Sunteți pe pagina 1din 9

Affinity selection-mass spectrometry screening techniques

for small molecule drug discovery


D Allen Annis, Elliot Nickbarg, Xianshu Yang, Michael R Ziebell
and Charles E Whitehurst

Affinity selection-mass spectrometry (AS-MS) techniques use MS as the detection system are of particular interest
assess the binding of candidate molecules to immobilized or because of the exquisite sensitivity and unique selectivity
soluble receptors, and these methods are gaining acceptance possible with MS. In some screening formats, the selec-
in high throughput screening laboratories as valuable tivity achieved by MS enables direct analysis of compound
complements to traditional drug discovery technologies. A mixtures such as combinatorial libraries and unpurified
diversity of receptor types have been evaluated by AS-MS, natural products extracts an advantage that is not easily
including those that are difficult to screen using traditional achieved using other analytical methods. MS-based
biochemical approaches. AS-MS techniques that couple liquid screening is primarily implemented in two ways: by
chromatography-MS with size-based separation methods, monitoring the functional output of a receptor-dependent
such as ultrafiltration, gel permeation, or size-exclusion biochemical reaction, or by using affinity-based methods
chromatography, are particularly amenable to the demands of that directly assess binding of a candidate molecule to its
MS-based screening and have demonstrated the greatest target receptor. Because of their expanding popularity,
success across a broad range of drug targets. MS both of these styles of MS-based drug discovery have been
measurements of receptor function have many of the same the subject of several timely reviews, special issues of
advantages as AS-MS screening and are increasingly used for topical journals, and the focus of at least two recent books
drug discovery as well. [2,3,47]. Herein we report on the recent contributions to
Addresses the field of MS-based drug discovery, with special empha-
Schering-Plough Research Institute, 320 Bent Street, Cambridge, sis on lead identification methods that couple an affinity
MA 02139, United States selection step with MS confirmation of receptorligand
binding.
Corresponding author: Annis, D. Allen (allen@allenannis.com),
Nickbarg, Elliot (elliot.nickbarg@spcorp.com), Yang, Xianshu
(xianshu.yang@spcorp.com), Ziebell, Michael R. Affinity selection-MS: direct detection of
(michael.ziebell@spcorp.com) and Whitehurst, Charles E. receptorligand complexes
(charles.whitehurst@spcorp.com) Affinity selection-MS (AS-MS) methods directly or
indirectly measure binding of small molecules to their
Current Opinion in Chemical Biology 2007, 11:518526 biomolecular target, and all varieties of AS-MS include
the following steps: (i) an affinity selection stage, where
This review comes from a themed issue on the protein is equilibrated with one or more potential
Analytical Techniques
Edited by Peter M Fischer
ligands, allowing the protein to form a complex with any
compound capable of binding; (ii) the resulting receptor
ligand complexes are separated from non-binding mix-
ture components; and (iii) ligands are identified by MS or
1367-5931/$ see front matter
# 2007 Elsevier Ltd. All rights reserved. MSMS.

DOI 10.1016/j.cbpa.2007.07.011 Direct AS-MS methods (Table 1) separate proteinligand


complexes from unbound components within the mass
spectrometer, then measure the mass of the non-covalent
Introduction or (less commonly) the covalent proteinligand complex in
Mass spectrometry (MS) is one of the most powerful the gas phase. Indirect methods (Table 2) use a separation
analytical techniques in modern science, and it plays a technique, typically chromatographic in nature, to isolate
key role in nearly every stage of the drug development the proteinligand complex from unbound components
process [1]. Recent advances in MS instrument design, before MS analysis. Both direct and indirect varieties of
especially the development of electrospray ionization AS-MS have been developed in an assortment of hardware
(ESI), have extended the reach of MS to the lead discovery configurations and successfully applied to screening com-
stages of drug development as well. Modern pharmaceu- pound mixtures, including combinatorial libraries and
tical discovery relies increasingly on target-based screening unpurified natural products extracts.
techniques to identify new lead compounds for receptors
that have known or suspected involvement in a disease Researchers at Sunesis developed a fragment-based, direct
pathway. High-throughput screening (HTS) methods that AS-MS technique to identify compounds covalently bound

Current Opinion in Chemical Biology 2007, 11:518526 www.sciencedirect.com


Affinity selection-mass spectrometry screening techniques for small molecule drug discovery Annis et al. 519

Table 1

Direct AS-MS screening approaches

Class Method Description Advantages Disadvantages


Covalent Fragment-based Ligand fragments bind Fragment library permits Requires a reactive side chain
complexes lead assembly covalently to the target interrogation of a large in vicinity of binding site.
and are identified by MS portion of chemical Assembled fragments may
diversity space not generate active compounds
Noncovalent Nano-electrospray-MS Gentle ionization High sensitivity permits Small molecule library size is
complexes technique enables covalent low compound library limited and HTS not yet
complexes to be preserved concentrations. A high demonstrated. Gas phase
in the gas phase density chip at the ESI measurements may not reflect
source increases throughput. biological interactions
Small sample volumes
Multi-target affinity/ Direct MS detection of Multiple targets may be Mass envelope of ligand-target
specificity screening noncovalent RNA-ligand screened in the same complex may not be
(MASS) complexes sample. Potential to observable by spectrometer
identify low affinity cmpds
missed by other approaches
Detection of Direct MS detection of Used as counter-screen to No information on specific
oligonucleotide-ligand ligands bound to single identify ligands with high oligonucleotide sites of interaction
complexes by ESI-MS or double stranded affinity to DNA. Automated
(DOLCE-MS) oligonucleotides sample prep permits >1000
samples/week
ESI-electron capture The sites of ligand-target Generates data on contact Useful primarily for high
dissociation contacts may be sites as well as putative affinity interactions. May
investigated using ECD mechanisms of binding. not work for large complexes
for complexes that remain Appropriate for ligand where conformation in the
intact in the gas phase optimization efforts gas phase is altered

to a target protein [8,9]. In their site-directed ligand recently published by Hofstadter and co-workers on
discovery approach, specific amino acids on a protein the detection of small molecule DNA and molecule
surface are modified to facilitate tethering of small mol- RNA binding by multi-target affinity/specificity
ecule building blocks (fragments). The protein guides screening, or multi-target affinity/specificity screening
fragment assembly within a binding pocket to select those (MASS) [13]. Zhang and co-workers have commercia-
building blocks with the best binding characteristics. The lized a chip-based ESI-MS system and demonstrated
resulting covalent proteinligand complexes are directly its utility for studying proteinligand interactions [14].
analyzed by MS, and the measured shift in molecular mass An especially useful feature of this system is its
indicates which building blocks yield the best ligand. ability to quantify binding affinities using a minimal
Medicinal chemistry efforts, often in conjunction with amount of purified receptor [15]. As an added benefit,
X-ray crystallographic analysis, are subsequently employed Loo and co-workers have shown that electron capture
to convert the covalently bound fragments into a non- dissociation analysis of the proteinligand complex
covalent ligand with better affinity and molecular proper- inside a Fourier transform ion cyclotron mass spec-
ties than the progenitor fragments. trometer can reveal the binding site of a small molecule
on its receptor [16].
AS-MS techniques that directly interrogate non-
covalent proteinligand complexes are of considerable ESI-MS methods for detecting receptorligand binding
value because the resulting leads are inherently more are appealing because they give direct evidence of the
drug-like than those arising from covalent approaches. existence of a complex. However, conflicting data on the
These methods require that the non-covalent com- correlation of gas-phase affinity measurements with
plexes survive the transition to the gas phase, and in solution-phase interactions have been shown [17], in-
1991 Ganem and Henion presented the first conclusive cluding reports by researchers using a new, laminar flow
evidence that the immunosuppressant FK506 and its technique that estimates receptorligand affinities by
biomolecule target FKBP can be directly observed as a ESI-MS while not requiring that non-covalent inter-
gas-phase complex using ESI-MS as a gentle ioniz- actions are preserved in the gas phase [18,19]. Also, these
ation technique [10]. Since this seminal report, many and other related direct AS-MS techniques do not tolerate
similar non-covalent receptorligand interactions have high concentrations of non-volatile salts, buffers, cofac-
been identified and characterized using ultra-sensitive tors, metal ions, or detergents that may be necessary for
nanospray ESI-MS [11,12]. An excellent review was proper protein folding and stability.

www.sciencedirect.com Current Opinion in Chemical Biology 2007, 11:518526


520 Analytical Techniques

Table 2

Indirect AS-MS screening approaches

Class Method Description Advantages Disadvantages


Size exclusion Automated ligand Target-ligand complex Large mixtures permit HTS. Samples are processed
chromatography identification captured by fast sec >10 000 compounds linearly (6 min/sample)
and gel permeation system (ALIS) followed by integrated screened per hour per limiting HTS of individual
chromatography LCMS ligand identification instrument compounds
SpeedScreen Target-ligand separation 96-well plates permit HTS Total organic content in
using 96-well SEC plates (400 000 cmpds/week) when bed volume is limited.
followed by LCMS screened in pools of 400 Receptor-library separation
compounds is not directly visualized
Solid phase- Frontal affinity Compounds infused through Generates affinity ranking Protein must be modified
immobilized target chromatography a column having a target for binders. Controls for for attachment. Library
with MS detection immobilized on the solid differences in MS sensitivity size is limited by elution
phase. Weak binders elute between cmpds. (10 000 overlap and competition
early, strong binders elute later compounds/day)
Affinity capture-MS Compounds combined with Permits interrogation of Protein must be modified
bead-bound target followed protein complexes as for attachment.
by washing, elution and well as poorly solubilized Background possible due
detection by ES-MS/MS receptors to nonspecific binding
Affinity capillary Target-ligand complexes are Flexible assay format. Ligand must modulate
electrophoresis-MS separated from other Potentially useful for a the target electrophoretic
components by electrophoretic range of targets. Short mobility. Requires volatile
mobility and bound ligands sample runtimes solvents to accommodate
identified by MS MS
Separation by Ultrafiltration-MS Enrichment of target-ligand Large library mixtures are Multiple steps and
ultrafiltration complexes by pressure-based possible increasing the incomplete separation
ultrafiltration, then ligand throughput to >200 000 of bound and free
dissociation & detection by compounds in 2 months ligands and possibility
ESI-MS of background noise
Pulsed Ligand-target complex retained Applicable for targets Ligand binding may be
ultrafiltration-MS on membrane is extracted and with poor chromatography disrupted by target
ligands are dissociated and characteristics when tried interactions with the
identified by flow-injection MS with other separation methods membrane. Washing
required

