You are on page 1of 8

Toxicology Mechanisms and Methods, 18:243–250, 2008

Copyright 
c Informa Healthcare USA, Inc.
ISSN: 1537-6516 print; 1537-6524 online
DOI: 10.1080/15376510701857189

Combined Computational Metabolite Prediction and


Automated Structure-Based Analysis of Mass
Spectrometric Data
David D. Stranz
Sierra Analytics, Inc., 5815 ABSTRACT As high-throughput technologies have developed in the phar-
Stoddard Road, Suite 601, maceutical industry, the demand for identification of possible metabolites
Modesto, CA 95356, USA
using predominantly liquid chromatographic/mass spectrometry-mass spec-
Shichang Miao trometry/mass spectrometry (LC/MS-MS/MS) for a large number of molecules
Amgen, Inc., Department of in drug discovery has also increased. In parallel, computational technologies
Pharmacokinetics and Drug have also been developed to generate predictions for metabolites alongside
Metabolism,1120 Veterans methods to predict MS spectra and score the quality of the match with
Blvd., South San Francisco,
experimental spectra. The goal of the current study was to generate metabolite
CA 94080;
Current address:
predictions from molecular structure with a software product, MetaDrug. In
ChemoCentryx Inc., 850 vitro microsomal incubations were used to ultimately produce MS data that
Maude Ave., Mountain View, could be used to verify the predictions with Apex, which is a new software tool
CA 94043, USA that can predict the molecular ion spectrum and a fragmentation spectrum,
automating the detailed examination of both MS and MS/MS spectra. For
Scott Campbell and
George Maydwell
the test molecule imipramine used to illustrate the combined in vitro/in silico
Sierra Analytics, Inc., 5815 process proposed, MetaDrug predicts 16 metabolites. Following rat microsomal
Stoddard Road, Suite 601, incubations with imipramine and analysis of the MSn data using the Apex
Modesto, CA 95356, USA software, strong evidence was found for imipramine and five metabolites and
weaker evidence for five additional metabolites. This study suggests a new
Sean Ekins
ACT LLC, 601 Runnymede Ave,
approach to streamline MS data analysis using a combination of predictive
Jenkintown, PA 19046; computational approaches with software capable of comparing the predicted
Department of Pharmaceutical metabolite output with empirical data when looking at drug metabolites.
Sciences, University of
Maryland, 20 Penn Street, KEYWORDS Apex; Metabolite; Mass Spectroscopy; MetaDrug; QSAR
Baltimore, MD 21201, USA
INTRODUCTION
Research in metabolism is aimed at answering the questions about how the molecule is
metabolized, which enzymes are involved, what are the sites of metabolism, the resulting
Received 21 July 2007; metabolites, the rate of metabolism (Austel and Kutter 1983), and whether the metabolites
accepted 28 August 2007. are biologically active or potentially toxic (Smith and Obach 2005). It is also important to
SE acknowledges GeneGo Inc. Sierra
understand species differences in metabolism and the impact this may have on extrapolation
Analytics acknowledges Rocco from preclinical species to man. As there have been increases in the throughput of
Falchetto of Novartis AG for initial experimental in vitro systems using various enzymes, data sets are being generated that
support for the development of Apex. can be used for computational models. As the majority of xenobiotics undergo phase I
Address correspondence to Sean Ekins
metabolism via the cytochrome P450 (P450) enzymes primarily in liver, intestine, and
M.Sc., Ph.D, D.Sc., ACT LLC, 601 kidney (Paine et al. 1997), this family of enzymes has been well studied. These enzymes have
Runnymede Ave, Jenkintown, PA therefore been extensively modeled using various computational and structural methods.
19046. E-mail: ekinssean@yahoo.com, The history of methods used for the computational prediction of human drug
sekins@arnoldllc.com metabolism includes several different approaches such as databases, quantitative structure

