You are on page 1of 8

Channels

ISSN: 1933-6950 (Print) 1933-6969 (Online) Journal homepage: http://www.tandfonline.com/loi/kchl20

Role of hERG potassium channel assays in drug


development

Birgit Priest, Ian M. Bell & Maria Garcia

To cite this article: Birgit Priest, Ian M. Bell & Maria Garcia (2008) Role of hERG potassium
channel assays in drug development , Channels, 2:2, 87-93, DOI: 10.4161/chan.2.2.6004

To link to this article: https://doi.org/10.4161/chan.2.2.6004

Copyright © 2008 Landes Bioscience

Published online: 15 May 2008.

Submit your article to this journal

Article views: 3583

Citing articles: 33 View citing articles

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=kchl20
[Channels 2:2, 87-93; March/April 2008]; ©2008 Landes Bioscience hERG assays

Review

Role of hERG potassium channel assays in drug development


Birgit T. Priest,1,* Ian M. Bell2 and Maria L. Garcia1
1Department of Ion Channels; Merck Research Laboratories; Rahway, New Jersey USA; 2Department of Medicinal Chemistry; Merck Research Laboratories; West Point,
Pennsylvania USA

Abbreviations: ECG, electrocardiogram; hERG, human ether-a-go-go related gene; LQTS, long QT syndrome; PPC, population patch
clamp

.
Key words: hERG, arrhythmia, drug development, torsades de pointes, potassium channel, assay development

E
UT
RIB
Numerous structurally and functionally unrelated drugs block What Is hERG?
the hERG potassium channel. HERG channels are involved in

IST
cardiac action potential repolarization, and reduced function of Technically, hERG is the name of a human gene and stands for
hERG lengthens ventricular action potentials, prolongs the QT human ether‑a‑go‑go related gene. The name is based on homology to
interval in an electrocardiogram, and increases the risk for poten‑ a gene found in Drosophila melanogaster that was named ether‑a‑go‑go

D
tially fatal ventricular arrhythmias. In order to reduce the risk (eag) because of the ether‑induced leg shaking observed in flies with
mutations in this gene.1 According to the new nomenclature, hERG

OT
of investing resources in a drug candidate that fails preclinical
safety studies because of QT prolongation, it is important to is KCNH2. The protein encoded by this gene will be referred to as the
screen compounds for activity on hERG channels early in the lead
optimization process. A number of hERG assays are ­available,
ON hERG channel, as is common practice, although the newer nomen-
clature refers to it as Kv11.1.2 The hERG channel is a voltage‑gated
potassium channel and, like other voltage‑gated potassium channels,
ranging from high throughput binding assays on stably expressed
.D
recombinant channels to very time consuming electrophysiological it is highly selective for potassium and is a tetramer formed of four
examinations in cardiac myocytes. Depending on the number subunits, each containing six transmembrane domains (Fig. 1). Two
CE

of compounds to be tested, binding assays or functional assays alternative splice forms, hERG1a and hERG1b, exist, which differ
measuring membrane potential or Rb+ flux, combined with electro‑ only at the N‑terminal domain.3 The two splice forms can assemble
IEN

physiology on a few compounds, can be used to efficiently develop to form functional homo‑ or hetero‑tetramers with somewhat
the structure‑function relationship of hERG ­interactions. different channel kinetics.4 Generally, hERG channels are involved
in action potential repolarization. In humans, the hERG channel
SC

Introduction is expressed widely, including in the brain, adrenal gland, thymus,


retina, and in cardiac and smooth muscle tissues.1,5 In the heart,
BIO

In drug development, it is advantageous to discover potential


safety liabilities associated with a structural series before significant ERG1 channels appear to be the only members of this potassium
resources have been invested. This process involves early counter channel family, whereas two related channels, ERG2 and ERG3, are
screening against related targets, such as a panel of kinases for a expressed in the nervous system.6 Although in heterologous expres-
ES

program targeting p38 kinase or other potassium channels for a sion systems, the hERG channel can associate with the β‑subunits
program directed against a member of this class of ion channels. minK (KCNE1) and MiRP1 (KCNE2),7,8 the existence of such
ND

Undesirable activities that are unrelated to the biological target of complexes in native tissues remains a subject of debate.
interest are much more difficult to address at the early stages and are HERG channels differ from other voltage‑gated potassium
LA

often only discovered during preclinical safety studies, after significant channels in the relative kinetics of activation and inactivation.9 In
resources have been devoted to the program. One off‑target activity hERG channels, the development and reversal of inactivation is
08

that is frequently encountered is block of hERG potassium channels, fast compared to the activation and deactivation steps, leading to a
which has the potential to cause life‑threatening arrhythmias. This is characteristic increase in current upon repolarization as illustrated in
20

a particular troublesome problem due to the fact that block of hERG Figure 2.
may be difficult to detect during routine preclinical and clinical
Why and When to Screen on hERG
©

safety studies. In this work, we discuss the need for hERG screening
and the types of hERG assays that are currently available. In the heart, hERG channels are the molecular correlate of the IKr
current which, together with other potassium currents, is involved
*Correspondence to: Birgit T. Priest; Department of Ion Channels; Merck Research
in action potential repolarization.9,10 Reduced function of hERG
Laboratories; Rahway, New Jersey USA; Email: birgit_priest@merck.com
causes action potential prolongation, which in rare cases can lead
Submitted: 05/02/08; Accepted: 05/05/08 to the potentially fatal ventricular tachyarrhythmia Torsades de
Previously published online as a Channels E-publication: Pointes.11,12 In a body surface electrocardiogram (ECG), ventricular
http://www.landesbioscience.com/journals/channels/article/6004 action potential prolongation manifests itself as a prolongation of

www.landesbioscience.com Channels 87
hERG assays

LQTS) and an increased risk of Torsades de


Pointes. This was first demonstrated in the
late 1980s and 1990s for the histamine H1
receptor antagonist terfenadine (Fig. 4). In
1989, overdoses of terfenadine were shown
to prolong the QT interval.15 Terfenadine
use was linked to Torsades de Pointes in
1990,16 and terfenadine was subsequently
shown to inhibit the delayed rectifier
potassium current in isolated myocytes,
and hERG channels expressed in Xenopus
oocytes.17,18 The link between QT prolon-
gation and Torsades de Pointes is complex,
and not all drugs that prolong the QT
interval carry the same risk of arrhythmia.
However, QT interval prolongation is the
only known surrogate for an increased
risk for Torsades de Pointes.19 Based on
this increased risk, a significant number of
drugs that prolong the QT interval, ranging
from the antihistamines terfenadine and
Figure 1. Voltage-gated potassium channel family. (A) Diagram showing the predicted topology of an
individual potassium channel α and the minK-related β subunit (left), and the arrangement of the pore
astemizole to the antipsychotic droperidol
forming subunits in a potassium channel tetramer (right). (B) Family tree for voltage-gated potassium (Fig. 4), have been withdrawn from the
channels. market, while others have received black
box warning labels.20 Although the correla-
tion between block of hERG and Torsades de Pointes is not perfect,
the potential for putting patients at risk means that an ­interaction of
a compound with the hERG channel must be taken very seriously.
The enormous cost of drug development, coupled with the desire
to avoid late‑stage compound failures, motivates pharmaceutical
companies to test compounds for inhibition of hERG early on in the
lead optimization process.

Medium and High Throughput hERG Assays


The ideal hERG assay provides a linear measure of channel
activity under physiologically relevant conditions. The closest to this
ideal would be voltage clamp recordings in cardiac myocytes using a
cardiac action potential waveform as the voltage command. However,
such a study is extremely laborious and only amenable to the detailed
characterization of very few selected compounds. As previously
noted, it is advantageous to screen compounds for hERG activity
early on in the lead evaluation and optimization process. However,
this approach requires testing of hundreds and potentially thousands
of compounds within a single drug discovery program. Although
Figure 2. hERG channel gating. Conformational state diagram (top), exem- the development of automated electrophysiology ­technologies has
plary current traces (middle) and voltage protocol (bottom) describing hERG improved the throughput of electrophysiological methods, radioli-
channel gating. gand binding, fluorescent or ion flux assays may serve as efficient and
robust assays for examining the potential hERG liabilities of a large
the QT interval or the period between the beginning of the QRS number of compounds.
complex and end of the T wave (Fig. 3). Electrophysiology. Electrophysiology can provide detailed and
A particular form of inherited long QT syndrome (LQTS), LQTS2, quantitative information on the potency and mechanism of hERG
has been linked to loss‑of‑function mutations in hERG.11,13,14 While block by a test compound. One of the unique advantages of such
LQTS2 is typically not associated with any clinical symptoms, voltage clamp recordings is the ability to control membrane poten-
except occasional fainting in some patients, it greatly increases an tial. Since activation and inactivation of hERG is dependent on
­individual’s risk for Torsades de Pointes. membrane potential, voltage clamp recordings can differentiate
In analogy with the inherited syndrome, certain drugs that between compounds that preferentially interact with different states
inhibit hERG channels can cause LQTS (acquired or drug‑induced of the channel.

88 Channels 2008; Vol. 2 Issue 2


hERG assays

A major limitation of all automated


e­ lectrophysiology platforms is the high cost of the
instruments and consumables. Another concern
with regard to the automated platforms is the poten-
tial for results that are inconsistent with those from
conventional voltage clamp assays. Typically, hERG
potencies measured by automated and manual
electrophysiology are in good agreement; however,
some compounds are known to appear less potent
when tested by automated electrophysiology.23‑26
Discrepancies typically arise from hydrophobic
compounds being adsorbed to the surface of the plate
used to hold the compound or to the patch plate
itself. Glass‑coated compound plates can help to
minimize the nonspecific binding to the compound
plate but come at a significant cost. Multiple
compound additions can mitigate problems caused
by compounds sticking to the patch plate substrate
and are common practice.
Flux assays. An alternative to either manual or
automated electrophysiology is a functional assay that
measures ion flux across cell or vesicle membranes.
This assay offers higher throughput than the auto-
Figure 3. Electrocardiogram and cardiac action potential trace corresponding to one heart- mated voltage clamp protocols and takes advantage
beat. Adapted with permission from Macmillan Publishers Ltd: Nature Reviews: Drug Discovery of the ability of Rb+ to permeate through hERG
(Fermini & Fossa, 2003, Nature Rev Drug Disc 2:439–447), copyright 2003. channels.27 Typically, cells are loaded with Rb+
overnight and hERG‑dependent Rb+ efflux is initi-
Several higher throughput automated electrophysiological ated by an addition of high (50–60 mM) extracellular potassium
­instruments have been developed and typically use a planar substrate concentrations to depolarize the cell and open hERG channels. The
with holes, illustrated in Figure 5, that replaces the traditional patch amount of Rb+ efflux can be calculated by using 86Rb+ as a radioac-
pipette.21,22 Cells that seal to the substrate around a hole may be tive tracer or by flame atomic absorption spectrometry (FAAS).27,28
voltage clamped, if the section of plasma membrane covering the Such flux assays, which can operate in 96‑ and 384‑well formats,
hole can be ruptured manually, by applying negative pressure, or afford the testing of several thousand compounds per day. They
perforated using an antibiotic such as amphotericin, allowing elec- offer a robust measure of channel activity, but lack voltage control
trical access to the cell’s cytoplasm. Currently, the highest throughput and temporal resolution. In two reports, using either HEK‑293 or
devices available are the IonWorks HT and IonWorks Quattro. Both CHO‑K1 cells stably expressing recombinant hERG channels, IC50
instruments use a disposable 384‑well PatchPlate™ and a common values determined in a Rb+ flux assay were approximately 10‑fold
ground chamber and achieve increased throughput through synchro- higher than those determined by electrophysiology, but the rank
nous recording from 48 wells combined with simple and versatile order of compounds according to potency was similar for the two
software. Typical seal resistances are on the order of 100 mega‑Ohms assay types.29,30
for IonWorks HT and 50 mega‑Ohms for IonWorks Quattro. Whole Fluorescence‑based assays. The development of improved
cell access is obtained by perfusion with a membrane‑perforating ­fluorescent dyes and plate readers has provided another approach to
agent, typically amphotericin B. The IonWorks Quattro features a high throughput screening of ion channel activities. Fluorescent dyes
similar hardware and software design but uses an approach called which are sensitive to changes in membrane potential have proved
population patch clamp (PPC). PPC involves a modified PatchPlate especially useful for studying the pharmacology of many ion channel
with 64 holes per well and is especially advantageous when ­expression types.31,32 However, studying hERG by this approach ­presents
levels are not homogeneous from cell to cell. However, recording in a challenge since this channel does not typically control a cell’s
PPC mode requires a minimum success rate of sealing to holes and a resting membrane potential. It has been possible, however, to select
sufficiently high seal resistance to avoid short circuit currents through HEK‑293 and CHO‑K1 cell lines stably expressing recombinant
holes not occluded by a cell. hERG channels that give rise to a depolarization signal in response to
Other automated electrophysiology instruments, such as the addition of extracellular potassium (50–60 mM).29,30 At least in the
PatchXpress (Molecular Devices), the QPatch (Sophion Biosciences) HEK‑293 background, this signal is the sum of hERG‑dependent
and the Patchliner (Nanion Technologies GmbH) also use planar and hERG‑independent components; a complication that prob-
substrates, but record asynchronously from individual wells. ably contributes to the low signal‑to‑noise ratio observed in this
Currently, 16‑well or 48‑well formats are available. High resistance assay. In both cell lines, IC50 values were significantly right‑shifted
seals and flexible voltage protocols result in recordings that rival compared to those determined by electrophysiology, regardless of
traditional manual electrophysiology in quality. whether DiBAC4(3) (Invitrogen, Carlsbad, CA), FMP or Blue

www.landesbioscience.com Channels 89
hERG assays

Figure 5. Generalized scheme depicting the recording configurations used in


conventional (manual) and automated electrophysiology recordings.

Figure 4. Structures of selected compounds that bind to the hERG channel. and are relatively inexpensive, which is why they are commonly
used in most large pharmaceutical companies. A number of
Membrane Potential Indicator Dye (both from Molecular Devices, well‑characterized radioligands for hERG have been described,
Sunnyvale, CA) was used to monitor membrane potential. The including [3H]‑dofetilide,34 [35S]‑MK‑499,35 [3H]‑astemizole,36 and
lack of sensitivity of these membrane potential assays resulted in [125I]‑BeKm‑1.37 In general, binding assays using [3H]‑dofetilide,
a high incidence of “false negatives”. Moreover, the rank order of [35S]‑MK‑499 and [3H]‑astemizole (Fig. 4) are very robust and
compounds by IC50 differed between the membrane potential assay reproducible and IC50 values typically correlate well with those
and ­electrophysiological determinations. determined in electrophysiology.30,36,38,39 These three non‑peptide
To avoid some of the problems arising from non‑hERG ­potassium radioligands, in analogy with most small molecule hERG blockers,
currents, it may be possible to take advantage of known activators, such are thought to bind in the large inner cavity of the hERG channel
as PD‑307243,33 to open hERG channels under physiological condi- and require channel opening to access their binding sites. In contrast,
tions which will yield a hERG‑dependent hyperpolarization. One the scorpion venom peptide BeKm‑1 preferentially blocks channels
concern associated with this assay format is the potential ­interference in the closed state and binds to the extracellular S5‑pore linker of
of the channel activator with binding of the test compound. the channel.40,41 The binding site for [125I]‑BeKm‑1 may be allos-
Yet another approach to hERG fluorescence‑based assays involves terically coupled to the inner cavity, since E‑4031, which is believed
dyes, such as FluxOR™ (Invitrogen, Carlsbad, CA), that are sensi- to bind to the inner cavity, inhibits binding of [125I]‑BeKm‑1 to
tive to intracellular thallium concentrations, since thallium is known ­recombinant hERG channels expressed in HEK293 cells.37
to permeate through hERG channels. Such an assay provides a more Binding assays do not provide detailed information regarding the
direct measurement of hERG activity and may be comparable to Rb+ nature of the interaction of the test compound with hERG, such
flux assays, but with the benefit of being amenable to a 1536‑well as the ability to block or activate the channel or a preference for a
screening format. particular state of the channel. Despite this limitation and the fact
In all fluorescent assays there is the potential for interactions that [35S]‑MK‑499 itself binds with a Kd of <  1 nM but blocks
between the test compound and the dye that result in quenching of hERG with an IC50 of approximately 30 nM, agreement between
the fluorescent signal, and this may limit the range of compound compound potencies in [35S]‑MK‑499 binding and voltage clamp
concentrations that can be tested reliably. assays is remarkably high (reviewed in ref. 38, personal observation).
Binding assays. Radioligand binding assays have been used Radioligand binding assays using isolated membranes differ from
extensively to screen for interaction with the hERG channel. These whole cell functional assays in that they are amenable to a range of
are non‑functional assays, usually performed using isolated cell assay conditions that may impact on the ability of test compounds to
membranes, and they rely on competition or allosteric coupling bind.38 These include temperature, osmolarity, relative concentration
between the binding sites for the test compound and for the radioli- of organic solvent, and K+ concentration. In addition, binding assays
gand. Although they do not provide a direct measure of IKr blockade, typically employ time periods of drug exposure significantly longer
such binding assays can test 50,000 to 100,000 compounds per day than most functional assays, which may allow for a more accurate

90 Channels 2008; Vol. 2 Issue 2


hERG assays

measurements is between the use of conscious animals implanted


with telemetry devices and anesthetized animals.51 Advantages of
the anesthetized animals include greater reproducibility of the data
and the potential for electrical pacing to study effects of heart rate.
Disadvantages include the preclusion of oral dosing and the potential
for interference of the anesthetic with normal cardiac function or
with metabolism of the test compound.
The effects of a test substance on QT interval can be obscured
by changes in heart rate since the QT interval is inversely correlated
with heart rate.52 Therefore, it is common to report QT intervals
corrected for the preceding RR interval (QTc). A number of correc-
tion methods exists that attempt to normalize QT intervals to an RR
interval of 1000 ms, corresponding to a heart rate of 60 beats per
minute.52‑55
While ECG measurements directly evaluate the effect of a
particular test compound on the QT interval, they may not be the
best choice to evaluate the potential off‑target activity of a class of
compounds on hERG channels since other factors may affect the
QT interval and either obscure an effect on IKr or complicate the
Figure 6. Homology model of hERG generated from the KcsA crystal structure
with MK-499 docked to its putative binding site.
interpretation of the results.56

In Silico Modeling Predictions


determination of IC50 values for very potent compounds that require Compounds representing a wide range of chemical structures
a significant period of time to equilibrate. (Fig.  4) have been shown to block hERG. Alanine‑scanning muta-
genesis studies on the pore‑lining S6 segment and parts of the pore
Evaluation of Cardiac Risk
helix identified a number of residues involved in conferring high
Measurements of action potential duration. As an alternative affinity for MK‑499,57 including threonine and valine (T623 and
to assays that utilize recombinant hERG channels expressed in V625) residues in the pore and glycine, tyrosine and phenylalanine
mammalian cell lines, it is possible to examine native hERG chan- residues in S6 (G648, Y652, F656). The two aromatic residues in S6,
nels in myocardial cells from several species. The most common Y652 and F656, also contribute to the block of hERG by terfenadine,
tissue sources are isolated Purkinje fibers from guinea pig, rabbit or cisapride, dofetilide and quinidine.57‑59 These residues are conserved
dog.42‑45 The contribution of hERG to repolarization and action in ERG and EAG channels, but are not found in other voltage‑gated
potential prolongation in these tissues is similar to the human heart, potassium channels. Modeling studies have suggested that F656 may
in contrast to the rat heart, for which hERG does not contribute be involved in π‑stacking interactions with aromatic rings in hERG
significantly to repolarization. ligands, while Y652 may interact with the charged amine that is
Although measurements of action potential repolarization are very present in many compounds that bind to this channel.60
labor intensive and can only be used to study a limited number of A homology model based on the crystal structure of the ­bacterial
compounds, they can provide valuable data since they examine effects potassium channel KcsA suggested that the cavity lined by the
on native cardiac channels. This is potentially important since the membrane‑spanning sections of hERG may be larger than the cavi-
precise molecular composition of the channel that controls the IKr ties found in most ion channels.57 Figure 6 shows a model of hERG
current remains a matter of debate. Additionally, these assays can detect with MK‑499 docked using the FLOG (flexible ligands oriented
effects on a number of cardiac channels in addition to hERG.43 on grid) procedure. Together with the interactions afforded by the
In vivo ECG measurements. The most reliable measure of unique aromatic residues in S6 of hERG, the larger size of the cavity
cardiac safety is probably afforded by ECG recordings in conscious may explain the ability of hERG channels to accommodate binding
or anesthetized animals,43,46,47 and new drug applications require of a wide variety of structurally distinct compounds. While hERG
these data. ECG measurements are only practical for a very small homology models can help to explain the promiscuity relative to
number of select compounds. At the same time, such in vivo studies other potassium channels, they are less useful in predicting the ability
are the only methods of assessing cardiac safety that take into account of test compounds to block hERG.
the safety liabilities of the test compound and all its metabolites A different modeling approach involves quantitative
combined. They also allow a direct evaluation of other potential ­structure‑activity relationship (QSAR) modeling.60 This approach
complications, such as plasma protein binding, tissue distribution takes advantage of known hERG blockers that are used as a training
and poly‑pharmacology. set to generate a pharmacophore model, which can then be tested
The most commonly used species for these measurements is the against a test set. Several pharmacophore models have been devel-
dog.47 Guinea pigs may also be used but are technically more chal- oped and can be used to prioritize compounds for hERG testing.
lenging.48 If metabolite profiles are expected to differ between man Since these models are generally better at predicting lack of hERG
and dog, ECG measurements can also be made in primates.49,50 In activity, they can be used as a screening tool to reduce the cost of
addition to the choice of species, the most important choice for ECG hERG testing in the in vitro assays.

