applications of various ms

Upload: gvikrant

Post on 30-May-2018

219 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/14/2019 Applications of Various MS

    1/18

    SPECTROSCOPIC METHODS OF ANALYSISMASS SPECTROMETRY

    Mike S. Lee

    Milestone Development Services, Newtown, Pennsylvania

    INTRODUCTION

    The dramatically increased expenditures for both in-house

    and outsourced pharmaceutical research and development

    (R&D) have led to a greater dependence on technology.

    New technologies are constantly introduced into drug

    development to address throughput issues and improve

    development cycles. The incorporation of new technol-

    ogies has resulted in fundamental change in the drug

    development paradigm. Recently, sample generating-

    based technologies such as high throughput biomolecularscreening and automated parallel synthesis have shifted

    the bottleneck to sample analysis-based technologies.

    The current focus on analytical techniques in the

    pharmaceutical industry emphasizes four primary figures

    of merit: sensitivity; selectivity; speed; and high

    throughput. Mass spectrometry (MS) provides each of

    these key attributes, and therefore, has been benchmarked

    an effective solution for pharmaceutical analysis in each

    stage of drug development (1). Perhaps more enabling than

    the MS-based technology itself is the diverse applications

    of MS in conjunction with sample preparation, chromato-

    graphic separation, and informatics. It is within thiscontext that MS has played an increasingly vital role in the

    pharmaceutical industry and has become the preferred

    analytical method for trace-mixture analysis (Fig. 1F1 ).

    A variety of MS formats are widely accepted and

    applied in the pharmaceutical industry. The specific MS

    application is often defined by the sample introduction

    technique. The pharmaceutical applications highlighted in

    this article feature two types of sample introduction

    techniques: dynamic and static. Dynamic sample intro-

    duction involves the use of high-performance liquid

    chromatography (HPLC) on-line with MS. The resulting

    liquid chromatography/mass spectrometry (LC/MS) for-

    mat provides unique and enabling capabilities for

    pharmaceutical analysis. The electrospray ionization

    (ESI) (2) and atmospheric pressure chemical ionization

    (APCI) (3) modes are the most widely used. Static sample

    introduction techniques primarily use matrix-assisted laser

    desorption/ionization (MALDI) (4).

    The advances in MS instrumentation (5) and role of MS

    within the pharmaceutical industry (1) have been recently

    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

    applications and describe the significant integration of this

    technology into drug development. A detailed and in-

    depth overview of current MS technologies and appli-

    cations can be obtained from the recent proceedings of the

    American Society for Mass Spectrometry Conference on

    Mass Spectrometry and Allied Topics (www.asms.org)

    and the Association of Biomolecular Resource Facilities

    (www.abrf.org).

    GENOMICS

    Though the contributions of MS have been somewhat

    limited in the field of genomics, there has been increased

    participation and interest (6). Certainly, the worldwide

    recognition received from the Human Genome Project

    created a sense of urgency toward determining genetic

    variation.

    Genomics refers to the study of genetic data to draw

    correlation between individual genetic inheritance andmedically or biologically important parameters. For

    example, these parameters may involve a patients

    response to a specific drug. Knowledge of the genetic

    basis of individual drug response may provide under-

    standing of the observed variability in drug response

    arising as a result of genetically determined differences in

    drug absorption, disposition, metabolism, or excretion (7).

    Furthermore, knowledge of genetic variation may be

    useful during the target selection process when multiple

    targets are available within a specific disease state. Thus,

    the pharmaceutical industry has great interest in

    determining the genetic variation in patient populations.

    Due to the existence of sequence variations, or

    polymorphisms, no two human genomes are identical.

    Single nucleotide polymorphisms (SNPs) are the most

    abundant genetic variation with an estimated frequency of

    1 SNP per 500 basepairs. Since SNPs are so prevalent in

    the genome, they can act as markers that are linked with a

    phenotype to provide a comprehensive measure of

    interaction with a specific drug. The validation of a

    ENCL 1002000128/10/200100012

    Encyclopedia of Pharmaceutical Technology

    Copyright q 2002 by Marcel Dekker, Inc. All rights reserved. 2545

  • 8/14/2019 Applications of Various MS

    2/18

    particular SNP represents an important stage for

    establishing SNPs as a routine clinical diagnostic marker.

    The validation of SNPs via MALDI-TOF MS is emergingas a valuable genotyping tool (8,9). A schematic of a

    MALDI-TOF MS instrument is shown in Fig. 2F2 .

    A recent study performed by Stroh et al. (10) compared

    the performance of MALDI-TOF MS with restriction

    fragment length polymorphism (RFLP) and fluorescence

    polarization (FP). The study involved the analysis of

    known mutations of the IL-1b gene. The procedure

    involved amplification of patient DNA samples using

    standard PCR techniques followed by a primer extension

    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

    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, theassay can be designed to extract complete information

    about the SNP regardless of its state. This emerging MS-

    based approach for SNP genotyping has potential to be

    highly automated without the requirement for fluorescent

    tags. Furthermore, multiplexing can be attained by

    selecting post-PCR primers of varying lengths to dedicate

    predetermined regions of the mass spectrum to specific

    SNPs.

    PROTEOMICS

    The study of protein structure, function, quantity, and

    interactions during maturation and progression of disease

    is referred to as proteomics. Analytical approaches that use

    a combination of two-dimensional (2-D) gel electrophor-

    esis for protein separation and MS analysis for protein

    identification followed by database searches is a widely

    practiced proteomics strategy (11). The tryptic peptides

    extracted from gels are analyzed by MALDI-TOF MS and

    microcolumn or capillary LC tandem mass spectrometry

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

    Fig. 1 Structure analysis matrix that illustrates pharmaceutical

    analysis preferences for four specific sample types: nontrace/-

    pure; nontrace/mixture; trace/pure; and trace/mixture. (Courtesy

    of Milestone Development Services, Newtown, PA.)

    X-ray

    NMR

    MS

    UV

    IR

    HPLC/UV

    LC/MS

    LC/NMR

    Trace

    Non-

    Trace

    Pure Mixture

    ENCL 1002000128/10/200100012

    Spectroscopic Methods of AnalysisMass Spectrometry2546

  • 8/14/2019 Applications of Various MS

    3/18

    (MS/MS) techniques. Typically, the MALDI-TOF MS

    techniques are used to quickly identify peptide fragments

    and confirm the presence of known proteins. Nano-scale

    capillary LC/MS/MS techniques (using 50 100 mmdiameter columns, operating at flow rates of 20 500

    nL/min) are used to further interrogate the complex protein

    mixture at the low femtomole level. These techniques

    require the use of a specialized ESI source shown in Fig. 3F3 .

    The combination of MALDI-TOF MS and capillary

    LC/MS/MS was recently described for the identification of

    disease state markers in human urine (12). In this study,

    urine proteins obtained from emphysema patients were

    separated on 2-D gels and selected spots were digested

    with trypsin and analyzed by MALDI-TOF. A database

    search using Protein Prospector identified a potential

    biomarker for emphysema as human alpha-1-antitrypsin

    (A1AT). The corresponding MALDI spectrum contained

    nine out of 18 peptides with masses that match the

    expected tryptic digest fragments for A1AT.

    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

    SEQUEST.F4 Fig. 4 illustrates the components of an ion

    trap mass spectrometer. The highly automated data-

    dependent MS/MS analysis provided excellent

    sequence coverage for 11 tryptic peptides related to

    A1AT in a single LC/MS run. A tryptic peptide that

    corresponds to the A1AT sequence SVLGQLGITKobserved at retention time (rt) 30.5 min. was observed

    in the spectrum. The LC/MS data also provided

    sequence information on unmatched MALDI peaks.

    The need to detect lower concentration of protein and

    peptide mixtures has resulted in the increased use of

    hybrid quadrupole/orthogonal TOF (QTOF) MS/MS

    instruments (13) in conjunction with microcolumn LC.

    Fig. 5F5 shows a schematic of a QTOF instrument. This

    LC/MS approach provides a resolution of ca. 0.1 mass

    units allowing for the analysis of complex product ion

    spectra (14,15). A recent publication by Chalmers and

    Gaskell highlights the current challenges in proteome

    analysis with regard to MS instrumentation (16).

    NATURAL PRODUCTS DEREPLICATION

    Historically, an excellent source of novel lead drug

    compounds is natural products. Natural product screening

    activities typically occur during drug discovery and

    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.)

    ENCL 1002000128/10/200100012

    Spectroscopic Methods of AnalysisMass Spectrometry 2547

  • 8/14/2019 Applications of Various MS

    4/18

    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-

    ution is retained.

    The key to natural products analysis using this approach

    is dependable molecular weight determination. This

    information is used with existing natural product databases

    that contain information on the bioactive compounds, the

    physical descriptions of the microorganisms from which

    they come, their spectrum of activity, the method of

    extraction and isolation, and physical data (i.e., molecular

    weight, UV absorption maxima). Molecular weight is the

    Fig. 4 Schematic of an ion trap MS instrument. This device

    consists of two endcap electrodes (entrance and exit) and a ring

    electrode. An ion trap MS separates ions based on mass-to-chargeratio (m/z). Once ions are introduced into the ion trap MS, the

    radiofrequency (rf) amplitude is increased so that ions are

    sequentially ejected (by increasing mass) and detected. This type

    of MS provides a routine (i.e., benchtop) and sensitive detector

    using either GC and LC interfaces. Furthermore, this instrument

    provides a unique format for multiple stages of MS analysis

    (MSn). (Courtesy of ThermoFinnigan, San Jose, CA.)

    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 1002000128/10/200100012

    Spectroscopic Methods of AnalysisMass Spectrometry2548

  • 8/14/2019 Applications of Various MS

    5/18

    most critical information for initial searches because of its

    link to structural specificity. This information is used to

    make pivotal decisions on whether or not to proceed to

    more time-consuming isolation steps based on novelty of

    the compound. In recently reported studies (17,18),

    LC/MS is used to increase sensitivity and accelerate

    analysis. These features serve to significantly reduce labor.A recent review described LC/MS-based approaches for

    the characterization of natural product mixtures in

    conjunction with high-throughput screening (19).

    The instrumental configuration of the LC/MS system

    developed by Ackermann et al. (17) features an HPLC, UV

    detector, fraction collector, ESI-tandem quadrupole

    (MS/MS), and MALDI-TOF MS. Filtered fermentation

    broths were extracted with butanol or ethyl acetate, and

    eluted on a gradient C18 reversed-phase HPLC separation.

    The eluent was split 1:10 between a tandem quadrupole

    MS/MS instrument (scanning 2502000 amu/3 sec in the

    full scan mode) and a single wavelength UV detector (254

    or 230 nm). One-min fractions were collected after the UV

    detector. Of these fractions, 2050 mL is used for

    MALDI-TOF analysis, and the remainder is concentrated

    for microbiological testing. The LC/UV chromatogram

    was compared to the bioactivity assay histogram to

    highlight the peaks that contain activity. The molecular

    weights of the active peaks were obtained for novelty

    assessment of the compounds.

    Similar approaches that use on-line LC/MS and

    LC/MS/MS techniques have been recently described for

    natural products dereplication (20,21). Approximately,000

    natural product extracts can be screened annually for in

    vivo and in vitro activity using LC/MS-based systems. Astandard approach for dereplication involves a comparison

    of retention time, full scan mass spectra (i.e., molecular

    weight information), and MS/MS spectra with those from

    known biologically active standards. Thus, previously

    identified components are rapidly eliminated and do not

    require time-consuming structure elucidation studies. The

    savings of effort allow researchers to focus efforts on

    novel chemistries. Samples of novel compounds can then

    be infused into an ion trap mass spectrometer, and a

    multiple stage mass analysis (MSn) fragmentation map is

    generated.

    COMBINATORIAL CHEMISTRY

    Recent reviews (22,,25) describe MS-based methods

    ranging from the analysis of complex molecular libraries

    (26) to open-access formats for drug discovery and

    development (27). High throughput criteria were central to

    each application.

    An important development in the quest for high

    throughput combinatorial library analysis was the multiple

    ESI interface described by Wang et al. (28). This novel

    ESI interface enabled effluent flow streams from an array

    of four HPLC columns to be sampled independently and

    sequentially using a quadrupole MS instrument. The

    interface featured a stepping motor and rotating plateassembly. The effluent flow from the HPLC columns was

    connected to a parallel arrangement of electrospray

    needles co-axial to the mass spectrometer entrance

    aperture. The individual spray tips were positioned 908

    relative to one another in a circular array. Each spray

    position was sampled multiple times per second by precise

    control of the stepping motor assembly.

    The parallel sample analysis format using a multiplexed

    LC/MS interface with an orthogonal time-of-flight (TOF)

    MS was described by de Biasi et al. (29). This approach

    illustrated the high-throughput capabilities of a multi-

    plexed ESI interface in combination with an MS format

    that accommodates fast chromatography methodologies.

    The system featured a four-way multiplexed electrospray

    interface attached directly with the existing source of the

    TOF-MS instrument. A rotating aperture driven by a

    variable speed stepper motor permitted the sampling of the

    spray from each electrospray probe tip. The data files were

    synchronized with the corresponding spray.

