You are on page 1of 16

REVIEWS

Innovative approaches to
anti-arrhythmic drug therapy
Stanley Nattel* and Leif Carlsson‡
Abstract | Normal cardiac function requires an appropriate and regular beating rate
(cardiac rhythm). When the heart rhythm is too fast or too slow, cardiac function can be
impaired, with derangements that vary from mild symptoms to life-threatening
complications. Irregularities, particularly those involving excessively fast or slow rates,
constitute cardiac ‘arrhythmias’. In the past, drug treatment of cardiac arrhythmias has
proven difficult, both because of inadequate effectiveness and a risk of serious
complications. However, a variety of recent advances have opened up exciting possibilities
for the development of novel and superior approaches to arrhythmia therapy. This article
will review recent progress and future prospects for treating two particularly important
cardiac arrhythmias: atrial fibrillation and ventricular fibrillation.

The normal heart beats approximately 100,000 times a of problems with side effects, particularly a paradoxi-
day, with beating rates finely tuned to the body’s needs. cal capacity to create more serious rhythm disorders
When the beating rate is inappropriately rapid or slow than the ones being treated, a phenomenon called ‘pro-
because of abnormalities in cardiac rhythm referred to as arrhythmia’6. Pro-arrhythmia is largely due to powerful
‘cardiac arrhythmias’, cardiac function can be impaired, effects of the drugs on ion channels, often in cardiac
producing derangements varying from mild symptoms regions other than the arrhythmic zone being treated,
to severe life-threatening complications. which create unexpected derangements in the presence
Two particularly important cardiac arrhythmias of a vulnerable substrate and induce rhythm instability.
with major clinical implications are atrial fibrillation Because of the potentially devastating effect of
(AF) and ventricular fibrillation (VF) (FIG. 1). AF is the arrhythmias, even low-probability events that occur
most common arrhythmia in the population1,2, and the in predisposed individuals at crucial times can be very
complex mechanisms leading to abnormal firing in AF dangerous, and pro-arrhythmic potential is a serious
are being rapidly elucidated3. Its age-related occurrence limitation of presently used anti-arrhythmic agents.
rate is growing alarmingly with the ageing of the popula- There is therefore a major unmet need for drugs that
tion, and it is estimated that if present trends continue will control arrhythmias more safely and effectively. New
more than 15 million Americans will be affected by AF developments in our understanding of the molecular
by 20504. It is also a common identifiable cause of stroke and biophysical factors controlling cardiac rhythm — as
(the single most common factor in stroke of the elderly) well as rapidly advancing approaches to high-through-
*Department of Medicine and is associated with particularly severe strokes5. VF is put screening (HTS) of molecular libraries, intelligent
and Research Center,
Montreal Heart Institute
the most common cause of sudden cardiac death, and is chemical modification of substance structure to achieve
and Université de Montréal, estimated to kill about 600,000 individuals per year in desired profiles of action, and cell and gene therapy
5000 Belanger Street, Europe and the United States combined. — are creating exciting opportunities in anti-arrhythmic
Montreal, Quebec, Anti-arrhythmic drugs have been used for more than therapeutic innovation. This paper reviews emerging
Canada H1T 1C8.

a hundred years, ever since quinidine was identified as findings that promise the development of new types of
AstraZeneca Research
& Development, the active ingredient in Cinchona bark and subsequently safer and more effective anti-arrhythmic compounds,
Integrative Pharmacology, achieved widespread use in the 1920s6. Currently avail- addressing in particular drugs that target the two very
Pepparedsleden 1, able anti-arrhythmic drug options include less than a important arrhythmias, AF and VF, illustrated in FIG. 1.
S-431 83 Mölndal, Sweden. dozen rather old agents, the majority of which were spe-
Correspondence to S.N.:
e-mail:
cifically designed to target ion channels, with the most Key basic concepts
stanley.nattel@icm-mhi.org recent launch more than 5 years ago. Anti-arrhythmic The cellular basis of cardiac electrical activity is the car-
doi:10.1038/nrd2112 drug use has decreased over the past 15 years because diac action potential (AP), which is based on ion fluxes

1034 | DECEMBER 2006 | VOLUME 5 www.nature.com/reviews/drugdisc


© 2006 Nature Publishing Group
REVIEWS

through specific membrane structures, particularly ion intracellular potential to a positive value (causing a phase
channels (BOX 1). The ‘firing’ or depolarization of cardiac of the AP called ‘phase 0’), it goes through a series of reg-
cells and closely associated cardiac electrical conduction ulated repolarizing steps (AP phases 1 and 3), separated
depends on the movement of positive ions into the cell by a relatively flat phase of the AP (phase 2), to get back
(Na+ for atrial and ventricular tissue, Ca2+ for nodal to its resting potential (BOX 1). Cardiac cells are gener-
structures such as the sino-atrial node (SAN) and the ally inexcitable once they have fired, and the time taken
atrioventricular (AV) node). Once a cardiac cell is fired from initial depolarization to repolarization (called AP
by being depolarized from its normally negative resting duration (APD)) imposes a limit (called the refractory
period (RP)) on how soon a cell can be re-excited. APD
is controlled by the rate of repolarization, which depends
a Normal heart
on the balance between inward movement of Na+ and
SAN LA AV node Ca2+ that tends to keep the cell depolarized and outward
0 mV SAN RA movement of K+ through a series of highly specialized
mV

mV
0 mV channels with typical time-dependent opening and
–50 –60 closing properties.
The underlying cause of cardiac arrhythmias is
AV node
Atrium
abnormal impulse formation or impulse propagation
Atrium Ventricle (FIG. 2) resulting from defective (overactive or underac-
30 mV 30 mV tive) function of cardiac ion channels and exchangers.
mV

mV Both causes can involve abnormal ion channel func-


RV LV tion and defective intracellular ion handling that can
–70 –80
Ventricle
accelerate or retard diastolic depolarization, produce
inward currents that generate after-depolarizations or
1 second
create conditions for re-entry arrhythmias. Enhanced
automaticity (FIG. 2A) refers to a regional exaggeration
b Atrial fibrillation c Ventricular fibrillation of normal cellular capacity to fire spontaneously, and
causes cardiac rhythm to be dictated by a site other
than the normal SAN pacemaker. After-depolarizations
are abnormal depolarizations falling during AP phases
2 or 3 (early after-depolarizations (EADs)), or phase
4 (delayed after-depolarizations (DADs)) (FIG. 2B,C).
EADs are caused by factors that impair repolariza-
tion, allowing for re-activation of inward currents that
depolarize the cell prematurely. DADs are caused by
abnormal Ca2+ leakage from subcellular stores during
the resting phase (phase 4), which causes enhanced
movement of Na+ into the cell in exchange for the excess
Ca2+. Re-entry (FIG. 3) is akin to an electrical short-cir-
cuit that depends on the balance between refractory
and conduction properties. Most ion channels deter-
Figure 1 | Electrical functioning of the heart. a | Normal heart. The heart has its mining cardiac electrical activity have been molecularly
own pacemaker: the sino-atrial node (SAN), which produces regular electrical impulses defined and the crucial ion currents involved in vari-
at an appropriate rate (normally 60–100 per min, but faster in situations of stress). The ous forms of cardiac arrhythmias are largely known7,
impulse spreads across the right atrium (RA) and left atrium (LA) and towards the atrio- as are the mechanisms by which presently used anti-
ventricular node (AV node), where its velocity is reduced before passing on to the left
arrhythmic agents work 8 (FIG. 4). If novel candidate
(LV) and right (RV) ventricles, in which contraction and therefore ventricular pumping
is triggered. Representative action potentials are shown from SAN and atria (left) and
currents for anti-arrhythmic action can be established,
from AV node and ventricles (right), respectively. b | Atrial fibrillation. The upper automated HTS methods can identify potent and selec-
cardiac chambers (atria) can develop sustained, very rapid (400–600 per min) and tive blockers from small-molecule libraries9. The key
irregular firing called atrial fibrillation (AF; for a detailed discussion, see REF. 1). These question that has to be answered in order to use this
waves of activity bombard the AV node, which filters the impulses, but they still reach powerful technology is: which ion channels are the
the ventricles at an inappropriately rapid rate (typically about 150 per min). Untreated, best candidates for new anti-arrhythmic drug devel-
the maintenance of such a rapid rate is likely to cause the heart to fail over time. In opment? In the discussion below, we present a number
addition, the atria do not contract effectively during AF, resulting in inadequate of promising new targets, including ion channels that
ventricular filling and a coagulation-favouring state, increasing the chances that a clot can be specifically targeted in the atria, ion channels
will dislodge and go to the brain (causing a stroke) or to other vital organs. c | Ventricular
that are specifically involved in triggering ventricular
fibrillation. Ventricular fibrillation (VF) resembles AF in producing rapid, irregular
waves that control ventricular activity. However, unlike AF, against which the ventricles
arrhythmias due to acute myocardial ischaemia, and
are protected by the filtering function of the AV node, VF directly fires the ventricles at ion channels that generate or modulate spontane-
400—600 per min. Such rapid activity is incompatible with effective cardiac pumping, ous activity. We also outline the lessons that can be
and VF is rapidly lethal if not arrested. The most common explanation of both VF and learned from inherited arrhythmia syndromes, and
AF is multiple simultaneous functional re-entry waves (for a presentation of the briefly discuss potential alternative strategies such as
mechanism of re-entry, see FIG. 3). gene therapy and ‘upstream therapy’ — the prevention

NATURE REVIEWS | DRUG DISCOVERY VOLUME 5 | DECEMBER 2006 | 1035


© 2006 Nature Publishing Group
REVIEWS

Box 1 | The action potential and ion currents during depolarization


The action potential (AP; red line), here depicted for a
typical atrial or ventricular cell, reflects the changing Ito↑
Early
electrical potential differences across cardiac cell repolarization Plateau (2)
membranes as a function of time, and is controlled by (1) ICaL↓
the flow of ions through ion channels (grey boxes with IKur↑
IKs↑
arrow up: outward currents; yellow boxes with arrow 0
down: inward currents). In the rested state (diastole, IKr↑
phase 4), ion pumps maintain a higher outside INa↓
concentration of Na+ and Ca2+ and a higher inside

mV
concentration of K+. The cell is impermeable to Na+ and Final
Ca2+, but is permeable to K+ through a channel Upstroke IKACh↑ repolarization (3)
+
(0)
designated IK1. As a result, positively charged K ions If↓
IK1↑
flow slowly out of the cell, keeping the interior
IKACh↑ Pacemaker
negative. When the membrane potential reaches a function
crucial threshold voltage, the cells ‘fire’ as positive ions IK1↑
(Na+ in the case of atrium and ventricle, Ca2+ in the APD
–80
case of the sino-atrial and atrioventricular nodes) Resting
NCX↓
(4) 0.2 seconds
enter the cell and rapidly bring the cell to a
‘depolarized’, positive potential (a process known as IK1
the AP upstroke or AP phase 0). The large current (INa) INa
through Na+ channels provides the electrical energy Ito
IKur
for rapid conduction in the mammalian heart. ICaL
2+
Following depolarization, Ca enters the cell through IKr
the L-type Ca2+-channel (ICaL), helping the cell to IKs
remain depolarized after initial depolarization and NCX
If
inducing mechanical contraction. During the ‘plateau’ IKACh
2+ +
phase of the AP (phase 2), inward Ca and outward K
currents are relatively balanced. To bring the cell back
to its resting state (‘repolarization’), an outward flow of K+ has to occur and is produced by a series of K+ currents known
by their time-dependent properties as transient-outward (Ito) and ultra-rapid (IKur), rapid (IKr), and slow (IKs) delayed-
rectifier currents. Rapidly activating K+ currents such as Ito and IKur are responsible for very rapid initial repolarization
(phase 1), whereas slower components (IKr and IKs) primarily determine terminal repolarization (phase 3), which brings
the cell back to its resting state. The time elapsed between AP upstroke and the return to the resting potential is
referred to as AP duration (APD; green bracket). Cells are unable to manifest a normal AP upstroke during the
depolarized phase of the AP, a period of relative inexcitability called the refractory period, corresponding roughly to the
APD. Some cells have a ‘pacemaker current’, If, allowing them to depolarize gradually during phase 4 to ‘threshold’,
causing repetitive spontaneous firing at a frequency characteristic of the tissue. The extra Na+ and Ca2+ which have
entered the cell during the AP are extruded (and the K+ which has left the cell is brought back in) by exchanging
intracellular Na+ for extracellular K+ (through the Na+,K+-pump) and Na+ for Ca2+ (by an exchanger called the Na+,Ca2+-
exchanger (NCX)). The acetylcholine-regulated K+ current IKACh is an inward rectifier current with time and voltage
dependent properties roughly similar to those of IK1. The timing of each current in relation to the AP is shown in the
schematic below.

of arrhythmogenic remodelling. A list of new anti- One interesting potential target is IKur, carried by
arrhythmic agents, along with molecular structures, Kv1.5 subunits (FIG. 4), which in humans has a significant
target, therapeutic indication, stage of development role in the atria12,13 but is absent in the ventricles13–15.
and manufacturer, is provided in TABLE 1. Its inhibition should delay atrial repolarization and RP
without affecting ventricular tissue (and therefore be
Atrial-selective drug targets free of the risk of ventricular pro-arrhythmia). Many
The most common mechanism of AF is complex atrial pharmaceutical companies have developed IKur block-
reentry1 (FIG. 3). An effective way to prevent or terminate ers16 and some compounds are approaching or have
re-entry is to increase the RP. The RP can be increased by entered clinical trials. One representative of this group is
drugs that inhibit K+-channels, making cells take longer AVE0118 (Sanofi-Aventis), which initially was believed
to repolarize to their rested state, and the drugs that to be a specific IKur blocker but later found to block Ito
have traditionally been used to prolong the RP (such as and IKACh as well. AVE0118 has shown excellent anti-
quinidine, sotalol and dofetilide) target IKr6. The problem arrhythmic efficacy in a goat model of persistent AF17.
with IKr as a target is that it is crucial for repolarization Another example that very recently entered clinical test-
in all parts of the heart, and in predisposed patients IKr ing is XEN-D0101 (Xention), which is highly selective
blockers destabilize ventricular repolarization and cause for IKur and has demonstrated anti-arrhythmic efficacy
potentially lethal EAD-related arrhythmias (FIG. 2C)10,11. in two canine AF models18,19.
There has therefore been a search for newer agents with A key issue will be whether IKur block effectively pro-
atrial-selective actions. longs the atrial AP, because the AP changes induced by

1036 | DECEMBER 2006 | VOLUME 5 www.nature.com/reviews/drugdisc


© 2006 Nature Publishing Group
REVIEWS

IKur block will predominantly delay early repolarization A Accelerated normal automaticity
(phases 1 and 2, BOX 1) and move the plateau (phase 2) to
a more positive potential. A more positive plateau activates
more IKr and consequently accelerates late repolarization
(phase 3). The acceleration of phase 3 can totally offset 0
the delaying of phases 1 and 2, causing no net AP pro-
longation20,21. However, in rapidly firing fibrillating tissue

mV
and with atrial remodelling the role of IKr is small and IKur
block prolongs the AP. Another issue is potential accessory
effects on non-cardiac tissue as Kv1.5 is found in vascu-
lar, neural and pancreatic tissues as well as the heart22–24. –80
However, no such effects have been seen in preclinical
studies and results in humans will be a crucial test. B Delayed after-depolarizations
Another potentially interesting target is the acetyl-
choline-related K+ current, IKACh (carried by Kir3.1/3.4
heterotetramers, FIG. 4). IKACh is normally small or absent
in the absence of the parasympathetic neurotransmitter 0
acetylcholine. When acetylcholine is released from para-
sympathetic nerves, it activates IKACh, which has strong
b

mV
atrial AP-shortening and AF-promoting effects. Recent
studies have shown that AF alters IKACh properties, caus-
ing the channel to open even in the absence of acetylcho-
a
line25,26 and resulting in APD abbreviation that promotes –80
AF27. IKACh is absent in the ventricles: blocking IKACh has no
effect on ventricular-cell currents or APs27. IKACh blockers C Early after-depolarizations
therefore might suppress AF without causing ventricular
pro-arrhythmia. Efforts to develop IKACh blockers are still
in their infancy, but one candidate molecular series is
being developed28 and is approaching clinical testing. 0
Na+-channel blockade is highly effective in terminat-
ing AF by causing primary re-entry waves to extinguish29.
RSD1235 (vernakalant; Cardiome) is a recently devel-
mV

oped atrial-selective agent30 that rapidly stops recent-


onset AF in ~50% of patients in clinical Phase II and
III studies31. Its mechanisms of action are incompletely
–80
understood, but INa inhibition with favourable kinetics32
probably has an important role. AZD7009 (AstraZeneca) Figure 2 | Cellular mechanisms of abnormal impulse
is another atrial-selective agent with both Na +- and formation and arrhythmia. A | Normal automaticity in a
K+-current blocking properties33, but it has been with- spontaneously depolarizing pacemaker cell: the cell
drawn from development because of extra-cardiac side reaches a critical threshold voltage (threshold potential,
effects. For a more detailed discussion of atrial-selective red line), at which the Na+ current is strong enough to lead
and other new targets for AF, the interested reader is to the initiation of an action potential (‘fire the cell’; solid
referred to a recent review34. line). Accelerated normal automaticity causes a faster
spontaneous firing rate than normal and results when cells
increase their rate of depolarization and, as a consequence,
Ischaemia-specific targets
spontaneous pacemaker frequency (dashed line), such as
Acute myocardial ischaemia causes highly arrhyth- when local disease induces an increased If (see BOX 1).
mogenic changes in cardiac electrical properties35,36 B | After-depolarizations of a typical ventricular cell: after-
which strongly promote ventricular tachycardias and depolarizations are abnormal ‘hump-like’ depolarizations.
ventricular fibrillation and lead to a high incidence of If they arise after the final repolarization of the cell they are
sudden death in minutes to hours36. called delayed after-depolarizations (DADs; dashed line, a)
In ischaemic tissue, APD is shortened, resting poten- and are generally due to abnormal intracellular Ca2+ release
tial is reduced (causing INa inactivation and reducing the that results in an excessive inward current carried by the
energy available for impulse conduction) and cell-to-cell Na+,Ca2+ exchanger. If the DAD is large enough to reach
coupling is impaired (FIG. 5). Attempts to prevent sud- threshold, it will cause abnormal extra beating (dashed
line, b) before the next expected action potential (solid
den death by suppressing premature beats that trigger
line). C | When after-depolarizations occur before
re-entry37,38 or by K+-channel blocking drugs that pre- complete AP repolarization, they are called ‘early after-
vent re-entry by increasing APD6,8 have failed. Efforts depolarizations’ (EADs) and result from abnormalities in
have shifted to implantable cardioverter-defibrillator repolarization that prolong the AP. APs of two cells are
devices in high-risk individuals39 and measures to com- shown: a cell (upper trace) that fails to repolarize,
bat ischaemia (such as β-adrenoceptor blockers and generating repeated EADs that cause the adjacent cell
revascularization procedures)40. to fire repeatedly (lower trace).

NATURE REVIEWS | DRUG DISCOVERY VOLUME 5 | DECEMBER 2006 | 1037


© 2006 Nature Publishing Group
REVIEWS

However, the occurrence of sudden arrhythmic death Re-entry


due to acute myocardial ischaemia and consequent ven-
tricular fibrillation, particularly as an initial presenting
I
event, remains significant. Developments in our under-
standing of the mechanisms causing ischaemia-induced
II
electrical dysfunction and arrhythmias (FIG. 5) have
opened up some interesting new therapeutic possibilities.
The adenosine-triphosphate sensitive (KATP) K+ channel,
composed of the Kir6.2 and SUR2A subunits (FIG. 4), is a
metabolic sensor that opens during myocardial ischaemia,
b d
and has a key role in ischaemic APD shortening41. KATP
has a complex role, mediating cardioprotective as well as 0 II
arrhythmogenic functions. Recent work has shown that
these actions can be dissociated, with cardioprotective

mV
function mediated largely by a mitochondrial KATP chan-
nel and electrical changes due to a sarcolemmal chan-
a c
nel42. Organic compounds (for example, HMR 1883 and –60
HMR1098/clamikalant; Sanofi-Aventis) selectively block –80 RP I
sarcolemmal KATP channels43, suppress ischaemia-induced
electrocardiographic changes44 and prevent associated Figure 3 | Re-entry. Re-entry as an electrical short-
ventricular fibrillation45. Because of the selectivity of circuit between two cells: re-entry depends on the
their action for acutely ischaemic tissue, they are poten- balance between refractory and conduction properties
of two adjacent tissue zones (I and II) that are
tially promising for the prevention of ischaemic sudden
connected, as shown in the inset. The solid lines
arrhythmic death. If such compounds prove to be suf- represent action potential recordings (a and b,
ficiently selective as to be both safe in relatively low-risk respectively) from zones I (lower trace) and II (upper
patients and effective against ischaemic arrhythmias, they trace) of the tissue without arrhythmia, and dashed
could be given chronically to patients with coronary artery potentials show recording with a re-entrant extra beat.
disease in order to prevent ischaemia-induced malignant Re-entry occurs when an impulse (b) leaves one cell (II
arrhythmias should an ischaemic event ensue. here) and arrives in another zone (I) before cells in the
The Brugada syndrome is a familial condition that, latter zone have fully repolarized (and therefore are
like acute myocardial ischaemia, involves elevation of refractory), setting up a delayed activation (c) of zone I
the ST segment on the electrocardiogram (reflecting (red dashed line). If a cell in zone II has now recovered its
excitability, this action potential (c) can propagate to
abnormal local repolarization; see also FIG. 6b) and
initiate an action potential (d) in zone II. This process can
an increased risk of sudden death due to ventricular occur repeatedly, causing a sustained rapid rhythm
fibrillation 46. As in acute myocardial ischaemia 47, (‘tachycardia’). In order for this mechanism to be
electrophysiological deterioration in the Brugada syn- maintained, the time for the impulse to traverse the
drome can be precipitated by Na+-channel blocking circuit has to be longer than the time required for cells
drugs48. Many genotyped cases involve loss-of-func- to regain excitability after initial firing as determined
tion mutations in cardiac Na+ channels and it has been by their refractory period (RP).
suggested that a key pathophysiological event is very
rapid repolarization in the outer (epicardial) layer of
heart muscle, which has a very large Ito early in the
AP, in the face of reduced inward INa to keep the cell Hyperpolarization-activated nonselective cation chan-
depolarized49. Similar pathophysiology might occur nels. A K+ current showing time-dependent inactiva-
in acute ischaemia, and ventricular tachyarrhythmic tion was initially believed to have a key role in cardiac
death in both acute ischaemia and Brugada syndrome pacemaking, but subsequent studies suggested that the
could be amenable to prevention by Ito blockers. At current involved carries both Na+ and K+ in a relatively
present, no selective Ito blockers are available; however, non-selective fashion and is activated in a time-depend-
the development of compounds targeting the under- ent way at negative potentials50. Because of its unusual
lying Kv4.3–KChIP2 subunit complex could produce ion-selectivity and voltage-dependent properties the
interesting new therapeutic possibilities. current was initially named If for ‘funny current’50. The
Finally, improved understanding of the basic mecha- underlying channel subunits have been cloned and
nisms of cell-to-cell coupling and their pharmacological named hyperpolarization-activated, cyclic nucleotide-
manipulation has yielded new approaches to ischaemia- gated cation channel (HCN) subunits. Four isoforms are
related uncoupling of intercellular communication. known, with HCN4 being particularly important in car-
diac pacemaking51, although various ionic currents con-
Targets that control spontaneous activity tribute to pacemaking function52. Recently developed,
A number of specific cardiac currents are involved in highly selective If inhibitors such as ivabradine (Servier)
the generation or regulation of spontaneous activity and (FIG. 4)53 might be useful for cardiac arrhythmias result-
abnormalities in these currents can cause abnormal local ing from abnormally increased pacemaker function in
cellular activity that generates arrhythmias (FIG. 2). regions that do not normally pace the heart54.

1038 | DECEMBER 2006 | VOLUME 5 www.nature.com/reviews/drugdisc


© 2006 Nature Publishing Group
REVIEWS

IKATP IKr IKs IKACh Ito IKur IK1 If


Na+

hERG KvLQT1 Kv4.3 Kv1.5 Kir2.1 HCN4


Kir6.2 Kir3.1 Kir3.4

MiRP1 KCR1 minK M2 G-protein KChIP2.2 MiRP1


SUR2A
K+ K+ K+ K+ K+ K+ K+ K+
Clamikalant NS1643 HMR-1556 NIP-151 XEN-D0101 (blocker) Ivabradine (blocker)
(blocker) (agonist) (blocker) (blocker) Phase I Phase III

L-364,373
(agonist)

Calstabin2
2+
SR Ca JTV519
RyR2 (stabilizer of
RyR channels)

RSD1235 (vernakalant)
(blocker) GsMTx4 Mibefradil Rotigaptide (agonist)
Phase III iv/II oral (blocker) (blocker) Phase II
INa + ICaT

Nav1.5 Cav3.1/3.2
Cytosol

β β SAC SAC ?
Gap junction Extracellular space
Na+ + Ca2+

Connexin
Connexon

Drug Preclinical Drug Clinical trials (phase indicated) Drug Withdrawn from market/clinical trials

Figure 4 | Ion currents, ion channel subunits and ion transporters implicated in arrhythmogenesis. Schematic of a
cardiac cell, depicting ion channels and ion transporters that are targeted by investigational drugs. SR, sarcoplasmic
reticulum; SAC, stretch activated channel.

Stretch-operated channels. The electrophysiological of stretch-activated cation channels61, was therefore a


characteristics of cardiomyocytes are sensitive to changes significant advance. The d-enantiomer is more resistant
in mechanical stress, and mechanical stimuli (such as to hydrolysis by endogenous proteases and possesses
tissue stretch) can modulate electrical activity55,56. This identical pharmacological activity to the l-enantiomer62.
interrelationship, commonly called ‘mechanoelectri- GsMTx4 is highly effective in suppressing AF promotion
cal feedback’, might be involved in the triggering of by stretch in an isolated rabbit heart model, at concentra-
various atrial and ventricular tachyarrhythmias55,56. tions without effects on action potential characteristics
For example, AF is often associated with atrial enlarge- or refractoriness63. GsMTx4 inhibition of the stretch-
ment57–59. Furthermore, in patients with diminished activated channel does not seem to involve traditional
ventricular function, transient myocardial stretch can lock-and-key protein-protein interactions, but more of
elicit ventricular tachycardias and promote fibrillation. a bilayer-dependent mechanism that modifies channel
Mechanoelectrical feedback involves several currents, gating64. Stretch-activated channels are sensitive to the
including cation fluxes across non-selective stretch-acti- local lipid environment, so increased membrane fluidity,
vated channels found in a variety of cardiac cell types60. as occurs following acute addition of polyunsaturated
Exploration of stretch-activated channels as targets for fatty acids, might attenuate stretch-induced vulnerability
anti-arrhythmic therapy has until recently been hampered to AF by altering physicochemical properties of cardiac
by a lack of specific blockers or modulators. The isolation membranes. A protective effect of dietary fish oil against
of GsMTx4, a peptide from the venom of the tarantula AF in the rabbit atrial-stretch model has recently been
Grammostola spatulata, a selective and potent inhibitor demonstrated65.

NATURE REVIEWS | DRUG DISCOVERY VOLUME 5 | DECEMBER 2006 | 1039


© 2006 Nature Publishing Group
REVIEWS

Table 1a | New anti-arrhythmic drugs in development


Agent and chemical structure Mechanism Therapeutic Stage of Company Refs
of action implications/ development
indications
IKr/IKs Maintenance of SR Phase III Procter &
O N N CH3 blockade Gamble
N
O N O
Cl N

Azimilide

Multichannel Maintenance of SR Phase III Sanofi Aventis


blockade
O O
H
O N
N
S
HCL
O
O

Dronedarone

Multichannel Conversion of AF/ Phase II Sanofi Aventis


O blockade maintenance of SR
O N

O
HO
OH
O

SSR149744C

IKur/Ito/IKACh Conversion of AF Phase II Sanofi Aventis 17


CH3
H blockade
O N
O
O
N N
H

AVE0118

Multichannel Conversion of AF Phase II Solvay


COO–
blockade Pharmaceuticals

–OOC
H COOH
+
N N •2
HOOC
2
Tedisamil

Na+/atrial Conversion of AF/ Phase II/III Cardiome 30


O potassium maintenance of SR Pharma/Astellas
O channel Pharma
blockade

N OH

RSD1235
(Vernakalant)

AF, atrial fibrillation; SR, sinus rhythm.

1040 | DECEMBER 2006 | VOLUME 5 www.nature.com/reviews/drugdisc


© 2006 Nature Publishing Group
REVIEWS

Table 1b | New anti-arrhythmic drugs in development


Agent and chemical structure Mechanism of Therapeutic Stage of Company Refs
action implications/ development
indications
Connexin Ventricular Phase II Wyeth/Zealand 84–86
O
H modulator arrhythmia Pharma
O N C
HO H3C
C NH2
O H
HN HN
O CH3
C C
H O O O
H N C C NH
N H

HO H

Rotigaptide

XEN-D0101 IKur blockade Maintenance of SR Phase I Xention


Structure not disclosed

JTV519 Promotes Animal studies Preclinical Japan Tobacco 18,19


Structure not disclosed calstabin 2– show efficacy in AF
RyR2 binding

NIP-151 IKACh blockade Maintenance of SR Preclinical Nissan 178


Structure not disclosed Pharmaceuticals

O SAN inhibitor SR control Phase III Servier 53


H3CO
OCH3
N N
H3CO CH3
OCH3
Ivabradine

GsMTx4 Blockade of Animal studies Preclinical 61


Polypeptide stretch-activated show efficacy in AF
channels
IKr activator Long QT syndrome Preclinical NeuroSearch AS 91
OH OH
H H
N N

CF3 CF3

NS1643

O IKs blockade Atrial arrhythmias Preclinical Sanofi-Aventis 103,104


S
F N
O
F O OH
F
O

HMR-1556

H3C IKs activator Long QT syndrome Preclinical Merck Research 92


O
N Labs

F N
H

L-364,373

AF, atrial fibrillation; SAN, sino-atrial node; SR, sinus rhythm

NATURE REVIEWS | DRUG DISCOVERY VOLUME 5 | DECEMBER 2006 | 1041


© 2006 Nature Publishing Group
REVIEWS

a Loss of plateau dome b Normal cell-to-cell coupling hypertrophy, and inherited arrhythmogenic diseases
because of INa/Ito imbalance: such as catecholaminergic polymorphic ventricular
block Ito and/or IKATP tachycardia (CPVT)66. By promoting after-depolariza-
0 tions (FIG. 2B), Ca2+-handling abnormalities can lead to
arrhythmias including atrial and ventricular fibrillation.
Ca 2+ handling is a crucial and complex cardio-
myocyte function. Effective contraction requires rapid
mV

Impaired coupling in ischaemia


increases in free cytoplasmic Ca2+ concentration, but
effective relaxation requires rapid restoration of low
–60
Ca2+ concentrations in diastole (the relaxed phase of the
cardiac cycle, corresponding roughly to AP phase 4).
–80
Efficient Ca2+ handling is maintained by a complex set
Cells depolarized by K+ loss
through IKATP: block IKATP Improve coupling with connexin activator of intracellular Ca2+ stores, transporters and channels.
Many membrane electrical functions are regulated by
Figure 5 | Promising targets for ischaemia-selective anti-arrhythmics. a | Diagram of Ca2+, so abnormalities in Ca2+ handling can be highly
a normal ventricular action potential (AP; grey) and an action potential affected by acute
arrhythmogenic. Key components of the Ca2+-handling
myocardial ischaemia (dashed line). Acute ischaemia shortens the duration of the AP and
causes depolarization of the resting membrane potential. The ion channels that carry IKATP
machinery include the sarcoplasmic reticulum (SR)
and Ito are potential drug targets. IKATP has It is well-documented role in ischaemia-induced Ca2+-release channel or ryanodine receptor (the cardiac
intracellular K+ loss and AP abbreviation. The role of Ito in acute ischaemia is not fully form is known as RyR2), the RyR2-binding molecule
established, but based on related physiology in the Brugada syndrome it might also be an calstabin 2, the SR Ca2+-ATPase (the cardiac form is
interesting target for ischaemia-induced arrhythmias. b | Impaired cell-to-cell coupling known as SERCA2A) and the SERCA2A-regulatory
in ischaemia. Cells (depicted by red boxes) are connected with each other by specialized molecule phospholamban (for details, see BOX 2).
gap junctions that maintain electrical continuity (top), allowing for the propagation of an Calstabin 2 is a crucial modulator of RyR2 func-
action potential between cells (direction of propagation shown by arrow). Coupling is tion (FIG. 4) and depletion of calstabin 2 from the RyR2
impaired by ischaemia, causing failure of propagation that results in an inability of the complex (as occurs in congestive heart failure) leads to
propagating impulse to initiate effective action potentials (bottom), promoting impulse
hyperactive RyRs and Ca2+ leak67,68. By contrast, strate-
blockade and the occurrence of re-entry. Connexin activators can reverse ischaemia-
induced coupling abnormalities, improving conduction and suppressing arrhythmias.
gies that increase calstabin 2 binding attenuate Ca2+ leak,
inhibit DADs and suppress triggered arrhythmias69. The
1,4-benzothiazepine JTV519 (Japan Tobacco), which
Targeting the Ca2+-handling machinery promotes calstabin 2–RyR2 binding, reduces diastolic
Abnormalities in intracellular Ca2+ handling occur in a Ca2+ leak, prevents adrenergically mediated polymor-
range of arrhythmogenic conditions, including congestive phic ventricular arrhythmias and sudden cardiac death
heart failure (CHF), ischaemic heart disease, myocardial in calstabin 2+/– haplosufficient mice70,71. JTV519 also

a Long QT syndromes b Brugada syndrome


ECG ECG

ST segment

AP Epicardial
AP
1s
Arrhythmic beat

Endocardial
Epicardial

Figure 6 | Examples of genetically based arrhythmia syndromes. a | Long QT syndrome. Long QT syndromes cause
paroxysmal ventricular tachyarrhythmias of characteristic, polymorphic morphology (Torsades de Pointes, electrocardiogram
(ECG) in upper panel) by impairing repolarization and causing arrhythmogenic early after-depolarizations (EADs) and
ectopic beats (action potential (AP) recordings s in lower panel). b | Brugada syndrome. The top panel shows a typical
Brugada ECG. The horizontal green lines delineate the ‘ST segment’. The characteristic electrocardiographic ST segment
elevation (dashed line, as opposed to normal ECG depicted by the solid line) is believed to be due to rapid repolarization in
the Brugada syndrome of the outer (epicardial) layer of the heart (see lower panel, the Brugada syndrome epicardial AP is
shown by the dashed line; a normal epicardial AP is shown by the black line). Rapid epicardial repolarization in Brugada
syndrome patients results from the large epicardial Ito, which overpowers a diminished Na+ current caused by the Na+
channel loss-of-function mutation that underlies Brugada syndrome. Electrical current flows from normally repolarizing
inner-layer (endocardial, red line) cells to the early-repolarizing epicardium. The resulting large voltage gradient produces
depolarization and reactivation of the endocardial cell, causing extra beats that can initiate ventricular tachyarrhythmias90.

1042 | DECEMBER 2006 | VOLUME 5 www.nature.com/reviews/drugdisc


© 2006 Nature Publishing Group
REVIEWS

Box 2 | Cardiac Ca2+ flux in health and disease


During depolarization of the cell membrane, calcium ICaL
Extracellular NCX
enters the cell via activated L-type Ca2+-channels. The Ca2+
2+ 2+
resulting Ca influx promotes intracellular Ca release
from subcellular stores in the sarcoplasmic reticulum
(SR) by Ca2+-induced Ca2+ release, which greatly
amplifies the initial signal118,119. Ca2+-induced Ca2+
release occurs via specialized Ca2+-release channels, also Na+
Ca2+ PKA
called ryanodine receptors (RyRs) because of their initial
identification via high-affinity binding of the toxin Calstabin 2 CamKII
PP1
ryanodine. Ca2+ release occurs via unitary events called RyR2
119 SERCA
‘sparks’, reflecting activation of clusters of RyRs . The
RyR (the cardiac form is RyR2) constitutes part of a large
macromolecular complex of key accessory proteins P –PLB P P PLB– P
including calmodulin, calstabin 2 (FKBP12.6), protein
2+
kinase A (PKA), Ca /calmodulin-dependent protein
kinase (CaMKII) and protein phosphatases 1 (PP1) and
Ca2+
2A120. Calmodulin is a key Ca2+-binding protein that
regulates the action of key intracellular enzymes such as
the kinase CaMKII and the protein phosphatase
calcineurin. Protein kinase A and CaMKII are both
stimulated by adrenergic activation and phosphorylate
key intracellular regulatory proteins. Calstabin 2 binds to Ca2+
and stabilizes the open and closed states of the RyR.
SR Ca2+ stores are determined by the rate of SR Ca2+
uptake and the rate of Ca2+ release. Ca2+ uptake occurs P –PLB P P PLB– P
via the SR Ca2+-ATP’ase, SERCA (cardiac form is
SERCA2A). SERCA function is negatively regulated by Cytosol SR PP1 SERCA
phospholamban (PLB), but PLB phosphorylation removes
2+
this inhibitory influence. Diastolic Ca leak from
dysfunctional RyRs causes a loss of SR Ca2+, reducing contractility, but might still facilitate delayed after-depolarizations
(DADs) by causing excess diastolic Ca2+ concentrations. In a seminal paper, Marx et al. suggested that in the failing heart RyR
‘hyperphosphorylation’ causes calstabin unbinding, increases RyR Ca2+ sensitivity and makes RyRs functionally ‘leaky’121. RyR
phosphorylation can result from PKA action at a specific serine120 or CaMKII-induced threonine-phosphorylation122.
Abnormalities in intracellular Ca2+ handling occurs in conditions such as congestive heart failure, ischaemic heart disease,
myocardial hypertrophy and atrial fibrillation, as well as in inherited arrhythmogenic diseases such as catecholaminergic
polymorphic ventricular tachycardia (CPVT) in which obvious structural heart disease is absent123. Spontaneous diastolic
Ca2+ release triggers Na+,Ca2+ exchange (NCX), which mitigates Ca2+ loading by extruding Ca2+ in exchange for extracellular
Na+. NCX carries three Na+-ions in for each single Ca2+ ion extruded, and therefore causes movement of one extra positive
ion into the cell for each functional cycle. This excess movement of positive ions into the cell depolarizes the cell
membrane, causing arrhythmic DADs as illustrated in FIG. 3. In addition, NCX activation by abnormal cellular Ca2+ handling
is believed to potentially participate in early after-depolarizations formation.

reduces AF inducibility in dogs with sterile pericardi- constitutes a potential novel anti-arrhythmic strategy,
tis72. Atrial cardiomyocytes from patients with chronic although we are not aware of effective and non-toxic lead
AF or from dogs with pacing-induced AF show RyR2 compounds presently under study.
hyperphosphorylation and calstabin 2 unbinding, which
are corrected by JTV51973. JTV519 might therefore be an Targeting intercellular coupling mechanisms
important potential lead structure for identification of Gap junctions contain clusters of closely packed
compounds targeting calstabin 2–RyR2 binding. hemichannel subunits that connect adjacent cells to
Another important enzyme which forms part of the allow intercellular communication by the passage of ions
RyR2 macromolecular complex is the Ca2+/calmodulin- and small molecules, producing the continuous syncytial
dependent protein kinase (CaMKII), which has been function of myocardial tissue. The conducting structures
shown to contribute to arrhythmogenesis in conditions formed by oligomerization of transmembrane spanning
such as Torsades de Pointes (TdP). TdP is a potentially life- proteins are named connexins, six of which constitute
threatening ventricular arrhythmia that is associated with an individual connexon. Of >20 connexin genes in the
abnormal repolarization, EADs and myocardial re-entry. genome, connexins 43, 40 and 45 are the most abundantly
Because of the very rapid rate of TdP, it can cause ventricu- expressed in the myocardium. Connexin 43 dominates in
lar ischaemia and/or irregular re-entry that degenerates working ventricular and atrial myocytes. Connexin 40 is
into ventricular fibrillation74. CaMKII increases L-type also expressed in atrium, and along with connexin 45 in
Ca2+-channel open probability, potentially facilitating conduction tissue. Changes in gap-junction organization,
channel reactivation that causes EADs, effects attenuated expression and function create a substrate for arrhyth-
by CaMKII inhibitors75,76. CaMKII inhibition therefore mias77, and abnormalities in connexins occur in a range

NATURE REVIEWS | DRUG DISCOVERY VOLUME 5 | DECEMBER 2006 | 1043


© 2006 Nature Publishing Group
REVIEWS

polymorphisms might reproduce some of the abnor-


Box 3 | Gap-junction coupling enhancers as anti-arrhythmic agents malities of monogenic arrhythmic syndromes and con-
In 1980, Aonuma and coworkers124 identified a bovine atrial hexapeptide with anti- tribute to an even more substantial portion of the overall
arrhythmic activity in cultured myocardial cell clusters, designating it anti-arrhythmic sudden death burden88. These insights have resulted in
peptide (AAP). The unique AAP amino-acid sequence (H2N-Gly-Pro-4Hyp-Gly-Ala-Gly- more specific tools for genetic testing, clinical diagnosis
COOH) was later determined. AAP was found not to act on ion currents; rather, anti- and risk stratification as, for example, in the long QT
arrhythmic actions were attributable to improved intercellular coupling by increased syndromes (LQTS). A number of specific paradigms for
gap-junctional conductivity125–127. Subsequently, synthetic analogues have been inherited arrhythmia mechanisms have been developed,
synthesized by altering the amino-acid sequence, exchanging single amino acids and including the LQTS, the short QT syndrome (SQTS), the
producing propionyl derivatives such as the pentapeptide HP-5128,129. Structure– Brugada syndrome and CPVT89,90.
activity relationships were established, allowing Dhein and others to synthesize the
The LQTS (FIG. 6a) promote sudden death by
structural analogue AAP10127. AAP10 inhibited ischaemia-induced conduction
impairing ventricular repolarization, and the genetic
alterations and reduced dispersion in epicardial repolarization127. AAP10 might act
through G-protein-coupled receptor-mediated activation of protein kinase Cα and abnormalities leading to this syndrome include loss-of-
phosphorylation of Connexin 43130. function mutations in subunits underlying IKr, IKs, IK1 and
Practical application remained elusive because of poor enzymatic stability of available gain-of-function mutations in INa and ICaL (for details, see
AAPs, rendering assessment of in vivo anti-arrhythmic efficacy difficult. A major step TABLE 2). Drugs are being developed that accelerate repo-
forward was the development of the stable AAP analogues cyclo-AAP10 and larization by directly enhancing function of IKr (NS1643)
rotigaptide (ZP123) by retro-all-d-amino acid design, resulting in markedly longer or IKs (L-364,373)91,92. Careful use of these compounds
plasma half-lives than traditional AAPs131,132. Rotigaptide attenuates atrial and will be needed to avoid creating a SQTS, but they are the-
ventricular conduction-slowing during acidosis and metabolic stress, reduces action oretically ideal agents for the targeted therapy of patients
potential dispersion and prevents re-entrant ventricular tachycardia induction during at risk of arrhythmias due to repolarization defects.
acute myocardial infarction133–136. By contrast, rotigaptide does not influence focal Arrhythmias in some LQTS, particularly the IKs-deficiency
ventricular tachycardia or triggered activity136. forms, are triggered by adrenergic stimulation, probably
Rotigaptide is currently in early clinical testing for management of atrial and
because of the crucial role of IKs as a ‘brake’ against exces-
ventricular arrhythmias. As for all peptide drugs, a major hurdle still to overcome is poor
sive ICaL-induced plateau prolongation by adrenergic
bioavailability resulting from intestinal absorption barriers137. Attempts to circumvent
this difficulty and to permit practical chronic oral therapy have not been very successful stimulation93. Anti-adrenergic therapy will become bet-
to date, hampered by factors such as toxicity of absorption enhancers, inter-individual ter targeted by evolving pharmacogenomic approaches
variability in absorption and manufacturing costs. If this challenge can be overcome, to guiding therapy. Defective channel-protein trafficking
gap-junction enhancers could become a practical and important addition to the causes most forms of the IKr-deficient LQTS (termed
anti-arrhythmic pharmacopoeia. LQT2)78,94,95, and might contribute to the causation of
other inherited arrhythmia syndromes96. Agents have
been identified that rescue defectively trafficked mutant
of arrhythmia models78–81. Conduction velocity begins to channels by improving their delivery to the cell mem-
be perceptibly reduced when gap-junction coupling is brane97,98. Although the initially identified compounds
reduced by >50% — for example, a two-thirds reduction (such as E-4031) were also channel blockers, which
in coupling decreases conduction speed by ~30%82. would have made dose titration difficult, recently iden-
Increasing gap-junctional coupling could be anti- tified agents (such as fexofenadine) promote channel
arrhythmic by improving cell-to-cell communication trafficking without directly affecting channel function95.
and thereby improving conduction velocity. Anti- Such agents could be used to correct the expression
arrhythmic peptides that act by enhancing coupling were of mutated ion channels in many LQTS patients and
first identified in 198083. By an interesting series of steps therefore provide safe and effective protection against
(BOX 3), this led to the identification of the stable ana- VF precipitated by TdP.
logue rotigaptide (Zealand Pharma/Wyeth). Rotigaptide By contrast, the SQTS is caused by gain-of-function
improves ventricular conduction and prevents arrhyth- mutations in K+ channels that accelerate repolarization
mia generation in the face of acute myocardial ischae- and reduce APD/RP, favouring re-entry (FIG. 3) by making
mia84–86. Rotigaptide might also improve other forms of it easier for the re-entering impulse to find excitable tis-
arrhythmias involving gap junction dysfunction, an area sue at all points in the potential re-entry circuit99–101. The
under active investigation. To date, no clear adverse or development of agents that specifically inhibit the result-
toxic effects of gap-junction enhancement have been ing abnormal current would seem to be an ideal therapy
reported, and the main obstacles to therapeutic applica- for SQTS. For IKr, highly selective blockers (such as dofe-
tion are pharmacokinetic: poor bioavailability and rapid tilide (Pfizer)) are well established102, and highly selective
breakdown in the plasma. IKs blockers (such as HMR-1566 (Sanofi-Aventis)) have
recently been developed103,104. Agents that selectively
Inherited arrhythmia syndromes as a paradigm for new inhibit IK1 might be of great interest for IK1 gain-of-func-
therapeutic approaches. There has been a remarkable tion SQTS and other arrhythmic conditions associated
increase in our understanding of the molecular and with IK1 enhancement, but none are presently known.
genetic basis of inherited lethal arrhythmias over the The Brugada syndrome (FIG. 6b) is associated with
past decade (TABLE 2)87, which has provided a wealth loss-of-function INa mutations, and is believed to pro-
of newer and more specific targets for anti-arrhythmic duce arrhythmias when Ito overpowers INa 90. There is
therapy. In addition to the relatively uncommon pure evidence that Ito inhibition can counteract the associ-
congenital arrhythmia syndromes, common genetic ated electrical and arrhythmic phenotype105, and the

1044 | DECEMBER 2006 | VOLUME 5 www.nature.com/reviews/drugdisc


© 2006 Nature Publishing Group
REVIEWS

Box 4 | Prevention of arrhythmogenic remodelling: ‘upstream therapy’


