You are on page 1of 14

Perioperative Medicine

ABSTRACT
Background: Numerous pharmacokinetic models have been published
aiming at more accurate and safer dosing of dexmedetomidine. The vast

Dexmedetomidine majority of the developed models underpredict the measured plasma concen-
trations with respect to the target concentration, especially at plasma concen-

Clearance Decreases with


trations higher than those used in the original studies. The aim of this article
was to develop a dexmedetomidine pharmacokinetic model in healthy adults

Increasing Drug Exposure:


emphasizing linear versus nonlinear kinetics.
Methods: The data of two previously published clinical trials with step-

Implications for Current wise increasing dexmedetomidine target-controlled infusion were pooled to
build a pharmacokinetic model using the NONMEM software package (ICON

Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/136/2/279/530456/20220200.0-00013.pdf by guest on 10 May 2022


Dosing Regimens and Development Solutions, USA). Data from 48 healthy subjects, included in a
stratified manner, were utilized to build the model.

Target-controlled Infusion Results: A three-compartment mamillary model with nonlinear elimination


from the central compartment was superior to a model assuming linear phar-

Models Assuming Linear macokinetics. Covariates included in the final model were age, sex, and total
body weight. Cardiac output did not explain between-subject or within-subject

Pharmacokinetics variability in dexmedetomidine clearance. The results of a simulation study


based on the final model showed that at concentrations up to 2 ng · ml–1,
the predicted dexmedetomidine plasma concentrations were similar between
Ricardo Alvarez-Jimenez, M.D., Ph.D.,
the currently available Hannivoort model assuming linear pharmacokinetics
Maud A. S. Weerink, M.D., Laura N. Hannivoort, M.D., Ph.D.,
and the nonlinear model developed in this study. At higher simulated plasma
Hong Su, Pharm.D., Michel M. R. F. Struys, M.D., Ph.D., F.R.C.A., concentrations, exposure increased nonlinearly with target concentration due
Stephan A. Loer, M.D., Ph.D., Pieter J. Colin, Pharm.D., Ph.D. to the decreasing dexmedetomidine clearance with increasing plasma con-
Anesthesiology 2022; 136:279–92 centrations. Simulations also show that currently approved dosing regimens
in the intensive care unit may potentially lead to higher-than-expected dexme-
detomidine plasma concentrations.
EDITOR’S PERSPECTIVE Conclusions: This study developed a nonlinear three-compartment phar-
macokinetic model that accurately described dexmedetomidine plasma
What We Already Know about This Topic concentrations. Dexmedetomidine may be safely administered up to tar-
• Dexmedetomidine pharmacokinetic models underpredict the mea- get-controlled infusion targets under 2 ng · ml–1 using the Hannivoort model,
sured plasma target-controlled infusion concentrations that are which assumed linear pharmacokinetics. Consideration should be taken
higher than those used in the model validation studies during long-term administration and during an initial loading dose when fol-
• The elimination clearance of high hepatic extraction ratio drugs like lowing the dosing strategies of the current guidelines.
dexmedetomidine is determined by liver blood flow and not enzyme
activity (ANESTHESIOLOGY 2022; 136:279–92)

What This Article Tells Us That Is New

D
• The data of 48 subjects from two published pharmacokinetic stud- exmedetomidine is widely known for its anxiolytic,
ies were pooled to build a three-compartment pharmacokinetic model
sedative, and analgesic effects due to its central α2-
with nonlinear elimination clearance that successfully predicted plasma
dexmedetomidine concentrations over a wide concentration range
adrenergic agonistic properties, which provide a unique
• Cardiac output did not explain between-subject or within-subject arousable sedation profile with minimal ventilatory effects
variability in dexmedetomidine elimination clearance when used in therapeutic ranges.1,2 Nowadays, dexmedeto-
• Dexmedetomidine elimination clearance may decrease with midine is approved for short-term use in mechanically ven-
increasing plasma concentrations because it alters the liver blood tilated patients admitted to the intensive care unit (ICU) and
flow–to–cardiac output ratio in a concentration-dependent manner for periprocedural sedation. A considerable amount of effort

This article is featured in “This Month in Anesthesiology,” page A1. This article is accompanied by an editorial on p. 258. Supplemental Digital Content is available for this article.
Direct URL citations appear in the printed text and are available in both the HTML and PDF versions of this article. Links to the digital files are provided in the HTML text of this
article on the Journal’s Web site (www.anesthesiology.org). This article has a visual abstract available in the online version.
Submitted for publication October 16, 2020. Accepted for publication October 4, 2021. Published online first on December 1, 2021. From the Department of Anesthesiology, Amsterdam
University Medical Center, Amsterdam, The Netherlands (R.A.-J., S.A.L.); the Department of Anesthesiology, University Medical Center Groningen, University of Groningen, Groningen, The
Netherlands (M.A.S.W., L.N.H., H.S., M.M.R.F.S., P.J.C.); and the Department of Basic and Applied Medical Sciences, Ghent University, Ghent, Belgium (M.M.R.F.S.).
Copyright © 2021, the American Society of Anesthesiologists. All Rights Reserved. Anesthesiology 2022; 136:279–92. DOI: 10.1097/ALN.0000000000004049

ANESTHESIOLOGY, V 136 • NO 2 February 2022 279


Copyright © 2021, the American Society of Anesthesiologists. All Rights Reserved. Unauthorized reproduction of this article is prohibited.
<zdoi;. DOI: 10.1097/ALN.0000000000004049>
PERIOPERATIVE MEDICINE

has been invested to elicit a deeper understanding and char- database (NCT03143972 and NCT01879865). Written
acterization of dexmedetomidine’s pharmacology among informed consent was obtained before performing a
all ages and clinical conditions. As a result, numerous and standard medical screening, which included a thorough
heterogeneous pharmacokinetic (and pharmacodynamic) medical history and a physical examination. Exclusion cri-
models have been published.These models aim to develop a teria included a body mass index greater than 30 kg ∙ m–2
more accurate and safer dosing regime that permits a faster or less than 18 kg ∙ m–2, an age less than 18 yr or above
onset of the desired effects while diminishing the risk for 70 yr, pregnancy, or currently breastfeeding. Alcohol
undesirable cardiovascular effects.3,4 Unfortunately, the vast consumption, smoking, and use of concomitant (ille-
majority of the developed models underpredict the mea- gal) drugs were not allowed during study participation.
sured plasma concentrations with respect to the target con- Inclusion was stratified according to age and sex in both
centration, especially at higher plasma concentrations than studies. Subjects were in fasting conditions during each
those originally used in the validation studies. For exam- study period.

Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/136/2/279/530456/20220200.0-00013.pdf by guest on 10 May 2022


ple, Hsu et al.5 using the model developed by Dyck et al.,6
Snapir et al.7 using the model developed by Talke et al.,8 and Trial by Hannivoort et al.
Weerink et al.9 using the model developed by Hannivoort
For the first study, 18 subjects were included. For a thor-
et al.10 underpredicted the measured plasma concentra-
ough description of the study demographics, data col-
tions at higher targets. All aforementioned models include
lection, and analytic methods, please refer to Hannivoort
a two- or three-compartment mammillary model struc-
et al.10 In short, during the trial, an initial 20-s 6-µg ∙ kg–1 ∙ h–1
ture with first-order kinetics. Dutta et al.11 have suggested
infusion was started. Ten minutes after the start of this
that dexmedetomidine’s pharmacokinetic profile exhibited
infusion, a dexmedetomidine target-controlled infusion
nonlinear kinetics. During a recent dexmedetomidine trial
was started with stepwise increasing targets of 1, 2, 3, 4, 6,
conducted at our hospital, we noticed that there was bias in
the predictions differing from the target-controlled infusion and 8 ng · ml–1. Each target was maintained for 30 min
model for plasma concentrations above the 3.0 ng · ml–1 using the model developed by Dyck et al.6 Arterial blood
target-controlled infusion targets,9 indicating a potential samples were obtained at baseline, 2 min after the initial
nonlinear increase in exposure with increasing dexmedeto- 20-s infusion, before each increase in target concentration
midine target concentrations. The aim of the article was to (at 10 min and every 30 min thereafter), during the target-
investigate whether dexmedetomidine clearance is nonlin- controlled infusion administration period, and at 0, 2, 5, 10,
ear, to establish whether between-subject or within-subject 20, 60, 120, and 300 min during the postinfusion period.To
differences in cardiac output explain this apparent nonlin- avoid hypertension, the infusion rate was limited to 6 µg ∙
earity, to determine the clinical importance of this phe- kg–1 ∙ h–1 for the first three infusion steps and was increased
nomenon when following the current recommended dose to 10 µg ∙ kg–1 ∙ h–1 for the two highest target-controlled
regimen guidelines, and to establish the potential impact infusion targets of 6 and 8 ng · ml–1. The upper and lower
on the use of the currently available (linear) Hannivoort limits of quantification for the analytical method were 0.02
dexmedetomidine model.10 and 20 ng · ml–1.

Materials and Methods Trial by Weerink et al.


In the second study, 30 subjects were included. For a thor-
Study Design and Data Collection ough description of the study demographics, data collec-
The data of two previously published clinical trials,9,10 tion, and analytic methods, please refer to Weerink et al.9
conducted in healthy subjects at the Department of Only data from the first period (dexmedetomidine alone
Anesthesiology at the University Medical Center and not in combination with remifentanil) were used for
Groningen (Groningen, The Netherlands) were pooled the current analysis. Target-controlled infusion was started
to build a pharmacokinetic model. Both trials were con- with stepwise increasing targets of 1, 3, 4, 5, and 8 ng · ml–1
ducted in accordance with the Declaration of Helsinki every 40 to 50 min using the previously developed dex-
and in compliance with good clinical practice and the medetomidine model by Hannivoort et al.10 Arterial blood
applicable regulatory requirements. Both trials explored samples were drawn at baseline and at pseudo-steady state
the effects of increasing target concentrations of dex- before changing the target concentration. Once the infu-
medetomidine stepwise using target-controlled infusion. sion was stopped, dexmedetomidine samples were drawn at
Ethical approval was obtained from an independent medi- 2, 5, 10, 20, 60, 120, 300, and 420 min. Dexmedetomidine
cal ethics review committee (Medisch Ethische Toetsings infusion was limited to 6 µg ∙ kg–1 ∙ h–1 for the first three
Commissie) by the Association for the Evaluation of infusion steps and was increased to 10 µg ∙ kg–1 ∙ h–1 for the
Ethics in Biomedical Research (Stichting Beoordeling two highest targets of 5 and 8 ng · ml–1. The protocol was,
Ethiek Biomedisch Onderzoek, Assen, The Netherlands). however, amended to lower the highest dexmedetomidine
Both studies were registered in the ClinicalTrials.gov target from 8 to 6 ng · ml–1 due to safety concerns. The

280 Anesthesiology 2022; 136:279–92 Alvarez-Jimenez et al.


Copyright © 2021, the American Society of Anesthesiologists. All Rights Reserved. Unauthorized reproduction of this article is prohibited.
Dexmedetomidine Elicits Nonlinear Kinetics

upper and lower limits of quantification for the analytical estimation of the effect of a covariate on between- and within-
method were 0.05 and 20 ng · ml–1. subject variability on a pharmacokinetic parameter.14

FCO = e ( BSV )
θ ⋅ (CObaseline − 6.6) +θWSV ⋅ (CO −CObaseline )
Study Drug Administration and Data Collection (1)

Dexmedetomidine administration was conducted using
RUGLOOP II software (Demed, Belgium) using a syringe In equation 1, FCO represents the effect of cardiac output (CO)
pump with an Orchestra module DPS (Orchestra Base A, on dexmedetomidine clearance. In the model, FCO was a pro-
Fresenius Kabi, Germany); the software also recorded all portional factor on dexmedetomidine clearance.The parame-
vital parameters, pump infusion rates, and eventual case ters θBSV and θWSV are estimated to quantify the influence on
report form annotations during the execution of the clini- dexmedetomidine clearance of between-subject variability in
cal trials. Continuous cardiac output was measured through baseline cardiac output (CObaseline) compared to the median
a FloTrac sensor connected to a Vigileo monitor (both from in the population (6.6 l ∙ min–1) and within-subject longitu-

Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/136/2/279/530456/20220200.0-00013.pdf by guest on 10 May 2022


Edwards Lifesciences, USA) in the trial by Hannivoort et dinal changes from baseline cardiac output. Cardiac output
al.10 and through a hemodynamic monitor (EV1000 moni- was included in the data set as a time-varying covariate with
tor with FloTrac sensor; Edwards Lifesciences, USA) in the backward constant interpolation between observations.
trial by Weerink et al.9
Model Evaluation
Pharmacokinetic Model Development In-sample predictive performance variability was evalu-
ated according to Varvel et al.15 using the median perfor-
Statistical Analysis mance error and the median absolute performance error as
Data set creation, goodness of fit plots, and simulations were described in equation 2.
performed using R statistical software (R Foundation for
Statistical Computing, 2018; version 3.5.0). Nonlinear mixed C p observed − C p predicted
PE = ⋅ 100% (2)
effects modeling was conducted using the NONMEM C p predicted

(version 7.3.0) software package (ICON Development
Solutions, USA). In the first trial by Hannivoort et al.,10 where CP indicates the dexmedetomidine plasma concen-
there were concentrations below the limit of quantification tration, and PE indicates the performance error.
limit.Values below the limit of quantification were handled Model precision and parameter identifiability along with
using the M3 method according to Ahn et al.12 the variance of the parameters random effects were used
to numerically assess the models. Goodness of fit plots and
Model Building and Parameter Estimation visual predictive check plots were used to visually assess the
The set of differential equations were solved using a sec- model performance.The visual predictive checks were con-
ond-order approximation method (LAPLACE option) structed according to Bergstrand et al.16 and were based on
using the ADVAN13 subroutine. The a priori model 1,000 simulations. To graphically represent the difference
assumed weight-based allometric scaling on all pharmaco- in performance between the linear Hannivoort model and
kinetic parameters using a fixed exponent of 1 on volume our final nonlinear model, individual level visual predic-
of distribution terms and an exponent of 0.75 on clearance tive checks were conducted for six subjects from our data
terms.13 Interindividual variability was incorporated using a set that had the highest median performance error values
log-normal distribution. Residual variability was modeled according to the linear Hannivoort model.
using a proportional and additive error model. Candidate
models were compared using the Akaike information cri- Simulations
terion, and the model with the lowest Akaike criterion was Visual predictive check simulations were based on a sample
taken forward. Covariates were screened by exploring plots of 1,000 virtual subjects with characteristics randomly sam-
of random effects versus covariates and were included in the pled from the multivariate distribution of observed patient
model if inclusion resulted in a decrease in the Akaike cri- characteristics from our study. Plasma target-controlled infu-
terion. The covariates that were tested for inclusion in the sion simulations were based on the Hannivoort model that
model were age (yr), total body weight (kg), height (cm), assumes first-order pharmacokinetics, taking into account
and sex. The derived parameters body surface area, body the maximal recommended infusion rate of 6 µg ∙ kg–1 ∙ h–1 .
mass index, and lean body mass were also tested. Effect-site target-controlled infusion simulations were based
on the ke0 from the dexmedetomidine pharmacokinetic–
Testing Cardiac Output as a Covariate on pharmacodynamic model for the Modified Observer’s
Dexmedetomidine Clearance Assessment of Alertness and Sedation (MOAA/S) score17
The influence of cardiac output on clearance was tested (ke0 = 4.85 ∙ 10–2 min–1) and were based on the PKPD
according to equation 1. This equation allows simultaneous Tools for Excel package.18 The ICU simulations consisted

