You are on page 1of 6

Sensor Issues

pubs.acs.org/acssensors

Harnessing the Versatility of Optical Biosensors for Target-Based


Small-Molecule Drug Discovery
Tim Kaminski, Anders Gunnarsson, and Stefan Geschwindner*
Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Pepparedsleden 1, S-43183 Mölndal,
Sweden

ABSTRACT: Optical biosensors entered target-based small-molecule drug discovery


more than two decades ago and have since transformed into a value-adding
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

component in the decision-making process. Here, we briefly highlight the major


application areas of optical biosensors and focus on desirable profiles of such platforms
in order to ensure their effective use in small molecule drug discovery. Furthermore,
we will emphasize current technology-based constraints and discuss experimental
strategies to address these limitations as well as provide a view of necessary technology
Downloaded via 141.0.9.202 on February 2, 2020 at 12:24:30 (UTC).

improvements for next generation platforms.

KEYWORDS: drug discovery, small molecule, kinetics, single molecule, surface-plasmon resonance, waveguide-grating,
biolayer interferometry

■ CURRENT USE AND IMPACT OF OPTICAL


BIOSENSORS IN TARGET-BASED
screening of much smaller compound libraries (<2000) and
typically monitors compound binding directly by employing,
SMALL-MOLECULE DRUG DISCOVERY e.g., fluidics-based SPR7 or BLI.5 This has recently gained in
Optical Biosensors have seen widespread application in target- importance, as FBDD has become a valuable alternative to
based drug discovery following the appearance of the first traditional hit finding approaches like high-throughput screen-
commercial platforms in the early 1990s.1,2 Instruments based ing (HTS) and typically involves screening campaigns using
on surface plasmon resonance (SPR),3 optical waveguide smaller compound fragments (∼200 Da) that are binding
grating (OWG),4 or biolayer interferometry (BLI)5 are weakly (Kd ∼ mM) to the target of interest.8 Thanks to their
predominantly applied in early drug discovery focusing on improved sensitivity, optical biosensors are effectively used
small-molecule screening and profiling, antibody character- alongside more traditional biophysical methods like nuclear
ization and selection in addition to biomarker detection, of magnetic resonance (NMR) or X-ray crystallography to fuel
which the latter two will not be discussed here. The major FBDD campaigns with novel chemistry.


application areas in the small molecule space aim to identify
and characterize the interaction of compounds with their target
protein, which encompass (i) primary screening, (ii) validation VALIDATION OF TARGET ENGAGEMENT
of target engagement, (iii) identification of the molecular mode Validation of primary hits or the assessment of target
of action (MoA), along with (iv) detailed characterization of engagement is predominantly used to confirm HTS actives.
compound binding such as binding kinetics and thermody- This often starts alongside HTS assay development and thus
namics. A summary of established and currently emerging prior to any screening campaign, in order to assess the quality
biosensor platforms that are used in the context of small of the assay (typically biochemical) and to understand
molecule drug discovery is provided in Table 1, including their
limitations as well as potential sources for HTS assay artifacts.9
primary application areas as well as the associated information
Even more important is the assessment of constructive target
content.


engagement of compounds identified by HTS, as those typically
PRIMARY SCREENING serve as starting points for extended and resource-intensive
medicinal chemistry campaigns.10 In this context, it is
Primary screening using optical biosensors can utilize plate-
based OWG systems to enable screening of relatively large important to note that target engagement is required but
compound libraries (>10 000). The experimental flexibility often not sufficient to elicit a pharmacological response.
allows equally for a biochemical4 or a cell-based readout,6 with
the notion that the later rather reflects the consequences of Received: November 15, 2016
compound binding through changes in cell morphology. In Accepted: December 15, 2016
contrast, Fragment-based drug discovery (FBDD) requires Published: December 15, 2016

© 2016 American Chemical Society 10 DOI: 10.1021/acssensors.6b00735


ACS Sens. 2017, 2, 10−15
ACS Sensors Sensor Issues

ref
■ UNDERSTANDING THE MOLECULAR MoA

11

12
3

4
To confirm compound-target engagement is only one of many
critical factors to drive compound development and ultimately

higher reagent consumption, de-


lower sensitivity, higher reagent

lower sensitivity, higher reagent

pendence on conformational
ensure clinical success. Equally important is to understand and
Table 1. Selection of Already Established and Currently Emerging Optical Biosensing Technologies Applied during Target-Based Small-Molecule Drug Discovery

confirm the compound-mechanism hypothesis, which means


limitations

building a detailed understanding of the molecular MoA.13 This


information can often help to bridge from simplistic in vitro
lower throughput

lower throughput
consumption

consumption observations to pharmacodynamics activity in a biologically


relevant disease setting. In the case of the α7 nicotinic

change
acetylcholine receptor, it is known that positive allosteric
modulators can provide a pharmacological advantage compared
affinity, kinetics, stoichi- to direct agonists.14 By tweaking the conditions in a biosensing
affinity, conformational
modynamics, stoichi-
information content

experiment, one is able to clearly distinguish compounds that


affinity, kinetics, ther-

directly act via the agonist binding site and those that modulate
affinity, kinetics

receptor activation via allosteric sites and thus might offer a


higher therapeutic potential.15 Such mechanistic studies
changes
ometry

ometry

frequently complement target engagement studies, as they


affinity

typically build on similar assay formats and platforms.


screening including whole cells, mode of
action studies including pathway anal-

screening (for conformational changes),

BINDING KINETICS AND THERMODYNAMICS


screening, target engagement, mode of

screening, target engagement, mode of

screening, target engagement, mode of

Finally, optical biosensors can provide a detailed character-


main application areas

ization of the dynamics of binding. A good example relates to


the generation of binding kinetic data, which can be used to
mode of action studies

drive structure−kinetic relationships during the compound


optimization process.16 Kinetic information is frequently
action studies

action studies

action studies

deduced through microfluidics-based biosensor platforms like


SPR, as the optimized fluidics and the high sampling rates allow
for an exact description of association and dissociation
ysis

processes. Information about extended dissociation processes


can eventually highlight possibilities to enhance compound
like SPR, detects refractive index changes within an evanescent field near the sensor surface, due

efficacy and safety and thus may help to ensure therapeutic


incident light couples into the waveguide resulting in a narrow band of reflected wavelengths,

induced conformational changes lead to a different relative orientation of the dye, which is
second-harmonic generation sensitive dyes are conjugated to surface-tethered targets; ligand-
analyzes the change in interference pattern of white light reflected from reference and active
detects changes in refractive index close to the sensor surface induced by analyte binding to

success if the pharmacokinetic features are compatible.17 In


analogy, thermodynamic data is generated by studying the
temperature-dependency of the binding affinity and can
eventually highlight compounds with a favorable enthalpy
allowing to detect changes in the sensing layer containing ligands or cells

profile, albeit the value of such data for more successful


compound design is disputable.18

■ DESIRED SPECIFICATIONS FOR OPTICAL


BIOSENSORS PLATFORMS
detection principles

Previously described application areas have some common


denominators for the optimal application of biosensor plat-
to mass changes induced by complex formation

forms in drug discovery. It can be essentially reduced to three


surface, containing surface-tethered ligand

main factors that make a successful assay: (i) the right


throughput, (ii) the right sensitivity, and (iii) the right reagent
(Figure 1). In order to effectively conduct primary screening
activities, higher throughput becomes a key factor, as the results
from such activities need to feed timely into the decision-
surface-tethered ligands

making process. This becomes even more important when


engaging in FBDD, as there is the frequent desire to conduct
this in parallel to HTS in order to get optimal synergies
between these activities.8 While biochemical assays used for
detected

HTS typically offer the desired throughput, they are frequently


not able to detect weak interactions, which displays a severe
shortcoming particular to FBDD. Consequently, biosensor
platforms need to provide both high detection sensitivity, i.e.,
second-harmonic gener-

ting-coupled interfer-
biolayer interferometry

optical waveguide gra-


surface plasmon reso-

waveguide-based gra-

acceptable signal-to-noise ratios together with an adequate


throughput of >1000 data points per day.
name

Once moving forward in this process, an ideal scenario is the


use of the same or a similar biosensor platform and/or assay
ometry
nance

ation
ting

configuration to continuously support the maturation of weak


binding fragments into more potent chemical leads. Simulta-
11 DOI: 10.1021/acssensors.6b00735
ACS Sens. 2017, 2, 10−15
ACS Sensors Sensor Issues

addressed. Nevertheless, such modifications are typically


tedious and tend to be very resource-intensive without
guarantee for success.
In summary, the throughput and the sensitivity of any
biosensor platform typically provides a framework for defining
optimal protein reagents or tools in order to meet the
individual project requirements. These can vary quite
substantially and thereby expose both strengths as well as
weaknesses of the individual biosensor platforms used in drug
discovery.

■ STRENGTHS AND WEAKNESSES OF MAIN


BIOSENSOR PLATFORMS IN DRUG DISCOVERY
The choice of optical biosensor platform is heavily dependent
on application and assay requirements. As plate-based
Figure 1. The three R’s of biosensing representing key factors for biosensor platforms like OWG offer a similar throughput as
successful generation of biosensor-derived data to support target-based biochemical assays used in HTS, they provide an attractive
small-molecule drug discovery: right throughput, right sensitivity, and alternative for biophysical screening. Nevertheless, those
right reagent. platforms typically experience relatively low sensitivity when
monitoring direct and specific small molecule binding, which is
neously, one often needs to enable hit validation originating in part a consequence of the limited tethering capacity of the
from alternative hit finding approaches such as HTS. functionalized biosensor surface. Thus, such platforms are more
Consequently, such platforms need to be able to cover a useful when trying to identify so-called “promiscuous” or
broad affinity range from millimolar- down to picomolar- aggregation-based inhibitors, as those signals tend to be much
binding affinities. This will ensure such platforms are applicable larger and are easily detectable.20
throughout the early drug discovery process from hit finding Changing the assay configuration can in some cases bypass
and validation via lead identification into lead optimization and the sensitivity limitations of OWG platforms and enables the
beyond. screening of weakly binding fragments to support FBDD. This
A key but often neglected aspect is the stringent require- is achieved through directed tethering of a functionalized tool
ments on biosensor-compatible protein reagents since they compound, which still retains good binding competence to the
make intimate use of the transducer surface. Hence, one of the respective target protein. Challenging the modified biosensor
most critical aspects of data quality is the ability to tether a with the target protein typically leads to a detectable signal, as
functional target with high stability to the surface. In addition, the amplitude scales with the change in molecular mass on the
the density of tethered functional target must be relatively high, biosensor. This signal can be subsequently modulated by
since the signal amplitude scales with the surface density of the compounds that compete for the same binding site in solution,
tethered ligand. This is particular critical in the case of small thus this assay configuration is also referred to as inhibition in
molecule interactions due to the relatively low signal generated solution assay (ISA).21
upon binding. In summary, optimization of protein reagent is A different picture emerges for SPR-based instruments.
key for successful deployment of any biosensor platform in While OWG platforms provide much higher throughput than
early drug discovery. sequentially operating systems (interrogating one compound or
There are a number of intrinsic and extrinsic factors that will concentration at a time), SPR systems typically offer much
influence this optimization workflow. Target stability can be higher sensitivity at the cost of lower throughput. This generally
affected by the buffer and its supplements, making screening for makes it the platform of choice for driving FBDD campaigns in
the right assay conditions often a prerequisite. Frequently, the a direct binding assay (DBA) configuration, as this allows
manipulation of the target protein itself is critical for success. reliable detection of weak interactions with small molecules to
This may include addition of short peptide tags (e.g., Avi- or surface-tethered proteins.7 As a consequence of the fluidics-
hexa-His) to allow for directed and thus more controlled target based nature of these platforms, there is the additional
tethering in contrast to nondirectional covalent- (e.g., amine or possibility to record binding kinetics data. While plate-based
maleimide chemistry) or noncovalent (charge, hydrophobic) systems typically operate at equilibrium, which only allows for
tethering approaches. For more challenging targets such as affinity (Kd) determination, fluidics-based systems such as SPR
membrane-bound receptors, iterative rounds of point mutations also provide the opportunity for kinetic readout (kon, koff) and
have sometimes been successful.19 While many native receptors thus offers higher information content. However, even for
are unstable and rapidly denature at the sensor surface, platforms with the same optical transducer, e.g. SPR, the
thermostabilized mutants of the same receptors can retain balance between sensitivity and throughput is crucial. Although
their functionality and stability in a surface-tethered config- “regular” SPR is unrivaled in terms of sensitivity, imaging SPR22
uration for a number of days. Another commonly employed offers accelerated throughput but at the expense of sensitivity,
strategy is the use of truncated proteins containing only the thus making the latter technology less attractive to small
domain of interest that is targeted with small molecules. In such molecule research. In order to address the sensitivity issue,
cases, it is very important to closely monitor that those some SPR imaging platforms are making use of chemical
variations and alterations do show similar ligand binding microarrays, which have shown to be particular useful for
patterns as to the native protein. Observed discrepancies could fragment screening.23 While following similar principles for
indicate a change in pharmacology when going into an in vivo sensitivity enhancement as the ISA, kinetic information is more
model to test the target hypothesis and thus need to be challenging to assess in this assay configuration.
12 DOI: 10.1021/acssensors.6b00735
ACS Sens. 2017, 2, 10−15
ACS Sensors Sensor Issues

Whereas throughput and sensitivity in addition to robustness incorporated into the lipid layer of fluorescently labeled
has continuously improved over time, all biosensor platforms liposomes. Using a standard TIRF microscope, the binding of
suffer from different limitations that are intrinsic to their build the target−liposome complex to the surface can be monitored
up. Besides the sensitivity limit that is dictated by the molecular with single molecule resolution.With each liposome containing
mass of the interactants, also throughput can be surprisingly at maximum a single copy of the target protein, each observed
affected by system-related factors like binding kinetics. As binding event reflects the interaction of a single target protein
optimized compounds gain dramatically in potency by several with a single tool compound at the surface. The ability to
orders of magnitude once progressed from an initial hit toward monitor each binding and release event at the surface under
a candidate drug, they frequently display residence times of equilibrium conditions allows for an estimation of association
hours or even days. For a reliable determination of binding and dissociation rates from a single experiment without
kinetics, the measurement time thus needs to be extended24 microfluidics or alternative liquid handling.29
and a complete dissociation is required to present a compound- This has many practical implications such as unrivaled
free biosensor for subsequent experiments. Even with sensitivity and thus provides a good rationale to eventually
throughput-enhancing strategies like single-cycle kinetics,25 it succeed existing biosensor platforms. The requirements on the
can easily take 12−24h to obtain affinity and binding kinetics tethered tool compounds are less stringent, as low affinity is not
data for a single compound. only tolerated but often even desirable, as it increases the
BLI can address some of those limitations, as it elegantly number of observable binding events in a given time frame due
combines some advantageous features of SPR and OWG by
to faster dissociation. Additionally, the use of tool compounds
offering a plate-based readout with the option for kinetic
in conjunction with single molecule sensitivity furnishes the
measurements.26 A range of biosensor tips (8−16) are dipping
opportunity to distinguish specific from nonspecific binding
into an analyte solution that is presented in a plate that can be
agitated to mimic a fluidics-based scenario for kinetic events, as they will show different kinetic profiles that can be
measurements. Those sensors can be discarded afterwards to deconvoluted. This is essentially impossible with technologies
enable a new round of measurements, thereby eliminating the that are based on ensemble measurements like SPR, OWG, or
need to wait for complete compound dissociation. This strive BLI. Furthermore, the use of liposomes offers the possibility to
for parallelization has also seen some recent advances in the study ligand binding even to labile membrane proteins in an
development of throughput-enhanced SPR platforms, enabling almost nativelike environment, without the need of extensive
eight analyte evaluations in parallel. protein purification or stability-enhancing mutations.29 This
Despite extensive optimization of protein reagents and assay strategy does not exclude soluble proteins, as they can be
conditions, it can sometimes be challenging to make a clear-cut tethered to liposomes following similar strategies as for
assessment of the binding specificity. While it is straightforward traditional biosensor experiments.
to identify compounds that deviate from the expected binding Finally and most importantly, performing experiments at the
behavior in terms of saturable, reversible binding following a single molecule level has surprising consequences for the limit
1:1 binding model,27 this is often much more difficult to of detection and thus the sensitivity. By increasing the
achieve for compounds that specifically engage with the target. observation time one is able to detect more binding and
In this case, one is forced to conduct additional experiments to release events to the biosensor. Therefore, the lower limit of
look for competition with known, well-characterized tool detection is constantly decreasing with increasing measurement
compounds or to perform a subsequent ISA experiment as time and is not a fixed and platform-dependent parameter
previously described. This obviously requires readily access to anymore.


such tool compounds, which preferably display high potency
and/or can be modified to accommodate a suitable linker for CONCLUSION AND OUTLOOK
directed tethering. Both approaches come at the cost of
additional reagent consumption and extended timelines for Over recent years, optical biosensors have been positioned as a
decision-making. vital part in small molecule drug discovery. The continuous


improvements of existing technologies have expanded their
NEXT-GENERATION BIOSENSOR PLATFORMS application areas significantly. For example, state-of-the-art SPR
ADDRESSING DRUG DISCOVERY NEEDS systems have now reached a level of sensitivity that readily
One common denominator in the experimental workflow of allows direct monitoring of small molecule interactions even at
the discussed biosensor platforms is the need for two separate suboptimal assay conditions (e.g., only a fraction of the tethered
experiments. First, the sensor surface is brought into contact target is functional). However, new platforms are also starting
with the analyte. Thereby, the convoluted association and to emerge in the drug discovery arena. One example is single
dissociation reaction is measured. Typically, this reaction is molecule microscopy, where evanescent-field illumination
observed until equilibrium coverage of the sensor surface is combined with surface functionalization provides a readout at
reached. Subsequently, the sensor surface is exposed to an the level of individual molecules. This provides unique
analyte-free solution to monitor the dissociation reaction alone. opportunities such as the possibility to monitor binding
The possibility of simultaneous quantification of association kinetics at equilibrium in a plate-based format, thereby
and dissociation rates would obviously represent a great combining the benefits from plate-based (throughput) and
technology enhancement, which can be realized with single flow-based systems (kinetics) in one platform. Another
molecule biosensor platforms. advantage lies in the extremely high sensitivity, which reduces
Such novel biosensor platforms and their potential value for protein consumption and allows measurements across a wide
drug discovery have recently been described by making use of affinity range. This together with the significantly reduced
the ISA configuration.28 While the tool compound is tethered reagent consumption furnishes the opportunity to generate
to the biosensor surface, the target protein is attached to or unique value for small molecule drug discovery.
13 DOI: 10.1021/acssensors.6b00735
ACS Sens. 2017, 2, 10−15
ACS Sensors Sensor Issues

■ AUTHOR INFORMATION
Corresponding Author
(13) Cook, D.; Brown, D.; Alexander, R.; March, R.; Morgan, P.;
Satterthwaite, G.; Pangalos, M. N. Lessons learned from the fate of
AstraZeneca’s drug pipeline: a five-dimensional framework. Nat. Rev.
*E-mail: stefan.geschwindner@astrazeneca.com. Drug Discovery 2014, 13 (6), 419−31.
ORCID (14) Williams, D. K.; Wang, J.; Papke, R. L. Positive allosteric
Stefan Geschwindner: 0000-0002-2154-8345 modulators as an approach to nicotinic acetylcholine receptor-targeted
Author Contributions therapeutics: advantages and limitations. Biochem. Pharmacol. 2011, 82
(8), 915−30.
The manuscript is written through contributions of all authors.
(15) Spurny, R.; Debaveye, S.; Farinha, A.; Veys, K.; Vos, A. M.;
All authors have given approval to the final version of the Gossas, T.; Atack, J.; Bertrand, S.; Bertrand, D.; Danielson, U. H.;
manuscript. Tresadern, G.; Ulens, C. Molecular blueprint of allosteric binding sites
Notes in a homologue of the agonist-binding domain of the alpha7 nicotinic
The authors declare no competing financial interest. acetylcholine receptor. Proc. Natl. Acad. Sci. U. S. A. 2015, 112 (19),

