You are on page 1of 9

Dexmedetomidine Sedation in Mechanically

Ventilated Critically Ill Children: A Pilot Randomized


Controlled Trial
Simon J. Erickson, MBBS, FRACP, FCICM1; Johnny Millar, MB ChB, PhD, MRCP, FRACP, FCICM2,3;
Brian J. Anderson, MB ChB, PhD, FANZCA, FCICM4,5; Marino S. Festa, MD, FCICM6;
Lahn Straney, PhD7; Yahya Shehabi, MBBS, PhD, FCICM8,9; Debbie A. Long, RN, PhD10,11; for the
Baby SPICE Investigators and the Australian and New Zealand Intensive Care Society Paediatric
Downloaded from http://journals.lww.com/pccmjournal by BhDMf5ePHKbH4TTImqenVIiuKVF7qTxs1W+oIWQuQ7XxwtmptojNIu7z81y9c1iI on 09/04/2020

Study Group (ANZICS-PSG)

Objectives: To assess the feasibility, safety, and efficacy of a se- Interventions: Children randomized to dexmedetomidine received
dation protocol using dexmedetomidine as the primary sedative in a dexmedetomidine-based algorithm targeted to light sedation
mechanically ventilated critically ill children. (State Behavioral Scale –1 to +1). Children randomized to usual
Design: Open-label, pilot, prospective, multicenter, randomized, care received sedation as determined by the treating clinician (but
controlled trial. The primary outcome was the proportion of se- not dexmedetomidine), also targeted to light sedation.
dation scores in the target sedation range in the first 48 hours. Measurements and Main Results: Sedation with dexmedetomi-
Safety outcomes included device removal, adverse events, and dine as the primary sedative resulted in a greater proportion of
vasopressor use. Feasibility outcomes included time to randomi- sedation measurements in the light sedation range (State Behav-
zation and protocol fidelity. ioral Scale –1 to +1) over the first 48 hours (229/325 [71%] vs
Setting: Six tertiary PICUs in Australia and New Zealand. 181/331 [58%]; p = 0.04) and the first 24 hours (66/103 [64%]
Patients: Critically ill children, younger than 16 years old, requiring vs 48/116 [41%]; p < 0.001) compared with usual care. Cumu-
intubation and mechanical ventilation and expected to be mechan- lative midazolam dosage was significantly reduced in the dexme-
ically ventilated for at least 24 hours. detomidine arm compared with usual care (p = 0.002).There were
more episodes of hypotension and bradycardia with dexmedeto-
midine (including one serious adverse event) but no difference in
1
Paediatric Critical Care, Perth Children’s Hospital, Perth, WA, Australia. vasopressor requirements. Median time to randomization after in-
2
Paediatric Intensive Care Unit, Royal Children’s Hospital, Melbourne, tubation was 6.0 hours (interquartile range, 2.0–9.0 hr) in the dex-
VIC, Australia. medetomidine arm compared with 3.0 hours (interquartile range,
3
Murdoch Children’s Research Institute, University of Melbourne, Melbourne, 1.0–7.0 hr) in the usual care arm (p = 0.24).
VIC, Australia.
Conclusions: A sedation protocol using dexmedetomidine as the
4
Paediatric Intensive Care Unit, Starship Children’s Hospital, Auckland,
New Zealand. primary sedative was feasible, appeared safe, achieved early, light
5
Department of Anaesthesiology, University of Auckland, Auckland, New sedation, and reduced midazolam requirements. The findings of
Zealand. this pilot study justify further studies of sedative agents in critically
6
Kids Critical Care Research, The Children’s Hospital at Westmead, ill children. (Pediatr Crit Care Med 2020; 21:e731–e739)
Westmead, NSW, Australia. Key Words: child; critical illness; dexmedetomidine; hypnotics and
7
Department of Epidemiology and Preventive Medicine, Monash University, sedatives; mechanical ventilation; midazolam
Melbourne, VIC, Australia.
8
School of Clinical Sciences, Monash University, Melbourne, VIC, Australia.
9
Clinical School of Medicine, University of New South Wales, Sydney,

O
NSW, Australia. ptimal sedation is an integral component of the com-
10
Paediatric Intensive Care Unit, Queensland Children’s Hospital, Bris- prehensive medical care of the critically ill child and
bane, QLD, Australia.
provides anxiolysis, amnesia, and facilitates mechan-
11
Children’s Health Research Centre, The University of Queensland, Bris-
bane, QLD, Australia. ical ventilation. There is no universally accepted approach to
Copyright © 2020 by the Society of Critical Care Medicine and the World the sedation of mechanically ventilated children due to lack of
Federation of Pediatric Intensive and Critical Care Societies high quality evidence to guide sedation practice in critically ill
DOI: 10.1097/PCC.0000000000002483 children (1–5). Sedative and analgesic medications used and

Pediatric Critical Care Medicine www.pccmjournal.org e731


Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.
Unauthorized reproduction of this article is prohibited
Erickson et al

