG) was purchased from Sigma-Aldrich (Schnelldorf, Germany). Methanol
CYPs are preferred to mammalian CYPs because they are soluble, show much higher (LC–MS grade) and formic acid (LC–MS grade) were from Biosolve (Valkenswaard, the
turnover rates and can be expressed at high levels in E. coli. A very promising candidate as Netherlands). All other chemicals were of analytical grade and were obtained from Sigma-
biocatalyst for the generation of large quantities of biotransformation products of drugs is Aldrich (Schnelldorf, Germany). Human liver microsomes (HLM) pooled from different
P450 BM3 (CYP102A1) from Bacillus megaterium. This enzyme shows the highest catalytic individual donors were obtained from BD Gentest TM (San Jose, USA) and contained 20
activity ever recorded for a CYP [9]. Recently, many research groups have succeeded in mg/mL protein (Cat. No. 452161).
expanding the substrate selectivity of P450 BM3 and improving its catalytic properties by
site-directed and/or random mutagenesis, as recently reviewed [10]. Expression and isolation of P450 BM3 mutants
Although several P450 BM3 mutants can perform oxygenation reaction with a high degree Expression of the CYP 102A1 mutants was performed by transforming competent E.Coli
of regio- and stereoselectivity [11,12], biosynthesis of drug metabolites using engineered BL21 cells with the corresponding pET28+-vectors, as described previously [27]. For
P450 BM3s can lead to the formation of multiple metabolites [1,13-15]. Structural expression, 600 mL Terrific Broth (TB) medium (24 g/L yeast extract, 12 g/L tryptone, 4 mL/L
identification and characterization of pharmacological and toxicological properties of the glycerol) with 30 mg/mL kanamicin was inoculated with 15 mL of an overnight culture. The
individual components in such mixtures requires elaborate semi-preparative fractionation. cells were grown at 175 rpm and 37°C until the OD600 reached 0.6. Then, protein expression
Alternatively, characterization of compounds in complex mixtures can be performed using was induced by the addition of 0.6 mM isopropyl-β-D-thiogalactopyranoside (IPTG). The
the high-resolution screening (HRS) approach, that combines analytical separation with temperature was lowered to 20°C and 0.5 mM of the heme precursor δ-aminolevulinic acid
post-column on-line bioaffinity profiling, based on enzyme inhibition or displacement was added. Expression was allowed to proceed overnight. Afterwards, cells were harvested
of tracer compounds, and parallel identity assessment with mass spectrometry (MS). by centrifugation (4600 × g, 4°C, 25 min), and the pellet was resuspended in 20 mL Kpi-
This approach has been successfully used for the identification of ligands/inhibitors of glycerol buffer (100 mM potassium phosphate (KPi) pH=7.4, 10% glycerol, 0.5 mM EDTA,
several drug targets, such as human estrogen receptors α and β, acetylcholinesterase, and 0.25 mM dithiothreitol). Cells were disrupted by French press (1000 psi, 3 repeats) and
acetylcholine binding protein, proteases and peptidases, and drug metabolizing enzymes the cytosolic fraction was separated from the membrane fraction by ultracentrifugation of
the lysate (120.000 × g, 4°C, 60 min). CYP concentrations were determined using a carbon
98 99
monoxide (CO) difference spectrum assay. comprised a LC–MS system from Shimadzu (‘s Hertogenbosch, the Netherlands), including
two LC-20AD and two LC-10AD isocratic pumps, an SIL-20AC, a CTO-20AC and a CTO-
Selection of the P450 BM3 mutant library 10AC column oven, an RF-10AXL fluorescence detector, an SPD-AD UV/VIS detector, and
In the present study, 33 mutants of P450 BM3 were selected which could be expressed at a CBM-20A controller coupled to an ion-trap time-of-flight hybrid mass spectrometer for HR-
good levels and which showed catalytic diversity towards a variety of drugs and steroids. MS equipped with an electrospray ionization (ESI) source. In short, the biotransformation
Mutants M01 (R47L, F87V, L188Q, E267V, G415S), M02 (R47L, L86I, F87V, L188Q, product mixtures were separated on an Xbridge C18 column 100×2.1  mm with 3.5  µm
N319T), M05 (R47L, F81I, F87V, L188Q, E267V, G415S) and M11 (R47L, E64G, F81I, particles (Waters, Milford, MA, USA) at 40°C and a flow rate of 113 µL/min.
F87V, E143G, L188Q, E267V, G415S) were previously constructed by a combination The gradient used was constructed by mixing the following mobile phases: eluent C (1 %
of three site-directed mutations and subsequent random-mutagenesis by error-prone methanol, 99 % water, and 0.01 % formic acid); eluent D (99 % methanol, 1 % water, and
PCR [13]. Mutants M01 and M11 where used as templates for additional site-directed 0.01 % formic acid). The gradient was programmed as follows: from 0 to 2 minutes isocratic
mutagenesis of active site residues, guided by available crystal structures of P450 BM3 at 20% D; from 2 to 18 minutes, linear increase of eluent D from 20% to 90%; from 18 to
and by computational modeling of the active site of P450 BM3 [28]. Positions which were 22 minutes, isocratic 90% D; from 22 to 23 minutes linear decrease from 90% to 20% of
selected for mutagenesis were residues 72, 74, 82, 87 and 437 which appear to be key eluent D; from 23 to 30 minutes re-equilibration isocratic at 20% eluent D. Post-column, the
active site residues which have profound influence on regio- and stereoselectivity of P450 flow was split in a ratio of 1:9 directing 13 µL/min to the bioaffinity detection and 100 µL/
BM3 mutants. min to UV/VIS and ESI-HR-MS analysis. The bioaffinity detection is based on competition
Position S72 and A74 are both located in the substrate binding channel [7,29] and have of analytes with a tracer showing fluorescence enhancement. Affinity towards p38α is
been shown to influence regioselectivity and activity [15,30]. The effect of a negatively assessed by mixing the analytes with 45 nM of enzyme and 630 nM of tracer, SKF86002.
charged residue (Asp or Glu) in both M01 and M11 in position 72 and 74 is evaluated in The readout occurred at excitation 355 nm and emission 425 nm, 15 nm bandwidth each,
this work. in the flow-through fluorescence detector. The UV/VIS detector was operated in dual
Mutation A82W was selected because it strongly influenced regioselectivity of steroid wavelength mode at 210 nm and 254 nm. For ESI, the needle voltage was set to 4.5 kV
hydroxylation when applied to M01 and M11 [11]. and the source heating block and the curved desolvation line were kept at 200°C. A drying
Mutations at position 87 have been extensively studied as the amino acid lies very close to gas pressure of 62 kPa and a nebulizing gas flow-rate of 1.5 L/min assisted the ionization.
3.2 the heme, according to available crystal structures of P450 BM3. In our previous studies, we
mutated this position in M11 to all possible amino acids, showing that mutation at position
Full spectra were obtained in the positive-ion mode between m/z 200 and 650. MS2 and
MS3 spectra were obtained in data-dependent mode between m/z 100 and 650 with an ion
3.2
87 has strong influence in the metabolism of alkoxyresorufins, and the regioselectivity of accumulation time of 10 ms, a precursor isolation width of 3 Da and a collision energy of
testosterone and clozapine metabolism [27,31]. 75%. For the analytes in the large-scale incubation mixture, MS2 and MS3 spectra were
Mutation of L437 was shown to have strong influence of regio- and stereoselectivity of generated with manual precursor selection and data-dependent analysis, respectively. The
α-ionone hydroxylation [12]. This position was mutated in M11 to the negatively charged IC50 determinations were done in flow-injection analysis (FIA) mode, which means the
residue Glu (L437E) and to three polar residues Asn (L437N), Ser (L437S), and Thr separation column was replaced by a low dead volume union (VICI, Schenkon, Switzerland).
(L437T), which differ in size and hydrogen bonding capabilities. The lower dead volume reduces analysis time and minimizes peak broadening compared
to a column elution without retention. In these experiments, an injection volume of 50 µL
Biotransformation of TAK-715 by P450 BM3 mutants and human liver microsomes was used to achieve adequate final concentrations for full inhibition. Negative peak heights
Incubations of TAK-715 were performed in 100 mM Kpi buffer pH 7.4 with 250 nM P450 were plotted against the corresponding final concentrations. The latter were calculated as
BM3 mutants. The final volume of the incubation was 200 μL, with 100 µM substrate described earlier [19].
concentration. The reactions were initiated by addition of a NADPH regenerating system
with final concentrations of 0.2 mM NADPH, 20 mM glucose-6-phosphate and 0.4 U/mL Large-scale production and isolation of biotransformation products by preparative
glucose-6-phosphate dehydrogenase. The reaction was allowed to proceed for 60 min HPLC
at 25°C and was terminated by the addition of 200 µL of cold methanol. For human liver The biotransformation products of TAK-715 were produced on a semi-preparative scale
microsomes (HLM), a final microsomal protein concentration of 5 mg/mL was used and by incubation with the most active P450 BM3 mutant as biocatalyst. A 100 mL reaction
these incubations were performed as described above, at 37°C instead of 25°C. Precipitated volume containing 1 µM P450 BM3, 100 µM TAK-715 and NADPH regenerating system (as
protein was removed by centrifugation (15 min, 14000 × g), and the supernatant was described above) was prepared in 100 mM KPi buffer at pH 7.4. The reaction was allowed
analyzed by UPLC using an Agilent Technologies 2000 system and a Zorbax Eclipse to continue for 5 h at 25°C. To achieve maximal conversion of TAK-715, the incubation
XDB-C18 column (1.8 mm, 50 mm × 4.6 mm; Agilent Technologies, USA). The gradient was supplemented every hour with 3 mL of 30 µM P450 BM3 M11 and 5 mL NADPH
used was constructed by mixing the following mobile phases: eluent A (0.8 % methanol, regenerating system (20× concentrated). The reaction mixture was extracted three times by
99 % water, and 0.2 % formic acid); eluent B (99 % methanol, 0.8 % water, and 0.2 % 100 mL ethyl acetate. The combined organic layers were collected in a round-bottom flask
formic acid) with a flow rate of 1 mL/min. The gradient was programmed as follows: from 0 and evaporated to dryness using a rotary evaporator. The residue was redissolved in 10
to 8 minutes linear increase of eluent B from 40% to 100%; from 8 to 9 minutes, isocratic mL of 50% MeOH/H2O and applied by manual injection on a preparative chromatography
100% B; from 9 to 9.5 minutes linear decrease from 100% to 40% of eluent B; from 9 to column Luna 5 µm C18 (250 × 100 mm i.d.) (Phenomenex, Torrance, CA, USA), which was
12 minutes isocratic 40% eluent B. The products and substrate were detected at 254 nm. previously equilibrated with 40% eluent B. A flow rate of 2 mL/min and a gradient using the
eluent A and B was applied for separation of formed TAK-715 biotransformation products.
HRS analysis of biotransformation products of TAK-715 using the LC-p38α kinase The gradient was programmed as follows: from 0 to 40 min linear increase of eluent B from
affinity/MS platform 40 to 100%; from 40 to 50 min isocratic 100% B, from 50 to 55 min linear decrease to 40%
The HRS analysis of the biotransformation products is conducted with an LC–enzyme B, and then re-equilibration was maintained until 65 min. Biotransformation products were
binding detection/MS platform which has been described previously [19]. The platform detected using UV detection at 254 nm and collected manually. Collected fractions were

100 101
first analyzed for purity and identity by the analytical UPLC. Fractions containing individual (Table S1). In particular, the mutants showing relatively low activity appeared to be highly
metabolites were evaporated to dryness under nitrogen stream and dissolved in 1 mL selective in the production of TAK-P5. The only mutant which showed a significantly
deuterium oxide to exchange acidic hydrogen atoms by deuterium atoms. Finally, after different profile was M11 L437N which was the only mutant to produce TAK-P6 as the
drying by a SpeedVac evaporator, the residues were redissolved in 500 µL of methanol-d4 major product (38%).
and 1H-NMR spectra were recorded at room temperature. 1H-NMR-analysis was performed Based on its high activity and its ability to produce most of the products at significant amounts,
on Bruker Avance 500 (Fallanden, Switzerland) at 500.23 MHz. Afterwards, the samples M11 was selected to further evaluate the affinity of the TAK-715 biotransformation products
were dried and redissolved in 30% MeOH and subjected to LC–enzyme binding detection/ towards p38α in the on-line HRS-affinity assay and to perform a large-scale incubation to
MS analysis. LogP values were calculated with ChemBioDraw Ultra version 12 from the obtain sufficient amounts of the products for 1H-NMR and further affinity assessments with
structures obtained. individual compounds.
From Table S1, it can also be concluded that the P450 BM3 mutants were able to form
almost all human-relevant metabolites although with different activities and with strongly
Results and discussion different product ratios. After 60 minutes of incubation, 33 ± 2 % of the substrate was
converted by HLM, corresponding to 25.2 nmol product/min/CYP. The main metabolite
Biocatalysed synthesis of lead compounds from TAK-715 by P450 BM3 mutants; was TAK-P7, which represented 29 ± 3% of the total metabolism, respectively. Metabolites
comparison to human liver metabolism TAK-P1, TAK-P2, TAK-P5 and TAK-P6 were formed at comparable amounts by HLM,
TAK-715 was incubated for 60 min with a library of 33 mutants of bacterial P450 BM3 ranging from 14 to 20% of the metabolites, whereas the minor metabolites TAK-P3, TAK-P4
and pooled human liver microsomes (HLM), and subsequently analysed by UPLC. The and TAK-P8 were formed at 3 to 5% of total metabolism.
results of these incubations, including the percentage of conversion of substrate and
profile of metabolites, are tabulated in Table S1 of the ESI. As shown in Figure 1, in total Parallel on-line bioaffinity assay and mass spectrometry of TAK-715 biotransforma-
eight metabolites were found after incubation of TAK-715 with HLM, encoded TAK-P1 to tion products produced by P450 BM3 M11 and HLM
TAK-P8 based on their order of elution, whereas six products were observed with P450 Although HLM incubations are not in the focus of this investigation, they produce the same
BM3 (TAK-P3 and TAK-P4 were not observed). The P450 BM3 only produced human products as BM3 M11 as well as a few other minor products. Affinity analysis was per-
3.2 relevant metabolites of TAK-715. The most active P450 BM3 were M11 and its mutants at formed for both HLM and P450 BM3 M11 incubations. Figure 2A shows the extracted-ion
and bioaffinity chromatograms for the TAK-715 metabolites produced by HLM, whereas
3.2
position 74 and 437. M11 produced TAK-P7 as major product, representing 42 ± 3% of the
total biotransformation. TAK-P5 was produced as 22 ± 2% of the total biotransformation, Figure 2B shows the same for the products generated by BM3 M11. Retention times differ
whereas TAK-P1, TAK-P2, TAK-P6 and TAK-P8 were formed at 13% or less. Mutation to from the chromatograms shown in Figure  1, because another HPLC system was used.
a negatively charged amino acid (Asp or Glu) or hydrogen bond donor (Thr) in positions However, the order of elution was unchanged. Next to TAK-715, four of its biotransforma-
74 or 437 of M11 has little to no effect on activity but changed the profile significantly by tion products and eight of its metabolites showed affinity towards p38α (Figure 2). Data on
producing threefold higher amounts of TAK-P1, making this the major product (Table S1). these compounds are summarized in Table S1.
In contrast, mutant M11 V87I did not produce any TAK-P1, making it more selective to As expected, TAK-715 (eluting at 39.5 min, [M+H]+ with m/z 400.151) showed a strong
produce TAK-P7 (60% of total biotransformation). bioaffinity. The second strongest decrease in fluorescence was found at 33 min, closely
TAK-P5 turned out to be the major product for 22 out of the 33 P450 BM3-mutants studied corresponding to TAK-P6 (32.7 min) and TAK-P7 (33.0 min). The lower resolution of the
HRS-system did not allow to attribute the bioaffinity signal to any of these metabolites.
MS analysis showed that TAK-P6 and TAK-P7 with m/z 416.146 and 430.123 correspond
to a mono-oxygenated TAK-715 and double oxygenated/dehydrogenated TAK-715,
respectively. The mono-oxygenated product TAK-P5 (31.4 min, m/z 416.146) and the
double oxygenated products TAK-P1 (23.0 min, m/z 432.138) and TAK-P4 (27.8 min) also
showed bioaffinity. TAK-P3 (24.3 min, m/z 430.123) only showed a very weak bioaffinity
signal. The products TAK-P2 (24.3 min, m/z 446.121) and TAK-P8 (34.5 min, m/z 416.146)
did not show significant bioaffinity.
Next to the eight metabolites previously found by UPLC, two additional metabolites were
identified based on small bioaffinity peaks in the HRS system. TAK-P9 (8.4 min, m/z
312.119) can be rationalized by hydrolysis of the amide-bond of one of the mono-
oxygenated TAK-715 products. TAK-P10 (17.4 min, m/z 448.138) apparently results from
triple oxygenation of TAK-715. The bioaffinity signal at 4.5 min appeared to be unrelated
to TAK-715.

Preparative scale biosynthesis of TAK-715 biotransformation products


To obtain sufficient amounts of TAK-715 biotransformation products for NMR structural
identification and affinity testing of isolated products to p38α, a large-scale (100 mL)
incubation was performed of TAK-715 with mutant P450 M11. Figure S1 of the ESI shows
Figure 1: Ultra-performance liquid chromatography chromatograms obtained after incubations of the preparative HPLC chromatogram of the product mixture obtained. After 5 hours of
TAK-715 (100 µM) incubated for 60 minutes with 200 nM P450 BM3 M11 (upper line) and 5 mg/mL incubation, with hourly additions of enzyme and cofactors, almost 90% conversion of 100
human liver microsomes (lower line). UV-Detection of metabolites was at 254 nm. μM of TAK-715 was obtained. LC–MS/MS measurements were performed to confirm the

102 103
identity of isolated products. The main product TAK-P1 (35.7 min) represents 39% of the
total biotransformation and corresponds to 1.5 mg of product formed. Two mono-oxygenated
products (TAK-P5 and TAK-P6, 42.6 min and 43.5 min) represent 27% (1.0 mg) and 12%
(0.4 mg) of the total biotransformation, respectively. TAK-P7 (43.8 min) represents 15%
of the total biotransformation (0.6 mg), whereas TAK-P2 (37 min) was formed at less than
5%. Interestingly, also a small amount of TAK-P4 was found in the large-scale incubation.
The relative product amounts obtained by large-scale incubation are slightly different from
that obtained in the screening experiment because of the longer incubation time (5h vs. 1h)
and higher enzyme/cofactor concentration. Also, the less efficient aeration of large-volume
incubations might contribute to the different profile.

Structure elucidation of TAK-715 biotransformation products


For five of the products of TAK-715 (m/z 400.148), sufficient material was obtained to
acquire a 1H-NMR spectrum, which together with the LC–MS/MS data allowed structure
elucidation. By comparison with the spectra and fragmentation patterns of TAK-715, it
was possible to assign the structural modifications for most of the products (Table 1). The
details of the 1H-NMR spectra can be found in Table S2 of the ESI. Data on interpretation of
the LC–MSn spectra are discussed in more detail elsewhere [32]. The proposed structures
of the products are summarized in Figure 3.
TAK-P5 and TAK-P6, both with m/z 416.146, indicate mono-oxygenation at different
positions. The integrals and coupling pattern of the 1H-NMR spectrum of TAK-P5 showed
that all 12 aromatic hydrogen atoms and the hydrogen atoms of the ethyl moiety were still
3.2 present (Table S2). The fact that the singlet of the benzylic methyl group was no longer
observed, whereas the chemical shifts of the aromatic protons were shifted upfield, indicates
3.2
that TAK-P5 results from hydroxylation of the benzylic methyl group. For TAK-P6, the
1
H-NMR-spectrum indicates hydroxylation at the methylene carbon of the ethyl side chain:
the triplet of the terminal methyl group in TAK-715 was changed to a doublet, whereas the
quartet of the original methylene group was shifted upfield and showed half the integral.
The secondary loss of acetaldehyde in the LC–MS/MS data of TAK-P6 was consistent with
hydroxylation at this position.
TAK-P7 (m/z 430.123) was the major metabolite formed by HLM. Its 1H-NMR spectrum
showed all aromatic hydrogen atoms and an unchanged ethyl side chain. Similar to TAK-P5,
the signal of the benzylic methyl group was not observed, whereas the signals of the

Table 1: Identification and characterization of TAK-715 biotransformation products


p38α kinase affinity (nM)
tr (min) a
code m/z of Structural IC50 95% Conf.
[M+H]+ modification interval
23 TAK-P1 432.138 + 2O 460 ± 165 240 to 890
24.3 TAK-P2 446.121 + 3O – 2H No affinity
25.2 TAK-P3 430.125 + 2O – 2H + (n.q.)b
27.8 TAK-P4 432.141 + 2O + (n.q.)
31.4 TAK-P5 416.146 +O 310 ± 33 250 to 380
32.7 TAK-P6 416.146 +O 38 ± 4 31 to 47
33 TAK-P7 430.123 + 2O – 2H 1500 ± 203 1100 to 1900
34.5 TAK-P8 416.146 +O No affinity
Figure 2: Correlation between biochemical detection (p38α kinase affinity trace, upper 8.4 TAK-P9 312.119 – C 7H 4O + O + (n.q.)
trace) and extracted ion chromatograms from the MS detection (lower traces) of oxidative 17.4 TAK-P10 448.138 + 3O + (n.q.)
metabolites of TAK-715 produced in incubations with (A) human liver microsomes (HLM), 39.4 TAK-715 400.151 71 ± 26 36 to 140
and (B) p450 BM3 mutant M11.
a
tret = retention time in the HRS analysis (Fig. 2A and 2B); b n.q., not quantified.

104 105
aromatic ring of the benzylic ring were strongly shifted upfield. This, in combination with the
LC–MS/MS data, indicates that the benzylic methyl group is metabolized to a carboxylate
group, e.g., by further oxidation of TAK-P5 by two successive oxidation reactions (Figure
3).
TAK-P1 (m/z 432.138, consistent with two sequential oxygenation reactions) showed a
1
H-NMR spectrum in which both the signals of the benzylic methyl group and the ethyl
side chain of TAK-715 were strongly affected, suggesting that this product results from
hydroxylation of both alkyl side chains. The coupling pattern and integrals of the signals of
the ethyl side chain show that hydroxylation occurred at the methylene group, similar to the
changes observed in the spectrum of TAK-P6.

Figure 3: Proposed structures of TAK-715 biotransformation products by P450 BM3 mutants and
Similarly, the 1H-NMR spectrum of TAK-P2 (m/z 446.138) showed strong changes in the
signals of the benzylic methyl group and the ethyl side chain. The signals of the ethyl
side chain point to hydroxylation at the methylene position, similar to TAK-P1 and TAK-P6,
whereas the signals of the benzylic group showed the same changes as observed in
TAK-P7, indicative for formation of a carboxylic acid by sequential oxidations (Figure 3).
For products TAK-P3, TAK-P4 and TAK-P9, no 1H-NMR data could be obtained; no P450
BM3-mutant produced these in sufficient amounts. Thus, no purification was attempted.
Structure elucidation for these products, based on the LC–MS/MS fragmentation [32], is
summarized in Table 1 and Figure 3. For metabolites TAK-P8 and TAK-P10, which result
from single and triple oxygenation reactions, respectively, no useful fragment ions were
found in LC–MS/MS.
Biotransformation of TAK-715 appeared to occur almost exclusively at its alkyl side chains.
3.2 TAK-P5 and TAK-P7 result from sequential oxygenation on the benzylic methyl group.
The intermediate aldehyde was not detected, suggesting that it extensively undergoes
3.2
further oxygenation to TAK-P7 and, to lesser extent, TAK-P3. Although conversion of the
alcohols to carboxylates is often catalysed by sequential alcohol dehydrogenase, aldehyde
dehydrogenase and oxidase reactions, conversion of TAK-P5 to TAK-P7 appears to be
fully catalysed by P450 BM3. So far, only few examples have been described on the
multistep oxidation of a methyl group to a carboxylic acid by CYPs. Oxidation of a methyl
of the t-butyl moiety of terfenatine and ebastine to their corresponding carboxylic acids
fexofenadine and carebastine in HLM was previously shown to be catalysed by CYP3A4
and CYP2J2 [33,34]. More recently, yet unidentified microbial P450s from Streptomyces
platensis were also shown to be capable to produce fenofexadine from terfenatine [35].
Similar to our P450 BM3 incubations, no intermediate aldehyde was found, indicating
that the conversion of aldehydes to acids by P450s is highly efficient. The availability of
biocatalysts for conversion of methyl groups to carboxylic acids is considered of high value
because chemical synthesis is laborious, and because incorporation of carboxylic acids in
human liver microsomes.

drug molecules might be beneficial for water solubility, might decrease the risk of hERG
inhibition [35], and might restrict passage of the blood-brain-barrier.
Combining the product profiles of the different P450 BM3 mutants (Table S1) with structural
modifications observed in the products (Figure 3), it can be concluded that oxygenation
of the benzylic methyl group is by far the major pathway for all mutants studied. The sum
of TAK-P5 and TAK-P7, both only involving modification of the benzylic methyl group,
represents from 50 to 100% of the products, dependent on the mutant. Because TAK-P1,
TAK-P2 and TAK-P3 might be secondary oxidation products of TAK-P5 and TAK-P7,
the total contribution of this pathway might even be underestimated. The most selective
mutants, which almost specifically produce the primary product TAK-P5, showed only very
low enzyme activity, explaining why no secondary product was observed. Only for mutants
M11 V87I and M11 V87F, the change in the product profile might be attributed to changes in
substrate orientation, because this is the only mutant which did not show any oxygenation
of the ethyl side chain: the benzylic methyl oxygenation appeared to represent 90% of the
biotransformation. Mutant M11 L437N showed the highest amount of product TAK-P6, and
relatively low levels of TAK-P5 and TAK-P7, which suggests that mutation L437N causes a
shift from benzylic methyl hydroxylation to hydroxylation of the ethyl side chain.

106 107
curves, the IC50-values and 95% confidence intervals can still be estimated for TAK-715
and TAK-P1 by fitting the data points by the same fitting procedure as was used for the
other compounds with complete binding curves. The IC50 obtained for TAK-715, 71 nM is
consistent with the value observed previously with this platform [19].
As shown in Figure 4, for three of the products, TAK-P1, TAK-P5, and TAK-P7, the binding
curves obtained were shifted to higher concentrations, compared to TAK-715. The largest
shift was observed with TAK-P7. This product showed an IC50-value of 1500 ± 203 nM
which is a 20-fold lower affinity than TAK-715. With IC50-values of 460 ± 165 nM and
310 ± 33 nM, respectively, TAK-P1 and TAK-P5 showed 5- and 7-fold decreased affinity.
Interestingly, TAK-P6 showed high affinity (IC50-value of 38 ± 4 nM). However, due to the
large standard error of the IC50-value of TAK-715, the difference in affinity of TAK-P6 is
not statistically significant. For TAK-P2, no binding curve was obtained, indicating that this
product completely lacked affinity to p38α.
These measurements indicate that oxygenation of the benzylic methyl group appears to
be detrimental for affinity to p38α. The fact that TAK-P7 shows a very low affinity, whereas
Figure 4: Concentration-dependent binding of TAK-715 and purified products TAK-P1, TAK-P2 shows no affinity at all, indicate that the carboxylic acid strongly disturbs binding to
TAK-P5, TAK-P6 and TAK-P7 to p38α kinase as determined by HRS in flow-injection analysis. p38α. These results are consistent with these results of Miwatashi et al. which showed that
Percentage of binding was calculated by the concentration-dependent displacement of the modifying the benzylic methyl group of TAK-715 by other substituents, or by positioning
tracer SKF86002 from p38α kinase. Error bars represent standard deviation. the methyl group to the ortho- or para-position, in all cases led to a strong loss in affinity
towards p38α [23]. Apparently, the benzylic methyl group at the meta-position of TAK-715 is
essential for its affinity. This is further confirmed by the high affinity of TAK-P6 in which only
the ethyl side chain is hydroxylated. Again, this seems consistent with the study of Miwatashi
3.2 Estimation of product affinity (IC50)
The products TAK-P1, TAK-P2, TAK-P5, TAK-P6 and TAK-P7 were isolated by preparative et al. [23] which showed that modification of the thiazole-2-ethyl moiety of TAK-715 to other
alkyl substituents (methyl, propyl) or a large substituent such as 4-methylsulfonylphenyl
3.2
HPLC. After checking their purity by 1H-NMR and analytical HPLC, the concentration of each
stock solution was determined by using a standard curve of TAK-715, assuming that the only showed little effect on p38α affinity.
extinction coefficients of each product was equivalent to that of TAK-715. This inaccuracy
can be resolved by correcting for the extinction coefficient once product standards are Conclusion
available. Subsequently, different concentrations of TAK-715 and TAK-715 metabolites
were analysed using the p38α HRS system in flow-injection mode in order to assess the The results of the present study show that the combination of a catalytically diverse set of
affinity of each compound. Figure 4 shows the concentration dependence of binding of the P450 BM3 mutants, as a toolbox to diversify a drug, and a HRS-systems capable of rapid
individual products and TAK-715 to p38α, after correction for the dilutions intrinsic to the screening for affinity to p38α kinase is a promising platform to generate potential novel lead
HRS-system. Calculated IC50-values of TAK-715 and its products are presented in Table 1. compounds which might have improved physicochemical properties (by improved water
For both TAK-715 and TAK-P1, not all data points of the full binding curve were considered solubility) and desired pharmacological properties. As exemplified by TAK-715 as model
in the IC50 determination, but those, which had been compromised by interferences, were compound, the P450 BM3 mutants were able to produce almost all metabolites produced by
excluded. This is neither due to a shortcoming in the assay nor due to experimental error. human liver microsomes at significant levels. Furthermore, profiling the effect of structural
On the contrary, these are two excellent examples of the power of the HRS platform modifications introduced by P450 on p38α kinase affinity gives valuable information on
to resolve interferences. In case of the highly lipophilic TAK-715, peak tailing due to which structural elements are critical and which are non-critical for affinity. This is an
nonspecific binding to materials of the HRS system deformed the Gaussian shape so essential tool in the construction of pharmacophore models and the lead optimization
severely at concentrations higher than 60 nM (5µM injected) that it was not possible to process. In addition, it would reduce the necessity of synthesizing large lead libraries by
calculate the local concentration in the assay. Also, the baseline for the following injection the medicinal chemist.
could not be recovered in a reasonable time. In the case of TAK-P1, (auto)fluorescence Finally, our results confirm that biocatalysis with CYPs can provide compounds with improved
at the assay wavelengths appeared to interfere with the p38α affinity measurement at pharmacological and/or physicochemical properties compared to the lead itself which are
high concentrations. Due to the different concentration-dependent behaviour of both not easily accessible by classic organic synthesis. This approach can be considered as an
contributions, e.g., increase of TAK-P1 (auto)fluorescence and decrease of fluorescence extra tool, next to the chemical synthesis in the typical medicinal chemistry approach.
enhancement of the tracer by displacement at increasing TAK-P1 concentration, a
W-shaped peak was obtained whose area no longer reflects the fraction of binding. It is Acknowledgement
important to note that these phenomena would also affect plate reader based assays,
albeit with different manifestations. Adsorption to the well plate surface and a mean signal This research was performed within the framework of project D2-102 “Metabolic stability
for positive and negative fluorescence contributions would result from high lipophilicity assessment as new tool in the Hit-to-Lead selection process and the generation of new
and auto-fluorescence, respectively. The first diminishes the actual concentration of the lead compound libraries” of the Dutch Top Institute Pharma.
binder while the latter results in an underestimation of the negative signal height. Both
effects will alter the dose-response curves and are difficult to detect in a plate reader setup.
However, in a HRS setup, these interferences are easily detected and the analysis can
thus be restricted to the uncompromised concentrations. Despite the incomplete binding
108 109
References 22. S. Miwatashi, Y. Arikawa, K. Naruo, K. Igaki, Y. Watanabe, H. Kimura, T. Kawamoto and S.
Ohkawa, Synthesis and biological activities of 4-phenyl-5-pyridyl-1,3-thiazole derivatives as p38
1. M. Sawayama, M. M. Chen, P. Kulanthaivel, M. S. Kuo, H. Hemmerle and F. H. Arnold, A panel
of cytochrome P450 BM3 variants to produce drug metabolites and diversify lead compounds, MAP kinase inhibitors, Chem. Pharm. Bull. (Tokyo), 2005, 53, 410-418.
Chemistry, 2009, 15, 11723-11729. 23. S. Miwatashi, Y. Arikawa, E. Kotani, M. Miyamoto, K. Naruo, H. Kimura, T. Tanaka, S. Asahi and
2. A. Fura, Y. Z. Shu, M. Zhu, R. L. Hanson, V. Roongta and W. G. Humphreys, Discovering drugs S. Ohkawa, Novel inhibitor of p38 MAP kinase as an anti-TNF-alpha drug: discovery of N-[4-[2-
through biological transformation: role of pharmacologically active metabolites in drug discovery, ethyl-4-(3-methylphenyl)-1,3-thiazol-5-yl]-2-pyridyl]benzamide (TAK-715) as a potent and orally
J. Med. Chem., 2004, 47, 4339-4351. active anti-rheumatoid arthritis agent, J. Med. Chem., 2005, 48, 5966-5979.
3. L. C. Wienkers and T. G. Heath, Predicting in vivo drug interactions from in vitro drug discovery 24. C. Ozdemir and C. A. Akdis, Discontinued drugs in 2006: pulmonary-allergy, dermatological,
data, Nature Rev. Drug Disc., 2005, 4, 825-833. gastrointestinal and arthritis drugs, Expert Opin. Investig. Drugs, 2007, 16, 1327-1344.
4. D. A. Smith and R. S. Obach, Metabolites in safety testing (MIST): considerations of mechanisms 25. F. Verkaar, A. A. van der Doelen, J. F. Smits, W. M. Blankesteijn and G. J. Zaman, Inhibition
of toxicity with dose, abundance, and duration of treatment, Chem. Res. Toxicol. 2009, 22, 267- of Wnt/β-catenin signaling by p38 MAP kinase inhibitors is explained by cross-reactivity with
279. casein kinase Iδ/ε., Chem. Biol., 2011, 18, 485-494.
5. C. M. Clouthier and J. N. Pelletier, Expanding the organic toolbox: a guide to integrating 26. N. Barker and H. Clevers, Mining the Wnt pathway for cancer therapeutics, Nature Rev. Drug
biocatalysis in synthesis, Chem. Soc. Rev. 2012, 41, 1585-1605. Discov., 2006, 5, 997-1014.
6. F. Hollmann, I. W. C. E. Arends, K. Buehler, A. Schallmey and B. Buhler, Enzyme-mediated 27. E. Vottero, V. Rea, J. Lastdrager, M. Honing, N. P. E. Vermeulen, J. N. M. Commandeur, Role of
oxidations for the chemist, Green Chemistry, 2011, 13, 226-265. residue 87 in substrate selectivity and regioselectivity of drug-metabolizing cytochrome P450
7. F. P. Guengerich, Cytochrome P450 enzymes in the generation of commercial products, Nature CYP102A1 M11, J. Biol. Inorg. Chem., 2011, 16, 899-912.
Rev. Drug Discov., 2002, 1, 359-366. 28. E. Stjernschantz, B. M. van Vugt-Lussenburg, A. Bonifacio, S. B. de Beer, G. van der Zwan, C.
8. S. Kumar, Engineering cytochrome P450 biocatalysts for biotechnology, medicine and Gooijer, J. N. M. Commandeur, N. P. E. Vermeulen and C. Oostenbrink, Structural rationalization
bioremediation, Expert Opin. Drug Metab. Toxicol., 2010, 6, 115-131. of novel drug metabolizing mutants of cytochrome P450 BM3., Proteins, 2008, 71, 336-352.
9. A. W. Munro, D. G. Leys, K. J. McLean, K. R. Marshall, T. W. Ost, S. Daff, C. S. Miles, S. K. 29. M. Dietrich, T. A. Do, R. D. Schmid, J. Pleiss and V. B. Urlacher, Altering the regioselectivity
Chapman, D. A. Lysek, C. C. Moser, C. C. Page and P. L. Dutton, P450 BM3: the very model of of the subterminal fatty acid hydroxylase P450 BM-3 towards gamma- and delta-positions, J.
a modern flavocytochrome, Trends Biochem Sci, 2002, 27, 250-257. Biotechnol., 2009, 139, 115-117.
10. C. J. Whitehouse and S. G. Bell, L. L. Wong, P450(BM3) (CYP102A1): connecting the dots, 30. H. Venkataraman, S. B. A. de Beer, L. A. van Bergen, N. van Essen, D. P. Geerke, N. P. E. Vermeulen,
3.2 Chem Soc Rev., 2012, 41, 1218-60.
11. V. Rea, A. J. Kolkman, E. Vottero, E. J. Stronks, K. A. Ampt, M. Honing, N. P. E. Vermeulen, S. S.
J. N. M. Commandeur, A single active site mutation inverts stereoselectivity of 16-hydroxylation
of testosterone catalyzed by engineered cytochrome P450 BM3, Chembiochem., 2012, 13, 520-
3.2
Wijmenga and J. N. M. Commandeur, Active site substitution A82W improves the regioselectivity 523.
of steroid hydroxylation by cytochrome P450 BM3 mutants as rationalized by spin relaxation 31. V. Rea, S. Dragovic, J. S. Boerma, F. de Kanter, N. P. E. Vermeulen and J. N. M. Commandeur, Role
nuclear magnetic resonance studies, Biochemistry, 2012, 51, 750-760. of residue 87 in the activity and regioselectivity of clozapine metabolism by drug-metabolizing
12. H. Venkataraman, S. B. A. de Beer, D. P. Geerke, N. P. E. Vermeulen and J. N. M. Commandeur, CYP102A1 M11H: application for structural characterization of clozapine GSH conjugates, Drug
Regio- and stereoselective hydroxylation of optically active α-ionone enantiomers by engineered Metab. Dispos., 2011, 39, 2411-20.
cytochrome P450 BM3 mutants, Adv. Synth. Catal., 2012, 354, 2172-2184. 32. D. Falck, J. Kool, M. Honing and W.M.A. Niessen, Tandem mass spectrometry study of p38α
13. B. M. A. van Vugt-Lussenburg, E. Stjernschantz, J. Lastdrager, C. Oostenbrink, N. P. E. Vermeulen kinase inhibitors and related substances, J. Mass Spectrom., 2013, 48, 718-731. [Chapter 4.1]
and J. N. M. Commandeur, Identification of critical residues in novel drug metabolizing mutants 33. K. H. Ling, G. A. Leeson, S. D. Burmaster, R. H. Hook, M. K. Reith and L. K. Cheng, Metabolism
of cytochrome P450 BM3 using random mutagenesis, J. Med. Chem., 2007, 50, 455-461. of terfenadine associated with CYP3A(4) activity in human hepatic microsomes, Drug. Metab.
14. C.H. Yun, K.H. Kim, D.H. Kim, H.C. Jung and J.G. Pan, The bacterial P450 BM3: a prototype for Dispos., 1995, 23, 631-636.
a biocatalyst with human P450 activities, Trends Biotechnol., 2007, 25, 289-98. 34. K. H. Liu, M. G. Kim, D. J. Lee, Y. J. Yoon, M. J. Kim, J. H. Shon, C. S. Choi, Y. K. Choi and Z.
15. J. Reinen, J. S. van Leeuwen, Y. Li, L. Sun, P. D. Grootenhuis, C. J. Decker, J. Saunders, N. P. E. Desta, J. G. Shin, Characterization of ebastine, hydroxyebastine, and carebastine metabolism
Vermeulen and J. N. M. Commandeur, Efficient screening of cytochrome P450 BM3 mutants for by human liver microsomes and expressed cytochrome P450 enzymes: major roles for CYP2J2
their metabolic activity and diversity toward a wide set of drug-like molecules in chemical space, and CYP3A, Drug Metab Dispos. 2006, 34, 1793-1797.
Drug Metab. Dispos., 2011, 39, 1568-1576. 35. B. Y. Zhu, Z. J. Jia, P. Zhang, T. Su, W. Huang, E. Goldman, D. Tumas, V. Kadambi, P. Eddy,
16. J. Kool, M. Giera, H. Irth and W. M. A. Niessen, Advances in mass spectrometry based post- U. Sinha, R. M. Scarborough and Y. Song, Inhibitory effect of carboxylic acid group on hERG
column bioaffinity profiling of mixtures, Anal. Bioanal. Chem., 2011, 399, 2655–2668. binding, Bioorg. Med. Chem. Lett., 2006, 16, 5507-5512.
17. J. S. B. de Vlieger, A. J. Kolkman, K. A. Ampt, J. N. M. Commandeur, N. P. E. Vermeulen, J.
Kool, S. S. Wijmenga, W. M. A. Niessen, H. Irth and M. Honing, Determination and identification
of estrogenic compounds generated with biosynthetic enzymes using hyphenated screening
assays, high resolution mass spectrometry and off-line NMR, J. Chromatogr. B, 2010, 878, 667-
674.
18. J. Reinen, L. L. Kalma, S. Begheijn, F. A. H. Heus, J. N. M. Commandeur and N. P. E. Vermeulen,
Application of cytochrome P450BM3 mutants as biocatalysts for the profiling of estrogen
receptor binding metabolites of the mycotoxin zearalenone, Xenobiotica, 2011, 41, 59-70.
19. D. Falck, J. S. B. de Vlieger, W. M. A. Niessen, J. Kool, M. Honing, M. Giera and H. Irth,
Development of an online p38α mitogen-activated protein kinase binding assay and integration
of LC–HR-MS, Anal. Bioanal. Chem., 2010, 398, 1771-1780. [Chapter 2.2]
20. P. Cohen, Protein kinases – the major drug targets of the twenty-first century?, Nature Rev. Drug
Discov., 2002, 1, 309-315.
21. A. Dorn, V. Schattel and S. Laufer, Design, synthesis and SAR of phenylamino-substituted
5,11-dihydro-dibenzo[a,d]cyclohepten-10-ones and 11H-dibenzo[b,f]oxepin-10-ones as p38
MAP kinase inhibitors, Bioorg. Med. Chem. Lett., 2011, 20, 3074-3077.

110 111
Supplementary Material Table S1: Percentage of conversion and ratio between the biotransformation products of TAK-715
incubations with human liver microsomes (HLM) and 33 mutants of P450 BM3α.

Enzyme % Conv TAK-P1 TAK-P2 TAK-P3 TAK-P4 TAK-P5 TAK-P6 TAK-P7 TAK-P8
m/z of [M+H]+ )b )a 432.138 446.121 430.123 432.138 416.146 416.146 430.123 416.146
HLM 33 19 14 5 3 17 10 28 4
M11 38 11 6 0 0 22 3 42 13
M11 L437E 36 35 9 0 0 23 15 16 2
M11 A74E 36 35 8 0 0 18 13 24 2
M11 A74D 33 34 7 0 0 20 14 24 3
M11 L437S 31 20 7 0 0 25 15 27 6
M11 L437T 30 33 5 0 0 23 15 20 3
M11 V87I 25 0 0 0 0 32 0 60 8
M01 L437E 20 18 0 0 0 42 26 9 5
M11 V87F L437S 20 21 0 0 0 44 20 9 6
M11 A82W 19 3 2 0 0 23 4 57 11
M05 19 6 0 0 0 34 6 40 14
M01 S72D 18 11 2 0 0 38 8 28 13
M11 V87F L437N 18 23 0 0 0 31 23 13 10
M01 A82W 18 5 0 0 0 38 5 48 4
3.2 M01 S72E
M01 A74D
17
17
14
13
0
0
0
0
0
0
48
48
26
26
7
7
5
6
3.2
M11 V87I L437N 16 4 0 0 0 47 6 28 15
M11 A82W V87F 15 0 0 0 0 78 0 0 14
Figure S1: Preparative HPLC-UV (254 nm) chromatogram of mixture of biotransformation products M11 V87F 15 0 0 0 0 82 5 6 7
obtained by large-scale incubation of TAK-715 with P450 BM3 mutant M11. M11 L437N 15 9 0 0 0 29 38 18 6
M11 S72D 12 12 6 0 0 52 17 6 7
M11 V87A 10 10 0 0 0 53 8 25 4
M11 V87I L437S 9 0 0 0 0 70 1 6 23
M01 7 0 0 0 0 60 14 6 20
M11 V87Y 7 0 0 0 0 76 6 6 12
M02 7 0 0 0 0 75 10 6 9
M11 V87L 6 0 0 0 0 79 5 3 13
M11 A82Y L437S 4 0 0 0 0 100 0 0 0
M11 A82Y V87I 4 0 0 0 0 85 15 0 0
M11 A82Y V87F 4 0 0 0 0 100 0 0 0
M11 V87Q 3 0 0 0 0 85 15 0 0
M01 A82C 3 0 0 0 0 100 0 0 0
M11 A82W L437S 2 0 0 0 0 100 0 0 0
a. All values represent averages of three individual experiments; RSDs were always less than 10%
b. [M+H]+-values as determined by accurate-mass measurements by HR-MS.

112 113
Table S2: 1H-NMR-spectra of TAK-715 and major biotransformation products formed by P450 BM3
M11.

TAK-715 TAK-P 1 TAK-P 2 TAK-P 5 TAK-P 6 TAK-P 7


Ha 1.45-1.48 1.61-1.63 1.64-1.65 1.45-1.48 1.62-1.64 1.44-1.49
(3H,t) (3H,d) (3H,d) (3H,t) (3H,d) (3H,t)
Hb 3.09-3.13 5.08-5.12 5.08-5.12 3.09-3.14 5.08-5.12 3.08-3.14
(2H,q) (1H,q) (1H,q) (2H,q) (1H,q) (2H,q)

3.2 Hc
Hd
2.33 (3H s)
7.19-7.26
~ 4.9* (s)
7.19-7.26
-
7.64-7.66
~ 4.9* (s)
7.35-7.39
2.32 (3H,s)
7.18-7.25
-
7.58-7.62
3.2
(3H,m) (3H,m) (1H,d) (3H,m) (3H,m) (1H,d)
He “ “ 7.43-7.46 “ “ 7.40-7.43
(1H,t) (1H,t)
Hf “ “ 7.99-8.01 “ “ 7.98-8.00
(1H,d) (1H,d)
Hg 7.34 (1H,s) 7.51-7.54 8.18 (1H,s) 7.51-7.54 7.34 (1H,s) 8.14 (1H,s)
(3H,m) (3H,m)
Hh 6.95-6.96 7.00-7.01 7.00 (1H,s) 6.98-6.99 6.97-6.98 6.95-6.96
(1H,d of d) (1H,d of d) (1H,d of d) (1H,d of d) (1H,d of d)
Hi 8.21-8.22 8.23-8.24 8.25-8.26 8.22-8.23 8.22-8.23 8.21-8.22
(1H,d) (1H,d) (1H,d) (1H,d) (1H,d) (1H,d)
Hj 8.31 (1H,d) 8.30 (1H,d) 8.33 (1H,d) 8.29 (1H,d) 8.31 (1H,d) 8.31 (1H,d)
Hk - - - - - -
Hl 7.94-7.96 7.94-7.95 7.94-7.95 7.94-7.95 7.94-7.96 7.94-7.95
(2H,d) (2H,d) (2H,d) (2H,d) (2H,d) (2H,d)
Hm 7.51-7.54 7.51-7.55 7.51-7.54 7.51-7.54 7.51-7.54 7.51-7.54
(2H,d of d) (3H,m) (2H,d of d) (3H,m) (2H,d of d) (2H,d of d)
Hn 7.59-7.62 7.59-7.62 7.58-7.60 7.59-7.62 7.59-7.62 7.58-7.62
(1H,t) (1H,t) (1H,t) (1H,t) (1H,t) (1H,t)
* Chemical shift and integral of benzylic protons could not be determined accurately due to interfer-
ence with solvent signal.

114 115
Chapter 3.3
Comparison of (bio-)
transformation methods
for the generation of
metabolite-like compound libraries
of p38α MAP kinase inhibitors
using high-resolution screening

David Falck1, Fatie Rahimi Pirkolachachi1, Martin Giera2, Maarten


Honing1,3, Jeroen Kool1 and Wilfried M. A. Niessen1,4

1
AIMMS Division of BioMolecular Analysis, VU University Amsterdam,
De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
2
Center for Proteomics and Metabolomics, Leiden University Medical Center,
Albinusdreef 2, 2300 RC Leiden, The Netherlands
3
DSM Resolve, Urmonderbaan 22, 6160 MD Geleen, The Netherlands
4
hyphen MassSpec, de Wetstraat 8, 2332 XT Leiden, The Netherlands
Abstract from different known p38α inhibitors. Chemical oxidation with hydrogen peroxide (H2O2),
which is generally used to simulate the influence of molecular oxygen on drugs during long
Four hydrophobic p38α mitogen-activated protein kinase inhibitors were refluxed with term storage [13, 14], may yield similar products as metabolic reactions, though not by the
7.5% hydrogen peroxide at 80°C and irradiated with visible light in order to generate same mechanisms [15]. Irradiation of drugs with intense visible light (Light), also applied in
more hydrophilic conversion products. The resulting mixtures were analysed in a high- stability testing, was investigated as well [13]. This approach might not be as promising for
resolution screening (HRS) platform, featuring liquid chromatographic separation coupled the generation of metabolite-like compounds, but Light can initiate photochemical reactions,
in parallel with a fluorescence enhancement based continuous-flow affinity bioassay which possibly modify the scaffold of the molecule, resulting in new active core structures
towards the p38α mitogen-activated protein kinase and with high-resolution (tandem) mass [14, 16]. Electrochemical conversion (EC) has been shown to be able to reproduce certain
spectrometry on an ion-trap–time-of-flight hybrid instrument. The results were compared metabolic reactions, especially N- and O-dealkylation or P- and S-oxidation [17]. HRS data
with similar data where chemical diversity was achieved by means of electrochemical on EC generated libraries of CPs of p38α inhibitors have been reported earlier [18]. An
conversion or incubation with either human liver microsomes or cytochrome P450s from interesting biosynthetic approach is the use of genetically engineered bacterial variants
Bacillus megaterium (BM3s). In total, more than 50 conversion products were identified. of metabolic enzymes, e.g., cytochrome P450s from Bacillus megaterium, especially one
The metabolite-like compound libraries studied are discussed in terms of the reactions called BM3 [19]. These BM3s can be engineered to be highly regio- and stereo-selective
enabled, the retention of affinity, and the change in hydrophilicity by modification, in [20] as well as for specific product profiles [21]. The library generation of CPs for the p38α
summary the ability to generate bioactive, more hydrophilic potential lead compounds. In inhibitor TAK-715 by means of BM3 mutants has been reported elsewhere [21]. In vitro
this context, HRS is demonstrated to be an effective tool as it reduces the effort directed metabolism simulation by human liver microsomal incubations (HLM) was investigated and
towards laborious synthesis and purification schemes. compared with the other methods in order to additionally explore the usefulness of HRS in
selecting suitable methods for metabolite synthesis in safety testing [22].
This manuscript is part of and actually concludes a larger study. Here, we present the
1. Introduction structure and affinity profiles of the lead libraries of CPs of the p38α inhibitors produced
with H2O2 and Light. In addition, these new results are critically compared to previously
Despite the efforts in, for example, combinatorial chemistry, drug discovery still relies mainly
reported data, obtained with the other modification methods, with respect to the reactions
on the utilization of purified compounds for biological affinity or activity testing, due to the
enabled, the retention of affinity, and the change in hydrophilicity by modification, based on
limitations of classical high-throughput assays [1]. However, almost every synthetic or
the results of our HRS platform. This enabled us to establish an initial qualitative Structure-
biosynthetic endeavour initially produces a mixture of compounds mainly because of side
Activity Relationship (SAR), which in turn allows us to assess the usefulness of employing
reactions. As a consequence, significant effort is directed towards optimization of synthesis
these methods as toolbox in the generation of metabolite-like lead libraries and judge the
[2] and purification [3] schemes for discovery compounds. An approach to overcome this
3.3 bottleneck is the use of high-resolution screening (HRS), which is based on the bioaffinity
assessment of individual compounds in a mixture [4] instead of requiring pure compounds
potential of their products as lead molecules. Thereby, we show that the implementation
of the HRS platform together with a variety of modification methods is likely to create an 3.3
effective lead optimization process as it reduces the effort directed towards laborious
or yielding a summed response like in high-throughput assays. By assessing all reaction
synthesis, purification and testing schemes.
products of a (parallel) synthesis, optimization and purification efforts can be directed
towards active products without having to make a pre-selection.
HRS relies on a combination of separation, mostly liquid chromatography (LC), and 2. Materials and Methods
hyphenated bioassays. The technology has matured in recent years by gradual improvements
in stability and reproducibility as well as by the integration of mass spectrometry (MS) for 2.1 Materials
structure elucidation [4]. We recently developed an HRS platform for the contemporary The human recombinant p38α mitogen-activated protein kinase as well as its inhibitors
drug target p38α mitogen-activated protein kinase (p38α) which was thoroughly validated, DMPIP (1-{6-chloro-5-[(2R,5S)-4-(4-fluorobenzyl)-2,5-dimethylpiperazine-1-carbonyl]-
for example by comparison of obtained IC50 values with other formats [5]. It enabled us to 3aHindol-3-yl}-2-morpholinoethane-1,2-dione), SB203580 (4-[4-(4-fluorophenyl)-2-(4-
simultaneously assess the structure and affinity towards p38α of individual small molecules methylsulfinylphenyl)-1H-imidazol-5-yl]pyridine) [23], BIRB796 (N-[3-(tert-butyl)-1-(4-
in a mixture. p38α is a key node in the cellular response to inflammatory stimuli and has methylphenyl)-1H-pyrazol-5-yl]-N’-[4-[2-(4-morpholinyl)ethoxy]-1-naphthalenyl]-urea)
thus been proposed as a drug target for therapy of chronic inflammatory diseases like [24], and TAK-715 (N-[4-[2-Ethyl-4-(3-methylphenyl)-5-thiazolyl]-2-pyridinyl]benzamide)
psoriasis, rheumatoid arthritis or Crohn’s disease [6]. Many high-affinity p38α inhibitors [25] come from various sources. Human liver microsomes (HLM) pooled from different
have been created and some of them have advanced as far as clinical phase III in the individual donors were obtained from BD Gentest TM (San Jose, CA, USA; Cat. No.
drug development pipeline [7, 8]. Current p38α inhibitors are often very lipophilic, which 452161) and contained 20 mg/mL protein. Methanol (LC-MS grade) and a 30% hydrogen
negatively influences their solubility and bioavailability [9]. peroxide solution were purchased from Biosolve (Valkenswaard, the Netherlands) and J.T.
We investigated modifications of know p38α inhibitors by various means aiming to improve Baker (Deventer, Holland), respectively. Formic acid was obtained from Merck (Darmstadt,
their physicochemical properties, which are pivotal aspects in steering bio-availability, Germany). All other chemicals were obtained from Sigma-Aldrich (Steinheim, Germany)
while retaining high affinities. Oxidative metabolism can decrease the lipophilicity of the at the highest purity available. Water was generated with a Milli-Q purification system
inhibitors and may retain their target affinity [10]. Therefore, techniques used for producing (Millipore, Amsterdam, Netherlands).
metabolites or metabolite-like compounds may present a promising route to lead libraries
with improved physicochemical properties. Next to the increase in hydrophilicity, for example 2.2 Chemical oxidation
by hydroxylation, these bioactive metabolites may have more selectivity as well [11, 12]. The p38α inhibitors were oxidized by refluxing (ca. 80°C) with hydrogen peroxide. Incubation
Therefore, we studied several methods that have been used to produce more hydrophilic times were 105 min for DMPIP, 180 min for SB203580, and 300 min for TAK-715 (refer to
metabolites, in order to generate metabolite-like lead libraries of conversion products (CPs) Section 1 of the electronic supplementary material (ESM)). Sampling was done by means
of a syringe from a three-neck flask via a septum. The reaction solutions were diluted
118 119
from 1  mM stock solutions in methanol to a solvent composition of 75% aq. hydrogen
peroxide and 25% MeOH. They contained an optimized hydrogen peroxide concentration
of 7.5% (w/w) and an inhibitor concentration of 100 µM. Control incubations under reflux
conditions containing no hydrogen peroxide were included to assess the influence of
thermal degradation.

2.3 Photochemical modification


Photochemical modification (Light) was induced by irradiating aliquots of an inhibitor with
intense light of the visible range (>310  nm [16]) at room temperature. The exclusion of
UV-wavelengths is expected to result in more specific reactions and prevent advanced
decomposition. The light was generated by a 150W Xenon lamp model L21 equipped with
an AEG transformer. The reaction solutions, diluted from 1 mM stock solutions in methanol
to a solvent composition of 75% water and 25% MeOH, were placed at a distance of 15 cm
from the lamp. The concentrations of the inhibitors were 100 µM for DMPIP and SB203580
and 10 µM for TAK-715 (see ESM Section 1), respectively. Incubation times were 120 min
for DMPIP, 300 min for SB203580, and 30 min for TAK-715 in Light (refer to ESM Section 1).
The use of closed Duran glass vessels excluded light of the UV range (<310 nm [16]) and
hindered evaporation. Samples were taken with a syringe. To protect the environment from
UV radiation, the whole setup was shielded with aluminium foil. Control incubations, which
assess the contribution of heat generated by the Xenon lamp, were prepared by wrapping
the samples in aluminium foil to exclude irradiation.

2.4 Microsomal incubations


In order to investigate how closely the products of the modification methods resemble
human metabolites, human phase I metabolism was simulated in vitro by incubation with
human liver microsomes (HLM). Phase I oxidative metabolites were generated by HLM
3.3 and cofactor NADPH using a modified version of a protocol described elsewhere [26]. In
brief, the reaction mixtures were prepared in 50 mM potassium phosphate buffer (pH 7.4) 3.3
including 5  mM magnesium chloride. The incubation mixtures contained 100  mM p38α
inhibitor, 2 mg/mL human liver microsomes and 6 mM NADPH, and were incubated for 2 h
at 37°C. Constant availability of NADPH was ensured by a regenerating system of 5 mM
glucose-6-phosphate and 5  U/mL glucose-6-phosphate dehydrogenase and by adding
10% (v/v) of a 10 mM NADPH solution in the above mentioned phosphate buffer after 30,
60 and 90 min. The reactions were stopped by addition of ice-cold acetonitrile 2:1 (v:v). The
samples were subsequently centrifuged at 16,000 × g for 5 min at 4°C. The supernatants
were taken, freeze-dried and stored at –20°C. For the HRS analysis, the samples were
re-dissolved in a 30% aqueous methanol solution, providing 10-fold higher concentrations.

2.5 HRS analysis


Figure 1: HRS chromatograms of A) TAK-715 in H2O2 and B) DMPIP in Light. The top trace is the
Analysis of structure and affinity towards p38α was conducted with an HRS platform fluorescence chromatogram (bioaffinity trace); affinity towards p38α is observed as a negative peak.
developed earlier [5]. The HRS platform consisted of two LC-20 AD pumps, two LC-10 The bottom traces are the extracted ion chromatograms (EIC) for the different CPs. The colors of the
AD pumps, a SIL-20 AC autosampler, a CTO-20 AC and a CTO-10 AC column oven, an EICs match their corresponding tags (CP numbers). For the m/z values of the EICs, refer to the CPs
RF-10AXL fluorescence detector, an SPD-AD UV/VIS detector, a CBM-20A controller, and in A) Table 2 and B) Table 1. Top and bottom chromatograms are aligned to correct for void volume
an ion-trap–time-of-flight mass spectrometer equipped with an ESI source, all products of differences. In this way, structure and affinity data can be correlated [18].
Shimadzu (’s Hertogenbosch, the Netherlands). In short, 10 µL (100 µL for TAK-715 in Light)
of the mixture of CPs were separated on a Symmetry C18 column (2.1 x 100 mm, 3.5 μm 3. Results
particles; Waters, Milford, MA, USA) at 40°C and a flow rate of 113 µL/min. LC solvents
included water, methanol and formic acid in the ratios 99% / 1% / 0.01% for solvent A and This paper integrates the new data on CPs generated by Light and H2O2 with data
1% / 99% / 0.01% for solvent B. Analytes were eluted with several gradients optimized per generated by other conversion methods. In this sense, it is part of and summarizes and
compound and per modification method (see ESM Section 2). The eluent was split, one concludes a larger study [5, 18, 21, 27]. Additionally, it provides a critical comparison of
part being analysed by electrospray ionization high-resolution (tandem) mass spectrometry the different methods and summarizes the role of HRS in the study. The conversion of
(HR-MSn) on the Shimadzu ion-trap–time-of-flight mass spectrometer, another constantly four p38α inhibitors (see Tables 1 to 4 for their structures) with five different modification
mixed with the bioassay reagents and detected by the fluorescence detector. Details on the strategies (EC, Light, H2O2, BM3, and HLM) results in a very large and complex data set.
HR-MSn conditions can be found elsewhere [27]. Therefore, interpretation of the HR-MSn spectra of the four parent drugs and the structure
120 121
elucidation of the more than 50 CPs found (see Tables 1 to 4) was reported separately [27]. the compound identity and the p38α affinity as well as with hydrophilicity information [28]
Here, we first report the results on the compound libraries of CPs generated using H2O2, as judged from relative retention time (RRT, [29]) (see Tables 1 to 4). As discussed before,
Light, and HLM. Proposed structures of the CPs are given in Section 3 to 6 of the ESM. this presents a strong basis for assembling a thorough SAR. This is nicely illustrated in the
Next, we review and compare the results from the five compound modification strategies typical HRS chromatograms shown in Figure 1. The p38α affinity is observed as a negative
in terms of the reactions enabled, the retention of affinity, and the change in hydrophilicity peak in the fluorescence chromatogram (top traces in Figure  1). The affinity peaks are
by modification, and thus the ability to generate valuable lead compounds, with potentially numbered according to the figure number, Roman numbers and a superscript BIO, e.g.,
improved efficacy, selectivity, and importantly bioavailability. 1A-IBIO for the first affinity peak in Figure 1A. The CPs are indicated with the m/z of their
protonated molecule, e.g., CP296 corresponds to a CP with m/z 296. Different extracted-
The HRS platform assesses in parallel the bioaffinity of the CPs by an on-line post-column ion chromatograms (EIC) are shown for the various CPs. Additionally, UV detection at
affinity assay and their structure by HR-MSn. This provides a direct correlation between 210 nm in line with the MS detection allows a (semi-)quantitative statement on abundance
of CPs. Minor and major CPs/metabolites are defined as having less or more than 20%
Table 1: Summary of the conversion products of DMPIP generated with the five applied modifi- peak area, respectively, as compared to the largest peak at UV 210 nm [30].
cation methods. The profile groups (A to D) used in structure elucidation are circled in grey [24].
Isomeric modifications were attributed to the individual profile groups. For structure proposals for
these CPs, see ESM Section 3. 3.1 Oxidation with hydrogen peroxide
First, the hydrogen peroxide concentration was optimized by testing 3.0%, 7.5%, and 15%
(w/w) at a reflux temperature of ca. 80°C to increase reaction speed [31]. With 3.0%, little
conversion was achieved, whereas 15% resulted in undesired advanced decomposition
of the molecule. As the controls showed that temperature increase alone did not result
in significant side reactions, refluxing with 7.5% hydrogen peroxide was applied. The
conversion efficiency, measured as the reduction in the parent compound peak area by
LC–UV, is ca. 50% after 180 min for SB203580, 30% after 300 min for TAK-715 and 90%
after 105 min for DMPIP (see ESM Section 1). BIRB796 was not included in this part of
the study, because initial experiments indicated that urea hydrolysis, due to the prevailing
Compound p38 RRT m/z Delta Modification Method acidic conditions, is the main reaction pathway.
affinitya
3.1.1 DMPIP
3.3 3.3
Parent X 1.00 541.204 x x x
The conversion of DMPIP ([M+H]+ with m/z 541; C28H31ClFN4O4+) by hydrogen peroxide
CP431 O 0.57 431.148 -C7H5F Dealkylation of A and de- EC (see Table  1) produces two abundant products, both with increased hydrophilicity. The
-H2 hydrogenation in B
major product CP557A (C28H31ClFN4O5+), containing a hydroxylation in the B part (cf.
CP433 O 0.34 433.165 -C7H5F Dealkylation of A BM3 Table 1), is the only one showing affinity, although significantly lower than DMPIP. Whereas
CP472 X 0.86 472.146 -C4H7N Amide hydrolysis EC MS and UV responses are 2 to 10 times higher, respectively, the affinity response is at
CP501 O 0.74, 0.67 501.170 -C3H4 Double dealkylation in B EC, H2O2 least 30 times lower. For the minor product CP501 (C25H27ClFN4O4+), a double dealkylation
CP505 ? 0.88 505.226 -HCl New 5-ring B and C Light product of the piperazine ring, no affinity signal is observed.
CP523A,C O 0.63 (A), 523.235 +H2O Light
0.69 (C), -HCl 3.1.2 TAK-715
CP523B, D O 0.67 (B), 523.235 “ Exchange of Cl for H in C Light In treatment of TAK-715 ([M+H]+ with m/z 400; C24H22N3OS+) with hydrogen peroxide, a large
0.72 (D) and OH in B number of minor products is yielded which almost exclusively have increased hydrophilicity
CP525 O 0.80 525.170 -CH4 Methyl loss from B EC (see Table  2 and Figure  1A). The strongest UV and MS signal comes from the amide
CP529 O 0.96 529.165 -C2H4+O All modifications in B EC hydrolysis product CP296 (C17H18N3S+) which is probably responsible for the first and highest
CP537 ? 0.88 537.252 -HCl Light of the product affinity peaks (1A-IBIO) and is also among the most hydrophilic products
+CH4O (RRT 0.35). At least 9 different mono-oxygenated CPs (CP416D to L, C24H22N3O2S+) can
CP539 X 1.22, 1.39 539.187 -H2 Dehydrogenation in B EC, Light be distinguished by reWtention time varying largely in hydrophilicity (RRTs range from 0.56
to 1.20). Additionally, two doubly-oxygenated compounds (CP432C and D, C24H22N3O3S+)
CP541A ? 0.88 541.204 x Light
are observed at RRT 0.57 and RRT 0.61. These and other CPs (Table 2) are discussed in
CP541B X 0.96 “ “ Light more detail in ESM Section 4. At RRTs between 0.5 and 0.8, overlapping affinity peaks are
CP541C X 1.34 “ “ Light observed (1A-IIBIO, 1A-IIIBIO and 1A-IVBIO), indicating that some of the mono-hydroxylated
CP557A X 0.75 557.197 +O OH in B H2O2 and possibly some of the double-oxygenated products have affinity for p38α. Due to the
CP557B X 0.95 “ “ Oxygenation in D EC high complexity of this sample (Table 2 and Figure 1A), separation is insufficient for exact
CP557C - 1.10 “ “ N-oxide in C HLM affinity assignment. Only CP432D and CP416G can clearly be attributed to 1A-IIIBIO and
CP559 X 0.80 559.213 +H2O Water addition to C or D Light 1A-IVBIO, respectively, as the onset of both affinity peaks cannot be attributed to any other
CP571 O 1.08 571.212 +CH2O Methoxylation in A or B EC CP. However, the width of 1A-IIIBIO and 1A-IVBIO suggests that there are other co-eluting CPs
CP573 ? 0.88 573.226 +CH4O Light with affinity. As this study is conducted under a screening paradigm, any further attempt
to achieve sufficient separation, thus allowing additional structure identification by MS and
a
X: affinity; O: no affinity observed; ?: unclear, mostly due to co-elution; -: not measured more precise affinity assignment, was considered to be out of its scope. Finally, 1A-VBIO
122 123
Table 2: Summary of the conversion products of TAK- in the bioaffinity trace indicated the presence of another CP, not initially recognized in the
715 generated with the four applicable modification MS data, corresponding to CP384 (C24H22N3O2+) in which sulphur has been exchanged for
methods. Individual carbon atoms, at which modifica- oxygen. CP384 only generates a very minor peak in the TIC, and as this sulphur-oxygen
tions take place, are numbered. For structure propos- exchange reaction was not expected, an EIC trace was initially not generated either. This is
als for these CPs, see ESM Section 4.
a good example of the power of the on-line post-column affinity assay [4]: a low-abundance
compound with relatively high affinity, missed in UV or MS detection, will be detected by
the affinity detection. Thus, the HRS platform is in a positive way biased towards binders.
a
X: affinity; O: no affinity observed;
?: unclear, mostly due to co-elution; 3.1.3 SB203580
The major CP of SB203580 ([M+H]+ with m/z 378; C21H17FN3OS+) is the S-oxidation product
CP394C (C21H17FN3O2S+). No affinity peaks were observed because these are masked
Compound p38 affinitya RRT m/z Delta Modification Method
Table 3: Summary of the conversion products of SB 203580 generated with the four applicable
Parent X 1.00 400.149 x x x modification methods. Individual carbon atoms, at which modifications take place, are numbered.
CP279 ? 1.14 279.096 -C7H7NO Benzamide loss and Light For structure proposals for these CPs, see ESM Section 5.
dehydrogenation
CP294A ? 0.37 294.107 -C7H4O Amide hydrolysis and H2O2
-H2 dehydrogenation
CP294B O 0.48 “ “ “ H2O2
CP296 O (Light 0.75, 296.123 -C7H4O Amide hydrolysis Light, HLM,
HLM), 0.35, +O H2O2
X (H2O2) 0.35
CP312 X 0.21 312.117 -C7H4O Amide hydrolysis and OH HLM
CP312B ? 0.33 “ “ H2O2
CP384 X 0.91 384.172 -S +O H2O2
CP398A ? 1.04 398.134 -H2 Light Compound RRT m/z Delta Modification Method
3.3 CP398B O 1.35 “ “ Ring formation between
carbon-9 and carbon-20
Light
Parent 1.00 378.108 x x x
3.3
CP398C ? 1.13 “ “ H2O2 CP243 1.26 243.094 -C7H5NS Sulfoxide replacement in CP305 Light
CP400 O 1.18 400.149 x Isomer of TAK-715 Light CP289 1.58 289.081 -C6H3N Sulfoxide to thioether in CP305 Light
CP414 ? 1.04 414.128 -H2 +O H2O2 CP305 1.16 305.077 -C6H3N +O See ESM Section 5 Light
CP416A X 0.80 416.144 +O OH at carbon-14 BM3, HLM CP316 1.18 316.125 -CH2OS Sulfoxide replacement H2O2
CP416B X 0.83 “ “ OH at carbon-2 BM3, HLM CP321 1.53 321.072 -C6H3N +2O Methyl hydroxylation in CP305 Light
CP416C O 0.87 “ “ OH? BM3, HLM CP362 1.44 362.113 -O Sulfoxide to thioether Light
CP416D ? 0.57 “ “ H2O2 CP376 0.60 376.113 -F+H+O -HF + H2O H2O2
CP416E+F ? 0.64 “ “ H2O2 CP392 0.68 392.108 -F+H+2O -HF + H2O + O H2O2
CP416G X 0.70 “ “ H2O2 CP394A 0.76 394.103 +O Aromatic OH H2O2
CP416H+I ? 0.74 “ “ H2O2 CP394B 0.80 “ “ Aromatic OH H2O2
CP416J O 0.87 “ “ H2O2 CP394C 1.05, 1.10, “ “ S-oxidation HLM, H2O2,
CP416K ? 1.11 “ “ H2O2 1.12 EC
CP416L ? 1.20 “ “ H2O2 CP394D 1.12, 1.24 “ “ N-oxidation HLM, H2O2
CP430A X 0.64 430.123 -H2 +2O OH at carbon-2 or-3 and HLM CP394E 1.36 “ “ OH in pyridine ortho to N H2O2
aldehyde at carbon-14 CP410A 0.83 410.098 +2O Aromatic OH (CP394A) and H2O2
CP430B X 0.84 “ “ Carboxylic acid at carbon-14 BM3, HLM S-oxidation
CP432A X 0.58 432.139 +2O OH at carbon-2 and BM3, HLM CP410B 0.88 “ “ Aromatic OH (CP394B) and H2O2
carbon-14 S-oxidation
CP432B X 0.70 “ “ Two OH in ethyl group (BM3), HLM CP410C 1.29 “ “ S- and N-oxidation H2O2
CP432C ? 0.57 “ “ H2O2 CP410D 1.43 “ “ S-oxidation and H2O2
CP432D X 0.61 “ “ H2O2 OH in pyridine ortho to N
CP446 O 0.62 446.121 -H2 +3O Carboxylic acid at carbon-14 BM3, HLM CP424A 1.37 424.115 +O +CH2O Both in fluorophenyl ring Light
and OH at carbon-2 or-3 CP424B 1.44 “ “ OH at carbon-14, Light
CP448 X 0.44 448.138 +3O HLM OCH3 in pyridine ring

124 125
by strong auto-fluorescence as indicated by positive fluorescence peaks throughout the 3.3.2 SB203580
bioaffinity trace. Next to CP394C, four other mono-oxygenation isomers and four different The in vitro metabolism of SB203580 ([M+H]+ with m/z 378; C21H17FN3OS+) has been
double-oxygenated CPs are observed (Table 3). The latter CPs result from a combination investigated by Henklova et al. [33]. However, in addition to the reported S-oxidation
of S-oxidation (CP394C) with each of the other mono-oxygenation reactions. Their metabolite (CP394C, C21H17FN3O2S+), we observed a minor metabolite showing N-oxidation
hydrophilicity nicely follows the hydrophilicity trend of their respective mono-oxygenation instead (CP394D, C21H17FN3O2S+) (Table 3). This additional finding is probably due to the
CPs. These and other CPs (Table 3) are discussed in more detail in ESM Section 5. higher sensitivity of HR-MSn, which significantly increases the signal-to-noise ratio when
analysing complex samples.
3.2 Photochemical modification
The conversion efficiency for the photochemical modification (Light) is ca. 20% after 3.3.3 BIRB796
300 min for SB203580, 90% after 300 min for TAK-715 and 70% after 120 min for DMPIP The transformation of BIRB796 ([M+H]+/[M+2H]2+ with m/z 528/265; C31H38N5O3+/
(determined by LC–UV; see ESM Section 1). BIRB796 was also not included in this part of C31H39N5O32+) resulted in a more complex mixture (Table  4). Two mono-oxygenation
the study, as initial experiments yielded only products of advanced decomposition. metabolites, CP544A and CP544B (C31H39N5O42+), were observed at RRTs of 0.84 and
0.87, respectively. These result from hydroxylation of the A part (CP544A) or of the A or B
3.2.1 DMPIP part (CP544B) (cf. Table 4). Other low-abundant mono-oxygenation products were present
DMPIP ([M+H]+ with m/z 541; C28H31ClFN4O4+) in Light yields a number of minor products at higher RRT. Furthermore, two co-eluting double-dealkylation metabolites were detected.
(Table  1). CP539 (C28H29ClFN4O4+), the product of a dehydrogenation in the B part (cf. CP502 (C29H37N5O32+) and CP458 (C27H33N5O22+) show the loss of ethyne and dihydrofuran,
Table  1), shows the highest affinity and the strongest UV response. As expected, this respectively, from part D. Additionally, three metabolites are partly co-eluting at RRTs of
modification increases the lipophilicity of the molecule (RRT 1.39). While the UV response 1.27 and 1.25. One of these less hydrophilic metabolites is the hydroquinone CP415 (RRT
of CP539 is only slightly more than 10% of the residual substrate, the height of the affinity 1.27, C25H27N4O2+) resulting from loss of the ethyl-morpholino group by O-dealkylation.
peak is almost 75% of that of DMPIP. Furthermore, three minor affinity signals are observed Additionally, CP1052 (C62H72N10O62+) is a dimerization product with two additional bonds
(Figure 1B). The shoulder (1B-IIIBIO) at the beginning of the substrate affinity peak (1B-SBIO) in the C and/or D part. CP459 (C27H31N4O3+) results from oxidative deamination of the
is induced by CP541B (C28H31ClFN4O4+), an isomer of DMPIP. Peak 1B-IIBIO might result from
up to four co-eluting compounds, being another substrate isomer CP541A (C28H31ClFN4O4+), Table 4: Summary of the conversion products of BIRB796 generated with the two applicable mod-
a dechlorination and ring formation product (CP505, C28H30FN4O4+), a product showing ification methods. The profile groups (A to D) used in structure elucidation are circled in grey [24].
the addition of methanol (CP573, C29H35ClFN4O5+), and CP537 (C29H34FN4O5+) combining Isomeric modifications were attributed to the individual profile groups. For structure proposals for
the modifications of CP505 and CP573. The structure elucidation of these four CPs was these CPs, see ESM Section 6.
3.3 hindered by their low abundance and limited fragmentation in MSn [27]. Peak 1B-IBIO seems
to correlate to a product of (at least apparent) water addition (CP559, C28H33ClFN4O5+). For
3.3
more information and additional CPs, see ESM Section 3.

