You are on page 1of 33

SPECIAL POPULATIONS Clin Pharmacokinet 1999 Nov; 37 (5): 399-431

0312-5963/99/0011-0399/$16.50/0

© Adis International Limited. All rights reserved.

Effects of Liver Disease


on Pharmacokinetics
An Update
Vanni Rodighiero
Department of Pharmacology, University of Padova, Padova, Italy

Contents
Abstract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 400
1. Liver Function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 401
2. Hepatic Blood Flow . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 401
3. Cytochrome P450 (CYP) System and Liver Disease . . . . . . . . . . . . . . . . . . . . . . . . . . 401
3.1 CYP1A . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 402
3.2 CYP2A . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 402
3.3 CYP2C . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 402
3.4 CYP3A . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 403
3.5 Relative Changes in CYP Enzyme Levels and Activity . . . . . . . . . . . . . . . . . . . . . . 404
4. Cardiovascular Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 405
4.1 Angiotensin Converting Enzyme (ACE) Inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . 405
4.2 Angiotensin II Receptor Antagonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 405
4.3 Calcium Antagonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 406
4.4 Ketanserin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 408
4.5 Antiarrhythmics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 409
4.6 Hypolipidaemics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 410
5. Diuretics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 410
5.1 Torasemide . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 410
6. Psychoactive and Anticonvulsant Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 411
6.1 Benzodiazepines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 411
6.2 Antidepressants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 412
6.3 Tiagabine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 414
7. Antiemetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 415
7.1 Metoclopramide . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 415
7.2 Serotonin Antagonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 415
8. Antiulcers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 416
8.1 Proton Pump Inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 416
9. Antimicrobials and Antiretrovirals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 417
9.1 Grepafloxacin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 417
9.2 Ornidazole . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 417
9.3 Pefloxacin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 418
9.4 Stavudine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 418
9.5 Zidovudine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 418
10. Immunosuppressants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 419
10.1 Cyclosporin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 419
10.2 Tacrolimus . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 419
11. Miscellaneous Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 420
11.1 Naltrexone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 420
400 Rodighiero

11.2 Tolcapone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 420


11.3 Toremifene . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 421
12. Clinical Implications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 421
12.1 Altered Pharmacokinetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 421
12.2 Altered Pharmacodynamics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 423
13. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 427

Abstract Liver disease can modify the kinetics of drugs biotransformed by the liver.
This review updates recent developments in this field, with particular emphasis
on cytochrome P450 (CYP). CYP is a rapidly expanding area in clinical pharma-
cology. The information currently available on specific isoforms involved in drug
metabolism has increased tremendously over the latest years, but knowledge
remains incomplete.
Studies on the effects of liver disease on specific isoenzymes of CYP have
shown that some isoforms are more susceptible than others to liver disease. A
detailed knowledge of the particular isoenzyme involved in the metabolism of a
drug and the impact of liver disease on that enzyme can provide a rational basis
for dosage adjustment in patients with hepatic impairment.
The capacity of the liver to metabolise drugs depends on hepatic blood flow
and liver enzyme activity, both of which can be affected by liver disease. In
addition, liver failure can influence the binding of a drug to plasma proteins. These
changes can occur alone or in combination; when they coexist their effect on drug
kinetics is synergistic, not simply additive. The kinetics of drugs with a low
hepatic extraction are sensitive to hepatic failure rather than to liver blood flow
changes, but drugs having a significant first-pass effect are sensitive to alterations
in hepatic blood flow.
The drugs examined in this review are: cardiovascular agents (angiotensin
converting enzyme inhibitors, angiotensin II receptor antagonists, calcium antag-
onists, ketanserin, antiarrhythmics and hypolipidaemics), diuretics (torasemide),
psychoactive and anticonvulsant agents (benzodiazepines, flumazenil, antide-
pressants and tiagabine), antiemetics (metoclopramide and serotonin antago-
nists), antiulcers (acid pump inhibitors), anti-infectives and antiretroviral agents
(grepafloxacin, ornidazole, pefloxacin, stavudine and zidovudine), immunosup-
pressants (cyclosporin and tacrolimus), naltrexone, tolcapone and toremifene.
According to the available data, the kinetics of many drugs are altered by liver
disease to an extent that requires dosage adjustment; the problem is to quantify
the required changes. Obviously, this requires the evaluation of the degree of
hepatic impairment. At present there is no satisfactory test that gives a quantitative
measure of liver function and its impairment. A critical evaluation of these meth-
ods is provided. Guidelines providing a rational basis for dosage adjustment are
illustrated.
Finally, it is important to consider that liver disease not only affects pharma-
cokinetics but also pharmacodynamics. This review also examines drugs with
altered pharmacodynamics.

This article updates a 1987 review[1] and pro- with liver disease. Other reviews on this subject
vides an overview of developments in the under- have been published previously.[2-5]
standing of how drug kinetics are altered in patients The capacity of the liver to metabolise drugs

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
Effects of Liver Disease on Pharmacokinetics 401

(hepatic clearance) depends on hepatic blood flow tion increases because of hypoproteinaemia, the
and enzyme activity. Factors that affect liver blood liver will remove all the drug anyway.
flow and enzyme activity can substantially alter Very few studies consider the effects of decreased
drug disposition and the therapeutic response to a plasma proteins on drug kinetics in patients with
drug. Liver disease is clearly a factor that can mod- liver disease.
ify the kinetics of a drug by changing hepatic blood
flow and reducing the activity of drug metabolising 2. Hepatic Blood Flow
enzymes. These changes can occur alone or in
Several drugs are subject to a significant oral first-
combination. When they coexist, their effect on
pass effect which leads to a reduced systemic avail-
drug kinetics is synergistic, not simply additive. In
ability. Liver blood flow can be reduced because
acute liver disease, the major alteration is in
of pathological alterations caused by liver disease,
hepatocellular function; in chronic liver disease,
as in cirrhosis. There can be spontaneous or surgi-
such as cirrhosis, the major abnormality is in liver
cal portacaval shunts. After oral administration of
blood flow and/or alteration in liver function.
a drug with a high first-pass effect to patients with
portacaval shunts, a fraction of the drug enters the
1. Liver Function shunt, thus bypassing the liver and reaching the
systemic circulation directly without being ex-
Drugs with a low hepatic extraction are not sen-
posed to the first-pass effect. This results in greater
sitive to liver blood flow changes. The fraction of
systemic availability, which may be increased
these drugs removed from the blood during a single
many-fold depending on the proportion of portal
passage through the liver, that is the extraction ratio,
blood flow shunted. Although drugs with a first-
is small, so hepatic clearance is not limited by the
pass effect are sensitive to changes in liver blood
amount of drug carried to the liver. Their clearance
flow, this is not the case for drugs with a low he-
depends on the activity of drug metabolising en-
patic extraction because the fraction of these drugs
zymes. Thus, hepatic failure can reduce the capacity
removed from the blood during a single passage
of the liver to metabolise the drug, resulting in a
through the liver is small.
decrease in hepatic drug clearance, an increase in
the area under the curve of plasma/serum concen-
3. Cytochrome P450 (CYP) System
tration versus time (AUC) and a prolongation of
and Liver Disease
the half-life (t1⁄2) of the drug. It is evident that if the
liver metabolises less drug, more drug is present in There are numerous pathways of drug metabo-
the systemic circulation and more time is needed lism in the liver, each affected to a different degree
for its elimination. by liver disease, oxidation being more sensitive
In blood many drugs are bound to plasma pro- than glucuronidation.[3] Oxidative metabolism plays
teins, mainly to albumin and/or α1-acid glycopro- a fundamental role and it is largely catalysed by
tein, whose synthesis can be decreased by hepatic enzymes belonging to the CYP family. One of the
failure with consequent reduction in drug binding. most rapidly expanding areas in clinical pharma-
The effect of hypoproteinaemia on drug kinetics cology is the study of hepatic drug biotransforma-
depends on the type of agent and degree of protein tion by the CYP system, and the identification and
binding. Drugs with a high extraction ratio are re- characterisation of individual CYP isoforms. The
moved by the liver as rapidly as they circulate in information currently available on specific CYP
hepatic blood because the liver is capable of meta- isoforms involved in the metabolism of a variety
bolising all the drug reaching the liver.[6] Thus, the of therapeutic agents has increased tremendously
hepatic clearance of bound and unbound drug is over recent years. However, knowledge remains
not sensitive to changes in drug protein binding but incomplete and for many agents the CYP isoform
is flow-dependent. If the unbound drug concentra- has not yet been identified. In humans the 3 fami-

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
402 Rodighiero

lies predominantly involved in hepatic drug meta- 3.2 CYP2A


bolism are CYP1, CYP2 and CYP3.[7]
Hepatic disease is associated with reduced me- Coumarin undergoes hydroxylation by CYP2A6
tabolic capacity for drugs that undergo oxidative to 7-hydroxycoumarin, which is conjugated with
biotransformation, but information on impairment glucuronic acid and rapidly excreted in the urine.[16]
of individual CYP isoforms is still limited.[8] Sev- Coumarin has been used as a probe drug for mea-
eral probe drugs have been used to investigate this suring liver function.[17] Oral coumarin 5mg was
point, but when drugs metabolised by CYP are used given to 36 patients with alcoholic liver disease
as a probe a common drawback in study design is classified as mild, moderate and severe according
the fact that CYP isoenzymes can be variably ex- to Child-Pugh score and histological diagnosis (11
pressed, being subject to inhibition and induction class A, 14 class B, 11 class C). The urinary excre-
by a variety of agents, as in the case of drug tion of 7-hydroxycoumarin was decreased and in-
coadministration to patients. In addition, CYP iso- versely correlated to Child-Pugh scores. However,
enzymes can exhibit genetic polymorphism. Thus, the coumarin test is not applicable in patients with
the presence of these factors may lead to erroneous impaired kidney function because the excretion of
conclusions when using a drug as a probe. 7-hydroxycoumarin is compromised.
Kraul et al.[18] measured the activity of 7-eth-
3.1 CYP1A oxycoumarin O-de-ethylase in liver biopsy speci-
mens from 23 patients with alcoholic liver disease
Caffeine is almost completely metabolised by and different degrees of structural liver damage
the liver; it is mainly demethylated by CYP1A2.[9] (fatty liver, chronic active hepatitis, cirrhosis). The
The drug has been used as a probe in patients with activity was reduced in proportion to the degree of
liver impairment, showing a decrease in CYP1A2 ac- liver damage.
tivity proportional to the severity of liver disease.
Holstege et al.[10] studied 27 patients with liver 3.3 CYP2C
cirrhosis who were given an oral dose of caffeine
7 mg/kg. The degree of hepatic impairment was There are 4 identified members belonging to
determined using the Child-Turcotte classifica- this subfamily: CYP2C8, CYP2C9, CYP2C10 and
tion[11] and the Child-Pugh scoring system[12] (7 class CYP2C19.[7] The CYP2C9 isoenzyme is not sensi-
A, 8 class B, 12 class C). The apparent caffeine tive to liver disease. Tolbutamide[7] and irbesar-
clearance decreased with increasing impairment of tan[19] are substrates for CYP2C9/10 and CYP2C9,
liver function, the correlation being highly signif- respectively. Mephenytoin is a racemic drug: the
icant. The demethylation of caffeine was severely R-enantiomer is metabolised by CYP2C9[20] and
impaired in these patients. the S-enantiomer by CYP2C19.[21] Liver disease
Scott et al.[13] obtained similar results in 19 pa- does not affect the disposition of irbesartan,[22] tol-
tients with cirrhosis (10 with Child-Pugh score ≤7, butamide[23] and R-mephenytoin, but it impairs the
9 with score 8 to 12). Upon oral administration of metabolism of S-mephenytoin.[21]
caffeine 400mg a significant linear inverse corre- S-Mephenytoin is a specific probe drug metabo-
lation between the degree of hepatic impairment lised by CYP2C19, a genetically polymorphic en-
and salivary caffeine clearance was observed. Sim- zyme.[21] After oral administration of racemic
ilarly, in 34 patients with mild, moderate and se- mephenytoin 100mg to 18 patients (poor metabo-
vere alcoholic liver disease who were given an oral lisers excluded) with mild or moderate liver cirrho-
dose of caffeine 300mg, salivary caffeine clearance sis (9 class A and 9 class B according to Pugh scores
strongly and inversely correlated with a modified and Child class) a marked decrease in the apparent
Child-Pugh score[14,15] (10 class A, 10 class B, 14 oral clearance (CL/F) of S-mephenytoin was ob-
class C). served with a corresponding reduction in the uri-

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
Effects of Liver Disease on Pharmacokinetics 403

nary excretion of the metabolite 4’-hydroxymephen- 3.4 CYP3A


ytoin.[21] The decrease in clearance was related to
the severity of the disease and exhibited an inverse The isoenzymes belonging to the CYP3A sub-
family are the most abundant form of CYP.[26] They
relation to Pugh scores: patients with moderate cir-
are generally referred to collectively as CYP3A be-
rhosis were affected more severely than patients
cause of the inability to readily distinguish among
with mild cirrhosis. Thus, CYP2C19 appears to be the various isoforms, especially in vivo. High levels
very sensitive to hepatic impairment. These results of CYP3A are present in the gastrointestinal tract
are consistent with those obtained in a previous and liver. Accordingly, the route of administration
study[24] which showed reduced elimination of S- of a drug becomes an extremely important factor.
mephenytoin in patients with chronic hepatitis. CYP3A is involved in the oxidative metabolism of
Adedoyin et al.[21] simultaneously evaluated in many important drugs. Although the activity of
the same group of patients the effect of liver im- CYP3A is subject to large interindividual varia-
pairment on CYP2D6, a genetically polymorphic tions, genetic polymorphism has not been demon-
enzyme, with the use of the specific probe drug strated.
debrisoquine (10mg orally). In contrast with S- The metabolism of drugs involving CYP3A is
affected in patients with liver disease: a reduction
mephenytoin, the metabolism of debrisoquine was
in activity (30 to 50%) has been reported.[26] Sev-
not affected by liver disease. Thus, this study showed eral drugs metabolised by CYP3A have been used
a selective effect of liver disease on the activity of to evaluate the effect of liver impairment on
specific CYP enzymes, CYP2C19 being more sen- CYP3A. Huang et al.[27] measured serum mono-
sitive than CYP2D6. This study has the advantage ethylglycinexylidide (MEGX) concentration 15
of simultaneously evaluating the effect of liver im- minutes after intravenous administration of a sin-
pairment on 2 isoenzymes in the same population. gle dose of lidocaine 1 mg/kg to 24 patients with
Because of the multiplicity of CYP enzymes, mul- chronic hepatitis and 47 patients with liver cirrho-
tiple studies are needed to assess different enzymes, sis (16 class A, 13 class B, 18 class C Child-Pugh
making it difficult to compare the results from in- classification). Lidocaine is metabolised to MEGX
via oxidative N-de-ethylation by CYP3A. The se-
vestigations using different study design and pop-
rum MEGX concentrations were inversely propor-
ulations.
tional to Child-Pugh classes and directly propor-
A new method (the ‘Pittsburgh cocktail’), based tional to galactose elimination capacity, although
on the simultaneous administration of low doses of there was no correlation with serum albumin or
multiple probes of drug-metabolising enzymes total bilirubin levels. There was a large variation in
(caffeine, chlorzoxazone, dapsone, debrisoquine MEGX concentrations, but this probably reflects
and mephenytoin) with the absence of metabolic the wide variation in CYP3A activity found in
interactions among the drugs used, has been re- healthy individuals. Patients taking drugs affecting
ported.[25] This method provides simultaneously lidocaine metabolism were excluded from the
independent estimates of respective individual study. Of course, lidocaine administration is not
suitable for patients with cardiovascular disorders.
metabolising enzyme activities in the same popu-
In conclusion, the MEGX formation test reflects
lation and it may be useful for the evaluation of
the severity of hepatic impairment and may serve
differential effects of a particular disease state or as a simple and rapid quantitative liver function test.
drug therapy on CYP activity. However, there is a Similar results were obtained by Shiffman et
drawback in the coadministration of probe drugs al.[28] In their study, serum concentrations of MEGX
as ‘cocktails’, since they can result in complex in- were determined 15 minutes after the intravenous
teractions. administration of a single dose of lidocaine 1

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
404 Rodighiero

mg/kg to 225 patients with chronic hepatitis (107 were noted in the CYP enzymes and CYP-specific
had biopsy-proven cirrhosis). A liver biopsy was mRNA content of hepatic tissue.
performed in all patients to assess liver histological CYP1A2 appears to be particularly susceptible
condition, which was scored according to the clas- to the effects of liver function impairment. The
sification of Knodell. The severity of cirrhosis was level of CYP1A2 was significantly reduced both in
further graded by Child classification as mild, cholestatic and noncholestatic cirrhosis, with a lin-
moderate and severe. MEGX production declined early correlated decrease in its catalytic activity
in parallel with deteriorating hepatic histology. A (ethoxyresorufin O-de-ethylase). CYP1A2 mRNA
decline in parallel with worsening Child class was levels were correspondingly reduced.
also observed in patients with cirrhosis. The range CYP2C and tolbutamide 4-hydroxylase activity
of MEGX values varied widely, probably in keep- were not reduced in either type of cirrhosis.
ing with the wide variation observed in healthy CYP2C activity was determined by the 4-hydrox-
individuals. The possibility of interference with the ylation of tolbutamide catalysed by CYP2C9/10.[7]
metabolism of lidocaine can be ruled out because The levels of mRNA were diminished in all cir-
patients taking known inducers or inhibitors of rhotic livers. However, the authors used a polyclo-
CYP3A were excluded from the study. nal antibody to identify CYP2C8/10; this antibody
The results reported above were confirmed by a is not specific and probably recognised other mem-
study of Sotaniemi et al.[17] in 36 patients with al- bers of the CYP2C subfamily.
coholic liver disease (11 class A, 14 class B, 11 CYP3A4 was significantly reduced with a par-
class C Child-Pugh classification) after intrave- allel decrease in catalytic activity (testosterone 6β-
nous administration of lidocaine 1 mg/kg. hydroxylase activity) in cirrhotic patients without
These findings are in agreement with the studies cholestasis, whereas in patients with cholestasis
that have shown reduced clearance of drugs meta- there was a reduction but it was not considered sig-
bolised by CYP3A in patients with liver impair-
nificant. Similarly, CYP3A4 mRNA was decreased
ment.
with a significant correlation with CYP3A4 content.
CYP2E1 and the related N,N-dimethylnitros-
3.5 Relative Changes in CYP Enzyme
amine N-demethylase activity were reduced only
Levels and Activity
in cirrhotic patients with cholestasis, whereas CYP-
George et al.[29,30] examined 50 livers removed 2E1 mRNA levels were lower in patients with both
for transplantation from patients with end-stage types of cirrhosis.
cirrhosis (18 with and 32 without cholestasis) to In conclusion, the studies showed that the extent
determine the levels of CYP1A2, 2C8/10, 2E1 and to which liver impairment affects drug metabolism
3A, their catalytic activities and the corresponding depends on the specific enzyme involved and the
levels of mRNA. The severity of liver disease was severity of the underlying liver disease. Some CYP
assessed using the Child-Pugh classification (4 isoforms are more susceptible than others. There is
class A, 17 class B, 29 class C). The total content a range of sensitivity in the presence of liver dis-
of CYPs was decreased but there was a selectivity ease, even though the mechanism responsible for
for individual enzymes, some being more affected this differential effect remains unknown. It is im-
than others. Moreover, the effect was related to the portant to have a detailed knowledge of the partic-
type of liver disease (cholestasis or no cholestasis). ular enzyme involved in the metabolism of a drug,
However, there is little information in the literature of the impact of liver disease on that enzyme and
about cholestatic liver disease. The reasons for of the degree of liver impairment. An adequate
these differences are unknown. It would be impor- amount of information on these factors can provide
tant to identify the mechanisms underlying this se- a rational basis for dosage adjustment in patients
lectivity of effects. Large interindividual variations with liver disease. Dosage adjustment depends on

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
Effects of Liver Disease on Pharmacokinetics 405

which specific enzyme mediates the metabolism of in patients with decompensated cirrhosis[35] [7 pa-
the drug involved and the severity of the disease tients given a single oral dose of 10mg, all with
itself. Since information is limited, studies are varices, splenomegaly and 37% retention of indo-
needed to investigate the effects of different de- cyanine green (ICG) at 15 min] as shown by a
grees of hepatic impairment on the activity of spe- marked reduction in the CL/F of enalapril resulting
cific metabolising enzymes so that specific guide- in a higher AUC and peak plasma drug concentra-
lines for dosage adjustment can be developed. tion (Cmax), while the AUC and Cmax of enalaprilat
were lower (CL/F for enalapril 39.2 vs 91.6 L/h in
4. Cardiovascular Agents healthy individuals, AUC for enalapril 276 vs 122
μg/L • h, Cmax for enalapril 123 vs 66 μg/L; AUC
4.1 Angiotensin Converting Enzyme for enalaprilat 173 vs 375 μg/L • h and Cmax of 16
(ACE) Inhibitors vs 46 μg/L).
The above studies demonstrated that the con-
Most ACE inhibitors are prodrugs, esters of the version of the prodrug to its active form is reduced
active form, because the active drug is poorly ab- when liver function is impaired. It is probably bet-
sorbed from the gastrointestinal tract.[31] The esters ter to avoid the use of ACE inhibitors, which are
are readily absorbed and converted in the liver to prodrugs, in patients with hepatic dysfunction
the active form, which is usually excreted in the since their dosage may need to be increased to en-
urine. It can be assumed that hepatic conversion to sure adequate concentrations of the active metabo-
the active form can be impaired by liver dysfunc- lite. The use of non-prodrug ACE inhibitors, such
tion. Although few studies are available, it is clear as lisinopril, a lysine analogue of enalaprilat, may
that the conversion of the prodrug is significantly be advocated in these clinical situations.
reduced in patients with impaired liver function.
Quinalapril is a prodrug converted in the liver 4.2 Angiotensin II Receptor Antagonists
to the active form, quinalaprilat. This biotransfor-
mation is impaired in patients with compensated Angiotensin II receptor antagonists are a new
alcoholic cirrhosis (as shown with a single oral class of drug for the treatment of patients with hy-
dose of 20mg),[32] producing a 3-fold higher AUC pertension. Losartan was the first agent of this class
of quinalapril than in matched healthy individuals, and has been followed by valsartan, irbesartan and
a 2-fold lower AUC of quinalaprilat and a 2-fold others. The number of studies on these drugs are
longer t1⁄2 of quinalapril (AUC for quinalapril 744 still limited and information is incomplete.
vs 257 μg/L • h and for quinalaprilat 1558 vs 3410 4.2.1 Losartan
μg/L • h; the t1⁄2 of quinalapril was 1.4 vs 0.7h). The Losartan is a nonpeptide angiotensin II receptor
t1⁄2 of quinalaprilat was not changed in these pa- antagonist[36,37] which binds competitively and se-
tients because quinalaprilat is largely eliminated in lectively to the angiotensin II subtype 1 receptor,
urine. thus blocking the physiological effects of angio-
Similar results have been obtained in 9 patients tensin II. The oral bioavailability of losartan is
with compensated liver cirrhosis given a single about 33% because of a considerable first-pass ef-
oral dose of cilazapril 1mg, a prodrug biotrans- fect. The protein binding is >90%. It is mainly me-
formed in the liver to cilazaprilat, because of a re- tabolised by hepatic CYP2C9 and CYP3A to sev-
duced clearance of cilazapril.[33] eral metabolites. The metabolite E-3174 is active
The conversion of enalapril to enalaprilat is re- and largely responsible for its antihypertensive ef-
duced in patients with well compensated liver cir- fect.
rhosis and impaired liver function compared with In patients with mild to moderate alcoholic cir-
healthy individuals.[34] The biotransformation of rhosis,[37] the total plasma clearance (CLp) of
enalapril to enalaprilat was considerably impaired losartan is decreased by approximately 50%, the

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
406 Rodighiero

oral bioavailability is doubled and the plasma con- The effect of hepatic impairment was assessed
centrations of losartan and E-3174 are increased 5- in 10 cirrhotic patients with mild or moderate he-
and 1.7-fold, respectively, as compared with patic impairment (Child-Pugh class A and B) given
healthy individuals. Therefore, a lower dosage oral irbesartan 300mg once daily for 7 consecutive
seems justified in patients with liver impairment; a days.[22] The pharmacokinetics were not signifi-
25mg once daily dose could be appropriate. It cantly altered; there were no significant differences
should be noted that the effects of losartan, between healthy individuals and patients with cir-
valsartan and irbesartan are dose-related. rhosis. This is in keeping with the fact that CYP-
2C9 is not sensitive to the effects of liver disease.
4.2.2 Valsartan Irbesartan could be a good choice for the treatment
Valsartan, a compound structurally related to of hypertension in patients with impaired liver
losartan, is a selective competitive angiotensin II function. However, more studies are needed to
antagonist which binds to the angiotensin II sub- evaluate the kinetics of irbesartan in severe liver
type 1 receptor.[38] The fraction of the oral dose disease.
absorbed by the gastrointestinal tract and systemi-
cally available is 23% for the capsule and 39% for 4.3 Calcium Antagonists
the solution. Binding to plasma proteins (albumin)
is >90%. Valsartan undergoes little hepatic meta- Calcium antagonists are widely used for the
bolism and is largely excreted unchanged in the treatment of patients with cardiovascular disease.
bile. The main route of elimination is biliary excre- They have some common pharmacokinetic charac-
tion. teristics:[40] they are subject to a high first-pass ef-
fect in the liver resulting in a low oral bioavailabil-
Patients with mild and moderate liver impair-
ity and are metabolised almost entirely by the liver.
ment (6 class A, 6 class B by the Child-Pugh clas-
All calcium antagonists reported in this review, ex-
sification) with different types of disease (primary
cept for gallopamil, are biotransformed by CYP3A.
biliary cirrhosis, alcoholic cirrhosis, alcoholic hep-
Hepatic disease alters, possibly markedly, the
atitis, hepatomegaly and extrahepatic biliary ob-
kinetics of calcium antagonists, the pharmacokinetic
struction) were given a single oral dose of valsartan
alterations depending on the severity of liver im-
160mg.[39] Exposure to valsartan, measured as
pairment. If given in normal dosages to patients
AUC, was increased approximately 2-fold com-
with liver disease, the pharmacological effect could
pared with matched healthy volunteers. The possi-
be excessive and dangerous. Dosage modifications
ble mechanism explaining the altered pharmaco-
are advisable in patients with significant hepatic
kinetics is the impaired transport of valsartan into
impairment, as for most calcium antagonists there
the bile. A lower dosage seems advisable in patients
is a relationship between plasma concentrations
with liver impairment. However, this study has
and effect.
drawbacks, since it grouped together patients with
very heterogeneous diseases and the number of 4.3.1 Felodipine
patients is small. Felodipine has a bioavailability of about 16%
and protein binding of >99%.[40] In 9 cirrhotic pa-
4.2.3 Irbesartan tients with impaired liver function after a single
Irbesartan is a nonpeptide selective and non- dose of felodipine 0.75mg intravenously or 10mg
competitive antagonist of angiotensin II receptor orally, the AUC and Cmax were almost twice that
subtype 1. Oral bioavailability is about 60 to 80% found in healthy individuals as a result of a reduced
and binding to plasma proteins is approximately CL/F (one-third of that in healthy individuals).[41]
90%.[19] It is primarily metabolised by hepatic The bioavailability in patients with cirrhosis, in-
CYP2C9 with the amount of the drug excreted un- cluding 2 patients with a portacaval anastomosis,
changed in the urine being about 1%. was almost the same as in healthy individuals. The

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
Effects of Liver Disease on Pharmacokinetics 407

most likely explanation is that the intestine plays a 4.3.4 Nifedipine


major role in the first-pass metabolism of felodip- Nifedipine has a bioavailability of about
ine.[42] This study did not specify the degree of liver 45%.[40] The protein binding is 96 to 98%. Its kinetics
impairment and included heterogeneous diseases. are grossly altered in patients with liver cirrhosis.
A reduction in dosage is recommended in patients In 7 patients with cirrhosis, all with varices and
with liver damage; a starting dosage of 5 mg/day mostly with ascites, the capacity of the liver to me-
seems appropriate. tabolise the drug (a single intravenous dose of ni-
fedipine 4.5mg) was reduced, as shown by a de-
4.3.2 Gallopamil creased systemic clearance (CL) and an almost
Gallopamil is a methoxy derivative of verapa- 4-fold prolongation of t1⁄2 (CL 13.98 vs 35.28 L/h
mil. Although it is almost completely absorbed af- in healthy controls; t1⁄2 7 vs 1.85h), while the oral
ter oral administration, oral bioavailability is low bioavailability (after a single oral dose of nifedi-
(15% after a single dose, 22.6% after repeated ad- pine 20mg) was almost doubled (90.5 vs 51.1%).[46]
ministration) because of its extensive first-pass he- In 3 patients with surgical portacaval shunts the
patic metabolism;[43] protein binding is about 93%. bioavailability was 100%. The unbound fraction of
It is largely metabolised by the liver. Liver disease nifedipine was almost doubled (8.5 vs 4.4%).
markedly alters the kinetics of gallopamil. In 6 pa- The capacity of the liver to metabolise nifedi-
tients with Child-Pugh class B cirrhosis who re- pine was reduced in those patients with cirrhosis,
ceived a single oral dose of gallopamil 25mg, the as shown by a decreased drug clearance. However,
CL/F was greatly reduced (1.82 L/min compared this reduction alone is not sufficient to account for
with 8.2 L/min in healthy individuals).[44] Conse- such a high bioavailability. Other factors include
quently, bioavailability increased to 60% and t1⁄2 the presence of portacaval shunts, with their syn-
was prolonged to 11 hours (compared with 3 to 6h ergistic effect on bioavailability.
in healthy individuals). The changes in pharmaco- Another study[47] reported a marked reduction
kinetics were accompanied by a greater drug effect in the elimination of nifedipine after a single oral
in patients with cirrhosis compared with that found dose of 10mg in patients with cirrhosis (all patients
in healthy volunteers. Based on these findings, I had portal hypertension and were largely decom-
suggest reduction of the oral dosage of gallopamil pensated) as shown by a 4-fold increase in the t1⁄2
to 25 to 50% of the normal dosage. and a 2-fold increase in AUC. Clearly, patients
with cirrhosis are at risk of nifedipine accumula-
4.3.3 Nicardipine tion and a dosage reduction (to half the normal dos-
Nicardipine has a low bioavailability (about age) is required.
11%) because of an extensive first-pass liver me-
tabolism.[40] It is 98 to 99% protein bound. After a 4.3.5 Nimodipine
single oral dose of nicardipine 30mg, 9 cirrhotic Nimodipine has a low bioavailability (of about
patients with impaired liver function (abnormal 11.6%) and protein binding of about 98%.[40]
routine laboratory tests, antipyrine AUC reduced In 6 patients with cirrhosis after a single oral
by 65%) showed a reduction in drug elimina- dose of nimodipine 60mg, CL/F was decreased by
tion,[45] demonstrated by substantially higher AUC almost 60% and AUC was 4 times greater than in
and much longer t1⁄2 than that found in the matched healthy individuals.[48] It was not reported whether
control group (AUC 432 vs 97 mg/L • h and t1⁄2 13.7 or not the patients had portacaval shunts, so it is
vs 1.6h). The pharmacological response to the drug not possible to determine the relative contribution
was also greater, which is in keeping with the of reduced clearance and blood shunting to the
higher plasma concentrations. These findings indi- greatly increased AUC. It seems that the decrease
cate that the dosage of nicardipine should be re- in clearance alone cannot account for a 4-fold in-
duced in patients with liver impairment. crease in AUC. In the patients with cirrhosis there

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
408 Rodighiero

was a significant reduction in mean arterial pres- nous nitrendipine the patients with cirrhosis had a
sure, which was significantly related to the nimodi- significantly altered drug kinetics; patients with
pine concentrations, while there were no signifi- chronic hepatitis had similar alterations but to a
cant changes in the healthy controls. In this study lesser extent [CL 51.2 L/h in patients with cirrho-
no data are reported to assess the degree of liver sis, 50.4 L/h in patients with chronic hepatitis and
impairment. 77.4 L/h in healthy individuals; AUC to 24 hours
Considering the marked changes in pharmaco- (AUC24) was 128, 118 and 61.8 μg/L • h, respectively,
kinetics, the dosage of nimodipine may be halved for the same groups; t1⁄2 was 7.67, 3.79 and 2.19h,
in patients with liver impairment. However, more respectively, for the same groups]. Following re-
detailed studies are needed. peated oral administration, significant accumula-
tion of the drug was observed in patients with liver
4.3.6 Nisoldipine
disease.
Nisoldipine has a bioavailability of about 3.9%
Accumulation of nitrendipine after repeated ad-
and protein binding of >99%.[40] The pharmacoki-
ministration was also found in another study.[52] In
netics of nisoldipine are markedly affected in pa-
6 patients with alcoholic cirrhosis (after a single
tients with liver disease. In 8 patients with cirrho-
10mg oral dose on day 1 and then 10mg twice daily
sis,[49] mostly with ascites and 2 with a mesocaval
for 6 days) there was a marked increase in AUC on
shunt, after a single dose of nisoldipine (as either
day 1 (119 vs 29 μg/L • h in healthy individuals)
an intravenous dose of 0.37mg or an oral dose of
and at steady state drug accumulation was evident
5mg) there was a 42% decrease in CL (29.64 vs
(AUC 165 vs 47 μg/L • h in healthy individuals),
50.82 L/h in the control group), a prolonged t1⁄2
while the t1⁄2 and the elimination rate constant were
(16.6 vs 9.7h in the control group) and a 4-fold
not significantly different in the 2 groups. These
increase in oral bioavailability (14.7 vs 3.7%),
data were considered consistent with a marked in-
probably because the portacaval shunts and the re-
crease in the systemic bioavailability of nitrendip-
duced hepatic function had a synergistic effect on
ine, probably as a result of portosystemic shunting.
bioavailability. Because of these alterations, a re-
duction in the dosage of nisoldipine was recom- As a result of the altered kinetics of nitrendip-
mended in patients with cirrhosis. However, in this ine, dosage adjustments should be made in patients
study very heterogeneous diseases were grouped with liver disease according to the haemodynamic
together. response.
In another study,[50] major modifications in the
kinetics of nisoldipine were observed in patients 4.4 Ketanserin
with chronic liver disease. AUC and t1⁄2 were in-
Ketanserin is a serotonin (5-hydroxytryptamine;
creased, and clearance was decreased.
5HT) 5HT2 receptor antagonist with a weak α1-
4.3.7 Nitrendipine adrenoceptor antagonistic activity used for the
Nitrendipine has a low oral bioavailability treatment of patients with hypertension and vaso-
(about 16%) and a protein binding of about spastic disorders.[53] After oral administration it is
98%.[40] Liver disease considerably modifies the subject to first-pass effect in the liver (bioavailabil-
kinetics of nitrendipine. Nitrendipine was given to ity of about 50%) and is extensively metabolised
11 cirrhotic patients with impaired liver function by the liver, only 1% being excreted unchanged in
and to 5 patients with chronic hepatitis (a single the urine. Ketanserin is about 95% protein bound,
5mg intravenous dose and 20mg orally once daily mainly to albumin. In 26 patients with cirrhosis
for 7 days).[51] Impairment of liver function was after a single intravenous dose of 5mg, or 20 or 40mg
quantified using antipyrine clearance (patients with orally, CLp was decreased by 48% compared with
cirrhosis 1.07 L/h and with chronic hepatitis 2.18 healthy controls, and the AUC was 3-fold higher
L/h; healthy individuals 2.50 L/h). After intrave- with the 20mg dose and 2- to 4-fold higher with the

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
Effects of Liver Disease on Pharmacokinetics 409

40mg dose.[54] The t1⁄2 was similar in the 2 groups. 4.5.2 Mexiletine
The decrease in clearance is not sufficient to ac- Mexiletine is a class Ib antiarrhythmic drug with
count for such a large increase in AUC. It is likely an oral bioavailability of about 90%.[58] It is exten-
that the increase in AUC may be because of the sively metabolised by hepatic CYP2D6 to inactive
existence of spontaneous portacaval anastomoses metabolites.[7] The plasma protein binding of mex-
in patients with cirrhosis. Unfortunately, the authors iletine is about 70%.
did not report the degree of severity of cirrhosis. The elimination of mexiletine is markedly im-
These findings suggest a reduction in the oral paired in patients with advanced alcoholic cirrho-
dosage of ketanserin or an increase in the adminis- sis after a single intravenous doses of 200mg.[59]
tration interval; the dosage should not exceed This study involved 6 patients with abnormal rou-
20mg twice daily. tine laboratory tests and antipyrine clearance
(0.013 L/h/kg vs 0.028 L/h/kg in healthy individu-
4.5 Antiarrhythmics als) and showed decreased CL, a greatly increased
AUC and prolonged t1⁄2 (CL 0.139 vs 0.496 L/h/kg
4.5.1 Flecainide in healthy controls, AUC 21.6 vs 6.25 mg/L • h and
Flecainide is a class Ic antiarrhythmic agent. It t1⁄2 28.7 vs 9.9h). On the basis of these findings the
is not subject to presystemic metabolism, the bio-
usual loading dose, if it is needed, can be given to
availability being approximately 95%.[55] Flecain-
patients with cirrhosis, but the maintenance dosage
ide is 30 to 40% bound to plasma proteins. It is
should be reduced to a quarter or one-third of the
metabolised by hepatic CYP2D6,[7] with about 25%
usual dosage. In this study the number of patients
being excreted unchanged in the urine. Liver dis-
was small and only patients with severe hepatic
ease markedly affects the kinetics of flecainide.
impairment were included.
Six cirrhotic patients with impaired liver func-
tion[56] (i.e. with abnormal values of routine labo-
4.5.3 Propafenone
ratory tests, antipyrine clearance 0.852 vs 3.192 Propafenone is a class Ic antiarrhythmic agent.
L/h in healthy individuals) were given a single in-
It has a low and variable oral bioavailability (rang-
travenous dose of flecainide 2 mg/kg. Liver im-
ing from 5 to 31%) because of its hepatic first-pass
pairment resulted in a decreased CLp for flecainide
effect[60] and protein binding of about 96%. It is
(0.23 vs 0.55 L/h/kg in healthy individuals), an in-
extensively metabolised by hepatic CYP2D6.[7] In
creased AUC (12 868 vs 3805 μg/L • h) and a
8 patients with moderate to severe hepatic dysfunc-
greatly prolonged t1⁄2 (49 vs 9.5h). Thus, in patients
tion caused by alcoholic cirrhosis (severity of dis-
with liver disease, flecainide can reach unaccept-
ably high plasma concentrations with the usual ease assessed with routine laboratory tests, pres-
dosage regimen. Caution should be used: therapy ence of varices and ICG clearance) given a single
should be started with a low dosage while monitor- dose (intravenous 1 mg/kg, oral 150mg), an in-
ing the therapeutic response and plasma drug con- crease in bioavailability, decrease in CL and ten-
centration. dency for an increase in free drug was observed,
Flecainide is partly excreted unchanged in the the changes being more marked with the increasing
urine and elimination is reduced in patients with severity of liver dysfunction.[61] On the basis of
kidney function impairment.[57] Thus, in patients these findings it is recommended to reduce the dos-
with liver disease both hepatic and renal function age of propafenone, depending on the severity of
should be assessed. If liver and kidney impairment hepatic impairment and to start with a low dose
coexist, the use of flecainide is dangerous and with ECG monitoring. Thus, at least a 50% reduc-
probably best avoided. tion in dosage is recommended.

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
410 Rodighiero

4.6 Hypolipidaemics nation, renal and hepatic, suggest a potential for


compensatory excretion by alternative routes in pa-
The HMG-CoA reductase inhibitors (statins) tients with renal or hepatic impairment. In patients
are reversible competitive inhibitors of HMG-CoA receiving pravastatin both renal and hepatic func-
reductase, the enzyme responsible for conversion tion should be evaluated. Caution should be used
of HMG-CoA to mevalonic acid, thereby reducing in administering pravastatin to patients with he-
endogenous cholesterol synthesis and lowering patic disease; I recommend that they are started at
plasma cholesterol levels. a lower dosage of 10 mg/day. In such patients, both
4.6.1 Fluvastatin
liver and kidney function should be evaluated.
Fluvastatin, which is almost completely ab-
sorbed (98%) from the gastrointestinal tract, has an 5. Diuretics
absolute oral bioavailability of between 20 and
30% because of its extensive first-pass metabo- 5.1 Torasemide
lism.[62] Fluvastatin is 99% bound to plasma pro-
teins, mainly albumin, and is almost completely Torasemide is a new loop diuretic used for the
metabolised to inactive metabolites, perhaps by treatment of patients with oedema caused by con-
CYP3A.[7] After a single oral dose of fluvastatin gestive heart failure, renal failure and cirrhosis and
40mg, 11 cirrhotic patients with hepatic insuffi- for the treatment of patients with hypertension.[66]
ciency (Child-Pugh score ≤10) had a plasma CL/F The absolute bioavailability is approximately 80%.
28% less than that of matched controls and a 2.5- It is highly bound (>99%) to plasma proteins. Torase-
fold increase in both AUC and Cmax, with no dif- mide differs from other loop diuretics because it is
ference in the t1⁄2 between the 2 groups (AUC 795 extensively metabolised by the hepatic CYP2C9;
vs 304 μg/L • h and Cmax 683 vs 269 μg/L).[63] only 20% of the drug is excreted unchanged in the
urine.
Lower dosages of fluvastatin are recommended
for patients with hepatic insufficiency. Several studies have shown that the kinetics of
torasemide are altered in patients with ascites
4.6.2 Pravastatin caused by cirrhosis. After a single intravenous or
Pravastatin has a low oral bioavailability (about oral dose of torasemide 10mg in 12 patients,[67] a
17 to 18%), which is attributed both to incomplete higher bioavailability (96%), decrease in CL and
absorption (34%) and extensive first-pass metabo- nonrenal clearance, increase in renal clearance
lism in the liver (extraction ratio 0.66).[64] About with a greater percentage of the dose excreted in
50% of the drug is bound to plasma proteins. Both the urine and prolongation of t1⁄2 were observed [CL
renal and hepatic routes are responsible for the 2.28 vs 2.58 L/h in healthy individuals, renal clear-
elimination of pravastatin and its metabolites, ac- ance (CLR) 0.546 vs 0.384 L/h, nonrenal clearance
counting for 47 and 53% of total clearance, respec- (CLNR) 1.752 vs 2.196 L/h and t1⁄2 8.1 vs 3.6h]. Pa-
tively. It is probably metabolised by CYP3A.[7] The tients had a diminished response to the drug.
2 major metabolites are active: 75% of the inhibi- Although this seems paradoxical, patients with
tory activity of pravastatin is attributable to the par- cirrhosis usually have a resistance to loop diuretics
ent drug. because of a pharmacodynamic abnormality.[66]
Patients with biopsy-proven alcoholic cirrhosis Because of the decreased CLNR of torasemide, a
and hepatic insufficiency have an AUC 34% higher greater amount of drug was delivered to the kidney.
than that of matched controls (dose not speci- Since renal function was preserved in patients with
fied).[65] This increase in AUC is attributed to re- ascites, there was a greater excretion of torasemide
duction in hepatic clearance. The impairment of in the urine, the fraction of the dose excreted being
hepatic clearance resulted in a compensatory in- about 70% higher than in healthy individuals.
crease in renal excretion. The dual routes of elimi- Thus, more drug for a longer time reached the renal

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
Effects of Liver Disease on Pharmacokinetics 411

site of action, offsetting the diminished pharmaco- almost completely biotransformed by CYP3A.
dynamics of response. In fact, the natriuretic effect When patients have moderate liver impairment the
was essentially the same as that in healthy individ- changes in midazolam kinetics are modest.[74] Af-
uals. Therefore, the pharmacokinetic changes of ter a single intravenous dose of midazolam 0.2
torasemide in patients with cirrhosis compensate mg/kg in 10 patients with alcoholic cirrhosis and a
for the pharmacodynamic abnormality. moderate degree of liver dysfunction (routine lab-
No change in dosage is needed in patients with oratory tests), CLp decreased by 37% compared
cirrhosis unless there is concomitant impairment with the control group, AUC increased by 57% and
of kidney function. It should be noted that in this t1⁄2 was prolonged by 25% (CLp 24.12 vs 38.22 L/h,
study individual data were widely scattered. The AUC 649 vs 414 μg/L • h and t1⁄2 2.8 vs 2.25h).
authors did not measure plasma protein concentra- The situation is different when patients have se-
tions in the patients; the presence of hypoprotein- vere liver cirrhosis.[75] After a single intravenous
aemia has important effects on a highly bound drug dose of midazolam 0.075 mg/kg, 7 patients with
such as torasemide. severe alcoholic cirrhosis (evaluated according to
In another study,[68] altered kinetics of torasem- routine laboratory tests, antipyrine and ICG clear-
ide were also observed in patients with ascites ance) had a 2-fold prolongation of t1⁄2 as a result of
caused by decompensated liver cirrhosis. a significant decrease in CL (CL 0.32 vs 0.62
L/h/kg, t1⁄2 3.9 vs 1.6h). The patients experienced
6. Psychoactive and greater sedation than the control group. Psychomo-
Anticonvulsant Agents tor function was also evaluated (reaction time and
critical flicker fusion time); there was a decrease
6.1 Benzodiazepines in this function, which was attributed to an in-
creased cerebral sensitivity to midazolam.
6.1.1 Alprazolam
Alprazolam has an oral bioavailability of about Similar results were obtained in 7 cirrhotic pa-
80 to 100%[69] with protein binding of between 79 tients with advanced impairment of liver function
and 83% (mainly to albumin). It is extensively me- as assessed with routine laboratory tests, antipy-
tabolised by hepatic CYP3A. In 17 patients with rine and ICG clearance.[76] The patients were given
alcoholic cirrhosis without ascites (abnormal rou- a single intravenous or oral dose of midazolam (7.5
tine laboratory tests, ICG t1⁄2 10.5 vs 2 to 4 min in or 15mg, respectively) and had a decrease in CL
healthy individuals)[70] given a single oral dose of (55.7 vs 93.8 L/h/kg in controls) with a consequent
alprazolam 1mg, the impairment of liver function prolongation of t1⁄2 (7.36 vs 3.80h) and increase in
resulted in a 54% decrease in the CL/F of al- AUC (543 vs 298 μg/L • h). The bioavailability was
prazolam with a corresponding increase in t1⁄2 and 76 vs 38%.
AUC as compared with healthy matched individu- Midazolam should be used with caution in pa-
als (CL/F 0.034 vs 0.073 L/h/kg, t1⁄2 19.7 vs 11.4h, tients with cirrhosis, and the dosage should be re-
AUC 529 vs 220 μg/L • h). A reduction in the daily duced by 50%.
dosage by half is suggested.
6.1.3 Triazolam
6.1.2 Midazolam Triazolam has an oral bioavailability of be-
Midazolam has an oral bioavailability in the tween 44 and 53% and is about 89% bound to
range of 34 to 68%, because the first-pass effect plasma proteins.[71] It is extensively metabolised
depends on the dose.[71] It undergoes significant by hepatic CYP3A. Eight patients with cirrhosis
presystemic metabolism in the small intestine by were given a single oral dose of triazolam 0.25mg
hepatic CYP3A;[72,73] high levels of CYP3A are (the severity of liver disease was assessed accord-
present in the intestine.[26] The plasma protein ing to ICG clearance, partial thromboplastin time
binding of midazolam is about 95%. Midazolam is and albumin concentration).[77] Liver impairment

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
412 Rodighiero

reduced the CL/F (0.299 vs 0.401 L/h/kg in healthy benzodiazepine receptor.[80] It is indicated when
individuals), the clearance being correlated with the central effects of a benzodiazepine need to be
the severity of liver disease. This was accompanied attenuated or terminated. It is 40 to 50% bound to
by an increase in AUC (15.5 vs 9.51 μg/L • h). The plasma proteins. The oral bioavailability averages
values of CL/F and AUC varied widely. Psychomo- 16% because of a high first-pass effect. Flumazenil
tor performance and memory tests were adminis- is extensively metabolised by the liver to inactive
tered; the patients with cirrhosis showed greater metabolites.
psychomotor impairment than healthy individuals. Eight patients with moderate alcoholic cirrhosis
Similar results have been found in another (Pugh’s score ranging from 5 to 11) had impaired
study.[78] Eight patients with stable cirrhosis treated elimination of flumazenil[81] after a single dose of
with a single oral dose of triazolam 0.25mg (sever- flumazenil (30mg oral or 2mg intravenous); this
ity of disease assessed with aminopyrine breath was caused by a decreased CLp with a consequent
test, galactose and ICG clearance) had a significant prolongation of t1⁄2 and a more than doubled AUC
increase in triazolam free fraction (16.9 vs 13.8% [CLp 42.3 vs 72.06 L/h in matched healthy volun-
in healthy individuals), reduced CL/F of the un- teers; t1⁄2 (intravenous) 1.4 vs 0.79h; t1⁄2 (oral) 1.3 vs
bound drug (0.888 vs 0.143 L/h/kg) which corre- 0.88h]. The oral bioavailability is more than dou-
lated with the aminopyrine breath test and pro- bled (65.2 vs 27.8%).
longed t1⁄2 (9.7 vs 3.9h). In patients with cirrhosis, In conclusion, patients with liver function im-
excessive sedation was observed and attributed not pairment have a reduced elimination of flumazenil.
only to impaired drug elimination but also to brain Acute intravenous administration of the drug to
hypersensitivity. The psychomotor capacity of the these patients only results in a longer duration of
patients was assessed with different psychometric action. Since the drug is well tolerated and has a
tests (flicker sensitivity, pursuit rotor and digit wide therapeutic index with a short half-life, no
symbol substitution test) when plasma concentra- serious problems should arise. If long term oral use
tions of unbound triazolam were the same in is anticipated, somewhat lower dosages are sug-
healthy individuals and patients with cirrhosis (i.e. gested, especially if blood shunting is present be-
2.25h after administration). The results of the tests cause flumazenil has a high first-pass effect. The
showed that the average impairment in the perfor- effects of this drug are dose-related.
mance of patients with cirrhosis was 30% greater
than in healthy individuals. 6.2 Antidepressants
Because of pharmacokinetic changes in patients
6.2.1 Selective Serotonin Reuptake Inhibitors
with cirrhosis, the dosage of triazolam should be Selective serotonin reuptake inhibitors (SSRIs)
adjusted: a 50% reduction of the dosage is advis- are antidepressant drugs with a common mecha-
able. nism of action.[82] They selectively and reversibly
The oxidative pathway is more sensitive to liver block the reuptake of serotonin, binding to the pre-
damage than glucuronidation, even though glucuron- synaptic serotonin reuptake carrier in the CNS. All
idation is not spared when liver disease is se- SSRIs are extensively metabolised in the liver. The
vere.[79] Thus, in patients with liver disease it is SSRIs discussed below are all biotransformed by
better to avoid benzodiazepines undergoing oxida- CYP2D6, although they have different chemical
tion and to use, in preference, benzodiazepines me- structures and different pharmacokinetic charac-
tabolised by conjugation with glucuronic acid. teristics.
6.1.4 Flumazenil Fluvoxamine
Flumazenil is a specific benzodiazepine antag- Fluvoxamine is subject to a significant first-pass
onist which reverses benzodiazepine-induced CNS effect, with an oral bioavailability of about
depression by competing with these drugs for the 50%;[83] protein binding is about 77%. The meta-

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
Effects of Liver Disease on Pharmacokinetics 413

bolites are inactive. In 13 patients with alcoholic dosage increases.[87] The kinetics of paroxetine in
liver cirrhosis [severity of disease assessed with 12 cirrhotic patients with moderate hepatic impair-
routine laboratory tests and bromosulphophthalein ment (routine laboratory tests) were investigated
(BSF) clearance], after a single oral dose of flu- by giving a single 20mg oral dose;[88] no signifi-
voxamine 100mg the AUC and the t1⁄2 were about cant difference was found as compared with
50% higher than in healthy volunteers.[84] This in- healthy individuals. However, a single dose study
crease is evidence that fluvoxamine metabolism is is not very meaningful because paroxetine is ad-
reduced (CL/F 54.8 L/h) in this patient group. ministered to patients for an extended period of
There is great interindividual variability. time: a multiple dose study is clinically more rele-
It is recommended that patients with liver dys- vant. In 12 patients with alcoholic cirrhosis and
function should be given a lower initial daily dos- reduced liver function,[89] repeated oral adminis-
age with a longer interval between doses and this tration of paroxetine 20 mg/day for 14 days re-
should be followed by careful monitoring. sulted in the doubling of AUC and t1⁄2 compared
with healthy individuals. The patients had a re-
Fluoxetine
duced clearance of antipyrine and galactose (1.74
The absolute bioavailability of oral fluoxetine
L/h and 78 nmol/h, respectively, vs 3.66 L/h and
in dogs is about 72% (human data is not avail-
150 nmol/h in controls). The data had a great inter-
able).[85] The main metabolite of fluoxetine is
individual variability. These findings suggest that
norfluoxetine, which has a similar potency and se-
in patients with hepatic impairment the initial
lectivity as the parent compound. The kinetics of
dosage of paroxetine should be at the lower end of
fluoxetine are nonlinear. Binding to plasma pro-
the range recommended for those with normal liver
teins is 95%. In 13 patients with stable alcoholic
function, starting with a daily dosage of 10mg.
cirrhosis (characterised by routine laboratory tests
and ICG clearance) given a single oral dose of
6.2.2 Nefazodone
fluoxetine 40 or 60mg, metabolism was signifi- Nefazodone is a new antidepressant agent with
cantly reduced,[86] as shown by a 56% decrease in a mechanism of action different from that of cur-
CL/F and prolongation of t1⁄2 (6.6 vs 2.2 days in rently available antidepressants.[90] It potently and
healthy individuals). selectively blocks postsynaptic serotonin recep-
The kinetics of norfluoxetine are also altered: tors. It undergoes extensive first-pass metabolism
CL/F is decreased by 30% and t1⁄2 prolonged (12 vs (oral bioavailability of about 20%) and it is largely
6.4 days in healthy individuals). Therefore, upon metabolised by hepatic CYP3A, the main metabo-
repeated administration, excessive accumulation lite (hydroxynefazodone) being active. It is highly
of the drug can be expected, thus increasing the risk bound to plasma proteins (>99%). Because of im-
of toxicity and exaggerated pharmacological re- paired hepatic function in patients with cirrhosis
sponse. (12 patients, 6 class A and 6 class B Child-Pugh
Based on the pharmacokinetic modifications classification; single oral dose of nefazodone
observed in the patients with cirrhosis, a lower dos- 100mg),[91] systemic exposure to nefazodone and
age (about a 50% reduction) or prolonged interval hydroxynefazodone is about 2-fold higher than in
should be used. healthy volunteers and approximately 25% higher
Paroxetine after repeated administration (100mg twice daily
Paroxetine has an oral bioavailability of about for 8 days), probably because nefazodone inhibits
50% and protein binding of approximately 95%. its own metabolism. The data have a large interin-
The main enzyme, CYP2D6, involved in the meta- dividual variability.
bolism of paroxetine is saturable. Its saturation ac- The results with the single dose reported above
counts for the nonlinear pharmacokinetics ob- are consistent with those reported in another study
served during repeated administration or after by the same authors.[92] 12 patients with cirrhosis

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
414 Rodighiero

and 12 healthy controls (antipyrine clearance 2.19 6.3 Tiagabine


vs 4.48 L/h, respectively) were given increasing
single oral doses of nefazodone (50, 100 and 200mg). Tiagabine, a new drug effective in the manage-
Increasing doses resulted in supraproportional in- ment of patients with refractory partial epilepsy,
creases in AUC (388, 1180 and 2890 μg/L • h, re- prevents γ-aminobutyric acid (GABA) uptake by
spectively, in patients with cirrhosis vs 195, 705 and inhibiting one of the GABA transporters (GAT-1)
2130 μg/L • h, respectively, in controls). This is responsible for the uptake of the neurotransmitter
indicative of nonlinear pharmacokinetics. In this into neurons and glial cells after synaptic release,
study, too, there was a large interindividual vari- thus increasing synaptic GABA concentrations and
ability of pharmacokinetic parameters. reducing neuronal excitability.[95]
In conclusion, the findings of these studies in- Tiagabine has an oral absolute bioavailability of
dicate that lower than usual dosages of nefazodone about 90% and is extensively metabolised by he-
should be considered in patients with hepatic im- patic CYP3A. About 96% of the drug is bound to
pairment, starting with 50mg twice daily with the plasma proteins.
drug response monitored. The elimination of tiagabine is reduced in pa-
tients with liver impairment.[96] Patients with mild
6.2.3 Moclobemide and moderate impairment of hepatic function (4
Moclobemide is a reversible and selective in- class A, 3 class B Child-Pugh classification) who
hibitor of the enzyme monoamine oxidase subtype were given oral tiagabine 4mg twice daily for 5
A (MAO-A).[93] It is subject to substantial hepatic days showed a decreased drug elimination caused
first-pass metabolism. The oral bioavailability of by liver function impairment, as shown by the sig-
moclobemide is about 55% with a single 100mg dose, nificantly lower elimination rate constants, in-
but is higher after repeated doses (about 85%), creased AUC and longer t1⁄2 (AUC 632 μg/L • h in
probably because the drug metabolism is a satura- mild impairment, 674 μg/L • h in moderate impair-
ble process. Moclobemide is almost completely ment and 395 μg/L • h in healthy individuals; t1⁄2
metabolised by hepatic CYP2C19. The binding to 11.7, 15.9 and 6.5h, respectively). The patients
plasma proteins (mainly albumin) is about 50%. with moderate hepatic impairment had a higher
The kinetics of moclobemide are altered in pa- fraction of unbound drug (5.13 vs 3.59% in healthy
tients with cirrhosis. Twelve cirrhotic patients with individuals) which was consistent with a reduced
a drastically reduced liver function,[94] assessed level of albumin (2.8 g/dl). Patients with hepatic
with antipyrine and ICG clearance, were given a disease had a higher incidence of adverse effect,
single oral 100mg dose or intravenous 90mg dose mostly neurological. The drug should be given
of moclobemide. They had a lower CL and in- with caution to epileptic patients with impaired
creases in bioavailability, maximum concentration liver function. Patients should be closely moni-
and t1⁄2 (CL 14.6 vs 37.3 L/h in healthy individuals, tored because of potential increased incidence of
F 84 vs 56%, Cmax 1607 vs 582 μg/L and t1⁄2 3.87 vs neurological adverse effects. Reduced dosages or a
1.67h). The parameters reported above show a sub- longer interval, or both, may be needed. Even
stantial interindividual variation, reflecting a wide though the number of patients studied was small, it
range of age in the group of patients with cirrhosis. can be said that if liver impairment causes a de-
In the presence of hepatic impairment the dos- crease in plasma proteins the fraction of unbound
age of moclobemide should be reduced to half or tiagabine will increase and therefore increase the
one-third of the usual dosage, or the administration effect of the drug, thus making it dangerous to ad-
interval should be prolonged, to avoid excessive ministration tiagabine to patients with severe he-
accumulation of the drug after repeated administra- patic impairment because of the very high inci-
tion. dence of neurological adverse effects.

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
Effects of Liver Disease on Pharmacokinetics 415

7. Antiemetics 7.2 Serotonin Antagonists

Antiemetic drugs are widely used to treat nau- Serotonin antagonists are a new class of anti-
sea and vomiting from a variety of causes, but es- emetics which are selective antagonists of 5HT3
pecially in patients undergoing cancer chemother- receptors. Ondansetron and tropisetron are repre-
apy. Little work has been done on the influence of sentatives of this class of drug.
liver disease on the kinetics of these drugs.
7.2.1 Ondansetron
7.1 Metoclopramide Ondansetron has an oral bioavailability of about
60% because of first-pass metabolism.[100] It is 70
Metoclopramide is subject to a first-pass effect, to 76% protein bound and extensively metabolised
but there are wide interindividual variations; the by hepatic CYP2D6, 1A2 and 3A.[7] Its kinetics are
oral bioavailability ranges from 32 to 97%.[97] It is affected by liver disease, depending on the degree
about 40% bound to plasma proteins and metabo- of hepatic impairment. This has been shown in 2
lised to sulphate and glucuronide conjugates, 20% studies in patients with varying degrees of hepatic
being excreted unchanged in the urine. impairment (mild, moderate and severe according
In 8 patients with severe alcoholic cirrhosis and to Child-Pugh classification).
marked hepatic impairment (Child-Pugh class C) 19 patients with chronic liver disease (type of
given single intravenous and oral doses of meto- disease not specified) and hepatic impairment (6
clopramide 20mg a 50% decrease in CL was ob- class A, 6 class B, 7 class C) were given a single
served.[98] This decrease resulted in a correspond- intravenous dose of ondansetron 8mg.[101] The pa-
ing prolongation of t1⁄2 (15 vs 7h in healthy tients with mild to moderate hepatic impairment
volunteers) and similar increases in AUC after both had similar pharmacokinetic changes: a reduction
intravenous and oral administration (AUCIV 1689 in CLp, an increase in AUC and a prolongation of
vs 749 μg/L • h, AUCoral 1559 vs 689 μg/L • h). t1⁄2 (CLp 12.7 to 17.9 vs 28.7 L/h in healthy individ-
Changes in metoclopramide kinetics were also uals, AUC 633 to 646 vs 279 μg/L • h, t1⁄2 9.1 to 9.2
found in another study.[99] 18 cirrhotic patients vs 3.6h); the changes were much more marked in
with varices, characterised with antipyrine and patients with severe hepatic impairment (CLp 5.76
ICG clearance, were studied after administration L/h, AUC 1383 μg/L • h, t1⁄2 of 20.6h).
of single intravenous and oral doses of metoclopra- Similar results have been obtained in another
mide 0.25 mg/kg. The presence of hepatic blood study[102] of 12 patients with chronic liver disease
shunting determined an increase in bioavailability (type of disease not specified) and varying degrees
and consequently a higher AUC (F 82 vs 60% in of hepatic impairment (4 class A, 4 class B, 4 class
controls and AUC 242 vs 180 μg/L • h). Dosage C) after single intravenous and oral doses of
adjustments are advisable in patients with cirrho- ondansetron 8mg. In patients with severe impair-
sis, especially in the case of prolonged therapy; a ment the bioavailability was almost complete
50% reduction of the dosage is recommended. (98%) with a decreased CL/F (14.1 L/h/kg), in-
In these 2 studies,[98,99] the authors did not re- creased Cmax (56 μg/L) and prolonged t1⁄2 (20h).
port the renal function of patients with cirrhosis. These changes were less pronounced in patients
This parameter is important because 20% of meto- with mild to moderate impairment (F 78 to 80% vs
clopramide is excreted unchanged in the urine. Pa- 60% in matched controls, CL/F 20.2 to 21.4 vs 44.6
tients with renal impairment have a reduced elim- L/h/kg, Cmax 39 to 40 vs 28 μg/L, t1⁄2 10 to 13 vs
ination of the drug. Thus, when severe hepatic 6h).
impairment and reduced kidney function coexist, On the basis of these results, when hepatic im-
the use of metoclopramide is probably best pairment is moderate to severe, a dosage adjust-
avoided. ment is recommended for ondansetron: a once

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
416 Rodighiero

daily dosage of less than 8mg would appear appro- 1.4h).[108] There was no substantial difference be-
priate. tween patients with compensated and uncompen-
sated cirrhosis.
7.2.2 Tropisetron
Tropisetron is subject to dose-dependent first- Similar results were obtained in another
pass metabolism because its kinetics are nonlin- study[109] after repeated oral administration of
ear;[103,104] thus, the bioavailability is 52% for the lansoprazole 30 mg/day for 9 days to 12 patients
20mg dose and 66% for the 100mg dose. Protein with cirrhosis (3 class A, 1 class B, 8 class C Child-
binding of tropisetron is 59 to 71%. Tropisetron is Pugh classification). There was no significant dif-
metabolised by hepatic CYP2D6 and, because of the ference in pharmacokinetics between the first and
polymorphism of this system, there are poor and last dose, indicating that the severity of cirrhosis
extensive metabolisers. In patients with liver cir- had little effect on accumulation of the drug.
rhosis the metabolic clearance (dose not reported) It is recommended that a lansoprazole dosage of
was 50% lower than that of healthy volunteers who 30 mg/day should not be exceeded in patients with
were extensive metabolisers. Accumulation of the impaired liver function.
drug in such patients was considered unlikely at the 8.1.2 Pantoprazole
recommended dosage of 5 mg/day.[104] The elimination of pantoprazole is also consid-
erably impaired in patients with cirrhosis. Twelve
8. Antiulcers patients with cirrhosis (Child class A and B) were
given an intravenous dose of pantoprazole 30mg
8.1 Proton Pump Inhibitors
once daily for at least 5 days and 40mg orally once
Omeprazole is a substituted benzimidazole daily for at least 7 days.[110] At steady state the pa-
which reduces gastric acid secretion by inhibition tients had a greatly increased AUC (33.8 mg/L • h)
of the H+,K+ ATPase in gastric parietal cells.[105] and t1⁄2 (7.8h) caused by the reduced CL (0.0161
Lansoprazole and pantoprazole, compounds which L/h/kg). No accumulation of the drug was observed.
are structurally similar to omeprazole, reduce acid Similar results have been obtained in 12 patients
gastric secretion by the same mechanism.[106,107] with cirrhosis with severe liver impairment given
Lansoprazole has an oral bioavailability of about pantoprazole 40mg orally for 7 days and 30mg in-
85%, pantoprazole about 77%, omeprazole about travenously for 5 days.[111]
60% and all 3 drugs have a low to medium extrac-
8.1.3 Omeprazole
tion ratio. These drugs are completely metabolised Cirrhosis causes marked changes in the pharma-
by hepatic CYP2C19. Plasma protein binding is cokinetics of omeprazole. Eight patients with liver
high for all 3 drugs (>95%). cirrhosis (3 class A, 4 class B, 1 class C Child clas-
8.1.1 Lansoprazole sification) given a single dose of omeprazole 20mg
The effects of cirrhosis on lansoprazole kinetics intravenously or 40mg orally had a 9-fold decrease
are considerable.[108] The results obtained in 16 pa- in CL as compared with healthy volunteers, an al-
tients with compensated or decompensated cirrho- most 2-fold increase in bioavailability, a 7-fold in-
sis and ascites after a single oral dose of lanso- crease in AUC and a 4-fold prolongation of t1⁄2.[112]
prazole 30mg were similar. The severity of liver However, the patients were older (40 to 80 years)
disease was established on the basis of routine lab- than the healthy volunteers (19 to 31 years). Thus,
oratory tests, sulfobromophthalein (SBP) clearance part of the changes observed in the patients may be
and the presence of ascites. The impairment of liver caused by age, as a more than 2-fold decrease in
function resulted in a considerable decrease in the clearance of omeprazole was found in elderly
lansoprazole CL/F (0.04 to 0.07 vs 0.26 L/h/kg in healthy volunteers.[113]
healthy individuals) and a several-fold higher AUC Similar results were obtained in another study[114]
(11.7 to 10.7 vs 2.67 mg/L • h) and t1⁄2 (6.1 to 7.2 vs in 8 patients with alcoholic cirrhosis (severity of

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
Effects of Liver Disease on Pharmacokinetics 417

disease not specified) after administration of a single erate hepatic impairment caused by different types
oral dose of omeprazole 20mg. No accumulation of of liver disease (10 class A and 12 class B Child-
the drug was reported in 2 separate studies[115,116] Pugh) received grepafloxacin 400mg orally for 7
after repeated administration to patients with cir- days. They had reduced CL/F [23.16 (mild impair-
rhosis because the t1⁄2, even though prolonged in ment) and 14.94 (moderate impairment) vs 35.82
patients with cirrhosis (about 3h), is still very short L/h in healthy controls] with an increased AUC (20
in relation to the administration interval (24h). and 28 vs 13 mg/L • h, respectively) and prolonga-
The substantial decreases in clearance of om- tion of t1⁄2 (13 and 19 vs 12h, respectively), these
eprazole, lansoprazole and pantoprazole observed changes being more pronounced in patients with
in patients with impaired liver function are similar moderate than in mild impairment.[121]
for all 3 drugs; this is expected because they are A dosage of grepafloxacin 400mg once daily is
metabolised by the same CYP isoform. As a clinical considered appropriate for patients with mild he-
consequence of reduced elimination, patients with patic impairment, whereas grepafloxacin should not
hepatic impairment would need lower dosages be used in patients with moderate or severe impair-
than those normally used because the degree of ment. One drawback of this study was that the
inhibition of gastric acid secretion is dose related. group of patients had heterogeneous types of liver
Proton pump inhibitors could be used in prefer- disease.
ence to cimetidine and ranitidine in patients with
liver disease because of the CNS adverse effects
experienced in these patients after administration 9.2 Ornidazole
of cimetidine and ranitidine. Several cases of men-
tal confusion have been reported after cimetidine Ornidazole is a nitroimidazole derivative with
administration.[117] There is enhanced penetration activity against protozoa and bacteria.[122] It is a
of cimetidine into the CNS in this patient group; low clearance drug with plasma protein binding of
the amount of cimetidine crossing the blood-brain 8 to 13% and is primarily metabolised by the liver.
barrier is almost doubled compared with that in Hepatic impairment affects the kinetics of this
healthy individuals.[118] Neuropsychiatric compli- drug. Ten patients with severe alcoholic liver cir-
cations in patients with liver dysfunction have been rhosis and ascites were given a single intravenous
reported in association with ranitidine (confusion, dose of ornidazole 500mg. The severity of the dis-
disorientation, hallucinations and delirium), but ease was assessed according to routine laboratory
with a lower incidence than with cimetidine.[119] tests. The patients had a significant reduction in
Ranitidine may cross the blood-brain barrier, but CLp (2.09 vs 3.04 L/h in healthy volunteers) lead-
to a lesser extent than cimetidine. ing to a prolonged t1⁄2 (21.9 vs 14.1h, respec-
tively).[123] Results in agreement with those reported
9. Antimicrobials and Antiretrovirals in the previous study were obtained in 3 groups of
patients with different hepatic diseases (acute viral
9.1 Grepafloxacin hepatitis, alcoholic cirrhosis and extrahepatic cho-
lestasis) after a single intravenous dose of or-
Grepafloxacin is a new broad spectrum fluoro- nidazole 500mg.[124] No significant difference was
quinolone antibacterial. It has an absolute oral bio- observed between pharmacokinetic parameters
availability of about 72% and protein binding of (CLp and t1⁄2) in these 3 groups of patients. What-
about 50%. Grepafloxacin is primarily metabolised ever the type of hepatic disease may be, ornidazole
by hepatic CYP1A2.[120] The kinetics are nonlinear, kinetics are markedly impaired.
probably because of saturation of metabolism. To avoid excessive accumulation of ornidazole
Impairment of liver function influences grepa- after repeated administration in patients with he-
floxacin kinetics. 22 patients with mild and mod- patic impairment, the dosage should be adjusted

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
418 Rodighiero

appropriately and the administration interval eliminated by both renal and nonrenal routes.
should be doubled. About 40% of a dose is excreted unchanged in the
urine, while the remaining is probably metabolised
9.3 Pefloxacin via glucuronidation or other routes. The pharmaco-
kinetics of stavudine are not altered in patients with
Pefloxacin is a broad spectrum antibacterial of
liver disease.
the quinolone group. The oral bioavailability is
In 5 cirrhotic patients with moderate to severe
about 100% and protein binding is between 20 and
hepatic impairment (Child-Pugh classification grade
30%. Biotransformation of pefloxacin in the liver
B or C) given a single oral dose of stavudine 40mg,
is extensive (85 to 90%).[125]
the pharmacokinetic parameters were similar to
Several studies on the pharmacokinetics of
those found in age-matched controls.[130] However,
pefloxacin in patients with hepatic impairment
the number of patients in the study was very small
have been published. The results of these studies
and further research is needed to clarify this is-
are in agreement and suggest hepatic impairment-
sue.
related pharmacokinetic modifications. In one
study,[126] 16 patients with alcoholic cirrhosis and, It is, however, necessary to evaluate the renal
mostly, with ascites were given a single intravenous function of patients with HIV infections because
dose of pefloxacin 8 mg/kg; the severity of disease the pharmacokinetics of stavudine are impaired
was not specified (routine laboratory tests). They when renal function is reduced.[131]
showed a marked decrease in CLp (2.66 vs 8.19
L/h/1.73m2 in healthy individuals) as a result of 9.5 Zidovudine
decreased hepatic metabolism and a significantly Zidovudine is a drug used for the treatment of
longer t1⁄2 (35 vs 11h). patients with the acquired immunodeficiency syn-
Two other studies[127,128] have reported similar drome (AIDS).[132] It undergoes significant first-
values in 25 patients with hepatocellular deficiency pass metabolism with an average bioavailability of
caused by alcoholism (2 class A, 9 class B, 14 class 63%, the binding to plasma proteins being about
C Child-Pugh classification) and in 10 cirrhotic pa- 25% (mainly to albumin). Zidovudine is primarily
tients with ascites (routine laboratory tests). In ad- metabolised via hepatic glucuronidation with a mi-
dition, in both studies a marked accumulation of nor contribution of CYP3A; the main metabolite is
the drug was observed after repeated administra- an inactive glucuronyl derivative (GZDV).
tion. There was also penetration of pefloxacin into
Patients with AIDS have a rather high incidence
the ascitic fluid with accumulation of the drug, the
of chronic hepatitis. Therefore, it is important to
concentration in the ascitic fluid being 72% of that
know how liver disease influences the pharmaco-
in plasma.
kinetics of zidovudine. Several studies have shown
The pharmacokinetic parameters in these 3 studies
that hepatic impairment affects, sometimes mark-
showed a great interindividual variability. In con-
edly, zidovudine disposition.
sideration of the altered pharmacokinetics in pa-
In 8 asymptomatic HIV-infected patients with
tients with hepatic impairment, dosage adjustments
liver disease (chronic hepatitis) and mild hepatic
are advisable: a single daily dose of pefloxacin
impairment (7 with a Child-Pugh score of 5 and 1
400mg should suffice.
with a score of 7) after single intravenous 120mg
9.4 Stavudine
and oral 200mg doses of zidovudine, the CL/F of
the drug was reduced and less GZDV metabolite
Stavudine is a pyrimidine nucleoside analogue formed.[133] The lower clearance (1.57 vs 2.28
with potent activity against HIV and is used in the L/h/kg in healthy individuals) resulted in an in-
treatment of patients with HIV infections. The oral creased AUC (1670 vs 1387 μg/L • h, respectively)
bioavailability is almost 100%.[129] Stavudine is and bioavailability (75%), a prolongation of t1⁄2

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
Effects of Liver Disease on Pharmacokinetics 419

(2.04 vs 1h) and a reduced AUC ratio of GZDV to formulation has been developed:[137] it is a micro-
zidovudine (GZDV/zidovudine 2.8 vs 4.6). emulsion which results in a more extensive and
The pharmacokinetic changes are more marked faster absorption. Even though some presystemic
in HIV seronegative patients with liver cirrhosis of metabolism occurs in the gastrointestinal mucosa,
varying degrees.[134] In these patients (3 grade A, the drug is mainly metabolised in the liver by
5 grade B and 6 grade C Child-Pugh score) after a CYP3A.
single oral dose of zidovudine 200mg, a 4-fold de- Impaired liver function results in a decrease in
crease in the CL/F of zidovudine compared with cyclosporin elimination. This has been shown in
healthy individuals was observed. As a conse- several studies. Renal transplant patients with im-
quence of the reduced clearance, there was a 4-fold pairment of liver function[138] had a reduction in
increase in AUC, a more than 2-fold prolongation CLp (0.525 vs 0.786 L/h/kg in patients with normal
of t1⁄2 and a 4-fold reduction in the AUC ratio of liver function) and a 30 to 50% increase in AUC.
GZDV to zidovudine. The patients were consid- The impaired hepatic function in 5 patients with
ered as whole group because there was a great inter- cirrhosis[139] after an infusion of cyclosporin 2
individual variability of kinetic parameters among mg/kg over more than 2 hours resulted in a marked
the 3 groups of patients and no clear difference in decrease in CL (0.146 L/h/kg) and in a longer t1⁄2
parameters could be established among the groups; (32h) as compared with patients with renal or bone
in addition, the number of patients was small. In marrow transplants.
another study,[135] AIDS patients with liver disease The kinetics of cyclosporin 3 mg/kg intrave-
(type of disease not specified) had pharmacoki- nously were studied in 5 liver transplant recipients
netic changes comparable to those observed in the prior to transplantation and 1 to 3 months post-
latter study.[134] transplantation, when hepatic function was consid-
The findings of these studies suggest that in ered stable.[140] Comparing pre- and post-transplant,
AIDS patients with hepatic impairment, dosage ad- there was an increase in CL (0.21 to 0.37 L/h/kg)
justments of zidovudine may be necessary. I rec- and a decrease in AUC (15 737 to 8811 μg/L • h)
ommend dosage reduction or prolongation of the as liver function stabilised, the post-transplant val-
dosage interval; the dosage could be halved or the ues being consistent with those in healthy volun-
interval between doses doubled. If this is not done, teers.
repeated administration may lead to an excessive Cyclosporin has a narrow therapeutic range
accumulation of the drug increasing the risk of tox- with a number of toxic effects, which are largely
icity. It is of note that the glucuronidation of concentration dependent. The risks of rejection and
zidovudine is affected whatever the severity of toxicity vary inversely with increases or decreases
liver disease, even when impairment is mild. in cyclosporin concentrations. Dosage adjustments
are necessary according to the status of liver func-
10. Immunosuppressants tion.

10.1 Cyclosporin 10.2 Tacrolimus

Cyclosporin is a lipophilic cyclic endecapeptide Tacrolimus is a novel macrocyclic lactone with


with potent immunosuppressive properties used to potent immunosuppressive properties used for pro-
prevent the rejection of transplanted organs and the phylaxis of organ rejection after transplanta-
treatment of autoimmune disorders.[136] Because tion.[141] Approximately 25% of the oral dose is
of the poor absorption of standard formulation available. This low bioavailability can be caused
cyclosporin from the intestine, bioavailability is by either poor oral absorption or gut metabolism.
about 30%, with the binding to plasma lipoproteins Tacrolimus is, in fact, a low clearance drug (blood
being about 34%. To overcome this problem a new clearance of about 6 L/h). The binding to plasma

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
420 Rodighiero

proteins is between 72 and 77%. Tacrolimus is liver via reduction and glucuronidation, the main
completely metabolised by hepatic CYP3A to sev- metabolite being the active 6β-naltrexol.
eral metabolites mainly excreted via the bile, which Heroin addicts are likely candidates for chronic
have a lower immunosuppressive activity than the liver disease. Thus, it is important to assess the ef-
parent drug. fects of liver impairment on the pharmacokinetics
Impairment of liver function affects the elimi- of naltrexone. A single oral dose of naltrexone
nation of tacrolimus. After infusion of tacrolimus 100mg was given to 11 patients with cirrhosis (6
0.15 mg/kg or an oral dose of 0.15 mg/kg, patients with decompensated cirrhosis, Child-Pugh class C,
with moderate to severe liver impairment had a re- and 5 with compensated cirrhosis, Child-Pugh
duced CL (one-third or less than normal), a many- class A and B).[149] Patients with decompensated
fold longer t1⁄2 (38.5h) and a higher oral bioavaila- cirrhosis had a marked increase in the systemic
bility (36%), the changes depending on the severity availability of naltrexone as shown by a greatly
of liver dysfunction.[142] During the immediate increased AUC (1610 vs 200 μg/L • 24h in controls)
postoperative period following orthotopic liver and an impaired conversion of naltrexone to 6β-
transplantation, 9 patients received a continuous naltrexol; patients with compensated cirrhosis had
infusion of tacrolimus 0.15 mg/kg/day for 3 to 6 similar changes but to a lesser extent. The pharma-
days.[143] There was a high variability in the phar- cokinetic alterations paralleled the degree of liver
macokinetic parameters, which was attributed to disease severity.
the variability in the functional status of the liver Adjustments of naltrexone dosage are advisable
in patients after transplantation. in patients with liver dysfunction. It is probably
Elevated plasma concentrations of tacrolimus better to avoid the use of naltrexone in patients with
have been found in transplanted patients with im- severe liver impairment because of these marked
paired liver function[144,145] and were associated alterations in pharmacokinetics.
with a higher incidence of nephrotoxicity (renal
failure).[145,146] In patients with liver dysfunction, 11.2 Tolcapone
high concentrations of tacrolimus metabolites have
been observed, indicating impaired biliary excre- Tolcapone is a potent, specific reversible inhib-
tion of metabolites.[147] itor of catechol-O-methyltransferase (COMT), an
Tacrolimus is a drug with a narrow therapeutic enzyme that metabolises levodopa. It is used for the
index. To avoid overdose and the resulting toxicity, management of patients with Parkinson’s disease.
close monitoring of patients with impaired liver The oral bioavailability is about 68% and the protein
function and appropriate dosage adjustments, with binding is approximately 87 to 88%.[150] Tolcapone
both oral and intravenous formulations, is neces- is a drug with a low extraction and is almost com-
sary. pletely metabolised, largely via glucuronidation to
an inactive metabolite.
11. Miscellaneous Agents Tolcapone was given as single doses, 200mg
orally and 50mg intravenously, to 8 patients with
11.1 Naltrexone moderate noncirrhotic liver disease (type of dis-
ease not specified), 8 patients with moderate liver
Naltrexone is a potent competitive antagonist of cirrhosis (Child-Pugh class A) and 8 healthy indi-
opioids at the receptor level and is used for the viduals.[150] Although total tolcapone bioavaila-
treatment of patients with heroin addiction to pre- bility, CL and volume of distribution were similar
vent relapse.[148] Its oral bioavailability ranges be- in the 3 groups, the kinetics of unbound tolcapone
tween 5 and 60% because of its extensive first-pass were modified in the 2 groups of patients, particu-
metabolism in the liver; the plasma protein binding larly in patients with moderate cirrhosis. The patients
is about 20%. It is mainly biotransformed by the had an increased unbound fraction of drug because

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
Effects of Liver Disease on Pharmacokinetics 421

of a decrease in albumin levels (unbound fraction There are different types of liver disease but most
0.20% in the patients with cirrhosis, 0.14% in non- studies have examined the disposition of drugs in
cirrhotic disease, 0.12% in controls). The CL/F and patients with liver cirrhosis, especially alcoholic
the volume of distribution at steady-state (Vss) of un- cirrhosis and, less frequently, chronic hepatitis.
bound drug were markedly decreased (CL/F 3830 This is probably because cirrhosis and chronic hep-
L/h in patients with cirrhosis, 5528 L/h in patients atitis are among the more widespread types of liver
with noncirrhotic liver disease and 6874 L/h in pathology. The data have shown that the pharmaco-
controls; Vss 4787, 6050 and 7371L, respectively). kinetics can be altered even in patients with mild
Thus, it can be expected that the average con- liver impairment. Data on hepatic drug elimination
centration of unbound tolcapone would be twice as in patients with primitive or metastatic cancer are
high in patients with cirrhosis than in those with scanty,[4] but the pharmacokinetics do not appear
healthy liver function. Therefore, the dosage of the to be affected when the only liver pathology is can-
drug can be reduced by 50% in patients with mod- cer without complications.
erate cirrhosis. Further studies are needed to assess Changes in pharmacokinetics depend on the se-
the kinetics of tolcapone in patients with severe verity of liver disease. However, many of the stud-
hepatic impairment. A drawback of this study is the ies compared patients with different degrees of
heterogeneity of liver diseases considered. liver impairment, in others the degree of severity
was not reported and in some studies patients with
11.3 Toremifene
different types of liver disease were grouped to-
Toremifene is a nonsteroidal estrogen antago- gether. Thus, it is not easy to generalise from indi-
nist used in the treatment of patients with breast vidual studies. In addition, the number of participants
cancer.[148] It exerts antitumour activity by specific included in several studies was small. There was a
and competitive inhibition of the binding of estro- tendency to use small samples in these studies, and
gen to its receptor. Toremifene has a bioavailability thus caution should be used in drawing conclu-
of about 100% and it is >99% bound to plasma sions.
proteins, mainly albumin. It is extensively metabo- Usually, the kinetics of drugs are evaluated in
lised in the liver by CYP3A. Thus, liver function patients having only liver disease. However, these
should be considered when evaluating the pharmaco- drugs are used to treat other diseases, such as car-
kinetics of toremifene in clinical practice. diovascular illnesses, psychiatric disorders and so
Ten patients with impaired liver function (de- on. These diseases can themselves cause patho-
gree not specified) due to alcoholic liver disease physiological changes in the body, which can af-
had a significantly decreased drug elimination af- fect the kinetics of the drug administered because
ter a single oral dose of toremifene 120mg as the drug is not working in a normal environment.
shown by the reduced CL/F (3.7 vs 5.1 L/h in It would be more appropriate, for instance, to study
healthy individuals), increased AUC (44.5 vs 28.4 the kinetics of an antidepressant agent in patients
mg/L • h) and prolonged t1⁄2 (10.9 vs 6.2 days).[149] with liver disease and depression using a control
Caution is advised when toremifene is adminis- group of patients with depression. The results
tered to patients with liver impairment; a lower would be more consistent.
dosage is recommended. Very often, in studies evaluating the influence
of liver disease on pharmacokinetics the total drug
12. Clinical Implications clearance is measured and drug binding to plasma
proteins is not considered. Liver disease can de-
12.1 Altered Pharmacokinetics
crease plasma proteins, with a consequent increase
The studies reported above have shown that in unbound drug for high extraction ratio drugs. A
liver disease impairs the kinetics of many drugs. significant decrease in the clearance of unbound

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
422 Rodighiero

drug, with little or no change in total drug clear- equate tissue is not obtained to exclude sampling
ance, was observed when the unbound drug con- errors.
centration was taken into consideration.[150,151] The Dynamic tests of liver function have long been
recommendation to lower the dosage should, there- used. Such tests include measuring the plasma
fore, be based on the clearance of the unbound drug clearance of antipyrine, ICG, galactose and SBP.
rather than on total clearance. To measure only total However, these tests are cumbersome for clinical
drug clearance could be deceiving. purposes and may not accurately reflect changes in
Most of the studies reported are single dose liver histological status and the severity of liver
studies. In clinical practice, patients are commonly disease. The SBP test has been largely abandoned
treated for a prolonged period of time. Upon repeated in clinical use because severe systemic reactions,
administration the drug accumulates until it reaches possibly fatal, can occur.[152] Another method
a steady-state concentration; the length of time to widely used is the Child-Turcotte and the Child-
steady state varies depending on the half-life of the Pugh classification.[11,12] These classifications are
drug. When drug clearance is reduced, and bio- semiquantitative tests of liver function, but do not
availability and half-life are increased by liver dis- provide an accurate evaluation of hepatic impair-
ease, the drug accumulates at a higher rate and ment.[153,154] They are somewhat empirical and the
reaches a higher steady-state concentration in a evaluation of the scores in clinical practice can be
longer time as compared with healthy individuals. subjective. More recently, because of the greater
understanding of pharmacokinetics, probe drugs
Thus, excessive accumulation of the drug can occur
have been used as quantitative tests of liver func-
after several doses, increasing the risk of toxicity
tion and the degree of liver impairment, especially
and exaggerating the pharmacological effect if a
in relation to CYPs.[155] These drugs are probes for
normal dosage of the drug is used.
specific enzymes, whereas antipyrine, ICG, galac-
According to the available data, the kinetics of
tose and SBP are considered general metabolic
many drugs are altered enough in patients with
probes. In addition to the ethical problems and safety
liver disease to require dosage adjustment. This can
of probe drugs, the metabolism of probe drugs is
be accomplished by reducing the dosage but main-
affected by genetic and environmental factors (to-
taining a normal administration interval, or by bacco smoking, alcohol consumption, concomitant
lengthening the interval with a normal dose, or drugs and genetic polymorphism). These factors
changing both the dose and the interval. The prob- should be taken into consideration to avoid mis-
lem is to quantify these changes. leading results when a drug is used as a probe.
It is obvious that the degree of hepatic impairment In conclusion, at present there is no single sat-
should be evaluated to adjust the dosage, but the isfactory test which gives a quantitative measure of
problem is how. Unlike creatinine clearance for the liver function and impairment. In the clinical set-
kidney, at present there is no clinical laboratory test ting more than one single test is needed to evaluate
which gives a quantitative measure of liver function the severity of liver disease.
and the degree of its impairment. In addition, there Besides the use of clinical tests, some criteria
is no clear correlation between routine liver func- can provide a rational basis for determining dosage
tion tests and the capacity of the liver to metabolise adjustment. It is important to make a careful eval-
drugs. Usually, changes in pharmacokinetic param- uation of the clinical situation of the patient to as-
eters do not correlate well with the indicators of certain what type of pathophysiological changes
liver function impairment. Liver biopsy is commonly has caused liver disease (hepatic failure, reduced
used in patients with chronic hepatitis and cirrhosis liver blood flow or portosystemic shunting). A fun-
to evaluate the severity of disease; however, histo- damental factor is the type of drug administered
logical findings can be misleading, especially if ad- (drug with a low or high hepatic extraction, protein

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
Effects of Liver Disease on Pharmacokinetics 423

binding), because the effect of liver disease de- these patients is lower than normal. The mecha-
pends on the type of drug. Drugs with a low hepatic nism of this pharmacodynamic abnormality is not
extraction are not sensitive to liver blood flow clear.[66] Probably, in these patients the reabsorp-
changes but to variations of the activity of drug- tion of sodium in the proximal tubule is increased,
metabolising enzymes, whereas drugs with a high leaving a smaller amount of sodium to the loop of
hepatic extraction are sensitive to liver blood flow Henle, which is the site of action of torasemide.
changes. In any case, a close observation of the Thus, the saluretic effect of torasemide is dimin-
patient’s response to the drug is mandatory; there ished.
is an interindividual variability in the response. There is evidence that patients with cirrhosis are
Plasma drug concentrations, mainly unbound more sensitive to triazolam than healthy individu-
drug, should be monitored whenever it is possible als; they have an increased cerebral sensitivity to
and useful. An adequate knowledge is needed of this drug.[78] Psychometric tests of CNS performance
the kinetic changes caused by liver disease for the (flicker sensitivity, digit symbol substitution test
particular drug. The pharmacokinetic parameters and pursuit rotor) were given to patients with cir-
of the drugs discussed in this review are reported rhosis and healthy individuals before triazolam ad-
in table I for ease of reference. In general, I would ministration and again when plasma concentra-
advise caution in the administration of drugs me- tions of unbound drug were the same in the 2
tabolised by the liver to patients with hepatic im- groups (i.e. 2.25h after administration). Prior to ad-
pairment. I recommend that the drug should be ministration the performance in the tests was sim-
started at lower dosages and that the dosage should ilar in both groups and it was not influenced by
be adjusted to the patient’s clinical response, with cirrhosis per se. At 2.25h after administration the
the help of plasma drug concentration monitoring
patients with cirrhosis had a significant higher
when applicable.
average impairment in the performance of tests
(30% greater than the controls). This impairment
12.2 Altered Pharmacodynamics can be considered evidence of the brain’s hyper-
Finally, it is important to consider that liver dis- sensitivity to triazolam in patients with cirrhosis
ease not only affects the pharmacokinetics but also and is not caused by altered pharmacokinetics. The
the pharmacodynamics of drugs. Hepatic patients mechanism underlying this hypersensitivity re-
have a greater sensitivity to a number of drugs, mains to be explained.
especially drugs acting on the CNS, resulting in an CNS effects represent an important adverse ef-
altered response.[156] Therefore, in adjusting drug fect of the quinolones.[158] They include headache,
dosage in these patients both pharmacokinetic and dizziness, agitation, sleep disorders and, rarely,
pharmacodynamic changes should be taken into convulsions. These effects have been correlated
consideration. Some drugs reported in this review with the binding of quinolones to GABA receptors
have abnormal pharmacodynamics in hepatic dis- in the brain, thus inhibiting GABA and increasing
ease; only these will be examined in detail. the excitability of CNS. Cases of grand mal sei-
Patients with cirrhosis usually have a resistance zures have been reported in patients with severe
to loop diuretics, a diminished response because of cirrhosis who received pefloxacin at the usual dos-
pharmacodynamic alterations.[157] This has been age of 400mg twice daily.[159] The increased pene-
observed with torasemide. In healthy individuals tration of pefloxacin into the CNS because of the
the relationship between the urinary excretion of increased permeability of the blood-brain barrier
torasemide and the natriuretic response is charac- in patients with cirrhosis could favour the develop-
terised by a sigmoidal curve,[157] but in cirrhotic ment of grand mal seizures. When liver cirrhosis is
patients with ascites the curve is shifted downward severe it is probably better to avoid the use of
and to the right.[67] Thus, the diuretic response in pefloxacin. Increased penetration into the CNS in

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
Table I. Pharmacokinetic parameters of drugs in patients with hepatic disease. Values for healthy individuals are in parentheses. Values for clearance are all corrected for 70kg
424
bodyweight and are systemic clearances (CL) unless otherwise indicated. Values for the apparent volume of distribution are corrected for 70kg bodyweight
Drug Disease n fm (%) fu (%) F (%) CL (L/h/70kg) CLu (L/h/70kg) Vss (L/70kg) t1 ⁄2 β (h) References

Cardiovascular agents
Angiotensin converting enzyme inhibitors
Quinalapril The conversion of these prodrugs to the active form is reduced in patients with cirrhosis 32-35
Cilazapril
Enalapril
Angiotensin II receptor antagonists
Irbesartan C, both mild and moderate 10 (10) >99 10a 22
Losartan C >90 <10 ↓50% 37

© Adis International Limited. All rights reserved.


Calcium antagonists
Felodipine C 9 (23) >99 <1 178b (580) 392 (721) 16.3 (11) 41
Gallopamil C 6 93 7 60 109 11 44
Nicardipine C 9 (8) >99 1-2 13.7 (1.6) 45
Nifedipine C 7 (7) 99 2-4 90.5 (51.1) 13.9 (35.3) 7 (1.85) 46
Nimodipine C 6 (5) >99 2 217b (519) 48
Nisoldipine C 8 (8) >90 <1 14.7 (3.7) 29.6b (50.8) 16.6 (9.7) 49
Nitrendipine C 11 >90 2 51.16c 51
CH 5 (6) 50.4c (77.36c)
54 41.2d
48 (40) 44.1d (82.7d)
Ketanserin C 26 (10) 99 5 17.5 (33.9) 10.1 (14.3) 54
Antiarrhythmics
Flecainide C 6 (6) 75 60-70 16.0 (38.2) 49 (9.5) 56
Mexiletine C 6 (6) 85-92 30 9.66 (34.7) 28.7 (9.9) 59
Propafenone C 11 >90 4 Higher Lower 61
Hypolipidaemics
Fluvastatin C 11 (11) 98 1 ↓28%b ↓31% 63
Pravastatin C 53 50 Lower 65

Diuretics
Torasemide C + ascites 12 (12) 80 <1 96 2.27 (2.52) CLR 0.55 (0.38) 24 (11.7) 8.1 (3.6) 67

Psychoactive and anticonvulsant agents


Benzodiazepines
Alprazolam C 17 (17) >90 17-21 2.35b (5.12) 19.7 (11.4) 70
Rodighiero

Clin Pharmacokinet 1999 Nov; 37 (5)


Midazolam C - moderate 10 (9) >90 5 24.1 (38.2) 2.8 (2.25) 74
C - severe 7 (8) 22.7 (54.1) 3.09 (1.6) 75
C - severe 7 (7) 76 (38) 14.0 (23.6) 7.36 (3.80) 76
Triazolam C 8 (7) >90 11 21.0b (28.1) 77
C - severe 8 (18) CL/Fu 62.2 (100.4) 78
Flumazenil C 8 (8) >99 50-60 65 (27) 42 (72) 64 (68) 81
Antidepressants
Fluvoxamine C 13 96 33 54.6b 24.7 84
Fluoxetine C 13 (12) 95-97 5 17.6b (40.3) 6.6 (2.2) days 86
Norfluoxetine C 13 (12) 5.9 (8.4) 12 (6.4) days 86
Paroxetine C 12 (6) 98-99 5 83 (36) 89

© Adis International Limited. All rights reserved.


Nefazodone C 12 >90 <1 Decreased 91
C 12 Decreased 92
Effects of Liver disease on Pharmacokinetics

Moclobemide C 12 (14) >99 50 84 (56) 14.6 (37.7) 75.7 (84.1) 94


Anticonvulsants
Tiagabine C - mild 7 97 4 11.7 96
C - moderate 6 15.9
(6) (6.5)

Antiemetics
Metoclopramide C - severe 8 (8) 80 60 84 (79) 11.2 (23.8) 3.1 (3.4) 15.4 (7.2) 98
C - severe 18 (12) 82 (60) 99
Ondansetron CLD - mild 19 >95 24-30 12.6 224 9.1 101
CLD - moderate 17.6 230 9.2
CLD - severe 5.38 179 20.6
(6) (28.6) (153) (3.6)
CLD - mild 12 80 1411b 10 102
CLD - moderate 78 1495b 13
CLD - severe 98 987b 20
(12) (60) (3121)b (6)
Tropisetron C - mild to moderate 10 (10)a 52-66 29-41 104

Antiulcers
Proton pump inhibitors
Lansoprazole C - compensated 8 >95 <5 2.77b 24.5 6.1 108
C - uncompensated 8 0.67b 38.5 7.2
(18) (18.2) 31.0 1.4
Continued over page
425

Clin Pharmacokinet 1999 Nov; 37 (5)


Table I. Contd
426
Drug Disease n fm (%) fu (%) F (%) CL (L/h/70kg) CLu (L/h/70kg) Vss (L/70kg) t1 ⁄2 β (h) References

Pantoprazole C 12 >95 <5 Lower Higher 110


Omeprazole C - mild 3 >95 <5 2.96 112
C - moderate 4 0.72
C - severe 1 98 4.0
(18) (56) (35.6)
C 8 (8) 5.84b (47.9b) 2.85 (0.73) 114

Antimicrobials and antiretrovirals


Grepafloxacin HI - mild 10 90-94 50 23.1b 13 121
HI - moderate 10 14.9b 19

© Adis International Limited. All rights reserved.


(12) (35.8b) (12)
Ornidazole C - severe 10 (10) >90 87-92 2.06 (3.02) 21.9 (14.1) 123
Pefloxacin C 16 (12) 85-90 70-80 186 (571) 35.1 (11.0) 126
Stavudine C 5 60a 130
Zidovudine CH 8 (6) 80-86 75 1.55b (2.27b) 2.04 (1.0) 133

Immunosuppressants
Cyclosporin HI >90 66 36.8 (55.0) 138
Pre-transplantation 14.7 140
Post-transplantation 25.9
Tacrolimus HI - moderate to severe 5 >99 23-28 36 38.5 142

Miscellaneous drugs
Tolcapone C 8 >80 12-13 70 7.27 3826 1.2 150
NCLD 8 (8) 72 7.48 5527 1.2
(8) (68) (8.27) (6871) (1.1)
Toremifene ALD 10 (10) High <1 3.70 (5.08) 10.9 (6.2) 149

a No significant differences compared to healthy individuals.


b Apparent clearance (CL/F).
c Single intravenous dose.
d Oral for 7 days.
ALD = alcoholic liver disease; C = cirrhosis; CH = chronic hepatitis; CLD = chronic liver disease; CLR = renal clearance; CLu = clearance of unbound drug; F = oral bioavailability;
fm = fraction metabolised; fu = fraction of unbound drug in plasma; HI = hepatic impairment; IV = intravenous; n = number of patients and (healthy individuals); NCLD = noncirrhotic
liver disease; t1⁄2β = elimination half-life; Vss = volume of distribution at steady state; ↓ = decrease.
Rodighiero

Clin Pharmacokinet 1999 Nov; 37 (5)


Effects of Liver Disease on Pharmacokinetics 427

hepatic disease has also been reported for cimeti- • Repeated administration of the drug under in-
dine and ranitidine, again with the occurrence of vestigation should be studied, as this would be
mental disturbances.[117,119] more clinically relevant than the use of a single
dose.
13. Conclusions • The clearance of the unbound drug rather than
total drug clearance should be measured, be-
The pharmacokinetics of many types of drugs cause unbound drug concentrations may be in-
metabolised by the liver are modified in patients creased by liver impairment.
with hepatic disease. Liver disease can affect the • When the drug being studied is used to treat a
pharmacokinetics by: (i) reducing the activity of concomitant disease (e.g. depression) then the
drug metabolising enzymes; (ii) reducing the syn- kinetics of the antidepressant agent should be
thesis of plasma proteins; or (iii) changing liver assessed in patients with liver disease and de-
blood flow. The pharmacokinetic modifications pression using a control group of patients with
depend on the characteristics of the drug (low or depression. This should be done as the concur-
high hepatic extraction, protein binding) and the rent disease may cause pathophysiological
severity of liver disease. The problem is to quantify changes in the body, which can affect the phar-
the degree of severity because, at present, there is macokinetics.
no single satisfactory test which gives a quantita-
tive measure of liver function and degree of im- References
pairment. A promising development is the study of 1. Rodighiero V. Effects of liver disease on pharmacokinetics. Ital
J Gastroenterol 1987; 19: 149-58
CYP isoforms, which is a rapidly expanding area 2. Bass NM, Williams RL. Guide to drug dosage in hepatic dis-
in pharmacology and is concerned with the identi- ease. Clin Pharmacokinet 1988; 15: 396-420
fication and characterisation of individual CYP 3. McLean AJ, Morgan DJ. Clinical pharmacokinetics in patients
with liver disease. Clin Pharmacokinet 1991; 21: 42-69
isoforms. Several drugs metabolised by specific 4. Morgan DJ, McLean AJ. Clinical pharmacokinetic and pharma-
CYP isoforms have been used as probes for quan- codynamic considerations in patients with liver disease: an
update. Clin Pharmacokinet 1995; 29 (5): 370-91
titative tests of liver function and degree of hepatic 5. Paintaud G, Bechtel Y, Brientini MP, et al. Effects of liver dis-
impairment. eases on drug metabolism. Therapie 1996; 51 (4): 384-9
6. Shargel L, Yu AB. Applied biopharmaceutics and pharmacoki-
Because the kinetics of many drugs are altered netics. Stamford (CT): Appleton and Lange, 1999: 353-98
by liver disease to an extent that requires dosage 7. Bertz RJ, Granneman GR. Use of in vitro and in vivo data to
adjustment, guidelines providing a rational basis estimate the likelihood of metabolic pharmacokinetic inter-
actions. Clin Pharmacokinet 1997; 32 (3): 210-58
for the required changes have been suggested. 8. Murray M. P450 enzymes: inhibition mechanisms, genetic reg-
However, it should be noted that liver disease af- ulation and effects of liver disease. Clin Pharmacokinet 1992;
23 (2): 132-46
fects not only pharmacokinetics but also pharma- 9. Gu L, Gonzalez FJ, Kalow W, et al. Biotransformation of caf-
codynamics. feine, paraxanthine, theobromine and theophylline by cDNA-
In order to improve our knowledge of the kinet- expressed human CYP1A2 and CYP2E1. Pharmacogenetics
1992; 2 (2): 73-7
ics of drugs in patients with liver disease, the follow- 10. Holstege A, Staiger M, Haag K, et al. Correlation of caffeine
ing aspects should be considered when designing elimination and Child’s classification in liver cirrhosis. Klin
Wochenschr 1989; 4 67 (1): 6-15
studies in this patient population. 11. Child CG. The liver and portal hypertension. Philadelphia (PA):
• The degree of liver impairment should be spec- WB Saunders, 1964
12. Pugh RN, Murray Lyon IM, Dawson JL, et al. Transection of
ified in the study. the oesophagus for bleeding oesophageal varices. Br J Surg
• Patients with heterogeneous liver diseases 1973; 60 (8): 646-9
should not be grouped together. However, pa- 13. Scott NR, Stambuk D, Chakraborty J, et al. The pharmacoki-
netics of caffeine and its dimethylxanthine metabolites in pa-
tients with varying degrees of hepatic impair- tients with chronic liver disease. Br J Clin Pharmacol 1989;
ment should be included as separate groups. 27 (2): 205-13
14. Lewis FW, Rector Jr WG. Caffeine clearance in cirrhosis: the
• An adequate number of patients should be in- value of simplified determinations of liver metabolic capac-
cluded to enable any differences to be identified. ity. J Hepatol 1992; 14 (2-3): 157-62

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
428 Rodighiero

15. Braillon A, Cales P, Valla D, et al. Influence of the degree of 33. Gross V, Schölmerich J, Treher E, et al. Altered kinetics of
liver failure on systemic and splanchnic haemodynamics and cilazapril (Ro 31-2848) and cilazaprilat (Ro 31-3113) in pa-
on response to propranolol in patients with cirrhosis. Gut tients with liver cirrhosis. Hepatology 1988; 8: 1382
1986; 27 (10): 1204-9 34. Hayes PC, Plevis JN, Bouchier IAD. Pharmacokinetics of en-
16. Pelkonen O, Raunio H, Rautio A, et al. Coumarin 7-hydroxyl- alapril and lisinopril in subjects with normal and impaired
ase: characteristics and regulation in mouse and man. J Ir Coll hepatic function. J Hum Hypertens 1989; 3: 153-8
Phys Surg 1993; 22 Suppl. 1: 24-8 35. Ohnishi A, Tsuboi Y, Ishizaki T. Kinetics and dynamics of en-
17. Sotaniemi EA, Rautio A, Backstrom M, et al. CYP3A4 and alapril in patients with liver cirrhosis. Clin Pharmacol Ther
CYP2A6 activities marked by the metabolism of lignocaine 1989; 45: 657-65
and coumarin in patients with liver and kidney diseases and 36. Lo MW, Golberg MR, McCrea JB, et al. Pharmacokinetics of
epileptic patients. Br J Clin Pharmacol 1995; 39 (1): 71-6 losartan, an angiotensin II receptor antagonist, and its active
18. Kraul H, Truckenbrodt J, Huster A, et al. A comparison of in metabolite EXP3174 in humans. Clin Pharmacol Ther 1995;
vitro and in vivo biotransformation in patients with liver dis- 58: 641-9
ease of differing severity. Eur J Clin Pharmacol 1991; 41 (5): 37. Goa KL, Wagstaff AJ. Losartan potassium. A review of its phar-
macology, clinical efficacy and tolerability in the manage-
475-80
ment of hypertension. Drugs 1996; 51: 820-45
19. Gillis JC, Markham A. Irbesartan: a review of its pharmacody-
38. Flesch G, Lloyd MP. Absolute bioavailability and pharmacoki-
namic and pharmacokinetic properties and therapeutic use in the
netics of valsartan, an angiotensin II receptor antagonist, in
management of hypertension. Drugs 1997; 54 (6): 885-902 man. Eur J Clin Pharmacol 1997; 52: 115-20
20. Relling MV, Aoyama T, Gonzalez FJ, et al. Tolbutamide and 39. Brookman LJ, Rolan PE, Benjamin IS, et al. Pharmacokinetics
mephenytoin hydroxylation by human cytochrome P450s in of valsartan in patients with liver disease. Clin Pharmacol
the CYP2C subfamily. J Pharmacol Exp Ther 1990; 252 (1): Ther 1997; 62: 272-8
442-7 40. Kelly JC, O’Malley K. Clinical pharmacokinetics of calcium
21. Adedoyin A, Arns PA, Richards WO, et al. Selective effect of antagonists: an update. Clin Pharmacokinet 1992; 22: 416-33
liver disease on the activities of specific metabolizing en- 41. Regärdh CG, Edgar B, Olsson R, et al. Pharmacokinetics of
zymes: investigation of cytochromes P450 2C19 and 2D6. felodipine in patients with liver disease. Eur J Clin Pharmacol
Clin Pharmacol Ther 1998; 64 (1): 8-17 1989; 36: 473-9
22. Marino MR, Langenbacher KM, Raymond RH, et al. Pharma- 42. Wang SX, Sutfin T, Baarnhielm C, et al. Contribution of the
cokinetics and pharmacodynamics of irbesartan in patients intestine to the first pass metabolism of felodipine in the rat.
with hepatic cirrhosis. J Clin Pharmacol 1998; 38(4): 347-56 J Pharmacol Exp Ther 1989; 250: 632-6
23. Howden CW, Birnie GG, Brodie MJ. Drug metabolism in liver 43. Brodgen RN, Benfield P. Gallopamil: a review of its pharmaco-
disease. Pharmacol Ther 1989; 40 (3): 439-74 dynamic and pharmacokinetic properties, and therapeutic po-
24. Arns PA, Adedoyin A, DiBisceglie AM, et al. Mephenytoin dis- tential in ischaemic heart disease. Drugs 1994; 47: 93-115
position and serum bile acids as indices of hepatic function in 44. Kaim AAH, Farker K. Pharmacokinetics and pharmacodynamics
chronic viral hepatitis. Clin Pharmacol Ther 1997; 62 (5): of gallopamil in patients with liver cirrhosis [abstract 36].
527-37 Naunyn Schmiedebergs Arch Pharmacol 1992; 345 Suppl. R9
25. Frye RF, Matzke GR, Adedoyin A, et al. Validation of the five- 45. Razak TA, McNeil JJ, Sewell RB, et al. The effect of hepatic
drug ‘Pittsburgh cocktail’ approach for assessment of selective cirrhosis on the pharmacokinetics and blood pressure re-
regulation of drug-metabolizing enzymes. Clin Pharmacol sponse to nicardipine. Clin Pharmacol Ther 1990; 47: 463-9
Ther 1997; 62 (4): 365-76 46. Kleinbloesem CH, van Harten J, Wilson JPH, et al. Nifedipine:
26. Wilkinson GR. Cytochrome P4503A (CYP3A) metabolism: kinetic and hemodynamic effects in patients with liver cirrho-
prediction of in vivo activity in humans. J Pharmacokinet sis after intravenous and oral administration. Clin Pharmacol
Biopharm 1996; 24 (5): 475-90 Ther 1986; 40: 21-8
27. Huang YS, Lee SD, Deng JF, et al. Measuring lidocaine meta- 47. Ene MD, Roberts CJC. Pharmacokinetics of nifedipine after
bolite monoethylglycinexylidide as a quantitative index of oral administration in chronic liver disease. J Clin Pharmacol
1987; 27: 1001-4
hepatic function in adults with chronic hepatitis and cirrhosis.
48. Gengo FM, Fagan SC, Krol G, et al. Nimodipine disposition
J Hepatol 1993; 19 (1): 140-7
and haemodynamic effects in patients with cirrhosis and age-
28. Shiffman ML, Luketic VA, Sanyal AJ, et al. Hepatic lidocaine
matched controls. Br J Clin Pharmacol 1987; 23: 47-53
metabolism and liver histology in patients with chronic hep-
49. van Harten J, van Brummelen P, Wilson JHP, et al. Nisoldipine:
atitis and cirrhosis. Hepatology 1994; 19 (4): 933-40 kinetics and effects on blood pressure and heart rate in patients
29. George J, Murray M, Byth K, et. al. Differential alterations of with liver cirrhosis after intravenous and oral administration.
cytochrome P450 proteins in livers from patients with severe Eur J Clin Pharmacol 1988; 34: 387-94
chronic liver disease. Hepatology 1995; 21 (1): 120-8 50. Davidsson GK, Edwards JS, Davidson C. The effect of age and
30. George J, Liddle C, Murray M, et al. Pre-translational regulation liver disease on the pharmacokinetics of the calcium antago-
of cytochrome P450 genes is responsible for disease-specific nist, nisoldipine. Curr Med Res Opin 1995; 13: 285-97
changes of individual P450 enzymes among patients with cir- 51. Dylevicz P, Kirch W, Santos SR, et al. Bioavailability and elim-
rhosis. Biochem Pharmacol 1995; 30; 49 (7): 873-81 ination of nitrendipine in liver disease. Eur J Clin Pharmacol
31. Burnier M, Biollaz J. Pharmacokinetic optimisation of angio- 1987; 32: 563-8
tensin converting enzyme (ACE) inhibitor therapy. Clin Phar- 52. Lasseter KC, Shamblen EC, Murdoch AA, et al. Steady-state
macokinet 1992; 22: 375-84 pharmacokinetics of nitrendipine in hepatic insufficiency. J
32. Horvath AM, Olson SC, Ferry JJ, et al. Pharmacokinetics of Cardiovasc Pharmacol 1984; 6 Suppl. 7: S977-81
quinalapril and its active metabolite, quinalaprilat, in patients 53. Persson B, Heykants J, Hedner T. Clinical pharmacokinetics of
with hepatic impairment. J Clin Pharmacol 1988; 28: 917 ketanserin. Clin Pharmacokinet 1991; 20: 263-79

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
Effects of Liver Disease on Pharmacokinetics 429

54. Lebrec D, Hadengue A, Gaudin C, et al. Pharmacokinetics of 75. MacGilchrist AJ, Birnie GG, Cook A, et al. Pharmacokinetics
ketanserin in patients with cirrhosis. Clin Pharmacokinet and pharmacodynamics of intravenous midazolam in patients
1990; 19: 160-6 with severe alcoholic cirrhosis. Gut 1986; 27: 190-5
55. Nestico PF, Morganroth J, Horowitz LN. New antiarrhythmic 76. Pentikäinen PJ, Välisalm L, Himberg JJ, et al. Pharmacokinet-
drugs. Drugs 1988; 35 (3): 286-319 ics of midazolam following intravenous and oral administra-
56. McQuinn RL, Pentikäinen PJ, Chang SF, et al. Pharmacokinet- tion in patients with chronic liver disease and in healthy
ics of flecainide in patients with cirrhosis of the liver. Clin subjects. J Clin Pharmacol 1989; 29: 272-7
Pharmacol Ther 1988; 44: 566-72 77. Kroboth PD, Smith RB, Van Thiel DH, et al. Nighttime dosing
57. Forland SC, Burgess E, Blair AD, et al. Oral flecainide phar- of triazolam in patients with liver disease and normal sub-
macokinetics in patients with impaired renal function. J Clin jects: kinetics and daytime effects. J Clin Pharmacol 1987;
Pharmacol 1988; 28 (3): 259-67 27: 555-60
58. Monk JP, Brogden RN. Mexiletine: a review of its pharmaco- 78. Bakti G, Fisch HU, Karlaganis G, et al. Mechanism of the ex-
dynamic and pharmacokinetic properties, and therapeutic use cessive sedative response of cirrhotics to benzodiazepines:
in the treatment of arrhythmias. Drugs 1990; 40: 374-411 model experiments with triazolam. Hepatology 1987; 7: 629-38
59. Pentikäinen PJ, Hietakorpi S, Halinen MO, et al. Cirrhosis of 79. Hoyumpa AM, Schenker S. Is glucuronidation truly preserved in
the liver markedly impairs the elimination of mexiletine. Eur patients with liver disease? Hepatology 1991; 13 (4): 786-95
J Clin Pharmacol 1986; 30: 83-8 80. Brodgen RN, Goa KL, Flumazenil: a reappraisal of its pharma-
60. Bryson HM, Palmer KJ, Langtry HD, et al. Propafenone: a cological properties and therapeutic efficacy as a benzo-
reappraisal of its pharmacology, pharmacokinetics and ther- diazepine antagonist. Drugs 1991; 42: 1061-89
apeutic use in cardiac arrhythmias. Drugs 1993; 45: 85-130 81. Janssen U, Walker S, Maier K, et al. Flumazenil disposition and
61. Lee JT, Yee YG, Dorian P, et al. Influence of hepatic dysfunction elimination in cirrhosis. Clin Pharmacol Ther 1989; 46: 317-23
on the pharmacokinetics of propafenone. J Clin Pharmacol 82. van Harten J. Clinical pharmacokinetics of selective serotonin
1987; 27; 384-9 reuptake inhibitors. Clin Pharmacokinet 1993; 24: 203-20
62. Plosker GL, Wagstaff A. Fluvastatin: a review of its pharmacol- 83. van Harten J. Overview of the pharmacokinetics of fluvoxam-
ogy and use in the management of hypercholesterolaemia. ine. Clin Pharmacokinet 1995; 29 Suppl. 1: 1-9
Drugs 1996; 51: 433-59 84. van Harten J, Duchier J, Devissaguet JP, et al. Pharmacokinetics
63. Deslypere JP. Clinical implications of biopharmaceutical prop- of fluvoxamine maleate in patients with liver cirrhosis after
erties of fluvastatin. Am J Cardiol 1994; 73: 12-7D single-dose oral administration. Clin Pharmacokinet 1993;
24: 177-82
64. Haria M, McTavish D. Pravastatin: a reappraisal of its pharma-
cological properties and clinical effectiveness in the manage- 85. Altamura AC, Moro AR, Percudani M. Clinical pharmacokinet-
ics of fluoxetine. Clin Pharmacokinet 1994; 26: 201-14
ment of coronary heart disease. Drugs 1997; 53: 299-336
65. Pan HY. Clinical pharmacology of pravastatin, a selective in- 86. Schenker S, Bergstrom RF, Wolen RL, et al. Fluoxetine dispo-
sition and elimination in cirrhosis. Clin Pharmacol Ther 1988;
hibitor of HMG-CoA reductase. Eur J Clin Pharmacol 1991;
44: 353-9
40 Suppl. 1: S15-8
87. Gunasekara NS, Noble S, Benfield P. Paroxetine: an update of
66. Knauf H, Mutschler E. Clinical pharmacokinetics and pharmaco-
its pharmacology and therapeutic use in depression and a re-
dynamics of torasemide. Clin Pharmacokinet 1998; 34: 1-24
view of its use in other disorders. Drugs 1998; 55: 85-120
67. Schwartz S, Brater DC, Pound D, et al. Bioavailability, phar-
88. Krastev Z, Terziivanov D, Vlahov V, et al. The pharmacokinet-
macokinetics, and pharmacodynamics of torasemide in pa-
ics of paroxetine in patients with liver cirrhosis. Acta Psy-
tients with cirrhosis. Clin Pharmacol Ther 1993; 54: 90-7 chiatr Scand 1989; 80 Suppl. 350: 91-2
68. Brunner G, von Bergmann K, von Möllendorff E. Comparison
89. Dahoff K, Almdal TP, Bjerrum K, et al. Pharmacokinetics of
of diuretic effects and pharmacokinetics of torasemide and paroxetine im patients with cirrhosis. Eur J Clin Pharmacol
furosemide after a single oral dose in patients with hydropic- 1991; 41: 351-4
ally decompensated cirrhosis of the liver. Arzneimittel 90. Davis R, Whittington R, Bryson HM. Nefazodone: a review of
Forschung Drug Res 1988; 38: 176-9 its pharmacology and clinical efficacy in the management of
69. Greenblatt DJ, Wright CE. Clinical pharmacokinetics of al- major depression. Drugs 1997; 53: 608-36
prazolam: therapeutic implications. Clin Pharmacokinet 91. Barbhaiya RH, Shukla UA, Natarajan XS, et al. Single- and
1993; 24: 453-71 multiple-dose pharmacokinetics of nefazodone in patients
70. Juhl RP, Van Thiel DH, Dittert LW, et al. Alprazolam pharma- with hepatic cirrhosis. Clin Pharmacol Ther 1995; 58: 390-8
cokinetics in alcoholic liver disease. J Clin Pharmacol 1984; 92. Barbhaiya RH, Shukla UA, Greene DS. Single-dose pharmaco-
24: 113-9 kinetics of nefazodone in healthy young and elderly subjects
71. Garzone PD, Krobot PD. Pharmacokinetics of the newer and in subjects with renal or hepatic impairment. Eur J Clin
benzodiazepines. Clin Pharmacokinet 1989; 16: 337-64 Pharmacol 1995; 49: 221-8
72. Thummel KE, O’Shea D, Paine MF, et al. Oral first-pass elim- 93. Fitton A, Faulds D, Goa KL. Moclobemide: a review of its
ination of midazolam involves both gastrointestinal and he- pharmacological properties and therapeutic use in depressive
patic CYP3A-mediated metabolism. Clin Pharmacol Ther illness. Drugs 1992; 43: 561-96
1996; 59: 491-502 94. Stoeckel K, Pfefen K, Mayersohn M, et al. Absorption and dis-
73. Paine MF, Shen DD, Kunze KL, et al. First-pass metabolism of position of moclobemide in patients with advanced age or
midazolam by the human intestine. Clin Pharmacol Ther reduced liver or kidney function. Acta Psychiatr Scand 1990;
1996; 60: 14-24 Suppl. 360: 94-7
74. Trouvin JH, Farinotti R, Haberer JP, et al. Pharmacokinetics of 95. Adkins JC, Noble S. Tiagabine: a review of its pharmacody-
midazolam in anaesthetized cirrhotic patients. Br J Anaesth namic and pharmacokinetic properties and therapeutic poten-
1988; 60: 762-7 tial in the management of epilepsy. Drugs 1998; 55: 437-60

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
430 Rodighiero

96. Lau AH, Gustavson LE, Sperelakis R, et al. Pharmacokinetics 118. Schentag JJ, Cerra FB, Calleri GM, et al. Age, disease, and
and safety of tiagabine in subjects with various degrees of cimetidine disposition in healthy subjects and chronically ill
hepatic function. Epilepsia 1997; 38: 448-51 patients. Clin Pharmacol Ther 1981 Jun; 29 (6): 737-43
97. Lauritsen K, Laursen LS, Rask-Madsen J. Clinical pharmaco- 119. Vial T, Goubier C, Bergeret A, et al. Side effects of ranitidine.
kinetics of drugs used in the treatment of gastrointestinal dis- Drug Saf 1991; 6 (2): 94-117
eases. Clin Pharmacokinet 1990; 19: 11-31 120. Efthymiopoulos C, Bramer SL, Maroli A. Pharmacokinetics of
98. Magueur E, Hagege H, Attali P, et al. Pharmacokinetics of meto- grepafloxacin after oral administration of single and repeated
clopramide in patients with liver cirrhosis. Br J Clin Phar- doses in healthy young males. Clin Pharmacokinet 1997; 33
macol 1991; 31: 185-7 Suppl. 1: 1-8
99. Hellstern A, Hellenbrecht D, Saller R, et al. Absolute bioavail- 121. Efthymiopoulos C, Bramer SL, Maroli A, et al. Grepafloxacin
ability of metoclopramide given orally or by enema in patients pharmacokinetics in individuals with hepatic dysfunction.
with normal liver function or with cirrhosis of the liver. Clin Pharmacokinet 1997; 33 Suppl. 1: 25-31
Arzneimittel Forschung Drug Res 1987; 37: 733-6 122. Lau AH, Lam NP, Piscitelli SC, et al. Clinical pharmacokinetics
100. Roila F, Del Favero A. Ondansetron clinical pharmacokinetics. of metronidazole and other nitroimidazole anti-infectives.
Clin Pharmacokinet 1995; 29: 95-109 Clin Pharmacokinet 1992; 23: 328-64
101. Blake JC, Palmer JL, Minton NA, et al. The pharmacokinetic 123. Taburet AM, Delion F, Attali P, et al. Pharmacokinetics of or-
of intravenous ondansetron in patients with hepatic impair- nidazole in patients with severe liver cirrhosis. Clin Phar-
ment. Br J Clin Pharmacol 1993; 35: 441-3 macol Ther 1986; 40: 359-64
102. Figg WD, Dukes GE, Pritchard JF, et al. Ondansetron (OND) 124. Taburet AM, Attali P, Bourget P, et al. Pharmacokinetics of or-
pharmacokinetics in chronic liver disease. Clin Pharmacol nidazole in patients with acute viral hepatitis, alcoholic cir-
Ther 1992; 51: 171 rhosis, and extrahepatic cholestasis. Clin Pharmacol Ther
103. Rhoda Lee C, Plosker GL, McTavish D. Tropisetron: a review 1989; 45: 373-9
of its pharmacodynamic and pharmacokinetic properties, and 125. Bressolle F, Gonçalves F, Gouby A, et al. Pefloxacin clinical
therapeutic potential as an antiemetic. Drugs 1993; 46: 925-43 pharmacokinetics. Clin Pharmacokinet 1994; 27: 418-46
104. de Bruijn KM. Tropisetron: a review of the clinical experience. 126. Danan G, Montay G, Cunci R, et al. Pefloxacin kinetics in cir-
Drugs 1992; 43: 11-22 rhosis. Clin Pharmacol Ther 1985; 38: 439-42
105. Andersson T, Cederberg C, Regärdh CG, et al. Pharmacokinet- 127. Galtier M, Bressolle F, de la Coussaye JE, et al. Multiple dose
ics of various single intravenous and oral doses of om- pharmacokinetics of pefloxacin in patients with hepatocellu-
eprazole. Eur J Clin Pharmacol 1990; 39: 195-7 lar deficiency. Clin Pharmacokinet 1993; 25: 415-23
106. Delhotal-Landes B, Petite JP, Flouvat B. Clinical pharmacoki- 128. Cardey J, Silvain C, Bouquet S, et al. Oral pharmacokinetics
netics of lansoprazole. Clin Pharmacokinet 1995; 28: 458-70 and ascitic fluid penetration of pefloxacin in cirrhosis. Eur J
107. Fitton A, Wiseman L. Pantoprazole: a review of its pharmaco- Clin Pharmacol 1987; 33: 469-72
logical properties and therapeutic use in acid-related disor- 129. Rana KZ, Dudley MN. Clinical pharmacokinetics of stavudine.
ders. Drugs 1996; 51: 460-8 Clin Pharmacokinet 1997; 33 (4): 276-84
108. Delhotal-Landes B, Flouvat B, Duchier J, et al. Pharmacokinet- 130. Schaad HJ, Petty BG, Grasela DM, et al. Pharmacokinetics and
ics of lansoprazole in patients with renal or liver disease of safety of a single dose of stavudine (d4T) in patients with
varying severity. Eur J Clin Pharmacol 1993; 45: 367-71 severe hepatic impairment. Antimicrob Agents Chemother
109. Coste T, Logeais C, Delhotal-Landes B, et al. Pharmacokinetics 1997; 41 (12): 2793-6
of lansoprazole after repeated administration in cirrhosis pa- 131. Grasela D, Christofalo B, Raymond E, et al. Safety and phar-
tients [abstract]. Gastroenterology 1993; 104 Suppl.: A59 macokinetics (PK) of stavudine (d4T) in subjects with mild,
110. Brunner G, Chang J, Hartmann M, et al. Pharmacokinetik von moderate or severe renal impairment [abstract]. 2nd National
pantoprazol bei patienten mit leberzirrhose. Med Klin 1994; Conference on Human Retroviruses and Related Infections;
8 Suppl. 1: 189 1995 Jan 29-Feb 2; Washington, DC. 145
111. Benet LZ, Zech K. Pharmacokinetics: a relevant factor for the 132. Acosta EP, Page LM, Fletcher CV. Clinical pharmacokinetics
choice of a drug? Aliment Pharmacol Ther 1994; 8 Suppl. 1: of zidovudine: an update. Clin Pharmacokinet 1996; 30:
25-32 251-62
112. Andersson T, Olsson R, Regärdh CG, et al. Pharmacokinetics 133. Moore KHP, Raasch RH, Bronwer KLR, et al. Pharmacokinet-
of [14C]omeprazole in patients with liver cirrhosis. Clin Phar- ics and bioavailability of zidovudine and its glucuronidated
macokinet 1993; 24: 71-8 metabolite in patients with human immunodeficiency virus
113. Landahl S, Andersson T, Larsson HE, et al. Pharmacokinetic infection and hepatic disease (AIDS Clinical Trials group pro-
study of omeprazole in elderly healthy volunteers. Clin Phar- tocol 062). Antimicrob Agents Chemother 1995; 39: 2732-7
macokinet 1992; 23: 469-76 134. Taburet AM, Naveau S, Zorza G, et al. Pharmacokinetics of
114. Rinetti M, Regazzi MB, Villani P, et al. Pharmacokinetics of zidovudine in patients with liver cirrhosis. Clin Pharmacol
omeprazole in cirrhotic patients. Arzneimittel Forschung Ther 1990; 47: 731-9
Drug Res 1991; 41: 420-2 135. Bareggi SR, Cinque P, Mazzei M, et al. Pharmacokinetics of
115. McKee RF, MacGilchrist AJ, Garden OJ, et al. The antisecre- zidovudine in HIV-positive patients with liver disease. J Clin
tory effect and pharmacokinetics of omeprazole in chronic Pharmacol 1994; 34: 782-6
liver disease. Aliment Pharmacol Ther 1988; 2: 429-37 136. Faulds D, Goa KL, Benfield P. Cyclosporin: a review of its
116. Caulin C, Gouéron H, Bretagne JF, et al. Tolerance de pharmacodynamic and pharmacokinetic properties, and ther-
l’omeprazole chez l’insuffisant hepatique. Etude ouverte chez apeutic use in immunoregulatory disorders. Drugs 1993; 45:
24 cirrhotiques. Gastroenterol Biol Clin 1987; 11: 42A 953-1040
117. Schentag JJ, Cerra FB, Calleri G, et al. Pharmacokinetic and 137. Noble S, Markham A. Cyclosporin: a review of the pharmaco-
clinical studies in patients with cimetidine-associated mental kinetic properties, clinical efficacy and tolerability of a
confusion. Lancet 1979 Jan 27; I (8109): 177-81 microemulsion-based formulation. Drugs 1995; 50: 924-41

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)
Effects of Liver Disease on Pharmacokinetics 431

138. Kahan BD, Kramer WG, Wideman C, et al. Demographic fac- liver and kidney function. Clin Pharmacol Ther 1995; 57:
tors affecting the pharmacokinetics of cyclosporine estimated 628-35
by radioimmunoassay. Transplantation 1986; 41: 459-64 150. Jorga KM, Kroodsma JM, Fotteler B, et al. Effect of liver im-
139. Venkataramanan R, Gray J, Ptachcinski RJ, et al. Cyclosporine pairment on the pharmacokinetics of tolcapone and its meta-
kinetics in liver disease. Clin Pharmacol Ther 1985; 37: 234 bolites. Clin Pharmacol Ther 1998; 63 (6): 646-54
140. Hebert MF, Walcher VJ, Benet LZ. Cyclosporine pharmacoki- 151. Demotes Mainard F, Vincon G, Amouretti M, et al. Pharmaco-
netics pre- and post-liver transplantation [abstract]. Clin kinetics and protein binding of cefpiramide in patients with
Pharmacol Ther 1995; 57: 187 alcoholic cirrhosis. Clin Pharmacol Ther 1991; 49 (3): 263-9
141. Venkataramanan R, Swaminathan A, Prasad T, et al. Clinical 152. Chopra S, Griffin PH. Laboratory tests and diagnostic proce-
pharmacokinetics of tacrolimus. Clin Pharmacokinet 1995; dures in evaluation of liver disease. Am J Med 1985; 79 (2):
29: 404-30 221-30
142. Jain AB, Venkataramanan R, Cadoff E, et al. Effect of hepatic 153. Barbare JC, Poupon RE, Jaillon P, et al. Intrinsic hepatic clear-
dysfunction and T tube clamping on FK506 pharmacokinetics ance and Child-Turcotte classification for assessment of liver
and trough concentrations. Transplant Proc 1990; 22 Suppl. function in cirrhosis. J Hepatol 1985; 1 (3): 253-9
1: 57-9 154. Conn HO. A peek at the Child-Turcotte classification. Hepato-
143. Jain AB, Abu-Elmagd K, Abdallah H, et al. Pharmacokinetics logy 1981; 1: 673-6
of FK506 in liver transplant recipients after continuous intra- 155. Tanaka E. Clinical importance of non-genetic and genetic cy-
venous infusion. J Clin Pharmacol 1993; 33: 606-11 tochrome P450 function tests in liver disease. J Clin Pharm
144. Winkler M, Ringe B, Rodeck B, et al. The use of plasma levels Ther 1998; 23 (3): 161-70
for FK506 dosing in liver-grafted patients. Transpl Int 1994; 156. Westphal JF, Brogard JM. Drug administration in chronic liver
7: 329-33 disease. Drug Saf 1997; 17 (1): 47-73
145. Abu-Elmagd K, Fung JJ, Alessiani M, et al. The effect of graft 157. Brater DC. Clinical pharmacology of loop diuretics in health
function on FK506 plasma levels, dosages, and renal func- and disease. Eur Heart J 1992; 13 Suppl. G: 10-4
tion, with particular reference to the liver. Transplantation 158. Domagala JM. Structure-activity and structure-side-effect rela-
1991; 52: 71-7 tionships for the quinolone antibacterials. J Antimicrob
146. Abu-Elmagd K, Fung JJ, Alessiani M, et al. Strategy of FK506 Chemother 1994; 33 (4): 685-706
therapy in liver transplant patients: effect of graft function. 159. Chapuis L, Cadranel JF, Nordmann P, et al. Grand mal seizures
Transplant Proc 1991; 23: 2777-80 as a complication of treatment with pefloxacin in patients
147. Christians U, Braun F, Schmidt M, et al. Specific and sensitive with cirrhosis: a report of three cases. J Hepatol 1993; 19 (3):
measurement of FK506 and its metabolites in blood and urine 383-4
of liver-graft recipients. Clin Chem 1992; 38: 2025-32
148. Wiseman LR, Goa KL. Toremifene: a review of its pharmaco-
logical properties and clinical efficacy in the management of
advanced breast cancer. Drugs 1997; 54: 141-60 Correspondence and reprints: Dr Vanni Rodighiero,
149. Anttila M, Laakso S, Nyländen P, et al. Pharmacokinetics of the Dipartimento di Farmacologia, Università di Padova,
novel antiestrogenic agent toremifene in subjects with altered Largo E. Meneghetti 2, 35131 Padova, Italy.

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Nov; 37 (5)

You might also like