The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×
Influential PublicationsFull Access

A Review of Pharmacologic Management and Prevention Strategies for Delirium in the Intensive Care Unit

Abstract

Background

The prevalence of delirium has been estimated at anywhere between 10% and 30% in general medical patients and in upwards of 80% in patients who are admitted to an intensive care unit (ICU). Given the high prevalence of delirium in the ICU population, it should not be surprising that a large percentage of psychiatric consults arise from this setting. While the mainstay of pharmacologic management of delirium centers on neuroleptic medications, such as haloperidol, recent studies using alternate agents have shown varying levels of promise.

Objective

Our purpose is to outline the major prospective studies looking at the efficacy of pharmacologic management and prevention strategies for delirium exclusively in adult ICU patients. Both conventional and novel pharmacotherapeutic interventions are discussed.

Method

Articles were obtained using the MEDLINE/PUBMED database looking specifically at pharmacologic interventions for delirium in the intensive care unit. A search was performed using the key words-“delirium,” “intensive care unit,” “treatment,” and “prophylaxis.” The authors limited their search to prospective studies, specifically randomized trials (both placebo-controlled and non-controlled) in the adult ICU population, and eliminated retrospective and observational studies. Relevant citations from the previously mentioned articles were also included in the review.

Conclusion

There is a plethora of studies on pharmacologic management strategies in general medical patients with delirium. Findings from these studies are often extrapolated to the ICU population; however, when looking at studies limited to ICU patients with delirium, there are far fewer credible prospective studies.

(Reprinted with permission from Psychosomatics, 2012; 53:203–211)

Delirium prevalence is estimated at 10%–30% in general medical population and as high as 80% in the ICU population. Given these statistics, it is not surprising that a large percentage, if not a majority, of delirium consults arise from the critical care setting. Definitive treatment of delirium in any setting centers on the correction of underlying physiologic and iatrogenic insults; however, the pathogenesis of delirium is often multifactorial and definitive precipitants are not always known. As a result, psychiatric consultants are often asked to suggest pharmacologic management strategies for the delirious patient while simultaneously addressing predisposing and precipitating risk factors. The higher incidence of morbidity (including injury to self and staff as well as long-term cognitive impairment) and mortality (3-fold increase in 6-month mortality) in ICU patients with delirium underscores the importance of early identification and treatment.1 With delirium often comes the added burden of prolonged mechanical ventilation and extended hospitalization. This leads to significantly increased risks for nosocomial infection and an increase in overall healthcare costs.

To underscore the importance of addressing delirium in the ICU, a recent study by Heymann and colleagues showed that in a cohort of 204 ICU patients, treating delirium within the first 24 hours of onset significantly reduced mortality (3-fold higher rate of mortality in the delayed treatment group vs. immediate treatment group).2 Studies such as these highlight the necessity for vigilance in identifying and treating delirium in the ICU setting and promote moving away from the archaic notion that delirium is a transient, benign process, with no long-term sequelae.

The purpose of this review is to familiarize the reader with the major prospective clinical trials that have looked at pharmacologic interventions for delirium in the adult ICU population (Table 1). Studies in non-ICU patients were excluded in an attempt to minimize confounding variables that are inherent when generalizing findings between very different study populations. Furthermore, this review serves as an update of the current evidence and rationale behind using various pharmacologic agents for the management of delirium in the ICU.

Table 1. Major Randomized Trials of Pharmacotherapeutic Interventions for Delirium in the ICU Population
StudyCharacteristicsPrimary Outcome MeasureResults
I. Antipsychotics



 Olanzapine vs. haloperidol (Skrobik et al14)
73 Medical/surgical ICU patients diagnosed with delirium randomized to olanzapine (n = 28) or haloperidol (n = 45)
Severity of delirium and benzodiazepine use over 5 days
No significant difference in delirium index or benzodiazepine use
 MIND Trial (Girard et al15)
101 Medical/surgical ICU patients randomized to haloperidol (n = 35), ziprasidone (n = 30), or placebo (n = 36) for up to 14 days
Days alive without delirium or coma
No significant differences between groups
Neither haloperidol nor ziprasidone were better than placebo in treating delirium
 Quetiapine (Devlin et al17)
36 Medical/surgical ICU patients diagnosed with delirium randomized to quetiapine (N = 18) or placebo (n = 18) until first resolution of delirium, for up to 10 days, or until ICU discharge, whichever came first
Time to first resolution of delirium
Quetiapine associated with a shorter time to first resolution of delirium
 Risperidone (Prakanrattana et al36)
126 Cardiac surgery patients (ICU) randomized to a single postoperative dose of risperidone (n = 63) or placebo (n = 63)
Incidence of post-operative delirium
Incidence of postop delirium lower in the risperidone group
II. Cholinesterase Inhibitors



 Rivastigmine (van Eijk et al18)
104 ICU patients diagnosed with delirium randomized to rivastigmine (n = 54) or placebo (n = 50) as an adjunct to haloperidol
Duration of delirium
Study prematurely stopped due to increased mortality in the rivastigmine group Median duration of delirium was longer in the rivastigmine group
 Rivastigmine (Gamberini et al38)
120 Cardiac surgery patients (ICU) randomized to rivastigmine (n = 59) or placebo (n = 61) for 6 postoperative days
Incidence of postoperative delirium
Incidence of postop delirium higher in the rivastigmine group
III. NMDA Antagonists



 Ketamine (Hudetz et al37)
58 Cardiac surgery patients (ICU) randomized to ketamine (n = 29) or placebo (n = 29) during anesthetic induction
Incidence of postoperative delirium
Incidence of postop delirium lower in the ketamine group
IV. α-2 Agonists



 Clonidine (Rubino et al40)
30 Surgical ICU patients who underwent acute repair of thoracic aortic dissection randomized to clonidine (n = 15) or placebo (n = 15) infusion during weaning from mechanical ventilation
Incidence and severity of postoperative delirium
No statistically significant difference in incidence of delirium between groups. severity of delirium lower in the clonidine group
 Dexmedetomidine vs. Haloperidol (Reade et al27)
20 Medical/surgical ICU patients who were difficult to extubate secondary to superimposed delirium randomized to a continuous infusion of dexmedetomidine (n = 10) or haloperidol (n = 10)
Time to extubation
Dexmedetomidine group had a significantly shorter time to extubation
Dexmedetomidine group had an increased proportion of time spent with minimal or no delirium symptoms
 Dexmedetomidine (Maldonado et al31)
118 Cardiac surgery patients (ICU) randomized to dexmedetomidine (n = 40), propofol (n = 38), or midazolam (n = 40) for postoperative sedation
Incidence of postoperative delirium
Incidence of delirium was significantly lower in the dexmedetomidine group
Incidence of delirium was markedly higher in the propofol and midazolam groups
 DEXCOM Study (Shehabi et al39)
306 Cardiac surgery patients (ICU) randomized to dexmedetomidine (n = 154) or morphine (n = 152) for postoperative sedation/analgesia
Incidence of postoperative delirium
Incidence of postop delirium was statistically similar between groups
Duration of delirium was shorter in the dexmedetomidine group
 MENDS Trial (Pandharipande et al24)
106 Medical/surgical ICU patients randomized to dexmedetomidine (n = 54) or lorazepam (n = 52) for sedation
Days alive without delirium or coma
Dexmedetomidine group had significantly more days alive without delirium or coma
Dexmedetomidine group had significantly more coma-free days but not delirium-free days
Table 1. Major Randomized Trials of Pharmacotherapeutic Interventions for Delirium in the ICU Population
Enlarge table

Antipsychotics

Antipsychotics are currently the mainstay of pharmacotherapy for the management of delirium in most settings. They are thought to exert their influence primarily via dopaminergic blockade as it relates to the dopamine excess/acetylcholine deficiency hypothesis of delirium pathophysiology.3 There is also some evidence that antipsychotics, in particular haloperidol, may also impart a neuroprotective effect by proposed σ-1 receptor antagonism, which helps attenuate oxidative stress that then leads to neuronal damage in delirium.4 Furthermore, there are studies showing that antipsychotics may also function as immunomodulators via indirect antagonism of IL-1.5 Even in sedative-hypnotic and alcohol withdrawal delirium, where GABA-receptor agonists are first-line agents, antipsychotics have been shown to be useful adjuncts.

To date, there are few large-scale studies of antipsychotics for delirium exclusively in the ICU population. Per Society of Critical Care Medicine guidelines, haloperidol remains the first-line pharmacotherapeutic agent for the management of delirium in the intensive care setting.6 Its use in managing delirium in the ICU is supported by a long track record with multiple observational studies and case series that have shown both efficacy and safety even at high doses administered via intravenous (IV) infusion.7,8 The use of haloperidol for delirium remains off-label, however, and relatively few placebo-controlled trials exist in the ICU population. Furthermore, though IV haloperidol is thought to carry less of a risk for extrapyramidal symptoms (EPS), its use warrants caution, given an increased risk for QTc prolongation and polymorphic ventricular tachyarrhythmias, such as torsades de pointes.9,10

With the advent of second generation antipsychotics, we now have more options in patients who are sensitive to dopamine antagonism (specifically D2 receptor antagonism). Studies thus far have not shown atypical antipsychotics to be superior in efficacy to haloperidol for the management of delirium in the ICU setting; however, there are multiple studies attesting to the equal efficacy of atypical antipsychotics and haloperidol.1113 With the added benefit of multiple dosage forms and routes of administration, however, haloperidol will likely remain a first-line agent in managing delirium in the ICU.

While studies comparing atypical antipsychotics and haloperidol have mostly been done in non-ICU populations, a recent study by Skrobik and colleagues looked at a cohort of 73 ICU patients who developed delirium.14 The study sample was subsequently randomized to receive either haloperidol or olanzapine. Comparable to non-ICU studies, findings showed that there were no significant differences in delirium index, benzodiazepine dose administered over time, or clinical improvement between the two study groups. As expected, the olanzapine group had a lower incidence of EPS. Limitations of this study included a lack of blinding, no placebo arm, as well as a relatively small sample size. In addition, both groups utilized additional PRN haloperidol, which may have confounded results.

To date, there are only two major randomized, placebo-controlled trials of antipsychotics for delirium exclusively in the ICU population. The MIND (Modifying the Incidence of Delirium) trial looked at 101 adult mechanically-ventilated medical and surgical ICU patients who were randomly assigned to receive haloperidol, ziprasidone, or placebo every 6 hours for up to 14 days.15 Delirium was then assessed for up to 21 days. Findings showed that there were no differences between groups in the duration of delirium or days alive without delirium. Interestingly, this study also showed that neither haloperidol nor ziprasidone was significantly better than placebo in treating delirium in this population. In addition, mortality, length of hospitalization, and ventilator-free days were all similar between groups. One must take these results with caution, however. There were major study limitations in the MIND trial including: small sample size, inclusion of non-delirious patients in the study group, exposure to open-label PRN haloperidol for breakthrough agitation in each study arm, and multiple rigid exclusion criteria.15,16

The second study, by Devlin and colleagues, looked at 36 ICU patients with delirium who were assigned to receive either quetiapine or placebo in addition to PRN haloperidol.17 Study findings showed that quetiapine was associated with a shorter “time to first resolution” of delirium (1 day vs. 4.5 days in the placebo arm) and reduced duration of delirium (36 hours vs. 120 hours, respectively). In addition, the quetiapine group required less PRN haloperidol than the placebo group. Mortality and length of ICU stay were similar between study groups. Again, major limitations included small sample size and multiple exclusion criteria. In addition, when measuring “time to first resolution” of delirium, the investigators may have inadvertently included patients that were in a lucid interval yet still delirious.

Generalizations cannot be made based on the findings of the study by Devlin and colleagues or on those of the MIND trial. Both studies were underpowered and excluded patients with multiple co-morbidities including: patients with primary neurologic conditions and those exposed to antipsychotics prior to the studies, in addition to patients on QTc-interval prolonging medications and those with various cardiac anomalies. Extrapolation of these findings to most ICU populations would be premature at this point. Furthermore, there remains an abundance of anecdotal data that supports the routine usage of antipsychotics for the management of delirium. Additional large-scale prospective studies are still needed to solidify practice guidelines in the ICU population. Both of the above studies did, however, show that prospective, placebo-controlled trials of antipsychotics for delirium in the ICU population are both possible and safe.

Cholinesterase Inhibitors

Cholinesterase inhibitors have garnered recent attention as possible pharmacotherapeutic options in treating delirium. The reasoning behind their use relates to the central cholinergic deficiency hypothesis of delirium.3 Cholinesterase inhibitors function primarily by inhibiting enzymatic breakdown of acetylcholine. Observational studies and anecdotal reports using physostigmine, a prototypical cholinesterase inhibitor, in the reversal of anticholinergicinduced delirium were promising and prompted further investigation into the potential utility of using newer agents (such as donepezil, galantamine, and rivastigmine). As with other agents, there is a relative lack of data in the ICU population. The results are currently mixed, with successful reports of cholinesterase inhibitors treating delirium in case studies and smaller prospective studies offset by relatively poor outcomes in larger prospective trials.

For the most part, recent studies of cholinesterase inhibitors in ICU patients who have been diagnosed with delirium have focused on rivastigmine. The single largest prospective study to date was a multi-center, randomized, placebo-controlled trial by van Eijk and colleagues in the Netherlands.18 This study initially planned to look at 440 ICU patients diagnosed with delirium who were randomized to receive an increasing dose of either rivastigmine or placebo as an adjunct to haloperidol for the management of delirium. The study was prematurely stopped, after the inclusion of 104 patients, due to an increase in mortality in the rivastigmine group (n = 12, 22%) compared with placebo (n = 4, 8%). Furthermore, the median duration of delirium was longer in the rivastigmine group (5 days) compared with placebo (3 days). In addition, the patients in the rivastigmine group had a higher severity of delirium, stayed longer in the ICU, and received higher cumulative doses of haloperidol, lorazepam, and propofol. Overall, the van Eijk study did not support the widespread use of cholinesterase inhibitors in treating delirium in the ICU. If anything, this study suggested that these agents should be used with the utmost caution in the critically ill, though further studies are still needed to make any definitive conclusions.

α-2 Agonists

Though most often used in sedation and analgesia protocols in the ICU, α-2 agonists have recently been shown to reduce the incidence of, and treat, delirium in the critically ill. These agents have the added benefit of minimizing respiratory suppression and facilitating the maintenance of a low heart rate, thereby minimizing hemodynamic fluctuations and reducing energy/demand expenditure that may contribute to global cerebral insult.19 These agents are also thought to be neuroprotective by inhibiting the release and production of neurotoxic glutamate.20

Recent studies investigating the efficacy in treating delirium with α-2 agonists have focused on the novel agent, dexmedetomidine. This is a centrally-acting α-2 agonist that is eight times more selective for α-2 adrenoreceptors than clonidine.21 Dexmedetomidine works both pre- and post-synaptically to decrease norepinephrine release and reduce sympathetic activity in the CNS.21 While initially approved for use in ICU patients for a duration of less than 24 hours, at least four recent clinical trials have shown that dexmedetomidine can be used safely for up to 30 days.2227 Though shown to have a lower incidence of rebound hypertension and tachycardia on abrupt discontinuation compared with clonidine, dexmedetomidine carries an increased risk for hypotension and bradycardia, especially with high-rate infusions.21

The proposed benefit of dexmedetomidine over standard agents for sedation/analgesia has centered on its lack of significant anticholinergic effects, promotion of sleep/wake cycle regulation, and reduced need for opioid analgesics (by as much as 40% in some studies) and GABA agonists.2833 These properties impart a unique profile to dexmedetomidine, thereby reducing the potential precipitating risk factors that play a role in delirium pathophysiology. Case in point, a recent study by Pandharipande and colleagues underscored the potential risks of benzodiazepine use in a cohort of 198 mechanically-ventilated ICU patients, showing that lorazepam use was an independent risk factor for progression to delirium in this at risk population.34 Findings showed that in addition to a substantial increase in the risk for developing delirium at low doses, the probability of progressing to delirium reached 100% after total daily lorazepam doses of ≥ 20 mg.34 Studies such as these underscore the importance of seeking out alternative agents for sedation and analgesia protocols.

There are numerous case studies examining the efficacy of using dexmedetomidine as both a primary and adjunctive therapy in sedation/analgesia protocols. Many of these studies examine the effectiveness of dexmedetomidine in treating symptoms attributable to delirium (such as agitation) as well as the incidence of delirium. Most prospective studies have evaluated the efficacy of using dexmedetomidine for sedation/analgesia protocols in mechanically-ventilated patients and looked at delirium incidence only as a secondary endpoint. There are few studies using dexmedetomidine as a primary agent in patients who were diagnosed with delirium at the onset on the study and even fewer have actually looked at the reduction in severity or resolution of delirium as primary endpoints.

The pivotal prospective study was performed by Reade and colleagues on 20 ICU patients exhibiting severe agitation presumed to be secondary to delirium at onset, then subsequently randomized to receive either dexmedetomidine or haloperidol infusions.27 Time to extubation was the primary outcome measure and delirium incidence and severity was a secondary measure. A total of 10 patients (five in each study arm) met criteria for delirium by the end of the study; however, the dexmedetomidine group had an increased proportion of time spent with minimal or no delirium symptoms as measured by the Intensive Care Delirium Screening Checklist (ICDSC), with the dexmedetomidine group spending 95.5% of the time with a ICDSC score of <4 and 61% of the time with a score <1 compared with the haloperidol group who spent 31.5% and 0% of the time with the same respective scores. In addition, the dexmedetomidine group had a shorter time to extubation (median length of 19.9 hours vs. 42.5 hours in the haloperidol group), shorter length of ICU hospitalization (4.5 days vs. 8 days in the haloperidol group), as well as an overall shorter time in mechanical restraints and a reduced necessity for supplemental propofol when compared to the haloperidol group. Though promising, this study had multiple limitations including: small sample size, lack of blinding, possible underdosing in the haloperidol arm, and the assumption that agitation was due to delirium when, in fact, agitation is a nonspecific symptom with multiple potential causes.27

Large-scale, randomized, placebo-controlled trials are still needed to establish the efficacy of using dexmedetomidine as a primary agent in treating delirium in the ICU population. Furthermore, the high cost of dexmedetomidine (on average $300–$400 per day) seems to be a limiting factor at first glance; however, a recent cost minimization analysis comparing dexmedetomidine and midazolam for sedation in a cohort of 366 mechanically-ventilated ICU patients showed that usage of dexmedetomidine resulted in a median total ICU cost savings of $9679 compared with midazolam.25,35 Findings such as these are promising and suggest that usage of dexmedetomide may in fact lower overall healthcare costs; however, further replication of such studies is necessary to make any definitive conclusions on this basis. Nonetheless, dexmedetomidine remains a viable alternative in managing treatment-refractory delirium in the critical care population.

Pharmacologic Prophylaxis

The increasingly apparent morbidity and mortality associated with delirium in the ICU promotes the search for possible pharmacologic prevention strategies. While such prophylactic measures are still experimental, some promising studies have shown a possible role for premedicating certain at-risk patient populations in the interest of reducing delirium incidence and resultant sequelae. Most of these studies have been done in non-ICU populations, are relatively underpowered, and remain mixed at best. Those studies limited to the ICU population are far fewer.

A large proportion of studies looking at pharmacologic prophylaxis measures for delirium have been limited to patients undergoing elective surgical procedures. Of these, only the studies examining prophylaxis in elective cardiac surgery have involved patients in the ICU setting.31,3639 While α-2 agonists are discussed below, the studies to date have also looked at antipsychotics, ketamine, and cholinesterase inhibitors. One such study, by Prakanrattana and colleagues, looked at 126 patients undergoing cardiac surgery who were randomized to receive a single dose of either risperidone or placebo post-surgically.36 The risperidone group was found to have a lower incidence of postoperative delirium (11.1% vs. 31.7% in the placebo arm). Similarly, a smaller study by Hudetz and colleagues examined the incidence of postoperative delirium in 58 cardiac surgery patients randomized to receive either ketamine or placebo during anesthetic induction with fentanyl and etomidate.37 Findings showed that patients who received ketamine had a lower incidence of postoperative delirium (3% vs. 31% in the placebo arm). Finally, Gamberini and colleagues looked at 120 patients undergoing cardiac surgery who were randomized to receive either rivastigmine (three doses daily starting the evening before surgery and continued until the sixth postoperative day) or placebo.38 Akin to the findings of most large-scale studies using cholinesterase inhibitors for delirium, there was no significant difference in the incidence of postoperative delirium between the rivastigmine and placebo groups (32% vs. 30%, respectively).

When looking exclusively at the ICU population, studies examining the efficacy of pharmacologic delirium prophylaxis have focused mainly on α-2 agonists. Despite one small-scale placebo-controlled study looking at the incidence of delirium with IV clonidine during ventilator weaning, most large-scale studies have focused on dexmedetomidine.40 A pivotal trial was performed by Pandharipande and colleagues and is known as the maximizing efficacy of targeted sedation and reducing neurologic dysfunction (MENDS) trial.24 In this double-blind trial, 106 mechanically-ventilated patients from two tertiary care centers were randomized to receive either dexmedetomidine or lorazepam for sedation. The primary outcome measure was days alive without delirium or coma. The dexmedetomidine group was found to have significantly more days alive without coma or delirium compared with the lorazepam group (7 days vs. 3 days). Furthermore, though not statistically significant, mortality measured after 28 days was found to be lower in the dexmedetomidine group compared with the lorazepam group (17% vs. 27%) as were ventilator-free days (22 days vs. 18 days, respectively). Subsequent subgroup analysis looking at septic vs. non-septic patients in the MENDS trial showed even greater differences, with septic patients in the dexmedetomidine group having significantly more days alive without delirium or coma, 70% less risk of mortality at 28 days, and more ventilator-free days compared with the septic patients in the lorazepam group.41

Limitations of the MENDS trial included the lack of a placebo arm and exclusion of patients with neurologic diseases (such as stroke or active seizures), severe liver disease, alcohol abuse, active myocardial infarction, second- or third-degree heart block, dementia, benzodiazepine dependence, severe hearing impairment, or being non-English speaking. Furthermore, per study design guidelines implemented by the FDA, dexmedetomidine could not be administered for more than 120 hours.24 This may have potentially introduced confounders given the necessity to switch to standard sedation (lorazepam or midazolam) in patients who were in the dexmedetomidine study arm and necessitated continued sedation beyond 120 hours.24

A similar study by Maldonado and colleagues looked at 118 mechanically ventilated cardiac surgery patients randomized to receive dexmedetomidine, propofol, or midazolam for postoperative sedation.31 The incidence of delirium was found to be significantly lower in the dexmedetomidine group (3%) compared with the propofol (50%) and midazolam groups (50%). The study lacked a placebo arm, was limited to patients undergoing valve replacement surgery, and excluded patients with the following conditions: dementia, on psychotropic medications, those with a history of substance abuse, advanced heart block, pregnancy, stroke within the past 6 months, and those age <18 years or >90 years. Furthermore, despite the lower incidence of delirium in the dexmedetomidine group, there were no significant differences in mean ICU or hospital stay.

Similarly, Shehabi and colleagues looked at 306 mechanically ventilated patients who underwent cardiac surgery and were randomized to receive either dexmedetomidine or morphine at equivalent levels of sedation/analgesia with delirium incidence as the primary endpoint.39 Findings showed that there was no statistically significant difference in delirium incidence between groups (8.6% in the dexmedetomidine vs. 15% in the morphine group); however, dexmedetomidine patients spent 3 fewer days in delirium compared with the morphine group and were extubated sooner. Upon subgroup analysis, those patients who required an intra-aortic balloon pump and were in the dexmedetomidine group had a significantly lower incidence of delirium compared with the same subgroup of patients in the morphine arm (15% vs. 36%, respectively). This particular study was limited by its homogeneous population (cardiac surgery patients aged 60 yrs and older), a lack of delirium surveillance beyond 5 days, and the fact that the use of open-label morphine in the dexmedetomidine study group may have confounded results.

Other studies with dexmedetomidine in various sedation/analgesia protocols have looked at delirium incidence and duration only as secondary endpoints and have yielded mixed results. Compared with standard sedation protocols, dexmedetomidine has been shown to be as effective for moderate sedation with the added benefit of a significantly lower incidence of delirium in one study.25 On the other hand, a similar study did not show a significant reduction in the incidence of delirium.26 Results remain mixed and further studies looking at delirium incidence as a primary endpoint are still needed to make any definitive conclusions. Dexmedetomidine remains a viable alternative to standard sedating medications in mechanically-ventilated ICU patients who are already at an increased risk for developing or exacerbating delirium; however, use in delirium prophylaxis alone should still be exercised with caution as efficacy is yet to be solidified.

Conclusions

This article summarizes the spectrum of pharmacologic intervention studies for delirium in the critical care population. It remains of paramount importance for the practicing psychiatrist to be familiar with the breadth of pharmacotherapeutic options for the management and potential prevention of delirium. Studies to date remain mixed at best and warrant further investigation in this population.

References

Ely EW, Shintani A, Truman B, Speroff T, Gordon SM, Harrell FE, et al.: Delirium as a predictor of mortality in mechanically ventilated patients in the intensive care unit. JAMA 2004; 291(14):1753–1762CrossrefGoogle Scholar

Heymann A, Radtke F, Schiemann A, Lütz A, MacGuill M, Wernecke KD, et al.: Delayed treatment of delirium increases mortality rate in intensive care unit patients. J Int Med Res 2010; 38(5):1584–1595CrossrefGoogle Scholar

Hshieh TT, Fong TG, Marcantonio ER, Inouye SK: Cholinergic deficiency hypothesis in delirium: a synthesis of current evidence. J Gerontol A Biol Sci Med Sci 2008; 63(7):764–772CrossrefGoogle Scholar

Schetz JA, Perez E, Liu R, Chen S, Lee I, Simpkins JW: A prototypical σ-l receptor antagonist protects against brain ischemia. Brain Res 2007; 1181:1–9CrossrefGoogle Scholar

Song C, Lin A, Kenis G, Bosmans E, Maes M: Immunosuppressive effects of clozapine and haloperidol: enhanced production of the interleukin-1 receptor antagonist. Schizophr Res 2000; 42(2):157–164CrossrefGoogle Scholar

Jacobi J, Fraser GL, Coursin DB, Riker RR, Fontaine D, Wittbrodt ET, et al.: Task Force of the American College of Critical Care Medicine (ACCM) of the Society of Critical Care Medicine (SCCM), American Society of Health-System Pharmacists (ASHP), American College of Chest Physicians. Clinical practice guidelines for the sustained use of sedatives and analgesics in the critically ill adult. Crit Care Med 2002; 30(1):l19–141; Erratum in: Crit Care Med 2002; 30(3):726CrossrefGoogle Scholar

Riker RR, Fraser GL, Cox PM: Continuous infusion of haloperidol controls agitation in critically ill patients. Crit Care Med 1994; 22(3):433–440CrossrefGoogle Scholar

Seneff MG, Mathews RA: Use of haloperidol infusions to control delirium in critically ill adults. Ann Pharmacother 1995; 29(7/8):690–693CrossrefGoogle Scholar

Menza MA, Murray GB, Holmes VF, Rafuls WA: Decreased extrapyramidal symptoms with intravenous haloperidol. J Clin Psychiatry 1987; 48(7):278–280Google Scholar

Glassman AH, Bigger JT.: Antipsychotic drugs: Prolonged QTc interval, torsades de pointes, and sudden death. Am J Psychiatry 2001; 158(11):1774–1782CrossrefGoogle Scholar

Lonergan E, Britton AM, Luxenberg J, Wyller T: Antipsychotics for delirium. Cochrane Database Syst Rev 2007; 2:CD005594. ReviewCrossrefGoogle Scholar

Rea RS, Battistone S, Fong JJ, Devlin JW: Atypical antipsychotics vs. haloperidol for treatment of delirium in acutely ill patients. Pharmacotherapy 2007; 27(4):588–594CrossrefGoogle Scholar

Han C, Kim Y: A double-blind trial of risperidone and haloperidol for the treatment of delirium. Psychosomatics 2004; 45(4):297–301CrossrefGoogle Scholar

Skrobik YK, Bergeron N, Dumont M, Gottfried SB: Olanzapine vs. haloperidol: treating delirium in a critical care setting. Intensive Care Med 2004; 30(3):444–449CrossrefGoogle Scholar

Girard TD, Pandharipande PP, Carson SS, Schmidt GA, Wright PE, Canonico AE, et al.: MIND Trial Investigators. Feasibility, efficacy, and safety of antipsychotics for intensive care unit delirium: the MIND randomized, placebo-controlled trial. Crit Care Med 2010; 38(2):428–437CrossrefGoogle Scholar

Balas MC. Free your MIND and the rest will follow: decoding delirium in the intensive care unit. Crit Care Med 2010; 38(2):697–698CrossrefGoogle Scholar

Devlin JW, Roberts RJ, Fong JJ, Skrobik Y, Riker RR, Hill NS, et al.: Efficacy and safety of quetiapine in critically ill patients with delirium: a prospective, multicenter, randomized, double-blind, placebo-controlled pilot study. Crit Care Med 2010; 38(2):419–427CrossrefGoogle Scholar

van Eijk MM, Roes KC, Honing ML, Kuiper MA, Karakus A, van der Jagt M, et al.: Effect of rivastigmine as an adjunct to usual care with haloperidol on duration of delirium and mortality in critically ill patients: a multicenter, double-blind, placebo-controlled randomised trial. Lancet 2010; 376(9755):1829–1837CrossrefGoogle Scholar

Aantaa R, Jalonen J: Perioperative use of α-2 adrenoceptor agonists and the cardiac patient. Eur J Anaesthesiol 2006; 23(5):361–372CrossrefGoogle Scholar

Huang R, Chen Y, Yu AC, Hertz L: Dexmedetomidine-induced stimulation of glutamine oxidation in astrocytes: a possible mechanism for its neuroprotective activity. J Cereb Blood Flow Metab 2000; 20(6):895–898CrossrefGoogle Scholar

Gertler R, Brown HC, Mitchell DH, Silvius EN: Dexmedetomidine: a novel sedative-analgesic agent. Proc (Bayl Univ Med Cent) 2001; 14(1):13–21CrossrefGoogle Scholar

Guinter JR, Kristeller JL. Prolonged infusions of dexmedetomidine in critically ill patients. Am J Health-Syst Pharm 2010; 67:1246–1253CrossrefGoogle Scholar

Gerlach AT, Murphy CV, Dasta JF: An updated focused review of dexmedetomidine in adults. Ann Pharmacother 2009; 43(12):2064–2074CrossrefGoogle Scholar

Pandharipande PP, Pun BT, Herr DL, Maze M, Girard TD, Miller RR, et al.: Effect of sedation with dexmedetomidine vs. lorazepam on acute brain dysfunction in mechanically ventilated patients: the MENDS randomized controlled trial. JAMA 2007; 298(22):2644–2653CrossrefGoogle Scholar

Riker RR, Shehabi Y, Bokesch PM, Ceraso D, Wisemandle W, Koura F, et al.: SEDCOM (Safety and Efficacy of Dexmedetomidine Compared with Midazolam) Study Group. Dexmedetomidine vs. midazolam for sedation of critically ill patients: a randomized trial. JAMA 2009; 301(5):489–499CrossrefGoogle Scholar

Ruokonen E, Parviainen I, Jakob SM, Nunes S, Kaukonen M, Shepherd ST, et al.: “Dexmedetomidine for Continuous Sedation” Investigators. Dexmedetomidine vs. propofol/midazolam for long-term sedation during mechanical ventilation. Intensive Care Med 2009; 35(2):282–290CrossrefGoogle Scholar

Reade MC, O’Sullivan K, Bates S, Goldsmith D, Ainslie WR, Bellomo R: Dexmedetomidine vs. haloperidol in delirious, agitated, intubated patients: a randomized open-label trial. Crit Care 2009; 13(3):R75CrossrefGoogle Scholar

Aho M, Lehtinen AM, Erkola O, Kallio A, Korttila K: The effect of intravenously administered dexmedetomidine on perioperative hemodynamics and isoflurane requirements in patients undergoing abdominal hysterectomy. Anesthesiology 1991; 74(6):997–1002CrossrefGoogle Scholar

Buttermann AE, Reid K, Maze M. Are cholinergic pathways involved in the anesthetic response to α-2 agonists. Toxicol Lett 1998; 100/101:17–22CrossrefGoogle Scholar

Hsu YW, Cortinez LI, Robertson KM, Keifer JC, Sum-Ping ST, Moretti EW, et al.: Dexmedetomidine pharmacodynamics: part I: crossover comparison of the respiratory effects of dexmedetomidine and remifentanil in healthy volunteers. Anesthesiology 2004; 101(5):1066–1076CrossrefGoogle Scholar

Maldonado JR, Wysong A, van der Starre PJ, Block T, Miller C, Reitz BA: Dexmedetomidine and the reduction of postoperative delirium after cardiac surgery. Psychosomatics 2009; 50(3):206–217CrossrefGoogle Scholar

Nelson LE, Lu J, Guo T, Saper CB, Franks NP, Maze M. The α-2 adrenoceptor agonist dexmedetomidine converges on an endogenous sleep-promoting pathway to exert its sedative effects. Anesthesiology 2003; 98(2):428–436CrossrefGoogle Scholar

Weinbroum AA, Ben-Abraham R: Dextromethorphan and dexmedetomidine: new agents for the control of perioperative pain. Eur J Surg 2001; 167(8):563–569CrossrefGoogle Scholar

Pandharipande P, Shintani A, Peterson J, Pun BT, Wilkinson GR, Dittus RS, et al.: Lorazepam is an independent risk factor for transitioning to delirium in intensive care unit patients. Anesthesiology 2006; 104(1):21–26CrossrefGoogle Scholar

Dasta JF, Kane-Gill SL, Pencina M, Shehabi Y, Bokesch P, Wisemandle W, et al.: A cost minimization analysis of dexmedetomidine compared with midazolam for long-term sedation in the intensive care unit. Crit Care Med 2010; 38(2):497–503CrossrefGoogle Scholar

Prakanrattana U, Prapaitrakool S. Efficacy of risperidone for prevention of postoperative delirium in cardiac surgery. Anaesth Intensive Care 2007; 35(5):714–719CrossrefGoogle Scholar

Hudetz JA, Patterson KM, Iqbal Z, Ganhi SD, Byrne AJ, Hudetz AG, et al.: Ketamine attenuates delirium after cardiac surgery with cardiopulmonary bypass. J Cardiothorac Vase Anesth 2009; 23(5):651–657CrossrefGoogle Scholar

Gamberini M, Bolliger D, Lurati Buse GA, Burkhart CS, Grapow M, Gagneux A, et al.: Rivastigmine for the prevention of postoperative delirium in elderly patients undergoing elective cardiac surgery—a randomized controlled trial. Crit Care Med 2009; 37(5):1762–1768CrossrefGoogle Scholar

Shehabi Y, Grant P, Wolfenden H, Hammond N, Bass F, Camp bell M, et al.: Prevalence of delirium with dexmedetomidine compared with morphine based therapy after cardiac surgery: a randomized controlled trial (DEXmedetomidine COmpared to Morphine-DEXCOM Study). Anesthesiology 2009; 111(5):1075–1084CrossrefGoogle Scholar

Rubino AS, Onorati F, Caroleo S, Galato E, Nucera S, Amantea B, et al.: Impact of clonidine administration on delirium and related respiratory weaning after surgical correction of acute type-A aortic dissection: results of a pilot study. Interact Cardiovasc Thorac Surg 2010; 10(l):58–62CrossrefGoogle Scholar

Pandharipande P, Sanders RD, Girard TD, McGrane S, Thompson JL, Shintani AK, et al., MENDS investigators: Effect of dexmedetomidine vs. lorazepam on outcome in patients with sepsis: an a priori-designed analysis of the MENDS randomized controlled trial. Crit Care 2010; 14(2):R38CrossrefGoogle Scholar