Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1094 word(s) 1094 2021-11-17 05:20:58 |
2 format correct Meta information modification 1094 2022-01-13 01:56:13 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Eldridge, S. Chemotherapy-Induced Peripheral Neuropathy (CIPN). Encyclopedia. Available online: https://encyclopedia.pub/entry/18153 (accessed on 23 July 2024).
Eldridge S. Chemotherapy-Induced Peripheral Neuropathy (CIPN). Encyclopedia. Available at: https://encyclopedia.pub/entry/18153. Accessed July 23, 2024.
Eldridge, Sandy. "Chemotherapy-Induced Peripheral Neuropathy (CIPN)" Encyclopedia, https://encyclopedia.pub/entry/18153 (accessed July 23, 2024).
Eldridge, S. (2022, January 12). Chemotherapy-Induced Peripheral Neuropathy (CIPN). In Encyclopedia. https://encyclopedia.pub/entry/18153
Eldridge, Sandy. "Chemotherapy-Induced Peripheral Neuropathy (CIPN)." Encyclopedia. Web. 12 January, 2022.
Chemotherapy-Induced Peripheral Neuropathy (CIPN)
Edit

Chemotherapy-induced peripheral neuropathy (CIPN) is widely recognized as a potentially severe toxicity that often leads to dose reduction or discontinuation of cancer treatment. Symptoms may persist despite discontinuation of chemotherapy and quality of life can be severely compromised. The clinical symptoms of CIPN, and the cellular and molecular targets involved in CIPN, are just as diverse as the wide variety of anticancer agents that cause peripheral neurotoxicity.

chemotherapy-induced peripheral neuropathy (CIPN)

1. Introduction

Chemotherapy-induced peripheral neuropathy (CIPN) is an adverse consequence of a wide variety of commonly used anticancer agents [1][2][3][4][5][6][7][8][9] and there are no gold standard therapeutics recommended for the prevention or treatment of CIPN [10]. CIPN frequently leads to dose reduction or discontinuation of therapy [4][11][12]. Clinical symptoms can persist long after completion of chemotherapy and severely diminish the quality of life of patients [1][13]. The pathophysiology of CIPN is complex and compounded by the fact that the various neurotoxic events culminating in CIPN are not necessarily related to the anticancer mechanisms of action for the agents that cause CIPN [9][14]. However, several lines of evidence point toward interactions involving various target components of the peripheral nervous system (PNS), including dorsal root ganglion (DRG), myelin, microtubules, mitochondria, ion channels, blood vessels, and nerve terminals [5][9][15][16][17][18]. A common pathology in CIPN is a “dying back” axon degeneration of distal nerve endings [9][19]

2. Anticancer Agents That Cause CIPN

CIPN is a debilitating adverse effect with a prevalence ranging from 19% to over 85% [8] and caused by a spectrum of classes of widely used anticancer therapeutics including platinum-based agents, microtubule disruptors (taxanes and vinca alkaloids), proteasome, and angiogenesis inhibitors (Table 1) [1][2][3][4][5][7][8][9][20][21][22]. Clinically, CIPN symptoms may be acute, worsen with cumulative drug dosing, or emerge late during the course of treatment, even long after cessation of treatment [18]. Although many genetic and clinical risk factors have been identified, CIPN surveillance during and post-chemotherapy is needed as well as further study to better understand the pathophysiology of CIPN [8][18].
Table 1. Anticancer agents known to cause CIPN and their proposed mechanisms and target sites of CIPN toxicity [1][2][3][4][5][7][8][9][20][21][22].
