Pharmacology D.

Michael

Classified

MD, FCP, and

Freedman,

is a unique

Amiadarone

antiarrhythmic

electrophysiologically

antisympathetic orally

and Pharmacokinetics of Amiodarone

and

as direct,

administered

fast

amiodarone

agent a Type

John

originally

negative

developed

111 antiarrhythmic,

channel-membrane (a

C. Somberg,

inotropic vasodilatian,

as

it also effects. agent)

MD,

has

a

both

vasadilator.

nonspecific

Hemadynamic are

usually

FCP

effects negligible,

of

and

are usually compensated for by induced Effects an thyroid and hepatic function may help to explain same of the unique pharmacologic as well as toxicologic effects of the drug. Amiodarone is poorly bioavailable (20-80%) and undergoes extensive enterohepatic circulation before entry into a central compartment. The principal metabalite, mono-n-desethyl amiadarone is also an antiarrhythmic. From this central compartment, it undergoes extensive tissue distribution (exceptionally high tissue/plasma partition coefficients). The distribution half-life of amiadarone out of the central compartment to peripheral and deep tissue compartments (tl/2,,) may be as short as 4 hours. The terminal half-life (t#{189}0) is both long and variable (9-77 days) secondary to the slow mobilization of the lipophilic medication out of (primarily) adipocytes. A phormocokinetically based loading scheme is described, and data suggesting a role for routine amiadarane plasma levels ore presented.

A

miodarone HC1 (2-butyl-3-benzofuranyl 4-[2(diethylyamino)-ethoxy]-3-5-diiodophenyl ketone HCL, molecular weight = 681.8) was developed by R. Charlier at Labaz Laboratories in Belgium.1. It is a di-iodinated benzofuran derivative, containing a diethylated tertiary amine chain. Originally recognized to be an antianginal agent (1967),2 within 2 years its antiarrhythmic abilities had become appreciated.3 Early investigations were primarily confined to Europe and South America, where the use of amiodarone as an antianginal and antiarrhythmic gained widespread acceptance.4’5’6 The pharmacokinetics of amiodarone and its metabolites are unlike those observed with any of the currently available antiarrhythmics (of any class). This is reflected by early clinical reports using amiodarone in which the number of dosing regimens appear to be only surpassed by the number of investigators. Although amiodarone has been existent since the early 1960s, an understanding of its biological/ metabolic fates and kinetics has emerged only within the last decade. This knowledge can be put to sound clinical use to improve our understanding of

appropriate loading (acute care) and maintenance therapy. Amiodarone is available in both oral and parenteral preparations. However, only the oral preparation (200 mg/tablet) has been approved by the Food and Drug Administration for use in the U.S. (Cordarone, Wyeth Laboratories, Philadelphia, PA). Insoluble in aqueous solutions, it is miscible in polysorbate-80 (Tween 80) and either ethanol or benzyl alcohol. An intravenous form (40 mg/mL), while available in Europe (Cordarone-X, Sanofi, Pharma S.A., Paris), remains in clinical trials in the U.S. This article focuses on the orally available form. The physical characteristics of pharmacology are outlined in Table I. This section serves as an outline to amiodarone pharmacology and assists in the conceptualization of amiodarone pharmacokinetics, rather than a definitive treatise on that subject. PHARMACOLOGY Electrophysiologic

From

the

New

York

Medical

College

(Dr. Freedman), Valhalla, New Division (Dr. Somberg), Chicago

York, and the Clinical Pharmacology Medical School, Chicago, Illinois.

Address

berg, MD. 3333

N. Chicago,

J COn Pharmacol

Green

Bay Road,

1991;31:1061-1069

for

reprints: IL 60064.

John

C. Som-

At a fundamental onstrated an demonstrated

Effects

(basic) level, amiodarone has antifibrillatory effect. This has in canine ventricular fibrillation7

dembeen and 1061

FREEDMAN

feline atrial fibrillation.8 Amiodarone is further characterized as possessing Vaughn-Williams “Class III” properties.91#{176} Depressing impulse initiation in the isolated rabbit sinus node,11 amiodarone appears to both prolong repolarization and refractoriness in all cardiac tissue.12 Sinus cycle length, (monophasic action potential and surface QT interval). AV nodal conduction time, His-Purkinje, and ventricular repolarization times are generally increased with increased refractoriness.13’14 Additionally, amiodarone appears to depress conduction velocity greater at higher rates of depolarization (repolarization time 30% greater after 6 weeks of therapy),15 suggesting that it (directly or indirectly) blocks inactivated cardiac sodium channels.16’17 Pathologic conduction is also slowed. In patients with Wolff-Parkinson-White syndrome, the anterograde effective refractory period (ERP) of the accessory pathway is lengthened.18 Amiodarone exerts antisympathetic effects to both alphaand beta-adrenergic responses after sympathetic stimulation or catecholamine injection.19 Additionally, amiodarone dissolved in ethanol noncompetitively antagonizes the chronotropic response of rabbit atria to isoproterenol and inhibits norepinephrine-induced contractions of myocardial strips in rats and rabbits. Table II summarizes the electrophysiologic actions of amiodarone. Hemodynamic

Actions

Hemodynamically, the weakly negative inotropic activity of amiodarone is usually more than offset by concurrently acting vasodilator effects.2#{176}Generally there is little change in either ejection fraction21 or systemic blood pressure after oral administration. It has been reported, however, that in patients with

AND

SOMBERG

TABLE Electrophysiologic

Amiodarone Physical

Heart

Activity

characteristics =

Pharmacology

Metabolic

oral absorption fate:

minimal,

principally

Extremely lipophilic Nonspecific antiadrenergic effects Direct membrane effects Class III antiarrhythmic Sodium/calcium channel-blocking

