Next Article in Journal
Acupuncture for Post-Operative Pain Relief and Functional Improvement in Tibial Fracture: A Systematic Review and Meta-Analysis
Previous Article in Journal
Barriers and Facilitators to Smart Healthcare Adoption Among Chinese Patients with Cardiovascular Disease and Their Caregivers: A Qualitative Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Dexmedetomidine’s Role in Adult ICU After 20 Years of Experience—A Narrative Review

by
Eleni N. Sertaridou
1,
Maria Fountoulaki
2,
Abhishek Jha
3,
Vasilios E. Papaioannou
4 and
Christina Alexopoulou
4,*
1
ICU Department, University Hospital of Alexandroupolis, 68100 Alexandroupolis, Greece
2
Second Department of Anaesthesiology, “Attikon” University Hospital, National and Kapodistrian University of Athens, 12461 Athens, Greece
3
King’s College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
4
ICU Department, Medical School, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece
*
Author to whom correspondence should be addressed.
Healthcare 2025, 13(22), 2882; https://doi.org/10.3390/healthcare13222882
Submission received: 1 October 2025 / Revised: 2 November 2025 / Accepted: 6 November 2025 / Published: 12 November 2025
(This article belongs to the Section Clinical Care)

Highlights

What are the main findings?
  • The unique arousable sedation in combination with mild opioid-spare analgesic effects, has confirmed to effectively minimize duration of intubation and mechanical ventilation, ICU and hospital length of stay the total hospital stay cost.
  • The anxiolytic and sympatholytic action have proved to sufficiently enhance sleep qualit, and has an important role on prevention and treatment of ICU delirium and post-ICU syndrome, especially among elderly patients.
What is the implication of the main finding?
  • Increasingly evident advocate promising neuro-, renal-, and cardio-protective and anti-inflammatory effects of Dex, which are attributed to autophagy and apoptosis inhibition, sympatholytic, and ischemia/reperfusion (I/R) injury protective effect.

Abstract

Background: Dexmedetomidine (Dex) is a well-known a2-adrenoceptor agonist with sedative, anxiolytic, sympatholytic, and analgesic effects that has been used principally as adjuvant sedation in the ICU. The enhanced clinical experience of Dex’s use and its physiological effects encourage its application beyond the initial indications. Aim: The purpose of this review is to summarize the current knowledge of Dex’s recently expanded applications in critically ill intensive care unit (ICU) adult patients. Methods: It is a narrative review that critically examines studies published since 2015 and referring to Dex’s use in ICU patients. Results: Despite the preliminary applications and the weak existing recommendation, the unique arousable sedation, in combination with mild opioid-spare analgesic effects, has been confirmed to effectively improve ICU outcomes. Moreover, the anxiolytic and sympatholytic actions have proved to sufficiently enhance sleep quality and prevent and treat ICU delirium and post-ICU syndrome, especially among elderly patients. Recently, increasing evidence advocates for promising neuro-, renal-, and cardio-protective and anti-inflammatory effects of Dex, which are attributed to autophagy and apoptosis inhibition and sympatholytic and ischemia/reperfusion (I/R) injury-protective effects. Conclusions: Beyond sedation, Dex seems to present promising neuroprotective, anti-inflammatory, and immunomodulating effects.

1. Introduction

Dexmedetomidine (Dex) is a highly selective α2-adrenoceptor agonist with sedative [1], antianxiety [2,3], sympatholytic, and mild analgesic opioid-sparing effects [4,5,6] without severe respiratory suppressive effects [7,8]. Unlike older commonly used sedative drugs, such as propofol and benzodiazepines, dexmedetomidine does not act at the gamma-aminobutyric acid (GABA) receptors. Compared with clonidine, another α2-agonist that has also been used for several decades, Dex has greater selectivity for α2-receptors (α2: α1 ratio of 1620:1 vs. 220:1), being a more potent sedative [9,10,11].
Its unique hypnotic action through the activation of central pre- and postsynaptic α2-receptors in the locus coeruleus induces a state of unconsciousness similar to natural sleep, while patients remain easily arousable and cooperative [7]. In addition, it has been shown to attenuate stress responses, minimizing the hemodynamic response to stressful events [3]. Its distinctive sedative and anxiolytic effects, with minimal influence on respiratory drive, support the consideration of Dex as a viable alternative sedative for critically ill patients in the intensive care unit (ICU).
In 1999, Dex was approved by the US Food and Drug Administration for short-term sedation of intubated and mechanically ventilated adults in the ICU and since 2008 in non-intubated patients during short-term surgical procedures, while since 2011 it has been approved in Europe for light to moderate ICU sedation [7,12]. Although the 2018 clinical practice guidelines for management of Pain, Agitation/Sedation, Delirium, Immobility and Sleep Disruption (PADIS) in adult ICU patients offered a conditional recommendation favoring the use of Dex over benzodiazepines and probably propofol in mechanically ventilated adults [13], in real clinical practice, midazolam is still frequently used, and propofol remains the main ICU sedative agent [14,15]. Moreover, the level of evidence for this recommendation was low since it was based on studies published between 2007 and 2015, and the quality of some of the included articles was questionable.
More recently, the Intensive Care Medicine Rapid Practice Guideline (ICM-RPG) issued a weak recommendation related to the use of Dex [16]. However, according to a recent systematic review and meta-analysis for PADIS treatment, Dex’s role and impact remain controversial due to the low quality of evidence [17]. The guideline task force provided moderate certainty recommendations for pharmacological management of agitation/sedation and sleep with Dex over propofol or benzodiazepines [18]. Moreover, Dex has gained popularity, appearing to be useful in multiple off-label applications in critically ill patients, broadening its clinical implementation.
The purpose of this narrative review is to summarize the current knowledge of dexmedetomidine’s more recently expanded applications in critically ill adult ICU patients, considering the physiological effects and highlighting properties that have not yet been implemented in common clinical practice.

2. Methods

This is a narrative review that critically examines recent literature indexed on PubMed since 2015 using the following key terms: “Dexmedetomidine”, “Intensive Care Unit”, “Critical care”, “Delirium”, “Adults” “Sleep in ICU”, “Sedation”, and “Sympatholytic”. The search was limited to peer-reviewed articles published in the last decade, in English, and involving clinical research or experimental studies, with a notable sample of adult critically ill patients, hospitalized in the ICU, and a few reviews and meta-analyses referring to the mechanism of action, pharmacodynamics, and pharmacokinetics.
After screening the titles for possible relevance, and excluding duplicates, 202 papers were reviewed; 2 papers were retracted and 62 papers were excluded because the full text was not in English, included pediatric population, non-ICU, or palliative care unit population, or Dex was used only for regional or intraoperative anesthesia. The references of the articles were also reviewed for additional potentially relevant papers (Figure 1). The literature review was focused on efficacy of Dex as sedative, anxiolytic, analgetic, sympatholytic, neuroprotective, and anti-inflammatory agent and its impact on delirium, post-ICU care syndrome (PICS), and alcohol withdrawal syndrome (AWS) treatment.

3. Mechanism of Action

Dexmedetomidine is a dextrorotatory isomer of the racemic mixture medetomidine. It is an imidazole with the chemical name 4-[(1R)-1-(2,3-dimethylphenyl) ethyl]-3H-imidazole hydrochloride, with G-protein-coupled α2 receptor agonistic action. The α2 adrenergic receptors are widely distributed in the central nervous system (CNS), peripheral nervous system (PNS), autonomic ganglia, and other organ tissues, such as blood vessels, liver, kidney, pancreas, and platelets [19]. Different subtypes of α2 adrenergic receptors (α2A, α2B, and α2C) have different functions. Namely, α2A is considered to be the major presynaptic inhibitory feedback receptor, controlling the exocytosis of adrenergic neurons. Thus, α2 agonists have sedative, analgesic, and antiepileptic effects. In the brainstem, the locus coeruleus contains many α2-adrenoreceptors and plays a key role in wakefulness and regulation of nociceptive neurotransmission. Its agonistic action in the locus coeruleus inhibits norepinephrine release, resulting in depression of alertness and sympathetic activity that manifests sedation, analgesia, hypotension, and bradycardia [20].
The activated α2B receptors that are located mainly in peripheral vascular smooth muscle lead to transient hypertension [9], while those distributed in the spinal cord modify nitrogen monoxide analgesia by inhibiting noradrenergic action [21]. The α2C receptors, which are allocated in the hippocampus, basal ganglia, olfactory bulb system, and cerebral cortex, modulate complex memory and behavioral functions [8]. Dexmedetomidine is a highly selective activator of the α2A receptor, acting on the nucleolus of the nucleus, having sedative and hypnotic effects, while its action on the spinal cord can produce analgesic effects, and its action on the peripheral and CNS can play a role in inhibiting sympathetic excitation [9].

4. Pharmacokinetics and Administration

Dexmedetomidine in critically ill ICU patients is administered purely intravenously, with an action onset within 15 min and reaching peak concentration after 1 h of continuous infusion [19]. It has a half-life of 2 h and follows first-order linear kinetics. It is a highly protein-bound drug, binding almost 94% to plasma albumin and α1-glycoprotein. It is rapidly distributed since a half-life distribution of about 6 min in healthy volunteers has been reported [7]. The volume of distribution was found to be related to age, body weight [22], fat (free) mass [23], and serum albumin levels [24], being estimated at approximately 1.31–2.46 L/kg (90–194 L) in healthy people, although revealing high variability in ICU patients (109–223 L) [7,25].
It is mainly hepatically metabolized into inactive metabolites by glucuronidation and hydroxylation since less than 1% is excreted unchanged, with metabolites being excreted renally (95%) and intestinally (4%) [7]. Thus, patients with hepatic dysfunction may require lower doses due to its prolonged elimination [19,26,27]. Renal impairment does not affect its pharmacokinetics substantially, although it has been described as longer-lasting sedation in such patients, without significant differences in Dex plasma protein binding, volume of distribution, or clearance elimination [7,26]. In the elderly, sedation seems to be more pronounced, with hemodynamic side effects, such as severe hypotension and bradycardia, appearing more frequently, especially after loading doses of >0.7 μg/kg [7,28].
The recommended loading dose of Dex is 1 μg/kg over 10 min, followed by a continuous infusion of 0.2 to 0.7 μg/kg per hour, titrated to the desired level of sedation. A higher dose (1.5 μg/kg per hour) has been administered without clinically significant adverse effects. Body size [23], age [28], hepatic [27], cardiac and renal impairment, and hypoalbuminemia [24] have been found to pronounce pharmacological actions [7]. When Dex is administered to replace another sedative, a loading dose is generally not required since bolus and rapid dosage adjustment to achieve the desired level of sedation have been associated with more adverse drug events [19].
Dexmedetomidine can be administered for more than 24 h, safely in combination with anesthetics, sedatives, hypnotics, neuromuscular blockade agents, and opioids, in reduced doses. Similarly, co-administration with antihypertensive agents could also increase its hypotensive and bradycardic effects [29,30].

5. Effects of Dexmedetomidine

5.1. Sedation

Dexmedetomidine, in plasma concentrations between 0.2 and 0.3 ng/mL, induces the so-called “arousable or cooperative sedation” since it appears as an easy transition from sleep to wakefulness, keeping the patient calm, cooperative, and communicative when stimulated [31]. This sedative property, which simulates natural sleep, is caused by suppression of noradrenergic neuronal firing of locus coeruleus in the brain stem, leading to loss of wakefulness via activation of an endogenous sleep-promoting pathway. In contrast to opioids, benzodiazepines [32,33,34], or propofol [34,35], it diminishes respiratory depression, maintaining the hypercapnic arousal phenomenon even at higher doses, providing significant benefits in discomfort restraint during weaning procedures [7,31] (Table 1).
However, Lodenius et al. reported that, equivalent to propofol, dexmedetomidine reduces both hypoxic and hypercapnic regulation of breathing in young healthy volunteers at plasma concentrations of around 0.66 ng/mL [51], while it has been noticed that elderly patients are more vulnerable to respiratory depression [7]. Concentrations above 1.9 ng/mL could cause unarousable deep sedation [7], raising the frequency of cardiovascular side effects, minimizing the only Dex-based anesthesia strategy [31]. Although initially the US Food and Drug Administration approved Dex for use up to 24 h only, multiple studies showed a safety profile when continuous Dex sedation is used for up to 30 days in ICU patients [12].
Dexmedetomidine, was reported in two large European randomized controlled trials (RCTs) (PRODEX and MIDEX) to be non-inferior to propofol and midazolam, respectively, in maintaining light to moderate sedation in mechanically ventilated ICU patients [36]. The post hoc economic evaluation analysis of these RCTs demonstrated that sedation with Dex significantly shortened the time of extubation and length of mechanical ventilation, reducing ICU resource needs and costs in comparison with midazolam and propofol [12]. Similarly, Aggarwal et al. demonstrated that Dex was associated with lower costs when compared to propofol or midazolam, used for short-term sedation in ICU, due to minimized ICU length of stay (LOS) [40].
The Japanese open-label multicenter RCT (DESIRE study) examined the efficacy of Dex in 201 septic mechanically ventilated patients. They concluded that, although the use of Dex showed an 8% reduction in 28-day mortality compared with other sedatives, which could be clinically important, it did not demonstrate statistical significance. Moreover, even if the Dex group was reported to have more sufficient sedation, the delirium risk and the duration of mechanical ventilation were not reduced. The authors attributed Dex’s non-superiority in delirium prevention and mortality reduction to the limitations of RASS (Richmond Agitation Sedation Scale) and CAM-ICU (Confusion Assessment Method for ICU Patients) scores’ subjective assessment and the small sample size, respectively [29].
Furthermore, Brandão et al., in a large cohort of cardiac surgery patients, found that the use of Dex was associated with lower risk of 30-day mortality, shorter ICU LOS, and fewer postsurgical neurological lesions [37]. Nevertheless, this retrospective cohort study has some important limitations since the Dex group included younger patients, with lower disease severity, shorter operating time, and more often underwent off-pump surgery.
Zhou et al. revealed a notable reduction in weaning time, faster extubation, and lower delirium risk when midazolam was switched to Dex during a spontaneous breathing trial in mechanically ventilated ICU patients compared with propofol or continuation of midazolam [34]. However, many other smaller RCTs, with noteworthy limitations, compared Dex’s efficacy with midazolam [26,32] or propofol [35,38,43,44] and failed to show a clear benefit of Dex sedation according to different outcomes (Table 1).
The use of dexmedetomidine as the sole or primary sedative agent in critically ill patients was assessed in the SPICE III open-label multi-national RCT, where 4000 critically ill mechanically ventilated adults, from 74 ICUs in eight different countries, were randomized to receive either Dex or the usual care (propofol, midazolam, or other sedatives). Those who received early Dex required supplemental propofol, midazolam, or both more often than the usual care patients to achieve the desired level of sedation, assessed by RASS score, while no outcome or delirium risk benefit and more adverse events were reported [39]. The authors attributed their results to the lack of predetermined protocol for pain management, sedative titration, and delirium prevention.
Subsequently, in the secondary analysis of the SPICE III trial, the authors included only ICU mechanically ventilated patients aged older than 65 years old [41]. In this cluster analysis, the early use of Dex resulted in lower 90-day mortality compared to usual care in elderly patients, while, in younger patients, it appeared likely to increase mortality, especially in non-operative critically ill patients with high severity of illness. Smaller clinical trials verified that perioperative Dex infusion may improve ICU and hospital mortality rates in elderly cardiac surgical patients [28].
Another American multicenter double-blind trial (the MENDS-2 trial) that compared light sedation with Dex or propofol in 432 mechanically ventilated septic patients failed to show any differences in ventilator-free days, 28- and 90-day mortality, delirium, and post-ICU cognition [42]. The lack of pain and delirium prevention strategies was a notable limitation in this study.
Subsequently, an English large multicenter RCT (the A2B trial) revealed a notable reduction in mechanical ventilation duration among ICU patients being sedated with Dex in comparison with clonidine or propofol, although agitation and hemodynamical instability were described more often, and there was no difference in the final outcome [1]. The post hoc economic evaluation of the A2B trial revealed similar total cost and quality of life among the studied groups [45].
A Cochrane meta-analysis of seven studies and 1624 mechanically ventilated ICU patients compared long-term Dex sedation with traditional sedatives, such as midazolam, lorazepam, and propofol, and reported that Dex reduced mechanical ventilation duration by 22% and ICU LOS by 14% [46]. The lack of evidence for a beneficial effect on the risk of delirium and mortality was attributed to the high heterogeneity of the included studies [46]. Moreover, Lewis et al. reported reduced risk of delirium and shorter duration of mechanical ventilation and ICU LOS upon systematically reviewing 77 RCTs including 11,997 mechanically ventilated critically ill patients [49]. Previously, they reported reduced risk of intubation, delirium, and ICU LOS after reviewing 12 RCTs including 738 ICU patients treated with non-invasive ventilation (NIV) [47]. Similar results were presented by Wen et al. in their systematic review and meta-analysis, where they included 16 RCTs and 2035 ICU mechanically ventilated patients. They verified Dex’s benefit in regard to the risk of delirium, length of mechanical ventilation and ICU stay, and the total cost compared to midazolam [50] (Table 1).

5.2. Analgesic Effects

Analgesic effects of α2-agonists are thought to be mediated by α2-receptor binding in central and spinal cord α2-receptors. Pain transmission is suppressed by hyperpolarization of interneurons and reduction in the release of pronociceptive transmitters, such as substance P and glutamate [9]. Early studies, investigating the analgesic properties of dexmedetomidine, found that mild to deep sedation lacks analgesic efficacy compared to opioids. Therefore, it has been proposed to be combined with an opioid agent [7]. Moreover, according to Peng et al., this combination is more effective in minimizing opioid side effects [52]. However, recently accumulating evidence has demonstrated that dexmedetomidine may have an opioid-sparing analgesic effect on ischemic, acute postoperative [5,22,52], and refractory cancer pain [8] but limited efficacy in posttraumatic pain [6].
The mechanism of dexmedetomidine analgesia has not been fully clarified. Probably, analgesic effects of Dex may partly be owing to an altered perception and reduced anxiety, although an opioid-sparing effect is described [7]. The peripheral analgesic effect might be due to inhibiting the transmission of pain signals through Aδ and C fibers. Moreover, Dex depolarizes the blue plaque and the descending noradrenergic pathway of the spinal cord to the presynaptic membrane, inhibiting the release of substance P and other nociceptive peptides in the presynaptic membrane, thereby inhibiting the spinal cord via the transmission of angular noxious stimuli, which in turn terminates the signaling of pain. Furthermore, Dex seems to have a local analgesic effect through modulation of hyperalgesia by stimulating the α2 receptor [8].

5.3. Hemodynamic Effects

Dexmedetomidine produces a typical biphasic hemodynamic response, resulting in an initial transient hypertension phase combined with marked reflex bradycardia due to α2-receptor activation and peripheral vasoconstriction due to initial peak plasma concentration. After a few minutes, the vasoconstriction attenuates, resulting in vasodilatation. Together with presynaptic α2-adrenoreceptors inhibiting sympathetic release of catecholamines and increased vagal activity, this results in a hypotensive phase. An average decrease in mean arterial blood pressure of 13–27% has been observed and is maintained for a prolonged period after the initial dose. A sustained dose-dependent reduction in circulating plasma catecholamines has been related to these long-lasting sympatholytic effects [7].
Furthermore, dexmedetomidine inhibits the antidiuretic action of vasopressin, increasing diuresis, enhancing osmolal clearance, and preventing cortical blood flow [31]. Moreover, it has been reported that high Dex plasma concentrations are associated with significant increases in systemic and pulmonary vascular resistance, resulting in pulmonary and systemic hypertension, which can be a relevant contraindication in patients with previous history of advanced heart block or severe ventricular dysfunction. In such cases, the loading dose sizes or the rate of infusion could be decreased [7]. The dose-dependent bradycardia seen with Dex treatment is mediated primarily by a decrease in sympathetic tone and partly by baroreceptor reflex and enhanced vagal activity [31]. This sympatholytic activity reduces myocardial oxygen consumption by decreasing metabolism, thereby minimizing the incidence of myocardial ischemia and improving survival. However, it should be mentioned that hypotension resulting from dexmedetomidine can also be potentially pro-ischemic.
The main side effects of Dex, as a selective α2-adrenoceptor agonist, are bradycardia [1] and hypotension [29,33,39,47,50], especially among elderly patients [48] or those with volume depletion [19]. However, Chang et al. compared the effects of Dex and propofol on hemodynamics in surgical ICU patients after major surgery and reported no significantly different incidences of bradycardia, hypotension, and cardiac index reduction between the two groups [53]. The combination with medication with negative chronotropic effects, such as beta-blockers or amiodarone, could worsen bradycardia, resulting in left anterior fascicular block, first-degree AV block, or asystole [54].
Compared with propofol in septic shock patients, the risk of hemodynamic instability and bradycardia seems not to be significantly higher [55]. Previously, Morelli et al. demonstrated that, when propofol was switched to Dex in septic patients, maintaining the same depth of sedation, vasopressor requirements were reduced [56]. According to the A2B trial, patients in the Dex group appeared to have similar hypotension risk to those in the clonidine group but higher than those in propofol [1], while, in the SPICE III post hoc analysis, Dex, in critically ill patients with septic shock, appeared to be associated with lower vasopressor requirements to achieve the target mean arterial blood pressure compared to standard sedatives [57]. However, subsequently, the French ADRESS (a2 Agonist Dexmedetomidine for Refractory Septic Shock) trial failed to demonstrate a reduction in vasopressor resistance in patients with refractory septic shock, and the study was terminated early due to high mortality [58].

5.4. Delirium in ICU

Agitated delirium among critically ill patients is a common source of increased morbidity and mortality, impaired cognitive function, and extended mechanical ventilation and length of ICU stay since it increases the risk of self-extubation and removal of other essential medical devices [19,59,60]. However, it seems that Dex has both preventive and therapeutic effects in delirium, unlike other sedatives (Table 2).

5.4.1. Delirium Prevention

It has been demonstrated that Dex-treated patients experience significantly less delirium in the ICU compared to patients treated with lorazepam [49], midazolam [49], or propofol [46,50,70,80] (Table 2). Many systematic reviews and meta-analyses support Dex’s possible perioperative delirium-preventive effect [80,81,82,83], especially among elderly patients [84], although they include a small number of heterogeneous RCTs, minimizing the strength of the evidence. This heterogeneity refers to the duration and time of Dex infusion (intra-, post-, or perioperative), the type of additional anesthesia (total intravenous or intravenous-inhaled combined), the severity and duration of the operation, age, and sample size.
A large number of clinical trials promote Dex’s possible delirium-preventive efficacy in ICU patients (Table 2). Particularly, MacLaren et al. reported that transitioning benzodiazepine sedation to Dex when patients qualify for daily awakenings may reduce delirium incidence and facilitate remembrance of ICU experiences without shortening the duration of mechanical ventilation [61]. Furthermore, Skrobik et al. evaluated Dex’s possible delirium preventive action through a two-center double-blind placebo-controlled RCT among ICU patients. They found that low-dose exclusively nocturnal Dex infusion reduced delirium incidence during ICU stay, although sleep quality remained unchanged, and length of mechanical ventilation and ICU and hospital LOS were similar between the two groups [62].
Moreover, a small Greek retrospective clinical trial compared Dex with standard of care in critically ill burn patients during weaning from mechanical ventilation. They found significantly lower rates of delirium and need for supplemental use of analgesic and antipsychotic agents in the Dex group, although the duration of mechanical ventilation was not reduced [44]. However, we have to mention that the patients in the Dex group were younger, suffering with a higher percentage of third-degree burn injury, evidence that could be a notable bias.
Conversely, after the Japanese DESIRE trial failed to reveal a clear delirium preventive benefit [29], Lee et al. also did not manage to prove any advantage of preventive infusion of Dex in postsurgical liver transplant patients, probably due to insufficient dose and duration and the unexpected low prevalence of delirium in both groups (<7.5%) [63]. In the DECADE study, Turan et al. suggested that the anti-inflammatory properties of a low dose of Dex may decrease postoperative delirium in cardiac surgery patients. However, the perioperative infusion of Dex did not decrease delirium onset in the treated group [64]. Similarly, He et al. could not demonstrate any delirium-preventive efficacy in postsurgical brain tumor patients, possibly because the incidence of delirium was not the primary endpoint of this study, the limited sample size, the presurgical affected neurological status, and the inadequate dose and duration of drug infusion [65].
Wang et al. compared the effect of sedation protocols with and without Dex on delirium risk and reported that the use of Dex could clinically slightly reduce the delirium risk, the ICU and hospital LOS, and the mechanical ventilation duration in ICU intubated patients [66]. They could not, however, find any benefit in mortality and the duration of delirium among the Dex-treated patients. They attributed these results to the small sample size and great heterogeneity of the 35 studies that were included in the meta-analysis [66].
The systematic review and meta-analysis by Heybati et al., which studied 41 trials including 3948 ICU mechanically ventilated patients, showed that Dex significantly reduced the duration of mechanical ventilation and the risk of ICU delirium in the cardiac surgical subgroup compared to propofol. Of note, the subgroup analysis also revealed that age might affect the incidence of hemodynamic side effects [48]. Another recent systematic review and meta-analysis, which included 16 controlled trials and 2035 ICU patients, compared Dex with midazolam and found a significantly shorter ICU LOS, duration of mechanical ventilation, and lower risk of delirium, showing a more obvious advantage in patients under 60 years old [50]. Of note, only 12 of the 16 RCTs (n = 1738 of 2035 patients) explored the incidence of delirium, while the included studies displayed statistical heterogeneity and small sample size.

5.4.2. Delirium Treatment

Dexmedetomidine is also promoted as an effective agent for controlling agitation among critically ill patients [69] (Table 2). The effectiveness of Dex as an agitated delirium treatment in mechanically ventilated ICU patients was determined by the DahLIA multicenter double-blind placebo-control trial, where Dex, when added to standard care, was associated with more brief delirium resolution, earlier extubation, and shorter length of mechanical ventilation and ICU stay, with the benefits of reduced opioid requirements and minimal memory and cognitive function impairments [59] (Table 2). Although there was a difference in the primary and several congruent secondary outcomes, this study was underpowered to detect significantly different endpoints, such as ICU LOS, due to its small sample size and the notable heterogeneity in baseline characteristics, such as duration of ventilation before randomization.
Moreover, Lu et al. revealed that early midazolam switched to Dex in agitated intubated ICU patients improved stress response and hemodynamic stability during extubation, reducing duration of mechanical ventilation and incidence of delirium and ICU and hospital LOS [67]. Another single-center RCT compared Dex’s efficacy in delirium treatment compared to benzodiazepine in 18 ICU polytrauma patients and found lower serum neuron-specific enolase (NSE), S100 calcium binding protein B (S100B), and brain-derived neurotrophic factor (BDNF) levels in the Dex group, evidence that appears to be associated with effective delirium treatment [85].
Subsequently, in an Australian target trial, emulation analysis reported that early initiation of Dex in ICU patients with delirium was more effective in agitation resolution (94% vs. 72% 30-day delirium resolution in Dex group vs. controls, respectively), minimizing the length of mechanical ventilation and the need for tracheostomy. Subgroup analyses revealed increased agitation resolution within 30 days in patients older than 65, mainly postsurgical, non-septic, and nonventilated, who were treated with Dex earlier than 12 h after the onset of agitation [68].
Moreover, Liu et al. verified the efficacy of Dex in treating delirium by reviewing data from ten RCTs and five non-RCTs including 1017 critically ill patients suffering from delirium. Dex treatment significantly reduced the duration of delirium compared with placebo or other sedative drugs [69]. However, this meta-analysis includes only limited trials, characterized by notable heterogeneity regarding the study population characteristics (postsurgical, cardiac-surgery, or pathological patients), the intervention (drug dose and infusion duration), the control group, and the endpoints [69].

5.4.3. Delirium Among Elderly Patients

Dexmedetomidine as both a preventive [70,71,73,74] and therapeutic agent for delirium has been examined more often among elderly ICU patients (Table 2). Djaiani et al. reported a significant prophylactic Dex benefit in frequency, time of onset, and duration of postoperative delirium in elderly patients after cardiac surgery compared to propofol [70]. However, patients who developed delirium in this single-center prospective RCT, which included 183 postsurgical cardiac patients aged >60 years, were older and had longer surgery and ICU LOS when compared with patients without delirium. The notable limitations of this study were the lack of blinding, the non-objective assessment of delirium using CAM and CAM-ICU scores, and the limited Dex infusion duration lasting less than 24 h, and then it was replaced by propofol. This practice may decrease Dex’s efficacy and probably explain the delayed delirium onset [70].
Moreover, Su et al. demonstrated that postoperatively prophylactic low-dose Dex (0.1 μg/kg/h) effectively prevented the occurrence of delirium and improved quality of sleep during the first 7 days in the ICU after non-cardiac surgery in elderly ICU patients aged >65 years (9% in Dex vs. 23% in placebo group; odds ratio 0.35, 95% CI 0.22–0.54; P < 0.0001) [71]. In addition, the DEXACET placebo-controlled RCT revealed that adequate postoperative analgesia and light sedation with Dex or propofol prevented delirium incidence among elderly cardiac surgical patients [73]. It has also been reported that postoperative Dex infusion via patient-controlled intravenous analgesia significantly minimized delirium prevalence, although it did not modify length of ICU and hospital LOS and mortality, among elderly patients undergoing thoracoabdominal tumor surgery [74].
Nevertheless, Deiner et al. reported that the intraoperative administration of Dex did not prevent postoperative delirium among elderly patients undergoing major elective non-cardiac surgery, possibly due to the short-acting nature of the drug and loss of salutary effects after discontinuation of the infusion [72]. The DIRECT trial was also underpowered to detect any preventive effect of Dex among elderly patients after undergoing cardiac surgery, probably due to its small sample size and the subjectivity of delirium diagnosis and assessment [43]. Similarly, Huet et al. failed to prove any delirium preventive benefit of Dex’s overnight infusion in older cardiac surgical patients, likely due to short-lasting drug action [75].
The effectiveness of Dex on delirium in elderly surgical patients was studied through a systematic review and meta-analysis of 21 trials including 6328 participants, where they concluded that Dex obviously decreased delirium occurrence in non-cardiac but not in cardiac elderly surgical patients [77]. Of note, only five of the twenty-one included studies assessed delirium in 1217 cardiac surgery patients, and the remaining sixteen studies included 5111 non-cardiac surgery patients. In addition, the authors remarked that there was moderate heterogeneity among all the RCTs regarding the time (intra-, peri-, or postoperative), the dose and the duration of Dex’s infusion, and the control group (placebo, propofol, or other sedative agent) [77].
Previously, Pereira et al. compared the delirium-preventive effect of Dex sedation with propofol through a systematic review of seven studies (six RCTs and two retrospective cohorts) that included 1249 elderly ICU patients and reported a lower delirium risk without raising adverse event frequency or outcome benefit [76]. It is important to note that this meta-analysis included notably heterogeneous studies regarding the study population (cardiac, non-cardiac surgical patients, or mixed medical and surgical ICU patients), the tool for delirium diagnosis, and the sedative protocol [76]. Recently, the protocol of the ALPHA 2 PRESENT Norwegian multicenter RCT was published, where the authors intended to evaluate the efficacy of Dex compared to clonidine or placebo in delirium prevention among elderly cardiac surgical patients [86]. Similarly, the protocol of the French EXACTUM placebo-controlled multicenter RCT has been announced, where Dex’s delirium-preventive effectiveness will also be assessed in cardiac postsurgical ICU patients older than 65 years [87].

5.4.4. Comparative Efficacy

Compared to clonidine, Dex has eight times more affinity for α2-adrenoreceptors, with more effective sedative and analgesic properties [19], while, in contrast to haloperidol, it has been described to be associated with fewer electrocardiographic changes in QT and QTc interval [88] (Table 2).
Carrasco et al. evaluated the clinical efficacy of Dex in non-intubated critically ill patients with agitated delirium refractory to haloperidol and reported better effectiveness, safety, and cost–benefit in cases where haloperidol failed to control delirium and was replaced by Dex [78]. Moreover, Shokri et al. evaluated the prophylactic efficacy of Dex and clonidine on rate and duration of delirium in older cardiac surgery patients. In this RCT, the authors found that postoperative Dex infusion was associated with lower risk and duration of delirium, shorter mechanical ventilation duration and ICU LOS, lower mortality rates, and lower morphine consumption than the clonidine group [79]. The major limitation of this study is that it is a single-center study with a small sample size, including possible bias regarding institutional standards of care. Furthermore, recently, the Japanese DEX-HD RCT was announced to compare the efficacy of Dex versus haloperidol in the treatment of nocturnal hyperactive delirium in non-intubated patients in high-dependency units [89].
Dexmedetomidine, as a sympatholytic a2-adrenergic agonist, is proposed to reduce stress response compared to commonly used sedatives in critically ill patients. Moore et al. measured plasma levels of stress response biomarkers in critically ill ventilated patients and showed that early sedation with Dex could not significantly change the physiological and serum parameters associated with the stress response as with midazolam or propofol [3]. On the other hand, Dex seems to decrease plasma stress hormones in cardiac surgery patients, when infused intraoperatively, compared with a placebo [2]. Zi et al. reported that perioperative Dex was related to significantly lower anxiety levels, after off-pump coronary artery bypass graft, compared with propofol [90].

5.4.5. Sleep Quality Improvement in ICU

Critically ill patients suffer from seriously disrupted sleep [91,92] and markedly segmented circadian rhythm [93]. Poor sleep quality has been related to delirium, long-lasting cognitive impairment, prolonged ICU and hospital stay, and increased morbidity and mortality [94]. Dexmedetomidine has been reported to more closely resemble natural non-REM sleep compared to GABA agonists, facilitating patient–caregiver interaction [91] (Table 3).
Wu et al. reported that prophylactic low-dose Dex infusion in non-intubated non-cardiac postsurgical elderly ICU patients improved and prolonged nocturnal sleep compared with a placebo [95]. Zhang et al. also randomized patients older than 65 years old, following non-cardiac surgery, to receive patient-controlled intravenous analgesia supplemented with either Dex or placebo [98]. In line with previous results [91,95,96], the authors showed that the Dex group had improved sleep structure and prolonged total sleep time [98]. In the MINDSS study, Qu et al. reported that a single bolus of Dex administered at nighttime could prevent delirium by promoting sleep. However, no difference was found regarding sleep quality as Dex is a short-acting drug and a single dose may not be sufficient to promote improved sleep quality [97].
Moreover, Dex treatment seems to increase total sleep duration and improve sleep efficiency in non-intubated ICU patients [94,95,98,99] and in mechanically ventilated patients [91,96] compared to a placebo or no sedation.
A recent systematic review of 29 trials including 5610 non-cardiac postoperative ICU patients explored the effects of perioperative Dex on postoperative sleep quality [100]. They concluded that Dex may improve the early postoperative quality pattern, although the evidence was of low quality.

5.4.6. Post-Intensive Care Syndrome

The term post-intensive care syndrome (PICS) refers to the current or worsening impairment of mental health, cognition, psychological, and physical vigor following critical illness [111]. Improvements in ICU patients’ short-term outcomes revealed the burden of survivors’ long-term quality-of-life issues. Perioperative treatment with dexmedetomidine has been associated with better postoperative analgesia and decreased opioid utilization [29], better quality of sleep [91,94], and minimized inflammatory response [112,113] —all of which could worsen post-ICU psychological and cognitive impairment [111]. Dong et al. reported significantly reduced rates of PICS and improved 6-month mortality in cardiac postsurgical patients treated with prophylactic nocturnal Dex administration [101] (Table 3).

