Morphine- and buprenorphine-induced analgesia and antihyperalgesia in a human inflammatory pain model: a double-blind, randomized, placebo-controlled, five-arm crossover study

Pernille Ravn, Erik L Secher, Ulrik Skram, Trine Therkildsen, Lona L Christrup, Mads U Werner, Pernille Ravn, Erik L Secher, Ulrik Skram, Trine Therkildsen, Lona L Christrup, Mads U Werner

Abstract

Purpose: Opioid therapy is associated with the development of tolerance and paradoxically increased sensitivity to pain. It has been suggested that buprenorphine is associated with a higher antihyperalgesia/analgesia ratio than μ-opioid receptor agonists. The primary outcome of this study was therefore to investigate relative differences in antihyperalgesia and analgesia effects between morphine and buprenorphine in an inflammatory pain model in volunteers. The secondary outcome was to examine the relationship between pain sensitivity and opioid-induced effects on analgesia, antihyperalgesia, and descending pain modulation.

Subjects and methods: Twenty-eight healthy subjects were included. The study was a double-blind, randomized, placebo-controlled, five-arm crossover study with a multimodal (electrical, mechanical, and thermal stimuli) testing technique. After baseline assessments, intravenous infusions of morphine (10/20 mg), buprenorphine (0.3/0.6 mg), or placebo (normal saline) were administered over a 210-minute period, during which a cold pressor test, heat injury (47°C, 7 minutes, 12.5 cm(2)), and the first postburn assessment were done. After completion of the drug infusions, two additional postburn assessments were done. The subjects were monitored during each 8-hour session by an anesthesiologist.

Results: For nearly all tested variables, significant dose-dependent analgesic effects were demonstrated. The median antihyperalgesia/analgesia ratio (secondary hyperalgesia/heat injury relative to placebo) for low-dose morphine was 0.01 (interquartile range: -6.2; 9.9), 0.00 (-2.4; 2.1) for high-dose morphine, 0.03 (-1.8; 2.1) for low-dose buprenorphine, and 0.00 (-3.2; 1.1) for high-dose buprenorphine (P > 0.466). There were no significant differences in opioid responses between high and low pain-sensitive subjects (P > 0.286). High-dose buprenorphine, compared to placebo, was associated with a significantly enhanced action of the descending inhibitory pain control system (P = 0.004).

Conclusion: The present study, using multimodal testing technique, could not demonstrate any significant differences between morphine and buprenorphine in the profiles of antihyperalgesia and analgesia. Only high-dose buprenorphine was associated with a significant effect on the descending inhibitory pain control system.

Keywords: analgesia; antihyperalgesia; experimental pain; opioid; pain sensitivity; randomized trial.

