You are on page 1of 15

ª 2009 John Wiley & Sons A/S

Chem Biol Drug Des 2010; 75: 3–17 doi: 10.1111/j.1747-0285.2009.00899.x

Review Article

Structure-based Drug Metabolism Predictions for


Drug Design

Hao Sun* and Dennis O. Scott oral drug bioavailability, which initiated the era of data mining and
computer-aided drug design in pharmaceutical industry (1). Another
Department of Pharmacokinetics, Dynamics, and Drug Metabolism, virtual 'prediction' example is the 'structure alert chart' summarized
Pfizer Inc., Groton, CT 06340, USA by medicinal chemists for drug design purpose. To develop compre-
*Corresponding author: Hao Sun hao.sun@pfizer.com hensive drug metabolism prediction methods will offer a powerful
means to identify and analyze potential pharmacokinetic and toxico-
Significant progress has been made in structure- logical problems.
based drug design by pharmaceutical companies
at different stages of drug discovery such as iden- Metabolism is a major consideration for modifying drug clearance
tifying new hits, enhancing molecule binding affin- and a primary source for drug metabolite-induced drug toxicity. An
ity in hit-to-lead, and reducing toxicities in lead appropriate pharmacokinetic profile such as a reasonable half-life is
optimization. Drug metabolism is a major consid- mainly controlled by drug metabolism, which should be adapted to
eration for modifying drug clearance and also a the desired purpose and is extremely important for drug develop-
primary source for drug metabolite-induced toxic- ment. Drug-metabolizing enzymes catalyze phase I (such as hydroly-
ity. With major cytochrome P450 structures identi-
sis, reduction and oxidation) and phase II (such as glucuronidation,
fied and characterized recently, structure-based
sulfation, acetylation, methylation and glutathione conjugation) reac-
drug metabolism prediction becomes increasingly
attractive. In silico methods based on molecular tions (2). The oxidation reactions in phase I often cause toxicities
and quantum mechanics such as docking, molecu- and ⁄ or drug–drug interactions. Cytochrome P450s are major enzymes
lar dynamics and ab initio chemical reactivity cal- to catalyze the oxidative bioactivation, and other enzymes are flavin
culations bring us closer to understand drug monooxygenase, aromatase, monoamine oxidase, and alcohol ⁄ alde-
metabolism and predict drug–drug interactions. In hyde dehydrogenase. To elucidate the underlying oxidation mecha-
this study, we review important progress in drug nisms will help improving pharmacokinetic and drug safety profiles.
metabolism and common in silico techniques To date, 57 human P450 isoforms are known (3). They catalyze the
adopted to predict drug regioselectivity, stereose-
metabolic activation of xenobiotics ⁄ drugs, sterols, fatty acids, eico-
lectivity, reactive metabolites, induction, inhibi-
sanoids and vitamins. Most of them are expressed in human livers
tion and mechanism-based inactivation, as well as
their implementation in hit-to-lead drug discovery. including CYP1A2, 2A6, 2B6, 2C8, 2C9, 2C18, 2C19, 2D6, 2E1, 3A4
and 3A5, and others are in human lungs such as CYP1A1, 1B1, 2A6,
Key words: cytochrome P450, drug design, drug metabolism, 2B6, 2E1, 2F1, 2J2, 2S1, 3A5 and 4B1 or in minor sites including kid-
structure-based ney, brain, small intestine, peripheral blood cells, platelets, neutroph-
ils, and seminal vesicles (3). CYP1A2, 2C9, 2C19, 2D6, 2E1, and
3A4 ⁄ 5 are major ones generally used for in vitro drug metabolism
The goal of drug discovery is to find best medicines to prevent, profiling studies in current discovery settings. They contribute to a
treat and cure diseases quickly and efficiently. To fulfill this goal, combined metabolism of over 90% of all drugs in the market, and
computational tools have helped medicinal chemists modify and their individual contribution to the total hepatic P450 expression is
optimize molecules to potent drug candidates, have led biologists approximate 15%, 20%, 5%, 5%, 10%, and 30%, respectively (3).
and pharmacologists to explore new disease genes and novel drug
targets, and have been also guiding drug metabolism scientists to Over the past 30 years, researchers have attempted to pinpoint the
achieve better pharmacokinetic profiles and avoid drug toxicities. nature and mechanism of P450-catalyzed drug metabolism by com-
These in silico approaches have been widely applied to predict drug paring substrate selectivity, identifying reaction intermediates, char-
absorption, distribution, metabolism, excretion and toxicity (ADMET). acterizing products, and most recently studying enzyme structures in
The optimization of chemical space in early discovery stage using a molecular level. The most relevant and accepted catalysis mecha-
these in silico tools will shorten the total drug discovery cycle time nism includes several major steps. In the catalytic cycle, it starts
and at the same time enhance the late-stage drug survival rate. with the binding of the substrate to the active site of P450s, then
Lipinski's 'Rule-of-Five' is a rule of thumb to predict small-molecule reduction of the ferric heme iron to its ferrous state by co-enzyme

Editor’s invited manuscript to celebrate the 4th Anniversary of Chemical Biology & Drug Design.

3
Sun and Scott

and reductase, then binding of molecular oxygen followed by a sec- To simplify the complexity of drug metabolism prediction, a hybrid
ond one-electron transfer and two-step protonation to form the method of combining molecular and quantum mechanics has been
iron-oxo intermediate (compound I), and then electron transfer for proposed, which is designed to predict the substrate binding and
catalysis (3). It is difficult to simulate this whole metabolic process rate-limiting steps of catalysis with cytochrome P450s. The primary
by computers accurately, but with the availability of human P450 goal of this review is to present these computational approaches
structures gradually, structure-based modeling has been developed on drug metabolism prediction. We first review drug-induced toxici-
to facilitate drug design. ties and mechanisms, then specific prediction examples for regiose-
lectivity, stereoselectivity, inhibition, induction, and mechanism-
In silico drug metabolism prediction methods were ligand-based such based inhibition, as well as the application in hit-to-lead drug
as building pharmacophore and QSAR quantitative structure–activity discovery.
relationship (QSAR) modeling before structure-based drug design
emerges (4). QSAR modeling still plays a big role in pharmaceutical
industry because of the significant growth of high-throughput screen- Drug Metabolite-induced Toxicities
ing data. In QSAR, neural networks, classification methods such as
recursive partitioning, decision trees, and support vector machines The attrition rate of drug candidates as a result of toxicities remains
are constructed with topological, structural and electronic descrip- high. Toxicity is still a major safety concern for drug withdrawal
tors. ADMET properties are thus predicted on the basis of these de- (such as drugs suprofen, terfenadine, rofecoxib, mibefradil, troglitaz-
scriptors such as lipophilicity and solubility. Usually QSAR predictions one, nefazodone, cisapride, remoxipride, tolcapone, and tienilic acid),
work particularly well with structurally similar compounds whereas the 'black box warning' (such as the most recent GlaxoSmithKline's
molecules from other regions of the chemical space can cause out- anti-diabetic drug rosiglitazone), and the discontinuation of clinical
liers. Furthermore, QSAR is limited by the quality of the available trials (such as Pfizer's hypercholesterolemia drug torcetrapib with-
experimental data. QSAR can sometimes provide hints about the drawal from Phase III) (7,8). An analysis of the first-in-human regis-
active site if according descriptors appear in the regression equation tration for ten big pharma companies demonstrated only a 10%
such as steric constraints and lipophilicity. Pharmacophore models total success rate leading to the final FDA approval (8). The failure
are constructed to overlay structures of all ligands also to simulate rate becomes even higher when all drug candidates in preclinical
the spatial and chemical properties of the binding site (5). Pharmaco- research are included in the statistics. In this regard, it becomes
phore models, structure-based docking and molecular dynamics are important to define strategies for drug safety assessment. The tradi-
all mechanism-based approaches. Recently, the prediction power has tional drug safety testing approaches include in vivo animal models
been improved in building 3D-QSAR pharmacophore models with and in vitro cell-based assays, and most recently in silico assess-
spatial atomic descriptors in consideration. These descriptors include ment is also introduced. QSAR-based expert systems are mainly
molecular interaction fields, electronic properties, and shapes of used in early drug discovery to predict toxicological endpoints includ-
active sites. The purpose of in silico ADMET prediction is that even- ing carcinogenicity, teratogenicity, mutagenicity, immunotoxicity, neu-
tually we should be able to predict drug absorption based on the rotoxicity, developmental toxicity, respiratory sensitization, and skin
combination of lipophilicity, solubility, permeability, metabolism, and irritation (9), but the quality of experimental datasets (10) is still a
transporter activity; to predict drug distribution based on the volume big challenge to develop a predictive model.
of distribution, plasma-protein binding and blood–brain barrier pene-
tration; and to predict clearance based on renal clearance, hepatic Drug metabolism continues to be a prominent contributor in drug
metabolism, biliary excretion, and gut stability. safety (11). To understand the molecular and cellular mechanisms
responsible for drug- and metabolite-induced toxicity will be critical
Structure-based methods have been an integral part of drug design. to reduce the attrition rate. In-target and off-target toxicities are
These mechanistic approaches are encroaching on drug metabolism two different mechanisms (12), in detail, the toxicity can be catego-
studies such as to predict the binding modes of substrates, conforma- rized into 4 types (13): type-1 is related to pharmacological path-
tion change of enzymes, catalysis, and their consequences on physio- ways either on primary target or non-primary target; type-2 refers
logical system. For example, docking method and local binding energy to the idiosyncratic toxicities such as drug-induced allergies; type-3
calculation were used to evaluate the relationship between the is due to chemical activation such as drug metabolism and the
metabolism and carcinogenicity (6). Indeed, structure-based modeling metabolites adducting with macromolecules; and type-4 is the
by combining molecular and quantum mechanics enables us to predict chronic toxicity such as carcinogenesis and teratogenesis. Mitochon-
substrate affinity, lability and metabolic pathways. In practice, mass drial dysfunction is also a mechanism. It is very possible that the
spectrometry, nuclear magnetic resonance spectroscopy, proteomics, withdrawal anti-diabetic drug troglitazone and cholesterol lowering
metabolomics, and other advanced bioanalytical techniques have drug cerivastatin caused mitochondrial dysfunction (14). Long-term
been improved significantly, which can help us validate these in silico mechanisms may come from the impaired mitochondrial replication
predictions quickly. There are still challenges using these tools such and protein synthesis, and short-term from the inactivation of the
as the difficulties in calculating free energies in the thermodynamic electron transport complexes in the inner membrane of mitochon-
cycles, enthalpy and entropy differences and solvation issues (7), but dria through the covalent binding with reactive metabolites (15).
they already saved us time and recourses discovering new drugs fas-
ter and more efficiently, and thus provide us more opportunities in Although in silico predictions of drug metabolism seem unlikely to
drug discovery. We believe, with breakthroughs, in silico methods will replace the well-established in vitro and in vivo methods in the
play an increasingly important role in drug discovery. near future, these computational tools help early drug design

