Impact of pregabalin on acute and persistent postoperative pain: a systematic review and meta-analysis

Similar documents
Benefit and harm of pregabalin in acute pain treatment: a systematic review with meta-analyses and trial sequential analyses

Combined spinalepidural. epidural analgesia in labour (review) By Neda Taghizadeh

Gabapentin Does Not Improve Analgesia Outcomes For Total Joint Replacement. Manyat Nantha-Aree, MD

Efficacy of postoperative epidural analgesia Block B M, Liu S S, Rowlingson A J, Cowan A R, Cowan J A, Wu C L

ENHANCED RECOVERY PROTOCOLS FOR KNEE REPLACEMENT

Effect of Single-dose Preoperative Pregabalin on Postoperative Pain after Cardiac Surgery: A Prospective Observational Randomized Double-blind Study

Systematic Review. Pain Physician 2017; 20: ISSN

Evaluation of a single preoperative dose of pregabalin for attenuation of postoperative pain after laparoscopic cholecystectomy

Screening - inclusion criteria

Screening - inclusion criteria

Lumbar Fusion. Reference Guide for PACU CLINICAL PATHWAY. All patient variances to the pathway are to be circled and addressed in the progress notes.

Oral Oxycodone for Acute Postoperative Pain: A Review of Clinical Trials. Pain Physician Opioid Special Issue 2017; 20:SE33-SE52

Nerve Blocks & Long Acting Analgesia for Plastic Surgeons. Karol A Gutowski, MD, FACS

Anaesthesia and Pain Management for Endo Exo Femoral Prosthesis (EEFP) Bridging the Gap from Surgery to Rehabilitation

Show Me the Evidence: Epidurals, PVBs, TAP Blocks Christopher L. Wu, MD Professor of Anesthesiology The Johns Hopkins Hospital

Satisfactory Analgesia Minimal Emesis in Day Surgeries. (SAME-Day study) A Randomized Control Trial Comparing Morphine and Hydromorphone

THE EFFECTS OF PREOPERATIVE PREGABALIN ON POSTOPERATIVE ANALGESIA AND MORPHI- NE CONSUMPTION AFTER ABDOMINAL HYSTERECTOMY

Postoperative pain management: Analgesics, algorithms and patient activation

Gabapentin and Pregabalin for the Acute Post-operative Pain Management. A Systematic-narrative Review of the Recent Clinical Evidences

Use of Pregabalin for Postoperative Pain: Outcomes in 2 Trials

Balanced Analgesia With NSAIDS and Coxibs. Raymond S. Sinatra MD, Ph.D

Intravenous lidocaine infusions. Dr Ian McConachie FRCA FRCPC

MD (Anaesthesiology) Title (Plan of Thesis) (Session )

Meta Analysis. David R Urbach MD MSc Outcomes Research Course December 4, 2014

Peri operative pain control. Disclosure. Objectives 9/1/2011. No current conflicts of interest

Nonsteroidal anti-inflammatory drugs and perioperative bleeding in paediatric tonsillectomy (Review)

Digital RIC. Rhode Island College. Linda M. Green Rhode Island College

Perioperative Pain Management

Systematic reviews and meta-analyses of observational studies (MOOSE): Checklist.

The Egyptian Journal of Hospital Medicine (July 2018) Vol. 72 (1), Page

Regional anaesthesia in paediatric day case surgery. PA Lönnqvist Karolinska Institutet Karolinska University Hospital Stockholm, Sweden

The intensity of preoperative pain is directly correlated with the amount of morphine needed for postoperative analgesia

Medicine. The Effect of Gabapentin on Acute Postoperative Pain in Patients Undergoing Total Knee Arthroplasty. A Meta-Analysis

Medicine. Chao Han (MD) a, Xiao-dan Li (MD) b, Hong-qiang Jiang (MD) a, Jian-xiong Ma (PhD) a, Xin-long Ma (MD) a, Systematic Review and Meta-Analysis

GUIDELINEs ON PAIN MANAGEMENT IN UROLOGY

Pharmacotherapy for the prevention of chronic pain after surgery in adults(review)

TITLE: Pregabalin for Acute Pain: A Review of the Clinical Effectiveness

Current evidence in acute pain management. Jeremy Cashman

Perioperative Pregabalin & Ketamine as Multimodal Pain Management Strategies

Post-caesarean analgesia. Genevieve Goulding Royal Brisbane & Women's Hospital 1

Over half of the patients using opioids chronically started with acute pain. [postoperative (27%) and injury-related pain (27%)]

Study of Efficacy of the Use of Peri-Operative Pregabalin and Dexamethasone on Post-Operative Pain in Patients undergoing Lumbar Laminectomy

PFIZER INC. THERAPEUTIC AREA AND FDA APPROVED INDICATIONS: See United States Package Insert (USPI)

Evidenzbasiertes, perioperatives Analgesie- Konzept in der Fast Track Chirurgie. Christoph Konrad Luzern

MD (Anaesthesiology) Title (Plan of Thesis) (Session )

Post Caesarean Analgesia An Update. Kim Ekelund MD, PhD, associate professor Rigshospitalet Copenhagen, Denmark

To staple or to sew. Zeng Xuan Hu

Acute Peri-Operative Pain Management Strategies

Acute painful crisis in patients with sickle cell disease: Clinical Guidelines (HN-506a)

Research and Reviews: Journal of Medical and Health Sciences

Screening - inclusion criteria

Opioid Free Anesthesia

Analgesia after c delivery - wound infusions, tap blocks and intrathecal opioids; what more can we offer our patients?

International Journal of Health Sciences and Research ISSN:

Mr David A McDonald Service Improvement Manager Whole System patient Flow Improvement Programme Scottish Government

Learning Objectives. Perioperative goals. Acute Pain in the Chronic Pain Patient for Ambulatory Surgery 9/8/16

(For National Authority Use Only) Name of Study Drug: to Part of Dossier:

Turlough O Hare, MD, FRCPC, MSc Assistant Clinical Professor, Department of Anesthesia, St. Joseph s Healthcare Hamilton McMaster University

A randomised, double-blind, parallel group, multicentre study to compare the tolerability, safety, and efficacy of oxycodone with morphine in

Kayalvizhi 1, J. Radhika 1* Original Research Article. Abstract

Specific Care Question : Question Originator: Plain Language Summary from The Office of Evidence Based Practice: Conditional Recommendation

Parecoxib, Celecoxib and Paracetamol for Post Caesarean Analgesia: A Randomised Controlled Trial

Professor Richard Langford Barts and The London NHS Trust. South Thames Acute Pain 10 th Nov 2011

Acute pain management in laparoscopic cholecystectomy: Is there a role for pregabalin? A review

Analgesia for ERAS programs. Dr Igor Lemech VMO Anaesthetist Wagga Wagga Base Hospital

Review Article Preoperative single dose of pregabalin alleviates postoperative pain: systematic review and meta-analysis

Effective pain management begins with OFIRMEV (acetaminophen) injection FIRST Proven efficacy with rapid reduction in pain 1

Peripheral regional anaesthesia and outcome: lessons learned from the last 10 years

Associate Professor Supranee Niruthisard Department of Anesthesiology Faculty of Medicine Chulalongkorn University January 21, 2008

Sufentanil Sublingual Tablet System 15mcg vs IV PCA Morphine: A Comparative Analysis of Patient Satisfaction and Drug Utilization by Surgery Type

Single-shot block for elective shoulder surgery Is it enough? Mr C Watkins, Miss L Onwordi Mr J Chakravarthy, Dr A Gupta. NESRA annual meeting 2016

Multimodal perioperative pain management protocols

Pre-op Interventions to Mitigate Post-op Acute and Chronic Pain

RECENT ADVANCES IN ANALGESIA

Local anesthetic infusion pump for pain management following total knee arthroplasty: a meta-analysis

Medicines Q&As. Medicines Q&As. What is the evidence to support the use of IV paracetamol for the treatment of pain? Background.

Advancement in the use of multimodal analgesia for acute postoperative pain

Dose: Metoclopramide -0.1 mg/kg (max 10 mg) IV, over 15 minutes

Pain relief after major surgery

Scottish Medicines Consortium

COBISS.SR-ID EFFECTIVNESS OF DEXAMETASONE VS. MAGNESIUM SULPHATE IN POSTOPERA- TIVE ANALGESIA (DEXAMETASONE VS. MAGNESIUM SULPHATE)

Patient controlled opioid analgesia versus conventional opioid analgesia for postoperative pain (Review)

META-ANALYSIS OF INTRATHECAL MORPHINE FOR LUMBAR SPINE SURGERY

CHAPTER 4 PAIN AND ITS MANAGEMENT

Effect of preoperative intravenous oxycodone administration on sufentanil consumption after retroperitoneal laparoscopic nephrectomy

BJF Acute Pain Team Formulary Group

NHS Grampian Protocol For The Prescribing And Administration Of Oral Opioids Following Trauma Or Surgery

Post-operative Analgesia for Caesarean Section

Deep vein thrombosis and its prevention in critically ill adults Attia J, Ray J G, Cook D J, Douketis J, Ginsberg J S, Geerts W H

SEEING KETAMINE IN A NEW LIGHT

NSAIDs and Tonsillectomy: Efficacy for Pain Relief

NEUROPATHIC CANCER PAIN STANDARDS AND GUIDELINES

Scottish Medicines Consortium

GUIDELINES ON THE MANAGEMENT OF PAIN DUE TO CANCER IN ADULTS

A Randomized Clinical Trial of Nefopam versus Ketorolac Combined With Oxycodone in Patient- Controlled Analgesia after Gynecologic Surgery

Clinical Study Synopsis for Public Disclosure

CONCERNED ABOUT TAKING OPIOIDS AFTER SURGERY?

Continuous Wound Infusion and Postoperative Pain Current status?

Clinical Trial Results with OROS Ò Hydromorphone

Transcription:

British Journal of Anaesthesia 114 (1): 10 31 (2015) Advance Access publication 10 September 2014. doi:10.1093/bja/aeu293 REVIEW ARTICLES EDITOR S CHOICE Impact of pregabalin on acute and persistent postoperative pain: a systematic review and meta-analysis B. M. Mishriky, N. H. Waldron and A. S. Habib* Department of Anesthesiology, Duke University Medical Center, Box 3094, Durham, NC 27710, USA * Corresponding author. E-mail: habib001@dm.duke.edu Editor s key points The authors reviewed the evidence for the use of pregabalin for pain relief in the perioperative period. They found a significant positive effect (in terms of improved pain scores, opioid-sparing and reduction in nausea, vomiting, and pruritus), but a slight increase in some side-effects. Summary. We performed this systematic review to assess the analgesic efficacyof perioperative pregabalin. Subgroup analyses and meta-regression were performed to assess the impact of individual dose and frequency of pregabalin administration on analgesic efficacy. We included 55 studies. When all doses and administration regimens were combined, pregabalin was associated with a significant reduction in pain scores at rest and during movement and opioid at compared with placebo {mean difference [95% confidence interval (CI)]¼20.38 (20.57, 20.20), 20.47 (20.76, 20.18), and 28.27 mg morphine equivalents (210.08, 26.47), respectively}. Patients receiving pregabalin had less postoperative nausea and vomiting and pruritus compared with placebo [relative risk (RR) (95% CI)¼0.62 (0.48, 0.80) and 0.49 (0.34, 0.70), respectively]. Sedation, dizziness, and visual disturbance were more common with pregabalin compared with placebo [RR (95% CI)¼1.46 (1.08, 1.98), 1.33 (1.07, 1.64), and 3.52 (2.05, 6.04), respectively]. All doses of pregabalin tested ( 75, 100 150, and 300 mg) resulted in opioid sparing at after. There were no significant differences in acute pain outcomes with pregabalin 100 300 mg between single preoperative dosing regimens and those including additional doses repeated after. Data were insufficient to reach conclusions regarding persistent pain, but limited data available from two studies suggested that pregabalin might be effective for the reduction of neuropathic pain. In conclusion, this review suggests that pregabalin improves postoperative analgesia compared with placebo at the expense of increased sedation and visual disturbances. Keywords: meta-analysis; postoperative pain; pregabalin Pregabalin is a g-aminobutyric acid analogue that binds to a 2 d subunits of the voltage-gated calcium channels. 1 It reduces the excitability of the dorsal horn neurones after tissue damage. 2 The use of pregabalin for the management of postoperative pain is off-label, and therefore, there are no dosing guidelines for this indication. For other indications, the recommended starting dose is 150 mg day 21 in two to three divided doses, increased within 1 week to 300 mg day 21 with a maximum recommended dose of 600 mg day 21. 2 Studies investigating the perioperative use of pregabalin used doses ranging from 50 to 300 mg and daily doses ranging from 50 to 750 mg. The efficacy of perioperative administration of pregabalin was investigated in previous meta-analyses, 3 5 with all showing better postoperative analgesia with pregabalin. Those metaanalyses grouped studies based on the total daily dose of pregabalin.zhang andcolleagues 5 reportedthat pregabalin dosesof,300and 300mgday 21 reduced 24h opioid but not pain scores after. Engelman and Cateloy 4 grouped the analysis over a wide time-frame (6 h 7 days after ) according to the daily dose of pregabalin (50 150, 225 300, and 600 750 mg) and reported that the lowest effective dose for reducing postoperative analgesic was 225 300 mg with no reduction in pain scores. Since doses were reported in those meta-analyses as total daily dose, it is not clear if the individual dose or frequency of administration of pregabalin affect outcome. For instance, it is not clear from those reviews if individual single doses lower than 225 300 mg have analgesic efficacy or if twice daily dosing of a particular dose of pregabalin would be more effective than single preoperative administration of the same dose. Some studies have investigated the impact of pregabalin on preoperative anxiety, but this was not addressed in those previous meta-analyses. More than 30 studies investigating perioperative pregabalin administration on acute pain outcomes have been published after the publicationofthose reviews,whichincluded 11 5 and18 4 studies. In addition, while one previous meta-analysis 3 assessed the impact of the perioperative administration of pregabalin on chronic pain, it included only three studies. 6 8 Seven other studies 9 15 addressing persistent pain after pregabalin administration have since been published. Therefore, we performed this systematic review to provide an updated meta-analysis of the impact of pregabalin & The Author 2014. Published by Oxford University Press on behalf of the British Journal of Anaesthesia. All rights reserved. For Permissions, please email: journals.permissions@oup.com

Pregabalin and postoperative pain BJA administration on postoperative pain scores and opioid and investigate whether those outcomes differ according to individual pregabalin dose, frequency of administration, type of anaesthesia, or type of. Secondary aims were to assess the impact of pregabalin administration on anxiety scores and persistent pain, and provide an updated meta-analysis of the side-effects of pregabalin administration. Methods We followed the recommendations of the PRISMA statement. 16 We searched MEDLINE (1966 2014), the Cochrane Central Register of Controlled Trials (CENTRAL), EMBASE (1947 2014), and CINAHL for randomized controlled trials (RCTs) that compared pregabalin with control in patients undergoing. Databases were searched using the term pregabalin combined with the MESH terms: Pain, postoperative, Postoperative period, Pain, acute, Pain, chronic, s, and Analgesics, opioid. The search was performed without language restriction. The last literature search was done on March 31, 2014. We also searched the bibliographies of retrieved articles for additional studies. Reviews, abstracts, letters to the editor, and retrospective studies were not included. Articles were included if pregabalin was administered before operation and pain scores, opioid, incidence of persistent pain, and/or time to first analgesia were reported. We excluded studies where pregabalin administration was initiated after operation, the endpoints of interest were not reported or if a placebo group was not included. The articles meeting the inclusion criteria were assessed separately by two authors (B.M.M. and N.H.W.) using the risk of bias table suggested by the Cochrane Collaboration. 17 A data collection sheet was created and two authors (B.M.M. and N.H.W.) extracted data on: (i) Patients: type of, type of anaesthesia, and number of patients. (ii) Interventions: pregabalin dose and frequency of administration. (iii) Comparison: control group regimen. (iv) Outcomes: (a) acute pain outcomes: pain scores at rest and during movement, opioid, and duration of post-anaesthesia care unit (PACU) and hospital stay, (b) preoperative anxiety scores, (c) adverse effects: nausea, vomiting, sedation, dizziness, confusion, headache, visual disturbance, pruritus, difficulty passing urine, dry mouth, fatigue, and request for rescue antiemetics, and (d) persistent pain: pain scores and incidence of persistent pain. Data presented in graphs were requested from the authors. If authors did not respond, data were extracted from the graph. Discrepancies between the two authors were resolved by discussion with the third author (A.S.H.). The primary outcomes of this meta-analysis were pain scores and opioid at 2 and. Secondary outcomes were duration of PACU and hospital stay, incidence of persistent pain at 1, 3, 6, and 12 months, preoperative anxiety scores, and side-effects. Analyses performed for the research questions and synthesis of data Acute pain outcomes In studies involving different doses of pregabalin, we combined all pregabalin doses for the main analysis assessing the impact of pregabalin administration on postoperative pain scores and opioid. Visual analogue scale (VAS) scores for pain reported as 0 100 was converted to the 0 10 scale for analysis (0, no pain; 10, worst possible pain). s were converted to morphine equivalents (ME) for analysis using a conversion factor of 3:1 for oxycodone, 18 0.15:1 for parenteral hydromorphone, 18 10:1 for fentanyl, 18 20:1 for codeine, 18 10:1 for tramadol, 5 and 1:1 for both ketobemidone 19 and piritramide. 20 If ketorolac was the only analgesic used, it was converted to ME using a conversion factor of 3:1. 21 If results were not reported at the time points specified in this analysis, those recorded close to those time points were used instead. To evaluate different pregabalin dosing regimens, we performed subgroup analyses for pain scores and opioid at 2 h after operation according to the individual dose of pregabalin administered before ( 75, 100 150, and 300mg).Forpainscoresandopioid at24h,we performed a subgroup analysis according to the dose and frequency of administration of pregabalin comparing the three dose levels ( 75, 100 150, and 300 mg) and single vs multiple dosing at each dose level. Single dosing refers to studies that administered a single preoperative dose of pregabalin, while multiple dosing refers to studies that used at least one postoperative dose of pregabalin in addition to the preoperative dose or administered more than one preoperative dose. We also performed sensitivity analyses according to the type of and type of anaesthesia (general vs regional) for the primary outcomes of pain scores and opioid at 2 and. To evaluate predictors that could impact our primary outcomes, we also performed a meta-regressionusing pregabalin dose, type of, and type of anaesthesia (general or regional) as predictors for the 2 h outcomes. The frequency of administration of pregabalin (single vs multiple dosing) was used as an additional predictor for outcomes. Preoperative anxiety We pooled preoperative anxiety scores after administration of pregabalin compared with placebo. VAS scores for anxiety reported as 0 100 were converted to the 0 10 scaleforanalysis. Side-effects of perioperative pregabalin administration We pooled adverse effects after administration of pregabalin compared with placebo. If an event rate was reported over multiple time intervals instead of the entire duration of the study, the highest recorded incidence over the duration of the study was used in the analysis. Sedation was defined as scores 3 6 on the Ramsay sedation scale (1, patient is anxious and 11

