You are on page 1of 10

Biodrugs 2005; 19 (6): 383-392

TECHNOLOGY 1173-8804/05/0006-0383/$34.95/0

© 2005 Adis Data Information BV. All rights reserved.

Application of Scintillation Proximity Assay in


Drug Discovery
Shaogui Wu1,2 and Bailing Liu1
1 Chengdu Institute of Organic Chemistry, Chinese Academy of Sciences, Chengdu, China
2 Graduate School of the Chinese Academy of Sciences, Beijing, China

Contents
Abstract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 383
1. The Principle of Scintillation Proximity Assay (SPA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 384
2. The Choice of Radioisotope . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 384
3. The Support Materials Used in SPA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 384
4. Scintillant Quenching . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 385
5. Applications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 386
5.1 Receptor-Ligand Binding Assay . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 386
5.2 Application in Drug Screening . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 386
5.3 Application in Radioimmunoassays . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 387
5.4 Application in Enzymatic Activity Measurement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 388
5.5 SPA Based on Other Radioisotopes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 389
5.6 Latest Advances . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 390
6. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 390

Abstract Scintillation proximity assay (SPA), characterized by its speed, sensitivity, reliability, and the fact that no
separation step is required, has become an important technique in high-throughput screening (HTS) for new
drugs, and for investigating their biological interactions. The SPA technique now plays a key role in HTS, in that
it can be used in many assay formats including radioimmunoassays (RIAs), ligand-receptor binding assays, and
enzyme assays. The SPA-based enzyme assay is usually designed in three formats corresponding to different
enzymes: signal removal format for hydrolytic enzymes, signal addition format for polymerase and transferase
enzymes, and product capture format for antibodies, DNA probes, receptors or other specific binding proteins.
The use of SPA in RIAs has been facilitated by new carriers, such as membranes that can be configured in
various shapes and sizes, allowing the assay to be performed on samples from many sources including tissue,
serum, plasma or cells. This review presents the principles of SPA, discusses supporting materials and quenching
effects, as well as detailed examples of the latest advances.

Modern high-throughput screening (HTS) technology for drug format. This assay technique was initially developed by Hart and
discovery and many other applications is a multidisciplinary field Greenwald in 1979[2] and has become a mature and well estab-
involving biology, biochemistry, molecular biology, analytical lished detection approach in HTS for new drug discovery. Com-
chemistry, synthetic chemistry, automation engineering, and com- mon therapeutic targets for HTS are enzymes, cell-surface recep-
puter science.[1] Scintillation proximity assay (SPA) is an emerg- tors, nuclear receptors, ion channels, and signal transduction pro-
ing technology that enables radioactive assays in high-throughput teins. Compounds that interact with these targets are usually
384 Wu & Liu

identified using in vitro biochemical assays.[3] Significant im- detected and no physical separation, such as centrifugation, wash-
provements in combinatorial chemistry over the last several years ing, and filtration, is necessary, making the operation more conve-
have enabled a large number of chemical compounds to be synthe- nient and easily amenable to automation than other binding assay
sized.[4-6] Meanwhile, there has also been a tremendous increase in techniques.[20-22]
the number of possible targets after completion of the Human
Genome Project (http://www.ornl.gov/sci/techresources/ 2. The Choice of Radioisotope
Human_Genome/home.shtml), providing both opportunities and For more than 50 years, biologists have successfully used
requirements for high-throughput assays.[7-10] These two factors radioactive isotopes (3H, 14C, 32P, 35S, and 125I) to study in vivo
have promoted the application of SPA in HTS,[11] and the SPA metabolism, distribution, and binding of small amounts of com-
technique has been widely applied to radioimmunoassays,[12,13] pounds.[23] Because their radiation energy is relatively strong, a
receptor-ligand binding assays,[14,15] enzymatic activity as- tiny amount will meet the requirement for an assay. However,
says,[15-18] RNA transcript detection,[19] protein-peptide interac- disadvantages include harmful radioactive rays, impracticality of
tions, among others. Beyond all of these, SPA has also highlighted long-term storage, and experiments involving compounds with
many possibilities for the design of new detection techniques in very short half-lives.
the future. Without doubt, new advances in SPA would further its The path length of the β particle is determined by its energy,
applications in HTS and related fields. and varies for different isotopes (see table I). For example, β
SPA is a solid phase homogeneous technique. A major advan- particles from 3H have an average path-length of approximately
tage of SPA over the conventional assay techniques is the conve- 1.5μm,[24] which easily meets the distance requirement for SPA.
nient operation, because no separation step is needed in the whole The Auger electrons emitted by 125I, which have energies in the
process of the assay and the basic assay relies on a ‘mix and read’ same range as β particles and travel between 1 and 17.5μm in
format. Consequently, the homogeneous format makes this assay aqueous media, also satisfy the distance requirement. Therefore
more simple, robust, and amenable for automation than a 3H and 125I are ideal isotopes for labeling ligands in SPA.
filter-based assay. SPA also offers high detection sensitivity and Other isotopes of interest, such as 14C, 32P, and 35S, have
can be operated in a 96- or 384-well format with minute sample longer path-lengths with mean ranges of approximately 58, 126,
volumes (5–150μL). Additionally, automated operation (using and 66μm, respectively, and are less suited to application of the
automated pipeting) increases the assay throughput. Therefore, proximity principle. Although SPA has recently been adapted to
SPA permits the design of high-flux assay formats for screening a utilize 35S and 32P,[25-28] the use of 14C is limited because the
large number of compounds in chemical collections. In this article, specific activities of 14C-labeled compounds are generally too low
the basic principles of SPA are reviewed and examples of its (approximately 60 ci/mmoL),[20] even though the path-length of
applications in HTS for drug discovery and other purposes are 14C is less than that of 35S. Nonetheless, some scientists have
described. successfully applied 14C in SPA (see section 5.5).

