Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

SPECIAL POPULATIONS Clin. Pha rmocokinet.

1996 Dec; 31 (6): 41 0-422


03 12-5963/ 96/001 2-D4 10/$06.5O/0

© Adis International Limited. All rights reseNed .

Pharmacokinetics of Opioids in
Renal Dysfunction
Graham Davies, Christopher Kingswood and Martin Street
University of Brighton and Royal Sussex County Hospital, Brighton, England

Contents
Summary . . . . . . . . . . . . . . . . . . . . . . . . ... . . . . . . . 410
1. Pharmacokinetic Properties of Opioid Analgesics in Individuals with
Normal Renal Function . . . . . . . . . . . . . . . . . . . . . . 411
2. Pharmacokinetic Properties of Opioid Analgesics in Patients
with Renal Insufficiency . . . . . . . 411
2.1 Morphine and its Metabolites . 411
2.2 Fentanyl . 414
2.3 Alfentanil .. . 415
2.4 Sufentanil .. 415
2.5 Remifentanil . 415
2.6 Codeine ... 416
2.7 Dihydrocodeine 417
2.8 Buprenorphine . 417
2.9 Pethidine (Meperidine) 417
2.10 Dextropropoxyphene (Propoxyphene) 418
2.11 Methadone 418
3. Conclusions . . . . . . . . . . . . . . . . . . . 418

Summary Patients with renal insufficiency commonly require the administration of an


opioid analgesic to provide adequate pain relief. The handling of morphine,
pethidine (meperidine) and dextropropoxyphene in patients with renal insuffi-
ciency is complicated by the potential accumulation of metabolites. While mor-
phine itself remains largely unaffected by renal failure, accumulation, as denoted by
an increase in both mean peak concentrations and the area under the concentration-
time curve, of both the active metabolite (morphine-6-glucuronide) and the prin-
cipal metabolite (morphine-3-glucuronide, thought to possess opiate antagonist
properties) have been reported. The increased elimination half-lives of the toxic
metabolites norpethidine and norpropoxyphene in patients with poor renal func-
tion administered pethidine and dextropropoxyphene, respectively, makes their
routine use ill advised.
Case reports of prolonged narcosis associated with the use of both codeine and
dihydrocodeine in patients with renal insufficiency call for care to be used when
prescribing these agents under such conditions. Although the pharmacokinetics
of buprenorphine, alfentanil , sufentanil and remifentanil change little in patients
with renal failure, the continuous administration of fentanyl can lead to prolonged
sedation.
Opioids in Renal Dysfunction 411

Opioid analgesics effectively control a spectrum parent compound (as with morphine-6-glucuronide)
of different types of pain experienced by a diverse while others produce adverse effects on accumula-
patient population. However, pain is a very indi- tion (as with norpethidine). While the majority of
vidual response; effective pain relief, therefore, de- metabolites are renally cleared, glucuronide conju-
mands a tailored approach to drug use to accommo- gates may, in addition, be excreted via the biliary
date the variation in pain intensity, tolerance and system. Studies investigating the pharmacokinetic
adverse effects experienced by patients. This can properties of some of the commonly prescribed op-
best be achieved by an understanding of not only ioids in individuals with normal renal function are
the pharmacology of this group of agents but also summarised in table I.l3-IS]
the likely impact of disease on their pharmacoki-
netic properties. 2. Pharmacokinetic Properties of
Research has described the importance of the Opioid Analgesics in Patients with
liver as a major site for the biotransformation of Renal Insufficiency
these compounds, so that most prescribers demonstr-
ate caution when using opioids in patients with liver 2.1 Morphine and its Metabolites
dysfunction)l] However, although research has
2. 1. 1 Summary of Pharmacokinetics
shown a relationship between renal function and
Although morphine is widely distributed
the removal of some opioid metabolites, the area is
throughout the body, great interindividual varia-
generally less well researched, resulting in a lack
tion has been reported, ranging from a mean vol-
of clear advice on how opioids should be used in
ume of distribution (Vd) of 1.1 Llkg in I study[16]
patients with renal impairment. This article reviews
to 5.3 Llkg in another.[17] Morphine is highly ex-
the pharmacokinetics of the commonly used op-
tracted by the liver and undergoes rapid glucuron-
ioids in patients with renal impairment and, where
idation when administered parenterally and exten-
possible, in those receiving conventional or contin-
sive first-pass metabolism when given by mouth.
uous renal support.
The high clearance (eL) values reported, for example
48 L· h- I in I study[ 18] to over 100 L. h- I in another, [9]
1. Pharmacokinetic Properties of result in a short elimination half-life (tI/2~) of approx-
Opioid Analgesics in Individuals imately 2 hours.[9.10]
with Normal Renal Function Two main metabolites have been described:
morphine-3-glucuronide (the principal metabolite)
While most opioids are well absorbed from the
and morphine-6-glucuronide which account for
gastrointestinal tract, their bioavailability is re-
approximately 40 and 10%, respectively, of the
duced, to some degree, by presystemic metabolism
morphine dose recovered in the urine following
in the gut wall or liver. Where systemic bioavaila-
intravenous administrationJ9,19] While morphine-
bility is extensively reduced, as in the case of
6-glucuronide is a more potent analgesic than mor-
buprenorphine where only 10% of the oral dose
phine,P 1,201 morphine-3-glucuronide has been shown,
reaches the systemic circulationp] administration
in animal models, to antagonise the analgesic ef-
via the oral route is inappropriate. All opioids bind
fects of both morphine[21] and morphine-6-glucu-
to plasma proteins (both albumin and y-globulin)
ronide. [22]
although the extent varies from 7% (codeine) to
around 93% (sufentanil). Most opioids are readily 2.1.2 Effect of Renal Insufficiency
converted to more polar compounds, predomi- The main findings of studies involving patients
nantly by the liver, although the extent of extrahep- with renal failure and those requiring dialysis are
atic metabolism continues to be a focus for re- summarised in table 11.[ 18,23-29]
search interest. Although these metabolites are Several early reports, employing a specific radio-
generally less active, some are more potent than the immunoassay (RIA) technique to measure plasma

© Adis International limited. All rights reserved. Clin. Pharmacokinet. 1996 Dec; 31 (6)
412 Davies et al.

Table I. Pharmacokinetic data for commonly used opioids in individuals with normal renal function
Dose (mg) n PB Cmax AUCo~ tl}z~ Cl Vss Reference
and route (%) (nmoVL) (nmoillo h) (h) (mllmin) (l)
Fentanyl
0.171V 7 83 3.1 5.46 335a 3
80 3.7 956 25 4
Alfentanil
O.UIV 7 79 1.6 0.84 27 3
Codeine
60 IV 6 4.0 1050a 244a 5
7 6
Pethidine
65-80 3-8 266a 7
Buprenorphine
0.3 IV 6 6.6 650.5 313 8
Morphine and metabolites
51V 10 273.2 ±65.2 228.7 ± 36.6 1.7 ± 0.8 1966±313 279.3 ± 99.5 9
M6G as a metabolite 10 79.5 ± 15.1 313.4 ± 87 2.6 ± 0.69 9
of morphine
0.12b1V 6 302.1 c ± 163.9 139.9d ± 48.9 2.0 ± 1.4 1057.4 ± 498.8 180.6 ± 81 .5 10
M6G as a metabolite 6 28.8c ± 16.1 96.7d ± 71.2 1.9± 1.3 10
of morphine
M6G
0.03b IV 6 155H ± 49.3 208.8d ± 55.4 2.1 ± 1.2 187.8 ± 37.4 29.4 ± 18.4 11
Sufentanil
0.005 b IV 10 92.5 2.7 ± 0.4 886 ± 55 a 122 ± 13a 12
Remifentanil
0.002-0.03 b IV 24 92 0.17-0.34 4117-4950 25-40 14,15
a Normalised for 70kg adult.
b mg/kg.
c ng/ml.
d ng/ml o h.
Abbreviations and symbols: AUCo~ = area under the concentration-time curve from zero to infinity; Cl = total body clearance of drug from
plasma; Cmax = peak plasma drug concentrations aiter a single-dose administration; IV = intravenous; M6G = morphine-6-glucuronide; n =
number of participants; PB =protein binding; tl2P =elimination half-life; Vss =apparent volume of distribution at steady-state.

concentrations, suggested that morphine accumu- of high-performance liquid chromatography has


lated in patients with renal failure[30-32] and led the subsequently made it the method of choice in the
authors to postulate that the kidney may play an analysis of morphine and its metabolitesp4]
important role in the metabolism of the drug. How- More recent work has confirmed the accumula-
ever the findings of other workers,[23,25,33] using tion of the glucuronide metabolites in patients with
chromatographic analytical techniques, high- renal failure following the administration of intra-
lighted the problem of crossreactivity between the venous morphine as a bolus dose (table II). Whilst
parent compound and its metabolites when the RIA patients with end-stage renal disease (ESRD)
technique was used. demonstrate a significant reduction in both mor-
This, therefore, cast doubt over the accuracy of phine Vd[26.27] and CL,[27] the difference in t1;2~ was
the plasma morphine concentrations measured in not significant when compared with healthy indi-
earlier studies and consequently the clearance val- viduals. However, the maximum plasma concen-
ues reported. The speed, precision and specificity trations and area under the concentration-time

© Adis International Limited . All rights reserved. Clin. Pharmacokinet. 1996 Dec; 31 (6)
Opioids in Renal Dysfunction 413

curve for both morphine-6-glucuronide and morphine- concentrationP7] The CSF morphine-6-glucuron-
3-glucuronide were significantly higher in patients ide concentrations, 24 hours post-dose, were up to
with ESRD when compared with those measured 15 times greater in patients with renal failure, lead-
in the control groups suggesting a decrease in ing the authors to conclude that the accumulation
clearance of these metabolitesJ26.27] Subsequent of morphine-6-glucuronide in the plasma, because
work has demonstrated a significant linear relation- of renal failure, leads to progressive accumulation
ship between creatinine clearance (CLCR) and the in the CSF, which may partially explain the re-
renal clearances of both glucuronides,l35.36] Inter- ported sensitivity of renal failure patients to mor-
estingly, peritoneal dialysis appears to have no sig- phine.[37]
nificant effect on the pharmacokinetic behaviour of Further evidence of the accumulation of mor-
morphine, morphine-3-glucuronide or morphine- phine-6-glucuronide when renal function is com-
6-glucuronide when compared with patients with promised is provided by a study of patients admin-
ESRD.[27] istered an intravenous bolus of the compound)29]
Other workers have demonstrated that the in- The authors compared the disposition of the drug
creased plasma concentrations of the glucuronides in patients with ESRD, those requiring peritoneal
reported in patients with renal failure leads to a dialysis and renal transplant patients with good
significant increase in cerebrospinal fluid (CSF) kidney function. The ty, was significantly shorter

Table II. Pharmacokinetic data for morphine and morphine-6-glucuronide (M6G) in patients with renal insufficiency
Dose n Renal AUCo~ tlr"p Cl Vss Reference
(mg) function (mg/l· h) (h) (mllmin) (l)
0.125" IV 11 NR 3.5 (1.7-7.4) 805b (539-1218) 196 (116-307) 18
0.125" IV 9 ESRD 3.2 (1.0-6.6) 721 (371 -1428) 126.7 (41.3-256.9) 18
13.3-15.3c 1M 3 NR 134.6 ± 14.3 2.2 ±0.1 1493b ± 163 281 b ± 30.1 23
10-14.7c 1M 4 ESRD 101 .5 ± 5.7 1.2 ± 0.2 1948b ±222 214b ± 38.5 23
0.2" IV 7 NR 3.1 ±0.9 1491 b ±441 259b ±84 24
0.2" IV 9 ESRD 3.1 ±0.9 1197b ± 294 196b ± 70 24
41V 6 NR 1.7±0.8 1960b ±392 280b ± 161 25
41V 7 ESRD 2.4±1.1 1477b ± 434 308b ± 119 25
10lV 5 NR 4.8±0.9 741 ± 169 240.8 ± 55.1 26
9 ESRD 4.8±3.2 533 ±298 140.9 ± 37.8 26
0.1" IV 10 NR 228.7d ± 36.6 1.7±0.8 1966±313 232.2 ±76.5 27
0.1" IV 8 ESRD 398d ± 95.2 2.3 ± 0.8 1171 ± 337 127.3 ± 19.4 27
0.1" IV 9 PO 469.1d ± 101.4 2.0 ± 0.4 971 ± 184 135.1 ±20 27
0.1" IV 9 Trans 302.1d±105.3 2.0±0.9 1596 ± 468 175.4 ±95.3 27
3.3" ± 1.41V 4 CAVH 5.7 ±3.1 1127b ± 504 28
0.03" (M6G) IV 6 ESRD 27.1 ± 15.8 10.6 ± 5.6 17.5b ±4.2 29
0.03 (M6G) IV 6 PO 17.3 ± 4.6 14.3±6.2 18.9b ±4.2 29
0.03 (M6G) IV 6 Trans 2.1 ±0.7 96.0 ± 34.9 13.3b ± 2.1 29
a mglkg.
b Normalised for 70kg adult.
c Papaveretum administered.
d nmolll· h.
e Morphine given as a continuous infusion.
Abbreviations and symbols: AUCo·_ = area under the concentration-time curve from time zero to infinity. CAVH = continuous arteriovenous
haemofiltration; Cl =total body clearance of drug from plasma; ESRD = end stage renal disease; 1M = intramuscular administration; IV =
intravenous administration; M6G =morphine-6-glucuronide; n =number of patients; NR =normal renal function ; PO =peritoneal dialysis;
hr"p =elimination half-life; Trans =post kidney transplant; Vss =apparent volume of distribution at steady-state.

© Adis Internaftonal Umited. All rights reserved. Clin. Pharmacokinet. 1996 Dec; 31 (6)
414 Davies et al.

and the clearance significantly greater in the trans- ronide and morphine-3 -glucuronide clearly do.
plant patients compared with both ESRD and peri- Further data on the pharmacodynamics of both
toneal dialysis groups, but no difference in clear- glucuronides are required in order to establish their
ance was found between the 2 renal failure groups relative clinical merits and the nature of their inter-
(ESRD and peritoneal dialysis). The clinical impli- action with both morphine and each other. Until
cations of this finding are, as yet, unresolved; how- such data become available, morphine should be
ever, it is interesting to note that no prolonged re- used with caution in patients with severe renal im-
spiratory depression was observed in the patients pairment or those requiring renal replacement
studied. therapy.
Research investigating the profile of morphine
in critically ill patients suggests a complex rela- 2.2 Fentanyl
tionship between the drug's pharmacokinetic and 2.2.1 Summary of Pharmacokinetics
pharmacodynamic profile and the severity of the Fentanyl is a potent, highly lipid-soluble, syn-
patients' condition. Although morphine clearance thetic opioid which is commonly used as an adjunct
appears to vary considerably in the presence of to general anaesthesia. It has a short duration of
acute renal failure, [28.38] the influence of critical ill- action (0.5 to 1 hours) when given as a single in-
ness and acute renal failure on the pharmacology travenous bolus because it is rapidly distributed,
of the glucuronides has not been described. How- metabolised and excreted (tY2 approximately 3.5
ever, the influence of a variety of renal replacement hours). Fentanyl is metabolised in the liver by N-
therapies on the clearance of both morphine and dealkylation and hydroxylation to compounds that
morphine-6-glucuronide has been studied.l 28 .38] are both inactive and nontoxic. The main metabo-
Intermittent haemodialysis for between 3 and 5 lites, 4-N-(N-propionylanilo )piperidine and 4-N-
hours produced significant falls in morphine con- (N-hydroxypropionylanilo )piperidine are excreted
centrations; a mean of 75% (47 to 100%) when in the urine. Less than 10% of the drug is excreted
combined with a period of haemofiltration and a unchanged in the urine.
mean of 48% (24 to 84%) when dialysis alone was The pharmacokinetic properties of fentanyl
used.l 28 ] In addition a significant correlation was vary greatly within healthy volunteers!3] and pa-
found between morphine tY2 and the haemofiltrate tients,l39] Its initial short duration of action is
volumes collected. Both continuous haemofiltra- thought to be concentration-dependent, so that re-
tion and continuous haemodiafiltration techniques peated doses lead to higher plasma concentrations
(commonly used in the management of acute renal and consequently a longer duration of action.!40]
failure) appear to extract both morphine and mor- The second peak plasma concentration observed
phine-6-glucuronide efficiently.f3 8] The mean siev- following fentanyl administration was initially
ing coefficient reported (a measure of the fraction thought to be caused by the enterohepatic recircu-
of the drug which is able to pass through the artifi- lation of the drug,f411 However, this hypothesis ap-
cial kidney membrane) for morphine and mor- pears unlikely due to the high first-pass extraction
phine-6-glucuronide exceeded 0.5 when either of the drug, and is probably related to the redistri-
technique was used. However, the authors con- bution of the drug from certain body stores.
cluded that methods used to provide continuous 2.2.2 effect of Renal Insufficiency
renal support make only a minor contribution to the The clearance of fentanyl has been found to be
overall clearance of morphine (less than 1%) but normal in surgical patients with renal failure,! 42 1
may be responsible for a larger proportion of mor- although studies involving the critically ill have
phine-6-glucuronide clearance.!38] reported an increase in both tl/2 (25 hours) and Vd
Although morphine itself does not accumulate (over 1700L).l43 1 No dosage modification appears
in patients with renal failure, morphine-6-glucu- necessary when fentanyl is administered as a bolus

© Adis International Umited. All rights reserved. Clin. Pharmacokinet. 1996 Dec; 31 (6)
Opioids in Renal Dysfunction 415

dose in patients with ESRD, although it would ap- having left the plasma within 30 minutes.[12]
pear prudent to closely monitor this group of pa- Sufentanil is highly protein bound (92.5%, mainly
tients for signs of intoxication when it is used as a to (XI-acid glycoprotein)[13] and has a large appar-
continuous infusion. Due to the reportedly increased ent Vd (approximately 120L)[12] suggesting exten-
tY2 and the lack of data relating to the clearance of sive uptake of the drug into body tissues . The com-
fentanyl (when administered as a continuous in- pound is extensively metabolised by the liver
fusion), fentanyl should be used with caution in (hepatic extraction ratio = 0.8) by N-dealkylation,
critically ill patients with acute renal failure and to products thought to be inactive, O-demethyla-
consideration should be given to the use of an alter- tion, to a metabolite possessing approximately
native agent. 10% of the activity of the parent compound, and
some additional conjugation)49] Although very lit-
2.3 Alfentonil tle sufentanil is excreted unchanged, the metabo-
2.3. 7 Summary of Pharmacokinetics
lites are found in both the urine and faeces.
Alfentanil is a very short acting opioid with an The t l/2~ of sufentanil (approximately 3 hours)[12,50]
analgesic effect lasting between 5 and 10 minutes; is greater than values reported for alfentanil but
although related to fentanyl, it is less potent and less than those offentanyl; some variation has been
less lipid soluble. When compared with fentanyl it observed in patients undergoing cardiac surgery
possesses a faster onset and shorter duration of ac- (approximately 10 hours)[ 5I l and hyperventilated
tion; a result of its smaller Vd (30L compared with surgical patients (approximately 4 hours»)52 l
over 300L) and shorter tY2 (l.5 hours compared 2.4.2 Effect of Renal Insufficiency
with over 3 hours).[2 l Although a case report observed that a patient
Alfentanil is metabolised in the liver, mainly by with ESRD experienced prolonged postoperative
N- and O-dealkylation,[44 l to inactive compounds respiratory depression as a result of elevated
which are renally cleared. plasma concentrations of sufentanil,[ 49 l other stud-
2.3.2 Effect of Renal Insufficiency ies have found no significant differences between
Although patients with chronic renal failure ap- the clearance, t Y2 ~ or apparent Vd at steady-state
pear to exhibit a reduced protein binding of alfen- (V d ss ) of the drug when comparing patients with
tanil, both Vd (28L in ESRD compared with 20L ESRD to those possessing normal renal func-
in healthy individuals) and tY2 (l.8 hours in ESRD tion. [53.55l One study[53l has suggests that, because
compared with l.5 hours in healthy individuals), of the wide variability in both clearance and tl/2~
values were found to be no different to values ob- observed in their study,[53] administration of
tained from patients with normal renal function) 45 l sufentanil should be individualised in patients with
Similar results have been reported elsewhere)46,47l renal failure . However, this variation may, in part,
No alteration to the administration regimen of al- be explained by the population studied, namely,
fentanil is required for patients with renal impair- patients aged between 10 and 15 years.
ment.
2.5 Remifentonil
2.4 Sufentonil
2.5. 7 Summary of Pharmacokinetics
2.4. 7 Summary of Pharmacokinetics Although remifentanil is another fentanyl de-
Sufentanil is a highly potent thienyl analogue of rivative its unique structure makes it a novel agent
fentanyl. It is up to 10 times as potent as fen- within the phenylpiperidine group of drugs. The
tanyl[ 48 l and more lipophilic (partition coefficient incorporation of a methyl ester into the N-acyl moi-
between n-octanol and water of 1754, twice the ety renders it susceptible to hydrolysis by blood
value quoted for fentanyl»)1 3l. It is rapidly distrib- and tissue nonspecific esterases. This structural
uted into most tissues, with 98 % of an injected dose manipulation makes remifentanil the first ultra-

© Adis Internatio nal Limited. All rights reserved . Clin. Pharmaco kinet. 1996 Dec; 31 (6)
416 Davies et al.

short-acting opioid available to supplement gen- piperidine derivatives in attempting to achieve a


eral anaesthesia. It is rapidly de-esterified to the balanced anaesthesia suggests that a knowledge of
primary carboxylic acid metabolite (GI-90291) the (l/2P of the drug is of limited value.[60] The au-
while N-dealkylation (to carboxylic acid) is only a thors suggest that using computer simulations to
minor metabolic pathway in humans.f 56 ] Approxi- select an appropriate agent based on the pharmaco-
mately 90% of the drug is recovered in the urine as kinetic and pharmacodynamic properties of the
the carboxylic acid metabolite.f 57 ] This is consider- drugs, together with a knowledge of the intended
ably less active than its parent compound (it pos- duration of infusion, may be a more rational ap-
sesses only 0.1 to 0.3% of the relative potency of proach, although more reliable measures of opioid
remifentanil), but mean steady-state plasma con- effect during general anaesthesia need to be devel-
centrations have been reported to be 12 times greater oped.
than those measured for remifentanil.[l4]
A recent study investigating the pharmacokinet-
2.6 Codeine
ics of remifentanil in patients receiving elective
surgery[14] assigned individuals to 4 groups (of 6
patients) in order to compare different remifentanil 2.6.7 Summary of Pharmacokinetics
doses (2,5, 15 and 30 Ilg/kg) administered as a I-min- Codeine, a naturally occurring phenathrene al-
ute infusion. The pharmacokinetics of remifentanil kaloid, is principally metabolised by the liver to
were described using a 3-compartment model. codeine-6-glucuronide.f 61 ] The drug also under-
Clearance was found to lie between 4 and 5 Llmin, goes N-demethylation to norcodeine, which has a
apparent Vd ss between 25 and 40L and the terminal similar potency to the parent drug, and approximately
tl/2 between 10 and 21 minutes. Both CL and V dss 10% of the drug is desmethylated to morphine.[62]
were independent of dose while CL was not signif- The terminal tl/2of codeine in healthy volunteers
icantly affected by body weight or gender. The tl/2 lies between 2.5 and 3.5 hours[63] with only 3 to
of the carboxylic acid was found to lie between 88 16% of the parent drug being excreted unchanged
and 137 minutes. in the urine.f 64 ] Codeine is less than 10% protein
bound[6J with a Vd of between 3 to 4 Llkg.
2.5.2 Effect of Renal Insufficiency
A recent study compared the pharmacokinetics 2.6.2 Effect of Renal Insufficiency
of remifentanil (administered as an infusion over 4 Codeine is known to cause profound hypoten-
hours) in patients receiving renal dialysis and sion[65] as well as both central nervous system
healthy volunteers. The authors concluded that re- (CNS) and respiratory depression. [66] Patients with
nal failure had no significant effect on the pharmaco- renal failure given conventional doses of codeine
kinetics of remifentanil, with no significant differ- have been reported to develop narcosis.[67,68]
ence between the 2 groups for both CL or Vd values,
Guay et aI.l5] studied the removal of codeine in
although the terminal tl/2P of the metabolite carbo-
6 patients receiving haemodialysis and 6 healthy
xylic acid was significantly longer in the group
volunteers, The tyzPof codeine was found to be 18,7
with renal failure.[58] Shlugman and colleagues[59]
+ 9,0 hours in the haemodialysis group compared
reported that patients with renal failure [defined as
with 4 + 0.6 hours in those individuals with normal
a CLCR of < 1.8 Lih «30 ml/min)] are not more
kidney function.
sensitive to the respiratory depressant effects of
This unexpected accumulation of codeine in re-
remifentanil and suggested that it may be an ideal
nal failure may represent not only a change in the
agent for use in such patients.
pharmacokinetic properties of the drug but also an
2.5.3 Phenylpiperidine Derivatives: A Special Note enhanced dynamic response. The influence of both
Recent work investigating the pharmacokinetic conventional and continuous renal replacement
and pharmacodynamic relationship of the phenyl- therapies on the removal of codeine is unknown.

© Adis International Limited. All rights reserved. Clin. Pharmac okinet. 1996 Dec; 31 (6)
Opioids in Renal Dysfunction 417

2.7 Dihydrocodeine longer t \l2 (but not statistically significant) in those


with normal renal function (mean 6.6 hours) com-
2.7. 1 Summary of Pharmacokinetics
pared with those with renal impairment (mean 4
Dihydrocodeine is thought to have similar elim-
hours). Plasma concentrations of the metabolites,
ination pathways to codeine, via glucuronidation
particularly buprenorphine-3-glucuronide, increase
and demethylation in the liver, although this has
dramatically in patients with significant renal im-
not been fully evaluated. Rowell et aJ.l69) reported
pairment. The 4-fold rise in the plasma concentra-
a mean v/,~ of 3.9 hours in patients with normal
tion of norbuprenorphine, the active metabolite,
renal function.
in renal failure patients is unlikely to result in clin-
2.7.2 Effect of Rena//nsufficiency ically significant prolongation of the drug's action.
Although the renal clearance of dihydrocodeine
has not been measured, reports exist of prolonged 2.9 Pethidine (Meperidine)
narcosis in patients with renal failure taking the
2.9. 1 Summary of Pharmacokinetics
drug.l70.7I) Both patients described in the case re-
Pethidine was one of the first synthetic opioid
ports responded to multiple doses of naloxone. In
analgesics to be used in clinical practice and is less
a pharmacokinetic study of oral dihydrocodeine,
potent than morphine itself. It is subject to exten-
Barnes et al.[72) found similar tl/2~ in patients with
sive first-pass metabolism with a mean oral bio-
renal failure compared with normal individuals.
availability of 56%.[73) The drug is demethylated
However, the authors observed significantly
in the liver to its sole active metabolite norpethid-
higher peak plasma concentrations and AUC in re-
ine, which is either further hydrolysed to norpeth-
nal failure patients. They postulated that this find-
idinic acid and its conjugates or excreted un-
ing may reflect a reduction in the systemic clear-
changed in the urine.[74) Some of the drug is also
ance of the drug or an increase in its bioavailability.
hydrolysed to pethidinic acid. Although less than
The administration of therapeutic doses of di-
5% of an administered dose of the parent drug is
hydrocodeine, appropriate for those with normal recovered from the urine at normal pH «5%),[75)
renal function, to patients with renal failure may, in
acidification of the urine increases the recovery of
some patients, result in adverse effects developing.
both pethidine and norpethidine,l76)
Pethidine is extensively protein bound (be-
2.8 Buprenorphine
tween 60 and 80%), mainly to <Xl-acid glycopro-
2.8.1 Summary of Pharmacokinetics tein,(77) has a large Vd (4 to 5 Llkg) and a t\l2 of
This is a partial opioid agonist, 30 times more approximately 3.5 hours. l77 ) The terminal t \l, of
potent than morphine. It undergoes extensive first- norpethidine ranges between 14 and 21 hours in
pass metabolism when administered orally. It is individuals with good renal function.l 78 )
metabolised, by glucuronidation and dealkylation,
2.9.2 Effect of Rena/Insufficiency
in the liver to produce buprenorphine-3-glucuron-
Although the terminal t\l2 of pethidine appears
ide, which is inactive, and norbuprenorphine
to be unaffected by changes in renal function,
which is 40 times less potent than the parent drug.
norpethidine clearance is significantly correlated
Both products are principally excreted via the bil-
with CLCR l79) and consequently accumulates in re-
iary system.
nal failure patients (terminal t\l2 of 35 hours).[78)
2.8.2 Effect of Rena//nsufficiency Although norpethidine is a CNS depressant,
A recent study comparing the clearance of intra- possessing twice the proconvulsant potency of
venous buprenorphine, both single bolus and con- pethidine,(77) individual susceptibility to the cen-
tinuous infusion, in patients with impaired and nor- tral effects of stupor and convulsions varies. For
mal renal function reported no significant difference example, in a study of 9 patients with moderate to
between the 2 groups. [7) The authors reported a severe renal failure (serum creatinine 300 to 2160

© Adis International Limite d. All rights reserve d. Clin. Pharmaco kine t. 1996 Dec; 31 (6)
418 Davies et al.

mmoIlL), only 2 patients developed seizures. One unchanged. [S4] The authors postulated that a reduc-
had mild and the other severe renal impairment.[SO] tion in presystemic clearance may explain the in-
Kaiko et aUSl] studied 48 patients with creased AUe reported for dextropropoxyphene.
norpethidine-induced neurological adverse effects. Studies involving animals have shown that the ac-
Although 14 of this group had some degree of renal cumulation of dextropropoxyphene can cause both
impairment (as measured by raised urea level), eNS and respiratory depression, while both com-
only 5 of the 10 patients who experienced a seizure pounds, in overdose, contribute to the depressed
were from this group. The authors commented that cardiac conduction witnessed.[S7]
although renal failure may predispose patients re- Haemodialysis appears to inefficiently remove
ceiving pethidine to neurological problems, other dextropropoxyphene and its metaboliteJSSl The re-
factors (such as the degree of exposure and plasma moval of the drug by other renal replacement tech-
norpethidine : pethidine ratios) may be as impor- niques has not been studied.
tant. One fatality, due to seizures associated with
high norpethidine plasma concentrations, has been 2.11 Methadone
reported in an addict with renal failure who re- 2. 11. 1 Summary of Pharmacokinetics
ceived repeated doses of the drug.f s2 ] Unlike many opioid analgesics, methadone pos-
Although little research has been undertaken, it sesses a high oral bioavailability (mean of approx-
would seem wise to use pethidine with caution in imately 80%),[S9] is highly protein bound (between
patients with renal failure. Whilst the administra- 71 and 87%)[90] and has an t1/2~ in excess of 30
tion of a single dose appears to carry little risk for hours.[91] Methadone is metabolised, mainly by N-
patients with renal failure, its administration on a methylation, in the liver to 1,5-dimethyl-2-ethyl-
regular basis should be avoided. Pethidine clear- 3,3-diphenyl-1-pyrroline, the main metabolite.
ance in patients receiving conventional or contin- However, renal excretion of the unchanged drug is
uous dialysis has not been studied. an important route of methadone elimination, ac-
counting for approximately 20% of an adminis-
2.10 Dextropropoxyphene tered single oral doseJ92] The majority of metha-
(Propoxyphene) done (as the metabolite) is excreted in the faeces.
2. 10.1 Summary of Pharmacokinetics 2.11 .2 Effect of Renal Insufficiency
Dextropropoxyphene is structurally similar to In a case report detailing 2 patients with chronic
methadone and is commonly used, in combination renal failure receiving maintenance methadone
with paracetamol, for the control of mild-to-mod- therapy, the plasma concentrations reported were
erate pain. The drug is primarily demethylated in similar to those seen in patients with normal renal
the liver to yield, norpropoxyphene which is ex- functionJ93] This has led to speculation that pa-
creted by the kidneys.[S3] The terminal tl/2 of the tients with poor renal function are able to effec-
parent compound and metabolite range between tively eliminate methadone (as the primary meta-
11.8 and 14.6 hours and 22.9 to 36.6 hours, respec- bolite) by increasing the quantity excreted through
tively.f s4 ,S5] Dextropropoxyphene is approximately the faeces. Until studies have been undertaken to
80% protein bound and is widely distributed (Vd confirm this hypothesis some authors recommend
16 Llkg).f6] that the dose of methadone administered in ESRD
should be reduced by up to 50% of normaU6]
2. 10.2 Effect of Renal Insufficiency
The peak plasma concentrations and AUe for
3. Conclusions
both dextropropoxyphene and norpropoxyphene
have been reported to be increased in anephric pa- The relationship between renal impairment and
tients.[S6] While the tl/ 2 of norpropoxyphene was the use of opioid analgesics remains a complex is-
prolonged, that for dextropropoxyphene remained sue because of the paucity of research. Sufficient

© Adis International Limited. All rights reserved. Clin. Phormac okinet. 1996 Dec; 31 (6)
Opioids in Renal Dysfunction 419

Table III. Recommendations for prescribing opioids in patients with renal insufficiency
Drug Problem in renal insufficiency Suggested action
Pethidine Active metabolite (norpethidine) accumulates. Can cause Avoid regular use of conventional doses on
seizures patients with ESRD, IHD, PD and CRRT
Dextropropoxyphene Both parent compound and its main metabolite Avoid use in patients with ESRD, IHD, PD and
(norpropoxyphene) accumulate in renal insufficiency. May CRRT
cause CNS or respiratory depression or cardiotoxicity
Morphine Accumulation of morphine-6-glucuronide, a metabolite more Use with care in patients with ESRD, IHD, PD
potent (as an analgesic) than morphine and renally cleared andCRRT
Fentanyl Prolonged sedation may occur when continuously When continuous administration is required in
administered to patients with chronic renal failure patients with poor renal function close
monitoring is required. Consider use of an
alternative agent
Codeine Case reports of patients with renal failure developing narcosis Use lower doses or an alternative analgesic for
patients with renal insufficiency
Buprenorphine No significant changes in pharmacokinetic parameters in Normal dose can be used
patients with renal insufficiency
Methadone Potential for accumulation of methadone and its primary Reduce dose by up to 50% in ESRD or
metabolite in renal failure. Postulated increase in faecal dialysis patients
excretion of methadone in patients with renal insufficiency
Dihydrocodeine Reports of raised peak plasma concentrations and AUC in Use lower doses or an alternative analgesic for
patients with renal failure, although t;;.~ remained the same patients with renal insufficiency
as for patients with normal renal function. Prolonged narcosis
reported in some patients with renal failure
Remifentanil No significant changes reported in the pharmacokinetics of Rapid breakdown of remifentanil by plasma
remifentanil in patients with renal failure. Accumulation of and tissue esterases makes it potentially the
active metabolite [carboxylic acid metabolite (GI-90291); ideal agent for use in patients with renal
0.1-0.3% potency of remifentanil] in patients with renal insufficiency
impairment. However, the degree of accumulation is unlikely
to result in the patient experiencing significant pharmacological
effects
Sufentanil No significant changes in pharmacokinetic parameters in Case report of prolonged respiratory
patients with renal failure. Possible accumulation of less depression in a patient with chronic renal
potent metabolite (desmethyl-sufentanil; 10% activity of failure. Use with caution in such patients until
sufentanil). Clinical significance of such accumulation is further information on the use of sufentanil in
unknown patients with renal failure becomes available
Alfentanil Reduced protein binding Its short duration of action and lack of active
metabolites make it an ideal agent for use in
patients with renal impairment
Abbreviations: AUC = area under the concentration-time curve; CRRT = continuous renal replacement therapies; ESRD = end-stage renal
disease; IHD = intermittent haemodialysis; PD = peritoneal dialysis; tlf.1~ = elimination half-life.

evidence exists, especially in relation to morphine, Acknowledgements


to suggest that patients with renal insufficiency
are more sensitive to the effects of these agents. We acknowledge Mrs S. Gotham for preparation of the
manuscript, and 2 anonymous referees for helpful comments
This increased sensitivity may, in part, result from on an earlier draft.
the accumulation of active metabolites in addition
to the pharmacokinetic changes conferred on the
References
parent compound by renal impairment itself. I. Corall I, Williams R. Management of liver failure. Br J Anaesth
Although table III attempts to summarise the 1986;58: 234-45
findings of the research to date, dosage regimens 2. Hughes TJ. A pharmacological basis to the rational use of potent
analgesics. Hosp Update 1988 Nov; 14 (II): 2159-69
should always be titrated against the response of 3. Bower S, Hull CJ. Comparative pharmacokinetics of fentanyl
the individual patient. and alfentanil. Br J Anaesth 1982; 54: 871-7

© Adis International Limited. All rights reserved. Clin. Pharmacokinet. 1996 Dec; 31 (6)
420 Davies et al.

4. McClain DA , Hugg Cc. Intravenous fentanyl kinetics. Clin 22. Watt JA, Cramond T, Smith MT. Morphine-6-glucuronide: an-
Pharmacol Ther 1980; 28: 106-14 algesic effects antagonised by morphine-3-glucuronide [ab-
5. Guay DRP, Matzke GR, Findlay JWA: et al. Codeine pharma- stract]. Clin Exp Pharmacol Physiol 1990; 17: 83
cokinetics and pharmacodynamics in hemodialysis patients 23. Woolner DF, Winter D, Frendin TJ, et al. Renal failure does not
[abstract]. Clin Pharmacol ther 1987; 41 : 222 impair the metabolism of morphine. Br J Clin Pharmacol
6. Jusko WJ, Gretch M. Plasma and tissue protein binding of drugs 1986; 22: 55-9
in pharmacokinetics. Drug Metab Rev 1976; 5: 43-140 24. Chauvin M, Sandouk P, Scherrmann JM , et al. Morphine
7. Bennett WM, Aronoff GR, Morrison G, et al. Drug prescribing pharmacokinetics in renal failure. Anesthesiology 1987;
in renal failure: dosing guidelines for adults. Am J Kidney Dis 66: 327-31
1983; 3: 155-93 25. Sawe J, Odar-Cederlof I. Kinetics of morphine in patients with
8. Hand CW, Sear JW, Uppington J, et al. Buprenorphine disposi- renal failure. Eur J Clin Pharmacol 1987; 32: 377-82
tion in patients with renal impairment: single and continuous 26. Sear JW, Hand CW, Moore RA, et al. Studies on morphine
dosing with special reference to metabolites. Br J Anaesth- disposition: influence of renal failure on the kinetics of mor-
esiol 1990; 64: 276-82 phine and its metabolites. Br J Anaesth 1989; 62: 28-32
9. Osborne R, Joel S, Trew D, et al. Morphine and metabolite 27. Osborne R, Joel S, Grebenik K, et al. The pharmokinetics of
behavior after different routes of morphine administration: morphine and morphine glucuronides in kidney failure. Clin
demonstration of the importance of the active metabolite mor- Pharmacol Ther 1993; 54: 158-67
phine-6-glucuronide. Clin Pharmacol Ther 1990; 47: 12-9 28. Bion JF, Logan BK, Newman PM, et al. Sedation in intensive
10. Hanna MH, Peat SJ , Knibb AA, et al. Disposition of morphine- care: morphine and renal function . Intensive Care Med 1986;
6-glucuronide and morphine in healthy volunteers. Br J An- 12: 359-65
aesth 1991 ; 66: 103-7 29. Hanna MH, D'Costa F, Peat SJ , et al. Morphine-6-glucuronide
II. Hanna MH , Peat SJ , Woodham M, et al. Analgesic efficacy disposition in renal impairment. Br J Anaesth 1993; 70: 511-4
30. Moore RA, Sear JW, Bullingham RES, et al. Morphine kinetics
and CSF pharmacokinetics of intrathecal morphine-6-glu-
in renal failure. In: Foley KM, Inturrisi CE, editors. Opioid
curonide: comparison with morphine. Br J Anaesth 1990;
analgesics in the management of cancer pain. New York: Ra-
64: 547-50
ven Press, 1986; 65-72
12. Bovill JG, Sebel PS, Blackburn CL, et al. The pharmokinetics
31. Ball M, McQuay HJ, Moore RA, et al. Renal failure and the use
of sufentanil in surgical patients. Anesthesiology 1984; 61:
of morphine in intensive care. Lancet 1985 Apr 6; I: 784-6
502-6
32. Shelly M, Park GR. Renal failure and use of morphine in inten-
13. Meuldermans WEG, Hurkmans RMA, Heykants JJP. Plasma
sive care [letter]. Lancet 1985 May 1\; I: 1100
protein binding and distribution of fentanyl, sufentanil , al-
33. Osborne RJ, Joel SP, Slevin ML. Morphine intoxication in renal
fentanil and lofentanil in blood. Arch Int Pharmacodyn Ther
failure : the role of morphine-6-glucuronide. BMJ 1986 Jun
1982; 257:4-19
14; 292: 1548-9
14. Westmoreland CL, Hoke JF, Sebel PS, et al. Pharmokinetics of
34. Glare PA, Walsh TD. Clinical pharmacokinetics of morphine.
remifentanil (GI87084B) and its major metabolite (GI9029I )
Ther Drug Monit 1991; 13: 1-23
in patients undergoing inpatient surgery. Anesthesiology
35. Milne RW, Nation RL, Somogyi AA, et al. The influence of
1993; 79: 893-903
renal function on the renal clearance of morphine and its glu-
15. Lemmens HJM. Pharmacokinetic-pharmacodynamic relation-
curonide metabolites in intensive-care patients. Br J Clin
ships for opioids in balanced anaesthesia. Clin Pharmacokinet Pharmacol 1992; 34: 53-9
1995; 29 (4): 231-42 36. Peterson GM, Randall CTC, Paterson J. Plasma levels of mor-
16. Moore A, Sear J, Baldwin D, et al. Morphine kinetics during phine and morphine glucuronides in the treatment of cancer
and after renal transplantation. Clin Pharmacol Ther 1984; 35: pain: relationship to renal function and route of administra-
641-5 tion. EurJ Clin Pharmacol 1990; 38: 121-4
17. Hoskin PJ, Hanks GW, Aherne GW, et al. The bioavailability 37. D' Honneur G, Gilton A, Sandouk P, et al. Plasma and cerebro-
and pharmacokinetics of morphine after intravenous, oral and spinal fluid concentrations of morphine and morphine glucu-
buccal administration in healthy volunteers [abstract]. Br J ron ides after oral morphine. Anesthesiology 1994 Jul; 81:
Clin Pharmacol 1989; 27: 499 87-93
18. Aitkinhead AR, Vater M, Achola K, et al. Pharmacokinetics of 38. Davies JG, Coombes ID, Kingswood C, et al. The clearance of
single-dose IV morphine in normal volunteers and patients morphine and morphine-6-glucuronide in critically ill pa-
with end-stage renal failure. Br J Anaesth 1984; 56: 813-9 tients receiving continuous renal replacement therapies [ab-
19. Sawe J. Morphine and its 3- and 6-glucuronides in plasma and stract). J Pharm Pharmacol 1994; 46 Suppl2: 1045
urine during chronic oral administration in cancer patients. In : 39. Bentley JB, Borel JD, Nenad RE, et al. Age and fentanyl phar-
Foley KM,lnturrisi CE, editors. Opioid analgesics in the man- macokinetics. Anesth Analg 1982; 61: 968-71
agement of cancer pain. New York: Raven Press, 1986: 45-55 40. Hug CC, Murphy MR. Fentanyl disposition in cerebrospinal
20. Osborne R, Joel S, Trew D, et al. Analgesic activity of mor- fluid and plasma and its relationship to ventilatory depression
phine-6-glucuronide. Lancet 1988; II : 828 in the dog. Anesthesiology 1979; 50: 342-9
21. Smith MT, Watt JA, Cramond T. Morphine-3-glucuronide: a 41. Stoeckel H, Hengstmann JH, Schuttler J. Pharmacokinetics of
potent antagonist of morphine analgesia. Life Sci 1990; 47: fentanyl as a possible explanation for recurrence of respira-
579-85 tory depression. B J Anaesth 1979; 51: 741-5

© Adis International Umited. All rights reserved. Clin. Pharmacokinet. 1996 Dec: 31 (6)
Opioids in Renal Dysfunction 421

42. Coral 1M, Moore AR, Strunin L. Plasma concentrations of fen- 61. Hull JH, Findlay JWA, Rogers JF, et al. An evaluation of the
tanyl in normal surgical patients with severe renal failure . Br effects of smoking on codeine pharmacokinetics and bio-
J Anaesth 1980; 52: 10 I P availability in normal human voluneteers. Drug Intell Clin
43. Alazia M, Levron JC, Guidon C, et al. Pharmacokinetics of Pharm 1982; 16: 849-54
fentanyl during continuous infusion in critically ill patients 62. Posey BL, Kimble SN . High-performance liquid chromato-
[abstract]. Anesthesiology 1987; 67: 665 graphic study of codeine, norcodeine and morphine as indi-
44. Meuldermans W, Van Peer A, Hendricks J, et al. Alfentanil cators of codeine ingestion. J Anal Toxicol 1984; 8: 68-74
pharmacokinetics and metabolism in humans. Anesthesiol- 63. Gilman AG, Goodman LS, Rail TW, et aI. , editors. Goodman
ogy 1988; 69: 527-34 and Gilman's the pharmacological basis of therapeutics. 7th
45. Chauvin M, Lebrault C, Levron JC, et al. Pharmacokinetics of ed. New York: Macmillan, 1985
alfentanil in chronic renal failure . Anesth Analg 1987; 66: 64. Way EL, Adler TK. The pharmacologic implications of the
53-6 fate of morphine and its surrogates. Pharmacol Rev 1968;
46. Bower S, Sear Jw. Disposition of alfentanil in patients receiv- 12: 383-446
ing a renal transplant. J Pharm Pharmacol 1989; 41 : 654-7 65. Parke TJ, Nandi PR, Bird KJ, et al. Profound hypotension fol-
47. Davis PJ, Stiller RL, Cook DR, et al. Effects of cholestatic lowing intravenous codeine phosphate. Anaesthesia 1992; 47:
hepatic disease and chronic renal failure on alfentanil phar- 852-4
macokinetics in children. Anesth Analg 1989; 68: 579-83 66. Pearson MA. A fatality due to the ingestion of codeine
48. Rolly G, Kay B, Cockx F. A double-blind comparison of high (methylmorphine). Clin Toxicol1979; 15: 267-71
doses of fentanyl and sufentanil in man. Acta Anaesthesiol 67. Levine OF. Hypocalcaemia increases the narcotic effect of co-
Belg 1979; 30: 247-56 deine. Postgrad Med J 1980; 56: 736-7
49. Wiggum DC, Cork RC, Weldon ST, et al. Postoperative respi- 68. Matzke GR, Chan GLC, Abrahim PA. Codeine dosage in renal
ratory depression and elevated sufentanil levels in a patient failure. Clin Pharm 1986; 5: 15-6
with chronic renal failure . Anesthesiology 1985; 63: 708-10 69. Rowell FJ, Seymour RA, Rawlins MD. Pharmacokinetics of
50. Lehmann KA, Sipakis K, Gasparini R, et al. Pharmacokinetics intravenous and oral dihydrocodeine and its acid metabolites.
of sufentanil in general surgical patients under different con- Eur J Clin Pharmacol 1983; 25: 419-24
ditions of anaesthesia. Acta Anaesthesiol Scand 1993; 37 (2): 70. Barnes IN, Goodwin FJ. Dihydrocodeine narcosis in renal fail-
176-80 ure . BMJ 1983; 286: 438-9
51. Howie MB, Smith OF, Harrington K, et al. Serum concentra- 71. Redfern N. Dihydrocodeine overdose treated with naloxone in-
tions of sufentanil and fentanyl in the post-operative course fusion . BMJ 1983; 287: 751-2
in cardiac surgery patients. Anesthesiology 1984; 61: A 131 72. Barnes IN, Williams AJ, Tomson MJF, et al. Dihydrocodeine in
52. Schwartz AE, Matteo RS, Ornsteine, et al. Pharmacokinetics of renal failure: further evidence for an important role of the
sufentanil in hyperventilated patients [abstract]. Anaesth An- kidney in the handling of opioid drugs. BM] 1985; 290: 740-2
alg 1987; 66 Suppl: 151 73. Mather LE, Tucker GT. Systemic availability of orally admin-
53. Davis PJ , Stiller RL, Cook DR, et al. Pharmacokinetics of istered meperidine. Clin Pharmacol Ther 1976; 20: 535-40
sufentanil in adolescent patients with chronic renal failure. 74. Mather LE, Meffin Pl Clinical pharmacokinetics of pethidine.
Anesth Analg 1988; 67: 268-71 Clin Pharmacokinet 1978; 3: 352-68
54. Fyman P, Avitable M, Moser F, et al. Sufentanil pharmacokinet- 75. Stambaugh JE, Wainer IW, Sanstead JK, et al. The clinical phar-
ics in patients undergoing renal transplantation [abstract]. An- macology of meperidine: comparison of routes of administra-
esth Analg 1987 ; 66 Suppl.: 62 tion. J Clin Pharmacol 1976; 16: 245-56
55. Sear Jw. Sufentanil disposition in patients undergoing renal 76. Asatoor AM , London DR , Milne MD, et al. The excretion of
transplantation: influence of choice of kinetic model. Br J pethidine and its derivatives. Br J Pharmacol 1963; 20:
Anaesth 1989; 63: 60-7 285-98
56. Egan TO, Lemmens HJM , Fiset P, et al. The pharmacokinet- 77. Edwards OJ, Svensson CK, Visco JP, et al. Clinical pharmaco-
ics of the new short-acting opioid remifentanil (GI87084B) kinetics of pethidine. Clio Pharmacokinet 1982; 7: 421-33
in healthy adult male volunteers. Anesthesiology 1993; 79: 78. Tang R, Shimomura SK, Rotblatt M. Meperidine-induced sei-
881-92 zures in sickle cell patients. Hosp Formul 1980; 15: 764-72
57. Glass PSA, Hardman 0 , Kamiyama Y, et al. Preliminary phar- 79. Odar-Cederlof I, Boreus LO, Bondesson U, et al. Comparison
macokinetics and pharmacodynamics of an ultra-short acting of renal excretion of pethidine (meperidine) and its metabo-
opioid: remifentanil (GI87084B). Anesth Analg 1993; 77: lites in old and young patients. Eur J Clin Pharmacol 1985;
1031-40 28: 171-5
58. Hoke J, Muir K, Glass P, et al. Pharmacokinetics of remifentanil 80. Szeto HH , Inturrisi E, Houde E, et al. Accumulation of
and its metabolite (G19029I ) in subjects with renal disease normeperidine, an active metabolite of meperidine, in pa-
[abstract]. Clin Pharmacol Ther 1995; 57: PI55 tients with renal failure or cancer. Ann Intern Med 1977; 86:
59. Shlugman 0 , Dufore S, Dershwitz M, et al. Respiratory effects 738-41
of remifentanil in subjects with severe renal impairment com- 81. Kaiko RF, Foley KM, Grabinski PY, et al. Central nervous sys-
pared to matched controls [abstract]. Anesthesiology 1994; tem excitatory effects of meperidine in cancer patients. Ann
81: 1417 Neurol 1982; 13: 180-5
60. Shafer SL, Varvel JR. Pharmacokinetics, pharmacodynamics, 82. Jiraki K. Lethal effects of normeperidine. Am J Forensic Med
and rational opioid selection. Anesthesiology 1991; 74: 53-63 Pathol 1992; 13 (I): 42-3

© Adis International Limited. All rights reserved. Clin. Pharmacokinet. 1996 Dec; 31 (6)
422 Davies et al.

83. Inturrisi CE, Colburn WA, Verebely K, et al. Propoxyphene and 89. Nilsson MI, Meresaar V, Anggard E. Clinical pharmacokinetics
norpropoxyphene kinetics after single and repeated doses of of methadone. Acta Anaesthesiol 1982; 74 Suppl: 66-9
propoxyphene. Clin Pharmacol Ther 1982; 31: 157-67 90. Berkowitz BA. The relationship of pharmacokinetics to phar-
84. Wolen RL, Ziege EA, Gruber CM. Determination of propoxy- macological activity: morphine, methadone and naloxone.
phene and norproxyphene by chemical ionization mass Clin Pharmacokinet 1976; I: 219-30
fragmentography. Clin Pharmacol Ther 1974; 17: 15-20 91. Gourley GK, Wilson PR, Glynn CJ. Pharmacodynamics and
85. Gram LF, Schou J, Way WL, et al. d-Propxyphene kinetics after pharmacokinetics of methadone during the perioperative pe-
riod. Anesthesiology 1982; 57: 458-67
single oral and intravenous doses in man. Clin Pharmacol
92. Inturrisi CE, Verebely K. Disposition of methadone in man after
Ther 1979; 26: 473-82
a single oral dose. Clin Pharmacol Ther 1972; 13: 923-30
86. Gibson TP, Giacomini KM , Briggs WA, et al. Propoxyphene and
93. Kreek MJ, Schecter AJ, Gutjahar CL, et al. Methadone use in
norpropoxyphene plasma concentrations in the anephric pa-
patients with chronic renal disease. Drug Alcohol Depend
tient. Clin Pharmacol Ther 1980; 27: 665-70 1980; 5: 197-205
87. Nickander R, Smits SE, Steinberg MI. Propoxyphene and
norpropoxyphene: pharmacologic and toxic effects in animals.
J Pharmacol Exp ther 1977; 200: 245-53
88. Giacomini KM, Gibson TP, Levy G. Effect of hemodialysis on Correspondence and reprints: Graham Davies, Department
propoxyphene and norpropoxyphene concentrations in blood of Pharmacy, University of Brighton, Moulsecoomb, Brigh- .
of anephric patients. Clin Pharmacol Ther 1980; 27: 508-14 ton BN2 4GJ, England.

© Adis International Umited. All rights reserved. Clin. Pharmacokinet. 1996 Dec; 31 (6)

You might also like