S-ketamine in patient-controlled analgesia reduces opioid consumption in a dose-dependent manner after major lumbar fusion surgery: A randomized, double-blind, placebo-controlled clinical trial

Elina C V Brinck, Taru Virtanen, Sanna Mäkelä, Venla Soini, Ville-Veikko Hynninen, Jukka Mulo, Urmas Savolainen, Juho Rantakokko, Kreu Maisniemi, Antti Liukas, Klaus T Olkkola, Vesa Kontinen, Pekka Tarkkila, Marko Peltoniemi, Teijo I Saari, Elina C V Brinck, Taru Virtanen, Sanna Mäkelä, Venla Soini, Ville-Veikko Hynninen, Jukka Mulo, Urmas Savolainen, Juho Rantakokko, Kreu Maisniemi, Antti Liukas, Klaus T Olkkola, Vesa Kontinen, Pekka Tarkkila, Marko Peltoniemi, Teijo I Saari

Abstract

Background: Spinal fusion surgery causes severe pain. Strong opioids, commonly used as postoperative analgesics, may have unwanted side effects. S-ketamine may be an effective analgesic adjuvant in opioid patient-controlled analgesia (PCA). However, the optimal adjunct S-ketamine dose to reduce postoperative opioid consumption is still unknown.

Methods: We randomized 107 patients at two tertiary hospitals in a double-blinded, placebo-controlled clinical trial of adults undergoing major lumbar spinal fusion surgery. Patients were randomly allocated to four groups in order to compare the effects of three different doses of adjunct S-ketamine (0.25, 0.5, and 0.75 mg ml-1) or placebo on postoperative analgesia in oxycodone PCA. Study drugs were administered for 24 hours postoperative after which oxycodone-PCA was continued for further 48 hours. Our primary outcome was cumulative oxycodone consumption at 24 hours after surgery.

Results: Of the 100 patients analyzed, patients receiving 0.75 mg ml-1 S-ketamine in oxycodone PCA needed 25% less oxycodone at 24 h postoperatively (61.2 mg) compared with patients receiving 0.5 mg ml-1 (74.7 mg) or 0.25 mg ml-1 (74.1 mg) S-ketamine in oxycodone or oxycodone alone (81.9 mg) (mean difference: -20.6 mg; 95% confidence interval [CI]: -41 to -0.20; P = 0.048). A beneficial effect in mean change of pain intensity at rest was seen in the group receiving 0.75 mg ml-1 S-ketamine in oxycodone PCA compared with patients receiving lower ketamine doses or oxycodone alone (standardized effect size: 0.17, 95% CI: 0.013-0.32, P = 0.033). The occurrence of adverse events was similar among the groups.

Conclusions: Oxycodone PCA containing S-ketamine as an adjunct at a ratio of 1: 0.75 decreased cumulative oxycodone consumption at 24 h after major lumbar spinal fusion surgery without additional adverse effects.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Fig 1. DoseRespKeta trial structure and CONSORT…
Fig 1. DoseRespKeta trial structure and CONSORT 2009 flow diagram.
CONSORT indicates Consolidated Standards of Reporting Trials.
Fig 2. Postoperative cumulative oxycodone ( top…
Fig 2. Postoperative cumulative oxycodone (top row) and S-ketamine (bottom row) consumption during the first 24 h in four patient-controlled analgesia (PCA) treatment groups (G1¬–G4).
PCA, patient-controlled analgesia. Box plots show the median and 25–75th percentiles, and the whiskers indicate the minimum and maximum. Blue diamonds show the mean oxycodone consumption in each plot.
Fig 3
Fig 3
A) Patient-reported numerical rating scale (NRS) values at rest during the first 24 h in four patient-controlled analgesia (PCA) treatment groups G1-G4. Box plots show the median and 25–75th percentiles, and the whiskers indicate the minimum and maximum. (B) Risk ratios (95% CI) for reporting NRS value 4 or higher. PACU, postoperative care unit; CI, confidence interval.

References

    1. Gerbershagen HJ, Aduckathil S, van Wijck AJM, Peelen LM, Kalkman CJ, Meissner W. Pain intensity on the first day after surgery: a prospective cohort study comparing 179 surgical procedures. Anesthesiology. 2013;118:934–944. doi: 10.1097/ALN.0b013e31828866b3
    1. Kehlet H, Jensen TS, Woolf CJ. Persistent postsurgical pain: risk factors and prevention. Lancet. 2006;367:1618–1625. doi: 10.1016/S0140-6736(06)68700-X
    1. Apfelbaum JL, Chen C, Mehta SS, Gan TJ. Postoperative pain experience: results from a national survey suggest postoperative pain continues to be undermanaged. Anesth Analg. 2003;97:534–540. doi: 10.1213/01.ane.0000068822.10113.9e
    1. Kurd MF, Kreitz T, Schroeder G, Vaccaro AR. The role of multimodal analgesia in spine surgery. J Am Acad Orthop Surg. 2017;25:260–268. doi: 10.5435/JAAOS-D-16-00049
    1. Cozowicz C, Bekeris J, Poeran J, Zubizarreta N, Schwenk E, Girardi F, et al.. Multimodal pain management and postoperative outcomes in lumbar spine fusion surgery: a population-based cohort study. Spine (Phila Pa 1976). 2020;45:580–589. doi: 10.1097/BRS.0000000000003320
    1. McNicol E, Ferguson MC, Hudcova J. Patient-controlled analgesia versus non-patient controlled analgesia for postoperative pain. Cochrane Database Syst Rev. 2015;6:CD003348. doi: 10.1002/14651858.CD003348.pub3
    1. Dinges HC, Otto S, Stay DK, Bäumlein S, Waldmann S, Kranke P, et al.. Side effect rates of opioids in equianalgesic doses via intravenous patient-controlled analgesia: a systematic review and network meta-analysis. Anesth Analg. 2019;129:1153–1162. doi: 10.1213/ANE.0000000000003887
    1. Peltoniemi MA, Hagelberg NM, Olkkola KT, Saari TI. Ketamine: a review of clinical pharmacokinetics and pharmacodynamics in anesthesia and pain therapy. Clin Pharmacokinet. 2016;55:1059–1077. doi: 10.1007/s40262-016-0383-6
    1. Basbaum AI, Bautista DM, Scherrer G, Julius D. Cellular and molecular mechanisms of pain. Cell. 2009; 139:267–284. doi: 10.1016/j.cell.2009.09.028
    1. Hirota K, Lambert DG. Ketamine: new uses for an old drug? Br J Anaesth. 2011;107:123–126. doi: 10.1093/bja/aer221
    1. Woolf CJ. Pain amplification—a perspective on the how, why, when, and where of central sensitization. J Appl Biobehav Res. 2018;23:e12124.
    1. Brinck EC, Tiippana E, Heesen M, Bell RF, Straube S, Moore RA, et al.. Perioperative intravenous ketamine for acute postoperative pain in adults. Cochrane Database Syst Rev. 2018;12:CD012033. doi: 10.1002/14651858.CD012033.pub4
    1. Assouline B, Tramèr MR, Kreienbühl L, Elia N. Benefit and harm of adding ketamine to an opioid in a patient-controlled analgesia device for the control of postoperative pain: systematic review and meta-analyses of randomized controlled trials with trial sequential analyses. Pain. 2016;157:2854–2864. doi: 10.1097/j.pain.0000000000000705
    1. Sveticic G, Gentilini A, Eichenberger U, Luginbühl M, Curatolo M. Combinations of morphine with ketamine for patient-controlled analgesia: a new optimization method. Anesthesiology. 2003;98:1195–11205. doi: 10.1097/00000542-200305000-00023
    1. Sveticic G, Eichenberger U, Curatolo M. Safety of mixture of morphine with ketamine for postoperative patient-controlled analgesia: an audit with 1026 patients. Acta Anaesthiol Scand. 2005;49:870–875. doi: 10.1111/j.1399-6576.2005.00740.x
    1. Sveticic G, Farzanegan F, Zmoos P, Zmoos S, Eichenberger U, Curatolo M. Is the combination of morphine with ketamine better than morphine alone for postoperative intravenous patient-controlled analgesia? Anesth Analg. 2008;106:287–293, table of contents. doi: 10.1213/01.ane.0000289637.11065.8f
    1. Schnider TW, Minto CF, Shafer SL, Gambus PL, Andresen C, Goodale DB, et al.. The influence of age on propofol pharmacodynamics. Anesthesiology. 1999; 90:1502–1516. doi: 10.1097/00000542-199906000-00003
    1. Minto CF, Schnider TW, Egan TD, Youngs E, Lemmens HJ, Billard V, et al.. Influence of age and gender on the pharmacokinetics and pharmacodynamics of remifentanil. I. Model development. Anesthesiology. 1997; 86:10–23. doi: 10.1097/00000542-199701000-00004
    1. Lin T-F, Yeh Y-C, Lin F-S, Wang Y-P, Sun W-Z, Fan S-F. Effect of combining dexmedetomidine and morphine for intravenous patient-controlled analgesia. Br J Anaesth. 2009;102:117–122. doi: 10.1093/bja/aen320
    1. Tomczak M, Tomczak E. The need to report effect size estimates revisited. An overview of some recommended measures of effect size. Trends Sport Sci. 2014;1:19–25.
    1. RStudio Team. RStudio: Integrated Development Environment for R. 2020. . Accessed: 13 March, 2020.
    1. R Core Team. R: A Language and Environment for Statistical Computing. 2019. . Accessed: 13 March 2020.
    1. Zanos P, Moaddel R, Morris PJ, Riggs LM, Highland JN, Georgiou P et al.. Ketamine and Ketamine Metabolite Pharmacology: Insights into Therapeutic Mechanisms. Pharmacol Rev. 2018;70:621–660. doi: 10.1124/pr.117.015198
    1. Pendi A, Field R, Farhan S-D, Eichler M, Bederman SS. Perioperative ketamine for analgesia in spine surgery: a meta-analysis of randomized controlled trials. Spine (Phila Pa 1976). 2018; 43:E299–E307. doi: 10.1097/BRS.0000000000002318
    1. Pacreu S, Fernández Candil J, Moltó L, Carazo J, Fernández Galinski S. The perioperative combination of methadone and ketamine reduces post-operative opioid usage compared with methadone alone. Acta Anaesthiol Scand. 2012;56:1250–1256. doi: 10.1111/j.1399-6576.2012.02743.x
    1. Yeom JH, Chon M-S, Jeon WJ, Shim J-H. Perioperative ketamine with the ambulatory elastometric infusion pump as an adjuvant to manage acute postoperative pain after spinal fusion in adults: a prospective randomized trial. Korean J Anesthesiol. 2012;63:54–58. doi: 10.4097/kjae.2012.63.1.54
    1. Javery KB, Ussery TW, Steger HG, Colclough GW. Comparison of morphine and morphine with ketamine for postoperative analgesia. Can J Anaesth. 1996;43:212–215. doi: 10.1007/BF03011736
    1. Nitta R, Goyagi T, Nishikawa T. Combination of oral clonidine and intravenous low-dose ketamine reduces the consumption of postoperative patient-controlled analgesia morphine after spine surgery. Acta Anaesthesiol Taiwan. 2013;51:14–17. doi: 10.1016/j.aat.2013.03.003
    1. Song JW, Shim JK, Song Y, Yang SY, Park SJ, Kwak YL. Effect of ketamine as an adjunct to intravenous patient-controlled analgesia, in patients at high risk of postoperative nausea and vomiting undergoing lumbar spinal surgery. Br J Anaesth. 2013;111:630–635. doi: 10.1093/bja/aet192
    1. Loftus RW, Yeager MP, Clark JA, Brown JR, Abdu WA, Sengupta DK. et al.. Intraoperative ketamine reduces perioperative opiate consumption in opiate-dependent patients with chronic back pain undergoing back surgery. Anesthesiology. 2010;113:639–646. doi: 10.1097/ALN.0b013e3181e90914
    1. Nielsen RV, Fomsgaard JS, Siegel H, Martuseviicius R, Nikolajsen L, Dahl JB, et al.. Intraoperative ketamine reduces immediate postoperative opioid consumption after spinal fusion surgery in chronic pain patients with opioid dependency: a randomized, blinded trial. Pain. 2017;158:463–470. doi: 10.1097/j.pain.0000000000000782
    1. Nielsen RV, Fomsgaard JS, Nikolajsen L, Dahl JB, Mathiesen O. Intraoperative S-ketamine for the reduction of opioid consumption and pain one year after spine surgery: a randomized clinical trial of opioid-dependent patients. Eur J Pain. 2019;23:455–460. doi: 10.1002/ejp.1317
    1. Boenigk K, Echevarria GC, Nisimov E, von Bergen Granell AE, Cuff GE, Wang J, et al.. Low-dose ketamine infusion reduces postoperative hydromorphone requirements in opioid-tolerant patients following spinal fusion: a randomised controlled trial. Eur J Anaesthesiol. 2019;36:8–15. doi: 10.1097/EJA.0000000000000877
    1. Wang L, Johnston B, Kaushal A, Cheng D, Zhu F, Martin J. Ketamine added to morphine or hydromorphone patient-controlled analgesia for acute postoperative pain in adults: a systematic review and meta-analysis of randomized trials. Can J Anaesth. 2016;63:311–325. doi: 10.1007/s12630-015-0551-4
    1. Carstensen M, Møller AM. Adding ketamine to morphine for intravenous patient-controlled analgesia for acute postoperative pain: a qualitative review of randomized trials. Br J Anaesth. 2010;104:401–406. doi: 10.1093/bja/aeq041
    1. Moore RA, Mhuircheartaigh RJN, Derry S, McQuay HJ. Mean analgesic consumption is inappropriate for testing analgesic efficacy in post-operative pain: analysis and alternative suggestion. Eur J Anaesthesiol. 2011;28:427–432. doi: 10.1097/EJA.0b013e328343c569
    1. Altman DG, Royston P. The cost of dichotomising continuous variables. BMJ. 2006;332:1080. doi: 10.1136/bmj.332.7549.1080
    1. Brinck ECV, Maisniemi K, Kankare J, Tielinen L, Tarkkila P, Kontinen VK. Analgesic effect of intraoperative intravenous S-ketamine in opioid-naïve patients after major lumbar fusion surgery is temporary and not dose-dependent: a randomized, double-blind, placebo-controlled clinical trial. Anesth Analg. 2021;132:69–79. doi: 10.1213/ANE.0000000000004729

Source: PubMed

3
Subscribe