Figures

Figure 1
Figure 1
Study algorithm (the order of the tests in the baseline assessments are as listed in the table). Abbreviations: CDT, cool detection threshold; EDT, electrical detection threshold; EPT, electrical pain threshold; EPTo, electrical pain tolerance; HPT, heat pain threshold; WDT, warmth detection threshold.
Figure 2
Figure 2
Participant flow-diagram. Notes: Participant flow-diagram of the study illustrating patient enrollment, inclusion, the two study days and analysis. The intention-to-treat (ITT) and per-protocol (PP) groups were 33/33 (females/males) and 13/14, respectively.
Figure 3
Figure 3
CPM-efficiency. Notes: The conditioned pain modulation (CPM) efficiency with repeated phasic heat stimuli (47°C, 4 s [1,3,4,5]) in relation to submersion of the non-dominant hand (2) in the cold pressor test (CPT [0.3°C–0.5°C, 30 s]). During the phasic heat stimuli, the subjects rated their maximal pain on a visual analog scale (VAS [0–10]).
Figure 4
Figure 4
Drug effect and drug like. Note: Drug effect and drug like scores for placebo, morphine and buprenorphine (low- and high-doses). *P < 0.005, **P < 0.001, ***P < 0.0001. Abbreviations: DE, drug effect; DL, drug like.
Figure 5
Figure 5
Electrical pain threshold and electrical pain tolerance. Notes: Electrical pain threshold and electrical pain tolerance for baseline, postburn 1–3 assessments and mean postburn assessments. The asterisks on baseline values indicates differences between baseline and its respective mean postburn value, while asterisks on mean postburn values indicate differences between the five sessions mean postburn assessments. Abbreviations: EPT, Electrical pain threshold; EPTo, electrical pain tolerance; Mean PB, mean of PB1, PB2 and PB3; PB1, 1 hour postburn; PB2, 2 hours postburn; PB3, 3 hours postburn.
Figure 6
Figure 6
Cool detection threshold and heat pain threshold. Notes: Cool Detection Threshold (CDT) and Heat Pain Threshold (HPT) for baseline, postburn 1–3 (PB1-3) and mean postburn (mean PB) assessments. The asterisks on baseline values indicate differences between baseline and its respective mean postburn value while asterisks on mean postburn values indicate differences between the five sessions mean postburn assessments. Abbreviations: CDT, cool detection threshold; HPT, heat pain threshold; Mean PB, mean of PB1, PB2 and PB3; PB1, 1 hour postburn; PB2, 2 hours postburn; PB3, 3 hours postburn.
Figure 7
Figure 7
Accumulated visual analog scale scores during heat injury. Notes: Accumulated VAS scores [0–10] during the heat injury. *P < 0.005, **P < 0.001, ***P < 0.0001. Abbreviation: VAS, visual analog scale.
Figure 8
Figure 8
Pressure algometry. Notes: Pressure pain tolerance before and after the cold pressor test, and delta pressure pain tolerance. *P < 0.005, **P < 0.001, ***P < 0.0001. Abbreviations: PTo1, pressure pain tolerance before CPT; PTo2, pressure pain tolerance after CPT; ΔPTo = PTo2 – PTo1.
Figure 9
Figure 9
Cold pressor test. Notes: Cold pressor test with registration of cold pressor pain and cold pressor tolerance. *P < 0.005, **P < 0.001, ***P < 0.0001. Abbreviations: CPP, cold pressor pain; CPT, cold pressor test; CPTo, cold pressor tolerance.
Figure 10
Figure 10
Conditioned pain modulation. Notes: Visual analog scale scores (0–10) during the test for CPM-efficiency. (Statistics only performed on ΔCPM). *P < 0.005, **P < 0.001, ***P < 0.0001. Abbreviations: CPM1, conditioned pain modulation rating before the cold pressor test; CPM2, conditioned pain modulation rating during the cold pressor test; CPM3, conditioned pain modulation rating 12 seconds after the cold pressor test; CPM4, conditioned pain modulation rating 24 seconds after the cold pressor test; ΔCPM, conditioned pain modulation efficiency; VAS, visual analog scale.
Figure 11
Figure 11
Secondary hyperalgesia areas. Note: Secondary hyperalgesia areas, in the three postburn assessments (PB1-3) and mean postburn values. Abbreviations: Mean PB, mean of PB1, PB2 and PB3; PB1, 1 hour postburn; PB2, 2 hours postburn; PB3, 3 hours postburn.
Figure 12
Figure 12
Anti-hyperalgesia and analgesia. Notes: Placebo-corrected differences for the active treatments in secondary hyperalgesia areas (Δ2HA[%], upper panel), pain-ratings during the heat injury (ΔPHI[%], middle panel), and electrical pain thresholds (ΔEPTo[%], lower panel). Columns are median (IQR). The anti-hyperalgesia/analgesia ratios (Δ2HA/ΔPHI and Δ2HA/ΔEPTo) for the four opioids relative to placebo. Abbreviations: Δ2HA(%), secondary hyperalgesia area relative to placebo; ΔPHI(%), heat injury relative to placebo; ΔEPTo(%), electrical pain tolerance relative to placebo; 2HA/PHI, ratio (secondary hyperalgesia/heat injury) relative to placebo; 2HA/EPTo, ratio (secondary hyperalgesia/electrical pain tolerance) relative to placebo; IQR, inter-quartile range

References

    1. Troster A, Ihmsen H, Singler B, Filitz J, Koppert W. Interaction of fentanyl and buprenorphine in an experimental model of pain and central sensitization in human volunteers. Clin J Pain. 2012;28(8):705–711.
    1. Angst MS, Clark JD. Opioid-induced hyperalgesia: a qualitative systematic review. Anesthesiology. 2006;104(3):570–587.
    1. Koppert W. Opioid-induced hyperalgesia – pathophysiology and clinical relevance. Acute Pain. 2007;9:21–34.
    1. Caroll IR, Angst MS, Clark JD. Management of perioperative pain in patients chronically consuming opioids. Reg Anesth Pain Med. 2004;29(6):576–591.
    1. Andresen T, Upton RN, Foster DJ, Christrup LL, Arendt-Nielsen L, Drewes AM. Pharmacokinetic/pharmacodynamic relationships of transdermal buprenorphine and fentanyl in experimental human pain models. Basic Clin Pharmacol Toxicol. 2011;108(4):274–284.
    1. Koppert W, Ihmsen H, Korber N, et al. Different profiles of buprenorphine-induced analgesia and antihyperalgesia in a human pain model. Pain. 2005;118(1–2):15–22.
    1. Angst MS, Phillips NG, Drover DR, et al. Pain sensitivity and opioid analgesia: a pharmacogenomic twin study. Pain. 2012;153(7):1397–1409.
    1. Abrishami A, Chan J, Chung F, Wong J. Preoperative pain sensitivity and its correlation with postoperative pain and analgesic consumption: a qualitative systematic review. Anesthesiology. 2011;114(2):445–457.
    1. Werner MU, Mjobo HN, Nielsen PR, Rudin A. Prediction of postoperative pain: a systematic review of predictive experimental pain studies. Anesthesiology. 2010;112(6):1494–1502.
    1. Clauw DJ, Williams D, Lauerman W, et al. Pain sensitivity as a correlate of clinical status in individuals with chronic low back pain. Spine (Phila Pa 1976) 1999;24(19):2035–2041.
    1. King CD, Wong F, Currie T, Mauderli AP, Fillingim RB, Riley JL., 3rd Deficiency in endogenous modulation of prolonged heat pain in patients with irritable bowel syndrome and temporomandibular disorder. Pain. 2009;143(3):172–178.
    1. Arendt-Nielsen L, Andresen T, Malver LP, Oksche A, Mansikka H, Drewes AM. A double-blind, placebo-controlled study on the effect of buprenorphine and fentanyl on descending pain modulation: a human experimental study. Clin J Pain. 2011;28(7):623–627.
    1. Yarnitsky D, Arendt-Nielsen L, Bouhassira D, et al. Recommendations on terminology and practice of psychophysical DNIC testing. Eur J Pain. 2010;14(4):339.
    1. Naert AL, Kehlet H, Kupers R. Characterization of a novel model of tonic heat pain stimulation in healthy volunteers. Pain. 2008;138(1):163–171.
    1. Pedersen JL, Kehlet H. Hyperalgesia in a human model of acute inflammatory pain: a methodological study. Pain. 1998;74(2–3):139–151.
    1. Werner MU, Duun P, Kehlet H. Prediction of postoperative pain by preoperative nociceptive responses to heat stimulation. Anesthesiology. 2004;100(1):115–119.
    1. Ravn P, Frederiksen R, Skovsen AP, Christrup LL, Werner MU. Prediction of pain sensitivity in healthy volunteers. J Pain Res. 2012;5:313–326.
    1. Yassen A, Olofsen E, Romberg R, Sarton E, Danhof M, Dahan A. Mechanism-based pharmacokinetic–pharmacodynamic modeling of the antinociceptive effect of buprenorphine in healthy volunteers. Anesthesiology. 2006;104(6):1232–1242.
    1. Mazoit JX, Butscher K, Samii K. Morphine in postoperative patients: pharmacokinetics and pharmacodynamics of metabolites. Anesth Analg. 2007;105(1):70–78.
    1. Gan TJ, Meyer TA, Apfel CC, et al. Society for Ambulatory Anesthesia guidelines for the management of postoperative nausea and vomiting. Anesth Analg. 2007;105(6):1615–1628.
    1. Bjelland I, Dahl AA, Haug TT, Neckelmann D. The validity of the Hospital Anxiety and Depression Scale. An updated literature review. J Psychosom Res. 2002;52(2):69–77.
    1. Sullivan MJL, Bishop SR, Kivik J. The pain catastrophizing scale: development and validation. Psychol Assess. 1995;7(4):524–532.
    1. Borly L, Anderson IB, Bardram L, et al. Preoperative prediction model of outcome after cholecystectomy for symptomatic gallstones. Scand J Gastroenterol. 1999;34(11):1144–1152.
    1. Jorgensen T, Teglbjerg JS, Wille-Jorgensen P, Bille T, Thorvaldsen P. Persisting pain after cholecystectomy. A prospective investigation. Scand J Gastroenterol. 1991;26(1):124–128.
    1. Zigmond AS, Snaith RP. The hospital anxiety and depression scale. Acta Psychiatr Scand. 1983;67(6):361–370.
    1. Bisgaard T, Klarskov B, Rosenberg J, Kehlet H. Characteristics and prediction of early pain after laparoscopic cholecystectomy. Pain. 2001;90(3):261–269.
    1. Gill DM, Reddon JR, Stefanyk WO, Hans HS. Finger tapping: effects of trials and sessions. Percept Mot Skills. 1986;62(2):675–678.
    1. Zacny JP, Gutierrez S. Subjective, psychomotor, and physiological effects profile of hydrocodone/acetaminophen and oxycodone/acetaminophen combination products. Pain Med. 2008;9(4):433–443.
    1. Stener-Victorin E, Kowalski J, Lundeberg T. A new highly reliable instrument for the assessment of pre- and postoperative gynecological pain. Anesth Analg. 2002;95(1):151–157.
    1. Moller KA, Johansson B, Berge OG. Assessing mechanical allodynia in the rat paw with a new electronic algometer. J Neurosci Methods. 1998;84(1–2):41–47.
    1. Werner MU, Rotboll-Nielsen P, Ellehuus-Hilmersson C. Humidity affects the performance of von Frey monofilaments. Acta Anaesthesiol Scand. 2011;55(5):577–582.
    1. Pud D, Granovsky Y, Yarnitsky D. The methodology of experimentally induced diffuse noxious inhibitory control (DNIC)-like effect in humans. Pain. 2009;144(1–2):16–19.
    1. Yarnitsky D, Crispel Y, Eisenberg E, et al. Prediction of chronic post-operative pain: pre-operative DNIC testing identifies patients at risk. Pain. 2008;138(1):22–28.
    1. Eisenberg E, Midbari A, Haddad M, Pud D. Predicting the analgesic effect to oxycodone by “static” and “dynamic” quantitative sensory testing in healthy subjects. Pain. 2010;151(1):104–109.
    1. Nielsen CS, Stubhaug A, Price DD, Vassend O, Czajkowski N, Harris JR. Individual differences in pain sensitivity: genetic and environmental contributions. Pain. 2008;136(1–2):21–29.
    1. Verbraecken J, Van de Heyning P, De Backer W, Van Gaal L. Body surface area in normal-weight, overweight, and obese adults. A comparison study. Metabolism. 2006;55(4):515–524.
    1. Koppert W, Angst M, Alsheimer M, et al. Naloxone provokes similar pain facilitation as observed after short-term infusion of remifentanil in humans. Pain. 2003;106(1–2):91–99.
    1. Pedersen JL, Kehlet H. Secondary hyperalgesia to heat stimuli after burn injury in man. Pain. 1998;76(3):377–384.
    1. Weidner C, Schmelz M, Schmidt R, Hansson B, Handwerker HO, Torebjork HE. Functional attributes discriminating mechano-insensitive and mechano-responsive C nociceptors in human skin. J Neurosci. 1999;19(22):10184–10190.
    1. Brennum J, Arendt-Nielsen L, Horn A, Secher NH, Jensen TS. Quantitative sensory examination during epidural anaesthesia and analgesia in man: effects of morphine. Pain. 1993;52(1):75–83.
    1. Lilleso J, Hammer NA, Pedersen JL, Kehlet H. Effect of peripheral morphine in a human model of acute inflammatory pain. Br J Anaesth. 2000;85(2):228–232.
    1. Warncke T, Stubhaug A, Jorum E. Ketamine, an NMDA receptor antagonist, suppresses spatial and temporal properties of burn-induced secondary hyperalgesia in man: a double-blind, cross-over comparison with morphine and placebo. Pain. 1997;72(1–2):99–106.
    1. Warncke T, Stubhaug A, Jorum E. Preinjury treatment with morphine or ketamine inhibits the development of experimentally induced secondary hyperalgesia in man. Pain. 2000;86(3):293–303.
    1. Werner MU, Pedersen KL, Rowbotham MC, Dahl JB. Can healthy volunteers be phenotyped using cutaneous sensitization models? 2012 Forthcoming.
    1. Klauenberg S, Maier C, Assion HJ, et al. Depression and changed pain perception: hints for a central disinhibition mechanism. Pain. 2008;140(2):332–343.
    1. Wang H, Fischer C, Chen G, Weinsheimer N, Gantz S, Schiltenwolf M. Does long-term opioid therapy reduce pain sensitivity of patients with chronic low back pain? Evidence from quantitative sensory testing. Pain Physician. 2012;15(Suppl 3):ES135–143.
    1. Werner MU, Perkins FM, Holte K, Pedersen JL, Kehlet H. Effects of gabapentin in acute inflammatory pain in humans. Reg Anesth Pain Med. 2001;26(4):322–328.
    1. Brennum J, Dahl JB, Moiniche S, Arendt-Nielsen L. Quantitative sensory examination of epidural anaesthesia and analgesia in man: effects of pre- and post-traumatic morphine on hyperalgesia. Pain. 1994;59(2):261–271.
    1. Park R, Wallace MS, Schulteis G. Relative sensitivity to alfentanil and reliability of current perception threshold vs von Frey tactile stimulation and thermal sensory testing. J Peripher Nerv Syst. 2001;6(4):232–240.
    1. Leung A, Wallace MS, Ridgeway B, Yaksh T. Concentration–effect relationship of intravenous alfentanil and ketamine on peripheral neurosensory thresholds, allodynia and hyperalgesia of neuropathic pain. Pain. 2001;91(1–2):177–187.
    1. Cowan A. Buprenorphine: new pharmacological aspects. Int J Clin Pract Suppl. 2003;133:3–8.
    1. Kuhlman JJ, Jr, Lalani S, Magluilo J, Jr, Levine B, Darwin WD. Human pharmacokinetics of intravenous, sublingual, and buccal buprenorphine. J Anal Toxicol. 1996;20(6):369–378.
    1. Belcheva MM, Barg J, McHale RJ, et al. Differential down- and up-regulation of rat brain opioid receptor types and subtypes by buprenorphine. Mol Pharmacol. 1993;44(1):173–179.
    1. Twycross RG. Opioid analgesics in cancer pain: current practice and controversies. Cancer Surv. 1988;7(1):29–53.
