FG 15

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 25

CHIRALITY 19:658–682 (2007)

Review Article
Enantiomeric Separation and Determination of Absolute
Stereochemistry of Asymmetric Molecules in Drug
Discovery—Building Chiral Technology Toolboxes
OLIVER MCCONNELL,1 * ALVIN BACH II,2 CARL BALIBAR,1 NEAL BYRNE,2 YANXUAN CAI,2 GUY CARTER,3
MICHAEL CHLENOV,2 LI DI,2 KRISTI FAN,2 IGOR GOLJER,1 YANAN HE,1 DON HEROLD,2 MICHAEL KAGAN,2
EDWARD KERNS,2 FRANK KOEHN,3 CHRISTINA KRAML,2 VASILIOS MARATHIAS,2 BRIAN MARQUEZ,1
LEONARD MCDONALD,3 LISA NOGLE,1 CHRISTOPHER PETUCCI,1 GERHARD SCHLINGMANN,3 GREGORY TAWA,2
MARK TISCHLER,3 R. THOMAS WILLIAMSON,3 ALAN SUTHERLAND,3 WILLIAM WATTS,1 MAIREAD YOUNG,3
MEI-YI ZHANG,2 YINGRU ZHANG,1 DAHUI ZHOU,2 AND DOUGLAS HO4
1
Wyeth Research, Chemical and Screening Sciences, Collegeville, Pennsylvania
2
Wyeth Research, Chemical and Screening Sciences, Princeton, New Jersey
3
Wyeth Research, Chemical and Screening Sciences, Pearl River, New York
4
Department of Chemistry, Princeton University, Princeton, New Jersey

ABSTRACT The application of Chiral Technology, or the (extensive) use of techni-


ques or tools for the determination of absolute stereochemistry and the enantiomeric or
chiral separation of racemic small molecule potential lead compounds, has been critical
to successfully discovering and developing chiral drugs in the pharmaceutical industry.
This has been due to the rapid increase over the past 10–15 years in potential drug can-
didates containing one or more asymmetric centers. Based on the experiences of one
pharmaceutical company, a summary of the establishment of a Chiral Technology tool-
box, including the implementation of known tools as well as the design, development,
and implementation of new Chiral Technology tools, is provided. Chirality 19:658–682,
2007. V C 2007 Wiley-Liss, Inc.

KEY WORDS: Chiral Technology; chiral separations; enantiomeric separations;


determination of absolute stereochemistry; supercritical fluid
chromatography; asymmetric transformations using enzymes; chiral
salt resolution; vibrational circular dichroism; electronic circular
dichroism; exciton coupling; optical rotation; modified Mosher’s or
Trost NMR method; chiral liquid crystal NMR

INTRODUCTION
In contrast to previous usage, Chiral Technology is defined in this review
In the United States, major pharmaceutical companies as the use of techniques or tools for the determination of absolute stereo-
are primarily responsible for discovering and bringing to chemistry, the chiral analysis or preparative separation of racemic small
the patient life-improving and life-saving drugs. To con- molecules containing one or more asymmetric centers into (all of) their
enantiomers (¼ enantiomeric or chiral separation), and the facilitation of
tinue to foster strong scientifically viable research pro- asymmetric transformations, but not asymmetric transformations them-
grams, as the ‘‘R’’ in ‘‘R&D’’ is sometimes a casualty when selves. The terms Chiral Technology and Chirotechnology have often
been used as a general category for asymmetric transformations using
a need arises to triage costs in difficult financial times, enzymes, chiral building blocks, chiral auxiliaries, and chiral catalysts, as
these companies must constantly reassess and improve well as chiral salt resolution.1–13
research and development strategies. For drug discovery Carl Balibar is currently at Harvard Medical School, Department of Biological
Chemistry and Molecular Pharmacology, Cambridge, Massachusetts.
programs in pharmaceutical companies, the need to Christina Kraml and Neal Byrne are at Accelapure, Princeton University, New
improve and produce creates tremendous challenges for Jersey.
all groups, including high throughput screening, explora- Brian Marquez is currently at Pfizer, Groton, Connecticut.
R. Thomas Williamson is currently at Roche Carolina Inc, Florence, South
tory chemistry, medicinal chemistry, analytical chemistry, Carolina.
discovery synthetic chemistry, computational chemistry, Yingru Zhang is currently at Bristol-Meyers-Squib, Lawrenceville, New Jersey.
*Correspondence to: Dr. Oliver J. McConnell, Discovery Analytical Chem-
biological chemistry, biology in a myriad of therapeutic istry, Chemical Sciences, Wyeth Research, 500 Arcola Road, Collegeville,
areas, structural biology, including protein expression, pu- Pennsylvania 19426, USA. E-mail: mcconno@wyeth.com
rification and characterization, translational medicine, and Received for publication 30 November 2006; Accepted 9 February 2007
DOI: 10.1002/chir.20399
discovery drug safety and metabolism. Barely scratching Published online 27 March 2007 in Wiley InterScience
the surface, a few of the challenges include addressing the (www.interscience.wiley.com).
C 2007 Wiley-Liss, Inc.
V
BUILDING CHIRAL TECHNOLOGY TOOLBOXES 659

complexity of identifying new biological targets involved in the requirement that each enantiomer must be evaluated
difficult-to-cure diseases, while maneuvering through a when developing stereoisomeric drugs.184 Consequently,
crowded chemical space and difficult intellectual property on top of all of the difficulties and hurdles involved in drug
environment, and making valiant efforts to manage, discovery that have been referred above, there are addi-
archive, and disseminate large numbers of compounds, as tional challenges from discovering and developing asym-
well as huge and multiple databases containing invaluable metric molecules. These include producing and analyzing
information needed across functions and often across dif- enantiomerically pure synthetic intermediates and final tar-
ferent research sites. At the same time, companies are try- gets, and determining the absolute stereochemistry of
ing to shorten the drug discovery timelines from target these molecules. In many pharmaceutical companies,
identification and high throughput screening through the these activities have begun in the early exploratory phase
exploratory and discovery phases into development to no of the discovery process and with greater intensity.
more than a few years. Companies are also determining Although the ultimate goal for all of the effort described
where outsourcing and partnering are an effective means above is seemingly quite simple—cure diseases in
of reducing overall costs without sacrificing the quality of humans, Figure 1 attempts to provide a visual context to
research and development, compromising intellectual position Chiral Technology in the highly complex and
property, and extending timelines. Consequently, pharma- evolving drug discovery environment with its numerous
ceutical companies are applying to their drug discovery challenges, hurdles, and tools.
efforts all available and appropriate chemical, molecular To prepare and analyze enantiomerically pure synthetic
biological and computational tools—virtual screening, in intermediates and final targets and determine their abso-
silico rational drug design,14–28 bioinformatics,29 chemo- lute stereochemistry, pharmaceutical companies have
metrics,30,31 the ’omics32–77—genomics, chemogenomics, needed to develop what we are describing as Chiral Tech-
proteomics, metabolomics, and metabonomics, parallel nology toolboxes. Similar to repairing a vehicle, where
synthesis,78–84 ultrafast purification and analysis,85–93 X-ray having sufficient knowledge to effectively troubleshoot the
crystallography of proteins and potential lead compounds problem, having the appropriate tools at one’s disposal,
from parallel synthesis,94 and novel applications of high- and knowing how to use the tools well are essential to an
powered and high field NMR95–98 and FT-MS,99–103 in efficient and successful repair, so it is with the various Chi-
addition to high throughput and high content screening ral Technology tools available to carry out the tasks men-
paradigms.104–114 tioned above. Because it is not possible to know the details
In the past, discovering a potential drug candidate of Chiral Technology toolboxes at other pharmaceutical
focused ideally on finding a compound with nanomolar or companies, described herein is what we do know, i.e., the
better activity and exquisite selectivity toward the target collective efforts of numerous chemists at Wyeth
receptor or enzyme. Now, the quality of compounds com- Research, whom we believe exemplify the practice in
ing from drug discovery and entering development must industry, to use these tools as well as develop new ones,
also be dramatically improved with respect to their drug- and then apply them in effective ways.
like properties,115–133 i.e., physicochemical, pharmacoki-
netic, and safety (or toxicity and drug-drug interaction)
properties, to reduce the attrition rate of drug candidates
later in clinical trials. Structure–activity-relationship (SAR) EARLY CHIRAL TECHNOLOGY TOOLBOX
studies are now carried out in parallel with structure–prop- Determination of Absolute Stereochemistry
erty-relationship (SPR) studies during lead optimization A decade ago, viable techniques for the determination of
from the early exploratory phase all the way through the absolute stereochemistry of small molecule drugs that were
discovery phase into the predevelopment phase.134–167 and are still available at our company include X-ray crystallog-
Further, chirality in the form of drugs containing one or raphy,185,186 comparison of specific optical rotation values and
more asymmetric centers plays a critical role in pharma- circular dichroism curves,187,188 circular dichroism using exci-
ceutical research and development.168–177 The evidence ton-coupling in combination with molecular modeling studies
for this is clear as in 1990, nearly a third of the drugs of the lowest energy conformers,189–197 and NMR using the
brought to market were either racemates or diastereom- (modified) Mosher’s or Trost approach,198–203 where func-
ers.178 Over the past several years, only one or two drugs tional groups such as secondary alcohols, primary amines, or
have been brought to market as racemic or diastereomeric carboxylic acids with substituents in the a- or b- positions are
mixtures.179,180 In 2004, five of the top ten selling drugs amenable to derivatization. For the determination of absolute
contained single-enantiomers, and this accounted for stereochemistry, X-ray crystallography185,186 is commonly
$32.4B or *61% of worldwide sales.181 (Although in race- viewed as the most reliable technique, however, the caveats
mic form, the active ingredients of three additional top ten- for this approach are the ability to prepare crystals suitable
selling drugs featured asymmetric centers.) The enantiomers for analysis and the recognition that X-ray analysis may be
that have been brought to market, include, of course, the incorrect, albeit rarely.204,205 X-ray crystallography and CD
so-called ‘‘chiral’’ or ‘‘racemic switches’’ where an enan- analysis have been available through important university col-
tiomer of a drug is (re)developed and marketed instead of laborations; several examples of compounds where absolute
the racemate, and, ideally, shortly before patent expiration stereochemistry has been determined by X-ray crystallogra-
of the racemate occurs.175–177,182,183 Several years ago, the phy have been published (Fig. 2).206–209 Application of the
US Food and Drug Administration (FDA) weighed in with exciton chirality CD method to compounds that already con-
Chirality DOI 10.1002/chir
660 MCCONNELL ET AL.

Fig. 1. Positioning Chiral Technology in a highly complex and evolving drug discovery environment.

tain two or more chromophores has been amply demon- preparative enantiomeric (or chiral) HPLC separation on the
strated on a variety of compounds,190,192,193 including micro- more commonly used polysaccharide, antibiotic, or Pirkle-
bially-derived natural products originating from the Wyeth brush type chiral stationary phases (CSPs).214–220 Besides
Research Natural Products Group (Fig. 3).210–213 the scientific literature and vendor’s brochures and catalogs,
searchable databases are also available, e.g., Chirbase'.221–225
Enantiomeric Analysis and Preparative Separation Using various molecular modeling and statistical techniques,
Over the past several decades, considerable research and the enantioselective binding properties of solutes with CSPs
experimentation have yielded a sizeable body of knowledge are slowly beginning to be understood.226–230 However, a
from which a chromatographer can extract exact conditions successful separation is often not predictable based on
or at least guidelines to carry out a successful analytical or analyte structure and CSP, and the common approach

Fig. 2. Examples of compounds where X-ray crystallographic analysis was used for determination of absolute stereochemistry.

Chirality DOI 10.1002/chir


BUILDING CHIRAL TECHNOLOGY TOOLBOXES 661

Fig. 3. Examples of natural products where CD was employed to determine absolute stereochemistry.

continues to rely simply (or perhaps not so simply) on ana- Chiral Technology would be timely and productive. Conse-
lyte screening by HPLC (or SFC - vide infra) on the avail- quently, an initiative was undertaken several years ago at
able CSPs in the analyst’s lab.231–234 our company with the expressed purpose of enhancing
In the area of chiral separations, capabilities available a chiral drug discovery by increasing or enhancing exper-
decade ago and primarily used in Discovery at our company tise, capabilities, and capacity in: (1) enantiomer separa-
included analytical and preparative chiral HPLC with stand- tion including purification, and chemical and biochemical
ard UV detection, which are commonly used techniques, resolution; (2) enantiomer analysis using HPLC, SFC, GC,
and chiral salt resolution. During this time, the analytical CE, and possibly MS; and (3) determination of absolute
chiral HPLC instrumentation featured automated method stereochemistry using X-ray crystallographic and spectro-
development using gradient solvent programming and scopic techniques, and computational means. The initiative
column switching devices. The chiral preparative HPLC resulted in several years of quarterly working group meet-
instrumentation featured automated injection and solvent ings at all of our research sites, seminars given by experts
recycling. In contrast to Development groups at most compa- in academia and select companies, symposia, evaluation
nies, the preparative setups in the Discovery Analytical Chemis- and implementation of various chiral separation and abso-
try (DAC) group at Wyeth Research employed primarily 2 cm lute configuration technologies, and original and ongoing
id columns, using for the most part polysaccharide-based and research to explore new techniques in these areas. Very
Pirkle-type CSPs, and injection quantities of the order of 10s importantly, the efforts were successful in large part by
to 100s of mg. Alternatively, enantiomeric separations were working closely with key external companies and aca-
achieved by preparation of chiral salts. In those cases where it demic groups. In-house research as well as collaborations
was deemed more efficient and cost effective, samples were with colleagues at universities and instrument companies
analyzed externally by a company specializing in chiral salt re- are ongoing.
solution using their unique process, Dutch Resolution, The current status of our Chiral Technology toolbox
whereby families of chemically related chiral salts are used derived from this initiative is found in Table 1.
for resolution.235–240 Because the analytical techniques used
to assess enantiomeric excess have been linked closely to
preparative separations, chiral capillary electrochromato- CURRENT TOOLBOX—CHIRAL SEPARATIONS
graphic techniques241–245 and chiral GC-MS246–249 have nor-
mally been used as secondary analytical approaches. A tremendous enhancement to the Discovery analyst’s
separation toolbox took place with the advent of commer-
cially-available packed-column sub- and supercritical auto-
mated SFC instrumentation with stacked injection.250–253
CHIRAL TECHNOLOGY INITIATIVE Sub- and supercritical fluids with modifiers and additives
The dramatic increase over the past decade of potential typically manifest an analyte selectivity that is not quite
drug leads with asymmetric centers, especially in the CNS the same as in HPLC. The greater diffusivity and lowered
area,169 made it apparent that an initiative focusing on viscosity of supercritical fluids lead to higher linear
Chirality DOI 10.1002/chir
662 MCCONNELL ET AL.

TABLE 1. Discovery chiral technology toolbox bed (SMB) instrumentation that has become available
commercially. SMC may become an especially useful
Enantiomeric (chiral) separations—Analytical and preparative:
(Automated) analytical and preparative HPLC
technique in those preparative cases where only partial
(Automated) analytical and preparative sub- and supercritical resolution is obtained rather quickly, and it is deemed
fluid chromatography (SFC) with stacked injection by the chromatographer more efficient to use a column
Chiral salt resolution switching approach than to exhaustively study station-
Semiautomated enzyme screening system for asymmetric ary phases, solvents, and additives to achieve ideal prep-
transformations using enzymes arative separation conditions, or to use SMB.264–271
Determination of absolute stereochemistry:  A novel strategy for rapid chiral method development
X-ray crystallography uses multicolumn parallel screening and circular
Circular dichroism (exciton chirality and porphyrin tweezers) dichroism signal pooling to shorten chiral method de-
Measured versus calculated and comparison methods:
velopment by at least one-fifth of the time.272 This
Vibrational circular dichroism (VCD)
Electronic circular dichroism (ECD)
approach splits the injection of one sample of race-
Optical rotation (OR) mate and pumps the split sample in parallel through
NMR methods: at least five different chiral stationary phases followed
(Modified) Mosher’s and Trost methods by an equal number of UV detectors. The eluent com-
Chiral liquid crystals ing out of each UV detector is then recombined by
flowing into a multiport valve. The single outlet from
this valve directs the recombined flow into a CD de-
tector. The signal pooling from the multiple columns
velocities and higher theoretical plates in SFC separations. and UV detectors allows for the effective use of a sin-
This translates typically to faster separations (per unit gle CD detector for multiple columns since unresolved
time) and fractions with higher purities when compared racemate has little CD signal, and observing the sign
with preparative HPLC. Further, the features of stacked of CD signal for one of the two-enantiomer UV peaks is
injection252 on 2 cm id columns and CO2-based purification sufficient for tracking the enantiomeric elution order.
systems provide high yields of fractionated materials and  A novel environmentally friendly fluorinated solvent,
in small (alcohol) solvent volumes and facilitate the proc- ethoxynonafluorobutane, was found to complement n-
essing of gram quantities of racemate per hour, with mini- hexane in normal-phase HPLC applications.273 Ethoxy-
mal follow-up processing. (Five cm id columns containing nonafluorobutane, which is miscible in all polar organic
CSPs are also currently available, but are used exclusively solvents, and its mixtures with methanol were de-
for separations by HPLC.) The primary CSPs now used for monstrated to have unique selectivity in chiral HPLC
enantiomeric preparative separation are the coated as well applications.
as the immobilized polysaccharide-based materials,254,255  The use of atmospheric pressure chemical ionization
although the Pirkle-type columns continue to find utility. (APCI)-mass spectrometry in combination with ethoxy-
Similar to identifying parameters for successful enantio- nonafluorobutane, was used effectively for normal-
meric separation by HPLC,231–234 screening CSPs, alcohol phase HPLC separations, including both achiral and
cosolvents, and acidic and basic modifiers are integral chiral separations, on a wide variety of structural
components in developing successful chiral separations by classes.274 (Hexane is not a suitable solvent for APCI-
SFC.256–258 A note of caution is appropriate as reversal of MS due to safety considerations.) These findings were
elution order of enantiomers is neither uncommon nor subsequently verified by a well-known, independent
currently predictive in HPLC and SFC separations.259–262 lab.275 A follow-up study in our labs examined this LC-
Therefore, spectroscopic characterization of the eluted (APCI) MS approach by carrying out mass-directed
enantiomers is paramount to successfully correlating con- (auto) purification on members of the same classes
figuration with eutomer (biologically active enantiomer) of compounds276 as well as on non-UV-active com-
and distomer (biologically inactive enantiomer). pounds.277 One example described the normal phase
Since our Chiral Technology initiative began, consider- separation on a CN stationary phase of optically-active
able effort has been made in the general area of enantio- diastereomers formed from a racemic acid that fea-
meric separations or chiral chromatography to become tures an a-methyl group and (1S)-()-2,10-camphorsul-
more efficient, productive, and creative. Brief descriptions tam, which appears to be a useful derivative in general
of many of these efforts are provided below: to effect enantiomeric separations of chiral acids.277
 The use of atmospheric pressure photoionization
 An HPLC technique titled ‘‘simulated moving columns’’ (APPI)-mass spectrometry to analyze a wide range of
(SMC)263 uses two or three shorter chiral columns compounds was found to be especially useful for non-
connected in series and forces partially resolved enan- functionalized and nonionizable compounds, primarily
tiomers to recycle exclusively through the columns until synthetic intermediates, that do not ionize well under
sufficient resolution is attained. (This technique was also electrospray (ESI) or APCI conditions.278 This tech-
coined ‘‘column leap frogging’’ in the mid-90s by W. Let- nique can be applied to analyze racemic or enantio-
ter (Chiralizer Services, L.L.C.) and implemented on an merically-enriched compounds.
HPLC sample peak recycler.) SMC (or column lead frog-  The carbobenzyloxy (cbz) protecting group was used
ging) complements new smaller-scale simulated moving successfully to enhance the resolution of chiral amine
Chirality DOI 10.1002/chir
BUILDING CHIRAL TECHNOLOGY TOOLBOXES 663

Fig. 4. Compounds 1, 2, and 3 that were subjected to chiral preparative SFC and/or HPLC. (Asymmetric center(s) represented by asterisk (‘‘’’).)

enantiomers using HPLC and SFC.279 Out of the using enzymes. This may also be due to the perception
numerous amines studied, the cbz derivatives were that the process of identifying an enzyme that affects an
amenable to enantiomeric separation on at least one asymmetric transformation, and then optimizing condi-
and in some cases up to five CSPs, which was in con- tions under which to run the reaction, may be time con-
trast to the parent amines where either enantiomeric suming and not straightforward. To ‘‘lower the energy
separation could not be achieved at all or, if achieved, barrier’’ in using enzymes in synthesis in Discovery (as a
only on one or sometimes two CSP supports. Further Biotransformation Group exists in Development at Wyeth
fine-tuning of analytical and preparative enantiomeric Research), a semi-automated enzyme screening system
separation of cbz derivatives of amines could be was developed that employs a user-friendly programmable
achieved through solvent selectivity using a variety of liquid sample handler, and originally exemplified with a
alcohols in addition to methanol.280 rapid (1.5 min) chiral SFC analysis.295 The method accel-
erates the enzyme selection process for screening biocata-
Examples of Compounds 1, 2, and 3 (Fig. 4) that were lysts, where a large number of enzymes were evaluated
subjected to chiral preparative SFC and HPLC can be for activity and enantioselectivity. Kinetic resolution of sec-
found in Figures 5–7. ondary alcohols by enzymatic transesterification was used
as a prototype for method development. The rapid auto-
mated method can be used effectively for screening
enzymes and optimizing reaction conditions in biocataly-
CURRENT TOOLBOX—ENZYME SCREENING
sis. This method has been extended to use chiral GC-MS
In general, chiral separations at our company are a col- as the detection method (C. Petucci, L. Di, and O.
laborative effort between the analytical, and medicinal or McConnell, unpublished).
synthetic chemists. The preference is to put into place an
asymmetric synthetic route or perhaps develop a chiral
salt resolution step for the more promising discovery leads
that require larger quantities of compound for biological
evaluation, and rely on the discovery analytical groups for CURRENT TOOLBOX—DETERMINATION OF
chiral analysis. This approach is not practical for most chi- ABSOLUTE STEREOCHEMISTRY
ral compounds in the exploratory and discovery phases as More recently, in addition to X-ray crystallographic anal-
it often takes too much time to develop an asymmetric syn- ysis, CD analysis with exciton coupling, and the modified
thesis, and time is of the essence, especially in the earlier Mosher’s or Trost NMR approach, several other techni-
exploratory phase. On the other hand, resources for chiral ques have become available at Wyeth Research for the
separations by HPLC or SFC are not without limitation. A determination of absolute stereochemistry. These techni-
well-known alternative approach to chiral HPLC and SFC ques include CD analysis of porphyrin tweezer conjugates,
and chiral salt resolution that can be used to effect a chiral calculated versus measured vibrational circular dichroism
resolution or asymmetric transformation is to take advant- (VCD), electronic circular dichroism (ECD), and optical
age of enzymes,281–284 e.g., the enantioselective acylation rotation (OR), new modified Mosher’s or Trost NMR
of an alcohol or amine,285–290 enantioselective esterifica- methods and the use of chiral liquid crystals as solvents
tion of an acid,291 or enantioselective hydrolysis of an ester for NMR-based determination of absolute stereochemistry.
to an acid.292–294 Consequently, enzymatic resolution or An important collaboration helped lead to the characteriza-
asymmetric transformation using enzymes represents a tion of the (zinc) porphyrin tweezer conjugates, which
viable, however, often underutilized approach in Discov- express exciton coupling in their circular dichroism (CD)
ery. In part, this may be due to a general lack of familiarity properties.296–299 These complexes have been shown to be
Chirality DOI 10.1002/chir
664 MCCONNELL ET AL.

Fig. 5. HPLC and SFC chromatograms for Compound 1: (a) analytical HPLC - Chiralcel OJ (4.6 3 250 mm), 100% MeOH, 1 ml/min, 258C; (b) analyt-
ical HPLC - Chiralpak AS (4.6 3 250 mm), 100% MeOH, 1 ml/min, 258C; (c) analytical SFC - Chiralpak OJ (4.6 3 250 mm), 30% MeOH/CO2, 1 ml/min,
100 bar; (d) analytical SFC - Chiralpak AS 20% MeOH/CO2, 1 ml/min, 100 bar; (e) Chiralpak AS (2 3 25 cm); 20% MeOH/CO2, 30 mg/0.4 ml injections,
(f) same as (e) 10 stacked injections, 115 sec intervals; (g) same as (e) 90 stacked injections, 110 sec intervals, 1 g processed/h.
BUILDING CHIRAL TECHNOLOGY TOOLBOXES 665

Fig. 6. HPLC and SFC chromatograms for Compound 2: (a) analytical HPLC - Chiralcel OD (4.6 id x 250 mm), 100% MeOH, 1 ml/min; b) analytical
SFC - Chiralpak OD 50% MeOH/CO2, 2 ml/min, 100 bar; (c) semipreparative HPLC - Chiralcel OD (2 3 25 cm) 100% MeOH 13 ml/min, 43 mg/injec-
tion; (d) Chiralcel OD (2 3 25 cm); 50% MeOH/CO2, 50 ml/min, 20 mg/0.4 ml injections, (e) 40 stacked injections, 115 sec intervals, 1 g processed/
1.5 h.

Chirality DOI 10.1002/chir


666 MCCONNELL ET AL.

Fig. 7. HPLC and SFC chromatograms for Compound 3: (a) semipreparative SFC - Chiralcel OJ (2 3 25 cm); 20% 2-BuOH/CO2, 50 ml/min, 40 mg/
0.4 ml injections, (b) 50 stacked injections, 205 sec intervals, 1 g processed/1.5 h.

highly effective in determining the absolute stereochemis- cifically to pharmaceutically-relevant molecules,325,335,336


try of secondary alcohols and primary amines. as well as numerous reports where various permutations
of calculated versus measured VCD, ECD and/or OR have
been used to determine absolute configuration.337–340 The
combination of calculated versus measured VCD and
CURRENT TOOLBOX—OPTICAL SPECTROSCOPY NMR,341 CD using porphyrin tweezers and NMR,342 and
APPROACHES calculated versus measured CD plus X-ray analysis have
The spectroscopic approach of comparing observed and also been reported.343 Sometimes, however, difficulties
calculated vibrational circular dichroism (VCD) spec- may be encountered when applying one or more of these
tra,300–305 electronic circular dichroism (ECD),197,306–308 or techniques, for example CD318,344 or OR.313,314,341
optical rotation (OR)309–318 values has gained popularity VCD has been used in our labs to determine the abso-
due to the ease of data acquisition and the ability to calcu- lute stereochemistry of compounds that are difficult to
late spectra and values with commercially-available soft- crystallize for X-ray analysis or lack functional groups ame-
ware running on supercomputers or modern, powerful nable to derivatization for analysis by NMR using either
PCs configured as Linux clusters or configured individu- the modified-Mosher’s or Trost approach. VCD is espe-
ally running under Microsoft operating systems. The VCD cially useful for conformationally rigid molecules with
approach entails comparing measured VCD and IR spectra relatively few low energy conformers, although conforma-
of one enantiomer with the spectra calculated for one abso- tionally flexible compounds aren’t precluded. Over the
lute configuration using ab initio calculations. When the past several years, VCD has been employed in Discovery
VCD spectrum of the target compound expresses bands at our company more than any other technique for the
similar in sign, wave number, and intensity (with the cal- determination of absolute stereochemistry. In general,
culated VCD spectrum), the absolute configuration chosen VCD calculations in our lab have used hybrid density func-
for the calculation is deduced as the same as that for the tional theory with the 6-31G(d) basis set as a minimum in
measured molecule. Besides determining absolute stereo- terms of the best trade-off between accuracy and computa-
chemistry of molecules containing one or more asym- tional cost. The B3LYP hybrid functional and 6-31G(d) ba-
metric centers, or planes or axes of asymmetry, i.e., atro- sis set utilizes the magnetic field perturbation method
pisomerism,319–324 additional information about solution with gauge invariant atomic orbitals; however, more
conformation can be obtained.304,325–327 Also, VCD can be recently, the B3PW91 hybrid functional and TZVP basis
used for real-time reaction monitoring.328–330 VCD has set have been found to be a useful alternative functional/
been used to determine the absolute stereochemistry of basis set when a suitable comparison has not been real-
natural products,320,331 compounds derived via asymmetric ized using B3LYP and 6-31G(d), and found to be especially
enzymatic catalysis,332 as well as carboxylic acids333,334 useful for comparing observed and calculated ECD and
and alcohols204 in derivatized form in order to improve cal- OR in those cases when use of B3LYP/6-31G(d) may
culations by obviating the presence of dimers or rigidify- effectively fail.
ing the molecule of interest, respectively. There are An example of VCD is provided by the differentiation
numerous published examples of VCD being applied spe- of the (R)- and (S)-enantiomers of Lodine1 (etodolac, 4)
Chirality DOI 10.1002/chir
BUILDING CHIRAL TECHNOLOGY TOOLBOXES 667

Fig. 8. Structure of Lodine (etodolac: (+)-1,8-diethyl-1,3,4,9-tetrahydropyrano-[3,4-b]indole-1-acetic acid), 4, and the four lowest energy (S)-etodolac
conformers with relative energy (RE in kcal/mol) and percent composition.

(D. Herold, K. Fan, and O. McConnell. Determination of The VCD spectra are overlaid with the Boltzmann distri-
the Absolute Configuration of Etodolac using VCD Analy- bution of the calculated spectra for the lowest energy
sis. The 43rd Eastern Analytical Symposium (EAS) and conformers. The calculated VCD spectrum for the (R)-
Exposition, November 15–18, 2004, Somerset, NJ), a non- enantiomer is found to closely resemble the spectrum of
steroidal anti-inflammatory drug (NSAID) that is used in (R)-etodolac and is quite different from the spectrum of
humans and for certain indications in animals.345–348 The (S)-etodolac; therefore, the enantiomer previously deter-
enantiomers of Lodine have been either separated by chi- mined to be (R)- was confirmed.
ral salt formation and identified by X-ray crystallographic VCD measurement of only one enantiomer is sufficient
analysis or have been synthesized.349–352 The structure to unambiguously determine its absolute stereochemistry;
and four low energy conformers for (R)-enantiomer of however, measurement of both enantiomers is preferred
Lodine are found in Figure 8. The relative energies shown in order to minimize potential artifacts, including any base-
in the figure are the energies calculated using DFT with line irregularities.
the B3LYP/6-31G(d) basis set.
The IR and VCD spectra of 5–10 mg each of (R)- and
(S)-etodolac (4) were measured for *4 h each in d6- CURRENT TOOLBOX—NMR APPROACHES
dmso. Composite IR and VCD spectra of each enantiomer, Various optically-active derivatizing reagents have been
over the range of 1150–1800 cm1, are shown in Figure 9. studied and used for over thirty years in the so-called
Chirality DOI 10.1002/chir
668 MCCONNELL ET AL.

Fig. 9. (a) Comparison of observed and calculated IR and VCD spectra between 1150 and 1800 cm1 for (S)-etodolac, 4 - (S), and Boltzmann mix of
the lowest energy conformers, and (b) comparison of observed and calculated VCD spectra between 1150 and 1800 cm1 for (R)-etodolac, 4 - (R), and
Boltzmann mix of the lowest energy conformers.

modified Mosher’s or Trost NMR approach, and include removed easily by treatment with tetrabutylammonium flu-
(+)-(R)- and ()-(S)-a-methoxy-a-(trifluoromethyl) phenyl- oroborate (TBAF) should recovery of the alcohol be desir-
acetic acid (MTPA acid), and ()-(R)- and (+)-(S)-a- able or necessary.
methoxyphenylacetic acid (MPA acid).198–203 These and In general, the caveats in the (modified) Mosher’s or
structurally-related acids as well as several types of opti- Trost NMR approach are that the chiral compound must
cally-active alcohols have found great utility to determine be amenable to derivatization and effectively derivatized,
the absolute stereochemistry of enantiomerically pure or the reaction mixture in some cases must be purified, and
enriched secondary alcohols353–358 and diols,359,360 b-chi- the solution conformations of the respective optically
ral alcohols,361,362 a-chiral amines,363–367 mixed chiral active diastereomers that are produced must be known in
amines and alcohols,368,369 chiral carboxylic acids,370–372 order to assign the configuration of the asymmetric center
and chiral 3-hydroxy b-lactams.373 These reagents have correctly. For a variety of reasons, derivatization may lead
also found combined utility in enantiomeric separation, to decomposition or a mixture, or may not occur at all. Pu-
including LC-NMR, plus determination of absolute stereo- rification may be difficult, or low yielding. Further,
chemistry of various types of compounds such as diaster- depending on the molecule, the derivatives may not adopt
eomeric alcohols374–377 and 2-hydroxypyrrolizidinones.378 the expected conformations based on literature precedent,
In our labs, MPTA, MPA, boc-phenylglycine, and ethyl- which, of course, may lead to the incorrect configurational
2-(9-anthryl)-2-hydroxyacetate have been used on various assignment. Approaches to checking solution conforma-
occasions to derivatize compounds for determination of tion may include performing NMR nOe (or 2D NOESY)
absolute stereochemistry. The reverse-Mosher’s approach and possibly additional 2D-NMR experiments to make the
using a-trifluoromethylbenzyl alcohol has also been used. necessary NMR assignments, as well as conducting geom-
Research has also been performed to find new and etry optimization using perhaps semiempirical or ab initio
improved reagents. A noteworthy development is the use calculations.
of dichlorodimethylsilane derivatized with ()-(R)- and A variety of reagents are available for NMR determina-
(+)-(S)-a-(trifluoromethyl)benzyl alcohol that is deemed tion of the percentage of enantiomeric enrichment (%ee) as
superior in many respects to other reagents specifically well as absolute stereochemistry,379,380 and chiral solvents
for the determination of absolute stereochemistry of sec- can be used with particular types of compounds for the
ondary alcohols (Fig. 10).353 This silane derivative can be determination of absolute stereochemistry by NMR.381–384
Chirality DOI 10.1002/chir
BUILDING CHIRAL TECHNOLOGY TOOLBOXES 669

Fig. 10. Synthesis and generalized structure of (+)-(S)-a-(trifluoromethyl)benzyl alcohol dimethylsilane derivatives used in new modified Mosher’s
or Trost NMR approach. (TEA ¼ triethylamine; TBAF ¼ tetrabutylammonium fluoride.)

CURRENT TOOLBOX—CHIRAL LIQUID CRYSTAL lar couplings (RDCs) was initiated by Lindon and Ems-
NMR METHOD ley.385 More recently, NMR spectroscopy in chiral poly-
peptide liquid crystal solvents has been used as a tool for
An NMR approach that is proving to be highly effective observing and analyzing enantiomeric excess in chiral
for absolute stereochemistry determination at our com- compounds.386–393 This method differentiates enantiomers
pany and that complements well calculated versus through their quadrupole and RDC constants.388 With
observed VCD is the use of chiral liquid crystals as an advances in NMR technology, experiments have been spe-
NMR solvent. Configurational and conformational analysis cifically generated for use in chiral polypeptide liquid crys-
of organic molecules in liquid crystals using residual dipo- tal solvents394 and the methodology has evolved to where

Fig. 11. A minimum of five independent RDCs from the stereocenter must be measured for the orientation of the molecule to be determined. Once
this is achieved, stereochemical information can be obtained. In this figure, the RDCs that can be measured from the chiral carbon of ibuprofen are
shown. The solid arrows indicate possible carbon–carbon RDCs, while dashed arrows indicate possible carbon–proton RDCs (a). The 1H NMR spectra
of (R)-ibuprofen in PBLG/CDCl3 alignment media (b), of (S)-ibuprofen in PBLG/CDCl3 alignment media (c) and racemic-ibuprofen in standard CDCl3
(d). The expansions in (b) and (c) show the proton–proton total coupling (T) between the methyl protons (14) and the methine proton (10) (small cou-
pling). The larger coupling arises from the methyl (14) to methyl (14) proton–proton interaction.
Chirality DOI 10.1002/chir
670 MCCONNELL ET AL.

Fig. 12. The RDCs were calculated from the lowest energy conformers produced from a conformational search conducted in a PBLG/CDCl3 simu-
lated environment of (R) and (S)-ibuprofen. The experimental RDCs are in good agreement with the calculated RDCs as indicated by the linear curve fit
of Dcalc ¼ Dobs + 0.3 Hz with a Pearson’s correlation factor of R2 ¼ 0.991 for the (R) enantiomer (a) and Dcalc ¼ 0.92Dobs + 0.43 Hz with R2 ¼ 0.992 for
the (S) enantiomer (b). When the configuration of the ibuprofen model structures was reversed from (R) to (S) and (S) to (R), the Dcalc to Dobs linear fit
was substantially lowered. This produced a correlation factor R2 ¼ 0.742 and Dcalc ¼ 0.58Dobs +1.7 Hz for the (R) enantiomer (c) and a R2 ¼ 0.783 with a
Dcalc ¼ 0.71Dobs +1.35 Hz for the (S) enantiomer (d).

RDCs measured in such alignment media have been used racemic-ibuprofen can be seen in Figure 11b–11d. The
to determine the relative and absolute stereochemistry of large difference in these spectra illustrates the contribu-
small molecules.395–397 A major advantage of using chiral tion from RDCs that are averaged and not observed under
polypeptide liquid crystal solvents as an alignment media normal (isotropic) NMR conditions. Using the experimen-
is that both the organic molecule of interest and polypep- tally observed RDCs along with RDCs calculated from the
tide are readily soluble in common low viscosity organic lowest energy conformers of (R)- and (S)-ibuprofen, it is
solvents.388 A method has been developed that uses the possible to distinguish between the two enantiomers
orientation media poly-g-benzyl-L-glutamate (PBLG) dis- (Figs. 12a and 12b). When the stereochemistry of these
solved in CDCl3 to discriminate the (R) and (S)-enantiom- conformers was changed from (R) to (S) and from (S) to
ers and to measure RDCs of small chiral and or racemic (R) the agreement between calculated and experimental
organic molecules. The RDCs of the enantiomers are then RDCs was substantially reduced allowing for stereochemi-
used, along with molecular modeling methods, to identify cal identification (Figs. 12c and 12d). With these results,
each enantiomer. As an example, this method has been we have shown that it is possible to use RDCs to discrimi-
applied to the (R) and (S) enantiomers of ibuprofen, whose nate between (R) and (S) enantiomers when the structural
structures have been well established by both NMR and geometry is fixed (as the enantiomers of ibuprofen adopt
X-ray crystallography.398 Only 15 mg of each enantiomer preferred conformations in the PBLG system).398 Experi-
of ibuprofen was required. With these amounts, most of mental RDCs obtained from both enantiomers are neces-
the NMR experimental acquisition times are less than sary for proper comparison to calculated RDCs. This
10 h long.399 The first step to use RDCs to obtain stereo- method is also effective for racemic mixtures of small or-
chemical assignments is to determine the orientation of ganic molecules. This method is generally useful for low
the molecule.397,400 As seen in Figure 11a, this is achieved molecular weight molecules that possess one or two chiral
by measuring five independent RDCs for each stereocen- centers that are soluble in low viscosity organic solvents
ter present in the molecule.397 The differences between and will not readily crystallize or cannot easily be derivat-
the enantiomers of ibuprofen present in PBLG/CDCl3 and ized with a Mosher’s or Trost reagent.
Chirality DOI 10.1002/chir
TABLE 2. Comparison of methods for determination of absolute stereochemistry

Method

CD (exciton chirality,
NMR (Mosher’s/Trost VCD (measured versus porphyrin tweezers, and OR (measured vs.
X-ray and chiral liquid crystal) calculated) measured vs. calculated) calculated)

Sample Type Single crystal (mg) Oil/solid (1 mg for Oil/solid (5–10 mg) Oil/solid (1 mg) Oil/solid (1–3 mg)
(typical quantity) Mosher’s or Trost
method; 15–50 mg for
chiral liquid crystal)
Derivatization Presence of heavy atom Yes for Mosher’s or Trost No, however one may No for calculation No
or chiral derivative of method; No for chiral consider making a approach, but Yes for
known stereochemistry liquid crystal method derivative to enhance exciton or porphyrin
required for solubility in CDCl3 tweezer method, e.g.,
determination of (solubility usually not benzoates, napthoates,
absolute stereochemistry an issue in d6-dmso) amides, or esters
Degree of sample Easy to very difficult Moderate for Easy Moderate for Easy
prep difficulty derivatization and derivatization to easy
liquid crystal prep
Typical (turnaround) Couple of days for relative Days for Mosher’s Days to weeks (depends Days to week (depends Days to weeks (depends
time stereochemistry; days approach to days to on number of on time for on number of
to week(s) for absolute week(s) for liquid conformers and conformational conformers and
stereochemistry crystal approach calculation time per analysis, derivatization calculation time per
BUILDING CHIRAL TECHNOLOGY TOOLBOXES

conformer) time) conformer)


Comments Greatest level of Mosher’s or Trost Best for small, rigid Derivatives may require Best for small, rigid
confidence; however, method may require molecules; requires chromatography; use molecules – requires
refinement takes time. chrom; Use on primary calculation. No tweezers on secondary calculation. Because
Initial crystals may or secondary OH, chromatography OH or primary amine; accuracy of calculation
require further amine, or acid; need to required assuming use modeling and is approx 20–308, need
purification and examine changes in pure compound to possibly NMR analysis relatively high rotation.
recrystallization chemical shifts; may begin with for ambiguous cases No chromatography
need modeling and nOe’s required

Chirality DOI 10.1002/chir


671
672 MCCONNELL ET AL.

SUMMARY AND CONCLUSIONS LITERATURE CITED


1. Grayson I, Drauz K. New applications of chiral technologies in the
Clearly, a wide variety of tools are available to affect sep- fine chemicals industry. Chim Oggi 2002;20:15–19.
aration of racemates into their enantiomers and to deter- 2. Vaidya N. Diastereomeric crystallization—the ‘‘classical’’ chiral tech-
mine the absolute stereochemistry of small molecules in nology. Innovat Pharma Technol 2001;1:82–85.
drug discovery. Each has its own advantages and disad- 3. Hollingsworth R. Carbohydrate-based chiral technology: challenging
vantages, and no universal solutions exist. In many cases, the petrochemistry paradigm. Chem Eng 2001;108:66–99.
the toolbox contains tools that are familiar and useful for 4. Blacker J. Recent developments in chiral technology. Innovat
the respective analysis. Often comfort and familiarity with Pharma Technol 2001;1:77–81.
the existing tools, and the pressure of deadlines in the 5. Hollingsworth R, Wang G. Optically-active three-carbon synthons:
drug discovery process minimizes the opportunity to Another milestone in the creation of a general carbohydrate chiral
technology platform. Chim Oggi 2000;18:40–42.
explore new methodology leading to the addition of valua-
6. McCague R. A chiral technology service to match outsourcing
ble new tools to the toolbox. At our company, an trends, Part 1. Speciality Chem 1998;18:402–403.
expressed effort has been made to build up our Chiral 7. McCague R. A chiral technology service to match outsourcing
Technology toolbox, and, where time and resources per- trends, Part 2. Speciality Chem 1998;18:440–441.
mit, to look for opportunities to refine current techniques 8. Hirohara H. Chiral technology using hydrolytic enzymes: feasibility
and pursue new techniques. and mechanism of enzyme action. Bio Industry 1999;16:27–33.
With respect to chiral separations, reliance on analytical 9. Richards A, McCague R. The impact of chiral technology on the
and preparative HPLC and SFC is especially strong. Valua- pharmaceutical industry. Chem Ind 1997;11:422–425.
ble analytical support is provided for %ee determination 10. Dyer R. Splitting it 50:50—racemization as a chiral technology.
Speciality Chem 1997;17:50–54.
during chiral salt resolution, asymmetric synthesis, and
11. Fadnavis N. Developments in chiral technology, Part II. Chem Ind
asymmetric transformations using enzymes. Preparative
Dig 1995;8:79–84.
chiral separations are carried out primarily using SFC, and
12. Divakar K, Ekkundi V. Developments in chiral technology, Part I.
the role of SFC continues to grow in the area of prepara- Chem Ind Dig 1995;8:65–77.
tive achiral separations as well. 13. Sheldon R. Chirotechnology. New York: Marcel Dekker; 1993. 423 pp.
With respect to the toolbox containing techniques to 14. Joseph-McCarthy D, Thomas IVB, Belmarsh M, Moustakas D,
determine absolute stereochemistry, the chiral liquid crys- Alvarez J. Pharmacophore-based molecular docking to account for
tal NMR approach and VCD are being employed much ligand flexibility. Protein Struct Funct Genet 2003;51:172–188.
more frequently, and complement well the techniques of 15. Joseph-McCarthy D, Alvarez J. Automated generation of MCSS-
X-ray diffraction analysis using a heavy atom, circular derived pharmacophoric site points for search multiconformation
databases. Protein Struct Funct Genet 2003;51:189–202.
dichroism with exciton coupling, and NMR analysis using
16. Jennings A, Tennant M. Discovery strategies in a biopharmaceutical
the modified Mosher’s and Trost approach. In the future, startup: maximizing your chances of success using computational fil-
it is anticipated that determination of absolute stereochem- ters. Curr Pharma Des 2005;11:335–344.
istry by computational means using electronic circular 17. Baringhaus K-H, Hessler G. Fast similarity searching and screening
dichroism and optical rotation will become commonplace. hit analysis. Drug Discov Today 2004;1:197–202.
From our perspective, key attributes of each technique are 18. Singh R, Ethayathulla A, Jabeen T, Sharma S, Kaur P, Singh T. Aspi-
found in Table 2. rin induces its anti-inflammatory effects through its specific binding
In conclusion, because the discovery and development to phospholipase A2: crystal structure of th ecomplex formed
between phospholipase A2 and aspirin at 1.9 angstrom resolution.
of single-enantiomer drugs are only expected to increase J Drug Target 2005;13:113–119.
in the coming years, it is anticipated that the building of 19. Dahl S, Sylte I. Molecular modelling of drug targets: the past, the
our Chiral Technology toolbox with the addition of new present and the future. Basic Clin Pharmacol Toxicol 2005;96:151–
tools and refinement of existing tools will be an ongoing 155.
activity for many years. Further, as in any company or aca- 20. Douguet D, Munier-Lehmann H, Labesse G, Pochet S. LEA3D: a
demic lab, resources are not without limit, so there is computer-aided ligand design for structure-based drug design.
heavy reliance on and strong encouragement for the suc- J Med Chem 2005;48:2457–2468.
cessful refinement of existing Chiral Technology tools and 21. Marrero-Ponce Y, Castillo-Garit JA, Olazabal E, Serrano HS, Morales
the discovery, design, and development of new tools A, Castanedo N, Ibarra-Velarde F, Huesca-Guillen A, Jorge E, del
Valle A, Torrens F, Castro EA. Tomocomd-Cardd, a novel approach
through academic labs and commercial organizations. for computer-aided ‘‘rational’’ drug design, Part I: theoretical and ex-
perimental assessment of a promising method for computational
ACKNOWLEDGMENTS screening and in silico design of new anthelmintic compounds.
We would like to thank Dr. M. Abou-Gharbia, Head of J Comput-Aided Mol Des2004;18:615–634.
22. Marrero-Ponce Y, Castillo-Garit JA, Olazabal E, Serrano HS, Morales
our Wyeth Research Chemical and Screening Sciences
A, Castanedo N, Ibarra-Velarde F, Huesca-Guillen A, Sanchez AM,
Division, for his continued support of our efforts, and Torrens F, Castro EA. Atom, atom-type and total molecular linear
acknowledge the continued support, input, and feedback indices as a promising approach for bioorganic and medicinal chem-
from our colleagues Dr. T. Berger (AccelaPure), Prof. N. istry: theoretical and experimental assessment of a novel method for
Berova (Columbia University), Prof. K. Nakanishi (Colum- virtual screening and rational design of new lead anthelmintic. Bio-
org Med Chem 2005;13:1005–20.
bia University), Dr. R. Dukor (BioTools, Inc.), Prof. T.
23. Keenan SM, Geyer JA, Welsh WJ, Prigge ST, Waters NC. Rational
Freedman (Syracuse University), Prof. L. Nafie (Syracuse inhibitor design and iterative screening in the identification of selec-
University), and Prof. P. Polavarapu (Vanderbilt Univer- tive plasmodial cyclin dependent kinase inhibitors. Comb Chem
sity). High Throughput Screen 2005;8:27–38.
Chirality DOI 10.1002/chir
BUILDING CHIRAL TECHNOLOGY TOOLBOXES 673

24. Lima LM, Barreiro EJ. Bioisosterism: a useful strategy for molecular 48. Brazeau DA. Combining genome-wide and targeted gene expression
modification and drug design. Curr Med Chem 2005;12:23–49. profiling in drug discovery: microarrays and real-time PCR. Drug
25. Pratt DJ, Endicott JA, Noble ME. The role of structure in kinase-tar- Discov Today 2004;9:838–845.
geted inhibitor design. Curr Opin Drug Discov Devel 2004;7:428–436. 49. Wong ML, Licinio J. From monoamines to genomic targets: a para-
26. Sims PA, Wong CF, McCammon JA. Charge optimization of the digm shift for drug discovery in depression. Nat Rev Drug Discov
interface between protein kinases and their ligands. J Comput Chem 2004;3:136–151.
2004;25:1416–1429. 50. Kley N, Ivanov I, Meier-Ewert S. Genomics and proteomics tools for
27. Geney R, Sun L, Pera P, Bernacki R, Xia X, Horowitz S, Simmerling compound mode-of-action studies in drug discovery. Pharmacoge-
C, Ojima I. Use of the tubulin bound paclitaxel conformation for nomics 2004;5:395–404.
structure-based rational drug. Chem Biol 2005;12:339–348. 51. Wulfkuhle J, Espina V, Liotta L, Petricoin E. Genomic and proteomic
28. Gonzalez HD, Bastida I, Castanedo N, Nasco O, Olazabal E, Morales technologies for individualisation and improvement of cancer treat-
A, Serrano H, Armas RRd. Simple stochastic fingerprints towards ment. Eur J Cancer 2004;40:2623–2632.
mathematical modelling in biology and medicine, Part 1: the treat- 52. Chen WG, White FM. Proteomic analysis of cellular signaling.
ment of coccidiosis. Bull Math Biol 2004;66:1285–1311. Expert Rev Proteomics 2004;1:343–354.
29. Rajappa M, Sharma A, Saxena A. Bioinformatics and its implications 53. Walgren JL, Thompson DC. Application of proteomic technologies
in clinical medicine: a review. Int Med J 2004;11:125–129. in the drug development process. Toxicol Lett 2004;149(1–3):377–
30. Hajduk PJ, Mendoza R, Petros AM, Huth JR, Bures M, Fesik SW, 385.
Martin YC. Ligand binding to domain-3 of human serum albumin: a 54. Gottfries J, Sjogren M, Holmberg B, Rosengren L, Davidsson P,
chemometric analysis. J Comput-Aided Mol Des 2003;17(2–4): Blennow K. Proteomics for drug target discovery. Chemom Intell
93–102. Lab Syst 2004;73:47–53.
31. van de Waterbeemd H. Chemometric methods used in drug discov- 55. Toledo-Sherman LM, DeSouza L, Hosfield CM, Liao L, Boutilier K,
ery. In: van de Waterbeemd H, editor. Structure–property correla- Taylor P, Climie S, McBroom-Cerajewski L, Moran MF. New targets
tions in drug research. London: Academic Press; 1996. p 55–80. for an old drug: a chemical proteomics approach to unraveling the
32. Manganaro V, Paratore S, Alessi E, Coffa S, Cavallaro S. Adding molecular mechanism of action of methotrexate. Clin Proteomics
semantics to gene expression profiles: new tools for drug discovery. 2004;1:45–67.
Curr Med Chem 2005;12:1149–1160. 56. Stoughton RB, Friend SH. How molecular profiling could revolution-
33. Buehler LK. Is genomics advancing drug discovery? Pharma Discov ize drug discovery. Nat Rev Drug Discov 2005;4:345–350.
2005;5:28–29. 57. Klebl BM, Daub H, Keri G. Chemical kinomics. Methods Principles
34. Orth AP, Batalov S, Perrone M, Chanda SK. The promise of Med Chem 2004;22:167–190.
genomics to identify novel therapeutic targets. Expert Opin Ther 58. Haar E, Walters WP, Pazhanisamy S, Talsimi P, Pierce AC, Bemis
Targets 2004;8:587–596. GW, Salituro FG, Harbeson SL. Kinase chemogenomics: targeting
35. Lavedan C, Birznieks G, Dressman M, McCullough K, Paczkowski the human kinome for target validation and drug discovery. Mini
R, Torres R, Wolfgang C, Polymeropoulos M. Translating the ge- Rev Med Chem 2004;4:235–253.
nome into individualized therapeutics. Drug Dev Res 2004;62:371– 59. Baringhaus K-H, Hessler G. A chemical genomics approach for ion
382. channel modulators. Methods Principles Med Chem 2004;22:221–
36. Lui DT. Expression genomics and cancer drug development. Drug 242.
Dev Res 2004;62:295–302. 60. Darvas F, Dorman G, Krajcsi P, Puskas LG, Kovari Z, Lorincz Z,
37. Shastry BS. Role of SNP/haplotype map in gene discovery and drug Urge L. Recent advances in chemical genomics. Curr Med Chem
development: an overview. Drug Dev Res 2004;62:143–150. 2004;11:3119–3145.
38. Janzen W, Bernasconi P, Cheatham L, Mansky P, Popa-Burke I, Wil- 61. Brown ED, Wright GD. New targets and screening approaches in
liams KP, Worley J, Hodge N. Optimizing the chemical genomics antimicrobial drug discovery. Chem Rev 2005;105:759–774.
process. In: Darvas F, Guttman A, Dorman G, editors. Chemical 62. Ryan TP, Watson DE, Berridge BR. Toxicology biomarkers in drug
genomics: Advances in drug discovery and functional genomics development. Delivering on the genomic promise. Pharma Discov
applications. New York: Marcel Dekker; 2004. p 59–100. 2004;4:22–28.
39. Chin K-V, Selevanayagam ZE, Vittal R, Yang CS, Kitta T, Kudoh K, 63. Triggle DJ. Drug development in the 21st century: Medicines, man
Shih WC, Yong L, Wong YF, Cheung TH, Chung TKH. Appplication and receptors. Med Chem Res 2004;13(3/4):238–248.
of expression genomics in drug development and genomic medi- 64. Comess KM, Schurdak ME. Affinity-based screening techniques for
cine. Drug Dev Res 2004;62:124–133. enhancing lead discovery. Curr Opin Drug Discov Dev 2004;7:411–
40. Lundstrom K. Structural genomics of GPCRs. Trends Biotechnol 416.
2005;23:103–108. 65. Wheeler TT, Hood KA. The mammalian innate immune system:
41. Sanchez-Martin M, Gonzalez-Herrero I, Sanchez-Garcia I. Cancer potential targets for drug development. Curr Drug Targets Immune
genetics and drug discovery in mice. Lett Drug Des Discov Endocr Metabol Disord 2005;5:237–247.
2004;1:353–360. 66. Penny MA, McHale D. Pharmacogenomics and the drug discovery
42. Auerbach D, Arnoldo A, Bogdan B, Fetchko M, Stagljar I. Drug dis- pipeline: when should it be implemented? Am J Pharmacogenomics
covery using yeast as a model system: a functional genomic and pro- 2005;5:53–62.
teomic view. Curr Proteomics 2005;2:1–13. 67. Harrigan GG, Brackett DJ, Boros LG. Medicinal chemistry, meta-
43. Bilello JA. The agony and ecstasy of ‘‘OMIC’’ technologies in drug bolic profiling and drug target discovery: a role for metabolic profil-
development. Curr Mol Med 2005;5:39–52. ing in reverse pharmacology and chemical genetics. Mini Rev Med
44. Kliger Y, Levanon EY, Gerber D. From genome to antivirals: SARS Chem 2005;5:13–20.
as a test tube. Drug Discov Today 2005;10:345–352. 68. Griffin JL. The potential of metabonomics in drug safety and toxicol-
45. Brunden K, Sherf B, Harrington J. The application of RAGE and ogy. Drug Discov Today Technol 2004;1:285–293.
GECKO in cell-based target discovery screens. Pharmacogenomics 69. Khandelwal P, Beyer CE, Lin Q, McGonigle P, Schechter LE, Bach
2005;6:383–392. AC II. Nanoprobe NMR spectroscopy and in vivo microdialysis:
46. Kramer R, Cohen D. Functional genomics to new drug targets. Nat new analytical methods to study brain neurochemistry. J Neurosci
Rev Drug Discov 2004;3:965–972. Methods 2004;133(1/2):181–189.
47. Zehender H, Le Goff F, Lehmann N, Filipuzzi I, Mayr LM. 70. Khandelwal P, Beyer CE, Lin Q, Schechter LE, Bach AC II. Study-
SpeedScreen: the ‘‘missing link’’ between genomics and lead discov- ing rat brain neurochemistry using nanoprobe NMR spectroscopy:
ery. J Biomol Screen 2004;9:498–505. A metabonomics approach. Anal Chem 2004;76:4123–4127.
Chirality DOI 10.1002/chir
674 MCCONNELL ET AL.

71. Parsons L, Orban J. Structural genomics and the metabolome: com- 91. Lee H, Sarko CR. Analysis of a combinatorial library synthesized
bining computational and NMR methods to identify target ligands. using a split-and-pool Irori MicroKan method for development and
Curr Opin Drug Discov Dev 2004;7:62–68. production. In: Kyranos JN, editor. High throughput analysis for
72. Verhoeckx KC, Bijlsma S, Jespersen S, Ramaker R, Verheij ER, Wit- early drug discovery. San Diego, CA: Elsevier; 2004. p 37–56.
kamp RF, van der Greef J, Rodenburg RJ. Characterization of anti- 92. Kyranos JN, Lee H, Goetzinger WK, Li LYT. One-minute full-gradi-
inflammatory compounds using transcriptomics, proteomics, and ent HPLC/UV/ELSD/MS analysis to support high-throughput paral-
metabolomics in combination with multivariate data analysis. Int lel synthesis. J Combinat Chem 2004;6:796–804.
Immunopharmacol 2004;4:1499–514. 93. Guttman A, Varoglu M, Khandurina J. Multidimensional separations
73. Goodacre R, Vaidyanathan S, Dunn WB, Harrigan GG, Kell DB. in the pharmaceutical arena. Drug Discov Today 2004;9:136–144.
Metabolomics by numbers: acquiring and understanding global 94. Chen JM, Jin G, Sung A, Levin JL. X-ray crystallography combined
metabolite data. Trends Biotechnol 2004;22:245–252. with parallel synthesis. Bioorg Med Chem Lett 2002;12:1195–1199.
74. Keun HC, Ebbels TM, Bollard ME, Beckonert O, Antti H, Holmes 95. Natt O, Frahm J. In vivo magnetic resonance imaging: insights into
E, Lindon JC, Nicholson JK. Geometric trajectory analysis of meta- structure and function of the central nervous system. Meas Sci
bolic responses to toxicity can define treatment specific profiles. Technol 2005;16:R17–R36.
Chem Res Toxicol 2004;17:579–587.
96. Villar HO, Yan J, Hansen MR. Using NMR for ligand discovery and
75. Bailey WJ, Ulrich R. Molecular profiling approaches for identifying optimization. Curr Opin Chem Biol 2004;8:387–391.
novel biomarkers. Expert Opin Drug Saf 2004;3:137–151.
97. Hajduk PJ, Huth JR, Fesik SW. Druggability indices for protein tar-
76. Burczynski ME, Oestreicher JL, Cahilly MJ, Mounts DP, Whitley gets derived from NMR-based screening data. J Med Chem 2005;48:
MZ, Speicher LA, Trepicchio WL. Clinical pharmacogenomics and 2518–2525.
transcriptional profiling in early phase oncology clinical trials. Curr
98. Huth JR, Yu L, Collins I, Mack J, Mendoza R, Isaac B, Braddock
Mol Med 2005;5:83–102.
DT, Muchmore SW, Comess KM, Fesik SW, Clore GM, Levens D,
77. Dougherty T, Miller P. Microbial genomics and drug discovery: Hajduk PJ. NMR-driven discoery of benzoylanthranilic acid inhibi-
exploring innovative routes of drug discovery in the postgenomic tors of far upstream element binding protein binding to the human
ear. IDrugs 2006;9:420–422. oncogene c-myc promoter. J Med Chem 2004;47:4851–4857.
78. Weller HN, Rubin AE, Moshiri B, Ruediger W, Li W-J, Allen J, Nolfo 99. Zhang J, McCombie G, Guenat C, Knochenmuss R. FT-ICR mass
J, Bertok A, Rosso VW. Development and commercialization of the spectrometry in the drug discovery process. Drug Discov Today
MiniBlock synthesizer family: a historical case study. JALA 2005;10: 2005;10(9):635–642.
59–71.
100. Siegel MM. Mass spectrometry based assays for drug screening in
79. Fokas D, Yu L, Baldino CM. Strategies for the synthesis of novel the early phases of drug discovery. Frontiers Med Chem 2004;
indole alkaloid-based screening libraries for drug discovery. Mol 1:273–292.
Divers 2005;9(1–3):81–89.
101. Truebenbach CS, Tong H, Huang N, Schnier PD, Siegel MM. High
80. Dallinger D, Kappe CO. Creating chemical diversity space by scaf- throughput flow injection analysis-mass spectrometry for combinato-
fold decoration of dihydropyrimidines. Pura Appl Chem 2005; 77: rial chemistry using electrospray ionization, atmospheric pressure
155–161. chemical ionization and exact-mass Fourier transform mass spec-
81. Weber A, von Roedern E, Stilz HU. SynCar: an approach to auto- trometry. In: Kyranos JN, editor. High throughput analysis for early
mated synthesis. J Comb Chem 2005;7:178–184. drug discovery. San Diego, CA: Elsevier; 2004. p 73–93.
82. Musonda CC, Chibale K. Application of combinatorial and parallel 102. Tabei K, Feng X, Vankatesan A, Abe T, Hideki U, Mansour TS,
synthesis chemistry methodologies to antiparasitic drug discovery. Siegel MM. Mechanism of inactivation of b-lactamases by novel 6-
Curr Med Chem 2004;11:2519–2533. methylidene penems elucidated using electrospray ionization mass
83. Laird ER, Blake JF. Structure-based generation of viable leads from spectrometry. J Med Chem 2004;47:3674–3688.
small combinatorial libraries. Curr Opin Drug Discov Dev 2004; 103. Kruppa G, Schnier PD, Tabei K, Van Orden S, Siegel MM. Multiple
7:354–359. ion isolation applications in FT-ICR MS: exact-mass MSn internal
84. Metcalf IIICA, Sawyer T. Src homology-2 domains and structure- calibration and purification/interrogation of protein-drug complexes.
based, small-molecule library approaches to drug discovery. In: Anal Chem 2002;74:3877–3886.
Makriyannis A, Biegel D, editors. Drug discovery strategies and 104. Braunschweig T, Chung J-Y, Hewitt SM. Perspectives in tissue
methods. New York: Marcel Dekker; 2004. p 23–59. microarrays. Comb Chem High Throughput Screen 2004;7:575–585.
85. Lee H. Pharmaceutical applications of liquid chromatography 105. Kleymann G, Werling H-O. A generally applicable, high-throughput
coupled with mass spectrometry (LC/MS). J Liquid Chromatogr screening-compatible assay to identify, evaluate, and optimize anti-
Relat Technol 2005;28(7/8):1161–1202. microbial agents for drug therapy. J Biomol Screen 2004;9:578–587.
86. Paulson JL. A stand-alone monitoring system for reliable unattended 106. Zhang H-Z, Kasibhatla S, Wang Y, Herich J, Guastella J, Tseng B,
operation of a commercial preparative LC/MS purification system. Drewe J, Cai SX. Discovery, characterization and SAR of gambogic
JALA 2005;10:6–15. acid as a potent apoptosis inducer by a HTS assay. Bioorg Med
87. Ventura M, Farrell W, Aurigemma C, Tivel K, Greig M, Wheatley J, Chem 2004;12:309–317.
Yanovsky A, Milgram KE, Dalesandro D, DeGuzman R, Tran P,
107. Yadavalli VK, Pishko MV. Biosensing in microfluidic channels using
Nguyen L, Chung L, Gron O, Koch CA. High-throughput preparative
fluorescence polarization. Anal Chim Acta 2004;501:123–128.
process utilizing three complementary chromatographic purification
technologies. J Chromatogr A 2004;1036:7–13. 108. Singh J, Chuaqui CE, Boriack-Sjodin PA, Lee W-C, Pontz T, Corbley
MJ, Cheung H-K, Arduini RM, Mead JN, Newman MN, Papadatos
88. Bolanos B, Greig M, Ventura M, Farrell W, Aurigemma CM, Li H,
Quenzer TL, Tivel K, Bylund JMR, Tran P, Pham C, Phillipson D. JL, Bowes S, Josiah S, Ling LE. Successful shape-based virtual
SFC/MS in drug discovery at Pfizer, La Jolla. Int J Mass Spectrom screening: the discovery of a potent inhibitor of the type I TGF re-
2003;238:85–97. ceptor kinase (T RI). Bioorg Med Chem Lett 2003;13:4355–4359.
89. Davis PW, Griffith MC. High throughput parallel LC/MS/ELSD of 109. Entzeroth M. Emerging trends in high-throughput screening. Curr
combinatorial libraries using the eight-channel LCT system with Opin Pharmacol 2003;3:522–529.
MUX technology. In: Kyranos JN, editor. High throughput analysis 110. Netzer R, Bischoff U, Ebneth A. HTS techniques to investigate the
for early drug discovery. San Diego, CA: Elsevier; 2004. p 113–123. potential effects of compounds on cardiac ion channels at early-
90. Delaney J, Julien LV, Kyranos JN, Salvatore C. Purity and quantity stages of drug discovery. Curr Opin Drug Discov Dev 2003;6:462–
determination of parallel synthesis compound libraries. In: Kyranos 469.
JN, editor. High throughput analysis for early drug discovery. San 111. Vaschetto M, Weissbrod T, Bodle D, Guener O. Enabling high-
Diego, CA: Elsevier; 2004. p 95–112. throughput discovery. Curr Opin Drug Discov Dev 2003;6:377–383.
Chirality DOI 10.1002/chir
BUILDING CHIRAL TECHNOLOGY TOOLBOXES 675

112. Kimple R, Jones M, Shutes A, Yerxa B, Siderovski D, Willard F. 134. Baringhaus K-H, Matter H. Efficient strategies for lead optimization by
Established and emerging fluroescence-based assays for G-protein simultaneously addressing affinity, selectivity and pharmacokinetic pa-
function: heterotrimeric G-protein alpha subunits and regulator of G- rameters. In: Manhold R, Kubinyi H, Folkers G, editors. Methods and
protein signaling (RGS) proteins. Combinat Chem High Throughput principles in medicinal chemistry, Vol. 23 (Chemoinformatics in drug
Screen 2003;6:399–407. discovery). Frankfurt: Wiley-VCH Verlag; 2005. p 1432–1436.
113. Asmild M, Oswald N, Kryzwkowski K, Friis S, Jacobsen R, Reuter 135. Takeda Y, Uoto K, Iwahana M, Jimbo T, Nagata M, Atsumi R, Onno
D, boryski RT, Kutchinsky J, Vestergaard R, Schroder R, Sørensen C, Tanaka N, Terasawa H, Soga T. New highly active taxoids from
CB, Bech M, Korsgaard MPG, Willumsen NJ. Upscaling and auto- 9b-dihydrobacctin-9.10-acetals, Part 5. Bioorg Med Chem Lett
mation of electrophysiology: toward high throughput screening in 2004;14:3209–3215.
ion channel drug discovery. Receptors Channels 2003;9:49–58. 136. Pinto D, Orwat M, Wang S, Fevig J, Quan M, Amparo E, Cacciola J,
114. Rychetsky P. Setting up a high-throughput screening operation for Rossi K, Alexander R, Smallwood A, Luettgen JM, Liang L, Aungst
combichem libraries doesn’t have to be a resource drain. Pharma- BJ, Wright MR, Knabb RM, Wong PC, Wexler RR, Lam PYS. Dis-
Chem 2002;1:2–5. covery of 1-[3-(aminomethyl)phenyl]-N-3-fluoro-20 -(methylsulfonyl)-
115. Kerns E, Di L. Automation in pharmaceutical profiling. JALA 2005; [1,10 -biphenyl]-4-yl]-3-(trifluoromethyl)-1H-pyrazole-5-carboxamide
10:114–123. (DPC423), a highly potent, selective, and orally bioavailable inhibi-
116. Jenkins K, Angeles R, Quintos M, Xu R, Kassel D, Rourick R. Auto- tor of blood coagulation factor Xa. J Med Chem 2001;44:566–578.
mated high throughput ADME assays for metabolic stability and 137. Fichert T, Yazdanian M, Proudfoot J. A structure–permeability study
cytochrome P450 inhibition profiling of combinatorial libraries. J of small drug-like molecules. Bioorg Med Chem Lett 2003;13:719–722.
Pharma Biomed Anal 2004;34:989–1004. 138. MacKenzie A, Marchington A, Middleton D, Newman S, Jones B.
117. Hersey A, Luscombe C, Modi S. In silico transporter modeling and Structure-activity relationships of 1-alkyl-5-(3,4-dichlorophenyl)-5-2-
its application in drug discovery. Biotechnol Pharma Aspects 2004; piperidones, Part 1: Selective antagonists of the neurokinin-2 recep-
1:53–68. tor. J Med Chem 2002;45:5365.
118. Bemporad D, Luttmann C, Essex J. Computer simulation of small 139. Hay M, Garnage S, Kovacs M, Pruijin F, Anderson R, Patterson A,
molecule permeation across a lipid bilayer: dependence on bilayer Wilson W, Brown J, Denny W. Structure–activity relationships of
properties and solute volume, size, and cross-sectional area. Biophys 1,2,4-benzotriazine 1,4-dioxides as hypoxia-selective analogues of
J 2004;87:1–13. tirapazamine J Med Chem 2003;46:169–182.
119. El-Gendy A, Adejare A. Membrane permeability related physico- 140. Peglion J-L, Goument B, Despaux N, Charlot V, Giraud H, Nisole C,
chemical properties of a novel g-secretase inhibitor. Int J Pharma Newman-Tancredi A, Dekeyne A, Bertrand M, Genissel P, Millan
2004;280(1/2):47–55. MJ. Improvement in the selectivity and metabolic stability of the se-
120. Nassar A-E, Kamel A, Clarimont C. Improving the decision-making rotonin 5-HT1A ligand, S 15535: a series of cis- and trans-2-(arycy-
process in the structural modification of drug candidates: enhancing cloalkylamine) 1-indanols. J Med Chem 2002;45:165–176.
metabolic stability. Drug Discov Today 2004;9:1020–1028. 141. Hutchinson I, Jennings S, Vishnuvajjala B, Westwell A, Stevens M.
121. Hartmann T, Schmitt J. Lipophilicity—Beyond octanol/water: a Antitumor benzothiazoles, Part 16: Synthesis and pharmaceutical
short comparison of modern technologies. Drug Discov Today properties of antitumor 2-(4-aminophenyl)benzothiazole amino acid
2004;1:431–439. prodrugs. J Med Chem 2002;45:744–747.
122. Kerns E, Di L. Automated property profiling for cellular drug deliv- 142. Liljebris C, Larsen S, Ogg D, Palazuk B, Bleasdale J. Investigation of
ery. Cell Drug Deliv 2004:345–365. potential bioisosteric replacements for the carboxyl groups of pepti-
domimetic inhibitors of protein tyrosine phosphatase 1B: identifica-
123. Di L, Kerns E. Profiling drug-like properties in discovery research.
tion of a tetrazole-containing inhibitor with cellular activity. J Med
Curr Opin Chem Biol 2003;7:402–408.
Chem 2002;45:1785–1798.
124. Kerns E. Pharmaceutical profiling in drug discovery. Drug Discov
143. Andersen H, Olsen O, Iversen L, Sorensen A, Mortensen S, Chris-
Today 2003;8:316–323.
tensen M, Branner S, Hansen T, Lau J, Jeppesen L, Moran EJ, Su J,
125. Di L, Kerns E, Fan K, McConnell O, Carter G. High throughput arti- Bakir F, Judge L, Shahbaz M, Collins T, Vo T, Newman MJ, Ripka
ficial membrane permeability assay for blood-brain barrier. Eur J WC, Moller NP. Discovery and SAR of a novel selective and orally
Med Chem 2003;38:223–232. bioavailable nonpeptide classical competitive inhibitor class of pro-
126. Didziapetris R, Japertas P, Avdeef A, Petrauskas A. Classification tein-tyrosine phosphatase 1B. J Med Chem 2002;45:4443–4459.
analysis of P-glycoprotein substrate specificity. J Drug Target 144. Madsen P, Ling A, Plewe M, Sams C, Knudsen L, Sidelmann U,
2003;11:391–406. Ynddal L, Brfand C, Andersen B, Murphy D, Teng M, Truesdale L,
127. Veber D, Johnson S, Cheng H-Y, Smith B, Ward K, Kopple K. Kiel D, May J, Kuki A, Shi S, Johnson MD, Teston KA, Feng J, Lakis
Molecular properties that influence the oral bioavailability of drug J, Anderes K, Gregor V, Lau J. Optimization of alkylidene hydrazide
candidates. J Med Chem 2002;45:2615–2623. based human glucagon receptor antagonists. Discovery of the
128. Gao F, Johnson D, Ekins S, Janiszewski J, Kelly K, Meyer R, West highly potent and orally available 3-cyano-4-hydroxybenzoic acid [1-
M. Optimizing higher throughput methods to assess drug-drug (2,3,5,6-tetramethylbenzyl)-1H-indol-4-ylmethylene]hydrazide. J Med
interactions for CYP1A2, CYP2C9, CYP2C19, CYP2D6, rCP2D6, and Chem 2002;45:5755–5775.
CYP3A4 in vitro using a single point IC50. J Biomol Screen 145. Al-awar R, Ray J, Schultz R, Andis S, Kennedy J, Moore R, Liang J,
2002;7:373–382. Golakoti T, Subbaraju G, Corbett T. A convergent approach to cryp-
129. Kerns E. High throughput physicochemical profiling for drug dis- tophycin 52 analogues: synthesis and biological evaluation of a novel
covery. J Pharm Sci 2001;90:1838–1858. series of fragment A epoxides and chlorohydrins. J Med Chem 2003;
46:2985–3007.
130. Van de Waterbeemd H, Smith D, Beaumont K, Walker D. Property-
based design: Optimization of drug absorption and pharmaco- 146. Chu-Moyer M, Ballinger W, Beebe D, Coutcher J, Day W, Li J,
kinetics. J Med Chem 2001;44:1313–1333. Oates P, Weekly R. SAR and species/stereo-selective metabolism of
the sorbitol dehydrogenase inhibitor, CP-470,711. Bioorg Med
131. Bullock P. Alternate methods for assessing absorption,
Chem Lett 2002;12:1477–1480.
metabolism, and routes of elimnation. Drugs Pharma Sci 2005;152:
303–347. 147. Fichert T, Yazdanian M, Proudfoot J. A structure-permeability study
132. Li A. Preclinical in vitro screening assays for drug-like properties. of small drug-like molecules. Bioorg Med Chem Lett 2003;13:719–722.
Drug Discov Today 2005;2(2):179–185. 148. Clark D. In silico prediction of blood-brain barrier permeation. Drug
133. Di L, Kerns E. Application of pharmaceutical profiling assays for Discov Today 2003;8:927–933.
optimization of drug-like properties. Curr Opin Drug Discov Devel 149. Singla A, Garg A, Aggarwal D. Paclitaxel and its formulations. Int J
2005;8:495–504. Pharm 2002;235:179–192.
Chirality DOI 10.1002/chir
676 MCCONNELL ET AL.

150. Regueiro-Ren A, Leavitt K, Kim S-H, Hofle G, Kiffe M, Gougoutas J, La Fargue J, Lapierre JM, Larrabee S, Li F, Papp E, McWeeney D,
DiMarco J, Lee F, Fairchild C, Long B, Vite GD. SAR and pH stabil- Ramesha C, Roberts R, Rotstein D, San Pablo B, Sjogren EB, So
ity of cyanosubstituted epothilones. Org Lett 2002;4:3815–3818. O-Y, Talamas FX, Tao W, Trejo A, Villasenor A, Welch M, Welch T,
151. Fray M, Bull D, Carr C, Gautier E, Mowbray C, Stobie A. Structure– Weller P, Whiteley PE, Young K, Zipfel S. S-[5-Amino-1-(4-fluoro-
activity relationships of 1,4-dihydro-(1H,4H)-quinoxaline-2,3-diones phenyl)-1H-pyrazol-4-yl]-[3-(2,3-dihydroxypropoxy) phenyl]methanone
as N-methyl-D-aspartate (glycine site) receptor antagonist, Part 1: (RO3201195), an orally bioavailable and highly selective inhibitor of
heterocyclic substituted 5-alkyl derivative. J Med Chem 2001;44: p38 map kinase. J Med Chem 2006;49:1562–1575.
1951–1962. 167. Stanton D, Mattioni B, Knittel J, Jurs P. Development and use of
152. Brooks G, Burgess W, Colthurst D, Hinks J, Hunt E, Pearson M, hydrophobic surface area (HSA) descriptors for computer-assisted
Shea B. Pleuromutilins, Part 1: the identification of novel mutilin 14- quantitative structure–activity and structure–property relationship
carbamates. Bioorg Med Chem 2001;9:1221–1231. studies. J Chem Inf Comput Sci 2004;44:1010–1023.
153. Goodwin JT, Conradi RA, Ho NFH, Burton PS. Physicochemical 168. Rouhi A. Chiral business. Chem Eng News 2003;81:45–55.
determinants of passive membrane permeability: role of solute 169. Rouhi A. Chiral roundup. Chem Eng News 2002;80:43–50.
hydrogen-bonding potential and volume. J Med Chem 2001;44: 170. Stinson C. Chiral pharmaceuticals. Chem Eng News 2001;79:79–97.
3721–3729. 171. Stinson C. Chiral chemistry. Chem Eng News 2001;79:45–57.
154. Ashwood A, Field MJ, Horwell DC, Julien-Larose C, Lewthwaite RA, 172. Maier N, Franco P, Lindner W. Separation of enantiomers: Needs,
McCleary S, Pritchard MC, Raphy J, Singh L. Utilization of an intra- challenges, perspectives. J Chromatogr A 2001;906:3–33.
molecular hydrogen bond to increase the CNS penetration of an
NK1 receptor antagonist. J Med Chem 2001;44:2276–2285. 173. Gerhard B. Synthesis of chiral drug substances. Synlett 2002;6:837–
850.
155. Brooks G, Burgess W, Colthurst D, Hinks JD, Hunt E, Pearson MJ,
Shea B, Takle AK, Wilson AM, Woodnutt G. Pleuromutilins. Part 1: 174. Prentis R, Lis Y, Walder S. Pharmaceutical innovation by the seven
the identification of novel mutilin 14-carbamates. J Med Chem 2001; U.K.-owned pharmaceutical companies (1964–1985). Br J Clin Phar-
9:1221–1231. macol 1998;25:387–396.
175. Agranat I, Caner H, Caldwell J. Putting chirality to work: Strategy of
156. Panchagnula R, Thomas N. Biopharmaceutics and pharmacokinetics
chiral switches. J Nat Rev Drug Discov 2002;1:753–768.
in drug research. Int J Pharm 2000;201:131–150.
176. Rouhi A. Chirality at work. Chem Eng News 2003;81:56–61.
157. Revesz L, Di Padova F, Buhl T, R F, Gram H, Hiestand P, Manning
U, Zimmerlin A. SAR of 4-hydroxypiperidine and hydroxyalkyl sub- 177. Rouhi A. Chiral chemistry. Chem Eng News 2004;82:47–62.
stituted heterocycles as novel p38 map kinase inhibitors. Bioorg 178. Cheng X-M. To market, to market—1993. Ann Rep Med Chem
Med Chem Lett 2000;10:1261–1265. 1994;29:331–354.
158. Bromidge S, Davies S, Duckworth D, Forbes I, Jones G, Jones J, 179. Hegde S, Carter J. To market, to market—2003. Ann Rep Med
King F, Blackburn T, Holland V, Kennett GA, Lightowler S, Middle- Chem 2003;39:337–368.
miss DN, Riley GJ, Trail B, Wood MD. 1-[2[(Heteroarylmethoxy)ar- 180. Hegde S, Schmidt M. To market, to market—2004. Ann Rep Med
yl]carbamoyl]indolines are selective and orally active 5-HT2C recep- Chem 2005;40:443–473.
tor inverse agonists. Bioorg Med Chem Lett 2000;10: 1867–1870.
181. Thayer A. Trial separations. Supercritical fluid chromatography
159. Milbank J, Tercel M, Atwell G, Wilson W, Hogg A, Denny W. Syn- gains favor in preparative-scale separations of enantiomers. Chem
thesis of 1-substituted 3-(chloromethyl)-6-aminoindoline(6-amino- Eng News 2005;83:49–53.
seco-CI) DNA minor groove alkylating agents and structure–activity
182. Tucker G. Chiral switches. Lancet 2000;355:1085–1087.
relationships for their cytotoxicity. J Med Chem 1999;42:649–658.
183. Agranat I, Caner H. Intellectual property and chirality of drugs.
160. Krise J, Zygmunt J, Georg G, Stella V. Novel prodrug approach for
Drug Discov Today 1999;4:313–321.
tertiary amines: synthesis and preliminary evaluation of N-phospho-
nooxymethyl prodruges. J Med Chem 1999;42:3094–3100. 184. Anonymous. FDA’s policy statement for the development of new
stereoisomeric drugs. (corrections made on 1/3/97. More informa-
161. Patt WC, Cheng X-M, Repine JT, Lee C, Reisdorph BR, Massa MA, tion on regulatory issues can be found at: http://www.fda.gov/cder/
Doherty AM, Welch KM, Bryant JW, Flynn MA, Walker DM, guidance/stereo.htm.) Chirality 1992;4:338–340.
Schroeder RL, Haleen SJ, Keiser JA. Butenolide endothelin antago-
nists with improved aqueous solubility. J Med Chem 1999;42:2162– 185. Maas G. Determination of absolute and relative configuration by
2168. X-ray and neutron diffraction methods. In: Houben-Weyl, editor.
Stuttgart: Thieme Verlag; 1996. p 399–497.
162. Bulat S, Bosio S, Papadopoulos M, Cerezo-Galvez S, Grabowski E,
Rosenbaum C, Matassa V, Ott I, Metz G, Schamberger J, Sekul R, 186. Stout G, Jensen L. X-ray structure determination: a practical guide,
Feurer A. Design and discovery of novel, potent pyrazinone-based 2nd ed. New York: Wiley; 1989. 480 pp.
thrombin inhibitors with a solubilizing amino P1-P2-linker. Lett 187. Schlingmann G, Roll D. Absolute stereochemistry of unusual biopol-
Drug Des Discov 2006;3:289–292. ymers from Ascomycete culture LL-W1278: examples that deriva-
tives of (S)-6-hydroxymellein are also natural fungal metabolites.
163. Morwick T, Berry A, Brickwood J, Cardozo M, Catron K, DeTuri M,
Emeigh J, Homon C, Hrapchak M, Jacober S, Jakes S, Kaplita P, Chirality 2005;17:S48–S51.
Kelly TA, Ksiazek J, Liuzzi M, Magolda R, Mao C, Marshall D, 188. Charan R, Schlingmann G, Bernan V, Feng X, Carter G. Additional
McNeil D, Prokopowicz III A, Sarko C, Scouten E, Sledziona C, Sun pyrrolomycins from cultures of Streptomyces fumanus. J Nat Prods
S, Watrous J, Wu JP, Cywin CL. Evolution of the thienopyridine 2005;68:277–279.
class of inhibitors of IkB kinase-b, Part I: hit-to-lead strategies. 189. Berova N, Nakanishi K, Woody R. Circular dichroism: principles
J Med Chem 2006;49:2898–2908. and applications. New York: Wiley-VCH; 2000. 878 pp.
164. Barrett D, Catalano J, Deaton D, Hassell A, Long S, Miller A, Miller 190. Bari LD, Mannuccci S, Pescitelli G, Salvadori P. Assignment of abso-
L, Ray J, Samano V, Shewchuk L, Wells-Knecht KJ, Willard Jr DH, lute configuration of chiral carboxylic acids via exciton-coupled CD
Wright LL. Novel, potent P2-P3 pyrrolidine derivatives of ketoamide- treatment: 4-phenylthioproline as a case study. Chirality 2002; 14:
based cathepsin K inhibitors. Bioorg Med Chem Lett 2006;16:1735– 611–617.
1739. 191. Meguro H, Kim J-H, Bai C, Nishida Y, Ohrui H. Some applications
165. Castanedo G, Clark K, Wang S, Tsui V, Wong M, Nicholas J, Wick- of a chiral fluorometric reagent, (S)-TBMB carboxylic acid. Chirality
ramasinghe D, Marsters J, Sutherlin D. DK2/cyclinA inhibitors: tar- 2001;13:441–445.
geting the cyclinA recruitment site with small molecules derived 192. Spodine E, Zolezi S, Calvo V, Decinti A. Studies of the circular
from peptide leads. Bioorg Med Chem Lett 2006;16:1716–1720. dichroism spectra of dissymmetric Schiff-bases by means of the
166. Goldstein D, Alfredson T, Bertrand J, Browner M, Clifford K, exciton chirality method. Tetrahedron: Asymmetry 2000;11:2277–
Dalrymple S, Dunn J, Freire-Moar J, Harris S, Labadie SS, 2288.
Chirality DOI 10.1002/chir
BUILDING CHIRAL TECHNOLOGY TOOLBOXES 677

193. Hartl M, Humpf H-U. 2-Naphthol as a powerful chromophore for the 212. Schlingmann G, Matile S, Berova N, Nakanishi K, Carter G. Abso-
configurational assignment of carboxylic acid groups via the CD lute stereochemistry of the diepoxins. Tetrahedron 1996;52:435–446.
exciton chirality method. Tetrahedron: Asymmetry 2000;11:1741–1747. 213. Lo L, Berova N, Nakanishi K, Schlingmann G, Carter G, Borders D.
194. Tanaka K, Pescitelli G, Di Bari L, Xiao TL, Nakanishi K, Armstrong Determination of the absolute stereochemistry of nemadictine
D, Berova N. Absolute stereochemistry of dihydrofuroangeoicins .alpha. 2 and .alpha. J Am Chem Soc 1991;114:7371–7374.
bearing C-8 substituted double bonds: a combined chemical/exciton 214. Francotte E. Enantioselective chromatography as a powerful alterna-
chirality protocol. Org Biomol Chem 2004;2:48–58. tive for the preparation of drug enantiomers. J Chromatogr A
195. Tanaka K, Pescitelli G, Nakanishi K, Berova N. Fluorescence 2001;906:379–397.
detected exciton coupled circular dichroism: Development of new 215. Zhang Y, Wu D-R, Want-Iverson D, Tymiak A. Enantioselective chro-
fluorescent reporter groups for structural studies. Monatshefte fuer matography in drug discovery. Drug Discov Today 2005;10:571–577.
Chemie 2005;136:367–395.
216. Liu Y, Lantz A, Armstrong D. High efficiency liquid and super-sub-
196. Tanaka K, Nakanishi K, Berova N. Absolute stereochemistry of
critical fluid-based enantiomeric separations: an overview. J Liquid
allylic alcohols, amines, and other ene moieties: a microscale cross
Chromatogr Relat Technol 2004;27(7–9):1121–1178.
metathesis/exciton chirality protocol. J Am Chem Soc 2003;125:
10802–10803. 217. Welch CJ, Leonard WR Jr, DaSilva J, Biba M, Albaneze-Walker J,
Henderson D, Laing B, Mathre D. Preparative chiral SFC as a green
197. Tanaka K, Itagaki Y, Satake M, Naoki H, Yasumoto T, Nakanishi K,
Berova N. Three challenges toward the assignment of absolute con- technolog for rapid access to enantiopurity in pharmaceutical pro-
figuration of gymnocin-B. J Am Chem Soc 2005;127:9561–9570. cess research. LC-GC 2005;23:16–29.
198. Dale J, Dull D, Mosher H. a-Methoxy-a-trifluoromethylphenyl 218. Andersson ME, Aslan D, Clarke A, Roeraade J, Hagman G. Evalua-
acetic acid, a versatile reagent for the determination of enantiomeric tion of generic chiral liquid chromatography screens for pharmaceu-
composition of alcohols and amines. J Org Chem 1969;34:2543– tical analysis. J Chromatogr A 2003;1005:83–101.
2549. 219. Andersson S, Allenmar S. Preparative chiral chromatographic reso-
199. Trost B, Belletire J, Godleski S, McDougal P, Balkovec J, Baldwin J, lution of enantiomers in drug discovery. J Biochem Biophys Meth-
Christy M, Ponticello G, Varga S, Springer ,JP. On the use of the O- ods 2002;54:11–23.
methylmandelate ester for establishment of absolute configuration 220. Zong-De Z, Yan-Ping S, Ting W. Development and validation of
of secondary alcohols. J Org Chem 1986;51:2370–2374. HPLC methods for enantioseparation of mirtazapine enantiomers at
200. Ohtani I, Kusumi T, Kashman Y, Kakisawa H. A new aspect of the analytical and semipreparative scale using polysaccharide chiral sta-
high-field NMR application of Mosher’s method. The absolute tionary phases. Anal Chim Acta 2005;550(1/2):123–129.
configuration of marine triterpene sipholenol-A. J Org Chem 221. Koppenhoefer B, Nothdurft A, Pierrot-Sanders J, Piras P, Popescu
1991;56:1296–1298. C, Roussel C, Stiebler M, Trettin U. CHIRBASE, a graphical molecu-
201. Seco J, Quinoa E, Riguera R. The assignment of absolute configura- lar database on the separation of enantiomers by liquid-, supercriti-
tion by NMR. Chem Rev 2004;104:17–117. cal fluid-, and gas chromatography. Chirality 1993;5:213–219.
202. Seco J, Quinoa E, Riguera R. A ppractical guide for the assignment 222. Koppenhoefer B, Graf R, Holzschuh H, Nothdurft A, Trettin U, Piras
of the absolute configuration of alcohols, amines and carboxylic P, Roussel C. CHIRBASE, a molecular database for the separation of
acids by NMR. Tetrahedron: Asymmetry 2001;12:2915–2925. enantiomers by chromatography. J Chromatogr A 1994;666(1/2):
203. Ohtani I, Kusumi T, Kashman Y, Kakisawa H. High-field FT NMR 557–563.
application of Mosher’s method. The absolute configurations of ma- 223. Roussel C, Piras P, Heitmann I. An approach to discriminating 25
rine terpenoids. J Am Chem Soc 1991;113:4092–4096. commercial chiral stationary phases from structural data sets extracted
204. Devlin F, Stephens P, Besse P. Are the absolute configurations of 2- from a molecular database. Biomed Chromatogr 1997; 11:311.
(1-hydroxyethyl)-chromen-4-one and its 6-bromo derivative deter- 224. Piras P, Roussel C, Pierrot-Sanders J. Reviewing mobile phases used
mined by X-ray crystallography correct? A vibrational circular on Chiralcel OD through an application of data mining tools to
dichroism study of their acetate derivatives. Tetrahedron: Asymme- CHIRBASE database. J Chromatogr A 2001;906(1/2):443–458.
try 2005;16:1557–1566. 225. Roussel C, Pierrot-Sanders J, Heitmann I, Piras P. CHIRBASE:
205. Li J, Burgett A, Amezcua C, Harran P. Total synthesis of nominal Database current status and derived research applications using mo-
diazonamides, Part 2: on the true structure and origin of the natural lecular similarity, decision tree and 3D ‘‘enantiophore’’ search. In:
isolates. Angew Chem Int Ed Eng 2001;40:4770–4773. Subramanian G, editor. Chiral separation techniques, 2nd ed. Wein-
206. Childers Jr W, Abou-Gharbia M, Kelly M, Andree T, Harrison B, heim, Germany: Wiley-VCH Verlag GmbH; 2001.
Ho D, Hornby G, Huryn D, Potestio L, Rosenzweig-Lipson SJ, 226. Del Rio A, Piras P, Roussel C. Enantiophore modeling in 3D-QSAR.
Schmid J, Smith DL, Sukoff SJ, Zhang G, Schechter LE. Synthesis A data mining application on Whelk-O1 chiral stationary phase.
and biological evaluation of benzodioxanylpiperazine derivatives as J Chirality 2006;18:498–508.
potent serotonin 5-HT1A antagonists: the discovery of Lecozotan. 227. Navratilova H, Opatrilova R, Kriz Z, Koca J. Enantioselective chro-
J Med Chem 2005;48:3467–3470. matography and molecular modeling of novel aryloxyaminopropan-
207. Yamashita A, Norton E, Petersen P, Rasmussen B, Singh G, Yang Y, 2-ols with the alkyl carbamate function. J Chirality 2004;16:139–146.
Mansour T, Ho D. Murayamycins, novel peptidoglycan biosynthesis 228. Del Rio A, Hayes J, Stein M, Piras P, Roussel C. Theoretical
inhibitors: synthesis and SAR of their analogues. Bioorg Med Chem reassessment of Whelk-O1 as an enantioselective receptor for 1-(4-
Lett 2003;13:3345–3350. halogeno-phenyl)-1-ethylamine derivatives. Chirality 2004;16(Suppl):
208. Yamashita A, Norton E, Williamson R, Ho D, Sinishtaj S, Mansour S1–S11.
T. Use of bis-(chiral-a-methylbenzyl)glycine esters for synthesis of 229. Kafri R, Lancet D. Probability rule for chiral recognition. Chirality
enantiopure a-hydroxyamino esters. Org Lett 2003;5:3305–3308. 2004;16:369–378.
209. Sabb A, Stein R, Vogel R, Tasse R, Amburn S, Fairman D, Kowal D, 230. Caetano S, Aires-de-Sousa J, Daszykowski M, Heyden Y. Prediction
Malhotra D, Boast C, Bartolomeo A, Morris H, Sailer T, Moyer JA, of enantioselectivity using chirality codes and classification and
Abou-Gharbia M, Ho DM. WAY-131256 is an orally active, effica- regression trees. Anal Chim Acta 2005;544:315–326.
cious, and in vivo functionally selective M1 agonist. Drug Dev Res 231. de la Puente M, White C, Rivera-Sagredo A, Reilly J, Burton K, Har-
1997;40:185–192. vey G. Impact of normal-phase gradient elution in chiral chromatog-
210. Wang T, Shirota O, Nakanishi K, Berova N, McDonald L, Barbieri raphy: a novel, robust, efficient and rapid chiral screening proce-
L, Carter G. Absolute stereochemistry of the spiroxins. Can J Chem dure. J Chromatogr A 2003;983:101–114.
2001;79:1786–1791. 232. Andersson M, Aslan D, Clarke A, Roeraade J, Hagman G. Evaluation
211. Guo J, Schlingmann G, Carter G, Nakanishi K, Berova N. Absolute of generic chiral liquid chromatography screens for pharmaceutical
stereochemistry of the strevertenes. J Chirality 2000;12:43–51. analysis. J Chromatogr A 2003;1005:83–101.
Chirality DOI 10.1002/chir
678 MCCONNELL ET AL.

233. Perrin C, Vu VA, Matthijs N, Maftouh M, Massart DL, Heyden YV. 253. Berger T. Analytical and semipreparative supercritical fluid chroma-
Screening approach for chiral separation of pharmaceuticals, Part I: tography in drug discovery. Drugs Pharma Sci 2004;138:497–538.
normal-phase liquid chromatography. J Chromatogr A 2002;947: 69–83. 254. Cirilli R, Orlando V, Ferretti R, Luciana T, Silvestri R, De Martino G,
234. Matthijs N, Perrin C, Maftouh M, Massart D, Heyden Y. Definition La Torre F. Direct HPLC enantioseparation of chiral aptazepine
and system implementation of strategies for method development of derivatives on coated and immobilized polysaccharide-based chiral
chiral separations in normal-or reversed-phase liquid chromatogra- stationary phases. J Chirality 2006;18:621–632.
phy using polysaccharide-based stationary phases. J Chromatogr A
255. Ghanem A, Hoenen H, Aboul-enein H. Application and comparison
2004;1041:119–133.
of immobilized and coated amylose tris-(3,5-dimethylphenylcarba-
235. Kellogg R, Nieuwenhuijzen J, Pouwer K, Vries T, Broxterman Q, mate) chiral stationary phases for the enantioselective separation of
Grimbergen R, Kaptein B, Crois FL, Wever Ed, Zwaagstra K, van b-blockers enantiomers by liquid chromatography. Talanta 2006;68:
der Laan AC. Dutch resolution: separation of enantiomers with fami-
602–609.
lies of resolving agents. A status report. Synthesis 2003;10:1626–1638.
256. Maftouh M, Granier-Loyaux C, Chavana E, Marini J, Pradines A,
236. Kaptein B, Vries T, Nieuwenhuijzen J, Kellogg R, Grimbergen R,
Heyden YV, Picard C. Screening approach for chiral separation of
Broxterman Q. Continuous developments in classical resolution:
pharmaceuticals, Part III: supercritical fluid chromatography for
Dutch resolution and asymmetric transformation. Life Sciences-
analysis and purification in drug discovery. J Chromatogr A 2005;
Advanced Synthesis & Catlysis. PharmaChem 2003;2:17–22.
1088:67–81.
237. Nieuwenhuijzen J, Grimbergen R, Koopman C, Kellogg R, Vries T,
Pouwer K, van Echten E, Kaptein B, Hulshof L, Broxterman Q. The 257. White C. Integration of supercritical fluid chromatography into drug
role of nucleation inhibition in optical resolutions with families of discovery as a routine support tool, Part I: fast chiral screening and
resolving agents. Angew Chem Int Ed Engl 2002;41:4281–4286. purification. J Chromatogr A 2005;1074:163–173.

238. Kaptein B, Boesten W, De Lange B, Grimbergen R, Hulshof L, Vries 258. Ye Y, Stringham R. The effect of acidic and basic additives on the
T, Seerden J-P, Broxterman Q. Production of enantiopure enantioseparation of basic drugs using polysaccharide-based chiral
compounds by crystallization-induced resolution. Chim Oggi stationary phases. J Chirality 2006;18:519–530.
2002;20(3/4):77–82. 259. Gyllenhaal O, Stefansson M. Reversal of elution order for profen
239. Kaptein B, Elsenber H, Grimbergen R, Broxterman Q, Hulshof L, acid enantiomers in packed-column SFC on chiralpak AD. Chirality
Pouwer K, Vries T. Dutch resolution of racemic 4-hydroxy- and 4-flu- 2005;17:257–265.
orophenylglycine with mixtures of phenylglycine and (+)-10-cam- 260. Roussel C, Vanthuyne N, Serradeil-Albalat M, Vallejos J-C. True or
phorsulfonic acid. Tetrahedron: Asymmetry 2000;11:1343–1351. apparent reversal of elution order during chiral high-performance
240. Vries T, Wynberg H, van Echten E, Koek J, ten Hoeve W, Kellog R, liquid chromatography monitored by a polarimetric detector under
Broxterman Q, Minnaard A, Kaptein B, van der Sluis S, Hulshof L, different mobile phase conditions. J Chromatogr A 2003;995:79–85.
Kooistra J. The family approach to the resolution of racemates. 261. Okamoto M. Reversal of elution order during the chiral separation
Angew Chem Int Ed Engl 1998;37:2349–2354. in high performance liquid chromatography. J Pharma Biomed Anal
241. Lammerhofer M. Chiral separations by capillary electromigratoin 2002;27:401–401.
techniques in nonaqueous media, Part I: enantioselective nonaqu- 262. Xiao T, Zhang B, Lee J, Hui F, Armstrong D. Reversal of enantio-
eous capillary electrophoresis. J Chromatogr A 2005;1068:3–30. meric elution order on macrocyclic glycopeptide chiral stationary
242. Lammerhofer M. Chiral separations by capillary electomigration phases. J Liquid Chrom Relat Technol 2001;24:2673–2684.
techniques in nonaqueous media, Part II: enantioselective nonaqu- 263. Zhang Y, McConnell O. Simulated moving columns technique for
eous capillary electrochromatography. J Chromatogr A 2005;1068: chiral liquid chromatography. J Chromatogr A 2004;1028:227–238.
31–57. 264. Rodrigues A, APais L. Design of SMB chiral separations using the
243. Kang J. Recent progress in enantiomeric separation by capillary concept of separation volume. Sep Sci Technol 2004;39:245–270.
electrochromatography. Electrophoresis 2002;23:4005–4021. 265. Chin C, Wang N-H. Simulated moving bed equipment designs. Sep
244. Scriba G. Selected fundamental aspects of chiral electromigration Purific Rev 2004;33:77–155.
techniques and their application to pharmaceutical and biomedical 266. Biressi G, Rajendran A, Mazzotti M, Morbidelli M. The GC-SMB
analysis. J Pharm Biomed Anal 2002;27:373–399. separation of the enantiomers of isoflurane. Sep Sci Technol
245. Enlund A, Andersson M, Hagman G. Improved quantification limits 2002;37:2529–2543.
in chiral capillary electrochromatography by peak compression 267. Imamoglu S. Simulated moving bed chromatography (SMB) for
effects. J Chromatogr A 2004;1028:333–338. application in bioseparation. Adv Biochem Eng/Biotechnol 2002;76:
246. Levkin P, Levkina A, Schurig V. Combining the enantioselectivities of 211–231.
L-valine diamide and permethylated b-cyclodextrin in one gas chro- 268. Huthmann E, Juza M. Modification of a commercial chiral stationary
matographic chiral stationary phase. Anal Chem 2006;78:5143–5148. phase. Influences on enantiomer separations using simulated mov-
247. Volker S. Contributions to the theory and practice of the chromato- ing bed chromatography. J Chromatogr A 2001;908(1/2):185–200.
graphic separation of enantiomers. Chirality 2005;17(Suppl):S205–S226. 269. Ludemann-Hombbourger O, Bailly M, Micoud R. Design of a simu-
248. Kasai H, Tsubuki M, Takahashi K, Shirao M, Matsumoto Y, Honda lated moving bed: optimal particle size of the stationary phase. Sep
Y, Seyama Y. Separation of stereoisomers of several furan deriva- Sci Technol 2000;35:1285–1305.
tives by capillary gas chromatography-mass spectrometry, supercriti- 270. Juza M, Mazzotti M, Morbidelli M. Simulated moving-bed chroma-
cal fluid chromatography, and liquid chromatography using chiral tography and its application to chirotechnology. Trends Biotechnol
stationary phases. J Chromatogr A 2002;977:125–134. 2000;18:108–118.
249. Schurig V. Separation of enantiomers by gas chromatography. J 271. Kuester E, BGerber G, Antia F. Enantioseparation of a chiral epox-
Chromatogr A 2001;906:275–299. ide by simulated moving bed chromatography using Chiralcel-OD.
250. Berger TA. Separation of polar solutes by packed column supercriti- Chromatographia 1995;40(7/8):387–393.
cal fluid chromatography. J Chromatogr A 1997;785:3–33. 272. Zhang Y, Watts W, Nogle L, McConnell O. Rapid method develop-
251. Berger T, Fogleman K, Staats T, Bente P, Crocket I, Farrell W, ment for chiral separation in drug discovery using multi-column par-
Osonubu M. The development of a semi-preparatory scale supercriti- allel screening and circular dichroism signal pooling. J Chromatogr
cal-fluid chromatograph for high-throughput purification of ‘combi- A 2004;1049(1/2):75–84.
chem’ libraries. J Biochem Biophys Methods 2000;43:87–111. 273. Kagan M. Normal-phase high-performance liquid chromatographic
252. Berger T, Smith J, Fogelman K, Kruluts K. Semipreparative chiral separations using ethoxynonafluorobutane as hexane alternative,
separations using supercritical fluid chromatography with stacked Part I: analytical and chiral applications. J Chromatogr A 2001;918:
injections. Am Lab 2002;34:14–20. 293–302.
Chirality DOI 10.1002/chir
BUILDING CHIRAL TECHNOLOGY TOOLBOXES 679

