Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

View Article Online / Journal Homepage / Table of Contents for this issue

Chemical Science Dynamic Article Links < C

Cite this: Chem. Sci., 2011, 2, 1656


www.rsc.org/chemicalscience MINIREVIEW
Molecular docking for virtual screening of natural product databases
Dik-Lung Ma,*a Daniel Shiu-Hin Chana and Chung-Hang Leung*b
Received 16th March 2011, Accepted 3rd June 2011
DOI: 10.1039/c1sc00152c
Published on 20 June 2011 on http://pubs.rsc.org | doi:10.1039/C1SC00152C

Molecular docking enables the extraordinary structural diversity of natural products to be harnessed in
an efficient manner. In this mini-review, we highlight recent examples of the use of molecular docking in
virtual screening for the identification of bioactive molecules from natural product databases.

Introduction a new golden age of drug discovery. However, medicinal chem-


ists were soon confronted with painfully disappointing results
The 1990’s heralded a paradigm shift in the way pharmaceutical and the realization dawned that the chemical space was too vast
research was conducted around the world. With new tools and to be systemically explored experimentally.8 Today, despite the
techniques supplied by molecular biology, millions of research
Downloaded on 04 December 2012

increasing financial investment into research and development,


dollars were invested into the development of highly efficient the number of new drugs approved for the market has fallen to
high-throughput screening (HTS) in vitro and in vivo methodol- an all-time low. Bereft of promising leads, large pharmaceutical
ogies that were capable of screening hundreds of thousands of companies have increasingly shifted towards mergers and
compounds each year.1–4 This growth was accompanied by acquisitions in order to fill their pipelines.9,10 Thus, researchers
exciting advances in the field of combinatorial chemistry.5–7 are continually seeking to develop novel methodologies for the
Whereas a typical bench chemist could be expected to possibly efficient discovery of new chemical scaffolds for pharmaceutical
make a few hundred novel compounds each year, the new investigations. This review focuses on the emerging use of
strategies afforded by combinatorial chemistry easily drove that computer-aided high-throughput molecular docking methodol-
number into the thousands. The pharmaceutical industry eagerly ogies to identify bioactive NPs against important therapeutic
adopted these new technologies, which promised to usher in targets, and aims to summarize the recent successes in this area.

a
Department of Chemistry, Hong Kong Baptist University, Kowloon Tong,
Hong Kong. E-mail: edmondma@hkbu.edu.hk Role of natural products in drug research
b
Centre for Cancer and Inflammation Research, School of Chinese
Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong. Nature has provided us with a fascinating cornucopia of complex
E-mail: duncanl@hkbu.edu.hk molecular scaffolds unparalleled in function and diversity. These

Dik-Lung Ma completed his Daniel Shiu-Hin Chan


PhD in 2004 at the University of completed his BSc degree in
Hong Kong under the supervi- Chemistry at the University of
sion of Prof. C.-M. Che. He New South Wales, Sydney,
spent the years 2005–09 at the Australia. He is currently
University of Hong Kong, the appointed as a Research Assis-
Hong Kong Polytechnic tant at the Department of
University, and the Scripps Chemistry at the Hong Kong
Research Institute with Prof. C.- Baptist University. His research
M. Che, Prof. K.-Y. Wong, and interests include development of
Prof. R. Abagyan. His research luminescent oligonucleotide-
mainly focuses on luminescent based assays for biomolecules
sensing for biomolecules and and metal ions.
Dik-Lung Ma metal ions, computer-aided drug Daniel Shiu-Hin Chan
discovery, and inorganic medi-
cines. In 2010 he was appointed
Assistant Professor at the Hong
Kong Baptist University.

1656 | Chem. Sci., 2011, 2, 1656–1665 This journal is ª The Royal Society of Chemistry 2011
View Article Online

natural products (NPs) have been refined over evolutionary time noted that the number of NP-based drugs in clinical trials has
scales for optimal interactions with biomolecules. As the fallen 30% between 2001 and 2008.34
production of any new compound by a living organism entails Tellingly, the shift away from NP research in pharmaceutical
increased costs, the natural selection hypothesis predicts that corporations was engendered not by any intrinsic faults with the
each natural product retained by an organism must be of value to class of natural compounds as a whole, but by the incompati-
the producer. Before the advent of modern synthetic medicinal bility of NP with the highly automated drug discovery method-
chemistry, natural products were the only source of medicinal ologies of HTS. The reasons are many-fold. Firstly, NPs are
compounds available to mankind. Pharmacological investiga- often regarded as highly structurally complex, containing any
tions into traditional and herbal remedies have yielded many number of chiral centres, fused ring systems, and reactive func-
significant discoveries for modern medicine.11–16 For example, tional groups like the hydroxyl. Pursuing the synthesis of
the herb Artemisia annua has been used in traditional Chinese a complicated NP or NP derivative is hardly a trivial task,
medicine for thousands of years for treating malaria and other requiring the construction of intricate ring systems together with
skin diseases, but it was only several decades ago that the active a laborious strategy of protection and deprotection steps that are
ingredient, artemisinin, was isolated.17–19 Huperzine A, isolated not readily amenable to combinatorial methods. Secondly, the
Published on 20 June 2011 on http://pubs.rsc.org | doi:10.1039/C1SC00152C

from the plant moss Huperzia serrta, is an acetylcholinesterase extraction and purification of NPs from plant, marine and
inhibitor investigated for Alzheimer’s disease, though the plant microbial sources can be very difficult, and the precious
itself has been used for centuries by Chinese herbalists for compounds are often obtained in only minute quantities. Issues
treating swelling, fever and blood disoders.20–22 Another highly with supply can become a major bottleneck in NP drug discovery
important drug of natural origin is the anti-cancer paclitaxel especially if the NP itself is to be utilized as a benchmark or as
(taxol), isolated from the tree bark of Taxus brevifolia, and used a synthetic intermediate for lead optimization and further in vivo
for the treatment of breast, ovarian, lung, head and neck testing. Furthermore, structural elucidation still has to be per-
cancer.23 The total number of natural compounds produced by formed on any assay hit obtained from a natural source, whether
plants has been estimated to be over 500 000.24 However, this is
Downloaded on 04 December 2012

from bioassay-guided fractionation or the isolation of a single


believed to represent only a small fraction of the total number of active compound, and its structure would have to be compared
existing NPs, considering the huge number of undocumented to the synthetically-generated NP. Thirdly, the HTS screening of
species in the vast unchartered regions of the biosphere. Marine crude NP extracts can sometimes be problematic due to highly
organisms, in particular, are a tremendous source of secondary polar or lipophilic components in the sample matrix, leading to
metabolites possessing unique structural diversities yet to be false positives or false negatives in the assay result. The screening
tapped by the medicinal chemist.25–27 of crude extract libraries also entails a stringent identification
Until recently, NPs were still regarded as a fundamental and dereplication program to prevent the expenditure of
cornerstone of pharmaceutical research.28–32 However, amidst resources on known or otherwise uninteresting compounds. A
the rapid developments and promises of HTS and combinatorial further complication arises due to the fact that individual
chemistry, the realm of natural products fell somewhat by the compounds in an extract are likely to be present at vastly
wayside. Many major pharmaceutical companies have halted or different concentrations, such that levels of trace components
significantly cut natural product research. A survey of new may be too low for detection, while inactive, highly abundant
chemical entities by Newman and Cragg between the years 1981– compounds may be registered due to non-specific inhibition.
2006 shows that NPs and drugs derived directly from natural Additionally, the intrinsic optical properties of uncharacterized
compounds occupied only 5% and 23% of this class, respectively, NPs in the extract may interfere with fluoresence-based assay
compared to 30% for synthetic compounds.33 It has also been methodologies. Lastly, the synergistic or antagonistic effects of
two or more components may vanish upon isolation of the
primary constituent. By comparison, the screening of large
Chung-Hang Leung completed libraries of pure, characterized, synthetic compounds present in
his PhD in 2002 at City known quantities is regarded as a much easier endeavour.
University of Hong Kong under However, the failure of combinatorial chemistry to yield
the supervision of Prof. W.-F. a significant increase in the number of drug candidates despite
Fong. After completing a five- the powerful testing capabilities of HTS prompted a rethink of
year post-doctoral fellowship at the nature and quality of the chemical libraries themselves. The
the Department of Pharma- major drawback to combinatorial compounds lies in the implicit
cology, Yale University with requirement for efficient reaction and purification methodologies
Prof. Y.-C. Cheng, he was that whilst able to produce large numbers of compounds at high
appointed Research Assistant yield and low cost, severely limits the structural diversity of the
Professor at the University of chemical library. According to a study performed by Feher and
Hong Kong and then at the co-workers, combinatorial compounds tend to contain fewer
Hong Kong Baptist University. chiral centers compared to natural compounds.35 They also
Chung-Hang Leung His primary research interests contain more rotable single bonds and aromatic rings, an artifact
are in anticancer and anti- of the construction process that involves joining many building
inflammatory drug discovery, blocks together in simple reactions. On the other hand, NPs tend
molecular pharmacology and to contain more rigid scaffolds comprising fused, unsaturated
immunology. ring systems, potentially exploiting the entropic advantages

This journal is ª The Royal Society of Chemistry 2011 Chem. Sci., 2011, 2, 1656–1665 | 1657
View Article Online

conferred by rigidity in order to achieve the high biochemical X-ray structure of the biomolecular target that is co-crystallized
affinities and specificities characteristic of NPs.35 with a ligand is especially advantageous since specific features of
Today, HTS is rarely performed using purely combinatorial, the ligand–target interaction can be readily detected. Some
unfocused libraries. Instead, efforts have focused on ‘‘privileged’’ computational softwares such as LIGANDSCOUT are able to
synthetic databases that seek to occupy a more biologically automatically construct a pharmacophore model by identifying
relevant space within the vast universe of possible chemical and performing calculations on the relevant interactions between
structures. Such libraries include databases of existing drugs the small molecule and the receptor. These ligand–targets are
(drug repurposing),36–38 as well as those based upon the principles classified into features such as hydrogen bonding, charge trans-
of ‘‘diversity-orientated synthesis’’ (DIOS)39–41 and ‘‘biology- fer, and lipophilic interactions to construct at 3D pharmaco-
orientated synthesis’’ (BIOS).42–44 A comprehensive review of all phore model. In ligand-based pharmacophore modeling, a 3D
these methodologies is outside the scope of this work, and the structure of the biomolecular target is not required. Rather,
reader is directed to the excellent reviews that have been pub- a collection of known ligands is assembled in order to derive
lished on these topics.36–44 However, despite the best of these representative electrostatic and steric features that are then used
efforts, HTS screening of such carefully constructed libraries can to construct the pharmacophore model. After the construction of
Published on 20 June 2011 on http://pubs.rsc.org | doi:10.1039/C1SC00152C

still realistically only sample but a small portion of the entire the 3D pharmacophore model, a chemical database is screened
chemical space at a time. This is where computer-aided drug against the model in order to extract ligands that possess the
discovery can make an important contribution. complementary functional groups in the correct spatial
arrangement. Since the affinity calculation is based only on the
geometric fit of ligand atoms and groups to the chemical features
Virtual screening in drug discovery
of the model, the strain on computational resources is signifi-
Virtual screening has recently emerged as a powerful technique cantly lower compared to molecular docking. This makes phar-
complementing traditional HTS technologies. Virtual screening macophore modeling suitable as a pre-filter for more demanding
virtual screening methodologies.53 However, a disadvantage is
Downloaded on 04 December 2012

can be broadly defined as the use of computational analysis of


a database of chemical structures to identify possible drug that the calculated affinity values are often not accurate,
candidates for a specific pharmaceutical target, often a particular however, they are still broadly useful for eliminating non-
enzyme or receptor. The major advantage derived from virtual binders. Ligand-based pharmacophore modeling also requires
screening is the tremendous reduction in time and resources prior knowledge of a set of active compounds, which makes it use
required to screen a chemical library of known compounds in unsuitable for novel biomolecular targets. An additional draw-
a drug discovery project. By identifying non-binding compounds back of the pharmacophoric method is that in the absence of
in silico, the number of compounds to be tested in vitro can be structural or mechanistic information about the target, it is not
dramatically reduced, sometimes by orders of magnitudes. Due always possible to anticipate which particular structural features
to this ‘‘weeding out’’ or elimination of inactive chemical struc- of the ligands is important for the receptor–ligand interaction.
tures, the hit rates in the in vitro assays are often much higher Lastly, due to the inherent nature of pharmacophore modeling,
compared to conventional HTS without preliminary virtual screening usually reveals hits that are structurally similar to the
screening. Furthermore, the number of compounds in even the known ligands, rather than leads with novel modes of binding.
largest chemical library is but a small fraction of the total The use of pharmacophore modeling in NP drug research is not
possible number of compounds that could be synthesized in the focus of this review, and the reader is directed to excellent
principle; estimates range up to 1020–24 for the total number of reviews published recently by the groups of Langer54 and
molecules accessible using known synthetic methods.45 Wolber.55
Computer-aided screening is thus a valuable tool to help Compared to the relatively established technique of pharma-
medicinal chemists decide what to synthesize, an advantage over cophore modeling, 3D molecular docking is considered more
the somewhat random nature of combinatorial libraries used complex and computer-intensive. However, exponential
previously. advances in computing processing power and capabilities have
The use of virtual screening methodologies in drug discovery increased the popularity of molecular docking methods (also
and development has been widely reviewed.46–51 Today, compu- called structure-based or receptor-based virtual screening) in
tational chemistry and chemoinformatics play a key role in early drug discovery and development. The use of molecular docking
phase drug research, by identifying the most promising candi- avoids some of the aforementioned drawbacks involved with
dates for experimental investigations. Two major strategies have pharmacophore modeling. Molecular docking requires knowl-
been employed for virtual screening: pharmacophore modeling edge of the 3D structure of the biomolecular target with or
and molecular docking (Fig. 1). Within the realm of pharmaco- without a bound ligand, at atomic resolution. Use of a co-crystal
phore modeling, the two techniques of structure-based phama- structure of a biomolecular target with a ligand entails several
cophore modeling and ligand-based pharmacophore modeling advantages. Firstly, the search area for the in silico docking can
can be distinguished. In the former, the knowledge of the three- be restricted to the binding site only (alternatively, the binding
dimensional (3D) structure of the biomolecular target is analyzed site may be predicted from homology with similar proteins). An
to identify features of the binding site that are important for unnecessarily large search area can increase the rate of false
ligand binding affinity and selectivity. The 3D structure may be positives due to the binding of virtual molecules outside the
solved by X-ray crystallography, NMR studies or constructed by actual binding site. Furthermore, this can also lead to wastage in
homology with related proteins, and over 73 000 of such struc- computer resources. Secondly, prior knowledge of critical
tures are freely available at the Protein Data Bank (PDB).52 An ligand–target interactions can help identify false positives in the

1658 | Chem. Sci., 2011, 2, 1656–1665 This journal is ª The Royal Society of Chemistry 2011
View Article Online
Published on 20 June 2011 on http://pubs.rsc.org | doi:10.1039/C1SC00152C

Fig. 1 Schematic flowchart outlining the two major strategies employed in in silico virtual screening.

virtual screening. Third, more accurate docking calculations can for screening natural compounds by bypassing many of these
be performed since the target is in its active or induced confor- tedious steps. For example, non-binders can be predicted in silico,
mation. A computational model of the target is constructed from avoiding the wastage of scarce NPs. Additionally, practical
the 3D structure, and ligands from the chemical database are difficulties associated with bioassay-guided fractionation and
sequentially docked against the receptor. Most docking dereplication are made irrelevant. One minor drawback is that
programs incorporate ligand flexibility into the docking calcu- it is no longer possible to isolate totally new bioactive
Downloaded on 04 December 2012

lations so that the binding geometry of the ligand can be cor- compounds, as from a NP extract.
rected predicted. However, the target is usually assumed to be It is common knowledge that the results of any HTS exercise is
mostly rigid, as the explicit inclusion of receptor flexibility in the ultimately predicated upon the quality of the compound collec-
docking calculations would be too computationally demanding. tion itself. Poorly designed libraries lacking sufficient diversity
Some docking algorithms attempt to model receptor flexibility in may result in few hits. Fortunately, NP libraries possess an
an indirect way, for example by tolerating some degree of steric intrinsic advantage in diversity of molecular scaffolds due to
clashing with the ligand without the explicit repacking of the their exquisite relationships with biomolecules that have been
receptor side chains, or by using several alternative receptor streamlined over evolutionary time scales. Furthermore, many
binding site conformations for docking and then merging the NP compound collections also contain NP-like structures that
results. Some modern docking algorithms are able to explicitly are based upon naturally occurring scaffolds. One should also
model receptor flexibility, but this is usually constrained to the note that NPs often contain more than one violation of Lip-
ligand binding domain in order to conserve computing inski’s ‘‘rule-of-fives’’,62 especially with regards to molecular
resources.56,57 A RMSD of <2 A  from the predicted binding pose weight and hydrogen bond acceptors, and too-strict adherence to
to the X-ray crystal structure is considered satisfactory. these rules in a screening campaign would certainly act to negate
After the optimal binding poses have been predicted for each the inherent advantages of structural diversity conferred by the
compound in the database, the next step is to rank or score the use natural compounds. A summary of some available NP
structures to determine their relative binding affinities. Scoring databases is presented in Table 1.
functions perform calculations involving statistical potentials or
weighted interaction terms that have been previously calibrated
Recent successes of molecular docking of natural
with ‘‘training sets’’ of known binders and non-binders. However,
it has been noted that scoring functions represent the major
products for drug discovery
weakness in docking programs; while the docking algorithm is We discuss here some recent examples of the use of molecular
relatively accurate at predicting the preferred binding mode, the docking for the discovery of bioactive NPs in the last decade. For
ability of the scoring functions to distinguish strong and weak ease of access, we have grouped docking targets into three
binders still leads to a significant number of false positives in the classes: enzyme–substrate interactions, receptor–ligand interac-
virtual screen. Consensus scoring, the use of multiple scoring tions (including protein–protein interactions) and DNA
functions in concert, has been found to significantly improve hit interactions.
rates compared to the use of a single scoring function.58–61
As discussed in the previous section, despite the astonishing
Natural products targeting the enzyme–substrate interaction
structural diversity and fascinating molecular architecture
exhibited NPs, their use in HTS has been somewhat neglected In 2004, Toney and co-workers screened the National Cancer
compared to purely synthetic libraries based on drug-likeness, Institute (NCI) diversity set of 1853 compounds of both natural
DIOS or BIOS. The screening of isolated natural compounds and synthetic origin against 3CLpro proteinase of the severe acute
and NP extracts has often been regarded as too ‘‘dirty’’ and too respiratory syndrome coronavirus (SARS-CoV), the causative
expensive, and involving additional time and labour-consuming agent for a pandemic that swept Southeast Asian regions in
efforts in isolation, fractionation, characterization, and 2003.63 The compounds were docked against the X-ray crystal
dereplication. Computational methods open up new possibilities structure of 3CLpro (PDB entry: 1P9S) using the AutoDock

This journal is ª The Royal Society of Chemistry 2011 Chem. Sci., 2011, 2, 1656–1665 | 1659
View Article Online

Table 1 Some available natural product databases for virtual screening

Number of
Database Company compounds Link

ZINC Bioinfomatics and Chemical Informatics 89 425 http://zinc.docking.org/vendor0/npd/


Research Center (BCIRC) index.html
MEGabolite Analyticon Discovery 4 700 http://www.ac-discovery.com
NatDiverse Analyticon Discovery >20 000 http://www.ac-discovery.com
IBS Database InterBioScreen Ltd >45 000 http://www.ibscreen.com/natural.
shtml
Super Natural Database Charite, Medical Faculty of the 45 917 http://bioinformatics.charite.de/
Humboldt-University supernatural/
CHEMnetBASE (Dictionary of Natural Chapman & Hall >226 000 http://www.chemnetbase.com/
Products)
CHEMnetBASE (Dictionary of Marine Chapman & Hall >34 000 http://www.chemnetbase.com/
Natural Products)
Published on 20 June 2011 on http://pubs.rsc.org | doi:10.1039/C1SC00152C

Natural Products Database (NPD) Molecular Diversity Preservation 22 048 http://www.mdpi.org/


International (MDPI)
Natural Compound Library (NCL) TimTec 640 http://www.timtec.net/
Natural Derivative Library (NDL) TimTec 3 000 http://www.timtec.net/
SPECS Natural Products SPECS, Inc. 400 http://www.specs.net/
CNPD NeoTrident Technology Ltd 57 000 http://www.neotrident.com/newweb/
index.asp
Downloaded on 04 December 2012

program. The highest scoring compound was found to be the utilized a consensus docking and scoring approach that includes
natural product sabadinine, which showed a binding energy of four docking algorithms (MOE-Dock, Glide, Fred and Gold)
11.6 kcal mol1 and a clustering of 9 out of 10 docked and five scoring functions (MOE-Score, GlideScore, GoldScore,
conformers within 0.5 A.  However, sabadinine did not affect ChemScore and Xscore) to dock and rank the NP database
murine coronavirus replication at 100 mM, as gauged by syncy- against the X-ray crystal structure of CK2 (PDB entry: 1JWH).
tium formation and cytopathic effects. Significantly, ellagic acid ranked in the top 5% of all five scoring
Sangma and co-workers (2005) utilized a combined approach functions. Furthermore, all four docking programs predicted the
of molecular docking and neural networks to identify inhibitors same lowest energy binding conformation for ellagic acid. This
of HIV-1 reverse transcriptase (RT) and HIV-1 protease (PR) promising in silico result was validated by the biological verifi-
from a Thai Medicinal Plants Database.64 2684 compounds were cation experiments, which showed that ellagic acid represented
docked against the X-ray co-crystal structures of HIV-1 RT with the most potent CK2 inhibitor reported at that time (Ki ¼
nevirapine or calanolide A (PDB entry: 1VRT) and HIV-1 PR 20 nM). Kinetic analysis revealed ellagic acid to be a competitive
with XK-263 (PDB entry: 1HVR) using AutoDock, and then the inhibitor with respect to ATP, which was consistent with the
results were applied to a neural network based on a self-orga- molecular docking results that showed binding of the small
nizing map (SOM) in order to reduce the size of the hit list. In the molecule to the ATP-binding domain. Importantly, ellagic acid
SOM approach, the reference structures are analyzed to find was shown to be at least 72-fold more potent against CK2 kinase
potential pharmacophoric groups, and for easier analysis, a map (IC50 ¼ 0.04 mM) compared to a panel of 11 other kinases (IC50 >
was generated that contained only the distances between three 2.9 mM), a remarkable result considering the ostentatiously
points of certain pharmacophoric groups. The same maps were promiscuous structural features of ellagic acid (a planar
generated from the docking hit list and then superimposed onto compound containing four hydroxyl moieties) and the fact that
the reference map, and only the compounds having their features this virtual screening hit was not optimized at all. This early work
represented in the common regions were reselected. This helped to demonstrate the power of molecular docking for
successive SOM screening could be repeated as many times as identifying potent and selective NPs against therapeutic biomo-
necessary (by using different features) to reduce the size of the lecular targets.
candidate pool. For example, AutoDock identified 562 (out of In 2008, Fu et al. identified Jadomycin C as an inhibitor of
2684 compounds) docking hits for HIV-1 PR, but this was Aurora B kinase by employing molecular docking of the
reduced to 135 and then 13 after successive rounds of SOM Microbial Natural Products Database containing about 15 000
screening. However, most of these compounds had already been natural compounds of microbial origin against the X-ray co-
reported to show anti-HIV activity; no biological validation was crystal structure of Aurora-B cocrystallized with the Hesperadin
performed in this work. This study demonstrates that using (PDB entry: 2BFY).66 Aurora kinases play key roles in cell
a pharmacophoric SOM neural network after molecular docking division through the regulation of centrosome maturation and
is a computationally inexpensive method of reducing the size of separation, microtubulue-kinetochore attachment, and chro-
the virtual screening hit list. mosome alignment and segregation. Consequently, Aurora
In 2006, the group of Moro identified ellagic acid, a naturally kinases have been found to be overexpressed in a variety of
occurring derivative of tannic acid, as an inhibitor of casein cancers, and inhibitors of such kinases, including Hesperadin,
kinase 2 (CK2), a putative oncogene in animal and cellular ZM447439 and VX-680, have been showed significant anticancer
models.65 Using an in-house database of around 2 000 NPs, they activity in vitro and in vivo. Using the docking program FlexX,

1660 | Chem. Sci., 2011, 2, 1656–1665 This journal is ª The Royal Society of Chemistry 2011
View Article Online

a hit list of the 150 top scoring structures were obtained from the 6,600 -biapigenin can be considered a new class of NAE inhibitor
virtual screening campaign, of which 22 were procured for (Fig. 2). Significantly, 6,600 -biapigenin was only the second NAE
experimental testing. The most promising candidate, Jadomycin inhibitor reported to date. While further kinetic or structural
B, could inhibit the activity of Aurora-B in vitro with an IC50 evidence would be required to definitively establish the mecha-
value of 10.5 mM and a Ki value of 6.8 mM. Jadomcyin was also nism of inhibition of NAE by 6,600 -biapigenin, this study high-
able to inhibit the proliferation of a variety of cancer cell lines lights some of the chief advantages of molecular docking over
with IC50 values in the range of 10–26 mM. However, because ligand-based pharmacophore screening. Firstly, for a relatively
Jadomycin B induced apoptosis without blocking the cell cycle, new biomolecular target such as NAE, ligand-based pharmaco-
the putative target mechanism of Aurora-B inhibition, the phore modeling is often impossible due to the fact that inhibitors
authors concluded that Jadomycin B could potentially inhibit of the target have not yet been discovered. Secondly, since the
other kinases more effectively resulting in apoptosis. Due to the structures in the screening library are computed against the
highly conserved nature of the kinase ATP-binding domain, pharmacophoric model of known inhibitors, pharmacophore
achieving selectivity amongst the 518 protein kinases in the screening is inherently unable to identify bioactive compounds
human genome is often the paramount concern in kinase inhib- with a novel mode of binding, and such compounds would be the
Published on 20 June 2011 on http://pubs.rsc.org | doi:10.1039/C1SC00152C

itor discovery projects. It would have therefore been interesting first to fall out of the virtual screening campaign due to their
for the authors to investigate the activity of Jadomycin B against incongruous binding mode. Besides its use in virtual screening,
panel of different kinases using both in silico and in vitro molecular docking can also be used to explore the mode and
methods, in order to evaluate the ability of molecular docking to mechanism of enzyme inhibition by natural products. For
predict the selectivity profile of their NP screening hit. example, Monti et al. have used Molsoft to analyze the binding
In 2011, our research group reported the discovery of the NP- of the marine natural products including scalaradial69 and
like 6,600 -biapigenin as only the second inhibitor of NEDD8- petrosaspongiolide M70 to phospholipase A2, a target for
activating enzyme (NAE) using molecular docking.67 NAE is an inflammation.
Downloaded on 04 December 2012

analogue of the ubiquitin E1 enzyme that is involved with


regulating the ubuiqitination and degradation of the subset of
Natural products targeting the receptor–ligand interaction
proteins regulated by E3 cullin-RING ligases, including cancer-
related substrates such as p-IkBa and c-myc. The first inhibitor of In 2003, Liu et al. screened the China Natural Products Database
NAE reported, MLN4924, displayed efficacy against both solid of 50 000 compounds against a structural model of the potassium
and hematological human cancer cell lines.68 We performed high- ion channel using Dock.71 The 3D model of eukaryotic Shaker
throughput molecular docking of the Analyticon MEGAbolite K+ channel was constructed based on homology with the crystal
and NatDiverse databases of over 20 000 NP and NP-like structure of prokaryotic Streptomyces Kcsa K+ channel (PDB
structures against the X-ray crystal structure of the quaternary entry: 1BL8). The search area for the molecular docking was
APPBP1-UBA3-NEDD8-ATP complex (PDB: 1R4N) using the restricted to the extracellular pore binding site formed by the
docking software Molsoft. The 10 highest-scoring compounds tetramer. Attention was focused on potential ligands that inter-
were purchased and tested in biological validation experiments, acted with the residues surrounding the entry of the ‘‘ion-selec-
and the biflavonoid 6,600 -biapigenin emerged as the top candi- tive filter’’ of the channel so that the compounds could inhibit K+
date, with micromolar potencies against NAE in enzyme and channel function. To reduce the size of the initial hit list, the top
cell-based assays. While 6,600 -biapigenin itself has not been found 200 compounds were reoptimized using the Sybyl molecular
in nature, it has been synthesized from 6,600 -biapigenin hexa- mechanics force field, yielding a final collection of 14 compounds
aceate, in turn obtained from the naturally occurring succeda- of which 4 were procured for biological testing. All 4 compounds
neaflavanone (6,600 -binarigenin). Interestingly, molecular inhibited the K+ channel in the electrophysiological assay by
modeling analysis of the ligand–receptor interaction indicated whole-cell voltage-clamp recording in dissociated hippocampal
a binding mode very different to that of ATP or the nucleotide rat neurons, with 20–1000-fold higher potency than tetraethyl-
mimic NAE inhibitor MLN4924, tentatively suggesting that ammonium, a well-known K+ channel blocker.