Class Agents Proposed Mechanism Main Target of CIPN Toxicity
Taxanes Paclitaxel
Docetaxel
Ixabepilone
Microtubule disruption Dorsal root ganglion; axons; distal nerve terminals
Platinum-based Cisplatin
Carboplatin
Oxaliplatin
DNA adducts Dorsal root ganglion
Alkylating agents Cyclophosphamide
Hexamethylmelamine
Ifosphamide
Procarbazine
Covalently bind to DNA Dorsal root ganglion
Vinca alkaloids Vincristine
Vinblastine
Vinorelbine
Vindesine
Dysfunction of mitochondria and endoplasmic reticulum; microtubule disruption Dorsal root ganglion; distal nerve terminals
Proteasome inhibitors Bortezomib
Carfilzomib
Ixazomib
Binds proteasome complex; mitochondrial disturbance; microtubule disruption Dorsal root ganglion and peripheral nerves
Immunomodulatory Thalidomide
Lenalidomide
Pomalidomide
Antiangiogenesis Dorsal root ganglion; distal nerve terminals
Among commonly used classes of cancer therapies for many blood and solid tumors, platinum analogs (e.g., cisplatin and oxaliplatin), proteasome inhibitors (e.g., bortezomib), immunomodulatory/antiangiogenic (e.g., thalidomide), and taxanes (e.g., paclitaxel) have markedly different chemical structures and mechanisms of actions. However, they all share a common adverse side effect: CIPN [16]. Clinically, CIPN involves the PNS that predominately leads to sensory axonal peripheral neuropathy characterized by a “stocking and glove” distribution of a plethora of potentially debilitating sensory effects [5][17]. Although the proposed pathogenesis of CIPN involves the cell bodies of the DRG concomitant with dying back axonal damage, the exact pathophysiology remains elusive [19][22]. Evidence suggests that neurotoxic chemotherapy drugs may involve various cellular components in the PNS by (1) forming DNA adducts, DNA damage, and alterations in DNA repair; (2) stabilization/disruption of microtubules; (3) targeting mitochondria; (4) functionally impairing ion channels; (5) production of oxidative stress; (6) dysregulation of calcium signaling; (7) altering cell signaling events; and/or (8) triggering immunological mechanisms through activation of satellite glial cells [9][14][15][16][17][18]. Development of an in vitro human peripheral neuronal cell model in which these various cellular components can be investigated will provide an urgently needed tool to dissect the cellular and molecular effects of potentially neurotoxic compounds.
The DRG of the PNS is vulnerable to neurotoxic damage since it is less protected by the blood–nerve barrier than the CNS [23][24]. This may partially explain the predominance of sensory involvement in patients with CIPN [9]. Platinum compounds form DNA adducts that can accumulate in the DRG [25][26], potentially leading to sensory neuronal cell death [27][28]. Paclitaxel has been reported to accumulate in the DRG through transmembrane transport mediated specifically by organic anion transporting polypeptide 1B (OATP1B) [29].
Central to the transport of proteins from the nerve cell body, down the length of the axon are microtubules [3]. A commonly used class of anticancer agents, taxanes, are microtubule binding agents, which produce polymerization that interferes with normal microtubule dynamics linked to disruption of axonal transport [21][30][31]. Another class of chemotherapy agents that cause CIPN are vinca alkaloids (vincristine, vinblastine, vinorelbine, and vinderine) that also bind tubulin and inhibit microtubule dynamics, leading to interference with the mitotic spindle [32]. A proteasome inhibitor, bortezomib, also affects microtubule polymerization independent of its mechanism as an anticancer agent [9][33].
Damage to the mitochondria that impairs mitochondrial function may play a pivotal role in CIPN [9][34]. For example, paclitaxel has been reported to cause functional impairment in axonal mitochondria [35]. Additionally, bortezomib has also been shown to cause accumulation of ubiquitin-conjugated proteins, mitochondrial dysfunction in peripheral sensory neurons (PNS) including Schwann cells, and endoplasmic reticulum stress particularly in Schwann cells [9][36][37][38].
There is reported evidence for direct toxicity to the distal axon terminals associated with peripheral neuropathy following cancer treatment with paclitaxel [39], thalidomide [40], and vincristine [41]. Oxaliplatin may affect the function of voltage-gated sodium (Na+) ion channels, inducing an acute peripheral neuropathy manifested by hyperexcitability [42][43][44].
Thalidomide is also associated with peripheral neuropathy through different proposed mechanisms [45]. Thalidomide-induced peripheral neuropathy is proposed to be mediated by its antiangiogenic effects [46]. Notably, attempts to establish a thalidomide rodent model both in vivo and in vitro have not been successful [9][47][48].
Although the underlying mechanisms responsible for the development of CIPN remain elusive and are further complicated by the diversity of anticancer agents that cause CIPN, there may be common degenerative pathways triggered when normal cellular processes and energy delivery mechanisms of the PNS become disrupted [9]. It is important to appreciate that mechanisms of CIPN may be shared by different classes of chemotherapeutic agents independent of their anticancer properties [3][4][5][6][7][8][9][12]. Some pathways, such as those activated by the mitogen-activated protein kinase (MAPK) family members and by mechanistic target of rapamycin (mTOR), could represent a common core of CIPN pathophysiology, as they have been demonstrated to be closely related to hyperalgesia and more in general to pain, a hallmark of CIPN [49][50][51][52][53]. The activation of MAPKs, and in particular of p38, has been observed to have a pivotal role in CIPN induced by different chemotherapeutic agents, such as paclitaxel, oxaliplatin, cisplatin, and vincristine [50][51][52][53] through their relation with toll-like receptor (TLR4) and nuclear factor kappa B (NF-kB) signaling pathways, not only in DRG neurons, but also in glial cells [49][50].

References

  1. Argyriou, A.A.; Kyritsis, A.P.; Makatsoris, T.; Kalofonos, H.P. Chemotherapy-induced peripheral neuropathy in adults: A comprehensive update of the literature. Cancer Manag. Res. 2014, 6, 135–147.
  2. Cioroiu, C.; Weimer, L.H. Update on Chemotherapy-Induced Peripheral Neuropathy. Curr. Neurol. Neurosci. Rep. 2017, 17, 47.
  3. Ferrier, J.; Pereira, V.; Busserolles, J.; Authier, N.; Balayssac, D. Emerging trends in understanding chemotherapy-induced peripheral neuropathy. Curr. Pain Headache Rep. 2013, 17, 364.
  4. Miltenburg, N.C.; Boogerd, W. Chemotherapy-induced neuropathy: A comprehensive survey. Cancer Treat. Rev. 2014, 40, 872–882.
  5. Park, S.B.; Goldstein, D.; Krishnan, A.V.; Lin, C.S.; Friedlander, M.L.; Cassidy, J.; Koltzenburg, M.; Kiernan, M.C. Chemotherapy-induced peripheral neurotoxicity: A critical analysis. CA Cancer J. Clin. 2013, 63, 419–437.
  6. Staff, N.P.; Grisold, A.; Grisold, W.; Windebank, A.J. Chemotherapy-induced peripheral neuropathy: A current review. Ann. Neurol. 2017, 81, 772–781.
  7. Starobova, H.; Vetter, I. Pathophysiology of Chemotherapy-Induced Peripheral Neuropathy. Front. Mol. Neurosci. 2017, 10, 174.
  8. Zajączkowska, R.; Kocot-Kępska, M.; Leppert, W.; Wrzosek, A.; Mika, J.; Wordliczek, J. Mechanisms of Chemotherapy-Induced Peripheral Neuropathy. Int. J. Mol. Sci. 2019, 20, 1451.
  9. Eldridge, S.; Guo, L.; Hamre, J., III. A Comparative Review of Chemotherapy-Induced Peripheral Neuropathy in In Vivo and In Vitro Models. Toxicol. Pathol. 2020, 48, 190–201.
  10. Loprinzi, C.L.; Lacchetti, C.; Bleeker, J.; Cavaletti, G.; Chauhan, C.; Hertz, D.L.; Kelley, M.R.; Lavino, A.; Lustberg, M.B.; Paice, J.A.; et al. Prevention and Management of Chemotherapy-Induced Peripheral Neuropathy in Survivors of Adult Cancers: ASCO Guideline Update. J. Clin. Oncol. 2020, 38, 3325–3348.
  11. Fehrenbacher, J.C. Chemotherapy-Induced Peripheral Neuropathy. In Progress in Molecular Biology and Translational Science; Price, T.J., Dussor, G., Eds.; Academic Press: Cambridge, MA, USA, 2015; Chapter 16; Volume 131, pp. 471–508.
  12. Han, Y.; Smith, M.T. Pathobiology of cancer chemotherapy-induced peripheral neuropathy (CIPN). Front. Pharmacol. 2013, 4, 156.
  13. Windebank, A.J.; Grisold, W. Chemotherapy-induced neuropathy. J. Peripher. Nerv. Syst. 2008, 13, 27–46.
  14. Hu, S.; Huang, K.M.; Adams, E.J.; Loprinzi, C.L.; Lustberg, M.B. Recent Developments of Novel Pharmacologic Therapeutics for Prevention of Chemotherapy-Induced Peripheral Neuropathy. Clin. Cancer Res. 2019, 25, 6295–6301.
  15. Balayssac, D.; Ferrier, J.; Descoeur, J.; Ling, B.; Pezet, D.; Eschalier, A.; Authier, N. Chemotherapy-induced peripheral neuropathies: From clinical relevance to preclinical evidence. Expert Opin. Drug Saf. 2011, 10, 407–417.
  16. Carozzi, V.A.; Canta, A.; Chiorazzi, A. Chemotherapy-induced peripheral neuropathy: What do we know about mechanisms? Neurosci. Lett. 2015, 596, 90–107.
  17. Hausheer, F.H.; Schilsky, R.L.; Bain, S.; Berghorn, E.J.; Lieberman, F. Diagnosis, management, and evaluation of chemotherapy-induced peripheral neuropathy. Semin. Oncol. 2006, 33, 15–49.
  18. Seretny, M.; Currie, G.L.; Sena, E.S.; Ramnarine, S.; Grant, R.; MacLeod, M.R.; Colvin, L.A.; Fallon, M. Incidence, prevalence, and predictors of chemotherapy-induced peripheral neuropathy: A systematic review and meta-analysis. Pain 2014, 155, 2461–2470.
  19. Fukuda, Y.; Li, Y.; Segal, R.A. A Mechanistic Understanding of Axon Degeneration in Chemotherapy-Induced Peripheral Neuropathy. Front. Neurosci. 2017, 11, 481.
  20. Addington, J.; Freimer, M. Chemotherapy-induced peripheral neuropathy: An update on the current understanding. F1000Research 2016, 5, 1466.
  21. Brzezinski, K. Chemotherapy-induced polyneuropathy. Part I. Pathophysiology. Contemp. Oncol. 2012, 16, 72–78.
  22. Grisold, W.; Cavaletti, G.; Windebank, A.J. Peripheral neuropathies from chemotherapeutics and targeted agents: Diagnosis, treatment, and prevention. Neuro-Oncology 2012, 14, iv45–iv54.
  23. Allen, D.; Kiernan, J. Permeation of proteins from the blood into peripheral nerves and ganglia. Neuroscience 1994, 59, 755–764.
  24. Reinhold, A.K.; Rittner, H.L. Barrier function in the peripheral and central nervous system—A review. Pflügers Arch. Eur. J. Physiol. 2017, 469, 123–134.
  25. Gregg, R.W.; Molepo, J.M.; Monpetit, V.J.; Mikael, N.Z.; Redmond, D.; Gadia, M.; Stewart, D.J. Cisplatin neurotoxicity: The relationship between dosage, time, and platinum concentration in neurologic tissues, and morphologic evidence of toxicity. J. Clin. Oncol. 1992, 10, 795–803.
  26. Krarup-Hansen, A.; Rietz, B.; Krarup, C.; Heydorn, K.; Rorth, M.; Schmalbruch, H. Histology and platinum content of sensory ganglia and sural nerves in patients treated with cisplatin and carboplatin: An autopsy study. Neuropathol. Appl. Neurobiol. 1999, 25, 29–40.
  27. Dzagnidze, A.; Katsarava, Z.; Makhalova, J.; Liedert, B.; Yoon, M.S.; Kaube, H.; Limmroth, V.; Thomale, J. Repair capacity for platinum-DNA adducts determines the severity of cisplatin-induced peripheral neuropathy. J. Neurosci. 2007, 27, 9451–9457.
  28. Ta, L.E.; Espeset, L.; Podratz, J.; Windebank, A.J. Neurotoxicity of oxaliplatin and cisplatin for dorsal root ganglion neurons correlates with platinum-DNA binding. Neurotoxicology 2006, 27, 992–1002.
  29. Leblanc, A.F.; Sprowl, J.A.; Alberti, P.; Chiorazzi, A.; Arnold, W.D.; Gibson, A.A.; Hong, K.W.; Pioso, M.S.; Chen, M.; Huang, K.M.; et al. OATP1B2 deficiency protects against paclitaxel-induced neurotoxicity. J. Clin. Investig. 2018, 128, 816–825.
  30. LaPointe, N.E.; Morfini, G.; Brady, S.T.; Feinstein, S.C.; Wilson, L.; Jordan, M.A. Effects of eribulin, vincristine, paclitaxel and ixabepilone on fast axonal transport and kinesin-1 driven microtubule gliding: Implications for chemotherapy-induced peripheral neuropathy. Neurotoxicology 2013, 37, 231–239.
  31. Theiss, C.; Meller, K. Taxol impairs anterograde axonal transport of microinjected horseradish peroxidase in dorsal root ganglia neurons in vitro. Cell Tissue Res. 2000, 299, 213–224.
  32. Topp, K.S.; Tanner, K.D.; Levine, J.D. Damage to the cytoskeleton of large diameter sensory neurons and myelinated axons in vincristine-induced painful peripheral neuropathy in the rat. J. Comp. Neurol. 2000, 424, 563–576.
  33. Arastu-Kapur, S.; Anderl, J.L.; Kraus, M.; Parlati, F.; Shenk, K.D.; Lee, S.J.; Muchamuel, T.; Bennett, M.K.; Driessen, C.; Ball, A.J.; et al. Nonproteasomal targets of the proteasome inhibitors bortezomib and carfilzomib: A link to clinical adverse events. Clin. Cancer Res. 2011, 17, 2734–2743.
  34. Canta, A.; Pozzi, E.; Carozzi, V.A. Mitochondrial Dysfunction in Chemotherapy-Induced Peripheral Neuropathy (CIPN). Toxics 2015, 3, 198–223.
  35. Flatters, S.J.L.; Bennett, G.J. Studies of peripheral sensory nerves in paclitaxel-induced painful peripheral neuropathy: Evidence for mitochondrial dysfunction. Pain 2006, 122, 245–257.
  36. Casafont, I.; Berciano, M.T.; Lafarga, M. Bortezomib induces the formation of nuclear poly(A) RNA granules enriched in Sam68 and PABPN1 in sensory ganglia neurons. Neurotox. Res. 2010, 17, 167–178.
  37. Cavaletti, G.; Gilardini, A.; Canta, A.; Rigamonti, L.; Rodriguez-Menendez, V.; Ceresa, C.; Marmiroli, P.; Bossi, M.; Oggioni, N.; D’Incalci, M.; et al. Bortezomib-induced peripheral neurotoxicity: A neurophysiological and pathological study in the rat. Exp. Neurol. 2007, 204, 317–325.
  38. Shin, Y.K.; Jang, S.Y.; Lee, H.K.; Jung, J.; Suh, D.J.; Seo, S.Y.; Park, H.T. Pathological adaptive responses of Schwann cells to endoplasmic reticulum stress in bortezomib-induced peripheral neuropathy. Glia 2010, 58, 1961–1976.
  39. Melli, G.; Jack, C.; Lambrinos, G.L.; Ringkamp, M.; Hoke, A. Erythropoietin protects sensory axons against paclitaxel-induced distal degeneration. Neurobiol. Dis. 2006, 24, 525–530.
  40. Isoardo, G.; Bergui, M.; Durelli, L.; Barbero, P.; Boccadoro, M.; Bertola, A.; Ciaramitaro, P.; Palumbo, A.; Bergamasco, B.; Cocito, D. Thalidomide neuropathy: Clinical, electrophysiological and neuroradiological features. Acta Neurol. Scand. 2004, 109, 188–193.
  41. Silva, A.; Wang, Q.; Wang, M.; Ravula, S.K.; Glass, J.D. Evidence for direct axonal toxicity in vincristine neuropathy. J. Peripher. Nerv. Syst. 2006, 11, 211–216.
  42. Adelsberger, H.; Quasthoff, S.; Grosskreutz, J.; Lepier, A.; Eckel, F.; Lersch, C. The chemotherapeutic oxaliplatin alters voltage-gated Na(+) channel kinetics on rat sensory neurons. Eur. J. Pharmacol. 2000, 406, 25–32.
  43. Sittl, R.; Lampert, A.; Huth, T.; Schuy, E.T.; Link, A.S.; Fleckenstein, J.; Alzheimer, C.; Grafe, P.; Carr, R.W. Anticancer drug oxaliplatin induces acute cooling-aggravated neuropathy via sodium channel subtype Na(V)1.6-resurgent and persistent current. Proc. Natl. Acad. Sci. USA 2012, 109, 6704–6709.
  44. Webster, R.G.; Brain, K.L.; Wilson, R.H.; Grem, J.L.; Vincent, A. Oxaliplatin induces hyperexcitability at motor and autonomic neuromuscular junctions through effects on voltage-gated sodium channels. Br. J. Pharmacol. 2005, 146, 1027–1039.
  45. Giannini, F.; Volpi, N.; Rossi, S.; Passero, S.; Fimiani, M.; Cerase, A. Thalidomide-induced neuropathy: A ganglionopathy? Neurology 2003, 60, 877–878.
  46. Kirchmair, R.; Tietz, A.B.; Panagiotou, E.; Walter, D.H.; Silver, M.; Yoon, Y.S.; Schratzberger, P.; Weber, A.; Kusano, K.; Weinberg, D.H.; et al. Therapeutic angiogenesis inhibits or rescues chemotherapy-induced peripheral neuropathy: Taxol- and thalidomide-induced injury of vasa nervorum is ameliorated by VEGF. Mol. Ther. 2007, 15, 69–75.
  47. Guo, L.; Hamre, J., III; Eldridge, S.; Behrsing, H.P.; Cutuli, F.M.; Mussio, J.; Davis, M. Editor’s Highlight: Multiparametric Image Analysis of Rat Dorsal Root Ganglion Cultures to Evaluate Peripheral Neuropathy-Inducing Chemotherapeutics. Toxicol. Sci. 2017, 156, 275–288.
  48. Hoke, A.; Ray, M. Rodent models of chemotherapy-induced peripheral neuropathy. ILAR J. 2014, 54, 273–281.
  49. Inyang, K.E.; McDougal, T.A.; Ramirez, E.D.; Williams, M.; Laumet, G.; Kavelaars, A.; Heijnen, C.J.; Burton, M.; Dussor, G.; Price, T.J. Alleviation of paclitaxel-induced mechanical hypersensitivity and hyperalgesic priming with AMPK activators in male and female mice. Neurobiol. Pain 2019, 6, 100037.
  50. Li, Y.; Zhang, H.; Kosturakis, A.K.; Cassidy, R.M.; Zhang, H.; Kennamer-Chapman, R.M.; Jawad, A.B.; Colomand, C.M.; Harrison, D.S.; Dougherty, P.M. MAPK signaling downstream to TLR4 contributes to paclitaxel-induced peripheral neuropathy. Brain Behav. Immun. 2015, 49, 255–266.
  51. Meng, J.; Zhang, Q.; Yang, C.; Xiao, L.; Xue, Z.; Zhu, J. Duloxetine, a Balanced Serotonin-Norepinephrine Reuptake Inhibitor, Improves Painful Chemotherapy-Induced Peripheral Neuropathy by Inhibiting Activation of p38 MAPK and NF-kappaB. Front. Pharmacol. 2019, 10, 365.
  52. Qin, B.; Li, Y.; Liu, X.; Gong, D.; Zheng, W. Notch activation enhances microglial CX3CR1/P38 MAPK pathway in rats model of vincristine-induced peripheral neuropathy. Neurosci. Lett. 2020, 715, 134624.
  53. Reyes-Gibby, C.C.; Wang, J.; Yeung, S.J.; Shete, S. Informative gene network for chemotherapy-induced peripheral neuropathy. BioData Min. 2015, 8, 24.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 557
Entry Collection: Neurodegeneration
Revisions: 2 times (View History)
Update Date: 13 Jan 2022
1000/1000
Video Production Service