Vous êtes sur la page 1sur 15

REVIEWS

COMPUTERBASED DE NOVO
DESIGN OF DRUGLIKE MOLECULES
Gisbert Schneider and Uli Fechner
Abstract | Ever since the first automated de novo design techniques were conceived only
15 years ago, the computer-based design of hit and lead structure candidates has emerged
as a complementary approach to high-throughput screening. Although many challenges
remain, de novo design supports drug discovery projects by generating novel
pharmaceutically active agents with desired properties in a cost- and time-efficient manner.
In this review, we outline the various design concepts and highlight current developments in
computer-based de novo design.

DE NOVO DESIGN Molecular DE NOVO DESIGN produces novel molecular only those candidate molecules that represent a local
The design of bioactive structures with desired pharmacological properties neighbourhood of the search agent are considered at
compounds by incremental from scratch. In this approach, a medicinal chemist a time. The map guiding the search to an optimum is
construction of a ligand model and, equally, de novo molecule-design software is constructed en passant along the search path, and there-
within a model of the receptor
or enzyme active site, the
confronted with a virtually infinite search space. The fore dynamically evolves during the search process.
structure of which is known number of chemically feasible, drug-like molecules The virtual search agent mimics a medicinal
from X-ray or NMR data106. has been estimated to be in the order of 106010100, chemist, and scoring functions perform a function
from which the most promising candidates have to analogous to virtual assays. In the ideal case, such an
be selected (cherry picked)13. Such a large space pro- in silico laboratory provides a road map that guides
hibits exhaustive searching, despite great advances in the agents to high-quality molecular structures via
high-throughput screening (HTS) technology. Instead tractable synthesis routes. Positive design restricts
of the systematic construction and evaluation of each this virtual optimization process to small regions
individual compound, navigation in the de novo design of chemical space that have a higher probability of
process relies on the principle of local optimization, containing drug-like molecules. Negative design, by
which does not necessarily lead to the globally optimal contrast, defines tabu zones that are characterized by
solution: the design process converges on a local or adverse properties and unwanted structures4,5. Still,
practical optimum. In fact, most software implemen- there is no guarantee that a molecule will be retrieved
tations are non-deterministic, and rely on some kind from chemical space that finds immediate appraisal
of stochastic structure optimization. from a synthetic chemist. In this context, it is essential
Johann Wolfgang Just as chemists with different backgrounds are that we learn to accept that de novo design will rarely
Goethe-University, likely to propose different molecules as promising yield novel lead structures with nanomolar activity
Institute of Organic solutions, multiple runs with stochastic de novo in the first instance. Rather, the designed structures
Chemistry and Chemical
design software will produce different compounds as will probably represent examples of a prospective new
Biology, Beilstein Endowed
Chair for Cheminformatics, a result of the nature of the search algorithm. The lead series with micromolar activities that require
Marie-Curie-Str. 11 trick is to incorporate as much chemical knowledge further optimization.
D-60439 Frankfurt as possible about the structure of the search space The artwork Development II by M. C. Escher pro-
am Main, Germany into the design algorithm to facilitate directed navi- vides an illustration of the concept of chemical space
Correspondence to G.S.
e-mail: gisbert. gation towards a goal location. As it is impossible to (FIG. 1). There might exist several activity islands in
schneider@modlab.de enumerate all possible virtual molecules in advance chemical space, represented by the well-shaped and
doi:10.1038/nrd1799 because of the problem of combinatorial explosion, formed lizards; that is, drug molecules with desired

NATURE REVIEWS | DRUG DISCOVERY VOLUME 4 | AUGUST 2005 | 649


2005 Nature Publishing Group
REVIEWS

de novo design provides a method for lead identification.


De novo design can therefore be regarded as a com-
plement to other virtual techniques, such as database
searching, and non-virtual techniques such as HTS.
We also accentuate strengths and weaknesses of current
de novo design approaches. The basic properties of
selected de novo design programs discussed in this
article are given in TABLES 1 and 2.

Concepts
Basically, three questions have to be addressed by a de
novo design program: how to assemble the candidate
compounds; how to evaluate their potential quality;
and how to sample the search space effectively. De novo
design is faced with the problem of combinatorial explo-
sion: the number of different element types and the way
they can be linked together is huge. Furthermore, there
are not only a large number of theoretically possible
Figure 1 | How drug-like chemical space might be topologies but also a variety of conformations for a
structured. M. C. Eschers Development II The M. C. single topology. This renders a simple enumeration
Escher Company (Baarn, Holland, 2004). All rights reserved.
of all solutions an exhaustive search impossible.
All algorithmic decisions of a de novo design program
are obviously assessed by the quality of their outcome,
pharmacological behaviour. Each island represents a which, in turn, crucially depends on a meaningful
distinct structural class of molecules that are consid- reduction of the search space.
ered to be isofunctional with regard to their primary
target. It is impossible to directly hop from island Primary target constraints
to island. Instead, one has to take a route through What kind of input is necessary with regard to a
areas that are populated by less desirable compounds, particular biological target before a de novo design
depicted by the blurred lizard patterns in the Escher run can be started? This question is directly con-
artwork. With increasing distance from an island nected to the quality assessment of candidate com-
the essential activity-defining molecular patterns pounds, because the constraints extracted from the
become less pronounced. In other words, SCAFFOLD input are used in scoring the generated structures.
HOPPING which might be the desired outcome of a All information that is related to the ligandreceptor
de novo design experiment to obtain new lead series interaction forms the PRIMARY TARGET CONSTRAINTS for
with potentially improved properties or to circum- candidate compounds. Such constraints can be
vent intellectual property constraints depends on a gathered both from the three-dimensional receptor
conceptual abstraction from chemical structure. Any structure and from known ligands of the particular
successful design attempt that aims to generate novel target. If the former is consulted, the design strategy
structures that interact with a given target will have is receptor-based; in the latter case, it is ligand-based.
to be grounded on a representation of molecules that Receptor-based design starts with the determination
allows an escape from one activity island to another. of the binding site. As complementarities in molecu-
Before reaching a new activity island, however, mol- lar shape and submolecular physical and chemical
ecules will be synthesized and tested that have only properties are important for specific binding, the
marginal activity; for example, binding constants in binding site is then examined to derive shape con-
SCAFFOLD HOPPING the medium- or even high-micromolar range. Such straints for a ligand, as well as specific non-covalent
The identification of candidates would usually not be followed up or might ligandreceptor interactions in the form of hypo-
isofunctional structures not even be recognized at all in a drug discovery thetical INTERACTION SITES. Interaction sites are typi-
with different backbone
architectures.
project. One reason for this is that wandering through cally subdivided into hydrogen bonds, electrostatic
terra incognita in chemical space can only be guided and hydrophobic interactions. Receptor groups
PRIMARY TARGET by maps that were created from previously existing capable of hydrogen-bonding are of special interest
CONSTRAINTS knowledge, and extrapolation from this knowledge owing to the strongly directional nature of the two
All information that is related
requires that small steps be made one at a time. interaction partners hydrogen-bond acceptor and
to the ligandreceptor
interaction that is, the This review gives an overview of computer-based donor and often form key interaction sites. They
binding affinity of a ligand to molecular de novo design methods on a conceptual level. allow the assignation of ligand atom positions with a
the particular biological target. We focus on the design of small, drug-like molecules. complementary hydrogen-bond type within a small
There are also attempts in the design of peptides68 and region of space and a defined orientation. Key inter-
INTERACTION SITE
other polymeric structures911 that are not considered action sites have a major role in the effort to reduce
A position in space that is not
occupied by the receptor and in
here. By means of several successful deployments the vast number of possible structures because they
which a ligand atom favourably of de novo design in the hit- and lead-finding stages define strong and explicit requirements for successful
interacts with the receptor. of the drug discovery process we demonstrate that receptorligand binding.

650 | AUGUST 2005 | VOLUME 4 www.nature.com/reviews/drugdisc


2005 Nature Publishing Group
REVIEWS

Table 1 | Selected de novo design programs with their basic properties in chronological order (continued in Table 2)
Name (year) Building Primary Search strategy Structure sampling
blocks target
constraints
At Fr Rc Li DFS BFS Rnd MC EA Gr Lk Lat MD Sto Scoring function
HSITE/2D X X X Fitting and clipping of planar Steric constraints and hydrogen
Skeletons12,31,95 skeletons bonds
(1989)
3D Skeletons32 X X X X Steric constraints and hydrogen
(1990) bonds
Diamond Lattice33 X X X X Steric constraints and hydrogen
(1990) bonds
BUILDER v128 X X X X X Steric constraints and key
(1992) interaction sites
LEGEND20 (1991) X X X X Force field
LUDI13,14,9698 X X X X X Empirical scoring function
(1992) (SCORE1; revised version
SCORE2 in 1998)
NEWLEAD30 X X X X X Steric constraints
(1993)
SPLICE60 (1993) X X X X Pharmacophore and steric
constraints
GenStar34 (1993) X X X X Steric constraints and ligand
enzyme contact
GroupBuild18 X X X X Force field
(1993)
CONCEPTS39 X X X X Empirical scoring function
(1993)
SPROUT17,57-59 X X X X X X Solvent accesible surface,
(1993) hydrogen bonds, electrostatic
and hydrophobic interactions
MCSS & X X X X Simplified van der Waals potential
HOOK25,27 (1994) of non-polar interactions
GrowMol21 (1994) X X X X X Simple empirical scoring function
61 X X X X Potential energy
MCDNLG (1995)
At, atoms; BFS, breadth-first search; DFS, depth-first-search; EA, evolutionary algorithms; Fr, fragments; Gr, grow; Lat, lattice; Li, ligand; Lk, link; MC, Monte Carlo
sampling with Metropolis criterion; MD, molecular dynamics; QSAR, quantitative structureactivity relationship; Rc, receptor; Rnd, random; Sto, stochastic.

Derivation of interaction sites by including interaction sites of covalent bonds and


Receptor-based de novo design uses a variety of bonds to metal ions. Moreover, the idea of complex
methods to deduce interaction sites from the three- hydrogen bonds was introduced: in the case of mul-
dimensional structure of the binding pocket. HSITE12, ticentred or bifurcated hydrogen bonds, individual
a rule-based method, was the first software specifi- hydrogen-bonding regions were intersected to obtain
cally developed for the derivation of primary target complex hydrogen-bonding regions. These regions
constraints in an automated, de novo design approach. are assumed to form particularly strong hydrogen
Only hydrogen-bond acceptors and donors are con- bonds and are therefore preferred over normal
sidered, but it attempted to identify intramolecular hydrogen-bonding regions.
hydrogen bonds of the receptor. Acceptordonor Several de novo design programs implement grid-
pairs that participate in such intramolecular binding based approaches for the derivation of primary target
have to be excluded from the list of potential interac- constraints. A grid of points is generated in the binding
tion sites. The outcome of an HSITE run is a map of site, and interaction energies are computed by placing
hydrogen-bonding regions. These regions are centred different probe atoms or fragments at each grid posi-
at ideal hydrogen-bond geometry and allow a certain tion. An appropriate selection of probes for example,
tolerance range of bond length and bond angle around with hydrogen-bonding capabilities or lipophilic prop-
the centres. The values for the ideal geometry and the erties leads to the determination of interaction sites.
tolerance ranges are empirically derived from crystal- Some de novo design programs13,14,18 perform the cal-
structure data of small molecules. Other rule-based culations with the software GRID19, whereas others2023
methods were developed later that added lipophilic contain their own implementation of this algorithm.
interaction sites1316. HIPPO17 augmented this concept LigBuilder24 carries out a statistical analysis after the

NATURE REVIEWS | DRUG DISCOVERY VOLUME 4 | AUGUST 2005 | 651


2005 Nature Publishing Group
REVIEWS

grid-based calculations to extract the most promising compromise has to be made. Two de novo approaches25,26
interaction sites. The performance of grid-based use Multiple Copy Simultaneous Search (MCSS)27 for
methods crucially depends on the resolution of the grid. the generation of primary target constraints.
It is evident that higher resolution leads to more grid MCSS determines energetically favourable posi-
points and therefore to greater computational costs, so a tions and orientations of functional groups in the

Table 2 | Selected de novo design programs with their basic properties in chronological order
Name (year) Building Primary Search strategy Structure sampling
blocks target
constraints
At Fr Rc Li DFS BFS Rnd MC EA Gr Lk Lat MD Sto Scoring function
Chemical X X X X X Combined score of shape, grid-
Genesis22 (1995) based and scalar constraints
DLD26,99 (1995) X X X X Potential-energy function without
electrostatic interactions
PRO_ X X X X X X Empirical scoring function
LIGAND15,44,100-103
(1995)
SMoG41,42,104 X X X Knowledge-based scoring function
(1996)
BUILDER v229 X X X X Steric constraints
(1995)
CONCERTS35 X X X X Force field
(1996)
RASSE23 (1996) X X X X Force field augmented by chemical
rules
PRO_SELECT16,40 X X X X Empirical scoring function
(1997)
SkelGen63,64 X X X X X Geometric, connectivity and
(1997) chemical constraints
Nachbar45,105 X X X X Target-specific QSAR model
(1998) based on topological connectivity
descriptor
Globus49 (1999) X X X X Molecular similarity based on
all-atom-pairs-shortest-path
descriptor
DycoBlock36,37 X X X X Force field and solvent-accessible
(1999) surface
LEA47 (2000) X X X X Target-specific QSAR model based
on three-dimensional descriptors
LigBuilder24 (2000) X X X X X Empirical scoring function
48 X X X X Molecular similarity based on
TOPAS (2000)
topological pharmacophore and
substructure fingerprints
F-DycoBlock38 X X X X Force field and solvent-accessible
(2001) surface
ADAPT67 (2001) X X X X Weighted sum of DOCK score,
clogP, MM, number of rotatable
bonds and hydrogen bonds
Pellegrini & Field46 X X X X X Target-specific QSAR model
(2003)
SYNOPSIS55 X X X X Two examples: electric dipole
(2003) moment and empirically derived
HIV-RT scoring
CoG50 (2004) X X X X X Molecular similarity based on
fingerprint descriptor
BREED62 (2004) X X X *
*Exhaustive recombination. Exhaustive enumeration; no internal scoring function. At, atoms; BFS, breadth-first search; DFS, depth-first-search; EA, evolutionary
algorithms; Fr, fragments; Gr, grow; HIV-RT, human immunodeficiency virus reverse transcriptase; Lat, lattice; Li, ligand; Lk, link; MC, Monte Carlo sampling with
Metropolis criterion; MD, molecular dynamics; MM, molecular mass; QSAR, quantitative structureactivity relationship; Rc, receptor; Rnd, random; Sto, stochastic.

652 | AUGUST 2005 | VOLUME 4 www.nature.com/reviews/drugdisc


2005 Nature Publishing Group
REVIEWS

assembly that is, favourable positions of specific


Box 1 | Multidimensional optimization
functional groups in the binding site are not only indi-
Many practical optimization problems face several objectives at once and drug design cated but are already placed at these positions. This
is no exception to this. Typically, optimization techniques reformulate multi-objective placement of chemical groups provides a starting point
problems as a single objective using a weighted scoring function. Weighted scoring for the next step in de novo design: the assembly of a
functions sum the individual objectives and associate a weighting factor to each complete ligand.
objective: f(p) = w1p1 + w2p2 + + wnpn, where wn is the nth weighting factor and pn the
nth property. The setting of the weights is non-trivial. Conflicting objectives give rise Receptor-based scoring
to competition among them. For example, the design of a focused combinatorial The evaluation of candidate compounds with scoring
library might be subject to chemical diversity and drug-like physico-chemical functions is a central task in the design process. The
properties88. Such a situation constitutes a pitfall for an optimization algorithm and application of a de novo design program yields more
can lead to unreasonable solutions. Moreover, the weights specify a single
than one candidate compound. These structures can
compromise of the objectives. But there usually exists a whole family of solutions,
either emerge from a single run of the program or from
each of which corresponds to a compromise in terms of the individual objectives.
several runs with one candidate compound per run.
The Multiobjective Genetic Algorithm (MOGA)89 parlays the concept of a genetic
algorithm to explicitly incorporate the optimization of several constraints. It attempts
Scoring functions rank the generated structures and
to map out a hypersurface in the search space such that all solutions are considered as thereby suggest which structures are the most promis-
equivalent. The solutions that are part of this hypersurface also termed Pareto ing ones. Moreover, during a run they also guide the
frontier are non-dominated or Pareto solutions. A solution is non-dominated if an design process through the search space by assigning
improvement in one objective leads to a degradation in one or more of the other fitness values to the sampled space.
objectives. In other words, a solution dominates another one if it is equivalent or Conceived approximately 15 years ago, the very
better in all objectives and better in at least one objective compared with all other first de novo design programs only applied steric
solutions of the same population. The ranking of a population is inversely constraints to guide the search procedure2834, but
proportional to the number of times a solution is dominated. Solutions can be more sophisticated approaches emerged in the
inspected to identify the ones that represent the most appropriate compromise of the following years. Basically, these quality-assessment
individual objectives for the task at hand. approaches can be subdivided into three different
The figure shows potential non-dominated (red circles) and dominated (grey types of receptor-based scoring functions: explicit
circles) solutions in a problem with two properties. As indicated by the large arrow force-field methods; empirical scoring functions;
the objective is a minimization and knowledge-based scoring functions. All of these
of both properties. Solutions are approaches attempt to approximate the binding
non-dominated if the rectangle free energy. Force fields are computationally more
formed by lines drawn parallel costly than the other two types of scoring functions.
1
to the axes contains no other LEGEND20 was the first program that used a force
solution (dotted lines). The non- 2 field to evaluate candidate compounds, but over the
dominated solutions map out
Property 2

0
years quite a few others followed this concept18,23,3538.
the Pareto frontier (orange line). 5 The first empirical scoring function in the field of
Numbers next to the solutions 1 2
indicate the number of times 0
de novo design was implemented in the program
they are dominated. The MOGA 1 LUDI13,14. Empirical scoring functions are a weighted
algorithm has been successfully 0 sum of individual ligandreceptor interaction types
applied to a number of problems commonly supplemented by penalty terms, such as
in cheminformatics9092 and the number of rotatable ligand bonds. The weights
recently made its debut in the correspond to the average free-energy contribution
field of de novo design50. Property 1 of a single interaction of that type and are obtained by
a regression analysis of a set of receptorligand com-
plexes. Interaction types include, for example, hydro-
binding site. Multiple copies of functional groups are gen bonds, electrostatic interactions and hydrophobic
randomly placed inside the binding pocket. All frag- interactions. The regression analysis requires both
ments are then minimized simultaneously using a force known structures and binding constants, and so the
field such that the forces among individual functional available datasets are limited in size and often feature
groups are not considered. Groups are discarded if the similar ligands and receptors. This can result in a bias
interaction energy between them and the protein is of empirical scoring functions towards specific struc-
above a certain threshold. An MCSS run yields a set of tural motifs. However, they are fast and have proved
pre-docked fragments that can be further investigated their suitability, and are therefore implemented in
to choose the most promising ones. The same outcome several de novo design programs15,16,21,24,39,40.
can be achieved by the use of docking software, which Knowledge-based scoring functions have become
is indeed the initial step in some de novo design pro- popular in the field of docking during the past few
grams2830. Rule- and grid-based methods only derive years, yet to date there is only a single de novo design
the primary target constraints: the outcome of these program, SmoG41,42, that uses its own implementation of
two methods is a map of the receptor that pinpoints this type of scoring function. Knowledge-based scoring
favourable interaction sites of a ligand. MCSS and the is grounded on a statistical analysis of ligandreceptor
pre-docking of fragments effectively amalgamate the complex structures. The frequencies of each possible
derivation procedure and the first steps of structure pair of atoms in contact to each other are determined.