243
metabolism relationships (QSMRs)/quantitative structure- similarity as a measure of similarity to training set molecules.
activity relationships (QSARs), pharmacophores (de Groot and Structural alerts for likely reactive metabolites (Li 2002; Uetrecht
Ekins 2002), statistical QSAR approaches (Shen et al. 2003; 2003; Williams and Park 2003) are also provided. MetaDrug
Balakin et al. 2004a, 2004b), rule-based approaches, electronic has been tested with 66 molecules and captured approximately
models (Singh et al. 2003; Korzekwa et al. 2004), homol- 79% of first-pass metabolites (Ekins et al. 2006). A more recent
ogy models, and crystal structures with docking approaches application of the underlying metabolism database has been for
(Jolivette and Ekins 2007). Metabolism knowledge bases (Darvas the creation of Kernel-Partial Least Squares (K-PLS) models for
1987; Klopman et al. 1994, 1997; Talafous et al. 1994; Boyer individual metabolic transformations that could be used to rank
and Zamora 2002; Langowski and Long 2002; Borodina et al. suggested metabolites (Embrechts and Ekins 2007).
2003, 2004; Button et al. 2003; Erhardt 2003) combine data In the drug discovery process, it is imperative for medicinal
or rules from many different mammalian species and tend to chemists to quickly find out the major metabolic “soft spots”
predict all the metabolic possibilities for a molecule (Nassar and in the lead molecule structure, so that analog compounds can
Talaat 2004), rather than the likely metabolites, thus limiting be designed and synthesized to address the metabolic stability
their usefulness in drug discovery. problems. The conventional method is to perform in vitro
These techniques when used individually have different and/or in vivo metabolic studies and identify the structures
levels of success and could be combined to improve predictions, of the metabolites using spectroscopic methods such as LC-
perhaps by deploying the most appropriate models based on MS/MS and NMR. This is often a costly and time-consuming
the molecule neighborhood for the test molecule compared to process. With considerable emphasis now on increasing the
training sets. P450–substrate/inhibitor recognition interactions efficiency of drug discovery, there is interest in using the types
have been studied extensively and have generally shown the of predictive computational approaches (as described above) in
importance of hydrophobic, hydrogen bonding and ionizable as many areas as possible, particularly in early drug discovery.
features for both substrates based on Km data and inhibitors Reliable and rapid metabolite prediction using computational
using Ki , IC50 , and percent inhibition data (Ekins et al. approaches, if achievable, could become a valuable tool for drug
2001). Molecular models that account for electronic effects discovery.
of ligands for P450-mediated metabolism have also been The most powerful tool for analysis of in vivo and in vitro
produced (Jones and Korzekwa 1996; Jones et al. 2002) and metabolite samples is mass spectrometry, typically coupled
these have combined aliphatic and aromatic oxidation reactions with liquid chromatographic separation in an online LC/MS
to generate predictions for metabolic regioselectivities. The instrument. Mass spectrometry can provide molecular weight
combination of approaches may also balance the strengths and information with high accuracy and sensitivity, and with
weaknesses of each approach and hence the introduction of such tandem MS/MS instruments, information about molecular
hybrid methods is ongoing. For example, a recent technique structure as well. Confirmation of the likely presence of pre-
called MetaSite (Molecular Discovery, Middlesex, UK) gener- dicted metabolites using LC/MS and LC/MS/MS is generally
ates GRID field descriptors used for determining energetically a straightforward procedure, but requires expert knowledge
favorable binding sites on molecules of known structure using in mass spectral interpretation. This manual interpretation is
crystal structures or homology models for the P450 enzymes and a bottleneck in the metabolite ID process, severely limiting
the interaction energy descriptors for the molecules evaluated throughput. Mass spectrometer manufacturers have responded
as substrates (Berellini et al. 2005). A reactivity component is to this need by providing some tools for metabolite detection,
also used in the MetaSite calculation to produce a probability but to our knowledge none of them explicitly takes the chemical
for an atom to be metabolized. This approach has been applied structure of the metabolite into account. Thus, there is always
with angiotensin II receptor antagonists predicting the site of some question where isomers are concerned, and a manual
metabolism for CYP2C9 and CYP3A4 (Berellini et al. 2005). interpretation is required.
A recent study has expanded the application to CYP2D6, With recent interest in using metabolite prediction software
CYP2C19, and CYP1A2 with 75% to 86% correct predictions with mass spectrometry data analysis (Anari et al. 2004; Nassar
(Cruciani et al. 2005). MetaSite has been compared with docking and Talaat 2004; Shu et al. 2004; Anari and Baillie 2005; Lin
of ligands into crystal structures and homology models of et al. 2005), we have used MetaDrug along with a second
CYP3A4, and MetaSite had 78% overall success compared with new software tool, Apex, which can predict the molecular
57% for docking (Zhou et al. 2006). MetaSite has a limitation of ion spectrum and a fragmentation spectrum that can in turn
not being able to predict the absolute or relative amounts of the be used to determine which predicted molecules are present
major and minor metabolites as well as the rate of metabolite in the analyte. This technology is compatible with all mass
formation for a molecule. A second hybrid method, MetaDrug, spectrometers and hence the combination of MetaDrug (or
includes a manually annotated Oracle database of human another predictive metabolism tool) and Apex enables the
drug metabolism information including xenobiotic reactions, streamlining of metabolite prediction and mass spectrometry
enzyme substrates, and enzyme inhibitors with kinetic data analysis. In silico metabolite prediction typically generates many
(Ekins et al. 2005a; Ekins et al. 2005b; Ekins et al. 2005c). more potential metabolites than are actually observed. This
The MetaDrug database has been used to predict some of presents a considerable problem for manual interpretation of
the major metabolic pathways and identify the involvement the mass spectral data. The analyst must examine each peak
of P450s using multiple QSAR models enabling the prediction detected in the liquid chromatogram and compare the mass
of affinity and rate of metabolism for numerous enzymes (Ekins spectra under that peak with the spectra expected for the
et al. 2005a, 2006). The user may also upload his or her own structures. Quite often, many of the predicted metabolites are
QSAR or QSMR data into the software to offer a further level of isomers, requiring a detailed examination of both MS and
utility. Steps have been undertaken to provide more confidence MS/MS spectra to determine which, if any, are correct. By
in predictions as the QSAR methods also provide Tanimoto automating this tedious process, Apex removes the potential

