Ketamine coadministration attenuates morphine tolerance and leads to increased brain concentrations of both drugs in the rat

T O Lilius, V Jokinen, M S Neuvonen, M Niemi, E A Kalso, P V Rauhala, T O Lilius, V Jokinen, M S Neuvonen, M Niemi, E A Kalso, P V Rauhala

Abstract

Background and purpose: The effects of ketamine in attenuating morphine tolerance have been suggested to result from a pharmacodynamic interaction. We studied whether ketamine might increase brain morphine concentrations in acute coadministration, in morphine tolerance and morphine withdrawal.

Experimental approach: Morphine minipumps (6 mg·day(-1) ) induced tolerance during 5 days in Sprague-Dawley rats, after which s.c. ketamine (10 mg·kg(-1) ) was administered. Tail flick, hot plate and rotarod tests were used for behavioural testing. Serum levels and whole tissue brain and liver concentrations of morphine, morphine-3-glucuronide, ketamine and norketamine were measured using HPLC-tandem mass spectrometry.

Key results: In morphine-naïve rats, ketamine caused no antinociception whereas in morphine-tolerant rats there was significant antinociception (57% maximum possible effect in the tail flick test 90 min after administration) lasting up to 150 min. In the brain of morphine-tolerant ketamine-treated rats, the morphine, ketamine and norketamine concentrations were 2.1-, 1.4- and 3.4-fold, respectively, compared with the rats treated with morphine or ketamine only. In the liver of morphine-tolerant ketamine-treated rats, ketamine concentration was sixfold compared with morphine-naïve rats. After a 2 day morphine withdrawal period, smaller but parallel concentration changes were observed. In acute coadministration, ketamine increased the brain morphine concentration by 20%, but no increase in ketamine concentrations or increased antinociception was observed.

Conclusions and implications: The ability of ketamine to induce antinociception in rats made tolerant to morphine may also be due to increased brain concentrations of morphine, ketamine and norketamine. The relevance of these findings needs to be assessed in humans.

© 2014 The Authors. British Journal of Pharmacology published by John Wiley & Sons Ltd on behalf of The British Pharmacological Society.

