ArticlePDF Available

Pharmacokinetics of Oral and Intravenous Paracetamol (Acetaminophen) When Co-Administered with Intravenous Morphine in Healthy Adult Subjects

Authors:

Abstract

Background and objective: Several features favor paracetamol (acetaminophen) administration by the intravenous rather than the oral route in the postoperative setting. This study compared the pharmacokinetics and bioavailability of oral and intravenous paracetamol when given with or without an opioid, morphine. Methods: In this randomized, single-blind, parallel, repeat-dose study in healthy adults, subjects received four repeat doses of oral or intravenous 1000 mg paracetamol at 6-h intervals, and morphine infusions (0.125 mg/kg) at the 2nd and 3rd intervals. Comparisons of plasma pharmacokinetic profiles were conducted before, during, and after opioid co-administrations. Results: Twenty-two subjects were included in the pharmacokinetic analysis. Observed paracetamol peak concentration (C max) and area under the plasma concentration-time curve over the dosing interval (AUC0-6) were reduced when oral paracetamol was co-administered with morphine (reduced from 11.6 to 7.25 µg/mL and from 31.00 to 25.51 µg·h/mL, respectively), followed by an abruptly increased C max and AUC0-6 upon discontinuation of morphine (to 13.5 µg/mL and 52.38 µg·h/mL, respectively). There was also a significantly prolonged mean time to peak plasma concentration (T max) after the 4th dose of oral paracetamol (2.84 h) compared to the 1st dose (1.48 h). However, pharmacokinetic parameters of paracetamol were not impacted when intravenous paracetamol was co-administered with morphine. Conclusions: Morphine co-administration significantly impacted the pharmacokinetics of oral but not intravenous paracetamol. The abrupt release of accumulated paracetamol at the end of morphine-mediated gastrointestinal inhibition following oral but not intravenous administration of paracetamol suggests that intravenous paracetamol provides a better option for the management of postoperative pain. CLINICALTRIALS. Gov identifier: NCT02848729.
ORIGINAL RESEARCH ARTICLE
Pharmacokinetics of Oral and Intravenous Paracetamol
(Acetaminophen) When Co-Administered with Intravenous
Morphine in Healthy Adult Subjects
Robert B. Raffa
1
Jayne Pawasauskas
2
Joseph V. Pergolizzi Jr
3
Luke Lu
4
Yin Chen
5
Sutan Wu
5
Brant Jarrett
4
Randi Fain
4
Lawrence Hill
4
Krishna Devarakonda
4
The Author(s) 2017. This article is an open access publication
Abstract
Background and Objective Several features favor parac-
etamol (acetaminophen) administration by the intravenous
rather than the oral route in the postoperative setting. This
study compared the pharmacokinetics and bioavailability
of oral and intravenous paracetamol when given with or
without an opioid, morphine.
Methods In this randomized, single-blind, parallel, repeat-
dose study in healthy adults, subjects received four repeat
doses of oral or intravenous 1000 mg paracetamol at 6-h
intervals, and morphine infusions (0.125 mg/kg) at the 2nd
and 3rd intervals. Comparisons of plasma pharmacokinetic
profiles were conducted before, during, and after opioid co-
administrations.
Results Twenty-two subjects were included in the phar-
macokinetic analysis. Observed paracetamol peak con-
centration (C
max
) and area under the plasma concentration-
time curve over the dosing interval (AUC
0–6
) were reduced
when oral paracetamol was co-administered with morphine
(reduced from 11.6 to 7.25 lg/mL and from 31.00 to
25.51 lgh/mL, respectively), followed by an abruptly
increased C
max
and AUC
0–6
upon discontinuation of
morphine (to 13.5 lg/mL and 52.38 lgh/mL, respec-
tively). There was also a significantly prolonged mean time
to peak plasma concentration (T
max
) after the 4th dose of
oral paracetamol (2.84 h) compared to the 1st dose
(1.48 h). However, pharmacokinetic parameters of parac-
etamol were not impacted when intravenous paracetamol
was co-administered with morphine.
Conclusions Morphine co-administration significantly
impacted the pharmacokinetics of oral but not intravenous
paracetamol. The abrupt release of accumulated paraceta-
mol at the end of morphine-mediated gastrointestinal
inhibition following oral but not intravenous administration
of paracetamol suggests that intravenous paracetamol
provides a better option for the management of postoper-
ative pain.
ClinicalTrials.gov Identifier NCT02848729.
Key Points
Morphine co-administration significantly impacts the
pharmacokinetics of oral but not intravenous
paracetamol by reducing/delaying its absorption and
substantially increasing the inter-individual
pharmacokinetic variability
Intravenous paracetamol produces more
predictable blood levels than oral paracetamol when
either are co-administered with morphine, and thus
provides a better option for the management of
postoperative pain in the context of multimodal
analgesia
&Krishna Devarakonda
krishna.devarakonda@mallinckrodt.com
1
University of Arizona College of Pharmacy, Tucson,
AZ 85718, USA
2
The University of Rhode Island College of Pharmacy,
Kingston, RI 02881, USA
3
NEMA Research, Inc., Naples, FL 34108, USA
4
Mallinckrodt Pharmaceuticals, Perryville III Corporate Park,
53 Frontage Road Third Floor, P.O. Box 9001, Hampton,
NJ 08827-9001, USA
5
Mallinckrodt Pharmaceuticals, Hazelwood, MO 63042, USA
Clin Drug Investig
https://doi.org/10.1007/s40261-017-0610-4
1 Introduction
Opioid agonists such as morphine continue to be important
analgesics in the treatment of pain in the immediate peri-
operative and critical care settings [1], although they are
associated with numerous adverse drug events [2]. In an
effort to reduce opioid exposure and thus minimize opioid-
related adverse drug events, non-opioid analgesics are
being incorporated as foundational therapy into multimodal
analgesia protocols for the management of postoperative
pain. Numerous surgical and nonsurgical medical societies
and accrediting and quality organization guidelines rec-
ommend the use of multimodal analgesia (MMA) in order
to reduce exposure to opioids [39]. Many also recommend
scheduled use of non-opioid analgesics (paracetamol [ac-
etaminophen], non-steroidal anti-inflammatory drugs
[NSAIDs]) as the first- line foundation of MMA.
A recent review [10] and the American Society of
Anesthesiologists (ASA) guidelines [11] have suggested
there is inadequate differentiation of intravenous and oral
paracetamol to warrant the higher acquisition cost and
longer time to administer the intravenous formulation.
Given that opioids remain a key component of post-
operative pain management, it is important to note that
opioids, along with surgical stress may have a negative
impact on orally administered drugs. Opioids inhibit gas-
trointestinal motility, including delaying gastric emptying
[12] and thus the route of administration of analgesics used
in multimodal regimens may be an important considera-
tion. Specifically, absorption of orally administered
paracetamol may be compromised in patients receiving
opioids, which could reduce efficacy. Furthermore, delayed
absorption of orally administered paracetamol in patients
receiving opioids could result in gastric accumulation of
paracetamol, thereby markedly changing the pharmacoki-
netic profile during and after opioid administration.
Paracetamol can significantly reduce the use of opioid
analgesics when concomitantly administered for the treat-
ment of acute pain [13]. The use of intravenous paraceta-
mol concomitantly with opioids has gained increased
popularity for postsurgical pain relief over orally admin-
istered paracetamol because it provides an immediate peak
plasma concentration [14] and is thought to provide a faster
analgesic effect. Bioavailability of intravenous paracetamol
is 100%, whereas the oral bioavailability of paracetamol
can be as low as 79% [15]. In a study comparing the
pharmacokinetics of paracetamol 1 g administered via
intravenous, oral, or rectal routes, cerebrospinal fluid (CSF)
area under the concentration-time curve from 0 to 6 h
(AUC
0–6
) was 24.9, 14.2, and 10.3 lgh/mL, respectively.
Absorption phase, variability in plasma and CSF were
greater with oral and rectal administration than with
intravenous administration [14].
Furthermore, in patients receiving opioids, the absorp-
tion of orally administered paracetamol may be delayed
and could result in gastric accumulation of paracetamol,
thereby markedly changing the pharmacokinetic profile.
Opioid-induced inhibition of gastrointestinal function
would not be expected to affect intravenous paracetamol
pharmacokinetics.
To assess potential interaction, the current study evalu-
ated whether an opioid, intravenous morphine, commonly
used to treat postsurgical pain, affects the absorption of
oral or intravenous paracetamol and results in altered
pharmacokinetics during and after co-administration in
healthy subjects.
2 Methods
This was an institutional review board (IRB)-approved,
randomized, single-blind, parallel, single-site, repeat-dose
study (NCT02848729) in healthy adult subjects from 18 to
55 years of age (inclusive). The study was completed with
four cohorts of subjects: both males and non-pregnant
females were enrolled in Cohort 1, and male subjects only
in Cohorts 2–4. Subjects were randomly assigned to 1 of 2
parallel groups: 4 repeat doses of 1000 mg oral paraceta-
mol (2 tablets, 500 mg/tablet; Mallinckrodt Inc., Hazel-
wood, Missouri, USA) at hours 0, 6, 12, and 18, and a
dummy 15-min intravenous infusion of saline at the same
time points, or 4 doses of intravenous paracetamol (1000
mg/100 mL; Ofirmev
, Mallinckrodt Inc., Hazelwood,
Missouri, USA) delivered as an infusion at hours 0, 6, 12,
and 18, and 2 placebo tablets at the same time points.
Intravenous morphine infusions (0.125 mg/kg in 100 mL
saline, approximately 15 min; morphine sulfate injection
10 mg/mL, 1 mL, West-Ward Pharmaceutical Corp.,
Eatontown, New Jersey, USA) occurred at hours 6 and 12
for all subjects. The total study duration for each subject
was approximately 38 days. A drop-out rate of 50% was
anticipated due to the required discontinuation of subjects
who experienced emesis and the known association
between morphine and emesis.
2.1 Bioanalytical Method to Determine Plasma
Concentration of Paracetamol
Venous blood was collected by venipuncture in K
2
EDTA-
containing tubes, and plasma was separated and stored at
-80 C. Quantification of paracetamol in plasma was
performed via high-performance liquid chromatography
(HPLC) with paired mass spectrometry (MS/MS). Analysis
of human plasma samples began on 16 March 2016 and
R. B. Raffa et al.
was completed on 01 April 2016. Plasma samples were
analyzed with a lower limit of quantitation of 0.100 lg/mL
and upper limit of quantitation of 50.0 lg/mL. Each cali-
bration curve was calculated using a linear (1/concentra-
tion
2
weighted) least-squares regression algorithm.
Precision and accuracy were evaluated by replicate anal-
yses of human plasma quality control pools prepared at five
concentrations spanning the calibration range. To demon-
strate reproducible quantitation of incurred subject sam-
ples, approximately 10% of the study samples were re-
assayed. Incurred sample repeats were considered accept-
able if the original and re-assay values from two-thirds of
the repeated samples had a relative percent difference of
B20%. The results of the incurred sample repeats met the
acceptance criteria.
2.2 Pharmacokinetics
Venous blood samples (6 mL each) for measuring parac-
etamol concentrations were collected at the following
times:
up to 30 min prior to the administration of the first dose
of paracetamol;
at 15, 30, and 45 min (±2 min), 1, 2, 3, 4, 6 h (±5 min
for hours 1, 2, 3, and 4; ±10 min for hour 6) after the
first paracetamol dose;
at 30 min (±2 min), 1, 2, 3, 4 and 6 h (±5 min for
hours 1, 2, 3, and 4; ±10 min for hour 6) after both
second and third paracetamol doses;
and at 15, 30, and 45 min (±2 min), 1, 2, 3, 4, 6, 8, 10,
12, and 18 h (±5 min for hours 1, 2, 3, 4, 8, 10, 12 and
18; ±10 min for hour 6) after the fourth dose of
paracetamol.
Blood draws at 6 h after the first, second, and third
paracetamol doses were collected before the next parac-
etamol dose that was to be administered.
A total of 33 blood samples were collected for each
subject. The schedules for blood samples collection were
the same for all study groups.
Approximately 200 mL of blood for determination of
plasma concentrations of paracetamol was collected, and
18 mL was collected for clinical laboratory tests at the
screening visit and check-in. The time (24-h clock) and
date of collection for each sample was recorded.
2.3 Safety
The following safety assessments were evaluated: medi-
cal/surgical history, physical examination, pregnancy test-
ing (Cohort 1 female subjects only) electrocardiogram,
vital signs, clinical laboratory testing (including standard
assessment of renal and hepatic function), pulse oximetry,
concomitant medication use, and assessment of adverse
events (AEs).
2.4 Statistical Analysis
This study was regarded as exploratory and thus sample
size was not determined by power analysis. Two popula-
tions were identified and analyzed in this study. The safety
population comprised subjects enrolled in the study that
received any quantity of study drug. The per-protocol
population comprised subjects who received all study drug
doses and provided all 33 protocol-specified blood samples
within required time frames without any major protocol
deviations.
Pharmacokinetic simulations were performed to predict
the paracetamol concentrations after the second, third, and
fourth doses of paracetamol assuming no co-administration
of morphine. The simulations were based on the observed
pharmacokinetic values after the first dose of orally or
intravenously administered paracetamol. Simulations were
performed using Phoenix
WinNonlin
V 6.4 (Certara,
Princeton, NJ).
Descriptive statistics for continuous variables included
number of values (N), mean, standard deviation (SD),
median, minimum, and maximum, unless otherwise noted.
Frequency and percentages were calculated for categorical
variables. In anticipation of having a limited number of
subjects available for final analysis (due to a high rate of
emesis-related drop-outs), the paired t-test was used in
comparisons within the oral paracetamol group or within
the intravenous paracetamol group, across paracetamol
doses, and the two-sample t-test was used in comparisons
across the oral and intravenous paracetamol groups. To
control for multiple comparisons, both raw pvalues from
the paired t-test and Hochberg adjusted pvalues were
calculated, and only the adjusted pvalues were presented.
All statistical significance testing was 2-tailed using
a=0.05.
Data summary and analyses were performed with SAS
9.2 or higher.
3 Results
3.1 Subject Disposition
A total of 50 subjects were enrolled in the study, 23 sub-
jects (46.0%) completed, and 27 subjects (54.0%) were
discontinued early. Twenty-six subjects (52.0%) discon-
tinued due to emesis and 1 subject withdrew consent to
participate in the study; no subjects discontinued due to any
other treatment-emergent adverse event (TEAE). A total of
22 subjects were included in the per-protocol population
PK of Oral and IV Paracetamol When Co-administered with IV Morphine
and used in the pharmacokinetic analysis. One subject
completed the study but did not provide all pharmacoki-
netic samples and thus was excluded from the per-protocol
population. No data imputation was performed.
3.2 Demographic and Baseline Characteristics
Of the 50 subjects enrolled, 34 (68%) were white and 16
(32%) were black or African American. The majority
(64%; 32/50) were not Hispanic or Latino in ethnicity.
Across all enrolled subjects, the mean age was 32.8 years
(range 18–55 years), and the mean body mass index was
27.78 kg/m
2
. There were 41 (82%) male and 9 (18%)
female subjects.
Of the 22 subjects included in the per-protocol popula-
tion, the majority were white (16 of 22 subjects; 72.7%),
and other subjects (6 of 22 subjects; 27.3%) were black or
African American. Subjects in the per-protocol population
had a mean age of 32.8 years (range 18–55 years), and had
a mean body mass index of 28.03 kg/m
2
. There were 21
male subjects (95.5%) and 1 female subject (4.5%).
3.3 Pharmacokinetics
Mean plasma pharmacokinetic parameters of oral parac-
etamol administered before, during, and after morphine
administration are presented in Table 1.
Observed paracetamol peak concentration (C
max
) and
area under the plasma concentration-time curve over the
6-h dosing interval (AUC
0–6
) for paracetamol following
oral administration were reduced when co-administered
with morphine, and increased after morphine was discon-
tinued. The mean AUC
0–6
was decreased from 31.00 lgh/
mL after the second dose (28.51 lgh/mL) and third dose
(25.31 lgh/mL) of oral paracetamol, and then sharply
increased following the fourth dose (52.38 lgh/mL),
resulting in statistically significant increases between
fourth and first dose (p\0.001), and fourth and third dose
(p=0.004). The mean C
max
followed a similar trend,
decreasing from 11.6 lg/mL after the first dose to 7.29 lg/
mL after the second dose and 7.25 lg/mL after the third
dose of paracetamol, and then increasing significantly after
the fourth dose (13.5 lg/mL, p=0.019, when compared
with the third dose).
Time to peak plasma concentration (T
max
) for oral
paracetamol was prolonged during and after co-adminis-
tration of morphine. Significantly prolonged mean T
max
(i.e., delayed C
max
) values were also observed after the
fourth (2.84 h, p=0.031) dose of oral paracetamol when
compared to the first dose (1.48 h).
Concomitant use of morphine also resulted in greater
variability in orally administered paracetamol exposures,
plasma concentrations, and T
max
(Table 1; Figs. 1and 2a).
The SDs of the pharmacokinetic parameters, AUC
0–6
,
C
max
, plasma concentration at 6 h (C
6
), and T
max
, after the
third and fourth doses of oral paracetamol, in most cases,
were greater than those after the first and second doses,
indicating greater inter-subject variability as a result of
interaction with co-administered morphine (Table 1).
Additionally, the concentration of the drug that produces
50% of the maximal effect (EC
50
), which for paracetamol
is purported to be in the range of 10–15 lg/mL [16,17],
was rarely reached following oral administration (Fig. 3a).
In contrast, the values of all critical pharmacokinetic
parameters after each intravenous dose of paracetamol,
administered before, during, and after morphine adminis-
tration, were found to be very similar (Fig. 2b). Further-
more, following intravenous administration of paracetamol,
EC
50
was reliably met in subjects following each dose
(Fig. 3b).
Mean plasma pharmacokinetic parameters of intra-
venous paracetamol are presented in Table 2. Because no
blood samples were collected at 0.25 h after intravenous
paracetamol, doses 2 and 3, C
max
and T
max
values reflect
the lack of sample collection before 0.25 h. Concentrations
at 0.5 h after each dose were measured, which were com-
parable across all four doses of paracetamol. Therefore,
C
0.5
is more relevant in representing the early phase of the
paracetamol concentrations when inter-dose comparison is
needed. Overall, the values of all critical pharmacokinetic
parameters after each intravenous dose of paracetamol
were found to be similar. The AUC
0–6
, AUC
0.5–6
,C
0.5
, and
C
6
for doses 2, 3, and 4 are within the bioequivalence limits
compared to the first dose but demonstrated a trend toward
small increases with each additional dose. This is consis-
tent with drug accumulation as expected in multi-dose
paracetamol administration.
3.4 Safety
Overall, 39 subjects (78.0%) experienced at least 1 TEAE,
all of which were mild or moderate in severity. The most
frequently reported TEAEs (experienced by C10% of
subjects) were vomiting (26 subjects, 52.0%), nausea
(22 subjects, 44.0%), dizziness (8 subjects, 16.0%), and
somnolence (5 subjects, 10.0%). No other TEAEs were
reported in more than 4 subjects. Although statistical
comparisons were not performed, there were fewer subjects
with TEAEs after receiving intravenous paracetamol
(73.9%) than after receiving oral paracetamol (81.5%).
There were fewer subjects who experienced the TEAE of
vomiting after receiving intravenous paracetamol (47.8%)
than after receiving oral paracetamol (55.6%). A total of 26
subjects discontinued the study due to emesis. Ten of the
26 subjects discontinued due to emesis after receiving their
first dose of morphine following their second paracetamol
R. B. Raffa et al.
dose and the remaining 16 subjects discontinued after the
second dose of morphine. Of the 10 subjects who discon-
tinued due to emesis after receiving their first dose of
morphine, 8 had previously received two doses of oral
paracetamol and 2 had received intravenous paracetamol
doses.
4 Discussion
Overall, this study demonstrated a substantial impact on the
pharmacokinetic profile of oral paracetamol, but not
intravenous paracetamol, when co-administered with
morphine. Greater pharmacokinetic variability was seen
following administration of paracetamol orally compared
to intravenously, particularly after subjects were exposed to
morphine. This variability could be attributed multiple
factors: (1) much of the inter-individual inherent physio-
logic variability could be contributing to higher variance in
absorption, (2) the uptake of paracetamol from the small
intestine is much faster than from the stomach due to the
greater surface area. An important consequence was that
the absorption would be determined by the rate at which
the drug was transferred from the stomach to the site of
rapid absorption in the upper small intestine, (3) absorption
also involved the process of dissolution from an orally
administered solid dosage form and either the rate of transit
from stomach or dissolution could be rate-limiting. Head-
ing and colleagues [18] found that rapid gastric emptying
in 14 convalescent patients was associated with the early
appearance of high peak plasma paracetamol concentra-
tions, whereas peak concentrations were low and appeared
late when gastric emptying was slow.
Observed peak paracetamol concentration (C
max
) and
area under the plasma concentration-time curve over the
6-h dosing interval (AUC
0–6
) were reduced in this study
when oral paracetamol was co-administered with mor-
phine, followed by an abruptly increased C
max
and AUC
0–6
upon the discontinuation of morphine, indicating the
absorption of the accumulated paracetamol entering the
small intestine after the effect of morphine dissipated.
Noticeable changes were also observed for time to reach
maximum drug concentration (T
max
) after the 3rd and 4th
doses. Variability in C
max
, AUC
0–6
,C
6
(paracetamol con-
centration before the following dosing time), as well as
T
max
around 3rd dose was significantly higher than that of
first two doses, indicating the impact of morphine on oral
paracetamol absorption varies substantially among indi-
vidual subjects. In contrast, intravenous paracetamol
demonstrated more predictable pharmacokinetics in the
setting of concomitant opioid use.
Table 1 Plasma pharmacokinetic parameters of oral paracetamol
Parameter Paracetamol dose
a
pvalue
c
First (before
b
) Second (during
b
) Third (during
b
) Fourth (after
b
)
n11 11 11 11
AUC
0–6
(lgh/mL)
d
Mean (SD) 31.00 (5.11) 28.51 (5.96) 25.31 (11.59) 52.38 (13.48) \0.001
CV% 16.5% 20.9% 45.8% 25.7%
AUC
0–18
(lgh/mL) Mean (SD) 82.50 (23.28)
C
max
(lg/mL) Mean (SD) 11.6 (4.11) 7.29 (1.82) 7.25 (3.95) 13.5 (3.31) 0.188
CV% 35.5% 25.0% 54.5% 24.6%
C
6
(lg/mL) Mean (SD) 2.93 (0.633) 3.71 (0.694) 4.83 (1.97) 6.83 (2.22) \0.001
CV% 21.6% 18.7% 40.8% 32.5%
T
max
(h) Mean (SD) 1.48 (0.61) 1.64 (0.78) 3.26 (2.30) 2.84 (1.05) 0.031
CV% 40.9% 47.5% 70.5% 37.0%
K
el
(/h) Mean (SD) 0.1904 (0.0171)
t
1/2
(h) Mean (SD) 3.67 (0.33)
CV coefficient of variation, AUC area under the plasma concentration-time curve, C
max
peak plasma concentration, C
6
plasma concentration at 6
h, T
max
time to peak plasma concentration, K
el
elimination rate constant, t
1/2
plasma half-life, SD standard deviation
a
Treatment A: first paracetamol dose: 1000 mg oral paracetamol (2 9500 mg tablets) and an intravenous infusion of saline at hour 0 (before
morphine). Second and third paracetamol doses: 1000 mg oral paracetamol (2 9500 mg tablets) and an intravenous infusion of saline at hours 6
and 12 (during morphine). Fourth paracetamol dose: 1000 mg oral paracetamol (2 9500 mg tablets) and an intravenous infusion of saline at hour
18 (after morphine)
b
Before, during, or after are relative to morphine administration
c
Treatment comparison: fourth/first paracetamol dose
d
AUC following each dose of paracetamol
PK of Oral and IV Paracetamol When Co-administered with IV Morphine
In this study, only two 0.125 mg/kg doses of morphine
were given, whereas in clinical practice, patients may need
more than two doses, which could have an even greater
impact on gastric emptying and gut motility, resulting in
greater accumulation of paracetamol following repeat oral
dose administration. Once gastric function is restored upon
the discontinuation of morphine, there is an abrupt release
of unabsorbed paracetamol that enters the small intestine
[19]. We observed a resultant approximate doubling in
AUC (after the 4th dose of oral paracetamol), which may
produce significant changes in paracetamol metabolism, as
common pathways such as glucuronidation/sulfation get
Fig. 1 Individual plasma concentration-time profiles of oral parac-
etamol showing large inter-subject variability. Subjects received
Treatment A, i.e., 4 repeat doses of 1000 mg oral paracetamol (2 9
500 mg tablets) and an intravenous infusion of saline every 6 h [hours
0, 6, 12, and 18 (blue arrows)], and 2 infusions of intravenous
morphine (0.125 mg/kg) at hours 6 and 12 (black arrows)
R. B. Raffa et al.
saturated and more drug is converted into the free radical
metabolite N-acetyl-p-benzoquinone imine (NAPQI),
which binds to and causes death of hepatocytes.
In general, the pharmacokinetic profile of paracetamol
following oral administration is known to exhibit consid-
erable inter-subject variability due to differences in normal
physiologic factors such as gastrointestinal movement [18].
The plasma concentrations of paracetamol and its absorp-
tion are apparently related to the rate of gastric emptying
since faster gastric emptying is associated with the rapid
appearance of high peak plasma concentrations, while the
peaks occur late and are lower in patients with delayed
gastric emptying. Gastric emptying likely influences
paracetamol absorption directly by controlling the rate at
which the drug is delivered to the small intestine. Consis-
tent with the high inter-subject variability in pharmacoki-
netic profiles seen in the current study in subjects who
received oral paracetamol, individual variation in the rate
of drug absorption may be due largely to differences in the
rate of gastric emptying [18]. Furthermore, concomitant
use of morphine in the current study introduced even
greater variability in orally administered paracetamol
exposures, plasma concentrations, and T
max
.
The interaction between morphine and orally adminis-
tered paracetamol could be even more pronounced in
postsurgical patients where factors such as concomitant
medications and surgical trauma may further impair gastric
function. This may result in inadequate pain control from
orally administered paracetamol during opioid co-treat-
ment and subsequent absorption of accumulated paraceta-
mol upon cessation of opioid treatment.
The results from this study are consistent with the
pharmacokinetic results of the study by Singla et al 2012
[14]. With respect to efficacy, plasma and CSF levels need
Fig. 2 Predicted (solid blue) and observed (dashed red) pharmacoki-
netic profiles for aoral and bintravenous paracetamol. Subjects
received Treatment A, i.e., 4 repeat doses of 1000 mg oral
paracetamol (2 9500 mg tablets) and an intravenous infusion of
saline every 6 h (hours 0, 6, 12, and 18), and 2 infusions of
intravenous morphine (0.125 mg/kg) at hours 6 and 12 Fig. 3 Paracetamol concentration-time profiles following aoral and
bintravenous paracetamol administration
PK of Oral and IV Paracetamol When Co-administered with IV Morphine
to be considered, though it is difficult to correlate parac-
etamol CSF levels with efficacy, —i.e., paracetamol has a
central effect but concentrations in CSF are not predictive
of (i.e., linearly related to) efficacy [20]. This is a limitation
of the study by Singla et al [14]. The ‘effect compartment’’
is unknown and could be multiple locations (e.g., brain and
spinal cord) [21]. Paracetamol conversion to the active
metabolite AM404 may result in active analgesia [22].
Low plasma concentrations following oral administra-
tion of paracetamol could impact efficacy relative to the
intravenous route of administration. Plasma C
max
may be a
better predictor of efficacy than AUC, in that the passive
diffusion of paracetamol into the CNS is highly dependent
on the concentration gradient across the blood brain barrier
[14]. Post-operative intravenous paracetamol previously
has been demonstrated to provide faster onset of analgesia
than a similar dose given orally [23] and, more recently,
that only two-thirds of patients given an oral dose of 1 g
paracetamol preoperatively achieved therapeutic plasma
concentrations at any point compared to 96% in those
given an intravenous dose of 1 g preoperatively [24].
In the context of multimodal analgesia, when choosing
medications to administer, increased efficacy, decreased
adverse effects, and opioid reduction are the primary
objectives [25]. Therefore, selection of non-opioids is
imperative, and route of administration should be factored
in, given opioid impact on gut motility, which impacts the
pharmacokinetics of agents administered orally; substantial
inter-subject variability could be the result of morphine’s
effect, or first- pass metabolism, or both.
Results of this study suggest intravenous paracetamol is
a better choice than oral paracetamol in patients receiving
concomitant opioids until normal gut function can be
demonstrated. A future study should evaluate the extent of
the ‘burst effect’ (i.e., the abrupt release of accumulated
paracetamol at the end of morphine-mediated GI inhibition
following oral administration), to evaluate both pharma-
cokinetics and safety (in particular, the impact on liver
enzyme levels). The study should also aim to measure
metabolites to determine the extent of NAPQI formation
following the burst effect.
5 Conclusion
This study demonstrated that morphine co-administration
significantly impacted the pharmacokinetics of oral
paracetamol, but not intravenous paracetamol, by
Table 2 Plasma pharmacokinetic parameters of intravenous paracetamol
Parameter Paracetamol dose
a
pvalue
c
First (before
b
) Second (during
b
) Third (during
b
) Fourth (after
b
)
n11 11 11 11
AUC
0–6
(lgh/mL)
d
Mean (SD) 42.56 (3.94) 44.37 (4.46) 43.59 (4.21) 49.05 (3.95) \0.001
CV% 9.2% 10.1% 9.7% 8.1%
AUC
0–18
(lgh/mL) Mean (SD) 63.58 (6.74)
C
max
(lg/mL Mean (SD) 22.6 (3.83) 17.0
e
(1.48) 17.5
e
(1.93) 28.5 (4.31) \0.001
CV% 17.0% 8.7% 11.0% 15.1%
C
0.5
(lg/mL) Mean (SD) 16.1 (1.41) 17.0 (1.48) 17.5 (1.93) 17.2 (1.60) 0.1054
C
6
(lg/mL) Mean (SD) 2.53 (0.659) 2.95 (0.621) 2.78 (0.544) 2.88 (0.616) 0.0465
CV% 24.7% 20.0% 18.8% 20.3%
T
max
(h) Mean (SD) 0.25 (0.01) 0.50 (0.00) 0.51 (0.02) 0.25 (0.01)
CV% 2.0% 0.0% 4.0% 4.0%
K
el
(/h) Mean (SD) 0.1704 (0.0193)
t
1/2
(h) Mean (SD) 4.11 (0.46)
AUC area under the plasma concentration-time curve, C
max
peak plasma concentration, C
6
plasma concentration at 6 h, T
max
time to peak plasma
concentration, K
el
elimination rate constant, t
1/2
plasma half-life, SD standard deviation
a
Treatment B: first paracetamol dose: intravenous infusion of paracetamol (1000 mg/100 mL) and 2 placebo tablets hour 0 (before morphine).
Second and third paracetamol doses: intravenous infusion of paracetamol (1000 mg/100 mL) and 2 placebo tablets at hours 6 and 12 (during
morphine). Fourth paracetamol dose: intravenous infusion of paracetamol (1000 mg/100 mL) and 2 placebo tablets at hour 18 (after morphine)
b
Before, during, or after are relative to morphine administration
c
Treatment comparison: fourth/first paracetamol dose
d
AUC following each dose of paracetamol
e
Concentration at first measured time point (0.5 h) post-dose
R. B. Raffa et al.
significantly reducing/delaying its absorption and substan-
tially increasing the inter-individual pharmacokinetic
variability. The abrupt release of the accumulated parac-
etamol at the end of morphine-mediated gastrointestinal
inhibition following oral administration of paracetamol is a
safety concern, particularly given that additional postsur-
gical patient factors may impact gastric function. Intra-
venous paracetamol thus provides a better option for the
management of postoperative pain in the context of mul-
timodal analgesia.
Compliance with Ethical Standards
Funding This study was funded by Mallinckrodt Pharmaceuticals.
These data were presented in part at the 15th Annual Pain Medicine
Meeting of the American Society of Regional Anesthesia and Pain
Medicine (ASRA), November 17–19, 2016, San Deigo, California.
Author contribution Michael G. Baker, PhD of Samorn Bio-
sciences, Inc., provided assistance with writing/formatting/proof
reading collation of the author comments and this assistance was
funded by Mallinckrodt Pharmaceuticals.
Conflict of interest Robert B. Raffa is a former employee of Johnson
& Johnson (1986–1996); he has received basic science laboratory
analgesics research funding from pharmaceutical companies; he
consults/presents on analgesics for several pharmaceutical companies,
including Mallinckrodt, but he receives no royalties for sales of any
products. Jayne Pawasauskas is a consultant to Mallinckrodt Phar-
maceuticals Inc., Joseph V Pergolizzi Jr. is a consultant/ speaker and/
or researcher for: Inspirion, Mallinckrodt, Collegium, Purdue Pharma
LLP, Grunenthal GmhB, Inspirion, ENDO Pharmaceuticals Iroko,
DepoMed and Mundipharma. Krishna Devarakonda, Jia Lu, Yin
Chen, Sutan Wu, Lawrence Hill, Brant Jarrett, and Randi Fain are
employees of Mallinckrodt Pharmaceuticals Inc.
Ethical approval Prior to initiation of this single-center study, the
protocol was reviewed and approved by the institutional review board
IntegReview, Austin, Texas. All procedures performed in studies
involving human participants were in accordance with the ethical
standards of the institutional and/or national research committee and
with the 1964 Helsinki declaration and its later amendments or
comparable ethical standards.
Informed consent Informed consent was obtained from all individ-
ual participants included in the study. Additional informed consent
was obtained from all individual participants for whom identifying
information is included in this article.
Open Access This article is distributed under the terms of the
Creative Commons Attribution-NonCommercial 4.0 International
License (http://creativecommons.org/licenses/by-nc/4.0/), which per-
mits any noncommercial use, distribution, and reproduction in any
medium, provided you give appropriate credit to the original
author(s) and the source, provide a link to the Creative Commons
license, and indicate if changes were made.
References
1. ASA Task Force on Acute Pain Management. Practice guidelines
for acute pain management in the perioperative setting: an
updated report by the American Society of Anesthesiologists
Task Force on Acute Pain Management. Anesthesiology.
2012;116:248–73.
2. Pasero C. Opioid-induced sedation and respiratory depression:
evidence-based monitoring guidelines. J Perianesth Nurs.
2012;27(3):208–11.
3. American Society of Anesthesiologists Task Force on Acute. Pain
Management. Practice guidelines for acute pain management in
the perioperative setting: an updated report by the American
Society of Anesthesiologists Task Force on Acute Pain Man-
agement. Anesthesiology. 2004;100:1573–81.
4. Jarzyna D, Jungquist CR, Pasero C, et al. American Society of
Pain Management Nursing guidelines on monitoring for opioid-
induced sedation and respiratory depression. Pain Manag Nurs.
2011;12(3):118–45.
5. Wells N, Pasero C, McCaffery M. Improving the quality of care
through pain assessment and management. In: Hughes RG, ed.
Patient safety and quality: an evidence-based handbook for nur-
ses. AHRQ Publication No. 08-0043. Rockville, MD: Agency for
Healthcare Research and Quality; March 2008.
6. The American Geriatrics Society. Pain management in the
elderly. http://www.americangeriatrics.org/gsr/anesthesiology/
pain_management.pdf. Accessed 10 Sep 2014.
7. Barr JU, Fraser GL, Puntillo K, et al. Clinical practice guidelines
for the management of pain, agitation, and delirium in adult
patients in the intensive care unit. Crit Care Med.
2013;41(1):263–306.
8. American Academy of Orthopaedic Surgeons. Management of
hip fractures in the elderly: evidence-based clinical practice
guideline. September 5, 2014. http://www.aaos.org/Research/
guidelines/HipFxGuideline_rev.pdf. Accessed 23 Mar 2015.
9. Feldman LS, Delaney CP, Ljungqvist O, Carli F. The SAGES/
ERAS
Society manual of enhanced recovery programs for
gastrointestinal surgery. New York: Springer; 2015.
10. Jibril F, Sharaby S, Mohamed A, Wilby KJ. Intravenous versus
oral acetaminophen for pain: systematic review of current evi-
dence to support clinical decision-making. Can J Hosp Pharm.
2015;68(3):238–47.
11. Chou R, Gordon DB, de Leon-Casasola OA, et al. Management
of postoperative pain: a clinical practice guideline from the
American Pain Society, the American Society of Regional
Anesthesia and Pain Medicine, and the American Society of
Anesthesiologists’ Committee on Regional Anesthesia, Executive
Committee, and Administrative Council. Pain.
2016;17(2):131–57.
12. Kennedy JM, Tyers NM, Davey AK. The influence of morphine
on the absorption of paracetamol from various formulations in
subjects in the supine position, as assessed by TDx measurement
of salivary paracetamol concentrations. J Pharm Pharmacol.
2003;55(10):1345–50.
13. Sinatra RS, Jahr JS, Reynolds LW, Viscusi ER, Groudine SB,
Payen-Champenois C. Efficacy and safety of single and repeated
administration of 1-gram intravenous acetaminophen injection
(paracetamol) for pain management after major orthopedic sur-
gery. Anesthesiology. 2005;102(4):822–31.
14. Singla NK, Parulan C, Samson R, et al. Plasma and cerebrospinal
fluid pharmacokinetic parameters after single-dose administration
of intravenous, oral, or rectal acetaminophen. Pain Pract.
2012;12(7):523–32.
15. Ameer B, Divoll M, Abernethy DR, Greenblatt DJ, Shargel L.
Absolute and relative bioavailability of oral acetaminophen
preparations. J Pharm Sci. 1983;72(8):955–8.
16. Anderson BJ, Holford NHG, Woollard GA, Kanagasundaram S,
Mahadevan M. Perioperative pharmacodynamics of acet-
aminophen analgesia in children. Anesthesiology.
1999;90(2):411–21.
PK of Oral and IV Paracetamol When Co-administered with IV Morphine
17. Gelotte CK. Cross-study pharmacokinetic and pharmacodynamic
modeling of acetaminophen: comparison of Tylenol
extended
relief caplets with regular-strength Tylenol
caplets. Fort
Washington, PA: McNeil Consumer & Specialty Pharmaceuti-
cals; 1995. Submitted to NDA 19-872 as Phase IV Commitment.
Cited in McNeil Consumer Healthcare, a Division of McNeil-
PPC, Inc. (a Johnson & Johnson Company), 2002 Submission to
the Non-Prescription Drug Advisory Committee.
18. Heading RC, Nimmo J, Prescott LF, Tothill P. The dependence of
paracetamol absorption on the rate of gastric emptying. Br J
Pharmacol. 1973;47:415–21.
19. Srinivas NR. Acetaminophen absorption kinetics in altered gas-
tric emptying: establishing a relevant pharmacokinetic surrogate
using published data. J Pain Palliat Care Pharmacother.
2015;29(2):115–9.
20. Anderson BJ, Holford NG, Woollard GA, Chan PS. Paracetamol
plasma and cerebrospinal fluid pharmacokinetics in children. Br J
Pharmacol. 1998;46(3):237–43.
21. Toussaint K, Yang XC, Zielinski MA, et al. What do we (not)
know about how paracetamol (acetaminophen) works? J Clin
Pharm Ther. 2010;35(6):617–38.
22. Mallet C, Barrie
`re DA, Ermund A, et al. TRPV1 in brain is
involved in acetaminophen-induced antinociception. PLoS One.
2010;5(9):e12748.
23. Møller PL, Sindet-Pedersen S, Petersen CT, Juhl GI, Dillen-
schneider A, Skoglund LA. Onset of acetaminophen analgesia:
comparison of oral and intravenous routes after third molar sur-
gery. Br J Anaesth. 2005;94(5):642–8.
24. van der Westhuizen J, Kuo PY, Reed PW, Holder K. Randomised
controlled trial comparing oral and intravenous paracetamol
(acetaminophen) plasma levels when given as preoperative
analgesia. Anaesth Intensive Care. 2011;39:242–6.
25. Raffa RB, Clark-Vetri R, Tallarida RJ, Wertheimer AI. Combi-
nation strategies for pain management. Expert Opin Pharma-
cother. 2003;4(10):1697–708.
R. B. Raffa et al.
... Also, the IM and SC routes are commonly employed for the treatment of acute pain; however, after surgery, they should not be considered first-line options since the poor perfusion and delayed distribution of the drugs is a risky possibility and can result in the occurrence of inadequate analgesia or the late occurrence of side effects [20]. Although IM and SC formulations may be appropriate for patients without IV access and with the oral route unavailable, IV administration is believed to provide the fastest relief [3,4,[22][23][24][25]. However, when compared to other routes of administration, the costs related to the use of IV forms are higher and with no benefits in terms of pain control [20]. ...
... Also, intravenously, the maximum plasma concentration of paracetamol is greater and achieved earlier. Since paracetamol enters the central nervous system (CNS) through passive diffusion, the gradient concentration between the plasma and CNS tissues is a very important factor for its infiltration into the CNS [23,24]. ...
... In the previously cited meta-analysis [93], only some clinical studies were selected where an ibuprofen/paracetamol combination was assessed through the administration of FDCs. Nonetheless, there are results of many other clinical trials in the literature where the combination of ibuprofen and paracetamol was performed either with an FDC or separately [23,24,[95][96][97][98][99][100][101][102][103][104][105][106][107][108][109][110]. In contrast with the results obtained by Abushanab D. & Al-Badriyeh D. [93], where the administration of ibuprofen/paracetamol FDCs seems to provide generally improved analgesia, no significant or clear advantages of an ibuprofen/paracetamol combination over ibuprofen and paracetamol in monotherapy are reported in the literature [104,[106][107][108][109]. ...
Article
Full-text available
The combination of non-steroidal anti-inflammatory drugs (NSAIDs) with non-opioid analgesics is common in clinical practice for the treatment of acute pain conditions like post-operative and post-traumatic pain. Besides that, and despite the very satisfactory results achieved by oral analgesics, parenteral analgesia is a key tool in the treatment of painful conditions when the enteral routes are not convenient. The parenteral ready-to-use fixed-dose combinations of NSAIDs with non-opioid analgesics like paracetamol or metamizole could play a central role in the treatment of painful conditions since they are able to gather into only one formulation the advantages of multimodal and parenteral analgesia. Surprisingly, only very recently a parenteral ready-to-use fixed-dose combination of Ibuprofen/Paracetamol was launched to the market. This review aims to review the current availability of NSAID-based Parenteral Fixed-dose combinations with paracetamol and metamizole in the European and American markets, and how the combination of such drugs could play a central role in a multimodal analgesia strategy. Also, it is explored how the parenteral formulations of NSAIDs, paracetamol, and metamizole could serve as starting elements for the development of new parenteral ready-to-use fixed-dose combinations. With this review, we concluded that, despite the well-recognized utility of combining NSAIDs with non-opioid analgesics, there are in the literature several randomized clinical trial studies unable to demonstrate clear advantages concerning their efficacy and safety. In the future, it is deemed necessary to perform clinical trials specifically designed to assess the efficacy and safety of these fixed-dose formulations to generate solid evidence of their clinical advantages.
... we have used this model system of early human brain development and investigated the effects of paracetamol exposures on transcriptional and epigenetic regulation. Paracetamol doses were selected to reflect therapeutic maternal doses and foetal in utero exposures (24)(25)(26). The time and dose effect of paracetamol exposure during differentiation was inferred by integrating multiple omics methods (bulk RNA-seq, bulk DNAm, single-cell RNA-seq and ATAC-seq). ...
... Culture media were replaced daily, and the cells were exposed to 100 or 200 μ M paracetamol during differentiation from Day 1 and onwards. These concentrations have been documented to be in the range of therapeutic plasma concentrations (24)(25)(26). All culture media (control and paracetamol-supplemented) were replaced daily, and unexposed (control) and paracetamol-exposed cells were harvested for downstream analyses on Day 7 and 20. ...
... Neuronal differentiation of hESCs and exposure to paracetamol hESCs HS360 were differentiated, as described by Samara et al, 2022 (23), in two separate time-course experiments, to investigate the in vitro effects of paracetamol exposure in physiologically concentrations relevant to long-term exposure in vivo. Since paracetamol crosses the placenta and the blood-brain barrier and has an insignificant plasma protein binding, maternal plasma/serum and cord blood concentrations can be used as estimates for the amount of paracetamol that reaches the developing foetal brain (104)(105)(106)(107). Concentrations of 100 μ M and 200 μ M paracetamol (Merck; A7085) were chosen as range for physiological plasma peak concentration in vivo, corresponding to one intermediate and one high peak plasma concentration (24)(25)(26). Thus, cells undergoing neuronal differentiation were exposed to media changes of medium only (control) or medium containing 100 or 200 μ M paracetamol from Day 1 to Day 20. ...
Preprint
Full-text available
Prenatal paracetamol exposure has been associated with neurodevelopmental outcomes in childhood. Pharmacoepigenetic studies show differences in cord blood DNA methylation between paracetamol exposed and unexposed neonates. However, causal implications and impact of long-term prenatal long-term paracetamol exposure on brain development remain unclear. Using a multi-omics approach, we investigated the effects of paracetamol on a model of early human brain development. We exposed human embryonic stem cells undergoing in vitro neuronal differentiation to daily media changes with paracetamol concentrations corresponding to maternal therapeutic doses. Single-cell RNA-seq and ATAC-seq integration identified paracetamol-induced chromatin-opening changes linked to gene expression. Differentially methylated and/or expressed genes were involved in signalling, neurotransmission, and cell fate-determination trajectories. Some genes involved in neuronal injury and development-specific pathways, such as KCNE3 , overlapped with differentially methylated genes previously identified in cord blood associated with prenatal paracetamol exposure. Our data suggest that paracetamol may play a causal role in impaired neurodevelopment. Graphical Abstract
... Paracetamol doses were selected to reflect therapeutic maternal doses and fetal in utero exposures. [31][32][33] By integrating multiple omics methods (bulk RNA-seq, bulk DNAm, single-cell RNA-seq and ATAC-seq, Table 1) we observed time and dose effects of paracetamol exposure during neuronal differentiation. ...
... These concentrations have been documented to be in the range of therapeutic plasma concentrations. [31][32][33] Unexposed (control) and paracetamol-exposed cells were harvested for downstream analyses on Day 7 and 20. We also harvested control cells at the onset of differentiation (hESCs; Day 0), and on the intermediate timepoint that cells were passaged (Day 13) to assess whether paracetamol exposed cells had mRNA abundance changes related to proliferation or delayed differentiation. ...
Article
Full-text available
Prenatal paracetamol exposure has been associated with neurodevelopmental outcomes in childhood. Pharmacoepigenetic studies show differences in cord blood DNA methylation between unexposed and paracetamol-exposed neonates, however, causality and impact of long-term prenatal paracetamol exposure on brain development remain unclear. Using a multi-omics approach, we investigated the ef- fects of paracetamol on an in vitro model of early human neurodevelopment. We exposed human em- bryonic stem cells undergoing neuronal differentiation with paracetamol concentrations corresponding to maternal therapeutic doses. Single-cell RNA-seq and ATAC-seq integration identified paracetamol- induced chromatin opening changes linked to gene expression. Differentially methylated and/or expressed genes were involved in neurotransmission and cell fate determination trajectories. Some genes involved in neuronal injury and development-specific pathways, such as KCNE3, overlap- ped with differentially methylated genes previously identified in cord blood associated with prenatal paracetamol exposure. Our data suggest that paracetamol may play a causal role in impaired neurodevelopment.
... It has also been shown that concomitant morphine administration delays and reduces oral acetaminophen absorption in adults while not affecting the pharmacokinetics of IV acetaminophen. 86 This is presumably because of an opioid-induced decrease in gastrointestinal motility and delayed gastric emptying. ...
Article
Acetaminophen is one of the oldest medications commonly administered in children. Its efficacy in treating fever and pain is well accepted among clinicians. However, the available evidence supporting the use of acetaminophen's different modes of administration remains relatively scarce and poorly known. This short report summarizes the available evidence and provides a framework to guide clinicians regarding a rational use of acetaminophen in children.
... During oral dosing, for example, both the rate and extent of substrate exposure are highly variable and do not exceed the responsible enzyme concentrations at therapeutic drug concentrations (for a review on CYP450., see Zanger and Schwab, 2013). Daily 400 mg oral doses of the anti-inflammatory drug acetaminophen (Mw 5 151.16 g/mol) corresponds to a plasma peak concentration of about 10 mg/L (66 mM) (Raffa et al., 2018). The most abundant hepatic responsible enzyme, CYP3A4, for the metabolism of acetaminophen has a total amount of 60 to 150 nmol/g hepatic tissue (60-150 mmol/kg liver or 90-225 mmol/ liver) (Zanger and Schwab, 2013), which exceeds drug exposure to acetaminophen at any time. ...
Article
Even though significant efforts have been spent in recent years to understand and define the determinants of in vivo potency and clearance, important pieces of information are still lacking. By introducing target turnover into the reasoning, we open up to further the understanding of central factors important to the optimization of translational dose-concentration-response predictions. We describe: i) new ('open' model) expressions of the in vivo potency and efficacy parameters which embody target turnover, binding and complex kinetics, also capturing full, partial, and inverse ago-nism, and antagonism, ii) a detailed examination of 'open' models to show what potency and efficacy parameters have in common and how they differ, and, iii) a comprehensive literature review showing that target turnover rate varies with age, species, tissue/subregion, treatment, disease state, hormonal and nutritional state, and day-night cycle. The new 'open' model expression which integrates system- and drug properties, shows that: fractional turnover rates rather than the absolute target or ligand-target complex expression determine necessary drug exposure via in vivo potency; absolute ligand-target expression determines the need of drug, based on the transduction Rho and in vivo efficacy parameters; the free enzyme concentration determines clearance and maximum metabolic rate; the fractional turnover rate determines time to equilibrium between substrate, free enzyme and complex; properties of substrate, target, and complex demonstrate non-saturable metabolic behavior at equilibrium; non-linear processes previously referred to as capacity- and time-dependent kinetics may occasionally have been disequilibria; and, the 'open' model may pinpoint why some subjects differ in their demand of drug. Significance Statement 'Understand the target turnover' is a central tenet in many translational dose-concentration-response predictions. New 'open' model expressions of in vivo potency, efficacy parameter and clearance are derived and anchored onto a comprehensive literature review showing that target turnover rate varies with age, species, tissue/subregion, treatment, disease, hormonal and nutritional state, day-night cycle, and more. Target turnover concepts will therefore significantly impact fundamental aspects of pharmacodynamics and pharmacokinetics, thereby also the basics of drug discovery, development, and optimization of clinical dosing.
... Value-added delivery systems are continuously being explored for the conveyance of drugs into the body with the aim to treat and combat the ever-increasing challenges [1]. However, the oral route has still been the priority for even newer/modified systems as it has many perks including the highest patient compliance, cost-effectiveness, no requirements for sterility, a cascade of options to construct dosage forms and the luxury of self-administration [2]. ...
Article
Full-text available
The aim of the current study was to enhance the oral bioavailability of Acyclovir (ACV) based on the papain-functionalized self-emulsifying drug delivery systems (SEDDS). The optimum control SEDDS formulation comprised of kolliphore (40%), transcutol (30%), propylene glycol (20%) and oleoyl chloride (10%). However, in the targeted SEDDS formulation, oleoyl chloride was replaced with oleoyl chloride-papain (OC-PAP) conjugate that was synthesized via an amide bond formation between the acyl halide groups of oleoyl chloride and the amino group of papain. Prior to adding in the SEDDS formulation, the newly synthesized conjugate was evaluated quantitatively by a Bradford assay that demonstrated 45 µg of papain contents per mg of the conjugate. Moreover, the conjugate formation was qualitatively confirmed through FTIR analysis and thin layer chromatography. ACV (a BCS class III drug) was incorporated into the SEDDS formulations after being hydrophobically ion paired with sodium deoxycholate, thereby making it lipophilic. The drug-loaded formulations were emulsified in the 0.1 M phosphate buffer (pH 6.8) and evaluated in vitro with respect to drug release and rabbit mucosal permeation studies. Both the formulations illustrated a very comparable drug release over a period of 4 h, afterwards, the OC-PAP-based formulation demonstrated a more sustaining effect. The extent of mucus diffusion evaluated via the silicon tube method demonstrated a 4.92-fold and a 1.46-fold higher penetration of the drug, a 3.21-fold and a 1.56-fold higher permeation through the rabbit intestinal mucus layer, and a 22.94-fold and a 2.27-fold higher retention of the drug over the intact mucosa of rabbit intestine, illustrated by OC-PAP-based nanoemulsions compared to the drug-free solution and controlled nanoemulsion, respectively. According to these in vitro results, papain-functionalized SEDDS is a promising approach for the oral delivery of ACV and many other drugs with oral bioavailability issues, however, in vivo studies in this respect have to be employed before making a comprehensive conclusion.
Article
Full-text available
Paracetamol has been recognized worldwide as a safe and effective agent for relieving pain and reducing fever in many patients. This study aimed to investigate the role of clinical pharmacist interventions in the rational and appropriate use of intravenous paracetamol in surgical patients and the impact of this rational use on hospital costs. A case-control study was conducted on 794 patients (400 in the intervention group and 394 in the control group). The appropriate and rational use of the drug was compared between baseline and post-intervention in the intervention group and between the two groups. The result showed a significant reduction in dispensed IV paracetamol vials after the pharmaceutical intervention (4,151 vials recovered by the intervention), which led to a reduction in the cost (8,302 USD reduction in the total cost). There was a significant reduction in the dose of IV paracetamol, use with or without adjunctive opioid analgesics, frequency of administration, duration of intravenous paracetamol use, daily max exceeding 4 g, and concomitant use with oral paracetamol after applying the clinical pharmacist intervention. In conclusion, the clinical pharmacist plays a vital role in various aspects of healthcare; clinical pharmacist involvement positively impacts the patient’s management plan by improving the optimal and rational use of intravenous paracetamol and decreasing hospital costs.
Article
Nonsteroidal anti‐inflammatory drugs (NSAIDs) are among the most frequently used drugs that can cause liver toxicity. The aim of this study was to integrate bioanalytical and population pharmacokinetic (PopPK) assay to rapidly screen and quantify the concentrations of NSAIDs in plasma and monitor clinical safety. A liquid chromatography–tandem mass spectrometry (LC–MS/MS) method was developed for the simultaneous quantification of acetaminophen (APAP), flurbiprofen (FLB), aspirin (ASP), and ibuprofen (IBP), four commonly used NSAIDs. The PopPK model of the signature toxicant was analyzed based on the published literature. The LC–MS/MS method was successfully validated and applied to determine NSAID concentrations in patient plasma samples. APAP, ASP, and IBP data were best fitted using a one‐compartment model, and FLB data were best fitted using a two‐compartment model. Bootstrapping and visual predictive checks suggested that a robust and reliable pharmacokinetic model was developed. A fast, simple, and sensitive LC–MS/MS method was developed and validated for determining APAP, FLB, ASP, and IBP in human plasma. Combined with the PopPK model, this method was applied to rapidly analyze the concentrations of NSAIDs in clinical samples from patients presenting to the emergency department with acute liver dysfunction and monitored NSAIDs clinical safety.
Article
Full-text available
Patients in intensive care units (ICUs) experience pain, which they describe as a significant cause of stress during treatment. It can progress to chronic pain and significantly affect the quality of life. Opioids have long been the backbone of ICU pain therapy. The consequences of their long-term use are known today, such as prolonged ICU stay and mechanical ventilation, resulting in increased treatment costs. Additionally, abstinence syndrome is a consequence of abrupt opioid withdrawal. Also, there is a risk of tolerance and hyperalgesia after prolonged opioid use. Globally, opioid dependence after hospital opioid treatment is alarming, although there is still a lack of data on its incidence after ICU. Multimodal analgesia enables comfort to the patient, opioid-sparing, and avoidance of side effects of non-opioid analgesics. Nonsteroidal anti-inflammatory drugs (NSAIDs) are a broad group of drugs recommended with paracetamol to treat mild to moderate acute postoperative pain. Although often prescribed by intensivists, their use in treating painful conditions in ICU is controversial due to the possible consequences on the organs of critically ill patients. Due to the inhibition of cyclooxygenases, NSAIDs indirectly cause vasoconstriction of the renal arteries and arterioles, leading to kidney damage. NSAIDs inhibit platelet aggregation and may predispose to bleeding. Analgesia of a critically ill patient is a important part of their treatment, however it can be challenging in certain patients. Numerous combinations of pharmacological and non-pharmacological approaches can be adapted to the patient's current characteristics.
Article
Full-text available
Unlabelled: Most patients who undergo surgical procedures experience acute postoperative pain, but evidence suggests that less than half report adequate postoperative pain relief. Many preoperative, intraoperative, and postoperative interventions and management strategies are available for reducing and managing postoperative pain. The American Pain Society, with input from the American Society of Anesthesiologists, commissioned an interdisciplinary expert panel to develop a clinical practice guideline to promote evidence-based, effective, and safer postoperative pain management in children and adults. The guideline was subsequently approved by the American Society for Regional Anesthesia. As part of the guideline development process, a systematic review was commissioned on various aspects related to various interventions and management strategies for postoperative pain. After a review of the evidence, the expert panel formulated recommendations that addressed various aspects of postoperative pain management, including preoperative education, perioperative pain management planning, use of different pharmacological and nonpharmacological modalities, organizational policies, and transition to outpatient care. The recommendations are based on the underlying premise that optimal management begins in the preoperative period with an assessment of the patient and development of a plan of care tailored to the individual and the surgical procedure involved. The panel found that evidence supports the use of multimodal regimens in many situations, although the exact components of effective multimodal care will vary depending on the patient, setting, and surgical procedure. Although these guidelines are based on a systematic review of the evidence on management of postoperative pain, the panel identified numerous research gaps. Of 32 recommendations, 4 were assessed as being supported by high-quality evidence, and 11 (in the areas of patient education and perioperative planning, patient assessment, organizational structures and policies, and transitioning to outpatient care) were made on the basis of low-quality evidence. Perspective: This guideline, on the basis of a systematic review of the evidence on postoperative pain management, provides recommendations developed by a multidisciplinary expert panel. Safe and effective postoperative pain management should be on the basis of a plan of care tailored to the individual and the surgical procedure involved, and multimodal regimens are recommended in many situations.
Article
Full-text available
As the complexity of analgesic therapies increases, priorities of care must be established to balance aggressive pain management with measures to prevent or minimize adverse events and to ensure high quality and safe care. Opioid analgesia remains the primary pharmacologic intervention for managing pain in hospitalized patients. Unintended advancing sedation and respiratory depression are two of the most serious opioid-related adverse events. Multiple factors, including opioid dosage, route of administration, duration of therapy, patient-specific factors, and desired goals of therapy, can influence the occurrence of these adverse events. Furthermore, there is an urgent need to educate all members of the health care team about the dangers and potential attributes of administration of sedating medications concomitant with opioid analgesia and the importance of initiating rational multimodal analgesic plans to help avoid adverse events. Nurses play an important role in: 1) identifying patients at risk for unintended advancing sedation and respiratory depression from opioid therapy; 2) implementing plans of care to assess and monitor patients; and 3) intervening to prevent the worsening of adverse events. Despite the frequency of opioid-induced sedation, there are no universally accepted guidelines to direct effective and safe assessment and monitoring practices for patients receiving opioid analgesia. Moreover, there is a paucity of information and no consensus about the benefits of technology-supported monitoring, such as pulse oximetry (measuring oxygen saturation) and capnography (measuring end-tidal carbon dioxide), in hospitalized patients receiving opioids for pain therapy. To date, there have not been any randomized clinical trials to establish the value of technologic monitoring in preventing adverse respiratory events. Additionally, the use of technology-supported monitoring is costly, with far-reaching implications for hospital and nursing practices. As a result, there are considerable variations in screening for risk and monitoring practices. All of these factors prompted the American Society for Pain Management Nursing to approve the formation of an expert consensus panel to examine the scientific basis and state of practice for assessment and monitoring practices for adult hospitalized patients receiving opioid analgesics for pain control and to propose recommendations for patient care, education, and systems-level changes that promote quality care and patient safety.
Article
Full-text available
Gastric absorption of oral paracetamol (acetaminophen) may be unreliable perioperatively in the starved and stressed patient. We compared plasma concentrations of parenteral paracetamol given preoperatively and oral paracetamol when given as premedication. Patients scheduled for elective ear, nose and throat surgery or orthopaedic surgery were randomised to receive either oral or intravenous paracetamol as preoperative medication. The oral dose was given 30 minutes before induction of anaesthesia and the intravenous dose given pre-induction. All patients were given a standardised anaesthetic by the same specialist anaesthetist who took blood for paracetamol concentrations 30 minutes after the first dose and then at 30 minute intervals for 240 minutes. Therapeutic concentrations of paracetamol were reached in 96% of patients who had received the drug parenterally, and 67% of patients who had received it orally. Maximum median plasma concentrations were 19 mg.l ⁻¹ (interquartile range 15 to 23 mg.l ⁻¹ ) and 13 mg.l ⁻¹ (interquartile range 0 to 18 mg.l ⁻¹ ) for the intravenous and oral group respectively. The difference between intravenous and oral groups was less marked after 150 minutes but the intravenous preparation gave higher plasma concentrations throughout the study period. It can be concluded that paracetamol gives more reliable therapeutic plasma concentrations when given intravenously.
Book
This volume presents a comprehensive, up-to-date and practical approach to creating an Enhanced Recovery Program for GI surgery. The first sections review the evidence underlying individual elements of Enhanced Recovery Programs, including evidence from laparoscopic procedures when available and pointing to evidence gaps where more research is required. These are written by experts in the field, including surgeons, anesthesiologists, nurses, and physiotherapists. The format is in the style of a narrative review, summarizing the evidence and concluding with key “take home messages” and references. Management of common complications and issues related to patient selection are also addressed. Subsequent chapters address practical concerns, including creation of a pathway team, project management and engaging administration. Experts contribute real-world examples of their pathways for a variety of procedures, including colorectal surgery, bariatric surgery, upper GI and hepatobiliary surgery, enabling the user to have a starting point for creating their own programs. The SAGES / ERAS® Society Manual of Enhanced Recovery Programs for Gastrointestinal Surgery will be of great value to fully trained surgeons, anesthesiologists, nurses and administrators interested in initiating an Enhanced Recovery Program.
Article
Intravenous (IV) acetaminophen is increasingly used around the world for pain control for a variety of indications. However, it is unclear whether IV administration offers advantages over oral administration. To identify, summarize, and critically evaluate the literature comparing analgesic efficacy, safety, and pharmacokinetics for IV and oral dosage forms of acetaminophen. A literature search of the PubMed, Embase, and International Pharmaceutical Abstracts databases was supplemented with keyword searches of Science Direct, Wiley Library Online, and Springer Link databases for the period 1948 to November 2014. The reference lists of identified studies were searched manually. Randomized controlled trials comparing IV and oral dosage forms of acetaminophen were included if they assessed an efficacy, safety, or pharmacokinetic outcome. For each study, 2 investigators independently extracted data (study design, population, interventions, follow-up, efficacy outcomes, safety outcomes, pharmacokinetic outcomes, and any other pertinent information) and completed risk-of-bias assessments. Six randomized clinical trials were included. Three of the studies reported outcomes pertaining to efficacy, 4 to safety, and 4 to pharmacokinetics. No clinically significant differences in efficacy were found between the 2 dosage forms. Safety outcomes were not reported consistently enough to allow adequate assessment. No evidence was found to suggest that increased bioavailability of the IV formulation enhances efficacy outcomes. For studies reporting clinical outcomes, the results of risk-of-bias assessments were largely unclear. For patients who can take an oral dosage form, no clear indication exists for preferential prescribing of IV acetaminophen. Decision-making must take into account the known adverse effects of each dosage form and other considerations such as convenience and cost. Future studies should assess multiple-dose regimens over longer periods for patients with common pain indications such as cancer, trauma, and surgery.
Article
Acetaminophen has been used as a tool for clinical and nonclinical experimental designs that evaluate gastric emptying because acetaminophen is not absorbed in stomach but efficiently absorbed from the small intestine. Published pharmacokinetic data of acetaminophen in subjects with normal gastric emptying vs. impaired gastric emptying (i.e., morphine treatment) were evaluated to select a key surrogate. Using Caverage (average concentration), computed from the exposure within the first hour, individual rank distribution was plotted across different studies. Caverage was highly correlated with Cmax (maximum concentration) in subjects with normal gastric emptying (R(2) = .7532) but not in those where gastric emptying was impaired (R(2) = .0213). The 50th percentile value of the distribution pattern of 1/Caverage in acetaminophen+morphine-treated group (coincided with the first shift in the slope) was considered as the cutoff point to figure out the impaired gastric emptying. The individual rank distribution plots for 1/Caverage across different studies supported similar trends in subjects with normal gastric emptying but showed a distinct distribution pattern in the cohort of impaired gastric emptying. Caverage, calculated within the first hour of dosing of acetaminophen (average concentration at 0-1 hour, C0-1havg), can be used as a key surrogate to distinguish the effects of gastric emptying on the absorption of acetaminophen. A 4 μg/mL C0-1havg of acetaminophen (dose: 1.5 g) may be used as cutoff point in future clinical investigations of acetaminophen to clarify the role of gastric emptying.
Article
Objective: To revise the "Clinical Practice Guidelines for the Sustained Use of Sedatives and Analgesics in the Critically Ill Adult" published in Critical Care Medicine in 2002. Methods: The American College of Critical Care Medicine assembled a 20-person, multidisciplinary, multi-institutional task force with expertise in guideline development, pain, agitation and sedation, delirium management, and associated outcomes in adult critically ill patients. The task force, divided into four subcommittees, collaborated over 6 yr in person, via teleconferences, and via electronic communication. Subcommittees were responsible for developing relevant clinical questions, using the Grading of Recommendations Assessment, Development and Evaluation method (http://www.gradeworkinggroup.org) to review, evaluate, and summarize the literature, and to develop clinical statements (descriptive) and recommendations (actionable). With the help of a professional librarian and Refworks database software, they developed a Web-based electronic database of over 19,000 references extracted from eight clinical search engines, related to pain and analgesia, agitation and sedation, delirium, and related clinical outcomes in adult ICU patients. The group also used psychometric analyses to evaluate and compare pain, agitation/sedation, and delirium assessment tools. All task force members were allowed to review the literature supporting each statement and recommendation and provided feedback to the subcommittees. Group consensus was achieved for all statements and recommendations using the nominal group technique and the modified Delphi method, with anonymous voting by all task force members using E-Survey (http://www.esurvey.com). All voting was completed in December 2010. Relevant studies published after this date and prior to publication of these guidelines were referenced in the text. The quality of evidence for each statement and recommendation was ranked as high (A), moderate (B), or low/very low (C). The strength of recommendations was ranked as strong (1) or weak (2), and either in favor of (+) or against (-) an intervention. A strong recommendation (either for or against) indicated that the intervention's desirable effects either clearly outweighed its undesirable effects (risks, burdens, and costs) or it did not. For all strong recommendations, the phrase "We recommend …" is used throughout. A weak recommendation, either for or against an intervention, indicated that the trade-off between desirable and undesirable effects was less clear. For all weak recommendations, the phrase "We suggest …" is used throughout. In the absence of sufficient evidence, or when group consensus could not be achieved, no recommendation (0) was made. Consensus based on expert opinion was not used as a substitute for a lack of evidence. A consistent method for addressing potential conflict of interest was followed if task force members were coauthors of related research. The development of this guideline was independent of any industry funding. Conclusion: These guidelines provide a roadmap for developing integrated, evidence-based, and patient-centered protocols for preventing and treating pain, agitation, and delirium in critically ill patients.
Article
Background: This is the first study to compare plasma and cerebrospinal fluid (CSF) pharmacokinetics of intravenous (IV), oral (PO), or rectal (PR) formulations of acetaminophen. Methods: Healthy male subjects (N = 6) were randomized to receive a single dose of IV (OFIRMEV®; Cadence) 1,000 mg (15 minute infusion), PO (2 Tylenol® 500 mg caplets; McNeil Consumer Healthcare), or PR acetaminophen (2 Feverall® 650 mg suppositories; Actavis) with a 1-day washout period between doses. The 1,300 mg PR concentrations were standardized to 1,000 mg. Acetaminophen plasma and CSF levels were obtained at T0, 0.25, 0.5, 0.75, 1, 2, 3, 4, and 6 hours. Results: IV acetaminophen showed earlier and higher plasma and CSF levels compared with PO or PR administration. CSF bioavailability over 6 hours (AUC0–6) for IV, PO, and PR 1 g was 24.9, 14.2, and 10.3 μg·h/mL, respectively. No treatment-related adverse events were reported. One subject was replaced because of premature failure of his lumbar spinal catheter. The mean CSF level in the IV group was similar to plasma from 3 to 4 hours and higher from 4 hours on. Absorption phase, variability in plasma, and CSF were greater in PO and PR groups than variability with IV administration. Conclusions: These results demonstrate that earlier and greater CSF penetration occurs as a result of the earlier and higher plasma peak with IV administration compared with PO or PR.