Alterations in cardiac structure and function — so-called arrhythmogenic remodelling — lead to acquired cardiac
arrhythmia syndromes138,139. Remodelling-induced abnormalities in cardiac structure and electrical function strongly
predispose to atrial fibrillation (AF) and ventricular fibrillation (VF). Treatments are being developed to target
remodelling, guided by the notion that more direct targeting of the arrhythmic substrate will produce novel treatment
approaches that are safe and effective. This approach, sometimes called ‘upstream therapy’, aims to prevent arrhythmia-
generating abnormalities upstream of the final electrical product.
Atrial structural remodelling
Many clinical conditions associated with AF are characterized by prominent atrial fibrosis. In experimental congestive
heart failure (CHF), atrial fibrosis associated with prominent local conduction slowing, which favours the occurrence of
re-entry, seems to be crucial to an increased capacity to maintain AF140,141. The atrial rennin–angiotensin system is strongly
activated by CHF142,143, and its inhibition by converting-enzyme inhibitors142,143 or angiotensin-receptor blockers144
prevents atrial fibrosis and associated AF promotion. Agents interfering with the rennin–angiotensin system have been
applied clinically for AF prevention with apparent success, particularly for patients with structural-remodelling
conditions such as CHF or hypertension145.
Effects of oxidative stress and inflammation
There is evidence for a role of oxidative stress146,147 and inflammation143 in AF-associated remodelling. Both tissue
oxidation and inflammation can alter ion-channel function and contribute to tissue fibrosis, and antioxidant interventions
might prevent atrial remodelling146. Although not indicated for maintenance of sinus rhythm in AF patients, anti-
inflammatory drugs such as glucocorticoids can prevent atrial remodelling148 and the recurrence of clinical AF149.
However, these are not accepted indications and are still experimental. Cholesterol-lowering statin drugs have anti-
oxidant and anti-inflammatory properties and are also effective in preventing atrial remodelling150. They are not indicated
or used specifically for prevention of AF recurrence but many AF patients are given statins for other reasons. As above,
these are not accepted indications and are still experimental.
Atrial-tachycardia remodelling
Rapid atrial firing rates, as occur during AF, change ion-channel function and atrial electrophysiology, and increase
susceptibility to AF induction and maintenance151. Mibefradil, a T-type Ca2+-channel blocker, and amiodarone, a
particularly effective anti-AF drug that also has T-type Ca2+-channel blocking action, reduce atrial tachycardia
remodelling and prevent remodelling-associated AF152,153. Ongoing work seeks to identify additional drugs that prevent
atrial-tachycardia remodelling and which might be useful in preventing AF recurrence. Anti-inflammatory and anti-
oxidant drugs might also work for this indication146,148,150.
Ventricular remodelling
Efforts to prevent ventricular arrhythmogenic remodelling have been more limited to date than those targeting atrial
remodelling. An endothelin-receptor A antagonist prevented prolongation of the duration of the action potential, ionic
remodelling, QT-interval prolongation and ventricular arrhythmias, and improved overall survival in cardiomyopathic
hamsters154. Most present effort is being targeted at functional alterations, like Ca2+-release channel phosphorylation and
SERCA downregulation, caused by ventricular remodelling, rather than remodelling development per se.

development of selective Ito blockers (none of which are treatment of AF and VF when it becomes possible to
presently known) would present a new and valuable tool identify patients with arrhythmias due to intracellular
for the Brugada syndrome and related arrhythmias. Ca2+ leak and treat them with agents that are specific to
CPVTs result when a diastolic Ca 2+ leak occurs, their pathophysiology70–73.
which is typically caused by mutations affecting RyR2
or the SR Ca 2+ -binding protein calsequestrin 106–109 Cell and gene therapy
(BOX 2). This can lead to DADs (FIG. 2B), which, when The limitations in currently available therapy for man-
large enough, can reach firing threshold and cause trig- agement of cardiac arrhythmias have led to considerable
gered activity. Because catecholamines enhance Ca2+ interest in exploring the potential utility of gene- and
entry into cardiac cells by stimulating β-adrenoceptors, cell-transfer therapy. Efforts to date have centred on
situations of enhanced adrenergic drive are the approaches to recreate or amplify cardiac pacemaker
most common triggers for arrhythmias related to activity, to modify electrical conduction and to influ-
abnormal diastolic Ca2+ release. CPVT responds to ence cardiac repolarization110,111. Strategies that have
β-adrenoceptor blockers, but arrhythmias can also been used to create or enhance biological pacemaker
occur in the absence of β-adrenergic drive and many activity in the target region include overexpression of
patients require implanted defibrillators to prevent β2-adrenoceptors to augment adrenergic responsive-
potentially lethal arrhythmias. The development of ness, suppression of IK1 to permit a greater effect of
drugs that stabilize the Ca2+-release channel and prevent endogenous pacemaker currents, enhancement of If by
diastolic Ca2+ leak, such as JTV51970,71, might produce transfer of underlying HCN subunits, and cell therapy
valuable new agents for arrhythmic conditions such as (in combination with genetic engineering) to create new
CPVT associated with diastolic Ca2+ leak-related DADs. pacemaker cells from mesenchymal or embryonic stem
These insights could affect future pharmacological cells111. Strategies to modify intracardiac conduction

NATURE REVIEWS | DRUG DISCOVERY VOLUME 5 | DECEMBER 2006 | 1045


© 2006 Nature Publishing Group
REVIEWS

Table 2 | Genes and functional abnormalities in inherited cardiac arrhythmia syndromes


Locus Gene Protein Functional alteration Mode of inheritance Refs
Long QT syndrome
11p15.5 KCNQ1 IKs potassium channel α-subunit Loss of function Dominant 155
7q35-q36 KCNH2 IKr potassium channel α-subunit Loss of function Dominant 156
3p21 SCN5A INa cardiac sodium channel α-subunit Gain of function Dominant 157
4q25-q27 ANK2 Ankyrin-B Loss of function Dominant 158
21q22.1-q22.2 KCNE1 IKs potassium channel β-subunit Loss of function Dominant 159
21q22.1 KCNE2 IKr potassium channel β-subunit Loss of function Dominant 160
17q23.1-q24.2 KCNJ2 IK1 potassium channel subunit Loss of function Dominant 161
12p13.3 CACNA1C Calcium channel α-subunit Gain of function De novo mutation 162
11p15.5 KCNQ1 IKs potassium channel α-subunit Loss of function Recessive 163
21q22.1-q22.2 KCNE1 IKs potassium channel β-subunit Loss of function Recessive 164
Short QT syndrome
7q35-q36 KCNH2 IKr potassium channel α-subunit Gain of function Dominant 165
11p15.5 KCNQ1 IKs potassium channel α-subunit Gain of function Unknown 166
17q23.1-q24.2 KCNJ2 IK1 potassium channel subunit Gain of function Dominant 167
Brugada syndrome
3p21 SCN5A INa cardiac sodium channel α-subunit Loss of function Dominant 168
Catecholaminergic polymorphic VT
1q42.1-q43 RYR2 Cardiac ryanodine receptor Gain of function Dominant 169
1p13.3-p11 CASQ2 Calsequestrin Gain of function Recessive 170
Idiopathic sick sinus syndrome
15q24-q25 HCN4 If pacemaker channel subunit Loss of function Unknown 171
3p21 SCN5A INa cardiac sodium channel α-subunit Loss of function Recessive 172
Cardiac conduction disease
3p21 SCN5A INa cardiac sodium channel α-subunit Loss of function Dominant 173
Familial atrial fibrillation
11p15.5 KCNQ1 IKs potassium channel α-subunit Gain of function Dominant 174
21q22.1 KCNE2 IKr potassium channel β-subunit Gain of function Dominant 175
17q23 KCNJ2 IK1 potassium channel subunit Gain of function Dominant 176
7q35-q36 KCNH2 IKr potassium channel α-subunit Gain of function Dominant 177

have focussed on interventions that suppress AV nodal Gene therapy has the promise to provide very specific
ICaL through gene transfer of adenoviral vectors carry- modulation of individual currents in selected cardiac
ing inhibitory G-protein (Gαi2) to the AV node or by regions. However, the area of arrhythmia gene- and
targeted Gem (a Ras-related small G-protein) gene cell-transfer therapy is presently still in its infancy. None
transfer to reduce trafficking of ICaL α-subunits to the of the applications described so far have been tested in
sarcolemma110,112. Both interventions reduced ventricu- humans and we are not aware of any ongoing clinical
lar response rate during AF, in pigs and guinea pigs, trials of gene therapy for cardiac arrhythmia treat-
respectively. A few studies have evaluated ion-channel ment. Major hurdles to circumvent include practical
or engineered cardiomyocyte gene transfer to alter approaches for successful transfer to the target region in
action potential morphology in canine ventricular humans, potential risks of pro-arrhythmia, the control
myocytes110. In vitro gene transfer of K+ currents accel- of amplitude, duration and location of gene expression,
erates repolarization in ventricular myocytes isolated and potential toxic effects of the vector or the transgene,
from mice, rats and rabbits113–116. Chamber-selective along with consequences of host immune responses.
gene therapy was recently reported in pigs: atrial gene
transfer of a dominant negative KCNH2 mutant elimi- Prevention of arrhythmogenic remodelling
nated IKr, specifically atrial repolarization, and left the During the past 10–15 years, the mechanisms by which a
ventricles unaffected117. variety of acquired conditions alter cardiac structure and

1046 | DECEMBER 2006 | VOLUME 5 www.nature.com/reviews/drugdisc


© 2006 Nature Publishing Group
REVIEWS

function to induce arrhythmias have been characterized, Conclusions


and are referred to collectively as ‘arrhythmogenic car- The field of anti-arrhythmic drug development was
diac remodelling’. A brief description of some aspects of greatly slowed by the recognition of the important
remodelling and its targeting by innovative anti-arrhythmic potential risks of classical approaches to anti-arrhyth-
therapies is provided in BOX 4. The prevention of arrhyth- mic therapy6. However, recent developments in under-
mogenic remodelling, sometimes called ‘upstream therapy’, standing the mechanisms of cardiac arrhythmias at the
differs from classical anti-arrhythmic drug approaches by molecular, biophysical and organ level have opened
targeting the development of the arrhythmic substrate, up unprecedented opportunities for the provision of
rather than trying to modify the electrical end-product. A safe and effective anti-arrhythmic drug therapy, as
major potential advantage of this approach is that it prom- well as for the development of newer pharmacological
ises to be relatively free of pro-arrhythmic risk, and raises approaches. It is hoped that these opportunities will
the prospect of providing beneficial effects not only for soon materialize into a broad expansion of the available
arrhythmias but for the underlying cardiac abnormalities. pharmaceutical armamentarium.

1. Wolf, P. A., Mitchell, J. B., Baker, C. S., Kannel, W. B. & 16. Peukert, S. et al. Pharmacophore-based search, 33. Carlsson, L., Chartier, D. & Nattel, S. Characterization
D’Agostino, R. B. Impact of atrial fibrillation on synthesis, and biological evaluation of anthranilic of the in vivo and in vitro electrophysiological effects of
mortality, stroke, and medical costs. Arch. Intern. amides as novel blockers of the Kv1.5 channel. Bioorg. the novel antiarrhythmic agent AZD7009 in atrial and
Med. 158, 229–234 (1998). Med. Chem. Lett. 14, 2823–2827 (2004). ventricular tissue of the dog. J. Cardiovasc.
2. van den Berg, M. P., van Gelder, I. C. & van 17. Blaauw, Y. et al. ‘Early’ class III drugs for the treatment Pharmacol. 47, 123–132 (2006).
Veldhuisen, D. J. Impact of atrial fibrillation on of atrial fibrillation: efficacy and atrial selectivity of 34. Page, R. L. & Roden, D. M. Drug therapy for atrial
mortality in patients with chronic heart failure. Eur. J. AVE0118 in remodeled atria of the goat. Circulation fibrillation: where do we go from here? Nature Rev.
Heart Fail. 4, 571–575 (2002). 110, 1717–1724 (2004). Drug Discov. 4, 899–910 (2005).
3. Nattel, S. New ideas about atrial fibrillation 50 years 18. Rivard, L. et al. Electrophysiological and atrial 35. Elharrar, V. & Zipes, D. P. Cardiac electrophysiologic
on. Nature 415, 219–226 (2002). antiarrhythmic effects of a novel IKur/Kv1.5 blocker in alterations during myocardial ischemia. Am. J. Physiol.
4. Miyasaka, Y. et al. Secular trends in incidence of atrial dogs. Heart Rhythm 2 (Suppl.), S180 (2005). 233, H329–H345 (1977).
fibrillation in Olmsted County, Minnesota, 1980 to 19. Shiroshita-Takeshita, A. et al. Electrophysiological and 36. Janse, M. J. & Wit, A. L. Electrophysiological
2000, and implications on the projections for future atrial antiarrhythmic effects of a novel IKur/Kv1.5 mechanisms of ventricular arrhythmias resulting from
prevalence. Circulation 114, 119–125 (2006). blocker in dogs with atrial tachycardia remodeling. myocardial ischemia and infarction. Physiol. Rev. 69,
5. Kimura, K., Minematsu, K., Yamaguchi, T. & Japan Heart Rhythm 3, S183 (2006). 1049–1169 (1989).
Multicenter Stroke Investigators’ Collaboration 20. Courtemanche, M., Ramirez, R. J. & Nattel, S. Ionic 37. Preliminary report: effect of encainide and flecainide
(J-MUSIC). Atrial fibrillation as a predictive factor for targets for drug therapy and atrial fibrillation-induced on mortality in a randomized trial of arrhythmia
severe stroke and early death in 15, 831 patients with electrical remodeling: Insights from a mathematical suppression after myocardial infarction. The Cardiac
acute ischaemic stroke. J. Neurol. Neurosurg. model. Cardiovasc. Res. 42, 477–489 (1999). Arrhythmia Suppression Trial (CAST) Investigators. N.
Psychiatry 76, 679–683 (2005). 21. Wettwer, E. et al. Role of IKur in controlling action Engl. J. Med. 321, 406–412 (1989).
6. Guerra, P. G. et al. Is there a future for antiarrhythmic potential shape and contractility in the human atrium: 38. Effect of the antiarrhythmic agent moricizine on
drug therapy? Drugs 56, 767–781 (1998). influence of chronic atrial fibrillation. Circulation 110, survival after myocardial infarction. The Cardiac
7. The Sicilian Gambit. A new approach to the 2299–2306 (2004). Arrhythmia Suppression Trial II Investigators. N. Engl.
classification of antiarrhythmic drugs based on their 22. Waldron, G. J. & Cole, W. C. Activation of vascular J. Med. 327, 227–233 (1992).
actions on arrhythmogenic mechanisms. Task Force of smooth muscle K+ channels by endothelium-derived 39. Zareba, W. Implantable cardioverter defibrillator
the Working Group on Arrhythmias of the European relaxing factors. Clin. Exp. Pharmacol. Physiol. 26, therapy in postinfarction patients. Curr. Opin. Cardiol.
Society of Cardiology. Circulation 84, 1831–1851 180–184 (1999). 19, 619–624 (2004).
(1991). 23. Chung, S., Saal, D. B. & Kaczmarek, L. K. Elimination 40. Nattel, S. & Waters, D. What is an antiarrhythmic
A classic description of the notion of of potassium channel expression by antisense drug? From clinical trials to fundamental concepts.
mechanistically based anti-arrhythmic therapy. oligonucleotides in a pituitary cell line. Proc. Natl Am. J. Cardiol. 66, 96–99 (1990).
8. Nattel, S. The molecular and ionic specificity of Acad. Sci. USA 92, 5955–5959 (1995). 41. Noma, A. ATP-regulated K+ channels in cardiac
antiarrhythmic drug actions. J. Cardiovasc. 24. Philipson, L. H. et al. Sequence and functional muscle. Nature 305, 147–148 (1983).
Electrophysiol. 10, 272–282 (1999). expression in Xenopus oocytes of a human insulinoma 42. Gross, G. J. The role of mitochondrial KATP channels
9. Wible, B. A. et al. HERG-Lite: a novel comprehensive and islet potassium channel. Proc. Natl Acad. Sci. USA in cardioprotection. Basic Res. Cardiol. 95, 280–284
high-throughput screen for drug-induced hERG risk. J. 88, 53–57 (1991). (2000).
Pharmacol. Toxicol. Methods 52, 136–145 (2005). 25. Ehrlich, J. R. et al. Characterization of a 43. Gogelein, H., Hartung, J., Englert, H. C. &
10. Roden, D. M. Taking the ‘idio’ out of ‘idiosyncratic’: hyperpolarization-activated time-dependent Scholkens, B. A. HMR 1883, a novel cardioselective
predicting torsades de pointes. Pacing Clin. potassium current in canine cardiomyocytes from inhibitor of the ATP-sensitive potassium channel.
Electrophysiol. 21, 1029–1034 (1998). pulmonary vein myocardial sleeves and left atrium. J. Part I: effects on cardiomyocytes, coronary flow and
An important and thoughtful analysis of the factors Physiol. 557, 583–597 (2004). pancreatic β-cells. J. Pharmacol. Exp. Ther. 286,
promoting one of the most important forms of pro- 26. Dobrev, D. et al. The G protein-gated potassium 1453–1464 (1998).
arrhythmia that limits the usefulness of present current I(K, ACh) is constitutively active in patients 44. Li, R. A., Leppo, M., Miki, T., Seino, S. & Marban, E.
anti-arrhythmic drug therapy. with chronic atrial fibrillation. Circulation 112, 3697– Molecular basis of electrocardiographic ST-segment
11. Marban, E. Cardiac channelopathies. Nature 415, 3706 (2005). elevation. Circ. Res. 87, 837–839 (2000).
213–218 (2002). 27. Cha, T. J., Ehrlich, J. R., Chartier, D., Xiao, L. & Nattel, 45. Billman, G. E., Englert, H. C. & Scholkens, B. A. HMR
12. Wang, Z., Fermini, B. & Nattel, S. Sustained S. Kir3-based inward rectifier potassium current: 1883, a novel cardioselective inhibitor of the ATP-
depolarization-induced outward current in human potential role in atrial tachycardia remodeling effects sensitive potassium channel. Part II: effects on
atrial myocytes. Evidence for a novel delayed rectifier on atrial repolarization and arrhythmias. Circulation susceptibility to ventricular fibrillation induced by
K+ current similar to Kv1.5 cloned channel currents. 113, 1730–1737 (2006). myocardial ischemia in conscious dogs. J. Pharmacol.
Circ. Res. 73, 1061–1076 (1993). 28. Matsuda, T. et al. Blockade by NIP-142, an Exp. Ther. 286, 1465–1473 (1998).
13. Feng, J., Wible, B., Li, G.-R., Wang, Z. & Nattel, S. antiarrhythmic agent, of carbachol-induced atrial 46. Brugada, J., Brugada, R. & Brugada, P. Right bundle-
Antisense oligodeoxynucleotides directed against action potential shortening and GIRK1/4 channel. J. branch block and ST-segment elevation in leads V1
Kv1.5 mRNA specifically inhibit ultrarapid delayed Pharmacol. Sci. 101, 303–310 (2006). through V3: a marker for sudden death in patients
rectifier K+ current in cultured adult human atrial 29. Kneller, J. et al. Mechanisms of AF termination by without demonstrable structural heart disease.
myocytes. Circ. Res. 80, 572–579 (1997). pure sodium channel blockade in an ionically-realistic Circulation 97, 457–460 (1998).
The definitive demonstration of the molecular basis mathematical model. Circ. Res. 96, e48–e57 (2005). 47. Elharrar, V., Gaum, W. E. & Zipes, D. P. Effect of drugs
and tissue selectivity of the human ultrarapid 30. Nattel, S., De Blasio, E., Wang, W.-Q. & Beatch, G. N. on conduction delay and incidence of ventricular
delayed rectifier current, which serve as the RSD1235: A novel antiarrhythmic agent with a unique arrhythmias induced by acute coronary occlusion in
underpinning for its development as an atrial- electrophysiological profile that terminates AF in dogs dogs. Am. J. Cardiol. 39, 544–549 (1977).
selective anti-arrhythmic drug target. (abstract). Eur. Heart J. 22 (Suppl.), 448 (2001). 48. Brugada, R. et al. Sodium channel blockers identify
14. Li, G.-R., Feng, J., Yue, L., Carrier, M. & Nattel, S. 31. Roy, D. et al. CRAFT Investigators. A randomized, risk for sudden death in patients with ST-segment
Evidence for two components of delayed rectifier K+ controlled trial of RSD1235, a novel anti-arrhythmic elevation and right bundle branch block but
current in human ventricular myocytes. Circ. Res. 78, agent, in the treatment of recent onset atrial structurally normal hearts. Circulation 101, 510–515
689–696 (1996). fibrillation. J. Am. Coll. Cardiol. 44, 2355–2361 (2000).
15. Han, W., Zhang, L., Schram, G. & Nattel, S. Properties (2004). 49. Di Diego, J. M., Fish, J. M. & Antzelevitch, C. Brugada
of potassium currents in human cardiac Purkinje cells 32. Fedida, D. et al. The mechanism of atrial syndrome and ischemia-induced ST-segment elevation.
of failing human hearts. Am. J. Physiol. (Heart & Circ. antiarrhythmic action of RSD1235. J. Cardiovasc. Similarities and differences. J. Electrocardiol. 38
Physiol.) 283, H2495–H2503 (2002). Electrophysiol. 16, 1227–1238 (2005). (Suppl. 4), 14–17 (2005).

NATURE REVIEWS | DRUG DISCOVERY VOLUME 5 | DECEMBER 2006 | 1047


© 2006 Nature Publishing Group
REVIEWS

50. Brown, H. & Difrancesco, D. Voltage-clamp 74. Anderson, C. L. et al. Most LQT2 mutations reduce 96. Delisle, B. P., Anson, B. D., Rajamani, S. &
investigations of membrane currents underlying pace- Kv11.1 (hERG) current by a class 2 (trafficking-deficient) January, C. T. Biology of cardiac arrhythmias: ion
maker activity in rabbit sino-atrial node. J. Physiol. mechanism. Circulation 113, 365–373 (2006). channel protein trafficking. Circ. Res. 94,
308, 331–351 (1980). 75. Dzura, I., Wu, Y., Colbram, R. J., Balser, J. R. & 1418–1428 (2004).
51. Stieber, J., Hofmann, F. & Ludwig, A. Pacemaker Anderson, M. E. Calmodulin kinase determines 97. Rajamani, S., Anderson, C. L., Anson, B. D. & January,
channels and sinus node arrhythmia. Trends calcium-dependent facilitation of L-type calcium C. T. Pharmacological rescue of human K+ channel
Cardiovasc. Med. 14, 23–28 (2004). channels. Nature Cell Biol. 2, 173–177 (2000). long QT2 mutations: human ether-a-go-go-related
52. Saint, D. A. Pacemaking in the heart: the interplay of 76. Anderson, M. E. et al. Brugada syndrome. Report gene rescue without block. Circulation 105,
ionic currents. Clin. Exp. Pharmacol. Physiol. 25, from the second consensus conference. Heart Rhythm 2830–2835 (2002).
841–846 (1998). 2, 429–440 (2005). 98. Gong, Q., Anderson, C. L., January, C. T. &
53. Difrancesco, D. Funny channels in the control of 77. Gutstein, D. E. et al. Conduction slowing and sudden Zhou, Z. Pharmacological rescue of trafficking
cardiac rhythm and mode of action of selective arrhythmic death in mice with cardiac-restricted defective HERG channels formed by coassembly of
blockers. Pharmacol. Res. 53, 399–406 (2006). inactivation of connexin43. Circ. Res. 88, 333–339 wild-type and long QT mutant N470D subunits.
54. Hsia, H. H. & Marchlinski, F. E. Electrophysiology (2001). Am. J. Physiol. Heart Circ. Physiol. 287, H652–
studies in patients with dilated cardiomyopathies. 78. De Groot, J. T. & Coronel, R. Acute ischemia-induced H658 (2004).
Card. Electrophysiol. Rev. 6, 472–481 (2002). gap junctional uncoupling and arrhythmogenesis. 99. Brugada, R. et al. Sudden death associated with
55. Nazir, S. A. & Lab, M. J. Mechanoelectric feedback Cardiovasc. Res. 62, 323–334 (2004). short-QT syndrome linked to mutations in HERG.
and atrial arrhythmias. Cardiovasc. Res. 32, 52–61 79. Akar, F. G., Spragg, D. D., Tunin, R. S., Kass, D. A. & Circulation 109, 30–35 (2004).
(1996). Tomaselli, G. F. Mechanisms underlying conduction 100. Bellocq, C. et al. Mutation in the KCNQ1 gene leading
56. Lab, M. J. Mechanoelectric feedback (transduction) in slowing and arrhythmogenesis in nonischemic dilated to the short QT-interval syndrome. Circulation 109,
heart: concepts and implications. Cardiovasc. Res. 32, cardiomyopathy. Circ. Res. 95, 717–725 (2004). 2394–2397 (2004).
3–14 (1996). 80. Firouzi, M. et al. Association of human connexin40 101. Priori, S. G. et al. A novel form of short QT syndrome
57. Henry, W. L. et al. Relation between gene polymorphisms with atrial vulnerability as a risk (SQT3) is caused by a mutation in the KCNJ2 gene.
echocardiographically determined left atrial size and factor for idiopathic atrial fibrillation. Circ. Res. 95, Circ. Res. 96, 800–807 (2005).
atrial fibrillation. Circulation 53, 273–279 (1976). e29–e33 (2004). 102. Tamargo, J., Caballero, R., Gomez, R.,
58. Vaziri, S. M., Larsson, M. G., Benjamin, E. J. & Levy, 81. van der Velden, H. M. et al. Altered pattern of Valenzuela, C. & Delpon, E. Pharmacology of cardiac
D. Echocardiographic predictors of nonrheumatic connexin40 distribution in persistent atrial fibrillation potassium channels. Cardiovasc. Res. 62, 9–33
atrial fibrillation: the Framingham Heart Study. in the goat. J. Cardiovasc. Electrophysiol. 9, 596–607 (2004).
Circulation 89, 724–730 (1994). (1998). 103. Thomas, G. P., Gerlach, U. & Antzelevitch. C. HMR
59. Zou, R., Kneller, J., Leon, L. J. & Nattel, S. Substrate 82. Shaw, R. M. & Rudy, Y. Ionic mechanisms of 1556, a potent and selective blocker of slowly
size as a determinant of fibrillatory activity propagation in cardiac tissue. Roles of the sodium and activating delayed rectifier potassium current. J.
maintenance in a mathematical model of canine L-type calcium currents during reduced excitability Cardiovasc. Pharmacol. 41, 140–147 (2003).
atrium. Am. J. Physiol. (Heart Circ. Physiol.) 289, and decreased gap junction coupling. Circ. Res. 81, 104. Butcher, J. W. et al. Novel 5-cyclopropyl-1, 4-
H1002–H1012 (2005). 727–741 (1997). benzodiazepin-2-ones as potent and selective I(Ks)-
60. Hu, H. & Sachs, F. Stretch-activated ion channels in 83. Aonuma, S. et al. Studies on heart. XIX. Isolation of an blocking class III antiarrhythmic agents. Bioorg. Med.
the heart. J. Mol. Cell. Cardiol. 29, 1511–1523 atrial peptide that improves the rhythmicity of Chem. Lett. 13, 1165–1168 (2003).
(1997). cultured myocardial cell clusters. Chem. Pharm. Bull. 105. Antzelevitch, C. et al. Brugada syndrome: report of the
61. Suchyna, T. M. et al. The identification of a peptide (Tokyo) 28, 3332–3339 (1980). second consensus conference. Heart Rhythm 2, 429–
toxin from Grammostola spatulata spider venom that 84. Kjølbye, A. L., Knudsen, C. B., Jepsen, T., Larsen, B. D. 440 (2005).
blocks cation-selective stretch-activated channels. J. & Petersen, J. S. Pharmacological characterization of 106. Marks, A. R., Priori, S., Memmi, M., Kontula, K. &
Gen. Physiol. 115, 583–598 (2000). the new stable antiarrhythmic peptide analog Ac-D-Pro Laitinen, P. J. Involvement of the cardiac ryanodine
62. Gottlieb, P. A., Suchyna, T. M., Ostrow, L. W. & Sachs, -D-Hyp-Gly-D-Ala-Gly-NH2 (ZP123): In vivo and in vitro receptor/calcium release channel in catecholaminergic
F. Mechanosensitive ion channels as drug targets. studies. J. Pharmacol. Exp. Ther. 306, 1191–1199 polymorphic ventricular tachycardia. J. Cell. Physiol.
Curr. Drug Targets-CNS Neurol. Disord. 4, 287–295 (2003). 190, 1–6 (2002).
(2004). 85. Xing, D. et al. ZP123 increases gap junctional 107. Francis, J., Sankar, V., Nair, V. K. & Priori, S. G.
63. Bode, F., Sachs, F. & Franz, M. R. Tarantula peptide conductance and prevents reentrant ventricular Catecholaminergic polymorphic ventricular
inhibits atrial fibrillation. Nature 409, 35–36 (2001). tachycardia during myocardial ischemia in open chest tachycardia. Heart Rhythm 2, 550–554 (2005).
A key description of the role of tissue stretch in dogs. J. Cardiovasc. Electrophysiol. 14, 510–520 108. Nam, G. B., Burashnikov, A. & Antzelevitch, C. Cellular
atrial fibrillation and of the potential of stretch- (2003). mechanisms underlying the development of
channel inhibitors to suppress the arrhythmia. 86. Haugan, K. et al. The antiarrhythmic peptide analog catecholaminergic ventricular tachycardia. Circulation
64. Suchyna, T. M. et al. Bilayer-dependent inhibition of ZP123 prevents atrial conduction slowing during 111, 2727–2733 (2005).
mechanosensitive channels by neuroactive peptide metabolic stress. J. Cardiovasc. Electrophysiol. 16, 109. Jiang, D. et al. Enhanced store overload-induced Ca2+
enantiomers. Nature 430, 235–240 (2004). 537–545 (2005). release and channel sensitivity to luminal Ca2+
65. Ninio, D. M., Murphy, K. J., Howe, P. R. & Saint, D. A. 87. Kääb, S. & Schulze-Bahr, E. Susceptibility genes and activation are common defects of RyR2 mutations
Dietary fish oil protects against stretch-induced modifiers for cardiac arrhythmias. Cardiovasc. Res. linked to ventricular tachycardia and sudden death.
vulnerability to atrial fibrillation in a rabbit model. J. 67, 397–413 (2005). Circ. Res. 97, 1173–1181 (2005).
Cardiovasc. Electrophysiol. 16, 1–6 (2005). 88. Bezzina, C. R. et al. Common sodium channel 110. Donahue, J. K., Kikuchi, K. & Sasano, T. Gene therapy
66. Scoote, M. & Williams, A. J. Myocardial calcium promoter haplotype in asian subjects underlies for cardiac arrhythmias. Trends Cardiovasc. Med. 15,
signalling and arrhythmia pathogenesis. Biochem. variability in cardiac conduction. Circulation 113, 219–224 (2005).
Biophys. Res. Comm. 322, 1286–1309 (2004). 338–344 (2006). A careful look at novel gene-transfer therapy
67. Marx, S. O. et al. PKA phosphorylation dissociates 89. Priori, S. G. Inherited arrhythmogenic diseases. The approaches for cardiac arrhythmias by one of the
FKBP12. 6 from the calcium release channel complexity beyond monogenic disorders. Circ. Res. leaders in the field.
(ryanodine receptor): defective regulation in failing 94, 140–145 (2004). 111. Rosen, M. R. Biological pacemaking: in our lifetime?
hearts. Cell 101, 365–376 (2000). 90. Antzelevitch, C., Brugada, P., Brugada, J. & Brugada, Heart Rhythm 2, 418–428 (2005).
68. Lehnart, S. E., Wehrens, X. H. T. & Marks, A. R. R. Brugada syndrome: from cell to bedside. Curr. A detailed review of a very promising new approach
Calstabin deficiency, ryanodine receptors, and sudden Probl. Cardiol. 30, 9–54 (2005). to a particular class of arrhythmias, with
death. Biochem. Biophys. Res. Comm. 322, 1267– 91. Xu, X. et al. Increasing I(Ks) corrects abnormal genetically engineered biological pacemakers that
1279 (2004). repolarization in rabbit models of acquired LQT2 and might replace the long-standard electronic
69. Doggrell, S. A. Stabilisation of calstabin2 — a new ventricular hypertrophy. Am. J. Physiol. Heart Circ. implantable devices.
approach in sudden cardiac death. Expert Opin. Ther. Physiol. 283, H664–H670 (2002). 112. Murata, M., Cingolani, E., McDonald, A. D., Donahue,
Targets 9, 955–962 (2005). 92. Casis, O., Olesen, S. P. & Sanguinetti, M. C. J. K. & Marbán, E. Creation of a genetic calcium
70. Yano, M. et al. FKBP12. 6 mediated stabilization of Mechanism of action of a novel human ether-a-go-go- channel blocker by targeted Gem gene transfer in the
calcium release channel (ryanodine receptors) as a related gene channel activator. Mol. Pharmacol. 69, heart. Circ. Res. 95, 398–405 (2004).
novel therapeutic strategy against heart failure. 658–665 (2006). 113. Johns, D. C. et al. Adenovirus-mediated expression of
Circulation 107, 477–484 (2003). Mechanistic description of a novel potassium- a voltage-gated potassium channel in vitro (rat cardiac
71. Wehrens, X. H. et al. Protection from cardiac channel-activating drug that could provide an myocytes) and in vivo (rat liver). A novel strategy for
arrhythmia through ryanodine-receptor stabilizing important new therapeutic approach. modifying excitability. J. Clin. Invest. 96, 1152–1158
protein calstabin2. Science 304, 292–296 93. Han, W., Wang, Z. & Nattel, S. Slow delayed rectifier (1995).
(2004). current and repolarization in canine cardiac Purkinje 114. Nuss, B., Marbán, E. & Johns, D. Overexpression of a
A clear demonstration of the potential value cells. Am. J. Physiol. (Heart Circ. Physiol.) 280, human potassium channel suppresses cardiac
of calcium-release channel stabilization in H1075–H1080 (2001). hyperexcitability in rabbit ventricular myocytes. J.
treating potentially malignant cardiac 94. Zhou, Z., Gong, Q., Epstein, M. L. & January, C. T. Clin. Invest. 103, 889–896 (1999).
arrhythmias. HERG channel dysfunction in human long QT 115. Ennis, I., Li, R., Murphy, A. & Nuss, H. Dual gene
72. Kumagai, K., Nakashima, H., Gondo, N. & Saku, K. syndrome: intracellular transport and functional therapy with SERCA1 and Kir 2.1 abbreviates
Antiarrhythmic effects of JVT-519, a novel defects. J. Biol. Chem. 273, 21061–21066 (1998). excitation without suppressing contractility. J. Clin.
cardiprotective drug, on atrial fibrillation/flutter in a 95. Robertson, G. A. & January, C. T. HERG trafficking and Invest. 109, 393–400 (2002).
canine sterile pericarditis model. J. Cardiovasc. pharmacological rescue of LQTS-2 mutant channels. 116. Kodirov, S., Brunner, M., Busconi, L. & Koren, G. Long-
Electrophysiol. 14, 880–884 (2003). Handb. Exp. Pharmacol. 171, 349–355 (2006). term restitution of 4-aminopyridine-sensitive currents
73. Vest, J. A. et al. Defective cardiac ryanodine receptor A detailed review of a novel pharmacological class: in Kv1DN ventricular myocytes using adeno-
regulation during atrial fibrillation. Circulation 111, drugs that rescue channels that fail to traffic associated virus-mediated delivery of Kv1.5. FEBS
2025–2032 (2005). normally to the cell membrane. Lett. 550, 74–78 (2003).

1048 | DECEMBER 2006 | VOLUME 5 www.nature.com/reviews/drugdisc


© 2006 Nature Publishing Group
REVIEWS

117. Kikuchi, K., McDonald, A. D., Sasano, T. & Donahue, 137. Hamman, J. H., Enslin, G. M. & Kotzé, A. F. Oral 159. Splawski, I., Tristani-Firouzi, M., Lehmann, M. H.,
J. K. Targeted modification of atrial electrophysiology delivery of peptide drugs. Barriers and developments. Sanguinetti, M. C. & Keating, M. T. Mutations in the
by homogenous transmural atrial gene transfer. Biodrugs 19, 165–177 (2005). hminK gene cause long QT syndrome and suppress
Circulation 111, 264–270 (2005). 138. Nattel, S., Maguy, A., Le Bouter, S. & Yeh, Y. H. I(Ks) function. Nature Genet. 17, 338–340 (1997).
118. Fabiato, A. & Fabiato, F. Contractions induced by a Arrhythmogenic ion-channel remodeling in the heart. 160. Abbott, G. W. et al. MiRP1 forms IKr potassium
calcium-triggered release of calcium from the Physiol. Rev. (in the press). channels with HERG and is associated with cardiac
sarcoplasmic reticulum of single skinned cardiac cells. 139. Nattel, S., Shiroshita-Takeshita, A., Cardin, S. & arrhythmia. Cell 97, 175–187 (1999).
J. Physiol. 249, 469–495 (1975). Pelletier, P. Mechanisms of atrial remodeling and 161. Plaster, N. M. et al. Mutations in Kir2.1 cause the
119. Bers, D. M. Cardiac excitation-contraction coupling. clinical relevance. Curr. Opin. Cardiol. 20, 21–25 developmental and episodic electrical phenotypes of
Nature 415, 198–205 (2002). (2005). Andersen’s syndrome. Cell 105, 511–519 (2001).
120. Lehnart, S. E., Wehrens, X. H. T. & Marks, A. R. A detailed analysis of the therapeutic implications 162. Splawski, I. et al. Ca(V)1.2 calcium channel
Calstabin deficiency, ryanodine receptors, and sudden of atrial remodelling, including implications for dysfunction causes a multisystem disorder including
death. Biochem. Biophys. Res. Comm. 322, 1267– anti-arrhythmic drug use and development and arrhythmia and autism. Cell 119, 19–31 (2004).
1279 (2004). potential for novel pharmaceutical targets in the 163. Neyroud, N. et al. A novel mutation in the potassium
121. Marx, S. O. et al. PKA phosphorylation dissociates treatment of atrial fibrillation. channel gene KVLQT1 causes the Jervell and Lange–
FKBP12. 6 from the calcium release channel 140. Li, D., Fareh, S., Leung, T. K. & Nattel, S. Promotion of Nielsen cardioauditory syndrome. Nature Genet. 15,
(ryanodine receptor): defective regulation in failing atrial fibrillation by heart failure in dogs: Atrial 186–189 (1997).
hearts. Cell 101, 365–376 (2000). remodeling of a different sort. Circulation 100, 87–95 164. Schulze-Bahr, E. et al. KCNE1 mutations cause Jervell
122. Ai, X., Curran, J. W., Shannon, T. R., Bers, D. M. (1999). and Lange–Nielsen syndrome. Nature Genet. 17,
& Pogwizd, SM. Ca2+(calmodulin-dependent protein 141. Cha, T. J. et al. Dissociation between ionic remodeling 267–268 (1997).
kinase modulates cardiac ryanodine receptor and ability to sustain atrial fibrillation during recovery 165. Brugada, R., Hong, K. & Dumaine, R. Sudden death
phosphorylation and sarcoplasmic reticulum Ca2+ from experimental congestive heart failure. Circulation associated with short-QT syndrome linked to
leak in heart failure. Circ. Res. 97, 1314–1322 109, 412–418 (2004). mutations in HERG. Circulation 109, 30–35 (2004).
(2005). 142. Li, D. et al. Effects of angiotensin-converting enzyme 166. Bellocq, C. et al. Mutation in the KCNQ1 gene leading
123. Scoote, M. & Williams, A. J. Myocardial calcium inhibition on the development of the atrial fibrillation to the short QT-interval syndrome. Circulation 109,
signalling and arrhythmia pathogenesis. Biochem. substrate in dogs with ventricular tachypacing-induced 2394–2397 (2004).
Biophys. Res. Comm. 322, 1286–1309 (2004). congestive heart failure. Circulation 104, 2608–2614 167. Priori, S. G. et al. A novel form of short QT syndrome
124. Aonuma, S. et al. Studies on heart. XIX. Isolation of an (2001). (SQT3) is caused by a mutation in the KCNJ2 gene.
atrial peptide that improves the rhythmicity of 143. Cardin, S. et al. Evolution of the atrial fibrillation Circ. Res. 96, 800–807 (2005).
cultured myocardial cell clusters. Chem. Pharm. Bull. substrate in experimental congestive heart failure: 168. Chen, Q. et al. Genetic basis and molecular
(Tokyo). 28, 3332–3339 (1980). Angiotensin-dependent and independent pathways. mechanism for idiopathic ventricular fibrillation.
125. Aonuma, S., Kohama. Y., Makino, T. & Fujisawa, T. Cardiovasc. Res. 60, 315–325 (2003). Nature 392, 293–296 (1998).
Studies of heart. XXI. Amino acid sequence of 144. Kumagai, K. et al: Effects of angiotensin II type 1 169. Priori, S. G. et al. Mutations in the cardiac ryanodine
antiarrhythmic peptide (AAP) isolated from atria. J. receptor antagonist on electrical and structural receptor gene (hRyR2) underlie catecholaminergic
Pharmacobiodyn. 5, 40–48 (1982). remodeling in atrial fibrillation. J. Am. Coll. Cardiol. polymorphic ventricular tachycardia. Circulation 103,
126. Argentieri, T., Cantor, E. & Wiggins, J. R. 41, 2197–2204 (2003). 196–200 (2001).
Antiarrhythmic peptide has no direct cardiac actions. 145. Healey, J. S. et al. Prevention of atrial fibrillation with 170. Viatchenko-Karpinski, S. et al. Abnormal calcium
Experientia 45, 737–738 (1989). angiotensin-converting enzyme inhibitors and signaling and sudden cardiac death associated with
127. Dhein, S. et al. A new synthetic antiarrhythmic peptide angiotensin receptor blockers: a meta-analysis. J. Am. mutation of calsequestrin. Circ. Res. 94, 471–477
reduces dispersion of epicardial activation recovery Coll. Cardiol. 45, 1832–1839 (2005). (2004).
interval and diminishes alterations of epicardial 146. Carnes, C. A. et al: Ascorbate attenuates atrial pacing- 171. Schulze-Bahr, E. et al. Pacemaker channel dysfunction
activation patterns induced by regional ischemia. induced peroxynitrite formation and electrical in a patient with sinus node disease. J. Clin. Invest.
Naunyn-Schmiedeberg´s Arch. Pharmacol. 350, remodeling and decreases the incidence of 111, 1537–1545 (2003).
174–184 (1994). postoperative atrial fibrillation. Circ. Res. 89, E32– 172. Benson, D. W. et al. Congenital sick sinus syndrome
128. Kohama, Y., Okimoto, N., Mimura, T., Fukaya, C., E38 (2001). caused by recessive mutations in the cardiac sodium
Watanabe, M., Yokoyama, K. A new antiarrhythmic 147. Kim, Y. M. et al. A myocardial Nox2 containing channel gene (SCN5A). J. Clin. Invest. 112, 1019–
peptide, N-3-(4-hydroxyphenyl)propionyl-Pro-Hyp-Gl NAD(P)H oxidase contributes to oxidative stress in 1028 (2003).
y-Ala-Gly. Chem. Pharm. Bull. 35, 3928–3930 human atrial fibrillation. Circ. Res. 97, 629–636 173. Schott, J. J. et al. Cardiac conduction defects
(1987). (2005). associate with mutations in SCN5A. Nature Genet.
129. Dhein, S. & Tudyka, T. Therapeutic potential of 148. Shiroshita-Takeshita, A., Brundel, B. J. J. M., Lavoie, J. 23, 20–21 (1999).
antiarrhythmic peptides. Cellular coupling as a new & Nattel, S. Prednisone prevents atrial fibrillation 174. Chen, Y. H. et al. KCNQ1 gain-of-function mutation in
antiarrhythmic target. Drugs. 49, 851–855 (1995). promotion by atrial tachycardia remodeling in dogs. familial atrial fibrillation. Science 299, 251–254
130. Weng, S., Lauven, M., Schaeffer, T., Polontchouk, L., Cardiovasc. Res. 69, 865–875 (2006). (2003).
Grover, R., Dhein, S. Pharmacological modification of 149. Dernellis, J. & Panaretou, M. Relationship between 175. Yang, Y. et al. Identification of a KCNE2 gain-of-
gap junction coupling by an antiarrhythmic peptide via C-reactive protein concentrations during function mutation in patients with familial atrial
protein kinase C activation. FASEB J. 16, 1114–1116 glucocorticoid therapy and recurrent atrial fibrillation. fibrillation. Am. J. Hum. Genet. 75, 899–905
(2002). Eur. Heart J. 25, 1100–1107 (2004). (2004).
131. Grover, R. & Dhein, S. Structure-activity relationships 150. Shiroshita-Takeshita, A., Schram, G., Lavoie, J. & 176. Xia, M. et al. A Kir2.1 gain-of-function mutation
of novel peptides related to the antiarrhythmic Nattel, S. The effect of simvastatin and antioxidant underlies familial atrial fibrillation. Biochem. Biophys.
peptide AAP10 which reduce the dispersion of vitamins on atrial fibrillation-promotion by atrial Res. Commun. 332, 1012–1019 (2005).
epicardial action potential duration. Peptides 22, tachycardia remodeling in dogs. Circulation 110, 177. Hong, K., Bjerregaard, P., Gussak, I. & Brugada, R.
1011–1021 (2001). 2313–2319 (2004). Short QT syndrome and atrial fibrillation caused by
132. Kjølbye, A. L., Knudsen, C. B., Jepsen, T., Larsen, B. D. 151. Wijffels, M. C. et al: Atrial fibrillation begets atrial mutation in KCNH2. J. Cardiovasc. Electrophysiol. 4,
& Petersen, J. S. Pharmacological characterization of fibrillation. A study in awake chronically instrumented 394–396 (2005).
the new stable antiarrhythmic peptide analog Ac-D-Pro goats. Circulation 92, 1954–1968 (1995). 178. Hashimoto, N. et al. A novel IKACh channel blocker ,
-D-Hyp-Gly-D-Ala-Gly-NH2 (ZP123): In vivo and in vitro 152. Shinagawa, K., Shiroshita-Takeshita, A., Schram, G. & NIP-151, terminates atrial fibrillation with atrial
studies. J. Pharmacol. Exp. Ther. 306, 1191–1199 Nattel, S. Effects of antiarrhythmic drugs on specific effective refractory period prolongation.
(2003). fibrillation in the remodeled atrium: Insights into the Circulation 112, Abs II–191 (2005).
The first description of the pharmacology of a mechanism of the superior efficacy of amiodarone.
stable gap-junction-enhancing anti-arrhythmic Circulation 107, 1440–1446 (2003). Ackowledgements
peptide. 153. Fareh, S., Bénardeau, A., Thibault, B. & Nattel, S. The Supported by the Canadian Institutes of Health Research, the
133. Xing, D. et al. ZP123 increases gap junctional T-type Ca2+ channel blocker mibefradil prevents the Quebec Heart and Stroke Foundation and the Mathematics
conductance and prevents reentrant ventricular development of a substrate for atrial fibrillation by of Complex Systems and Information Technology (MITACS)
tachycardia during myocardial ischemia in open chest tachycardia-induced atrial remodeling in dogs. Network of Centers of Excellence.
dogs. J. Cardiovasc. Electrophysiol. 14, 510–520 Circulation 100, 2191–2197 (1999).
(2003). 154. Matsumoto, Y. et al. Long-term endothelin a receptor Competing financial interests
134. Eloff, B. C., Gilat, E., Wan, X. & Rosenbaum D. S. blockade inhibits electrical remodeling in The authors declare competing financial interests: see Web
Pharmacological modulation of cardiac gap junctions cardiomyopathic hamsters. Circulation 106, 613–619 version for details.
to enhance cardiac conduction. Evidence supporting a (2002).
novel target for antiarrhythmic therapy. Circulation 155. Wang, Q. et al. Positional cloning of a novel potassium
108, 3157–3163 (2003). channel gene: KVLQT1 mutations cause cardiac DATABASES
135. Haugan, K. et al. The antiarrhythmic peptide analog arrhythmias. Nature Genet. 12, 17–23 (1996). The following terms in this article are linked online to:
ZP123 prevents atrial conduction slowing during 156. Curran, M. E. et al. A molecular basis for cardiac Entrez Gene:
metabolic stress. J. Cardiovasc. Electrophysiol. 16, arrhythmia: HERG mutations cause long QT http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=gene
537–545 (2005). syndrome, Cell 80, 795–803 (1995). Calstabin 2 | Connexin 40 | Connexin 43 | Connexin 45 |
136. Xing, D., Kjølbye, A. L., Petersen, J. S. & Martins, J. B. 157. Bennett, PB., Yazawa, K. & Makita, N. Molecular RyR2 | SERCA2A
Pharmacological stimulation of cardiac gap junction mechanism for an inherited cardiac arrhythmia. OMIM:
coupling does not affect ischemia-induced focal Nature 376, 683–685 (1995). http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=OMIM
ventricular tachycardia or triggered activity in dogs. 158. Mohler, P. J. et al. Ankyrin-B mutation causes type 4 Brugada syndrome
Am. J. Physiol. Heart Circ. Physiol. 288, H511–H516 long-QT cardiac arrhythmia and sudden cardiac death. Access to this links box is available online.
(2005). Nature 421, 634–639 (2003).

NATURE REVIEWS | DRUG DISCOVERY VOLUME 5 | DECEMBER 2006 | 1049


© 2006 Nature Publishing Group

You might also like