Figures

Figure 1
Figure 1
The design of the morphine tolerance and withdrawal experiments. In experiment sets 1 (blue) and 2 (red), 48 and 24 Sprague–Dawley rats were used respectively.
Figure 2
Figure 2
Development of antinociceptive tolerance after morphine minipump implantations. On day 0, minipumps delivering morphine 6 mg·day–1 or vehicle were implanted s.c. under brief isoflurane anaesthesia. The means (±SEM) of the tail flick (A) and hot plate (B) latencies are shown after 1, 3 and 5 days of pump implantation. The mean raw nociceptive latencies are presented to allow assessment of the data quality. No extra morphine doses were given before the behavioural measurements. *P < 0.05, ***P < 0.001; significantly different from the vehicle control group; n = 8 in the vehicle group and n = 16 in the morphine group.
Figure 3
Figure 3
Effects of an acute low dose of ketamine on antinociception in morphine-tolerant rats under chronic morphine treatment. Rats under an ongoing morphine (6 mg·day–1, Mo) or vehicle (Veh) pump treatment received an acute s.c. dose of ketamine (10 mg·kg–1, Ket) or vehicle (Veh) on day 5. Antinociception was measured using tail flick (A) and hot plate (B) tests. The mean of the maximum possible effect (MPE%) ±SEM is plotted. In the rotarod test (C), the mean (±SEM) survival time (seconds) is plotted. From separate animals having had the same pretreatments, whole brain, serum and liver samples were collected after 90 min of ketamine administration. The tissue concentrations and serum levels of the experimental drugs and their main metabolites (morphine, MO; M3G; ketamine, KET; norketamine, NORKET) in the tissues were quantified and the means of the groups (±SEM) are presented in graphs D–I. The percentage difference between the treatment groups is shown for each compound. n = 8. **P < 0.01, ***P < 0.001; significantly different from the vehicle control group. #P < 0.05, ##P < 0.01, ###P < 0.001; significantly different from the morphine pump treated group that received acute vehicle. §§P < 0.01, §§§P < 0.001; significantly different from the vehicle-pretreated group that received acute ketamine.
Figure 4
Figure 4
Effects of an acute low dose of ketamine, morphine and their combination on antinociception in morphine-tolerant rats under morphine withdrawal. Rats had morphine pumps that delivered morphine (Mo) or vehicle (Veh) 6 mg·day–1 for 5 days in total. On the evening of day 5, the morphine pumps were removed. On day 7, rats in morphine withdrawal received s.c. doses of morphine, ketamine or a combination. Control animals received vehicle or ketamine. Morphine was administered 15 min before ketamine. Antinociception was measured using tail flick (A) and hot plate (B) tests. The mean of the maximum possible effect (MPE%) ±SEM is plotted. In the rotarod test (C), the mean (±SEM) survival time (seconds) is plotted. Whole brain, serum and liver samples were collected after 90 min of ketamine administration. The tissue concentrations and serum levels of the experimental drugs and their main metabolites (morphine, MO; M3G; ketamine, KET; norketamine, NORKET) in the tissues were quantified and the means of the groups (±SEM) are presented in graphs D–I. The percentage difference between the treatment groups is shown for each compound. n = 8. *P < 0.05, **P < 0.01, ***P < 0.001; significantly different from the vehicle control group. #P < 0.05, ###P < 0.001; significantly different from the morphine pump treated group that received acute morphine only. §P < 0.05, §§P < 0.01, §§§P < 0.001; significantly different from the vehicle-pretreated group that received acute ketamine.
Figure 5
Figure 5
Effects of an acute low dose of ketamine on antinociception in acute coadministration with morphine. Rats received an acute s.c. dose of ketamine (10 mg·kg–1, Ket) or vehicle (Veh). Morphine was given 15 min before ketamine. Antinociception was measured using tail flick (A) and hot plate (B) tests. The mean of the maximum possible effect (MPE%) ±SEM is plotted. In the rotarod test (C), the mean (±SEM) survival time (seconds) is plotted. Whole brain, serum and liver samples were collected after 90 min of ketamine administration. The tissue concentrations and serum levels of the experimental drugs and their main metabolites (morphine, MO; M3G; ketamine, KET; norketamine, NORKET) in the tissues were quantified and the means of the groups (±SEM) are presented in graphs D–I. The percentage difference between the treatment groups is shown for each compound. n = 8, except n = 7 in the morphine-treated group. *P < 0.05, **P < 0.01, ***P < 0.001; significantly different from the vehicle control group. #P < 0.05, ##P < 0.01, ###P < 0.001; significantly different from morphine only. §P < 0.05, §§§P < 0.001; significantly different from acute ketamine only.

