Evaluation of anti-hyperalgesic and analgesic effects of two benzodiazepines in human experimental pain: a randomized placebo-controlled study

Pascal H Vuilleumier, Marie Besson, Jules Desmeules, Lars Arendt-Nielsen, Michele Curatolo, Pascal H Vuilleumier, Marie Besson, Jules Desmeules, Lars Arendt-Nielsen, Michele Curatolo

Abstract

Background and aims: Compounds that act on GABA-receptors produce anti-hyperalgesia in animal models, but little is known on their effects in humans. The aim of this study was to explore the potential usefulness of GABA-agonism for the control of pain in humans. Two agonists at the benzodiazepine-binding site of GABAA-receptors (clobazam and clonazepam) were studied using multiple experimental pain tests. Positive results would support further investigation of GABA agonism for the control of clinical pain.

Methods: In a randomized double-blind crossover design, 16 healthy male volunteers received clobazam 20 mg, clonazepam 1 mg and tolterodine 1 mg (active placebo). The area of static hyperalgesia after intradermal capsaicin injection was the primary endpoint. Secondary endpoints were: area of dynamic hyperalgesia, response to von Frey hair stimulation, pressure pain thresholds, conditioned pain modulation, cutaneous and intramuscular electrical pain thresholds (1, 5 and 20 repeated stimulation), and pain during cuff algometry.

Results: For the primary endpoint, an increase in the area of static hyperalgesia was observed after administration of placebo (p<0.001), but not after clobazam and clonazepam. Results suggestive for an anti-hyperalgesic effect of the benzodiazepines were obtained with all three intramuscular pain models and with cuff algometry. No effect could be detected with the other pain models employed.

Conclusions: Collectively, the results are suggestive for a possible anti-hyperalgesic effect of drugs acting at the GABAA-receptors in humans, particularly in models of secondary hyperalgesia and deep pain. The findings are not conclusive, but support further clinical research on pain modulation by GABAergic drugs. Because of the partial results, future research should focus on compounds acting selectively on subunits of the GABA complex, which may allow the achievement of higher receptor occupancy than unselective drugs. Our data also provide information on the most suitable experimental models for future investigation of GABAergic compounds.

Trial registration: ClinicalTrials.gov NCT01011036.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Flowchart of the study.
Figure 1. Flowchart of the study.
Figure 2. Capsaicin model.
Figure 2. Capsaicin model.
The dots represent the points where stimulation was applied. The circle in the center of the hectagon is the site of capsaicin injection.
Figure 3. Time plan of the experiment.
Figure 3. Time plan of the experiment.
Horizontal arrow: Testing time; ZZ: Resting time; EX: Medical examination, installation of the testing equipment and training measures; CT: Measures on the area of capsaicin hyperalgesia; ET: Cutaneous electrical stimulation; ED: Intramuscular electrical stimulation, pressure stimulation, cold pressor test, conditioned pain modulation and cuff algometry; BT: Psychomotor performance test; b: Blood sample.
Figure 4. Static pinprick hyperalgesia after capsaicin.
Figure 4. Static pinprick hyperalgesia after capsaicin.
The area of static hyperalgesia significantly increased with the active placebo tolterodine (p

Figure 5. Intramuscular repeated electrical stimulation (5…

Figure 5. Intramuscular repeated electrical stimulation (5 stimuli).

There was a statistically significant increase in…

Figure 5. Intramuscular repeated electrical stimulation (5 stimuli).
There was a statistically significant increase in the temporal summation thresholds for clonazepam (p = 0.021) and clobazam (p = 0.021) between baseline measures and 120 minutes, whereas no statistically significant change in threshold after the active placebo tolterodine was observed.
Figure 5. Intramuscular repeated electrical stimulation (5…
Figure 5. Intramuscular repeated electrical stimulation (5 stimuli).
There was a statistically significant increase in the temporal summation thresholds for clonazepam (p = 0.021) and clobazam (p = 0.021) between baseline measures and 120 minutes, whereas no statistically significant change in threshold after the active placebo tolterodine was observed.

References

    1. Woolf CJ, Salter MW (2000) Neuronal plasticity: increasing the gain in pain. Science 288: 1765–1769.
    1. Curatolo M, Arendt-Nielsen L, Petersen-Felix S (2006) Central hypersensitivity in chronic pain: mechanisms and clinical implications. Phys Med Rehabil Clin N Am 17: 287–302.
    1. Zeilhofer HU (2005) The glycinergic control of spinal pain processing. Cell Mol Life Sci 62: 2027–2035.
    1. Knabl J, Zeilhofer UB, Crestani F, Rudolph U, Zeilhofer HU (2009) Genuine antihyperalgesia by systemic diazepam revealed by experiments in GABAA receptor point-mutated mice. Pain 141: 233–238.
    1. Reichl S, Augustin M, Zahn PK, Pogatzki-Zahn EM (2011) Peripheral and spinal GABAergic regulation of incisional pain in rats. Pain
    1. Knabl J, Witschi R, Hosl K, Reinold H, Zeilhofer UB, et al. (2008) Reversal of pathological pain through specific spinal GABAA receptor subtypes. Nature 451: 330–334.
    1. Staahl C, Olesen AE, Andresen T, Arendt-Nielsen L, Drewes AM (2009) Assessing efficacy of non-opioid analgesics in experimental pain models in healthy volunteers: an updated review. Br J Clin Pharmacol 68: 322–341.
    1. Wildin JD, Pleuvry BJ, Mawer GE, Onon T, Millington L (1990) Respiratory and sedative effects of clobazam and clonazepam in volunteers. Br J Clin Pharmacol 29: 169–177.
    1. van der Meyden CH, Bartel PR, Sommers DK, Blom M, Pretorius LC (1989) Effects of clobazam and clonazepam on saccadic eye movements and other parameters of psychomotor performance. Eur J Clin Pharmacol 37: 365–369.
    1. Wiffen P, Collins S, McQuay H, Carroll D, Jadad A, et al. (2005) Anticonvulsant drugs for acute and chronic pain. Cochrane Database of Systematic Reviews 20: CD001133.
    1. Hugel H, Ellershaw JE, Dickman A (2003) Clonazepam as an adjuvant analgesic in patients with cancer-related neuropathic pain. Journal of Pain and Symptom Management 26: 1073–1074.
    1. Riss J, Cloyd J, Gates J, Collins S (2008) Benzodiazepines in epilepsy: pharmacology and pharmacokinetics. Acta Neurol Scand 118: 69–86.
    1. Nilvebrant L, Hallen B, Larsson G (1997) Tolterodine–a new bladder selective muscarinic receptor antagonist: preclinical pharmacological and clinical data. Life Science 60: 1129–1136.
    1. Whitwam JG, Amrein R (1995) Pharmacology of flumazenil. Acta Anaesthesiol Scand Suppl 108: 3–14.
    1. Gazerani P, Andersen OK, Arendt-Nielsen L (2007) Site-specific, dose-dependent, and sex-related responses to the experimental pain model induced by intradermal injection of capsaicin to the foreheads and forearms of healthy humans. J Orofac Pain 21: 289–302.
    1. Scanlon GC, Wallace MS, Ispirescu JS, Schulteis G (2006) Intradermal capsaicin causes dose-dependent pain, allodynia, and hyperalgesia in humans. J Investig Med 54: 238–244.
    1. Torebjörk HE, Lundberg LE, LaMotte RH (1992) Central changes in processing of mechanoreceptive input in capsaicin-induced secondary hyperalgesia in humans. Journal of Physiology (London) 448: 765–780.
    1. Witschi R, Punnakkal P, Paul J, Walczak JS, Cervero F, et al. (2011) Presynaptic alpha2-GABAA receptors in primary afferent depolarization and spinal pain control. Journal of Neuroscience 31: 8134–8142.
    1. Chitour D, Dickenson AH, Le Bars D (1982) Pharmacological evidence for the involvement of serotonergic mechanisms in diffuse noxious inhibitory controls (DNIC). Brain Res 236: 329–337.
    1. Ge HY, Madeleine P, Arendt-Nielsen L (2004) Sex differences in temporal characteristics of descending inhibitory control: an evaluation using repeated bilateral experimental induction of muscle pain. Pain 110: 72–78.
    1. Arendt-Nielsen L, Sonnenborg FA, Andersen OK (2000) Facilitation of the withdrawal reflex by repeated transcutaneous electrical stimulation: an experimental study on central integration in humans. European Journal of Applied Physiology 81: 165–173.
    1. Polianskis R, Graven-Nielsen T, Arendt-Nielsen L (2002) Spatial and temporal aspects of deep tissue pain assessed by cuff algometry. Pain 100: 19–26.
    1. Hindmarch I (1980) Psychomotor function and psychoactive drugs. Br J Clin Pharmacol 10: 189–209.
    1. Wang H, Bolognese J, Calder N, Baxendale J, Kehler A, et al. (2008) Effect of morphine and pregabalin compared with diphenhydramine hydrochloride and placebo on hyperalgesia and allodynia induced by intradermal capsaicin in healthy male subjects. J Pain 9: 1088–1095.
    1. Pullar T, Haigh JR, Peaker S, Feely MP (1987) Pharmacokinetics of N-desmethylclobazam in healthy volunteers and patients with epilepsy. British Journal of Clinical Pharmacology 24: 793–797.
    1. LaMotte RH, Shain CN, Simone DA, Tsai EF (1991) Neurogenic hyperalgesia: psychophysical studies of underlying mechanisms. Journal of Neurophysiology 66: 190–211.
    1. Poyhia R, Vainio A (2006) Topically administered ketamine reduces capsaicin-evoked mechanical hyperalgesia. Clin J Pain 22: 32–36.
    1. Wallace MS, Ridgeway B, Leung A, Schulteis G, Yaksh TL (2002) Concentration-effect relationships for intravenous alfentanil and ketamine infusions in human volunteers: effects on acute thresholds and capsaicin-evoked hyperpathia. Journal of Clinical Pharmacology 42: 70–80.
    1. Gottrup H, Juhl G, Kristensen AD, Lai R, Chizh BA, et al. (2004) Chronic oral gabapentin reduces elements of central sensitization in human experimental hyperalgesia. Anesthesiology 101: 1400–1408.
    1. Wallace MS, Barger D, Schulteis G (2002) The effect of chronic oral desipramine on capsaicin-induced allodynia and hyperalgesia: a double-blinded, placebo-controlled, crossover study. Anesthesia and Analgesia 95: 973–978.
    1. Kraft B, Frickey NA, Kaufmann RM, Reif M, Frey R, et al. (2008) Lack of analgesia by oral standardized cannabis extract on acute inflammatory pain and hyperalgesia in volunteers. Anesthesiology 109: 101–110.
    1. Wallace M, Schulteis G, Atkinson JH, Wolfson T, Lazzaretto D, et al. (2007) Dose-dependent effects of smoked cannabis on capsaicin-induced pain and hyperalgesia in healthy volunteers. Anesthesiology 107: 785–796.
    1. Curatolo M, Petersen-Felix S, Gerber A, Arendt-Nielsen L (2000) Remifentanil inhibits muscular more than cutaneous pain in humans. British Journal of Anaesthesia 85: 529–532.
    1. Dickenson AH, Sullivan AF (1987) Evidence for a role of the NMDA receptor in the frequency dependent potentiation of deep rat dorsal horn nociceptive neurones following C fibre stimulation. Neuropharmacology 26: 1235–1238.
    1. Ho KM, Ismail H (2008) Use of intrathecal midazolam to improve perioperative analgesia: a meta-analysis. Anaesthesia and Intensive Care 36: 365–373.
    1. Corrigan R, Derry S, Wiffen PJ, Moore RA (2012) Clonazepam for neuropathic pain and fibromyalgia in adults. Cochrane Database of Systematic Reviews 5: CD009486.
    1. Staahl C, Olesen AE, Andresen T, Arendt-Nielsen L, Drewes AM (2009) Assessing analgesic actions of opioids by experimental pain models in healthy volunteers - an updated review. Br J Clin Pharmacol 68: 149–168.
    1. Neziri AY, Curatolo M, Nuesch E, Scaramozzino P, Andersen OK, et al. (2011) Factor analysis of responses to thermal, electrical, and mechanical painful stimuli supports the importance of multi-modal pain assessment. Pain 152: 1146–1155.
    1. Di Lio A, Benke D, Besson M, Desmeules J, Daali Y, et al. (2011) HZ166, a novel GABAA receptor subtype-selective benzodiazepine site ligand, is antihyperalgesic in mouse models of inflammatory and neuropathic pain. Neuropharmacology 60: 626–632.
    1. Zeilhofer HU, Mohler H, Di Lio A (2009) GABAergic analgesia: new insights from mutant mice and subtype-selective agonists. Trends in Pharmacological Sciences 30: 397–402.
    1. Rudolph U, Knoflach F (2011) Beyond classical benzodiazepines: novel therapeutic potential of GABA(A) receptor subtypes. Nature Reviews Drug Discovery 10: 685–697.
    1. de Haas SL, de Visser SJ, van der Post JP, de Smet M, Schoemaker RC, et al. (2007) Pharmacodynamic and pharmacokinetic effects of TPA023, a GABA(A) alpha(2,3) subtype-selective agonist, compared to lorazepam and placebo in healthy volunteers. Journal of Psychopharmacology 21: 374–383.

Source: PubMed

3
Suscribir