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

A comparation of dexmedetomidine and midazolam for sedation in patients with mechanical ventilation in ICU: A systematic review and meta-analysis

  • Jiaxuan Wen,

    Roles Conceptualization, Data curation, Formal analysis, Writing – original draft, Writing – review & editing

    Affiliation School of Nursing, Weifang Medical University, Weifang, P. R. China

  • Xueying Ding ,

    Contributed equally to this work with: Xueying Ding

    Roles Conceptualization, Data curation

    Affiliation School of Nursing, Weifang Medical University, Weifang, P. R. China

  • Chen Liu,

    Roles Formal analysis, Methodology, Supervision

    Affiliation School of Nursing, Weifang Medical University, Weifang, P. R. China

  • Wenyu Jiang,

    Roles Formal analysis

    Affiliation School of Public Health, Weifang Medical University, Weifang, P. R. China

  • Yingrui Xu,

    Roles Formal analysis

    Affiliation School of Nursing, Weifang Medical University, Weifang, P. R. China

  • Xiuhong Wei,

    Roles Methodology, Supervision

    Affiliation School of Nursing, Weifang Medical University, Weifang, P. R. China

  • Xin Liu

    Roles Data curation, Writing – review & editing

    xinliuwf@163.com

    Affiliation Department of Neonatology, Weifang People’s Hospital, P. R. China

Abstract

Background

The use of dexmedetomidine rather than midazolam may improve ICU outcomes. We summarized the available recent evidence to further verify this conclusion.

Methods

An electronic search of PubMed, Medline, Embase, Cochrane Library, and Web of Science was conducted. Risk ratios (RR) were used for binary categorical variables, and for continuous variables, weighted mean differences (WMD) were calculated, the effect sizes are expressed as 95% confidence intervals (CI), and trial sequential analysis was performed.

Results

16 randomized controlled trials were enrolled 2035 patients in the study. Dexmedetomidine as opposed to midazolam achieved a shorter length of stay in ICU (MD = -2.25, 95%CI = -2.94, -1.57, p<0.0001), lower risk of delirium (RR = 0.63, 95%CI = 0.50, 0.81, p = 0.0002), and shorter duration of mechanical ventilation (MD = -0.83, 95%CI = -1.24, -0.43, p<0.0001). The association between dexmedetomidine and bradycardia was also found to be significant (RR 2.21, 95%CI 1.31, 3.73, p = 0.003). We found no difference in hypotension (RR = 1.44, 95%CI = 0.87, 2.38, P = 0.16), mortality (RR = 1.02, 95%CI = 0.83, 1.25, P = 0.87), neither in terms of adverse effects requiring intervention, hospital length of stay, or sedation effects.

Conclusions

Combined with recent evidence, compared with midazolam, dexmedetomidine decreased the risk of delirium, mechanical ventilation, length of stay in the ICU, as well as reduced patient costs. But dexmedetomidine could not reduce mortality and increased the risk of bradycardia.

Introduction

Sedation is often required for critically ill patients in the intensive care unit (ICU) to reduce patient-ventilator asynchrony and minimize delirium and agitation for comfort [1, 2]. Conventional sedatives include benzodiazepines, dexmedetomidine, propofol, and opioids [3], of which benzodiazepines are gamma aminobutyrate receptor agonists of the central nervous system and have anti-anxiety, amnesia, sedation, hypnosis, and anticonvulsant effects [4]. As the most common drug in this class, midazolam has the characteristic of rapid onset and short duration [5]. As a selective Alpha-2 adrenergic receptor agonist, by inhibiting the release of norepinephrine from the nucleus coeruleus and competitively binding to the Alpha-2 receptor, dexmedetomidine plays a role in relieving calming, antianxiety and mild analgesic sedation [6].

Although the recent international guidelines offered a conditional recommendation favoring the use of dexmedetomidine over benzodiazepines in mechanically ventilated adults [3], in real clinical practice, midazolam is still frequently used [710], especially when the COVID-19 pandemic caused a shortage of medicines [11]. Besides, this recommendation was based on studies published between 2007 and 2015 [12, 13], and the quality of some of these articles was questionable. Because of the low level of evidence, international guidelines issued only a conditional recommendation [3, 14]. Some previous analysis suggested that dexmedetomidine provided an advantage over midazolam in terms of shorter sedation duration and fewer adverse events over midazolam, the low quality of inclusion articles [15] or few outcome measures [16] prompted us to update our analysis. In addition, although previous studies confirmed that dexmedetomidine was superior to benzodiazepines for mechanical ventilation in the ICU [17, 18], these previous researches included studies comparing dexmedetomidine to lorazepam. As a medium-effect benzodiazepine central nervous system inhibitor, the clinical pharmacology of lorazepam is significantly different from that of midazolam. Even short-term injection of lorazepam for sedation may lead to significant delays in extubation and discharge of ICU patients [19]. Thus, this comparison may overstate the adverse effects of midazolam.

Based on the latest evidence, we attempted to expand the previous analysis to comprehensively analyze the efficacy and adverse effects of the two sedatives. Moreover, we attempted to determine whether the results reached the size of the information required to draw conclusions in conjunction with the trial sequence analysis.

Materials and methods

We followed the PRISMA statement and the guidelines in the Cochrane Handbook for reporting systematic reviews and meta-analyses in order to ensure the reliability and authenticity of the results. [20, 21]. Our meta-analysis had been registered with the PROSPERO database with registration number CRD42022379612. The PRISMA checklist is shown in S1 Table.

Electronic search strategy

A search of the databases Medline, Embase, PubMed, Cochrane Library, Web of Science, EBSCO and Scopus was performed by two researchers. The search identified relevant articles without language restriction from inception to October 15th, 2023. During the electronic database search, Among the MeSH terms and keywords were "dexmedetomidine", "midazolam", "mechanical ventilation" and "intensive care". To obtain additional trials of high relevance and to ensure the accuracy of subsequent analysis, we filtered the additional research in the reference list of relevant articles.

Eligibility criteria

Only randomized controlled trials published as full articles were included without language restrictions. These randomized controlled trials adhered to the following criteria: (a) patients over the age of 18, (b) admitted to the ICU requiring mechanical ventilation, (c) clinically needed sedation, (e) and received dexmedetomidine in the intervention group and midazolam with or without other sedatives in the control group. (f) trials with outcome indicators included: delirium, intensive care unit length of stay, duration of mechanical ventilation, hospital length of stay, all-cause mortality, proportion of time at target sedation, additional sedatives use, and adverse events including bradycardia, hypotension, hypertension, tachycardia.

Data extraction and analysis

Data were extracted and cross-checked by two researchers after reviewing the literature independently. If there was a disagreement, it should be resolved through discussion or consultation with a third reviewer. A designed data form was used to extract and manage data from all qualified literature. We collected data on title, author, year of publication, patient age, gender, number of cases, acute physiology and chronic health evaluation (APACHE) II, target sedation, drug dose and outcome indicators. Trials that report a continuous outcome as a median and dispersion were converted into a mean and standard deviation [22, 23]. To obtain missing data from a study, the original author was contacted via phone or email.

Literature quality assessment

As per the recommendations in the Cochrane Handbook for the Systematic Review of Interventions 5.1.0 [24], two reviewers independently and in duplicate assessed trials for bias. We resolved disagreements through discussion and consensus. Among the contents assessed are the randomization method, the allocation concealment design, the blinding methods used, integrity of the results report, selective reporting of outcome and the existence of other sources of bias, etc. Following are the results: An answer of "Yes" indicated correct methodology or complete data, indicating a low risk of bias; "unclear" indicated a medium risk of bias; and "No" indicated a high risk of bias. The article was considered low risk of bias if the bias in each domain was low, otherwise it was regarded as high risk. A risk of bias assessment plot was exported using RevMan 5.4 software.

Statistical analysis

Revman5.4 was used to conduct data analyses. During the meta-analysis, we used the DerSimonian and Laird random-effect models [25]. For dichotomous outcomes, we calculated pooled relative risks (RR), for continuous outcomes, we calculated mean differences (MD), and we calculated corresponding confidence interval (CI) of 95%. In order to assess the statistical heterogeneity, the I2 index was calculated, low heterogeneity was defined as I2<50% [26]. Publication bias was checked through visual inspection of funnel plots and by Egger’s or Egger’s tests using STATA software V.16.0. [27, 28].

In order to examine whether baseline factors influenced treatment effects and explore heterogeneity between trials, we performed a subgroup analysis. Subgroup analyses were conducted for the outcome by age of participants, region, as well as sedation duration. We conducted meta-regression analyses in Stata 16.0 to examine the relationship between delirium and the mean age of patients. To evaluate whether the results were robust, we performed a sensitivity analysis by excluding studies with small sample sizes (n<100) or studies with a high risk of bias.

To determine if the required sample size for statistical significance was met, we performed trial sequential analysis (TSA) using TSA software v. 0.9.5.10 Beta [29]. A conclusion could be considered firmly accepted or refuted when the required information size (RIS) was larger than accrued information size (AIS) or the cumulative z-curve crossed the sequential monitoring boundary. Otherwise, further trials were needed to verify the results. The type-I error (Alpha-2) and power were set as 0.05 and 0.80, respectively. The relative risk reduction in analysis of All dichotomous outcomes is 30%, for continuous data, the “empirical” item or minimal difference was set for MD and variance, and the “model-based variance” item was applied.

GRADE (Grading of Recommendations Assessment, Development, and Evaluation) was used by two reviewers in evaluating the quality of evidence [30]. GRADEpro software was used to estimate the quality of our results on the basis of risk of bias, inconsistency, indirectness, imprecision, and publication bias. The quality of the results was categorized into four grades: high, moderate, low, or very low [31].

Results

Search results

A preliminary search identified 788 citations (Pubmed:42; Embase:124; Cochrane Library:86; Web of Science:109; EBSCO:307; Scopus:120), and 549 records remained after the removal of duplicate records. We screened the titles and abstracts. A further 524 records were removed after that. The full-text of 25 articles was carefully read for eligibility. The full-text of 9 articles was excluded for a variety of reasons. Finally, 16 trials were included in our analysis. The overview of the search strategy is shown in the S2 Table in S1 File. Fig 1 represents the PRISMA flow chart of the search process.

Study characteristics

Overall, 16 eligible trials enrolled a total of 2035 patients. Table 1 describes the baseline characteristics of included literature. These trials were published between 2009 and 2022. Study participants ranged from 23 to 500. In all cases, patients required mechanical ventilation in the intensive care unit. The overall mean age of patients was 60.52±15.99 years. An estimated 40.3% of participants were female. And the mean APACHE II score reported in 14 trials was 19.88±7.28. 9 of the studies were conducted in Asian countries, three in the United States, three in European countries and one in Australia. 11 trials showed sedation lasting longer than 24 hours. A maximum rate of 0.7μg/kg/h was used for infusion of dexmedetomidine in most trials, and the infusion rate of midazolam varied greatly among different trials. All the trials maintained a light level of sedation although the evaluation methods were different.

Explanations

1. Duration of Mechanical Ventilation; 2. ICU Length of Stay; 3. Delirium; 4. Hypotension; 5. Bradycardia; 6. Mortality; 7. Hospital Length of Stay; 8. Proportion of Time at Target Sedation; 9. Percent of Patients that Required Supplemental Sedatives; 10. Hypertension; 11. Tachycardia;12. ICU cost

Risk of bias

7 trials were found to be low risk of bias according to the Cochrane risk of bias tool. There was a moderate level of quality among the included trials. We evaluated the mentioned-above items. An inadequate method of random sequence generation was applied in 1 trial and conceal allocation in 1 trial was improper. Patient or caregiver blinding was absent in 3 trials, and 4 did not follow a blinding procedure for outcome assessment. We deemed two trials to have incomplete results. Risk of bias assessment details are shown in S1 Fig in S1 File.

Meta-analysis

Meta-analyses were performed for the intensive Care Unit Length of Stay, Duration of Mechanical Ventilation, delirium, bradycardia, hypotension, mortality and some other outcomes. In the following paragraphs, we present the results for effect sizes comparing the dexmedetomidine and midazolam.

ICU length of stay

A total of 13 studies with 1779 patients were analyzed. There was relatively large heterogeneity (I2 = 53%, p = 0.01). The use of dexmedetomidine can reduce the ICU length of stay compared to midazolam (MD = -2.25 days, 95%CI = -2.94, -1.57, p<0.0001, moderate certainty, S1 Table in S1 File and Fig 2A). The sensitivity analyses excluding high risk of bias or small sample size studies, as well as the subgroup analysis, indicated no effect modification, which proved the robustness of our results (Fig 3A and S12-S17 Figs in S1 File). In trial sequential analysis, the AIS was larger than the RIS. The cumulative Z curves crossed both the conventional boundary and the benefit boundary, so the conclusion is conclusive and favors dexmedetomidine (Fig 4A). By employing Egger’s test, publication bias was not detected (P = 0.938; S2 Fig in S1 File).

Delirium

12 studies (n = 1738) explored the difference between dexmedetomidine and midazolam in the incidence of delirium. The studies displayed statistical heterogeneity (I2 = 51%, p = 0.03). When dexmedetomidine was used instead of midazolam, the percentage of delirious patients was lower (RR = 0.63, 95%CI = 0.50, 0.81, p = 0.0002, moderate certainty, S1 Table in S1 File and Fig 2C). The results of the subgroup analysis and sensitivity analysis suggested no effect modification (Fig 3C and S18-S23 Figs in S1 File). Additionally, we performed a meta-regression analysis to determine the risk of delirium based on age, but our results did not reach statistical significance (P = 0.213, S3 Fig in S1 File). Although the cumulative Z curves in TSA showed that the required amount of information was not enough, the Z curve crossed traditional and benefit boundaries, indicating a true positive result in favor of dexmedetomidine usage (Fig 4C). This analysis did not exhibit publication bias according to Egger’s test (P = 0.246, S2C Fig in S1 File).

Duration of mechanical ventilation

Two sedatives were compared in 11 trials (1712 patients) in terms of duration of mechanical ventilation. The I2 of 97% indicated substantial heterogeneity. Our analysis found that dexmedetomidine was associated with shorter mechanical ventilation (MD = -0.83 h, 95%CI = -1.24, -0.43, p<0.0001, very low certainty, S1 Table in S1 File and Fig 2B). It was unclear whether dexmedetomidine would reduce the duration of mechanical ventilation among long-term sedation patients or North Americans in the subgroup analysis. Significant subgroup differences were detected in subgroup analyses by age and APACHE II, but the results of the subgroup were consistent with those of the primary analysis. Sensitivity analyses also did not alter the conclusions (Fig 3C and S24-S29 Figs in S1 File). The cumulative Z curve exceeded the monitoring boundary despite the information size not reaching the required level, so the outcome of TSA demonstrated that the result was conclusive (Fig 4B). With Egger’s test, there were no indications of publication bias (P = 0.200; S2B Fig in S1 File).

Bradycardia

The pooled estimate of 10 RCTs (n = 1578 patients) showed that a high risk of bradycardia was associated with dexmedetomidine when compared to midazolam (RR 2.21, 95% CI 1.31, 3.73, p = 0.003, high certainty, S1 Table in S1 File and Fig 2D). The heterogeneity among studies was low (I2 = 42%, p = 0.08). Neither subgroup analysis nor sensitivity analysis suggested effect modification (Fig 3D and S30-S35 Figs in S1 File). While the cumulative Z curve did not reach the required information size, it crossed both the conventional and harm boundaries, concluding the TSA conclusion. (Fig 4D). Egger’s test revealed no publication bias (P = 0.507, S2D Fig in S1 File). However, according to the analysis of two trials (n = 428), there was no increased risk of bradycardia requiring intervention (RR 1.98, 95% CI 0.14, 28.72, S1 Table in S1 File and S10 Fig in S1 File).

Hypotension

In 9 studies (n = 1427 patients), hypotension was included in their evaluation index. We observed heterogeneity between studies (I2 = 64%, p = 0.005). Meta analysis indicated that dexmedetomidine did not increase the risk of hypotension (RR = 1.39, 95%CI = 0.84, 2.32, P = 0.2, low certainty, S1 Table in S1 File and Fig 2E). Subgroup analysis and sensitivity analysis did not alter the primary conclusions (Fig 3E and S36-S41 Figs in S1 File). TSA conclusion is inconclusive, since the cumulative Z curves did not cross any boundaries and the AIS is smaller than the RIS, so further study is necessary to uncover firm evidence (Fig 4E). The Egger’s test (P = 0.239) indicated no publication bias (S2E Fig in S1 File). The results from 5 trials (n = 609) showed no statistically significant increase in the risk of hypotension requiring intervention (RR 1.45, 95% CI 0.70, 3.00; S9 Fig in S1 File).

Mortality

To explore the difference in mortality between dexmedetomidine and midazolam, we comprehensively analyzed 9 literatures (n = 1662 patients) Heterogeneity among these studies was low (I2 = 0%, p = 0.48). Our analysis revealed that compared with midazolam, dexmedetomidine did not show an advantage in reducing patient mortality (RR = 0.99, 95%CI = 0.81, 1.21, P = 0.92, high certainty, S1 Table in S1 File and Fig 2F). According to the results of the subgroup and sensitivity analyses, the primary analysis was reliable (Fig 3F and S42-S47 Figs in S1 File). The TSA conclusion is inconclusion. The Z curves crossed no boundaries, so further research may be needed to confirm this conclusion (Fig 4F). Based on Egger’s test (P = 0.325), publication bias was not detected (S2F Fig in S1 File).

Other outcomes

4 trials (n = 972) included the outcome of proportion of time at target sedation. Dexmedetomidine was the same as midazolam in terms of sedative effects (MD = -0.91, 95%CI = -3.90, 5.72, p = 0.71, low certainty, S1 Table in S1 File and S4 Fig in S1 File). This had also been demonstrated with regard to the outcome of using additional sedatives in 6 trials (n = 1149, RR = 1.04, 95%CI = 0.86, 1.26, p = 0.67, low certainty, S1 Table in S1 File and S5 Fig in S1 File). 4 trials included the length of the hospital stay(n = 747), there was no difference in the effects of the two sedatives on this outcome (MD = 0.11, 95%CI = -5.90, 6.11, p = 0.97, very low certainty, S1 Table in S1 File and S8 Fig in S1 File). 3 trials (n = 1016) reported hypertension. As with hypotension, the incidence of hypertension did not differ between the two sedatives (n = 1016, RR = 0.98, 95%CI = 0.74, 1.29, p = 0.87, low certainty, S1 Table in S1 File and S7 Fig in S1 File). However, midazolam was associated with an increased risk of tachycardia, according to the reports in four trials (n = 1039, RR = 0.73, 95%CI = 0.51, 1.04, p = 0.08, S1 Table in S1 File and S6 Fig in S1 File).

Discussion

Sedation has always been a challenge in mechanical ventilation in the ICU. Our meta-analysis combined the latest evidence to analyze the merits of dexmedetomidine versus midazolam in critically ill patients, using trial sequence analysis and a GEADE system to verify the reliability of our results. Based on the results of our systematic review and meta-analysis of 16 trials including 1,998 critically ill patients, we find high certainty evidence that dexmedetomidine increases bradycardia risk compared with midazolam. It was moderately certain that dexmedetomidine reduced ICU hospitalization and delirium incidences, but did not improve survival. Low evidence indicated that it did not increase hypotension risks. Evidence that dexmedetomidine reduced the duration of mechanical ventilation was of very low certainty.

In patients with delirium, prolonged mechanical ventilation, hospital stays, increased medical costs and mortality [5052]. There is a significant increase in the incidence of cognitive impairment during long-term follow-up of patients with delirium [53]. PADIS noted that benzodiazepine use is a modifiable factor associated with delirium [3], but several recent studies have also shown that there does not appear to be a relationship between short-acting benzodiazepines and the incidence of delirium [5456]. Previous studies have also indicated that midazolam seems to be unrelated to the incidence of delirium [57]. Interestingly, midazolam had also been reported to have a brain protective effect [58, 59]. In contrast to the embarrassment of midazolam, dexmedetomidine was widely believed to have delirium prevention effects [17]. In fact, this was recently questioned in a large, randomized clinical trials [60]. We compared the two sedatives in our meta-analysis. Moderate evidence was found that patients on dexmedetomidine did have a lower incidence of delirium than on midazolam. Subgroup analysis by region, APACHE II score and duration of sedation suggested no effect modification. The delirium prophylactic effect of dexmedetomidine may relate to these aspects. Dexmedetomidine leads to a lower depth of sedation [61], provides a more natural sleep-like sedation pattern [62]. However, in the subgroup analyses by age, dexmedetomidine showed a more obvious advantage over midazolam in patients aged under 60 years old. In the meta-regression analysis, we found that neither age nor baseline APACHE II score was associated with delirium efficacy, which is similar to recent studies [63, 64]. We have the following assumptions for this result. PADIS points out that increasing age is a nonmodifiable risk factor for an association with delirium [3]. Previous studies also indicated that midazolam was a risk factor for delirium. Therefore, in the face of these patients, caregivers may increase the priority of delirium monitoring and prevention, thus reducing the incidence of delirium in the midazolam group. In particular, the odds of delirium are similar between non-benzodiazepines and intermittent bolus dosing benzodiazepines [65].Many studies have also shown that delirium prevention will depend on the implementation of non-drug interventions, which have shown the greatest potential for success [6668]. A comfortable patient without psychosis needs a suitable balance between pain medication synergy and other effective modes of non-medication management, anti-anxiety, and restorative sleep, such as family involvement. Delirium should not be a problem in the ICU in the future [69].

Sedative choice can have a potential impact on overall cost per episode [70]. In the present study, Dexmedetomidine on ICU cost showed cost-effectiveness. Although dexmedetomidine is more expensive than midazolam, we observed statistically significant reductions in ICU length of stay and mechanical ventilation time in patients sedated with dexmedetomidine. Subgroup analysis and sensitivity analysis proved that the results were reliable. Therefore, the total cost was cheaper instead. Prolonged mechanical ventilation or ICU hospitalization increased the risk of delirium and other adverse events, and increased the financial burden and emotional stress of the patient. Post-intensive care syndrome (PICS) seriously reduces the quality of life after patient discharge [71]. Negative ICU experiences and delirium were considered major risk factors [72]. Dexmedetomidine can improve sleep quality [61], decrease rates of psychological impairment, and thereby significantly lower rates of PICS [73].

Our meta-analysis found no difference between two sedatives in maintaining patient target sedation rates and no difference in the need for additional sedatives. However, dexmedetomidine has the biphasic effects of temporarily raising blood pressure through the transient constriction of peripheral blood vessels [74]. When administered in a slow manner, sedation was induced by the activation of the Alpha-2 receptor. Inhibition of sympathetic nerve activity and an increase in cardiac vagus nerve activity may lead to hypotension and bradycardia [75]. Our meta-analysis suggested that dexmedetomidine results in little to no difference from midazolam in hypotension, but there is an increasing tendency. A higher risk of bradycardia was associated with dexmedetomidine. However, there was no difference in hypotension and bradycardia requiring intervention, which was consistent with previous studies [17, 76]. Subgroup analysis and sensitivity analysis also demonstrated no modification. Furthermore, previous studies had shown that dexmedetomidine has a higher risk of hypertension than midazolam [77], nor was it observed in our meta-analysis. Although midazolam is thought to have a negligible effect on hemodynamics [78]. We found an increased tendency to tachycardia in the midazolam group. Although dexmedetomidine may increase the risk of hypotension and bradycardia, it is closely related to the loading dose and infusion speed [79, 80]. Therefore, these adverse reactions can be prevented and reversed. In fact, many clinicians tend to avoid loading dose administration, especially in critically ill patients [81]. Therefore, the correct dose and method of dexmedetomidine can completely avoid bradycardia and hypotension.

Several limitations were also observed in this meta-analysis. First, we wanted to include the risk of self-extubation and delirium-free days as our outcome indicators, but these outcomes were not described in the included RCTS. There were too few trials describing some outcomes, which affects the level of evidence for these outcomes, and we cannot detect publication bias. Due to the high heterogeneity of some outcome indicators, the certainty of outcome is reduced. Second, most of the ICU types in the trial were mixed, so the subgroup analysis we planned to compare the medical and surgical populations was not available. Some subgroup results lacked individual patient data, and we were unable to conduct meta-regression of APACHE II score and patient weight. Due to the small number of contribution trials and participants, there was insufficient persuasion for specific subgroups.

Conclusion

In conclusion, combined with evidence from recent studies, it suggests that dexmedetomidine is comparable to midazolam in sedation, with lower delirium, shorter mechanical ventilation duration, and shorter ICU stay. In spite of this, it may increase the risk of adverse events such as hypotension and bradycardia, and does not improve patient survival.

References

  1. 1. Hughes CG, McGrane S, Pandharipande PP. Sedation in the intensive care setting. Clinical pharmacology: advances and applications. 2012;4:53–63. Epub 2012/12/04. pmid:23204873; PubMed Central PMCID: PMC3508653.
  2. 2. Mehta S, McCullagh I, Burry L. Current sedation practices: lessons learned from international surveys. Anesthesiol Clin. 2011;29(4):607–24. Epub 2011/11/15. pmid:22078912.
  3. 3. Devlin JW, Skrobik Y, Gélinas C, Needham DM, Slooter AJC, Pandharipande PP, 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(9):e825–e73. Epub 2018/08/17. pmid:30113379.
  4. 4. Young CC, Prielipp RC. Benzodiazepines in the intensive care unit. Crit Care Clin. 2001;17(4):843–62. Epub 2002/01/05. pmid:11762264.
  5. 5. Fragen RJ. Pharmacokinetics and pharmacodynamics of midazolam given via continuous intravenous infusion in intensive care units. Clin Ther. 1997;19(3):405–19; discussion 367–8. Epub 1997/05/01. pmid:9220206.
  6. 6. Keating GM. Dexmedetomidine: A Review of Its Use for Sedation in the Intensive Care Setting. Drugs. 2015;75(10):1119–30. Epub 2015/06/13. pmid:26063213.
  7. 7. Wang J, Peng ZY, Zhou WH, Hu B, Rao X, Li JG. A National Multicenter Survey on Management of Pain, Agitation, and Delirium in Intensive Care Units in China. Chinese medical journal. 2017;130(10):1182–8. Epub 2017/05/10. pmid:28485318; PubMed Central PMCID: PMC5443024.
  8. 8. Kotfis K, Zegan-Barańska M, Żukowski M, Kusza K, Kaczmarczyk M, Ely EW. Multicenter assessment of sedation and delirium practices in the intensive care units in Poland—is this common practice in Eastern Europe? BMC anesthesiology. 2017;17(1):120. Epub 2017/09/04. pmid:28865447; PubMed Central PMCID: PMC5581441.
  9. 9. Talsi O, Kiiski Berggren R, Johansson G, Winsö O. A national survey on routines regarding sedation in Swedish intensive care units. Ups J Med Sci. 2019;124(3):199–202. Epub 2019/05/24. pmid:31119971; PubMed Central PMCID: PMC6758647.
  10. 10. Richards-Belle A, Canter RR, Power GS, Robinson EJ, Reschreiter H, Wunsch H, et al. National survey and point prevalence study of sedation practice in UK critical care. Critical care (London, England). 2016;20(1):355. Epub 2016/10/30. pmid:27788680; PubMed Central PMCID: PMC5084331.
  11. 11. Chanques G, Constantin JM, Devlin JW, Ely EW, Fraser GL, Gélinas C, et al. Analgesia and sedation in patients with ARDS. Intensive Care Med. 2020;46(12):2342–56. Epub 2020/11/11. pmid:33170331; PubMed Central PMCID: PMC7653978.
  12. 12. 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. Epub 2007/12/13. pmid:18073360.
  13. 13. MacLaren R, Preslaski CR, Mueller SW, Kiser TH, Fish DN, Lavelle JC, et al. A randomized, double-blind pilot study of dexmedetomidine versus midazolam for intensive care unit sedation: patient recall of their experiences and short-term psychological outcomes. J Intensive Care Med. 2015;30(3):167–75. Epub 2013/11/15. pmid:24227448.
  14. 14. Møller MH, Alhazzani W, Lewis K, Belley-Cote E, Granholm A, Centofanti J, et al. Use of dexmedetomidine for sedation in mechanically ventilated adult ICU patients: a rapid practice guideline. Intensive care medicine. 2022;48(7):801–10. Epub 2022/05/20. pmid:35587274.
  15. 15. Zhou WJ, Liu M, Fan XP. Differences in efficacy and safety of midazolam vs. dexmedetomidine in critically ill patients: A meta-analysis of randomized controlled trial. Experimental and therapeutic medicine. 2021;21(2):156. Epub 2021/01/19. pmid:33456523; PubMed Central PMCID: PMC7791964.
  16. 16. Zhang Z, Chen K, Ni H, Zhang X, Fan H. Sedation of mechanically ventilated adults in intensive care unit: a network meta-analysis. Scientific reports. 2017;7:44979. Epub 2017/03/23. pmid:28322337; PubMed Central PMCID: PMC5359583.
  17. 17. Lewis K, Alshamsi F, Carayannopoulos KL, Granholm A, Piticaru J, Al Duhailib Z, 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(7):811–40. Epub 2022/06/02. pmid:35648198.
  18. 18. Wang S, Hong Y, Li S, Kuriyama A, Zhao Y, Hu J, et al. 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. Epub 2020/12/10. pmid:33296787.
  19. 19. Barr J, Zomorodi K, Bertaccini EJ, Shafer SL, Geller E. A double-blind, randomized comparison of i.v. lorazepam versus midazolam for sedation of ICU patients via a pharmacologic model. Anesthesiology. 2001;95(2):286–98. Epub 2001/08/17. pmid:11506097.
  20. 20. Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS medicine. 2009;6(7):e1000097. Epub 2009/07/22. pmid:19621072; PubMed Central PMCID: PMC2707599.
  21. 21. Cumpston M, Li T, Page MJ, Chandler J, Welch VA, Higgins JP, et al. Updated guidance for trusted systematic reviews: a new edition of the Cochrane Handbook for Systematic Reviews of Interventions. The Cochrane database of systematic reviews. 2019;10:Ed000142. Epub 2019/10/24. pmid:31643080.
  22. 22. Luo D, Wan X, Liu J, Tong T. Optimally estimating the sample mean from the sample size, median, mid-range, and/or mid-quartile range. Statistical methods in medical research. 2018;27(6):1785–805. Epub 2016/09/30. pmid:27683581.
  23. 23. Wan X, Wang W, Liu J, Tong T. Estimating the sample mean and standard deviation from the sample size, median, range and/or interquartile range. BMC medical research methodology. 2014;14:135. Epub 2014/12/20. pmid:25524443; PubMed Central PMCID: PMC4383202.
  24. 24. Higgins JP, Altman DG, Gøtzsche PC, Jüni 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. Epub 2011/10/20. pmid:22008217; PubMed Central PMCID: PMC3196245 at www.icmje.org/coi_disclosure.pdf (available on request from the corresponding author) and declare support from the Cochrane Collaboration for the development and evaluation of the tool described; they have no financial relationships with any organisations that might have an interest in the submitted work in the previous three years and no other relationships or activities that could appear to have influenced the submitted work.
  25. 25. DerSimonian R, Laird N. Meta-analysis in clinical trials. Controlled clinical trials. 1986;7(3):177–88. Epub 1986/09/01. pmid:3802833.
  26. 26. Higgins JP, Thompson SG, Deeks JJ, Altman DG. Measuring inconsistency in meta-analyses. BMJ (Clinical research ed). 2003;327(7414):557–60. Epub 2003/09/06. pmid:12958120; PubMed Central PMCID: PMC192859.
  27. 27. Egger M, Davey Smith G, Schneider M, Minder C. Bias in meta-analysis detected by a simple, graphical test. BMJ (Clinical research ed). 1997;315(7109):629–34. Epub 1997/10/06. pmid:9310563; PubMed Central PMCID: PMC2127453.
  28. 28. Begg CB, Mazumdar M. Operating characteristics of a rank correlation test for publication bias. Biometrics. 1994;50(4):1088–101. Epub 1994/12/01. pmid:7786990.
  29. 29. Wetterslev J, Jakobsen JC, Gluud C. Trial Sequential Analysis in systematic reviews with meta-analysis. BMC medical research methodology. 2017;17(1):39. Epub 2017/03/08. pmid:28264661; PubMed Central PMCID: PMC5397700.
  30. 30. Guyatt GH, Oxman AD, Vist GE, Kunz R, Falck-Ytter Y, Alonso-Coello P, et al. GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. BMJ (Clinical research ed). 2008;336(7650):924–6. Epub 2008/04/26. pmid:18436948; PubMed Central PMCID: PMC2335261 GRADE’s success has a positive influence on their academic career. Authors listed in the byline have received travel reimbursement and honorariums for presentations that included a review of GRADE’s approach to rating quality of evidence and grading recommendations. GHG acts as a consultant to UpToDate; his work includes helping UpToDate in their use of GRADE. HJS is documents editor and methodologist for the American Thoracic Society; one of his roles in these positions is helping implement the use of GRADE. He is supported by “The human factor, mobility and Marie Curie actions scientist reintegration European Commission grant: IGR 42192—GRADE.”.
  31. 31. Guideline Development Tool.https://gdt.gradepro.org/app/#projects. Available from: https://gdt.gradepro.org/app/#projects.
  32. 32. Zhou Y, Yang J, Wang B, Wang P, Wang Z, Yang Y, et al. Sequential use of midazolam and dexmedetomidine for long-term sedation may reduce weaning time in selected critically ill, mechanically ventilated patients: a randomized controlled study. Critical Care. 2022;26(1). WOS:000790169200002. pmid:35505432
  33. 33. Nader ND, Shadvar K, Pilehvar N, Sanaie S, Iranpour A, Hamishehkar H, et al. Long-term Dexmedetomidine versus midazolam in patients under mechanical ventilation: A double-blinded randomized clinical trial. Journal of Cellular and Molecular Anesthesia. 2021;6(3):259–68.
  34. 34. Shu A, Fu Y, Luo Y, Cao J, Jian W, Sun B, et al. An investigation on delirium and hemodynamics influenced by dexmedetomidine for sedating elderly patients in mechanical ventilation. International journal of clinical and experimental medicine. 2019;12(2):1942‐6.
  35. 35. Geng Y, Jiang X. A clinical study on the effects of dexmedetomidine on off-line extubation of invasive mechanical ventilation. International journal of clinical and experimental medicine. 2018;11(4):4094‐100.
  36. 36. Kawazoe Y, Miyamoto K, Morimoto T, Yamamoto T, Fuke A, Hashimoto A, et al. Effect of Dexmedetomidine on Mortality and Ventilator-Free Days in Patients Requiring Mechanical Ventilation With Sepsis: a Randomized Clinical Trial. JAMA. 2017;317(13):1321‐8. pmid:28322414.
  37. 37. Miyagawa N, Kawazoe Y, Sato T, Kushimoto S, Miyamoto K, Ohta Y, et al. Comparison between midazolam and propofol in acute phase for ventilated patients with sepsis: a post-hoc analysis of the DESIRE trial. Acute medicine & surgery. 2022;9(1):e746. Epub 2022/04/14. pmid:35414941; PubMed Central PMCID: PMC8982504.
  38. 38. Li Y, Yu ZX, Ji MS, Yan J, Cai Y, Liu J, et al. A Pilot Study of the Use of Dexmedetomidine for the Control of Delirium by Reducing the Serum Concentrations of Brain-Derived Neurotrophic Factor, Neuron-Specific Enolase, and S100B in Polytrauma Patients. J Intensive Care Med. 2019;34(8):674–81. Epub 2017/06/02. pmid:28569132.
  39. 39. 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(1):92–8. Epub 2015/03/20. pmid:25788780; PubMed Central PMCID: PMC4353161.
  40. 40. Srivastava VK, Agrawal S, Kumar S, Mishra A, Sharma S, Kumar R. Comparison of dexmedetomidine, propofol and midazolam for short-term sedation in postoperatively mechanically ventilated neurosurgical patients. J Clin Diagn Res. 2014;8(9):Gc04-7. Epub 2014/11/12. pmid:25386451; PubMed Central PMCID: PMC4225903.
  41. 41. Shehabi Y, Bellomo R, Reade MC, Bailey M, Bass F, Howe B, et al. Early goal-directed sedation versus standard sedation in mechanically ventilated critically ill patients: a pilot study*. Crit Care Med. 2013;41(8):1983–91. Epub 2013/07/19. pmid:23863230.
  42. 42. Jakob SM, Ruokonen E, Grounds RM, Sarapohja T, Garratt C, Pocock SJ, et al. Dexmedetomidine vs Midazolam or Propofol for Sedation During Prolonged Mechanical Ventilation Two Randomized Controlled Trials. Jama-Journal of the American Medical Association. 2012;307(11):1151–60. WOS:000301708000023. pmid:22436955
  43. 43. Turunen H, Jakob SM, Ruokonen E, Kaukonen KM, Sarapohja T, Apajasalo M, et al. Dexmedetomidine versus standard care sedation with propofol or midazolam in intensive care: an economic evaluation. Critical care (London, England). 2015;19(1):67. Epub 2015/04/19. pmid:25887576; PubMed Central PMCID: PMC4391080.
  44. 44. Huang Z, Chen Y, Yang Z, Liu J. Dexmedetomidine versus midazolam for the sedation of patients with non-invasive ventilation failure. Internal Medicine. 2012;51(17):2299–305. pmid:22975538
  45. 45. Ruokonen E, Parviainen I, Jakob SM, Nunes S, Kaukonen M, Shepherd ST, et al. Dexmedetomidine versus propofol/midazolam for long-term sedation during mechanical ventilation. Intensive Care Med. 2009;35(2):282–90. Epub 2008/09/17. pmid:18795253.
  46. 46. 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. Epub 2009/02/04. pmid:19188334.
  47. 47. Dasta JF, Kane-Gill SL, Pencina M, Shehabi Y, Bokesch PM, Wisernandle W, et al. A cost-minimization analysis of dexmedetomidine compared with midazolam for long-term sedation in the intensive care unit. Critical Care Medicine. 2010;38(2):497–503. WOS:000273927700019. pmid:19789442
  48. 48. Maldonado JR, Wysong A, van der Starre PJ, Block T, Miller C, Reitz BA. Dexmedetomidine and the reduction of postoperative delirium after cardiac surgery. Psychosomatics. 2009;50(3):206–17. Epub 2009/07/02. pmid:19567759.
  49. 49. Esmaoglu A, Ulgey A, Akin A, Boyaci A. Comparison between dexmedetomidine and midazolam for sedation of eclampsia patients in the intensive care unit. J Crit Care. 2009;24(4):551–5. Epub 2009/03/31. pmid:19327948.
  50. 50. Milbrandt EB, Deppen S, Harrison PL, Shintani AK, Speroff T, Stiles RA, et al. Costs associated with delirium in mechanically ventilated patients. Crit Care Med. 2004;32(4):955–62. Epub 2004/04/09. pmid:15071384.
  51. 51. Sanchez D, Brennan K, Al Sayfe M, Shunker SA, Bogdanoski T, Hedges S, et al. Frailty, delirium and hospital mortality of older adults admitted to intensive care: the Delirium (Deli) in ICU study. Critical care (London, England). 2020;24(1):609. Epub 2020/10/17. pmid:33059749; PubMed Central PMCID: PMC7565834.
  52. 52. Salluh JI, Wang H, Schneider EB, Nagaraja N, Yenokyan G, Damluji A, et al. Outcome of delirium in critically ill patients: systematic review and meta-analysis. BMJ (Clinical research ed). 2015;350:h2538. Epub 2015/06/05. pmid:26041151; PubMed Central PMCID: PMC4454920 at www.icmje.org/coi_disclosure.pdf and declare: no support from any organization for the submitted work; no financial relationships with any organizations that might have an interest in the submitted work in the previous three years; no other relationships or activities that could appear to have influenced the submitted work.
  53. 53. van den Boogaard M, Schoonhoven L, Evers AW, van der Hoeven JG, van Achterberg T, Pickkers P. Delirium in critically ill patients: impact on long-term health-related quality of life and cognitive functioning. Crit Care Med. 2012;40(1):112–8. Epub 2011/09/20. pmid:21926597.
  54. 54. Wang ML, Min J, Sands LP, Leung JM. Midazolam Premedication Immediately Before Surgery Is Not Associated With Early Postoperative Delirium. Anesth Analg. 2021;133(3):765–71. Epub 2021/03/16. pmid:33721875; PubMed Central PMCID: PMC8373629.
  55. 55. Athanassoglou V, Cozowicz C, Zhong H, Illescas A, Poeran J, Liu J, et al. Association of perioperative midazolam use and complications: a population-based analysis. Reg Anesth Pain Med. 2022;47(4):228–33. Epub 2022/01/14. pmid:35022262.
  56. 56. Memtsoudis S, Cozowicz C, Zubizarreta N, Weinstein SM, Liu J, Kim DH, et al. Risk factors for postoperative delirium in patients undergoing lower extremity joint arthroplasty: a retrospective population-based cohort study. Reg Anesth Pain Med. 2019:934–43. Epub 2019/07/16. pmid:31302641.
  57. 57. Skrobik Y, Leger C, Cossette M, Michaud V, Turgeon J. Factors predisposing to coma and delirium: fentanyl and midazolam exposure; CYP3A5, ABCB1, and ABCG2 genetic polymorphisms; and inflammatory factors. Crit Care Med. 2013;41(4):999–1008. Epub 2013/02/07. pmid:23385102.
  58. 58. Nugent M, Artru AA, Michenfelder JD. Cerebral metabolic, vascular and protective effects of midazolam maleate: comparison to diazepam. Anesthesiology. 1982;56(3):172–6. Epub 1982/03/01. pmid:7059025.
  59. 59. Liu JY, Guo F, Wu HL, Wang Y, Liu JS. Midazolam anesthesia protects neuronal cells from oxidative stress-induced death via activation of the JNK-ERK pathway. Molecular medicine reports. 2017;15(1):169–79. Epub 2016/12/14. pmid:27959401; PubMed Central PMCID: PMC5355727.
  60. 60. Shehabi Y, Howe BD, Bellomo R, Arabi YM, Bailey M, Bass FE, et al. Early Sedation with Dexmedetomidine in Critically Ill Patients. The New England journal of medicine. 2019;380(26):2506–17. Epub 2019/05/22. pmid:31112380.
  61. 61. 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. Epub 2016/10/19. pmid:27571256.
  62. 62. Venn RM, Bradshaw CJ, Spencer R, Brealey D, Caudwell E, Naughton C, et al. Preliminary UK experience of dexmedetomidine, a novel agent for postoperative sedation in the intensive care unit. Anaesthesia. 1999;54(12):1136–42. Epub 1999/12/14. pmid:10594409.
  63. 63. Sato T, Kawazoe Y, Miyagawa N, Yokokawa Y, Kushimoto S, Miyamoto K, et al. Effect of age on dexmedetomidine treatment for ventilated patients with sepsis: a post-hoc analysis of the DESIRE trial. Acute medicine & surgery. 2021;8(1):e644. Epub 2021/04/17. pmid:33859826; PubMed Central PMCID: PMC8033411.
  64. 64. Shehabi Y, Serpa Neto A, Howe BD, Bellomo R, Arabi YM, Bailey M, 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(4):455–66. Epub 2021/03/10. pmid:33686482; PubMed Central PMCID: PMC7939103.
  65. 65. Zaal IJ, Devlin JW, Hazelbag M, Klein Klouwenberg PM, van der Kooi AW, Ong DS, et al. Benzodiazepine-associated delirium in critically ill adults. Intensive Care Med. 2015;41(12):2130–7. Epub 2015/09/26. pmid:26404392.
  66. 66. Rood PJT, Zegers M, Ramnarain D, Koopmans M, Klarenbeek T, Ewalds E, et al. The Impact of Nursing Delirium Preventive Interventions in the ICU: A Multicenter Cluster-randomized Controlled Clinical Trial. Am J Respir Crit Care Med. 2021;204(6):682–91. Epub 2021/06/26. pmid:34170798.
  67. 67. Chen TJ, Traynor V, Wang AY, Shih CY, Tu MC, Chuang CH, et al. Comparative effectiveness of non-pharmacological interventions for preventing delirium in critically ill adults: A systematic review and network meta-analysis. International journal of nursing studies. 2022;131:104239. Epub 2022/04/26. pmid:35468538.
  68. 68. Girard TD, Kress JP, Fuchs BD, Thomason JW, Schweickert WD, Pun BT, et al. Efficacy and safety of a paired sedation and ventilator weaning protocol for mechanically ventilated patients in intensive care (Awakening and Breathing Controlled trial): a randomised controlled trial. Lancet (London, England). 2008;371(9607):126–34. Epub 2008/01/15. pmid:18191684.
  69. 69. Kotfis K, van Diem-Zaal I, Williams Roberson S, Sietnicki M, van den Boogaard M, Shehabi Y, et al. The future of intensive care: delirium should no longer be an issue. Critical care (London, England). 2022;26(1):200. Epub 2022/07/06. pmid:35790979; PubMed Central PMCID: PMC9254432.
  70. 70. Aggarwal J, Lustrino J, Stephens J, Morgenstern D, Tang WY. Cost-Minimization Analysis of Dexmedetomidine Compared to Other Sedatives for Short-Term Sedation During Mechanical Ventilation in the United States. Clinicoecon Outcomes Res. 2020;12:389–97. Epub 2020/08/18. pmid:32801809; PubMed Central PMCID: PMC7395701.
  71. 71. Yuan C, Timmins F, Thompson DR. Post-intensive care syndrome: A concept analysis. International journal of nursing studies. 2021;114:103814. Epub 2020/11/22. pmid:33220570.
  72. 72. Lee M, Kang J, Jeong YJ. Risk factors for post-intensive care syndrome: A systematic review and meta-analysis. Australian critical care: official journal of the Confederation of Australian Critical Care Nurses. 2020;33(3):287–94. Epub 2019/12/17. pmid:31839375.
  73. 73. Dong CH, Gao CN, An XH, Li N, Yang L, Li DC, et al. Nocturnal dexmedetomidine alleviates post-intensive care syndrome following cardiac surgery: a prospective randomized controlled clinical trial. BMC Med. 2021;19(1):306. Epub 2021/12/07. pmid:34865637; PubMed Central PMCID: PMC8647374.
  74. 74. Talke P, Chen R, Thomas B, Aggarwall A, Gottlieb A, Thorborg P, et al. The hemodynamic and adrenergic effects of perioperative dexmedetomidine infusion after vascular surgery. Anesth Analg. 2000;90(4):834–9. Epub 2000/03/29. pmid:10735784.
  75. 75. Ebert TJ, Hall JE, Barney JA, Uhrich TD, Colinco MD. The effects of increasing plasma concentrations of dexmedetomidine in humans. Anesthesiology. 2000;93(2):382–94. Epub 2000/07/26. pmid:10910487.
  76. 76. Lewis K, Piticaru J, Chaudhuri D, Basmaji J, Fan E, Møller MH, et al. Safety and Efficacy of Dexmedetomidine in Acutely Ill Adults Requiring Noninvasive Ventilation: A Systematic Review and Meta-analysis of Randomized Trials. Chest. 2021;159(6):2274–88. Epub 2021/01/13. pmid:33434496; PubMed Central PMCID: PMC8579314.
  77. 77. 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. Epub 2013/08/06. pmid:23910886.
  78. 78. Raza SM, Zsigmond EK, Barabas E. Midazolam causes no adverse hemodynamic effects in cardiac patients. Clin Ther. 1987;10(1):40–3. Epub 1987/01/01. pmid:3502455.
  79. 79. Bloor BC, Ward DS, Belleville JP, Maze M. Effects of intravenous dexmedetomidine in humans. II. Hemodynamic changes. Anesthesiology. 1992;77(6):1134–42. Epub 1992/12/01. pmid:1361311.
  80. 80. Kallio A, Scheinin M, Koulu M, Ponkilainen R, Ruskoaho H, Viinamäki O, et al. Effects of dexmedetomidine, a selective alpha 2-adrenoceptor agonist, on hemodynamic control mechanisms. Clinical pharmacology and therapeutics. 1989;46(1):33–42. Epub 1989/07/01. pmid:2568211.
  81. 81. Dasta JF, Kane-Gill SL, Durtschi AJ. Comparing dexmedetomidine prescribing patterns and safety in the naturalistic setting versus published data. The Annals of pharmacotherapy. 2004;38(7–8):1130–5. Epub 2004/06/03. pmid:15173557.