sedation goals may vary widely between and even within hospi- b)  Heart rate less than 2 sds below normal mean for age (23)
tals and optimal levels of sedation are often not achieved (6, 7).
A complete list of the exclusion criteria is shown in Supple-
Over-sedation may result in prolonged mechanical venti-
mentary Table 1 (Supplemental Digital Content 1, http://links.
lation, delirium, and drug tolerance and withdrawal, while
lww.com/PCC/B437).
under-sedation may lead to loss or displacement of IV access
The trial protocol was registered with Australian and New
and drains, unplanned extubation, and emotional distress. A
systematic review of sedation practices protocols and algo- Zealand Clinical Trials Registry (number 12614000225617).
rithms demonstrated that sedation guidelines can reduce drug Ethical approval was obtained at each participating site, and
exposure and PICU length of stay (8), and nurse-driven consent was obtained from the parent/guardian for all study
sedation and analgesia protocols have demonstrated reduc- participants. Subjects were randomly assigned, in permuted
tions in drug exposure. However, the impact of sedation blocks of 2–6 via concealed envelopes, in a 1:1 ratio to dex-
protocols on patient-centered outcomes such as duration of medetomidine as the primary sedative or to usual sedation
ventilation and neurocognitive outcome is still unclear (9–11). practice.
Midazolam is still widely used as a primary sedative in chil- Background Treatments
dren (7, 12–14). Midazolam has been linked to neurotoxicity Pain relief for all children was provided by an infusion or bolus
in animal studies (15, 16) and may be associated with delirium
of an opioid (e.g., morphine or fentanyl) or other drugs (e.g.,
and drug withdrawal, in addition to long-term neurocognitive
ketamine) at the discretion of the treating clinician. The de-
sequelae in children (17–21).
fault sedation target was light sedation, as defined by State
Dexmedetomidine, a selective α2-adrenoreceptor agonist,
Behavioral Scale (SBS) (24) range of –1 to +1 (responsive to
exerts its therapeutic effects via reduction of norepinephrine
gentle touch to restless) unless otherwise clinically indicated
in the sympathetic nerve endings in the central and peripheral
(e.g., painful procedures or neuromuscular blockade). Patients
nervous systems, resulting in sedation, anxiolysis, and anal-
who required neuromuscular blockade after randomization
gesia, and has not been linked to neurotoxicity (22).
received additional sedatives (if required) to ensure SBS –2
We conducted a randomized controlled pilot feasibility trial
to –3 to prevent awareness during neuromuscular blockade.
to compare the use of dexmedetomidine as a primary sedative,
compared with usual sedation practice in mechanically venti- Concurrent dexmedetomidine infusion was continued during
lated, critically ill children. this period. Once there was no need for further neuromuscular
blockade, sedative management continued as per protocol.
Agitation (SBS > +1) was managed by increasing the pri-
MATERIALS AND METHODS mary sedative agent. Persistent agitation was then managed by
Study Design the addition of second-line agents (such as propofol, ketamine,
The trial was an open-label, randomized, controlled pilot trial chloral hydrate, phenobarbital, and antihistamines) at the dis-
conducted in six tertiary PICUs in Australia and New Zealand. cretion of the treating physician. In refractory agitation, ben-
zodiazepines or dexmedetomidine use was permitted in both
Study Participants groups. All other routine care, including ventilation manage-
Inclusion Criteria. Children, younger than 16 years old, who ment and weaning, cardiovascular support and monitoring,
were mechanically ventilated via an endotracheal tube and had was performed using local unit protocols.
been intubated (excluding time spent intubated within an op- Pain and sedation (using SBS) were assessed by the
erating or procedural theater), for less than 12 hours, and the bedside nurse every four hours or more frequently if re-
treating clinician anticipated that: 1) the patient required im- quired. Withdrawal was assessed daily using the Withdrawal
mediate and ongoing sedative medication for comfort, safety, Assessment Tool-1 (WAT-1) (25, 26) after 72 hours following
and to facilitate the delivery of life support measures and 2) the randomization or when sedation was being weaned. Patients
patient was expected to remain intubated for at least 24 hours were assessed daily for delirium if SBS greater than or equal
after enrollment. to –1, using Preschool Confusion Assessment Method for the
ICU if 6 months to 5 years old (27) or Pediatric Confusion
Main Exclusion Criteria. Assessment Method for ICU if greater than 5 years old (28).
1) Primary diagnosis of proven or suspected acute brain in- Intervention Arm—(Dexmedetomidine)
jury resulting in impaired conscious level or cognition Following randomization, dexmedetomidine was commenced
2) Primary diagnosis of proven or suspected neurologic injury as the primary sedative agent at 1.0 µg/kg/hr without a load-
or pathology resulting in permanent or prolonged weakness ing dose and titrated to achieve target sedation up to an age-
3) Requirement or anticipated requirement for continuous dependent maximum dose (29, 30) (Supplemental Fig. 1,
neuromuscular blockade Supplemental Digital Content 2, http://links.lww.com/PCC/
4) Cardiovascular instability manifested by either of the following:
B438; legend, Supplemental Digital Content 4, http://links.
a) Mean arterial blood pressure less than 2 sds below lww.com/PCC/B440).
normal mean for age despite fluid and vasopressor re- Dexmedetomidine infusion was continued until sedation was
suscitation (23) no longer required or to a maximum of 14 days after enrollment.
e732 www.pccmjournal.org September 2020 • Volume 21 • Number 9
Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.
Unauthorized reproduction of this article is prohibited
Online Clinical Investigations

Supplemental sedative agents such as propofol (maximum light sedation. A sample size of 60 patients was estimated to pro-
dose 4 mg/kg/hr, maximum duration 24 hr), ketamine, chloral vide over 700 SBS assessments in the first 48 hours and 3,600
hydrate, sedating antihistamines, and phenobarbital according sedation hours to allow meaningful assessment of efficacy and
to physician preference were used if dexmedetomidine at the safety. All analyses were performed on an intention to treat prin-
maximum allowable and tolerable dose did not provide suffi- ciple. Data were collected for 14 days or until ICU discharge.
cient sedation. Data were presented as percentages and numbers, medians with
Benzodiazepine use was permitted only for salvage treat- interquartile ranges (IQRs), or means with sd. Two-sample Wil-
ment of refractory agitation. Clonidine was prohibited as a coxon rank-sum (Mann-Whitney U) tests were used to compare
sedative agent with use reserved for management of suspected subgroups. Two-sample t tests were used to compare normally
dexmedetomidine withdrawal (after cessation for 96 hr). distributed variables, and chi-square tests were used to compare
categorical variables between the intervention and standard
Usual Care Arm treatment arms. A two-sample test of proportions was used to
The primary sedative agent was at the discretion of the treat- compare the proportion of children achieving target sedation
ing clinician, given by bolus dose or infusion (excluding dex- between treatment arms. The difference in proportions was cal-
medetomidine or any other alpha-2 agonist) titrated to target culated with odds ratios and 95% CIs. A p value of less than 0.05
sedation range. Sedative agents could include propofol (max- were considered statistically significant. To account for within-
imum dose 4 mg/kg/hr, maximum duration 24 hr), benzodiaz- patient correlation for repeated assessments, comparisons have
epines, chloral hydrate, ketamine, and barbiturates, guided by been made with generalized linear modeling for repeated meas-
individual unit practice. Clonidine was used only as a supple- ures using a binomial distribution.
mental (rather than primary) sedative in this group. Time to randomization was the time between eligibility (in-
Dexmedetomidine could be used at the discretion of tubation) and randomization. All drug dosages were adjusted
the treating intensivist if the target sedation range was not for weight. All statistical analyses were conducted using Stata
achieved despite maximum titration of conventional sedative (Version 15.0; StataCorp, College Station, TX).
agents (except when clonidine had been used).
RESULTS
Outcome Measures
The primary outcome measure was the percentage of sedation Study Participants
scores in the target light sedation range (SBS –1 to +1) over the The study was conducted over an 18-month period from Jan-
first 48 hours of sedation in intensive care. uary 2015 to July 2016. There were 60 children included; 29
Safety outcomes were drug withdrawal (defined as WAT-1 were randomized to the dexmedetomidine arm and 31 to the
score > 3) (25, 26), unplanned extubation and reintubation, usual care arm.
loss of vascular cannulae, and adverse events including hypo- (Supplemental Fig. 2, Supplemental Digital Content 3,
tension and bradycardia (defined as heart rate or blood pres- http://links.lww.com/PCC/B439; legend, Supplemental Digital
sure more than two sds below mean for age) (23). Content 4, http://links.lww.com/PCC/B440)
Feasibility outcomes were the proportion of screened and Baseline demographic characteristics are shown in Table 1.
eligible patients who were recruited, time from eligibility to The median age was higher in the dexmedetomidine arm (16 mo
randomization, compliance with study protocol, and propor- [IQR, 4–30 mo]) compared with the usual care arm (3 mo [IQR,
tion of patients who were intubated for longer than 24 hours 1–13 mo) (p = 0.02). There were no other baseline imbalances.
and loss to follow-up at 90 days. Primary Outcome. During the first 48 hours of sedation
Additional clinical outcome measures included the per- after randomization, 229 of 325 (71%) of SBS measurements
centage of sedation scores in the target light sedation range in were in the light sedation range in the dexmedetomidine arm
the first 24 hours, the proportion of daily sedation score assess- compared with 181 of 311 (58%) in the usual care arm (odds
ments in SBS categories –2 to –3 (deep sedation), –1 to +1 ratio, 2.20; 95% CI, 1.03–4.72; p = 0.04). During the first 24
(light sedation) and agitation (+2), daily cumulative weight- hours, 66 of 103 (64%) of SBS measurements were in the light
adjusted doses of IV sedative agents and opioids given, occur- sedation range in the dexmedetomidine arm compared with 48
rence rate of delirium, the duration of mechanical ventilation, of 116 (41%) in the usual care arm (odds ratio, 2.5; 95% CI, 9.8–
intensive care and hospital length of stay, and vital status at 50.3%; p < 0.001). Time to achieve light sedation was shorter
hospital discharge and after 90 days of follow-up. with dexmedetomidine (median, 5.0  hr; IQR, 2.6–8.5  hr)
compared with usual care (9.0; 6.6–13.8; p = 0.02).
Statistical Analysis The distribution of SBS scores for each study arm over the
The primary outcome was the difference in proportion of mea- first 24 and 48 hours are shown in Figure 1.
surements in the target light sedation range in the first 48 hours Clinical and Safety Outcomes. Feasibility, safety, and clin-
in each arm. The main efficacy outcomes included the difference ical outcomes including median duration of ventilation, length
in the number of patients receiving the primary sedative agents, of ICU stay, and hospital stay are shown in Table 2.
number of days, and cumulative doses of each agent. Feasibility There were eight episodes of bradycardia (in seven patients)
outcomes included time to randomization and time to achieve in the dexmedetomidine arm, six were classified as mild with no

Pediatric Critical Care Medicine www.pccmjournal.org e733


Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.
Unauthorized reproduction of this article is prohibited
Erickson et al

TABLE 1.Baseline Characteristics and arm compared with a median (IQR) of 3.0 (1.0–7.0) in the
Patients Demographics usual care arm (p = 0.24).
There was very high compliance with the protocol with only
Dexmedetomidine Usual Care four protocol violations reported (one patient given dexme-
Patient Characteristics (n = 29) (n = 28)
detomidine prior to other sedatives in the usual care arm and
Age, mo, median (IQR) 16 (4–30) 3 (1–13) three subjects given midazolam prior to other sedatives in the
16 (55) 16 (57)
dexmedetomidine arm).
Gender, male, n (%)
There were 56 of 60 children (93%) ventilated and sedated
Weight, kg, median (IQR) 9.15 (5–14) 5.65 (3.7–9.4) for more than 24 hours. One patient lost to follow-up.
Nonoperative, n (%) 22 (76) 23 (82)
Sedative Medications
Pre admission cardiac 7 (24) 5 (18)
surgery, n (%) The sedation protocol was successfully adhered to with 28 of 29
children (96.5%) in the dexmedetomidine arm receiving dexme-
ICU admission source, n (%) detomidine compared with two of 28 children (7.1%) in usual
  Operating theater/recovery 11 (38) 8 (29) care (p < 0.0001). Midazolam was used more frequently in usual
  Emergency department 2 (7) 6 (21) care (26/28 [92.8%]) compared with the dexmedetomidine arm
(11/29 [37.9%]) (p < 0.0001). There were no significant differ-
 Ward 4 (14) 4 (14)
ences between the groups in the proportion of children receiving
  Other ICU/neonatal ICU 1 (3) 0 (0) propofol, ketamine, morphine, or fentanyl (Table 3).
same hospital Apart from dexmedetomidine and midazolam, there were
  Direct ICU admission 11 (38) 10 (36) no significant differences in the cumulative doses of other IV
  Elective admission 8 (28) 5 (18)
sedatives or opioids over the study period (Table 3) or the first
48 hours (Fig. 2A).
Diagnostic group, n (%) Children in the dexmedetomidine arm received propofol
  Cardiovascular (incl 7 (24) 6 (21) on more sedation days (48/165 [29%]) compared with usual
postoperative) care (22/144 [15%] sedation days; p = 0.004) and ketamine on
 Respiratory 11 (38) 13 (46) more sedation days (28/165 [17%]) compared with usual care
(1/144 [0.6%]) sedation days; p < 0.001).
 Postoperative 6 (21) 3 (11)
Daily exposure (number of subjects) to IV sedatives and
 Neurologic 1 (3) 0 (0) opioids over the first 7 days are shown in Figure 2B.
 Trauma 0 (0) 1 (4)
 Miscellaneous 4 (14) 5 (18) DISCUSSION
Children sedated with dexmedetomidine as the primary sed-
Pediatric Index of Mortality 0.032 (0.034) 0.035 (0.065)
3 risk of death, mean (sd) ative achieved target sedation levels more often in the first 48
hours following initiation of mechanical ventilation when com-
IQR = interquartile range, n = number of children (percentages).
All p were > 0.05 except for age (p = 0. 02). pared with usual care. Children treated with dexmedetomidine
were also more likely to achieve target sedation levels in the first
intervention needed, one was classified as moderate requiring 24 hours, and time from randomization to achieve target seda-
cessation of dexmedetomidine. There were five episodes of mild tion levels was shorter. Children in the dexmedetomidine arm
hypotension (in three patients) in the dexmedetomidine arm, were less likely to receive midazolam, although almost a third
none requiring intervention. These events occurred across all age of these children required the addition of a second-line seda-
ranges (mean age, 27 mo). There was one severe adverse event: a tive during the first 48 hours to achieve target sedation levels
2.5-year-old patient in the dexmedetomidine arm required a brief despite the administration of maximum age-specific doses of
period of cardiopulmonary resuscitation for bradycardia during dexmedetomidine. Dexmedetomidine is known to have both
an airway intervention. This was classified as possibly related to sedative and analgesic properties (31) but not amnestic proper-
the study drug (independent data monitoring and safety commit- ties as described for midazolam. Despite the use of higher age-
tee review). There was no difference in vasopressor use between appropriate maximum doses than previously suggested (based
the groups (p = 0.4). There was no difference in the occurrence on pharmacokinetic/pharmacodynamic [PKPD] analysis)
rate of drug withdrawal or delirium between the groups. in this study (28, 29), it is unclear whether the need for addi-
Feasibility Outcomes. The number of children eligible for tional sedatives (fentanyl, propofol, and ketamine) in a third of
randomization was 254, of which 60 patients (23.6%) were children was due to inadequate sedative, analgesic or amnestic
randomized. The consent rate was 58% (103 families were properties, or inadequate dosing of dexmedetomidine. It is also
approached for consent). unclear whether use of higher dexmedetomidine doses would
Randomization occurred within a median (IQR) of 6.0 be limited by adverse effects, as shown in a minority of patients
hours (2.0–9.0 hr) after intubation in the dexmedetomidine receiving dexmedetomidine in this study.

e734 www.pccmjournal.org September 2020 • Volume 21 • Number 9


Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.
Unauthorized reproduction of this article is prohibited
Online Clinical Investigations

other adverse events such as


accidental removal of airway
or vascular invasive devices
were seen. Data from other
studies suggest that seda-
tive choice may impact on
a pediatric delirium (32).
No differences between the
groups with respect to drug
withdrawal or delirium were
observed in our study, al-
though delirium data were
limited by more than 50% of
children excluded from de-
lirium assessment as they were
less than 6 months old. Hence,
our study cannot make any
Figure 1. Distribution of State Behavioral Scale (SBS) scores over the first 24 and 48 hr in the confident conclusions about
dexmedetomidine arm versus usual care. Values are expressed as percentage (%) of total daily assessments.
the relative occurrence rate of
At the doses used in this study, all but one episode of hy- delirium with dexmedetomidine and other sedatives.
potension or bradycardia in the dexmedetomidine arm were Three previous randomized controlled trials in children
self-limiting and required no specific intervention and overall have compared dexmedetomidine sedation to other agents.
vasopressor requirements were similar between study arms. Tobias and Berkenbosch (33) demonstrated a reduction in
One patient in the dexmedetomidine arm required a brief pe- rescue morphine requirements in children receiving dexme-
riod of cardiac massage for bradycardia during an airway in- detomidine compared with midazolam. Aydogan et al (34)
tervention, and it is possible that dexmedetomidine may have showed that lighter sedation, reduced pain scores, and reduced
contributed to the severity of bradycardia in this case. No fentanyl consumption was achieved with dexmedetomidine

Clinical Outcome Differences Between Those Children Given Dexmedetomidine


TABLE 2.
and Those Managed With Usual Care
Dexmedetomidine, Usual Care,
Clinical Outcomes n = 29 n = 28 p

Time to randomization, hr, median (IQR) 6 (2–9) 3 (1–7) 0.2


Neuromuscular blockade, n (%) 20 (69.0) 17 (60.7) 0.4
Physical restraints, n (%) 10 (34.5) 7 (25.0) 0.4
Vasopressor, n (%) 9 (31.0) 12 (42.9) 0.4
Accidental device removal, n (%) 0 0 Not applicable
Renal replacement therapy, n (%) 0 (0.0) 4 (2.8) 0.2
Bradycardia, n (%) 9 (31.0) 0 (0.0) 0.002
Hypotension, n (%) 5 (17.2) 0 (0.0) 0.05
Withdrawal Assessment Tool-1 > 3, n (%) 5 (17.2) 3 (10.7) 0.5
Preschool Confusion Assessment Method for the ICU/Pediatric 3/18 (16.7) 1/10 (10.0) 0.2
Confusion Assessment Method for ICU positivea, n (%)
Intubation duration, hr, median (IQR) 69.5 (31.2–111.5) 57.3 (43.4–82.5) 0.6
ICU LOS, hr, median (IQR) 115.0 (71.0–219.8) 90.5 (69.1–185.9) 0.5
Hospital LOS, hr, median (IQR) 332.2 (222.4–416.9) 286.5 (163.4–436.9) 0.4
Mortality in ICU, n (%) 0 (0.0) 1 (3.6) 0.5
IQR = interquartile range, LOS = length of stay, n = number of children (percentages).
Twenty-nine points excluded from having assessments as under 6 mo old (11 from dexmedetomidine and 18 from usual care).
a

Although bradycardia was more common in those managed with dexmedetomidine, there was weak evidence for an increased occurrence rate of hypotension.

Pediatric Critical Care Medicine www.pccmjournal.org e735


Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.
Unauthorized reproduction of this article is prohibited
Erickson et al

TABLE 3. Cumulative IV Sedative and Opioid per/kg Body Weight and Duration of
Treatment
Dexmedetomidine, Usual Care,
Drugs Given Dexmedetomidine Usual Care p n = 29, n (%) n = 28, n (%) p

Dexmedetomidine (μg) 47.8 (35.87–70.97) 3.51 (2.96–4.05) 0.005 28 (96.6) 2 (7.1) < 0.0001
  Days on dexmedetomidine 3 (2–5) 0 (0–0) < 0.0001
Midazolam (mg) 0.17 (0.09–0.56) 2.72 (1.16–5.10) 0.002 11 (37.9) 26 (92.9) < 0.0001
  Days on midazolam 0 (0–1) 3 (2–4) < 0.0001
Propofol (mg) 58 (15.65–67.53) 15.42 (8.50–49.25) 0.3 18 (62.1) 11 (39.3) 0.1
  Days on propofol 1 (0–2) 0 (0–1.25) 0.06
Ketamine (mg) 0.84 (0.36–6.21) 0.12 (0.12–0.12) 0.4 8 (27.6) 1 (3.6) 0.03
  Days on ketamine 0 (0–1) 0 (0–0) 0.01
Morphine (mg) 1.90 (1.12–3.96) 1.55 (1.11–2.34) 0.3 26 (89.7) 27 (96.4) 0.6
  Days on morphine 3 (2–5) 3 (2–4.25) 0.7
Fentanyl (μg) 42.2 (25.73–83.54) 21.49 (6.55–53.42) 0.4 16 (55.2) 8 (28.6) 0.06
  Days on fentanyl 1 (0–2) 0 (0–1) 0.03
Cumulative doses shown as median (interquartile range).

compared with midazolam following scoliosis surgery. Prasad on the duration of ventilation in a protocolized nursing se-
et al (35) showed a shorter time to extubation after cessation dation regimen. Nevertheless, protocolized care improved
of dexmedetomidine compared with fentanyl postcardiac sur- achievement of sedation targets. Our study protocol com-
gery. Other data concerning pediatric sedation comprises case pared dexmedetomidine as the primary sedative to the usual
series, case control studies, and surveys (36–40). Our study is care group in which the primary sedative was at the discretion
notable for inclusion of a wide range infants and children in of the treating physician. Although the approach to analgesia,
multiple centers across a region. Our findings are consistent neuromuscular blockade, and sedation monitoring and tar-
with the study by Aydogan et al (34) in that lighter sedation geted sedation levels were identical between the groups, it is
was achieved with dexmedetomidine, In contrast to the study possible that the protocolized dosing regimen used for dex-
by Tobias and Berkenbosch (33), there was no opioid-sparing medetomidine may have improved achievement of sedation
effect and no difference in time to extubation as shown in the targets compared with the usual care group. Alternatively and
study by Prasad et al (35). more likely, the choice of primary sedative or combination of
This pilot study was designed to test the feasibility of a sedative agents were more important factors in achieving the
sedation strategy using dexmedetomidine as the primary lighter sedation levels observed in our study. This is supported
sedative when compared with usual care. Our results demon- by a post hoc analysis of the RESTORE trial by Grant et al
strated that a strategy using dexmedetomidine was feasible, (41), which showed efficacy of dexmedetomidine as a single
practical, and safe. Our study was not blinded as it was felt sedative agent in noninvasively ventilated children.
that blinding in a process of care sedation study would be Our trial was a centrally coordinated, collaborative study
difficult and a potential safety risk, and we acknowledge the with good protocol adherence and successful implementa-
increased potential for bias in our results. Although the use tion of and the randomization and recruitment processes
of sedative agents in the usual care arm was not protocolized by the study team. An important strength of our trial is the
as such, other conduct including management of sedation, early enrollment and commencement of dexmedetomidine
withdrawal and delirium scores in both arms was identical, within hours of initiation of mechanical ventilation. Patients
as was the protocolized use of sedation targets. The dosing of were correctly predicted to require prolonged ventilation,
dexmedetomidine was necessarily prescriptive as it was a rel- with 93% of patients randomized were ventilated for more
atively new drug, it was being used off-label (not approved than 24 hours. Consent rate was high and study participants
by the regulatory body, the Australian Therapeutic Goods were quickly randomized. However, almost 40% of eligible
Administration for use in children). We were using higher children were missed, primarily due to the lack of availability
doses in some age groups, based on PKPD analysis (28, 29), of a research coordinator, competing studies, or ineligibility
and for longer periods than previously described. Curley et due to unit protocols (e.g., individual unit sedation pro-
al (11), in the Randomized Evaluation of Sedation Titration tocol mandating clonidine infusion as primary sedative for
for Respiratory Failure (RESTORE) trial, showed no benefit infants).