3.2.2 TAK-715
TAK-715 ([M+H]+ with m/z 400; C24H22N3OS+) in Light produces three major (CP398A and B
and CP400) and two minor (CP279 and CP296) CPs (Table 2), none of which show affinity,
although affinity of CP398A and CP279 may be masked by that of TAK-715. The fact that
the amide hydrolysis product (CP296, C17H18N3S+) does not show affinity here, as opposed
to H2O2, is most likely a concentration effect. Some other CPs (Table 2) are discussed in Compound p38 RRT m/zb Delta Modification Method
ESM Section 4. Except for CP296, all products were less hydrophilic than the substrate. affinitya

3.2.3 SB203580 Parent X 1.00 264.653 (2) x x x


SB203580 ([M+H]+ with m/z 378; C21H17FN3OS+) in Light forms only minor products. Again, CP230 O 0.95 230.165 (1) -C17H18N2O3 AB EC
no affinity peaks were observed (see 3.1.3). Some CPs (Table 3) are discussed in ESM CP273 O 1.04 273.171 (1) -C16H17NO2 AB+CO+NH3 EC
Section 5. CPs of Light show exclusively decreased hydrophilicity. CP413 X 1.22 413.199 (1) -C6H13NO quinoneimine EC
CP415 X 1.19, 1.27 415.215 (1) -C6H11NO ether hydrolysis EC,HLM
3.3 In vitro metabolism CP458 - 0.90 229.632 (2) -C4H6O 2x N-dealkylation in D HLM
HLM incubations of the four kinase inhibitors will be discussed except for TAK-715 which CP459 - 1.25 459.242 (1) -C4H7N alcohol for morpholino HLM
have been described elsewhere [21], but the results are included in Table 2. This addresses CP502 - 0.90 251.645 (2) -C2H2 O-and N-dealkylation HLM
the question whether the other modification methods produce real metabolites in addition in D
to interesting CPs, because HLM incubations are still the gold standard in cell-free in vitro CP544A - 0.84 272.650 (2) +O OH in A or B HLM
metabolism [32]. CP544B - 0.87 “ “ OH in A HLM
CP1052 - 1.25 526.285 (2) dimerization with two HLM
3.3.1 DMPIP bonds in part C or D
The incubation of DMPIP with HLM results in only one minor mono-oxygenation metabolite
CP557C (C28H31ClFN4O5+), produced by N-oxidation of the C ring (cf. Table 1). a
X: affinity; O: no affinity observed; -: not measured
b
Charge state (n): [M+nH]n+
126 127
morpholino group, exchanging the latter for a hydroxyl group. Most metabolites show CP539 shows decreased hydrophilicity, but the reduction of stereochemical complexity
relatively low UV peak areas, except for the peak that corresponds to the three co-eluting, might be advantageous. Furthermore, CP472 and CP557A are of special interest because
less hydrophilic metabolites (CP415, CP459, and CP1052). This peak has about 20% of they show bioaffinity and increased hydrophilicity. Additionally, CP472 is simpler and the
the UV area of the residual substrate peak. hydroxyl group of CP557A might result in more specific interactions with the target, for
example through hydrogen bonding.
4. Discussion
4.1.2 TAK-715
The CPs related to TAK-715 are listed in Table  2 and ESM Section  4. Electrochemical
4.1 Comparison of compound modification methods conversion was not achieved under the tested conditions [18]. In the other methods,
The results of the five modification methods (H2O2, Light, EC, HLM and BM3) are discussed generally, four types of modifications were observed. Dehydrogenation was found in all
and compared in different ways. First, we look at the structures of the CPs generated methods, whereas oxygenation occurred in HLM, BM3 and H2O2, and the hydrolysis of the
and correlate them to affinity. The parallel setup of on-line post-column affinity assay and amide bond in Light, H2O2 and HLM. A combination of oxygenation and dehydrogenation
MSn enables a straightforward structure-affinity comparison and significantly reduces the was also frequently observed. Additionally, in Light, a loss of the complete benzamide moiety
risk of errors in the correlation. Hydrophilicity, as a physicochemical parameter of major as opposed to simple hydrolysis is observed. This is especially interesting as a similar
importance in modern drug discovery [9], is used in addition to affinity in order to assess reaction is observed in collision-induced dissociation (CID) of the protonated molecule [27].
the quality of the products as potential new leads. To this end, CPs are marked as more HLM and BM3 show the most similar profile, HLM having slightly more products, including
hydrophilic (RRT<0.90), unchanged (0.90≥RRT≤1.10) or less hydrophilic (RRT>1.10) those with amide hydrolysis [21]. However, H2O2 also yields many products with similar
compared to the substrate based on their RRTs. The clogP values of the substrates were modification to HLM and BM3, but with much more isomeric mono-oxygenated products.
calculated with ChemBioDraw version 12 to be 5.8 for BIRB796 (logP 5.2 [34]), 6.5 for Additionally, only H2O2 and HLM show a combination of amide hydrolysis and oxygenation.
TAK-715, 2.7 for SB203580 and 2.3 for DMPIP. Finally, the most interesting CPs as seen Finally, Light shows three products not observed with the other methods.
from various viewpoints are highlighted. The ability to generate human relevant metabolites Twelve of the 32 CPs showed bioaffinity, six did not and for another fourteen the analysis
using the modification methods is also briefly discussed. was inconclusive. The higher number of CPs results in a higher uncertainty in the affinity
determination, mainly owing to co-elution, especially of the mono-oxygenation isomers in
4.1.1 DMPIP H2O2. HLM and BM3 showed the most impressive results in generating CPs with affinity,
An overview of the CPs yielded from DMPIP is given in Table  1 and ESM Section  3. In but also H2O2 created quite a number of affinity compounds. Correlating structure and
general, there is little overlap in the products of DMPIP between the different modification affinity indicates that many of the oxygenation products retain affinity, even after multiple
methods, except the double N-dealkylation product CP501, generated by H2O2 as well
3.3 as EC, and the dehydrogenation product CP539, appearing in Light and EC. CP433 was
almost exclusively produced by BM3, whereas a similar product CP431 with an additional
reactions (up to three were observed). CP416B even has similar affinity as TAK-715 [21].
Loss of the phenyl ketone group by amide hydrolysis seems to have some effect on affinity, 3.3
because the affinity of CP296 is only observed at high concentration in H2O2 but not at the
double bond in the B ring was produced by the EC. Light is the only method inducing lower concentration in Light. This discrepancy might also result from (possible) affinity of
chlorine hydrogen exchange (CP523A to D), isomerisation (CP541A to D) and the addition co-eluting CP294A and/or CP312B. Dehydrogenation by ring formation in CP398B exerts a
of water (CP559). In contrast, methyl abstraction (CP525) and amide hydrolysis (CP472) strong negative influence on affinity, whereas formation of a carbonyl group at the aromatic
are exclusively found in EC. H2O2, EC and HLM all produce an oxygenated compound methyl group in CP430A and CP430B only results in lower affinity, e.g., CP430B affinity is
(CP557A to C). However, all three modifications are found in different parts of the molecule about 20 times lower than TAK-715 [21]. The TAK-715 isomer (CP400) also does not show
and while EC produces hydroxylation, HLM results in N-oxidation. affinity.
Seven of the 22 CPs showed bioaffinity, ten did not and for another five the analysis was By generating many oxygenated CPs, HLM, BM3 and H2O2 also produced a compound
inconclusive. From correlating structure and affinity, we can conclude that the fluorobenzyl library with many more hydrophilic CPs. In Light, this was only the case for CP296. In total,
ring is more important for affinity than the morpholino group, as removal of the former in 23 of the 32 CPs were more hydrophilic, whereas six had decreased hydrophilicity. The
CP433 and CP431 leads to complete affinity loss while upon loss or modification of the latter were equally contributed by Light and H2O2, but accounting for as much as 60% of
latter in CP472 and CP557B affinity is retained. Furthermore, the dimethyl substitution at products in Light and only 16% in H2O2.
the piperazine ring seems crucial as all dealkylations such as the loss of a methyl group A rather unusual reaction for BM3 is the formation of a carboxylic acid from the aromatic
in CP525, and the hydrocarbon losses in CP501 and CP529 delete affinity. Addition of methyl group in CP430B [21]. Though possessing the right elemental composition, the
a hydroxyl group (CP557A) on the other hand seems to be at least less detrimental to aldehyde CP414 from H2O2 is unlikely to be the missing link between the alcohol CP416A
affinity. Dehydrogenation even produces the high-affinity compound CP539. Interestingly, and the carboxylic acid CP430B, as under aqueous conditions an aldehyde would be
the exchange of the chlorine for a hydrogen atom has a huge negative effect on affinity. transformed into the carboxylic acid by a strong oxidizing agent like hydrogen peroxide. The
Additionally, at least two of the three substrate isomers (CP541A to C) are binders. These most interesting CP is CP416B, which not only shows significantly increased hydrophilicity
could be promising modified scaffolds for further lead library diversification. but also similarly high affinity as TAK-715.
Many CPs showed an improvement in physicochemical properties: fifteen of the 22 CPs
were more hydrophilic, five had similar hydrophilicity and only two were less hydrophilic 4.1.3 SB203580
than the substrate. Interestingly, the only metabolite from HLM was less hydrophilic than The CPs of SB203580 which were produced are summarized in Table 3 and ESM Section 5.
the substrate. No affinity signals for CPs of SB203580 have been observed in any of the measurements,
The most interesting compound of this series is probably CP539 as it showed by far the possibly due to the strong auto-fluorescence of SB203580 at the assay wavelength.
highest product affinity peak in all DMPIP derived mixtures. Whether this was due to higher Oxygenation was introduced by all modification methods. Next to the major metabolite, i.e.,
abundance and/or higher affinity than other products cannot be determined. Unfortunately, the S-oxidation product CP394C, also reported by Henklova et al [33], many other mono-

128 129
and double-oxygenated isomers have been generated. In Light, the oxygenation was often identities are not yet confirmed. EC confirmed its value in the generation of reactive
accompanied by additional reactions, and in H2O2 by dehalogenation. Interestingly, the CP metabolites, exemplified by CP413 and CP415, but is also useful for the generation of
profile generated by EC is most similar to HLM, whereas H2O2 produces all products of HLM stable metabolites, as is shown for SB203580. Thus, the combination of all four methods
but also many more, such as CPs involving loss of the sulfonyl group (CP316). The most represents a valuable toolbox for the generation of metabolite standards. HRS contributes
uncommon reactions are again observed with Light, such as S-reduction to the thioether to the application of this toolbox by rapidly identifying the method(s) of choice for a specific
CP362, methoxylation in CP424A and CP424B, and the breakdown of the imidazole ring to substrate. The standards can then be produced by up-scaling and purification which can
CP305 and its related products. also be monitored with the HRS system.
Generating more hydrophilic CPs was less successful: Only six of the 19 CPs were more
hydrophilic. Interestingly, all six more hydrophilic compounds were from H2O2, as were six 5. Conclusion
of the less hydrophilic compounds.
Four modification methods were investigated for their potential to produce metabolite-
4.1.4 BIRB796 like compounds. The main goal was to find CPs with retained affinity and increased
CPs of BIRB796 were only generated by EC and HLM. Initial tests indicated that BM3, hydrophilicity, which may act as lead compounds in further drug discovery. Therefore, the
H2O2 and Light were not suitable to produce interesting compound libraries. The ten CPs data evaluation emphasized the reactions occurring, the affinity of the CPs towards p38α
observed are listed in Table 4 and ESM Section 6. Only the two hydroxylation isomers in and the hydrophilicity of the CPs.
HLM are more hydrophilic. First of all, it can be concluded that the HRS system is very useful for the described type
There is little overlap between HLM and EC CPs. Where HLM mainly catalyses hydroxylation of study. An efficient analysis of the modifications introduced via the different methods
and N- and O-dealkylation, EC is mostly active around the urea function. However, two CPs was enabled. Affinity and identity of the CPs could be assessed simultaneously. However,
generated by both methods are important in metabolism studies: the reactive quinoneimine ambiguity remains in some cases. Limited chromatographic resolution may hinder
CP413 and its corresponding unstable hydroquinone CP415. Both CPs showed affinity. identification of the CP with affinity, especially in very complex mixtures, such as with TAK-715
They are the main products in EC whereas CP415 seems to be among the most abundant in H2O2 and DMPIP in Light (Figure 1). Whereas many conversion reactions like oxygenation,
CPs in HLM. The fact, that CP413 is only detectable in HLM close to the limit of detection, dehydrogenation and amide hydrolysis are readily identified, limited fragmentation (and/or
does not mean that CP413 is not an important metabolite. On the contrary, quinoneimines sensitivity) may hinder structure elucidation of CPs, for example in distinguishing between
may react with free cysteines in proteins to form covalent drug-protein adducts [35]. They N-oxidation and aromatic hydroxylation, or between different oxygenated isomers. Direct
can be detected as GSH adducts in specially designed experiments [36] which were outside quantitation of CPs from a mixture is still missing in the HRS technology, as is also true
the scope of this investigation. for early metabolic profiling, unless radioactivity detection is applied [38]. However, the
3.3 4.2 Conversion products as metabolite standards
qualitative affinity data in combination with the MS structure analysis were sufficient to
achieve an initial structure-activity assessment. The combination of hydrophilicity, affinity
3.3
Another interesting aspect of this study is the possibility to generate metabolites formed and structure efficiently starts the characterization of CPs concerning their usefulness as
using HLM by other conversion methods (H2O2, EC, Light, and/or BM3s), as this may yield potential new lead compounds. The strong link between the three properties, ensured by
an alternative to conventional organic synthesis. The synthesis of standards for metabolic the HRS platform, increases the confidence in all results. All this is possible, and was in
analysis often is a bottleneck in the drug discovery phase. It might be assumed that BM3, fact achieved, without fractionation, directed synthesis or extensive sample preparation.
as another biotransformation method, shows the closest resemblance of product profiles. Mostly, mixtures of related compounds are analysed in a single chromatographic run. This
As discussed in detail elsewhere [21], the HLM and BM3 profiles for TAK-715 were very leads to an extremely efficient collection of a multitude of relevant data on the prospective
closely related for some mutants. Herein, the flexibility of the BM3 approach is crucial. lead compounds. In earlier work, we demonstrated that this provides a very efficient basis
However, H2O2 was found to produce an HLM metabolite of TAK-715 which BM3 did not. to focus purification efforts on favourable compounds which can then be followed up by in-
For DMPIP, the HLM metabolite CP557C was not produced by BM3 or any other method, depth structural characterization and further biological testing [21].
although H2O2 and EC did give other oxygenation products. For SB203580, H2O2 was Although H2O2 and Light converted all tested substrates, the CPs produced did not always
the only method to produce both HLM metabolites, the S- and the N-oxidation product, show affinity or increased hydrophilicity. EC failed to convert TAK-715. After pre-screening,
although in addition to a large number of other compounds. EC worked very well here as BM3, was only applied to TAK-715 and DMPIP. In terms of the number of products,
its only major product was the main metabolite CP394C. For BIRB796, HLM can only be successful combinations of modification method and substrate were EC/DMPIP, Light/
compared to EC. At first glance, the overlap seems quite limited. However, EC produces DMPIP, Light/SB203580, H2O2/TAK715, H2O2/SB203580, BM3/TAK715, HLM/TAK715 and
one (possibly two) very important metabolites as main product(s), CP413 and CP415, HLM/BIRB796.
which, being reactive species might be very interesting for the toxicology of BIRB796 [37]. Retention of affinity is more or less evenly distributed among the tested conversion methods.
Another interesting observation in this study was that, contrary to general expectation, There is no single one which exclusively or predominantly forms affinity compounds. With
many HLM metabolites with decreased hydrophilicity were observed. Only for TAK-715, 50-70% of the related CPs showing retention of affinity, the screening of DMPIP and
all metabolites were more hydrophilic, while for DMPIP and SB203580 exclusively less TAK-715 was quite successful in this respect. All methods, except HLM, are reasonably
hydrophilic metabolites were observed. This underlines the importance of taking into well scalable. H2O2 only requires low cost reagents and Light and EC minor investment
account metabolites with decreased hydrophilicity when developing analytical methods for in apparatus. BM3, however, necessitates expensive purified enzymes and cofactors. Of
metabolite identification. course, this comparison applies to the analytical scale. It might be very different at larger-
Though BM3 has the most possibilities to be tuned for the production of specific metabolites, scale production. High cost and limited availability, apart from ethical considerations, in
the tendency of H2O2 to produce many metabolite-like isomers can also be exploited. combination with a low catalytic activity exclude HLM as an alternative for biosynthesis.
With adequate fractionation technology, it might even be more advantageous to produce H2O2 yielded mostly more hydrophilic CPs, with the exception of some N-oxides and
many standards simultaneously with H2O2, especially at a stage when the true metabolite the general tendency for less hydrophilic CPs in SB203580. H2O2 also produced more
130 131
isomeric species than the other methods. Although this increases the chance of finding Comm. 2 (2011) 349-355.
affinity compounds, it reduces the method’s value in the production of specific CPs. BM3 10. A. Fura, Y.Z. Shu, M. Zhu, R.L. Hanson, V. Roongta, W.G. Humphreys, Discovering drugs
incubation seems less generally applicable but, when applicable, is efficient in generating through biological transformation: role of pharmacologically active metabolites in drug discovery,
more hydrophilic CPs with high affinity. Furthermore, it is possible to tune these enzymes J. Med. Chem. 47 (2004) 4339-4351.
11. D.P. Benfield, C.M. Harries, D.K. Luscombe, Some pharmacological aspects of
for the production of either complex mixtures with a variety of CPs or specific CPs [21].
desmethylclomipramine, Postgrad. Med. J. 56 Suppl 1 (1980) 13-18.
EC is complementary to the other methods by producing unique products, as is most 12 M.J. Kang, W.H. Song, B.H. Shim, S.Y. Oh, H.Y. Lee, E.Y. Chung, Y. Sohn, J. Lee,
obvious in DMPIP and BIRB796. It shows its strength in the conversion of BIRB796 where Pharmacologically active metabolites of currently marketed drugs: potential resources for new
it enables the analysis of the reactive quinoneimine [18]. While it is reasonable to assume drug discovery and development, Yakugaku Zasshi 130 (2010) 1325-1337.
that this metabolite is also produced by HLM, it is the low matrix content and direct analysis 13. M. Bakshi, S. Singh, Development of validated stability-indicating assay methods--critical
capabilities featured by the EC which readily allowed structure and affinity analysis of the review, J. Pharm. Biomed. Anal. 28 (2002) 1011-1040.
reactive species. In this respect, the complexity and reactivity of the sample matrix increase 14. L. Azevedo Marques, M. Giera, H. Lingeman, W.M. Niessen, Analysis of acetylcholinesterase
in the order Light<EC<H2O2<BM3~HLM. A more complex matrix will not only hinder the inhibitors: bioanalysis, degradation and metabolism, Biomed. Chromatogr. 25 (2011) 278-299.
analysis, especially of reactive products, but also increases the difficulty of purification, 15. M. Thevis, I. Moller, A. Thomas, S. Beuck, G. Rodchenkov, W. Bornatsch, H. Geyer, W.
if desired at a later stage. Therefore, EC is the most promising method to study relevant Schanzer, Characterization of two major urinary metabolites of the PPARdelta-agonist GW1516
and implementation of the drug in routine doping controls, Anal. Bioanal. Chem. 396 (2010)
reactive species. Unfortunately, the CPs from EC and Light show no general tendency
2479-2491.
towards more hydrophilicity, although such CPs are still frequently observed. However, 16. L.A. Marques, M. Giera, F.J. de Kanter, W.M. Niessen, H. Lingeman, H. Irth, Photohuperzine
Light opens up a wider range of reaction pathways which often results in at least some A-A new photoisomer of huperzine A: structure elucidation, formation kinetics and activity
unique products. Structural changes induced in Light are not easily identified by HR-MSn as assessment, J Pharm Biomed Anal 52 (2010) 190-194.
Light displays the unique possibility to induce strong structural isomerism even in the core of 17. A. Baumann, U. Karst, Online electrochemistry/mass spectrometry in drug metabolism studies:
the molecule. Inducing structural changes to the molecules core, while maintaining affinity principles and applications, Expert Opin. Drug Metab. Toxicol. 6 (2010) 715-731.
(see CP541B and CP541C of DMPIP), can lead to a new scaffold for optimization which 18. D. Falck, J.S.B. de Vlieger, M. Giera, M. Honing, H. Irth, W.M.A. Niessen, J. Kool, On-line
could be desirable, for example when selectivity of the old scaffold has proven insufficient. electrochemistry-bioaffinity screening with parallel HR-LC-MS for the generation and
Therefore, the four modification methods are highly complementary with regard to the characterization of modified p38alpha kinase inhibitors, Anal. Bioanal. Chem. 403 (2012) 367-
substrates converted, the reactions observed, the isomers formed from similar reactions, 375. [Chapter 3.1]
19. C.J. Whitehouse, S.G. Bell, L.L. Wong, P450(BM3) (CYP102A1): connecting the dots, Chem.
the retention of affinity in different substrates and the tendencies for more hydrophilic CPs.
Soc. Rev. 41 (2012) 1218-1260.
All of them created interesting CPs that have the potential to be further explored as lead 20. H. Venkataraman, S.B.A. de Beer, D.P. Geerke, N.P.E. Vermeulen, J.N.M. Commandeur, Regio-
3.3 compounds. and Stereoselective Hydroxylation of Optically Active alpha-Ionone Enantiomers by Engineered
Cytochrome P450 BM3 Mutants, Adv. Synth. Catal. 354 (2012) 2172-2184.
3.3
6. Acknowledgements 21. V. Rea, D. Falck, J. Kool, F.J.J. de Kanter, J.N.M. Commandeur, N.P.E. Vermeulen, W.M.A.
Niessen, M. Honing, Combination of biotransformation by P450 BM3 mutants with on-line
This research was performed within the framework of project D2-102 “Metabolic stability post-column bioaffinity and mass spectrometric profiling as a novel strategy to diversify and
characterize p38α kinase inhibitors, Med. Chem. Comm. 4 (2013) 371-377. [Chapter 3.2]
assessment as new tool in the Hit-to-Lead selection process and the generation of new
22. D.A. Smith, R.S. Obach, Metabolites in safety testing (MIST): considerations of mechanisms of
lead compound libraries” of the Dutch Top Institute Pharma. Vanina Rea is acknowledged toxicity with dose, abundance, and duration of treatment, Chem. Res. Toxicol. 22 (2009) 267-
for her contribution to the BM3 data. 279.
23. S.E. de Laszlo, D. Visco, L. Agarwal, L. Chang, J. Chin, G. Croft, A. Forsyth, D. Fletcher, B.
References Frantz, C. Hacker, W. Hanlon, C. Harper, M. Kostura, B. Li, S. Luell, M. MacCoss, N. Mantlo,
E.A. O’Neill, C. Orevillo, M. Pang, J. Parsons, A. Rolando, Y. Sahly, K. Sidler, S.J. O’Keefe,
1. A. Barker, J.G. Kettle, T. Nowak, J.E. Pease, Expanding medicinal chemistry space, Drug
Discov. Today 18 (2013) 298-304. Pyrroles and other heterocycles as inhibitors of p38 kinase, Bioorg Med Chem Lett 8 (1998)
2. A. Sharma, P. Appukkuttan, E. Van der Eycken, Microwave-assisted synthesis of medium-sized 2689-2694.
heterocycles, Chem. Commun. (Camb.) 48 (2012) 1623-1637. 24. J. Regan, S. Breitfelder, P. Cirillo, T. Gilmore, A.G. Graham, E. Hickey, B. Klaus, J. Madwed,
3. K. Ebinger, H.N. Weller, J. Kiplinger, P. Lefebvre, Evaluation of a new preparative supercritical M. Moriak, N. Moss, C. Pargellis, S. Pav, A. Proto, A. Swinamer, L. Tong, C. Torcellini, Pyrazole
fluid chromatography system for compound library purification: the TharSFC SFC-MS Prep-100 urea-based inhibitors of p38 MAP kinase: from lead compound to clinical candidate, J Med
system, J. Lab Autom. 16 (2011) 241-249. Chem 45 (2002) 2994-3008.
4. J. Kool, M. Giera, H. Irth, W.M. Niessen, Advances in mass spectrometry-based post-column 25. S. Miwatashi, Y. Arikawa, E. Kotani, M. Miyamoto, K. Naruo, H. Kimura, T. Tanaka, S. Asahi, S.
bioaffinity profiling of mixtures, Anal. Bioanal. Chem. 399 (2011) 2655-2668. Ohkawa, Novel inhibitor of p38 MAP kinase as an anti-TNF-alpha drug: discovery of N-[4-[2-
5. D. Falck, J.S. de Vlieger, W.M. Niessen, J. Kool, M. Honing, M. Giera, H. Irth, Development of an ethyl-4-(3-methylphenyl)-1,3-thiazol-5-yl]-2-pyridyl]benzamide (TAK-715) as a potent and orally
online p38alpha mitogen-activated protein kinase binding assay and integration of LC–HR-MS, active anti-rheumatoid arthritis agent, J Med Chem 48 (2005) 5966-5979.
Anal Bioanal Chem 398 (2010) 1771-1780. [Chapter 2.2] 26. J. Kool, R. Ramautar, S.M. van Liempd, J. Beckman, F.J. de Kanter, J.H. Meerman, T. Schenk,
6. R.J. Mayer, J.F. Callahan, p38 MAP kinase inhibitors: A future therapy for inflammatory diseases, H. Irth, J.N. Commandeur, N.P. Vermeulen, Rapid on-line profiling of estrogen receptor binding
Drug Discov. Today 3 (2006) 49-54. metabolites of tamoxifen, J. Med. Chem. 49 (2006) 3287-3292.
7. S. Kumar, J. Boehm, J.C. Lee, p38 MAP kinases: key signalling molecules as therapeutic targets 27. D. Falck, J. Kool, M. Honing, W.M.A. Niessen, Tandem mass spectrometry study of p38α kinase
for inflammatory diseases, Nat Rev Drug Discov 2 (2003) 717-726. inhibitors and related substances, J. Mass Spectrom. 48 (2013) 718-731. [Chapter 4.1]
8. R.M. Eglen, T. Reisine, The current status of drug discovery against the human kinome, Assay 28. J.V. Odovic, B.D. Markovic, R.D. Injac, S.M. Vladimirov, K.D. Karljikovic-Rajic, Correlation
Drug Dev. Technol. 7 (2009) 22-43. between ultra-high performance liquid chromatography-tandem mass spectrometry and
9. M.M. Hann, Molecular obesity, potency and other addictions in drug discovery, Med. Chem. reversed-phase thin-layer chromatography hydrophobicity data for evaluation of angiotensin-

132 133
converting enzyme inhibitors absorption, J. Chromatogr. A 1258 (2012) 94-100.
29. M. Holcapek, L. Kolarova, M. Nobilis, High-performance liquid chromatography-tandem mass
Supplementary Material
spectrometry in the identification and determination of phase I and phase II drug metabolites,
Anal. Bioanal. Chem. 391 (2008) 59-78. Section 1 Experimental details
30. L. Fang, M. Wan, M. Pennacchio, J. Pan, Evaluation of evaporative light-scattering detector for The optimization of temperature and hydrogen peroxide concentration for H2O2 and of the
combinatorial library quantitation by reversed phase HPLC, J Comb Chem 2 (2000) 254-257. incubation time for H2O2 and Light was monitored on an LC–UV setup. This consisted
31. L.A. Marques, I. Maada, F.J. de Kanter, H. Lingeman, H. Irth, W.M. Niessen, M. Giera, Stability- of two LC-20AD pumps, an SIL-20AC autosampler, an SPD-20AD UV/VIS detector, a
indicating study of the anti-Alzheimer’s drug galantamine hydrobromide, J. Pharm. Biomed. CBM-20A controller (all products of Shimadzu, ’s Hertogenbosch, The Netherlands) and a
Anal. 55 (2011) 85-92.
32. P. Fasinu, P.J. Bouic, B. Rosenkranz, Liver-based in vitro technologies for drug biotransformation
Mistral column oven (Spark Holland, Emmen, The Netherlands). With an injection volume
studies - a review, Curr. Drug. Metab. 13 (2012) 215-224. of 10 μL, chromatographic separations were achieved on a Symmetry C18 column (2.1 x
33. P. Henklova, R. Vrzal, B. Papouskova, P. Bednar, P. Jancova, E. Anzenbacherova, J. Ulrichova, 100 mm, 3.5 μm particles; Waters, Milford, MA, USA) at 40°C and a flow rate of 113 µL/min.
P. Maurel, P. Pavek, Z. Dvorak, SB203580, a pharmacological inhibitor of p38 MAP kinase LC solvents included water, methanol and formic acid in the ratios 99% / 1% / 0.01% for
transduction pathway activates ERK and JNK MAP kinases in primary cultures of human solvent A and 1% / 99% / 0.01% for solvent B. The following gradient was used: 2 min iso-
hepatocytes, Eur. J. Pharmacol. 593 (2008) 16-23. cratic at 20%B, then a linear gradient to 90% at 18 min, washing at 90% for 4 min (22 min)
34. M.J. Barnes, R. Conroy, D.J. Miller, J.S. Mills, J.G. Montana, P.K. Pooni, G.A. Showell, L.M. and afterwards re-equilibration at 20% from 23 min to 30 min. UV detection was achieved
Walsh, J.B. Warneck, Trimethylsilylpyrazoles as novel inhibitors of p38 MAP kinase: a new use at 280 nm.
of silicon bioisosteres in medicinal chemistry, Bioorg Med Chem Lett 17 (2007) 354-357. 10 µl injections of samples with a 100 µM starting concentration were used in all H2O2
35. L. Switzar, M. Giera, H. Lingeman, H. Irth, W.M. Niessen, Protein digestion optimization for and Light conversion experiments with the exception of TAK-715 in Light. TAK-715 was not
characterization of drug-protein adducts using response surface modeling, J. Chromatogr. A
1218 (2011) 1715-1723.
soluble at 100 µM without the presence of hydrogen peroxide which decreased the pH and
36. X. Ma, E.C. Chan, Fluorescence-based liver microsomal assay for screening of pharmaceutical thereby most probably increased the polarity of TAK-715 by protonation in H2O2. Therefore,
reactive metabolites using a glutathione conjugated 96-well plate, Bioconjug. Chem. 21 (2010) the starting concentration of TAK-715 in Light was decreased to 10 µM. Consequentially,
46-55. the injection volume was increased to 100 µL to yield similar concentrations in the LC–UV
37. S. Zhou, E. Chan, W. Duan, M. Huang, Y.Z. Chen, Drug bioactivation, covalent binding to target and HRS analysis.
proteins and toxicity relevance, Drug Metab. Rev. 37 (2005) 41-213. The substrate consumption was quantified by LC–UV using calibration curves of the inhibi-
38. R.F. Staack, G. Hopfgartner, New analytical strategies in studying drug metabolism, Anal. tors at concentrations between 5 and 100 µM for DMPIP and SB203580. For TAK-715, the
Bioanal. Chem. 388 (2007) 1365-1380. injected concentrations were between 50 and 100 µM or between 1 and 10 µM in the cali-
bration for H2O2 and for Light, respectively. All calibration solutions were freshly prepared in
3.3 the same way as the respective sample solutions. This was important, because significant 3.3
matrix effects were observed when comparing calibration curves with and without hydro-
gen peroxide. The injection volumes for the calibration solutions were the same as for the
respective conversion experiments.
For optimization of the incubation time, aliquots were taken at different time points.

In Light at 0, 10, 20, 30, 45, 60, 75, 90, 105 and 120 min for DMPIP;
every 30 min from 0 to 300 min for SB203580; and
at 0, 3, 6, 9, 12, 15, 20, 25 and 30 min for TAK-715.
In H2O2 every 15 min from 0 to 105 min for DMPIP;
every 30 min from 0 to 180 min for SB203580; and
at 0, 75, 120, 165, 210, 255 and 300 min for TAK-715.

This yielded the optimized incubation times and the corresponding conversion rates de-
scribed in the manuscript. Both calibration solutions and samples were measured tripli-
cate. In H2O2, the standard deviation was generally higher than in Light, probably because
photochemical reactions are started and stopped much more reliably by introducing and
removing the light source than the oxidation reaction are by heating and cooling.

Section 2: Overview chromatographic conditions


In the measurements of samples from EC, BM3 and HLM (TAK-715 only), an Xbridge
C18 column (100×2.1 mm with 3.5 µm particles; Waters, Milford, MA, USA) was used.
For samples from H2O2, Light and HLM (DMPIP, SB203580 and BIRB796) experiments,
a Symmetry column (same dimensions and manufacturer) was applied. Because the
results reported here originate from several independent studies, different gradient pro-
grams were used. A flow rate of 113 μL/min and a column temperature of 40°C were ap-
plied in all cases.
134 135
Table S1: Overview of the binary solvent gradients used in different parts of the study, depending on CP505 CP539
conversion method and starting compound investigated.

Time %B Time %B Time %B Time %B Time %B Time %B


0 20 0 40 0 20 0 20 0 20 0 20
2 20 2 40 2 20 2 20 2 20 2 20
18 90 45 90 10 90 8 50 20 50 20 50
22 90 52 90 13 90 10 50 21 50 21 50 CP523A and C
Four isomers CP523A to D (C28H32FN4O5+) are CP541A, B and C
23 20 53 40 14 20 26 90 26 90 26 90 produced by dechlorination and hydroxylation in These are isomers of the substrate DMPIP. Even
30 20 60 40 20 20 28 90 33 90 33 90 Light, none of which show affinity. In contrast to where fragmentation is different among the iso-
CP523B and D, for CP523A and C the spectral mers, interpretation of the fragmentation trees
29 20 34 20 34 20 data was insufficient for a structure proposal. is extremely challenging. Thus, as in this case,
35 20 40 20 40 20 structural isomers of the substrate are often not
CP523B and D identified by LC–MSn.

Used EC, BM3 BM3 H2O2 Light H2O2 Light CP557A


for: (DMPIP) (TAK-715) (DMPIP) (DMPIP, (SB203580) (TAK-715)
and HLM and HLM SB203580)
(DMPIP, (TAK-715)
SB203580,
BIRB796)

Section 3: Structure proposals of DMPIP related CPs CP525


The structure elucidation by LC–MSn was described in detail elsewhere [1]. For the con-
venience of the reader, structure proposals are drawn here on the basis of that analysis. CP557B
3.3 In this way, the structure-activity discussion can be followed more visually. Note that the
structures of the corresponding molecules are depicted, not the ions detected by LC–MSn.
3.3
Additionally, CP537 and CP573, which were not yet discussed, are examined.
DMPIP CP472

CP529
CP557C

CP431 CP501

CP537 CP559
CP537 (C29H34FN4O5+) is a combination of the
dechlorination and ring formation observed in
CP505 and the addition of methanol found in
CP573. The structure elucidation of CP537 was
CP433 hindered by low abundance and limited fragmen-
tation in MSn.

136 137
CP571 CP279 cupies the same position in the new molecule is CP416B
unclear.

CP398A

CP279 (C17H15N2S+) corresponds to a loss of the


benzamide moiety including the amine and an
CP573 additional dehydrogenation. Whether this is a
This product, showing the addition of methanol direct reaction or occurs via the amide hydrolysis
(CP573, C29H35ClFN4O5+), did not provide suffi- product CP296, which is also present in minor
cient fragmentation in MSn to propose a structure. amounts in Light, is unclear.
CP416C
CP294A and B
Section 4: Structure proposals of CP398B
TAK-715 related CPs
The structure elucidation by LC–MSn was de-
scribed in detail elsewhere [1]. For the conve-
nience of the reader, structure proposals are
drawn here on the basis of that analysis. In this
way, the structure-activity discussion can be fol-
lowed more visually. Note that the structures of
the corresponding molecules are depicted, not the CP296
ions detected by LC–MSn. Additionally, some CPs,
which were not yet discussed, are examined.
The low-abundance CPs partially derived from CP416D to J
3.3 amide hydrolysis are relatively polar and show
RRTs similar to CP296. These include CP294A
Unfortunately, due to extensive co-elution
of these seven mono-oxygenation isomers
3.3
and B, CP310, CP312B and CP328. It is reason- CP398C (C24H22N3O2S+), structure elucidation was not
able to assume that these are present as a num- A dehydrogenation product (C24H20N3OS+). which achieved by LC–MSn. As this study was focused
ber of isomers which are not chromatographically cannot be further evaluated due to insufficient on a screening paradigm, further separation was
resolved. LC–MSn data. not attempted. However, their RRTs between
CP310 0.57 and 0.87 indicate that most of them will be
TAK-715 This low-abundance CP of H2O2 combines three CP400 hydroxylated [2].
different modifications, being amide hydrolysis, This CP is one of the major products in Light and
oxygenation and dehydrogenation (m/z 310.102, an isomer of TAK-715 (C24H22N3OS+). CP416K and L
C17H16N3OS+). The RRTs of over 1.1 of these two mono-oxygen-
CP414 ation CPs indicate N-oxidation [2].
CP312 and B A product of combined oxygenation and dehydro-
genation (C24H20N3O2S+). As it was produced by CP430A
H2O2, CP414 is unlikely to be an aldehyde as this
would most likely be converted to the carboxylic
acid under these strong oxidizing conditions.

CP416A

CP328
This low abundant product of amide hydrolysis
and double-oxygenation (m/z 328.113,
C17H18N3O2S+) is made in H2O2.

CP384
In this CP, the sulphur has been exchanged for
oxygen (C24H22N3O2+). Whether the oxygen oc-

138 139
CP430B CP448 CP305 and double-oxygenation (C21H18N3O3S+), this
is one of the two most polar CPs derived from
SB203580 in H2O2.

CP394A and B
These CPs result from aromatic hydroxylation
(RRT 0.76 and 0.80).

CP394C

CP432A One of the two most abundant CPs (by MS re-


Section 5: Structure proposals of sponse) in Light.

SB203580 related CPs CP316


The structure elucidation by LC–MSn was de- CP394D
scribed in detail elsewhere [1]. For the conve- CP394D is an N-oxidation product (RRT=1.24).
nience of the reader, structure proposals are
drawn here on the basis of that analysis. In this CP394E
way, the structure-activity discussion can be fol-
lowed more visually. Note that the structures of
the corresponding molecules are depicted, not the
ions detected by LC–MSn. Additionally, some CPs,
which were not yet discussed, are examined. Interestingly, CP316 (C20H15FN3+) is similar
The CPs marked with an asterisk are related to to a gas phase CID fragment (m/z 315.117,
CP432B CP305 for which two alternate structure propos- C20H14FN3+•). The methyl-sulfoxide group has
3.3 als are given [1]. For the sake of brevity, only one
possible form is given for the related CPs.
been replaced by hydrogen decreasing polarity.
3.3
CP321* CP394E shows strongly reduced polarity which
SB203580 matches cLogP calculation of an ortho-hydroxyl-
ated pyridine [1].

CP410A
Combines the S-oxidation of CP394C and the
aromatic hydroxylation of CP394A.

CP362 CP410B
Combines the S-oxidation of CP394C and the
aromatic hydroxylation of CP394B.
CP432C and D CP243*
These two double-oxygenation isomers CP410C
(C24H22N3O2S+) did not yield sufficient LC–MSn Combines the S-oxidation of CP394C and the
data for structure elucidation. N-oxidation of CP394D.

CP446 CP410D
One of the two most abundant CPs (by MS re-
CP289* sponse) in Light.

CP376
Resulting from a combination of dehalogena-
tion and mono-oxygenation (C21H18N3O2S+), this
is one of the two most polar CPs derived from
SB203580 in H2O2.

CP392
Resulting from a combination of dehalogenation

140 141
CP424A CP273 CP502

CP424B

CP413 CP544A

Section 6: Structure proposals of


BIRB796 related CPs
The structure elucidation by LC–MSn was de-
scribed in detail elsewhere [1]. For the conve- CP415
nience of the reader. structure proposals are CP544B
drawn here on the basis of that analysis. In this
way, the structure-activity discussion can be fol-
lowed more visually. Note that the structures of
3.3 the corresponding molecules are depicted, not the
ions detected by LC–MSn. Additionally, some CPs, 3.3
which were not yet discussed, are examined.

BIRB796

CP458
CP1052
This CP is essentially a dimer of two BIRB796
molecules. The two substrate molecules are
probably linked by two new bonds resulting in
the loss of two times H2 compared to two full
BIRB796 molecules. Both links are found in part
C and/or part D.

CP230
References
1. D. Falck, J. Kool, M. Honing, W.M.A. Niessen, Tandem mass spectrometry study of p38α kina-
CP459 se inhibitors and related substances, J. Mass Spectrom. 48 (2013) 718-731. [Chapter 4.1]
2. M. Holcapek, L. Kolarova, M. Nobilis, High-performance liquid chromatography-tandem mass
spectrometry in the identification and determination of phase I and phase II drug metabolites,
Anal. Bioanal. Chem. 391 (2008) 59-78.

142 143
Section 4 Chapter 4.1

Integrated structure elucidation Tandem mass spectrometry study


approaches for of p38α kinase inhibitors and
high-resolution screening related substances

D. Falck1, J. Kool1, M. Honing1,2, W.M.A. Niessen1,3

1
AIMMS Division of BioMolecular Analysis, VU University Amsterdam,
De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands.
2
DSM Resolve, Urmonderbaan 22, 6160 MD Geleen, The Netherlands.
3
hyphen MassSpec, de Wetstraat 8, 2332 XT Leiden, The Netherlands.
Abstract generated enzymatically, e.g., biosynthetically using (human) microsomal incubations or
by incubation with specific bacterial mutants (BM3) of cytochrome P450s [14]. Alternatively,
The p38 mitogen-activated protein kinase α (p38α) is an important drug target widely other (chemical) means such as photochemistry or oxidative reagents can be applied.
investigated for therapy of chronic inflammatory diseases. Its inhibitors are rather lipophilic, These different means to create chemical diversity of p38α kinase inhibitors are compared
and as such not very favourable lead compounds in drug discovery. Therefore, we explored in a separate study, involving assessment of bioaffinity and identity by the HRS platform
various approaches to access new chemical space, create diversity, and generate lead [15].
libraries with improved solubility and reduced lipophilicity, based on known p38α inhibitors, This paper focuses on the structure elucidation aspect of the HRS platform with special
e.g., BIRB796 and TAK-715. Compound modification strategies include incubation with attention on MS fragment interpretation. The study resulted in a wealth of MS/MS data,
human liver microsomes and bacterial cytochrome P450 mutants from Bacillus megaterium not only for the kinase inhibitors itself, but also for their related substances, that is: their
(BM3) and treatment by electrochemical oxidation, H2O2, and intense light irradiation. conversion products (CPs). The interpretation of these data and the identification of most
The MS/MS fragmentation pathways of p38α inhibitors and their conversion products (CPs) of the CPs of the typical small-molecule kinase inhibitors TAK-715, BIRB796, SB203580,
have been studied in an ion-trap–time-of-flight MSn instrument. Interpretation of accurate- and DMPIP are discussed in more detail in this paper. Emphasis is on specific pathways,
mass MSn data for four sets of related compounds revealed unexpected and peculiar fragments crucial in structure elucidation, noticeable observations of the gas-phase
fragmentation pathways which are discussed in detail. Emphasis is put on the usefulness chemistry, and the potential of using an ion-trap–time-of-flight hybrid instrument in a
of HR-MSn based structure elucidation in a screening setting and on peculiarities of the screening setting, where fragmentation methods have to be comprehensive. Some of the
fragmentation with regard to the analytes and the MS instrument. applied conversion methods enabled the generation of sufficient material of the related
In one example, an intramolecular rearrangement reaction accompanied by the loss of a substances in order to confirm the HRMS identification with 1H NMR spectroscopy data.
bulky group is observed. For BIRB796, the double-charge precursor ion is used in MS2,
providing a wider range of fragment ions in our instrument. For TAK-715, a number of Materials and methods
related compounds could be produced in a large-scale incubation with a BM3 mutant,
thus enabling comparison of the structure elucidation by 1H-NMR and MSn. A surprisingly Chemicals
large number of homolytic cleavages is observed. Competition between two fragmentation The human recombinant mitogen-activated protein kinase p38α isoform and its inhibitors
pathways involving either the loss of CH3• or OH• radicals was observed for SB203580 and DMPIP (1-{6-chloro-5-[(2R,5S)-4-(4-fluorobenzyl)-2,5-dimethylpiperazine-1-carbonyl]-
its conversion products. 3aHindol-3-yl}-2-morpholinoethane-1,2-dione), SB 203580 (4-[4-(4-fluorophenyl)-2-
(4-methylsulfinylphenyl)-1H-imidazol-5-yl]pyridine), BIRB796 (N-[3-(tert-butyl)-1-(4-
Introduction methylphenyl)-1H-pyrazol-5-yl]-N’-[4-[2-(4-morpholinyl)ethoxy]-1-naphthalenyl]-urea),
TAK-715 (N-[4-[2-Ethyl-4-(3-methylphenyl)-5-thiazolyl]-2-pyridinyl]benzamide) were
The mitogen-activated protein kinase p38, especially its isoform p38α, is an important target obtained from various sources. Methanol (LC–MS grade) and formic acid (LC–MS grade)
in drug discovery and development directed at chronic inflammatory diseases like Crohn’s were from Biosolve (Valkenswaard, the Netherlands). All other chemicals were of analytical
disease and rheumatoid arthritis [1-3]. It is involved in signal transduction pathways through grade and were obtained from Sigma-Aldrich (Schnelldorf, Germany).

