Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Pharmacology & Therapeutics 119 (2008) 199–209

Contents lists available at ScienceDirect

Pharmacology & Therapeutics


j o u r n a l h o m e p a g e : w w w. e l s ev i e r. c o m / l o c a t e / p h a r m t h e r a

Preclinical Torsades-de-Pointes Screens: Advantages and limitations of surrogate and


direct approaches in evaluating proarrhythmic risk
Gary A. Gintant ⁎
Department of Integrative Pharmacology, Abbott Laboratories (Dept. R46R, Bldg AP-9), 100 Abbott Park Road, Abbott Park, IL 60064-6119, United States

A R T I C L E I N F O A B S T R A C T

Keywords: The successful development of novel drugs requires the ability to detect (and avoid) compounds that may
Torsades-de-Pointes provoke Torsades-de-Pointes (TdeP) arrhythmia while endorsing those compounds with minimal torsado-
QT genic risk. As TdeP is a rare arrhythmia not readily observed during clinical or post-marketing studies,
Preclinical studies numerous preclinical models are employed to assess delayed or altered ventricular repolarization (surrogate
hERG
markers linked to enhanced proarrhythmic risk). This review evaluates the advantages and limitations of
Proarrhythmia model
Delayed repolarization
selected preclinical models (ranging from the simplest cellular hERG current assay to the more complex in vitro
perfused ventricular wedge and Langendorff heart preparations and in vivo chronic atrio-ventricular (AV)-
node block model). Specific attention is paid to the utility of concentration–response relationships and
“risk signatures” derived from these studies, with the intention of moving beyond predicting clinical QT
prolongation and towards prediction of TdeP risk. While the more complex proarrhythmia models may be
suited to addressing questionable or conflicting proarrhythmic signals obtained with simpler preclinical
assays, further benchmarking of proarrhythmia models is required for their use in the robust evaluation of
safety margins. In the future, these models may be able to reduce unwarranted attrition of evolving compounds
while becoming pivotal in the balanced integrated risk assessment of advancing compounds.
© 2008 Elsevier Inc. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 199
2. General considerations (and limitations) of using surrogate markers to evaluate proarrhythmic risk . . . . 200
3. hERG assay . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 201
4. In vitro arterially-perfused left ventricular wedge preparations . . . . . . . . . . . . . . . . . . . . . 202
5. In vitro Langendorff perfused isolated heart preparations . . . . . . . . . . . . . . . . . . . . . . . . 203
6. A comparison of in vitro proarrhythmia assays . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 204
6.1. Effects of experimental conditions on the sensitivity of the Langendorff rabbit heart model . . . . . 204
7. In vivo chronic AV-blocked heart models . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 205
8. Conclusions. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 206
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 207

1. Introduction a rare arrhythmia that can lead to ventricular fibrillation and sudden
cardiac death. Since becoming a prominent safety issue with such
The pharmaceutical industry and regulators are under increasing drugs as the antianginal agent prenylamine and the antihistamine
pressure to quickly develop novel, effective, and safe therapeutics. One terfenadine in the late 1980's and early 1990's, at least ten drugs have
vexing problem that plagues drug development is that of avoiding been removed from the market, with warnings added to the product
compounds with a propensity of eliciting Torsades-de-Pointes (TdeP), labels of others due to issues of cardiac proarrhythmia. Such actions
demonstrate the urgent need to avoid this safety hurdle while pro-
ducing novel therapeutic agents.
⁎ Tel.: 847 935 1688; fax: 847 938 5286. TdeP is a rare arrhythmia not readily observed during clinical trials
E-mail address: Gary.Gintant@Abbott.com. or post-marketing surveillance with most drugs. For example, the

0163-7258/$ – see front matter © 2008 Elsevier Inc. All rights reserved.
doi:10.1016/j.pharmthera.2008.04.010
200 G.A. Gintant / Pharmacology & Therapeutics 119 (2008) 199–209

estimated incidence of adverse cardiac events with cisapride is about 1 expression system, myocytes, or (arguably) a wedge of ventricular
in 111,000 prescriptions (Malik & Camm, 2001). Most drugs linked to tissue. As would be expected, more complex assays provide for greater
TdeP are associated with delayed or altered repolarization, manifest numbers of surrogate markers for evaluation (ion channel b myocytes b
clinically as prolongation of the QT interval on the surface ECG. tissue b organ b animal). Multiple surrogate endpoints complicate the
Consequently, regulatory guidances (such as the E14-Guidance, (E-14 evaluation of torsadogenic risk, as weighting factors must be assigned
Guidance for Industry (2005a))) expect a rigorous clinical study to to the different markers in order to provide “risk signatures” that
evaluate the ability of drug candidates to prolong the QT interval in match preclinical and clinical perceptions. The relative clinical risk of
humans (a so-called thorough QT study). Such studies are designed to TdeP is also difficult to discern, due to the rare incidence of TdeP. In
provide robust and precise estimates of QT prolongation with addition, multiple additional factors (for example, drug–drug interac-
therapeutic (and supratherapeutic) drug exposures. These studies are tions that may prominently increase drug concentrations) may
also expensive in terms of resources and additional development time. contribute to the perceived clinical risk independent of direct effects
Despite the key role they have assumed in drug development, it is on myocardium. Finally, different definitions of TdeP are often used
recognized that QT prolongation remains a surrogate marker for with comparing TdeP in preclinical models with the clinical experi-
proarrhythmia. Preclinical and clinical data suggest that there is no ence. For example, a clinical definition of TdeP cited by Napolitano et al.
fixed relation between the extent of QT prolongation and the risk of TdeP (1994) (“progressive twisting of QRS axis around an imaginary base-
(see Belardinelli et al., 2003; Roden, 2004; Shah, 2005a, 2005b, 2005c). line, complete 180 degree twist, and markedly prolonged QT interval in
The purpose of this article is to provide perspectives on the the last sinus beat preceding the onset of arrhythmia” (Drugs)) is very
strengths and limitations of selected preclinical proarrhythmia models different than that cited for ventricular wedge preparations by Shimizu
used to evaluate torsadogenic risk. For comparison purposes, we start and Antzelevitch (1998) (“programmed electrical stimulation-induced
first with a discussion of the simplest in vitro surrogate marker of polymorphic ventricular tachycardia displaying characteristics of
delayed repolarization, namely the hERG current assay. Subsequently, TdeP”(Circ )). Together, these above considerations make quantitative
increasingly complex in vitro models are introduced, including the in comparisons of concentration–response relationships across preclini-
vitro ventricular wedge and Langendorff-perfused rabbit heart cal and clinical studies difficult, and highlight the need for wide safety
preparations, and the in vivo chronic AV-node blocked model. Specific margins derived from preclinical studies.
attention is provided to discerning the utility of quantitative It is generally accepted that TdeP results from the culmination of
(concentration–response) relationships to guide drug discovery and multiple factors acting synergistically to initiate (and sustain) this
avoid TdeP risk. This review is limited to studies of acute electro- arrhythmia. TdeP was initially considered an idiosyncratic drug reac-
physiologic drug effects, and does not consider drugs that may have tion (that is, an adverse drug reaction that does not occur in most
more long-term effects on repolarization, such as those that alter hERG people at doses used clinically), and undoubtedly some cases of drug-
channel trafficking and channel density (see Kuryshev et al., 2005; induced Torsades are likely idiosyncratic (for example, those involving
Dennis et al., 2007). Available data suggests that the more complex genetic mutations affecting cardiac potassium channels). However,
proarrhythmia models are presently most useful to resolve conflicting most cases can be attributed to the contributions of multiple risk
or questionable proarrhythmic signals generated in simpler assays. factors that include a drug (pharmacodynamic) effect (Roden, 1998;
Further benchmarking of proarrhythmic models (required for more Zeltser et al., 2003; DeBruin et al., 2005). Consequently, it would be
precise estimates of model sensitivity and specificity) may support expected that the most useful surrogate marker(s) to interrogate
their role in reducing attrition of advancing compounds and a place in would be those most frequently linked to TdeP and that also play a key
the integrated risk assessment for cardiac safety. role in the initiation of TdeP, with other markers assuming lesser (but
still important) roles. Given that multiple risk factors are recognized to
2. General considerations (and limitations) of play a role in the initiation of TdeP (female gender, bradycardia,
using surrogate markers to evaluate proarrhythmic risk hypokalemia, hypomagnesia, and structural heart disease), it is not
surprising that simple surrogate markers do not provide 100%
It is instructive to consider screening assays for TdeP based on the sensitivity or specificity with regards to predictability of clinical
endpoints acquired with the different models, namely, either surrogate outcome (TdeP). Thus, in order to provide a robust quantitative
(or indirect) markers of TdeP or direct measures of the incidence of (concentration–response relationship) of the TdeP risk, it is imperative
TdeP (see Table 1). Examples of surrogate endpoints used to evaluate to understand 1) the role and limitations of each surrogate marker, and
drug effects range from the deceptively simple IC50 value for hERG 2) the influence (which may be dynamic and nonlinear) of contributing
current block to characterizing changes in the duration and shape of risk factors on the predictability of the surrogate marker under study.
the cardiac action potential (from tissues or multiple cardiac regions) The identification of discrepancies between preclinical “signals” and
to changes in the configuration of T waves from ECG (or ECG-like) clinical findings, while highlighting less than perfect assay sensitivity
recordings. Only more complex models can be used to directly measure or specificity, represents an important first step towards under-
the incidence of TdeP, as one cannot elicit TdeP using a single channel standing the limitations of any surrogate marker.

Table 1
Summary of parameters typically evaluated in preclinical “QT” models

Model Parameters measured or emphasized

Preparation Curr. Repolariz. EADs Triang. Reverse use-dep. Instability/variability Disper. Pseudo-ECG-QT ECGQT QTMorph. TdeP(like)

hERG Channel X
APD Repolarization Tissue APD X X X
QT measures Animals X
Wedge Tissue slice APD X X X X X
Langendorff Organ (Hearts) MAPD X X X X X X X
Chronic AV-block Animals MAPD X X X X X

Models are arranged according to increasing complexity. Measures of the incidence of TdeP (rightmost column) is only one of eleven parameters evaluated in the six selected assays.
Repolariz. = repolarization; EADs = early afterdepolarizations, Triang. = triangulation, QT Morph. = QT morphology (Tpeak–Tend), MAPD = monophasic action potential duration,
Disper. = APD dispersion, ADP = transmembrane action potential, TdeP = Torsades-de-Pointes.
G.A. Gintant / Pharmacology & Therapeutics 119 (2008) 199–209 201

3. hERG assay higher than 4, arrhythmias may be expected. For drugs shown to
appreciably accumulate in myocardium, it may be difficult to translate
IKr is a voltage- and time-dependent outward (repolarizing) hERG block measured in simple CHO or HEK cell expression systems to
current that is activated upon depolarization and plays a prominent electrophysiologic effects in vivo.
role in triggering final repolarization in ventricular tissue (Sanguinetti IC50 values for hERG block are typically compared to predicted (or
et al., 1995). In humans, iKr is encoded by hERG, and is typically actual) clinical Cmax values in order to characterize safety margins for
referred to as hERG current. Numerous studies have demonstrated delayed repolarization. Redfern et al. (2003) have proposed that an
that hERG block may delay repolarization and prolong the QT interval IC50 value for hERG block 30-fold greater than the maximal calculated
on the ECG. For most drugs, block of hERG current results from drug unbound effective therapeutic plasma concentration (ETPCunbound)
binding within the channel pore after gaining access from the cyto- provides a reasonable, balanced compromise for delineating torsado-
plasmic pool (for a review, see Sanguinetti & Tristani-Firouzi, 2006). genic risk and calculating safety margins. These authors noted excep-
Virtually all drugs associated with TdeP block iKr/hERG current tions to this rule (see below for some examples) and also commented
(Roden, 2004). that this margin should be increased for future drug discovery pro-
It is now relatively easy to study drug effects on hERG current using grams and for drugs aimed at non-debilitating diseases. Safety
voltage clamp techniques and either manual patch or automated margins based on IC20 values, while providing for greater sensitivity,
planar patch techniques applied to heterologous expression systems. also are likely more variable due to the reduced slope in that range of
In its simplest form, block is evaluated based on changes in the am- the concentration–response relationship.
plitude of the deactivating tail current measured upon repolarization Not all drugs that block hERG current result in delayed repolariza-
(preceded by a depolarizing square pulse typically applied at slow tion. For example, verapamil is not associated with either QT prolon-
frequencies). Block potency is typically compared based on IC50 values gation or TdeP despite blocking hERG at concentrations comparable to
derived from fitting concentration–response curves; Hill coefficients therapeutic (total) plasma concentrations. This result is most easily
for derived fits and confidence intervals should also be provided. attributed to concomitant block of L-type (inward) calcium current
Testing an accepted standard to benchmark assay sensitivity is essen- mitigating hERG current (Zhang et al., 1999). Fluoxetine and citalopram
tial, as is measuring bath concentrations; the later is crucial to prevent are other examples of HERG blocking drugs expected to reduce hERG
underestimating block potency resulting from reduced bath concen- current at (or moderately above) therapeutic concentrations that do
trations (resulting, for example, from drug absorption to tubing). not typically affect QT prolongation or TdeP clinically (Witchel et al.,
Experimental conditions (for example, temperature) as well as 2002; Fossa et al., 2007). It has been suggested that fluoxetine would
different voltage clamp protocols (different amplitude and duration of not have been developed in the 1970's if the hERG current assay were
depolarizing square pulses, square vs. repolarizing ramp clamp pulses in place (Fermini & Fossa, 2003). In general, these exceptions may be
[to approximate ventricular repolarization]) may affect IC50 determi- attributed (at least in part) to drug effects on non-hERG cardiac
nations. While comparable values IC50 values are attained using such channels that may mitigate or obscure hERG block (Bril et al., 1996, see
varied conditions with most drugs, a few compounds provide sub- also Gintant et al., 2006). Such effects, termed multi-channel block
stantially different values (Kirsch et al., 2004; Yao et al., 2005). More (Martin et al., 2004), are readily apparent when comparing hERG block
recent studies have highlighted the effect of stimulation rate on hERG and delayed repolarization with high content cellular assays or tissue
current block (Stork et al., 2007). Modulation of block by stimulation repolarization studies. Fig. 1 compares the effects of 10 drugs on hERG
rate likely relates to unique kinetics of drug binding and unbinding to current block and prolongation of canine Purkinje fiber action
the channel. These considerations argue for a wide therapeutic win- potential duration. In these experiments, identical concentrations of
dow if calculations are based solely on hERG current measures. each drug were tested in both assays. With drugs such as E-4031 and
For insoluble compounds, the hERG assay is typically conducted
with concentrations to the limit of solubility. The hERG assay should
be conducted in the absence of plasma proteins, since drug binding
within the myocardial cell near the hERG channel is unknown. This
practice also avoids generating parallel datasets of IC50 values in the
absence and presence of plasma proteins. In the case of highly plasma
protein bound compounds, “corrected” IC50 reflecting calculated
(equivalent) total plasma drug concentrations are sometimes pro-
vided. However, it should be recognized that plasma protein binding
varies with species, is not necessarily linear across drug concentra-
tions, and represents only one factor that determines drug distribu-
tion to the myocardium. Controversy exists in regards to “correcting”
for plasma protein binding with in vitro studies. In a hERG study with
a series of five antipsychotic drugs with high plasma protein binding
(83–99%), the ratio of hERG IC50 values to total plasma concentration
corresponded well with QT prolongation, while free (unbound) drug
plasma levels did not provide a reliable correlation (Kongsamut et al.,
2002). In contrast, the ratio of hERG IC50 values to free plasma con-
centrations was predictive of QT prolongation for seven fluoroquino-
lone antibiotics with weak plasma protein binding (range 20–30%,
Fig. 1. A comparison of APD prolongation and hERG current inhibition with ten
Kang et al., 2001). In an in vitro repolarization study using canine
structurally diverse drugs. Plotted are changes in APD obtained from (abscissa) versus
Purkinje fibers, plasma protein binding did reduce APD prolongation hERG current block (ordinate). Matching concentrations of each drug representing
seen with bound drugs, but the extent of the effect was not well therapeutic values and higher multiples (see Martin et al., 2004) were tested in each
correlated with the calculated clinical free fraction (Limberis et al., assay. Nine of 10 drugs demonstrated prominent concentration-dependent hERG block
2007). Some drugs have been shown to accumulate in cardiac tissues; at supratherapeutic concentrations; however, only 4 demonstrated convincing
monotonic concentration-dependent APD prolongation with increasing hERG block
cardiac tissue/plasma ratios have been reported for clozapine, (2.2), (E-4031, erythromycin, moxifloxacin, and telithromycin). Comparable levels of hERG
olanzapine (2.7), sertindole (3.1), risperidone (4.5), and haloperidol block did not predict the extent of APD prolongation. Values represent means of a
(6.4) (Titier et al., 2004). These authors suggested that with ratios minimum of 6 determinations per drug for each assay.
202 G.A. Gintant / Pharmacology & Therapeutics 119 (2008) 199–209

moxifloxacin, there is a clear increase in APD with increased hERG erogeneity in the in situ heart compared to wedge preparations (see
current block. We call these drugs overt hERG blockers. In contrast, Anyukhovsky et al., 1999; Taggart et al., 2003; Ueda et al., 2004, also
increasing hERG block is not associated with increasing APD prolonga- Conrath & Opthof, 2006 for a review). Changes in the configuration of
tion for such drugs as haloperidol and fluoxetine. We call these drugs the “T waves” from pseudo-ECG recordings (measured as the interval
covert hERG blocking agents. Cisapride represents a special case in that between the peak and end of the T wave [Tpeak–Tend]) have been
increasing drug concentrations initially prolong and then shorten the proposed as an index of transmural dispersion of repolarization in this
APD (a bell-shaped concentration–response curve). This effect is likely model (Yan & Antzelevitch, 1998; Antzelevitch & Shimizu, 2002).
due to block of L-type calcium current only at higher drug concentra- However, the validity of this marker has been questioned, at least
tions. Such studies emphasize that hERG block represents only one when applied to in situ preparations (Opthof et al., 2007). In the canine
possible ion channel (off-target) effect. Clearly, there is a need to wedge, transmural dispersion of repolarization is sensitive to the
mechanistically define the level of torsadogenic risk with covert hERG stimulation site, with dispersion increasing with epicardial vs. endo-
blocking agents. As hERG is presently positioned in lead optimization to cardial stimulation (Di Diego et al., 2003; Circ.). This effect modifies the
differentiate (and eliminate) compounds early in drug discovery, the sensitivity of preparations to a Torsades-like arrhythmia with low doses
number of compounds wrongly excluded based solely on the early of cisapride, and needs to be considered when evaluating results with
application of hERG screening assays is difficult to determine. this model.
Given results from a well-conducted hERG study one would expect In a blinded validation of the isolated arterially-perfused rabbit
it possible to estimate concentration-dependent effects on QTc ventricular wedge preparation, Liu and colleagues (2006) compared the
clinically (and hence, therapeutic margins for delayed repolarization) effects of 13 (8 positive, 5 negative controls) drugs with varying pro-
based upon comparisons between IC50 values for block and therapeutic pensity for causing QT prolongation or TdeP. Electrophysiologic effects
drug concentrations. For some specific hERG blocking drugs this may were studied over a wide range of drug concentrations including those
be the case. Dofetilide is a drug used to treat atrial arrhythmias that is corresponding to calculated free clinical therapeutic plasma concentra-
also a potent iKr blocker (IC50 values approximately 10–20 ng/mL) and tions (see Fig. 2 from Liu et al., below). These investigators were also
that delays ventricular repolarization. A modeling study comparing careful to validate bath concentrations to ensure drug exposures, a factor
dofetilide's effect on hERG current with results from four recent clinical not considered in similar studies. Relative TdP risk assessment was based
thorough QT studies demonstrates striking sensitivity of QT prolonga- on a scoring system evaluating QT interval prolongation, Tpeak–end/QT
tion to hERG current block: 7% block of hERG current affects 5 msec ms ratio, and development of early afterdepolarizations with and without a
mean prolongation of QT, and 13% block affecting 20 ms prolongation closely coupled extrasystole; the development of EAD-induced extra-
(Jonker et al., 2005). This exquisite sensitivity may be attributed (at systoles received the greatest weight in calculation of TdeP risk scores.
least in part) to specificity of iKr block by dofetilide. In contrast, the QT The authors reported a high sensitivity and specificity for identifying
prolonging response with cisapride is not as sensitive as that of drugs with a potential for causing TdP. As shown in Fig. 2 (below), all
dofetilide (Wallis, 2008), likely due to multi-channel block. Obviously, five “negative” compounds maintain low TdeP risk scores throughout the
one must also consider the likelihood of additional off-target drug 0.1–100× range of free therapeutic plasma Cmax concentrations.
effects in vivo that may indirectly affect cardiac repolarization (for Liu et al. did note differences in the TdeP risk scores for the 8
example, extracardiac alpha1-adrenergic stimulation, see Farkas et al., “offender” compounds associated with either QT prolongation or TdeP
2006). in humans, especially near concentrations 10-fold free therapeutic
plasma Cmax values. For example, in the approximate range of 7–9-
4. In vitro arterially-perfused left ventricular wedge preparations fold free therapeutic plasma Cmax concentrations, the composite risk
score ranking was terfenadine b moxifloxacin b sparfloxacin; moxiflox-
A more complex preclinical assay used to interrogate delayed acin demonstrated greater TdeP risk scores than did terfenadine over
repolarization and torsadogenic risk is the arterially-perfused ventricular the entire 0.1–100+-fold range of free therapeutic plasma Cmax
wedge preparation (Wu et al., 2005; Yan et al., 1998a, 1998b; 2001). This concentration multiples. It is debatable as to whether this ranking
model has proven useful in delineating mechanisms of proarrhythmia, reflects the intrinsic ability of these drugs to elicit TdeP clinically
including spatial and temporal dispersion of repolarization, drug-induced
enhancement of repolarization heterogeneity (leading to transmural
dispersion), and triggered activity (early afterdepolarizations). Electro-
physiological recordings (obtained with loosely-held (“floating”) micro-
electrodes) allow for comparisons of action potentials recorded from
different myocardial layers (epi-, mid-, and endocardial regions) as well
as recordings of pseudo-ECG (derived either from either microelectrode
recordings or extracellular field electrodes for transmural signals). While
TdeP-like recordings have been published from these wedge prepara-
tions, typical surrogate markers include: delayed repolarization (QT or
APD prolongation), dispersion of repolarization (Tpeak–end/QT ratio or
TDR), and incidence of EADs. The rabbit wedge preparation is claimed
advantageous compared to the Langendorff rabbit heart (see below) in
regards to electrical stability (more than 4 h for wedge compared to lesser
times for the Langendorff perfused rabbit heart), allowing for four 40 min
drug equilibration periods (Chen et al., 2006). One challenge of this model
(or any model evaluating multiple endpoints) is to correctly align the
measured parameters to optimize sensitivity and specificity when
defining proarrhythmic risk.
With wedge preparations, particular attention is often focused on
greater drug-induced prolongation within the ventricular myocardium Fig. 2. Effect of thirteen compounds in a rabbit ventricular wedge preparation. Eight
compounds chosen on the putative basis of ability to prolong QTc and/or induce TdeP in
(“M cells”, see for example Yan et al., 1998; Di Diego et al., 2003) that humans (closed symbols) increase TdP risk score, while all five drugs associated with
is exaggerated at excessively slow stimulation rates. It should be noted, cardiac safety in clinical usage (open symbols) do not affect the TdP risk score. From Liu
however, that some reports find lesser evidence of repolarization het- et al. (2006).
G.A. Gintant / Pharmacology & Therapeutics 119 (2008) 199–209 203

based solely on direct cardiac electrophysiologic activity (pharmaco- Moxifloxacin and cisapride are the only two compounds tested by
dynamic factors), or whether the clinical experience is biased by the both Lu et al. and Chen et al. in wedge preparations. The Chen study
possible range of high exposures achieved clinically with both com- differentiates moxifloxacin as having higher proarrhythmic risk
pounds (pharmacokinetic factors). compared to cisapride over the entire range of Cmax multiples tested.
For each of the five tested drugs recognized clinically for cardiac In contrast, the Liu study ranks the two drugs equal at multiples up to
safety (verapamil, clozapine, captopril, fexofenadine, and desipra- 9-fold Cmax; above this multiple, risk with cisapride increase 5-fold,
mine), there was a clear lack of TdeP risk (open symbols, TdP risk while scores with moxifloxacin remain essentially constant. It should
scores consistently below zero at up to 100× the free therapeutic also be noted that cisapride produced EAD's in the rabbit model at
plasma Cmax). Interestingly, of the 5 “safe” compounds tested, only concentrations near 100 nM, while no EAD's were observed at higher
fexofenadine had a hERG ratio (defined as hERG IC50/free therapeutic concentrations (5 μM) in the dog wedge preparation (Di Diego et al.,
plasma concentration and calculated based on values from Table 2 of 2003). These comparisons demonstrate the possible variability across
Liu et al.) greater than 30 (respective values of 20, 5, and 1.75 are laboratories and species when attempting to quantify proarrhythmic
calculated for desipramine, clozapine, and verapamil). Despite these risk, and highlights the need for benchmarking of each specific model
low margins, all five did not demonstrate enhanced TdeP risk scores. with known standards to demonstrate assay sensitivity.
These results suggest that this model provides evidence of safety of
hERG blocking drugs with relatively low hERG safety margins. 5. In vitro Langendorff perfused isolated heart preparations
In a separate study also published in 2006, Chen and colleagues
independently validated the arterially-perfused rabbit left ventricular The Langendorff perfused isolated rabbit heart is perhaps the most
wedge model and examined its use in predicting proarrhythmic widely used in vitro preparation used to evaluate drug-induced
risk. This work described results obtained with a set of six drugs delayed repolarization and proarrhythmia. This model extends studies
(dofetilide, DL-sotalol, cisapride, risperidone, and moxifloxacin as posi- pioneered by Carlson et al. with the in vivo methoxamine-sensitized
tive controls, fluoxetine as a negative control), along with a fairly detailed rabbit heart proarrhythmia assay used to assess proarrhythmic effects
methodological description of the preparation. In this study, the scoring of Class III antiarrhythmic agents (Carlsson et al., 1993). Developed on
scheme was based on QT interval prolongation, Tpeak–Tend, and the an automated platform by Hondeghem, the SCREENIT Langendorff
appearance of phase 2 EAD's (comparable to a scheme employed by Liu assay measures ventricular monophasic action potential recordings
et al., 2006, above). Overall, this model could distinguish between the from endocardial and epicardial surfaces (as well as pseudo-ECG's)
negative and positive control compounds across a minimum 30-fold obtained from methoxamine-pretreated female rabbit hearts subject
range of concentrations encompassing calculated free Cmax values to escalating drug concentrations (see Hondeghem et al., 2001;
(see Fig. 3, below). Concentration-dependent increases in arrhyth- Hondeghem & Hoffmann, 2003 and earlier references contained
mogenic scores was noted for each of the five positive controls. therein). Dispersion of repolarization (measured as difference in
Dofetilide, DL-sotalol, cisapride, risperidone and moxifloxacin in- monophasic action potential duration recorded from ventricular
creased endo- and epicardial APD90, QT interval and TP-E (peak-to endocardial and epicardial surface) may also be measured as an
end time of the T wave) in a reverse use-dependent manner within index of proarrhythmia. Complex pacing protocols (encompassing
clinically relevant concentration ranges. Phase 2 early afterdepolar- rapid and slow stimulation periods) are also employed, and transient
izations (EADs) were observed at 1.6, 2.3, 16.7, 37.5 and 7.9 fold, changes in APD in response to rate changes are monitored. Such
respectively, their corresponding unbound therapeutic concentra- measures provide for an evaluation of repolarization instability,
tions. Based on multiples of free Cmax values, ranking of TdeP risk was defined as difference between upper and lower quartile estimates of
sotalol = dofetilide N moxifloxacin Nrisperidone = cisapride. Somewhat APD values of MAP recordings from a sequence of 60 action potentials.
surprisingly, at approximately four-fold free Cmax concentrations, Instability is one of the components of “TRIaD”, an acronym used to
moxifloxacin demonstrated a 2-fold greater proarrhythmic score com- describe multiple drug-induced electrophysiologic effects; T refers to
pared to cisapride (though both fall below the indicated “threshold” triangulation of the action potential configuration, R refers to reverse
that would indicate a high risk for proarrhythmia). use-dependence, I refers to instability of repolarization and D refers to
dispersion of repolarization (added in later versions of SCREENIT). Each
of the elements of TRIaD have been implicated in the genesis of
proarrhythmia (Shah & Hongehem, 2005, see also Millberg et al.,
2004), and may act synergistically. As with other assays utilizing
multiple parameters as endpoints, it was unclear as to how to
optimally rank factors towards a composite score when assessing
proarrhythmic risk.
In an early blinded validation of this assay using 14 drugs (8 asso-
ciated with QT prolongation clinically, 3 linked to QT shortening, the
remaining 3 with no QT duration effects), SCREENIT identified
potentially proarrhythmic drugs at selected concentrations based on
measures of repolarization instability, triangulation, and/or reverse
use-dependence (Hondeghem & Hoffmann, 2003); none of the drugs
considered as safe were labeled as proarrhythmic. In some cases,
however, concentrations necessary to identify proarrhythmic risk
greatly exceeded therapeutic concentrations. For example, terfena-
dine demonstrated effects only at high free drug concentrations
Fig. 3. Arrhythmogenic scores of drugs in relation to their clinically relevant (calculated
free) concentrations in a rabbit ventricular wedge preparation. Drugs tested included (from (5 mcg/mL), much higher than those encountered clinically either in
left to right);DL-sotalol (squares), dofetilide (circles), moxifloxacin (left triangles), cisapride the absence or presence of metabolic inhibition in humans (approx
(up triangles) = risperidone (down triangles), and fluoxetine (diamonds). The dashed box 13 ng/mL total [0.4 ng/mL calculated free concentration], associated
indicated threshold for alarm that was obtained by averaging the scores of preparations with a mean QTc prolongation of 42.4 msecms, Abernethy et al., 2001).
demonstrating EADs; the lower line reflects the greater sensitivity of cisapride to initiate
EADs (observed in 2 of 4 preparations with low (2%) QT prolongation). It should also be
In a separate validation study, 55 blinded compounds (grouped
noted that near the threshold scoring point substantial increases in average QTc values according to the five torsadogenic categories following the outline of
(89%) and Tpeak–end (211%) were observed. Adopted from Chen et al. (2006). Redfern et al., 2003) were subjected to a rigorous quantitative analysis
204 G.A. Gintant / Pharmacology & Therapeutics 119 (2008) 199–209

using SCREENIT to evaluate it's predictive value for TdeP liability assays (hERG current, Purkinje fiber repolarization, left ventricular
(Lawrence et al., 2006). Compounds were evaluated using four or five wedge and arterially-perfused Langendorff heart proarrhythmia
escalating concentrations spaced at either full or half-log concentra- models) was recently published by Lu et al. (2007); with the exception
tions at multiples of 1–300 the highest effective free therapeutic of the hERG assay, preparations studied were all derived from the
clinical plasma concentration (EFTPCmax). Of eight drugs generally rabbit. The antibiotics were tested at concentrations based on total
associated with high TdeP risk (Category 1), two false negative therapeutic Cmax concentrations, and included sparfloxacin (removed
compounds (azimilide and disopyramide) were detected. Of 21 drugs from the market for TdeP). Different rankings were found for the
generally considered free of TdeP risk, 3 false positives (moxifloxacin, antibiotics dependent on the assay employed (see Fig. 4, below). For
vardenafil, and risperidone) were determined. For drugs comprising example, moxifloxacin and erythromycin had comparable rankings in
the middle three categories (representing graded levels of putative the hERG assay, while erythromycin demonstrated lowest score and
proarrhythmic risk), the model was described as “less precise”. moxifloxacin the highest score in the Langendorff-perfused heart. The
Within this Lawrence study, a subset analysis was performed for authors suggested that the hERG current, Purkinje fiber, and isolated
seven drugs that spanned the five TdeP risk categories described by Langendorff-perfused rabbit hearts could be used to assess the ability
Redfern yet demonstrated an approximate 10-fold hERG IC50/ of antibiotic compounds to delay ventricular repolarization, while the
EFTPCmax ratio. Using hERG potency as a filter, a clear separation wedge preparation appeared more predictive and suitable for detect-
between Category 1 (procainamide, most risk example) and Category 5 ing torsadogenic effects of antibiotics. These conclusions are critically
(ketoconazole, least risk example) risk scores was apparent across the dependent on the correct ranking of clinical (pharmacodynamic) risk.
1–100 multiple range of EFTPCmax. In contrast, no clear separation Lu et al. also demonstrated that grepafloxacin elicited QT prolongation
between drugs in the intermediate risk categories (2, 3, and 4) was in the arterially-perfused rabbit left ventricular wedge model, but
apparent; domperidone (Category 4) had a higher proarrhythmic score failed to do so in the Langendorff-perfused heart, emphasizing the
than grepafloxacin or sertindole (Categories 2 and 3) at a 10-fold need to benchmark assay sensitivity across different models.
multiple of EFTPCmax. In conclusion, this validation study by Lawrence
et al. demonstrated that with drugs taken as groups, the Langendorff 6.1. Effects of experimental conditions
SCREENIT model could predict clinical outcome, particularly for drugs on the sensitivity of the Langendorff rabbit heart model
associated with either very high or very low torsadogenic risk.
However, the model was less precise in predicting risk for drugs at An understanding of the sensitivity of any biological assay is
other than the extreme margins of perceived TdeP liabilities. Following essential to understanding the utility of that model. This concept is
the lead of Redfern et al., the authors proposed that a 30 fold margin exemplified in two studies utilizing the in vitro female Langendorff
between risk scores and EFTPCmax would provide confidence that a rabbit heart model in the absence and presence of the sea anemone
new chemical entity should proceed through development without toxin (ATX-II, an agent that increases cardiac late sodium current). In
incurring substantial risk of eliminating potentially beneficial drugs. the first study, the proarrhythmic effects of moxifloxacin were
A recent study by Steidl-Nichols et al. (2008) provided further evaluated (Wu et al., 2006). ATX-II alone caused neither EAD's nor
insights into the utility and limitations of the Langendorff perfused VT, while ATX-II (3 nM) and moxifloxacin (400 ng/mL(1 μM)) together
rabbit heart proarrhythmia model. In their investigations, a blinded set caused spontaneous and pause-triggered EAD's, ectopic ventricular
of nine compounds of varying proarrhythmic risk (along with a beats, and polymorphic VT in each of seven hearts studied. This
negative control) was independently evaluated in the SCREENIT model moxifloxacin concentration is considered subtherapeutic and falls far
using a series of experimental protocols that explored a), effects below the range of typical Cmax values (4 mcg/mL) measured during
measured across overlapping concentration ranges, b) increased “safe” thorough QT studies or following oral administration of a 400 mg
number of concentration ranges, c) extended perfusion times, and d) therapeutic dose (Moxifloxacin product label) and is considered
increasing the number of experiments. Proarrhythmia scores were subtherapeutic. In the second study with the Langendorff rabbit
derived based on those described by Lawrence et al. (2006). While the heart model, Wu et al. (2008) demonstrated that acutely administered
model correctly identified proarrhythmic agents qualitatively, the escalating doses of a wide range of amiodarone concentrations (1 nM–
study design was shown to significantly impact the proarrhythmic risk 10 μM) caused an insignificant increase in the monophasic action
assessment. Of note was the finding that the concentration at which
action potential prolongation, triangulation, instability, reverse use-
dependence and ectopic beats occurred varied based on the over-
lapping concentration ranges used in the different protocols. This
result, which is likely due to inadequate equilibration time with cardiac
tissue, demonstrated the critical role of study protocol in defining
preclinical safety margins in this model. It should also be noted that
some limitations demonstrated by Steidl-Nichols et al. were previously
recognized by Hondeghem as model limitations (for example, duration
of equilibration period for steady-state effect and use of predefined,
standardized concentrations, (Hondeghem & Hoffmann, 2003). Col-
lectively, the above data suggests that a strength of the Langendorff
perfused rabbit heart assay lies in delineating proarrhythmic actions of
drugs in a more complex, integrated system (though not necessarily in
providing precise concentration–response relationships to define
proarrhythmic risk), and to aid in demonstrating (or refuting) potential
proarrhythmic effects of compounds showing prominent effects in
simpler assays (i.e., potent hERG current block).
Fig. 4. Summary of rank order of proarrhythmic scores of four antibiotics in four different
6. A comparison of in vitro proarrhythmia assays preclinical assays. Test concentrations of each antibiotic were chosen based on
therapeutic Cmax plasma concentrations. For drugs with approximately equal scores,
drug abbreviations were placed between rank levels (1 least liability, 4 greatest liability).
A study directly comparing the electrophysiologic effects of four Spar = sparfloxacin, Moxi = moxifloxacin, Ery = erythromycin, Teli = telithromycin. Data
antibiotics (two fluoroquinolones and two macrolides) in four in vitro derived from Lu et al. (2007), (with clinical rankings based on Torsades.org).
G.A. Gintant / Pharmacology & Therapeutics 119 (2008) 199–209 205

potential duration and did not elicit TdeP. In contrast, in the presence of sen et al., 2003; van Opstal et al., 2001c); ectopic beats were observed
3 nM ATX-II, low amiodarone concentrations (1–30 nM) increased in the high dose, as was a tendency for increased interventricular
transmural dispersion of repolarization, beat-to-beat variability, and dispersion. Three interventions to promote repolarization (infusion of
prolonged MAP duration. Higher concentration ranges (30–300 nM KCl, pharmacologic activation of iKATP with levcromakalim, and
(0.02–0.2 mcg/mL)) induced TdP in 16/17 hearts, while clearly steady ventricular pacing (60 bpm)) abolished TdeP without reducing
excessive concentrations (1–10 μM) abbreviated APD, decreased drug-induced QT prolongation. Thus, QT prolongation does not always
beat-to-beat variability, and suppressed TdeP. It should be noted that correlate with the risk of TdeP in this diseased-based model.
the higher concentrations tested (0.02–0.2 mcg/mL) represent values Studies with the anesthetized CAVB dog model (albeit with a
12-fold lower than the high therapeutic concentration range for somewhat limited number of drugs and drug concentrations) have
amiodarone (1–2.5 mcg/mL, Jurgens et al., 2003); the demonstrated demonstrated proarrhythmia consistent with the clinical perception of
excessive sensitivity is even greater if one considers plasma protein TdeP risk. Thomsen et al. (2006) compared QT prolongation and the
binding of amiodarone (approx 95%, Jurgens et al., 2003). Amiodarone incidence of TdeP with two doses of either moxifloxacin or azithro-
is a drug that is recognized as prolonging QT interval but essentially mycin administered i.v. using a randomized cross-over experimental
devoid of TdeP liability in man (Lazzara, 1989; Auer et al., 2002; Singh, design (n = 5–6 dogs); dofetilide was used as a positive control. Mea-
2006) and following acute administration in the chronic AV-blocked sured total plasma concentrations of moxifloxacin and azithromycin
dog model (van Opstal et al., 2001a; Thomsen et al., 2004). These in were approximately 1–6 fold and 7–30 fold the expected mean clinical
vitro studies emphasize the importance of experimental conditions in Cmax concentrations (low and high dose infusions, respectively).
determining assay sensitivity and possibly defining false-positive Results demonstrated greater prolongation of QTc with dofetilide
compounds when evaluating new chemical entities. (29%) compared to b) low and high exposures of moxifloxacin (7 and
21% prolongation); azithromycin demonstrated essentially no QTc
7. In vivo chronic AV-blocked heart models prolongation. While dofetilide and moxifloxacin elicited QT prolonga-
tion, dofetilide elicited TdeP in each of five dogs studied, while no TdeP
Perhaps the most extensively studied in vivo large animal was observed with either dose of moxifloxacin (consistent with the
proarrhythmia model is that of the chronic atrio-ventricular blocked perceived clinical risk). The CAVB model has also been used to
(CAVB) dog. In this model, the AV node is ablated, and animals demonstrate minimal torsadogenic risk with AVE-0118, a novel atrial-
(typically dogs or rabbits) are studied after sufficient time is allowed active drug (Oros et al., 2006); despite effects on atrial repolarization,
for bradycardia-induced electrophysiologic ventricular remodeling this agent does not affect ventricular repolarization and does not
leading to decreased repolarization reserve (Volders et al., 1998, 1999; induce TdeP.
Van Opstal et al., 2001b, Tsuji et al., 2002; Gross, 2006). Conscious or More recent studies with the anesthetized CAVB dog model have
anesthetized animals may be studied, with more invasive instrumen- suggested that beat-to-beat variability of repolarization (BVR), a
tation possible with anesthetized animals. Parameters evaluated in measure related to temporal dispersion of repolarization, is a novel
this model can include QT prolongation, ectopic beats, and mono- surrogate marker of proarrhythmia. BVR can be explored graphically
phasic action potentials typically recorded with catheters positioned using Poincare diagrams that plot consecutive pairs of x,y values of
on the left and/or right ventricular endocardium. Early afterdepolar- parameters (typically APD values) from the nth and nth + 1 beat (Fig. 5,
izations (EADs) can be recorded from monophasic action potentials, above). As used previously with MAP recordings from Langendorff
and interventricular dispersion calculated by comparing MAPs from perfused hearts (Hondeghem et al., 2001), the polygon formed by
left and right ventricles. Additional stressors that have been applied consecutive x–y points widens around the line of identity with greater
include female gender, hypokalemia (as with furosemide), high drug beat-to-beat variability. BVR can be characterized based on measures
concentrations (by IV or oral administration), catecholamine chal- of short term variability (STV), representing the width of the Poincare
lenge (epinephrine), and additional potassium channel blockers to diagram from the line of identity (see Thomsen et al., 2004; van der
reduce repolarization reserve (for example, iKs blockade). Being an Linde et al., 2005 and references therein).
in vivo model, potential additional off-target drug effects (heart rate, In anesthetized CAVB dogs, MAP measures of left ventricular STV
blood pressure changes) that may contribute to proarrhythmia may be are preferred to quantify repolarization liability, as MAP measures of
manifest; while off-target effects may render mechanistic interpreta- right ventricular STV are a poor predictor of drug-induced TdeP
tions more difficult, such effects may provide a more realistic (Thomsen et al., 2004, 2005). The increase in baseline STV in the
assessment of proarrhythmic risk. anesthetized, drug-free CAVB dog model has been attributed to long-
The CAVB model has been useful in exploring mechanisms of TdeP, term electrical remodeling and blunted beta-adrenergic receptor
defining novel surrogate markers, as well as providing direct measures signaling (Stengl et al., 2006), and not to changes in heart rate or
of the incidence of TdeP. Dose–response relationships are not typically average measures of variability in RR interval (Thomsen et al., 2007).
studied due (at least in part) to the time required for achieving steady- However, in remodeled hearts, the additional contribution of heart rate
state plasma (or tissue) concentrations in vivo with anesthetized, variability to drug-induced increases in STV (and TdeP) is uncertain. It
instrumented animals. With this model, drug-induced QT prolonga- should be noted that CAVB dogs can be divided into two phenotypes;
tion has been observed without TdeP. For example, despite QT those susceptible and resistant to TdeP with infusion of dofetilide
lengthening with chronic amiodarone (21%) and augmented inter- (Thomsen et al., 2003; van Opstal et al., 2001c). As proarrhythmia
ventricular dispersion (difference between left and right endocardial resistance may be related to the extent of ventricular remodeling, dogs
MAP recordings, 3 of 7 anesthetized dogs) no TdeP was observed must be prescreened to reduce baseline variability when evaluating
(consistent with clinical experience) (van Opstal et al., 2001a). The proarrhythmic risks of novel compounds.
absence of TdeP with amiodarone (despite QT prolongation) was Studies with D-sotalol in the anesthetized CAVB model demonstrated
attributed to homogeneous lengthening of APD in most animals as that increases in STV (but not QT prolongation) were predictive of drug-
well as the lack of EAD's and ventricular ectopy. In contrast, the induced TdeP (see Fig. 5A and B, above; Thomsen et al., 2004). A recent
noniodinated congener dronedarone elicited QT prolongation (31%), paper by Oosterhoff et al. (2007) summarized studies with the CAVB
increased interventricular dispersion in 7 of 8 dogs, and elicited TdeP model from the Vos lab regarding the utility of increased BVR (based
in 4 of 8 dogs with the highest interventricular dispersion. Subsequent on endocardial MAP recordings) in screening for drug-induced TdeP.
studies with the antipsychotic drug sertindole in CAVB anesthetized A graphic summary of these studies (derived from reviewed data) is
dogs demonstrated no TdeP with a low dose (but not high dose) of i.v. shown in Fig. 5C. More prominent increases in STV (abscissa) with
administered sertindole despite comparable increases in QTc (Thom- dofetilide, NS-7,D-sotalol are associated with increasing incidence of
206 G.A. Gintant / Pharmacology & Therapeutics 119 (2008) 199–209

Fig. 5. A. Representative Poincare plots of left ventricular monophasic action potential duration recorded from anesthetized CAVB dog under control conditions (open arrows)
and during infusion ofD-sotalol (2 mg/kg, left, 4 mg/kg right). Closed arrows indicate time of first extrasystole. B. Changes in short term variability (shown in A) plotted on time
axis. Whereas no TdeP was observed at the 2 mg/kg dose, TdeP was observed with the 4 mg/kg dose at 19 min after start ofD-sotalol infusion. Results obtained from same dog
(adopted from Thomsen et al. Circ, 2004). C. Summary of effects of six compounds on short term variability (STV) and incidence of Torsades (TdeP). For drugs that do not elicit
TdeP, STV does not increase (amiodarone, moxifloxacin, azithromycin). In contrast, STV increases withD-sotalol, dofetilide, and NS-7 are associated with increased incidence of
TdeP. However, the absolute STV value does not correlate with the TdeP incidence (74% incidence of TdeP with dofetilide associated with a relatively low STV (4.3), vs. 25%
incidence of TdeP with D-sotalol associated with a relatively high (5.5) msec ms STV value. STV measured from LV MAPsMAP's in anesthetized dogs. Panel C derived from review
data provided by Oosterhoff et al. (2007).

TdeP (ordinate), while smaller STV increases elicited with amiodarone, model are essential to evaluate it's sensitivity and specificity with
moxifloxacin, and azithromycin did not result in TdeP. In most cases, therapeutic and supratherapeutic drug concentrations.
TdeP was observed only when statistically significant increases were Finally, the extent to which STV measures are predictive of TdeP
observed. However, absolute STV values were not good predictors of may also depend on the model under study. A recent study with
TdeP incidence; D-sotalol (5 ms increase in STV) affected a 25% inci- anesthetized, open-chest, phenylephrine-stimulated “normal” rabbits
dence of TdeP, while dofetilide (4 msec ms increase in STV) affected demonstrated that TdeP elicited by a variety of single or combination
a 74% TdeP incidence. Thus, STV qualitatively (but not necessarily agents (E-4031, HMR1556, ATX-II) was not preceded by an increase in
quantitatively) demonstrated a strong predictive value in the screening STV values derived from left ventricular epicardial MAP's (Michael et
for proarrhythmia. al., 2007). These results contrast with those of the anesthetized CAVB
It is recognized that the above summary spans a period of multiple dog model (where STV increases measured from left ventricular
years; a blinded, prospective comparative study with select drugs endocardial MAP's predict TdeP); it is uncertain whether these
would be necessary to rigorously assess the precision of STV measures disparate results relate to species or experimental details. The value
in predicting TdeP with multiple compounds. of increased STV measures derived from QT intervals (rather than MAP
Collectively, the above studies suggest that the CAVB dog model recordings) require further study; a recent pilot clinical study suggests
represents a stringent (yet not overly sensitive) model for predicting risk that in the absence of QTc prolongation, baseline BVR (based on QT
of TdeP that does not rely on measures of QT prolongation. The utility of interval measures) is of potential value for identifying patients at risk
this model is likely due to electrical remodeling reducing repolarization for proarrhythmia with QT prolonging drugs (Hinterseer et al., 2008);
reserve to a level comparable to that found in humans, as well as a block of multiple cardiac potassium channels has been shown to
comparable range of heart rates to those of humans. Additional drug increase short term QT interval variability and provoke TdeP in
studies with this model are limited by the low throughput, expense, and conscious dogs (Lengyel et al., 2007).
technical expertise required to successfully complete such studies. On
the issue of in vivo model dependency, Chiba et al. (2004a,b) compared 8. Conclusions
the sensitivity of surrogate markers of drug-induced TdeP in halothane-
anesthetized dogs without chronic AV block. Using sparfloxacin (a non- In 1997, the Points to Consider Document suggested additional
specific IKr channel blocker that elicits TdeP in experimental and clinical preclinical in vitro repolarization studies to assess the proarrhythmic
studies), they demonstrated prolongation of the QT and QTc intervals, risk of TdeP (CPMP, 1997). Specifically, this document suggested
effective refractory period, and endocardial MAP90 values from both cellular electrophysiologic investigations to evaluate the potential to
ventricles. In spite of significant prolongation of the QT interval and MAP evaluated delayed ventricular repolarization of drugs based on action
duration, no early afterdepolarizations, ectopic beats, or dispersion of potentials and repolarization abnormalities such (as early after-
ventricular repolarization (measured as interventricular dispersion of depolarizations). Since that time, further progress has been made
endocardial MAPs) was observed; delayed repolarization of phase 3 elucidating the mechanisms responsible for the initiation (and
repolarization (without triangulation) was observed. This study suggests perpetuation) of TdeP. Such studies have focused on the “permissive”
that the sensitivity of surrogate markers may vary dramatically for role of hERG block (the most prevalent cause of delayed repolariza-
bradycardia-remodeled and “non-remodeled” normal hearts. An in vivo tion), demonstrated that multi-channel block may affect repolariza-
rabbit heart failure model to characterize torsadogenic risk has recently tion and effectively modulate the untoward effects expected of hERG
been described (Kijtawornrat et al., 2006); further studies with this block alone, and provided an appreciation of the role of repolarization
G.A. Gintant / Pharmacology & Therapeutics 119 (2008) 199–209 207

heterogeneity (arising from spatial and temporal dispersion) in setting interactions that may result in excessive drug concentrations and
the stage for TdeP. Additional models established to evaluate corresponding electrophysiologic effects). Thus, the perceived risk of
proarrhythmic risk include more complex preparations such as the TdeP may not be reflected in the risk signature of compounds mea-
in vitro ventricular wedge and Langendorff heart preparations, and in sured directly in a complex proarrhythmia model.
vivo chronic AV-node block models. One measured parameter shared Based on the studies reviewed above, the more complex proar-
between these more popular models is that of cellular repolarization rhythmia models are presently most useful for qualitatively clarifying
(evaluated either using transmembrane or monophasic action the torsadogenic risk of drugs, especially those linked to proarrhyth-
potentials). From action potentials, measures of spatial dispersion of mia based on simpler surrogate markers. This approach is exemplified
repolarization are derived by comparing repolarization recorded from by published in vitro studies (with rabbit ventricular wedge prepara-
multiple cardiac regions, while measures of temporal dispersion are tions (demonstrating low TdP risk scores for compounds with narrow
provided by following dynamic changes in action potentials evoked hERG margins) and in vivo studies with the anesthetized CAVB dog
with irregular rhythms or pacing protocols. A second measured model (evaluating the effects of amiodarone, van Opstal et al., 2001a;
parameter shared between these models is that of either standard or selected antibiotics, Thomsen et al., 2006; and dose-dependent effects
“pseudo” ECG recordings to evaluate QT prolongation. Simple changes of sertindole, Thomsen et al., 2003; van Opstal et al., 2001c). Indeed,
in the morphology of the QT interval may provide an index of spatial one might consider proarrhythmia models in the category of “follow-
changes in repolarization heterogeneity. Together, these markers (in up” studies as part of the integrated risk assessment in the ICH S7B
various formats) provide a more comprehensive “risk signature” to guidelines (Guidance for Industry, 2005b, S7B). Proarrhythmia models
define the potential of drug-induced TdeP. could prove useful in reducing attrition of drugs based primarily on
It is appreciated that defects in cardiac channels for iKr (hERG), IKs, hERG assay results, as well as clarifying “weak signals” obtained from
and INa account for the majority of cases of congenital long QT extremely precise (and expensive) thorough clinical QT studies.
syndromes (LQT2, LQT1, and LQT3 respectively) and congenital However, clarification of both the sensitivity and specificity of the
proarrhythmia. Similarly, drug effects on these three channels (as preclinical models must be provided in support of this role. PK/PD
well as the inward rectifier iK1) can lead to acquired long QT comparisons drawn from preclinical and early clinical studies (see for
syndromes and proarrhythmia. While most drugs that delay repolar- example, Garnett et al., 2008) will also be necessary to provide the
ization block hERG block, and potency of block can be measured with necessary contextual link and guide the common interpretation of
great precision, hERG block is not always predictive of either delayed study results.
repolarization or torsadogenic risk. This may be due (at least in part) to A drug classification system based on drug effects on surrogate
the fact that Torsades results from “multiple hits” from recognized risk markers would be useful in providing a more global perspective of
factors superimposed on an altered or diseased electrophysiologic safety profiles and evolving risk signatures. This system could
substrate (i.e., cardiac hypertrophy, bradycardia-induced electrical categorize simpler concentration-dependent effects on multiple
remodeling) and dynamic conditions (such as the short–long–short cardiac ion channels as well as more complex parameters derived
rhythm sequence) that may act synergistically to initiate TdeP. Based from increasingly integrated assay systems (including cellular repo-
on this complex interplay of factors leading to TdeP, it is unrealistic to larization changes (action potentials, spatial and temporal dispersion)
expect that any simple single assay or surrogate marker would and global (QT) repolarization changes (such as Tpeak–Tend).
provide either 100% sensitivity or specificity to detect proarrhythmic Concordance of risk signatures with preclinical or perceived clinical
risk or provide a precise dose–response relationship in regards to TdeP risk would be based on comparable drug concentrations.
drug-induced proarrhythmia. Similar arguments can be made regard- Additional modulating “off-target” effects would also have to be
ing the utility of QT interval prolongation to quantitatively predict considered (for example, alpha-adrenergic agonism/antagonism). This
proarrhythmic risk. As most drugs that have warranted removal from necessitates a detailed accounting of experimental conditions (tissues
the market have demonstrated hERG liability, this assay remains studied, K+ levels, measured drug concentrations, acute vs. chronic
invaluable in early screening of proarrhythmic risk. However, evolving exposures, etc.) to provide context. A simple starting question for such
drugs (especially those displaying a mixture of modest or conflicting a classification could be delineating characteristics of hERG blocking
preclinical QT signals) must be evaluated beyond simpler hERG and QT drugs that do not delay repolarization or prolong the QT interval
prolongation studies in order to avoid discarding efficacious and (covert hERG blocking agents). Our future challenge is to devise and
potentially safe. This will be best accomplished using high content implement a reliable, robust testing strategy that extends beyond
cellular assays and/or evolving proarrhythmia models. hERG block and QT prolongation surrogate markers to provide a more
Proarrhythmia models of increasing complexity (by inclusion of comprehensive integrative risk assessment of torsadogenic risk.
known risk factors for TdeP) have the potential to more realistically
evaluate drug-induced proarrhythmic risk. However, such models References
require considerable expertise and thus are not easily amenable to
Abernethy, D. R., Barbey, J. T., Franc, J., Brown, K. S., Feirrera, I., Ford, N., et al. (2001).
routine screening efforts. It is presently difficult to determine how to
Loratadine and terfenadine interaction with nefazodone: both antihistamines are
meaningfully rank various proarrhythmic risk indices derived from associated with QTc prolongation. Clin Pharmacol Ther 69(3), 96−103.
these complex models to provide a realistic, multifactorial measure of Antzelevitch, C., & Shimizu, W. (2002). Cellular mechanisms underlying the long QT
proarrhythmic liability (and hence an accurate therapeutic margin). syndrome. Curr Opin Cardiol 17, 43−51.
Anyukhovsky, E. P., Susunov, E. A., Gainullin, R. Z., & Rosen, M. R. (1999). The controver-
Furthermore, rankings of indices may vary with experimental con- sial M cell. J Cardiovasc Electrophysiol 10, 244−260.
ditions or drug concentrations. Furthermore, additional “stressors” Auer, J., Berent, R., & Eber, B. (2002). Amiodarone in the prevention and treatment of
may increase sensitivity at the expense of specificity (for example, arrhythmia. Curr Opin Investig Drugs 3(7), 1037−1044.
Belardinelli, L., Antzelevitch, C., & Vos, M. A. (2003). Assessing predictors of drug-
addition of ATXII to in vitro Langendorff models). Despite such issues, induced Torsade de Pointes. Trends Pharmacol Sci 24(12), 619−625.
the more complex in vivo proarrhythmia models may prove advanta- Bril, A., Gout, B., Bonhomme, M., Landais, L., Faivre, J. F., Linee, P., et al. (1996). Combined
geous in that plasma protein binding and metabolites (an integral part potassium and calcium channel blocking activities as a basis for antiarrhythmic
efficacy with low proarrhythmic risk: experimental profile of BRL-32872.
of the experiment) may be comparable to clinical studies. Benchmark- J Pharmacol Exp Ther 276, 637−646.
ing the appropriate experimental conditions to mimic clinical risk is Carlsson, L., Abrahamsson, C., Andersson, B., Duker, G., & Schiller-Linhardt, G. (1993).
essential to establish the utility of proarrhythmia models. This exercise Proarrhythmic effects of the class III agent almokalant: importance of infusion rate,
QT dispersion, and early afterdepolarizations. Cardiovasc Res 27(12), 2186−2193.
is presently hampered by the fact that the perceived clinical risk of
Chen, X., Cordes, J. S., Bradley, J. A., Sun, Z., & Zhou, J. (2006). Use of arterially perfused
TdeP is related to both direct cardiac effects as well as indirect effects rabbit ventricular wedge in predicting arrhythmogenic potentials of drugs.
(for example, “metabolic multiplication” resulting from drug–drug J Pharmacol Toxicol Methods 54(3), 261−272.
208 G.A. Gintant / Pharmacology & Therapeutics 119 (2008) 199–209

Chiba, K., Sugiyama, A., Hagiwara, T., Takahashi, S. -I., Takasuna, K., & Hashimoto, K. Liu, T., Brown, B. S., Wu, Y., Antzelevitch, C., Kowey, P. R., & Yan, G. -X. (2006). Blinded
(2004a). In-vivo experimental approach for the risk assessment of fluoroquinolone validation of the isolated arterially perfused rabbit ventricular wedge in preclinical
antibacterial agents-induced long QT syndrome. Eur J Pharmacol 486, 189−200. assessment of drug-induced proarrhythmias. Heart Rhythm 3, 948−956.
Chiba, K., Sugiyama, A., Takasuna, K., Hashimoto, K. (2004b). Comparison of sensitivity Lu, H. R., Vlaminckx, E., Van de Water, A., Rohrbacher, J., Hermans, A., & Gallacher, D. J.
of surrogate markers of drug-induced Torsades de Pointes in canine hearts. Eur J (2007). In-vitro experimental models for the risk assessment of antibiotic-induced
Pharmacol. 11;502(1–2):117–22. QT prolongation. Eur J Pharmacol 577(1–3), 222−232.
Committee for Proprietary Medicinal Products (CPMP) (1997). Points to consider: the Malik, M., & Camm, A. J. (2001). Evaluation of drug-induced QT interval prolongation:
assessment of the potential for QT interval prolongation by non-cardiovascular Implications for drug approval and labeling. Drug Safety 24, 323−351.
medicinal products. European Agency for the Evaluation of Medicinal Products. Human Martin, R. L., McDermott, J. S., Salmen, H. J., Palmatier, J., Cox, B. F., & Gintant, G. A.
Medicines Evaluation Unit Accessed on April 4, 2008 at CPMP/986/96http://www. (2004). The utility of hERG and repolarization assays in evaluating delayed cardiac
tga.health.gov.au/docs/pdf/euguide/swp/098696en.pdf repolarization: influence of multi-channel block. J Cardiovasc Pharmacol 43(3),
Conrath, C. E., & Opthof, T. (2006). Ventricular repolarization. An overview of (patho) 369−379.
physiology, sympathetic effects, and genetic aspects. Prog Biophys Mol Biol 92, Michael, G., Dempster, J., Kane, K. A., & Coker, S. J. (2007). Potentiation of E-4031-
269−307. induced Torsade de Pointes by HMR1556 or ATX-II is not predicted by action
DeBruin, M. L., Pettersson, M., Meyboom, R. H., Hoes, A. S. W., & Leufkens, H. G. (2005). potential short-term variability or triangulation. Br J Pharmacol 152(8), 1215−1227.
Anti-HERG activity and the risk of drug-induced arrhythmias and sudden death. Eur Millberg, P., Ramtin, S., Monnig, G., Osada, N., Wasmer, K., Berithardt, G., et al. (2004).
Heart J 26(6), 590−597. Comparison of the in vitro electrophysiology and proarrhythmic effects of
Dennis, A., Wang, L., Wan, X., & Ficker, E. (2007). HERG channel trafficking: novel targets amiodarone and sotalol in a rabbit model of acute atrioventricular block. J Cardiovasc
in drug-induced long QT syndrome. Biochem Soc Trans 35, 1060−1063. Pharm 44, 278−286.
Di Diego, J. M., Belardinelli, L., & Antzelevitch, C. (2003). Cisapride-induced transmural Moxifloxacin product label. (2001). Accessed at http://www.fda.gov/cder/foi/nda/2001/
dispersion of repolarization and Torsade de Pointes in the canine left ventricular 21-085S010_Avelox_prntlbl.pdf on 2008, March 29.
wedge preparation during epicardial stimulation. Circulation 108(8), 1027−1033. Napolitano, C., Priori, S. G., & Schwartz, P. J. (1994). Torsade de Pointes. Mechanisms and
Farkas, A.S., Acsai, K., Tóth, A., Dézsi, L., Orosz, S., Forster, T.et al. (2006). Importance of Management. Drugs 47(1), 51−65.
extracardiac alpha1-adrenoceptor stimulation in assisting dofetilide to induce Oosterhoff, P., Oros, A., & Vos, M. A. (2007). Beat-to-beat variability of repolarization: a
Torsade de Pointes in rabbit hearts. Eur J Pharmacol. 10;537(1–3):118–25. new parameter to determine arrhythmic risk of an individual or identify
Fermini, B., & Fossa, A. A. (2003). The impact of drug-induced QT interval prolongation proarrhythmic drugs. Anadolu Kardiyol Derg 7(Suppl 1), 73−78.
on drug discovery and development. Nat Rev Drug Discov 2(6), 439−447. Opthof, T., Coronel, R., Wilms-Schopman, F. J., Plotnikov, A. N., Shlapakova, I. N., Danilo,
Fossa, A. A., Gorczyca, W., Wisialowski, T., Yasgar, A., Wang, E., Crimin, K., et al. (2007). P., Jr., et al. (2007). Dispersion of repolarization in canine ventricle and the
Electrical alternans and hemodynamics in the anesthetized guinea pig can electrocardiographic T wave: Tp-e interval does not reflect transmural dispersion.
discriminate the cardiac safety of antidepressants. J Pharmacol Toxicol Methods 55(1), Heart Rhythm 4(3), 341−348.
78−85. Oros, A., Volders, P. G. A., Beekman, J. D. M., van der Nagel, T., & Vos, M. A. (2006). Atrial-
Garnett, C. E., Beasley, N., Bhattaram, V. A., Jadhav, P. R., Madabushi, R., Stockbridge, N., et al. specific drug AVE-0118 is free of Torsades de Pointes in anesthetized dogs with
(2008). Concentration–QT relationships play a key role in the evaluation of chronic complete atrioventricular block. Heart Rhythm 3(11), 1339−1345.
proarrhythmic risk during regulatory review. J Clin Pharmacol 48(1), 13−18. Redfern, W. S., Carlsson, L., Davis, A. S., Lynch, W. G., MacKenzie, Il, Palethorpe, S., et al.
Gintant, G. A., Su, Z., Martin, R. L., & Cox, B. F. (2006). Utility of hERG assays as surrogate (2003). Relationships between preclinical cardiac electrophysiology, clinical QT
markers of delayed cardiac repolarization and QT safety. Toxicol Pathol 34(1), 81−90. interval prolongation and Torsade de Pointes for a broad range of drugs: evidence
Gross, G. J. (2006). Bradycardia-mediated ventricular electrical remodeling. J Cardiovasc for a provisional safety margin in drug development. Cardiovasc Res 58(1), 32−45.
Electrophysiol 17(Suppl 1), S178−S182. Roden, D. M. (1998). Taking the “idio” out of “idiosyncratic”. Pacing Clin Electrophysiol 21(5),
Guidance for Industry (2005a, Oct). E14 Clinical Evaluation of QT/QTc Interval 1029−1034.
Prolongation and Proarrhythmic Potential for Non-antiarrhythmic Drugs. ICH. Roden, D. M. (2004). Drug-induced prolongation of the QT interval. N Engl J Med 350
accessed at http://www.fda.gov/cber/gdlns/iche14qtc.pdf on March 29, 2008. (10), 1013−1022.
Guidance for Industry (2005b, Oct). S7B Nonclinical Evaluation of the Potential for Sanguinetti, M.C., Tristani-Firouzi, M. (2006). hERG potassium channels and cardiac
Delayed Ventricular Repolarization (QT Interval Prolongation) by Human Pharmaceu- arrhythmia. Nature. 23;440(7083):463–9.
ticals. ICH. Oct accessed at http://www.fda.gov/cder/guidance/6885fnl.pdf on Sanguinetti, M. C., Jiang, C., Curran, M. E., & Keating, M. T. (1995). A mechanistic link
March 29, 2008. between an inherited and an acquired cardiac arrhythmia: HERG encodes the IKr
Hinterseer, M., Thomsen, M. B., Beckmann, B. M., Pfeufer, A., Schimpf, R., Wichmann, H. E., potassium channel. Cell 81, 299−307.
et al. (2008). Beat-to-beat variability of QT intervals is increased in patients with drug- Shah, R. R. (2005a). Drugs, QT interval prolongation and ICH E14. The need to get it right.
induced long-QT syndrome: a case control pilot study. Eur Heart J 29(2), 185−190. Drug Safety 28(2), 115−125.
Hondeghem, L. M., & Hoffman, P. (2003). Blinded test in isolated female rabbit heart Shah, R. R. (2005b). Drug-induced QT interval prolongation—regulatory guidance and
reliably identifies action potential duration prolongation and proarrhythmic drugs: perspectives on hERG channel studies. Novartis Found Symp 266, 251−280.
Importance of triangulation reverse-use dependence and instability. J Cardiovasc Shah, R. R. (2005c). Drugs, QTc interval prolongation and final ICH E14 guideline: an
Pharmacol 41, 14−24. important milestone with challenges ahead. Drug Safety 28(11), 1009−1028.
Hondeghem, L.M., Carlsson, L., Duker, G. (2001). Instability and triangulation of the Shah, R. R., & Hongehem, L. M. (2005). Refining detection of drug-induced proarrhythmia:
action potential predict serious proarrhythmia, but action potential duration QT interval and TRIaD. Heart Rhythm 2, 758−772.
prolongation is antiarrhythmic. Circulation 17;103(15), 2004–2013. Shimizu, W., & Antzelevitch, C. (1998). Sodium channel blockers on transmural
Jonker, D. M., Kenna, L. A., Leishmann, D., Wallis, R., Milligan, P. A., & Jonsson, E. N. dispersion of repolarization and Torsade de Syndrome: effects of β-adrenergic
(2005). A pharmacokinetic-pharmacodynamic model for the quantitative predic- agonists and antagonists and sodium cellular basis for the ECG features of the LQT1
tion of dofetilide clinical QT prolongation from human ether-a-go-go-related gene form of the long-QT. Circulation 98, 2314−2322.
current inhibition data. Clin Pharmacol Ther 77(6), 572−582. Singh, B. N. (2006). Amiodarone: a multifaceted antiarrhythmic drug. Curr Cardiol Rep 8(5),
Jurgens, G., Graudal, N. A., & Kampmann, J. P. (2003). Therapeutic drug monitoring of 349−355.
antiarrhythmic drugs. Clin Pharmacokinet 42(7), 647−663. Steidl-Nichols, J. V., Hanton, G., Leaney, J., Liu, R. -C., Leishman, D., McHarg, A. S., et al.
Kang, J., Wang, L., Chen, X. L., Triggle, D. J., & Rampe, D. (2001). Interactions of a series of (2008). Impact of Study Design on proarrhythmia prediction in the SCREENIT rabbit
fluoroquinolone antibacterial drugs with the human cardiac K+ channel HERG. Mol isolated heart model. J Pharmacol Toxicol Methods 57(1), 9−22.
Pharmacol 59(1), 122−126. Stengl, M., Ramakers, C., Donker, D. W., Nabar, A., Rybin, A. V., Spätjens, R. L., et al. (2006).
Kijtawornrat, A., Nishijima, Y., Roche, B. M., Keene, B. W., & Hamlin, R. L. (2006). Use of a Temporal patterns of electrical remodeling in canine ventricular hypertrophy: focus
failing rabbit heart as a model to predict torsadogenicity. Toxicol Sci 93(1), 205−212. on IKs downregulation and blunted beta-adrenergic activation. Cardiovasc Res 72(1),
Kirsch, G. E., Trepakova, E. S., Brimecombe, J. C., Sidach, S. S., Erickson, H. D., Kochan, M. C., 90−100.
et al. (2004). Variability in the measurement of hERG potassium channel inhibition: Stork, D., Timin, E. N., Berjukow, S., Huber, C., Hohaus, A., Auer, M., et al. (2007). State
effects of temperature and stimulus pattern. J Pharmacol Toxicol Methods 50(2), dependent dissociation of HERG channel inhibitors. Br J Pharmacol 151(8), 1368−1376.
93−101. Taggart, P., Sutton, P., Opthof, T., Coronel, R., & Kallis, P. (2003). Electrotonic cancellation
Kongsamut, S., Kang, J., Chen, X.L., Roehr, J., Rampe, D. (2002). A comparison of the of transmural electrical gradients in the left ventricle in man. Prog Biophys Mol Biol
receptor binding and HERG channel affinities for a series of antipsychotic drugs. Eur 82, 243−254.
J Pharmacol. 16;450(1):37–41. Thomsen, M. B., Volders, P. A., Stengl, M., Spätjens, R. L., Beekman, J. D. M., Bischoff, U., et al.
Kuryshev, Y. A., Ficker, E., Wang, L., Hawryluk, P., Dennis, A. T., Wible, B. A., et al. (2005). (2003). Electrophysiological safety of sertindole in dogs with normal and remodeled
Pentamidine-induced long QT syndrome and block of hERG trafficking. J Pharmacol hearts. J Pharmacol Exp Ther 307, 776−784.
Exp Ther 312(1), 316−323. Thomsen, M.B., Verduyn, S.C., Stengl, M., Beekman, J.D., de Pater, G., van Opstal, J.et al.
Lawrence, C. L., Bridgland-Taylor, M. H., Pollard, C. E., Hammond, T. G., & Valentin, J. -P. (2004). Increased short-term variability of repolarization predictsD-sotalol-induced
(2006). A rabbit Langendorff heart proarrhythmia model: predictive value for Torsades de Pointes in dogs. Circulation. 19;110(16):2453–9.
clinical identification of Torsades de Pointes. Br J Pharmacol 149, 845−860. Thomsen, M. B., Truin, M., van Opstal, J. M., Beekman, J. D., Volders, P. G., Stengl, M.,
Lazzara, R. (1989). Amiodarone and Torsade de Pointes. Ann Intern Med 111(7), 549−551. et al. (2005). Sudden cardiac death in dogs with remodeled hearts is associated
Lengyel, C., Varró, A., Tábori, K., Papp, J. G., & Baczkó, I. (2007). Combined pharma- with larger beat-to-beat variability of repolarization. Basic Res Cardiol 100,
cological block of I(Kr) and I(Ks) increases short-term QT interval variability and 279−287.
provokes Torsades de Pointes. Br J Pharmacol 151(7), 941−951. Thomsen, M. B., Beekman, J. D. M., Attevelt, N. J. M., Takahara, A., Sugiyama, A., Chibba, K.,
Limberis, J. T., McDermott, J. S., Salmen, H. J., Su, Z., Mikhail, A., Green, J. R., et al. (2007). et al. (2006). No proarrhythmic properties of the antibiotics moxifloxacin or
The effects of plasma proteins on delayed repolarization in vitro with cisapride, azithromycin in anesthetized dogs with chronic AV block. Br J Pharmacol 149,
risperidone, andDL, -sotalol. J Pharmacol Toxicol Methods 56(1), 11−17. 1039−1048.
G.A. Gintant / Pharmacology & Therapeutics 119 (2008) 199–209 209

Thomsen, M. B., Oros, A., Schoenmakers, M., van Opstal, J. M., Maas, J. N., Beekman, J. D., Witchel, H.J., Pabbathi, V.K., Hofmann, G., Paul, A.A., Hancox, J.C. (2002). Inhibitory
et al. (2007). Proarrhythmic electrical remodeling is associated with increased beat- actions of the selective serotonin re-uptake inhibitor citalopram on HERG and
to-beat variability of repolarization. Cardiovasc Res 73, 521−530. ventricular L-type calcium currents. FEBS Lett 13;512(1–3):59–66.
Titier, K., Canal, M.., Déridet, E., Abouelfath, A., Gromb, S., Molimard, M., & Moore, N. Wu, Y., Carlsson, L., Liu, T., Kowey, P. R., & Yan, G. X. (2005). Assessment of the
(2004). Determination of myocardium to plasma concentration ratios of five proarrhythmic potential of the novel antiarrhythmic agent AZD7009 and dofetilide
antipsychotic drugs: Comparison with their ability to induce arrhythmia and in experimental models of Torsades de Pointes. J Cardiovasc Electrophysiol 16(8),
sudden death in clinical practice. Toxicol Appl Pharmacol 199(1), 52−60. 898−904.
Tsuji, Y., Opthof, T., Yasui, K., Inden, Y., Takemura, H., Niwa, N., et al. (2002). Ionic Wu, L., Shryock, J. C., Song, Y., & Belardinelli, L. (2006). An increase in late sodium
mechanisms of acquired QT prolongation and Torsades de Pointes in rabbits with current potentiates the proarrhythmic activities of low risk QT prolonging drugs in
chronic complete atrioventricular block. Circulation 106(15), 2012−2018. female rabbit hearts. J Pharmacol Exp Ther 316, 718−726.
Ueda, N., Zipes, D. P., & Wu, J. (2004). Functional and transmural modulation of M cell Wu, L., Rajamani, S., Shryock, J. C., Li, H., Ruskin, J., Antzelevitch, C., et al. (2008).
behavior in canine ventricular wall. Am J Physiol Heart Circ Physiol 287, H2569−2574. Augmentation of late sodium current unmasks the proarrhythmic effects of
van der Linde, H., van de Water, A., Loots, W., van Deuren, B., Lu, H. R., van Ammel, K., et al. amiodarone. Cardiovasc Res 77, 1−8.
(2005). A new method to calculate the beat-to-beat instability of QT duration in Yan, G. X., & Antzelevitch, C. (1998). Cellular basis for the normal T wave and the
drug-induced long QT in anesthetized dogs. J Pharmacol Toxicol Methods 52(1), electrocardiographic manifestations of the Long QT syndrome. Circulation 98,
168−177. 1928−1936.
Van Opstal, J. M., Leunissen, J. D., Wellens, H. J., Vos, M. A., et al. (2001a). Azimilide and Yan, G. X., Shimizu, W., & Antzelevitch, C. (1998). Characteristics and distribution of M
dofetilide produce similar electrophysiologic and proarrhythmic effects in a canine cells in arterially perfused canine left ventricular wedge preparations. Circulation
model of Torsade de Pointes arrhythmias. Eur J Pharmacol 412, 67−76. 98(18), 1921−1927.
van Opstal, J. M., Schoenmakers, M., Verduyn, S. C., de Groot, S. H., Leunissen, J. D., van Yan, G. X., Su, Y., Liu, T., Wang, J., Marinchak, R. A., & Kowey, P. R. (2001). Phase 2 early
Der Hulst, F. F., et al. (2001b). Chronic amiodarone evokes no Torsade de Pointes afterdepolarization as a trigger of polymorphic ventricular tachycardia in acquired
arrhythmias despite QT lengthening in an animal model of acquired long-QT long-QT syndrome: direct evidence from intracellular recording in the intact left
syndrome. Circulation 104, 2722−2727. ventricular wall. Circulation 103, 2851−2856.
van Opstal, J. M., Verduyn, S. C., Leunissen, H. D. M., de Groot, S. H. M., Wellens, H. J. J., & Yao, J. A., Du, X., Lu, D., Baker, R. L., Daharsh, E., & Atterson, P. (2005). Estimation of
Vos, M. A (2001c). Electrophysiological parameters indicative of sudden cardiac potency of HERG channel blockers: impact of voltage protocol and temperature.
death in the dog with chronic complete AV-block. Cardiovasc Res 50(2), 354−361. J Pharmacol Toxicol Methods 52(1), 146−153.
Volders, P. G., Sipido, K. R., Vos, M. A., Kulcsár, A., Verduyn, S. C., & Wellens, H. J. (1998). Zeltser, D., Justo, D., Halkin, A., Prokhorov, V., Heller, K., & Viskin, S. (2003). Torsade de
Cellular basis of biventricular hypertrophy and arrhythmogenesis in dogs with Pointes due to noncardiac drugs. Most patients have easily identifiable risk factors.
chronic complete atrioventricular block and acquired Torsade de Pointes. Circulation Medicine (Baltimore) 82, 282−290.
98(11), 1136−1147. Zhang, S., Zhou, Z., Gong, Q., Makielski, J. C., & January, C. T. (1999). Mechanism of block
Volders, P. G., Sipido, K. R., Vos, M. A., Spätjens, R. L., Leunissen, J. D., Carmeliet, E., et al. and identification of the verapamil binding domain to hERG potassium channels.
(1999). Downregulation of delayed rectifier K(+) currents in dogs with chronic Circ Res 84(9), 989−998.
complete atrioventricular block and acquired Torsades de Pointes. Circulation 100(24),
2455−2461.
Wallis, Robert M. (2008). Preclinical Testing vs. Clinical Results; Implications for Internal
Decision Making and Regulatory Actions. Drug Information Association Meeting;
QT and Arrhythmia Issues in Drug Development. Bethesda MD (April 7, 2008).

You might also like