Affinity selection-MS: indirect (hyphenated) Brennan and co-workers have immobilized nicotinic
methods acetylcholine receptors on monolithic FAC columns
Immobilized ligand or receptor and confirmed the activity of the bound receptor using
In part to enable the use of non-volatile buffers in the epibatidine as control ligand. In this manner they used
receptorligand binding reaction, indirect AS-MS screen- FAC-MS to optimize protein immobilization methods for
ing methods have been developed that couple a separ- on-line enzyme assays [24,25]. Researchers at Protana
ation technique with ESI-MS detection. The simplest have also reported FAC-MS as a global kinase-binding
variants are those where one binding partner, either the assay with a throughput of up to 10 000 compounds per
receptor or the small molecule ligand, is immobilized on a day [26]. They demonstrated their method by discovering
solid support. Familiar non-MS methods infer receptor inhibitors of the EphB2 tyrosine kinase receptor, and
ligand interactions by measuring changes in surface plas- described other applications of the platform in a recent
mon resonance (Biacore) or wavelength shift (Cornings review [27,28]
EPIC and SRU BioSystems BIND platforms) [20,21].
MS-based techniques detect ligands that have been cap- Affinity capillary electrophoresis (ACE) is a well-estab-
tured by an immobilized receptor; for example, Schriemer lished technique for drug discovery from complex mix-
and co-workers have developed a frontal affinity chroma- tures, including natural products [29]. Despite improved
tography (FAC)-MS platform for lead discovery. In sensitivity and selectivity by using MS as a detection
FAC-MS, a sample containing a set of ligands and a system for ACE, it requires the use of volatile buffers in
non-binding marker compound are passed through a order to interface with MS, so this application is not
column onto which a receptor has been attached. The routinely used [30].
non-binding marker compounds elutes in the void
volume, while each ligand elutes at a later time depend- Affinity chromatography techniques, including ACE-MS
ing on its affinity for the immobilized receptor [22,23]. and FAC-MS, work well for analyzing compound mixtures,

Current Opinion in Chemical Biology 2007, 11:518526 www.sciencedirect.com


Affinity selection-mass spectrometry screening techniques for small molecule drug discovery Annis et al. 521

and can also yield quantitative binding affinity estimates these latter two reports are interesting examples of ultra-
for a variety of receptor types. Additionally, the affinity filtration-based AS-MS, as described these methodologies
columns can be reused. However, these methods require are not immediately amenable to HTS.
that the receptor be immobilized, which can affect its
binding behavior relative to the native, soluble target, Gel permeation chromatography (GPC) spin columns are
and may cause false positives from compounds binding also used to separate receptorligand complexes from
non-specifically to the stationary phase or linker system. unbound ligands for AS-MS, and Siegel recently provided
While direct MS analysis of the affinity column is chal- a detailed review of this approach as practiced by Amgen,
lenged by nonvolatile buffers, integration with matrix- Novartis, and Wyeth Pharmaceuticals [41]. A limitation of
assisted laser desorption/ionization mass spectrometry this approach is that spin-column handling and centrifu-
(MALDI-MS) may overcome this limitation [31]. gation may be more difficult to automate and scale than
other methods; however, overall throughput can be
Solution-based methods increased by pooling the eluants from several spin col-
Solution-based AS-MS methods do not require immobil- umns before ESI-MS analysis. Mayr recently reviewed
ization of the target receptor or ligands on a solid support. the successful use of SpeedScreen, an AS-MS method
This feature avoids the alteration of receptor properties developed at Novartis that uses a centrifuged 96-well
by tagging or chemical linkage, and it allows greater plate assembly of GPC columns, including a top sample
compound diversity since immobilizing functional groups loading plate with pinholes in the bottom of each well; a
is not necessary. The primary challenge for solution- central filter plate pre-loaded with GPC resin (Sephadex,
based AS-MS methods is the development of efficient Pharmacia); and a bottom collection plate for the capture
techniques to separate receptorligand complexes from of effluents containing separated receptorligand com-
unbound small molecules. plexes [42]. Brown et al. reported on the chemical nature
of hits identified from the SpeedScreen-based HTS of 26
Separation technologies that use ultrafiltration mem- targets [43]. They concluded that SpeedScreen yields
branes are commonly used to isolate high molecular compounds with drug-like properties, albeit with greater
weight biomolecules (>10 kDa proteins and DNA) from lipophilicity and higher molecular weights than the pro-
small molecules. Researchers at Abbot used ultrafiltration genitor compound screening library. The authors empha-
to develop an AS-MS screening method by (i) combining sized the value of using cheminformatic-based filters to
a library of compounds with a protein, then (ii) separating eliminate promiscuous binders, false positives, and dis-
non-binders by pressure-based ultrafiltration, (iii) re- favored chemotypes from primary hit lists, and further
equilibrating with library-free buffer and re-separating emphasized the importance of hit validation by rigorous
by ultrafiltration to enrich higher affinity ligands, and (iv) follow-up SpeedScreen assays.
denaturing the retained proteinligand complexes and
analyzing the freed ligands by liquid chromatography Size exclusion chromatography (SEC) separates protein
(LC)MS [32,33]. These researchers identified hits to ligand complexes from non-binding small molecules by the
the important oncology targets Bcl-xL and the mamma- same principles as GPC. However, SEC holds several
lian checkpoint kinase CHK1, as well as the anti-infective advantages relative to the spin-column method, including
target Streptococcus enzyme MurF and the oncology target its amenability to automation and the ability to directly
MetAP2 [34,35,36,37]. A disadvantage of ultrafiltration is couple the SEC stage with an LCMS system for ligand
that unbound compounds are not completely removed detection. The first fully integrated SEC-LCMS platform
unless subsequent washing steps are employed, which for HTS was developed by researchers at NeoGenesis
can confound MS detection of true ligands unless they are (later acquired by Schering-Plough) and dubbed the auto-
significantly enriched relative to chemical noise caused mated ligand identification system, or ALIS [44]. The
by unbound compounds. In other reports, Sun and van ALIS process includes the following steps (see Figure 1): a
Breemen investigated the relative binding of compounds target receptor plus compound mixture are first combined
to estrogen receptors using ultrafiltration-MSMS [38]. in a 96-well plate format and allowed to reach equilibrium,
Hannewald et al. reported that centrifugal ultrafiltration then the plate is loaded into a chromatography system
followed by MALDI-MS analysis of the retentate fitted with a reusable SEC column containing a proprietary
enabled the identification of the control compounds resin for rapid separation (<20 s) of target-ligand com-
colchicine, vinblastine and vincristine binding to their plexes from unbound pool components. Proteinligand
target receptor tubulin [39]. Cheng and van Breemen also complexes in the SEC eluant are monitored by UV detec-
reported an unorthodox, low-throughput AS-MS screen- tion, and an automated valving system directs the protein
ing approach where a ligand-induced increase in target peak to a reverse-phase chromatography column for dis-
solubility, in this case A140 from amyloid precursor sociation, desalting, and elution of any ligands into an ESI-
protein, was measured by ultrafiltration-based separation MS system for identification. The ALIS platform has been
of soluble from aggregated A140 followed by detection used to screen a variety of target classes, including integral
of soluble A140 using flow injection ESI-MS [40]. While membrane proteins (Table 3). A collaboration between

www.sciencedirect.com Current Opinion in Chemical Biology 2007, 11:518526


522 Analytical Techniques

Figure 1

Schematic diagram of the ALIS process.

NeoGenesis and Merck yielded a novel inhibitor for the the quality of different protein preparations. ALIS
protease -Secretase, an important target for Alzheimers enables the simultaneous detection of multiple target-
disease therapy [45]. Whitehurst et al. reported on the use bound ligands in a single binding reaction, allowing the
of ALIS to screen the muscarinic M2 acetylcholine re- determination of allosteric or isosteric competitive bind-
ceptor (M2R), a G-protein coupled receptor (GPCR) ing profiles versus known ligands or cofactors, while also
[46]. This report was the first to describe screening an providing mixture-based affinity measurements that
integral membrane receptor by solution-based AS-MS [47]. enable potency optimization and the development of
structureactivity relationships directly from combinator-
Beyond HTS, the ALIS platform has been demonstrated ial chemical libraries [49]. These ALIS-based analytical
to be a valuable analytical tool for characterizing protein techniques were highlighted in the report of the ALIS-
ligand interactions [48]. For example, saturation bind- based screening of M2R, where ALIS was applied to
ing assays can be performed where a receptor preparation three phases of lead discovery: (i) biochemical validation
at constant concentration is titrated with increasing con- of the target before HTS screening; (ii) HTS screening of
centrations of a ligand and then analyzed by ALIS to a diverse combinatorial small molecule library; and (iii)
provide absolute binding affinity estimates and to assess the characterization of newly discovered hits to determine

Table 3

Protein types screened by solution-based AS-MS

SEC-RPC-ESI-MS GPC Spin FAC-ESI-MS Ultrafiltration-ESI-MS


column-ESI-MS or -MALDI-TOF-MS
Schering-Plough, Amgen, Novartis, Protana Abbott, Van
Merck Wyeth Breemen
Kinases U U U U
Proteases, peptidases U U U U
Other enzyme classes U U U U
Nuclear hormone receptors U U U U
Protein complexes U
GPCRs, integral membrane proteins U
Proteinprotein interaction targets U U U U
Transcription factors U U

Current Opinion in Chemical Biology 2007, 11:518526 www.sciencedirect.com


Affinity selection-mass spectrometry screening techniques for small molecule drug discovery Annis et al. 523

