Sie sind auf Seite 1von 24

SPECIAL POPULATIONS Clin Pharmacokinet 1999 Jul; 37 (1): 17-40

0312-5963/99/0007-0017/$12.00/0

© Adis International Limited. All rights reserved.

Pharmacokinetics of Opioids in
Liver Disease
Irmgard Tegeder, Jörn Lötsch and Gerd Geisslinger
Center of Pharmacology, Institute of Clinical Pharmacology,
Johann Wolfgang Goethe-University of Frankfurt, Frankfurt am Main, Germany

Contents
Abstract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
1. Overview of the Pharmacokinetics of Opioids in Patients with Normal Liver Function . . . . . . . . . 20
2. Pharmacokinetics of Specific Opioids in Patients with Liver Disease . . . . . . . . . . . . . . . . . 20
2.1 Morphine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
2.2 Methadone . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 23
2.3 Pethidine (Meperidine) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
2.4 Dextropropoxyphene . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 25
2.5 Codeine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
2.6 Dihydrocodeine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
2.7 Buprenorphine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
2.8 Pentazocine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 27
2.9 Tramadol . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 28
2.10 Tilidine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
2.11 Fentanyl . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
2.12 Sufentanil . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 30
2.13 Alfentanil . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
2.14 Remifentanil . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
3. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33

Abstract The liver is the major site of biotransformation for most opioids. Thus, the
disposition of these drugs may be affected in patients with liver insufficiency.
The major metabolic pathway for most opioids is oxidation. The exceptions are
morphine and buprenorphine, which primarily undergo glucuronidation, and
remifentanil, which is cleared by ester hydrolysis.
Oxidation of opioids is reduced in patients with hepatic cirrhosis, resulting in
decreased drug clearance [for pethidine (meperidine), dextropropoxyphene, pen-
tazocine, tramadol and alfentanil] and/or increased oral bioavailability caused by
a reduced first-pass metabolism (for pethidine, dextropropoxyphene, pentazocine
and dihydrocodeine). Although glucuronidation is thought to be less affected in
liver cirrhosis, the clearance of morphine was found to be decreased and oral
bioavailability increased.
The consequence of reduced drug metabolism is the risk of accumulation in
the body, especially with repeated administration. Lower doses or longer admin-
istration intervals should be used to remedy this risk. Special risks are known for
pethidine, with the potential for the accumulation of norpethidine, a metabolite
18 Tegeder et al.

that can cause seizures, and for dextropropoxyphene, for which several cases of
hepatotoxicity have been reported. On the other hand, the analgesic activity of
codeine and tilidine depends on transformation into the active metabolites,
morphine and nortilidine, respectively. If metabolism is decreased in patients with
chronic liver disease, the analgesic action of these drugs may be compromised.
Finally, the disposition of a few opioids, such as fentanyl, sufentanil and
remifentanil, appears to be unaffected in liver disease.

Opioids are extensively used in the treatment of the effect may also change with respect to its mag-
pain in a diverse patient population. As the experi- nitude and time course.
ence of pain varies greatly among patients, and op- The rate of change of the amount (A) of drug in
ioid effects may vary between individuals, therapy the body can be described by:
needs to be individualised in order to achieve ade-
dA/dt = –kA (Eq. 2)
quate pain control. In addition to pain relief, opioids
induce several adverse effects, such as respiratory where k is the elimination rate constant. The half-
depression and sedation, and have actions on the life of a drug can be calculated as ln(2)/k. The
gastrointestinal tract that induce nausea, vomiting amount of drug in the body is related to the plasma
and constipation. A phenomenon still incompletely concentration by the volume of distribution (Vd):
understood but complicating the therapy is the de- A = Vd • Cp (Eq. 3)
velopment of tolerance.
It has been suggested that patients with severe If clearance (CL) = k • Vd, the rate of change of
liver disease have an increased sensitivity to opioid amount A of drug in the body is related to the
analgesics. Administration of opioids may cause ce- plasma concentration by:
rebral dysfunction or worsen the condition of a pa- dA/dt = CL • Cp (Eq. 4)
tient with pre-existing hepatic encephalopathy.[1,2]
Additionally, impaired liver function may translate The total clearance (CL) is a sum of several par-
into changes in pharmacokinetics. It is generally tial clearances, named after the site or mechanism
accepted that opioid effects are initiated by the of drug elimination, such as hepatic metabolic
binding of an opioid to specific receptors. The clearance (CLH) or renal excretory clearance
higher the number of opioid receptors occupied by (CLR).
opioid molecules, the higher the effect until a max- The effect of liver impairment becomes more
imum is reached. The effect is thus a function of pronounced as the contribution of hepatic clear-
the opioid concentration at the effect site. After sys- ance to total clearance increases. The efficiency of
temic administration, the time course of the con- drug removal by the liver is determined by hepatic
centration at the effect site (Ceff) is a function of the blood flow, hepatic enzyme capacity and plasma
plasma concentration (Cp): protein binding. Liver disease may affect any of
these conditions and, in addition, may lead to the
Ceff = ke0 e−ke0t ∗ Cp (Eq. 1) formation of porto-systemic shunts by which a
drug can be spared from hepatic elimination.
where ke0 is the rate constant for the transfer from Hepatic clearance may be described as:
the site of drug effect to the environment[3] and the
CLH = QH • EH (Eq. 5)
asterisk denotes convolution (the mathematical op-
eration by which the time courses of drug concentra- where QH is the hepatic blood flow and EH is the
tion in plasma and at the effect site are combined). hepatic extraction ratio describing the efficiency of
When the plasma concentration time course changes, drug removal by the liver. When EH approaches 1,

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)
Opioids in Liver Disease 19

the hepatic clearance approximately equals the he- In contrast to high-extraction drugs, the hepatic
patic blood flow (CLH ≈ QH). Thus, the clearance clearance of drugs with a low extraction ratio (EH
of drugs highly extracted by the liver (EH > 0.7), < 0.3), such as methadone, largely depends on the
such as pethidine (meperidine), pentazocine and intrinsic hepatic clearance (CLint), which describes
dextropropoxyphene, largely depends on liver the metabolic activity, and on plasma protein bind-
ing, because drug unbound to proteins is available
blood flow.
for elimination and the drop in drug concentration
In patients with liver disease, the clearance of
across the liver is small. When EH is small, hepatic
these drugs is mainly affected by reduced liver clearance may be described as:
blood flow or porto-systemic intra- and extra-hepatic
shunting. However, in liver cirrhosis the clearance CLH = CLint • fu (Eq. 6)
of some high-extraction drugs, such as propranolol,
where fu denotes the fraction of drug that is not
were found to be reduced primarily by decreased
bound to proteins. Therefore, the clearance of low-
enzyme capacity.[4,5] Another effect of reduced extraction drugs in patients with liver disease is
liver clearance is an increase in the oral bioavaila- mainly affected by a reduced enzyme capacity or
bility of high-extraction drugs because of reduced altered protein binding (for drugs with fu < 0.1).
first-pass metabolism and/or portal blood bypassing An increased free fraction of a drug caused by
the liver via porto-caval shunts. a reduced level of plasma proteins may counter-

Table I. Pharmacokinetic parameters of commonly used opioids in patients with normal liver and renal function
Drug F (%) PB (%) CL (ml/min) Vss (L) t1⁄2β (h) References
Alfentanil 88-92 300-500a 30-70a 1.5-2 14-18
Buprenorphine 50-55b ~96 650-1300 200-400 3-6 (-23c) 19-24
Codeine 50-55 4-7 210-350a 2.5-3.5 25-29
O-Demethyl-tramadol ~9 30
Dextropropoxyphene 30-70 80 600-1200 700-1800 11-16 31-33
Dihydrocodeine 12-34 80-90a 3.3-4.5 34
Fentanyl 80-86 600-1000 200-560a 3-4 14, 35-39
(-15d)
(-8.7e)
Methadone 41-99 70-90 50-200 240-330a 19-58 40-44
Morphine 15-50 20-35 800-2000 70-330 1.5-4.5 (~2) 45-55
Morphine-3-glucuronide 150-190 8-30 1-2 47, 56
Norpethidine 12-24 57, 58
Norpropoxyphene 220-450 23-37 32, 33
Nortilidine 99 3.3-4.9 59
Pentazocine <20 1200-1700 350-500 2-5 (-10c) 8, 60-62
Pethidine (meperidine) 48-56 60-80 470-730a 260-320a 3-7 63-66
Remifentanil 3000-5000 25-40 10-20 min 67-70
Sufentanil 91-93 700-1500a 120-380a 2.5-3.5 (-12.8e) 14, 17, 71-75
Tilidine 6 59
Tramadol 65-75 440-490 200-300 5-6 30, 76
a Normalised for 70kg adult.
b Sublingual.
c Enterohepatic recirculation.
d Patients >60 years of age.
e Long sampling time to define the terminal elimination phase.
CL = clearance; F = oral bioavailability; PB = protein binding; t1⁄2β = terminal elimination half-life; Vss = volume of distribution at steady state.

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)
20 Tegeder et al.

balance the effect of a decrease in enzyme activity. of binding varies from <10% for codeine to >95%
In addition, when the protein binding of drugs that for buprenorphine. Opioids distribute rapidly in all
are usually highly bound to proteins is reduced, body tissues. Most opioids have a large Vd (of
more than the normal amount of the drug can dis- about 3 to 6 L/kg).
tribute into tissues. Since the amount of drug in the Opioids are metabolised to more polar com-
body remains unaffected, the Vd can increase. In pounds, predominantly by the liver, although extra-
addition, since the drug trapped in the tissues is not hepatic metabolism, especially in the kidneys, con-
eliminated, the half-life of those drugs may in- tinues to be a focus for research. Most of the
crease. metabolites are less active than the parent com-
With intermediately extracted drugs (0.3 < EH < pounds and do not have clinically relevant pharma-
0.7), such as codeine and alfentanil, hepatic clear- cological actions. Some metabolites, however, are
ance may be altered by liver blood flow, protein as or more potent as opioid agonists as the parent
binding and enzyme activity. drug (e.g. morphine as a metabolite of codeine,
The function of various metabolising enzymes morphine-6-glucuronide or O-demethyl-tramadol)
appears to be differently affected in patients with and may produce analgesic effects as well as ad-
liver disease. It has been shown that the clearance verse effects such as respiratory depression. On ac-
of low-extraction drugs oxidised by the micro- cumulation, norpethidine may even cause severe
somal cytochrome P450 (CYP) enzyme system is adverse effects not typical for pethidine itself.
reduced in patients with chronic liver disease.[4-8] The majority of metabolites are excreted via the
In contrast, the clearance of low-extraction drugs kidneys and may accumulate in patients with renal
that undergo predominantly glucuronidation was failure. In addition to renal excretion, glucuronide
not reduced.[9-12] However, in patients with severe metabolites may be excreted via the biliary system;
cirrhosis there is some evidence that glucuronida- enterohepatic recirculation has also been suggested
tion is also impaired.[13] for some of these drugs (e.g. buprenorphine and
As the liver is the major site of opioid metabo- morphine).
lism, alterations in drug disposition may be expected
in patients with liver disease. This article reviews 2. Pharmacokinetics of Specific
the pharmacokinetics of the commonly used opioids Opioids in Patients with Liver Disease
in patients with liver disease.

1. Overview of the Pharmacokinetics 2.1 Morphine


of Opioids in Patients with Normal
Liver Function 2.1.1 Pharmacokinetics
Morphine is well absorbed from the gastrointes-
The pharmacokinetic parameters of the com- tinal tract. However, it undergoes substantial first-
monly used opioids in patients with normal liver pass metabolism, predominantly in the liver, result-
and renal function are summarised in table I. When ing in an oral bioavailability of only 20 to
not mentioned otherwise, half-life means terminal 40%.[45,77,78] Peak plasma drug concentrations (Cmax)
elimination half-life (t1⁄2β) and Vd is at steady state are reached within 15 to 20 minutes of intramuscu-
(i.e. Vss). lar and subcutaneous administration, [40] and within
Although most opioids are quite well absorbed 30 to 90 minutes of oral administration of a typical
from the gastrointestinal tract, their oral bioavaila- immediate-release formulation.[45]
bility is reduced by presystemic metabolism in the After absorption, morphine is rapidly distributed
gut wall and the liver (first-pass effect). All opioids in the body. It easily crosses the blood-brain bar-
bind to plasma proteins, generally to albumin and rier, but at a considerably lower rate than more lipo-
α1-acid glycoprotein (AAG); however, the extent philic opioids such as methadone or fentanyl.[79-81]

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)
Opioids in Liver Disease 21

Approximately one-third of morphine binds to bolised to normorphine, another active metabo-


plasma proteins.[40] Its Vd is 1 to 6 L/kg.[45,77,46,47,56] lite.[110] Its concentrations are usually too small to
The primary site of morphine metabolism is the be detected in plasma. However, normorphine may
liver, where it undergoes rapid glucuronidation. be neurotoxic.[111] The amount of morphine ex-
The high extraction of morphine (60 to 70%)[82,83] creted unchanged in urine is less than 10%.[45,77]
results in a high systemic clearance (about 1.5 to 2 Enterohepatic recirculation is thought to account
L/min) and a short t1⁄2β of approximately 2 for traces of morphine still found in urine and fae-
hours.[47,77,84,85] Extrahepatic metabolism was found ces several days after administration.[84,112] How-
to contribute considerably (up to 38%) to the total ever, it is not clear whether this is of clinical im-
body clearance of morphine.[83,86] A simple calcu- portance. In patients with renal insufficiency, the
lation shows that the assumption of extrahepatic glucuronide metabolites may accumulate because
morphine metabolism is reasonable: assuming an they are mainly excreted by the kidneys.[48]
EH of 0.7 and a QH of 25 ml/min/kg,[87] hepatic There is still controversy surrounding the con-
clearance as calculated by CLH = EH • QH should tribution of M6G to the effects seen after the ad-
not exceed 17.5 ml/min/kg. Systemic morphine ministration of morphine. M6G is a potent opioid[107]
clearance, however, exceeds this value by about 30%, and there is strong evidence that it contributes to
strongly indicating that the liver cannot be the only the clinical effects[113] of morphine in humans.[114]
site of morphine metabolism, which is an indirect After intrathecal administration in humans it pro-
evidence for extrahepatic metabolism of the drug. duced analgesic effects with potency approxi-
The principal metabolite, morphine-3-glucuron- mately 2.6 times greater than that of morphine.[104]
ide (M3G), has no opioid agonistic effects.[88,89] Its Although previous clinical investigations sug-
role is still controversial. There are several reports gested analgesic effects after systemic administra-
that M3G antagonises the analgesic activity of tion,[115,116] it was recently demonstrated in a study
morphine and morphine-6-glucuronide (M6G), with placebo and positive (morphine) control that
contributing to morphine tolerance.[90-93] This an- M6G had neither clinical nor analgesic effects
tagonism appears not to be mediated by opioid re- when administered as an intravenous bolus plus a
ceptors.[94-98] On the other hand, several other re- 4-hour intravenous infusion.[117] This points to-
ports question the hyperalgesic action of M3G.[99-103] wards a slow and low brain uptake of plasma
M6G is produced in smaller amounts than M3G. M6G,[118] thereby challenging earlier reports of its
In contrast with M3G, the opioid agonist activity ability to easily cross the blood-brain barrier. This
of M6G is not in doubt. M6G appears to be at least had previously been explained by the existence of
as potent as morphine.[104-108] Both M3G and M6G 2 energetically favourable conformations of the
are excreted primarily by the kidney and account M6G-molecule: extended in hydrophilic environ-
for between 55 and 60%, and approximately 10%, ments and folded in lipophilic environments.[119]
respectively, of a given dose recovered in urine. However, with the long-term administration of
After the administration of oral morphine the area morphine, more M6G might enter the brain with
under the plasma concentration-time curve (AUC) time and it may thus become more relevant. This
of M6G exceeded that of morphine by a factor of view is supported by the observation that after
4 : 1[45,89,109] to 9 : 1.[77] In contrast, after the intra- short-term administration oral morphine is only
venous administration of morphine, the M6G : one-sixth to one-eighth as potent as parenterally
morphine ratio is about 1 : 1 because of the ab- administered morphine, but that after repeated oral
sence of first-pass formation of M6G.[47] administration it is half to one-third as potent.[120]
M6G has a plasma clearance of about 10.2 L/h In addition, M6G was reported to be a substrate of
and a Vd of 10 to 30L, resulting in a t1⁄2β of 1 to 2 P-glycoprotein,[121,122] as is morphine.[123] It is,
hours.[47,56] Small amounts of morphine are meta- therefore, possible that coadministration of inhib-

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)
22 Tegeder et al.

Table II. Pharmacokinetics of morphine in patients with hepatic cirrhosis


Route Dose n Liver function F (%) CL (ml/min) Vss (L) t1⁄2β (h) Reference
Intravenous 0.1 mg/kg 6 NL 2345a ± 560 295a ± 65 1.85 ± 0.53 49
8 LCI 1470a ± 525 314a ± 62 3.35 ± 0.65 49
0.15 mg/kg 6 NL 1233 ± 427 203a ± 168 2.5 ± 1.5 124
6 LCI 1153 ± 345 161a ± 91 2.2 ± 1.3 124
1mg 8 NL 735a ± 98 1.67 ± 0.37 83
8 LCI 546a ± 49 2.02 ± 0.23 83
4mg 6 NL 1960a ± 161 287a ± 35 4.2 ± 0.2 125
7 LCI 798a ± 91 280a ± 70 1.7 ± 0.3 125
Oral 20mg 6 NL 47 ± 5.8 3.3 ± 0.6 125
10mg 6 LCI 101 ± 16.4 5.5 ± 0.8 125
a Normalised for 70kg adult.
CL = clearance; F = oral bioavailability; LCI = liver cirrhosis; NL = normal liver function; t1⁄2β = terminal elimination half-life; Vss = volume of
distribution at steady state.

itors of P-glycoprotein, such as verapamil or cyclo- clinical data presented by Patwardhan et al.[124]
sporin, lead to an increased brain uptake of M6G, suggest that their patients had less severe hepatic
thereby enhancing its effects on the central nervous dysfunction (hypoalbuminaemia and hyperbilirubin-
system. aemia, normal prothrombin time, 3 patients with
ascites) compared with other studies that included
2.1.2 Effect of Liver Disease patients in Child-Pugh class C, encephalopathy or
The main findings of studies involving patients bleeding from oesophageal or gastric varices.[83,125]
with hepatic cirrhosis are summarised in table II. Using hepatic vein catheterisation, Crotty et al.[83]
An early report suggested that morphine can cause have provided a direct measurement of morphine
hepatic encephalopathy in patients with liver cir-
hepatic extraction in 8 patients with hepatic cirrho-
rhosis.[1] Although this was not confirmed by a
sis and 8 controls. The extraction ratio was 0.52 in
more recent study,[125] several authors have shown
the control group and was reduced by 25% in those
that the metabolism of morphine is significantly
with cirrhosis. The hepatic plasma flow was almost
impaired in patients with severe hepatic cirrho-
identical in both patient groups, so that the reduc-
sis.[49,83,125,126] The t1⁄2β was prolonged in those pa-
tients with cirrhosis who had marked decreases in tion in extraction ratio was probably because of
hepatic function. Compared with controls, t1⁄2β was impaired enzyme capacity and/or the presence of
approximately doubled (3 to 4 vs 1.5 to 2h),[49,125] intrahepatic shunts. However, the effect of cirrho-
which was attributable to a reduction in total body sis was less than that reported in similar studies of
clearance. Vd was not significantly altered. [49,125] high-extraction oxidised drugs such as proprano-
Hasselstrom et al.[125] have also reported that the lol[5] and lidocaine[4] (44 and 52% reduction of he-
bioavailability of oral morphine increased to 100% patic extraction in cirrhosis, respectively). This
in patients with cirrhosis as compared with a bio- supports the concept that glucuronidation may be
availability of about 30% in the control group. In relatively spared in cirrhosis. Specifically, the sys-
contrast to these results, Patwardhan et al.[124] temic clearance of low-extraction drugs that are
found that the systemic clearance and t1⁄2β of mor- metabolised exclusively by glucuronidation was
phine after intravenous administration were similar not reduced in cirrhosis.[9-11]
in both patients with cirrhosis and healthy volun- At present, this phenomenon may be explained
teers. These contradictory findings may be ex- as follows. The isoenzymes of glucuronyltrans-
plained by differences in the severity of liver dis- ferase are thought to be located in the membrane
ease in the patients enrolled in the studies. The of the endoplasmic reticulum.[127] The addition of

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)
Opioids in Liver Disease 23

detergents or other membrane-perturbing agents 2.2 Methadone


produces an increase in the enzyme activity in
2.2.1 Pharmacokinetics
vitro.[128] It has been postulated that the sparing of The analgesic activity of methadone is almost
glucuronidation in liver disease may be because of entirely produced by l-methadone, which is much
a similar activation of the enzyme by the disease more potent as an opioid agonist than the d-iso-
process.[129-133] mer.[140] Most kinetic studies are performed with
Crotty et al.[83] found a discrepancy between the racemic methadone. It is well absorbed from the
systemic clearance and the hepatic clearance in gastrointestinal tract, and unlike many other opioid
both the controls and patients with cirrhosis. The analgesics it has a considerably high oral bioavail-
percentage of systemic clearance unaccounted for ability (approximately 80%)[41,42] because its he-
patic extraction is low (0.09).[43]
by hepatic clearance was greater in the patients
At therapeutic concentrations, about 90% of
with liver disease (33%) than in the control group
methadone is bound to plasma proteins.[43] Metha-
(10%). In a study by Mazoit et al.,[86] total body done has a long t1⁄2β of about 30 hours (with a range
clearance exceeded hepatic clearance by 38%. These of 8.5 to 58h).[43,44,141,142] It appears to be firmly
findings provide indirect support for the idea that bound to proteins in various tissues with a gradual
morphine has an extrahepatic metabolism; this is accumulation after repeated administration.[142,143]
further substantiated by several studies that have Its Vd is about 2 to 5.5 L/kg.[42,43] Methadone
shown that morphine is conjugated with glucuronic undergoes extensive biotransformation in the liver.
acid in homogenates of human intestine,[134,135] The major metabolic pathways are N-demethyla-
kidney microsomes[136,137] and brain tissue.[138] tion and cyclisation to pyrrolidines and pyrrolines.
Thus, extrahepatic metabolism might contribute sub- The metabolites are excreted in urine and bile along
with small amounts of unchanged drug.[144,145] The
stantially to the overall clearance of morphine, es-
mean total plasma clearance is about 3 to 12 L/h,
pecially in patients with liver cirrhosis. renal clearance accounting only for 0.24 to 1.8
Olsen et al.[139] have shown that the plasma pro- L/h.[43,146]
tein binding of morphine was decreased in patients
2.2.2 Effect of Liver Disease
with hepatic failure. The decreased binding was
Kreek et al.[147] reported that the total 24-hour
attributed to lower albumin levels and the displace-
urinary excretion of methadone and its metabolites
ment of the bound drug caused by the high binding was significantly lower (32.6%) in patients with
affinity of bilirubin. The higher free fraction of liver disease compared with controls (48.3%). The
morphine in plasma, however, is not expected to urinary excretion of the pyrrolidone metabolite, an
increase the Vd because morphine is only moder- end-product of 2 pathways of methadone metabo-
ately protein bound.[124] lism, was also reduced. In a second study the same
Because the metabolism of morphine is reduced authors found the faecal excretion of methadone
in patients with liver cirrhosis, it should be used and its unconjugated metabolites to be unaltered in
with caution in these patients to avoid accumula- patients with liver disease as compared with a
control group.[148] The patients in these 2 studies
tion of the drug. A 1.5- to 2-fold increase in the
had chronic active, chronic persistent or alcoholic
administration interval has been suggested.[49] If
hepatitis without severe liver dysfunction.[147,148]
administered orally, the increased bioavailability In patients with biopsy-proven mild to moder-
has to be considered. Whether the lower formation ate chronic liver disease, Novick et al.[149] found
of M6G in patients with cirrhosis is associated with no change in the pharmacokinetic parameters of
a lower pharmacological effect has not yet been methadone compared with healthy individuals. In
evaluated. patients with more severe liver disease, the t1⁄2β was

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)
24 Tegeder et al.

somewhat longer, but drug disposition was not sig- tein were reduced in livers from patients with
nificantly altered.[149] In patients with severe alco- hepatocellular diseases.[155] Since excessive long
holic liver cirrhosis, however, the t1⁄2β of methadone term alcohol consumption may induce enzymes,
was significantly prolonged (32h) compared with the results obtained in patients with alcoholic liver
a control group of methadone-maintained alcohol cirrhosis might not be representative of patients
abusers with normal liver function tests (t1⁄2β with severe liver cirrhosis of other aetiologies.
19.7h).[150] The Vd was larger in patients with cir-
rhosis than in controls (716 vs 438L) whereas the 2.3 Pethidine (Meperidine)
apparent clearance after oral administration was 2.3.1 Pharmacokinetics
similar in both groups.[150] The patients with cirrhosis Pethidine was one of the first synthetic opioid
had somewhat lower dose-adjusted mean plasma analgesics introduced into clinical practice. It is
concentrations than the controls (2.8 vs 3.6 μg/L/ less potent than morphine. Pethidine is metabo-
mg),[150] which might have been a consequence of lised predominantly by the liver; its t1⁄2β is about 3.5
the greater Vd and possibly increased tissue storage hours.[63,64] The major metabolic pathway is hydro-
of the drug. None of the patients with severe liver lysis to meperidinic acid, which is consecutively
cirrhosis had signs or symptoms of methadone conjugated with glucuronic acid and excreted by
overdosage.[150] the kidney. Pethidine is also N-demethylated to
Since most disposition parameters of methadone norpethidine which is either metabolised to nor-
were normal in patients with hepatic cirrhosis, meperidinic acid or excreted unchanged in urine
Novick et al.[150] suggested that the usual metha- (about 6%).[65] Elimination of the pharmacologi-
done maintenance dosage may be continued even cally active norpethidine is slower (t1⁄2β 12 to 24h)
in patients with severe hepatic dysfunction. How- than that of pethidine, and the metabolite can accu-
ever, with alcohol abusers as controls the pharmaco- mulate in patients receiving repeated doses of
kinetic differences between patients with and with- pethidine.[57,58] High plasma concentrations of
out cirrhosis might have been underestimated, norpethidine have been related to the development
because this kind of control is more likely to have of neuromuscular irritability and seizures.[58]
compensated cirrhosis with normal liver function At normal urine pH, less than 5% of an admin-
tests than truly healthy individuals.[151] On the istered pethidine dose is excreted unchanged in
other hand, long-term excessive alcohol (ethanol) urine.
ingestion appears to increase the metabolism of Taken orally, pethidine is subject to extensive
methadone by stimulating its degradation in liver first-pass metabolism with a mean oral bioavaila-
microsomes, whereas acute alcohol intake inhibits bility of about 50%.[156] Approximately 60 to 80%
the metabolism of methadone at liver microsomal of pethidine in plasma is bound to proteins, mainly
sites,[152] leading to elevated methadone plasma to AAG.[157] When AAG levels increase (as in
concentrations.[153] These potential interactions stress) pethidine binding may also increase.[157]
and a missing second control group of truly healthy Pethidine has a large Vd (3 to 5 L/kg).[63,64]
individuals makes it difficult to interpret the results 2.3.2 Effect of Liver Disease
presented by Novick et al.[149,150] Using isolated perfused rat livers, Callaghan et
It has been demonstrated that 4 heterologously al.[158] have examined the effect of liver disease on
expressed CYP enzymes were able to catalyse the the hepatic extraction and clearance of pethidine,
N-demethylation of methadone. Referring to their using chronic carbon tetrachloride-induced cirrho-
relative content in the liver, however, it can be as- sis as a model of liver disease. The extraction ratio
serted that CYP3A4 is the major enzyme involved of pethidine was reduced from 0.91 in controls to
in the N-demethylation of methadone.[154] It has 0.79 in cirrhosis, and the intrinsic hepatic clearance
been shown that both the CYP3A4 mRNA and pro- was reduced from approximately 10.8 to 4.2 L/h.

