Вы находитесь на странице: 1из 18

ENCL 100200012—8/10/2001——00012

SPECTROSCOPIC METHODS OF ANALYSIS—MASS SPECTROMETRY

Mike S. Lee
Milestone Development Services, Newtown, Pennsylvania

INTRODUCTION reviewed. This article will focus on MS technologies with


regard to specific applications in drug development. The
intent of this article is to provide an overview of MS
The dramatically increased expenditures for both in-house
applications and describe the significant integration of this
and outsourced pharmaceutical research and development
technology into drug development. A detailed and in-
(R&D) have led to a greater dependence on technology.
depth overview of current MS technologies and appli-
New technologies are constantly introduced into drug
cations can be obtained from the recent proceedings of the
development to address throughput issues and improve
American Society for Mass Spectrometry Conference on
development cycles. The incorporation of new technol-
Mass Spectrometry and Allied Topics (www.asms.org)
ogies has resulted in fundamental change in the drug
and the Association of Biomolecular Resource Facilities
development paradigm. Recently, sample generating-
(www.abrf.org).
based technologies such as high throughput biomolecular
screening and automated parallel synthesis have shifted
the bottleneck to sample analysis-based technologies.
GENOMICS
The current focus on analytical techniques in the
pharmaceutical industry emphasizes four primary figures
of merit: sensitivity; selectivity; speed; and high Though the contributions of MS have been somewhat
throughput. Mass spectrometry (MS) provides each of limited in the field of genomics, there has been increased
these key attributes, and therefore, has been benchmarked participation and interest (6). Certainly, the worldwide
an effective solution for pharmaceutical analysis in each recognition received from the Human Genome Project
stage of drug development (1). Perhaps more enabling than created a sense of urgency toward determining genetic
the MS-based technology itself is the diverse applications variation.
of MS in conjunction with sample preparation, chromato- Genomics refers to the study of genetic data to draw
graphic separation, and informatics. It is within this correlation between individual genetic inheritance and
context that MS has played an increasingly vital role in the medically or biologically important parameters. For
pharmaceutical industry and has become the preferred example, these parameters may involve a patient’s
½F1 analytical method for trace-mixture analysis (Fig. 1). response to a specific drug. Knowledge of the genetic
A variety of MS formats are widely accepted and basis of individual drug response may provide under-
applied in the pharmaceutical industry. The specific MS standing of the observed variability in drug response
application is often defined by the sample introduction arising as a result of genetically determined differences in
technique. The pharmaceutical applications highlighted in drug absorption, disposition, metabolism, or excretion (7).
this article feature two types of sample introduction Furthermore, knowledge of genetic variation may be
techniques: dynamic and static. Dynamic sample intro- useful during the target selection process when multiple
duction involves the use of high-performance liquid targets are available within a specific disease state. Thus,
chromatography (HPLC) on-line with MS. The resulting the pharmaceutical industry has great interest in
liquid chromatography/mass spectrometry (LC/MS) for- determining the genetic variation in patient populations.
mat provides unique and enabling capabilities for Due to the existence of sequence variations, or
pharmaceutical analysis. The electrospray ionization polymorphisms, no two human genomes are identical.
(ESI) (2) and atmospheric pressure chemical ionization Single nucleotide polymorphisms (SNPs) are the most
(APCI) (3) modes are the most widely used. Static sample abundant genetic variation with an estimated frequency of
introduction techniques primarily use matrix-assisted laser 1 SNP per 500 basepairs. Since SNPs are so prevalent in
desorption/ionization (MALDI) (4). the genome, they can act as markers that are linked with a
The advances in MS instrumentation (5) and role of MS phenotype to provide a comprehensive measure of
within the pharmaceutical industry (1) have been recently interaction with a specific drug. The validation of a
Encyclopedia of Pharmaceutical Technology
Copyright q 2002 by Marcel Dekker, Inc. All rights reserved. 2545
ENCL 100200012—8/10/2001——00012

2546 Spectroscopic Methods of Analysis—Mass Spectrometry

Pure Mixture
step where a separate post-PCR primer is hybridized
directly adjacent to the SNP site. The resulting MALDI-
TOF MS data provided a direct confirmation of molecular
Non- X-ray LC/NMR
Trace NMR
weight for fast analysis of polymorphisms.
Knowledge of the DNA sequence flanking the SNP site
allows for the optimum choice of post-PCR primer size,
primer location, and dideoxy-nucleotide(s). Thus, the
MS HPLC/UV
assay can be designed to extract complete information
Trace
UV LC/MS about the SNP regardless of its state. This emerging MS-
IR based approach for SNP genotyping has potential to be
highly automated without the requirement for fluorescent
tags. Furthermore, “multiplexing” can be attained by
Fig. 1 Structure analysis matrix that illustrates pharmaceutical selecting post-PCR primers of varying lengths to dedicate
analysis preferences for four specific sample types: nontrace/-
predetermined regions of the mass spectrum to specific
pure; nontrace/mixture; trace/pure; and trace/mixture. (Courtesy
SNPs.
of Milestone Development Services, Newtown, PA.)

particular SNP represents an important stage for


establishing SNPs as a routine clinical diagnostic marker. PROTEOMICS
The validation of SNPs via MALDI-TOF MS is emerging
as a valuable genotyping tool (8,9). A schematic of a The study of protein structure, function, quantity, and
½F2 MALDI-TOF MS instrument is shown in Fig. 2. interactions during maturation and progression of disease
A recent study performed by Stroh et al. (10) compared is referred to as proteomics. Analytical approaches that use
the performance of MALDI-TOF MS with restriction a combination of two-dimensional (2-D) gel electrophor-
fragment length polymorphism (RFLP) and fluorescence esis for protein separation and MS analysis for protein
polarization (FP). The study involved the analysis of identification followed by database searches is a widely
known mutations of the IL-1b gene. The procedure practiced proteomics strategy (11). The tryptic peptides
involved amplification of patient DNA samples using extracted from gels are analyzed by MALDI-TOF MS and
standard PCR techniques followed by a primer extension microcolumn or capillary LC tandem mass spectrometry

Fig. 2 Schematic of a MALDI-TOF MS instrument. MALDI-TOF samples are


prepared with a matrix that contains a small organic molecule capable of absorbing
ultraviolet light. A laser is used to desorb ions from the sample plate and the resulting
ions are forced into the flight tube by application of the acceleration voltage from
extraction grids. All ions leave the source with the same kinetic energy and travel
down the flight tube toward an ion reflector. Separation is based on mass with lighter
ions traveling faster than heavier ions. The ion reflector is used to correct for small
kinetic energy differences between ions of the same mass resulting in improved
resolution and mass accuracy. (Courtesy of Applied Biosystems, Framingham, MA.)
ENCL 100200012—8/10/2001——00012

Spectroscopic Methods of Analysis—Mass Spectrometry 2547

Fig. 3 Schematic of a nano-scale capillary ESI interface. This specialized LC/MS


interface, operating at flow rates from 20 to 500 nL/min and using 50 to 100 mm ID
columns, typically provides low femtomole sensitivity. Fully automated sample
handling and preparation procedures (i.e., desalting and preconcentration) combined
with specialized devices for high separation and variable nL gradient flow rates
provide unique capabilities for high-throughput analysis of proteins. (Courtesy of
New Objective, Cambridge, MA.)