1. The Principle of Scintillation Proximity Assay (SPA) 3. The Support Materials Used in SPA

The core of this technique is the support material: the SPA


When a radioisotope such as tritium decays, it releases high-
bead. The most commonly used SPA beads are polyvinyltoluene
energy β particles, which have a limited transferring path-length in
aqueous solutions (1.5μm for 3H and ~17.5μm for 125I Auger a b

electrons) [figure 1]. If the radioligand is not ‘captured’ onto the β emission
SPA bead (figure 1a), energy from radioactive decay is dissipated
β emission
into the aqueous environment and gives no light emission, leaving
the disintegration undetected. Conversely, if the radioligand is SPA bead SPA bead

captured on the surface of the SPA bead (figure 1b), the radioiso-
tope is brought into close proximity to the scintillant and the β
particles effectively transfer their energy to the scintillant, result-
ing in light emission that can be detected by liquid scintillation Fig. 1. The principle of scintillation proximity assay (SPA). (a) Free: no
counters.[3] Hence, the bound and free radiolabeled ligand can be signal generated; (b) bound: signal generated.

© 2005 Adis Data Information BV. All rights reserved. Biodrugs 2005; 19 (6)
Application of Scintillation Proximity Assay in Drug Discovery 385

Table I. Radioactive isotopes in common use


Isotope characteristic 3H 14C 32P 35S 125I

Half-life 12.32 (y) 5730 (y) 14.3 (d) 87.4 (d) 60 (d)
Radioactive rays β β β β Auger electron
Average energy (keV) 5.7 49 690 49 32
Transmission distance in air (cm) 0.6 22 7.2 24 2.4
Path-length in aqueous solution (μm) 1.5 58 126 66 1–17.5

(PVT) beads (diameter ~5μm; density ~1.05 g/cm3) and yttrium which contribute to background noise. Therefore, these plates are
silicate (YSi) beads, which are commercially available from also accommodated to SPA based on other long path-length iso-
Amersham Biosciences (GE Healthcare).[29-31] High-efficiency topes such as 33P, 35S, etc.
scintillant has been incorporated inside the PVT matrix, which can
be excited by the high-energy β emission generated from the decay
4. Scintillant Quenching
of radioactive isotopes when the SPA bead is within the effective
path length for energy transference.[32] The surface of this SPA
bead is coated with hydrophilic polyhydroxy film that reduces Although SPA is an accepted and versatile technique, the
hydrophobicity of the bead to reduce nonspecific interactions. This quenching effect is a serious shortcoming. Some chemical or
film has been chemically derived to covalently couple generic colored samples absorb a proportion of the light emission from the
capture molecules, which can selectively bind molecules of inter- scintillator, which causes a severe reduction (quenching) of signal
est including receptors, proteins, or other disease targets. SPA and subsequently interferes with the efficiency of the scintillation
counter. Two types of quenching occur with SPA: chemical
beads with the following capture molecules are available commer-
quenching and color quenching.
cially: protein A, avidin, streptavidin, wheatgerm agglutinin
Chemical quenching takes place when the energy is transfer-
(WGA), glutathione, and various polyclonal secondary antibo-
ring between solvent and scintillator. Some chemical quenching
dies.[33,34] All of these capture molecules are routinely used as one
agents adsorb a part of the energy and give off heat that is then
member of a detection-pair system. These beads are easily pipetted
dissipated in the medium, which reduces the transformation effi-
using automated liquid handling devices into 96-well plates and
ciency of energy to light emission. However, the effect of chemi-
therefore are easily accommodated in HTS operation. Because
cal quenching is slight and can be ignored in many conditions
PVT beads have a lower density than YSi beads, they can remain
(such as in YSi-based SPA).
in suspension longer, which is beneficial for automated HTS.
Color quenching takes place when light emitted by the stimulat-
However, some ligands bind nonspecifically to the surface of PVT
ed scintillator is absorbed or dissipated by the colored solution.
beads and may increase the background noise. The heavier density
Most colored or fluorescent compounds, especially dyes, have
of YSi beads allows the beads to settle for bottom counting, which
absorption spectra that overlap the emission spectra of the scintil-
can reduce nonspecific interference.
lant, which results in quenching of the light emission.
Other support materials include scintillating microplates, in The degree of quenching can be calculated by analyzing the
which scintillant is directly incorporated into the polymeric matrix quenched and unquenched samples. The standard curve can be
or coated on the inner surface of the wells.[32,35,36] Two kinds of made from a series of standards of the same activity but with
these plates, Flash-plate® 1 and Scintistrip-plate® are commercial- different quenching levels. Therefore, it is possible to compensate
ly available from NEN Life Science Products (Boston, MA, USA) for and correct the quenching effect. A color quench correction
and Wallac-Oy (Turku, Finland), respectively. When SPA is car- program can be applied to automatically overcome this issue by
ried out in these microplates, it is necessary to remove all liquid the appropriate scintillation counter. If some compounds with
from the wells. With an appropriate washing/air-drying procedure false positive activity are picked up by SPA, further assays, such
before measuring (but not a ‘mix and measure’ technique) these as absorbance or filtration-based radioactive assays, will be neces-
plates offer the advantage of minimizing nonproximity effects, sary to eliminate them and identify the genuine ‘hits’.

1 The use of trade names is for product identification purposes only and does not imply endorsement.

© 2005 Adis Data Information BV. All rights reserved. Biodrugs 2005; 19 (6)
386 Wu & Liu

a
The ligand binding and protein coating are achieved by mixing
ligand, protein, and SPA beads in sequence. No additional steps
β emission
such as pre-coating and washing procedures are required. For the
+
purpose of measuring the quantitative relation in interactions,
equation 1 was derived to accurately determine the binding affini-
ty for non-tight and tight binding inhibitors. For non-tight binding
Receptor Radioligand
inhibitors, the binding affinity Ki can be calculated from its
relation to IC50 (the concentration of inhibitor at which 50% of the
radioligand binding is reduced).
+ IC50
Ki =
1 + L/KL
Inhibitor (Eq. 1)
For tight binding inhibitors, Ki can be obtained by fitting a plot
b of RL/RL0 versus [I0] in light of equation 2:
R+L RL RL − (Kiapp + I0 − R0) + (Kiapp + I0 − R0)2 + 4 × R0 × Kiapp
+ KL =
I RL0 2 × R0
Ki (Eq. 2)
RI where = Ki × (1 + L0/KL).
Kiapp
Fig. 2. Schematic diagram of a competitive binding assay (reproduced This quantitative assay provided an important theoretical basis
from Sun et al.,[39] with permission from Elsevier). I = inhibitor; Ki = equilibri-
for high-throughput assays to screen chemical structures in a
um dissociation constant for I; KL = equilibrium dissociation constant for L;
L = radioligand; R = receptor; RI = inhibitor-bound receptor; RL = radioli- chemical library.
gand-bound receptor.
5.2 Application in Drug Screening
5. Applications
The SPA technique has been used successfully in HTS for drug
discovery. Many novel chemical structures have been identified as
5.1 Receptor-Ligand Binding Assay ‘hits’ or active molecules, which can be developed into potential

[3H]-dihydronovobiocin
Conventional receptor-ligand binding assays are often complex
hv
and time consuming,[23,37,38] but the application of SPA can greatly
GyrB43
facilitate this type of assay. Receptors are directly immobilized
onto the surface of SPA beads via a number of coupling approach- β emission
es. When a suitable radioligand binds to a receptor, it will be in
proximity to stimulate the bead to emit light. Unbound ligands are
too distant from the beads to transfer their energy and thus are not
SPA bead
*
Av-Bio
detected. Sun and colleagues[39] described an optimization of a
generic binding assay to determine the ligand-receptor interactions
based on SPA. They immobilized peroxisome proliferator-activat-
ed receptors (PPARs) on the surface of SPA beads, and light Fig. 3. Schematic diagram of the gyrase B (GyrB) subunit scintillation
emission was generated by excitation of the scintillant inside the proximity assay (SPA). An ATP-binding site is located in GyrB43 and both
coumarin and cyclothialidine antibacterials act through binding to it. When
bead as a result of binding of a radiolabeled ligand to immobilized radiolabeled inhibitors such as [3H] dihydronovobiocin bind to this site, the
PPARs. The intrinsic binding affinity of unlabeled ligands is radioisotope is in proximity to the SPA bead, and the scintillant inside the
bead is excited by the β particles from the radioactive decay to emit light. A
determined by competitive displacement of the radioligand (figure
scintillation counter can be used to record the emission signal (reproduced
2a). The binding equilibrium of receptor, radioligand, and inhibi- from Gevi and Domenici,[50] with permission from Elsevier). Av-Bio = avi-
tor can be established as shown in figure 2b. din-biotin; hv = photon emission.

© 2005 Adis Data Information BV. All rights reserved. Biodrugs 2005; 19 (6)
Application of Scintillation Proximity Assay in Drug Discovery 387

5.3 Application in Radioimmunoassays


Membrane containing
secondary antibody
The application of the SPA technique to radioimmunoassays is
referred to as the scintillation proximity radioimmunoassay
[125I]cAMP (SPRIA). Characterized by its speed, simple operation, accepted
cAMP precision, and preferable reproducibility, SPRIA has inherited
2° antibody many advantages of SPA. It is apparent that this high-throughput
1° antibody homogeneous assay is superior to the conventional radioimmu-
noassay that uses ammonium sulfate precipitation as the separa-
tion method. SPRIAs are amenable to automation and processing
of a large number of samples. Such assays may be applied in place
of traditional separation-based radioimmunoassays. Tadepalli and
Quinn[51] applied this technique to quantitate acyclovir in samples
from pre-clinical and clinical studies after the administration of
valaciclovir (a prodrug of acyclovir).[51] Serres et al.[52] used this
technique in determination of phosphrothioate antisense
Fig. 4. Principle of scintillation proximity radioimmunoassay with micro-
porous membranes. A secondary antibody is immobilized in the pore sur- oligonucleotide plasma concentrations.
face of the membranes, onto which a primary antibody was bound. When a SPA beads are also the main supporting materials currently
radioligand such as [125I] cyclic adenosine monophosphate (cAMP) binds
used in SPRIAs. Recently, Mansfield et al.[13] reported a new
to the primary antibody, the scintillant inside the polymeric membrane is
stimulated by the radioactive rays to emit light that can be measured easily SPRIA based on the use of microporous membranes (figure 4).
by a liquid scintillation counter (reproduced from Mansfield et al.,[13] with Organic or inorganic fluors were incorporated into the matrix of
permission from Elsevier). microporous polymeric membranes. A secondary antibody was
immobilized onto their surfaces prior to use. These membranes
drugs with a desired therapeutic activity, such as agonist, antago- were employed in competitive binding assays in which a separa-
nist, antibacterial, etc.[40-45] Recently, much attention has been
focused on the development of novel coumarin and noncoumarin
antibacterials.[46-48] Bacterial DNA gyrase is the site of action of