4.1 successive activation by phosphorylation [4]. Several p38α inhibitors are in different stages
of (pre)clinical drug development [2,5]. Typical examples are TAK-715 [6], BIRB796 [7,8] Generation of related substances of p38α kinase inhibitors 4.1
and SB203580 [9] (see structures in Figures 4-6). Various methods of compound conversion were used. Each of these methods is discussed
In the past few years, we worked on the development of an integrated platform based briefly but has been described in detail elsewhere. Electrochemistry adds (reduction) or
on high-resolution screening (HRS) to assist in lead optimization and enhancing lead removes (oxidation) electrons from the substrate on a surface electrode which results in
diversity in drug discovery processes, e.g., involving an on-line bioaffinity assay for steroid further reaction for example with the solvent [13]. During incubation with (human) liver
metabolites [10]. HRS is the parallel analysis of molecular structure and affinity/activity microsomes [14,15] or with mutants of cytochrome P450 BM3 [14] the enzymes present
towards a target protein performed on a mixture of related compounds and assessing both catalyse specific reactions like hydroxylation. Oxidation with hydrogen peroxide occurs
qualities individually for each compound. Therefore, the platform consists of a combination mainly via reactive oxygen species [15] while photochemical conversion is the result of the
of liquid chromatography and parallel on-line continuous-flow bioactivity/bioaffinity excitation of electrons in the substrate by intense visible light irradiation [15].
screening and high-resolution mass spectrometry (HRMS) [11]. In this way, simultaneous
bioactivity/bioaffinity assessment and (tentative) molecular structure elucidation in mixtures HRS platform: LC with parallel bioaffinity screening and high-resolution MS
of related compounds is possible. The HRS analysis of structure and bioaffinity of the CPs is conducted with the integrated
Recently, we reported an HRS platform to study p38α binding, which enables high-quality LC–p38α binding assay/MS platform shown in Figure 1 which has been previously reported
bioaffinity assessment (Z’=0.8; S/N up to 100) [12]. This enables us to investigate mixtures [12]. In short, the platform consists of a Shimadzu (‘s Hertogenbosch, the Netherlands)
of p38α inhibitors and their related substances, generated by various means of metabolism- LC–MS system, including two LC-20AD and two LC-10AD isocratic pumps, an SIL-20AC
like compound conversion. Such an approach can be especially useful for highly lipophilic autosampler, a CTO-20AC or a CTO-10AC column oven, an RF-10AXL fluorescence
compounds like kinase inhibitors, as the compound modification may result in more polar, detector, an SPD-AD UV/VIS detector, a CBM-20A controller, and an ion-trap–time-of-flight
but still bioactive analogues that are commonly more difficult to generate by conventional hybrid mass spectrometer (IT–TOF) for HRMS, operated with an electrospray ionization
organic synthesis. (ESI) source. The mixtures of CPs were separated in the chromatographic part of the HRS
For example, the use of electrochemistry in an on-line combination with the HRS platform platform on an Xbridge C18 column 100×2.1 mm with 3.5 mm particles (Waters, Milford,
resulted in a number of related substances for p38α kinase inhibitors, including unstable MA, USA) at 40°C using a flow rate of 113  mL/min. The following mobile phases were
and reactive conversion products [13]. Mixtures of related substances can also be employed: eluent A (1 % methanol, 99 % water, and 0.01 % formic acid); eluent B (99 %
methanol, 1 % water, and 0.01 % formic acid). With these, the gradient was constructed as
146 147
elemental composition of the precursor ion. With these restrictions, most often only one
elemental composition was yielded within 5 ppm error. In rare cases, where in spite of the
restrictions several elemental compositions were obtained, the false compositions could
be easily excluded on basis of carbon-hydrogen ratio, carbon-nitrogen ratio and/or the
absence of a sensible modification route in the case of the parent ions and/or the absence
of sensible neutral losses in the case of the fragments. The structures of the CPs should
be viewed as carefully compiled proposals as confirmation by a second, independent
analytical technique or through synthesis of standards was out of the scope of this study.
An exception is presented by the five purified TAK-715 derived CPs analysed by NMR
where such confirmation has been demonstrated.
Where applicable, 1H-NMR analysis was performed on a Bruker Avance 500 (Fallanden,
Switzerland) at 500.23 MHz.

Results
The molecular structure of the major conversion products (CPs) originating from four
structurally different p38α inhibitors was elucidated using accurate-mass measurement
Figure 1: Schematic setup of the HRS platform. The analysis of MS fragmentation and the resulting
and MS2 and MS3 data from an IT–TOF MS instrument. These CPs were generated by
structure proposals of the CPs are the topic of this manuscript. Therefore, the HR-MS part is
highlighted because it provides the accurate mass fragmentation data.

follows: from 0 to 2 min isocratic at 20% B; from 2 to 18 min, linear increase of eluent B from
20% to 90%; from 18 to 22 min, isocratic at 90% B; from 22 to 23 min linear decrease from
90% to 20% of eluent B; from 23 to 30 min re-equilibration to isocratic conditions at 20%
eluent B. Because the different modification techniques were assessed in several studies,
both gradient profile and stationary phase varied. For example, the reaction mixtures were
most complex in the Light and H2O2 incubations, so specialized gradients were used per
substrate. An overview about chromatographic conditions can be found in Section 1 of the
Supporting Information and the associated text.
The eluent was split post-column in a ratio of 1:9 using 13 mL/min in the bioaffinity detection

4.1 and 100 mL/min in ESI-HRMS analysis. The p38α binding assay is not described here as
the bioaffinity data is irrelevant to the structure elucidation and the discussion of the MS 4.1
fragmentation. Ionization by ESI was achieved with the following settings: needle voltage
4.5 kV; source heating block and the curved desolvation line temperature 200°C; drying
gas pressure 62 kPa; nebulizing gas flow-rate 1.5 L/min. The IT–TOF instrument provides a
resolution of ~10,000 (FWHM) in both MS and MSn modes. External calibration on sodium
trifluoroacetate clusters was applied. The mass accuracy achieved for precursor ions and
fragments was generally within ±5 ppm; otherwise, the actual mass accuracy obtained for
a particular ion is specified. Deviations larger than ±5 ppm can mostly be explained by low
abundance or overlap with the isotope pattern of other species.
Full spectra were obtained in the positive-ion mode between m/z 200 and 650. MS2 and
MS3 spectra were obtained in data-dependent mode between m/z  100 and 650 with an
ion accumulation time of 10 ms and a precursor isolation width of 3 Da. MS, MS2 and MS3
spectra as well as bioaffinity data are collected in a single run of the HRS platform. Where
necessary, the collection of MS2 and MS3 spectra was repeated with manual precursor
selection in MS2 using the HRS platform with inactive bioaffinity detection. The collision
energy was set to 75% for TAK-715 and its products and to 50% for all other compounds.
Structure elucidation was based on calculating the elemental composition of precursor and
fragment ions from the accurate-mass measurements. Several restrictions were applied
to limit the analysis to relevant elemental compositions. For the parent ions, the elemental
composition of the substrate plus a reasonable number of atoms which can be added
by the respective modification techniques was used as a cut off. Mostly, this is restricted Figure 2: MS/MS spectra of the four substrates. The precursor ions are selected in the ion trap within
to oxygen, but also carbon and hydrogen were used where for example methoxylation a window of ±1.5 amu around the following m/z values A) m/z 541.2 for DMPIP, B) m/z 378.1 for
was possible. For the analysis of fragment ions, the restriction criterion applied was the SB203580, C) m/z 529.3 for BIRB796 and D) m/z 400.1 for TAK-715.

148 149
different modification methods, being enzymatic incubation with human liver microsomes

Table 1: DMPIP and its conversion products (CPs), showing elemental composition of the CP and the change (Delta) compared to the parent com-
pound, the occurrence of some specific fragments indicated in the text, proposed identification and site of modification (refer to Figure  3), relative
(HLM) and a bacterial cytochrome P450 BM3 mutant as well as by chemical treatment

Technique
using electrochemical oxidation, H2O2, and intense light irradiation. The resulting mixtures

EC, Light
EC, H2O2
of CPs were analysed by LC coupled to a continuous-flow p38α enzyme binding assay and

Light
Light

Light

Light

Light
Light

Light
H2O2

HLM
BM3
parallel HRMS to obtain MS2 and MS3 data. The interpretation of MS3 data is discussed in

EC

EC

EC
EC

EC

EC
this paper, whereas affinity data for the CPs as well as comparison between the various

x
modification strategies is discussed elsewhere [15].

0.74, 0.67

1.22, 1.39
0.69 (C),
0.63 (A),

0.67 (B),
The structure elucidation of CPs generally proceeds in three steps. First, the MS2 and

0.72 (D)
RRT
MS3 data of the parent compounds were interpreted in considerable detail, trying to

1.00
0.57

0.34
0.86

0.88

0.80
0.96

0.88

0.96
1.34
0.75

0.95
1.10
0.80
1.08
recognize representative fragments to act as profile groups in the molecule [16,17]. In
the second step, from a comparison of the elemental composition of the CPs with that of
the parent compound, the nature of the modification was established. In the third step,

Water addition to C or D
double dealkylation in B

Methoxylation in A or B
the fragmentation of the CPs was compared to that of the parent to assess modification

dehydrogenation in B

dehydrogenation in B
All modifications in B
dealkylation of A and

new 5-ring B and C


of the representative fragments in order to narrow down the site of modification and to

Methyl loss from B


Modification

oxygenation in D
amide hydrolysis
dealkylation of A
propose structures of the CPs. Data on relative retention time (RRT) were used to assist
in identifying the modification, e.g., to differentiate between hydroxylation and N-oxide

Exchange of

N-oxide in C
and OH in B
Cl for H in C
formation [18]. In the case of five BM3-generated CPs of TAK-715, a fourth step involved
purification of the CPs by preparative LC and subsequent acquisition and interpretation of

OH in B
the 1H-NMR spectra, guided by the knowledge obtained from the MSn data [14]. The data
are summarized in the Tables 1 to 4. For most CPs, not all fragments are tabulated, but

x
rather the fragments important for structure elucidation. MSn spectra of most of the CPs are

See Figure 3 + text


included in the Supporting Information. The discussion is not comprehensive as it does not

218.000, 218-CO
-H2O-CH2O, CD-

CD or C+ C3H5N
include structure elucidation of all CPs found.

CD-Cl+C3H8N→

CD+C3H7N →
ACD, AC-CO,
BCD•, BC-H2,
Other

C-Cl+C3H8N
CD-Cl+H or
Structure elucidation of DMPIP conversion products

BCD•-HCl,

D, -CH2O
AB-H2-H•
BC-C3H4
The fragmentation of DMPIP (m/z 541.204, C28H31ClFN4O4+) has partly been described

A, -CO
elsewhere [13]. The fragmentation is more readily discussed, if the molecule is divided

-CO2

-H2O
C-H2
retention time (RRT) and the technique by which a particular CP is generated.

H 2O
n.a.

n.a.
n.a.
into four parts (A to D, see Figure 3) at the main fragmentation sites. The major fragment

-CH4
n.a.

n.a.
n.a.

-2x
AB

H2
X

X
4.1 4.1

* CPs generated by EC are discussed in more detail elsewhere.13


n.a.

n.a.
n.a.

+O
C

X
X

X
-C4H7N

-Cl +H

+H2O
n.a.

n.a.
n.a.

n.a.
CD

+O
+O
X

X
X
X

X
-C2H4+O
-C4H7N

+CH2O
-C7H5F

-C7H5F

-C3H4

+H2O

+H2O
Delta

-CH4
-HCl

-HCl
-H2

-H2

+O
x

x




C28H31ClFN4O5+
C28H31ClFN4O4+

C24H24ClFN3O4+
C25H27ClFN4O4+

C27H27ClFN4O4+
C26H27ClFN4O5+
C28H29ClFN4O4+

C28H31ClFN4O4+

C28H33ClFN4O5+
C29H33ClFN4O5+
Composition

C21H24ClN4O4+

C21H26ClN4O4+

C28H30FN4O4+
C28H32FN4O5+
Elemental




CP523B, D
Compound

CP523A,C

CP557B*
CP541C

CP557C
CP541A

CP541B

CP557A
CP431*

CP472*
CP501*

CP525*
CP529*
CP539*

CP571*
CP433

CP505

CP559
Parent

Figure 3: Structure and profile groups of DMPIP as well as proposed structures for the fragment ions.
150 151
observed in MS2 (see Figure 2A) is due to the CD-part (m/z 319.048, C15H12ClN2O4+); the consistent with the addition of H2O (Δ +18.001 Da) to the C or D part, and another showing
complementary AB-part (m/z 223.161, C13H20FN2+) is also observed as well as the C-part the loss of HCl (CP505, m/z 505.226, C28H30FN4O4+). The latter modification probably
(m/z 206.000, C10H5ClNO2+). Two other fragments can be explained by an intramolecular involves the generation of a five-membered ring between carbon formerly carrying the
rearrangement of the fluorobenzyl group, [19,20] which in effect results in the loss of chlorine atom and the B part. At least four isomers are observed of CP523 (m/z 523.235,
dimethylpiperazine (the B-part, C6H14N2) and the formation of the fragment ion with m/z C28H32FN4O5+), which are consistent with a combination of both modifications. The
427.087 (ACD; C22H17ClFN2O4+). The fragment ion with m/z 286.043 (AC-CO; C16H10ClFNO+) observation of several isomers already proves that the chlorine is not directly exchanged
is a secondary fragment of the ACD-ion, involving the loss of both the D-part and CO (see by a hydroxyl group. Despite significant co-elution, two of the major isomers, CP523B and
Figure 3). In MS3 of ACD, a fragment ion with m/z 312.024 (AC-H2; C17H8ClFNO2+, 6 ppm) CP523D, can be identified as loss of Cl from the C-ring and presence of OH in the B-ring.
is formed which is consistent with the loss of part D as morpholine-N-carbaldehyde and This is concluded from an accordingly modified CD-fragment and similarities to CP557A
thus requires ring formation in the A and/or C part. The two other ions observed in MS3, m/z fragmentation, respectively.
284.029 (C16H8ClFNO+, 6 ppm) and m/z 257.041 (most likely C17H7NO2+•, –24 ppm), are The example of the mono-oxygenated CPs nicely demonstrates the strength of the profile-
probably fragments of this AC-H2 fragment resulting from the loss of CO and a combination group approach [16,17]. The structural isomers CP557A, B and C can be shown to be
of HF and Cl•, respectively. MS3 of CD results in the C-ion (m/z 206.000, C10H5ClNO2+). The distinctively modified in part B, D and C, respectively. On top of that, the RRT [18] is very
MS3 spectrum of AC-CO can be readily explained in terms of neutral losses, that is: m/z helpful to identify CP557A as a hydroxylation and CP557C as an N-oxidation product.
258.048 due to the loss of CO, m/z 222.072 due to the loss of CO and HCl, m/z 196.069 However, if the RRT of the product is close to 1.00, as for CP557B, the latter approach is
due to the loss of CO, HCN, and Cl•, and m/z 109.044 due to C7H6F+ (–7 ppm; A-part), but less helpful. In CP539, the addition of a single double bond remarkably changes the pattern
proposing fragmentation routes and fragment structures is more difficult. of proton/hydride rearrangement during CID and even leads to the onset of homolytic
The DMPIP CPs found with all conversion methods are summarized in detail in Table 1; cleavage of the amide bond. Another interesting change in CID fragmentation, caused
MSn spectra of most of the CPs are included in the Section 5 of the Supporting Information. by modification, is observed in the CPs characterized by hydroxylation of the B part,
In general, only a few of the above identified fragments of DMPIP are actually observed namely CP557A and CP523B and D. This modification activates an internal fragmentation
for the CPs, especially the CD-ion. CPs generated by EC are discussed in more detail of the piperazine ring not observed in any of the other CPs, including DMPIP itself. The
elsewhere [13]. interpretation of structural isomers of the substrate remains a challenge as the fragmentation
In HLM incubations, DMPIP gave only one minor mono-oxygenated CP (CP557C; m/z trees are either to similar or the changes are extremely difficult to interpret.
557.197, C28H31ClFN4O5+). Both the (CD+O)-ion in MS2 (m/z 335.045, C15H12ClN2O5+) and
the (C+O)-ion (m/z 221.995, C10H5ClNO3+) in MS3 contain this modification. However, the Structure elucidation of SB203580 conversion products
C-ion is mainly observed with an additional water loss (m/z 203.986, C10H3ClNO2+). The The fragmentation of SB203580 (m/z 378.108, C21H17FN3OS+) in the IT–TOF is similar to
water loss and RRT indicate N-oxidation rather than aromatic hydroxylation in the C-ring that in the Q–TOF [21]. The only fragment ions observed (see also Figure 2B) are due to
[18]. the loss of CH3• (F363; m/z 363.084, C20H14FN3OS•+) and of SOCH3• (F315; m/z 315.117,
In BM3 incubations, DMPIP almost exclusively produced CP433 (m/z 433.165, C20H14FN3•+). The SB203580 CPs found with all applicable conversion methods are
C21H26ClN4O4+), consistent with dealkylation of the fluorobenzyl group (loss of A), which summarized in detail in Table 2; MSn spectra of most of the CPs are included in Section 6
could be identified based on accurate mass only. of the Supporting Information.
4.1 In H2O2, DMPIP produces two abundant CPs. The major product is CP557A (m/z 557.196,
C28H31ClFN4O5+), which is a mono-oxygenated CP. The MS2 spectrum indicates the oxidation
In H2O2, EC and HLM conversion, the major product formed (CP394C; m/z 394.103,
C21H17FN3O2S+) is consistent with transformation of the sulfoxide to a sulfone group [21].
4.1
must have taken place in the B-part, as the m/z-values for the ACD-, CD-, and AC-CO-ions From the unmodified F315 fragment and the absence of F363 (or a corresponding F363+O;
are not changed (see Figure 3). The fragment ion with m/z 376.108 (C18H19ClN3O4+) results m/z 379.080, C20H14FN3O2S+•) alone, S-oxidation cannot be distinguished from CH3-
from the loss of both the A-part (C7H6F) and cleavage in the oxidated dimethylpiperazine hydroxylation. However, under Light, a compound with CH3-hydroxylation was observed
ring, involving the loss of C3H6NO. This ring cleavage, which is not observed for the DMPIP for which H2C=O loss was strongly favored over SO2CH3• loss (see CP321 below). Having
itself, apparently is stimulated by the presence of the extra O at the dimethylpiperazine the fragmentation patterns of both modifications allows to distinguish them. A minor product
group. In MS3, this ion shows losses of either C3H7N (57.056 Da) to an ion with m/z 319.052 in HLM and H2O2 conversion is another mono-oxygenation product (CP394D). The major
(CD; C15H12ClN2O4+) or of the D-part to an ion with m/z 261.043 (C13H10ClN2O2+). This fragment (F363+O) excludes both CH3-hydroxylation and S-oxidation. In MS3, the odd-
behavior in MS3, the RRT of 0.75, and the absence of losses of H2O, OH• or O suggest electron ion F363+O is further fragmented by OH• loss (m/z 362.078, C20H13FN3OS+),
hydroxylation rather than N-oxidation. suggesting N-oxidation [22,23]. This is consistent with the comparably high RRT [18]. The
The minor product (CP501, m/z 501.170, C25H27ClFN4O4+) was also observed in EC strong preference for the CH3• loss over OH• loss from the N-oxide suggests that the OH•
experiments and was identified as resulting from the loss of C3H4 from the dimethylpiperazine loss is significantly easier from the odd-electron ion than from the even-electron ion [24-26].
group [13]. Minor products of H2O2 conversion include two mono-oxygenation isomers (CP394A and
Photochemical conversion of DMPIP yields a product CP539 (m/z 539.187, C28H29ClFN4O4+), CP394B), which also show F363+O as major fragment. However, these two species involve
also observed in EC experiments, [13] which is due to dehydrogenation of the B part. hydroxylation at the aromatic system, as F315+O (m/z 331.112, C20H14FN3O•+) is observed.
This interpretation is supported by the increased lipophilicity, and the observation of an The latest eluting isomer (CP394E) shows the same fragmentation pattern, consistent
unmodified CD fragment in MS2 as well as a neutral loss of the unmodified A part as a with either a lipophilic aromatic hydroxyl product or an unusually stable N-oxide. cLogP
radical leading to a BCD fragment (m/z 430.139, C21H23ClN4O4+•). Interestingly, the modified calculations predict that hydroxylation in ortho-position to the nitrogen of the pyridine ring
AB fragment (m/z 223.161, C13H20FN2+), is observed with m/z 219.129 (C13H16FN2+) results in significantly increased lipophilicity.
and 220.135 (C13H17FN2+•, –10 ppm), thus showing an additional H2 loss, maybe due to Four double-oxygenated isomers (m/z 410.098, C21H17FN3O3S+) were observed in H2O2
conjugation. conversion. The two earliest eluting isomers (CP410A and CP 410B) show the loss of
Other photochemical CPs observed include a product (CP559, m/z 559.213, C28H33ClFN4O5+), SO2CH3• to generate F315+O. This indicates a combination of S-oxidation and aromatic

152 153
hydroxylation; the RRT are in good agreement with assuming an S-oxidation of CP394A

compound, the occurrence of some specific fragments indicated in the text, proposed identification and site of modification (refer to Figure 4), relative
and CP394B. The third isomer (CP410C) shows the loss of either OH• (m/z 393.096,

HLM, H2O2,
Technique

HLM, H2O2
Table 2: SB 203580 and its conversion products (CPs), showing elemental composition of the CP and the change (Delta) compared to the parent
C21H16FN3O2S+•) or SO2CH3• to F315+O, indicating an N-oxide-S-oxide product, which is
also consistent with the RRT, as both modifications increase lipophilicity. F315+O alone

Light

Light
Light

Light

Light

Light

Light
H2O2

H2O2
H2O2
H2O2
H2O2

H2O2
H2O2
H2O2
H2O2

H2O2
does not establish S-oxidation. The fragment might also result from an SOCH2 loss from

EC
m/z 393, in the case of an unmodified sulfoxide group. However, S-oxidation is proven

x
by fragmentation of m/z 393, thus after OH• loss, resulting in the loss of the sulfonyl

1.05, 1.10,

1.12, 1.24
moiety either as SO2CH3• (m/z 314.106, C20H13FN3+, 11 ppm) or as SO2CH4 (m/z 313.101,

RRT
C20H12FN3+•), but not as SOCH2 leading to F315+O. It is interesting to observe that,
because the S-oxidation completely suppresses the CH3• loss, the OH• loss in this case

1.00
1.26

1.58
1.16

1.18
1.53

1.44
0.60
0.68
0.76
0.80

1.12

1.36
0.83
0.88
1.29

1.43
1.37

1.44
occurs from an even-electron ion. Based on RRT, the last isomer (CP410D) seems to be a
combination of the lipophilic aromatic hydroxylation product CP394E and the S-oxidation.
Like for CP410A and CP410B, the only MS2 fragment is F315+O, thus the nature of the

CP394C and CP394D


sulfoxide replacement

CP394B and CP394C

CP394C and CP394E


CP394A and CP394C

OCH3 in pyridine ring


sulfoxide to thioether
methyl hydroxylation
modifications must be the same.

in fluorophenyl ring
CP305 and CP316

CP305 and CP362

Both modifications
Modification

OH at carbon-14,
Photochemical conversion of SB203580 results in two major and four minor products.

-HF + H2O + O
One major product (CP362; m/z 362.113, C21H17FN3S+) results from the conversion of the

see figure S3

aromatic OH
aromatic OH

N-oxidation
sulfoxide into the corresponding thioether. Fragment ions consistent with the loss of CH3•

CP305 and

S-oxidation
-HF + H2O
and CH3S• confirm this interpretation.

see text
Based on its elemental composition and the assumption that no major rearrangement
takes place, the other major product (CP305; m/z 305.077, C15H14FN2O2S+) involves a

x
decomposition of the imidazole ring, that is: replacing pyridine and its linking carbon in
the imidazole ring by an O-atom (see structure proposals & comments in Section 3 of the

-C8H7NOS, C7H4FO+
-OH•→-SO2CH3• or
analogous CP305

analogous CP305

analogous CP305
Supporting Information). The position of the O-atom is not easy to assess as MSn fragments

(F363+O)→-OH•
show an arrangement of the N-atoms which is different from the original imidazole ring. Both

see figure S3
Other
phenyl rings are observed with a free amide, thus suggesting that carbon-3 and -5, each with

-C8H7NOS,
an N-atom linked to it, are bridged by the O-atom. However, whether this rearrangement of

C8H8FO2+
-SO2CH4
-CH2O,
the modified imidazole ring occurs in the liquid phase during light irradiation or in the gas

retention time (RRT) and the technique by which a particular CP is generated.


phase during ionization and/or fragmentation is not clear. Three of the minor products can

n.a.

n.a.
n.a.
be considered to be secondary products of CP305. CP243 (m/z 243.094, C14H12FN2O+)
is consistent with the replacement of the methylsulfoxide with H, CP289 (m/z 289.081,

F315
C15H14FN2OS+) with conversion of the sulfoxide into the corresponding thioether, whereas

n.a.

n.a.
n.a.
+O
+O

+O
+O
+O
+O

+O
4.1 4.1

X
CP321 (m/z 321.072, C15H14FN2O3S+) involves hydroxylation of the methyl group, as
indicated by the loss of H2C=O to yield the ion with m/z 291.059 (C14H12FN2O2S+) rather
than the loss of SO2CH3• that would occur after S-oxidation (see CP394C).

-C6H3N

+CH2O
F363

n.a.

n.a.
n.a.
Two minor isomeric products (CP424A and CP424B; m/z 424.115, C22H19FN3O3S+) result

+O

+O
+O

+O
+O

+O
-O
X
from combined hydroxylation and methoxylation. In MS2, both CPs generate an ion with
m/z 259.088, due to the loss of C8H7NOS (H3C–S(=O)–C6H4–CºN), indicating that the
modifications are either in the pyridine or in the fluorophenyl ring. In the MS2 spectrum of the

-SOCH3+H

-SOCH3+H

-F+H+2xO
-C6H3N+O

+O+CH2O
CP305+O
CP305-O

-F+H+O
earlier eluting CP424A, the ion with m/z 155.052 (C8H8FO2+) indicates both modifications

CP305

+2xO
Delta

+O
-O
have occurred in the fluorophenyl ring. A similar fluorophenyl fragment was not observed





for other CPs of SB203580. For CP424B, the fluorophenyl fragment is observed with m/z

C15H14FN2O2S+

C15H14FN2O3S+

C21H17FN3O2S+

C21H17FN3O3S+

C22H19FN3O3S+
Composition
C21H17FN3OS+

C15H14FN2OS+

C21H18N3O2S+
C21H18N3O3S+
Elemental

C14H12FN2O+

C21H17FN3S+
C20H15FN3+





Compound

CP394C

CP394D

CP410C

CP410D
CP394A
CP394B

CP394E
CP410A
CP410B

CP424A

CP424B
CP243

CP289
CP305

CP316
CP321

CP362
CP376
CP392
Parent

Figure 4: Structure of SB203580.


154 155
123.025 (C7H4FO+) in MS3. This correlates to a quinone like structure of this carbocation CD-part with cleavage on either

compound, the occurrence of some specific fragments indicated in the text, proposed identification and site of modification (refer to Figure 5), relative

Technique
including carbon-3 and an added oxygen. As the para-position is blocked by fluorine, the side of the carbonyl. This results in

EC,HLM
aromatic hydroxylation must be ortho to carbon-3, otherwise, there would be no quinone. the fragments AB with m/z 230.164

Table 3: BIRB796 and its conversion products (CPs), showing elemental composition of the CP and the change (Delta) compared to the parent
Since the methoxylation is present in the ion with m/z 259 but not in the ion with m/z 123, (C14H20N3+), AB+CO with m/z 256.143

HLM
HLM
HLM
HLM
HLM
HLM
EC
EC
EC
it must have occurred in the pyridine ring. (C15H18N3O+), CD with m/z 273.158

x
Interestingly, the loss of SO2CH3• indicates S-oxidation (CP394C) while methyl hydroxylation (C16H21N2O2+, –7 ppm), and CD+CO

1.19, 1.27
leads to different fragmentation (CP321), but only because the methyl hydroxylation is more with m/z 299.138 (C17H19N2O3+). Both

RRT
unambiguously interpretable. This means that both structures can be determined without AB fragments show the loss of the

1.00
0.95
1.04
1.22

0.90
1.25
0.90
0.84
0.87
1.25
standards when both fragmentation trees are available. CP424B is one of the rare cases B-part (C4H8) to the A-part, that is: m/z
were an aromatic hydroxylation can be linked to a single carbon atom (14a and 14b are 200.082, (A+CO; C11H10N3O+) and m/z
chemically identical). The reason is that a very small fragment containing the hydroxylation 174.103 (A; C10H12N3+) (see Figure 5).

O-and N-dealkylation in D
can be generated whose quinone-like structure can only be explained by ortho-, but not In our instrument, BIRB796 also

alcohol for morpholino


by meta-substitution. Most importantly, the ability of the IT–TOF instrument, as opposed to generated a double-charge ion

2x N-dealkylation in D

dimerization with two


bonds in part C or D
a Q–TOF type instrument, to provide a parent daughter relationship among the fragments ([M+2H]2+; m/z 264.652, C31H39N5O32+).

Modification
is crucial in elucidating the structure of some CPs, especially CP410C. The MS2 spectrum with the double-

ether hydrolysis
charge ion as precursor ion partly

quinoneimine
AB+CO+NH3

OH in A or B
Structure elucidation of BIRB796 conversion products yields all the fragments observed in
For a discussion on its fragmentation, BIRB796 (m/z 529.300; C31H38N5O3+) can be divided the MS2 of the single-charge ion, but

OH in A
into four parts (see Figure 5). Major fragments (see Figure 2C) result from cleavages in in addition fragment ions with m/z

AB
the urea moiety, leading to two sets of complementary fragments: the AB-part and the 414.206 (ABC+•; C25H26N4O2+•), m/z

x
413.197 (ABC-2H; C25H25N4O2+),
both due to the loss of the D-part,

-H2O, AB+•-H2
See Figure 5
as well as fragment ions with m/z

Other

n.a.
n.a.
n.a.

AB, AB+•
229.156 (AB+•; C14H19N3+•, –6 ppm),
m/z 114.092 (D; C6H12NO+, 6 ppm),

C+•
and m/z 100.076 (D–CH2; C5H10NO+)

C
(see Figure 5). It is quite intriguing to

retention time (RRT) and the technique by which a particular CP is generated.


observe the formation of pairs of odd-

n.a.
n.a.
n.a.
D

X
X
electron and even-electron ions for
m/z 414/413 and for m/z 229/230 from
the double-charge ion. No double-
4.1 4.1

A+CO

n.a.
n.a.
n.a.

+O
charge fragment ions were observed.

X
X

X
In this particular case, the MS2 on
the double-charge precursor ion also
enables to get around the low-m/z

AB+CO

n.a.
n.a.
n.a.

-H2
-H2
cut-off typical of the ion trap: for the

X
X
X
X

X
double-charge precursor ion with m/z
264.7, fragment ions with m/z 100 and

+C31H37N5O3
114 are observed, whereas they were

-C17H18N2O3
-C16H17NO2
-C6H13NO
-C6H11NO
-C4H6O
-C4H8N
not for the single-charge precursor

-2xH2
-C2H2
Delta

+O
ion with m/z 528.3. Further MS3


fragmentation of the various fragment
ions is summarized in Section 2 of the
Supporting Information. In the MS data

Composition

C62H72N10O62+
of the CPs, not all these fragments

C31H39N5O32+

C27H33N5O22+

C29H37N5O32+
C31H39N5O42+
Elemental

C25H25N4O2+
C25H27N4O2+

C27H31N4O3+
C15H21N4O+
are actually observed; AB+CO, A+CO

C14H20N3+
and D were observed most frequently.


As the fragmentation of the double-
charge ion provides better abundance
and information of the four profile
Compound

groups recognized in BIRB796, it

CP544A
CP544B
CP1052
CP230 was chosen as precursor ion in the
CP273
CP413
CP415
CP458
CP459
CP502
Parent

Figure 5: Structure and profile groups of BIRB796 as well as proposed structures for the fragment structure elucidation of most CPs. The
ions in MS2 with [M+H]+ or [M+2H]2+ as precursor ions. BIRB796 CPs found with all conversion

156 157
methods are summarized in detail in Table 3; MSn spectra of most of the CPs are included also a consequence of the stabilization of the amide bond. This cleavage is also frequently
in Section 7 of the Supporting Information. found in the MS2 spectra of the CPs, and can thus be used to exclude modification in
In HLM incubations, BIRB796 generates a complicated mixture of CPs. Two major mono- this phenyl ring. Additionally, several minor fragments are observed. One is the ion F297
oxygenated CPs (m/z 272.650, C31H39N5O42+; [M+H]+ with m/z 544.293) were observed, (m/z 297.106, C17H17N2OS+) which can only be explained by an unusual rearrangement
CP544A and CP544B, for which RRTs suggest hydroxylation. For CP544A, in-source of the amide oxygen from the hydroxylimine form, first leading to the structure proposed
fragmentation yields an ion with m/z 263.646, consistent with an easy H2O loss. In MS2, in Section 4 of the Supporting Information and then to the loss of benzonitrile (C6H5–CºN,
three fragments are observed, m/z 114.093, indicating no change took place in the D-part, 103.042 Da). Secondary fragmentation is considered uncommon in a single fragmentation
as well as m/z 254.129 (C15H16N3O+) and m/z 227.141 (C14H17N3+•), consistent with AB+CO– experiment in ion-trap MS instruments. However, several fragments of TAK-715, which
H2 and AB+•–H2, respectively, most likely due to a H2O loss, indicating hydroxylation in require two bond cleavages for formation, were observed both in MS2 and MS3. The
the AB-part. For CP544B, the modification can be attributed to the A-part, because an fragment F264 (m/z  264.072, C16H12N2S+•) originates from a loss of CH3• from F279.
A+CO+O fragment (m/z 216.077, C11H10N3O2+) is observed. This is supported by observing the fragmentation pathway m/z  400.148 → m/z  279.095
Another major conversion product is CP502 (m/z 251.645, C29H37N5O32+; [M+H]+ with m/z → m/z 264.072 in the MS3 experiments. Based solely on this fragmentation, it is unclear
502.283), consistent with a combined O- and N-dealkylation (effective loss of C2H2) from whether the methyl radical is lost from the methyl or the ethyl group. However, the fragment
the morpholine ring. The observation of AB+CO, AB, AB+•, and A+CO fragments in the F224 (m/z  224.053, C14H10NS+), which is observed in MS2 and in MS3 via the pathway
MS2 spectrum confirms this interpretation. m/z 400.148 → m/z 279.095 → m/z 224.053, is helpful in this respect. The difference in
CP458 (m/z 229.632, C27H33N5O22+; [M+H]+ with m/z 458.255) is a minor CP, resulting from elemental composition between F279 and F224 is C3H5N which is consistent with the loss
a double N-dealkylation in the morpholine ring, thus involving the effective loss of C4H6O of propionitrile from the ethylthiazole part (carbon-1, carbon-2, carbon-3 and nitrogen-7;
(dihydrofuran). This modification is supported by the observation of an unmodified AB+CO see Figure 6). The ethyl group is thus no longer present in the F224 fragment. By observing
fragment. an additional pathway m/z  400.148 (→ m/z  279.095) → m/z  224.053 → m/z  209.029,
A major product CP415, observed as single-charge ion (m/z 415.215, C25H27N4O2+; effective where CH3• is lost from the F224 fragment, the ethyl group is excluded as source of this
loss of C6H11NO), results from O-dealkylation of the ethyl-morpholino group (loss of D). This radical. The remaining two minor fragments in MS2, F252 (m/z 252.084, C16H14NS+) and
and the related CP413 (m/z 413.199, C25H25N4O2+; effective loss of C6H13NO), which is F197 (m/z  197.042, C13H19S+), result from the loss of HCN from the pyridine ring of the
consistent with subsequent hydroquinone formation, have been observed by EC conversion ions F279 and F224, respectively. Next to the analysis of the parent fragmentation, the
[13]. Although the modification into CP415 can be quite unambiguously identified from the 1
H-NMR signals of TAK-715 were assigned [14]. Large-scale incubation of TAK-715 with
accurate mass, further confirmation is obtained from MS2 data, showing fragments due BM3 mutant M11 and subsequent preparative LC resulted in sufficient material of five CPs
to AB+CO and A+CO, indicating the changes are not in the AB-part, and an ion with m/z to allow 1H-NMR spectroscopy. These data are shown in Table 5 and discussed here only
160.078 (C10H10NO+, 11 ppm) consistent with the C-part. in context of the identification by MSn; MSn spectra of most of the CPs are included in
Another CP (m/z 526.285, C62H72N10O62+ and m/z 351.192, C62H73N10O63+; [M+H]+ with m/z Section 8 of the Supporting Information.
1051.555) is a dimeric product involving two new (C–C) bonds, as four hydrogen atoms Three fragments were picked as profile groups, representative of different parts of TAK-715:
are missing. In MS2, fragments AB+CO and A+CO are observed, indicating the linkages (1) F279 or its corresponding fragments, which is due to the loss of benzylamide, was used
must be in part C or D. to observe or exclude modification in the cleaved phenyl ring, (2) F264 was employed as
4.1 This compound shows a strong preference for the double-charge ion, both in the parent
and some CPs. This is especially striking considering the relatively small differences in
representative of the CH3 group (carbon-14), and (3) F224 or its corresponding fragments
which is representative of the ethyl group (carbon-2 and -3). The TAK-715 CPs found with
4.1
relevant parameters such as size, polarity or number of nitrogen atoms as compared to, for all conversion methods are summarized in detail in Table 4.
example, DMPIP. In fact, the exact position of one of the protonation sites can be deduced CP416A (m/z  416.143, C24H22N3O2S+) is one of the mono-oxygenated products. In MS2,
by following the change from double-charge to single-charge preference in the CPs (see it shows a fragment with m/z  277.080 (C17H13N2S+) which corresponds to F279+O–
Table 3). All CPs retaining the morpholine-N prefer the double-charge state, but as soon as H2O. Although both an OH group and an N-oxide can show the loss of H2O, the RRT
the morpholine-N is replaced by a hydroxyl group (CP459) the single-charge ion becomes points to hydroxylation [18]. The H2O loss suggests the presence of an α-H, enabling
more abundant. Thus, the morpholine group is protonated at the N-atom. a 1,2-elimination, thus an OH group in an aliphatic rather than in an aromatic system.
Furthermore, an MS3 fragment (m/z 262.056, C16H10N2S•+), corresponding to F264+O–H2O,
Structure elucidation of TAK-715 conversion products seems to exclude the modification of the CH3 group at carbon-14 (Figure 6). This would
Fragmentation analysis of TAK-715 and its conversion products is not trivial. TAK-715 lead to the conclusion that one of the ethyl carbons has been hydroxylated. However, NMR
shows a very complicated fragmentation tree with a large number of fragments. On top of analysis [14]. suggests otherwise: All aromatic protons are still present, only the m-methyl-
that, the fragmentation pattern changes drastically in some CPs. This can give additional phenyl ring protons are shifted to a lower field. The triplet and the quartet of the ethyl
information, but restricts the usefulness of characteristic parent fragments in the profile- group are unchanged, whereas the singlet of the methyl group has shifted to a higher
group approach [16,17]. In MS2, TAK-715 (m/z  400.148, C24H22N3OS+) shows two major field. Unfortunately, as the signal is masked by a solvent signal, it cannot be integrated.
fragments (see Figure 6 and Figure 2D). A loss of water (m/z 382.137) is probably favoured Nevertheless, it still proves hydroxylation of the CH3 group at carbon-14. This apparent
by an equilibrium between the amide moiety and its hydroxylimine resonance structure, mismatch between the MSn and the NMR data can be explained by assuming that the H2O
stabilized by coupling of the aromatic systems of the phenyl and the pyridine ring and an loss is accompanied by the rearrangement of a remote hydride to carbon-14. This would
intramolecular hydrogen bond to the pyridine nitrogen (see Figure  6). The other major explain the H2O loss as well as the loss of the apparently unmodified CH3 group as CH3•. In
fragment F279 (m/z 279.095, C17H15N2S+) corresponds to the loss of the benzamide (C6H5– conclusion, the hydroxylation in CP416A was assigned to carbon-14.
C(=O)NH2, 121.053 Da). Although fragmentation of the amide bond would be expected CP416B (m/z  416.143, C24H22N3O2S+) is another mono-oxygenated product. In the MS2
with complementary fragment ions with m/z 105.034 due to C6H5–CºO+ and m/z 296.122 spectrum of CP416B, an ion with m/z  295.092 (C17H15N2OS+, 6 ppm), corresponding to
[27], the fragment ion F279 is observed instead. This unexpected fragmentation is probably P297+O–H2O, is more dominant than the ion with m/z 277.080, corresponding to P279+O–

158 159
H2O. A strong indication for hydroxylation of the ethyl moiety is found in a fragment

compound, the occurrence of some specific fragments indicated in the text, proposed identification and site of modification (refer to Figure 6), relative

Light, HLM
Technique

BM3, HLM
BM3, HLM
BM3, HLM

BM3, HLM

BM3, HLM

BM3, HLM
with m/z  372.118 (C22H18N3OS+), consistent with a loss of acetaldehyde (C2H4O); no
corresponding fragment is observed for the parent. Ethyl hydroxylation is supported by

(BM3),
Table 4: TAK-715 and its conversion products (CPs), showing elemental composition of the CP and the change (Delta) compared to the parent
NMR data: [14]. The triplet of the three protons on carbon-3 has changed to a doublet at

Light

Light
Light

Light
HLM

HLM

HLM

HLM
slightly lower field and the proton at carbon-2 is still split into a quartet but the signal has

x
shifted to a lower field by almost 2 ppm. Unfortunately, integration of this signal was not

RRT

0.75,
successful because of a very strong overlapping water signal at ca. 4.9  ppm. All other

1.00
1.14

0.35
0.21

1.04
1.35

1.18
0.80
0.83
0.87
0.64

0.84

0.58
0.70

0.62

0.44
protons were accounted for by integration: the methyl singlet and all aromatic signals are
unchanged. In conclusion, hydroxylation in CP416B has taken place at carbon-2 in the
ethyl group.

amide hydrolysis and OH

OH at carbon-2 or-3 and


carbon-9 and carbon-20
CP432A (m/z  432.138, C24H22N3O3S+) is a double-oxygenated product. The fragment

ring formation between

aldehyde at carbon-14

CP416A and CP416B


two OH in ethyl group
ion in MS2 with m/z  293.074 (C17H13N2OS+) corresponds to F279+2xO–H2O. In MS3 of

benzamide loss and


Modification

CP430B and OH at
Isomer of TAK-715
this fragment, a fragment ion with m/z 250.056 (C15H10N2S+•), consistent with a homolytic

carboxylic acid at
dehydrogenation
amide hydrolysis

OH at carbon-14
cleavage of a C2H3O•, reveals a modification in the ethyl group. At first, this could suggest

OH at carbon-2

carbon-2 or-3
that both modifications took place in the ethyl group as otherwise the loss of an ethoxy

carbon-14
radical (C2H5O•) would have been expected. However, as already discussed for CP416A,
H2O loss from a hydroxylation at carbon-14 can result in hydride rearrangement from

OH?
remote positions. In the fragment with m/z 293.074, the water loss from the methyl group
has already occurred, probably forming a carbonyl group from the hydroxyl group in

x
the ethyl moiety, which could explain the elimination of the C2H3O•. The NMR data [14].

-C7H4O-H2O→-CO

See CP430B (+O)

F279+3xO-2xH2O
F264-H2, F224-H2

-C2H4O2-HSCN
-C7H4O-H2O→
-H2O→-C3H5N

-CO→-C3H3N
F264+O-H2O
See Figure 6

F279-C2H3O•
F297-HCNO
Other

n.a.

n.a.

-C2H4O2,
-C7H4O,
-C2H4O
retention time (RRT) and the technique by which a particular CP is generated.

+2xO-H2O

+3xO-H2O
+2xO -H2

-H2-H2O
+O-H2O
+O-H2O

+3xO
F279

n.a.

n.a.

-H2
-H2

+O
X

X
4.1 4.1

+2xO-H2

-H2-H2O
+O-H2O

+2xO

+3xO
F297

-H2O
n.a.

n.a.

-H2
-H2
X

X
-C7H5NO

+2xO-H2

+3xO-H2
-C7H4O

-C7H4O

+2xO

+3xO
Delta

-H2

-H2
+O

+O
x

x



C24H22N3O2S+

C24H20N3O3S+

C24H22N3O3S+

C24H20N3O4S+

C24H22N3O4S+
Composition
C24H22N3OS+

C17H18N3OS+

C24H20N3OS+

C24H22N3OS+
Elemental

C17H15N2S+

C17H18N3S+


10ppm
TAK-

P10
P9

P5
P6
P8
P3

P7

P1
P4

P2
Compound

CP416C
CP398A
CP398B

CP416A
CP416B

CP430A

CP430B

CP432A
CP432B
CP279

CP296

CP312

CP400

CP446

CP448
Parent
Figure 6: Structure of TAK-715 and proposed structures for its fragment ions in MS2 and MS3. The
fragmentation pathway is based on ion-trap MSn data, as outlined in the text.
160 161
Table 5: 1H-NMR-spectra of TAK-715 and major CPs formed by BM3. m/z 326.097 (C17H16N3O2S+), consistent with a common amide bond cleavage. This results
in a fragment ion with an amine group at the pyridine ring and the loss of a neutral (C7H4O).
In MS3, the fragment with m/z 308.085 shows the loss of CO (m/z 280.091, C16H14N3S+).
This serial loss of water and CO points to the presence of a carboxylic acid function. In the
NMR spectrum, [14]. again, no changes are observed in the number of aromatic protons or
in the shifts for the protons in the phenyl and the pyridine ring. Furthermore, the ethyl group
signals are unchanged. This indicates a carboxylic acid group in CP430B on carbon-14.
Indeed, this also explains the shift to an even lower field than in CP416A of the m-methyl-
phenyl ring protons.
CP446 (m/z 446.118, C24H20N3O4S+), the fifth compound of which NMR data were obtained,
contains a triple-oxygenation and a dehydrogenation. Its fragmentation pathway m/z 446.118
→ m/z 324.080 → m/z 296.083 is equivalent to the pathway 430.123 → m/z 308.085 →
m/z  280.091 observed in CP430B, keeping in mind the difference of one oxygen atom
TAK-715 CP432A CP446 CP416A CP416B CP430B between the CPs. Therefore, the carboxylic acid group at carbon-14 is expected to be
Ha 1.45-1.48 1.61-1.63 1.64-1.65 1.45-1.48 1.62-1.64 1.44-1.49 present in CP446 as well. Further analogies between CP446 and CP430B are observed
(3H,t) (3H,d) (3H,d) (3H,t) (3H,d) (3H,t) in the aromatic region of the NMR spectra where shifts, integrals and coupling are almost
Hb 3.09-3.13 5.08-5.12 5.08-5.12 3.09-3.14 5.08-5.12 3.08-3.14 identical. This suggests not only a carboxylic acid on carbon-14 but also the localization of
(2H,q) (1H,q) (1H,q) (2H,q) (1H,q) (2H,q) the third additional oxygen atom in the ethyl group. Indeed, the proton signals for the protons
Hc 2.33 (3H s) ~ 4.9* (s) - ~ 4.9* (s) 2.32 (3H,s) - at carbon-2 and carbon-3 are identical to those in CP416B and CP432A. To summarize,
CP446 is modified to a carboxylic acid on carbon-14 and a hydroxyl moiety at carbon-2. A
Hd 7.19-7.26 7.19-7.26 7.64-7.66 7.35-7.39 7.18-7.25 7.58-7.62
(3H,m) (3H,m) (1H,d) (3H,m) (3H,m) (1H,d) combination of the modifications found in CP416B and CP430B possibly leads to CP446,
keeping in mind the undefined stereochemistry at carbon-2.
He “ “ 7.43-7.46 “ “ 7.40-7.43
(1H,t) (1H,t) These five CPs were observed with both BM3 mutant M11 and HLM incubations. In
HLM incubations, five other CPs were observed, two of which were also produced by
Hf “ “ 7.99-8.01 “ “ 7.98-8.00
(1H,d) (1H,d)
BM3, however, not abundant enough in the large-scale incubation to justify purification.
Therefore, their possible structures have to be derived from MS3 data only.
Hg 7.34 (1H,s) 7.51-7.54 8.18 (1H,s) 7.51-7.54 7.34 (1H,s) 8.14 (1H,s)
(3H,m) (3H,m)
CP432B (m/z  432.138, C24H22N3O3S+) is another double-oxygenated product. The MSn
data suggests that both hydroxylations have taken place in the ethyl group, probably one
Hh 6.95-6.96 7.00-7.01 7.00 (1H,s) 6.98-6.99 6.97-6.98 6.95-6.96
at each carbon. The main clue is a fragment with m/z 372.119 (C22H18N3OS+), consistent
(1H,dd) (1H,dd) (1H,dd) (1H,dd) (1H,dd)
with a loss of 60.021 Da, due to either C2H4O2 or two consecutive losses of H2C=O. An
Hi 8.21-8.22 8.23-8.24 8.25-8.26 8.22-8.23 8.22-8.23 8.21-8.22
additional fragment with m/z 313.136 (C21H17N2O+), consistent with a loss of HS=C=N from
4.1 Hj
(1H,d)
8.31 (1H,d)
(1H,d)
8.30 (1H,d)
(1H,d)
8.33 (1H,d)
(1H,d)
8.29 (1H,d)
(1H,d)
8.31 (1H,d)
(1H,d)
8.31 (1H,d)
the ion with m/z 372.119, links the 60.021 Da loss to the ethyl group as the HS=C=N loss
from the thiazole ring is only possible after the loss of the ethyl moiety as C2H4O2.
4.1
Hk - - - - - - CP430A (m/z  430.125, C24H20N3O3S+, 6 ppm) is another double-oxygenated and
Hl 7.94-7.96 7.94-7.95 7.94-7.95 7.94-7.95 7.94-7.96 7.94-7.95 dehydrogenated product. In MS2, a fragment ion with m/z  308.086 (C17H14N3OS+) is
(2H,d) (2H,d) (2H,d) (2H,d) (2H,d) (2H,d) observed, corresponding to the loss of water and 104 Da (C7H4O), similar to CP430B.
Hm 7.51-7.54 7.51-7.55 7.51-7.54 7.51-7.54 7.51-7.54 7.51-7.54 The ion with m/z 308.086 yields informative MS3 data. A neutral loss of CO to m/z 280.092
(2H,d of d) (3H,m) (2H,d of d) (3H,m) (2H,d of d) (2H,d of d) (C16H14N3S+), also observed in CP430B, indicates a carbonyl group. The subsequent loss
Hn 7.59-7.62 7.59-7.62 7.58-7.60 7.59-7.62 7.59-7.62 7.58-7.62 of CH2=CHCºN (acrylonitrile) to an ion with m/z  227.063 (C13H11N2S+) indicates that the
(1H,t) (1H,t) (1H,t) (1H,t) (1H,t) (1H,t) earlier water loss involves two protons from the ethyl group. Otherwise, a propionitrile loss
(C2H5CºN), as observed for the parent (F279 > F224; m/z 279.095 → m/z 224.053), would
* Chemical shift and integral of benzylic protons could not be determined accurately due to interfer-
ence with solvent signal. be expected. Thus, with an OH group at the ethyl, an aldehyde at carbon-14 is the only
option to explain both the mass shift and the CO loss.
show that the second hydroxylation has indeed taken place at carbon-14 as the aromatic The photolytic conversion of TAK-715 produces four major and one minor CPs. One of the
signals are equivalent to CP416A, whereas the other hydroxylation is in the ethyl group major products is CP400 (m/z 400.149, C24H22N3OS+), an isomer of TAK-715. Remarkably,
as this modification did not influence the aromatic signals in CP416B either. Furthermore, in MS2 of CP400, the H2O loss, which is dominant in TAK-715 and most of its CPs reported
in CP432A the doublet (3H) and the quartet (1H) for the hydroxylated ethyl group are here, is no longer favoured. As a result, F297 is now the most abundant fragment, rather than
observed at exactly the same shifts as those in CP416B. In the NMR spectrum of CP432A, F279. Instead of F224 and F197, a fragment ion with m/z 254.100 (C16H16NS+) is observed,
integration is possible because the water signal at 4.9  ppm is far less pronounced. In which is due to the HCNO loss from F297. A structure proposal for the isomer is given in
conclusion, CP432A most likely contains a combination of the two hydroxylations from Section 4 of the Supporting Information. In fact, in MS2 of this TAK-715 isomer, generated
CP416A and CP416B although absolute stereochemistry at carbon-2 was not elucidated. by intense light irradiation in solution, was predicted as a gas-phase rearrangement product
The MS data for CP430B (m/z  430.123, C24H20N3O3S+) are consistent with double- (see above). Two other major products are isomers from a dehydrogenation reaction
oxygenation and a dehydrogenation. The major fragment (m/z  308.085, C17H14N3OS+), (CP398A and CP398B; m/z 398.134, C24H20N3OS+). The modification did not occur in the
which is observed next to a H2O loss, probably results from a H2O loss from a fragment with benzamide part, because in MS2 both CPs yield the fragment ions F297-H2 (m/z 295.089,

162 163
C17H15N2OS+) and F279-H2 (m/z 277.079, C17H13N2S+). From the low-abundance fragments CPs of SB203580, the competition between CH3• and OH• loss from an N-oxide CP394D
F264-H2 (m/z 262.055, C16H10N2S+, –6 ppm) and F224-H2 (m/z 222.036, C14H8NS+, –8 ppm) of SB203580, the hydride rearrangement in the water loss from a hydroxylated aromatic
in the MS2 spectrum of CP398B, it may be concluded that CP398B most likely results methyl group (carbon-14) in TAK-715, the influence of the presence of the morpholino
from the formation of six-member aromatic ring by bond formation between carbon-9 and group on double-charge ion generation in BIRB796, and the intermolecular rearrangement
carbon-20. of the fluorobenzyl group in DMPIP. It sometimes turned out to be essential to know the
Although the profile-group approach is less useful for the structure elucidation of TAK- parent daughter relationship between the fragments which was achieved by using an ion
715 CPs than for the other substrates, it still solved cases like CP398B. The possibility trap instead of a quadrupole for fragmentation. The best example is found in CP410C
to be able to propose a structure for the substrate isomer CP400 is a rare case of luck. where SO2CH3• (S-oxidation) was observed as neutral loss instead of OH• and SOCH2
Intriguingly, the same rearrangement as observed in solution during Light incubations is, (unmodified sulfoxide). While known for a methylsulfinyl moiety in SB203580, the
in the gas phase, key to explaining the fragment F297 of TAK-715. The interpretation of occurrence of (unusual) homolytic cleavages and radical losses in the fragmentation
the fragmentation of the TAK-715 CPs included a warning that gas phase chemistry can of the other compound classes is of interest. As such, this study does not only lead to
be very different from solution phase chemistry. Though water loss in solution proceeds structure elucidation of the related substances found, but also extend our understanding of
via proton rearrangement and therefore requires an α–proton, this does not seem to be fragmentation of protonated molecules in MSn.
necessary in the gas phase. In the water loss from the hydroxylated aromatic methyl group
(carbon-14), a hydride rearrangement seems to be equally capable of restoring the charge Acknowledgement
neutrality. The need for an E1 mechanism due to the absence of a base in the gas phase
and the extraordinary stability of water might level the playing field between proton and This research was performed within the framework of project D2-102 “Metabolic stability
hydride rearrangement. The extended conjugated system might additionally enable the assessment as new tool in the Hit-to-Lead selection process and the generation of new
hydride rearrangement in the case of TAK-715 CPs. lead compound libraries” of the Dutch Top Institute Pharma. Vanina Rea, Frans de Kanter,
Jan Commandeur, and Nico Vermeulen are thanked for their involvement in the part of
Occurrence of homolytic cleavages the work related to incubations with BM3 mutants of cytochrome p450s. Jon de Vlieger is
To a large extent, the data comply to the general finding that fragmentation of even-electron acknowledged for his initial involvement in the elucidation of the substrate fragmentation
ions predominantly leads to even-electron fragment ions, which is described as the parity trees.
or even-electron rule [18,29]. However, (unusual) homolytic cleavages of even-electron
ions are observed for SB203580, BIRB796 and TAK-715 as well as for several of their
CPs. For SB203580, homolytic cleavage occurs exclusively in C-S bonds, as is common References
in methylsulfinyl or methylsulfonyl analogues [29,30]. The fragment ABC+• of BIRB796 is [1] J. Zhang, B. Shen, A. Lin, Novel strategies for inhibition of the p38 MAPK pathway. Trends
Pharmacol. Sci. 2007, 28, 286-295.
stabilized by the formation of a semi-quinone structure, but notably is only observed with [2] S. Kumar, J. Boehm, J. C. Lee, p38 MAP kinases: key signalling molecules as therapeutic
the double-charge precursor ion. The second homolytic cleavage from ABC+• to AB+• is targets for inflammatory diseases. Nat. Rev. Drug Discov. 2003, 2, 717-726.
then probably catalyzed by the existing radical. The methyl radical loss in TAK-715 from [3] R. J. Mayer, J. F. Callahan, p38 MAP kinase inhibitors: A future therapy for inflammatory
F279 to F264 and from F224 to F209 is more surprising. It might be speculated that this is diseases. Drug. Discov. Today 2006, 3, 49-54.
4.1 again related to the sulfur in the aromatic system. The only expected homolytic cleavage
in the CPs is the hydroxyl radical loss from the N-oxide CP394D. Although, it is clear
[4] J. Saklatvala, The p38 MAP kinase pathway as a therapeutic target in inflammatory disease.
Curr. Opin. Pharmacol. 2004, 4, 372-377.
4.1
that in DMPIP dehydrogenation induces homolytic cleavage, the uncertainty in locating the [5] D. M. Goldstein, A. Kuglstatter, Y. Lou, M. J. Soth, Selective p38alpha Inhibitors Clinically
modification prevents an explanation. Amongst the TAK-715 CPs, we observe a homolytic Evaluated for the Treatment of Chronic Inflammatory Disorders. J. Med. Chem. 2010, 53, 2345-
cleavage of the hydroxylated ethyl group in CP432A. Interestingly, the hydroxylation of 2353.
the methyl group does not promote its homolytic cleavage. On the contrary, CP416A still [6] S. Miwatashi, Y. Arikawa, E. Kotani, M. Miyamoto, K. Naruo, H. Kimura, T. Tanaka, S. Asahi, S.
Ohkawa, Novel inhibitor of p38 MAP kinase as an anti-TNF-alpha drug: discovery of N-[4-[2-
losses a methyl radical only after eliminating the additional hydroxyl group via dehydration ethyl-4-(3-methylphenyl)-1,3-thiazol-5-yl]-2-pyridyl]benzamide (TAK-715) as a potent and orally
and hydride rearrangement. Surprisingly, CP416B shows the loss of the hydroxylated ethyl active anti-rheumatoid arthritis agent. J. Med. Chem. 2005, 48, 5966-5979.
group by heterolytic cleavage indicating that the methyl hydroxylation is also involved in [7] J. Regan, S. Breitfelder, P. Cirillo, T. Gilmore, A. G. Graham, E. Hickey, B. Klaus, J. Madwed,
mediating the homolytic cleavage of CP432A. M. Moriak, N. Moss, C. Pargellis, S. Pav, A. Proto, A. Swinamer, L. Tong, C. Torcellini, Pyrazole
urea-based inhibitors of p38 MAP kinase: from lead compound to clinical candidate. J. Med.
Conclusion Chem. 2002, 45, 2994-3008.
[8] C. Pargellis, L. Tong, L. Churchill, P. F. Cirillo, T. Gilmore, A. G. Graham, P. M. Grob, E. R.
Hickey, N. Moss, S. Pav, J. Regan, Inhibition of p38 MAP kinase by utilizing a novel allosteric
This paper reports the structure elucidation of a wide variety of related substances of binding site. Nat. Struct. Biol. 2002, 9, 268-272.
four kinase inhibitors, generated in different ways. Comparison between these compound [9] A. Cuenda, J. Rouse, Y. N. Doza, R. Meier, P. Cohen, T. F. Gallagher, P. R. Young, J. C. Lee,
conversion tools and the effect of conversion on bioactivity is reported elsewhere [15]. This SB 203580 is a specific inhibitor of a MAP kinase homologue which is stimulated by cellular
discussion may serve as an example to demonstrate the power of MSn in combination with stresses and interleukin-1. FEBS Lett. 1995, 364, 229-233.
HRMS in structure elucidation of related substances, but can evenly well be used, if one [10] J. S. B. de Vlieger, A. J. Kolkman, K. A. Ampt, J. N. M. Commandeur, N. P. E. Vermeulen, J.
wants, to highlight the limitations of such strategy. Although the profile-group approach [16, Kool, S. S. Wijmenga, W. M. A. Niessen, H. Irth, M. Honing, Determination and identification
17] was applied as much as possible, each substrate required some adaptations in the of estrogenic compounds generated with biosynthetic enzymes using hyphenated screening
elucidation strategy. Especially, the frequently observed deviations in the fragmentation assays, high resolution mass spectrometry and off-line NMR. J. Chromatogr. B Biomed. Sci.
behavior of the CPs compared to the parent compound lead to challenges in interpretation. Appl. 2010, 878, 667-674.
In this respect, we may highlight the competition between H2C=O and SO2CH3 losses in [11] J. Kool, M. Giera, H. Irth, W. M. A. Niessen, Advances in mass spectrometry-based post-column

164 165
bioaffinity profiling of mixtures. Anal. Bioanal. Chem. 2011, 399, 2655-2668.
[12] D. Falck, J. S. B. de Vlieger, W. M. A. Niessen, J. Kool, M. Honing, M. Giera, H. Irth, Development
Supporting Information
of an online p38alpha mitogen-activated protein kinase binding assay and integration of LC–HR-
MS. Anal. Bioanal. Chem. 2010, 398, 1771-1780. [Chapter 2.2] Section 1: Overview chromatographic conditions
[13] D. Falck, J. S. B. de Vlieger, M. Giera, M. Honing, H. Irth, W. M. A. Niessen, J. Kool, On- Next to the Xbridge C18 column 100×2.1 mm with 3.5 µm particles (Waters, Milford, MA,
line electrochemistry-bioaffinity screening with parallel HR-LC-MS for the generation and USA), a Symmetry column (same dimensions and manufacturer) was used. The former
characterization of modified p38alpha kinase inhibitors. Anal. Bioanal. Chem. 2012, 403, 367- was employed in the measurements of samples from EC, BM3 and HLM (TAK-715 only)
375. [Chapter 3.1] experiments, while the latter was used for samples from H2O2, Light and HLM (DMPIP,
[14] V. Rea, D. Falck, J. Kool, F. J. J. de Kanter, J. N. M. Commandeur, N. P. E. Vermeulen, W. M. SB203580 and BIRB796) experiments. Additionally, the gradients differ, mainly because
A. Niessen, M. Honing, Combination of biotransformation by P450 BM3 mutants with on-line
they originate from several independent studies, and because separation on the generic
post-column bioaffinity and mass spectrometric profiling as a novel strategy to diversify and
gradient was generally not sufficient for the analysis of most samples. Flow rate and tem-
characterize p38α kinase inhibitors. Med. Chem. Comm. 2013, 4, 371-377. [Chapter 3.2]
[15] D. Falck, F. Rahimi Pirkolachachi, M. Giera, M. Honing, J. Kool, W. M. A. Niessen, Comparison perature were the same in all cases.
of (bio-)transformation methods for the generation of metabolite-like compound libraries of p38α
Table S1: Binary gradients.
MAP kinase inhibitors using high-resolution screening. J. Pharm. Biomed. Anal. 2013,
http://dx.doi.org/10.1016/j.jpba.2013.08.045 [Chapter 3.3] Time %B Time %B Time %B Time %B Time %B Time %B
[16] E. H. Kerns, K. J. Volk, S. E. Hill, M. S. Lee, Profiling New Taxanes Using LC/MS and LC/MS/
MS Substructural Analysis Techniques. Rapid Commun. Mass Spectrom. 1995, 9, 1539-1545. 0 20 0 40 0 20 0 20 0 20 0 20
[17] E. H. Kerns, R. A. Rourick, K. J. Volk, M. S. Lee, Buspirone metabolite structure profile using a 2 20 2 40 2 20 2 20 2 20 2 20
standard liquid chromatographic-mass spectrometric protocol. J. Chromatogr. B Biomed. Sci. 18 90 45 90 10 90 8 50 20 50 20 50
Appl. 1997, 698, 133-145.
[18] M. Holcapek, L. Kolarova, M. Nobilis, High-performance liquid chromatography-tandem mass 22 90 52 90 13 90 10 50 21 50 21 50
spectrometry in the identification and determination of phase I and phase II drug metabolites. 23 20 53 40 14 20 26 90 26 90 26 90
Anal. Bioanal. Chem. 2008, 391, 59-78. 30 20 60 40 20 20 28 90 33 90 33 90
[19] S. Crotti, L. Stella, I. Munari, F. Massaccesi, L. Cotarca, M. Forcato, P. Traldi, Claisen 29 20 34 20 34 20
rearrangement induced by low-energy collision of ESI-generated, protonated benzyloxy indoles.
35 20 40 20 40 20
J. Mass Spectrom. 2007, 42, 1562-1568.
[20] X. Z. Qin, Tandem mass spectrum of a farnesyl transferase inhibitor--gas-phase rearrangements Used EC, BM3 BM3 H2O2 (DMPIP) Light (DMPIP, H2O2 Light
involving imidazole. J. Mass Spectrom. 2001, 36, 911-917. for: (DMPIP) (TAK-715) and SB203580) (TAK-715)
(SB203580)
[21] P. Henklova, R. Vrzal, B. Papouskova, P. Bednar, P. Jancova, E. Anzenbacherova, J. Ulrichova, and HLM HLM (TAK-
P. Maurel, P. Pavek, Z. Dvorak, SB203580, a pharmacological inhibitor of p38 MAP kinase (DMPIP, 715)
transduction pathway activates ERK and JNK MAP kinases in primary cultures of human SB203580,
hepatocytes. Eur. J. Pharmacol. 2008, 593, 16-23. BIRB796)
[22] W. Danikiewicz, Electron ionization-induced fragmentation of N-alkyl-o-nitroanilines: observation
4.1 of new types of ortho-effects. Eur. Mass Spectrom. 1998, 4, 167-179.
[23] N. J. Bunce, H. S. Mckinnon, R. J. Schnurr, S. R. Keum, E. Buncel, Fragmentation Pathways in
4.1
the Mass-Spectra of Isomeric Phenylazoxypyridine-N-Oxides. Can. J. Chem. 1992, 70, 1028-
1032.
[24] R. Ramanathan, A. D. Su, N. Alvarez, N. Blumenkrantz, S. K. Chowdhury, K. Alton, J.
Patrick, Liquid chromatography/mass spectrometry methods for distinguishing N-oxides from
hydroxylated compounds. Anal. Chem. 2000, 72, 1352-1359.
[25] A. T. Aberg, H. Lofgren, U. Bondesson, M. Hedeland, Structural elucidation of N-oxidized
clemastine metabolites by liquid chromatography/tandem mass spectrometry and the use of
Cunninghamella elegans to facilitate drug metabolite identification. Rapid Commun. Mass
Spectrom. 2010, 24, 1447-1456.
[26] H. Cerecetto, M. Gonzalez, G. Seoane, C. Stanko, O. E. Piro, E. E. Castellano, Mass spectrometry
of 1,2,5-oxadiazole N-oxide derivatives. Use of deuterated analogues in fragmentation pattern
studies. J. Braz. Chem. Soc. 2004, 15, 232-240.
[27] W. M. A. Niessen, Group-specific fragmentation of pesticides and related compounds in liquid
chromatography-tandem mass spectrometry. J. Chromatogr. A 2010, 1217, 4061-4070.
[28] M. Karni, A. Mandelbaum, The ‘Even-Electron Rule’. Org. Mass Spectrom. 1980, 15, 53-64.
[29] E. M. Thurman, I. Ferrer, O. J. Pozo, J. V. Sancho, F. Hernandez, The even-electron rule in
electrospray mass spectra of pesticides. Rapid Commun. Mass Spectrom. 2007, 21, 3855-
3868.
[30] W. M. A. Niessen, Fragmentation of toxicologically relevant drugs in positive-ion liquid
chromatography-tandem mass spectrometry. Mass Spectrom. Rev. 2011, 30, 626-663.

166 167
Section 2: Additional information on BIRB796 MS3 fragmentation (b)

Table S2: Further MS3 fragmentation of the various fragment ions of BIRB796.
Precursor in MS3 Fragment ions in MS3 Identification/Comments
m/z 414 (ABC+•) m/z 256.144 AB+CO
m/z 230.165 & 229.157 AB & AB+•
m/z 174.102 A
m/z 299 (CD+CO) m/z 212.069 Loss of C4H9NO
m/z 114.093 D
m/z 273 (CD) m/z 186.091 Loss of C4H9NO
m/z 100.077 C5H10NO+ (part of D)
m/z 256 (AB+CO) m/z 200.081 A+CO (Loss of B)
m/z 173.072 A+CO – HCN
m/z 157.076 A+CO – HNCO
m/z 133.076 C 8H 9N 2+
m/z 230 (AB) m/z 174.103 A (Loss of B)
m/z 147.093 A – HCN
m/z 133.076 C 8H 9N 2+
m/z 229 (AB+•) m/z 214.132 Loss of CH3•
m/z 172.087 Loss of B• The fragmentation of CP305 (m/z 305.077, C15H14FN2O2S+) is explained starting from
m/z 145.076 m/z 172 – HCN structure proposal B. MS2 results in major fragments with m/z 290.052 (C14H11FN2O2S+•),
m/z 200 (A+CO) m/z 173.071 Loss of HCN consistent with CH3• loss, and m/z 184.044 (C8H10NO2S+), consistent with an amide group
m/z 157.076 Loss of HNCO at the (methylsulfinyl)benzene. MS3 on the ion with m/z 184.044 results in losses of NH3
m/z 147.092 C9H11N2+
and HN=C=O, that is ions with m/z 167 and 141, respectively, and in loss of CH3• to an
m/z 133.076 C 8H 9N 2+
ion with m/z 169.021 (C7H7NO2S+•). MS3 on the ion with m/z 290.052 surprisingly results
m/z 174 (A) m/z 157.076 Loss of NH3 in fragments with m/z 140 and 123, consistent with protonated fluorobenzamide and the
m/z 147.091 C9H11N2+
m/z 133.076 C 8H 9N 2+
acylium ion due to the consecutive loss of NH3, as well as the ion with m/z 169.021 (loss
m/z 120.082 C8H10N+ of CH3•). The observation of two (protonated) amides, m/z 184 and m/z 140, favors the
structure proposal B as there is no other way to link an oxygen to both carbon-3 and -5.
However, this might also be created from A in the gas phase via an intramolecular nucle-
4.1 ophilic attack of an oxygen lone pair on the imine carbon. 4.1
Section 3: Details on the structure assignment of CP305 Section 4: Structure proposal for TAK-715 isomer and fragmentation
intermediate
Figure S1: (a) Structure proposals for CP305 and (b) interpretation of its fragmentation.
Figure S2: Isomer of TAK-715 observed with Light and proposed as intermediate for TAK-715 CID
fragmentation
H3C

S
N
O

(a)
NH2+
Depicted are two structure proposals for CP305. A involves oxygenation of carbon-3, N

opening of the imidazole ring, and loss of methylpyridine. B has the same modifications
but the atoms of the modified imidazole ring are rearranged. While A can be explained
without any heavy atom rearrangements, B is more consistent with the fragmentation H3C
without gas-phase rearrangements in CID. Of both structures, a 5-ring analogue can be
proposed in which the O-atom and the imine N-atom (A) or the two N-atoms are linked For Sections 5 to 8 please refer to the supplementary material which is available on-line
(B). However, the easy fragmentation makes the open variants more plausible. with the journal version and can be downloaded free-of-charge.

168 169
Chapter 4.2

EC–SPE–stripline-NMR analysis of
reactive products:
A feasibility study

David Falck1, Anna J. Oosthoek - de Vries2, Ard Kolkman2,3,


Henk Lingeman1, Maarten Honing1,3, Sybren S. Wijmenga2,
Arno P.M. Kentgens2 and Wilfried M. A. Niessen1,4
1
AIMMS Division of BioMolecular Analysis, VU University Amsterdam, The Netherlands
2
Institute for Molecules and Materials, Radboud University Nijmegen, The Netherlands
3
DSM Resolve, Urmonderbaan 22, 6160 MD Geleen, The Netherlands
4
hyphen MassSpec, de Wetstraat 8, 2332 XT Leiden, The Netherlands
Abstract decreasing the detection volume in order to achieve high concentrations for mass-limited
samples, because the same number of molecules is more effectively measured in a smaller
Flow-through electrochemical conversion (EC) of drug-like molecules was hyphenated volume [12]. As EC–NMR is somewhat underdeveloped, the implementation of miniatur-
to miniaturized nuclear magnetic resonance spectroscopy (NMR) via on-line solid phase ization and cryo technology in EC–NMR probes has not yet been reported. A relatively new
extraction (SPE). After EC of the prominent p38α mitogen-activated protein kinase inhib- trend in chip-based NMR is the stripline geometry featuring a planar resonator, which is
itor BIRB796 into its reactive products, the SPE step provided preconcentration of the optimised on radio frequency (rf) homogeneity, sensitivity and spectral resolution [13]. Pre-
EC products and solvent exchange for NMR analysis. The acquisition of NMR spectra viously, the stripline-NMR chip has been successfully used to analyse low concentrations
of the mass limited samples was achieved in a stripline probe with a detection volume of of metabolites in human cerebrospinal fluid [14]. Moreover, the microfluidic stripline chip
150 nL offering superior mass sensitivity. This hyphenated EC–SPE–stripline-NMR setup is perfectly suited for in situ measurements, for instance in a flow of analyte directed from
enabled the detection of the reactive products using only minute amounts of substrate. the SPE to the NMR. Therefore, we chose a stripline-NMR setup for the detection of the
Furthermore, the integration of conversion and detection into one flow setup counteracts relatively low sample amount produced by our EC setup.
incorrect assessments caused by the degradation of reactive products. However, apparent Though requiring low absolute amounts of sample, the stripline-NMR setup demands high
interferences of the NMR magnetic field with the EC, leading to a low product yield, so far analyte concentrations. To this end, the intermediate SPE step enables significant sample
demanded relatively long signal averaging. A critical assessment of what is and what is not preconcentration next to providing a favourable solvent exchange. The complete integra-
(yet) possible with this approach is presented, for example in terms of structure elucidation tion into one flow system in the EC–SPE–stripline-NMR platform greatly contributes to
and the estimation of concentrations. Additionally, promising routes for further improvement limiting the time elapsing between generation and analysis, thus preventing degradation.
of EC–SPE–stripline-NMR are discussed. In order to achieve a more realistic assessment of the current state of this EC–SPE–
stripline-NMR technology, we studied EC reactions of the p38α mitogen-activated protein
kinase (p38α) inhibitors BIRB796 and SB203580, which are highly relevant in contempo-
Introduction rary drug discovery [15,16]. BIRB796 is currently studied in clinical phase III for the treat-
ment of rheumatoid arthritis and Crohn’s disease [15]. The EC production of reactive or
Whereas hyphenation of electrochemical conversion (EC) and mass spectrometry (MS)
unstable conversion products of BIRB796 has been reported [11].
has become an established technology, e.g., for studying drug metabolism to simulate
cytochrome P450 biotransformation [1], this is not true for hyphenation of EC and nuclear
magnetic resonance spectroscopy (NMR). Although EC–NMR is not an entirely new idea
[2,3], it has not been extensively studied. To some extent, this may be due to the mutual in-
fluence EC and NMR conditions may have, that is disturbance of the magnetic-field homo-
geneity in NMR and/or of the EC processes by induction of additional currents [4]. Despite
this, EC–NMR has been applied frequently and successfully to the study of processes at
electrode surfaces by solid-state NMR techniques [5]. However, in the context of pharma-
ceutical applications, such as studying of redox pairs in vitamins [6] or structure elucidation
of electrochemical products of drug-like compounds, the liquid state and bulk view is more
desirable. Since the 1970s, a few groups have designed in situ EC–NMR probes where EC
is carried out directly in the field region of the NMR, either in static [4] or continuous-flow
approaches [2]. This allows a direct look at the EC reaction, its products and intermediates
without delay. This approach has several disadvantages and limitations, such as the need
4.2 for a dedicated NMR probe, the need to use the same (inert deuterated) solvent in NMR
and EC, and the possible overlap of analyte peaks with signals from additives like buffers
4.2
and electrolytes. Recently, EC–NMR was reported for the analysis of EC products of ac-
etaminophen, using a conventional 60 µL NMR flow cell under steady state conditions [7].
Though high-quality spectra were achieved with commercially available equipment, the
other disadvantages remain in effect. As demonstrated in hyphenation of liquid chromatog-
raphy (LC) and NMR [8,9], some of these problems can be solved by the application of an
on-line solid phase extraction (SPE) step to decouple EC and NMR.
With respect to the application of EC–NMR in the drug discovery process, where initially
only a few mg of a compound is available, a number of different setups are available [10].
Given the small sample amounts, an amperometric setup with a thin-layer cell is to be
preferred over a coulometric setup. In addition, a flow-through approach is advantageous
for the detection of unstable/reactive products [10,11]. Obviously, limiting the amount of
sample used results in high demands on the NMR sensitivity, which is a weak spot of NMR.
To enhance NMR sensitivity, there is a strong trend towards capillary or chip-based NMR
techniques. While higher sample concentrations are required in capillary NMR to achieve
comparable performance in terms of spectrum quality, the total amount of sample neces- Figure 1: BIRB796 and its initial EC products. Electrochemical O-dealkylation at oxygen 9 results in
sary can be reduced up to 100-fold [8]. In microfluidic NMR, the sensitivity is improved by the three major products shown. If not measured directly (on-line), these products undergo degrada-
tion reactions [11].

172 173
Materials and Methods capillary, a shut-off valve (P-732, Upchurch Scientific) was used to prevent the sample
Chemicals from leaking out of the measurement area during measurements lasting longer than one
Acetonitrile (ACN, LC-MS grade) and formic acid (ULC-MS grade) were delivered by Bio- hour. The homebuilt probe consists of an aluminum cylinder, the top of which is divided into
solve (Valkenswaard, The Netherlands). Water was purified by a Milli-Q academic from Mil- halves in which the chip and the electronics circuit are placed [14].
lipore (Amsterdam, The Netherlands). BIRB796 (N-[3-(tert-butyl)-1-(4-methylphenyl)-1H-
pyrazol-5-yl]-N’-[4-[2-(4-morpholinyl)ethoxy]-1-naphthalenyl]-urea) (structure in Figure  1) Acquisition and processing
and SB203580 (4-[4-(4-fluorophenyl)-2-(4-methylsulfinylphenyl)-1H-imidazol-5-yl]pyridine) The stripline-NMR measurements were performed at room temperature in a VNMRS
were obtained from various sources. All other chemicals were obtained from Sigma-Aldrich 600MHz Varian NMR spectrometer, operated with VNMRJ software (Agilent, St. Clara, CA,
(Schnelldorf, Germany). USA). The 90° pulse length was 1.95 µs at 10 W power. At the start of the experiments, the
shiming of the stripline-NMR probe was once performed on a 70% ethanol solution. Then,
Electrochemical conversion the chip was thoroughly flushed with solvent. With the sample solution in the probe, the
EC was achieved in a ROXY EC system (Antec, Zoeterwoude, The Netherlands) consist- shims were only slightly adjusted. Tuning and matching was performed on the sample in
ing of a µ-PrepCell and a ROXY potentiostat. The µ-PrepCell consists of a three electrode the probe.
setup with a glassy carbon working electrode, a titanium counter electrode and a HyREF™ The elution monitoring experiments by SPE–stripline-NMR (see Figure  3) were carried
(Pd/H2) reference electrode. A spacer thickness of 150  µm was employed resulting in a out by averaging 4 scans for one data point every 15 s. All other NMR experiments were
volume of 12 µL in the cell. The EC reaction mixture was supplied by a syringe pump at performed in stopped-flow mode. The 3 mM BIRB796 standard spectrum (Figure 4B) was
a flow of 5 µL/min, converted at the working electrode at a direct current voltage of 0.7 V, measured in 256 scans with a relaxation delay of 1 s; the acquisition took 9 min. The SPE–
and directed to the SPE. The mixture consisted of 10% aqueous ACN containing 370 µM stripline-NMR spectrum (Figure 4C) was measured by accumulating 1024 scans, taking 51
ammonium formate / 630 µM formic acid (pH 3.5) and 200 µM BIRB796. min. The EC–SPE–stripline-NMR sample (Figure 4D) was measured in an array of 25 times
The EC of BIRB796 was monitored at 254 nm at four different flow rates of 5, 10, 20 and 256 scans with a relaxation delay of 2 s. The scans were summed afterwards. The complete
40 µL/min by LC–UV (LC-20 system, Shimadzu, ‘s Hertogenbosch, the Netherlands) and experiment took 12  hr and 25  min. A total correlated spectroscopy (TOCSY) spectrum
analysed using a calibration curve of the standard (0–400 µM). The gradient, solvents and (Figure 5) was measured acquiring 128 t1 increments of 16 scans with a relaxation time
stationary phase used were the same as published earlier [11]. of 2 s. A mixing time of 50 ms with MLEV-17 spinlock was used [17]. The experiment took
approximately 2 hr and 45 min. A correlation spectroscopy (COSY) spectrum (Figure S2 in
On-line Solid Phase Extraction the ESM) was measured using 256 t1 increments of 256 scans on the SPE trapped sample
A SecurityGuard 4 × 2.0 mm C18 cartridge (Phenomenex, Utrecht, The Netherlands) was [18]. The experiment took 45 hr.
used. All steps were executed at room temperature at a flow rate of 5 µL/min. The trapping The 1  mM standard of BIRB796 depicted in Figure  4A was measured accumulating
of the standards from the EC reaction mixture was done for 30 min, but the product mixture
of BIRB796 was trapped for 60 min in order to achieve comparable concentrations for the
competing quinoneimine and hydroquinone (See Figure  1). After a wash step with 99%
D2O and 1% ACN-d3 for 10 min, the analytes were eluted with DMSO-d6 for 11 min. Finally,
the wash step was repeated for re-equilibration. Additionally, a 200 µM 10% aqueous ACN
solution of SB203580 containing 2% DMSO (due to solubility issues with SB203580) and
1 mM ammonium bicarbonate (titrated to pH 10 with sodium hydroxide) was trapped and
eluted, but not converted, using the same protocol.
4.2 Stripline chip, probe and microfluidics
4.2
The stripline structured rf coil consists of a stack of glass or fused silica substrates with
etched microfluidic channels (see Figure S1 of the electronic supplementary material, ESM)
[14]. The copper structures, two ground planes and the rf coil resonator, are sputtered and
electroplated. A constriction in the middle of the rf detector locally enhances the current
density, which results in a high rf field at the position of detection. The rf field is concentrated
and rf homogeneity is enhanced by the two copper ground planes above and below the
stripline. The layers are assembled and merged together with a bounding process so that
they are leakage-free and stable. In the fused silica substrates, two microfluidic channels
are etched, with a volume of 150 nl each, which may contain the sample; only one channels
is used here. Fused silica capillaries (75  µm ID, 150  µm OD, ~15 cm long, Polymicro
Technologies, Phoenix, AZ, USA) are glued into the top and bottom of the chip into the
microfluidic channels to function as inlet and outlet. Via nanotight PEEK unions (P-779,
Upchurch Scientific, Oak Harbor, WA, USA), both fused silica capillaries were connected
to larger capillaries (150 µm ID, 360 µm OD at the inlet side and 250 µm ID, 360 µm OD
at the outlet side) at the base of the magnet in order to protect the μ-PrepCell by avoiding
a pressure buildup in the system higher than 10 bar. The inlet capillary was connected to
the six-port switching valve (made in house) following the SPE unit. At the end of the outlet Figure 2: EC–SPE–stripline-NMR setup. The void volume of all connecting capillaries is specified.

174 175
618 scans with a relaxation delay of 1 s. The product mixture shown in Figure 6B and 6C, rationalized by comparing the trapped analyte weight of 32 µg with the estimated cartridge
before and after storage at room temperature, was measured with a relaxation delay of loading capacity of ~1 mg.
2.3 s using 130 and 160 scans, respectively. The COSY of the product mixture (data not
shown) is measured obtaining 256 t1 increments of 8 scans with a relaxation delay of 2 s. SPE–stripline-NMR
More details on the off-line EC experiments measured by conventional NMR can be found This paragraph deals with the optimization of the flow system and especially the hyphen-
in Section 3 of the ESM. ation of the SPE and the stripline-NMR step. Although the experiments were carried out in
Data processing was done with MatNMR [19]. A zero filling up to 32000 points and a line the complete EC–SPE–stripline-NMR setup, EC was not yet applied. Contrary to off-line
broadening of 1 Hz were used for noise reduction. An asymmetric least squares function SPE, where the sample is obtained as a homogeneous solution in a predefined volume, the
was employed for baseline correction [20]. For peak-picking a Lorentzian to Gaussian elution of the analytes in on-line SPE is comparable to an LC experiment without retention.
resolution enhancement (exp -0.7, Gauss -1.5 Hz) was applied. The 2D spectra were Therefore, the determination of the void time equivalent to the elution time of the analytes
plotted using MatNMR and ACD/NMR Processor (Advanced Chemistry Development Inc, is critical to the success of the stopped-flow stripline-NMR measurements. With the void
Toronto, ON, Canada). time known, the flow can be stopped when the top of the elution peak is in the stripline-
NMR channel. Obviously, an accurate and repeatable elution is one major determinant of
EC–SPE–stripline-NMR setup the sensitivity of the system. The 1H signals of the tert-butyl group of BIRB796 and of the
A schematic diagram of the setup is given in Figure 2. The three syringe pumps (Model methyl group of SB203580 allowed us to follow the elution of both compounds by stripline-
11 Plus Advanced, Harvard Apparatus, Holliston, MA, US), which were placed at ca. 1 m NMR in the final EC–SPE–stripline-NMR setup, albeit with the voltage switched off. The
distance to the magnet, were linked to a four-way connector (VICI, Schenkon, Switzerland) reconstructed SPE–stripline-NMR chromatograms and their Gaussian fits are shown in
and delivered in turn the EC reaction mixture, the wash solution and DMSO-d6. The EC Figure  3. The effective elution time, including the time to reach the stripline chip, was
reaction mixture passed the µ-PrepCell before arriving at the connector. The liquid from the around 11 min for both compounds, its difference of 0.7 min being lower than the accuracy
four-way connector was guided to the SPE cartridge, which was connected to a six-port of the Gaussian fits (≤0.9 min). The elution time is expected to be the same for all com-
switching valve. During the trapping and half of the wash step, the SPE effluent was direct- pounds not showing significant retention under these strong eluting conditions. This makes
ed to waste in order to prevent contamination of the NMR probe with buffer salts and pro- the SPE step widely applicable to compounds with medium to low polarity. The full width
tonated solvents. When the wash step was halfway finished, the valve was switched and at half maximum (FWHM) for BIRB796 and SB203580 is 2.2 min or 11 µL and 1.4 min or
the wash solvent cleared the probe of analyte from the previous measurement. Afterwards, 7  µL, respectively. The resulting concentration factor at half height should therefore be
the new sample was eluted from the SPE cartridge with DMSO-d6 into the stripline-NMR around 15 to 20 fold. On the one hand, this results in a rather inefficient use of the sample,
probe. When the elution was complete, the valve was switched again and both the re-equil- considering that the detection volume is only 150 nL. To some extent, this is due to the low
ibration and the trapping step could be performed during sample measurement. Because flow rate applied, in combination with the large void volume which results from the use of a
the correct timing of the elution is essential and the void volume negatively correlates with 2 mm I.D. SPE cartridge, a conventional six-way switching valve, and the long transfer tub-
repeatability, all connection volumes have been optimized (see Figure 2), within the limits ing allowing the necessary distance between the SPE setup and the stripline-NMR probe.
of the above mentioned pressure restrictions. The comparably large void volumes V1 and On the other hand, it reduces the strain on the repeatability of the elution, because even a
V2 are necessary for bubble-free filling of the µ-PrepCell.

Results and Discussion


The initial step in the EC–SPE–stripline-NMR setup was the EC of the substrate at a
4.2 thin-layer electrode in the flow-through setup. Simultaneously, the products and residual
substrate were trapped on the on-line SPE cartridge. After conversion/trapping, buffer salts
4.2
and protonated solvents were washed away. Finally, substrate and products were eluted
with DMSO-d6 into the stripline-NMR probe. There, 1H NMR as well as COSY and TOCSY
spectra were recorded with excellent mass sensitivity.

Optimization of EC and SPE steps


The EC conditions were transferred from a method published earlier [11] which used the
same EC setup but with a ReactorCell instead of the µ-PrepCell. It was expected that a
higher concentration of substrate could be converted with the µ-PrepCell. Indeed, instead
of the low to medium conversion at 10 µM with the ReactorCell [11], ~90% consumption
of BIRB796 was observed at 200 µM and 5 µL/min with the µ-PrepCell as assessed by
LC–UV. The manufacturer indicates the optimal flow rate of the μ-PrepCell to be between
20 and 50 µL/min. As a decrease in product consumption to ca. 50, 30 and 5% was ob-
served for 10, 20 and 40 µL/min, respectively, the original flow rate of 5 µL/min used in the
ReactorCell was considered optimal. Figure 3: Reconstructed SPE–stripline-NMR chromatograms. Elution of BIRB796 (dots) and
Trapping of the 200 µM standard of BIRB796 was possible for ca. 60 min. If trapping was SB203580 (boxes) from the SPE was monitored in real time by stripline-NMR using 1H signals from a
continued after this period, breakthrough was observed. That indeed breakthrough was the tert-butyl and a methyl group, respectively. The calculated elution time served as basis for the align-
cause of the detected analyte signal and not overloading of the stationary phase can be ment of analyte peak and measurement area.

176 177
deviation of ~1 min leads to only 50% signal reduction, and on the sensitivity of the NMR In order to assess whether the SPE step has an influence on the stripline-NMR, we first
experiment used to follow it, because multiple NMR spectra can be added to create one measured a 3 mM standard of BIRB796 by direct infusion into the stripline-NMR probe. The
data point while following the elution. Both advantages are not to be underestimated when results were compared to a 30 min trapping and elution of a 200 µM standard. The aromatic
using such a highly experimental setup. However, the experimental data suggests that the region of the BIRB796 spectra obtained under different experimental conditions is present-
signal would be strong enough to follow the elution using single scans instead of the four ed in Figure 4 whereas the corresponding chemical shifts and J-couplings are summarized
scans used per data point in these experiments. This would allow increasing the data point in Table S1 of the ESM. All C-H signals are found at the same chemical shifts in both mea-
frequency from 0.07 to 0.33 Hz, thus enabling the detection of peaks with FWHM ≥12 s (7 surements; the maximum deviation is about 0.05 ppm for proton 35. Similar differences are
data points above 10% signal). Thus, the elution volume at half height could in principle be observed between conventional spectra in either DMSO-d6 or ACN-d3 which indicate that
reduced to 1 µL which would result in a ca. 10-fold gain in analyte concentration or equal most likely small differences in residual ACN concentration cause these effects. Additional-
reduction in substrate consumption through improvement of the SPE dimensions. At the ly, the chemical shifts of BIRB796 measured by conventional NMR in DMSO-d6 do not devi-
moment, the substrate consumption of the EC–SPE–stripline-NMR is six times less than ate by more than 0.05 ppm from the stripline-NMR measurements in the same solvent (see
in the conventional NMR. When considering that only one or two percent of the sample Figure 4 and Table S1 of the ESM), except for protons 12, 16 and 17 whose peaks change
was actually measured in the EC–SPE–stripline-NMR, this difference theoretically rises to positions in the spectra between Figure 4A and Figure 4D. This allows comparison to off-
approximately 300-fold. line EC samples measured with conventional NMR. The exchangeable protons attached to
the urea nitrogen atoms (N20 and N23) are no longer observed in the sample measured by
SPE–stripline-NMR which indicates a very efficient exchange with deuterons. By compar-
ing the average signal-to-noise ratios (S/N) of several peaks in the stripline-NMR and in the

4.2 4.2

Figure 4: NMR spectra of the aromatic region of BIRB796. For the numbering, please refer to Fig-
ure 1. Spectra measured A) in conventional NMR (1mM, 618 scans), B) in the stripline-NMR setup
(3  mM, 256  scans), C) in the SPE–stripline-NMR setup (200 µM, 1024  scans), and D) in the full
EC–SPE–stripline-NMR setup (product mixture generated from 200 µM, 25x256 scans). The spectra
displayed are limited to the aromatic region for better visibility. The chemical shifts and J-couplings
are described in Table S1 of the ESM. The product mixture (D) is further described in the next section
and Table S2 of the ESM. Figure 5: TOCSY spectrum of BIRB796 standard in the stripline-NMR
178 179
SPE–stripline-NMR spectrum, a concentration of 2.4 mM (12-fold) could be estimated for cannot be assigned with confidence, because the aliphatic region shows a lot of interfer-
the SPE–stripline-NMR experiment which fits well to the previously calculated 15–20-fold ences and only minor differences with the parent chemical shifts are expected. Having the
concentration at half-height. fingerprint of the hydroquinone and the quinoneimine from the off-line experiment enables
Furthermore, we recorded a COSY spectrum of BIRB796 by SPE–stripline-NMR. All ex- us to identify some of their peaks in the EC–SPE–stripline-NMR spectrum and thereby con-
pected coupling peaks were observed (see Figure S2 in the ESM) in effectively measuring firm their presence. This is shown in Figure 6A which is a zoom of Figure 4D. Comparison
as little as 360  pmol of sample. Unfortunately, an extensive measurement time (45  hr) of the data in Figure 6B and 6C shows that the peaks of the reactive/instable quinoneimine
proved necessary, as an attempt at recording a shorter COSY spectrum on the 3  mM disappear after storage as is expected from earlier studies [11].
standard was not successful. In the stripline-NMR experiment, relatively broad lines are
found to which the long measurement time without lock might contribute. For a COSY, the Conclusions and Perspectives
diagonal peaks are in-phase whereas the cross-peaks have an anti-phase character, which
may lead to vanishing signals in the case of broad peaks. This is in contrast to a TOCSY This study revealed the potential of the hyphenation of EC and miniaturized NMR in the
experiment, in which all peaks show in-phase absorptive lineshapes. It makes the TOCSY proposed way. The EC–SPE–stripline-NMR platform allowed the acquisition of 1H NMR
a more suitable experiment in our case. Therefore, we tried a TOCSY experiment on the and COSY as well as TOCSY proton-proton correlation spectra at substrate concentrations
standard, which took only 2 hr and 45 min and provided a much clearer correlation. As can compatible with a high conversion rate in EC. The spectral resolution and S/N is not as
be seen in Figure 5 and Figure S2 (see ESM), the same 3J-couplings between protons 11 high as in the off-line experiments, but the differences are subtle considering that for the
and 12, 15 and 16, and 17 and 18 are observed in the COSY and in the TOCSY spectrum. EC–SPE–stripline-NMR experiments six times less substrate was consumed. Employing a
test compound which shows simpler NMR spectra, more pronounced spectral differences
EC–SPE–stripline-NMR
After an initial validation of the SPE–stripline-NMR part of the integrated platform, the next
step was the measurement and structural analysis of the EC products of BIRB796. For
assessment of the possibilities and limitations of the platform as well as for validation of the
structural analysis, the on-line spectra are compared to reference spectra obtained off-line
by collection, evaporation and reconstitution with a conventional NMR probe. There is a
significant difference in the overall time consumption of the experiments which stresses the
advantage of the EC–SPE–stripline-NMR over the conventional approach for the detection
of reactive products and is shared by the larger scale flow approach [7]. The preparation
for the conventional NMR measurements often took more than a regular working day (see
ESM Section 3), meaning that for between 8 and 24 hr the sample was subjected to deg-
radation and for at least 7 hr at room temperature. In the EC–SPE–stripline-NMR, the time
delay between generation and analysis was ca. 80 min. Though the NMR measurement it-
self was significantly longer, the products are far less likely to degrade in the elution solvent
than in the reaction solvent, for example by acid-catalysis hydrolysis or dehydration [11].
Figure 6A shows that degradation is not observed in the EC–SPE–stripline-NMR setup.
For BIRB796, three main products are expected from EC–MS experiments [11]. The EC
reaction is an oxidative O-dealkylation at the ether function. It produces 2-morpholinoetha-
4.2 nol, a quinoneimine and a hydroquinone structure (Figure 1). 2-morpholinoethanol was not
observed in the EC–SPE–stripline-NMR spectrum, probably because it is too polar to be 4.2
trapped by reversed-phase SPE. In the EC–SPE–stripline-NMR measurement, mainly the
substrate and weak signals of the two other products are found, indicating low conversion
efficiency in the EC–SPE–stripline-NMR, which conflicts with the high conversion rate mea-
sured by LC–UV. This might be due to interference of the magnetic field with the electrical
currents of the EC. Unfortunately, the low conversion significantly hinders the routine appli-
cation of the current setup for the structure elucidation of the products. Some improvement
in the product S/N, either by increased EC conversion, by an improved SPE method or by
improved stripline-NMR sensitivity, is still needed before the EC–SPE–stripline-NMR can
be used for product structure elucidation independently.
However, if the structure of the products is elucidated with the off-line EC experiments mea-
sured by conventional NMR, the fingerprint can be used to identify the products present Figure 6: Aromatic region of the product mixture. A) The sample measured by EC–SPE–stripline-
in the EC–SPE–stripline-NMR spectrum (see Table S2 in the ESM). Figure 6B shows the NMR is magnified to reveal small product peaks. B) The sample is measure off-line by conventional
peak assignments in the aromatic region after off-line EC. These assignments were sup- NMR directly after re-dissolving and C) again the next day after storage at room temperature to show
ported by a COSY spectrum (data not shown) and analysis of the J-couplings of these sam- the stability of the different products. The substrate peaks are marked with ‘s’, the hydroquinone
marked with ‘h’ and the quinoneimine with ‘q’. The quinoneimine is less stable than the hydroquinone
ples. Some peaks overlap with peaks from solvent constituents in the EC reaction mixture
[11]. After storage, the quinoneimine is not detected any more. This can be used additionally to dis-
which complicates a clear assignment. Next to the substrate, BIRB796, the hydroquinone tinguish the two products. An overview of the NMR signals of the two products in a numerical format
and quinoneimine conversion products are observed. The peaks for 2-morpholinoethanol can be found in Table S2 in the ESM.
180 181
between substrate and product, and a higher conversion rate in EC might have lead to Acknowledgements
even higher quality NMR data, but consequently might have overestimated the usefulness
of this technology at its current state of development in a drug discovery/development This work was performed partly within the framework of the Dutch Top Institute Pharma
setting. The fact that every step of this hyphenated technology as well as the hyphenation project D2-102 (Metabolic stability assessment as new tool in the Hit-to-Lead selection
itself can be improved significantly, either with existing technology or with easily imaginable process and the generation of new lead compound libraries) and partly within the research
technological improvements, promises this technology a future place in the analytical tool- programme ACTS - Process on a Chip (PoaC), financed by the Netherlands Organisa-
box. Even despite these possibilities, the integration and miniaturization already allows the tion for Scientific Research (NWO). Agnieszka Kraj, Hendrik-Jan Brouwer and Jean-Pierre
use of 10-fold lower substrate concentrations at comparable sample volumes than recently Chervet (Antec, Zoeterwoude, The Netherlands) are acknowledged for their support of the
published combinations of EC with conventional flow probe NMR [7]. Additionally, both electrochemistry part of the project. Hans Janssen, Jan van Os and Jan van Bentum (Rad-
flow-based approaches limit the time delay between product generation and analysis, thus boud University Nijmegen, The Netherlands) are credited for technical and organizational
counteracting interferences by degradation. support with regard to set-up and ongoing development of the stripline probe. Additionally,
Possible improvements of the EC–SPE–stripline-NMR platform can be readily indicated. Roald Tiggelaar, Jacob Bart and Han Gardeniers (Twente University/Mesoscale Chemical
The relatively new stripline-NMR technology still awaits integration of recent improvements Systems, Enschede, The Netherlands) are acknowledged for their support of the stripline-
in NMR systems. For example, the addition of a deuterium lock channel would allow lon- NMR chip fabrication. Frans J.J. de Kanter and Andreas W. Ehlers are thanked for their
ger measurement times without compromising the resolution, thus improving sensitivity. input concerning the conventional NMR measurements.
However, interestingly, the maximum measurement time of 12.5 hr (12800 scans) for a 1H
NMR spectrum, applied in this study, did not result in increased line width for the 9H peak
compared with the 512  scan or even the single scan spectrum of the same EC–SPE– References
stripline-NMR sample. Measurements of a 3 mM BIRB796 standard gave a line broadening 1. Jahn S, Karst U (2012) Electrochemistry coupled to (liquid chromatography/) mass spectrome-
try--current state and future perspectives. J Chromatogr A 1259:16-49
of 0.2 Hz from single scan up to 256 scans. At longer measurement times, double lines are 2. Richards JA, Evans DH (1975) Flow Cell for Electrolysis within Probe of a Nuclear Magnet-
observed in some rare cases which may be prevented by use of a lock channel. A further ic-Resonance Spectrometer. Anal Chem 47 (6):964-966
advancement could be achieved by integration of cryotechnology, which has led to an up 3. Albert K, Dreher EL, Straub H, Rieker A (1987) Monitoring Electrochemical Reactions by C-13
to 6-fold increase in sensitivity in other miniaturized probes [21]. This might fill an existing Nmr-Spectroscopy. Magn Reson Chem 25 (10):919-922
gap in quantitative integration which requires an S/N of 150 in order to achieve less than 4. Zhang XC, Zwanziger JW (2011) Design and applications of an in situ electrochemical NMR
1% error [22]. In the EC–SPE–stripline-NMR (12800 scans), the conversion mixture gave cell. J Magn Reson 208 (1):136-147
an S/N of 75 for the doublet of proton 18 of the substrate and of only 5 for the hydroqui- 5. Wu JJ, Day JB, Franaszczuk K, Montez B, Oldfield E, Wieckowski A, Vuissoz PA, Ansermet
none. Thus, the SPE–stripline-NMR combination is only a factor of two short of achieving JP (1997) Recent progress in surface NMR-electrochemistry. J Chem Soc, Faraday Trans 93
sufficient quality 1D spectra from 300 µL sample material at 200 µM concentration. An ad- (6):1017-1026
ditional way to improve sensitivity is found in the stripline-NMR chip. The limit of detection 6. Mairanovsky VG, Yusefovich LY, Filippova TM (1983) Nmr Electrolysis Combined Method
(Nmrel) - Basic Principles and Some Applications. J Magn Reson 54 (1):19-35
(LOD), which is given by the number of spins in 1 Hz bandwidth that need to resonate to 7. Simon H, Melles D, Jacquoilleot S, Sanderson P, Zazzeroni R, Karst U (2012) Combination
achieve an S/N of 1 in a single scan [13], is with 4•1013 [spins/sqrt(Hz)] a factor of three of Electrochemistry and Nuclear Magnetic Resonance Spectroscopy for Metabolism Studies.
higher than the theoretical value. This discrepancy is mainly attributed to losses due to the Anal Chem 84 (20):8777-8782
use of glass as a chip substrate. Therefore, other chip substrates might improve the sen- 8. Gokay O, Albert K (2012) From single to multiple microcoil flow probe NMR and related capil-
sitivity significantly. A fused silica chip is currently under development, and the first tests lary techniques: a review. Anal Bioanal Chem 402 (2):647-669
indicate that lower LOD, thus higher sensitivity, can be expected. Another contribution to 9. Corcoran O, Spraul M (2003) LC-NMR-MS in drug discovery. Drug Discov Today 8 (14):624-

4.2 higher sensitivity may come from an improved resolution of the chip, since the line width
negatively correlates with S/N. We found an FWHM of around 3 Hz, although in a previous 10.
631
Baumann A, Karst U (2010) Online electrochemistry/mass spectrometry in drug metabolism 4.2
stripline-NMR chip we have found an FWHM of less than 1 Hz [23]. Thus, improvements studies: principles and applications. Expert Opin Drug Metab Toxicol 6 (6):715-731
in line width are also anticipated for the newly designed chip. Together with the change 11. Falck D, de Vlieger JSB, Giera M, Honing M, Irth H, Niessen WMA, Kool J (2012) On-line elec-
trochemistry-bioaffinity screening with parallel HR-LC-MS for the generation and characteriza-
in chip substrate, this is expected to improve S/N by at least a factor of five. The main tion of modified p38alpha kinase inhibitors. Anal Bioanal Chem 403 (2):367-375 [Chapter 3.1]
challenge in this study was the unexpectedly low conversion rate. If the conversion rate of 12. van Bentum PJM, Janssen JWG, Kentgens APM, Bart J, Gardeniers JGE (2007) Stripline
60% (long collection, estimated) to 90% (short conversion, measured) of the off-line exper- probes for nuclear magnetic resonance. J Magn Reson 189 (1):104-113
iments would have been achieved in the EC–SPE–stripline-NMR setup, the resulting 4 to 13. Bart J, Janssen JWG, van Bentum PJM, Kentgens APM, Gardeniers JGE (2009) Optimization
6-fold sensitivity increase would be sufficient for acceptable product spectra. of stripline-based microfluidic chips for high-resolution NMR. J Magn Reson 201 (2):175-185
Furthermore, improved tuning of the elution volume of the on-line SPE step to the stripline- 14. Bart J, Kolkman AJ, Oosthoek-de Vries AJ, Koch K, Nieuwland PJ, Janssen JWG, van Bentum
NMR probe, bridging the 50-fold gap between measurement and elution volume, would al- JPM, Ampt KAM, Rutjes FPTJ, Wijmenga SS, Gardeniers JGE, Kentgens APM (2009) A micro-
low increased sensitivity or decreased substrate consumption and trapping time as well. Al- fluidic high-resolution NMR flow probe. J Am Chem Soc 131 (14):5014-5015
though a second alternative would be the use of commercially available 5 µL NMR probes, 15. Eglen RM, Reisine T (2009) The current status of drug discovery against the human kinome.
these do not promise additional mass sensitivity as reported concentrations and measure- Assay Drug Dev Technol 7 (1):22-43
16. Kumar S, Boehm J, Lee JC (2003) p38 MAP kinases: key signalling molecules as therapeutic
ment times are comparable to those reported for the stripline-NMR herein [24]. Therefore, targets for inflammatory diseases. Nat Rev Drug Discov 2 (9):717-726
it is likely more beneficial to bridge this gap by miniaturization of the on-line SPE step. 17. Bax A, Davis DG (1985) MLEV-17-Based Two-Dimensional Homonuclear Magnetization
In conclusion, the proposed improvements provide ample opportunity to close the small Transfer Spectroscop. J Magn Reson 65:355-360
sensitivity gap to achieve a productive hyphenation of EC to miniaturized NMR, provided 18. Aue WP, Karhan J, Ernst RR (1976) Homonuclear broad band decoupling and two-dimension-
the conversion rate in EC can be brought to the off-line level. al J-resolved NMR spectroscopy. J Chem Phys 64 (10):4226

182 183
19. van Beek JD (2007) matNMR: a flexible toolbox for processing, analyzing and visualizing mag-
netic resonance data in Matlab. J Magn Reson 187 (1):19-26
20. Eilers PH (2003) A perfect smoother. Anal Chem 75 (14):3631-3636
21. http:\\www.bruker.com/products/mr/nmr/probes/cryoprobes/microcryoprobe/ overview.html.
Bruker Corporation. Accessed 11.03.2013 2013
22. Malz F, Jancke H (2005) Validation of quantitative NMR. J Pharm Biomed Anal 38 (5):813-823
23. Kentgens APM, Bart J, van Bentum PJM, Brinkmann A, van Eck ERH, Gardeniers JGE,
Janssen JWG, Knijn P, Vasa S, Verkuijlen MHW (2008) High-resolution liquid- and solid-state
nuclear magnetic resonance of nanoliter sample volumes using microcoil detectors. J Chem
Phys 128 (5):052202
24. Jansma A, Chuan T, Albrecht RW, Olson DL, Peck TL, Geierstanger BH (2005) Automated
microflow NMR: routine analysis of five-microliter samples. Anal Chem 77 (19):6509-6515

Supplementary Material

4.2 Figure S2: COSY Spectrum of BIRB796 measured in the SPE–stripline-NMR setup


4.2

Section 3: Off-line EC and conventional NMR experiments


BIRB796 was converted to the product mixture in 6 hr under the conditions described in
‘Electrochemical conversion‘. After 2 and 4 hr, the EC cell was cleaned and refilled as ope-
ration for longer than two hours resulted in significant decrease of conversion efficiency.
The product mixture was dried at 30°C under a nitrogen stream. The dried sample was then
stored at –20°C overnight and reconstituted in DMSO-d6 directly prior to the NMR mea-
surements. These measurements were executed on an Advance 500 NMR spectrometer
using a 5 mm CryoProbeTM (Bruker, Fallanden, Switzerland) at 500.23 MHz.
Figure S1: 400 µm stripline, showing the stripline structure (with cap and ground planes removed)
on top of the probe.

184 185
4.2
Table S1: NMR data on BIRB796. Chemical shifts and J-couplings are compared between the different NMR setups. The corresponding spectra are

186
found in Figure 4.

Proton number 2,6 3,5 7 8 11 12 15 16,17 18 20 23 28 30,34 31,33 35 37,38,39


stripline-NMR
Chem. Shift (ppm) 3.60 * 2.86 4.27 6.96 ** 7.85 7.55 8.19 8.76 8.55 6.34 7.42 7.36 2.45 1.27
multiplet t t br d d m d s s s dd dd s s
8.0, 8.5,
J-coupling 4.1 5.4 8.6 7.8 8.0 8.1
32.7 32.7
SPE–stripline-NMR
Chem. Shift (ppm) 3.61 2.57 2.88 4.28 6.96 ** 7.84 7.56 8.20 - - 6.34 7.41 7.37 2.40 1.27
multiplet br br br br d d m d s dd dd br s s
8.3, 7.1,
J-coupling 8.3 7.1 8.9, 4.0 7.1
25.9 24.7
EC–SPE–stripline-NMR
Chem. Shift (ppm) 3.62 2.55 2.89 4.28 6.96 7.50 7.81 7.56 8.21 8.74 8.53 6.33 7.40 7.36 2.39 1.27
multiplet t s t t d d d m d s br s s dd dd s S
7.4, 8.1, 8.1,
J-coupling 4.1 5.4 4.5 8.2 7.9 7.0 7.6
4.5, 0.4 20.4 20.4
conventional NMR
Chem. Shift (ppm) 3.59 2.55 2.85 4.26 6.96 7.61 7.89 7.57 8.18 8.76 8.56 6.35 7.44 7.36 2.39 1.27
multiplet t br t t t d d d dt d s s s dd dd s s
8.3, 8.2,
J-coupling 4.6 4.5 5.6 5.6 8.4 8.3 8.3 8.2, 6.8 8.2
39.1 39.0
* overlap with DMSO
** overlap of peaks 12, 16 and 17
For multiplets averaged values are taken

Table S2: NMR data on the two EC products of BIRB796. The chemical shift, coupling pattern and J-coupling strength are compared between the EC–
SPE–stripline-NMR setup and the experiments using the conventional NMR.

Proton number 2,6 3,5 7 8 11 12 15 16,17 18 20 23 28 30,34 31,33 35 37,38,39

hydroquinone
EC–SPE–stripline-NMR
Chem. Shift (ppm) x x x x 6.78 * 7.84 * 8.06 ** ** 6.46 *** *** *** 1.37
multiplet d *** d s s
J-coupling 10.6 8.3
off-line, fresh
Chem. Shift (ppm) x x x x 7.24 * 7.95 7.54 8.09 ** ** 6.42 7.35 7.42 2.35 1.37
multiplet d d t d s dd dd s s
J-coupling 8.3 8.6 8.1 8.4 6.4, 36.6 7.2, 37.5
off-line, aged
Chem. Shift (ppm) x x x x 7.24 * 7.95 7.53 8.09 ** ** 6.42 7.43 7.35 2.35 1.37
multiplet d d dt d s dd dd s s
J-coupling 8.2 8.4 1.3, 6.0 8.4 6.7, 36.8 6.7, 36.8

quinoneimine
EC–SPE–stripline-NMR
Chem. Shift (ppm) x x x x 6.83 * 7.76 * 8.14 ** ** 6.36 *** *** *** ***
multiplet d d d s
J-coupling 8.0 8.6 8.3
off-line, fresh
Chem. Shift (ppm) x x x x 6.81 * 7.83 7.49 8.13 ** ** 6.31 7.35 7.44 *** 1.27
multiplet d d dt d s dd dd s
J-coupling 8.1 8.4 8.3, 17.8, 1.3 8.0 13.5 8.1, 45.7 8.2, 45.5
* overlap of peaks 12, 16 and 17
** multiple broad peaks in this region
***overlap with parent compound/solvent peaks
x product does not contain these Hydrogen atoms
187
4.2
Section 5 Chapter 5
High temperature liquid
chromatography hyphenated with
ESI–MS and ICP–MS detection
for the structural characterization
and quantification of halogen
containing drug metabolites

Jon S.B. de Vliegera, Mark J.N. Giezenb, David Falcka, Cornelis Tumpb,
Fred van Heuvelnb, Martin Gieraa, Jeroen Koola, Henk Lingemana,
Jaap Wielingb, Maarten Honingc, Hubertus Irtha, Wilfried M.A. Niessena
a
BioMolecular Analysis Group, Faculty of Sciences, VU University Amsterdam,
De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
b
QPS Netherlands B.V., Petrus Campersingel 123, 9713 AG Groningen, The Netherlands
c
DSM Resolve, PO Box 18, 6160 MD Geleen, The Netherlands
Abstract emental standard are features of ICP-MS that can be used complementary to e.g., ESI-
MS. Cuykens et al. reported the use of ICP-MS with isotope dilution for profiling a drug in
In this paper we describe the hyphenation of high temperature liquid chromatography with development [16] as well as the use of ICP-MS for the detection of glutathione adducts of
ICP-MS and ESI-MS for the characterization of halogen containing drug metabolites. The clozapine based on S and Cl [17]. Wilson et al. reported interesting work on metabolite de-
use of temperature gradients up to 200°C enabled the separation of metabolites with low tection in biological fluids with ICP-MS [18–20]. In this paper, we describe the application of
organic modifier content. This specific property allowed the use of detection methods that HTLC in the identification and quantification of drug metabolites based on ESI-HR-MS and
suffer from (significant) changes in analyte response factors as a function of the organic ICP-MS. Oxidation products of two kinase inhibitors, SB-203580-Iodo and MAPK inhibitor
modifier content such as ICP-MS. Metabolites of two kinase inhibitors (SB-203580-Iodo VIII, were generated with human liver microsomes, bacterial cytochrome P450 BM3 mu-
and MAPK inhibitor VIII) produced by bacterial cytochrome P450 BM3 mutants and hu- tants and electrochemical oxidation.
man liver microsomes were identified based on high resolution MSn data. Quantification
was done using their normalized and elemental specific response in the ICP-MS. The im- Experimental
portance of these kinds of quantification strategies is stressed by the observation that the
difference of the position of one oxygen atom in a structure can greatly affect its response 2.1. Materials
in ESI-MS and UV detection. Human liver microsomes (HLM), pooled from 50 donors were obtained from Xenotech (Lot
no. 0710619) and contained 20 mg mL−1 protein. Drug metabolising bacterial cytochrome
Introduction P450 BM3 mutants M02 [21] and M11 V87A [22] were provided by Dr. J.N.M. Comman-
deur (section Molecular Toxicology, VU University Amsterdam). Acetonitrile (ACN), metha-
In early drug discovery programs, so-called metabolic stability assessment of drug candi- nol (MeOH) both LC-MS grade and formic acid (F) ULC-MS grade were obtained from
dates, it is important to predict the clearance of a drug by cytochrome P450 (and other) Biosolve (Valkenswaard, the Netherlands). Purified water was produced by a Milli-Q device
enzymes in the human liver. Early in vitro metabolic stability screening provides the me- of Millipore (Amsterdam, The Netherlands). Applichem (Lokeren, Belgium) supplied be-
dicinal chemists with information on the sites in the lead structures where metabolism is ta-nicotinamide adenine dinucleotide phosphate (NADPH) tetrasodium salt. Isoclozapine
likely to occur (the so-called metabolic soft spots) [1]. The identification and quantification was obtained from Axon Medchem (Groningen, The Netherlands), Clothiapine from Enzo
of metabolites requires advanced analytical technologies and the development in this area Life Sciences (Raamsdonkveer, the Netherlands) and loxapine from Sigma–Aldrich (Zwi-
is an ongoing process [2]. Recently, we introduced a platform for the simultaneous bio- jndrecht, the Netherlands). SB-203580-Iodo (SB-I) and p38 MAP kinase inhibitor VIII were
activity testing and identification of drug metabolites [3]. Depending on the properties of obtained from Calbiochem (Amsterdam, the Netherlands). Monosodium dihydrogen phos-
the drugs investigated and the bioassay applied, the platform may be adapted to include phate, disodium monohydrogen phosphate, glucose-6-phosphate, glucose-6-phosphate
alternative separation and/or detection technologies. One of the limitations of an on-line dehydrogenase, magnesium chloride, ammonium acetate and dimethylsulfoxide (DMSO)
bioassay is the compatibility with LC mobile-phase composition [4], especially with respect were purchased at Sigma-Aldrich (Schnelldorf, Germany). The structures of all analytes
to the organic modifier content. High-temperature liquid chromatography (HTLC) has been are provided in Supplementary Figure S1.
demonstrated to enable compound separation at constant (low) organic modifier content.
Teutenberg et al. described the fundamentals and practical issues of HTLC in detail [5,6], 2.2. Methods
including the use of low organic modifier concentrations. The benefits of HTLC combined Metabolic incubations with the P450 BM3 mutants had a final volume of 500 µL and con-
with on-line bioassays were readily demonstrated [7]. However, a constant organic modi- sisted of 500 nM enzyme and 100 µM substrate in 100 mM potassium phosphate buffer pH
fier content is beneficial in other instances as well. Several analyte detection strategies 7.4 and were performed at 24°C. Of the substrates, stock solutions were made of 10 mM
suffer from (significant) changes in analyte response factors as a function of the organic in DMSO. The DMSO concentration in the metabolic incubations was always below 2%.
modifier content. This may lead to under- or over-estimated concentrations of drug metabo- An NADPH regenerating system was used to initiate the reactions, resulting in final con-
lites present in the sample. This is certainly true for attractive tools like evaporative light centrations of 0.2 mM NADPH, 0.3 mM glucose-6-phosphate, and 0.4 units mL−1 glucose-
scattering detection (ELSD) [8,9], electrospray ionization mass spectrometry (ESI-MS) [10] 6-phosphate dehydrogenase.
and inductively coupled plasma-mass spectrometry (ICP-MS). In the latter case, a solvent Metabolite generation with HLM was performed at 37 ◦C. The incubations had a final vol-
5 gradient does not only influence the response factor but also the stability of the ICP plasma.
HTLC–ICP-AES was recently reported for the analysis of food ingredients [11]. Smith et
ume of 500 µLand consisted of 1 mg mL−1 protein and 100 µM substrate in 100 mM potas-
sium phosphate buffer pH 7.4 containing 5 mM MgCl2. At time points 0 min and 90 min, 250
5
al. pioneered the integration of HTLC with ICP-MS [12] for the quantification of a glycine µLsamples were taken from the incubation and added to 250 µL of ice-cold ACN, vortexed
conjugate of bromobenzoic acid. for 30 s and centrifuged for 15 min at 6000 rpm. The supernatants were filtered through a
Since its development, ICP-MS is mainly used for quantitative trace element determina- Phenex RC syringe filter 0.2 µm from Phenomenex (Utrecht, the Netherlands).
tion. The main application area of elemental speciation is found in environmental studies. Electrochemical oxidation of SB-I was done by infusing a 10 µM standard in 25% ACN, and
In pharmaceutical applications, the use of ICP-MS started with the profiling of inorganic 1 mM ammonium acetate buffer with pH 5.0 through a Roxy reactor cell equipped with a
impurities and nowadays is expanding to bioanalysis as well, especially involving phar- glassy carbon electrode controlled by a Decade II module and Dialogue software (Antec
macokinetic studies of platinum based cytostatics [13,14]. More recently, ICP-MS has to Leyden, Zoeterwoude, the Netherlands). Products were formed at +1.5 V with a flow-rate
a limited extent been applied in drug metabolism studies of drugs readily detectable by of 10 µL min−1 and collected in an autosampler vial insert.
ICP-MS, especially drugs containing Cl and Br, as reviewed by Gammelgaard et al. [15].
Although fluorine is an even more occurring halogen in drug molecules, low-level detection 2.2.1. HTLC–ESI-MS
of fluorine by ICP-MS is hampered by its relatively high ionization energy. The elemental A Shimadzu LC Ion Trap–Time of Flight (IT-TOF) MS system (Shimadzu, ‘s Hertogenbosch,
specificity, the large linear range, and its ability to quantify with an species-unspecific el- the Netherlands) was equipped with an HT-HPLC 200 oven from SIM GmbH (Oberhausen,

190 191
2.2.2. ICP-MS
An Agilent 7500c ICP-MS equipped with a collision/reaction cell (Agilent Technologies, Am-
stelveen, the Netherlands) was used for the ICP-MS analysis. Tuning of the instrument was
done using direct infusion and pneumatic nebulisation of the analyte solution in the mobile
phase corresponding to its isocratic elution parameters. The plasma parameters for Br and
I detection were as follows: RF power 1600 W, RF Matching 1.72 V, Sample depth 6.0 mm,
Torch horizontal positioning 0.2 mm and vertical 0.6 mm. The carrier gas was 0.82 L min−1
and the makeup gas was set to 0.10 L min−1, both gases were argon with 5.0 purity. For the
detection of SO+, the RF power was 1600 W, the RF matching 1.70 V, sample depth 12.0
Figure 1: Schematic representation of the HTLC-MS setup with, (1) injector and gradient pumps, mm, torch horizontal and vertical position were set at −0.4 mm. The carrier gas was 0.90
(2) solvent preheating compartment, (3) column heating block, (4) post column cooling unit and (5) L min−1 and the makeup gas 0.20 L min−1. The optional gas (O2) was set at 5% and was
ICP-MS or ESI-MS. added to the plasma. The transfer line between the HTLC oven and the inlet of the ICP-MS
was 50 cm of PEEK tubing with an internal diameter of 0.005 in.
Germany). A Waters Acquity UPLC BEH C18 1.7 µm, 2.1×50 mm column was used for
separation. The column was mounted inside the oven with an in house made aluminium
holder for optimal heat transfer (see Figure 1 for schematic representation of the setup). A Results and discussion
stainless steel frit (1 µm) fixed into a Valco union (DaVinci Europe, Rotterdam,The Nether-
lands) was used as an inline filter. The integrated solvent preheating area and post-column Thermal degradation of the standards was not observed under the conditions used. In the
cooling unit consisted of 25 cm and 30 cm stainless steel tubing (internal diameter of 0.3 supporting information (Supplementary Figure S2) the extracted ion chromatograms of
mm). In all HTLC experiments, the solvent preheating area was set 5°C above the starting SB-I (as an example) are shown under increasing temperatures while isocratic elution was
temperature of the gradient while the post column cooling unit was set at a fixed value of performed. The calculated peak areas had a standard deviation of less than 5%, indicating
25°C. that the compound was stable throughout the analysis.
The IT-TOF-MS was equipped with an electrospray source and operated in positive ion
mode. The interface voltage was set at 4.5 kV while the source heating block and curved 3.1. Solvent effect on electrospray ionization
desolvation line (CDL) temperature were at 200°C. Drying gas pressure was set at 62 kPa For the three standards, isoclozapine (C18H19ClN4, [M + H]+ with m/z 327.1371), loxapine
and nebulising gas was applied. MS3 spectra were acquired in data dependent mode with
the following settings: fullspectrum MS from m/z 200–650, MS2 and MS3 acquisition from
m/z 80–650 with ion accumulation of 10 ms and collision energy set at 50%. The first min-
ute of the chromatogram was diverted to waste.
A standard mixture of 100 µM isoclozapine, clothiapine and loxapine in 3% DMSO, 97%
H2O was analysed in isocratic mode, with eluent composition of 95% H2O, 0.2% FA and
5% ACN or 5% MeOH (solvent Aa and Am, respectively). If not stated otherwise, a flow-
rate of 300 µL min−1 was applied. With 5% ACN as eluent, the temperature gradient started
with 0.5 min at 110°C followed by a linear increase up to 175°C in 6 min where it was kept
constant for 2.5 min. With 5% MeOH as eluent, the temperature gradient
started with 0.5 min at 130 ◦C followed by a linear increase up to 175°C in 4 min where it
was kept constant for 2.5 min. The system was ready for the next injection after 12.5 min.
For reference chromatograms, the following solvent gradient was applied: 1 min isocratic
at 5% B linear increase in 11.5 min to 75% B, back to 5% B in 0.5 min where kept constant
5 for 3 min. Solvent Ba and Bm were ACN, 0.2% FA and MeOH, 0.2% FA, respectively. The
column was thermostated at 50°C. 5
The analysis of the SB-I samples was achieved using the following temperature gradient:
5 min at 110°C, then a linear increase to 180°C in 8 min where kept constant for another 5
min. The system was cooled down and after a total runtime of 20 min, it was ready for the
next injection. An isocratic solvent Aa was used with a flow-rate of 600 µL min−1.
MAPK inhibitor VIII samples were analysed with a temperature gradient starting from
120°C for 5 min, followed by a linear increase in 5 min to 200°C where kept constant for 5
min. An isocratic solvent composition of 15% MeOH 0.2% FA was used with a flow-rate of
600 µL min−1.
Thermostability of the standards was assessed by determining the peak areas of the stand-
ards in a set of experiments where isocratic and isothermal elution was performed. With
increasing the temperature up to 200°C, this experiment can indicate possible on-column
degradation. Fig. 2. Comparison of ESI-MS peak areas (arbitrary units) of standards with temperature
and organic solvent gradients.
192 193
(C18H18ClN3O, [M + H]+ with m/z 328.1211) and clothiapine (C18H18ClN3S, [M + H]+ with m/z C19H19ClN4O, corresponding to additional CO in the molecule. Unfortunately, in MS/MS of
344.0983), a comparison was made between the peak area in ESI-MS using either a tem- the impurity, only the loss of H2O is observed. Therefore, it is difficult to make a structure
perature or a solvent gradient. When using a temperature gradient at 5% of organic modi- proposal. Peaks 3, 4, and 5 were identified based on the HR-MS data as isoclozapine,
fier, the peak areas for the standards were affected by the type of organic modifier (MeOH loxapine and clothiapine, respectively.
or ACN) to a very small extend, as can be seen in Figure 2. In the solvent gradient, the peak For bromine with m/z of 79, the detection limit in ICP-MS was 0.5 µM and iodine could be
areas in MeOH were higher than those in ACN (24% for isoclozapine, 17% for loxapine and detected down to 80 nM. For iodine, a linear calibration plot was achieved in the range of
16% for clothiapine), which is somewhat to be expected because ACN is the stronger sol- 80 nM–80 µM (y = 3.96e4 x + 2.17e3 with R2 0.9976), the experimental conditions of the
vent, making the analytes elute at a lower ACN than MeOH concentration in the gradient. HTLC part of this are described above in Section 2.2.1. The detection limits were not as
The solvent effect on the ESI-MS response obviously is a bit of a disadvantage in HTLC, low as reported by others [15], but are sufficiently low for the detection of metabolites and
where only 5% of organic modifier is used. Especially loxapine and clothiapine, eluting at impurities present in the in vitro incubation samples. Moreover, the separation method dif-
higher concentrations of organic modifier in the solvent gradient, show a factor of 1.9 and fers significantly and our injection volumes are a factor of 2–10 smaller.
2.3, decreased ESI-MS peak areas in the temperature gradient (Figure 2), respectively.
The chromatograms of using a temperature gradient and a solvent gradient with ACN can 3.3. Profiling bromine containing MAPK inhibitor VIII products
be found in the supporting information, Supplementary Figure S3. In practical applications, The ESI-MSn fragmentation pattern of MAPK inhibitor VIII ([M + H]+ with m/z 415.0207,
not only the peak area but also the signal-to-noise ratio (S/N) is important. In the total-ion C20H17BrClN2O+) in the IT-TOF was elucidated to facilitate the identification of its metabo-
chromatogram (TIC), there is not much difference in S/N for the three compounds, when lites. Figure 4a shows the ESI-MSn spectra and Figure 4b the proposed fragmentation
comparing the solvent and the temperature gradient. In the extracted-ion chromatograms pathway. MAPK inhibitor VIII contains both a chlorine and a bromine atom; the characteris-
(EIC), the S/N in HTLC mode is slightly lower than the S/N of the solvent gradient. This is tic isotopic pattern is observed in ESI-MS. This feature is useful for the search of structur-
consistent with the difference in ESI-MS response between the two modes as described ally related metabolites in the incubation mixture by e.g., software assisted isotope pattern
above. recognition and isotope filtering strategies [23]. In this case, we used HTLC–ICP-MS to
search for all bromine containing compounds in the metabolic incubations. Subsequently,
3.2. Hyphenation of HTLC to ICP-MS these bromine containing metabolites were identified based on their MSn spectra in the
The detection limits of the ICP-MS for Cl+ (m/z 35), Br+ (m/z 79), S+ (m/z 32) and I+ (m/z ESI-MS measurements.
127) were determined by flow injection in a 5% MeOH or ACN, 0.2% FA solution. Using Two metabolites of MAPK inhibitor VIII were found in the P450 BM3 mutant incubation
optional gas flow, the detection of SO+ (m/z 48, LOD 18 µM) was found to be more sensi- samples when analysed with HTLC–ICP-MS (Figure 5) and LC–ESI-MS. The accurate
tive than that of S+ (227 µM). This is due to the occurrence of a polyatomic interference masses of these metabolites are in accordance with a hydroxylated substrate ([M + H]+
of O2+ (m/z 31.9898) with S+ (m/z 31.9721), both having m/z 32 in the ICP-MS due to its with m/z 431.0156, peak 2 in Figure 5) and a quinone-type structure ([M + H]+ with m/z
limited resolution. Isoclozapine, loxapine and clothiapine were readily separated and de- 444.9949, peak 1 in Figure 5). The fragmentation patterns of these metabolites indicate
tected with the HTLC–ICP-MS method. The chlorine specific detection demonstrated the that the oxidations took place in the toluene substructure, present in MAPK inhibitor VIII.
hyphenation of HTLC to ICP-MS, with an LOD of 128 µM. As can be seen in Figure 3, the The singly oxygenated metabolite (peak 2) could be tentatively identified from a different
chromatogram of m/z 35 of the HTLC–ICP-MS analysis of a 250 µM mixture of clothiapine fragmentation behaviour in MSn compared to the parent drug. It showed a loss of H2O in
(5), iso-clozapine (3) and loxapine (4) resulted in five peaks with an S/N ratio above 3. MS2 and subsequent loss of a bromine radical in MS3, and not the toluene loss observed
Peak number 1 is due to polyatomic interference of 34S + H+ (m/z 35) resulting from the for the parent drug. Therefore, we propose that hydroxylation occurred at the methyl group
high percentage of DMSO in the sample, or to the breakthrough of 35Cl+. (The unit-mass and the water loss involves a six-member ring formation between the bromo-phenyl group
resolution ICP-MS used in this study does not allow discrimination of these two.) Peak 2 is and the chlorine substituted benzene ring. The loss of a halogen radical from an aromatic
due to an impurity in the isoclozapine standard. HR-MS data indicate an [M + H]+ with m/z systems is often observed in ESI-MS/MS experiments [24]. The other metabolite (peak
355.1325 and a monochlorine isotope pattern for this impurity. The molecular formula is 1) shows a mass difference of 29.9731 u, which is consistent with the loss of H2 and the
gain of O2 (29,9742 u), thus C20H15BrClN2O3+. In MS2, this metabolite shows the fragment
ion with m/z 296.9789, indicating the loss of the (modified) toluene ring. The most likely
way to explain the observed changes in the toluene ring is by quinone formation [25]. This
5 5
2000

1800 3 4 5 apparently results in an easier loss of the complete side chain (C8H4O3) with hydrogen re-
1 arrangement rather than the stepwise loss of toluene and CO, as observed for the parent
m/z 35
1600
compound. The HTLC–ICP-MS chromatogram showed an additional bromine containing
ICP-MS response

1400 compound in the sample (peak 3, Figure 5), and was found to be an impurity in the ana-
1200
2
lytical standard (m/z 439.0192, proposed formula C22H17N2OClBr+, 4.8 ppm error). The ac-
quired MS2 data allowed us to propose a structure for this impurity (Figure 5). The formation
1000
of a six membered ring by adding C=C to connect the secondary substituted amine with the
800
primary amine fits the accurate mass and matches the fragmentation spectra. Peak 4 is the
600 substrate, MAPK inhibitor VIII with m/z 415.0207. In the HTLC–ESI-MS chromatograms,
0 2 4 6 8 10
we did not observe additional metabolites or impurities missing the characteristic bromine
Retention time in minutes
and/or chlorine isotope pattern of MAPK inhibitor VIII.
Figure 3: HTLC-ICP-MS chromatogram for chlorine (m/z 35) in a 250 µM standard mix of iso-clozap-
ine, loxapine and clothiapine using 5% ACN 0.2% FA as mobile phase. (1) Polyatomic inference of 34S 3.4. Analysis of iodine containing SB-I and conversion products
+ H; (2) chlorine containing impurity with m/z 355.1325; (3) isoclozapine; (4) loxapine; (5) clothiapine. The major fragment in MS2 of SB-I (C21H17IN3OS+, [M + H]+ with m/z 486.0132) in the IT-

194 195
a
O Cl

Inten.(x1,000,000) 4500 O Br
417.0172
7.5
415.0198 Full MS 1
4000 O N
H

5.0 NH2

4
3500 m/z 79 O Cl
2.5
Br

ICP-MS response in counts


0.0 3000
405.0 410.0 415.0 420.0 425.0 430.0 m/z N
H
Inten.(x100,000) OH NH2
322.9559 2500
2.5 MS2 415.0 O Cl

2000 1 Br

2 3
0.0
250 500 750 m/z N
1500
Inten.(x10,000) 3 NH
259.9933
2.5 294.9576 MS3 322.9 1000 O Cl
181.0755
Br

0.0
250 500 750 m/z
500 4
N
H

NH2
0

b O Cl
0 1 2 3 4 5 6 7 8 9 10

Br Retention time in minutes

N
Figure 5: HTLC-ICP-MS chromatogram (m/z 79, Br+) of the incubation sample having MAPK inhib-
H2+ itor VIII as substrate.
Chemical Formula: C20H17BrClN2O+ NH2
Exact Mass: 415.0207 TOF instrument is the loss of a methyl radical, resulting in the fragment with m/z 470.9894.
Performing MS3 experiments on this radical cation produces a major fragment with m/z
O Cl
-C 7H8 423.0227 corresponding to the loss of SO. In addition, a minor fragment with m/z 344.0843
∆ m/z 92.0626 is observed due to the loss of an iodine radical. The limited fragmentation under these
C+ Br conditions indicates that unambiguous metabolite identification by MSn will not be readily
possible. Incubation of SB-I with the P450 BM3 mutants M02, M11 V87A and with HLM
resulted in the formation of several metabolites (see Figure 6). This figure may be used
N
H to assess the ease at which HTLC is implemented: ESI-MSn and ICP-MS were performed
NH2 -Cl • on different days and in two different laboratories (Amsterdam and Groningen). In order
-CO Chemical Formula: C13H9BrClN2O+ -Br • to achieve this, the HTLC part of the system was dismantled in the first and reinstalled
∆ m/z 27.9949 Exact Mass: 322.9581 -CO in the second laboratory. Nevertheless, highly comparable chromatographic results were
∆ m/z 141.8821 achieved (Figure 6).
Cl
-CO MSn spectra obtained by HTLC–ESI-MSn analysis of the metabolites of SB-I, indicate oxi-
-Cl • dation at the sulphur, oxidation of the dehalogenated SB-I (m/z 376.1144) and two more
5 C+
Br
∆ m/z 62.9637
C+ oxidation products (m/z 502.0088). S-oxidation has also been reported for the fluorine 5
Br substituted analogue SB-203580 [26]. Dehalogenation of the standard (m/z 360.1181) was
C+
N observed in all samples, including the controls and dilutions from a freshly prepared stock
N
H
H
solution.
Chemical Formula: C12H9BrClN2+ NH2
N
H
NH2
Peak 1 in the ICP-MS chromatogram (Figure 6) is observed in the enzyme blank and in
Chemical Formula: C12H9N2+
Exact Mass: 294.9632 Chemical Formula: C12H9BrN2•+ NH2 Exact Mass: 181.0760 the t = 0 control as well, indicating a background signal of the human liver microsomal
Exact Mass: 259.9944 matrix and is therefore not included in Table 1. Peak 2 in the ESI-MS chromatogram with
m/z 360.1181 can be identified as the dehalogenated SB-I and is present as an impurity in
all samples containing SB-I. Increased ESI-MS peak areas in the active incubations com-
Figure 4: a) Full HR-ESI-MS, MS/MS and MS3 spectra on MAPK inhibitor VIII. b) Proposed fragmen- pared to control experiments indicate that dehalogenation is catalysed by the P450s. Since
tation pattern and fragment structures of MAPK inhibitor VIII.
iodine is no longer present in the structure, this compound is not detected in the ICP-MS
analysis. The presence of sulphur provides an additional feature to quantify these metabo-
lites, although we were able to detect S+ and SO+, these detection limits were not sufficient-
ly low enough to be used for quantification of these low abundant metabolites. Peak 3 is a
196 197
metabolite present in all active enzymatic incubations but the P450 BM3 mutants produce
significantly more of this oxidized dehalogenated metabolite (m/z 376.1136, see Table 1).
The loss of a CH3SO2 radical indicates oxidation of the sulphur. There are two possible
explanations for the formation of this metabolite, either oxidation of the dehalogenated
impurity or the reductive dehalogenation of the main metabolite, the sulphonated SB-I.
More interesting is peak 4, present in both ICP-MS and ESI-MS traces of the HLM incuba-
tion. This indicates the presence of iodine in the structure. The m/z value is in accordance
with hydroxylation of the parent compound (m/z 502.0088). This metabolite is only found
in the HLM incubations and not in the mutant ones. Peak 5 is the parent compound with
m/z 486.0132. Based on the ESI-MS data, the main metabolite was found to be oxidized
at the sulphur into a sulphone, peak number 6, as is indicated by the subsequent losses of
a CH3 radical and SO2 in two stages of MSn. This metabolite was found in all incubations,
but at different concentrations (Table 1). In incubations with HLM and with the mutant M11
V87A only, an additional oxidated product with m/z 502.0088 was found (peak 7). Given the
fact that the retention time for this product is higher than that of the parent, the compound
might be an N-oxide [27]. With the formation of this specific compound, the M11 V87A BM3
mutant mimics the in vitro human metabolism the most.

3.5. Response comparison of ICP-MS and ESI-MS


Table 1 not only provides information on the concentrations of metabolites, based on the
response in ICP-MS, but also enables a comparison of the response factor in ESI-MS and
ICP-MS. For example, the metabolite peaks 6 and 7 have the same chemical formula and
the oxygen incorporated by metabolism results in only a relatively small difference in the
Figure 6: Comparison of HTLC-ICP-MS chromatogram with m/z 127 (lower trace) and HTLC–ESI- chemical structure. However, whereas the concentrations of these metabolites are very
MS chromatogram with extracted ions of SB-I and HLM related products. (m/z 502.0088 5× similar (0.75 µM peak 6 and 0.72 µM for peak 7), the response in ESI-MS differs by a fac-
magnified). See Table 1 for peak areas and peak identification. tor of ~25. (25.5×106 for peak 6 and only 1.0×106 for peak 7). The ICP-MS is a normalized
detector for specific elements and therefore not sensitive to the difference in structure of
these two metabolites. Additionally, the results of comparing the same calibration samples
measured on both instruments show the difference in linearity, especially with the higher
Table 1: Formation of SB-203580-Iodo related products by different enzymatic systems including
ICP-MS and ESI-MS data. For chromatograms of the HLM incubation see Figure 6. concentrations of SB-I measured. Calibration curves were measured from 0 to 80 µM of
SB-I. The LOD for SB-I with HTLC–ICP-MS was 80 nM while with ESI-MS the LOD was as
Peak Rt Enzyme ICP-MS ESI-MS low as 10 nM. The linear range of the ICP-MS was found to be from 80 nM up to 80 µM,
nr (min) while in HTLC–ESI-MS linearity was observed from 20 nM to 10 µM. This demonstrates
the potential of over- and/or underestimating the concentration of a metabolite based on
Area Conc. in µM Area [M+H]+ Molecular its response in a regular LC–ESI-MS run. In this case, the small structural changes in the
formula
molecule and the limited linear response in ESI-MS require complementary normalized
2 1.4 M02 – – 15693726 360.1181 C21H17N3OS detection methods to provide an accurate assessment of the concentrations present in the
M11 V87A – – 14731403 sample.
HLM – – 11823036 Next to incubations with enzymes, we apply electrochemical oxidation to generate oxida-
3 1.8 M02 – – 4046358 376.1136 C21H17N3O2S tion products of drugs. In some cases, several human relevant metabolites can be pro-
5 M11 V87A
HLM




3843774
582953
duced in this way [28]. This is a relatively clean method to produce oxidation products and
therefore may support metabolite identification studies. We applied electrochemical oxida-
5
4 3.2 M02 – – – 502.0088 C21H16IN3O2S tion to the iodine containing SB-I and use this additional sample to illustrate the strength of
M11 V87A – – – hyphenating HTLC to ICP-MS. The electrochemical oxidation products were analysed in
HLM 3999 <0.08 (0.05) 437422
both ICP-MS and ESI-MS configuration. The total compound concentration in this sample
5 7.7 M02 917471 23.1 113167098 486.0132 C21H16IN3OS was 5–10 times lower than in the enzymatic incubations since the optimum electrochemical
M11 V87A 876649 22.1 104141596
conversion conditions were found to include a substrate concentration of only 10 µM (data
HLM 746022 18.8 82157923
will be published elsewhere). Figure 7 shows the ICP-MS trace of iodine and the UV chro-
6 8.4 M02 31814 0.75 25544346 502.0086 C21H16IN3O2S
matogram at 254 nm. Unfortunately, in ESI-MS only the substrate was found, indicating
M11 V87A 20698 0.47 15789179
HLM 17126 0.38 8225095 that the concentrations of the formed products were below their ESI-MS detection limits.
The latter was verified using a conventional solvent gradient analysis. Nevertheless, the
7 11.0 M02 – – – 502.0086 C21H16IN3O2S
M11 V87A 27128 0.63 998657
concentrations of the products could be determined by the HTLC–ICP-MS analysis (Table
HLM 30589 0.72 1044683 2). The electrochemical oxidation process converted more than 60% of the initial SB-I into
six iodine containing products. The major product was formed at a concentration of 4 µM,

198 199
Conclusions
The combined use of temperature gradients and isocratic solvent compositions enabled
the profiling and quantification of several oxidation products of SB-I and MAPK inhibitor
VIII by means of ICP-MS. Information on structures of the metabolites was obtained by
ESI-HR-MSn experiments. The detection of drug metabolites by normalized and selective
detection of a halogen using ICP-MS provides valuable and complementary information
on the analytes. Whenever the halogen remains incorporated within the metabolites, they
are detected, can be quantified, and even mass balancing is possible. However, if oxida-
tive dehalogenation occurs, mass balancing approaches become more difficult, especially
when more than one metabolite looses the halogen from its structure. Nevertheless, the
hyphenation of HTLC to structure independent detection methods such as ICP-MS and
ELSD can contribute to the toolbox of researchers for the quantification of drug metabolites
and other drug related molecules, e.g., process impurities, degradation products. The im-
portance of these kinds of quantification strategies is stressed by the observation that the
difference of the position of one oxygen atom in a structure can greatly affect its response
in ESI-MS and UV detection. Whether the incorporation of this atom is by P450 metabo-
lism, electrochemical oxidation, photodegradation or any other process is irrelevant for the
strategy presented in this paper.

Figure 7: Overlay of HTLC-ICP-MS (solid upper trace) and HTLC-UV (dashed lower trace) chromato-
Acknowledgements
grams of the electrochemical oxidation products of SB-I.
This research was performed within the framework of project D2-102 ‘Metabolic stabil-
while other products were formed in concentrations of 1 µM or lower. ity assessment’ of the Dutch Top Institute Pharma. The authors thank Andre Pantophlet,
In this particular case, the total concentration of all products and the parent drug was found Dick van Iperen, Klaas van Altena and Roald Boegschoten for their technical assistance.
to be the same as the total concentration of the parent drug before oxidation. This means Dr. Jelle Reinen of the VU University section of Molecular Toxicology is thanked for the
that no dehalogenation occurred in the electrochemical oxidation and that all products expression of the BM3 enzymes. Antec Leyden is acknowledged for the loan of the Roxy
detected contained iodine. If this would be the case with in vitro metabolism studies, mass EC system.
balancing can be done which significantly contributes to the elucidation of the drug metabo-
lism and to the metabolic and pharmacokinetic profiling. Moreover, the data in Table 2 and
Figure 7 also indicate that not only the ESI-MS response but also the molar extinction coef-
References
1. A.P. Watt, R.J. Mortishire-Smith, U. Gerhard, S.R. Thomas, Curr. Opin. Drug Discovery Dev. 6
ficient may significantly change upon oxidation. For instance, peaks 4 and 5 show similar (2003) 57.
response in ICP-MS, whereas their response in UV is significantly different. Therefore, in 2. A.N.R. Nedderman, Biopharm. Drug Dispos. 30 (2009) 153. [3] J.S.B. de Vlieger, A.J. Kolk-
this specific case, the assumption that conversion products have the same UV extinction man, K.A.M. Ampt, J.N.M. Commandeur, N.P.E. Vermeulen, J. Kool, S.S. Wijmenga, W.M.A.
coefficient cannot be made since it would significantly underestimate the concentrations of Niessen, H. Irth, M. Honing, J. Chromatogr. B 878 (2010) 667.
products formed by oxidation. 4. J. Kool, M. Giera, H. Irth, W. Niessen, Anal. Bioanal. Chem. (2011) 1.
5. T. Teutenberg, Anal. Chim. Acta 643 (2009) 1.
6. T. Teutenberg, H.J. Goetze, J. Tuerk, J. Ploeger, T.K. Kiffmeyer, K.G. Schmidt, W.G. Kohorst,
T. Rohe, H.D. Jansen, H. Weber, J. Chromatogr. A 1114 (2006) 89.
Table 2: Retention time, peak area and determined concentration by ICP-MS of electrochemical 7. A.R. de Boer, J.M. Alcaide-Hidalgo, J.G. Krabbe, J. Kolkman, C.N.V. Boas, W.M.A. Niessen,
5 oxidation products of SB-I from Figure 7.
8.
H. Lingeman, H. Irth, Anal. Chem. 77 (2005) 7894.
D. Guillarme, S. Rudaz, C. Schelling, M. Dreux, J.L. Veuthey, J. Chromatogr. A 1192 (2008)
5
Peak Peak retention area (counts) concentration µM 103.
time(min) 9. D. Guillarme, S. Heinisch, J.Y. Gauvrit, P. Lanteri, J.L. Rocca, J. Chromatogr. A 1078 (2005)
22.
1 0.5 2311 <0.08 (0.003) 10. J. Wang, A. Aubry, M.S. Bolgar, H. Gu, T.V. Olah, M. Arnold, M. Jemal, Rapid Commun. Mass
2 0.7 17516 0.39 Spectrom. 24 (2010) 3221.
3 1.8 9953 0.20 11. A. Terol, E. Paredes, S.E. Maestre, S. Prats, J.L. Todoli, J. Chromatogr. A 1217 (2010) 6195.
4 5.1 160583 4.00 12. C.J. Smith, S. Shillingford, A.M. Edge, C. Bailey, I.D. Wilson, Chromatographia 67 (2008) 673.
13. E.E.M. Brouwers, M. Tibben, H. Rosing, J.H.M. Schellens, J.H. Beijnen, Mass Spectrom. Rev.
5 7.7(SB-I) 151373 3.76
27 (2008) 67.
6 7.9 29748 0.70 14. M. Breda, M. Maffini, A. Mangia, C. Mucchino, M. Musci, J. Pharm. Biomed. Anal. 48 (2008)
7 11.7 44491 1.07 435.
Total 415975 10.1 15. B. Gammelgaard, H.R. Hansen, S. Sturup, C. Moller, Exp. Opin. Drug Metab. Toxicol. 4 (2008)
1187.

200 201
16. F. Cuyckens, L.I.L. Balcaen, K. De Wolf, B. De Samber, C. Van Looveren, R. Hurkmans, F.
Vanhaecke, Anal. Bioanal. Chem. 390 (2008) 1717.
17. K. De Wolf, L. Balcaen, E. Van De Walle, F. Cuyckens, F. Vanhaecke, J. Anal. At. Spectrom. 25
(2010) 419.
18. O. Corcoran, J.K. Nicholson, E.M. Lenz, F. Abou-Shakra, J. Castro-Perez, A.B. Sage, I.D. (x10,000,000)
Wilson, Rapid Commun. Mass Spectrom. 14 (2000) 2377. 200°C Figure S2: Temperature Stability of SB-203580 -Iodo
19. C.J. Duckett, I.D. Wilson, H. Walker, F. Abou-Shakra, J.C. Lindon, J.K. Nicholson, Rapid Com- 2.5 Temperature in °C peak area
175°C 200 4.06E+08
mun. Mass Spectrom. 17 (2003) 1855. 2.0 175 4.31E+08
20. B.P. Jensen, C.J. Smith, C.J. Bailey, C. Rodgers, I.D. Wilson, J.K. Nicholson, Rapid Commun. 150 4.44E+08
150°C
Mass Spectrom. 19 (2005) 519. 1.5
125 4.29E+08
21. M.C. Damsten, B.M.A. van Vugt-Lussenburg, T. Zeldenthuis, J.S.B. de Vlieger, J.N.M. Com- 100 4.25E+08
mandeur, N.P.E. Vermeulen, Chem. -Biol. Interact. 171 (2008) 96. 1.0
125°C 75 4.01E+08
50 4.36E+08
22. E. Vottero, V. Rea, J. Lastdrager, M. Honing, N.P. Vermeulen, J.N.M. Commandeur, J. Biol. 100°C 75°C
Inorg. Chem., 487, in press, doi:10.1007/s00775-011-0789-4. 0.5 50°C
23. F. Cuyckens, R. Hurkmans, J.M. Castro-Perez, L. Leclercq, R.J. Mortishire-Smith, Rapid Com-
mun. Mass Spectrom. 23 (2009) 327. 0.0
0.0 5.0 10.0 15.0 20.0 25.0 30.0 35.0 40.0 45.0 50.0 55.0 60.0 65.0 70.0 75.0 min
24. M. Holcapek, R. Jirasko, M. Lisa, J. Chromatogr. A 1217 (2010) 3908.
25. P. Deng, D.F. Zhong, F.J. Nan, S. Liu, D. Li, T. Yuan, X.Y. Chen, J.A. Zheng, Chem. Res. Toxi-
col. 23 (2010) 1617.
26. P. Henklova, R. Vrzal, B. Papouskova, P. Bednar, P. Jancova, E. Anzenbacherova, J. Ulricho-
va, P. Maurel, P. Pavek, Z. Dvorak, Eur. J. Pharmacol. 593 (2008) 16.
27. M. Holcapek, L. Kolarova, M. Nobilis, Anal. Bioanal. Chem. 391 (2008) 59.
28. A. Baumann, U. Karst, Exp. Opin. Drug Metab. Toxicol. 6 (2010) 715.
Figure S3: Comparison of HTLC-ESI-MS and LC-ESI-MS
(x1,000,000)
327.1371 (1.00)
6.0 328.1211 (1.00)

A
344.0983 (1.00)
Supplementary Material 5.0

Figure S1: Structures of the compounds used in this study 4.0


A: Temperature gradient
3.0 2 with 5 % ACN
Isoclozapine Loxapine Clothiapine 2.0 1 3
N N N
1.0

N N N 0.0
1 Isoclozapine
N N N
(x1,000,000)
3.0 4.0 5.0 6.0 7.0 8.0 9.0 min
2 Loxapine
327.1371 (1.00)
6.0 328.1211 (1.00) 3 Clothiapine
B
344.0983 (1.00)

2 3
Cl Cl Cl
N O S 5.0
H

4.0
B: ACN gradient
Chemical Formula: C18H19ClN4 Chemical Formula: C18H18ClN3O Chemical Formula: C18H18ClN3S
1
5 3.0 isothermal 50 °C
5
SB- 203580 - Iodo P38 MAPK Inhibitor VIII 2.0

N
1.0
O Cl

H
N O Br 0.0
3.0 4.0 5.0 6.0 7.0 8.0 9.0 min
S

N
N
H
Chemical Formula: C21H16IN3OS
Chemical Formula: C20H16BrClN2O NH2

202 203
Section 6 Chapter 6.1

Summary Conclusions and perspectives


This thesis aims at exploring new applications of high-resolution screening (HRS). Therefore, developed for the estrogen receptors α and β [1], for binding selectivity analysis should
our goal was to improve the corresponding platforms and extend their possibilities, mainly be mentioned in this context. These could be applied to investigate selectivity over close
by further integration of chromatography, mass spectrometry, bioassays and especially family members or generally problematic off-targets. For example, in the case of p38α,
innovative synthetic methods. We developed HRS platforms for two targets, p38α mitogen- the closely related kinases JNK-1, JNK-2 and JNK-3 could be simultaneously monitored
activated protein kinase (p38α) and soluble epoxide hydrolase (sEH), which were previously for selectivity during lead optimization [2]. Additionally, detection of selectivity for the
not accessible by HRS [Chapter  2]. Furthermore, by integrating four complementary different p38 isoforms might lead to molecular probes which could be used to investigate
modification methods with the p38α HRS platform, we achieved an efficient and integrated the biological role of p38β, γ and δ in more detail [3]. Concerning off-targets, the increasing
approach to generate and screen metabolite-like lead libraries, yielding structure-affinity interest in the novel concept of translational research may lead to the exploitation of the
relationships [Chapter  3]. We thoroughly analyzed the possibilities and limitations of large body of knowledge on p38 inhibitors in future drug discovery campaigns. For example,
structure elucidation of related substances by liquid chromatography–(ion trap–time-of- the suspected off-target interactions, related to certain side effects observed in the clinic,
flight) high-resolution mass spectrometry (LC–HR-MSn) within the framework of HRS on might be included in selectivity studies.
basis of the kinase inhibitor derived metabolite-like lead libraries [Chapter 4.1]. Finally, we In any case, next to selectivity detection, future developments of HRS bioassays lie in
peaked into the future of HRS which may feature miniaturized NMR for additional structure miniaturization, because the reduction in both reagent consumption and sample volume
elucidation [Chapter  4.2] and/or inductively coupled plasma MS (ICP-MS) for absolute has important advantages [4]. Using less target protein helps to control the cost of HRS and
quantitation [Chapter 5] integrated directly into the HRS platform. thus improves its competitive position versus HTS. Enhanced mass sensitivity, possibly in
As far as the development of HRS bioassays is concerned, on the example of p38α, we combination with pre-concentration through the integration of on-line SPE, might finally
demonstrated the first on-line post-column bioaffinity assay reported for any enzyme target allow HRS analysis of clinical samples in the future.
and integrated it into an HRS platform [Chapter 2.2]. For the sEH HRS platform, we were We further advanced integration in HRS platforms, synthesizing metabolite-like lead libraries
mainly interested in making the possibilities of HRS for metabolism studies available to this and analyzing them for structure and affinity in one platform or even in one hyphenated
novel target and therefore took a very classic approach [Chapter 2.3]. system. Combining synthesis and analysis in one workflow resulted in an efficient exploration
For the p38α HRS platform, we additionally introduced new solutions for the screening of of neglected chemical space around a substrate scaffold, consequently yielding structure-
highly lipophilic compounds. The use of fused silica capillaries demonstrates that there activity relationships with the input of no more than the substrates. These substrates
are still possibilities to improve upon current HRS bioassays technology, even in aspects were important p38α inhibitors, including a lead (DMPIP), a widely used molecular probe
that have been as extensively studied as post-column reactors. In this example, the (SB203580) and clinical candidates (BIRB796and TAK-715). As we focused on metabolite-
enhanced peak shapes improve chromatographic resolution and dilution calculation. We like lead libraries, it seemed prudent to additionally investigate, whether the platforms would
achieved excellent performance of the p38α and the sEH HRS platforms. For the p38α be useful in selecting production strategies for actual metabolites. To this end, we included
HRS platform, this is indicated by excellent figures-of-merit, such as a Z’-factor of ca. in vitro metabolism by human liver microsomes (HLM) into the method comparison.
0.8 and an S/N of up to 100, and an affinity ranking in accordance with literature values. The electrochemical conversion (EC) HRS platform was certainly the most interesting
Unfortunately, the only tested compound of which affinity data on non-phosphorylated integrated synthesis approach from an analytical point of view [Chapter 3.1]. The EC setup
p38α was available (BIRB796), showed unconventionally slow binding kinetics, rendering employed had already been successfully used in EC–MS and EC–LC–MS setups and
a quantitative comparison of the IC50 values for validation purposes impossible, due to the required no intermediate sample pre-treatment step. Therefore, a completely integrated
limited incubation times achievable in HRS. Nonetheless, its affinity was detected as were EC–LC–p38α binding/MS platform was achieved. This proved to be especially useful for
those of representative compounds of all three types of ATP-competitive binding. the detection of reactive EC products [Chapter 3.1] which were also (partly) observed later
The sEH HRS platform featured an S/N>60, though quality would have certainly profited in human liver microsomal incubations [Chapter 3.3].
from using state-of-the-art chromatographic equipment. We could very reliably match affinity The electrophilic reactivity of the observed quinoneimine also offers the possibility for a
and small-molecule structure and even tackle isobaric and many isomeric compounds. new synthetic approach. A pre-column incubation step with a mixture of nucleophiles could
This holds for both HRS platforms, but was more extensively demonstrated for p38α be integrated into the EC–HRS platform, allowing the production of extensive lead libraries
[Chapter 2.2, Chapter 3] than for sEH [Chapter 2.3]. The clear advantages of HRS over with a broad chemical diversity in a rapid and automated fashion as well as the direct
HTS with respect to the bioassay level are exemplified by a decreased p38α concentration analysis of the reaction mixtures in the same system. Especially interesting might be the
and a similar sEH concentration at significantly reduced incubation time. Obviously, the integration of a second electrochemical (detection) step between column and MS which
ability to analyze mixtures is an important advantage of HRS over HTS as well. would serve to probe the redox stability of the new compounds. Through this approach, it
For both HRS platforms, we successfully tackled issues with enzyme stability. In addition, might be possible to find leads with a reduced tendency to form reactive products [5]. This
for the sEH HRS platform, we overcame issues with low substrate solubility. Furthermore, is especially interesting, if adverse effects caused by reactive metabolites are known or
during the development and the HRS campaigns [Chapter 2, Chapter 3], we observed a anticipated. The general approach of EC–HRS might also be interesting in stages of drug
6.1 number of interferences which are clearly visualized by the HRS bioassays. Examples are
the auto-fluorescence and peak tailing in the p38α HRS platform, which interfere with the
discovery other than lead optimization. When fitting an EC–HRS platform with a bioassay
for a notorious off-target, such as hERG [6], it could be very useful as a rapid metabolic 6.1
IC50 measurements [Chapter 2.2, Chapter 3.2], and to some extend the precipitation issues alert screening. Though EC is not a complete mimic of human metabolism, redox weak
during the development of the sEH HRS bioassay [Chapter 2.3]. While the visualization spots and the toxicological consequences of the associated modifications may be identified
of interferences might be seen as a disadvantage, we believe that it in fact is infinitely early on in drug discovery.
preferable to spot and address these issues than to put confidence in faulty data. Therefore, The use of biocatalysts to generate metabolite-like lead libraries proved very successful
we regard the ability of the HRS platform to resolve interferences as one of the major as well. The library of Cytochrome P450 BM3 (BM3) mutants applied produced a number
strength of this approach. of conversion products. The products showed increased hydrophilicity and one of them
There are several interesting directions in which both the p38α and the sEH HRS platform even retained full p38α affinity [Chapter 3.2]. Because large parts of the BM3 mutant library
may be further developed. Parallel bioassays (two or even multiple), similar to the one were “humanized”, a significant overlap with products from HLM incubation was found. The

206 207
fact that, through up-scaling and purification, IC50 determination and NMR measurements showed, using ion-trap fragmentation in combination with HR-MS readout by time-of-flight,
were enabled, greatly enhanced the detail of information obtained. In addition, we thereby the value of clearly assigning precursor-product ion relationships for structure elucidation
demonstrated how the initial results of the HRS platform can be used to guide further [Chapter 4.1].
structural and pharmacological investigation of promising conversion products. Next to the HR-MSn structure elucidation, we pursued an intriguing possibility to incorporate
Subsequently, we compared the EC and the biocatalytic approach to chemically (H2O2) and NMR structure elucidation into an HRS platform in the future [Chapter  4.2]. As starting
photochemically (visible light) induced transformations [Chapter 3.3]. H2O2 was very good points, we took the reactive products of BIRB796 discovered in the EC–HRS platform
in making many different, more hydrophilic products, e.g., numerous oxygenated isomers. [Chapter  3.1]. We set out to elucidate their structure by NMR. The logical choice for an
Photochemical conversion showed some unique products and can induce isomerism in approach that can be integrated with HRS was a miniaturized flow probe NMR in order to
the core structure, which may result in novel pharmacophores. One consequence of that match the small sample volume and the flow-through EC system.
isomerism is a more limited success in structure elucidation by MS [Chapter 4.1]. In the end, we combined an up-scaled electrode, an SPE step for pre-concentration
In comparison, all methods have their specific profile of strength and weaknesses. For and solvent exchange, and a stripline-NMR chip probe equipped to a 600  MHz NMR
example, biocatalysis and H2O2 are most successful in increasing hydrophilicity. While instrument. The system was well suited for the detection of the BIRB796 standard, and the
biocatalysis is the most laborious method, but can also be the most selective, H2O2 is solvent exchange was efficient enough to be able to compare the EC–SPE–stripline-NMR
very straightforward but often produces the most (isomeric) products. Photochemistry and results to conventional (2D-)1H-NMR. Unfortunately, the conversion rate of BIRB796 to its
EC do not favor increased hydrophilicity in the products, but photochemistry yields the two reactive products was significantly lower than in the off-line experiments. Therefore,
most unique products. However, the EC–HRS platform is very favorable for the analysis of identification of the products could only be achieved by comparison with the conventional
reactive products, due to its direct analysis capabilities. 1
H-NMR spectra, instead of by the desired independent structure elucidation. However,
In conclusion, all conversion methods are highly complementary and thus together present the platform showed promising results and there are numerous possibilities to close the
an interesting toolbox for the generation of metabolite-like lead libraries. We could show the existing sensitivity gap in order to allow direct integration into the HRS platform.
impressive value of integrating synthesis of mixtures into the HRS strategy. The p38α HRS Integrated LC–MS/NMR platforms are already used in pharmaceutical and other applications
platform rapidly delivered an initial structure-activity relationship for the lead libraries and [7,8]. As combined MS and NMR structure elucidation is crucial in HRS applications as
can be considered as a solid starting point for further pharmacological and toxicological well [1], HRS platforms featuring this combined structure elucidation power are certainly a
investigation. worthwhile aim.
Next to producing lead libraries with favorable properties, perhaps unsurprisingly, A major enhancement of the possibilities and a major step towards a wider applicability
especially the biocatalytic and the chemical approach showed potential for the synthesis of HRS is the integration of absolute quantitation into HRS platforms. For example, this
of metabolite standards. The novelty is that the implementation of the HRS platform allows may enable a quantitative structure-activity relationship (QSAR) directly from mixtures
a focus on active metabolites for further evaluation while at the same time still providing a generated by the described modification methods. A simple injection of a dilution series of
comprehensive overview. Again, this might also be an interesting application for off-target the mixture might suffice in order to obtain IC50 curves. Mainly due to the lack of standards,
HRS bioassays. detection techniques focused on atoms rather than on molecules will be the methods of
However, the biocatalytic and the chemical approach will require different strategies choice when dealing with unknown compounds. Detection techniques like MS or UV/VIS,
for metabolite standard production. By mutation, a biocatalyst can be generated which which focus on molecular properties, show a large variation in response, depending on
exclusively or predominantly produces one metabolite that is then easily purified. The the molecular structure. While the response also shows large variation between different
strategy for H2O2 will rely on simultaneous purification of all (major) products, as conversion atoms when atomic properties are detected, it is relatively easy to find a common atom in
is undirected (high number of isomers) and there is little opportunity to tune the product many different organic molecules.
profile. However, this will also provide standards which would be ignored in a focused One atom property based method, which was already discussed, is NMR. However, NMR
approach, but might still be useful, for example when metabolites are detected in human is influenced by the chemical environment of the detected atom which has consequences
that have not been predicted in in vitro or animal models. for the response. Fortunately, these are much less pronounced than in molecular property
Being an integral part of the HRS platforms, we closely investigated the possibilities and based detection techniques and can be minimized by careful consideration of experimental
challenges of structure elucidation by LC–HR-MSn under the special circumstances of parameters such as relaxation time. The real issue with NMR is its currently inadequate
a (HRS) screening paradigm [Chapter  4.1]. The resulting restrictions include the use of sensitivity. Yet, if this hurdle can be overcome, NMR promises quantitation and additional
“generic” settings for the ionization and fragmentation experiments as well as prohibiting structure information at the same time.
the use of elaborate MS experiments like stable isotope labeling and H/D-exchange ICP-MS is much more sensitive and its response is virtually independent of the chemical
experiments. Nonetheless, the attribution of modifications to specific parts of the molecule environment, due to the constant atomic ionization efficiency and the huge difference
is often successful and in special cases allows absolute structure elucidation, for example between molecular bond energy and the atomic ionization energy [9]. Unfortunately, the
6.1 for CP472, a conversion product of DMPIP in the EC experiments [Chapter 3.1].
The sometimes drastic changes in fragmentation initiated by comparatively minor
main elements of organic molecules, hydrogen, carbon, oxygen, and nitrogen, cannot be
targeted by LC–ICP-MS for various reasons. However, many pharmaceuticals contain a 6.1
modifications and unexpected intra-molecular rearrangements presented challenges in the halogen and/or sulfur atom and there are even examples containing metal atoms. While we
LC–HR-MSn structure elucidation. These rearrangements were also interesting examples measured bromine and iodine very sensitively by ICP-MS, this was not the case for sulfur
of gas-phase chemistry, such as the electrocyclic rearrangement in the fragmentation of and chlorine [Chapter 5]. The observed isobaric polyatomic interferences may be resolved
TAK-715, the rearrangement underlying the formation of CP305 (or its fragmentation) using reaction/collision cells or an ICP-HR-MS instrument [9]. This might allow sensitive
from SB203580, the hydride rearrangement leading to water elimination from the aromatic quantification of sulfur or chlorine containing molecules, where detection limits with unit
hydroxymethyl group in CP416A (derived from TAK-715), and the rearrangement of the mass resolution ICP-MS were a factor of 30 or 200, respectively, short of HRS compatible
whole para-fluoro-benzyl group in DMPIP. Additionally, we detected an unexpectedly high quantitation [Chapter 5]. ICP-MS relies on stable solvent conditions and is therefore not
number of homolytic cleavages in violation of the even-electron rule. In the process, we compatible with classic solvent gradients [10]. High-temperature LC (HTLC) presents an