D. D. Stranz et al. 244


for error. Every structure is compared against every spectrum ACN. The eluent was ionized in the mass spectrometer using
in a fast and comprehensive procedure. Predictions containing electrospray ionization. Spectra were recorded in positive ion
tens to thousands of structures are evaluated in a few seconds, mode.
without bias, and metabolites that are actually present in the
sample are easily confirmed. The purpose of the current study
was to illustrate how these independently developed software MetaDrug Method for Metabolite
tools can be utilized to identify likely metabolites (using
imipramine as an example), and ultimately frees resources for Prediction
the identification of other metabolites that are currently not The development of MetaDrug (GeneGo Inc, St. Joseph,
suggested by available metabolism knowledge bases. MI) has been described in detail previously (Ekins et al. 2005a,
2005d). Approximately 80 human metabolic reaction rules
(Ekins et al. 2006) were included in MetaDrug at the time of
EXPERIMENTAL the current study. The molecular structure of imipramine was
used in the mol file format and uploaded into MetaDrug. Using
Reagents and Chemicals the metabolite rules coded into the software, likely metabolites
Rat microsomes were purchased from BD Gentest (Woburn, were predicted. These molecules were then exported as an sdf
MA) and were stored at −80◦ C until use. NADPH and file.
imipramine were purchased from Sigma (St. Louis, MO) and
Aldrich Chemical Company (Milwaukee, WI), respectively.
Data Processing Using Apex
The underlying equivalence between chemical structure,
In Vitro Incubations chemical formula, and molecular weight is fundamental to the
Incubations were conducted in 0.5 mL of 0.1 M phosphate ability of mass spectrometry to distinguish among molecules
buffer (pH 7.4) with 10 µM imipramine, 1.0 mg/mL rat liver based on mass (more precisely, the mass-to-charge ratio, or
microsomes, and 1.3 mM NADPH for 1 h at 37◦ C in open m/z, as is measured by the instrument). Apex (Sierra Analytics,
polypropylene tubes in a shaking water bath. The reaction was Modesto, CA) is fully structure based and requires a minimum
terminated with two volumes of acetonitrile and centrifuged at of user expertise in mass spectral interpretation. The software is
4,000 rpm for 20 min. The supernatant was dried down in a written using the C++ programming language for the Microsoft
speed vac and the residue was reconstituted in 400 µL water- Windows operating system. All algorithms for chromatographic
methanol (2:1) for LCMS injection. and spectral data processing and structure-spectrum correlation
were developed in-house by Sierra Analytics.
In use, Apex imports the substrate and metabolite structures
contained in the sdf file produced by MetaDrug. From these
Instrumentation structures, MS and MS/MS spectra are predicted for comparison
The reconstituted sample was examined in the positive with the experimental data. The software can take into account
electrospray ionization mode on a Thermo Electron (San multiple adducts, charge states, oligomers, and neutral losses
Jose, CA) LCQ instrument, using a data-dependent LC/MSn when predicting spectral features. For MS/MS, Apex uses heuris-
acquisition with six scan events in each scan cycle (event tic rules to create a straightforward estimate of the fragmentation
1: MS full scan; events 2 to 3: MS/MS without and with of a molecule: from the two-dimensional chemical structure,
wide band activation, respectively; Events 4 to 6: MS3 on the find all rings and determine the order of all bonds. Then, for
three most abundant MS/MS fragment ions from event 3). In each acyclic single bond, cleave the molecule into two parts at
this experiment, molecular ion (MS) spectra are continuously that bond, and compute the mass of each part. Additional rules
recorded during the LC separation. When a molecular ion is are applied to predict cross-ring cleavages in saturated rings. The
observed with intensity above a predetermined threshold, an fragmentation “spectrum” is the set of all such pairs of masses,
MS/MS fragmentation spectrum of that ion is automatically where each feature in this “spectrum” has the same intensity.
generated and recorded (collision energy = 35%; activation Q = Since the primary discriminant among candidate structures is
0.25; activation time = 30 ms). Ions observed in the MS/MS molecular weight, this simulated spectrum is generally sufficient
spectrum are then isolated and subjected to further stages of to distinguish among isomers or isobars by matching one or
fragmentation (MS3 ). The result is a combined MS, MS/MS, more predicted and observed fragment ions.
and MS3 data set with molecular weight and fragmentation Next, the experimental MS data set is imported directly from
information on all significant species that can be detected under the MS instrument’s raw format. Apex supports most of the
the experimental conditions. major vendors and MS data system formats. The spectra are
For these experiments, 40 µL of sample was injected onto peak detected and centroided for comparison with the spectra
a Shiseido Capcell PAK C18 AQ 3µ 2 × 100 mm reverse predicted from the structures. Using a comprehensive scoring
phase column (Phenomenex, Torrance, CA) with equilibration and correlation algorithm, the mass spectral data is searched for
at 100% solvent A over 1.5 min, binary gradient elution to 25% the presence of each of the predicted structures. Each structure
solvent B over 20 min, followed by isocratic elution at 100% is given a composite score based on the degree of match between
solvent B for 2 min and re-equilibration at 100% solvent A the predicted and experimental spectra. The structures are then
over 1.5 min for a total run time of 25 min. The flow rate was ranked by correlation score. These three basic mass spectral
400 µL/min throughout. Solvent A was 0.1% formic acid (FA) properties of molecules, their mass, isotopic distribution, and
in 95% water/acetonitrile (ACN); solvent B was 0.08% FA in estimated fragmentation, can be used to derive scores for the