Fig. 2 Molecular model of a) virtual screening hit 6,600 -biapigenin, b) MLN4924 and c) ATP bound to the NAE heterodimer generated by virtual ligand
docking. Molecular modeling analysis revealed a putatively different binding mode of 6,600 -biapigenin compared to MLN4924 or ATP.67

This journal is ª The Royal Society of Chemistry 2011 Chem. Sci., 2011, 2, 1656–1665 | 1661
View Article Online

Brinton and co-workers (2005) docked an in-house database of only 6 out of the 29 compounds showed biological activity, and
25 000 plant-based NP and NP derivatives against the X-ray co- all 6 were from the flavonoid family, with activities approxi-
crystal structure of estrogen receptor-b (ERb) with the isoflavone mately 3.6–21-fold times less potent than rosiglitazone. Consid-
genistein (PDB entry: 1QKM) using the docking software Gold.72 ering the considerable flexibility exhibited by the PPAR-g ligand
ERb is postulated to play key roles in maintaining estrogen- binding domain, the authors used an induced-fit docking (IFD)
inducible neuronal morphological plasticity, brain development, protocol to further analyze the binding mode of the flavonoids.
and cognition. On the other hand, ERa is more predominantly Receptor flexibility was modeled using the protein structure
expressed in the breast and the endometrium. Therefore, ER prediction and refinement package Prime in combination with
agonists for the treatment of neurodegenerative diseases should Glide. Significantly, induced-fit docking of the flavonoids into
display high selectivity for ERb over ERa to avoid unwanted the inferior model (the rosiglitazone-induced receptor) resulted
profilerative effects in other tissues or organs. The top 500 scoring in significant structural rearrangements in the protein side chains
molecules from the molecular docking were filtered by visual of the ligand-binding domain, reproducing the superior, lower-
inspection and 100 compounds were manually selected for further energy binding pose observed in the farglitazar model. This study
analysis by the docking program Affinity, to refine the binding showed that induced-fit docking can be used to reveal a more
Published on 20 June 2011 on http://pubs.rsc.org | doi:10.1039/C1SC00152C

modes predicted by Gold. The final result was that 31 compounds plausible binding mode than by rigid-docking alone, allowing for
were selected that possessed the critical hydrogen bond with even significant rearrangements of the protein scaffold in order
His475 of ERb as well as necessary hydrophilic and hydrophobic to better accommodate the docked ligand. However, the protein
characteristics matching that of endogenous 17b-estradiol or side-chain prediction and geometry minimization calculations
genistein. Of the 12 tested compounds, 5 molecules displayed are more computationally demanding compared to those in
significant selectivity for ERb over ERa (3 of these exhibited over rigid-receptor docking. Furthermore, as with all molecular
100-fold selectivity). However, the neurobiological effects of these models, the induced-fit binding pose would still have to be veri-
compounds will require further in vivo investigations as the fied with experimental techniques such as NMR or X-ray
Downloaded on 04 December 2012

agonistic or antagonistic activities of weak estrogenic binders are crystallography.


known to be dependent on a number of factors, such as the In 2008, the group of Ngai and Archer reported the discovery
concentration of the drug, the concentrations of endogenous of high-affinity agonists of an olfactory G protein-coupled
estrogens, as well as the level of transcriptional coactivators or receptor (GPCR) using both receptor-based and ligand-based
corepressers present in the particular tissue.73 virtual screening.76 The GPCR responds preferentially to long
Peroxisome proliferator-activated receptor-gamma (PPAR-g) chain amino acids such as lysine and arginine, as well as to other
plays a critical role in lipid and glucose homeostasis, and inhib- amino acids with lower affinity. This relatively non-specific
itors of PPAR-g, such as the synthetic thiazolidinediones, are binding profile is characteristic of odorant receptors, which
potential therapeutic agents for the treatment of type II dia- allows the olfactory system to recognize a diversity of chemical
betes.74 Notable side effects associated with such compounds, structures exceeding the actual number of olfactory receptors in
including fluid retention, weight gain, and cardiac hypertropy the human genome. The authors wished to identify novel high-
have recently stimulated Hibbs and co-workers (2008) to affinity agonists of the V2R vomeronasal receptor-like goldfish
discover new NP scaffolds as PPAR-g agonists.75 The authors receptor 5.24 for use as molecular probes to study receptor
docked an in-house natural product database of 200 compounds structure and olfactory function. Interestingly, while a database
against two structurally distinct X-ray co-crystal structures of comprising over 1.6 million commercially available compounds
PPAR-g using rigid-receptor docking with Glide. One PPAR-g was used for the molecular docking and pharmacophore-
structure was co-crystallized with the thiazolidinedione rosigli- screening, the most active hit compounds were all naturally
tazone (PDB entry: 1FM6), while the other incorporated the occurring, linear amino acid derivatives, and these were found to
tyrosine-based analogue farglitazar (PDB entry: 1FM9), one of be more potent agonists than the natural amino acids. For
the earliest anti-diabetic lead compounds. Termed ‘‘multiple example, diaminopimelic acid is a component of Gram-negative
rigid-receptor docking’’, the authors proposed that the use of two bacterial cell walls and is used as an intermediate bacterial
structurally distinct induced-fit conformations of the protein biosynthetic pathways for lysine and peptidoglycans. Further-
would admit a wider range of ligands, particularly if the hits more, the hit rate for the common virtual hits that scored highly
achieved ligand–receptor interactions comparable to one or both in both the molecular docking and pharmacophore screening was
of the known inhibitors. The 20 best-scoring compounds for each extremely high (16/36 ¼ 44%), with activity defined as >40%
of the two conformations were pooled together, and duplicates maximum receptor activity at 20 mM of the compound. Four of
(those compounds scoring highly for both conformations) were these compounds were active as olfactory receptor agonists in
removed, to achieve a hit list of 29 unique candidates, that were vivo as measured by electrophysiological recordings of goldfish
observed to fall into three structural classes: the flavonoids, the olfactory epithelium. Furthermore, molecular modeling was
gingeroids, and the ginkolides. However, visual inspection of the performed to analyse specific features of the amino acid ligands
binding poses revealed that only the flavonoids were able to that may confer agonist or antagonist activities, based on the
engage key protein residues in the transcriptional activation structural analysis of the receptor’s ligand binding domain.
funtion 2 domain (AF-2 helix) characteristic of PPAR-g Compared to enzyme–substrate or ligand–receptor interac-
agonists, and all of the flavonoids achieved much better docking tions, targeting the protein–protein interface is usually regarded
scores with the farglitazar-induced receptor rather than the as more difficult due to the relatively large and featureless nature
rosiglitazone-induced receptor. Biological validation using of most protein–protein binding pockets, that lack clearly-
a cellular transcriptional factor assay confirmed this analysis as defined binding crevices or mechanism-based contacts.77 In 2010,

1662 | Chem. Sci., 2011, 2, 1656–1665 This journal is ª The Royal Society of Chemistry 2011
View Article Online

our research group wondered if the enormous structural diversity (IC50 ¼ ca. 5 mM) had comparable activity to SPD304 (IC50 ¼ ca.
of NPs could be harnessed to discover novel small molecule 3 mM) against cellular TNF-a induced NF-kB luciferase activity.
inhibitors of tumor necrosis factor-a (TNF-a).78 TNF-a is These compounds also represented only the third and fourth
a multifunctional cytokine that acts as a central biological examples of TNF-a inhibition by a small molecule inhibitor at
mediator for critical immune responses, including inflammation, that time. This study demonstrates that despite the seemingly
infection, and apoptotic cell death. Dysregulation of TNF-a has intractable nature of the protein–protein interface, molecular
been implicated in cases of tumorigenesis, diabetes, and espe- modeling can be an effective tool for harnessing the structural
cially in autoinflammatory diseases such as rheumatoid arthritis, diversity of NPs towards identifying novel inhibitors of the
psoriatic arthritis and Crohn’s disease.79 Using the software protein–protein interaction.
Molsoft, over 20 000 NP and NP-like structures from the Ana-
lyticon MEGAbolite and NatDiverse databases were docked
Natural products targeting DNA
against the X-ray co-crystal structure of TNF-a dimer with
SPD304 (PDB code: 2AZ5), the strongest TNF-a small molecule DNA-targeting NPs are some of the most well-known anti-
inhibitor reported to date. The top 16 compounds from the cancer drugs. For example, mitomycin is a potent DNA cross-
Published on 20 June 2011 on http://pubs.rsc.org | doi:10.1039/C1SC00152C

virtual screening were obtained and tested in vitro, and two NP- linker from Streptomyces caespitosus or Streptomyces lavendulae
like compounds, a pyrazole-linked quinuclidine and an indolo that finds use as a chemotherapeutic against gastrointestinal,
[2,3-a]quinolizidine, emerged as the top candidates. Despite the breast, and bladder cancers,80 while the actinomycins, also from
mostly hydrophobic, formless nature of the TNF-a binding Streptomyces, are chemotherapeutics and antibiotics that bind
pocket, both compounds occupied a similar region in the binding DNA at the transcription initiation complex and inhibiting RNA
site compared to SPD304 (Fig. 3). Furthermore, both polymerase.81 Recently, non-canonical secondary structures of
compounds were observed to be large enough to contact both DNA such as the G-quadruplex have emerged as attractive
subunits of the TNF-a dimer simultaneously, thus occupying targets for therapeutic intervention due to their putative associ-
Downloaded on 04 December 2012

and blocking the binding site for the third TNF-a subunit and ation with oncogene promoter sequences such as c-myc, bcl-2,
preventing the formation of the biologically active TNF-a trimer VEGF, KRAS, and c-kit.82–84 While the NP telomestatin is the
complex. Molecular modeling analysis indicated the absence of most potent G-quadruplex ligand known to date,85 the majority
extensive hydrogen bonding or salt bridge formation which was of G-quadruplex binding ligands reported in the literature have
consistent with our knowledge of the TNF-a binding pocket and been synthetic in origin. In 2010, our research group applied
with the reported binding mode SPD304. The indoloquinolizi- high-throughput structure-based virtual screening methods to
dine (IC50 ¼ ca. 10 mM) was found to be more potent against identify natural product ligands of the c-myc G-quadruplex, as
TNF-a in the receptor binding assay compared to SPD304 potential chemotherapeutic agents.86 In order to develop a high-
(IC50 ¼ 22 mM by a comparable ELISA), while the quinuclidine throughput screening platform for G-quadruplex binding

Fig. 3 Low-energy binding conformations of a) screening hit quinuclidine, b) screening hit indolo[2,3-a]quinolizidine, c) SPD304 bound to TNF-
a dimer and d) superimposition of quinuclidine (orange), indolo[2,3-a]quinolizidine (yellow) and SPD304 (blue) generated by virtual ligand docking.78

This journal is ª The Royal Society of Chemistry 2011 Chem. Sci., 2011, 2, 1656–1665 | 1663
View Article Online

ligands, a model of the intramolecular c-myc G-quadruplex pharmacophore modeling, has the potential to accurately predict
1 : 2 : 1 loop isomer was constructed using the X-ray crystal binding affinities of screening hits as well as potentially reveal
structure of the closely related intramolecular human telomeric lead structures with novel modes of binding. As scoring algo-
G-quadruplex DNA (PDB entry: 1KF1), since neither the NMR rithms become more refined, together with the continuous
nor X-ray crystallographic information for the predominant c- improvement in computer processing power and capabilities, we
myc 1 : 2 : 1 loop isomer was available. Using Molsoft, we believe that molecular docking has great promise in virtual lead
docked the Analyticon MEGAbolite and NatDiverse databases discovery. In this review, we have highlighted some recent
of over 20 000 NP and NP-like structures against our the model examples of the use of molecular docking for the identification of
of the c-myc G-quadruplex, and the top 5 scoring compounds bioactive compounds from natural product databases. These
were tested in an in vitro assay. The naphthopyrone pigment works demonstrate that molecular modeling can be used to
fonsecin B, isolated from the fungus Aspergillus fonsecaeus, reveal natural products as highly potent and selective enzyme
emerged as the top candidate. Fonsecin B was found to inhibit inhibitors, blockers of protein-protein interactions, as well as
Taq-mediated extension through stabilization of the G-quad- ligands targeting non-canonical DNA structures. Given that
ruplex secondary structure (IC50 ¼ ca. 20 mM), with potency most of these studies were published within the last three years,
Published on 20 June 2011 on http://pubs.rsc.org | doi:10.1039/C1SC00152C

comparable to the well-known G-quadruplex binder TMPyP4. we are confident that this powerful combination of molecular
The molecular docking analysis revealed that fonsecin B was docking and natural compounds will continue to thrive as an
stacked against the end of the G-quadruplex at the 30 -terminus, active area of research in the coming years.
with potential electrostatic interactions with a potassium ion in This work is supported by the Hong Kong Baptist University
the central channel (Fig. 4). Importantly, this study represented (FRG2/09-10/070 and FRG2/10-11/008), Centre for Cancer and
the first large scale high-throughput virtual screening of a NP Inflammation Research, School of Chinese Medicine (CCIR-
database against the c-myc G-quadruplex. To our knowledge, no SCM, HKBU) and The Hong Kong Anti-Cancer Society
other study has been published that utilizes high-throughput (HKACS) Research Grant.
Downloaded on 04 December 2012

