Skip to main content

Graphical Abstract

Abstract

Rationale:

The class Ic antiarrhythmic drug flecainide prevents ventricular tachyarrhythmia in patients with catecholaminergic polymorphic ventricular tachycardia (CPVT), a disease caused by hyperactive RyR2 (cardiac ryanodine receptor) mediated calcium (Ca) release. Although flecainide inhibits single RyR2 channels in vitro, reports have claimed that RyR2 inhibition by flecainide is not relevant for its mechanism of antiarrhythmic action and concluded that sodium channel block alone is responsible for flecainide’s efficacy in CPVT.

Objective:

To determine whether RyR2 block independently contributes to flecainide’s efficacy for suppressing spontaneous sarcoplasmic reticulum Ca release and for preventing ventricular tachycardia in vivo.

Methods and Results:

We synthesized N-methylated flecainide analogues (QX-flecainide and N-methyl flecainide) and showed that N-methylation reduces flecainide’s inhibitory potency on RyR2 channels incorporated into artificial lipid bilayers. N-methylation did not alter flecainide’s inhibitory activity on human cardiac sodium channels expressed in HEK293T cells. Antiarrhythmic efficacy was tested utilizing a Casq2 (cardiac calsequestrin) knockout (Casq2−/−) CPVT mouse model. In membrane-permeabilized Casq2−/− cardiomyocytes—lacking intact sarcolemma and devoid of sodium channel contribution—flecainide, but not its analogues, suppressed RyR2-mediated Ca release at clinically relevant concentrations. In voltage-clamped, intact Casq2−/− cardiomyocytes pretreated with tetrodotoxin to inhibit sodium channels and isolate the effect of flecainide on RyR2, flecainide significantly reduced the frequency of spontaneous sarcoplasmic reticulum Ca release, while QX-flecainide and N-methyl flecainide did not. In vivo, flecainide effectively suppressed catecholamine-induced ventricular tachyarrhythmias in Casq2−/− mice, whereas N-methyl flecainide had no significant effect on arrhythmia burden, despite comparable sodium channel block.

Conclusions:

Flecainide remains an effective inhibitor of RyR2-mediated arrhythmogenic Ca release even when cardiac sodium channels are blocked. In mice with CPVT, sodium channel block alone did not prevent ventricular tachycardia. Hence, RyR2 channel inhibition likely constitutes the principal mechanism of antiarrhythmic action of flecainide in CPVT.

Introduction

Editorial, see p 332
In This Issue, see p 305
Meet the First Author, see p 306
Catecholaminergic polymorphic ventricular tachycardia (CPVT) is a genetic arrhythmia syndrome caused by mutations in proteins that constitute the calcium (Ca) release unit of the cardiac sarcoplasmic reticulum (SR), such as RyR2 (cardiac ryanodine receptor) SR Ca release channel,1 Casq2 (cardiac calsequestrin),2,3 and calmodulin.4,5 Mutations in these proteins cause spontaneous SR Ca release during adrenergic stress that can trigger potentially fatal ventricular arrhythmias.6,7 The class Ic antiarrhythmic drug flecainide, a cardiac sodium channel blocker,8 exhibits striking efficacy for the prevention of ventricular arrhythmias in patients with CPVT.9–12 Despite its proven clinical efficacy in CPVT, the mechanism of flecainide action remains controversial. Based on our discovery that flecainide inhibits RyR2 Ca release channels, we have suggested that RyR2 block contributes independently to its mechanism of action in CPVT.9 This concept was supported by in vitro experiments showing that clinically relevant concentrations of flecainide prevented spontaneous Ca release in CPVT cardiomyocytes even in the absence of extracellular sodium and Ca,9 or after cell membrane permeabilization which renders sodium channels inactive.13,14 Subsequent studies have disputed the importance or action of RyR2 block by flecainide and concluded that sodium channel block alone is responsible for flecainide’s efficacy in CPVT.15–18 Those conclusions were based on observations that flecainide’s inhibitory potency in single RyR2 channel assays is too low to explain its clinical efficacy and that sodium channel blockers lacking single-channel RyR2-blocking properties, such as tetrodotoxin18 or QX-flecainide (QX-FL) also suppressed spontaneous SR Ca release in intact cardiomyocytes.16
Flecainide’s mechanism of action is of major importance; hyperactive RyR2 channels are implicated in a variety of heart diseases associated with atrial and ventricular tachyarrhythmias,19–21 but currently there is no clinically available RyR2-specific inhibitor. Uncovering whether RyR2 inhibition by flecainide plays a role for its antiarrhythmic activity would validate RyR2 as therapeutic target and facilitate the drug development of specific RyR2 inhibitors. Because of its proposed dual mode of action—sodium channel block and RyR2 block—it is difficult to resolve flecainide’s mechanism of action. It is well established that sodium channel blockers inhibit RyR2-mediated Ca release indirectly via the Na/Ca exchanger by altering intracellular sodium and hence cytosolic and intraSR Ca concentrations.18 Remarkably, studies claiming that RyR2 block is not therapeutically relevant have not examined the effect of flecainide on RyR2-mediated SR Ca release in the complete absence of sodium channel activity.
To distinguish between these 2 mechanisms (sodium channel block versus RyR2 inhibition) and isolate flecainide’s direct effect on RyR2, we synthesized flecainide analogues that retain sodium channel block but exhibit substantially lower potency of single-channel RyR2 inhibition. We then tested the effects of flecainide and its analogues on spontaneous Ca release in cardiomyocytes from Casq2−/− mice, an established model of severe CPVT.22 To investigate the contribution of RyR2 block to its antiarrhythmic efficacy in vivo, we utilized our single N-methylated flecainide analogue that has pharmacological properties amenable to in vivo administration, inhibits sodium channel with similar potency as flecainide but does not block RyR2 Ca release in isolated cardiomyocytes.

Methods

The materials and data from this study are available from the corresponding author upon reasonable request.
See Data Supplement for expanded methods. The use of animals was approved by the Animal Care and Use Committee of Vanderbilt University Medical Center (animal protocol No. M1900057-00) and performed in accordance with National Institutes of Health guidelines. Representative images/traces were selected from records that had values close to the mean/median measure. Statistical analyses were performed using Prism v6.01 (GraphPad Software, Inc) or R (https://www.R-project.org/). Statistical tests were used as reported in the figure legends.

Results

Generation of Flecainide Analogues With Reduced RyR2 Inhibitory Activity

To isolate the specific effects of flecainide on RyR2-mediated Ca release, we synthesized 2 flecainide analogues, a tetraalkylammonium salt derivative of flecainide, which is a quaternary ammonium cation also known as QX-FL,23 and a single-methylated flecainide derivative, N-methyl flecainide (NM-FL), shown in Figure 1A. We hypothesized that the secondary amine on the piperidine ring in flecainide is necessary for its activity in RyR2 channels but not sodium channels. This idea stems from work in which QX-FL has been shown to not block RyR2 in single-channel studies performed at +40 mV membrane potential difference, where an effect with flecainide is observed.16 While both QX-FL and NM-FL have similar benzamide-derived structures compared with flecainide, there are key differences in the piperidine moiety. QX-FL, synthesized using a modified literature procedure, is doubly methylated at the piperidine nitrogen, resulting in a quaternary ammonium salt.24 This modification renders QX-FL permanently charged and removes its ability to participate in hydrogen bonding at that nitrogen center. To test the hypothesis that the secondary amine is critical for flecainide RyR2 block as well as make a substrate suitable for in vivo administration, we synthesized NM-FL using an adapted Eschweiler-Clarke reaction to install a single methyl at the piperidine nitrogen.24 This substrate maintains similar physicochemical properties to flecainide yet also has the key N-H donor removed. Flecainide has a pKa of 9.3, resulting in 99% ionization at physiological pH (7.4). Although an exact pKa value for NM-FL has not been experimentally determined, the effect of N-methylation of amines on pKa has been extensively studied and documented. Hall shows that the pKa of 2-methylpiperidine and that of 1,2-dimethyl piperidine are <1 pKa unit apart.25,26 This can be extended to the case of flecainide and NM-FL. Hence, like flecainide, NM-FL is predicted to be predominately ionized at physiological pH. Sodium channel or RyR2 block by NM-FL has not been characterized previously.
Figure 1. N-Methylation of flecainide reduces its inhibitory activity on the single-channel cardiac RyR2 (ryanodine receptor). A, Chemical structures for flecainide and 2 N-methyl flecainide (NM-FL) analogues: double NM-FL, a quaternary ammonium cation (QX-flecainide [QX-FL]) and single NM-FL. B, Representative single-channel recordings from sheep RyR2 incorporated into lipid bilayers and treated with flecainide (top), QX-FL (middle), or NM-FL (bottom). Recordings were made in the presence of 0.1 µmol/L free Ca and 2 mmol/L ATP, and the membrane potential was held at +40 mV. Open (o) and closed (c) states are labeled in the first recording. C, RyR2 open probability (Po) at various concentrations of flecainide or its analogues (normalized to maximum Po for each channel without drug). NFlecainide=13 (5 µmol/L), 10 (10 µmol/L), 7 (20 µmol/L), 6 (30 µmol/L), 8 (50 µmol/L). NNM-FL=8 (10 µmol/L), 10 (20 µmol/L), 5 (50 µmol/L), 6 (100 µmol/L), 3 (150 µmol/L), 4 (200 µmol/L). NQX-FL=4 (10 µmol/L), 4 (20 µmol/L), 5 (50 µmol/L), 5 (100 µmol/L), 6 (150 µmol/L), 12 (200 µmol/L). Extra sum-of-squares F test was used to compare IC50 values. P=3.71×10−9 for Flecainide vs NM-FL. P=2.55×10−5 for Flecainide vs QX-FL. P=4.86×10−2 for NM-FL vs QX-FL.

N-Methylation of Flecainide Reduces Its Inhibitory Activity on RyR2 Channels

To determine the structure-activity relationship of the N-methylated flecainide analogues, we recorded single-channel RyR2 openings and measured open probability (RyR2 Po) from sheep RyR2 incorporated into lipid bilayers (representative traces shown in Figure 1B). At end-diastolic cytoplasmic Ca concentrations (0.1 µmol/L), RyR2 open probability had a skewed distribution with a median of 0.042 and 25 to 75 percentiles at 0.009 and 0.18 (n=60). Addition of flecainide to the cytoplasmic bath at a membrane potential of +40 mV inhibited RyR2 with an IC50 of 15.9 µmol/L (CI95%=[10.9–21.2]). Phosphorylation of single RyR2 channels had no significant effect on the potency of flecainide block (Figure I in the Data Supplement). N-Methylation of flecainide substantially reduced its inhibitory potency on RyR2 channels. The IC50 of the single-methylated NM-FL was 99 µmol/L (CI95%=[68.7–144], P=3.71×10−9 versus flecainide). Double methylation further reduced the inhibitory potency, with an IC50 of 182 µmol/L for QX-FL (CI95%=[128–259], P=2.55×10−5 versus flecainide, P=4.86×10−2 versus NM-FL for QX-FL, Figure 1B and 1C).
We next tested flecainide and its analogues on RyR2 at a Ca concentration of 100 µmol/L, which is the cytoplasmic Ca concentration in the dyadic cleft during systole.27 Under those conditions, RyR2 was near maximally activated (median Po=0.68, 25–75 percentiles at 0.21 and 0.86, n=32). As previously reported,9 high cytosolic Ca reduces the potency of RyR2 block by flecainide (IC50=88 µmol/L (CI95%=[78.5–99.5]), Figure II in the Data Supplement), which may explain why flecainide does not inhibit systolic SR Ca release during normal excitation-contraction coupling.28 N-Methylation significantly reduced the inhibitory potency of flecainide also at elevated Ca concentrations, with an IC50 of 357 µmol/L (CI95%=[254–502]) for the single-methylated NM-FL (P=2.34×10−14 versus flecainide), and 773 µmol/L (CI95%=[626–952]) for the double-methylated QX-FL (P=5.19×10−24 versus flecainide, Figure II in the Data Supplement).
To test whether reduced activity on single RyR2 channels translates into altered drug activity when the RyR2 channel is incorporated in its biological SR membrane, we recorded spontaneous Ca waves—which are generated by RyR2-mediated SR Ca release—in Casq2−/− cardiomyocytes subjected to saponin to selectively permeabilize the sarcolemma and enable equivalent access of flecainide or its analogues to RyR2. Importantly, treatment with saponin produces large pores in the sarcolemma, effectively clamping the sarcolemmal membrane potential at 0 mV and eliminating any effects of sodium channel block or sarcolemmal polarity on SR Ca release, while leaving the SR membrane intact.29 Representative line scans of Ca waves are shown in Figure 2A. We observed a concentration-dependent decrease in Ca wave frequency (Figure 2B) following treatment with flecainide (IC50=7.0 µmol/L, CI95%=[4.69–10.3]). QX-FL and NM-FL did not inhibit Ca waves at the highest concentration tested, consistent with their reduced potency in the RyR2 single-channel assay (Figure 1C). Taken together, these results indicate that N-methylation of flecainide reduces its ability to inhibit single RyR2 channels and RyR2-mediated Ca release.
Figure 2. N-Methylation of flecainide abolishes its ability to inhibit sarcoplasmic reticulum (SR) calcium (Ca) waves in permeabilized Casq2−/− cardiomyocytes. A, Isolated Casq2−/− cardiomyocytes were subjected to saponin (40 µg/mL for 45 s) to selectively permeabilize the sarcolemma and enable equivalent access of flecainide or its analogues to cardiac RyR2 (ryanodine receptors). Representative line scans show spontaneous Ca release events in the form of Ca waves after 5 min incubation in vehicle (Veh.) or 25 µmol/L flecainide, QX-flecainide (QX-FL), or N-methyl flecainide (NM-FL). B, Ca wave frequency decreased in a dose-dependent manner following treatment with flecainide but not NM-FL or QX-FL. N=28, 24, 31, 29, and 38 cells for flecainide; N=23, 21, 40, and 63 cells for QX-FL; N=21, 23, 26, and 34 cells for NM-FL. Two-tailed t-tests were used to compare with vehicle. P=1.96×10−6 for a. P=1.07×10−10 for b. P=8.59×10−7 for c. Data are normalized to vehicle and shown as mean±SEM. Normality was examined before statistical testing.