(MS/MS) techniques. Typically, the MALDI-TOF MS dependent MS/MS analysis provided excellent
techniques are used to quickly identify peptide fragments sequence coverage for 11 tryptic peptides related to
and confirm the presence of known proteins. Nano-scale A1AT in a single LC/MS run. A tryptic peptide that
capillary LC/MS/MS techniques (using 50 – 100 mm corresponds to the A1AT sequence SVLGQLGITK
diameter columns, operating at flow rates of 20 –500 observed at retention time (rt) 30.5 min. was observed
nL/min) are used to further interrogate the complex protein in the spectrum. The LC/MS data also provided
mixture at the low femtomole level. These techniques sequence information on unmatched MALDI peaks.
½F3 require the use of a specialized ESI source shown in Fig. 3. The need to detect lower concentration of protein and
The combination of MALDI-TOF MS and capillary peptide mixtures has resulted in the increased use of
LC/MS/MS was recently described for the identification of hybrid quadrupole/orthogonal TOF (QTOF) MS/MS
disease state markers in human urine (12). In this study, instruments (13) in conjunction with microcolumn LC.
urine proteins obtained from emphysema patients were ½F5 Fig. 5 shows a schematic of a QTOF instrument. This
separated on 2-D gels and selected spots were digested LC/MS approach provides a resolution of ca. 0.1 mass
with trypsin and analyzed by MALDI-TOF. A database units allowing for the analysis of complex product ion
search using Protein Prospector identified a potential spectra (14,15). A recent publication by Chalmers and
biomarker for emphysema as human alpha-1-antitrypsin Gaskell highlights the current challenges in proteome
(A1AT). The corresponding MALDI spectrum contained analysis with regard to MS instrumentation (16).
nine out of 18 peptides with masses that match the
expected tryptic digest fragments for A1AT.
NATURAL PRODUCTS DEREPLICATION
The same tryptic digest protein sample was analyzed
by capillary LC/MS/MS using an ion trap mass
spectrometer followed by a database search with Historically, an excellent source of novel lead drug
½F4 SEQUEST. Fig. 4 illustrates the components of an ion compounds is natural products. Natural product screening
trap mass spectrometer. The highly automated data- activities typically occur during drug discovery and
ENCL 100200012—8/10/2001——00012

2548 Spectroscopic Methods of Analysis—Mass Spectrometry

involve the testing of crude extracts obtained from


microbial fermentation broths, plants, or marine organ-
isms. When activity above a certain level is detected,
active components are isolated and purified for identifi-
cation. This process is often time-consuming, where the
physicochemical characteristics of the active components
are determined, known compounds are identified (dere-
plication), and the novel compounds are scaled-up for
more detailed investigation.
Analysis strategies that use on-line ESI-LC/MS
approaches provide an integrated format for natural
product dereplication by combining traditional fraction
collection, sample preparation, and multi-component
analysis into a single step. In this way, crude extracts are
screened without extensive purification and chemical
analysis. Furthermore, less material is required due to the
sensitivity of the technique and chromatographic resol-
Fig. 4 Schematic of an ion trap MS instrument. This device ution is retained.
consists of two endcap electrodes (entrance and exit) and a ring The key to natural products analysis using this approach
electrode. An ion trap MS separates ions based on mass-to-charge is dependable molecular weight determination. This
ratio (m/z). Once ions are introduced into the ion trap MS, the information is used with existing natural product databases
radiofrequency (rf) amplitude is increased so that ions are
that contain information on the bioactive compounds, the
sequentially ejected (by increasing mass) and detected. This type
physical descriptions of the microorganisms from which
of MS provides a routine (i.e., benchtop) and sensitive detector
using either GC and LC interfaces. Furthermore, this instrument they come, their spectrum of activity, the method of
provides a unique format for multiple stages of MS analysis extraction and isolation, and physical data (i.e., molecular
(MSn). (Courtesy of ThermoFinnigan, San Jose, CA.) weight, UV absorption maxima). Molecular weight is the

Fig. 5 Schematic of a QTOF MS instrument. Ions formed in the source region are
introduced into a quadrupole mass filter (see Fig. 7) that separates ions based on
mass-to-charge ratio (m/z). Selected ions are then transferred into the TOF MS for
detailed analysis (i.e., high resolution capabilities). (Courtesy of Micromass,
Manchester, UK.)
ENCL 100200012—8/10/2001——00012

Spectroscopic Methods of Analysis—Mass Spectrometry 2549

most critical information for initial searches because of its An important development in the quest for high
link to structural specificity. This information is used to throughput combinatorial library analysis was the multiple
make pivotal decisions on whether or not to proceed to ESI interface described by Wang et al. (28). This novel
more time-consuming isolation steps based on novelty of ESI interface enabled effluent flow streams from an array
the compound. In recently reported studies (17,18), of four HPLC columns to be sampled independently and
LC/MS is used to increase sensitivity and accelerate sequentially using a quadrupole MS instrument. The
analysis. These features serve to significantly reduce labor. interface featured a stepping motor and rotating plate
A recent review described LC/MS-based approaches for assembly. The effluent flow from the HPLC columns was
the characterization of natural product mixtures in connected to a parallel arrangement of electrospray
conjunction with high-throughput screening (19). needles co-axial to the mass spectrometer entrance
The instrumental configuration of the LC/MS system aperture. The individual spray tips were positioned 908
developed by Ackermann et al. (17) features an HPLC, UV relative to one another in a circular array. Each spray
detector, fraction collector, ESI-tandem quadrupole position was sampled multiple times per second by precise
(MS/MS), and MALDI-TOF MS. Filtered fermentation control of the stepping motor assembly.
broths were extracted with butanol or ethyl acetate, and The parallel sample analysis format using a multiplexed
eluted on a gradient C18 reversed-phase HPLC separation. LC/MS interface with an orthogonal time-of-flight (TOF)
The eluent was split 1:10 between a tandem quadrupole MS was described by de Biasi et al. (29). This approach
MS/MS instrument (scanning 250 – 2000 amu/3 sec in the illustrated the high-throughput capabilities of a multi-
full scan mode) and a single wavelength UV detector (254 plexed ESI interface in combination with an MS format
or 230 nm). One-min fractions were collected after the UV that accommodates fast chromatography methodologies.
detector. Of these fractions, 20– 50 mL is used for The system featured a four-way multiplexed electrospray
MALDI-TOF analysis, and the remainder is concentrated interface attached directly with the existing source of the
for microbiological testing. The LC/UV chromatogram TOF-MS instrument. A rotating aperture driven by a
was compared to the bioactivity assay histogram to variable speed stepper motor permitted the sampling of the
highlight the peaks that contain activity. The molecular spray from each electrospray probe tip. The data files were
weights of the active peaks were obtained for novelty synchronized with the corresponding spray.
assessment of the compounds. As the preparation of large libraries for lead discovery
Similar approaches that use on-line LC/MS and became routine, the burden placed on analysis techniques
LC/MS/MS techniques have been recently described for focused mainly on throughput and quality (30). However,
natural products dereplication (20,21). Approximately,000 biological assay requirements typically required pure
natural product extracts can be screened annually for in compounds. Thus, the focus shifted toward the use of
vivo and in vitro activity using LC/MS-based systems. A automated high throughput purification methods applied to
standard approach for dereplication involves a comparison libraries of discrete compounds (31). Reverse-phase
of retention time, full scan mass spectra (i.e., molecular analytical and preparative HPLC formats in conjunction
weight information), and MS/MS spectra with those from with MS techniques have been critical for the high
known biologically active standards. Thus, previously throughput purification approaches for parallel synthesis
identified components are rapidly eliminated and do not libraries. A variety of approaches that featured the use of
require time-consuming structure elucidation studies. The gradient methods, short columns, and high flow rates have
savings of effort allow researchers to focus efforts on been described (32). Highly automated LC/MS approaches
novel chemistries. Samples of novel compounds can then for purification at the multimilligram level were described
be infused into an ion trap mass spectrometer, and a for a quadrupole system by Zeng et al. (33). These
multiple stage mass analysis (MSn) fragmentation map is methods featured the use of short columns that were
generated. operated at ultra high flow rates. Preparative columns were
operated at flow rates in excess of 70 ml/min to match the
linear velocity of the short analytical columns (4.0
COMBINATORIAL CHEMISTRY
ml/min). Analytical LC/MS analyses of compound
libraries were achieved in 5 min for chromatographically,
Recent reviews (22,,25) describe MS-based methods well-behaved compounds. Slightly longer preparative
ranging from the analysis of complex molecular libraries LC/MS analysis times (8 –10 min/sample) were required
(26) to open-access formats for drug discovery and for compounds that exhibited poor chromatographic peak
development (27). High throughput criteria were central to shapes and/or for compound mixtures the required higher
each application. resolution separations. The fraction collection process is
ENCL 100200012—8/10/2001——00012

2550 Spectroscopic Methods of Analysis—Mass Spectrometry

initiated in real-time once the reconstructed ion current is used to calculate the binding affinity of the components. In
observed for a specific m/z value that corresponds to the the example described above, two incubation cycles
compound of interest. This design permitted the collection followed by the SEC separation provided an enhancement
of one sample per fraction. Thus, the need for very large of strong binders to weak binders. This LC/MS-based
fraction collector beds and postpurification screening and method provides a unique approach to obtain information
pooling was eliminated. Unattended and automated in situations when lower concentrations of tighter binding
operation of this system led to the purification of ligands are present in the same mixture with higher
overcompounds (mg quantities) per day. concentrations of weaker binding ligands. Furthermore,
this method is more efficient than synthetic deconvolution
procedures and does not require the use of radioligands.
BIOAFFINITY SCREENING
Combinatorial chemistry initiatives have created a
tremendous challenge for activities that deal with the
With the integration of highly automated parallel synthesis screening of these mixtures for activity against a specified
techniques into drug discovery programs, hundreds of target (37). MS-based approaches that use affinity
thousands of compounds are now screened against a selection (38), encoding methodologies (39 – 41), pulsed
particular biological target. Once activity is determined for ultrafiltration (42), and anti-aggregatory approaches (43)
a mixture, the identification of the active component(s) is have been described.
necessary. A recent study described the use of bioaffinity The use of MS formats that provide accurate mass
selection LC/MS methods for the identification of active capabilities have been recently illustrated for screening
mixture component(s) (34). This approach features an combinatorial libraries (44 –47). The unambiguous con-
integrated bioaffinity-based LC/MS screening method to firmation/identification of combinatorial library com-
separate and identify compounds from mixtures. ponents from small quantities of material have been
A mixture of compounds is incubated with the target illustrated using a hybrid quadrupole/orthogonal TOF
protein and the components bound to the protein are (QTOF) (44,45) and Fourier transform ion cyclotron
selected by using a size exclusion chromatography (SEC) resonance (FTICR) (47) mass spectrometry. A schematic
“spin column.” In this experiment, the unbound com- ½F6 of a FTICR-MS system is shown in Fig. 6. Accurate
pounds are retained on the column. The bound components isotope patterns or “isotopic signature” and unique mass
are eluted and identified with LC/MS. Increased specificity differences between isobaric compounds can be obtained
is obtained by dissociating the bound compounds and using these two MS formats.
performing a second equilibration incubation with the
protein. This procedure preferentially selects for the
OPEN-ACCESS SYSTEMS
compounds with higher affinity, and results in an
enhancement of the quantitative LC/MS response.
Iterative stages of incubation, size-exclusion, and Chemists now routinely use open-access MS systems in
LC/MS allow the tighter binding components to be the same way that they previously used thin-layer
enriched relative to weaker binding components. chromatography (TLC) to monitor reaction mixtures for
In this study, the peroxisome proliferator-activated the desired product and to optimize reaction conditions. In
receptor (PPARg), which is a target for anti-diabetic drugs practice, medicinal chemists require only molecular
(construct molecular weight of 32,537 Da), is incubated weight data, and are comfortable with a variety of MS
with 10 ligands that range in molecular weight from 283 to ionization methods to obtain this information. However,
587 units. A spin column of 6000 Da cutoff is used for confidence in the actual method and procedure is a
SEC purposes. The retained mixture of components is requisite. Today, molecular mass measurement has
analyzed by fast perfusive chromatography (35,36), using quickly become a preferred means of structure confir-
a standard full-scan LC/MS strategy. This analysis mation over NMR and IR during the early stages of
procedure allows for the identification and quantitation synthetic chemistry activities (i.e., drug discovery), where
of the protein and the ligands, compared to their responses sample quantities are limited.
prior to incubation. The ligand-protein complex that In the open-access LC/MS procedure described by
dissociated under the reversed-phase chromatographic Pullen et al. (48), the samples are directly introduced from
conditions is selectively detected. solution for ease of automation and sample preparation.
This analysis scheme provided a quick measurement of Chemists prepare samples in solvent to a suggested
binding affinity, and serves as a screening tool during drug concentration range, then log the samples into the system.
candidate selection. Spreadsheets were constructed and The sample log-in is done at any time during the
ENCL 100200012—8/10/2001——00012

Spectroscopic Methods of Analysis—Mass Spectrometry 2551

Fig. 6 Schematic of a FTICR MS instrument. This type of MS consists of an ion cyclotron resonance (ICR) analyzer cell that is situated
in the homogeneous region of a large magnet. The ions introduced into the ICR analyzer are constrained (trapped) by the magnetic field to
move in circular orbits with a specific frequency that corresponds to a specific mass-to-charge ratio (m/z). Mass analysis occurs when
radiofrequency (rf) potential is applied (pulsed) to the ICR analyzer so that all ions are accelerated to a larger orbit radius. After the pulse
is turned off, the transient image current is acquired and a Fourier transform separates the individual cyclotron frequencies. Repeating this
pulsing process to accumulate several transients is used to improve the signal-to-noise ratio. (Courtesy of Bruker Daltonics, Billerica,
MA.)

continuous automated queue. Autosampler vials are used compounds; 2) reaction monitoring; 3) reaction optimiz-
to hold the samples, and autosamplers are used to directly ation; 4) analysis of preparative HPLC fractions; and 5)
deliver samples in solution to the mass spectrometer. The analysis of TLC plate spots. The availability of these
system uses a standard method to analyze the samples in methods led to the increased use of LC/MS for structural
queue, average spectra according to a preset scheme, and analysis. The short analysis time and reliable structure
print out a spectrum for the chemist. Fail-safe procedures confirmation resulted in the use of LC/MS as a first choice
for untrained users and instrument self-maintenance at for structure characterization for synthetic chemistry
start-up and shutdown were also developed. applications.
Taylor et al. (49) further demonstrated the value of Open-access LC/MS formats have spawned new
open-access LC/MS systems for generating a widened dimensions in access and data management. The use of a
scope of pharmaceutical analysis applications, including direct exposure probe (DEP) for automated sample
1) characterization of synthetic intermediates and target introduction has been developed for quick (ca. 3 min)
ENCL 100200012—8/10/2001——00012

2552 Spectroscopic Methods of Analysis—Mass Spectrometry

molecular weight determination of new lead compounds analysis of synthetic products, intermediates, reactants,
½F7 and quantitative analysis (50). Fig. 7 illustrates an reagents, and contaminants. A list of observed ions along
automated direct probe system for molecular weight with known artifact ions is generated and used to provide a
determination. Versatile software packages for data measure of the quality-of-fit to the predicted product(s).
manipulation and processing has been a popular approach Open-access MS systems provide an effective means for
for integrating analysis and information (51 –53). These maintaining the high-throughput characterization of
software programs are efficiently implemented with stand- synthetic compounds. These systems offer an efficient
alone computers and servers that are networked with open- laboratory- to bench-scale integration of sample gener-
access mass spectrometer data systems. In this configur- ation and analysis activities. Advances in analytical
ation, the data are generated, visualized, processed, and instrumentation and electronic communication have also
automatically reported for the chemist. The program played a major role in the emergence and acceptance of
compares a template of predicted molecular ions with the MS as a front-line tool for structure characterization.
actual ions generated by ESI and APCI for the quick

Fig. 7 Schematic of an automated direct probe system for molecular weight determination that features an ion trap MS. Samples are
dissolved in a suitable solvent and injected via an automated syringe system onto the DEP wire. The probe is injected into the MS via an
automated isolation valve system and the temperature is ramped to the programmed temperature. After sample analysis the probe is
removed from the source and heated to a high temperature to clean the DEP wire in preparation for the next sample. This type of
integrated, open-access MS-based application provides routine, unattended support for medicinal chemistry needs such as reaction
monitoring and the optimization of reaction conditions. (Courtesy of Scientific Instrument Services, Ringoes, NJ.)
ENCL 100200012—8/10/2001——00012

Spectroscopic Methods of Analysis—Mass Spectrometry 2553

IN VIVO DRUG SCREENING dependent on the above parameters, as well as on the


degree of tolerance to which the specific screen is set.
The simultaneous pharmacokinetic assessment of
The application of APCI-LC/MS techniques for the rapid
multiple drug candidates in one animal has been termed
determination of protein binding and pharmacokinetics in
“n-in-one” or “cassette dosing.” As discussed for the
drug discovery were recently described by Allen et al.
previous example, this parallel approach results in an
using a single quadrupole instrument (54). A “cocktail”
increased productivity for bioanalysis during drug
approach consisted of four experimental compounds and a
discovery. Beaudry et al. (55) recently investigated the
control compound dosed orally at 1 mg/kg with plasma
extension of this methodology to study larger numbers of
samples obtained at 0.5, 1, 2, 4, and 8 h post dose. To
compounds in each mixture, and to integrate sample
insure reproducibility, the control compound was tested
preparation with the LC/MS/MS system for increased
with each cocktail. This approach generated timely
efficiency.
systemic exposure (AUC and Cmax) data on 44 test
The number of analytes studied in parallel was extended
compounds in three work days, using two laboratory
to 63 plus an internal standard. The increased number of
scientists.
analytes was made possible due to improvements to the
The use of LC tandem quadrupole MS/MS-based
collision region of the MS/MS system that provide
screening approaches for quantitative bioanalytical
increased sensitivity and reduced “memory effects.” In
measurements allow a large, chemically diverse, range
addition, robotic systems for sample handling and on-line
of potential drug candidates to be analyzed quickly and
(solid phase extraction) SPE of plasma samples were
confidently. A schematic of a tandem quadrupole MS/MS
integrated with the LC/MS/MS system. An isocratic
½F8 instrument is shown in Fig. 8. The development of unique
reversed-phase HPLC method provided a cycle time of 4.5
LC/MS-based systems for in vivo pharmacokinetic
min per sample. The on-line sample preparation and short
screening reduces the analysis to a manageable number
analysis resulted in an increased sample throughput that
of samples, and results in a cost-effective approach to
required less time from the scientist. The method produced
evaluate new lead compounds. Approaches to this type of
good performance, in terms of extraction efficiency,
methodology will likely vary, according to the behavior of
linearity, and limit of detection (LOD), and has the
the molecules of interest, standard operating procedures
capability of analyzing 320 –960 samples per day. The
(SOPs), performance capabilities of the mass spec-
strategic emphasis of this approach is on providing high
trometer, and integration of automated sample preparation,
throughput LC/MS methods for evaluating large numbers
and data analysis procedures. Success will likely be
of drug candidates during drug discovery to eliminate poor
pharmacokinetic performers.

METABOLIC STABILITY SCREENING

The use of fast gradient elution LC/MS techniques on a


single quadrupole instrument was described for high
throughput metabolic stability screening (56). The method
uses as HPLC column-switching apparatus to desalt and
analyze lead candidates incubated with human liver
microsomes. Substrates were selected whose in vivo
Fig. 8 Schematic of a tandem quadrupole MS/MS instrument. clearance is controlled predominantly by phase I oxidative
A tandem quadrupole MS/MS instrument consists of two metabolism as opposed to phase II metabolism or renal
quadrupole MS filters, MS1 and MS2, separated by a collision clearance. In this way, the resulting data could be resolved
cell. Each quadrupole MS filter consists of four cylindrical or into four categories of metabolic stability: high ($60%);
hyperbolic shaped rods. A unique combination of direct current
moderate ($30 –59%); low ($10– 29%); and very low
(dc) potential and radiofrequency (rf) potential is applied to each
pair of rods (one pair 1808 out of phase with the other). A mass
(,10%).
spectrum results by varying the voltages at a constant rf/dc ratio. The rapid structure identification of metabolites is a
A variety of scan modes (e.g., full scan, product ion, precursor powerful complement to previously described quantitative
ion, neutral loss) provide unique capabilities for quantitative and approaches. The utility of an automated metabolite
qualitative structure analysis. (Courtesy of Micromass, Manche- identification approach, using LC/MS/MS with an ion
ster, UK.) trap mass spectrometer has been demonstrated (57). In this
ENCL 100200012—8/10/2001——00012

2554 Spectroscopic Methods of Analysis—Mass Spectrometry

study, MSn analysis is automated to provide maximum virtually all the ion current is consolidated into an adduct
structural information in combination with predictive of the molecular ion (i.e., [M 1 H]1, [M 1 NH3]1).
strategies for biotransformation. Automated data-depen- Full-scan mass spectra generally contain an abundant
dent scan functions are used to generate full scan, MS/MS, [M 1 H]1 ion signal with little detectable fragmentation.
and MSn mass spectra of metabolites within a single Product-ion spectra are obtained to reveal product ions and
chromatographic analysis. This feature is unique and neutral losses that are associated with diagnostic
avoids the multiple (2 – 4) injections that are necessary substructures of the buspirone molecule. To assist with
with other MS/MS configurations (e.g., tandem quadru- the MS/MS structure identification, the gross substructure
pole). Along with the significant savings in time, detailed of buspirone is categorized into profile groups (61). Profile
structure information is generated, which enables a groups directly correlate specific product ions and neutral
comprehensive analysis of substructure relationship to be losses with the presence, absence, substitution, and
constructed for each metabolite. These automated studies molecular connectivity (62) of specific buspirone sub-
provide unique advantages during drug discovery, and structures and their modifications. The profile groups of
provide an early perspective on the metabolically labile buspirone are identified with abbreviations that correspond
sites, or “soft spots” of a drug candidate. This knowledge to the three specific substructures: azaspirone decane
is useful during lead optimization activities, and can lead dione (A), butyl piperazine (B), and pyrimidine (P).
to the initiation of proactive research efforts that deal with Substituted substructures are designated with a subscript
metabolism-guided structural modification and toxicity. (s), and a dash (–) denotes substructure connectivity.
Thus, the buspirone molecule is represented by A – B –P.
The As –B – P designation refers to metabolite structures
that contain the azaspirone decane dione, butyl piperazine,
METABOLITE PROFILING AND IDENTIFICATION
and pyrimidine substructures with substitution on the
azaspirone decane dione substructure. The profile group
The application of LC/MS-based techniques for the categorization within a corresponding database allows the
structure identification of drug metabolites has played a rapid visual recognition of primary substructures affected
significant role in drug development. The early identifi- by metabolism.
cation of drug metabolites provides valuable insights into Metabolite structure databases can be easily constructed
the pathways of metabolism and biotransformation. Once and contain information on the structure, molecular
metabolites are identified/confirmed, metabolism-guided weight, UV characteristics, RRT, and product ions of
structural modification during the drug discovery stage is metabolites obtained from rat bile, urine, and liver S9
initiated to facilitate the selection of drug candidates for samples. Using this format, Kerns et al. reported the
subsequent development. predominant buspirone metabolite profile groups as As –
The identification of metabolite structures with LC/MS B – P, A – B – Ps, and As –B– Ps. These profile groups
and LC/MS/MS techniques using quadrupole-based MS indicate azaspirone decane dione and pyrimidine as
instruments are an effective approach due to their ability to metabolically active sites of attack and the presence of
analyze trace mixtures from complex samples of urine, multiple substitution sites on each of these substructures.
bile, and plasma. The key to structure identification
approaches is based on the fact that metabolites generally
retain most of the core structure of the parent drug (58,59).
IMPURITY PROFILING AND IDENTIFICATION
Therefore, the parent drug and its corresponding
metabolites would be expected to undergo similar
fragmentation and to produce mass spectra that indicate Synthetic impurities are of particular concern during
major substructures. process research and safety evaluation activities. Often,
Kerns et al. demonstrated the application of LC/MS and impurities are the result of synthetic by-products or
LC/MS/MS standard method approaches in preclinical starting materials of the scale-up process. Impurities
development for the metabolite identification of buspir- provide a comprehensive indicator of the chemical process
one, a widely used anxiolytic drug (60). The success of this and are diagnostic of overall quality. Process chemists use
method relies on the performance of the LC/MS interface this information to guide process optimization. Knowledge
and the ability to generate abundant ions that correspond to of the identity and relative amount of impurities is used to
the molecular weight of the drug and drug metabolites. diagnose process reactions so that changes in reagents and
The production of abundant molecular ions is an ideal reaction conditions leads to better yields and higher quality
situation for molecular weight confirmation because material.
ENCL 100200012—8/10/2001——00012

Spectroscopic Methods of Analysis—Mass Spectrometry 2555

With an increasing number of novel lead candidates that A standard reversed-phase HPLC method was used for
enter into preclinical development, considerable resources all the samples that are associated with a drug candidate to
are needed to identify impurities. LC/MS-based reduce time-consuming method development/method
approaches provide integrated sample clean-up and refinement procedures. Standard reversed-phase methods
structure analysis procedures for the rapid analysis of typically involve a 20 – 30 min cycle time and provide
impurities. This advantage was demonstrated during the information for a wide range of compounds. The
preclinical development of TAXOLw (18). LC/MS played incorporation of a standard method strategy allows the
an important role for the identification of impurities use of autosampling procedures and standard system
contained in extracts and process intermediates from Taxus software for data analysis.
brevifolia and T. baccata biomass. Because drugs derived During the development of TAXOLw, 90 taxane
from natural sources often have a very diverse set of impurities were rapidly identified and added to the
structural analogs, it is important to determine which structure database. This MS/MS information was routinely
analogs are carried through the purification process and obtained for impurities down to the 100 ng level (injected),
ultimately appear as impurities. This task presents a unique and required approximately 2 –3 h for the analysis of each
challenge during the early stages of drug development due sample. The compounds are structurally categorized with
to the highly complex nature of the samples. profile group terminology. The LC/MS-based methods
Kerns and coworkers described a structure identification were significantly faster than the previously used
strategy that incorporates LC/MS and LC/MS/MS analytical methods based on scale-up, isolation, fraction-
techniques using quadrupole-based instruments for rapid, ation, and individual structural analysis. Software tools
sensitive, and high-throughput impurity analysis (18). This capable of sample tracking, interpretation, and data
approach integrates traditional steps of sample prep- storage facilitate the structure profiling of impurities,
aration, separation, analysis, and data management into a degradants, and metabolites (63). Key pharmaceutical
single instrumental method. The resulting multidimen- analysis elements that deal with sample preparation, real-
sional data include retention time, molecular weight, UV, time analysis decisions, databasing, distribution/visualiza-
and substructure information. A structure database is ½F9 tion of results (Fig. 9) and prediction of fragmentation are
developed for each candidate and is used to rapidly now highly integrated.
identify the same impurities in new samples. Structures are
proposed based on using the drug candidate as a structural
DEGRADANT PROFILING AND IDENTIFICATION
template and, with the use of a standard method approach,
consistency for comparison of results throughout the
preclinical development process is ensured. During the course of drug development, the bulk drug and
Nearly all of the impurities contained the characteristic drug formulation are studied under a variety of stress
paclitaxel core substructure as indicated by the character- conditions such as temperature, humidity, acidity, basicity,
istic product ion at m/z 509 with variations due to oxidization, and light. Qin et al. described the utilization of
modifications. Many of these taxanes contained a side- stressing conditions that may cause degradation (64). The
chain similar to paclitaxel, with variations occurring on the resulting samples may be used to validate analytical
terminal amide of the side chain. The product ions that monitoring methods and to serve as predictive tools for
differed from the characteristic side-chain ions of future formulation and packaging studies.
paclitaxel (m/z 286) by values indicative of specific A traditional approach to study degradant formation
substructures were used to identify these terminal amide involves similar time-consuming scale-up and preparation
variations. A comparison with the paclitaxel substructural steps as described for metabolite and impurity analysis.
template indicated structural differences beyond the Similarly, this area of pharmaceutical analysis has
position of the amide group in the side-chain substructure. experienced the issues associated with faster drug
When a new impurity was encountered during chemical development cycles. Rourick and coworkers recently
process research, retention time and molecular weight described proactive approaches to obtain degradant
information were compared to the database for rapid information with quadrupole LC/MS methods during the
identification. This approach is similar to the procedure preclinical development stage (65). The corresponding
described for natural product dereplication. If the structural information provides insight for decisions on
compound is not contained in the structure database, which leads to further develop for clinical testing. The
then the corresponding LC/MS/MS analysis is performed early structural information on degradants of a drug
to obtain substructural detail and the proposal of a new candidate offers a unique capability for synthetic
structure. modification to minimize degradation. Structural infor-
ENCL 100200012—8/10/2001——00012

2556 Spectroscopic Methods of Analysis—Mass Spectrometry

Fig. 9 Visualization of molecular fragments using a “lasso tool” application. The lasso tool is used to identify a particular fragment and,
if a signal corresponding to its mass is present in the spectrum, the fragment is highlighted and the corresponding assignment is added to
an assignment table. (Courtesy of Advanced Chemistry Development, Toronto, Ontario, Canada.)

mation can also facilitate planning of preclinical drug moisture) as a predictive profile. The coordinated use of
development in process research, formulation develop- LC/MS and LC/MS/MS provide structure identification
ment, and safety assessment. for speed, sensitivity, and high throughput. Standard
The strategy for impurity and degradant identification methods, useful for 80% of the compounds, are applied.
described by Rourick et al. subjects lead candidates to Various types of structural data are obtained for
various development conditions followed by LC/MS and elucidation purposes (e.g., retention time, molecular
LC/MS/MS analysis protocols. A structure database is weight, MS/MS), and unknown compounds are elucidated
constructed from the corresponding results and is used to with the candidate drug as a structural template. The
reveal unstable regions within the drug structure as well as LC/MS analysis provides retention time and molecular
to ascertain which candidate or homologous series of drug weight data, whereas LC/MS/MS provides substructural
candidates may be the most favorable for further detail for structure identification. Drug candidates are
development. High capacity and throughput speed are incubated under drug processing, storage, and physiologi-
necessary so that many lead candidates may be evaluated. cal conditions that were expected to occur throughout drug
Applicability of the method to a wide range of compound lifetime.
classes is desirable. Once the drug candidate enters clinical Using this approach, 10 degradants of cefadroxil, an
development and manufacturing, the structure database is orally effective semisynthetic cephalosporin antibiotic,
useful for the rapid identification of impurities and were elucidated in a 2-day study. The use of standard
degradants in samples generated during these stages of LC/MS methods provided consistency from sample to
development. sample throughout the development process, and allowed
The method exposes drug candidates to forced for the construction and use of a structural database for the
degradation conditions, (e.g., acid, base, heat, and rapid identification of impurities and degradants during
ENCL 100200012—8/10/2001——00012

Spectroscopic Methods of Analysis—Mass Spectrometry 2557

development. The reversed-phase HPLC conditions times of the drug and internal standard were 3.1 and 3.4
provided a general measure of the polarity of each min, respectively.
compound, useful for interpretation of substructural At the time, this application provided a powerful
differences between related compounds. Due to the benchmark for the use of quadrupole LC/MS-based
mass-resolving capability of the mass spectrometer, methods in the pharmaceutical industry and paved the
chromatographic resolution of co-eluting or unresolved way for the tremendous growth of MS-based applications
components was not required. Abundant protonated in support of clinical development. This particular assay
molecule ions, [M 1 H]1, provided reliable molecular successfully supported several clinical studies with
weight information, and product-ion spectra generated sensitive and reliable results. This performance was
valuable substructure information for each degradant. The benchmarked on more than 4000 clinical samples, and led
product-ion spectrum of cefadroxil was used as a template to a widened scope of MS application for quantitative
for interpretation; specific product ions and neutral losses bioanalysis (67 – 71).
were compared to the spectra obtained from the unknown
degradants. Product ions common to each spectrum
provided evidence of substructures unchanged by the
QUANTITATIVE BIOANALYSIS—SELECTED
degradation conditions and differences were indicative of
REACTION MONITORING
structural variations.

The use of selected reaction monitoring (SRM) methods


QUANTITATIVE BIOANALYSIS—SELECTED ION
for quantitative bioanalysis represents increased dimen-
MONITORING
sions of mass spectrometry analysis. A SRM method that
features a tandem quadrupole MS/MS instrument for the
The quantitative analysis of targeted components in quantitative analysis of an antipsychotic agent, clozapine,
physiological fluids is a major requirement in clinical in human plasma was recently described by Dear et al.
development. In 1991, Fouda et al. (66) pioneered the use (72). Preclinical development studies of clozapine in rats
of APCI-LC/MS on a single quadrupole instrument for the and dogs used HPLC with fluorescence detection (FLD).
quantitative determination of the renin inhibitor, CP- With this method, a better limit of quantitation (LOQ) of 1
80,794, in human serum. Because the pharmacological ng/ml was obtained. As the compound moved into the
action is below 200 pg/ml, a quantitative assay in the low clinical stages of development, a more sensitive method of
pg/ml range was required to monitor the drug’s analysis was required to obtain rapid metabolic infor-
pharmacokinetic and pharmacodynamic properties. Also, mation in support of drug safety evaluation studies. As a
the structure of the CP-80,794 molecule lacked a result, a standard LC/MS/MS method was developed for
significant chromophore for UV detection with conven- the quantitative analysis of clozapine (I) and four
tional HPLC methods. Furthermore, the low volatility and metabolites (II-V) in human plasma.
thermal instability precluded analyses with GC/MS The LC/MS/MS strategy deployed is similar to
methods. previously described approaches for protein, natural
Quantitative LC/MS assays in clinical development products, metabolite, and impurity identification. An
generally involve four intensive steps: sample prep- ionization technique that generates abundant molecular
aration; assay calibration; sample analysis; and data ion species with very little fragmentation is desirable. The
management. In the method developed by Fouda and product-ion spectrum is obtained to generate the
coworkers, human serum samples were prepared with a substructural template of the molecule. Abundant and
liquid – liquid extraction procedure. Assay calibration structurally unique transitions (molecular ion ! product
involved the use of human serum samples fortified with ion) are identified from the spectrum, and are used in the
CP-80,794 at 11 concentrations (6 replicates per corresponding SRM experiment for quantitation. The
concentration) ranging from 0.05 to 10 ng/ml. The SRM experiment provides a high degree of selectivity and
LC/MS analysis involved the use of the SIM mode to better LOD than full-scan or SIM experiments for the
monitor the molecular ions[M–H]2 that correspond to the analysis of complex mixtures (73,74). The selectivity of
drug (m/z 619) and internal standard (m/z 633). In this MS/MS reduces the requirements for complete chromato-
particular LC/MS application, the negative ion mode was graphic resolution of each component. Therefore,
highly sensitive for this class of compound. Samples were LC/MS/MS experiments for quantitation typically
loaded onto an HPLC autosampler and 80 mL aliquots are emphasize short analytical run times to provide high
injected onto the column at 4-min intervals. The elution sample throughput.
ENCL 100200012—8/10/2001——00012

2558 Spectroscopic Methods of Analysis—Mass Spectrometry

The inter- and intra-assay precision (% C.V.) of this medical device company to identify the leachables and to
method were reported to be less than 8% across the range provide adequate testing of their toxicity. Monitoring
of the limits of quantification (0.05 – 10 ng/ml). The methods must be developed and validated to effectively
accuracy (% bias) for all spiked control concentrations did control toxic leachables during the manufacture of high
not exceed ^4%. Same-day turnaround of results for over quality pharmaceuticals.
100 samples was possible and was used to support an acute As a result, materials for medical devices and drug
dose tolerance and pharmacokinetic study that involved products must be tested for leachable components. Once a
the analysis of 1600 samples. known toxic compound is discovered, it must be identified
for the assessment of toxicity, followed by the monitoring
of levels using validated methods as required by the FDA.
PEPTIDE MAPPING IN QUALITY CONTROL
This identification procedure could be a time-consuming
process with traditional methods that are based on
Quality control involves a carefully designed series of fractionation and individual component analysis.
analysis and protocols. The purpose of this activity during Tiller et al. demonstrated an analytical strategy with on-
the manufacturing stage of drug development is to ensure line LC/UV/MS and LC/MS/MS to rapidly obtain
the production of safe, high quality drug products. These structural information for leachables from a drug-delivery
measures are helpful for the producers of the product as device (76). Similar to proteomics-based applications, the
well as the regulators (i.e., FDA). In this way, adherence to analysis strategy makes use of “data-dependent” analysis,
protocols and procedure are carefully monitored on a wherein the mass spectrometer first obtains molecular ions
routine basis. When any uncertainty in the manufacturing using full-scan techniques, and makes real-time decisions
process occurs, procedures are referenced and data are about MS/MS product-ion spectra that must be obtained.
analyzed to determine the specific stage of manufacturing In this way, molecular weight and substructural infor-
to begin examining. Thus, the responsibility of drug mation are both obtained for many components during a
manufacturers and regulating agencies are to determine single HPLC run.
when and how a process went awry. The ability to do so in Many components were readily observed from the ESI-
an efficient, straightforward manner is helpful to both LC/MS chromatogram, and several polyester leachables
parties, and ultimately, the consumer. Thus, the ability to were identified. The ESI LC/MS chromatogram revealed
provide this information is highly dependent on the 15 components, compared to the three components that
manufacturer’s ability to control the process. were observed in the 220 nm UV chromatogram. This
The use of LC/MS in a manufacturing quality control difference illustrates the capability of ESI-LC/MS to
environment for biologicals was reported by Chang et al. provide a more universal detection when the analytes do
(75). Their approach involved three steps. First, the not contain strongly UV-absorbing substructures (e.g.,
expected cleavage sites (in this case, trypsin) within the aromatic). The MS-based method proved to be highly
amino acid sequence of the protein somidobove are efficient because molecular weight and substructural
indicated. This “tryptic map” serves as a template for the information via the full-scan and product-ion experiments,
expected peptide fragments. Second, an analytical method respectively, could both be obtained for the sample
using chromatography columns and conditions that components.
provide the best resolution and reproducibility is
developed. An opportunity exists to optimize the analysis
CONCLUSIONS AND FUTURE PROSPECTS
based on chromatography, digestion, and LC/MS per-
formance. Finally, the resulting LC/MS data are profiled,
according to amino acid sequence, peak number, and [M The understanding and application of MS in the
1 H]1. Other properties such as relative retention time can pharmaceutical industry has experienced tremendous
also be added in this format. growth due to unprecedented sample requirements (i.e.,
trace mixture analysis) and commercial pressure (i.e.,
faster development time-lines). Today, MS is an essential
IMPURITY IDENTIFICATION IN QUALITY
component of the modern pharmaceutical laboratory as
CONTROL
well as an important complement to traditional methods of
analysis.
Regulatory authorities strictly scrutinize the leachables An expanded role for highly sensitive assays for both
(e.g., plasticizers, impurities) that may come from medical quantitative and qualitative analysis will likely continue.
devices and drugs. It is the responsibility of the drug or Novel pharmaceutically relevant formats and systems that
ENCL 100200012—8/10/2001——00012

Spectroscopic Methods of Analysis—Mass Spectrometry 2559

Fig. 10 Schematic of an LC/NMR/MS system. In this configuration, NMR is used to provide structural, stereochemical, or ligand
binding information while MS is used to provide molecular weight information. (Courtesy of Bruker Daltonics, Billerica, MA.)

expand current approaches as well as create low-flow, 3. Bruins, A.P.; Covey, T.R.; Henion, J.D. Anal. Chem. 1987,
miniaturized formats (77 –79) are envisioned. Continued 59, 2642– 2646.
4. Hillenkamp, F.; Karas, M.; Beavis, R.C.; Chait, B.T. Anal.
advances in integrated approaches that feature MS will Chem. 1991, 63, 1193A – 1203A.
generate significant interest with regard to pharmaceutical 5. Burlingame, A.L.; Boyd, R.K.; Gaskell, S.J. Anal. Chem.
analysis. For example, the use of integrated systems that 1998, 70, 647R – 716R.
feature NMR/MS have been described for applications that 6. Kelleher, N.L. Chem. Biol. 2000, 7, R37– R45.
involve LC/NMR/MS (80,81) and MS/NMR (82). A 7. Wolf, C.R.; Smith, G. Pharmacogenetics. Br. Medical Bull.
½F10 schematic of an LC/NMR/MS system is shown in Fig. 10. 1999, 55, 366– 386.
8. Ross, P.; Hall, L.; Smirnov, I.; Haff, L. Nat. Biotechnol.
In these configurations, NMR is used to provide structural 1998, 18, 1347– 1351.
and stereochemical information (80,81) or to verify 9. Haff, L.A.; Smirnov, I.P. Genome Res. 1997, 7, 378– 388.
binding/interaction (82) while MS is used to provide 10. Pezzullo, L.II.; Hall, S.K.; Affourtit, J.P.; Seymour, A.B.;
molecular weight information. New challenges that deal Stroh, J.G. Proceedings of the 48th ASMS Conference on
specifically with probing mechanism of action will likely Mass Spectrometry and Allied Topics, Long Beach
California, 2000; 1552.
generate the need for a broader application of MS as well
11. Yates, J.R. J. Mass Spectrom. 1998, 33, 1 – 19.
as spur the development of novel technologies for sample 12. Gale, B.L.; Bleibaum, J.L.; MacKenzie, R.T.; Martin, R.L.;
preparation, chromatography, and information Straub, K.M. Proceedings 48th ASMS Conference Mass
management. Spectrometry and Allied Topics, Long Beach, California,
Q1 2000; 62.
13. Morris, H.R.; Paxton, T.; Dell, A.; Langhorne, J.; Berg, M.;
Bordoli, R.S.; Hoyes, J.; Bateman, R.H. J. Rapid Commun.
REFERENCES Mass Spectrom. 1996, 10, 889– 896.
14. Stevenson, T.I.; Loo, J.A. Proceedings of the 47th ASMS
Conference on Mass Spectrometry and Allied Topics,
1. Lee, M.S.; Kerns, E.H. Mass Spectrom. Rev. 1999, 18, Q1 Dallas, Texas, 1999; 215.
187– 279. 15. Buko, A.; Tang, Q.; Trevillyan, J.; Chiou, G. Proceedings
2. Fenn, J.B.; Mann, M.; Meng, C.K.; Wong, S.F.; White- of the 47th ASMS Conference on Mass Spectrometry and
house, C.M. Science 1989, 246, 64 – 71. Allied Topics, Dallas, Texas, 1999; 248.
ENCL 100200012—8/10/2001——00012

2560 Spectroscopic Methods of Analysis—Mass Spectrometry

16. Chalmers, M.J.; Gaskell, S.J. Curr. Opin. Biotechnol. 2000, 43. Park, S.; Wanna, L.; Johnson, M.E.; Venton, D.L. J. Comb.
11, 384– 390. Chem. 2000, 2, 314– 317.
17. Ackermann, B.L.; Regg, B.T.; Colombo, L.; Stella, S.; 44. Blom, K.F.; Combs, A.P.; Rockwell, A.L.; Oldenburg,
Coutant, J.E. J. Am. Soc. Mass Spectrom. 1996, 7, K.R.; Zhang, J.H.; Chen, T. Rapid Commun. Mass
1227– 1237. Spectrom. 1998, 12, 1192– 1198.
18. Kerns, E.H.; Volk, K.J.; Hill, S.E.; Lee, M.S. J. Nat. Prod. 45. Lane, S.J.; Pipe, A. Rapid Commun. Mass Spectrom. 1999,
1994, 57, 1391– 1403. 13, 798– 814.
19. Strege, M.A. J. Chromatogr. B 1999, 725, 67 – 78. 46. Fang, A.S.; Vouros, P.; Stacey, C.C.; Kruppa, G.H.;
20. Gilbert, J.R.; Lewer, P. Proceedings of the 46th ASMS Laukien, F.H.; Wintner, E.A.; Carell, T.; Rebek, J., Jr.
Conference on Mass Spectrometry and Allied Topics, Comb. Chem. High Throughput Screen 1998, 1, 23 – 33.
Orlando, Florida, 1998; 21. 47. Speir, J.P.; Perkins, G.; Berg, C.; Pullen, F. Rapid
21. Janota, K.; Carter, G.T. Proceedings of the 46th ASMS Commun. Mass Spectrom. 2000, 14, 1937–1942.
Conference on Mass Spectrometry and Allied Topics, 48. Pullen, F.S.; Kerkins, G.L.; Burton, K.I.; Ware, R.S.;
Orlando, Florida, 1998; 557. Teague, M.S.; Kiplinger, J.P. J. Am. Soc. Mass Spectrom.
22. Burdick, D.J.; Stults, J.T. Methods Enzymol. 1997, 289, 1995, 6, 394– 399.
499– 519. 49. Taylor, L.C.E.; Johnson, R.L.; Raso, R. J. Am. Soc. Mass
23. Fitch, W.L. Mol. Divers. 1998-99, 4, 39 – 45. Spectrom. 1995, 6, 387– 393.
24. Sübmuth, R.D.; Jung, G. J. Chromatogr. B Biomed. Sci. 50. Scientific Instrument Services. AutoProbeTM Application
Appl. 1999, 725, 49 – 65. Note; Ringoes: New Jersey, 2000.
25. Swali, V.; Langley, G.J.; Bradley, M. Curr. Opin. Chem. 51. Whitney, J.L.; Kerns, E.H.; Rourick, R.A.; Hail, M.E.;
Biol. 1999, 3, 337– 341. Volk, K.J.; Fink, S.W.; Lee, M.S. Pharm. Tech. 1998, May,
26. Dunayevskiy, Y.; Vouros, P.; Carell, T.; Wintner, E.A.; 76 – 82.
Rebek, J., Jr. Anal. Chem. 1995, 67, 2906– 2915. 52. Tong, H.; Bell, D.; Keiko, T.; Siegel, M.M. J. Am. Soc.
27. Cepa, S.; Searle, P. Proceedings of the 46th ASMS Mass Spectrom. 1999, 10, 1174– 1187.
Conference on Mass Spectrometry and Allied Topics, 53. Richmond, R.; Gorlach, E.; Seifert, J.M. J. Chromatogr. A
Orlando, Florida, 1998; 283. 1999, 835, 29– 39.
28. Wang, T.; Cohen, J.; Kassel, D.B.; Zeng, L. Comb. Chem. 54. Allen, M.C.; Shah, T.S.; Day, W.W. Pharm. Res. 1998, 15,
High Throughput Screen 1999, 2, 327–334. 93 – 97.
29. de Biasi, V.; Haskins, N.; Organ, A.; Bateman, R.; Giles, 55. Beaudry, F.; Yves Le Blanc, J.C.; Coutu, M.; Brown, N.K.
K.; Jarvis, S. Rapid Commun. Mass Spectrom. 1999, 13, Rapid Commun. Mass Spectrom. 1998, 12, 1216– 1222.
1165– 1168. 56. Ackermann B.L.; Ruterbories K.J.; Hanssen B.R.; Lind-
30. Van Hijfte, L.; Marciniak, G.; Froloff, N. J. Chromatogr. B strom, T.D. Proceedings of the 46th ASMS Conference on
Biomed. Sci. Appl. 1999, 725, 3 – 15. Mass Spectrometry and Allied Topics, Orlando, Florida,
31. Weller, H.N. Mol. Divers. 1998-99, 4, 47 –52. 1998; 16.
32. Weller, H.N.; Young, M.G.; Michalczyk, S.J.; Reitnauer, 57. Lopez, L.L.; Yu, X.; Cui, D.; Davis, M.R. Rapid Commun.
G.H.; Cooley, R.S.; Rahn, P.C.; Loyd, D.J.; Fiore, D.; Mass Spectrom. 1998, 12, 1756– 1760.
Fischman, S.J. Mol. Divers. 1997, 3, 61 – 70. 58. Perchalski, R.J.; Yost, R.A.; Wilder, B.J. Anal. Chem.
33. Zeng, L.; Wang, X.; Wang, T.; Kassel, D.B. Comb. Chem. 1982, 54, 1466 –1471.
High Throughput Screen 1998, 1, 101–111. 59. Lee, M.S.; Yost, R.A.; Perchalski, R.J. Annu. Rep. Med.
34. Davis, R.G.; Anderegg, R.J.; Blanchard, S.G. Tetrahedron Chem. 1986, 21, 313– 321.
1999, 55, 1653– 1667. 60. Kerns, E.H.; Rourick, R.A.; Volk, K.J.; Lee, M.S.
35. Regnier, F.E. Nature 1991, 350, 634– 635. J. Chromatogr. B 1997, 698, 133– 145.
36. Afeyan, N.B.; Fulton, S.P.; Regnier, F.E. J. Chromatogr. 61. Kerns, E.H.; Volk, K.J.; Hill, S.E.; Lee, M.S. J. Rapid
1991, 544, 267– 279. Commun. Mass Spectrom. 1995, 9, 1539– 1545.
37. Schriemer, D.C.; Hindsgaul, O. Comb. Chem. High 62. Lee, M.S.; Klohr, S.E.; Kerns, E.H.; Volk, K.J.; Leet, J.E.;
Throughput Screen 1998, 1, 155– 170. Schroeder, D.R.; Rosenberg, I.E. J. Mass Spectrom. 1996,
38. Kaur, S.; McGuire, L.; Tang, D.; Dollinger, G.; Huebner, V. 31, 1253– 1260.
J. Prot. Chem. 1997, 16, 505– 511. 63. Williams, A.; Lee, M.S.; Lashin, V. Spectroscopy 2001, 16,
39. Geysen, H.M.; Wagner, C.D.; Bodnar, W.M.; Markworth, 38 – 49.
C.J.; Parke, G.J.; Schoenen, F.J.; Wagner, D.S.; Kinder, 64. Qin, X.Z.; Ip, D.P.; Chang, K.H.; Dradransky, P.M.;
D.S. Chem. Biol. 1996, 3, 679– 688. Brooks, M.A.; Sakuma, T. J. Pharm. Biomed. Anal. 1994,
40. Hughes, I. J. Med. Chem. 1998, 41, 3804–3811. 12, 221– 233.
41. Wagner, D.S.; Markworth, C.J.; Wagner, C.D.; Schoenen, 65. Rourick, R.A.; Volk, K.J.; Kerns, E.K.; Lee, M.S. J. Pharm.
F.J.; Rewerts, C.E.; Kay, B.K.; Geysen, H.M. Comb. Chem. Biomed. Anal. 1996, 14, 1743– 1752.
High Throughput Screen 1998, 1, 143–153. 66. Fouda, H.; Nocerini, M.; Schneider, R.; Gedutis, C. J. Am.
42. van Breeman, R.B.; Huang, C.-R.; Nikolic, D.; Woodbury, Soc. Mass Spectrom. 1991, 2, 164–167.
C.P.; Zhao, Y.-Z.; Venton, D.L. Anal. Chem. 1997, 69, 67. Wang-Iverson, D.; Arnold, M.E.; Jemal, M.; Cohen, A.I.
2159– 2164. Biol. Mass Spectrom. 1992, 21, 189– 194.
ENCL 100200012—8/10/2001——00012

Spectroscopic Methods of Analysis—Mass Spectrometry 2561

68. Ayrton, J.; Dear, G.J.; Leavens, W.J.; Mallett, D.N.; Plumb, 75. Chang, J.P.; Kiehl, D.E.; Kennington, A. Rapid Commun.
R.S. Rapid Commun. Mass Spectrom. 1997, 11, Mass Spectrom. 1997, 11, 1266– 1270.
1953– 1958. 76. Tiller, P.R.; El Fallah, Z.; Wilson, V.; Juysman, J.; Patel, D.
69. Kaye, B.; Herron, W.J.; Macrae, P.V.; Robinson, S.; Rapid Commun. Mass Spectrom. 1997a, 11, 1570– 1574.
Stopher, D.A.; Venn, R.F.; Wild, W. Anal. Chem. 1996, 68, 77. Schultz, G.A.; Corso, T.N.; Prosser, S.J.; Zhang, S. Anal.
1658– 1660. Chem. 2000, 72, 4058– 4063.
70. Olah, T.V.; McLoughlin, D.A.; Gilbert, J.D. Rapid 78. Li, J.; Wang, C.; Kelly, J.F.; Harrison, D.J.; Thibault, P.
Commun. Mass Spectrom. 1997, 11, 17 – 23. Electrophoresis 2000, 21, 198– 210.
71. Ayrton, J.; Plumb, R.; Leavens, W.J.; Mallett, D.; Dickins, 79. Li, J.; Kelly, J.F.; Chernushevich, I.; Harrison, D.J.;
M.; Dear, G.J. Rapid Commun. Mass Spectrom. 1998, 12, Thibault, P. Anal. Chem. 2000, 72, 599– 609.
217– 224. 80. Holt, R.M.; Newman, M.J.; Pullen, F.S.; Richards, D.S.;
72. Dear, G.J.; Fraser, T.J.; Patel, D.K.; Long, J.; Pleasance, S. Swanson, A.G. J. Mass Spectrom. 1997, 32, 64 – 70.
J. Chromatogr. A 1998, 794, 27 – 36. 81. Shockcor, J.P.; Unger, S.E.; Savina, P.; Nicholson, J.K.;
73. Johnson, J.V.; Yost, R.A. Anal. Chem. 1985, 57, Lindon, J.C. J. Chromatogr. B Biomed. Sci. Appl. 2000,
758A– 768A. 748, 269– 279.
74. Kusmierz, J.J.; Sumrada, R.; Desiderio, D.M. Anal. Chem. 82. Moy, F.J.; Haraki, K.; Mobilio, D.; Walker, G.; Powers, R.;
1990, 62, 2395 –2400. Tabei, K.; Tong, H.; Siegel, M.M. Anal. Chem. in press.
ENCL 100200012—8/10/2001——00012

2562 Spectroscopic Methods of Analysis—Mass Spectrometry

Author Queries
JOB NUMBER: 100200012
JOURNAL: 2820-3

Q1 Reference No.12 and 14 not in MSS. Author


please check.

Вам также может понравиться