e736 www.pccmjournal.org September 2020 • Volume 21 • Number 9


Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.
Unauthorized reproduction of this article is prohibited
Online Clinical Investigations

investigators commented that


the disappointing recruitment
rate reflected a reluctance of
families to provide consent
once sedation had been es-
tablished and a reluctance of
clinicians to recruit unstable
patients. We estimated that a
phase III trial comparing dex-
medetomidine to standard
sedation with a primary out-
come of reduction in duration
of ventilation would require
approximately 600 patients.
Based on our recruitment rate
for this pilot study, recruit-
ment would take many years;
however, we anticipate recruit-
ment would be improved by
better research infrastructure
and resources and by minimiz-
ing exclusion criteria, in par-
ticular those with acute and
chronic brain injury, given the
increased use of dexmedeto-
midine in this patient group in
clinical practice.
This pilot study showed
that randomization of venti-
lated children in intensive care
to alternative sedation arms is
feasible and that a treatment
algorithm with dexmedeto-
midine as the primary seda-
tive agent may be superior to
achieve more rapid safe seda-
tion levels. Although a collab-
orative, binational, multicenter
approach was achievable in this
pilot study, adequate recruit-
ment to a definitive randomized
controlled trial comparing the
efficacy and safety of dexme-
detomidine as the primary sed-
ative, particularly its effect on
Figure 2. Comparison of IV sedative and opioid use between dexmedetomidine and usual care groups.
A, Cumulative total IV sedative and opioid given over the first 48 hr comparing dexmedetomidine to usual care.
patient-centered outcomes, will
All cumulative doses expressed per kg body weight. Median cumulative dose expressed by black bars. likely require a multinational
B, Exposure to IV sedative and opioid drugs over the first 7 d comparing dexmedetomidine to usual care. approach. Notwithstanding
Values are expressed as the total number of children given each drug on each day.
the varied and ever-changing
approaches to sedation in
Randomized trials in pediatric critical care sedation have PICU, and difficulties in gaining consensus on study design,
proven difficult, mainly due to recruitment issues. The mul- a large randomized trial to confirm the main findings of this
ticenter European clonidine study (42) recruited 200 patients pilot study is essential to support evidence-based sedation pro-
in over 5 years, and the U.K. clonidine trial (43) recruited 129 tocols which are safe and effective. Such a study would provide a
patients of a planned 1,000. Both trials were unable to show unique opportunity to understand optimal sedation practice in
positive outcomes due to inadequate sample size. The trial improving long-term outcomes for critically ill children.

Pediatric Critical Care Medicine www.pccmjournal.org e737


Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.
Unauthorized reproduction of this article is prohibited
Erickson et al

The Baby SPICE Investigators and the Australian and New Zealand In- 14. Taffarel P, Bonetto G, Jorro Barón F, et al: Sedation and analgesia in
tensive Care Society Paediatric Study Group (ANZICS PSG) are listed patients on mechanical ventilation in pediatric intensive care units in
in Appendix 1. Argentina. Arch Argent Pediatr 2018; 116:e196–e203
Supplemental digital content is available for this article. Direct URL cita- 15. Young C, Jevtovic-Todorovic V, Qin YQ, et al: Potential of ketamine
tions appear in the printed text and are provided in the HTML and PDF and midazolam, individually or in combination, to induce apoptotic
versions of this article on the journal’s website (http:/journals.lww.com/ neurodegeneration in the infant mouse brain. Br J Pharmacol 2005;
pccmjournal). 146:189–197
The study was aided by Perth Children’s Hospital Foundation Grant and 16. Jevtovic-Todorovic V, Hartman RE, Izumi Y, et al: Early exposure to
common anesthetic agents causes widespread neurodegeneration in
Hospira (Pfizer; unrestricted study grant).
the developing rat brain and persistent learning deficits. J Neurosci
Drs. Millar’s and Long’s institution received funding from Princess Mar- 2003; 23:876–882
garet Hospital Foundation. Drs. Millar’s, Festa’s, Shehabi’s, and Long’s 17. Colville G, Kerry S, Pierce C: Children’s factual and delusional memo-
institutions received funding from Pfizer (Hospira). Dr. Festa disclosed ries of intensive care. Am J Respir Crit Care Med 2008; 177:976–982
off-label product use of dexmedetomidine infusion. Dr. Shehabi received
18. Mody K, Kaur S, Mauer EA, et al: Benzodiazepines and development
competitive grant funding from the National Health and Medical Research
of delirium in critically ill children: Estimating the causal effect. Crit
Council of Australia. The remaining authors have disclosed that they do
Care Med 2018; 46:1486–1491
not have any potential conflicts of interest.
19. Amigoni A, Vettore E, Brugnolaro V, et al: High doses of benzodiaze-
For information regarding this article, E-mail: simon.erickson@health. pine predict analgesic and sedative drug withdrawal syndrome in pae-
wa.gov.au diatric intensive care patients. Acta Paediatr 2014; 103:e538–e543
20. Best KM, Boullata JI, Curley MA: Risk factors associated with iatro-
genic opioid and benzodiazepine withdrawal in critically ill pediatric
REFERENCES patients: A systematic review and conceptual model. Pediatr Crit
1. Playfor S, Jenkins I, Boyles C, et al; United Kingdom Paediatric Care Med 2015; 16:175–183
Intensive Care Society Sedation; Analgesia and Neuromuscular 21. Traube C, Silver G, Reeder RW, et al: Pediatric delirium in critically-
Blockade Working Group: Consensus guidelines on sedation ill children: An international point prevalence study. Crit Care Med
and analgesia in critically ill children. Intensive Care Med 2006; 2017; 45:584–590
32:1125–1136 22. Hayden JC, Breatnach C, Doherty DR, et al: Efficacy of α2-agonists
2. Playfor SD, Thomas DA, Choonara I: Sedation and neuromuscular for sedation in pediatric critical care: A systematic review. Pediatr Crit
blockade in paediatric intensive care: A review of current practice in Care Med 2016; 17:e66–e75
the UK. Paediatr Anaesth 2003; 13:147–151 23. Shann F: Drug Doses. 17th Edition. Melbourne, VIC, Australia,
3. Rhoney DH, Murry KR: National survey on the use of sedatives and Collective, 2017
neuromuscular blocking agents in the pediatric intensive care unit. 24. Curley MA, Harris SK, Fraser KA, et al: State Behavioral Scale (SBS)
Pediatr Crit Care Med 2002; 3:129–133 a sedation assessment instrument for infants and young children sup-
ported on mechanical ventilation. Pediatr Critical Care Med 2006;
4. Marx CM, Rosenberg DI, Ambuel B, et al: Pediatric intensive care
7:107–114
sedation: Survey of fellowship training programs. Pediatrics 1993;
91:369–378 25. Franck LS, Harris SK, Soetenga DJ, et al: The Withdrawal Assessment
Tool-1 (WAT-1): An assessment instrument for monitoring opioid and
5. Twite MD, Rashid A, Zuk J, et al: Sedation, analgesia, and neuromus- benzodiazepine withdrawal symptoms in pediatric patients. Pediatr
cular blockade in the pediatric intensive care unit: Survey of fellow- Crit Care Med 2008; 9:573–580
ship training programs. Pediatr Crit Care Med 2004; 5:521–532
26. Franck LS, Scoppettuolo LA, Wypij D, et al: Validity and generalizability
6. Vet NJ, Ista E, de Wildt SN, et al: Optimal sedation in pediatric inten- of the Withdrawal Assessment Tool-1 (WAT-1) for monitoring iatrogenic
sive care patients: A systematic review. Intensive Care Med 2013; withdrawal syndrome in pediatric patients. Pain 2012; 153:142–148
39:1524–1534
27. Smith HA, Gangopadhyay M, Goben CM, et al: The Preschool
7. Long D, Erickson S; for the ANZICS Paediatric Study Group: Baby Confusion Assessment Method for the ICU (psCAM-ICU): Valid and
Spice: Sedation practice in paediatric intensive care evaluation in reliable delirium monitoring for critically ill infants and children. Crit
Australia and New Zealand. Pediatr Crit Care Med 2014; 15:19 Care Med 2016; 44:592–600
(abstract) 28. Smith HA, Boyd J, Fuchs DC, et al: Diagnosing delirium in critically ill
8. Poh YN, Poh PF, Buang SN, et al: Sedation guidelines, protocols, children: Validity and reliability of the pediatric confusion assessment
and algorithms in PICUs: A systematic review. Pediatr Crit Care Med method for the intensive care unit. Crit Care Med 2011; 39:150–157
2014; 15:885–892 29. Potts AL, Anderson BJ, Warman GR, et al: Dexmedetomidine phar-
9. Keogh SJ, Long DA, Horn DV: Practice guidelines for sedation and macokinetics in pediatric intensive care–a pooled analysis. Paediatric
analgesia management of critically ill children: A pilot study evaluat- Anaesth 2009; 19:1119–1129
ing guideline impact and feasibility in the PICU. BMJ Open 2015; 30. Potts AL, Warman GR, Anderson BJ: Dexmedetomidine disposition in
5:e006428 children: A population analysis. Paediatr Anaesth 2008; 18:722–730
10. Gaillard-Le Roux B, Liet J-M, Bourgoin P, et al: Implementation of a 31. Gertler R, Brown HC, Mitchell DH, et al: Dexmedetomidine: A
nurse-driven sedation protocol in a PICU decreases daily doses of novel sedative-analgesic agent. Proc (Bayl Univ Med Cent) 2001;
midazolam. Pediatr Critical Care Med 2017; 18:e9–e17 14:13–21
11. Curley MA, Wypij D, Watson RS, et al; RESTORE Study Investigators 32. Smith HAB, Gangopadhyay M, Goben CM, et al: Delirium and benzo-
and the Pediatric Acute Lung Injury and Sepsis Investigators Network: diazepines associated with prolonged ICU stay in critically ill infants
Protocolized sedation vs usual care in pediatric patients mechanically and young children. Crit Care Med 2017; 45:1427–1435
ventilated for acute respiratory failure: A randomized clinical trial. 33. Tobias JD, Berkenbosch JW: Sedation during mechanical ventilation
JAMA 2015; 313:379–389 in infants and children: Dexmedetomidine versus midazolam. South
12. Kudchadkar SR, Yaster M, Punjabi NM: Sedation, sleep promotion, Med J 2004; 97:451–455
and delirium screening practices in the care of mechanically venti- 34. Aydogan MS, Korkmaz MF, Ozgül U, et al: Pain, fentanyl consumption,
lated children: A wake-up call for the pediatric critical care commu- and delirium in adolescents after scoliosis surgery: Dexmedetomidine
nity*. Crit Care Med 2014; 42:1592–1600 vs midazolam. Paediatr Anaesth 2013; 23:446–452
13. Garcia Guerra G, Joffe AR, Cave D, et al; Sedation Withdrawal and 35. Prasad SR, Simha PP, Jagadeesh AM: Comparative study between
Analgesia Team, and the Canadian Critical Care Trials Group: Survey dexmedetomidine and fentanyl for sedation during mechanical venti-
of sedation and analgesia practice among Canadian pediatric critical lation in post-operative paediatric cardiac surgical patients. Indian J
care physicians. Pediatr Crit Care Med 2016; 17:823–830 Anaesth 2012; 56:547–552

e738 www.pccmjournal.org September 2020 • Volume 21 • Number 9


Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.
Unauthorized reproduction of this article is prohibited
Online Clinical Investigations

36. Czaja AS, Zimmerman JJ: The use of dexmedetomidine in critically ill 41. Grant MJ, Schneider JB, Asaro LA, et al; Randomized Evaluation
children. Pediatr Crit Care Med 2009; 10:381–386 of Sedation Titration for Respiratory Failure Study Investigators:
37. Chrysostomou C, Di Filippo S, Manrique AM, et al: Use of dexme- Dexmedetomidine use in critically ill children with acute respiratory
detomidine in children after cardiac and thoracic surgery. Pediatr Crit failure. Pediatr Crit Care Med 2016; 17:1131–1141
Care Med 2006; 7:126–131
42. Hünseler C, Balling G, Röhlig C, et al; Clonidine Study Group:
38. Hosokawa K, Shime N, Kato Y, et al: Dexmedetomidine sedation in Continuous infusion of clonidine in ventilated newborns and
children after cardiac surgery. Pediatr Crit Care Med 2010; 11:39–43
infants: A randomized controlled trial. Pediatr Crit Care Med 2014;
39. Bejian S, Valasek C, Nigro JJ, et al: Prolonged use of dexmedeto-
15:511–522
midine in the paediatric cardiothoracic intensive care unit. Cardiol
Young 2009; 19:98–104 43. Wolf A, McKay A, Spowart C, et al: Prospective multicentre ran-
40. Kleiber N, de Wildt SN, Cortina G, et al: A comparative analysis of domised, double-blind, equivalence study comparing clonidine and
preemptive versus targeted sedation on cardiovascular stability after midazolam as intravenous sedative agents in critically ill children: The
high-risk cardiac surgery in infants. Pediatr Crit Care Med 2016; SLEEPS (Safety profiLe, Efficacy and Equivalence in Paediatric inten-
17:321–331 sive care Sedation) study. Health Technol Assess 2014; 18:1–212

APPENDIX 1. BABY SPICE INVESTIGATORS The Australian and New Zealand Intensive Care So-
AND THE AUSTRALIAN AND NEW ZEALAND ciety Paediatric Study Group (ANZICS PSG) are as fol-
INTENSIVE CARE SOCIETY PAEDIATRIC lows: Anusha Ganeshalingam, Claire Sherring, Starship
STUDY GROUP (ANZICS PSG) Children’s Hospital, Auckland, New Zealand; Simon Erick-
The Baby SPICE Investigators are as follows: Brian Anderson, son, Samantha Barr, Perth Children`s Hospital, Perth, Aus-
Claire Sherring (Starship Hospital, Auckland, New Zealand), tralia; Andreas Schibler, Debbie Long, Luregn Schlapbach
Debbie Long, Anthony Slater (Queensland Children’s Hospital, (Chair), Jan Alexander (ANZPIC registry), Queensland
Brisbane, QLD, Australia), Johnny Millar, Carmel Delzoppo Children’s Hospital, Brisbane, Australia; Shane George,
(Royal Children’s Hospital, Melbourne, VIC, Australia), Marino Gold Coast University Hospital; Gary Williams, Vicky
Festa (Children’s Hospital at Westmead, Westmead, NSW, Aus- Smith, Sydney Children’s Hospital, Randwick, Australia;
tralia), Mary Lou Morritt, Janelle Young, John Awad (Sydney Warwick Butt, Carmel Delzoppo, Johnny Millar (ANZPIC
Children’s Hospital, Sydney, NSW, Australia), Simon Erickson, registry lead), Ben Gelbart (Vice Chair), Royal Children’s
Sara Shea, Samantha Barr (Perth Children’s Hospital, Perth, Hospital, Melbourne, Australia; Felix Oberender, Monash
WA, Australia), Belinda Howe (Australian New Zealand Inten- Children`s Hospital, Melbourne, Australia; Subodh Ganu,
siveCare Research Centre, School of Public Health and Preven- Georgia Letton, Women’s and Children’s Hospital, Ad-
tive Medicine, Monash University, Melbourne, VIC, Australia), elaide, Australia; Jonathan Egan, Gail Harper, Marino
and Elizabeth MacKinnon (Statistics-Telethon Kids Institute: Festa (Past Chair), Westmead Children’s Hospital, Sydney,
Child and Adolescent Health Service, Perth, WA, Australia). Australia.

Pediatric Critical Care Medicine www.pccmjournal.org e739


Copyright © 2020 by the Society of Critical Care Medicine and the World Federation of Pediatric Intensive and Critical Care Societies.
Unauthorized reproduction of this article is prohibited

You might also like