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)
Opioids in Liver Disease 25

In patients with cirrhosis, it has been shown that widely prescribed, often in combination with para-
the presystemic metabolism of pethidine is strik- cetamol (acetaminophen) or aspirin (acetylsali-
ingly impaired, and its oral bioavailability may in- cylic acid), for the treatment of mild to moderate
crease to as much as 80%.[159] The reduced hepatic pain. The analgesic effect is produced by the dextro
extraction of pethidine in cirrhosis results in a de- isomer of propoxyphene, which is thought to be
crease of approximately 50% in plasma clearance half as potent as an analgesic as codeine when
and approximate doubling of the half-life of the given orally.
parent compound.[160] A prolonged half-life was Following oral administration, about 30 to 70%
also noticed in patients with acute viral hepati- of the drug escapes first-pass metabolism to enter
tis.[161] The use of normal doses of pethidine in these the circulation.[31] Plasma concentrations vary
patients may result in excessive sedation, respira- widely between individuals.[31] Dextropropoxy-
tory depression or other adverse opioid effects. phene is about 80% bound to proteins[163] and has
Pond et al.[159] have shown that the generation a large Vd (approximately 16 L/kg).[31,164] The ma-
of norpethidine is impaired in patients with cirrho- jor route of metabolism is N-demethylation to
sis, suggesting that these patients may be relatively
norpropoxyphene, which is excreted by the kid-
protected from its toxicity. Mean norpethidine
neys.[32] Norpropoxyphene is pharmacologically
plasma concentrations after intravenous and oral
active. It has, however, substantially lower CNS-
pethidine were 36 and 45% of the concentrations
depressant effects than dextropropoxyphene but is
in healthy individuals. However, further elimina-
thought to have a greater local anaesthetic ef-
tion of norpethidine was slower than in healthy
individuals, leading to an increased risk of cumu- fect.[165]
lative toxicity when repeated doses are adminis- The t1⁄2β of the parent compound and the meta-
tered. This is substantiated by a case of CNS tox- bolite range from 11 to 16 hours and 23 to 37 hours,
icity in a patient with cirrhosis after multiple days respectively,[31-33] with much longer half-lives re-
of pethidine use. The patient was receiving approx- ported in elderly patients (dextropropoxyphene 24
imately 400mg daily of intramuscular pethidine to 50h, norpropoxyphene 25 to 76h).[166] After re-
when symptoms of CNS toxicity (diaphoresis, ag- peated doses, the accumulation of norpropoxy-
itation, tremors and muscle spasms) occurred. The phene may be responsible for some of the observed
patient had normal renal function.[162] toxicity, such as cardiac dysrhythmias.[165,167-169]
Since the Vd of pethidine in patients with cir- Patients with cardiac diseases may be at a higher
rhosis was not affected[159] a single intravenous risk for this adverse effect.
dose should not be changed. When taken orally, the
2.4.2 Effect of Liver Disease
dose of pethidine should be reduced as the bio-
After oral administration of dextropropoxy-
availability is increased. Because the elimination
phene in patients with cirrhosis, plasma concentra-
of norpethidine is decreased in patients with liver
tions of dextropropoxyphene were considerably
disease, those patients may be at risk of norpethid-
higher and norpropoxyphene concentrations were
ine accumulation and neurotoxicity with repeated
doses, although less norpethidine is produced. much lower than in control patients.[170] This is
Therefore, pethidine should not be used for long probably because of a decreased first-pass metabo-
term pain management in any patient. lism of orally administered dextropropoxyphene
in these patients. The AUC ratio of norpropoxy-
2.4 Dextropropoxyphene phene : dextropropoxyphene was significantly lower
in patients with cirrhosis (0.5 to 0.9) than in con-
2.4.1 Pharmacokinetics trols (2.5 to 4)[170,171] and was significantly corre-
Dextropropoxyphene is chemically related to lated with the liver function.[171] The reduction of
methadone but has a low analgesic activity. It is norpropoxyphene generation may reflect both

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)
26 Tegeder et al.

porto- systemic shunting and reduced liver enzyme O-demethylation of codeine to morphine is catalysed
capacity. by the genetically polymorphic CYP2D6,[179,180,186]
Patients with cirrhosis, unlike healthy individuals, which is expressed in phenotypes, the most impor-
experienced considerable sedation after oral dextro- tant of which are extensive (EMs) and poor (PMs)
propoxyphene,[170] probably because of increased metabolisers of sparteine and debrisoquine. PMs
bioavailability. Several cases of hepatotoxicity account for about 7% of the healthy Caucasian pop-
with jaundice, upper abdominal pain and malaise ulation. PMs produce only trace amounts of mor-
attributable to dextropropoxyphene have been re- phine and, therefore, no analgesia is achieved after
ported.[172-175] The histological changes seen at bio- the administration of codeine.[180,182,187,188]
psy consisted of centrilobular cholestasis, portal Conflicting results have been reported concern-
tract inflammation and bile duct abnormalities, ing adverse drug reactions in EMs and PMs. In the
mimicking large bile duct obstruction.[172] Dextro- study of Poulsen et al.,[188] only EMs experienced
propoxyphene, therefore, should be avoided in pa- adverse drug reactions, whereas Eckhardt et al.[180]
tients with hepatic insufficiency. found no difference between the phenotypes. Since
the oxidative enzyme capacity of the liver has been
2.5 Codeine shown to be impaired in patients with hepatic cir-
rhosis[4-8] it can be assumed that patients with cir-
2.5.1 Pharmacokinetics
rhosis will experience little or no analgesia from
Codeine, which is chemically methylmorphine,
codeine. Even worse, the use of codeine in these
is a naturally occurring phenanthrene alkaloid. Co-
patients might expose them to the adverse effects
deine is principally metabolised by the liver to co-
caused by codeine itself. However, this is chal-
deine-6-glucuronide (approximately 60%) which
lenged by reports that the CYP isoenzymes are af-
is excreted by the kidneys.[25] The drug also under-
fected to a different extent by cirrhosis,[189,190] with
goes N-demethylation to norcodeine.[176] A small
CYP2D6 being relatively spared.[190] Thus, until
fraction (about 10%) of codeine is O-demethylated
data on the pharmacokinetics and analgesic effects
to morphine.[176] Both morphine and its glucuronide
of codeine in patients with cirrhosis become avail-
conjugates can be found in the urine after therapeu-
able, alternative analgesics should be preferred in
tic doses of codeine.[177]
these patients.
Only 4 to 12% of a given codeine dose is ex-
creted unchanged in urine.[176,177] Codeine has an
exceptionally low affinity for opioid receptors.[178] 2.6 Dihydrocodeine
Its analgesic effect is thought to be caused by its
conversion to morphine.[179-183] However, its anti-
2.6.1 Pharmacokinetics
tussive actions might involve distinct receptors that
Dihydrocodeine has elimination pathways sim-
interact with codeine itself.[184] Probably because
ilar to codeine, that is, glucuronidation and de-
of first-pass metabolism, morphine : codeine AUC
methylation. In EMs of sparteine and debrisoquine
ratios were greater after oral than intramuscular ad-
(CYP2D6), the following metabolites were recov-
ministration.[25]
ered in urine after oral administration of dihydro-
The bioavailability of orally administered codeine
codeine 60mg: dihydrocodeine glucuronide (27.7%),
is approximately 50 to 55%.[25,26,185] In plasma,
nordihydrocodeine (15.8%), nordihydrocodeine
less than 10% of codeine is bound to proteins.[164]
glucuronide (6.3%) dihydromorphine (0.5%) and
Codeine has a Vd of 3 to 4 L/kg. The t1⁄2β is 2 to 4
dihydromorphine glucuronide (8.4%). Approxi-
hours.[26,27]
mately 30% of the dihydrocodeine dose was excreted
2.5.2 Effect of Liver Disease unchanged in the urine.[191] In PMs of sparteine,
As mentioned in section 2.5.1, codeine analge- dihydrocodeine O-demethylation to dihydromor-
sia requires the formation of morphine.[179,180] The phine was found to be impaired.[191]

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)
Opioids in Liver Disease 27

Orally administered dihydrocodeine undergoes approximately 35 hours,[19] respectively. About


extensive first-pass metabolism, resulting in a bio- 96% of the circulating drug is bound to protein,[23]
availability of approximately 20%.[34] The half-life and its Vd is 200 to 400L.[19,22,24]
varies between 3.3 and 4.5 hours.[34]
2.7.2 Effect of Liver Disease
2.6.2 Effect of Liver Disease
The N-dealkylation of buprenorphine to nor-
In contrast to codeine, quinidine, an inhibitor of
buprenorphine is catalysed by the hepatic CYP en-
CYP2D6, did not change the effects of dihydro-
zyme system, with CYP3A4 as the major isoen-
codeine on pain thresholds.[192] From that it was
zyme.[196,197] Since it has been shown that the
concluded that dihydrocodeine produces analgesia
expression of the CYP3A proteins is significantly
independent of its biotransformation to dihydro-
reduced in patients with severe chronic liver dis-
morphine.[192,193] Patients with cirrhosis can, there-
ease,[189] the metabolism of buprenorphine is sus-
fore, be expected to experience pain relief from
pected to be altered in liver cirrhosis. Therefore,
dihydrocodeine. However, since oral bioavailabil-
dosage adjustments may be required in patients
ity may be increased and hepatic clearance de-
with liver insufficiency. Until sufficient pharmaco-
creased in patients with cirrhosis, dihydrocodeine
kinetic data on buprenorphine and its metabolites
should be used with caution in these patients.
in patients with liver disease become available, the
drug should be used cautiously in these patients.
2.7 Buprenorphine

2.7.1 Pharmacokinetics
2.8 Pentazocine
Buprenorphine is a semisynthetic, highly lipo-
philic, opioid derived from thebaine which has
been categorised as a partial agonist at μ opioid 2.8.1 Pharmacokinetics
receptors. It appears to display a ceiling of its ef- Pentazocine acts as a κ agonist and a partial ag-
fects at high doses. onist at μ opioid receptors. Its analgesic actions are
When administered orally, buprenorphine under- produced by the l isomer; the d-isomer has little
goes extensive first-pass metabolism and its oral affinity for opioid receptors.
bioavailability is insufficient to achieve analgesic Pentazocine is well absorbed from the gastroin-
drug concentrations. Administered sublingually, testinal tract, but first-pass metabolism in the liver
however, about 50 to 55% of a buprenorphine dose is extensive. Less than 20% of a dose of pentazo-
may escape first-pass metabolism and enter the cine enters the systemic circulation after oral ad-
systemic circulation.[19-21] Buprenorphine is meta- ministration.[60] The half-life in plasma is approx-
bolised by glucuronidation and dealkylation pre- imately 3 hours[60,61] and the Vd is 4 to 5 L/kg.[60,61]
dominantly in the liver to buprenorphine-3-glucu- The action of the drug is terminated largely by
ronide, which is inactive, and norbuprenorphine, biotransformation in the liver, with only 5 to 8% of
which has an analgesic effect of approximately a dose excreted unchanged in the urine. The meta-
one-fiftieth of that of the parent drug.[194] Both the bolites, products of the oxidation of the terminal
N-dealkylated and conjugated metabolites can be methyl groups and glucuronide conjugates, are ex-
detected in the urine (about 10% of a dose).[195] creted largely by the kidney.[198] Individuals who
However, most of the drug (approximately 80 to smoke metabolise pentazocine 40% more than
90%) is excreted as its metabolites via the biliary nonsmokers.[199] There is a considerable interindi-
system,[195] and enterohepatic recirculation was vidual variability of the rate of pentazocine meta-
suggested.[195] bolism. This may account for the variability of the
After intravenous administration, the t1⁄2β of analgesic response, which has been found to be
buprenorphine and norbuprenorphine have been quite unpredictable when the drug was adminis-
reported to be approximately 3 to 6 hours,[20,22] and tered orally.[200]

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)
28 Tegeder et al.

2.8.2 Effects of Liver Disease methylated compounds undergo further sulpha-


In patients with hepatic cirrhosis, a 2- to 3-fold tion or glucuronidation.[205] 5 metabolites arising
increase in the bioavailability of orally adminis- from phase I reactions and 6 from phase II reactions
tered pentazocine has been reported, i.e. approxi- are known.[205] The main metabolite (M1), O-
mately 70% of a given dose entered the circulation demethyl-tramadol, is an agonist at the μ opioid
in patients with cirrhosis compared with approxi- receptor with a higher affinity than the parent com-
mately 20% in a healthy control group.[8,201] Com- pound.[178] Tramadol and its metabolites are
pared with healthy individuals, systemic clearance mainly excreted via the kidneys (90%); the remain-
was approximately halved and t1⁄2β doubled, whereas ing 10% appear in faeces.[205] Approximately 15%
Vd was unchanged.[8,201] The AUC ratio between of a dose of tramadol is excreted unchanged in the
patients with healthy liver function and patients urine.[205] The t1⁄2β of tramadol and its O-demethyl
with cirrhosis (AUChealthy : AUCcirrhosis) was 2.0 derivative (M1) were reported to be 5 to 6 and 9
after intravenous administration and 8.3 when pen- hours, respectively.[30,76,206,207] Pharmacokinetic
tazocine was taken orally.[8] differences between the enantiomers have not been
Assuming that the total response is related to the reported.
AUC, a remarkable increase of the effects of pen-
tazocine is expected in patients with cirrhosis, es- 2.9.2 Effects of Liver Disease
pecially when the drug is given orally. This agrees O-Demethylation of tramadol to the active M1
with the observations that hepatic insufficiency ap- metabolite seems to depend on the polymorphic
pears to predispose patients to more pronounced enzyme CYP2D6.[206] O-Demethyl-tramadol has a
pentazocine adverse effects (marked apprehension, higher affinity for opioid receptors than tramadol
anxiety, dizziness and sleepiness).[202] itself, and has been shown to have analgesic activ-
Because of a higher bioavailability, oral doses ity in mice and rats as assessed by the tail flick
of pentazocine should be reduced substantially in response.[178] In PMs of sparteine, analgesic effects
patients with cirrhosis. When multiple oral or par- of tramadol on experimental pain were reduced
enteral doses are required, administration intervals compared with those seen with EMs.[208] From that,
should be lengthened or subsequent doses should it was concluded that the opioid activity of tramadol
be reduced below the initial dose because of the may well be a consequence of the metabolism of
lower systemic clearance of the drug.[201] the parent drug to the active demethylated metabo-
lite.[208] Since the oxidative enzyme capacity ap-
2.9 Tramadol pears to be reduced in patients with liver cirrhosis,
the analgesic activity of tramadol might be less
2.9.1 Pharmacokinetics than expected in patients with severe liver disease.
Tramadol, a racemic mixture of 2 enantiomers, However, this has not yet been shown directly.
is a synthetic 4-phenylpiperidine analogue of co- The plasma concentration and t1⁄2β of tramadol
deine. It is a central analgesic with low affinity for in patients with liver cirrhosis was increased by a
opioid receptors. Its analgesic effects result in large factor of 2 to 3 compared with volunteers with
part from activation of monoaminergic spinal inhi- healthy liver function.[209] The same source reports
bition of pain pathways.[203,204] an increase in the renal clearance of unchanged
Oral tramadol is rapidly absorbed, with a bio- drug to 30% in these patients, compared with ap-
availability of 65 to 75%. Cmax is reached within 2 proximately 10% in healthy volunteers.
hours. Plasma protein binding is 20% and Vd about Since it is not yet known whether patients with
200 to 300L.[30,76] liver cirrhosis experience sufficient pain relief after
Tramadol undergoes extensive biotransforma- recommended doses of tramadol, and because of
tion in the liver via 2 main metabolic pathways to the potential for delayed elimination and accumu-
O- and N-demethylated compounds. The O-de- lation of the drug, it appears advisable to consider

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)
Opioids in Liver Disease 29

alternative agents until more information becomes Since the analgesic effects of tilidine depend on
available. demethylation to nortilidine, and since systemically
available naloxone antagonises analgesic effects of
2.10 Tilidine nortilidine, it may be assumed that patients with
severe liver cirrhosis experience none, or only mi-
2.10.1 Pharmacokinetics
nor, analgesia after the administration of tilidine.
Tilidine is a prodrug from which the active met-
Although this assumption has not yet been confirmed
abolite nortilidine is formed by demethylation,
by clinical studies, it appears wise to use alterna-
mainly during first-pass metabolism in the liver
tive analgesics in patients with severe liver disease.
after oral administration of the drug.[59,210] The
systemic oral bioavailability is only about 6% for
2.11 Fentanyl
the parent substance. About 99% is converted into
the active metabolite nortilidine.[59] Nortilidine is
2.11.1 Pharmacokinetics
further metabolised in the liver to bisnortilidine Fentanyl is a highly lipid-soluble synthetic op-
and other polar compounds, which are eliminated ioid related to the phenylpiperidines, and is com-
by the kidneys. Only 3% of an oral dose of tilidine monly used as a supplement to general anaesthesia.
is recovered in the urine as nortilidine and less than It is primarily a μ agonist and is estimated to be 80
0.1% as tilidine.[59] The t1⁄2β of nortilidine and times as potent as morphine as an analgesic.[212]
bisnortilidine are 3.5 to 5 hours, and 5 to 7 hours, Following intravenous administration of fen-
respectively.[59] tanyl, the onset of action is within one circulation
Tilidine is often combined with naloxone as an time, suggesting a rapid and extensive uptake by
oral formulation in order to prevent intravenous the brain, the site of action.[213] The short duration
abuse. Because of a high first-pass metabolism, of effect (about 30 min) after the administration of
naloxone normally does not reach the systemic cir- a single intravenous bolus dose is because of its
culation after oral administration. Therefore, given rapid redistribution from the sites of action in the
orally, it does not interfere with the analgesic effects brain to storage sites (muscle and fat) and biotrans-
of nortilidine. However, with intravenous admin- formation (liver).[213] With repeated administra-
istration, the opioid agonistic affects of nortilidine tion, continuous infusion or high doses, accumula-
would be antagonised by naloxone. tion may lead to a prolonged duration of action.
2.10.2 Effects of Liver Disease The apparent Vd of fentanyl is about 3 to 5
Data presented here were obtained from the L/kg[35,214] but has been shown to increase with
manufacturer.[211] In a nonblind single-dose trial, continuous infusion resulting in a prolongation of
the pharmacokinetic parameters of tilidine + nalox- the t1⁄2β.[215-217] Additionally, after prolonged con-
one were evaluated in 8 patients with compensated tinuous infusion of fentanyl, the duration of effect
hepatic cirrhosis. Compared with the results of pre- may not be limited by redistribution, but by the
vious studies in healthy individuals, the AUC (812 clearance of the drug which is much slower than
vs 993 μg/L • h in patients with cirrhosis and redistribution.[37] In plasma, about 85% of fentanyl
healthy individuals, respectively) and the Cmax of is bound to proteins, mainly to albumin (50 to
nortilidine (88.6 vs 158.0 μg/L) were reduced in 60%);[14,218] binding to AAG is of minor impor-
the patients with cirrhosis, whereas the t1⁄2β was tance.[218]
prolonged (9.9 vs 4.6h). In contrast to studies with The elimination of fentanyl from the body is
healthy individuals, naloxone could be measured governed by its reuptake from storage sites and its
in plasma from patients with liver cirrhosis. Thus, metabolism in the liver. Fentanyl undergoes N-
it can be assumed that the capacity for oxidative dealkylation and hydroxylation to compounds that
metabolism of tilidine and for glucuronidation of are both inactive and nontoxic. The main metabo-
naloxone is reduced in these patients. lites, 4-N-(N-propionylanilino)-piperidine and 4-N-

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)
30 Tegeder et al.

(N-hydroxypropionylanilino)-piperidine, and a small blood flow.[228] Since fentanyl is a high-extraction


amount of unchanged drug (<10%) are excreted in drug, its clearance will be relatively sensitive to
the urine.[38,219] Fentanyl is eliminated with a half-life changes of liver blood flow.
of about 3.5 hours,[35,36,38,220] but there is consider- The disposition of fentanyl appears to be rela-
able variability interindividually.[221,222] In patients tively unaffected by hepatic cirrhosis, although
undergoing cardiac[223,224] or abdominal aortic[39] pharmacokinetic alterations might occur with se-
surgery, longer t1⁄2β values of 5 to 7 and 8.7 hours, vere liver dysfunction or reduced hepatic blood
respectively, have been reported. flow. It is not yet known whether accumulation of
Studies investigating the pharmacokinetic and fentanyl during continuous infusion is more pro-
pharmacodynamic relationship of the phenylpiper- nounced in patients with liver cirrhosis compared
idine derivatives, however, suggest that a knowl- with patients with normal liver function.
edge of the terminal elimination half-life of the
drug is of limited value[225] because it does not help 2.12 Sufentanil
to predict to what extent the concentration decline
of a drug might be prolonged after repetitive dos- 2.12.1 Pharmacokinetics
ages or drug infusion. Hughes et al.[226] therefore Sufentanil is a synthetic, highly lipophilic. op-
proposed the use of a ‘context-sensitive half-time’, ioid chemically related to fentanyl. It is approxi-
defined as the time required for the plasma drug mately 5 to 10 times more potent than fentanyl as
concentration to decline by 50% starting from the an analgesic.[229]
plasma concentration found at the end of the infu- Sufentanil distributes rapidly and extensively to
sion. The word ‘context’ refers to the infusion du- all tissues, with a Vd of about 3 to 5 L/kg.[71,72,230]
ration. ‘Context-sensitive half-time’ means how In plasma, sufentanil is 90 to 93% bound to pro-
long it takes for the plasma concentration measured teins, mainly to AAG.[14,231] In neonates, protein
at the end of an infusion of a certain duration to binding is only about 80% because of lower AAG
drop by 50%. plasma levels.[232] Protein binding is independent
of sufentanil plasma concentration but is affected
2.11.2 Effects of Liver Disease by plasma pH. An increase in plasma pH from 7.4
In patients with histologically confirmed he- to 7.8 decreases binding by 28%, and conversely,
patic cirrhosis who underwent surgery, Haberer et lowering the pH increases the binding.[14]
al.[35] found no change in the pharmacokinetics of Sufentanil is extensively metabolised by the
fentanyl compared with controls with healthy liver liver (hepatic extraction ratio 0.8) by N-dealkylat-
and kidney function. However, none of the patients ion to products thought to be inactive, by O-de-
had profound hepatic insufficiency, as indicated by methylation to a metabolite possessing approxi-
the clinical data of the patients presented in that mately 10% of the activity of the parent compound
paper (normal serum albumin and total protein lev- and by some additional conjugation reactions. The
els, only minor reductions of the prothrombin time metabolites are found in both urine and faeces.
in most patients).[35] Only 1 to 2% of an injected dose of sufentanil is
In patients undergoing abdominal aortic surgery, recovered unchanged in urine.[233]
Hudson et al.[39] reported a lower total body clear- The t1⁄2β in patients who underwent general sur-
ance and larger Vd of fentanyl resulting in a pro- gery was reported to be about 3 hours[71,72,230] and
longed t1⁄2β (8.7h), compared with previously reported approximately 5 hours in adults undergoing car-
mean values (3 to 4h). The aortic cross-clamping diac surgery.[234] However, the t1⁄2β might have been
in that study might have altered the distribution and underestimated because the duration of sampling
elimination of fentanyl. Aortic cross-clamping ac- (8 hours or less) might have missed the last expo-
tivates the renin-angiotensin system[227] which can nential of the disposition function. In a study where
result in a decrease of the mesenteric and hepatic blood samples were taken up to 48 hours after the

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)
Opioids in Liver Disease 31

administration of sufentanil, the terminal elimina- and brain (t1⁄2,ke0 0.96 min[237] compared with 6.6
tion half-life was approximately 13 to 16 hours.[73,235] and 6.2 min for fentanyl and sufentanil[238,239]),
which leads to a faster onset of effects. The Vd of
2.12.2 Effects of Liver Disease
alfentanil is smaller than that of fentanyl, ranging
In patients with liver cirrhosis undergoing sur-
between 0.3 and 1 L/kg.[15,16,225,240-243] In plasma,
gery, Chauvin et al.[74] found no change in sufen-
alfentanil is 88 to 92% bound to proteins, [14,244]
tanil kinetics compared with age-matched con-
mostly to AAG.[14] This proportion is independent
trols. Plasma protein binding was similar in the
of plasma alfentanil concentrations or changes in
control group (91.7%) and in patients with cirrho-
plasma pH,[14] but may be affected by changes in
sis (90.4%). However, neither the plasma albumin
plasma levels of AAG.[244]
nor AAG plasma levels in these patients with cir-
rhosis differed from those of the controls, which Alfentanil is almost exclusively eliminated by
may partly explain why the sufentanil free fraction metabolism. Only 0.4% of a dose is recovered in
remained unaffected. urine as unchanged drug.[245] The main metabolic
pathways are oxidative N- and O-dealkylation,[243]
In addition to plasma protein concentrations,
leading to inactive compounds that are cleared by
binding of sufentanil may be influenced by pH
the kidneys.[243] The enzyme primarily responsible
changes possibly leading to alterations of tissue
for the metabolism is CYP3A4.[246] The estimated
distribution. Sufentanil has a pKa of 8.0, and there-
hepatic extraction ratio of 0.3 to 0.5[247] suggests
fore at physiological pH only a small fraction (ap-
that the total body clearance of alfentanil could be
proximately 1.6%) is in the un-ionised form. With
influenced by changes in both hepatic blood flow
alkalosis, which may occur in patients with severe
and intrinsic clearance. In contrast to fentanyl, the
liver dysfunction because of elevated ammonia
duration of effect of a single dose of alfentanil de-
levels, the proportion of the un-ionised form in-
pends more on total body clearance than on redis-
creases and protein binding decreases. In neurosur-
tribution to tissues, probably because of its lower
gical patients undergoing hyperventilation, it has
lipid solubility when compared with fentanyl.
been shown that an increase in the plasma pH from
7.37 in controls to 7.51 in the patients with hypo- The t1⁄2β of alfentanil is approximately 1.5 hours
capnia resulted in an increase in the total Vd (5.4 after a single intravenous dose.[15,16,225,241-243,248]
vs 3.5 L/kg in controls) and a prolongation of the And with continuous infusion in critically ill patients
t1⁄2β (3.9 vs 2.4h in controls).[236] needing intensive care the t1⁄2β was 2.5 to 3
The results of Chauvin et al.[74] suggest that sin- hours.[249-251]
gle dose sufentanil should have a similar duration
2.13.2 Effects of Liver Disease
of action in patients with cirrhosis and patients
In patients with liver dysfunction, disposition of
with healthy liver function. In patients with cirrho-
alfentanil is affected in several ways. This con-
sis with reduced plasma proteins or alkalosis, the
trasts to the reports of unaltered kinetics of fentanyl
Vd and t1⁄2β might increase.
and sufentanil in patients with cirrhosis. Ferrier et
al.[241] reported that patients with biopsy-proven
2.13 Alfentanil
liver cirrhosis had an t1⁄2β of 3.65 hours compared
2.13.1 Pharmacokinetics with 1.5 hours in controls, and plasma protein
Alfentanil is a very short acting opioid; its an- binding was significantly reduced from 88.5% in
algesic effects last 5 to 10 minutes after adminis- controls to 81.4% in patients with liver disease.
tration of a single intravenous dose. Its analgesic Similar results were demonstrated by Bower et
potency is only one-third to one-quarter of that of al.[240] The increase in the free fraction might be
fentanyl and it is less lipid soluble than fentanyl. attributable to lower AAG levels or alterations of
When compared with fentanyl and sufentanil, al- binding sites in patients with cirrhosis.[241] When
fentanil has a faster equilibration between blood the pharmacokinetic parameters were corrected for

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)
32 Tegeder et al.

protein binding, the unbound Vd and free drug related to the fentanyl family of short-acting 4-an-
clearance were decreased significantly in both ilidopiperidine derivatives, remifentanil is struc-
studies.[240,241] These changes in pharmacokinetics turally unique among currently available opioids
were observed in patients with a moderate degree because of an ester linkage that is crucial to its
of hepatic insufficiency; a more delayed elimina-
opioid activity. As an ester, remifentanil is suscep-
tion can be expected in patients with pronounced
tible to hydrolysis by blood and tissue nonspecific
hepatic failure.
In patients with liver dysfunction, alfentanil esterases, resulting in rapid metabolism to essen-
should be administered with caution. Its effects tially inactive compounds. The primary carboxylic
may not only be prolonged but also enhanced, be- acid metabolite (GI-90291) had only 1/4600 of the
cause of the increased free fraction of the drug. μ opioid agonist potency of the parent compound
in anaesthetised dogs.[254]
2.14 Remifentanil GI-90291 is eliminated primarily by renal ex-
2.14.1 Pharmacokinetics
cretion, with a t1⁄2β of approximately 1.5 to 2
Remifentanil is a synthetic μ opioid agonist[252] hours[67,255] and steady-state plasma concentra-
with an analgesic potency 20 to 30 times higher tions about 12 times higher than those measured for
than that of alfentanil.[237,253] Although chemically remifentanil.[67] In addition to ester hydrolysis as

Table III. Recommendations for prescribing opioids in patients with hepatic insufficiency
Drug Problem in liver disease Suggested action
Alfentanil Reduced protein binding. CL decreased by ~50% and t1⁄2β Reduce dose in patients with severe liver
prolonged dysfunction
Buprenorphine Insufficient data
Codeine O-Demethylation to form morphine, the active compound, may Do not use for analgesia
be reduced
Dextropropoxyphene Increased F of oral dextropropoxyphene. Several reports of Avoid in patients with liver disease
hepatotoxicity due to dextropropoxyphene
Dihydrocodeine Insufficient data
Fentanyl Pharmacokinetics of a single intravenous dose unaltered Normal single dose can be used. With
continuous administration, recovery time after
termination of the infusion may be prolonged
Methadone Prolongation of t1⁄2β and increase of Vd in patients with severe Normal dosage can be used in mild to moderate
hepatic dysfunction. Chronic alcohol abuse may increase liver disease. In severe liver dysfunction
methadone metabolism accumulation may occur
Morphine Reduced hepatic glucuronidation leads to an increase of oral F, Use with care in patients with severe liver
decrease of CL and prolongation of t1⁄2β cirrhosis and reduce oral dosage
Pentazocine 2- to 3-fold increase in F. CL reduced by ~50% and t1⁄2β doubled Use lower dosage or an alternative analgesic
Pethidine Reduced hepatic CL of pethidine and norpethidine resulting in Reduce oral dosage and avoid regular use
(meperidine) an increased F of oral pethidine and prolongation of t1⁄2β of
pethidine and norpethidine. Accumulation of norpethidine may
occur. Can cause seizures
Remifentanil Pharmacokinetics unaltered Normal dose can be used
Sufentanil Pharmacokinetics unaltered. Reduced protein binding with Normal dose can be used. Use with care when
alkalosis associated with an increase of Vd and t1⁄2β plasma pH is elevated
Tilidine Generation of the active compound nortilidine reduced (tilidine is Do not use as an analgesic in severe liver
a prodrug). Probably no analgesic effects cirrhosis
Tramadol Reduced generation of the main metabolite O-demethyl-tramadol Prefer alternative analgesic until analgesic
that appears to be responsible for some of the analgesic actions. activity of tramadol in patients with liver disease
t1⁄2β of tramadol and O-demethyl-tramadol approximately doubled has been confirmed
CL = clearance; F = oral bioavailability; t1⁄2β = terminal elimination half-life; Vd = volume of distribution.

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)
Opioids in Liver Disease 33

the major metabolic pathway, a minor proportion tramadol or tilidine. However, there is sufficient
of remifentanil undergoes N-dealkylation.[68] evidence that the hepatic metabolism of morphine,
Remifentanil distributes rapidly into tissues, as pethidine, pentazocine and alfentanil is signifi-
indicated by the very short time required for equil- cantly reduced in patients with cirrhosis, leading to
ibration between blood and effect site (t1⁄2,ke0 0.75 changes in drug disposition that should be consid-
to 1.3 min).[69,70,237] Its Vd is 25 to 40L, similar to ered in dosage regimens. Table III summarises the
that of alfentanil (30 to 60 L/kg).[237,68-70] Its clear- findings of the research to date and attempts to give
ance is about an order of magnitude higher than some practical advice. The choice of the most ap-
that of alfentanil (about 180 vs about 18 L/h).[67,237] propriate drug and dose, however, depends on the
Because of its high clearance and small Vd, individual situation of the patient and the kind of
remifentanil has a very short t1⁄2β of 10 to 20 min- pain to be treated.
utes.[67,256] As a result, recovery from the effects of
remifentanil is rapid and almost independent of References
dose or duration of infusion.[68] 1. Laidlaw J, Read AE, Sherlock S. Morphine tolerance in hepatic
cirrhosis. Gastroenterology 1961; 40: 389-96
2.14.2 Effects of Liver Disease 2. Fraser CL, Arieff AI. Hepatic encephalopathy. N Engl J Med
1985; 313 (14): 865-73
Data on the influence of liver disease on the 3. Holford NH, Sheiner LB. Pharmacokinetic and pharmacody-
pharmacokinetics of remifentanil are limited. In namic modeling in vivo. CRC Crit Rev Bioeng 1981; 5 (4):
patients awaiting liver transplantation, Dershwitz 273-322
4. Huet PM, Villeneuve JP. Determinants of drug disposition in
et al.[257] showed that the disposition of remifen- patients with cirrhosis. Hepatology 1983; 3 (6): 913-8
tanil was unaltered. Hepatic function in these pa- 5. Pessayre D, Lebrec D, Descatoire V, et al. Mechanism for re-
duced drug clearance in patients with cirrhosis. Gastroenter-
tients was severely compromised, as indicated by ology 1978; 74 (3): 566-71
hypoalbuminemia, prolongation of prothrombin 6. Klotz U, Avant GR, Hoyumpa A, et al. The effects of age and
time and portal hypertension. There were also no liver disease on the disposition and elimination of diazepam
in adult man. J Clin Invest 1975; 55 (2): 347-59
differences in the disposition of the main metabo- 7. Roberts RK, Wilkinson GR, Branch RA, et al. Effect of age and
lite GI-90291 between the control group and pa- parenchymal liver disease on the disposition and elimination
tients with liver disease. However, in the hepatic of chlordiazepoxide (librium). Gastroenterology 1978; 75
(3): 479-85
dysfunction group the remifentanil concentration 8. Neal EA, Meffin PJ, Gregory PB, et al. Enhanced bioavailabil-
needed to depress CO2-stimulated ventilation by ity and decreased clearance of analgesics in patients with cir-
rhosis. Gastroenterology 1979; 77 (1): 96-102
50% was significantly reduced (1.56 vs 2.52 μg/L), 9. Ghabrial H, Desmond PV, Watson KJ, et al. The effects of age
suggesting that the patients with liver disease were and chronic liver disease on the elimination of temazepam.
more sensitive to the opioid effects of remifentanil. Eur J Clin Pharmacol 1986; 30 (1): 93-7
10. Kraus JW, Desmond PV, Marshall JP, et al. Effects of aging and
Because the drug is cleared so rapidly, regardless liver disease on disposition of lorazepam. Clin Pharmacol
of the presence of liver disease, the apparent dif- Ther 1978; 24 (4): 411-9
ference in sensitivity to the ventilatory depres- 11. Shull HJ, Wilkinson GR, Johnson R, et al. Normal disposition
of oxazepam in acute viral hepatitis and cirrhosis. Ann Intern
sant effect of remifentanil is unlikely to cause Med 1976; 84 (4): 420-5
meaningful changes in the safety of the drug in the 12. Pacifici GM, Viani A, Franchi M, et al. Conjugation pathways
in liver disease. Br J Clin Pharmacol 1990; 30 (3): 427-35
recovery period. At present, dose adjustment of 13. Macdonald JI, Wallace SM, Mahachai V, et al. Both phenolic
remifentanil appears to be unnecessary in pa- and acyl glucuronidation pathways of diflunisal are impaired
tients with hepatic dysfunction. in liver cirrhosis. Eur J Clin Pharmacol 1992; 42 (5): 471-4
14. Meuldermans WE, Hurkmans RM, Heykants JJ. Plasma protein
binding and distribution of fentanyl, sufentanil, alfentanil and
lofentanil in blood. Arch Int Pharmacodyn Ther 1982; 257
3. Conclusion (1): 4-19
The relationship between liver impairment and 15. Bovill JG, Sebel PS, Blackburn CL, et al. The pharmacokinetics
the use of opioid analgesics remains a complex of alfentanil (R39209): a new opioid analgesic. Anesthesiol-
ogy 1982; 57 (6): 439-43
issue, partly because of the paucity of research for 16. Hug Jr CC, Burm AG, de Lange S. Alfentanil pharmacokinetics
some opioid analgesics such as dihydrocodeine, in cardiac surgical patients. Anesth Analg 1994; 78 (2): 231-9

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)
34 Tegeder et al.

17. Davis PJ, Cook DR. Clinical pharmacokinetics of the newer 37. Mather LE. Clinical pharmacokinetics of fentanyl and its newer
intravenous anaesthetic agents. Clin Pharmacokinet 1986; 11 derivatives. Clin Pharmacokinet 1983; 8 (5): 422-46
(1): 18-35 38. McClain DA, Hug Jr CC. Intravenous fentanyl kinetics. Clin
18. Goresky GV, Koren G, Sabourin MA, et al. The pharmacoki- Pharmacol Ther 1980; 28 (1): 106-14
netics of alfentanil in children. Anesthesiology 1987; 67 (5): 39. Hudson RJ, Thomson IR, Cannon JE, et al. Pharmacokinetics
654-9 of fentanyl in patients undergoing abdominal aortic surgery.
19. Kuhlman Jr JJ, Lalani S, Magluilo Jr J, et al. Human pharma- Anesthesiology 1986; 64 (3): 334-8
cokinetics of intravenous, sublingual, and buccal buprenorph- 40. Berkowitz BA. The relationship of pharmacokinetics to phar-
ine. J Anal Toxicol 1996; 20 (6): 369-78 macological activity: morphine, methadone and naloxone.
20. Bullingham RE, McQuay HJ, Porter EJ, et al. Sublingual Clin Pharmacokinet 1976; 1 (3): 219-30
buprenorphine used postoperatively: ten hour plasma drug 41. Nilsson MI, Meresaar U, Anggard E. Clinical pharmacokinetics
concentration analysis. Br J Clin Pharmacol 1982; 13 (5): of methadone. Acta Anaesthesiol Scand Suppl 1982; 74: 66-9
665-73 42. Meresaar U, Nilsson MI, Holmstrand J, et al. Single dose phar-
21. Mendelson J, Upton RA, Everhart ET, et al. Bioavailability macokinetics and bioavailability of methadone in man studied
of sublingual buprenorphine. J Clin Pharmacol 1997; 37 with a stable isotope method. Eur J Clin Pharmacol 1981; 20
(1): 31-7 (6): 473-8
22. Hand CW, Sear JW, Uppington J, et al. Buprenorphine disposi- 43. Inturrisi CE, Colburn WA, Kaiko RF, et al. Pharmacokinetics
tion in patients with renal impairment: single and continuous and pharmacodynamics of methadone in patients with chronic
dosing, with special reference to metabolites. Br J Anaesth pain. Clin Pharmacol Ther 1987; 41 (4): 392-401
1990; 64 (3): 276-82 44. Gourlay GK, Wilson PR, Glynn CJ. Pharmacodynamics and
23. Heel RC, Brogden RN, Speight TM, et al. Buprenorphine: a pharmacokinetics of methadone during the perioperative pe-
review of its pharmacological properties and therapeutic effi- riod. Anesthesiology 1982; 57 (6): 458-67
cacy. Drugs 1979; 17 (2): 81-110 45. Sawe J. High-dose morphine and methadone in cancer patients:
24. Bullingham RE, McQuay HJ, Moore A, et al. Buprenorphine clinical pharmacokinetic considerations of oral treatment.
kinetics. Clin Pharmacol Ther 1980; 28 (5): 667-72 Clin Pharmacokinet 1986; 11 (2): 87-106
25. Hull JH, Findlay JW, Rogers JF, et al. An evaluation of the 46. Aitkenhead AR, Vater M, Achola K, et al. Pharmacokinetics of
effects of smoking on codeine pharmacokinetics and bioavail- single-dose i.v. morphine in normal volunteers and patients
ability in normal human volunteers. Drug Intell Clin Pharm
with end-stage renal failure. Br J Anaesth 1984; 56 (8): 813-9
1982; 16 (11): 849-54
47. Lotsch J, Weiss M, Kobal G, et al. Pharmacokinetics of mor-
26. Findlay JW, Butz RF, Welch RM. Codeine kinetics as deter-
phine-6-glucuronide and its formation from morphine after
mined by radioimmunoassay. Clin Pharmacol Ther 1977; 22
intravenous administration. Clin Pharmacol Ther 1998; 63
(4): 439-46
(6): 629-39
27. Band CJ, Band PR, Deschamps M, et al. Human pharmacoki-
48. Osborne R, Joel S, Grebenik K, et al. The pharmacokinetics of
netic study of immediate-release (codeine phosphate) and
morphine and morphine glucuronides in kidney failure. Clin
sustained-release (codeine Contin) codeine. J Clin Pharmacol
1994; 34 (9): 938-43 Pharmacol Ther 1993; 54 (2): 158-67
49. Mazoit JX, Sandouk P, Zetlaoui P, et al. Pharmacokinetics of
28. Gibson TP. Pharmacokinetics, efficacy, and safety of analgesia
with a focus on tramadol HCl. Am J Med 1996; 101 (1A): unchanged morphine in normal and cirrhotic subjects. Anesth
47S-53S Analg 1987; 66 (4): 293-8
29. Bodd E, Beylich KM, Christophersen AS, et al. Oral adminis- 50. Mather LE. Opioid pharmacokinetics in relation to their effects.
tration of codeine in the presence of ethanol. a pharmacoki- Anaesth Intensive Care 1987; 15 (1): 15-22
netic study in man. Pharmacol Toxicol 1987; 61 (5): 297-300 51. Portenoy RK, Foley KM, Stulman J, et al. Plasma morphine and
30. Lintz W, Barth H, Becker R, et al. Pharmacokinetics of tramadol morphine-6-glucuronide during chronic morphine therapy for
and bioavailability of enteral tramadol formulations: 2nd cancer pain. Plasma profiles, steady-state concentrations and
communication. Drops with ethanol. Arzneimittel Forschung the consequences of renal failure. Pain 1991; 47 (1): 13-9
1998; 48 (5): 436-45 52. Hasselstrom J, Alexander N, Bringel C, et al. Single-dose and
31. Gram LF, Schou J, Way WL, et al. d-Propoxyphene kinetics steady-state kinetics of morphine and its metabolites in cancer
after single oral and intravenous doses in man. Clin Phar- patients: a comparison of two oral formulations. Eur J Clin
macol Ther 1979; 26 (4): 473-82 Pharmacol 1991; 40 (6): 585-91
32. Inturrisi CE, Colburn WA, Verebey K, et al. Propoxyphene and 53. Khojasteh A, Evans W, Reynolds RD, et al. Controlled-release
norpropoxyphene kinetics after single and repeated doses of oral morphine sulfate in the treatment of cancer pain with
propoxyphene. Clin Pharmacol Ther 1982; 31 (2): 157-67 pharmacokinetic correlation. J Clin Oncol 1987; 5 (6): 956-61
33. Brosen K, Gram LF, Schou J, et al. Dextropropoxyphene kinet- 54. Hoskin PJ, Hanks GW, Aherne GW, et al. The bioavailability
ics after single and repeated oral doses in man. Eur J Clin and pharmacokinetics of morphine after intravenous, oral and
Pharmacol 1985; 29 (1): 79-84 buccal administration in healthy volunteers. Br J Clin Phar-
34. Rowell FJ, Seymour RA, Rawlins MD. Pharmacokinetics of macol 1989; 27 (4): 499-505
intravenous and oral dihydrocodeine and its acid metabolites. 55. Lotsch J, Stockmann A, Kobal G, et al. Pharmacokinetics of
Eur J Clin Pharmacol 1983; 25 (3): 419-24 morphine and its glucuronides after intravenous infusion of
35. Haberer JP, Schoeffler P, Couderc E, et al. Fentanyl pharmaco- morphine and morphine-6-glucuronide in healthy volunteers.
kinetics in anaesthetized patients with cirrhosis. Br J Anaesth Clin Pharmacol Ther 1996; 60 (3): 316-25
1982; 54 (12): 1267-70 56. Hanna MH, Peat SJ, Knibb AA, et al. Disposition of morphine-
36. Bentley JB, Borel JD, Nenad Jr RE, et al. Age and fentanyl 6-glucuronide and morphine in healthy volunteers. Br J An-
pharmacokinetics. Anesth Analg 1982; 61 (12): 968-71 aesth 1991; 66 (1): 103-7

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)
Opioids in Liver Disease 35

57. Verbeeck RK, Branch RA, Wilkinson GR. Meperidine disposi- 76. Lintz W, Barth H, Osterloh G, et al. Bioavailability of enteral
tion in man. influence of urinary pH and route of administra- tramadol formulations: 1st communication. Capsules.
tion. Clin Pharmacol Ther 1981; 30 (5): 619-28 Arzneimittel Forschung 1986; 36 (8): 1278-83
58. Szeto HH, Inturrisi CE, Houde R, et al. Accumulation of 77. Osborne R, Joel S, Trew D, et al. Morphine and metabolite
normeperidine, an active metabolite of meperidine, in pa- behavior after different routes of morphine administration:
tients with renal failure of cancer. Ann Intern Med 1977; 86 demonstration of the importance of the active metabolite mor-
(6): 738-41 phine-6-glucuronide. Clin Pharmacol Ther 1990; 47 (1): 12-9
59. Vollmer KO, Thomann P, Hengy H. Pharmacokinetics of tilid- 78. Brunk SF, Delle M. Morphine metabolism in man. Clin Phar-
ine and metabolites in man. Arzneimittel Forschung 1989; 39 macol Ther 1974; 16 (1): 51-7
(10): 1283-8 79. D’Honneur G, Gilton A, Sandouk P, et al. Plasma and cerebro-
60. Ehrnebo M, Boreus LO, Lonroth U. Bioavailability and first- spinal fluid concentrations of morphine and morphine glucu-
pass metabolism of oral pentazocine in man. Clin Pharmacol ronides after oral morphine. The influence of renal failure.
Ther 1977; 22 (6): 888-92 Anesthesiology 1994; 81 (1): 87-93
61. Hamunen K, Olkkola KT, Seppala T, et al. Pharmacokinetics 80. Wolff T, Samuelsson H, Hedner T. Morphine and morphine
and pharmacodynamics of pentazocine in children. Phar- metabolite concentrations in cerebrospinal fluid and plasma
macol Toxicol 1993; 73 (2): 120-3 in cancer pain patients after slow-release oral morphine ad-
62. Ehrnebo M, Boreus LO, Lonroth U. Single-dose kinetics and ministration. Pain 1995; 62 (2): 147-54
bioavailability of pentazocine. Acta Anaesthesiol Scand 81. Wolff T, Samuelsson H, Hedner T. Concentrations of morphine
1982; 74 Suppl.: 70-1 and morphine metabolites in CSF and plasma during contin-
63. Herman RJ, McAllister CB, Branch RA, et al. Effects of age on uous subcutaneous morphine administration in cancer pain
meperidine disposition. Clin Pharmacol Ther 1985; 37 (1): patients. Pain 1996; 68 (2-3): 209-16
19-24 82. Stanski DR, Greenblatt DJ, Lowenstein E. Kinetics of intrave-
64. Koska AJd, Kramer WG, Romagnoli A, et al. Pharmacokinetics nous and intramuscular morphine. Clin Pharmacol Ther
of high-dose meperidine in surgical patients. Anesth Analg 1978; 24 (1): 52-9
1981; 60 (1): 8-11 83. Crotty B, Watson KJ, Desmond PV, et al. Hepatic extraction of
65. Stambaugh JE, Wainer IW, Sanstead JK, et al. The clinical phar- morphine is impaired in cirrhosis. Eur J Clin Pharmacol 1989;
macology of meperidine: comparison of routes of administra- 36 (5): 501-6
tion. J Clin Pharmacol 1976; 16 (5-6): 245-56 84. Hasselstrom J, Sawe J. Morphine pharmacokinetics and meta-
66. Mather LE, Tucker GT, Pflug AE, et al. Meperidine kinetics in bolism in humans. Enterohepatic cycling and relative contri-
man: intravenous injection in surgical patients and volun- bution of metabolites to active opioid concentrations. Clin
teers. Clin Pharmacol Ther 1975; 17 (1): 21-30 Pharmacokinet 1993; 24 (4): 344-54
67. Westmoreland CL, Hoke JF, Sebel PS, et al. Pharmacokinetics 85. Sawe J, Odar-Cederlof I. Kinetics of morphine in patients with
of remifentanil (GI87084B) and its major metabolite renal failure. Eur J Clin Pharmacol 1987; 32 (4): 377-82
(GI90291) in patients undergoing elective inpatient surgery. 86. Mazoit JX, Sandouk P, Scherrmann JM, et al. Extrahepatic me-
Anesthesiology 1993; 79 (5): 893-903 tabolism of morphine occurs in humans. Clin Pharmacol Ther
68. Egan TD, Lemmens HJ, Fiset P, et al. The pharmacokinetics of 1990; 48 (6): 613-8
the new short-acting opioid remifentanil (GI87084B) in 87. Richardson PD, Withrington PG. Liver blood flow: I. Intrinsic
healthy adult male volunteers. Anesthesiology 1993; 79 (5): and nervous control of liver blood flow. Gastroenterology
881-92 1981; 81 (1): 159-73
69. Glass PS, Hardman D, Kamiyama Y, et al. Preliminary pharma- 88. Westerling D, Persson C, Hoglund P. Plasma concentrations of
cokinetics and pharmacodynamics of an ultra-short-acting morphine, morphine-3-glucuronide, and morphine-6-glucu-
opioid, remifentanil (GI87084B). Anesth Analg 1993; 77 (5): ronide after intravenous and oral administration to healthy
1031-40 volunteers. relationship to nonanalgesic actions. Ther Drug
70. Minto CF, Schnider TW, Egan TD, et al. Influence of age and Monit 1995; 17 (3): 287-301
gender on the pharmacokinetics and pharmacodynamics of 89. Glare PA, Walsh TD. Clinical pharmacokinetics of morphine.
remifentanil: I. Model development. Anesthesiology 1997; Ther Drug Monit 1991; 13 (1): 1-23
86 (1): 10-23 90. Gong QL, Hedner J, Bjorkman R, et al. Morphine-3-glucuron-
71. Bovill JG, Sebel PS, Blackburn CL, et al. The pharmacokinetics ide may functionally antagonize morphine-6-glucuronide in-
of sufentanil in surgical patients. Anesthesiology 1984; 61 duced antinociception and ventilatory depression in the rat
(5): 502-6 [see comments]. Pain 1992; 48 (2): 249-55
72. Lehmann KA, Sipakis K, Gasparini R, et al. Pharmacokinetics 91. Smith MT, Watt JA, Cramond T. Morphine-3-glucuronide: a
of sufentanil in general surgical patients under different con- potent antagonist of morphine analgesia. Life Sci 1990; 47
ditions of anaesthesia. Acta Anaesthesiol Scand 1993; 37 (2): (6): 579-85
176-80 92. Faura CC, Olaso MJ, Garcia Cabanes C, et al. Lack of mor-
73. Gepts E, Shafer SL, Camu F, et al. Linearity of pharmacokinet- phine-6-glucuronide antinociception after morphine treat-
ics and model estimation of sufentanil. Anesthesiology 1995; ment. Is morphine-3-glucuronide involved? Pain 1996; 65
83 (6): 1194-204 (1): 25-30
74. Chauvin M, Ferrier C, Haberer JP, et al. Sufentanil pharma- 93. Smith GD, Smith MT. Morphine-3-glucuronide. evidence to
cokinetics in patients with cirrhosis. Anesth Analg 1989; support its putative role in the development of tolerance to
68 (1): 1-4 the antinociceptive effects of morphine in the rat. Pain 1995;
75. Sear JW. Sufentanil disposition in patients undergoing renal 62 (1): 51-60
transplantation: influence of choice of kinetic model. Br J 94. Makman MH, Dobrenis K, Surratt CK. Properties of mu 3 opi-
Anaesth 1989; 63 (1): 60-7 ate alkaloid receptors in macrophages, astrocytes, and HL-60

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)
36 Tegeder et al.

human promyelocytic leukemia cells. Adv Exp Med Biol 113. Tiseo PJ, Thaler HT, Lapin J, et al. Morphine-6-glucuronide
1998; 437: 137-48 concentrations and opioid-related side effects. a survey in
95. Loser SV, Meyer J, Freudenthaler S, et al. Morphine-6-O-beta- cancer patients. Pain 1995; 61 (1): 47-54
D-glucuronide but not morphine-3-O-beta-D-glucuronide 114. Hasselstrom J, Berg U, Lofgren A, et al. Long lasting respiratory
binds to mu-, delta- and kappa-specific opioid binding sites depression induced by morphine-6-glucuronide? Br J Clin
in cerebral membranes. Naunyn Schmiedebergs Arch Phar- Pharmacol 1989; 27 (4): 515-8
macol 1996; 354 (2): 192-7 115. Osborne R, Thompson P, Joel S, et al. The analgesic activity
96. Makman MH, Dvorkin B, Stefano GB. Murine macrophage cell of morphine-6-glucuronide. Br J Clin Pharmacol 1992; 34
lines contain mu 3-opiate receptors. Eur J Pharmacol 1995; (2): 130-8
273 (3): R5-6 116. Thompson PI, Joel SP, John L, et al. Respiratory depression
97. Bartlett SE, Smith MT. The apparent affinity of morphine-3- following morphine and morphine-6-glucuronide in normal
glucuronide at mu1-opioid receptors results from morphine subjects. Br J Clin Pharmacol 1995; 40 (2): 145-52
contamination. demonstration using HPLC and radioligand 117. Lotsch J, Kobal G, Stockmann A, et al. Lack of analgesic activ-
binding. Life Sci 1995; 57 (6): 609-15 ity of morphine-6-glucuronide after short-term intravenous
98. Lipkowski AW, Carr DB, Langlade A, et al. Morphine-3-glucu- administration in healthy volunteers. Anesthesiology 1997;
ronide: silent regulator of morphine actions. Life Sci 1994; 55 87 (6): 1348-58
(2): 149-54 118. Bickel U, Schumacher OP, Kang YS, et al. Poor permeability
99. Gardmark M, Karlsson MO, Jonsson F, et al. Morphine-3- of morphine 3-glucuronide and morphine 6-glucuronide
glucuronide has a minor effect on morphine antinocicep- through the blood-brain barrier in the rat. J Pharmacol Exp
tion: pharmacodynamic modeling. J Pharm Sci 1998; 87 Ther 1996; 278 (1): 107-13
(7): 813-20 119. Carrupt PA, Testa B, Bechalany A, et al. Morphine 6-glucuron-
100. Ouellet DM, Pollack GM. Effect of prior morphine-3-glucuron- ide and morphine 3-glucuronide as molecular chameleons
ide exposure on morphine disposition and antinociception. with unexpected lipophilicity. J Med Chem 1991; 34 (4):
Biochem Pharmacol 1997; 53 (10): 1451-7 1272-5
101. Bian JT, Bhargava HN. Effects of morphine-3-glucuronide on 120. Hanks GW, Hoskin PJ, Aherne GW, et al. Explanation for po-
the antinociceptive activity of peptide and nonpeptide opioid tency of repeated oral doses of morphine? Lancet 1987; II
receptor agonists in mice. Peptides 1996; 17 (8): 1415-9 (8561): 723-5
102. Suzuki N, Kalso E, Rosenberg PH. Intrathecal morphine-3-glu- 121. Huwyler J, Drewe J, Klusemann C, et al. Evidence for P-glyco-
curonide does not antagonize spinal antinociception by mor- protein-modulated penetration of morphine-6-glucuronide
phine or morphine-6-glucuronide in rats. Eur J Pharmacol into brain capillary endothelium. Br J Pharmacol 1996; 118
1993; 249 (2): 247-50 (8): 1879-85
103. Hewett K, Dickenson AH, McQuay HJ. Lack of effect of mor- 122. Huwyler J, Drewe J, Gutmann H, et al. Modulation of mor-
phine-3-glucuronide on the spinal antinociceptive actions of phine-6-glucuronide penetration into the brain by P-glycopro-
morphine in the rat. an electrophysiological study. Pain 1993; tein. Int J Clin Pharmacol Ther 1998; 36 (2): 69-70
53 (1): 59-63 123. Letrent SP, Pollack GM, Brouwer KR, et al. Effect of
104. Hanna MH, Peat SJ, Woodham M, et al. Analgesic efficacy and GF120918, a potent P-glycoprotein inhibitor, on morphine
CSF pharmacokinetics of intrathecal morphine-6-glucuron- pharmacokinetics and pharmacodynamics in the rat. Pharm
ide. comparison with morphine. Br J Anaesth 1990; 64 (5): Res 1998; 15 (4): 599-605
547-50 124. Patwardhan RV, Johnson RF, Hoyumpa Jr A, et al. Normal me-
105. Boerner U. The metabolism of morphine and heroin in man. tabolism of morphine in cirrhosis. Gastroenterology 1981; 81
Drug Metab Rev 1975; 4 (1): 39-73 (6): 1006-11
106. Krzanowska EK, Rossi GC, Pasternak GW, et al. Potency ratios 125. Hasselstrom J, Eriksson S, Persson A, et al. The metabolism and
of morphine and morphine-6beta-glucuronide analgesia elic- bioavailability of morphine in patients with severe liver cir-
ited from the periaqueductal gray, locus coeruleus or rostral rhosis. Br J Clin Pharmacol 1990; 29 (3): 289-97
ventromedial medulla of rats. Brain Res 1998; 799 (2): 329-33 126. Watson KJR, Ghabrial H, Breen KJ, et al. Morphine disposition
107. Paul D, Standifer KM, Inturrisi CE, et al. Pharmacological char- in cirrhosis [abstract]. Gastroenterology 1986; 88: 1704
acterization of morphine-6 beta-glucuronide, a very potent 127. Meech R, Mackenzie PI. Structure and function of uridine
morphine metabolite. J Pharmacol Exp Ther 1989; 251 (2): diphosphate glucuronosyltransferases. Clin Exp Pharmacol
477-83 Physiol 1997; 24 (12): 907-15
108. Pasternak GW, Bodnar RJ, Clark JA, et al. Morphine-6-glucu- 128. Zakim D. The role of membrane lipids in the regulation of mem-
ronide, a potent mu agonist. Life Sci 1987; 41 (26): 2845-9 brane-bound enzymes. Prog Liver Dis 1986; 8: 65-80
109. Goucke CR, Hackett LP, Ilett KF. Concentrations of morphine, 129. Desmond PV, Smyth FE, Mashford ML. Release of latent
morphine-6-glucuronide and morphine-3-glucuronide in se- glucuronosyltransferase activity contributes to the sparing of
rum and cerebrospinal fluid following morphine administra- glucuronidation in experimental liver injuries. J Gastroenterol
tion to patients with morphine-resistant pain. Pain 1994; 56 Hepatol 1994; 9 (4): 350-4
(2): 145-9 130. Debinski HS, Mackenzie PI, Lee CS, et al. UDP glucurono-
110. Hasselstrom J, Svensson JO, Sawe J, et al. Disposition and an- syltransferase in the cirrhotic rat liver. J Gastroenterol Hepatol
algesic effects of systemic morphine, morphine-6-glucuron- 1996; 11 (4): 373-9
ide and normorphine in rat. Pharmacol Toxicol 1996; 79 (1): 131. Aitio A. Effect of chrysene and carbon tetrachloride adminis-
40-6 tration on rat hepatic microsomal monooxygenase and
111. Glare PA, Walsh TD, Pippenger CE. Normorphine, a neurotoxic UDPglucuronosyltransferase activity. FEBS Lett 1974; 42
metabolite? Lancet 1990; 335 (8691): 725-6 (1): 46-9
112. Hanks GW, Hoskin PJ, Aherne GW, et al. Enterohepatic circu- 132. Boyer TD, Zakim D. The effect of choline deficiency on the
lation of morphine. Lancet 1988; I (8583): 469 activity of a phosphatidylcholine-requiring enzyme activity

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)
Opioids in Liver Disease 37

and properties of UDP-glucuronyltransferase in choline-de- 150. Novick DM, Kreek MJ, Arns PA, et al. Effect of severe alco-
ficient rats. Biochem Biophys Res Commun 1983; 114 (1): holic liver disease on the disposition of methadone in main-
418-24 tenance patients. Alcohol Clin Exp Res 1985; 9 (4): 349-54
133. Otani G, Abou-El-Makarem MM, Bock KW. UDP-glucuron- 151. Orrego H, Israel Y, Blendis LM. Alcoholic liver disease: in-
yltransferase in perfused rat liver and in microsomes: III. Ef- formation in search of knowledge? Hepatology 1981; 1 (3):
fects of galactosamine and carbon tetrachloride on the 267-83
glucuronidation of 1-naphthol and bilirubin. Biochem Phar- 152. Borowsky SA, Lieber CS. Interaction of methadone and ethanol
macol 1976; 25 (11): 1293-7 metabolism. J Pharmacol Exp Ther 1978; 207 (1): 123-9
134. Pacifici GM, Bencini C, Rane A. Presystemic glucuronidation 153. Donnelly B, Balkon J, Lasher C, et al. Evaluation of the
of morphine in humans and rhesus monkeys. subcellular dis- methadone-alcohol interaction: I. Alterations of plasma con-
tribution of the UDP-glucuronyltransferase in the liver and centration kinetics. J Anal Toxicol 1983; 7 (5): 246-8
intestine. Xenobiotica 1986; 16 (2): 123-8 154. Iribarne C, Berthou F, Baird S, et al. Involvement of cyto-
135. Dechelotte P, Sabouraud A, Sandouk P, et al. Uptake, 3-, and chrome P450 3A4 enzyme in the N-demethylation of
6-glucuronidation of morphine in isolated cells from stom- methadone in human liver microsomes. Chem Res Toxicol
ach, intestine, colon, and liver of the guinea pig. Drug Metab 1996; 9 (2): 365-73
Dispos 1993; 21 (1): 13-7 155. George J, Liddle C, Murray M, et al. Pre-translational regula-
136. Pacifici GM, Rane A. Renal glucuronidation of morphine in the tion of cytochrome P450 genes is responsible for disease-spe-
human foetus. Acta Pharmacol Toxicol 1982; 50 (2): 155-60 cific changes of individual P450 enzymes among patients
137. Vree TB, Hekster YA, Anderson PG. Contribution of the human with cirrhosis. Biochem Pharmacol 1995; 49 (7): 873-81
kidney to the metabolic clearance of drugs. Ann Phar- 156. Mather LE, Tucker GT. Systemic availability of orally admin-
macother 1992; 26 (11): 1421-8 istered meperidine. Clin Pharmacol Ther 1976; 20 (5): 535-40
138. Wahlstrom A, Lenhammar L, Ask B, et al. Tricyclic antidepres- 157. Julius HC, Levine HL, Williams WD. Meperidine binding to
sants inhibit opioid receptor binding in human brain and he- isolated alpha 1-acid glycoprotein and albumin. Drug Intell
patic morphine glucuronidation. Pharmacol Toxicol 1994; 75 Clin Pharm 1989; 23 (7-8): 568-72
(1): 23-7 158. Callaghan R, Desmond PV, Paull P, et al. Hepatic enzyme ac-
139. Olsen GD, Bennett WM, Porter GA. Morphine and phenytoin tivity is the major factor determining elimination rate of high-
binding to plasma proteins in renal and hepatic failure. Clin clearance drugs in cirrhosis. Hepatology 1993; 18 (1): 54-60
Pharmacol Ther 1975; 17 (6): 677-84 159. Pond SM, Tong T, Benowitz NL, et al. Presystemic metabolism
of meperidine to normeperidine in normal and cirrhotic sub-
140. Shimoyama N, Shimoyama M, Elliott KJ, et al. d-Methadone
jects. Clin Pharmacol Ther 1981; 30 (2): 183-8
is antinociceptive in the rat formalin test. J Pharmacol Exp
Ther 1997; 283 (2): 648-52 160. Klotz U, McHorse TS, Wilkinson GR, et al. The effect of cir-
rhosis on the disposition and elimination of meperidine in
141. Verebely K, Volavka J, Mule S, et al. Methadone in man. phar-
man. Clin Pharmacol Ther 1974; 16 (4): 667-75
macokinetic and excretion studies in acute and chronic treat-
161. McHorse TS, Klotz U, Wilkinson G, et al. Impaired elimination
ment. Clin Pharmacol Ther 1975; 18 (2): 180-90
of meperidine in patients with liver disease. Trans Assoc Am
142. Anggard E, Nilsson MI, Holmstrand J, et al. Pharmacokinetics Physicians 1974; 87: 281-7
of methadone during maintenance therapy. pulse labeling
162. Danziger LH, Martin SJ, Blum RA. Central nervous system
with deuterated methadone in the steady state. Eur J Clin
toxicity associated with meperidine use in hepatic disease.
Pharmacol 1979; 16 (1): 53-7 Pharmacotherapy 1994; 14 (2): 235-8
143. Kreek MJ, Hachey DL, Klein PD. Stereoselective disposition 163. Giacomini KM, Gibson TP, Levy G. Plasma protein binding of
of methadone in man. Life Sci 1979; 24 (10): 925-32 d-propoxyphene in normal subjects and anephric patients. J
144. Kreek MJ, Kalisman M, Irwin M, et al. Biliary secretion of Clin Pharmacol 1978; 18 (2-3): 106-9
methadone and methadone metabolites in man. Res Commun 164. Jusko WJ, Gretch M. Plasma and tissue protein binding of drugs
Chem Pathol Pharmacol 1980; 29 (1): 67-78 in pharmacokinetics. Drug Metab Rev 1976; 5 (1): 43-140
145. Anggard E, Gunne LM, Homstrand J, et al. Disposition of 165. Lund-Jacobsen H. Cardio-respiratory toxicity of propoxyphene
methadone in methadone maintenance. Clin Pharmacol Ther and norpropoxyphene in conscious rabbits. Acta Pharmacol
1975; 17 (3): 258-66 Toxicol 1978; 42 (3): 171-8
146. Nilsson MI, Anggard E, Holmstrand J, et al. Pharmacokinetics 166. Crome P, Gain R, Ghurye R, et al. Pharmacokinetics of dex-
of methadone during maintenance treatment. adaptive tropropoxyphene and nordextropropoxyphene in elderly
changes during the induction phase. Eur J Clin Pharmacol hospital patients after single and multiple doses of dis-
1982; 22 (4): 343-9 talgesic. Preliminary analysis of results. Hum Toxicol
147. Kreek MJ, Bencsath FA, Field FH. Effects of liver disease on 1984; 3: 41S-48S
urinary excretion of methadone and metabolites in mainte- 167. Stork CM, Redd JT, Fine K, et al. Propoxyphene-induced wide
nance patients. Quantitation by direct probe chemical ioniza- QRS complex dysrhythmia responsive to sodium bicarbon-
tion mass spectrometry. Biomed Mass Spectrom 1980; 7 (9): ate: a case report. J Toxicol Clin Toxicol 1995; 33 (2): 179-83
385-95 168. Whitcomb DC, Gilliam FRD, Starmer CF, et al. Marked QRS
148. Kreek MJ, Bencsath FA, Fanizza A, et al. Effects of liver disease complex abnormalities and sodium channel blockade by pro-
on fecal excretion of methadone and its unconjugated meta- poxyphene reversed with lidocaine. J Clin Invest 1989; 84
bolites in maintenance patients: quantitation by direct probe (5): 1629-36
chemical ionization mass spectrometry. Biomed Mass 169. Chan TY. Propoxyphene overdose in Chinese subjects. J Tox-
Spectrom 1983; 10 (10): 544-9 icol Clin Toxicol 1996; 34 (2): 251-2
149. Novick DM, Kreek MJ, Fanizza AM, et al. Methadone dispo- 170. Giacomini KM, Giacomini JC, Gibson TP, et al. Propoxyphene
sition in patients with chronic liver disease. Clin Pharmacol and norpropoxyphene plasma concentrations after oral pro-
Ther 1981; 30 (3): 353-62 poxyphene in cirrhotic patients with and without surgically

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)
38 Tegeder et al.

constructed portacaval shunt. Clin Pharmacol Ther 1980; 28 189. George J, Murray M, Byth K, et al. Differential alterations of
(3): 417-24 cytochrome P450 proteins in livers from patients with severe
171. Horsmans Y, Desager JP, Daenens C, et al. D-propoxyphene and chronic liver disease. Hepatology 1995; 21 (1): 120-8
norpropoxyphene kinetics after the oral administration of D- 190. Adedoyin A, Arns PA, Richards WO, et al. Selective effect of
propoxyphene. a new approach to liver function? J Hepatol liver disease on the activities of specific metabolizing en-
1994; 21 (3): 283-91 zymes: investigation of cytochromes P450 2C19 and 2D6.
172. Rosenberg WM, Ryley NG, Trowell JM, et al. Dextropropoxy- Clin Pharmacol Ther 1998; 64 (1): 8-17
phene induced hepatotoxicity: a report of nine cases. J 191. Fromm MF, Hofmann U, Griese EU, et al. Dihydrocodeine: a
Hepatol 1993; 19 (3): 470-4 new opioid substrate for the polymorphic CYP2D6 in hu-
173. Bassendine MF, Woodhouse KW, Bennett M, et al. Dextropro- mans. Clin Pharmacol Ther 1995; 58 (4): 374-82
poxyphene induced hepatotoxicity mimicking biliary tract 192. Wilder-Smith CH, Hufschmid E, Thormann W. The visceral and
disease. Gut 1986; 27 (4): 444-9 somatic antinociceptive effects of dihydrocodeine and its me-
174. Lee TH, Rees PJ. Hepatotoxicity of dextropropoxyphene. BMJ tabolite, dihydromorphine: a cross-over study with extensive
and quinidine-induced poor metabolizers. Br J Clin Phar-
1977; 2 (6082): 296-7
macol 1998; 45 (6): 575-81
175. Desruelles F, Chichmanian RM, Castanet J, et al. Hepatotoxic-
193. Jurna I, Komen W, Baldauf J, et al. Analgesia by dihydrocodeine
ity caused by dextropropoxyphene: two cases, one of them
is not due to formation of dihydromorphine: evidence from
with recurrence [letter; in French]. Therapie 1994; 49 (6):
nociceptive activity in rat thalamus. J Pharmacol Exp Ther
521-2 1997; 281 (3): 1164-70
176. Chen ZR, Somogyi AA, Reynolds G, et al. Disposition and 194. Ohtani M, Kotaki H, Sawada Y, et al. Comparative analysis of
metabolism of codeine after single and chronic doses in one buprenorphine- and norbuprenorphine-induced analgesic ef-
poor and seven extensive metabolisers. Br J Clin Pharmacol fects based on pharmacokinetic-pharmacodynamic modeling.
1991; 31 (4): 381-90 J Pharmacol Exp Ther 1995; 272 (2): 505-10
177. Yue QY, Svensson JO, Alm C, et al. Interindividual and inter- 195. Cone EJ, Gorodetzky CW, Yousefnejad D, et al. The metabo-
ethnic differences in the demethylation and glucuronidation lism and excretion of buprenorphine in humans. Drug Metab
of codeine. Br J Clin Pharmacol 1989; 28 (6): 629-37 Dispos 1984; 12 (5): 577-81
178. Hennies HH, Friderichs E, Schneider J. Receptor binding, an- 196. Iribarne C, Picart D, Dreano Y, et al. Involvement of cytochrome
algesic and antitussive potency of tramadol and other selected P450 3A4 in N-dealkylation of buprenorphine in human liver
opioids. Arzneimittel Forschung 1988; 38 (7): 877-80 microsomes. Life Sci 1997; 60 (22): 1953-64
179. Cleary J, Mikus G, Somogyi A, et al. The influence of pharma- 197. Kobayashi K, Yamamoto T, Chiba K, et al. Human buprenorph-
cogenetics on opioid analgesia: studies with codeine and oxy- ine N-dealkylation is catalyzed by cytochrome P450 3A4.
codone in the Sprague-Dawley/Dark Agouti rat model. J Drug Metab Dispos 1998; 26 (8): 818-21
Pharmacol Exp Ther 1994; 271 (3): 1528-34 198. Pittman K. Human metabolism of orally administered pentazo-
180. Eckhardt K, Li S, Ammon S, et al. Same incidence of adverse cine. Biochem Pharmacol 1970; 19 (5): 1833-6
drug events after codeine administration irrespective of the 199. Vaughan DP, Beckett AH, Robbie DS. The influence of smoking
genetically determined differences in morphine formation. on the intersubject variation in pentazocine elimination. Br J
Pain 1998; 76 (1-2): 27-33 Clin Pharmacol 1976; 3 (2): 279-83
181. Sindrup SH, Brosen K, Bjerring P, et al. Codeine increases pain 200. Hoskin PJ, Hanks GW. Opioid agonist-antagonist drugs in acute
thresholds to copper vapor laser stimuli in extensive but not and chronic pain states. Drugs 1991; 41 (3): 326-44
poor metabolizers of sparteine. Clin Pharmacol Ther 1990; 48 201. Pond SM, Tong T, Benowitz NL, et al. Enhanced bioavailability
(6): 686-93 of pethidine and pentazocine in patients with cirrhosis of the
182. Desmeules J, Gascon MP, Dayer P, et al. Impact of environmen- liver. Aust NZ J Med 1980; 10 (5): 515-9
tal and genetic factors on codeine analgesia. Eur J Clin Phar- 202. Pentazocine (Talwin®) [product information]. Munich: Sanofi
macol 1991; 41 (1): 23-6 Winthrop, 1995
183. Chen ZR, Irvine RJ, Bochner F, et al. Morphine formation from 203. Raffa RB, Friderichs E, Reimann W, et al. Complementary and
codeine in rat brain: a possible mechanism of codeine analge- synergistic antinociceptive interaction between the en-
antiomers of tramadol. J Pharmacol Exp Ther 1993; 267 (1):
sia. Life Sci 1990; 46 (15): 1067-74
331-40
184. Kamei J. Role of opioidergic and serotonergic mechanisms in
204. Raffa RB, Friderichs E, Reimann W, et al. Opioid and nonopioid
cough and antitussives. Pulm Pharmacol 1996; 9 (5-6): 349-56
components independently contribute to the mechanism of
185. Rogers JF, Findlay JW, Hull JH, et al. Codeine disposition in action of tramadol, an ‘atypical’opioid analgesic. J Pharmacol
smokers and nonsmokers. Clin Pharmacol Ther 1982; 32 (2): Exp Ther 1992; 260 (1): 275-85
218-27 205. Lintz W, Erlacin S, Frankus E, et al. Biotransformation of
186. Yue QY, Sawe J. Different effects of inhibitors on the O- and tramadol in man and animal [in German]. Arzneimittel
N-demethylation of codeine in human liver microsomes. Eur Forschung 1981; 31 (11): 1932-43
J Clin Pharmacol 1997; 52 (1): 41-7 206. Dayer P, Desmeules J, Collart L. Pharmacology of tramadol.
187. Persson K, Sjostrom S, Sigurdardottir I, et al. Patient-controlled Drugs 1997;Suppl. 2: 18-24
analgesia (PCA) with codeine for postoperative pain relief in 207. Lee CR, McTavish D, Sorkin EM. Tramadol: a preliminary re-
ten extensive metabolisers and one poor metaboliser of view of its pharmacodynamic and pharmacokinetic proper-
dextromethorphan. Br J Clin Pharmacol 1995; 39 (2): 182-6 ties, and therapeutic potential in acute and chronic pain states.
188. Poulsen L, Brosen K, Arendt-Nielsen L, et al. Codeine and mor- Drugs 1993; 46 (2): 313-40
phine in extensive and poor metabolizers of sparteine: phar- 208. Poulsen L, Arendt-Nielsen L, Brosen K, et al. The hypoalgesic
macokinetics, analgesic effect and side effects. Eur J Clin effect of tramadol in relation to CYP2D6. Clin Pharmacol
Pharmacol 1996; 51 (3-4): 289-95 Ther 1996; 60 (6): 636-44

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)
Opioids in Liver Disease 39

209. Tramadol (Ultram®) [product information]. Raritan (NJ): Ortho- cular, respiratory and metabolic parameters. Acta An-
McNeil Pharmaceutical, 1997 aesthesiol Belg 1979; 30 (4): 247-54
210. Vollmer KO, Poisson A. On the metabolism of ethyl-DL-trans- 230. Schwartz AE, Matteo RS, Ornstein E, et al. Pharmacokinetics
2-dimethylamino-1-phenyl-cyclohex-3-ene-trans-1-carbox of sufentanil in obese patients. Anesth Analg 1991; 73 (6):
ylate-hydrochloride (Tilidine-HCl): 2nd communication. 790-3
Studies with 14C-labelled substance on rats and dogs. 231. Meistelman C, Benhamou D, Barre J, et al. Effects of age on
Arzneimittel Forschung 1976; 26 (10): 1827-36 plasma protein binding of sufentanil. Anesthesiology 1990;
211. Data on file, report no. RR 423-00080, Gödecke 72 (3): 470-3
212. Paix A, Coleman A, Lees J, et al. Subcutaneous fentanyl and 232. Greeley WJ, de Bruijn NP, Davis DP. Sufentanil pharmacoki-
sufentanil infusion substitution for morphine intolerance in netics in pediatric cardiovascular patients. Anesth Analg
cancer pain management. Pain 1995; 63 (2): 263-9 1987; 66 (11): 1067-72
213. Hug Jr CC, Murphy MR. Tissue redistribution of fentanyl and 233. Rosow CE. Sufentanil citrate: a new opioid analgesic for use in
termination of its effects in rats. Anesthesiology 1981; 55 (4): anesthesia. Pharmacotherapy 1984; 4 (1): 11-9
369-75 234. Howie MB, Smith DF, Reilley TE, et al. Postoperative course
214. Singleton MA, Rosen JI, Fisher DM. Pharmacokinetics of fen- after sufentanil or fentanyl anesthesia for coronary artery sur-
tanyl in the elderly. Br J Anaesth 1988; 60 (6): 619-22 gery. J Cardiothorac Vasc Anesth 1991; 5 (5): 485-9
215. Santeiro ML, Christie J, Stromquist C, et al. Pharmacokinetics 235. Scholz J, Bause H, Schulz M, et al. Pharmacokinetics and ef-
of continuous infusion fentanyl in newborns. J Perinatol fects on intracranial pressure of sufentanil in head trauma
1997; 17 (2): 135-9 patients. Br J Clin Pharmacol 1994; 38 (4): 369-72
216. Katz R, Kelly HW. Pharmacokinetics of continuous infusions 236. Schwartz AE, Matteo RS, Ornstein E, et al. Pharmacokinetics
of fentanyl in critically ill children. Crit Care Med 1993; 21 of sufentanil in neurosurgical patients undergoing hyperven-
(7): 995-1000 tilation. Br J Anaesth 1989; 63 (4): 385-8
217. Hoffmann P. Continuous infusions of fentanyl and alfentanil in 237. Egan TD, Minto CF, Hermann DJ, et al. Remifentanil versus
intensive care [abstract]. Eur J Anaesthesiol Suppl. 1987; 1: alfentanil: comparative pharmacokinetics and pharmacody-
71-5 namics in healthy adult male volunteers. Anesthesiology
218. Wiesner G, Taeger K, Peter K. Serum protein binding of fen- 1996; 84 (4): 821-33
tanyl. The effect of postoperative acute phase reaction with 238. Scott JC, Cooke JE, Stanski DR. Electroencephalographic
elevated alpha 1-acid glycoprotein and methodologic prob- quantitation of opioid effect: comparative pharmacodynam-
lems in determination by equilibrium dialysis. Anaesthesist ics of fentanyl and sufentanil. Anesthesiology 1991; 74 (1):
1996; 45 (4): 323-9 34-42
219. Goromaru T, Matsuura H, Yoshimura N, et al. Identification and 239. Scott JC, Ponganis KV, Stanski DR. EEG quantitation of nar-
quantitative determination of fentanyl metabolites in patients cotic effect: the comparative pharmacodynamics of fentanyl
by gas chromatography: mass spectrometry. Anesthesiology and alfentanil. Anesthesiology 1985; 62 (3): 234-41
1984; 61 (1): 73-7 240. Bower S, Sear JW, Roy RC, et al. Effects of different hepatic
220. Murphy MR, Hug Jr CC, McClain DA. Dose-independent pathologies on disposition of alfentanil in anaesthetized pa-
pharmacokinetics of fentanyl. Anesthesiology 1983; 59 tients. Br J Anaesth 1992; 68 (5): 462-5
(6): 537-40 241. Ferrier C, Marty J, Bouffard Y, et al. Alfentanil pharmacokinet-
221. Miller RS, Peterson GM, Abbott F, et al. Plasma concentrations ics in patients with cirrhosis. Anesthesiology 1985; 62 (4):
of fentanyl with subcutaneous infusion in palliative care pa- 480-4
tients. Br J Clin Pharmacol 1995; 40 (6): 553-6 242. Bower S, Hull CJ. Comparative pharmacokinetics of fentanyl
222. Reilly CS, Wood AJ, Wood M. Variability of fentanyl pharma- and alfentanil. Br J Anaesth 1982; 54 (8): 871-7
cokinetics in man: computer predicted plasma concentrations 243. Meuldermans W, Van Peer A, Hendrickx J, et al. Alfentanil
for three intravenous dosage regimens. Anaesthesia 1985; 40 pharmacokinetics and metabolism in humans. Anesthesiol-
(9): 837-43 ogy 1988; 69 (4): 527-34
223. Bovill JG, Sebel PS. Pharmacokinetics of high-dose fentanyl: 244. Meuldermans W, Woestenborghs R, Noorduin H, et al. Protein
a study in patients undergoing cardiac surgery. Br J Anaesth binding of the analgesics alfentanil and sufentanil in maternal
1980; 52 (8): 795-801 and neonatal plasma. Eur J Clin Pharmacol 1986; 30 (2):
224. Koska AJd, Romagnoli A, Kramer WG. Effect of cardiopulmo- 217-9
nary bypass on fentanyl distribution and elimination. Clin 245. Schüttler J, Stoeckel H. Clinical pharmacokinetics of al-
Pharmacol Ther 1981; 29 (1): 100-5 fentanyl. Anaesthesist 1982; 31 (1): 10-4
225. Shafer SL, Varvel JR. Pharmacokinetics, pharmacodynamics, 246. Kharasch ED, Russell M, Mautz D, et al. The role of cyto-
and rational opioid selection. Anesthesiology 1991; 74 (1): chrome P450 3A4 in alfentanil clearance: implications for
53-63 interindividual variability in disposition and perioperative
226. Hughes MA, Glass PS, Jacobs JR. Context-sensitive half-time drug interactions. Anesthesiology 1997; 87 (1): 36-50
in multicompartment pharmacokinetic models for intrave- 247. Chauvin M, Bonnet F, Montembault C, et al. The influence of
nous anesthetic drugs. Anesthesiology 1992; 76 (3): 334-41 hepatic plasma flow on alfentanil plasma concentration pla-
227. Grant RP, Jenkins LC. Modification by preoperative beta- teaus achieved with an infusion model in humans: measure-
blockade of the renin response to infrarenal aortic cross- ment of alfentanil hepatic extraction coefficient. Anesth
clamping. Can Anaesth Soc J 1983; 30 (5): 480-6 Analg 1986; 65 (10): 999-1003
228. Jackson EK, Garrison JC. Renin and angiotensin. New York: 248. Camu F, Gepts E, Rucquoi M, et al. Pharmacokinetics of al-
McGraw-Hill, 1996 fentanil in man. Anesth Analg 1982; 61 (8): 657-61
229. Rolly G, Kay B, Cockx F. A double blind comparison of high 249. Bodenham A, Park GR. Alfentanil infusions in patients requir-
doses fentanyl and sufentanil in men: influence on cardiovas- ing intensive care. Clin Pharmacokinet 1988; 15 (4): 216-26

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)
40 Tegeder et al.

250. Yate PM, Thomas D, Short SM, et al. Comparison of infusions failure compared with healthy volunteers. Anesthesiology
of alfentanil or pethidine for sedation of ventilated patients on 1997; 87 (3): 533-41
the ITU. Br J Anaesth 1986; 58 (10): 1091-9 256. Kapila A, Glass PS, Jacobs JR, et al. Measured context-sensitive
251. Sinclair ME, Sear JW, Summerfield RJ, et al. Alfentanil infu- half-times of remifentanil and alfentanil. Anesthesiology
sions on the intensive therapy unit. Intensive Care Med 1988; 1995; 83 (5): 968-75
14 (1): 55-9 257. Dershwitz M, Hoke JF, Rosow CE, et al. Pharmacokinetics and
252. James MK, Feldman PL, Schuster SV, et al. Opioid receptor
pharmacodynamics of remifentanil in volunteer subjects with
activity of GI 87084B, a novel ultra-short acting analgesic, in
isolated tissues. J Pharmacol Exp Ther 1991; 259 (2): 712-8 severe liver disease. Anesthesiology 1996; 84 (4): 812-20
253. Jhaveri R, Joshi P, Batenhorst R, et al. Dose comparison of
remifentanil and alfentanil for loss of consciousness. Anes-
thesiology 1997; 87 (2): 253-9
254. Hoke JF, Cunningham F, James MK, et al. Comparative phar- Correspondence and reprints: Dr Gerd Geisslinger, Center of
macokinetics and pharmacodynamics of remifentanil, its Pharmacology, Institute of Clinical Pharmacology, Johann
principle metabolite (GR90291) and alfentanil in dogs. J
Pharmacol Exp Ther 1997; 281 (1): 226-32 Wolfgang Goethe-University, Theodor Stern Kai 7, 60590
255. Hoke JF, Shlugman D, Dershwitz M, et al. Pharmacokinetics Frankfurt am Main, Germany.
and pharmacodynamics of remifentanil in persons with renal E-mail: geisslinger@em.uni-frankfurt.de

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1999 Jul; 37 (1)

Das könnte Ihnen auch gefallen