275
Views
6
CrossRef citations to date
0
Altmetric
Review

Pharmacologic agents directed at the treatment of pain associated with maladaptive neuronal plasticity

, ORCID Icon, ORCID Icon, , ORCID Icon, , , ORCID Icon, ORCID Icon, & show all
Pages 105-116 | Received 10 Feb 2021, Accepted 16 Aug 2021, Published online: 30 Aug 2021

References

  • Perry VH, Teeling J. Microglia and macrophages of the central nervous system: the contribution of microglia priming and systemic inflammation to chronic neurodegeneration. Semin Immunopathol. 2013;35(5):601–612.
  • Castren E, Antila H. Neuronal plasticity and neurotrophic factors in drug responses. Mol Psychiatry. 2017;22(8):1085–1095.
  • Melzack R. From the gate to the neuromatrix. Pain. 1999;Suppl 6:S121–126. DOI:https://doi.org/10.1016/S0304-3959(99)00145-1.
  • IASP terminology: international association for the study of pain. 2020. [cited 2020 Sep 10]. Available from: https://www.iasp-pain.org/Education/Content.aspx?ItemNumber=1698
  • Trouvin AP, Perrot S. New concepts of pain. Best Pract Res Clin Rheumatol. 2019;33(3):101415.
  • von Hehn CA, Baron R, Woolf CJ, et al. Deconstructing the neuropathic pain phenotype to reveal neural mechanisms. Neuron. 2012;73(4):638–652.
  • Kim DS, Choi JO, Rim HD, et al. Downregulation of voltage-gated potassium channel alpha gene expression in dorsal root ganglia following chronic constriction injury of the rat sciatic nerve. Brain Res Mol Brain Res. 2002;105(1–2):146–152.
  • Yam MF, Loh YC, Tan CS, et al. General pathways of pain sensation and the major neurotransmitters involved in pain regulation. Int J Mol Sci. 2018;19(8). DOI:https://doi.org/10.3390/ijms19082164.
  • Baldwin KT, Eroglu C. Molecular mechanisms of astrocyte-induced synaptogenesis. Curr Opin Neurobiol. 2017;45:113–120.
  • Citri A, Malenka RC. Synaptic plasticity: multiple forms, functions, and mechanisms. Neuropsychopharmacology. 2008;33(1):18–41.
  • Ji RR, Suter MR. P38 mapk, microglial signaling, and neuropathic pain. Mol Pain. 2007;3:33.
  • Skaper SD, Facci L. Mast cell-glia axis in neuroinflammation and therapeutic potential of the anandamide congener palmitoylethanolamide. Philos Trans R Soc Lond B Biol Sci. 2012;367(1607):3312–3325.
  • Varrassi G, Fusco M, Coaccioli S, et al. Chronic pain and neurodegenerative processes in elderly people. Pain Pract. 2015;15(1): 1–3.
  • Fusco M, Skaper SD, Coaccioli S, et al. Degenerative joint diseases and neuroinflammation. Pain Pract. 2017;17(4): 522–532.
  • Boche D, Perry VH, Nicoll JA, et al. Review: activation patterns of microglia and their identification in the human brain. Neuropathol Appl Neurobiol. 2013;39(1):3–18.
  • Varrassi G, Fusco M, Skaper SD, et al. A pharmacological rationale to reduce the incidence of opioid induced tolerance and hyperalgesia: a review. Pain Ther. 2018;7(1): 59–75.
  • Xie F, Zhang JC, Fu H, et al. Age-related decline of myelin proteins is highly correlated with activation of astrocytes and microglia in the rat cns. Int J Mol Med. 2013;32(5):1021–1028.
  • Biggs JE, Lu VB, Stebbing MJ, et al. Is bdnf sufficient for information transfer between microglia and dorsal horn neurons during the onset of central sensitization?. Mol Pain. 2010;6(44). DOI:https://doi.org/10.1186/1744-8069-6-44.
