SlideShare a Scribd company logo
1 of 7
Download to read offline
285
NMR, already some 50 years old, has long been an invaluable
analytical method in industry for verification of chemical
synthesis and compound characterisation. The range of
molecular information accessible through NMR, however,
offers a far larger horizon of applications. Of these, ligand
screening by NMR has emerged as a very promising new
method in drug discovery. Its unmatched screening sensitivity,
combined with the abundance of available information on the
structure and nature of molecular binding, justifies the growing
interest in this dynamically expanding NMR application.
Addresses
*NOVASPIN Biotech GmbH, Mühlfeldweg 46, 85748 Garching,
Germany; e-mail: info@novaspin.de
†Institut für Organische Chemie und Biochemie, Technische Universität
München, Lichtenbergstraße 4, 85747 Garching, Germany;
e-mail : kessler@ch.tum.de
Current Opinion in Chemical Biology 2001, 5:285–291
1367-5931/01/$ — see front matter
© 2001 Elsevier Science Ltd. All rights reserved.
Abbreviations
HPLC high-performance liquid chromatography
MAS magic-angle spinning
NOE nuclear Overhauser enhancement
NOESY NOE spectroscopy
RDC residual dipolar couplings
S3 spin-state selection
STD saturation transfer difference
TOCSY total correlation spectroscopy
trNOE transferred intramolecular NOE
TROSY transverse relaxation-optimized spectroscopy
Introduction
NMR has gained widespread acceptance over recent years
as a most versatile tool for industrial drug research. The
relevance of NMR in this regard rests on two pillars — its
use as a powerful, universal and fast-screening technique
to detect intermolecular interactions with unparalleled
sensitivity, and its potential to yield unique information
with atomic resolution to guide structure-based drug
design. This review reflects this dual importance,
focussing not only on NMR screening techniques, but also
including structural applications and a brief survey of
NMR hardware and general methodological developments
that are contributing to advances in the field. The reader is
also referred to other reviews [1–5].
Basic developments in NMR
The application of NMR to industrial drug research is
greatly facilitated by recent hardware developments
(reviewed in [6]). Most important for screening purposes
are developments that reduce sample amounts and dead-
times for sample handling — notorious shortcomings of
‘wasteful’ NMR. Cryoprobe technology can substantially
increase the signal-to-noise ratio, which also translates into
lower accessible binding affinities. Flow probes alleviate the
need for NMR tubes and their time-consuming handling
[7••], enabling direct coupling with separation techniques
such as (HP)LC. Microcoil probes reduce the required
sample volume and also offer superior radio-frequency field
homogeneity [8], benefiting difference-based NMR
screening methods.
Screening techniques are also beginning to benefit from
recent fundamental advances in NMR that have already
proven of value in structure determination. These include
spin-state selection (S3), which is most often employed to
optimise relaxation properties (i.e. in transverse relaxation-
optimized spectroscopy; TROSY [9]), extending molecular
sizes amenable to NMR well beyond 100 kDa, primarily by
enhancing spectral resolution. S3 experiments are also
employed [10] to precisely measure residual dipolar couplings
(RDC) [11], a new set of unique structural constraints that
have proven useful in orienting (hydrogen-poor) ligands
bound to macromolecular targets. RDC result from partial
molecular alignment that can be brought about by adding
magnetically orienting media such as bicelles, filamentous
phages, and others [12]. Connectivities across hydrogen
bonds [13] also yield new structural constraints that can com-
plement the scarce nuclear Overhauser enhancement (NOE)
data in hydrogen-poor systems. Last but not least, various
new strategies for isotopic labelling of proteins, crucial to
NMR, have been developed (for a recent review, see [14]).
General aspects of NMR screening
NMR offers several key advantages for drug research:
1. NMR can detect the weakest ligand–target interactions
of any method so far and thus discover initial hits with
even millimolar binding constants.
2. NMR is a universal screening technique that, unlike
bioassays, requires no knowledge of a protein’s function and
therefore no target-specific set-up. Thus, protein targets that
have been identified purely on a genomics basis and so far lack
a functional bioassay can immediately be screened by NMR.
3. NMR concomitantly enables a determination of
binding constants [15].
4. NMR spectrally separates individual components,
allowing the direct screening and deconvolution of
mixtures from natural sources or combinatorial chemistry.
5. In addition to the mere binary binding information,
NMR returns crucial structural information for both target
and ligand with atomic resolution for the subsequent
optimisation of weak initial hits into strongly binding
drug candidates.
Applications of NMR in drug discovery
Tammo Diercks*, Murray Coles* and Horst Kessler†
NMR screening exploits the fact that many NMR observ-
ables change even upon temporary binding because of
global and local effects (see Figures 1 and 2). The effects
of binding may be monitored either on the target or on the
ligand and are summarised in Table 1. The observed
molecule primarily dictates inherent limitations such as
sample availability (possibly with appropriate isotope
labelling), solubility, stability and molecular size.
Ligand-observed screening
Detection with these techniques generally takes place on the
dissociate, free form of the small ligand (implying a minimal
Kd of ca. 10–7 M), allowing the direct identification of bind-
ing species from mixtures. The unobserved target molecule
is not subject to size limitations and may even be immo-
bilised. In general, isotopic labelling is not required. Many of
these techniques rely on size-dependent effects, which can
be difficult to resolve for weak binding (implying a maximal
Kd of ca. 10–3 M) and small relative molecular size differ-
ences between ligand and target. These effects can be
selected for in 1D filtered spectra, in which tuning the filter
selects for different ligand affinities. Alternatively, the size of
the effect could be evolved in a second indirect dimension,
yielding pseudo-2D edited spectra. The information for
structure-based ligand optimisation obtainable through these
techniques is more limited than for target-observed tech-
niques. It may, however, reveal a ligand’s binding epitope, its
bound conformation, and even contacts to other ligands
binding at adjacent sites.
Use of molecular diffusion
Detection of transiently binding ligands by their decreased
diffusion coefficients is an established screening technique
(for exhaustive reviews see [16,17•]) Pseudo-2D diffusion-
edited experiments (diffusion-ordered spectroscopy,
DOSY), where molecules are separated according to their
individual diffusion coefficients, are particularly useful for
deconvoluting mixtures. Alternatively, 1D diffusion filters
can be combined with standard 2D correlation experi-
ments, such as in the diffusion-modulated gradient COSY
(correlation spectroscopy) [18] and diffusion-encoded total
correlation spectroscopy (TOCSY; DECODES) [19], to
elucidate spin systems and facilitate mixture analysis.
Diffusion filtering or editing might also be enhanced by
combination with other screening techniques, such as
NOE pumping [20,21] (see below). In a promising appli-
cation for combinatorial chemistry, diffusion filtering has
recently been used in solid-state MAS (magic-angle
spinning) NMR to identify and distinguish resin-bound
molecules from impurities [22].
Use of relaxation
Screening based on binding-induced slowing of global
dynamics, leading to a general decrease in NMR relaxation
times, is conceptually very similar to diffusion-based
screening. The diffusion filter is replaced by an inversion-
recovery delay, a spin-lock period or a CPMG echo to filter
for the different types of relaxation times, T1, T1ρ and T2,
respectively. One-dimensional relaxation-based screening
usually requires subtraction of a reference spectrum of the
target-free ligand mixture, and measures must be taken to
avoid artefacts due to binding-induced lineshape distor-
tions, chemical-shift changes and residual target signals
[23]. T2-based ligand screening may also be performed by
comparing spectral linewidths [24•] that increase upon
binding. Complications can arise from superposition of
line-broadening due to chemical exchange or slow
286 Combinatorial chemistry
Figure 1
Global NMR consequences of ligand–target
interactions. Observation is restricted to the
ligand where the effects are most
pronounced. Their strength depends on
molecular size differences and binding
kinetics.
kA
kD
+
Complex properties
similar to
target properties
• Slow Relaxation
• Fast diffusion
• Positive NOE
• Fast relaxation
• Slow diffusion
• Negative NOE
Small ligands ComplexLarge target
Current Opinion in Chemical Biology
Figure 2
Local NMR effects of complex formation may be observed on the
ligand and/or the target. Structural information is available as effects
are mostly localised to the binding site.
Surface protection
Current Opinion in Chemical Biology
Intermolecular
magnetisation
transfer
Chemical shift perturbation
Perturbation of local dynamics{
conformational motions (on the microsecond and
millisecond timescales), which are often relevant for
induced-fit during intermolecular interactions and damped
upon binding. Thus, a size-dependent decrease in T2
times might be compensated for by a flexibility-dependent
reduction in line-broadening.
In contrast, the recently proposed concept of transferred
T1 times [25] relies on the fact that relaxation rates
measured on a fast-exchanging ligand in the free state also
contain contributions from its local dynamics in the bound
state, as immobilisation of flexible regions of the ligand
generally increases relaxation rates. Transferred relaxation
rates may therefore be seen as a dynamic analogue of
transferred NOE (see below). Individual differences in
relaxation rates must be compared with their average in
order to distinguish local from global dynamic changes
[25]. Another complication arises from local relaxation
contributions because of molecular anisotropy.
Intramolecular magnetisation transfer: transfer-NOE
Transferred intramolecular NOEs (trNOEs) are well
established as unique sources of structural information on
the bound ligand (for a review see [26]) where spin-diffu-
sion should be suppressed to obtain optimal structural
precision [27,28]). The observation of trNOE has proved
to be equally useful in the fast screening of ligand
mixtures [24•], forming the basis of ‘bioaffinity NMR’
[29]. The technique relies on the size-dependence of the
intramolecular NOE, which shows slow NOE build-up
with weak positive maxima for free ligands and rapid
build-up with strong negative maxima for the bound state.
If dissociation of the ligand occurs quickly enough (i.e.
Kd > 10–7 M), a sufficient percentage of observable free
ligand will retain intense negative trNOE as a ‘memory’ of
the bound state and thus quickly indicate binding. If the
residence time is too short (i.e. Kd > 10–3 M), however,
trNOE will build up too weakly for detection [30].
Routinely, trNOE are recorded as 2D NOESY (NOE
spectroscopy) spectra with short mixing times favouring
build-up of trNOE over direct NOE. Relaxation filtering
suppresses residual target signals. Notable are applica-
tions of trNOE-based screening [30,31•,32] and recent
experimental improvements [33].
Screening by trNOE imposes upper limits on the ligands
(< 1 kDa), which have to fall into the positive NOE
regime. For borderline cases, this regime may be extend-
ed, for example, by increasing the temperature [31•].
Another problem is the possible cancellation of positive
direct and negative transferred NOE, which could be
resolved by more lengthy 3D experiments such as the 3D
TOCSY–trNOESY [34], or by resorting to other tech-
niques such as saturation transfer difference (see below).
Intermolecular magnetisation transfer
The intermolecular magnetisation transfer is always posi-
tive and cancellation cannot occur. For its measurement,
non-equilibrium magnetisation must first be created asym-
metrically (i.e. on one component only, usually the drug
target). It is then transferred to the binding partner during
a mixing period. The intermolecular magnetisation trans-
fer results in altered signal intensities for both components
and can finally be monitored on either the starting or the
receiving molecule (usually the ligand). Remaining target
resonances can be suppressed (e.g. by isotope filtering or
relaxation filtering). Subtraction of a reference spectrum
(recorded without initially disturbing the magnetisation) is
usually required to reveal these intensity changes and
entails the risk of subtraction artefacts.
Asymmetric non-equilibrium magnetisation on the target
molecule may generally be created by selective irradiation,
requiring the existence of a unique NMR frequency band.
Applications of NMR in drug discovery Diercks, Coles and Kessler 287
Table 1
NMR observable effects of ligand binding.
Global effects of binding Ligand observed Screening Applications References
Translational dynamics Diffusion rates Affinity NMR [17•,19]
Rotational dynamics Relaxation rates
Line broadening [23,24•]
Sign of transferred intramol. NOE Bioaffinity NMR (trNOE) [29,30,31•,32,34]
Molecular orientation Residual dipolar couplings *
Local effects of binding Ligand- or target-observed
Chemical shift perturbation Chemical shifts SAR-by-NMR (HSQC screening) [44•,48,49,51,53•]
Local dynamics perturbation Relaxation rates *
Surface protection Water exchange rates *
Paramagnetic surface probing *
Intermolecular magnetisation transfer Via scalar couplings *
(e.g. intermol. H-bonds)
Via nuclear dipolar couplings Bioaffinity NMR (STD) [32,35,37•]
(intermol. NOE) (Reverse) NOE pumping [20,21]
WaterLOGSY [38]
Via electron dipolar couplings
(paramagnetic relaxation) SLAPSTIC [39•,41•]
*No application to NMR screening reported yet. Intermol., intermolecular.
Initially localised, it is spread across the entire molecule by
spin diffusion such that magnetisation may be picked up
by a ligand binding anywhere on the surface. Alternatively,
non-equilibrium magnetisation may be created symmetri-
cally, followed by asymmetric suppression making use of
the large size difference between target and ligands. For
instance, a diffusion filter would suppress magnetisation
on the ligands, whereas a relaxation filter would suppress
magnetisation on the target.
Saturation transfer difference (STD) [35] as the special
case of intermolecular steady-state NOE is observed after
frequency-selective irradiation of the target and subtrac-
tion of a reference spectrum with off-resonance irradiation.
Although usually run as 1D spectra, STD can be coupled to
standard 2D correlation experiments such as TOCSY, COSY
and HMQC (heteronuclear multiple quantum correlation) to
alleviate spectral overlap [32,35]. The distance-dependence
of the observed signal intensities identifies those nuclei
closest to the target–ligand interface. This ‘epitope mapping’
[28,36] may be seen as a ligand-based complement to the
‘SAR-by-NMR’ approach. A remarkable application extends
STD screening to solid-state NMR [37•], in which the target
was immobilized on controlled-pearl glass, facilitating its
recovery. From a comparison of STD integrals, it was
possible to assess relative binding strengths of the dissolved
ligands. A reference spectrum with pure target-free resin
must rule out direct ligand–resin interactions.
NOE pumping [20] employs the scheme of symmetric
magnetisation inversion, followed by asymmetric suppres-
sion. Classically, the ligand magnetisation is suppressed via
diffusion filtering, and its recovery directly indicates inter-
molecular magnetisation transfer from the target, hence
binding. A suppression of remaining target magnetisation
prior to acquisition greatly facilitates identification, but the
diffusion filter employed may entail serious intensity losses
through concomitant relaxation of the target magnetisation.
This problem is alleviated in the reverse case (reverse
NOE pumping) [21], in which ligand magnetisation is
preserved while target magnetisation is suppressed via
relaxation or isotope filtering. The ligand magnetisation is
then reduced because of transfer onto the target, and signal
reductions observed on a ligand thus indicate binding.
These reductions, however, are only revealed after subtrac-
tion of a reference spectrum, which also takes account of
unspecific magnetisation losses through T1 relaxation.
WaterLOGSY [38] exploits the surface layer of target-
bound water molecules as a pool of selectively invertible
magnetisation that can be transferred onto binding ligands.
After simultaneous selective water suppression and
relaxation filtering (for target suppression), only signals of
binding ligands are detected.
Paramagnetic spin labels
Magnetic spin interactions can be enhanced drastically by
introducing paramagnetic spin labels. The induced NMR
effects — resonance shifts and line broadening — can
thus be observed over larger distances (up to some 20 Å),
shorter contact times, and at lower protein concentrations.
Soluble paramagnetic reagents have classically been
employed to search for metal ion binding sites and probe
solvent-accessible surfaces. Intermolecular binding, which
blocks part of the solvent-accessible surface, may be
detected and localised in a similar manner.
In a recent application to second-site screening, a
confirmed binding ligand was spin labelled and used to
search for ligands concomitantly binding to the target
protein at an adjacent site [39•]. The method is analogous
to the observation of transferred inter-ligand NOE
contacts [40], yet offers larger specificity and sensitivity,
establishing connectivity between more distant binding
sites. The need to synthesize spin-labelled ligands without
affecting their natural binding properties is, however, a
major drawback. A blind test must furthermore rule out
that ligands interact in the absence of the target.
In the SLAPSTIC method, the target protein is spin-
labelled to assist in identifying binding ligands [41•]. The
line-broadening of ligand signals upon binding is thus
strongly emphasized and allows for a reduction in protein
(and ligand) amounts by one or two orders of magnitude. It
is critical to spin-label the target near, but not within the
binding site, as this would invariably lead to altered
binding properties. Moreover, no structural information
can be derived from this method.
Use of residual dipolar couplings
Alignment of the target molecule also causes alignment of
the ligand upon temporary binding. RDC may thus be
transferred from the bound to the isotropic free state if
dissociation is fast enough (Kd in the millimolar range)
[42,43]. In contrast to NOE data, the structural information
available from RDC does not suffer from spin-diffusion
and is therefore precise even for large, immobilised targets.
RDC yield information on both the bound ligand’s
structure and its orientation relative to the target without
the need for assigning target resonances as demonstrated
in two recent applications [42,43]. This technique has not,
however, been applied to ligand screening yet.
Target-observed screening
Observation on the target, rather than on dissociated
ligands, is neither restricted by the ligands’ size nor by an
upper affinity limit. It also immediately reveals different
binding sites, enables direct distinction between specific
and unspecific binding and provides a wealth of structural
information for ligand optimisation. This information,
however, can only be extracted if the spectral assignments
for the target are known. Inherent limitations often
preclude observation on the large target molecule, although
relaxation-optimised techniques (TROSY) may be of
benefit. The amounts of protein required, which must be
isotope-labelled, can be reduced significantly by employing
288 Combinatorial chemistry
cryoprobe technology and screening larger mixtures of
compounds [44•]. Mixtures of ligands can not, however, be
directly deconvoluted.
Chemical-shift perturbations
Most target-observed screening applications rely on
chemical-shift changes as indicators for intermolecular
binding. After assignment, these changes can be mapped
upon the protein structure, revealing its binding sites
[45,46] and guiding structure-based ligand optimisation.
This forms the basis of the ‘SAR-by-NMR’ approach
[47,48], in which optimised ligands are constructed by
linking weakly binding fragments. The approach contin-
ues to be successfully used in drug development [49–52].
Standard screening using 2D 15N,1H correlation spectra
has recently been complemented by 13C,1H HSQC-based
screening on proteins bearing 13C-labeled methyl groups
[53•]. The selective labelling, using inexpensive starting
materials, avoids the detrimental effects of 13C–13C
homonuclear coupling while greatly reducing the cost of
sample preparation. This application benefits from the
higher proton multiplicity and slower transverse relaxation
of the methyl groups. Consequently, intensities were
reported to be on average three-times higher. On the other
hand, screening based on methyl groups suffers from
assignment difficulties and lower spectral dispersion. Note
that the distribution and accessibility of (polar) amide and
(non-polar) methyl groups are not equivalent, possibly
leading to different screening sensitivities.
Equating chemical-shift perturbations with spatial proximity
to the ligand may be misleading, especially in the presence
of large shielding anisotropies in a ligand or because of con-
formational changes in the target. Therefore, an alternative
has been proposed in which the binding site and the bound
ligand’s orientation can be determined more reliably [54]
without the need for a complete re-determination of the
complex structures. It is based on the comparison of chemi-
cal-shift changes in the proximity of the binding site
induced by a series of closely related ligands. This can be
seen as trading synthetic for spectroscopic efforts.
Intermolecular magnetisation transfer
The general scheme for intermolecular magnetisation
transfer experiments (see above) can also be employed for
target-detected screening. In one concept [55], target mag-
netisation is selectively suppressed by relaxation filtering
after symmetric excitation and then recovers by intermolec-
ular transfer from binding ligands. Prior to observation on the
target, the ligand signals are filtered out by selective multiple
frequency suppression. This, however, precludes the direct
screening of ligands with more complex 1H spectra.
Screening for macromolecular interactions
Screening for macromolecular interactions is a special case
in which the ‘ligand’ and ‘target’ have comparable molecu-
lar sizes, and size discrimination is impracticable. Other
than that, many of the techniques mentioned above can be
and have been applied to the problem. For instance, STD
was used to probe protein–nucleic-acid interactions by
saturating well-separated protons of the latter [56]. A varia-
tion of the scheme scans for protein–protein interactions
[57] with molecular distinction made by perdeuterating and
15N-labelling only one partner. The unlabelled protein
remains susceptible to saturation of the aliphatic protons,
while intermolecular STD is observed on the 1H,15N
moieties of the labelled protein. The low proton density in
this perdeuterated species precludes efficient spin-diffu-
sion and the STD is largely confined to the binding site.
Design of NMR screening libraries
In contrast to other screening techniques, NMR screening
requires relatively high sample concentrations and sampling
times. With its unprecedented sensitivity (detecting interac-
tions with Kd up to 10–2 M) and atomic resolution, however,
NMR is particularly suited for guiding the ‘bottom-up’
construction of complex ligands from small, weakly binding
molecular scaffolds. In line with this, the principles for com-
posing a dedicated ligand library are availability, good
solubility, low molecular mass (i.e. < 1–2 kDa), and limita-
tion to some 102–103 compounds of maximal diversity. For
mixture screening, batches should contain (non-reacting)
ligands with isolated NMR signals for easy monitoring.
Various attempts have been made to construct libraries using
consensus fragments identified from statistical analyses of
known drugs (e.g. see [24•,58,59]) and protein-binding
ligands [60], and from fragmenting an existing lead
compound [52]. Pre-selection of small ligands for NMR
screening can also be based on the scoring results obtained
from virtual docking [61].
Conclusions
NMR screening is now being recognised by industry as a
powerful technique for lead discovery. The underlying prin-
ciples have mostly been discovered and developed long ago
by academic scientists, such that the novelty of NMR
screening lies mostly with its applications. In this review, we
have offered a classification of the available NMR methods
and outlined possible future developments by a reduction to
basic NMR properties and principles. The distinction
between ligand- and target-observed screening techniques
is most important. In practice, a dual strategy that combines
the complementary strengths and detectable affinity ranges
of both techniques should prove advantageous. Most impor-
tantly, ligand-observed techniques may produce false
negatives in cases of very strong binding where exchange
between target-bound and detected free ligand is too slow.
These techniques nevertheless have clear advantages for
pre-screening large libraries as they implicitly deconvolute
ligand mixtures, require no isotope labelling and pose few
limitations on the target. Consecutive target-observed ‘fine-
’screening of the identified binding ligands can then rule out
unspecific binding, identify different binding sites and
modes, and guide structure-based ligand optimisation. By
Applications of NMR in drug discovery Diercks, Coles and Kessler 289
screening a carefully composed small library of basic
molecular scaffolds, the pre-screening step may be omitted
for amenable targets.
References and recommended reading
Papers of particular interest, published within the annual period of review,
have been highlighted as:
• of special interest
••of outstanding interest
1. Moore JM: NMR screening in drug discovery. Curr Opin Biotechnol
1999, 10:54-58.
2. Moore JM: NMR techniques for characterization of ligand binding:
utility for lead generation and optimization in drug discovery.
Biopolymers 1999, 51:221-243.
3. Roberts GCK: NMR spectroscopy in structure-based drug design.
Curr Opin Biotechnol 1999, 10:42-47.
4. Roberts GCK: Applications of NMR in drug discovery. Drug Discov
Today 2000, 5:230-240.
5. Stockman BJ: NMR spectroscopy as a tool for structure-based
drug design. Progr NMR Spectrosc 1998, 33:109-151.
6. Keifer PA: NMR tools for biotechnology. Curr Opin Biotechnol
1999, 10:34-41.
7. Ross A, Schlotterbeck G, Klaus W, Senn H: Automation of NMR
•• measurements and data evaluation for systematically screening
interactions of small molecules with target proteins. J Biomol
NMR 2000, 16:139-146.
This reference gives valuable practical advice on performing industrial-scale
ligand screening by NMR, presenting a fully integrated hardware set-up with
robotized sample preparation, flow-through probehead and automated data
analysis of the acquired 15N,1H-HSQC. A pre-check of the protein–ligand
mixtures for aggregation and solubility is required to prevent clogging of the
flow-through probe. The authors conclude that screening individual ligands,
rather than mixtures, justifies the extra time by alleviating the need for char-
acterizing possible ligand–ligand interactions and deconvoluting mixtures.
The problem of using too low buffer concentrations, entailing artefactual
chemical shift changes, is also discussed.
8. Haner RL, Llanos W, Mueller L: Small volume flow probe for
automated direct-injection NMR analysis: design and
performance. J Mag Res 2000, 143:69-78.
9. Pervushin K, Riek R, Wider G, Wüthrich K: Attenuated T2 relaxation
by mutual cancellation of dipole-dipole coupling and chemical
shift anisotropy indicates an avenue to NMR structures of very
large biological macromolecules in solution. Proc Natl Acad Sci
USA 1997, 94:12366-12371.
10. Permi P, Annila A: Transverse relaxation optimised spin-state
selective NMR experiments for measurement of residual dipolar
couplings. J Biomol NMR 2000, 16:221-227.
11. Prestegard JH: New techniques in structural NMR — anisotropic
interactions. Nat Struct Biol 1998, (NMR suppl):517-522.
12. Rückert M, Otting G: Alignment of biological macromolecules in
novel nonionic liquid crystalline media for NMR experiments.
J Am Chem Soc 2000, 122:7793-7797.
13. Cordier F, Grzesiek S: Direct observation of hydrogen bonds in
proteins by interresidue h3JNC’ scalar couplings. J Am Chem Soc
1999, 121:1601-1602.
14. Goto NK, Kay LE: New developments in isotope labeling
strategies for protein solution NMR spectroscopy. Curr Opin
Struct Biol 2000, 10:585-592.
15. Fielding L: Determination of association constants (Ka) from
solution NMR data. Tetrahedron 2000, 56:6151-6170.
16. Johnson CS: Diffusion ordered nuclear magnetic resonance
spectroscopy: principles and applications. Progr NMR Spectrosc
1999, 34:203-256.
17. Chen A, Shapiro MJ: Affinity NMR. Anal Chem 1999,
• 71:669A-675A.
A review describing NMR screening, in particular diffusion-based affinity
NMR, in a modular way.
18. Gmeiner WH, Hudalla CJ, Soto AM, Marky L: Binding of ethidium to
DNA measured using a 2D diffusion-modulated gradient COSY
NMR experiment. FEBS Lett 2000, 465:148-152.
19. Bleicher K, Lin M, Shapiro MJ, Wareing JR: Diffusion edited NMR:
screening compound mixtures by affinity NMR to detect binding
ligands to vancomycin. J Org Chem 1998, 63:8486-8490.
20. Chen A, Shapiro MJ: NOE pumping: a novel NMR technique for
identification of compounds with binding affinity to
macromolecules. J Am Chem Soc 1998, 120:10258-10259.
21. Chen A, Shapiro MJ: NOE pumping. 2. A high-throughput method
to determine compounds with binding affinity to macromolecules
by NMR. J Am Chem Soc 2000, 122:414-415.
22. Shapiro MJ, Chin J, Chen A, Wareing JR, Tang Q, Tommasi RA,
Marepalli HR: Covalent or trapped? PFG diffusion MAS NMR for
combinatorial chemistry. Tetrahedron Lett 1999, 40:6141-6143.
23. Hajduk PJ, Olejniczak ET, Fesik SW: One-dimensional relaxation-
and diffusion-edited NMR methods for screening compounds that
bind to macromolecules. J Am Chem Soc 1997, 119:12257-12261.
24. Fejzo J, Lepre CA, Peng JW, Bemis GW, Ajay, Murcko MA, Moore JM:
• The SHAPES strategy: An NMR-based approach for lead
generation in drug discovery. Chem Biol 1999, 6:755-769.
Although no new techniques are described, a clear overview on the set-up
of NMR screening, including library design is provided.
25. LaPlante SR, Aubry N, Deziel R, Ni F, Xu P: Transferred 13C T1
relaxation at natural isotopic abundance: a practical method for
determining site-specific changes in ligand flexibility upon
binding to a macromolecule. J Am Chem Soc 2000,
122:12530-12535.
26. Ni F: Recent developments in transferred NOE methods. Progr
NMR Spectrosc 1994, 26:517-606.
27. Vincent SJF, Zwahlen C, Post CB, Burgner JW, Bodenhausen G: The
conformation of NAD++ bound to lactate dehydrogenase
determined by nuclear magnetic resonance with suppression of
spin-diffusion. Proc Natl Acad Sci USA 1997, 94:4383-4388.
28. Maaheimo H, Kosma P, Brade L, Brade H, Peters T: Mapping the
binding of synthetic disaccharides representing epitopes of
chlamydial lipopolysaccharide to antibodies with NMR.
Biochemistry 2000, 39:12778-12788.
29. Meyer B, Weimar T, Peters T: Screening mixtures for biological
activity by NMR. Eur J Biochem 1997, 246:705-709.
30. Mayer M, Meyer M: Mapping the active site of angiotensin-
converting enzyme by transferred NOE spectroscopy. J Med
Chem 2000, 43:2093-2099.
31. Henrichsen D, Ernst B, Magnani JL, Wang W-T, Meyer B, Peters T:
• Bioaffinity NMR spectroscopy: identification of an E-selectin
antagonist in a substance mixture by transfer NOE. Angew Chem
Int Ed Engl 1999, 38:98-102.
This article describes an application of trNOE-based screening to a mixture
of ligands, also discussing important details to be considered with this tech-
nique. Prior to screening, the library was tested for the positive NOE regime,
which was eventually reached only after increasing the temperature. The
binding ligand then identified by negative trNOE displayed strong spin-
diffusion, which in this case helped its identification, but generally blurs
structural information. Suppression of residual target protein signals by
means of a relaxation filter was largely complete, except for signals from
a highly flexible, attached carbohydrate chain.
32. Vogtherr M, Peters T: Application of NMR based binding assays to
identify key hydroxy groups for intermolecular recognition. J Am
Chem Soc 2000, 122:6093-6099.
33. Weimar T: Improving bioaffinity NMR spectra by means of zero-
quantum dephasing. Magn Reson Chem 2000, 38:315-318.
34. Herfurth L, Weimar T, Peters T: Application of 3D-TOCSY-tr-NOESY
for the assignment of bioactive ligands from mixtures. Angew
Chem Int Ed Engl 2000, 39:2097-2099.
35. Mayer M, Meyer B: Characterization of ligand binding by saturation
transfer difference NMR spectroscopy. Angew Chem Int Ed Engl
1999, 38:1784-1788.
36. Mayer M, Meyer B: Group epitope mapping (GEM) to identify
segments of a ligand in direct contact with a protein: STD NMR as
a tool to characterize binding to Ricinus communis agglutinin.
J Am Chem Soc 2001, in press.
290 Combinatorial chemistry
37. Klein J, Meinecke R, Mayer M, Meyer B: Detecting binding affinity to
• immobilized receptor proteins in compound libraries by HR-MAS
STD NMR. J Am Chem Soc 1999, 121:5336-5337.
This article describes an exciting extension of the STD screening method to
high-resolution MAS NMR, using dissolved ligands and a target immobilized
on controlled pearl glass for easy protein recovery. A reference spectrum
with pure, target-free resin must be recorded to rule out ligand–resin inter-
actions. Relative binding strengths of competitive ligands can be assessed
from a comparison of STD integrals. Structural information, however, is
restricted to the binding epitope of the ligand.
38. Dalvit C, Pevarello P, Tato M, Veronesi M, Vulpetti A, Sundström M:
Identification of compounds with binding affinity to proteins via
magnetization transfer from bulk water. J Biomol NMR 2000, 18:65-68.
39. Jahnke W, Perez LB, Paris CG, Strauss A, Fendrich G, Nalin CM:
• Second-site NMR screening with a spin-labelled first ligand. J Am
Chem Soc 2000, 122:7394-7395.
This interesting alternative to the so-called ‘SAR-by-NMR’ approach uses a
spin-labelled known binding ligand to induce detectable signal broadening
or extinction on another ligand binding to an adjacent site on the target pro-
tein. This novel method for the exclusive observation of second-site binding
is highly sensitive and also provides some structural information.
40. Li D, DeRose EF, London RE: The inter-ligand Overhause effect: a
powerful new NMR approach for mapping structural relationships
of macromolecular ligands. J Biomol NMR 1999, 15:71-76.
41. Jahnke W, Rüdisser S, Zurini M: Spin label enhanced NMR
• screening. J Am Chem Soc 2001, 123:3149-315.
This article describes a novel technique, SLAPSTIC, which uses a modified,
spin-labelled target protein for ligand screening. Ligand binding is detected
by signal broadening and extinction on the free form of a temporarily binding
ligand. The strong magnetic marker allows a drastic reduction in the required
protein and ligand concentrations, which also translates into lower minimum
ligand solubilities and access to lower binding affinities.
42. Koenig BW, Mitchell DC, König S, Grzesiek S, Litman BJ, Bax A:
Measurement of dipolar couplings in a transducin peptide
fragment weakly bound to oriented photo-activated rhodopsin.
J Biomol NMR 2000, 16:121-125.
43. Thompson GS, Shimizu H, Homans SW, Donohue-Rolfe A:
Localization of the binding site for the oligosaccharide moiety of
GB3 on verotoxin 1 using NMR residual dipolar coupling
measurements. Biochemistry 2000, 39:13153-13156.
44. Hajduk PJ, Gerfin T, Boehlen J-M, Häberli M, Marek D, Fesik SW:
• High-throughput nuclear magnetic resonance-based screening.
J Med Chem 1999, 42:2315-2317.
This article presents a profound analysis of the benefits of cryoprobe tech-
nology to 15N-HSQC-based NMR screening. The merits alternatively trans-
late into lower detectable ligand affinities (with up to millimolar dissociation
rates), substantially reduced protein and ligand concentrations, or faster
screening rates. The authors claim that libraries with up to 200,000 ligands
(administered in mixtures of 100 compounds) could thus be screened effi-
ciently within less than a month.
45. Sakamoto T, Tanaka T, Ito Y, Rajesh S, Iwamoto-Sugai M, Kodera Y,
Tsuchida M, Shibata T, Kohno T: An NMR analysis of ubiquitin
recognition by yeast ubiquitin hydrolase: evidence for novel
substrate recognition by a cysteine protease. Biochemistry 1999,
38:11634-11642.
46. Sun C, Cai M, Gunasekera AH, Meadows RP, Wang H, Chen J, Zhang H,
Wu W, Xu N, Ng SC et al.: NMR structure and mutagenesis of the
inhibitor-of-apoptosis protein XIAP. Nature 1999, 401:818-822.
47. Hajduk PJ, Sheppard G, Nettesheim D, Olejniczak ET, Shuker SB,
Meadows RP, Steinman DH, Carrera GM, Marcotte PA, Severin J
et al.: Discovery of potent nonpeptide inhibitors of
stromelysin using SAR by NMR. J Am Chem Soc 1997,
119:5818-5827.
48. Shuker SB, Hajduk PJ, Meadows RP, Fesik SW: Discovering high-
affinity ligands for proteins: SAR by NMR. Science 1996,
274:1531-1534.
49. Hajduk PJ, Zhou M-M, Fesik SW: NMR-based discovery of
phosphotyrosine mimetics that bind to the Lck SH2 domain.
Bioorg Med Chem Lett 1999, 9:2403-2406.
50. Hajduk PJ, Dinges J, Schkeryantz JM, Janowick D, Kaminski M,
Tufano M, Augeri DJ, Petros A, Nienaber V, Zhong P et al.: Novel
inhibitors of Erm methyltransferases from NMR and parallel
synthesis. J Med Chem 1999, 42:3852-3859.
51. Hajduk PJ, Boyd S, Nettesheim D, Nienaber V, Severin J, Smith R,
Davidson D, Rockway T, Fesik SW: Identification of novel inhibitors
of urokinase via NMR-based screening. J Am Chem Soc 2000,
43:3862-3866.
52. Hajduk PJ, Gomtsyan A, Didomenico S, Cowart M, Bayburt EK,
Solomon L, Severin J, Smith R, Walter K, Holzman TF et al.: Design of
adenosine kinase inhibitors from the NMR-based screening of
fragments. J Med Chem 2000, 43:4781-4786.
53. Hajduk PJ, Augeri DJ, Mack J, Mendoza R, Yang J, Betz SF, Fesik SW:
• NMR-based screening of proteins containing 13C-labeled methyl
groups. J Am Chem Soc 2000, 122:7898-7904.
This article evaluates the advantages of 13C-HSQC-based screening on a
target protein selectively 13C-labelled at the methyl groups, including a com-
parison with 15N-HSQC- and 15N-TROSY-based screening.
54. Medek A, Hajduk PJ, Mack J, Fesik SW: The use of differential
chemical shifts for determining the binding site location and
orientation of protein-bound ligands. J Am Chem Soc 2000,
122:1241-1242.
55. Dalvit C, Floersheim P, Zurini M, Widmer A: Use of organic solvents
and small molecules for locating binding sites on proteins in
solution. J Biomol NMR 1999, 14:23-32.
56. Ramos A, Kelly G, Hollingworth D, Pastore A, Frenkiel T: Mapping the
interface of protein-nucleic acid complexes using cross-
saturation. J Am Chem Soc 2000, 122:11311-11314.
57. Takahashi H, Nakanishi T, Kami K, Arata Y, Shimada I: A novel NMR
method for determining the interfaces of large protein–protein
complexes. Nat Struct Biol 2000, 7:220-223.
58. Bemis GW, Murcko MA: The properties of known drugs. 1.
Molecular frameworks. J Med Chem 1996, 39:2887-2893.
59. Bemis GW, Murcko MA: Properties of known drugs. 2. Side chains.
J Med Chem 1999, 42:5095-5099.
60. Hajduk PJ, Bures M, Praestgaard J, Fesik SW: Privileged molecules
for protein binding identified from NMR-based screening. J Med
Chem 2000, 43:3443-3447.
61. Muegge I, Martin YC, Hajduk PJ, Fesik SW: Evaluation of PMF
scoring in docking weak ligands to the FK506 binding protein.
J Med Chem 1999, 42:2498-2503.
Applications of NMR in drug discovery Diercks, Coles and Kessler 291

More Related Content

What's hot

Gel matrix dependence on the dose response properties and diffusion phenomena...
Gel matrix dependence on the dose response properties and diffusion phenomena...Gel matrix dependence on the dose response properties and diffusion phenomena...
Gel matrix dependence on the dose response properties and diffusion phenomena...
inventionjournals
 
SPECTROSCOPIC METHOD DEVELOPMENT FOR LOSARTON POTASSIUM IN TABLET By Ninad M...
SPECTROSCOPIC METHOD DEVELOPMENT FOR LOSARTON POTASSIUM IN TABLET  By Ninad M...SPECTROSCOPIC METHOD DEVELOPMENT FOR LOSARTON POTASSIUM IN TABLET  By Ninad M...
SPECTROSCOPIC METHOD DEVELOPMENT FOR LOSARTON POTASSIUM IN TABLET By Ninad M...
Ninad Phadke
 
Simultaneous loading of 5-florouracil and SPIONs in HSA nanoparticles: Optimi...
Simultaneous loading of 5-florouracil and SPIONs in HSA nanoparticles: Optimi...Simultaneous loading of 5-florouracil and SPIONs in HSA nanoparticles: Optimi...
Simultaneous loading of 5-florouracil and SPIONs in HSA nanoparticles: Optimi...
Nanomedicine Journal (NMJ)
 

What's hot (19)

CADD
CADDCADD
CADD
 
Gel matrix dependence on the dose response properties and diffusion phenomena...
Gel matrix dependence on the dose response properties and diffusion phenomena...Gel matrix dependence on the dose response properties and diffusion phenomena...
Gel matrix dependence on the dose response properties and diffusion phenomena...
 
MAGNETICALLY MODULATED DRUG DELIVERY SYSTEMS
MAGNETICALLY MODULATED DRUG DELIVERY SYSTEMSMAGNETICALLY MODULATED DRUG DELIVERY SYSTEMS
MAGNETICALLY MODULATED DRUG DELIVERY SYSTEMS
 
De novo drug design
De novo drug designDe novo drug design
De novo drug design
 
Cadd
CaddCadd
Cadd
 
Structure based in silico virtual screening
Structure based in silico virtual screeningStructure based in silico virtual screening
Structure based in silico virtual screening
 
Drug Discovery Today: Fighting TB with Technology
Drug Discovery Today: Fighting TB with TechnologyDrug Discovery Today: Fighting TB with Technology
Drug Discovery Today: Fighting TB with Technology
 
Computer Aided Drug Design ppt
Computer Aided Drug Design pptComputer Aided Drug Design ppt
Computer Aided Drug Design ppt
 
SPECTROSCOPIC METHOD DEVELOPMENT FOR LOSARTON POTASSIUM IN TABLET By Ninad M...
SPECTROSCOPIC METHOD DEVELOPMENT FOR LOSARTON POTASSIUM IN TABLET  By Ninad M...SPECTROSCOPIC METHOD DEVELOPMENT FOR LOSARTON POTASSIUM IN TABLET  By Ninad M...
SPECTROSCOPIC METHOD DEVELOPMENT FOR LOSARTON POTASSIUM IN TABLET By Ninad M...
 
Fragment based drug design
Fragment based drug designFragment based drug design
Fragment based drug design
 
UV-vis. spectroscopy N HPLC (rilpivirine) by RJcharan.
UV-vis. spectroscopy N HPLC (rilpivirine) by RJcharan.UV-vis. spectroscopy N HPLC (rilpivirine) by RJcharan.
UV-vis. spectroscopy N HPLC (rilpivirine) by RJcharan.
 
Superimposition method- ligand based drug design
Superimposition method- ligand based drug designSuperimposition method- ligand based drug design
Superimposition method- ligand based drug design
 
Presentation (1)
Presentation (1)Presentation (1)
Presentation (1)
 
Simultaneous loading of 5-florouracil and SPIONs in HSA nanoparticles: Optimi...
Simultaneous loading of 5-florouracil and SPIONs in HSA nanoparticles: Optimi...Simultaneous loading of 5-florouracil and SPIONs in HSA nanoparticles: Optimi...
Simultaneous loading of 5-florouracil and SPIONs in HSA nanoparticles: Optimi...
 
In silico drug design/Molecular docking
In silico drug design/Molecular dockingIn silico drug design/Molecular docking
In silico drug design/Molecular docking
 
Image analysis and Laser Particle Diffraction study of ProRoot-MTA, Portland ...
Image analysis and Laser Particle Diffraction study of ProRoot-MTA, Portland ...Image analysis and Laser Particle Diffraction study of ProRoot-MTA, Portland ...
Image analysis and Laser Particle Diffraction study of ProRoot-MTA, Portland ...
 
In Silico methods for ADMET prediction of new molecules
 In Silico methods for ADMET prediction of new molecules In Silico methods for ADMET prediction of new molecules
In Silico methods for ADMET prediction of new molecules
 
Denovo Drug Design
Denovo Drug DesignDenovo Drug Design
Denovo Drug Design
 
Cadd and molecular modeling for M.Pharm
Cadd and molecular modeling for M.PharmCadd and molecular modeling for M.Pharm
Cadd and molecular modeling for M.Pharm
 

Viewers also liked (14)

Presentacion blady
Presentacion bladyPresentacion blady
Presentacion blady
 
Power point informtica
Power point informticaPower point informtica
Power point informtica
 
Tabajo powerpoint de tic nuevo
Tabajo powerpoint de tic  nuevoTabajo powerpoint de tic  nuevo
Tabajo powerpoint de tic nuevo
 
Tratamiento de imagen
Tratamiento de imagenTratamiento de imagen
Tratamiento de imagen
 
Tabajo powerpoint de tic nuevo
Tabajo powerpoint de tic  nuevoTabajo powerpoint de tic  nuevo
Tabajo powerpoint de tic nuevo
 
Sports Nutrition_Chapter 3_Carbohydrates
Sports Nutrition_Chapter 3_CarbohydratesSports Nutrition_Chapter 3_Carbohydrates
Sports Nutrition_Chapter 3_Carbohydrates
 
Presentacion blady
Presentacion bladyPresentacion blady
Presentacion blady
 
Presentación1(trabajo de tecnologia ) javier justavino
Presentación1(trabajo de tecnologia ) javier justavinoPresentación1(trabajo de tecnologia ) javier justavino
Presentación1(trabajo de tecnologia ) javier justavino
 
Sports Nutrition_Chapter 2_Nutrients
Sports Nutrition_Chapter 2_NutrientsSports Nutrition_Chapter 2_Nutrients
Sports Nutrition_Chapter 2_Nutrients
 
Sports Nutrition_Chapter 5_Protein
Sports Nutrition_Chapter 5_ProteinSports Nutrition_Chapter 5_Protein
Sports Nutrition_Chapter 5_Protein
 
Conservation, distribution and types of forests and wildlife
Conservation, distribution and types of forests and wildlifeConservation, distribution and types of forests and wildlife
Conservation, distribution and types of forests and wildlife
 
Computers
ComputersComputers
Computers
 
Sound
SoundSound
Sound
 
Camouflage
Camouflage Camouflage
Camouflage
 

Similar to วิจัยต้นอ้อ

Some Biophysical Methods for Demonstrating Comparability of Conformation and ...
Some Biophysical Methods for Demonstrating Comparability of Conformation and ...Some Biophysical Methods for Demonstrating Comparability of Conformation and ...
Some Biophysical Methods for Demonstrating Comparability of Conformation and ...
KBI Biopharma
 
Medicamente si abuzul de medicamente.pdf
Medicamente si abuzul de medicamente.pdfMedicamente si abuzul de medicamente.pdf
Medicamente si abuzul de medicamente.pdf
Carmen180502
 
ACSPosterMurphyBrownPradel2015
ACSPosterMurphyBrownPradel2015ACSPosterMurphyBrownPradel2015
ACSPosterMurphyBrownPradel2015
Megan Murphy
 

Similar to วิจัยต้นอ้อ (20)

SAR BY NMR (Structure Activity Relationship by Using NMR)
SAR BY NMR (Structure Activity Relationship by Using NMR)SAR BY NMR (Structure Activity Relationship by Using NMR)
SAR BY NMR (Structure Activity Relationship by Using NMR)
 
Nuclear magnetic resonance (enzymology)
Nuclear magnetic resonance (enzymology) Nuclear magnetic resonance (enzymology)
Nuclear magnetic resonance (enzymology)
 
Fragment screening library workshop (IQPC 2008)
Fragment screening library workshop (IQPC 2008)Fragment screening library workshop (IQPC 2008)
Fragment screening library workshop (IQPC 2008)
 
Characterizing aptamer small molecule interactions with back-scattering inter...
Characterizing aptamer small molecule interactions with back-scattering inter...Characterizing aptamer small molecule interactions with back-scattering inter...
Characterizing aptamer small molecule interactions with back-scattering inter...
 
Some Biophysical Methods for Demonstrating Comparability of Conformation and ...
Some Biophysical Methods for Demonstrating Comparability of Conformation and ...Some Biophysical Methods for Demonstrating Comparability of Conformation and ...
Some Biophysical Methods for Demonstrating Comparability of Conformation and ...
 
Crimson Publishers-Potential Application of Raman Micro-Spectroscopy as an In...
Crimson Publishers-Potential Application of Raman Micro-Spectroscopy as an In...Crimson Publishers-Potential Application of Raman Micro-Spectroscopy as an In...
Crimson Publishers-Potential Application of Raman Micro-Spectroscopy as an In...
 
Nuclear Magnetic Resonance
Nuclear Magnetic ResonanceNuclear Magnetic Resonance
Nuclear Magnetic Resonance
 
Solid state NMR Princple and application .
Solid state NMR Princple and application . Solid state NMR Princple and application .
Solid state NMR Princple and application .
 
Medicamente si abuzul de medicamente.pdf
Medicamente si abuzul de medicamente.pdfMedicamente si abuzul de medicamente.pdf
Medicamente si abuzul de medicamente.pdf
 
Characterization Techniques for Nanoparticles
Characterization Techniques for NanoparticlesCharacterization Techniques for Nanoparticles
Characterization Techniques for Nanoparticles
 
Proteomics Practical (NMR and Protein 3D software
Proteomics Practical (NMR and Protein 3D softwareProteomics Practical (NMR and Protein 3D software
Proteomics Practical (NMR and Protein 3D software
 
Structural proteomics
Structural proteomicsStructural proteomics
Structural proteomics
 
2D NMR.pptx
2D NMR.pptx2D NMR.pptx
2D NMR.pptx
 
Poster
Poster Poster
Poster
 
J.1747 0285.2009.00940.x
J.1747 0285.2009.00940.xJ.1747 0285.2009.00940.x
J.1747 0285.2009.00940.x
 
3d qsar
3d qsar3d qsar
3d qsar
 
Pharmacophore Modeling and Docking Techniques.ppt
Pharmacophore Modeling and Docking Techniques.pptPharmacophore Modeling and Docking Techniques.ppt
Pharmacophore Modeling and Docking Techniques.ppt
 
ACSPosterMurphyBrownPradel2015
ACSPosterMurphyBrownPradel2015ACSPosterMurphyBrownPradel2015
ACSPosterMurphyBrownPradel2015
 
Mayuri shitre
Mayuri shitreMayuri shitre
Mayuri shitre
 
Design of fragment screening libraries (IQPC 2008)
Design of fragment screening libraries (IQPC 2008)Design of fragment screening libraries (IQPC 2008)
Design of fragment screening libraries (IQPC 2008)
 

Recently uploaded

Recently uploaded (20)

04-2024-HHUG-Sales-and-Marketing-Alignment.pptx
04-2024-HHUG-Sales-and-Marketing-Alignment.pptx04-2024-HHUG-Sales-and-Marketing-Alignment.pptx
04-2024-HHUG-Sales-and-Marketing-Alignment.pptx
 
Workshop - Best of Both Worlds_ Combine KG and Vector search for enhanced R...
Workshop - Best of Both Worlds_ Combine  KG and Vector search for  enhanced R...Workshop - Best of Both Worlds_ Combine  KG and Vector search for  enhanced R...
Workshop - Best of Both Worlds_ Combine KG and Vector search for enhanced R...
 
GenCyber Cyber Security Day Presentation
GenCyber Cyber Security Day PresentationGenCyber Cyber Security Day Presentation
GenCyber Cyber Security Day Presentation
 
A Year of the Servo Reboot: Where Are We Now?
A Year of the Servo Reboot: Where Are We Now?A Year of the Servo Reboot: Where Are We Now?
A Year of the Servo Reboot: Where Are We Now?
 
Boost PC performance: How more available memory can improve productivity
Boost PC performance: How more available memory can improve productivityBoost PC performance: How more available memory can improve productivity
Boost PC performance: How more available memory can improve productivity
 
Handwritten Text Recognition for manuscripts and early printed texts
Handwritten Text Recognition for manuscripts and early printed textsHandwritten Text Recognition for manuscripts and early printed texts
Handwritten Text Recognition for manuscripts and early printed texts
 
Automating Google Workspace (GWS) & more with Apps Script
Automating Google Workspace (GWS) & more with Apps ScriptAutomating Google Workspace (GWS) & more with Apps Script
Automating Google Workspace (GWS) & more with Apps Script
 
Powerful Google developer tools for immediate impact! (2023-24 C)
Powerful Google developer tools for immediate impact! (2023-24 C)Powerful Google developer tools for immediate impact! (2023-24 C)
Powerful Google developer tools for immediate impact! (2023-24 C)
 
🐬 The future of MySQL is Postgres 🐘
🐬  The future of MySQL is Postgres   🐘🐬  The future of MySQL is Postgres   🐘
🐬 The future of MySQL is Postgres 🐘
 
08448380779 Call Girls In Greater Kailash - I Women Seeking Men
08448380779 Call Girls In Greater Kailash - I Women Seeking Men08448380779 Call Girls In Greater Kailash - I Women Seeking Men
08448380779 Call Girls In Greater Kailash - I Women Seeking Men
 
Finology Group – Insurtech Innovation Award 2024
Finology Group – Insurtech Innovation Award 2024Finology Group – Insurtech Innovation Award 2024
Finology Group – Insurtech Innovation Award 2024
 
The 7 Things I Know About Cyber Security After 25 Years | April 2024
The 7 Things I Know About Cyber Security After 25 Years | April 2024The 7 Things I Know About Cyber Security After 25 Years | April 2024
The 7 Things I Know About Cyber Security After 25 Years | April 2024
 
2024: Domino Containers - The Next Step. News from the Domino Container commu...
2024: Domino Containers - The Next Step. News from the Domino Container commu...2024: Domino Containers - The Next Step. News from the Domino Container commu...
2024: Domino Containers - The Next Step. News from the Domino Container commu...
 
08448380779 Call Girls In Diplomatic Enclave Women Seeking Men
08448380779 Call Girls In Diplomatic Enclave Women Seeking Men08448380779 Call Girls In Diplomatic Enclave Women Seeking Men
08448380779 Call Girls In Diplomatic Enclave Women Seeking Men
 
Real Time Object Detection Using Open CV
Real Time Object Detection Using Open CVReal Time Object Detection Using Open CV
Real Time Object Detection Using Open CV
 
From Event to Action: Accelerate Your Decision Making with Real-Time Automation
From Event to Action: Accelerate Your Decision Making with Real-Time AutomationFrom Event to Action: Accelerate Your Decision Making with Real-Time Automation
From Event to Action: Accelerate Your Decision Making with Real-Time Automation
 
Apidays Singapore 2024 - Building Digital Trust in a Digital Economy by Veron...
Apidays Singapore 2024 - Building Digital Trust in a Digital Economy by Veron...Apidays Singapore 2024 - Building Digital Trust in a Digital Economy by Veron...
Apidays Singapore 2024 - Building Digital Trust in a Digital Economy by Veron...
 
Data Cloud, More than a CDP by Matt Robison
Data Cloud, More than a CDP by Matt RobisonData Cloud, More than a CDP by Matt Robison
Data Cloud, More than a CDP by Matt Robison
 
Raspberry Pi 5: Challenges and Solutions in Bringing up an OpenGL/Vulkan Driv...
Raspberry Pi 5: Challenges and Solutions in Bringing up an OpenGL/Vulkan Driv...Raspberry Pi 5: Challenges and Solutions in Bringing up an OpenGL/Vulkan Driv...
Raspberry Pi 5: Challenges and Solutions in Bringing up an OpenGL/Vulkan Driv...
 
Understanding Discord NSFW Servers A Guide for Responsible Users.pdf
Understanding Discord NSFW Servers A Guide for Responsible Users.pdfUnderstanding Discord NSFW Servers A Guide for Responsible Users.pdf
Understanding Discord NSFW Servers A Guide for Responsible Users.pdf
 

วิจัยต้นอ้อ

  • 1. 285 NMR, already some 50 years old, has long been an invaluable analytical method in industry for verification of chemical synthesis and compound characterisation. The range of molecular information accessible through NMR, however, offers a far larger horizon of applications. Of these, ligand screening by NMR has emerged as a very promising new method in drug discovery. Its unmatched screening sensitivity, combined with the abundance of available information on the structure and nature of molecular binding, justifies the growing interest in this dynamically expanding NMR application. Addresses *NOVASPIN Biotech GmbH, Mühlfeldweg 46, 85748 Garching, Germany; e-mail: info@novaspin.de †Institut für Organische Chemie und Biochemie, Technische Universität München, Lichtenbergstraße 4, 85747 Garching, Germany; e-mail : kessler@ch.tum.de Current Opinion in Chemical Biology 2001, 5:285–291 1367-5931/01/$ — see front matter © 2001 Elsevier Science Ltd. All rights reserved. Abbreviations HPLC high-performance liquid chromatography MAS magic-angle spinning NOE nuclear Overhauser enhancement NOESY NOE spectroscopy RDC residual dipolar couplings S3 spin-state selection STD saturation transfer difference TOCSY total correlation spectroscopy trNOE transferred intramolecular NOE TROSY transverse relaxation-optimized spectroscopy Introduction NMR has gained widespread acceptance over recent years as a most versatile tool for industrial drug research. The relevance of NMR in this regard rests on two pillars — its use as a powerful, universal and fast-screening technique to detect intermolecular interactions with unparalleled sensitivity, and its potential to yield unique information with atomic resolution to guide structure-based drug design. This review reflects this dual importance, focussing not only on NMR screening techniques, but also including structural applications and a brief survey of NMR hardware and general methodological developments that are contributing to advances in the field. The reader is also referred to other reviews [1–5]. Basic developments in NMR The application of NMR to industrial drug research is greatly facilitated by recent hardware developments (reviewed in [6]). Most important for screening purposes are developments that reduce sample amounts and dead- times for sample handling — notorious shortcomings of ‘wasteful’ NMR. Cryoprobe technology can substantially increase the signal-to-noise ratio, which also translates into lower accessible binding affinities. Flow probes alleviate the need for NMR tubes and their time-consuming handling [7••], enabling direct coupling with separation techniques such as (HP)LC. Microcoil probes reduce the required sample volume and also offer superior radio-frequency field homogeneity [8], benefiting difference-based NMR screening methods. Screening techniques are also beginning to benefit from recent fundamental advances in NMR that have already proven of value in structure determination. These include spin-state selection (S3), which is most often employed to optimise relaxation properties (i.e. in transverse relaxation- optimized spectroscopy; TROSY [9]), extending molecular sizes amenable to NMR well beyond 100 kDa, primarily by enhancing spectral resolution. S3 experiments are also employed [10] to precisely measure residual dipolar couplings (RDC) [11], a new set of unique structural constraints that have proven useful in orienting (hydrogen-poor) ligands bound to macromolecular targets. RDC result from partial molecular alignment that can be brought about by adding magnetically orienting media such as bicelles, filamentous phages, and others [12]. Connectivities across hydrogen bonds [13] also yield new structural constraints that can com- plement the scarce nuclear Overhauser enhancement (NOE) data in hydrogen-poor systems. Last but not least, various new strategies for isotopic labelling of proteins, crucial to NMR, have been developed (for a recent review, see [14]). General aspects of NMR screening NMR offers several key advantages for drug research: 1. NMR can detect the weakest ligand–target interactions of any method so far and thus discover initial hits with even millimolar binding constants. 2. NMR is a universal screening technique that, unlike bioassays, requires no knowledge of a protein’s function and therefore no target-specific set-up. Thus, protein targets that have been identified purely on a genomics basis and so far lack a functional bioassay can immediately be screened by NMR. 3. NMR concomitantly enables a determination of binding constants [15]. 4. NMR spectrally separates individual components, allowing the direct screening and deconvolution of mixtures from natural sources or combinatorial chemistry. 5. In addition to the mere binary binding information, NMR returns crucial structural information for both target and ligand with atomic resolution for the subsequent optimisation of weak initial hits into strongly binding drug candidates. Applications of NMR in drug discovery Tammo Diercks*, Murray Coles* and Horst Kessler†
  • 2. NMR screening exploits the fact that many NMR observ- ables change even upon temporary binding because of global and local effects (see Figures 1 and 2). The effects of binding may be monitored either on the target or on the ligand and are summarised in Table 1. The observed molecule primarily dictates inherent limitations such as sample availability (possibly with appropriate isotope labelling), solubility, stability and molecular size. Ligand-observed screening Detection with these techniques generally takes place on the dissociate, free form of the small ligand (implying a minimal Kd of ca. 10–7 M), allowing the direct identification of bind- ing species from mixtures. The unobserved target molecule is not subject to size limitations and may even be immo- bilised. In general, isotopic labelling is not required. Many of these techniques rely on size-dependent effects, which can be difficult to resolve for weak binding (implying a maximal Kd of ca. 10–3 M) and small relative molecular size differ- ences between ligand and target. These effects can be selected for in 1D filtered spectra, in which tuning the filter selects for different ligand affinities. Alternatively, the size of the effect could be evolved in a second indirect dimension, yielding pseudo-2D edited spectra. The information for structure-based ligand optimisation obtainable through these techniques is more limited than for target-observed tech- niques. It may, however, reveal a ligand’s binding epitope, its bound conformation, and even contacts to other ligands binding at adjacent sites. Use of molecular diffusion Detection of transiently binding ligands by their decreased diffusion coefficients is an established screening technique (for exhaustive reviews see [16,17•]) Pseudo-2D diffusion- edited experiments (diffusion-ordered spectroscopy, DOSY), where molecules are separated according to their individual diffusion coefficients, are particularly useful for deconvoluting mixtures. Alternatively, 1D diffusion filters can be combined with standard 2D correlation experi- ments, such as in the diffusion-modulated gradient COSY (correlation spectroscopy) [18] and diffusion-encoded total correlation spectroscopy (TOCSY; DECODES) [19], to elucidate spin systems and facilitate mixture analysis. Diffusion filtering or editing might also be enhanced by combination with other screening techniques, such as NOE pumping [20,21] (see below). In a promising appli- cation for combinatorial chemistry, diffusion filtering has recently been used in solid-state MAS (magic-angle spinning) NMR to identify and distinguish resin-bound molecules from impurities [22]. Use of relaxation Screening based on binding-induced slowing of global dynamics, leading to a general decrease in NMR relaxation times, is conceptually very similar to diffusion-based screening. The diffusion filter is replaced by an inversion- recovery delay, a spin-lock period or a CPMG echo to filter for the different types of relaxation times, T1, T1ρ and T2, respectively. One-dimensional relaxation-based screening usually requires subtraction of a reference spectrum of the target-free ligand mixture, and measures must be taken to avoid artefacts due to binding-induced lineshape distor- tions, chemical-shift changes and residual target signals [23]. T2-based ligand screening may also be performed by comparing spectral linewidths [24•] that increase upon binding. Complications can arise from superposition of line-broadening due to chemical exchange or slow 286 Combinatorial chemistry Figure 1 Global NMR consequences of ligand–target interactions. Observation is restricted to the ligand where the effects are most pronounced. Their strength depends on molecular size differences and binding kinetics. kA kD + Complex properties similar to target properties • Slow Relaxation • Fast diffusion • Positive NOE • Fast relaxation • Slow diffusion • Negative NOE Small ligands ComplexLarge target Current Opinion in Chemical Biology Figure 2 Local NMR effects of complex formation may be observed on the ligand and/or the target. Structural information is available as effects are mostly localised to the binding site. Surface protection Current Opinion in Chemical Biology Intermolecular magnetisation transfer Chemical shift perturbation Perturbation of local dynamics{
  • 3. conformational motions (on the microsecond and millisecond timescales), which are often relevant for induced-fit during intermolecular interactions and damped upon binding. Thus, a size-dependent decrease in T2 times might be compensated for by a flexibility-dependent reduction in line-broadening. In contrast, the recently proposed concept of transferred T1 times [25] relies on the fact that relaxation rates measured on a fast-exchanging ligand in the free state also contain contributions from its local dynamics in the bound state, as immobilisation of flexible regions of the ligand generally increases relaxation rates. Transferred relaxation rates may therefore be seen as a dynamic analogue of transferred NOE (see below). Individual differences in relaxation rates must be compared with their average in order to distinguish local from global dynamic changes [25]. Another complication arises from local relaxation contributions because of molecular anisotropy. Intramolecular magnetisation transfer: transfer-NOE Transferred intramolecular NOEs (trNOEs) are well established as unique sources of structural information on the bound ligand (for a review see [26]) where spin-diffu- sion should be suppressed to obtain optimal structural precision [27,28]). The observation of trNOE has proved to be equally useful in the fast screening of ligand mixtures [24•], forming the basis of ‘bioaffinity NMR’ [29]. The technique relies on the size-dependence of the intramolecular NOE, which shows slow NOE build-up with weak positive maxima for free ligands and rapid build-up with strong negative maxima for the bound state. If dissociation of the ligand occurs quickly enough (i.e. Kd > 10–7 M), a sufficient percentage of observable free ligand will retain intense negative trNOE as a ‘memory’ of the bound state and thus quickly indicate binding. If the residence time is too short (i.e. Kd > 10–3 M), however, trNOE will build up too weakly for detection [30]. Routinely, trNOE are recorded as 2D NOESY (NOE spectroscopy) spectra with short mixing times favouring build-up of trNOE over direct NOE. Relaxation filtering suppresses residual target signals. Notable are applica- tions of trNOE-based screening [30,31•,32] and recent experimental improvements [33]. Screening by trNOE imposes upper limits on the ligands (< 1 kDa), which have to fall into the positive NOE regime. For borderline cases, this regime may be extend- ed, for example, by increasing the temperature [31•]. Another problem is the possible cancellation of positive direct and negative transferred NOE, which could be resolved by more lengthy 3D experiments such as the 3D TOCSY–trNOESY [34], or by resorting to other tech- niques such as saturation transfer difference (see below). Intermolecular magnetisation transfer The intermolecular magnetisation transfer is always posi- tive and cancellation cannot occur. For its measurement, non-equilibrium magnetisation must first be created asym- metrically (i.e. on one component only, usually the drug target). It is then transferred to the binding partner during a mixing period. The intermolecular magnetisation trans- fer results in altered signal intensities for both components and can finally be monitored on either the starting or the receiving molecule (usually the ligand). Remaining target resonances can be suppressed (e.g. by isotope filtering or relaxation filtering). Subtraction of a reference spectrum (recorded without initially disturbing the magnetisation) is usually required to reveal these intensity changes and entails the risk of subtraction artefacts. Asymmetric non-equilibrium magnetisation on the target molecule may generally be created by selective irradiation, requiring the existence of a unique NMR frequency band. Applications of NMR in drug discovery Diercks, Coles and Kessler 287 Table 1 NMR observable effects of ligand binding. Global effects of binding Ligand observed Screening Applications References Translational dynamics Diffusion rates Affinity NMR [17•,19] Rotational dynamics Relaxation rates Line broadening [23,24•] Sign of transferred intramol. NOE Bioaffinity NMR (trNOE) [29,30,31•,32,34] Molecular orientation Residual dipolar couplings * Local effects of binding Ligand- or target-observed Chemical shift perturbation Chemical shifts SAR-by-NMR (HSQC screening) [44•,48,49,51,53•] Local dynamics perturbation Relaxation rates * Surface protection Water exchange rates * Paramagnetic surface probing * Intermolecular magnetisation transfer Via scalar couplings * (e.g. intermol. H-bonds) Via nuclear dipolar couplings Bioaffinity NMR (STD) [32,35,37•] (intermol. NOE) (Reverse) NOE pumping [20,21] WaterLOGSY [38] Via electron dipolar couplings (paramagnetic relaxation) SLAPSTIC [39•,41•] *No application to NMR screening reported yet. Intermol., intermolecular.
  • 4. Initially localised, it is spread across the entire molecule by spin diffusion such that magnetisation may be picked up by a ligand binding anywhere on the surface. Alternatively, non-equilibrium magnetisation may be created symmetri- cally, followed by asymmetric suppression making use of the large size difference between target and ligands. For instance, a diffusion filter would suppress magnetisation on the ligands, whereas a relaxation filter would suppress magnetisation on the target. Saturation transfer difference (STD) [35] as the special case of intermolecular steady-state NOE is observed after frequency-selective irradiation of the target and subtrac- tion of a reference spectrum with off-resonance irradiation. Although usually run as 1D spectra, STD can be coupled to standard 2D correlation experiments such as TOCSY, COSY and HMQC (heteronuclear multiple quantum correlation) to alleviate spectral overlap [32,35]. The distance-dependence of the observed signal intensities identifies those nuclei closest to the target–ligand interface. This ‘epitope mapping’ [28,36] may be seen as a ligand-based complement to the ‘SAR-by-NMR’ approach. A remarkable application extends STD screening to solid-state NMR [37•], in which the target was immobilized on controlled-pearl glass, facilitating its recovery. From a comparison of STD integrals, it was possible to assess relative binding strengths of the dissolved ligands. A reference spectrum with pure target-free resin must rule out direct ligand–resin interactions. NOE pumping [20] employs the scheme of symmetric magnetisation inversion, followed by asymmetric suppres- sion. Classically, the ligand magnetisation is suppressed via diffusion filtering, and its recovery directly indicates inter- molecular magnetisation transfer from the target, hence binding. A suppression of remaining target magnetisation prior to acquisition greatly facilitates identification, but the diffusion filter employed may entail serious intensity losses through concomitant relaxation of the target magnetisation. This problem is alleviated in the reverse case (reverse NOE pumping) [21], in which ligand magnetisation is preserved while target magnetisation is suppressed via relaxation or isotope filtering. The ligand magnetisation is then reduced because of transfer onto the target, and signal reductions observed on a ligand thus indicate binding. These reductions, however, are only revealed after subtrac- tion of a reference spectrum, which also takes account of unspecific magnetisation losses through T1 relaxation. WaterLOGSY [38] exploits the surface layer of target- bound water molecules as a pool of selectively invertible magnetisation that can be transferred onto binding ligands. After simultaneous selective water suppression and relaxation filtering (for target suppression), only signals of binding ligands are detected. Paramagnetic spin labels Magnetic spin interactions can be enhanced drastically by introducing paramagnetic spin labels. The induced NMR effects — resonance shifts and line broadening — can thus be observed over larger distances (up to some 20 Å), shorter contact times, and at lower protein concentrations. Soluble paramagnetic reagents have classically been employed to search for metal ion binding sites and probe solvent-accessible surfaces. Intermolecular binding, which blocks part of the solvent-accessible surface, may be detected and localised in a similar manner. In a recent application to second-site screening, a confirmed binding ligand was spin labelled and used to search for ligands concomitantly binding to the target protein at an adjacent site [39•]. The method is analogous to the observation of transferred inter-ligand NOE contacts [40], yet offers larger specificity and sensitivity, establishing connectivity between more distant binding sites. The need to synthesize spin-labelled ligands without affecting their natural binding properties is, however, a major drawback. A blind test must furthermore rule out that ligands interact in the absence of the target. In the SLAPSTIC method, the target protein is spin- labelled to assist in identifying binding ligands [41•]. The line-broadening of ligand signals upon binding is thus strongly emphasized and allows for a reduction in protein (and ligand) amounts by one or two orders of magnitude. It is critical to spin-label the target near, but not within the binding site, as this would invariably lead to altered binding properties. Moreover, no structural information can be derived from this method. Use of residual dipolar couplings Alignment of the target molecule also causes alignment of the ligand upon temporary binding. RDC may thus be transferred from the bound to the isotropic free state if dissociation is fast enough (Kd in the millimolar range) [42,43]. In contrast to NOE data, the structural information available from RDC does not suffer from spin-diffusion and is therefore precise even for large, immobilised targets. RDC yield information on both the bound ligand’s structure and its orientation relative to the target without the need for assigning target resonances as demonstrated in two recent applications [42,43]. This technique has not, however, been applied to ligand screening yet. Target-observed screening Observation on the target, rather than on dissociated ligands, is neither restricted by the ligands’ size nor by an upper affinity limit. It also immediately reveals different binding sites, enables direct distinction between specific and unspecific binding and provides a wealth of structural information for ligand optimisation. This information, however, can only be extracted if the spectral assignments for the target are known. Inherent limitations often preclude observation on the large target molecule, although relaxation-optimised techniques (TROSY) may be of benefit. The amounts of protein required, which must be isotope-labelled, can be reduced significantly by employing 288 Combinatorial chemistry
  • 5. cryoprobe technology and screening larger mixtures of compounds [44•]. Mixtures of ligands can not, however, be directly deconvoluted. Chemical-shift perturbations Most target-observed screening applications rely on chemical-shift changes as indicators for intermolecular binding. After assignment, these changes can be mapped upon the protein structure, revealing its binding sites [45,46] and guiding structure-based ligand optimisation. This forms the basis of the ‘SAR-by-NMR’ approach [47,48], in which optimised ligands are constructed by linking weakly binding fragments. The approach contin- ues to be successfully used in drug development [49–52]. Standard screening using 2D 15N,1H correlation spectra has recently been complemented by 13C,1H HSQC-based screening on proteins bearing 13C-labeled methyl groups [53•]. The selective labelling, using inexpensive starting materials, avoids the detrimental effects of 13C–13C homonuclear coupling while greatly reducing the cost of sample preparation. This application benefits from the higher proton multiplicity and slower transverse relaxation of the methyl groups. Consequently, intensities were reported to be on average three-times higher. On the other hand, screening based on methyl groups suffers from assignment difficulties and lower spectral dispersion. Note that the distribution and accessibility of (polar) amide and (non-polar) methyl groups are not equivalent, possibly leading to different screening sensitivities. Equating chemical-shift perturbations with spatial proximity to the ligand may be misleading, especially in the presence of large shielding anisotropies in a ligand or because of con- formational changes in the target. Therefore, an alternative has been proposed in which the binding site and the bound ligand’s orientation can be determined more reliably [54] without the need for a complete re-determination of the complex structures. It is based on the comparison of chemi- cal-shift changes in the proximity of the binding site induced by a series of closely related ligands. This can be seen as trading synthetic for spectroscopic efforts. Intermolecular magnetisation transfer The general scheme for intermolecular magnetisation transfer experiments (see above) can also be employed for target-detected screening. In one concept [55], target mag- netisation is selectively suppressed by relaxation filtering after symmetric excitation and then recovers by intermolec- ular transfer from binding ligands. Prior to observation on the target, the ligand signals are filtered out by selective multiple frequency suppression. This, however, precludes the direct screening of ligands with more complex 1H spectra. Screening for macromolecular interactions Screening for macromolecular interactions is a special case in which the ‘ligand’ and ‘target’ have comparable molecu- lar sizes, and size discrimination is impracticable. Other than that, many of the techniques mentioned above can be and have been applied to the problem. For instance, STD was used to probe protein–nucleic-acid interactions by saturating well-separated protons of the latter [56]. A varia- tion of the scheme scans for protein–protein interactions [57] with molecular distinction made by perdeuterating and 15N-labelling only one partner. The unlabelled protein remains susceptible to saturation of the aliphatic protons, while intermolecular STD is observed on the 1H,15N moieties of the labelled protein. The low proton density in this perdeuterated species precludes efficient spin-diffu- sion and the STD is largely confined to the binding site. Design of NMR screening libraries In contrast to other screening techniques, NMR screening requires relatively high sample concentrations and sampling times. With its unprecedented sensitivity (detecting interac- tions with Kd up to 10–2 M) and atomic resolution, however, NMR is particularly suited for guiding the ‘bottom-up’ construction of complex ligands from small, weakly binding molecular scaffolds. In line with this, the principles for com- posing a dedicated ligand library are availability, good solubility, low molecular mass (i.e. < 1–2 kDa), and limita- tion to some 102–103 compounds of maximal diversity. For mixture screening, batches should contain (non-reacting) ligands with isolated NMR signals for easy monitoring. Various attempts have been made to construct libraries using consensus fragments identified from statistical analyses of known drugs (e.g. see [24•,58,59]) and protein-binding ligands [60], and from fragmenting an existing lead compound [52]. Pre-selection of small ligands for NMR screening can also be based on the scoring results obtained from virtual docking [61]. Conclusions NMR screening is now being recognised by industry as a powerful technique for lead discovery. The underlying prin- ciples have mostly been discovered and developed long ago by academic scientists, such that the novelty of NMR screening lies mostly with its applications. In this review, we have offered a classification of the available NMR methods and outlined possible future developments by a reduction to basic NMR properties and principles. The distinction between ligand- and target-observed screening techniques is most important. In practice, a dual strategy that combines the complementary strengths and detectable affinity ranges of both techniques should prove advantageous. Most impor- tantly, ligand-observed techniques may produce false negatives in cases of very strong binding where exchange between target-bound and detected free ligand is too slow. These techniques nevertheless have clear advantages for pre-screening large libraries as they implicitly deconvolute ligand mixtures, require no isotope labelling and pose few limitations on the target. Consecutive target-observed ‘fine- ’screening of the identified binding ligands can then rule out unspecific binding, identify different binding sites and modes, and guide structure-based ligand optimisation. By Applications of NMR in drug discovery Diercks, Coles and Kessler 289
  • 6. screening a carefully composed small library of basic molecular scaffolds, the pre-screening step may be omitted for amenable targets. References and recommended reading Papers of particular interest, published within the annual period of review, have been highlighted as: • of special interest ••of outstanding interest 1. Moore JM: NMR screening in drug discovery. Curr Opin Biotechnol 1999, 10:54-58. 2. Moore JM: NMR techniques for characterization of ligand binding: utility for lead generation and optimization in drug discovery. Biopolymers 1999, 51:221-243. 3. Roberts GCK: NMR spectroscopy in structure-based drug design. Curr Opin Biotechnol 1999, 10:42-47. 4. Roberts GCK: Applications of NMR in drug discovery. Drug Discov Today 2000, 5:230-240. 5. Stockman BJ: NMR spectroscopy as a tool for structure-based drug design. Progr NMR Spectrosc 1998, 33:109-151. 6. Keifer PA: NMR tools for biotechnology. Curr Opin Biotechnol 1999, 10:34-41. 7. Ross A, Schlotterbeck G, Klaus W, Senn H: Automation of NMR •• measurements and data evaluation for systematically screening interactions of small molecules with target proteins. J Biomol NMR 2000, 16:139-146. This reference gives valuable practical advice on performing industrial-scale ligand screening by NMR, presenting a fully integrated hardware set-up with robotized sample preparation, flow-through probehead and automated data analysis of the acquired 15N,1H-HSQC. A pre-check of the protein–ligand mixtures for aggregation and solubility is required to prevent clogging of the flow-through probe. The authors conclude that screening individual ligands, rather than mixtures, justifies the extra time by alleviating the need for char- acterizing possible ligand–ligand interactions and deconvoluting mixtures. The problem of using too low buffer concentrations, entailing artefactual chemical shift changes, is also discussed. 8. Haner RL, Llanos W, Mueller L: Small volume flow probe for automated direct-injection NMR analysis: design and performance. J Mag Res 2000, 143:69-78. 9. Pervushin K, Riek R, Wider G, Wüthrich K: Attenuated T2 relaxation by mutual cancellation of dipole-dipole coupling and chemical shift anisotropy indicates an avenue to NMR structures of very large biological macromolecules in solution. Proc Natl Acad Sci USA 1997, 94:12366-12371. 10. Permi P, Annila A: Transverse relaxation optimised spin-state selective NMR experiments for measurement of residual dipolar couplings. J Biomol NMR 2000, 16:221-227. 11. Prestegard JH: New techniques in structural NMR — anisotropic interactions. Nat Struct Biol 1998, (NMR suppl):517-522. 12. Rückert M, Otting G: Alignment of biological macromolecules in novel nonionic liquid crystalline media for NMR experiments. J Am Chem Soc 2000, 122:7793-7797. 13. Cordier F, Grzesiek S: Direct observation of hydrogen bonds in proteins by interresidue h3JNC’ scalar couplings. J Am Chem Soc 1999, 121:1601-1602. 14. Goto NK, Kay LE: New developments in isotope labeling strategies for protein solution NMR spectroscopy. Curr Opin Struct Biol 2000, 10:585-592. 15. Fielding L: Determination of association constants (Ka) from solution NMR data. Tetrahedron 2000, 56:6151-6170. 16. Johnson CS: Diffusion ordered nuclear magnetic resonance spectroscopy: principles and applications. Progr NMR Spectrosc 1999, 34:203-256. 17. Chen A, Shapiro MJ: Affinity NMR. Anal Chem 1999, • 71:669A-675A. A review describing NMR screening, in particular diffusion-based affinity NMR, in a modular way. 18. Gmeiner WH, Hudalla CJ, Soto AM, Marky L: Binding of ethidium to DNA measured using a 2D diffusion-modulated gradient COSY NMR experiment. FEBS Lett 2000, 465:148-152. 19. Bleicher K, Lin M, Shapiro MJ, Wareing JR: Diffusion edited NMR: screening compound mixtures by affinity NMR to detect binding ligands to vancomycin. J Org Chem 1998, 63:8486-8490. 20. Chen A, Shapiro MJ: NOE pumping: a novel NMR technique for identification of compounds with binding affinity to macromolecules. J Am Chem Soc 1998, 120:10258-10259. 21. Chen A, Shapiro MJ: NOE pumping. 2. A high-throughput method to determine compounds with binding affinity to macromolecules by NMR. J Am Chem Soc 2000, 122:414-415. 22. Shapiro MJ, Chin J, Chen A, Wareing JR, Tang Q, Tommasi RA, Marepalli HR: Covalent or trapped? PFG diffusion MAS NMR for combinatorial chemistry. Tetrahedron Lett 1999, 40:6141-6143. 23. Hajduk PJ, Olejniczak ET, Fesik SW: One-dimensional relaxation- and diffusion-edited NMR methods for screening compounds that bind to macromolecules. J Am Chem Soc 1997, 119:12257-12261. 24. Fejzo J, Lepre CA, Peng JW, Bemis GW, Ajay, Murcko MA, Moore JM: • The SHAPES strategy: An NMR-based approach for lead generation in drug discovery. Chem Biol 1999, 6:755-769. Although no new techniques are described, a clear overview on the set-up of NMR screening, including library design is provided. 25. LaPlante SR, Aubry N, Deziel R, Ni F, Xu P: Transferred 13C T1 relaxation at natural isotopic abundance: a practical method for determining site-specific changes in ligand flexibility upon binding to a macromolecule. J Am Chem Soc 2000, 122:12530-12535. 26. Ni F: Recent developments in transferred NOE methods. Progr NMR Spectrosc 1994, 26:517-606. 27. Vincent SJF, Zwahlen C, Post CB, Burgner JW, Bodenhausen G: The conformation of NAD++ bound to lactate dehydrogenase determined by nuclear magnetic resonance with suppression of spin-diffusion. Proc Natl Acad Sci USA 1997, 94:4383-4388. 28. Maaheimo H, Kosma P, Brade L, Brade H, Peters T: Mapping the binding of synthetic disaccharides representing epitopes of chlamydial lipopolysaccharide to antibodies with NMR. Biochemistry 2000, 39:12778-12788. 29. Meyer B, Weimar T, Peters T: Screening mixtures for biological activity by NMR. Eur J Biochem 1997, 246:705-709. 30. Mayer M, Meyer M: Mapping the active site of angiotensin- converting enzyme by transferred NOE spectroscopy. J Med Chem 2000, 43:2093-2099. 31. Henrichsen D, Ernst B, Magnani JL, Wang W-T, Meyer B, Peters T: • Bioaffinity NMR spectroscopy: identification of an E-selectin antagonist in a substance mixture by transfer NOE. Angew Chem Int Ed Engl 1999, 38:98-102. This article describes an application of trNOE-based screening to a mixture of ligands, also discussing important details to be considered with this tech- nique. Prior to screening, the library was tested for the positive NOE regime, which was eventually reached only after increasing the temperature. The binding ligand then identified by negative trNOE displayed strong spin- diffusion, which in this case helped its identification, but generally blurs structural information. Suppression of residual target protein signals by means of a relaxation filter was largely complete, except for signals from a highly flexible, attached carbohydrate chain. 32. Vogtherr M, Peters T: Application of NMR based binding assays to identify key hydroxy groups for intermolecular recognition. J Am Chem Soc 2000, 122:6093-6099. 33. Weimar T: Improving bioaffinity NMR spectra by means of zero- quantum dephasing. Magn Reson Chem 2000, 38:315-318. 34. Herfurth L, Weimar T, Peters T: Application of 3D-TOCSY-tr-NOESY for the assignment of bioactive ligands from mixtures. Angew Chem Int Ed Engl 2000, 39:2097-2099. 35. Mayer M, Meyer B: Characterization of ligand binding by saturation transfer difference NMR spectroscopy. Angew Chem Int Ed Engl 1999, 38:1784-1788. 36. Mayer M, Meyer B: Group epitope mapping (GEM) to identify segments of a ligand in direct contact with a protein: STD NMR as a tool to characterize binding to Ricinus communis agglutinin. J Am Chem Soc 2001, in press. 290 Combinatorial chemistry
  • 7. 37. Klein J, Meinecke R, Mayer M, Meyer B: Detecting binding affinity to • immobilized receptor proteins in compound libraries by HR-MAS STD NMR. J Am Chem Soc 1999, 121:5336-5337. This article describes an exciting extension of the STD screening method to high-resolution MAS NMR, using dissolved ligands and a target immobilized on controlled pearl glass for easy protein recovery. A reference spectrum with pure, target-free resin must be recorded to rule out ligand–resin inter- actions. Relative binding strengths of competitive ligands can be assessed from a comparison of STD integrals. Structural information, however, is restricted to the binding epitope of the ligand. 38. Dalvit C, Pevarello P, Tato M, Veronesi M, Vulpetti A, Sundström M: Identification of compounds with binding affinity to proteins via magnetization transfer from bulk water. J Biomol NMR 2000, 18:65-68. 39. Jahnke W, Perez LB, Paris CG, Strauss A, Fendrich G, Nalin CM: • Second-site NMR screening with a spin-labelled first ligand. J Am Chem Soc 2000, 122:7394-7395. This interesting alternative to the so-called ‘SAR-by-NMR’ approach uses a spin-labelled known binding ligand to induce detectable signal broadening or extinction on another ligand binding to an adjacent site on the target pro- tein. This novel method for the exclusive observation of second-site binding is highly sensitive and also provides some structural information. 40. Li D, DeRose EF, London RE: The inter-ligand Overhause effect: a powerful new NMR approach for mapping structural relationships of macromolecular ligands. J Biomol NMR 1999, 15:71-76. 41. Jahnke W, Rüdisser S, Zurini M: Spin label enhanced NMR • screening. J Am Chem Soc 2001, 123:3149-315. This article describes a novel technique, SLAPSTIC, which uses a modified, spin-labelled target protein for ligand screening. Ligand binding is detected by signal broadening and extinction on the free form of a temporarily binding ligand. The strong magnetic marker allows a drastic reduction in the required protein and ligand concentrations, which also translates into lower minimum ligand solubilities and access to lower binding affinities. 42. Koenig BW, Mitchell DC, König S, Grzesiek S, Litman BJ, Bax A: Measurement of dipolar couplings in a transducin peptide fragment weakly bound to oriented photo-activated rhodopsin. J Biomol NMR 2000, 16:121-125. 43. Thompson GS, Shimizu H, Homans SW, Donohue-Rolfe A: Localization of the binding site for the oligosaccharide moiety of GB3 on verotoxin 1 using NMR residual dipolar coupling measurements. Biochemistry 2000, 39:13153-13156. 44. Hajduk PJ, Gerfin T, Boehlen J-M, Häberli M, Marek D, Fesik SW: • High-throughput nuclear magnetic resonance-based screening. J Med Chem 1999, 42:2315-2317. This article presents a profound analysis of the benefits of cryoprobe tech- nology to 15N-HSQC-based NMR screening. The merits alternatively trans- late into lower detectable ligand affinities (with up to millimolar dissociation rates), substantially reduced protein and ligand concentrations, or faster screening rates. The authors claim that libraries with up to 200,000 ligands (administered in mixtures of 100 compounds) could thus be screened effi- ciently within less than a month. 45. Sakamoto T, Tanaka T, Ito Y, Rajesh S, Iwamoto-Sugai M, Kodera Y, Tsuchida M, Shibata T, Kohno T: An NMR analysis of ubiquitin recognition by yeast ubiquitin hydrolase: evidence for novel substrate recognition by a cysteine protease. Biochemistry 1999, 38:11634-11642. 46. Sun C, Cai M, Gunasekera AH, Meadows RP, Wang H, Chen J, Zhang H, Wu W, Xu N, Ng SC et al.: NMR structure and mutagenesis of the inhibitor-of-apoptosis protein XIAP. Nature 1999, 401:818-822. 47. Hajduk PJ, Sheppard G, Nettesheim D, Olejniczak ET, Shuker SB, Meadows RP, Steinman DH, Carrera GM, Marcotte PA, Severin J et al.: Discovery of potent nonpeptide inhibitors of stromelysin using SAR by NMR. J Am Chem Soc 1997, 119:5818-5827. 48. Shuker SB, Hajduk PJ, Meadows RP, Fesik SW: Discovering high- affinity ligands for proteins: SAR by NMR. Science 1996, 274:1531-1534. 49. Hajduk PJ, Zhou M-M, Fesik SW: NMR-based discovery of phosphotyrosine mimetics that bind to the Lck SH2 domain. Bioorg Med Chem Lett 1999, 9:2403-2406. 50. Hajduk PJ, Dinges J, Schkeryantz JM, Janowick D, Kaminski M, Tufano M, Augeri DJ, Petros A, Nienaber V, Zhong P et al.: Novel inhibitors of Erm methyltransferases from NMR and parallel synthesis. J Med Chem 1999, 42:3852-3859. 51. Hajduk PJ, Boyd S, Nettesheim D, Nienaber V, Severin J, Smith R, Davidson D, Rockway T, Fesik SW: Identification of novel inhibitors of urokinase via NMR-based screening. J Am Chem Soc 2000, 43:3862-3866. 52. Hajduk PJ, Gomtsyan A, Didomenico S, Cowart M, Bayburt EK, Solomon L, Severin J, Smith R, Walter K, Holzman TF et al.: Design of adenosine kinase inhibitors from the NMR-based screening of fragments. J Med Chem 2000, 43:4781-4786. 53. Hajduk PJ, Augeri DJ, Mack J, Mendoza R, Yang J, Betz SF, Fesik SW: • NMR-based screening of proteins containing 13C-labeled methyl groups. J Am Chem Soc 2000, 122:7898-7904. This article evaluates the advantages of 13C-HSQC-based screening on a target protein selectively 13C-labelled at the methyl groups, including a com- parison with 15N-HSQC- and 15N-TROSY-based screening. 54. Medek A, Hajduk PJ, Mack J, Fesik SW: The use of differential chemical shifts for determining the binding site location and orientation of protein-bound ligands. J Am Chem Soc 2000, 122:1241-1242. 55. Dalvit C, Floersheim P, Zurini M, Widmer A: Use of organic solvents and small molecules for locating binding sites on proteins in solution. J Biomol NMR 1999, 14:23-32. 56. Ramos A, Kelly G, Hollingworth D, Pastore A, Frenkiel T: Mapping the interface of protein-nucleic acid complexes using cross- saturation. J Am Chem Soc 2000, 122:11311-11314. 57. Takahashi H, Nakanishi T, Kami K, Arata Y, Shimada I: A novel NMR method for determining the interfaces of large protein–protein complexes. Nat Struct Biol 2000, 7:220-223. 58. Bemis GW, Murcko MA: The properties of known drugs. 1. Molecular frameworks. J Med Chem 1996, 39:2887-2893. 59. Bemis GW, Murcko MA: Properties of known drugs. 2. Side chains. J Med Chem 1999, 42:5095-5099. 60. Hajduk PJ, Bures M, Praestgaard J, Fesik SW: Privileged molecules for protein binding identified from NMR-based screening. J Med Chem 2000, 43:3443-3447. 61. Muegge I, Martin YC, Hajduk PJ, Fesik SW: Evaluation of PMF scoring in docking weak ligands to the FK506 binding protein. J Med Chem 1999, 42:2498-2503. Applications of NMR in drug discovery Diercks, Coles and Kessler 291