Scolaris Content Display Scolaris Content Display

Cochrane Database of Systematic Reviews

Infusión continua de lidocaína intravenosa perioperatoria para el dolor posoperatorio y la recuperación en adultos

Información

DOI:
https://doi.org/10.1002/14651858.CD009642.pub3Copiar DOI
Base de datos:
  1. Cochrane Database of Systematic Reviews
Versión publicada:
  1. 04 junio 2018see what's new
Tipo:
  1. Intervention
Etapa:
  1. Review
Grupo Editorial Cochrane:
  1. Grupo Cochrane de Anestesia

Copyright:
  1. Copyright © 2018 The Cochrane Collaboration. Published by John Wiley & Sons, Ltd.

Cifras del artículo

Altmetric:

Citado por:

Citado 0 veces por enlace Crossref Cited-by

Contraer

Autores

  • Stephanie Weibel

    Department of Anaesthesia and Critical Care, University of Würzburg, Würzburg, Germany

  • Yvonne Jelting

    Department of Anaesthesia and Critical Care, University of Würzburg, Würzburg, Germany

  • Nathan L Pace

    Department of Anesthesiology, University of Utah, Salt Lake City, USA

  • Antonia Helf

    Department of Anaesthesia and Critical Care, University of Würzburg, Würzburg, Germany

  • Leopold HJ Eberhart

    Department of Anaesthesiology & Intensive Care Medicine, Philipps‐University Marburg, Marburg, Germany

  • Klaus Hahnenkamp

    Department of Anesthesiology, University Hospital, Greifswald, Germany

  • Markus W Hollmann

    Department of Anaesthesiology, Academic Medical Center (AMC) University of Amsterdam, Amsterdam, Netherlands

  • Daniel M Poepping

    Department of Anesthesiology and Intensive Care, University Hospital Münster, Münster, Germany

  • Alexander Schnabel

    Department of Anaesthesia and Critical Care, University of Würzburg, Würzburg, Germany

  • Peter Kranke

    Correspondencia a: Department of Anaesthesia and Critical Care, University of Würzburg, Würzburg, Germany

    [email protected]

Contributions of authors

Stephanie Weibel (SW), Yvonne Jelting (YJ), Nathan Leon Pace (NLP), Antonia Helf (AH), Leopold HJ Eberhart (LE), Klaus Hahnenkamp (KH), Markus W Hollmann (MH), Daniel M Poepping (DP), Alexander Schnabel (AS), Peter Kranke (PK)

(Original review: Kranke 2015)

Conceiving the review: AS, PK

Co‐ordinating the review: original review ‐ PK; update ‐ SW, PK

Undertaking manual searches: original review ‐ JJ, SW; update ‐ YJ, SW

Screening search results: original review ‐ JJ, SW; update ‐ SW, YJ, AH

Organizing retrieval of papers: original review ‐ JJ, SW, PK; update ‐ SW, YJ, PK

Screening retrieved papers against inclusion criteria: original review ‐ JJ, SW; update ‐ SW, YJ, AH

Appraising quality of papers: original review ‐ JJ, SW; update ‐ SW; YJ, AH

Abstracting data from papers: original review ‐ SW, JJ, PK, DP, LE, AS, KH, MH; update ‐ SW, YJ, AH

Writing to authors of papers for additional information ‐ original review: SW; update ‐ SW, YJ

Providing additional data about papers: SW

Obtaining and screening data on unpublished studies: SW

Data management for the review: SW

Entering data into Review Manager 5 (RevMan 5; Review Manager 2014): SW

RevMan statistical data: SW, NLP

Other statistical analysis not using RevMan: original review ‐ PK, NLP; update ‐ SW, NLP

Double entry of data: original review ‐ (data entered by person one: SW; data checked by person two: JJ); update ‐ data entered by person one: SW; data checked by person two: YJ)

Interpretation of data: original review ‐ PK, SW, LE, MH, KH, DP, NLP; update ‐ PK, SW, LE, MH, KH, DP, NLP, AS, YJ, AH

Statistical inferences: original review ‐NLP, PK, SW; update ‐ NLP, PK, SW

Writing the review: original review: SW, PK, JJ, MH; update ‐ SW, PK, YJ, AH

Securing funding for the review: PK

Performing previous work that was the foundation of the present study: original review ‐ MH, KH, PK; update: PK

Guarantor for the review (one author): PK

Person responsible for reading and checking review before submission: PK

Sources of support

Internal sources

  • Departmental resources only, Germany.

External sources

  • No sources of support supplied

Declarations of interest

Stephanie Weibel: none known.

Yvonne Jelting: none known.

Nathan Leon Pace: has no conflict of interest regarding the topic of this review. Nathan L Pace has received payment for development of educational presentations (Barash, Cullen, Stoelting Clinical Anesthesia 8th edition) and provided consultancy (St Marks Hospital, Salt Lake City, UT) on topics not related to the current review. He has received supplements to attend Cochrane meetings. He also has stocks and shares in companies which have no interest in the topic of this review (TIAA‐CREF, Fidelity, Vanguard, USAA, Morgan Stanley).

Antonia Helf: none known.

Leopold HJ Eberhart: has no conflict of interest regarding the topic of this review. Leopold HJ Eberhart has received lecture fees (from
Baxter GmbH and Fresenius GmbH), payment for lectures (from Grünenthal GmbH, Baxter GmbH and Fresenius, GmbH) and has
provided consultancy (for Grünenthal GmbH, Baxter GmbH, ratiopharm GmbH) for topics not related to the current review. He
holds a board membership (with Grünenthal GmbH Deutschland) who do not have an interest in the topic of this review.

Klaus Hahnenkamp: is working in this research area and has participated in a clinical study that is relevant for this systematic review (Herroeder 2007). Critical appraisal and data extraction were done by JJ and SW.

Markus W Hollmann: is working in this research area and has participated in a clinical study that is relevant for this systematic review (Herroeder 2007). Critical appraisal and data extraction were done by JJ and SW.

Daniel M Poepping: none known.

Alexander Schnabel: none known.

Peter Kranke: has no conflict of interest regarding the topic of this review. Peter Kranke has received lecture fees (from FreseniusKabi,
MSD, Ratiopharm, Covidien) and has provided consultancy (to MSD, FreseniusKabi, Ratiopharm, Covidien) on topics not related
to the current review. He has been involved in the conduct of Phase II and phase III clinical trials not related to the current review.

Acknowledgements

We would like to thank Jane Cracknell (Managing Editor, Cochrane Anaesthesia, Critical and Emergency Care Group (ACE), formerly Cochrane Anaesthesia Review Group (CARG)), Mathew Zacharias (Content Editor), Marialena Trivella (Statistical Editor), Jane Ballantyne and William Zempsky (Peer Reviewers), and Janet Wale (Consumer Editor) for their help and editorial advice during the preparation of the protocol for the review (Selig 2012); and Karen Hovhannisyan (ACE Information Specialist) for performing the professional literature search.

We would like to thank Andy Smith (Content Editor), Vibeke E Horstmann (Statistical Editor), Andrew Moore and Marc Gentili (Peer Reviewers), and Janet Wale (Consumer Editor) for their help and editorial advice during the preparation of the review (Kranke 2015).

We would like to thank Andrew Smith (content editor), Vibeke E Horstmann (statistical editor), Marc Gentili,Tim R McCormick (peer reviewers), Janet Wale (consumer editor) for their help and editorial advice during the preparation of the updated systematic review.

We would like to thank Elisabeth Friedrich‐Würstlein (University library, Wuerzburg) for her assistance during the literature search and organizing full texts of the relevant publications.

We thank Bita Mesgarpour for translation and data abstraction of the included Persian study (Ismail 2008); and Taru Jokinen for helping with the translation of Spanish articles during the search process.

We thank Wolfgang Viechtbauer, author of the R metafor package, for advice on multivariate meta‐analytic methods for independent tau2 estimation in subgroups.

We would like to explicitly thank Laurence Weinberg for the willingness to answer all of our open questions on his manuscript patiently until the end (Weinberg 2016). He was the only author that showed this commitment.

We would like to thank Johanna Jokinen for her contribution to the original review (Kranke 2015).

Version history

Published

Title

Stage

Authors

Version

2018 Jun 04

Continuous intravenous perioperative lidocaine infusion for postoperative pain and recovery in adults

Review

Stephanie Weibel, Yvonne Jelting, Nathan L Pace, Antonia Helf, Leopold HJ Eberhart, Klaus Hahnenkamp, Markus W Hollmann, Daniel M Poepping, Alexander Schnabel, Peter Kranke

https://doi.org/10.1002/14651858.CD009642.pub3

2015 Jul 16

Continuous intravenous perioperative lidocaine infusion for postoperative pain and recovery

Review

Peter Kranke, Johanna Jokinen, Nathan Leon Pace, Alexander Schnabel, Markus W Hollmann, Klaus Hahnenkamp, Leopold HJ Eberhart, Daniel M Poepping, Stephanie Weibel

https://doi.org/10.1002/14651858.CD009642.pub2

2012 Feb 15

Continuous intravenous perioperative lidocaine infusion for postoperative pain and recovery

Protocol

Christian Selig, Alexander Schnabel, Markus W. Hollmann, Klaus Hahnenkamp, Leopold H. J. Eberhart, Daniel Poepping, Niklas Hahn, Nathan Leon Pace, Peter Kranke

https://doi.org/10.1002/14651858.CD009642

Differences between protocol and review

Changes to the authors of the review since publication of the protocol (Selig 2012)

In 2015 (Kranke 2015)

  1. Two authors (C Selig and N Hahn) were no longer involved with the review and were removed from the list of authors.

  2. Two new authors (J Jokinen and S Weibel) were added to the authors list and contributed to the review, as described in the Contributions of authors section.

For the current update:

  1. Peter Kranke moved from first to last author (contact author). Stephanie Weibel became the new first author.

  2. Johanna Jokinen left the review team.

  3. Yvonne Jelting and Antonia Helf were newly added.

Differences in the methods used between the protocol (Selig 2012), and the review (Kranke 2015)

  1. Criteria for considering studies for this review: we added the following to the review 'The IV lidocaine infusion, must have been started intraoperatively (with or without an IV bolus) prior to incision and continued until the end of surgery.' In the protocol we only described 'to have been continued postoperatively'.

  2. At the protocol stage we planned to include quasi‐RCTs if it were found that few RCTs were available for meta‐analysis. However, in the review we did not include quasi‐RCTs due to the large number of available RCTs, which present the best available evidence, regarding the topic of interest.

  3. We did not pre‐specify in the protocol for this review at which time periods the relevant outcomes of this review should be analysed. Based on pharmacodynamic and pharmacokinetic considerations and the logic in many included trials in which the postoperative observation period was divided into at least two distinct time intervals, we decided to subdivide outcome reporting for pain, postoperative opioid consumption, nausea and vomiting, etc. into different postoperative time points (e.g. 'early' and 'late'/'overall') to cover most of the reported data adequately.

  4. In the protocol we planned to include pain data reported on VAS 0 to 100 mm scale. Due to the large proportion of data reported on other scales, we decided to include all pain data presented on a VAS 0 to 100 mm scale, NRS 0 to 10 and VRS 0 to 10 (0 = no pain, 10 = worst pain), and VAS 0 to 10 cm.

  5. We broadened the outcome 'time to first bowel sounds' and included data on 'time to first bowel movement'.

  6. The outcomes 'intraoperative and postoperative opioid requirements' were not previously considered in the published protocol. However, after intensive study of the relevant published trials dealing with perioperative lidocaine infusion for reduction of postoperative pain, we recognized that this outcome was widely analysed within the studies and we believe that opioid consumption is another relevant outcome to understand the effect of lidocaine in the perioperative setting since it may also affect the postoperative recovery and occurrence of side effects, e.g. ileus, nausea or vomiting.

  7. Measurement of treatment effects: in the protocol we planned for data on pain scores, neuropsychological status or patient satisfaction that are reported on disparate scales, to calculate the standardized mean difference (SMD) obtained from the MD and SD. In the published review we combined, for the outcome 'pain', all data presented on either VAS 0 to 10 cm scale, VAS 0 to 100 mm, NRS 0 to 10, or VRS 0 to 10 (0 = no pain, 10 = worst pain) and transformed the first three into VAS 0 to 10 cm and presented the effect estimates as MD.

  8. Dealing with missing data: in the protocol we planned to perform complete‐case analyses if there were exclusions/dropouts in the study flow. We intended to perform sensitivity analyses by inputting missing data (best case and worst case) in instances of more than trivial missing data. To the review we added the following statement which explains the handling of missing data which are obviously not crucial for the overall estimation of the treatment effect: 'If data were missing due to random events and the impact of missing data was considered marginal, we included data in the analysis only on those participants whose results were known. Studies with incomplete reporting of their study flow or disputable exclusions were subsequently excluded in a sensitivity meta‐analysis to assess bias. The potential impact of the missing data on the results was considered in the interpretation of the results of the review.'

  9. In the protocol we did not pre‐specify that we will include median values and IQR. However, during data extraction we recognized that the data in large part were reported as median and IQR. Since we wanted to include as much data as possible, we calculated in the review the mean and SD from median and IQR in accordance with Higgins 2011. To assess the impact of the median data on the summary statistics, we performed a sensitivity analysis.