NATURE REVIEWS | DRUG DISCOVERY VOLUME 4 | AUGUST 2005 | 653


2005 Nature Publishing Group
REVIEWS

A Link/grow strategy a Place fragments

N
H O
Ile56
O Link
N
H O

OH
O
O OH
Ki = 16 M
O
O
Phe46 Asp37
b Place first fragment
Define binding pocket Determine interaction sites N
H O

O Grow

OH
O O
O O
B Lattice strategy
N N
H H

OH

O O
O O

Fill pocket with lattice points Find and connect interaction points Assign molecular framework Build molecule
Figure 2 | Principles of structure-based ligand assembly. A | The link (Aa) and grow (Ab) concepts are displayed for the
example of FK506-binding protein (FKBP12) ligand de novo design. On the basis of an X-ray model of the binding pocket
(PDB-identifier: 1fkf), interaction sites were identified. Selected interaction centres are indicated by blue dots (lipophilic), green
(acceptor) and red (donor) lines. A micromolar inhibitor of FKBP12 was designed using the software package LUDI, representing
one of the first successful prospective applications of de novo design93. The magenta-coloured substructures highlight the
linker in the link approach, and the grown part of the molecule in the grow scenario, respectively. B | The lattice strategy
provides an alternative approach that is grounded on a grid representation of potential positions of ligand atoms within the
binding pocket. Lattice points are indicated by grey dots. Ligand candidates are formed from those points that lie along the
shortest path through the lattice connecting interaction sites (the particular molecule shown is an artificial example).

PHARMACOPHORE
The ensemble of steric and
electronic features that is
Interactions found to occur more frequently than receptors (GPCRs), which are the most successful drug
necessary to ensure the optimal would be randomly expected are considered attractive; targets in terms of therapeutic benefit and potential
supramolecular interactions interactions that occur less frequently are considered sales43. Receptor-based structure generation is inevi-
with a specific biological target repulsive. Only structural information is necessary tably confronted with the problem of conformational
structure and to trigger (or to
block) its biological response106.
to derive these frequencies so that a greater number complexity. A ligand-based strategy, in contrast, can
of structures can be included in the analysis. As the either consider the three-dimensional or the topological
QUANTITATIVE STRUCTURE available structures are also more diverse than those structure of one or more known ligands.
ACTIVITY RELATIONSHIPS with known binding affinities, less bias is expected One way to use the information inherent to the
(QSAR). Mathematical
relationships linking chemical
compared with empirical scoring functions. known actives is the derivation of a three-dimensional
structure and pharmacological ligand PHARMACOPHORE model. Once established, it can
activity in a quantitative Ligand-based scoring be used to obtain a pseudo-receptor model44. This
manner for a series of If a three-dimensional structure of a particular biological facilitates the application of de novo design programs
compounds. Methods that can
be used in QSAR include
target is unavailable but one or more binding molecules that were originally developed with a receptor-based
various regression and pattern- are known, ligand-based design provides an alternative strategy in mind to ligand-based design. Alternatively,
recognition techniques106. strategy. This scenario holds true for G-protein-coupled the three-dimensional ligand pharmacophore model

654 | AUGUST 2005 | VOLUME 4 www.nature.com/reviews/drugdisc


2005 Nature Publishing Group
REVIEWS

ligands. A common binding mode of all deployed


ligands is a prerequisite for the building of a substantial
model. A set of known ligands can also be taken as an
Binding input for the development of a target-specific QUANTITA
Initial state
pocket TIVE STRUCTUREACTIVITY RELATIONSHIP (QSAR) model. The
established model then serves as a scoring function4547.
Another scoring approach is the computation of molec-
ular similarity between a known active compound and
the candidate compounds4850. This requires the choice
of a molecular descriptor and a similarity index. Both
a QSAR model and molecular similarity can be based
not only on the three-dimensional ligand structures
but also on their topological structures. This choice is
made by the selection of appropriate descriptors.
Level 1 NH HN

Secondary target constraints


An effectual drug molecule is subject to more objectives
than the binding affinity. Essential drug properties,
such as suitable absorption, distribution, metabo-
lism, excretion and toxicity (the so-called ADMET
properties), clearly emphasize the multi-dimensional
optimization of drug development. Constraints other
O
than the binding affinity are secondary constraints.
HO
The overall score of structures proposed by a de novo
Level 2 NH NH NH design program is sometimes calculated as a weighted
N
sum of the estimated binding affinity and other terms
N O that directly or indirectly contribute to the sec-
ondary constraints. Candidate compounds with an
increased chance of oral bioavailability can be obtained
by filtering the designed structures with Lipinskis rule-
of-five51. As the proposed molecules are considered
to be potential leads, a lowered upper bound for the
molecular mass of 350 Da and clogP of 3 might be
more appropriate52. In silico prediction systems are also
able to assist with the avoidance of severe side effects,
End state NH Designed molecule
such as hERG-mediated sudden death, though their
accuracy is still coarse-grained at best53.
O
The sole regard of valence rules does not ensure
the generation of chemically reasonable and stable
Figure 3 | Tree model of search space exploration by an automated structure- structures. The issue of unstable chemical groups was
generation method. A binding pocket and predicted key interaction sites are given as an
addressed early in the development of de novo design
input and are represented by the initial state (root node of the tree). In this example, key
interaction sites are pharmacophore points at which yellow circles indicate sites for a ligand with a set of chemical rules for example, a list of
hydrogen-bond donor and blue circles represent regions for lipophilic ligand parts. Candidate undesirable substructures. More difficult to handle,
compounds are assembled by cycles of fragment placing and subsequent scoring of the but also of major importance, is the ease of synthesis,
resultant partial structures (grow strategy). Partial structures are partial solutions (shown in light because the ability to predict binding affinity is still
green) if they satisfy the primary target constraints. In the example, some partial structures are limited. It cannot be expected from the outcome of a
rejected due to boundary violation (Level 1 and 2) or due to mismatching interaction types
design study that all the candidate compounds have
(Level 2). Two partial solutions emerge on Level 1. A breadth-first search would follow both
nodes in the search graph simultaneously, whereas the tree shown here selects a single node
the proposed properties. What can be expected is a
for expansion (depth-first search). Selection of this single node could be guided by the score of significantly increased hit rate compared with the
the partial solution, by chance or by a combination of both. A path through the search space screening of arbitrary compound collections.
tree from the initial state to an end state leads to a candidate compound. This only underlines the relevance of validation with
biological assays and the consequent need to design
synthetically accessible structures. Synthetic accessibil-
can be used directly in a similarity design method44. ity subsumes the availability of starting materials and
Whereas a (pseudo)receptor guides the design of the synthetic feasibility of the final product by amena-
structures that are complementary to the primary ble reactions. Even though this issue was recognized
target constraints, a ligand pharmacophore model can early in the history of de novo design, it was addressed
be applied to designing structures that are similar to only recently. The common basic motif of the few de
these constraints. The generalization quality of a three- novo design programs that consider ease of synthesis
dimensional pharmacophore model improves with a is to assemble the building blocks in accordance with
high degree of structural diversity in the set of known a set of virtual organic reaction schemes.

NATURE REVIEWS | DRUG DISCOVERY VOLUME 4 | AUGUST 2005 | 655


2005 Nature Publishing Group
REVIEWS

For example, the building blocks of TOPAS48 are positioned by the de novo program itself or provided
obtained by virtual retro-synthesis of a drug-molecule by another program (pre-docked building blocks). The
collection with a set of 11 common organic reactions positioned building blocks are automatically connected
the so-called RECAP54 reactions. The same set of to each other by so-called linkers to yield a complete
reaction schemes is then used to assemble candidate molecule that satisfies all key interaction sites. Linkers
compounds. PRO_SELECT 16 adds the template- are selected with the objective of forming favourable
substituent idea of combinatorial chemistry. The most interactions with the receptor.
sophisticated approach is undertaken by SYNOPSIS55. The growing procedure1315,18,20,21,23,32,34,41,42,5759 starts
In this program, the building blocks are constituted by with a single building block at one of the key interac-
a database of available molecules. Structure assembly tion sites of the receptor (FIG. 2). This starting point is
is guided by 70 different simulated organic synthesis selected by the program, the user or is given by a pre-
steps so that a synthesis route is proposed for every docked fragment. The structure is then grown from the
generated structure. Furthermore, the acceptance of a initial building block in an attempt to provide suitable
reaction incorporates information about neighbours interactions for both the key interaction sites of the
of the functional group, other functional groups that receptor and regions of the receptor between two key
might prevent the reaction, and reactivity rankings interaction sites. PRO_SELECT16 introduced the idea
if a functional group is present more than once in a of combinatorial chemistry to the linking method.
molecule. An alternative approach is the application Substituents are linked to a pre-docked scaffold with
of external software that attempts to assess the syn- user-defined attachment sites. Both the growing and the
thetic accessibility of a set of candidate compounds linking strategy have their strengths and weaknesses.
for example, CAESA17 or SEEDS56. These programs The growing strategy can run into difficulties if the
automatically analyse generalized synthetic routes and active site contains two or more distinct (sub)pockets
select potential precursors from databases of available separated by a large gap in which the possible interac-
compounds. CAESA additionally provides an estima- tions between small-molecule ligands and the protein
tion of the ease of synthesis for example, recogni- are limited18. When fragments are used in combination
tion of complex ring systems or stereocentres by an with the linking approach, slightly misplaced fragments
expert rule system. or fragments with no strictly defined spatial orienta-
tion can lead to ambiguity during linking. An example
Structure sampling is a phenyl ring, which has no preferred orientation in
The basic building blocks for the assembly of candi- a lipophilic binding pocket.
date structures can be either single atoms or fragments. Another concept is the placement of an atomic
Atom-based approaches are superior to fragment-based lattice in the binding site (FIG. 2). This lattice is made
methods in terms of the structural variety that can up of regularly arranged sp3 carbon atoms (diamond
be generated. But this increase in potential solutions lattice)33, randomly and evenly distributed atoms29, or
makes it harder to find suitable candidate compounds pre-docked fragments28. Lattice atoms in the vicinity
among the ones that are amenable. Fragment-based of different interaction sites are then joined by find-
design strategies, on the other hand, significantly ing the shortest path through the lattice atoms. Atoms
reduce the size of the search space. This reduction can that are part of such a shortest path are connected by
be called a meaningful reduction if fragments are used newly formed bonds. DLD26 and MCDNLG61 start with
that commonly occur in drug molecules. Additionally, a binding site that is filled with a non-physical arrange-
the definition of fragment is variable: a fragment can ment of atoms. The initial atom arrangement is termed
be anything from an atom to a polycyclic ring system, non-physical because atoms are placed or connected
which means that atoms are essentially a subset of in a way that is non-existent in reality. Examples are
fragments. A composition of building blocks that participation of atoms in a number of bonds that far
contains small fragments and larger ones allows for exceed typical chemical bond valences or which com-
structural changes to a different extent depending pletely disregard van der Waals radii. Then, an atom
on the specific requirements. Many of the early de is randomly selected and a randomly chosen transi-
novo design strategies were atom-based. But as the tion is applied to this atom. Such a transition can be a
combinatorial problem that is coupled with atoms translation or rotation operation26,61, a change of atom
as building blocks became more and more evident, type or bond type, or the appearance or disappearance
fragments were generally used. Today, we commonly of an atom26. The guidance of iterated transitions by a
find building-block sets that are mainly composed of potential-energy function finally yields a chemically
fragments with more than one atom and padded with valid molecule.
a few single-atom fragments. A few de novo design methods implement structure
There are several general concepts of structure sam- sampling that is driven by a molecular dynamics simu-
pling: linking, growing, lattice-based sampling, random lation. Covalent connections are formed among the
structure mutation, transitions driven by molecular building blocks in a stochastic and reversible manner
dynamics simulations, and graph-based sampling. to dynamically evolve candidate compounds. Initially,
The linking approach1315,24,25,30,5760 starts with the building blocks are randomly positioned in the binding
placement of building blocks at key interaction sites site. Subsequent free movement of the building blocks
of the receptor (FIG. 2). These building blocks are either is guided by molecular dynamics equilibria. Every

656 | AUGUST 2005 | VOLUME 4 www.nature.com/reviews/drugdisc


2005 Nature Publishing Group
REVIEWS

few molecular dynamics steps, one or more building Later approaches3639 exclusively regarded interactions
blocks are randomly chosen, all bonds are cleaved and between the building blocks and the receptor (consen-
empty valences are filled with bonds to nearby building sus molecular dynamics). A further addition to this
blocks. The first such program, CONCERTS35, consid- method of structure sampling was a repeated cycle of
ered interactions between the building blocks and the construction and deconstruction: after a certain period
receptor, as well as between different building blocks. of molecular dynamics simulation, all candidate com-
pounds are dissected into their respective fragments.
Initial state Before each deconstruction, high-scoring structures
HO
O are stored in a list for later inspection38.
Ligand-based de novo design is not provided with
N NH O
interaction sites as primary target constraints. The
OH majority of ligand-based methods operate on the topo-
HN O logical molecular graphs and feature an evolutionary
algorithm for optimization. This choice implicitly paves
O the way for their structure sampling technique: genetic
O operators that are specifically tailored for molecular
O HN O graphs. Whereas Globus49 only implemented a recom-
+
N N bination operator, TOPAS48 solely applies a mutation
O N operator that randomly substitutes whole fragments,
OH
thereby obeying the rules of virtual chemical reaction
O 0.47
schemes. Nachbar45 and Brown50 developed both muta-
HO
O
tion and recombination operators. Candidate com-
pounds are mutated by changing an atom element type
O
or bond order, opening or closing rings and expand-
ing or contracting rings. Chemical Genesis22 applies
O genetic operators to the three-dimensional molecular
Tanimoto index (similarity to the template structure)

0.57 structures that led to the development of translational


and rotational mutation operators. BREED62, a recent
N
H
N NH
contribution to the field of de novo design, utilizes a
N
N N
completely different idea: a set of known ligands for
N N a particular target in their three-dimensional active
N O conformation is exhaustively recombined. The recom-
0.66 bination is carried out by overlaying the known ligands
O and swapping the fragments of different ligands on
each side of overlapping bonds. This procedure is car-
N NH N ried out recursively, so that the candidate compounds
that emerge from recombination are added to the pool
Br NH
of known actives and participate in subsequent cycles
of recombination. A prerequisite for BREED is knowl-
0.70
O edge of the receptor-relevant, presumed active ligand
H
N N conformation. As the known ligands are superimposed
N N NH N in their active conformation, the generated structures
N inherit this active conformation and are effectively
N Br NH
pre-docked to the receptor.
O
OH 0.81
Figure 4 | Progress of a de novo design exercise
HN following the concept of the design software TOPAS48
N N for assembling drug-like structures. The task was to
F N HN (re)generate the structure of imatinib (Gleevec; Novartis), an
O inhibitor of Abelson tyrosine kinase94. The algorithm started
off with a population of 100 randomly assembled molecules
O N 0.92 and navigated through the search space by stochastic
sampling. For molecule assembly, building blocks were used
N N N N that originated from pseudo-retrosynthetic fragmentation of a
H H
N database of drug-like molecules. Only the best partial
N F
solutions of selected generations are shown. The Tanimoto
index, which is based on Daylight Fingerprints, served as the
O N fitness function. Larger values indicate a higher degree of
1.0
similarity between the intermediate solutions and the
N N N N template structure, imatinib. After 50 generations, the
H H
N algorithm successfully reproduced the template. Individual
N End state
runs differed in terms of the highest-scoring candidate.

NATURE REVIEWS | DRUG DISCOVERY VOLUME 4 | AUGUST 2005 | 657


2005 Nature Publishing Group
REVIEWS

O
OH
HO O H O N
O N N N
S H
N
O O H
O H2N S
N
OH

1 2 3

N O
HN F3C
HN
O CF3

O
4 5

Figure 5 | Examples of pharmacologically active substances that were designed de novo using computer algorithms.

Combinatorial search strategies paths reaches an end state. During a breadth-first


De novo design has to tackle the issue of combinatorial search all nodes are systematically examined so that
explosion, which leads to problems that are NPHARD. identifying the optimal solution is guaranteed.
This class of problems is believed not to be solvable How is such an exhaustive search in a de novo design
with provably good run times and provably optimal run possible? Most of the programs that implement the
solution quality. Combinatorial search algorithms breadth-first strategy deal with a smaller problem space:
offer a practical solution by giving up one or both of they use the linking method for structure assembly.
these two aims. In other words, a combinatorial search Given that the key interaction sites are already satis-
strategy usually finds pretty good solutions, but there is fied with favourable ligand fragments, and a small set
no proof that the solutions will not become arbitrarily of fragments is used to link them, an exhaustive search
bad; it usually runs reasonably quickly, but there is no is feasible. RASSE23, an atom-based growing approach
argument this will always be the case. A combinatorial with a breadth-first search, implements another strategy.
search algorithm is able to solve instances of combina- It limits the search space by choosing only the 100 best
torial problems by reducing the size of the search space partial solutions at each growing step. An ancestor of
and by exploring it efficiently. HEURISTIC algorithms SPROUT32 and SPROUT itself5759 use the A* search
represent one way to achieve this. algorithm, a particular type of best-first search. The A*
The search space for structure generation can be algorithm optimizes a depth-first search by estimat-
represented by a graph in which each possible state of ing the costs of reaching a specific partial solution and
the system is assigned a node (FIG. 3). The root node reaching an end state from this partial solution. The
represents the initial state (that is, the start of the cost estimation is based on a heuristic that incorporates
problem); terminal nodes of the graph correspond knowledge about the problem domain in otherwise
to end states (that is, acceptable solutions of the general search algorithms. A refined version of the
problem); and all other nodes represent intermediate combinatorial search algorithm of SPROUT led to an
states. Application of a rule to a given node leads to interesting amalgamation of a breadth-first and a depth-
the generation of a successor node at the next level of first search17. Another application of depth-first search is
the graph. The objective of any search algorithm is to the random selection of one partial solution at each tree
find a path between the root node and the end nodes level among the highest-scoring partial solutions18,34 or
NPHARD by selecting the nodes that are expanded to the next among all partial solutions40.
Non-deterministic level of the graph.
polynomial-time hard Monte Carlo and the Metropolis criterion
(NP-hard) refers to a class of
decision problems of which
Breadth-first and depth-first search Pure random sampling, also called Monte Carlo
current knowledge provides Several de novo design programs implement either a search, is implemented in the program LEGEND20.
no way to obtain or derive a breadth-first strategy23,2830,5759 or a depth-first strat- Random sampling can also be combined with a
solution time that is less than egy15,18,28,3234,5759. The depth-first strategy retains only Metropolis criterion. In this case, after each struc-
exponential in problem size.
one of a variety of possible partial solutions at each ture-modification step, the change is evaluated
HEURISTIC level of the search space graph until an end state is to decide whether it is accepted or rejected. If the
Application of probabilistic reached. Even if the highest-scoring partial solution modification results in a fitter candidate compound,
rules grounded on knowledge is selected each time, it is not guaranteed to find the it is immediately accepted. If, on the other hand, the
of a particular problem domain overall best solution, but the search space is reduced modification yields a less fit candidate compound, it
to obtain an algorithm that
performs reasonably well in
significantly. A breadth-first strategy retains all partial can still be accepted with a probability that is based
many cases, but without proof solutions at one level of the search space graph and on the scoring function difference between the
that it is always fast. explores, sequentially, other levels until each of these modified and unmodified structure and a random

658 | AUGUST 2005 | VOLUME 4 www.nature.com/reviews/drugdisc


2005 Nature Publishing Group
REVIEWS

In structure generation, the phenotype itself is a


molecular graph that renders a distinction between
an evolutionary strategy and a genetic program-
ming algorithm difficult. Moreover, extensions to
W60D NH genetic algorithms allow the use of extensible and
Y60A
H57 contractible chromosomes that are similar to those in
O
genetic programming. We therefore do not attempt
L99
S195
to classify the following examples any further than as
evolutionary algorithms.
NH NH2 The program LEA47 uses a tailored form of chro-
6 mosomes. It encodes the molecular structures in the
N98 form of SMILES strings66 and the genetic operators are
carried out on these strings. In TOPAS48, the search for
A190 candidate compounds is guided by an evolutionary algo-
G216
W215 G219 rithm that features adaptive step-size control. The vari-
ance distribution of offspring is determined by a special
D189 parameter. This parameter is also subject to evolution
so that the width of the search-space sampling that is,
Figure 6 | Experimentally determined binding mode of benzamidine within the S1
the chemical diversity of the population adapts to the
substrate-recognition pocket of thrombin (by X-ray, resolution 3.16 , PDB identifier:
1DWB). Residues flanking the binding pocket are shown, including the catalytic Ser195. The
local requirements of the fitness landscape.
arrow indicates the direction of fragment growing by de novo design. The binding pocket offers The crossover operator can give rise to problems if
several potential interaction points with the ligand. applied to cyclic graphs. As the name suggests, a cyclic
graph contains one or more cycles, which translate
to chemical ring systems in the context of molecular
number. CONCEPTS39 was the first de novo design structures. Cycles are a common property of molecular
program that made use of a Monte Carlo search with structures and abundant in drug molecules. A crossover
the Metropolis criterion; several others followed operator takes two molecular graphs as an input, dis-
suit21,26,35,41,42,46,61,63,64. connects each graph into two subgraphs, and merges
two subgraphs from different input graphs. This pro-
Evolutionary algorithms cedure results in two molecular graphs, each of which
Evolutionary algorithms are based on the ideas comprises parts of the first and the second input graph.
described by Charles Darwin in 185965. They are In graphs with cycles, the removal of a single edge does
population-based optimization algorithms that mimic not necessarily lead to two disconnected fragments.
biological evolution with the genetic operators repro- Cyclic graphs therefore significantly complicate matters.
duction, mutation and recombination (crossover). Several authors have faced this problem and developed
Mutation introduces new information into a popula- different solutions by modifying the crossover operator
tion, whereas recombination exploits the information appropriately22,45,49,50,67. There are other programs that
inherent in the population. Candidate compounds are mainly utilize the population-based variation and selec-
represented by individuals in a population so that a set tion cycle of evolutionary algorithms without directly
of solutions is obtained in a single run of the algorithm. relating to the genetic operators24,55.
The scoring function, which in the context of evolu-
tionary algorithms is often called a fitness function, Recent examples of de novo design
assesses the fitness of the individuals and therefore There exist several published examples of successful
determines which structures are chosen as parents. A de novo design projects, and comprehensive lists of
parent structure is then modified by one of the genetic applications have been compiled elsewhere6870. Here,
operators and the resultant child structure is put in the we present selected examples of more recent de novo
population of the next generation. This cyclic process design efforts. Our intention is to emphasize attempts
of variation and selection is iterated until a termination aiming at synthetic accessibility and the incorpora-
criterion is reached (FIG. 4). tion of SECONDARY TARGET CONSTRAINTS to increase the
Evolutionary algorithms can be further subdi- drug-likeness of the designed structures (FIG. 5).
vided into genetic algorithms, genetic programming In a semi-automatic design exercise, MCSS 27
and evolution strategies. Genetic algorithms encode was used to explore key regions in the active sites
the phenotype (molecular structure) by means of a of Candida albicans and Mycobacterium tuberculosis
chromosome, which usually comprises either binary lanosterol 14-demethylase (CYP51), a member of the
SECONDARY TARGET
CONSTRAINTS
or real values. In genetic programming, the chro- cytochrome P450 superfamily. Through this approach,
Essential drug properties apart mosomes are represented as trees rather than the energetically favourable positions and orientations of
from the binding affinity to a fixed-length strings of genetic algorithms. This tree functional groups were obtained71. LUDI13,14 fragment
biological target for example, representation facilitates the extension and contrac- placing helped further explore important regions of
absorption, distribution,
metabolism, excretion and
tion of chromosomes. Evolutionary strategies omit the binding sites. Initial lead molecules were manu-
toxicity properties, or binding the encoding of phenotypic features by a chromo- ally constructed from a selection of the MCSS minima
selectivity. some and operate directly on the phenotype instead. that took into account synthetic feasibility. Subsequent

NATURE REVIEWS | DRUG DISCOVERY VOLUME 4 | AUGUST 2005 | 659


2005 Nature Publishing Group
REVIEWS

Consideration of automatically docked molecules


would further extend the numbers of ideas generated
by this algorithm62,72.
O The structure-based SYNOPSIS55 approach was
Y60A W60D used to find new inhibitors of the HIV-1 protein reverse
N
N
transcriptase (RT). Several runs of the design algo-
O rithm produced a shortlist of 28 candidates predicted
F
to have low IC50 values. The designs were assessed
3.1
by an organic chemist for both synthetic feasibility
and sufficient dissimilarity from known HIV-1 RT
N98 HN NH2 inhibitors. Of 18 molecules synthesized and tested, 10
compounds were identified with inhibitory in vitro
G219 7
activity below 100 M, four were cytotoxic molecules
W215 and four were inactive. Compound 3 (FIG. 5) gives an
G216
example of a designed HIV-1 RT inhibitor (IC50 =
A190
4.4 M). The authors stress that the outcome of the
D189
design project and hit rate compares favourably with
HTS experiments despite the fact that a rather high
Figure 7 | Experimentally determined binding mode of compound 7 within an X-ray
activity threshold was used52,55,73.
model of the thrombin active site (resolution 1.67 , PDB identifier: 1OYT). Amino-
acid residues interacting with the ligand are highlighted. An FHC hydrogen bond was
LEGEND20 was applied to the design of new cyclin-
shown to significantly enhance the proteinligand interaction. The hydrogen bond pattern dependent kinase 4 (CDK4) inhibitors in a structure-
between the benzamidine moiety of the inhibitor and protein side chains in the S1 based manner from a homology model of CDK456. The
recognition pocket is indicated by dotted lines. designs were not synthesized; instead, combinatorial
scaffolds with molecular masses below 350 Da were
extracted from the candidate molecules and served
energy minimization and LUDI scoring led to the as templates for library design. In a first step, a novel
discovery of a novel series of antifungal agents (such class of CDK4 inhibitors was found with nanomolar
as compound 1 in FIG. 5) with micromolar activity. inhibitory activity (compound 4; the scaffold is shown
Compound 1 specifically interacts with active-site in red in FIG. 5). Subsequently, a combination of LUDI
residues and therefore potentially would not cause and LeapFrog (Tripos Inc.) was used to design a
toxicity arising from coordination binding with pyrazol-3-ylurea library of selective low-nanomolar
the haeme of mammalian P450s. The secondary CDK4 inhibitors74. It is noteworthy that the design
constraint of avoiding P450 toxicity was indirectly algorithms were only used for the identification of
considered by restricting the ligandreceptor inter- side chains of the combinatorial scaffold that show
action to only those functional groups that form favourable interactions with enzyme residues around
non-covalent bonds between the ligand and residues the ATP-binding pocket. These examples demonstrate
of the binding site. This example shows that manual the potential of combining established chemistry that
intervention at the level of fragment linking can ensures synthetic accessibility with de novo design for
facilitate the identification of drug-like agents. side-chain optimization.
Embarking on a strict fragment-based approach, A related, entirely ligand-based, concept was
the design software BREED62 was used to produce pursued using TOPAS48 for the design of combina-
novel human immunodeficiency virus 1 (HIV-1) torial libraries directed against human cannabinoid
protease and kinase inhibitors following a two-step receptor 1 (CB1)75. A stock of building blocks was
protocol. In the first step, four known ligands of generated from pseudo-retrosynthetic fragmentation
each class were processed by the BREED recombina- of known GPCR ligands. These building blocks were
tion algorithm, and in the second step the resulting used for subsequent de novo design. A single known
hybrids were fused with additional known reference CB1 ligand served as the reference for pharmacoph-
molecules, resulting in more than 100 potential new ore-based fitness calculations. From several TOPAS
ligands. Compound 2 (FIG. 5) represents an example runs, two prominent scaffolds were identified, and
of such a designed, potent HIV-1 protease inhibitor small, focused libraries were devised by similarity
(Ki = 42 nM). Both synthetic feasibility and potential searching for promising and physically available side
drug-like properties of the designs were gained by chains. Hit rates of 6% and 10% were obtained, respec-
starting off from drug-like reference molecules. In a tively, applying an activity threshold of Ki = 10 M.
strict sense, the resulting molecules do not represent The most potent ligand was compound 5 (Ki = 0.3
real de novo designed structures; rather, they could M; scaffold shown in red in FIG. 5). This study dem-
be regarded as chimeric structures that were generated onstrates that novel lead-structure candidates can be
from exhaustive recombination of existing ligands. found using ligand-based de novo design even in the
The BREED concept demonstrates the usefulness absence of receptor-structure information by taking
of re-using already known, potentially preferred, a single known reference ligand as a template. Again,
fragments. It is noteworthy that BREED recombined the combination of de novo design and combinato-
experimentally determined ligand conformations. rial optimization was pivotal to success. The hit rate

660 | AUGUST 2005 | VOLUME 4 www.nature.com/reviews/drugdisc


2005 Nature Publishing Group
REVIEWS

was up to two orders of magnitude higher than that a direct consequence of our lack of knowledge about
expected for random screening of historical corporate function-determining features of desired ligand mol-
compound collections. Lengthy assay development ecules in the early stages of the discovery process79.
and costly, logistically complex, HTS efforts were not We expect de novo design to increasingly become a
required in order to generate two novel, selective and complementary strategy to HTS through a number of
patentable hit series whose chemical simplicity can routes: by suggesting new chemical entities that were
serve as a starting point for further refinement and designed taking into consideration several aspects of
lead-optimization activities. lead- and drug-likeness and synthetic accessibility;
How do such automated, entirely computer-gener- by making full use of pre-existing knowledge, such
ated designs compare with rational, non-automated as reference ligands or receptor models; and by facili-
design approaches for example, by manual molec- tating the design of activity-enriched screening sets
ular modelling, docking and piecemeal structure with increased hit rates.
modification? The design of non-peptidic thrombin Automated de novo design has proven its value
inhibitors has been the focus of many de novo for hit and lead-structure identification. Designed
design projects during recent years and can help molecules provide sometimes astonishing ideas for
answer this question. the medicinal chemist and aid in the development
Thrombin is a trypsin-like serine protease with a of novel and patentable leads with desired property
central role in the blood-clotting cascade. A pivotal profiles. To gain further acceptance of computer-
part of current thrombin inhibitors is a functional generated molecules it will be essential to understand
group interacting with Asp189 at the bottom of the current limitations of the design techniques. One
S1 recognition pocket of the enzyme. As thrombin weakness is the inability of de novo design software
cleaves fibrinogen after an arginine residue, guanidin- to sufficiently consider the flexibility of the target
ium-mimicking fragments are usually selected by de protein. Although examples of docking methods exist
novo design software at this position. The best result that consider receptor flexibility, examples in the field
of an automated combinatorial docking and design of de novo design are sparse38,80. This is probably a
approach using LUDI is compound 6 (Ki = 10 nM), result of the combinatorial problem de novo design
which contains a benzamidine in this position (FIG. 6)76. is faced with; receptor flexibility adds just another
In this study, p-amino-benzamidine (Ki = 34 M) was aspect of combinatorics on top of this problem. The
identified as a preferred core fragment for subsequent same holds for flexible alignments of ligand ensembles
combinatorial optimization by LUDI (unsubstituted that provide the basis for a QSAR, pharmacophore
benzamidine alone yields a Ki of 250 M). or pseudo-receptor model81. One cannot expect good
For comparison, a carefully, rationally de novo designs from poor initial alignments, because these
designed molecule is compound 7 (FIG. 7), which was guide the search through chemical space.
optimized in a step-wise manner to display selective Most pharmaceutical leads are part of a limited set
thrombin inhibition77. Also revealing the benzami- of chemotypes8284. Many pharmaceutical companies
dine residue in the S1 pocket, this compound was direct their attention towards this limited number of
developed from a rigid tricyclic core structure that structural classes. The chemical diversity of potential
pre-organizes this chemotype for thrombin bind- leads discovered by HTS is therefore restricted by
ing78. As a result of a fluorine scan of the precursor the diversity of the screening libraries that are used.
molecule, an FHC hydrogen bond was shown to De novo design offers a broader exploration of
significantly enhance the proteinligand interaction chemical space and therefore makes it possible to
(Ki = 6 nM; 67-fold selectivity compared with trypsin identify novel ligand scaffolds, which can be a major
inhibition) (FIG. 7). The increased positive polarization competitive advantage85. Fragment-based screening
of the hydrogen in the ortho position to the fluor sub- strategies, such as NMR and high-throughput X-ray
stituent is also supposed to enhance the edge-to-face crystallography, can be used to help identify new
interaction with Trp215. Such effects and other types chemotypes by suggesting starting orientations of
of ligandreceptor interactions, such as arenearene molecular building blocks for subsequent computer-
and cation interactions, or water-mediated con- assisted linking and growing86,87. Still, the prediction
tacts, are often neglected by de novo design software. of crucial properties of a drug molecule primary
These non-covalent interactions can influence the target constraints (binding behaviour) as well as
binding mode and affinity of a ligand, and therefore secondary constraints such as pharmacokinetic
demonstrate limitations of current automated de novo properties is limited. Molecules that are predicted
design methods. Starting the amalgamation of frag- to be the best from a de novo design run rarely rep-
ments from small seed structures, such as the ben- resent the preferred choice of a medicinal chemist.
zamidine fragment in the thrombin example, might Even though there is fully automated de novo design
even prevent better solutions from being found. software, it remains a crucial human task to pick the
most promising candidates. An important goal of de
Conclusions novo design is to inspire medicinal chemists through
The drug discovery pipelines of many pharmaceu- the chemical motifs that are identified. Ultimately, the
tical companies are fuelled by HTS as one of the aim is to offer support for hit and lead identification
major sources of new hit-to-lead candidates. This is and widen the chemical horizon.

