Skip to main content
Erschienen in: Annals of Intensive Care 1/2018

Open Access 01.12.2018 | Research

Dexmedetomidine in prevention and treatment of postoperative and intensive care unit delirium: a systematic review and meta-analysis

verfasst von: Julian Flükiger, Alexa Hollinger, Benjamin Speich, Vera Meier, Janna Tontsch, Tatjana Zehnder, Martin Siegemund

Erschienen in: Annals of Intensive Care | Ausgabe 1/2018

Abstract

Background

To determine the preventive and therapeutic effect of dexmedetomidine on intensive care unit (ICU) delirium.

Methods

The literature search using PubMed and the Cochrane Central Register of Controlled Trials was performed (August 1, 2018) to detect all randomized controlled trials (RCTs) of adult ICU patients receiving dexmedetomidine. Articles were included if they assessed the influence of dexmedetomidine compared to a sedative agent on incidence of ICU delirium or treatment of this syndrome. Accordingly, relevant articles were allocated to the following two groups: (1) articles that assessed the delirium incidence (incidence comparison) or articles that assessed the treatment of delirium (treatment comparison). Incidence of delirium and delirium resolution were the primary outcomes. We combined treatment effects comparing dexmedetomidine versus (1) placebo, (2) standard sedatives, and (3) opioids in random-effects meta-analyses. Risk of bias for each included RCT was assessed following Cochrane standards.

Results

The literature search resulted in 28 articles (25 articles/4975 patients for the incidence comparison and three articles/166 patients for the treatment comparison). In the incidence comparison, heterogeneity was present in different subgroups. Administration of dexmedetomidine was associated with significantly lower overall incidence of delirium when compared to placebo (RR 0.52; 95% CI 0.39–0.70; I2 = 37%), standard sedatives (RR 0.63; 95% CI 0.46–0.86; I2 = 69%), as well as to opioids (RR 0.61; 95% CI 0.44–0.83; I2 = 0%). Use of dexmedetomidine significantly increased the risks of bradycardia and hypotension. Limited data were available on circulatory insufficiency and mortality. In the treatment comparison, the comparison drugs in the three RCTs were placebo, midazolam, and haloperidol. The resolution of delirium was measured differently in each study. Two out of the three studies indicated clear favorable effects for dexmedetomidine (i.e., compared to placebo and midazolam). The study comparing dexmedetomidine with haloperidol was a pilot study (n = 20) with high variability in the results.

Conclusion

Findings suggest that dexmedetomidine reduces incidence and duration of ICU delirium. Furthermore, our systematic searches show that there is limited evidence if a delirium shall be treated with dexmedetomidine.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s13613-018-0437-z) contains supplementary material, which is available to authorized users.
Julian Flükiger and Alexa Hollinger have contributed equally to study design and writing of the manuscript
Abkürzungen
CAM
confusion assessment method
CENTRAL
Cochrane Central Register of Controlled Trials
CI
confidence interval
GABA
gamma-amino-butyric acid
ICDSC
intensive care delirium screening checklist
ICU
intensive care unit
IQR
interquartile range
MV
mechanical ventilation
OR
odds ratio
PAD
pain, agitation, and delirium
RCT
randomized controlled trial
SD
standard deviation

Background

Delirium is experienced in 20% to 40% of the critically ill and up to 80% of mechanically ventilated (MV) medical or surgical patients [14]. It is a confusional state that has been described as a transient global disorder of cognition, awareness, and attention and as such is not only challenging for the treating medical team, but also has a considerable impact on affected patients. It is associated with prolonged hospital length of stay and time on MV, deterioration in cognition, and increased morbidity and mortality causing additional health-care expenses [46]. Its pathophysiological mechanisms are highly heterogeneous—as perceived by the high number of risk factors [7]—and have yet to be fully understood. Imbalanced neurotransmitter systems such as the reduction of acetylcholine activity, excess of serotonin and dopamine, or the release of gamma-amino-butyric acid (GABA) seem essential. Certain hospital care characteristics (patient immobilization, bladder catheter, or sleep deprivation) further contribute to the development of delirium [8, 9].
Treatment approach with sedation offers several benefits: First, it reduces discomfort, anxiety, and stress. Second, it facilitates the daily intensive care unit (ICU) procedures for the treating nurses and physicians, providing a calm, cooperative patient who is easy to rouse and capable of communicating pain and other needs [1012]. So far, there is no ideal sedative agent that fulfills the criteria of being cheap, rapid in onset and offset, and without local or systemic adverse effects [10].
Although the 2013 PAD guidelines suggested against the use of antipsychotics such as haloperidol for the treatment of delirium due to their side effects (e.g., extrapyramidal symptoms, neuroleptic malignant syndrome, and QTc interval prolongation), they are still commonly applied today [1, 13, 14]. Moreover, the most recent 2013 American College of Critical Care Medicine Guidelines for the management of pain, agitation, and delirium (PAD) in adult ICU patients recommend the use of a non-benzodiazepine-based sedation approach [15].
Dexmedetomidine with its broad range of effects including easily controllable sedation, analgesia, and anxiolysis still enables the caring medical team to interact with the patient. It reduces the activity while still maintaining the reactivity of neurons in the locus coeruleus. Therefore, it is an appealing alternative to traditional sedatives such as propofol and benzodiazepines [16]. As a highly selective \(\propto\)2-receptor agonist with no effect on the GABA receptor, it interacts with transmembrane G-protein-binding adrenoreceptors in the periphery (\(\propto\)2A), as well as in the brain (\(\propto\)2B) and the spinal cord (\(\propto\)2C). Inducing a sleep-like state without respiratory depression may explain the beneficial effects of dexmedetomidine [17, 18], since disturbed circadian rhythm is a known contributing factor of delirium [19]. These characteristics lead to an easily arousable, communicative, and cooperative patient and render dexmedetomidine a potential therapeutic option for the ICU delirium, in addition to its suggested use for delirium prevention [16, 20]. However, inhibition of sympathetic activity in the periphery leads to sequential decreases in blood pressure and heart rate [19], the most commonly reported adverse events associated with dexmedetomidine [21].
A review from 2013 suggested dexmedetomidine could be suitable for both prevention and treatment of ICU-associated delirium [14]. In 2014, a systematic review from Pasin et al. [20] including 14 RCTs with a total of 3029 patients demonstrated that use of dexmedetomidine for anesthesia and sedation was associated with a significant reduction of the incidence of delirium. Similar results were demonstrated in a previous review from Xia et al. where only propofol was used as a comparator [22]. Very recently, Duan et al. [23] performed a meta-analysis, showing that dexmedetomidine can reduce postoperative delirium incidence in adult cardiac and non-cardiac surgery patient. Based on fact that in the meantime a number of additional randomized controlled trials (RCTs) were published, we wanted to provide an up-to-date meta-analysis on dexmedetomidine for both treatment and prevention of ICU delirium to define areas of future research concerning ICU delirium and dexmedetomidine treatment. Randomized controlled trials (RCTs) were included, assessing the effect of dexmedetomidine on delirium in adult ICU patients. The goal was to summarize current evidence on the potential of dexmedetomidine to lower the incidence and duration of ICU delirium.

Methods

For this systematic review and meta-analysis, we adhered to the PRISMA statement for reporting systematic reviews and meta-analyses [24].
Authors performed an electronic database search of PubMed and the Cochrane Central Register of Controlled Trials (CENTRAL). The detailed search strategy is available in appendix (Additional file 1: Appendix). We used standard filters to search for RCTs. The last date of search was August 1, 2018.

Inclusion criteria

Two independent investigators (JF and AH) identified all potentially relevant studies in PubMed and CENTRAL based on a screening of titles and abstracts. Of these, full texts were obtained and reviewed for eligibility by the same two investigators. Any conflict of opinion was resolved by consensus with a third reviewer (MS).
All studies that met the following inclusion criteria were selected: (1) RCT; (2) adult (≥ 18 years) medical or surgical ICU patients; (3) sedation with dexmedetomidine versus any comparator either for prevention or treatment of ICU delirium, regardless of dose, duration, or time of administration; (4) incidence of delirium as a mandatory outcome measurement regarding incidence comparison; and (5) full text available in English. The exclusion criteria were duplicate publications, missing indication of delirium incidence, study design other than RCT, focus on withdrawal delirium, oral administration of dexmedetomidine, or dexmedetomidine administered in both intervention and control groups.

Data extraction

Study-relevant information was extracted by two independent investigators (JF and AH) for each included RCT. Any conflict of opinion was resolved by consensus with a third reviewer (MS).
Interventional (dexmedetomidine) and control drugs, publication date, study location and date of study conduct, as well as patient characteristics, total number of patients, and conducted procedure were considered relevant for data extraction. Control drugs were divided into three groups: placebo, standard sedatives (including propofol, midazolam, and lorazepam), and opioids (including morphine and remifentanil). Delirium assessment tools (e.g., confusion assessment method for the ICU = CAM–ICU or the intensive care delirium screening checklist = ICDSC) were assessed. Furthermore, we recorded if delirium was assessed as the primary, a secondary, or no specific endpoint.
Although our inclusion criteria focused only on the incidence of delirium, we also conducted further analyses of other patient-relevant factors, such as adverse events (i.e., mortality, bradycardia, tachycardia, hypotension, hypertension, and circulatory insufficiency) and clinical outcome data (i.e., ICU length of stay, and time to extubation or duration of MV). These analyses have only an explorative character and raise no claim to completeness.
According to whether dexmedetomidine and control drugs were applied for treatment or prevention of ICU delirium, included trials were assigned to incidence comparison or delirium treatment (treatment comparison) and analyzed according to their affiliation.
For assessment of adverse events, we also considered studies reporting adverse events during anesthesia and subsequent intensive care sedation to increase the reliability of the study since only a few studies reported adverse events during intensive care sedation with dexmedetomidine.
The primary outcome measure in incidence comparison was incidence of delirium. The primary outcome measure in treatment comparison was delirium resolution.

Risk of bias assessment

Risk of bias for each included RCT was assessed using the Cochrane Collaboration’s tool [25]. Two investigators (JF and BS) reviewed each RCT for risk of bias considering the following five key domains: (1) random sequence generation (selection bias); (2) allocation concealment (selection bias); (3) blinding of participants and personnel (performance bias); (4) blinding of outcome assessment (detection bias); and (5) incomplete outcome data (attrition bias). Each domain was judged as “low risk,” “high risk,” or “unclear risk” of bias. Any disagreement in opinion was resolved by consensus. We estimated the overall risk of bias for a RCT as low if the risk of bias was low in all key domains, as unclear if the risk of bias was unclear in at least one key domain, and as high if the risk of bias was high in at least one key domain.

Statistical analysis

Meta-analyses were conducted using the Cochrane Review Manager 5 (RevMan5). Differences in binary outcomes were presented as risk ratios (RR) including 95% confidence intervals (CI) based on random and fixed effect models. RRs below 1 indicate that the event occurred less often in patients who received dexmedetomidine compared to patients who received a control drug. In incidence comparison, meta-analyses were performed to compare the incidence of delirium, adverse events, and clinical outcomes for each control drug separately. For the incidence of delirium, the following subgroup and sensitivity analyses were conducted: analyzing only studies with low risk of bias according to risk of bias assessment, population (i.e., cardiac surgery, MV, and non-cardiac surgery), and using a trim-and-fill method [26, 27] if there was an indication of publication bias.
Higgin’s I2 was conducted to assess the heterogeneity of the studies [28]. If a subgroup consisted of sufficient RCTs, Egger’s test was performed and funnel plot was visually assessed to determine the risk of publication bias. Symmetric statistics and trim-and-fill methods were conducted using R version 3.4.1 (meta package) following the Cochrane recommendations [29]. Furthermore, meta-analyses were conducted for the following adverse events and clinical outcomes: mortality, bradycardia, tachycardia, hypotension, hypertension, circulatory insufficiency, ICU length of stay, and time to extubation or duration of MV. If both time to extubation and duration of MV were presented, we used time to extubation data for the meta-analyses. In case no means and standard deviations (SDs) were reported (for ICU length of stay, time to extubation or duration of MV), we used medians, interquartile ranges (IQRs), or ranges to estimate these attributes, forcing us to make several assumptions (Additional file 1: Tables S1–S3) [30, 31].
Mortality rates assessed up to 45 days and the remaining adverse events were pooled regardless of the time they occurred (intraoperatively, postoperatively, during sedation or MV).
For treatment comparison, all articles reported different outcomes. Therefore, results are summarized descriptively and no meta-analysis was performed.

Results

Trial identification

After excluding duplicate studies, our literature search on PubMed and CENTRAL retrieved 232 articles. By screening titles and abstracts, 171 publications were excluded at the title abstract level and an additional 33 were excluded after consulting the full text (Fig. 1). The remaining 28 articles were included and divided into incidence comparison consisting of 25 RCTs [3256] and treatment comparison consisting of three articles [5759] (Fig. 1).

Study characteristics

In incidence comparison, all RCTs were published between 2005 and 2018 (being conducted between 2002 and 2017; Table 1). Two RCTs did not state study dates [39, 40]. Sixteen RCTs dealt with surgical patients, nine of which were cardiac surgery RCTs [36, 39, 41, 42, 4547, 51, 53] and seven non-cardiac surgery RCTs, meaning microvascular free flap surgery [34], joint replacement [49] or total hip arthroplasty [54], major abdominal surgery [52], major laparoscopic surgery [55], and elective non-cardiac surgery under general anesthesia [33, 48]. The remaining nine RCTs consisted of six studies examining MV patients [32, 38, 40, 43, 44, 50] two examining noninvasively ventilated patients [35, 37] and one where dexmedetomidine was used for nightly sedation [56]. Applied procedures for administering intervention and control drugs were highly heterogeneous (Table 1). Comparators were placebo in eight RCTs [3335, 45, 48, 51, 55, 56], propofol in six RCTs [42, 46, 47, 5254], midazolam in two RCTs [37, 50], and lorazepam [32], morphine [41], and remifentanil [36] in one RCT each. All of these RCTs used a two-arm design. Two RCTs compared dexmedetomidine sedation to a sedation regimen of either propofol or midazolam [43, 44]. Two RCTs used a three-arm design comparing dexmedetomidine to midazolam and propofol [39, 40]. One RCT used a two-arm design comparing dexmedetomidine to placebo, but did not report overall results. Instead, only results for the two subgroups “normal-aged” patients and patients with amnestic mild cognitive impairment (aMCI) after joint replacement were reported [49]. One article consisted of two separate RCTs comparing dexmedetomidine to midazolam and propofol, respectively [38]. The total number of patients in incidence comparison was 4975. The used tools for delirium assessment are listed in Table 1. In treatment comparison, all RCTs were published between 2009 and 2016 (study dates ranged from 2005 to 2013; Table 1). Study populations of all RCTs consisted of patients with agitated delirium. Two RCTs dealt with MV patients in whom extubation was considered unsafe because of agitated delirium [57, 59]. One RCT examined patients presenting with delirium following extubation failure after cardiac surgery [58]. Interventional and control drugs were administered according to the desired RASS score [57, 59], except for one RCT, where they were administered according to blood pressure and heart rate [58]. Comparators were placebo [57], midazolam [58], or haloperidol [59]. The total number of patients in treatment comparison was 166. Delirium assessment was performed using validated tools (Table 1).
Table 1
Study characteristics of all included RCTs
 
Study
Study location
Study date
Population
Procedure
Control medication
Total number of patients
Mean age (years)
Male (%)
Delirium assessment
Endpoint delirium
Sedation comparison
Chang [52]
Taiwan
2014–2015
Non-cardiac surgery
Postoperatively for a maximum of 24 h, RASS-2-0
Propofol
60
70.5
58.3
CAM-ICU
2
Lee [55]
South Korea
2016–2017
Non-cardiac surgery
Intraoperatively as continuous infusion or bolus
Placebo
318
73
44.3
CAM
1
Mei [54]
China
2016–2017
Non-cardiac surgery
Intraoperatively as bolus and subsequent continuous infusion
Propofol
296
75
45.6
CAM
1
Sheikh [53]
India
2014–2016
Cardiac surgery
Intraoperatively as bolus and subsequent continuous infusion
Propofol
60
34.6
N/A
*
2
Skrobik [56]
USA
2013–2016
MV**
9:30 in the evening until 6:15 the next morning, continuous infusion without bolus, RASS-1
Placebo
100
62.3
64
ICDSC
1
Deiner [33]
USA
2008–2014
Non-cardiac surgery
Entering OR until 2 h in recovery room
Placebo
390
74***
48.7
CAM CAM-ICU
1
Kawazoe [44]
Japan
2013–2016
MV
Continuously until end of MV, RASS-0 by day, RASS-2 by night
Propofol midazolam
201
68.5
63.2
CAM
2
Li [45]
China
2014–2015
Cardiac surgery
Intraoperatively and continuously until end of MV
Placebo
285
67
69.1
CAM CAM-ICU
1
Li [43]
China
2015–2016
MV
Sedation for at least 48 h until end of MV, RASS-0–1 in intervention, RASS − 2/− 3 in control group
Propofol, midazolam
126
44
56.3
CAM-ICU
1
Djaiani [46]
Canada
2011–2014
Cardiac surgery
Postoperatively and continuously until end of MV
Propofol
183
72.6
75.4
CAM CAM-ICU
1
Liu [47]
China
2015
Cardiac surgery
Postoperatively on ICU admission and continuously until end of MV, RASS-3-0
Propofol
61
54***
41
CAM
2
Liu [49]
China
2014–2016
Non-cardiac surgery
Intraoperatively until 20 min before end of surgery
Placebo
197
73
48.7
CAM
1
Su [58]
China
2011–2013
Non-cardiac surgery
Postoperatively until 8:00 the next morning
Placebo
700
74.4
60.4
CAM
1
MacLaren [50]
USA
2009–2012
MV
Continuously until end of MV, according to desired SAS
Midazolam
23
58.1
56.5
ICDSC
2
Priye [51]
India
2012–2013
Cardiac surgery
Postoperatively for 12 h
Placebo
64
43.3
51.6
RASS
2
Yang [34]
China
2013
Non-cardiac surgery
Intraoperatively until 1 h before end of surgery and postoperatively until 6:00 the next morning
Placebo
79
50.5
53.2
CAM-ICU
2
Devlin [35]
USA
2008–2012
NIV
2 h stable after NIV mask removal, SAS 3–4
Placebo
33
65
51.5
ICDSC
2
Park [36]
South Korea
2012–2013
Cardiac surgery
Postoperatively until end of MV, RSS 2–3
Remifentanil
142
52.7
55.6
CAM-ICU
1
Huang [37]
China
2008–2011
NIV
Until end of sedation, RSS 2–3
Midazolam
62
64.5
41.9
N/A
2
Jakob [38]
Europe + Russia
2007–2010
MV
Continuously until end of MV, RASS-3-0
Propofol, midazolam
998
65***
65.5
CAM-ICU
2
Maldonado [39]
USA
N/A
Cardiac surgery
Postoperatively until end of MV
Propofol, midazolam
118
57.7
63.6
CAM CAM-ICU
1
Ruokonen [40]
Switzerland + Finland
N/A
MV
Continuously until end of MV, according to desired RASS
Propofol, midazolam
85
66***
82.4
CAM-ICU
2
Shehabi [41]
Australia
2004–2007
Cardiac surgery
Postoperatively until ICU discharge, MAAS 2–4
Morphine
299
71.3***
75.3
CAM-ICU
1
Pandharipande [32]
USA
2004–2006
MV
Continuously until end of MV, according to desired RASS
Lorazepam
103
59.5***
51.5
CAM-ICU
1
Corbett [42]
USA
2002–2004
Cardiac surgery
Postoperatively until end of MV, RSS 5 the first 2 h, RSS 3–4 afterward
Propofol
89
63
82
N/A
3
Treatment comparison
Reade [57]
Australia + New Zealand
2011–2013
AD (MV)
According to desired RASS = 0
Placebo
74
57.3***
75.7
CAM-ICU
3
Yapici 2010
Turkey
2005–2007
AD (cardiac surgery)
According to blood pressure and heart rate
Midazolam
72
60
37.5
CAM-ICU
3
Reade [59]
Australia
2006–2008
AD (MV)
According to desired RASS = 0
Haloperidol
20
60.3***
85
ICDSC
3
Incidence comparison, normal matter; treatment comparison, italic matter
AD agitated delirium, CAM confusion assessment method, CAM-ICU confusion assessment method for intensive care unit, h hours, ICDSC intensive care delirium screening checklist, MAAS Motor Activity Assessment Scale, MV mechanical ventilation, N/A not available, NIV noninvasive ventilation, OR operating room, RASS Richmond Agitation and Sedation Scale, RSS Ramsay Sedation Scale, SAS Riker Sedation and Agitation Scale
1, primary endpoint; 2, secondary endpoint; 3, no specific endpoint
*Delirium defined as shourt course illusions, confusion, and cerebral excitement in the postoperative period; **Most; ***Median age
Further information on the drug doses used throughout the studies is provided in Additional file 2: Table S4.

Quality of evidence

The Cochrane Collaboration’s risk of bias assessment is provided in Table 2. Six [56] articles consisting of seven RCTs in the incidence comparison had low risk of bias [3335, 38, 45]. Nine RCTs had an unclear risk of bias in at least one domain due to poor reporting (1808 patients) [32, 36, 37, 40, 41, 48, 50, 51, 53], and ten RCTs had a high risk of bias in at least one of the five assessed domains [39, 4244, 46, 47, 49, 52, 54, 55]. In the treatment comparison, there was one RCT with low risk of bias [57], one with unclear risk of bias in all five domains [58], and one with high risk of bias in two of the five domains [59].
Table 2
Risk of bias assessment of included RCTs
https://static-content.springer.com/image/art%3A10.1186%2Fs13613-018-0437-z/MediaObjects/13613_2018_437_Tab2_HTML.png
aPatient blinded
bBlocked randomization in unblinded trial
cPatients with delayed recovery were excluded
dStudy reported as double blind; it is likely that patients and personnel were blinded, but this is unclear for the outcome assessor
eStudy reported as double blind, but blinding of patients is not explicitly mentioned
fNo flow chart provided; unclear if more patients were originally included

Incidence comparison

Nine articles consisting of ten data sets compared dexmedetomidine to placebo; 14 articles consisting of 15 data sets compared dexmedetomidine to standard sedatives; and two RCTs (two data sets) compared dexmedetomidine to opioids (Table 3).
Table 3
Subgroup and sensitivity analyses for incidence of delirium
Subgroup and sensitivity analyses
Number of data sets
Number of patients
Risk ratio (95% CI) random effect model
I 2
Placebo
Overall
10
2071
0.52 (0.42–0.63)
37%
Studies with no risk of bias according to our risk of bias assessment
5
887
0.66 (0.43–1.03)
20%
Cardiac surgery
2
349
0.53 (0.22–1.24)
1%
Mechanical ventilation
0
   
Non-cardiac surgery
6
1589
0.53 (0.35–0.80)
61%
Standard sedatives
Overall
15
2463
0.63 (0.46–0.86)
69%
Studies with no risk of bias according to our risk of bias assessment
2
998a*
0.64 (0.41–0.98)
0%
Cardiac surgery
5
509
0.40 (0.23–0.69)
11%
Mechanical ventilation
7
1536
0.82 (0.60–1.11)
66%
Non-cardiac surgeryb
2
356
0.46 (0.23–0.90)
NA
Trim-and-fill method
20c
0.77 (0.57–1.05)
64%
Opioids
Overall
2
441
0.61 (0.44–0.83)
0%
aPropofol or midazolam was used in the comparison group
bOne out of two studies had no events
cFive hypothetical studies added with by a trim-and-fill function or R package meta

Incidence of delirium

Overall incidence of delirium in the dexmedetomidine group was significantly lower compared to placebo (RR 0.52; 95% CI 0.39–0.70; I2 = 37%; Fig. 2), compared to standard sedatives (RR 0.63; 95% CI 0.46–0.86; I2 = 69%; Fig. 3), as well as compared to opioids (RR 0.61; 95% CI 0.44–0.83; I2 = 0%; Fig. 4). When only RCTs with a low risk of bias were included, we retrieved a RR of 0.66 (95% CI 0.43–1.03; five data sets included) for the placebo comparison and a RR of 0.64 (0.41–0.98; two data sets included) for the standard sedative comparison (Table 3). It is important to mention that the two data sets from the standard sedatives comparison were presented in the same article [38]. In the opioids group, there was no RCT without risk of bias.
Visual inspection of the funnel plot from the analysis of standard sedatives reveals some deviation from the funnel shape (Additional file 1: Figure S1), indicating the presence of publication bias (Egger’s test: p = 0.015). This was, however, not the case for the placebo comparison (Egger’s test: p = 0.62; Additional file 1: Figure S2).
Results from the trim-and-fill method (dexmedetomidine vs. standard sedatives) were less clear about the favorable effect compared to the overall result (RR 0.77; 95% CI 0.57–1.05; Table 3). Further subgroup and sensitivity results are presented in Table 3. Considerable heterogeneity was found in the subgroup analysis.

Adverse events

Explorative analysis of adverse events raising no claim for completeness is presented in Additional file 1: Table S5. When compared to placebo, dexmedetomidine sedation showed a RR of 0.92 for mortality (95% CI 0.51–1.64; I2 = 0%) [33, 35, 45, 48, 56] and a RR of 0.80 for circulatory insufficiency (95% CI 0.32–2.00; Additional file 1: Table S5) [48]. The risks of bradycardia [3335, 45, 48, 56] and hypotension [3335, 45, 48, 56] were significantly higher in the dexmedetomidine group, as opposed to tachycardia [35, 48] and hypertension [33, 48].
When compared to standard sedatives, only risks of bradycardia (RR 2.05; 95% CI 1.31–3.22; I2 = 36%) [32, 37, 38, 40, 44, 49, 50, 52] and hypotension (RR 1.26; 95% CI 1.04–1.54; I2 = 9%) [37, 38, 40, 42, 47, 50, 52] were significantly higher in the dexmedetomidine group.
When compared to opioids, dexmedetomidine showed a RR of 0.48 for mortality (95% CI 0.09–2.60) [41] and a higher risk of bradycardia with a RR of 2.03 (95% CI 1.07–3.83; I2 = 22%) [36, 41].