if any are interesting leads [46]. Interestingly, the ALIS- quantifying the bound marker after sequential filter iso-
based screening of M2R resulted in the discovery of both lation and liberation steps. Fragmentation of the marker
orthosteric antagonists and a putative allosteric modu- in a triple quadrupole MS and quantitation of the result-
lator, consistent with the proposed capability of AS-MS to ing MSMS ions relative to an isotopically labeled (but
discover ligands with various binding modes for a receptor non-radioactive) standard yielded excellent reproducibil-
[50]. ity and good correlation with results from a traditional
radioligand analysis [55].
To date, Schering-Plough is the only company to have
reported the routine use of on-line SEC-LCMS for Throughput can also be a challenge for MS-based func-
HTS. However, several proof-of-concept studies have tional screening. Despite the advent of multi-head injec-
been reported recently that demonstrate other appli- tors for sequential switching between LC-purified
cations of on-line SEC-LCMS. For example, Flarakos samples, MS is an inherently serial analysis technique,
and Vouros reported that custom-made SEC columns since only one sample at-a-time can be introduced to the
packed with Sephadex G-25 media (to mimic centrifugal mass analyzer. (This contrasts with UV- or fluorescence-
spin-columns) could be connected by an on-line switch- based spectroscopic techniques, where array detectors
ing system to UV-based detectors, enabling protein allow simultaneous measurement from many samples.)
ligand complex monitoring and capture for LCMS To overcome this limitation, researchers at BioTrove have
detection of bound ligandsin this case, compounds that developed an ultra-fast sample switching inlet for a triple-
bound to human serum albumin [51]. These investigators quadrupole mass spectrometer, and commercialized its
reported that improvements in miniaturization are application for monitoring enzyme reaction products by
required to reduce protein and compound consumption LCMS. Their Rapid Fire system can analyze over
in their system to enable HTS. Researchers at Merck 10 000 samples in a day, and was demonstrated by the
recently described an integrated SEC-LCMS platform discovery of several potent inhibitors of acetylcholinester-
to affinity-rank ligands present in mixtures by competi- ase, the Akt1 kinase, and the membrane-associated protein
tive affinity selection to dipeptidyl peptidase IV [52]. phosphatidylserine decarboxylase (PISD) [56,57,58].
They showed that the relative affinities of the ligands
determined by their AS-MS technique corresponded well In another approach to automated MS-based enzyme
with the known relative biochemical IC50 values of the assays, Brennan and co-workers have developed a sol
ligands, but did not report on HTS efforts. gel entrapment system to immobilize proteins for on-line
inhibitor screening [59]. By eluting enzyme substrates
MS measurements of receptor function plus possible inhibitors over the trapped proteins, these
MS-based techniques that monitor functional assays have researchers measure the effect of the inhibitor on enzyme
advantages that parallel those of AS-MS. Target-based activity or receptor binding, as exemplified by a screen of
functional assays identify lead compounds by measuring 49 compounds against adenosine deaminase (ADA) with
the outcome of a biomolecule-mediated transformation, IC50 determination for active inhibitors. They have
such as production or depletion of the starting materials or demonstrated the entrapment technique for a variety
products of an enzymatic reaction, or displacement of a of target systems, including membrane proteins such as
labeled marker compound from a biomolecular receptor. the nicotinic acetylcholine receptor (nAChR) and dopa-
These measurements are traditionally done using radio- mine D2 receptor [60].
chemical methods, fluorescence readouts, or UV spec-
troscopy. By using MS analysis, the products of enzymatic Enzyme inhibitors have also been characterized in high-
reactions or displaced ligands can be directly identified by throughput fashion by MALDI-MS techniques, in-
virtue of their molecular mass, which eliminates chemical cluding desorption/ionization on silicon (DIOS-MS)
interferences and obviates the need for radioactive or [61] and from carbon nanotube-based matrices [62]. A
fluorescent labeling of compounds or marker agents. particular advantage of the MALDI approach is demon-
strated by its application in kinase assays. Traditionally
A considerable challenge to MS-based functional screen- the phosphorylation of a protein or substrate is monitored
ing is that most assays require high concentrations of salts, by incorporation of radioactive phosphate into the pro-
buffers, cofactors or detergents, which are nonvolatile duct. MALDI-MS allows modification of the native sub-
components that interfere with MS analysis coupling strate even whole proteins to be monitored directly
LC with the mass spectrometer enables nonvolatile com- without the use of a labeled probe [63]. Unfortunately,
ponent removal, and Wanner and co-workers used this these approaches are limited to use with volatile buffers,
capability to develop competitive MS-binding assays for example, ammonium carbonate.
[53,54]. In this system, LCMS detection is used to assess
the ability of test compounds to compete a native (non- Conclusions and future directions
labeled) marker ligand from its pharmacological receptor, As shown in this report, the pharmaceutical industry
by either analyzing the free (unbound) marker or by is enthusiastically using MS-based HTS methods to

www.sciencedirect.com Current Opinion in Chemical Biology 2007, 11:518526


524 Analytical Techniques

discover and characterize new lead compounds, and we 12. Sinz A: Investigation of Proteinligand Interactions by Mass
Spectrometry. Chem Med Chem 2007.
anticipate these applications will continue and expand in
13. Hofstadler SA, Sannes-Lowery KA: Applications of ESI-MS in
the future. Presently, no one-size-fits-all AS-MS solution  drug discovery: interrogation of noncovalent complexes.
is expected to satisfy all future screening demands, and no Nat Rev Drug Discov 2006, 5:585-595.
commercial product specifically tailored for AS-MS This report provides a comprehensive overview of affinity selection-MS
techniques for discovering DNA and RNA ligands. The authors provide
screening is available to interested researchers. Rather, examples from their laboratories that demonstrate drug discovery by
a diversity of methods is implemented by different com- direct MS detection of noncovalent drug-target complexes.
mercial and academic laboratories, each with its particular 14. Zhang S, Van Pelt CK: Chip-based nanoelectrospray mass
spectrometry for protein characterization. Expert Rev
advantages and disadvantages, suggesting that no single Proteomics 2004, 1:449-468.
technique can solve the broad range of requirements that
15. Zhang S, Van Pelt CK, Wilson DB: Quantitative determination of
modern pharmaceutical discovery requires. As analytical noncovalent binding interactions using automated
instrumentation becomes miniaturized and more auto- nanoelectrospray mass spectrometry. Anal Chem 2003,
75:3010-3018.
mated, especially MS and LC components, their ease of
use for non-specialists will naturally allow more wide- 16. Xie Y, Zhang J, Yin S, Loo JA: Top-down ESI-ECD-FT-ICR mass
spectrometry localizes noncovalent proteinligand binding
spread and creative applications and may expand the sites. J Am Chem Soc 2006, 128:14432-14433.
already considerable impact of AS-MS on drug discovery.
17. Raji MA, Frycak P, Beall M, Sakrout M, Ahn JM, Bao Y,
Armstrong DW, Schug KA: Development of an ESI-MS
Acknowledgements screening method for evaluating binding affinity between
We thank Satish Jindal, Jerry Shipps, Arshad Siddiqui, and our integrin fragments and RGD-based peptides. Int J Mass
colleagues at SPRI for critical review of this manuscript. Spectrom 2007, 262:232-240.
18. Clark SM, Konermann L: Determination of ligand-protein
References and recommended reading dissociation constants by electrospray mass spectrometry-
Papers of particular interest, published within the annual period of based diffusion measurements. Anal Chem 2004,
review, have been highlighted as: 76:7077-7083.