    1. Sittl R. Transdermal buprenorphine in cancer pain and palliative care. Palliat Med. 2006;20(Suppl 1):s25–s30.
    1. Andresen T, Staahl C, Oksche A, Mansikka H, Arendt-Nielsen L, Drewes AM. Effect of transdermal opioids in experimentally induced superficial, deep and hyperalgesic pain. Br J Pharmacol. 2011;164(3):934–945.
    1. Yarnitsky D. Conditioned pain modulation (the diffuse noxious inhibitory control-like effect): its relevance for acute and chronic pain states. Curr Opin Anaesthesiol. 2010;23(5):611–615.
    1. Julien N, Goffaux P, Arsenault P, Marchand S. Widespread pain in fibromyalgia is related to a deficit of endogenous pain inhibition. Pain. 2005;114(1–2):295–302.
    1. Johannesson U, de Boussard CN, Brodda Jansen G, Bohm-Starke N. Evidence of diffuse noxious inhibitory controls (DNIC) elicited by cold noxious stimulation in patients with provoked vestibulodynia. Pain. 2007;130(1–2):31–39.
    1. Olesen SS, Brock C, Krarup AL, et al. Descending inhibitory pain modulation is impaired in patients with chronic pancreatitis. Clin Gastroenterol Hepatol. 2010;8(8):724–730.
    1. Le Bars D, Willer JC, De Broucker T. Morphine blocks descending pain inhibitory controls in humans. Pain. 1992;48(1):13–20.
    1. Willer JC, Le Bars D, De Broucker T. Diffuse noxious inhibitory controls in man: involvement of an opioidergic link. Eur J Pharmacol. 1990;182(2):347–355.
    1. Ram KC, Eisenberg E, Haddad M, Pud D. Oral opioid use alters DNIC but not cold pain perception in patients with chronic pain – new perspective of opioid-induced hyperalgesia. Pain. 2008;139(2):431–438.
    1. Nielsen CS, Staud R, Price DD. Individual differences in pain sensitivity: measurement, causation, and consequences. J Pain. 2009;10(3):231–237.
    1. Kindler LL, Valencia C, Fillingim RB, George SZ. Sex differences in experimental and clinical pain sensitivity for patients with shoulder pain. Eur J Pain. 2011;15(2):118–123.
    1. Petersen-Felix S, Arendt-Nielsen L, Bak P, et al. Ondansetron does not inhibit the analgesic effect of alfentanil. Br J Anaesth. 1994;73(3):326–330.
    1. Stubhaug A, Romundstad L, Kaasa T, Breivik H. Methylprednisolone and ketorolac rapidly reduce hyperalgesia around a skin burn injury and increase pressure pain thresholds. Acta Anaesthesiol Scand. 2007;51(9):1138–1146.
    1. Greffrath W, Baumgartner U, Treede RD. Peripheral and central components of habituation of heat pain perception and evoked potentials in humans. Pain. 2007;132(3):301–311.
    1. Granot M, Weissman-Fogel I, Crispel Y, et al. Determinants of endogenous analgesia magnitude in a diffuse noxious inhibitory control (DNIC) paradigm: do conditioning stimulus painfulness, gender and personality variables matter? Pain. 2008;136(1–2):142–149.
    1. Yarnitsky D, Granot M, Nahman-Averbuch H, Khamaisi M, Granovsky Y. Conditioned pain modulation predicts duloxetine efficacy in painful diabetic neuropathy. Pain. 2012;153(6):1193–1198.
    1. Treister R, Eisenberg E, Gershon E, Haddad M, Pud D. Factors affecting – and relationships between – different modes of endogenous pain modulation in healthy volunteers. Eur J Pain. 2010;14(6):608–614.

Source: PubMed

Подписаться