Cleavage site
naturally occurring antibacterials, including the broad-spectrum
SPA bead
coumarin antibiotics and the cyclic peptide inhibitors. They both
act through binding to a site located on the gyrase B (GyrB)
+ Protease + Protease
subunit and consequent inhibition of the adenosine triphosphatase inhibitor
(ATPase) reaction. The principle of the GyrB SPA is illustrated in
figure 3. A radiolabeled ligand such as [3H] dihydronovobiocin
binds to GyrB, which is in turn captured by the SPA bead via the
streptavidin-biotin (Av-Bio) system. The radioactive decay is in
SPA bead
Av-Bio
+

proximity of the bead, which results in the excitation of the
scintillant to emit photons that can be measured by a scintillation Fig. 5. Schematic diagram of the principle of scintillation proximity assay
(SPA) used in detecting human cytomegalovirus (HCMV) protease inhibi-
counter. From this assay, IC50s of 42, 64, and 11nM were obtained
tor. The peptide substrate (the thick horizontal black line) is attached to the
for novobiocin, dihydronovobiocin, and the cyclothialidine analog SPA bead through a streptavidin-biotin (Av-Bio) system, which contains a
GR 122222X, respectively, which is consistent with the results cleavage site (black rectangle). The peptide is phosphorylated with radio-
labeled adenosine triphosphate (ATP) at the serine residue near its car-
from other methods.[49] However, the SPA technique is rapid,
boxyl terminus. The scintillant inside the SPA bead is stimulated by the
robust, and reliable compared with conventional HTS techniques. radioactive decay and a high radioactive signal can be detected. The
This assay can be conducted in 96-well (384-well or more) micro- cleavage of the peptide by HCMV separates the phosphate label from the
part related to the SPA bead and the counts per minute (CPM) will de-
titer plates and no additional separation steps are required, which
crease. However, the presence of protease inhibitors can prevent this
greatly facilitates drug discovery efforts and is amenable for cleavage. The radioactive signal changes can be used to detect protease
automated HTS of large chemical collections for novel drugs. inhibitors (reproduced from Baum et al.,[25] with permission from Elsevier).

© 2005 Adis Data Information BV. All rights reserved. Biodrugs 2005; 19 (6)
388 Wu & Liu

tion procedure was not necessary for bound and unbound radioli- formats of SPA enzyme assays have been developed: signal re-
gands.[13] The results were nearly identical to those obtained using moval, signal addition, and product capture. In the first format
the standard radioimmunoassay, although less time was used and (signal removal), the radiolabeled substrate is biotinylated to the
the assay procedure was simplified greatly. This membrane-based SPA bead, which results in the scintillation of the bead. When the
SPRIA is applicable in the detection of various radioanalytes and catalysis of a specific enzyme separates the radioactive moiety
can be configured in a number of sizes and shapes. from the biotinylated portion of the substrate molecule, the light
signal will decrease. This assay is suitable for hydrolase activities
5.4 Application in Enzymatic Activity Measurement such as those of proteases, nucleases, phospholipases, and ester-
ases.
Conventional approaches for quantitating enzymatic activity
In the signal addition format, which is suitable for molecular
are often time consuming, labor intensive, or not sensitive enough,
transfer activities (e.g. transferases, polymerases, and kinases), the
especially for high-throughput studies. These limitations prompted
acceptor substrate is biotinylated and the donor substrate is radio-
the development of a new, versatile, simple, solid-phase technique
labeled. The radiolabel is transferred from the donor substrate to
based on SPA. SPA has been a routine tool used in enzymatic
the receptor substrate, which is in turn captured by the SPA bead
activity measurement and is well documented in the litera-
and results in the generation of a signal.
ture.[20,53-55] For example, Zhang et al.[56] reported a 96-well homo-
geneous SPA for the study of DnaB-stimulated Escherichia coli The last assay format is product capture, in which the product
primase activity and the identification of E. coli primase inhibi- generated from the action of the enzyme is captured via bio-
tors.[56] Macarron et al.[57] used SPA in the measurement of the specific recognition by antibodies, DNA probes, receptors, or
aminoacyl-tRNA synthetase aminoacylation activity.[57] They other specific binding proteins on the beads. The antibody or probe
found it was faster and more amenable to HTS than traditional must be able to discriminate adequately between the generated
approaches. product and the substrate. The choice of the format and detailed
In SPA enzyme assays, the conversion of the substrate to design for the enzymatic activity assay are in accordance with
product is monitored by either removal or addition of radioisotope actual conditions such as the properties of the enzyme and corre-
to the component, which is captured on the SPA bead. Three basic sponding substrates.

Ph O Ph

R
OH
ST1 R= O ST10 R= O

ST11 R= OH
ST2 R= O O
Et
OH
ST3 R= O ST12 R=
N Et Br
OH Et
ST4 R= O
Et
ST13 R= O
N H Br
ST5 R= O
Et
OH O N
ST14 R= O
O
ST6 R= O O
O
O

ST7 R= O

O OH O
ST8 R= O
O ST15 R=
O O
O
ST9 R= O NH 2
O ST16 R= N
H
Fig. 6. Chemical structures of membrane-soluble scintillators (reproduced from Culliford et al.,[65] with permission from Elsevier). ST = structure.

© 2005 Adis Data Information BV. All rights reserved. Biodrugs 2005; 19 (6)
Application of Scintillation Proximity Assay in Drug Discovery 389

Liposomes
Scintillant molecules

Incorporation

Incubation with
Fusion
HeLa cells
HeLa cell membrane

Influx/incorporation Efflux assay Radioligand binding


assay

Scintillant molecule
Light emitted from scintillant
Movement of substrate
Radiolabeled substrate/ligand
Monolayer of cultured cells

Fig. 7. Schematic diagram of the principle of cell-based scintillation proximity assay following application of membrane soluble scintillators (MSS) to
cultured cells (reproduced from Culliford et al.,[65] with permission from Elsevier). The scintillation of MSS cannot occur in an aqueous environment but can
proceed in a non-aqueous environment (such as lipid bilayer) in the presence of a radioactive decay. MSS can be incorporated into liposomes and then
fused into the membrane of HeLa cells. Therefore, the radiolabeled ligand or substrate in close proximity to the cell membrane (e.g. when the [14C]-
methionine is taken up by the HeLa cells) will cause the light emission of scintillator in the cell membrane, which can be monitored by a scintillation counter.
This system can be used in radiochemical and radioligand binding assays either in vivo or on microsomal preparations obtained from tissues.

5.5 SPA Based on Other Radioisotopes bead-related moiety and results in a concomitant decrease in the
radioactive signal, which is prevented by the presence of a prote-
The path-lengths of the β emissions for 33P and 35S are 126 and ase inhibitor. This new SPA is amenable to other proteases with a
66μm, respectively, which seem less suitable for the application in known cleavage site.
SPA than 3H (1.5μm) and 125I (~17.5μm), because longer path-
The path-length of 35S emission is 66μm, which is more
lengths will often bring about the ‘no-proximity effect’ and se-
applicable for SPA than the 126μm path length of 33P. 35S-based
verely reduce the signal-to-noise ratio. However, Baum et al.[25]
SPA seems to be more common than that of other radioactive
successfully applied the SPA technique in detecting human
cytomegalovirus (HCMV) protease UL80 inhibitor using 33P.[25] isotopes except 3H and 125I.[58-60] Delapp and colleagues[61,62]
This is the first report that 33P is indeed compatible with the SPA reported an antibody-capture [35S] GTPγS SPA and used it in the
system. The peptide substrate used in the assay contains an HCMV analysis of G-protein-coupled receptors (GPCRs) pharmacolo-
protease cleavage site and is biotinylated to the SPA bead at its gy.[61,62]
amino terminus (figure 5). The peptide was also phosphorylated Although the specific activities of 14C-labeled compounds are
with radioactive ATP at its carboxyl terminus. The peptide is generally too low, reports about 14C-based SPA have appeared
incubated with protease prior to binding to SPA beads. Cleavage frequently in the literature in recent years. For example, Williams
of the peptide by the HCMV separates the radiolabel from the SPA and coworkers[63] reported the application of 14C-based SPA for

© 2005 Adis Data Information BV. All rights reserved. Biodrugs 2005; 19 (6)
390 Wu & Liu

analysis of Na+/Cl–-dependent neurotransmitter transporter activi- yet to be resolved include how to reduce the nonspecific binding
ty. Mattingly et al.[64] developed a new membrane-based SPA for and increase the signal-to-noise ratio so as to enhance the reliabili-
detecting and quantifying 14CO2.[64] They used a polymeric mem- ty of the detection result, how to avoid the use of radioisotopes,
brane, which could simultaneously capture and quantify 14CO2 and how to protect the protein/enzyme (acceptor or donor) from
successfully, as the support material. Compared with traditional inactivation due to immobilization or labeling during the assay.
detection approaches for 14CO2, it exhibited obvious advantages Currently, development of non-isotopic detection systems, cell-
such as easy operation, low cost, and minimization of radioactive based assays, and low-cost assays are concentrated on resolving
waste. these disadvantages. It is anticipated that the rapid progress of
SPA will combine with the latest advances in scientific technology
5.6 Latest Advances to continuously improve this technique and expand its applica-
tions.
Further advances in SPA are continuously occurring. A novel
cell-based SPA for studying protein function and activity in vi-
tro was reported by Culliford et al.,[65] Chapman et al.,[66] and Acknowledgments
McCairn et al.[67] They used membrane-soluble scintillants
This project was supported by the National Natural Science Foundation of
(MMS), which have a scintillant ‘head’ group (2,5-diphenylox-
China (20574071).
azole) and a lipophilic ‘tail’ (figure 6). In the presence of a The authors have no conflicts of interest relevant to the contents of this
radioactive source, these MSS have no scintillant activity in an article.
aqueous environment, but do in a non-aqueous environment, such
as a lipid bilayer (e.g. liposome or cell membrane). MSS li-
posomes are incorporated and fused with the plasma membranes References
1. Liu BL, Li SJ, Hu J. Technological advances in high-throughput screening. Am J
of cells in culture and can be excited by radiolabeled molecules in Pharmacogenomics 2004; 4: 263-76
close proximity to the cell membrane to elicit a scintillation signal. 2. Hart HE, Greenwald EB. Scintillation proximity assay (SPA): a new method of
immunoassay. Direct and inhibition mode detection with human albumin and
This system has been used to successfully monitor [14C] methio- rabbit antihuman albumin. Mol Immunol 1979; 16: 265-7
nine uptake in HeLa cells, and may be used in radiochemical and 3. Cook ND. Scintillation proximity assay: a versatile high-throughput screening
radioligand binding assays either in vivo or on microsomal prepa- technology. Drug Discov Today 1996; 1 (7): 287-94
4. Arya P, Roth HJ. Combinatorial chemistry. Curr Opin Chem Biol 2005; 9: 229-31
rations obtained from tissues (figure 7). This is obviously a great 5. Weber L. In vitro combinatorial chemistry to create drug candidates. Drug Discov
technical advance in SPA, since even the live cell can be used as Today 2004; 1: 261-7
6. Ganesan A. Natural products as a hunting ground for combinatorial chemistry.
the support material and no protein purification is needed. In other
Curr Opin Biotechnol 2004; 15: 584-90
words, this new improvement not only further simplifies the SPA 7. Ratti E, Trist D. The continuing evolution of the drug discovery process in the
procedure, but also effectively avoids the inactivation that often pharmaceutical industry. Farmaco 2001; 56: 13-9
8. Hurko O. Genetics and genomics in neuropsychopharmacology: the impact on
results from purification and immobilization to the disease target/ drug discovery and development. Eur Neuropsychopharmacol 2001; 11: 491-9
receptor. It can be used in situations where the assays should be 9. Ryu DDY, Nam DH. Biomolecular engineering: a new frontier in biotechnology. J
Mol Catal B Enzym 2000; 10: 23-37
performed in a cellular environment to ensure the activity of
10. Grippo JF, Burn P. Obesity genes: molecular genetic approaches to drug target
proteins. This methodology has been successfully employed in identification. Farmaco 1998; 53: 262-5
radioligand-binding studies and in a variety of flux and uptake 11. Sittampalam GS, Kahl SD, Janzen WP. High-throughput screening: advances in
assay technologies. Curr Opin Chem Biol 1997; 1: 384-91
assays and incorporation assays. This technique is clearly a prom- 12. Kominami G, Nakamura M, Chomei N, et al. Radioimmunoassay for a novel
ising development in the utilization of SPA. benzodiazepine inverse agonist, S-8510, in human plasma and urine. J
Pharmaceut Biomed 1999 Jun; 20 (1-2): 145-53
13. Mansfield RK, Bhattacharyya D, Hartman NG, et al. Scintillation proximity
6. Conclusion radioimmunoassay with microporous membranes. Appl Radiat Isot 1996; 47
(3): 323-8
SPA plays an important role in HTS, radioimmunoassays, 14. Crane K, Shih DT. Development of a homogeneous binding assay for histamine
receptors. Anal Biochem 2004; 335: 42-9
ligand-receptor binding assays, enzyme assays, and so on. Howev-
15. Dautzenberg FM, Huber G, Higelin J, et al. Evidence for the abundant expression
er, SPA has many shortcomings. For example, SPA is a costly of arginine 185 containing human CRF2α receptors and the role of position 185
assay (including the expensive SPA reagents) and scintillation for receptor-ligand selectivity. Neuropharmacology 2000; 39: 1368-76
16. Mádi A, Kárpáti L, Kovacs A, et al. High-throughput scintillation proximity assay
cocktails maybe radioactive and toxic. Disposal of experimental for transglutaminase activity measurement. Anal Biochem 2005 Aug 15; 343
waste is also costly and environmentally unfriendly. Issues that are (2): 256-62

© 2005 Adis Data Information BV. All rights reserved. Biodrugs 2005; 19 (6)
Application of Scintillation Proximity Assay in Drug Discovery 391

17. Latour D, Fung K, Michelotti E, et al. 991 Development of a homogeneous 38. Patel S, Harris A, O’Beirne G, et al. Kinetic analysis of inositol trisphosphate
scintillation proximity assay (SPA) for determining activity of HCV polymer- binding to pure inositol trisphosphate receptors using scintillation proximity
ase and screening for enzyme inhibitors. Hepatology 2003; 38: 633-9 assay. Biochem Biophys Res Commun 1996; 221: 821-5
18. Kumar CC, Yin ZZ, Liu YH, et al. Expression, purification, characterization and 39. Sun SX, Almaden J, Carlson TJ, et al. Assay development and data analysis of
homology modeling of active Akt/PKB, a key enzyme involved in cell survival receptor–ligand binding based on scintillation proximity assay. Metab Eng
signaling. BBA-Gen Subjects 2001; 1526: 257-8 2005; 7 (1): 38-44
19. Liu JW, Feldman PA, Lippy JS, et al. A scintillation proximity assay for RNA 40. Yang F, Dicker IB, Kurilla MG, et al. PolC-type polymerase III of Streptococcus
detection. Anal Biochem 2001; 289 (2): 239-45 pyogenes and its use in screening for chemical inhibitors. Anal Biochem 2002;
20. Skorey KI, Kennedy BP, Friesen RW, et al. Development of a robust scintillation 304 (1): 110-6
proximity assay for protein tyrosine phosphatase 1B using the catalytically 41. Tanitame A, Oyamada Y, Ofuji K, et al. Synthesis and antibacterial activity of
inactive (C215S) mutant. Anal Biochem 2001; 291: 269-78 novel and potent DNA gyrase inhibitors with azole ring. Bioorg Med Chem
21. Mueller JP, Reynolds KA. Development of a scintillation proximity assay for b- 2004; 12: 5515-24
ketoacyl-acyl carrier protein synthase III. Anal Biochem 2000; 282: 107-14 42. DeLapp NW, McKinzie JH, Sawyer BD, et al. Determination of [35S] gua-
nosine-5′-O- (3-thio) triphosphate binding mediated by cholinergic muscarinic
22. Whitfield J, Harada K, Bardelle C, et al. High-throughput methods to detect
receptors in membranes from chinese hamster ovary cells and rat striatum using
dimerization of Bcl-2 family proteins. Anal Biochem 2003; 322 (2): 170-8
an anti-G protein scintillation proximity assay. J Pharmacol Exp Ther 1999; 289
23. Higelin J, Lang GP, Paternoster C, et al. 125I-Antisauvagine-30: a novel and
(2): 946-55
specific high-affinity radioligand for the characterization of corticotropin-
43. Porter AC, Sauer JM, Knierman MD, et al. Characterization of a novel endocan-
releasing factor type 2 receptors. Neuropharmacology 2000; 40 (1): 114-22
nabinoid, virodhamine, with antagonist activity at the CB1 receptor. J
24. Zhou V, Han S, Brinker A, et al. A time-resolved fluorescence resonance energy Pharmacol Exp Ther 2002; 301 (3): 1020-4
transfer-based HTS assay and a surface plasmon resonance-based binding assay
44. Bymaster FP, Falcone JF, Bauzon D, et al. Potent antagonism of 5-HT3 and 5-HT6
for heat shock protein 90 inhibitors. Anal Biochem 2004; 331 (2): 349-57
receptors by olanzapine. Eur J Pharmacol 2001; 430 (2-3): 341-9
25. Baum EZ, Johnston SH, Bebernitz GA, et al. Development of a scintillation
45. Cussac D, Pasteau V, Millan MJ. Characterisation of Gs activation by dopamine
proximity assay for human cytomegalovirus protease using 33Phosphorous.
D1 receptors using an antibody capture assay: antagonist properties of
Anal Biochem 1996; 237 (1): 129-34
clozapine. Eur J Pharmacol 2004; 485 (1-3): 111-7
26. Zaworski PG, Alberts GL, Pregenzer JF, et al. Efficient functional coupling of the
46. Mei HY, Mack DP, Galan AA, et al. Discovery of selective, small-molecule
human D3 dopamine receptor to G(o) subtype of G proteins in SH-SY5Y cells.
inhibitors of RNA complexes: 1. The Tat protein/TAR RNA complexes re-
Br J Pharmacol 1999; 128 (6): 1181-8
quired for HIV-1 transcription. Bioorg Med Chem 1997; 5 (6): 1173-84
27. Yang QD, Qureshi SA, Xie D, et al. Detection of glucagon-dependent GTPgam- 47. Laurin P, Ferroud D, Schio L, et al. Structure-activity relationship in two series of
maS binding in high-throughput format. Anal Biochem 2002 Feb 1; 301 (1): aminoalkyl substituted coumarin inhibitors of gyrase B. Bioorg Med Chem Lett
156-9 1999; 9 (19): 2875-80
28. Simpson PR, Yu XH, Redza ZM, et al. Quantification of hepatitis B virus DNA 48. Laurin P, Ferroud D, Klich M, et al. Synthesis and in vitro evaluation of novel
using competitive PCR and a scintillation proximity assay. J Virol Methods highly potent coumarin inhibitors of gyrase B. Bioorg Med Chem Lett 1999; 9
1997; 69 (1-2): 197-208 (14): 2079-84
29. Pachter JA, Zhang RM, Mayerezell R. Scintillation proximity assay to measure 49. Kyono K, Miyashiro M, Taguchi I. Detection of hepatitis C virus helicase activity
binding of soluble fibronectin to antibody-captured 51 integrin. Anal Biochem using the scintillation proximity assay system. Anal Biochem 1998 Mar 15; 257
1995; 230: 101-7 (2): 120-6
30. McDonald OB, Chen WJ, Ellis B, et al. A scintillation proximity assay for the Raf/ 50. Gevi M, Domenici E. A scintillation proximity assay amenable for screening and
MEK/ERK kinase cascade: high-throughput screening and identification of characterization of DNA gyrase B subunit inhibitors. Anal Biochem 2002 Jan 1;
selective enzyme inhibitors. Anal Biochem 1999; 268 (2): 318-29 300 (1): 34-9
31. Pai JJK, Kirkup MP, Frank EA, et al. Compounds capable of generating singlet 51. Tadepalli SM, Quinn RP. Scintillation proximity radioimmunoassay for the mea-
oxygen represent a source of artifactual data in scintillation proximity assays surement of Acyclovir. J Pharm Biomed Anal 1996; 15: 157-63
measuring phosphopeptide binding to SH2 domains [published erratum appears 52. Serres MD, McNully MJ, Christensen L, et al. Development of a novel scintillation
in Anal Biochem 1999 Dec 15; 276 (2): 262]. Anal Biochem 1999 May 15; 270 proximity competitive hybridization assay for the determination of
(1): 33-40 phosphorothioate antisense oligonucleotide plasma concentrations in a tox-
32. Dayton BD, Chiou WJ, Opgenorth TJ, et al. Direct determination of endothelin icokinetic study. Anal Biochem 1996; 233: 228-33
receptor antagonist levels in plasma using a scintillation proximity assay. Life 53. Zheng W, Carroll SS, Inglese J, et al. Miniaturization of a hepatitis C virus RNA
Sci 2000; 66: 937-45 polymerase assay using a –102°C cooled CCD camera-based imaging system.
33. Zhang J, Wu P, Kuvelkar R, et al. A scintillation proximity assay for human Anal Biochem 2001; 290 (2): 214-20
interleukin-5 (hIL-5) high-affinity binding in insect cells coexpressing hIL-5 54. Nare B, Allocco JJ, Kuningas R, et al. Development of a scintillation proximity
receptor α and β subunits. Anal Biochem 1999; 268 (1): 134-42 assay for histone deacetylase using a biotinylated peptide derived from histone-
34. Jeffery JA, Sharom JR, Fazekas M, et al. An ATPase assay using scintillation H4. Anal Biochem 1999; 267 (2): 390-6
proximity beads for high-throughput screening or kinetic analysis. Anal Bi- 55. Hood CM, Kelly VA, Bird MI, et al. Measurement of α (1-3) fucosyltransferase
ochem 2002; 304 (1): 55-62 activity using scintillation proximity. Anal Biochem 1998; 255 (1): 8-12
35. Sonatore LM, Wisniewski D, Frank LJ, et al. The utility of FK506-binding protein 56. Zhang Y, Yang F, Kao YC, et al. Homogenous assays for Escherichia coli DnaB-
as a fusion partner in scintillation proximity assays: application to SH2 do- stimulated DnaG primase and DnaB helicase and their use in screening for
mains. Anal Biochem 1996; 240: 289-97 chemical inhibitors. Anal Biochem 2002; 304 (2): 174-9
36. Antonsson B, Marshall CJ, Montessuit S, et al. An in vitro 96-well plate assay of 57. Macarron R, Mensah L, Cid C, et al. A homogeneous method to measure aminoa-
the mitogen-activated protein kinase cascade. Anal Biochem 1999; 267: 294-9 cyl-tRNA synthetase aminoacylation activity using scintillation proximity as-
37. Nichols JS, Parks DJ, Consler TG, et al. Development of a scintillation proximity say technology. Anal Biochem 2000; 284 (2): 183-90
assay for peroxisome proliferator-activated receptor γ ligand binding domain 58. Sawyer BD, Silbernagel A, McKinzie JB, et al. Comparison of GTP-γ-35S and 3H-
[published erratum appears in Anal Biochem 1998 Oct 1; 263 (1): 126]. Anal NMS binding in CHO cells stably expressing human M1–M5 muscarinic
Biochem 1998; 257 (2): 112-9 receptors using scintillation proximity assays. Life Sci 1999; 64: 568

© 2005 Adis Data Information BV. All rights reserved. Biodrugs 2005; 19 (6)
392 Wu & Liu

59. Milligan G. Principles: extending the utility of [35S] GTPγS binding assays. 65. Culliford SJ, McCauley P, Sutherland AJ, et al. A novel cell-based scintillation
Trends Pharmacol Sci 2003; 24: 87-90 proximity assay for studying protein function and activity in vitro using
60. Wang XY, Bergdahl K, Heijbel A, et al. Analysis of in vitro interactions of protein membrane-soluble scintillants. Biochem Biophys Res Commun 2002; 296 (4):
tyrosine phosphatase 1B with insulin receptors. Mol Cell Endocrinol 2001; 173 857-63
(1-2): 109-20 66. Chapman RL, Stanley TB, Hazen R, et al. Small molecule modulators of HIV Rev/
61. DeLapp NW, McClure D, McKinzie J, et al. A study of M1 receptor coupling to Rev response element interaction identified by random screening. Antiviral Res
inhibitory G proteins in CHO cells via GTPγS binding determined with anti-G 2002; 54 (3): 149-62
protein antibodies. Life Sci 1999; 64: 569
67. McCairn MC, Culliford SJ, Kozlowski RZ, et al. Synthesis, evaluation and
62. DeLapp NW. The antibody-capture [35S] GTPγS scintillation proximity assay: a
incorporation into liposomes of 4-functionalised-2, 5-diphenyloxazole deriva-
powerful emerging technique for analysis of GPCR pharmacology. Trends
tives for application in scintillation proximity assays. Tetrahedron Lett 2004;
Pharmacol Sci 2004; 25: 400-1
45: 2163-6
63. Williams JB, Mallorga PJ, Lemaire W, et al. Development of a scintillation
proximity assay for analysis of Na+/Cl–-dependent neurotransmitter transporter
activity. Anal Biochem 2003; 321 (1): 31-7
64. Mattingly CD, Mansfield RK, Bhattacharyya D, et al. Membrane-based scintilla- Correspondence and offprints: Professor Bailing Liu, Chengdu Institute of
tion proximity assays: I. Detection and quantification of 14CO2. J Memb Sci Organic Chemistry, Chinese Academy of Sciences, Chengdu 610041, China.
1995; 98: 275-80 E-mail: Blliuchem@hotmail.com

© 2005 Adis Data Information BV. All rights reserved. Biodrugs 2005; 19 (6)

You might also like