NATURE REVIEWS | DRUG DISCOVERY VOLUME 4 | AUGUST 2005 | 661


2005 Nature Publishing Group
REVIEWS

1. Dobson, C. M. Chemical space and biology. Nature 432, 27. Miranker, A. & Karplus, M. Functionality maps of binding 51. Lipinski, C. et al. Experimental and computational
824828 (2004). sites: a multiple copy simultaneous search method. approaches to estimate solubility and permeability in drug
2. Lipinski, C. & Hopkins, A. Navigating chemical space for Proteins 11, 2934 (1991). discovery and development settings. Adv. Drug. Deliv. Rev.
biology and medicine. Nature 432, 855861 (2004). 28. Lewis, R. A. et al. Automated site-directed drug design 23, 325 (1997).
3. Schneider, G. Trends in virtual combinatorial library design. using molecular lattices. J. Mol. Graphics 10, 6678 52. Teague, S. J. et al. The design of leadlike combinatorial
Curr. Med. Chem. 9, 20952101 (2002). (1992). libraries. Angew. Chem. Int. Ed. Engl. 38, 37433747
4. Richardson, J. S. & Richardson, D. C. The de novo design 29. Roe, D. C. & Kuntz, I. D. BUILDER v.2: improving the (1999).
of protein structures. Trends Biochem. Sci. 14, 304309 chemistry of a de novo design strategy. J. Comput. Aided 53. Aronov, A. M. Predictive in silico modeling for hERG
(1989). Mol. Des. 9, 269282 (1995). channel blockers. Drug Discov. Today 10, 149155 (2005).
5. Richardson, J. S. et al. Looking at proteins: 30. Tschinke, V. & Cohen, N. C. The NEWLEAD program: 54. Lewell, X. O., Budd, D. B., Watson, S. P. & Hann, M. M.
representations, folding, packing, and design. Biophys. J. a new method for the design of candidate structures from RECAP Retrosynthetic Combinatorial Analysis
63, 11851209 (1992). pharmacophoric hypothesis. J. Med. Chem. 36, Procedure: a powerful new technique for identifying
6. Moon, J. B. & Howe, W. J. Computer design of bioactive 38633870 (1993). privileged molecular fragments with useful applications in
molecules: a method for receptor-based de novo ligand 31. Lewis, R. A. & Dean, P. M. Automated site-directed drug combinatorial chemistry. J. Chem. Inf. Comput. Sci. 38,
design. Proteins 11, 314328 (1991). design: the formation of molecular templates in primary 511522 (1998).
7. Schneider, G. & Wrede, P. The rational design of amino structure generation. Proc. R. Soc. Lond. B 236, 141162 55. Vinkers, H. M. et al. SYNOPSIS: SYNthesize and OPtimize
acid sequences by artificial neural networks and simulated (1989). System in Silico. J. Med. Chem. 46, 27652773 (2003).
molecular evolution: de novo design of an idealized leader 32. Gillett, V. A., Johnson, A. P., Mata, P. & Sike, S. Automated 56. Honma, T. et al. Structure-based generation of a new class
peptidase cleavage site. Biophys. J. 66, 335344 (1994). structure design in 3D. Tetrahedron Comput. Method. 3, of potent Cdk4 inhibitors: new de novo design strategy and
8. Schneider, G. et al. Peptide design by artificial neural 681696 (1990). library design. J. Med. Chem. 44, 46154627 (2001).
networks and computer-based evolutionary search. Proc. 33. Lewis, R. A. Automated site-directed drug design: 57. Gillett, V., Johnson, P., Mata, P., Sike, S. & Williams, P.
Natl Acad. Sci. USA 95, 1217912184 (1998). approaches to the formation of 3D molecular graphs. SPROUT: a program for structure generation. J. Comput.
9. Venkatasubramanian, V., Chan, K. & Caruthers, J. M. J. Comput. Aided Mol. Des. 4, 205210 (1990). Aided Mol. Des. 7, 127153 (1993).
Computer-aided molecular design using genetic 34. Rotstein, S. H. & Murcko, M. A. GenStar: a method for de 58. Gillet, V. et al. P. SPROUT: recent developments in the de
algorithms. Computers Chem. Eng. 18, 833844 (1994). novo drug design. J. Comput. Aided. Mol. Des. 7, 2343 novo design of molecules. J. Chem. Inf. Comput. Sci. 34,
10. Venkatasubramanian, V., Sundaram, A., Chan, K. & (1993). 207217 (1994).
Caruthers, J. M. in Genetic Algorithms in Molecular 35. Pearlman, D. A. & Murcko, M. A. CONCERTS: dynamic 59. Mata, P. et al. SPROUT: 3D structure generation using
Modelling (ed. Devillers, J.) 271302 (Academic, London, connection of fragments as an approach to de novo ligand templates. J. Chem. Inf. Comput. Sci. 35, 479493 (1995).
1996). design. J. Med. Chem. 39, 16511663 (1996). 60. Ho, C. M. W. & Marshall, G. R. SPLICE: a program to
11. Sundaram, A. & Venkatasubramanian, V. Parametric Introduces the concept of consensus molecular assemble partial query solutions from three-dimensional
sensitivity and search-space characterization studies of dynamics as a method for structure sampling to de database searches into novel ligands. J. Comput. Aided
genetic algorithms for computer-aided polymer design. novo design. Mol. Des. 7, 623647 (1993).
J. Chem. Inf. Comput. Sci. 38, 11771191 (1998). 36. Liu, H., Duan, Z., Luo, Q. & Shi, Y. Structure-based ligand 61. Gelhaar, D. K. et al. De novo design of enzyme inhibitors
12. Danziger, D. J. & Dean, P. M. Automated site-directed drug design by dynamically assembling molecular building by monte carlo ligand generation. J. Med. Chem. 38,
design: a general algorithm for knowledge acquisition blocks at binding site. Proteins 36, 462470 (1999). 466472 (1995).
about hydrogen-bonding regions at protein surfaces. 37. Zhu, J., Yu, H., Fan, H. Liu, H. & Shi, Y. Design of selective 62. Pierce, A. C., Rao, G., & Bemis, G. W. BREED: generating
Proc. R. Soc. Lond. B 236, 101113 (1989). inhibitors of cyclooxygenase-2 dynamic assembly of novel inhibitors through hybridization of known ligands.
First work about interaction site derivation from a molecular building blocks. J. Comput. Aided Mol. Des. 15, application to CDK2, P38, and HIV protease. J. Med.
receptor structure tailored for the use in automated 447463 (2001). Chem. 47, 27682775 (2004).
de novo design. 38. Zhu, J., Fan, H., Liu, H. & Shi, Y. Structure-based ligand 63. Todorov, N. P. & Dean, P. M. Evaluation of a method for
13. Bhm, H.-J. The computer program LUDI: a new simple design for flexible proteins: application of new F-DycoBlock. controlling molecular scaffold diversity in de novo ligand
method for the de-novo design of enzyme inhibitors. J. Comput. Aided Mol. Des. 15, 979996 (2001). design. J. Comput. Aided. Mol. Des. 11, 175192 (1997).
J. Comput. Aided Mol. Des. 6, 6178 (1992). 39. Pearlman, D. A. & Murcko, M. A. CONCEPTS: new 64. Todorov, N. P. & Dean, P. M. A branch-and-bound
14. Bhm, H.-J. LUDI: rule-based automatic design of new dynamic algorithm for de novo design suggestion. method for optimal atom-type assignment in de novo
substituents for enzyme inhibitor leads. J. Comput. Aided J. Comput. Chem. 14, 11841193 (1993). ligand design. J. Comput. Aided. Mol. Des. 12, 335350
Mol. Des. 6, 593606 (1992). 40. Eldridge, M. D., Murray, C. W., Auton, T. R., Paolini, G. V. & (1998).
15. Clark, D. E. et al. PRO LIGAND: an approach to de novo Mee, R. P. Empirical scoring functions: I. The development 65. Darwin, C. On the Origin of Species (Facsimile of the First
molecular design. 1. Application to the design of organic of a fast empirical scoring function to estimate the binding Edition) (Harvard Univ. Press, Cambridge, Massachusetts,
molecules. J. Comput. Aided Mol. Des. 9, 1332 (1995). affinity of ligands in receptor complexes. J. Comput. Aided 1859/1975).
A comprehensive approach that adopts a lot of Mol. Des. 11, 425445 (1997). 66. Weininger, D. SMILES, a chemical language and
earlier ideas and provides new concepts. 41. DeWitte, R. S. & Shakhnovich, E. I. SMoG de novo design information system. 1. Introduction to methodology and
16. Murray, C. W. et al. PRO_SELECT: combining structure- method based on simple, fast, and accurate free energy encoding rules. J. Chem. Inf. Comput. Sci. 28, 3136
based drug design and combinatorial chemistry for rapid estimates. 1. Methodology and supporting evidence. (1988).
lead discovery. 1. Technology. J. Comp. Aided Mol. Des. J. Am. Chem. Soc. 118, 1173311744 (1996). 67. Pegg, S. C.-H., Haresco, J. J. & Kuntz, I. D. A genetic
11, 193207 (1997). 42. Ishchenko, A. V. & Shakhnovich, E. I. SMall Molecule algorithm for structure-based de novo design. J. Comput.
17. Gillett, V. J., Myatt, G., Zsoldos, Z. & Johnson, A. P. Growth 2001 (SMoG2001): an improved knowledge- Aided Mol. Des. 15, 911933 (2001).
SPROUT, HIPPO and CAESA: tools for de novo structure based scoring function for proteinligand interactions. 68. Schneider, G. & Bhm, H.-J. Virtual screening and fast
generation and estimation of synthetic accessibility. J. Med. Chem. 45, 27702780 (2002). automated docking methods. Drug Discov. Today 7,
Perspect. Drug Discov. Des. 3, 3450 (1995). 43. Wise, A., Gearing, K. & Rees, S. Target validation of 6470 (2002).
18. Rotstein, S. H. & Murcko, M. A. GroupBuild: a fragment- G-protein coupled receptors. Drug Discov. Today 7, 69. Hou, T. & Xu, X. Recent development and application of
based method for de novo drug design. J. Med. Chem. 235246 (2002). virtual screening in drug discovery: an overview. Curr.
36, 17001710 (1993). 44. Waszkowycz, B. et al. PRO LIGAND: an approach to de Pharm. Des. 10, 10111033 (2004).
19. Goodford, P. J. A computational procedure for determining novo molecular design. 2. design of novel molecules from 70. Honma, T. Recent advances in de novo design strategy for
energetically favorable binding sites on biologically important molecular field analysis (MFA) models and practical lead identification. Med. Res. Rev. 23, 606632
macromolecules. J. Med. Chem. 28, 849857 (1985). pharmacophores. J. Med. Chem. 37, 39944002 (1994). (2003).
20. Nishibata, Y. & Itai, A. Automatic creation of dug candidate 45. Nachbar, R. B. Molecular evolution: automated 71. Ji, H. et al. Structure-based de novo design, synthesis,
structures based on receptor structure. Starting point for manipulation of hierarchical chemical topology and its and biological evaluation of non-azole inhibitors specific for
artificial lead generation. Tetrahedron 47, 89858990 application to average molecular structures. Genet. lanosterol 14-demethylase of fungi. J. Med. Chem. 46,
(1991). Programming Evolvable Machines 1, 5794 (2000). 474485 (2003).
21. Bohacek, R. S. & McMartin, C. Multiple highly diverse Development of genetic operators for graph-based 72. Perola, E., Walters, W. P. & Charifson, P. S. A detailed
structures complementary to enzyme binding sites: results structure sampling and detailed description of the comparison of current docking and scoring methods on
of extensive application of a de novo design method problems that have to be solved. systems of pharmaceutical relevance. Proteins 56,
incorporating combinatorial growth. J. Am. Chem. Soc. 46. Pellegrini, E. & Field, M. J. Development and testing of a 235249 (2003).
116, 55605571 (1994). de novo drug-design algorithm. J. Comp. Aided Mol. Des. 73. Schuffenhauer, A. et al. Molecular diversity management
22. Glen, R. C. & Payne, A. W. R. A genetic algorithm for the 17, 621641 (2003). strategies for building and enhancement of diverse and
automated generation of molecules within constraints. 47. Douguet, D., Thoreau, E. & Grassy, G. A genetic algorithm focused lead discovery compound screening collections.
J. Comput. Aided. Mol. Des. 9, 181202 (1995). for the automated generation of small organic molecules: Comb. Chem. High Throughput Screen. 7, 771781
23. Luo, Z., Wang, R. & Lai, L. RASSE: a new method for drug design using an evolutionary algorithm. J. Comput. (2004).
structure-based drug design. J. Chem. Inf. Comput. Sci. Aided Mol. Des. 14, 449466 (2000). 74. Honma, T. et al. A novel approach for the development of
36, 11871194 (1996). 48. Schneider, G., Lee, M.-L., Stahl, M. & Schneider, P. De selective Cdk4 inhibitors: library design based on locations
24. Wang, R., Gao, Y. & Lai, L. LigBuilder: a multi-purpose novo design of molecular architectures by evolutionary of Cdk4 specific amino acid residues. J. Med. Chem. 44,
program for structure-based drug design. J. Mol. Model. assembly of drug-derived building blocks. J. Comput. 46284640 (2001).
6, 498516 (2000). Aided Mol. Des. 14, 487494 (2000). 75. Rogers-Evans, M., Alanine, A. I., Bleicher, K. H., Kube, D.
25. Eisen, M. B., Wiley, D. C., Karplus, M. & Hubbard, R. E. 49. Globus, A., Lawton, J. & Wipke, W. T. Automatic Molecular & Schneider, G. Identification of novel cannabinoid
HOOK: a program for finding novel molecular design using evolutionary algorithms. Nanotechnology 10, receptor ligands via evolutionary de novo design and rapid
architectures that satisfy the chemical and steric 290299 (1999). parallel synthesis. QSAR Comb. Sci. 23, 426430 (2004).
requirements of a macromolecule binding site. Proteins 50. Brown, N., McKay, B., Gilardoni, F. & Gasteiger, J. A 76. Bhm, H.-J., Banner, D. W. & Weber, L. Combinatorial
19, 199221 (1994). graph-based genetic algorithm and its application to the docking and combinatorial chemistry: design of potent
26. Miranker, A. & Karplus, M. An automated method for multiobjective evolution of median molecules. J. Chem. Inf. non-peptide thrombin inhibitors. J. Comput. Aided Mol.
dynamic ligand design. Proteins 23, 472490 (1995). Comput. Sci. 44, 10791087 (2004). Des. 13, 5156 (1999).

662 | AUGUST 2005 | VOLUME 4 www.nature.com/reviews/drugdisc


2005 Nature Publishing Group
REVIEWS

77. Obst, U., Banner, D. W., Weber, L. & Diederich, F. 89. Fonseca, C. M. & Fleming, P. J. in Genetic Algorithms: 101. Frenkel, D. et al. PRO LIGAND: an approach de novo
Molecular recognition at the thrombin active site: Proceedings of the Fifth International Conference (ed. molecular design. 4. Application to the design of peptides.
structure-based design and synthesis of potent and Forrest, S. 416423 (Morgan Kaufmann: San Mateo, J. Comput. Aided Mol. Des. 9, 213225 (1995).
selective thrombin inhibitors and the X-ray crystal CA, 1993). 102. Clark, D. E. & Murray, C. W. PRO LIGAND: an approach to
structures of two thrombin-inhibitor complexes. Chem. 90. Handschuh, S., Wagener, M. & Gasteiger, J. Superposition de novo molecular design. 5. Tools for the Analysis of
Biol. 4, 287295 (1997). of three-dimensional chemical structures allowing for Generated Structures. J. Chem. Inf. Comput. Sci. 35,
78. Olsen, J. A. et al. A fluorine scan of thrombin inhibitors to conformational flexibility by a hybrid method. J. Chem. Inf. 914923 (1995).
map the fluorophilicity/fluorophobicity of an enzyme active Comput. Sci. 38, 220232 (1998). 103. Murray, C. W., Clark, D. E., Byrne, D. G. PRO LIGAND:
site: evidence for CF...C=O interactions. Angew. Chem. 91. Agrafiotis, D. K. Multiobjective optimisation of combinatorial an approach to de novo molecular design. 6. Flexible
Int. Ed. Eng. 42, 25072511 (2003). libraries. IBM J. Res. DeV. 45, 545566 (2001). fitting in the design of peptides. J. Comput. Aided Mol.
79. Gribbon, P. & Sewing A., High-throughput drug discovery: 92. Wright, T., Gillet, V. J., Green, D. V. S. & Pickett, S. D. Des. 9, 381395 (1995).
what can we expect from HTS? Drug Discov. Today 10, Optimizing the size and configuration of combinatorial 104. Grzybowski, B. A. et al. Combinatorial computational
1722 (2005). libraries. J. Chem. Inf. Comput. Sci. 43, 381390 (2003). method gives new picomolar ligands for a known enzyme.
80. Anderson, A. C. & Wright, D. L. The design and docking of 93. Babine, R. E. et al. Design, synthesis and X-ray Proc. Natl Acad. Sci. USA 99, 12701273 (2002).
virtual compound libraries to structures of drug targets. crystallographic studies of novel FKBB-12 ligands. Bioorg. Design of a picomolar human carbonic anhydrase II
Curr. Comp. Aided Drug Des. 1, 103127 (2005). Med. Chem. Lett. 5, 17191724 (1995). inhibitor, the highest-affinity inhibitor to date, with
An excellent overview of current developments in 94. Schindler, T. et al. Structural mechanism of STI-571 the program SMoG.
molecular docking and scoring and its relation to de inhibition of Abelson tyrosine kinase. Science 289, 105. Nachbar, R. B. Molecular evolution: a hierarchical
novo design. 19381942 (2000). representation for chemical topology and its automated
81. Doweyko, A. M. 3D-QSAR illusions. J. Comp. Aided Mol. 95. Lewis, R. A. & Dean, P. M. Automated site-directed drug manipulation. Proc. 3rd Ann. Genetic Programming
Des. 18, 587596 (2004). design: the concept of spacer skeletons for primary structure Conf. 246253 (Univ. of Wisconsin, Madison, Wisconsin
82. Bemis, G. W. & Murcko, M. A. The properties of known generation. Proc. R. Soc. Lond. B 236, 125140 (1989). 1998).
drugs. 1. Molecular frameworks. J. Med. Chem. 39, Pioneering theoretical outline to tackle the problem 106. Wermuth, C. G., Gannelin, C. R., Lindberg, P. and
28872893 (1996). of automated drug design from first principles. Mitscher, L. A. Glossary of terms used in medicinal
83. Mller, G. in Chemogenomics in Drug Discovery (eds 96. Bhm, H.-J. A novel computational tool for automated chemistry. Pure Appl. Chem. 70, 11291143 (1998).
Kubinyi, H. & Mller, G.) 741 (Wiley-VCH, Weinheim, 2004). structure-based drug design. J. Mol. Recognit. 6, 131
84. Jenkins, J. L., Glick, M. & Davies, J. W. A 3D similarity 137 (1993). Acknowledgements
method for scaffold hopping from known drugs or natural Concise overview of the early developments of the H. Kubinyi is thanked for helpful discussion and kind support. This
ligands to new chemotypes. J. Med. Chem. 47, program LUDI. work was supported by the Beilstein-Institut zur Frderung der
61446159 (2004). 97. Bhm, H.-J. The development of a simple empirical Chemischen Wissenschaften, Frankfurt am Main. U.F. is thankful
85. Bailey, D. & Brown, D. High-throughput chemistry and scoring function to estimate the binding constant for a for a fellowship granted by Aventis Pharma Deutschland GmbH,
structure-based design: survival of the smartest. Drug protein-ligand complex of known three-dimensional a company of the Sanofi-Aventis group.
Discov. Today 6, 5759 (2001). structure. J. Comput. Aided Mol. Des. 8, 243256 (1994).
86. Verdonk, M. L. & Hartshorn, M. J. Structure-guided 98. Bhm, H.-J. Prediction of binding constants of protein Competing interests statement
fragment screening for lead discovery. Curr. Opin. Drug ligands: a fast method for the prioritization of hits obtained The authors declare no competing financial interests.
Discov. Devel. 7, 404410 (2004). from de novo design or 3D database search programs.
87. Villar, H. O., Yan, J. & Hansen, M. R. Using NMR for ligand J. Comput. Aided Mol. Des. 12, 309323 (1998).
discovery and optimization. Curr. Opin. Chem. Biol. 8, 99. Stultz, C. M. & Karplus, M. Dynamic ligand design and Online links
387391 (2004). combinatorial optimization: designing inhibitors to
88. Gillet, V. J., Khatib, W., Willett, P., Fleming, P. J. &. Green, endothiapepsin. Proteins 40, 258289 (2000). FURTHER INFORMATION
D. V. S. Combinatorial library design using a multiobjective 100. Westhead, D. R. et al. PRO LIGAND: an approach to de Glossary of terms used in medicinal chemistry:
genetic algorithm. J. Chem. Inf. Comput. Sci. 42, novo molecular design. 3. A genetic algorithm for structure http://www.chem.qmul.ac.uk/iupac/medchem/
375385 (2002). refinement. J. Comput. Aided Mol. Des. 9, 139148 (1995). Access to this interactive links box is free online.

NATURE REVIEWS | DRUG DISCOVERY VOLUME 4 | AUGUST 2005 | 663


2005 Nature Publishing Group

Vous aimerez peut-être aussi