 of special interest 19. Clark SM, Konermann L: Screening for noncovalent ligand-
 of outstanding interest receptor interactions by electrospray ionization mass
spectrometry-based diffusion measurements. Anal Chem
2004, 76:1257-1263.
1. Korfmacher WA: Principles and applications of LCMS in new
drug discovery. Drug Discov Today 2005, 10:1357-1367. 20. Makara GM, Athanasopoulos J: Improving success rates for
lead generation using affinity binding technologies. Curr Opin
2. Lee MS (Ed): Integrated Strategies for Drug Discovery Using Mass Biotechnol 2005, 16:666-673.
Spectrometry, Hoboken, NJ: Wiley-Interscience; 2005.
21. Cunningham BT, Laing L: Microplate-based, label-free
3. Wanner K, Hofner G (Eds): Mass Spectrometry in Medicinal
detection of biomolecular interactions: applications in
 Chemistry, Weinheim: Wiley-VCH; 2007.
proteomics. Expert Rev Proteomics 2006, 3:271-281.
This timely book includes contributions from several leaders in the field of
affinity selection-mass spectrometry and MS-based drug discovery, plus 22. Ng ES, Yang F, Kameyama A, Palcic MM, Hindsgaul O,
sections on modern MS instrumentation, principles, and applications in Schriemer DC: High-throughput screening for enzyme
pharmacokinetics. inhibitors using frontal affinity chromatography with liquid
4. Schermann SM, Simmons DA, Konermann L: Mass chromatography and mass spectrometry. Anal Chem 2005,
77:6125-6133.
spectrometry-based approaches to proteinligand
interactions. Expert Rev Proteomics 2005, 2:475-485. 23. Chan N, Lewis D, Kelly M, Ng ESM, Schriemer DC: Frontal affinity
5. Deng G, Sanyal G: Applications of mass spectrometry in early chromatography mass spectrometry for ligand discovery
stages of target based drug discovery. J Pharm Biomed Anal and characterization. In Mass Spectrometry in Medicinal
2006, 40:528-538. Chemistry. Edited by Wanner K, Hofner G. Wiley-VCH; 2007:217-
246 . [Mannhold R, Kubinyi H, Folkers G (Series Editor): Methods
6. Geoghegan KF, Kelly MA: Biochemical applications of and Principles in Medicinal Chemistry.]
mass spectrometry in pharmaceutical drug discovery.
Mass Spectrom Rev 2005, 24:347-366. 24. Besanger TR, Hodgson RJ, Green JRA, Brennan JD: Immobilized
enzyme reactor chromatography: Optimization of protein
7. Hofstadler SA, Lee M (Eds): International Journal of Mass retention and enzyme activity in monolithic silica stationary
Spectrometry, Special Issue: Drug Discovery: Elsevier; 2004. phases. Anal Chim Acta 2006, 564:106-115.

8. Cancilla MT, Erlanson DA: Tethering: fragment-based drug 25. Besanger TR, Hodgson RJ, Guillon D, Brennan JD: Monolithic
discovery by mass spectrometry. In Mass Spectrometry in membrane-receptor columns: Optimization of column
Medicinal Chemistry. Edited by Wanner K, Hofner G. Wiley-VCH; performance for frontal affinity chromatography/mass
2007:305-320 . [Mannhold R, Kubinyi H, Folkers G (Series Editor): spectrometry applications. Anal Chim Acta 2006, 561:107-118.
Methods and Principles in Medicinal Chemistry.]
26. Slon-Usakiewicz JJ, Dai JR, Ng W, Foster JE, Deretey E,
9. Erlanson DA, Hansen SK: Making drugs on proteins: site- Toledo-Sherman L, Redden PR, Pasternak A, Reid N: Global
 directed ligand discovery for fragment-based lead assembly. kinase screening. Applications of frontal affinity
Curr Opin Chem Biol 2004, 8:399-406. chromatography coupled to mass spectrometry in drug
In this article Erlanson and Hansen describe the state-of-the-art in discovery. Anal Chem 2005, 77:1268-1274.
tethering approaches to drug discovery.
27. Slon-Usakiewicz JJ, Ng W, Dai JR, Pasternak A, Redden PR:
10. Ganem B, Li YT, Henion JD: Observation of noncovalent  Frontal affinity chromatography with MS detection (FAC-MS)
enzyme-substrate and enzyme-product complexes by in drug discovery. Drug Discov Today 2005, 10:409-416.
ion-spray mass spectrometry. J Am Chem Soc 1991, This overview of FAC-MS provides an in-depth discussion of its applica-
113:7818-7819. tions and compares the features of this important method with those of
other affinity selection-mass spectrometry techniques.
11. Ganem B, Henion JD: Going gently into flight: analyzing
noncovalent interactions by mass spectrometry. Bioorg Med 28. Slon-Usakiewicz JJ, Ng W, Foster JE, Dai JR, Deretey E, Toledo-
Chem 2003, 11:311-314. Sherman L, Redden PR, Pasternak A, Reid N: Frontal affinity

Current Opinion in Chemical Biology 2007, 11:518526 www.sciencedirect.com


Affinity selection-mass spectrometry screening techniques for small molecule drug discovery Annis et al. 525

chromatography with MS detection of EphB2 tyrosine kinase combinatorial libraries: Discovery of a novel antagonist of E.
receptor. 1. Comparison with conventional ELISA. J Med Chem coli dihydrofolate reductase. Int J Mass Spectrom 2004,
2004, 47:5094-5100. 238:77-83.
29. Gullo VP, McAlpine J, Lam KS, Baker D, Petersen F: Drug 45. Coburn CA, Stachel SJ, Li YM, Rush DM, Steele TG, Chen-
discovery from natural products. J Ind Microbiol Biotechnol Dodson E, Holloway MK, Xu M, Huang Q, Lai MT et al.:
2006, 33:523-531. Identification of a small molecule nonpeptide active site beta-
secretase inhibitor that displays a nontraditional binding
30. Schou C, Heegaard NH: Recent applications of affinity mode for aspartyl proteases. J Med Chem 2004, 47:6117-6119.
interactions in capillary electrophoresis. Electrophoresis 2006,
27:44-59. 46. Whitehurst CE, Nazef N, Annis DA, Hou Y, Murphy DM,
 Spacciapoli P, Yao Z, Ziebell MR, Cheng CC, Shipps GW Jr et al.:
31. Kovarik P, Hodgson RJ, Covey T, Brook MA, Brennan JD: Discovery and characterization of orthosteric and allosteric
Capillary-scale frontal affinity chromatography/MALDI muscarinic M2 acetylcholine receptor ligands by affinity
tandem mass spectrometry using protein-doped monolithic selection-mass spectrometry. J Biomol Screen 2006,
silica columns. Anal Chem 2005, 77:3340-3350. 11:194-207.
This report describes the first use of AS-MS for the discovery of agonists
32. Comess KM, Schurdak ME: Affinity-based screening and antagonists to a G protein-coupled receptor (GPCR), an important
techniques for enhancing lead discovery. Curr Opin Drug class of targets for modern drug discovery.
Discov Devel 2004, 7:411-416.
47. Whitehurst CE, Annis DA: Affinity selection-mass spectrometry
33. Cloutier TE, Comess KM: Library screening using ultrafiltration and its emerging application to the high throughput screening
and mass spectrometry. In Mass Spectrometry in Medicinal of G-protein-coupled receptors. Comb Chem High Throughput
Chemistry. Edited by Wanner K, Hofner G. Wiley-VCH; 2007:157- Screen, in press.
184 . [Mannhold R, Kubinyi H, Folkers G (Series Editor): Methods
and Principles in Medicinal Chemistry.] 48. Annis DA, Chuang C-C, Nazef N: ALIS: An affinity selection
 mass spectrometry system for the discovery and
34. Qian J, Voorbach MJ, Huth JR, Coen ML, Zhang H, Ng SC, characterization of proteinligand Interactions. In Mass
Comess KM, Petros AM, Rosenberg SH, Warrior U et al.: Spectrometry in Medicinal Chemistry. Edited by Wanner K, Hofner
Discovery of novel inhibitors of Bcl-xL using multiple G. Wiley-VCH; 2007:121-184 . [Mannhold R, Kubinyi H, Folkers G
high-throughput screening platforms. Anal Biochem 2004, (Series Editor): Methods and Principles in Medicinal Chemistry.].
328:131-138. This book chapter provides a definitive overview of ALIS as both a drug
35. Comess KM, Schurdak ME, Voorbach MJ, Coen M, Trumbull JD, discovery engine and a valuable tool for characterizing proteinligand
 Yang H, Gao L, Tang H, Cheng X, Lerner CG et al.: An interactions.
ultraefficient affinity-based high-throughout screening 49. Annis DA, Nazef N, Chuang CC, Scott MP, Nash HM: A general
process: application to bacterial cell wall biosynthesis enzyme technique to rank proteinligand binding affinities and
MurF. J Biomol Screen 2006, 11:743-754. determine allosteric versus direct binding site competition in
In this article, Commess and co-workers describe their GPC-spin column compound mixtures. J Am Chem Soc 2004, 126:15495-15503.
process for rapid screening of compound mixtures to discover inhibitors
of the important antibacterial target MurF. 50. Falb D, Jindal S: Chemical genomics: bridging the gap between
the proteome and therapeutics. Curr Opin Drug Discov Devel
36. Comess KM, Trumbull JD, Park C, Chen Z, Judge RA, 2002, 5:532-539.
Voorbach MJ, Coen M, Gao L, Tang H, Kovar P et al.: Kinase drug
discovery by affinity selection/mass spectrometry (ASMS): 51. Flarakos J, Morand KL, Vouros P: High-throughput solution-
application to DNA damage checkpoint kinase Chk1. J Biomol based medicinal library screening against human serum
Screen 2006, 11:755-764. albumin. Anal Chem 2005, 77:1345-1353.
37. Kawai M, BaMaung NY, Fidanze SD, Erickson SA, Tedrow JS, 52. Adam GC, Meng J, Athanasopoulos J, Zhang X, Chapman KT:
Sanders WJ, Vasudevan A, Park C, Hutchins C, Comess KM et al.: Affinity-based ranking of ligands for DPP-4 from mixtures.
Development of sulfonamide compounds as potent Bioorg Med Chem Lett 2007.
methionine aminopeptidase type II inhibitors with
antiproliferative properties. Bioorg Med Chem Lett 2006, 53. Zepperitz C, Hofner G, Wanner KT: MS-binding assays: kinetic,
16:3574-3577. saturation, and competitive experiments based on
quantitation of bound marker as exemplified by the GABA
38. Sun Y, Gu C, Liu X, Liang W, Yao P, Bolton JL, van Breemen RB: transporter mGAT1. Chem Med Chem 2006, 1:208-217.
Ultrafiltration tandem mass spectrometry of estrogens for
characterization of structure and affinity for human estrogen 54. Hofner G, Wanner KT: Competitive binding assays made easy
receptors. J Am Soc Mass Spectrom 2005, 16:271-279. with a native marker and mass spectrometric quantification.
Angew Chem Int Ed Engl 2003, 42:5235-5237.
39. Hannewald P, Maunit B, Muller JF: Tubulin-binding drug
screening by MALDI-TOFMS. Anal Chem 2006, 78:4390-4397. 55. Niessen KV, Hofner G, Wanner KT: Competitive MS binding
assays for dopamine D2 receptors employing spiperone as a
40. Cheng X, van Breemen RB: Mass spectrometry-based native marker. Chembiochem 2005, 6:1769-1775.
screening for inhibitors of beta-amyloid protein aggregation.
Anal Chem 2005, 77:7012-7015. 56. Ozbal CC, LaMarr WA, Linton JR, Green DF, Katz A, Morrison TB,
 Brenan CJ: High throughput screening via mass spectrometry:
41. Siegel MM: Drug screening using gel permeation a case study using acetylcholinesterase. Assay Drug Dev
chromatography spin columns coupled with ESI-MS. In Mass Technol 2004, 2:373-381.
Spectrometry in Medicinal Chemistry. Edited by Wanner K, Hofner Ultra-fast LCMS analysis enables the discovery of novel ligands to this
G. Wiley-VCH; 2007:65-120 . [Mannhold R, Kubinyi H, Folkers G very challenging protein target.
(Series Editor): Methods and Principles in Medicinal Chemistry.]
57. Forbes CD, Toth JG, Ozbal CC, LaMarr WA, Pendleton JA, Rocks
42. Mayr LM: Tackling the chemogenomic space by novel S, Gedrich RW, Osterman DG, Landro JA, Lumb KJ: High-
screening technologies. Ernst Schering Res Found Worksho throughput mass spectrometry screening for inhibitors of
2006:111-173. phosphatidylserine decarboxylase. J Biomolec Screen 2007,
Published Online May 3, 2007 as doi:10.1177/1087057107301320
43. Brown N, Zehender H, Azzaoui K, Schuffenhauer A, Mayr LM,
Jacoby E: A chemoinformatics analysis of hit lists obtained 58. Quercia AK, Myung J, LaMarr WA, Ozbal CC, Landro JA, Lumb KJ:
from high-throughput affinity-selection screening. J Biomol High-throughput screening by mass spectrometry:
Screen 2006, 11:123-130. comparison with the scintillation proximity assay with a
focused file screen of AKT1/PKB. J Biomolec Screen 2007,
44. Annis DA, Athanasopoulos J, Curran PJ, Felsch JS, Kalghatgi K, Published Online May 3, 2007 as doi:10.1177/1087057107300647
Lee WH, Nash HM, Orminati J-PA, Rosner KE, Shipps JGW et al.:
An affinity selection-mass spectrometry method for the 59. Hodgson RJ, Besanger TR, Brook MA, Brennan JD: Inhibitor
identification of small molecule ligands from self-encoded  screening using immobilized enzyme reactor

www.sciencedirect.com Current Opinion in Chemical Biology 2007, 11:518526


526 Analytical Techniques

chromatography/mass spectrometry. Anal Chem 2005, desorption/ionization on silicon (DIOS) platform.


77:7512-7519. Chembiochem 2004, 5:921-927.
This innovative, in-line method for monitoring enzyme reactions was used
to identify inhibitors for adenosine deaminase and directly determine the 62. Hu L, Jiang G, Xu S, Pan C, Zou H: Monitoring enzyme reaction
Ki values for the inhibitors. and screening enzyme inhibitor based on MALDI-TOF-MS
platform with a matrix of oxidized carbon nanotubes. J Am Soc
60. Besanger TR, Brennan JD: Entrapment of membrane proteins in Mass Spectrom 2006, 17:1616-1619.
solgel derived silica. J SolGel Sci Technol 2006, 40:209-225.
63. Greis KD, Zhou S, Burt TM, Carr AN, Dolan E, Easwaran V,
61. Shen Z, Go EP, Gamez A, Apon JV, Fokin V, Greig M, Ventura M, Evdokimov A, Kawamoto R, Roesgen J: Davis GF: MALDI-TOF
Crowell JE, Blixt O, Paulson JC et al.: A mass spectrometry plate MS as a label-free approach to rapid inhibitor screening. J Am
reader: monitoring enzyme activity and inhibition with a Soc Mass Spectrom 2006, 17:815-822.

Current Opinion in Chemical Biology 2007, 11:518526 www.sciencedirect.com

S-ar putea să vă placă și