245 Metabolite Prediction and Mass Spectrometric Data Analysis


TABLE 1 Metabolites of imipramine identified with MetaDrug and Apex
Experimentally
Metabolites Predicted determined Comments

N-demethylation (desipramine) Yes Yes Best match overall; all MS/MS features
matched
Aromatic hydroxylation (e.g., Yes (4 isomers) Yes Moderate score; isomers are
2-hydroxylation, 10-hydroxylation) indistinguishable by MS and MS/MS
Alkyl hydroxylation Yes (3 isomers) Yes High score, but biased by lack of observed
MS/MS fragments
N-oxidation Yes (2 isomers) Yes Oxidation of dimethylamine most likely
2-hydroxy desipramine No∗ N/A
Didesmethylimipramine No∗ N/A
Iminodibenzyl Yes No
∗ These metabolites would be predicted if MetaDrug was used in the sequential mode.
† Apex cannot determine presence of structures de novo. The only structures that can be determined are those imported from the prediction software.

correlation of a molecule against experimental data. Apex uses the spectra in the data set. (4) MS/MS similarity score: Using the
each of these properties in several ways to create a set of scores. estimated fragmentation spectrum, match the predicted features
Each score is in the range of zero (no match) to one (perfect against those observed and weight according to the intensity of
match). (1) Similarity score: Using predicted mass and isotope the matched features. The weighting takes into account the
cluster, compute the “distance” between the predicted features expectation that the major features in the spectrum should be
and those observed in a spectrum. If the spectrum contains accounted for by a predicted fragment. (5) In addition, external
none of the predicted features, the score is zero; if the spectrum signals (such as from an LC/UV or radiochemical detector) can
is identical to the prediction, the score is one. A partial match be incorporated as “scores,” where the signal is normalized to
to the prediction, either by mass or relative isotopic abundance, a 0–1 range. Each of the structure-based scores is computed
yields a score between zero and one. (2) Purity score: Compute for each spectrum in the data set. If a score is plotted for each
the similarity score, and then weight it by the summed intensity spectrum in time order, the result is a score chromatogram. Peaks
of the matched spectral features divided by the total intensity appear in the score chromatogram where the underlying spectra
of the spectrum. A high score results when the isotope cluster have high score values for that metric. Each individual structure
is both a good match and is the most intense set of features will have a set of score chromatograms, one for each computed
in the spectrum; the spectrum is pure for that species. (3) score.
Molecular ion cluster (MIC) score: Sum the intensities of the When a number of potential metabolite structures are
matched spectral features, and then normalize this over all of presented to Apex, as is the case with MetaDrug, Apex

FIGURE 1 The integrated in silico/in vitro metabolite prediction protocol using MetaDrug and Apex for predicting and verifying
metabolites with mass spectrometry data.

D. D. Stranz et al. 246


FIGURE 2 Apex identification of imipramine metabolites showing detailed match results for predicted and experimentally observed
metabolites, with the correlation score for each of the potential metabolites.

247 Metabolite Prediction and Mass Spectrometric Data Analysis


computes this set of score chromatograms for every predicted distinguish among the alkyl-hydroxylated metabolite isomers.
structure. A typical metabolite prediction result may contain The aromatic-hydroxylated isomers cannot be distinguished due
ten to a hundred or more candidate structures, while a typical both to lack of observed fragments and because Apex does
chromatographically separated mass spectrometric data set not predict fragmentation across aromatic rings. In both cases,
might contain a thousand to several thousand individual mass analysis of bona fide standards of these metabolites would likely
spectra, and usually several individual scores are combined to be required for a definitive identification.
yield the final score for any one molecule. The total number
of score chromatogram points that must be computed is the
product of the number of molecules times the number of CONCLUSION
scores times the number of individual mass spectra. This total Software tools for predicting potential metabolites of small
will usually be in the hundreds of thousands to millions of molecule substrates in early drug discovery are important in
score computations. Apex has been optimized to make this guiding lead optimization in drug discovery to produce drug
computational process as fast as possible; a typical calculation candidates with desirable metabolic and toxicological proper-
with 100 structures and 1,000 MS and MS/MS spectra takes a ties. We suggest that the integration of available computational
second or two on a 2-GHz Pentium PC. tools for prediction of metabolites such as MetaDrug with
MS and software such as Apex for combining the outputs of
both represents a method for improving the throughput of
RESULTS AND DISCUSSION metabolite identification studies and builds on earlier studies
(Anari et al. 2004; Nassar and Talaat 2004; Anari and Baillie
While there has been some interest in using different metabo-
2005). This combined process will likely become a standard
lite prediction software programs with mass spectrometry data
practice in the near future for the pharmaceutical industry. In
analysis (Anari et al. 2004; Nassar and Talaat 2004; Shu et al.
fact, we are already seeing the development of tools such as Mass
2004; Anari and Baillie 2005; Lin et al. 2005), to our knowledge
Frontier that have more predictive capabilities. Future studies
there has been no detailed discussion of the integration of
may likely include using additional compounds incubated with
such approaches. The current study briefly highlights how two
human microsomal or other in vitro systems, extending the
separate software products could be used to process a molecular
present work performed with rat microsomes. While we can
structure, suggest metabolites, and ultimately verify which of
certainly not claim to have solved the problem of metabolite
these metabolites are observed from experimental data. While
prediction, there are many limitations to be aware of. Our
we would not suggest that our approach is ideal or in fact the
knowledge base or “training set” from which the metabolite
only tool available, we think it is important to bring some of
prediction rules have been derived could still be considered
the difficulties inherent in these approaches to the attention of
tiny compared to chemistry space. Hence, these methods tend
the reader.
to overpredict metabolites. The impact of methods to limit the
number of predicted metabolites to those that are most likely
and ranking them (Embrechts and Ekins 2007) will need to
Application Example be assessed to provide further confidence in the predictions.
The metabolism of imipramine, a tricyclic antidepressant, In this previous study K-PLS was used with augmented atom
has been characterized extensively in vitro and in vivo in human descriptors to generate models to classify whether a metabolite
and rat (Strandgarden and Gunnarsson 1994; van Breemen et al. is likely to be produced from a particular starting structure alone.
1998). It is known to undergo N-demethylation, hydroxylation, Over 300 molecules, including parent drugs and their primary
and N-oxidation. This molecule therefore represents a good and secondary (sequential) metabolites, were used to build
example for evaluating the approach taken for metabolite iden- these models corresponding to individual metabolism rules.
tification in the current study. MetaDrug was used to generate Each model was internally validated to assess the capability
an sdf file containing 15 unique predicted metabolite structures to classify other molecules. Receiver operator curve statistics
plus the substrate itself (Table 1). The predicted structures and models for N-dealkylation and O-dealkylation had AUC values
experimental MS data were then imported into Apex (Fig. 1). from 0.75 to 0.84 and were able to predict between 61%
After selection of appropriate scoring criteria for Apex, the and 79% of active molecules upon leave-out testing. Other
set of structures were correlated against all of the MS and efforts at adding prediction confidence or domains such as
MS/MS spectra generated from the rat liver microsomal samples Support Vector Machine models for P450s have been described
(Fig. 2). In this example, three scores were computed for each previously (Ekins et al. 2007). The metabolite prediction
structure: MS similarity (match between actual and predicted efforts outlined to date and used for validation of software
molecular ion spectrum), isotopic purity (percentage of the are relatively simple compared with some of the known
overall spectral intensity due to the structure), and MS/MS metabolic pathways of drugs and endogenous compounds that
similarity (match between actual and predicted fragmentation can involve multiple phase I and phase II reactions. The
pattern for the structure). After scoring, an overall correlation relative rates of formation of particular metabolites may be
for each structure was computed as the product of the three important to predict. Some of these metabolites may even be
individual scores and a threshold was applied to eliminate inhibitors for other enzymes involved in the same metabolic
structures with low correlation. Of the 16 structures, six had high pathways and it would be useful to assess or predict this
and five more had moderate correlation scores (Table 1). These impact.
included the known primary metabolites: N-demethylation to As virtually all computational metabolism prediction efforts
form desipramine, aromatic 2-hydroxylation of imipramine to have been on human, we should address the considerable
form 2-hydroxyimipramine, and imipramine itself. A general amount of metabolism data for mouse and rat, which may
scarcity of fragments in the MS/MS spectra made it difficult to enable us to understand species differences (Nedelcheva and

D. D. Stranz et al. 248


Gut 1994). Compilation of metabolite rules for different species Ekins, S., Andreyev, S., Ryabov, A., Kirilov, E., Rakhmatulin, E. A., Bugrim,
will also be important to consider in future software. Four A., and Nikolskaya, T. 2005a. Computational prediction of human
different rule-based methods have been described in a recent drug metabolism. Exp. Opin. Drug Metab. Toxicol. 1:303–324.
review and a comparison of the predictive ability of the methods Ekins, S., Bugrim, A., Nikolsky, Y., and Nikolskaya, T. 2005b. Systems
Biology: Applications in Drug Discovery. Drug Discovery Handbook,
was limited to seven phenolic compounds (Kulkarni et al.
ed. Gad, S., Wiley, New York, pp. 123–183
2005). To be of relevance to the pharmaceutical industry, these
Ekins, S., de Groot, M., and Jones, J. P. 2001. Pharmacophore and three
rule-based methods need more extensive testing with diverse dimensional quantitative structure activity relationship methods
drug-like structures. An in-depth comparison of the different for modeling cytochrome P450 active sites. Drug Metab. Dispos.
metabolite prediction algorithms is beyond the scope of this 29:936–944.
current study but is also needed in order to ensure the optimal Ekins, S., Embrechts, M. J., Breneman, C. M., Jim, K., Wery J.-P.
selection of software components for the integrated in silico/in 2007. Novel applications pf Kernel-partial least squares to mod-
vitro metabolite prediction protocol described herein. eling a comprehensive array of properties for drug discovery. In
Computational Toxicology: Risk Assessment for Pharmaceutical
and Environmental Chemicals, ed. Ekins, S., Wiley-Interscience,
REFERENCES Hoboken, NJ, pp. 403–432
Ekins, S., Kirillov, E., Rakhmatulin, E., and Nikolskaya, T. 2005c. A novel
Anari, M. R., and Baillie, T. A. 2005. Bridging cheminformatic metabolite method for visualizing nuclear hormone receptor networks relevant
prediction and tandem mass spectrometry. Drug Discov. Today to drug metabolism. Drug Metab. Dispos. 33:474–481.
10:711–717. Ekins, S., Nikolsky, Y., and Nikolskaya, T. 2005d. Techniques: application
Anari, M. R., Sanchez, R. I., Bakhtiar, R., Franklin, R. B., and Baillie, T. A. of systems biology to absorption, distribution, metabolism, excre-
2004. Integration of knowledge-based metabolic predictions with tion, and toxicity. Trends Pharmacol. Sci. 26:202–209.
liquid chromatography data-dependent tandem mass spectrometry Embrechts, M. J., and Ekins, S. 2007. Classification of metabolites with
for drug metabolism studies: application to studies on the biotrans- Kernel-Partial Least Squares (K-PLS). Drug Metab. Dispos. 35:325–
formation of indinavir. Anal. Chem. 76:823–832. 327.
Austel, V., Kutter, E. 1983. Absorption, Distribution, and Metabolism of Erhardt, P. W. 2003. A human drug metabolism database: potential roles
drugs. Quantitative Structure-Activity Relationships of Drugs, ed. in the quantitative predictions of drug metabolism and metabolism-
Topliss, J. G., Academic Press, New York, pp. 437–496 related drug-drug interactions. Curr. Drug Metab. 4:411–422.
Balakin, K. V., Ekins, S., Bugrim, A., Ivanenkov, Y. A., Korolev, D., Jolivette, L. J., and Ekins, S. 2007. Methods for predicting human drug
Nikolsky, Y., Ivashchenko, A. A., Savchuk, N. P., and Nikolskaya, T. metabolism. Adv. Clin. Chem. 43:131–176.
2004a. Quantitative structure-metabolism relationship modeling of Jones, J. P., Korzekwa, K. R. 1996. Predicting the Rates and Regioselec-
the metabolic N-dealkylation rates. Drug Metab. Dispos. 32:1111– tivity of Reactions Mediated by the P450 Superfamily, Academic
1120. Press, Inc., New York.
Balakin, K. V., Ekins, S., Bugrim, A., Ivanenkov, Y. A., Korolev, D., Jones, J. P., Mysinger, M., and Korzekwa, K. R. 2002. Computational
Nikolsky, Y., Skorenko, S. A., Ivashchenko, A. A., Savchuk, N. models for cytochrome P450: a predictive electronic model for
P., and Nikolskaya, T. 2004b. Kohonen maps for prediction of aromatic oxidation and hydrogen abstraction. Drug Metab. Dispos.
binding to human cytochrome P450 3A4. Drug Metab. Dispos. 30:7–12.
32:1183–1189. Klopman, G., Dimayuga, M., and Talafous, J. 1994. META. 1. A program
Berellini, G., Cruciani, G., and Mannhold, R. 2005. Pharmacophore, drug for the evaluation of metabolic transformations of chemicals. J.
metabolism, and pharmacokinetics models on non-peptide AT1, Chem. Inf. Comput. Sci. 34:1320–1325.
AT2, and AT1/AT2 angiotensin II receptor antagonists. J. Med. Klopman, G., Tu, M., and Talafous, J. 1997. META. 3. A genetic
Chem. 48:4389–4399. algorithm for metabolic transform priorities optimization. J. Chem.
Borodina, Y., Rudik, A., Filimonov, D., Kharchevnikova, N., Dmitriev, A., Inf. Comput. Sci. 37:329–334.
Blinova, V., and Poroikov, V. 2004. A new statistical approach to Korzekwa, K., Ewing, T. J., Kocher, J. P., Carlson, T. J. 2004. Models
predicting aromatic hydroxylation sites. Comparison with model- for cytochrome P450-mediated metabolism. In Pharmaceutical
based approaches. J. Chem. Inf. Comput. Sci. 44:1998–2009. Profiling in Drug Discovery for Lead Selection. Borchardt, R. T, Kerns,
Borodina, Y., Sadym, A., Filimonov, D., Blinova, V., Dmitriev, A., and E. H., Lipinski, C. A., Thakker, D. R., and Wang, B., (Eds.). Arlington,
Poroikov, V. 2003. Predicting biotransformation potential from VA: AAPS Press, pp. 69–80
molecular structure. J. Chem. Inf. Comput. Sci. 43:1636–1646. Kulkarni, S. A., Zhu, J., and Blechinger, S. 2005. In silico techniques
Boyer, S., and Zamora, I. 2002. New methods in predictive metabolism. for the study and prediction of xenobiotic metabolism: a review.
J. Comp. Aided Mol. Des. 16:403–413. Xenobiotica 35:955–973.
Button, W. G., Judson, P. N., Long, A., and Vessey, J. D. 2003. Using Langowski, J., and Long, A. 2002. Computer systems for the prediction
absolute and relative reasoning in the prediction of the potential of xenobiotic metabolism. Adv. Drug Del. Rev. 54:407–415.
metabolism of xenobiotics. J. Chem. Inf. Compu. Sci. 43:1371– Li, A. P. 2002. A review of the common properties of drugs with idiosyn-
1377. cratic hepatotoxicity and the “multiple determinant hypothesis” for
Cruciani, G., Carosati, E., De Boeck, B., Ethirajulu, K., Mackie, C., Howe, the manifestation of idiosyncratic drug toxicity. Chem. Biol. Interact.
T., and Vianello, R. 2005. MetaSite: understanding metabolism in 142:7–23.
human cytochromes from the perspective of the chemist. J. Med. Lin, T., Pang, J., Hao, C., Stranz, D., Campbell, S., Mordenti, J., and
Chem. 48:6970–6979. Pan, L. 2005. Semi-automated metabolite identification using
Darvas, F. 1987. MetabolExpert: an expert system for predicting the information-dependent LC/MS/MS acquisition, in silico metabolite
metabolism of substances. In QSAR in Environmental Toxicology, prediction, and structure based data analysis. Drug Metab. Rev.
ed. Kaiser, K. L., Reidel Co., Dordrecht, pp. 71–81. 37(suppl 2):399.
de Groot, M. J., and Ekins, S. 2002. Pharmacophore modeling of Nassar, A. E., and Talaat, R. E. 2004. Strategies for dealing with metabolite
cytochromes P450. Adv. Drug Del. Rev. 54:367–383. elucidation in drug discovery and development. Drug Discov. Today
Ekins, S., Andreyev, S., Ryabov, A., Kirillov, E., Rakhmatulin, E. A., 9:317–327.
Sorokina, S., Bugrim, A., and Nikolskaya, T. 2006. A combined Nedelcheva, V., and Gut, I. 1994. P450 in the rat and man: methods
approach to drug metabolism and toxicity assessment. Drug Metab. of investigation, substrate specificities and relevance to cancer.
Dispos. 34:495–503. Xenobiotica 24:1151–1175.

249 Metabolite Prediction and Mass Spectrometric Data Analysis


Paine, M. F., Khalighi, M., Fisher, J. M., Shen, D. D., Kunze, K. L., Marsh, Strandgarden, K., and Gunnarsson, P. O. 1994. Metabolism of
C. L., Perkins, J. D., and Thummel, K. E. 1997. Characterization lofepramine and imipramine in liver microsomes from rat and man.
of interintestinal and intraintestinal variations in human CYP3A- Xenobiotica 24:703–711.
dependent metabolism. J. Pharmacol. Exper. Ther. 283:1552–1562. Talafous, J., Sayre, L. M., Mieyal, J. J., and Klopman, G. 1994. META. 2.
Shen, M., Xiao, Y., Golbraikh, A., Gombar, V. K., and Tropsha, A. 2003. A dictionary model of mammalian xenobiotic metabolism. J. Chem.
Development and validation of k-nearest neighbour QSPR models Inf. Comput. Sci. 34:1326–1333.
of metabolic stability of drug candidates. J. Med. Chem. 46:3013– Uetrecht, J. 2003. Screening for the potential of a drug candidate to
3020. cause idiosyncratic drug reactions. Drug Discov. Today 8:832–837.
Shu, Y.-Z., Arora, V., Alberts, J., Belcastro, J., and Skiles, G. 2004. Bridging van Breemen, R. B., Nikolic, D., and Bolton, J. L. 1998. Metabolic screening
metabolite prediction and metabolite identification- the use of using on-line ultrafiltration mass spectrometry. Drug Metab. Dispos.
metabolite prediction software for LC/MS data mining of potential 26:85–90.
metabolites. Drug Metab. Rev. 36(suppl 1):103. Williams, D. P., and Park, B. K. 2003. Idiosyncratic toxicity: the role
Singh, S. B., Shen, L. Q., Walker, M. J., and Sheridan, R. P. 2003. A of toxicophores and bioactivation. Drug Discov. Today 8:1044–
model for likely sites of CYP3A4-mediated metabolism on drug-like 1050.
molecules. J. Med. Chem. 46:1330–1336. Zhou, D., Afzelius, L., Grimm, S. W., Andersson, T. B., Zauhar, R. J.,
Smith, D. A., and Obach, R. S. 2005. Seeing through the mist: abundance and Zamora, I. 2006. Comparison of methods for the prediction
versus percentage. Commentary on metabolites in safety testing. of the metabolic sites for CYP3A4-mediated metabolic reactions.
Drug Metab. Dispos. 33:1409–1417. Drug Metab. Dispos. 34:976–983.

D. D. Stranz et al. 250

You might also like