274. Kagan M, Chlenov M, Kraml C. Normal-phase high-performance liq- 295. Di L, McConnell O, Kerns E, Sutherland A. Rapid, automated
uid chromatographic separations using ehtoxynonafluorobutane as screening method for enzymatic transformations using a robotic sys-
hexane alternative, Part II: liquid chromatography-atmospheric pres- tem and supercritical fluid chromatography. J Chromatogr B 2004;
sure chemical ionization-mass spectrometry applications with metha- 809:231–235.
nol gradients. J Chromatogr A 2004;1033(1/2):321–331.
296. Huang X, Fujioka N, Pescitelli G, Koehn F, Williamson R, Nakanishi
275. Ding J, Desai M, Armstrong D. Evaluation of ethoxynonafluorobu- K, Berova N. Absolute configurational assignments of secondary
tane as a safe and environmentally friendly solvent for chiral normal- amines by CD-sensitive dimeric zinc porphyrin host. J Am Chem
phase LC-atmospheric pressure chemical ionization/electrospray
Soc 2002;124:10320–10335.
ionization-mass spectrometry. J Chromatogr A 2005;1076(1/2):34–43.
297. Kurtan T, Nesnas N, Li Y-Q, Huang X, Nakanishi K, Berova N. Chiral
276. Kagan M, Chlenov M, Bach A, McConnell O. Mass-directed normal-
recognition by CD-sensitive dimeric zinc porphyrin host, Part 1: chirop-
phase preparative HPLC with atmospheric pressure chemical ioniza-
tion detection. J Liquid Chrom Relat Technol 2004;27:1817–1834. tical protocol for absolute configurational assignments of monoalcohols
and primary monamines. J Am Chem Soc 2001;123: 5962–5973.
277. Kagan M, Chlenov M, Greenfield A, Ho D. Normal-phase chiral liq-
uid chromatography-mass spectrometry of non-UV-active com- 298. Kurtan T, Nesnas N, Koehn F, Li Y-Q, Nakanishi K, Berova N.
pounds: Applications for pharmaceutically relevant racemates. J Chiral recognition by CD-sensitive dimeric zinc porphyrin host. 2.
Chromatogr A 2006;1120:82–88. Structural studies of host-guest with chiral alcohol and monamine
conjugates. J Am Chem Soc 2001;123:5974–5982.
278. Cai Y, Kingery D, McConnell O, Bach IIIA. Advantages of atmos-
pheric pressure photoionization mass spectrometry in support of 299. Huang T, Nakanishi K, Berova N. Porphyrins and metalloporphyr-
drug discovery. Rapid Commun Mass Spectrom 2005;19:1717–1724. ins: versatile circular dichroic reporter groups for structural studies.
J Chirality 2000;12:237–255.
279. Kraml C, Zhou D, Byrne N, McConnell O. Enhanced chromato-
graphic resolution of amine enantiomers as carbobenzyloxy deriva- 300. Freedman T, Cao X, Dukor R, Nafie L. Absolute configuration deter-
tives in high-performance liquid chromatography and supercritical mination of chiral molecules in the solution state using vibrational
fluid chromatography. J Chromatogr A 2005;1100:108–115. circular dichroism. Chirality 2003;15:743–758.
280. Nogle L, Mann C, Watts W Jr, Zhang Y. Preparative separation and 301. Stephens P. Vibrational circular dichroism spectroscopy: a new tool
identification of derivatized b-methylphenylalanine enantiomers by for the stereochemical characterization of chiral molecules. Comput
chiral SFC, HPLC and NMR for development of new peptide ligand Med Chem Drug Discov 2004:699–725.
mimetics in drug discovery. J Pharm Biomed Anal 2006;40:901–909. 302. Polavarapu P, He J. Chiral analysis using mid-IR vibrational CD
281. Krishna S. Developments and trends in enzyme catalysis in noncon- spectroscopy. Anal Chem 2004:61A–67A.
ventional media. Biotechnol Adv 2002;20(3/4):239–267. 303. Kuppens T, Herrebout W, Van der Veken B, Corens D, De Groot A,
282. Koeller K, Wong C-H. Enzymes for chemical synthesis. Nature Doyon J, Van Lommen G, Bultinck P. Elucidation of the absolute
2001;409:232–240. configuration of JNJ-27553292, a CCR2 receptor antagonist, by vibra-
tional circular dichroism analysis of two precursors. Chirality 2006;
283. Klibanov A. Improving enzymes by using them in organic solvents. 18:609–620.
Nature 2001;409:241–246.
304. Freedman T, Cao X, Phillips L, Cheng P, Dalterio R, Shu Y-Z, Zhang
284. Thayer A. Enzymes at work. Chem Eng News 2006;84:15–25. H, Zhao N, Shukla R, Tymiak A, Gozo SK, Nafie LA, Gougoutas JZ.
285. Vieira T, Ferraz H, Andrade L, Porto A. Highly enantioselective enzy- Determination of the absolute configuration and solution conforma-
matic resolution of cis-fused octalols mediated by Candida Antarctica tion of a novel disubstituted pyrrolidine acid a by vibrational circular
lipase (Novozym 435). Tetrahedron: Asymmetry 2006;17:1990–1994. dichroism. Chirality 2006; 18:746–753.
286. Fransson A-B, Xu Y, Leijondahl K, Baeckvall J-E. Enzymatic resolu- 305. Morita H, Kodama T, Tanaka T. Chirality of camphor derivatives by
tion, desymmetrization, and dynamic kinetic asymmetric transforma- density functional theory. Chirality 2006;18:783–789.
tion of 1,3-cycloalkanediols. J Org Chem 2006;71:6309–6316. 306. Giorgio E, Tanaka K, Ding W, Krishnamurthy G, Pitts K, Ellestad G,
287. Easwar S, Argade N. A facile synthesis and enzymatic resolution of Rosini C, Berova N. Theoretical simulation of the electronic circular
naturally occurring remotely functionalized alkylmethylmaleic anhy- dichroism spectrum of calicheamicin. Bioorg Med Chem 2005;13:
drides from Aspergillus wentii: aspergillus acids A–D. Synthesis 5072–5079.
2006;5:831–838. 307. Schuhly W, Crockett SL, Fabian WMF. Hyperolactone C: determina-
288. Miyazawa T, Kaito E, Yukawa T, Murashima T, Yamada T. Enzy- tion of its absolute configuration by comparison of experimental and
matic resolution of 2-aryloxy-1-propanols via lipase-catalyzed enantio- calculated CD spectra. Chirality 2005;17:250–256.
selective acylation using acid anhydrides as acyl donors. J Mol Catal 308. Superchi S, Giogio E, Rosini C. Structural determinations by circular
B Enzym 2005;37(1–6):63–67. dichroism spectra analysis using coupled oscillator methods: an
289. Garcia-Urdiales E, Rebolledo F, Gotor V. Enzymatic one-pot resolu- update of the applications of the DeVoe polarizability model. Chiral-
tion of two nucleophiles: alcohol and amine. Tetrahedron: Asymme- ity 2004;16:422–451.
try 2000;11:1459–1463. 309. Polavarapu P. Protocols for the analysis of theoretical optical rota-
290. Lou W-Y, Zong M-H. Efficient kinetic resolution of (R,S)-1-trimethyl- tions. Chirality 2006;18:348–356.
silylethanol via lipase-mediated enantioselective acylation in ionic 310. Polavarapu P, Petrovic A, Wang F. Intrinsic rotation and molecular
liquids. Chirality 2006;18:814–821. structure. Chirality 2003;15(Suppl):S143–S149.
291. Bellur E, Boettcher D, Bornscheuer U, Langer P. Enantioselective 311. Polavarapu P. Optical rotation: Recent advances in determining the
synthesis of 2-alkylidenetetrahydrofurans based on a cyclization/enzy- absolute configuration. Chirality 2002;14:768–781.
matic resolution strategy. Tetrahedron: Asymmetry 2006;17:892–899. 312. Giorgio E, Roje M, Tanaka K, Hamersak Z, Sunjic V, Nakanishi K,
292. Shaw S-Y, Chen Y-J, Ou J-J, Ho L. Enzymatic resolution of methyl Rosini C, Berova N. Determination of the absolute configuration of
DL-b-acetylthioisobutyrate and DL-b-acetylthioisobutyramide using a flexible molecules by ab initio ORD calculations: a case study with
stereoselective esterase from Pseudomonas putida IFO12996. J Mol Cytoxazones and Isocytoxazones. J Org Chem 2005;70:6557–6563.
Catal B Enzym 2006;38(3–6):163–170. 313. Stephens P, McCann D, Cheeseman J, Frisch F. Determination of
293. Truppo M, Journet M, Shafiee A, Moore J. Optimization and scale- absolute configurations of chiral molecules using ab inito time-de-
up of a lipase-catalyzed enzymatic resolution of an indole ester inter- pendent density functional theory calculations of optical rotation:
mediate for a prostaglandin D2 (DP) receptor antagonist targeting how reliable are absolute configurations obtained for molecules with
allergic rhinitis. Org Process Res Dev 2006;10:592–598. small rotations? Chirality 2005;17:S52–S64.
294. Xin J, Zhao Y, Zhao G, Zheng Y, Ma X, Xia C, Li S. Enzymatic resolu- 314. McCann D, Stephens P, Cheeseman J. Determination of absolute
tion of (R,S)-Naproxen in water-saturated ionic liquid. Biocatal Bio- configuration using density functional theory calculation of optical
transform 2005;23:353–361. rotation: chiral alkanes. J Org Chem 2004;69:8709–8717.
Chirality DOI 10.1002/chir
680 MCCONNELL ET AL.

315. Stephens P, Devlin F, Cheeseman J, Frisch M, Bortoline O, Besse 332. Vass E, Hollosi M, Forro E, Fulop F. VCD spectroscopic investigation
P. Determination of absolute configuration using ab initio calculation of enantiopure cyclic b-lactams obtained through lipolase-catalyzede-
of optical rotation. Chirality 2003;15(Suppl):S57–S64. nantioselective ring-opening reaction. Chirality 2006;18:733–740.
316. Stephens P, Devlin F, Cheeseman J, Frisch F, Rosini C. Determina- 333. He J, Wang F, Polavarapu P. Absolute configurations of chiral herbi-
tion of absolute configuration using optical rotation calculated using cides determined from vibrational circular dichroism. J Chirality 2005;
density functional theory. Org Lett 2002;4:4595–4598. 17:S1–S8.
317. Coriani S, Baranowska A, Ferrighi L, Forzato C, Marchesan D, Nitti 334. Devlin F, Stephens P, Bortolini O. Determination of absolute config-
P, Pitacco G, Rizzo A, Ruud K. Solvent effects on the conformational uration using vibrational circular dichroism spectroscopy: phenyl
distribution and optical rotation of g-methylparaconic acids and glycidic acid derivatives obtained via asymmetric epoxidation using
esters. Chirality 2006;18:357–369. oxone and a keto bile acid. Tetrahedron: Asymmetry 2005;16:2653–
318. Specht K, Nam J, Ho D, Berova N, Kondru R, Beratan D, Wipf P, 2663.
Pascal R Jr, Kahne D. Determining absolute configuration in flexible 335. Freedman T, Dukor R, Hoof Pv, Bach EK, Nafie L. Determination of
molecules: a case study. J Am Chem Soc 2001;123:8961–8966. the absolute configuration of ()-mirtazapine by vibrational circular
319. Freedman T, Cao X, Nafie L, Kalbermatter M, Linden A, Rippert A. dichroim. Helvetica Chim Acta 2002;85:1160–1165.
Determination of the atropisomeric stability and solution conforma- 336. Shah R, Nafie L. Spectroscopic methods for determining enantio-
tion of asymmetrically substituted biphenyls by means of vibrational meric purity and absolute configuration in chiral pharmaceutical
circular dichroism (VCD). Helvetic Chim Acta 2005;88:2302–2314. molecules. Curr Opin Drug Discov Dev 2001;4:764–775.
320. Cichewicz R, Clifford L, Lassen P, Freedman XCT, Nafie L, 337. Petrovic A, He J, Polavarapu P, Xiao L, Armstrong D. Absolute con-
Deschamps J, Kenyon V, Flanary J, Holman T, Crews P. Stereo- figuration and predominant conformations of 1,1-dimethyl-2-phenyl-
chemical determination and bioactivity assessment of (S)-(+)-curcu- ethyl phenyl sulfoxide. Org Biomol Chem 2005;3:1977–1981.
phenol dimers isolated from the marine sponge Didiscus aceratus 338. Petrovic A, Polavarapu P, Drabowicz J, Zhang Y, McConnell O, Dud-
and synthesized through laccase biocatalysis. Bioorg Med Chem deck H. Absolute configuration of C2-symmetric spiroselenurane:
2005;13:5600–5612. 3,3,30 30 -tetramethyl-1,10 spriobi[3H. 2.1]benzoxaselenole. Chem Eur J
321. Freedman T, Cao X, Oliveira R, Cass Q, Nafie L. Determination of 2005;11:4257–4262.
the absolute configuration and solution conformation of gossypol by 339. Stephens P, McCann D, Butkus E, Stoncius S, Cheeseman J, Frisch
vibrational circular dichroism. Chirality 2003;15:196–200. M. Determination of absolute configuration using concerted ab initio
322. Freedman T, Nafie XCL, Kalbermatter M, Linden A, Rippert A. An DFT calculations of electronic circular dichroism and optical rota-
unexpected atropisomerically stable 1,1-biphenyl at ambient temper- tion: bicyclo[3.3.1]nonane diones. J Org Chem 2004;69:1948–1958.
ature in solution, elucidated by vibrational circular dichroism 340. Stephens P, McCann D, Devlin F, Cheeseman J, Frisch M. Determi-
(VCD). Helvetica Chim Acta 2003;86:3141–3155. nation of the absolute configuration of [3.2](1,4)barrelenophanedi-
323. Urbanova M, Setnicka V, Devlin F, Stephens P. Determination of carbonnitrile using concerted time-dependent density functional
molecular structure in solution using vibrational circular dichroism theory calculations of optical rotation and electronic circular dichro-
spectroscopy: the supramolecular tetramer of S-2,20 -dimethyl- ism. J Am Chem Soc 2004;126:7514–7521.
biphenyl-6,60 dicarboxylic acid. J Am Chem Soc 2005;127:6700–6711. 341. Stephens P, McCann D, Devlin F, Flood T, Butkus E, Stoncius S,
324. Mazaleyrat J-P, Wright K, Gaucher A, Wakselman M, Oancea S, Cheeseman J. Determination of molecular structure using vibra-
Formaggio F, Toniolo C, Setnicka V, Kapitan J, Keiderling T. Syn- tional circular dichroism spectroscopy: the keto-lactone product of
thesis and conformational study of homo-peptides based on (S)-bin, Baeyer-Villiger oxidation of (+)-(1R,5S)-bicyclo[3.3.1]nonane-2,7-
a C2-symmetric binaphthyl-derived Caa-disubstituted blycine with dione. J Org Chem 2005;70:3903–3913.
only axial chirality. Tetrahedron: Asymmetry 2003;14:1879–1893. 342. Van Klink JW, Baek S-H, Barlow AJ, Ishii H, Nakanishi K, Berova
325. Dunmire D, Freedman T, Nafie L, Aeschlimann C, Gerber J, Gal J. N, Perry NB, Weavers RT. Determination of the absolute configura-
Determination of the absolute configuration and solution conforma- tion of Anisotome irregular diterpenes: application of CD and NMR
tion of the antifungal agents ketoconazole, itraconazole, and mi- methods. Chirality 2004;16:549–558.
conazole with vibrational circular dichroism. Chirality 2005;17: 343. Vogensen SB, Greenwood JR, Varming AR, Brehm L, Pickering DS,
S101–S108. Nielsen B, Liljefors T, Clausen RP, Johansen TN, Krogsgaard-
326. Freedman T, Cao X, Nafie L, Solladie-Cavallo A, Jierry I, Bouerat L. Larsen P. A stereochemical anomaly: the cyclized (R)-AMPA
VCD configuration of enantiopure/-enriched tetrasubstituted a-fluo- analogue (R)-3-hydroxy-4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridine-5-
rocyclohexanones and their use for epoxidation of trans-olefins. carboxylic acid [(R)-5-HPCA] resembles (S)-AMPA at glutamate
Chirality 2004;16:467–474. receptors. Org Biomol Chem 2004;2:206–213.
327. Bour P, Navratilova H, Setnicka V, Urbanova M, Karel V. (3R,4S)-4- 344. Solladie-Cavallo A, Marsol C, Pescitelli G, DiBari L, Salvadori P,
(4-fluorophenyl)-3-hydroxymethyl-1-methylpiperidine: conformation Huang X, Fujioka N, Berova N, Cao X, Freedman T, et al. (R)-(+)-
and structure monitoring by vibrational circular dichroism. J Org and (S)-()-1-(9-phenanthryl)ethylamine: assignment of absolute
Chem 2002;67:161–168. configuration by CD tweezer and VCD methods, and difficulties
encountered with the CD exciton chirality method. Eur J Org Chem
328. Guo C, Shah R, Dukor R, Cao X, Freedman T, Nafie L. Determina-
2002;11:1788–1796.
tion of enantiomeric excess in samples of chiral molecules using
Fourier transform vibrational circular dichroism spectroscopy: simu- 345. Lodine Monograph; 1993. 2581–2584 p.
lation of real-time reaction monitoring. Anal Chem 2004;76:6956– 346. Demerson C, Humber L, Dobson T, Jirkovsky I. Pyrano[3,4-b]-in-
6966. dolederivatives, pharmaceutical compositions and methods of use.
329. Maryanoff B, McComsey D, Dukor R, Nafie L, Freedman T, Cao X, US Patent No. 4,076,831, February 28, 1978.
Day V. Structural studies on McN-5652-X, a high-affinity ligand for 347. Demerson C, Humber L, Phillip A. Etodolic acid and related com-
the serotonin transporter in mammalian brain. Bioorg Med Chem pounds. Chemistry and antiinflammatory actions of some potent
2003;11:2463–2470. di- and trisubstituted 1,3,4,9-tetrahydropyrano[3,4-b]indole-1-acetic
330. Guo C, Shah R, Mills J, Dukor R, Cao X, Freedman T, Nafie L. Fou- acids. J Med Chem 1976;19:391–395.
rier transform near-infrared vibrational circular dichroism used for 348. Gonzalez A. A convenient preparation of 8-ethyl-4,9-dihydro-3H-pyr-
on-line monitoring the epimerization of 2,2-dimethyl-1,3-dioxolane-4- ano[3,4-b]indol-1-one, key intermediate of the antiinflammatory
methanol: A pseudo racemization reaction. Chirality 2006;18:775– agent etodolac. Synth Commun 1991;21:669–674.
782. 349. Humber L, Demerson C, Swaminathan P, Bird P. Etodolac (1,8-
331. Monde K, Taniguchi T, Miura N, Vairappan C, Suzuki M. Absolute diethyl-1,3,4,9-tetrahydropyrano[3,4-b]indole-1-acetic acid): a potent
configurations of brominated sesquiterpenes determined by vibra- antiinflammatory drug. Conformation and absolute configuration of
tional circular dichroism. Chirality 2006;18:335–339. its active enantiomer. J Med Chem 1986;29:871–874.
Chirality DOI 10.1002/chir
BUILDING CHIRAL TECHNOLOGY TOOLBOXES 681

350. Brenna E, Fuganti C, Fuganti D, Grasseli P, Malpezzi L, Pedrocchi- 370. Hoye T, Hamad A-S, DO K, MA T. An NMR method for determina-
Fantoni G. New enzymatic and chemical approaches to enantiopure tion of configuration of b-substituted carboxylic acids. Tetrahedron
etodolac. Tetrahedron 1997;53(52):17769–17780. Lett 2000;41:2289–2293.
351. Chou S-Y, Tseng C-L, Chang L-S. Exploration of an efficient method 371. Ferreiro M, Latypov S, Quinoa E, Riguera R. The use of ethyl 2-(9-
for optical resolution of etodolac. J Chin Chem Soc 2001;48:229–234. anthryl)-2-hydroxyacetate for assignment of the absolute configura-
352. Chou S-Y, Tseng C-L, Chen S-F. The formal synthesis of chiral eto- tion of carboxylic acids by 1H NMR. Tetrahedron: Asymmetry
dolac using chiral 1,2-di(alkylcarbonyl)oxypentan-3-one as chiral 1997;8:1015–1018.
building block. Heterocycles 1999;51:1527–1541. 372. Hoye T, Koltun D. An NMR strategy for determination of configura-
353. Williamson R, Barrios Sosa A, Mitra A, Seaton P, Weibel D, tion of remote stereogenic centers: 3-methylcarboxylic acids. J Am
Schroeder F, Meinwald J, Koehn F. New silyl ether reagents for the Chem Soc 1998;120:4638–4643.
absolute stereochemical determination of secondary alcohols. Org 373. Yang Y, Kayser M, Hooper D. Assignment of absolute configuration
Lett 2003;5:1745–1748. of 3-hydroxy b-lactams by the NMR ‘‘mix and shake’’ method. Chir-
354. Weibel D, Walker T, Schroeder F, Meinwald J. Chiral silylation ality 2005;17:131–134.
reagents for the determination of absolute configuration by NMR 374. Kasai Y, Taji H, Fujita T, Yamamoto Y, Akagi M, Sugio A, Kuwahara
spectroscopy. Org Lett 2000;2:2381–2383. S, Watanabe M, Harada N, Ichikawa A, Schurig V. MaNP acid, a
powerful chiral molecular tool for preparation of enantiopure alco-
355. Seco J, Latypov S, Quinoa E, Riguera R. Determining factors in the
hols by resolution and determination of their absolute configura-
assignment of the absolute configuration of alcohols by NMR. The
tions by the 1H NMR anisotropy method. Chirality 2004;16:569–
use of anisotropic effects on remote positions. Tetrahedron 1997;53:
585.
8541–8564.
375. Taji H, Kasai Y, Sugio A, Kuwahara S, Watanabe M, Harada N,
356. Latypov S, Seco J, Quinoa E, Riguera R. MTPA vs PMA in the deter-
Ichikawa A. Practical enantioresolution of alcohols with 2-methoxy-2-
mination of the absolute configuration of chiral alcohols by 1H
(1-naphthyl)propionic acid and determination of their absolute
NMR. J Org Chem 1996;61:8569–8577.
configurations by the 1H NMR anisotropy method. Chirality 2002;
357. Reiser M, Hui Y, Rupprech J, Koxlowski J, Wood K, McLaughlin J, 14:81–84.
Hanson P, Zhuang Z, Hoye T. Determination of absolute configura-
376. Seco J, Tseng L-H, Godejohann M, Quinoa E, Riguera R. Simultane-
tion of stereogenic carbinol centers in Annonaceous acetogenins by
1 ous enantioresolution and assignment of absolute configuration of
H- and 19F-NMR analysis of mosher ester derivatives. J Am Chem
secondary alcohols by directly coupled HPLC-NMR of 9-AMA
Soc 1992;114:10203–10213.
esters. Tetrahedron: Asymmetry 2002;13:2149–2153.
358. Kelly D. A new method for the determination of the absolute stereo-
377. Harada N, Watanabe M, Kuwahara S, Sugio A, Kasai Y, Ichikawa A.
chemistry of aromatic and heteraromatic alkanols using Mosher’s
2-Methoxy-2-(1-naphthyl)propionic acid, a powerful chiral auxiliary
esters. Tetrahedron: Asymmetry 1999;10:2927–2934.
for enantioresolution of alcohols and determination of their absolute
359. Freire F, Seco J, Quinoa E, Riguera R. Determining the absolute configurations by the 1H NMR anisotropy method. Tetrahedron:
stereochemistry of secondary diols by 1H NMR: basis and applica- Asymmetry 2000;11:1249–1253.
tions. J Org Chem 2005;70:3778–3790. 378. Cordero F, Pisaneschi F, Salvati M, Valenza S, Faggi C, Brandi A.
360. Konno K, Fujishima T, Liu Z, Takayama H. Determination of abso- Application of Mosher’s method for absolute configuration assign-
lute configuration of 1,3-diols by the modified Mosher’s method ment and resolution of 2-hydroxypyrrolizidinones. Chirality 2005;17:
using their di-MTPA esters. Chirality 2002;13:72–80. 149–153.
361. Latypov S, Ferreiro M, Quinoa E, Riguera R. Assignment of the 379. Wentzel T, Wilcox J. Chiral reagents for the determination of enan-
absolute configuration of b-chiral primary alcohols by NMR: scope tiomeric excess and absolute configuration using NMR spectros-
and limitations. J Am Chem Soc 1998;120:4741–4751. copy. Chirality 2003;15:256–270.
362. Ferreiro M, Latypov S, Quinoa E, Riguera R. Determination of the 380. Lovely A, Wenzel T. Chiral NMR discrimination of pyrrolidines
absolute configuration and enantiomeric purity of chiral primary using (18-crown-6)-2,3,11,12–tetracarboxylic acid. Tetrahedron:
alcohols by 1H NMR of 9-anthrylmethoxyacetates. Tetrahedron: Asymmetry 2006;17:2642–2648.
Asymmetry 1996;7:2195–2198. 381. Kobayashi Y, Czechtizky W, Kishi Y. Complete stereochemistry of
363. Rodriguez-Escrich S, Popa D, Jimeno C, Vidal-Ferran A, Pericas M. tetrafibricin. Org Lett 2003;5:93–96.
(S)-20[(R)-Fluoro(phenyl)methyl]oxirane: a general reagent for 382. Kobayashi N, Hayashi N, Kishi Y. Toward the creation of NMR data-
determining the ee of a-chiral amines. Org Lett 2005;7:3829– bases in chiral solvents: bidentate chiral NMR solvents for assign-
3832. ment of the absolute configuration of acyclic secondary alcohols.
364. Seco J, Quinoa E, Riguera R. Boc-phenylglycine: the reagent of Org Lett 2002;4:411–414.
choice for the assignment of the absolute configuration of a-chiral 383. Kobayashi N, Hayashi M, Kishi Y. Application of chiral bidentate
primary amines by 1H NMR spectroscopy. J Org Chem 1999;64: NMR solvents for assignment of the absolute configuration of alco-
4669–4675. hols: scope and limitation. Tetrahedron Lett 2003;44:7489–7491.
365. Seco J, Latypov S, Quinoa E, Riguera R. Choosing the right reagent 384. Kobayashi Y, Hayashi N, Kishi Y. Application of chiral bidentate
for the determination of the absolute configuration of amines by NMR solvents for assignment of the absolute configuration of alco-
NMR: MTPA or MPA? J Org Chem 1997;62:7569–7574. hols: Scope and limitation. Tetrahedron Lett 2003;44:7489–7491.
366. Latypov S, Seco J, Quinoa E, Riguera R. Determination of the abso- 385. Emsley W, Lindon J. NMR spectroscopy using liquid crystal sol-
lute stereochemistry of chiral amines by 1H NMR of arylmethoxy- vents. Oxford: Pergamon Press; 1975.
acetic acid amides: the conformational model. J Org Chem 386. Farjon J, Merlet D, Philippe L, J C. Enantiomeric excess measure-
1995;60:1538–1545. ments in weakly oriented chiral liquid crystal solvents through 2D
367. Trost B, Bunt R, Pulley S. On the use of O-methylmandelic acid for 1H selective refocusing experiments. J Magn Res 2002;158(1/2):
the establishment of absolute configuration of a-chiral primary 169–172.
amines. J Org Chem 1994;59:4202–4205. 387. Parenty A, Campagne J-M, Aroulanda C, Lesot P. Routine use of nat-
368. Porto S, Duran J, Seco J, Quinoa E, Riguera R. ‘‘Mix and shake’’ ural abundance deuterium NMR in a polypeptidic chiral oriented sol-
method for configurational assignment by NMR: application to chiral vent for the determination of the enantiomeric composition of chiral
amines and alcohols. Org Lett 2003;5:2979–2982. building blocks. Org Lett 2002;4:1663–1666.
369. Latypov S, Galiullina N, Aganov A, Kataev V, Riguera R. Determina- 388. Sarfati M, Lesot P, Merlet D, Courtieu J. Theoretical and experimen-
tion of the absolute stereochemistry of alcohols and amines by tal aspects of enantiomeric differentiation using natural abundance
NMR of the group directly linked to the chiral derivatizing reagent. multinuclear NMR spectroscopy in chiral polypeptide liquid crystals.
Tetrahedron 2001;57:2231–2236. Chem Commun 2000;21:2069–2081.
Chirality DOI 10.1002/chir
682 MCCONNELL ET AL.

389. Canlet C, Merlet D, Lesot P, Meddour A, Lowewenstein A, Courtieu tal solvent using a natural abundance HSQC experiment. Magn
J. Deuterium NMR stereochemical analysis of threo-erythro isomers Reson Chem 2005;43:512–519.
bearing remote stereogenic centers in racemic and non-racemic liq- 395. Thiele C, Berger S. Probing the diasterotopicity of methylene pro-
uid crystalline solvents. Tetrahedron: Asymmetry 2000;11:1911– tons in strychnine using residual dipolar couplings. Org Lett 2003;
1918. 5:705–708.
390. Jakubcova M, Meddour A, Pechine J-M, Baklouti A, Courtieu J.
396. Thiele C. Poly-g-ethyl-glutamate as alignment medium for the mea-
Measurement of the optical purity of fluorinated compounds using
surement of RDCs in organic substrates. J Org Chem 2004;69:7403–
proton decoupled 19F-NMR spectroscopy in a chiral liquid crystal
7413.
solvent. J Fluorine Chem 1997;86:149–153.
397. Verdier L, Sakhaii P, Zweckstetter M, Griesinger C. Measurement
391. Meddour A, Berdague P, Hedli A, Courtieu J, Lesot P. Proton-
of long range H,C couplings in natural products in orientation
decoupled carbon-13 NMR spectroscopy in a lyotropic chiral
media: a tool for structure elucidation of natural products. J Magn
nematic solvent as an analytical tool for the measurement of the
Res 2003;163:353–359.
enantiomeric excess. J Am Chem Soc 1997;119:4502–4508.
392. Chevtchouk T, Ollivier J, Salaun J, Merlet D, Courtieu J. Determi- 398. Walsh R, Brander M, Fleischman S, Morales L, Moulton B,
nation of the regio- and enantioselectivity of the enzymatic hydrol- Rodriguez-Hornedo N, Zaworotko M. Crystal engineering of the
yses of succinates based on H,C COLOC analysis and deuterium composition of pharmaceutical phases. Chem Commun 2003:
NMR in a chiral liquid crystal. Tetrahedron: Asymmetry 1997; 186.
8(7):999. 399. Bax A, Freeman R, Kempsell S. Natural abundance 13C– 13C cou-
393. Canet J-L, Canet I, Courtieu J, Da Silva S, Gelas J, Troin Y. Acetyl-d3 pling observed via double quantum coherence. J Am Chem Soc
chloride: A convenient nonchiral derivatizing agent for a facile enan- 1980;102:4849–4851.
tiomeric excess determination of Amines through deuterium NMR. 400. Clore G, Gronenborn A, Bax A. A robust method for determining
J Org Chem 1996;61:9035–9037. the magnitude of the fully asymmetric alignment tensor of oriented
394. Marathias V, Goljer I, Bach AI. Simultaneous Determination of macromolecules in the absence of structural information. J Magn
1
H– 1H and 13C– 1H residual dipolar couplings in a chiral liquid crys- Res 1998;133:216–221.

Chirality DOI 10.1002/chir

You might also like