Clinical outcomes

Explorative analysis of clinical outcomes raising no claim for completeness is presented in Additional file 1: Table S6. Two outcome measures (i.e., ICU length of stay and time to extubation or duration of mechanical ventilation) were estimated for dexmedetomidine compared to placebo, standard sedatives, and opioids with overall results in favor of dexmedetomidine (Additional file 1: Table S6).

Treatment comparison

Three RCTs were identified, which reported different clinical outcomes (Additional file 1: Table S7). Control drugs were placebo [57], midazolam [58], and haloperidol [59], each in one RCT. Comparing dexmedetomidine to placebo, Reade et al. [59] found a significantly accelerated resolution of delirium in patients receiving dexmedetomidine (median, 23.3 h; n = 39 vs. 40.0 h; n = 32; median difference between groups, 16.0 h [95% CI 3.0–28.0 h]; P = 0.01). The risk of bias according to our assessment was low in this study. Using midazolam as control medication, Yapici et al. (2011) who assessed delirium based on CAM-ICU at 36 h and 60 h postoperatively had an unclear risk of bias in all five domains. While after 36 h all patient still had a delirium (i.e., 37 dexmedetomidine treated and 34 midazolam treated patients), they stated significantly lower number of patients with delirium in the intervention group 60 h postoperatively (1/37 patients; 2.7% vs. 7/34 patients; 21%; P < 0.05). Comparing dexmedetomidine to haloperidol, Reade et al. [59] measured proportion of time with satisfactory ICDSC below 4 and desirable ICDSC below 1 as clinical outcome. Patients who received dexmedetomidine “tended to spend a greater proportion of time with satisfactory scores” (median [IQR], 95.5% [51–100%] vs. 31.5% [17–97%]; P = 0.122 and 61% [0–100%] vs. 0% [0–0%]; P = 0.134, respectively) [59]. The RCT was a non-blinded pilot study (including 10 patients in each treatment arm) which had to goal to assess the feasibility of the trial design and the safety of the two treatments.

Discussion

This meta-analysis suggests dexmedetomidine to be an efficient and reliable sedative agent to reduce the incidence of delirium in critically ill patients when compared to placebo, standard sedatives, and opioids. Our results indicate that the overall incidence of delirium in the dexmedetomidine group was significantly lower compared to placebo, standard sedatives, and opioids. The 2009 study by Maldonado et al. [39] proposed two theories to explain the decreased rates of delirium associated with sedation of dexmedetomidine: The first theory is based on the intrinsic delirium-sparing property determined by multiple characteristics of dexmedetomidine. Since GABA, the primary inhibitory neurotransmitter in the central nervous system seems to play a key role in the pathogenesis of delirium, it is plausible that GABAergic agents such as benzodiazepines and propofol are strongly involved in the development and prolongation of delirium [60, 61]. The second theory is that dexmedetomidine-induced sedation provides a more natural sleep-like sedation pattern, which might reduce the risk of developing delirium [19, 62]. Furthermore, dexmedetomidine has very little effect on the cholinergic system, which is strongly linked to cognitive functions and the development of delirium [63]. Finally, when used as an alternative to sedation with GABAergic agents and/or opioids, dexmedetomidine may decrease delirium incidence.
When analyzing cardiac and non-cardiac surgery patients separately, the risk ratios for placebo and standard sedatives seem to be similar to the overall results. As the number of included data sets becomes small in all sensitivity-and subgroup analyses, the results have to be interpreted with caution.
Reduction of overall delirium risk by dexmedetomidine compared to standard sedatives confirms the findings from a meta-analysis conducted and published by Xia et al. [22]. Moreover, a 2014 meta-analysis by Pasin et al. [20] reported that dexmedetomidine significantly decreased delirium incidence when compared to control patients and in a subgroup analysis comparing dexmedetomidine to midazolam. A subgroup of RCTs assessing only cardiac surgery patients showed also results favoring dexmedetomidine. This finding supports Herr et al.’s [64] statement, suggesting a potentially positive effect of dexmedetomidine on the incidence of ICU delirium when compared to standard sedatives. The only article [38] with a low risk of bias included two data sets which have similar results when compared to the 12 included data sets.
When compared to opioids, the overall risk of delirium seems to be significantly lower favoring dexmedetomidine. The result has to be interpreted with caution because the opioid-controlled group in this meta-analysis consists of only two RCTs with unclear risk of bias [36, 41].
Of note, it can be interpreted from the subgroup and sensitivity analysis that all data sets point into the same direction, but there are not many studies of high level of evidence estimated by the five dimensions of the risk of bias assessment.
Our additional explorative meta-analyses assessing clinical outcomes confirmed that the use of dexmedetomidine significantly increased the risks of bradycardia and hypotension [3238, 4042, 44, 45, 47, 48, 50]. However, we had only limited data to assess the mortality risk or risk of circulatory insufficiency. Large uncertainties exist as indicated by wide 95% CIs. Here a comprehensive meta-analysis for this endpoint is warranted.
For the treatment of delirium, we identified only three RCTs including all different comparators. Therefore, it is difficult to draw any conclusion about the potential of dexmedetomidine in the treatment of delirium. This systematic review demonstrated the essential need for a standardized measurement for the clinical recovery from delirium. Otherwise, it is impossible to meta-analyze subsequent RCTs on this topic.
Several limitations in our study need to be acknowledged. First, since we focused on incidence of delirium as our primary outcome, we excluded all RCTs not containing delirium data. Of these studies, some might probably have evaluated adverse events and clinical outcome. Therefore, our meta-analysis of adverse events and clinical outcome raises no claim to completeness. Second, we only search in PubMed and CENTRAL for articles published in English. Therefore, it is possible that we missed relevant RCTs because they were published in other languages or published in journals which are not indexed in PubMed or CENTRAL. However, a recently published meta-epidemiological study showed that searching only PubMed and CENTRAL leads in the vast majority to the same conclusion (eventually with less certainty) when compared to more comprehensive Cochrane reviews [65]. Third, only seven out of 28 RCTs could be classified as low risk of bias RCTs. Fourth, there were many significant variations across all RCTs, potentially affecting their comparability. These include different ICU populations, severities of illness, and sedation protocols, leading to heterogeneity in drug doses and potentially influencing outcomes, or different durations of drug administration. Fifth, publication bias could not be assessed in one of the three main comparisons due to the limited number of studies [29]. The found effect for the comparison between standard sedatives and dexmedetomidine seemed to be affected by publication bias. Therefore, the results from the trim-and-fill method might be more valid. A further potential limitation, common in all sedation trials, is a masking of differences in sedation if opioids have been administered for clinical reasons [40].
Overall, our systematic review and meta-analysis are the first to focus on the role of dexmedetomidine in the prevention and treatment of the ICU delirium without making any restrictions concerning comparator drugs or types of ICU population. It uses a systematic approach to evaluate the hypothesis that dexmedetomidine is more effective than common therapeutic strategies for the treatment and prevention of delirium.
The results of our meta-analysis suggest that dexmedetomidine may be more efficient in reducing ICU delirium incidence than placebo standard sedatives and opioids. The evidence on treatment of ICU delirium with dexmedetomidine is limited, as only three trials with different comparators each were suitable for our investigation, and needs to be further investigated [66]. Additionally, it is necessary to standardize clinical outcomes in general, and especially in the treatment section to facilitate meta-analyses, thereby ensuring robust evidence.

Authors’ contributions

JF, AH, MS contributed to study concept and design; JF, AH, BS, MS took part in acquisition of data including risk of bias assessment; JF, AH, BS, MS involved in analysis and interpretation of data; JF, AH, MS took part in drafting of the manuscript; all declared authors participated in critical revision of the manuscript for important intellectual content; administrative, technical, or material support is not applicable; MS involved in study supervision. All authors read and approved the final manuscript.

Acknowledgements

The authors thank Allison Dwileski, for providing us with the literature and for proofreading the final manuscript.

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

No review protocol for this systematic review and meta-analysis was registered online. Prof. Siegemund had full access to all data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.
Not applicable.
Not applicable.

Funding

Not applicable.

Authors’ information

Julian Flükiger works as a MD candidate at the University Hospital Basel. Alexa Hollinger works as a postdoctoral research fellow at the Saint Louis Lariboisière University Hospitals. Benjamin Speich works as a postdoctoral research at the Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, UK. Vera Meier, Janna Tontsch, and Tatjana Zehnder are medical students at the University of Basel and are all part of the research team of the Department for Anesthesia, Surgical Intensive Care, Prehospital Emergency Medicine and Pain Therapy of the University Hospital Basel. Martin Siegemund works as deputy head of the surgical intensive care unit at the University Hospital Basel.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.
Literatur
1.
Zurück zum Zitat Barr J, Fraser GL, Puntillo K, Ely EW, Gelinas C, Dasta JF, et al. Clinical practice guidelines for the management of pain, agitation, and delirium in adult patients in the intensive care unit. Crit Care Med. 2013;41(1):263–306.CrossRef Barr J, Fraser GL, Puntillo K, Ely EW, Gelinas C, Dasta JF, et al. Clinical practice guidelines for the management of pain, agitation, and delirium in adult patients in the intensive care unit. Crit Care Med. 2013;41(1):263–306.CrossRef
2.
Zurück zum Zitat Pandharipande P, Cotton BA, Shintani A, Thompson J, Pun BT, Morris JA, et al. Prevalence and risk factors for development of delirium in surgical and trauma intensive care unit patients. J Trauma. 2008;65:34–41.CrossRef Pandharipande P, Cotton BA, Shintani A, Thompson J, Pun BT, Morris JA, et al. Prevalence and risk factors for development of delirium in surgical and trauma intensive care unit patients. J Trauma. 2008;65:34–41.CrossRef
3.
Zurück zum Zitat McPherson JA, Wagner CE, Boehm LM, Hall JD, Johnson DC, Miller LR, et al. Delirium in the cardiovascular ICU: exploring modifiable risk factors. Crit Care Med. 2013;41:405–13.CrossRef McPherson JA, Wagner CE, Boehm LM, Hall JD, Johnson DC, Miller LR, et al. Delirium in the cardiovascular ICU: exploring modifiable risk factors. Crit Care Med. 2013;41:405–13.CrossRef
4.
Zurück zum Zitat Ely EW, Shintani A, Truman B, Speroff T, Gordon S, Harrell F, et al. Delirium as a predictor of mortality in mechanically ventilated patients in the intensive care unit. JAMA J Am Med Assoc. 2004;291:1753–62.CrossRef Ely EW, Shintani A, Truman B, Speroff T, Gordon S, Harrell F, et al. Delirium as a predictor of mortality in mechanically ventilated patients in the intensive care unit. JAMA J Am Med Assoc. 2004;291:1753–62.CrossRef
5.
Zurück zum Zitat Lonergan E, Luxenberg J, Sastre AA, Wyller TB. Benzodiazepines for delirium. Cochrane Database Syst Rev. 2009;1:CD006379. Lonergan E, Luxenberg J, Sastre AA, Wyller TB. Benzodiazepines for delirium. Cochrane Database Syst Rev. 2009;1:CD006379.
7.
Zurück zum Zitat Hollinger A, Siegemund M, Goettel N, Steiner LA. Postoperative delirium in cardiac surgery: an unavoidable menace? J Cardiothorac Vasc Anesth. 2015;29(6):1677–87.CrossRef Hollinger A, Siegemund M, Goettel N, Steiner LA. Postoperative delirium in cardiac surgery: an unavoidable menace? J Cardiothorac Vasc Anesth. 2015;29(6):1677–87.CrossRef
8.
Zurück zum Zitat Inouye SK, Viscoli CM, Horwitz RI, Hurst LD, Tinetti ME. A predictive model for delirium in hospitalized elderly medical patients based on admission characteristics. Ann Intern Med. 1993;119:474–81.CrossRef Inouye SK, Viscoli CM, Horwitz RI, Hurst LD, Tinetti ME. A predictive model for delirium in hospitalized elderly medical patients based on admission characteristics. Ann Intern Med. 1993;119:474–81.CrossRef
9.
Zurück zum Zitat Inouye SK, Charpentier PA. Precipitating factors for delirium in hospitalized elderly persons. JAMA J Am Med Assoc. 1996;275:852–7.CrossRef Inouye SK, Charpentier PA. Precipitating factors for delirium in hospitalized elderly persons. JAMA J Am Med Assoc. 1996;275:852–7.CrossRef
10.
Zurück zum Zitat Tan JA, Ho KM. Use of dexmedetomidine as a sedative and analgesic agent in critically ill adult patients: a meta-analysis. Intensive Care Med. 2010;36:926–39.CrossRef Tan JA, Ho KM. Use of dexmedetomidine as a sedative and analgesic agent in critically ill adult patients: a meta-analysis. Intensive Care Med. 2010;36:926–39.CrossRef
11.
Zurück zum Zitat Riker RR, Fraser GL. Altering intensive care sedation paradigms to improve patient outcomes. Anesthesiol Clin. 2011;29:663–74.CrossRef Riker RR, Fraser GL. Altering intensive care sedation paradigms to improve patient outcomes. Anesthesiol Clin. 2011;29:663–74.CrossRef
12.
Zurück zum Zitat Mantz J, Josserand J, Hamada S. Dexmedetomidine: new insights. Eur J Anaesthesiol. 2011;28:3–6.CrossRef Mantz J, Josserand J, Hamada S. Dexmedetomidine: new insights. Eur J Anaesthesiol. 2011;28:3–6.CrossRef
13.
Zurück zum Zitat Girard TD, Pandharipande PP, Ely EW. Delirium in the intensive care unit. Crit Care. 2008;12:S3.CrossRef Girard TD, Pandharipande PP, Ely EW. Delirium in the intensive care unit. Crit Care. 2008;12:S3.CrossRef
14.
Zurück zum Zitat Mo Y, Zimmermann AE. Role of dexmedetomidine for the prevention and treatment of delirium in intensive care unit patients. Ann Pharmacother. 2013;47:869–76.CrossRef Mo Y, Zimmermann AE. Role of dexmedetomidine for the prevention and treatment of delirium in intensive care unit patients. Ann Pharmacother. 2013;47:869–76.CrossRef
15.
Zurück zum Zitat Barr J, Fraser GL, Puntillo K, Ely EW, Gélinas C, Dasta JF, et al. Clinical practice guidelines for the management of pain, agitation, and delirium in adult patients in the intensive care unit. Crit Care Med. 2013;41:263–306.CrossRef Barr J, Fraser GL, Puntillo K, Ely EW, Gélinas C, Dasta JF, et al. Clinical practice guidelines for the management of pain, agitation, and delirium in adult patients in the intensive care unit. Crit Care Med. 2013;41:263–306.CrossRef
16.
Zurück zum Zitat Chen K, Lu Z, Xin Y, Cai Y, Chen Y, Pan S. Alpha-2 agonists for long-term sedation during mechanical ventilation in critically ill patients. Cochrane Database Syst Rev. 2015;1:CD010269.PubMed Chen K, Lu Z, Xin Y, Cai Y, Chen Y, Pan S. Alpha-2 agonists for long-term sedation during mechanical ventilation in critically ill patients. Cochrane Database Syst Rev. 2015;1:CD010269.PubMed
17.
Zurück zum Zitat Paris A, Tonner PH. Dexmedetomidine in anaesthesia. Curr Opin Anaesthesiol. 2005;18:412–8.CrossRef Paris A, Tonner PH. Dexmedetomidine in anaesthesia. Curr Opin Anaesthesiol. 2005;18:412–8.CrossRef
18.
Zurück zum Zitat Huupponen E, Maksimow A, Lapinlampi P, Sarkela M, Saastamoinen A, Snapir A, et al. Electroencephalogram spindle activity during dexmedetomidine sedation and physiological sleep. Acta Anaesthesiol Scand. 2008;52(2):289–94.CrossRef Huupponen E, Maksimow A, Lapinlampi P, Sarkela M, Saastamoinen A, Snapir A, et al. Electroencephalogram spindle activity during dexmedetomidine sedation and physiological sleep. Acta Anaesthesiol Scand. 2008;52(2):289–94.CrossRef
19.
Zurück zum Zitat Venn RM, Bradshaw CJ, Spencer R, Brealey D, Caudwell E, Naughton C, et al. Preliminary UK experience of dexmedetomidine, a novel agent for postoperative sedation in the intensive care unit. Anaesthesia. 1999;54:1136–42.CrossRef Venn RM, Bradshaw CJ, Spencer R, Brealey D, Caudwell E, Naughton C, et al. Preliminary UK experience of dexmedetomidine, a novel agent for postoperative sedation in the intensive care unit. Anaesthesia. 1999;54:1136–42.CrossRef
20.
Zurück zum Zitat Pasin L, Landoni G, Nardelli P, Belletti A, Di Prima AL, Taddeo D, et al. Dexmedetomidine reduces the risk of delirium, agitation and confusion in critically Ill patients: a meta-analysis of randomized controlled trials. J Cardiothorac Vasc Anesth. 2014;28(6):1459–66.CrossRef Pasin L, Landoni G, Nardelli P, Belletti A, Di Prima AL, Taddeo D, et al. Dexmedetomidine reduces the risk of delirium, agitation and confusion in critically Ill patients: a meta-analysis of randomized controlled trials. J Cardiothorac Vasc Anesth. 2014;28(6):1459–66.CrossRef
21.
Zurück zum Zitat Szumita PM, Baroletti SA, Anger KE, Wechsler ME. Sedation and analgesia in the intensive care unit: evaluating the role of dexmedetomidine. Am J Health Syst Pharm AJHP. 2007;64:37–44.CrossRef Szumita PM, Baroletti SA, Anger KE, Wechsler ME. Sedation and analgesia in the intensive care unit: evaluating the role of dexmedetomidine. Am J Health Syst Pharm AJHP. 2007;64:37–44.CrossRef
22.
Zurück zum Zitat Xia Z-Q, Chen S-Q, Yao X, Xie C-B, Wen S-H, Liu K-X. Clinical benefits of dexmedetomidine versus propofol in adult intensive care unit patients: a meta-analysis of randomized clinical trials. J Surg Res. 2013;185:833–43.CrossRef Xia Z-Q, Chen S-Q, Yao X, Xie C-B, Wen S-H, Liu K-X. Clinical benefits of dexmedetomidine versus propofol in adult intensive care unit patients: a meta-analysis of randomized clinical trials. J Surg Res. 2013;185:833–43.CrossRef
23.
Zurück zum Zitat Duan X, Coburn M, Rossaint R, Sanders RD, Waesberghe JV, Kowark A. Efficacy of perioperative dexmedetomidine on postoperative delirium: systematic review and meta-analysis with trial sequential analysis of randomised controlled trials. Br J Anaesth. 2018;121(2):384–97.CrossRef Duan X, Coburn M, Rossaint R, Sanders RD, Waesberghe JV, Kowark A. Efficacy of perioperative dexmedetomidine on postoperative delirium: systematic review and meta-analysis with trial sequential analysis of randomised controlled trials. Br J Anaesth. 2018;121(2):384–97.CrossRef
24.
Zurück zum Zitat Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gøtzsche PC, Ioannidis JPA, et al. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: explanation and elaboration. PLoS Med. 2009;6:e1000100.CrossRef Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gøtzsche PC, Ioannidis JPA, et al. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: explanation and elaboration. PLoS Med. 2009;6:e1000100.CrossRef
25.
Zurück zum Zitat Higgins JPT, Altman DG, Gotzsche PC, Juni P, Moher D, Oxman AD, et al. The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials. BMJ. 2011;343:d5928.CrossRef Higgins JPT, Altman DG, Gotzsche PC, Juni P, Moher D, Oxman AD, et al. The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials. BMJ. 2011;343:d5928.CrossRef
26.
Zurück zum Zitat Duval S, Tweedie R. Trim and fill: a simple funnel-plot-based method of testing and adjusting for publication bias in meta-analysis. Biometrics. 2000;56(2):455–63.CrossRef Duval S, Tweedie R. Trim and fill: a simple funnel-plot-based method of testing and adjusting for publication bias in meta-analysis. Biometrics. 2000;56(2):455–63.CrossRef
27.
Zurück zum Zitat Duval STRA. Nonparametric “trim and fill” method of accounting for publication bias in meta-analysis. J Am Stat Assoc. 2000;95(449):89–98. Duval STRA. Nonparametric “trim and fill” method of accounting for publication bias in meta-analysis. J Am Stat Assoc. 2000;95(449):89–98.
28.
Zurück zum Zitat Higgins JPT, Thompson SG. Quantifying heterogeneity in a meta-analysis. Stat Med. 2002;21:1539–58.CrossRef Higgins JPT, Thompson SG. Quantifying heterogeneity in a meta-analysis. Stat Med. 2002;21:1539–58.CrossRef
29.
Zurück zum Zitat Higgins J, Green S. Cochrane handbook for systematic reviews of interventions. London: The Cochrane Collaboration; 2011. Higgins J, Green S. Cochrane handbook for systematic reviews of interventions. London: The Cochrane Collaboration; 2011.
30.
Zurück zum Zitat Wan X, Wang W, Liu J, Tong T. Estimating the sample mean and standard deviation from the sample size, median, range and/or interquartile range. BMC Med Res Methodol. 2014;14:135.CrossRef Wan X, Wang W, Liu J, Tong T. Estimating the sample mean and standard deviation from the sample size, median, range and/or interquartile range. BMC Med Res Methodol. 2014;14:135.CrossRef
31.
Zurück zum Zitat Luo D, Wan X, Liu J, Tong T. Optimally estimating the sample mean from the sample size, median, mid-range and/or mid-quartile range. Stat Methods Med Res. 2016;27:1785.CrossRef Luo D, Wan X, Liu J, Tong T. Optimally estimating the sample mean from the sample size, median, mid-range and/or mid-quartile range. Stat Methods Med Res. 2016;27:1785.CrossRef
32.
Zurück zum Zitat Pandharipande PP, Pun BT, Herr DL, Maze M, Girard TD, Miller RR, et al. Effect of sedation with dexmedetomidine vs lorazepam on acute brain dysfunction in mechanically ventilated patients: the MENDS randomized controlled trial. JAMA. 2007;298:2644–53.CrossRef Pandharipande PP, Pun BT, Herr DL, Maze M, Girard TD, Miller RR, et al. Effect of sedation with dexmedetomidine vs lorazepam on acute brain dysfunction in mechanically ventilated patients: the MENDS randomized controlled trial. JAMA. 2007;298:2644–53.CrossRef
33.
Zurück zum Zitat Deiner S, Luo X, Lin H-M, Sessler DI, Saager L, Sieber FE, et al. Intraoperative infusion of dexmedetomidine for prevention of postoperative delirium and cognitive dysfunction in elderly patients undergoing major elective noncardiac surgery. JAMA Surg. 2017;152:e171505.CrossRef Deiner S, Luo X, Lin H-M, Sessler DI, Saager L, Sieber FE, et al. Intraoperative infusion of dexmedetomidine for prevention of postoperative delirium and cognitive dysfunction in elderly patients undergoing major elective noncardiac surgery. JAMA Surg. 2017;152:e171505.CrossRef
34.
Zurück zum Zitat Yang X, Li Z, Gao C, Liu R. Effect of dexmedetomidine on preventing agitation and delirium after microvascular free flap surgery: a randomized, double-blind, control study. J Oral Maxillofac Surg. 2015;73:1065–72.CrossRef Yang X, Li Z, Gao C, Liu R. Effect of dexmedetomidine on preventing agitation and delirium after microvascular free flap surgery: a randomized, double-blind, control study. J Oral Maxillofac Surg. 2015;73:1065–72.CrossRef
35.
Zurück zum Zitat Devlin JW, Al-Qadheeb NS, Chi A, Roberts RJ, Qawi I, Garpestad E, et al. Efficacy and safety of early dexmedetomidine during noninvasive ventilation for patients with acute respiratory failure: a randomized, double-blind, placebo-controlled pilot study. Chest. 2014;145:1204–12.CrossRef Devlin JW, Al-Qadheeb NS, Chi A, Roberts RJ, Qawi I, Garpestad E, et al. Efficacy and safety of early dexmedetomidine during noninvasive ventilation for patients with acute respiratory failure: a randomized, double-blind, placebo-controlled pilot study. Chest. 2014;145:1204–12.CrossRef
36.
Zurück zum Zitat Park JB, Bang SH, Chee HK, Kim JS, Lee SA, Shin JK. Efficacy and safety of dexmedetomidine for postoperative delirium in adult cardiac surgery on cardiopulmonary bypass. Korean J Thorac Cardiovasc Surg. 2014;47(3):249–54.CrossRef Park JB, Bang SH, Chee HK, Kim JS, Lee SA, Shin JK. Efficacy and safety of dexmedetomidine for postoperative delirium in adult cardiac surgery on cardiopulmonary bypass. Korean J Thorac Cardiovasc Surg. 2014;47(3):249–54.CrossRef
37.
Zurück zum Zitat Huang Z, Chen Y-S, Yang Z-L, Liu J-Y. Dexmedetomidine versus midazolam for the sedation of patients with non-invasive ventilation failure. Intern Med. 2012;51:2299–305.CrossRef Huang Z, Chen Y-S, Yang Z-L, Liu J-Y. Dexmedetomidine versus midazolam for the sedation of patients with non-invasive ventilation failure. Intern Med. 2012;51:2299–305.CrossRef
38.
Zurück zum Zitat Jakob SM, Ruokonen E, Grounds RM, Sarapohja T, Garratt C, Pocock SJ, et al. Dexmedetomidine vs midazolam or propofol for sedation during prolonged mechanical ventilation. JAMA. 2012;307:1151–60.CrossRef Jakob SM, Ruokonen E, Grounds RM, Sarapohja T, Garratt C, Pocock SJ, et al. Dexmedetomidine vs midazolam or propofol for sedation during prolonged mechanical ventilation. JAMA. 2012;307:1151–60.CrossRef
39.
Zurück zum Zitat Maldonado JR, Wysong A, van der Starre PJ, Block T, Miller C, Reitz BA. Dexmedetomidine and the reduction of postoperative delirium after cardiac surgery. Psychosomatics. 2009;50(3):206–17.CrossRef Maldonado JR, Wysong A, van der Starre PJ, Block T, Miller C, Reitz BA. Dexmedetomidine and the reduction of postoperative delirium after cardiac surgery. Psychosomatics. 2009;50(3):206–17.CrossRef
40.
Zurück zum Zitat Ruokonen E, Parviainen I, Jakob SM, Nunes S, Kaukonen M, Shepherd ST, et al. Dexmedetomidine versus propofol/midazolam for long-term sedation during mechanical ventilation. Intensive Care Med. 2009;35:282–90.CrossRef Ruokonen E, Parviainen I, Jakob SM, Nunes S, Kaukonen M, Shepherd ST, et al. Dexmedetomidine versus propofol/midazolam for long-term sedation during mechanical ventilation. Intensive Care Med. 2009;35:282–90.CrossRef
41.
Zurück zum Zitat Shehabi Y, Grant P, Wolfenden H, Hammond N, Bass F, Campbell M, et al. Prevalence of delirium with dexmedetomidine compared with morphine based therapy after cardiac surgery: a randomized controlled trial (DEXmedetomidine COmpared to Morphine-DEXCOM Study). Anesthesiology. 2009;111(5):1075–84.CrossRef Shehabi Y, Grant P, Wolfenden H, Hammond N, Bass F, Campbell M, et al. Prevalence of delirium with dexmedetomidine compared with morphine based therapy after cardiac surgery: a randomized controlled trial (DEXmedetomidine COmpared to Morphine-DEXCOM Study). Anesthesiology. 2009;111(5):1075–84.CrossRef
42.
Zurück zum Zitat Corbett SM, Rebuck JA, Greene CM, Callas PW, Neale BW, Healey MA, et al. Dexmedetomidine does not improve patient satisfaction when compared with propofol during mechanical ventilation. Crit Care Med. 2005;33:940–5.CrossRef Corbett SM, Rebuck JA, Greene CM, Callas PW, Neale BW, Healey MA, et al. Dexmedetomidine does not improve patient satisfaction when compared with propofol during mechanical ventilation. Crit Care Med. 2005;33:940–5.CrossRef
43.
Zurück zum Zitat Li Y, Yu Z-X, Ji M-S, Yan J, Cai Y, Liu J, et al. A pilot study of the use of dexmedetomidine for the control of delirium by reducing the serum concentrations of brain-derived neurotrophic factor, neuron-specific enolase, and S100B in polytrauma patients. J Intensive Care Med. 2017;1:885066617710643. Li Y, Yu Z-X, Ji M-S, Yan J, Cai Y, Liu J, et al. A pilot study of the use of dexmedetomidine for the control of delirium by reducing the serum concentrations of brain-derived neurotrophic factor, neuron-specific enolase, and S100B in polytrauma patients. J Intensive Care Med. 2017;1:885066617710643.
44.
Zurück zum Zitat Kawazoe Y, Miyamoto K, Morimoto T, Yamamoto T, Fuke A, Hashimoto A, et al. Effect of dexmedetomidine on mortality and ventilator-free days in patients requiring mechanical ventilation with sepsis. JAMA. 2017;317:1321–8.CrossRef Kawazoe Y, Miyamoto K, Morimoto T, Yamamoto T, Fuke A, Hashimoto A, et al. Effect of dexmedetomidine on mortality and ventilator-free days in patients requiring mechanical ventilation with sepsis. JAMA. 2017;317:1321–8.CrossRef
45.
Zurück zum Zitat Li X, Yang J, Nie X-L, Zhang Y, Li X-Y, Li L-H, et al. Impact of dexmedetomidine on the incidence of delirium in elderly patients after cardiac surgery: a randomized controlled trial. PLoS ONE. 2017;12:e0170757.CrossRef Li X, Yang J, Nie X-L, Zhang Y, Li X-Y, Li L-H, et al. Impact of dexmedetomidine on the incidence of delirium in elderly patients after cardiac surgery: a randomized controlled trial. PLoS ONE. 2017;12:e0170757.CrossRef
46.
Zurück zum Zitat Djaiani G, Silverton N, Fedorko L, Carroll J, Styra R, Rao V, et al. Dexmedetomidine versus propofol sedation reduces delirium after cardiac surgery. Anesthesiology. 2016;124:362–8.CrossRef Djaiani G, Silverton N, Fedorko L, Carroll J, Styra R, Rao V, et al. Dexmedetomidine versus propofol sedation reduces delirium after cardiac surgery. Anesthesiology. 2016;124:362–8.CrossRef
47.
Zurück zum Zitat Liu X, Zhang K, Wang W, Xie G, Cheng B, Wang Y, et al. Dexmedetomidine versus propofol sedation improves sublingual microcirculation after cardiac surgery: a randomized controlled trial. J Cardiothorac Vasc Anesth. 2016;30:1509–15.CrossRef Liu X, Zhang K, Wang W, Xie G, Cheng B, Wang Y, et al. Dexmedetomidine versus propofol sedation improves sublingual microcirculation after cardiac surgery: a randomized controlled trial. J Cardiothorac Vasc Anesth. 2016;30:1509–15.CrossRef
48.
Zurück zum Zitat Su X, Meng Z-T, Wu X-H, Cui F, Li H-L, Wang D-X, et al. Dexmedetomidine for prevention of delirium in elderly patients after non-cardiac surgery: a randomised, double-blind, placebo-controlled trial. Lancet. 2016;388:1893–902.CrossRef Su X, Meng Z-T, Wu X-H, Cui F, Li H-L, Wang D-X, et al. Dexmedetomidine for prevention of delirium in elderly patients after non-cardiac surgery: a randomised, double-blind, placebo-controlled trial. Lancet. 2016;388:1893–902.CrossRef
49.
Zurück zum Zitat Liu Y, Ma L, Gao M, Guo W, Ma Y. Dexmedetomidine reduces postoperative delirium after joint replacement in elderly patients with mild cognitive impairment. Aging Clin Exp Res. 2016;28:729–36.CrossRef Liu Y, Ma L, Gao M, Guo W, Ma Y. Dexmedetomidine reduces postoperative delirium after joint replacement in elderly patients with mild cognitive impairment. Aging Clin Exp Res. 2016;28:729–36.CrossRef
50.
Zurück zum Zitat MacLaren R, Preslaski CR, Mueller SW, Kiser TH, Fish DN, Lavelle JC, et al. A randomized, double-blind pilot study of dexmedetomidine versus midazolam for intensive care unit sedation. J Intensive Care Med. 2015;30:167–75.CrossRef MacLaren R, Preslaski CR, Mueller SW, Kiser TH, Fish DN, Lavelle JC, et al. A randomized, double-blind pilot study of dexmedetomidine versus midazolam for intensive care unit sedation. J Intensive Care Med. 2015;30:167–75.CrossRef
51.
Zurück zum Zitat Priye S, Jagannath S, Singh D, Shivaprakash S, Reddy D. Dexmedetomidine as an adjunct in postoperative analgesia following cardiac surgery: a randomized, double-blind study. Saudi J Anaesth. 2015;9:353–8.CrossRef Priye S, Jagannath S, Singh D, Shivaprakash S, Reddy D. Dexmedetomidine as an adjunct in postoperative analgesia following cardiac surgery: a randomized, double-blind study. Saudi J Anaesth. 2015;9:353–8.CrossRef
52.
Zurück zum Zitat Chang YF, Chao A, Shih PY, Hsu YC, Lee CT, Tien YW, et al. Comparison of dexmedetomidine versus propofol on hemodynamics in surgical critically ill patients. J Surg Res. 2018;228:194–200.CrossRef Chang YF, Chao A, Shih PY, Hsu YC, Lee CT, Tien YW, et al. Comparison of dexmedetomidine versus propofol on hemodynamics in surgical critically ill patients. J Surg Res. 2018;228:194–200.CrossRef
53.
Zurück zum Zitat Sheikh TA, Dar BA, Akhter N, Ahmad N. A comparative study evaluating effects of intravenous sedation by dexmedetomidine and propofol on patient hemodynamics and postoperative outcomes in cardiac surgery. Anesth Essays Res. 2018;12(2):555–60.CrossRef Sheikh TA, Dar BA, Akhter N, Ahmad N. A comparative study evaluating effects of intravenous sedation by dexmedetomidine and propofol on patient hemodynamics and postoperative outcomes in cardiac surgery. Anesth Essays Res. 2018;12(2):555–60.CrossRef
54.
Zurück zum Zitat Mei B, Meng G, Xu G, Cheng X, Chen S, Zhang Y, et al. Intraoperative sedation with dexmedetomidine is superior to propofol for elderly patients undergoing hip arthroplasty: a prospective randomized controlled study. Clin J Pain. 2018;34(9):811–7.PubMed Mei B, Meng G, Xu G, Cheng X, Chen S, Zhang Y, et al. Intraoperative sedation with dexmedetomidine is superior to propofol for elderly patients undergoing hip arthroplasty: a prospective randomized controlled study. Clin J Pain. 2018;34(9):811–7.PubMed
55.
Zurück zum Zitat Lee C, Lee CH, Lee G, Lee M, Hwang J. The effect of the timing and dose of dexmedetomidine on postoperative delirium in elderly patients after laparoscopic major non-cardiac surgery: a double blind randomized controlled study. J Clin Anesth. 2018;47:27–32.CrossRef Lee C, Lee CH, Lee G, Lee M, Hwang J. The effect of the timing and dose of dexmedetomidine on postoperative delirium in elderly patients after laparoscopic major non-cardiac surgery: a double blind randomized controlled study. J Clin Anesth. 2018;47:27–32.CrossRef
56.
Zurück zum Zitat Skrobik Y, Duprey MS, Hill NS, Devlin JW. Low-dose nocturnal dexmedetomidine prevents ICU delirium. A randomized, placebo-controlled trial. Am J Respir Crit Care Med. 2018;197(9):1147–56.CrossRef Skrobik Y, Duprey MS, Hill NS, Devlin JW. Low-dose nocturnal dexmedetomidine prevents ICU delirium. A randomized, placebo-controlled trial. Am J Respir Crit Care Med. 2018;197(9):1147–56.CrossRef
57.
Zurück zum Zitat Reade MC, Eastwood GM, Bellomo R, Bailey M, Bersten A, Cheung B, et al. Effect of dexmedetomidine added to standard care on ventilator-free time in patients with agitated delirium. JAMA. 2016;315:1460–8.CrossRef Reade MC, Eastwood GM, Bellomo R, Bailey M, Bersten A, Cheung B, et al. Effect of dexmedetomidine added to standard care on ventilator-free time in patients with agitated delirium. JAMA. 2016;315:1460–8.CrossRef
58.
Zurück zum Zitat Yapici N, Coruh T, Kehlibar T, Yapici F, Tarhan A, Can Y, et al. Dexmedetomidine in cardiac surgery patients who fail extubation and present with a delirium state. Heart Surg Forum. 2011;14:E93–8.CrossRef Yapici N, Coruh T, Kehlibar T, Yapici F, Tarhan A, Can Y, et al. Dexmedetomidine in cardiac surgery patients who fail extubation and present with a delirium state. Heart Surg Forum. 2011;14:E93–8.CrossRef
59.
Zurück zum Zitat Reade MC, O’Sullivan K, Bates S, Goldsmith D, Ainslie WR, Bellomo R. Dexmedetomidine vs. haloperidol in delirious, agitated, intubated patients: a randomised open-label trial. Crit Care. 2009;13(3):R75.CrossRef Reade MC, O’Sullivan K, Bates S, Goldsmith D, Ainslie WR, Bellomo R. Dexmedetomidine vs. haloperidol in delirious, agitated, intubated patients: a randomised open-label trial. Crit Care. 2009;13(3):R75.CrossRef
61.
Zurück zum Zitat Pandharipande P, Shintani A, Peterson J, Pun BT, Wilkinson GR, Dittus RS, et al. Lorazepam is an independent risk factor for transitioning to delirium in intensive care unit patients. Anesthesiology. 2006;104:21–6.CrossRef Pandharipande P, Shintani A, Peterson J, Pun BT, Wilkinson GR, Dittus RS, et al. Lorazepam is an independent risk factor for transitioning to delirium in intensive care unit patients. Anesthesiology. 2006;104:21–6.CrossRef
62.
Zurück zum Zitat Coull JT, Jones MEP, Egan TD, Frith CD, Maze M. Attentional effects of noradrenaline vary with arousal level: selective activation of thalamic pulvinar in humans. NeuroImage. 2004;22:315–22.CrossRef Coull JT, Jones MEP, Egan TD, Frith CD, Maze M. Attentional effects of noradrenaline vary with arousal level: selective activation of thalamic pulvinar in humans. NeuroImage. 2004;22:315–22.CrossRef
63.
Zurück zum Zitat Zaal IJ, Slooter AJC. Delirium in critically ill patients: epidemiology, pathophysiology, diagnosis and management. Drugs. 2012;72:1457–71.CrossRef Zaal IJ, Slooter AJC. Delirium in critically ill patients: epidemiology, pathophysiology, diagnosis and management. Drugs. 2012;72:1457–71.CrossRef
64.
Zurück zum Zitat Herr DL, Sum-Ping STJ, England M. ICU sedation after coronary artery bypass graft surgery: dexmedetomidine-based versus propofol-based sedation regimens. J Cardiothorac Vasc Anesth. 2003;17:576–84.CrossRef Herr DL, Sum-Ping STJ, England M. ICU sedation after coronary artery bypass graft surgery: dexmedetomidine-based versus propofol-based sedation regimens. J Cardiothorac Vasc Anesth. 2003;17:576–84.CrossRef
65.
Zurück zum Zitat Nussbaumer-Streit B, Klerings I, Wagner G, Heise TL, Dobrescu AI, Armijo-Olivo S, et al. Abbreviated literature searches were viable alternatives to comprehensive searches: a meta-epidemiological study. J Clin Epidemiol. 2018;102:1.CrossRef Nussbaumer-Streit B, Klerings I, Wagner G, Heise TL, Dobrescu AI, Armijo-Olivo S, et al. Abbreviated literature searches were viable alternatives to comprehensive searches: a meta-epidemiological study. J Clin Epidemiol. 2018;102:1.CrossRef
66.
Zurück zum Zitat Hollinger A, Ledergerber K, Von Felten S, Sutter R, Rüegg S, Gantner L, et al. Comparison of propofol and dexmedetomidine infused overnight to treat hyperactive and mixed ICU delirium: a protocol for the Basel ProDex clinical trial. BMJ Open. 2017;7:e015783.CrossRef Hollinger A, Ledergerber K, Von Felten S, Sutter R, Rüegg S, Gantner L, et al. Comparison of propofol and dexmedetomidine infused overnight to treat hyperactive and mixed ICU delirium: a protocol for the Basel ProDex clinical trial. BMJ Open. 2017;7:e015783.CrossRef
Metadaten
Titel
Dexmedetomidine in prevention and treatment of postoperative and intensive care unit delirium: a systematic review and meta-analysis
verfasst von
Julian Flükiger
Alexa Hollinger
Benjamin Speich
Vera Meier
Janna Tontsch
Tatjana Zehnder
Martin Siegemund
Publikationsdatum
01.12.2018
Verlag
Springer International Publishing
Erschienen in
Annals of Intensive Care / Ausgabe 1/2018
Elektronische ISSN: 2110-5820
DOI
https://doi.org/10.1186/s13613-018-0437-z

Weitere Artikel der Ausgabe 1/2018

Annals of Intensive Care 1/2018 Zur Ausgabe

Ein Drittel der jungen Ärztinnen und Ärzte erwägt abzuwandern

07.05.2024 Medizinstudium Nachrichten

Extreme Arbeitsverdichtung und kaum Supervision: Dr. Andrea Martini, Sprecherin des Bündnisses Junge Ärztinnen und Ärzte (BJÄ) über den Frust des ärztlichen Nachwuchses und die Vorteile des Rucksack-Modells.

Häufigste Gründe für Brustschmerzen bei Kindern

06.05.2024 Pädiatrische Diagnostik Nachrichten

Akute Brustschmerzen sind ein Alarmsymptom par exellence, schließlich sind manche Auslöser lebensbedrohlich. Auch Kinder klagen oft über Schmerzen in der Brust. Ein Studienteam ist den Ursachen nachgegangen.

Aquatherapie bei Fibromyalgie wirksamer als Trockenübungen

03.05.2024 Fibromyalgiesyndrom Nachrichten

Bewegungs-, Dehnungs- und Entspannungsübungen im Wasser lindern die Beschwerden von Patientinnen mit Fibromyalgie besser als das Üben auf trockenem Land. Das geht aus einer spanisch-brasilianischen Vergleichsstudie hervor.

Endlich: Zi zeigt, mit welchen PVS Praxen zufrieden sind

IT für Ärzte Nachrichten

Darauf haben viele Praxen gewartet: Das Zi hat eine Liste von Praxisverwaltungssystemen veröffentlicht, die von Nutzern positiv bewertet werden. Eine gute Grundlage für wechselwillige Ärzte und Psychotherapeuten.

Update AINS

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.