molecular docking to discover novel NP or NP-like nucleic acid-


binding compounds. With a growing awareness of the important
regulatory roles played by non-canonical DNA or RNA motifs
in human biology, as well as the ready availability of such nucleic
Notes and references
acid structures in the PDB, we believe that this is an area that 1 J. J. Burbaum and N. H. Sigal, Curr. Opin. Chem. Biol., 1997, 1, 72–
definitely deserves further attention in the literature. 78.
2 R. P. Hertzberg and A. J. Pope, Curr. Opin. Chem. Biol., 2000, 4, 445–
451.
3 K. H. Bleicher, H.-J. B€ohm, K. M€ ullee and A. I. Alanine, Nat. Rev.
Conclusions Drug Discovery, 2003, 2, 369–378.
4 P. B. Fernandes, Curr. Opin. Chem. Biol., 1998, 2, 597–603.
Computational technologies are a practical solution to the
5 J. W. Szostak, Chem. Rev., 1997, 97, 347–348.
horrific experimental costs associated with high-throughput 6 H. Mario Geysen, F. Schoenen, D. Wagner and R. Wagner, Nat. Rev.
screening of large compound libraries. A variety of modeling Drug Discovery, 2003, 2, 222–230.
techniques are available for today’s medicinal chemists for the 7 E. M. Gordon, M. A. Gallop and D. V. Patel, Acc. Chem. Res., 1996,
29, 144–154.
rapid and efficient discovery of lead molecules against biomo- 8 S. S. Phatak, C. C. Stephan and C. N. Cavasotto, Expert Opin. Drug
lecular targets. Meanwhile, natural products are re-emerging as Discovery, 2009, 4, 947–959.
a valuable source of bioactive scaffolds that display remarkable 9 S. Shibayama, K. Tanikawa, R. Fujimoto and H. Kimura, Drug
chemical diversity in structure and function. The use of virtual Discovery Today, 2008, 13, 86–93.
10 L. Schweizer and D. Z. Knyphausen-Aufsess, in Handbook of
screening technologies ameliorates many of the problems asso- Bioentrepreneurship, ed. Z. J. Acs and D. B. Audretsch, Springer
ciated with the incompatability of natural products with high- New York, 2008, pp. 129–144.
throughput screening. The combination of virtual screening and 11 B. Patwardhan and R. A. Mashelkar, Drug Discovery Today, 2009,
14, 804–811.
natural products allows the medicinal chemist to harness the 12 D. Normile, Science, 2003, 299, 188–190.
extraordinary potential of natural products in an efficient and 13 W. Y. Jiang, Trends Pharmacol. Sci., 2005, 26, 558–563.
inexpensive manner. Molecular docking, while regarded as more 14 M. Heinrich, Phytochem. Lett., 2008, 1, 1–5.
complex and computationally demanding compared to 15 M. Heinrich, Frontiers in Pharmacology, 2010, 1, 8.
16 L. Bohlin, U. Goeransson, C. Alsmark, C. Weden and A. Backlund,
Phytochem. Rev., 2010, 9, 279–301.
17 N. J. White, Science, 2008, 320, 330–334.
18 D. L. Klayman, Science, 1985, 228, 1049–1055.
19 S. R. Meshnick, T. E. Taylor and S. Kamchonwongpaisan, Microbiol.
Rev., 1996, 60, 301–305.
20 D. H. Cheng, H. Ren and X. C. Tang, NeuroReport, 1996, 8, 97–101.
21 J.-S. Liu, Y.-L. Zhu, C.-M. Yu, Y.-Z. Zhou, Y.-Y. Han, F.-W. Wu
and B.-F. Qi, Can. J. Chem., 1986, 64, 837–839.
22 A. P. Kozikowski and W. Tueckmantel, Acc. Chem. Res., 1999, 32,
641–650.
23 M. C. Wani, H. L. Taylor, M. E. Wall, P. Coggon and A. T. McPhail,
J. Am. Chem. Soc., 1971, 93, 2325–2327.
24 L. Zhang, A. L. Demain, A. L. Demain and L. Zhang, in Natural
Products, Humana Press, 2005, pp. 3–29.
Fig. 4 Hypothetical molecular model of virtual screening hit fonsecin B 25 A. M. Burja, B. Banaigs, E. Abou-Mansour, J. G. Burgess and
with the c-myc G-quadruplex.86 P. C. Wright, Tetrahedron, 2001, 57, 9347–9377.

1664 | Chem. Sci., 2011, 2, 1656–1665 This journal is ª The Royal Society of Chemistry 2011
View Article Online

26 J. W. Blunt, B. R. Copp, M. H. G. Munro, P. T. Northcote and 60 P. S. Charifson, J. J. Corkery, M. A. Murcko and W. P. Walters, J.
M. R. Prinsep, Nat. Prod. Rep., 2006, 23, 26–78. Med. Chem., 1999, 42, 5100–5109.
27 B. Haefner, Drug Discovery Today, 2003, 8, 536–544. 61 M. Feher, Drug Discovery Today, 2006, 11, 421–428.
28 A. L. Harvey, Drug Discovery Today, 2008, 13, 894–901. 62 C. A. Lipinski, F. Lombardo, B. W. Dominy and P. J. Feeney, Adv.
29 W. R. Strohl, Drug Discovery Today, 2000, 5, 39–41. Drug Delivery Rev., 1997, 23, 3–25.
30 M. S. Butler, J. Nat. Prod., 2004, 67, 2141–2153. 63 J. H. Toney, S. Navas-Martin, S. R. Weiss and A. Koeller, J. Med.
31 F. E. Koehn and G. T. C. Carter, Nat. Rev. Drug Discovery, 2005, 4, Chem., 2004, 47, 1079–1080.
206–220. 64 C. Sangma, D. Chuakheaw, N. Jongkon, K. Saenbandit, P. Nunrium,
32 D. J. Newman, J. Med. Chem., 2008, 51, 2589–2599. P. Uthayopas and S. Hannongbua, Comb. Chem. High T. Scr., 2005,
33 D. J. Newman and G. M. Cragg, J. Nat. Prod., 2007, 70, 461–477. 8, 417–429.
34 J. W.-H. Li and J. C. Vederas, Science, 2009, 325, 161–165. 65 G. Cozza, P. Bonvini, E. Zorzi, G. Poletto, M. A. Pagano, S. Sarno,
35 M. Feher and J. M. Schmidt, J. Chem. Inf. Comput. Sci., 2003, 43, A. Donella-Deana, G. Zagotto, A. Rosolen, L. A. Pinna, F. Meggio
218–227. and S. Moro, J. Med. Chem., 2006, 49, 2363–2366.
36 M. S. Boguski, K. D. Mandl and V. P. Sukhatme, Science, 2009, 324, 66 D.-H. Fu, W. Jiang, J.-T. Zheng, G.-Y. Zhao, Y. Li, H. Yi, Z.-R. Li,
1394–1395. J.-D. Jiang, K.-Q. Yang, Y. Wang and S.-Y. Si, Mol. Cancer Ther.,
37 D. W. Carley, IDrugs, 2005, 8, 306–309. 2008, 7, 2386–2393.
38 D. W. Carley, IDrugs, 2005, 8, 310–313. 67 C.-H. Leung, D. S.-H. Chan, H. Yang, R. Abagyan, S. M.-Y. Lee and
Published on 20 June 2011 on http://pubs.rsc.org | doi:10.1039/C1SC00152C

39 S. L. Schreiber, Science, 2000, 287, 1964–1969. D.-L. Ma, Chem. Commun., 2011, 47, 2511–2513.
40 M. D. Burke and S. L. Schreiber, Angew. Chem., Int. Ed., 2004, 43, 68 T. A. Soucy, P. G. Smith, M. A. Milhollen, A. J. Berger, J. M. Gavin,
46–58. S. Adhikari, J. E. Brownell, K. E. Burke, D. P. Cardin, S. Critchley,
41 D. S. Tan, Nat. Chem. Biol., 2005, 1, 74–84. C. A. Cullis, A. Doucette, J. J. Garnsey, J. L. Gaulin,
42 K. H. Altmann, Chem. Biol., 2007, 14, 443–451. R. E. Gershman, A. R. Lublinsky, A. McDonald, H. Mizutani,
43 A. N€ oren-M€ uller, I. Reis-Corr^ea, H. Prinz, C. Rosenbaum, U. Narayanan, E. J. Olhava, S. Peluso, M. Rezaei, M. D. Sintchak,
K. Saxena, H. J. Schwalbe, D. Vestweber, G. Cagna, S. Schunk, T. Talreja, M. P. Thomas, T. Traore, S. Vyskocil,
O. Schwarz, H. Schiewe and H. Waldmann, Proc. Natl. Acad. Sci. G. S. Weatherhead, J. Yu, J. Zhang, L. R. Dick, C. F. Claiborne,
U. S. A., 2006, 103, 10606–10611. M. Rolfe, J. B. Bolen and S. P. Langston, Nature, 2009, 458, 732–736.
44 I. Reis-Corr^ea, A. N€ oren-M€uller, H.-D. Ambrosi, S. Jakupovic, 69 M. C. Monti, A. Casapullo, C. N. Cavasotto, A. Napolitano and
K. Saxena, H. Schwalbe, M. Kaiser and H. Waldmann, Chem. – R. Riccio, ChemBioChem, 2007, 8, 1585–1591.
Downloaded on 04 December 2012

Asian J., 2007, 2, 1109–1126. 70 M. C. Monti, A. Casapullo, C. N. Cavasotto, A. Tosco, P. F. Dal,


45 P. Ertl, J. Chem. Inf. Comput. Sci., 2003, 43, 374–380. A. Ziemys, L. Margarucci and R. Riccio, Chem.–Eur. J., 2009, 15,
46 R. Abagyan and M. Totrov, Curr. Opin. Chem. Biol., 2001, 5, 375– 1155–1163.
382. 71 H. Liu, Y. Li, M. Song, X. Tan, F. Cheng, S. Zheng, J. Shen, X. Luo,
47 B. K. Shoichet, S. L. McGovern, B. Wei and J. J. Irwin, Curr. Opin. R. Ji, J. Yue, G. Hu, H. Jiang and K. Chen, Chem. Biol., 2003, 10,
Chem. Biol., 2002, 6, 439–446. 1103–1113.
48 C. McInnes, Curr. Opin. Chem. Biol., 2007, 11, 494–502. 72 L. Zhao and R. D. Brinton, J. Med. Chem., 2005, 48, 3463–3466.
49 D. B. Kitchen, H. Decornez, J. R. Furr and J. Bajorath, Nat. Rev.  Gustafsson, Curr. Opin. Chem. Biol., 1998, 2, 508–511.
73 J.-A.
Drug Discovery, 2004, 3, 935–949. 74 M. Ricote, A. C. Li, T. M. Willson, C. J. Kelly and C. K. Glass,
50 S. Ghosh, A. Nie, J. An and Z. Huang, Curr. Opin. Chem. Biol., 2006, Nature, 1998, 391, 79–82.
10, 194–202. 75 N. K. Salam, T. H. W. Huang, B. P. Kota, M. S. Kim, Y. Li and
51 C. N. Cavasotto and A. J. W. Orry, Curr. Top. Med. Chem., 2007, 7, D. E. Hibbs, Chem. Biol. Drug Des., 2008, 71, 57–70.
1006–1014. 76 N. Triballeau, E. Van Name, G. Laslier, D. Cai, G. Paillard,
52 F. C. Bernstein, T. F. Koetzle, G. J. B. Williams, J. E. F. Meyer, P. W. Sorensen, R. Hoffmann, H.-O. Bertrand, J. Ngai and
M. D. Brice, J. R. Rodgers, O. Kennard, T. Shimanouchi and F. C. Acher, Neuron, 2008, 60, 767–774.
M. Tasumi, J. Mol. Biol., 1977, 112, 535–542. 77 J. A. Wells and C. L. McClendon, Nature, 2007, 450, 1001–1009.
53 M. Loewer and E. Proschak, Molecular Informatics, 2011, 30, 398– 78 D. S.-H. Chan, H.-M. Lee, F. Yang, C.-M. Che, C. C. L. Wong,
404. R. Abagyan, C.-H. Leung and D.-L. Ma, Angew. Chem. Int. Ed.,
54 F. Petersen, R. Amstutz, J. M. Rollinger, H. Stuppner and T. Langer, 49, pp. 2860–2864.
in Natural Compounds as Drugs Volume I, Birkh€auser Basel, 2008, pp. 79 B. B. Aggarwal, Nat. Rev. Immunol., 2003, 3, 745–756.
211–249. 80 S. T. Crooke and W. T. Bradner, Cancer Treat. Rev., 1976, 3, 121–139.
55 D. Schuster and G. Wolber, Curr. Pharm. Des., 2010, 16, 1666–1681. 81 R. P. Perry and D. E. Kelley, J. Cell. Physiol., 1970, 76, 127–139.
56 C. N. Cavasotto and N. Singh, Curr. Comput.-Aided Drug Des., 2008, 82 H. J. Lipps and D. Rhodes, Trends Cell Biol., 2009, 19, 414–422.
4, 221–234. 83 A. Arola and R. Vilar, Curr. Top. Med. Chem., 2008, 8, 1405–1415.
57 F. Spyrakis, A. B. Chanal, X. Barril and F. J. Luque, Curr. Top. Med. 84 J.-L. Mergny and C. Helene, Nat. Med., 1998, 4, 1366–1367.
Chem. (Sharjah, United Arab Emirates), 2011, 11, 192–210. 85 M.-Y. Kim, H. Vankayalapati, K. Shin-ya, K. Wierzba and
58 R. D. Clark, A. Strizhev, J. M. Leonard, J. F. Blake and L. H. Hurley, J. Am. Chem. Soc., 2002, 124, 2098–2099.
J. B. Matthew, J. Mol. Graphics Modell., 2002, 20, 281–295. 86 H.-M. Lee, D. S.-H. Chan, F. Yang, H.-Y. Lam, S.-C. Yan,
59 J.-M. Yang, Y.-F. Chen, T.-W. Shen, B. S. Kristal and D. F. Hsu, J. C.-M. Che, D.-L. Ma and C.-H. Leung, Chem. Commun., 2010, 46,
Chem. Inf. Model., 2005, 45, 1134–1146. 4680–4682.

This journal is ª The Royal Society of Chemistry 2011 Chem. Sci., 2011, 2, 1656–1665 | 1665

You might also like