    As the preparation of large libraries for lead discovery

    became routine, the burden placed on analysis techniques

    focused mainly on throughput and quality (30). However,

    biological assay requirements typically required pure

    compounds. Thus, the focus shifted toward the use of

    automated high throughput purification methods applied tolibraries of discrete compounds (31). Reverse-phase

    analytical and preparative HPLC formats in conjunction

    with MS techniques have been critical for the high

    throughput purification approaches for parallel synthesis

    libraries. A variety of approaches that featured the use of

    gradient methods, short columns, and high flow rates have

    been described (32). Highly automated LC/MS approaches

    for purification at the multimilligram level were described

    for a quadrupole system by Zeng et al. (33). These

    methods featured the use of short columns that were

    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

    ml/min). Analytical LC/MS analyses of compound

    libraries were achieved in 5 min for chromatographically,

    well-behaved compounds. Slightly longer preparative

    LC/MS analysis times (810 min/sample) were required

    for compounds that exhibited poor chromatographic peak

    shapes and/or for compound mixtures the required higher

    resolution separations. The fraction collection process is

    ENCL 1002000128/10/200100012

    Spectroscopic Methods of AnalysisMass Spectrometry 2549

  • 8/14/2019 Applications of Various MS

    6/18

    initiated in real-time once the reconstructed ion current is

    observed for a specific m/z value that corresponds to the

    compound of interest. This design permitted the collection

    of one sample per fraction. Thus, the need for very large

    fraction collector beds and postpurification screening and

    pooling was eliminated. Unattended and automated

    operation of this system led to the purification ofovercompounds (mg quantities) per day.

    BIOAFFINITY SCREENING

    With the integration of highly automated parallel synthesis

    techniques into drug discovery programs, hundreds of

    thousands of compounds are now screened against a

    particular biological target. Once activity is determined for

    a mixture, the identification of the active component(s) is

    necessary. A recent study described the use of bioaffinity

    selection LC/MS methods for the identification of active

    mixture component(s) (34). This approach features an

    integrated bioaffinity-based LC/MS screening method to

    separate and identify compounds from mixtures.

    A mixture of compounds is incubated with the target

    protein and the components bound to the protein are

    selected by using a size exclusion chromatography (SEC)

    spin column. In this experiment, the unbound com-

    pounds are retained on the column. The bound components

    are eluted and identified with LC/MS. Increased specificity

    is obtained by dissociating the bound compounds and

    performing a second equilibration incubation with the

    protein. This procedure preferentially selects for the

    compounds with higher affinity, and results in anenhancement of the quantitative LC/MS response.

    Iterative stages of incubation, size-exclusion, and

    LC/MS allow the tighter binding components to be

    enriched relative to weaker binding components.

    In this study, the peroxisome proliferator-activated

    receptor (PPARg), which is a target for anti-diabetic drugs

    (construct molecular weight of 32,537 Da), is incubated

    with 10 ligands that range in molecular weight from 283 to

    587 units. A spin column of 6000 Da cutoff is used for

    SEC purposes. The retained mixture of components is

    analyzed by fast perfusive chromatography (35,36), using

    a standard full-scan LC/MS strategy. This analysis

    procedure allows for the identification and quantitation

    of the protein and the ligands, compared to their responses

    prior to incubation. The ligand-protein complex that

    dissociated under the reversed-phase chromatographic

    conditions is selectively detected.

    This analysis scheme provided a quick measurement of

    binding affinity, and serves as a screening tool during drug

    candidate selection. Spreadsheets were constructed and

    used to calculate the binding affinity of the components. In

    the example described above, two incubation cycles

    followed by the SEC separation provided an enhancement

    of strong binders to weak binders. This LC/MS-based

    method provides a unique approach to obtain information

    in situations when lower concentrations of tighter binding

    ligands are present in the same mixture with higherconcentrations of weaker binding ligands. Furthermore,

    this method is more efficient than synthetic deconvolution

    procedures and does not require the use of radioligands.

    Combinatorial chemistry initiatives have created a

    tremendous challenge for activities that deal with the

    screening of these mixtures for activity against a specified

    target (37). MS-based approaches that use affinity

    selection (38), encoding methodologies (3941), pulsed

    ultrafiltration (42), and anti-aggregatory approaches (43)

    have been described.

    The use of MS formats that provide accurate mass

    capabilities have been recently illustrated for screening

    combinatorial libraries (4447). The unambiguous con-

    firmation/identification of combinatorial library com-

    ponents from small quantities of material have been

    illustrated using a hybrid quadrupole/orthogonal TOF

    (QTOF) (44,45) and Fourier transform ion cyclotron

    resonance (FTICR) (47) mass spectrometry. A schematic

    of a FTICR-MS system is shown in Fig. 6F6 . Accurate

    isotope patterns or isotopic signature and unique mass

    differences between isobaric compounds can be obtained

    using these two MS formats.

    OPEN-ACCESS SYSTEMS

    Chemists now routinely use open-access MS systems in

    the same way that they previously used thin-layer

    chromatography (TLC) to monitor reaction mixtures for

    the desired product and to optimize reaction conditions. In

    practice, medicinal chemists require only molecular

    weight data, and are comfortable with a variety of MS

    ionization methods to obtain this information. However,

    confidence in the actual method and procedure is a

    requisite. Today, molecular mass measurement has

    quickly become a preferred means of structure confir-

    mation over NMR and IR during the early stages of

    synthetic chemistry activities (i.e., drug discovery), where

    sample quantities are limited.

    In the open-access LC/MS procedure described by

    Pullen et al. (48), the samples are directly introduced from

    solution for ease of automation and sample preparation.

    Chemists prepare samples in solvent to a suggested

    concentration range, then log the samples into the system.

    The sample log-in is done at any time during the

    ENCL 1002000128/10/200100012

    Spectroscopic Methods of AnalysisMass Spectrometry2550

  • 8/14/2019 Applications of Various MS

    7/18

    continuous automated queue. Autosampler vials are used

    to hold the samples, and autosamplers are used to directly

    deliver samples in solution to the mass spectrometer. The

    system uses a standard method to analyze the samples in

    queue, average spectra according to a preset scheme, and

    print out a spectrum for the chemist. Fail-safe procedures

    for untrained users and instrument self-maintenance at

    start-up and shutdown were also developed.

    Taylor et al. (49) further demonstrated the value of

    open-access LC/MS systems for generating a widened

    scope of pharmaceutical analysis applications, including

    1) characterization of synthetic intermediates and target

    compounds; 2) reaction monitoring; 3) reaction optimiz-

    ation; 4) analysis of preparative HPLC fractions; and 5)

    analysis of TLC plate spots. The availability of these

    methods led to the increased use of LC/MS for structural

    analysis. The short analysis time and reliable structure

    confirmation resulted in the use of LC/MS as a first choice

    for structure characterization for synthetic chemistry

    applications.

    Open-access LC/MS formats have spawned new

    dimensions in access and data management. The use of a

    direct exposure probe (DEP) for automated sample

    introduction has been developed for quick (ca. 3 min)

    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.)

    ENCL 1002000128/10/200100012

    Spectroscopic Methods of AnalysisMass Spectrometry 2551

  • 8/14/2019 Applications of Various MS

    8/18

  • 8/14/2019 Applications of Various MS

    9/18

    IN VIVO DRUG SCREENING

    The application of APCI-LC/MS techniques for the rapid

    determination of protein binding and pharmacokinetics in

    drug discovery were recently described by Allen et al.

    using a single quadrupole instrument (54). A cocktail

    approach consisted of four experimental compounds and acontrol compound dosed orally at 1 mg/kg with plasma

    samples obtained at 0.5, 1, 2, 4, and 8 h post dose. To

    insure reproducibility, the control compound was tested

    with each cocktail. This approach generated timely

    systemic exposure (AUC and Cmax) data on 44 test

    compounds in three work days, using two laboratory

    scientists.

    The use of LC tandem quadrupole MS/MS-based

    screening approaches for quantitative bioanalytical

    measurements allow a large, chemically diverse, range

    of potential drug candidates to be analyzed quickly and

    confidently. A schematic of a tandem quadrupole MS/MSinstrument is shown in Fig. 8F8 . The development of unique

    LC/MS-based systems for in vivo pharmacokinetic

    screening reduces the analysis to a manageable number

    of samples, and results in a cost-effective approach to

    evaluate new lead compounds. Approaches to this type of

    methodology will likely vary, according to the behavior of

    the molecules of interest, standard operating procedures

    (SOPs), performance capabilities of the mass spec-

    trometer, and integration of automated sample preparation,

    and data analysis procedures. Success will likely be

    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

    multiple drug candidates in one animal has been termed

    n-in-one or cassette dosing. As discussed for the

    previous example, this parallel approach results in an

    increased productivity for bioanalysis during drugdiscovery. Beaudry et al. (55) recently investigated the

    extension of this methodology to study larger numbers of

    compounds in each mixture, and to integrate sample

    preparation with the LC/MS/MS system for increased

    efficiency.

    The number of analytes studied in parallel was extended

    to 63 plus an internal standard. The increased number of

    analytes was made possible due to improvements to the

    collision region of the MS/MS system that provide

    increased sensitivity and reduced memory effects. In

    addition, robotic systems for sample handling and on-line

    (solid phase extraction) SPE of plasma samples were

    integrated with the LC/MS/MS system. An isocratic

    reversed-phase HPLC method provided a cycle time of 4.5

    min per sample. The on-line sample preparation and short

    analysis resulted in an increased sample throughput that

    required less time from the scientist. The method produced

    good performance, in terms of extraction efficiency,

    linearity, and limit of detection (LOD), and has the

    capability of analyzing 320 960 samples per day. The

    strategic emphasis of this approach is on providing high

    throughput LC/MS methods for evaluating large numbers

    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

    clearance is controlled predominantly by phase I oxidative

    metabolism as opposed to phase II metabolism or renal

    clearance. In this way, the resulting data could be resolved

    into four categories of metabolic stability: high ($60%);

    moderate ($3059%); low ($1029%); and very low

    (,10%).

    The rapid structure identification of metabolites is a

    powerful complement to previously described quantitative

    approaches. The utility of an automated metabolite

    identification approach, using LC/MS/MS with an ion

    trap mass spectrometer has been demonstrated (57). In this

    Fig. 8 Schematic of a tandem quadrupole MS/MS instrument.

    A tandem quadrupole MS/MS instrument consists of two

    quadrupole MS filters, MS1 and MS2, separated by a collision

    cell. Each quadrupole MS filter consists of four cylindrical orhyperbolic shaped rods. A unique combination of direct current

    (dc) potential and radiofrequency (rf) potential is applied to each

    pair of rods (one pair 1808 out of phase with the other). A mass

    spectrum results by varying the voltages at a constant rf/dc ratio.

    A variety of scan modes (e.g., full scan, product ion, precursor

    ion, neutral loss) provide unique capabilities for quantitative and

    qualitative structure analysis. (Courtesy of Micromass, Manche-

    ster, UK.)

    ENCL 1002000128/10/200100012

    Spectroscopic Methods of AnalysisMass Spectrometry 2553

  • 8/14/2019 Applications of Various MS

    10/18

    study, MSn analysis is automated to provide maximum

    structural information in combination with predictive

    strategies for biotransformation. Automated data-depen-

    dent scan functions are used to generate full scan, MS/MS,

    and MSn mass spectra of metabolites within a single

    chromatographic analysis. This feature is unique and

    avoids the multiple (2 4) injections that are necessarywith other MS/MS configurations (e.g., tandem quadru-

    pole). Along with the significant savings in time, detailed

    structure information is generated, which enables a

    comprehensive analysis of substructure relationship to be

    constructed for each metabolite. These automated studies

    provide unique advantages during drug discovery, and

    provide an early perspective on the metabolically labile

    sites, or soft spots of a drug candidate. This knowledge

    is useful during lead optimization activities, and can lead

    to the initiation of proactive research efforts that deal with

    metabolism-guided structural modification and toxicity.

    METABOLITE PROFILING AND IDENTIFICATION

    The application of LC/MS-based techniques for the

    structure identification of drug metabolites has played a

    significant role in drug development. The early identifi-

    cation of drug metabolites provides valuable insights into

    the pathways of metabolism and biotransformation. Once

    metabolites are identified/confirmed, metabolism-guided

    structural modification during the drug discovery stage is

    initiated to facilitate the selection of drug candidates for

    subsequent development.The identification of metabolite structures with LC/MS

    and LC/MS/MS techniques using quadrupole-based MS

    instruments are an effective approach due to their ability to

    analyze trace mixtures from complex samples of urine,

    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).

    Therefore, the parent drug and its corresponding

    metabolites would be expected to undergo similar

    fragmentation and to produce mass spectra that indicate

    major substructures.

    Kerns et al. demonstrated the application of LC/MS and

    LC/MS/MS standard method approaches in preclinical

    development for the metabolite identification of buspir-

    one, a widely used anxiolytic drug (60). The success of this

    method relies on the performance of the LC/MS interface

    and the ability to generate abundant ions that correspond to

    the molecular weight of the drug and drug metabolites.

    The production of abundant molecular ions is an ideal

    situation for molecular weight confirmation because

    virtually all the ion current is consolidated into an adduct

    of the molecular ion (i.e., [M 1 H]1, [M 1 NH3]1).

    Full-scan mass spectra generally contain an abundant

    [M 1 H]1 ion signal with little detectable fragmentation.

    Product-ion spectra are obtained to reveal product ions and

    neutral losses that are associated with diagnostic

    substructures of the buspirone molecule. To assist withthe MS/MS structure identification, the gross substructure

    of buspirone is categorized into profile groups (61). Profile

    groups directly correlate specific product ions and neutral

    losses with the presence, absence, substitution, and

    molecular connectivity (62) of specific buspirone sub-

    structures and their modifications. The profile groups of

    buspirone are identified with abbreviations that correspond

    to the three specific substructures: azaspirone decane

    dione (A), butyl piperazine (B), and pyrimidine (P).

    Substituted substructures are designated with a subscript

    (s), and a dash ( ) denotes substructure connectivity.

    Thus, the buspirone molecule is represented by ABP.

    The AsBP designation refers to metabolite structures

    that contain the azaspirone decane dione, butyl piperazine,

    and pyrimidine substructures with substitution on the

    azaspirone decane dione substructure. The profile group

    categorization within a corresponding database allows the

    rapid visual recognition of primary substructures affected

    by metabolism.

    Metabolite structure databases can be easily constructed

    and contain information on the structure, molecular

    weight, UV characteristics, RRT, and product ions of

    metabolites obtained from rat bile, urine, and liver S9

    samples. Using this format, Kerns et al. reported the

    predominant buspirone metabolite profile groups as AsB P, A B Ps, and As B Ps. These profile groups

    indicate azaspirone decane dione and pyrimidine as

    metabolically active sites of attack and the presence of

    multiple substitution sites on each of these substructures.

    IMPURITY PROFILING AND IDENTIFICATION

    Synthetic impurities are of particular concern during

    process research and safety evaluation activities. Often,

    impurities are the result of synthetic by-products or

    starting materials of the scale-up process. Impurities

    provide a comprehensive indicator of the chemical process

    and are diagnostic of overall quality. Process chemists use

    this information to guide process optimization. Knowledge

    of the identity and relative amount of impurities is used to

    diagnose process reactions so that changes in reagents and

    reaction conditions leads to better yields and higher quality

    material.

    ENCL 1002000128/10/200100012

    Spectroscopic Methods of AnalysisMass Spectrometry2554

  • 8/14/2019 Applications of Various MS

    11/18

    With an increasing number of novel lead candidates that

    enter into preclinical development, considerable resources

    are needed to identify impurities. LC/MS-based

    approaches provide integrated sample clean-up and

    structure analysis procedures for the rapid analysis of

    impurities. This advantage was demonstrated during the

    preclinical development of TAXOLw

    (18). LC/MS playedan important role for the identification of impurities

    contained in extracts and process intermediates from Taxus

    brevifolia and T. baccata biomass. Because drugs derived

    from natural sources often have a very diverse set of

    structural analogs, it is important to determine which

    analogs are carried through the purification process and

    ultimately appear as impurities. This task presents a unique

    challenge during the early stages of drug development due

    to the highly complex nature of the samples.

    Kerns and coworkers described a structure identification

    strategy that incorporates LC/MS and LC/MS/MS

    techniques using quadrupole-based instruments for rapid,

    sensitive, and high-throughput impurity analysis (18). This

    approach integrates traditional steps of sample prep-

    aration, separation, analysis, and data management into a

    single instrumental method. The resulting multidimen-

    sional data include retention time, molecular weight, UV,

    and substructure information. A structure database is

    developed for each candidate and is used to rapidly

    identify the same impurities in new samples. Structures are

    proposed based on using the drug candidate as a structural

    template and, with the use of a standard method approach,

    consistency for comparison of results throughout the

    preclinical development process is ensured.

    Nearly all of the impurities contained the characteristicpaclitaxel core substructure as indicated by the character-

    istic product ion at m/z 509 with variations due to

    modifications. Many of these taxanes contained a side-

    chain similar to paclitaxel, with variations occurring on the

    terminal amide of the side chain. The product ions that

    differed from the characteristic side-chain ions of

    paclitaxel (m/z 286) by values indicative of specific

    substructures were used to identify these terminal amide

    variations. A comparison with the paclitaxel substructural

    template indicated structural differences beyond the

    position of the amide group in the side-chain substructure.

    When a new impurity was encountered during chemical

    process research, retention time and molecular weight

    information were compared to the database for rapid

    identification. This approach is similar to the procedure

    described for natural product dereplication. If the

    compound is not contained in the structure database,

    then the corresponding LC/MS/MS analysis is performed

    to obtain substructural detail and the proposal of a new

    structure.

    A standard reversed-phase HPLC method was used for

    all the samples that are associated with a drug candidate to

    reduce time-consuming method development/method

    refinement procedures. Standard reversed-phase methods

    typically involve a 2030 min cycle time and provide

    information for a wide range of compounds. The

    incorporation of a standard method strategy allows theuse of autosampling procedures and standard system

    software for data analysis.

    During the development of TAXOLw, 90 taxane

    impurities were rapidly identified and added to the

    structure database. This MS/MS information was routinely

    obtained for impurities down to the 100 ng level (injected),

    and required approximately 23 h for the analysis of each

    sample. The compounds are structurally categorized with

    profile group terminology. The LC/MS-based methods

    were significantly faster than the previously used

    analytical methods based on scale-up, isolation, fraction-

    ation, and individual structural analysis. Software tools

    capable of sample tracking, interpretation, and data

    storage facilitate the structure profiling of impurities,

    degradants, and metabolites (63). Key pharmaceutical

    analysis elements that deal with sample preparation, real-

    time analysis decisions, databasing, distribution/visualiza-

    tion of results (Fig. 9F9 ) and prediction of fragmentation are

    now highly integrated.

    DEGRADANT PROFILING AND IDENTIFICATION

    During the course of drug development, the bulk drug and

    drug formulation are studied under a variety of stressconditions such as temperature, humidity, acidity, basicity,

    oxidization, and light. Qin et al. described the utilization of

    stressing conditions that may cause degradation (64). The

    resulting samples may be used to validate analytical

    monitoring methods and to serve as predictive tools for

    future formulation and packaging studies.

    A traditional approach to study degradant formation

    involves similar time-consuming scale-up and preparation

    steps as described for metabolite and impurity analysis.

    Similarly, this area of pharmaceutical analysis has

    experienced the issues associated with faster drug

    development cycles. Rourick and coworkers recently

    described proactive approaches to obtain degradant

    information with quadrupole LC/MS methods during the

    preclinical development stage (65). The corresponding

    structural information provides insight for decisions on

    which leads to further develop for clinical testing. The

    early structural information on degradants of a drug

    candidate offers a unique capability for synthetic

    modification to minimize degradation. Structural infor-

    ENCL 1002000128/10/200100012

    Spectroscopic Methods of AnalysisMass Spectrometry 2555

  • 8/14/2019 Applications of Various MS

    12/18

    mation can also facilitate planning of preclinical drugdevelopment in process research, formulation develop-

    ment, and safety assessment.

    The strategy for impurity and degradant identification

    described by Rourick et al. subjects lead candidates to

    various development conditions followed by LC/MS and

    LC/MS/MS analysis protocols. A structure database is

    constructed from the corresponding results and is used to

    reveal unstable regions within the drug structure as well as

    to ascertain which candidate or homologous series of drug

    candidates may be the most favorable for further

    development. High capacity and throughput speed are

    necessary so that many lead candidates may be evaluated.

    Applicability of the method to a wide range of compound

    classes is desirable. Once the drug candidate enters clinical

    development and manufacturing, the structure database is

    useful for the rapid identification of impurities and

    degradants in samples generated during these stages of

    development.

    The method exposes drug candidates to forced

    degradation conditions, (e.g., acid, base, heat, and

    moisture) as a predictive profile. The coordinated use ofLC/MS and LC/MS/MS provide structure identification

    for speed, sensitivity, and high throughput. Standard

    methods, useful for 80% of the compounds, are applied.

    Various types of structural data are obtained for

    elucidation purposes (e.g., retention time, molecular

    weight, MS/MS), and unknown compounds are elucidated

    with the candidate drug as a structural template. The

    LC/MS analysis provides retention time and molecular

    weight data, whereas LC/MS/MS provides substructural

    detail for structure identification. Drug candidates are

    incubated under drug processing, storage, and physiologi-

    cal conditions that were expected to occur throughout drug

    lifetime.

    Using this approach, 10 degradants of cefadroxil, an

    orally effective semisynthetic cephalosporin antibiotic,

    were elucidated in a 2-day study. The use of standard

    LC/MS methods provided consistency from sample to

    sample throughout the development process, and allowed

    for the construction and use of a structural database for the

    rapid identification of impurities and degradants during

    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.)

    ENCL 1002000128/10/200100012

    Spectroscopic Methods of AnalysisMass Spectrometry2556

  • 8/14/2019 Applications of Various MS

    13/18

    development. The reversed-phase HPLC conditions

    provided a general measure of the polarity of each

    compound, useful for interpretation of substructural

    differences between related compounds. Due to the

    mass-resolving capability of the mass spectrometer,

    chromatographic resolution of co-eluting or unresolved

    components was not required. Abundant protonatedmolecule ions, [M 1 H]1, provided reliable molecular

    weight information, and product-ion spectra generated

    valuable substructure information for each degradant. The

    product-ion spectrum of cefadroxil was used as a template

    for interpretation; specific product ions and neutral losses

    were compared to the spectra obtained from the unknown

    degradants. Product ions common to each spectrum

    provided evidence of substructures unchanged by the

    degradation conditions and differences were indicative of

    structural variations.

    QUANTITATIVE BIOANALYSISSELECTED IONMONITORING

    The quantitative analysis of targeted components in

    physiological fluids is a major requirement in clinical

    development. In 1991, Fouda et al. (66) pioneered the use

    of APCI-LC/MS on a single quadrupole instrument for the

    quantitative determination of the renin inhibitor, CP-

    80,794, in human serum. Because the pharmacological

    action is below 200 pg/ml, a quantitative assay in the low

    pg/ml range was required to monitor the drugs

    pharmacokinetic and pharmacodynamic properties. Also,

    the structure of the CP-80,794 molecule lacked asignificant chromophore for UV detection with conven-

    tional HPLC methods. Furthermore, the low volatility and

    thermal instability precluded analyses with GC/MS

    methods.

    Quantitative LC/MS assays in clinical development

    generally involve four intensive steps: sample prep-

    aration; assay calibration; sample analysis; and data

    management. In the method developed by Fouda and

    coworkers, human serum samples were prepared with a

    liquid liquid extraction procedure. Assay calibration

    involved the use of human serum samples fortified with

    CP-80,794 at 11 concentrations (6 replicates per

    concentration) ranging from 0.05 to 10 ng/ml. The

    LC/MS analysis involved the use of the SIM mode to

    monitor the molecular ions[MH]2 that correspond to the

    drug (m/z 619) and internal standard (m/z 633). In this

    particular LC/MS application, the negative ion mode was

    highly sensitive for this class of compound. Samples were

    loaded onto an HPLC autosampler and 80 mL aliquots are

    injected onto the column at 4-min intervals. The elution

    times of the drug and internal standard were 3.1 and 3.4

    min, respectively.

    At the time, this application provided a powerful

    benchmark for the use of quadrupole LC/MS-based

    methods in the pharmaceutical industry and paved the

    way for the tremendous growth of MS-based applications

    in support of clinical development. This particular assaysuccessfully supported several clinical studies with

    sensitive and reliable results. This performance was

    benchmarked on more than 4000 clinical samples, and led

    to a widened scope of MS application for quantitative

    bioanalysis (6771).

    QUANTITATIVE BIOANALYSISSELECTED

    REACTION MONITORING

    The use of selected reaction monitoring (SRM) methods

    for quantitative bioanalysis represents increased dimen-

    sions of mass spectrometry analysis. A SRM method that

    features a tandem quadrupole MS/MS instrument for the

    quantitative analysis of an antipsychotic agent, clozapine,

    in human plasma was recently described by Dear et al.

    (72). Preclinical development studies of clozapine in rats

    and dogs used HPLC with fluorescence detection (FLD).

    With this method, a better limit of quantitation (LOQ) of 1

    ng/ml was obtained. As the compound moved into the

    clinical stages of development, a more sensitive method of

    analysis was required to obtain rapid metabolic infor-

    mation in support of drug safety evaluation studies. As a

    result, a standard LC/MS/MS method was developed forthe quantitative analysis of clozapine (I) and four

    metabolites (II-V) in human plasma.

    The LC/MS/MS strategy deployed is similar to

    previously described approaches for protein, natural

    products, metabolite, and impurity identification. An

    ionization technique that generates abundant molecular

    ion species with very little fragmentation is desirable. The

    product-ion spectrum is obtained to generate the

    substructural template of the molecule. Abundant and

    structurally unique transitions (molecular ion ! product

    ion) are identified from the spectrum, and are used in the

    corresponding SRM experiment for quantitation. The

    SRM experiment provides a high degree of selectivity and

    better LOD than full-scan or SIM experiments for the

    analysis of complex mixtures (73,74). The selectivity of

    MS/MS reduces the requirements for complete chromato-

    graphic resolution of each component. Therefore,

    LC/MS/MS experiments for quantitation typically

    emphasize short analytical run times to provide high

    sample throughput.

    ENCL 1002000128/10/200100012

    Spectroscopic Methods of AnalysisMass Spectrometry 2557

  • 8/14/2019 Applications of Various MS

    14/18

  • 8/14/2019 Applications of Various MS

    15/18

    expand current approaches as well as create low-flow,

    miniaturized formats (7779) are envisioned. Continued

    advances in integrated approaches that feature MS will

    generate significant interest with regard to pharmaceutical

    analysis. For example, the use of integrated systems that

    feature NMR/MS have been described for applications that

    involve LC/NMR/MS (80,81) and MS/NMR (82). A

    schematic of an LC/NMR/MS system is shown in Fig. 10F10 .

    In these configurations, NMR is used to provide structural

    and stereochemical information (80,81) or to verify

    binding/interaction (82) while MS is used to provide

    molecular weight information. New challenges that deal

    specifically with probing mechanism of action will likely

    generate the need for a broader application of MS as well

    as spur the development of novel technologies for sample

    preparation, chromatography, and information

    management.

    REFERENCES

    1. Lee, M.S.; Kerns, E.H. Mass Spectrom. Rev. 1999, 18,187279.

    2. Fenn, J.B.; Mann, M.; Meng, C.K.; Wong, S.F.; White-house, C.M. Science 1989, 246, 6471.

    3. Bruins, A.P.; Covey, T.R.; Henion, J.D. Anal. Chem. 1987,59, 26422646.

    4. Hillenkamp, F.; Karas, M.; Beavis, R.C.; Chait, B.T. Anal.

    Chem. 1991, 63, 1193A 1203A.5. Burlingame, A.L.; Boyd, R.K.; Gaskell, S.J. Anal. Chem.

    1998, 70, 647R 716R.

    6. Kelleher, N.L. Chem. Biol. 2000, 7, R37R45.7. Wolf, C.R.; Smith, G. Pharmacogenetics. Br. Medical Bull.

    1999, 55, 366386.

    8. Ross, P.; Hall, L.; Smirnov, I.; Haff, L. Nat. Biotechnol.

    1998, 18, 13471351.9. Haff, L.A.; Smirnov, I.P. Genome Res. 1997, 7, 378388.

    10. Pezzullo, L.II.; Hall, S.K.; Affourtit, J.P.; Seymour, A.B.;Stroh, J.G. Proceedings of the 48th ASMS Conference on

    Mass Spectrometry and Allied Topics, Long BeachCalifornia, 2000; 1552.

    11. Yates, J.R. J. Mass Spectrom. 1998, 33, 1 19.

    12. Gale, B.L.; Bleibaum, J.L.; MacKenzie, R.T.; Martin, R.L.;Straub, K.M. Proceedings 48th ASMS Conference MassSpectrometry and Allied Topics, Long Beach, California,

    2000; 62.Q113. Morris, H.R.; Paxton, T.; Dell, A.; Langhorne, J.; Berg, M.;

    Bordoli, R.S.; Hoyes, J.; Bateman, R.H. J. Rapid Commun.

    Mass Spectrom. 1996, 10, 889896.14. Stevenson, T.I.; Loo, J.A. Proceedings of the 47th ASMS

    Conference on Mass Spectrometry and Allied Topics,

    Dallas, Texas, 1999; 215.Q1

    15. Buko, A.; Tang, Q.; Trevillyan, J.; Chiou, G. Proceedingsof the 47th ASMS Conference on Mass Spectrometry and

    Allied Topics, Dallas, Texas, 1999; 248.

    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.)

    ENCL 1002000128/10/200100012

    Spectroscopic Methods of AnalysisMass Spectrometry 2559

  • 8/14/2019 Applications of Various MS

    16/18

    16. Chalmers, M.J.; Gaskell, S.J. Curr. Opin. Biotechnol. 2000,11, 384390.

    17. Ackermann, B.L.; Regg, B.T.; Colombo, L.; Stella, S.;Coutant, J.E. J. Am. Soc. Mass Spectrom. 1996, 7,12271237.

    18. Kerns, E.H.; Volk, K.J.; Hill, S.E.; Lee, M.S. J. Nat. Prod.1994, 57, 13911403.

    19. Strege, M.A. J. Chromatogr. B 1999, 725, 6778.20. Gilbert, J.R.; Lewer, P. Proceedings of the 46th ASMS

    Conference on Mass Spectrometry and Allied Topics,Orlando, Florida, 1998; 21.

    21. Janota, K.; Carter, G.T. Proceedings of the 46th ASMSConference on Mass Spectrometry and Allied Topics,Orlando, Florida, 1998; 557.

    22. Burdick, D.J.; Stults, J.T. Methods Enzymol. 1997, 289,499519.

    23. Fitch, W.L. Mol. Divers. 1998-99, 4, 3945.24. Submuth, R.D.; Jung, G. J. Chromatogr. B Biomed. Sci.

    Appl. 1999, 725, 4965.25. Swali, V.; Langley, G.J.; Bradley, M. Curr. Opin. Chem.

    Biol. 1999, 3, 337341.26. Dunayevskiy, Y.; Vouros, P.; Carell, T.; Wintner, E.A.;

    Rebek, J., Jr. Anal. Chem. 1995, 67, 29062915.27. Cepa, S.; Searle, P. Proceedings of the 46th ASMS

    Conference on Mass Spectrometry and Allied Topics,Orlando, Florida, 1998; 283.

    28. Wang, T.; Cohen, J.; Kassel, D.B.; Zeng, L. Comb. Chem.High Throughput Screen 1999, 2, 327334.

    29. de Biasi, V.; Haskins, N.; Organ, A.; Bateman, R.; Giles,K.; Jarvis, S. Rapid Commun. Mass Spectrom. 1999, 13,11651168.

    30. Van Hijfte, L.; Marciniak, G.; Froloff, N. J. Chromatogr. BBiomed. Sci. Appl. 1999, 725, 3 15.

    31. Weller, H.N. Mol. Divers. 1998-99, 4, 4752.32. Weller, H.N.; Young, M.G.; Michalczyk, S.J.; Reitnauer,

    G.H.; Cooley, R.S.; Rahn, P.C.; Loyd, D.J.; Fiore, D.;Fischman, S.J. Mol. Divers. 1997, 3, 6170.

    33. Zeng, L.; Wang, X.; Wang, T.; Kassel, D.B. Comb. Chem.High Throughput Screen 1998, 1, 101111.

    34. Davis, R.G.; Anderegg, R.J.; Blanchard, S.G. Tetrahedron

    1999, 55, 16531667.35. Regnier, F.E. Nature 1991, 350, 634635.36. Afeyan, N.B.; Fulton, S.P.; Regnier, F.E. J. Chromatogr.

    1991, 544, 267279.37. Schriemer, D.C.; Hindsgaul, O. Comb. Chem. High

    Throughput Screen 1998, 1, 155170.38. Kaur, S.; McGuire, L.; Tang, D.; Dollinger, G.; Huebner, V.

    J. Prot. Chem. 1997, 16, 505511.39. Geysen, H.M.; Wagner, C.D.; Bodnar, W.M.; Markworth,

    C.J.; Parke, G.J.; Schoenen, F.J.; Wagner, D.S.; Kinder,D.S. Chem. Biol. 1996, 3, 679688.

    40. Hughes, I. J. Med. Chem. 1998, 41, 3804 3811.41. Wagner, D.S.; Markworth, C.J.; Wagner, C.D.; Schoenen,

    F.J.; Rewerts, C.E.; Kay, B.K.; Geysen, H.M. Comb. Chem.High Throughput Screen 1998, 1, 143153.

    42. van Breeman, R.B.; Huang, C.-R.; Nikolic, D.; Woodbury,C.P.; Zhao, Y.-Z.; Venton, D.L. Anal. Chem. 1997, 69,21592164.

    43. Park, S.; Wanna, L.; Johnson, M.E.; Venton, D.L. J. Comb.Chem. 2000, 2, 314317.

    44. Blom, K.F.; Combs, A.P.; Rockwell, A.L.; Oldenburg,K.R.; Zhang, J.H.; Chen, T. Rapid Commun. MassSpectrom. 1998, 12, 11921198.

    45. Lane, S.J.; Pipe, A. Rapid Commun. Mass Spectrom. 1999,13, 798814.

    46. Fang, A.S.; Vouros, P.; Stacey, C.C.; Kruppa, G.H.;Laukien, F.H.; Wintner, E.A.; Carell, T.; Rebek, J., Jr.Comb. Chem. High Throughput Screen 1998, 1, 2333.

    47. Speir, J.P.; Perkins, G.; Berg, C.; Pullen, F. RapidCommun. Mass Spectrom. 2000, 14, 19371942.

    48. Pullen, F.S.; Kerkins, G.L.; Burton, K.I.; Ware, R.S.;Teague, M.S.; Kiplinger, J.P. J. Am. Soc. Mass Spectrom.1995, 6, 394399.

    49. Taylor, L.C.E.; Johnson, R.L.; Raso, R. J. Am. Soc. MassSpectrom. 1995, 6, 387393.

    50. Scientific Instrument Services. AutoProbeTM ApplicationNote; Ringoes: New Jersey, 2000.

    51. Whitney, J.L.; Kerns, E.H.; Rourick, R.A.; Hail, M.E.;Volk, K.J.; Fink, S.W.; Lee, M.S. Pharm. Tech. 1998, May,7682.

    52. Tong, H.; Bell, D.; Keiko, T.; Siegel, M.M. J. Am. Soc.Mass Spectrom. 1999, 10, 11741187.

    53. Richmond, R.; Gorlach, E.; Seifert, J.M. J. Chromatogr. A

    1999, 835, 2939.54. Allen, M.C.; Shah, T.S.; Day, W.W. Pharm. Res. 1998, 15,

    9397.55. Beaudry, F.; Yves Le Blanc, J.C.; Coutu, M.; Brown, N.K.

    Rapid Commun. Mass Spectrom. 1998, 12, 12161222.56. Ackermann B.L.; Ruterbories K.J.; Hanssen B.R.; Lind-

    strom, T.D. Proceedings of the 46th ASMS Conference onMass Spectrometry and Allied Topics, Orlando, Florida,1998; 16.

    57. Lopez, L.L.; Yu, X.; Cui, D.; Davis, M.R. Rapid Commun.Mass Spectrom. 1998, 12, 17561760.

    58. Perchalski, R.J.; Yost, R.A.; Wilder, B.J. Anal. Chem.

    1982, 54, 14661471.59. Lee, M.S.; Yost, R.A.; Perchalski, R.J. Annu. Rep. Med.

    Chem. 1986, 21, 313321.60. Kerns, E.H.; Rourick, R.A.; Volk, K.J.; Lee, M.S.

    J. Chromatogr. B 1997, 698, 133145.61. Kerns, E.H.; Volk, K.J.; Hill, S.E.; Lee, M.S. J. Rapid

    Commun. Mass Spectrom. 1995, 9, 15391545.62. Lee, M.S.; Klohr, S.E.; Kerns, E.H.; Volk, K.J.; Leet, J.E.;

    Schroeder, D.R.; Rosenberg, I.E. J. Mass Spectrom. 1996,31, 12531260.

    63. Williams, A.; Lee, M.S.; Lashin, V. Spectroscopy 2001, 16,3849.

    64. Qin, X.Z.; Ip, D.P.; Chang, K.H.; Dradransky, P.M.;Brooks, M.A.; Sakuma, T. J. Pharm. Biomed. Anal. 1994,12, 221233.

    65. Rourick, R.A.; Volk, K.J.; Kerns, E.K.; Lee, M.S. J. Pharm.Biomed. Anal. 1996, 14, 17431752.

    66. Fouda, H.; Nocerini, M.; Schneider, R.; Gedutis, C. J. Am.Soc. Mass Spectrom. 1991, 2, 164167.

    67. Wang-Iverson, D.; Arnold, M.E.; Jemal, M.; Cohen, A.I.Biol. Mass Spectrom. 1992, 21, 189194.

    ENCL 1002000128/10/200100012

    Spectroscopic Methods of AnalysisMass Spectrometry2560

  • 8/14/2019 Applications of Various MS

    17/18

    68. Ayrton, J.; Dear, G.J.; Leavens, W.J.; Mallett, D.N.; Plumb,R.S. Rapid Commun. Mass Spectrom. 1997, 11,19531958.

    69. Kaye, B.; Herron, W.J.; Macrae, P.V.; Robinson, S.;Stopher, D.A.; Venn, R.F.; Wild, W. Anal. Chem. 1996, 68,16581660.

    70. Olah, T.V.; McLoughlin, D.A.; Gilbert, J.D. RapidCommun. Mass Spectrom. 1997, 11, 1723.

    71. Ayrton, J.; Plumb, R.; Leavens, W.J.; Mallett, D.; Dickins,M.; Dear, G.J. Rapid Commun. Mass Spectrom. 1998, 12,217224.

    72. Dear, G.J.; Fraser, T.J.; Patel, D.K.; Long, J.; Pleasance, S.J. Chromatogr. A 1998, 794, 2736.

    73. Johnson, J.V.; Yost, R.A. Anal. Chem. 1985, 57,758A768A.

    74. Kusmierz, J.J.; Sumrada, R.; Desiderio, D.M. Anal. Chem.1990, 62, 23952400.

    75. Chang, J.P.; Kiehl, D.E.; Kennington, A. Rapid Commun.Mass Spectrom. 1997, 11, 12661270.

    76. Tiller, P.R.; El Fallah, Z.; Wilson, V.; Juysman, J.; Patel, D.Rapid Commun. Mass Spectrom. 1997a, 11, 15701574.

    77. Schultz, G.A.; Corso, T.N.; Prosser, S.J.; Zhang, S. Anal.Chem. 2000, 72, 40584063.

    78. Li, J.; Wang, C.; Kelly, J.F.; Harrison, D.J.; Thibault, P.Electrophoresis 2000, 21, 198210.

    79. Li, J.; Kelly, J.F.; Chernushevich, I.; Harrison, D.J.;Thibault, P. Anal. Chem. 2000, 72, 599609.

    80. Holt, R.M.; Newman, M.J.; Pullen, F.S.; Richards, D.S.;Swanson, A.G. J. Mass Spectrom. 1997, 32, 6470.

    81. Shockcor, J.P.; Unger, S.E.; Savina, P.; Nicholson, J.K.;Lindon, J.C. J. Chromatogr. B Biomed. Sci. Appl. 2000,748, 269279.

    82. Moy, F.J.; Haraki, K.; Mobilio, D.; Walker, G.; Powers, R.;Tabei, K.; Tong, H.; Siegel, M.M. Anal. Chem. in press.

    ENCL 1002000128/10/200100012

    Spectroscopic Methods of AnalysisMass Spectrometry 2561

  • 8/14/2019 Applications of Various MS

    18/18

    Author QueriesJOB NUMBER: 100200012

    JOURNAL: 2820-3

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

    please check.

    ENCL 1002000128/10/200100012

    Spectroscopic Methods of AnalysisMass Spectrometry2562