4 Chem Biol Drug Des 2010; 75: 3–17


Structure-based Drug Metabolism Predictions

greatly. One potential toxicological factor the can be eliminated in or destruct the prosthetic heme group of P450s. Some
early stage is the mechanism-based toxicity. Xenobiotics may be known mechanism-based inactivators are acetylenes (such as
directly toxic such as nicotine, whereas the toxicity of drugs is lar- 2-ethynylnaphatalene, 9-ethynylphenathrene, 7-ethynylcoumarin and
gely due to their metabolites, either highly reactive electrophiles or 5-phenyl-1-pentyne), organosulfur compounds (such as disulfiram,
free radicals. Various mechanisms were proposed such as reactive cimetidine, tienilic acid, ticlopidine, and thiazolidinediones), arylam-
metabolite formation and mechanism-based inactivation. Primarily, ines (such as 1-aminobenzotriazole), cyclic tertiary amines (such as
metabolic activation process depends on physicochemical properties phencyclidine), and furanocoumarins (such as bergamottin, 8-gera-
of compounds, the interactions between compounds and active site nyloxypsoralen, and 8-methoxypsoralen). Others include tamoxifen
amino acid residues of drug-metabolizing enzymes, and catalysis. In and raloxifene, both of which are the selective estrogen receptor
addition, transporters such as P-glycoprotein or organic ion-trans- modulators, glabridin (an isoflavan) and anticancer drug N,N ¢,N ¢¢-
porting polypeptides can transport drug metabolites to the off-site triethylenethiophosphoramide (19). In addition, the pneumotoxin
target for toxicity. Moreover, drug toxicity can be idiosyncratic or 3-methylindole was found to be bioactivated by CYP2F1 in human
caused by genetic diversity. Polymorphisms in P450, N-acetyltrans- lung to form electrophilic methylene imine reactive intermediates
ferase and transporter genes impact individual responses to drugs that cause mechanism-based inactivation. Studies showed that 3-
because of the population genetic difference of these enzymes. The methylindole structural analogues are also potential mechanism-
ultimate objective of drug metabolism prediction is to evaluate the based inactivators (Figure 1). For example, zafirlukast (a leukotriene
toxicity, but currently it focuses on the selection and optimization of receptor antagonist), MK-0524 (a prostaglandin D2 receptor antago-
drug candidates for the advancement of drug discovery pipelines, nist), and SPD-304 (a tumor necrosis factor-a inhibitor) were found
also with the goal of reducing recourses and recreating more reli- to form methylene imine reactive intermediates, and the mecha-
able candidates (11). nism-based inactivation of CYP3A4 was observed in zafirlukast,
SPD-304 and tadalafil.
One of the key roles of preclinical drug metabolism assessment is
to identify potential liabilities in new chemical series as early as
possible. For example, the identification of reactive metabolites Drug Metabolism Prediction: insights
offers promise for decreasing the high rate of attrition, because from Docking, Molecular Dynamics and
reactive metabolites may bind covalently to the locus of their for- Quantum Chemical Calculations
mation or at a distant site to inactivate target proteins, change the
biochemistry, signal transduction, or even trigger immunological The success of structure-based drug design is well known, by which
response. Various forms of reactive metabolites are identified and approximately 50 compounds have been introduced to the clinical
used for the in vitro reactive metabolite screening. These moieties trials and ⁄ or approved until 2004 (20). To date, many more drug
include quinones, quinone methides, quinone imines, imine met- candidates have been discovered with the help of advanced compu-
hides, epoxides, anilines and derivatives, furans, benzylamines, thi- tational techniques for the reason that in silico tools have been
ophenes, glitazones, thioureas, alkenes, and alkynes (16). However, improved considerably and more widely used by pharmaceutical
some reactive metabolites still escape from the detection even with companies in the last several years. For example, using AutoDock
the most advanced bioanalytical instruments, and also there is a combined with relaxed complex method has led to the approval of
gap between reactive metabolite formation and its toxicological the first HIV integrase inhibitor raltegravir (Isentress) by Merck in
consequences (15). A solution especially in design is to incorporate 2007 (21). Furthermore, the advantage of structure-based drug
in silico tools as reviewed here. metabolism prediction not only predicts the site of metabolism
(metabolic liability) but also elucidates important molecular interac-
Several P450-related pharmacological processes can cause drug– tions between substrates and active site residues of drug-metabo-
drug interactions including induction, inhibition, and mechanism- lizing enzymes, which produce invaluable information for chemical
based inactivation. Induction refers to a process that causes the design and redesign to enhance pharmacokinetic profiles and
increase of P450 enzyme expression, an adaptive response to xeno- reduce toxicities.
biotics or certain drugs by human body, which is mediated by
nuclear hormone receptors such as constitutive androstane receptor To perform structure-based drug metabolism prediction, experimen-
(CAR), pregnane X-receptor (PXR), peroxisome proliferator activated tal determined (such as using X-ray crystallography) structures of
receptor (PPAR) and glucocorticoid receptor (GR). Induction-caused P450s or other drug-metabolizing enzymes are required. If no corre-
drug–drug interactions can reduce the efficacy and ⁄ or increase the sponding X-ray crystal structures are available, appropriate homol-
toxicity of co-administrated drugs (17). Inhibition occurs during sub- ogy models are required, but need to be carefully evaluated during
strate or molecular oxygen binding and mechanistically, P450 inhibi- the construction process based on enzyme kinetics data and ⁄ or
tors are categorized as reversible or irreversible ones that are site-directed mutagenesis results. The first mammalian P450 struc-
measured by analyzing the inhibition of typical substrates of P450s ture, rabbit microsomal CYP2C5 was determined in 2003 by Dr. Eric
by inhibitors such as using Michaelis–Menten kinetics to determine Johnson's group in the Scripps Research Institute (22) and subse-
a reversible inhibition (18). Mechanism-based inactivation is also quently, the crystal structures of major human forms become avail-
referred as 'suicide' or 'time-dependent' inactivation. It is a process able including CYP1A2, 2A6, 2A13, 2C8, 2C9, 2D6, 2E1, and 3A4
that reactive intermediates inactivate the enzyme without leaving (Table 1). These structures yielded invaluable molecular information
the active site of P450s (19). Mechanism-based inactivators can of P450 enzymes, and also revealed the coming of fast crystalliza-
either covalently bind to the amino acid residues of apoprotein tion of drug-metabolizing enzymes.

Chem Biol Drug Des 2010; 75: 3–17 5


Sun and Scott

O
O HN S
P450 O
O
H
N N O N
H H
3-methylindole 3-methyleneindolenine O
N

Zafirlukast (Accolate)

O O
N N
N
F O

O N
OH O
N
N
O S O N
H
Cl F F O Figure 1: 3-Methylindole and
O its structural analogues are poten-
F
MK-0524 tial mechanism-based inactivators
SPD-304 Tadalafil (Cialis) of cytochrome P450s (86-91).

Table 1: Available structures of human cytochrome P450s for public access, as determined by X-ray crystallography (37,77–85)

CYPs PDB CODE Resolution () Notes

CYP1A2 2HI4 1.95 Bound with Alpha-Naphthoflavone


CYP2A6 1Z10, 1Z11 1.90, 2.05 Bound with Coumarin and Methoxsalen
2FDU, 2FDV, 2FDW, 2FDY 1.65 – 2.05 Bound with N,N-Dimethyl(5-(pyridin-3-yl)furan-2-yl)methanamine,
N-Methyl(5-(pyridin-3-yl)furan-2-yl)methanamine, (5-(Pyridin-3- yl)furan-2-yl)methanamine,
and Adrithiol
2PG6, 2PG7, 2PG5 1.95 – 2.80 2A6 Mutants L240C ⁄ N297Q, N297Q ⁄ I300V and N297Q
CYP2A13 2P85 2.35 Bound with Indole
CYP2C8 1PQ2 2.70 No Substrate Bound
2NNI, 2NNJ, 2NNH 2.28 – 2.80 Bound with Montelukast, Felodipine, and 9-Cis-Retinoic Acid
CYP2C9 1OG2, 1OG5 2.55, 2.60 Bound with Warfarin
1R9O 2.00 Bound with Flurbiprofen
CYP2D6 2F9Q 3.00 No Substrate Bound
CYP2E1 3E4E, 3E6I 2.60, 2.20 Bound with 4-Methylpyrazole and Indazole
CYP3A4 1TQN 2.05 No Substrate Bound
1W0E, 1W0F, 1W0G 2.65 – 2.80 No Substrate Bound, and Bound with Metyrapone and Progesterone
2J0D, 2V0M 2.75, 3.80 Bound with Erythromycin and Ketoconazole

Modeling drug metabolism requires predicting substrate binding to by other computational chemists and drug metabolism scientists to
the active sites and adapting computational approaches to account predict drug metabolism.
for the catalysis. There is no single fully integrated structure-based
computational tool that is able to simulate all the possible pro- One of the goals of drug metabolism prediction is to find the low-
cesses of drug metabolism. However, a combined computational est-energy binding structure complex containing drug-metabolizing
strategy was designed to predict the regioselectivity of macrolide enzyme and substrate, which in many cases may account for one of
immunosuppressants sirolimus and everolimus: their hydroxylation the most possible orientations for catalysis. Other goals include the
and O-dealkylation reactions catalyzed by CYP3A4 (23). In address- prediction of the rates of metabolism based on drug molecule
ing the molecular mechanisms of sirolimus and everolimus bioacti- activation mechanisms. The success of prediction requires the
vation, the proposed prediction method consisted of three separate development of an accurate energy potential function in the above-
steps. First, the substrates were docked into the active site of mentioned sequential calculations. Simplification of the process is
CYP3A4 with energetically favored binding poses selected. Second, invariably necessary in considering the computing time; however, a
the chosen docking poses were placed into AMBER force field for single one-step modeling is not adequate from a mechanistic view,
molecular dynamic simulation with the substrate binding orientation but sometimes still produces relative or comparable energy informa-
and the distance to the heme iron calculated, respectively. Third, tion that is sufficient for drug design purpose especially for the
the hydrogen atom abstraction of selected fragments of both sub- compounds within the same chemical series. For example, docking
strates was calculated by quantum chemistry method (Figure 2). was successfully used to predict CYP2C9 metabolic activation of
This prediction strategy has been gradually accepted and adopted the COX-2 inhibitor celecoxib and its 13 analogues (26) Figure 3

6 Chem Biol Drug Des 2010; 75: 3–17


Structure-based Drug Metabolism Predictions

Figure 2: Energy change from


substrate binding to product forma-
tion in cytochrome P450-catalyzed
drug metabolism. A combination of
docking, molecular dynamics and
quantum chemical calculation is
proposed for in silico prediction of
drug metabolism.

demonstrated the predicted two binding pose clusters of midazolam ing comparing to stochastic methods, which apply random changes
with docking method applied alone, and Figure 4 demonstrated the of substrates and then evaluate a predefined probability function.
quantum chemistry-based calculation for the activation energy of Monte-Carlo and genetic algorithms are classic stochastic algo-
testosterone carbon hydrogens indicating the 6-b position is the rithms. Simulation method like molecular dynamics accommodates
most likely CYP bioactivation place. ligands in local minima of the energy surface that will be discussed
later.
Docking method which involves the prediction of substrate confor-
mation and orientation within the active site of enzymes (27) is A scoring function is required to rank the favored ligand conforma-
composed of a tandem prediction process: posing and ranking. Pos- tions, such as using force field-, empirical-, or knowledge-based
ing searches energetically favored binding orientations of substrates methodologies. Fundamentally, these scoring functions make various
that is often determined by a certain computational algorithm. On assumptions; therefore, the evaluation process is simplified and can
the contrary, ranking scores various poses derived from posing with only roughly simulate the biological process. Researchers have tried
different interaction energy calculation functions such as electro- to eliminate assumptions and combine as many as energy parame-
static, hydrogen bonding, van der Waals, entropy, and explicit solva- ters to make it more reliable. In force field-based scoring functions,
tion (24). Searching protocols may be systematic, stochastic or a energetic terms are derived from classic mechanics such as
simulation. A systematic searching usually incrementally grows the AMBER95 or CHARMm22, and the atomic interactions between pro-
substrate within the active site of CYPs, in other words, rigid cores tein and ligands are simulated based on known factors controlling
of substrates are docked in first and then the flexible parts, based the molecular recognition. Both enzyme–substrate interaction energy
on libraries of pre-generated conformations. They are less challeng- and substrate internal energy are calculated, but the internal pro-

A B

Figure 3: Two major energeti-


cally favored binding clusters of
midazolam in the active site of
CYP3A4 from molecular docking
prediction (A) 1¢-hydroxylation Helix l Helix I
position (B) 4-hydoxylation position.
CYP3A4 is shown in a cartoon
format (transparency gray), and
midazolam (yellow) and heme (pink)
are shown with colored sticks: Heme Heme
nitrogen, blue; oxygen, red;
chlorine, green; fluorine, cyan.

Chem Biol Drug Des 2010; 75: 3–17 7


Sun and Scott

A B

Figure 4: Dynamic analysis of active site residues of CYP3A4 crystal structures (A) active site of CYP3A4 (B) overlapped selected active
site residues from various crystal structures with different substrates bound.

tein energy are generally excluded. For example, the van der Waals substrate binding. Ito et al. simulated the motion of active site resi-
energy is given by a Lennard–Jones potential function and electro- dues of CYP2D6 in the process of propranolol binding, and found a
static energy is calculated using Coulombic formulation with a dis- significant movement of F-G loop and H-I loop, with several resi-
tance-dependent dielectric function. Empirical scoring functions dues identified as key sites such as Phe120, Glu216, Asp301,
depend on the molecular datasets used for regression analysis and Phe483, Phe219, and Glu222 (25). Their theoretical analysis was
fitting that are obtained from experimentally determined binding indeed correlated with site-directed mutagenesis results. In another
energies. It is usually simpler than force field-based scoring func- study, molecular dynamic analysis was placed in-line with neural
tion but does not provide mechanistic information for the purpose network modeling to predict the binding of 82 compounds to
of molecular modification. Knowledge-based scoring is designed to CYP2D6, which demonstrated well-defined predictive behavior (28).
reproduce experimental structures rather than binding energies. The Another molecular dynamic analysis with spectroscopic study
advantage is its computational simplicity, but the function is only showed the large-scale structural transition of CYP2B4 when the
based on limited sets of protein–ligand complex structures and also substrate 4-(4-chlorophenyl)imidazole binds. Interestingly, Muralidha-
limited by the atom-types being used to derive the corresponding ra and coworkers proposed a theory of 'open from' and 'closed
potential of mean force. form' of P450 enzymes (29), which states that the structural varia-
tions are mainly located in the B' helix and F-G region.
Our perception of the dynamic change of P450 active site grows
with the expanding X-ray crystal structures, and also from site- A continuous time-course structural determination would be an
directed mutagenesis and molecular dynamic analysis. Molecular ideal method to visualize the dynamic change of P450 active site
dynamics simulate the flexibility of CYP active site residues in a residues, but still, not possible in the current experimental settings.
time scale, generally in the order of nanoseconds. Depending on However, the co-crystallization with different substrate bound has
the size of the binding pocket, multiple binding poses are possible created a small database illustrating the dynamic change of P450
to give rise to different metabolic products. This might help explain enzymes especially the dynamic orientations of side chains. Over-
why regioselectivity or stereoselectivity often occurs for P450 sub- lapping several CYP3A4 structures, we analyzed the dynamic
strates. With molecular dynamics combined, docking methods can change of active site residues. Primary interests have centered on
also be enhanced. Although our understanding of P450-catalyzed different substrate binding, which showed some residues are extre-
drug metabolism and our ability to model the process are expand- mely flexible such as the obvious movement of Arg212, Ser312,
ing, the prediction of active site residue dynamic change in a time Phe304, and Glu308 (Figure 5). The interplay between these resi-
scale remains a challenge. The stability of the substrate-P450 com- dues makes it difficult, if not impossible, to fully grasp the protein
plex is likely the result of attractive van de Waals interactions, sol- dynamics when individual substrate binds. But the information is
vation free energy, hydrogen bonds as well as hydrophobic and important for us to determine docking parameters such as to set
polar interactions. In light of these interactions, molecular dynamic part of protein flexible and the rest in rigid to increase the predic-
approaches are often applied to identify specific residues that direct tion accuracy and reduce computing time.

8 Chem Biol Drug Des 2010; 75: 3–17


Structure-based Drug Metabolism Predictions

OH ation and epoxidation reaction pathway ratios and they


(i) concluded both electrostatic and hydrogen bonding interactions
(j) are important for the difference, especially for propene, the
(h) penta-radical and ⁄ or tri-radical determine the mechanisms of
(k) (g)
hydrogenation and epoxidation. For compound I, the Fe–S bond
(m) length changes when the substrate binds, which can be calculated
(n) (l) by QM ⁄ MM methods, for example, it was found the Fe–S bond
(e) (f) of CYP3A4 compound I tends to elongate (34). Moreover, the
(o) parameters calculated by quantum mechanics were tested as
(d)
theoretical descriptors to build QSAR models such as molecular
orbital derived descriptors for flavone substrates binding to CYP1A
(c) (35).
O
(a) (b)
Water molecules often play an essential role in drug–enzyme
Position of H ab initio Position of H ab initio interactions: the displacement of water molecules from binding
Abstration (kcal/mol) Abstration (kcal/mol) site is one principal source of binding free energy (24). Water
molecules were found in the binding pockets of drug-metabolizing
a 116.23 i 107.33
enzymes, which can make favorable interactions with either
b 83.54 j 101.98
c 102.12 k 98.01
ligands or protein active site residues. Therefore, active-site water
d 95.52 l 94.62 molecules are often included in computational methods as one of
e 95.05 m 106.31 the strategies to predict drug metabolism. But current efforts in
f 102.90 n 102.49 the simulation of these water molecules mainly focus on the
g 103.05 o 95.85 molecular dynamics of the 'on' and 'off' states to unravel their
h 96.48 significant roles in substrate binding as well as their effects on
docking and virtual screening accuracy. In some docking protocols
Figure 5: Hydrogen atom abstraction of testosterone calculated water molecules are allowed to rotate around their three principal
by density functional theory. The DFT energy shows 6b position axes. With this consideration, a study on the prediction of 65
hydrogen is the most energetically favored for abstraction. CYP2D6 substrates binding and their sites of metabolism demon-
strated that water molecules are located in the energetically
favored locations. Applying this strategy, the accuracy of docking
Quantum chemical calculation is a major tool for predicting P450 algorithms was significantly enhanced (36). Thus, an improved
catalysis. From the calculated energy barrier value, we can tell drug metabolism prediction strategy could plausibly include water
the absolute or relative oxidation potential in xenobiotic bioactiva- molecules to the settings of simulation. Not all water molecules
tion. The ab initio calculations using Hartree–Fock or density func- are resolved in crystal structures of cytochrome P450s, but
tional theory are often used to calculate the reactivity of researchers have identified some of these structural water mole-
substrates. For aliphatic hydroxylation and N,O-dealkylation reac- cules recently (Figure 6). For example, an ordered water (wat733)
tions, the hydrogen atom abstraction to form a radical is the rate- was found in the active site of CYP1A2, which is located in an
limiting step for catalysis. However, the mechanism of aromatic appropriate distance range to form hydrogen bonds with the car-
hydroxylation is still unclear. The arene epoxides are generally bonyl group of the inhibitor a-naphthoflavone (2.92 ) and also
accepted intermediates for the next-step hydrogen migration from the carbonyl group of Gly316 (2.84 ) of helix I (83). Several
hydroxylation site to the adjacent carbon ('NIH shift') (3). Except water molecules were identified to be important factors for
the activation energy, the orientation of the aromatic ring is also CYP2C9 substrate binding: one (wat600) is located between the
important, which is orientated for either 'side-on' addition (ring substrate flurbiprofen and the heme iron at a distance of <3.0 
vertical to the heme porphyrin) or 'face-on' addition (ring parallel that accounts for the hydrogen bond with the backbone carbonyl
to the heme porphyrin). To ensure the active site environment group of Ala297, one (wat819) is located at the distortion region
parameters included to determine the orientation, heme-oxygen of helix I, and another one (wat842) is between flurbiprofen and
species and other interacting active site residues are needed to the turn region of the substrate recognition site-6 (85). In CYP3A4,
be incorporated into quantum chemical calculation. The hybrid a cluster of water molecules was also observed above the heme
quantum mechanical ⁄ molecular mechanical (QM ⁄ MM) method is porphyrin (37), which can be replaced totally or partially that
ideal to do the calculation (30). For example, a hybrid QM ⁄ MM depends on the size of substrates.
method was used to simulate the benzene hydroxylation by
CYP2C9 by Bathelt et al. and the results suggested a roughly Regioselectivity
equal 'side-on' and 'face-on' pathways, which posed a theoreti- Many drug metabolism prediction efforts have focused on the
cal challenge because early opinions differed because the 'face- prediction of P450 regioselectivity or metabolic pathways (38,39).
on' addition was considered as the dominant mechanism for The derived information is important for modifying metabolic lia-
p-stacking interactions (31). Quantum chemical calculations also bility in hit-to-lead. Camitro's metabolism models were widely
shed light on the regioselectivity studies of cyclohexene and pro- used in early years to predict enzyme-substrate binding affinities
pene. Cohen et al. (32) and Hirao et al. (33) studied the hydroxyl- and metabolic sites using empirical and quantum chemical

Chem Biol Drug Des 2010; 75: 3–17 9


Sun and Scott

A B C

wat733
wat842 Helix I R212

ANF G316 FBP wat819

Helix I A297 Helix I


wat600
Heme Heme Heme

Figure 6: Essential water molecules in the active sites of cytochrome P450s. (A) CYP1A2 and inhibitor a-naphthoflavone (ANF) (B) CYP2C9
and substrate flurbiprofen FBP, and (C) CYP3A4. Water molecules are depicted as red spheres.

approaches (40). By definition, the regioselectivity of P450-cata- Stereoselectivity


lyzed reactions is the preference of one or several substrate Structure-based drug metabolism prediction is also a good option to
sites of metabolism over others. Zhang et al. investigated the elucidate the metabolic differences in enantiomers. Indeed, many
metabolism of the C3¢ substituents of a series of taxane ana- drugs are synthesized for one enantiomer only to avoid the stere-
logues by CYP2C8 and 3A4 (41) using docking approaches, and oselectivity by drug-metabolizing enzymes (Figure 7). The difference
found the binding of molecules were re-oriented from in favor of of metabolic profiles of phenylahistin enantiomers was evaluated
3¢-p-hydroxylation to 4¢¢-hydroxylation reaction when replacing the using the energy calculation of docked enantiomers by molecular
C3¢ aromatic ring (paclitaxel) with an aliphatic chain (cephalo- mechanics and Poisson–Boltzmann surface area approaches. It dem-
mannine). In addition, several combined models were used to onstrated that (-)-phenylahistin is 1.5–8 times less metabolized than
predict regioselectivity. In one model, docking was combined with (+)-phenylahistin by CYP3A4 (46). Docking of (+)-phenylahistin in the
an empirical activation energy model as well as reaction active site of CYP3A4 showed that both isoprenyl group and nitro-
enthalpy and ionization potential descriptors to predict the sub- gen atom at the imidazolyl ring can interact with the heme iron for
strate reactivity (42). The authors evaluated 72 well-known the hydroxylation reaction; however, for (-)-phenylahistin, the predic-
CYP3A4 substrates and produced a 76% success rate. In another tion results differed dramatically (isoprenyl group is far from the
model, docking and ligand-based in silico methods were applied heme and the lone pair of the imidazolyl nitrogen does not point
to predict metabolism sites of 70 known CYP2C9 substrates suc- toward the heme iron), because they possess very similar physico-
cessfully (43). These studies also indicated that the regioselectivi- chemical properties. The difference of enantiomer metabolites from
ty is probably controlled by one or several active site residues. non-steric compounds can also cause enantioselectivity. Bikadi and
For example, Lafite et al. investigated the hydroxylation of terfen- Hazai studied CYP2C-catalyzed O-demethylation of methoxychlor
adine derivatives by CYP2J2 (44) and found the presence of (47), and demonstrated that both CYP2C9 and 2C19 favored the for-
bulky residues Ile127, Ala311, Ile375, Ile376, and Val380 above mation of S-mono-OH-methoxychlor, no enantioselectivity found for
the heme iron to form a hydrophobic core, and a hydrogen bond CYP2C3, but CYP2C5 favored the formation of R-mono-OH-methoxy-
formed between the keto groups of terfenadine derivatives and chlor. The chiral preference was further studied by molecular
Arg117. These results demonstrated that the control of substrate dynamics, and the authors found it was caused by the variance and
accessibility can direct the regioselectivity, although improved cal- flexibility of active site residues. In addition, deoxypodophyllotoxin
culations are needed to further test the hypothesis. In another was found to be stereoselectively bioactivated by CYP3A4 to epip-
study, the bioactivation of tobacco procarcinogen 4-(methylnitrosa- odophyllotoxin but not to podophyllotoxin (48). The docking and
mino)-1-(3-pyridyl)-1-butanone (NNK) by CYP2A6 and 2A13 was Monte-Carlo-based refinement revealed that the epipodophyllotoxin
compared by docking approaches. It was found that both Met365 formation is in a more appropriate orientation, or specifically, the
and Ile366 of CYP2A13 provided more space for NNK positioning hydroxylation is mediated by the hydrogen bonding between de-
in the active site for a-methyl hydroxylation and a-methylene oxypodophyllotoxin and active site residues Arg212, Glu374 and
hydroxylation reactions but not in CYP2A6 (84), which explains Arg105.
the regioselectivity difference of these two enzymes. The regiose-
lectivity was also observed using enzyme mutants. Keizers et al.
found that the O-demethylation of 3,4-methylenedioxy-N-alkylam- Reactive metabolites
phetamines was dominant in wild type CYP2D6 but N-demethyla- The mechanism of reactive metabolite formation by CYPs has
tion and N-hydroxylation reactions occurred instead in its F120A been extensively studied computationally, mainly by quantum
mutants (45). chemical calculations. With different substituents added to lead

10 Chem Biol Drug Des 2010; 75: 3–17


Structure-based Drug Metabolism Predictions

O O

NH N NH N
NH NH
HN HN

O O

(–)-phenylahistin (+)-phenylahistin

H CCl3 H CCl3 H CCl3


(R) (S)

MeO OMe HO OMe MeO OH

Methoxychlor R-mono-OH-methoxychlor S-mono-OH-methoxychlor

H H HO H H OH
O O O
O O O
O O O

MeO OMe MeO OMe MeO OMe


OMe OMe OMe

Deoxypodophyllotoxin Podophyllotoxin Epipodophyllotoxin

Figure 7: Enantioselectivity and stereoselectivity of several known compounds.

compounds, the relative free energy change for activation is calcu- examples, we know by calculating the oxidation potential of reac-
lated to guide lead optimization. Docking and molecular dynamics tive metabolite formation, we can modify molecules to enhance
are often combined with quantum chemical calculations to drug metabolic profiles or reduce toxicity issues.
increase the prediction power. Many successful applications have
been documented. For example, the mechanism of phenacetin bio-
activation to form reactive metabolite quinone imine was studied Inhibition
with ab initio energy and spin distribution calculation, to compare The mechanism of inhibition was studied by docking and molecular
the hydrogen atom abstraction at the a-methylene carbon atom dynamic approaches. Quinidine is a potent competitive inhibitor of
and electron abstraction from the nitrogen atom at the acetylami- CYP2D6 but not a substrate. McLaughlin et al. investigated the
no side (49). A similar study on the metabolic activation of acet- binding modes of quinidine together with site-directed mutagenesis
aminophen to quinone imine by Loew and Goldblum predicted the experiments (55). They found in CYP2D6's F120A and E216F
hydrogen atom abstraction from the phenol group is more proba- mutants, quinidine indeed became a substrate, indicating the impor-
ble than from the amide nitrogen (50), which was also confirmed tance of these two residues in controlling substrate ⁄ inhibitor
by in vitro studies (51). In another study, the bioactivation of car- switching. Both substrates and inhibitors bind to enzymes in ener-
bamazepine by CYP3A4 showed that the reactive metabolite10,11- getically favored manner, but for inhibitors they are not in the
epoxide formation is more probable than the hydroxylation of the appropriate orientation for catalysis or bind to no-catalysis sites
aromatic ring calculated by density functional theory (52). With such as the ligand entrance region of P450s. Structurally diverse
these mechanistic results, we can design new compounds with compounds were evaluated by several research groups for their
reduced oxidation potential and thus eliminate the reactive metab- inhibition of CYP3A4 using docking methods and also considering
olite formation. For example, the bioactivation energy of the anti- ligand conformation, orientation, stereoisomer, and protonation
psychotic drug remoxipride to from a hydroquinone metabolite and state. The results demonstrated that docking approach is indeed a
further to an electrophilic quinonone was calculated by hybrid good option to screen a library for potential inhibition and drug-
density functional theory (53). The authors then redesigned drug interactions (56,57). In addition, co-operativity properties that
remoxipride by adding amide and methoxy groups. In addition, the may determine the inhibition showed the homotropic and ⁄ or hetero-
ab initio calculation was used to compare the oxidation potential tropic co-operativity of 1-alkoxy-4-nitrobenzenes and 1,4-phenylene
of nefazodone and buspirone to form reactive intermediate diisocyanide in CYP1A2 (58). These calculations were used to
quinone imines (54). It demonstrated buspirone is much more diffi- design new compounds with modified inhibition. For example, sub-
cult to undergo two-electron oxidation than nefazodone, and thus stitution of terfenadine benzylic alcohol group to ketone demon-
less reactive metabolite is expected. From these drug design strated 10-fold inhibition increase with CYP2J2 (59).

Chem Biol Drug Des 2010; 75: 3–17 11


Sun and Scott

Application in hit-to-lead Drug Discovery

It is important to maintain both quality and quantity of early lead


series to discover a new therapeutic agent in a reasonable time
frame (68). In hit-to-lead drug discovery, high-throughput screening
requires a suitable assay to be developed; as attractive alternatives
fragment-based and docking methods are especially useful for
focused screening. The application of better hit-to-lead approaches
can help us achieve the goal of fast and efficient drug discovery.
Structure-based drug metabolism prediction has the potential to
offer new drug design approaches based on metabolic profiles for
Figure 8: Structure of pregnane X receptor. The monomers are hit-to-lead discovery, which should facilitate our endeavor on
in yellow and green, ligands in hot pink, and steroid receptor coac- expanding chemical space and searching high-quality drug candi-
tivator 1 (SRC-1) in red on the back. dates. Drug metabolism prediction using docking, molecular dynamic
and quantum chemical methods provided a powerful insight into
the application of these in silico tools in drug discovery. We have
Induction found great success in predicting drug metabolism using the similar
CYP3A4 is induced by the activation of PXR, and its structure has methods in our drug discovery projects. Indeed, we need a cost-
been determined recently (60). Docking showed the addition of effective and high-efficient hit-to-lead process to expand the chemi-
polar groups to one end of the activator attenuated the activation cal space and find the best lead compounds with minimum
because the new molecules destabilize the hydrophobic interactions resources, so the in silico drug metabolism approaches described in
in the binding pocket (Figure 8) (61). With more structures of key this review can be a great benefit for the optimization of chemical
induction regulators determined, we expect to predict the induction space with the purpose of exploring best-in-class medicines
using the similar in silico tools above. (Figure 9).

Substantial attention has been given to use docking-related molecu-


Mechanism-based inactivation lar mechanical approaches to make molecules more potent. The
Heme porphyrin ring and some active site residues are primary tar- modification such as adding or changing functional groups primarily
gets for the binding of reactive metabolites that can consequently focuses on the increase of ligand–protein interactions, which gener-
cause the mechanism-based inactivation before their release from ally lowers binding energies or increases the potency. For examples,
P450 active site (19). Recent structural modeling and proteomic a docking and scoring method was applied to explore potent b-sec-
studies have shed light on possible inactivation mechanisms for retase inhibitors (69). More importantly, to develop a safer com-
either heme porphyrin or apoprotein residue alkylation with drug pound means more chance to move a drug candidate into the
molecules. Raloxifene is a mechanism-based inhibitor of CYP3A4, market. For example, the cyclooxygenase-2 inhibitor celecoxib is
which is bioactivated to either diquinone methide or epoxide reac- less potent than rofecoxib, but much safer according to meta-analy-
tive metabolites. Several research teams have identified these sis postmarket studies, which was the main reason it is still in the
adduction sites such as Cys239 and Tyr75 for quenching reactive market but the rest of same-family drugs such as rofecoxib got
intermediates (62–64), in particular, it was postulated that the withdrawal for increased cardiovascular toxicity. As discussed
nucleophilic OH group of Tyr75 or the sulfur group of Cys239 can above, the current trend of hit-to-lead and lead optimization is the
adduct with reactive metabolite directly. In addition, CYP3A4 and early evaluation of ADMET properties such as the optimization of
CYP3A5 were compared to elucidate the mechanism of raloxifene clearance profiles and elimination of P450 inhibition (70). The trade-
bioactivation. It was found that the mechanism-based inactivation off for improved ADMET properties is sometimes decrease the
was only present in CYP3A4 but not in CYP3A5 because Cys239 is potency, and vice versa. Therefore, to find the overlap region of AD-
replaced by a serine residue in CYP3A5 (64). Kang et al. showed MET and potency properties early trade-off is essential for reducing
that the formation of reactive 10,11-epoxide, 2,3-arene oxide and chemical liabilities and accelerating hit-to-lead process. Lead com-
iminoquinone metabolites of carbamazepine adducted with Cys239 pounds can then be optimized based on the collected pharmacologi-
for mechanism-based inactivation (65). The cysteine-specific modify- cal and ADMET properties. It was noted that the receptor binding
ing reagent iodoacetamide was also applied to test the importance sites are difficult to define for many drug targets (71), but for P450
of the cysteine residue, and the results showed that iodoacetamide enzymes they are highly conserved in a fixed region. Considering
can prevent the mechanism-based inactivation (63), and thus indi- this, the success rate for drug metabolism modeling can be higher
cated Cys239 may be one of the cysteine to quench the reactive than target protein simulation. Overall, these in silico approaches
metabolite. Moreover, the apoprotein alkylation by reactive metabo- are helpful for the identification of tighter or specific binders with
lites also depends on the structures of substrates. Docking showed better ADMET properties (72).
the furan ring of bergamottin is bioactivated to an epoxide that
inactivates CYP3A5 and CYP2B6. The furan site was observed as The strategy of blocking the metabolic labile sites of compounds
the primary site for the exposure to the oxyferryl center of heme was driven by attaching a metabolic stable group such as a fluorine
(66). Ser360 was also found to be the adduction site by the reac- atom or a trifluoromethyl group. For example, the metabolic activa-
tive metabolites of 17-a-ethynylestrodiol (67). tion of a series of celecoxib structural analogues showed that

12 Chem Biol Drug Des 2010; 75: 3–17


Structure-based Drug Metabolism Predictions

Figure 9: Schematic represen-


tation of hit-to-lead and lead opti-
mization approaches to defining
chemical space based on both drug
receptor space and ADME space.

replacing the methyl group of 4-methylphenyl moiety with a trifluo- ation. The trifluoromethylpyrimidine series were found to be pro-
romethyl group totally blocked the methyl hydroxylation by CYP2C9 line-rich tyrosine kinase-2 inhibitors but also positive in reactive
(but COX-2 inhibition rate remains the same) (73), which indicate metabolite screening. Clearly, to remove the potential toxicities
that new compounds have improved metabolic properties without was required for further development. The modification of the
losing the potency and selectivity. However, the merit of adding a carbonyl group of the 5-aminooxindole moiety to a stronger elec-
trifluoromethyl or fluorine group is unpredictable because new met- tron-withdrawing sulfone group was found to eliminate reactive
abolic lability can be introduced by switching the metabolic path- metabolite formation, which was predicted correctly by density
ways. Therefore, the blind practice of including them for every drug functional theory calculations (92). These successful hit-to-lead
design without considering substrate-protein interactions can block efforts in drug metabolic profile improvement suggest that the
our vision during the exploration of wider chemical space. Once the combination use of in silico tools at both receptor space and drug
structures of receptors or P450 enzymes are available, we can use metabolism space will be likely to turn the serendipity of drug
structure-based computational tools to expand the chemical space discovery into powerful drug design strategies.
and also achieve better pharmacokinetic profiles for safer drugs.
Several limitations of these tools should be noticed. Crystal struc-
Therefore, a promising strategy to reduce attrition rate and ture resolution, accuracy of the force field, docking algorithms and
shorten discovery cycle is to harness these in silico tools to refine quantum chemical calculation all determine the prediction power.
ADMET properties in the early stage. Indeed, many recent drug The quality of crystal structures are different because the flexible
discovery efforts have focused on drug metabolism prediction regions of some proteins are difficult to be determined. A higher-
early. In the discovery of chemokine receptor CCR5 antagonists, quality structure will generally mean higher prediction power. For
the original hits contained imidazopyridine moieties that are com- trend analysis purpose, the high-throughput docking methods have
mon CYP2D6 inhibitors (74). The authors redesigned the candi- been developed for fast and economic discovery process (71). To
dates after modeling the binding of selected compounds in the avoid these limitations various strategies have been applied.
active site of CYP2D6, which demonstrated that the pyridine nitro- Scientists at Merck found that the virtual screening success rate
gen atom is the closest atom to the heme iron. Based on the was enhanced by the combination of two docking methods (93). In
interaction information, imidazopyridine moieties were replaced by addition, free-energy perturbation calculation in the context of
the carbon analogues benzimidazoles. Consequently, the drug-drug Monte-Carlo statistical mechanic simulation was used concurrently
interaction was avoided early, and thus a high quality lead was to optimize docked compounds. For example, modification of core
discovered. The development of metabotropic glutamate receptor heterocyclic moieties of 2-anilinyl-5-benzyloxadiazole with free-
antagonists tells a similar story. The pyrazolo[3,4-d]pyrimidin-4-one energy calculation led to potent non-nucleoside inhibitor of human
series were found to be original hits as the potent and selective immunodeficiency virus reverse transcriptase (76). Moreover, de
antagonists but showed high clearance. The authors then novo ligand design such as fragment-based design and 'scaffold
improved the ADMET properties by adding electron-withdrawing hopping' are attractive approaches that ultimately allow more hit
groups to the molecule (75). In addition, rational approaches to series explored, and thus produce better chance of successful hit-
eliminating reactive metabolite formation are worthy of consider- to-lead drug discovery.

Chem Biol Drug Des 2010; 75: 3–17 13


Sun and Scott

In summary, recent drug metabolism prediction efforts reviewed 16. Kalgutkar A.S., Obach R.S., Maurer T.S. (2007) Mechanism-based
here highlighted the importance of computational methods for drug inactivation of cytochrome P450 enzymes: chemical mechanisms,
design and hit-to-lead drug discovery. Given the rapid accumulation structure-activity relationships and relationship to clinical drug-
of new structures of drug-metabolizing enzymes, it is increasingly drug interactions and idiosyncratic adverse drug reactions. Curr
clear that computational tools to predicting drug metabolism will be Drug Metab;8:407–447.
crucial in faster and more efficient drug discovery. To conclude, con- 17. Sinz M., Wallace G., Sahi J. (2008) Current industrial practices
siderable challenges remain for in silico drug metabolism prediction in assessing CYP450 enzyme induction: preclinical and clinical.
using structure-based methods, but the opportunities for in silico AAPS J;10:391–400.
drug metabolism prediction to become a mainstream component of 18. Fowler S., Zhang H. (2008) In vitro evaluation of reversible and
drug design are now apparent, and the accurate prediction now irreversible cytochrome P450 inhibition: current status on meth-
appears to be an achievable goal in the near future. odologies and their utility for predicting drug-drug interactions.
AAPS J;10:410–424.
19. Hollenberg P.F., Kent U.M., Bumpus N.N. (2008) Mechanism-
References based inactivation of human cytochromes p450s: experimental
characterization, reactive intermediates, and clinical implications.
1. Lajiness M.S., Vieth M., Erickson J. (2004) Molecular properties Chem Res Toxicol;21:189–205.
that influence oral drug-like behavior. Curr Opin Drug Discov 20. Jorgensen W.L. (2004) The many roles of computation in drug
Devel;7:470–477. discovery. Science;303:1813–1818.
2. Pratt W.B., Taylor P. (1990) Principles of Drug Action: the Basis 21. Schames J.R., Henchman R.H., Siegel J.S., Sotriffer C.A., Ni H.,
of Pharmacology. Churchill Livingstone, New York, NY, USA. McCammon J.A. (2004) Discovery of a novel binding trench in
3. Ortiz de Montellano P.R. (2005) Cytochrome P450: Structure, HIV integrase. J Med Chem;47:1879–1881.
Mechanism, and Biochemistry, 3rd edn. Kluwer Academic ⁄ Plenum 22. Wester M.R., Johnson E.F., Marques-Soares C., Dansette P.M.,
Publishers, New York, NY, USA. Mansuy D., Stout C.D. (2003) Structure of a substrate complex
4. van de Waterbeemd H., Gifford E. (2003) ADMET in silico model- of mammalian cytochrome P450 2C5 at 2.3 A resolution: evi-
ling: towards prediction paradise? Nat Rev Drug Discov;2:192– dence for multiple substrate binding modes. Biochemis-
204. try;42:6370–6379.
5. de Groot M.J. (2006) Designing better drugs: predicting cyto- 23. Kuhn B., Jacobsen W., Christians U., Benet L.Z., Kollman P.A.
chrome P450 metabolism. Drug Discov Today;11:601–606. (2001) Metabolism of sirolimus and its derivative everolimus by
6. Fratev F., Benfenati E. (2008) A combination of 3D-QSAR, dock- cytochrome P450 3A4: insights from docking, molecular dynam-
ing, local-binding energy (LBE) and GRID study of the species ics, and quantum chemical calculations. J Med Chem;44:2027–
differences in the carcinogenicity of benzene derivatives chemi- 2034.
cals. J Mol Graph Model;27:147–160. 24. Kroemer R.T. (2007) Structure-based drug design: docking and
7. Leach A.R. (2001) Molecular Modelling: principles and Applica- scoring. Curr Protein Pept Sci;8:312–328.
tions, 2 edn. Pearson Prentice Hall, London, UK. 25. Ito Y., Kondo H., Goldfarb P.S., Lewis D.F. (2008) Analysis of
8. Kola I., Landis J. (2004) Can the pharmaceutical industry reduce CYP2D6 substrate interactions by computational methods. J Mol
attrition rates? Nat Rev Drug Discov;3:711–715. Graph Model;26:947–956.
9. Schnecke V., Bostrom J. (2006) Computational chemistry-driven 26. Ahlstrom M.M., Ridderstrom M., Zamora I. (2007) CYP2C9 struc-
decision making in lead generation. Drug Discov Today;11:43– ture-metabolism relationships: substrates, inhibitors, and metab-
50. olites. J Med Chem;50:5382–5391.
10. Bhogal N., Grindon C., Combes R., Balls M. (2005) Toxicity test- 27. Kitchen D.B., Decornez H., Furr J.R., Bajorath J. (2004) Docking
ing: creating a revolution based on new technologies. Trends and scoring in virtual screening for drug discovery: methods and
Biotechnol;23:299–307. applications. Nat Rev Drug Discov;3:935–949.
11. Baillie T.A. (2008) Metabolism and toxicity of drugs. Two dec- 28. Bazeley P.S., Prithivi S., Struble C.A., Povinelli R.J., Sem D.S.
ades of progress in industrial drug metabolism. Chem Res Toxi- (2006) Synergistic use of compound properties and docking
col;21:129–137. scores in neural network modeling of CYP2D6 binding: predicting
12. Stevens J.L. (2006) Future of toxicology–mechanisms of toxicity affinity and conformational sampling. J Chem Inf
and drug safety: where do we go from here? Chem Res Toxi- Model;46:2698–2708.
col;19:1393–1401. 29. Muralidhara B.K., Negi S., Chin C.C., Braun W., Halpert J.R.
13. Smith D.A., Obach R.S. (2005) Seeing through the mist: abun- (2006) Conformational flexibility of mammalian cytochrome P450
dance versus percentage. Commentary on metabolites in safety 2B4 in binding imidazole inhibitors with different ring chemistry
testing. Drug Metab Dispos;33:1409–1417. and side chains. J Biol Chem;281:8051–8061.
14. Dykens J.A., Will Y. (2007) The significance of mitochondrial tox- 30. de Graaf C., Vermeulen N.P., Feenstra K.A. (2005) Cytochrome
icity testing in drug development. Drug Discov Today;12:777– p450 in silico: an integrative modeling approach. J Med
785. Chem;48:2725–2755.
15. Baillie T.A. (2006) Future of toxicology-metabolic activation and 31. Bathelt C.M., Mulholland A.J., Harvey J.N. (2008) QM ⁄ MM
drug design: challenges and opportunities in chemical toxicology. modeling of benzene hydroxylation in human cytochrome P450
Chem Res Toxicol;19:889–893. 2C9. J Phys Chem A;112:13149–13156.

14 Chem Biol Drug Des 2010; 75: 3–17


Structure-based Drug Metabolism Predictions

32. Cohen S., Kozuch S., Hazan C., Shaik S. (2006) Does substrate chromes P450: a possible explanation for their different cytotox-
oxidation determine the regioselectivity of cyclohexene and pro- icity. Drug Metab Dispos;36:2381–2392.
pene oxidation by cytochrome p450?. J Am Chem 47. Bikadi Z., Hazai E. (2008) In silico description of differential e-
Soc;128:11028–11029. nantioselectivity in methoxychlor O-demethylation by CYP2C
33. Hirao H., Kumar D., Thiel W., Shaik S. (2005) Two states and enzymes. Biochim Biophys Acta;1780:1070–1079.
two more in the mechanisms of hydroxylation and epoxidation 48. Julsing M.K., Vasilev N.P., Schneidman-Duhovny D., Muntendam
by cytochrome P450. J Am Chem Soc;127:13007–13018. R., Woerdenbag H.J., Quax W.J. et al. (2008) Metabolic stere-
34. Fishelovitch D., Hazan C., Hirao H., Wolfson H.J., Nussinov R., oselectivity of cytochrome P450 3A4 towards deoxypodophyllo-
Shaik S. (2007) QM ⁄ MM study of the active species of the toxin: in silico predictions and experimental validation. Eur J
human cytochrome P450 3A4, and the influence thereof of the Med Chem;43:1171–1179.
multiple substrate binding. J Phys Chem B;111:13822–13832. 49. Koymans L., Van Lenthe J.H., Donne-op Den Kelder G.M., Ver-
35. Iori F., da Fonseca R., Ramos M.J., Menziani M.C. (2005) Theo- meulen N.P. (1990) Mechanisms of activation of phenacetin to
retical quantitative structure-activity relationships of flavone reactive metabolites by cytochrome P-450: a theoretical study
ligands interacting with cytochrome P450 1A1 and 1A2 iso- involving radical intermediates. Mol Pharmacol;37:452–460.
zymes. Bioorg Med Chem;13:4366–4374. 50. Loew G.H., Goldblum A. (1985) Metabolic activation and toxicity
36. de Graaf C., Oostenbrink C., Keizers P.H., van der Wijst T., Jon- of acetaminophen and related analogs. A theoretical study. Mol
gejan A., Vermeulen N.P. (2006) Catalytic site prediction and vir- Pharmacol;27:375–386.
tual screening of cytochrome P450 2D6 substrates by 51. Potter D.W., Hinson J.A. (1987) Mechanisms of acetaminophen
consideration of water and rescoring in automated docking. J oxidation to N-acetyl-P-benzoquinone imine by horseradish per-
Med Chem;49:2417–2430. oxidase and cytochrome P-450. J Biol Chem;262:966–973.
37. Yano J.K., Wester M.R., Schoch G.A., Griffin K.J., Stout C.D., 52. Hata M., Tanaka Y., Kyoda N., Osakabe T., Yuki H., Ishii I. et al.
Johnson E.F. (2004) The structure of human microsomal cyto- (2008) An epoxidation mechanism of carbamazepine by CYP3A4.
chrome P450 3A4 determined by X-ray crystallography to 2.05-A Bioorg Med Chem;16:5134–5148.
resolution. J Biol Chem;279:38091–38094. 53. Erve J.C., Svensson M.A., von Euler-Chelpin H., Klasson-Wehler
38. de Groot M.J., Ackland M.J., Horne V.A., Alex A.A., Jones B.C. E. (2004) Characterization of glutathione conjugates of the rem-
(1999) A novel approach to predicting P450 mediated drug oxipride hydroquinone metabolite NCQ-344 formed in vitro and
metabolism. CYP2D6 catalyzed N-dealkylation reactions and detection following oxidation by human neutrophils. Chem Res
qualitative metabolite predictions using a combined protein and Toxicol;17:564–571.
pharmacophore model for CYP2D6. J Med Chem;42:4062–4070. 54. Kalgutkar A.S., Vaz A.D., Lame M.E., Henne K.R., Soglia J., Zhao
39. de Groot M.J., Ackland M.J., Horne V.A., Alex A.A., Jones B.C. S.X., Abramov Y.A, Lombardo F., Collin C., Hendsch Z.S., Hop
(1999) Novel approach to predicting P450-mediated drug metab- C.E. (2005) Bioactivation of the nontricyclic antidepressant ne-
olism: development of a combined protein and pharmacophore fazodone to a reactive quinone-imine species in human liver mi-
model for CYP2D6. J Med Chem;42:1515–1524. crosomes and recombinant cytochrome P450 3A4. Drug Metab
40. Langowski J., Long A. (2002) Computer systems for the predic- Dispos;33:243–253.
tion of xenobiotic metabolism. Adv Drug Deliv Rev;54:407–415. 55. McLaughlin L.A., Paine M.J., Kemp C.A., Marechal J.D., Flana-
41. Zhang J.W., Ge G.B., Liu Y., Wang L.M., Liu X.B., Zhang Y.Y., Li gan J.U., Ward C.J., Sutcliffe M.J., Roberts G.C., Wolf C.R.
W., He Y.Q., Wang Z.T., Sun J., Xiao H.B., Yang L. (2008) Tax- (2005) Why is quinidine an inhibitor of cytochrome P450 2D6?
ane's substituents at C3¢ affect its regioselective metabolism: The role of key active-site residues in quinidine binding J Biol
different in vitro metabolism of cephalomannine and paclitaxel. Chem;280:38617–38624.
Drug Metab Dispos;36:418–426. 56. Lill M.A., Dobler M., Vedani A. (2006) Prediction of small-mole-
42. Oh W.S., Kim D.N., Jung J., Cho K.H., No K.T. (2008) New com- cule binding to cytochrome P450 3A4: flexible docking combined
bined model for the prediction of regioselectivity in cytochrome with multidimensional QSAR. ChemMedChem;1:73–81.
P450 ⁄ 3A4 mediated metabolism. J Chem Inf Model;48:591–601. 57. Marechal J.D., Yu J., Brown S., Kapelioukh I., Rankin E.M., Wolf
43. Sykes M.J., McKinnon R.A., Miners J.O. (2008) Prediction of C.R., Roberts G.C., Paine M.J., Sutcliffe M.J. (2006) In silico and
metabolism by cytochrome P450 2C9: alignment and docking in vitro screening for inhibition of cytochrome P450 CYP3A4 by
studies of a validated database of substrates. J Med comedications commonly used by patients with cancer. Drug
Chem;51:780–791. Metab Dispos;34:534–538.
44. Lafite P., Andre F., Zeldin D.C., Dansette P.M., Mansuy D. (2007) 58. Isin E.M., Sohl C.D., Eoff R.L., Guengerich F.P. (2008) Cooperativi-
Unusual regioselectivity and active site topology of human cyto- ty of cytochrome P450 1A2: interactions of 1,4-phenylene diiso-
chrome P450 2J2. Biochemistry;46:10237–10247. cyanide and 1-isopropoxy-4-nitrobenzene. Arch Biochem
45. Keizers P.H., de Graaf C., de Kanter F.J., Oostenbrink C., Feenstra Biophys;473:69–75.
K.A., Commandeur J.N., Vermeulen N.P. (2005) Metabolic regio- 59. Li W., Tang Y., Liu H., Cheng J., Zhu W., Jiang H. (2008) Probing
and stereoselectivity of cytochrome P450 2D6 towards 3,4- ligand binding modes of human cytochrome P450 2J2 by homol-
methylenedioxy-N-alkylamphetamines: in silico predictions and ogy modeling, molecular dynamics simulation, and flexible mole-
experimental validation. J Med Chem;48:6117–6127. cular docking. Proteins;71:938–949.
46. Perrin L., Aninat C., Hamon V., Hayashi Y., Abadie C., Heyd B. 60. Xue Y., Chao E., Zuercher W.J., Willson T.M., Collins J.L., Redin-
et al. (2008) Metabolism of phenylahistin enantiomers by cyto- bo M.R. (2007) Crystal structure of the PXR-T1317 complex pro-

Chem Biol Drug Des 2010; 75: 3–17 15


Sun and Scott

vides a scaffold to examine the potential for receptor antago- 76. Zeevaart J.G., Wang L., Thakur V.V., Leung C.S., Tirado-Rives J.,
nism. Bioorg Med Chem;15:2156–2166. Bailey C.M. et al. (2008) Optimization of azoles as anti-human
61. Gao Y.D., Olson S.H., Balkovec J.M., Zhu Y., Royo I., Yabut J., immunodeficiency virus agents guided by free-energy calcula-
Evers R., Tan E.Y., Tang W., Hartley D.P., Mosley R.T. (2007) tions. J Am Chem Soc;130:9492–9499.
Attenuating pregnane X receptor (PXR) activation: a molecular 77. Rowland P., Blaney F.E., Smyth M.G., Jones J.J., Leydon V.R.,
modelling approach. Xenobiotica;37:124–138. Oxbrow A.K. et al. (2006) Crystal structure of human cytochrome
62. Yukinaga H., Takami T., Shioyama S.H., Tozuka Z., Masumoto H., P450 2D6. J Biol Chem;281:7614–7622.
Okazaki O., Sudo K. (2007) Identification of cytochrome P450 78. Williams P.A., Cosme J., Vinkovic D.M., Ward A., Angove H.C.,
3A4 modification site with reactive metabolite using linear ion Day P.J. et al. (2004) Crystal structures of human cytochrome
trap-Fourier transform mass spectrometry. Chem Res Toxi- P450 3A4 bound to metyrapone and progesterone. Sci-
col;20:1373–1378. ence;305:683–686.
63. Baer B.R., Wienkers L.C., Rock D.A. (2007) Time-dependent inac- 79. Ekroos M., Sjogren T. (2006) Structural basis for ligand promis-
tivation of P450 3A4 by raloxifene: identification of Cys239 as cuity in cytochrome P450 3A4. Proc Natl Acad Sci
the site of apoprotein alkylation. Chem Res Toxicol;20:954–964. USA;103:13682–13687.
64. Pearson J.T., Wahlstrom J.L., Dickmann L.J., Kumar S., Halpert 80. Schoch G.A., Yano J.K., Sansen S., Dansette P.M., Stout C.D.,
J.R., Wienkers L.C., Foti R.S, Rock D.A. (2007) Differential time- Johnson E.F. (2008) Determinants of cytochrome P450 2C8 sub-
dependent inactivation of P450 3A4 and P450 3A5 by raloxifene: strate binding: structures of complexes with montelukast, troglit-
a key role for C239 in quenching reactive intermediates. Chem azone, felodipine, and 9-cis-retinoic acid. J Biol
Res Toxicol;20:1778–1786. Chem;283:17227–17237.
65. Kang P., Liao M., Wester M.R., Leeder J.S., Pearce R.E., Corre- 81. Schoch G.A., Yano J.K., Wester M.R., Griffin K.J., Stout C.D.,
ia M.A. (2008) CYP3A4-Mediated carbamazepine (CBZ) metabo- Johnson E.F. (2004) Structure of human microsomal cytochrome
lism: formation of a covalent CBZ-CYP3A4 adduct and P450 2C8. Evidence for a peripheral fatty acid binding site.
alteration of the enzyme kinetic profile. Drug Metab Dis- J Biol Chem;279:9497–9503.
pos;36:490–499. 82. Yano J.K., Hsu M.H., Griffin K.J., Stout C.D., Johnson E.F. (2005)
66. Kent U.M., Lin H.L., Noon K.R., Harris D.L., Hollenberg P.F. (2006) Structures of human microsomal cytochrome P450 2A6 com-
Metabolism of bergamottin by cytochromes P450 2B6 and 3A5. plexed with coumarin and methoxsalen. Nat Struct Mol
J Pharmacol Exp Ther;318:992–1005. Biol;12:822–823.
67. Kent U.M., Sridar C., Spahlinger G., Hollenberg P.F. (2008) Modi- 83. Sansen S., Yano J.K., Reynald R.L., Schoch G.A., Griffin K.J.,
fication of serine 360 by a reactive intermediate of 17-alpha- Stout C.D., Johnson E.F. (2007) Adaptations for the oxidation of
ethynylestradiol results in mechanism-based inactivation of cyto- polycyclic aromatic hydrocarbons exhibited by the structure of
chrome P450s 2B1 and 2B6. Chem Res Toxicol;21:1956–1963. human P450 1A2. J Biol Chem;282:14348–14355.
68. Keseru G.M., Makara G.M. (2006) Hit discovery and hit-to-lead 84. Smith B.D., Sanders J.L., Porubsky P.R., Lushington G.H., Stout
approaches. Drug Discov Today;11:741–748. C.D., Scott E.E. (2007) Structure of the human lung cytochrome
69. Katharine Holloway M., McGaughey G.B., Coburn C.A., Stachel P450 2A13. J Biol Chem;282:17306–17313.
S.J., Jones K.G., Stanton E.L. et al. (2007) Evaluating scoring 85. Wester M.R., Yano J.K., Schoch G.A., Yang C., Griffin K.J., Stout
functions for docking and designing beta-secretase inhibitors. C.D., Johnson E.F. (2004) The structure of human cytochrome
Bioorg Med Chem Lett;17:823–827. P450 2C9 complexed with flurbiprofen at 2.0-A resolution. J Biol
70. Davis A.M., Keeling D.J., Steele J., Tomkinson N.P., Tinker A.C. Chem;279:35630–35637.
(2005) Components of successful lead generation. Curr Top Med 86. Kartha J.S., Skordos K.W., Sun H., Hall C., Easterwood L.M.,
Chem;5:421–439. Reilly C.A., Yost G.S. (2008) Single mutations change CYP2F3
71. Joseph-McCarthy D., Baber J.C., Feyfant E., Thompson D.C., from a dehydrogenase of 3-methylindole to an oxygenase. Bio-
Humblet C. (2007) Lead optimization via high-throughput molecu- chemistry;47:9756–9770.
lar docking. Curr Opin Drug Discov Devel;10:264–274. 87. Kartha J.S., Yost G.S. (2008) Mechanism-based inactivation of
72. Shaikh S.A., Jain T., Sandhu G., Latha N., Jayaram B. (2007) lung-selective cytochrome P450 CYP2F enzymes. Drug Metab
From drug target to leads–sketching a physicochemical pathway Dispos;36:155–162.
for lead molecule design in silico. Curr Pharm Des;13:3454– 88. Kassahun K., Skordos K., McIntosh I., Slaughter D., Doss G.A.,
3470. Baillie T.A., Yost G.S. (2005) Zafirlukast metabolism by
73. Ahlstrom M.M., Ridderstrom M., Zamora I., Luthman K. (2007) cytochrome P450 3A4 produces an electrophilic alpha,beta-
CYP2C9 structure-metabolism relationships: optimizing the meta- unsaturated iminium species that results in the selective
bolic stability of COX-2 inhibitors. J Med Chem;50:4444–4452. mechanism-based inactivation of the enzyme. Chem Res Toxicol;
74. Armour D., de Groot M.J., Edwards M., Perros M., Price D.A., 18:1427–1437.
Stammen B.L. et al. (2006) The discovery of CCR5 receptor 89. Levesque J.F., Day S.H., Chauret N., Seto C., Trimble L., Bate-
antagonists for the treatment of HIV infection: hit-to-lead stud- man K.P., Silva J. M., Berthelette C., Lachance N., Boyd M., Li
ies. ChemMedChem;1:706–709. L., Sturino C.F., Wang Z., Zamboni R., Young R.N., Nicoll-Griffith
75. Wang X., Kolasa T., El Kouhen O.F., Chovan L.E., Black-Shaefer D.A. (2007) Metabolic activation of indole-containing prostaglan-
C.L., Wagenaar F.L. et al. (2007) Rapid hit to lead evaluation of din D2 receptor 1 antagonists: impacts of glutathione trapping
pyrazolo[3,4-d]pyrimidin-4-one as selective and orally bioavail- and glucuronide conjugation on covalent binding. Bioorg Med
able mGluR1 antagonists. Bioorg Med Chem Lett;17:4303–4307. Chem Lett;17:3038–3043.

16 Chem Biol Drug Des 2010; 75: 3–17


Structure-based Drug Metabolism Predictions

90. Sun H., Yost G.S. (2008) Metabolic activation of a novel 3-sub- catalyzed bioactivation of 5-aminooxindole and 5-aminobenzsul-
stituted indole-containing TNF-alpha inhibitor: dehydrogenation tam scaffolds in proline-rich tyrosine kinase 2 (PYK2) inhibitors:
and inactivation of CYP3A4. Chem Res Toxicol;21:374–385. retrospective analysis by CYP3A4 molecular docking, quantum
91. Ring B.J., Patterson B.E., Mitchell M.I., Vandenbranden M., Gil- chemical calculations and glutathione adduct detection using lin-
lespie J., Bedding A.W., Jewell H., Payne C.D., Forgue S.T., Eck- ear ion trap ⁄ orbitrap mass spectrometry. Bioorg Med Chem
stein J., Wrighton S.A., Phillips D.L. (2005) Effect of tadalafil on Lett;19:3177–3182.
cytochrome P450 3A4-mediated clearance: studies in vitro and 93. Maiorov V., Sheridan R.P. (2005) Enhanced virtual screening by
in vivo. Clin Pharmacol Ther;77:63–75. combined use of two docking methods: getting the most on a
92. Sun H., Sharma R., Bauman J., Walker D.P., Aspnes G.E., limited budget. J Chem Inf Model;45:1017–1023.
Zawistoski M.P., Kalgutkar A.S. (2009) Differences in CYP3A4

Chem Biol Drug Des 2010; 75: 3–17 17

You might also like