You are on page 1of 15

British Journal of Anaesthesia, 118 (2): 167–81 (2017)

doi: 10.1093/bja/aew411
Review Article

Evidence basis for using perineural dexmedetomidine


to enhance the quality of brachial plexus nerve blocks:
a systematic review and meta-analysis of randomized
controlled trials
L. Vorobeichik1,2, R. Brull1,3 and F. W. Abdallah1,2,4,*
1
Department of Anaesthesia, University of Toronto, Toronto, Canada, 2Department of Anaesthesia, St.
Michael’s Hospital, University of Toronto, Toronto, Canada, 3Department of Anaesthesia, Women’s College
Hospital, Toronto, Canada and 4The Li Ka Shing Knowledge Institute, University of Toronto, Toronto, Canada

*Corresponding author. E-mail: abdallahf@smh.ca

Abstract
Background. Dexmedetomidine has been proposed as a perineural local anaesthetic (LA) adjunct to prolong peripheral
nerve block duration; however, results from our previous meta-analysis in the setting of brachial plexus block (BPB) did not
support its use. Many additional randomized trials have since been published. We thus conducted an updated meta-
analysis.
Methods. Randomized trials investigating the addition of dexmedetomidine to LA compared with LA alone (Control) in BPB
for upper extremity surgery were sought. Sensory and motor block duration, onset times, duration of analgesia, analgesic
consumption, pain severity, patient satisfaction, and dexmedetomidine-related side-effects were analysed using random-
effects modeling. We used ratio-of-means (lower confidence interval [point estimate]) for continuous outcomes.
Results. We identified 32 trials (2007 patients), and found that dexmedetomidine prolonged sensory block (at least 57%,
P < 0.0001), motor block (at least 58%, P < 0.0001), and analgesia (at least 63%, P < 0.0001) duration. Dexmedetomidine expe-
dited onset for both sensory (at least 40%, P < 0.0001) and motor (at least 39%, P < 0.0001) blocks. Dexmedetomidine also
reduced postoperative oral morphine consumption by 10.2mg [-15.3, -5.2] (P < 0.0001), improved pain control, and enhanced
satisfaction. In contrast, dexmedetomidine increased odds of bradycardia (3.3 [0.8, 13.5](P ¼ 0.0002)), and hypotension (5.4
[2.7, 11.0] (P < 0.0001)). A 50-60mg dexmedetomidine dose maximized sensory block duration while minimizing haemo-
dynamic side-effects. No patients experienced any neurologic sequelae. Evidence quality for sensory block was high accord-
ing to the GRADE system.
Conclusions. New evidence now indicates that perineural dexmedetomidine improves BPB onset, quality, and analgesia.
However, these benefits should be weighed against increased risks of motor block prolongation and transient bradycardia
and hypotension.

Key words: adjunct; adjuvant; axillary; brachial plexus block; infraclavicular; interscalene; nerve block; perineural
dexmedetomidine; regional anaesthesia; sensory block; supraclavicular

C The Author 2017. Published by Oxford University Press on behalf of the British Journal of Anaesthesia.
V
All rights reserved. For Permissions, please email: journals.permissions@oup.com

167
Downloaded from https://academic.oup.com/bja/article-abstract/118/2/167/2924212/Evidence-basis-for-using-perineural
by guest
on 26 September 2017
168 | Vorobeichik et al.

wrist. Additional non-indexed articles were retrieved using


Editor’s key points Google Scholar; and the bibliographies of retrieved trials were
• Dexmedetomidine, an alpha-2 agonist, can be expected hand-searched for additional relevant studies. Our search was
limited to randomized trials published in the English language.
to extend the duration of local anaesthetic blocks but it
Only trials including adults (age > 18 yr) published in full-
has been unclear whether this has clinical benefit.
manuscript form between January 1985 and February 2016 were
• This systematic review provides sufficient evidence to
considered. Abstracts were excluded.
properly test this concept, finding enhanced onset and
longer duration of block with minimal side-effects.
• The optimal local anaesthetic adjunct dose for brachial Inclusion criteria
plexus block seems to be 50-60 mg. We included randomized trials with parallel group design
• There is some uncertainty as to overall analgesic effect- examining the effects of adding perineural dexmedetomidine
iveness and patient outcomes. as an adjunct to LA (Dex group), compared with LA alone
(Control group) on BPB characteristics, postoperative analgesia and
dexmedetomidine-related side-effects, in patients undergoing
upper extremity surgery with BPB. Specifically, interscalene (ISB),
Anaesthetists have sought strategies to extend the benefits of supraclavicular (SCB), infraclavicular (ICB) and axillary (AXB) level
single-shot peripheral nerve blocks beyond the duration of com- blocks of the brachial plexus performed for either anaesthesia or
monly available local anaesthetics (LA).1 Perineural adjuncts are postoperative analgesia were considered. Randomized and quasi-
one technically simple strategy that can be used for this pur- randomized, and single- and double-blinded trials were included.
pose.2 Dexmedetomidine, an alpha-2 agonist,3 has been pro- Randomized trials without a control group were excluded. We also
posed as a safe4 and effective56 adjunct capable of extending the excluded trials if non-perineural routes of dexmedetomidine ad-
duration of single-shot block. ministration were used (e.g. intra-articular injection);13 if surgeries
Hyperpolarization-activated cation currents normally bring involved anatomical areas other than the upper extremity (e.g. ab-
neurons back to the resting potential and normal functional activ- dominal);14 and if blocks other than BPB were performed.15 Studies
ity during the refractory phase in an action potential. By blocking of i.v. regional anaesthesia16 17 and distal peripheral nerve blocks
these currents, dexmedetomidine can accentuate inhibition of (e.g. medial, radial or ulnar blocks)18 19 were also excluded.
neuronal conduction and produce analgesia.6 However, despite
early promising evidence from animal56 and human7 studies that Trial selection and methodological assessment
have signaled efficacy, our quantitative systematic review of dex-
The two authors (LV and FWA) independently evaluated the
medetomidine as an adjunct to brachial plexus block (BPB), pub-
identified abstracts. Inclusion of qualifying studies in the review
lished in 2013,8 was unable to demonstrate any clinically important
was taken by consensus between the two authors.
benefits. Some of the trials7 9–11 were marked by substantial clinical
Disagreements were resolved by re-evaluating the full manu-
and statistical heterogeneities that may have undermined a precise
script of the source studies and consulting with the third author
estimation of the dexmedetomidine treatment effect. Many add-
(RB). Trials that failed to meet the inclusion criteria were
itional trials of dexmedetomidine as a BPB adjunct have been since
excluded.
published, thus prompting a re-examination specifically to assess
The quality of the reviewed trials was independently as-
the role of dexmedetomidine in prolonging sensory block duration.
sessed using the Cochrane Collaboration Risk of Bias tool20 by
two of the authors (LV and FWA). The tool evaluates trials for
Methods biases, among which are selection (randomization and alloca-
tion), performance and detection (blinding), attrition, reporting,
We followed PRISMA statement guidelines12 in the preparation and other forms of bias. A score was assigned to each trial by
of this manuscript. Randomized trials examining the effect of consensus; if an agreement could not be reached, the third au-
dexmedetomidine on the duration of sensory block after single- thor (RB) was consulted. Considering the limited number of
shot BPB were evaluated using a predefined protocol, but this studies and that our earlier meta-analysis failed to demonstrate
was not previously published. the effects of dexmedetomidine on nerve blocks, we decided not
to exclude studies based on the quality scores, but rather to take
Literature search an inclusive approach towards studies with a view towards an-
swering the question of interest. However, trials were excluded
Two of the authors (LV and FWA) independently sought and if they had fewer than 10 subjects per group, to reduce the pos-
retrieved relevant studies from electronic databases including sibility of chance in estimating a treatment effect.
the US National Library of Medicine database, MEDLINE; the
Excerpta Medica database, EMBASE; the Cochrane Databases of
systematic reviews; the Cochrane central register of controlled
Data extraction
clinical trials; Cumulative Index of Nursing and Allied Health The authors independently extracted relevant data using a
Literature (CINAHL); Scopus; Web of Science; MEDLINE In- standardized data sheet; discrepancies were resolved by re-
Process; and other non-indexed citations. The medical subject examining the source data as a first resort, then consulting with
headings (MeSH), text words, and controlled vocabulary terms the third author. Data extracted included primary author, yr of
relating to Dexmedetomidine and Medetomidine were sought. publication, comparative groups, sample size, nature of primary
Results were combined using the Boolean operator “AND” with outcome, surgical site, nature of surgical anaesthetic, level of
the search terms analgesia, anaesthesia, adjunct, adjuvant, BPB, nerve localization technique, type and dose of LA, dose of
anaesthetics local, nerve block, perineural, regional anaesthesia perineural dexmedetomidine, block characteristics, analgesic
and terms designating upper extremity such as arm, brachial effects, and dexmedetomidine-related side-effects. Data on
plexus, forearm, elbow, hand, humerus, radius, shoulder, and additional study arms examining other perineural additives

Downloaded from https://academic.oup.com/bja/article-abstract/118/2/167/2924212/Evidence-basis-for-using-perineural


by guest
on 26 September 2017
Perineural dexmedetomidine for brachial plexus block | 169

(e.g. dexamethasone21, fentanyl22) were extracted but excluded proportion of patients who experienced a certain side-effect at
from the final analysis. least once.
Data from trials with more than two intervention groups
receiving different doses of perineural dexmedetomidine were
Outcomes assessed combined into a single group as per the Cochrane Handbook.30
We designated sensory block duration (min), defined as time We analysed results on an intention-to-treat basis by analysing
from completion of LA injection to full recovery from sensory the data available from all participants in each study group, re-
block, as a primary outcome. Secondary outcomes included gardless of compliance or attrition, to estimate the influence on
block characteristics, namely motor block duration (min), and treatment effect.31 Continuous data describing patient satisfac-
sensory and motor block onset times (min), defined as time tion were converted to odds ratios (OR) to facilitate quantitative
from completion of LA injection to achieving full sensory and analysis.32
motor block, respectively. We also evaluated analgesic out-
comes comprising duration of analgesia (min), defined as time
Meta-analysis
to first analgesic request, or as defined by authors; cumulative
analgesic consumption during the first 24 h postoperatively, ex- Data entry was performed by one author (LV) and checked by
pressed as oral morphine equivalents (mg);23 rest and dynamic another (FWA). Meta-analytic techniques (Revman 5.3,
postoperative pain severity (visual analogue scale, VAS; 0 ¼ no Cochrane Library, Oxford, UK, and OpenMeta[Analyst] software
pain, 10 ¼ worst pain imaginable) at 12 and 24 h postoperatively; version: Beta 3.13, Tufts Medical Centre)33 were used to combine
patient satisfaction with postoperative pain relief, (VAS; the data where possible. As a variety of BPB and upper extremity
0 ¼ least satisfied, 10 ¼ most satisfied); and length of hospital procedures were examined, we selected the DerSimonian ran-
stay. Also included were frequency of dexmedetomidine-related dom effect modeling to pool the results.34
adverse effects (bradycardia, hypotension, excessive sedation, The strength of evidence pooled from the trials reviewed
hypoxemia)24 and postoperative nausea and vomiting (PONV), was assessed using Grades of Recommendation, Assessment,
as defined by authors, and block-related complications. Development, and Evaluation (GRADE) guidelines.35 In contrast
For the purpose of this review, trials reporting the range or to other assessment tools that evaluate quality across outcomes
interquartile range (IQR) were included using an estimate of the within a given trial, the GRADE tool evaluates quality across tri-
standard deviation (SD), using the formulas: SD¼ Range/4 and als for each outcome. Based on key elements including study
SD¼ IQR/1.35, respectively, as described by the Cochrane quality, consistency, directness, precision, and publication bias,
Handbook for Systematic Reviews.25 Data reported as 95% confi- the GRADE tool classifies the strength of synthesized evidence
dence intervals (CI) were similarly used to estimate the range, into four categories: i) high quality: further research is very un-
and were converted to SD. The median was used to estimate the likely to change the confidence in the estimate of effect; ii) mod-
mean if its value was not provided.26 Postoperative pain severity erate quality: further research is likely to have an important
reported as numerical rating scale scores or verbal rating scale impact on the confidence in the estimate of effect and may
scores was converted to VAS scores.27 change the estimate; iii) low quality: further research is very
likely to have an important impact on the confidence in the esti-
mate of effect and is likely to change the estimate; and iv) very
Predefined sources of heterogeneity low quality: we are very uncertain about the estimate. For con-
To explore potential causes of heterogeneity in our results, we sistency in specific, we resorted to examining the similarity of
pre-identified the clinical characteristics of individual trials and point estimates, and the extent of overlap of confidence inter-
known confounders that may lead to variations in our primary vals, in addition to testing heterogeneity using the I2 statistic.
outcome results (sensory block duration). The variables of inter- All time-to-event outcomes, including block characteristics
est included i) level of BPB, ii) LA dose, iii) dexmedetomidine (sensory and motor block onset and duration), and the duration
dose, iv) epinephrine dose, v) block localization technique, and of analgesia, the ratio of means, standard error, and 95% CIs
vi) nature of surgical anaesthetic used (general anaesthesia vs were calculated for all continuous outcomes that examined
nerve block). Based on the clinical assumption that different lev- change from baseline.36 37 For the remaining outcomes, the OR
els of BPB will lead to different block characteristics and anal- and 95% CIs were reported for dichotomous outcomes, while
gesic effects, we planned to separately analyse outcomes the weighted mean difference and 95% CI were reported for con-
according to the type of BPB performed (interscalene, tinuous outcomes. Differences were considered statistically sig-
supraclavicular, infraclavicular, and axillary). The degree to nificant when the P-value was < 0.05 and the 95% CI did not
which the remaining factors can predict the duration of sensory include 1 for OR and 0 for the standardized mean difference.
block (primary outcome) was evaluated using meta-regression Heterogeneity of the pooled results was assessed using the I2
analysis. statistic.38 When heterogeneity was significant (I2 > 50%), we
planned to explore the sources of heterogeneity of the primary
outcome data (i.e. sensory block duration), by examining the as-
Statistical analysis sociation with pre-specified confounders. The risk of publica-
We used data presented in tables as the primary source for ex- tion bias was evaluated by checking for asymmetry of the
traction; when data were not presented in tables, we contacted funnel plots, as described in the Egger regression test.39
authors for additional information. Graphical data not other-
wise available in the text or from the authors were estimated
from figures. As a final resort, when SD values were not re-
Results
ported for an outcome (e.g. postoperative pain),9 28 these values Our database search strategy retrieved 137 potentially relevant
were imputed.29 30 Dichotomous data relating to side-effects records published between 2010 and 2016, including 15 from
were converted to incidence (n/N) during a given time interval; non-indexed citations. Of these, 102 records were excluded after
and the single highest incidence was used to capture the initial screening; and another record was excluded for

Downloaded from https://academic.oup.com/bja/article-abstract/118/2/167/2924212/Evidence-basis-for-using-perineural


by guest
on 26 September 2017
170 | Vorobeichik et al.

Identification

Records identified through Additional records identified


database search through other sources
(n= 122) (n= 15)

Records after removing duplicates Records excluded


(n = 137) (n= 102)
4: Abstract, full text not available
Screening

1: Abstract, full text in different


record
30: Design: Reviews
Records screened 8: Population: Animal studies
(n = 137) 38: Intervention: Dex not
administered perineurally
9: Intervention: No Dex group
1: Intervention: Perineural Dex in
Full-text articles assessed other nerve blocks
for eligibility 2: Comparator: No control group
Eligibility

(n= 35) 1: Language (Chinese)

Studies included in Full-text articles excluded


qualitative synthesis (n= 1)
(n = 34) 1: Duplicated work
Included

Studies included in
quantitative synthesis
(meta-analysis)
(n = 34)

Fig 1 Preferred reporting items for systematic reviews and meta-analyses (PRISMA)12 flow diagram summarizing retrieved, included, and excluded randomized
trials.

duplicated work.40 None of the remaining records involved less anaesthesia in 23 trials, postoperative analgesia in four trials,55
60 62 64
than 10 subjects per group. A total of 34 full-text randomized tri- and seven trials46 48 50–53 56 did not provide sufficient de-
als7 9–11 21 22 28 41–67 were included in the final analysis. Figure 1 tails regarding the role of blocks. The mode of nerve block local-
represents the preferred reporting items for systematic reviews ization was anatomical (landmark) in six trials,10 42 43 45–48 nerve
and meta-analyses (PRISMA)12 flow diagram, and summarizes stimulation in 20 trials, ultrasound in three trials,8 55 63 a com-
the reasons for exclusion of records. bination of nerve stimulation and ultrasound in four trials,9 58 59
65
and not defined in one trial.44 With the exception of one trial
that used mepivacaine,66 all trials used long-acting LAs (ropiva-
Trial characteristics caine, bupivacaine, or levobupivacaine) alone or in conjunction
Data from a total of 2007 patients, including 1026 in the Dex with short acting LAs; and three trials used epinephrine in all
group and 981 in the Control group, were available for analysis. study arms.60 64 65 Perineural dexmedetomidine was used in ei-
Details of the 34 trials, intervention arms, sample size, and out- ther weight-based doses (0.75 to 1.0 mg/kg) or flat doses (10 to
comes assessed are summarized in Table 1. These 34 trials 150 mg). A combination of perineural bolus and infusion of dex-
examined single-shot nerve blocks at all levels of brachial medetomidine was used in one study; in this case, we excluded
plexus, with five trials at the interscalene level,43 52 55 60 64 18 at all outcomes influenced by the infusion.60 Eight trials examined
the supraclavicular level,10 22 28 44–49 51 53 56–58 62 63 65 67 three at adjuncts other than perineural dexmedetomidine;8 21 43 51 59
the infraclavicular level,9 59 66 and eight at the axillary level.7 11 these study arms were excluded from the analysis as they
21 41 42 50 54 61
The blocks were used to provide surgical did not meet the inclusion criteria. Two trials included two

Downloaded from https://academic.oup.com/bja/article-abstract/118/2/167/2924212/Evidence-basis-for-using-perineural


by guest
on 26 September 2017
by guest
on 26 September 2017
Table 1 Trial characteristics and outcomes examined. Abbreviations: Dex, dexmedetomidine; Epi, epinephrine; kg, kilogram; ml, milliliter; ND, not defined; NS, normal saline; PONV, postopera-
tive nausea and vomiting; lg, microgram. (*) excluded from analysis

Author Surgery Block use N Groups (n) Local Anaesthetic Dex dose Block Primary outcome
Concentration – Total localization
volume, and Epi dose

Interscalene Block
Abdallah 201664 Shoulder, arthroscopic Analgesic 99 1. Ropivacaine þ NS (32) 0.5% - 15 ml epi 0.5 mg/kg Ultrasound Duration of analgesia
2. Ropivacaine þ Dex þ NS IV (33) 1:200,000
3. Ropivacaine þ NS þ Dex IV (34) *
Bengisun 201460 Shoulder, arthroscopic Analgesic 48 1. Levobupivacaine (25) 0.5% - 20 ml epi 50 mg 10 mg Stimulator Pain scores area
2. Levobupivacaine þ Dex (23) under the curve
Fritsch 201455 Shoulder, arthroscopic, Analgesic 61 1. Ropivacaine þ NS (30) 0.5% - 10 ml 150 mg Ultrasound Duration of sensory
open 2. Ropivacaine þ Dex (31)
Kumar 201443 Shoulder Surgical 90 1. Bupivacaine þ NS (30) 0.25% - 40 ml 50 mg Landmark N/D
2. Bupivacaine þ Dex (30)
3. Bupivacaine þ Dexamethasone (30) *
Rashmi 201652 Upper limb ND 60 1. Ropivacaine þ NS (30)2. Ropivacaine 0.75% - 30 ml 50 mg Stimulator Haemodynamic side-effects
þ Dex (30)
Supraclavicular Block
Agarwal 201456 Upper limb ND 50 1. Bupivacaine þ NS (25) 0.325% - 30 ml 100 mg Stimulator N/D
2. Bupivacaine þ Dex (25)
Bharti 201565 Upper limb Surgical 54 1. Ropivacaine þ lidocaine (27) Ropivacaine 0.325% 1 mg/kg Ultrasound Duration of analgesia
2. Ropivacaine þ lidocaine þ Dex (27) Lidocaine þ stimulator
1% 40 ml epi 1:200,000
Biswas 201457 Forearm, hand Surgical 60 1. Levobupivacaine þ NS (30) 0.5% - 35 ml 100 mg Stimulator N/D
2. Levobupivacaine þ Dex mg (30)
Das 201428 Distal arm, forearm, hand Surgical 84 1. Ropivacaine þ NS (42) 0.5% - 30 ml 100 mg Stimulator Duration of analgesia
2. Ropivacaine þ Dex (42)
Das 201667 Upper limb Surgical 80 1. Ropivacaine þ NS (40) 0.5% - 30 ml 1 mg/kg Stimulator Duration of analgesia
2. Ropivacaine þ Dex (40)
Dixit 201544 Upper limb Surgical 40 1. Levobupivacaine þ NS (20) 0.5% - 29 ml 1 mg/kg ND N/D
2. Levobupivacaine þ Dex (20)