5.4.7. Alcohol Withdrawal Syndrome

Alcohol withdrawal syndrome (AWS) occurs after a period of absolute or relative abstinence from alcohol, where blood alcohol levels decrease significantly in habituated individuals [114]. Diagnosis of AWS in ICU patients is associated with the need for excessive doses of sedatives, longer mechanical ventilation, ICU and hospital length of stay, and inflated healthcare costs [114]. Approximately 10–30% of ICU patients experience AWS, typically beginning within 6 to 24 h after alcohol cessation, peaking 36 h post-abstinence. The first-line treatment includes γ-aminobutyric acid agonists, such as benzodiazepines, for controlling persistent autonomic hyperactivity and preventing complications like seizures or delirium tremens [107,115]. The adjunctive agents evaluated include α2-agonists.
Intravenous dexmedetomidine has been reported to reduce neuronal loss in the locus coeruleus and catecholamine neuron degeneration, improving AWS symptoms [103]. Because of its anxiolytic and sedating effects, it can reduce benzodiazepine dosages, as shown in two retrospective cohort studies [102,103] (Table 3). In the first study, the 12-h change in benzodiazepine requirements differed significantly in 20 AWS patients treated with Dex compared to 22 treated with benzodiazepines alone [102]. The second cohort included 77 patients admitted to the medical ICU with severe AWS, showing that Dex improved Clinical Institute Withdrawal Assessment (CIWA) scores and benzodiazepine requirements, although it prolonged ICU length of stay [103].
In line with the above results, the systematic review of Woods et al., which included four studies with 55 AWS ICU patients, found that adjuvant Dex treatment added to benzodiazepine-based therapy reduced delirium risk and improved AWS control more effectively than benzodiazepine therapy alone [108]. Similarly, a second review by Wong, which studied thirteen articles (eight case reports/series and five RCTs), also reported the possible Dex adjuvant’s role in benzodiazepine-based therapy in AWS ICU patients [107]. It is important to note that both these reviews included heterogenic, small, and low-quality studies.
However, combining Dex with benzodiazepines or phenobarbital seems to prolong mechanical ventilation, ICU stay, and duration of delirium [104,106] without expected efficacy; studies have failed to demonstrate notable changes in CIWA score, intubation risk, or seizure control [104,105]. Dex possibly suppressed AWS signs and symptoms without treating the underlying withdrawal physiology as it has no GABA modulation effects, altering the kinetics of withdrawal in a manner that prolonged its duration. Additionally, Dex’s infusion requires ICU level of care due to potential cardiovascular complications. Thus, those patients who received prolonged Dex infusion probably remained in ICU because of the drug monitoring requirements [104].
Similarly to the above, Polintan et al., in a review of 12 studies, reported no significant benefit of adjunctive Dex added to benzodiazepine over benzodiazepine monotherapy regarding ICU and AWS control [109]. More recently, Fiore et al. also evaluated Dex as an adjunctive therapy for AWS and found that Dex failed to reduce intubation rates in patients with AWS, while it increased bradycardia risk [110]. Both of these meta-analyses included small low-quality heterogeneous studies.
Dexmedetomidine has been reported to have potential neuroprotective effects since it seems to dose-dependently decrease intracranial pressure, cerebral blood flow, and cerebral metabolic rate [21,84,116]. The underlying possible pathophysiologic mechanism for these neuroprotective effects is complicated and unclear. Sympatholytic effects of dexmedetomidine could minimize hypoxic brain damage, which is worsened through catecholamine release in nerve synapses since the latter increases neuronal sensitivity to glutamate during neuronal ischemia and promotes oxidative stress, worsening the existing nerve tissue impairment [8]. It also seems to reduce neuronal damage by inhibiting neurotransmitter release, which improves ischemic perfusion and metabolic disorders [117] (Table 4).
Animal studies presented reductions in neuronal cell death in the cortex and the hippocampus and a prevention effect on axonal damage and synaptic degeneration in the cortex while improving functional recovery after traumatic brain injury (TBI) [118]. A remarked limitation of this in vivo study is that the mice treated with Dex were obviously hypothermic. Thus, the demonstrated neuroprotective effects on TBI may not only be due to Dex’s activation of the signal transduction cascade but also the hypothermia induced by Dex [118]. Moreover, Feng et al. reported a significant alleviation of neurological deficits and brain edema in a TBI animal model, which was attributed to Dex-mediated inhibition of autophagy and neuroinflammation, ameliorating neuronal apoptosis related to the ROS/Nrf2 pathway [119]. Similarly, Li et al. demonstrated that Dex minimizes post-TBI inflammatory response by restriction of NF-κB activation through the Nrf2 signaling pathway [85], while Huang et al. imputed its anti-inflammatory and neuroprotective effects to its possible action on the TLR4/NF-κB pathway [120].
It has also been reported that dexmedetomidine presents neuroprotection in hypoxic injury through inhibition of NF-κB/COX-2 pathway activation [126]. Moreover, a protective effect on ischemia/reperfusion (I/R) injury has been described, preventing neuronal cells’ apoptosis through an intrinsic Bax–mitochondria–cytochrome c-caspase protease pathway [118].
Furthermore, retrospective studies based on the MIMIC-IV database found that adjuvant sedation with Dex was correlated with better outcomes among patients with aneurysmal subarachnoid hemorrhage (aSAH) [85,124], ischemic stroke [122,123], and TBI [116,121]. Possibly, Dex presented a protective effect in SAH patients either by anti-inflammatory or inflammation-modulatory properties, regulating three specific hub genes (MyD88, AR, and AREG), which have been identified as potential mediators of its protective effects [85], or by improving hemodynamics and cerebral perfusion, minimizing delayed cerebral ischemia [124]. Of note, Dex infusion did not improve the prognosis of patients with a low Charlson score ischemic stroke but instead provided greater clinical benefits for patients with multiple comorbidities, suggesting that, except from its neuroprotective effects, Dex may reveal benefits for other chronic diseases [123].
In line with this evidence, Lu et al. assessed the neuroprotective efficacy of Dex in TBI patients through internal jugular vein catheterization and evaluation of SjvO2, CEO2, and serum interleukin 6 (IL-6) and −1β (IL-1β) levels [125]. In the Dex group, they found significantly enhanced cerebral oxygen metabolism and reduced inflammatory biomarkers [125]. Moreover, a meta-analysis, which included 19 RCTs and 879 patients, amplified Dex’s possible neuroprotection effect on ischemic brain injury, attributed to hemodynamic stability, control of inflammatory and neuroendocrine response, and maintenance of intracranial homeostasis [21].

5.4.8. Anti-Inflammatory Effects

Dexmedetomidine is proposed to have cardio- and renoprotective and anti-inflammatory properties [7] (Table 5).

5.4.9. Cardio-Protective Properties

Dexmedetomidine as a sedative adjuvant has been proposed to modulate the inflammatory response after coronary artery bypass surgery (Table 5). Bolow et al. reported a significant reduction in postsurgical inflammatory biomarkers (IL-1, -6, TNF-α, INF-γ, and C-reactive protein) when Dex was used as an adjuvant sedative in cardiopulmonary bypass patients compared with non-Dex sedation. These findings indicate that Dex possibly modifies the inflammatory response in such cardiac surgery [112]. Moreover, Liu et al. compared the effects of Dex and propofol on sublingual microcirculation in patients after cardiac surgery and showed an earlier and greater improvement in the Dex group compared with propofol [127]. The possible mechanisms they proposed include inflammatory attenuation, decrease in leukocyte-endothelial interactions, mild hypocoagulation, and reduction in capillary perfusion deficits [127].
Furthermore, another small prospective RCT indicated a notable anti-inflammatory and myocardial protective effect when Dex was administered before anesthesia induction in patients during cardiac valve replacement [128]. In addition, the review of Zhang et al. evaluated the effect of Dex on myocardial I/R injury in patients undergoing cardiopulmonary bypass [129]. They included 17 retrospective studies, which involved 843 patients, and reported that the addition of Dex can significantly reduce the serum levels of CK-MB and troponin after cardiopulmonary bypass, suggesting its possible myocardial tissue protective effect [129].
In addition, further studies reveal that dexmedetomidine exerts dose-dependent renoprotective effects following surgery or during sepsis, likely attributed to anti-inflammatory, cytoprotective, and sympatholytic effects (Table 5). Cakir et al. demonstrated, in a kidney I/R experimental model, that Dex reduced I/R injury (lower levels of malondialdehyde, catalase, and glutathione), antioxidant enzyme activity, kidney dysfunction (lower levels of creatinine (Cr) and blood urea nitrogen (BUN)), and histologic injury score [130]. Of note, according to their results, applying 100 μg/kg of Dex instead of 10 μg/kg seems to be more effective in terms of healing kidney I/R damage [130]. A second animal study also investigated the possible protective effects of Dex in rat sepsis models [131]. They found that, when rats were treated with Dex, Acute Kidney Injury (AKI), induced by sepsis, was decreased significantly. In addition, animal model exposure to an a2-adrenergic receptor antagonist (yohimbine) eliminated this reduction [131].
The results of these experimental studies have been verified by two RCTs [113,132]. The first one, a small prospective randomized controlled placebo trial, showed that pretreatment with Dex in patients undergoing cardiac valve replacement under cardiopulmonary bypass attenuated renal injury and decreased the incidence of AKI [113]. This possibly renoprotective effect was associated with increased superoxidase activity [113]. The second RCT aimed to compare Dex with propofol and the incidence of AKI in septic mechanically ventilated patients [132]. The authors reported a significantly lower level of renal injury (Cr and BUN) and inflammatory biomarkers (TNF-α, IL-1, CD4+, and CD8+ T lymphocytes) in the Dex group, indicating a significant decrease in AKI incidence and renal replacement demands due to the possible anti-inflammatory and immunoregulatory action of Dex [132].
Dexmedetomidine has been shown to confer possible renoprotection by stabilizing the sympathetic system, exerting anti-inflammatory effects, and attenuating I/R injury. Liu et al. verified these findings after systematically reviewing 10 trials including 1575 postsurgical cardiac patients, reporting a significant reduction in postoperative AKI incidence [133]. However, this meta-analysis is based on low-quality evidence since it examined a limited number of trials with great heterogeneity regarding the studied population (coronary artery bypass or valve replacement; on- or off-pump), the intervention (Dex vs. placebo, or Dex vs. different sedatives), and adjuvant sedatives and analgesics [133]. A second meta-analysis by Zhao et al. that studied 15 trials, enrolling 2907 cardiac surgery patients, verified that perioperative Dex reduced the incidence of postoperative AKI [134]. Of note, the subgroup analyses revealed similar trends regardless of age, comorbidities, and the dose and duration of Dex infusion [134].
Finally, a recent high-level-of-evidence meta-analysis of 25 trials including 3997 postoperative patients revealed a significant reduction in AKI occurrence among individuals who were administered Dex in contrast to the control group [135]. The following subgroup analyses demonstrated that Dex did not have a statistically significant influence on subjects who underwent non-cardiac operations. This could be linked to the relatively lower incidence of AKI in non-cardiac surgery patients compared to cardiac patients, while the sample-size of the meta-analysis for non-cardiac patients was small [135].

5.4.10. Dexmedetomidine in Sepsis

Dexmedetomidine has been shown to minimize inflammatory response during sepsis in experimental models of sepsis [131]. However, data from clinical trials are limited (Table 5). An open-label pilot RCT by Iten et al., which compared Dex-based sedation with propofol- or midazolam-based sedation, failed to show any significant difference in septic encephalopathy biomarkers (S100-β) in critically ill patients with sepsis requiring mechanical ventilation [137]. These results were attributed to the small sample size (34 patients in the Dex group and 36 in the control group) and the increased dose of added propofol or midazolam in the Dex group in order to achieve sedation effectively [137]. Similarly, Patidar et al. revealed that Dex improves regional cerebral oxygen saturation in septic patients compared to propofol (utilizing Near-Infrared Spectroscopy monitor-NIRS), although no differences in delirium occurrence and duration were detected [138].
Moreover, the DecatSepsis trial proved to be underpowered to detect any significant reduction in mortality or norepinephrine dose in septic patients treated with Dex [136]. In addition, the ADRESS multicenter RCT, which was designed to evaluate Dex’s efficacy on the vasopressor response in patients with refractory septic shock, also failed to reveal any benefit and was terminated prematurely due to significantly higher mortality in the Dex arm [58]. Of note, sepsis is a complicated and heterogenic syndrome caused by a dysregulated host response to infection [142]. It is at least simplistic to try to correlate sepsis’ outcome with a single intervention, like Dex infusion.
Furthermore, Chen at al. examined the literature in order to determine Dex’s influence on mechanical ventilation duration among septic patients [139]. They reviewed only four studies and enrolled 349 patients, but they reported a significant increase in ventilator-free days and better 28-day survival in those patients treated with Dex [139]. In line with the above results, a second meta-analysis by Wang et al. failed to reveal any benefit of Dex use in septic patients compared with common sedative agents regarding mortality, ICU LOS, delirium incidence, and duration [140].
Subsequently, another meta-analysis by Zhang et al. included 19 RCTs that enrolled 1929 ICU septic patients treated either with Dex or other sedatives [141]. They reported significantly better survival in the Dex group compared with midazolam but not with propofol, and there was a notable elimination of inflammatory response. In line with the previous results, there were no differences in ICU LOS, mechanical ventilation duration, incidence of delirium, and rate of organ dysfunction [141].

6. Recommendations for Practice and/or Further Research

Dexmedetomidine seems to be an attractive alternative anesthetic for critically ill patients, with a variety of clinical indications. However, certain extended applications of Dex, such as neuroprotection or immunomodulation, require further evaluation.

7. Conclusions

In conclusion, compared with the current pharmacological management strategies, dexmedetomidine appears to have some advantages considering the pharmacokinetic profile of the drug. It has high specificity and selectivity for α2-adrenoreceptors and blocks norepinephrine, disrupting neurotransmitter pathways. It presents sedative effects that are comparable to GABA agonists, without respiratory depression, promoting a more physiological sleep–wake cycle, thereby prolonging sleep and improving its quality. As a result, Dex seems to eliminate the risk of delirium and post-ICU syndrome, especially among elderly and cardiac surgery ICU patients. Moreover, the latest evidence suggests a variety of extended indications for its use, such as neuroprotection as well as anti-inflammatory and immunomodulatory properties. These clinical applications require further evaluation and careful patient selection.

Author Contributions

C.A. conceived and designed the study, researched and reviewed the literature, and revised, reviewed, edited, and approved the final manuscript before submission. E.N.S., M.F., A.J. and V.E.P. contributed to the research and review of the literature, wrote the manuscript, and revised, edited, and approved the final manuscript before submission. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing is not applicable to this article as no new data were created or analyzed in this article.

Conflicts of Interest

All authors declare no competing interests.

References

  1. Walsh, T.S.; Parker, R.A.; Aitken, L.M.; McKenzie, C.A.; Emerson, L.; Boyd, J.; Macdonald, A.; Beveridge, G.; Giddings, A.; Hope, D.; et al. A2B Trial Investigators. Dexmedetomidine- or Clonidine-Based Sedation Compared With Propofol in Critically Ill Patients: The A2B Randomized Clinical Trial. JAMA 2025, 334, 32–45. [Google Scholar] [CrossRef] [PubMed]
  2. Wu, H.; Tang, J.; Pan, J.; Han, M.; Cai, H.; Zhang, H. Effects of dexmedetomidine on stress hormones in patients undergoing cardiac valve replacement: A randomized controlled trial. BMC Anesth. 2020, 20, 142. [Google Scholar] [CrossRef] [PubMed]
  3. Moore, J.P.R.; Shehabi, Y.; Reade, M.C.; Bailey, M.; Fraser, J.F.; Murray, L.; Anstey, C.; Singer, M. Stress response during early sedation with dexmedetomidine compared with usual-care in ventilated critically ill patients. Crit. Care 2022, 26, 359. [Google Scholar] [CrossRef] [PubMed]
  4. Patel, A.; Garg, R.; Bharti, S.J.; Kumar, V.; Gupta, N.; Mishra, S.; Bhatnagar, S.; Kumar, A. Comparison of sedation efficacy of intravenous infusion of dexmedetomidine versus propofol in terms of opioid consumption in patients requiring postoperative mechanical ventilation after head and neck onco-surgeries—A randomized prospective study. Indian. J. Cancer 2024, 61, 81–89. [Google Scholar] [CrossRef] [PubMed]
  5. Ibrahim, E.S.; Metwally, A.A.; Abdullatif, M.; Eid, E.A.; Mousa, M.G.; Sultan, A.A. Opioid sparing anesthesia in patients with liver cirrhosis undergoing liver resection: A controlled randomized double-blind study. BMC Anesth. 2025, 25, 53. [Google Scholar] [CrossRef] [PubMed]
  6. Nahmias, J.; Stopenski, S.; Jebbia, M.; Atallah, S.; Kirby, K.A.; Alvarez, C.A.; Aryan, N.; Tay-Lasso, E.; Dolich, M.; Lekawa, M.; et al. Dexmedetomidine for Analgesia in Nonintubated Patients With Traumatic Rib Fractures: A Randomized Clinical Trial. JAMA Surg. 2025, 160, 1047–1056. [Google Scholar] [CrossRef] [PubMed]
  7. Weerink, M.A.S.; Struys, M.M.R.F.; Hannivoort, L.N.; Barends, C.R.M.; Absalom, A.R.; Colin, P. Clinical Pharmacokinetics and Pharmacodynamics of Dexmedetomidine. Clin. Pharmacokinet. 2017, 56, 893–913. [Google Scholar] [CrossRef]
  8. Zhao, Y.; He, J.; Yu, N.; Jia, C.; Wang, S. Mechanisms of Dexmedetomidine in Neuropathic Pain. Front. Neurosci. 2020, 14, 330. [Google Scholar] [CrossRef]
  9. Giovannitti, J.A., Jr.; Thoms, S.M.; Crawford, J.J. Alpha-2 adrenergic receptor agonists: A review of current clinical applications. Anesth. Prog. 2015, 62, 31–39. [Google Scholar] [CrossRef] [PubMed]
  10. Nguyen, V.; Tiemann, D.; Park, E.; Salehi, A. Alpha-2 Agonists. Anesth. Clin. 2017, 35, 233–245. [Google Scholar] [CrossRef]
  11. Glaess, S.S.; Attridge, R.L.; Gutierrez, G.C. Clonidine as a strategy for discontinuing dexmedetomidine sedation in critically ill patients: A narrative review. Am. J. Health Syst. Pharm. 2020, 77, 515–522. [Google Scholar] [CrossRef] [PubMed]
  12. Turunen, H.; Jakob, S.M.; Ruokonen, E.; Kaukonen, K.M.; Sarapohja, T.; Apajasalo, M.; Takala, J. Dexmedetomidine versus standard care sedation with propofol or midazolam in intensive care: An economic evaluation. Crit. Care 2015, 19, 67. [Google Scholar] [CrossRef]
  13. Devlin, J.W.; Skrobik, Y.; Gélinas, C.; Needham, D.M.; Slooter, A.J.C.; Pandharipande, P.P.; Watson, P.L.; Weinhouse, G.L.; Nunnally, M.E.; Rochwerg, B.; et al. Clinical Practice Guidelines for the Prevention and Management of Pain, Agitation/Sedation, Delirium, Immobility, and Sleep Disruption in Adult Patients in the ICU. Crit. Care Med. 2018, 46, e825–e873. [Google Scholar] [CrossRef]
  14. Kotfis, K.; Zegan-Barańska, M.; Żukowski, M.; Kusza, K.; Kaczmarczyk, M.; Ely, E.W. Multicenter assessment of sedation and delirium practices in the intensive care units in Poland—Is this common practice in Eastern Europe? BMC Anesth. 2017, 17, 120. [Google Scholar] [CrossRef]
  15. Wang, J.; Peng, Z.Y.; Zhou, W.H.; Hu, B.; Rao, X.; Li, J.G. A National Multicenter Survey on Management of Pain, Agitation, and Delirium in Intensive Care Units in China. Chin. Med. J. 2017, 130, 1182–1188. [Google Scholar] [CrossRef]
  16. Møller, M.H.; Alhazzani, W.; Lewis, K.; Belley-Cote, E.; Granholm, A.; Centofanti, J.; McIntyre, W.B.; Spence, J.; Al Duhailib, Z.; Needham, D.M.; et al. Use of dexmedetomidine for sedation in mechanically ventilated adult ICU patients: A rapid practice guideline. Intensive Care Med. 2022, 48, 801–810. [Google Scholar] [CrossRef] [PubMed]
  17. Hume, N.E.; Zerfas, I.; Wong, A.; Klein-Fedyshin, M.; Smithburger, P.L.; Buckley, M.S.; Devlin, J.W.; Kane-Gill, S.L. Clinical Impact of the Implementation Strategies Used to Apply the 2013 Pain, Agitation/Sedation, Delirium or 2018 Pain, Agitation/Sedation, Delirium, Immobility, Sleep Disruption Guideline Recommendations: A Systematic Review and Meta-Analysis. Crit. Care Med. 2024, 52, 626–636. [Google Scholar] [CrossRef]
  18. Lewis, K.; Balas, M.C.; Stollings, J.L.; McNett, M.; Girard, T.D.; Chanques, G.; Kho, M.E.; Pandharipande, P.P.; Weinhouse, G.L.; Brummel, N.E.; et al. A Focused Update to the Clinical Practice Guidelines for the Prevention and Management of Pain, Anxiety, Agitation/Sedation, Delirium, Immobility, and Sleep Disruption in Adult Patients in the ICU. Crit. Care Med. 2025, 53, e711–e727. [Google Scholar] [CrossRef]
  19. Ungarian, J.; Rankin, J.A.; Then, K.L. Delirium in the Intensive Care Unit: Is Dexmedetomidine Effective? Crit. Care Nurse 2019, 39, e8–e21. [Google Scholar] [CrossRef]
  20. Scholz, J.; Finnerup, N.B.; Attal, N.; Aziz, Q.; Baron, R.; Bennett, M.I.; Benoliel, R.; Cohen, M.; Cruccu, G.; Davis, K.D.; et al. Classification Committee of the Neuropathic Pain Special Interest Group (NeuPSIG). The IASP classification of chronic pain for ICD-11, chronic neuropathic pain. Pain 2019, 160, 53–59. [Google Scholar] [CrossRef] [PubMed]
  21. Jiang, L.; Hu, M.; Lu, Y.; Cao, Y.; Chang, Y.; Dai, Z. The protective effects of dexmedetomidine on ischemic brain injury: A meta-analysis. J. Clin. Anesth. 2017, 40, 25–32. [Google Scholar] [CrossRef]
  22. Abu-Halaweh, S.; Obeidat, F.; Absalom, A.R.; AlOweidi, A.; Abeeleh, M.A.; Qudaisat, I.; Robinson, F.; Mason, K.P. Dexmedetomidine versus morphine infusion following laparoscopic bariatric surgery: Effect on supplemental narcotic requirement during the first 24 h. Surg. Endosc. 2016, 30, 3368–3374. [Google Scholar] [CrossRef] [PubMed]
  23. Obara, S.; Morimoto, I.; Iseki, Y.; Oishi, R.; Mogami, M.; Imaizumi, T.; Hosono, A.; Hakozaki, T.; Nakano, Y.; Isosu, T.; et al. The effect of obesity on dose of dexmedetomidine when administered with fentanyl during postoperative mechanical ventilation-retrospective. Fukushima J. Med. Sci. 2015, 61, 38–46. [Google Scholar] [CrossRef]
  24. Zhang, T.; Deng, Y.; He, P.; He, Z.; Wang, X. Effects of mild hypoalbuminemia on the pharmacokinetics and pharmacodynamics of dexmedetomidine in patients after major abdominal or thoracic surgery. J. Clin. Anesth. 2015, 27, 632–637. [Google Scholar] [CrossRef] [PubMed]
  25. Cortı’nez, L.I.; Anderson, B.J.; Holford, N.H.G.; Puga, V.; de la Fuente, N.; Auad, H.; Solari, S.; Allende, F.A. Dexmedetomidine pharmacokinetics in the obese. Eur. J. Clin. Pharmacol. 2015, 71, 1501–1508. [Google Scholar] [CrossRef]
  26. Wang, L.; Zhang, T.; Huang, L.; Peng, W. Comparison between Dexmedetomidine and Midazolam for Sedation in Patients with Intubation after Oral and Maxillofacial Surgery. Biomed. Res. Int. 2020, 2020, 7082597. [Google Scholar] [CrossRef]
  27. Yang, L.; Zhu, L.; Qi, B.; Zhang, Y.; Ni, C.; Zhang, Y.; Shi, X.; Xia, Q.; Masters, J.; Ma, D.; et al. Dexmedetomidine use during orthotopic liver transplantation surgery on early allograft dysfunction: A randomized controlled trial. Int. J. Surg. 2024, 110, 5518–5526. [Google Scholar] [CrossRef] [PubMed]
  28. Cheng, H.; Li, Z.; Young, N.; Boyd, D.; Atkins, Z.; Ji, F.; Liu, H.J. The Effect of Dexmedetomidine on Outcomes of Cardiac Surgery in Elderly Patients. J. Cardiothorac. Vasc. Anesth. 2016, 30, 1502–1508. [Google Scholar] [CrossRef] [PubMed]
  29. Kawazoe, Y.; Miyamoto, K.; Morimoto, T.; Yamamoto, T.; Fuke, A.; Hashimoto, A.; Koami, H.; Beppu, S.; Katayama, Y.; Itoh, M.; et al. Dexmedetomidine for Sepsis in Intensive Care Unit Randomized Evaluation (DESIRE) Trial Investigators. Dexmedetomidine for Sepsis in Intensive Care Unit Randomized Evaluation (DESIRE) Trial Investigators. Effect of dexmedetomidine on mortality and ventilator-free days in patients requiring mechanical ventilation with sepsis: A randomized clinical trial. JAMA 2017, 317, 1321–1328. [Google Scholar]
  30. Buckley, M.S.; Agarwal, S.K.; MacLaren, R.; Kane-Gill, S.L. Adverse Hemodynamic Events Associated With Concomitant Dexmedetomidine and Propofol for Sedation in Mechanically Ventilated ICU Patients. J. Intensive Care Med. 2020, 35, 1536–1545. [Google Scholar] [CrossRef] [PubMed]
  31. Lee, S. Dexmedetomidine: Present and future directions. Korean J. Anesth. 2019, 72, 323–330. [Google Scholar] [CrossRef] [PubMed]
  32. Gupta, S.; Singh, D.; Sood, D.; Kathuria, S. Role of dexmedetomidine in early extubation of the intensive care unit patients. J. Anaesthesiol. Clin. Pharmacol. 2015, 31, 92–98. [Google Scholar] [CrossRef] [PubMed]
  33. Zhou, W.J.; Liu, M.; Fan, X.P. Differences in efficacy and safety of midazolam vs. dexmedetomidine in critically ill patients: A meta-analysis of randomized controlled trial. Exp. Ther. Med. 2021, 21, 156. [Google Scholar] [CrossRef]
  34. Zhou, Y.; Yang, J.; Wang, B.; Wang, P.; Wang, Z.; Yang, Y.; Liang, G.; Jing, X.; Jin, X.; Zhang, Z.; et al. use of midazolam and dexmedetomidine for long-term sedation may reduce weaning time in selected critically ill, mechanically ventilated patients: A randomized controlled study. Crit. Care 2022, 26, 122. [Google Scholar] [CrossRef]
  35. Winings, N.A.; Daley, B.J.; Bollig, R.W.; Roberts, R.F., Jr.; Radtke, J.; Heidel, R.E.; Taylor, J.E.; McMillen, J.C. Dexmedetomidine versus propofol for prolonged sedation in critically ill trauma and surgical patients. Surgeon 2021, 19, 129–134. [Google Scholar] [CrossRef] [PubMed]
  36. Jakob, S.M.; Ruokonen, E.; Grounds, R.M.; Sarapohja, T.; Garratt, C.; Pocock, S.J.; Bratty, J.R.; Takala, J. Dexmedetomidine for Long-Term Sedation Investigators. Dexmedetomidine vs midazolam or propofol for sedation during prolonged mechanical ventilation: Two randomized controlled trials. JAMA 2012, 307, 1151–1160. [Google Scholar] [CrossRef]
  37. Brandão, P.G.M.; Lobo, F.R.; Ramin, S.L.; Sakr, Y.; Machado, M.N.; Lobo, S.M. Dexmedetomidine as an Anesthetic Adjuvant in Cardiac Surgery: A Cohort Study. Braz. J. Cardiovasc. Surg. 2016, 31, 213–218. [Google Scholar] [CrossRef]
  38. Elgebaly, A.S.; Sabry, M. Sedation effects by dexmedetomidine versus propofol in decreasing duration of mechanical ventilation after open heart surgery. Ann. Card. Anaesth. 2018, 21, 235–242. [Google Scholar] [CrossRef] [PubMed]
  39. Shehabi, Y.; Howe, B.D.; Bellomo, R.; Arabi, Y.M.; Bailey, M.; Bass, F.E.; Kadiman, S.B.; McArthur, C.J.; Murray, L.; Reade, M.C.; et al. Early Sedation with Dexmedetomidine in Critically Ill Patients. N. Engl. J. Med. 2019, 380, 2506–2517. [Google Scholar] [CrossRef] [PubMed]
  40. Aggarwal, J.; Lustrino, J.; Stephens, J.; Morgenstern, D.; Tang, W.Y. Cost-Minimization Analysis of Dexmedetomidine Compared to Other Sedatives for Short-Term Sedation During Mechanical Ventilation in the United States. Clin. Outcomes Res. 2020, 12, 389–397. [Google Scholar] [CrossRef]
  41. Shehabi, Y.; Serpa Neto, A.; Howe, B.D.; Bellomo, R.; Arabi, Y.M.; Bailey, M.; Bass, F.E.; Kadiman, S.B.; McArthur, C.J.; Reade, M.C.; et al. Early sedation with dexmedetomidine in ventilated critically ill patients and heterogeneity of treatment effect in the SPICE III randomised controlled trial. Intensive Care Med. 2021, 47, 455–466. [Google Scholar] [CrossRef]
  42. Hughes, C.G.; Mailloux, P.T.; Devlin, J.W.; Swan, J.T.; Sanders, R.D.; Anzueto, A.; Jackson, J.C.; Hoskins, A.S.; Pun, B.T.; Orun, O.M.; et al. Dexmedetomidine or Propofol for Sedation in Mechanically Ventilated Adults with Sepsis. N. Engl. J. Med. 2021, 384, 1424–1436. [Google Scholar] [CrossRef]
  43. Chitnis, S.; Mullane, D.; Brohan, J.; Noronha, A.; Paje, H.; Grey, R.; Bhalla, R.K.; Sidhu, J.; Klein, R. Dexmedetomidine Use in Intensive Care Unit Sedation and Postoperative Recovery in Elderly Patients Post-Cardiac Surgery (DIRECT). J. Cardiothorac. Vasc. Anesth. 2022, 36, 880–892. [Google Scholar] [CrossRef] [PubMed]
  44. Stangaciu, B.; Tsotsolis, S.; Papadopoulou, S.; Lavrentieva, A. Sedation With Dexmedetomidine in Critically Ill Burn Patients Reduced Delirium During Weaning From Mechanical Ventilation. Cureus 2022, 14, e31813. [Google Scholar] [CrossRef] [PubMed]
  45. Morris, S.; Lone, N.I.; McKenzie, C.A.; Weir, C.J.; Walsh, T.S. A2B trial investigators. Cost-Effectiveness of α2 Agonists for Intravenous Sedation in Patients With Critical Illness. JAMA Netw. Open 2025, 8, e2517533. [Google Scholar] [CrossRef] [PubMed]
  46. Chen, K.; Lu, Z.; Xin, Y.C.; Cai, Y.; Chen, Y.; Pan, S.M. Alpha-2 agonists for long-term sedation during mechanical ventilation in critically ill patients. Cochrane Database Syst. Rev. 2015, 1, CD010269. [Google Scholar] [CrossRef]
  47. Lewis, K.; Piticaru, J.; Chaudhuri, D.; Basmaji, J.; Fan, E.; Møller, M.H.; Devlin, J.W.; Alhazzani, W. Safety and Efficacy of Dexmedetomidine in Acutely Ill Adults Requiring Noninvasive Ventilation: A Systematic Review and Meta-analysis of Randomized Trials. Chest 2021, 159, 2274–2288. [Google Scholar] [CrossRef]
  48. Heybati, K.; Zhou, F.; Ali, S.; Deng, J.; Mohananey, D.; Villablanca, P.; Ramakrishna, H. Outcomes of dexmedetomidine versus propofol sedation in critically ill adults requiring mechanical ventilation: A systematic review and meta-analysis of randomised controlled trials. Br. J. Anaesth. 2022, 129, 515–526. [Google Scholar] [CrossRef]
  49. Lewis, K.; Alshamsi, F.; Carayannopoulos, K.L.; Granholm, A.; Piticaru, J.; Al Duhailib, Z.; Chaudhuri, D.; Spatafora, L.; Yuan, Y.; Centofanti, J.; et al. Dexmedetomidine vs other sedatives in critically ill mechanically ventilated adults: A systematic review and meta-analysis of randomized trials. Intensive Care Med. 2022, 48, 811–840. [Google Scholar] [CrossRef] [PubMed]
  50. Wen, J.; Ding, X.; Liu, C.; Jiang, W.; Xu, Y.; Wei, X.; Liu, X. A comparation of dexmedetomidine and midazolam for sedation in patients with mechanical ventilation in ICU: A systematic review and meta-analysis. PLoS ONE 2023, 18, e0294292. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  51. Lodenius, Å.; Ebberyd, A.; Hårdemark Cedborg, A.; Hagel, E.; Mkrtchian, S.; Christensson, E.; Ullman, J.; Scheinin, M.; Eriksson, L.I.; Fagerlund, M.J. Sedation with dexmedetomidine or propofol impairs hypoxic control of breathing in healthy male volunteers: A nonblinded, randomized crossover study. Anesthesiology 2016, 125, 700–715. [Google Scholar] [CrossRef]
  52. Peng, K.; Liu, H.-Y.; Wu, S.-R.; Cheng, H.; Ji, F.-H. Effects of combining dexmedetomidine and opioids for postoperative intravenous patient-controlled analgesia: A systematic review and meta-analysis. Clin. J. Pain 2015, 31, 1097–1104. [Google Scholar] [CrossRef]
  53. Chang, Y.F.; Chao, A.; Shih, P.Y.; Hsu, Y.C.; Lee, C.T.; Tien, Y.W.; Yeh, Y.C.; Chen, L.W. NTUH Center of Microcirculation Medical Research (NCMMR). Comparison of dexmedetomidine versus propofol on hemodynamics in surgical critically ill patients. J. Surg. Res. 2018, 228, 194–200. [Google Scholar] [CrossRef] [PubMed]
  54. Lazzari, L.; Donzelli, S.; Tordini, A.; Parise, A.; Pirozzi, C.; Di Meo, F.; Marallo, C.; Pace, V.; Marini, C.; Carreras, G. The point on the treatment of arrhythmic storm. Eur. Heart J. Suppl. 2024, 26 (Suppl. S1), i44–i48. [Google Scholar] [CrossRef]
  55. Nelson, K.M.; Patel, G.P.; Hammond, D.A. Effects from Continuous Infusions of Dexmedetomidine and Propofol on Hemodynamic Stability in Critically Ill Adult Patients with Septic Shock. J. Intensive Care Med. 2020, 35, 875–880. [Google Scholar] [CrossRef] [PubMed]
  56. Morelli, A.; Sanfilippo, F.; Arnemann, P.; Hessler, M.; Kampmeier, T.G.; D’Egidio, A.; Orecchioni, A.; Santonocito, C.; Frati, G.; Greco, E.; et al. The Effect of Propofol and Dexmedetomidine Sedation on Norepinephrine Requirements in Septic Shock Patients: A Crossover Trial. Crit. Care Med. 2019, 47, e89–e95. [Google Scholar] [CrossRef]
  57. Cioccari, L.; Luethi, N.; Bailey, M.; Shehabi, Y.; Howe, B.; Messmer, A.S.; Proimos, H.K.; Peck, L.; Young, H.; Eastwood, G.M.; et al. The effect of dexmedetomidine on vasopressor requirements in patients with septic shock: A subgroup analysis of the Sedation Practice in Intensive Care Evaluation [SPICE III] Trial. Crit. Care 2020, 24, 441. [Google Scholar] [CrossRef]
  58. Dargent, A.; Bourredjem, A.; Jacquier, M.; Bohe, J.; Argaud, L.; Levy, B.; Fournel, I.; Cransac, A.; Badie, J.; Quintin, L.; et al. Dexmedetomidine to Reduce Vasopressor Resistance in Refractory Septic Shock: α2 Agonist Dexmedetomidine for REfractory Septic Shock (ADRESS): A Double-Blind Randomized Controlled Pilot Trial. Crit. Care Med. 2025, 53, e884–e896. [Google Scholar] [CrossRef] [PubMed]
  59. Reade, M.C.; Eastwood, G.M.; Bellomo, R.; Bailey, M.; Bersten, A.; Cheung, B.; Davies, A.; Delaney, A.; Ghosh, A.; van Haren, F.; et al. Effect of Dexmedetomidine Added to Standard Care on Ventilator-Free Time in Patients With Agitated Delirium: A Randomized Clinical Trial. JAMA 2016, 315, 1460–1468. [Google Scholar] [CrossRef] [PubMed]
  60. Raquer, A.P.; Fong, C.T.; Walters, A.M.; Souter, M.J.; Lele, A.V. Delirium and Its Associations with Critical Care Utilizations and Outcomes at the Time of Hospital Discharge in Patients with Acute Brain Injury. Medicina 2024, 60, 304. [Google Scholar] [CrossRef]
  61. MacLaren, R.; Preslaski, C.R.; Mueller, S.W.; Kiser, T.H.; Fish, D.N.; Lavelle, J.C.; Malkoski, S.P. A randomized, double-blind pilot study of dexmedetomidine versus midazolam for intensive care unit sedation: Patient recall of their experiences and short-term psychological outcomes. J. Intensive Care Med. 2015, 30, 167–175. [Google Scholar] [CrossRef]
  62. Skrobik, Y.; Duprey, M.S.; Hill, N.S.; Devlin, J.W. Low-Dose Nocturnal Dexmedetomidine Prevents ICU Delirium. A Randomized, Placebo-controlled Trial. Am. J. Respir. Crit. Care Med. 2018, 197, 1147–1156. [Google Scholar] [CrossRef]
  63. Lee, H.; Yang, S.M.; Chung, J.; Oh, H.W.; Yi, N.J.; Suh, K.S.; Oh, S.Y.; Ryu, H.G. Effect of Perioperative Low-Dose Dexmedetomidine on Postoperative Delirium After Living-Donor Liver Transplantation: A Randomized Controlled Trial. Transpl. Proc. 2020, 52, 239–245. [Google Scholar] [CrossRef]
  64. Turan, A.; Duncan, A.; Leung, S.; Karimi, N.; Fang, J.; Mao, G.; Hargrave, J.; Gillinov, M.; Trombetta, C.; Ayad, S.; et al. Dexmedetomidine for reduction of atrial fibrillation and delirium after cardiac surgery (DECADE): A randomised placebo-controlled trial. Lancet 2020, 396, 177–185. [Google Scholar] [CrossRef]
  65. He, X.; Cheng, K.M.; Duan, Y.Q.; Xu, S.S.; Gao, H.R.; Miao, M.Y.; Li, H.L.; Chen, K.; Yang, Y.L.; Zhang, L.; et al. Feasibility of low-dose dexmedetomidine for prevention of postoperative delirium after intracranial operations: A pilot randomized controlled trial. BMC Neurol. 2021, 21, 472. [Google Scholar] [CrossRef] [PubMed]
  66. Wang, S.; Hong, Y.; Li, S.; Kuriyama, A.; Zhao, Y.; Hu, J.; Luo, A.; Sun, R. Effect of dexmedetomidine on delirium during sedation in adult patients in intensive care units: A systematic review and meta-analysis. J. Clin. Anesth. 2021, 69, 110157. [Google Scholar] [CrossRef]
  67. Lu, X.; Li, J.; Li, T.; Zhang, J.; Li, Z.B.; Gao, X.J.; Xu, L. Clinical study of midazolam sequential with dexmedetomidine for agitated patients undergoing weaning to implement light sedation in intensive care unit. Chin. J. Traumatol. 2016, 19, 94–96. [Google Scholar] [CrossRef] [PubMed]
  68. Serpa Neto, A.; Young, M.; Phongphithakchai, A.; Maeda, A.; Hikasa, Y.; Pattamin, N.; Kitisin, N.; Premaratne, G.; Chan, G.; Furler, J.; et al. A target trial emulation of dexmedetomidine to treat agitation in the intensive care unit. Crit. Care Sci. 2025, 37, e20250010. [Google Scholar] [CrossRef]
  69. Liu, X.; Xiong, J.; Tang, Y.; Gong, C.C.; Wang, D.F. Role of dexmedetomidine in the treatment of delirium in critically ill patients: A systematic review and meta-analysis. Minerva Anestesiol. 2021, 87, 65–76. [Google Scholar] [CrossRef] [PubMed]
  70. Djaiani, G.; Silverton, N.; Fedorko, L.; Carroll, J.; Styra, R.; Rao, V.; Katznelson, R. Dexmedetomidine versus propofol sedation reduces delirium after cardiac, surgery: A randomized controlled trial. Anesthesiology 2016, 124, 362–368. [Google Scholar] [CrossRef]
  71. Su, X.; Meng, Z.T.; Wu, X.H.; Cui, F.; Li, H.L.; Wang, D.X.; Zhu, X.; Zhu, S.N.; Maze, M.; Ma, D. Dexmedetomidine for prevention of delirium in elderly patients after non-cardiac surgery: A randomised, double-blind, placebo-controlled trial. Lancet 2016, 388, 1893–1902. [Google Scholar] [CrossRef]
  72. Deiner, S.; Luo, X.; Lin, H.M.; Sessler, D.I.; Saager, L.; Sieber, F.E.; Lee, H.B.; Sano, M.; Jankowski, C.; the Dexlirium Writing Group; et al. Intraoperative Infusion of Dexmedetomidine for Prevention of Postoperative Delirium and Cognitive Dysfunction in Elderly Patients Undergoing Major Elective Noncardiac Surgery: A Randomized Clinical Trial. JAMA Surg. 2017, 152, e171505. [Google Scholar] [CrossRef]
  73. Subramaniam, B.; Shankar, P.; Shaefi, S.; Mueller, A.; O’Gara, B.; Banner-Goodspeed, V.; Gallagher, J.; Gasangwa, D.; Patxot, M.; Packiasabapathy, S.; et al. Effect of Intravenous Acetaminophen vs Placebo Combined With Propofol or Dexmedetomidine on Postoperative Delirium Among Older Patients Following Cardiac Surgery: The DEXACET Randomized Clinical Trial. JAMA 2019, 321, 686–696. [Google Scholar] [CrossRef]
  74. Xie, K.; Chen, J.; Tian, L.; Gu, F.; Pan, Y.; Huang, Z.; Fang, J.; Yu, W.; Zhou, H. Postoperative infusion of dexmedetomidine via intravenous patient-controlled analgesia for prevention of postoperative delirium in elderly patients undergoing surgery. Aging Clin. Exp. Res. 2023, 35, 2137–2144. [Google Scholar] [CrossRef] [PubMed]
  75. Huet, O.; Gargadennec, T.; Oilleau, J.F.; Rozec, B.; Nesseler, N.; Bouglé, A.; Kerforne, T.; Lasocki, S.; Eljezi, V.; Dessertaine, G.; et al. EXACTUM and the Atlanrea Study Group. Prevention of post-operative delirium using an overnight infusion of dexmedetomidine in patients undergoing cardiac surgery: A pragmatic, randomized, double-blind, placebo-controlled trial. Crit. Care 2024, 28, 64. [Google Scholar] [CrossRef]
  76. Pereira, J.V.; Sanjanwala, R.M.; Mohammed, M.K.; Le, M.L.; Arora, R.C. Dexmedetomidine versus propofol sedation in reducing delirium among older adults in the ICU: A systematic review and meta-analysis. Eur. J. Anaesthesiol. 2020, 37, 121–131. [Google Scholar] [CrossRef] [PubMed]
  77. Lin, C.; Tu, H.; Jie, Z.; Zhou, X.; Li, C. Effect of Dexmedetomidine on Delirium in Elderly Surgical Patients: A Meta-analysis of Randomized Controlled Trials. Ann. Pharmacother. 2021, 55, 624–636. [Google Scholar] [CrossRef]
  78. Carrasco, G.; Baeza, N.; Cabré, L.; Portillo, E.; Gimeno, G.; Manzanedo, D.; Calizaya, M. Dexmedetomidine for the Treatment of Hyperactive Delirium Refractory to Haloperidol in Nonintubated ICU Patients: A Nonrandomized Controlled Trial. Crit. Care Med. 2016, 44, 1295–1306. [Google Scholar] [CrossRef]
  79. Shokri, H.; Ali, I. A randomized control trial comparing prophylactic dexmedetomidine versus clonidine on rates and duration of delirium in older adult patients undergoing coronary artery bypass grafting. J. Clin. Anesth. 2020, 61, 109622. [Google Scholar] [CrossRef] [PubMed]
  80. Liu, X.; Xie, G.; Zhang, K.; Song, S.; Song, F.; Jin, Y.; Fang, X. Dexmedetomidine vs propofol sedation reduces delirium in patients after cardiac surgery: A meta-analysis with trial sequential analysis of randomized controlled trials. J. Crit. Care 2017, 38, 190–196. [Google Scholar] [CrossRef]
  81. Flükiger, J.; Hollinger, A.; Speich, B.; Meier, V.; Tontsch, J.; Zehnder, T.; Siegemund, M. Dexmedetomidine in prevention and treatment of postoperative and intensive care unit delirium: A systematic review and meta-analysis. Ann. Intensive Care 2018, 8, 92. [Google Scholar] [CrossRef]
  82. Zhang, J.; Yu, Y.; Miao, S.; Liu, L.; Gan, S.; Kang, X.; Zhu, S. Effects of peri-operative intravenous administration of dexmedetomidine on emergence agitation after general anesthesia in adults: A meta-analysis of randomized controlled trials. Drug Des. Devel. Ther. 2019, 13, 2853–2864. [Google Scholar] [CrossRef] [PubMed]
  83. Fu, Q.; Sandeep, B.; Li, H.; Wang, B.S.; Huang, X. Impact of perioperative dexmedetomidine on postoperative delirium in adult undergoing cardiac surgery: A comprehensive bibliometrix and meta-analysis. Asian J. Psychiatr. 2025, 108, 104522. [Google Scholar] [CrossRef]
  84. Pan, H.; Liu, C.; Ma, X.; Xu, Y.; Zhang, M.; Wang, Y. Perioperative dexmedetomidine reduces delirium in elderly patients after non-cardiac surgery: A systematic review and meta-analysis of randomized-controlled trials. Can. J. Anaesth. 2019, 66, 1489–1500. [Google Scholar] [CrossRef]
  85. Li, F.; Wang, X.; Zhang, Z.; Zhang, X.; Gao, P. Dexmedetomidine Attenuates Neuroinflammatory-Induced Apoptosis after Traumatic Brain Injury via Nrf2 signaling pathway. Ann. Clin. Transl. Neurol. 2019, 6, 1825–1835. [Google Scholar] [CrossRef] [PubMed]
  86. Neerland, B.E.; Busund, R.; Haaverstad, R.; Helbostad, J.L.; Landsverk, S.A.; Martinaityte, I.; Norum, H.M.; Ræder, J.; Selbaek, G.; Simpson, M.R.; et al. Alpha-2-adrenergic receptor agonists for the prevention of delirium and cognitive decline after open heart surgery (ALPHA2PREVENT): Protocol for a multicentre randomised controlled trial. BMJ Open 2022, 12, e057460. [Google Scholar] [CrossRef] [PubMed]
  87. Gargadennec, T.; Oilleau, J.F.; Rozec, B.; Nesseler, N.; Lasocki, S.; Futier, E.; Amour, J.; Durand, M.; Bougle, A.; Kerforne, T.; et al. Dexmedetomidine after Cardiac Surgery for Prevention of Delirium (EXACTUM) trial protocol: A multicentre randomised, double-blind, placebo-controlled trial. BMJ Open 2022, 12, e058968. [Google Scholar] [CrossRef]
  88. Haspolat, A.; Polat, F.; Köprülü, A.Ş. A Comparative Analysis of the Effects of Haloperidol and Dexmedetomidine on QTc Interval Prolongation during Delirium Treatment in Intensive Care Units. J. Crit. Care Med. 2024, 10, 222–231. [Google Scholar] [CrossRef]
  89. Minami, T.; Watanabe, H.; Kato, T.; Ikeda, K.; Ueno, K.; Matsuyama, A.; Maeda, J.; Sakai, Y.; Harada, H.; Kuriyama, A.; et al. Dexmedetomidine versus haloperidol for sedation of non-intubated patients with hyperactive delirium during the night in a high dependency unit: Study protocol for an open-label, parallel-group, randomized controlled trial (DEX-HD trial). BMC Anesth. 2023, 23, 193. [Google Scholar] [CrossRef]
  90. Zi, J.; Fan, Y.; Dong, C.; Zhao, Y.; Li, D.; Tan, Q. Anxiety administrated by dexmedetomidine to prevent new-onset of postoperative atrial fibrillation in patients undergoing off-pump coronary artery bypass graft. Int. Heart J. 2020, 61, 263–272. [Google Scholar] [CrossRef]
  91. Georgopoulos, D.; Kondili, E.; Alexopoulou, C.; Younes, M. Effects of Sedatives on Sleep Architecture Measured With Odds Ratio Product in Critically Ill Patients. Crit. Care Explor. 2021, 3, e0503. [Google Scholar] [CrossRef]
  92. Showler, L.; Deane, A.M.; Litton, E.; Ankravs, M.J.; Wibrow, B.; Barge, D.; Goldin, J.; Hammond, N.; Saxena, M.K.; Young, P.J.; et al. A multicentre point prevalence study of nocturnal hours awake and enteral pharmacological sleep aids in patients admitted to Australian and New Zealand intensive care units. Crit. Care Resusc. 2024, 26, 192–197. [Google Scholar] [CrossRef] [PubMed]
  93. Sertaridou, E.N.; Chouvarda, I.G.; Arvanitidis, K.I.; Filidou, E.K.; Kolios, G.C.; Pnevmatikos, I.N.; Vasilios E Papaioannou, V.E. Melatonin and cortisol exhibit different circadian rhythm profiles during septic shock depending on timing of onset: A prospective observational study. Ann. Intensive Care 2018, 8, 118. [Google Scholar] [CrossRef] [PubMed]
  94. Lu, W.; Fu, Q.; Luo, X.; Fu, S.; Hu, K. Effects of dexmedetomidine on sleep quality of patients after surgery without mechanical ventilation in ICU. Medicine 2017, 96, e7081. [Google Scholar] [CrossRef]
  95. Wu, X.H.; Cui, F.; Zhang, C.; Meng, Z.T.; Wang, D.X.; Ma, J.; Wang, G.F.; Zhu, S.N.; Ma, D. Low-dose Dexmedetomidine Improves Sleep Quality Pattern in Elderly Patients after Noncardiac Surgery in the Intensive Care Unit: A Pilot Randomized Controlled Trial. Anesthesiology 2016, 125, 979–991. [Google Scholar] [CrossRef] [PubMed]
  96. Sun, Y.M.; Zhu, S.N.; Zhang, C.; Li, S.L.; Wang, D.X. Effect of low-dose dexmedetomidine on sleep quality in postoperative patients with mechanical ventilation in the intensive care unit: A pilot randomized trial. Front. Med. 2022, 9, 931084. [Google Scholar] [CrossRef]
  97. Qu, J.Z.; Mueller, A.; McKay, T.B.; Westover, M.B.; Shelton, K.T.; Shaefi, S.; D’Alessandro, D.A.; Berra, L.; Brown, E.N.; Houle, T.T.; et al. Nighttime dexmedetomidine for delirium prevention in non-mechanically ventilated patients after cardiac surgery (MINDDS): A single-centre, parallel-arm, randomised, placebo-controlled superiority trial. EClinicalMedicine 2023, 56, 101796. [Google Scholar] [CrossRef]
  98. Zhang, Z.F.; Su, X.; Zhao, Y.; Zhong, C.L.; Mo, X.Q.; Zhang, R.; Wang, K.; Zhu, S.N.; Shen, Y.E.; Zhang, C.; et al. Effect of mini-dose dexmedetomidine supplemented intravenous analgesia on sleep structure in older patients after major noncardiac surgery: A randomized trial. Sleep. Med. 2023, 102, 9–18. [Google Scholar] [CrossRef]
  99. Sun, P.; Liang, X.Q.; Chen, N.P.; Ma, J.H.; Zhang, C.; Shen, Y.E.; Zhu, S.N.; Wang, D.X. Impact of mini-dose dexmedetomidine supplemented analgesia on sleep structure in patients at high risk of obstructive sleep apnea: A pilot trial. Front. Neurosci. 2024, 18, 1426729. [Google Scholar] [CrossRef]
  100. Wang, L.; Liang, X.Q.; Sun, Y.X.; Hua, Z.; Wang, D.X. Effect of perioperative dexmedetomidine on sleep quality in adult patients after noncardiac surgery: A systematic review and meta-analysis of randomized trials. PLoS ONE 2024, 19, e0314814. [Google Scholar] [CrossRef]
  101. Dong, C.H.; Gao, C.N.; An, X.H.; Li, N.; Yang, L.; Li, D.C.; Tan, Q. Nocturnal dexmedetomidine alleviates post-intensive care syndrome following cardiac surgery: A prospective randomized controlled clinical trial. BMC Med. 2021, 19, 306. [Google Scholar] [CrossRef] [PubMed]
  102. VanderWeide, L.A.; Foster, C.J.; MacLaren, R.; Kiser, T.H.; Fish, D.N.; Mueller, S.W. Evaluation of early dexmedetomidine addition to the standard of care for severe alcohol withdrawal in the ICU: A retrospective controlled cohort study. J. Intensive Care Med. 2016, 31, 198–204. [Google Scholar] [CrossRef]
  103. Beg, M.; Fisher, S.; Siu, D.; Rajan, S.; Troxell, L.; Liu, V.X. Treatment of alcohol withdrawal syndrome with and without dexmedetomidine. Perm. J. 2016, 20, 49–53. [Google Scholar] [CrossRef] [PubMed]
  104. Yavarovich, E.R.; Bintvihok, M.; McCarty, J.C.; Breeze, J.L.; LaCamera, P. Association between dexmedetomidine use for the treatment of alcohol withdrawal syndrome and intensive care unit length of stay. J. Intensive Care 2019, 7, 49. [Google Scholar] [CrossRef]
  105. Collier, T.E.; Farrell, L.B.; Killian, A.D.; Kataria, V.K. Effect of Adjunctive Dexmedetomidine in the Treatment of Alcohol Withdrawal Compared to Benzodiazepine Symptom-Triggered Therapy in Critically Ill Patients: The EvADE Study. J. Pharm. Pract. 2022, 35, 356–362. [Google Scholar] [CrossRef]
  106. Ware, L.R.; Schuler, B.R.; Goodberlet, M.Z.; Marino, K.K.; Lupi, K.E.; DeGrado, J.R. Evaluation of Dexmedetomidine as an Adjunct to Phenobarbital for Alcohol Withdrawal in Critically Ill Patients. J. Intensive Care Med. 2023, 38, 553–561. [Google Scholar] [CrossRef]
  107. Wong, A.; Smithburger, P.L.; Kane-Gill, S.L. Review of adjunctive dexmedetomidine in the management of severe acute alcohol withdrawal syndrome. Am. J. Drug Alcohol. Abus. 2015, 41, 382–391. [Google Scholar] [CrossRef] [PubMed]
  108. Woods, A.D.; Giometti, R.; Weeks, S.M. The use of dexmedetomidine as an adjuvant to benzodiazepine-based therapy to decrease the severity of delirium in alcohol withdrawal in adult intensive care unit patients: A systematic review. JBI Database Syst. Rev. Implement. Rep. 2015, 13, 224–252. [Google Scholar] [CrossRef] [PubMed][Green Version]
  109. Polintan, E.T.T.; Danganan, L.M.L.; Cruz, N.S.; Macapagal, S.C.; Catahay, J.A.; Patarroyo-Aponte, G.; Azmaiparashvili, Z.; Lo, K.B. Adjunctive Dexmedetomidine in Alcohol Withdrawal Syndrome: A Systematic Review and Meta-analysis of Retrospective Cohort Studies and Randomized Controlled Trials. Ann. Pharmacother. 2023, 57, 696–705. [Google Scholar] [CrossRef]
  110. Fiore, M.; Alfieri, A.; Torretta, G.; Passavanti, M.B.; Sansone, P.; Pota, V.; Simeon, V.; Chiodini, P.; Corrente, A.; Pace, M.C. Dexmedetomidine as Adjunctive Therapy for the Treatment of Alcohol Withdrawal Syndrome: A Systematic Review and Meta-Analysis. Pharmaceuticals 2024, 17, 1125. [Google Scholar] [CrossRef]
  111. Yuan, C.; Timmins, F.; Thompson, D.R. Post-intensive care syndrome: A concept analysis. Int. J. Nurs. Stud. 2021, 114, 103814. [Google Scholar] [CrossRef]
  112. Bulow, N.M.H.; Colpo, E.; Pereira, R.P.; Correa, E.F.M.; Waczuk, E.P.; Duarte, M.F.; Rocha, J.B.T. Dexmedetomidine decreases the inflammatory response to myocardial surgery under mini-cardiopulmonary bypass. Braz. J. Med. Biol. Res. 2016, 49, e4646. [Google Scholar] [CrossRef]
  113. Zhai, M.; Kang, F.; Han, M.; Huang, X.; Li, J. The effect of dexmedetomidine on renal function in patients undergoing cardiac valve replacement under cardiopulmonary bypass: A double-blind randomized controlled trial. J. Clin. Anesth. 2017, 40, 33–38. [Google Scholar] [CrossRef] [PubMed]
  114. Maldonado, J.R. Novel algorithms for the prophylaxis and management of alcohol withdrawal syndromes—Beyond benzodiazepines. Crit. Care Clin. 2017, 33, 559–599. [Google Scholar] [CrossRef]
  115. Glahn, A.; Proskynitopoulos, P.J.; Bleich, S.; Hillemacher, T. Pharmacotherapeutic management of acute alcohol withdrawal syndrome in critically Ill patients. Expert. Opin. Pharmacother. 2020, 21, 1083–1092. [Google Scholar] [CrossRef]
  116. Wang, J.; Sun, J.Q.; Lu, Y.; Yang, Q.L.; Zhao, P.L.; Hang, C.H.; Li, W. Delirium as a mediating factor in the survival benefits of dexmedetomidine in acute brain injury management. Sci. Rep. 2025, 15, 30937. [Google Scholar] [CrossRef]
  117. Wang, K.; Wu, M.; Xu, J.; Wu, C.; Zhang, B.; Wang, G.; Ma, D. Effects of dexmedetomidine on perioperative stress, inflammation, and immune function: Systematic review and meta-analysis. Br. J. Anaesth. 2019, 123, 777–794. [Google Scholar] [CrossRef] [PubMed]
  118. Wu, J.; Vogel, T.; Gao, X.; Lin, B.; Kulwin, C.; Chen, J. Neuroprotective effect of dexmedetomidine in a murine model of traumatic brain injury. Sci. Rep. 2018, 8, 4935. [Google Scholar] [CrossRef] [PubMed]
  119. Feng, X.; Ma, W.; Zhu, J.; Jiao, W.; Wang, Y. Dexmedetomidine alleviates early brain injury following traumatic brain injury by inhibiting autophagy and neuroinflammation through the ROS/Nrf2 signaling pathway. Mol. Med. Rep. 2021, 24, 661. [Google Scholar] [CrossRef]
  120. Huang, G.R.; Hao, F.G. Dexmedetomidine Inhibits Inflammation to Alleviate Early Neuronal Injury via TLR4/NF-κB Pathway in Rats with Traumatic Brain Injury. Crit. Rev. Eukaryot. Gene Expr. 2021, 31, 41–47. [Google Scholar] [CrossRef]
  121. Liu, S.Y.; Kelly-Hedrick, M.; Komisarow, J.; Hatfield, J.; Ohnuma, T.; Treggiari, M.M.; Colton, K.; Arulraja, E.; Vavilala, M.S.; Laskowitz, D.T.; et al. Association of Early Dexmedetomidine Utilization With Clinical Outcomes After Moderate-Severe Traumatic Brain Injury: A Retrospective Cohort Study. Anesth. Analg. 2024, 139, 366–374. [Google Scholar] [CrossRef]
  122. Chen, S.; Ren, S.; Li, X.; Liu, K. Dexmedetomidine administration is associated with a reduced risk of ICU mortality in critically ill patients with ischemic stroke. Front. Neurol. 2025, 16, 1571957. [Google Scholar] [CrossRef]
  123. Yang, Y.; Zhang, J.; Wang, H.; Zhang, Y.; Zhang, J. Association between dexmedetomidine sedation and mortality in critically ill patients with ischaemic stroke: A retrospective study based on MIMIC-IV database. BMJ Open 2025, 15, e101395. [Google Scholar] [CrossRef] [PubMed]
  124. Liu, Y.; Peng, J.; Zhang, Y.H.; Liu, H.T. Dexmedetomidine is Associated with Reduced In-Hospital Mortality Risk of Patients with Subarachnoid Hemorrhage Undergoing Surgery. World Neurosurg. 2025, 194, 123539. [Google Scholar] [CrossRef]
  125. Lu, S.; Song, H.; Lin, Y.; Song, B.; Lin, S. A randomized controlled trial investigating the impact of early goal-directed sedation dominated by dexmedetomidine on cerebral oxygen metabolism and inflammatory mediators in patients with severe brain injury. Neurol. Sci. 2025, 46, 1741–1750. [Google Scholar] [CrossRef]
  126. Pan, W.; Lin, L.; Zhang, N.; Yuan, F.; Hua, X.; Wang, Y.; Mo, L. Neuroprotective Effects of Dexmedetomidine Against Hypoxia-Induced Nervous System Injury are Related to Inhibition of NF-κB/COX-2 Pathways. Cell Mol. Neurobiol. 2016, 36, 1179–1188. [Google Scholar] [CrossRef]
  127. Liu, X.; Zhang, K.; Wang, W.; Xie, G.; Cheng, B.; Wang, Y.; Hu, Y.; Fang, X. Dexmedetomidine Versus Propofol Sedation Improves Sublingual Microcirculation After Cardiac Surgery: A Randomized Controlled Trial. J. Cardiothorac. Vasc. Anesth. 2016, 30, 1509–1515. [Google Scholar] [CrossRef] [PubMed]
  128. Yuan, B.; Huang, X.; Wen, J.; Peng, M. Dexmedetomidine Pretreatment Confers Myocardial Protection and Reduces Mechanical Ventilation Duration for Patients Undergoing Cardiac Valve Replacement under Cardiopulmonary Bypass. Ann. Thorac. Cardiovasc. Surg. 2024, 30, oa-23. [Google Scholar] [CrossRef] [PubMed]
  129. Zhang, G.R.; Peng, C.M.; Liu, Z.Z.; Leng, Y.F. The effect of dexmedetomidine on myocardial ischemia/reperfusion injury in patients undergoing cardiac surgery with cardiopulmonary bypass: A meta-analysis. Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 7409–7417. [Google Scholar] [CrossRef]
  130. Cakir, M.; Polat, A.; Tekin, S.; Vardi, N.; Taslidere, E.; Duran, Z.R.; Tanbek, K. The effect of dexmedetomidine against oxidative and tubular damage induced by renal ischemia reperfusion in rats. Ren. Fail. 2015, 37, 704–708. [Google Scholar] [CrossRef]
  131. Tan, F.; Chen, Y.; Yuan, D.; Gong, C.; Li, X.; Zhou, S. Dexmedetomidine protects against acute kidney injury through downregulating inflammatory reactions in endotoxemia rats. Biomed. Rep. 2015, 3, 365–370. [Google Scholar] [CrossRef]
  132. Liu, J.; Shi, K.; Hong, J.; Gong, F.; Mo, S.; Chen, M.; Zheng, Y.; Jiang, L.; Xu, L.; Tu, Y.; et al. Dexmedetomidine protects against acute kidney injury in patients with septic shock. Ann. Palliat. Med. 2020, 9, 224–230. [Google Scholar] [CrossRef] [PubMed]
  133. Liu, Y.; Sheng, B.; Wang, S.; Lu, F.; Zhen, J.; Chen, W. Dexmedetomidine prevents acute kidney injury after adult cardiac surgery: A meta-analysis of randomized controlled trials. BMC Anesth. 2018, 18, 7. [Google Scholar] [CrossRef] [PubMed]
  134. Zhao, C.; Liu, S.; Zhang, H.; Gao, M. Does dexmedetomidine reduce the risk of acute kidney injury after cardiac surgery? A meta-analysis of randomized controlled trials. Braz. J. Anesth. 2024, 74, 744446. [Google Scholar] [CrossRef]
  135. Zhao, J.; Tang, M.H.; Shen, Q.H.; Xu, D.C. The effectiveness of dexmedetomidine for preventing acute kidney injury after surgery: A systematic review and meta-analysis. Front. Med. 2024, 11, 1414794. [Google Scholar] [CrossRef]
  136. Al-Regal, A.R.E.; Ramzy, E.A.; Atia, A.A.A.; Emara, M.M. Dexmedetomidine for Reducing Mortality in Patients With Septic Shock: A Randomized Controlled Trial (DecatSepsis). Chest 2024, 166, 1394–1405. [Google Scholar] [CrossRef]
  137. Iten, M.; Bachmann, K.; Jakob, S.M.; Grandgirard, D.; Leib, S.L.; Cioccari, L. Adjunctive Sedation with Dexmedetomidine for the Prevention of Severe Inflammation and Septic Encephalopathy: A Pilot Randomized Controlled Study. Crit. Care Med. 2025, 53, e1377–e1388. [Google Scholar] [CrossRef] [PubMed]
  138. Patidar, A.K.; Khanna, P.; Kashyap, L.; Ray, B.R.; Maitra, S. Utilization of NIRS Monitor to Compare the Regional Cerebral Oxygen Saturation Between Dexmedetomidine and Propofol Sedation in Mechanically Ventilated Critically ill Patients with Sepsis- A Prospective Randomized Control Trial. J. Intensive Care Med. 2025, 40, 379–387. [Google Scholar] [CrossRef]
  139. Chen, P.; Jiang, J.; Zhang, Y.; Li, G.; Qiu, Z.; Levy, M.M.; Hu, B. Effect of Dexmedetomidine on duration of mechanical ventilation in septic patients: A systematic review and meta-analysis. BMC Pulm. Med. 2020, 20, 42. [Google Scholar] [CrossRef]
  140. Wang, C.; Chen, Q.; Wang, P.; Jin, W.; Zhong, C.; Ge, Z.; Xu, K. The Effect of Dexmedetomidine as a Sedative Agent for Mechanically Ventilated Patients With Sepsis: A Systematic Review and Meta-Analysis. Front. Med. 2021, 8, 776882. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  141. Zhang, T.; Mei, Q.; Dai, S.; Liu, Y.; Zhu, H. Use of dexmedetomidine in patients with sepsis: A systematic review and meta-analysis of randomized-controlled trials. Ann. Intensive Care 2022, 12, 81. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  142. Singer, M.; Deutschman, C.S.; Seymour, C.W.; Shankar-Hari, M.; Annane, D.; Bauer, M.; Bellomo, R.; Bernard, G.R.; Chiche, J.D.; Coopersmith, C.M.; et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 2016, 315, 801–810. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Flow diagram. This review included peer-reviewed articles in English, published since 2015, in PubMed. It contained all kinds of original clinical research (animal experimental studies, observational studies, retrospective studies, and randomized clinical trials (RCTs)) and a few relevant reviews and meta-analyses, principally referring to Dex’s mechanism of action, pharmacodynamics, and pharmacokinetics.
Figure 1. Flow diagram. This review included peer-reviewed articles in English, published since 2015, in PubMed. It contained all kinds of original clinical research (animal experimental studies, observational studies, retrospective studies, and randomized clinical trials (RCTs)) and a few relevant reviews and meta-analyses, principally referring to Dex’s mechanism of action, pharmacodynamics, and pharmacokinetics.
Healthcare 13 02882 g001
Table 1. Dexmedetomidine as sedative in ICU patients.
Table 1. Dexmedetomidine as sedative in ICU patients.
Study DesignStudy PopulationIntervention Comparison Main OutcomesSecondary OutcomesAdverse EventsStudy Limitations
Turunen H, et al., 2015 [12]Post hoc analysis of MIDEX and PRODEX study (Jakob SM, et al. JAMA 2012;307:1151–60) [36]990 ICU adult patients requiring prolonged MV493 Dex group
250 midazolam (MIDEX)
247 propofol (PRODEX)
Dex vs. propofol or midazolamReduce total ICU resource utilization and respective costs- Shortening the extubation time and
- MV duration, especially in comparison with midazolam
Not referred - Great heterogeneity of study population.
- ICU patients with severe neurological disorders were excluded.
Gupta S, et al., 2015 [32]RCT40 ICU MV patients0.2–0.7 mcg/kg/h Dex vs. 0.04–0.2 mg/kg/h midazolamDex vs. midazolamDex significantly minimized time to extubationEasy arousability,
Lack of respiratory depression
Not referred A small single-center study
Cheng H, et al., 2016 [28]Single-center Retrospective clinical trial505 ICU CAP patients, aged > 65 years0.24–0.6 μgr/kg/h Dex Dex vs. other sedativesDex decreased in-hospital (0.90 vs. 2.83%) and operative mortality (1.35 vs. 3.18%)Dex reduced risk of
stroke (0.9 vs. 1.77%),
delirium (7.21% vs. 10.95%)
Not referred A single-center non-randomized cohort study, with a relatively small sample size
Brandão PGM, et al., 2016 [37]Single-center Retrospective clinical trial1302 ICU CAP patients0.5 μgr/kg/h DexDex vs. other sedativesDex was associated with lower 30-day mortality (3.4 vs. 9.7%)Shorter ICU LOS (3.1 vs. 7.3%)
Decreased incidence of neurological problems
Not referred
-
A single-center retrospective study
-
Dex patients were younger and underwent more often off-pump surgery
-
Time of CPB was shorter in DEX group
Kawazoe Y, et al., 2017 [29]Multicenter RCT (8 ICUs in Japan) (DESIRE trial)201 septic MV patientsSedation with Dex vs. without DexSame 28-day mortality (22.8 vs. 30.8%) and
Ventilator-free days (20 vs. 18)
Dex group had higher rate of well-controlled sedation but
no reduction in delirium
Higher rates of bradycardia in Dex group
-
An open-label study
-
The end points were assessed by physicians at discharge
-
RASS and CAM-ICU scores were assessed by “not blinded” nurses
-
Only short-term outcomes were analyzed
Elgebaly AS, et al. Ann Card Anae [38]Single-center RCT50 ICU CAP patients, aged 18–55 years 0.8 μgr/kg/h Dex vs. 1.5 mg/kg/h propofolDex vs. propofolNo difference between groups in extubation time,
analgesic requirements, or ICU and hospital LOS
Same hemodynamic stability,
Increased financial costs in Dex group
Not referred
-
Small sample size
-
Different cardiovascular operations (CABG or valve/CABG) with different possibility for postsurgical respiratory failure
-
Many subjects required more than one sedative agent, increasing MV duration
Shehabi Y, et al., 2019 [39]Multicenter RCT (SPICE III trial)4000 patientsDex as sole or primary sedative or usual careDex vs. other sedativesSame 90-day mortality,Dex group required supplemental sedativesHigher rates of bradycardia and hypotension in Dex group
-
Unblinded RCT
-
Patients that required deep sedation were not excluded
-
No sedation daily interruption
-
No sedation managing protocol
-
No delirium prevention protocol
Wang L, et al., 2020 [26]Single-center RCT40 oral and maxillofacial postsurgical ICU, intubated patients0.1 μgr/kg/h Dex vs. 0.2 mg/kg/h midazolamDex vs. midazolamBPS and Ramsay scores were higher in midazolam group.
Shorter time of extubation in Dex group.
Lower incidence of delirium and respiratory depression in Dex group.Higher rates of bradycardia in Dex groupA small single-center study
Aggarwal J, et al., 2020 [40]A single-center cost analysis studyShort term sedation in ICU, MV patientsDex vs. other sedativesDex reduced costs/person
Dex reduced ICU LOS
Not referred
-
A single-center open label trial
-
The cost analysis model was suitable for US national healthcare system
Winings NA, et al.,2021 [35]Single-center RCT57 trauma MV ICU patientsDex vs. propofolNo significant difference in MV duration (125 vs. 130 h)
Dex achieved better target sedation
No difference in mortality,
ICU and hospital LOS,
or incidence of delirium.
Similar incidences of bradycardia and hypotension
-
Single-center open label study
-
Patients in Dex group spent more time in target sedation
Shehabi Y, et al., 2021 [41]Secondary analysis of SPICE III trial1825 ICU patients, MV more than 24 h, aged > 65 yearsDex vs. standard sedationDex vs. other sedativesReduced 90-day mortality in patients >65 years oldHigh probability to increase 90-day mortality in younger non-operative patientsNo difference
-
A secondary post hoc analysis
-
Lack of blinding
-
Absence of a strictly protocolized strategy fer sedation or delirium management
Hughes CH, et al., 2021 [42]Multicenter RCT (MENDS 2 trial)432 ICU MV septic patientsDex vs. propofolNo difference in mortality,
Delirium risk, or
ventilator-free days
Not referredLack of pain and delirium prevention strategy
Zhou Y, et al., 2022 [34]Single-Center RCT252 ICU, MV patients, long lasting weaning0.2–0.7μ g/kg/h Dex plus midazolam vs. plus propofol vs. midazolam alone during SBTWhen midazolam was switched to Dex, faster extubationLess weaning time,
Lower delirium,
No difference in adverse events rate
-
Unblinded single-center trial
-
The use of midazolam was not in accordance with the PAD 2013 and PADIS 2018 guidelines
Chitnis S, et al., 2022 [43]Single-Center RCT (DIRECT study)70 postsurgical cardiac patients, older than 75 years oldDex vs. propofolDecrease in MCAS score in Dex groupNo difference in delirium rate, time to extubation, length of hospital stayNo difference in adverse events rate
-
A small open-label single-center study,
-
Only postsurgical cardiac patients.
Stangaciu B, et al., 2022 [44]Single-Center Retrospective clinical trial56 MV, burn ICU patients, during weaningDex 1 mcg/kg bolus and 0.4–0.1 mcg/kg/h iv vs. 0.08 mg/kg/h midazolam or 15–30 mcg/kg/min propofol Dex vs. midazolam or propofolNo significantly shorter duration of MV (9.3 vs. 7.5 days),
Lower delirium rate (38.4 vs. 53.3%) (assessed with CAM-ICU score),
Less need for supplemental analgesia (23.1 vs. 53.3%) (assessed with RASS score),
Less need for antipsychotic agents (15.4 vs. 53.3%)
- Bradycardia
- Abdominal distention
- A small retrospective single-center study.
- Patients with Dex sedation were younger and had higher percentage of 3rd-degree burn injuries.
Walsh TS, et al., 2025 [1]Multicenter Open-label RCT (41 ICUs in UK)
(A2B trial)
1404 adult, MV, ICU patientsPropofol + opioids vs. Dex or clonidine + opioidDex or clonidine vs. propofolReduced MV duration (Dex: 136 h, clonidine: 146 h and propofol: 162 h)More agitation in Dex group and clonidine vs. propofol,
Similar mortality
Higher rate of bradycardia in Dex and clonidine group
Morris S, et al., 2025 [45]Multicenter Open-label RCT (41 ICUs in UK) (economic evaluation of A2B trial)1404 adult, MV, ICU patientsPropofol + opioids vs. Dex or clonidine + opioidDex or clonidine vs. propofolSimilar costs and QALYs Not referredA post hoc economic evaluation of previous RCTs
Chen K, et al., 2015 [46]Systematic Review of 7 studies1624 adult patients requiring prolonged MVDex vs. propofol, or midazolam, or lorazepamReduced duration of MV by 22%Reduced length of ICU stay by 14%
Zhou WJ, et al., 2021 [33]Systematic Review of 8 studies1379 patientsDex vs. midazolamDex reduced LOS in ICU,
time to extubation,
delirium
No difference in hypotension incidence and mortalityHigher incidence of bradycardia with Dex
Lewis K, et al., 2021 [47]Systematic Review of 12 RCTs738 NIV patientsDex vs. other sedativesReduced the risk of intubation
Reduced ICU LOS
Reduced risk of delirium- Bradycardia
- Hypotension
-
Heybati K, et al., 2022 [48]Systematic Review of 41 studies3948 MV ICU patientsDex vs. propofolNo difference in ICU LOSReduced MV duration and delirium risk only among cardiac surgical patients- Bradycardia,
- Hypotension, especially among elderly
Lewis K, et al., 2022 [49]Systematic Review of 77 RCTs11,997 patients MV ICU patientsDex vs. other sedativesReduced MV duration
Reduced ICU LOS
Reduced delirium risk
-
Bradycardia,
-
Hypotension
-
Wen J, et al., 2023 [50]Systemic Review of 16 RCTs2035 patientsDex vs. midazolam
-
Shorter LOS ICU
-
Lower risk of delirium
-
Shorter duration of MV
- No difference in mortality and
- length of hospital stay
- Bradycardia,
- No difference in hypotension
Abbreviations: BPS: Behavioral Pain Scale, CABG: coronary artery bypass graft, CAP: cardiopulmonary bypass, Dex: dexmedetomidine, h: h., ICU: intensive care unit, LOS: length of stay, MCAS score: Minnesota Cognitive Acuity Screen score, MV: mechanical ventilation, PAD: Pain, Anxiety and Delirium, PADIS: Pain, Anxiety, Agitation/Sedation, Delirium, Immobility and Sleep Disruption, QALYs: quality-adjusted life years, RCT: randomized controlled trial, SBT: spontaneous breathing trial, UK: United Kingdom.
Table 2. Indications for dexmedetomidine in ICU delirium.
Table 2. Indications for dexmedetomidine in ICU delirium.
Study DesignStudy PopulationComparison Assessment of DeliriumMain OutcomesSecondary OutcomesStudy Limitations
2.1 Delirium Prevention
MacLaren R, et al., 2015 [61]Single-center double-blind RCT11 ICU MV patients in weaning time received Dex vs. 12 midazolam0.61 μg/kg/h Dex vs. 3.7 mg/h midazolam- CAM-ICUDex reduced delirium incidences
-
Rates of anxiety and depression were similar.
-
Higher risk of hypotension in Dex group
-
A small single-center trial
Kawazoe Y, et al., 2017 [29] Open-label, multicenter RCT (8 ICUs in Japan) (DESIRE trial)201 ICU septic MV patientsSedation with Dex vs. without Dex- CAM-ICUSame 28-day mortality (22.8 vs. 30.8%) and
Ventilator-free days (20 vs. 18)
-
Dex group had higher rate of well-controlled sedation but
-
no reduction in delirium
-
An open-label study
-
The end points were assessed by physicians at discharge
-
RASS and CAM-ICU scores were assessed by “not blinded” nurses
-
Only short-term outcomes were analyzed
Skrobik Y, et al., 2018 [62]Two-center double-blind RCT100 ICU patientsi.v. 0.2 μg/kg/h Dex from 9:30 pm to 6:15 am vs. placebo, until ICU discharge- ICU Delirium Screening Checklist every 12 h.
- LSEQ
Decrease incidence of ICU delirium (20% Dex group vs. 46% placebo group)No difference in
-
sleep quality
-
incidence of hypotension or bradycardia
-
More ICU days free of coma in Dex group
-
Early mobility was infrequently used
-
Patients with obstructive sleep apnea were not excluded
Lee H, et al., 2020 [63]Single-center RCT217 Postsurgical liver transplant patientsPerioperative i.v. 0.1 μg/kg/h Dex for 48 h vs. placebo- CAM-ICU every 8 h postoperativelyNo difference in delirium incidence (9% Dex group vs. 5.9% placebo group)No difference in
-
Delirium duration
-
Length of MV
-
ICU LOS
-
Hospital LOS
-
In-hospital mortality
-
3-month mortality
-
Relatively small sample size
-
Only elective living-donor liver transplant
-
Unexpected low total delirium incidence in both groups
-
Probably insufficient Dex’s dose and duration infusion
Turan A, et al., 2020 [64]Multicenter RCT
(DECADE trial)
794 cardiac surgery patientsPerioperative infusion of Dex vs. placebo
-
CAM-ICU
-
LSEQ
No significant difference in atrial fibrillation incidence (30% in Dex group vs. 34% in placebo group)No significant difference in delirium incidence (17% in Dex group vs. 12% in placebo group)
-
Subjective delirium assessment
-
Underestimated delirium incidence
He X, et al., 2021 [65]Single-center RCT60 Postsurgical brain-surgical patientsPerioperative i.v. 0.1 μg/kg/h Dex for <24 h vs. placebo- CAM-ICU every 12 h. postoperativelyDrug interruption rateNo difference in
-
delirium incidence
-
bradycardia
-
hypotension
-
respiratory depression
-
Relatively small sample size
-
Only brain tumor patients, with preoperative neurological dysfunction
-
Studies’ primary endpoint was the study-drug interruption
Wang S, et al., 2021 [66]Systematic Review of 36 studies9623 ICU patientsDex vs. non-Dex sedation Dex was associated with
-
reduced of delirium risk
-
higher incidence of bradycardia
-
and hypotension
Dex was associated with
-
shorter ICU LOS
-
shorter hospital LOS
-
shorter MV duration
-
No improved mortality
-
Low- or very low-quality evidence
-
Great heterogeneity of included trails
Stangaciu B, et al., 2022 [44]Single-center Retrospective Clinical Trial56 MV, severe burn ICU patients, during weaningDex 1 mcg/kg bolus and 0.4–0.1 mcg/kg/h iv vs. 0.08 mg/kg/h midazolam or 15–30 mcg/kg/min propofol- CAM-ICU
-
No significantly shorter duration of MV (9.3 vs. 7.5 days),
-
Lower delirium rate in Dex group (38.4 vs. 53.3%),
-
less need for supplemental analgesia (23.1 vs. 53.3%),
-
less need for antipsychotic agents (15.4 vs. 53.3%)
-
more incidence of bradycardia in Dex group
-
Single-center retrospective study
-
Limited sample size
-
Dex patients were younger with higher percentage of 3rd-degree burn injury
Heybati K, et al. [48]Systematic review of 41 trials3948 ICU, MV patientsDex vs. propofol
-
No significant difference in ICU LOS
-
Reduced MV duration
-
Reduced delirium risk among cardiac surgical patients
-
Dex was associated with greater risk of bradycardia, especially among older patients
-
Included publications with high risk of bias
Wen J, et al. [50]Systematic review of 16 RCTs2035 ICUDex vs. midazolam
-
Dex achieved shorter ICU LOS
-
Lower risk of delirium
-
Shorter duration of MV
-
More incidence of bradycardia
No difference in
-
Hypotension
-
Mortality
-
12 of the 16 trials (n = 1738) explored the incidence of delirium
-
The studies displayed statistical heterogeneity and small sample size
2.2 Delirium Treatment
Reade MC, et al. [59]Multicenter, double-blind RCT
(DahLIA study)
74 adult intubated ICU patients with agitated deliriumDex initially at a rate of 0.5 μg/kg/h and then titrated to rates 0–1.5 μg/kg/h vs. placebo- CAM-ICU
- MAAS score
- Increased ventilator-free h at 7 days (144.8 vs. 127.5 h.),
- Earlier extubation (21.9 vs. 44.3 h.)
- Accelerated resolution of delirium (23.3 vs. 40.0 h.)
- Lower quantities of other sedatives and opioids
- Shorter ICU LOS (2.9 vs. 4.1 days)
- no difference in incidence of bradycardia, hypotension, or temporarily agitation between the 2 groups
- Relatively small sample size
- Different duration of MV before randomization (144.8 h in Dex group vs. 127.5 in placebo)
Lu X, et al. [67]Single-center double-blind RCT80 agitated intubated ICU patientsGroup A: i.v. 0.3–3 mg/kg/h midazolam for 24 h and then i.v. 0.2–1 μg/kg/h Dex
Group B: only i.v. 0.3–3 mg/kg/h midazolam
-
HR and MAP were significantly higher in group B during extubation
-
Lower incidence of delirium in Dex group (20% vs. 45%) after extubation
-
Lower extubation time in group A
-
Lower hospital LOS in group A
-
Single-center study
-
Small sample size
-
The timing, dosage, and Dex’s target group need to be validated
Serpa Neto A, et al., 2025 [68]Target-trial emulation2052 ICU patients’ records with delirium (314 treated with Dex)Dex vs. other sedatives
-
Early initiation of Dex had higher rates of resolution of ICU agitation (94% 30-days delirium resolution vs. 72%)
-
Lower risk of tracheostomy by day 30
-
Lower mortality (5% vs. 9%)
-
Dex patients were younger, had more severe illness, and were more likely to have unplanned department admissions
-
Potential confounders, such pain level, RASS score, or history of mental illness, medications, or addiction, were not included in the model
Liu X, et al., 2021 [69]Systematic Review of 10 RCTs and 5 non-RCTs1017 critically ill patients with deliriumDex vs. other agents for delirium treatment
-
Reduced delirium frequency,
-
Shorter time of delirium resolution
-
Higher rate of bradycardia
-
Only a few studies were included in this comparison
-
High heterogeneity in outcome and interventions
2.3 Delirium among elderly patients
Djaiani G, et al., 2016 [70]Single-center RCT183 Cardiac postsurgical patients > 60 years oldi.v. 0.4 μg/kg bolus and 0.2–0.7 μg/kg /h Dex max for 24 h.
vs. 25–50 μg/kg/min propofol
- CAM-ICU 12 h postoperatively
- CAM when discharged ICU
Decrease incidence of postoperative delirium (17 vs. 31.5%)
-
Earlier onset of delirium in propofol group (1st vs. 2nd postoperatively day)
-
Shorter duration of delirium in Dex group (2 vs. 3 days)
-
Dex-based sedation was cost savings
-
Lack of blinding of drugs infusion
-
Assessment of CAM and CAM-ICU were not objective
-
The duration of Dex’s infusion was less than 24 h.
Su X, et al., 2016 [71]Multicenter, double-blind RCT700 ICU non-cardiac surgical patients > 65 years oldi.v. 0.1 μg/kg/h Dex vs. placeboCAM-ICU every 12 h.Decrease incidence of postoperative delirium (9 vs. 23%)Hypertension and tachycardia more frequently in placebo (18 vs. 10% and 14 vs. 7%, respectively)
-
Lack of baseline delirium assessment, before surgery
-
CAM-ICU is not a sensitive tool for delirium assessment
Deiner S, et al., 2017 [72]Multicenter double-blind RCT404 patients > 65 years old, undergoing non-cardiac surgeryIntraoperative use of i.v. 0.5 μg/kg/h Dex and 2 h postoperatively vs. placebo
-
MMSE
-
CAM every day
-
CAM-ICU every day
Dex did not significantly reduce postoperative delirium (12.2% vs. 11.4%)
-
Daily delirium assessment, during working h, could resalt to undetected evening delirium
Subramaniam B, et al., 2019 [73]RCT120 cardiac surgical patients aged > 60 yearsPostoperative sedation with propofol vs. Dex and analgesia with acetaminophen vs. placebo
-
CAM-ICU every day
-
Patients treated with acetaminophen had less delirium incidence than placebo group (10 vs. 28%)
-
Patients treated with Dex had no significant difference in delirium rates vs. propofol (17 vs. 21%)
-
No difference in ICU and hospital LOS
-
A relatively small sample size
-
Possible undetected delirium during evening
Chitnis S, et al., 2022 [43]Single-center RCT (DIRECT study)70 postsurgical cardiac patients, >75 years oldDex vs. propofolNot referred Decrease in MCAS score in Dex groupNo difference in
-
delirium rate,
-
time to extubation,
-
hospital LOS
-
A small open-label single-center study,
-
Only postsurgical cardiac patients.
Xie K, et al., 2023 [74]Single-center RCT236 patients > 60 years old undergoing thoracoabdominal tumor surgeryPostoperative Dex + sufentanil via PCIA vs. only sufentanil via PCIANot referredThe incidence of delirium was significant lower in Dex group (3.4 vs. 10.1%)No difference in ICU and hospital LOS and
mortality
-
A small single-center study,
-
Subjective delirium assessment
Huet Q, et al., 2024 [75]Double-blind RCT333 postsurgical cardiac patients, >65 years oldOvernight Dex infusion vs. placebo
-
CAM-ICU,
-
LSEQ
The incidence of delirium was not significant different between the two groups (12.6% Dex vs. 12.4% placebo) Dex group had significantly more hypotensive events (7.3% vs. 0.6%)
-
Subjective delirium assessment
-
Underestimated delirium incidence
Pereira J, et al., 2020 [76]Systematic Review of 6 RCTs and 2 retrospective cohorts 1249 ICU patients, aged > 60 yearsDex vs. propofol
-
CAM-ICU
-
CAM
Lower delirium riskNo reduced in
- ICU LOS,
- hospital LOS or
- MV duration
- No difference in bradycardia or hypotension
-
High heterogeneity of included studies and small sample size
-
High risk of type I and II error
Lin C, et al., 2021 [77]Systematic Review of 21 studiesElderly surgical ICU patients with deliriumDex vs. other sedatives
-
Dex reduced delirium incidents in non-cardiac surgical patients,
- No difference in delirium frequency among cardiac surgical patients
-
Decrease mortality,
-
Shortened ICU LOS,
- Shortened hospital LOS
- Increased bradycardia
-
Moderate heterogeneity in outcome and interventions
2.4 Comparative efficacy
Carrasco G, et al., 2016 [78]Single-center non-randomized study132 non-intubated patients in ICU with agitated deliriumDex vs. Haloperidol
-
Dex achieved higher levels sedation satisfactory
-
Reduced ICU LOS
-
Reduced the total ICU cost
Haloperidol associated with
-
more cases of oversedation
-
more case of QT prolongation
-
single-center study non-RCT
Shokri H, et al., 2020 [79]Prospective observation RCT286 cardiac surgery patients >60 years oldi.v. 0.7–1.2 μg/kg/h Dex for 72 h vs. 0.5 μg/kg clonidine- RASS scoreDex achieved
-
lower risk of delirium
-
Shorter MV duration
-
ICU LOS
-
lower mortality rate
-
lower morphine consumption
-
single-center study
Abbreviations: BDNF: brain-derived neurotrophic factor, CAM-ICU: Confusion Assessment Method for ICU, Dex: dexmedetomidine, h: h., HR: heart rate, ICU: intensive care unit, LOS: length of stay, LSEQ: Leeds Sleep Evaluation Questionnaire, MAAS score: Motor Activity Assessment Scale, MAP: Mean Arterial Pressure, MCAS score: Minnesota Cognitive Acuity Screen score, MMSE: Mini-Mental State Examination, MV: mechanical ventilation, NSE: neuron-specific enolase, PCIA: patient-controlled intravenous analgesia, QALYs: quality-adjusted life years, RCT: randomized controlled trial, S100B: S100 calcium binding protein, SBT: spontaneous breathing trial, UK: United Kingdom.5.4.5. Anxiolytic Effect.
Table 3. Clinical indications for dexmedetomidine in ICU patients.
Table 3. Clinical indications for dexmedetomidine in ICU patients.
Study DesignMaterialComparison Main OutcomesStudy Limitations
3.1. Sleep quality improvement in ICU
Wu XH, et al., 2016 [95]RCT61 non-cardiac postsurgical, non-intubated, ICU patients > 65 years oldDex 0.1 μg/kg/h vs. placebo for 15 h postsurgically
-
Increased the percentage of stage N2 sleep (43.5 vs. 15.8%),
-
Prolonged the total sleep time,
-
Decreased the percentage of stage N1 sleep,
-
Increased the sleep efficiency,
-
Improved sleep quality
-
Increased the hypotension incidence
-
Single-center pilot RCT
-
Limited sample size
Lu W, et al., 2017 [94]Observational Study20 non-intubated, non-MV, ICU, postsurgical patients11 in Dex group vs. 9 in no-sedation group
-
Sleep efficiency and
-
Sleep time of patients in the sedation group was significantly higher during the night,
-
No difference in heart and respiratory rates,
-
No respiratory depression
-
Single-center observational study
-
Limited sample size
Georgopoulos D, et al., 2021 [91]Retrospective study23 MV, ICU patients Dex vs. propofol vs. no sedation
-
In non-sedated patients, sleep quality was poor, with frequent wake intrusions and little stable sleep,
-
Light sedation with propofol or Dex resulted in a shift in sleep architecture toward normal.
-
Small single-center retrospective study
Sun YM, et al., 2022 [96]RCT80 non-cardiac, postsurgical, MV, patientsDex 0.1–0.2 μg/kg/h vs. placebo for >72 h
-
Better sleep quality (RCSQ score: 61 vs. 52),
-
Longer total sleep time,
-
Higher sleep efficiency,
-
Lower percentage of stage N1 sleep,
-
Higher percentage of stage N3 sleep,
-
Lower arousal index.
-
Small sample size
-
15% sleep monitoring failure
Qu JZ, et al., 2023 [97]Single-center RCT (The MINDDS trial) 394 cardiac, postsurgical patients > 60 years oldA short nighttime dose of iv Dex (1 μg/kg in 40 min)
-
Non-significant delirium incidence reduction in Dex group (8.8 vs. 14.1%)
-
Evaluation of delirium only the first postoperative day
-
The trial stopped early because of the COVID-19 pandemic
Zhang ZF, et al., 2023 [98]RCT118 non-cardiac, postsurgical, non-intubated patients > 65 years oldDex 0.02 μg/kg/h vs. placebo + opioid analgesia for up to 3 days
-
Increased the percentage of stage N2 sleep,
-
Prolonged total sleep time,
-
Increased sleep efficiency,
-
Decrease percentage of N1 sleep,
-
Lowered sleep fragmentation index
-
Single-center RCT
-
They only monitored sleep quality the night of surgery.
Sun PS, et al., 2024 [99]RCT123 non-cardiac, postsurgical, non-intubated patients, with OSADex 0.02 μg/kg/h vs. placebo + opioid analgesia
-
Increased the percentage of stage N2 sleep,
-
Decreased percentage of N1 sleep,
-
Slightly improved sleep quality
-
Single-center RCT
-
Small sample size
Wang L, et al., 2024 [100]Systematic review of 29 trials5610 non-cardiac, postsurgical patientsPerioperative Dex vs. placebo
-
Improved sleep quality,
-
Increased sleep quality,
-
Low and very low quality of evidence
3.2. Post-Intensive Care Syndrome
Zi J, et al., 2020 [90]Single-center RCT196 patients underwent off-pump coronary artery bypassDex vs. propofol perioperatively
-
Lower incidence of atrial fibrillation,
-
Lower anxiety level (51.6 vs. 67.2%)
-
Did not evaluate the effects of other related sedatives or analgesics
-
Small sample size
Dong CH, et al., 2021 [101]Single-center RCT508 patients underwent off-pump coronary artery bypassProphylactic nocturnal Dex vs. placebo
-
Lower incidence of PICS (21.5 vs. 31.1%),
-
cognitive impairment (3,98 vs. 6,61%),
-
disability (2,79 vs. 5,06%),
-
psychological impairment (18.7 vs. 26.8%),
-
6-months mortality (1.2 vs. 1.6%)
-
Lack of standard PICS definition
-
Additional sedatives were used
3.3. Alcohol withdrawal syndrome
VanderWeide LA, et al., 2016 [102]Retrospective cohort study42 AWS ICU patientsBenzodiazepine alone vs. benzodiazepine + Dex
-
Reduced benzodiazepine’s requirement,
-
More cases of bradycardia in Dex group,
-
No difference in AWS control.
-
Small single = center retrospective study
-
The analysis had bias against Dex
Beg M, et al., 2016 [103]Retrospective cohort study77 AWS ICU patientsBenzodiazepine alone vs. benzodiazepine + Dex
-
Dex group improved CIWA score,
-
Dex group increased ICU LOS
-
Small single-center retrospective study
-
The initiation, titration, and discontinuation of the drugs were not control
-
Additional sedatives were used
Yavarovich ER, et al. [104]Multicenter retrospective cohort study (8 ICUs)438 AWS ICU patientsBenzodiazepine alone vs. benzodiazepine + Dex
-
Dex group had higher CIWA,
-
Longer ICU LOS,
-
Higher rate of delirium.
-
Retrospective study
-
Lack of standardization of Dex prescribing and additional adjunctive medications
Collier TE, et al., 2022 [105]Single-center retrospective cohort study (The EvADE study)147 AWS ICU patientsBenzodiazepine alone vs. benzodiazepine + Dex
-
No significant change in CIWA-Ar score in Dex group: (3.8 vs. 5.4),
-
Increased benzodiazepine requirements in Dex group,
-
Prolonged ICU LOS,
-
Higher risk of new seizures onset and
-
Intubation.
-
Retrospective study
-
Lack of standardization of Dex prescribing and additional adjunctive medications
-
Inability to access CIWA-Ar in intubated patients
Ware LR, et al., 2023 [106]Single-center retrospective cohort study197 AWS ICU patientsPhenobarbital alone vs. phenobarbital + Dex
-
No-Dex group had reduced ICU LOS (47.5 vs. 97.2 h),
-
Dex group had higher rates of total delirium days (9208 vs. 143 days),
-
Dex group had longer MV duration.
-
Retrospective cohort
Wong A, et al., 2015 [107]Review of 13 studies Dex as an adjunctive agent
-
Dex decreased benzodiazepine’s requirement,
-
Better controlled hypertension and tachycardia,
-
No difference in seizures control
-
Limited number of small trials
-
Low quality of studies (8 case reports/series and 5 RCTs)
-
Heterogeneity in AWS assessment and treatment
Woods D, et al., 2015 [108]Systematic review of 4 studies55 AWS ICU patientsDex as adjunctive therapy + standard of care vs. benzodiazepine
-
Dex decreased CIWA score,
-
Decreased delirium more effectively.
-
Limited number of small trials (3 retrospective and 1 prospective)
-
Low-quality studies
-
Heterogeneity in AWS assessment
Polintan ETT, et al., 2023 [109]Systematic review of 12 studiesAWS ICU patientsDex as adjunctive therapy + standard of care vs. benzodiazepine
-
Dex showed no significant difference for ICU LOS,
-
AWS control,
-
Bradycardia, or
-
Hypotension.
-
Limited number of small trials (3 retrospective and 1 prospective)
-
Low-quality studies
-
Heterogeneity in AWS assessment
Fiore M, et al., 2024 [110]Systematic review of 9 studies (RCTs and non-RCTs)AWS ICU patientsDex as adjunctive therapy + standard of care vs. benzodiazepine
-
Dex as adjunctive therapy is not more effective than standard of care in reducing intubation,
-
Higher risk of bradycardia,
-
Same risk of hypotension.
-
Limited number of small trials
-
Low-quality studies
-
Heterogeneity in AWS treatment and assessment
Abbreviations: AWS: alcohol withdrawal syndrome, CIWA: Clinical Institute Withdrawal Assessment Score, CIWA-Ar: Clinical Institute Withdrawal Assessment Score for alcohol, Dex: dexmedetomidine, h: h., ICU: intensive care unit, LOS: length of stay, MV: mechanical ventilation, OSA: obstructive sleep apnea, PICS: post-intensive care syndrome, RCT: randomized controlled trial, RCSQ: Richards–Campbell Sleep Questionnaire, SBT: spontaneous breathing trial, UK: United Kingdom.
Table 4. Neuroprotective effects of dexmedetomidine in ICU patients.
Table 4. Neuroprotective effects of dexmedetomidine in ICU patients.
Study DesignMaterialIntervention OutcomesStudy Limitations
Wu J, et al., 2018 [118]Animal study76 male C57 BL/6 mice (TBI model)Different doses of Dex or placebo in addition, 1 and 12 h after TBI
-
Reduced loss of cortical tissue,
-
Reduced cell death in the cortex and hippocampus
-
Prevented axonal degeneration
-
Protected synapses from elimination
Mice treated with Dex were hypothermic
Li F, et al., 2019 [85]Animal studyMale Sprague-Dawley rats (TBI model)25 μg/kg Dex 30 min after TBI vs. placebo
-
Lower NSS scores
-
Reduced apoptotic factor expression
-
Decreased proinflammatory cytokine expression
-
Increased Nrf2 protein levels
-
Induced Nrf2 downstream factor expression
-
Improved TBI induced Bax upregulation and Bcl-2 downregulation
Not refereed
Feng X, et al., 2021 [119]Animal studyC57 BL/6 mice (TBI model)30 μg/kg Dex vs. normal saline
-
Increased the survival rate and neurological score,
-
Increased neuron survival,
-
decreased the expression of the LC3, Beclin-1, and NF-κB proteins, and the cytokines IL-1β, IL-6, and TNF-α
Not refereed
Huang GR, et al., 2021 [120]Animal studyRat (BI model)Dex vs. normal saline
-
Inhibit inflammation and
-
Attenuate early neuronal injury through activation on TLR4/NF-κB pathway.
Not refereed
Liu SY, et al. A 2024 [121]Retrospective study (Premier dataset)19,751 ICU patients with TBIDex vs. other sedatives
-
Reduced hospital mortality
-
Increased risk for liberation from MV
-
Reduced LOS in ICU
-
Retrospective analysis of MIMIC IV database
-
Relatively small sample size
-
Low statistical power
Chen S, et al., 2025 [122]Retrospective study (MIMIC-IV database)646 ICU patients with ischemic strokeDex vs. other sedatives
-
Reduced ICU mortality
-
Retrospective analysis of MIMIC IV database
Yang Y, et al., 2025 [123]Retrospective study (MIMIC-IV database)2816 ICU patients with ischemic stroke Dex vs. other sedatives
-
Reduced 28-day mortality (27.8 vs. 36.6%)
-
Reduced 180-day mortality, among discharged patients
-
Retrospective analysis of MIMIC IV database
-
Heterogeneity regarding severity of ischemic stroke
Liu Y, et al., 2025 [124]Retrospective study (MIMIC-IV database)527 patients with aSAHDex vs. midazolam or propofol
-
Reduced in-hospital mortality
-
Retrospective analysis of MIMIC IV database
-
Limited statistical power due to relatively small sample size
Lu S, et al., 2025 [125]Single-center RCT60 TBI patientsDex vs. propofol
-
Dex reduced MV duration and ICU LOS
-
Dex elevated SjvO2 and reduced CERO2
-
Dex reduced IL-6 levels but not IL-1β
-
Single-center
-
Small sample size
Jiang L, et al., 2017 [21]Systematic review and meta-analysis19 RCTs including 879 patientsDex during operation
-
Reduces the release of inflammatory mediators and neuroendocrine hormones.
-
Maintains intracranial homoeostasis
-
Alleviates ischemic brain injury
-
exerts an effect on brain protection.
-
Abbreviations: aSAH: aneurysmal subarachnoid hemorrhage, BI: brain injury, Dex: dexmedetomidine, ICU: intensive care unit, IL-6: interleukin-6, INF-γ: interferon-γ, I/R: ischemia/reperfusion, LOS: length of stay, MV: mechanical ventilation, NSS score: Neurological Severity Score System, RCT: randomized controlled trial, TBI: traumatic brain injury, TNF-α: tumor necrosis factor-α.
Table 5. Anti-inflammatory effects of dexmedetomidine in ICU patients.
Table 5. Anti-inflammatory effects of dexmedetomidine in ICU patients.
Study DesignMaterialComparisonOutcomesStudy Limitations
5.1. Cardio-protective properties
Bulow NMH et al., 2016 [112]RCT12 CPB patients in Dex group vs. 11 CPB controlsDex perioperatively in CPB patients plus other sedatives vs. only other sedatives
-
Reduced circulating IL-1, IL-6, TNF-α, and INF-γ levels
-
Small sample size
-
The dose of Dex used was low
Liu X, et al., 2016 [127]RCTE60 elective valve and CAP surgery patients0.2–1.5 μg/kg/h Dex or 5–50 μg/kg/min propofol
-
Dex group had significantly greater changes in perfused small-vessel density and the De Backer score
-
Possible interaction with other used drugs
-
Single-center RCT
-
Small sample size
Yuan B, et al., 2024 [128]RCT52 CPB patients vs. 52 controls0.5 μg/kg Dex before anesthesia induction + 0.5 μg/kg/h vs. 0.125 mL/kg NaCl 0.9% before aortic occlusion
-
Serum cTnI, CK-MB,
-
MDA, and
-
TNF-α were lower in the Dex group
-
Dex group had fewer interventions on the mitral valve
Zhang GR, et al., 2021 [129]Systematic Review of 17 studies866 CPB patientsDex vs. other sedatives
-
Reduced serum CK-MB and cTn-I
-
Shorted ICU LOS
-
Myocardial protection from I/R injury
-
Small sample size
-
Notable heterogeneity of the included studies
5.2. Renoprotective properties
Cakir M, et al., 2015 [130]Animal studyGroup 1: 10 I/R injury rats Group 2: 10 controls
Group 3: I/R + 10 μg/kg Dex
Group 4: I/R + 100 μg/kg Dex
10 μg/kg Dex vs. 100 μg/kg Dex
-
Lower levels of MDA, CAT, and GHH in Dex groups
-
Increased SOD activity in Dex group
-
In Dex100 group, the elevated BUN levels were significantly lower than in I/R group
-
In DEX groups, the elevated Cr levels were significantly lower than in I/R group
-
Lower histological injury scores in Dex groups, but not between different doses of Dex
-
It is an experimental laboratory animal study
-
Only 2 doses of Dex were compared
-
The efficacy of Dex was evaluated only in kidney I/R injury
Tan F, et al., 2015 [131]Animal studyGroup 1: 8 sepsis model rats
Group 2: 8 controls
Group 3: 8 Dex + sepsis model
Group 4: 8 Dex + yohimbine + sepsis model
10 μg/kg Dex before sepsis vs. a2-adrenergic receptor agonist (yohimbine)
-
When rats were pretreated with Dex, sepsis induced AKI was decreased significantly
-
Exposure of yohimbine eliminated this reduction
-
Sepsis AKI is a multifactorial AKI
Zhai M, et al., 2017 [113]Single-center RCT72 valve replacement CPB patients0.6 μg/kg Dex before anesthesia induction + 0.2 μg/kg/h vs. NaCl 0.9%
-
The levels of BUN and Cr and the NGAL significantly lower in Dex group,
-
the intraoperative urine output was significantly increased and
-
the postoperative incidence of AKI was significantly lower in Dex group.
-
AKI was defined according to RIFLE criteria
-
Single-center small RCT
Liu J, et al., 2020 [132]RCT200 MV septic ICU patientsDex vs. propofol
-
Less AKI
-
Less CRRT duration
-
Less LOS ICU
-
No significant difference in MV duration
-
Single-center RCT
-
AKI was defined according to RIFLE criteria
Liu Y, et al., 2018 [133]Systematic review of 10 studies1575 postoperative cardiac surgery ICU patientsDex vs. other sedatives
-
Significantly reduced postoperative AKI
-
No difference in mortality
-
No difference in MV duration
-
No difference in ICU LOS and
-
Hospital LOS
-
Relatively small number of RCTs
-
Small sample size
-
Great heterogeneity regarding study population or comparison (Dex vs. placebo, or Dex vs. other sedatives)
-
Different adjuvant sedatives and analgesics
Zhao C, et al., 2024 [134]Systematic review of 15 RCTs2907 postoperative cardiac surgical patientsDex vs. other sedatives
-
Dex reduced the incidence of postoperative AKI
-
No difference in mortality,
-
No difference in MV duration
-
No difference in ICU LOS
-
No difference in hospital LOS
-
Moderate study heterogeneity
-
4 different AKI definitions
-
Different RRT initiation criteria
Zhao J, et al., 2024 [135]Systematic review of 25 trials3997 postsurgical patientsDex vs. controls
-
Dex reduced the incidence of postoperative AKI
-
Dex reduced AKI incidence only in cardiac surgical patients
-
Dex reduced ICU and hospital LOS
-
Dex reduced postoperative delirium occurrence
- 3 studies focused on pediatric patients
- Notably study heterogeneity
- Small sample size of non-cardiac surgical patients
5.3. Dexmedetomidine in sepsis
Al-Regal ARE, et al., 2024 [136]RCT
(The DecatSepsis)
90 Septic ICU patientsDex infusion for 48 h to maintain the HR:60–90/min
-
Underpowered to detect significant reduction in in-hospital mortality and dose of noradrenaline
-
Both mechanically and non-MV ICU patients
-
Small sample size
Iten M, et al., 2025 [137]Single-Center RCT70 Septic, MV, ICU patients0.1–1.4 μg/kg/h Dex vs. propofol/midazolam group
-
No difference in S100-β levels at 48 h or upon ICU discharge
-
This is a pilot RCT
-
Small sample size
-
The duration of Dex sedation was max 7 days
-
Added propofol or midazolam in Dex group
Patidar AK, et al., 2025 [138]Single-center RCT54 septic ICU patients5 mcg/mL Dex vs. 10 mg/mL propofol, aiming RASS: –2 to 0
-
Dex group showed higher mean regional cerebral oxygen saturation
-
No difference in MV and delirium duration
-
No difference in ICU LOS
-
Single-center RCT
-
Small sample size
Dargent A, et al., 2025 [58]The ADRESS Multicenter RCT (5 French ICUs)Septic shock, MV, ICU patientsDex vs. placebo in patients with refractory septic shock
-
Higher mortality in Dex group
-
The trial stopped earlier
-
Early study elimination
Chen P, et al., 2020 [139]Systematic Review of 4 studies349 MV septic patientsDex vs. other sedatives
-
No difference in duration of MV
-
Significant difference in ventilator-free days and
-
28-day mortality
-
Limited sample size
-
Notable study heterogeneity
Wang C, et al., 2021 [140]Systematic Review of 9 studies1134 septic ICU patientsDex vs. other sedatives
-
No difference in mortality, LOS ICU, delirium
-
Significantly reduced MV duration and inflammatory response
-
Limited sample size
-
Notable study heterogeneity
Zhang T, et al., 2022 [141]Systematic Review of 19 RCTs1929 septic patientsDex vs. other sedatives
-
Decreased mortality
-
Decreased inflammatory response
-
Increased risk of arrhythmias
-
Not significantly reduce ICU LOS, duration of MV, incidence of delirium, Cr, and ALT
-
Relatively small number of RCTs
-
Limited sample size
-
Notable study heterogeneity
Abbreviations: AKI: Acute Kidney Injury, ALT: Alanine Aminotransferase, BUN: blood urea nitrogen, CAT activity: catalase activity, CRP: C-reactive protein, Cr: creatinine, CRRT: continues renal replacement therapy, Dex: dexmedetomidine, CK-MB: creatinine-kinase-MB, CPB: cardiopulmonary bypass, Cr: creatinine, cTnI: cardiac troponin I, HR: heart rate, ICU: intensive care unit, IL-6: interleukin-6, INF-γ: interferon-γ, I/R: ischemia/reperfusion, GSH: glutathione, LOS: length of stay, MDA: malondialdehyde, MV: mechanical ventilation, NGAL: Neutrophil Gelatinase-Associated Lipocalin, RCT: randomized controlled trial, RRT: renal replacement therapy, SOD activity: superoxidase dismutase, TNF-α: tumor necrosis factor-α.5.4.9. Neuroprotective Effects.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sertaridou, E.N.; Fountoulaki, M.; Jha, A.; Papaioannou, V.E.; Alexopoulou, C. Dexmedetomidine’s Role in Adult ICU After 20 Years of Experience—A Narrative Review. Healthcare 2025, 13, 2882. https://doi.org/10.3390/healthcare13222882

AMA Style

Sertaridou EN, Fountoulaki M, Jha A, Papaioannou VE, Alexopoulou C. Dexmedetomidine’s Role in Adult ICU After 20 Years of Experience—A Narrative Review. Healthcare. 2025; 13(22):2882. https://doi.org/10.3390/healthcare13222882

Chicago/Turabian Style

Sertaridou, Eleni N., Maria Fountoulaki, Abhishek Jha, Vasilios E. Papaioannou, and Christina Alexopoulou. 2025. "Dexmedetomidine’s Role in Adult ICU After 20 Years of Experience—A Narrative Review" Healthcare 13, no. 22: 2882. https://doi.org/10.3390/healthcare13222882

APA Style

Sertaridou, E. N., Fountoulaki, M., Jha, A., Papaioannou, V. E., & Alexopoulou, C. (2025). Dexmedetomidine’s Role in Adult ICU After 20 Years of Experience—A Narrative Review. Healthcare, 13(22), 2882. https://doi.org/10.3390/healthcare13222882

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop