SlideShare una empresa de Scribd logo
1 de 15
Descargar para leer sin conexión
Guest Editor: Tim Baker 
MASS SPECTROMETRY ANALYSIS OF NEW CHEMICAL ENTITIES 
FOR PHARMACEUTICAL DISCOVERY 
Annette S. Fang,1 Xiusheng Miao,1 Peter W. Tidswell,2 Marc H. Towle,3 
Wolfgang K. Goetzinger,4 and James N. Kyranos5* 
1ArQule, Inc., Woburn, MA 
2Pfizer Global Research and Development, St. Louis, MO 
3Vertex Pharmaceuticals, Cambridge, MA 
4Amgen, Cambridge, MA 
5Wolfe Laboratories, 134 Coolidge Avenue, Watertown, MA 02472 
Received 28 February 2007; received (revised) 20 August 2007; accepted 24 September 2007 
Published online 21 November 2007 in Wiley InterScience (www.interscience.wiley.com) DOI 10.1002/mas.20153 
In this Section, we review the applications of mass spectrometry 
for the analysis and purification of new chemical entities 
(NCEs) for pharmaceutical discovery. Since the speed of 
synthesis of NCEs has dramatically increased over the last 
few years, new high throughput analytical techniques have 
been developed to keep pace with the synthetic developments. 
In this Section, we review both novel, as well as modifications 
of commonly used mass spectrometry techniques that have 
helped increase the speed of the analytical process. Part of the 
review is devoted to the purification of NCEs, which has 
undergone significant development in recent years, and the 
close integral association between characterization and 
purification to drive high throughput operations. At the end 
of the Section, we review potential future directions based on 
promising and exciting new developments. # 2007 Wiley 
Periodicals, Inc., Mass Spec Rev 27:20–34, 2008 
Keywords: mass spectrometry; characterization; purification; 
new chemical entities 
I. BACKGROUND 
In recent years, the development of high throughput synthesis 
techniques has significantly changed the drug discovery process. 
The successful implementation of automation technology that 
allows medicinal chemists to generate 10–1,000 times more 
compounds for screening has augmented the traditional synthesis 
of single compounds. Today’s approach for early discovery hit 
identification is geared towards generating hundreds or even 
thousands of new chemical entities (NCEs) every week. By 
leveraging automation technology, larger numbers of compounds 
can be generated with minimal additional effort. However, in 
order to effectively utilize the subsequent screening information, 
these compounds must be characterized to assess purity and 
quantity. Moreover, screening larger collections of closely 
related analogs together, yields more data per cycle and leads 
to more complete structure activity relationships. By conducting 
the initial cycles of design, synthesis, analysis and screening in a 
high throughput manner, the typical medicinal chemistry process 
is converted to a more efficient and cost effective discovery 
process (Kassel, 2001). 
Traditionally, every compound synthesized would be 
analyzed by techniques such as nuclear magnetic resonance 
(NMR), thin layer chromatography (TLC), and elemental 
analysis. Although these techniques provide the medicinal 
chemist with the necessary characterization information, they 
are not readily amenable to unattended high throughput 
automation and easily automated data interpretation. The ability 
to acquire and interpret data in an automated fashion is an 
important consideration to effectively handle the thousands of 
NCEs being synthesized. While NMR is clearly the technique 
of choice for most traditional medicinal chemists, the need for 
spectral interpretation by an experienced professional limit the 
overall throughput of the technique in a high throughput setting. 
In addition, higher capital costs, limited automation capabilities, 
large sample requirements and the use of deuterated solvents 
impose further limitations on the use ofNMRfor high throughput 
analysis (Haner, Llanos, & Mueller, 2000; Keifer et al., 2000; 
Lindon, Nicholson,&Wilson, 2000; Louden et al., 2000;Wilson, 
Lindon, & Nicholson, 2000). 
II. MASS SPECTROMETRY—CENTRAL 
ANALYTICAL TECHNIQUE 
Simultaneous to the development of high throughput synthesis, 
mass spectrometry (MS) has emerged as the key analytical 
technique to characterize NCEs (Carrell et al., 1995; Enjalbal, 
Martinez, & Aubagnac, 2000; Hauser-Fang & Vouros, 2000; 
Su¨ssmuth & Jung, 1999; Triolo et al., 2001a,b). The inherent 
high sensitivity of MS leads to small sample requirements. 
Moreover, the relative ease of unattended automation and 
automated data interpretation contribute toMSbeing the primary 
characterization technique for high throughput analysis. Below 
———— 
*Correspondence to: James N. Kyranos, Wolfe Laboratories, 134 
Coolidge Avenue, Watertown, MA 02472. 
E-mail: james.kyranos@wolfelabs.com 
Mass Spectrometry Reviews, 2008, 27, 20–34 
# 2007 by Wiley Periodicals, Inc.
we will discuss some of the most common mass spectrometry-based 
techniques along with advantages and disadvantages. 
Depending on the focus of the investigation, a variety of 
analytical data may be desired, such as overall purity, number 
and type of contaminants or the identification of other com-ponents 
in mixtures (Kassel, 2001). In such cases, mass 
spectrometry is often used in combination with other techniques 
such as ultraviolet (UV) or evaporative light scattering detectors 
(ELSD) to collect a range of complementary data for higher 
confidence results (Yan et al., 2000). 
Adapting to the new challenges presented by modern drug 
discovery, all analytical instrumentation and especially mass 
spectrometry have evolved. The most notable changes in MS 
have been associated with the continued refinement of ionization 
techniques and the successful coupling of various separation 
methods with mass spectrometry. Most analyses involving small 
organic compounds to large biomolecules have benefited from 
advances in ionization techniques such as electrospray ionization 
(ESI), atmospheric pressure chemical ionization (APCI) and 
matrix-assisted laser desorption/ionization (MALDI). The most 
pronounced advantage of MS regardless of the ionization 
technique noted above is that resulting spectra are simpler to 
interpret with very few fragments observed compared to electron 
impact (Hauser-Fang & Vouros, 2000). 
In addition to generating simpler spectra, atmospheric 
pressure ionization techniques allow much easier coupling of 
high performance liquid chromatography (HPLC), capillary 
electrophoresis (CE) or supercritical fluid chromatography 
(SFC) with a mass spectrometer. Earlier publications reported 
successful coupling HPLC instruments with MALDI and fast 
atom bombardment (FAB) ionization sources, however, ESI and 
APCI have become the dominant ionization techniques in the 
field of on-line HPLC–MS. 
The role of MS and HPLC–MS for the characterization of 
combinatorial libraries has been described by several authors, 
and rapid HPLC–MS for the analysis of small molecules has 
been reviewed in the past byWehr (2003), Tiller et al. (2003), and 
Hsieh, Brisson, and Wang (2003). Typically, an initial MS 
characterization run provides a quick check for synthetic 
success and an assessment of compound purity. The resulting 
information may then be used to set up a variety of parameters for 
subsequent purification or to further investigate potential 
synthesis failures. Regardless of the motivation or approach, 
given the increased pressure to generate more high quality 
compounds in a shorter time frame, high throughput MS 
methods for both characterization and purification have become 
commonplace. 
III. IONIZATION TECHNIQUES FOR A HIGH 
THROUGHPUT ENVIRONMENT 
Since mass spectrometric detection is very specific in providing 
compound structural information, it is an irreplaceable tool in 
drug discovery (Kleintop, Zhang, & Ray, 2004). In addition, fast 
and accurate analytical methods are essential to keep pace with 
compound libraries produced with high throughput synthesis 
techniques. To facilitate these needs there have been many 
improvements and innovations of ionization sources. 
MASS SPECTROMETRY ANALYSIS & 
A. MUX Source 
One of the most innovative approaches to increase the effective 
utilization of the mass spectrometer, and increase the overall 
analysis speed, is the development of the multi-channel ESI 
(MUX) source (Biasi et al., 1999;Wang et al., 1999; Fang et al., 
2002). The MUX interface consists of multiple electrospray 
probes connected to separate HPLC pumps and one sampling 
cone. Multi-channel electrospray sources have been designed 
with up to eight individual parallel channels ending in an 
electrospray tip at an angle to the sampling cone. A sampling 
rotor connects the spray of each channel to the sampling cone in 
rapid succession. Data acquisition occurs separately for each 
channel with as many data traces as channels used (usually four or 
eight). This approach leverages a single more expensive mass 
spectrometer coupled with multiple separation systems to 
generate an overall cost-effective solution (Sage, Little, & 
Giles, 2000; Burg, 2004). However, because of the fast 
acquisition times that are needed to effectively switch between 
channels, this type of setup is only possible with instruments 
having high scan speeds. Orthogonal acceleration time of flight 
(TOF) mass spectrometer is the instrument of choice for this 
application. As the speed of analysis is increased, the per-sample 
capital expenses are correspondingly reduced. In order to reduce 
the overall per sample analysis time, this type of system has been 
applied for both pre-purification and post-purification analysis 
of high throughput synthesis products (Yan et al., 2004). 
The general approach, with appropriate modification made for 
preparative HPLC, has also been used for high throughput 
purification of diastereomers (Irving et al., 2004). 
B. Combination Sources 
It is estimated that about 80% of small molecular weight 
compounds that are of the same structural class readily ionize 
using a similar electrospray ionization method. For samples that 
do not ionize well using a typical acid modified mobile phase and 
ESI, one can investigate increasing the pH of the mobile phase or 
change the ionization technique. Gallagher et al. (2003) have 
developed a new combined ESI–APCI source (ESCi) for use 
with on-line HPLC applications. This combined source can 
generate clearly differentiated and reproducible ESI and APCI 
spectra. Furthermore, positive and negative spectra can readily be 
obtained by switching polarity. The qualitative performance of 
the combined source has been compared to existing ESI and 
APCI interfaces. The ESI mode of the new source has been found 
to be equivalent to conventional ESI and the APCI mode 
produces less thermal fragmentation. This new source operates 
effectively at flow rates from 50 to 1,000 mL/min and can be used 
with a variety of separation techniques. By eliminating the need 
to repeat chromatographic separations in order to generate 
data using different ionization modes or polarity, the overall 
analysis time is reduced and throughput is increased. 
Similarly, in order to gain an advantage in the ionization of 
low polarity compounds, an APCI/APPI (atmospheric pressure 
chemical ionization/atmospheric pressure photoionization) 
source has been developed. The advantage of this approach, for 
example, is that this source can easily be coupled with normal 
Mass Spectrometry Reviews DOI 10.1002/mas 21
phase HPLC systems for the analysis of very nonpolar 
compounds such as steroids (Robb, Covey, & Bruins, 2000). 
C. MALDI 
MALDI–TOF has become a widely used and powerful tool 
for the analysis of large peptides, biomolecules, and synthetic 
polymers. However, the analysis of small molecules by MALDI 
has remained a challenge. This is primarily due to the technique’s 
relatively poor ionization efficiency and the presence of a variety 
of abundant matrix-related ions in the low mass range. In 
addition, proper sample preparation requires significantly more 
operator experience and thus is not amenable to open access 
walk-up systems that are available to the medicinal chemists. 
Regardless of the potential limitations, MALDI has many 
advantages including low detection limits that are in the 
femtomole range. This level of sensitivity is ideal for the analysis 
of resin bound compounds obtained from individual beads. 
MALDI characterization of single beads has been successfully 
used for characterization of split-and-mix combinatorial libra-ries. 
(Egner, Cardno, & Bradley, 1995; Egner, Langley, & 
Bradley, 1995). By using appropriate photocleavable linkers 
during synthesis, products can be analyzed directly off the solid 
support (Fitzgerald et al., 1996). The MALDI laser irradiation 
provides enough energy to cleave the compound off the bead and 
ionize it at the same time. 
Although low molecular weight interference produced by 
conventional matrices, has limited the utility of MALDI for NCE 
characterization (Cohen & Gusev, 2002; Krutchinsky & Chait, 
2002), recent introductions of new matrices are beginning to 
overcome this issue (Iijima, 1991; Iijima & Ichihashi, 1993; 
Dale, Knochenmuss,&Zenobi, 1996;Ayorinde et al., 1999;Wei, 
Buriak, & Siuzdak, 1999; Han & Sunner, 2000; Kinumi et al., 
2000; Cuiffiet al., 2001; Shen et al., 2001; Zhang et al., 2001; Guo 
et al., 2002; Cai et al., 2003; Fu & Sun, 2003; Lewis et al., 2003; 
Peng et al., 2003; Reyzer et al., 2003; Xu et al., 2003; Pan et al., 
2004; Go et al., 2005; Gobey et al., 2005; Pan et al., 2005; Sunner, 
Dratz, & Chen, 1995). Stolzberg and Patel (2004), for example, 
were able to produce clean negative ion mass spectra of 
molecules in the 100–300 Da range using a light-absorbing 
electrically conductive polymer matrix. Peterson et al. (2004) 
used porous polymer monoliths for MALDI analysis and were 
able to transfer sufficient energy to the analyte to induce 
desorption and ionization. The performance of the monolith 
hydrophobic porous surfaces was demonstrated with several 
small molecules, including drugs, explosives and acid labile 
compounds. 
Separation combined with MALDI–MS for small molecule 
analysis has been reported by several authors using ultra-thin 
layer chromatography (TLC–MALDI) (Busch, 1992; Somsen, 
Morden,&Wilson, 1995;Weins& Hauck, 1996; Chen, Shiea,& 
Sunner, 1998; Wilson, 1999; Gusev, 2000; Mehl & Hercules, 
2000; Hauck et al., 2001; Ka´lasz & Ba´thori, 2001; Hauck & 
Schulz, 2002;Wu&Chen, 2002; Crecelius, Clench,&Richards, 
2003; Salo et al., 2003; Santos et al., 2004). In this approach, 
analytes were separated on TLC plates, which were then coated 
with matrix and subsequently introduced into theMALDIsource. 
Salo et al. (2003) used an atmospheric pressure MALDI (AP-MALDI) 
source in order to reduce possible contamination of the 
mass spectrometer from introducing chromatographic material 
into a vacuum. All analyte spots were successfully ionized 
directly from the TLC plate. 
Gobey et al. (2005) have shown that the use of a high 
repetition rate laser can increase sample throughput while 
filtering out matrix interferences through the use of tandem mass 
spectrometry. The authors especially emphasize that this type of 
MALDI analysis is the ultimate high-throughput technique 
with an analysis time limited only by the speed of the plate 
readers of the MALDI instrument rather than the solvent 
handling capacity of an HPLC–MS system. Limitations include 
ion suppression effects from complex matrices and the need for 
additional SPE cleanup procedures for complex and impure 
samples. 
In a comparison of MALDI and ESI using 14 compounds 
that failed ESI analysis, only four of them were successfully 
analyzed by MALDI. These data suggest that MALDI and ESI 
are generally not orthogonal techniques. In a recent publication 
by Corra et al. (2006), ESI and MALDI were compared with 
regards to several operating considerations, including low-molecular 
weight sensitivities, analysis speed, throughput and 
instrument robustness. They found that although MALDI has a 
faster acquisition speed, detection limits are higher compared to 
HPLC/ESI–MS. However, by developing methods to concen-trate 
samples on the target, detection limits can be significantly 
improved. 
Lee, Chen, and Gebler (2004) have shown that MALDI–MS 
can be used both as a qualitative or quantitative technique. 
MALDI can certainly be used in an automated high-throughput 
mode with individual compounds loaded onto the target. 
However, the typical spot-to-spot sample reproducibility and 
matrix interferences are still limiting factors, especially for small 
molecule analysis. Sleno (2005) have reviewed the fundamental 
and technical aspects of MALDI and its limitations in greater 
detail. If the limitations of the technique can be overcome, 
MALDI analysis might one day provide extremely fast analysis 
of NCEs while consuming only small sample quantities. 
D. Nano and Chip Based Techniques 
In a unique approach Pan et al. (2004, 2005) used oxidized and 
nonoxidized carbon nanotubes as a matrix for small molecule 
MALDI analysis (Iijima, 1991; Iijima & Ichihashi, 1993; Sunner 
et al., 1995; Dale, Knochenmuss,&Zenobi, 1996; Han&Sunner, 
2000; Cai et al., 2003; Fu&Sun, 2003; Peng et al., 2003;Xu et al., 
2003). Compared to conventional matrices, they observed 
dramatically reduced matrix interferences for both types of 
nanotubes. Oxidized carbon nanotubes enhanced signal intensity 
compared to nonoxidized ones. The advantages of oxidized 
carbon nanotubes seem to be associated with greater water 
solubility that leads to a more homogeneous matrix mixture. 
By using silicon nanowires (Go et al., 2005), the sensitivity 
of the technique was increased down to the attomole level. 
Moreover, the 40 nm silicon nanowires were used to conduct a 
chromatographic separation by taking advantage of their high 
surface area and fluid wicking capabilities. The nanowires 
provide a unique platform for both separation and mass 
spectrometric analysis. 
& FANG ET AL. 
22 Mass Spectrometry Reviews DOI 10.1002/mas
Zhang et al. (2001) described quantitative analysis of small 
molecules using chip-based nano ESI–MS/MS. This system is 
reported to have advantages in limiting carryover, sample 
consumption, cycle time and the ability to be fully automated. 
Chip-based nano-electrospray that is integrated into a micro 
purification/collection system has also been reported and will be 
discussed further in the purification section. 
IV. SEPARATIONS COUPLED WITH 
MASS SPECTROMETRY 
Speed is an important consideration in modern drug discovery. 
The more time that is reduced from the discovery and develop-ment 
cycle, the more patent protection the company can enjoy 
once the drug is on the market. Extending the patent protection by 
6 months to a year translates into hundreds of millions of dollars 
in total revenue. It is these types of incentives that drive the 
industry to greater speed and efficiency. For high throughput 
analysis, the need for speed usually means that large numbers of 
compounds have to be analyzed in a matter of days and the results 
have to be interpreted quickly thereafter. 
For practical purposes, high throughput analysis is usually 
further divided into high throughput characterization and 
purification. In characterization, the focus lies in determining 
the contents and/or assessing the purity of a large number of 
samples. Characterization analysis is used as an initial check for 
synthesis completion, quality control purposes or to collect 
important data about samples that need to be purified later. For 
purification, the data obtained in characterization is typically 
used to optimize the subsequent preparative analyses. Given the 
robustness of the single quadrupole systems and the lower cost 
relative to the high end mass spectrometers, low-resolution 
HPLC–ESI–MS instruments have become the industry standard 
for high throughputNCEcharacterization. There have been some 
examples of high throughput high resolution work but usually 
this process is significantly more complex and in most cases 
unnecessary (Walk et al., 1999). 
A. FIA–MS 
Flow injection analysis mass spectrometry (FIA-MS) is the 
simplest form of rapid sample introduction into the mass 
spectrometer. This approach was one of the earliest examples 
of characterization and has been widely used in the analysis of 
high throughput synthesis products. If separation is not needed, 
FIA can be used to confirm synthesis by the presence of the mass-to- 
charge ratio (m/z) of the expected compound ions. Since flow 
injection analysis is not limited by separation run times, it has real 
speed advantages. FIA–MS provides the highest throughput and 
greatest ease of automation and use for large numbers of samples 
(Yu & Balogh, 2000). A major drawback to FIA–MS is ion 
suppression due to co-eluting sample components. The ion 
suppression can be quite significant. In many cases there is 
significant chemical interference and the expected molecular ion 
of the desired compounds is not observed, even though the 
synthesis may have been successful. Moreover, without separa-tion 
of the synthesis mixture components, there is no reliable 
purity determination. 
MASS SPECTROMETRY ANALYSIS & 
One of the early issues arising in FIA–MS analysis was 
sample to sample carry-over. This issue can be a serious concern 
for any analysis, however, it is of particular issue when running 
large number of samples very quickly, where wash cycles may be 
abbreviated to gain cycle time speed. Earlier instrumentation was 
limited in its control of injector parameters and thus more prone 
to carryover when trying to maximize analysis speed. Richmond 
(2000) and Richmond and Go¨rlach (1999a) described minimiz-ing 
sample carryover in reaction monitoring using FIA–MS. 
Morand et al. (2001) reported an improved method with a 
throughput of 4 sec/sample by using a parallel sampling system 
with an eight-probe injector. Others have worked on improving 
speed or the analytical design (Felten et al., 2001) and also on 
theoretically analyzing inter-sample carryover (Richmond & 
Go¨rlach, 1999b; Richmond, 2000). The increasing numbers of 
samples for analysis required a further reduction in the sample 
measurement duty cycle while maintaining the carry-over below 
1%. Lazar et al. (1999) introduced high-speed autosamplers with 
duty cycles that could be reduced from 168 to 44 sec. Further 
optimization of syringe and loop wash steps could reduce the 
median inter-sample carry-over to 0.01%. Wang et al. (1998) 
shortened the sample injection duty cycle by using a multiprobe 
autosampler for flow-injection analysis of eight samples within 
1 min. Using this type of autosampler, it has been shown that 
more than eight flow-injection peaks can be generated in 1 min, 
which translates into analyzing one whole 96-microtiter well 
plate in less than 12 min (Wang et al., 1998). 
Shah et al. (2000) described a quantitation method with 
FIA–MS and chemiluminescence nitrogen detection (CLND) 
for simultaneous proof of structure and estimation of purity, 
respectively. Characterization of one compound was completed 
every 60 sec, allowing for more than 1,000 compounds analyzed 
in a single day. However, when large amounts of impurities were 
present or the compound of interest was present at a very low 
concentration, purity estimation was less reliable. In these types 
of cases, HPLC–CLND and HPLC–MS may be required. 
Although, as described above, there have been many 
publications focusing on high-throughput injections and the 
reduction of carryover in FIA–MS, most commercial systems 
have been optimized for approximately one injection per minute. 
This injection speed is adequate for typical HPLC analysis but 
not high throughput FIA–MS operations. The approaches 
described above that were used for multiple injections per 
minute, require extensive modifications of injector hardware and 
software. 
B. HPLC–MS 
Flow–injection analysis provides the shortest run time for a high 
throughput system but does not yield confident purity determi-nation. 
In order to assure the highest possible data quality with 
more complex samples, chromatographic separation is neces-sary. 
There is significant opportunity to develop the appropriate 
analytical methods by coupling HPLC to mass spectrometry, 
however, usually at the expense of longer analysis times. In an 
effort to decrease HPLC–MS run times, rapid mobile phase 
gradients and/or modifiers, as well as packing materials and 
column size are among the more flexible components that have 
Mass Spectrometry Reviews DOI 10.1002/mas 23
been adjusted to fit individual analysis. All these parameters have 
been used to develop effective high quality and rapid character-ization 
and purification methods. Of course, chromatographic 
analysis times can also be dramatically shortened by using 
different techniques such as capillary electrophoresis (CE), 
capillary electrochromatography (CEC) or supercritical fluid 
chromatography (SFC). Some of these techniques will be 
reviewed further in subsequent sections. 
In general, modifiers are compounds that alter the 
physicochemical properties of the mobile phase. Modifiers have 
been used quite extensively to optimize specific chromatographic 
separations. Although a variety of modifiers have been developed 
and reported, only those that are volatile are compatible with the 
needs of the mass spectrometer. This requirement has limited 
the use of modifiers primarily to trifluoroacetic acid (TFA) and 
formic acid (FA) for acidic methods and ammonium buffers for 
basic separations. 
In addition to optimizing chromatographic resolution, 
mobile phase additives can be used to improve MS sensitivity 
in both ESI and APCI detection modes. Mallet, Lu, and Mazzeo 
(2004) evaluated ion suppression/enhancement in negative-ion 
mode by using several additives and ion-pairing agents at 
different concentrations for test compounds. The intensity of a 
novel indolocarbazol compound increased by a factor of four 
in negative-ion mode when the pH of the mobile phase was 
increased from 8 to 10 using 2mMammonium hydroxide (Wang 
et al., 2002). Similarly, the sensitivity of detection was increased 
up to tenfold for isorhamnetin with a decrease in pH of the mobile 
phase ranging from 4.5 to 2.3 in positive-ion mode (Rauha, 
Vuorela, & Kostiainen, 2001). The addition of alkali metal salts 
to enhance the intensity of molecular ion adducts by forming 
stable Liþ, Naþ, or Kþ complexes of organic compounds has 
been observed in FAB, MALDI, and ESI mass spectrometry of 
various compounds (Williams et al., 2003). The extreme form of 
mobile phase modification can be considered micelle formation 
used with electrophoresis, which affects the fundamental 
separation processes. There have been reports of various 
approaches, such as micellar electrokinetic chromatography 
in combination with mass spectrometry (Goetzinger & Cai, 
2005). 
Chromatographic resolution and mass spectrometric sensi-tivity 
are important considerations for all analytical methods. 
However, the real speed limitation for any high throughput 
analysis using HPLC–MS is the chromatographic separation 
time. Analysis speed has been enhanced by reducing the column 
size from typical column dimensions of 4.6150 mm to 
4.630 mm. These shorter columns can be used with higher 
flow rates without exceeding the operating pressures limits. 
HPLC instrumentation has also been adapted to enable high-speed 
gradients. Presently, gradients of 5–95% acetonitrile/ 
aqueous buffers within 1–5 min with fast column regeneration 
are quite common. These developments have increased the 
throughput from 24 to 48 samples per day using a 30–60 min 
analysis to more than 250 samples per day using a 5 min analysis. 
One to five minute run times have become common for many 
laboratories. For our operations, run times of 2.5 min are the 
standard with no apparent negative effects on peak shape or 
resolution (Kyranos et al., 2001a). Such short run times make 
high throughput HPLC–MS analysis a practical and effective 
analytical solution to support high throughput synthesis. 
Goetzinger and Kyranos (1998) investigated packing 
materials, gradient methods and sample solvents to allow 
FIGURE 1. QC of combinatorial library sample with rapid HPLC. a: 1504.6 mm, 5 mm Zorbax SB-C8, 
1.5 mL/min, 15–95% acetonitrile in 16 min; (b) 504.6 mm, 3.5 mm Zorbax SB-C8, 2 mL/min, 15–95% 
acetonitrile in 4 min; (c) 304.6 mm, 3.5 mm Zorbax SB-C8, 4 mL/min, 15–95% acetonitrile in 0.6 min. 
 FANG ET AL. 
24 Mass Spectrometry Reviews DOI 10.1002/mas
ultra-fast gradient HPLC–MS methods down to 1 min per 
sample. Similarly, Hsieh et al. (2001) used fast gradients within 
1 min for quantitative screening of drug discovery compounds 
in monkey plasma. Using generic methods to avoid method 
development is another trend to enhance the throughput for the 
analysis of combinatorial libraries. Figure 1 is an example of 
rapid chromatography using fast gradients and high flow without 
compromising chromatographic resolution. Although the run 
time is significantly decreased from 20 to 1 min, the overall peak 
capacity for all the separations is still acceptable to determine 
sample purity. The significant developments in separation speed 
have shifted the efforts for further optimization and time savings 
toward the autosampler and injector components (Zweigenbaum 
et al., 1999). 
As previously discussed, the MUX interface has been used 
successfully with FIA–MS to increase the overall throughput. 
Davis and Griffith (2004) described a high-throughput parallel 
HPLC–MS–ELSD method for the analysis of combinatorial 
libraries with an eight-channel TOF system. This configuration 
could analyze approximately 300,000 compounds per year. Fang 
et al. (2003) investigated parallel high-throughput accurate mass 
measurement using a nine-channel multiplexed HPLC–UV– 
TOF–MS system. Accurate mass measurements were simulta-neously 
obtained from compounds eluting from eight parallel 
columns. Mass accuracy better than 10 ppm was achieved for 
80% of the samples investigated. 
In other examples of higher throughput, Cremin and Zeng 
(2002) applied an eight-way fully automated parallel LC–MS– 
ELSD system to enhance the throughput for the analysis of a 
natural product library. This method has been successfully 
applied to analyze a library of 36,000 partially purified fractions 
derived from plants. 
The quest for additional separation speed and higher 
efficiency, which in turn will increase the overall analysis 
throughput, continues with the development of new stationary 
phase materials. Much of the early development in this area has 
been focused on decreasing the particle size of the packing 
material. However, the higher operating pressure associated with 
particle size reduction has limited this advancement. Monolithic 
columns are a single solid piece of stationary material with pores 
through the main skeleton. These structures can increase column 
performance by increasing the column permeability and 
efficiency (Tanaka et al., 2002). There have been a number of 
applications in related research areas using monolithic columns 
that exemplify the beneficial performance of these materials 
(Asperger et al., 2002; Borges et al., 2004). 
C. CE–MS 
Capillary electrophoresis (CE) is a powerful separation techni-que 
that has been used to separate ionic compounds (Foulon et al., 
2004; Liu, Jo¨nsson,  Jiang, 2005) or chiral mixtures (Millot, 
2003; Erny  Cifuentes, 2006; Souverain et al., 2006). The 
different mode of separation of CE results in a unique selectivity 
compared to reverse phase HPLC. Moreover, high efficiency 
associated with CE promise enhanced separation resolution, 
which will lead to even faster analyses. This technique can be 
used as an orthogonal complimentary approach to HPLCor as the 
primary analysis. However, the difficulties in interfacing CE to 
the mass spectrometer and the limited amount of material that can 
be loaded on the column have hindered wider use of CE–MS. 
Dunayevskiy et al. (1996) was one of the first to apply CE– 
MS to combinatorial chemistry. They successfully identified the 
171 individual components of a combinatorial library mixture 
using CE–MS. Visky et al. (2005) discussed the application of 
CE–MS and its role in impurity profiling of pharmaceutical 
products. More recently, Smyth reviewed the broader applica-tions 
of CE–MS in drug analysis (Smyth, 2005). Generally, CE– 
MScan be a valuable technique for rapid compound separation. It 
is also useful for analyzing extremely small sample volumes with 
high separation efficiency and has been applied to the analysis of 
complex biological matrixes (Moini, 2002; Wan  Thompson, 
2005). 
Goetzinger and Cai (2005) described a newway of preparing 
a micellar buffer system for the separation of neutral compounds 
based on micellar electrokinetic chromatography (MEKC). The 
substitution of the traditionally used alkyl sulfates, such as SDS, 
with carboxylates together with the substitution of inorganic 
cations with organic bases provided a different separation system 
with interesting properties. It was shown that these novel buffer 
systems could be used as micellar separation systems with lower 
conductivity compared to SDS buffers and thus provided more 
flexibility for separation conditions. Although this article was 
conducted using UV detection, the buffers used are volatile and 
amenable to interfacing with MS. The inevitable coupling of this 
novel MEKC system with MS could be used to increase 
separation speed or alternatively to improve the detection limit 
by using larger diameter capillaries. 
D. SFC–MS 
Supercritical fluid chromatography (SFC) has been used for 
chiral separations (Mukherjee, 2007). Recently there has been a 
growing trend in the application of SFC–MS to the analysis of 
NCEs (Berger et al., 2000). SFC offers the potential benefit of 
high-speed separations that can be complementary to reverse 
phase HPLC–MS (Berger, 1997; Coe, Rathe,  Lee, 2006). Due 
to the lower viscosity of supercritical fluids, SFC can be run at 
higher flow rates compared to HPLC. Bolan˜os, Ventura, and 
Greig (2003) demonstrated the advantage of SFC–TOF MS 
for ultrafast qualitative applications. High-speed MS detection 
allows the superior resolving power of SFC to be exploited 
in high-throughput analytical settings. Bolanos et al. (2004) 
extended their work and summarized the applications of SFC– 
MS in drug discovery at Pfizer, La Jolla, CA. They rely heavily 
on SFC–MS for analysis, as well as, purification of diverse 
compound sets. 
SFC for purification applications is especially appealing due 
to rapid evaporation of the carbon dioxide mobile phase (Toribio 
et al., 2006; Zhang et al., 2006). During separation, the carbon 
dioxide mobile phase is under high pressure and behaves like a 
fluid. However, once the pressure is reduced after the detector, the 
fluid rapidly expands into a gas that quickly evaporates leaving 
behind the analyte and any organic modifier such as methanol. 
The rapid evaporation of the solvent after purification signifi-cantly 
increases the overall throughput of preparative SFC 
compared to HPLC. 
MASS SPECTROMETRY ANALYSIS  
Mass Spectrometry Reviews DOI 10.1002/mas 25
Although there are many theoretical and practical advan-tages 
to SFC, most laboratories have not embraced the technique 
for characterization nor purification. Much of the resistance is 
probably due to the perception that SFC is more complicated than 
HPLC. In addition, there are significantly fewer vendor options 
for instrumentation and application support. Finally, SFC more 
closely resembles the separation of normal phase chromatog-raphy 
and many scientists may assume that SFC will not be 
applicable to many of the polar and ionic compounds typically 
associated with pharmaceutical research and development. 
Pinkston et al. (2006) have recently shown that approximately 
75% of a 2266 compound subset, representing their corporate 
collection, could be adequately assessed by SFC–MS. This 
compares to approximate 80% success of the same subset by 
HPLC–MS. These results further reinforce the value of SFC as an 
important technique for high throughput analysis in support of 
modern drug discovery and development. 
E. Multiple Hyphenated Detection Techniques 
One of the benefits of HPLC–MS is the ability to couple a variety 
of detectors to generate a range of qualitative, as well as 
quantitative information. It is quite common for one or more 
auxiliary detection methods to be combined with HPLC–MS to 
provide a better overall assessment of sample purity. A typical 
approach has been to couple UV and ELSD. Although the 
different detection methods often vary regarding detection 
capabilities, selectivity, sensitivity range, and linear response, 
collectively they provide complementary information. 
Due to variations of quantitative data from different 
detection techniques, Letot et al. (2005) investigated HPLC– 
CLND and HPLC–NMR for quantification of combinatorial 
library compounds. In general, he found that purity values 
obtained by NMR were more accurate than those obtained by 
CLND. However, the differences in purity are not always of the 
same order for the two methods. For low concentration 
quantification, NMR is less accurate than typically expected 
with higher sample concentrations. 
Yurek, Branch, and Kuo (2002) developed a new assay 
system for the simultaneous determination of identity, purity and 
concentration of sample components from combinatorial libra-ries 
produced by parallel synthesis. The system employed HPLC 
with photodiode array (PDA), ELSD, CLND, and TOF–MS 
detectors. The use of TOF–MS with CLND provides a 
synergistic combination, enabling target and side-product 
structures to be identified and quantified. This approach allows 
characterization, purity analysis and quantification in a single 
experiment from a synthesis solution containing microgram 
levels of materials. Recently, Peake et al. (2005) described a high-throughput 
screening method using ELSD, CLND and accurate 
mass HPLC–MS–MS to obtain both quantitative data and 
structural information. 
Although the complementary nature of the different 
detection methods is often a benefit, it can also be a challenge. 
In particular, sensitivity and detector response times may vary 
significantly among the various detectors. Therefore, it is very 
important to assure that all operating parameters are appropri-ately 
optimized. This is especially important for rapid separation 
methods, which produce chromatographic peaks with 1–2 sec 
widths. These systems are very susceptible to operating vari-ations 
and must be diligently optimized for peak performance. 
Another consideration when using multiple detectors to 
assess purity is developing protocols to deal with major purity 
inconsistencies between multiple detectors. Of course the 
definition of purity depends on the application and can vary for 
different experiments. In general, however, purity values of 
around 80–85% for both UV and ELSD have been found 
acceptable for many early discovery applications. It is important 
to note that for hyphenated detection, all purity values have to be 
above some predefined threshold. One failed value can indicate 
the presence of major impurities that have not been identified by 
the other methods. In our experience, we found that ELSD tends 
to overestimate the larger components in a mixture at the expense 
of the smaller ones. UVon the other hand, even monitoring low 
wavelengths, may exhibit more variability depending on 
structural differences of the mixture components. This is the 
advantage of the complementary detector approach, which 
generates higher confidence data (Letot et al., 2005). 
F. Open-Access HPLC–MS 
Although not a true high-throughput technique in the typical 
sense, the open-access or walk-up HPLC–MS system does 
provide quick single sample HPLC–MS analysis. This system 
has presently become the standard setup in many laboratories. 
The open-access system can be used at any time by the medicinal 
chemist in need of rapid confirmation of synthetic products or to 
monitor the extent of reactions. The quick single sample analysis 
can provide information about the completeness of a reaction, 
byproducts or unexpected reaction failure. These data add 
another layer of information to that obtained from NMR analysis 
or other traditional low throughput approaches. Once imple-mented, 
open-access systems accept individual samples in an 
ongoing queue and can be run without much support from the 
analytical department. 
The open-access approach was pioneered by Hayward, 
Snodgrass, and Thomson (1993) and optimized by Pullen and 
coworkers at Pfizer Central Research (Pullen et al., 1995a,b). 
Initially, an automated column-bypass thermospray MS system 
was used but later this was reconfigured into an ESI–MS system. 
The instrument is usually set up to email the results as completely 
processed data to the submitting chemists (Mallis et al., 2002). 
Because of its success, open-access software modules are 
currently offered by most instrument manufacturers to be used 
with single-quadrupole and TOF instruments. The use of an 
open-access facility can enhance sample throughput and allow 
the analytical chemists to spend more time on nonroutine work. 
Spreen and Schaffter (1996) have extensively reviewed applica-tions 
of open-access MS. 
It has been shown that open-access MS works extremely 
well for fast characterization of synthesis samples. Most small 
molecules that are of interest for pharmaceutical research ionize 
well using a standard set of instrument conditions and time 
consuming adjustments are typically not needed. Although 
the typical setup for a walk-up mass spectrometer includes a 
low resolution instrument, high resolution instrumentation has 
also been used effectively (Dykes et al., 2003). There have also 
been attempts to extend the walk-up approach to purification 
 FANG ET AL. 
26 Mass Spectrometry Reviews DOI 10.1002/mas
systems (Blom et al., 2004). However to date, purification 
systems have not been adopted as readily as the characteriza-tion 
systems. 
V. MIXTURE ANALYSIS 
Most high throughput synthesis approaches being used generate 
a single discrete compound per reaction vessel. Solid phase 
mix-and-split combinatorial synthesis was one of the earliest 
synthesis techniques that generate mixtures of closely related 
analogs in each reaction vessel. The emphasis for this approach is 
to synthesize as many compounds as possible in a small number 
of reaction pools and then screen these mixtures to identify hits. 
Once a hit is identified in a mixture pool, that mixture pool is 
deconvoluted to identify the active component(s). Alternatively, 
the active mixture can be resynthesized in a series of smaller 
mixture pools and these pools screened for activity. By iteratively 
synthesizing and screening ever smaller mixture pools, the active 
compound(s) can ultimately be identified. This approach does not 
require complete characterization of the individual members 
of a mixture pool. The emphasis for analysis here is a quick 
assessment of how well the reaction has proceeded in general. 
The intent is often to eliminate complete synthesis failures rather 
than identifying and characterizing an individual compounds. 
Although HPLC–MS has been used in characterizing these types 
of complex mixtures, it is quite difficult and labor intensive. For 
such a general evaluation of a synthetic procedure, direct analysis 
of the mixture using mass spectrometry has been used quite 
effectively. By knowing the compounds expected in a synthesis 
pool and assuming that each compound is equally represented in 
the mixture, theoretical mass distributions can be generated. 
These theoretical mass spectra can then be compared to the actual 
spectra and conclusions about the success of the synthesis can be 
drawn. These types of analyses have been successfully conducted 
using both lowand high resolution instrumentation (Carrell et al., 
1995; Dunayevskiy et al., 1995; Nawrocki et al., 1996). 
Fourier transform ion cyclotron resonance (FTICR) instru-mentation, 
also known simply as Fourier transform mass 
spectrometry (FTMS), are high field magnetic instruments that 
are usually installed in a laboratory to handle nonroutine 
applications. The high resolution capabilities of FTMS and 
its more complex operation make it somewhat of a specialty 
analytical tool. Although FTMS can be used as an HPLC detec-tor, 
as described earlier in this review, the high costs and the more 
complex operation associated with it have limited this use. The 
primary advantages of FTMS are its high sensitivity (sub fmol), 
high mass accuracy (sub ppm levels), high resolution for complex 
mixtures (600,000 FWHM) and sequencing capabilities (exact 
MS(n) capability). 
The high resolution capabilities of FTMS are particularly 
appealing for the characterization of mixture analysis since 
accurate mass measurements can be used to correctly identify 
potential isobaric compounds without the use of chromato-graphic 
separation. Furthermore, the ability to conduct sequential 
MS–MS and obtain accurate mass assignment for the fragments 
helps to accurately identify the molecular structure of specific 
compounds in a complex synthesis mixture. Although HPLC– 
MS–MS may be able to adequately separate and characterize 
MASS SPECTROMETRY ANALYSIS  
individual structures in a complex matrix, direct analysis of 
the synthesis mixture using high resolution accurate mass FTMS 
in some cases may be easier and faster. On the other hand, 
conducting sequential MS–MS for each component of a large 
complex mixture is quite tedious, time consuming and not 
particularly amenable to high throughput environment. Several 
publications have focused on FTMS as a technique to character-ize 
(Winger  Campana, 1996; Fang et al., 1998; Schmid et al., 
2001) or screen (Poulsen, 2006) combinatorial library mixtures 
of discrete small molecules (Bristow  Webb, 2003; Herniman 
et al., 2004, 2005; Zhang et al., 2005). 
VI. PURIFICATION OF NCES 
The number of new chemical entities undergoing purification 
before any biological assays are undertaken has increased over 
the last few years.(Ripka, Barker,  Krakover, 2001). This is 
mostly due to the recognition that valuable resources are often 
wasted when impure compounds are submitted for screening 
only to end up generating false positives. This issue has led many 
companies to institute specific purity thresholds for compounds 
supporting medicinal chemistry projects. An industry standard of 
at least 80–85% purity by UVat 210–214 nm has become fairly 
typical (Letot et al., 2005). 
A. Preparative HPLC–MS 
Preparative HPLC using small efficient columns that allow for 
rapid separations have become the mainstay of the industry. 
Many laboratories use traditional UV detection to collect all 
fractions above a certain threshold. The collected fractions are 
then rapidly analyzed by FIA–MS to identify the component(s) 
of interest. Due to collecting multiple fractions from one sample, 
the overall throughput of this approach is severely constrained. 
The collection decks can accommodate only a limited number of 
fraction collection tubes and once those tubes are used, the 
operation stops. In an effort to alleviate this limitation, most of the 
current high throughput purification setups use some variation of 
HPLC–MS instrumentation as the method of fraction collection. 
The specificity of the mass spectrometer is used to trigger 
collection only when the expected mass of the product is seen in 
the sample. This approach significantly reduces the total number 
of fractions collected per sample and allows more samples to be 
purified per run.We have pushed the throughput to the maximum 
by allowing only one fraction to be collected for each sample. 
This type of setup is becoming more common and can purify 
approximately 3,000 compounds each month in a completely 
automated fashion using a single instrument. Setups for this type 
of analysis include simple single channel HPLC–MS systems as 
well as MUX systems, as described above. 
For purification of crude synthesis products, the most critical 
step in the process is the mass directed fraction collection. After 
separation by the HPLC column and detection by UV, the eluant 
is connected to a tee that splits the chromatographic flow. The 
majority of the HPLC eluant is directed to a switching valve 
connected to the fraction collector. This valve, which is 
controlled by the MS output signal, toggles between the 
collection tubes and the waste line. A smaller portion of the 
eluant is coupled to the MS system via a split flow interface. 
Mass Spectrometry Reviews DOI 10.1002/mas 27
The mass spectrometer monitors the expected m/z trace of 
the component of interest. When the expected m/z rises above a 
predetermined threshold value, the collector valve is switched 
‘‘on’’ to collect the fraction of interest. When the monitored m/z 
trace drops below a certain threshold, flow through the switching 
valve is directed to waste. In our approach, the waste line is 
connected to a second UV detector and the eluant is monitored 
prior to discharging it in the waste system. The overall efficiency 
of the collection process can be monitored for each sample in 
real time by comparing the trace of the UVimmediately after the 
HPLC column with the chromatographic trace from the waste 
UV (Goetzinger et al., 2004). 
The amount of sample being split and infused into the mass 
spectrometer must be small. Since mass spectrometry consumes 
the sample during detection, only the absolute minimum of the 
sample should be sacrificed. Moreover, the sensitivity of MS is 
quite high, therefore, the amount necessary for good signal-to-noise 
detection is relatively small. In fact, given the high sample 
concentration eluting off the preparative HPLC column and 
the minimal sample necessary for good MS detection, a 1:10,000 
split ratio is fairly typical. To effectively and reproducibly 
accommodate this split, small lengths of capillary tubing ranging 
from 5 to 25 mm in diameter are typically used. The high split 
ratio necessary to limit the amount of material entering the mass 
spectrometer requires additional make up flow provided by a 
separate pump. 
It has been suggested that a major drawback of mass directed 
fraction collection is the lack of ruggedness. This apparent 
limitation is more of a perception issue because of the more 
complex nature of MS and the experience necessary to set up and 
use split flow systems. Our experience has been that the system 
will be fairly stable and perform well as long as performance is 
continually monitored and regular preventive maintenance is 
conducted. Our protocols include running daily standards, 
checking system performance and recovery every month and 
regularly replacing in-line filters to prevent clogging of the flow 
split capillary. It has been shown by us and others that using the 
setup described above, more than 200 compounds can be purified 
in unattended overnight runs (Isbell et al., 2002, Kyranos et al., 
2001b). In addition to implementing diligent preventive main-tenance 
procedures, custom monitoring systems can be 
employed that trigger an alarm and/or action when a variety of 
potential failures are detected. Potential system failures include 
high back pressure, injector clogs, collector failures, no anti-cipated 
masses to trigger collection are found or data acquisition 
errors. By properly programming these system monitoring 
devises, responses to the aforementioned failures can be 
customized, ranging from system shutdown to selectively 
rebooting an unresponsive computer to allow for continued 
operation. This type of custom sensor can be highly effective for 
unattended overnight runs. It can prevent samples from being 
injected into a failing system or will reboot a nonresponsive 
acquisition system and get it back into operation without wasting 
much time during the purification run (Paulson, 2005). 
B. Connecting Characterization and Purification 
The focus of the initial characterization analysis for synthesis 
samples is on developing high performance methods that can be 
used as general universal methods for all samples. Speed with 
maximum resolution of all the components in the mixture across 
the entire run time are important considerations. Once the pre-purification 
analysis is completed, the emphasis shifts to 
optimizing the separation conditions exclusively for the peak of 
interest. It is irrelevant if several other peaks are overlapping or if 
the majority of the peaks are closely eluting either early or later in 
the chromatogram. As long as the product peak is well separated 
from other components and can be collected with ease, 
purification will be successful. Ideally, in a high throughput 
environment where compound diversity is the rule, each 
synthesis mixture needs to be treated differently. This challenge 
contributed to developing automation routines that use the 
information obtained from initial characterization data to adjust 
the separation gradients used for purification. 
For example, if the compound of interest was found co-eluting 
with impurities late in the gradient, then the purification 
gradient is automatically adjusted to start with a high percentage 
of organic solvent followed by a shallow gradient until the 
desired product is collected. Using this approach, co-eluting 
impurities are adequately separated from the desired peak more 
effectively and a cleaner fraction is collected. This process is 
illustrated with the example in Figure 2. Figure 2a represents the 
pre-purification chromatogram, identifying the peak of interest 
(shaded). Based on this separation, the gradient was modified in 
order to further separate the product from the side products 
during the preparative run (Figure 2b). 
In reality, not all characterization data will identify perfect 
gradient assignments for all samples. There can be challenges 
regarding continued co-elution or target peaks that are broadened 
by a shallow gradient. But compared to running a routine 
universal gradient of 5–95% acetonitrile for all compounds, this 
method of adjusting the gradients based on characterization data 
routinely outperforms the rigid traditional approach. Thus it 
provides an effective way to optimize purification conditions for 
the large majority of compounds without significant operator 
effort. 
C. Post-Purification Challenges 
Other recent advances have been in the combination of analytical 
and preparative scale instrumentation that yields an analytical-to-prep 
combination. This instrument has the ability to perform an 
initial characterization analysis and under preset guidelines, 
utilizes the processed data from the first run to automate a 
purification analysis. In some cases this has been extended to 
subsequent characterization of the collected fraction(s). 
Since many high-throughput environments now need to 
handle large amounts of liquids from collected fractions, some 
efforts have been made to simplify these tasks. One such effort is 
the design of a fraction pooling station as described by Koppitz, 
Brailsford, andWenz (2005). In their approach, product fractions 
are automatically identified from their corresponding mass 
spectra, even accounting for sodium adducts or dimers. The 
key elements of this approach are automated identification of the 
target compounds. This information is then used to optimize 
preparative gradients for purification of the target compounds 
followed by automated purity assessment of fractions with 
 FANG ET AL. 
28 Mass Spectrometry Reviews DOI 10.1002/mas
MASS SPECTROMETRY ANALYSIS  
FIGURE 2. a: Pre-purification QC analysis using a gradient of 5–95% acetonitrile. The product peak 
(4) partially co-elutes with an impurity at 1.43 min. b: Purification run with conditions adjusted to a gradient 
of 30–95% acetonitrile based on pre-purification results. The product peak is completely resolved from the 
co-eluting impurity that was detected in pre-purification QC run. 
subsequent pooling of validated product fractions. Purity 
assessment of product fractions allows cherry-picking of pure 
fraction tubes on the basis of an algorithm that calculates MS 
purities and considers fragmentation. Using this type of system, 
one person can easily process 100–200 compounds per day. 
D. Microscale Separations and Collection 
Besides the potential for sample path blockages, the sample loss 
associated with the use of small diameter capillaries for flow 
splitting is not a major factor for mass directed fraction collection 
with semi-preparative HPLC flows. Flow splitting can indeed 
become a problem once the scale and the flow rate become 
significantly smaller than traditional semi-preparative or ana-lytical 
applications. This has been reported byWachs and Henion 
(2001) and is part of the driver for the development of an 
integrated nano scale fraction collection system that is being 
marketed commercially as an ultra-low-volume fraction collec-tor 
and analyzer. This type of system monitors mass traces using 
an automated chip-based nanospray and collects fractions that 
can subsequently be reanalyzed automatically. 
VII. CONCLUSION AND FUTURE DIRECTIONS 
In the early 1990s, there was an explosive surge of new 
technology development primarily focused on characterization 
of NCE’s in support of early pharmaceutical discovery. During 
this period, mass spectrometry emerged as the central analytical 
technique in the development of modern high throughput 
Mass Spectrometry Reviews DOI 10.1002/mas 29
analysis. The MS technologies covered in this review have made 
a leap from primarily research purposes to becoming an integral 
part of modern medicinal chemistry. However, over the last 
5 years there has been greater emphasis on fine tuning the existing 
characterization techniques and making them more robust and 
reliable rather than pioneering new technologies. The emphasis 
on future developments in MS characterization is anticipated to 
be continuing to streamline and further increase throughput. 
There is still opportunity for significant improvements. Data 
handling and analysis can be automated further. Instrument and 
method troubleshooting can be automated. Development of 
additional methods that can accommodate more complex 
samples and new MALDI matrices that are more amenable to 
low molecular weight compounds are also needed. 
In contrast toMScharacterization, there has been significant 
development of high throughput purification using mass directed 
fractionation. Areas of future development are anticipated in 
automatic gradient or column assignments specific to the need 
of each sample, as well as improved automated fraction 
handling. 
Successful chip-based MS analytical techniques for both, 
characterization and purification have been reported. This area of 
research is promising to be the most significant development in 
NCE characterization and purification. The small volumes that 
are typically used with chip based techniques have the potential 
for rapid and ultra-high throughput while maintaining opera-tional 
and consumable costs low. However, the rate of future 
development of these techniques and how fast they may be 
adopted in mainstream applications is yet to be determined. 
REFERENCES 
Asperger A, Efer J, Koal T, Engewald W. 2002. Trace determination of 
priority pesticides in water by means of high-speed on-line solid-phase 
extraction-liquid chromatography-tandem mass spectrometry using 
turbulent-flow chromatography columns for enrichment and a short 
monolithic column for fast liquid chromatographic separation. 
J Chromatogr A 960:109. 
Ayorinde FO, Hambright P, Porter TN, Keith QL Jr. 1999. Use of meso 
tetrakis(pentafluorophenyl)porphyrin as a matrix for low molecular 
weight alkylphenol ethoxylates in laser desorption/ionization time-of-flight 
mass spectrometry. Rapid Commun Mass Spectrom 13:2474. 
Berger TA. 1997. Separation of polar solutes by packed column supercritical 
fluid chromatography. J Chromatogr 785:3–33. 
Berger TA, Fogleman K, Staats T, Bente P, Crocket I, FarrellW, Osonubi M. 
2000. The development of a semi-preparatory scale supercritical-fluid 
chromatograph for high-through purification of ‘‘combi-chem’’ libra-ries. 
J Biochem Biophys Methods 43:87–111. 
Biasi VD, Haskins N, Organ A, Bateman R, Giles K, Jarvis S. 1999. High 
throughput liquid chromatography/mass spectrometric analyses using 
a novel multiplexed electrospray interface. Rapid Commun Mass 
Spectrom 13:1165–1168. 
Blom KF, Glass B, Sparks R, Combs AP. 2004. Preparative LC-MS 
purification: Improved compound-specific method optimization. 
J Comb Chem 6:874–883. 
Bolan˜os BJ, Ventura MC, Greig MJ. 2003. Preserving the chromatographic 
integrity of high-speed supercritical fluid chromatography separations 
using time-of-flight mass spectrometry. J Comb Chem 5:451–455. 
Bolanos B, Greig M, Ventura M, Farrell W, Aurigemma CM, Li H, Quenzer 
TL, TivelK, Bylund JMR, Tran P, Pham C, Phillipson D. 2004. SFC/MS 
in drug discovery at Pfizer, La Jolla. J Mass Spectrom 238:85–97. 
Borges V, Yang E, Dunn J, Henion J. 2004. High-throughput liquid 
chromatography-tandem mass spectrometry determination of bupro-pion 
and its metabolites in human, mouse and rat plasma using a 
monolithic column. J Chromatogr B 804:277–287. 
Bristow AWT, Webb KS. 2003. Intercomparison study on accurate mass 
measurement of small molecules in mass spectrometry. JAm Soc Mass 
Spectrom 14:1086–1098. 
Burg J. 2004. Parallel LC/MS detection for automatic purification. 
Nachrichten aus der Chemie 52:623–624. 
Busch KL. 1992. Coupling thin layer chromatography with mass spectrom-etry. 
J Planar Chromatogr 5:72–79. 
Cai YQ, Jiang GB, Liu JF, Zhou QX. 2003. Multiwalled carbon nanotubes 
as a solid-phase extraction adsorbent for the determination of bisphenol 
A, 4-n-nonylphenol, and 4-tert-octylphenol. Anal Chem 75:2517– 
2521. 
Carrell T,Wintner EA, Sutherland AJ, Rebek J Jr, Dunayevskiy Y, Vouros P. 
1995. New promise in combinatorial chemistry: Synthesis, character-ization, 
and screening of small-molecule libraries in solution. Chem 
Biol 3:171–183. 
Chen YC, Shiea J, Sunner J. 1998. Thin-layer chromatography mass 
spectrometry using activated carbon, surface-assisted laser desorption/ 
ionization. J Chromatogr A 826:77–86. 
Coe RA, Rathe JO, LeeJW. 2006. Supercritical fluid chromatography-tandem 
mass spectrometry for fast bioanalysis of R/S-warfarin in human 
plasma. J Pharm Biomed Anal 42:573–580. 
Cohen LH, Gusev AI. 2002. Small molecule analysis by MALDI mass 
spectrometry. Anal Bioanal Chem 373:571–586. 
Corra JJ, Kovarika P, Schneidera BB, Hendriksea J, Lobodaa A, Covey TR. 
2006. Design considerations for high speed quantitative mass 
spectrometry with MALDI ionization. J Am Soc Mass Spectrom 17: 
1129–1141. 
Crecelius A, Clench MR, Richards D. 2003. TLC-MALDI in pharmaceutical 
analysis. LC-GC Eur 16:225–229. 
Cremin PA, Zeng L. 2002. High-throughput analysis of natural product 
compound libraries by parallel LC-MS evaporative light scattering 
detection. Anal Chem 74:5492–5500. 
Cuiffi JD, Hayes DJ, Fonash SJ, Brown KN, Jones AD. 2001. Desorption-ionization 
mass spectrometry using deposited nanostructured silicon 
films. Anal Chem 73:1292. 
Dale MJ, Knochenmuss R, Zenobi R. 1996. Graphite/liquid mixed matrices 
for laser desorption/ionization mass spectrometry. Anal Chem 68: 
3321–3329. 
Davis PW, Griffith MC. 2004. High throughput parallel LC/MS/ELSD of 
combinatorial libraries using eight-channel LCT system with MUX 
technology. In: Kyranos JN, editor. High throughput analysis for early 
drug discovery. San Diego: Elsevier. pp. 112–123. 
Dunayevskiy Y, Vouros P, Carrell T, Wintner E, Rebek J Jr. 1995. 
Characterization of the complexity of small molecule libraries by 
electrospray ionization mass spectrometry. Anal Chem 67:2906–2915. 
Dunayevskiy YM, Vouros P, Wintner EA, Shipps GW, Carell T, Rebek J Jr. 
1996. Application of capillary electrophoresis-electrospray ionization 
mass spectrometry in the determination of molecular diversity. Proc 
Natl Acad Sci USA 93:6152–6157. 
Dykes S, Fancy SA, Perkins GL, Pullen FS. 2003. The automation of a 
commercial fourier transform mass spectrometer to provide a quick and 
robust method for determining exact mass for the synthetic chemist. Eur 
J Mass Spectrom 9:73–80. 
Egner BJ, Cardno M, BradleyM. 1995a. Linkers for combinatorial chemistry 
and reaction analysis using solid phase in situ mass spectrometry. 
J Chem Soc Chem Commun 21:2163–2164. 
Egner BJ, Langley GJ, Bradley M. 1995b. Solid phase chemistry: Direct 
monitoring by matrix-assisted laser desorption/ionization time of flight 
mass spectrometry. A tool for combinatorial chemistry. J Org Chem 60: 
2652–2653. 
 FANG ET AL. 
30 Mass Spectrometry Reviews DOI 10.1002/mas
Enjalbal C, Martinez J, Aubagnac JL. 2000. Mass spectrometry in 
combinatorial chemistry. Mass Spectrom Rev 19:139–161. 
Erny GL, Cifuentes A. 2006. Liquid separation techniques coupled with mass 
spectrometry for chiral analysis of pharmaceutical compounds and their 
metabolites in biological fluids. J Pharma Biomed Anal 40: 509– 
515. 
Fang AS, Vouros P, Stacey CC, Kruppa GH, Laukien FH, Wintner EA, 
Carell T, Rebek J. 1998. Characterization of combinatorial libraries 
using electrospray ionization fourier transform ion cyclotron resonance 
mass spectrometry. Comb Chem High Throughput Screen 1:22–33. 
Fang L, Cournoyer J, Demee M, Zhao J, Tokushige D, Yan B. 2002. High-throughput 
liquid chromatography ultraviolet/mass spectrometric 
analysis of combinatorial libraries using an eight-channel multiplexed 
electrospray time-of-flight mass spectrometer. Rapid Commun Mass 
Spectrom 16:1440–1447. 
Fang L, Demee M, Cournoyer J, Sierra T, Young C, Yan B. 2003. Parallel 
high-throughput accurate mass measurement using a nine-channel 
multiplexed electrospray liquid chromatography ultraviolet time-of-flight 
mass spectrometry system. Rapid Commun Mass Spectrom 17: 
1425–1432. 
Felten C, Foret F, Minarik M, Goetzinger W, Karger BL. 2001. Automated 
high-throughput infusion ESI-MS with direct coupling to a microtiter 
plate. Anal Chem 7:1449–1454. 
Fitzgerald MC, Harris K, Shevlin CG, Siuzdak G. 1996. Direct character-ization 
of solid phase resin-bound molecules by mass spectrometry. 
Bioorg Med Chem Lett 6:979–982. 
Foulon C, Danel C, Vaccher C, Yous S, Bonte J-P, Goossens J-F. 2004. 
Determination of ionization constants of N-imidazole derivatives, 
aromatase inhibitors, using capillary electrophoresis and influence of 
substitutes on pKa shifts. J Chromatogr A 131–136. 
Fu KF, Sun YP. 2003. Dispersion and solubilization of carbon nanotubes. 
J Nanosci Nanotechnol 3:351–364. 
Gallagher RT, Balogh MP, Davey P, Jackson MR, Sinclair I, Southern LJ. 
2003. Combined electrospray ionization-atmospheric pressure chem-ical 
ionization source for use in high-throughput LC-MS applications. 
Anal Chem 75:973–977. 
Go EP, Apon JV, Luo G, Saghatelian A, Daniels RH, Sahi V, Dubrow R, 
Cravatt BF,Vertes A, Siuzdak G. 2005. Desorption/ionization on silicon 
nanowires. Anal Chem 77:1641–1646. 
Gobey J, Cole M, Janiszewski J, Covey T, Chau T, Kovarik P, Corr J. 2005. 
Characterization and performance of MALDI on a triple quadrupole 
mass spectrometer for analysis and quantification of small molecules. 
Anal Chem 77:5643–5654. 
GoetzingerW, Kyranos J. 1998. Fast gradient RP-HPLC for high-throughput 
quality control analysis of spatially addressable combinatorial libraries. 
Am Lab 30:27–37. 
Goetzinger WK, Cai H. 2005. Buffer system for the separation of neutral 
and charged small molecules using micellar electrokinetic chromatog-raphy 
with mass spectrometric detection. J Chromatogr A 1079:372– 
381. 
GoetzingerW, Zhang X, Bi G, Towle M, Cherrak D, Kyranos JN. 2004. High 
throughput HPLC/MS purification in support of drug discovery. Int J 
Mass Spectrom 238:153–162. 
Guo Z, Zhang Q, Zou H, Guo B, Ni J. 2002. A method for the analysis of low-mass 
molecules by MALDI-TOF mass spectrometry. Anal Chem 
74:1637–1641. 
Gusev AI. 2000. Interfacing matrix-assisted laser desoption/ionization mass 
spectrometry with column and planar separations. Fresenius J Anal 
Chem 266:691–700. 
Han M, Sunner J. 2000. An activated carbon substrate surface for laser 
desorption mass spectrometry. Am Soc Mass Spectrom 11:644–649. 
Haner RL, Llanos W, Mueller L. 2000. Small volume flow probe for 
automated direct-injection NMR analysis: Design and performance. 
J Magn Reson 143:69–78. 
MASS SPECTROMETRY ANALYSIS  
Hauck HE, Schulz M. 2002. Ultrathin-layer chromatography. J Chromatogr 
Sci 40:1–3. 
Hauck HE, Bund O, Fischer M, Schulz M. 2001. Ultra-thin layer 
chromatography (UTLC)-a new dimension in thin-layer chromatog-raphy. 
J Planar Chromatogr 14:234–236. 
Hauser-Fang A, Vouros P. 2000. The use of mass spectrometry. In: Swartz 
ME, editor. Analytical techniques in combinatorial chemistry. New 
York: Marcel Dekker. pp. 29–63. 
Hayward MJ, Snodgrass JT, Thomson ML. 1993. Flow injection Thermos-pray 
mass spectrometry for the automated analysis of potential 
agricultural chemicals. Rapid Commun Mass Spectrom 7:85–91. 
Herniman JM, Bristow TWT, O’Connor G, Jarvis J, Langley GJ. 2004. 
Improved precision and accuracy for high-performance liquid chroma-tography/ 
fourier transform ion cyclotron resonance mass spectrometric 
exact mass measurement of small molecules from the simultaneous and 
controlled introduction of internal calibrants via a second electrospray 
nebuliser. Rapid Commun Mass Spectrom 18:3035–3040. 
Herniman JM, Langley GJ, Bristow TWT, O’Connor G. 2005. The validation 
of exact mass measurements for small molecules using FT-ICRMS for 
improved confidence in the selection of elemental formulas. J Am Soc 
Mass Spectrom 16:1100–1108. 
Hsieh Y, Chintala M, Mei H, Agans J, Brisson J-M, Ng KI, KorfmacherWA. 
2001. Quantitative screening and matrix effect studies of drug discovery 
compounds in monkey plasma using fast-gradient liquid chromatog-raphy/ 
tandem mass spectrometry. Rapid Commun Mass Spectrom 
15:2481. 
Hsieh Y, Brisson J-M,Wang G. 2003. Fast HPLC-MS/MS analyses for small 
molecules. Am Pharm Rev 6:14–20. 
Iijima S. 1991. Helical microtubules of graphitic carbon. Nature 354:56– 
5658. 
Iijima S, Ichihashi T. 1993. Single-shell carbon nanotubes of 1-nm diameter. 
Nature 363:603–605. 
Irving M, Krueger CA,Wade JV, Hodges JC, Leopold K, Collins N, Chan C, 
Shaqair S, Shornikov A, Yan B. 2004. High-throughput purification of 
combinatorial libraries II: Automated separation of single diastereom-ers 
from a 4-amido-pyrrolidone library containing intentional diaster-eomer 
pairs. J Comb Chem 6:478–486. 
Isbell J, Xu R, Cai Z, Kassel DB. 2002. Realities of high-throughput liquid 
chromatography/mass spectrometry purification of large combinatorial 
libraries: A report on overall sample throughput using parallel 
purification. J Comb Chem 4:600–611. 
Ka´lasz H, Ba´thori M. 2001. Pharmaceutical applications of TLC. LC-GC Eur 
14:311–321. 
Kassel DB. 2001. Combinatorial chemistry and mass spectrometry in the 21st 
century drug discovery laboratory. Chem Rev 101:255–268. 
Keifer PA, Smallcombe SH,Williams EH, Salomon KE, Mendez G, Belletire 
JL, Moore CD. 2000. Direct-injection NMR (DI-NMR): A flow NMR 
technique for the analysis of combinatorial chemistry libraries. J Comb 
Chem 2:151–171. 
Kinumi T, Saisu T, Takayama M, Niwa H. 2000. Matrix-assisted laser 
desorption/ionization time-of-flight mass spectrometry using as 
inorganic particle matrix for small molecule analysis. J Mass Spectrom 
35:417. 
Kleintop BL, Zhang H, Ray KL. 2004. Leveraging the use of modern mass 
spectrometry instrumentation in small molecule drug development.Am 
Pharm Rev 7:124–127. 
Koppitz M, Brailsford A,Wenz M. 2005. Maximizing automation in LC/MS 
high-throughput analysis and purification. J Comb Chem 7:714– 
720. 
Krutchinsky AN, Chait BT. 2002. On the nature of chemical noise in MALDI 
mass spectra. J Am Soc Mass Spectrom 13:129. 
Kyranos JN, Cai H, Zhang B, Goetzinger WK. 2001a. High-throughput 
techniques for compound characterization and purification. Curr Opin 
Drug Discov Dev 4:719–728. 
Mass Spectrometry Reviews DOI 10.1002/mas 31
Kyranos JN, Cai H, Wei D, Goetzinger WK. 2001b. High-throughput high-performance 
liquid chromatography/mass spectrometry for modern 
drug discovery. Curr Opin Biotech 12:105–111. 
Lazar IM, Ramsey RS, Sundberg S, Ramsey JM. 1999. Subattomole-sensitivity 
microchip nanoelectrospray source with time-of-flight mass 
spectrometry detection. Anal Chem 71:3627. 
Lee PJ, Chen W, Gebler JC. 2004. Qualitative and quantitative analysis of 
small amine molecules by MALDI-TOF mass spectrometry through 
charge derivatization. Anal Chem 76:4888–4893. 
Letot E, Koch G, Falchetto R, Bovermann G, Oberer L, Roth H-J. 2005. 
Quality control in combinatorial chemistry: Determinations of amounts 
and comparison of the ‘‘purity’’ of LC-MS-purified samples by NMR, 
LC-UVand CLND. J Comb Chem 7:364–371. 
Lewis WG, Shen ZX, Finn MG, Siuzdak G. 2003. Desorption/ionization on 
silicon (DIOS) mass spectrometry: Background and applications. Int J 
Mass Spectrom 226:107–116. 
Lindon JC, Nicholson JK, Wilson ID. 2000. Directly coupled HPLC-NMR 
and HPLC-NMR-MS in pharmaceutical research and development. 
J Chromatogr B 748:233–258. 
Liu J-F, Jo¨nsson JA° , Jiang G-B. 2005. Application of ionic liquids in 
analytical chemistry. Trends Anal Chem 24:20–27. 
Louden D, Handley A, Taylor S, Lenz E, Miller S,Wilson ID, Sage A. 2000. 
Flow injection spectroscopic analysis of model drugs using on-lineUV-diode 
array, FT-infrared and 1H-nuclear magnetic resonance spectro-scopy 
and time-of-flight mass spectrometry. Analyst 125:927–931. 
Mallet CR, Lu Z, Mazzeo JR. 2004. A study of ion suppression effects in 
electrospray ionization from mobile phase additives and solid-phase 
extracts. Rapid Commun Mass Spectrom 18:49. 
Mallis LM, Sarkahian AB, Kulishoff JMJr,Watts WL Jr. 2002. Open-access 
liquid chromatography/mass spectrometry in a drug discovery environ-ment. 
J Mass Spectrom 37:889–896. 
Mehl JT, Hercules DM. 2000. Direct TLC-MALDI coupling using a hybrid 
plate. Anal Chem 72:68–73. 
Millot MC. 2003. Separation enantiomers by liquid chromatography 
and capillary electrophoresis, using immobilized proteins as chiral 
selectors. J Chromatogr B 797:131–159. 
Moini M. 2002. Capillary electrophoresis mass spectrometry and its 
application to the analysis of biological mixtures. Anal Bioanal Chem 
373:466–480. 
Morand KL, Burt TM, Regg BT, Chester TL. 2001. Techniques for increasing 
the throughput of flow injection mass spectrometry. Anal Chem 
73:247–252. 
Mukherjee PS. 2007. Validation of direct assay of an aqueous formulation of 
drug compound AZY by chiral supercritical fluid chromatography 
(SFC). J Pharm Biomed Anal 43:464–470. 
Nawrocki JP, Wigger M, Watson CH, Hayes TW, Senko MW, Benner SA, 
Eyler JR. 1996. Analysis of combinatorial libraries using electrospray 
ionization fourier transform ion cyclotron resonance mass spectrom-etry. 
Rapid Commun Mass Spectrom 10:1860–1864. 
Pan C, Xu S, Zou H, Guo Z, Zhang Y, Guo B. 2004. Carbon nanotubes as 
adsorbent of solid-phase extraction and matrix for laser desorption/ 
ionization mass spectrometry. J Am Soc Mass Spectrom 16:263– 
270. 
Pan C, Xu S, Hu L, Su X, Ou J, Zou H, Guo Z, Zhang Y, Guo B. 2005. Using 
oxidized carbon nanotubes as matrix for analysis of small molecules by 
MALDI-TOF MS. J Am Soc Mass Spectrom 16:883–892. 
Paulson J. 2005. A stand-alone monitoring system for reliable unattended 
operation of a commercial preparative LC/MS purification system. 
JALA 10:6–15. 
Peake DA, Duckworth DC, Perun TJ, Scott WL, Kulanthaivel P, Strege MA. 
2005. Analytical and biological evaluation of high throughput screen 
actives using evaporative light scattering, chemiluminescent nitrogen 
detection, and accurate mass LC-MS-MS. Comb Chem High 
Throughput Screen 8:477–487. 
Peng XJ, Li YH, Luan ZK, Di ZC, Wang HY, Tian BH, Jia ZP. 2003. 
Adsorption of 1,2-dichlorobenzene from water to carbon nanotubes. 
Chem Phys Lett 376:154–158. 
Peterson DS, Luo Q, Hilder EF, Svec F, Fre´chet JMJ. 2004. Porous polymer 
monolith for surface-enhanced laser desorption/ionization time-of-flight 
mass spectrometry. Rapid Commun Mass Spectrom 18:1504– 
1512. 
Pinkston JD,Wen D, Morand KL, Tirey DA, Stanton DT. 2006. Comparison 
of LC/MS and SFC/MS for screening of a large and diverse library of 
pharmaceutically relevant compounds. Anal Chem 78:7467–7472. 
Poulsen SA. 2006. Direct screening of a dynamic combinatorial library using 
mass spectrometry. J Am Soc Mass Spectrom 17:1074–1080. 
Pullen FS, Perkins GL, Burton KI,Ware RS, Taegue MS, Kiplinger JP. 1995a. 
Putting mass spectrometry in the hands of the end user. JAm Soc Mass 
Spectrom 6:394–399. 
Pullen FS, Swanson AG, Newman MJ, Richards DS. 1995b. ‘Online’ liquid 
chromatography/nuclear magnetic resonance mass spectrometry—A 
powerful spectroscopic tool for the analysis of mixtures of pharma-ceutical 
interest. Rapid Commun Mass Spectrom 9:1003–1006. 
Rauha J, Vuorela H, Kostiainen R. 2001. Effect of eluent on the ionization 
efficiency of flavonoids by ion spray, atmospheric pressure chemical 
ionization, and atmospheric pressure photoionization mass spectrom-etry. 
J Mass Spectrom 36:1269. 
Reyzer ML, Hsieh Y, Ng K, Korfmacher WA, Caprioli RM. 2003. Direct 
analysis of drug candidates in tissue by matrix-assisted laser desorption/ 
ionization mass spectrometry. J Mass Spectrom 38:1081–1092. 
Richmond R. 2000. The analytical characterization of sub-minute measure-ment 
duty cycles in flow-injection analysis mass spectrometry, by their 
carry-over. Anal Chim Acta 403:287–294. 
Richmond R, Go¨rlach E. 1999a. The automatic visualization of carry-over in 
high-throughput flow injection analysis mass spectrometry. Anal Chim 
Acta 390:175. 
Richmond R, Go¨rlach E. 1999b. Sorting measurement queues to speed up the 
flow injection analysis mass spectrometry of combinatorial chemistry 
syntheses. Anal Chim Acta 394:33–342. 
Ripka WC, Barker G, Krakover J. 2001. High-throughput purification of 
compound libraries. Drug Disc Today 6:471–477. 
Robb DB, Covey TR, Bruins AP. 2000. Atmospheric pressure photo-ionization: 
An ionization method for liquid chromatography-mass 
spectrometry. Anal Chem 72:3653–3659. 
Sage AB, Little D, Giles K. 2000. Using parallel LC-MS and LC-MS-MS to 
increase sample throughput. LC-GC Mag Separation Sci 18:S20–S29. 
Salo PK, Pertovaara AM, Salo VMA, Salomies HEM,Konstiainen RK. 2003. 
High-performance thin-layer chromatography method for assessment 
of the quality of combinatorial libraries, and comparison with liquid 
chromatography-ultraviolet-mass spectrometry. J Comb Chem 5:223– 
232. 
Santos LS, Haddad R, Hoehr NF, Pilli RA, Eberlin MN. 2004. Fast screening 
of low molecular weight compounds by thin-layer chromatography and 
‘‘on-spot’’ MALDI-TOF mass spectrometry. Anal Chem 76:2144– 
2147. 
Schmid DG, Grosche P, Bandel H, Jung G. 2001. FTICR-mass spectrometry 
for high-resolution analysis in combinatorial chemistry. Biotechnol 
Bioeng (Comb Chem) 71:149–161. 
Shah N, Gao M, Tsutsui K, Lu A, Davis J, Scheuerman R, FitchW. 2000. A 
novel approach to high-throughput quality control of parallel synthesis 
libraries. J Comb Chem 2:453–460. 
Shen Z, Thomas JJ, Averbuj C, Broo KM, Engelhard M, Crowell JE, Finn 
MG, Siuzdak G. 2001. Porous silicon as a versatile platform for laser 
desorption/ionization mass spectrometry. Anal Chem 73:612. 
SlenoLVD. 2005. Some fundamental and technical aspects of the quantitative 
analysis of pharmaceutical drugs by matrix-assisted laser desorption/ 
ionization mass spectrometry. Rapid Commun Mass Spectrom 19: 
1928–1936. 
 FANG ET AL. 
32 Mass Spectrometry Reviews DOI 10.1002/mas
Smyth WF. 2005. Recent applications of capillary electrophoresis-electro-spray 
ionisation-mass spectrometry in drug analysis. Electrophoresis 
26:1334–1357. 
Somsen GW, Morden W, Wilson ID. 1995. Planar chromatography coupled 
with spectroscopic techniques. J Chromatogr A 703:613–665. 
Souverain S, Geiser L, Rudaz S,Veuthey J-L. 2006. Strategies for rapid chiral 
analysis by capillary electrophoresis. J Pharm Biomed Anal 40:235– 
241. 
Spreen RC, Schaffter LM. 1996. Open access MS: A walk-up MS service. 
Anal Chem 68:414A–419A. 
Stolzberg LJ, Patel P. 2004. Small molecule matrix-assisted laser desorption/ 
ionization time-of-flight mass spectrometry using a polymer matrix. 
Rapid Commun Mass Spectrom 18:1455–1458. 
Sunner J, Dratz E, Chen YC. 1995. Graphite surface assisted laser desorption/ 
ionization time-of-flight mass spectrometry of peptides and proteins 
form liquid solutions. Anal Chem 67:4335–4342. 
Su¨ssmuth RD, Jung G. 1999. Impact of mass spectrometry on combinatorial 
chemistry. J Chromatogr B 725:49–65. 
Tanaka N, Kobayashi H, Ishizuka N, Minakuchi H, Nakanishi K, Hosoya K, 
Ikehami T. 2002. Monolithic silica columns for high-efficiency 
chromatographic separations. J Chromatogr A 965:35. 
Tiller PR, Romanyshyn LA, Leslie A, Neue UD. 2003. Fast LC/MS in the 
analysis of small molecules. Anal Bioanal Chem 377:788–802. 
Toribio L, Alonso C, del Nozal MJ, Bernal JL, Martin MT. 2006. 
Sempreparative enantiomeric separation of omeprazole by supercritical 
fluid chromatography. J Chromatogr A 1137:30–35. 
Triolo A, Altamura M, Cardinali F, Sisto A, Maggi CA. 2001a. Mass 
spectrometry and combinatorial chemistry: A short outline. J Mass 
Spectrom 36:1249–1259. 
Triolo A, Altamura M, Cardinale F, Sisto A, Maggi CA. 2001b. Mass 
spectrometry and combinatorial chemistry: A short outline. J Mass 
Spectrom 36:1249. 
Visky D, Jimidar I, Van Ael W, Vennekens T, Redlich D, De Smet M. 2005. 
Capillary electrophoresis-mass spectrometry in impurity profiling of 
pharmaceutical products. Electrophoresis 26:1541–1549. 
Wachs T, Henion J. 2001. Electrospray device for coupling microscale 
separations and other miniaturized devices with electrospray mass 
spectrometry. Anal Chem 73:632–638. 
Walk TB, Trautwein AW, Richter H, Jung G. 1999. ESI Fourier transform ion 
cyclotron resonance mass spectrometry (ESI-FT-ICR-MS): A rapid 
high-resolution analytical method for combinatorial compound libra-ries. 
Angew Chem Int Ed Engl 38:1763–1765. 
Wan H, Thompson RA. 2005. Capillary electrophoresis technologies for 
screening in drug discovery. Drug Discov Today Technol 2:171–178. 
Wang T, Zeng L, Strader T, Burton L, Kassel DB. 1998. A new ultra-high 
throughput method for characterizing combinatorial libraries incorpo-rating 
a multiple probe autosampler coupled with flow injection mass 
spectrometry analysis. Rapid Commun Mass Spectrom 12:1123. 
Wang T, Zeng L, Cohen J, Kassel DB. 1999.Amultiple electrospray interface 
for parallel mass spectrometric analyses of compound libraries. Comb 
Chem High Throuput Screen 2:327–334. 
Wang AQ, Zeng W, Musson DG, Rogers JD, Fisher AL. 2002. A rapid and 
sensitive liquid chromatography/negative ion tandem mass spectrom-etry 
MASS SPECTROMETRY ANALYSIS  
method for the determination of an indolocarbazole in human 
plasma using internal standard (IS) 96-well diatomaceous earth plates 
for solid-liquid extraction. Rapid Commun Mass Spectrom 16:975. 
Wehr T. 2003. Fast LC for high-throughput LC-MS. LCGC North America 
21, Part 2 (Suppl):75–78. 
Wei J, Buriak JM, Siuzdak G. 1999. Desorption-ionization mass spectrometry 
on porous silicon. Nature 399:243–246. 
Weins C, Hauck HE. 1996. Advances and developments in thin layer 
chromatography. LC-GC Int 9:710–717. 
Williams SM, Brodbelt JS, Huang Z, Lai H, Marchand AP. 2003. Complex-ation 
of silver and co-recovered metals with novel aza-crown ether 
macrocycles by electrospray ionization mass spectrometry. Analyst 
128:1352–1359. 
Wilson ID. 1999. The state of the art in thin-layer chromatography-mass 
spectrometry: A critical appraisal. J Chromatogr A 856:429–442. 
Wilson ID, Lindon JC, Nicholson JK. 2000. Advancing hyphenated 
chromatographic systems. Anal Chem 72:534A–542A. 
Winger BE, Campana JE. 1996. Characterization of combinatorial peptide 
libraries by electrospray ionization fourier transform mass spectrom-etry. 
Rapid Commun Mass Spectrom 10:1811–1813. 
Wu JY, Chen Y. 2002. A novel approach of combining thin-layer 
chromatography with surface-assisted laser desorption/ionization 
(SALDI) time-of-flight mass spectrometry. J Mass Spectrom 37:85–90. 
Xu SY, Li YF, Zou HF, Qiu JS, Guo Z, Guo BC. 2003. Carbon nanotubes as 
assisted matrix for laser desorption/ionization time-of-flight mass 
spectrometry. Anal Chem 75:6191–6195. 
Yan B, Chu YH, Shapiro M, Richmond R, Chin J, Liu L, Yu Z. 2000. 
Analytical methods in combinatorial chemistry. In: Fenniri H, editor. 
Combinatorial chemistry: a practical approach. Oxford UK: Oxford 
University Press. pp. 263–286. 
Yan B, Collins N, Wheatley J, Irving M, Leopold K, Chan C, Shornikov A, 
Fang L, Lee A, Stock M, Zhao J. 2004. High-throughput purification of 
combinatorial libraries I: A high-throughput purification system using 
an accelerated retention window approach. J Comb Chem 6:255–261. 
Yu K, Balogh M. 2000. A protocol for high-throughput drug mixture 
quantitation: Fast LC-MS or flow injection analysis-MS? LC-GC 19: 
60–72. 
Yurek D, Branch DL, Kuo M-S. 2002. Development of a system to evaluate 
compound identity, purity, and concentration in a single experiment and 
its application in quality assessment of combinatorial libraries and 
screening hits. J Comb Chem 4:138–148. 
Zhang QC, Zou HF, Guo Z, Zhang Q, Chen XM, Ni JY. 2001. Matrix-assisted 
laser desorption/ionization mass spectrometry using porous silicon and 
silica gel as matrix. Rapid Commun Mass Spectrom 15:217–223. 
Zhang LK, Rempel D, Pramanik BN, Gross ML. 2005. Accurate mass 
measurements by fourier transform mass spectrometry. Mass Spectrom 
Rev 24:286–309. 
Zhang X, Towle MH, Felice CF, Flament JH, Goetzinger WK. 2006. 
Development of a mass-directed preparative supercritical fluid 
chromatography purification system. J Comb Chem 8:705–714. 
Zweigenbaum J, Heinnig K, Steinborner S, Wachs T, Henion J. 1999. High-throughput 
bioanalytical LC/MS/MS determination of benzodiazepines 
in human urine:1000 samples per 12 hours. Anal Chem 71:2294–2300. 
Annette S. Fang, Ph.D. Dr. Fang received her undergraduate education in Germany and her 
Ph.D. degree in Analytical Chemistry from Northeastern University. She was a Staff 
Investigator in the Analytical Department at ArQule, primarily focused on purification of 
new chemical entities synthesized for early drug discovery. Her prior work experiences 
include positions at Research Biochemicals, Inc., Natick, MA and Bruker Daltonics, 
Billerica, MA. 
Mass Spectrometry Reviews DOI 10.1002/mas 33
Xiusheng Miao, Ph.D., Senior Investigator, Preclinical Development and Clinical 
Pharmacology, ArQule, Inc., Woburn, MA. Dr. Miao is currently responsible for in vitro 
and in vivo drug metabolite identification and bioanalytical method development. He 
obtained his Ph.D. from the Chinese Academy of Sciences, China and has co-authored over 
40 journal articles and book sections. 
Peter W. Tidswell, Ph.D., Associate Research Fellow, Pfizer, Inc. Dr. Tidswell leads the 
purification laboratory at the St. Louis research site, which supports the medicinal chemistry 
department and therapeutic programs. Prior to joining Pfizer, he was a Group Leader in the 
Chemical Technologies’ analytical department for ArQule, where he led the Character-ization 
group. His other experience encompasses positions at the Bristol-Myers Squibb 
Pharmaceutical Research Institute, EvotecOAI and ChiroScience. Dr. Tidswell received his 
Ph.D. under the direction of Dr. Mel Kilner in Organometallic Chemistry from the 
University of Durham, UK. 
Marc H. Towle, Scientist, Discovery Analytical,Vertex Pharmaceuticals, Inc., Cambridge, 
MA. Mr. Towle provides analytical development support to assess the purity, potency and 
stability of drug substances and drug products for late research, pre-clinical, and phase I 
development. Prior to joining Vertex, he was a Senior Investigator at ArQule, where he 
managed the analytical purification group responsible for supporting high throughput 
production of chemical libraries for early drug discovery. His prior experience includes 
research positions with BioDevelopment Laboratories, Arthur D. Little and Groundwater 
Technology. Mr. Towle received his BS in Biochemistry from the University of New 
Hampshire. 
Wolfgang K. Goetzinger, Ph.D., Director Research, Molecular Structure, Amgen, Inc., 
Cambridge, MA. Dr. Goetzinger is responsible for the Discovery Analytical group at 
Amgen, Inc. where he leads a team supporting all analytical aspects of small molecule 
pharmaceutical discovery. Prior to joining Amgen, he was Director of Analytical Chemistry 
at ArQule, Inc., where he managed a group focused on high throughput analytical 
approaches to support parallel synthesis efforts. Dr. Goetzinger worked as a Post Doctoral 
Fellowunder Professor Barry L. Karger at Northeastern University, where he focused on life 
science applications of capillary electrophoresis. He received his Ph.D. from the University 
Saarland under Professor Heinz Engelhardt working on the synthesis and characterization 
of chiral stationary phases for the separation of enantiomers. 
James N. Kyranos, Ph.D., Vice President, Preclinical Development, Wolfe Laboratories, 
Watertown, MA. Dr. Kyranos leads the technical and business operations of WLI’s 
preclinical development services focused on bioanalytical and pharmaceutical sciences 
support. Prior to joining WLI, he was Vice President of Chemical Technologies for ArQule, 
where he managed the chemistry services business focused on providing early discovery 
services to the pharmaceutical industry. His other experience includes leadership positions 
at BioDevelopment Laboratories and Arthur D. Little. Dr.Kyranos received his Ph.D. under 
the direction of Professor Paul Vouros in Analytical Chemistry from Northeastern 
University. 
 FANG ET AL. 
34 Mass Spectrometry Reviews DOI 10.1002/mas

Más contenido relacionado

La actualidad más candente

EXPERIMENTAL IMPLEMENTATION OF EMBARRASINGLY PARALLEL PROCESS IN ANALYSIS OF ...
EXPERIMENTAL IMPLEMENTATION OF EMBARRASINGLY PARALLEL PROCESS IN ANALYSIS OF ...EXPERIMENTAL IMPLEMENTATION OF EMBARRASINGLY PARALLEL PROCESS IN ANALYSIS OF ...
EXPERIMENTAL IMPLEMENTATION OF EMBARRASINGLY PARALLEL PROCESS IN ANALYSIS OF ...ijesajournal
 
Analytical method development and validation
Analytical method development and validationAnalytical method development and validation
Analytical method development and validationCreative Peptides
 
Mass Spectrometry: Protein Identification Strategies
Mass Spectrometry: Protein Identification StrategiesMass Spectrometry: Protein Identification Strategies
Mass Spectrometry: Protein Identification StrategiesMichel Dumontier
 
Harnessing the power of CCS for routine screening applications on the UNIFI i...
Harnessing the power of CCS for routine screening applications on the UNIFI i...Harnessing the power of CCS for routine screening applications on the UNIFI i...
Harnessing the power of CCS for routine screening applications on the UNIFI i...Waters Corporation - Chemical Materials
 
Vol 1,issue 7 Radiation therapy treatment unit dose-rate effects on metal–oxi...
Vol 1,issue 7 Radiation therapy treatment unit dose-rate effects on metal–oxi...Vol 1,issue 7 Radiation therapy treatment unit dose-rate effects on metal–oxi...
Vol 1,issue 7 Radiation therapy treatment unit dose-rate effects on metal–oxi...IJAMHC
 
An algorithm for detection of tuberculosis bacilli in Ziehl-Neelsen sputum sm...
An algorithm for detection of tuberculosis bacilli in Ziehl-Neelsen sputum sm...An algorithm for detection of tuberculosis bacilli in Ziehl-Neelsen sputum sm...
An algorithm for detection of tuberculosis bacilli in Ziehl-Neelsen sputum sm...IJECEIAES
 
Mass Spectrometry Applications and spectral interpretation: Basics
Mass Spectrometry Applications and spectral interpretation: BasicsMass Spectrometry Applications and spectral interpretation: Basics
Mass Spectrometry Applications and spectral interpretation: BasicsShreekant Deshpande
 
Innovative Technique for Gene Selection in Microarray Based on Recursive Clus...
Innovative Technique for Gene Selection in Microarray Based on Recursive Clus...Innovative Technique for Gene Selection in Microarray Based on Recursive Clus...
Innovative Technique for Gene Selection in Microarray Based on Recursive Clus...AM Publications
 
An Introduction to:In Vitro-In VivoExtrapolation (IVIVE)
An Introduction to:In Vitro-In VivoExtrapolation (IVIVE)An Introduction to:In Vitro-In VivoExtrapolation (IVIVE)
An Introduction to:In Vitro-In VivoExtrapolation (IVIVE)mjamei
 
Simultaneous loading of 5-florouracil and SPIONs in HSA nanoparticles: Optimi...
Simultaneous loading of 5-florouracil and SPIONs in HSA nanoparticles: Optimi...Simultaneous loading of 5-florouracil and SPIONs in HSA nanoparticles: Optimi...
Simultaneous loading of 5-florouracil and SPIONs in HSA nanoparticles: Optimi...Nanomedicine Journal (NMJ)
 
"Neurotech seminar" on Mass Spectrometry
"Neurotech seminar" on Mass Spectrometry"Neurotech seminar" on Mass Spectrometry
"Neurotech seminar" on Mass SpectrometryIvan Weinsanto
 
Aapm tg63 on hip prosthesis consideration during pelvic radiotherapy
Aapm tg63 on hip prosthesis consideration during pelvic radiotherapyAapm tg63 on hip prosthesis consideration during pelvic radiotherapy
Aapm tg63 on hip prosthesis consideration during pelvic radiotherapykaruppusamya
 
Foley 2011 Cochrane protocol
Foley 2011 Cochrane protocolFoley 2011 Cochrane protocol
Foley 2011 Cochrane protocolRuth Foley
 
Mass spectrometry basic principles
Mass spectrometry basic principlesMass spectrometry basic principles
Mass spectrometry basic principlesRania Elsharkawy
 
A Novel Eclectic Approach for Cancer Therapy with Liquid Knife & Immuno Thera...
A Novel Eclectic Approach for Cancer Therapy with Liquid Knife & Immuno Thera...A Novel Eclectic Approach for Cancer Therapy with Liquid Knife & Immuno Thera...
A Novel Eclectic Approach for Cancer Therapy with Liquid Knife & Immuno Thera...CrimsonpublishersCancer
 

La actualidad más candente (20)

EXPERIMENTAL IMPLEMENTATION OF EMBARRASINGLY PARALLEL PROCESS IN ANALYSIS OF ...
EXPERIMENTAL IMPLEMENTATION OF EMBARRASINGLY PARALLEL PROCESS IN ANALYSIS OF ...EXPERIMENTAL IMPLEMENTATION OF EMBARRASINGLY PARALLEL PROCESS IN ANALYSIS OF ...
EXPERIMENTAL IMPLEMENTATION OF EMBARRASINGLY PARALLEL PROCESS IN ANALYSIS OF ...
 
Analytical method development and validation
Analytical method development and validationAnalytical method development and validation
Analytical method development and validation
 
Mass Spectrometry: Protein Identification Strategies
Mass Spectrometry: Protein Identification StrategiesMass Spectrometry: Protein Identification Strategies
Mass Spectrometry: Protein Identification Strategies
 
Harnessing the power of CCS for routine screening applications on the UNIFI i...
Harnessing the power of CCS for routine screening applications on the UNIFI i...Harnessing the power of CCS for routine screening applications on the UNIFI i...
Harnessing the power of CCS for routine screening applications on the UNIFI i...
 
Vol 1,issue 7 Radiation therapy treatment unit dose-rate effects on metal–oxi...
Vol 1,issue 7 Radiation therapy treatment unit dose-rate effects on metal–oxi...Vol 1,issue 7 Radiation therapy treatment unit dose-rate effects on metal–oxi...
Vol 1,issue 7 Radiation therapy treatment unit dose-rate effects on metal–oxi...
 
An algorithm for detection of tuberculosis bacilli in Ziehl-Neelsen sputum sm...
An algorithm for detection of tuberculosis bacilli in Ziehl-Neelsen sputum sm...An algorithm for detection of tuberculosis bacilli in Ziehl-Neelsen sputum sm...
An algorithm for detection of tuberculosis bacilli in Ziehl-Neelsen sputum sm...
 
Paper1
Paper1Paper1
Paper1
 
AAPM REPORT 84 TG 43
AAPM REPORT 84 TG 43AAPM REPORT 84 TG 43
AAPM REPORT 84 TG 43
 
Mass Spectrometry Applications and spectral interpretation: Basics
Mass Spectrometry Applications and spectral interpretation: BasicsMass Spectrometry Applications and spectral interpretation: Basics
Mass Spectrometry Applications and spectral interpretation: Basics
 
Innovative Technique for Gene Selection in Microarray Based on Recursive Clus...
Innovative Technique for Gene Selection in Microarray Based on Recursive Clus...Innovative Technique for Gene Selection in Microarray Based on Recursive Clus...
Innovative Technique for Gene Selection in Microarray Based on Recursive Clus...
 
An Introduction to:In Vitro-In VivoExtrapolation (IVIVE)
An Introduction to:In Vitro-In VivoExtrapolation (IVIVE)An Introduction to:In Vitro-In VivoExtrapolation (IVIVE)
An Introduction to:In Vitro-In VivoExtrapolation (IVIVE)
 
Simultaneous loading of 5-florouracil and SPIONs in HSA nanoparticles: Optimi...
Simultaneous loading of 5-florouracil and SPIONs in HSA nanoparticles: Optimi...Simultaneous loading of 5-florouracil and SPIONs in HSA nanoparticles: Optimi...
Simultaneous loading of 5-florouracil and SPIONs in HSA nanoparticles: Optimi...
 
"Neurotech seminar" on Mass Spectrometry
"Neurotech seminar" on Mass Spectrometry"Neurotech seminar" on Mass Spectrometry
"Neurotech seminar" on Mass Spectrometry
 
Aapm tg63 on hip prosthesis consideration during pelvic radiotherapy
Aapm tg63 on hip prosthesis consideration during pelvic radiotherapyAapm tg63 on hip prosthesis consideration during pelvic radiotherapy
Aapm tg63 on hip prosthesis consideration during pelvic radiotherapy
 
Biotech 2012 spring-2-protein_chips
Biotech 2012 spring-2-protein_chipsBiotech 2012 spring-2-protein_chips
Biotech 2012 spring-2-protein_chips
 
nanomagnetosols
nanomagnetosolsnanomagnetosols
nanomagnetosols
 
Foley 2011 Cochrane protocol
Foley 2011 Cochrane protocolFoley 2011 Cochrane protocol
Foley 2011 Cochrane protocol
 
Mass spectrometry basic principles
Mass spectrometry basic principlesMass spectrometry basic principles
Mass spectrometry basic principles
 
Mass spectroscopy
Mass spectroscopyMass spectroscopy
Mass spectroscopy
 
A Novel Eclectic Approach for Cancer Therapy with Liquid Knife & Immuno Thera...
A Novel Eclectic Approach for Cancer Therapy with Liquid Knife & Immuno Thera...A Novel Eclectic Approach for Cancer Therapy with Liquid Knife & Immuno Thera...
A Novel Eclectic Approach for Cancer Therapy with Liquid Knife & Immuno Thera...
 

Similar a Mass Spectrometry reviews 2008 v27 p20

Crimson Publishers -A Sensor Multiplatform for Non Invasive Diagnosis of Pros...
Crimson Publishers -A Sensor Multiplatform for Non Invasive Diagnosis of Pros...Crimson Publishers -A Sensor Multiplatform for Non Invasive Diagnosis of Pros...
Crimson Publishers -A Sensor Multiplatform for Non Invasive Diagnosis of Pros...CrimsonPublishers-SBB
 
Dorobantu_A_Luminex-bead-based assay - Feedback.pdf
Dorobantu_A_Luminex-bead-based assay - Feedback.pdfDorobantu_A_Luminex-bead-based assay - Feedback.pdf
Dorobantu_A_Luminex-bead-based assay - Feedback.pdfAdinaGeorgiana7
 
Limitations and Advantages in Assessing Adenovirus Homogeneity by Laser Light...
Limitations and Advantages in Assessing Adenovirus Homogeneity by Laser Light...Limitations and Advantages in Assessing Adenovirus Homogeneity by Laser Light...
Limitations and Advantages in Assessing Adenovirus Homogeneity by Laser Light...KBI Biopharma
 
Leibniz: A Digital Scientific Notation
Leibniz: A Digital Scientific NotationLeibniz: A Digital Scientific Notation
Leibniz: A Digital Scientific Notationkhinsen
 
Journal Combinatorial Chemistry 2006 v8 p820
Journal Combinatorial Chemistry 2006 v8 p820Journal Combinatorial Chemistry 2006 v8 p820
Journal Combinatorial Chemistry 2006 v8 p820Peter Tidswell
 
Mass Spectrometry Basic By Inam
Mass Spectrometry Basic By InamMass Spectrometry Basic By Inam
Mass Spectrometry Basic By InamInamul Hasan Madar
 
ANALYTICAL TECHNIQUES (CHROMATOGRAPHY, SPECTROSCOPY, ELECTROPHOROSIS) IN PHAR...
ANALYTICAL TECHNIQUES (CHROMATOGRAPHY, SPECTROSCOPY, ELECTROPHOROSIS) IN PHAR...ANALYTICAL TECHNIQUES (CHROMATOGRAPHY, SPECTROSCOPY, ELECTROPHOROSIS) IN PHAR...
ANALYTICAL TECHNIQUES (CHROMATOGRAPHY, SPECTROSCOPY, ELECTROPHOROSIS) IN PHAR...imran khan
 
Analytical Considerations When Monitoring Pain Medications by LC-MS/MS
Analytical Considerations When Monitoring Pain Medications by LC-MS/MSAnalytical Considerations When Monitoring Pain Medications by LC-MS/MS
Analytical Considerations When Monitoring Pain Medications by LC-MS/MSDavid Masters-Moore
 
Quantitative mass spec imaging
Quantitative mass spec imagingQuantitative mass spec imaging
Quantitative mass spec imagingSteve Cepa
 
Mass spectrometry it's use applications methods
Mass spectrometry it's use applications methodsMass spectrometry it's use applications methods
Mass spectrometry it's use applications methodsSadRajput
 
A review of use of enantiomers in homeopathy
A review of use of enantiomers in homeopathyA review of use of enantiomers in homeopathy
A review of use of enantiomers in homeopathyhome
 
Hdat pdf-draft
Hdat pdf-draftHdat pdf-draft
Hdat pdf-draftshassant2
 
Chromatographic Analysis of Pharmaceuticals Second Edition by John A. Adamovics
Chromatographic Analysis of Pharmaceuticals Second Edition by John A. AdamovicsChromatographic Analysis of Pharmaceuticals Second Edition by John A. Adamovics
Chromatographic Analysis of Pharmaceuticals Second Edition by John A. AdamovicsRohit K.
 

Similar a Mass Spectrometry reviews 2008 v27 p20 (20)

Crimson Publishers -A Sensor Multiplatform for Non Invasive Diagnosis of Pros...
Crimson Publishers -A Sensor Multiplatform for Non Invasive Diagnosis of Pros...Crimson Publishers -A Sensor Multiplatform for Non Invasive Diagnosis of Pros...
Crimson Publishers -A Sensor Multiplatform for Non Invasive Diagnosis of Pros...
 
Dorobantu_A_Luminex-bead-based assay - Feedback.pdf
Dorobantu_A_Luminex-bead-based assay - Feedback.pdfDorobantu_A_Luminex-bead-based assay - Feedback.pdf
Dorobantu_A_Luminex-bead-based assay - Feedback.pdf
 
Limitations and Advantages in Assessing Adenovirus Homogeneity by Laser Light...
Limitations and Advantages in Assessing Adenovirus Homogeneity by Laser Light...Limitations and Advantages in Assessing Adenovirus Homogeneity by Laser Light...
Limitations and Advantages in Assessing Adenovirus Homogeneity by Laser Light...
 
Leibniz: A Digital Scientific Notation
Leibniz: A Digital Scientific NotationLeibniz: A Digital Scientific Notation
Leibniz: A Digital Scientific Notation
 
Journal Combinatorial Chemistry 2006 v8 p820
Journal Combinatorial Chemistry 2006 v8 p820Journal Combinatorial Chemistry 2006 v8 p820
Journal Combinatorial Chemistry 2006 v8 p820
 
Mass Spectrometry Basic By Inam
Mass Spectrometry Basic By InamMass Spectrometry Basic By Inam
Mass Spectrometry Basic By Inam
 
Chenomx
ChenomxChenomx
Chenomx
 
ANALYTICAL TECHNIQUES (CHROMATOGRAPHY, SPECTROSCOPY, ELECTROPHOROSIS) IN PHAR...
ANALYTICAL TECHNIQUES (CHROMATOGRAPHY, SPECTROSCOPY, ELECTROPHOROSIS) IN PHAR...ANALYTICAL TECHNIQUES (CHROMATOGRAPHY, SPECTROSCOPY, ELECTROPHOROSIS) IN PHAR...
ANALYTICAL TECHNIQUES (CHROMATOGRAPHY, SPECTROSCOPY, ELECTROPHOROSIS) IN PHAR...
 
Metabolomics
MetabolomicsMetabolomics
Metabolomics
 
Analytical Considerations When Monitoring Pain Medications by LC-MS/MS
Analytical Considerations When Monitoring Pain Medications by LC-MS/MSAnalytical Considerations When Monitoring Pain Medications by LC-MS/MS
Analytical Considerations When Monitoring Pain Medications by LC-MS/MS
 
SURE poster
SURE posterSURE poster
SURE poster
 
The Future of Computational Models for Predicting Human Toxicities
The Future of Computational Models for Predicting Human ToxicitiesThe Future of Computational Models for Predicting Human Toxicities
The Future of Computational Models for Predicting Human Toxicities
 
Quantitative mass spec imaging
Quantitative mass spec imagingQuantitative mass spec imaging
Quantitative mass spec imaging
 
Metabolomics
MetabolomicsMetabolomics
Metabolomics
 
OGurel THz-Bio v4
OGurel THz-Bio v4OGurel THz-Bio v4
OGurel THz-Bio v4
 
HTS by mukesh
HTS by mukeshHTS by mukesh
HTS by mukesh
 
Mass spectrometry it's use applications methods
Mass spectrometry it's use applications methodsMass spectrometry it's use applications methods
Mass spectrometry it's use applications methods
 
A review of use of enantiomers in homeopathy
A review of use of enantiomers in homeopathyA review of use of enantiomers in homeopathy
A review of use of enantiomers in homeopathy
 
Hdat pdf-draft
Hdat pdf-draftHdat pdf-draft
Hdat pdf-draft
 
Chromatographic Analysis of Pharmaceuticals Second Edition by John A. Adamovics
Chromatographic Analysis of Pharmaceuticals Second Edition by John A. AdamovicsChromatographic Analysis of Pharmaceuticals Second Edition by John A. Adamovics
Chromatographic Analysis of Pharmaceuticals Second Edition by John A. Adamovics
 

Último

Natural Polymer Based Nanomaterials
Natural Polymer Based NanomaterialsNatural Polymer Based Nanomaterials
Natural Polymer Based NanomaterialsAArockiyaNisha
 
Recombinant DNA technology (Immunological screening)
Recombinant DNA technology (Immunological screening)Recombinant DNA technology (Immunological screening)
Recombinant DNA technology (Immunological screening)PraveenaKalaiselvan1
 
Cultivation of KODO MILLET . made by Ghanshyam pptx
Cultivation of KODO MILLET . made by Ghanshyam pptxCultivation of KODO MILLET . made by Ghanshyam pptx
Cultivation of KODO MILLET . made by Ghanshyam pptxpradhanghanshyam7136
 
Nightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43b
Nightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43bNightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43b
Nightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43bSérgio Sacani
 
Unlocking the Potential: Deep dive into ocean of Ceramic Magnets.pptx
Unlocking  the Potential: Deep dive into ocean of Ceramic Magnets.pptxUnlocking  the Potential: Deep dive into ocean of Ceramic Magnets.pptx
Unlocking the Potential: Deep dive into ocean of Ceramic Magnets.pptxanandsmhk
 
A relative description on Sonoporation.pdf
A relative description on Sonoporation.pdfA relative description on Sonoporation.pdf
A relative description on Sonoporation.pdfnehabiju2046
 
Artificial Intelligence In Microbiology by Dr. Prince C P
Artificial Intelligence In Microbiology by Dr. Prince C PArtificial Intelligence In Microbiology by Dr. Prince C P
Artificial Intelligence In Microbiology by Dr. Prince C PPRINCE C P
 
Spermiogenesis or Spermateleosis or metamorphosis of spermatid
Spermiogenesis or Spermateleosis or metamorphosis of spermatidSpermiogenesis or Spermateleosis or metamorphosis of spermatid
Spermiogenesis or Spermateleosis or metamorphosis of spermatidSarthak Sekhar Mondal
 
CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service 🪡
CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service  🪡CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service  🪡
CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service 🪡anilsa9823
 
GFP in rDNA Technology (Biotechnology).pptx
GFP in rDNA Technology (Biotechnology).pptxGFP in rDNA Technology (Biotechnology).pptx
GFP in rDNA Technology (Biotechnology).pptxAleenaTreesaSaji
 
Grafana in space: Monitoring Japan's SLIM moon lander in real time
Grafana in space: Monitoring Japan's SLIM moon lander  in real timeGrafana in space: Monitoring Japan's SLIM moon lander  in real time
Grafana in space: Monitoring Japan's SLIM moon lander in real timeSatoshi NAKAHIRA
 
Recombination DNA Technology (Nucleic Acid Hybridization )
Recombination DNA Technology (Nucleic Acid Hybridization )Recombination DNA Technology (Nucleic Acid Hybridization )
Recombination DNA Technology (Nucleic Acid Hybridization )aarthirajkumar25
 
Is RISC-V ready for HPC workload? Maybe?
Is RISC-V ready for HPC workload? Maybe?Is RISC-V ready for HPC workload? Maybe?
Is RISC-V ready for HPC workload? Maybe?Patrick Diehl
 
Formation of low mass protostars and their circumstellar disks
Formation of low mass protostars and their circumstellar disksFormation of low mass protostars and their circumstellar disks
Formation of low mass protostars and their circumstellar disksSérgio Sacani
 
SOLUBLE PATTERN RECOGNITION RECEPTORS.pptx
SOLUBLE PATTERN RECOGNITION RECEPTORS.pptxSOLUBLE PATTERN RECOGNITION RECEPTORS.pptx
SOLUBLE PATTERN RECOGNITION RECEPTORS.pptxkessiyaTpeter
 
Traditional Agroforestry System in India- Shifting Cultivation, Taungya, Home...
Traditional Agroforestry System in India- Shifting Cultivation, Taungya, Home...Traditional Agroforestry System in India- Shifting Cultivation, Taungya, Home...
Traditional Agroforestry System in India- Shifting Cultivation, Taungya, Home...jana861314
 
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...Sérgio Sacani
 
Nanoparticles synthesis and characterization​ ​
Nanoparticles synthesis and characterization​  ​Nanoparticles synthesis and characterization​  ​
Nanoparticles synthesis and characterization​ ​kaibalyasahoo82800
 
Botany 4th semester series (krishna).pdf
Botany 4th semester series (krishna).pdfBotany 4th semester series (krishna).pdf
Botany 4th semester series (krishna).pdfSumit Kumar yadav
 

Último (20)

Natural Polymer Based Nanomaterials
Natural Polymer Based NanomaterialsNatural Polymer Based Nanomaterials
Natural Polymer Based Nanomaterials
 
Recombinant DNA technology (Immunological screening)
Recombinant DNA technology (Immunological screening)Recombinant DNA technology (Immunological screening)
Recombinant DNA technology (Immunological screening)
 
Cultivation of KODO MILLET . made by Ghanshyam pptx
Cultivation of KODO MILLET . made by Ghanshyam pptxCultivation of KODO MILLET . made by Ghanshyam pptx
Cultivation of KODO MILLET . made by Ghanshyam pptx
 
Nightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43b
Nightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43bNightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43b
Nightside clouds and disequilibrium chemistry on the hot Jupiter WASP-43b
 
Unlocking the Potential: Deep dive into ocean of Ceramic Magnets.pptx
Unlocking  the Potential: Deep dive into ocean of Ceramic Magnets.pptxUnlocking  the Potential: Deep dive into ocean of Ceramic Magnets.pptx
Unlocking the Potential: Deep dive into ocean of Ceramic Magnets.pptx
 
A relative description on Sonoporation.pdf
A relative description on Sonoporation.pdfA relative description on Sonoporation.pdf
A relative description on Sonoporation.pdf
 
Artificial Intelligence In Microbiology by Dr. Prince C P
Artificial Intelligence In Microbiology by Dr. Prince C PArtificial Intelligence In Microbiology by Dr. Prince C P
Artificial Intelligence In Microbiology by Dr. Prince C P
 
Spermiogenesis or Spermateleosis or metamorphosis of spermatid
Spermiogenesis or Spermateleosis or metamorphosis of spermatidSpermiogenesis or Spermateleosis or metamorphosis of spermatid
Spermiogenesis or Spermateleosis or metamorphosis of spermatid
 
CELL -Structural and Functional unit of life.pdf
CELL -Structural and Functional unit of life.pdfCELL -Structural and Functional unit of life.pdf
CELL -Structural and Functional unit of life.pdf
 
CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service 🪡
CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service  🪡CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service  🪡
CALL ON ➥8923113531 🔝Call Girls Kesar Bagh Lucknow best Night Fun service 🪡
 
GFP in rDNA Technology (Biotechnology).pptx
GFP in rDNA Technology (Biotechnology).pptxGFP in rDNA Technology (Biotechnology).pptx
GFP in rDNA Technology (Biotechnology).pptx
 
Grafana in space: Monitoring Japan's SLIM moon lander in real time
Grafana in space: Monitoring Japan's SLIM moon lander  in real timeGrafana in space: Monitoring Japan's SLIM moon lander  in real time
Grafana in space: Monitoring Japan's SLIM moon lander in real time
 
Recombination DNA Technology (Nucleic Acid Hybridization )
Recombination DNA Technology (Nucleic Acid Hybridization )Recombination DNA Technology (Nucleic Acid Hybridization )
Recombination DNA Technology (Nucleic Acid Hybridization )
 
Is RISC-V ready for HPC workload? Maybe?
Is RISC-V ready for HPC workload? Maybe?Is RISC-V ready for HPC workload? Maybe?
Is RISC-V ready for HPC workload? Maybe?
 
Formation of low mass protostars and their circumstellar disks
Formation of low mass protostars and their circumstellar disksFormation of low mass protostars and their circumstellar disks
Formation of low mass protostars and their circumstellar disks
 
SOLUBLE PATTERN RECOGNITION RECEPTORS.pptx
SOLUBLE PATTERN RECOGNITION RECEPTORS.pptxSOLUBLE PATTERN RECOGNITION RECEPTORS.pptx
SOLUBLE PATTERN RECOGNITION RECEPTORS.pptx
 
Traditional Agroforestry System in India- Shifting Cultivation, Taungya, Home...
Traditional Agroforestry System in India- Shifting Cultivation, Taungya, Home...Traditional Agroforestry System in India- Shifting Cultivation, Taungya, Home...
Traditional Agroforestry System in India- Shifting Cultivation, Taungya, Home...
 
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...
Discovery of an Accretion Streamer and a Slow Wide-angle Outflow around FUOri...
 
Nanoparticles synthesis and characterization​ ​
Nanoparticles synthesis and characterization​  ​Nanoparticles synthesis and characterization​  ​
Nanoparticles synthesis and characterization​ ​
 
Botany 4th semester series (krishna).pdf
Botany 4th semester series (krishna).pdfBotany 4th semester series (krishna).pdf
Botany 4th semester series (krishna).pdf
 

Mass Spectrometry reviews 2008 v27 p20

  • 1. Guest Editor: Tim Baker MASS SPECTROMETRY ANALYSIS OF NEW CHEMICAL ENTITIES FOR PHARMACEUTICAL DISCOVERY Annette S. Fang,1 Xiusheng Miao,1 Peter W. Tidswell,2 Marc H. Towle,3 Wolfgang K. Goetzinger,4 and James N. Kyranos5* 1ArQule, Inc., Woburn, MA 2Pfizer Global Research and Development, St. Louis, MO 3Vertex Pharmaceuticals, Cambridge, MA 4Amgen, Cambridge, MA 5Wolfe Laboratories, 134 Coolidge Avenue, Watertown, MA 02472 Received 28 February 2007; received (revised) 20 August 2007; accepted 24 September 2007 Published online 21 November 2007 in Wiley InterScience (www.interscience.wiley.com) DOI 10.1002/mas.20153 In this Section, we review the applications of mass spectrometry for the analysis and purification of new chemical entities (NCEs) for pharmaceutical discovery. Since the speed of synthesis of NCEs has dramatically increased over the last few years, new high throughput analytical techniques have been developed to keep pace with the synthetic developments. In this Section, we review both novel, as well as modifications of commonly used mass spectrometry techniques that have helped increase the speed of the analytical process. Part of the review is devoted to the purification of NCEs, which has undergone significant development in recent years, and the close integral association between characterization and purification to drive high throughput operations. At the end of the Section, we review potential future directions based on promising and exciting new developments. # 2007 Wiley Periodicals, Inc., Mass Spec Rev 27:20–34, 2008 Keywords: mass spectrometry; characterization; purification; new chemical entities I. BACKGROUND In recent years, the development of high throughput synthesis techniques has significantly changed the drug discovery process. The successful implementation of automation technology that allows medicinal chemists to generate 10–1,000 times more compounds for screening has augmented the traditional synthesis of single compounds. Today’s approach for early discovery hit identification is geared towards generating hundreds or even thousands of new chemical entities (NCEs) every week. By leveraging automation technology, larger numbers of compounds can be generated with minimal additional effort. However, in order to effectively utilize the subsequent screening information, these compounds must be characterized to assess purity and quantity. Moreover, screening larger collections of closely related analogs together, yields more data per cycle and leads to more complete structure activity relationships. By conducting the initial cycles of design, synthesis, analysis and screening in a high throughput manner, the typical medicinal chemistry process is converted to a more efficient and cost effective discovery process (Kassel, 2001). Traditionally, every compound synthesized would be analyzed by techniques such as nuclear magnetic resonance (NMR), thin layer chromatography (TLC), and elemental analysis. Although these techniques provide the medicinal chemist with the necessary characterization information, they are not readily amenable to unattended high throughput automation and easily automated data interpretation. The ability to acquire and interpret data in an automated fashion is an important consideration to effectively handle the thousands of NCEs being synthesized. While NMR is clearly the technique of choice for most traditional medicinal chemists, the need for spectral interpretation by an experienced professional limit the overall throughput of the technique in a high throughput setting. In addition, higher capital costs, limited automation capabilities, large sample requirements and the use of deuterated solvents impose further limitations on the use ofNMRfor high throughput analysis (Haner, Llanos, & Mueller, 2000; Keifer et al., 2000; Lindon, Nicholson,&Wilson, 2000; Louden et al., 2000;Wilson, Lindon, & Nicholson, 2000). II. MASS SPECTROMETRY—CENTRAL ANALYTICAL TECHNIQUE Simultaneous to the development of high throughput synthesis, mass spectrometry (MS) has emerged as the key analytical technique to characterize NCEs (Carrell et al., 1995; Enjalbal, Martinez, & Aubagnac, 2000; Hauser-Fang & Vouros, 2000; Su¨ssmuth & Jung, 1999; Triolo et al., 2001a,b). The inherent high sensitivity of MS leads to small sample requirements. Moreover, the relative ease of unattended automation and automated data interpretation contribute toMSbeing the primary characterization technique for high throughput analysis. Below ———— *Correspondence to: James N. Kyranos, Wolfe Laboratories, 134 Coolidge Avenue, Watertown, MA 02472. E-mail: james.kyranos@wolfelabs.com Mass Spectrometry Reviews, 2008, 27, 20–34 # 2007 by Wiley Periodicals, Inc.
  • 2. we will discuss some of the most common mass spectrometry-based techniques along with advantages and disadvantages. Depending on the focus of the investigation, a variety of analytical data may be desired, such as overall purity, number and type of contaminants or the identification of other com-ponents in mixtures (Kassel, 2001). In such cases, mass spectrometry is often used in combination with other techniques such as ultraviolet (UV) or evaporative light scattering detectors (ELSD) to collect a range of complementary data for higher confidence results (Yan et al., 2000). Adapting to the new challenges presented by modern drug discovery, all analytical instrumentation and especially mass spectrometry have evolved. The most notable changes in MS have been associated with the continued refinement of ionization techniques and the successful coupling of various separation methods with mass spectrometry. Most analyses involving small organic compounds to large biomolecules have benefited from advances in ionization techniques such as electrospray ionization (ESI), atmospheric pressure chemical ionization (APCI) and matrix-assisted laser desorption/ionization (MALDI). The most pronounced advantage of MS regardless of the ionization technique noted above is that resulting spectra are simpler to interpret with very few fragments observed compared to electron impact (Hauser-Fang & Vouros, 2000). In addition to generating simpler spectra, atmospheric pressure ionization techniques allow much easier coupling of high performance liquid chromatography (HPLC), capillary electrophoresis (CE) or supercritical fluid chromatography (SFC) with a mass spectrometer. Earlier publications reported successful coupling HPLC instruments with MALDI and fast atom bombardment (FAB) ionization sources, however, ESI and APCI have become the dominant ionization techniques in the field of on-line HPLC–MS. The role of MS and HPLC–MS for the characterization of combinatorial libraries has been described by several authors, and rapid HPLC–MS for the analysis of small molecules has been reviewed in the past byWehr (2003), Tiller et al. (2003), and Hsieh, Brisson, and Wang (2003). Typically, an initial MS characterization run provides a quick check for synthetic success and an assessment of compound purity. The resulting information may then be used to set up a variety of parameters for subsequent purification or to further investigate potential synthesis failures. Regardless of the motivation or approach, given the increased pressure to generate more high quality compounds in a shorter time frame, high throughput MS methods for both characterization and purification have become commonplace. III. IONIZATION TECHNIQUES FOR A HIGH THROUGHPUT ENVIRONMENT Since mass spectrometric detection is very specific in providing compound structural information, it is an irreplaceable tool in drug discovery (Kleintop, Zhang, & Ray, 2004). In addition, fast and accurate analytical methods are essential to keep pace with compound libraries produced with high throughput synthesis techniques. To facilitate these needs there have been many improvements and innovations of ionization sources. MASS SPECTROMETRY ANALYSIS & A. MUX Source One of the most innovative approaches to increase the effective utilization of the mass spectrometer, and increase the overall analysis speed, is the development of the multi-channel ESI (MUX) source (Biasi et al., 1999;Wang et al., 1999; Fang et al., 2002). The MUX interface consists of multiple electrospray probes connected to separate HPLC pumps and one sampling cone. Multi-channel electrospray sources have been designed with up to eight individual parallel channels ending in an electrospray tip at an angle to the sampling cone. A sampling rotor connects the spray of each channel to the sampling cone in rapid succession. Data acquisition occurs separately for each channel with as many data traces as channels used (usually four or eight). This approach leverages a single more expensive mass spectrometer coupled with multiple separation systems to generate an overall cost-effective solution (Sage, Little, & Giles, 2000; Burg, 2004). However, because of the fast acquisition times that are needed to effectively switch between channels, this type of setup is only possible with instruments having high scan speeds. Orthogonal acceleration time of flight (TOF) mass spectrometer is the instrument of choice for this application. As the speed of analysis is increased, the per-sample capital expenses are correspondingly reduced. In order to reduce the overall per sample analysis time, this type of system has been applied for both pre-purification and post-purification analysis of high throughput synthesis products (Yan et al., 2004). The general approach, with appropriate modification made for preparative HPLC, has also been used for high throughput purification of diastereomers (Irving et al., 2004). B. Combination Sources It is estimated that about 80% of small molecular weight compounds that are of the same structural class readily ionize using a similar electrospray ionization method. For samples that do not ionize well using a typical acid modified mobile phase and ESI, one can investigate increasing the pH of the mobile phase or change the ionization technique. Gallagher et al. (2003) have developed a new combined ESI–APCI source (ESCi) for use with on-line HPLC applications. This combined source can generate clearly differentiated and reproducible ESI and APCI spectra. Furthermore, positive and negative spectra can readily be obtained by switching polarity. The qualitative performance of the combined source has been compared to existing ESI and APCI interfaces. The ESI mode of the new source has been found to be equivalent to conventional ESI and the APCI mode produces less thermal fragmentation. This new source operates effectively at flow rates from 50 to 1,000 mL/min and can be used with a variety of separation techniques. By eliminating the need to repeat chromatographic separations in order to generate data using different ionization modes or polarity, the overall analysis time is reduced and throughput is increased. Similarly, in order to gain an advantage in the ionization of low polarity compounds, an APCI/APPI (atmospheric pressure chemical ionization/atmospheric pressure photoionization) source has been developed. The advantage of this approach, for example, is that this source can easily be coupled with normal Mass Spectrometry Reviews DOI 10.1002/mas 21
  • 3. phase HPLC systems for the analysis of very nonpolar compounds such as steroids (Robb, Covey, & Bruins, 2000). C. MALDI MALDI–TOF has become a widely used and powerful tool for the analysis of large peptides, biomolecules, and synthetic polymers. However, the analysis of small molecules by MALDI has remained a challenge. This is primarily due to the technique’s relatively poor ionization efficiency and the presence of a variety of abundant matrix-related ions in the low mass range. In addition, proper sample preparation requires significantly more operator experience and thus is not amenable to open access walk-up systems that are available to the medicinal chemists. Regardless of the potential limitations, MALDI has many advantages including low detection limits that are in the femtomole range. This level of sensitivity is ideal for the analysis of resin bound compounds obtained from individual beads. MALDI characterization of single beads has been successfully used for characterization of split-and-mix combinatorial libra-ries. (Egner, Cardno, & Bradley, 1995; Egner, Langley, & Bradley, 1995). By using appropriate photocleavable linkers during synthesis, products can be analyzed directly off the solid support (Fitzgerald et al., 1996). The MALDI laser irradiation provides enough energy to cleave the compound off the bead and ionize it at the same time. Although low molecular weight interference produced by conventional matrices, has limited the utility of MALDI for NCE characterization (Cohen & Gusev, 2002; Krutchinsky & Chait, 2002), recent introductions of new matrices are beginning to overcome this issue (Iijima, 1991; Iijima & Ichihashi, 1993; Dale, Knochenmuss,&Zenobi, 1996;Ayorinde et al., 1999;Wei, Buriak, & Siuzdak, 1999; Han & Sunner, 2000; Kinumi et al., 2000; Cuiffiet al., 2001; Shen et al., 2001; Zhang et al., 2001; Guo et al., 2002; Cai et al., 2003; Fu & Sun, 2003; Lewis et al., 2003; Peng et al., 2003; Reyzer et al., 2003; Xu et al., 2003; Pan et al., 2004; Go et al., 2005; Gobey et al., 2005; Pan et al., 2005; Sunner, Dratz, & Chen, 1995). Stolzberg and Patel (2004), for example, were able to produce clean negative ion mass spectra of molecules in the 100–300 Da range using a light-absorbing electrically conductive polymer matrix. Peterson et al. (2004) used porous polymer monoliths for MALDI analysis and were able to transfer sufficient energy to the analyte to induce desorption and ionization. The performance of the monolith hydrophobic porous surfaces was demonstrated with several small molecules, including drugs, explosives and acid labile compounds. Separation combined with MALDI–MS for small molecule analysis has been reported by several authors using ultra-thin layer chromatography (TLC–MALDI) (Busch, 1992; Somsen, Morden,&Wilson, 1995;Weins& Hauck, 1996; Chen, Shiea,& Sunner, 1998; Wilson, 1999; Gusev, 2000; Mehl & Hercules, 2000; Hauck et al., 2001; Ka´lasz & Ba´thori, 2001; Hauck & Schulz, 2002;Wu&Chen, 2002; Crecelius, Clench,&Richards, 2003; Salo et al., 2003; Santos et al., 2004). In this approach, analytes were separated on TLC plates, which were then coated with matrix and subsequently introduced into theMALDIsource. Salo et al. (2003) used an atmospheric pressure MALDI (AP-MALDI) source in order to reduce possible contamination of the mass spectrometer from introducing chromatographic material into a vacuum. All analyte spots were successfully ionized directly from the TLC plate. Gobey et al. (2005) have shown that the use of a high repetition rate laser can increase sample throughput while filtering out matrix interferences through the use of tandem mass spectrometry. The authors especially emphasize that this type of MALDI analysis is the ultimate high-throughput technique with an analysis time limited only by the speed of the plate readers of the MALDI instrument rather than the solvent handling capacity of an HPLC–MS system. Limitations include ion suppression effects from complex matrices and the need for additional SPE cleanup procedures for complex and impure samples. In a comparison of MALDI and ESI using 14 compounds that failed ESI analysis, only four of them were successfully analyzed by MALDI. These data suggest that MALDI and ESI are generally not orthogonal techniques. In a recent publication by Corra et al. (2006), ESI and MALDI were compared with regards to several operating considerations, including low-molecular weight sensitivities, analysis speed, throughput and instrument robustness. They found that although MALDI has a faster acquisition speed, detection limits are higher compared to HPLC/ESI–MS. However, by developing methods to concen-trate samples on the target, detection limits can be significantly improved. Lee, Chen, and Gebler (2004) have shown that MALDI–MS can be used both as a qualitative or quantitative technique. MALDI can certainly be used in an automated high-throughput mode with individual compounds loaded onto the target. However, the typical spot-to-spot sample reproducibility and matrix interferences are still limiting factors, especially for small molecule analysis. Sleno (2005) have reviewed the fundamental and technical aspects of MALDI and its limitations in greater detail. If the limitations of the technique can be overcome, MALDI analysis might one day provide extremely fast analysis of NCEs while consuming only small sample quantities. D. Nano and Chip Based Techniques In a unique approach Pan et al. (2004, 2005) used oxidized and nonoxidized carbon nanotubes as a matrix for small molecule MALDI analysis (Iijima, 1991; Iijima & Ichihashi, 1993; Sunner et al., 1995; Dale, Knochenmuss,&Zenobi, 1996; Han&Sunner, 2000; Cai et al., 2003; Fu&Sun, 2003; Peng et al., 2003;Xu et al., 2003). Compared to conventional matrices, they observed dramatically reduced matrix interferences for both types of nanotubes. Oxidized carbon nanotubes enhanced signal intensity compared to nonoxidized ones. The advantages of oxidized carbon nanotubes seem to be associated with greater water solubility that leads to a more homogeneous matrix mixture. By using silicon nanowires (Go et al., 2005), the sensitivity of the technique was increased down to the attomole level. Moreover, the 40 nm silicon nanowires were used to conduct a chromatographic separation by taking advantage of their high surface area and fluid wicking capabilities. The nanowires provide a unique platform for both separation and mass spectrometric analysis. & FANG ET AL. 22 Mass Spectrometry Reviews DOI 10.1002/mas
  • 4. Zhang et al. (2001) described quantitative analysis of small molecules using chip-based nano ESI–MS/MS. This system is reported to have advantages in limiting carryover, sample consumption, cycle time and the ability to be fully automated. Chip-based nano-electrospray that is integrated into a micro purification/collection system has also been reported and will be discussed further in the purification section. IV. SEPARATIONS COUPLED WITH MASS SPECTROMETRY Speed is an important consideration in modern drug discovery. The more time that is reduced from the discovery and develop-ment cycle, the more patent protection the company can enjoy once the drug is on the market. Extending the patent protection by 6 months to a year translates into hundreds of millions of dollars in total revenue. It is these types of incentives that drive the industry to greater speed and efficiency. For high throughput analysis, the need for speed usually means that large numbers of compounds have to be analyzed in a matter of days and the results have to be interpreted quickly thereafter. For practical purposes, high throughput analysis is usually further divided into high throughput characterization and purification. In characterization, the focus lies in determining the contents and/or assessing the purity of a large number of samples. Characterization analysis is used as an initial check for synthesis completion, quality control purposes or to collect important data about samples that need to be purified later. For purification, the data obtained in characterization is typically used to optimize the subsequent preparative analyses. Given the robustness of the single quadrupole systems and the lower cost relative to the high end mass spectrometers, low-resolution HPLC–ESI–MS instruments have become the industry standard for high throughputNCEcharacterization. There have been some examples of high throughput high resolution work but usually this process is significantly more complex and in most cases unnecessary (Walk et al., 1999). A. FIA–MS Flow injection analysis mass spectrometry (FIA-MS) is the simplest form of rapid sample introduction into the mass spectrometer. This approach was one of the earliest examples of characterization and has been widely used in the analysis of high throughput synthesis products. If separation is not needed, FIA can be used to confirm synthesis by the presence of the mass-to- charge ratio (m/z) of the expected compound ions. Since flow injection analysis is not limited by separation run times, it has real speed advantages. FIA–MS provides the highest throughput and greatest ease of automation and use for large numbers of samples (Yu & Balogh, 2000). A major drawback to FIA–MS is ion suppression due to co-eluting sample components. The ion suppression can be quite significant. In many cases there is significant chemical interference and the expected molecular ion of the desired compounds is not observed, even though the synthesis may have been successful. Moreover, without separa-tion of the synthesis mixture components, there is no reliable purity determination. MASS SPECTROMETRY ANALYSIS & One of the early issues arising in FIA–MS analysis was sample to sample carry-over. This issue can be a serious concern for any analysis, however, it is of particular issue when running large number of samples very quickly, where wash cycles may be abbreviated to gain cycle time speed. Earlier instrumentation was limited in its control of injector parameters and thus more prone to carryover when trying to maximize analysis speed. Richmond (2000) and Richmond and Go¨rlach (1999a) described minimiz-ing sample carryover in reaction monitoring using FIA–MS. Morand et al. (2001) reported an improved method with a throughput of 4 sec/sample by using a parallel sampling system with an eight-probe injector. Others have worked on improving speed or the analytical design (Felten et al., 2001) and also on theoretically analyzing inter-sample carryover (Richmond & Go¨rlach, 1999b; Richmond, 2000). The increasing numbers of samples for analysis required a further reduction in the sample measurement duty cycle while maintaining the carry-over below 1%. Lazar et al. (1999) introduced high-speed autosamplers with duty cycles that could be reduced from 168 to 44 sec. Further optimization of syringe and loop wash steps could reduce the median inter-sample carry-over to 0.01%. Wang et al. (1998) shortened the sample injection duty cycle by using a multiprobe autosampler for flow-injection analysis of eight samples within 1 min. Using this type of autosampler, it has been shown that more than eight flow-injection peaks can be generated in 1 min, which translates into analyzing one whole 96-microtiter well plate in less than 12 min (Wang et al., 1998). Shah et al. (2000) described a quantitation method with FIA–MS and chemiluminescence nitrogen detection (CLND) for simultaneous proof of structure and estimation of purity, respectively. Characterization of one compound was completed every 60 sec, allowing for more than 1,000 compounds analyzed in a single day. However, when large amounts of impurities were present or the compound of interest was present at a very low concentration, purity estimation was less reliable. In these types of cases, HPLC–CLND and HPLC–MS may be required. Although, as described above, there have been many publications focusing on high-throughput injections and the reduction of carryover in FIA–MS, most commercial systems have been optimized for approximately one injection per minute. This injection speed is adequate for typical HPLC analysis but not high throughput FIA–MS operations. The approaches described above that were used for multiple injections per minute, require extensive modifications of injector hardware and software. B. HPLC–MS Flow–injection analysis provides the shortest run time for a high throughput system but does not yield confident purity determi-nation. In order to assure the highest possible data quality with more complex samples, chromatographic separation is neces-sary. There is significant opportunity to develop the appropriate analytical methods by coupling HPLC to mass spectrometry, however, usually at the expense of longer analysis times. In an effort to decrease HPLC–MS run times, rapid mobile phase gradients and/or modifiers, as well as packing materials and column size are among the more flexible components that have Mass Spectrometry Reviews DOI 10.1002/mas 23
  • 5. been adjusted to fit individual analysis. All these parameters have been used to develop effective high quality and rapid character-ization and purification methods. Of course, chromatographic analysis times can also be dramatically shortened by using different techniques such as capillary electrophoresis (CE), capillary electrochromatography (CEC) or supercritical fluid chromatography (SFC). Some of these techniques will be reviewed further in subsequent sections. In general, modifiers are compounds that alter the physicochemical properties of the mobile phase. Modifiers have been used quite extensively to optimize specific chromatographic separations. Although a variety of modifiers have been developed and reported, only those that are volatile are compatible with the needs of the mass spectrometer. This requirement has limited the use of modifiers primarily to trifluoroacetic acid (TFA) and formic acid (FA) for acidic methods and ammonium buffers for basic separations. In addition to optimizing chromatographic resolution, mobile phase additives can be used to improve MS sensitivity in both ESI and APCI detection modes. Mallet, Lu, and Mazzeo (2004) evaluated ion suppression/enhancement in negative-ion mode by using several additives and ion-pairing agents at different concentrations for test compounds. The intensity of a novel indolocarbazol compound increased by a factor of four in negative-ion mode when the pH of the mobile phase was increased from 8 to 10 using 2mMammonium hydroxide (Wang et al., 2002). Similarly, the sensitivity of detection was increased up to tenfold for isorhamnetin with a decrease in pH of the mobile phase ranging from 4.5 to 2.3 in positive-ion mode (Rauha, Vuorela, & Kostiainen, 2001). The addition of alkali metal salts to enhance the intensity of molecular ion adducts by forming stable Liþ, Naþ, or Kþ complexes of organic compounds has been observed in FAB, MALDI, and ESI mass spectrometry of various compounds (Williams et al., 2003). The extreme form of mobile phase modification can be considered micelle formation used with electrophoresis, which affects the fundamental separation processes. There have been reports of various approaches, such as micellar electrokinetic chromatography in combination with mass spectrometry (Goetzinger & Cai, 2005). Chromatographic resolution and mass spectrometric sensi-tivity are important considerations for all analytical methods. However, the real speed limitation for any high throughput analysis using HPLC–MS is the chromatographic separation time. Analysis speed has been enhanced by reducing the column size from typical column dimensions of 4.6150 mm to 4.630 mm. These shorter columns can be used with higher flow rates without exceeding the operating pressures limits. HPLC instrumentation has also been adapted to enable high-speed gradients. Presently, gradients of 5–95% acetonitrile/ aqueous buffers within 1–5 min with fast column regeneration are quite common. These developments have increased the throughput from 24 to 48 samples per day using a 30–60 min analysis to more than 250 samples per day using a 5 min analysis. One to five minute run times have become common for many laboratories. For our operations, run times of 2.5 min are the standard with no apparent negative effects on peak shape or resolution (Kyranos et al., 2001a). Such short run times make high throughput HPLC–MS analysis a practical and effective analytical solution to support high throughput synthesis. Goetzinger and Kyranos (1998) investigated packing materials, gradient methods and sample solvents to allow FIGURE 1. QC of combinatorial library sample with rapid HPLC. a: 1504.6 mm, 5 mm Zorbax SB-C8, 1.5 mL/min, 15–95% acetonitrile in 16 min; (b) 504.6 mm, 3.5 mm Zorbax SB-C8, 2 mL/min, 15–95% acetonitrile in 4 min; (c) 304.6 mm, 3.5 mm Zorbax SB-C8, 4 mL/min, 15–95% acetonitrile in 0.6 min. FANG ET AL. 24 Mass Spectrometry Reviews DOI 10.1002/mas
  • 6. ultra-fast gradient HPLC–MS methods down to 1 min per sample. Similarly, Hsieh et al. (2001) used fast gradients within 1 min for quantitative screening of drug discovery compounds in monkey plasma. Using generic methods to avoid method development is another trend to enhance the throughput for the analysis of combinatorial libraries. Figure 1 is an example of rapid chromatography using fast gradients and high flow without compromising chromatographic resolution. Although the run time is significantly decreased from 20 to 1 min, the overall peak capacity for all the separations is still acceptable to determine sample purity. The significant developments in separation speed have shifted the efforts for further optimization and time savings toward the autosampler and injector components (Zweigenbaum et al., 1999). As previously discussed, the MUX interface has been used successfully with FIA–MS to increase the overall throughput. Davis and Griffith (2004) described a high-throughput parallel HPLC–MS–ELSD method for the analysis of combinatorial libraries with an eight-channel TOF system. This configuration could analyze approximately 300,000 compounds per year. Fang et al. (2003) investigated parallel high-throughput accurate mass measurement using a nine-channel multiplexed HPLC–UV– TOF–MS system. Accurate mass measurements were simulta-neously obtained from compounds eluting from eight parallel columns. Mass accuracy better than 10 ppm was achieved for 80% of the samples investigated. In other examples of higher throughput, Cremin and Zeng (2002) applied an eight-way fully automated parallel LC–MS– ELSD system to enhance the throughput for the analysis of a natural product library. This method has been successfully applied to analyze a library of 36,000 partially purified fractions derived from plants. The quest for additional separation speed and higher efficiency, which in turn will increase the overall analysis throughput, continues with the development of new stationary phase materials. Much of the early development in this area has been focused on decreasing the particle size of the packing material. However, the higher operating pressure associated with particle size reduction has limited this advancement. Monolithic columns are a single solid piece of stationary material with pores through the main skeleton. These structures can increase column performance by increasing the column permeability and efficiency (Tanaka et al., 2002). There have been a number of applications in related research areas using monolithic columns that exemplify the beneficial performance of these materials (Asperger et al., 2002; Borges et al., 2004). C. CE–MS Capillary electrophoresis (CE) is a powerful separation techni-que that has been used to separate ionic compounds (Foulon et al., 2004; Liu, Jo¨nsson, Jiang, 2005) or chiral mixtures (Millot, 2003; Erny Cifuentes, 2006; Souverain et al., 2006). The different mode of separation of CE results in a unique selectivity compared to reverse phase HPLC. Moreover, high efficiency associated with CE promise enhanced separation resolution, which will lead to even faster analyses. This technique can be used as an orthogonal complimentary approach to HPLCor as the primary analysis. However, the difficulties in interfacing CE to the mass spectrometer and the limited amount of material that can be loaded on the column have hindered wider use of CE–MS. Dunayevskiy et al. (1996) was one of the first to apply CE– MS to combinatorial chemistry. They successfully identified the 171 individual components of a combinatorial library mixture using CE–MS. Visky et al. (2005) discussed the application of CE–MS and its role in impurity profiling of pharmaceutical products. More recently, Smyth reviewed the broader applica-tions of CE–MS in drug analysis (Smyth, 2005). Generally, CE– MScan be a valuable technique for rapid compound separation. It is also useful for analyzing extremely small sample volumes with high separation efficiency and has been applied to the analysis of complex biological matrixes (Moini, 2002; Wan Thompson, 2005). Goetzinger and Cai (2005) described a newway of preparing a micellar buffer system for the separation of neutral compounds based on micellar electrokinetic chromatography (MEKC). The substitution of the traditionally used alkyl sulfates, such as SDS, with carboxylates together with the substitution of inorganic cations with organic bases provided a different separation system with interesting properties. It was shown that these novel buffer systems could be used as micellar separation systems with lower conductivity compared to SDS buffers and thus provided more flexibility for separation conditions. Although this article was conducted using UV detection, the buffers used are volatile and amenable to interfacing with MS. The inevitable coupling of this novel MEKC system with MS could be used to increase separation speed or alternatively to improve the detection limit by using larger diameter capillaries. D. SFC–MS Supercritical fluid chromatography (SFC) has been used for chiral separations (Mukherjee, 2007). Recently there has been a growing trend in the application of SFC–MS to the analysis of NCEs (Berger et al., 2000). SFC offers the potential benefit of high-speed separations that can be complementary to reverse phase HPLC–MS (Berger, 1997; Coe, Rathe, Lee, 2006). Due to the lower viscosity of supercritical fluids, SFC can be run at higher flow rates compared to HPLC. Bolan˜os, Ventura, and Greig (2003) demonstrated the advantage of SFC–TOF MS for ultrafast qualitative applications. High-speed MS detection allows the superior resolving power of SFC to be exploited in high-throughput analytical settings. Bolanos et al. (2004) extended their work and summarized the applications of SFC– MS in drug discovery at Pfizer, La Jolla, CA. They rely heavily on SFC–MS for analysis, as well as, purification of diverse compound sets. SFC for purification applications is especially appealing due to rapid evaporation of the carbon dioxide mobile phase (Toribio et al., 2006; Zhang et al., 2006). During separation, the carbon dioxide mobile phase is under high pressure and behaves like a fluid. However, once the pressure is reduced after the detector, the fluid rapidly expands into a gas that quickly evaporates leaving behind the analyte and any organic modifier such as methanol. The rapid evaporation of the solvent after purification signifi-cantly increases the overall throughput of preparative SFC compared to HPLC. MASS SPECTROMETRY ANALYSIS Mass Spectrometry Reviews DOI 10.1002/mas 25
  • 7. Although there are many theoretical and practical advan-tages to SFC, most laboratories have not embraced the technique for characterization nor purification. Much of the resistance is probably due to the perception that SFC is more complicated than HPLC. In addition, there are significantly fewer vendor options for instrumentation and application support. Finally, SFC more closely resembles the separation of normal phase chromatog-raphy and many scientists may assume that SFC will not be applicable to many of the polar and ionic compounds typically associated with pharmaceutical research and development. Pinkston et al. (2006) have recently shown that approximately 75% of a 2266 compound subset, representing their corporate collection, could be adequately assessed by SFC–MS. This compares to approximate 80% success of the same subset by HPLC–MS. These results further reinforce the value of SFC as an important technique for high throughput analysis in support of modern drug discovery and development. E. Multiple Hyphenated Detection Techniques One of the benefits of HPLC–MS is the ability to couple a variety of detectors to generate a range of qualitative, as well as quantitative information. It is quite common for one or more auxiliary detection methods to be combined with HPLC–MS to provide a better overall assessment of sample purity. A typical approach has been to couple UV and ELSD. Although the different detection methods often vary regarding detection capabilities, selectivity, sensitivity range, and linear response, collectively they provide complementary information. Due to variations of quantitative data from different detection techniques, Letot et al. (2005) investigated HPLC– CLND and HPLC–NMR for quantification of combinatorial library compounds. In general, he found that purity values obtained by NMR were more accurate than those obtained by CLND. However, the differences in purity are not always of the same order for the two methods. For low concentration quantification, NMR is less accurate than typically expected with higher sample concentrations. Yurek, Branch, and Kuo (2002) developed a new assay system for the simultaneous determination of identity, purity and concentration of sample components from combinatorial libra-ries produced by parallel synthesis. The system employed HPLC with photodiode array (PDA), ELSD, CLND, and TOF–MS detectors. The use of TOF–MS with CLND provides a synergistic combination, enabling target and side-product structures to be identified and quantified. This approach allows characterization, purity analysis and quantification in a single experiment from a synthesis solution containing microgram levels of materials. Recently, Peake et al. (2005) described a high-throughput screening method using ELSD, CLND and accurate mass HPLC–MS–MS to obtain both quantitative data and structural information. Although the complementary nature of the different detection methods is often a benefit, it can also be a challenge. In particular, sensitivity and detector response times may vary significantly among the various detectors. Therefore, it is very important to assure that all operating parameters are appropri-ately optimized. This is especially important for rapid separation methods, which produce chromatographic peaks with 1–2 sec widths. These systems are very susceptible to operating vari-ations and must be diligently optimized for peak performance. Another consideration when using multiple detectors to assess purity is developing protocols to deal with major purity inconsistencies between multiple detectors. Of course the definition of purity depends on the application and can vary for different experiments. In general, however, purity values of around 80–85% for both UV and ELSD have been found acceptable for many early discovery applications. It is important to note that for hyphenated detection, all purity values have to be above some predefined threshold. One failed value can indicate the presence of major impurities that have not been identified by the other methods. In our experience, we found that ELSD tends to overestimate the larger components in a mixture at the expense of the smaller ones. UVon the other hand, even monitoring low wavelengths, may exhibit more variability depending on structural differences of the mixture components. This is the advantage of the complementary detector approach, which generates higher confidence data (Letot et al., 2005). F. Open-Access HPLC–MS Although not a true high-throughput technique in the typical sense, the open-access or walk-up HPLC–MS system does provide quick single sample HPLC–MS analysis. This system has presently become the standard setup in many laboratories. The open-access system can be used at any time by the medicinal chemist in need of rapid confirmation of synthetic products or to monitor the extent of reactions. The quick single sample analysis can provide information about the completeness of a reaction, byproducts or unexpected reaction failure. These data add another layer of information to that obtained from NMR analysis or other traditional low throughput approaches. Once imple-mented, open-access systems accept individual samples in an ongoing queue and can be run without much support from the analytical department. The open-access approach was pioneered by Hayward, Snodgrass, and Thomson (1993) and optimized by Pullen and coworkers at Pfizer Central Research (Pullen et al., 1995a,b). Initially, an automated column-bypass thermospray MS system was used but later this was reconfigured into an ESI–MS system. The instrument is usually set up to email the results as completely processed data to the submitting chemists (Mallis et al., 2002). Because of its success, open-access software modules are currently offered by most instrument manufacturers to be used with single-quadrupole and TOF instruments. The use of an open-access facility can enhance sample throughput and allow the analytical chemists to spend more time on nonroutine work. Spreen and Schaffter (1996) have extensively reviewed applica-tions of open-access MS. It has been shown that open-access MS works extremely well for fast characterization of synthesis samples. Most small molecules that are of interest for pharmaceutical research ionize well using a standard set of instrument conditions and time consuming adjustments are typically not needed. Although the typical setup for a walk-up mass spectrometer includes a low resolution instrument, high resolution instrumentation has also been used effectively (Dykes et al., 2003). There have also been attempts to extend the walk-up approach to purification FANG ET AL. 26 Mass Spectrometry Reviews DOI 10.1002/mas
  • 8. systems (Blom et al., 2004). However to date, purification systems have not been adopted as readily as the characteriza-tion systems. V. MIXTURE ANALYSIS Most high throughput synthesis approaches being used generate a single discrete compound per reaction vessel. Solid phase mix-and-split combinatorial synthesis was one of the earliest synthesis techniques that generate mixtures of closely related analogs in each reaction vessel. The emphasis for this approach is to synthesize as many compounds as possible in a small number of reaction pools and then screen these mixtures to identify hits. Once a hit is identified in a mixture pool, that mixture pool is deconvoluted to identify the active component(s). Alternatively, the active mixture can be resynthesized in a series of smaller mixture pools and these pools screened for activity. By iteratively synthesizing and screening ever smaller mixture pools, the active compound(s) can ultimately be identified. This approach does not require complete characterization of the individual members of a mixture pool. The emphasis for analysis here is a quick assessment of how well the reaction has proceeded in general. The intent is often to eliminate complete synthesis failures rather than identifying and characterizing an individual compounds. Although HPLC–MS has been used in characterizing these types of complex mixtures, it is quite difficult and labor intensive. For such a general evaluation of a synthetic procedure, direct analysis of the mixture using mass spectrometry has been used quite effectively. By knowing the compounds expected in a synthesis pool and assuming that each compound is equally represented in the mixture, theoretical mass distributions can be generated. These theoretical mass spectra can then be compared to the actual spectra and conclusions about the success of the synthesis can be drawn. These types of analyses have been successfully conducted using both lowand high resolution instrumentation (Carrell et al., 1995; Dunayevskiy et al., 1995; Nawrocki et al., 1996). Fourier transform ion cyclotron resonance (FTICR) instru-mentation, also known simply as Fourier transform mass spectrometry (FTMS), are high field magnetic instruments that are usually installed in a laboratory to handle nonroutine applications. The high resolution capabilities of FTMS and its more complex operation make it somewhat of a specialty analytical tool. Although FTMS can be used as an HPLC detec-tor, as described earlier in this review, the high costs and the more complex operation associated with it have limited this use. The primary advantages of FTMS are its high sensitivity (sub fmol), high mass accuracy (sub ppm levels), high resolution for complex mixtures (600,000 FWHM) and sequencing capabilities (exact MS(n) capability). The high resolution capabilities of FTMS are particularly appealing for the characterization of mixture analysis since accurate mass measurements can be used to correctly identify potential isobaric compounds without the use of chromato-graphic separation. Furthermore, the ability to conduct sequential MS–MS and obtain accurate mass assignment for the fragments helps to accurately identify the molecular structure of specific compounds in a complex synthesis mixture. Although HPLC– MS–MS may be able to adequately separate and characterize MASS SPECTROMETRY ANALYSIS individual structures in a complex matrix, direct analysis of the synthesis mixture using high resolution accurate mass FTMS in some cases may be easier and faster. On the other hand, conducting sequential MS–MS for each component of a large complex mixture is quite tedious, time consuming and not particularly amenable to high throughput environment. Several publications have focused on FTMS as a technique to character-ize (Winger Campana, 1996; Fang et al., 1998; Schmid et al., 2001) or screen (Poulsen, 2006) combinatorial library mixtures of discrete small molecules (Bristow Webb, 2003; Herniman et al., 2004, 2005; Zhang et al., 2005). VI. PURIFICATION OF NCES The number of new chemical entities undergoing purification before any biological assays are undertaken has increased over the last few years.(Ripka, Barker, Krakover, 2001). This is mostly due to the recognition that valuable resources are often wasted when impure compounds are submitted for screening only to end up generating false positives. This issue has led many companies to institute specific purity thresholds for compounds supporting medicinal chemistry projects. An industry standard of at least 80–85% purity by UVat 210–214 nm has become fairly typical (Letot et al., 2005). A. Preparative HPLC–MS Preparative HPLC using small efficient columns that allow for rapid separations have become the mainstay of the industry. Many laboratories use traditional UV detection to collect all fractions above a certain threshold. The collected fractions are then rapidly analyzed by FIA–MS to identify the component(s) of interest. Due to collecting multiple fractions from one sample, the overall throughput of this approach is severely constrained. The collection decks can accommodate only a limited number of fraction collection tubes and once those tubes are used, the operation stops. In an effort to alleviate this limitation, most of the current high throughput purification setups use some variation of HPLC–MS instrumentation as the method of fraction collection. The specificity of the mass spectrometer is used to trigger collection only when the expected mass of the product is seen in the sample. This approach significantly reduces the total number of fractions collected per sample and allows more samples to be purified per run.We have pushed the throughput to the maximum by allowing only one fraction to be collected for each sample. This type of setup is becoming more common and can purify approximately 3,000 compounds each month in a completely automated fashion using a single instrument. Setups for this type of analysis include simple single channel HPLC–MS systems as well as MUX systems, as described above. For purification of crude synthesis products, the most critical step in the process is the mass directed fraction collection. After separation by the HPLC column and detection by UV, the eluant is connected to a tee that splits the chromatographic flow. The majority of the HPLC eluant is directed to a switching valve connected to the fraction collector. This valve, which is controlled by the MS output signal, toggles between the collection tubes and the waste line. A smaller portion of the eluant is coupled to the MS system via a split flow interface. Mass Spectrometry Reviews DOI 10.1002/mas 27
  • 9. The mass spectrometer monitors the expected m/z trace of the component of interest. When the expected m/z rises above a predetermined threshold value, the collector valve is switched ‘‘on’’ to collect the fraction of interest. When the monitored m/z trace drops below a certain threshold, flow through the switching valve is directed to waste. In our approach, the waste line is connected to a second UV detector and the eluant is monitored prior to discharging it in the waste system. The overall efficiency of the collection process can be monitored for each sample in real time by comparing the trace of the UVimmediately after the HPLC column with the chromatographic trace from the waste UV (Goetzinger et al., 2004). The amount of sample being split and infused into the mass spectrometer must be small. Since mass spectrometry consumes the sample during detection, only the absolute minimum of the sample should be sacrificed. Moreover, the sensitivity of MS is quite high, therefore, the amount necessary for good signal-to-noise detection is relatively small. In fact, given the high sample concentration eluting off the preparative HPLC column and the minimal sample necessary for good MS detection, a 1:10,000 split ratio is fairly typical. To effectively and reproducibly accommodate this split, small lengths of capillary tubing ranging from 5 to 25 mm in diameter are typically used. The high split ratio necessary to limit the amount of material entering the mass spectrometer requires additional make up flow provided by a separate pump. It has been suggested that a major drawback of mass directed fraction collection is the lack of ruggedness. This apparent limitation is more of a perception issue because of the more complex nature of MS and the experience necessary to set up and use split flow systems. Our experience has been that the system will be fairly stable and perform well as long as performance is continually monitored and regular preventive maintenance is conducted. Our protocols include running daily standards, checking system performance and recovery every month and regularly replacing in-line filters to prevent clogging of the flow split capillary. It has been shown by us and others that using the setup described above, more than 200 compounds can be purified in unattended overnight runs (Isbell et al., 2002, Kyranos et al., 2001b). In addition to implementing diligent preventive main-tenance procedures, custom monitoring systems can be employed that trigger an alarm and/or action when a variety of potential failures are detected. Potential system failures include high back pressure, injector clogs, collector failures, no anti-cipated masses to trigger collection are found or data acquisition errors. By properly programming these system monitoring devises, responses to the aforementioned failures can be customized, ranging from system shutdown to selectively rebooting an unresponsive computer to allow for continued operation. This type of custom sensor can be highly effective for unattended overnight runs. It can prevent samples from being injected into a failing system or will reboot a nonresponsive acquisition system and get it back into operation without wasting much time during the purification run (Paulson, 2005). B. Connecting Characterization and Purification The focus of the initial characterization analysis for synthesis samples is on developing high performance methods that can be used as general universal methods for all samples. Speed with maximum resolution of all the components in the mixture across the entire run time are important considerations. Once the pre-purification analysis is completed, the emphasis shifts to optimizing the separation conditions exclusively for the peak of interest. It is irrelevant if several other peaks are overlapping or if the majority of the peaks are closely eluting either early or later in the chromatogram. As long as the product peak is well separated from other components and can be collected with ease, purification will be successful. Ideally, in a high throughput environment where compound diversity is the rule, each synthesis mixture needs to be treated differently. This challenge contributed to developing automation routines that use the information obtained from initial characterization data to adjust the separation gradients used for purification. For example, if the compound of interest was found co-eluting with impurities late in the gradient, then the purification gradient is automatically adjusted to start with a high percentage of organic solvent followed by a shallow gradient until the desired product is collected. Using this approach, co-eluting impurities are adequately separated from the desired peak more effectively and a cleaner fraction is collected. This process is illustrated with the example in Figure 2. Figure 2a represents the pre-purification chromatogram, identifying the peak of interest (shaded). Based on this separation, the gradient was modified in order to further separate the product from the side products during the preparative run (Figure 2b). In reality, not all characterization data will identify perfect gradient assignments for all samples. There can be challenges regarding continued co-elution or target peaks that are broadened by a shallow gradient. But compared to running a routine universal gradient of 5–95% acetonitrile for all compounds, this method of adjusting the gradients based on characterization data routinely outperforms the rigid traditional approach. Thus it provides an effective way to optimize purification conditions for the large majority of compounds without significant operator effort. C. Post-Purification Challenges Other recent advances have been in the combination of analytical and preparative scale instrumentation that yields an analytical-to-prep combination. This instrument has the ability to perform an initial characterization analysis and under preset guidelines, utilizes the processed data from the first run to automate a purification analysis. In some cases this has been extended to subsequent characterization of the collected fraction(s). Since many high-throughput environments now need to handle large amounts of liquids from collected fractions, some efforts have been made to simplify these tasks. One such effort is the design of a fraction pooling station as described by Koppitz, Brailsford, andWenz (2005). In their approach, product fractions are automatically identified from their corresponding mass spectra, even accounting for sodium adducts or dimers. The key elements of this approach are automated identification of the target compounds. This information is then used to optimize preparative gradients for purification of the target compounds followed by automated purity assessment of fractions with FANG ET AL. 28 Mass Spectrometry Reviews DOI 10.1002/mas
  • 10. MASS SPECTROMETRY ANALYSIS FIGURE 2. a: Pre-purification QC analysis using a gradient of 5–95% acetonitrile. The product peak (4) partially co-elutes with an impurity at 1.43 min. b: Purification run with conditions adjusted to a gradient of 30–95% acetonitrile based on pre-purification results. The product peak is completely resolved from the co-eluting impurity that was detected in pre-purification QC run. subsequent pooling of validated product fractions. Purity assessment of product fractions allows cherry-picking of pure fraction tubes on the basis of an algorithm that calculates MS purities and considers fragmentation. Using this type of system, one person can easily process 100–200 compounds per day. D. Microscale Separations and Collection Besides the potential for sample path blockages, the sample loss associated with the use of small diameter capillaries for flow splitting is not a major factor for mass directed fraction collection with semi-preparative HPLC flows. Flow splitting can indeed become a problem once the scale and the flow rate become significantly smaller than traditional semi-preparative or ana-lytical applications. This has been reported byWachs and Henion (2001) and is part of the driver for the development of an integrated nano scale fraction collection system that is being marketed commercially as an ultra-low-volume fraction collec-tor and analyzer. This type of system monitors mass traces using an automated chip-based nanospray and collects fractions that can subsequently be reanalyzed automatically. VII. CONCLUSION AND FUTURE DIRECTIONS In the early 1990s, there was an explosive surge of new technology development primarily focused on characterization of NCE’s in support of early pharmaceutical discovery. During this period, mass spectrometry emerged as the central analytical technique in the development of modern high throughput Mass Spectrometry Reviews DOI 10.1002/mas 29
  • 11. analysis. The MS technologies covered in this review have made a leap from primarily research purposes to becoming an integral part of modern medicinal chemistry. However, over the last 5 years there has been greater emphasis on fine tuning the existing characterization techniques and making them more robust and reliable rather than pioneering new technologies. The emphasis on future developments in MS characterization is anticipated to be continuing to streamline and further increase throughput. There is still opportunity for significant improvements. Data handling and analysis can be automated further. Instrument and method troubleshooting can be automated. Development of additional methods that can accommodate more complex samples and new MALDI matrices that are more amenable to low molecular weight compounds are also needed. In contrast toMScharacterization, there has been significant development of high throughput purification using mass directed fractionation. Areas of future development are anticipated in automatic gradient or column assignments specific to the need of each sample, as well as improved automated fraction handling. Successful chip-based MS analytical techniques for both, characterization and purification have been reported. This area of research is promising to be the most significant development in NCE characterization and purification. The small volumes that are typically used with chip based techniques have the potential for rapid and ultra-high throughput while maintaining opera-tional and consumable costs low. However, the rate of future development of these techniques and how fast they may be adopted in mainstream applications is yet to be determined. REFERENCES Asperger A, Efer J, Koal T, Engewald W. 2002. Trace determination of priority pesticides in water by means of high-speed on-line solid-phase extraction-liquid chromatography-tandem mass spectrometry using turbulent-flow chromatography columns for enrichment and a short monolithic column for fast liquid chromatographic separation. J Chromatogr A 960:109. Ayorinde FO, Hambright P, Porter TN, Keith QL Jr. 1999. Use of meso tetrakis(pentafluorophenyl)porphyrin as a matrix for low molecular weight alkylphenol ethoxylates in laser desorption/ionization time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 13:2474. Berger TA. 1997. Separation of polar solutes by packed column supercritical fluid chromatography. J Chromatogr 785:3–33. Berger TA, Fogleman K, Staats T, Bente P, Crocket I, FarrellW, Osonubi M. 2000. The development of a semi-preparatory scale supercritical-fluid chromatograph for high-through purification of ‘‘combi-chem’’ libra-ries. J Biochem Biophys Methods 43:87–111. Biasi VD, Haskins N, Organ A, Bateman R, Giles K, Jarvis S. 1999. High throughput liquid chromatography/mass spectrometric analyses using a novel multiplexed electrospray interface. Rapid Commun Mass Spectrom 13:1165–1168. Blom KF, Glass B, Sparks R, Combs AP. 2004. Preparative LC-MS purification: Improved compound-specific method optimization. J Comb Chem 6:874–883. Bolan˜os BJ, Ventura MC, Greig MJ. 2003. Preserving the chromatographic integrity of high-speed supercritical fluid chromatography separations using time-of-flight mass spectrometry. J Comb Chem 5:451–455. Bolanos B, Greig M, Ventura M, Farrell W, Aurigemma CM, Li H, Quenzer TL, TivelK, Bylund JMR, Tran P, Pham C, Phillipson D. 2004. SFC/MS in drug discovery at Pfizer, La Jolla. J Mass Spectrom 238:85–97. Borges V, Yang E, Dunn J, Henion J. 2004. High-throughput liquid chromatography-tandem mass spectrometry determination of bupro-pion and its metabolites in human, mouse and rat plasma using a monolithic column. J Chromatogr B 804:277–287. Bristow AWT, Webb KS. 2003. Intercomparison study on accurate mass measurement of small molecules in mass spectrometry. JAm Soc Mass Spectrom 14:1086–1098. Burg J. 2004. Parallel LC/MS detection for automatic purification. Nachrichten aus der Chemie 52:623–624. Busch KL. 1992. Coupling thin layer chromatography with mass spectrom-etry. J Planar Chromatogr 5:72–79. Cai YQ, Jiang GB, Liu JF, Zhou QX. 2003. Multiwalled carbon nanotubes as a solid-phase extraction adsorbent for the determination of bisphenol A, 4-n-nonylphenol, and 4-tert-octylphenol. Anal Chem 75:2517– 2521. Carrell T,Wintner EA, Sutherland AJ, Rebek J Jr, Dunayevskiy Y, Vouros P. 1995. New promise in combinatorial chemistry: Synthesis, character-ization, and screening of small-molecule libraries in solution. Chem Biol 3:171–183. Chen YC, Shiea J, Sunner J. 1998. Thin-layer chromatography mass spectrometry using activated carbon, surface-assisted laser desorption/ ionization. J Chromatogr A 826:77–86. Coe RA, Rathe JO, LeeJW. 2006. Supercritical fluid chromatography-tandem mass spectrometry for fast bioanalysis of R/S-warfarin in human plasma. J Pharm Biomed Anal 42:573–580. Cohen LH, Gusev AI. 2002. Small molecule analysis by MALDI mass spectrometry. Anal Bioanal Chem 373:571–586. Corra JJ, Kovarika P, Schneidera BB, Hendriksea J, Lobodaa A, Covey TR. 2006. Design considerations for high speed quantitative mass spectrometry with MALDI ionization. J Am Soc Mass Spectrom 17: 1129–1141. Crecelius A, Clench MR, Richards D. 2003. TLC-MALDI in pharmaceutical analysis. LC-GC Eur 16:225–229. Cremin PA, Zeng L. 2002. High-throughput analysis of natural product compound libraries by parallel LC-MS evaporative light scattering detection. Anal Chem 74:5492–5500. Cuiffi JD, Hayes DJ, Fonash SJ, Brown KN, Jones AD. 2001. Desorption-ionization mass spectrometry using deposited nanostructured silicon films. Anal Chem 73:1292. Dale MJ, Knochenmuss R, Zenobi R. 1996. Graphite/liquid mixed matrices for laser desorption/ionization mass spectrometry. Anal Chem 68: 3321–3329. Davis PW, Griffith MC. 2004. High throughput parallel LC/MS/ELSD of combinatorial libraries using eight-channel LCT system with MUX technology. In: Kyranos JN, editor. High throughput analysis for early drug discovery. San Diego: Elsevier. pp. 112–123. Dunayevskiy Y, Vouros P, Carrell T, Wintner E, Rebek J Jr. 1995. Characterization of the complexity of small molecule libraries by electrospray ionization mass spectrometry. Anal Chem 67:2906–2915. Dunayevskiy YM, Vouros P, Wintner EA, Shipps GW, Carell T, Rebek J Jr. 1996. Application of capillary electrophoresis-electrospray ionization mass spectrometry in the determination of molecular diversity. Proc Natl Acad Sci USA 93:6152–6157. Dykes S, Fancy SA, Perkins GL, Pullen FS. 2003. The automation of a commercial fourier transform mass spectrometer to provide a quick and robust method for determining exact mass for the synthetic chemist. Eur J Mass Spectrom 9:73–80. Egner BJ, Cardno M, BradleyM. 1995a. Linkers for combinatorial chemistry and reaction analysis using solid phase in situ mass spectrometry. J Chem Soc Chem Commun 21:2163–2164. Egner BJ, Langley GJ, Bradley M. 1995b. Solid phase chemistry: Direct monitoring by matrix-assisted laser desorption/ionization time of flight mass spectrometry. A tool for combinatorial chemistry. J Org Chem 60: 2652–2653. FANG ET AL. 30 Mass Spectrometry Reviews DOI 10.1002/mas
  • 12. Enjalbal C, Martinez J, Aubagnac JL. 2000. Mass spectrometry in combinatorial chemistry. Mass Spectrom Rev 19:139–161. Erny GL, Cifuentes A. 2006. Liquid separation techniques coupled with mass spectrometry for chiral analysis of pharmaceutical compounds and their metabolites in biological fluids. J Pharma Biomed Anal 40: 509– 515. Fang AS, Vouros P, Stacey CC, Kruppa GH, Laukien FH, Wintner EA, Carell T, Rebek J. 1998. Characterization of combinatorial libraries using electrospray ionization fourier transform ion cyclotron resonance mass spectrometry. Comb Chem High Throughput Screen 1:22–33. Fang L, Cournoyer J, Demee M, Zhao J, Tokushige D, Yan B. 2002. High-throughput liquid chromatography ultraviolet/mass spectrometric analysis of combinatorial libraries using an eight-channel multiplexed electrospray time-of-flight mass spectrometer. Rapid Commun Mass Spectrom 16:1440–1447. Fang L, Demee M, Cournoyer J, Sierra T, Young C, Yan B. 2003. Parallel high-throughput accurate mass measurement using a nine-channel multiplexed electrospray liquid chromatography ultraviolet time-of-flight mass spectrometry system. Rapid Commun Mass Spectrom 17: 1425–1432. Felten C, Foret F, Minarik M, Goetzinger W, Karger BL. 2001. Automated high-throughput infusion ESI-MS with direct coupling to a microtiter plate. Anal Chem 7:1449–1454. Fitzgerald MC, Harris K, Shevlin CG, Siuzdak G. 1996. Direct character-ization of solid phase resin-bound molecules by mass spectrometry. Bioorg Med Chem Lett 6:979–982. Foulon C, Danel C, Vaccher C, Yous S, Bonte J-P, Goossens J-F. 2004. Determination of ionization constants of N-imidazole derivatives, aromatase inhibitors, using capillary electrophoresis and influence of substitutes on pKa shifts. J Chromatogr A 131–136. Fu KF, Sun YP. 2003. Dispersion and solubilization of carbon nanotubes. J Nanosci Nanotechnol 3:351–364. Gallagher RT, Balogh MP, Davey P, Jackson MR, Sinclair I, Southern LJ. 2003. Combined electrospray ionization-atmospheric pressure chem-ical ionization source for use in high-throughput LC-MS applications. Anal Chem 75:973–977. Go EP, Apon JV, Luo G, Saghatelian A, Daniels RH, Sahi V, Dubrow R, Cravatt BF,Vertes A, Siuzdak G. 2005. Desorption/ionization on silicon nanowires. Anal Chem 77:1641–1646. Gobey J, Cole M, Janiszewski J, Covey T, Chau T, Kovarik P, Corr J. 2005. Characterization and performance of MALDI on a triple quadrupole mass spectrometer for analysis and quantification of small molecules. Anal Chem 77:5643–5654. GoetzingerW, Kyranos J. 1998. Fast gradient RP-HPLC for high-throughput quality control analysis of spatially addressable combinatorial libraries. Am Lab 30:27–37. Goetzinger WK, Cai H. 2005. Buffer system for the separation of neutral and charged small molecules using micellar electrokinetic chromatog-raphy with mass spectrometric detection. J Chromatogr A 1079:372– 381. GoetzingerW, Zhang X, Bi G, Towle M, Cherrak D, Kyranos JN. 2004. High throughput HPLC/MS purification in support of drug discovery. Int J Mass Spectrom 238:153–162. Guo Z, Zhang Q, Zou H, Guo B, Ni J. 2002. A method for the analysis of low-mass molecules by MALDI-TOF mass spectrometry. Anal Chem 74:1637–1641. Gusev AI. 2000. Interfacing matrix-assisted laser desoption/ionization mass spectrometry with column and planar separations. Fresenius J Anal Chem 266:691–700. Han M, Sunner J. 2000. An activated carbon substrate surface for laser desorption mass spectrometry. Am Soc Mass Spectrom 11:644–649. Haner RL, Llanos W, Mueller L. 2000. Small volume flow probe for automated direct-injection NMR analysis: Design and performance. J Magn Reson 143:69–78. MASS SPECTROMETRY ANALYSIS Hauck HE, Schulz M. 2002. Ultrathin-layer chromatography. J Chromatogr Sci 40:1–3. Hauck HE, Bund O, Fischer M, Schulz M. 2001. Ultra-thin layer chromatography (UTLC)-a new dimension in thin-layer chromatog-raphy. J Planar Chromatogr 14:234–236. Hauser-Fang A, Vouros P. 2000. The use of mass spectrometry. In: Swartz ME, editor. Analytical techniques in combinatorial chemistry. New York: Marcel Dekker. pp. 29–63. Hayward MJ, Snodgrass JT, Thomson ML. 1993. Flow injection Thermos-pray mass spectrometry for the automated analysis of potential agricultural chemicals. Rapid Commun Mass Spectrom 7:85–91. Herniman JM, Bristow TWT, O’Connor G, Jarvis J, Langley GJ. 2004. Improved precision and accuracy for high-performance liquid chroma-tography/ fourier transform ion cyclotron resonance mass spectrometric exact mass measurement of small molecules from the simultaneous and controlled introduction of internal calibrants via a second electrospray nebuliser. Rapid Commun Mass Spectrom 18:3035–3040. Herniman JM, Langley GJ, Bristow TWT, O’Connor G. 2005. The validation of exact mass measurements for small molecules using FT-ICRMS for improved confidence in the selection of elemental formulas. J Am Soc Mass Spectrom 16:1100–1108. Hsieh Y, Chintala M, Mei H, Agans J, Brisson J-M, Ng KI, KorfmacherWA. 2001. Quantitative screening and matrix effect studies of drug discovery compounds in monkey plasma using fast-gradient liquid chromatog-raphy/ tandem mass spectrometry. Rapid Commun Mass Spectrom 15:2481. Hsieh Y, Brisson J-M,Wang G. 2003. Fast HPLC-MS/MS analyses for small molecules. Am Pharm Rev 6:14–20. Iijima S. 1991. Helical microtubules of graphitic carbon. Nature 354:56– 5658. Iijima S, Ichihashi T. 1993. Single-shell carbon nanotubes of 1-nm diameter. Nature 363:603–605. Irving M, Krueger CA,Wade JV, Hodges JC, Leopold K, Collins N, Chan C, Shaqair S, Shornikov A, Yan B. 2004. High-throughput purification of combinatorial libraries II: Automated separation of single diastereom-ers from a 4-amido-pyrrolidone library containing intentional diaster-eomer pairs. J Comb Chem 6:478–486. Isbell J, Xu R, Cai Z, Kassel DB. 2002. Realities of high-throughput liquid chromatography/mass spectrometry purification of large combinatorial libraries: A report on overall sample throughput using parallel purification. J Comb Chem 4:600–611. Ka´lasz H, Ba´thori M. 2001. Pharmaceutical applications of TLC. LC-GC Eur 14:311–321. Kassel DB. 2001. Combinatorial chemistry and mass spectrometry in the 21st century drug discovery laboratory. Chem Rev 101:255–268. Keifer PA, Smallcombe SH,Williams EH, Salomon KE, Mendez G, Belletire JL, Moore CD. 2000. Direct-injection NMR (DI-NMR): A flow NMR technique for the analysis of combinatorial chemistry libraries. J Comb Chem 2:151–171. Kinumi T, Saisu T, Takayama M, Niwa H. 2000. Matrix-assisted laser desorption/ionization time-of-flight mass spectrometry using as inorganic particle matrix for small molecule analysis. J Mass Spectrom 35:417. Kleintop BL, Zhang H, Ray KL. 2004. Leveraging the use of modern mass spectrometry instrumentation in small molecule drug development.Am Pharm Rev 7:124–127. Koppitz M, Brailsford A,Wenz M. 2005. Maximizing automation in LC/MS high-throughput analysis and purification. J Comb Chem 7:714– 720. Krutchinsky AN, Chait BT. 2002. On the nature of chemical noise in MALDI mass spectra. J Am Soc Mass Spectrom 13:129. Kyranos JN, Cai H, Zhang B, Goetzinger WK. 2001a. High-throughput techniques for compound characterization and purification. Curr Opin Drug Discov Dev 4:719–728. Mass Spectrometry Reviews DOI 10.1002/mas 31
  • 13. Kyranos JN, Cai H, Wei D, Goetzinger WK. 2001b. High-throughput high-performance liquid chromatography/mass spectrometry for modern drug discovery. Curr Opin Biotech 12:105–111. Lazar IM, Ramsey RS, Sundberg S, Ramsey JM. 1999. Subattomole-sensitivity microchip nanoelectrospray source with time-of-flight mass spectrometry detection. Anal Chem 71:3627. Lee PJ, Chen W, Gebler JC. 2004. Qualitative and quantitative analysis of small amine molecules by MALDI-TOF mass spectrometry through charge derivatization. Anal Chem 76:4888–4893. Letot E, Koch G, Falchetto R, Bovermann G, Oberer L, Roth H-J. 2005. Quality control in combinatorial chemistry: Determinations of amounts and comparison of the ‘‘purity’’ of LC-MS-purified samples by NMR, LC-UVand CLND. J Comb Chem 7:364–371. Lewis WG, Shen ZX, Finn MG, Siuzdak G. 2003. Desorption/ionization on silicon (DIOS) mass spectrometry: Background and applications. Int J Mass Spectrom 226:107–116. Lindon JC, Nicholson JK, Wilson ID. 2000. Directly coupled HPLC-NMR and HPLC-NMR-MS in pharmaceutical research and development. J Chromatogr B 748:233–258. Liu J-F, Jo¨nsson JA° , Jiang G-B. 2005. Application of ionic liquids in analytical chemistry. Trends Anal Chem 24:20–27. Louden D, Handley A, Taylor S, Lenz E, Miller S,Wilson ID, Sage A. 2000. Flow injection spectroscopic analysis of model drugs using on-lineUV-diode array, FT-infrared and 1H-nuclear magnetic resonance spectro-scopy and time-of-flight mass spectrometry. Analyst 125:927–931. Mallet CR, Lu Z, Mazzeo JR. 2004. A study of ion suppression effects in electrospray ionization from mobile phase additives and solid-phase extracts. Rapid Commun Mass Spectrom 18:49. Mallis LM, Sarkahian AB, Kulishoff JMJr,Watts WL Jr. 2002. Open-access liquid chromatography/mass spectrometry in a drug discovery environ-ment. J Mass Spectrom 37:889–896. Mehl JT, Hercules DM. 2000. Direct TLC-MALDI coupling using a hybrid plate. Anal Chem 72:68–73. Millot MC. 2003. Separation enantiomers by liquid chromatography and capillary electrophoresis, using immobilized proteins as chiral selectors. J Chromatogr B 797:131–159. Moini M. 2002. Capillary electrophoresis mass spectrometry and its application to the analysis of biological mixtures. Anal Bioanal Chem 373:466–480. Morand KL, Burt TM, Regg BT, Chester TL. 2001. Techniques for increasing the throughput of flow injection mass spectrometry. Anal Chem 73:247–252. Mukherjee PS. 2007. Validation of direct assay of an aqueous formulation of drug compound AZY by chiral supercritical fluid chromatography (SFC). J Pharm Biomed Anal 43:464–470. Nawrocki JP, Wigger M, Watson CH, Hayes TW, Senko MW, Benner SA, Eyler JR. 1996. Analysis of combinatorial libraries using electrospray ionization fourier transform ion cyclotron resonance mass spectrom-etry. Rapid Commun Mass Spectrom 10:1860–1864. Pan C, Xu S, Zou H, Guo Z, Zhang Y, Guo B. 2004. Carbon nanotubes as adsorbent of solid-phase extraction and matrix for laser desorption/ ionization mass spectrometry. J Am Soc Mass Spectrom 16:263– 270. Pan C, Xu S, Hu L, Su X, Ou J, Zou H, Guo Z, Zhang Y, Guo B. 2005. Using oxidized carbon nanotubes as matrix for analysis of small molecules by MALDI-TOF MS. J Am Soc Mass Spectrom 16:883–892. Paulson J. 2005. A stand-alone monitoring system for reliable unattended operation of a commercial preparative LC/MS purification system. JALA 10:6–15. Peake DA, Duckworth DC, Perun TJ, Scott WL, Kulanthaivel P, Strege MA. 2005. Analytical and biological evaluation of high throughput screen actives using evaporative light scattering, chemiluminescent nitrogen detection, and accurate mass LC-MS-MS. Comb Chem High Throughput Screen 8:477–487. Peng XJ, Li YH, Luan ZK, Di ZC, Wang HY, Tian BH, Jia ZP. 2003. Adsorption of 1,2-dichlorobenzene from water to carbon nanotubes. Chem Phys Lett 376:154–158. Peterson DS, Luo Q, Hilder EF, Svec F, Fre´chet JMJ. 2004. Porous polymer monolith for surface-enhanced laser desorption/ionization time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 18:1504– 1512. Pinkston JD,Wen D, Morand KL, Tirey DA, Stanton DT. 2006. Comparison of LC/MS and SFC/MS for screening of a large and diverse library of pharmaceutically relevant compounds. Anal Chem 78:7467–7472. Poulsen SA. 2006. Direct screening of a dynamic combinatorial library using mass spectrometry. J Am Soc Mass Spectrom 17:1074–1080. Pullen FS, Perkins GL, Burton KI,Ware RS, Taegue MS, Kiplinger JP. 1995a. Putting mass spectrometry in the hands of the end user. JAm Soc Mass Spectrom 6:394–399. Pullen FS, Swanson AG, Newman MJ, Richards DS. 1995b. ‘Online’ liquid chromatography/nuclear magnetic resonance mass spectrometry—A powerful spectroscopic tool for the analysis of mixtures of pharma-ceutical interest. Rapid Commun Mass Spectrom 9:1003–1006. Rauha J, Vuorela H, Kostiainen R. 2001. Effect of eluent on the ionization efficiency of flavonoids by ion spray, atmospheric pressure chemical ionization, and atmospheric pressure photoionization mass spectrom-etry. J Mass Spectrom 36:1269. Reyzer ML, Hsieh Y, Ng K, Korfmacher WA, Caprioli RM. 2003. Direct analysis of drug candidates in tissue by matrix-assisted laser desorption/ ionization mass spectrometry. J Mass Spectrom 38:1081–1092. Richmond R. 2000. The analytical characterization of sub-minute measure-ment duty cycles in flow-injection analysis mass spectrometry, by their carry-over. Anal Chim Acta 403:287–294. Richmond R, Go¨rlach E. 1999a. The automatic visualization of carry-over in high-throughput flow injection analysis mass spectrometry. Anal Chim Acta 390:175. Richmond R, Go¨rlach E. 1999b. Sorting measurement queues to speed up the flow injection analysis mass spectrometry of combinatorial chemistry syntheses. Anal Chim Acta 394:33–342. Ripka WC, Barker G, Krakover J. 2001. High-throughput purification of compound libraries. Drug Disc Today 6:471–477. Robb DB, Covey TR, Bruins AP. 2000. Atmospheric pressure photo-ionization: An ionization method for liquid chromatography-mass spectrometry. Anal Chem 72:3653–3659. Sage AB, Little D, Giles K. 2000. Using parallel LC-MS and LC-MS-MS to increase sample throughput. LC-GC Mag Separation Sci 18:S20–S29. Salo PK, Pertovaara AM, Salo VMA, Salomies HEM,Konstiainen RK. 2003. High-performance thin-layer chromatography method for assessment of the quality of combinatorial libraries, and comparison with liquid chromatography-ultraviolet-mass spectrometry. J Comb Chem 5:223– 232. Santos LS, Haddad R, Hoehr NF, Pilli RA, Eberlin MN. 2004. Fast screening of low molecular weight compounds by thin-layer chromatography and ‘‘on-spot’’ MALDI-TOF mass spectrometry. Anal Chem 76:2144– 2147. Schmid DG, Grosche P, Bandel H, Jung G. 2001. FTICR-mass spectrometry for high-resolution analysis in combinatorial chemistry. Biotechnol Bioeng (Comb Chem) 71:149–161. Shah N, Gao M, Tsutsui K, Lu A, Davis J, Scheuerman R, FitchW. 2000. A novel approach to high-throughput quality control of parallel synthesis libraries. J Comb Chem 2:453–460. Shen Z, Thomas JJ, Averbuj C, Broo KM, Engelhard M, Crowell JE, Finn MG, Siuzdak G. 2001. Porous silicon as a versatile platform for laser desorption/ionization mass spectrometry. Anal Chem 73:612. SlenoLVD. 2005. Some fundamental and technical aspects of the quantitative analysis of pharmaceutical drugs by matrix-assisted laser desorption/ ionization mass spectrometry. Rapid Commun Mass Spectrom 19: 1928–1936. FANG ET AL. 32 Mass Spectrometry Reviews DOI 10.1002/mas
  • 14. Smyth WF. 2005. Recent applications of capillary electrophoresis-electro-spray ionisation-mass spectrometry in drug analysis. Electrophoresis 26:1334–1357. Somsen GW, Morden W, Wilson ID. 1995. Planar chromatography coupled with spectroscopic techniques. J Chromatogr A 703:613–665. Souverain S, Geiser L, Rudaz S,Veuthey J-L. 2006. Strategies for rapid chiral analysis by capillary electrophoresis. J Pharm Biomed Anal 40:235– 241. Spreen RC, Schaffter LM. 1996. Open access MS: A walk-up MS service. Anal Chem 68:414A–419A. Stolzberg LJ, Patel P. 2004. Small molecule matrix-assisted laser desorption/ ionization time-of-flight mass spectrometry using a polymer matrix. Rapid Commun Mass Spectrom 18:1455–1458. Sunner J, Dratz E, Chen YC. 1995. Graphite surface assisted laser desorption/ ionization time-of-flight mass spectrometry of peptides and proteins form liquid solutions. Anal Chem 67:4335–4342. Su¨ssmuth RD, Jung G. 1999. Impact of mass spectrometry on combinatorial chemistry. J Chromatogr B 725:49–65. Tanaka N, Kobayashi H, Ishizuka N, Minakuchi H, Nakanishi K, Hosoya K, Ikehami T. 2002. Monolithic silica columns for high-efficiency chromatographic separations. J Chromatogr A 965:35. Tiller PR, Romanyshyn LA, Leslie A, Neue UD. 2003. Fast LC/MS in the analysis of small molecules. Anal Bioanal Chem 377:788–802. Toribio L, Alonso C, del Nozal MJ, Bernal JL, Martin MT. 2006. Sempreparative enantiomeric separation of omeprazole by supercritical fluid chromatography. J Chromatogr A 1137:30–35. Triolo A, Altamura M, Cardinali F, Sisto A, Maggi CA. 2001a. Mass spectrometry and combinatorial chemistry: A short outline. J Mass Spectrom 36:1249–1259. Triolo A, Altamura M, Cardinale F, Sisto A, Maggi CA. 2001b. Mass spectrometry and combinatorial chemistry: A short outline. J Mass Spectrom 36:1249. Visky D, Jimidar I, Van Ael W, Vennekens T, Redlich D, De Smet M. 2005. Capillary electrophoresis-mass spectrometry in impurity profiling of pharmaceutical products. Electrophoresis 26:1541–1549. Wachs T, Henion J. 2001. Electrospray device for coupling microscale separations and other miniaturized devices with electrospray mass spectrometry. Anal Chem 73:632–638. Walk TB, Trautwein AW, Richter H, Jung G. 1999. ESI Fourier transform ion cyclotron resonance mass spectrometry (ESI-FT-ICR-MS): A rapid high-resolution analytical method for combinatorial compound libra-ries. Angew Chem Int Ed Engl 38:1763–1765. Wan H, Thompson RA. 2005. Capillary electrophoresis technologies for screening in drug discovery. Drug Discov Today Technol 2:171–178. Wang T, Zeng L, Strader T, Burton L, Kassel DB. 1998. A new ultra-high throughput method for characterizing combinatorial libraries incorpo-rating a multiple probe autosampler coupled with flow injection mass spectrometry analysis. Rapid Commun Mass Spectrom 12:1123. Wang T, Zeng L, Cohen J, Kassel DB. 1999.Amultiple electrospray interface for parallel mass spectrometric analyses of compound libraries. Comb Chem High Throuput Screen 2:327–334. Wang AQ, Zeng W, Musson DG, Rogers JD, Fisher AL. 2002. A rapid and sensitive liquid chromatography/negative ion tandem mass spectrom-etry MASS SPECTROMETRY ANALYSIS method for the determination of an indolocarbazole in human plasma using internal standard (IS) 96-well diatomaceous earth plates for solid-liquid extraction. Rapid Commun Mass Spectrom 16:975. Wehr T. 2003. Fast LC for high-throughput LC-MS. LCGC North America 21, Part 2 (Suppl):75–78. Wei J, Buriak JM, Siuzdak G. 1999. Desorption-ionization mass spectrometry on porous silicon. Nature 399:243–246. Weins C, Hauck HE. 1996. Advances and developments in thin layer chromatography. LC-GC Int 9:710–717. Williams SM, Brodbelt JS, Huang Z, Lai H, Marchand AP. 2003. Complex-ation of silver and co-recovered metals with novel aza-crown ether macrocycles by electrospray ionization mass spectrometry. Analyst 128:1352–1359. Wilson ID. 1999. The state of the art in thin-layer chromatography-mass spectrometry: A critical appraisal. J Chromatogr A 856:429–442. Wilson ID, Lindon JC, Nicholson JK. 2000. Advancing hyphenated chromatographic systems. Anal Chem 72:534A–542A. Winger BE, Campana JE. 1996. Characterization of combinatorial peptide libraries by electrospray ionization fourier transform mass spectrom-etry. Rapid Commun Mass Spectrom 10:1811–1813. Wu JY, Chen Y. 2002. A novel approach of combining thin-layer chromatography with surface-assisted laser desorption/ionization (SALDI) time-of-flight mass spectrometry. J Mass Spectrom 37:85–90. Xu SY, Li YF, Zou HF, Qiu JS, Guo Z, Guo BC. 2003. Carbon nanotubes as assisted matrix for laser desorption/ionization time-of-flight mass spectrometry. Anal Chem 75:6191–6195. Yan B, Chu YH, Shapiro M, Richmond R, Chin J, Liu L, Yu Z. 2000. Analytical methods in combinatorial chemistry. In: Fenniri H, editor. Combinatorial chemistry: a practical approach. Oxford UK: Oxford University Press. pp. 263–286. Yan B, Collins N, Wheatley J, Irving M, Leopold K, Chan C, Shornikov A, Fang L, Lee A, Stock M, Zhao J. 2004. High-throughput purification of combinatorial libraries I: A high-throughput purification system using an accelerated retention window approach. J Comb Chem 6:255–261. Yu K, Balogh M. 2000. A protocol for high-throughput drug mixture quantitation: Fast LC-MS or flow injection analysis-MS? LC-GC 19: 60–72. Yurek D, Branch DL, Kuo M-S. 2002. Development of a system to evaluate compound identity, purity, and concentration in a single experiment and its application in quality assessment of combinatorial libraries and screening hits. J Comb Chem 4:138–148. Zhang QC, Zou HF, Guo Z, Zhang Q, Chen XM, Ni JY. 2001. Matrix-assisted laser desorption/ionization mass spectrometry using porous silicon and silica gel as matrix. Rapid Commun Mass Spectrom 15:217–223. Zhang LK, Rempel D, Pramanik BN, Gross ML. 2005. Accurate mass measurements by fourier transform mass spectrometry. Mass Spectrom Rev 24:286–309. Zhang X, Towle MH, Felice CF, Flament JH, Goetzinger WK. 2006. Development of a mass-directed preparative supercritical fluid chromatography purification system. J Comb Chem 8:705–714. Zweigenbaum J, Heinnig K, Steinborner S, Wachs T, Henion J. 1999. High-throughput bioanalytical LC/MS/MS determination of benzodiazepines in human urine:1000 samples per 12 hours. Anal Chem 71:2294–2300. Annette S. Fang, Ph.D. Dr. Fang received her undergraduate education in Germany and her Ph.D. degree in Analytical Chemistry from Northeastern University. She was a Staff Investigator in the Analytical Department at ArQule, primarily focused on purification of new chemical entities synthesized for early drug discovery. Her prior work experiences include positions at Research Biochemicals, Inc., Natick, MA and Bruker Daltonics, Billerica, MA. Mass Spectrometry Reviews DOI 10.1002/mas 33
  • 15. Xiusheng Miao, Ph.D., Senior Investigator, Preclinical Development and Clinical Pharmacology, ArQule, Inc., Woburn, MA. Dr. Miao is currently responsible for in vitro and in vivo drug metabolite identification and bioanalytical method development. He obtained his Ph.D. from the Chinese Academy of Sciences, China and has co-authored over 40 journal articles and book sections. Peter W. Tidswell, Ph.D., Associate Research Fellow, Pfizer, Inc. Dr. Tidswell leads the purification laboratory at the St. Louis research site, which supports the medicinal chemistry department and therapeutic programs. Prior to joining Pfizer, he was a Group Leader in the Chemical Technologies’ analytical department for ArQule, where he led the Character-ization group. His other experience encompasses positions at the Bristol-Myers Squibb Pharmaceutical Research Institute, EvotecOAI and ChiroScience. Dr. Tidswell received his Ph.D. under the direction of Dr. Mel Kilner in Organometallic Chemistry from the University of Durham, UK. Marc H. Towle, Scientist, Discovery Analytical,Vertex Pharmaceuticals, Inc., Cambridge, MA. Mr. Towle provides analytical development support to assess the purity, potency and stability of drug substances and drug products for late research, pre-clinical, and phase I development. Prior to joining Vertex, he was a Senior Investigator at ArQule, where he managed the analytical purification group responsible for supporting high throughput production of chemical libraries for early drug discovery. His prior experience includes research positions with BioDevelopment Laboratories, Arthur D. Little and Groundwater Technology. Mr. Towle received his BS in Biochemistry from the University of New Hampshire. Wolfgang K. Goetzinger, Ph.D., Director Research, Molecular Structure, Amgen, Inc., Cambridge, MA. Dr. Goetzinger is responsible for the Discovery Analytical group at Amgen, Inc. where he leads a team supporting all analytical aspects of small molecule pharmaceutical discovery. Prior to joining Amgen, he was Director of Analytical Chemistry at ArQule, Inc., where he managed a group focused on high throughput analytical approaches to support parallel synthesis efforts. Dr. Goetzinger worked as a Post Doctoral Fellowunder Professor Barry L. Karger at Northeastern University, where he focused on life science applications of capillary electrophoresis. He received his Ph.D. from the University Saarland under Professor Heinz Engelhardt working on the synthesis and characterization of chiral stationary phases for the separation of enantiomers. James N. Kyranos, Ph.D., Vice President, Preclinical Development, Wolfe Laboratories, Watertown, MA. Dr. Kyranos leads the technical and business operations of WLI’s preclinical development services focused on bioanalytical and pharmaceutical sciences support. Prior to joining WLI, he was Vice President of Chemical Technologies for ArQule, where he managed the chemistry services business focused on providing early discovery services to the pharmaceutical industry. His other experience includes leadership positions at BioDevelopment Laboratories and Arthur D. Little. Dr.Kyranos received his Ph.D. under the direction of Professor Paul Vouros in Analytical Chemistry from Northeastern University. FANG ET AL. 34 Mass Spectrometry Reviews DOI 10.1002/mas