Downloaded from https://academic.oup.com/bja/article-abstract/118/2/167/2924212/Evidence-basis-for-using-perineural


Gandhi 201210 Upper limb Surgical 70 1. Bupivacaine þ NS (35) 0.25% - 38 ml 30 mg Landmark Duration of analgesia
2. Bupivacaine þ Dex (35)
Gurajala 201562 Distal arm, forearm, hand Analgesic 36 1. Ropivacaine þ NS (18) 0.5% - 35 ml 50 mg Stimulator Onset of motor block
2. Ropivacaine þ Dex (18)
Kathuria 201563 Distal arm, forearm, hand Surgical 60 1. Ropivacaine (20) 0.5% - 30 ml 50 mg Ultrasound N/D
2. Ropivacaine þ Dex (20)
3. Ropivacaine þ Dex i.v. (20) *
Kaur 201545 Upper limb Surgical 92 1. Levobupivacaine þ lidocaine þ NS (45) Levobupivacaine 0.375% 1 mg/kg Landmark Onset of sensory block
2. Levobupivacaine þ lidocaine þ Dex (45) Lidocaine 0.25% 40 ml
Perineural dexmedetomidine for brachial plexus block

Khade 201346 Forearm ND 40 1. Bupivacaine þ lidocaine þ NS (20) Bupivacaine 0.33% 50 mg Stimulator N/D
|

2. Bupivacaine þ lidocaine þ Dex (20) Lidocaine 0.66% 30 ml


Kwon 201558 Forearm, hand Surgical 60 1. Ropivacaine þ NS (30) 0.5% - 40 ml 1 mg/kg Ultrasound Bispectral index changes
171

2. Ropivacaine þ Dex (30) þ stimulator


(continued)
by guest
Table 1 Continued
Author Surgery Block use N Groups (n) Local Anaesthetic Dex dose Block Primary outcome

on 26 September 2017
Concentration – Total localization
172

volume, and Epi dose


|

Manohar 201522 Distal arm, forearm Surgical 90 1. Bupivacaine þ NS (30) 0.5% - 30 ml 50 mg Stimulator N/D
2. Bupivacaine þ Dex (30)
3. Bupivacaine þ Fentanyl 50 mg (30) *
Nema 201447 Forearm Surgical 60 1. Ropivacaine þ NS (30) 0.75% - 30 ml 50 mg Landmark N/D
2. Ropivacaine þ Dex (30)
Patki 201548 Forearm ND 60 1. Ropivacaine þ NS (30) 0.5% - 30 ml 1 mg/kg Landmark Duration of analgesia
2. Ropivacaine þ Dex (30)
Vorobeichik et al.

Saraf 201653 Upper limb ND 90 1. Ropivacaine þ NS (30) 0.75% - 20 ml 2. 25 mg Stimulator N/D


2. Ropivacaine þ Dex 25 mg (30) 3. 50 mg
3. Ropivacaine þ Dex 50 mg (30)
Singh 201649 Upper limb Surgical 60 1. Levobupivacaine þ NS (30) 0.5% - 30 ml 100 mg Stimulator Duration of analgesia
2. Levobupivacaine þ Dex (30)
Tanden 201651 Upper limb ND 90 1. Levobupivacaine þ NS (30) 0.5% - 30 ml 100 mg Stimulator N/D
2. Levobupivacaine þ Dex (30)
3. Levobupivacaine þ Clonidine 150 mg (30) *
Infraclavicular Block
Ammar 20129 Forearm, hand Surgical 60 1. Bupivacaine þ NS (30) 0.33% - 30 ml 0.75 m/kg Ultrasound þ Duration of analgesia
2. Bupivacaine þ Dex (30) Stimulator
Mirkheshti 201459 Distal arm, forearm Surgical 103 1. Lidocaine þ NS (34) 1.5% - 30 ml 100 mg Ultrasound þ N/D
2. Lidocaine þ Dex (34) stimulator
3. Lidocaine þ ketorolac 50 mg (35) *
Song 201466 Upper limb Surgical 30 1. Mepivacaine þ NS (10) 1% - 40 ml 1 mg/kg Stimulator Duration of sensory block
2. Mepivacaine þ Dex (10)
3. Mepivacaine þ epi 200 mg (30) *
Axillary block
Arun 201650 Forearm and hand ND 60 1. Ropivacaine þ NS (30) 0.75% - 25 ml 50 mg Stimulator N/D
2. Ropivacaine þ Dex (30)
Bangera 201661 Forearm and hand Surgical 80 1. Ropivacaine þ NS (40) 0.5% - 40 ml 100 mg Stimulator N/D
2. Ropivacaine þ Dex (40)
Esmaoglu 20107 Forearm and hand Surgical 60 1. Levobupivacaine þ NS (30) 0.25% - 40 ml 100 mg Stimulator Duration of sensory
2. Levobupivacaine þ Dex (30) and motor block
Hanoura 201341 Forearm and hand Surgical 48 1. Bupivacaine þ NS (24) 0.5% - 39 ml 100 mg Stimulator N/D

Downloaded from https://academic.oup.com/bja/article-abstract/118/2/167/2924212/Evidence-basis-for-using-perineural


2. Bupivacaine þ Dex (24)
Karthik 201542 Forearm and hand Surgical 100 1. Levobupivacaine þ NS (50) 0.375% - 39 ml 1 mg/kg Landmark N/D
2. Levobupivacaine þ Dex (50)
Kaygusuz 201211 Forearm and hand Surgical 60 1. Levobupivacaine þ NS (30) 0.5% - 39 ml 1 mg/kg Stimulator Duration Of sensory block
2. Levobupivacaine þ Dex (30)
Lee 201621 Forearm and hand Surgical 51 1. Ropivacaine þ NS (17) 0.5% - 22 ml 100 mg Stimulator Duration of sensory block
2. Ropivacaine þ Dex (17)
3. Ropivacaine þ dexamethasone 10 mg (17) *
Zhang 201454 Forearm and hand Surgical 45 1. Ropivacaine þ NS (15) 0.33% - 40 ml 2. 50 mg Stimulator Duration of analgesia
2. Ropivacaine þ Dex 50 lg (15) 3. 100 mg
3. Ropivacaine þ Dex 100 lg (15)
Perineural dexmedetomidine for brachial plexus block | 173

perineural dexmedetomidine arms54 with different doses,


which were combined into a single group for the purpose of this Table 2 Risk of bias summary: review authors’ judgements about
each risk of bias item for each included study. Green circle, low risk of
analysis. All trials reported block characteristics, analgesic out-
bias; orange circle, high risk of bias; yellow circle, unclear risk of bias
comes, and dexmedetomidine-related complications.

Blinding of participants and personnel (performance bias)


Risk of bias assessment

Blinding of outcome assessment (detection bias)


Some of the studies reviewed lacked sufficient details to permit

Random sequence generation (selection bias)


full evaluation of the risk of bias; in such cases, we were conser-

Incomplete outcome data (attrition bias)


Allocation concealment (selection bias)
vative in our risk of bias evaluation by tending to classify trials

Selective reporting (reporting bias)


as having an “unclear risk of bias” when the complete details
that allow the exclusion of selection, performance and detection
biases were not reported. Furthermore, trials that used general-
izations such as “similar side-effects between study groups”, or
presented haemodynamic outcomes data in a graphical format
that precluded determining the actual risk of side-effects, were

Other bias
considered to be at a high risk for selective reporting. The re-
viewers’ consensus assessment of the risk is detailed in Table 2.
Considering our conservative approach, and the fact that the Abdallah 2016 (64) + + + + + + +
main outcomes of interest, namely block characteristics, were
Agarwal 2014 (56) + ? ? + + + ?
less likely to be affected by the aforementioned biases, we con-
sidered the methodological quality for the majority of the 34 tri- Ammar 2012 (9) + + + ? + + +
als included to be acceptable, and rated the overall risk of bias
Arun 2016 (50) + ? ? ? ? – +
across the studies as moderate. Most of trials had low risk for
Bangera 2016 (61) + ? ? + ? + +
selection bias, attrition bias, and other biases; moreover, the
majority of trials were assigned unclear risk of selection and Bengisun 2014 (60) ? ? ? + + – +
performance bias, because authors did not provide sufficient Bharti 2015 (65) + + ? + + + +
details regarding blinding and concealment of sequence
Biswas 2014 (57) + ? + + + – +
allocation. Bias as a result of selective reporting was classified
as high in most of the trials, because of the aforementioned Das 2014 (28) + ? ? + + – ?
conservative approach in assessing the reporting of the Das 2016 (67) ? ? ? + + + +
dexmedetomidine-related haemodynamic side-effects. None of
Dixit 2015 (44) ? ? – – ? + +
the trials were quasi-randomized.
Esmaoglu 2010 (7) + ? ? ? + – +

Fritsch 2014 (55) + + + + + ? ?


Sensory block duration
Ghandi 2012 (10) ? ? ? + + – +
Data describing the primary outcome, sensory block duration
(time to full recovery from sensory block), were available from Gurajala 2015 (62) + ? + + + + –
all trials (910 patients in Dex group) except two,8 60 and are pre- Hanoura 2013 (41) – – – – – + ?
sented in Figure 2. Administering perineural dexmedetomidine
Karthik 2015 (42) – – ? ? + – –
as an LA adjunct to BPB prolonged sensory block duration com-
pared with Control. Expressed as lower CI [point estimate], the Kathuria 2015 (63) + ? ? + ? + +
prolongation was at least 44% [57%], (P < 0.0001, I2 ¼ 99%) for ISB; Kaur 2015 (45) + ? ? + + – +
54% [82%], (P < 0.0001, I2 ¼ 100%) for SCB; 21% [37%], (P < 0.0001,
Kaygusuz 2012 (11) + ? ? ? + ? +
I2 ¼ 75%) for ICB; and 34% [37%], (P < 0.0001, I2 ¼ 92%) for AXB. The
overall treatment effect for all levels of BPB suggested that dex- Khade 2013 (46) ? ? ? ? ? – ?

medetomidine prolonged sensory block duration by at least 57% Kumar 2014 (43) ? ? – – ? – ?
[65%], (P < 0.0001, I2 ¼ 100%), or by 3.8 h (from 7.7 h to 11.5 h).
Kwon 2015 (58) + + ? ? + ? +
The primary outcome results were characterized by signifi-
cant heterogeneity for all four subgroups. Performing meta- Lee 2016 (21) + ? ? ? + + +

regression analysis using the a priori pre-specified confounders, Manohar 2015 (22) ? ? ? ? ? ? ?
namely the LA dose, dexmedetomidine dose, epinephrine dose,
Mirkheshti 2014 (59) + ? ? ? ? + +
block localization technique, and nature of surgical anaesthetic,
yielded omnibus P-values of 0.047, 0.948, 0.37, 0.09, and 0.215, re- Nema 2014 (47) ? ? ? ? + – ?
spectively, suggesting that the total LA dose is a predictor of Patki 2015 (48) ? ? – – + – –
sensory block prolongation.
Rashmi 2016 (52) + ? ? ? + – ?
Based on the fact that 15 trials were contributed by non-
indexed citations, we elected to perform an additional subgroup Saraf 2016 (53) ? ? ? ? ? – ?

analysis to assess the influence of potential selection bias intro- Singh 2016 (49) + ? ? ? + – ?
duced by including non-indexed trials. This analysis indicated
Song 2014 (66) ? – – – + – ?
that dexmedetomidine prolonged sensory block duration by at
least 46% [60], (P < 0.0001, I2 ¼ 99%) and 58% [70], (P < 0.0001, Tandon 2016 (51) ? ? ? ? + – +
I2 ¼ 100%) for the indexed and non-indexed trials, respectively. Zhang 2014 (54) + ? ? ? ? – ?
There was no difference between the two subgroups (P ¼ 0.3),

Downloaded from https://academic.oup.com/bja/article-abstract/118/2/167/2924212/Evidence-basis-for-using-perineural


by guest
on 26 September 2017
174 | Vorobeichik et al.

Ratio of means Ratio of means


Study or subgroup Ratio of means SE Weight IV, random, 95% CI IV, random, 95% CI
1.3.1 ISB
Fritsch 2014 (55) 1.2857 0.0085 3.3% 1.29 [1.27, 1.30]
Kumar 2014 (43) 2.2466 0.0689 3.0% 2.25 [2.11, 2.38]
Rashmi 2016 (52) 1.3673 0.0048 3.3% 1.37 [1.36, 1.38]
Subtotal (95% CI) 9.6% 1.57 [1.44, 1.69]
Heterogeneity: Tau2=0.01; χ2=239.51, df=2 (P<0.00001); I2=99%
Test for overall effect: Z=24.35 (P<0.00001)

1.3.2 SCB
Agarwal 2014 (56) 3.2181 0.0528 3.1% 3.22 [3.11, 3.32]
Bharti 2015 (65) 1.3636 0.0388 3.2% 1.36 [1.29, 1.44]
Biswas 2014 (57) 1.3922 0.0209 3.3% 1.39 [1.35, 1.43]
Das 2014 (28) 1.793 0.0425 3.2% 1.79 [1.71, 1.88]
Das 2016 (67) 1.5562 0.025 3.3% 1.56 [1.51, 1.61]
Dixit 2015 (44) 1.4009 0.029 3.2% 1.40 [1.34, 1.46]
Ghandi 2012 (10) 4.9993 0.0435 3.2% 5.00 [4.91, 5.08]
Gurajala 2015 (62) 2.119 0.0807 2.9% 2.12 [1.96, 2.28]
Kathuria 2015 (63) 1.7481 0.0773 2.9% 1.75 [1.60, 1.90]
Kaur 2015 (45) 1 0.0219 3.3% 1.00 [0.96, 1.04]
Khade 2013 (46) 2.0025 0.0453 3.2% 2.00 [1.91, 2.09]
Kwon 2015 (58) 1.4334 0.0528 3.1% 1.43 [1.33, 1.54]
Manohar 2015 (22) 1 0.0154 3.3% 1.00 [0.97, 1.03]
Nema 2014 (47) 1.4044 0.058 3.1% 1.40 [1.29, 1.52]
Patki 2015 (48) 1.2862 0.0084 3.3% 1.29 [1.27, 1.30]
Saraf 2016 (53) 1.5323 0.0319 3.2% 1.53 [1.47, 1.59]
Singh 2016 (49) 2.0937 0.057 3.1% 2.09 [1.98, 2.21]
Tandon 2016 (51) 1.4097 0.0155 3.3% 1.41 [1.38, 1.44]
Subtotal (95% CI) 57.1% 1.82 [1.54, 2.10]
Heterogeneity: Tau2=0.36; χ2=9789.54, df=17 (P<0.00001); I2=100%
Test for overall effect: Z=12.85 (P<0.00001)

1.3.3 ICB
Ammar 2012 (9) 1.4621 0.0269 3.2% 1.46 [1.41, 1.51]
Mirkheshti 2014 (59) 1.3139 0.135 2.4% 1.31 [1.05, 1.58]
Song 2014 (66) 1.2699 0.0654 3.0% 1.27 [1.14, 1.40]
Subtotal (95% CI) 8.7% 1.37 [1.21, 1.52]
Heterogeneity: Tau2=0.01; χ2=8.16, df=2 (P=0.02); I2=75%
Test for overall effect: Z=17.69 (P<0.00001)

1.3.4 AXB
Arun 2016 (50) 1.4236 0.0053 3.3% 1.42 [1.41, 1.43]
Bangera 2016 (61) 1.3699 0.0324 3.2% 1.37 [1.31, 1.43]
Esmaoglu 2010 (7) 1.318 0.0242 3.3% 1.32 [1.27, 1.37]
Hanoura 2013 (41) 1.0885 0.0456 3.2% 1.09 [1.00, 1.18]
Karthik 2015 (42) 1.4004 0.0024 3.3% 1.40 [1.40, 1.41]
Kaygusuz 2012 (11) 1.3905 0.0229 3.3% 1.39 [1.35, 1.44]
Lee 2016 (21) 1.8031 0.1171 2.6% 1.80 [1.57, 2.03]
Zhang 2014 (54) 1.447 0.1292 2.5% 1.45 [1.19, 1.70]
Subtotal (95% CI) 24.6% 1.37 [1.34, 1.40]
Heterogeneity: Tau2=0.00; χ2=89.31, df=7 (P<0.00001); I2=92%
Test for overall effect: Z=82.48 (P<0.00001)

Total (95% CI) 100.0% 1.65 [1.57, 1.73]


Heterogeneity: Tau2=0.05; χ2=10251.31, df=31 (P<0.00001); I2=100%
–2 –1 0 1 2
Test for overall effect: Z=40.09 (P<0.00001)
Favours control Favours dexmedetomidine
Test for subgroup differences: χ2=18.23, df=3 (P =0.0004); I2=83.5%

Fig 2 Forest plot depicting sensory block duration. The individual trials’ ratio of means, standard error, and the pooled estimates of the ratio of means are shown.
The 95% confidence intervals are shown as lines for individual studies and as diamonds for pooled estimates. Abbreviations: AXB, axillary block; CI, confidence
interval; ICB, infraclavicular block; ISB, interscalene block; SCB, supraclavicular block; SE, standard error.

Downloaded from https://academic.oup.com/bja/article-abstract/118/2/167/2924212/Evidence-basis-for-using-perineural


by guest
on 26 September 2017
Perineural dexmedetomidine for brachial plexus block | 175

and heterogeneity remained high. Finally, the funnel plot did was not defined in six trials.45 46 48 51–53 Dexmedetomidine pro-
not suggest significant publication bias (P ¼ 0.08) for our primary longed the duration of analgesia by at least 60% [74%],
outcome. (P < 0.0001, I2 ¼ 100%) for ISB; 69% [91%], (P < 0.0001, I2 ¼ 100%) for
When the strength of the synthesized evidence was eval- SCB; 0% [39%], (P < 0.0001, I2 ¼ 97%) for ICB; and 12% [33%],
uated using the GRADE guidelines, there was high evidence that (P < 0.0001, I2 ¼ 99%) for AXB (Table 3). The overall treatment ef-
mixing dexmedetomidine with long-acting LA used in a BPB, re- fect for all levels of BPB suggested that dexmedetomidine pro-
gardless of the level, prolongs the duration of sensory block dur- longed the duration of analgesia by at least 63% [72%],
ation compared with LA alone, in patients undergoing upper (P < 0.0001, I2 ¼ 100%), or from 7.5 h to 11.9 h. The level of evi-
extremity surgery (Table 3). The overall quality assessment was dence for this finding was rated as moderate. (Table 3,
downgraded by quality limitations; but was also upgraded by Supplementary Fig. S3) The overall quality assessment was
the large treatment effect and presence of a dose response. downgraded by quality, consistency, and directness limitations;
but was also upgraded by the large treatment effect and dose-
response.
Block characteristics Cumulative 24-h postoperative analgesic consumption was
The effect of perineural dexmedetomidine on motor block dur- reported in eight trials belonging to the ISB43 55 60 64 and SCB28 48
63 65
ation was evaluated in 31 trials.7 9–11 21 22 28 41–54 56–59 61–63 65–67 subgroups; and none of the trials examining ICB or AXB re-
Adding dexmedetomidine to BPB prolonged motor block dur- ported this outcome. Combining dexmedetomidine with LA
ation by at least 26% [50%], (P < 0.0001, I2 ¼ 100%) for ISB; 61% reduced oral morphine equivalent consumption by a mean [95%
[90%], (P < 0.0001, I2 ¼ 100%) for SCB; 19% [38%], (P < 0.0001, CI] of -9.6 mg [-19.4, 0.1], (P ¼ 0.05, I2 ¼ 92%) and -11.6 mg [-15.6, -
I2 ¼ 85%) for ICB; and 11% [35%], (P < 0.0001, I2 ¼ 100%) for AXB 7.6], (P < 0.0001, I2 ¼ 30%) for the ISB and SCB subgroups, respect-
(Fig. 3). The overall treatment effect for all levels of BPB sug- ively. The overall treatment effect for these two levels of BPB
gested that dexmedetomidine prolongs motor block duration by suggested that dexmedetomidine reduced postoperative anal-
at least 58% [68%], (P < 0.0001, I2 ¼ 100%), or from 6.9 h to 10.1 h. gesic consumption by -10.2 mg [-15.3, -5.2], (P< 0.0001, I2 ¼ 83%)
The level of evidence for this finding was rated as high. (Table 3) of oral morphine equivalents. The level of evidence for this find-
The overall quality assessment was downgraded by quality ing was rated as low. (Table 3) The overall quality assessment
limitations; but was also upgraded by the large treatment effect. was downgraded by quality and sparse data limitations.
The effect of perineural dexmedetomidine as a LA adjunct o The effect of perineural dexmedetomidine on posto perative
n the sensory block onset was evaluated in 31 trials.7 9–11 21 22 28 rest pain scores at 24 h was reported in seven trials.28 41 43 55 60 64
41 42 44–56 58–63 65 67 68 65
Dexmedetomidine hastened sensory block Dexmedetomidine reduced pain scores by -0.5 cm [0.9, 0.0],
onset by at least 77% [32%], (P ¼ 0.003, I2 ¼ 99%) for ISB; 44% (P ¼ 0.03, I2 ¼ 0%) and 0.8 cm [1.6, 0.0], (P ¼ 0.04, I2 ¼ 0) for SCB
[33%], (P < 0.0001, I2 ¼ 94%) for SCB; 38% [31%], (P < 0.0001, I2 ¼ 0%) and AXB subgroups, respectively but there were no differences
for ICB; and 24% [18%], (P < 0.0001, I2 ¼ 64%) for AXB (Table 3). in the ISB and ICB subgroups. The level of evidence for this find-
The overall treatment effect for all levels of BPB suggested that ing was rated as low. (Table 3) The overall quality assessment
dexmedetomidine shortened sensory block onset time by at was downgraded by quality and sparse data limitations. Rest
least 40% [28%], (P < 0.0001, I2 ¼ 98%), or from 20.0 min to pain at 12 h and length of hospital stay time were inconsistently
10.8 min. The level of evidence for this finding was rated as assessed in the reviewed trials; while dynamic pain was re-
moderate. (Table 3, Supplementary Fig. S1) The overall quality ported in one trial only,55 precluding any conclusions regarding
assessment was downgraded by quality and consistency limita- these outcomes.
tions; but was also upgraded by the large treatment effect. Patient satisfaction with the pain management received
The effect of perineural dexmedetomidine on motor block and/or willingness to receive the same management in the fu-
onset was assessed in 27 trials.7 9–11 22 28 42 44–56 58 59 61–63 65 67 ture was assessed in six trials.8 22 41 45 52 60 The proportion of pa-
Dexmedetomidine hastened motor block onset by at least 35% tients with high satisfaction was greater among those who
[26%], (P < 0.0001, I2 ¼ 96%) for SCB; 40% [31%], (P < 0.0001, I2 ¼ 0%) received dexmedetomidine, with an OR [95% CI] of 2.5 [1.2, 5.3],
for ICB; and 17% [15%], (P < 0.0001, I2 ¼ 26%) for AXB (Table 3). (P ¼ 0.02, I2 ¼ 49%), compared with Control. The level of evidence
Though both trials52 55 examining the effect of dexmedetomi- for this finding was rated as very low. (Table 3) The overall qual-
dine on motor block onset time in ISB showed faster motor ity assessment was downgraded by quality, consistency and
block onset, the pooled results for this specific brachial plexus sparse data limitations.
level was not significant. The overall treatment effect for all lev-
els of BPB suggested that dexmedetomidine shortened motor
block onset time by at least 39% [27%], (P < 0.00001, I2 ¼ 99%), or
Dexmedetomidine-related adverse effects
from 21.2 min to 13.4 min. The level of evidence for this finding The definitions of dexmedetomidine-related adverse effects in
was rated as moderate. (Table 3, Supplementary Fig. S2) The the reviewed trials were diverse; therefore, we reported these
overall quality assessment was downgraded by quality and con- outcomes as ‘standardized units’. Dexmedetomidine increased
sistency limitations; but was also upgraded by the large treat- the odds of bradycardia by an OR of 3.3 [0.8, 13.5], (P ¼ 0.0002,
ment effect. I2 ¼ 70%); it also increased the odds of hypotension by an OR of
5.4 [2.7, 11.0], (P < 0.0001, I2 ¼ 0%). The level of evidence for these
findings was rated as moderate. (Table 3) The overall quality as-
Analgesic outcomes sessment was downgraded by quality and consistency limita-
The effect of perineural dexmedetomidine on duration of post- tions. Notably, these side-effects were transient, reversible, did
operative analgesia was evaluated in 26 trials.7 9–11 22 41 43 45–53 56 not require any intervention, and did not cause any long-term
57 59 61–67
The definition of duration of analgesia in these trials consequence in any of the patients. Our additional subgroup
varied to include time to first analgesic request,7 9 11 41 47 49 50 56 analysis suggested that a dose of 50-60 mg maximizes sensory
57 61 63 65 67
to attain a pain VAS score > 359 or > 4,22 43 62 or to first block duration (by at least 52% [61%], P < 0.0001), while minimiz-
patient report of postoperative pain at surgical site,10 64 66 and ing the haemodynamic side-effects to none [i.e. by an OR of 1.7

Downloaded from https://academic.oup.com/bja/article-abstract/118/2/167/2924212/Evidence-basis-for-using-perineural


by guest
on 26 September 2017
by guest
Table 3 Summary of results and GRADE35 of evidence. Abbreviations: cm, centimetre; Dex, Dexmedetomidine; h, hours; min, minute; NA, not applicable; PONV, postoperative nausea and
vomiting

on 26 September 2017
Time-to-event Number of References of studies Dex N Dex Control Control Ratio of Means P-Value for P-Value for I2 Test for Quality of
176

outcomes studies included Mean N Mean [95% Confidence statistical heterogeneity heterogeneity evidence
|

included Interval] significance (GRADE)

Sensory block 31 7, 9, 10, 11, 21, 22, 28, 41, 953 10.8 909 20.0 0.7 [0.6, 0.8] < 0.00001 < 0.00001 98% 丣丣丣両, Moderate
onset (min) 42, 43, 44, 45, 46, 47, 48,
49, 50, 51, 52, 53, 54, 55,
56, 58, 59, 60, 61, 62, 65,
67, 63
Sensory block 32 7, 9, 10, 11, 21, 22, 28, 41, 970 691.8 924 460.6 1.7 [1.6, 1.7] < 0.00001 < 0.00001 100% 丣丣丣丣, High
Vorobeichik et al.

duration (min) 42, 43, 44, 45, 46, 47, 48,


49, 50, 51, 52, 53, 54, 55,
56, 57, 58, 59, 61, 62, 63,
65, 66, 67
Motor block 27 7, 9, 10, 11, 22, 28, 42, 44, 860 13.4 815 21.2 0.7 [0.6, 0.9] < 0.00001 < 0.00001 99% 丣丣丣両, Moderate
onset (min) 45, 46, 47, 48, 49, 50, 51,
52, 53, 54, 55, 56, 58, 59,
61, 62, 63, 65, 67
Motor block 31 9, 7, 10, 11, 22, 28, 41, 42, 957 605.9 909 412.9 1.7 [1.6, 1.8] < 0.00001 < 0.00001 100% 丣丣丣丣, High
duration (min) 43, 44, 45, 46, 47, 48, 49,
50, 51, 52, 53, 54, 56, 57,
58, 59, 61, 62, 63, 64, 65,
66, 67
Duration of 26 7, 9, 10, 11, 22, 28, 41, 43, 787 716.5 754 452.4 1.7 [1.6, 1.8] < 0.00001 < 0.00001 100% 丣丣丣両, Moderate
analgesia (min) 45, 46, 47, 48, 49, 50, 51,
52, 53, 56, 57, 59, 61, 62,
63, 64, 65, 66
Analgesic Number of References of studies Dex N Dex Control Control Odds Ratio or P-Value for P-Value for I2 Test for Quality of
outcomes studies included (Mean N (Mean Weighed Mean[95% statistical heterogeneity heterogeneity evidence
included or n/N) or n/N) Confidence Interval) significance (GRADE)
Analgesic 8 28, 43, 48, 55, 60, 63, 64, 65 234 26.7 234 36.0 10.2 [-15.3, -5.2] < 0.0001 < 0.00001 83% 丣丣両両, Low
consumption
Rest pain scores 7 9, 28, 41, 55, 60, 64, 65 207 3.4 206 3.9 0.6 [-0.9, -0.3] 0.0002 0.5 0% 丣丣両両, Low
at 24 h (cm)
Dynamic pain 1 55 31 3.9 30 3.5 0.5 [-0.9, 1.8] 0.48 NA NA N/A
scores at 24 h (cm)

Downloaded from https://academic.oup.com/bja/article-abstract/118/2/167/2924212/Evidence-basis-for-using-perineural


Patient satisfaction 6 22, 41, 45, 52, 60, 64 183 151/183 186 124/186 2.5 [1.2, 5.3] 0.02 0.08 49% 丣両両両, Very low
Dex-related Number of References of studies Dex N Dex Control Control Odds Ratio or Weighed P-Value for P-Value for I2 Test for Quality of
adverse effects studies included (Mean N (Mean Mean[95% Confidence statistical heterogeneity heterogeneity evidence
included or n/N) or n/N) Interval) significance (GRADE)
Bradycardia 19 7, 10, 11, 21, 22, 42, 45, 49, 540 69/540 525 10/525 4.8 [1.2, 19.2] 0.03 0.001 66% 丣丣丣両, Moderate
51, 54, 56, 57, 58, 60, 61,
62, 63, 64, 65
Hypotension 14 7, 10, 11, 21, 42, 49, 54, 57, 410 50/410 395 15/395 5.4 [2.7, 11.0] < 0.00001 0.99 0% 丣丣丣両, Moderate
60, 61, 62, 63, 64, 65
Excessive sedation 8 9, 28, 44, 46, 49, 55, 56, 65 222 53/222 220 4/220 17.2 [1.0, 286.5] 0.05 0.001 78% 丣両両両, Very low
Hypoxemia 11 7, 11, 21, 22, 42, 49, 56, 57, 328 0/328 327 0/327 NA NA NA NA NA
61, 63, 65
PONV 15 7, 9, 11, 21, 22, 43, 51, 55, 386 19/386 386 43/386 0.4 [0.2, 1.0] 0.06 0.08 49% 丣丣両両, Low
56, 57, 60, 63, 64, 65
Perineural dexmedetomidine for brachial plexus block | 177

Ratio of means Ratio of means


Study or subgroup Ratio of means SE Weight IV, random, 95% CI IV, random, 95% CI
1.4.1 ISB
Abdallah 2016 (64) 1.0649 0.0015 3.3% 1.06 [1.06, 1.07]
Kumar 2014 (43) 2.1685 0.0684 3.2% 2.17 [2.03, 2.30]
Rashmi 2016 (52) 1.3371 0.0068 3.3% 1.34 [1.32, 1.35]
Subtotal (95% CI) 9.9% 1.50 [1.26, 1.74]
Heterogeneity: Tau2=0.04; χ2=1782.28, df=2 (P<0.00001); I2=100%
Test for overall effect: Z=12.37 (P<0.00001)

1.4.2 SCB
Agarwal 2014 (56) 3.375 0.0386 3.3% 3.38 [3.30, 3.45]
Bharti 2015 (65) 1.3 0.0539 3.2% 1.30 [1.19, 1.41]
Biswas 2014 (57) 1.6406 0.0241 3.3% 1.64 [1.59, 1.69]
Das 2014 (28) 1.6892 0.0404 3.3% 1.69 [1.61, 1.77]
Das 2016 (67) 1.2078 0.0697 3.2% 1.21 [1.07, 1.34]
Dixit 2015 (44) 1.522 0.0282 3.3% 1.52 [1.47, 1.58]
Ghandi 2012 (10) 6.5561 0.0825 3.1% 6.56 [6.39, 6.72]
Gurajala 2015 (62) 2.1538 0.1221 2.9% 2.15 [1.91, 2.39]
Kathuria 2015 (63) 1.9456 0.0918 3.1% 1.95 [1.77, 2.13]
Kaur 2015 (45) 0.6627 0.0271 3.3% 0.66 [0.61, 0.72]
Khade 2013 (46) 1.9993 0.0452 3.3% 2.00 [1.91, 2.09]
Kwon 2015 (58) 1.2679 0.0587 3.2% 1.27 [1.15, 1.38]
Manohar 2015 (22) 1.2408 0.0293 3.3% 1.24 [1.18, 1.30]
Nema 2014 (47) 1.402 0.0668 3.2% 1.40 [1.27, 1.53]
Patki 2015 (48) 1.3144 0.0067 3.3% 1.31 [1.30, 1.33]
Saraf 2016 (53) 1.6442 0.0359 3.3% 1.64 [1.57, 1.71]
Singh 2016 (49) 1.9085 0.1034 3.0% 1.91 [1.71, 2.11]
Tandon 2016 (51) 1.5165 0.0209 3.3% 1.52 [1.48, 1.56]
Subtotal (95% CI) 58.0% 1.90 [1.61, 2.20]
Heterogeneity: Tau2=0.42; χ2=7985.11, df=17 (P<0.00001); I2=100%
Test for overall effect: Z=12.46 (P<0.00001)

1.4.3 ICB
Ammar 2012 (9) 1.4711 0.0336 3.3% 1.47 [1.41, 1.54]
Mirkheshti 2014 (59) 1.453 0.1482 2.7% 1.45 [1.16, 1.74]
Song 2014 (66) 1.2333 0.0551 3.2% 1.23 [1.13, 1.34]
Subtotal (95% CI) 9.2% 1.38 [1.19, 1.56]
Heterogeneity: Tau2=0.02; χ2=13.67, df=2 (P=0.001); I2=85%
Test for overall effect: Z=14.40 (P<0.00001)

1.4.4 AXB
Arun 2016 (50) 1.0065 0.0065 3.3% 1.01 [0.99, 1.02]
Bangera 2016 (61) 1.3546 0.0289 3.3% 1.35 [1.30, 1.41]
Esmaoglu 2010 (7) 1.3443 0.0274 3.3% 1.34 [1.29, 1.40]
Hanoura 2013 (41) 1.0519 0.0375 3.3% 1.05 [0.98, 1.13]
Karthik 2015 (42) 1.4849 0.003 3.3% 1.48 [1.48, 1.49]
Kaygusuz 2012 (11) 1.489 0.0245 3.3% 1.49 [1.44, 1.54]
Zhang 2014 (54) 1.7305 0.1021 3.0% 1.73 [1.53, 1.93]
Subtotal (95% CI) 22.9% 1.35 [1.11, 1.58]
Heterogeneity: Tau2=0.10; χ2=4582.20, df=6 (P<0.00001); I2=100%
Test for overall effect: Z=11.36 (P<0.00001)

Total (95% CI) 100.0% 1.68 [1.58, 1.79]


Heterogeneity: Tau2=0.09; χ2=27788.77, df=30 (P<0.00001); I2=100%
–2 –1 0 1 2
Test for overall effect: Z=30.83 (P<0.00001)
Favours control Favours dexmedetomidine
Test for subgroup differences: χ2=10.17, df=3 (P=0.02); I2=70.5%

Fig 3 Forest plot depicting motor block duration. The individual trials’ ratio of means, standard error, and the pooled estimates of the ratio of means are shown.
The 95% confidence intervals are shown as lines for individual studies and as diamonds for pooled estimates. Abbreviations: AXB, axillary block; CI, confidence
interval; ICB, infraclavicular block; ISB, interscalene block; SCB, supraclavicular block; SE, standard error.

Downloaded from https://academic.oup.com/bja/article-abstract/118/2/167/2924212/Evidence-basis-for-using-perineural


by guest
on 26 September 2017
178 | Vorobeichik et al.

(0.3, 9.6), P ¼ 0.5 and OR of 6.9 (0.8, 60.8), P ¼ 0.08 for bradycardia and treated. Furthermore, these side-effects may preclude use
and hypotension, respectively]. (Supplementary Fig. S4) in higher risk patients and in procedures inherently associated
Excessive postoperative sedation was reported using various with changes in heart rate and bp, such as surgery in the sitting
scales, including Richmond,49 56 Modified Wilson,46 University position.73
of Michigan,44 and four-point sedation scales;65 it was un- This meta-analysis has positive safety implications. While
defined in three trials.9 55 67 Patients who received perineural our previous meta-analysis8 could not draw conclusions about
dexmedetomidine had greater odds of experiencing excessive the safety of the clinical use of dexmedetomidine, the present
postoperative sedation, with an OR of 17.2 [1.04, 286.5], (P ¼ 0.05, review incorporates data from 1026 patients who received peri-
I2 ¼ 78%). The level of evidence for this finding was rated as very neural dexmedetomidine for the BPB, and none developed any
low. (Table 3) The overall quality assessment was downgraded neurotoxicity symptoms, or any other neurologic sequalae. In
by quality, consistency and sparse data limitations. fact, there is further evidence from in vitro and animal studies
Hypoxaemia was defined as oxygen saturation below 90%7 11 suggesting that the perineural application of dexmedetomidine
21 42 49 56 65
, 93%61 or was not defined.22 57 63 None of the patients may be neuroprotective against the LA-induced inflammatory
in the reviewed trials experienced hypoxemic events. response.5 74 75 While this has also been shown for clonidine,76
Furthermore, data from 15 trials7 9 11 21 22 43 44 51 55–57 60 63–65 sug- the latter may carry a risk of neurotoxicity when combined with
gested that there was no statistically significant difference in local anaesthetics.4 Nonetheless, recent evidence suggesting
the incidence of PONV between the two groups. The level of evi- that systemic dexmedetomidine may be as effective as perineu-
dence for this finding was rated as low. The overall quality as- ral dexmedetomidine in prolonging the duration of analgesia
sessment was downgraded by quality and sparse data after BPB, is a promising new area of research,8 though the
limitations. underlying mechanisms and the comparative risk of haemo-
Finally, none of the patients reported any block-related dynamic side-effects have yet to be explored.
complications.

Limitations
Our review has several limitations. First, clinical heterogeneity
Discussion characterized the data reviewed as it originated from different
This is the first study to provide a high level of evidence from surgical, anaesthetic, and analgesic settings. While we stratified
clinical trials supporting the efficacy of perineural dexmedeto- studies according to the level of BPB, considerable variability
midine as peripheral nerve block adjunct. Our updated review may persist within each subgroup. For example, postoperative
demonstrates that using perineural dexmedetomidine as a BPB pain after shoulder rotator cuff repair is likely more severe than
adjunct is associated with important facilitatory effects regard- simple shoulder arthroscopy; but lack of sufficient data pre-
less of the BPB level, specifically prolonged sensory and motor cluded any further stratification by surgical procedure. Second,
block durations, and faster sensory and motor block onset. The definitions of some outcomes of interest, such as postoperative
analgesic benefits of using dexmedetomidine include prolonged analgesia duration, hypoxaemia, and excessive sedation varied
duration of analgesia, reduced cumulative 24 h postoperative between trials, which may have contributed to the observed
analgesic consumption, improved pain control and enhanced heterogeneity. Third, the trials reviewed were small, with sam-
satisfaction with pain relief. However, perineural dexmedetomi- ple sizes of 10-50 patients/group, which increases the chances
dine was also associated with increased risk of transient hypo- of type I error and publication bias. Fourth, only two trials origi-
tension, bradycardia, and postoperative sedation. The levels of nated from North America64 and Europe,55 which may represent
evidence relating to sensory block duration and duration of an- another source of publication bias.7778 Fifth, while a variety of
algesia were high and moderate, respectively. doses of dexmedetomidine were used, we could not detect a
The observed differences in the magnitude of dexmedetomi- dose-response, although evidence suggests that dexmedetomi-
dine effects on the various BPB levels may be attributed to in- dine produces a dose-dependent prolongation in sensory block
trinsic differences in the systemic uptake69 and neuraxial duration.19 This may be as a result of the clinical heterogeneity
spread70 between these levels. This updated systematic review of nerve block techniques and LA doses used. We also did not
and meta-analysis overcomes the limitations of our earlier seek abstracts from meeting and trial results from clinical trial
meta-analysis8 of a small number of trials that had suggested registries, and we restricted our search to trials published in the
lack of efficacy of dexmedetomidine in prolonging sensory block English language. Lastly, it is noteworthy that the methodo-
duration or expediting block onset. However, the present results logical shortcomings of studies and inconsistencies in the defin-
are similarly characterized by high heterogeneity, and while LA ition and assessment of outcomes were main reasons why the
dose-response account for some of this heterogeneity, our find- strength of evidence was down-graded for some outcomes.
ings should still be interpreted with caution. Despite these inconsistencies, the methods of assessing out-
The benefits of dexmedetomidine should be carefully comes indicative of dexmedetomidine effectiveness (i.e. dur-
weighed against prolonged motor block duration and the ations of sensory block, motor block, and postoperative
increased risks of sedation, bradycardia and hypotension. analgesia), were marked by good internal and external validity.
Practically, while extended motor block duration may be desir- Furthermore, factors such as the strength of the treatment ef-
able in certain populations, using dexmedetomidine in lower fect, presence of a dose-response, and successful identification
extremity blocks may delay recovery after ambulatory surgery of confounders served to offset these limitations for some
and increase the risk of falls.71 72 Additionally, sedation may outcomes.
interfere with fast-tracking, recovery room bypass, or other In contrast, our review has several points of strength. The lit-
pathways aimed to expedite discharge. The potential for erature review we conducted was exhaustive and included all
haemodynamic side-effects may limit the administration of relevant databases. Our inclusion criteria were limited to
blocks inclusive of dexmedetomidine to monitored settings, randomized trials. The primary outcome results maintained
where bradycardia and hypotension can be readily identified their robustness despite our attempt to explore statistical

Downloaded from https://academic.oup.com/bja/article-abstract/118/2/167/2924212/Evidence-basis-for-using-perineural


by guest
on 26 September 2017
Perineural dexmedetomidine for brachial plexus block | 179

heterogeneity. These factors underscore the validity of our 7. Esmaoglu A, Yegenoglu F, Akin A, Turk CY.
findings. Dexmedetomidine added to levobupivacaine prolongs axil-
lary brachial plexus block. Anesth Analg 2010; 111: 1548–51
8. Abdallah FW, Brull R. Facilitatory effects of perineural dex-
Conclusion medetomidine on neuraxial and peripheral nerve block: a
Our study provides strong evidence that using perineural dex- systematic review and meta-analysis. Br J Anaesth 2013; 110:
medetomidine improves BPB onset, quality, and analgesia. 915–25
These results differ considerably from our earlier meta-analysis 9. Ammar AS, Mahmoud KM. Ultrasound-guided single injec-
that did not support the use of perineural dexmedetomidine. tion infraclavicular brachial plexus block using bupivacaine
There is now strong evidence to support its effect in prolonging alone or combined with dexmedetomidine for pain control
sensory block duration, and moderate evidence to supports its in upper limb surgery: a prospective randomized controlled
effect in hastening block onset and extending the duration of trial. Saudi J Anaesth 2012; 6: 109–14
analgesia. However, these benefits should be weighed against 10. Gandhi R, Shah A, Patel I. Use of dexemedetomidine along
the increased risks of motor block prolongation and transient with bupivacaine for brachial plexus block. Natl J Med Res
bradycardia and hypotension. 2012; 2: 67–9
11. Kaygusuz K, Kol IO, Duger C, et al. Effects of adding dexmede-
tomidine to levobupivacaine in axillary brachial plexus
Authors’ contributions block. Curr Ther Res Clin Exp 2012; 73: 103–11
12. Moher D, Liberati A, Tetzlaff J, Altman DG. Preferred report-
Study design/planning: F.W.A.
ing items for systematic reviews and meta-analyses: the
Study conduct: L.V., F.W.A.
PRISMA statement. J Clin Epidemiol 2009; 62: 1006
Data analysis: L.V., R.B., F.W.A.
13. Al-Metwalli RR, Mowafi HA, Ismail SA, et al. Effect of intra-
Writing paper: L.V., R.B., F.W.A.
articular dexmedetomidine on postoperative analgesia after
Revising paper: all authors
arthroscopic knee surgery. Br J Anaesth 2008; 101: 395–9
14. Luan H, Zhang X, Jfeng J, Zhu P, Li J, Zhao Z. Effect of dexme-
Supplementary material detomidine added to ropivacaine on ultrasound-guided
transversus abdominis plane block for postoperative
Supplementary material is available at British Journal of
analgesia after abdominal hysterectomy surgery: a prospect-
Anaesthesia online.
ive randomized controlled trial. Minerva Anestesiol 2016; 82:
981–8
Declaration of interest 15. Mohamed SA, Fares KM, Mohamed AA, Alieldin NH.
Dexmedetomidine as an adjunctive analgesic with bupiva-
None declared. caine in paravertebral analgesia for breast cancer surgery.
Pain Physician 2014; 17: E589–98
16. Memis D, Turan A, Karamanlioglu B, Pamukcu Z, Kurt I.
Funding
Adding dexmedetomidine to lidocaine for intravenous re-
This work was supported by departmental funding. Both gional anesthesia. Anesth Analg 2004; 98: 835–40
F.W.A.and R.B. are supported by the Merit Award Program, 17. Esmaoglu A, Mizrak A, Akin A, Turk Y, Boyaci A. Addition of
Department of Anaesthesia, University of Toronto. dexmedetomidine to lidocaine for intravenous regional an-
aesthesia. Eur J Anaesthesiol 2005; 22: 447–51
18. Marhofer D, Kettner SC, Marhofer P, Pils S, Weber M,
References Zeitlinger M. Dexmedetomidine as an adjuvant to ropiva-
1. Boezaart AP, Davis G, Le-Wendling L. Recovery after ortho- caine prolongs peripheral nerve block: a volunteer study. Br J
pedic surgery: techniques to increase duration of pain con- Anaesth 2013; 110: 438–42
trol. Curr Opin Anaesthesiol 2012; 25: 665–72 19. Keplinger M, Marhofer P, Kettner SC, Marhofer D, Kimberger
2. Opperer M, Gerner P, Memtsoudis SG. Additives to local an- O, Zeitlinger M. A pharmacodynamic evaluation of dexme-
esthetics for peripheral nerve blocks or local anesthesia: a detomidine as an additive drug to ropivacaine for peripheral
review of the literature. Pain Manag 2015; 5: 117–28 nerve blockade: a randomised, triple-blind, controlled study
3. Kamibayashi T, Maze M. Clinical uses of alpha2 -adrenergic in volunteers. Eur J Anaesthesiol 2015; 32: 790–6
agonists. Anesthesiology 2000; 93: 1345–9 20. Higgins JP, Altman DG, Gotzsche PC, et al. The Cochrane
4. Williams BA, Hough KA, Tsui BY, Ibinson JW, Gold MS, Collaboration’s tool for assessing risk of bias in randomised
Gebhart GF. Neurotoxicity of adjuvants used in perineural trials. Br Med J 2011; 343: d5928
anesthesia and analgesia in comparison with ropivacaine. 21. Lee MJ, Koo DJ, Choi YS, Lee KC, Kim HY. Dexamethasone or
Reg Anesth Pain Med 2011; 36: 225–30 dexmedetomidine as local anesthetic adjuvants for
5. Brummett CM, Norat MA, Palmisano JM, Lydic R. Perineural ultrasound-guided axillary brachial plexus blocks with nerve
administration of dexmedetomidine in combination with stimulation. Korean J Pain 2016; 29: 29–33
bupivacaine enhances sensory and motor blockade in sciatic 22. Manohar P, Prakash M. Comparison of the effects of fentanyl
nerve block without inducing neurotoxicity in rat. and dexmedetomidine in supraclavicular brachial plexus
Anesthesiology 2008; 109: 502–11 block achieved with 0.5% bupivacaine in Karpaga Vinayaga
6. Brummett CM, Hong EK, Janda AM, Amodeo FS, Lydic R. Medical College and Hospital, Maduranthagam. JMSCR 2015;
Perineural dexmedetomidine added to ropivacaine for sci- 3: 7131–8
atic nerve block in rats prolongs the duration of analgesia by 23. Anonymous Opioids, CPS 2016. Compendium of
blocking the hyperpolarization-activated cation current. Pharmaceuticals and Specialties, Jovaisas B, ed. Ottawa,
Anesthesiology 2011; 115: 836–43 Canada: Canadian Pharmacists Association, 2016; 2334

Downloaded from https://academic.oup.com/bja/article-abstract/118/2/167/2924212/Evidence-basis-for-using-perineural


by guest
on 26 September 2017
180 | Vorobeichik et al.

24. Ebert TJ, Hall JE, Barney JA, Uhrich TD, Colinco MD. The ef- 50 lg Dexmedetomidine as an adjuvant for interscalene bra-
fects of increasing plasma concentrations of dexmedetomi- chial plexus block: prospective clinical study. J Evol Med Dent
dine in humans. Anesthesiology 2000; 93: 382–94 Sci 2014; 3: 13111–9
25. Higgins JPT, Deeks JJ. Chapter 7: Selecting Studies and Collecting 44. Dixit A, Singhal S, Neema C, Sanwatsarkar S, Bhatia M. An
Data, Cochrane Handbook for Systematic Reviews of Interventions. evaluation of the addition of Dexmedetomidine to
JPT Higgins, JJ Deeks and S Green, eds. Sussex, England: The Levobupivacaine for supraclavicular brachial plexus block in
Cochrane Collaboration, 2011 upper limb orthopaedic surgeries. Asian Pac J Health Sci 2015;
26. Hozo SP, Djulbegovic B, Hozo I. Estimating the mean and 2: 148–53
variance from the median, range, and the size of a sample. 45. Kaur H, Singh G, Rani S, et al. Effect of dexmedetomidine as
BMC Med Res Methodol 2005; 5: 13 an adjuvant to levobupivacaine in supraclavicular brachial
27. Breivik EK, Bjornsson GA, Skovlund E. A comparison of pain plexus block: a randomized double-blind prospective study.
rating scales by sampling from clinical trial data. Clin J Pain J Anaesthesiol Clin Pharmacol 2015; 31: 333–8
2000; 16: 22–8 46. Khade AR, Makwana JC, Jethva NK, Bansal S, Chudasama P,
28. Das A, Majumdar S, Halder S, et al. Effect of dexmedetomi- Chadha IA. Evaluation of effect of Dexmeditomidine as an
dine as adjuvant in ropivacaine-induced supraclavicular adjuvant to bupivacaine in supraclavicular brachial plexus
brachial plexus block: a prospective, double-blinded and block. NJIRM 2013; 4: 122–7
randomized controlled study. Saudi J Anaesth 2014; 8: S72–7 47. Nema N, Badgaiyan H, Raskaran S, et al. Effect of addition of
29. Furukawa TA, Barbui C, Cipriani A, Brambilla P, Watanabe N. Dexmedetomidine to ropivacaine hydrochloride (0.75%) in
Imputing missing standard deviations in meta-analyses can brachial plexus block through supraclavicular route in upper
provide accurate results. J Clin Epidemiol 2006; 59: 7–10 limb surgeries: a clinical comparative study. JEMDS 2014; 3:
30. Higgins JPT, Deeks JJ, Altman DG. Chapter 16: Special Topics In 12612–21
Statistics, Cochrane Handbook for Systematic Reviews of 48. Patki YS, Bengali R, Patil T. Efficacy of Dexmedetomidine as
Interventions. JPT Higgins and JJ Deeks, eds. Sussex, England: an adjuvant to 0.5% ropivacaine in supraclavicular brachial
The Cochrane Collaboration, 2011 plexus block for postoperative analgesia. IJSR 2015; 4: 2345
31. Newell DJ. Intention-to-treat analysis: implications for quan- 49. Singh AP, Mahindra M, Gupta R, Singh Bajwa SJ.
titative and qualitative research. Int J Epidemiol 1992; 21: Dexmedetomidine as an adjuvant to levobupivacaine in
837–41 supraclavicular brachial plexus block: a novel anesthetic ap-
32. Moore A, Moore O, McQuay H, Gavaghan D. Deriving dichot- proach. Anesth Essays Res 2016; 10: 414–19
omous outcome measures from continuous data in rando- 50. Arun S. Effect of dexmedetomidine as an adjuvant to 0.75%
mised controlled trials of analgesics: use of pain intensity ropivacaine in axillary brachial plexus block for forearm and
and visual analogue scales. Pain 1997; 69: 311–5 hand surgeries. IJBR 2016; 7: 187–92
33. Wallace BC, Schmid CH, Lau J, Trikalinos TA. Meta-analyst: 51. Tandon N. A comparative clinical study to evaluate the effi-
software for meta-analysis of binary, continuous and diag- cacy of levobupivacaine with clonidine and levobupivacaine
nostic data. BMC Med Res Methodol 2009; 9: 2288–9 with dexmedetomidine in supraclavicular brachial plexus
34. DerSimonian R, Laird N. Meta-analysis in clinical trials re- block. JEMDS 2016; 5: 925
visited. Contemp Clin Trials 2015; 45: 139–45 52. Rashmi HD, Komala HK. Effect of Dexmedetomidine As an Adjuvant
35. Atkins D, Best D, Briss PA, et al. Grading quality of evidence to 0.75% Ropivacaine in Interscalene Brachial Plexus Block Using
and strength of recommendations. Br Med J 2004; 328: 1490 Nerve Stimulator: A Prospective, Randomized Double-Blind Study.
36. Friedrich JO, Adhikari NK, Beyene J. Ratio of geometric means Mumbai, India: Anesthesia Essays and Researches. In press
to analyze continuous outcomes in meta-analysis: compari- 53. Saraf S. Dexmedetomidine as adjuvant to ropivacaine in bra-
son to mean differences and ratio of arithmetic means using chial plexus block-a dose response study. PIJR 2016; 5: 397
empiric data and simulation. Stat Med 2012; 31: 1857–86 54. Zhang Y, Wang CS, Shi JH, et al. Perineural administration of
37. Friedrich JO, Adhikari NK, Beyene J. Ratio of means for ana- dexmedetomidine in combination with ropivacaine pro-
lyzing continuous outcomes in meta-analysis performed as longs axillary brachial plexus block. Int J Clin Exp Med 2014; 7:
well as mean difference methods. J Clin Epidemiol 2011; 64: 680–5
556–64 55. Fritsch G, Danninger T, Allerberger K, et al. Dexmedetomi-
38. Higgins JP, Thompson SG. Quantifying heterogeneity in a dine added to ropivacaine extends the duration of intersca-
meta-analysis. Stat Med 2002; 21: 1539–58 lene brachial plexus blocks for elective shoulder surgery
39. Egger M, Davey Smith G, Schneider M, Minder C. Bias in when compared with ropivacaine alone a single-center, pro-
meta-analysis detected by a simple, graphical test. Br Med J spective, triple-blind, randomized controlled trial. Reg Anesth
1997; 315: 629–34 Pain Med 2014; 39: 37–47
40. Dar FA, Najar MR, Jan N. Dexmedetomidine added to ropiva- 56. Agarwal S, Aggarwal R, Gupta P. Dexmedetomidine prolongs
caine prolongs axillary brachial plexus block. IJBAR 2013; 4: the effect of bupivacaine in supraclavicular brachial plexus
719–22 block. J Anaesthesiol Clin Pharmacol 2014; 30: 36–40
41. Hanoura S, Elsayed M, Abdullah A, Elsayed H, Nor Eldeen T. 57. Biswas S, Das RK, Mukherjee G, Ghose T. Dexmedetomidine
Dexmedetomidine improves the outcome of a bupivacaine an adjuvant to levobupivacaine in supraclavicular brachial
brachial plexus axillary block: a prospective comparative plexus block: a randomized double blind prospective study.
study. Ain-Shams J Anaesthesiol 2013; 6: 58–62 Ethiop J Health Sci 2014; 24: 203–8
42. Anandh K, Bhupal JPS, Grewal TK, Bindra TK, Verma R. 58. Kwon Y, Hwang SM, Lee JJ, Kim JH. The effect of dexmedeto-
Dexmedetomidine prolongs the effect of 0.5% isobaric levo- midine as an adjuvant to ropivacaine on the bispectral index
bupivacaine in axillary brachial plexus block. J Evol Med Dent for supraclavicular brachial plexus block. Korean J Anesthesiol
Sci 2015; 4: 1015–22 2015; 68: 32–6
43. Kumar AN. Comparative study between 0.25% Bupivacaine 59. Mirkheshti A, Saadatniaki A, Salimi A, Manafi Rasi A,
with 8 M.G Dexamethasone and 0.25% Bupivacaine with Memary E, Yahyaei H. Effects of dexmedetomidine versus

Downloaded from https://academic.oup.com/bja/article-abstract/118/2/167/2924212/Evidence-basis-for-using-perineural


by guest
on 26 September 2017
Perineural dexmedetomidine for brachial plexus block | 181

ketorolac as local anesthetic adjuvants on the onset and dur- techniques of brachial plexus blockade. Anaesthesia 2007; 62:
ation of infraclavicular brachial plexus block. Anesth Pain 1008–14
Med 2014; 4: e17620 70. Stundner O, Meissnitzer M, Brummett CM, et al. Comparison
60. Bengisun ZK, Ekmekci P, Akan B, Koroglu A, Tuzuner F. The of tissue distribution, phrenic nerve involvement, and epi-
effect of adding dexmedetomidine to levobupivacaine for dural spread in standard- vs low-volume ultrasound-guided
interscalene block for postoperative pain management after interscalene plexus block using contrast magnetic reson-
arthroscopic shoulder surgery. Clin J Pain 2014; 30: 1057–61 ance imaging: a randomized, controlled trial. Br J Anaesth
61. Bangera A, Manasa M, Krishna P. Comparison of effects of 2016; 116: 405–12
ropivacaine with and without dexmedetomidine in axillary 71. Johnson RL, Kopp SL, Hebl JR, Erwin PJ, Mantilla CB. Falls and
brachial plexus block: a prospective randomized double- major orthopaedic surgery with peripheral nerve blockade: a
blinded clinical trial. Saudi J Anaesth 2016; 10: 38–44 systematic review and meta-analysis. Br J Anaesth 2013; 110:
62. Gurajala I, Thipparampall AK, Durga P, Gopinath R. Effect of 518–28
perineural dexmedetomidine on the quality of supraclavicular 72. Atkinson HD, Hamid I, Gupte CM, Russell RC, Handy JM.
brachial plexus block with 0.5% ropivacaine and its interaction Postoperative fall after the use of the 3-in-1 femoral nerve
with general anaesthesia. Indian J Anaesth 2015; 59: 89–95 block for knee surgery: a report of four cases. J Orthop Surg
63. Kathuria S, Gupta S, Dhawan I. Dexmedetomidine as an ad- (Hong Kong) 2008; 16: 381–4
juvant to ropivacaine in supraclavicular brachial plexus 73. Laflam A, Joshi B, Brady K, et al. Shoulder surgery in the
block. Saudi J Anaesth 2015; 9: 148–54 beach chair position is associated with diminished cerebral
64. Abdallah FW, Dwyer T, Chan VW, et al. IV and perineural autoregulation but no differences in postoperative cognition
dexmedetomidine similarly prolong the duration of anal- or brain injury biomarker levels compared with supine pos-
gesia after interscalene brachial plexus block: a randomized, itioning: the anesthesia patient safety foundation beach
three-arm, triple-masked, placebo-controlled trial. chair study. Anesth Analg 2015; 120: 176–85
Anesthesiology 2016; 124: 683–95 74. Tufek A, Kaya S, Tokgoz O, Firat U, Evliyaoglu O, Celik F,
65. Bharti N, Sardana DK, Bala I. The analgesic efficacy of dex- Karaman H. The protective effect of dexmedetomidine on
medetomidine as an adjunct to local anesthetics in supracla- bupivacaine-induced sciatic nerve inflammation is medi-
vicular brachial plexus block: a randomized controlled trial. ated by mast cells. Clin Invest Med 2013; 36: E95–102
Anesth Analg 2015; 121: 1655–60 75. Huang Y, Lu Y, Zhang L, Yan J, Jiang J, Jiang H. Perineural dex-
66. Song JH, Shim HY, Lee TJ, et al. Comparison of dexmedetomi- medetomidine attenuates inflammation in rat sciatic nerve
dine and epinephrine as an adjuvant to 1% mepivacaine in via the NF-kappaB pathway. Int J Mol Sci 2014; 15: 4049–59
brachial plexus block. Korean J Anesthesiol 2014; 66: 283–9 76. Lavand’homme PM, Eisenach JC. Perioperative administra-
67. Das B, Lakshmegowda M, Sharma M, Mitra S, Chauhan R. tion of the alpha2-adrenoceptor agonist clonidine at the site
Supraclavicular brachial plexus block using ropivacaine of nerve injury reduces the development of mechanical
alone or combined with dexmedetomidine for upper limb hypersensitivity and modulates local cytokine expression.
surgery: a prospective, randomized, double-blinded, com- Pain 2003; 105: 247–54
parative study. Rev Esp Anestesiol Reanim 2016; 63: 135–40 77. Sleem H, Abdelhai RA, Al-Abdallat I, et al. Development of an
68. Kumar A, Sharma DK, Datta B. Addition of ketamine or dex- accessible self-assessment tool for research ethics commit-
medetomidine to lignocaine in intravenous regional anes- tees in developing countries. J Empir Res Hum Res Ethics 2010;
thesia: a randomized controlled study. J Anaesthesiol Clin 5: 85–96; quiz 97-8
Pharmacol 2012; 28: 501–4 78. Abou-Zeid A, Afzal M, Silverman HJ. Capacity mapping of na-
69. Rettig HC, Lerou JG, Gielen MJ, Boersma E, Burm AG. The tional ethics committees in the Eastern Mediterranean
pharmacokinetics of ropivacaine after four different Region. BMC Med Ethics 2009; 108: 6939–10

Handling editor: P. S. Myles

Downloaded from https://academic.oup.com/bja/article-abstract/118/2/167/2924212/Evidence-basis-for-using-perineural


by guest
on 26 September 2017

You might also like