www.landesbioscience.com Channels 91
hERG assays

References
hERG Trafficking 1. Warmke JW, Ganetzky B. A family of potassium channel genes related to eag in Drosophila
and mammals. Proc Natl Acad Sci 1994; 91:3438‑42.
More recently, effects on hERG trafficking have been identified as 2. Gutman GA, Chandy KG, Grissmer S, Lazdunski M, McKinnon D, Pardo LA, Robertson GA,
a mechanism for drug‑induced LQTS.61 Drugs that reduce hERG/ Rudy B, Sanguinetti MC, Stuhmer W, Wang X. International Union of Pharmacology LIII.
IKr currents through interfering with hERG trafficking include Nomenclature and molecular relationships of voltage‑gated potassium channels. Pharmacol
Rev 2005; 57:473‑508.
arsenic trioxide, pentamidine and the cardiac glycosides digoxin, and 3. London B, Trudeau MC, Newton KP, Beyer AK, Copeland NG, Gilbert DJ, Jenkins NA,
ouabain.62‑64 A number of other drugs block hERG directly and Satler CA, Robertson GA. Two isoforms of the mouse ether‑a‑go‑go‑related gene coassemble
to form channels with properties similar to the rapidly activating component of the cardiac
interfere with trafficking. These include the antidepressant fluoxetine delayed rectifier K+ current. Circul Res 1997; 81:870‑8.
and the cholesterol‑lowering agent probucol.65,66 The mechanisms 4. Jones EM, Roti Roti EC, Wang J, Delfosse SA, Robertson GA. Cardiac IKr channels mini-
by which drugs interfere with hERG trafficking are largely unknown. mally comprise hERG 1a and 1b subunits. J Biol Chem 2004; 279:44690‑4.
5. Farrelly AM, Ro S, Callaghan BP, Khoyi MA, Fleming N, Horowitz B, Sanders KM, Keef KD.
In some cases, interactions with chaperones such as Hsp90 may Expression and function of KCNH2 (HERG) in the human jejunum. Am J Physiol
prevent proper folding of hERG;67 whereas in other cases drugs Gastrointest Liver Physiol 2003; 284:883‑95.
appear to trap hERG in the endoplasmic reticulum.64 Most in vitro 6. Shi W, Wymore RS, Wang HS, Pan Z, Cohen IS, McKinnon D, Dixon JE. Identification of
two nervous system‑specific members of the erg potassium channel gene family. J Neurosci
and in vivo assays currently used to detect hERG inhibition and 1997; 17:9423‑32.
QT prolongation focus on acute effects and are not likely to detect 7. McDonald TV, Yu Z, Ming Z, Palma E, Meyers MB, Wang KW, Goldstein SA, Fishman GI.
drug effects on channel trafficking. Electrophysiology, Rb+ flux and A minK‑HERG complex regulates the cardiac potassium current IKr. Nature 1997;
388:289‑92.
whole cell binding assays yield signals that are proportional to surface 8. Abbott GW, Sesti F, Splawski I, Buck ME, Lehmann MH, Timothy KW, Keating MT,
channel expression and could therefore be adapted to detect drug‑in- Goldstein SA. MiRP1 forms IKr potassium channels with HERG and is associated with
cardiac arrhythmia. Cell 1999; 97:175‑87.
duced changes in hERG trafficking. In addition, an antibody‑based 9. Sanguinetti MC, Jiang C, Curran ME, Keating MT. A mechanistic link between an inher-
chemiluminescent assay has been developed to specifically monitor ited and an acquired cardiac arrhythmia: HERG encodes the IKr potassium channel. Cell
effects on hERG cell surface expression.68 1995; 81:299‑307.
10. Keating MT, Sanguinetti MC. Molecular and cellular mechanisms of cardiac arrhythmias.
hERG Activators Cell 2001; 104:569‑80.
11. Curran ME, Splawski I, Timothy KW, Vincent GM, Green ED, Keating MT. A molecu-
lar basis for cardiac arrhythmia: HERG mutations cause long QT syndrome. Cell 1995;
Recently, several hERG activators have been identified.69‑71 As 80:795‑803.
expected these compounds shorten the ventricular action ­potential 12. Priori SG, Cantu F, Schwartz PJ. The long QT syndrome: new diagnostic and therapeutic
approach in the era of molecular biology. Schweizerische medizinische Wochenschrift 1996;
duration and the QT interval. Several forms of familial short QT 126:1727‑31.
syndrome have been identified, one of which has been linked 13. Huang FD, Chen J, Lin M, Keating MT, Sanguinetti MC. Long‑QT syndrome‑associ-
to mutations in hERG.72 Familial short QT syndrome is associ- ated missense mutations in the pore helix of the HERG potassium channel. Circ 2001;
104:1071‑5.
ated with atrial and ventricular arrhythmias and sudden death, 14. Sanguinetti MC, Tristani‑Firouzi M. hERG potassium channels and cardiac arrhythmia.
suggesting that activation of hERG carries similar safety liabilities Nature 2006; 440:463‑9.
as hERG blockade. 15. Davies AJ, Harindra V, McEwan A, Ghose RR. Cardiotoxic effect with convulsions in
terfenadine overdose. BMJ (Clinical research ed) 1989; 298:325.
Summary 16. Monahan BP, Ferguson CL, Killeavy ES, Lloyd BK, Troy J, Cantilena LR Jr. Torsades de
pointes occurring in association with terfenadine use. JAMA 1990; 264:2788‑90.
17. Woosley RL, Chen Y, Freiman JP, Gillis RA. Mechanism of the cardiotoxic actions of ter-
In order to reduce the risk of a drug candidate failing in preclinical fenadine. JAMA 1993; 269:1532‑6.
safety studies because of blockade of hERG channels and associated 18. Roy M, Dumaine R, Brown AM. HERG, a primary human ventricular target of the nonse-
QT prolongation, it is important to screen compounds for activity dating antihistamine terfenadine. Circ 1996; 94:817‑23.
19. Redfern WS, Carlsson L, Davis AS, Lynch WG, MacKenzie I, Palethorpe S, Siegl PK,
on hERG channels early in the lead optimization process. However, Strang I, Sullivan AT, Wallis R, Camm AJ, Hammond TG. Relationships between preclini-
the earlier in the process hERG screening is to occur, the higher the cal cardiac electrophysiology, clinical QT interval prolongation and torsade de pointes for
a broad range of drugs: evidence for a provisional safety margin in drug development.
required assay throughput. Although binding assays do not measure Cardiovascular Res 2003; 58:32‑45.
hERG function, they offer a rapid, robust and inexpensive measure 20. Fermini B, Fossa AA. The impact of drug‑induced QT interval prolongation on drug dis-
of the ability of compounds to interact with the hERG channel. covery and development. Nature Rev 2003; 2:439‑47.
21. Priest BT, Swensen AM, McManus OB. Automated electrophysiology in drug discovery.
Combined with electrophysiology on a few benchmark compounds Curr Pharm Des 2007; 13:2325‑37.
to characterize the nature of the compound‑channel interaction, 22. Dunlop J, Bowlby M, Peri R, Vasilyev D, Arias R. High‑throughput electrophysiol-
ogy: an emerging paradigm for ion‑channel screening and physiology. Nature Rev 2008;
binding assays can be used as an efficient means to develop the 7:358‑68.
­structure‑function relationship of hERG interactions within a 23. Kiss L, Bennett PB, Uebele VN, Koblan KS, Kane SA, Neagle B, Schroeder K. High
structural class of test compounds. This strategy should facilitate throughput ion‑channel pharmacology: planar‑array‑based voltage clamp. Assay Drug Dev
Technol 2003; 1:127‑35.
the development of new small molecule drugs with improved 24. Tao H, Santa Ana D, Guia A, Huang M, Ligutti J, Walker G, Sithiphong K, Chan F,
­cardiovascular safety profiles. Guoliang T, Zozulya Z, Saya S, Phimmachack R, Sie C, Yuan J, Wu L, Xu J, Ghetti A.
Automated tight seal electrophysiology for assessing the potential hERG liability of phar-
Acknowledgements maceutical compounds. Assay Drug Dev Technol 2004; 2:497‑506.
The authors thank Jill Williams and Chris Culberson for 25. Sorota S, Zhang XS, Margulis M, Tucker K, Priestley T. Characterization of a hERG screen
using the IonWorks HT: comparison to a hERG rubidium efflux screen. Assay Drug Dev
­assistance in the preparation of figures. Technol 2005; 3:47‑57.
26. Dubin AE, Nasser N, Rohrbacher J, Hermans AN, Marrannes R, Grantham C, Van Rossem K,
Cik M, Chaplan SR, Gallacher D, Xu J, Guia A, Byrne NG, Mathes C. Identifying modula-
tors of hERG channel activity using the PatchXpress planar patch clamp. J Biomol Screen
2005; 10:168‑81.
27. Weir SW, Weston AH. The effects of BRL 34915 and nicorandil on electrical and mechani-
cal activity and on 86Rb efflux in rat blood vessels. Br J Pharmacol 1986; 88:121‑8.

92 Channels 2008; Vol. 2 Issue 2


hERG assays

28. Terstappen GC. Functional analysis of native and recombinant ion channels using a 53. Franz MR. Time for yet another QT correction algorithm? Bazett and beyond. J Am Coll
high‑capacity nonradioactive rubidium efflux assay. Anal Biochem 1999; 272:149‑55. Cardiol 1994; 23:1554‑6.
29. Tang W, Kang J, Wu X, Rampe D, Wang L, Shen H, Li Z, Dunnington D, Garyantes T. 54. Molnar J, Weiss J, Zhang F, Rosenthal JE. Evaluation of five QT correction formulas using a
Development and evaluation of high throughput functional assay methods for HERG software‑assisted method of continuous QT measurement from 24‑hour Holter recordings.
potassium channel. J Biomol Screen 2001; 6:325‑31. Am J Cardiol 1996; 78:920‑6.
30. Murphy SM, Palmer M, Poole MF, Padegimas L, Hunady K, Danzig J, Gill S, Gill R, Ting A, 55. Luo S, Michler K, Johnston P, Macfarlane PW. A comparison of commonly used QT correc-
Sherf B, Brunden K, Stricker Krongrad A. Evaluation of functional and binding assays in tion formulae: the effect of heart rate on the QTc of normal ECGs. J Electrocardiol 2004;
cells expressing either recombinant or endogenous hERG channel. J Pharmacol Toxicol 37:81‑90.
Meth 2006; 54:42‑55. 56. Schneider J, Hauser R, Andreas JO, Linz K, Jahnel U. Differential effects of human
31. Gonzalez JE, Maher MP. Cellular fluorescent indicators and voltage/ion probe reader ether‑a‑go‑go‑related gene (HERG) blocking agents on QT duration variability in conscious
(VIPR) tools for ion channel and receptor drug discovery. Receptors Channels 2002; dogs. Eur J Pharmacol 2005; 512:53‑60.
8:283‑95. 57. Mitcheson JS, Chen J, Lin M, Culberson C, Sanguinetti MC. A structural basis for
32. Felix JP, Williams BS, Priest BT, Brochu RM, Dick IE, Warren VA, Yan L, Slaughter RS, drug‑induced long QT syndrome. Proc Natl Acad Sci USA 2000; 97:12329‑33.
Kaczorowski GJ, Smith MM, Garcia ML. Functional assay of voltage‑gated sodium chan- 58. Lees‑Miller JP, Duan Y, Teng GQ, Duff HJ. Molecular determinant of high‑affinity
nels using membrane potential‑sensitive dyes. Assay Drug Dev Technol 2004; 2:260‑8. dofetilide binding to HERG1 expressed in Xenopus oocytes: involvement of S6 sites. Mol
33. Gordon E, Lozinskaya IM, Lin Z, Semus SF, Blaney FE, Willette RN, Xu X. 2‑[2‑(3,4‑di- Pharm 2000; 57:367‑74.
chloro‑phenyl)‑2,3‑dihydro‑1H‑isoindol‑5‑ylamino]‑nicotinic acid (PD‑307243) causes 59. Kamiya K, Niwa R, Mitcheson JS, Sanguinetti MC. Molecular determinants of HERG
instantaneous current through human ether‑a‑go‑go‑related gene potassium channels. Mol
channel block. Mol Pharm 2006; 69:1709‑16.
Pharmacol 2008; 73:639‑51.
60. Aronov AM. Predictive in silico modeling for hERG channel blockers. Drug Discovery
34. Finlayson K, Turnbull L, January CT, Sharkey J, Kelly JS. [3H]dofetilide binding to HERG
Today 2005; 10:149‑55.
transfected membranes: a potential high throughput preclinical screen. Eur J Pharmacol
2001; 430:147‑8. 61. Dennis A, Wang L, Wan X, Ficker E. hERG channel trafficking: novel targets in drug‑in-
duced long QT syndrome. Biochem Soc Trans 2007; 35:1060‑3.
35. Raab CE, Butcher JW, Connolly TM, Karczewski J, Yu NX, Staskiewicz SJ, Liverton N,
Dean DC, Melillo DG. Synthesis of the first sulfur‑35‑labeled hERG radioligand. Bioorg 62. Ficker E, Kuryshev YA, Dennis AT, Obejero‑Paz C, Wang L, Hawryluk P, Wible BA, Brown AM.
Med Chem Lett 2006; 16:1692‑5. Mechanisms of arsenic‑induced prolongation of cardiac repolarization. Mol Pharm 2004;
36. Chiu PJ, Marcoe KF, Bounds SE, Lin CH, Feng JJ, Lin A, Cheng FC, Crumb WJ, Mitchell R. 66:33‑44.
Validation of a [3H]astemizole binding assay in HEK293 cells expressing HERG K+ chan- 63. Kuryshev YA, Ficker E, Wang L, Hawryluk P, Dennis AT, Wible BA, Brown AM, Kang J,
nels. J Pharmacol Sci 2004; 95:311‑9. Chen XL, Sawamura K, Reynolds W, Rampe D. Pentamidine‑induced long QT syndrome
37. Angelo K, Korolkova YV, Grunnet M, Grishin EV, Pluzhnikov KA, Klaerke DA, Knaus HG, and block of hERG trafficking. J Pharm Exp Therap 2005; 312:316‑23.
Moller M, Olesen SP. A radiolabeled peptide ligand of the hERG channel, [125I]‑BeKm‑1. 64. Wang L, Wible BA, Wan X, Ficker E. Cardiac glycosides as novel inhibitors of human
Pflugers Arch 2003; 447:55‑63. ether‑a‑go‑go‑related gene channel trafficking. J Pharm Exp Therap 2007; 320:525‑34.
38. Wang J, Della Penna K, Wang H, Karczewski J, Connolly TM, Koblan KS, Bennett PB, 65. Rajamani S, Eckhardt LL, Valdivia CR, Klemens CA, Gillman BM, Anderson CL, Holzem KM,
Salata JJ. Functional and pharmacological properties of canine ERG potassium channels. Delisle BP, Anson BD, Makielski JC, January CT. Drug‑induced long QT syndrome: hERG
Am J Physiol 2003; 284:256‑67. K+ channel block and disruption of protein trafficking by fluoxetine and norfluoxetine. Br
39. Diaz GJ, Daniell K, Leitza ST, Martin RL, Su Z, McDermott JS, Cox BF, Gintant GA. The J Pharm 2006; 149:481‑9.
[3H]dofetilide binding assay is a predictive screening tool for hERG blockade and proar- 66. Guo J, Massaeli H, Li W, Xu J, Luo T, Shaw J, Kirshenbaum LA, Zhang S. Identification of
rhythmia: Comparison of intact cell and membrane preparations and effects of altering [K+] IKr and its trafficking disruption induced by probucol in cultured neonatal rat cardiomyo-
o. J Pharmacol Toxicol Meth 2004; 50:187‑99. cytes. The Journal of pharmacology and experimental therapeutics 2007; 321:911‑20.
40. Milnes JT, Dempsey CE, Ridley JM, Crociani O, Arcangeli A, Hancox JC, Witchel HJ. 67. Ficker E, Dennis AT, Wang L, Brown AM. Role of the cytosolic chaperones Hsp70 and
Preferential closed channel blockade of HERG potassium currents by chemically synthesised Hsp90 in maturation of the cardiac potassium channel HERG. Circ Res 2003; 92:87‑100.
BeKm‑1 scorpion toxin. FEBS Letters 2003; 547:20‑6. 68. Wible BA, Hawryluk P, Ficker E, Kuryshev YA, Kirsch G, Brown AM. HERG‑Lite: a novel
41. Korolkova YV, Bocharov EV, Angelo K, Maslennikov IV, Grinenko OV, Lipkin AV, comprehensive high‑throughput screen for drug‑induced hERG risk. J Pharmacol Toxicol
Nosyreva ED, Pluzhnikov KA, Olesen SP, Arseniev AS, Grishin EV. New binding site on Meth 2005; 52:136‑45.
common molecular scaffold provides HERG channel specificity of scorpion toxin BeKm‑1.
69. Kang J, Chen XL, Wang H, Ji J, Cheng H, Incardona J, Reynolds W, Viviani F, Tabart M,
J Biol Chem 2002; 277:43104‑9.
Rampe D. Discovery of a small molecule activator of the human ether‑a‑go‑go‑related gene
42. Davie C, Pierre Valentin J, Pollard C, Standen N, Mitcheson J, Alexander P, Thong B. (HERG) cardiac K+ channel. Mol Pharm 2005; 67:827‑36.
Comparative pharmacology of guinea pig cardiac myocyte and cloned hERG (IKr) channel.
70. Zhou J, Augelli‑Szafran CE, Bradley JA, Chen X, Koci BJ, Volberg WA, Sun Z, Cordes JS.
J Cardiovasc Electrophysiol 2004; 15:1302‑9.
Novel potent human ether‑a‑go‑go‑related gene (hERG) potassium channel enhancers and
43. Picard S, Lacroix P. QT interval prolongation and cardiac risk assessment for novel drugs.
their in vitro antiarrhythmic activity. Mol Pharm 2005; 68:876‑84.
Curr Op Investig Drugs 2003; 4:303‑8.
71. Hansen RS, Diness TG, Christ T, Demnitz J, Ravens U, Olesen SP, Grunnet M. Activation
44. Brown AM. HERG block, QT liability and sudden cardiac death. Novartis Foundation
of human ether‑a‑go‑go‑related gene potassium channels by the diphenylurea 1,3‑bis‑(2‑hy-
Symposium 2005; 266:118‑31.
droxy‑5‑trifluoromethyl‑phenyl)‑urea (NS1643). Mol Pharm 2006; 69:266‑77.
45. Lu HR, Vlaminckx E, Van de Water A, Rohrbacher J, Hermans A, Gallacher DJ. In‑vitro
experimental models for the risk assessment of antibiotic‑induced QT prolongation. Eur J 72. Gussak I, Brugada P, Brugada J, Wright RS, Kopecky SL, Chaitman BR, Bjerregaard P.
Pharmacol 2006; 553:229‑39. Idiopathic short QT interval: a new clinical syndrome? Cardiology 2000; 94:99‑102.
46. Mizuno H, Adachi H, Kimura J, Sawa Y, Kakiki M, Lansdell K, Saito M, Kerns WD.
Cardiovascular assessment of ER‑118585, a selective phosphodiesterase 5 inhibitor. Biol
Pharm Bull 2003; 26:1661‑7.
47. Chen X, Cass JD, Bradley JA, Dahm CM, Sun Z, Kadyszewski E, Engwall MJ, Zhou J.
QT prolongation and proarrhythmia by moxifloxacin: concordance of preclinical models in
relation to clinical outcome. Br J Pharmacol 2005; 146:792‑9.
48. Testai L, Calderone V, Salvadori A, Breschi MC, Nieri P, Martinotti E. QT prolongation in
anaesthetized guinea‑pigs: an experimental approach for preliminary screening of torsado-
genicity of drugs and drug candidates. J Appl Toxicol 2004; 24:217‑22.
49. Ando K, Hombo T, Kanno A, Ikeda H, Imaizumi M, Shimizu N, Sakamoto K, Kitani S,
Yamamoto Y, Hizume S, Nakai K, Kitayama T, Yamamoto K. QT PRODACT: in vivo QT
assay with a conscious monkey for assessment of the potential for drug‑induced QT interval
prolongation. J Pharmacol Sci 2005; 99:487‑500.
50. Chaves AA, Keller WJ, O’Sullivan S, Williams MA, Fitzgerald LE, McPherson HE,
Goykhman D, Ward PD, Hoe CM, Mixson L, Briscoe RJ. Cardiovascular monkey
telemetry: sensitivity to detect QT interval prolongation. J Pharmacol Toxicol Meth 2006;
54:150‑8.
51. Nolan ER, Feng MR, Koup JR, Liu J, Turluck D, Zhang Y, Paulissen JB, Olivier NB, Miller T,
Bailie MB. A novel predictive pharmacokinetic/pharmacodynamic model of repolariza-
tion prolongation derived from the effects of terfenadine, cisapride and E‑4031 in the
conscious chronic av node—ablated, His bundle‑paced dog. J Pharmacol Toxicol Meth
2006; 53:1‑10.
52. Ahnve S. Correction of the QT interval for heart rate: review of different formulas and the
use of Bazett’s formula in myocardial infarction. American heart journal 1985; 109:568‑74.

www.landesbioscience.com Channels 93

You might also like