BJA agitated or restless; 2, patient is co-operative, oriented, and tranquil; 3, patient responds to commands only; 4, patient exhibits brisk response to light glabellar tap or loud auditory stimulus; 5, patient exhibits a sluggish response to light glabellar tap or loud auditory stimulus; 6, patient exhibits no response), 2 5 on the five-point scale (1, completely awake; 2, awake but drowsy; 3, asleep but responsive to verbal commands; 4, asleep but responsive to tactile stimulus; 5, asleep and not responsive to any stimulus), and 2 4 on the four-point scale (1, awake; 2, mild sedation; 3, sleepy but rousable; and 4, very sleepy), while severe sedation was considered for scores 4 6, 4 5, and 3 4 on the Ramsay, five-point scale, and four-point scale, respectively. In studies investigating sedation on the four-point scale (none, mild, moderate, and severe), sedation was defined as any sedation (mild, moderate, or severe). If sedation was not reported, somnolence or drowsiness was used instead for the analysis. Persistent pain We compared the incidence of persistent pain at 1, 3, 6, and 12 months after after pregabalin vs placebo administration. We also compared pain scores at 1 and 3 months after between pregabalin and placebo. Continuous data were summarized as mean difference (MD) with 95% confidence interval (CI). If the 95% CI included a value of 0, we considered that the difference between pregabalin and placebo was not statistically significant. Dichotomous data were summarized as relative risk (RR) with 95% CI. If the 95% CI included a value of 1, we considered the difference not statistically significant. If the pooled results were not statistically significant and the CIs included values that exceeded a 30% difference in the pregabalin group compared with the control group, we considered that no conclusion could be derived from the pooled results due to the wide CIs. Analyses were performed using the Review Manager (RevMan), Version 5.1, Copenhagen: The Nordic Cochrane Centre, The Cochrane Collaboration, 2011, and Comprehensive Meta-Analysis Software (version 3.0). A random effects model (which assumes that the effects being estimated in the different studies are not identical, but follow some distribution) was used. 17 We assessed heterogeneity using the I 2 -test. Heterogeneity was assumed to be present if the I 2 was.50%. Forest plots were used to graphically represent and evaluate treatment effects. Subgroup analysis was performed using the Q-test. We assessed for publication bias for the primary outcomes using the Egger s test. 22 We also performed a sensitivity analysis for the primary outcomes after removing papers with an unclear or high risk of bias. To exclude a small study effect, we compared the results of the random effects and fixed effect models for our primary outcomes. We assessed the proportion of the total variance explained by each of the covariates (R 2 ) included in the meta-regression (pregabalin dose, frequency of administration, type of anaesthesia, and type of ) for the primary outcomes. Meta-regression was performed using the method of moments. Results Six hundred and ninety-five studies were assessed for inclusion in this review (Fig. 1). Fifty-five studies 6 15 23 67 with 4155 patients (2270 received pregabalin and 1885 served as control) were included in the final analysis. Additional data from 16 studies 13 15 24 27 36 37 40 42 46 48 50 57 were provided by the authors. Forty-nine studies investigated acute pain, 7 11 13 15 23 29 31 57 59 61 64 66 68 and 10 chronic pain. 6 15 The characteristics of the included studies are shown in Table 1. The risk of bias of the included studies is shown in Table 2. Primary outcomes Mishriky et al. Pain scores Pain scores at 2 h Pain scores at rest at 2 h (Fig. 2) were investigated in 33 9 11 13 15 24 27 28 34 38 40 44 46 49 51 57 61 63 66 68 studies and during movement in 14 9 11 13 14 29 36 37 40 42 44 51 57 studies. Pooled results showed a statistically significant reduction in pain scores at rest [MD (95% CI)¼20.81 (21.07, 20.51, I 2 ¼88%)] and during movement [MD (95% CI)¼ 20.58 (20.94, 20.21, I 2 ¼82%)] in pregabalin-treated patients. There was no evidence of publication bias for pain scores at rest or movement (P¼0.07 and 0.71, respectively). For pain scores at rest, 9% of the total variance was explained by the dose of pregabalin used, 11% by the type of, and 1% by the type of anaesthesia. For pain scores on movement, 16% of the total variance was explained by the dose of pregabalin, 10% by type of, and none was explained by type of anaesthesia. In subgroup analysis, pain scores at rest were reduced with all doses of pregabalin (Fig. 2). Pain scores with movement were only reduced with the 300 mg dose. There were no significant differences between the three dose groups for pain at rest (P¼0.95) or with movement (P¼0.29). Sensitivity analysis according to type of showed a reduction in pain scores at rest for all types of except minor and cardiac (Table 3). Pain scores on movement were only reduced in open abdominal and head and neck surgeries. Pain scores at rest and on movement were reduced in studies using general anaesthesia but not regional anaesthesia. The meta-regression found type of (P¼0.02), but not pregabalin dose (P¼0.74) or type of anaesthesia (0.16) to be a significant predictor of 2 h pain scores at rest. The type of anaesthesia was a significant predictor of pain scores on movement at 2 h (P¼0.046), but not type of (P¼0.10) or pregabalin dose (0.81). Pain scores at Pain scores at rest at (Fig. 3) were 79 11 13 15 23 26 28 31 32 34 38 40 57 investigated in 40 studies 59 62 64 66 79 11 13 14 23 29 and during movement in 18 studies. 31 36 37 40 42 46 51 57 59 Pooled results showed a statistically significant reduction in pain scores at rest [MD (95% CI)¼20.38 (20.57, 20.20, I 2 ¼77%)] and during movement [MD (95% CI)¼20.47 (20.76, 20.18, I 2 ¼70%)] with the perioperative administration of pregabalin. There was no evidence of publication bias (P¼0.94 and 0.65 for pain scores 12

Pregabalin and postoperative pain BJA Literature search Databases: MEDLINE, EMBASE, CENTRAL, and CINAHL Last date of search: 03/31/2014 695 studies were identified 614 Excluded 254 Reviews 181 Duplicates 52 Conference abstract 48 Case-reports 32 Editorial, letter, or correspondence 27 Not studying pregabalin 7 Surveys 8 Animal studies 4 Practice guidelines 1 News letter 82 studies assessed for inclusion 24 Excluded 6 Full text could not be obtained 6 No control group 4 Endpoints other than specified by our analysis 2 Pregabalin administered postoperatively 2 Study completed but not published 1 Cross over study 1 Retracted 1 Retrospective study 1 Non-surgical 58 studies met the inclusion criteria 3 Excluded 2 Inconsistent data 1 Not reporting standard deviations 55 studies included in the final analysis Fig 1 PRISMA flow chart detailing retrieved, excluded, assessed, and included trials. at rest and on movement, respectively). For pain scores at rest, 1% of the total variance was explained by the dose of pregabalin used, 32% by the type of, 0.28% by the frequency of pregabalin administration, and none was explained by the type of anaesthesia. For pain scores on movement, type of explained 26% of the total variance and type of anaesthesia 0.22%. The dose of pregabalin and frequency of administration did not explain any of the total variance. In subgroup analysis, pain scores at rest were reduced with pregabalin doses 100 mg, but not 75 mg (Fig. 3), with no significant differences between dose levels (P¼0.87). When accounting for dosing frequency (Table 4), pain scores at rest were not reduced by any of the doses of pregabalin compared with placebo when given in single doses, but were reduced with multiple dosing of pregabalin 75 mg (P¼0.03) and 100 150 mg (P¼0.0001). There were no significant differences between single and multiple dosing at the 75 mg (P¼0.17), 100 150 mg (P¼0.86), or 300 mg (P¼0.27) dose levels. Pain scores at rest were only reduced in open abdominal and head and neck surgeries (Table 3). Sensitivity analysis according to type of anaesthesia showed a reduction in pain scores at rest with general but not regional anaesthesia. The meta-regression found type of (P¼0.008), but not type of anaesthesia (P¼0.21), pregabalin dose (P¼0.67), or pregabalin frequency (P¼0.61) to be a significant predictor of pain scores at rest. Pain scores on movement were reduced only with doses of 300 mg compared with placebo, but there were no significant differences between the three dose levels of pregabalin (P¼0.42). When accounting for dosing frequency (Table 3), 13

14 Table 1 Characteristics of included studies. n, number of patients in the group; PGB, pregabalin;, general anaesthesia; POD, postoperative day; Dex, dexamethasone; PCA, patient-controlled analgesia; PCEA, patient-controlled epidural analgesia; VAS, verbal analogue scale; NSAID, non-steroidal anti-inflammatory drug; PACU, post-anaesthesia care unit; ICU, intensive care unit; CSE, combined spinal epidural. *Group not included in the analysis Participant characteristics Interventions Comparison Outcomes Reference Surgery type Anaesthesia Postoperative analgesia Experimental groups (n) Acin and Mesh hernia repair Non-steroidal colleagues 12 anti-inflammatory drugs Agarwal and colleagues 23 Alimian and colleagues 52 Balaban and colleagues 24 Bekawi and colleagues 53 Bornemann-Cimenti and colleagues 25 Burke and Shorten 7 Buvanendran and colleagues 6 Laparoscopic cholecystectomy Dacrocystorhinostomy Laparoscopic cholecystectomy Laparoscopic cholecystectomy Transperitoneal nephrectomy Lumbar discectomy Total knee arthroplasty PGB administration time PGB 75 mg (70) 75 mg at night for 3 days before operation and 75 mg at night till POD 12 PCA fentanyl PGB 150 mg (27) before PRN opioid PGB 300 (40) before I.V. fentanyl 25 mg as needed PGB 150 mg (30); PGB 300 mg (30) I.M. meperidine and diclofenac PRN PGB 150 mg (30) before 2 h before and then every 12 h for 2 days PCA piritramide PGB 300 mg (13) before PACU: i.v. morphine; ward: regular oral codeine phosphate, paracetamol, and diclofenac+i.m. opioids (dihydrocodeine, tramadol, and morphine) for breakthrough pain PCEA followed by oral opioids as needed to keep pain score,4 PGB 300 mg (18) PGB 300 mg (120) 300 mg 90 min before operation and 150 mg at 12 and after operation 300 mg 1 2 h before, 150 mg twice daily for 10 days, 75 mg twice daily on days 11 and 12, and 50 mg twice daily on days 13 and 14 Control group (n) Primary outcome Study follow-up Placebo (70) Persistent pain 12 months Placebo (29) Pain scores and opioid Placebo (40) Pain scores Placebo (30) Placebo (30); gabapentin 1200 mg* (30) Placebo (13) Pain scores and opioid Pain scores and opioid Placebo (20) Pain score up to 3 months after operation Placebo (120) Neuropathic pain at 6 months 48 h 3 months 6 months BJA Mishriky et al.

15 Buvanendran and colleagues 26 Cabrera Schulmeyer and colleagues 27 Chang and colleagues 28 Clendenen and colleagues 32 Chaparro and colleagues 29 Choi and colleagues 31 Total knee replacement Laparoscopic sleeve gastrectomy Laparoscopic cholecystectomy Arthroscopic rotator cuff repair Liposuction+ augmentation mammoplasty/ abdominoplasty Lumbar spinal Spinal Patient-controlled intrathecal analgesia Infusion of ketoprofen 300 mg 21 +i.v. morphine as rescue therapy I.V. ketorolac 30 mg as needed Oral celecoxib, oxycodone, and paracetamol 325 mg every 4 6 h as needed PACU: i.v. morphine; then: oral paracetamol and codeine or tramadol or hydrocodone+ibuprofen or diclofenac for rescue Continuous infusion of i.v. fentanyl until 48 h+i.v. ketorolac 30 mg for VAS pain 5 PGB 150 mg multi-dose (16); PGB 150 mg single dose (16) PGB 150 mg (39) PGB 300 mg (39) PGB 150 mg (23) PGB 75 mg (50) PGB 150 mg+placebo (36); PGB 150 mg+dex* (36) Demirhan and Rhinoplasty Tramadol PCA PGB 300 mg+placebo colleagues 55 (20); PGB 300 mg+dex* (20) El Rahmawy and colleagues 65 Eskandar and Ebeid 56 Elective general surgeries below the umbilicus (inguinal hernia, varicocele, varicose veins) Shoulder arthroscopy 1 h before (single and multiple dosing groups)+150 mg at 12 and in multi-dose group Single dosing: 2 h before 1 h before induction of anaesthesia and 12 h after the initial dose 150 mg before sedation then every 12 h for a total of 4 doses 75 mg the night before and 1 h before, then every 12 h through POD 4 1 h before and every 12 h after initial dose until POD 3 before Spinal I.M. diclofenac every 12 h PGB 150 (43) Single dosing: 2 h before PRN nalbuphine PGB 300 mg (40) 12 and 1 h before Placebo (16) Placebo (40) Placebo (38) Placebo (24) Placebo (49) Placebo (36) Placebo (20) Placebo (43) Placebo (40) Level of spinal neurotransmitters Shoulder pain and abdominal pain Pain score during movement Pain scores and need for rescue analgesia and pain scores Incidence of post-dural puncture headache Postoperative pain scores 32 h 48 h 48 h 96 h 6 months Continued Pregabalin and postoperative pain BJA

16 Table 1 Continued Participant characteristics Interventions Comparison Outcomes Reference Surgery type Anaesthesia Postoperative analgesia Experimental groups (n) Fassoulaki and colleagues 13 George and colleagues 57 Ghai and colleagues 33 Ghoneim Hegazy 58 Gianesello and colleagues 14 Gonano and colleagues 34 Ittichaikulthol and colleagues 35 Jain and colleagues 59 Jo and colleagues 11 Jokela and colleagues 36 Abdominal hysterectomy or myomectomy Abdominal hysterectomy Abdominal hysterectomy Cystectomy with urinary diversion Major spinal Minor orthopaedic Abdominal hysterectomy Unilateral total knee arthroplasty Abdominal hysterectomy Day-case gynaecological laparoscopic Epidural First 2 days: PCA morphine, then oral codeine with paracetamol as needed PCA morphine, scheduled oral NSAID I.M. diclofenac sodium; i.v. tramadol if pain not controlled Morphine PCA and i.v. paracetamol Continuous infusion of morphine+ketorolac until 48 h after, VAS 3: i.v. morphine PACU: i.v. piritramide; after PACU: oral mefenamic acid PACU: i.v. morphine; ward: PCA morphine Bupivacaine/morphine PCEA, oral diclofenac rescue PACU: i.v. fentanyl; after PACU: PCA fentanyl I.V. fentanyl (in PACU) or oral paracetamol and codeine (after PACU) as needed PGB 150 mg (39) PGB 75 mg (31); PGB 150 mg (28) PGB 300 mg (30) PGB 75 mg (30) PGB 300 mg (30) PGB 300 mg (20) PGB 300 mg (38) PGB 75 (20) Remifentanil PGB 150 mg (20) PGB 75 mg (30); PGB 150 mg (26) PGB administration time 8hourly administration starting on the day before and continued to POD 5 2 h before and 12 h after initial dose Single dosing: 1 2 h before 75 mg every 12 h for 10 days before 300 mg 1 h before then 150 mg twice daily for 48 h Single dosing: at least 1 h before before 75 mg twice daily starting preop and continued through POD2 before before Control group (n) Primary outcome Study follow-up Placebo (41) Placebo (30) Placebo (30); gabapentin 900 mg* (30) Placebo (30) Placebo (30) Placebo (20) VAS anxiety immediately before anaesthesia induction 3 months 48 h 48 h Placebo (40) Pain scores Placebo (20) PCEA morphine use 48 h Remifentanil (20); placebo* (20) Placebo (28) PACU opioid Pain scores and opioid 48 h BJA Mishriky et al.

17 Jokela and colleagues 37 Joshi and Jagadeesh 9 Khurana and colleagues 60 Kim and colleagues 15 Laparoscopic hysterectomy Off-pump coronary artery bypass grafting Lumbar discectomy Robot-assisted endoscopic thyroidectomy I.V. oxycodone then PCA oxycodone until next morning, then oral ibuprofen and paracetamol/codeine for breakthrough pain I.V. paracetamol every 6 h, breakthrough pain treated with tramadol first, then i.v. diclofenac I.V. tramadol for breakthrough pain PACU: i.v. fentanyl; ward: i.m. tramadol Kim and colleagues 10 Mastectomy PACU: i.v. fentanyl; ward: i.m. tramadol Kim and colleagues 38 Lumbar spinal fusion PGB 150 mg (27); PGB 300 mg (29) PGB 150 mg (20) PGB 75 mg (30) PGB 150 mg (47) PGB 75 mg (42) PCA fentanyl+ketorolac PGB 75 mg (28); PGB 150 mg (28) Kohli and Hysterectomy Spinal Not reported PGB 150 mg (50); PGB colleagues 39 300 mg (50) Koyuncu and colleagues 54 Kumar and colleagues 61 Lee and colleagues 62 Martinez and colleagues 63 Modified radical mastectomy Lumbar discectomy Laparoscopic urologic Total hip arthroplasty 1 h before and 12 h after the initial dose 150 mg before operation, then 75 mg every 12 h for 2 postoperative days 75 mg before operation, then 75 mg every 8 h for 7 postoperative days 1 h before and 12 h after the initial dose 1 h before and 12 h after the initial dose 1 h before and 12 h after before PCA morphine PGB 150 (30) 150 mg preop and 75 mg 12 h postop I.V. fentanyl, i.v. diclofenac, or both for breakthrough pain PCA with solution containing morphine, ketorolac, and 5-HT 3 antagonist PGB 150 (25); tramadol 100 (25)* PGB 300+high dose (0.3 mgkg 21 min 21 ) remifentanil (31) Morphine PCA PGB 150 mg (35); PGB 150 mg+intraoperative ketamine (35)*; Intraoperative ketamine (34)* before operation before operation before operation Placebo (29) Pain scores and opioid 5days Placebo (20) Pain scores 3 months Placebo (30); gabapentin 300 mg* (30) Pain scores Placebo (47) Pain scores 48 h Placebo (42) Placebo (28) Pain scores on movement Analgesic, pain scores, and need for rescue analgesics 3 months 48 h 48 h Placebo (50) VAS anxiety scores Placebo (30) Pain scores and opioid 12 h Placebo (25) Pain scores 6 h High-dose remifentanil (0.3 mgkg 21 min 21 ) (29); low-dose remifentanil (0.05 mgkg 21 min 21 )* (30) Pain scores, PCA use, and hyperalgesia Placebo (38) Pain scores 48 h Continued Pregabalin and postoperative pain BJA

18 Table 1 Continued Participant characteristics Interventions Comparison Outcomes Reference Surgery type Anaesthesia Postoperative analgesia Experimental groups (n) Mathiesen and Tonsillectomy Paracetamol+i.v. morphine colleagues 41 and oral ketobemidone upon patient request Mathiesen and colleagues 42 Abdominal hysterectomy PACU: i.v. morphine; after PACU: regular paracetamol+pca morphine Mathiesen and Hip arthroplasty Spinal Regular oral colleagues 40 paracetamol+pca morphine Nutthachote and colleagues 64 Ozgencil and colleagues 43 Paech and colleagues 68 Peng and colleagues 44 Laparoscopic gynaecologic Lumbar laminectomy and discectomy Minor gynaecological Laparoscopic cholecystectomy Scheduled oral NSAID, oral paracetamol, and i.v. meperidine for breakthrough pain Paracetamol+PGB 300 mg+placebo (45); paracetamol+pgb 300 mg+dex* (43) Paracetamol+PGB 300 mg+placebo (43); Paracetamol+PGB 300 mg+dex* (42) PGB 300 mg+placebo (40); PGB 300 mg+dex* (38) PGB 75 mg (27) PGB administration time before before before 2 h before and every 12 h for total of three doses PCA morphine PGB 150 mg (30) 2 h before and every 12 h from initial dose for 2 days PACU: i.v. fentanyl then i.v. tramadol followed by oral diclofenac if target pain score not achieved. After discharge: paracetamol as needed PACU: i.v. fentanyl as needed; after PACU: paracetamol/ codeine orally upon request Pesonen and Cardiac ICU: i.v. oxycodone; ward: colleagues 8 oral or i.m. oxycodone; after discharge: oral paracetamol PGB 100 mg (45) PGB 50 mg (48); PGB 75 mg (48) PGB 150 mg (32) Sagit and Septoplasty I.M. diclofenac PGB 75 mg (50); PGB colleagues 66 150 mg (46) before 1 h before and every 12 h after initial dose for a total of 3 doses 150 mg 1 h before then 75 mg every 12 h from POD 1 to POD 5 before Control group (n) Primary outcome Study follow-up Paracetamol+placebo +placebo (43) Paracetamol+placebo +placebo (43) Placebo+placebo (42) Placebo (27) Placebo (30); gabapentin 600 mg* (30) Pain during swallowing at 2 h Pain scores (shoulder) 48 h Placebo (45) Pain scores Placebo (46) Pain scores 7 days Placebo (32) Placebo (47) Pain scores 3 months BJA Mishriky et al.

19 Sahu and colleagues 45 Sarakatsianou and colleagues 67 Spreng and colleagues 46 Sundar and colleagues 47 Infra-umbilical surgeries Laparoscopic cholecystectomy Lumbar discectomy Off-pump coronary artery bypass grafting Wang and Bunionectomy Regional PCA hydromorphone then colleagues 48 oral hydrocodone/ paracetamol White and colleagues 49 Yadeau and colleagues 50 Yucel and colleagues 51 Elective ambulatory and short stay surgical procedures Foot or ankle Abdominal hysterectomy Spinal Not reported PGB 150 mg (35) 12 and 1 h before PCA morphine PGB 300 (20) the night before and immediately before operation PCA morphine PGB 150 mg (22) before I.V. fentanyl PGB 150 mg (30) before PGB 300 mg (32) I.V. fentanyl PGB 75 mg (27); PGB 150 mg (27); PGB 300 mg (27) Popliteal nerve block+cse PCA hydromorphone then oral oxycodone or hydrocodone or hydromorphone and paracetamol PGB 100 mg (30) PCA morphine PGB 150 mg (30); PGB 300 mg (30) 300 mg 1 h before then 150 mg every 8 h up to 40 h Single dosing: 60 90 min before 100 mg 1 h before and 50 mg every 12 h for 3days 4 h before and at 12 h after operation Placebo (35) Pain scores Placebo (20) Pain scores Placebo (24) Pain scores Placebo (30) Placebo (28); naproxen sodium* (29) Haemodynamic response to intubation and opioid and time to first PCA use 48 h Placebo (27) VAS anxiety 7 days Placebo (30) Placebo (30) Number of hours of moderate to severe pain 48 h Pregabalin and postoperative pain BJA

BJA Mishriky et al. Table 2 Risk of bias table. Low, low risk of bias; high, high risk of bias; unclear, unclear risk of bias Reference Randomization sequence generation Allocation concealment Blinding of participants and personnel Blinding of outcome assessment Incomplete outcome data Selective reporting Acin and colleagues 12 Unclear High High High Low Low Agarwal and colleagues 23 Low Low Low Low Low Low Alimian and colleagues 52 Unclear Low Low Low Low Low Balaban and colleagues 24 Low Low Low Low Low Low Bekawi and colleagues 53 Low Low Low Low Unclear Low Bornemann-Cimenti and Low Low Low Low Low Low colleagues 25 Burke and Shorten 7 Unclear Low Unclear Unclear Low Low Buvanendran and Low Low Low Low Low Low colleagues 6 Buvanendran and Low Low Low Low Low Low colleagues 26 Cabrera Schulmeyer and Low Unclear Low Low Low Low colleagues 27 Chang and colleagues 28 Unclear Low Low Low Low Low Clendenen and colleagues 32 Low Low Low Unclear Low Low Chaparro and colleagues 29 Low Low Low Low Low Low Choi and colleagues 31 Low Low Low Low Low Low Demirhan and colleagues 55 Low Low Low Low Low Low El Rahmawy and colleagues 65 Low Low Low Low Unclear Low Eskandar and Ebeid 56 Low Low Low Unclear Low Low Fassoulaki and colleagues 13 Low Low Low Unclear Low Low George and colleagues 57 Low Low Low Low Low Low Ghai and colleagues 33 Low Low Low Low Low Low Ghoneim and Hegazy 58 Low Low Low Low Low Low Gianesello and colleagues 14 Low Low Low Low Low Low Gonano and colleagues 34 Low Unclear Low Low Low Low Ittichaikulthol and Unclear Unclear Low Low Low Low colleagues 35 Jain and colleagues 59 Low Low Low Low Low Low Jo and colleagues 11 Low Unclear Low Low Low Low Jokela and colleagues 36 Low Low Low Low Low Low Jokela and colleagues 37 Low Low Low Low Low Low Joshi and Jagadeesh 9 Low Low Low Low Low Low Khurana and colleagues 60 Low Low Low Low Unclear Unclear Kim and colleagues 15 Unclear Low Low Low Low Low Kim and colleagues 10 Low Low Low Low Low Low Kim and colleagues 38 Low Low Low Low Low Low Kohli and colleagues 39 Low Low Unclear Unclear Low Low Koyuncu and colleagues 54 Low Low Unclear Unclear Unclear Unclear Kumar and colleagues 61 Low Low Low Low Low Low Lee and colleagues 62 Unclear Unclear Unclear Unclear Low Low Martinez and colleagues 63 Low Low Low Low Low Low Mathiesen and colleagues 41 Low Low Low Low Low Low Mathiesen and colleagues 42 Low Low Low Low Low Low Mathiesen and colleagues 40 Low Low Low Low Low Low Nutthachote and Low Low Low Low Low Low colleagues 64 Ozgencil and colleagues 43 Low Unclear Low Unclear Low Low Paech and colleagues 68 Low Low Low Low Low Low Peng and colleagues 44 Low Low Low Low Low Low Continued 20

Pregabalin and postoperative pain BJA Table 2 Continued Reference Randomization sequence generation Allocation concealment Blinding of participants and personnel Blinding of outcome assessment Incomplete outcome data Pesonen and colleagues 8 Low Low Low Low Low Low Sagit and colleagues 66 Low Unclear Low Low Low Low Sahu and colleagues 45 Low Unclear Unclear Low Low Low Sarakatsianou and Low Low Low Low Low Low colleagues 67 Spreng and colleagues 46 Low Low Low Low Low Low Sundar and colleagues 47 Low Unclear Low Low Low Low Wang and colleagues 48 Low Low Low Low Low Low White and colleagues 49 Low Low Low Low Low Low Yadeau and colleagues 50 Low Low Low Low Low Low Yucel and colleagues 51 Low Low Low Low Low Low Selective reporting pain on movement was not reduced byanyof the doses of pregabalin compared with placebo when given in single doses, but were reduced by multiple doses at the 75 mg (P¼0.02) and 300 mg dose levels (P¼0.04). Multiple dosing was significantly more effective than single dosing at the 75 mg level (P¼0.02), but not at the 100 150 mg (P¼0.39) or the 300 mg (P¼0.89) doses. Pain scores on movement were reduced in open abdominal, orthopaedic, and cardiac (Table 3). Sensitivity analysis according to type of anaesthesia showed a reduction in pain scores with movement with general but not regional anaesthesia. None of the covariates was a significant predictor in the meta-regression model [type of (P¼0.08), pregabalin dose (P¼0.14), pregabalin frequency (P¼0.27), and type of anaesthesia (P¼0.77)]. at 2 h at 2 h 711131424282934 36 38 (Fig. 4) was investigated in 23 studies. 40 44 46 49 51 55 57 68 Pooled results showed a statistically significant reduction in opioid [MD (95% CI)¼22.09 mg ME (22.87, 21.30, I 2 ¼94%)] with the administration of pregabalin. There was no evidence of publication bias (P¼0.83). Twenty per cent of the total variance was explained by the dose of pregabalin. Type of anaesthesia or did not explain anyof the total variance. In subgroup analysis, opioid was reduced by the 100 150 and 300 mg doses compared with placebo, but not the 75 mg dose (Fig. 4). This reduction was significantly different among the three groups (P¼0.005) with pairwise comparisons showing significantly lower opioid sparing in the 75 mg group compared with the 100 150 mg (P¼0.001) and the 300 mg (P¼0.0007) groups, and no difference between the 100 150 and 300 mg groups. Sensitivity analysis according to type of showed a reduction in opioid at 2 h in orthopaedic, open abdominal, and minor surgical procedures. Two hours opioid sparing was seen with general anaesthesia but not regional anaesthesia (Table 3). In the meta-regression model, pregabalin dose was a significant predictor of 2 h opioid (P¼0.02), but not type of (P¼0.34) or type of anaesthesia (0.34). at at 8111314232527 29 33 (Fig. 5) was investigated in 29 studies. 35 38 40 44 46 48 50 51 54 58 Pooled results showed a statistically significant reduction in opioid with the administration of pregabalin [MD (95% CI)¼28.27 mg ME (210.08, 26.47, I 2 ¼95%)]. There was no evidence of publication bias (P¼0.21). Fourteen per cent of the total variance was explained by the dose of pregabalin, 3% by the type of anaesthesia, whereas type of and frequency of pregabalin administration did not explain any of the variance. In subgroup analysis, all doses of pregabalin reduced opioid compared with placebo when given as a single preoperative dose or as multiple doses (Table 3). There were no statistically significant differences between administration of a single preoperative dose and administration of multiple doses for the 75 mg (P¼0.87), 100 150 mg (P¼0.44), and the 300 mg (P¼0.66) dose levels. There were also no significant differences between the three dose levels of pregabalin when all studies were combined (P¼0.25, Fig. 5). at was reduced in all types of except minor surgical procedures and head and neck. Pooled results of both general anaesthesia and regional anaesthesia studies showed a reduction in opioid (Table 3). The type of anaesthesia was a significant predictor of opioid (P¼0.0496) in the meta-regression model, but not type of (P¼0.11), pregabalin dose (P¼0.92), or frequency of pregabalin administration (P¼0.26). Sensitivity analysis according to study risk of bias assessment Our sensitivity analysis showed no difference in primary outcomes when papers with unclear risk of bias 711 13 15 27 28 32 34 35 39 43 45 47 52 54 56 60 62 65 66 in any of the risk of bias assessments were removed from the analysis. 21

BJA Mishriky et al. Pregabalin Control Study or Subgroup Mean SD Total Mean SD Total Weight 1.1.1 Pregabalin 75 mg George 4.1 2 31 5.4 2.2 30 2.2% Jokela BJA 2008 2.95 1.2 30 2.81 1.37 28 2.7% Kim 2011 5 2 42 6 2 42 2.4% Kim JC 2011 2 2.22 28 3 1.48 28 2.3% Peng 1.5 2.32 96 2 2.96 46 2.3% Sagit 4.46 2.19 50 5.72 2.19 47 2.4% White 4 3 27 4 3 27 1.6% Subtotal (95% Cl) 304 248 15.8% Heterogeneity: Tau 2 = 0.17; Chi 2 = 10.61, df = 6 (P = 0.10); I 2 = 43% Test for overall effect: Z = 2.91 (P = 0.004) Mean Difference IV, Random, 95% Cl 1.30 [ 2.36, 0.24] 0.14 [ 0.52, 0.80] 1.00 [ 1.86, 0.14] 1.00 [ 1.99, 0.01] 0.50 [ 1.47, 0.47] 1.26 [ 2.13, 0.39] 0.00 [ 1.60, 1.60] 0.71 [ 1.18, 0.23] Mean Difference IV, Random, 95% Cl 1.1.2 Pregabalin 100 150 mg Balaban 0.7 0.8 30 1 0.8 30 3.0% Bekawi 2.4 1.28 30 3.73 1.7 30 2.6% Cabrera Schulmeyer 3.31 3.11 39 5.93 2.98 41 1.8% Fassoulaki 2.01 6.4 2.16 40 2.3% George 2.1 28 5.4 2.2 30 2.1% Jo 2.7 0.8 20 3.65 1.46 20 2.6% Jokela BJA 2008 2.58 1.61 26 2.81 1.37 28 2.5% Jokela Pain 2008 3.18 2.01 27 2.53 1.71 2.3% Joshi 2.2 0.89 20 2.4 0.82 20 2.8% Kim 2010 2.5 Kim JC 2011 3 1.85 28 3 1.48 28 2.4% Koyuncu 2 0.74 30 2.5 0.74 30 3.0% Kumar 3.64 1.28 25 6.04 0.93 25 2.7% Ozgencil 3.5 1.13 30 5.36 1.03 30 2.8% Paech 1.6 2.67 2.2% Sagit 4 2.3 46 5.72 2.19 2.4% Spreng 1.17 0.96 22 2.25 1.44 24 2.6% White 4 2 27 4 3 27 1.8% Yucel 6.02 0.66 30 6.23 0.56 30 3.0% Subtotal (95% Cl) 587 601 47.4% Heterogeneity: Tau 2 = 0.52; Chi 2 = 105.20, df = 18 (P < 0.00001); I 2 =83% Test for overall effect: Z = 4.16 (P < 0.0001) 1.1.3 Pregabalin 300 mg 4.7 4.3 5.5 Alimian 2.7 1.4 40 5.4 1.6 40 Balaban 0.3 0.7 30 1 0.8 30 Chang 5.2 1.9 39 4.9 1.8 38 Demirhan 1 2.96 Eskander 4.65 1.53 Gianesello 1.8 0.4 30 3.7 0.5 30 Gonano 2.7 1.1 20 2.6 1.2 20 lttichaikuthol 3.8 1.9 38 5.4 2.9 40 Jokela Pain 2008 2.38 1.8 29 2.53 1.71 29 Lee 5.48 0.89 Mathiesen 2008 0.45 0.93 40 0.56 1.01 38 Mathiesen 2009 3.83 2.2 39 4.03 2.04 40 Mathiesen 2011 3.73 2.52 45 4.5 2.38 43 White 4 4 27 4 3 27 Yucel 4.37 0.49 30 6.23 0.56 30 Subtotal (95% Cl) 498 494 Heterogeneity: Tau 2 = 0.74; Chi 2 = 163.74, df = 14 (P < 0.00001); I 2 =91% Test for overall effect: Z = 3.25 (P = 0.001) Total (95% Cl) 1389 1343 100.0% Heterogeneity: Tau 2 = 0.65; Chi 2 = 321.89, df = 40 (P < 0.00001); I 2 =88% Test for overall effect: Z = 5.51 (P < 0.00001) Test for subgroup differences: Chi 2 = 0.11, df = 2 (P = 0.95); I 2 =0% 37 20 40 31 1 5.8 5.76 4.44 1.32 0.91 29 47 5.7 2.1 47 2.3% 45 1 2.19 45 47 20 40 29 2.7% 3.0% 2.5% 1.0% 2.7% 3.1% 2.6% 2.1% 2.4% 2.9% 2.9% 2.3% 2.2% 1.3% 3.1% 36.8% 0.30 [ 0.70, 0.10] 1.33 [ 2.09, 0.57] 2.62 [ 3.96, 1.28] 1.70 [ 2.63, 0.77] 1.10 [ 2.21, 0.01] 0.95 [ 1.68, 0.22] 0.23 [ 1.03, 0.57] 0.65 [ 0.33, 1.63] 0.20 [ 0.73, 0.33] 0.20 [ 1.13, 0.73] 0.00 [ 0.88, 0.88] 0.50 [ 0.87, 0.13] 2.40 [ 3.02, 1.78] 1.86 [ 2.41, 1.31] 0.60 [ 0.41, 1.61] 1.72 [ 2.63, 0.81] 1.08 [ 1.78, 0.38] 0.00 [ 1.36, 1.36] 0.21 [ 0.52, 0.10] 0.79 [ 1.16, 0.42] 2.70 [ 3.36, 2.04] 0.70 [ 1.08, 0.32] 0.30 [ 0.53, 1.13] 0.00 [ 2.34, 2.34] 1.15 [ 1.78, 0.52] 1.90 [ 2.13, 1.67] 0.10 [ 0.61, 0.81] 1.60 [ 2.68, 0.52] 0.15 [ 1.05, 0.75] 0.28 [ 0.74, 0.18] 0.11 [ 0.54, 0.32] 0.20 [ 1.14, 0.74] 0.77 [ 1.79, 0.25] 0.00 [ 1.89, 1.89] 1.86 [ 2.13, 1.59] 0.81 [ 1.30, 0.32] 0.79 [ 1.07, 0.51] 4 2 0 2 4 Favours pregabalin Favours control Fig 2 Forest plot for pain scores at rest at 2 h. SD, standard deviation; CI, confidence interval; IV, inverse variance. Comparing random effects and fixed effect models for primary outcomes The fixed effect model yielded very comparable results with the random effects model for the primary outcomes of our review. Secondary outcomes Duration of PACU and hospital stay 28 34 46 49 The duration of PACU stay was reported in four studies and duration of hospital stay or time to achieve hospital discharge criteria in five studies. 68143249 There was no difference 22

23 Table 3 Sensitivity analysis according to type of and type of anaesthesia. Data are MD (95% CI) (number of studies included in the analysis). ME, morphine equivalents; NA, not applicable; NE, not estimable Type of or anaesthesia Orthopaedic Open abdominal Laparoscopic abdominal Minor surgical procedures Head and neck Pain scores at rest at 2 h 21.13 (21.77, 20.48, I 2 ¼92%) (8) 56 61 14 34 38 40 43 46 Pain scores during movement at 2 h at 2 h (mg ME) Pain scores at rest at 21.07 (22.73, 0.58, 21.99 (23.48, 20.49, 20.25 (20.52, 0.03, I 2 ¼95%) (2) 14 40 I 2 ¼94%) (8) 714343840434656 I 2 ¼53%) (14) 7142631323438 40 43 46 48 50 56 59 Pain scores during movement at 20.65 (21.27, 20.02, at (mg ME) 14 38 40 43 46 48 50 56 211.70 (215.41, 27.99, I 2 ¼69%) (6) 71440465931 I 2 ¼84%) (8) 21.05 (21.34, 20.76, 20.58 (20.85, 20.32, 23.13 (24.54, 21.72, 20.67 (21.12, 20.21, 20.31 (20.60, 20.02, 212.70 (217.41, 27.99, I 2 ¼16%) (6) 11 13 35 42 51 57 I 2 ¼0%) (5) 11 13 42 51 57 I 2 ¼80%) (6) 11 13 35 42 51 57 I 2 ¼73%) (7) 11 13 35 42 51 45 57 I 2 ¼0%) (5) 11 13 42 51 57 I 2 ¼94%) (9) 20.47 (20.90, 20.03, I 2 ¼70%) (8) 53 62 24 27 28 36 37 44 20.00 (20.49, 0.49, 22.17 I 2 36 37 44 ¼0%) (3) (24.98, 0.64, I 2 ¼97%) 20.39 (20.84, 0.07, (4) 24 28 36 44 I 2 ¼83%) (9) 62 64 23 27 28 37 44 36 53 57 58 11 13 33 35 42 51 25 20.05 (20.83, 0.73, 26.59 (211.65, 21.52, I 2 ¼73%) (3) 23 36 37 I 2 ¼90%) (6) 23 27 28 36 37 44 0.38 (20.42, 1.18, I 2 ¼0%) NA 20.99 (21.78, 20.19, I 2 ¼0%) 1.33 (0.42, 2.24, I 2 ¼NE) NA 21.78 (26.98, 3.42, I 2 ¼83%) (2) 49 68 (2) 49 68 (1) 49 29 36 (2) 21.18 (22.23, 20.13, 21.71 (22.79, 20.63, I 2 ¼83%) (5) 15 41 52 55 66 I 2 ¼NE) (1) 41 20.74 (22.11, 0.63, I 2 ¼83%) 20.94 (21.25, 20.64, 20.80 (21.80, 0.20, 22.29 (23.52, 21.06, (2) 41 55 I 2 ¼18%) (5) 15 41 52 66 55 I 2 ¼NE) (1) 41 I 2 41 55 ¼0%) (2) Cardiac 20.20 (20.73, 0.33, 20.20 (20.52, 0.12, NA 20.36 (21.22, 0.49, I 2 ¼91) 21.30 (21.88, 20.72, 23.87 (25.27, 22.47, I 2 ¼NE) (1) 9 I 2 ¼NE) (1) 9 (2) 947 I 2 ¼NE) (1) 9 I 2 ¼0%) (2) 847 Breast and plastic General anaesthesia Regional anaesthesia 20.60 (20.98, 20.21, 20.76 (23.21, 1.69, 0.00 I 2 ¼9%) (2) 10 54 I 2 10 29 ¼94%) (2) 20.88 (21.17, 20.58, I 2 ¼86%) (32) 9 11 13 15 24 27 28 34 38 41 44 46 49 51 57 61 63 66 68 20.69 (21.15, 20.23, I 2 9 11 13 14 ¼85%) (13) 29 36 37 41 42 44 51 57 20.11 (20.54, 0.32, 20.21 (20.80, 0.38, I 2 ¼NE%) (1) 40 I 2 ¼NE%) (1) 40 0.48 (20.58, 0.58, I 2 ¼NE%) 20.49 (21.47, 0.49, 21.08 (22.94, 0.78, (1) 10 29 I 2 ¼77%) (2) 10 54 I 2 10 29 ¼89%) (2) 22.05 (22.89, 21.21, I 2 ¼94%) (22) 7111314242829 34 36 38 41 44 46 49 51 55 57 68 20.40 (20.60, 20.20, I 2 ¼75%) (34) 79 11 13 15 23 27 28 31 32 34 38 41 44 46 47 49 51 57 62 64 66 20.54 (20.90, 20.18, I 2 ¼73%) (16) 7911131423 29 31 36 38 41 42 46 51 57 (20.43, 1.40, I 2 ¼0%) 29 54 (2) 28.04 (210.03, 26.05, I 2 ¼95%) (26) 81113142325 27 29 33 35 38 41 44 46 47 51 20.70 (21.48, 0.08, I 2 ¼NE) 20.71 (21.42, 0.00, 20.77 (21.89, 0.34, 219.24 (226.49, 211.98, (1) 40 I 2 ¼77%) (6) 26 40 45 48 50 59 I 2 ¼74%) (2) 40 59 I 2 40 48 50 ¼38%) (3) 54 58 Pregabalin and postoperative pain BJA

BJA Mishriky et al. Pregabalin Control Study or Subgroup Mean SD Total Mean SD Total Weight 2.1.1 Pregabalin 75 mg George jain Jokela BJA 2008 Kim 2011 Kim JC 2011 Nutthachote Peng Sagit White Subtotal (95% Cl) 1.4 2.2 1.29 2 3 3.17 3 1.38 3 1.4 0.69 1.01 1.75 2.22 1.21 2.59 1.17 2 31 20 30 42 28 27 96 50 27 351 1.9 3.5 0.81 3 3 2.84 4 2.09 2 1.6 1.2 0.98 1.5 2.22 1.21 2.22 1.45 2 30 20 28 42 28 27 46 47 27 295 Heterogeneity: Tau 2 = 0.46; Chi 2 = 37.94, df = 8 (P < 0.00001); I 2 =79% Test for overall effect: Z = 1.28 (P = 0.20) 2.1% 2.4% 2.6% 2.2% 1.4% 2.3% 2.0% 2.5% 1.6% 19.0% Mean Difference IV, Random, 95% Cl 0.50 [ 1.26, 0.26] 1.30 [ 1.91, 0.69] 0.48 [ 0.03, 0.99] 1.00 [ 1.70, 0.30] 0.00 [ 1.16, 1.16] 0.33 [ 0.32, 0.98] 1.00 [ 1.82, 0.18] 0.71 [ 1.24, 0.18] 1.00 [ 10.07, 2.07] 0.33 [ 0.85, 0.18] Mean Difference IV, Random, 95% Cl 2.1.2 Pregabalin 100 150 mg Agarwal 2 1.48 27 3.5 2.96 29 1.4% 1.50 [ 2.71, 0.29] Bekawi 0.6 0.62 30 1.13 0.63 30 2.9% 0.53 [ 0.85, 0.21] Buvanendran 2012 3.44 1.9 32 3.32 2.55 16 1.1% 0.12 [ 1.29, 1.53] Cabrera Schulmeyer 0.77 1.33 39 2.1 2.83 41 1.7% 1.33 [ 2.29, 0.37] Choi 2 1.48 36 3 2.96 36 1.5% 1.00 [ 2.08, 0.08] Clendenen 3.1 2.4 23 3.9 2.7 24 1.1% 0.80 [ 2.26, 0.66] Fassoulaki 2.5 1.92 36 3.1 1.79 39 1.9% 0.60 [ 1.44, 0.24] George 1.1 1.2 28 1.9 1.6 30 2.1% 0.80 [ 1.52, -0.08] Jo 1.6 0.7 20 1.7 0.7 20 2.7% 0.10 [ 0.53, 0.33] Jokela BJA 2008 1.37 1.26 26 0.81 0.98 28 2.4% 0.56 [ 0.05, 1.17] Jokela Pain 2008 1.47 1.46 27 1.11 1.37 29 2.1% 0.36 [ 0.38, 1.10] Joshi 1 0.59 20 1.8 0.59 20 2.8% 0.80 [ 1.17, 0.43] Kim 2010 1.6 1.1 47 2.2 1.2 47 2.6% 0.60 [ 1.07, 0.13] K1mJC 2011 3 1.48 28 3 2.22 28 1.7% 0.00 [ 0.99, 0.99] Koyuncu 1 0.96 30 1 1.48 30 2.3% 0.00 [ 0.63, 0.63] Ozgencil 1.1 1.18 30 0.77 30 2.6% 0.40 [ 0.90, 0.10] Sagit 0.33 0.66 46 2.09 1.45 47 2.7% 1.76 [ 2.22, 1.30] Sahu 1.97 1.75 3.93 2.24 35 1.8% 1.96 [ 2.90, 1.02] Spreng 2.36 1.53 22 2.62 1.39 24 1.9% 0.26 [ 1.11, 0.59] Sundar 2.07 0.74 30 2 0.64 30 2.8% 0.07 [ 0.28, 0.42] White 3 2 27 2 2 27 1.6% 1.00 [ 0.07, 2.07] YaDeau 3.4 2.4 21 3 1.9 21 1.2% 0.40 [ 0.91, 1.71] Yucel 1.57 0.6 30 1.73 0.9 30 2.8% 0.16 [ 0.55, 0.23] Subtotal (95% Cl) 690 691 47.8% 0.44 [ 0.71, 0.16] Heterogeneity: Tau 2 = 0.29; Chi 2 = 94.68, df = 22 (P < 0.00001); I 2 = 77% Test for overall effect: Z = 3.14 (P = 0.002) 35 1.5 2.1.3 Pregabalin 300 mg Alimian Burke Chang Demirhan Eskander Gianesello Gonano lttichaikuthol Jokela Pain 2008 Lee Mathiesen 2008 Mathiesen 2009 Mathiesen 2011 Wang White Yucel 0.6 1.73 2.1 0 2.1 0.6 2.6 2 0.86 1.42 1.28 1.39 3.71 4.08 4 1.47 0.8 2.02 1.8 0 0.79 0.8 1.2 1.3 1.06 0.5 1.12 1.78 2.56 1.5 3 0.5 2.5% 1.4% 2.2% 2.8% 2.9% 2.1% 2.2% 2.3% 2.9% 2.5% 2.0% 1.5% 2.1% 1.2% 2.8% Subtotal (95% Cl) 514 511 33.3% Heterogeneity: Tau 2 = 0.28; Chi 2 = 66.36, df = 14 (P < 0.00001); I 2 = 79% Test for overall effect: Z = 2.02 (P =0.04) 40 18 39 20 40 30 20 38 29 31 40 35 45 32 27 30 1.6 2.38 1.8 0 1.95 0.5 2.6 3.4 1.11 2.38 1.29 1.65 4.67 5.12 2 1.73 1.5 1.77 1.4 2.96 0.83 0.5 1.2 1.8 1.37 0.68 1.29 1.8 2.83 1.54 2 0.9 40 20 38 20 40 30 20 40 29 29 38 39 43 28 27 30 1.00 [ 1.53, 0.47] 0.65 [ 1.86, 0.56] 0.30 [ 0.42, 1.02] Not estimable 0.15 [ 0.21, 0.51] 0.10 [ 0.24, 0.44] 0.00 [ 0.74, 0.74] 1.40 [ 2.09, 0.71] 0.25 [ 0.88, 0.38] 0.96 [ 1.26, 0.66] 0.01 [ 0.55, 0.53] 0.26 [ 1.08, 0.56] 0.96 [ 2.09, 0.17] 1.04 [ 1.81, 0.27] 2.00 [0.64, 3.36] 0.26 [ 0.63, 0.11] 0.33 [ 0.65, 0.01] Total (95% Cl) 1555 1497 100.0% 0.38 [ 0.57, 0.20] Heterogeneity: Tau 2 = 0.29; Chi 2 = 200.02, df = 46 (P < 0.00001); I 2 = 77% Test for overall effect: Z = 4.02 (P < 0.0001) Test for subgroup differences: Chi 2 = 0.29, df = 2 (P =0.87); I 2 =0% 2 1 0 1 2 Favours pregabalin Favours control Fig 3 Forest plot for pain scores at rest at. SD, standard deviation; CI, confidence interval; IV, inverse variance. 24