Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Dexmedetomidine for the prevention of postoperative delirium in elderly patients undergoing noncardiac surgery: A meta-analysis of randomized controlled trials

  • Hai Zeng,

    Roles Conceptualization, Data curation, Formal analysis, Methodology, Software, Writing – review & editing

    Affiliation The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China

  • Zunjiang Li,

    Roles Data curation, Formal analysis, Investigation, Project administration, Software, Writing – original draft

    Affiliation The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China

  • Jianbin He,

    Roles Data curation, Investigation, Project administration, Software, Writing – original draft

    Affiliation School of Basic Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China

  • Wenbin Fu

    Roles Conceptualization, Formal analysis, Methodology, Software, Supervision, Writing – review & editing

    zjkzh@139.com

    Affiliations Department of Acupuncture and Moxibustion, The Second Affiliated hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong Province, China, Shenzhen Bao’an Research Center for Acupuncture and Moxibustion, Shenzhen, Guangdong Province, China, Sanming Project of Medicine in Shenzhen (No. SZSM201806077), Shenzhen, Guangdong Province, China

Abstract

Background

Postoperative delirium (POD) among the elderly population that undergoes noncardiac surgery is significantly associated with adverse clinical outcomes. We conducted this meta-analysis to evaluate the effectiveness and safety of dexmedetomidine for the prophylaxis of POD among the elderly population after noncardiac surgery.

Methods

We searched Embase, PubMed, and the Cochrane Library from inception date to March 2019 for randomized controlled trials (RCTs) that compared dexmedetomidine and placebo for the prevention of POD and evaluated the major cardiovascular outcomes among elderly people after noncardiac surgery. Two authors independently screened the studies and extracted data from the published articles. The main outcome was the incidence of POD. The secondary outcomes included the occurrence of bradycardia, hypotension, hypertension, tachycardia, myocardial infarction, stroke, hypoxaemia, and all-cause mortality.

Results

A total of 6 RCTs with 2102 participants were included. Compared with placebo, dexmedetomidine significantly reduced the prevalence of POD (RR = 0.61, 95% CI 0.34–0.76, P = 0.001, I2 = 66%), and the risk of tachycardia (RR = 0.48, 95% CI 0.30–0.76, P = 0.002, I2 = 0%), hypertension (RR = 0.59, 95% CI 0.44–0.79, P < 0.001, I2 = 20%), stroke (RR = 0.22, 95% CI 0.06–0.76, P = 0.02, I2 = 0%), and hypoxaemia (RR = 0.50, 95% CI 0.32–0.78, P = 0.002, I2 = 0%) in elderly patients who underwent noncardiac surgery. However, dexmedetomidine accelerated the occurrence of bradycardia (RR = 1.36, 95% CI 1.11–1.67, P = 0.003, I2 = 0%). Furthermore, no significant differences were observed in the incidence of hypotension, myocardial infarction, and all-cause mortality between the dexmedetomidine and placebo groups.

Conclusions

Among elderly patients after noncardiac surgery, the prophylactic use of dexmedetomidine, compared with the use of placebo, was related to a decline in the incidence of POD.

Introduction

Delirium is usually characterized as a multifactorial syndrome of acute attention and cognitive disorders; delirium is also a common, serious, underrecognized, and even lethal condition, especially for geriatric patients [1]. Delirium is associated with an elevated risk of mortality, complication morbidity, and dementia, an extended length of hospital stay, and a worsening in health-related quality of life [26]. The prevalence of postoperative delirium (POD) in elderly patients after noncardiac surgery is approximately 13% to 50% [1]. To decrease the incidence and adverse outcomes associated with delirium, multicomponent nonpharmacologic approaches that help control multiple risk factors of delirium are recommended [7, 8]. However, to date, the use of pharmacologic intervention to prevent delirium remains controversial [1, 811].

Dexmedetomidine serves as a potent sedative and has positive sedation and analgesic effects with modest anxiolytic ability, minimal respiratory depression and neurotoxicity; the mechanism of dexmedetomidine is closely related to its highly selective stimulation of the alpha-2 adrenoreceptors [12, 13]. In recent years, the usage of dexmedetomidine has increased for critically ill patients, particularly for surgical patients [14].

Two published meta-analyses [15, 16] showed that dexmedetomidine might decrease the occurrence of POD, compared with other active sedative drugs, in adult patients who underwent cardiac surgery. However, it is not clear whether dexmedetomidine exerts benefits on delirium prevention after noncardiac surgery among the elderly population when the drug is compared with placebo. Recent randomized controlled trials (RCTs) have reported inconsistent conclusions about the prophylactic effect of dexmedetomidine on POD in elderly patients. As new studies continue to provide extensive new data and insights into the potential effects of dexmedetomidine on POD and other major clinical outcomes, this review was performed to estimate the updated influence of dexmedetomidine on POD by comparing the drug with placebo in elderly patients after noncardiac surgery.

Materials and methods

The meta-analysis was reported in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) criteria [17]. The protocol of our review was registered in the International Prospective Register of Systematic Reviews (PROSPERO) (registration no. CRD42018105508).

Search strategy

Two independent reviewers conducted systematic literature searches. The searches included PubMed, Embase, and the Cochrane Library databases. The literature search of each database was up to date as of March 2019. A language restriction on studies published in English was imposed. The study design was limited to RCTs. The following Medical Subject Headings (MeSH) or non-MeSH terms and their combinations were searched in the title and abstract: “dexmedetomidine”, “precedex”, “placebo”, “saline”, “Sodium Chloride”, “salt water”, “placebos”, “elder”, “aged”, “elderly”, “geriatric”, “old”, “older”, “elders”, “geriatrics”, “intraoperative”, “surgery”, “surgical”, “operation”, “postoperative”, “anesthesia”, “anaesthesia”, “operative”, “perioperative”, “clinical”, “trial”, and “random”. The complete search strategies of PubMed and Embase are presented in the supplementary (S1 File). Any disagreements reached a consensus by discussing with a third reviewer.

Inclusion criteria

Patients: elderly population (aged 65 years or older) who underwent noncardiac surgery under general anesthesia; intervention: dexmedetomidine; comparison: placebo (normal saline); primary outcome: the incidence of POD; and secondary outcomes: the occurrence of major cardiovascular events (bradycardia, hypotension, hypertension, tachycardia, stroke, and myocardial infarction), hypoxaemia, and all-cause death. The study design was limited to RCTs.

Exclusion criteria

Duplicated reports, trials that did not report the outcomes of interest, and case reports were excluded. We also eliminated trials in which other interventions were conducted in addition to the use of dexmedetomidine (e.g., nonpharmacological interventions and use of antipsychotics).

Data extraction

Any selected study that fulfilled the inclusion criteria was included. The data from each eligible study were extracted by two independent reviewers. Discrepancies were reconciled after discussing with a third reviewer. The data extracted included the year of publication, the first author’s name, basic characteristics of the participants (average age, sex ratio, and sample size), countries where the trials were conducted, surgery and anesthesia type, assessment method and time of POD, intervention time of dexmedetomidine, the sedative dose and rate of dexmedetomidine infusion, and the primary and secondary outcomes mentioned above.

Quality assessment

Using the Cochrane Collaboration’s tool for evaluating the risk of bias, the quality of each included trial was independently estimated by two investigators, with any discrepancies reconciled by a third reviewer. The tool had 7 quality items: random sequence generation, allocation concealment, blinding of participants and personnel, blinding of outcome assessment, incomplete outcome data, selective reporting, and other bias [18]. Each item was classified as a low, unclear, or high risk of bias [18].

Data analysis

Statistical analyses were completed with RevMan (version 5.3.5) and Stata (version 14.0) software. For dichotomous outcomes, summary relative risks (RRs) with corresponding 95% CIs were calculated. A P value of less than 0.05 was deemed statistically significant. Heterogeneity was calculated quantitatively using the I2 statistic. The I2 statistic was deemed to represent a no (0%), low (0%-25%), moderate (25%-75%), and high (75%-100%) likelihood of heterogeneity [19]. A random-effects model was adopted since there was heterogeneity among studies. We used the Egger regression test and the Begg’s rank correlation test to examine the publication bias in addition to visual measurements of the funnel plots.

Results

Search results

The initial literature search yielded 1117 studies. The number of duplicated articles removed was 290. A total of 813 studies were excluded after screening the titles and abstracts. Thirteen studies were reviewed for eligibility by scrutinizing full-text articles. Of those studies, 6 studies [2025] reported no relevant clinical outcomes of interest, and one study [26] involved participants who were younger than 65 years. Finally, 6 eligible RCTs (2102 elderly patients) [2732] were included in our review. The PRISMA flowchart is presented in Fig 1. Additionally, the interrater reliability measures between the two reviewers showed high agreement with kappa values of 0.83 for systematic searches and 0.89 for study selection.

Study characteristics

The basic characteristics of the six included RCTs are listed in S1 Table in the supporting information; these RCTs included a total study population of 2102 randomized participants (sample sizes ranging from 76 [32] to 700 patients [31]). The process of data extraction had a kappa value of 0.92, demonstrating high agreement between the two authors. Three studies [27, 28, 31] were multicenter trials. The results of these trials were published during or after 2015. All studies enrolled participants aged 65 years or older. All studies were randomized placebo-controlled trials. In all included studies, the diagnosis of delirium was based on the confusion assessment method (CAM) or CAM for the intensive care unit (CAM-ICU). Both the CAM and CAM-ICU are well-established delirium assessment instruments that have been widely used in clinical trials [1, 9, 33, 34]. In the included trials, the types of noncardiac surgery varied and included intra-abdominal, intra-thoracic, spinal and extremital, orthopedic, superficial, and urologic surgery. Additionally, the intervention time of dexmedetomidine was different among the included studies. In three studies [27, 29, 30], dexmedetomidine was used during the intraoperative period. In two trials [31, 32], dexmedetomidine was administered after surgery. In another study [28], dexmedetomidine was administered in both the intraoperative and postoperative periods.

Quality assessment

The results of the methodological quality assessment are summarized in the Fig 2. Following strict judgments of each included study according to the Cochrane handbook, three trials [28, 31, 32] were assessed as high quality as a result of having low risks of methodological bias across all seven domains. One study [29] was considered low quality; there was a high risk of bias in the blinding of the researchers, because the anesthesiologists and nurses were not blinded to anesthetic agents. In one study [27], there was inadequate information to decide whether a risk of selective reporting bias existed; thus, this study was considered as a moderate-quality study. In addition, another study [30] was considered to be of moderate quality due to an unclear risk of bias as a consequence of inadequate information in both assignment concealment and blinding of the outcome evaluation.

Results of the meta-analysis

POD.

The incidence of POD was reported in six studies [2732], with 320 events (15.2%) among 2102 patients. The reported incidence of POD among the included studies ranged from 6.6% [32] to 29.4% [30]. The overall pooled crude incidence of POD differed significantly between the dexmedetomidine and placebo groups. Only 106 of the 1040 patients in the dexmedetomidine group were reported to have POD, which accounted for 10.2% of the patients, while 214 of the 1062 patients in the placebo group were reported to have POD, which was a rate of 20.2%. The Pooled data from the six trials revealed that dexmedetomidine could significantly decrease the incidence of POD compared with placebo (RR = 0.61, 95% CI 0.34–0.76, P = 0.001, I2 = 66%; Fig 3).

Bradycardia.

Three studies [28, 31, 32] reported the incidence of bradycardia, with 269 events (23.1%) among 1166 patients. Dexmedetomidine, compared to placebo, was associated with a significant increase in the risk of perioperative bradycardia (RR = 1.36, 95% CI 1.11–1.67, P = 0.003, I2 = 0%; Fig 4).

Hypotension.

Three studies [28, 31, 32] reported the occurrence of hypotension, with 423 events (36.3%) among 1166 patients. Dexmedetomidine had no significant influence on the occurrence of perioperative hypotension when compared with placebo (RR = 1.27, 95% CI 0.98–1.64, P = 0.07, I2 = 54%; Fig 5).

Hypertension.

Three studies [28, 31, 32] reported the incidence of hypertension, with 220 events (18.7%) among 1166 patients. The pooled analysis demonstrated that dexmedetomidine, compared to placebo, could significantly decrease the incidence of perioperative hypertension (RR = 0.59, 95% CI 0.44–0.79, P < 0.001, I2 = 20%; Fig 6).

Tachycardia.

Two studies [31, 32] reported the occurrence of tachycardia, with 74 events (9.5%) among 776 patients. The pooled analysis demonstrated that dexmedetomidine, compared to placebo, was associated with a statistically significant decrease in the prevalence of perioperative tachycardia (RR = 0.48, 95% CI 0.30–0.76, P = 0.002, I2 = 0%; Fig 7).

Myocardial infarction.

Three studies [27, 28, 31] reported the occurrence of myocardial infarction, with 17 events (1.1%) among 1539 patients. The pooled data from these studies showed that dexmedetomidine had no significant influence on the incidence of myocardial infarction when compared with placebo (RR = 0.70, 95% CI 0.27–1.82, P = 0.46, I2 = 0%; Fig 8).

Stroke.

Three studies [27, 28, 31] reported the incidence of stroke, with 15 events (1.0%) among 1539 patients. The pooled results suggested that the use of dexmedetomidine might be associated with a reduced incidence of stroke when compared with placebo (RR = 0.22, 95% CI 0.06–0.76, P = 0.02, I2 = 0%; Fig 9).

Hypoxaemia.

Two studies [31, 32] reported the incidence of hypoxaemia, with 81 events (10.4%) among 776 patients. The pooled data from three studies showed that dexmedetomidine had no significant influence on the occurrence of hypoxaemia when compared with placebo (RR = 0.50, 95% CI 0.32–0.78, P = 0.002, I2 = 0%; Fig 10).

All-cause mortality.

Four studies [27, 28, 31, 32] reported the incidence of all-cause mortality, including 11 events (0.6%) among a total of 1701 patients. The pooled analysis showed that dexmedetomidine did not cause a significant decrease in the risk of all-cause mortality when compared with placebo (RR = 0.40, 95% CI 0.10–1.56, P = 0.18, I2 = 0%; Fig 11).

Publication bias

Using the incidence of POD as the primary outcome, a visual inspection of the funnel plot (Fig 12) showed no apparent evidence of publication bias. Furthermore, no obvious evidence of publication bias was identified, with an Egger’s test P value of 0.89 and a Begg’s rank correlation of 0.71.

Discussion

The meta-analysis incorporated six RCTs [2732] that met the inclusion criteria and included 2102 elderly patients in total. The main conclusion of this review was that elderly patients who underwent noncardiac surgery and received dexmedetomidine intervention had a significantly lower occurrence of POD than those who received placebo. Even though dexmedetomidine could increase the risk of bradycardia, the drug might reduce the occurrence of tachycardia, hypertension, stroke, and hypoxaemia; no significant differences were discovered in the incidence of hypotension, myocardial infarction, and all-cause mortality between the dexmedetomidine and placebo groups.

Postoperative delirium is an acute cognitive disturbance that plays an important role in postoperative outcomes and remains a serious and costly condition, particularly in elderly patients [35]. The etiology of delirium is multifactorial (e.g., history of dementia or cognitive impairment and old age), the prominent potential pathophysiologic contributors of developing delirium involve abnormal levels of neurotransmitters and pro-inflammatory markers, physiologic stressors, metabolic derangements, and electrolyte disorders plus genetic factors [1]. There might be an interplay between the inflammatory response of the peripheral innate immune system and the human brain that is potentially associated with cognitive function impairments after surgery with general anesthesia [36].

Dexmedetomidine, a highly selective alpha-2 adrenoreceptor agonist, has the positive sedation, anti-anxiety, and analgesic effects [12, 13, 37]. Thus, dexmedetomidine has been widely used in surgical patients. Because of the complex etiology and pathophysiology of delirium, the accurate mechanisms by which dexmedetomidine could decrease the risk of delirium remain poorly understood, although there are some studies on this issue. Dexmedetomidine could suppress systemic inflammatory processes through the downregulation of the HMGB1-TLR4-NF-κB signaling pathway by activating alpha-2 adrenergic receptors and stimulating the vagus nerve via a vagal- and alpha-7 nicotinic acetylcholine receptor-dependent mechanism [38, 39]. Dexmedetomidine might confer protective effects against transient cerebral ischemia or ischemic reperfusion impairment via restraining inflammation and anesthesia-induced (e.g., propofol) neurotoxicity in the brain [12, 40]. The findings of these studies are consistent with the results of our meta-analysis, which suggest that perioperative intervention with dexmedetomidine might be a promising option for ameliorating the occurrence of delirium following surgery with general anesthesia.

Previous reviews [15, 16] demonstrated that the perioperative usage of dexmedetomidine was related to a reduced prevalence of delirium in adult patients who underwent cardiac surgery. In addition, Duan et al. [41] assessed the efficacy of dexmedetomidine for delirium prevention among adult patients after noncardiac surgery and revealed similar results. However, these studies [15, 16, 41] compared dexmedetomidine with other sedative drugs (e.g., midazolam and propofol) that activate γ-aminobutyric acid A (GABAA) receptors. Notably, the current evidence indicates that the use of GABAA agonists is associated with a high risk of developing delirium [9, 11, 4245]. One plausible interpretation is that dexmedetomidine does not induce delirium as do these GABAA agonists, but also does not prevent delirium [31]. To clearly illuminate this controversial topic, we conducted this meta-analysis and confirmed that dexmedetomidine could effectively prevent POD among elderly patients after noncardiac surgery when compared with placebo.

Among the included studies, four RCTs [27, 2931] found that dexmedetomidine treatment could reduce the incidence of delirium while the results observed in the remaining studies [28, 32] were in contrast with these findings. In the study by Su and his colleagues [31], patients who received a continuous intravenous infusion of dexmedetomidine from study recruitment on the day of surgery until postoperative day 1 were observed to have a decreased risk of delirium. However, Deiner and his colleagues [28] limited dexmedetomidine administration to intraoperative period and 2 hours into recovery. The lack of salutary effects might be partly due to the short-acting nature of the drug and the unsuitable timing of drug infusion. In addition, the incidence of delirium was not the primary endpoint of the relatively small study by Wu et al [32], which had a sample size of 61 patients.

The most common adverse effects of dexmedetomidine that have been reported are bradycardia and hypotension, which are the adverse consequences of stimulating alpha-2 adrenoreceptors [46, 47]. Dexmedetomidine is an excellent drug that could not only reduce the magnitude of the hemodynamic response to anesthetic induction, tracheal intubation, surgery and extubation, but could also decrease the consumption of opioids and isoflurane to achieve appropriate analgesia and anesthesia [48, 49]. Our study also discovered that dexmedetomidine could reduce the risk of tachycardia and hypertension in elderly patients who underwent noncardiac surgery, but dexmedetomidine might increase the incidence of perioperative bradycardia; these findings are in accordance with the results of two previous meta-analyses [15, 16]. Notably, these adverse effects are both multifactorial and dosage-dependent and are mediated via both central and peripheral mechanisms [47].

Myocardial infarction and stroke are common cardiovascular complications following surgery and are strongly related to a high risk of mortality [50, 51]. Our meta-analysis did not observe any significant differences between the dexmedetomidine and placebo groups in the occurrence of myocardial infarction and all-cause mortality. A prior Cochrane review [52] revealed similar conclusions. Moreover, as observed in our study, the usage of dexmedetomidine in the perioperative period might be associated with a reduced incidence of stroke, but more large-scale cardiovascular outcome trials are required to confirm this finding. In addition, dexmedetomidine was related to a decreased risk of hypoxaemia, which indicated that dexmedetomidine might not result in respiratory depression. This result is in line with another study [53] that showed that dexmedetomidine had no harmful clinical effects on respiration when used in the surgical population that required intensive care.

However, this review has several limitations. First, some restrictions exist in number and quality of the included trials, since only six RCTs were included in our study. A high risk of bias in the blinding of researchers was identified in one study [29], and two other studies [27, 30] were considered moderate quality in consequence of unclear risk from inadequate descriptions of the allocation concealment process, blinding of outcome assessments or selective reporting. Second, we identified moderate heterogeneity among the included trials, including in variables such as intervention time (intraoperative, intraoperative plus postoperative, and postoperative), type of surgery, sedative dose and rate of dexmedetomidine infusion, and patient characteristics. Moreover, there was an inadequate number of trials to apply meta-regression methods that could assess the variables potentially related to heterogeneity. Finally, although no statistical evidence of publication bias was observed, the probability of bias still exists as a consequence of the low statistical power caused by the limited quantity of included studies.

Despite the limitations above, to the best of our knowledge, this review was the first meta-analysis to estimate the prophylaxis efficacy of dexmedetomidine on POD when compared with placebo in elderly patients after noncardiac surgery. Furthermore, our review only included randomized double-blind placebo-controlled clinical trials, whose rigorous study designs strengthens the creditability of the main outcomes.

Conclusion

In summary, the results of our review indicated that perioperative prophylactic intervention with dexmedetomidine, compared with placebo, could significantly reduce the prevalence of POD, tachycardia, hypertension, and hypoxaemia in elderly patients following non-cardiac surgery. However, the use of dexmedetomidine was associated with an elevated risk of bradycardia. Additional high-quality, large-scale multicenter RCTs are still warranted for the purpose of exploring the optimal dose and timing of dexmedetomidine on POD prevention among the elderly population after noncardiac surgery.

References

  1. 1. Inouye SK, Westendorp RG, Saczynski JS. Delirium in elderly people. Lancet. 2014;383(9920):911–22. http://dx.doi.org/10.1016/s0140-6736(13)60688-1 pmid:23992774
  2. 2. Witlox J, Eurelings LS, de Jonghe JF, Kalisvaart KJ, Eikelenboom P, van Gool WA. Delirium in elderly patients and the risk of postdischarge mortality, institutionalization, and dementia: a meta-analysis. Jama. 2010;304(4):443–51. http://dx.doi.org/10.1001/jama.2010.1013 pmid:20664045
  3. 3. Siddiqi N, House AO, Holmes JD. Occurrence and outcome of delirium in medical in-patients: a systematic literature review. Age and ageing. 2006;35(4):350–64. http://dx.doi.org/10.1093/ageing/afl005 pmid:16648149
  4. 4. Abelha FJ, Luis C, Veiga D, Parente D, Fernandes V, Santos P, et al. Outcome and quality of life in patients with postoperative delirium during an ICU stay following major surgery. Crit Care. 2013;17(5):R257. http://dx.doi.org/10.1186/cc13084 pmid:24168808
  5. 5. Saczynski JS, Marcantonio ER, Quach L, Fong TG, Gross A, Inouye SK, et al. Cognitive trajectories after postoperative delirium. The New England journal of medicine. 2012;367(1):30–9. http://dx.doi.org/10.1056/NEJMoa1112923 pmid:22762316
  6. 6. Pandharipande PP, Girard TD, Jackson JC, Morandi A, Thompson JL, Pun BT, et al. Long-term cognitive impairment after critical illness. The New England journal of medicine. 2013;369(14):1306–16. http://dx.doi.org/10.1056/NEJMoa1301372 pmid:24088092
  7. 7. O’Mahony R, Murthy L, Akunne A, Young J. Synopsis of the National Institute for Health and Clinical Excellence guideline for prevention of delirium. Annals of internal medicine. 2011;154(11):746–51. http://dx.doi.org/10.7326/0003-4819-154-11-201106070-00006 pmid:21646557
  8. 8. Oh ES, Fong TG, Hshieh TT, Inouye SK. Delirium in Older Persons: Advances in Diagnosis and Treatment. Jama. 2017;318(12):1161–74. http://dx.doi.org/10.1001/jama.2017.12067 pmid:28973626
  9. 9. Barr J, Fraser GL, Puntillo K, Ely EW, Gelinas C, Dasta JF, et al. Clinical practice guidelines for the management of pain, agitation, and delirium in adult patients in the intensive care unit. Crit Care Med. 2013;41(1):263–306. http://dx.doi.org/10.1097/CCM.0b013e3182783b72 pmid:23269131
  10. 10. Al-Qadheeb NS, Balk EM, Fraser GL, Skrobik Y, Riker RR, Kress JP, et al. Randomized ICU trials do not demonstrate an association between interventions that reduce delirium duration and short-term mortality: a systematic review and meta-analysis. Crit Care Med. 2014;42(6):1442–54. http://dx.doi.org/10.1097/ccm.0000000000000224 pmid:24557420
  11. 11. The American Geriatrics Society Expert Panel on Postoperative Delirium in Older Adults. American Geriatrics Society abstracted clinical practice guideline for postoperative delirium in older adults. Journal of the American Geriatrics Society. 2015;63(1):142–50. http://dx.doi.org/10.1111/jgs.13281 pmid:25495432
  12. 12. Li J, Xiong M, Nadavaluru PR, Zuo W, Ye JH, Eloy JD, et al. Dexmedetomidine Attenuates Neurotoxicity Induced by Prenatal Propofol Exposure. J Neurosurg Anesthesiol. 2016;28(1):51–64. http://dx.doi.org/10.1097/ana.0000000000000181 pmid:25844953
  13. 13. Mo Y, Zimmermann AE. Role of dexmedetomidine for the prevention and treatment of delirium in intensive care unit patients. The Annals of pharmacotherapy. 2013;47(6):869–76. http://dx.doi.org/10.1345/aph.1AR708 pmid:23719785
  14. 14. Wunsch H, Kahn JM, Kramer AA, Wagener G, Li G, Sladen RN, et al. Dexmedetomidine in the care of critically ill patients from 2001 to 2007: an observational cohort study. Anesthesiology. 2010;113(2):386–94. http://dx.doi.org/10.1097/ALN.0b013e3181e74116 pmid:20613466
  15. 15. Liu X, Xie G, Zhang K, Song S, Song F, Jin Y, et al. Dexmedetomidine vs propofol sedation reduces delirium in patients after cardiac surgery: A meta-analysis with trial sequential analysis of randomized controlled trials. Journal of critical care. 2017;38:190–6. http://dx.doi.org/10.1016/j.jcrc.2016.10.026 pmid:27936404
  16. 16. Geng J, Qian J, Cheng H, Ji F, Liu H. The Influence of Perioperative Dexmedetomidine on Patients Undergoing Cardiac Surgery: A Meta-Analysis. PLoS One. 2016;11(4):e0152829. http://dx.doi.org/10.1371/journal.pone.0152829 pmid:27049318
  17. 17. Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gotzsche PC, Ioannidis JP, et al. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate healthcare interventions: explanation and elaboration. BMJ (Clinical research ed). 2009;339:b2700. http://dx.doi.org/10.1136/bmj.b2700 pmid:19622552
  18. 18. Higgins JP, Altman DG, Gotzsche PC, Juni P, Moher D, Oxman AD, et al. The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials. BMJ (Clinical research ed). 2011;343:d5928. http://dx.doi.org/10.1136/bmj.d5928 pmid:22008217
  19. 19. Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ (Clinical research ed). 2003;327(7414):557–60. http://dx.doi.org/10.1136/bmj.327.7414.557 pmid:12958120
  20. 20. Chen J, Yan J, Han X. Dexmedetomidine may benefit cognitive function after laparoscopic cholecystectomy in elderly patients. Exp Ther Med. 2013;5(2):489–94. http://dx.doi.org/10.3892/etm.2012.811 pmid:23403854
  21. 21. Kim DJ, Kim SH, So KY, Jung KT. Effects of dexmedetomidine on smooth emergence from anaesthesia in elderly patients undergoing orthopaedic surgery. BMC anesthesiology. 2015;15:139. http://dx.doi.org/10.1186/s12871-015-0127-4 pmid:26446479
  22. 22. Lee CW, Kim M. Effects of preanesthetic dexmedetomidine on hemodynamic responses to endotracheal intubation in elderly patients undergoing treatment for hypertension: a randomized, double-blinded trial. Korean journal of anesthesiology. 2017;70(1):39–45. http://dx.doi.org/10.4097/kjae.2017.70.1.39 pmid:28184265
  23. 23. Mansouri N, Nasrollahi K, Shetabi H. Prevention of Cognitive Dysfunction after Cataract Surgery with Intravenous Administration of Midazolam and Dexmedetomidine in Elderly Patients Undergoing Cataract Surgery. Advanced biomedical research. 2019;8:6. http://dx.doi.org/10.4103/abr.abr_190_18 pmid:30820427
  24. 24. Zhang DF, Su X, Meng ZT, Li HL, Wang DX, Li XY, et al. Impact of Dexmedetomidine on Long-term Outcomes After Noncardiac Surgery in Elderly: 3-Year Follow-up of a Randomized Controlled Trial. Annals of surgery. 2018. http://dx.doi.org/10.1097/sla.0000000000002801 pmid:29742525
  25. 25. Zhan-Ying G, Chang-Ming W, Shuai T, Lin-Lin T, Yu-Feng H. Comparison of Effects of Different Doses Dexmedetomidine on Inhibiting Tracheal Intubation-Evoked Haemodynamic Response in the Elderly Patients. Journal of clinical and diagnostic research: JCDR. 2015;9(9):Uc10–3. http://dx.doi.org/10.7860/jcdr/2015/14624.6455 pmid:26500982
  26. 26. Li Y, He R, Chen S, Qu Y. Effect of dexmedetomidine on early postoperative cognitive dysfunction and peri-operative inflammation in elderly patients undergoing laparoscopic cholecystectomy. Experimental and therapeutic medicine. 2015;10(5):1635–42. http://dx.doi.org/10.3892/etm.2015.2726 pmid:26640530
  27. 27. Cheng XQ, Mei B, Zuo YM, Wu H, Peng XH, Zhao Q, et al. A multicentre randomised controlled trial of the effect of intra-operative dexmedetomidine on cognitive decline after surgery. Anaesthesia. 2019. http://dx.doi.org/10.1111/anae.14606 pmid:30835822
  28. 28. Deiner S, Luo X, Lin HM, Sessler DI, Saager L, Sieber FE, et al. Intraoperative Infusion of Dexmedetomidine for Prevention of Postoperative Delirium and Cognitive Dysfunction in Elderly Patients Undergoing Major Elective Noncardiac Surgery: a Randomized Clinical Trial. JAMA surgery. 2017;152(8):e171505. http://dx.doi.org/10.1001/jamasurg.2017.1505 pmid:28593326
  29. 29. Lee C, Lee CH, Lee G, Lee M, Hwang J. The effect of the timing and dose of dexmedetomidine on postoperative delirium in elderly patients after laparoscopic major non-cardiac surgery: a double blind randomized controlled study. Journal of clinical anesthesia. 2018;47:27–32. http://dx.doi.org/10.1016/j.jclinane.2018.03.007 pmid:29549829
  30. 30. Liu Y, Ma L, Gao M, Guo W, Ma Y. Dexmedetomidine reduces postoperative delirium after joint replacement in elderly patients with mild cognitive impairment. Aging clinical and experimental research. 2016;28(4):729–36. http://dx.doi.org/10.1007/s40520-015-0492-3 pmid:26559412
  31. 31. Su X, Meng ZT, Wu XH, Cui F, Li HL, Wang DX, et al. Dexmedetomidine for prevention of delirium in elderly patients after non-cardiac surgery: a randomised, double-blind, placebo-controlled trial. Lancet (london, england). 2016;388(10054):1893–902. http://dx.doi.org/10.1016/S0140-6736(16)30580-3.
  32. 32. Wu XH, Cui F, Zhang C, Meng ZT, Wang DX, Ma J, et al. 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(5):979–91. http://dx.doi.org/10.1097/ALN.0000000000001325 pmid:27571256
  33. 33. De J, Wand AP. Delirium Screening: A Systematic Review of Delirium Screening Tools in Hospitalized Patients. The Gerontologist. 2015;55(6):1079–99. http://dx.doi.org/10.1093/geront/gnv100 pmid:26543179
  34. 34. Luetz A, Heymann A, Radtke FM, Chenitir C, Neuhaus U, Nachtigall I, et al. Different assessment tools for intensive care unit delirium: which score to use? Crit Care Med. 2010;38(2):409–18. http://dx.doi.org/10.1097/CCM.0b013e3181cabb42 pmid:20029345
  35. 35. Muller A, Lachmann G, Wolf A, Morgeli R, Weiss B, Spies C. Peri- and postoperative cognitive and consecutive functional problems of elderly patients. Current opinion in critical care. 2016;22(4):406–11. http://dx.doi.org/10.1097/mcc.0000000000000327 pmid:27272100
  36. 36. Forsberg A, Cervenka S, Jonsson Fagerlund M, Rasmussen LS, Zetterberg H, Erlandsson Harris H, et al. The immune response of the human brain to abdominal surgery. Annals of neurology. 2017;81(4):572–82. http://dx.doi.org/10.1002/ana.24909 pmid:28253549
  37. 37. Clark IA, Vissel B. The Inflammatory Nature of Post-surgical Delirium Predicts Benefit of Agents With Anti-TNF Effects, Such as Dexmedetomidine. Frontiers in neuroscience. 2018;12:257. http://dx.doi.org/10.3389/fnins.2018.00257 pmid:29725287
  38. 38. Xiang H, Hu B, Li Z, Li J. Dexmedetomidine controls systemic cytokine levels through the cholinergic anti-inflammatory pathway. Inflammation. 2014;37(5):1763–70. http://dx.doi.org/10.1007/s10753-014-9906-1 pmid:24803295
  39. 39. Yang YF, Peng K, Liu H, Meng XW, Zhang JJ, Ji FH. Dexmedetomidine preconditioning for myocardial protection in ischaemia-reperfusion injury in rats by downregulation of the high mobility group box 1-toll-like receptor 4-nuclear factor kappaB signalling pathway. Clin Exp Pharmacol Physiol. 2017;44(3):353–61. http://dx.doi.org/10.1111/1440-1681.12711 pmid:27998004
  40. 40. Kim E, Kim HC, Lee S, Ryu HG, Park YH, Kim JH, et al. Dexmedetomidine confers neuroprotection against transient global cerebral ischemia/reperfusion injury in rats by inhibiting inflammation through inactivation of the TLR-4/NF-kappaB pathway. Neuroscience letters. 2017;649:20–7. http://dx.doi.org/10.1016/j.neulet.2017.04.011 pmid:28392361
  41. 41. Duan X, Coburn M, Rossaint R, Sanders RD, Waesberghe JV, Kowark A. Efficacy of perioperative dexmedetomidine on postoperative delirium: systematic review and meta-analysis with trial sequential analysis of randomised controlled trials. Br J Anaesth. 2018;121(2):384–97. http://dx.doi.org/10.1016/j.bja.2018.04.046 pmid:30032877
  42. 42. 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):2644–53. http://dx.doi.org/10.1001/jama.298.22.2644 pmid:18073360
  43. 43. Riker RR, Shehabi Y, Bokesch PM, Ceraso D, Wisemandle W, Koura F, et al. Dexmedetomidine vs midazolam for sedation of critically ill patients: a randomized trial. Jama. 2009;301(5):489–99. http://dx.doi.org/10.1001/jama.2009.56 pmid:19188334
  44. 44. Xia ZQ, Chen SQ, Yao X, Xie CB, Wen SH, Liu KX. Clinical benefits of dexmedetomidine versus propofol in adult intensive care unit patients: a meta-analysis of randomized clinical trials. The Journal of surgical research. 2013;185(2):833–43. http://dx.doi.org/10.1016/j.jss.2013.06.062 pmid:23910886
  45. 45. Kahraman S, Zup SL, McCarthy MM, Fiskum G. GABAergic mechanism of propofol toxicity in immature neurons. J Neurosurg Anesthesiol. 2008;20(4):233–40. http://dx.doi.org/10.1097/ANA.0b013e31817ec34d pmid:18812886
  46. 46. Kamibayashi T, Maze M. Clinical uses of alpha2 -adrenergic agonists. Anesthesiology. 2000;93(5):1345–9. pmid:11046225
  47. 47. Nguyen V, Tiemann D, Park E, Salehi A. Alpha-2 Agonists. Anesthesiology clinics. 2017;35(2):233–45. http://dx.doi.org/10.1016/j.anclin.2017.01.009 pmid:28526145
  48. 48. Kunisawa T, Nagata O, Nagashima M, Mitamura S, Ueno M, Suzuki A, et al. Dexmedetomidine suppresses the decrease in blood pressure during anesthetic induction and blunts the cardiovascular response to tracheal intubation. J Clin Anesth. 2009;21(3):194–9. http://dx.doi.org/10.1016/j.jclinane.2008.08.015 pmid:19464613
  49. 49. Bajwa SJ, Kaur J, Singh A, Parmar S, Singh G, Kulshrestha A, et al. Attenuation of pressor response and dose sparing of opioids and anaesthetics with pre-operative dexmedetomidine. Indian J Anaesth. 2012;56(2):123–8. http://dx.doi.org/10.4103/0019-5049.96303 pmid:22701201
  50. 50. Smilowitz NR, Gupta N, Guo Y, Berger JS, Bangalore S. Perioperative acute myocardial infarction associated with non-cardiac surgery. European heart journal. 2017;38(31):2409–17. http://dx.doi.org/10.1093/eurheartj/ehx313 pmid:28821166
  51. 51. Hood R, Budd A, Sorond FA, Hogue CW. Peri-operative neurological complications. Anaesthesia. 2018;73 Suppl 1:67–75. http://dx.doi.org/10.1111/anae.14142 pmid:29313909
  52. 52. Duncan D, Sankar A, Beattie WS, Wijeysundera DN. Alpha-2 adrenergic agonists for the prevention of cardiac complications among adults undergoing surgery. The Cochrane database of systematic reviews. 2018;3:Cd004126. http://dx.doi.org/10.1002/14651858.CD004126.pub3 pmid:29509957
  53. 53. Venn RM, Hell J, Grounds RM. Respiratory effects of dexmedetomidine in the surgical patient requiring intensive care. Crit Care. 2000;4(5):302–8. http://dx.doi.org/10.1186/cc712 pmid:11056756