NM-FL Inhibits Cardiac Sodium Channels With Properties Comparable to Flecainide

Cardiac sodium channel block by QX-FL has been extensively characterized.23 QX-FL is permanently charged at any pH, rendering it membrane impermeable. This requires it to be introduced intracellularly to exert any effect. Flecainide, while predominantly ionized at physiological pH, is in equilibrium with a small fraction that is neutral and can diffuse across the cell membrane. Once inside the cell, flecainide again reaches equilibrium with most of the compound being charged. Only the ionized form of flecainide is responsible for its characteristic sodium channel use-dependent block (UDB; ie, the channel inhibition increases with higher channel activity).23 UDB of sodium channels is an important mechanism of action for class I antiarrhythmic drugs such as flecainide.8
Since NM-FL had not previously been characterized biologically, we compared its effect on the cardiac sodium channel (NaV1.5) to that of flecainide. Experiments were carried out using whole cell patch clamp of human embryonic kidney (HEK293T) cells stably expressing human NaV1.5.30 Flecainide or NM-FL were applied in the extracellular perfusate and sodium currents were measured using standard protocols. Both flecainide and NM-FL exhibited strong UDB, with a similar concentration dependence (Figure 3A). With an IC50=7.7 µmol/L (CI95%=[6.31–9.49]) for UDB, NM-FL was modestly more potent than flecainide (IC50=10.6 µmol/L, CI95%=[8.11–13.9], P=0.07), which is consistent with a slight difference in pKa. Rate of recovery from UDB (Figure III in the Data Supplement) and voltage dependence of UDB (Figure IV in the Data Supplement) were not statistically different between flecainide and NM-FL. To assess the tonic block of sodium channels, we measured the effect of flecainide and NM-FL on the current-voltage relationship of INa activation (Figure 3B) and steady-state inactivation (Figure V in the Data Supplement). At 6 µmol/L, there were no statistically significant differences between flecainide and NM-FL (Figure 3B peak current t test, P=0.93). Taken together, these results demonstrate that N-methylation of flecainide’s piperidine moiety does not significantly alter its activity on cardiac sodium channels.
Figure 3. Characterization of INa inhibition by flecainide and N-methyl flecainide (NM-FL) in HEK293T cells expressing human NaV1.5. A, Representative current records (left) and concentration-response relationship (right) of use-dependent block (UDB) by flecainide and NM-FL. Currents were recorded before (control) and after (test, arrow) a train of conditioning pulse (300 pulses, 10 Hz frequency) in the presence of various concentrations (0.3, 1, 3, 10, and 30 µmol/L) of flecainide or NM-FL. Data are normalized to the initial test pulse. IC50 was 10.6 µmol/L for flecainide (n=3 cells for 0.3, 1, and 3 µmol/L; n=4 cells for 10 µmol/L; n=5 cells for 30 µmol/L) and 7.7 µmol/L for NM-FL (n=4 cells/concentration). Extra sum-of-squares F test was used to compare IC50 values. P=0.067 for flecainide vs NM-FL. B, Current-voltage relationship of INa activation in the absence (vehicle, n=10 cells) and presence of 6 µmol/L flecainide (n=5 cells) or NM-FL (n=5 cells) applied to the bath solution (5 min incubation). Left side of the panel shows voltage protocol for the experiment, as well as representative example traces. Peak current was compared using a 2-tailed t test. P=0.93 for flecainide vs NM-FL.

Flecainide, but Not QX-FL or NM-FL, Inhibits Spontaneous Ca Release in Intact Cardiomyocytes Under Conditions of Complete Sodium Channel Block

We next examined spontaneous SR Ca release inhibition in intact Casq2−/− cardiomyocytes using whole-cell patch clamp. Because QX-FL is permanently charged and does not cross cell membranes, flecainide, QX-FL, or NM-FL (6 µmol/L) were applied in both the extracellular (perfused solution) and intracellular solutions (dialyzed via patch pipette) for 5 minutes. Cyclic AMP (200 µmol/L) was included in the patch pipette to maximally activate PKA (protein kinase A) and mimic the catecholamine-mediated phosphorylation state that occurs during adrenergic response. Spontaneous Ca waves were recorded during a 48-second test period following termination of a short conditioning train of membrane-depolarizing steps (Figure 4A, Figure VI in the Data Supplement). To abolish any contribution of sodium channel block by flecainide and its analogues and isolate potential effects from RyR2 inhibition, cells were pretreated with 30 µmol/L tetrodotoxin for 15 minutes, which blocked INa completely (Figure VII in the Data Supplement). Tetrodotoxin pretreatment (ie, complete sodium channel block) significantly reduced Ca wave frequency by ≈45% compared with vehicle treatment (Figure 4A). In the presence of tetrodotoxin, flecainide further reduced Ca wave frequency (by ≈90%, Figure 4A), which was significantly more than that of tetrodotoxin alone (Figure 4A), indicating an additive effect. QX-FL and NM-FL, on the other hand, did not significantly reduce the frequency of spontaneous Ca waves beyond that observed with tetrodotoxin alone (Figure 4A). Flecainide, QX-FL, and NM-FL had no effect on the SR Ca content, as estimated by a rapid 10 mmol/L caffeine spritz at the end of every recording.
Figure 4. Flecainide inhibits spontaneous Ca waves in intact Casq2−/− cardiomyocytes independently of sodium channel block. A, Top: representative Ca fluorescent records from voltage-clamped ventricular myocytes after 5 min extracellular application and intracellular dialysis of either DMSO vehicle (Veh.), or 6 µmol/L flecainide, QX-flecainide (QX-FL), or N-methyl flecainide (NM-FL). In 4 out of 5 experimental groups, cells were preincubated for 15 min with 30 µmol/L tetrodotoxin (TTX) to completely block sodium channels. To elicit spontaneous calcium waves, a short conditioning train of 4 membrane depolarization steps from −70 mV to +10 mV with 1 Hz frequency was applied at the beginning of each recording (blue hash marks). Bottom: spontaneous Ca wave frequency for each treatment. N=11, 12, 13, 10, and 12 cells from 9 mice, respectively. B, Top: representative traces showing the effect of flecainide and NM-FL in the absence of TTX. Bottom: spontaneous Ca wave frequency for each treatment. Note the additive contributions from sodium channel and RyR2 block. N=17, 11, and 12 cells from 9 mice. Statistical comparisons were made using a mixed model with Bonferroni correction to account for independent cardiomyocyte isolations.46
We next compared flecainide and NM-FL in the absence of tetrodotoxin (Figure 4B). The magnitude of Ca wave inhibition by flecainide was significantly greater than that of NM-FL, further supporting the additive effect of RyR2 block on Ca wave inhibition (Figure 4B). Taken together, these results establish that flecainide inhibits arrhythmogenic RyR2-mediated SR Ca release in intact cardiomyocytes independent of its sodium channel blocking properties.

Flecainide, but Not NM-FL, Prevents Ventricular Tachycardia in Casq2−/− Mice

We next sought to determine if and to what extent the RyR2 block by flecainide influences its antiarrhythmic efficacy in vivo. Given its activity by extracellular application (Figure 3), equivalent block of cardiac sodium channels (Figure 3), and lack of direct effect on RyR2-mediated Ca waves (Figures 2 and 4) compared with flecainide, we used NM-FL as a tool to answer this question. Using the well-established Casq2−/− CPVT mouse model, Casq2−/− mice were randomized to a triple crossover study design and pretreated via intraperitoneal injection with vehicle, 15 mg/kg flecainide (Flec.), or 15 mg/kg NM-FL, followed by catecholamine challenge with isoproterenol (3 mg/kg intraperitoneal) 90 minutes later. Informed by our previous studies of flecainide pharmacokinetics in mice,9 the dose was chosen to produce flecainide plasma concentrations in mice that are within the therapeutic range (0.4–1 mg/L) used clinically to prevent ventricular arrhythmias in patients with CPVT.10 Figure 5A shows representative time courses of the heart rate in Casq2−/− mice before and after catecholamine challenge (time=0). Injection of isoproterenol causes development of ventricular arrhythmia (inset), observed as a fluctuating heart rate because of variable R-R intervals with premature ventricular complexes and ventricular tachycardia. In vivo, use-dependent sodium channel block manifests itself as a widening of the QRS complex because of the reduction in depolarization rate and hence slowed conduction velocity.31 Figure 5B shows representative QRS waveforms from a Casq2−/− mouse. Both flecainide and NM-FL prolonged the QRS complex to a comparable degree (Figure 5C), indicating that the doses were bioequivalent and both drugs exhibit equivalent potency of use-dependent sodium channel block in vivo. Arrhythmia burden was classified on an ordinal scale from 0 to 4. Only flecainide, but not NM-FL, which lacks RyR2 blocking properties, significantly decreased arrhythmia burden (Figure 5D and 5E, Figure VIIIA in the Data Supplement) and premature ventricular complexes (Figure VIIIB in the Data Supplement), despite both drugs equivalently inhibiting the sodium channel. There were no differences in heart rates between treatment groups (Figure VIIIC through VIIIE in the Data Supplement). Taken together, these results establish the importance of RyR2 inhibition by flecainide for its antiarrhythmic activity in vivo.
Figure 5. Flecainide, but not N-methyl flecainide (NM-FL), prevents ventricular tachycardia in Casq2−/− mice. A, Heart rate in a Casq2−/− mouse pretreated with DMSO (vehicle), 15 mg/kg flecainide, or 15 mg/kg NM-FL, before and after intraperitoneal (IP) injection with 3 mg/kg isoproterenol (time=0). Arrythmias produce a variable beat-to-beat heart rate on ECG. B, QRS width for each drug treatment. Left: representative ECG traces illustrating QRS morphology from the same mouse treated with DMSO (Veh.), flecainide (Flec.), or N-methyl flecainide (NM-FL). Waveforms are aligned to the initial moment of depolarization. Right: QRS width for each animal. Casq2−/− mice were randomized to a triple crossover design and injected intraperitoneal with 15 mg/kg DMSO (equivalent volume), flecainide, or NM-FL. Repeated measures ANOVA with Tukey post hoc test. Median values displayed as blue crossbars. C, Representative arrhythmias observed following catecholamine challenge. Bigeminy=alternating sinus beats with premature ventricular complexes (PVCs), couplet=2 consecutive PVCs, nonsustained ventricular tachycardia (NSVT)=3 or more consecutive PVCs. Scale bar=250 ms. D, Classification of arrhythmia risk for each treatment group. E, Arrhythmia risk score. Normality was examined before statistical testing. Data in D and E compared with pairwise Wilcoxon rank-sum test with Bonferroni correction. N=10 mice in B, D, and E.

Discussion

Here, we report that flecainide remains a highly effective inhibitor of RyR2-mediated arrhythmogenic Ca release even when cardiac sodium channels are blocked. Flecainide analogues that retain sodium channel block but have significantly reduced activity on RyR2 single channels fail to inhibit RyR2-mediated SR Ca release. These results indicate that RyR2 block by flecainide is important to suppress spontaneous Ca release in ventricular cardiomyocytes. In mice with CPVT, sodium channel block in the absence of RyR2 block did not prevent catecholamine-induced ventricular tachyarrhythmias. Hence, our results demonstrate that potent and direct inhibition of RyR2-mediated SR Ca release is required for flecainide’s antiarrhythmic action in preventing CPVT and thus resolves an ongoing controversy in the field.
Previously, some investigators had concluded that RyR2 block is not a relevant mechanism of flecainide action,17 and that sodium channel block is solely responsible for its antiarrhythmic efficacy in CPVT.15,17 This interpretation was based exclusively on in vitro experiments, mostly on single RyR2 channel data showing that flecainide does not inhibit RyR2 at a SR membrane potential of 0 mV but only at positive SR membrane potentials,17 which are considered nonphysiological based on modeling studies.32,33 However, the SR membrane potential during Ca release has never been measured experimentally and may not be 0 mV, at least not during initial phase of spontaneous SR Ca release. Thus, we carried out a careful biological assessment of flecainide inhibition of RyR2 in its native cellular environment as an extension of single-channel studies, utilizing flecainide analogues that retain sodium channel inhibitory activity but compared with flecainide have reduced activity on RyR2 single channels at +40 mV. Treatment with saponin permeabilized cardiomyocytes and removed the contribution of sarcolemmal sodium channels—clamping intracellular (sodium) and rendering sodium channels inactivated—enabling us to resolve flecainide’s effect on RyR2-mediated Ca release directly (Figure 2). The direct action of flecainide on RyR2-mediated Ca release was confirmed in intact cardiomyocytes with complete inhibition of sodium channels by tetrodotoxin (Figure 4). Importantly, our results in cardiomyocytes mirror the different inhibitory potency of flecainide and QX-FL or NM-FL on single RyR2 channels at positive membrane potentials (Figure 1 and study by Bannister et al16). Given that flecainide RyR2 block is steeply voltage-dependent,34 our experimental results suggest that the junctional SR membrane polarizes to positive potentials during spontaneous Ca release, at least transiently. Future studies will have to confirm this hypothesis experimentally. The higher potency and efficacy of flecainide in intact (Figure 4) compared with permeabilized cardiomyocytes (Figure 3) can be explained by the additive effect of the sodium channel block in intact cardiomyocytes, as illustrated in Figure 4B.
Our discovery that methylation of a single amine residue abolishes flecainide’s activity against RyR2-mediated SR Ca release provides important information on flecainide’s structure-activity relationship of RyR2 inhibition. It establishes that the N-H in the piperidine ring of flecainide is necessary for its activity on RyR2. Additionally, the results of the methylation studies suggest that flecainide binding to the RyR2 macromolecular complex is selective and supports the existence of a discrete binding site in the RyR2 macromolecular complex. Future studies are warranted to identify the flecainide binding site on RyR2, which will enable further optimization of the RyR2-blocking properties of the flecainide molecule. This is important, because hyperactive RyR2 channels are implicated as an important arrhythmia mechanism in structural heart disease such as heart failure and ischemic heart disease19,20 where sodium channel blockers are contraindicated.8

What Is The Relative Need For Sodium Channel Block Versus RyR2 Inhibition to Achieve Therapeutic Benefit From Flecainide Therapy?

To answer this question, one should consider the mechanisms underlying the generation of catecholamine-induced ventricular arrhythmias in CPVT.35 At the level of the cardiomyocyte, hyperactive RyR2 channels cause spontaneous RyR2 openings during diastole that coalesce into a propagated Ca wave, which in turn activates the electrogenic sodium calcium exchanger, resulting in a cell membrane depolarization. Membrane depolarizations of sufficient amplitude will activate voltage-gated sodium channels, which in turn will trigger a full cardiac action potential. The therapeutic benefit of RyR2 block by flecainide stems from its drastic reduction or complete prevention of arrhythmogenic calcium waves (Figure 4 and studies by Watanabe et al,9 Galimberti and Knollmann,13 and Hilliard et al28), which is the primary cellular event responsible for the generation of the spontaneous action potential in CPVT.35 The therapeutic benefit of sodium channel block stems from 2 following independent actions: (1) the reduction of cellular sodium and hence Ca loading, reducing the likelihood of spontaneous SR Ca release16,18 and (2) increasing the cell membrane potential threshold that can trigger a spontaneous action potential.15 Which of these mechanisms predominates depends largely on the experimental conditions, which has contributed to different investigators coming to various conclusions about the relative importance of each mechanism. For example, rapid pacing protocols favor sodium channel block as the primary mechanism because of the strong use-dependence of flecainide, leading to an overestimation of the contribution of sodium channel block.36 To overcome this limitation, here we used a train of voltage-clamp pulses to stimulate cardiomyocytes and elicit arrhythmogenic Ca waves. We find that a complete block of sodium channels with tetrodotoxin reduced the rate of spontaneous Ca waves by ≈45% (Figure 4). A similar reduction was achieved by sodium channel block with NM-FL, which blocks sodium channels with similar potency as flecainide (Figure 4B) but does not inhibit RyR2 at the concentration tested. On the other hand, flecainide reduced spontaneous Ca waves by over 90% (Figure 4), indicating that the additive effect of RyR2 block is substantial. In contrast, Bannister et al16 reported that sodium channel block by flecainide is solely responsible for its effect on Ca waves, based on a comparison of intracellular application of flecainide and QX-FL, which does not block RyR2 (Figure 1). However, QX-FL is a much more potent sodium channel blocker than flecainide when selectively applied to the intracellular side of the membrane because flecainide rapidly diffuses out of the cell whereas quaternary ammonium QX-FL does not.23 In our experiments, QX and flecainide were applied to both the intracellular and extracellular side of the sarcolemma at equal concentrations (Figure 4). Taken together, our results indicate that at least half of the therapeutic benefit at the level of the cardiomyocyte is derived from flecainide’s inhibition of RyR2-mediated Ca release, with a contribution by sodium channel inhibition that depends on the experimental conditions.

What Is The Relative Contribution of Sodium Channel Block Versus RyR2 Inhibition for Therapeutic Benefit From Flecainide Therapy In Vivo in Patients With CPVT?

The collective evidence from experimental and clinical studies suggests that while RyR2 inhibition is beneficial, treatment with pure sodium channel blockers does not provide benefit and may even harm patients with CPVT. In experimental studies, selective RyR2 block with ent-verticilide prevented catecholamine-induced ventricular arrhythmia in a murine CPVT model.37 In clinical studies, based on case-control studies9,11,12 and a randomized clinical trial,9 the dual RyR2 and sodium channel blocker flecainide prevented ventricular arrythmias in patients with CPVT when used as a monotherapy or in combination with β-blockers, and since 2013 is considered the standard of care in the treatment of patients with CPVT.38 Propafenone, another class Ic agent that inhibits RyR2 channels similar to flecainide, is also effective in patients with CPVT.39 Clinical experience with class Ia and Ib sodium channel blockers which lack activity on RyR213 is limited, but in a case series of 29 patients with CPVT, class Ia and Ib agents given alone or in combination with a β blocker were not protective but rather significantly increased the risk of sudden death.40 Two of the 3 cases given mexiletine and one case given disopyramide died suddenly. Our data testing the antiarrhythmic efficacy of class I agents in CPVT mouse models also indicate that sodium channel block alone is not effective in CPVT. Compared with flecainide or (R)-propafenone, sodium channel blockers that have less ((S)-propafenone) or no RyR2 blocking properties (lidocaine, procainamide) did not significantly reduce the incidence of exercise or catecholamine-induced ventricular tachycardia in CPVT mice.9,39 The current study demonstrates that the N-methyl flecainide analogue, which lacks RyR2 block at clinically relevant concentrations (Figures 1 and 2), did not reduce the severity of ventricular tachyarrhythmias in CPVT mice (Figure 5), although the degree of sodium channel block as evidenced by the QRS widening on the ECG was comparable to that of flecainide (Figure 5). Why do sodium channel blockers reduce triggered activity in isolated CPVT cardiomyocytes15–18 but do not protect against ventricular tachycardia in vivo? The answer is not clear but may relate to the well-established proarrhythmic liability of sodium channel blockers at the tissue level,6 which could explain why treatment with pure sodium channel blockers increased the risk of fatal arrhythmias in patients with CPVT.40 Collectively, these clinical and experimental data indicate that sodium channel block, by itself, likely confers little or no therapeutic benefit in CPVT. Rather, inhibition of RyR2 is the principal mechanism of antiarrhythmic action of flecainide in CPVT. Whether or not sodium channel block adds any therapeutic benefit when combined with RyR2 block—as is the case for flecainide—remains to be determined.

Acknowledgments

The authors thank Paul Johnson for his assistance with the RyR2 single channel studies.

Novelty and Significance

What Is Known?

The class Ic antiarrhythmic drug flecainide is highly effective in preventing adrenergic stress–induced arrhythmias in mouse models of catecholaminergic polymorphic ventricular tachycardia (CPVT) and in patients with CPVT refractory to standard drug treatment.
Despite flecainide’s proven clinical efficacy in CPVT, the mechanism of its therapeutic action remains controversial: it is debated whether flecainide’s direct effect on RyR2 (ryanodine receptor) is responsible for its antiarrhythmic efficacy or whether its efficacy can be solely attributed to flecainide’s inhibition of cardiac sodium channels.

What New Information Does This Article Contribute?

In vitro, flecainide analogues that retain sodium channel block but have significantly reduced activity on RyR2 single channels fail to inhibit spontaneous RyR2-mediated sarcoplasmic reticulum Ca release when sodium channel activity is eliminated.
In mice with CPVT, a flecainide analogue that lacks RyR2-inhibitory properties but is a potent Na channel blocker does not prevent ventricular tachycardia, whereas flecainide is highly effective in reducing arrhythmic events under the same experimental conditions.
RyR2 channel inhibition likely is the principal mechanism of flecainide’s antiarrhythmic action.
Hyperactive RyR2 channels are implicated in a variety of heart diseases associated with atrial and ventricular tachyarrhythmias. The genetic syndrome of RyR2 hyperactivity, CPVT, is caused by mutations in RYR2 or in its binding partner Casq2 (calsequestrin). CPVT is characterized by spontaneous sarcoplasmic reticulum Ca release during adrenergic stress that can trigger atrial and ventricular tachyarrhythmia. Flecainide has been clinically proven to be effective in treating patients with CPVT, but its mechanism of action remains debated. To distinguish between the 2 proposed mechanisms of flecainide action (sodium channel vs RyR2 inhibition), we synthesized flecainide analogues that retain sodium channel block, but exhibit substantially lower potency of RyR2 channel inhibition. In CPVT cardiomyocytes, flecainide remains an effective inhibitor of RyR2-mediated arrhythmogenic Ca release even when cardiac sodium channels are blocked. In mice with CPVT, sodium channel block alone did not prevent ventricular tachycardia. Hence, RyR2 channel inhibition likely constitutes the principal mechanism of flecainide action in CPVT and may also be responsible for flecainide’s antiarrhythmic action in other atrial and ventricular arrhythmias.

Footnote

Nonstandard Abbreviations and Acronyms

Casq2
cardiac calsequestrin
CPVT
catecholaminergic polymorphic ventricular tachycardia
NM-FL
N-methyl flecainide
PKA
protein kinase A
QX-FL
QX-flecainide
RyR2
cardiac ryanodine receptor
SR
sarcoplasmic reticulum
UDB
use-dependent block

Supplemental Material

File (circres_circres-2020-316819_supp2.pdf)
File (circres_circres-2020-316819_supp3.pdf)

References

1.
Priori SG, Napolitano C, Memmi M, Colombi B, Drago F, Gasparini M, DeSimone L, Coltorti F, Bloise R, Keegan R, et al. Clinical and molecular characterization of patients with catecholaminergic polymorphic ventricular tachycardia. Circulation. 2002;106:69–74. doi: 10.1161/01.cir.0000020013.73106.d8
2.
Lahat H, Pras E, Olender T, Avidan N, Ben-Asher E, Man O, Levy-Nissenbaum E, Khoury A, Lorber A, Goldman B, et al. A missense mutation in a highly conserved region of CASQ2 is associated with autosomal recessive catecholamine-induced polymorphic ventricular tachycardia in Bedouin families from Israel. Am J Hum Genet. 2001;69:1378–1384. doi: 10.1086/324565
3.
Postma AV, Denjoy I, Hoorntje TM, Lupoglazoff JM, Da Costa A, Sebillon P, Mannens MM, Wilde AA, Guicheney P. Absence of calsequestrin 2 causes severe forms of catecholaminergic polymorphic ventricular tachycardia. Circ Res. 2002;91:e21–e26. doi: 10.1161/01.res.0000038886.18992.6b
4.
Nyegaard M, Overgaard MT, Søndergaard MT, Vranas M, Behr ER, Hildebrandt LL, Lund J, Hedley PL, Camm AJ, Wettrell G, et al. Mutations in calmodulin cause ventricular tachycardia and sudden cardiac death. Am J Hum Genet. 2012;91:703–712. doi: 10.1016/j.ajhg.2012.08.015
5.
Hwang HS, Nitu FR, Yang Y, Walweel K, Pereira L, Johnson CN, Faggioni M, Chazin WJ, Laver D, George AL, et al. Divergent regulation of ryanodine receptor 2 calcium release channels by arrhythmogenic human calmodulin missense mutants. Circ Res. 2014;114:1114–1124. doi: 10.1161/CIRCRESAHA.114.303391
6.
Knollmann BC, Roden DM. A genetic framework for improving arrhythmia therapy. Nature. 2008;451:929–936. doi: 10.1038/nature06799
7.
Leenhardt A, Lucet V, Denjoy I, Grau F, Ngoc DD, Coumel P. Catecholaminergic polymorphic ventricular tachycardia in children. A 7-year follow-up of 21 patients. Circulation. 1995;91:1512–1519. doi: 10.1161/01.cir.91.5.1512
8.
Brunton, LL, Knollmann, BC, Hilal-Dandan, R. Goodman & Gilman’s the Pharmacological Basis of Therapeutics. 13th edition, ed. New York: McGraw Hill Medical; 2018.
9.
Watanabe H, Chopra N, Laver D, Hwang HS, Davies SS, Roach DE, Duff HJ, Roden DM, Wilde AA, Knollmann BC. Flecainide prevents catecholaminergic polymorphic ventricular tachycardia in mice and humans. Nat Med. 2009;15:380–383. doi: 10.1038/nm.1942
10.
Kannankeril PJ, Moore JP, Cerrone M, Priori SG, Kertesz NJ, Ro PS, Batra AS, Kaufman ES, Fairbrother DL, Saarel EV, et al. Efficacy of flecainide in the treatment of catecholaminergic polymorphic ventricular tachycardia: a randomized clinical trial. JAMA Cardiol. 2017;2:759–766. doi: 10.1001/jamacardio.2017.1320
11.
Watanabe H, van der Werf C, Roses-Noguer F, Adler A, Sumitomo N, Veltmann C, Rosso R, Bhuiyan ZA, Bikker H, Kannankeril PJ, et al. Effects of flecainide on exercise-induced ventricular arrhythmias and recurrences in genotype-negative patients with catecholaminergic polymorphic ventricular tachycardia. Heart Rhythm. 2013;10:542–547. doi: 10.1016/j.hrthm.2012.12.035
12.
van der Werf C, Kannankeril PJ, Sacher F, Krahn AD, Viskin S, Leenhardt A, Shimizu W, Sumitomo N, Fish FA, Bhuiyan ZA, et al. Flecainide therapy reduces exercise-induced ventricular arrhythmias in patients with catecholaminergic polymorphic ventricular tachycardia. J Am Coll Cardiol. 2011;57:2244–2254. doi: 10.1016/j.jacc.2011.01.026
13.
Galimberti ES, Knollmann BC. Efficacy and potency of class I antiarrhythmic drugs for suppression of Ca2+ waves in permeabilized myocytes lacking calsequestrin. J Mol Cell Cardiol. 2011;51:760–768. doi: 10.1016/j.yjmcc.2011.07.002
14.
Savio-Galimberti E, Knollmann BC. Channel activity of cardiac ryanodine receptors (RyR2) determines potency and efficacy of flecainide and R-propafenone against arrhythmogenic calcium waves in ventricular cardiomyocytes. PLoS One. 2015;10:e0131179. doi: 10.1371/journal.pone.0131179
15.
Liu N, Denegri M, Ruan Y, Avelino-Cruz JE, Perissi A, Negri S, Napolitano C, Coetzee WA, Boyden PA, Priori SG. Short communication: flecainide exerts an antiarrhythmic effect in a mouse model of catecholaminergic polymorphic ventricular tachycardia by increasing the threshold for triggered activity. Circ Res. 2011;109:291–295. doi: 10.1161/CIRCRESAHA.111.247338
16.
Bannister ML, Alvarez-Laviada A, Thomas NL, Mason SA, Coleman S, du Plessis CL, Moran AT, Neill-Hall D, Osman H, Bagley MC, et al. Effect of flecainide derivatives on sarcoplasmic reticulum calcium release suggests a lack of direct action on the cardiac ryanodine receptor. Br J Pharmacol. 2016;173:2446–2459. doi: 10.1111/bph.13521
17.
Bannister ML, Thomas NL, Sikkel MB, Mukherjee S, Maxwell C, MacLeod KT, George CH, Williams AJ. The mechanism of flecainide action in CPVT does not involve a direct effect on RyR2. Circ Res. 2015;116:1324–1335. doi: 10.1161/CIRCRESAHA.116.305347
18.
Sikkel MB, Collins TP, Rowlands C, Shah M, O’Gara P, Williams AJ, Harding SE, Lyon AR, MacLeod KT. Flecainide reduces Ca(2+) spark and wave frequency via inhibition of the sarcolemmal sodium current. Cardiovasc Res. 2013;98:286–296. doi: 10.1093/cvr/cvt012
19.
Marks AR. Calcium cycling proteins and heart failure: mechanisms and therapeutics. J Clin Invest. 2013;123:46–52. doi: 10.1172/JCI62834
20.
Wehrens XH, Marks AR. Novel therapeutic approaches for heart failure by normalizing calcium cycling. Nat Rev Drug Discov. 2004;3:565–573. doi: 10.1038/nrd1440
21.
Dobrev D, Carlsson L, Nattel S. Novel molecular targets for atrial fibrillation therapy. Nat Rev Drug Discov. 2012;11:275–291. doi: 10.1038/nrd3682
22.
Knollmann BC, Chopra N, Hlaing T, Akin B, Yang T, Ettensohn K, Knollmann BE, Horton KD, Weissman NJ, Holinstat I, et al. Casq2 deletion causes sarcoplasmic reticulum volume increase, premature Ca2+ release, and catecholaminergic polymorphic ventricular tachycardia. J Clin Invest. 2006;116:2510–2520. doi: 10.1172/JCI29128
23.
Liu H, Atkins J, Kass RS. Common molecular determinants of flecainide and lidocaine block of heart Na+ channels: evidence from experiments with neutral and quaternary flecainide analogues. J Gen Physiol. 2003;121:199–214. doi: 10.1085/jgp.20028723
24.
Banitt EH, Bronn WR, Coyne WE, Schmid JR. Antiarrhythmics. 2. Synthesis and antiarrhythmic activity of N-(piperidylalkyl)trifluoroethoxybenzamides. J Med Chem. 1977;20:821–826. doi: 10.1021/jm00216a016
25.
Hall HK. Correlation of the base strengths of amines1. J Am Chem Soc. 1957;79:5441–5444.
26.
Hall HK. Steric effects on the base strengths of cyclic amines1. J Am Chem Soc. 1957;79:5444–5447.
27.
Cannell MB, Kong CH, Imtiaz MS, Laver DR. Control of sarcoplasmic reticulum Ca2+ release by stochastic RyR gating within a 3D model of the cardiac dyad and importance of induction decay for CICR termination. Biophys J. 2013;104:2149–2159. doi: 10.1016/j.bpj.2013.03.058
28.
Hilliard FA, Steele DS, Laver D, Yang Z, Le Marchand SJ, Chopra N, Piston DW, Huke S, Knollmann BC. Flecainide inhibits arrhythmogenic Ca2+ waves by open state block of ryanodine receptor Ca2+ release channels and reduction of Ca2+ spark mass. J Mol Cell Cardiol. 2010;48:293–301. doi: 10.1016/j.yjmcc.2009.10.005
29.
Endo M, Iino M. Specific perforation of muscle cell membranes with preserved SR functions by saponin treatment. J Muscle Res Cell Motil. 1980;1:89–100. doi: 10.1007/BF00711927
30.
Potet F, Vanoye CG, George AL Use-dependent block of human cardiac sodium channels by GS967. Mol Pharmacol. 2016;90:52–60. doi: 10.1124/mol.116.103358
31.
Vaughan Williams EM. Relevance of cellular to clinical electrophysiology in interpreting antiarrhythmic drug action. Am J Cardiol. 1989;64:5J–9J. doi: 10.1016/0002-9149(89)91189-2
32.
Zsolnay V, Fill M, Gillespie D. Sarcoplasmic reticulum Ca2+ release uses a cascading network of intra-SR and channel countercurrents. Biophys J. 2018;114:462–473. doi: 10.1016/j.bpj.2017.11.3775
33.
Gillespie D, Fill M. Intracellular calcium release channels mediate their own countercurrent: the ryanodine receptor case study. Biophys J. 2008;95:3706–3714. doi: 10.1529/biophysj.108.131987
34.
Mehra D, Imtiaz MS, van Helden DF, Knollmann BC, Laver DR. Multiple modes of ryanodine receptor 2 inhibition by flecainide. Mol Pharmacol. 2014;86:696–706. doi: 10.1124/mol.114.094623
35.
Wleklinski MJ, Kannankeril PJ, Knollmann BC. Molecular and tissue mechanisms of catecholaminergic polymorphic ventricular tachycardia. J Physiol. 2020;598:2817–2834. doi: 10.1113/JP276757
36.
Watanabe H, Steele DS, Knollmann BC. Mechanism of antiarrhythmic effects of flecainide in catecholaminergic polymorphic ventricular tachycardia. Circ Res. 2011;109:712–713. doi: 10.1161/CIRCRESAHA.111.251322
37.
Batiste SM, Blackwell DJ, Kim K, Kryshtal DO, Gomez-Hurtado N, Rebbeck RT, Cornea RL, Johnston JN, Knollmann BC. Unnatural verticilide enantiomer inhibits type 2 ryanodine receptor-mediated calcium leak and is antiarrhythmic. Proc Natl Acad Sci U S A. 2019;116:4810–4815. doi: 10.1073/pnas.1816685116
38.
Priori SG, Wilde AA, Horie M, Cho Y, Behr ER, Berul C, Blom N, Brugada J, Chiang CE, Huikuri H, et al. HRS/EHRA/APHRS expert consensus statement on the diagnosis and management of patients with inherited primary arrhythmia syndromes: document endorsed by HRS, EHRA, and APHRS in May 2013 and by ACCF, AHA, PACES, and AEPC in June 2013. Heart Rhythm. 2013;10:1932–1963. doi: 10.1016/j.hrthm.2013.05.014
39.
Hwang HS, Hasdemir C, Laver D, Mehra D, Turhan K, Faggioni M, Yin H, Knollmann BC. Inhibition of cardiac Ca2+ release channels (RyR2) determines efficacy of class I antiarrhythmic drugs in catecholaminergic polymorphic ventricular tachycardia. Circ Arrhythm Electrophysiol. 2011;4:128–135. doi: 10.1161/CIRCEP.110.959916
40.
Sumitomo N, Harada K, Nagashima M, Yasuda T, Nakamura Y, Aragaki Y, Saito A, Kurosaki K, Jouo K, Koujiro M, et al. Catecholaminergic polymorphic ventricular tachycardia: electrocardiographic characteristics and optimal therapeutic strategies to prevent sudden death. Heart. 2003;89:66–70. doi: 10.1136/heart.89.1.66
41.
Laver DR, Roden LD, Ahern GP, Eager KR, Junankar PR, Dulhunty AF. Cytoplasmic Ca2+ inhibits the ryanodine receptor from cardiac muscle. J Membr Biol. 1995;147:7–22. doi: 10.1007/BF00235394
42.
O’Neill ER, Sakowska MM, Laver DR. Regulation of the calcium release channel from skeletal muscle by suramin and the disulfonated stilbene derivatives DIDS, DBDS, and DNDS. Biophys J. 2003;84:1674–1689. doi: 10.1016/S0006-3495(03)74976-5
43.
Li J, Imtiaz MS, Beard NA, Dulhunty AF, Thorne R, vanHelden DF, Laver DR. ß-Adrenergic stimulation increases RyR2 activity via intracellular Ca2+ and Mg2+ regulation. PLoS One. 2013;8:e58334. doi: 10.1371/journal.pone.0058334
44.
Gomez-Hurtado N, Boczek NJ, Kryshtal DO, Johnson CN, Sun J, Nitu FR, Cornea RL, Chazin WJ, Calvert ML, Tester DJ, et al. Novel CPVT-associated calmodulin mutation in CALM3 (CALM3-A103V) activates arrhythmogenic Ca waves and sparks. Circ Arrhythm Electrophysiol. 2016;9:10.1161/CIRCEP.116.004161 e004161. doi: 10.1161/CIRCEP.116.004161
45.
Conard GJ, Ober RE. Metabolism of flecainide. Am J Cardiol. 1984;53:41B–51B. doi: 10.1016/0002-9149(84)90501-0
46.
Sikkel MB, Francis DP, Howard J, Gordon F, Rowlands C, Peters NS, Lyon AR, Harding SE, MacLeod KT. Hierarchical statistical techniques are necessary to draw reliable conclusions from analysis of isolated cardiomyocyte studies. Cardiovasc Res. 2017;113:1743–1752. doi: 10.1093/cvr/cvx151

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.

Information & Authors

Information

Published In

Go to Circulation Research
Go to Circulation Research
Circulation Research
Pages: 321 - 331
PubMed: 33297863

Versions

You are viewing the most recent version of this article.

History

Received: 12 February 2020
Revision received: 4 December 2020
Accepted: 8 December 2020
Published online: 10 December 2020
Published in print: 5 February 2021

Permissions

Request permissions for this article.

Keywords

  1. arrhythmia
  2. calsequestrin
  3. flecainide
  4. ryanodine receptor
  5. tachycardia, ventricular

Subjects

Authors

Affiliations

Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN (D.O.K., D.J.B., C.L.E., B.C.K.).
Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN (D.O.K., D.J.B., C.L.E., B.C.K.).
Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN (D.O.K., D.J.B., C.L.E., B.C.K.).
Department of Chemistry, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN (A.N.S., S.M.B., J.N.J.).
Department of Chemistry, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN (A.N.S., S.M.B., J.N.J.).
Department of Chemistry, Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN (A.N.S., S.M.B., J.N.J.).
School of Biomedical Sciences and Pharmacy, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW, Australia (D.R.L.).
Vanderbilt Center for Arrhythmia Research and Therapeutics, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN (D.O.K., D.J.B., C.L.E., B.C.K.).

Notes

*
D.O.K. and D.J.B. contributed equally to this article.
The Data Supplement is available with this article at Supplemental Material.
For Sources of Funding and Disclosures, see page 330.
Correspondence to: Bjorn C. Knollmann, MD, PhD, Vanderbilt Center for Arrhythmia Research and Therapeutics (VanCART), Vanderbilt University School of Medicine, Medical Research Bldg IV, Room 1265, 2215B Garland Ave, Nashville, TN 37232-0575. Email [email protected]

Disclosures

Disclosures None.

Sources of Funding

This work was supported in part by National Institutes of Health (NIH) grants NHLBI R35 HL144980 (B.C. Knollmann), NHLBI R01 HL151223 (J.N. Johnston, B.C. Knollmann), NHLBI F32 HL140874 (D.J. Blackwell), T32—5T32GM007569-44 (B.C. Knollmann, C.L. Egly), NHLBI F31 HL151125 (A.N. Smith), the Leducq Foundation grant 18CVD05 (B.C. Knollmann), the American Heart Association grant 19SFRN34830019 (B.C. Knollmann), PhRMA Foundation Postdoctoral Award (D.J. Blackwell), and NSW Health infrastructure grant through the Hunter Medical Research Institute (D.R. Laver). Ca wave measurements were performed using the VUMC Cell Imaging Shared Resource (supported by NIH grants CA68485, DK20593, DK58404, DK59637, and Ey008126).

Metrics & Citations

Metrics

Citations

Download Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.

  1. Catecholaminergic Polymorphic Ventricular Tachycardia: Clinical Characteristics, Diagnostic Evaluation and Therapeutic Strategies, Journal of Clinical Medicine, 13, 6, (1781), (2024).https://doi.org/10.3390/jcm13061781
    Crossref
  2. Novel variants in TECRL leading to catecholaminergic polymorphic ventricular tachycardia , Life Science Alliance, 7, 8, (e202402572), (2024).https://doi.org/10.26508/lsa.202402572
    Crossref
  3. Management of Catecholaminergic Polymorphic Ventricular Tachycardia, Current Pediatrics, 23, 2, (63-70), (2024).https://doi.org/10.15690/vsp.v23i2.2740
    Crossref
  4. ent -Verticilide B1 Inhibits Type 2 Ryanodine Receptor Channels and is Antiarrhythmic in Casq2 −/− Mice , Molecular Pharmacology, 105, 3, (194-201), (2024).https://doi.org/10.1124/molpharm.123.000752
    Crossref
  5. The proarrhythmogenic role of autonomics and emerging neuromodulation approaches to prevent sudden death in cardiac ion channelopathies, Cardiovascular Research, 120, 2, (114-131), (2024).https://doi.org/10.1093/cvr/cvae009
    Crossref
  6. Drug Repurposing Patent Applications July–September 2024, ASSAY and Drug Development Technologies, (2024).https://doi.org/10.1089/adt.2024.126
    Crossref
  7. Backbone-Determined Antiarrhythmic Structure–Activity Relationships for a Mirror Image, Oligomeric Depsipeptide Natural Product, Journal of Medicinal Chemistry, 67, 14, (12205-12220), (2024).https://doi.org/10.1021/acs.jmedchem.4c00923
    Crossref
  8. Enhanced Ca2+-Driven Arrhythmogenic Events in Female Patients With Atrial Fibrillation, JACC: Clinical Electrophysiology, 10, 11, (2371-2391), (2024).https://doi.org/10.1016/j.jacep.2024.07.020
    Crossref
  9. Ventricular Tachycardia Due to Triggered Activity, JACC: Clinical Electrophysiology, 10, 2, (379-401), (2024).https://doi.org/10.1016/j.jacep.2023.10.033
    Crossref
  10. Structure-activity optimization of ryanodine receptor modulators for the treatment of catecholaminergic polymorphic ventricular tachycardia, Heart Rhythm, (2024).https://doi.org/10.1016/j.hrthm.2024.09.062
    Crossref
  11. See more
Loading...

View Options

View options

PDF and All Supplements

Download PDF and All Supplements

PDF/EPUB

View PDF/EPUB
Login options

Check if you have access through your login credentials or your institution to get full access on this article.

Personal login Institutional Login
Purchase Options

Purchase this article to access the full text.

Purchase access to this journal for 24 hours

Restore your content access

Enter your email address to restore your content access:

Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

Media

Figures

Other

Tables

Share

Share

Share article link

Share

Comment Response