Вы находитесь на странице: 1из 9

review article

published online: 18 march 2013 | doi: 10.1038/nchembio.1199

Target identification and mechanism of action in


chemical biology and drug discovery
Monica Schenone1, Vlado Dank2, Bridget K Wagner2 & Paul A Clemons2*

Target-identification and mechanism-of-action studies have important roles in small-molecule probe and drug discovery.
Biological and technological advances have resulted in the increasing use of cell-based assays to discover new biologically
active small molecules. Such studies allow small-molecule action to be tested in a more disease-relevant setting at the outset,
but they require follow-up studies to determine the precise protein target or targets responsible for the observed phenotype.
Target identification can be approached by direct biochemical methods, genetic interactions or computational inference.
In many cases, however, combinations of approaches may be required to fully characterize on-target and off-target effects and
to understand mechanisms of small-molecule action.

M
odern chemical biology and drug discovery each seek to approach is characterized by identifying, often under experimen-
2013 Nature America, Inc. All rights reserved.

identify new small molecules that potently and selectively tal selection pressure, a phenotype of interest, followed by identi-
modulate the functions of target proteins. Historically, fication of the gene (or genes) responsible for the phenotype (see
nature has been an important source for such molecules, with knowl- refs. 9, 10). Modern molecular biological methods, particularly
edge of toxic or medicinal properties often long predating knowledge genetic engineering approaches, have given rise to reverse genetics
of precise target or mechanism. Natural selection provides a slow (sometimes equated with molecular genetics), in which a specific
and steady stream of bioactive small molecules, but each of these gene of interest is targeted for mutation, deletion or functional abla-
molecules must perforce confer reproductive advantage in order for tion (for example, with RNAi11), followed by a broad search for the
nature to invest in its synthesis. In recent decades, investments in resulting phenotype (see refs. 12, 13).
finding new small-molecule probes and drugs have expanded to a By analogy to genetics, there are two fundamental approaches to
paradigm of screening large numbers (typically 103106) of com- understanding the action of small molecules on biological systems14,15.
pounds for those that elicit a desired biological response1,2. In some Biochemical screening approaches are analogous to reverse genet-
cases, these studies interrogate natural products3,4, but more often ics (Fig. 1a). In advance of conducting a high-throughput screen,
they involve collections of synthetic small molecules prepared by the protein target is selected and (typically) purified before expo-
organic chemistry strategies5,6 that rapidly yield large collections of sure to small molecules16,17. This target validation or credentialing
relatively pure compounds. Thus, the overall discovery paradigm is a time-consuming process that involves demonstrating the rel-
involves investments both in organic synthesis and in biological evance of the protein for a particular biological pathway, pro-
testing of large compound collections. cess or disease of interest18,19. Once a target has been validated, it
Since the revolution in molecular biology, the biological test- is presumed that binders or inhibitors of this protein will affect
ing component of screening-based discovery has overwhelm- the desired process. Often, however, such an impact needs to be
npg

ingly involved testing compounds for effects on purified proteins. characterized more completely in cells or animals by observing
However, with advances in assay technology, many research pro- compound-induced phenotypes; hence, this approach has been
grams are increasingly turning (or returning) to cell- or organism- termed reverse chemical genetics.
based phenotypic assays that benefit from preserving the cellular In contrast, forward chemical genetics refers to the process of
context of protein function7. The cost paid for this benefit is that testing small molecules directly for their impact on biological pro-
the precise protein targets or mechanisms of action responsible cesses, often in cells or even in animals (Fig. 1b)20,21. Phenotypic
for the observed phenotypes remain to be determined. Even screens expose candidate compounds to proteins in more biologi-
after a relevant target is established, additional functional studies cally relevant contexts than screens involving purified proteins7,22.
may help to identify unwanted off-target effects or establish new Because these screens measure cellular function without imposing
roles for the target protein in biological networks. In this review, preconceived notions of the relevant targets and signaling pathways,
we address these important steps in the discovery process, they offer the possibility of discovering new therapeutic targets.
termed target identification or deconvolution8, illustrating Indeed, several important drug programs have been inspired by
methods available to approach the problem, highlighting recent phenotypic screening results, including the effects of cyclosporine A
advances and discussing how findings from multiple approaches and FK506 on T-cell receptor signaling7,23, leading to the discoveries
are integrated. of FKBP12 (ref. 24), calcineurin25 and mTOR26, and the performance
of trapoxin A in differentiation and proliferation assays27, leading to
Background the discovery of histone deacetylases28,29. Importantly, such assays
Historically, genetics has provided powerful biological insights, prevalidate the small molecule and its (initially unknown) protein
allowing characterization of protein function by manipulation target as an effective means of perturbing the biological process or
of genetic sequence. A forward genetics (or classical genetics) disease model under study.

Proteomics Platform, Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA. 2Chemical Biology Program, Broad Institute of Harvard and
1

MIT, Cambridge, Massachusetts, USA. *e-mail: pclemons@broadinstitute.org

232 nature chemical biology | vol 9 | april 2013 | www.nature.com/naturechemicalbiology


Nature chemical biology doi: 10.1038/nchembio.1199 review article
a Target-based b Phenotype-based of a compound can be generated by gene expression profiling in
(reverse chemical genetics) (forward chemical genetics)
the presence or absence of compound treatment. Many target-
identification projects actually proceed through a combination of
Target Disease
validation model these methods, where researchers use both direct measurements
and inferences to test increasingly specific target hypotheses. Indeed,
we suggest that the problem of target identification will not gener-
ally be solved by a single method but rather by analytical integration
Biochemical Phenotypic
of multiple, complementary approaches.
assay assay
Direct biochemical methods
Biochemical affinity purification provides the most direct approach
to finding target proteins that bind small molecules of interest37.
Candiate small
molecules
Because they are based on physical interactions involving mam-
malian or human proteins, biochemical methods can lead to infor-
mation about molecular mechanisms of efficacy or toxicity highly
Target
identification relevant to human disease. Similarly, small-molecule optimiza-
tion efforts can be complemented by biochemical methods when
Mechanism-of-
action studies three-dimensional structure information about the target is known.
Finally, unbiased protein identification, especially from lysates
Figure 1 | Mechanism-of-action and target identification in chemical
containing intact protein complexes, potentially allows evaluation
genetics. (a) Target-based approaches (reverse chemical genetics) begin
of polypharmacology.
with target validation, in which a role is established for a protein in a
Pioneering work in affinity purification involved monitor-
pathway or disease, followed by a biochemical assay to find candidate
ing chromatographic fractions for enzyme activity after exposure
2013 Nature America, Inc. All rights reserved.

small molecules; mechanism-of-action studies are still required to validate


of extracts to compound immobilized on a column, followed by
cellular activities of candidates and evaluate possible side effects.
elution43. In general, such an approach requires large amounts of
(b) Phenotype-based approaches (forward chemical genetics) begin
extract, possibly prefractionated, and stringent wash conditions.
with a phenotype in a model system and an assay for small molecules
Such approaches have been used with success to identify certain pro-
that can perturb this phenotype; candidate small molecules must then
tein targets, including those of both natural29 and synthetic44 small
undergo target-identification and mechanism-of-action studies to
molecules, and one such approach has been used to elucidate the
determine the protein responsible for phenotypic change.
mechanism of the teratogenic side effect of thalidomide35. However,
these methods seem best suited for situations where a high-affinity
ligand binds a relatively abundant target protein. High-stringency
However, phenotypic assays require a subsequent effort to dis- washes can bias proteins identified to those with the highest-
cover the molecular targets of bioactive small molecules, which affinity interactions, decreasing the likelihood of finding additional
can be a complex endeavor30. We often assume that direct inter- targets that might be important in cellular contexts, as suggested
action with a single target is responsible for phenotypic observa- by proteomic profiling studies (for example, those described in
tions, but this need not be the case. Many drugs show side effects refs. 45, 46). Furthermore, stringent washing will also reduce the
owing to interactions with off-target proteins31,32, and even small ability to identify protein complexes in which the direct target par-
moleculeinduced phenotypes observed in cell culture may repre- ticipates and whose members identities might help illuminate the
sent the superposition of effects on multiple targets33,34. For drug connection between the direct target and the cellular activity of the
development, target identification is important to follow-up studies, small molecule.
aiding medicinal chemistry efforts. Furthermore, identifying both Affinity purification experiments also involve the challenge of
npg

the therapeutic target and other targets that might cause unwanted preparing immobilized affinity reagents that retain cellular activ-
side effects enables optimization of small-molecule selectivity35. ity, so that target proteins will still interact with the small molecule
Conversely, polypharmacology can be considered a new tool, lever- while it is bound to a solid support. A related issue is the identifica-
aging multiple small-molecule effects to gain maximal effect, once tion of appropriate controls and tethers. Different approaches are
again underscoring the benefit of an unbiased approach to screen- possible; control beads loaded with an inactive analog47,48 or capped
ing and the need for target deconvolution36. without compound have been used49. These control experiments
have limitations, most notably the availability of related inactive
Approaches to target identification compounds. The inactive analog must be sufficiently different from
In this review, we will cover three distinct and complementary the compound of interest to fail to bind the target, raising the pos-
approaches for discovering the protein target of a small molecule: sibility that it will have different physicochemical properties and
direct biochemical methods, genetic interaction methods and com- therefore different nonspecific interactions with proteins. In the
putational inference methods. Direct methods involve labeling the case of capped beads, the results are confounded by the background
protein or small molecule of interest, incubation of the two popula- of high-abundance, low-affinity proteins with slight differential
tions and direct detection of binding, usually following some type binding to the control bead. Elution from the bead-bound small
of wash procedure (reviewed in ref. 37). Genetic manipulation can molecule24 or preincubation of lysate with compound is a viable
also be used to identify protein targets by modulating presumed tar- alternative control35,49,50 but is limited by compound solubility. The
gets in cells, thereby changing small-molecule sensitivity (reviewed choice of tether, influencing the type of background proteins iden-
in ref. 38). Target hypotheses, in contrast, can be generated by com- tified, also becomes a critical parameter5155. These challenges can
putational inference, using pattern recognition to compare small- frustrate individual attempts at affinity purification, particularly if a
molecule effects to those of known reference molecules or genetic biochemical approach is used in isolation.
perturbations3941. Mechanistic hypotheses, rather than targets per se, Recent affinity-based methods have attempted to overcome
for new compounds emerge from such tests. The target pathway or one or more of these challenges. Approaches based on chemical or
protein of a new small molecule is inferred but remains to be con- ultraviolet lightinduced cross-linking56,57 use covalent modifica-
firmed42. Similarly, hypotheses regarding the mechanism of action tion of the protein target to increase the likelihood of capturing

nature CHEMICAL BIOLOGY | vol 9 | april 2013 | www.nature.com/naturechemicalbiology 233


review article Nature chemical biology doi: 10.1038/nchembio.1199

low-abundance proteins or those with low affinity for the small


molecule. This method requires prior knowledge of the enzyme
activity being targeted, making it a slightly biased approach. If there
is no bias, cross-linking of the small molecule to proteins is bur-
Light-isotope Heavy-isotope
dened with the possibility of high, nonspecific background owing to label label
the cross-linking event itself. A variation on the use of photoaffinity
reagents couples covalent modification to two-dimensional gel elec-
trophoresis in an attempt to deconvolve nonspecific interactions58. Immobilized SM Immobilized SM
+ soluble competitor
A nonselective universal coupling method that enables attachment
to a solid support by a photoaffinity reaction resulted in the identi-
fication of an inhibitor of glyoxalase I59. This method assumes that
a compound could bind the solid support in multiple ways while
some functional relevant sites remain available to interact with
the protein target. This approach, however, runs the risk of false-
negative results when the functional group is masked in the coupling Mixing, washing, electrophoresis,
reaction. Another method for immobilizing small molecules, that trypsinization
is, coupling them to peptides that allow them to recover the probe-
protein complex by immunoaffinity purification60, was devised to
address this issue. Specific, indirect Nonspecific Specific, direct
Some small-molecule libraries are prepared with synthetic
handles primed for making affinity matrices after an activity is Quantitative MS

identified61,62. Because these methods rely on advance modification


of the compound structure, they require additional chemistry exper-
2013 Nature America, Inc. All rights reserved.

tise and may not be possible for all compound classes. Similarly,
if a small molecule can be fluorescently labeled, it can be used Figure 2 | Illustration of stable isotope labeling and quantitative MS.
to probe proteins separated by microarray (reviewed in ref. 63). Cells are labeled with either heavy- or light-isotope labels. One sample is
However, such methods are limited to proteins that can be readily exposed to bead-immobilized small molecules (SM) in the presence of
manipulated, usually in heterologous expression systems. The use of soluble competitor compound and the other in the absence of competitor.
purified proteins does not necessarily ensure physiological expres- Following mixing, washing and electrophoresis, samples are digested
sion levels, giving incorrect information about relative binding to using trypsin and peptide fragments analyzed by quantitative MS49. Ratios
alternative targets in cells and masking effects due to the formation of heavy- and light-labeled peptides are used to determine specificity of
of protein complexes. interactions for the small molecule, potentially including both direct and
Two interesting new target-identification approaches that cir- indirect targets (for example, members of complexes including the direct
cumvent the need to immobilize compounds have emerged in the target), but not to differentiate them.
last few years. One of them uses changes in protein susceptibility
to proteolytic degradation upon small-molecule binding (reviewed and absolute quantification (iTRAQ)75, coupled with free soluble
in ref. 64). The other is based on a characteristic shift in the chro- competitor for elution as a control, has been used to profile kinases
matographic retention-time profile after a compound binds a pro- enriched by affinity purification with nonselective kinase-binding
tein target65. Although the generality of these approaches remains small molecules76. There are variations on chemical-labeling strate-
to be determined, their combination with quantitative proteomics gies, such as mass differential tags for relative and absolute quanti-
is quite promising. fication (mTRAQ)75, tandem mass tags (TMT)77 and stable isotope
Affinity chromatography has been coupled to powerful new dimethyl labeling78,79. These chemical labeling strategies provide
npg

techniques in MS, which can possibly provide the most sensitive more versatility regarding the type of samples that can be labeled,
and unbiased methods of finding target proteins. Quantitative but a weakness lies in the fact that they generally rely on labeling
proteomics66 (reviewed in ref. 67) has been effective in identifying at the peptide level, later in the proteomic workflow, and thus are
specific protein-protein interactions by affinity methods68,69 and has prone to more variation and less accuracy80.
been increasingly applied to proteinsmall molecule interactions These unbiased approaches show great promise but will require
(reviewed in ref. 70). In the context of target identification, two dif- new software8183 and analytical techniques84 to quantify the relative
ferent quantitative techniques have been used; these can be broadly expression of members of candidate target lists to aid in experimen-
divided into metabolic and chemical labeling. tal prioritization. In contrast, biased approaches to target deconvo-
Metabolic labeling, more specifically stable-isotope labeling by lution based on profiling small-molecule activity against a panel of
amino acids in cell culture (SILAC)71, has been effectively used, in enzymes are now commercially available and widely used. These
experiments with gentle washing and free soluble competitor pre- methods rely on prior knowledge of the enzyme family (kinases,
incubation of lysates, to provide unbiased assessment of multiple ubiquitinases, demethylases and so on). For example, assay-
direct and indirect targets (Fig. 2)49. It has also been used in com- performance profiling and compounds with a known mode of
bination with serial drug-affinity chromatography to characterize action were used to predict kinase inhibitory activity for new com-
the quantitative changes of the kinome in different phases of the pounds emerging from a phenotypic screen42. A commercial kinase
cell cycle72. Metabolic labeling has the advantage of allowing sample profiling panel85 then confirmed activity against a subset of kinases.
pooling early in the process, eliminating quantification errors due Having information from genetic studies or computational methods
to sample handling. A disadvantage is that it limits the workflow to to decide which set of enzymes to investigate provides a valuable
immortalized cell lines73. tool, again underscoring the importance of integrating all methods
Chemical labeling has also been successfully used in the past. at the researchers disposal.
Isotope-coded affinity tag (ICAT) technology74, coupled with
beads loaded with active and inactive compound as controls, has Genetic interaction and genomic methods
been used to identify malate dehydrogenase as a specific target of Target identification based on genetic or genomic methods lever-
the anticancer compound E7070 (ref. 47). Isobaric tags for relative ages the relative ease of working with DNA and RNA to perform

234 nature chemical biology | vol 9 | april 2013 | www.nature.com/naturechemicalbiology


Nature chemical biology doi: 10.1038/nchembio.1199 review article
a Panel of single-gene deletions b Strain 1 Mating Strain 2 c ORF library

Gene-specific SM-resistant
SM sensitivity muntant

Recombinant
strains

Spores

Compare sensitivity Differential


to double knockouts Map sensitivity
fitness
to genetic loci

Figure 3 | Illustrations of yeast genomic methods for target-identification and mechanism-of-action studies. (a) A panel of viable single-gene deletions
is tested for small-molecule (SM) sensitivity, indicating synthetic-lethal interactions between potential targets and the original deletion; mechanisms are
interpreted by comparing interactions to double-knockout strains146. (b) Different strains of diploid yeast are mated to form F1 recombinants, and meiotic
progeny are subjected to small molecules; segregation frequencies allow mapping of small-molecule sensitivity to genetic loci147. (c) A recessive small
moleculeresistant mutant is transformed with a wild-type open reading frame library; transformants obtaining a wild-type copy of the mutant gene are
selectively sensitive to small molecules, resulting in their depletion among pooled transformants, as quantified by microarray89.
2013 Nature America, Inc. All rights reserved.

large-scale modifications and measurements. These methods often small-molecule screens95, which could serve as a follow-up method
use the principle of genetic interaction, relying on the idea of genetic for the other techniques described in this review.
modifiers (enhancers or suppressors) to generate target hypotheses. Genetic target-identification efforts are increasingly focused
Gene knockout organisms, RNAi (reviewed in ref. 86) and small on mammalian cells. For example, examination of compound-
molecules with well-defined mechanisms can each be used to resistant clones of cells, using transcriptome sequencing (RNA-seq),
alter the functions of putative targets, uncovering dependen- identified intracellular targets of normally cytotoxic compounds96.
cies on activity. For example, if a gene knockdown phenocopies a Advantages of this approach include the ability to perform cell
compounds effects, the evidence that the protein could be the typespecific analyses and not having to chemically modify the
target relevant to that phenotype would be strengthened. In this compound to perform the analysis. Clones of HCT116 colon
case, hypersensitivity of individual mutants to sublethal concentra- cancer cells resistant to the polo-like kinase 1 (PLK1) inhibitor
tions of compound demonstrates a chemical-genetic interaction BI 2356 were sequenced and compared to the parental line. Although
(Fig. 3a). Similarly, mating of laboratory and wild yeast strains can PLK1 was not mutated in every clone, it was the only gene mutated
reveal patterns of small-molecule sensitivity with specific genetic in more than one group; moreover, mutations were present in
loci (Fig. 3b)87. the known binding site of BI 2356. This proof-of-principle study
These concepts have been expanded to include molecularly may pave the way for more rapid target identification in mamma-
barcoded libraries of open reading frames88 and detection of small lian cells, although this approach is currently limited to cell viability
moleculeresistant clones by microarray (Fig. 3c). Importantly, these as a phenotype.
studies provide a conceptual framework for understanding complex Finally, recent efforts to use gene expression signatures to deter-
diseases. Although direct translation to human biology may not be mine compounds mechanisms of action illustrate the close relation-
npg

forthcoming, owing to the lack of conservation of some pathways ship between genetic techniques and computational methods. Using
between yeast and human, this framework enables us to envision transcription profiling data from the Connectivity Map97, a recent
technical advances that facilitate this type of analysis in mammalian study described a weighting scheme to rank order lists of genes
systems. An accompanying perspective in this series89 addresses across multiple cell lines, resulting in what was termed a prototype
these approaches in more detail. ranked list98. The authors then used gene-set enrichment analysis99
A promising genetics-based technique for target identification to compute pairwise distances between the ranked lists for each
involves combining results from small-molecule and RNAi pertur- compound and constructed a network in which each compound
bations. This approach enables parallel testing of small-molecule was a node. Clustering revealed communities of nodes connected
and RNAi libraries for induction of the same cellular phenotype90,91. to each other, two-thirds of which were enriched for similar mecha-
RNAi experiments can be performed on a genome-wide scale nisms of action, as determined by anatomical therapeutic chemical
to find phenotypes similar to those induced by small molecules codes. A limitation of this approach is the reliance on accurate anno-
(Fig. 4a)92. Alternatively, when some mechanistic clues already tation of small-molecule activity, but even so, this type of sophisti-
exist, a more focused set of RNAi reagents can be used to find path- cated approach will most likely uncover similarities between known
way members that alter the effects of a small molecule (Fig. 4b)93. bioactive compounds and new screening hits. The line separating
The main strength of combining small-molecule and RNAi pertur- genetic and computational approaches increasingly blurs as the
bations is the ability to measure phenotypic effects in more physi- technical hurdles to generating massive data sets are surmounted.
ologically relevant cellular contexts, using mammalian, or even
human, cells. Clearly, genetic perturbations cannot always reca- Computational inference methods
pitulate or phenocopy the effect of a small molecule94, for example On their own, computational methods are used to infer protein
because of the risk of genetic compensation. Using RNAi libraries targets of small molecules, in addition to providing analytical sup-
that contain entire families of genes may help alleviate this problem. port for proteomic and genetic techniques. These methods can
Another way to dissect these effects is to use a suboptimal concen- also be used to find new targets for existing drugs, with the goal of
tration of small molecule in combination with genetic knockdown. drug repositioning or explaining off-target effects. Profiling meth-
An elegant study provided proof of concept for RNAi-sensitized ods rely on pattern recognition to integrate results of parallel or

nature CHEMICAL BIOLOGY | vol 9 | april 2013 | www.nature.com/naturechemicalbiology 235


review article Nature chemical biology doi: 10.1038/nchembio.1199

a Genome-wide new activities for small molecules111. Such approaches represented


RNAi reagents
important conceptual advances in this field because they considered
multidimensional profiling as a method to identify small-molecule
mechanisms of action.
Gene expression technology has also had an important role in
profile-based inference of small-molecule targets and mechanisms.
One early study used gene expression profiles of yeast treated with
the immunosuppressants FK506 and cyclosporine A. This study not
only recapitulated the importance of calcineurin in immunosup-
+ pression but also identified a number of other genes that were sug-
gested to be secondary targets112. Later, a compendium of yeast gene
expression profiles was derived from both genetic mutations and
Effect based RNAi phenocopies small-molecule perturbations to annotate uncharacterized genes
on SM perturbation SM effect
and pathways40. Commercial databases of rat tissuebased expres-
b Pathway-specic sion profiles following drug treatment were developed to identify
RNAi reagents
toxicities of new chemical entities113. Subsequently, the Connectivity
Map was developed as a public database of gene expression pro-
SM inhibits files derived from small-molecule perturbations of human cell lines
target or pathway
(Fig. 5a), which enables the functions of new small molecules to be
identified by pattern matching97.
Several groups have developed profiling methods based on
other measurement technologies114,115, including affinity profiling of
Pathway Pathway
activator repressor biochemical assays to predict ligand binding to proteins116, and on
2013 Nature America, Inc. All rights reserved.

similarities among side-effect profiles117. Affinity profiling was also


used to design biologically diverse screening libraries118. Several
promising studies have involved profiling by high-throughput
microscopy (sometimes called high-content screening), allowing
Differential SM
sensitivity phenotypes to be clustered, in a manner analogous to transcriptional
profiles, to discover potential small-molecule targets119,120. More
Figure 4 | Illustrations of RNAi-based methods for target-identification generally, public databases such as ChemBank105 or PubChem106
and mechanism-of-action studies. (a) In one implementation, phenotypes provide extensible environments for accumulating rich phenotypic
from genome-wide RNAi are compared to those induced by a small profiles for many compounds exposed to a diverse set of assays
molecule (SM) of interest; full or partial phenocopy of the small-molecule and have been exploited to understand both bioinformatic121 and
effect by RNAi provides evidence that the gene product is a small- cheminformatic101,122 relationships (Fig. 5b).
molecule target92. (b) When prior evidence suggests a particular target More focused profiling methods investigate only particular
pathway, focused sets of RNA reagents can help to generate mechanistic aspects of biological systems. For example, high-throughput screen-
hypotheses. In general, RNAi can enhance or suppress small-molecule ing (HTS) activity profiling and a guilt-by-association approach
effects, as in genetic epistasis analysis; in practice, more complex were used to determine mechanisms of action for hits in an antima-
relationships among proteins than those illustrated may also exist93. larial screen123. Another study124 examined small moleculeinduced
cell death and systematically characterized lethal compounds on the
multiplexed experiments, typically from small-molecule phenotypic basis of modulatory profiles, created experimentally by measuring
profiling100,101. Ligand-based methods also incorporate chemical cell viability. Comparison of modulatory profile clusters with gene
npg

structures to predict targets. Structure-based methods rely on three- expression and structure-based profiles demonstrated that that
dimensional protein structures to predict proteinsmall molecule modulatory profiles revealed additional relationships. Bioactivity
interactions102,103. Owing to their limitation to proteins with known profile similarity search (BASS) is another method that uses profiles
structures, structure-based methods will not be discussed in detail. of dose-response cell-based assay results to associate targets with
In general, bioactivity profiling methods are based on the prin- small molecules125. It is increasingly likely that single measurements
ciple that compounds with the same mechanism of action will will be inadequate to the task of determining mechanism of action.
have similar behavior across different biological assays. Public
databases of, for example, gene expression97,104 or small-molecule a Connectivity using b Connectivity using
screening105,106 data sets house measurements that record pheno- gene expression cellular assays

typic consequences of small-molecule perturbations. Target or Reference


mechanism inference frequently relies on the fact that molecules databases

with known mechanisms of action (sometimes called landmark


compounds100) are also profiled, allowing new compounds to be
assessed for similar patterns of performance107,108. Small-molecule
profiling data can be mined most effectively when there exists a crit- Similarity to
ical mass of data generated using a common set of compounds and Gene expression landmark SMs Assay performance
signature with known signature
cell states (multidimensional screening109). A prototypical example mechanism
of this type of study is the US National Cancer Institutes NCI-60 set
of 60 cancer cell lines that was exposed to a large collection of small Figure 5 | Illustration of computational inference methods for target-
molecules110. In a seminal study using the NCI-60 data set, protein identification and mechanism-of-action studies. In general, data sets
targets were connected (via their degree of expression in each cell that provide multidimensional readouts of small molecule (SM)-induced
line) to small-molecule sensitivity patterns in the same cell lines39 phenotypes can be used to provide connections between new small-
(reviewed in ref. 101). Similar studies, including those using the molecule signatures and reference databases by similarity to landmark
COMPARE algorithm110, have allowed researchers to hypothesize compounds with known mechanisms of action.

236 nature chemical biology | vol 9 | april 2013 | www.nature.com/naturechemicalbiology


Nature chemical biology doi: 10.1038/nchembio.1199 review article
Recent profiling methods have taken full advantage of high- Small-molecule Genetic interaction
throughput data available in public and proprietary screening discovery methods
databases, for example by developing HTS fingerprints (HTS-FP)
Global expression
with a goal of facilitating virtual screening and scaffold hopping126. profiling
Enrichment of gene ontology terms among known protein targets Target
was observed within HTS-FP clusters. Further, HTS-FP can be used Computational
identification

to select biologically diverse compounds for screening when testing inference methods

the full library is not possible. Direct biochemical


Using ligand-based predictive modeling to classify com- methods

pounds is a well-established practice in computational chemistry.


Independently, two groups have extended predictive-modeling Figure 6 | Illustration of a conceptual workflow for integrated target-
approaches to explore global relationships among biological tar- identification and mechanism-of-action studies. Small-molecule
gets. In one case127, protein-ligand interactions were explored with discovery often starts with phenotypic screening, and, depending on the
a goal of predicting targets for new compounds. Using Laplacian- expertise available to the researchers, target identification could proceed
modified naive Bayesian modeling, trained on 964 target classes in using any combination of direct biochemical methods, genetic or genomic
the WOMBAT database, the researchers were able to predict the methods or using computational inference methods. A key component for
top three most likely protein targets and validated their approach success is the integration of data from all available methods to produce the
by examining therapeutic activities of compounds in the MDL most reliable target and mechanistic hypotheses.
Drug Data Report database. Such modeling can also be used to
explain off-target effects or to design target- or family-focused protein sequences and drug-target network topology resulted in
libraries. In a separate but similar approach128, a large collection the creation of a unified pharmacological space that could pre-
of structure-activity relationship data was assembled from vari- dict ligand-target interactions for new compounds and proteins137.
ous public and proprietary sources, resulting in a set of 836 human Several additional studies report recent advances in network-based
2013 Nature America, Inc. All rights reserved.

genes that are targets of small molecules with binding affinities less approaches to target prediction138,139.
than 10 M. These connections were used to generate a polyphar- Inference-based methods arguably have the least bias of any
macology interaction network of proteins in chemical space. This target-identification method as they often rely on experiments done
network enables a deeper understanding of compound and target by others. The analyst is distant from the original experimental
cross-reactivity (promiscuity) and provides rational approaches to design and is therefore poised to reveal unanticipated relationships.
lead hopping and target hopping. In contrast, such studies rely on data sets that require substantial time
Chemical similarity is often used as a metric of success for and investment, however distributed, to realize. Fortunately, compu-
pattern-recognition approaches. The similarity ensemble approach tational techniques have increasingly emerged to take advantage of
(SEA)129 works by quantitatively grouping related proteins on the the explosion of public data sources. Structured publicly accessible
basis of the chemical similarity of their ligands. For each pair of databases are the best hope for the success of such methods; they not
targets, chemical similarity was computed and properly normalized only promote the availability of data from multiple sources but also
according to a null model of random similarity. Even though only encourage rigor among experimentalists by exposing their data to
ligand chemical information was used, biologically related clusters of critical evaluation by the scientific community at large.
proteins emerged. Importantly, in the SEA results, there were cases
of ligand-based clusters differing from protein sequencebased Summary and outlook
clusters; for example, ion channels and G proteincoupled receptors Each of the described approaches to target-identification and
are ligand related, but they have no structure or sequence similarity. mechanism-of-action studies has strengths and limitations, and,
In contrast, many neurological receptors with related sequences importantly, different laboratories will have different technical
lack pharmacological similarity. The authors documented the utility strengths. Laboratories with expertise in chemistry or biochemis-
npg

of SEA by predicting and confirming off-target activities of metha- try might gravitate toward direct biochemical approaches, whereas
done, loperamide and emetine. Large-scale prediction of off-target genetic or cell biology laboratories might favor genetic interaction
activities using SEA was subsequently described130. approaches, and groups with a high degree of computational experi-
SEA is well suited to provide insights into the mechanisms of ence might first pursue methods that investigate databases for clues.
action of small molecules. With that purpose in mind, SEA was Of course, there is no right answer about which method is best.
used on the MDL Drug Data Report database to predict new Rather, we suggest that a combination of approaches93,140 is most
ligand-target interactions131. The predictions were either confirmed likely to bear fruit (Fig. 6).
by literature or database search or investigated experimentally. There have been a few examples of successful integration of
Out of 30 experimentally tested predictions, 23 were confirmed. different methods to help determine the mechanism of action of
This work was extended to de-orphan seven US Food and Drug a small molecule. An integrated approach was used to characterize
Administrationapproved drugs with unknown targets132. SEA has the ability of a well-known natural product, K252a, to potentiate
also been used to predict targets of compounds that were active in a Nrg1-induced neurite outgrowth141. Integrating quantitative pro-
zebrafish behavioral assay133. Out of 20 predictions that were tested teomic results with a lentivirus-mediated loss-of-function screen
experimentally, the authors confirmed 11, with activities ranging to validate candidate target proteins, the authors found that knock-
from 1 nM to 10 M. These results suggest that chemical informa- down of AAK-1 reproducibly potentiated Nrg-1driven neurite
tion alone can be sufficient to make predictions using this approach. outgrowth. Similarly, the mechanism by which another natural prod-
Additional ligand-based target-identification methods have been uct, piperlongumine, selectively kills cancer cells was determined
reported (reviewed in ref. 103). by a combination of direct proteomic affinity-enrichment and short
Network-based approaches extend systems biology to drug- hairpin RNA methods142.
target and ligand-target networks and are now known as systems To determine whether bortezomib neurotoxicity could be attrib-
chemical biology134, network pharmacology135 or systems phar- uted to off-target effects, secondary targets of the proteasome inhibi-
macology136. Such approaches are necessary, as many phenotypes tors bortezomib and carfilzomib were explored143. The authors used
are caused by effects of compounds on multiple targets. An explo- a multifaceted approach, involving screening a panel of purified
ration of the relationship between drug chemical structures, target enzymes, activity-based probe profiling in cells and cell lysates and

nature CHEMICAL BIOLOGY | vol 9 | april 2013 | www.nature.com/naturechemicalbiology 2 37


review article Nature chemical biology doi: 10.1038/nchembio.1199

mining a database of known and predicted proteases, to show that 18. Wyatt, P.G., Gilbert, I.H., Read, K.D. & Fairlamb, A.H. Target validation:
bortezomib inhibits several serine proteases and to hypothesize that linking target and chemical properties to desired product profile. Curr. Top.
its neurotoxicity may be caused by off-target effects. Med. Chem. 11, 12751283 (2011).
19. Kauselmann, G., Dopazo, A. & Link, W. Identification of disease-relevant
Computational data integration and network analysis were genes for molecularly-targeted drug discovery. Curr. Cancer Drug Targets
instrumental in determining Aurora kinase A as a relevant tar- 12, 113 (2012).
get of dimethylfasudil in acute megakaryoblastic leukemia144. 20. Stockwell, B.R. Chemical genetics: ligand-based discovery of gene function.
Dimethylfasudil is a broad-spectrum kinase inhibitor; integrating Nat. Rev. Genet. 1, 116125 (2000).
quantitative proteomics data, kinase inhibition profiling and RNA- 21. Stockwell, B.R. Exploring biology with small organic molecules. Nature 432,
846854 (2004).
silencing data allowed the researchers to generate a testable hypoth- 22. Clemons, P.A. Complex phenotypic assays in high-throughput screening.
esis which not only identified the physiologically relevant target Curr. Opin. Chem. Biol. 8, 334338 (2004).
of dimethylfasudil but also a potential therapeutic target for acute 23. Schreiber, S.L. & Crabtree, G.R. The mechanism of action of cyclosporin A
megakaryoblastic leukemia. Another elegant application of inte- and FK506. Immunol. Today 13, 136142 (1992).
grating transcriptional data with proteomic data was used to dissect 24. Harding, M.W., Galat, A., Uehling, D.E. & Schreiber, S.L. A receptor for
the immunosuppressant FK506 is a cis-trans peptidyl-prolyl isomerase.
the synergy between two multikinase inhibitors in chronic myelog- Nature 341, 758760 (1989).
enous leukemia cell lines145. A combination of proteomic methods 25. Liu, J. et al. Inhibition of T cell signaling by immunophilin-ligand
to measure drug binding in cell lysates, global phosphoproteomics complexes correlates with loss of calcineurin phosphatase activity.
and genome-wide transcriptomics demonstrated synergy between Biochemistry 31, 38963901 (1992).
26. Brown, E.J. et al. A mammalian protein targeted by G1-arresting
danusertib and bosutinib in chronic myelogenous leukemia cells
rapamycin-receptor complex. Nature 369, 756758 (1994).
harboring the BCR-ABLT315I gatekeeper mutation. 27. Yoshida, M., Nomura, S. & Beppu, T. Effects of trichostatins on differentiation
These very recent examples show how powerful the ability to of murine erythroleukemia cells. Cancer Res. 47, 36883691 (1987).
effectively and systematically integrate large sets of disparate data 28. Yoshida, M., Kijima, M., Akita, M. & Beppu, T. Potent and specific
will be in understanding the molecular mechanisms of a small inhibition of mammalian histone deacetylase both in vivo and in vitro by
trichostatin A. J. Biol. Chem. 265, 1717417179 (1990).
molecule in biological systems. When done in a disciplined and
2013 Nature America, Inc. All rights reserved.

29. Taunton, J., Hassig, C.A. & Schreiber, S.L. A mammalian histone
thoughtful manner, such data integration represents a modern deacetylase related to the yeast transcriptional regulator Rpd3p. Science 272,
instantiation of the scientific method, relying on high-throughput 408411 (1996).
technology, data integration and multidisciplinary approaches 30. McNamara, C. & Winzeler, E.A. Target identification and validation of
to provide clues and avenues to new targets and mechanisms of novel antimalarials. Future Microbiol. 6, 693704 (2011).
31. Whitebread, S., Hamon, J., Bojanic, D. & Urban, L. Keynote review: in vitro
small-molecule action. safety pharmacology profiling: an essential tool for successful drug
development. Drug Discov. Today 10, 14211433 (2005).
Received 10 December 2012; accepted 28 January 2013; 32. Xie, L. & Bourne, P.E. Structure-based systems biology for analyzing
published online 18 March 2013 off-target binding. Curr. Opin. Struct. Biol. 21, 189199 (2011).
33. Chen, S. et al. Self-renewal of embryonic stem cells by a small molecule.
References Proc. Natl. Acad. Sci. USA 103, 1726617271 (2006).
1. Sundberg, S.A. High-throughput and ultra-high-throughput screening: 34. Apsel, B. et al. Targeted polypharmacology: discovery of dual inhibitors
solution- and cell-based approaches. Curr. Opin. Biotechnol. 11, 4753 of tyrosine and phosphoinositide kinases. Nat. Chem. Biol. 4, 691699
(2000). (2008).
2. Mayr, L.M. & Bojanic, D. Novel trends in high-throughput screening. 35. Ito, T. et al. Identification of a primary target of thalidomide teratogenicity.
Curr. Opin. Pharmacol. 9, 580588 (2009). Science 327, 13451350 (2010).
3. Koehn, F.E. High impact technologies for natural products screening. 36. Knight, Z.A., Lin, H. & Shokat, K.M. Targeting the cancer kinome through
Prog. Drug Res. 65, 175, 177210 (2008). polypharmacology. Nat. Rev. Cancer 10, 130137 (2010).
4. Lachance, H., Wetzel, S., Kumar, K. & Waldmann, H. Charting, navigating, 37. Burdine, L. & Kodadek, T. Target identification in chemical genetics: the
and populating natural product chemical space for drug discovery. J. Med. (often) missing link. Chem. Biol. 11, 593597 (2004).
Chem. 55, 59896001 (2012). 38. Zheng, X.S., Chan, T.F. & Zhou, H.H. Genetic and genomic approaches to
5. Nielsen, T.E. & Schreiber, S.L. Towards the optimal screening collection: identify and study the targets of bioactive small molecules. Chem. Biol. 11,
npg

a synthesis strategy. Angew. Chem. Int. Edn Engl. 47, 4856 (2008). 609618 (2004).
6. OConnor, C.J., Beckmann, H.S. & Spring, D.R. Diversity-oriented synthesis: 39. Weinstein, J.N. et al. An information-intensive approach to the molecular
producing chemical tools for dissecting biology. Chem. Soc. Rev. 41, pharmacology of cancer. Science 275, 343349 (1997).
44444456 (2012). 40. Hughes, T.R. et al. Functional discovery via a compendium of expression
7. Swinney, D.C. & Anthony, J. How were new medicines discovered? profiles. Cell 102, 109126 (2000).
Nat. Rev. Drug Discov. 10, 507519 (2011). 41. Young, D.W. et al. Integrating high-content screening and ligand-target
8. Terstappen, G.C., Schlupen, C., Raggiaschi, R. & Gaviraghi, G. Target prediction to identify mechanism of action. Nat. Chem. Biol. 4, 5968
deconvolution strategies in drug discovery. Nat. Rev. Drug Discov. 6, (2008).
891903 (2007). 42. Fomina-Yadlin, D. et al. Small-molecule inducers of insulin expression in
9. Garca-Garca, M.J. et al. Analysis of mouse embryonic patterning and pancreatic alpha-cells. Proc. Natl. Acad. Sci. USA 107, 1509915104 (2010).
morphogenesis by forward genetics. Proc. Natl. Acad. Sci. USA 102, 43. Cuatrecasas, P., Wilchek, M. & Anfinsen, C.B. Selective enzyme purification
59135919 (2005). by affinity chromatography. Proc. Natl. Acad. Sci. USA 61, 636643 (1968).
10. Muto, A. et al. Forward genetic analysis of visual behavior in zebrafish. 44. Hirota, T. et al. Identification of small molecule activators of cryptochrome.
PLoS Genet. 1, e66 (2005). Science 337, 10941097 (2012).
11. Fire, A. et al. Potent and specific genetic interference by double-stranded 45. Brehmer, D., Godl, K., Zech, B., Wissing, J. & Daub, H. Proteome-wide
RNA in Caenorhabditis elegans. Nature 391, 806811 (1998). identification of cellular targets affected by bisindolylmaleimide-type
12. Pickart, M.A. et al. Genome-wide reverse genetics framework to identify protein kinase C inhibitors. Mol. Cell Proteomics 3, 490500 (2004).
novel functions of the vertebrate secretome. PLoS ONE 1, e104 (2006). 46. Wissing, J. et al. Chemical proteomic analysis reveals alternative modes of
13. Ecker, A., Bushell, E.S., Tewari, R. & Sinden, R.E. Reverse genetics screen action for pyrido[2,3-d ]pyrimidine kinase inhibitors. Mol. Cell Proteomics
identifies six proteins important for malaria development in the mosquito. 3, 11811193 (2004).
Mol. Microbiol. 70, 209220 (2008). 47. Oda, Y. et al. Quantitative chemical proteomics for identifying candidate
14. Mitchison, T.J. Towards a pharmacological genetics. Chem. Biol. 1, 36 drug targets. Anal. Chem. 75, 21592165 (2003).
(1994). 48. Wang, G., Shang, L., Burgett, A.W., Harran, P.G. & Wang, X. Diazonamide
15. Schreiber, S.L. Chemical genetics resulting from a passion for synthetic toxins reveal an unexpected function for ornithine delta-amino transferase
organic chemistry. Bioorg. Med. Chem. 6, 11271152 (1998). in mitotic cell division. Proc. Natl. Acad. Sci. USA 104, 20682073 (2007).
16. Inglese, J. et al. High-throughput screening assays for the identification of 49. Ong, S.E. et al. Identifying the proteins to which small-molecule probes and
chemical probes. Nat. Chem. Biol. 3, 466479 (2007). drugs bind in cells. Proc. Natl. Acad. Sci. USA 106, 46174622 (2009).
17. Macarron, R. et al. Impact of high-throughput screening in biomedical 50. Fleischer, T.C. et al. Chemical proteomics identifies Nampt as the target of
research. Nat. Rev. Drug Discov. 10, 188195 (2011). CB30865, an orphan cytotoxic compound. Chem. Biol. 17, 659664 (2010).

238 nature chemical biology | vol 9 | april 2013 | www.nature.com/naturechemicalbiology


Nature chemical biology doi: 10.1038/nchembio.1199 review article
51. Shiyama, T., Furuya, M., Yamazaki, A., Terada, T. & Tanaka, A. Design and 80. Mertins, P. et al. iTRAQ labeling is superior to mTRAQ for quantitative
synthesis of novel hydrophilic spacers for the reduction of nonspecific global proteomics and phosphoproteomics. Mol. Cell Proteomics 11,
binding proteins on affinity resins. Bioorg. Med. Chem. 12, 28312841 M111.014423 (2012).
(2004). 81. Mortensen, P. et al. MSQuant, an open source platform for mass
52. Speers, A.E. & Cravatt, B.F. A tandem orthogonal proteolysis strategy for spectrometry-based quantitative proteomics. J. Proteome Res. 9, 393403
high-content chemical proteomics. J. Am. Chem. Soc. 127, 1001810019 (2010).
(2005). 82. Tsou, C.C. et al. MaXIC-Q Web: a fully automated web service using
53. van der Veken, P. et al. Development of a novel chemical probe for the statistical and computational methods for protein quantitation based on stable
selective enrichment of phosphorylated serine- and threonine-containing isotope labeling and LC-MS. Nucleic Acids Res. 37, W661W669 (2009).
peptides. Chembiochem. 6, 22712280 (2005). 83. Cox, J. et al. Andromeda: A peptide search engine integrated into the
54. Fonoviis, M., Verhelst, S.H., Sorum, M.T. & Bogyo, M. Proteomics MaxQuant environment. J. Proteome Res. 10, 17941805 (2011).
evaluation of chemically cleavable activity-based probes. Mol. Cell 84. Margolin, A.A. et al. Empirical Bayes analysis of quantitative proteomics
Proteomics 6, 17611770 (2007). experiments. PLoS ONE 4, e7454 (2009).
55. Verhelst, S.H., Fonovic, M. & Bogyo, M. A mild chemically cleavable linker 85. Fabian, M.A. et al. A small moleculekinase interaction map for clinical
system for functional proteomic applications. Angew. Chem. Int. Ed. Engl. kinase inhibitors. Nat. Biotechnol. 23, 329336 (2005).
46, 12841286 (2007). 86. Boutros, M. & Ahringer, J. The art and design of genetic screens: RNA
56. Evans, M.J., Saghatelian, A., Sorensen, E.J. & Cravatt, B.F. Target discovery interference. Nat. Rev. Genet. 9, 554566 (2008).
in small-molecule cell-based screens by in situ proteome reactivity profiling. 87. Perlstein, E.O., Ruderfer, D.M., Roberts, D.C., Schreiber, S.L. & Kruglyak, L.
Nat. Biotechnol. 23, 13031307 (2005). Genetic basis of individual differences in the response to small-molecule
57. Cisar, J.S. & Cravatt, B.F. Fully functionalized small-molecule probes for drugs in yeast. Nat. Genet. 39, 496502 (2007).
integrated phenotypic screening and target identification. J. Am. Chem. Soc. 88. Pierce, S.E. et al. A unique and universal molecular barcode array.
134, 1038510388 (2012). Nat. Methods 3, 601603 (2006).
58. Park, J., Oh, S. & Park, S.B. Discovery and target identification of an 89. Roemer, T. & Boone, C. Systems-level antimicrobial drug and drug synergy
antiproliferative agent in live cells using fluorescence difference in discovery. Nat. Chem. Biol. 9, 222231 (2013).
two-dimensional gel electrophoresis. Angew. Chem. Int. Ed. Engl. 51, 90. Moffat, J. et al. A lentiviral RNAi library for human and mouse genes
54475451 (2012). applied to an arrayed viral high-content screen. Cell 124, 12831298 (2006).
59. Kawatani, M. et al. The identification of an osteoclastogenesis inhibitor 91. Wang, J. et al. Cellular phenotype recognition for high-content RNA
2013 Nature America, Inc. All rights reserved.

through the inhibition of glyoxalase I. Proc. Natl. Acad. Sci. USA 105, interference genome-wide screening. J. Biomol. Screen. 13, 2939 (2008).
1169111696 (2008). 92. Eggert, U.S. et al. Parallel chemical genetic and genome-wide RNAi screens
60. Saxena, C. et al. Capture of drug targets from live cells using a multipurpose identify cytokinesis inhibitors and targets. PLoS Biol. 2, e379 (2004).
immuno-chemo-proteomics tool. J. Proteome Res. 8, 39513957 (2009). 93. Guertin, D.A., Guntur, K.V., Bell, G.W., Thoreen, C.C. & Sabatini, D.M.
61. Khersonsky, S.M. et al. Facilitated forward chemical genetics using a tagged Functional genomics identifies TOR-regulated genes that control growth
triazine library and zebrafish embryo screening. J. Am. Chem. Soc. 125, and division. Curr. Biol. 16, 958970 (2006).
1180411805 (2003). 94. Knight, Z.A. & Shokat, K.M. Chemical genetics: where genetics and
62. Kim, Y.K. & Chang, Y.T. Tagged library approach facilitates forward pharmacology meet. Cell 128, 425430 (2007).
chemical genetics. Mol. Biosyst. 3, 392397 (2007). 95. Castoreno, A.B. et al. Small molecules discovered in a pathway screen target
63. Tao, S.C., Chen, C.S. & Zhu, H. Applications of protein microarray the Rho pathway in cytokinesis. Nat. Chem. Biol. 6, 457463 (2010).
technology. Comb. Chem. High Throughput Screen. 10, 706718 (2007). 96. Wacker, S.A., Houghtaling, B.R., Elemento, O. & Kapoor, T.M. Using
64. Lomenick, B., Olsen, R.W. & Huang, J. Identification of direct protein transcriptome sequencing to identify mechanisms of drug action and
targets of small molecules. ACS Chem. Biol. 6, 3446 (2011). resistance. Nat. Chem. Biol. 8, 235237 (2012).
65. Chan, J.N. et al. Target identification by chromatographic co-elution: 97. Lamb, J. et al. The Connectivity Map: using gene-expression signatures to
monitoring of drug-protein interactions without immobilization or connect small molecules, genes, and disease. Science 313, 19291935 (2006).
chemical derivatization. Mol. Cell. Proteomics 11, M111.016642 (2012). 98. Iorio, F. et al. Discovery of drug mode of action and drug repositioning
66. Aebersold, R. & Mann, M. Mass spectrometrybased proteomics. Nature from transcriptional responses. Proc. Natl. Acad. Sci. USA 107, 14621
422, 198207 (2003). 14626 (2010).
67. Ong, S.E. & Mann, M. Mass spectrometrybased proteomics turns 99. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based
quantitative. Nat. Chem. Biol. 1, 252262 (2005). approach for interpreting genome-wide expression profiles. Proc. Natl.
68. Blagoev, B. et al. A proteomics strategy to elucidate functional protein- Acad. Sci. USA 102, 1554515550 (2005).
protein interactions applied to EGF signaling. Nat. Biotechnol. 21, 315318 100. Feng, Y., Mitchison, T.J., Bender, A., Young, D.W. & Tallarico, J.A.
(2003). Multi-parameter phenotypic profiling: using cellular effects to characterize
npg

69. Ranish, J.A. et al. The study of macromolecular complexes by quantitative small-molecule compounds. Nat. Rev. Drug Discov. 8, 567578 (2009).
proteomics. Nat. Genet. 33, 349355 (2003). 101. Wagner, B.K. & Clemons, P.A. Connecting synthetic chemistry decisions
70. Rix, U. & Superti-Furga, G. Target profiling of small molecules by chemical to cell and genome biology using small-molecule phenotypic profiling.
proteomics. Nat. Chem. Biol. 5, 616624 (2009). Curr. Opin. Chem. Biol. 13, 539548 (2009).
71. Ong, S.E. et al. Stable isotope labeling by amino acids in cell culture, 102. Haupt, V.J. & Schroeder, M. Old friends in new guise: repositioning of
SILAC, as a simple and accurate approach to expression proteomics. known drugs with structural bioinformatics. Brief. Bioinform. 12, 312326
Mol. Cell Proteomics 1, 376386 (2002). (2011).
72. Daub, H. et al. Kinase-selective enrichment enables quantitative 103. Koutsoukas, A. et al. From in silico target prediction to multi-target drug
phosphoproteomics of the kinome across the cell cycle. Mol. Cell 31, design: current databases, methods and applications. J. Proteomics 74,
438448 (2008). 25542574 (2011).
73. Bendall, S.C. et al. Prevention of amino acid conversion in SILAC 104. Barrett, T. et al. NCBI GEO: mining tens of millions of expression
experiments with embryonic stem cells. Mol. Cell Proteomics 7, profilesdatabase and tools update. Nucleic Acids Res. 35, D760D765
15871597 (2008). (2007).
74. Gygi, S.P. et al. Quantitative analysis of complex protein mixtures using 105. Seiler, K.P. et al. ChemBank: a small-molecule screening and cheminformatics
isotope-coded affinity tags. Nat. Biotechnol. 17, 994999 (1999). resource database. Nucleic Acids Res. 36, D351D359 (2008).
75. Ross, P.L. et al. Multiplexed protein quantitation in Saccharomyces cerevisiae 106. Wang, Y. et al. PubChem: a public information system for analyzing
using amine-reactive isobaric tagging reagents. Mol. Cell Proteomics 3, bioactivities of small molecules. Nucleic Acids Res. 37, W623W633 (2009).
11541169 (2004). 107. Stegmaier, K. et al. Gene expressionbased high-throughput screening
76. Bantscheff, M. et al. Quantitative chemical proteomics reveals mechanisms (GE-HTS) and application to leukemia differentiation. Nat. Genet. 36,
of action of clinical ABL kinase inhibitors. Nat. Biotechnol. 25, 10351044 257263 (2004).
(2007). 108. Hieronymus, H. et al. Gene expression signaturebased chemical genomic
77. Thompson, A. et al. Tandem mass tags: a novel quantification strategy for prediction identifies a novel class of HSP90 pathway modulators. Cancer
comparative analysis of complex protein mixtures by MS/MS. Anal. Chem. Cell 10, 321330 (2006).
75, 18951904 (2003); erratum 75, 4942 (2003); erratum 78, 4235 (2006). 109. Kim, Y.K. et al. Relationship of stereochemical and skeletal diversity of
78. Hsu, J.L., Huang, S.Y., Chow, N.H. & Chen, S.H. Stable-isotope dimethyl small molecules to cellular measurement space. J. Am. Chem. Soc. 126,
labeling for quantitative proteomics. Anal. Chem. 75, 68436852 (2003). 1474014745 (2004).
79. Boersema, P.J., Raijmakers, R., Lemeer, S., Mohammed, S. & Heck, A.J. 110. Paull, K.D. et al. Display and analysis of patterns of differential activity of
Multiplex peptide stable isotope dimethyl labeling for quantitative drugs against human tumor cell lines: development of mean graph and
proteomics. Nat. Protoc. 4, 484494 (2009). COMPARE algorithm. J. Natl. Cancer Inst. 81, 10881092 (1989).

nature CHEMICAL BIOLOGY | vol 9 | april 2013 | www.nature.com/naturechemicalbiology 239


review article Nature chemical biology doi: 10.1038/nchembio.1199

111. Zaharevitz, D.W. et al. Discovery and initial characterization of the 132. Gregori-Puigjan, E. et al. Identifying mechanism-of-action targets for
paullones, a novel class of small-molecule inhibitors of cyclin-dependent drugs and probes. Proc. Natl. Acad. Sci. USA 109, 1117811183 (2012).
kinases. Cancer Res. 59, 25662569 (1999). 133. Laggner, C. et al. Chemical informatics and target identification in a
112. Marton, M.J. et al. Drug target validation and identification of secondary zebrafish phenotypic screen. Nat. Chem. Biol. 8, 144146 (2012).
drug target effects using DNA microarrays. Nat. Med. 4, 12931301 (1998). 134. Oprea, T.I., Tropsha, A., Faulon, J.L. & Rintoul, M.D. Systems chemical
113. Ganter, B. et al. Development of a large-scale chemogenomics database to biology. Nat. Chem. Biol. 3, 447450 (2007).
improve drug candidate selection and to understand mechanisms of 135. Hopkins, A.L. Network pharmacology: the next paradigm in drug
chemical toxicity and action. J. Biotechnol. 119, 219244 (2005). discovery. Nat. Chem. Biol. 4, 682690 (2008).
114. Fliri, A.F., Loging, W.T., Thadeio, P.F. & Volkmann, R.A. Biological spectra 136. Boran, A.D. & Iyengar, R. Systems pharmacology. Mt. Sinai J. Med. 77,
analysis: Linking biological activity profiles to molecular structure. Proc. 333344 (2010).
Natl. Acad. Sci. USA 102, 261266 (2005). 137. Yamanishi, Y., Araki, M., Gutteridge, A., Honda, W. & Kanehisa, M.
115. Berg, E.L., Kunkel, E.J., Hytopoulos, E. & Plavec, I. Characterization of Prediction of drug-target interaction networks from the integration of
compound mechanisms and secondary activities by BioMAP analysis. chemical and genomic spaces. Bioinformatics 24, i232i240 (2008).
J. Pharmacol. Toxicol. Methods 53, 6774 (2006). 138. He, Z. et al. Predicting drug-target interaction networks based on
116. Kauvar, L.M. et al. Predicting ligand binding to proteins by affinity functional groups and biological features. PLoS ONE 5, e9603 (2010).
fingerprinting. Chem. Biol. 2, 107118 (1995). 139. Mei, J.P., Kwoh, C.K., Yang, P., Li, X.L. & Zheng, J. Drug-target interaction
117. Campillos, M., Kuhn, M., Gavin, A.C., Jensen, L.J. & Bork, P. Drug target prediction by learning from local information and neighbors. Bioinformatics
identification using side-effect similarity. Science 321, 263266 (2008). 29, 238245 (2013).
118. Dixon, S.L. & Villar, H.O. Bioactive diversity and screening library selection 140. Bach, S. et al. Roscovitine targets, protein kinases and pyridoxal kinase.
via affinity fingerprinting. J. Chem. Inf. Comput. Sci. 38, 11921203 (1998). J. Biol. Chem. 280, 3120831219 (2005).
119. Perlman, Z.E. et al. Multidimensional drug profiling by automated 141. Kuai, L. et al. AAK1 identified as an inhibitor of neuregulin-1/ErbB4
microscopy. Science 306, 11941198 (2004). dependent neurotrophic factor signaling using integrative chemical
120. Carpenter, A.E. Image-based chemical screening. Nat. Chem. Biol. 3, genomics and proteomics. Chem. Biol. 18, 891906 (2011).
461465 (2007). 142. Raj, L. et al. Selective killing of cancer cells by a small molecule targeting
121. Chen, B., Wild, D. & Guha, R. PubChem as a source of polypharmacology. the stress response to ROS. Nature 475, 231234 (2011).
J. Chem. Inf. Model. 49, 20442055 (2009). 143. Arastu-Kapur, S. et al. Nonproteasomal targets of the proteasome inhibitors
122. Tanikawa, T. et al. Using biological performance similarity to inform bortezomib and carfilzomib: a link to clinical adverse events. Clin. Cancer
2013 Nature America, Inc. All rights reserved.

disaccharide library design. J. Am. Chem. Soc. 131, 50755083 (2009). Res. 17, 27342743 (2011).
123. Plouffe, D. et al. In silico activity profiling reveals the mechanism of action 144. Wen, Q. et al. Identification of regulators of polyploidization presents
of antimalarials discovered in a high-throughput screen. Proc. Natl. Acad. therapeutic targets for treatment of AMKL. Cell 150, 575589 (2012).
Sci. USA 105, 90599064 (2008). 145. Winter, G.E. et al. Systems-pharmacology dissection of a drug synergy in
124. Wolpaw, A.J. et al. Modulatory profiling identifies mechanisms of small imatinib-resistant CML. Nat. Chem. Biol. 8, 905912 (2012).
molecule-induced cell death. Proc. Natl. Acad. Sci. USA 108, E771E780 146. Parsons, A.B. et al. Integration of chemical-genetic and genetic interaction
(2011). data links bioactive compounds to cellular target pathways. Nat. Biotechnol.
125. Cheng, T., Li, Q., Wang, Y. & Bryant, S.H. Identifying compound-target 22, 6269 (2004).
associations by combining bioactivity profile similarity search and public 147. Perlstein, E.O. et al. Revealing complex traits with small molecules and
databases mining. J. Chem. Inf. Model. 51, 24402448 (2011). naturally recombinant yeast strains. Chem. Biol. 13, 319327 (2006).
126. Petrone, P.M. et al. Rethinking molecular similarity: comparing compounds
on the basis of biological activity. ACS Chem. Biol. 7, 13991409 (2012).
127. Nidhi, Glick, M, Davies, J.W. & Jenkins, J.L. Prediction of biological targets Acknowledgments
for compounds using multiple-category Bayesian models trained on This work was supported by US National Institutes of Health Genomics Based Drug
chemogenomics databases. J. Chem. Inf. Model. 46, 11241133 (2006). DiscoveryTarget ID Project grant RL1HG004671, which is administratively linked to the
128. Paolini, G.V., Shapland, R.H., van Hoorn, W.P., Mason, J.S. & Hopkins, A.L. US National Institutes of Health grants RL1CA133834, RL1GM084437 and UL1RR024924.
Global mapping of pharmacological space. Nat. Biotechnol. 24, 805815
(2006). Competing financial interests
129. Keiser, M.J. et al. Relating protein pharmacology by ligand chemistry. The authors declare no competing financial interests.
Nat. Biotechnol. 25, 197206 (2007).
130. Lounkine, E. et al. Large-scale prediction and testing of drug activity on
side-effect targets. Nature 486, 361367 (2012). Additional information
131. Keiser, M.J. et al. Predicting new molecular targets for known drugs. Reprints and permissions information is available online at http://www.nature.com/
npg

Nature 462, 175181 (2009). reprints/index.html. Correspondence should be addressed to P.A.C.

24 0 nature chemical biology | vol 9 | april 2013 | www.nature.com/naturechemicalbiology

Вам также может понравиться