References

    1. Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, CGTP Collaborators et al. The Concise Guide to PHARMACOLOGY 2013/14: Ligand-gated ion channels. Br J Pharmacol. 2013a;170:1582–1606.
    1. Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The Concise Guide to PHARMACOLOGY 2013/14: Enzymes. Br J Pharmacol. 2013b;170:1797–1867.
    1. Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The Concise Guide to PHARMACOLOGY 2013/14: Transporters. Br J Pharmacol. 2013c;170:1706–1796.
    1. Amphoux A, Vialou V, Drescher E, Brüss M, La Cour CM, Rochat C, et al. Differential pharmacological in vitro properties of organic cation transporters and regional distribution in rat brain. Neuropharmacology. 2006;50:941–952.
    1. Arroyo-Novoa CM, Figueroa-Ramos MI, Miaskowski C, Padilla G, Paul SM, Rodríguez-Ortiz P, et al. Efficacy of small doses of ketamine with morphine to decrease procedural pain responses during open wound care. Clin J Pain. 2011;27:561–566.
    1. Bell RF. Low-dose subcutaneous ketamine infusion and morphine tolerance. Pain. 1999;83:101–103.
    1. Bell RF. Ketamine for chronic noncancer pain: concerns regarding toxicity. Curr Opin Support Palliat Care. 2012;6:183–187.
    1. Bell RF, Dahl JB, Moore RA, Kalso EA. Perioperative ketamine for acute postoperative pain. Cochrane Database Syst Rev. 2006;(1) CD004603.
    1. Bell RF, Eccleston C, Kalso EA. Ketamine as an adjuvant to opioids for cancer pain. Cochrane Database Syst Rev. 2012;(11) CD003351.
    1. Caraceni A, Hanks G, Kaasa S, Bennett MI, Brunelli C, Cherny N, et al. Use of opioid analgesics in the treatment of cancer pain: evidence-based recommendations from the EAPC. Lancet Oncol. 2012;13:e58–e68.
    1. Coffman BL, Rios GR, Tephly TR. Purification and properties of two rat liver phenobarbital-inducible UDP-glucuronosyltransferases that catalyze the glucuronidation of opioids. Drug Metab Dispos. 1996;24:329–333.
    1. Coffman BL, Rios GR, King CD, Tephly TR. Human UGT2B7 catalyzes morphine glucuronidation. Drug Metab Dispos. 1997;25:1–4.
    1. Doan KMM. Passive permeability and P-glycoprotein-mediated efflux differentiate central nervous system (CNS) and non-CNS marketed drugs. J Pharmacol Exp Ther. 2002;303:1029–1037.
    1. Dworkin RH, O'Connor AB, Backonja M, Farrar JT, Finnerup NB, Jensen TS, et al. Pharmacologic management of neuropathic pain: evidence-based recommendations. Pain. 2007;132:237–251.
    1. Ebert B, Mikkelsen S, Thorkildsen C, Borgbjerg FM. Norketamine, the main metabolite of ketamine, is a non-competitive NMDA receptor antagonist in the rat cortex and spinal cord. Eur J Pharmacol. 1997;333:99–104.
    1. Edwards SR, Minto CF, Mather LE. Concurrent ketamine and alfentanil administration: pharmacokinetic considerations. Br J Anaesth. 2002;88:94–100.
    1. González P, Cabello P, Germany A, Norris B, Contreras E. Decrease of tolerance to physical dependence on morphine by, glutamate receptor antagonists. Eur J Pharmacol. 1997;332:257–262.
    1. Grant IS, Nimmo WS, Clements JA. Pharmacokinetics and analgesic effects of i.m. and oral ketamine. Br J Anaesth. 1981;53:805–810.
    1. Hardy J, Quinn S, Fazekas B, Plummer J, Eckermann S, Agar M, et al. Randomized, double-blind, placebo-controlled study to assess the efficacy and toxicity of subcutaneous ketamine in the management of cancer pain. J Clin Oncol. 2012;30:3611–3617.
    1. Hassan HE, Myers AL, Lee IJ, Mason CW, Wang D, Sinz MW, et al. Induction of xenobiotic receptors, transporters, and drug metabolizing enzymes by oxycodone. Drug Metab Dispos. 2013;41:1060–1069.
    1. Hijazi Y. Contribution of CYP3A4, CYP2B6, and CYP2C9 isoforms to N-demethylation of ketamine in human liver microsomes. Drug Metab Dispos. 2002;30:853–858.
    1. Holtman JR, Crooks PA, Johnson-Hardy JK, Hojomat M, Kleven M, Wala EP. Effects of norketamine enantiomers in rodent models of persistent pain. Pharmacol Biochem Behav. 2008a;90:676–685.
    1. Holtman JR, Crooks P, Johnson-Hardy JK, Wala E. Interaction between morphine and norketamine enantiomers in rodent models of nociception. Pharmacol Biochem Behav. 2008b;90:769–777.
    1. Honarmand A, Safavi M, Karaky H. Preincisional administration of intravenous or subcutaneous infiltration of low-dose ketamine suppresses postoperative pain after appendectomy. J Pain Res. 2012;5:1–6.
    1. Kavalali ET, Monteggia LM. Synaptic mechanisms underlying rapid antidepressant action of ketamine. Am J Psychiatry. 2012;169:1150–1156.
    1. Kilkenny C, Browne W, Cuthill IC, Emerson M, Altman DG. Animal research: reporting in vivo experiments: the ARRIVE guidelines. Br J Pharmacol. 2010;160:1577–1579.
    1. King CD, Rios GR, Green MD, MacKenzie PI, Tephly TR. Comparison of stably expressed rat UGT1.1 and UGT2B1 in the glucuronidation of opioid compounds. Drug Metab Dispos. 1997;25:251–255.
    1. Laskowski K, Stirling A, McKay WP, Lim HJ. A systematic review of intravenous ketamine for postoperative analgesia. Can J Anesth. 2011;58:911–923.
    1. Letrent SP, Pollack GM, Brouwer KR, Brouwer KL. Effects of a potent and specific P-glycoprotein inhibitor on the blood-brain barrier distribution and antinociceptive effect of morphine in the rat. Drug Metab Dispos. 1999;27:827–834.
    1. Li Y, Coller JK, Hutchinson MR, Klein K, Zanger UM, Stanley NJ, et al. The CYP2B6*6 allele significantly alters the N-demethylation of ketamine enantiomers in vitro. Drug Metab Dispos. 2013;41:1264–1272.
    1. Mathews TJ, Churchhouse AMD, Housden T, Dunning J. Does adding ketamine to morphine patient-controlled analgesia safely improve post-thoracotomy pain? Interact Cardiovasc Thorac Surg. 2012;14:194–199.
    1. McGrath J, Drummond G, McLachlan E, Kilkenny C, Wainwright C. Guidelines for reporting experiments involving animals: the ARRIVE guidelines. Br J Pharmacol. 2010;160:1573–1576.
    1. Miyamoto H, Saito Y, Kirihara Y, Hara K, Sakura S, Kosaka Y. Spinal coadministration of ketamine reduces the development of tolerance to visceral as well as somatic antinociception during spinal morphine infusion. Anesth Analg. 2000;90:136–141.
    1. Morgan CJA, Curran HV Independent Scientific Committee on Drugs. Ketamine use: a review. Addiction. 2012;107:27–38.
    1. Nesher N, Ekstein MP, Paz Y, Marouani N, Chazan S, Weinbroum AA. Morphine with adjuvant ketamine vs higher dose of morphine alone for immediate postthoracotomy analgesia. Chest. 2009;136:245–252.
    1. Pawson AJ, Sharman JL, Benson HE, Faccenda E, Alexander SP, Buneman OP, et al. NC-IUPHAR. The IUPHAR/BPS Guide to PHARMACOLOGY: an expert-driven knowledge base of drug targets and their ligands. Nucl. Acids Res. 2014;42(Database Issue):D1098–D1106.
    1. Pritchard M, Fournel-Gigleux S, Siest G, Mackenzie P, Magdalou J. A recombinant phenobarbital-inducible rat liver UDP-glucuronosyltransferase (UDP-glucuronosyltransferase 2B1) stably expressed in V79 cells catalyzes the glucuronidation of morphine, phenols, and carboxylic acids. Mol Pharmacol. 1994;45:42–50.
    1. Qi X, Evans AM, Wang J, Miners JO, Upton RN, Milne RW. Inhibition of morphine metabolism by ketamine. Drug Metab Dispos. 2010;38:728–731.
    1. Salas S, Frasca M, Planchet-Barraud B, Burucoa B, Pascal M, Lapiana J-M, et al. Ketamine analgesic effect by continuous intravenous infusion in refractory cancer pain: considerations about the clinical research in palliative care. J Palliat Med. 2012;15:287–293.
    1. Schinkel AH, Wagenaar E, van Deemter L, Mol CA, Borst P. Absence of the mdr1a P-Glycoprotein in mice affects tissue distribution and pharmacokinetics of dexamethasone, digoxin, and cyclosporin A. J Clin Invest. 1995;96:1698–1705.
    1. Sharma HS, Ali SF. Alterations in blood-brain barrier function by morphine and methamphetamine. Ann N Y Acad Sci. 2006;1074:198–224.
    1. Shimoyama M, Shimoyama N, Gorman AL, Elliott KJ, Inturrisi CE. Oral ketamine is antinociceptive in the rat formalin test: role of the metabolite, norketamine. Pain. 1999;81:85–93.
    1. Shimoyama N, Shimoyama M, Inturrisi CE, Elliott KJ. Ketamine attenuates and reverses morphine tolerance in rodents. Anesthesiology. 1996;85:1357–1366.
    1. Sinner B, Graf BM. Ketamine. In: Schuttler J, Schwilden H, editors. Modern Anesthetics. Berlin Heidelberg: Springer; 2008. pp. 313–333. . In: (eds).
    1. Su W, Pasternak GW. The role of multidrug resistance-associated protein in the blood-brain barrier and opioid analgesia. Synapse. 2013;67:609–619.
    1. Suppa E, Valente A, Catarci S, Zanfini B, Draisci G. A study of low-dose S-Ketamine infusion as ‘preventive’ pain treatment for cesarean section with spinal anesthesia: benefits and side effects. Minerva Anestesiol. 2012;7:774–781.
    1. Trujillo KA, Akil H. Inhibition of morphine tolerance and dependence by the NMDA receptor antagonist MK-801. Science. 1991;251:85–87.
    1. Trujillo KA, Akil H. Inhibition of opiate tolerance by non-competitive N-d-aspartate receptor antagonists. Brain Res. 1994;633:178–188.
    1. Tzvetkov MV, dos Santos Pereira JN, Meineke I, Saadatmand AR, Stingl JC, Brockmöller J. Morphine is a substrate of the organic cation transporter OCT1 and polymorphisms in OCT1 gene affect morphine pharmacokinetics after codeine administration. Biochem Pharmacol. 2013;86:666–678.
    1. Uchaipichat V, Raungrut P, Chau N, Janchawee B, Evans AM, Miners JO. Effects of ketamine on human UDP-glucuronosyltransferases in vitro predict potential drug-drug interactions arising from ketamine inhibition of codeine and morphine glucuronidation. Drug Metab Dispos. 2011;39:1324–1328.
    1. Vachon P, Hélie P, Veilleux-Lemieux D, Beaudry F. Effects of endotoxemia on the pharmacodynamics and pharmacokinetics of ketamine and xylazine anesthesia in Sprague–Dawley rats. Vet Med Res Rep. 2012;3:99–109.
    1. Varma MVS, Sateesh K, Panchagnula R. Functional role of p-glycoprotein in limiting intestinal absorption of drugs: contribution of passive permeability to P-glycoprotein mediated efflux transport. Mol Pharmaceutics. 2005;2:12–21.
    1. Visser E, Schug SA. The role of ketamine in pain management. Biomed Pharmacother. 2006;60:341–348.
    1. White PF. Clinical pharmacology of intravenous induction drugs. Int Anesthesiol Clin. 1988;26:98–104.
    1. Xie R, Hammarlund-Udenaes M, de Boer AG, de Lange EC. The role of P-glycoprotein in blood-brain barrier transport of morphine: transcortical microdialysis studies in mdr1a (–/–) and mdr1a (+/+) mice. Br J Pharmacol. 1999;128:563–568.
    1. Zheng M, McErlane KM, Ong MC. High-performance liquid chromatography-mass spectrometry-mass spectrometry analysis of morphine and morphine metabolites and its application to a pharmacokinetic study in male Sprague-Dawley rats. J Pharm Biomed Anal. 1998;16:971–980.
    1. Zimmermann M. Ethical guidelines for investigations of experimental pain in conscious animals. Pain. 1983;16:109–110.

Source: PubMed

3
구독하다