1062

S

J CIin Pharmacol

rate

RR PR

QRS QT

Observation Slowed Prolonged Prolonged Unchanged/prolonged

PA

Lengthened Prolonged

AH Atrial ERP* HV conductiont

Prolonged Prolonged Prolonged

AV nodal ERP AV nodal FRPI: Ventricular ERP

Prolonged Prolonged Prolonged

AVNW U waves

Prolonged Often present,

#{149} ERP = effective refractory period. f His.ventricular. t FRP = functional refractory period. § Atrial pacing cycle length producing Wenkebach. From: Zipes DP, et al, Singh BN, Vaughan Williams Zipes DP et 81,22 and Harris L.

merging

into

I

EM,1#{176} Rosen MR, eta!,12

severely decompensated left ventricular function, critically dependent on augmented sympathetic tone, that the nonspecific beta-blocking effect of amiodarone may be deleterious.15’22 (NB: The aforementioned summary of amiodarone’s hemodynamic actions are in contrast to those observed when the intravenous material is administered. In both experimental animals and humans administered IV amiodarone, significant decreases in systemic blood pressure and total vascular resistance are accompanied by negligible increase in cardiac output that may last 15 minutes or longer after injection.) Pharmacologic

Actions

I

Molecular weight 681.8 Structure: dilodinated benzofuran derivative Highly protein bound, (albumin, /3-lipoprotein) Incomplete

Effects

Parameter

Other TABLE

II

metabolized

actions

1991;31:1061-1069

by the liver

A potent inhibitor of thyroxine 5’deiodinase, amiodarone inhibits the peripheral formation of 3,5,3’,5’tetraiodothyronine (T4) to 3,5, 3’-triiodothyronine (T3) causing a doseand duration-dependent increase in serum reverse T3.23’24’25 This action suggested to Singh that this might be a primary mechanism of action1#{176}since changes produced by amiodarone were similar to those seen after thyroid ablation.26 Serum reverse T3 levels were initially used as an alternative to measuring plasma levels of amiodarone directly (since it was believed to be linearly drug dependent and correlate with therapeutic response as well as with the rate of adverse reaction formation). This test has been largely precluded by

PHARMACOLOGY

AND

newer (widely available and accurate) HPLC assays directly measuring plasma drug levels. In addition to inhibition of T4-T3 converting enzyme, amiodarone use has also been associated with multiple aberrations of thyroid function (hypoand hyperthyroidism). Although it is believed to be directly dependent on the pharmacology of amiodarone, both hyperand hypothyroidism could result directly from the supratherapeutic iodine load associated with amiodarone therapy (approximately 75 mg organic iodine/tablet). Amiodarone use has also been associated with elevation in serum transaminasis and appears to induce a phospholipidosis and consequent fibrosis within the liver.27 A similar, systemically occurring phospholipidosis (characterized by foamy macrophages and lamellated inclusion bodies) has been observed in other tissues (mesenteric lymph nodes and lungs) after prolonged administration of the drug to experimental animals.28 The alveolitis observed (also characterized by foamy macrophages) as an adverse reaction with amiodarone may be consequent to this pharmacologic action(s). PHARMACOKINETICS Bioavailability, and Metabolism

AND Tissue

METABOLISM

Distribution,

Amiodarone has been shown to have variable (2080%) oral bioavailability.29’3#{176} After absorption, the drug undergoes extensive enterohepatic circulation before distribution into the central compartment and subsequently to tissue compartments. Examination of hepatic and portal vein samples after oral administration of amiodarone indicates a large first-pass (de-ethylation) effect resulting in mono-N-desethyl amiodarone. Peak amiodarone serum levels after oral dosing are achieved within 3-7 hours. Amiodarone is highly protein bound (96_99%),3132 predominately to albumin, but also to 3-lipoprotein.33 (Figure 1) Hepatic metabolism has been demonstrated to be partially the result of a P-450 cytochrome oxidasedependent oxidative de-ethylation. Both phenobarbita134 and 3-methyl cholanthrene35 have been established as inducing agents for this reaction(s). MonoN-desethyl amiodarone, the primary de-ethylated metabolite formed, has also been recognized to have antiarrhythmic abilities.36 A second dealkylation may then occur (N-dealkylation), resulting in the primary amine. Other metabolic pathways shown to exist include deiodinations (monoand di-), 0-dealkylation, and hydroxylation.37 Finally a glucuronidation reaction has been found to be present, proba-

LV DYSFUNCTION

AND ARRHYTHMIAS

OF AMIODARONE

PHARMACOKINETICS

bly accounting for the major mechanism of amiodarone clearance. Renal clearance is negligible, and the dose of the drug is not reduced in patients with renal failure or those undergoing dialysis.38 An outline of the aforementioned metabolic pathways is found in Figure 2. Comprehension of amiodarone pharmacokinetics further requires a cognizance of its tissue distribution. Tissue distribution also explains the particular and characteristic toxicity of the drug. Fortunately, by using refined HPLC techniques, both plasma and tissue drug levels can be evaluated.39’40 Tissue distribution of amiodarone and its active metabolites have been studied in both animals and humans. Highest amiodarone and mono-N-desethyl amiodarone levels have been found in adipose tissue, lung, liver, and lymph nodes (deep compartments). Lowest levels have been found in brain, thyroid, and muscles (peripheral compartments).4142’43 The tremendous lipid solubility of amiodarone is expressed by an estimated log P0 = 6.66, for octanol/ water partition of the neutral (non ionized) form. Amiodarone myocardial concentrations, determined in patients who are undergoing coronary artery bypass grafting, were found to be higher in the cardiac tissue than in the plasma,45 with a partition coefficient (PC) of 35. The partition coefficient for the metabolite mono-N-desethyl amiodarone has been demonstrated to be 91 (2.6 x greater than for the parent molecule), and thus is even more lipophilic than its precursor. Initial

Drug

Distribution

Because of the differing solubilities of the drug within many tissues (characterized by the differing partition coefficients), amiodarone pharmacokinetics are usually described using multi-compartmental models (Figure 1). In the simplest of these models, the tissue compartments are distinguished as being central, deep, or peripheral depending on the solubility of amiodarone within that space. With a limited volume of distribution (Vd) and restricted solubility, serum levels in the central or plasma compartment increase rapidly after absorption. Drug levels in the other more poorly perfused tissue compartments (both deep and peripheral) with large Vds associated with relatively large drug solubility would be expected to rise at a much slower rate. Similarly, total tissue load would decrease at a slower rate in the tissue (peripheral and deep) compartments as described by the rate constants (K4.K7.K6 Figure 4) and prolonged t#{189}8. Early information on pharmacokinetic parameters was obtained using radiolabeled material bY Broek-

1063

FREEDMAN

SOMBERG

AND

BIO*ILABILITY

30-80%

ABSORPTION

HEPATIC K1

METABOLISM

CENTRAL

BILIARY

COMMRTMENT

KhiIIIIiJ ‘(4

PERIPHERAL

K6\

COMRTMENT

/

> MED.

GOLUSILITY

COUP..

REL

LARGE

Vd)

EXCRETION

/

DEEP NIGH

COMRTMENT SOLUSILITY

c9IF.

bIa

K8

Figure

1. Amiodarane

S

.1 Clin

Vd

AdIpo.#{149}

#{149} Ilvsr

pharmacakinetics.

huysen in 1969.46 After administration of radiolabeled amiodarone to humans the kinetics of (1251 and 14C)-amiodarone were evaluated (whole body radioscintigraphy). Although the first elimination (distribution) phase was found to be variable from one subject to another, the importance of this phase progressively decreased with the term of therapy.47 After single-dose administration of 200-mg amiodarone, Anastasciou-Nana evaluated several pharmacokinetic parameters using drug levels obtained by HPLC and found an apparent t#{189}of 18.7 ± 4.0 hours. Riva, administering 400 mg (single dose study), noted a t#{189} of 35.9 ± 38.0 hours.48’49 Haffajee et al. also measured serum amiodarone levels after administration of a single 800-mg dose of amiodarone in eight patients and found wide interpatient variability with a mean t#{189}=4.62 hours.5#{176}The apparent half-lives observed in these acute models represent the distribution (a phase, t#{189}a) into tissue compartments, in which amiodarone is sequestered. (Table ifi) Since poorly perfused compartments, in particular the deep compartments, represent areas with much greater affinity for amiodarone (denoted by their partition coefficients), they represent enormous “sinks” or reservoirs for amiodarone storage.

1064

LARGE

Pharmacol

1991;31:1061-1069

Elimination

(Terminal)

Kinetics

The existence of deep compartments, their respectively enormous distribution volumes (Vds), and slow individual clearance(s) has been demonstrated in several studies. Holt3#{176}and other investigators51’52 have shown that the (terminal) elimination pharmacokinetics of amiodarone appear to be first order, with a linear relationship between the amount of drug administered and the steady-state plasma level (Table IV). When 400 mg orally per day (typically a therapeutic dose) is delivered, the steady-state plasma concentration of amiodarone was shown to be 1.93 ± 0.80 mg/L and the serum concentration of mono-N-desethyl amiodarone was 1.79 ± 0.65 mg/L (Holt et al.). Studying plasma concentrations from six healthy volunteers after intravenous injection of 400 mg amiodarone, Holt found plasma concentrations were best fitted to a multicompartmental (multi-exponential function) model with a distribution t#{189}a of 4 minutes (consistent with earlier data from Haffajee et al.)41 and elimination (terminal, 3 phase) t#{189}$ of 24.8 ± 11.7 days53 (single dose, IV administration). The total body clearance was 8.6 ± 1.9 mL/min and the total body Vd of amiodarone at steady state was 4936

PHARMACOLOGY

L

AND

OF AMIODARONE

PHARMACOKINETICS

C2HS

/ -O-CH2CH2H C2HB

IIjC4HS

H

O-DEALKYLATION

/-O-CH2CH2N C2HE

DE ODINATI

/ \ C 2H6

C4H9

N-DEALKYLATION

AMIODARONE HYDROXYLATION C2HE

PH

-0 -C H2CH2N

OH

I/

C4H9

\ C2HS

IIXIJC4HS H2 C H 2 N\ C 2H6

-0-C

\

I

C2H6

,

A /

-O-CH2CH2NHE

5;o

/ O-CH2CH2N

\ OH

Figure

2. Major

metabolic

pathways-amiodarone.

± 3290L. This investigator also carried out an analysis of plasma concentrations from eight patients who withdrew from chronic oral amiodarone therapy. Under these conditions, amiodarone t#{189}fl=52.6± 23.7 days and mono-N-desethylamiodarone t#{189}$= 61.2 ± 31.1 days. Differences between the results of the volunteers and the patients were suggested to be secondary to an underestimation of projected steady-state plasma levels in the computerized simulation (volunteers), as well as to a time-dependent pharmacokinetic factor(s). Other investigators have similarly documented lengthy terminal half lives (t#{189}fl)after cessation of maintenance therapy (Table III). Differences in both the rate of acquisition of amiodarone into body stores (range: t#{189}fl = 9-77 days) and subsequently the required loading dose(s) address the tremendous interindividual variability present in the treated population.

