You are on page 1of 9

Arch. Pharm. Chem. Life Sci. 2007, 340, 169 – 177 S.-Y. Han and S.-H.

Kim 169

Review Article
Introduction to Chemical Proteomics for Drug Discovery and
Development

Sung-Young Han1, 2, Seong Hwan Kim2

1
Department of Chemistry, University of Texas at Dallas, Richardson, TX, USA
2
Laboratory of Chemical Genomics, Bio-Organic Science Team, Drug Discovery Research Hub, Korea
Research Institute of Chemical Technology, Daejeon, Korea

A fundamental goal of chemical proteomics is to identify target proteins for bioactive small
molecules and then apply them to drug discovery and development as valid and drugable tar-
gets. Here, we introduce integrated technologies for the rapid identification of target proteins,
methodologies for validating them as drugable targets, and applications of chemical proteomics
in drug discovery and development.

Keywords: Photoreactivity / Rational drug design / Structure elucidation /

Received: September 19, 2006; accepted: February 1, 2007

DOI 10.1002/ardp.200600153

Introduction

Chemical proteomics is a currently emerging field with a


mission to identify and validate a target protein that
directly binds with a binder such as a bioactive small
molecule in a rapid, systematic, and comprehensive man-
ner by design, synthesis, and application of relevant che-
mical probes. As a multidisciplinary science, chemical
proteomics proposes the integration of tools and technol-
ogies from a variety of disciplines, chemistry, biochemis-
try, biology, structural biology, proteomics, and infor-
matics (Fig. 1). Thus, an integrated strategy in chemical
proteomics can provide the information about the bind-
ing site of bioactive small molecule in its target protein
and, importantly, it could be the starting point to
develop the drug screening systems based on the struc-
ture of target protein.
Mostly, the discovery of signal transduction pathway
contributing to the disease state has been done in the
first step of the processes for developing new drugs. The Figure 1. Chemical proteomics as a multidisciplinary science.
understanding of signaling pathways and further map-
ping of the key signaling molecules in biochemical path- ways have provided how cells, organs, and the body are
aberrantly controlled in disease state, and this had an
Correspondence: Seong Hwan Kim, Ph.D., Laboratory of Chemical
enormous impact on the development of new therapeu-
Genomics, Bio-Organic Science Team, Drug Discovery Research Hub, tic approaches for the treatment of nearly every human
Korea Research Institute of Chemical Technology, P. O. Box 107, Yu- disease [1]. Thus, the efforts to reveal the signaling path-
seong-gu, Daejeon 305-600, Korea
E-mail: hwan@krict.re.kr ways and molecules that are deeply involved in certain
Fax: +82 42 861-0307 disease states have continuously developed the mechan-

i 2007 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim


170 S.-Y. Han and S.-H. Kim Arch. Pharm. Chem. Life Sci. 2007, 340, 169 – 177

ism-based drug discovery, but it is not enough to develop this review, the importance of chemical proteomics and
new drugs for treating patients since the communication several tools used in this field will be introduced and,
among the disease-related signaling pathways is too finally, its application in the process of drug discovery
sophisticated to easily decipher the pathophysiological and development will be discussed briefly.
network of the disease-specific signals.
Actually, as well as the understanding of the cross-reac-
tion between a bioactive small molecule and its target Impact of chemical proteomics
protein(s) that can elucidate the biological actions of
both, the structural information about candidate target High-throughput screening assays used routinely is for
may prove useful in ultimately predicting the interaction identifying a bioactive small molecule that can bind its
of a bioactive small molecule with its non-targeted pro- target candidates and modulate their activities or func-
tein(s), which can lead to multiple side effects. Therefore, tions, but a very large proportion of the hits identified
the structure-based drug design approach based on the are false positives that do not bind to the target binding
,
understanding of what disease-related targets are’ in the site. This limitation can be overcome with the use of vali-
outset of the drug development process could cover the dated target proteins that are identified and evaluated by
shortage of mechanism-based drug discovery including chemical proteomics approaches.
unpredicted side effects. Chemical proteomics study on the target proteins of
As a rapid growth of structural biology for determining bioactive small molecules can also reveal alternative cel-
the protein structure and an advance in mass spectrome- lular modes of small molecule action, which was an
try (MS) analysis, a structure-based drug design approach unrecognized therapeutic potential previously. For
is considered a central component in the process of drug example, pyrido[2,3-d]pyrimidines, which were initially
discovery and development. In fact, advances in process developed as anti-proliferative agents targeting protein
automation and informatics have aided the development tyrosine kinases involved in cancer [7], were found to
of high-throughput X-ray crystallography and the use of lack selectivity for tyrosine kinases. Instead, these inhibi-
this technique for structure-based lead discovery [2, 3]. tors were established as potent inhibitors of several ser-
Thus, virtual screening coupled with high-throughput X- ine/threonine kinases such as p38a and RIPK2. The identi-
ray crystallography is now focused on identifying one or fication of alternative target proteins for bioactive small
more binding small molecule fragments from chemical molecules can provide new insights into their cellular
libraries consisting of hundreds of small molecule frag- modes of action. This could be highly associated with the
ments. The information about target proteins/peptides following; the identification of target proteins for an
can also make several other applications feasible. Two MS- approved medication by using chemical proteomics
based strategies, function-based and affinity-based, have approaches play a pivotal role in the process of finding
been employed in recent years for screening and evalua- new uses outside the scope of the original medical indica-
tion of compounds [4]. In the function-based approach, tion for existing drugs that is known as redirecting,
the effects of compounds on the biological activity of a tar- repurposing, repositioning, and reprofiling [8]. Reposi-
get molecule are measured, and in the affinity-based tioning success stories and companies leveraging reposi-
approach, compounds are screened based on their bind- tioning strategies are increasing in number, but it should
ing affinities to target molecules. Therefore, the elucida- not be discussed here.
tion of interaction between a binder (e. g. substrate for To summarize, the elucidation of the specific interac-
enzyme, ligand for receptor, or bioactive small molecule) tion between bioactive small molecules and their part-
and its target protein(s) could be a promising way to speed ners, target proteins by using integrated chemical proteo-
up and further industrialize target-based drug discovery mics strategies contributes to our understanding of how
[5]. On the other hand, the efforts in chemical proteomics bioactive small molecules work in cells and the body, and
would help to prioritize candidate targets, streamline the consequently to the development of medicines.
drug development pipeline, and hopefully reduce the
number of failures downstream.
With the fact, that chemical proteomics approaches Target proteins and binding sites
have gained more interests in the early stage of drug dis-
covery and development together with chemical geno- The biological function of most proteins depends on
mics that is to find and optimize chemical compounds direct and specific binding of their binders to particular
for its use to directly test the therapeutic relevance of spatial areas, the binding sites. Since the interaction of
new targets revealed through genome sequencing [6], in binders with target proteins affects their functional

i 2007 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.archpharm.com


Arch. Pharm. Chem. Life Sci. 2007, 340, 169 – 177 Chemical Proteomics in Drug Discovery 171

activities by changing the protein conformation, trigger- considered first, how its binding partners can be purified
ing (or blocking) the protein post-translational modifica- from the complex proteome. Several approaches such as
tion such as phosphorylation or competing with sub- the use of nanoparticle, photoaffinity probe, activity-
strates, the understanding of thermodynamics (or ener- based probe (ABP), and chemical affinity matrix have
getic stability) of interaction is important to elucidate been developed to purify/enrich and identify target pro-
the biological activities of small molecules in cells. teins/peptides that can directly interact with a bioactive
Binding site (or pocket) can be defined as the region of small molecule [11 – 13].
a protein that associates with a binder and it usually con-
sists of a cavity in the protein surface formed by a particu- Nanoparticles
lar arrangement of amino acids. Binding depends on for- Several forms of nanoparticles have been used in biologi-
mation of contacts between chemical groups of a binder cal separation. In particular, biomolecule-conjugated
and those in the binding site of the target protein. In par- gold nanoparticles (AuNPs) are the most popular probes
ticular, the cavity in the structure of a protein deter- because of their advantages such as the readily assem-
mines the recognition features of the target protein. bling with thiolated molecules, the large area/volume
Thus, the characteristics of a binding site can provide the ratio for investigating 3D interactions, and the ease of
missing link needed for a thorough understanding of the separation by centrifugation [14, 15]. Recently, a new
correlation between chemistry space and genome space. approach of using carbohydrate-encapsulated AuNP has
Recently, advanced and integrated methods for charac- been introduced [11]. This allows rapid mapping of carbo-
terizing functional binding sites from a three-dimen- hydrate-recognition peptide sequences in the target pro-
sional (3D) perspective have been developed and widely teins.
used; the variety of methods using computer algorithms Surface-modified magnetic nanoparticles can act as a
have been continuously developed and can be commer- general agent to separate, transport, and anchor a pro-
cially purchased from several companies such as Accelrys tein with several advantages such as their high surface/
Software, Inc. (http://www.accelrys.com) and Tripos, Inc. volume ratio, good solubility, faster movement/easy
(http://www.tripos.com). Using these methods, the virtual entry into cells, and magnetically controllable aggrega-
binding sites of proteins can be identified or a suffi- tion behavior that allow the intensive steps of centrifuga-
ciently reliable homology model can be built [9], but the tion omitted [16].
main limitation such as a high number of false positives
can not be excluded. This is the major bottleneck of in Photoaffinity probe
silico approaches. Much attention has been devoted to the application of
In order to upgrade the quality of the protein models, photoaffinity labeling to the study of small molecule-tar-
the effort to improve the experimental methods such as get protein interactions since photoaffinity techniques
X-ray crystallography and nuclear magnetic resonance are especially useful to elucidate the protein structure at
(NMR) has been continuously made. The recent advances the interface of bioactive small molecules by photoche-
in the automation of protein crystallization and in X-ray mically labeling interacting sites [17 – 19].
data collection/analysis have lead to the rapidly growing Conceptually, the structure of photoaffinity probes
number of protein structures collected in the Protein consists of four distinct functional elements; (a) a bioac-
Data Bank (PDB; http://www.pdb.org) [10]. Parallel with tive small molecule, (b) tags (such as biotin) for identify-
this effort, the experimental identification and valida- ing and/or purifying the probe-target protein complex,
tion of target proteins and/or binding sites by using inte- (c) a linker region, and (d) a photoreactive moiety capable
grated strategies in chemical proteomics should be also of covalently binding to target proteins (Fig. 2). Impor-
performed since the precise elucidation of interaction tantly, the design of chemical probes runs parallel to
between binders and their target candidates can help us structure – activity relationships (SAR) study because
to determine the crystal structure of proteins. probes should retain the biological activity as its parent
compound does [20, 21]. Generally, the introduction of a
photoreactive moiety and a biotin into the structure of a
Technologies for purifying/enriching target bioactive small molecule causes the decrease of biologi-
proteins/peptides cal activity, but probes with as much as 1000-times lower
activity can still be useful for investigating binder-pro-
For elucidating the physical mode of action for bioactive tein interactions [17].
small molecules identified by using cell-based, mechan- Biotin is a powerful tag for purifying the probe-target
ism-based or effect-based screening system, it should be protein/peptide complex based on the biotin-avidin inter-

i 2007 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.archpharm.com


172 S.-Y. Han and S.-H. Kim Arch. Pharm. Chem. Life Sci. 2007, 340, 169 – 177

Figure 2. Structure of a biotin-tagged photoaffinity probe. A bio-


tin-tagged photoaffinity probe consists of four distinct functional
elements, a bioactive small molecule, biotin, a linker region (e. g.
PEG or alkyl), and a photoreactive moiety (e. g. azide, diazirine,
or benzophenone).

action that is the strongest non-covalent interaction


(Kd = 10 – 15 M) [17, 18]. Biotin-tagged photoaffinity probes
can be useful not only for the separation of the probe-tar-
get protein / peptide complex in mixture (Fig. 3), but also
for the indirect visualization of this complex by using
the chemiluminescent reaction of horse-radish peroxi-
dase conjugated avidin [22, 23].
The linker region between biotin, photoreactive moi- Figure 3. Identification of target protein(s) by using biotin-tagged
ety, and the bioactive small molecule can serve multiple photoaffinity probes. The UV-cross-linked complex of a biotin-
purposes; (a) to provide enough space to prevent steric tagged photoaffinity probe with target protein(s) can be purified
and identified by using an avidin column and MS analysis,
hindrance that could decrease interactions between bin- respectively.
der and active site of target protein in living cells, (b) to
probe the structural requirements for optimal biological
activity. In order to modulate the membrane permeabil- group due to its many useful features; (a) good chemical
ity of a chemical probe in living cells and tissues, the stability in various reaction conditions that may facili-
alkyl linker spacer can be used. Polyethylene glycol (PEG) tate the use of diazirines as carbene precursors; (b) photo-
can also increase the bioavailability of probes by improv- activatability under long-wavelength UV light (wave-
ing its solubility in aqueous solutions. length 350 – 360 nm) that minimize damage to pro-
Photoreactive moieties such as an azide, a diazirine, teins;(c) generation of highly reactive carbene that vir-
and a benzophenone group, have been used as precursors tually reacts with proteins with no intramolecular rear-
for the highly reactive intermediates, which are gener- rangements. Benzophenone has been also used for photo-
ated upon photolysis [17 – 19]. Perfluorophenyl azide is affinity-labeling experiments because of its photoactivat-
the most widely used photolabeling reagent since it can ability at long wavelength (e. g. 365 nm). Benzophenone
be prepared easily. It belongs to a new class of photolabel- reacts preferentially with C-H bonds within 3.1  of the
ing reagents with improved C-H insertion efficiency com- carbonyl oxygen and its high lipophilicity can be feasible
pared with nonfluorinated analogues. The fluorine sub- to modulate the hydrophobicity and cell permeability of
stitution on the ortho-position of an azide group can elim- probes.
inate the unfavorable ketenimine formation, which is
less reactive with target proteins, and sufficiently extend Activity-based probe (ABP)
,
the lifetime of the highly reactive singlet nitrene, which The field activity-based proteomics’ makes use of ABP for
is important to obtain a stable covalent linkage. Trifluor- profiling the functional state of enzymes in complex pro-
omethylaryldiazirine has been used as the photoreactive teome [24 – 27]. ABP generally consists of three basic ele-

i 2007 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.archpharm.com


Arch. Pharm. Chem. Life Sci. 2007, 340, 169 – 177 Chemical Proteomics in Drug Discovery 173

Figure 5. Activity-based proteomics by using ABP. A typical


ABP consisting of an electrophilic reactive group (RG, arrow)
and a tag (fluorophore; circle) can be labeled with its target
enzyme(s) in a complex proteome and the activity-dependent
Figure 4. Two basic designs of ABP-based on the target selec- labeling event can be visualized by using in-gel fluorescence
tivity. In the direct ABP approach, well-characterized affinity scanning.
labels can be incorporated as the reactive group (RG) to direct
probe reactivity toward enzymes sharing a similar catalytic
mechanism and/or substrate specificity, but in the non-direct able binding groups appended to a common RG, and
ABP approach, libraries of candidate probes are synthesized these reagents are screened against complex proteome to
with variable binding groups appended to a common RG. identify activity-based labeling events. This method can
facilitate the identification of compounds possessing
ments (Fig. 4); (a) an electrophilic reactive group (RG) that both selective proteome reactivities and novel bioactiv-
is able to covalently attach to the active site of an ities [31].
enzyme, (b) a tag (e. g. biotin or fluorophore) that is useful
for subsequent purification or visualization of probe- Chemical affinity matrix
labeled enzymes, (c) a linker that connects these two ele- In order to identify target protein(s) for a bioactive small
ments, provides selective binding interactions, and pre- molecule by using chemical affinity matrix, suitable
vents steric congestion [28]. attachment sites for immobilization should be predicted
Based on a range of chemical scaffolds, ABPs have been in a small molecule from SAR data obtained with collec-
developed for more than a dozen enzyme classes includ- tions of structurally related compounds. However, in the
ing proteases, kinases, phosphatases, glycosidases, and absence of any of such data, several potential attachment
oxidoreductases. The selectivity of a chemically reactive sites can be selected to ensure that at least one of them
group in ABP allows specific proteins or protein subsets enables the immobilization of a bioactive small molecule
to be tagged and purified or visualized (Fig. 5). As a result, without conferring steric hindrance with respect to target
the application of ABP is able to identify novel enzymatic binding through a systematic trial-and-error approach.
proteins and characterize the mode of action of bioactive Small molecules possessing a functional group such as
small molecule. The design and biological application of a solvent-exposed amino, carboxyl, or hydroxyl moiety
ABP has been reviewed in detail by Evans and Cravatt can be covalently coupled to a suitable chromatography
[29]. resin; a variety of chromatography resins are commer-
Two general strategies have been introduced for the cially available that permit the immobilization of bioac-
design of ABP as shown in Fig. 4 [25, 26]. In the direct ABP tive small molecules to the terminal functional groups of
approach, well-characterized affinity labels can be incor- hydrophilic spacer molecules. After verifying retention
porated as the RG to direct probe reactivity toward of the biological activity, the small molecule-conjugated
enzymes sharing a similar catalytic mechanism and/or chemical affinity resin can be utilized for the preparative
substrate specificity. This can be feasible to identify novel purification of target proteins from the relevant biologi-
members of enzyme family to react with well-character- cal extracts [32 – 34].
ized RG [30]. Whereas, in a second strategy referred to Importantly, two kinds of chemical affinity matrices
non-direct ABP, libraries of candidate probes (biotiny- (positive and negative affinity matrices) should be run
lated sulfonate ester in Fig. 4) are synthesized with vari- simultaneously; despite of structural modification, posi-

i 2007 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.archpharm.com


174 S.-Y. Han and S.-H. Kim Arch. Pharm. Chem. Life Sci. 2007, 340, 169 – 177

separated by gel (e. g. SDS-PAGE) and subsequently ana-


lyzed by MS instruments. However, due to the exclusion
limitation in the sample preparation step and the lim-
ited sensitivity of commonly used protein staining
reagents, the traditional separation method (e. g. two-
dimensional gel electrophoresis) can not offer a lot of
information in the lower molecular weight fraction and
the low-abundance proteins of biological samples. To
overcome this problem, the enriched target proteins can
be digested with trypsin, and the resulting mixture is
then subjected to nanoflow reverse phase LC-MS/MS anal-
ysis. Liquid-based separation can easily reach into the
lower molecular weight protein and peptide range, an
area that is largely inaccessible to standard gel-based pro-
teomics. Therefore, advanced and integrated technolo-
gies such as electrospray ionization-MS/MS-integrated
nanoflow reverse phase HPLC for the separation/identifi-
cation of peptide mixtures are now being increasingly
used in the field of chemical proteomics [34, 37]. As
shown in Fig. 6, target proteins can be captured by che-
mical affinity matrices and after the following processes
of washing, elution, and trypsin digestion, the compara-
tive profiling of bound peptides allows rapid mapping of
positive chemical probe-binding peptide sequences by
nanoflow LC-MS/MS.
Consequently, the successful implementation of che-
Figure 6. Identification of target protein(s) by using chemical mical proteomics critically depends on not only the effi-
affinity matrix. Target proteins can be captured by chemical affi- ciency of affinity purification procedures, but also the
nity matrices and, after the following processes of washing, elu-
sensitivity of MS analysis. For reference, the basic of MS,
tion and trypsin digestion, the comparative profiling of bound
peptides allows rapid sequence mapping of positive chemical MS-based proteomics, and its applications (including tar-
probe-binding peptides (arrow-indicated) by LC-MS/MS. get identification and characterization, structure eluci-
dation of synthetic compounds, and early drug metabo-
tive affinity matrix retains the significant activity and lism and pharmacokinetics) in early stage of target-based
specificity, but negative affinity matrix does not exhibit drug discovery have been well-reviewed in the literatures
any potent activity [13]. This approach can differentiate [4, 38, 39].
positive affinity matrix-specific binding proteins/pep-
tides from those bound nonspecifically to both matrices
(Fig. 6). Using affinity matrices, several proteins such as Target validation by surface plasmon
FK506-binding protein and mammalian histone deacety- resonance (SPR) technology
lase 1 have been identified [35, 36].
To verify the specificity of the observed target proteins,
simple experiments such as western blot analysis and
Integration of liquid chromatography (LC) and competition binding assay were traditionally used
MS analysis [12, 23, 40]. However, these technologies do not provide
quantitative results of the relative sensitivities for the
The combination of LC and MS has had a significant bioactive small molecule to the target protein and these
impact on drug development over the past decade. Con- are not enough to turn out the initially identified pro-
tinual improvements in LC/MS interface technologies teins (or candidates for target proteins) to be a valid tar-
combined with powerful features for structure analysis get. Therefore, a need for target assessment that can
have resulted in a widened scope of application. prioritize a target candidate from an increasing number
In general, the proteins that are enriched by using che- of feasible targets being discovered through several tech-
mical tools such as probes or affinity matrices can be nologies is required.

i 2007 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.archpharm.com


Arch. Pharm. Chem. Life Sci. 2007, 340, 169 – 177 Chemical Proteomics in Drug Discovery 175

Figure 7. SPR sensorgram presenting SPR response (or reso-


nance signal) plotted against time. The sensorgram displays the
association and dissociation of complexes over the entire course
of an interaction with the kinetics.
Figure 8. Chemical proteomics and its applications in drug dis-
covery and development.
Physicochemical properties for molecules binding to
its target protein affect the biological function of target covery), but also for identifying/evaluating chemical inhi-
protein. Thus, the data showing the kinetics between bitors thereof (inhibitor discovery) as shown in Fig. 8 [26].
small molecules and proteins could be particularly help- The identification and functional annotation of the
ful in target assessment. relevant binding site(s) in a target protein for a bioactive
Without any labeling, SPR imaging permits monitor- small molecule can provide new strategies in the process
ing of biomolecular interactions by detecting the inten- of drug discovery and development such as structure-
sity change of reflected lights resulting from changes in based drug design and lead optimization [24]. Using
the refractive index on the gold surface [41, 42]; the direct binding SPR assays, compounds can be rapidly
change in mass concentration at the surface can be identified to be with specificity in the binding site of a
detected in real time with data presented as a sensor- validated target protein or candidates. For example, the
gram (SPR response plotted against time), which displays kinetic data obtained from SPR biosensors has been used
the association and dissociation of complexes with the in the screening of kinase inhibitors or potential prion
kinetics (Fig. 7). However, before examining the kinetics, disease therapeutics [43, 44]. After compounds screened,
the purified or recombinant target proteins should be the approach to find and optimize the lead compounds
prepared and then immobilized onto the sensor surface can be followed [45]. A new strategy for improved second-
as an interaction partner. Immobilization occurs by ary screening and lead optimization using high-resolu-
direct coupling to the surface or via a capture molecule tion SPR characterization of compound-target interac-
coupled to the surface. tions has been recently reviewed [46]. In this report, the
Recent SPR instruments have improved sensitivity and structure-based biophysical analysis method integrating
sample capacity, and better tools for sample recovery and SPR with bioinformatic approaches and mutation studies
the interface with MS. Therefore, the kinetics between in the early drug discovery process suggested to be used
small molecules and target proteins can be simulta- as a cost-effective alternative to high-throughput screen-
neously analyzed and/or those with high affinity and spe- ing methods in cases when detailed binding site informa-
cificity to the target can be directly identified in the fol- tion is available. Binding site-modified proteins can be
lowing MS analysis. used as reference targets in direct binding SPR assays
aimed at identifying compounds with specificity to bind-
ing site of real target protein.
Applications of chemical proteomics In addition, ABP per se has recently been shown to have
a great potential to facilitate the process of lead drug
Methods for chemical probes can provide a new avenue identification [24]; for example, of hypothetical lead
not only for identifying novel target proteins (target dis- drug candidates, the lead drug can show the most specifi-

i 2007 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.archpharm.com


176 S.-Y. Han and S.-H. Kim Arch. Pharm. Chem. Life Sci. 2007, 340, 169 – 177

city for the intended drug target in a competition bind- tion with target proteins. Importantly, the elucidation of
ing assay with the chemical probe. the relevant binding site(s) in a target protein for a bioac-
Together with secondary screening and lead optimiza- tive small molecule can provide new strategic applica-
tion, SPR technology can also be applied in a variety of tions for lead finding, lead optimization, and develop-
fields including activity characterization, compound spe- ment of structure-based drug screening system.
cificity studies, early in vitro ADMET (adsorption, distribu- In conclusion, together with technologies developed
tion, metabolism, excretion, and toxicity) evaluation and in a variety of disciplines and improved chemical and bio-
quantitative SAR analysis [45]. In clinical analyses such as logical information provided from websites (e. g. Prous
immunogenic study and food industry, the presence of Science Integrity, http://www.prous.com; Discovery Gate,
pathogens, toxins, veterinary drugs, and nutritional http://www.discoverygate.com), the advances in chemical
additives in food samples is being investigated with SPR proteomics and its expansion in drug discovery and
biosensors [47, 48]. development could provide new potential for efficient
The information about the binding site or 3D structure discovery of new classes of drugs in the near future.
of target proteins that is extracted from chemical proteo-
,
mics approaches can be used for providing virtual hits by SHK was supported by Chemical Genomics Research Project‘,
in silico screening of large substance library. Virtual Korea Research Institute of Chemical Technology, Republic of
,
screening methods have increasingly been used as a com- Korea and Chemical Genomics R&D Project‘, Ministry of
plementary means of finding small molecules that are Science and Technology, Republic of Korea.
active with a particular target. In this screening, detailed
knowledge about the binding site of the target protein
provides a promising starting point for finding com- References
pounds with selectivity that are suitable as lead candi-
dates and their biological activity can be directly evalu- [1] A. Persidis, Nat. Biotechnol. 1998, 16, 1082 – 1083.
ated in in-vivo studies. [2] J. M. Thornton, A. E. Todd, D. Milburn, N. Borkakoti, C. A.
Consequently, the contribution of chemical proteo- Orengo, Nat. Struct. Biol. 2000, 7 Suppl, 991 – 994.
mics to the pipeline in drug discovery and development [3] T. L. Blundell, H. Jhoti, C. Abell, Nat. Rev. Drug Discov.
with its several applications are being deployed in new 2002, 1, 45 – 54.
ways to boost the efficiency of discovery and develop- [4] G. Deng, G. Sanyal, J. Pharm. Biomed. Anal. 2006, 40, 528 –
ment, the quality of compounds moving through the 538.
pipeline, and the speed with which higher-quality drug [5] K. Russel, W. F. Michne in Chemogenomics in Drug Discov-
candidates enter into the clinic. ery. A Medicinal Chemistry Perspective (Ed.: H. Kubinyi, G.
Mller), Wiley-VCH, Weinheim, 2004, chapter 3.
[6] F. R. Salemme, Pharmacogenomics 2003, 4, 257 – 267.
[7] R. L. Panek, G. H. Lu, S. R. Klutchko, B. L. Batley, et al., J.
Conclusion Pharmacol. Exp. Ther. 1997, 283, 1433 – 1444.
It is very clear that the success in the development of new [8] T. T. Ashburn, K. B. Thor, Nat. Rev. Drug Discov. 2004, 3,
673 – 683.
small molecule therapeutics could be decided in the step
to choose a suitable target because a bad choice makes us [9] M. R. Chance, A. R. Bresnick, S. K. Burley, J. S. Jiang, et al.,
Protein Sci. 2002, 11, 723 – 738.
waste time and money in competitive drug markets. In
[10] J. Westbrook, Z. Feng, L. Chen, H. Yang, H. M. Berman,
drug discovery and development, chemical proteomics
Nucleic Acids Res. 2003, 31, 489 – 491.
strongly presents the possibility of speeding up target
[11] Y. J. Chen, S. H. Chen, Y. Y. Chien, Y. W. Chang, et al.,
identification and getting higher-quality leads in a rapid,
ChemBioChem 2005, 6, 1169 – 1173.
systematic, and comprehensive manner by design, synth-
[12] T. Sugimoto, T. Fujii, Y. Idutu, S. Yamamura, M. Ueda,
esis, and application of relevant chemical probes. In this Tetrahedron Lett. 2004, 45, 335 – 338.
promising approach, the interplay of chemistry and biol- [13] Y. Oda, T. Owa, T. Sato, B. Boucher, et al., Anal. Chem.
ogy drives target selection and lead identification with 2003, 75, 2159 – 2165.
integrated strategy; chemistry can provide the critical [14] J. M. de La Fuente, A. G. Barrientos, T. C. Rojas, J. Rojo, et
technologies in the step to design and synthesize the che- al., Angew. Chem. Int. Ed. Engl. 2001, 40, 2257 – 2261.
mical tools, and then, the advanced disciplines such as [15] E. Katz, I. Willner, Angew. Chem. Int. Ed. Engl. 2004, 43,
cell biology, biochemistry, proteomics, structural biol- 6042 – 6108.
ogy, bioinformatics, and physical chemistry provide pro- [16] C. Xu, K. Xu, H. Gu, X. Zhong, et al., J. Am. Chem. Soc.
found knowledge of compound selectivity and its interac- 2004, 126, 3392 – 3393.

i 2007 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.archpharm.com


Arch. Pharm. Chem. Life Sci. 2007, 340, 169 – 177 Chemical Proteomics in Drug Discovery 177

[17] G. Dorman, G. D. Prestwich, Trends Biotechnol. 2000, 18, [32] M. Knockaert, N. Gray, E. Damiens, Y. T. Chang, et al.,
64 – 77. Chem. Biol. 2000, 7, 411 – 422.
[18] Y. Hatanaka, Y. Sadakane, Curr. Top. Med. Chem. 2002, 2, [33] K. Godl, J. Wissing, A. Kurtenbach, P. Habenberger, et al.,
271 – 288. Proc. Natl. Acad. Sci. USA 2003, 100, 15434 – 15439.
[19] F. Kotzyba-Hibert, I. Kapfer, M. Goeldner, Angew. Chem. [34] J. Wissing, K. Godl, D. Brehmer, S. Blencke, et al., Mol. Cell
Int. Ed. Engl. 1995, 34, 1296 – 1312. Proteomics 2004, 3, 1181 – 1193.
[20] S. Y. Han, S. S. Park, W. G. Lee, Y. K. Min, B. T. Kim, Bioorg. [35] M. W. Harding, A. Galat, D. E. Uehling, S. L. Schreiber,
Med. Chem. Lett. 2006, 16, 129 – 133. Nature 1989, 341, 758 – 760.
[21] J. S. Lee, S. Y. Han, M. H. Kim, C. M. Yu, et al., Bioorg. Med. [36] J. Taunton, C. A. Hassig, S. L. Schreiber, Science 1996, 272,
Chem. Lett. 2006, 16, 4733 – 4737. 408 – 411.
[22] W. Frick, A. Bauer-Schafer, J. Bauer, F. Girbig, et al., [37] D. Brehmer, Z. Greff, K. Godl, S. Blencke, et al., Cancer Res.
Bioorg. Med. Chem. 2003, 11, 1639 – 1642. 2005, 65, 379382.
[23] S. Y. Han, S. H. Choi, M. H. Kim, W. G. Lee, et al., Tetrahe- [38] G. L. Glish, R. W. Vachet, Nat. Rev. Drug Discov. 2003, 2,
dron Lett. 2006, 47, 2915 – 2919. 140 – 150.
[24] D. A. Jeffery, M. Bogyo, Curr. Opin. Biotechnol. 2003, 14, [39] R. Aebersold, M. Mann, Nature 2003, 422, 198 – 207.
87 – 95.
[40] K. Saravanan, M. Paramasivam, S. Dey, T. P. Singh, A. Sri-
[25] N. Jessani, B. F. Cravatt, Curr. Opin. Chem. Biol. 2004, 8, nivasan, J. Cardiovasc. Pharmacol. 2004, 44, 287 – 293.
54 – 59.
[41] R. Gambari, Am. J. Pharmacogenomics 2001, 1, 119 – 135.
[26] A. E. Speers, B. F. Cravatt, ChemBioChem 2004, 5, 41 – 47.
[42] H. Zhu, M. Snyder, Curr. Opin. Chem. Biol. 2003, 7, 55 – 63.
[27] D. A. Campbell, A. K. Szardenings, Curr. Opin. Chem. Biol.
2003, 7, 296 – 303. [43] H. Nordin, M. Jungnelius, R. Karlsson, O. P. Karlsson,
Anal. Biochem. 2005, 340, 359 – 368.
[28] S. H. L. Verhelst, M. Bogyo, Biotechniques 2005, 38, 175 –
177. [44] T. R. K. Reddy, R. Mutter, W. Heal, K. Guo, et al., J. Med.
Chem. 2006, 49, 607 – 615.
[29] M. J. Evans, B. F. Cravatt., Chem. Rev. 2006, 106, 3279 –
3301. [45] S. Lofas, Assay Drug Dev. Technol. 2004, 2, 407 – 415.
[30] A. Borodovsky, H. Ovaa, N. Kolli, T. Gan – Erdene, et al., [46] W. Huber, J. Mol. Recogit. 2005, 18, 273 – 281.
Chem. Biol. 2002, 9, 1149 – 1159. [47] R. Karlsson, J. Mol. Recogit. 2004, 17, 151 – 161.
[31] G. C. Adam, B. F. Cravatt, E. J. Sorensen, Chem. Biol. 2001, [48] D. Nedelkov, A. Rasooly, R. W. Nelson, J. Food Microbiol.
8, 81 – 95. 2000, 60, 1 – 13.

i 2007 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim www.archpharm.com

You might also like