208 209
elegant way to achieve efficient separation at isocratic solvent conditions [Chapter 5]. HTLC WK, Evans DC, Kumar S (2003) Addressing the metabolic activation potential of new leads in
is not only interesting for ICP-MS compatibility, but may also present a solution to reduce drug discovery: a case study using ion trap mass spectrometry and tritium labeling techniques.
the organic modifier influence on post-column bioassays. Though the ESI-MS response is J Mass Spectrom 38 (2):211-221
decreased at low organic modifier concentrations, the high sensitivity of ESI-MS usually 6. Piper DR, Duff SR, Eliason HC, Frazee WJ, Frey EA, Fuerstenau-Sharp M, Jachec C, Marks BD,
Pollok BA, Shekhani MS, Thompson DV, Whitney P, Vogel KW, Hess SD (2008) Development
allows a favorable compromise.
of the predictor HERG fluorescence polarization assay using a membrane protein enrichment
In its application to drug discovery and related fields, we have demonstrated many approach. Assay Drug Dev Technol 6 (2):213-223
strengths and possibilities of HRS and encountered some of its limitations. However, the 7. Corcoran O, Spraul M (2003) LC-NMR-MS in drug discovery. Drug Discov Today 8 (14):624-631
role HRS platforms will play in the future depends on many factors. At the moment, the 8. Tang H, Xiao C, Wang Y (2009) Important roles of the hyphenated HPLC-DAD-MS-SPE-NMR
development status of HRS platforms limits their application to niches where separation technique in metabonomics. Magn Reson Chem 47 Suppl 1:S157-162
of complex mixtures is a key element, for example screening of natural product libraries 9. Ammann AA (2007) Inductively coupled plasma mass spectrometry (ICP MS): a versatile tool. J
[11,12]. The specific niche will determine the HRS platform’s outward appearance, e.g., Mass Spectrom 42 (4):419-427
whether pre-column affinity selection or post-column bioassays are employed, or whether 10. Gammelgaard B, Hansen HR, Sturup S, Moller C (2008) The use of inductively coupled plasma
on-line or at-line hyphenation is favored [Chapter  1.1.2]. Miniaturization may present a mass spectrometry as a detector in drug metabolism studies. Expert Opin Drug Metab Toxicol 4
way to reduce HRS cost [4] and thus enhance its commercial competitive position versus (9):1187-1207
11. Heus F, Vonk F, Otvos RA, Bruyneel B, Smit AB, Lingeman H, Richardson M, Niessen WM, Kool
HTS. However, knowledge presents a much greater hurdle towards the implementation of
J (2013) An efficient analytical platform for on-line microfluidic profiling of neuroactive snake
HRS platforms. Currently, a user has to be trained in all analytical and pharmacological venoms towards nicotinic receptor affinity. Toxicon 61:112-124
disciplines related to the HRS platform. This will only change, if commercial solutions for 12. Potterat O, Hamburger M (2013) Concepts and technologies for tracking bioactive compounds
robust and fully integrated systems as well as automated data analysis tools become in natural product extracts: generation of libraries, and hyphenation of analytical processes with
available in the future. For HRS to be recognized as a real alternative in pharmaceutical bioassays. Nat Prod Rep 30 (4):546-564
screening and to create a demand for these solutions in the first place, next to maturity 13. Crommelin D, Stolk P, Besancon L, Shah V, Midha K, Leufkens H (2010) Pharmaceutical
and user friendliness, one hurdle is potentially all-important. HRS does not smoothly fit the sciences in 2020. Nat Rev Drug Discov 9 (2):99-100
high-throughput batch approach dominating contemporary pharmaceutical industry and is 14. Kola I, Landis J (2004) Can the pharmaceutical industry reduce attrition rates? Nat Rev Drug
thus very rarely seriously considered. However, this may very well change in the not so Discov 3 (8):711-715
far future. The pharmaceutical industry is under great pressure to critically evaluate their
R&D strategies and present more viable alternatives [13,14]. More flexible, quality-focused
drug discovery campaigns and an increasing interest in flow chemistry as alternative to
batch approaches may make HRS platforms an attractive tool. Flow chemistry might be
integrated as demonstrated in the EC–HRS platform [Chapter 3.1] or with an intermediate
on-line SPE step. With appropriate flow splitting and at substantially decreased cost, HRS
might also be useful beyond discovery, for example for monitoring up-scaling or even for
quality control of active pharmaceutical ingredient production based on the (side) effect
rather than molecular composition. Thus, the “ideal” HRS platform of the future consists
of a (multiple) modification method(s), state-of-the-art (multidimensional) separation,
multiple bioassays for selectivity analysis, two independent structure analysis techniques
like ESI-HR-MS (including MS/MS or MSn capabilities) and NMR, and two independent
quantitation techniques such as ICP-MS and NMR, operates at micro- or even (partly)
nano-scale, and can be used by scientists with diverse backgrounds. Applications might
include fully automated quantitative structure-activity relationship studies and, without a
modification method, the quantitative elucidation of the pharmacokinetic contributions of all
pharmacologically or toxicologically relevant metabolites for metabolic profiling.

References
1. de Vlieger JS, Kolkman AJ, Ampt KA, Commandeur JN, Vermeulen NP, Kool J, Wijmenga SS,
Niessen WM, Irth H, Honing M (2010) Determination and identification of estrogenic compounds
6.1 generated with biosynthetic enzymes using hyphenated screening assays, high resolution mass
spectrometry and off-line NMR. J Chromatogr B 878 (7-8):667-674 6.1
2. Goldstein DM, Kuglstatter A, Lou Y, Soth MJ (2010) Selective p38alpha Inhibitors Clinically
Evaluated for the Treatment of Chronic Inflammatory Disorders. J Med Chem 53 (6):2345-2353
3. Zhang J, Shen B, Lin A (2007) Novel strategies for inhibition of the p38 MAPK pathway. Trends
Pharmacol Sci 28 (6):286-295
4. Heus F, Giera M, de Kloe GE, van Iperen D, Buijs J, Nahar TT, Smit AB, Lingeman H, de Esch
IJ, Niessen WM, Irth H, Kool J (2010) Development of a microfluidic confocal fluorescence
detection system for the hyphenation of nano-LC to on-line biochemical assays. Anal Bioanal
Chem 398 (7-8):3023-3032
5. Samuel K, Yin W, Stearns RA, Tang YS, Chaudhary AG, Jewell JP, Lanza T, Jr., Lin LS, Hagmann
210 211
Chapter 6.2

Nederlandse samenvatting
Hoge-resolutie screening [Hoofdstuk 2, Hoofdstuk 3] een aantal verstoringen gezien welke juist duidelijker zichtbaar
waren in de HRS bioassays. Voorbeelden zijn de auto-fluorescentie en het peak tailing in
voor metaboliet-gerelateerde lead-bibliotheken het p38α HRS bioassay, welke de IC50 metingen verstoren [Hoofdstuk 2.2, Hoofdstuk 3.2],
en gedeeltelijk de neerslagproblemen tijdens de ontwikkeling van het sEH HRS bioassay
In dit proefschrift worden nieuwe applicaties van hoge-resolutie screening (HRS) onder-
[Hoofdstuk 2.3]. De zichtbaarheid van de verstoringen zou als een nadeel aangemerkt kun-
zocht. Daarvoor streefden wij naar verbetering van bestaande systemen en verbreding van
nen worden, maar wij geven sterk de voorkeur aan het visualiseren en rekening houden
hun mogelijkheden. Hierbij hoorden geavanceerde integratie van chromatografie, mas-
met dit soort verstoringen boven het vertrouwen op onjuiste gegevens. Daarom zien wij het
saspectrometrie, bioassays en in het bijzonder innovatieve synthetische methodes. Wij
zichtbaar maken van verstoringen door het HRS systeem als een van zijn grote voordelen.
ontwikkelden nieuwe HRS systemen voor twee doelproteïnen, p38α mitogen-geactiveerd
Een andere belangrijke ontwikkelingsstap is de integratie in HRS systemen van methoden
proteïne kinase (p38α) en soluble epoxide hydrolase (sEH) [Hoofdstuk 2]. Verder bereikten
voor de synthese van metaboliet-gerelateerde lead-bibliotheken, waarmee we zowel de
wij een efficiënte en geïntegreerde aanpak voor het genereren en screenen van metabo-
synthese als de analyse van structuur en affiniteit in één aanpak of zelfs in een geheel
liet-gerelateerde lead-bibliotheken, door integratie van vier complementaire modificatieme-
geïntegreerd systeem realiseren. Dit resulteerde in een efficiëntere bestudering van de
thodes met het p38α HRS systeem, welke uiteindelijk resulteerden in structuur-affiniteits-
chemische ruimte rond een substraatkernstructuur, met als eindpunt een structuur-affini-
relaties [Hoofdstuk 3]. Wij analyseerden grondig de mogelijkheden en beperkingen van
teitsrelatie met niet meer dan het substraat als inbreng. De substraten waren belangrijke
structuuropheldering van gerelateerde stoffen met behulp van vloeistofchromatografie–
p38α remmers, waaronder een lead-verbinding (DMPIP), een veelgebruikte moleculaire
(ion-trap–time-of-flight) hoge-resolutie massaspectrometrie (LC–HR–MSn) in relatie met
sonde (SB203580) en klinische kandidaat moleculen (BIRB796 en TAK-715). Omdat wij
HRS gebaseerd op de metaboliet-gerelateerde lead-bibliotheken van de kinase remmers
gefocusseerd waren op metaboliet-gerelateerde lead-bibliotheken, hebben wij ook uitge-
[Chapter 4.1]. Uiteindelijk waagden wij een poging om de (mogelijke) toekomst van HRS
zocht of de synthesesystemen geschikt waren als productiestrategieën voor daadwerke-
te exploreren. Deze zou mogelijk bestaan in integreren in het HRS systeem van gemini-
lijke metabolieten. Daarom hebben wij ook in vitro metabolisme met behulp van humane
aturiseerde kernspinresonantie spectroscopie (NMR) voor verdere structuuropheldering
lever mikrosomen (HLM) in de methodenvergelijking opgenomen.
[Hoofdstuk 4.2] en/of inductief-gekoppelde plasma MS (ICP-MS) voor absolute kwantifice-
Het elektrochemische omzetting (EC) HRS systeem was analytisch gezien zeker de mee-
ring [Hoofdstuk 5].
st interessante geïntegreerde synthese-aanpak [Hoofdstuk 3.1]. Het EC apparaat was al
Het p38α systeem is het eerste on-line post-column bioaffiniteitsassay systeem voor een
succesvol ingezet bij EC–MS en EC–LC–MS systemen en vereiste geen intermediaire
enzym, een belangrijke stap in de ontwikkeling van HRS bioassays [Hoofdstuk 2.2]. Bij
monsterbewerkingsstappen. Daardoor kon een volledig geïntegreerde EC–LC–p38α bin-
het sEH HRS systeem waren wij voornamelijk geïnteresseerd in het toegankelijk maken
ding/MS systeem worden opgezet. Dit bleek bijzonder nuttig bij de detectie van reactieve
van de mogelijkheden van HRS in metabolismestudies voor dit doelenzym en hebben we
EC producten [Hoofdstuk 3.1] welke (deels) ook later in de HLM incubaties teruggevonden
daarom voor een klassieke benadering gekozen [Hoofdstuk 2.3].
worden [Hoofdstuk 3.3].
In het p38α HRS systeem hebben wij bovendien nieuwe oplossingen voor het screenen
Het gebruik van biokatalyse voor het genereren van metaboliet-gerelateerde lead-biblio-
van zeer lipofiele stoffen geïntroduceerd. Het gebruik van fused-silica capillairen laat zien
theken bleek ook heel succesvol te zijn. De gebruikte bibliotheek van Cytochrome P450
dat er nog steeds mogelijkheden zijn om de HRS bioassay technologie te verbeteren,
BM3 (BM3) mutanten produceerde een aantal omzettingsproducten. Deze producten toon-
zelfs in zo veelvuldig bestudeerde aspecten als post-kolom reactoren. In dit geval resul-
den verhoogde hydrofiliciteit en één ervan behield zelfs de volle p38α affiniteit [Hoofdstuk
teerde de betere piekvorm in een betere chromatografische resolutie en nauwkeurigere
3.2]. Omdat grote delen van de BM3 bibliotheek ‘gehumaniseerd’ waren, was er een signi-
verdunningsberekeningen. Wij bereikten uitstekende prestaties van zowel het p38α als het
ficante overlap met producten van de HLM incubaties. Het feit dat wij door schaalvergroting
sEH HRS systeem. Voor het p38α HRS systeem blijkt dit uit uitstekende kengetallen zoals
en opzuivering ook IC50 waardes konden bepalen en NMR metingen konden uitvoeren
een Z‘-factor van rond 0.8 en een signaal/ruis verhouding (S/N) van 100. Verder kwam
heeft de informatiediepte duidelijk verhoogd. Verder hebben wij daardoor kunnen aantonen
de gemeten affiniteitsranking overeen met de literatuurwaarden. Helaas toonde de enige
hoe de aanvankelijke resultaten van het HRS systeem kunnen worden gebruikt om verdere
teststof waarvoor affiniteitsdata op niet-phosphoryleerde p38α beschikbaar zijn (BIRB796)
structuuropheldering en farmacologisch onderzoek van veelbelovende omzettingsproduc-
een buitengewoon langzaam kinetiek. Dat voorkwam een kwantitatieve vergelijking van
ten te sturen.
de IC50 waarden voor validatiedoeleinden omdat vanwege de beperkte incubatietijden in
Vervolgens vergeleken wij de EC en de biokatalytische aanpak met chemisch (H2O2) of fo-
HRS het evenwicht niet bereikt werd. Toch kon de affiniteit van BIRB796 en die van andere
tochemisch (zichtbaar licht) induceerde omzettingen [Hoofdstuk 3.3]. H2O2 bleek zeer goed
representatieve stoffen van alle drie types van ATP-competitieve binding gemeten worden.
in staat om veel verschillende en meer hydrofiele producten te maken, bijvoorbeeld een
Het sEH HRS systeem toonde een S/N van boven de 60 maar de kwaliteit zou veel beter
grote aantal geoxydeerde isomeren. Fotochemische omzetting resulteerde in enige unieke
zijn geweest als er modernere chromatografische apparaten waren gebruikt. Wij konden
producten en induceerde isomerie in de kernstructuur, wat mogelijk tot nieuwe farmacofo-
zeer betrouwbaar de affiniteit en de moleculestructuur combineren en daardoor zelfs iso-
ren zou kunnen leiden. Een consequentie van isomerie is het wat beperktere succes in MS
bare en vele isomere componenten onderscheiden. Dit was in beide HRS systemen reali-
6.2 seerbaar maar kon in het p38α systeem [Hoofdstuk 2.2, Hoofdstuk 3] nog duidelijker wor-
den aangetoond dan in het sEH systeem [Hoofdstuk 2.3]. De duidelijke voordelen van HRS
structuuropheldering [Hoofdstuk 4.1].
Iedere gebruikte synthesemethode heeft haar eigen profiel van sterke punten en beper- 6.2
kingen. Biokatalyse en H2O2 zijn bijvoorbeeld het meest succesvol in het verhogen van hy-
ten opzichte van hoge-doorvoer screening (HTS) met betrekking tot het bioassay gedeelte
drofiliciteit. Terwijl biokatalyse de meest arbeidsintensieve methode is, maar ook de meest
worden onder ander duidelijk in de lagere p38α concentratie en de vergelijkbare sEH con-
selectieve, is H2O2 zeer eenvoudig te gebruiken, maar produceert het vaak ook de meeste
centratie ondanks de significant gereduceerde incubatietijd. De mogelijkheid om mengsels
(isomere) producten. Fotochemie en EC hebben geen voorkeur voor hogere hydrofiliciteit
te analyseren is overduidelijk ook een belangrijke voordeel van HRS ten opzichte van HTS.
in de producten, maar fotochemie levert de meeste unieke producten. Aan de andere kant
Verder hebben wij voor beide systemen enzymstabiliteitgerelateerde problemen opgelost
is het EC–HRS systeem vanwege zijn directe analysemogelijkheden zeer krachtig voor de
en een oplossing gevonden voor de substraatoplosbaarheidsproblemen die bij het sEH
analyse van reactieve producten.
systeem speelden. Ook hebben wij bij het opzetten van en tijdens de HRS experimenten
Wij concluderen dat alle omzettingsmethoden zeer complementair zijn en dus samen een

214 215
interessante verzameling vormen voor het genereren van metaboliet-gerelateerde lead-bi- kelijk is van de chemische omgeving van een atoomkern, vanwege de constant atomaire
bliotheken. Zo waren wij in staat om de meerwaarde van de integratie van de synthese van ionisatie en de grote verschillen tussen moleculaire bindingsenergie en atomaire ionisa-
mengsels met het HRS strategie te demonstreren. Het p38α systeem leverde in korte tijd tie-efficiëntie. Helaas kunnen de hoofdbestanddelen van organische moleculen (water-
een initiele structuur-affiniteitsrelatie voor de lead-bibliotheken op en kan als een solide stof, koolstof, zuurstof en stikstof) om verschillende redenen niet met LC–ICP-MS worden
startpunt voor verder farmacologisch en toxicologisch onderzoek gezien worden. gemeten. Maar veel medicijnen bevatten halogeen- en/of zwavelatomen en er zijn zelfs
Naast het produceren van lead-bibliotheken met gunstige eigenschappen hebben, mis- voorbeelden welke metaalatomen bevatten. Broom en jodium konden zeer gevoelig met
schien niet onverwacht, vooral de biokatalytische en de chemische aanpak potentieel voor ICP-MS worden gemeten, maar dat was niet het geval bij zwavel en chloor [Hoofdstuk 5].
de synthese van metabolietstandaarden getoond. De innovatie ligt hierbij vooral in de mo- De waargenomen isobare polyatomaire verstoringen zouden met reactie/botsingscellen of
gelijkheid om zich voor gedetailleerd onderzoek op actieve metabolieten te focusseren en ICP-HR-MS instrumenten kunnen worden voorkomen. Daardoor zou gevoelige kwantifice-
toch tegelijkertijd een breder overzicht te verwerven. ring van zwavel en/of chloor bevattende verbindingen mogelijk kunnen worden. Tot nu toe
Omdat het een integraal bestanddeel van het HRS systeem is, hebben wij in meer detail waren de detektielimieten op het lage-resolutie ICP-MS 30 respectievelijk 200 keer te laag
naar mogelijkheden en beperkingen van structuuropheldering met behulp van LC–HR-MSn om een met HRS verenigbare kwantificering te leveren [Hoofdstuk 5]. ICP-MS is gebaseerd
gekeken, zoals die in de bijzondere omstandigheden van een (HRS) screeningsomgeving op een gelijkmatige stroom van oplosmiddel en is daarom niet geschikt om met klassieke
optreden [Hoofdstuk 4.1]. De bijhorende beperkingen omvatten het gebruik van ‘generieke’ oplosmiddelgradiënten gecombineerd te worden. Hoge-temperatuur LC (HTLC) biedt een
parameters voor ionisatie- en fragmentatie-experimenten en verhinderen van ingewikkelde elegante mogelijkheid om een efficiente scheiding bij constante oplosmiddelsamenstelling
MS experimenten zoals stabiele-isotooplabeling en H/D-uitwisseling. Niettemin was het te realiseren [Hoofdstuk 5]. Daarbij is HTLC niet alleen in verband met ICP-MS interessant,
relateren van modificatieposities naar specifieke delen van de moleculen vaak succesvol maar het kan ook een oplossing zijn om de invloed van de oplosmiddelgradiënt in een
en kon in speciale gevallen zelfs de absolute structuur opgehelderd worden, bijvoorbeeld post-kolom bioassay te beperken. Ook hebben lage organische oplosmiddelconcentraties
voor CP472, een omzettingsproduct van DMPIP in de EC experimenten [Hoofdstuk 3.1]. een negatieve invloed op de ESI-MS respons, maar maakt de hoge gevoeligheid van ESI-
De soms drastische veranderingen in fragmentatie als gevolg van relatief kleine modifica- MS toch meestal een aantrekkelijk compromis mogelijk.
ties en onverwachte intra-moleculaire omleggingen waren uitdagingen in de LC–HR-MSn
structuuropheldering. Deze omleggingen waren tegelijkertijd interessante voorbeelden
van gasfase-chemie, bijvoorbeeld de elektrocyclische omlegging in de fragmentatie van
TAK-715, de omlegging voorgaande aan de vorming van CP305 (of zijn fragmentatie) uit
SB203580, de hydride-omlegging met als gevolg watereliminatie vanuit de aromatische
hydroxymethyl groep in CP416A (afkomstig van TAK-715), en de omlegging van de hele
para-fluoro-benzyl groep in DMPIP. Bovendien detecteerden wij een onverwacht groot
aantal homolytische breukreacties welke in strijd zijn met de even-elektron regel. Daarbij
hebben wij, door gebruik te maken van de ion-trap fragmentatie in combinatie met time-of-
flight HR-MS detectie, het belang van een heldere relatie tussen precursor ion en product
ionen voor structuuropheldering aangetoond [Hoofdstuk 4.1].
Naast de HR-MSn structuuropheldering verkenden wij een veelbelovende mogelijkheid om
in de toekomst structuuropheldering met behulp van NMR te integreren met het HRS sys-
teem [Hoofdstuk 4.2]. Hierbij stonden de reactieve producten van BIRB796 centraal die
in het EC–HRS systeem ontdekt werden [Hoofdstuk 3.1]. Hun structuur wilden wij met
behulp van NMR ophelderen. De logische aanpak gericht op integratie met HRS zou een
miniaturiseerde flow-sonde NMR kunnen zijn, waarmee de kleine monstervolumes en een
doorstroom-EC-apparaat goed te combineren zijn. Uiteindelijk gebruikten wij een vergrote
werkelektrode en een vaste-fase extractie (SPE) stap ten behoeve van voorconcentratie
en oplosmiddeluitwisseling in combinatie met een stripline-NMR chip sonde en een 600
MHz NMR instrument. Het systeem was goed geschikt om de BIRB796 standaard te detec-
teren. De oplosmiddeluitwisseling was efficiënt genoeg om een vergelijking van EC–SPE–
stripline-NMR resultaten en conventioneel (2D-)1H-NMR mogelijk te maken. Helaas was de
omzettingsgraad van BIRB796 naar zijn twee reactieve producten in de on-line experimen-
6.2 ten duidelijk lager dan in de off-line experimenten. Daardoor was productidentificatie alleen
mogelijk door het vergelijken met de conventionele 1H-NMR spectra in plaats van door de 6.2
bedoelde onafhankelijke structuuropheldering. Toch liet het systeem veelbelovende resul-
taten zien en zijn er ook vele mogelijkheden om de bestaande gevoeligheid te verbeteren
en zo een directe integratie met het HRS systeem te bereiken.
Een grote stap voorwaarts in HRS zou de mogelijkheid zijn om absolute kwantificering
van gemeten componenten in een HRS systeem te integreren. Dan zou bijvoorbeeld een
kwantitatieve structuur-affiniteitsrelatie (QSAR) gemaakt kunnen worden direct op basis
van het mengsel gesynthetiseerd met de beschreven omzettingsmethoden. ICP-MS biedt
zich hiervoor aan omdat het heel gevoelig kan zijn en zijn respons vrijwel volledig onafhan-

216 217
I. List of publications
Section 2: High-Resolution Screening Platforms
1. D. Falck, J. Kool, W.M.A. Niessen, A practical guide to high-resolution screening. In
preparation.

Appendix
2. D. Falck, J.S.B. de Vlieger, W.M.A. Niessen, J. Kool, M. Honing, M. Giera, H. Irth,
Development of an on-line p38α mitogen-activated protein kinase binding assay and
integration of LC–HR-MS. Anal. Bioanal. Chem.,398 (2010) 1771-1780.
3. D. Falck, N.H. Schebb, S. Prihatiningtyas, J. Zhang, F. Heus, C. Morisseau, J. Kool,
B.D. Hammock. W.M.A. Niessen, Development of on-line liquid chromatography–bio-
chemical detection for soluble epoxide hydrolase inhibitors in mixtures. Chromato-
graphia, 76 (2013) 13-21.

Section 3: Integration of modification methods and HRS workflows


for the study of metabolite-like lead libraries
4. D. Falck, J.S.B. de Vlieger, M. Giera, M. Honing, H. Irth, W.M.A. Niessen, J. Kool,
On-line electrochemistry–bioaffinity screening with parallel HR-LC-MS for the gener-
ation and characterization of modified p38α kinase inhibitors. Anal. Bioanal.Chem.,
403 (2012) 367-375.
5. D. Falck, V. Rea, J. Kool, F.J.J. de Kanter, J.N.M. Commandeur, N.P.E. Vermeulen,
W.M.A. Niessen, M. Honing, Combination of biotransformation by P450 BM3 mutants
with on-line post-column bioaffinity and mass spectrometric profiling as a novel strat-
egy to diversify and characterize p38α kinase inhibitors. MedChemComm, 4 (2013)
371-377.
6. D. Falck, F.Rahimi Pirkolachachi, M. Giera, M. Honing, J. Kool, W.M.A. Niessen,
Comparison of (bio-)transformation methods for the generation of metabolite-like
compound libraries of p38α MAP kinase inhibitors using high-resolution screening. J.
Pharm. Biomed. Anal. (2013), In Press, http://dx.doi.org/10.1016/j.jpba.2013.08.045

Section 4: Integrated structure elucidation approaches for high-reso-


lution screening
7. D. Falck, J. Kool, M. Honing, W.M.A. Niessen, Tandem mass spectrometry study of
p38α kinase inhibitors and related substances. J. Mass Spectrom., 48 (2013) 718-
731
8. D. Falck, A.J. Oosthoek-de Vries, A. Kolkman, H. Lingeman, M. Honing, S. Wijmen-
ga, A. Kentgens, W.M.A. Niessen, EC–SPE–stripline-NMR analysis of reactive prod-
ucts: a feasibility study. Anal. Bioanal. Chem. 405 (2013) 6711-6720.

Section 5 : High temperature liquid chromatography hyphenated with


ESI–MS and ICP–MS detection for the structural characterization and
quantification of halogen containing drug metabolites
9. J.S.B. de Vlieger, M.J.N. Giezen, D. Falck, C. Tump, F. van Heuveln, M. Giera, J.
Kool, H. Lingeman, J. Wieling, M. Honing, H. Irth , W.M.A. Niessen, High tempera-
ture liquid chromatography hyphenated with ESI-MS and ICP-MS detection for the
structural characterization and quantification of halogen containing drug metabolites.
Anal. Chim. Acta, 698 (2011) 69-76.
AP
Other publications, not in the thesis:
1. M. Mladic, D. Scholten, S. Bayrak, D. Falck, WM.A. Niessen, M.J. Smit, J. Kool, Liq-
uid chromatography–mass spectrometry with parallel bioaffinity assessment for met-
abolic profiling of small ligands towards CXCR3 receptor. J. Exp. Pharmacol. Ther.,
219
in preparation.
2. J. Kool, A.F. Rudebeck, F. Fleurbaaij, S. Nijmeijer, D. Falck, R.A. Smits, H.F. Vischer,
R. Leurs, W.M.A. Niessen, High-resolution metabolic profiling towards G-protein cou-
II. Curriculum vitae
pled receptors: rapid and comprehensive screening of histamine H4 receptor ligands.
J. Chromatogr. A, 1259 (2012) 213-220. May 2013 - current
3. S. Nijmeijer, H.F. Vischer, A.F. Rudebeck, F. Fleurbaaij, D. Falck, R. Leurs, W.M.A.
Niessen, J. Kool, Development of a profiling strategy for metabolic mixtures by com- Researcher
bining chromatography and mass spectrometry with cell-based GPCR signaling. J.
Biomol. Screen., 17 (2012) 1329-1338. Glycomics and Glycoproteomics group
4. N.H. Schebb, D. Falck, H. Faber, E.M. Hein, U. Karst, H. Hayen, Fast method for Center for Proteomics and Metabolomics
monitoring phospholipase A2 activity by liquid chromatography-electrospray ioniza- Leiden University Medical Center
tion mass spectrometry. J Chromatogr A. 1216 (2009) 5249-5255. Leiden, The Netherlands

Topic: - Targeted glycoproteomics and glycopeptide derivatization

April 2009 – March 2013


PhD student
AIMMS Division of BioMolecular Analysis
VU University Amsterdam
Amsterdam, The Netherlands

Topic: - Development and application of high-resolution screening platforms


(Integrated combination of LC, MS and biological assays)
- Small molecule structure elucidation by tandem MS, supported by NMR

October 2003 - February 2009


Student in Chemistry

Faculty of Chemistry and Pharmacy


University of Münster
Münster, Germany

Thesis: - Development of a fast liquid chromatography – mass spectrometry


method for the detection of Phospholipase A2 activity
Title: - Diplom Chemiker (equivalent to Master of Science)
Grade: - Sehr gut (highest grade)

AP AP

220 221
III. Acknowledgements mile. When my MS was not working and I could not fix it myself, there was Ben. I appre-
ciated that very much. Thanks also for the SciFi talks. Dick-Paul, we were sort of on/off
colleagues. I was glad that I could always go to you for IT advice. Thanks for holding up
the office prank tradition and for the Dutch lessons. To my last year office mates Dina, In-
At last, I have the pleasure of looking back at the last four plus years and reminisce on all geborg, Jan-Hein, Reka, Petra and Willem, I would like to say thank you for your company,
the friendships formed and acquaintances made. I learned so much from almost everyone for talking and listening and for the extracurricular activities. With Bart and Dik, I first and
I met, from Bachelor student to Noble prize winner, that I am afraid I cannot do justice to foremost shared the pleasant and unpleasant parts of the practical courses. Thank you that
each and every one of them in this limited space. I thank everyone who I met on the way I never had to do it alone. For Niels M. I remember our long discussions, for example on
for making me feel welcome in this foreign and sometimes strange country. You had a great the differences of the Dutch and German education system which I enjoyed a lot. I regret
part in making my PhD studies one of the best experiences of my life and for that I owe you that we did not have more talks, but who knows how much work we would have gotten
many thanks. done, if we would have had a longer overlap. Niels J. and Linda, though we were never
My many supervisors are the first I will mention by name. Even though, there have been technically colleagues I am glad we kept in touch. Thanks for showing me the world and
quite some changes in my supervision, thanks to their efforts I never experienced this as a art of whiskey. Thank you, Lygia, for the introduction to the Brazilian cuisine and for the
disadvantage, rather the opposite. I always found open doors and I had a wealth of experi- games. Marek, thank you for sharing my amazement of ‘the strange Dutch’. My gratitude
ence in so many different topics to choose from to get my questions answered. also goes to the spectroscopists Cecilia, Cees, Freek, Gert, Ivonne, Joost and Silvia for
Dear Wilfried: If you had not agreed to become ‘Interim Head’, my PhD would certainly being great colleagues, sharing events and doing spectroscopy (Unfortunately, rarely with
have been very different and I cannot imagine it would have been as successful. You sacri- my participation). Erika, Eelco, Govert, Jordy, Lionel, Marc, Nel and Rob, thanks for being
ficed a lot in order to be a fulltime supervisor on a two day contract and for that I am deeply equally great colleagues.
thankful. Thank you for sharing your knowledge of and enthusiasm for MS, for always mak- I would also like to thank all of the students I had the pleasure of supervising during this
ing me a priority, for being an example of integrity and organisation, for providing structure period. This goes for the many students in the practical courses, but especially the longer
where I could not yet myself, and for making all this enjoyable. internship students, who certainly made an impression on this thesis. Thank you, Anders,
Dear Maarten: You were one of the constants during my thesis; from my first day of work Dana, Fatie, Frank, Harsha, Jiawen, Johann, Junella, Laurens, Saurabh and Setyo. I hope
at the TIPharma group meeting in Nijmegen till the last project with DSM. Constantly and you learned half as much from me as I learned from supervising you.
actively encouraging us to spend time in the labs in Oss and later in Geleen, you were our Much gratitude is due to my colleagues of the TIPharma D2-102 project. Being whole-
breach in the ivory tower and the key to the fabled land of corporate science. Even more heartedly welcomed into such an excellent interdisciplinary team was surely a key to the
than that, you were a constant source of inspiration by placing our daily work in a bigger success of my PhD work. The intense, but open and friendly scientific discussions at our
context, clarifying its importance and supporting new ideas. meetings were a constant source of inspiration and guidance for my work. Thank you, Ard,
Dear Hubertus: Thank you for giving me this opportunity, despite my own doubts at our first Cornelis, Eduardo, Galvin, Geert, Jaap, Jan, Jelle, Jon, Kirsten, Lionel, Lutgarde, Nico, Pa-
meeting, to work in the exciting D2-102 project. Although, from the start our daily contact tricia, Sybren and Vanina for the exchange of ideas and the fruitful collaborations. Though
was limited, I still feel I learned a lot from you. When I started in 2009, the group was still the author lists of the chapters speak for themselves, I would like to thank Ard and Sybren
heavily influenced by your ideas and I am sure I learned many lessons from you also indi- for hosting me in Nijmegen, Vanina, Eduardo, Jelle, Galvin, Jan and Nico for introducing
rectly through the other group members. me to the world of biocatalysis, Cornelis and Jaap for enabling the nice ICP data and Li-
Dear Jeroen: You were my daily supervisor during the later part of my thesis, but even be- onel, Geert and Lutgarde for getting me interested in the possibilities of Chemometrics.
fore that I learned many things from you about screening. Thanks for giving me the freedom An analytical scientist needs good instrumentation and support when things do not work
that a senior PhD student needs. You were a bottomless well of ideas and could always the way he wants them to or when he just wants to try something new. The Shimadzu
make me enthusiastic about new projects. IT-Tof LC–MS system was at the heart of my work and after coming to grips with (most
Dear Martin: Your daily supervision at the beginning of my thesis was very important to me. of) the possibilities of the software, I became quite fond of it. Chapter 4.1 would certainly
You always had a strong and clear opinion and whether I followed it or not that gave me the have been far less exciting with a ‘normal’ q-Tof. Thanks Evert, Johan, Tom and Vincent for
guidance and orientation I needed. As I also experienced later with Wilfried, we had similar your support with the equipment and of new ideas. I am also very glad I choose to work on
attitudes towards many things which made it very easy and enjoyable to work with you. electrosynthesis when we got the ROXY in 2010. Thanks to Agnieszka, Jean-Pierre and
Dear Henk: Thanks a lot for the lessons in teaching, academic politics and Dutch humour. Hendrik-Jan from Antec for sharing their know-how, the nice conversations and the quick
Thanks also for letting me try myself at lecturing, for just talking and for sharing my fond- and uncomplicated support which meant a lot to me as a novice in electrochemistry.
ness of cakes. I do not want to forget my collaborators outside of the TIPharma project who made a big
My gratitude goes to my Paranimfen Linda and Marija for supporting me on this last stretch difference to this thesis. Thanks to Anna-Jo and Arno for the exciting experiments (finally
of the road. Thank you, Linda, for the chance to see the ceremony from the podium and for a chance to do some NMR in my PhD) and the hospitality at the RUN in Nijmegen. Nils,
keeping the watch with Ferry and me during the summer of 2011. Thank you, Marija, for Christophe and Bruce at UC Davis for the fruitful collaboration on soluble epoxide hydro-
bringing life back to the group after that and the fun evenings in Rotterdam. Thanks also to lase. Thanks to Daniel, Iwona and Martin at DSM Resolve for giving me an excuse to do
Jon for the smooth entrance into a project running at full speed, for all the collaborations some practical work in my last year. Thanks to Bert and Mario for the second batch of p38α.
and for passing on a marvellously well maintained LC–MS/HRS system. I do not know what I would have done without it. Thanks, Guido for the discussion and
AP My thanks are also due to everyone who was part of the group (ACAS, BMA, BAS, etc...)
for help, chatting about science or other important issues and generally creating a pleasant
updates on p38. Andreas and Frans, I thank you for your NMR measurements of my ridicu-
lously low concentrated samples. Thanks to Anne, Martin, Uwe and all the other analytical
AP
work atmosphere. Thanks to Ansgar for being a fellow spirit in games, books and music, chemists from Münster for keeping in touch, enjoying ASMS 2010 together and discussions
something I sorely needed when I started. Ferry, with you the weeks never got boring. about electrochemistry. Thanks to Dick, Klaas and Roald for fixing everything the students
Thanks for the biking lessons ‘Amsterdam style’ and for carrying the sEH project the last and I could break and making everything we could think of.

222 223
My new colleagues at the LUMC deserve a thank you as well for making me feel welcome,
for patiently teaching me a new science and for listening to my complaints about my ‘sec-
ond job’. Agnes, Albert, Anton, Aswin, Bas, Bart, Carolien, Cees, Dana, Dennis, Dick-Paul,
Florent, Frank, Karli, Linda, Genadij, Guinevere, Hans, Manfred, Manu, Marco, Martin,
Maurice, Simone, Oleg, Rico, Rosina, Stephi, Toma, Vincenco, Yoann and Yuri: Thank you.
The challenge, excitement and frustration of a PhD thesis cannot be weathered without an
intact social life. Although I was also friends with many of my colleagues, it was good to
have friends outside of work, to share my ups and downs from a different perspective and
to forget work for a couple of hours here and there. To all the tennis and Jugger players, the
gamers, the (ex-)Booking.com people and the ones left behind in Germany: Yes, I mean
you. Thank you. For my cousins, aunts, uncles and grandparents, I could say the same as
I said for my friends except that they have already known and influenced me quite a while
longer. Für meine Cousins, Tanten, Onkel und Großeltern: Vielen Dank, dass ihr so getan
habt, als würde es euch interessieren. Vielen Dank für das gute Essen und alles andere.
Georg, my little brother: Thank you for listening. I am really happy you choose a similar
route as me and we have so many things to talk about these days.
My parents: How to find the right words? An meine Eltern: Ihr habt mich immer unterstützt
in meiner Neugier und auf der Suche nach meinem eigenen Weg. Ihr habt mir in genau
den richtigen Portionen Freiheit und Sicherheit gegeben. Ich weiß, das ist nicht selbstver-
ständlich. Euer Anteil daran, dass ich es soweit gebracht habe, ist wahrscheinlich kaum zu
überschätzen. Alles was ich tun kann ist aus tiefstem Herzen Danke zu sagen.
However, Lisa, you had the most non-scientific influence to get me here. You already sup-
ported me through the ups and downs of my studies and you gave me the courage to take
the risk and move to Amsterdam. I am very glad it turned out so well for the both of us. You
hardly complained about the business trips, the long days or the weekends, but you were
not afraid to tell me when it was enough. For both, I am thankful. Most of all, I am very
grateful for your inexhaustible patience and your loving support.

AP

224

You might also like