throbber
yduosnueyJounyWd-HIN
`
`
`yduosnuewsounyWd-HIN
`
`yduosnuewJounyWd-HIN
`
`NIH Public Access
`
`QinAuthorManuscript
`
`Kreps
`
`Published in final edited formas:
`Curr Pharm Des. 2004 . 10(20): 2463-2475,
`
`Cardiovascular Side Effects of New Antidepressants and
`Antipsychotics: New Drugs, old Concerns?
`
`Pal Pacher!:2:" and Valeria Kecskemeti2”
`1 National Institutes ofHealth, National Institute on Alcohol Abuse & Alcoholism, Laboratory Physiologic
`Studies, Bethesda,MD 20892-8115, USA
`
`2Department ofPharmacology and Pharinacotherapy, Semmelweis University ofMedicine, Faculty of
`Medicines, Budapest Hungary
`
`Abstract
`
`The cardiovasculartoxicity of older generation of tricyclic antidepressants (e.g. imipramine,
`desipramine, amitriptyline, clomipramine) and neuroleptics (e.g. haloperidol, droperidol,
`thioridazine, pimozide) is well established. These drugs inhibit cardiovascular Na*, Ca?” and K~
`channels oficn leading to life-threatening arrhythmia.
`
`To overcomethe toxicity of old generation of antidepressants and antipsychotics, selective serotonin
`reuptake inhibitor antidepressants (SSRIs: fluoxetine, fluvoxamine, paroxetine, sertraline,
`citalopram, venlafaxin) and several newantipsychotics (c.g. clozapine, olanzapine, risperidone,
`sertindolc,. aripiprazole. ziprasidone, quetiapine) were introduced during the past decade. Although
`these new compoundsare not more effective in treating psychiatric disorders than older medications,
`they gained incredible popularity since they have been reported to have fewer and more benign side
`effect profile (including cardiovascular) than predecessors.
`
`Surprisingly. an increasing numberofcase reports have demonstrated that the use of SSRIs and new
`antipsychotics (c.g. clozapine, olanzapine, risperidone, sertindole, aripiprazole, ziprasidone,
`quetiapine) is associated with cases of arrhythmias, prolonged QTc interval on electrocardiogram
`(ECG) and orthostatic hypotensionin patients lacking cardiovascular disorders, raising new concerns
`about the putative cardiovascularsafety of these compounds. In agreementwith these clinical reports
`these new compounds indeed showmarked cardiovascular depressant effects in different mammalian
`and humancardiovascular preparations by inhibiting cardiac and vascular Na‘, Ca?* and K*
`channels. Taken together, these results suggest that the newgeneration of antidepressants and
`antipsychotics also have clinically important cardiac as well as vascular effects. Clinicians should
`be more vigilant about these potential adverse reactions and ECG control maybe suggested during
`therapy. especially in patients with cardiovascular disorders.
`
`The primarygoal ofthis reviewis to shed light on the recently observed clinically important
`cardiovascular effects of newantidepressants and antipsychotics and discuss the mechanism beyond
`this phenomenon.
`
`*Address Corresspondanceto these authors at the Department of Pharmacologyand Pharmacotherapy, Semmelweis University, Faculty
`of Medicine, Nagyvaradtér 4, P-O.Box 370, Budapest H 1445, Hungary, Tel: 36-1-2102930/6265; Fax: 36-1-2104412; E-mail:
`keesval(@pharma.sote.hu and NationalInstitutes of Health, NIAAA,Park Bldg., Rm. 445, 12420 Parklawn Drive, MSC-8$115, Bethesda,
`MD2 20892-8115 USA;Tel: 301-496-6777; Fax: 301-480-0257, E-mails: ppacher@lycos.com.
`
`1
`
`Exhibit 2055
`Slayback v. Sumitomo
`IPR2020-01053
`
`Exhibit 2055
`Slayback v. Sumitomo
`IPR2020-01053
`
`

`

`adiosnue;JounyWd-HIN
`
`
`
`yduosnuewJouInyWd-HIN
`WJouINYWd-HIN
`
`duosnue}
`
`Pacher and Keeskemeti
`
`Keywords
`
`Page 2
`
`antidepressants; neuroleptics; antipsychotics, QT prolongation; arrhythmia; cardiac ion channels;
`repolarization
`
`INTRODUCTION
`
`Cardiovascular mortality in psychiatric patients is high. Reports of sudden unexplained death
`in those taking psychotropic drugs, including neuroleptics and antidepressants, have raised the
`concern that part of this excess maybe duc to drug-induced arrhythmias, since manyof these
`drugs have cardiac electrophysiological effects similar to those of quinidine. Indeed, it has
`recently been established that old generation of antidepressants (tricyclic antidepressants
`(TCAs) and antipsychotics (c.g. haloperidol, droperidol, thioridazine, pimozide) can be
`associated with increased risk of cardiac arrhythmias and sudden death [reviewed in 1-7),
`
`In contrast, newgeneration of selective serotonin reuptake inhibitor antidepressants (SSRIs:
`fluoxetine, fluvoxamine, paroxetine, sertraline, citalopram, venlafaxin) and several new
`antipsychotics (e.g. clozapine, olanzapine, risperidone, sertindole, aripiprazole, ziprasidone,
`quetiapine risperidal) are considered to be free from the cardiotoxicity of their predecessors.
`However, there are increasing numberof case reports on various arrhythmias and syncope
`associated with the use of these new compounds [reviewedin: 3, 5-9) . Inadditionrecentstudies
`have demonstrated that the new SSRIs and antipsychotics also exert potent cardiovascular
`depressanteffects in various mammalian and human cardiovascular preparations byinhibiting
`cardiac and vascular Na’. Ca?’ and K* channels. This reviewis concerned with the
`cardiovascular effects of newantidepressants and antipsychotics.
`
`1. CARDIOVASCULAR EFFECTS OF ANTIDEPRES-SANTS
`
`1.1. Clinical Evidence
`
`1.1.1. Cardiovascular Effects of Tricyclic Antidepressants (TCAs)—The
`cardiovascular effects and toxicity of tricyclic antidepressants have been well documentedin
`depressed patients without pre-existing cardiac disease | 1,3, 10-11 |. The most common
`manifestation of such effect is the slowing ofintraventricular conduction, manifested by
`prolonged PR, QRSand QTintervals on the standard ECG,and othostatic hypotension j2-
`5}. The prolonged conduction can be dangerous in overdose and depressed patients with
`preexisting conduction defect and in paticnts who have already been treated with a classI
`(Na*-channel blocking) antiarrhythmic agent jl6-I 71. In overdose, delayed conduction may
`result ina complete heart block or ventricular reentry arrhythmias. Any of these complications,
`or a combinationof both, maylead to death [8-2 |. Depressed patients with conduction
`disease, particularly bundle branchblock, being treated with TCAsat therapeutic plasma levels,
`are at a higherrisk of developing symptomatic AV block than those of free from conduction
`disorders jlO-17). Tricyclic antidepressants have also been found to exert I/A class
`antiarrhythmic effects (2 I ~23), Children seemto beespecially vulnerable to cardiotoxic effects
`of high doses of tricyclic compounds, Death has occurred in children after accidental or
`deliberate overdose with only a few hundred milligrams of drug (24). Sincetricyclic
`antidepressants can cause orthostatic hypotension. induce arrhythmiaat higher doses ortissue
`concentrations, and interact unpredictably with other drugs, as do the serotonin-reuptake
`inhibitors, they must be used with great cautionin patients with cardiac disease [! I .
`
`1.1.2. Cardiovascular Effects of Selective Serotonin ReuptakeInhibitors (SSRIs)
`—The most human clinical studies with SSRIs like fluoxetine, fluvoxamin, paroxetine,
`sertaline and citalopram showedsignificant advantages over TCAsin producing fewer
`
`Curr Pharm Des. Author manuscript, available in PMC 2008 August 1.
`
`

`

`Pacher and Keeskemeti
`
`Page 3
`
`cardiotonic, anticholinergic and antihistaminergic side effects in the treatment of major
`depressive disorders [reviewed in-3}. These newercompoundsexhibited lowerrisk ofinchicg
`hypotension and a higher margin ofsafety in acute overdose thantricyclics [reviewedin -3}.
`However,it is interesting to note that the results of some animalstudies were not always so
`clear cut. For example, early preclinical studies in cats with the highly selectiveom
`reuptake inhibitor, citalopram, showed TCA-like cardiac effects at high doses|25), and the
`developmentof citalopram was delayed byreports ofcarters in dogs, eventually
`attributed to a species-specific metabolite not found in humans (2 |.
`
`The SSRI drug,ofthat the mostinformation is available, is fluoxetine [77] used for oral
`administration;it is chemically unrelated to tricyclic, tetracyclic antidepressantagents. Several
`clinical studies showed that comparedto tricyclic antidepressants, fluoxetine causes
`significantly fewereinen antihistaminergic and cardiovascularside effects
`[reviewed in: 3, 8-9|. However, even with fluoxetine one must be cautious in the conclusions
`drawnbecausethe patients that have been carefully studied are, in general, depressed patients
`free ofcardiovasculardisease, endoonly verylimited informationis available in paticnts having
`cardiovascular disease as well (28-31). The SSRIs do have cardiac effects, the best
`demonstrated of those being a mild bradycardia observed during chronic treatment with
`fluoxetine, fluvoxamin, paroxetine [reviewed in 3,88). This usually amountsto onlya fewbeats
`per minute butit is the opposite ofthe tachycardia that has beenassociated with tricyclic ies
`Analysing large number of ECG recordings from citalopram-treated patients Enemark |~32)
`reported that citalopram-treatment also reduced the heart rate. This reduction occurred within
`the first week of the treatment without further reduction later. In a small group of citalopram-
`treated patients (3—4%) with normalheart rate at baseline bradycardia was developed.
`Furthermore, citalopram treatment was associated with a non-specific, insignificant
`prolongation of QTinterval irrespective of age. In younger groupof the paticntsa statistically
`significant decrease in T-wave amplitude was also demonstrated [- 2). Moreover.there are
`increasing numberofcase reports on dyshyoa and syncope associated with fluoxetine and
`another SSRIs treatment and overdose ps 58}. A multicenter case-control study has shown
`that in the elderly the consumption of noe wassignificantly associated with an excess
`risk ofsyncope and orthostatic hypotension >91. A penton blood pressure lowering effect
`of fluoxetine was reported in DOCA-hypertensive rats (6!1, The authors suggested that a
`central action of fluoxetine on vasomotorcenter maybe responsible for the reduction of blood
`pressure, but the possible direct cardiac and/or vascular effects of fluoxetine were not excluded
`or determined.Interestingly, several recent studies have provided evidence that fluoxetine and
`citalopram directly inhibit Ca?* entry into vascular and intestinal smooth musclesresulting in
`vasodilation = intestinal relaxation, effects, which could be ofsignificant therapeutic
`importance. (° 1-64|. Surprisinglyresults from recently published retrospective studies show
`that the use of new SSRIs,similarly e the old TCAs,increasesthe risk offalls and hip fracture
`among elderly people [reviewed in 1.
`
`1.2, Cellular Electrophysiological Effects
`
`Electrophysiological studies (using a broad range ofin vitro models) demonstrated that both
`antidepressants and antipsychotics exerted their cardiac actions by modifying the different
`cardiac ionic currents during the action potential (Fig. 1).
`
`1.2.1. Cellular Electrophysiological Effects of TCAs—Inclectrophysiological studies
`on isolated mammalian multicellular cardiac preparations and single myocytes, TCAs, such as
`imipramine, chlorimipramine amitriptyline, desipramine, dibenzepin, lofepramine and
`
`amoxapine, were demonstrated to reduce the maximum velocity ofCopokatagiona of
`the action potential, an indirect indexof the fast inward sodium current, Ina.| 65-6
`Furthermore, imipramine also blocks the outward delayed rectifier K* current (Ix) is the
`
`Curr Pharm Des. Author manuscript, available in PMC 2008 August 1.
`
`
`
`|wosnueyJ
`
`id
`
` WoSnueWJOUINYWd-HIN
`
`
`
`W40uRNYWd-HIN
`
`duosnue
`
`qc
`
`

`

`Pacher and Keeskemeti
`
`Page 4
`
`inward slow Ca2* (Icq) currents in eulncsapis ventricular myocytes and transient outward
`K* current (Ito) in rabbit atrial cells {7°
`]. These direct membraneeffects explain a variety
`ofcharacteristic ECG abnormalitieslike prolongation of PQ. QRS, and QT,and cardiac adverse
`effects including tachyarrhythmias, heart block, oae nae failure, observed during
`tricyclic antidepressants treatment and overdose ji. 3 10-14, 19). The effects of imipramine
`onaction potential Suetion (APD) showimportant species peetcr In bovine ventricular
`[75 and Purkinje fibers |65|, guinea-pig papillary muscles {7 7 and isolated vee
`myocytes | /70) imipramine shortened the APD, whereas in rabbit andratatrial fibers [78“79
`it lengthened the APD. Thedifferenteffects of imipramine on APD can be explained bythe
`important differences in the ionic currents responsible for the snares among animal
`species. In guinea-pig ventricular myocytes where |, is relativelylittle (’4), the APDis
`controlled bythe interaction oeeen inward (Ix and I¢q) and outwardes (x and Tx)).
`Imipramine decreased the Ix, [8 ly, Ix andIg, but did not modify the Ix, (70» 75 |. The reduction
`of the APD in bovineaepues pig ventricular preparations could be an mainly by
`inhibition ofIca j5, 70
`1]. In contrast, in rat, rabbit and human atria (o4 80, 82) gand rat
`ventricular myocytesthe Ix is negligible and ],, appears to be the most important outward
`K’ current responsible for action potential repolarization. Thus the reduction ofI, could
`explain the prolongation of the APD observed in above-mentioned species. More recently
`several antidepressants with different chemical structures (imipramine, amitriptyline,
`naianeesine, maprotiline and trazodone) were reported to block transient outward K~ current
`(Ito) é3).
`
`1.2.2. Cellular Electrophysiological Effects of SSRIs
`
`1.2.2.1. Effects of SSRIs on Cardiac Action Potentials (APs) In vitro: Wepreviously
`demonstrated that fluoxetine elicited a concentration dependent depression of the amplitude
`ofaction potential (APA), overshoot (OS) and the maximumrateofrise of depolarization phase
`(Vmax) in multicellular ventricular kc
`tions of rats, rabbits and dogs without changing the
`resting membrane potential (RP) [s4- 5}. The significant threshold concentrations were more
`or less similar (3—10 4M)in various species (including the most sensitive isolated canine
`myocytes). Fluoxctine caused a nearlysimilar shortening of the duration of ventricular action
`potential (APD)in three species (guinea pig. rabbit, canine), but not in rats. Fluoxetine caused
`a concentration-dependentdecreasein force of contractionin rat right ventricular papillary
`muscle with a calculated [C59 value of 9.86 uM. Citalopramsimilarly to fluoxetine elicited a
`concentration-dependent (10-100 1M)reduction of Vmax, decrease ofAPA, OS and
`shortening of APA in guinea-pig ventricular papillary muscle (86). Fluoxetine and citalopram
`produced a dose-dependent decrease of V max (an indirect indicator of the fast Na’ channel
`activity), which suggests that they inhibited the activation of fast Na* channels and exhibited
`class | anti-arrhythmic effects. A possible explanation of the decrease in APA and OS and
`shortening ofthe early part of repolarization (APDso) can be the inhibition of the calcium
`current (I¢q). This latter mechanism mayalso be responsible for the negative inotropic effect
`of fluoxetine. The inhibitory effect of fluoxetine on peak Ca?* current wasprovenin voltage
`clamped canine ventricular myocytes by ICs value of 5.4 1M. This effect may cause
`lengthening ofatrioventricular conduction. Considering its Na* and Ca?* currents inhibitory
`action, fluoxetine mayhave antiarrhythmic as well as pro-arrhythmic properties (due to
`impairementof atrioventricular or intraventricular conduction). As far as the different effects
`of fluoxetine on rat ventricular APD are concerned, these can be explained bythe unique ion
`regulation characteristic to ventricular repolarization phase of rat markedlydifferent from that
`ofother mammalian species [87]. Similar cardiac clectrophysiological effects with venlafaxine
`were observedin guinea-pig cardiac myocytes (88). Thesedirect cardiac effects of fluoxetine
`and citalopramare similar to those found byus for TCA clomipramine (8) and previously
`reported for the tri- and tetracyclic antidepressants |” ].
`
`Curr Pharm Des. Author manuscript, available in PMC 2008 August 1.
`
`|wosnueyJ
`
`By
`
`osnuey|JounyWd-HIN
`
`
`
`W40UINYWd-HIN
`
`duosnue}
`
`

`

`Pacher and Keeskemeti
`
`Page 5
`
`1.2.2.2. Effects of SSRIs on Cardiac lon Channels: Previous and recent studies demonstrated
`that fluoxetine and other SSRIs —aiantagonistic properties on celaeee
`ion channels in different tissues ps4
`6). The ICsg values of SSRIs for Na*, Ca?* and
`K* channels of mainly cardiac tissues are summarized in Table 1. Fluoxetine inhibited L-type
`of Ca?*(Cay?*) currentin both rat and canine ventricular myocytes, be itspotency was twice
`as high in rat (C59 2.8 uM) than in caninesyonyite (IC59 5.4 uM) (34> 99), It is interesting
`to note that sertraline also inhibited the Ca; 2° current in rat myocytesandits inhibitoryactivity
`(I1Csg= 2.3 4M)wassimilarto that of fluoxetine, while citalopram inhibited Ca;?* current of
`guinea-pig myocytes at much higher concentration (100 wM) [25> 195). These data provide
`evidence that inhibition of cardiac Cay2* current could play an importantrole in reducing
`cardiac contractility, heart rate and atrio-ventricularconduction. The proposed mechanism may
`explain the prolonged PRinterval, AV block, hypotension, which are commoncardiovascular
`complications of fluoxetine therapy.
`
`Fluoxetine and citalopram have a highaporn (ICsg=3.1, 1.5 and 3.97 yM,
`respectively) on HERG potassium channel (9+
`5]. The Auman ether-a-go-go-related gence,
`HERG, is believed to encodethe protein, which underlies the rapid componentof the delayed
`rectifier K* current Ix,. HERG encoded I x, plays an important part in the repolarization of the
`cardiac action potential. Pharmacological inhibition ofeither heterologously expressed HERG
`or native Ix, would thus be expectedto correlate with ventricular action potential prolongation
`and associated prolongation of the QTc interval on ECG. Thus the HERGcurrent inhibition
`byfluoxetine and citalopram may give an explanation for the arrhythmogenic side effects
`(ventricular tachycardias) of these drugs. It is very important to note that this current inhibition
`can occurat nearly therapeutic levels of these drugs, thus this effect should be considered
`during the therapy.
`
`SSRIs also exhibit potent inhibitoryeffects on various voltage-dependent ion channels in non-
`cardiac tissues. Some of these effects are summarized in the Table 1, but the detailed description
`is beyond the scopeofthis review.
`
`The inhibitory concentrations of SSRIs on cardiac APs and ion current were in the upper range
`of the therapeutic plasma levels p107 |. However, it is difficult to relate in vivo plasma
`concentrations to én viiro concentrations as pharmacokinetic properties (tissue accumulation,
`metabolites) of the drug must also be considered. Undercertain conditions (c.g. incase of drug
`interactions or reduced metabolism in elderly) the plasma concentration of SSRIs can reach
`even higherlevels. Thus, a significant inhibition of various cardiovascular ion channels by
`SSRIs mayoccurin patients chronicallytreated with these compounds,resulting in certain pro-
`or arrhythmiceffects. [reviewed in ~].
`
`2. CARDIOVASCULAR EFFECTS OF NEUROLEPTICS
`
`2.1, Clinical Evidence
`
`The aim ofthis part of the reviewis to organise the available evidences on cardiac/
`cardiovascularside effects; proarrhythmic potential of antipsychotic drugs andto discusstheir
`actions on cardiac ion currents as proposed explanationoftheir proarrhythmic effects.
`
`Antipsychotic drugs represent a chemically various group of compounds. Antipsychotic drugs
`canbe classified typical (older drugs acting on dopamine Dj, D>, adrenergic a). muscarinic
`cholinergic, 5-HT> and histamine H)-receptors and associated with different side effects) and
`atypical (newerdrugs inhibiting mainly both D2 and 5-HT2,receptors and have a higher
`efficacy and fewerside effects) groups. Amongatypical antipsychotics clozapine shows
`marked esgee fromthe others binding more to Dy, 5-HTand a, receptors than to D2
`receptor. jo8). Aripiprazole is a first memberof a newclass of atypical antipsychotics have
`
`Curr Pharm Des. Author manuscript, available in PMC 2008 August 1.
`
`¥osnuey|JounyWd-HIN
`
`
`
`W40UINYWd-HIN
`
`duosnue}
`
`

`

`
`
`Pacher and Keeskemeti
`
`Page 6
`
`also unique properties showing a combinedipartialagonist activity at Dz and 5-HT|,, receptors
`with an antagonism at 5-HT2,receptors !
`Vy
`
`Antipsychotic drugs have long been knownto be associated with risk of cardiac arrhythmia
`and cardiac arrest. These arrhythmias are often reflected as changes in the electrocardiogram
`(ECG),prolongation of the QT interval, ventricular tachycardias, torsades de pointes (TdP).
`TdPisa potentially life-threatening ventricular tachyarrhythmia thatis associated with sy ncope
`and sudden death. TdP is characterized by a twisting morphology of the QRS complex around
`the isoelectric baseline and can occur in congenital and acquired form induced byvarious
`
`cardiac and non-cardiac drugs. Among antipsychotics taleperel, dro1gees pimozide,
`sertindole,thioridazine were foundto cause definitively TdP pil 2-118).
`Several other
`antipsychotics includingtypical (chlorpromazine, fluphenazine, vente.
`ocienen Ree, sultopride)| 119-12 ] (quetiapine, olanzapine, risperidone,
`ziprasidone)| 122-127 | have beenreported to prolong the corrected OT interval (corrected for
`heart rate)(QTc). Both an Australian and a Finnish study ofneuroleptic poisoning demonstrated
`that thioridazine caused the moet frequently tachycardia, prolonged QTc. widened QRS,
`arrhythmias and sudden death (220, 28). Thioridazine and Spero:wear found to be
`associated with prolongation of QTc even at dosage used for therapy [2| and basedonthis
`studythe indications ofthioridazine were cyge and droperidol was voluntarily
`discontinued by the manufacturer in UK pil4), Pimozide, sultopiride and droperidole also
`prolong QTc ehpeat have been associated with TdP and sudden death, but far fewerdata
`areavailable jl2
`30), The high-potency drug ena can prolong QTc interval, causes
`TdP and sudden death at normal therapeuticdoses [*” I), but the frequency by which these
`effects occuris less than with thioridazine | 1288). Similar cardiovascular risks of traditional
`antipsychotics used at therapeutic dosage were published in the USA i-a retrospective study
`investigating 481.744 persons (aged 15-84 years, from 1988 to 1993) [}32}.
`
`The newatypical antipsychotics have greater efficacy and fewerside effects than older
`neuroleptics and with the exceptions of sertindole and ziprasidone they have not caused
`consistentoesignificant lengthening ofQT or sudden cardiac death at therapeutic
`concentrations il
`3]. Sertindole has becn provento be associated with a QT prolongation
`at therapeutic concentrations it 17, 124) and bothincreasing evidence of unexplained sudden
`cardiac death and serious arrhythmias found by the Committee on Safety ofMedicine imane
`United Kingdomresulted in a voluntary withdrawal of the drug by the manufacturer ps4).
`Albeit the known correlation between schizophrenia and increased cardiovascular mortalityit
`ma oe to estimate the sudden death due to particular neuroleptics at therapeutic doses
`| 135, 1 36), Clozapine beyond the well-known agranulocytosisrisk, is being associated with
`myocarditis, cardiomyopathyand arrhythmogenesis risk 137. 138 He also reduced measures
`of heart rate variability associated with parasympathetic control jl24]. In the study of
`overdoses, clozapine overdose was associated with sinus tachycardia (more than 66% ofthe
`patients) anaesit the case of risperidone overdose more than 66% ofthe patients were
`asyReet (39. However,there are data suggesting that risperidone could cause sudden
`death pl25}. Neither olanzapine nor quetiapine had been implicated in cases of TdP or sudden
`death.
`
`Ziprasidoneis a newatypical drug with less side effects and in comparison with olanzapine
`and risperidone it does not appear to cause weight gain, hyperlipidemia and hyperglycaemia
`
`(12 rolongs the QT interval more than haloperidol, olanzapine, quetiapine and risperidone
`27). Althoughit was not associated with cardiac events during premarketing trials the
`ene ofunexpected life-threatening arrhythmias can not be excluded when the drug
`
`enters widespread use.
`
`Curr Pharm Des. Author manuscript, available in PMC 2008 August 1.
`
`

`

`Pacher and Keeskemeti
`
`Page 7
`
`2.2. Mechanism of the Lengthening of QTInterval
`
`The QTinterval includes both depolarization and repolarization. Q waverepresents the onset
`of ventricular depolarization, while T waveis the sign of the repolarization. Because the QT
`interval shortens with increasing heart rates, it is usually corrected for heart rate (QTc).
`Depolarization ofventricularcells is the result ofa rapid influx of sodium ions through selective
`Na’ channel and its duration measured by the QRS interval. Repolarization involves calcium,
`sodium, and different potassium channels. Whereas the participation of these ion channels in
`the repolarization is highly dependent on species, mainly potassium channels are responsible
`for this parameter. Concerning the specificity of QT prolongation as a markerof an effect on
`cardiac repolarisation, it should be kept in mind that the duration of the QT interval maybe
`affected byboth the velocity of repolarisation and ventricular conduction velocity. Class I
`antiarrhythmics as sodium channel blockers, decrease ventricular aia velocity, cause
`widening of QRS complexand therefore lengthen the QT interval (491. Similar action can be
`observedin the case of tricyclic antidepressants which by blocking Na” as well K* channels
`widen both the QRS and the QTc. The potassium channels (among them I,) are most often
`involved in drug-induced QT prolongation and TdP. Drug!Lacking the IKrchannel can induce
`QTprolongation and TdP and sometimes sudden death ji+ y. However. there is no close
`correlation between QTc interval prolongation and occurrence of TdP. Notall drugs that
`prolong the QTc interval produce TdP. Amiodarone,a class III antiarrhythmic drug, produces
`marked prolongation of QTc interval but docs not evoke TdP. The calcium-channel blocker
`verapamil has been alown to prolong QTinterval in a mannerthatis linearly correlatedto its
`spc concentration [!42} but there are fewdescribed casesofverapamil-induced TdP
`it 43), Noclear-cut dose-dependencycanalso be observed for QT prolongation or occurrence
`of TdP. In some cases the QT prolongation and occurrence of TdP is dose dependentbut these
`parameters can also be observed at normal plasma levels ofdrugs, too ji44). Inthe latter several
`factors (hypokalaemia/magnesaemia, mutation of K* channels) reducing the repolarization
`eh=a given subject greatly increase the proarrhythmic potential of relatively low plasma
`level [?>
`‘].
`
`The link between the lengthening of QT interval and TdP is seemingly very complex and
`affected byseveral factors including electrolyte imbalance, age, gender, disease (myocardial
`ischemia, infarction, hypertension, hypothyroidism, diabetes, renal or hepatic dysfunction) and
`concomitant medications.
`
`2.3. Cellular Electrophysiological Effects of Neuroleptics
`Most ofantipsychotics are generally lengthenthe action potential duration (APD)and inhibit
`the rapid componentof the delayed rectifier current (I,,) but some of the typical antipsychotics
`including ipo andpagent, beyond their inhibitory effect on K~ current inhibit
`also Na* and Ca?* channels (8 1). Such effects could be antiarrhythmic orcardiotoxic,
`depending on the health (e.g. post myocardial infarct) of the myocardium. Thenet effect on
`APDofantipsychotics depend on the overall balance between inward and outward currents
`during the plateau phase of AP andtheir relative sensitivity to the particular agent in question.
`Table 2. summarizes the inhibitory potencyof antipsychotic drugs on K“(Ik. Ito. Ik. HERG)
`and other ion (Na”, Ca") currents. In humanIx,is carried by the humanether-a-go-go (HERG)
`K* channel, which can be expressed in homologousand heterologouscells in orderto assess
`the potency (ICs9) of a drug in inhibiting this channel. Haloperidol and droperidol prolong
`APDinguinea-pig ventricular myocytes and inhibit Ix, and HERG with ICs9 values of 20
`nM-1.36 uMand 32.2 1M,respectively and the effects of haloperidol on eS are overfive
`aoeenty times more potent than its effects on Iy, and Icy), respectively (3!» 145-146, 150,
`63}. Thioridazine also lengthened APD in guinea-pig ventricular myory-ae potently
`inhibited I, and HERG (IC«q values of 1.25 uM, 191 nMand | yM) [!47-
`!49), Comparative
`
`Curr Pharm Des. Author manuscript, available in PMC 2008 August 1.
`
`¥osnuey|JounyWd-HIN
`
`
`
`W40UINYWd-HIN
`
`duosnue}
`
`

`

`
`
`Pacher and Keeskemeti
`
`Page $
`
`study showed that neweratypical antipsychotic ziprasidone, olaseapine, risperidone block
`HERGandIx; ina more orless similar concentration range (}
`if
`
`Figures (2 and 3) showthat risperidone concentration-dependently increased APDin both
`guinea-pig ventricular muscle (Fig. 2A) and canine ventricular myocytes (Fig. 3B). This effect
`was most prominent on terminal phase of repolarization (APDog) (with ECso values of 0.29
`uM and 0.48 iM in guinea pig and canine myocytes,respectively) (Fig. 3C) and showed reverse
`tate dependence (Fig. 2B). Haloperidol had similar effect on APD (Fig. 2C) but reduced also
`the maximum velocityofdepolarization (V,,,,)(indirect indicator ofNa* channelactivity)(Fig.
`2D) while risperidone was ineffective on this parameter. We found thatrisperidone
`concentration-dependently inhibited Ix, with an IC59 of 0.92 4M and practically had no effect
`on the other K‘ currents(1, with ICs >10 wM,Ixy with ICsg >100 uM) [!51}. Similareffects
`of re on both APD andIx,in rabbit ventricular myocardium and myocytes were
`observed | 152lwme lowerICsgvalues (167 and 261 nM, respectively) were found in HERG
`channelbyothers | 147, 155),
`
`Sertindole was found to be a high affinity antagonist of the human cardiac K' channel HERG
`(ICsg= 3 and 14 nM)but wasless active at blocking other K* currents (Kv 1.5, Ijo with
`ICso=2.1 and 10 uM, respectively) [147. 148, 153],
`pimozide potentlyinhibited cardiagHERGK*channel (IC59 values of 18 and 174 nM) ie
`oe the inhibitory action onHERGchannel sertindole and pimozide also blocked the
`30), increased the risk of TdP pls4 and also blocked Ica; in rat ventricular myocytes[ 56|.
`human brain K* channelerg3. Sertindole blockeder;ef 3 channel currents with an ICs9 of 43
`nM, while pimozide had an ICs9 value of 103 nM il 7]. It was suggested that this inhibition
`of erg3 related K* channels in the brain might contribute to their cfficacy/side effect profiles.
`
`Comparing the HERG channel inhibitory activity of seven antipsychotics drugs (olanzapine,
`pimozide. quetiapine. risperidone, sertindole. thioridazine, ziprasidone)to their binding
`affinities for Dy and 5-HT}, receptors the following selectivity rank was found: olanzepine >
`risperidone > ziprasidone > thioridazine > pimozide > sertindole. Sertindole and pimozide had
`the highest HERG channelinhibitory activity, while the lowest inhibitory activity can be
`observed in the case of olanzapine and quetiapine. These results also showedthatsertindole,
`pimozide, thioridazine displayed little or no selectivity for dopamine Dz or 5-HT2, receptors
`relative to their HERG channelaffinities, and olanzapine had the greatestselectivity for
`dopamine D, and 5-HT>, receptor binding compared to the HERG channel. In the case of
`quetiapine the selectivity was not calculated due to its lack of affinity for the dopamine D>
`recepm,Esamainien the relationship between plasma levels and QT prolongation for these
`drugs ji27 the authors also found a good correlation between ae)ang oftotal plasma drug
`concentration to HERG ICs, and their QTc prolongation effect i+7. Based uponthis in
`vilro results drug’s selectivity (between their target receptoraffinity and their HERG channel
`ICs value) seems to be a predictive factor for appearance ofQT prolongation in clinic. Itwould
`be expected that olanzepine and risperidone, displaying highselectivity, would havethe least
`potential to produce QT prolongationin clinical settings.
`
`However,it is widely accepted that most QT prolonging drugsinhibit Ix, but the potency of
`drug-induced block of Ix, does not showa clear correlation with the risk of QT prolongation
`and moran ofTdP.In isolated feline hearts haloperidol prolongs QT interval more than
`sertindole pi59}. while sertindole is 10-300 times more potent as a blocker ofHERGthan
`haloperidole (Table 2). In addition, some aoeeee that thioridazine and chlorpromazine
`have similar potenciesfor inhibition of HERG pis8) and in stepwise regression analysis of 495
`beyctentpatients thioridazine wasassociatedwithQT prolongation, butchlorpromazine was
`not| 128|. These d

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket