Korean J Pain.  2014 Apr;27(2):103-111. 10.3344/kjp.2014.27.2.103.

Rediscovery of Nefopam for the Treatment of Neuropathic Pain

Affiliations
  • 1Department of Anesthesia and Pain Medicine, School of Medicine, Pusan National University, Yangsan, Korea.
  • 2Department of Pain Medicine, Division of Anesthesiology and Critical Care, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. SAbdi@mdanderson.org

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.

Keyword

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

MeSH Terms

Administration, Intravenous
Administration, Oral
Analgesia
Analgesics, Non-Narcotic
Anticonvulsants
Antidepressive Agents
Calcium
Carbamazepine
Dizziness
Drug-Related Side Effects and Adverse Reactions
Hiccup
Long-Term Potentiation
Molecular Mechanisms of Pharmacological Action
N-Methylaspartate
Nausea
Nefopam*
Neuralgia*
Neurotransmitter Agents
Nociceptive Pain
Norepinephrine
Shivering
Sleep Stages
Sodium Channels
Sweat
Sweating
Tachycardia
Analgesics, Non-Narcotic
Anticonvulsants
Antidepressive Agents
Calcium
Carbamazepine
Molecular Mechanisms of Pharmacological Action
N-Methylaspartate
Nefopam
Neurotransmitter Agents
Norepinephrine
Sodium Channels

Figure

  • Fig. 1 Similarity of structural formulae of (A) orphenadrine, (B) diphenhydramine, and (C) nefopam.

  • 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 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 The roles and deficit states of 3 important neurotransmitters (serotonin, norepinephrine, and dopamine) and balanced and unbalanced states of these neurotransmitters in human body.


Cited by  14 articles

Effects on postoperative nausea and vomiting of nefopam versus fentanyl following bimaxillary orthognathic surgery: a prospective double-blind randomized controlled trial
Eunhye Choi, Myong-Hwan Karm, Eunsun So, Yoon Ji Choi, Sookyung Park, Yul Oh, Hye Joo Yun, Hyun Jeong Kim, Kwang-Suk Seo
J Dent Anesth Pain Med. 2019;19(1):55-66.    doi: 10.17245/jdapm.2019.19.1.55.

Comparison of effect of electroacupuncture and nefopam for prevention of postanesthetic shivering in patients undergoing urologic operation under spinal anesthesia
Jun-Ho Hong, Su-Jin Kim, Min-Sub Hwang
Korean J Anesthesiol. 2016;69(6):579-586.    doi: 10.4097/kjae.2016.69.6.579.

Emergence agitation: current knowledge and unresolved questions
Seok-Jin Lee, Tae-Yun Sung
Korean J Anesthesiol. 2020;73(6):471-485.    doi: 10.4097/kja.20097.

For the peace of postanesthesia care unit
Sung Mi Hwang
Korean J Anesthesiol. 2018;71(3):173-174.    doi: 10.4097/kja.d.18.00113.

Effects of nefopam on catheter-related bladder discomfort in patients undergoing ureteroscopic litholapaxy
Yong Woo Cheon, Seon Hwan Kim, Jin Hyub Paek, Jin A Kim, Yong Kyung Lee, Jin Hye Min, Hyung Rae Cho
Korean J Anesthesiol. 2018;71(3):201-206.    doi: 10.4097/kja.d.18.27113.

All about pain pharmacology: what pain physicians should know
Kyung-Hoon Kim, Hyo-Jung Seo, Salahadin Abdi, Billy Huh
Korean J Pain. 2020;33(2):108-120.    doi: 10.3344/kjp.2020.33.2.108.

The Potential Role of Intrathecal Nefopam in the Management of Neuropathic Pain
Mohamed Amin Ghobadifar, Navid Kalani
Korean J Pain. 2014;27(3):301-302.    doi: 10.3344/kjp.2014.27.3.301.

The Effect of Nefopam on Postoperative Fentanyl Consumption: A Randomized, Double-blind Study
Jee Youn Moon, Sang Sik Choi, Shin Young Lee, Mi Kyung Lee, Jung Eun Kim, Ji Eun Lee, So Hyun Lee
Korean J Pain. 2016;29(2):110-118.    doi: 10.3344/kjp.2016.29.2.110.

The Role of Spinal Dopaminergic Transmission in the Analgesic Effect of Nefopam on Rat Inflammatory Pain
Do Yun Kim, Joo Wung Chae, Chang Hun Lim, Bong Ha Heo, Keun Suk Park, Hyung Gon Lee, Jeong Il Choi, Myung Ha Yoon, Woong Mo Kim
Korean J Pain. 2016;29(3):164-171.    doi: 10.3344/kjp.2016.29.3.164.

Tapentadol: Can It Kill Two Birds with One Stone without Breaking Windows?
Eun Jung Chang, Eun Ji Choi, Kyung Hoon Kim
Korean J Pain. 2016;29(3):153-157.    doi: 10.3344/kjp.2016.29.3.153.

Nefopam Reduces Dysesthesia after Percutaneous Endoscopic Lumbar Discectomy
Young Min Ok, Ji Hyun Cheon, Eun Ji Choi, Eun Jung Chang, Ho Myung Lee, Kyung Hoon Kim
Korean J Pain. 2016;29(1):40-47.    doi: 10.3344/kjp.2016.29.1.40.

Use of Nefopam in Perioperative Pain Management; Keeping Nefopam in between
Jeong Il Choi
Korean J Pain. 2016;29(2):71-72.    doi: 10.3344/kjp.2016.29.2.71.

Earlier treatment improves the chances of complete relief from postherpetic neuralgia
Dong Hee Kang, Su Young Kim, Hyuck Goo Kim, Jung Hyun Park, Tae Kyun Kim, Kyung Hoon Kim
Korean J Pain. 2017;30(3):214-219.    doi: 10.3344/kjp.2017.30.3.214.

Status Epilepticus Caused by Nefopam
Yong-sook Park, Young-baeg Kim, Jeong-min Kim
J Korean Neurosurg Soc. 2014;56(5):448-450.    doi: 10.3340/jkns.2014.56.5.448.


Reference

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. PMID: 22711801.
Article
2. 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. PMID: 21901726.
Article
3. 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. PMID: 14695722.
Article
4. 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. PMID: 6991238.
5. Podranski T, Bouillon TW, Riva T, Kurz AM, Oehmke MJ. Compartmental pharmacokinetics of nefopam during mild hypothermia. Br J Anaesth. 2012; 108:784–791. PMID: 22331396.
Article
6. 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. PMID: 5343358.
Article
7. Tobin WE, Gold RH. Nefopam hydrochloride: a novel muscle relaxant. J Clin Pharmacol New Drugs. 1972; 12:230–238. PMID: 4555883.
Article
8. 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. PMID: 4842015.
Article
9. 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. PMID: 994022.
10. Cohen A. Nefopam hydrochloride for pain relief. Curr Ther Res Clin Exp. 1974; 16:184–193. PMID: 4206725.
11. Klotz AL. Long-term safety of Nefopam hydrochloride (Acupan), a new analgesic formulation. Curr Ther Res Clin Exp. 1974; 16:602–608. PMID: 4211140.
12. Workmon FC, Winter L Jr. A clinical evaluation of nefopam hydrochloride (Acupan): a new analgesic. Curr Ther Res Clin Exp. 1974; 16:609–616. PMID: 4211141.
13. Kolodny AL, Winter L Jr. Further clinical evaluations of nefopam hydrochloride, a new analgesic. Curr Ther Res Clin Exp. 1975; 17:519–524. PMID: 808373.
14. 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. PMID: 19588431.
Article
15. Izzo V, Mariconti P, Tiengo M. Action and effectiveness of nefopam chloride in the control of postoperative shivering. Minerva Anestesiol. 1991; 57:760–762. PMID: 1798568.
16. 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. PMID: 23560188.
Article
17. 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. PMID: 22890247.
18. Bilotta F, Rosa G. Nefopam for severe hiccups. N Engl J Med. 2000; 343:1973–1974. PMID: 11186682.
Article
19. Bilotta F, Pietropaoli P, Rosa G. Nefopam for refractory postoperative hiccups. Anesth Analg. 2001; 93:1358–1360. PMID: 11682430.
Article
20. 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. PMID: 17933805.
Article
21. Barrot M. Tests and models of nociception and pain in rodents. Neuroscience. 2012; 211:39–50. PMID: 22244975.
Article
22. Jaggi AS, Jain V, Singh N. Animal models of neuropathic pain. Fundam Clin Pharmacol. 2011; 25:1–28. PMID: 20030738.
Article
23. Hunskaar S, Hole K. The formalin test in mice: dissociation between inflammatory and non-inflammatory pain. Pain. 1987; 30:103–114. PMID: 3614974.
Article
24. 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. PMID: 23342202.
Article
25. 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. PMID: 11735363.
Article
26. 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. PMID: 12007921.
Article
27. 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. PMID: 19608840.
Article
28. Evans MS, Lysakowski C, Tramèr MR. Nefopam for the prevention of postoperative pain: quantitative systematic review. Br J Anaesth. 2008; 101:610–617. PMID: 18796441.
Article
29. 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. PMID: 397711.
Article
30. 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. PMID: 7864317.
Article
31. Gasser JC, Bellville JW. Respiratory effects of nefopam. Clin Pharmacol Ther. 1975; 18:175–179. PMID: 1097153.
Article
32. Guindon J, Walczak JS, Beaulieu P. Recent advances in the pharmacological management of pain. Drugs. 2007; 67:2121–2133. PMID: 17927280.
Article
33. Torres GE, Gainetdinov RR, Caron MG. Plasma membrane monoamine transporters: structure, regulation and function. Nat Rev Neurosci. 2003; 4:13–25. PMID: 12511858.
Article
34. 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. p. 1710–1726.
35. 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. PMID: 15714253.
Article
36. 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. PMID: 15193535.
Article
37. 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. PMID: 12906743.
Article
38. 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. PMID: 17021653.
Article
39. 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. PMID: 21857079.
Article
40. Löscher W, Schmidt D. New Horizons in the development of antiepileptic drugs: innovative strategies. Epilepsy Res. 2006; 69:183–272. PMID: 16835945.
Article
41. 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. PMID: 2699187.
Article
42. 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. PMID: 7938165.
43. Fink KB, Göthert M. 5-HT receptor regulation of neurotransmitter release. Pharmacol Rev. 2007; 59:360–417. PMID: 18160701.
Article
44. Barnes NM, Sharp T. A review of central 5-HT receptors and their function. Neuropharmacology. 1999; 38:1083–1152. PMID: 10462127.
Article
45. Bannister K, Bee LA, Dickenson AH. Preclinical and early clinical investigations related to monoaminergic pain modulation. Neurotherapeutics. 2009; 6:703–712. PMID: 19789074.
Article
46. Cotecchia S, Stanasila L, Diviani D. Protein-protein interactions at the adrenergic receptors. Curr Drug Targets. 2012; 13:15–27. PMID: 21777184.
Article
47. Girault JA, Greengard P. The neurobiology of dopamine signaling. Arch Neurol. 2004; 61:641–644. PMID: 15148138.
Article
48. Benzon HT. The neuropathic pain scales. Reg Anesth Pain Med. 2005; 30:417–421. PMID: 16135344.
Article
49. 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. PMID: 10689295.
Article
50. 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. PMID: 17868209.
Article
Full Text Links
  • KJP
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr