You are on page 1of 30

Kimia & Biologi, Vol.

10, 787-797, September, 2003,2003 Elsevier Science


Ltd. Semua hak dilindungi. DOI 10,1016 / j c dia
m ol bi. 20 03. 09 0,0 0 2

Proses Struktur Berbasis Ulasan


Desain obat
pengantar
During the early 1980s, the ability
Amy C. Anderson * to rationally design drugs using
Dartmouth College protein structures was an
Departemen Kimia unrealized goal for many structural
Burke Laboratories biologists. The first projects were
Hanover, New Hampshire 03755 underway in the mid-80s, and by
the early 1990s the first success
stories were published [1–3]. Today,
Ringkasan even though there is still quite a bit
of fine-tuning necessary to perfect
Bidang desain obat berbasis the process, structure-based drug
struktur merupakan daerah design is an integral part of most
berkembang pesat di mana industrial drug discovery pro-grams
banyak keberhasilan telah [4] and is the major subject of
terjadi dalam beberapa tahun research for many academic
terakhir. Ledakan informasi laboratories.
genomik, proteomik, dan The completion of the human
struktural telah memberikan genome project, the start of both
ratusan target dan peluang baru the proteomics and structural
untuk memimpin obat genomics revolutions, and
penemuan masa depan. Ulasan developments in information
ini merangkum proses desain technol-ogy are fueling an even
obat berbasis struktur dan greater opportunity for structure-
termasuk, terutama, pilihan based drug design to be part of the
target, evaluasi struktur dari success story in the discovery of
target itu, pertanyaan- new drug leads. Excellent drug
pertanyaan penting untuk targets are identified at an
dipertimbangkan dalam Choo- increased pace using
ing metode untuk memimpin developments in bioinformatics.
obat penemuan, dan evaluasi The genes for these targets can be
obat memimpin. prinsip-prinsip cloned quickly, and the protein
kunci dalam bidang desain obat expressed and purified to
berbasis struc-mendatang akan homogeneity. Advances in high-
digambarkan melalui studi throughput crystal-lography, such
kasus yang mengeksplorasi as automation at all stages, more
desain obat untuk AmpC intense synchrotron radiation, and
-lactamase. new developments in phase
determination, have shortened the
timeline for determin-ing
speed at which drug leads can be
structures. Structure determination
identified and evalu-ated in silico.
using nuclear magnetic resonance
(NMR) has also seen a number of Structure-based drug design is
advances in the past years, most powerful when it is a part of
including magnet and probe an entire drug lead discovery
improvements, automated process. A review by J. Antel [9]
assignment [5–7], and new states that the combination of
experimental methods to combinatorial chemistry and
determine larger structures structure-based design can lead to
the parallel synthesis of focused
[8]. Faster computers and the
compound libraries. It is also
availability of relatively
important to consider that
inexpensive clusters of computers
structure-based drug design
have increased the
directs the discovery of a drug
lead, which is not a drug product
*Correspondence: but, specifically, a com-pound with
amy.c.anderson@dartmouth.edu at least micromolar affinity for a
target [10]. The time devoted to
the structure-based drug design
process, as outlined in this review,
may represent only a fraction of
the total time toward developing a
marketable drug product. Many
years of research may be
necessary to convert a drug lead
into a drug that will be both
effective and tolerated by the
human body. Additional years of
research and development will
bring the drug through clinical
trials to finally reach the market.
This review is intended to
provide an overview of the
process of structure-based drug
design from the selec-tion of a
target to the generation and
evaluation of lead compounds. An
in-depth discussion or evaluation
of the computational methods
involved in drug discovery will not
be provided here, since that
subject has been cov-ered in increase potency. Addi-tional
reviews elsewhere [11–17]. cycles include synthesis of the
optimized lead, structure
Overview of the Process determination of the new
The process of structure-based target:lead complex, and further
drug design is an itera-tive one optimization of the lead
(see Figure 1) and often compound. After several cycles of
proceeds through multi-ple the drug design process, the
cycles before an optimized lead optimized compounds usually
goes into phase I clinical trials. show marked improvement in
The first cycle includes the bind-ing and, often, specificity for
cloning, purifi-cation and the target.
structure determination of the
target protein or nucleic acid by Choice of a Drug Target
one of three principal methods: The choice of a drug target is
X-ray crystallography, NMR, or primarily made on a biolog-ical
homology modeling. Using and biochemical basis. The ideal
computer algorithms, compounds target macromole-cule for
or fragments of com-pounds from structure-based drug design is
a database are positioned into a one that is closely linked to human
selected region of the structure. disease and binds a small mole-
These compounds are scored cule in order to carry out a
and ranked based on their steric function. The target molecule
and electrostatic inter-actions Chemistry & Biology
with the target site, and the best 788
compounds are tested with
biochemical assays. In the
second cycle, structure Figure 1. Th
determination of the target in Iterative
complex with a promising lead Process of
from the first cycle, one with at Structure-
least micromolar inhibition in Based Drug
vitro, reveals sites on the com-
Design
pound that can be optimized to
pathogen, and be able to be inhibited
by a

usually has a well-defined


binding pocket. Other de-signed
small molecules can compete, at
a required level of potency, with
the natural small molecule in
order to modulate the function of
the target. Many good drug
targets are proteins; however,
drug design against RNA targets
with well-defined secondary
structure, like the bacterial
ribosome and portions of the HIV
genome, has also been effective.
Recent reviews highlight some of
the RNA structure-based projects
underway [18, 19]. In diseases
caused by the malfunction of
human proteins, small molecule
drugs against G protein coupled
recep-tors (GPCRs) represent at
least 25% of the currently
marketed drugs [20]. Small
molecules that modulate the
function of ion channels,
proteases, kinases, and nuclear
hormone receptors make up
another 22% of the market.
The goal in developing drugs
against the targets listed above
is often to modulate the function
of the human protein; the goal in
developing drugs against
pathogenic organisms is total
inhibition, leading to the death of
the pathogen. Antimicrobial drug
targets should be essen-tial,
have a unique function in the
pathogen, be present only in the
biokimia antara bentuk patogen
dan manusia. Akhirnya, molekul
target harus dapat dihambat
dengan mengikat molekul kecil.
Enzim sering menjadi sasaran
obat yang sangat baik akan
penyebab senyawa dapat
dirancang untuk muat dalam
saku situs aktif.
Cancer targets can be difficult
because the targets are often
somatic cell mutants of proteins
that regulate essential cellular
functions, resulting in the loss of
a function. Of course, it is difficult
for a small molecule to potentiate
the recovery of a function.
However, as pointed out in a
perspective by W. Kaelin [21], a
loss of function in one molecule
is often correlated with a gain of
function in another. The
disruption of oncogenic com-
molekul kecil. target harus Review
penting, dalam hal ini 789
adalah bagian dari siklus
penting dalam sel, dan
eliminasi harus plexes is another difficult problem for
menyebabkan kematian anticancer drug design. For example,
patogen. target harus unik: a chromosomal translocation in core
tidak ada jalur lain harus binding factor b causes the formation
bisa luwes-ment fungsi of a novel chimeric protein that
target dan mengatasi pres- sequesters necessary transcrip-tion
ence inhibitor. Jika factor subunits [22]. Despite the
makromolekul yang difficulty of design-ing a small
memenuhi semua kriteria molecule to disrupt an unwanted
yang digariskan untuk protein association, the specific
menjadi target obat tetapi interface between the fusion protein
fungsi dalam sel manusia and the transcription factor does
yang sehat serta patogen, provide a target that can be
spesifisitas sering dapat exploited. Finally, malignancy often
direkayasa menjadi inhibitor alters the target from its normal
dengan memanfaatkan behavior, leading to interest in the
perbedaan struktural atau
design of specificity for the electron den-sity map is
malignant state. unambiguous. The R factor and Rfree
reported for a model are measures of
Evaluating a Structure the correlation between the model
for Structure-Based and experimental data. The Rfree
Drug Design value should be below 28% and
ideally below 25%, and the R factor
Once a target has been
should be well below 25% in order to
identified, it is necessary to
use the structure in drug design. If
obtain accurate structural
the only structure available for a
information. There are three
particular target does not meet the
primary methods for structure resolution or R factor criteria, drug
determination that are useful for design projects can still be
drug design: X-ray considered, but the results should be
crystallography, NMR, and judged carefully.
homology modeling. The Low coordinate error in a crystal
evaluation of structures from structure is crucial since van der
each method will be discussed. Waals interactions modulate with the
Crystal structures are the most sixth power of the distance between
common source of structural atoms, and directional bonds, such
information for drug design, as hydrogen bonds and electrostatic
since structures determined to inter-actions, have a narrow
high resolution may be tolerance for both the angle and
available, and the method is distance (approximately 0.2 A˚).
useful for proteins that range in Coordinate error can be measured in
size from a few amino acids to many different ways, but two
significant methods are the Luzzati
998 kDa [23]. Another
method [24], based on averag-ing
advantage of crystallography is
coordinate error as a function of R
that ordered water molecules factors that vary with resolution, and
are visible in the experimental methods in which expected errors are
data and are often useful in drug calculated based on the temperature
lead design. A crystal structure factor, or B factor, of an atom and the
should be evalu-ated for the atom:reflection ratio [25]. The Luzzati
resolution of the diffracted coordinate error is often reported in
amplitudes (often simply called coordinates deposited with the
resolution); reliability, or R Protein Data Bank (PDB) and should
factors; coordi-nate error; be in the range of 0.2–0.3 A˚. For
temperature factors; and further accuracy in error
chemical “correct-ness.” determination, the B factor (B) and
Typically, crystal structures atom:reflection ratio (atom/refl) can
determined with data extending be included, as in the Stroud and
to beyond 2.5 A˚ are acceptable Fauman method:
for drug de-sign purposes since
they have a high data to
parameter ratio, and the
placement of residues in the
nucleic acid in solution, are also
valuable sources for drug design.
Since the target is in solution, it is
Expected error 5 sometimes possible to interpret the
0.642 1 0.00852e 7.88B 2 0.687 2 0.00223e dynamics of the target from the data
6.16B e(22)(atom/refl). [28]. Ensembles of structures are
deposited in the PDB, all of which
Temperature factors of atoms satisfy the distance restraints from
in the region of interest should
the ex-perimental data and show
be no greater than the average
reasonable stereochemical
temperature fac-tor for the
molecule. High temperature parameters. There is no analogous
factors can reflect disorder due reliability factor as in
to motion of the residue or crystallography, but the quality of the
ligand or a general indication of structure is often measured by the
error, adding to the inaccuracy rms deviations of the coordinates of
of atomic positions. In a study the members of the ensemble from
reported by Carson et al. [26], the average struc-ture (often divided
the temperature factor was the into main chain and side positions)
most highly corre-lated and overall stereochemical
determinant of R factor. Finally, soundness, including van der Waals
the molecule should be refined violations, phi/psi conformational
to be consistent with all rules of angle anal-ysis, side chain torsion
ste-reochemical “correctness” angle analysis, bond lengths, bond
known from small molecule angles, and planarity. NMR data are
structures; deviations from often col-lected by measuring
ideal bond lengths should be nuclear Overhauser effect (NOE)
no greater than 0.015 A˚ or 38
peaks between resonant nuclei that
for bond angles. Planar atoms
are a distance of less than 5
should be no more than 0.015
A˚ out of the plane, and there A˚terpisah dalam struktur tersier.
should be no incorrect chiral statistik penting untuk mengevaluasi
centers. Finally, at least 90% of NMR yang diturunkan struc-
the backbone w and c angles membangun struktur adalah jumlah
should fall into the most pembatasan NOE terpenuhi, lain-
favored regions of the bijaksana disebut pelanggaran.
Ramachandran plot. The PDB pelanggaran NOE yang crosspeaks
header record lists these antara inti resonansi yang muncul
statistics, and they should be dalam data eksperimen tetapi
evaluated before drug design dijelaskan dalam model. Evaluasi
attempts con-tinue. The results statistik terakhir adalah jumlah total
of a structure evaluation pengekangan NOE per resi-akibat,
program, PROCHECK [27], are atau data: rasio parameter.
also available from the PDB In a survey of 97 deposited NMR
and provide additional detail. structures in the PDB [29],
Structures determined by
Doreleijers et al. found that the
nuclear magnetic reso-nance,
average struc-ture had 11.3
using a concentrated protein or
restraints per residue and 61
NOE viola-tions. The precision
of the structures, as defined by
the circular variance of the
backbone dihedral angles, is
clearly correlated with the
number of restraints per resi-
due. The number of residues in
the most favored regions of the
Ramachandran plot is also
correlated with the number of
restraints per residue and a low
number of NOE violations. The
programs PROCHECK-NMR
[30] and WHAT IF [31], the
results of which are available
from
Chemistry & Biology dalam struktur kristal ditentukan
790 dengan data pada resolusi yang lebih
rendah dari 1,0 A˚. The protonasi dan
tautomerik negara residu serta
keadaan residu histidin (e. D, Atau
the PDB, provide additional keduanya nitrogen terprotonasi) harus
structure-based details for ditetapkan. molekul kecil, seperti ion
evaluating NMR structures. One dan molekul air, dapat dimasukkan
other note to consider is that the selama memimpin fase Wegener-asi
average structure from the dalam kasus di mana mereka
ensemble may or may not actually memainkan peran struktural yang
exist; therefore, one of the sangat penting untuk konformasi dari
members of the ensemble or the target, lain-bijaksana mereka biasanya
entire ensemble itself may be a diangkat untuk memungkinkan setiap
better target choice. potensi menyebabkan menempati
If no experimentally determined posisi mereka.
structure is available, a homology
model can be used for drug Identifikasi Target Site
design [32–34]. To evaluate a Structure-based design begins with the
homology model, SWISS-MODEL identification of a potential ligand
[35] out-puts a confidence factor binding site on the target molecule.
per residue that reflects the Ideally, the target site is a pocket or
amount of structural information protuberance with a variety of potential
used to create that portion of the hydrogen bond donors and ac-ceptors,
model. A higher confidence hydrophobic characteristics, and sizes
number re-flects a lower number of mo-lecular surfaces. The ligand
of templates and therefore a de- binding site can be the ac-tive site, as
creased accuracy. All other in an enzyme, an assembly site with
methods for judging protein another macromolecule, or a
structures, such as communication site necessary in the
stereochemical soundness (bond mechanism of the molecule. In addition
lengths, bond angles, planarity, to the well-accepted protein target
and packing) and resi-dues in the sites, RNA secondary structural
most favored regions of the elements can provide excellent target
Ramachandran plot, apply to sites since they are species specific,
analyzing a homology model as bind ligands, and can be specific for a
well as to experimentally derived disease state [36, 37]. Target sites for
models. protein-protein interactions can be
Menggunakan informasi difficult to locate since these surfaces
struktural yang diperoleh melalui are often flat, large, and hydrophobic,
teknik di atas, struktur ini but even these difficulties can be
kemudian disiapkan untuk surmounted [38–40]. Cocrystal-lization
program desain obat dengan studies in which the target
terlebih dahulu menambahkan macromolecule is crystallized with an
atom hidrogen, biasanya tidak initial small molecule inhibitor can be
invaluable for the determination of
a good target site.

Drug Design Methods torial chemistry, in which


Once the structure and target site thousands of compounds are
are identified, there are several tested for biochemical effects.
paths to developing a good lead The computer-aided methods
based on the structure of the can be further classi-fied into at
target. These paths can be least three categories: inspection,
broadly classified as computer virtual screening, and de novo
aided versus experimental. Com- generation. In the first category,
puter-aided methods will be the inspection, known molecules that
main focus of this re-view. An bind the site, such as substrates
example of an experimental or cofactors in the case of
method, by way of contrast, is enzymes, or peptides in the case
high-throughput screening with of protein:protein or protein:nucleic
combina- acid interactions, are modified to
become inhibitors based on
maximizing complementary
interactions in the target site [1, 3,
41, 42]. In virtual screening,
databases of available small
molecules are docked into the
region of interest in silico and
scored based on predicted inter-
actions with the site. Finally, for de
novo generation small fragments
of molecules, such as benzene
rings, carbonyl groups, amino
groups, etc., are positioned in the
site, scored, and linked in silico.
The final com-pounds, created in
silico from the linked fragments,
then must be synthesized in the
laboratory. There is some overlap
between the virtual screening and
de novo gen-eration
classifications. Some programs,
for example, LUDI, which is
usually used to dock fragments of
com-pounds, are also capable of
docking and scoring entire
compounds. The programs are
classified in Table 1 ac- inhibitors [1, 3, 41, 42, 44, 45]
cording to their primary use. (see Figure 2 for an example).
There are many excellent Initially, the crystal structure is
drug design software meth- solved in the presence of a
ods available capable of substrate, cofac-tor, or drug lead.
either virtual screening or de Then, modifications to direct the
novo generation. This review small molecule toward being a
will focus on a few of the potent inhibitor are designed in
major points necessary to silico based on the interactions of
decide on a particular route the molecule with the target site.
for lead generation. The newly designed compounds
Extensive reviews of the are then scored for binding using
software are available [11, evaluative scoring algorithms
12, 14, 15, 43] and are highly available in virtual screening
recom-mended for further methods.
reading. Docking Available Small
Questions that are pivotal in Molecules versus De Novo
deciding on a method for Generation
lead generation are as The main advantage to docking
follows: (1) are molecules compounds from data-bases such
available which can be as the Available Chemicals
modified to be inhibitors, (2) Database (ACD) into the target
is there a means for site is that hit compounds can be
synthesizing novel pur-chased and tested using
molecules, and (3) what is biochemical assays. Alterna-tively,
the degree of accuracy instead of testing the entire
required at a particular stage database, a database can be
of the design process versus refined to select molecules with a
the time needed for the specific motif.
calculation? Factors such as
the inclusion of protein or
ligand flexibility and the
effects of solvent increase
the time needed for the
calculation but also increase
the predictive value. Each of
these questions will be
discussed with reference to
available drug design algo-
rithms.
Modifying an Initial Compound
Substrates and cofactors for
many proteins have been
modified to become excellent
Review
791

Table 1. Algorithms for Docking Small Molecules or Fragments against a


Target
Flexib Flexi
le ble
Program Protei Ligan Description Referen
n? d? ce

Virtual DOCK no yes docks either small [46–49]


screening molecules or
fragments,
includes solvent effects
FlexX no yes incremental [51]
construction
FlexE yes yes incremental [52]
construction; samples
ensem-
bles of receptor
structures
SLIDE yes yes anchor fragments [50]
placed, remainder of
ligand added;
backbone flexibility
Flo98 no yes can rapidly dock a [76]
large number of ligand
molecules, graphically
view results
ADAM no yes fragments aligned [77]
based on hydrogen
bonding
Hammer no yes genetic algorithms to [78]
head link tail fragments
to anchor fragments
MCSA- yes yes uses simulated [64]
PCR annealing to generate
conformations of target
AUTOD yes yes uses averaged [79]
OCK interaction energy grid
to account for receptor
conformations
and simulated
annealing for ligand
conformations
MCDOC no yes Monte Carlo to sample [80]
K ligand placement
ProDOC yes yes Monte Carlo [81]
K minimization for flexible
ligand, flexible site
ICM yes yes Monte Carlo [82]
minimization for
protein-
ligand docking
DockVis no no Monte Carlo [83]
ion minimization
De novo LUDI no yes docks and scores [54]
generation fragments
of ligands
GRID no yes calculates binding [55]
energies for functional
groups
MCSS no yes exhaustive search of [56]
binding site for
functional group
minima
SMoG no yes knowledge-based [58]
scoring function;
molecules built by
joining rigid fragments
CONCE no yes fills active site with [57]
RTS molecular fragments,
links fragments
Legend no yes grows molecule atom [84]
by atom
DLD no yes saturates binding site [85]
with sp3 carbons,
later linked
GrowMo no yes builds ligands from a [86]
l library of atom types
GenStar no yes builds ligands from sp3 [87]
carbons
GROW no yes constructs a peptide by [88]
residue addition
GroupB no yes builds ligand from a [89]
uild predefined library of
fragments
HOOK no yes searches database of [90]
molecular skeletons
for fit to binding site;
hooks two MCSS
functional groups to
skeleton
SPROU no yes generates skeletons [91]
T that fit site, substi-
tutes atoms into
skeleton to give
molecule
with correct properties
CAVEAT no yes searches database of [92]
small molecules to
connect fragments

compounds are currently being


Programs such as DOCK [46–49], incorporated in a version of LUDI
SLIDE [50], FlexX [51], or FlexE [14, 53]. LUDI [54], GRID [55],
[52] and others (see Table 1) dock MCSS [56], CON-CERTS [57],
databases of compounds and SMoG [58], and others represent
score them according to their examples of de novo lead
inter-actions with the site. Novel generation programs (see Table
scaffolds for inhibitors can be 1).
discovered in this way. Time of Calculation versus
De novo lead generation can Predictive Value
give rise to novel com-pounds; In an initial lead generation run,
however, it does require a team one common goal is to determine
member who can actually the feasibility of the project and
synthesize the intended product in the classes of possible leads that
the labo-ratory. Fragments of may result. Most programs can be
molecules, usually small functional run in a “basic” mode which allows
groups, are docked into the site, this determina-tion. For instance,
scored, and linked together. DOCK [46–49] can position and
Ideally, the fragments can more score all of the compounds in the
fully explore the binding site than ACD quite quickly when run with a
a predefined compound. Means single rigid target, rigid ligands,
for predicting the synthetic and no solvent modeling.
accessibility of the novel However, the predictive value of
these pro-
Chemistry & Biology
792

Figure 2.
Inhibitors for
Thymidylate Several
Synthase potent
Were inhibitors a
Designed shown: (B
Based on
CB3717, (C
Modifications
of the OSI
Cofactor 5,10- 1843U89,
Methylene and (D
Tetrahydrofolat ZD1694
e (Tomudex).
Many programs that allow protein
flexibility incorpo-rate information from
multiple protein structures. En-
sembles of structures can be
grams can be greatly increased experimentally deter-mined, such as
when routines that model protein NMR ensembles (see Figure 3) or
and ligand flexibility as well as multiple crystal structures [61],
solvent contribution are added. computationally pre-
Protein and Ligand Flexibility.
There have been many reports
which emphasize the crucial
effects of including protein and
ligand flexibility in the docking
and scoring process [15, 43, 59].
Most proteins and most ligands
are quite flexible in solution and
may experience a full ensemble
of possible conformations. As a
result, leads generated from a
single, rigid structure may have
dif-fering results in solution than
in silico [60]. In order to account
for the landscape of protein and
ligand confor-mations, several Figure 3. An Ensemble of Six
drug design algorithms Structures of Dihydrofolate Reductase
incorporate protein and/or ligand Six (out of a total of 24 reported)
flexibility. However, modeling mo- structures of dihydrofolate reduc-tase
lecular flexibility, especially for bound to trimethoprim (red) and
the target macromole-cule, NADPH (orange) (1LUD; [93]) are
drastically increases the compute shown. Each member of the
time required for the structure- ensemble is separately colored, and
based drug design (SBDD)
hydrogens are omitted for clarity.
search.
maximized [66]. In a third
capacity, the effect of the solvent
can be incorporated into the
scoring scheme for the
dicted by molecular target:ligand interaction. The
dynamics [62], or generated steps of increased accuracy in
using rotamers of protein modeling the solvent effect
side chains [50, 63]. Using a during scoring are as follows: (1)
molecu-lar dynamics making the assumption that the
simulation to generate molecules are in a vacuum, i.e.,
multiple protein no solvent modeling;
conformations, Carlson et (2) using a fixed dielectric
al. have experimentally veri- constant in estimating electro-
fied a dynamic static contributions; (3) explicit
pharmacophore model for solvation models; and
HIV-1 integ-rase [62]. (4) modeling the Born
Programs which mimic equation. The Born equation cal-
protein flexibility through the culates the polarization
use of ensembles include contribution to solvation when a
SLIDE [50], FlexE [52], and charge is placed within a
MCSA-PCR [64]. spherical solvent cavity. In
Solvent Effects. Solvent general, increased accuracy
plays an important role in comes with increased
ligand binding in several computational cost.
ways. In one capacity, The correct value for the
ordered water molecules dielectric constant of the medium
seen in the structure can be is critical in properly evaluating
incorpo-rated into the electrostatic effects and
designed ligand, effectively estimating binding affinity. In the
increasing ligand binding by Northwest-ern University version
increasing the entropy of of DOCK [49], a solvation correc-
the system (releasing the tion can be added to the score.
bound water molecule). As Possible approaches to achieve
an example, inhibitors for an exact solution to the solvent
HIV protease [65] problem include solving the
incorporate an oxygen atom Poisson-Boltzmann equation,
to substitute for a key water often by using finite differences,
molecule coordinated by or using a free-energy pertur-
residues of the flap region bation technique. Three
of the active site (see approaches have been used in
Figure 4). In a second practice: a modified Born
capacity, ordered water equation [49] to calculate
molecules can be treated solvation energies, an
as bound ligands, and approximation to the electrostatic
contacts with them can be desolvation by modeling the first
solvation shell at the Where a
binding interface [67], and Bound
an implicit model which ac- Water
counts for desolvation by Molecule
computationally generating Was
possible positions of water Visualized in
molecules in the binding
X-Ray
pocket [68].
Structures
Drug Lead Evaluation Nonpeptide
Once a small molecule has HIV
been identified as protease
potentially binding to the inhibitors
target molecule, it must be based on
evaluated
cyclic urea
Review
793 compounds
incorporate
an oxy-gen
atom
Figure 4.
(noted)
Nonpeptide HIV
where a
Protease
bound wate
Inhibitors Based
mole-cule
on Cyclic Urea
was
Compounds
visualized in
Incorpo-rate an
X-ray
Oxygen Atom
structures.
ently an approximation. Both the
solvent effect and the effects of
target and ligand flexibility are
usually impre-cisely described.
Usually, several molecules which
scored well during the docking run
are evaluated in further tests since
even the top scoring molecule
could fail in vitro assays. Leads are
before proceeding to further first evaluated visually with
stages. It is important to consider computer graphics and can often
that the ranking assigned by the be optimized at this step for
scoring func-tion is not always increased affinity. Leads are also
indicative of a true binding evaluated for their likelihood to be
constant, since the model of the orally bioavailable using the “Rule
target:ligand interaction is inher- of 5” [69], which states that good
leads generally have less than five
hydrogen bond donors and less
than ten hydrogen bond
acceptors, a molecular weight less
than 500, and a calculated log of
the partition coefficient (clogP)
less than 5. Rigidifying the lead
can also impart a lower binding
constant by decreasing the
conforma-tional entropy in the ful, karena enzim sering target
unbound state to approach the obat yang baik dan situs aktif
presumably very low memberikan ligan situs mengikat
conformational entropy in the yang sangat baik untuk desain
bound state. Veber and obat. Amprenavir (Agenerase) dan
colleagues [70] state that the nelfinavir (Vira-kecuali bahwa)
number of rotatable bonds should [72], yang dikembangkan
be less than ten in order to terhadap protease HIV, adalah de-
increase the potential for oral ditandatangani menggunakan
bioavailability. Other factors, such metode terutama struktur berbasis
as chemical and metabolic dan adalah dua obat pertama
stability and the ease of synthesis, yang mencapai pasar
can also factor into the deci-sion menggunakan SBDD. Baru-baru
to proceed with a particular ini, zanamivir (Relenza)
candidate lead. Finally, leads are dikembangkan terhadap
brought into the wet lab for neuraminidase [73], Tomudex
biochemical evalu-ation. dikembangkan terhadap timidilat
Promising leads reenter the sintase [44], dan imitinab mesylate
structural determination process to (Glivec) menghambat Abl tyrosine
find the exact binding mode and to kinase [74]. Dengan devel-ngunan
evaluate any further optimization desain berbasis struktur terhadap
that becomes evident. A few sasaran narkoba yang sulit seperti
examples of designed leads have asam nukleat dan protein: protein
shown significant dif-ferences antar-tindakan, terobosan menarik
between predicted and actual baru-baru ini terjadi di lapangan.
binding modes [71], but in many desain obat berbasis struktur-telah
cases the docked and mengungkapkan tertentu, inhibitor
experimental conformations are mikromolar terhadap HIV-1 RNA
within 2 A˚ rmsd [16]. target yang TAR [36, 37], IL-2 / IL-
AmpC-Lactamase Case Study 2RSebuahInteraksi reseptor [39],
There have been many important yang VEGF / VEGF reseptor [40],
successes in struc-ture-based dan Bcl2 [33]. desain berbasis
drug design. Struc-mendatang melawan target
The discovery of enzyme enzim AmpCb-lactamase
inhibitors has been success- menggambarkan prinsip-prinsip
desain obat out-berbaris di Versi Northwestern University of
ulasan ini dan akan dibahas lebih DOCK [47, 49] digunakan untuk
lanjut dalam bagian ini. menyaring ACD terhadap
b-lactamases adalah enzim- konsensus struc-mendatang, “hot
enzim bakteri yang spot” model AmpC b-lactamase.
menyebabkan re-sistance untuk Struktur konsensus dimasukkan
bantibiotik -lactam seperti com- eksperimen dan
monly resep penisilin obat- Kimia & Biologi
obatan dan sefalosporin. b- 794
lactamase adalah target obat
yang baik karena unik patogen,
dapat dihambat oleh molekul
kecil, dan sangat penting untuk
ketahanan patogen untuk
bantibiotik -lactam. Itu benzim
-lactamase memiliki serin
nukleofil di situs aktif yang
membelah bcincin -lactam
antibiotik, secara efektif
menghancurkan setiap manfaat
farmasi. binhibitor -lactamase,
seperti asam klavulanat, sering
diberikan bersama b-lactam Gambar 5. Desain Obat terhadap
antibiotik, tetapi inhibitor ini b- AmpC b-Lactamase
lactams mereka-diri,
(A) Bola-dan-tongkat representasi dari
menyebabkan peningkatan
senyawa 1 (merah), ditemukan
regulasi ekspresi b-lactamase. dengan layar DOCK, terikat untuk
Novel binhibitor -lactamase yang AmpC b-lactamase.
tidak meningkatkan ekspresi (B) Senyawa 1 (ruang mengisi) terikat
yang diperlukan untuk mencegah AmpC b-lactamase (residu dalam
resistensi antibiotik. waktu 7 A˚ ditunjukkan dengan
permukaan van der Waals).
inhibitors. The top 500 scoring
komputasi berasal Data ligan molecules from the DOCK run were
mengikat dari 13 AmpC b- examined graphically for complemen-
lactamase structures [75]. The tarity, polar interactions, and
consensus bind-ing sites for agreement with the identi-fied binding
AmpC b-lactamase include an sites. Fifty-six compounds were
amide recog-nition site, an purchased and tested with in vitro
oxyanion hole, hydroxyl and assays. Three compounds inhibit with
carboxyl binding sites, and, Ki 5 650 mM or better. Compound 1
finally, four ordered water was shown to be selective for AmpC
molecules shown to consistently b-lactamase over other serine
bind either the enzyme or the
nucleophile enzymes and was found to be essential. The
selected for further study. addition of a piperidine ring to
Powers et al. [66] determined the distal aryl ring increased
the cocrystal structure of AmpC binding by 2-fold. Finally,
b-lactamase and compound 1
(Figure 5). The structure was compound 1 is relatively “drug-
determined to a resolution of like,” according to Lip-inski’s
1.94 A˚, with R factor 17.3% and rules [69], and has sites for
Rfree 20.7%, coordinate error
0.19 A˚, average B factor 23 A˚2, future synthetic elabo-ration.
and average B factor for com- In summary, AmpC b-
pound 1, 37 A˚2. The structure is lactamase is an excellent drug
stereochemically cor-rect, citing
an rmsd from ideality for bond target with accurate structural
lengths 5 0.009 A˚ and bond information. The North-western
angles 5 1.58. The DOCK- University version of DOCK was
predicted conformation of used to screen the ACD to find
compound 1 closely resembles
the crystallographically novel inhibitor scaffolds. The top-
determined conformation of com- scoring compounds were novel
pound 1. In fact, the rmsd for all and predicted to have comple-
inhibitor atoms is 1.87 A˚ for one
molecule in the asymmetric unit mentary interactions with the
of the crystal and 1.75 A ˚ for the target site, but were shown to
second molecule in the have relatively low binding
asymmetric unit. The predicted
interactions were also highly constants in solution. Fur-ther
corre-lated with the improvement will be needed
crystallographically determined before the drug lead can
interac-tions: of nine hydrogen proceed into future trials.
bonds observed in the crystal
structure, seven were predicted, Structural studies of the selected
and of eight hydrogen bonds inhibitor and the enzyme are
predicted, only one was not invaluable in fu-ture chemical
observed crystallo-graphically.
elaboration.
Compound 1 was tested in
The results of the AmpC b-
microbiology experiments and
lactamase case study also
found to reduce the minimum
exemplify the sort of reasonable
inhibitory concentra-tion (MIC) of
expectations one should have for
ampicillin by 4-fold in b-
initial structure-based drug
lactamase-positive bacteria.
design stud-ies. One, micromolar
Analogs of compound 1 were
inhibitors were discovered
tested to deter-mine which
through the docking procedure
functional moieties were
and will serve as lead com-
essential. The car-boxylate
pounds requiring further
group, the proton donating ability
modification for increased po-
of the sulfon-amide, and the
tency. It is very rare that
atom order of the sulfonamide
extremely potent inhibitors (nM
were
inhibition or better) are
discovered during docking
screens. Two, 56 top-scoring
compounds were pur- process of reen-tering structural
chased and tested in vitro determination and reevaluation for
after the initial docking opti-mization. Focused libraries of
screen. Due to synthesized compounds based on
approximations in the the structure-based lead can create a
models of protein and very promising lead which can
ligand interactions in the continue to phase I clinical trials.
scoring algorithms, the As structural genomics,
docked compounds may be bioinformatics, and computa-tional
ranked in slightly different power continue to explode with new
order than their in vitro advances, further successes in
assays reveal. In fact, some structure-based drug design are likely
of the hits from the docking to follow. Each year, new targets are
study may not exhibit being identi-fied, structures of those
successful in vitro results at targets are being determined at an
all. Structure-based drug amazing rate, and our capability to
design methods increase capture a quan-titative picture of the
the chance that a “hit” will interactions between macromole-
be found in the top-ranked cules and ligands is accelerating.
ligands.
References
Promise for the Future
1. Roberts, N., Martin, J.,
Structure-based drug
design is a powerful Kinchington, D., Broadhurst, A.,
method, es-pecially when Craig, J., Duncan, I., Galpin, S.,
used as a tool within an Handa, B., Kay, J., Krohn, A., et al.
armamentarium, for (1990). Rational design of peptide-
discovering new drug leads based HIV proteinase inhibi-tors.
against important tar-gets. Science 248, 358–361.
After a target and a 2. Erickson, J., Neidhart, D., VanDrie,

structure of that target are J., Kempf, D., Wang, X.,


Review Norbeck, D., Plattner, J.,
795 Rittenhouse, J., Turon, M.,
Wideburg, N., et al. (1990). Design,
activity and 2.8 A˚ crystal structure
of a C2 symmetric inhibitor
complexed to HIV-1 protease.
chosen, new leads can be Science 249, 527–533.
designed from chemical prin- 3. Dorsey, B.D., Levin, R.B.,
ciples or chosen from a subset
McDaniel, S.L., Vacca, J.P., Guare,
of small molecules that scored
well when docked in silico J.P., Darke, P.L., Zugay, J.A.,
against the target. After a Emini, E.A., Schleif, W.A.,
preliminary assessment of Quintero, J.C., et al. (1994). L-
bioavailability, the candidate 735,524: the design of a potent and
leads continue in an iterative orally available HIV protease
inhibitor. J. Med. Chem. 37, macromolecules in solution. Proc.
3443–3451. Natl. Acad. Sci. USA 94, 12366–
4. Mountain, V. (2003). Astex, 12371.
Structural Genomix, and 9. Antel, J. (1999). Integration of
Syrrx. Chem. Biol. 10, 95–98. combinatorial chemistry and
5. Zheng, D., Huang, Y., structure-based drug design. Curr.
Moseley, H., Xiao, R., Aramini, Opin. Drug Discov. Dev. 2, 224–
J., Swapna, G., and 233.
Montelione, G. (2003). 10. Verlinde, C., and Hol, W. (1994).

Automated protein fold Structure-based drug design:


determi-nation using a progress, results and challenges.
minimal NMR constraint Structure 2, 577–587.
strategy. Protein Sci. 12, 11. Taylor, R., Jewsbury, P., and Essex,

1232–1246. J. (2002). A review of pro-tein-small


6. Oezguen, N., Adamian, L., Xu, molecule docking methods. J.
Y., Rajarathnam, K., and Comput. Aided Mol. Des. 16, 151–
Braun, W. (2002). Automated 166.
assignment and 3D structure 12. Joseph-McCarthy, D. (1999).
calculations using Computational approaches to
combinations of 2D structure-based ligand design.
homonuclear and 3D Pharmacol. Ther. 84, 179–191.
heteronuclear NOESY 13. Carlson, H., and McCammon, J.

spectra. J. Biomol. NMR 22, (1999). Method for including the


249–263. dynamic fluctuations of a protein in
7. Bailey-Kellogg, C., Widge, A., computer-aided drug design. J.
Kelley, J., Berardi, M., Phys. Chem. A 103, 10213–10219.
Bushweller, J., and Donald, B. 14. Bohacek, R., and McMartin, C.
(2000). The NOESY jigsaw: (1997). Modern computational
automated protein secondary chemistry and drug discovery:
structure and main-chain structure generating programs.
assignment from sparse, Curr. Opin. Chem. Biol. 1, 157–
unassigned NMR data. J. 161.
Comput. Biol. 7, 537–558. 15. Carlson, H., dan McCammon, J.

8. Pervushin, K., Riek, R., Wider,


(2000). protein akomodatif
G., and Wutrich, K. (1997).
Attenu-ated T2 relaxation by
mutual cancellation of dipole-
dipole cou-pling and chemical
shift anistropy indicates an
avenue to NMR structures of
very large biological
K., Liu, P., Laue, T., Biltonen, R.,
et al. (2002). Altered affinity of
CBFb-SMMHC for Runx1 explains
fleksibilitas dalam desain its role in leukemogenesis. Nat.
obat komputasi. Mol. Struct. Biol. 9, 674–679.
Pharmacol. 57, 213-218. 23. Nissen, P., Hansen, J., Ban, N.,
16. Shoichet, B., McGovern, S.,
Moore, P., and Steitz, T. (2000).
Wei, B., dan Irwin, J. (2002). The structural basis of ribosome
Memimpin penemuan activity in peptide bond synthe-sis.
menggunakan docking Science 289, 920–930.
molekuler. Curr. Opin. Chem. 24. Luzzati, V. (1952). The statistical
Biol. 6, 439-446. treatment of errors in crystal
17. Klebe, G., dan Bohm, H. structures. Acta Crystallogr. 5,
(1997). faktor energik dan 802–810.
entropis menentukan afinitas 25. Stroud, R., and Fauman, E.
mengikat dalam kompleks (1995). Significance of structural
protein-ligan. J. Re-kecuali changes in proteins: Expected
bahwa. Sinyal Transduct. errors in refined protein struc-
Res. 17, 459-473. tures. Protein Sci. 4, 2392–2404.
18. Gallego, J., dan Varani, G.
26. Carson, M., Bugg, C., DeLucas,
(2001). Menargetkan RNA L., and Narayana, S. (1994).
dengan obat-molekul kecil: Comparison of homology model to
janji terapi dan tantangan the experimental structure of a
kimia. Acc. Chem. Res. 34, novel serine protease. Acta
836-843. Crystallogr. D Biol. Crystallogr. 50,
19. Afshar, M., Prescott, C., dan
889–899.
Varani, G. (1999). Struktur 27. Laskowski, R., MacArthur, M.,
berbasis dan pencarian Moss, D., and Thornton, J. (1993).
kombinasi obat RNA- PROCHECK: a program to check
mengikat baru. Curr. Opin. the stereochemical quality of
Biotechnol. 10, 59-63. protein structures. J. Appl.
20. Hopkins, A., dan Groom, C.
Crystallogr. 26, 283–291.
(2002). The druggable 28. Pellecchia, M., Sern, D., and
genom. Nat. Rev. Obat Wutrich, K. (2002). NMR in drug
Discov. 1, 727-730. discovery. Nat. Rev. Drug Discov.
21. Kaelin, W. (1999). Choosing
1, 211–219.
anticancer drug targets in the 29. Doreleijers, J., Rullman, J., and
post-genomic era. J. Clin. Kaptein, R. (1998). Quality as-
Invest. 104, 1503–1506. sessment of NMR structures: a
22. Lukasik, S., Zhang, L., statistical survey. J. Mol. Biol. 281,
Corpora, T., Tomanicek, S., 149–164.
Li, Y., Kundu, M., Hartman,
30. Laskowski, R., Rullman, J., Func-tional Genomics, M. Wilkins,
MacArthur, M., Kaptein, R., K. Williams, R. Appel, and D.
and Thornton, J. (1996). Hoch-strasser, eds. (New York:
AQUA and PROCHECK- Springer), pp. 177–186.
NMR: programs for checking 36. Lind, K., Du, Z., Fujinaga, K.,
the quality of protein Peterlin, B., and James, T. (2002).
structures solved by NMR. J. Structure-based computational
Biomol. NMR 8, 477–486. database screening, in vitro
31. Vriend, G. (1990). assay, and NMR assessment of
BAGAIMANA JIKA: a compounds that target TAR RNA.
pemodelan molekul dan Chem. Biol. 9, 185–193.
program desain obat. J. Mol. 37. Filikov, A., Mohan, V., Vickers, T.,

Grafik. 8, 52-56. Griffey, R., Cook, P., Abagyan, R.,


32. Enyedy, I., Lee, S., Kuo, A., and James, T. (2000).
Dickson, R., Lin, C., dan Identification of ligands for RNA
Wang, S. (2001). pendekatan targets via structure-based virtual
berbasis struktur-untuk screening: HIV-1 TAR. J. Comput.
penemuan bis-benzamidines Aided Mol. Des. 14, 593–610.
sebagai inhibitor novel 38. Gadek, T., and Nicholas, J.

matriptase. J. Med. Chem. (2003). Small molecule


44, 1349-1355. antagonists of proteins. Biochem.
33. Enyedy, I., Ling, Y., Nacro, Pharmacol. 65, 1–8.
K., Tomita, Y., Wu, X., Cao, 39. Tilley, J., Chen, L., Fry, D.,

Y., Guo, R., Li, B., Zhu, X., Emerson, S., Powers, G., Biondi,
Huang, Y., et al. (2001). D.,
Penemuan inhibitor-molekul
kecil dari Bcl-2 melalui
screening komputer berbasis
struktur. J. Med. Chem. 44,
4313-4324.
34. Schapira, M., Raaka, B.,
Samuels, H., dan Abagyan,
R. (2001). Dalam penemuan
silico novel asam retinoat
reseptor agonis struc-
membangun struktur. BMC
Struct. Biol. 1, 1.
35. Peitsch, M., and Guex, N.

(1997). Large-scale
comparative pro-tein
modeling. In Proteome
Research: New Frontiers in
Chemistry & Biology bagaimana untuk memukul target
796 bergerak. Curr. Opin. Chem. Biol. 6,
447-452.
44. Rutenber, E., dan Stroud, R. (1996).

Varnell, T., Trilles, R., Guthrie, Pengikatan antikanker ZD1694 obat


R., Mennona, F., et al. (1997). untuk E. coli timidilat sintase: menilai
Identification of a small tertentu-ity dan afinitas. Struktur 4,
molecule inhibitor of the IL-2/IL- 1317-1324.
2R re-ceptor interaction which 45. Stout, T., dan Stroud, R. (1996).
Kompleks dari terapi anti-kanker,
binds IL-2. J. Am. Chem. Soc. BW1843U89, dengan timidilat
119, 7589–7590. ˚
sintase pada 2,0 A Resolusi: implikasi
40. Wiesmann, C., Christinger, H., untuk modus baru penghambatan.
Cochran, A., Cunningham, B., Struktur 4, 67-77.
46. Kuntz, I., Blaney, J., Oatley, S.,
Fairbrother, W., Keenan, C.,
Meng, G., and DeVos, A. Langridge, R., and Ferrin, T. (1982).
(1998). Crystal structure of the A geometric approach to
complex between VEGF and a macromolecular-ligand inter-actions.
receptor-blocking peptide. J. Mol. Biol. 161, 269–288.
47. Lorber, D., and Shoichet, B. (1998).
Biochemistry 37, 117765–
117772. Flexible ligand docking using
41. Varney, M., Marzoni, G., Palmer,
conformational ensembles. Protein
C., Deal, J., Webber, S., Welsh, Sci. 7, 938–950.
48. Ewing, T., Makino, S., Skillman, G.,
K., Bacquet, R., Bartlett, C.,
Morse, C., Booth, C., et al. and Kuntz, I. (2001). DOCK 4.0:
(1992). Crystal structure-based search strategies for automated
drug design and synthesis of molecular docking of flexi-ble
benz[c-d]indole-containing molecule databases. J. Comput.
inhibitors of thymidylate Aided Mol. Des. 15, 411–428.
49. Shoichet, B., Leach, A., and Kuntz, I.
synthase. J. Med. Chem. 35,
663–676. (1999). Ligand solvation in molecular
42. Chan, D., Laughton, C., docking. Proteins 34, 4–16.
50. Schnecke, V., Swanson, C., Getzoff,
Queener, S., dan Stevens, M.
(2001). Studi struktural pada E., Tainer, J., and Kuhn, L. (1998).
senyawa bioaktif. 34. Desain, Screening a peptidyl database for
Synthe-sis, dan evaluasi potential ligands to proteins with
biologis triazenyl tersubstitusi side-chain flexibility. Proteins 33, 74–
inhibitor pyrimeth-amina dari 87.
51. Kramer, B., Metz, G., Rarey, M., and
Pneumocystis carinii dihidrofolat
reduc-tase. J. Med. Chem. 44, Lengauer, T. (1999). Ligand docking
2555-2564. and screening with FlexX. Med.
43. Carlson, H. (2002). Protein Chem. Res. 9, 463–478.
fleksibilitas dan desain obat:
52. Claussen, H., Buning, C., Rarey, estimates. 1. Methodology and
M., and Lengauer, T. (2001). supporting evidence. J. Am. Chem.
FlexE. Efficient molecular Soc. 118, 11733–11744.
docking considering protein 59. Davis, A., and Teague, S. (1999).

structure variations. J. Mol. Biol. Hydrogen bonding, hydropho-bic


308, 377–395. interactions, and failure of the rigid
53. Boehm, H. (1996). Towards the receptor hypothesis. Angew. Chem.
automatic design of synthetically Int. Ed. Engl. 38, 736–749.
accessible protein ligands: 60. Anderson, A., O’Neil, R., Surti, T.,

Peptides, amides and and Stroud, R. (2001). Ap-proaches


peptidomi-metics. J. Comput. to solving the rigid receptor problem
Aided Mol. Des. 10, 265–272. by identifying a minimal set of flexible
54. Boehm, H. (1992). The residues during ligand docking.
computer program, LUDI: a new Chem. Biol. 8, 445–457.
method for the de novo design
of enzyme inhibitors. J. Comput.
Aided Mol. Des. 6, 61–78.
55. Goodford, P. (1985). A
computational procedure for
determining energetically
favorable binding sites on
biologically important
macromolecules. J. Med. Chem.
28, 849–857.
56. Caflisch, A., Miranker, A., and

Karplus, M. (1993). Multiple


copy simultaneous search and
construction of ligands in
binding sites: applications to
inhibitors of HIV-1 aspartic
proteinase. J. Med. Chem. 36,
2142–2167.
57. Pearlman, D., and Murcko, M.

(1996). CONCERTS: dynamic


con-nection of fragments as an
approach to de novo ligand
design. J. Med. Chem. 39,
1651–1663.
58. DeWitte, R., and Shakhnovich,

E. (1996). SMoG: a de novo de-


sign method based on simple,
fast, and accurate free energy
66. Powers, R., Morandi, F., and
Shoichet, B. (2002). Structure-
based discovery of a novel,
61. Knegtel, R., Kuntz, I., and noncovalent inhibitor of AmpC
Oshiro, C. (1997). beta-lactamase. Structure 10,
Molecular docking to 1013–1023.
ensembles of protein 67. Majeux, N., Scarsi, M., and
structures. J. Mol. Biol. Caflisch, A. (2001). Efficient
266, 424–440. electro-static solvation model for
62. Carlson, H., Masukawa, K.,
protein-fragment docking.
Jorgensen, W., Lins, R., Proteins 42, 256–268.
Briggs, J., and 68. Bohm, H. (1998). Prediction of
McCammon, J. (2000). binding constants of protein li-
Developing a dynamic gands: A fast method for the
pharmaco-phore model for prioritization of hits obtained
HIV-1 integrase. J. Med. from de novo design or 3D
Chem. 43, 2100–2114. database search programs. J.
63. Lovell, S., Word, J.,
Comput. Aided Mol. Des. 12,
Richardson, J., and 309–323.
Richardson, D. (2000). The 69. Lipinski, C., Lombardo, F.,
penultimate rotamer library. Dominy, B., and Feeney, P.
Proteins 40, 389–408. (1997). Experimental and
64. Ota, N., and Agard, D. computational approaches to
(2001). Binding mode estimate solu-bility and
prediction for a flexible permeability in drug discovery
ligand in a flexible pocket and development set-tings. Adv.
using multi-conformation Drug Deliv. Rev. 23, 3–25.
sim-ulated annealing 70. Veber, D., Johnson, S., Cheng,
pseudo crystallographic
H., Smith, B., Ward, K., and
refinement. J. Mol. Biol.
Kopple, K. (2002). Molecular
314, 607–617.
properties that influence the oral
65. Lam, P., Jadhav, P.,
bioavailability of drug
Eyerman, C., Hodge, C.,
Ru, Y., Bacheler, L., Meek, candidates. J. Med. Chem. 45,
J., Otto, M., Rayner, M., 2615–2623.
71. Fritz, T., Tondi, D., Finer-Moore,
and Wong, Y. (1994).
Rational design of potent, J., Costi, M., and Stroud, R.
bioavailable, nonpeptide (2001). Predicting and
cyclic ureas as HIV harnessing protein flexibility in
protease inhibitors. the design of species-specific
Science 263, 380–384. inhibitors of thymidylate
synthase. Chem. Biol. 8, automatic search method for
981–995. stable docking models of protein
72. Kaldor, S., Kalish, V., and ligand. J. Mol. Biol. 243,
Davies, J., Shetty, B., Fritz, 310–326.
J., Appelt, K., Burgess, J., 78. Welch, W., Ruppert, J., and
Campanale, K., Chirgadze, Jain, A. (1996). Hammerhead:
N., Clawson, D., et al. fast, fully automated docking of
(1997). Viracept (Nelfinavir flexible ligands to protein
Mesylate AG1343): A binding sites. Chem. Biol. 3,
potent, orally bioavailable 449–462.
inhibitor of HIV-1 protease. 79. Goodsell, D., Morris, G., and

J. Med. Chem. 40, 3979– Olson, A. (1996). Automated


3985. dock-ing of flexible ligands:
73. Varghese, J. (1999). applications of AutoDock. J.
Development of Mol. Recog-nit. 9, 1–5.
neuraminidase inhibitors 80. Liu, M., and Wang, S. (1999).

as anti-influenza virus MCDOCK: A Monte Carlo


drugs. Drug Dev. Res. 46, simula-tion approach to the
176–196. molecular docking problem. J.
74. Schindler, T., Bornmann, Comput. Aided Mol. Des. 13,
W., Pellicena, P., Miller, W., 435–451.
Clarkson, B., and Kuriyan, 81. Trosset, J., and Scheraga, H.

J. (2000). Structural (1999). Prodock: software


mechanism for STI-571 package for protein modeling
inhibition of abelson and docking. J. Comput. Chem.
tyrosine kinase. Science 20, 412–427.
289, 1938–1942. 82. Abagyan, R., Totrov, M., and

75. Powers, R., and Shoichet, Kuznetsov, D. (1994). ICM—a


B. (2002). Structure-based new method for protein
approach for binding site modeling and design—
identification on AmpC b- applications to dock-ing and
lactamase. J. Med. Chem. structure prediction from the
45, 3222–3234. distorted native conforma-tion.
76. McMartin, C., and J. Comput. Chem. 15, 488–506.
Bohacek, R. (1997). QXP: 83. Hart, T., and Read, R. (1992).

Powerful, rapid computer Proteins 13, 206–222.


algorithms for structure- 84. Nishibata, Y., and Itai, A. (1993).

based drug design. J. Confirmation of usefulness of a


Com-put. Aided Mol. Des. structure construction program
11, 333–344. based on three-dimensional
77. Mitzutani, M., Tomioka, N.,

and Itai, A. (1994). Rational


Review
797

receptor structure for rational lead generation. J. Med. Chem.


36, 2921–2928.
85. Miranker, A., and Karplus, M.

(1995). An automated method


for dynamic ligand design.
Proteins 23, 472–490.
86. Bohacek, R., and McMartin, C.

(1994). Multiple highly diverse


structues complementary to
enzyme binding sites: Results of
extensive application of a de
novo design method
incorporating combinatorial
growth. J. Am. Chem. Soc. 116,
5560–5571.
87. Rotstein, S., and Murcko, M.

(1993). GenStar: a method for


de novo drug design. J.
Comput. Aided Mol. Des. 7, 23–
43.
88. Moon, J., and Howe, W. (1991).

Computer design of bioactive


molecules: a method for
receptor-based de novo ligand
design. Proteins 11, 29–34.
89. Rotstein, S., and Murcko, M.

(1993). GroupBuild: a fragment-


based method for de novo drug
design. J. Med. Chem. 36,
1700–1710.
90. Eisen, M., Wiley, D., Karplus,

M., and Hubbard, R. (1994).


HOOK: a program for finding
novel molecular architectures
that satisfy the chemical and
steric requirements of
macromolecule binding sites.
Proteins 19, 199–221.
91. Gillet, V., Johnson, P., Mata, P.,

Sike, S., and Williams, P.


(1993). SPROUT: a program for
structure generation. J. Comput.
Aided Mol. Des. 7, 127–153.
92. Bartlett, P., Shea, G., Telfer, S.,

and Waterman, S. (1989). CA-


VEAT: a program to facilitate the
structure-derived design of
biologically active molecules. In
Molecular Recognition: Chemi-
cal and Biological Problems, S.
Roberts, ed. (Cambridge: Royal
Society of Cambridge), pp. 182–
196.
NMR-based
dihydrofolate
67–70. solution structure
reductase of the complex
with trimethoprim and of Lac-tobacillus
NADPH. casei
J. Biomol. NMR 24,

You might also like