  10. Assessment of heterogeneity: in the protocol we planned to perform meta‐regressions to explain heterogeneity. In the present review, we did not perform these calculations.

  11. 'Summary of findings' table: at the protocol stage we planned to present results on pain scores and gastrointestinal recovery within 'Summary of findings' tables. We decided post‐analysis to additionally present nausea as an outcome of public interest. We presented further the results of the different surgical subgroups (open abdominal, laparoscopic abdominal, and other surgeries) for the outcome 'pain (early)' to reflect the specific benefit for abdominal surgery patients.

  12. Sensitivity analysis: in the protocol we planned to perform a sensitivity analysis using the inverse variance weighted fixed‐effect model. Due to the large heterogeneity observed between the studies the random‐effects model fits much better than the fixed‐effect model. Therefore, we did not perform this sensitivity analysis.

  13. Sensitivity analysis: we analysed the impact of data reported as median and IQR on the overall effect estimation to each outcome to judge the robustness of the summary statistics.

  14. Sensitivity analysis: we planned in the protocol to perform a sensitivity analysis including only low risk of bias studies to test the robustness of the summary statistics. Since only few studies received an overall low risk classification, we reconsidered that point in the review and proceeded to exclude the high risk of bias studies to judge the robustness of the summary statistics.

Differences in the methods used between the published review (Kranke 2015), and the updated review

  1. We have changed the title from 'Continuous intravenous perioperative lidocaine infusion for postoperative pain and recovery' to 'Continuous intravenous perioperative lidocaine infusion for postoperative pain and recovery in adults' to reflect the focus on the adult population.

  2. We have made changes to the list of primary outcomes: due to the high priority of adverse effects in the investigation of medical interventions, we moved the outcome 'adverse events' from secondary to primary outcomes.

  3. We separated the outcome 'time to first bowel movement/sounds' into 'time to first bowel movement' and 'time to first bowel sounds'. We combined the outcomes 'time to first defaecation' and 'time to first bowel movement' into one outcome named as 'time to first defaecation/bowel movement' since both outcomes measure the same clinical condition.

  4. We have made changes to the 'Summary of findings' table (GRADE‐relevant outcomes). We have reduced the number of 'Summary of findings' tables to one per comparison and created the following outcome categories: pain (early, intermediate, and late time points), gastrointestinal recovery (postoperative ileus, time to first defaecation/bowel movement), adverse events, postoperative nausea (overall), postoperative opioid consumption (overall). The outcomes 'time to first bowel sound', 'time to first flatus', and 'postoperative nausea (early)' are no longer GRADE‐relevant outcomes.

  5. We performed meta‐analyses for the following new outcomes due to availability of more than three studies: length of hospital stay (outpatient surgery, mins), surgical complications (anastomotic leak), surgical complications (bleeding), patient satisfaction, and intraoperative remifentanil consumption.

  6. We omitted studies with suspected small variance reporting for the outcomes: pain, gastrointestinal recovery, and opioid consumption.

  7. Assessment of risk of other bias: we no longer assessed a lack of sample size calculation in trials as high risk of other bias.

  8. In contrast to the original review, we calculated the SMD as summary statistics for all pain outcomes since several different scales were used in the individual trials. The use of SMD as summary statistics was originally described in the protocol.

  9. We changed 'dealing with missing data' to the method described in the original protocol.

  10. We introduced the 95% prediction interval (PI) to understand the uncertainty associated with an intervention about whether an intervention works or not in the light of between‐study heterogeneity.

  11. We used the Mantel‐Haenszel method for RRs instead of inverse variance weighting.

  12. We have changed the 'Summary of findings' table with respect to presented outcomes (we removed subgroups for pain 'early' and added adverse events plus postoperative opioid consumption 'overall') and to the approach for assessing inconsistency and imprecision (see 95% PI).

  13. Sensitivity analysis (fixed‐effect model): as described in the protocol, we included in the current update sensitivity analyses using the fixed‐effect model.

  14. Sensitivity analysis (risk of bias): as in the protocol described, we performed in the current update sensitivity analyses, including only low risk of bias studies to test the robustness of the summary statistics.

  15. Sensitivity analysis (suspected variance reporting): we added studies with suspected variance reporting to the meta‐analyses of relevant outcomes to explore the impact on the effect estimates in sensitivity meta‐analyses.

Keywords

MeSH

PICO

Population
Intervention
Comparison
Outcome

El uso y la enseñanza del modelo PICO están muy extendidos en el ámbito de la atención sanitaria basada en la evidencia para formular preguntas y estrategias de búsqueda y para caracterizar estudios o metanálisis clínicos. PICO son las siglas en inglés de cuatro posibles componentes de una pregunta de investigación: paciente, población o problema; intervención; comparación; desenlace (outcome).

Para saber más sobre el uso del modelo PICO, puede consultar el Manual Cochrane.

Study flow diagram.
Figuras y tablas -
Figure 1

Study flow diagram.

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.
Figuras y tablas -
Figure 2

Risk of bias summary: review authors' judgements about each risk of bias item for each included study.

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.
Figuras y tablas -
Figure 3

Risk of bias graph: review authors' judgements about each risk of bias item presented as percentages across all included studies.

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 1 Pain score at rest, 'early time points' (1 h to 4 h, PACU).
Figuras y tablas -
Analysis 1.1

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 1 Pain score at rest, 'early time points' (1 h to 4 h, PACU).

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 2 Pain score at rest, 'intermediate time points' (24 h).
Figuras y tablas -
Analysis 1.2

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 2 Pain score at rest, 'intermediate time points' (24 h).

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 3 Pain score at rest, 'late time points' (48 h).
Figuras y tablas -
Analysis 1.3

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 3 Pain score at rest, 'late time points' (48 h).

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 4 Postoperative ileus (dichotomous).
Figuras y tablas -
Analysis 1.4

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 4 Postoperative ileus (dichotomous).

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 5 Time to first defaecation/bowel movement (h).
Figuras y tablas -
Analysis 1.5

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 5 Time to first defaecation/bowel movement (h).

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 6 Time to first flatus (h).
Figuras y tablas -
Analysis 1.6

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 6 Time to first flatus (h).

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 7 Time to first bowel sounds (h).
Figuras y tablas -
Analysis 1.7

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 7 Time to first bowel sounds (h).

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 8 Length of hospital stay (days).
Figuras y tablas -
Analysis 1.8

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 8 Length of hospital stay (days).

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 9 Length of hospital stay (outpatient surgery, mins).
Figuras y tablas -
Analysis 1.9

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 9 Length of hospital stay (outpatient surgery, mins).

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 10 Surgical complications ‐ anastomotic leak.
Figuras y tablas -
Analysis 1.10

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 10 Surgical complications ‐ anastomotic leak.

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 11 Surgical complications ‐ bleeding.
Figuras y tablas -
Analysis 1.11

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 11 Surgical complications ‐ bleeding.

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 12 Surgical complications ‐ postoperative infection.
Figuras y tablas -
Analysis 1.12

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 12 Surgical complications ‐ postoperative infection.

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 13 Patient satisfaction.
Figuras y tablas -
Analysis 1.13

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 13 Patient satisfaction.

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 14 Postoperative nausea, 'early time points' (PACU).
Figuras y tablas -
Analysis 1.14

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 14 Postoperative nausea, 'early time points' (PACU).

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 15 Postoperative nausea, 'overall' (0 to 24 h, to 48 h, to 72 h).
Figuras y tablas -
Analysis 1.15

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 15 Postoperative nausea, 'overall' (0 to 24 h, to 48 h, to 72 h).

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 16 Postoperative vomiting, 'early time points' (PACU).
Figuras y tablas -
Analysis 1.16

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 16 Postoperative vomiting, 'early time points' (PACU).

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 17 Postoperative vomiting, 'overall' (0 to 24 h, to 48 h, to 72 h).
Figuras y tablas -
Analysis 1.17

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 17 Postoperative vomiting, 'overall' (0 to 24 h, to 48 h, to 72 h).

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 18 Intraoperative opioid consumption (MEQ, mg).
Figuras y tablas -
Analysis 1.18

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 18 Intraoperative opioid consumption (MEQ, mg).

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 19 Intraoperative remifentanil consumption (µg).
Figuras y tablas -
Analysis 1.19

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 19 Intraoperative remifentanil consumption (µg).

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 20 Postoperative opioid consumption, PACU (MEQ, mg).
Figuras y tablas -
Analysis 1.20

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 20 Postoperative opioid consumption, PACU (MEQ, mg).

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 21 Postoperative opioid consumption, overall (MEQ, mg).
Figuras y tablas -
Analysis 1.21

Comparison 1 Intravenous (IV) lidocaine versus placebo, Outcome 21 Postoperative opioid consumption, overall (MEQ, mg).

Comparison 2 Intravenous (IV) lidocaine versus thoracic epidural analgesia (TEA), Outcome 1 Pain score (VAS 0 to 10 cm) at rest, 'intermediate time points' (24 h).
Figuras y tablas -
Analysis 2.1

Comparison 2 Intravenous (IV) lidocaine versus thoracic epidural analgesia (TEA), Outcome 1 Pain score (VAS 0 to 10 cm) at rest, 'intermediate time points' (24 h).

Comparison 2 Intravenous (IV) lidocaine versus thoracic epidural analgesia (TEA), Outcome 2 Pain score (VAS 0 to 10 cm) at rest, 'late time points' (48 h).
Figuras y tablas -
Analysis 2.2

Comparison 2 Intravenous (IV) lidocaine versus thoracic epidural analgesia (TEA), Outcome 2 Pain score (VAS 0 to 10 cm) at rest, 'late time points' (48 h).

Comparison 2 Intravenous (IV) lidocaine versus thoracic epidural analgesia (TEA), Outcome 3 Time to first bowel movement (h).
Figuras y tablas -
Analysis 2.3

Comparison 2 Intravenous (IV) lidocaine versus thoracic epidural analgesia (TEA), Outcome 3 Time to first bowel movement (h).

Comparison 2 Intravenous (IV) lidocaine versus thoracic epidural analgesia (TEA), Outcome 4 Length of hospital stay (days).
Figuras y tablas -
Analysis 2.4

Comparison 2 Intravenous (IV) lidocaine versus thoracic epidural analgesia (TEA), Outcome 4 Length of hospital stay (days).

Comparison 2 Intravenous (IV) lidocaine versus thoracic epidural analgesia (TEA), Outcome 5 Intraoperative opioid consumption (MEQ, mg).
Figuras y tablas -
Analysis 2.5

Comparison 2 Intravenous (IV) lidocaine versus thoracic epidural analgesia (TEA), Outcome 5 Intraoperative opioid consumption (MEQ, mg).

Summary of findings for the main comparison. Intravenous (IV) lidocaine compared to placebo or no treatment in patients undergoing any elective or urgent surgical procedure under general anaesthesia

IV Lidocaine compared to placebo or no treatment in patients undergoing any elective or urgent surgical procedure under general anaesthesia

Patient or population: adult patients undergoing any elective or urgent surgical procedure under general anaesthesia
Settings: Asia (24 trials); USA, Canada, and South America (18 trials); Europe (15 trials); Middle East (7 trials); New Zealand and Australia (4 trials)
Intervention: IV lidocaine
Comparison: placebo or no treatment

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect (95% CI)

Prediction interval
(95% PI)

No. of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk with placebo or no treatment

Corresponding risk with IV lidocaine

1. Pain (VAS 0 to 10 cm, 0 to 100 mm, NRS 0 to 10)

Pain score at rest, 'early time points' (1 hto 4 hpostoperatively, or in the PACU)

(1.61 lower to 0.62 higher)

1656
(29 RCTs)

⊕⊝⊝⊝
Very lowa,b,c,d

A SMD of 0.50 fewer is equivalent to a range of 0.37 cm fewer (SD = 0.74 cm) to 2.48 cm fewer (SD = 4.95 cm) on a VAS 0 to 10 cm scale in the intervention group.

The range of mean effects that can be expected in a future study (95% PI) includes both benefit and harmk.

The standardized mean pain score in the intervention group was 0.50 lower (0.72 lower to 0.28 lower)

Pain score at rest, 'intermediate time points' (24 hpostoperatively)

(0.44 lower to 0.16 higher)

1847
(33 RCTs)

⊕⊕⊕⊝
Moderatee,f,g,h,i

A SMD of 0.14 fewer is equivalent to a range of 0.10 cm fewer (SD = 0.74 cm) to 0.48 cm fewer (SD = 3.42 cm) on a VAS 0 to 10 cm scale in the intervention group.

The range of mean effects that can be expected in a future study (95% PI) is clinically not relevantk.

The standardized mean pain score in the intervention group was 0.14 lower (0.25 lower to 0.04 lower)

Pain score at rest, 'late time points' (48 hpostoperatively)

(0.60 lower to 0.38 higher)

1404
(24 RCTs)

⊕⊕⊕⊝
Moderatee,f,g,h,i

A SMD of 0.11 fewer is equivalent to a range of 0.08 cm fewer (SD = 0.7 cm) to 0.42 cm fewer (SD = 3.8 cm) on a VAS 0 to 10 cm scale in the intervention group.

The range of mean effects that can be expected in a future study (95% PI) is clinically not relevantk.

The standardized mean pain score in the intervention group was 0.11 lower (0.25 lower to 0.04 higher)

2. Gastrointestinal recovery

Postoperative ileus (dichotomous)

The number of participants with postoperative ileus

RR 0.37 (0.15 to 0.87)

(0.05 lower to 2.43 higher)

273
(4 RCTs)

⊕⊝⊝⊝
Very lowa,b,c

The range of mean effects that can be expected in a future study (95% PI) includes both benefit and harm.

131 per 1000

48 per 1000 (20 to 114)

Time to first defaecation/bowel movement (h)

(22.19 h shorter to 6.36 h longer)

684
(12 RCTs)

⊕⊝⊝⊝
Very lowa,b,c

The range of mean effects that can be expected in a future study (95% PI) includes both benefit and clinical non‐relevance.

The mean time to first defaecation/bowel movement in the control group ranged from 24 h to 94 h

The mean time to first defaecation/bowel movement in the intervention group was 7.92 h shorter (12.71 h shorter to 3.13 h shorter)

3. Adverse events

(e.g. the number of participants that died, or had arrhythmias, other heart rate disorders, or showed any signs of lidocaine toxicity)

See comment

See comment

See comment

⊕⊝⊝⊝
Very Lowj

Adverse events that were investigated in a few trials are death, arrhythmia, light‐headedness, perioral numbness, and dizziness. The effect of lidocaine on these adverse effects is uncertain.

4. Postoperative nausea, 'overall' (0 to 24 h, to 48 h, to 72 h)

350 per 1000

273 per 1000 (235 to 319)

RR 0.78 (0.67 to 0.91)

(0.49 lower to 1.23 higher)

1903
(35 RCTs)

⊕⊝⊝⊝
Very lowa,b,c,d

The range of mean effects that can be expected in a future study (95% PI) includes both benefit and clinical non‐relevance.

5. Postoperative opioid consumption, 'overall' (MEQ, mg)

The mean postoperative opioid consumption in the control group ranged from 1.13 mg to 233.93 mg

The mean postoperative opioid consumption in the intervention group was 4.52 mg lower (6.25 mg lower to 2.79 mg lower)

(12.03 mg lower to 3.00 mg higher)

2201

(40 RCTs)

⊕⊝⊝⊝
Very lowa,b,c,d

The range of mean effects that can be expected in a future study (95% PI) includes both benefit and clinical non‐relevance.

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; MD: mean difference; MEQ: morphine equivalents; NRS: numeric rating scale; PACU: postanaesthesia care unit; RCT: randomized controlled trial; RR: risk ratio; SMD: standardized mean difference; SD: standard deviation; VAS: visual analogue scale

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: we are moderately confident in the effect estimate; the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low quality: our confidence in the effect estimate is limited; the true effect may be substantially different from the estimate of the effect.
Very low quality: we have very little confidence in the effect estimate; the true effect is likely to be substantially different from the estimate of effect.

aDowngrading for study limitations: substantial information is derived from studies at high or unclear risk of bias (selection bias, blinding, attrition bias). Exclusion of high and unclear risk of bias studies affected the robustness of the estimated effect.

bDowngrading for inconsistency: the 95% PI is significantly wider than the 95% CI (we assume between‐study heterogeneity).

cDowngrading for imprecision: we downgraded for imprecision due to the fact that the 95% PI crosses the line of identity in contrast to the 95% CI.

dPublication bias: test for publication bias suggested funnel plot asymmetry and trim and fill analysis changed the conclusion. We did not downgrade for publication bias since we have already downgraded for inconsistency (true heterogeneity may be a source of funnel plot asymmetry).

eStudy limitations: substantial information is derived from studies at high or unclear risk of bias (selection bias, blinding, attrition bias). However, exclusion of high and unclear risk of bias studies did not affect the robustness of the estimated effect (95% CI: clinical non‐relevant range of effects).

fInconsistency: the 95% PI is wider than the 95% CI (we assume between‐study heterogeneity), but the range of effects lie in areas of clinical non‐relevance. Therefore, we did not downgrade for inconsistency.

gImprecision: we did not downgrade for imprecision since the 95% PI and the 95% CI around the effect size are narrow (precise result with no clinical relevance).

hPublication bias: test for publication bias suggested funnel plot asymmetry but trim and fill analysis did not change the conclusion (95% CI: clinical non‐relevant range of effects).

iDowngrading for study limitations and publication bias: we downgraded by one level for the combination of study limitations and funnel plot asymmetry, because of the uncertain risk of bias domains for over half of the studies and the evidence for publication bias shown by funnel plot asymmetry.

jThere are few trials investigating adverse events with a great heterogeneity in the investigated adverse events and with a lack of systematic assessment and reporting of adverse events which limits quality of evidence. Data of adverse events were not pooled in any meta‐analysis. Downgrading for inconsistency, imprecision, and study quality.

kClinical relevance is assumed if the minimally important difference on the 0 to 10 cm pain scale is approximately 1 cm.

Figuras y tablas -
Summary of findings for the main comparison. Intravenous (IV) lidocaine compared to placebo or no treatment in patients undergoing any elective or urgent surgical procedure under general anaesthesia
Summary of findings 2. Intravenous (IV) lidocaine compared to thoracic epidural analgesia (TEA) in patients undergoing any elective or urgent surgical procedure under general anaesthesia

IV lidocaine compared to TEA in adult patients undergoing any elective or urgent surgical procedure under general anaesthesia

Patient or population: adult patients undergoing any elective or urgent surgical procedure under general anaesthesia
Settings: USA and Canada (two trials)
Intervention: IV lidocaine
Comparison: TEA

Outcomes

Illustrative comparative risks* (95% CI)

Relative effect (95% CI)

Prediction interval
(95% PI)

No. of Participants
(studies)

Quality of the evidence
(GRADE)

Comments

Assumed risk with placebo or no treatment

Corresponding risk with IV lidocaine

1. Pain (VAS 0 to 10 cm, 0 to 100 mm, NRS 0 to 10

Pain score at rest, 'early time points' (1 hto 4 hpostoperatively, or in the PACU)

(0 RCTs)

No trial assessed this outcome.

See comment

See comment

Pain score at rest, 'intermediate time points' (24 hpostoperatively)

(VAS 0 to 10 cm)

Not estimable*

102
(2 RCTs)

⊕⊝⊝⊝
Very lowa,b,c

The estimated effect (95% CI) includes both benefit and harmg.

The mean pain score 'intermediate time points' ranged across control groups from 0 to 3.3 cm

The mean pain score 'intermediate time points' in the intervention group was 1.51 cm higher (0.29 lower to 3.32 higher)

Pain score at rest 'late time points' (48 hpostoperatively)

(VAS 0 to 10 cm)

Not estimable*

102
(2 RCTs)

⊕⊝⊝⊝
Very lowa,b,c

The estimated effect (95% CI) includes both benefit and harmg.

The mean pain score 'late time points' ranged across control groups from 0 to 2.7 cm

The mean pain score 'late time points' in the intervention group was 0.98 cm higher (1.19 lower to 3.16 higher)

2. Gastrointestinal recovery

Postoperative ileus (dichotomous)

The number of participants with postoperative ileus

Not estimable

Not estimable*

60

(1 RCT)

⊕⊝⊝⊝
Very lowa,d

Only one small trial assessed this outcome.

Two out of 30 participants in the control group and one out of 30 in the lidocaine group had postoperative ileus

Time to bowel movements (h)

Not estimable*

102
(2 RCTs)

⊕⊝⊝⊝
Very lowa,e

The estimated effect (95% CI) includes both benefit and harm.

The mean time to first bowel movements (h) ranged across control groups from 39 h to 72 h

The mean time to first bowel movements (h) in the intervention group was 1.66 h shorter (10.88 shorter to 7.56 longer)

3. Adverse events

(e.g. the number of participants that died, or had arrhythmias, other heart rate disorders, or showed any signs of lidocaine toxicity)

See comment

See comment

See comment

⊕⊝⊝⊝
Very lowf

All adverse events that are reported in the individual studies were listed in Table 12.

4. Postoperative nausea, 'overall' (0 to 24 h, to 48 h, to 72 h)

17 out of 30 participants in the control group and 11 out of 30 in the lidocaine group had nausea

Not estimable

Not estimable*

60

(1 RCT)

⊕⊝⊝⊝
Very lowa,d

Only one small trial assessed this outcome.

5. Postoperative opioid consumption, 'overall' (MEQ, mg)

See comment

See comment

(0 RCTs)

No trial assessed this outcome.

*The risk in the intervention group (and its 95% CI) is based on the assumed risk in the comparison group and the relative effect of the intervention (and its 95% CI).

CI: confidence interval; MEQ: morphine equivalents; NRS: numeric rating scale; PACU: postanaesthesia care unit; RCT: randomized controlled trial; VAS: visual analogue scale

GRADE Working Group grades of evidence
High quality: we are very confident that the true effect lies close to that of the estimate of the effect.
Moderate quality: we are moderately confident in the effect estimate; the true effect is likely to be close to the estimate of the effect, but there is a possibility that it is substantially different.
Low quality: our confidence in the effect estimate is limited; the true effect may be substantially different from the estimate of the effect.
Very low quality: we have very little confidence in the effect estimate; the true effect is likely to be substantially different from the estimate of effect.

* The estimate of the PI is imprecise when based on only a few studies with small sample size (IntHout 2016). In this case, we did not provide the 95% PI.

aDowngrading for study limitations: substantial information is derived from studies at high or unclear risk of bias (selection bias, blinding, attrition bias). Exclusion of high and unclear risk of bias studies affected the robustness of the estimated effect.

bDowngrading for inconsistency: between‐study heterogeneity was high for this outcome.

cDowngrading for imprecision: we downgraded for imprecision due to the fact that the 95% CI around the effect size was large.

dDowngrading for imprecision: we double‐downgraded for imprecision since information is derived from only one small trial.

eDowngrading for imprecision: we double‐downgraded for imprecision since the 95% CI around the effect size was large, including benefit and harm. There is a high uncertainty associated with this effect estimate.

fThere is great heterogeneity in the investigated adverse events in the individual trials with a lack of systematic assessment and reporting of adverse events which limits quality of evidence.

gClinical relevance is assumed if the minimally important difference on the 0 to 10 cm pain scale is approximately 1 cm.

Figuras y tablas -
Summary of findings 2. Intravenous (IV) lidocaine compared to thoracic epidural analgesia (TEA) in patients undergoing any elective or urgent surgical procedure under general anaesthesia
Table 1. Sensitivity analyses ‐ risk of bias (incomplete outcome data)

All studies

Without high/unclear risk of bias studies (incomplete outcome data)

Outcome

Statistical method

Studies

Effect estimate

Studies

Effect estimate

Pain score, rest, 'early time points' (1 hr to 4 hrs, PACU)

SMD (IV, Random, 95% CI)

29

−0.50 (−0.72 to −0.28)

17

−0.45 (−0.77 to −0.14)

Pain score, rest, 'intermediate time points' (24 hrs)

SMD (IV, Random, 95% CI)

33

−0.14 (−0.25 to −0.04)

18

−0.12 (−0.26 to 0.01)

Pain score, rest, 'late time points' (48 hrs)

SMD (IV, Random, 95% CI)

24

−0.11 (−0.25 to 0.04)

11

−0.06 (−0.27 to 0.15)

Postoperative ileus (dichotomous)

RR (MH, Random, 95% CI)

4

0.37 (0.15 to 0.87)

4

0.37 (0.15 to 0.87)

Time to first defaecation/bowel movement (hrs)

MD (IV, Random, 95% CI)

12

−7.92 (−12.71 to −3.13)

8

−7.5 (−14.38 to −0.63)

Time to first flatus (hrs)

MD (IV, Random, 95% CI)

13

−4.09 (−6.30 to −1.87)

9

−3.98 (−7.03 to −0.93)

Time to first bowel sounds (hrs)

MD (IV, Random, 95% CI)

2

−6.08 (−13.77 to 1.60)

2

−6.08 (−13.77 to 1.60)

Length of hospital stay (days)

MD (IV, Random, 95% CI)

32

−0.37 (−0.60 to −0.15)

17

−0.23 (−0.49 to 0.02)

Length of hospital stay (outpatient surgery, mins)

MD (IV, Random, 95% CI)

3

−10.81 (−36.93 to 15.31)

3

−10.81 (−36.93 to 15.31)

Surgical complications ‐ anastomotic leak

RR (MH, Random, 95% CI)

3

0.61 (0.08 to 4.80)

3

0.61 (0.08 to 4.80)

Surgical complications ‐ bleeding

RR (MH, Random, 95% CI)

3

1.79 (0.41 to 7.89)

3

1.79 (0.41 to 7.89)

Surgical complications ‐ postoperative infection

RR (MH, Random, 95% CI)

5

1.64 (0.41 to 6.52)

4

1.19 (0.25 to 5.67)

Patient satisfaction

MD (IV, Random, 95% CI)

6

0.76 (0.46 to 1.06)

2

0.59 (−0.09 to 1.26)

Postoperative nausea, early (PACU)

RR (MH, Random, 95% CI)

