Back to Journals » Journal of Pain Research » Volume 12

Remifentanil-Induced Secondary Hyperalgesia Is Not Prevented By Preoperative Acetazolamide Administration In Patients Undergoing Total Thyroidectomy: A Randomized Controlled Trial

Authors Gutiérrez R, Contreras F, Blanch A, Bravo D, Egaña JI , Rappoport D, Cabané P, Rodríguez F, Penna A 

Received 28 June 2019

Accepted for publication 17 September 2019

Published 6 November 2019 Volume 2019:12 Pages 2991—2997

DOI https://doi.org/10.2147/JPR.S221131

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 3

Editor who approved publication: Professor E Alfonso Romero-Sandoval



Rodrigo Gutiérrez,1,2 Felipe Contreras,1 Alonso Blanch,1 Daniela Bravo,1 José I Egaña,1 Daniel Rappoport,3 Patricio Cabané,3 Francisco Rodríguez,3 Antonello Penna1,2

1Department of Anesthesiology and Perioperative Medicine, Hospital Clínico Universidad de Chile, Santiago, Chile; 2Centro de Investigación Clínica Avanzada (CICA), Facultad de Medicina and Hospital Clínico Universidad de Chile, Santiago, Chile; 3Head and Neck Surgery, Department of Surgery, Hospital Clínico Universidad de Chile, Santiago, Chile

Correspondence: Antonello Penna
Departmento de Anestesiología y Medicina Perioperatoria, Hospital Clínico, Universidad de Chile, Santos Dumont 999, Santiago 838 0456, Chile
Tel +56-2-29788209
Email [email protected]

Purpose: Acute administration of remifentanil may lead to opioid-induced hyperalgesia (OIH). Studies in mice suggest that OIH is mediated by impaired anionic homeostasis in spinal lamina I neurons due to a down-regulation of the K+-Cl co-transporter KCC2, which was reverted using acetazolamide (ACTZ), a carbonic anhydrase inhibitor. We propose that ACTZ prevents remifentanil-mediated OIH in humans.
Patients and methods: We conducted a randomized, double-blind, placebo-controlled clinical trial between December 2016 and September 2018. Patients were randomly allocated to receive ACTZ (250 mg of ACTZ 2 h before surgery) or placebo. To detect hyperalgesia, mechanical pain threshold (MPT) were measured before and after surgery using hand-held von Frey filaments in the forearm. Anesthesia was maintained with remifentanil at a target effect site of 4.5 ± 0.5 ng/mL, and sevoflurane at an end-tidal concentration of 0.8 MAC corrected for age.
Results: In total, 47 patients completed the study. Both groups were comparable in the baseline characteristics and intraoperative variables. Baseline MPT were similar in both groups. However, MPT in the forearm significantly diminished in the time in both groups. Finally, postoperative pain and morphine consumption were similar between groups.
Conclusion: Both groups developed remifentanil-mediated OIH at 12–18 h after surgery. However, ACTZ did not prevent the MPT reduction in patients undergoing total thyroidectomy.

Keywords: anesthesia, chloride dysregulation, carbonic anhydrase, pain

Introduction

Opioid-induced hyperalgesia (OIH) is a sensitization process in which opioids, paradoxically, cause pain hypersensitivity.1,2 A hypersensitivity in a distant place from the surgery site is secondary hyperalgesia, which is manifested as a decline in the mechanical pain threshold (MPT).2,3 The relevance of OIH and secondary hyperalgesia in humans is controversial. Nonetheless, clinical trials show consistently that high-dose of opioids during surgery increases postoperative pain and opioid consumption after surgery.3 Particularly in patients who receive the ultrashort-acting μ-opioid receptor agonist, remifentanil.46 Moreover, a high intraoperative dose of remifentanil is associated with OIH manifested as a diminished in the MPT surrounding the surgical wound on the day after surgery (ie, primary hyperalgesia).7

The canonical mechanism related to OIH is the activation of N-methyl-D-aspartate (NMDA)-dependent pronociceptive systems leading to a reduction in the nociceptive thresholds.2,8 However, some studies suggest that OIH might also be explained by alternative mechanisms (Figure 1A and B) in animal models.9 Briefly, it has been proposed that OIH may be mediated, at least in part, by a downregulation of the K-Cl co-transporter 2 (KCC2) expression in the spinal dorsal horn in rats.10 This would lead to an intracellular accumulation of chloride (Cl) in neurons, and consequently, GABAA receptor inhibition might be abolished by depolarization of the Cl reversal potential (ie, chloride dysregulation).11 In normal conditions, GABAA receptors inhibition depends on the influx of Cl, which is counterbalanced by an efflux of bicarbonate (HCO3) as GABAA receptors are permeable to Cl and HCO3 in a 4:1 permeability ratio.1214 Thus, chloride dysregulation could be reverted by reducing the HCO3 efflux (Figure 1C). One way to attenuate HCO3 efflux is to block the reaction catalyzed by carbonic anhydrase that restores the intracellular HCO3 levels.15 Indeed, spinal inhibition of carbonic anhydrase reduced neuropathic allodynia in rats,16 and also avoided morphine-induced hyperalgesia in mice.11 To demonstrate that chloride dysregulation is an alternative mechanism of OIH in humans, we propose that inhibition of carbonic anhydrase by acetazolamide (ACTZ) might revert OIH.

Figure 1 Scheme representing the molecular mechanism of acetazolamide (ACTZ) to revert hyperalgesia generated by chloride-dysregulation induced for opioids. (A) Normal condition: i) GABAA receptors activation leads to a Cl influx and HCO3 efflux; ii) KCC2 maintains a low Cl intracellular concentration, and thus, the reversal potential for GABAA receptor (EGABA) is approximately −60 mV. EGABA depends on Cl and HCO3 with 4:1 ratio of permeability. (B) Remifentanil-induced hyperalgesia by downregulation of the KCC2: i) Remifentanil leads to a downregulation of the KCC2 expression by a BDNF-mediated mechanism11 generating an increase in the intracellular Cl concentration and therefore changing the Cl gradient; ii) EGABA is depolarized to approximately −50 mV. (C) Acetazolamide reverses the chloride-dysregulation by inhibition of the carbonic anhydrase: i) Acetazolamide inhibits carbonic anhydrase leading to a reduction in the intracellular HCO3 concentration; ii) EGABA is consequently hyperpolarized to approximately −60 mV.

Thus, the aim of this clinical trial was to test whether preoperative administration of 250 mg of ACTZ during sevoflurane-remifentanil anesthesia diminishes the postoperative remifentanil-induced secondary hyperalgesia in patients undergoing total thyroidectomy.

Materials And Methods

This trial was registered in ClinicalTrials.gov, ID number: NCT02992938 and was conducted in accordance with the Declaration of Helsinki. After obtaining ethics committee approval (Hospital Clínico de la Universidad de Chile, Santiago, Chile) we conducted a randomized, double-blind, placebo-controlled clinical trial between December 2016 and September 2018. We included patients between 18 and 65 years, American Society of Anesthesiologist (ASA) I or II, scheduled for total thyroidectomy without neck dissection. Exclusion criteria were chronic pain history, previous partial thyroidectomy, analgesic use in the last 48 h, body mass index > 30 kg·m-2, contraindication to receive ACTZ or a sevoflurane-based anesthesia, and unable to use a patient-controlled analgesia (PCA) device. After signed a written informed consent, patients were randomly allocated to the ACTZ group (250 mg of ACTZ 2 h before surgery) or to the control group (placebo 2 h before surgery). Placebo tablets were the same in terms of color and shape to those containing ACTZ. Patients were allocated to each group using a random-number table whose information was kept in sealed envelopes. After the enrollment of a patient, the respective envelope was opened, and the medication was administered according to the information in the envelope.

To determine the secondary hyperalgesia (ie, in a distant place from surgical site), the MPT was measured using hand-held von Frey filaments in the right forearm as previously described.7,17 All the evaluations were performed by the same operator (RG). Baseline MPT measurement was performed before the surgery, and at 2 h and 12–18 h after surgery in the forearm without peripheral venous catheter. Since, we did not include patients scheduled at first hour in the morning, the 12–18 h interval evaluation corresponds to the morning at the day after surgery. General anesthesia induction was standardized as follows: remifentanil infusion was guided by Minto’s model18 to an effect-site target of 6 ng·mL−1, propofol 2 mg·kg−1, lidocaine 40 mg, and rocuronium 0.6 mg·kg−1 to facilitate orotracheal intubation. After tracheal intubation, patients were protectively ventilated to normocapnia with 40–50% oxygen without nitrous oxide. Patients received intravenous ketorolac 60 mg and acetaminophen 1 g before the incision. Anesthesia was maintained with remifentanil at target effect-site of 4.5 ± 0.5 ng·mL−1, and sevoflurane at an end-tidal concentration of 0.8 MAC corrected for age. Thirty minutes before the anticipated end of surgery, a 0.1 mg·kg−1 bolus dose of morphine was given intravenously, and then, a morphine PCA device was connected, which was programmed with a 1 mg bolus of morphine and a lockout interval of 8 min. All participants were preoperatively instructed on the PCA use. Ketamine was not used in any patient in this protocol.

The primary endpoint was to detect a higher MPT at 12–18 h in the ACTZ group respect to control group. Based on previous results17 the number of patients needed to find a difference of 20 g (65 g vs 45 g) in the MPT was 19 per group, with a power of 0.80 and an α of 0.05. Assuming a dropout rate of 30%, we calculated a sample size of 25 patients per group. Secondary outcomes were pain evaluated with a numeric pain rating scale (NPRS) and morphine consumption during the first 24 h after surgery. Any adverse event was registered and follow up during the hospital stay.

The analysis of the von Frey measurement, NPRS, and morphine consumption was performed using two-way ANOVA. Demographics and intraoperative data were analyzed with Fisher exact test or t test as corresponded. A p value <0.05 was considered statically significant.

Results

Fifty patients were enrolled in the study. Three patients were excluded due to the suspension of surgery (n=2) and unplanned neck dissection (n=1). Therefore, 47 patients completed the study and were included in the final analysis (Figure 2). There were no differences between groups in comorbidities, morphometric and demographic characteristics, while women were predominant in our population (Table 1). Between ACTZ administration and the anesthesia induction, the mean time was the same in both groups (Control 113.7 ± 38.8 min vs ACTZ 113.1 ± 30.8 min, p=0.9). Duration of surgery and anesthesia were also similar between groups. Finally, both groups received the same total dose of remifentanil during surgery (Control 1186 ± 341.8 μg vs ACTZ 1286 ± 359.4 μg, p=0.4).

Figure 2 CONSORT diagram of patients’ recruitment and retention through the trial.

Table 1 Demographic And Clinical Characteristics Of Enrolled Patients

There were no differences in the baseline forearm MPT between patients receiving placebo and those receiving ACTZ (Figure 3). In the postoperative evaluations at 2 and 12–18 h, we did not find any differences in the forearm MPT between the groups. However, MPT in the forearm significantly diminished in the time in both groups (two way ANOVA, effect of time: F(2,44) = 6.8, p=0.02; effect of treatment: F(1,22) = 0.13, p=0.7; and effect of interaction: F(2,44) = 0.2, p = 0.2. Tukey post hoc test, Placebo: 2 h vs 12–18 h, p<0.05; ACTZ: baseline vs 12–18 h, p=0.001 and 2 h vs 12–18 h, p<0.05).

Figure 3 Mechanical pain threshold (MPT) measured using von Frey filaments at different times in both groups of patients. Values are presented as mean (95% CI) and statistical analyses was performed with two-way ANOVA and Tukey’s test for multiple comparisons. *p<0.05, ***p=0.001.

Finally, we found no differences between groups in the postoperative pain evaluated using the NPR scale (Figure 4A) or in morphine consumption (Figure 4B). Both were evaluated during the first 24 h after surgery. Total morphine consumption was 7.7 ± 6.7 mg in the control group and 7.6 ± 11.0 mg in the ACTZ group. There was no serious adverse event related to the ACTZ administration. Three patients reported headache in the ACTZ group (3/23, 13%) and one patient in the control group (1/24, 4%). The headaches were not severe in any case. There were also no differences in the incidence of postoperative nausea and vomiting (Control: 4/24 (16%) vs ACTZ: 5/23 (21%), p=0.9).

Figure 4 Pain and morphine consumption at different times in both groups of patients. (A) Postoperative pain scores using the numeric pain rating scale (NPRS). Values are presented as median (IQR). (B) Postoperative cumulative morphine consumption. Values are presented as mean (SD).

Discussion

This is the first clinical trial aimed to test whether a single preoperative dose of ACTZ prevents remifentanil-induced secondary hyperalgesia in patients undergoing total thyroidectomy. We found an intraoperative remifentanil-induced hyperalgesia, which was defined as a decreased in the MPT 12–18 h after surgery, but it was not prevented by ACTZ.

The rationale for using ACTZ comes mainly from results obtained in pre-clinical studies. ACTZ has been widely used in different clinical scenarios and exhibits a secure profile in terms of adverse effects, thus ACTZ was potentially an excellent agent to prevent remifentanil-induced hyperalgesia. However, our results indicate that ACTZ did not avoid remifentanil-induced hyperalgesia. On the other hand, remifentanil-mediated OIH occurred in both groups of patients, even when thyroidectomy is a short-duration surgery (approximately 1.5 h) and the total dose of remifentanil was less than in other studies.7,19 Remifentanil-induced secondary hyperalgesia has been previously reported by Schmidt et al.19 They observed that an acute remifentanil administration was associated with the induction of hyperalgesia to painful pressure stimuli in a different site to the surgery localization and our data show that this phenomenon occurs also in this short-duration surgery.

Several factors could explain our negative results. First, ACTZ dose may not have been enough to avoid OIH. A larger dose, multiple doses or other timing of administration could have been beneficial. Nonetheless, we define the dose (250 mg) and the timing (2 h before surgery) of ACTZ administration based on a pharmacokinetic study in healthy volunteers.20,21 The peak plasma concentration is reached after 2–3 h from oral administration and its elimination half-life is about 4–8 h.20 Indeed, a single 250 mg dose led to a plasma concentration above 5 μg·mL1 for at least 10 h since its administration in healthy Chilean volunteers21 which is a concentration in the therapeutic range to inhibit carbonic anhydrase.22 This plasma concentration is over the therapeutic range to inhibit carbonic anhydrase. Second, since the theoretical mechanism of ACTZ is based on restoring the Cl reversal potential in neurons, this drug will act once the chloride dysregulation has already been established. For the design of this trial, we expected that a single preoperative dose would be enough to prevent the Cl reversal potential after the surgery, based of its pharmacokinetics properties.21 However, since we failed to demonstrate the hypothesis with our trial design, future studies with another timing and dosages of ACTZ, including a postoperative administration (eg, when a change in MPT is actually observed), will be necessary to elucidate its potential role in reverting the OIH in humans. Indeed, in animals’ models ACTZ is administered when the OIH is established, and therefore its role is not to prevent its occurrence, instead it is to revert it.11 Third, it is possible that the KCC2-Chloride dysregulation mechanism of hyperalgesia may not be substantial in humans. Fourth, there is a potential confounder effect derived from morphine postoperative consumption, which is a potential limitation of this study. Morphine may probably affect pain perceptions compared with baseline measurements in areas distant to skin incision and possibly mask the OIH. However, in order to reduce this bias, we decided to conduct this trial in patients undergoing thyroidectomy, which has a mild surgical injury compared to other type of surgeries. In fact, morphine consumption was low in our patients and it principally occurred during the first postoperative hours. Fifth, our sample size could have been small to find a significant difference between groups. However, it was enough to detect a statically significant drop out in the MPT between the baseline and 12–18 h after surgery in a similar magnitude in both groups. Finally, the restoration of the MPT to the basal condition was not evaluated and it could have been different between the two groups.

Conclusion

In conclusion, both groups presented a reduction in the MPT in the forearm at 12–18 h compared to baseline, developing remifentanil-induced secondary hyperalgesia. However, ACTZ failed to prevent the MPT reduction in patients undergoing total thyroidectomy.

Data Sharing Statement

Individual participant data that underlie the results reported in this article, after deidentification will be shared upon request. The study protocol, statistical analysis plan, informed consent form and the analytic code will be also available immediately following publication and ending 5 years. This information will be shared to researches who provide a methodologically sound proposal to achieve the respective aims. Proposals should be directed to [email protected].

Acknowledgments

This project was supported by research fund from Laboratorios Saval in association with Faculty of Medicine, University of Chile (“Proyectos Saval de investigación 2015”) and by FONDECYT (Fondo Nacional de Desarrollo Científico y Tecnológico) de Iniciación 2015 Number 11150416.

Disclosure

The authors report no conflicts of interest in this work.

References

1. Angst M, Clark D. Opioid-induced hyperalgesia. Anesthesiology. 2006;104:570–587. doi:10.1097/00000542-200603000-00025

2. Chu LF, Angst MS, Clark D. Opioid-induced hyperalgesia in humans: molecular mechanisms and clinical considerations. Clin J Pain. 2008;24:479–496. doi:10.1097/AJP.0b013e31816b2f43

3. Fletcher D, Martinez V. Opioid-induced hyperalgesia in patients after surgery: a systematic review and a meta-analysis. Br J Anaesth. 2014;112:991–1004. doi:10.1093/bja/aeu137

4. Kim SH, Stoicea N, Soghomonyan S, et al. Intraoperative use of remifentanil and opioid induced hyperalgesia/acute opioid tolerance: systematic review. Front Pharmacol. 2014;5:108. doi:10.3389/fphar.2014.00108

5. Lee HC, Ryu HG, Kim HJ, et al. Excessive remifentanil during total intravenous anesthesia is associated with increased risk of pain after robotic thyroid surgery. PLoS ONE. 2018;13:e0209078. doi:10.1371/journal.pone.0209078

6. Park JH, Lee YC, Lee J, et al. The influence of high-dose intraoperative remifentanil on postoperative sore throat: a prospective randomized study: a CONSORT compliant article. Medicine (Baltimore). 2018;97:e13510. doi:10.1097/MD.0000000000013510

7. Joly V, Richebe P, Guignard B, et al. Remifentanil-induced postoperative hyperalgesia and its prevention with small-dose ketamine. Anesthesiology. 2005;103:147–155. doi:10.1097/00000542-200507000-00022

8. Hahnenkamp K, Nollet J, Van Aken HK, et al. Remifentanil directly activates human N-methyl-D-aspartate receptors expressed in Xenopus laevis oocytes. Anesthesiology. 2004;100:1531–1537. doi:10.1097/00000542-200406000-00028

9. Doyon N, Vinay L, Prescott SA, et al. Chloride regulation: a dynamic equilibrium crucial for synaptic inhibition. Neuron. 2016;89:1157–1172. doi:10.1016/j.neuron.2016.02.030

10. Gu W, Zhang W, Lei Y, et al. Activation of spinal alpha-7 nicotinic acetylcholine receptor shortens the duration of remifentanil-induced postoperative hyperalgesia by upregulating KCC2 in the spinal dorsal horn in rats. Mol Pain. 2017;13:1–16. doi:10.1177/1744806917704769

11. Ferrini F, Trang T, Mattioli TA, et al. Morphine hyperalgesia gated through microglia-mediated disruption of neuronal Cl homeostasis. Nat Neurosci. 2013;16:183–192. doi:10.1038/nn.3295

12. Staley KJ, Soldo BL, Proctor WR. Ionic mechanisms of neuronal excitation by inhibitory GABAA receptors. Science. 1995;269:977–981. doi:10.1126/science.7638623

13. Kaila K, Lamsa K, Smirnov S, et al. Long-lasting GABA-mediated depolarization evoked by high-frequency stimulation in pyramidal neurons of rat hippocampal slice is attributable to a network-driven, bicarbonate-dependent K+ transient. J Neurosci. 1997;17:7662–7672. doi:10.1523/JNEUROSCI.17-20-07662.1997

14. Kaila K, Voipio J, Paalasmaa P, et al. The role of bicarbonate in GABAA receptor-mediated IPSPs in rat neocortical neurons. J Physiol (Lond). 1993;464:273–289. doi:10.1113/jphysiol.1993.sp019634

15. Lee KY, Prescott SA. Chloride dysregulation and inhibitory receptor blockade yield equivalent disinhibition of spinal neurons yet are differentially reversed by carbonic anhydrase blockade. Pain. 2015;156:2431–2437. doi:10.1097/j.pain.0000000000000301

16. Asiedu M, Ossipov MH, Kaila K, et al. Acetazolamide and midazolam act synergistically to inhibit neuropathic pain. Pain. 2010;148:302–308. doi:10.1016/j.pain.2009.11.015

17. Echevarría G, Elgueta F, Fierro C, et al. Nitrous oxide (N2O) reduces postoperative opioid-induced hyperalgesia after remifentanil-propofol anaesthesia in humans. Br J Anaesth. 2011;107:959–965. doi:10.1093/bja/aer323

18. Minto CF, Schnider TW, Egan TD, 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

19. Schmidt S, Bethge C, Förster MH, et al. Enhanced postoperative sensitivity to painful pressure stimulation after intraoperative high dose remifentanil in patients without significant surgical site pain. Clin J Pain. 2007;23:605–611. doi:10.1097/AJP.0b013e318122d1e4

20. Granero GE, Longhi MR, Becker C, et al. Biowaiver monographs for immediate release solid oral dosage forms: acetazolamide. J Pharm Sci. 2008;97:3691–3699. doi:10.1002/jps.21282

21. Ritschel WA, Paulos C, Arancibia A, et al. Pharmacokinetics of acetazolamide in healthy volunteers after short- and long-term exposure to high altitude. J Clin Pharmacol. 1998;38:533–539. doi:10.1002/j.1552-4604.1998.tb05791.x

22. Aim A, Berggren L, Hartvig P, et al. Monitoring acetazolamide treatment. Acta Ophthalmol. 1982;60:24–34.

Creative Commons License © 2019 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.