  • Raghavendra V, Tanga F, DeLeo JA, et al. Inhibition of microglial activation attenuates the development but not existing hypersensitivity in a rat model of neuropathy. J Pharmacol Exp Ther. 2003;306(2):624–630.
  • Sandkühler J. Models and mechanisms of hyperalgesia and allodynia. Physiol Rev. 2009;89(2):707–758.
  • Chessell IP, Hatcher JP, Bountra C, et al. Disruption of the p2x7 purinoceptor gene abolishes chronic inflammatory and neuropathic pain. Pain. 2005;114(3):386–396.
  • Bushong EA, Martone ME, Jones YZ, et al. Protoplasmic astrocytes in ca1 stratum radiatum occupy separate anatomical domains. J Neurosci. 2002;22(1):183–192.
  • Silver R, Curley JP. Mast cells on the mind: new insights and opportunities. Trends Neurosci. 2013;36(9):513–521.
  • Skaper SD, Giusti P, Facci L, et al. Microglia and mast cells: two tracks on the road to neuroinflammation. Faseb J. 2012;26(8): 3103–3117.
  • Héron A, Dubayle D. A focus on mast cells and pain. J Neuroimmunol. 2013;264(1–2):1–7.
  • Matsuda H, Kawakita K, Kiso Y, et al. Substance p induces granulocyte infiltration through degranulation of mast cells. J Immunol. 1989;142(3):927–931.
  • Zuo Y, Perkins NM, Tracey DJ, et al. Inflammation and hyperalgesia induced by nerve injury in the rat: a key role of mast cells. Pain. 2003;105(3):467–479.
  • Mika J, Zychowska M, Popiolek-Barczyk K, et al. Importance of glial activation in neuropathic pain. Eur J Pharmacol. 2013;716(1–3):106–119.
  • Alfonso Romero-Sandoval E, Sweitzer S. Nonneuronal central mechanisms of pain: glia and immune response. Prog Mol Biol Transl Sci. 2015;131:325–358.
  • Rajchgot T, Thomas SC, Wang JC, et al. Neurons and microglia; a sickly-sweet duo in diabetic pain neuropathy. Front Neurosci. 2019;13:25.
  • Liampas A, Rekatsina M, Vadalouca A, et al. Pharmacological management of painful peripheral neuropathies: a systematic review. Pain Ther. 2021;10(1):55-68.
  • Zhuang ZY, Wen YR, Zhang DR, et al. A peptide c-jun n-terminal kinase (jnk) inhibitor blocks mechanical allodynia after spinal nerve ligation: respective roles of jnk activation in primary sensory neurons and spinal astrocytes for neuropathic pain development and maintenance. J Neurosci. 2006;26(13):3551–3560.
  • Zhang J, De Koninck Y. Spatial and temporal relationship between monocyte chemoattractant protein-1 expression and spinal glial activation following peripheral nerve injury. J Neurochem. 2006;97(3):772–783.
  • Deogracias R, Yazdani M, Dekkers MP, et al. Fingolimod, a sphingosine-1 phosphate receptor modulator, increases bdnf levels and improves symptoms of a mouse model of rett syndrome. Proc Natl Acad Sci U S A. 2012;109(35):14230–14235.
  • Ferrini F, Salio C, Boggio E, et al. Interplay of bdnf and gdnf in the mature spinal somatosensory system and its potential therapeutic relevance. Curr Neuropharmacol. 2020 Nov 16. doi: https://doi.org/10.2174/1570159X18666201116143422.
  • Smith PA. Bdnf: no gain without pain?. Neuroscience. 2014;283:107–123.
  • Bartus RT, Johnson EM Jr. Clinical tests of neurotrophic factors for human neurodegenerative diseases, part 1: where have we been and what have we learned?. Neurobiol Dis. 2017;97(Pt B):156–168.
  • Nibuya M, Morinobu S, Duman RS, et al. Regulation of bdnf and trkb mrna in rat brain by chronic electroconvulsive seizure and antidepressant drug treatments. J Neurosci. 1995;15(11):7539–7547.
  • Castrén E, Antila H. Neuronal plasticity and neurotrophic factors in drug responses. Mol Psychiatry. 2017;22(8):1085–1095.
  • Bigal ME, Walter S. Monoclonal antibodies for migraine: preventing calcitonin gene-related peptide activity. CNS Drugs. 2014;28(5):389–399.
  • Bigal ME, Walter S, Rapoport AM, et al. Calcitonin gene-related peptide (cgrp) and migraine current understanding and state of development. Headache. 2013;53(8):1230–1244.
  • Yeh J-F, Akinci A, Al Shaker M, et al. Monoclonal antibodies for chronic pain: a practical review of mechanisms and clinical applications. Mol Pain. 2017;13:1744806917740233.
  • Romero-Sandoval A, Nutile-McMenemy N, DeLeo JA, et al. Spinal microglial and perivascular cell cannabinoid receptor type 2 activation reduces behavioral hypersensitivity without tolerance after peripheral nerve injury. Anesthesiology. 2008;108(4):722–734.
  • Alkaitis MS, Solorzano C, Landry RP, et al. Evidence for a role of endocannabinoids, astrocytes and p38 phosphorylation in the resolution of postoperative pain. PLoS One. 2010;5(5): e10891.
  • Speth C, Dierich MP, Gasque P, et al. Neuroinvasion by pathogens: a key role of the complement system. Mol Immunol. 2002;38(9):669–679.
  • Le Friec G, Kemper C. Complement: coming full circle. Arch Immunol Ther Exp (Warsz). 2009;57(6):393–407.
  • Morgan BP, Gasque P. Expression of complement in the brain: role in health and disease. Immunol Today. 1996;17(10):461–466.
  • West EE, Kolev M, Kemper C, et al. Complement and the regulation of t cell responses. Annu Rev Immunol. 2018;36(1):309–338.
  • Mika J. Modulation of microglia can attenuate neuropathic pain symptoms and enhance morphine effectiveness. Pharmacol Rep. 2008;60(3):297–307.
  • Strand V, Burmester GR, Ogale S, et al. Improvements in health-related quality of life after treatment with tocilizumab in patients with rheumatoid arthritis refractory to tumour necrosis factor inhibitors: results from the 24-week randomized controlled radiate study. Rheumatology (Oxford). 2012;51(10):1860–1869.
  • Scholz J, Abele A, Marian C, et al. Low-dose methotrexate reduces peripheral nerve injury-evoked spinal microglial activation and neuropathic pain behavior in rats. Pain. 2008;138(1):130–142.
  • Kersten C, Cameron MG. Cetuximab alleviates neuropathic pain despite tumour progression. BMJ Case Rep. 2012;2012:bcr1220115374.
  • Kersten C, Cameron MG, Laird B, et al. Epidermal growth factor receptor-inhibition (egfr-i) in the treatment of neuropathic pain. Br J Anaesth. 2015;115(5):761–767.
  • Martin LJ, Smith SB, Khoutorsky A, et al. Epiregulin and egfr interactions are involved in pain processing. J Clin Invest. 2017;127(9):3353–3366.
  • Christidis N, Kang I, Cairns BE, et al. Expression of 5-ht3 receptors and ttx resistant sodium channels (na(v)1.8) on muscle nerve fibers in pain-free humans and patients with chronic myofascial temporomandibular disorders. J Headache Pain. 2014;15(1):63.
  • Nilius B, Szallasi A. Transient receptor potential channels as drug targets: from the science of basic research to the art of medicine. Pharmacol Rev. 2014;66(3):676–814.
  • Chincholkar M. Analgesic mechanisms of gabapentinoids and effects in experimental pain models: a narrative review. Br J Anaesth. 2018;120(6):1315–1334.
  • Waszkielewicz AM, Gunia A, Szkaradek N, et al. Ion channels as drug targets in central nervous system disorders. Curr Med Chem. 2013;20(10):1241–1285.
  • Abdallah CG, Sanacora G, Duman RS, et al. Ketamine and rapid-acting antidepressants: a window into a new neurobiology for mood disorder therapeutics. Annu Rev Med. 2015;66(1): 509–523.
  • Duman RS, Aghajanian GK, Sanacora G, et al. Synaptic plasticity and depression: new insights from stress and rapid-acting antidepressants. Nat Med. 2016;22(3):238–249.
  • Corasaniti MT, Amantea D, Russo R, et al. The crucial role of neuronal plasticity in pain and cell death. Cell Death & Differentiation. 2006;13(3):534–536.
  • Tencheva A, Liu R, Volkova TV, et al. Synthetic analogues of memantine as neuroprotective and influenza viral inhibitors: in vitro and physicochemical studies. Amino Acids. 2020;52(11–12):1559–1580.
  • Scholz J, Woolf CJ. The neuropathic pain triad: neurons, immune cells and glia. Nat Neurosci. 2007;10(11):1361–1368.
  • Patel RN, Attur MG, Dave MN, et al. A novel mechanism of action of chemically modified tetracyclines: inhibition of cox-2-mediated prostaglandin e2 production. J Immunol. 1999;163(6):3459–3467.
  • Ismail CAN, Suppian R, Aziz CBA, et al. Minocycline attenuates the development of diabetic neuropathy by modulating dream and bdnf protein expression in rat spinal cord. J Diabetes Metab Disord. 2019;18(1):181–190.
  • Jin SX, Zhuang ZY, Woolf CJ, et al. P38 mitogen-activated protein kinase is activated after a spinal nerve ligation in spinal cord microglia and dorsal root ganglion neurons and contributes to the generation of neuropathic pain. J Neurosci. 2003;23(10):4017–4022.
  • Patel MK, Kaye AD, Urman RD, et al. Tanezumab: therapy targeting nerve growth factor in pain pathogenesis. J Anaesthesiol Clin Pharmacol. 2018;34(1):111–116.
  • Bramson C, Herrmann DN, Carey W, et al. Exploring the role of tanezumab as a novel treatment for the relief of neuropathic pain. Pain Med. 2015;16(6):1163–1176.
  • Chang DS, Hsu E, Hottinger DG, et al. Anti-nerve growth factor in pain management: current evidence. J Pain Res. 2016;9:373–383.
  • Watson JJ, Allen SJ, Dawbarn D, et al. Targeting nerve growth factor in pain: what is the therapeutic potential?. BioDrugs. 2008;22(6):349–359.
  • Bannwarth B, Kostine M. Targeting nerve growth factor (ngf) for pain management: what does the future hold for ngf antagonists?. Drugs. 2014;74(6):619–626.
  • Kan SL, Li Y, Ning GZ, et al. Tanezumab for patients with osteoarthritis of the knee: a meta-analysis. PLoS One. 2016;11(6):e0157105.
  • Chen J, Li J, Li R, et al. Efficacy and safety of tanezumab on osteoarthritis knee and hip pains: a meta-analysis of randomized controlled trials. Pain Med. 2017;18(2):374–385.
  • Webb MP, Helander EM, Menard BL, et al. Tanezumab: a selective humanized mab for chronic lower back pain. Ther Clin Risk Manag. 2018;14:361–367.
  • P2y12 nucleotide receptor antagonioss, including clopidogrel, for the treatment of stroke. Global Summit on Stroke, Birmingham, UK, 2015. https://www.longdom.org/proceedings/p2y12-nucleotide-receptor-antagonists-including-clopidogrel-for-the-treatment-of-stroke-16555.html
  • Neumann A, Müller CE, Namasivayam V, et al. P2y1-like nucleotide receptors—structures, molecular modeling, mutagenesis, and oligomerization. Wiley Interdiscip Rev Comput Mol Sci. 2020;10(4):e1464.
  • Inoue K. Atp receptors of microglia involved in pain. Novartis Found Symp. 2006;276: 263–272. discussion 273-281.
  • Watkins LR, Hutchinson MR, Rice KC, et al. The “toll” of opioid-induced glial activation: improving the clinical efficacy of opioids by targeting glia. Trends Pharmacol Sci. 2009;30(11):581–591.
  • Muccioli GG, Stella N. Microglia produce and hydrolyze palmitoylethanolamide. Neuropharmacology. 2008;54(1):16–22.
  • Esposito E, Paterniti I, Mazzon E, et al. Effects of palmitoylethanolamide on release of mast cell peptidases and neurotrophic factors after spinal cord injury. Brain Behav Immun. 2011;25(6):1099–1112.
  • Lo Verme J, Fu J, Astarita G, et al. The nuclear receptor peroxisome proliferator-activated receptor-alpha mediates the anti-inflammatory actions of palmitoylethanolamide. Mol Pharmacol. 2005;67(1):15–19.
  • Del Giorno R, Skaper S, Paladini A, et al. Palmitoylethanolamide in fibromyalgia: results from prospective and retrospective observational studies. Pain Ther. 2015;4(2):169–178.
  • Smart D, Jonsson KO, Vandevoorde S, et al. ‘Entourage’ effects of n-acyl ethanolamines at human vanilloid receptors. Comparison of effects upon anandamide-induced vanilloid receptor activation and upon anandamide metabolism. Br J Pharmacol. 2002;136(3):452–458.
  • De Petrocellis L, Davis JB, Di Marzo V, et al. Palmitoylethanolamide enhances anandamide stimulation of human vanilloid vr1 receptors. FEBS Lett. 2001;506(3):253–256.
  • Richardson JD, Vasko MR. Cellular mechanisms of neurogenic inflammation. J Pharmacol Exp Ther. 2002;302(3):839–845.
  • Cerrato S, Brazis P, della Valle MF, et al. Effects of palmitoylethanolamide on immunologically induced histamine, pgd2 and tnfalpha release from canine skin mast cells. Vet Immunol Immunopathol. 2010;133(1):9–15.
  • Loría F, Petrosino S, Mestre L, et al. Study of the regulation of the endocannabinoid system in a virus model of multiple sclerosis reveals a therapeutic effect of palmitoylethanolamide. Eur J Neurosci. 2008;28(4):633–641.
  • Genovese T, Esposito E, Mazzon E, et al. Effects of palmitoylethanolamide on signaling pathways implicated in the development of spinal cord injury. J Pharmacol Exp Ther. 2008;326(1):12–23.
  • Costa B, Conti S, Giagnoni G, et al. Therapeutic effect of the endogenous fatty acid amide, palmitoylethanolamide, in rat acute inflammation: inhibition of nitric oxide and cyclo-oxygenase systems. Br J Pharmacol. 2002;137(4):413–420.
  • Calignano A, La Rana G, Piomelli D, et al. Antinociceptive activity of the endogenous fatty acid amide, palmitylethanolamide. Eur J Pharmacol. 2001;419(2–3):191–198.
  • Mazzari S, Canella R, Petrelli L, et al. N-(2-hydroxyethyl)hexadecanamide is orally active in reducing edema formation and inflammatory hyperalgesia by down-modulating mast cell activation. Eur J Pharmacol. 1996;300(3):227–236.
  • Paladini A, Varrassi G, Bentivegna G, et al. Palmitoylethanolamide in the treatment of failed back surgery syndrome. Pain Res Treat. 2017;2017:1486010.
  • Scaturro D, Asaro C, Lauricella L, et al. Combination of rehabilitative therapy with ultramicronized palmitoylethanolamide for chronic low back pain: an observational study. Pain Ther. 2020;9(1):319–326.
  • Paladini A, Fusco M, Cenacchi T, et al. Palmitoylethanolamide, a special food for medical purposes, in the treatment of chronic pain: a pooled data meta-analysis. Pain Physician. 2016;19(2):11–24.
  • Mizuno T, Kurotani T, Komatsu Y, et al. Neuroprotective role of phosphodiesterase inhibitor ibudilast on neuronal cell death induced by activated microglia. Neuropharmacology. 2004;46(3):404–411.
  • Kawasaki A, Hoshino K, Osaki R, et al. Effect of ibudilast: a novel antiasthmatic agent, on airway hypersensitivity in bronchial asthma. J Asthma. 1992;29(4):245–252.
  • Liu J, Feng X, Yu M, et al. Pentoxifylline attenuates the development of hyperalgesia in a rat model of neuropathic pain. Neurosci Lett. 2007;412(3):268–272.
  • Lundblad R, Ekstrøm P, Giercksky KE, et al. Pentoxifylline improves survival and reduces tumor necrosis factor, interleukin-6, and endothelin-1 in fulminant intra-abdominal sepsis in rats. Shock. 1995;3(3):210–215.
  • Raghavendra V, Tanga F, Rutkowski MD, et al. Anti-hyperalgesic and morphine-sparing actions of propentofylline following peripheral nerve injury in rats: mechanistic implications of spinal glia and proinflammatory cytokines. Pain. 2003;104(3):655–664.
  • Tawfik VL, Nutile-McMenemy N, Lacroix-Fralish ML, et al. Efficacy of propentofylline, a glial modulating agent, on existing mechanical allodynia following peripheral nerve injury. Brain Behav Immun. 2007;21(2):238–246.
  • Frampton M, Harvey RJ, Kirchner V, et al. Propentofylline for dementia. Cochrane Database Syst Rev. 2003;2:Cd002853.
  • Sansone RA, Sansone LA. Pain, pain, go away: antidepressants and pain management. Psychiatry (Edgmont). 2008;5(12):16–19.
  • Coluzzi F, Mattia C. Mechanism-based treatment in chronic neuropathic pain: the role of antidepressants. Curr Pharm Des. 2005;11(23):2945–2960.
  • Maya Vetencourt JF, Sale A, Viegi A, et al. The antidepressant fluoxetine restores plasticity in the adult visual cortex. Science. 2008;320(5874):385–388.
  • Chollet F, Tardy J, Albucher JF, et al. Fluoxetine for motor recovery after acute ischaemic stroke (flame): a randomised placebo-controlled trial. Lancet Neurol. 2011;10(2):123–130.
  • Squillace S, Spiegel S, Salvemini D, et al. Targeting the sphingosine-1-phosphate axis for developing non-narcotic pain therapeutics. Trends Pharmacol Sci. 2020;41(11):851–867.
  • Food and Drug Administration: fda expands approval of gilenya to treat multiple sclerosis in pediatric patients. Rockville, Maryland: Food and Drug Administration; 2018. Available from: https://www.fda.gov/news-events/press-announcements/fda-expands-approval-gilenya-treat-multiple-sclerosis-pediatric-patients
  • Leung L, Cahill CM. Tnf-alpha and neuropathic pain–a review. J Neuroinflammation. 2010;7:27.
  • Hess A, Axmann R, Rech J, et al. Blockade of tnf-α rapidly inhibits pain responses in the central nervous system. Proc Natl Acad Sci U S A. 2011;108(9):3731–3736.
  • Schnitzer TJ, Easton R, Pang S, et al. Effect of tanezumab on joint pain, physical function, and patient global assessment of osteoarthritis among patients with osteoarthritis of the hip or knee: a randomized clinical trial. Jama. 2019;322(1):37–48.
  • Coluzzi F, Fornasari D, Pergolizzi J, et al. From acute to chronic pain: tapentadol in the progressive stages of this disease entity. Eur Rev Med Pharmacol Sci. 2017;21(7):1672–1683.
  • Kucyi A, Davis KD. The neural code for pain: from single-cell electrophysiology to the dynamic pain connectome. Neuroscientist. 2017;23(4):397–414.
  • Kucyi A, Davis KD. The dynamic pain connectome. Trends Neurosci. 2015;38(2):86–95.
  • Costigan M, Scholz J, Woolf CJ, et al. Neuropathic pain: a maladaptive response of the nervous system to damage. Annu Rev Neurosci. 2009;32(1):1–32.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.