Alvarez-Jimenez et al. Anesthesiology 2022; 136:279–92 281


Copyright © 2021, the American Society of Anesthesiologists. All Rights Reserved. Unauthorized reproduction of this article is prohibited.
PERIOPERATIVE MEDICINE

of a loading dose of 1 µg ∙ kg–1 over 10 min followed by Nonlinear dexmedetomidine clearance was tested in the
a constant rate of 0.7 μg ∙ kg–1 ∙ h–1 for 1 h and, alterna- model using equation 3:
tively, followed by the maximum recommended constant
rate infusion of 1.4 µg ∙ kg–1 ∙ h–1 over 24 h according to  I MAX ⋅ C p γ 
CL = CL0 ⋅ 1 − γ  (3)
the European Medicinal Agency Summary of Medicinal
Product Characteristics19 for sedation in the ICU.  ( γ
C p + C 50 ) 

where the clearance at a particular plasma concentration
Results
ranges between CL0 and CL0 ∙ (1 – IMAX) depending on
Analysis Data Set the plasma concentration of dexmedetomidine (Cp). C50
determines the concentration at which 50% of inhibition
The final analysis data set included 48 subjects, 18 subjects takes place, and the exponent (γ) determines the steepness
from the trial by Hannivoort et al.,10 and 30 were from

Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/136/2/279/530456/20220200.0-00013.pdf by guest on 10 May 2022


of the relationship. IMAX represents the theoretical maxi-
the trial by Weerink et al.9 From the trial by Hannivoort mum inhibitory effect at which dexmedetomidine clear-
et al.,10 two sessions, separated by a washout, were included. ance decreases depending on the plasma concentration. In
From a total of 762 observations, 29 plasma concentration the tested models, IMAX was fixed to 1 (i.e., dexmedetomi-
were below the lower limit of quantification in the trial dine clearance decrease to 0 at very high concentrations) to
by Hannivoort et al.10 There were no samples below the avoid numerical difficulties with the parameter estimation.
lower limit of quantification in the trial by Weerink et al.9 Inclusion of equation 3 in the three-compartment model
Dexmedetomidine concentrations ranged from 0.02 to resulted in a significant decrease in the Akaike criterion of
14.4 ng · ml–1. A stratified inclusion resulted in a balanced 169 points. Figure 1 depicts the post hoc clearance for all
population: 24 subjects were male, and 24 subjects were subjects in the data set as a function of the dexmedetomi-
female. The mean age was 42.2 yr (range, 18 to 70 yr). The dine plasma concentration. Supplemental Digital Content
mean total body weight was 74.8 kg (SD, 13.3 kg; and range, figure S3 (http://links.lww.com/ALN/C743) shows that
49 to 110 kg), and height was 176.2 cm (SD, 10.7 cm; and this nonlinear kinetic model adequately described both
range, 155 to 203 cm). In total, 17 subjects were between 18 data sets. Interoccasion variability was tested on all phar-
and 34 yr old, 15 were between 35 and 50 yr old, and 16 macokinetic parameters but did not result in a decrease in
were between 51 and 70 yr old . Cardiac output measure- the model Akaike criterion. The additive error term in the
ments were available at a resolution of 1 measurement every model for the trial by Weerink et al.9 was negligible and was
10 s, resulting in a median of 728 (range, 194 to 1,375) car- therefore removed.
diac output measurements per subject included in the data
set. Figure S1 of Supplemental Digital Content (http:// Covariate Analysis
links.lww.com/ALN/C743) shows the measured cardiac
output against the measured dexmedetomidine plasma Covariates were tested after accepting the model with non-
concentrations for all subjects included in the analysis. linear elimination and once interindividual variability was
incorporated. In our final model, as shown in equations 4
to 8 and table 2, the volumes of distribution (V1,V2, and V3)
Structural Model Development
scaled linearly with FSIZE, the intercompartment clearance
Linear versus Nonlinear Elimination Kinetics (Q1 and Q2) scaled with size according to an exponent
of 0.75, and CL0 scaled with FSIZE according to an expo-
A three-compartment mamillary model with linear phar- nent of 0.75 and decreased with increasing age as function
macokinetics from the central compartment was used as of FAGE.
the starting point for the model development. This first
model showed considerable bias in the highest predicted  WGT
concentrations when compared to the measured observa-  Male =
70 (4)
tions (Supplemental Digital Content fig. S2, http://links. FSIZE 
WGT
lww.com/ALN/C743). Although the linear pharmacoki- Female = ⋅ (1 + θSEX ~ FSIZE )
 70 
netic model adequately described the data from the trial by
Hannivoort et al.10 (the data set on which this model was
FAGE = e (
−θ AGE ~CL 0 ⋅ ( AGE − 35))
developed, upper panel in Supplemental Digital Content (5)

fig. S2, http://links.lww.com/ALN/C743), it did not ade-
quately describe the observations from the trial by Weerink V = θV · FSIZE  (6)
et al.9 (lower panel in Supplemental Digital Content fig. S2,
http://links.lww.com/ALN/C743). Subsequent modifica- CL0 = θCL 0 · FSIZE 0.75 ⋅ FAGE  (7)
tions to this starting model are summarized in the model
development log as presented in table 1. Q = θQ ⋅ FSIZE 0.75 (8)


282 Anesthesiology 2022; 136:279–92 Alvarez-Jimenez et al.


Copyright © 2021, the American Society of Anesthesiologists. All Rights Reserved. Unauthorized reproduction of this article is prohibited.
Dexmedetomidine Elicits Nonlinear Kinetics

Table 1. Dexmedetomidine Model Development Log

Akaike Change in Reference


Model Characteristics Criterion Akaike Criterion Model

1 Three-compartment, clearance with linear kinetics –663.1


2 Three-compartment, clearance with nonlinear kinetics –831.8 –168.7 Model 1
3 Three-compartment, clearance with nonlinear kinetics and sex on FSize –842.8 –11.0 Model 2
4 Final model: three-compartment, clearance with nonlinear kinetics, sex on FSize, and age on clearance –849.8 –7.00 Model 3

The estimates of the final model can be found in table 2. Akaike criterion = –11). In addition, age was a significant
Figure 2 presents the goodness of fit plots of the final covariate on CL0 (change in Akaike criterion = –7), and

Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/136/2/279/530456/20220200.0-00013.pdf by guest on 10 May 2022


model, which showed no clear systematic trend or bias, con- CL0 decreased with age as shown in equation 5. On aver-
firming the adequacy of the model to describe the concen- age, for every 10 yr of age, the clearance decreased by 5.1%.
tration-time profiles of dexmedetomidine. The final (log-) There were no significant correlations between C50 and the
likelihood profiles can be found in Supplemental Digital tested covariates.
Content figure S4 (http://links.lww.com/ALN/C743). We found that cardiac output did not explain
Supplemental Digital Content figure S5 (http://links.lww. between-subject or within-subject variability in dex-
com/ALN/C743) depicts the prediction- and variance-cor- medetomidine clearance. Implementation of equation 1
rected visual predictive check for the final model. The in the model did not significantly decrease the objec-
parameter uncertainty was considered acceptable and ranged tive function value (change in Akaike criterion = +1.2; P
between 4 and 32.7% for the fixed effects and 30.5 to 40.8% value for likelihood ratio test = 0.25 for 2 degrees of free-
for the random effects.The final model had a median absolute dom). The estimated parameters for θBSV and θWSV were
performance error of 21.7% and a median performance error 0.0211 and 0.0037 and indicate that the estimated effect
of –2.3%. Figure 3 presents the individual-level visual predic- of cardiac output on clearance is negligible. According
tive check for the linear pharmacokinetic model (blue lower to equation 1, these estimates denote (1) that an individ-
panels) and the nonlinear pharmacokinetic model (gray upper ual with a cardiac output that is 50% of the population
panels) for six subjects from our data set. In this graph, it is median (6.6 l ∙ min–1) has a dexmedetomidine clearance
clear that the highest concentrations are not contained in the that is 6.7% lower compared to the typical clearance and
95% prediction interval for the linear pharmacokinetic model. (2) that a relative change from baseline cardiac output
We found that females had a higher estimated FSIZE by of 50% is associated with a 1.2% decrease in dexmede-
a factor of 1.18 compared to males (equation 4; change in tomidine clearance. Prediction-variance–corrected visual

1.5
Post hoc clearance (L/min)

1.0

0.5

0.0
0 5 10 15
Plasma concentration (ng/mL)

Fig. 1. Post hoc clearance versus plasma concentrations. The gray-shaded area denotes the 95% prediction interval according to the final
model with nonlinear elimination. The red line denotes the predicted median values at every plasma concentration of the model. The gray
lines around the mean depict the post hoc predicted clearance estimates against the plasma concentrations for all subjects included in our
analysis.

Alvarez-Jimenez et al. Anesthesiology 2022; 136:279–92 283


Copyright © 2021, the American Society of Anesthesiologists. All Rights Reserved. Unauthorized reproduction of this article is prohibited.
PERIOPERATIVE MEDICINE

Table 2. Dexmedetomidine Final Pharmacokinetic Parameters

Pharmacokinetic Parameter Estimate (Relative Standard Error %) [95% CI]*

FSIZE  WGT
 Male =
70

Female = WGT ⋅(1 + θ
 SEX ~FSIZE )
70

θSEX~FSIZE 1.80 ∙ 10–1 (26.2) [0.91 ∙ 10–1 to 2.76 ∙ 10–1]


FAGE
e
( −θ AGE ~CL ⋅ ( AGE − 35))
θAGE~CL 5.19 ∙ 10–3 (32.7) [1.90 ∙ 10–3 to 8.57 ∙ 10–3]

Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/136/2/279/530456/20220200.0-00013.pdf by guest on 10 May 2022


V1 (l) θV 1 ⋅ FSIZE
θV1 1.87 (13.8) [1.38 to 2.40]
V2 (l) θV 2 ⋅ FSIZE
θV2 17.8 (9.11) [15.3 to 21.7]
V3 (l) θV 3 ⋅ FSIZE
θV3 67.4 (3.33) [63.5 to 72.3]
CL0 (l ∙ min–1)
θCL 0 ⋅ FSIZE 0.75 ⋅ FAGE
θCL0 7.99 ∙ 10–1 (5.28) [7.25 ∙ 10–1 to 8.91 ∙ 10–1]
Q2 (l ∙ min–1)
θQ 2 ⋅ FSIZE 0.75
θQ2 2.97 (11.2) [2.45 to 3.77]
Q3 (l ∙ min–1)
θQ 3 ⋅ FSIZE 0.75
θQ3 7.54 ∙ 10–1 (4.02) [6.92 ∙ 10–1 to 8.10 ∙ 10–1]
C50 (ng ∙ ml–1) 5.75 (15.0) [4.27 to 7.64]
γ 1.34 (11.4) [1.08 to 1.67]
Between-subject variability (coefficient of variation %)†
V2 39.9 (35.1) [28.3 to 54.8]
CL0 15.1 (32.8) [11.0 to 20.4]
C50 57.9 (30.5) [44.2 to 77.4]
Residual unexplained variability (SD)‡
Proportional error – Hannivoort study 1.62 ∙ 10–1 (4.31) [1.50 ∙ 10–1 to 1.78 ∙ 10–1]
Proportional error – Weerink study 1.44 ∙ 10–1 (4.43) [1.33 ∙ 10–1 to 1.58 ∙ 10–1]
Additive error – Hannivoort study 2.84 ∙ 10–2 (12.5) [2.25 ∙ 10–2 to 3.65 ∙ 10–2]

*Relative standard error (%) is calculated from the log-likelihood profiles. †Coefficient of variation (%) is calculated according to ω 2 * 100%, where ω2 is the estimated variance in
the NONMEM software package (version 7.3.0; ICON Development Solutions, USA). ‡SD is calculated as the square root of the estimated variance in NONMEM.
Age, subject age (yr); C50, dexmedetomidine concentration where the clearance is half-maximal; CL0, clearance; FAGE, factor to account for age-related changes in pharmacokinetic
parameters; FSIZE, factor to account for size-related changes in pharmacokinetic parameters; Q2, distributional clearance between the central and the fast peripheral compartment; Q3,
distributional clearance between the central and the slow peripheral compartment; V1, volume of distribution of the central compartment; V2, volume of distribution of the fast peripheral
compartment; V3, volume of distribution of the slow peripheral compartment; WGT, subject weight (kg); γ, steepness of the relationship between the dexmedetomidine concentration
and resulting decrease in dexmedetomidine clearance.

predictive checks stratified by covariate for total body to the linear pharmacokinetic model by Hannivoort et
weight (Supplemental Digital Content fig. S6, http:// al.10 with increasing target concentrations. The aim of
links.lww.com/ALN/C743), sex (Supplemental Digital the simulation was to identify the concentration range in
Content fig. S7, http://links.lww.com/ALN/C743), and which predictions from our final nonlinear model would
age (Supplemental Digital Content fig. S8, http://links. deviate from the expected concentrations according to
lww.com/ALN/C743) can be found in the supplemental a target-controlled infusion system based on the linear
materials. pharmacokinetic model by Hannivoort et al.10 Figure 4
shows the simulated plasma concentrations for the target-
Simulations controlled infusion system based on the linear pharma-
cokinetic model by Hannivoort et al.10 and the predicted
Simulation of Target-controlled Infusion Using the plasma concentrations and the 95% prediction interval
Hannivoort Model for our final nonlinear model. The predicted concentra-
To better illustrate the differences between the models and tions from the final nonlinear model and the target-con-
the possible implications in the clinical setting, we simu- trolled infusion model are in good agreement until 2 ng
lated target-controlled infusion administrations according · ml–1. Beyond this target concentration, a discrepancy

284 Anesthesiology 2022; 136:279–92 Alvarez-Jimenez et al.


Copyright © 2021, the American Society of Anesthesiologists. All Rights Reserved. Unauthorized reproduction of this article is prohibited.
Dexmedetomidine Elicits Nonlinear Kinetics

Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/136/2/279/530456/20220200.0-00013.pdf by guest on 10 May 2022


Fig. 2. Goodness of fit diagnostic plots for the final model. The top panels show the population (left) and individual (right) predictions on the
y-axis plotted against the observed dexmedetomidine plasma concentrations. The continuous red line is a polynomial logistic regression used
as an aid to identify biased from the line of unity (when applicable). The middle panels show the individual conditional weighted residuals
plotted against the individual predictions (left) and time (right). The bottom panels show the conditional weighted residuals plotted against
the individual predictions (left) and time (right).

is seen between the predicted concentrations accord- pharmacokinetic model by Hannivoort et al.10 (blue lines
ing to the target-controlled infusion model and the pre- and shaded area) and our final nonlinear model (gray lines
dictions according to the nonlinear model with 62.1% and shaded area).The median predicted CMAX was 1.28-fold
of the simulated individuals being above the target at a higher (95% prediction interval: 0.4 to 2.8) at 0.1 ng · ml–1,
target of 3 ng · ml–1 to 80.4% at 5 ng · ml–1. The inac- 1.28-fold higher (0.4 to 2.5) at 0.2 ng · ml–1, 1.27-fold higher
curacy of the linear target-controlled infusion model, (0.4 to 2.2) at 0.5 ng · ml–1, 1.29-fold higher (0.4 to 2.2)
expressed as the ratio between the predicted concentra- at 1.0 ng · ml–1, 1.26-fold higher (0.4 to 2.0) at 2.0 ng · ml–1,
tion according to our final nonlinear model and the tar- and 1.22-fold higher (0.4 to 2.0) at 3.0 ng · ml–1.
get concentration, was 0.89 (95% prediction interval = 0.7
to 1.2) at 1 ng · ml–1, 0.97 (0.7 to 1.6) at 2 ng · ml–1, 1.1 (0.7 Critical Care Guidelines Recommended Dosing
to 2.0) at 3 ng · ml–1, 1.2 (0.7 to 2.4) at 4 ng · ml–1, and 1.4 Simulation
(0.8 to 2.9) at 5 ng · ml–1. To further illustrate the implications of the nonlinear
Another scenario that we explored was effect-site target- pharmacokinetics of dexmedetomidine we simulated the
controlled infusion. Figure 5 shows effect-site target- current European Medicines Agency (Amsterdam, The
controlled infusion for increasing effect-site targets and the Netherlands)–approved dosing regimen for sedation in the
resulting predicted plasma concentrations for the linear ICU,19 consisting of a weight-based constant rate infusion

Alvarez-Jimenez et al. Anesthesiology 2022; 136:279–92 285


Copyright © 2021, the American Society of Anesthesiologists. All Rights Reserved. Unauthorized reproduction of this article is prohibited.
PERIOPERATIVE MEDICINE

Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/136/2/279/530456/20220200.0-00013.pdf by guest on 10 May 2022


Fig. 3. Visual predictive check comparing linear and nonlinear pharmacokinetics models. Six subjects were simulated using both the final
nonlinear model (gray) and the previous linear model (blue). The subjects were selected based on the highest individual median performance
error on the linear kinetics model. The shaded area surrounding the model prediction median is the 95% prediction interval (based on 500
simulations). The red dots are the observations.

(µg ∙ kg–1 ∙ h–1). The upper panel of figure 6 shows that the were the first to suggest nonlinearity in dexmedetomi-
predicted CMAX according to our final nonlinear model reaches dine pharmacokinetics. In line with Dyck et al.,6 Dutta
3.2 ng · ml–1 (95% prediction interval, 2.2 to 4.8) after the ini- et al.11 hypothesized that a dose-related reduction in car-
tial loading dose, whereas the predicted CMAX according to the diac output resulted in a decrease in hepatic blood flow
linear pharmacokinetic model by Hannivoort et al.10 is 1.5 ng and consequently a reduced dexmedetomidine clearance.
· ml–1 (95% prediction interval, 1.1 to 2.0). The lower panel Interestingly, Dutta et al.11 concluded that based on their
in figure 6 depicts the maximum recommended continuous data from 10 healthy volunteers, there was no statistically
administration of dexmedetomidine and shows a good agree- significant difference in the fit of a cardiac output inde-
ment between the median predicted plasma concentrations for pendent and dependent pharmacokinetic model. In 2012,
both models. However, figure 6 suggests that with time, the Iirola et al.20 reported a positive association between cardiac
variability in the population increases, with the 95% prediction output and dexmedetomidine clearance in eight patients
interval increasing from 0.95 to 2.02 ng · ml–1 1 h after the dose admitted to the ICU with a continuous infusion of dexme-
to 1.80 to 20.84 ng · ml–1 toward the end of the 24-h infusion detomidine. Unfortunately, Iirola et al.20 did not compare a
period.At the end of the 24-h infusion, 62.8% of the simulated model with cardiac output against a nonlinear pharmacoki-
subjects had plasma concentrations above the median concen- netic model without cardiac output, like the model we are
tration predicted with the linear model, 24.3% twice as high proposing. Such a comparison would have given an indica-
and 7.3% five times higher than the concentrations predicted tion of whether cardiac output is a better surrogate than the
by the linear model. dexmedetomidine plasma concentration for describing the
nonlinear dexmedetomidine pharmacokinetic model. At
Discussion the same time, it is noteworthy that the subjects in the study
In this pooled analysis, we have shown that dexmedeto- by Iirola et al.20 received concomitant medication known/
midine administered to healthy subjects exhibits nonlinear suspected to decrease cardiac output (e.g., propofol), which
clearance, which may lead to higher than expected plasma might have confounded the association between dexmede-
concentrations with increasing drug exposure. Dyck et al.6 tomidine plasma concentration and cardiac output.

286 Anesthesiology 2022; 136:279–92 Alvarez-Jimenez et al.


Copyright © 2021, the American Society of Anesthesiologists. All Rights Reserved. Unauthorized reproduction of this article is prohibited.
Dexmedetomidine Elicits Nonlinear Kinetics

15
80.4 %

Dexmedetomidine concentration (ng/mL)

10
73.5 %

62.1 %
5

45.9 %

Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/136/2/279/530456/20220200.0-00013.pdf by guest on 10 May 2022


36 %
0

0 200 400 600 800 1000

time (min)

Fig. 4. Target-controlled infusion simulation comparing linear and nonlinear pharmacokinetics models. Plasma simulated concentrations
against time in an increasing dose scheme. Simulations were performed using the linear model as the driving model (blue); i.e., the needed
dose to accomplish as soon as possible the given target concentration (1 to 5 ng · ml–1) was calculated using the target-controlled infusion
algorithm. Using this same dose as a fixed variable, the theoretical real concentration was calculated to be used in the nonlinear parameters
(gray). The shaded area surrounding the median prediction is the 95% prediction interval. Percentages at each dose target depict the per-
centages of individuals with concentrations above the target plasma concentration. For the simulations, 1,000 randomly generated subjects
were used. Simulated demographics (age, weight, and sex) were also sampled from the model.

Our analysis based on continuous cardiac output mea- over 10 min, i.e., 6 µg · kg–1 · h–1), and the finding of
surements in 48 healthy volunteers has shown that cardiac decreased cardiac output in these dose groups is therefore
output is not a significant predictor for between-subject less representative of the current use of dexmedetomidine.
variability in dexmedetomidine clearance. We also showed Our model, most notably equation 3, implies that dex-
that changes from baseline cardiac output are not correlated medetomidine clearance decreases with increasing dexme-
with within-subject longitudinal changes in dexmedetomi- detomidine plasma concentrations. Saturation of metabolic
dine clearance. clearance at higher concentrations is not expected to have
Bloor et al.21 studied the hemodynamic changes, includ- an influence on dexmedetomidine clearance because dex-
ing cardiac output, induced after IV dexmedetomidine medetomidine is considered a high extraction ratio drug.
bolus doses of 0.25, 0.5, 1.0, and 2.0 µg · kg–1 administered According to the well stirred liver model, liver blood flow
over 2 min. These authors found that cardiac output (and and not enzyme activity is the main determinant of clear-
systolic blood pressure) in the lowest dose group was not ance for high extraction ratio drugs. This reasoning is sup-
different from placebo. In the trial by Hannivoort et al.10 ported by experimental work by Kohli et al.22 and Wang
and the trial by Weerink et al.,9 the dexmedetomidine infu- et al.,23 who observed no influence of CYP2A6 metabolizer
sion rate was limited to 6 µg · kg–1 · h–1 for target concen- status on dexmedetomidine pharmacokinetics.
trations less than 5 ng · ml–1 and to 10 µg · kg–1 · h–1 for We hypothesize that dexmedetomidine alters the liver
higher targets. These rate limitations imply that the bolus blood flow–to–cardiac output ratio in a concentration-
doses given by the target-controlled infusion system imme- dependent manner (in line with the other hemodynamic
diately after changing the target-controlled infusion target effects of dexmedetomidine21,24,25). Such a phenomenon
were limited to a rate of 0.20 µg · kg–1 and 0.33 µg · kg–1 per could account for the apparent lack of correlation between
2 min, respectively. In that respect, the absence of a signifi- cardiac output and dexmedetomidine clearance and the
cant dexmedetomidine-induced decrease in cardiac output concentration-dependent nature of the decrease in clear-
in our data (as shown in Supplemental Digital Content fig. ance. Interestingly, a similar phenomenon was observed
S1, http://links.lww.com/ALN/C743) is not surprising for propofol, another high extraction ratio drug. Peeters
and completely in line with the seminal work by Bloor et et al.26 demonstrated that in critically ill patients, there was
al.21 The 0.5, 1.0, and 2.0 µg ∙ kg–1 bolus doses studied by no relationship between propofol clearance and cardiac
Bloor et al.21 were infused at higher infusion rates than what output, whereas hepatic blood flow (measured by sorbitol)
is recommended nowadays on the drug label (1 µg · kg–1 was positively correlated with propofol clearance. Although

Alvarez-Jimenez et al. Anesthesiology 2022; 136:279–92 287


Copyright © 2021, the American Society of Anesthesiologists. All Rights Reserved. Unauthorized reproduction of this article is prohibited.
PERIOPERATIVE MEDICINE

Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/136/2/279/530456/20220200.0-00013.pdf by guest on 10 May 2022


Fig. 5. Effect-site target-controlled infusion simulations comparing linear and nonlinear pharmacokinetics models. Plasma simulated con-
centrations against time in an increased dosing scheme. Simulations were performed using the concentrations in the effect-site compartment
as the driving model. Six target concentrations were explored: 0.1, 0.2, 0.5, 1, 2, and 3 ng · ml–1. The theoretical concentration was calculated
using both the nonlinear (gray) and linear (blue) kinetic models. The effect-site concentration in the right vertical axis is represented by the
purple discontinuous line. The shaded area surrounding the median prediction is the 95% prediction interval. For the simulations, 1,000
randomly generated subjects were used. Simulated demographics (age, weight, and sex) were also sampled from the model.

the analysis by Peeters et al.26 might have been confounded which may account for the pharmacokinetic differences
by the presence of extrinsic factors, such as comedication, between the sexes. Supplemental Digital Content figure S9
their work supports our hypothesis that, in some cases, the (http://links.lww.com/ALN/C743) depicts the result of a
liver blood flow–to–cardiac output ratio is not constant, simulation in subjects with different covariates, providing
leading to no (or poor) correlation between cardiac output evidence that male subjects with increased age and a low
and drug clearance. total body weight might be subjects at risk of nonlinear
Our final nonlinear three-compartment pharmacoki- pharmacokinetics.
netic model successfully predicted dexmedetomidine con- Our final model assumes that clearance is completely
centrations over a wide concentration range. We found that inhibited at very high dexmedetomidine concentrations.
weight, age, and sex are significant covariates for the phar- This assumption was challenged by an attempt to estimate
macokinetics of dexmedetomidine. In addition to weight- an IMAX parameter in the model, describing the maximum
based allometric scaling, we identified the importance of proportional decrease in dexmedetomidine clearance
incorporating age into the model to enhance accuracy at very high concentrations. However, we encountered
of allometric scaling for the estimation of the clearance. numerical difficulties when trying to estimate IMAX, and this
Clearance was also inversely proportional to the age in sev- approach was abandoned. According to our final model,
eral previous models.3 Despite the lack of a clear consensus the mean maximal decrease in dexmedetomidine clear-
as to whether age correlates with the clearance or volumes ance across individuals was 55.2% (range, 23.3 to 82.7%).To
of distribution, three studies20,27,28 have reported positive estimate an IMAX parameter from experimental data, higher
results of age as covariate over a wide age range.With refer- dexmedetomidine concentrations than the ones reported in
ence to sex, this is the first data set that demonstrates varia- our study are likely necessary.
tion between females and males. Differences have also been In our study, the plasma concentrations were from arte-
reported in cardiac output and liver perfusion between rial blood samples taken during (45% of total number of
females and males in addition to the body fat composition,29 samples) or after (55%) drug infusion. Incomplete mixing

288 Anesthesiology 2022; 136:279–92 Alvarez-Jimenez et al.


Copyright © 2021, the American Society of Anesthesiologists. All Rights Reserved. Unauthorized reproduction of this article is prohibited.
Dexmedetomidine Elicits Nonlinear Kinetics

dexmedetomodine loading dose of 1 ug/kg in 10 min followed by a continuous


4
Dexmedetomidine concentration (ng/mL)

one−hour 0.7 ug/kg/hr infusion according to the Summary of Medical Product


Characteristics (SmPC) for sedation in ICU patients
non−linear elimination model
3

linear elimination model


2
1

Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/136/2/279/530456/20220200.0-00013.pdf by guest on 10 May 2022


0

0 50 100 150 200

time (min)
Dexmedetomidine concentration (ng/mL in log scale)

20.0
5.0
2.0
1.0
0.5

dexmedetomidine continuous dose (1.4 ug/kg/hr) in 24 hrs


non−linear elimination model
0.2

linear elimination model


0.1

0 500 1000 1500 2000

time (min)

Fig. 6. Weight dosed simulation using the current recommended dosing strategies for dexmedetomidine in clinical practice. The upper
panel illustrates the weight-only (and not target-controlled infusion) currently recommended dosing strategy for patients in the intensive care
unit (ICU) receiving sedation using dexmedetomidine. A dexmedetomidine loading dose of 1 μg · kg–1 administered in 10 min followed by a
continuous 0.7 μg · kg–1 · h–1 infusion for 1 h was simulated in 1,000 randomly generated 40-yr-old subjects with a total body weight of 80 kg.
The lower panel simulates the maximum recommended administration dose of 1.4 μg · kg–1 · h–1 without a loading dose for 24 h in 1,000
subjects with the same demographics as above.

of first-pass and recirculated drug concentrations, most nonlinear model on the remaining 419 (55% of total number
notably in the presence of reduced cardiac output, affects of observations) measured dexmedetomidine concentra-
measured plasma concentrations from samples taken during tions. The results showed strong statistical evidence in favor
drug infusion.30 As a consequence, the apparent nonlin- of the nonlinear model (Δ objective function value = –63;
earity that we described could have originated from the P value likelihood-ratio test < 0.001), thereby demonstrat-
amplifying effect of incomplete mixing rather than from a ing that potential incomplete mixing of arterial samples
concentration-dependent decrease in clearance. To test this during drug infusion is not confounding our conclusions.
hypothesis, we excluded all observations that were collected To identify the prediction discrepancies between our
during or shortly after (less than 5 min) drug infusion and final nonlinear pharmacokinetic model and the linear phar-
reestimated the linear three-compartmental model and the macokinetic model by Hannivoort et al.10 that is currently

Alvarez-Jimenez et al. Anesthesiology 2022; 136:279–92 289


Copyright © 2021, the American Society of Anesthesiologists. All Rights Reserved. Unauthorized reproduction of this article is prohibited.
PERIOPERATIVE MEDICINE

available in target-controlled infusion systems, we simu- the ICU for patients at risk during long-term administra-
lated different scenarios. For plasma target-controlled infu- tion and during an initial loading dose when following the
sion, we found that the model predictions for both models dosing strategies of the current dosing guidelines.
for plasma targets less than 2 ng · ml–1 were very consis-
tent. Conversely, when aiming for higher targets, caution Acknowledgments
is advised, and small increments in targets should be used
The authors thank Rob Spanjersberg, R.N. (University
due to nonlinear clearance. Effect-site target-controlled
Medical Center Groningen, Groningen, The Netherlands),
infusion simulations highlighted the discrepancies between
for coordinating and facilitating this research project, as well
both models when utilizing specific effect compartment
as all research personnel that collected the data of the two
concentrations. Effect-site target-controlled infusion differs
clinical studies. The authors also thank all of the volunteers
from the previous plasma concentration target-controlled
who contributed to the two previous clinical trials and
infusion because the aimed concentration is to be found
David A. Newhall, M.D. (North Bristol National Health

Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/136/2/279/530456/20220200.0-00013.pdf by guest on 10 May 2022


at the (theoretical) compartment concentration where
Service Trust, Bristol, United Kingdom), for his help with
the desired effect is expected, in this case sedative effect
thoroughly reviewing the article.
in the central nervous system. The effect-site target-con-
trolled infusion has the advantage of a quicker appearance
of the desired effect because the system administers an ini-
Research Support
tial bolus to achieve the target concentration at the effect Supported by the Department of Anesthesiology, University
site. Unfortunately, this initial bolus can lead to adverse of Groningen and University Medical Center Groningen,
effects as the plasma concentrations can abruptly peak. Groningen, The Netherlands.
Even though there was a difference between the linear
and nonlinear model plasma concentrations, the CMAX did Competing Interests
not increase in the target range from 0.1 to 3.0 ng · ml–1 Dr. Struys’s research group/department received (over the
with a maximum ratio between 1.22 and 1.28. As a final last 3 yr) research grants and consultancy fees from the
example of the implications of the model, we simulated the Medicines Company (Parsippany, New Jersey), Masimo
current guideline-recommended doses for sedation for ven- (Irvine, California), Fresenius (Bad Homburg, Germany),
tilated patients in the ICU.19 Figure 6 clearly demonstrates Dräger (Lübeck, Germany), Paion (Aachen, Germany),
that the predicted concentration would have doubled in the and Medtronic (Dublin, Ireland). Dr. Struys also receives
nonlinear model resembling the real plasma concentrations royalties on intellectual property from Demed Medical
compared to the previous linear model during the initial (Temse, Belgium) and Ghent University (Ghent,
1 µg · kg–1 loading dose administration. In addition, fig- Belgium). He is an editorial board member and director
ure 6 also identifies that long-term infusion (24 h) can cause for the British Journal of Anesthesia and associate editor
higher plasma concentrations in a minority of subjects with for Anesthesiology. He was not involved in the editorial
a low individual C50 value. Subjects in the extremes of age process of this publication. The other authors declare no
were not included in the trial. Future studies should exam- competing interests.
ine very elderly as well as pediatric subjects and multiple
comorbid patients. An enhanced understanding of the eti-
Correspondence
ologic mechanisms of nonlinearity are needed to further
understand the pharmacology of dexmedetomidine, and if Address correspondence to Dr. Colin: University
proven favorable for patient outcome, commercially avail- Medical Center Groningen, University of Groningen,
able target-controlled infusion pumps should include a P.O. Box 30001, 9700 RB Groningen, The Netherlands.
more advanced algorithm to include the solution of non- p.j.colin@umcg.nl. This article may be accessed for per-
linear kinetics. Today, target-controlled infusion steering sonal use at no charge through the Journal Web site,
algorithms implemented in clinically available pumps sup- www.anesthesiology.org.
port only linear kinetics.31
In this study, we developed a nonlinear three-compart- References
ment pharmacokinetic model that predicted dexmede-
tomidine at various concentration ranges. As opposed to 1. Hall JE, Uhrich TD, Barney JA, Arain SR, Ebert TJ:
previous studies, our model may permit a more accurate Sedative, amnestic, and analgesic properties of small-
dosing at high plasma concentrations (greater than 2 ng dose dexmedetomidine infusions. Anesth Analg 2000;
· ml–1). We identified weight, age, and sex as significant 90:699–705
covariates in our pharmacokinetic model. Administration 2. Belleville JP, Ward DS, Bloor BC, Maze M: Effects of
of up to 2 ng · ml–1 may be safe to utilize in the previously intravenous dexmedetomidine in humans: I. Sedation,
developed dexmedetomidine pharmacokinetic model with ventilation, and metabolic rate. Anesthesiology 1992;
linear or first-order pharmacokinetics. Caution is advised in 77:1125–33

290 Anesthesiology 2022; 136:279–92 Alvarez-Jimenez et al.


Copyright © 2021, the American Society of Anesthesiologists. All Rights Reserved. Unauthorized reproduction of this article is prohibited.
Dexmedetomidine Elicits Nonlinear Kinetics

3. Weerink MAS, Struys MMRF, Hannivoort LN, 15. Varvel JR, Donoho DL, Shafer SL: Measuring the pre-
Barends CRM, Absalom AR, Colin P: Clinical phar- dictive performance of computer-controlled infusion
macokinetics and pharmacodynamics of dexmedeto- pumps. J Pharmacokinet Biopharm 1992; 20:63–94
midine. Clin Pharmacokinet 2017; 56:893–913 16. Bergstrand M, Hooker AC, Wallin JE, Karlsson MO:
4. Dyck JB, Maze M, Haack C,Vuorilehto L, Shafer SL:The Prediction-corrected visual predictive checks for diag-
pharmacokinetics and hemodynamic effects of intrave- nosing nonlinear mixed-effects models. AAPS J 2011;
nous and intramuscular dexmedetomidine hydrochlo- 13:143–51
ride in adult human volunteers. Anesthesiology 1993; 17. Colin PJ, Hannivoort LN, Eleveld DJ, Reyntjens
78:813–20 KMEM, Absalom AR,Vereecke HEM, Struys MMRF:
5. Hsu YW, Cortinez LI, Robertson KM, Keifer JC, Dexmedetomidine pharmacokinetic–pharmacody-
Sum-Ping ST, Moretti EW, Young CC, Wright DR, namic modelling in healthy volunteers: 1. Influence of
Macleod DB, Somma J: Dexmedetomidine phar- arousal on bispectral index and sedation. Br J Anaesth

Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/136/2/279/530456/20220200.0-00013.pdf by guest on 10 May 2022


macodynamics: Part I. Crossover comparison of the 2017; 119:200–10
respiratory effects of dexmedetomidine and remifen- 18. Schnider TW, Minto CF: PKPD Tools for Excel.
tanil in healthy volunteers. Anesthesiology 2004; Available at: http://www.pkpdtools.com/excel.
101:1066–76 Accessed June 24, 2020.
6. Dyck JB, Maze M, Haack C, Azarnoff DL, Vuorilehto 19. Orion Pharma: Dexdor Product Information: Annex
L, Shafer SL: Computer-controlled infusion of intrave- I. Summary of Product Characteristics. Available at:
nous dexmedetomidine hydrochloride in adult human https://www.ema.europa.eu/en/documents/prod-
volunteers. Anesthesiology 1993; 78:821–8 uct-information/dexdor-epar-product-information_
7. Snapir A, Posti J, Kentala E, Koskenvuo J, Sundell J, en.pdf. Accessed June 23, 2021.
Tuunanen H, Hakala K, Scheinin H, Knuuti J, Scheinin 20. Iirola T, Ihmsen H, Laitio R, Kentala E, Aantaa R,
M: Effects of low and high plasma concentrations of Kurvinen JP, Scheinin M, Schwilden H, Schüttler
dexmedetomidine on myocardial perfusion and cardiac J, Olkkola KT: Population pharmacokinetics of
function in healthy male subjects. Anesthesiology dexmedetomidine during long-term sedation
2006; 105:902–10; quiz 1069–70 in intensive care patients. Br J Anaesth 2012;
8. Talke P, Richardson CA, Scheinin M, Fisher DM: 108:460–8
Postoperative pharmacokinetics and sympatholytic effects 21. Bloor BC, Ward DS, Belleville JP, Maze M: Effects
of dexmedetomidine. Anesth Analg 1997; 85:1136–42 of intravenous dexmedetomidine in humans: II.
9. Weerink MAS, Barends CRM, Muskiet ERR, Hemodynamic changes. Anesthesiology 1992;
Reyntjens KMEM, Knotnerus FH, Oostra M, 77:1134–42
van Bocxlaer JFP, Struys MMRF, Colin PJ: 22. Kohli U, Pandharipande P, Muszkat M, Sofowora GG,
Pharmacodynamic interaction of remifentanil and Friedman EA, Scheinin M, Wood AJ, Ely EW, Tyndale
dexmedetomidine on depth of sedation and tol- RF, Choi L, Stein CM, Kurnik D: CYP2A6 genetic
erance of laryngoscopy. Anesthesiology 2019; variation and dexmedetomidine disposition. Eur J Clin
131:1004–17 Pharmacol 2012; 68:937–42
10. Hannivoort LN, Eleveld DJ, Proost JH, Reyntjens KM, 23. Wang L, Wang S, Qi J,Yu R, Zhuang J, Zhuang B, Lou
Absalom AR,Vereecke HE, Struys MM: Development Y, Ruan J,Ye H, Lin F: Impact of CYP2A6 gene poly-
of an optimized pharmacokinetic model of dexmede- morphism on the pharmacokinetics of dexmedetomi-
tomidine using target-controlled infusion in healthy dine for premedication. Expert Rev Clin Pharmacol
volunteers. Anesthesiology 2015; 123:357–67 2018; 11:917–22
11. Dutta S, Lal R, Karol MD, Cohen T, Ebert T: Influence 24. Ebert TJ, Hall JE, Barney JA, Uhrich TD, Colinco MD:
of cardiac output on dexmedetomidine pharmacoki- The effects of increasing plasma concentrations of
netics. J Pharm Sci 2000; 89:519–27 dexmedetomidine in humans. Anesthesiology 2000;
12. Ahn JE, Karlsson MO, Dunne A, Ludden TM: 93:382–94
Likelihood based approaches to handling data below 25. Colin PJ, Hannivoort LN, Eleveld DJ, Reyntjens
the quantification limit using NONMEM VI. J KMEM, Absalom AR,Vereecke HEM, Struys MMRF:
Pharmacokinet Pharmacodyn 2008; 35:401–21 Dexmedetomidine pharmacodynamics in healthy vol-
13. Anderson BJ, Holford NH: Mechanism-based con- unteers: 2. Haemodynamic profile. Br J Anaesth 2017;
cepts of size and maturity in pharmacokinetics. Annu 119:211–20
Rev Pharmacol Toxicol 2008; 48:303–32 26. Peeters MY, Aarts LP, Boom FA, Bras LJ, Tibboel D,
14. Wählby U, Thomson AH, Milligan PA, Karlsson MO: Danhof M, Knibbe CA: Pilot study on the influence of
Models for time-varying covariates in population liver blood flow and cardiac output on the clearance of
pharmacokinetic–pharmacodynamic analysis. Br J Clin propofol in critically ill patients. Eur J Clin Pharmacol
Pharmacol 2004; 58:367–77 2008; 64:329–34

Alvarez-Jimenez et al. Anesthesiology 2022; 136:279–92 291


Copyright © 2021, the American Society of Anesthesiologists. All Rights Reserved. Unauthorized reproduction of this article is prohibited.
PERIOPERATIVE MEDICINE

27. Kuang Y, Xiang YX, Guo CX, Zhang RR, Yang G, 29. Soldin OP, Mattison DR: Sex differences in pharmaco-
Hou GF, Tan HY, Huang L, Huang J, Ouyang W, Duan kinetics and pharmacodynamics. Clin Pharmacokinet
KM, Wang SY, Li JL, Pei Q: Population pharmacoki- 2009; 48:143–570
netics study of dexmedetomidine in Chinese adult 30. Upton RN: The two-compartment recirculatory
patients during spinal anesthesia. Int J Clin Pharmacol pharmacokinetic model: An introduction to recircu-
Ther 2016; 54:200–7 latory pharmacokinetic concepts. Br J Anaesth 2004;
28. Lee S, Kim BH, Lim K, Stalker D, Wisemandle W, Shin 92:475–84
SG, Jang IJ, Yu KS: Pharmacokinetics and pharmaco- 31. Struys MM, De Smet T, Glen JI,Vereecke HE, Absalom
dynamics of intravenous dexmedetomidine in healthy AR, Schnider TW: The history of target-controlled
Korean subjects. J Clin Pharm Ther 2012; 37:698–703 infusion. Anesth Analg 2016; 122:56–69

ANESTHESIOLOGY REFLECTIONS FROM THE WOOD LIBRARY-MUSEUM

Downloaded from http://pubs.asahq.org/anesthesiology/article-pdf/136/2/279/530456/20220200.0-00013.pdf by guest on 10 May 2022


Conjuring Childhood Dreams, Pond’s Extract “Cures All”

Patients in the late nineteenth century often feared the expense and inconsistency of physician remedies, reach-
ing for drugstore “cure-alls” instead. Store clerks in the United States handed out trade cards that advertised
these “patent medicines,” some of which contained high levels of alcohol, morphine, or cocaine. Capturing the
hope and innocence of youth, these cheerful collectibles became a scrapbooking favorite. In the example above,
a popular trade card celebrates “Pond’s Extract: Vegetable Pain Destroyer,” brainchild of pharmacist Theron T.
Pond. A dream of childhood imbues the image; an angel-winged girl holds a bright umbrella over a monocled
boy on an iridescent seashore. “What’s on the back?” the card beckons (bottom). Its reverse unveils the acronym
“POND’S EXTRACT” with its corresponding indications: “Piles, Old sores, Neuralgia, Diphtheria, Sprains,
Eyes inflamed, Xternal Use, Tonsilitis, Rheumatism, Accidents, Catarrh, Throat sore.” A topical remedy of witch
hazel (Hamamelis virginiana) and dilute alcohol, Pond’s Extract was beloved for its skin-protectant and anti-in-
flammatory properties. In the early twentieth century, the T.T. Pond Company would also develop Pond’s Cold
Cream, now considered a classic beauty product. (Copyright © the American Society of Anesthesiologists’
Wood Library-Museum of Anesthesiology, Schaumburg, Illinois. www.woodlibrarymuseum.org)
Jane S. Moon, M.D., Assistant Clinical Professor, Department of Anesthesiology and Perioperative Medicine, University
of California, Los Angeles, California.

292 Anesthesiology 2022; 136:278–92 Alvarez-Jimenez et al.


Copyright © 2021, the American Society of Anesthesiologists. All Rights Reserved. Unauthorized reproduction of this article is prohibited.

You might also like