■ ACKNOWLEDGMENTS
We would like to acknowledge all colleagues within the global
E2543−52.
(16) Vilums, M.; Zweemer, A. J.; Yu, Z.; de Vries, H.; Hillger, J. M.;
Wapenaar, H.; Bollen, I. A.; Barmare, F.; Gross, R.; Clemens, J.;
Krenitsky, P.; Brussee, J.; Stamos, D.; Saunders, J.; Heitman, L. H.;
AstraZeneca Biophysics community for inspiration and their Ijzerman, A. P. Structure-kinetic relationships–an overlooked param-
continuous strive to unleash the full potential and maximise the eter in hit-to-lead optimization: a case of cyclopentylamines as
value of optical biosensors in drug discovery. Furthermore, we chemokine receptor 2 antagonists. J. Med. Chem. 2013, 56 (19), 7706−
would like to thank the internal AstraZeneca postdoc program 14.
for the great support it has given to Tim Kaminski and Anders (17) Dahl, G.; Akerud, T. Pharmacokinetics and the drug-target
Gunnarsson. In addition, we want to thank Helen Boyd for residence time concept. Drug Discovery Today 2013, 18 (15−16),
input on the manuscript. 697−707.


(18) Geschwindner, S.; Ulander, J.; Johansson, P. Ligand Binding
ABBREVIATIONS Thermodynamics in Drug Discovery: Still a Hot Tip? J. Med. Chem.
2015, 58 (16), 6321−35.
BLI, biolayer inteferometry; DBA, direct binding assay; FBDD, (19) Congreve, M.; Rich, R. L.; Myszka, D. G.; Figaroa, F.; Siegal, G.;
fragment-based drug discovery; HTS, high-throughput screen- Marshall, F. H. Fragment screening of stabilized G-protein-coupled
ing; ISA, inhibition in solution assay; MoA, mode of action; receptors using biophysical methods. Methods Enzymol. 2011, 493,
OWG, optical waveguide grating; SPR, surface plasmon 115−136.
resonance; TIRF, total internal reflection fluorescence


(20) Chan, L. L.; Lidstone, E. A.; Finch, K. E.; Heeres, J. T.;
Hergenrother, P. J.; Cunningham, B. T. A Method for Identifying
REFERENCES Small-Molecule Aggregators Using Photonic Crystal Biosensor
(1) Cooper, M. A. Optical biosensors in drug discovery. Nat. Rev. Microplates. JALA (1998-2010) 2009, 14 (6), 348−359.
Drug Discov 2002, 1 (7), 515−28. (21) Karlsson, R.; Kullman-Magnusson, M.; Hamalainen, M. D.;
(2) Geschwindner, S.; Carlsson, J. F.; Knecht, W. Application of Remaeus, A.; Andersson, K.; Borg, P.; Gyzander, E.; Deinum, J.
optical biosensors in small-molecule screening activities. Sensors 2012, Biosensor analysis of drug-target interactions: direct and competitive
12 (4), 4311−23. binding assays for investigation of interactions between thrombin and
(3) Myszka, D. G.; Rich, R. L. Implementing surface plasmon thrombin inhibitors. Anal. Biochem. 2000, 278 (1), 1−13.
resonance biosensors in drug discovery. Pharm. Sci. Technol. Today (22) Wong, C. L.; Olivo, M. Surface Plasmon Resonance Imaging
2000, 3 (9), 310−317. Sensors: A Review. Plasmonics 2014, 9 (4), 809−824.
(4) Cunningham, B. T.; Li, P.; Schulz, S.; Lin, B.; Baird, C.; (23) Neumann, T.; Junker, H. D.; Schmidt, K.; Sekul, R. SPR-based
Gerstenmaier, J.; Genick, C.; Wang, F.; Fine, E.; Laing, L. Label-free fragment screening: advantages and applications. Curr. Top. Med.
assays on the BIND system. J. Biomol. Screening 2004, 9 (6), 481−490. Chem. 2007, 7 (16), 1630−42.
(5) Wartchow, C. A.; Podlaski, F.; Li, S.; Rowan, K.; Zhang, X.; Mark, (24) Katsamba, P. S.; Navratilova, I.; Calderon-Cacia, M.; Fan, L.;
D.; Huang, K. S. Biosensor-based small molecule fragment screening Thornton, K.; Zhu, M.; Bos, T. V.; Forte, C.; Friend, D.; Laird-
with biolayer interferometry. J. Comput.-Aided Mol. Des. 2011, 25 (7), Offringa, I.; Tavares, G.; Whatley, J.; Shi, E.; Widom, A.; Lindquist, K.
669−76. C.; Klakamp, S.; Drake, A.; Bohmann, D.; Roell, M.; Rose, L.;
(6) Fang, Y. Label-free cell-based assays with optical biosensors in Dorocke, J.; Roth, B.; Luginbuhl, B.; Myszka, D. G. Kinetic analysis of
drug discovery. Assay Drug Dev. Technol. 2006, 4 (5), 583−95. a high-affinity antibody/antigen interaction performed by multiple
(7) Danielson, U. H. Fragment Library Screening and Lead Biacore users. Anal. Biochem. 2006, 352 (2), 208−21.
Characterization Using SPR Biosensors. Curr. Top. Med. Chem. (25) Karlsson, R.; Katsamba, P. S.; Nordin, H.; Pol, E.; Myszka, D. G.
2009, 9 (18), 1725−1735. Analyzing a kinetic titration series using affinity biosensors. Anal.
(8) Erlanson, D. A.; Fesik, S. W.; Hubbard, R. E.; Jahnke, W.; Jhoti, Biochem. 2006, 349 (1), 136−47.
H. Twenty years on: the impact of fragments on drug discovery. Nat. (26) Concepcion, J.; Witte, K.; Wartchow, C.; Choo, S.; Yao, D. F.;
Rev. Drug Discovery 2016, 15 (9), 605−19. Persson, H.; Wei, J.; Li, P.; Heidecker, B.; Ma, W. L.; Varma, R.; Zhao,
(9) Folmer, R. H. Integrating biophysics with HTS-driven drug L. S.; Perillat, D.; Carricato, G.; Recknor, M.; Du, K.; Ho, H.; Ellis, T.;
discovery projects. Drug Discovery Today 2016, 21 (3), 491−8. Gamez, J.; Howes, M.; Phi-Wilson, J.; Lockard, S.; Zuk, R.; Tan, H.
(10) Bergsdorf, C.; Ottl, J. Affinity-based screening techniques: their Label-Free Detection of Biomolecular Interactions Using BioLayer
impact and benefit to increase the number of high quality leads. Expert Interferometry for Kinetic Characterization. Comb. Chem. High
Opin. Drug Discovery 2010, 5 (11), 1095−107. Throughput Screening 2009, 12 (8), 791−800.
(11) Moree, B.; Connell, K.; Mortensen, R. B.; Liu, C. T.; Benkovic, (27) Giannetti, A. M.; Koch, B. D.; Browner, M. F. Surface plasmon
S. J.; Salafsky, J. Protein Conformational Changes Are Detected and resonance based assay for the detection and characterization of
Resolved Site Specifically by Second-Harmonic Generation. Biophys. J. promiscuous inhibitors. J. Med. Chem. 2008, 51 (3), 574−80.
2015, 109 (4), 806−15. (28) Gunnarsson, A.; Snijder, A.; Hicks, J.; Gunnarsson, J.; Hook, F.;
(12) Kozma, P.; Kehl, F.; Ehrentreich-Forster, E.; Stamm, C.; Bier, F. Geschwindner, S. Drug discovery at the single molecule level:
F. Integrated planar optical waveguide interferometer biosensors: a inhibition-in-solution assay of membrane-reconstituted beta-secretase
comparative review. Biosens. Bioelectron. 2014, 58, 287−307. using single-molecule imaging. Anal. Chem. 2015, 87 (8), 4100−3.

14 DOI: 10.1021/acssensors.6b00735
ACS Sens. 2017, 2, 10−15
ACS Sensors Sensor Issues

(29) Gunnarsson, A.; Dexlin, L.; Wallin, P.; Svedhem, S.; Jonsson, P.;
Wingren, C.; Hook, F. Kinetics of ligand binding to membrane
receptors from equilibrium fluctuation analysis of single binding
events. J. Am. Chem. Soc. 2011, 133 (38), 14852−5.

15 DOI: 10.1021/acssensors.6b00735
ACS Sens. 2017, 2, 10−15

You might also like