Rediscovery of nefopam for the treatment of neuropathic pain

Kyung Hoon Kim, Salahadin Abdi, Kyung Hoon Kim, Salahadin Abdi

Abstract

Nefopam (NFP) is a non-opioid, non-steroidal, centrally acting analgesic drug that is derivative of the non-sedative benzoxazocine, developed and known in 1960s as fenazocine. Although the mechanisms of analgesic action of NFP are not well understood, they are similar to those of triple neurotransmitter (serotonin, norepinephrine, and dopamine) reuptake inhibitors and anticonvulsants. It has been used mainly as an analgesic drug for nociceptive pain, as well as a treatment for the prevention of postoperative shivering and hiccups. Based on NFP's mechanisms of analgesic action, it is more suitable for the treatment of neuropathic pain. Intravenous administration of NFP should be given in single doses of 20 mg slowly over 15-20 min or with continuous infusion of 60-120 mg/d to minimize adverse effects, such as nausea, cold sweating, dizziness, tachycardia, or drowsiness. The usual dose of oral administration is three to six times per day totaling 90-180 mg. The ceiling effect of its analgesia is uncertain depending on the mechanism of pain relief. In conclusion, the recently discovered dual analgesic mechanisms of action, namely, a) descending pain modulation by triple neurotransmitter reuptake inhibition similar to antidepressants, and b) inhibition of long-term potentiation mediated by NMDA from the inhibition of calcium influx like gabapentinoid anticonvulsants or blockade of voltage-sensitive sodium channels like carbamazepine, enable NFP to be used as a therapeutic agent to treat neuropathic pain.

Keywords: adverse drug reactions; molecular mechanisms of pharmacological action; nefopam; neuropathic pain; nonopioid analgesics.

Figures

Fig. 1
Fig. 1
Similarity of structural formulae of (A) orphenadrine, (B) diphenhydramine, and (C) nefopam.
Fig. 2
Fig. 2
Schematic presentation of dopamine (DA), noradrenalin (NA), and 5-hydroxytryptamine (5-HT) synaptic terminals drawn in the same neuron. In human body, they are actually located in the different neurons and locations: (1) The dopamine transporter (DAT) expression is dominant in the cell bodies of the substantia nigra and ventral tegmental area: (2) The noradrenalin transporter (NET) expression is abundant in the locus coeruleus and other brain stem nuclei; (3) The 5-HT transporter (SERT) expression is frequently found in the median and dorsal raphe nuclei. They are also found peripherally, especially in the dorsal root ganglia related to descending inhibition of pain. Monoamine transporters are localized to presynaptic sites, where they are crucial for the termination of monoamine transmission and the maintenance of presynaptic monoamine storage. Nefopam (NFP) has an ability of these 3 receptors reuptake inhibition (Modified from Torres GE, Gainetdinov RR, Caron MG. Plasma membrane monoamine transporters: structure, regulation and function. Nat Rev Neurosci 2003; 4: 13-25.).
Fig. 3
Fig. 3
Model synapse illustrating interaction of Na+ channel blocking anticonvulsants with voltage-activated Na+ channels and putative sites of action of newer anticonvulsants (gabapentin, pregabalin, and levetiracetam) that may more directly interact with release machinery. Gabapentin and pregabalin bind to α2-δ, which may inhibit voltage-activated Ca2+ entry through high voltage-activated Ca2+ channels or affect the way in which Ca2+ channels interact with vesicular release. Levetiracetam may also affect release by binding to synaptic vesicles protein SV2A. In contrast, action potentials are mediated by voltage-activated Na+ and K+ channels; Na+ channel blocking anticonvulsants suppress epileptiform action potential firing, which leads to inhibited release. Smaller yellow circles represent glutamate within synaptic vesicles (larger blue circles) and free in the synaptic cleft. Glutamate acts on ionotropic receptors of the NMDA, AMPA and kainate types to generate an excitatory postsynaptic potential (EPSP) in the postsynaptic neuron. Nefopam (NFP) shows an activity of the inhibition of long-term potentiation mediated by NMDA from the inhibition of calcium influx like gabapentinoid anticonvulsants or blockade of voltage-sensitive sodium channels like carbamazepine (Modified by Löscher W, Schmidt D. New Horizons in the development of antiepileptic drugs: Innovative strategies. Epilepsy Res 2006; 69: 183-272.).
Fig. 4
Fig. 4
The roles and deficit states of 3 important neurotransmitters (serotonin, norepinephrine, and dopamine) and balanced and unbalanced states of these neurotransmitters in human body.

References

    1. Gregori-Puigjané E, Setola V, Hert J, Crews BA, Irwin JJ, Lounkine E, et al. Identifying mechanism-of-action targets for drugs and probes. Proc Natl Acad Sci U S A. 2012;109:11178–11183.
    1. Moore RA, Derry S, McQuay HJ, Wiffen PJ. Single dose oral analgesics for acute postoperative pain in adults. Cochrane Database Syst Rev. 2011;(9):CD008659.
    1. Alfonsi P, Adam F, Passard A, Guignard B, Sessler DI, Chauvin M. Nefopam, a nonsedative benzoxazocine analgesic, selectively reduces the shivering threshold in unanesthetized subjects. Anesthesiology. 2004;100:37–43.
    1. Heel RC, Brogden RN, Pakes GE, Speight TM, Avery GS. Nefopam: a review of its pharmacological properties and therapeutic efficacy. Drugs. 1980;19:249–267.
    1. Podranski T, Bouillon TW, Riva T, Kurz AM, Oehmke MJ. Compartmental pharmacokinetics of nefopam during mild hypothermia. Br J Anaesth. 2012;108:784–791.
    1. Bassett JR, Cairncross KD, Hacket NB, Story M. Studies on the peripheral pharmacology of fenazoxine, a potential antidepressant drug. Br J Pharmacol. 1969;37:69–78.
    1. Tobin WE, Gold RH. Nefopam hydrochloride: a novel muscle relaxant. J Clin Pharmacol New Drugs. 1972;12:230–238.
    1. Bolt AG, Graham G, Wilson P. Stereoselective demethylation of the enantiomers of nefopam, an experimental antidepressant and skeletal muscle relaxant. Xenobiotica. 1974;4:355–363.
    1. Koe BK. Molecular geometry of inhibitors of the uptake of catecholamines and serotonin in synaptosomal preparations of rat brain. J Pharmacol Exp Ther. 1976;199:649–661.
    1. Cohen A. Nefopam hydrochloride for pain relief. Curr Ther Res Clin Exp. 1974;16:184–193.
    1. Klotz AL. Long-term safety of Nefopam hydrochloride (Acupan), a new analgesic formulation. Curr Ther Res Clin Exp. 1974;16:602–608.
    1. Workmon FC, Winter L., Jr A clinical evaluation of nefopam hydrochloride (Acupan): a new analgesic. Curr Ther Res Clin Exp. 1974;16:609–616.
    1. Kolodny AL, Winter L., Jr Further clinical evaluations of nefopam hydrochloride, a new analgesic. Curr Ther Res Clin Exp. 1975;17:519–524.
    1. Kakkar M, Derry S, Moore RA, McQuay HJ. Single dose oral nefopam for acute postoperative pain in adults. Cochrane Database Syst Rev. 2009;(3):CD007442.
    1. Izzo V, Mariconti P, Tiengo M. Action and effectiveness of nefopam chloride in the control of postoperative shivering. Minerva Anestesiol. 1991;57:760–762.
    1. Kim YA, Kweon TD, Kim M, Lee HI, Lee YJ, Lee KY. Comparison of meperidine and nefopam for prevention of shivering during spinal anesthesia. Korean J Anesthesiol. 2013;64:229–233.
    1. Park SM, Mangat HS, Berger K, Rosengart AJ. Efficacy spectrum of antishivering medications: meta-analysis of randomized controlled trials. Crit Care Med. 2012;40:3070–3082.
    1. Bilotta F, Rosa G. Nefopam for severe hiccups. N Engl J Med. 2000;343:1973–1974.
    1. Bilotta F, Pietropaoli P, Rosa G. Nefopam for refractory postoperative hiccups. Anesth Analg. 2001;93:1358–1360.
    1. Pajot S, Geeraerts T, Leblanc PE, Duranteau J, Benhamou D. Hiccup during weaning from mechanical ventilation: the use of nefopam. Br J Anaesth. 2007;99:748–749.
    1. Barrot M. Tests and models of nociception and pain in rodents. Neuroscience. 2012;211:39–50.
    1. Jaggi AS, Jain V, Singh N. Animal models of neuropathic pain. Fundam Clin Pharmacol. 2011;25:1–28.
    1. Hunskaar S, Hole K. The formalin test in mice: dissociation between inflammatory and non-inflammatory pain. Pain. 1987;30:103–114.
    1. Cho SY, Park AR, Yoon MH, Lee HG, Kim WM, Choi JI. Antinociceptive effect of intrathecal nefopam and interaction with morphine in formalin-induced pain of rats. Korean J Pain. 2013;26:14–20.
    1. Girard P, Pansart Y, Coppe MC, Gillardin JM. Nefopam reduces thermal hypersensitivity in acute and postoperative pain models in the rat. Pharmacol Res. 2001;44:541–545.
    1. Buritova J, Besson JM. Effects of nefopam on the spinal nociceptive processes: a c-Fos protein study in the rat. Eur J Pharmacol. 2002;441:67–74.
    1. Laboureyras E, Chateauraynaud J, Richebé P, Simonnet G. Long-term pain vulnerability after surgery in rats: prevention by nefopam, an analgesic with antihyperalgesic properties. Anesth Analg. 2009;109:623–631.
    1. Evans MS, Lysakowski C, Tramèr MR. Nefopam for the prevention of postoperative pain: quantitative systematic review. Br J Anaesth. 2008;101:610–617.
    1. Tigerstedt I, Tammisto T, Leander P. Comparison of the analgesic dose-effect relationships of nefopam and oxycodone in postoperative pain. Acta Anaesthesiol Scand. 1979;23:555–560.
    1. Dordoni PL, Della Ventura M, Stefanelli A, Iannace E, Paparella P, Rocca B, et al. Effect of ketorolac, ketoprofen and nefopam on platelet function. Anaesthesia. 1994;49:1046–1049.
    1. Gasser JC, Bellville JW. Respiratory effects of nefopam. Clin Pharmacol Ther. 1975;18:175–179.
    1. Guindon J, Walczak JS, Beaulieu P. Recent advances in the pharmacological management of pain. Drugs. 2007;67:2121–2133.
    1. Torres GE, Gainetdinov RR, Caron MG. Plasma membrane monoamine transporters: structure, regulation and function. Nat Rev Neurosci. 2003;4:13–25.
    1. Max MB, Gilron IH. Antidepressants, muscle relaxants, and N-methyl-D-aspartate receptor antagonists. In: Loeser JD, Bonica JJ, editors. Bonica's management of pain. 3rd ed. Philadelphia (PA): Lippincott Williams & Wilkins; 2001. pp. 1710–1726.
    1. Novelli A, Díaz-Trelles R, Groppetti A, Fernández-Sánchez MT. Nefopam inhibits calcium influx, cGMP formation, and NMDA receptor-dependent neurotoxicity following activation of voltage sensitive calcium channels. Amino Acids. 2005;28:183–191.
    1. Verleye M, André N, Heulard I, Gillardin JM. Nefopam blocks voltage-sensitive sodium channels and modulates glutamatergic transmission in rodents. Brain Res. 2004;1013:249–255.
    1. Biella GE, Groppetti A, Novelli A, Fernández-Sánchez MT, Manfredi B, Sotgiu ML. Neuronal sensitization and its behavioral correlates in a rat model of neuropathy are prevented by a cyclic analog of orphenadrine. J Neurotrauma. 2003;20:593–601.
    1. Novelli A, Groppetti A, Rossoni G, Manfredi B, Ferrero-Gutiérrez A, Pérez-Gómez A, et al. Nefopam is more potent than carbamazepine for neuroprotection against veratridine in vitro and has anticonvulsant properties against both electrical and chemical stimulation. Amino Acids. 2007;32:323–332.
    1. Czuczwar M, Czuczwar K, Cięszczyk J, Kiś J, Saran T, Łuszczki JJ, et al. Nefopam enhances the protective activity of antiepileptics against maximal electroshock-induced convulsions in mice. Pharmacol Rep. 2011;63:690–696.
    1. Löscher W, Schmidt D. New Horizons in the development of antiepileptic drugs: innovative strategies. Epilepsy Res. 2006;69:183–272.
    1. Hoebel BG, Hernandez L, Schwartz DH, Mark GP, Hunter GA. Microdialysis studies of brain norepinephrine, serotonin, and dopamine release during ingestive behavior. Theoretical and clinical implications. Ann N Y Acad Sci. 1989;575:171–191.
    1. Hoyer D, Clarke DE, Fozard JR, Hartig PR, Martin GR, Mylecharane EJ, et al. International Union of Pharmacology classification of receptors for 5-hydroxytryptamine (Serotonin) Pharmacol Rev. 1994;46:157–203.
    1. Fink KB, Göthert M. 5-HT receptor regulation of neurotransmitter release. Pharmacol Rev. 2007;59:360–417.
    1. Barnes NM, Sharp T. A review of central 5-HT receptors and their function. Neuropharmacology. 1999;38:1083–1152.
    1. Bannister K, Bee LA, Dickenson AH. Preclinical and early clinical investigations related to monoaminergic pain modulation. Neurotherapeutics. 2009;6:703–712.
    1. Cotecchia S, Stanasila L, Diviani D. Protein-protein interactions at the adrenergic receptors. Curr Drug Targets. 2012;13:15–27.
    1. Girault JA, Greengard P. The neurobiology of dopamine signaling. Arch Neurol. 2004;61:641–644.
    1. Benzon HT. The neuropathic pain scales. Reg Anesth Pain Med. 2005;30:417–421.
    1. Mather GG, Labroo R, Le Guern ME, Lepage F, Gillardin JM, Levy RH. Nefopam enantiomers: preclinical pharmacology/toxicology and pharmacokinetic characteristics in healthy subjects after intravenous administration. Chirality. 2000;12:153–159.
    1. Durrieu G, Olivier P, Bagheri H, Montastruc JL French Network of Pharmacovigilance Centers. Overview of adverse reactions to nefopam: an analysis of the French Pharmacovigilance database. Fundam Clin Pharmacol. 2007;21:555–558.

Source: PubMed

3
Suscribir