8

0.72 (0.53 to 0.98)

7

0.66 (0.47 to 0.91)

Postoperative nausea, overall (0 to 24 hrs, to 48 hrs, to 72 hrs)

RR (MH, Random, 95% CI)

35

0.78 (0.67 to 0.91)

19

0.87 (0.72 to 1.06)

Postoperative vomiting, early (PACU)

RR (MH, Random, 95% CI)

4

0.49 (0.16 to 1.48)

3

0.75 (0.15 to 3.80)

Postoperative vomiting, overall (0 to 24 hrs, to 48 hrs, to 72 hrs)

RR (MH, Random, 95% CI)

19

0.83 (0.63 to 1.08)

7

0.88 (0.58 to 1.31)

Intraoperative opioid consumption (MEQ, mg)

MD (IV, Random, 95% CI)

18

−2.14 (−3.87 to −0.40)

10

−1.52 (−4.13 to 1.09)

Intraoperative opioid consumption with remifentanil (MEQ, mg)

MD (IV, Random, 95% CI)

6

−14.17 (−35.27 to 6.92)

5

−16.08 (−41.41 to 9.25)

Postoperative opioid consumption, PACU (MEQ, mg)

MD (IV, Random, 95% CI)

21

−3.10 (−3.87 to −2.32)

12

−2.59 (−3.76 to −1.42)

Postoperative opioid consumption, overall (MEQ, mg)

MD (IV, Random, 95% CI)

40

−4.52 (−6.25 to −2.79)

25

−2.84 (−4.45 to −1.22)

Acronyms and abbreviations used in the table:

CI = confidence interval, hrs = hours, IV = inverse variance, MD = mean difference, MEQ = morphine equivalent dose, mins = minutes, MH = Mantel Haenszel, PACU = post anaesthesia care unit, RR = risk ratio, SMD = standardized mean difference

Figuras y tablas -
Table 1. Sensitivity analyses ‐ risk of bias (incomplete outcome data)
Table 2. Sensitivity analyses ‐ median + interquartile range

Mean + SD and median + IQR values

Only mean + SD values

Outcome

Statistical method

Studies

Effect estimate

Studies

Effect estimate

Pain score , rest, 'early time points' (1 hr to 4 hrs, PACU)

SMD (IV, Random, 95% CI)

29

−0.50 (−0.72 to −0.28)

23

−0.64 (−0.89 to −0.38)

Pain score , rest, 'intermediate time points' (24 hrs)

SMD (IV, Random, 95% CI)

33

−0.14 (−0.25 to −0.04)

27

−0.16 (−0.29 to −0.04)

Pain score , rest, 'late time points' (48 hrs)

SMD (IV, Random, 95% CI)

24

−0.11 (−0.25 to 0.04)

20

−0.12 (−0.29 to 0.04)

Time to first defaecation/bowel movement (hrs)

MD (IV, Random, 95% CI)

12

−7.92 (−12.71 to −3.13)

7

−6.03 (−10.98 to −1.08)

Time to first flatus (hrs)

MD (IV, Random, 95% CI)

13

−4.09 (−6.30 to −1.87)

10

−4.40 (−6.30 to −2.50)

Time to first bowel sounds (hrs)

MD (IV, Random, 95% CI)

2

−6.08 (−13.77 to 1.60)

2

−6.08 (−13.77 to 1.60)

Length of hospital stay (days)

MD (IV, Random, 95% CI)

32

−0.37 (−0.60 to −0.15)

16

−0.32 (−0.54 to −0.10)

Length of hospital stay (outpatient surgery, mins)

MD (IV, Random, 95% CI)

3

−10.81 (−36.93 to 15.31)

0

Not estimable

Patient satisfaction

MD (IV, Random, 95% CI)

6

0.76 (0.46 to 1.06)

1

0.30 (−0.21 to 0.81)

Intraoperative opioid consumption (MEQ, mg)

MD (IV, Random, 95% CI)

18

−2.14 (−3.87 to −0.40)

13

−2.32 (−4.33 to −0.32)

Intraoperative opioid consumption with remifentanil (MEQ, mg)

MD (IV, Random, 95% CI)

6

−14.17 (−35.27 to 6.92)

4

−20.45 (−52.10 to 11.19)

Postoperative opioid consumption, PACU (MEQ, mg)

MD (IV, Random, 95% CI)

21

−3.10 (−3.87 to −2.32)

15

−2.88 (−3.80 to −1.96)

Postoperative opioid consumption, overall (MEQ, mg)

MD (IV, Random, 95% CI)

40

−4.52 (−6.25 to −2.79)

28

−4.64 (−6.72 to −2.56)

Acronyms and abbreviations used in this table:

CI = confidence interval, hrs = hours, IQR = interquartile range, IV = inverse variance, MD = mean difference, MEQ = morphine equivalent dose, mins = minutes, PACU = post anaesthesia care unit, SD = standard deviation, SMD = standardized mean difference

Figuras y tablas -
Table 2. Sensitivity analyses ‐ median + interquartile range
Table 3. Subgroup analyses ‐ type of surgery

Main meta‐analyses

Open abdominal surgery

Laparoscopic surgery

Other surgery

Test for subgroup difference (P)

Outcome

n

Effect estimate

(I2)

n

Effect estimate

(I2)

n

Effect estimate

(I2)

Pain score, rest, 'early time points' (1 hr to 4 hrs, PACU)

8

−0.54 (−0.82 to −0.26)

(51%)

10

−0.78 (−1.34 to −0.21)

(89%)

11

−0.21 (−0.44 to 0.02)

(56%)

0.07

Pain score, rest, 'intermediate time points' (24 hrs)

No subgroup analysis performed (I2 < 30%)

Pain score, rest, 'late time points' (48 hrs)

7

0.03 (−0.17 to 0.23)

(0%)

7

−0.30 (−0.74 to 0.13)

(74%)

10

−0.10 (−0.27 to 0.08)

(20%)

0.35

Postoperative ileus (dichotomous)

No subgroup analysis performed (I2 < 30%)

Time to first defaecation/bowel movement (hrs)

6

−7.09 (−10.33 to −3.86)

(0%)

5

−6.23 (−18.07 to 5.62)

(85%)

1

−6.10 (−24.49 to 12.29)

NE

0.41

Time to first flatus (hrs)

6

−4.49 (−7.38 to −1.60)

(6%)

5

−3.07 (−8.28 to 2.15)

(78%)

2

−2.15 (−3.56 to −0.74)

(0%)

0.36

Time to first bowel sounds (hrs)

1

−10.00 (−17.13 to −2.87)

NE

1

−2.16 (−9.30 to 4.98)

NE

0

NE

0.13

Length of hospital stay (days)

6

−0.59 (−0.99 to −0.18)

(27%)

12

−0.15 (−0.58 to 0.28)

(77%)

14

−0.48 (−0.84 to −0.11)

(69%)

0.32

Length of hospital stay (outpatient surgery, mins)

0

NE

3

−10.81 (−36.93 to 15.31)

0

NE

NE

Surgical complications ‐ anastomotic leak

No subgroup analysis performed (I2 < 30%)

Surgical complications ‐ bleeding

No subgroup analysis performed (I2 < 30%)

Surgical complications ‐ postoperative infection

No subgroup analysis performed (I2 < 30%)

Patient satisfaction

No subgroup analysis performed (I2 < 30%)

Postoperative nausea, early (PACU)

No subgroup analysis performed (I2 < 30%)

Postoperative nausea, overall (0 to 24 hrs, to 48 hrs, to 72 hrs)

No subgroup analysis performed (I2 < 30%)

Postoperative vomiting, early (PACU)

No subgroup analysis performed (I2 < 30%)

Postoperative vomiting, overall (0 to 24 hrs, to 48 hrs, to 72 hrs)

No subgroup analysis performed (I2 < 30%)

Intraoperative opioid consumption (MEQ, mg)

7

−1.93 (‐4.61 to 0.75)

(78%)

3

−0.71 (−7.95 to 6.53)

(93%)

8

−2.03 (−4.14 to 0.07)

(40%)

0.94

Intraoperative opioid consumption with remifentanil (MEQ, mg)

No subgroup analysis performed (I2 < 30%)

Postoperative opioid consumption, PACU (MEQ, mg)

5

−3.03 (−4.82 to −1.23)

(0%)

7

−3.84 (−4.57 to −3.11)

(0%)

9

−2.66 (−4.19 to −1.13)

(61%)

0.33

Postoperative opioid consumption, overall (MEQ, mg)

11

−3.56 (−6.76 to −0.35)

(40%)

16

−4.85 (−7.46 to −2.23)

(77%)

13

−5.54 (−9.35 to −1.72)

(77%)

0.71

Acronyms and abbrviations used in this table:

hrs = hours, MEQ = morphine equivalent dose, mins = minutes, NE = not estimable, PACU =post anaesthesia care unit

Figuras y tablas -
Table 3. Subgroup analyses ‐ type of surgery
Table 4. Subgroup analyses ‐ time and dosing of lidocaine

Main meta‐analyses

< 2 mg/kg/hr until end of surgery to PACU

≥ 2 mg/kg/hr until end of surgery to PACU

< 2 mg/kg/hr
for ≥ 24 hrs

≥ 2 mg/kg/hr
for ≥ 24 hrs

Test for subgroup
difference (P)

Outcome

n

Effect estimate

(I2)

n

Effect estimate

(I2)

n

Effect estimate

(I2)

n

Effect estimate

(I2)

Pain score, rest, 'early time points' (1 hr to 4 hrs, PACU)

8

−0.36 (−0.70 to −0.02)

(67%)

21

−0.54 (−0.82 to −0.27)

(82%)

0.42

Pain score, rest, 'intermediate time points' (24 hrs)

No subgroup analysis performed (I2 < 30%)

Pain score, rest, 'late time points' (48 hrs)

5

−0.15 (−0.39 to 0.09)

(0%)

13

−0.18 (−0.34 to −0.02) (12%)

5

0.03 (−0.45 to 0.51) (78%)

1

0.11 (−0.39 to 0.61)

NE

0.66

Postoperative ileus (dichotomous)

No subgroup analysis performed (I2 < 30%)

Time to first defaecation/bowel movement (hrs)

4

−7.06 (−11.37 to −2.75) (10%)

3

−7.27 (−13.54 to −1.00) (0%)

4

−6.97 (−20.09 to 6.16)

(86%)

1

−20.00 (−50.62 to 10.62)

NE

0.62

Time to first flatus (hrs)

4

−5.72 (−9.58 to −1.87) (28%)

4

−3.63 (−6.07 to −1.20) (64%)

4

−0.43 (−9.46 to 8.61) (84%)

1

−6.50 (−17.05 to 4.05)

NE

0.65

Time to first bowel sounds (hrs)

1

−10.00 (−17.13 to −2.87)

NE

1

−2.16 (−9.30 to 4.98)

NE

0

NE

0

NE

0.13

Length of hospital stay (days)

7

−0.51 (−0.84 to −0.19) (2%)

16

−0.26 (‐0.50 to −0.03) (58%)

7

−0.25 (−1.04 to 0.54) (83%)

2

−1.29 (−4.47 to 1.89)

(94%)

0.59

Length of hospital stay (outpatient surgery, mins)

1

−44.00 (−75.57 to −12.43)

NE

2

−2.97 (−11.33 to 5.39)

(0%)

0

NE

0

NE

0.01

Surgical complications ‐ anastomotic leak

No subgroup analysis performed (I2 < 30%)

Surgical complications ‐ bleeding

No subgroup analysis performed (I2 < 30%)

Surgical complications ‐ postoperative infection

No subgroup analysis performed (I2 < 30%)

Patient satisfaction

No subgroup analysis performed (I2 < 30%)

Postoperative nausea, early (PACU)

No subgroup analysis performed (I2 < 30%)

Postoperative nausea, overall (0 to 24 hrs, to 48 hrs, to 72 hrs)

No subgroup analysis performed (I2 < 30%)

Postoperative vomiting, early (PACU)

No subgroup analysis performed (I2 < 30%)

Postoperative vomiting, overall (0 to 24 hrs, to 48 hrs, to 72 hrs)

No subgroup analysis performed (I2 < 30%)

Intraoperative opioid consumption (MEQ, mg)

7

−0.68 (−3.51 to 2.15) (80%)

11

−3.11 (−5.74 to −0.47) (79%)

0.22

Intraoperative opioid consumption with remifentanil (MEQ, mg)

No subgroup analysis performed (I2 < 30%)

Postoperative opioid consumption, PACU (MEQ, mg)

6

−3.55 (−5.43 to −1.67) (63%)

15

−3.02 (−3.86 to −2.18) (30%)

0.61

Postoperative opioid consumption, overall (MEQ, mg)

8

−2.33 (−5.05 to −0.13) (59%)

21

−7.41 (−10.91 to −3.91) (76%)

8

−2.88 (−6.25 to 0.49) (72%)

3

−3.90 (−10.18 to 2.38)

(18%)

0.14

Acronyms and abbreviations used in this table:

hrs = hours, MEQ = morphine equivalent dose, mins = minutes, NE = not estimable, PACU =post anaesthesia care unit

Figuras y tablas -
Table 4. Subgroup analyses ‐ time and dosing of lidocaine
Table 5. Subgroup analyses with independent tau2 (type of surgery)

Outcome

Meta‐regression model

(random‐effects model, tau2 estimator: REML)

Open abdominal surgery

Laparoscopic surgery

Other surgery

Test of moderators

(P)

Likelihood ratio test

(P)

n

Effect estimate

(tau2)

n

Effect estimate

(tau2)

n

Effect estimate

(tau2)

Pain score, rest, 'early time points' (1 hr to 4 hrs, PACU)

Individual tau2

8

−0.55 (−0.83 to −0.27)

(0.08)

10

−0.78(−1.35 to −0.21)

(0.76)

11

−0.21 to (−0.44 0.03)

(0.09)

0.07

0.017

Pain score, rest, 'late time points' (48 hrs)

Individual tau2

7

0.03 (−0.17 to 0.23)

(0.00)

7

−0.03 (−0.73 to 0.12)

(0.24)

10

−0.10 (−0.27 to 0.08)

(0.02)

0.34

0.049

Time to first defaecation/bowel movement (hrs)

Individual tau2

6

−8.16 (−12.44 to −3.87)

(5.94)

5

−6.22 (−18.42 to 5.98)

(127.06)

1

−6.10 (−24.49 to 12.29)

(0.00)

0.94

0.097

Time to first flatus (hrs)

Individual tau2

6

−4.36 (−6.99 to −1.72)

(0.00)

5

−2.55 (−9.31 to 4.21)

(47.86)

2

−2.15 (−3.56 to −0.74)

(0.00)

0.35

0.234

Length of hospital stay (days)

Individual tau2

6

−0.62 (−1.07 to −0.18)

(0.11)

12

−0.16 (−0.66 to 0.33)

(0.43)

14

−0.47 (−0.83 to −0.12)

(0.18)

0.39

0.592

Intraoperative opioid consumption (MEQ, mg)

Individual tau2

7

−2.00 (−4.30 to 0.30)

(4.34)

3

0.04 (−11.99 to 12.08)

(107.18)

8

−1.86 (−3.34 to −0.38)

(0.55)

0.95

0.027

Postoperative opioid consumption, PACU (MEQ, mg)

Individual tau2

5

−3.03 (−4.82 to −1.23)

(0.00)

7

−3.84 (−4.57 to −3.11)

(0.00)

9

−2.71 (−4.32 to −1.09)

(3.45)

0.37

0.211

Postoperative opioid consumption, overall (MEQ, mg)

Individual tau2

11

−3.43 (−6.01 to −0.85)

(3.08)

16

−5.78 (−9.33 to −2.23)

(32.11)

13

−6.42 (−11.60 to −1.24)

(50.54)

0.43

0.285

Subgroup‐analyses are based on multivariate meta‐analysis models (method: REML, R package 'metafor'; Viechtbauer 2010). We tested for subgroup differences (test of moderators). Tests on subgroup differences are based on the assumption that the tau2 (between‐study heterogeneity) varies across the subgroups. We estimated the tau2s and tested if they have a common value (likelihood ratio test). We rejected the null hypothesis for P < 0.05.

Acronyms and abbreviations used in this table:

MEQ = morphine equivalent dose, PACU =post anaesthesia care unit, REML = restricted maximum likelihood approach

Figuras y tablas -
Table 5. Subgroup analyses with independent tau2 (type of surgery)
Table 6. Sensitivity analyses ‐ risk of bias (selection bias)

All studies

Without high/unclear risk of bias studies (selection bias)

Outcome

Statistical method

Studies

Effect estimate

Studies

Effect estimate

Pain score, rest, 'early time points' (1 hr to 4 hrs, PACU)

SMD (IV, Random, 95% CI)

29

−0.50 (−0.72 to −0.28)

6

−0.23 (−0.51 to 0.05)

Pain score, rest, 'intermediate time points' (24 hrs)

SMD (IV, Random, 95% CI)

33

−0.14 (−0.25 to −0.04)

6

0.09 (−0.13 to 0.30)

Pain score, rest, 'late time points' (48 hrs)

SMD (IV, Random, 95% CI)

24

−0.11 (−0.25 to 0.04)

3

0.03 (−0.24 to 0.29)

Postoperative ileus (dichotomous)

RR (MH, Random, 95% CI)

4

0.37 (0.15 to 0.87)

0

Not estimable

Time to first defaecation/bowel movement (hrs)

MD (IV, Random, 95% CI)

12

−7.92 (−12.71 to −3.13)

2

−6.18 (−11.19 to −1.18)

Time to first flatus (hrs)

MD (IV, Random, 95% CI)

13

−4.09 (−6.30 to −1.87)

2

−3.27 (−6.33 to −0.21)

Time to first bowel sounds (hrs)

MD (IV, Random, 95% CI)

2

−6.08 (−13.77 to 1.60)

0

Not estimable

Length of hospital stay (days)

MD (IV, Random, 95% CI)

32

−0.37 (−0.60 to −0.15)

3

−0.13 (−0.70 to 0.44)

Length of hospital stay (outpatient surgery, mins)

MD (IV, Random, 95% CI)

3

−10.81 (−36.93 to 15.31)

3

−10.81 (−36.93 to 15.31)

Surgical complications ‐ anastomotic leak

RR (MH, Random, 95% CI)

3

0.61 (0.08 to 4.80)

0

Not estimable

Surgical complications ‐ bleeding

RR (MH, Random, 95% CI)

3

1.79 (0.41 to 7.89)

0

Not estimable

Surgical complications ‐ postoperative infection

RR (MH, Random, 95% CI)

5

1.64 (0.41 to 6.52)

0

Not estimable

Patient satisfaction

MD (IV, Random, 95% CI)

6

0.76 (0.46 to 1.06)

2

0.59 (−0.09 to 1.26)

Postoperative nausea, early (PACU)

RR (MH, Random, 95% CI)

8

0.72 (0.53 to 0.98)

3

0.64 (0.30 to 1.37)

Postoperative nausea, overall (0 to 24 hrs, to 48 hrs, to 72 hrs)

RR (MH, Random, 95% CI)

35

0.78 (0.67 to 0.91)

8

0.99 (0.69 to 1.42)

Postoperative vomiting, early (PACU)

RR (MH, Random, 95% CI)

4

0.49 (0.16 to 1.48)

2

0.39 (0.11 to 1.38)

Postoperative vomiting, overall (0 to 24 hrs, to 48 hrs, to 72 hrs)

RR (MH, Random, 95% CI)

19

0.83 (0.63 to 1.08)

3

1.33 (0.50 to 3.53)

Intraoperative opioid consumption (MEQ, mg)

MD (IV, Random, 95% CI)

18

−2.14 (−3.87 to −0.40)

3

−3.28 (−6.56 to −0.00)

Intraoperative opioid consumption with remifentanil (MEQ, mg)

MD (IV, Random, 95% CI)

6

−14.17 (−35.27 to 6.92)

2

−9.53 (−59.18 to 40.12)

Postoperative opioid consumption, PACU (MEQ, mg)

MD (IV, Random, 95% CI)

21

−3.10 (−3.87 to −2.32)

6

−2.69 (−4.13 to −1.24)

Postoperative opioid consumption, overall (MEQ, mg)

MD (IV, Random, 95% CI)

40

−4.52 (−6.25 to −2.79)

10

−2.74 (−5.60 to 0.13)

Acronyms and abbreviations used in this table:

CI = confidence interval, IV = inverse variance, MD = mean difference, MEQ = morphine equivalent dose, MH = Mantel Haenszel, PACU = postanaesthesia care unit, RR = risk ratio, SMD = standardized mean difference

Figuras y tablas -
Table 6. Sensitivity analyses ‐ risk of bias (selection bias)
Table 7. Sensitivity analyses ‐ risk of bias (blinding)

All studies

Without high/unclear risk of bias studies (blinding)

Outcome

Statistical method

Studies

Effect estimate

Studies

Effect estimate

Pain score, rest, 'early time points' (1 hr to 4 hrs, PACU)

SMD (IV, Random, 95% CI)

29

−0.50 (−0.72 to −0.28)

21

−0.62 (−0.88 to −0.35)

Pain score, rest, 'intermediate time points' (24 hrs)

SMD (IV, Random, 95% CI)

33

−0.14 (−0.25 to −0.04)

22

−0.19 (−0.33 to −0.05)

Pain score, rest, 'late time points' (48 hrs)

SMD (IV, Random, 95% CI)

24

−0.11 (−0.25 to 0.04)

19

−0.17 (−0.31 to −0.04)

Postoperative ileus (dichotomous)

RR (MH, Random, 95% CI)

4

0.37 (0.15 to 0.87)

2

0.55 (0.16 to 1.88)

Time to first defaecation/bowel movement (hrs)

MD (IV, Random, 95% CI)

12

−7.92 (−12.71 to −3.13)

5

−8.87 (−20.51 to 2.78)

Time to first flatus (hrs)

MD (IV, Random, 95% CI)

13

−4.09 (−6.30 to −1.87)

6

−3.63 (−6.59 to −0.67)

Time to first bowel sounds (hrs)

MD (IV, Random, 95% CI)

2

−6.08 (−13.77 to 1.60)

1

−2.16 (−9.30 to 4.98)

Length of hospital stay (days)

MD (IV, Random, 95% CI)

32

−0.37 (−0.60 to −0.15)

19

−0.32 (−0.59 to −0.04)

Length of hospital stay (outpatient surgery, mins)

MD (IV, Random, 95% CI)

3

−10.81 (−36.93 to 15.31)

1

−4.00 (−12.64 to 4.64)

Surgical complications ‐ anastomotic leak

RR (MH, Random, 95% CI)

3

0.61 (0.08 to 4.80)

1

1.00 (0.07 to 15.26)

Surgical complications ‐ bleeding

RR (MH, Random, 95% CI)

3

1.79 (0.41 to 7.89)

1

Not estimable

Surgical complications ‐ postoperative infection

RR (MH, Random, 95% CI)

5

1.64 (0.41 to 6.52)

3

0.69 (0.11 to 4.33)

Patient satisfaction

MD (IV, Random, 95% CI)

6

0.76 (0.46 to 1.06)

5

1.00 (0.63 to 1.37)

Postoperative nausea, early (PACU)

RR (MH, Random, 95% CI)

8

0.72 (0.53 to 0.98)

6

0.75 (0.53 to 1.05)

Postoperative nausea, overall (0 to 24 hrs, to 48 hrs, to 72 hrs)

RR (MH, Random, 95% CI)

35

0.78 (0.67 to 0.91)

23

0.78 (0.66 to 0.93)

Postoperative vomiting, early (PACU)

RR (MH, Random, 95% CI)

4

0.49 (0.16 to 1.48)

3

0.52 (0.16 to 1.68)

Postoperative vomiting, overall (0 to 24 hrs, to 48 hrs, to 72 hrs)

RR (MH, Random, 95% CI)

19

0.83 (0.63 to 1.08)

15

0.70 (0.50 to 0.96)

Intraoperative opioid consumption (MEQ, mg)

MD (IV, Random, 95% CI)

18

−2.14 (−3.87 to −0.40)

13

−1.86 (−3.74 to 0.02)

Intraoperative opioid consumption with remifentanil (MEQ, mg)

MD (IV, Random, 95% CI)

6

−14.17 (−35.27 to 6.92)

5

−16.08 (−41.41 to 9.25)

Postoperative opioid consumption, PACU (MEQ, mg)

MD (IV, Random, 95% CI)

21

−3.10 (−3.87 to −2.32)

18

−2.93 (−3.75 to −2.11)

Postoperative opioid consumption, overall (MEQ, mg)

MD (IV, Random, 95% CI)

40

−4.52 (−6.25 to −2.79)

24

−7.29 (−10.38 to −4.19)

Acronyms and abbreviations used in this table:

CI = confidence interval, IV = inverse variance, MD = mean difference, MEQ = morphine equivalent dose, MH = Mantel Haenszel, PACU = postanaesthesia care unit, RR = risk ratio, SMD = standardized mean difference

Figuras y tablas -
Table 7. Sensitivity analyses ‐ risk of bias (blinding)
Table 8. Sensitivity analyses ‐ random‐effects versus fixed‐effect model

Random‐effects model

Fixed‐effect model

Outcome

Statistical method

Studies

Effect estimate

Studies

Effect estimate

Pain score, rest, 'early time points' (1 hr to 4 hrs, PACU)

SMD (IV, Random, 95% CI)

29

−0.50 (−0.72 to −0.28)

29

−0.40 (−0.50 to −0.30)

Pain score, rest, 'intermediate time points' (24 hrs)

SMD (IV, Random, 95% CI)

33

−0.14 (−0.25 to −0.04)

33

−0.13 (−0.22 to −0.04)

Pain score, rest, 'late time points' (48 hrs)

SMD (IV, Random, 95% CI)

24

−0.11 (−0.25 to 0.04)

24

−0.09 (−0.19 to 0.02)

Postoperative ileus (dichotomous)

RR (MH, Random, 95% CI)

4

0.37 (0.15 to 0.87)

4

0.35 (0.15 to 0.82)

Time to first defaecation/bowel movement (hrs)

MD (IV, Random, 95% CI)

12

−7.92 (−12.71 to −3.13)

12

−6.01 (−8.53 to −3.49)

Time to first flatus (hrs)

MD (IV, Random, 95% CI)

13

−4.09 (−6.30 to −1.87)

13

−3.63 (−4.59 to −2.68)

Time to first bowel sounds (hrs)

MD (IV, Random, 95% CI)

2

−6.08 (−13.77 to 1.60)

2

−6.09 (−11.13 to −1.04)

Length of hospital stay (days)

MD (IV, Random, 95% CI)

32

−0.37 (−0.60 to −0.15)

32

−0.21 (−0.30 to −0.12)

Length of hospital stay (outpatient surgery, mins)

MD (IV, Random, 95% CI)

3

−10.81 (−36.93 to 15.31)

3

−5.66 (−13.74 to 2.43)

Surgical complications ‐ anastomotic leak

RR (MH, Random, 95% CI)

3

0.61 (0.08 to 4.80)

3

0.58 (0.08 to 4.24)

Surgical complications ‐ bleeding

RR (MH, Random, 95% CI)

3

1.79 (0.41 to 7.89)

3

1.86 (0.43 to 8.05)

Surgical complications ‐ postoperative infection

RR (MH, Random, 95% CI)

5

1.64 (0.41 to 6.52)

5

1.69 (0.53 to 5.33)

Patient satisfaction

MD (IV, Random, 95% CI)

6

0.76 (0.46 to 1.06)

6

0.76 (0.46 to 1.06)

Postoperative nausea, early (PACU)

RR (MH, Random, 95% CI)

8

0.72 (0.53 to 0.98)

8

0.72 (0.53 to 0.99)

Postoperative nausea, overall (0 to 24 hrs, to 48 hrs, to 72 hrs)

RR (MH, Random, 95% CI)

35

0.78 (0.67 to 0.91)

35

0.77 (0.68 to 0.88)

Postoperative vomiting, early (PACU)

RR (MH, Random, 95% CI)

4

0.49 (0.16 to 1.48)

4

0.51 (0.18 to 1.44)

Postoperative vomiting, overall (0 to 24 hrs, to 48 hrs, to 72 hrs)

RR (MH, Random, 95% CI)

19

0.83 (0.63 to 1.08)

19

0.78 (0.60 to 1.01)

Intraoperative opioid consumption (MEQ, mg)

MD (IV, Random, 95% CI)

18

−2.14 (−3.87 to −0.40)

18

−1.05 (−1.47 to −0.62)

Intraoperative opioid consumption with remifentanil (MEQ, mg)

MD (IV, Random, 95% CI)

6

−14.17(−35.27 to 6.92)

6

−13.68 (−33.53 to 6.17)

Postoperative opioid consumption, PACU (MEQ, mg)

MD (IV, Random, 95% CI)

21

−3.10 (−3.87 to −2.32)

21

−3.14 (−3.67 to −2.61)

Postoperative opioid consumption, overall (MEQ, mg)

MD (IV, Random, 95% CI)

40

−4.52 (−6.25 to −2.79)

40

−1.52 (−2.14 to −0.90)

Acronyms and abbreviations used in this table:

CI = confidence interval, hrs = hours, IV = inverse variance, MD = mean difference, MEQ = morphine equivalent dose, MH = Mantel Haenszel, PACU = postanaesthesia care unit, mins = minutes, RR = risk ratio, SMD = standardized mean difference

Figuras y tablas -
Table 8. Sensitivity analyses ‐ random‐effects versus fixed‐effect model
Table 9. Sensitivity analyses ‐ with studies with 'suspected variance reporting'

Without suspicious studies

With suspicious studies

Outcome

Statistical method

Studies

Effect estimate

Studies

Effect estimate

Pain score, rest, 'early time points' (1 hr to 4 hrs, PACU)

SMD (IV, Random, 95% CI)

29

−0.50 (−0.72 to −0.28)

37

−0.88 (−1.18 to −0.57)

Pain score, rest, 'intermediate time points' (24 hrs)

SMD (IV, Random, 95% CI)

33

−0.14 (−0.25 to −0.04)

41

−0.29 (−0.44 to −0.15)

Pain score, rest, 'late time points' (48 hrs)

SMD (IV, Random, 95% CI)

24

−0.11 (−0.25 to 0.04)

30

−0.22 (−0.40 to −0.03)

Time to first defaecation/bowel movement (hrs)

MD (IV, Random, 95% CI)

12

−7.92 (−12.71 to −3.13)

14

−7.09 (−10.06 to −4.11)

Time to first flatus (hrs)

MD (IV, Random, 95% CI)

13

−4.09 (−6.30 to −1.87)

16

−5.02 (−7.73 to −2.31)

Time to first bowel sounds (hrs)

MD (IV, Random, 95% CI)

2

−6.08 (−13.77 to 1.60)

4

−4.28 (−10.32 to 1.76)

Postoperative opioid consumption, PACU (MEQ, mg)

MD (IV, Random, 95% CI)

21

−3.10 (−3.87 to −2.32)

25

−3.51 (−4.88 to −2.15)

Postoperative opioid consumption, overall (MEQ, mg)

MD (IV, Random, 95% CI)

40

−4.52 (−6.25 to −2.79)

43

−4.81 (−6.55 to −3.07)

Acronyms and abbreviations used in this table:

CI = confidence interval, hrs = hours, IV = inverse variance, MD = mean difference, MEQ = morphine equivalent dose, PACU = postanaesthesia care unit, SMD = standardized mean difference

Figuras y tablas -
Table 9. Sensitivity analyses ‐ with studies with 'suspected variance reporting'
Table 10. Study drug administration

Study ID

Surgical procedure

Start infusion

End infusion

Duration of infusion

Bolus dose

Infusion dose

Total dose

Ahn 2015

Laparoscopic colectomy

2 mins before intubation

End of the operaton

216.60 mins (surgery)

1.5 mg/kg

2 mg/kg/hr

NA

Baral 2010

Abdominal surgery

30 mins before skin incision

1 hr after the end of surgery

157.80 min (infusion)

1.5 mg/kg

1.5 mg/kg/hr

NA

Bryson 2010

Abdominal hysterectomy

Prior to induction

Skin closure

105.0 mins (anaesthesia)

1.5 mg/kg

3 mg/kg/hr

NA

Cassuto 1985

Cholecystectomy

30 mins before skin incision

24 hrs postop

105 mins (surgery) + 30 mins (prior) + 24 hrs (postop)

100 mg

2 mg/min

NA

Chen 2015

Spine surgery

After induction of anaesthesia

End of surgery

129.2 mins (surgery)

1 mg/kg

1.5 mg/kg/hr

NA

Choi SJ 2012

Breast plastic surgeries

30 mins before skin incision

Skin closure

295 mins (surgery) + 30 mins (prior)

1.5 mg/kg

1.5 mg/kg/hr

NA

Choi GJ 2016

Elective total thyroidectomy

Prior to anaesthesia

End of surgery

135 mins (anaesthesia)

1.5 mg/kg

2 mg/kg/hr

NA

Choi KW 2016

Thyroidectomy

Immediately after induction

Extubation

148.9 mins (anaesthesia)

2 mg/kg

3 mg/kg/hr

NA

Cui 2010

Thoracic surgery

At induction

Skin closure

244 mins (anaesthesia)

No bolus

33 µg/kg/mins

NA

Dale 2016

Laparoscopic fundoplication

At induction

24 hrs after start of continuous infusion

24 hrs

1 mg/kg

2 mg/kg/hr

NA

De Oliveira 2012

Outpatient laparoscopic surgery

Prior to induction

End of the surgical procedure

105.5 mins (time of induction to skin incision)

1.5 mg/kg

2 mg/kg/hr

NA

De Oliveira 2014

Laparoscopic
bariatric surgery

Prior to induction

End of the surgical procedure

144 mins (surgery)

1.5 mg/kg

2 mg/kg/hr

NA

Dewinter 2016

Laparoscopic sterilisation in women

At induction

30 mins after arrival at PACU

77 mins

1.5 mg/kg

1.5 mg/kg/hr

240 mg

El‐Tahan 2009

Caesarean delivery

30 mins before induction

60 mins after skin closure

43.2 mins (anaesthesia) + 60 mins (postop)

1.5 mg/kg

1.5 mg/kg/hr

NA

Farag 2013

Spine surgery

At induction

Discharge from the PACU or a maximum of 8 hrs

8.5 hrs

No bolus

2 mg/kg/hr

NA

Grady 2012

Abdominal hysterectomy

At induction

24 hours postop

NA

1.5 mg/kg

2 mg/kg/hr

NA

Grigoras 2012

Surgery for breast cancer

Prior to induction

60 mins after skin closure

60.6 mins (surgery) + 60 mins (postop)

1.5 mg/kg

1.5 mg/kg/hr

328.1 mg

Groudine 1998

Radical retropubic prostatectomy

Prior to induction

60 mins after skin closure

NA

1.5 mg/kg

1.5 mg/kg/hr

NA

Herroeder 2007

Colorectal surgery

Prior to induction

4 hours
postop

194.3 mins (surgery) + NA (induction to skin incision) + 4 hrs (postop)

1.5 mg/kg

2 mg/mins

NA

Insler 1995

CABG

After induction of anaesthesia and before surgical incision

Up to 48 hours in the ICU
unless discharged earlier

NA

1.5 mg/kg

30 μg/kg/min

NA

Ismail 2008

Lumbar discectomy

30 mins before induction

Until 10 mins after extubation

NA

1.5 mg/kg

1.5 mg/kg

NA

Jain 2015

Laparoscopic cholecystectomy

10 mins prior to induction

End of first postop hr, max. 180 mins

NA

1.5 mg/kg

1.5 mg/kg/hr

NA

Kaba 2007

Laparoscopic colectomy

At induction

24 hrs postop

169 mins (anaesthesia) + 24 hrs (postop)

1.5 mg/kg

2 mg/kg/hr intraop and 1.33 mg/kg/h for 24 hrs
postop

NA

Kang 2011

Inguinal herniorrhaphy

2 mins before induction

End of the surgical procedure

66.03 mins (anaesthesia) + 2 mins (before induction)

1.5 mg/kg

2 mg/kg/hr

NA

Kasten 1986

CABG

2 mins before induction

NA

NA

3 mg/kg

0.05 mg/kg/min

NA

Kim TH 2011

Laparoscopic appendectomy

2 mins before induction

End of the surgical procedure

70.0 mins (anaesthesia) or 55.0 (surgery) + 2 mins (before induction)

1.5 mg/kg

2 mg/kg/hr

240.3 mg

Kim TH 2013

Laparoscopic gastrectomy

Preop

End of the surgical procedure

324 mins (anaesthesia) or 282.06 mins (surgery)

1.5 mg/kg

2 mg/kg/hr

NA

Kim HJ 2014

Coronary artery bypass graft

Before induction

24 hrs after end of surgery

339 mins (anaesthesia) + 24 hrs

1.5 mg/kg

2 mg/kg/hr

3917 mg

Kim HO 2014

Laparoscopic colectomy

Prior to incision

After 24 hrs

24 hrs

1 mg/kg

1 mg/kg/hr

NA

Kim KT 2014

Elective one‐level laminectomy and discectomy

Preop

End of surgery

110 min (surgery)

1.5 mg/kg

2 mg/kg/hr

NA

Koppert 2004

Major abdominal surgery

30 mins before skin incision

1 hr after the end of surgery

6.2 hrs (infusion)

1.5 mg/kg

5 mg/kg/hr

NA

Kuo 2006

Surgery for colon cancer

30 mins before surgery

End of the surgical procedure

157.8 mins (surgery) + 30 min (before surgery)

2 mg/kg

3 mg/kg/hr

NA

Lauwick 2008

Outpatient laparoscopic cholecystectomy

At induction

End of the surgical procedure

60 mins (surgery) + NA (induction to skin incision)

1.5 mg/kg

2 mg/kg/hr

NA

Lauwick 2009

Laparoscopic prostatectomy

At induction

End of the surgical procedure

262.5 mins (surgery) + NA (induction to skin incision)

1.5 mg/kg

2 mg/kg/hr

NA

Lee 2011

Off‐pump coronary artery bypass graft surgery

At induction

End of the surgical procedure

208.9 mins (surgery) + NA (induction to skin incision)

1.5 mg/kg

2 mg/kg/hr

NA

Maquoi 2016

Prostatectomy

Before induction

24 hrs postop

173 mins (anaesthesia) + 24 hrs

1.5 mg/kg

2 mg/kg/hr during surgery,
then 1.33 mg/kg/hr

NA

Martin 2008

Hip
arthroplasty

30 mins before skin incision

1 hr after the end of surgery

NA

1.5 mg/kg

1.5 mg/kg/hr

NA

Mathew 2009

Cardiac surgery

After induction

48 hrs postop

NA

1 mg/kg

4 mg/min for 1 hr,
2 mg/min for the second hr,
1 mg/min for the rest

NA

McKay 2009

Outpatient surgery

After induction

1 hr after arrival in the PACU

NA

1.5 mg/kg

2 mg/kg/hr

517 mg

Mitchell 1999

Cardiac surgery

At induction

48 hrs postop

NA

1 mg/kg

240 mg over the first hr and
120 mg over the second hr,
and then 60 mg/h thereafter if the patient
was receiving lidocaine

NA

Mitchell 2009

Cardiac surgery

At induction

Total
of 12 hours

NA

1 mg/kg

2 mg/min for 2 hrs, and 1 mg/min thereafter

NA

Oliveira 2015

Hysterectomy

At induction

End of surgery

145.1 mins (anaesthesia)

No bolus

2 mg/kg/hr

NA

Omar 2013

Functional endoscopic sinus surgery

After induction

End of the surgical procedure

87 mins (anaesthesia) or 62 mins (surgery)

1.5 mg/kg

1.5 mg/kg/hr

NA

Ortiz 2016

Laparoscopic cholecystectomy

Before incision

1 hr after end of surgery

105.23 mins (surgery) + 1 hr

1.5 mg/kg

3 mg/kg/hr

NA

Peng 2016

Supratentorial tumour surgery

After induction

End of surgery

254 mins (surgery)

1.5 mg/kg

2 mg/kg/hr

NA

Rimbäck 1990

Cholecystectomy

Prior to induction

24 hrs postop

109 mins (surgery) + NA (induction to skin incision) + 24 hrs (postop)

100 mg

3 mg/min

NA

Saadawy 2010

Laparoscopic cholecystectomy

Prior to induction

End of the surgical procedure

80.3 mins (surgery) + NA (induction to skin incision)

2 mg/kg

2 mg/kg/hr

NA

Samimi 2015

Abdominal hysterectomy

30 mins before incision

1 hr after surgery

30 mins + 95 min (surgery) + 60 mins

1.5 mg/kg

2 mg/kg/hr

NA

Slovack 2015

VATS

At induction

End of the surgical procedure

NA

1.5 mg/kg

3 mg/min if the
patient’s total body weight was more than 70 kg or 2 mg/min if weight was less than 70
kg

239.6 mg

Soltani 2013

Ophthalmologic surgeries

NA

Intraoperatively

No bolus

2.5 mg/kg/hr

Sridhar 2015

Open abdominal surgery

Time of intubation

1 hr after surgery

145.8 mins (surgery) + 60 min

1.5 mg/kg

1.5 mg/kg/hr

NA

Staikou 2014

Large bowel surgery

Before induction

Before skin suturing

122 mins

1.5 mg/kg

2 mg/kg/hr

NA

Striebel 1992

Tonsillectomy

30 mins before skin incision

24 hrs

57 mins (surgery) + 30 min (before skin incision) + 24 hrs (postop)

1.5 mg/kg

2 mg/kg/hr over 6 hrs and
0.5 mg/kg/hr for another 18 hrs

NA

Swenson 2010

Colon resection

Prior to induction

Until the day after return of bowel function or fifth postop
day

69 hrs 54 mins (infusion)

No bolus

11 patients: 2 mg/min in patients < 70 kg,
3 mg/min in patients > 70 kg,
and 11 patients: 1 mg/min in patients < 70 kg,
2 mg/min in patients > 70 kg

NA

Terkawi 2014

Breast cancer surgery

Before induction

2 hrs after arrival in PACU or at discharge from PACU

85 mins

1.5 mg/kg, max. 150 mg

2 mg/kg/h, max 200 mg/hr

NA

Tikuisis 2014

Laparoscopic colon resection

Prior to induction

24 hrs postop

115 mins (anaesthesia) + 24 hrs (postop)

1.5 mg/kg

2 mg/kg/hr during surgery, 1 mg/
kg/hr for 24 hrs

NA

Wallin 1987

Cholecystectomy

30 mins before skin incision

24 hrs postop

110 mins (surgery) + 30 min (before skin incision) + 24 hrs

100 mg

2 mg/min

NA

Wang 2002

CABG

At the opening of the
pericardium

End of the surgical procedure

NA

1.5 mg/kg,

second dose (4 mg/kg) was administered to the priming solution
of CPB

4 mg/min

NA

Wang 2015

Hysterectomy

10 mins prior to induction

Discharge from the operating room

152.3 (anaesthesia) + 10 mins

1.5 mg/kg

1.5 mg/kg/hr

NA

Weinberg 2016

Radical retropubic prostatectomy

Before induction

End of surgery

NA

1.5 mg/kg

1.5 mg/kg/hr

NA

Wongyingsinn 2011

Laparoscopic colorectal surgery

Prior to induction

48 hrs postop

220 mins (surgery) + NA (induction to skin incision) + 48 (postop)

1.5 mg/kg, max: 100 mg

2 mg/kg/hr during surgery,
1 mg/kg/hr for 48 hrs

NA

Wu 2005

Laparoscopic cholecystectomy

30 mins before skin incision

End of the surgical procedure

81.4 mins (surgery) + 30 mins (before incision)

No bolus

3 mg/kg/hr

NA

Wuethrich 2012

Laparoscopic transperitoneal renal surgery

At induction

24 hrs postop

293 mins (anaesthesia) + 24 hrs (postop)

1.5 mg/kg

2 mg/kg/hr during surgery,
1.3 mg/kg/hr for 24 hrs

NA

Xu 2017

Abdominal hysterectomy

10 mins before induction

Wound closure

118.7 mins (anaesthesia)

1.5 mg/kg

1.5 mg/kg/hr

185.7 mg

Yang 2014

Laparoscopic
cholecystectomy

2 mins before induction

End of the surgical procedure

65 mins (anaesthesia) + 2 mins (before induction)

1.5 mg/kg

2 mg/kg/hr

228.71 mg

Yardeni 2009

Transabdominal hysterectomy

20 mins before skin incision

End of the surgical procedure

109 mins (surgery) + 20 mins (before skin incision)

2 mg/kg

1.5 mg/kg/hr

NA

Yon 2014

Subtotal gastrectomy

Preop (protocol: 2 mins before intubation)

End of surgery

271.27 mins (surgery)

1.5 mg/kg

2 mg/kg/hr

NA

Zengin 2015

Laparotomy

At induction

Wound closure

114.1 mins (surgery)

1.0 mg/kg

2 mg/kg/hr

NA

Acronyms and abbreviations used in this table:

CABG = coronary artery bypass graft , CPB = cardiopulmonary bypass, hr = hour, ICU = intensive care unit, min = minute, NA = not available, preop = preoperatively, postop =postoperatively, VATS = Video‐assisted thoracoscopic surgery

Figuras y tablas -
Table 10. Study drug administration
Table 11. Heterogeneity/prediction intervals/publication bias

Outcome

Statistical method

Studies

Effect estimate (95% CI)

Prediction interval (95% PI)

I2

Adjusted effect estimate (trim and fill), (number of added studies)

Pain score, rest, 'early time points' (1 hr to 4 hrs, PACU)

SMD (IV, Random)

29

−0.50 (−0.72 to −0.28)

(−1.61 to 0.62)

79%

−0.26 (−0.52 to −0.004), (6)*

Pain score, rest, 'intermediate time points' (24 hrs)

SMD (IV, Random)

33

−0.14 (−0.25 to −0.04)

(−0.44 to 0.16)

20%

0.007 (−0.12 to 0.13), (11)*

Pain score, rest, 'late time points' 48 hrs)

SMD (IV, Random)

24

−0.11 (−0.25 to 0.04)

(−0.60 to 0.38)

42%

−0.015 (−0.17 to 0.14), (4)*

Time to first defaecation/bowel movement (hrs)

MD (IV, Random)

12

−7.92 (−12.71 to −3.13)

(−22.19 to 6.36)

62%

−4.06 (−9.07 to 0.95), (4)

Time to first flatus (hrs)

MD (IV, Random)

13

−4.09 (−6.30 to −1.87)

(−10.431 to 2.26)

63%

−3.63(−5.88 to −1.37), (1)

Length of hospital stay (days)

MD (IV, Random)

32

−0.37 (−0.60 to −0.15)

(−1.26 to 0.52)

69%

−0.19 (−0.42 to −0.04), (8)*

Postoperative nausea, overall (0 to 24 hrs, to 48 hrs, to 72 hrs)

RR (MH, Random)

35

0.78 (0.67 to 0.91)

(0.49 to 1.23)

22%

0.87 (0.74 to 1.03), (9)*

Postoperative vomiting, overall (0 to 24 hrs, to 48 hrs, to 72 hrs)

RR (MH, Random)

19

0.83 (0.63 to 1.08)

(0.62 to 1.10)

0%

0.89 (0.69 to 1.15), (3)

Intraoperative opioid consumption (MEQ, mg)

MD (IV, Random)

18

−2.14 (−3.87 to −0.40)

(−8.13 to 3.86)

80%

−2.10 (−3.83 to −0.38), (1)

Postoperative opioid consumption, PACU (MEQ, mg)

MD (IV, Random)

21

−3.10 (−3.87 to −2.32)

(−5.43 to −0.77)

40%

−2.91 (−3.72 to −2.11), (2)

Postoperative opioid consumption, overall (MEQ, mg)

MD (IV, Random)

40

−4.52 (−6.25 to −2.79)

(−12.03 to 3.00)

73%

−1.09 (−2.97 to 0.79), (16)*

Acronyms and abbreviations used in this table:

CI = confidence interval, hr = hour, IV = inverse variance, MD = mean difference, MEQ =morphine equivalent dose, MH = Mantel Haenszel, PACU = postanaesthesia care unit, PI = prediction interval, RR = risk ratio, SMD = standardized mean difference

We analysed all studies with 10 or more studies for funnel plot asymmetry. The asterisk (*) indicates that we rejected the null hypothesis of funnel plot asymmetry (P < 0.1).

Figuras y tablas -
Table 11. Heterogeneity/prediction intervals/publication bias
Table 12. Adverse events

Study ID

Type of adverse event/side effect ‐ lidocaine group

Type of adverse event/side effect ‐ control group

No adverse events/side effects detectable (statement)

Ahn 2015

NA

NA

“Not one patient had a postoperative complication related to lidocaine infusion.”

Baral 2010

Light headache (3), cardiac arrhythmias (0), perioral numbness (0), hypotension (0)

Cardiac arrhythmias (0), perioral numbness (0), hypotension (0)

NA

Bryson 2010

Light‐headedness, tinnitus, dysgeusia, etc. (11)

Light‐headedness, tinnitus, dysgeusia, etc. (21)

NA

Cassuto 1985

Light‐headedness (1)

Light‐headedness (1)

"No adverse reactions to lidocaine were observed."

Chen 2015

NA

NA

NA

Choi SJ 2012

Dizziness 24 hrs/ 48 hrs/ 72 hrs (1/1/1),

itching 24 hrs/ 48 hrs/ 72 hrs (8/3/0),

respiratory repression 24 hrs/ 48 hrs/ 72 hrs (0/0/0)

Dizziness 24 hrs/ 48 hrs/ 72 hrs (3/2/1),

itching 24 hrs/ 48 hrs/ 72 hrs (6/2/1),

respiratory repression 24 hrs/ 48 hrs/ 72 hrs (0/0/0)

NA

Choi GJ 2016

NA

NA

“There were no adverse events related to the investigational procedure or systemic administration of lidocaine such as arrhythmia, perioral numbness, visual disturbance, metal taste, or light‐headedness.”

Choi KW 2016

NA

NA

"In addition, none of the patients showed symptoms or signs associated with lidocaine toxicity during the perioperative period."

Cui 2010

Drowsiness (0), metal taste (0), perioral numbness (0), visual disturbances (0)

Drowsiness (0), metal taste (0), perioral numbness (0), visual disturbances (0)

"No patient reported any side effect of lidocaine toxicity."

Dale 2016

Severe bradycardia (1), perioral paraesthesia (1), restless legs (1)

Severe bradycardia (0), perioral paraesthesia (0), restless legs (0)

NA

De Oliveira 2012

NA

NA

"We did not observe any potential cardiovascular or neurological side effects associated with the infusion of systemic lidocaine in our investigation."

De Oliveira 2014

NA

NA

"We did not observe any potential cardiovascular or neurological side effects associated with the infusion of systemic lidocaine in our investigation."

Dewinter 2016

NA

NA

“Both groups did not differ with respect to the incidence of other AEs. No patient receiving lidocaine reported subjective symptoms of local anesthetic systemic toxicity.”

El‐Tahan 2009

Perioperative arrhythmia (0), light‐headedness (0), headache (0), perioral numbness (0), tunnel vision (0), seizures (0)

Perioperative arrhythmia (0), light‐headedness (0), headache (0), perioral numbness (0), tunnel vision (0), seizures (0)

"There were no reported serious side effects during the study."

Farag 2013

Pneumonia (0), respiratory failure (0), cardiac arrest (0), arrhythmia (0), heart failure (0), stroke (0), intravascular coagulopathy (0), thromboembolism (0), delirium (0), monoplegia (0), upper gastrointestinal bleeding (0), sepsis (0), readmission (2)

Pneumonia (0), respiratory failure (0), cardiac arrest (0), arrhythmia (0), heart failure (0), stroke (0), intravascular coagulopathy (0), thromboembolism (0), delirium (0), monoplegia (1), upper gastrointestinal bleeding (0), sepsis (0), readmission (3)

NA

Grady 2012

NA

NA

NA

Grigoras 2012

NA

NA

"No side effects related to lidocaine were observed."

Groudine 1998

NA

NA

"No patient experienced identifiable adverse events related to the lidocaine infusion."

Herroeder 2007

NA

NA

NA

Insler 1995

Death (1), myocardial infarction (0)

Death (1), myocardial infarction (0)

NA

Ismail 2008

NA

NA

NA

Jain 2015

Drowsiness (3)

Drowsiness (0)

“None of the patients complained of lignocaine‐related side effects such as perioral numbness or metallic taste. The incidence of light‐headedness and nausea was comparable in both the groups. Three patients in Group B demonstrated drowsiness in the postoperative period lasting between 10 and 17 mins.”

Kaba 2007

NA

NA

NA

Kang 2011

NA

NA

NA

Kasten 1986

NA

NA

NA

Kim TH 2011

NA

NA

"There was no adverse effect from intravenous lidocaine throughout the study."

Kim TH 2013

NA

NA

"In our study, no neuropsychiatric events were observed throughout the process."

Kim HJ 2014

NA

NA

“No specific complication or side effect regarding lidocaine or dexmedetomidine was reported.”

Kim HO 2014

Hospital mortality (0)

Hospital mortality (0)

“There were no significant lidocaine‐related adverse events during our trial.”

Kim KT 2014

NA

NA

“There were no side effects from the lidocaine, such as arrhythmia, hypotension, and hypersensitivity.”

Koppert 2004

NA

NA

"No anaesthesiologist noted adverse events related to the lidocaine infusion during surgery. Furthermore, no patient after having regained consciousness complained of lidocaine‐related side effects such as perioral numbness or metallic taste. The incidences of drowsiness, light‐headedness, and nausea were comparable in the lidocaine and control groups."

Kuo 2006

Bradycardia (3)

Bradycardia (0)

"No patient experienced an identifiable adverse event related to IV lidocaine infusion."

Lauwick 2008

NA

NA

NA

Lauwick 2009

Bleeding (1), sepsis (1), chest infection (1)

Bladder leakage (1)

NA

Lee 2011

Atrial fibrillation (9), other arrhythmia (7), myocardial infarction (0), death (0)

Atrial fibrillation (5), other arrhythmia (10), myocardial infarction (0), death (0)

"All patients started on lidocaine completed their full course of drug and did not experience any adverse events related to the local anaesthetic, such as severe bradycardia (< 40 beats min‐1), asystole, or neurological symptoms."

Maquoi 2016

NA

NA

NA

Martin 2008

NA

NA

"No patient reported lidocaine toxicity side effects and no adverse events were reported in both groups"

Mathew 2009

Serious adverse events (12.3%), no detailed description

Serious adverse events (10.2%), no detailed description

"Adverse events were not significantly different between treatment groups."

McKay 2009

Dizziness and visual disturbances (1)

NA

"There were no serious adverse events recorded."

Mitchell 1999

Death (1)

Death (1)

NA

Mitchell 2009

Death due to multiorgan failure (3) and acute graft occlusion (1)

Death (0)

NA

Oliveira 2015

NA

NA

NA

Omar 2013

Hypotension (0)

Hypotension (0)

NA

Ortiz 2016

NA

NA

"There was no arrhythmia or adverse effect occurrence."

Peng 2016

Hypertension (3), coronary heart disease (0)

Hypertension (4), coronary heart disease (0)

“There were no seizures or other symptoms of potential lidocaine toxicity found in patients who received lidocaine infusion. There was no significant difference in the number of cases complicated by hypertension, tachycardia, dysphoria, or PONV between the normal saline group and the lidocaine group.”

Rimbäck 1990

Sedation (2)

NA

"No adverse reactions to lidocaine were reported."

Saadawy 2010

NA

NA

NA

Samimi 2015

NA

NA

"...also none of the patients experienced lidocaine‐related adverse effects."

Slovack 2015

Confusion (1), sedation (2), light‐headedness/dizziness (0), blurred vision (0), hypotension (0), respiratory depression (0), pruritus (0)

Confusion (0), sedation (0), light‐headedness/dizziness (0), blurred vision (1), hypotension (1), respiratory depression (0), pruritus (0)

NA

Soltani 2013

NA

NA

NA

Sridhar 2015

NA

NA

NA

Staikou 2014

Transient confusion in PACU (1), bradycardia requiring treatment (0)

Transient confusion in PACU (0), bradycardia requiring treatment (0)

NA

Striebel 1992

NA

NA

No signs of urticaria, dermatitis, asthma bronchiale, anaphylactic shock, restlessness, anxiety, lalopathy, tinnitus, metallic taste, dizziness, visual disturbance, and tremor.

Swenson 2010

Wound infection (0), anaemia (1), anxiety (1), supraventricular tachycardia (3), back pain (0), bradycardia (0), confusion (2), decreased oxygen saturation level (1), dizziness/light‐headedness (1), fever (1), hyperglycaemia (3), hypertension (3), itching (3), lower extremity numbness (1), intravascular device infection (0), syncope (0), arrhythmia severe (1), confusion severe (1), facial numbness severe (1), shortness of breath (1)

Wound infection (1), anaemia (1), anxiety (0), supraventricular tachycardia (1), back pain (1), bradycardia (1), confusion (0), decreased oxygen saturation level (0), dizziness/light‐headedness (1), fever (1), hyperglycaemia (0), hypertension (0), itching (3), lower extremity numbness (6), intravascular device infection (1), syncope (1), arrhythmia severe (1), confusion severe (0), facial numbness severe (0), shortness of breath (0)

NA

Terkawi 2014

NA

NA

"...no toxicity cases were reported in our cohort...."

Tikuisis 2014

Light‐headedness (0), perioral numbness (0), metallic taste (0), dizziness (0), and visual disturbances (0)

NA

"Lidocaine‐associated haemodynamic changes such as severe hypotension, bradycardia, and arrhythmia were not observed in any lidocaine group patient during surgery."

Wallin 1987

Drowsiness (2)

NA

"Aside from drowsiness in two patients of the lidocaine group, no side effects due to possible lidocaine overdosage were reported."

Wang 2002

Death (2)

Death (4)

NA

Wang 2015

NA

NA

NA

Weinberg 2016

Pruritus (6), dizziness (14), visual disturbances (4), perioral numbness (2), muscle weakness (1), constipation (4)

Pruritus (9), dizziness (20), visual disturbances (6), perioral numbness (2), muscle weakness (3), constipation (10)

NA

Wongyingsinn 2011

NA

NA

"No patients showed signs of lidocaine toxicity in the postoperative period."

Wu 2005

NA

NA

"No patient experienced an identifiable adverse event related to the lidocaine infusion, except that an occasional arrhythmia with stable vital signs was noted in one patient in both groups."

Wuethrich 2012

Light‐headedness (0), drowsiness (0), perioral numbness (0), visual disturbances (0), metal taste (0), pathological cardiac rhythm disturbances (0), and seizures (0)

NA

"No postoperative complications and no adverse events related to systemic administration of lidocaine were observed."

Xu 2017

NA

NA

NA

Yang 2014

Blurred vision (0), hearing problems (0), peripheral paraesthesia (0), dizziness (0), uncontrolled muscle contraction (0), convulsions (0), hypotension (0),
bradycardia (0), headache (0), and itching (0)

NA

NA

Yardeni 2009

NA

NA

NA

Yon 2014

Shivering (0), tinnitus (0)

Shivering (1), tinnitus (0)

NA

Zengin 2015

Pruritus (1)

Pruritus (4)

NA

Acronyms and abbreviations used in this table:

AE = adverse events, IV = intravenous, NA = not available, PACU = postanaesthesia care unit, PONV = postoperative nausea and vomiting

Figuras y tablas -
Table 12. Adverse events
Comparison 1. Intravenous (IV) lidocaine versus placebo

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain score at rest, 'early time points' (1 h to 4 h, PACU) Show forest plot

29

1656

Std. Mean Difference (IV, Random, 95% CI)

‐0.50 [‐0.72, ‐0.28]

1.1 open abdominal surgery

8

448

Std. Mean Difference (IV, Random, 95% CI)

‐0.54 [‐0.82, ‐0.26]

1.2 laparoscopic abdominal surgery

10

518

Std. Mean Difference (IV, Random, 95% CI)

‐0.78 [‐1.34, ‐0.21]

1.3 other surgery

11

690

Std. Mean Difference (IV, Random, 95% CI)

‐0.21 [‐0.44, 0.02]

2 Pain score at rest, 'intermediate time points' (24 h) Show forest plot

33

1847

Std. Mean Difference (IV, Random, 95% CI)

‐0.14 [‐0.25, ‐0.04]

3 Pain score at rest, 'late time points' (48 h) Show forest plot

24

1404

Std. Mean Difference (IV, Random, 95% CI)

‐0.11 [‐0.25, 0.04]

4 Postoperative ileus (dichotomous) Show forest plot

4

273

Risk Ratio (M‐H, Random, 95% CI)

0.37 [0.15, 0.87]

5 Time to first defaecation/bowel movement (h) Show forest plot

12

684

Mean Difference (IV, Random, 95% CI)

‐7.92 [‐12.71, ‐3.13]

6 Time to first flatus (h) Show forest plot

13

785

Mean Difference (IV, Random, 95% CI)

‐4.09 [‐6.30, ‐1.87]

7 Time to first bowel sounds (h) Show forest plot

2

110

Mean Difference (IV, Random, 95% CI)

‐6.08 [‐13.77, 1.60]

8 Length of hospital stay (days) Show forest plot

32

2077

Mean Difference (IV, Random, 95% CI)

‐0.37 [‐0.60, ‐0.15]

9 Length of hospital stay (outpatient surgery, mins) Show forest plot

3

191

Mean Difference (IV, Random, 95% CI)

‐10.81 [‐36.93, 15.31]

10 Surgical complications ‐ anastomotic leak Show forest plot

3

188

Risk Ratio (M‐H, Random, 95% CI)

0.61 [0.08, 4.80]

11 Surgical complications ‐ bleeding Show forest plot

3

222

Risk Ratio (M‐H, Random, 95% CI)

1.79 [0.41, 7.89]

12 Surgical complications ‐ postoperative infection Show forest plot

5

352

Risk Ratio (M‐H, Random, 95% CI)

1.64 [0.41, 6.52]

13 Patient satisfaction Show forest plot

6

306

Mean Difference (IV, Random, 95% CI)

0.76 [0.46, 1.06]

14 Postoperative nausea, 'early time points' (PACU) Show forest plot

8

511

Risk Ratio (M‐H, Random, 95% CI)

0.72 [0.53, 0.98]

15 Postoperative nausea, 'overall' (0 to 24 h, to 48 h, to 72 h) Show forest plot

35

1903

Risk Ratio (M‐H, Random, 95% CI)

0.78 [0.67, 0.91]

16 Postoperative vomiting, 'early time points' (PACU) Show forest plot

4

305

Risk Ratio (M‐H, Random, 95% CI)

0.49 [0.16, 1.48]

17 Postoperative vomiting, 'overall' (0 to 24 h, to 48 h, to 72 h) Show forest plot

19

1026

Risk Ratio (M‐H, Random, 95% CI)

0.83 [0.63, 1.08]

18 Intraoperative opioid consumption (MEQ, mg) Show forest plot

18

1116

Mean Difference (IV, Random, 95% CI)

‐2.14 [‐3.87, ‐0.40]

19 Intraoperative remifentanil consumption (µg) Show forest plot

6

490

Mean Difference (IV, Random, 95% CI)

‐14.17 [‐35.27, 6.92]

20 Postoperative opioid consumption, PACU (MEQ, mg) Show forest plot

21

1219

Mean Difference (IV, Random, 95% CI)

‐3.10 [‐3.87, ‐2.32]

21 Postoperative opioid consumption, overall (MEQ, mg) Show forest plot

40

2201

Mean Difference (IV, Random, 95% CI)

‐4.52 [‐6.25, ‐2.79]

Figuras y tablas -
Comparison 1. Intravenous (IV) lidocaine versus placebo
Comparison 2. Intravenous (IV) lidocaine versus thoracic epidural analgesia (TEA)

Outcome or subgroup title

No. of studies

No. of participants

Statistical method

Effect size

1 Pain score (VAS 0 to 10 cm) at rest, 'intermediate time points' (24 h) Show forest plot

2

102

Mean Difference (IV, Random, 95% CI)

1.51 [‐0.29, 3.32]

2 Pain score (VAS 0 to 10 cm) at rest, 'late time points' (48 h) Show forest plot

2

102

Mean Difference (IV, Random, 95% CI)

0.98 [‐1.19, 3.16]

3 Time to first bowel movement (h) Show forest plot

2

102

Mean Difference (IV, Random, 95% CI)

‐1.66 [‐10.88, 7.56]

4 Length of hospital stay (days) Show forest plot

2

102

Mean Difference (IV, Random, 95% CI)

‐0.02 [‐0.38, 0.33]

5 Intraoperative opioid consumption (MEQ, mg) Show forest plot

2

100

Mean Difference (IV, Random, 95% CI)

7.27 [‐13.92, 28.47]

Figuras y tablas -
Comparison 2. Intravenous (IV) lidocaine versus thoracic epidural analgesia (TEA)