ASSESSMENT AMIODARONE

OF THE RESPONSE ADMINISTRATION

Optimization of amiodarone a rapid assessment of the

LV DYSFUNCTION

C4HS

C 1H5

ri

administration response of the

AND ARRHYTHMIAS

TO

requires myocardial

conduction tissues to appropriate therapy. Appropriate therapy is generally reflected by establishing that certain plasma levels (therapeutic window of activity) have been attained. Originally it was hypothesized that the steady-state serum levels of amiodarone did not require monitoring since a known therapeutic range could not be established for this drug,54 as measured by plasma levels.

TABLE Terminal

Half- Life (t112)

III

After

Oral Administration Terminal

Number Studied

Amiodarone

et al.

8

53 ± 24

Marchiset, et al. Haffajee, et al. Kannon, et al. Harris, et al.

12 3

47 ± 13 19 ± 9 39 ± 17 @52

Holt,

From: a!.

,85

Holt OW et and Harris et al.

4

70 Marchiset

D et 81,83

Haffajee

Half-life

(d)

Monodesetylamiodarone

61 ± 8 54 ± 23

@60 et al.,#{176}Kannen

et

1065

FREEDMAN

TABLE Plasma

Levels

Dose Administered

IV

at Varying

Chronic

Mono-Desethyl. Amiodarone (mg/I)

Amiodarone (mg/I)

n

Dosages

200 mg/day Holt

et al.’#{176}1

400

1.45 1.1

± 0.43 ± 0.50 ± 0.30

1.04 1.38

1.93 3.05 2.2

± 0.80 ± 1.36 ± 0.9

1.79 2.33

3.46 3.78 3.9

± 1.50 ± 1.61 ± 1.6

2.79 2.92

1.06

Heger et al.55 Bopanna et al.

± 0.34 ± 0.74 -

mg/day

Holt et al.5’ Heger et al.55 Bopanna et al. 600 mg/day Holt et al.’8 Heger et al.55 Bopanna et al. From:

Halt et al.,42 Heger

et a!.

52

and Bopanna

± 0.65 ± 1.22 -

± 1.17 ± 1.40 -

et al.8’

As experience has been gained, a vast amount of contradictory evidence, supporting as well as mitigating the role of tissue and plasma levels of amiodarone, has been published. One of the primary difficulties has been the standardization of methodologies employed to determine drug level and outcome events (both efficacy as well as toxicity events). Essentially the three criteria of efficacy that have evolved are acute surface electrocardiographic response, response to programmed electrical stimulation, and response during intermittent subchronic surface electrocardiography (Holter monitoring). The first evidence of efficacy were obtained utilizing acute surface electrocardiographic confirmation of arrhythmia suppression. Early studies and reviews by Horowitz55 and Haffajee5#{176} suggested that the antiarrhythmic effect of amiodarone does not correlate to any degree with plasma level. Others have shown that amiodarone plasma levels (1.0-1.5 mg/ L)56’57’58’59 have been associated with a decrease in the amount of ventricular ectopy, and that doses in excess of 2.5 mg/L do not appear to provide any additional antiarrhythmic benefit. Efficacy of amiodarone has also been determined by prevention of ventricular arrythmia induction during programmed electrical stimulation. The predicative value of inducibility (as an indicator of efficacy) has been noted to range from poor60’61’62’63 to reasonable (predictive in 50-75% of patients).TM’65 Difficulties in deriving efficacy data using programmed electrical stimulation are probably secondary to varied stimulation protocols as well as times of testing. Many authors have confirmed that patients

1066

#{149} J COn Pharmacol

1991;31:1061-1069

SOMBERG

AND

treated with amiodarone frequently remain inducible and that furthermore, inducibility does not (necessarily) predict a bad outcome.TM67SSTM6970 The Collaborative Group for Amiodarone Evaluation71 analyzed the plasma amiodarone levels in patients who were considered to be responders (based on Holter monitor results) and found no differences between responders and nonresponders. Kim et al.72 found that the predictive value of Holter monitoring was low in several studies, and that suppression did not guarantee a good outcome. Nevertheless, even in these groups, patients who appeared to be responders (in terms of ventricular arrythmia) generally had plasma levels in the range of 1.0 to 3.5 mg/L, whereas plasma levels of 2.5 mg/L are associated with increased neurologic and gastrointestinal adverse reactions.5#{176} Additionally, a mass of literature supports the hypothesis that the risk of pulmonary toxicity, ocular changes, and cutaneous changes increases in a dose-duration-dependent (non-acute, and probably not serum-level related) manner, becoming particularly obvious at higher daily doses over increased lengths of time (>500mg/d).50’76 Therefore, plasma levels of amiodarone appear to be most useful during the acute stage of administration for determination of which patients are receiving either too little drug (2.5 mg/L) without attendant increase in efficacy. In this way, a therapeutic window has been defined based on maximization of acute risk/benefit ratio. This definition has allowed for a more rational approach to initiation and maintenance of therapy, based on drug levels, and hopefully an individualization of therapy. INITIATION MAINTENANCE

OF

THERAPY DOSING

AND

Loading doses used when chronic therapy is contemplated, have largely been developed empirically. Given the lengthy t1,,2, both the amounts used and the time required for appropriate loading become easier to understand. Early in its history, oral doses as

large

as several

for up to I week to maintenance

grams

before doses

per

day

were

administered

the patient was switched of 400 or 200 mg/day.77

over One

PHARMACOLOGY

AND

(more) rational oral dosing schedule (based on pharmacokinetic modeling) for the treatment of ventricular arrhythmias has been proposed by Siddoway et al.78 In this schedule, 2,000 mg is administered on day 1. This is followed by 3 days of 1400 mg/day and then 7 days of 1,000 mg/day. 800 mg/day is given for another 1 to 2 weeks before the dose is reduced to 400 mg/day. Using this program, a total loading dose of 6,200 mg is delivered. In the case of treatment of supraventricular arrhythmias, some authors have suggested the aforementioned regimen be reduced 25 to 50% (1,000 mg/day x 1 day, followed by 700800 mg X 3 days then 7 days of 600 mg/day followed by maintenance of 200-400 mg/day) because of the increased sensitivity of supraventricular arrhythmias to amiodarone.79 Alternatively, a loading dose schedule developed by Ward et al.8#{176} allows a total loading dose as low as 1,200 mg administered as 600 mg/d X 7 days followed by 400 mg/day X 7 days was also found to be effective. Using either regimen antiarrhythmic effect is extremely variable. After loading, it is only necessary to administer the chronic therapy of 200 to 400 mg per day to achieve therapeutic efficacy. Many authors believe that 400 mg is in fact the maximum “prudent” dose that should be used for prolonged periods.81 Terminal half-lives after cessation of prolonged oral therapy are found in Table III. CONCLUSIONS The riddle of ariiodarone pharmacokinetics is slowly being elucidated. They are in fact enigmatic of a pharmacokinetically unique drug. One with multiple metabolic fates, each of which probably has multiple modes of action. A firm understanding of pharmacokinetics becomes essential since the usefulness of amiodarone is limited by its time-effect and adverse reaction profiles (both of which are probably related). Future questions include the determination of the role of following tissue as well as plasma levels of amiodarone, the role of following plasma levels to possibly deter adverse reactions and utilization of the unique kinetic profile of amiodarone to allow for alternatives in dosing that would lead to greater patient compliance without loss of efficacy. REFERENCES 1. Charlier R, Deltour serie des benzofurannes du butyl-2 (diiodo-3’, benzofuranne. Arch 2. Vastesaeger velle Medication 3.

LV

Van

M,

G, Tondeur R, Binion R: Reserches dans VII. Etude pharmacologique preliminaire 5-b-N-diethylamino-ethoxy-4’ benzaly)-3 Intl Pharmacodyn 1962:139:255.

Gillot P. Rasson G: Etude Antiangoreuse. Acta Cardiol

Schepdael

DYSF’JNCTION

J,

Solvay

AND

H: Etude

ARRHYTHMIAS

Clinique

Clinique (Brux) De

OF

PHARMACOKINETICS

d’une Nou1967:22:483. L’Amiodarone

la

Dans

Les

AMIODARONE

Troubles

du

Rhythme

Cardiaque.

Presse

Med

1970:78:1849. 4. Ward DE, Camm AJ, Spurrell fects of amiodarone in patients cardia. Br Heart 1 1980:44:91. 5. Rosenbaum amiodarone 1976:38:934.

MB, as

6. Rosenbaum RS, Lazrri antiarrhythmic

an

RA: Clinical with resistant

Chiale PA, Halpern antiarrhythmic

MB. Chiale

PA, Halper MVS: Clinical Am J Cardial

JO, Ekiazari agent.

antiarrhythmic paroxysmal

MS: agent.

Clinical Am

eftachyefficacy

of

Cordial

J

J, Levi

N. Nau GJ. Prybylski efficacy of amiodarone 1976:38:934.

as an

7. Patterson E, Eller BT, Abrams GD, Vasiliades J, Lucches BR: Ventricular fibrillation in a conscious canine preparation of sudden coronary death - Prevention by short and long-term amiodarone administration, Circulation 1983:68:857-864. 8. Winslow E: Hemodynamic and arrhythmogenic effects of aconitine applied to left atria of anesthetized cats: Effects of amiodarone and atropine. J Cardiovasc Phormacol 1981:3:87-100. 9. Zipes DP, Troup P1: New antiarrhythmic agents: Amiodarone, aprindine, disopyramide. ethmozin, mexiletine, tocainide, verapamil. Am I Curdial 1978:57:845-853. 10. Singh BN, new antianginal I 970;39:675-687.

Vaughan drug,

11. Goupil node activity

N,

12. Rosen drugs. Am

MR. Heart

Williams on the

EM: cardiac

The

Lenfant J: The effects of the rabbit heart. Eur Wit

AL:

effect muscle.

of amiodarone, Br I Pharmacal

of amiodarone I Pharmacol

Electropharmacology

a

on the sinus 1976:39:23-31. of antiarrhythmic

J 1983;106:829-839.

13. Singh BN, Jewitt DE, Downey JM, et al: Effects of amiodarone and L 8040, novel antianginal and antiarrhythmic drugs, on cardiac and coronary hemodynamics and cardiac intracellular potentials. Clin Exp Pharmacol Physiol 1976:3:427-442. 14. Singh BN, Vaughan Williams mic action. Effects on atrial and tials, and other pharmacological 1999 and AH 3474. Br 1 Pharmacol 15. Singh BN: cologic profile. 16. Mason inactivated 81.

17. vated

Amiodarone: Am Heart

Historical 11983:106:788-797.

JW. Hondeghem cardiac sodium

Eksp Biol Med

development

LM, Katzung BC: channels. Pjlugers

SV,

Revenko sodium

EM: A third class ofanti-arrhythventricular intracellular potenactions on cardiac muscle of MJ 1970:39:675-687.

Khodorov channels by 1980:89:702-704.

and

pharma-

Amiodarone blocks Arch 1983:396:79-

BI, Avrutskii MA: the antiarrhythmic

Blocking of inacticordarone, Bull

18.

McKenna WJ, Rowland E, Holt DW, Krikler DM: Electrophysiological assessment of amiodarone in atrial fibrillation complicating Wolff-Parkinson-White Syndrome. Br Heart J1981;45:617. 19. Charlier of amiodarone, Arzneimittelforsch

20. Schwartz A, Shen Chatterjee K: Hemadynamic patients with depressed ventricular tachycardia. 21. Pfisterer one depresses Circulation

apy.

with

J,

MI, Burkart F, Muller-Brand cardiac function acutely 1983:68:1122.

Nademanee K, Singh JAMA 1982:247:217-222.

a new

C: Pharmacology biological profile,

E, Morady F, Gillespie K, Scheinman effects of intravenous amiodarone left ventricular function and recurrent Am Heart 11983:106:849-855.

22. Zipes DP, Prystowsky EN, ologic actions, pharmacokinetics Cordial 1984:3:1059-1071.

23.

1’ Deltour

R. Delaunois C. Bauthier and antianginal drug 1968;18:1408-1417.

BN:

Heger

but

Kiowski W: Amiodardoes not chronically.

JJ: Amiodarone: and

clinical

Advances

in

A, in

effects.

ElectrophysiI Am

antiarrhythmic

Call ther-

1067

FREEDMAN

24.

Sogeol

PB,

Hershman

of amiodarone 5’-monodeiodination

25.

JM,

on serum

Balsam

A, Sexton paired in vitro generation ine in the livers 1979;105:1115-1121.

26. Freedberg altered thyroid

AS.

AW,

Dillman

WH:

The

death

GY,

Vaughan

Williams

intracellular

EM:

The

effect

45.

46.

E, Larratt L, phos-

47.

J Physiol

1970:207:357.

27. Poucell S, Ireton Patterson J, Blendis pholipidosis

and

28. Mazue

J, Valencia-Mayoral P, Downar L, Phillips MJ: Amiodarone-associated

fibrosis

of the

C, Vic P, Gouy

Barchewitz.

Gagnol

animals:

29. Andreasen F, Agerback cokinetics of amiodarone tion. Eur J Clin Pharmacol Tucker

Berger

Fund

H, Bjerregaard after intravenous

P, Gotzsche and oral

I

ligands.

Phorm

AppI

Taxi-

H: Pharmaadministra-

1981:19:293-299.

CT, Jackson PR, Storey Am Heart J 1983;106:840-847.

CGA:

Amiodarone

E, Zipes D: Plasma during chronic therapy.

Pharmacology Effects.

Paris:

Pharmacol

Pharmaco1986;990-

-

of amiodarone and other

by inter-

1984;36:366-372.

34. Fruncillo R, Bernhard B. Swanson B, Viasses P. Fergusson R: The effect of phenobarbital on the pharmacokinetics and tissue levels of amiodarone. Clin Pharmacol Ther 1984;35:241. 35. Bernhard R, Fruncillo R, Swanson B, Fergusson R: The effect of 3-methylcholanthrene on the pharmacokinetics of amiodarone in the rat. Fed Proc 1984;43:749. 36.

Nattal S, Davies M. Quantz M: The antiarrhythmic efficacy amiodarone and desethyl amiodarone. alone and in combination, in dogs with myocardial infarction. Circulation 1988:77:200-208.

37.

Berger Y, Harris - Toxicology

netics

38. Harris GCA, Halt metabolite.

L: Amiodarone, -

Clinical

Effects.

Pharmacology Paris: Medsi,

of

- Pharmacoki1986: 59.

L, Hind CRK, McKenna WJ, Savage C, Krikler SJ. Storey BW: Renal elimination of amiodarone and its desethyl Postgrad Med J 1983:59:440-442.

39. Mostow ND, Noon DL, Myers CM, Rakita L, Blumer JL: Determination of amiodarone and its N-deethylated metabolite in serum by high-performance liquid chromatography. I Chromatogr 1983:227: 229-237. 40. Flanigan RJ, Storey GCA, Holt DW: Rapid high-performance liquid chromatographic method for the measurement of amiodarone in blood plasma or serum at the concentrations attained during

therapy.

41.

Plomp

darone

and

I

Chromatogr

W. Maes

desethylamiodarone

R: Tissue in rats

neal administration of various forschung 1985;35:122-129.

after

amiodarone

distribution

of amio-

multiple

dosages.

intraperito-

Arzneimittel-

1068

#{149} J Clln Pharmacol

1991;31:1061-1069

V. Tognoni

and

D, Bory

C, Ciani

Pharmacology Paris: Medsi,

M, Bailie

after

sudden

1983;52:217-218. -

Pharmacoki-

1986;49.

Y, Serradimigni

A, Cano

plasma drug levels of amiodarone Eur Heart I 1984;5 (Suppl. 1):112.

P. Heusghem

C: Usefulness

studies.

tachyarrythmias.

51. Plomp T, Van R: Pharmacokinetics Arzneimittelforschung

Arch

and

of whole body Pharmocodyn

Intl

AT,

U.

efficacy

Circulation

Rossum and

53. Holt D, Storey rone pharmacokinetics.

Lesko

and

its de-

counting Ther

Asdourian

C, Al-

of amiodarone

1983:67:6,

C, Jackson

D:

for

1347-1355.

J, Robles de Medina E, Van Lier body distribution of amiodarone 1984:34:513-520.

52. Heger , Prystowsky, Zipes darone dosage, drug concentrations, Heart 11983:106:931-935.

Relationships and adverse

T, Maes in man.

between amioside effects. Am

P. Tucker C, McKenna W: Pharmacol 1983;16:206.

Amioda-

Br J Clin

54. Greenberg ML, Lerman BB, Shipe JR. Kaiser DL, Relation between amiodarone and desethylamiodarone concentrations and electrophysiologic effects, efficacy ity. I Am Call Cardiol 1987:9:1148-1155.

DiMarco JP: plasma and toxic-

55. Horowitz LN, Greenspan AM, Spielman SR, Webb CR, Morganroth J, Rotmensch H, Sokoloff NM, Rae AP, Segal BL. Kay HR: Usefulness of electrophysiologic testing in evaluation of amiodarone therapy for sustained ventricular tachyarrhythmias associated with coronary heart disease. Am 1 Cordial 1985:55:367-371. 56. Mostow ND, darone: Correlation complex ventricular 574.

Rakita L, Vrobel TR, Noon of serum concentration ectopic activity. Am

I

DL, Blumer J: Amiowith suppression of Cordial 1984:54:569-

57. Escoubet B, Coumel P. Poirier JM, Maison-Blanche P. Leclercq IF, Menasche P. Cheymol C, Pinnica Slama R: Suppression of arrhythmias within hours loading dose of amiodarone and relation to plasma dial concentrations. Am I Cardiol 1985:55:696-702. 58.

Nattel

5, Davies

amiodarone, alone cardial infarction. Nattel

its active 1986:8:771-777. 60. tric and

Heart

A. d’Aranno

Delwaide

refractory

43.

A, Volpi

L: Amiodarone, Clinical Effects.

P. Marchiset

tissues

I Cordial

48. Anastasiou-Nana M, Lewis C, Moulopoulos 5: Plasma levels of amiodarone in humans after intravenous and oral administration. I Clin Chem Clin Biochem 1981:19:599-600. 49. Riva E, Gerna M, Latini R, Ciani P. Volpi A, Maggioni A: Pharmacokinetics of amiodarone in man. J Cardiovasc Pharmacol 1982:4: 264-269.

61.

A. Maggi

-

in pharmacokinetic 1972:196:142-146.

42. Holt D. Adams P. Campbell R, Morley A, Callaghan J, Storey G: Amiodarone and its desethyl metabolite: Tissue distribution and ultrastructural changes in amiodarone-treated patients. Br I Clin Pharmacol 1984;17:195P-196P. Maggioni

in human Am

Broekhuysen J, Laurel R, Sion R: Recheres dans Ia serie des benzofurannes. XXXVII. Etude comparee du transit et du metabolisme de l’amiodarone chez diverses especes animales et chez l’homme. Arch Intl Pharmacodyn 1969;177:340-359.

59.

1980;187:391-398.

T, Wiersinga

Djiane

distribution recording.

50. Haffajee CI, Love IC, Canada pert JS: Clinical pharmacokinetics

and red cell Am I Car-

Medsi,

33. Lalloz M, Byfield P. Himsworth R: Binding serum proteins and the effects of drugs, hormones acting

J,

lipid-

study.

Y, Harris L: Amiodarane, Toxicology Clinical

-

F, Berthe

amiodarone-induced

A toxicological

Heger J, Solow E, Prystowsky concentrations of amiodarone diol 53: 91 2-917. kinetics 91.

86:926-936

B, Lacheretz

from

31.

32.

Gastroenterology

D, Remandet

JP: Recovery

osis in laboratory col 1984:4:992-999.

30. HoIt DW, pharmacokinetics.

liver.

Halter

JP: Myocardial sethyl metabolite.

of

potentials.

tissue

during

44. Berger Y, Harris netics - Toxicology

Ingbar SH: On the mechanism of imof 3,5,3’-triiodothyronine from thyroxof hypothyroid rats. Endocrinology

on atrial

SOMBERG

P: Amiodarone

effects

and hepatic thyroxine 1983;113:1464-1469.

F.

Papp

state

Reed

thyroid hormones in rats. Endocrinology

AND

M, Quantz

M: The

Reddy CP, induction, its relation

antiarrhythmic

Kuo CS, morphology, to clinical

Jivrajka and efficacy.

studies

I V: Effect rate of PACE

of

with

acute

of

myo-

amiodarone

Cardiovasc

and

Pharmacol

of amiodarone on elecventricular tachycardia 1984:7:1055-1062.

DiCarlo LA, Marady F, deBuiteleir M, Baerman Annesley T: Effects of chronic amiodarone therapy tachycardia induced by programmed ventricular

11987:113:57-64.

efficacy

and in combination. in dogs Circulation 1988:77:200-208.

5: Pharmacodynamics N-desethyl metabolite.

P. Jaillon A, Lagier C, after a single and myocar-

JM, Schurig U. on ventricular stimulation. Am

PHARMACOLOGY

62.

K. Hendrickson J, Kannon and electrophysiologic life-threatening ventricular

Nademanee rhythmic efficacy in patients with

suppression Am Heart 63.

of spontaneously J 1982;103: 950-959.

occurring

64.

HU: The efficacy cannot be predicated Intl J Cordial 1983:3:71.

McGovern

tricular ologic

B, Ruskin

arrhythmias studies. Intl

65.

Horowitz

KR:

Ventricular

Am

Heart

UN.

can

efficacy

be predicated 1983;3:71.

Speilman arrhythmias:

1 1983;106:4,

ventricular

tachycardia.

75.

for ventricular electrophysiologic

of amiodarone

with

clinical

76. nary

electrophysiCR. Kay studies.

one.

long-term fibrillation.

therapy N

for EngI

Med

1981:305:539-545. 67.

Heger

darone

JJ. Prystawsky in the treatment

EN,

Zipes

DP:

of recurrent

fibrillation.

68. Waxman dowski UM,

HL, Croh WC, Marchlinski Horowitz UN, Josephson ME,

of sustained

electrophysiologic

1982;50:1066-1074. 69. McGovern

Heart

ventricular effects

51

H, Malacoff Long

80.

of amio-

tachycardia

and

fects

Sa-

81.

Heart

FE. Kastor

Buxton AE, JA: Amiodarone

tachyrhythmia:

in

B, Caran

Sellers TD, Ruskin JN: tachycardia or fibrillation 1984;53:1558-1563.

efficacy

J 1983:106:887-894.

ventricular

for control

Am

Clinical

ventricular

term treated

patients.

RF, Di Marco

clinical outcome with amiodarone.

71.

Collaborative

group

72.

Kim

SG, Felder of Holter

JD: and

Value inefficacy

SD, Figura

monitoring of amiodarone

class IA antiarrhythmic cardia. I Am Coil Cordial 73. Kim ventricular

or Holter

LV

for amiodarone of a low-dose ventricular

JP, Grant

SC:

agents

The management tachyarrythmias: monitoring (either

DYSFUNCTION

observation: regimen arrhythmias.

I, Johnston in used

predicting alone or

in patients

Multicenter

long-term in combination with ventricular

UE. Fisher efficacy with tachy-

1987:9:169-174. of patients with life threatening Programmed electrical stimulation or both)? Circulation 1987:76:1-5.

AND ARRHYTHMIAS

Mostow 1106:4,

N, Vrobel 906-914.

ND, Vrobel RT, Noon ventricular arrhytmias Circulation 1986:73:1231-1238.

Ward

DE, Comm

of amiodarone I 1980;44:91-95. Rotmensch

82.

C,

of amiodarone for Am I Cordial

DR, Waspe

SM, Heart

HH.

with

CR, based

and pulmo-

concepts.

Roden DM, on its pharma-

and toxicity tachyarrythmias.

of amiodaProg

antiarrhythmic tachycardia.

paroxysmal

B: Amiodarone

Current

PP:

D, Rakita L: Rapid suppression with high-dose oral amiodar-

RAJ: Clinical

patients

Belhassen

arrhythmias:

T: Amiodarone

CB, Wilkinson A proposal 951-956.

AJ. Spureli in

DL, DiMarco

desethylamiodarone effects, efficacy

efBr

in the management Med Gun North

Am

72:321-358.

of ventricular Am I Cordial

70. Hamer AW, Fine rman WB Jr, Peter T, et al: Disparity between the clinical and electrophysiologic effects of amiodarone in the treatment of recurrent ventricular tachyarrythmias. Am Heart 1981;102:992-1000. controlled observation treatment of severe 1984:53:1564-1569.

Rakita U, Sobel toxicity. Am

of cardiac

Clinical and J Cordial

Am

JR. Kaiser

79. Kopeiman I-IA, Horowitz LN: Efficacy rone for the treatment of supraventricular Cardiovasc Dis 1989;31:355-366.

recurrent

J

BB. Shipe

78. Siddoway LA, McAllister Woosley RU: Amiodarone dosing: cokinetics. Am Heart 11983:106:

881-885.

during ventricular

MU, Uerman

between amiodarone and concentrations and electrophysiologic Am Coil Cardiol 1987:9:1148-1155.

77. Mostow of complex

66. Heger JJ. Prystowsky EN, Jackman WM, Naccarelli CV, Warfel KA, Rinkenberger RU, Zipes DP: Amiodarone: Clinical efficacy and electrophysiology tachycardia or

Greenberg

plasma toxicity.

ar-

for ven-

SR, Creenspan AM. Webb Use of electrophysiologic

OF AMIODARONE

74. Haffajee CI, Love JC, Alpert JS, Asdourian CK, Sloan KC: Efficacy and safety or long term amiodarone in treatment of cardiac arrythmia: Dosage experience. Am Heart I 1983;106:935-943.

R, Singh BN: Antiaractions of amiodarone arrhythmias: Potent

of amiodarone with clinical

JN: The

I Cordial

PHARMACOKINETICS

Relationship

Waxman

rhythmias studies.

AND

Harris cokinetics 131.

83. migni netics nance

I

Marchiset

U, Munoz Toxicology

-

A: in: Amiadarone, Clinical Effects.

D, Bruno

R, Dijiane

P, Cano

Pharmacology Paris:Medsi.

J, Benichou

-

Pharma-

1986:100-

M, Serradi-

A: Amiodarone and desethylamiodarone elimination kifollowing withdrawal of long-term amiodarone maintetherapy. Biopharm Drug Dispos 1985:6:209-215.

84. Kannan R, Nademanee JA, Hendrickson JA, Rostami H, Singh BN: Plasma levels of amiodarone and its elimination half-life following acute and chronic dosing in patients with ventricular arrhythmias. Circulation 1981 ;64(Suppl IV):264. 85. Kannan R, Nademanee BN: Amiodarone kinetics 1982;31: 438-444. 86. Harris U, McKenna Krickier DM, Halt DW: rone levels in chronic IV):263A.

K, Hendrickson after oral doses.

J, Rostami Clin

WJ, Rowland E, Storey Plasma amiodarone and oral therapy. Circulation

Pharmacol

HJ, Singh Ther

CCA, Storey DM, desethyl amioda1981;64(Supp

87. Boppana VK, Greenspan A, Swanson BN, Speilman 5, Ferguson RK, Beihassen B, Rotmensch HH: Clinical efficacy and serum concentrations of amiodarone. Clin Pharm Ther 1983:33:209.

1069

Pharmacology and pharmacokinetics of amiodarone.

Amiodarone is a unique antiarrhythmic agent originally developed as a vasodilator. Classified electrophysiologically as a Type III antiarrhythmic, it ...
2MB Sizes 0 Downloads 0 Views