2,522
Views
11
CrossRef citations to date
0
Altmetric
Neurology

Chemotherapy-induced peripheral neuropathy: limitations in current prophylactic strategies and directions for future research

ORCID Icon
Pages 981-984 | Received 10 Oct 2016, Accepted 16 Jan 2017, Published online: 15 Feb 2017

Introduction

Different types of neurologic complications, including central neurotoxicity conditions ranging from minor cognitive deficits to encephalopathy with dementia or even coma, and peripheral neurotoxicity, may be associated with antineoplastic drug therapy. Peripheral neuropathy is the most common neurological complication of cancer treatment, representing a set of symptoms ranging from minor, and temporary symptoms, to severe and permanent forms of polyneuropathy. Because this kind of neurotoxicity is due to the administration of anticancer drugs it is commonly indicated as chemotherapy-induced peripheral neuropathy (CIPN).

Epidemiology

CIPN is a common treatment-related adverse effect, affecting up to 48% of cancer patients who received chemotherapyCitation1, and this serious complication can negatively impact the long-term quality of life (QoL) of cancer survivors, because up to 40% of these individuals may experience permanent symptoms, mainly as paresthesia, but also as pain and other types of disability, after the end of the therapeutic courseCitation2,Citation3.

Anticancer drugs involved

Many anticancer drugs, mostly platinum compounds, antitubulins (taxanes, vinca alkaloids, eribulin), and proteasome inhibitors (bortezomib), but also immunomodulatory agents (thalidomide, lenalidomide, pomalidomide), are known to cause peripheral neuropathyCitation4. Other drugs, such as etoposide, methotrexate, 5-fluorouracile (5-FU), and gemcitabine are less commonly or not associated with CIPN, whereas the use of biological agents has also been occasionally associated with CIPN. For instance, about 50% of patients with relapsed or refractory Hodgkin’s lymphoma treated with brentuximab vedotin developed mild sensory neuropathy, although mostly not clinically relevantCitation5. The role of synergistic neurotoxicity caused by previously given chemotherapies and concomitant chemotherapies is not clear, indeed approximately 6% of patients treated with the biologic agent alemtuzumab, combined with fludarabine and melphalan, developed a progressive sensorimotor radiculoneuropathy and/or a myelitisCitation6, and up to 40% of patients who received cisplatin and taxol developed CIPNCitation7. Again, 92% of patients underwent the drug combination FOLFOX (folinic acid, 5-FU and oxaliplatin) suffered by sensory CIPNCitation8.

Clinical features

Although CIPN is generally a temporary manifestation, it may represent a permanent side effect from the chemotherapy treatment employed, and painful CIPN can also persist from months to years after the chemotherapy conclusion, leading to significant challenges for cancer survivors because of its negative influence on functional activity and QoLCitation9. A frequent question asked by patients regards the time course of symptom resolution. We can respond using data from the most important trials published on the topic, assuming that clinical features depend on several conditions, including chemotherapy regimens, duration of exposure, and assessment methods. Thus, the APEX trial showed that of patients with grade 2, or higher, bortezomib-induced peripheral neuropathy, 64% experienced improvement or resolution to baseline at a median of 110 daysCitation10, whereas cumulative oxaliplatin-induced peripheral neuropathy is reported to be partially reversible in approximately 80% of patients and completely resolves in approximately 40%, at 6 to 8 months after cessation of treatmentCitation11.

Prophylactic agents proposed

American Society of Clinical Oncology (ASCO) on the topic, based on a systematic review of 42 randomized controlled trials (RCTs) investigating 18 agents, found that there are no agents that have shown consistent clinically meaningful benefits for CIPN preventionCitation12. Moreover, a Cochrane review states that chemoprotective agents do not seem to prevent CIPN caused by cisplatin and related compoundsCitation13.

Furthermore, several preclinical and clinical investigations have been conducted to verify the neuroprotective effects of a wide range of agents, such as glutathioneCitation14, amifostineCitation15, N-acetylcysteineCitation16, the calcium-channel antagonist nimodipineCitation17, nutraceuticals, such as glutamineCitation18, acetyl-L-carnitineCitation19, vitamin ECitation20, omega-3 fatty acidsCitation21, all-trans retinoic acidCitation22, calcium and magnesiumCitation23, curcuminCitation24, quercetinCitation25, matricaria chamomillaCitation25, ginkgo biloba extractCitation26, green teaCitation27, the aromatic plant in the family Lamiaceae ocimum sanctumCitation28, acorus calamusCitation29 and salvia officinalisCitation30, as well as the Kampo medicine GoshajinkiganCitation31 and other herbal combinationsCitation32. In addition, anti-seizure drugs, such as carbamazepine and pregabalinCitation33 and antidepressants, such as venlafaxine, are ineffective or seem to have more results in the treatment of CIPN rather than in its preventionCitation34. In Argyriou et al.’s analysis oxcarbazepine has been shown to be modestly effective in prophylaxis against cumulative oxaliplatin-induced neuropathyCitation35. Briefly, there are no effective agents, or protocol, with strong evidence of effectiveness to prevent this serious treatment-related toxicity.

Drug regimen strategies

CIPN prevention mainly consists of several proposed strategies, including the application of a stop-and-go regimen, consisting in chemotherapy (e.g. oxiplatinum) free intervals to reduce advanced sensory neuropathyCitation36, cumulative dose reduction, or the use of lower dose intensities, especially in patients which are at higher risk to develop neurotoxic side effects, or in patients which already have neuropathic symptoms due to diabetes mellitus, hereditary neuropathies, or earlier treatment with neurotoxic chemotherapy.

This background is particularly important as it introduces two questions. Why does CIPN represent such a paramount side effect, so difficult to prevent and/or to treat? And, what are promising future directions for research in this field?

The response to the first question must be addressed through the explanation of CIPN mechanisms. While much research has been conducted on the topic, the precise pathophysiology of CIPN has not yet been defined. Probably, the key factor lies within the clinical observation that any group of anticancer drugs exhibits a different mechanism of damage to the peripheral nervous systemCitation37. Thus, the pathogenetic chain of CIPN could comprehend initial steps that differ between different chemotherapies – which would explain the differences in clinical manifestations – and ending stages with many points in common.

Usually, CIPN manifests itself after a typical cumulative dose (dose-dependent mechanism) but, in some cases, such as those following the use of oxaliplatin, or taxanes, immediate toxic effects may occur (acute neurotoxicity). Thus, several drug-dependent pathogenetic mechanisms exist, whereas different mechanisms may be involved in the development of the clinical manifestations (acute/chronic) due to the same agent. For instance, the proposed mechanism of action of acute oxaliplatin-induced CIPN includes alteration in the current of voltage-gated sodium channels in response to oxalate, a metabolic by-product of oxaliplatin, as well as the indirect interaction of oxalate with voltage-gated sodium channels through chelation of calcium and magnesiumCitation38. On the other hand, it seems that chronic oxaliplatin-induced CIPN could be caused by a dose-dependent accumulation of platinum compounds in the dorsal root ganglia (DRG) causing neuronal atrophy and apoptosis, indeed studies have demonstrated a strong affinity of platinum compounds to the deoxyribonucleic acid (DNA) of spinal ganglion cellsCitation39.

With respect to taxanes, the so-called paclitaxel acute pain syndrome, which is an expression of acute neurotoxicity, has been linked with a sensitization of nociceptors and their fibers by proinflammatory cytokines (IL-6, IL-8, IL1β, TNF-α), whereas the promotion of microtubule polymerization and inhibition of depolymerization, leading to inhibition of axonal transport within neurons, are involved in dose-dependent taxane neurotoxicityCitation40.

Furthermore, preclinical studies suggest that peripheral neuropathy due to proteasome inhibitors (e.g. bortezomib) is probably the result of different pathogenetic mechanisms, including pathological changes in Schwann cells and myelin – affecting the flow of information related to proprioception, touch, pain and temperature through myelinated (Aα, Aβ, Aδ) fibers – combined with axonal degeneration in all major primary afferent myelinated and unmyelinated (C) fibersCitation41, and DRG neuron changes, manifested as nuclear and nucleolar atrophy and/or loss of neuronsCitation42.

But explaining the CIPN pathogenesis is a very complex matter, because other variables, such as other cancer treatments, like surgery or radiation, neural compression or invasion by the tumor, diabetes, alcohol abuse, low vitamin B levels, and peripheral vascular disease come into play. Moreover, the results of Schneider’s and co-authors’ studyCitation43 showed a significant association between neuropathy and African-American race, and some genetic studies suggested that certain genes were related to CIPN. A very interesting review on the topic focused on several potential genes associated with CIPNCitation44. These genes are involved in different functions, including regulation of the intracellular drug concentrations (GSTP1, GSTM1 and ABCB1), response to DNA damage (ERCC1, FANCD2, BCL2, and SOX10), cellular stress response (BCL2), inflammation processes (ABCC1, ABCC2, ABCG2, ITGA1, ITGB3, TAC1, ABCB1, ABCC2, EPHA4, EPHA6, SLCO1B1, TUBB2A, ABCA1, BCL2, OPRM1 and TRPV1), and neuronal plasticity (ERCC1 and TAC1)Citation44. However, until now, no conclusive data has been found, and the precise gene–phenotype association is not fully understood.

Whatever the precise mechanism(s), neurotoxic chemotherapy agents usually induce mitochondrial dysfunction – as suggested by the loss of mitochondrial mobility in bortezomib, paclitaxel, and vinca alkaloid – with progressive axonal neuropathy and combined DRG neuronal cell body changes, which are the last steps of the pathogenetic chain.

On these bases, research should be finalized: 1) to better understand all the steps of the pathogenetic mechanisms of CIPN; 2) to identify patients who are at high risk for CIPN; 3) to explore more about molecule or combinations, already tested; 4) to develop novel strategies in order to offer more effectiveness drug molecules for the future; 5) to design more effectiveness protocols.

Research on CIPN mechanisms is a flourishing field of study. For instance, several lines of evidence support the notion that DNA damage may be a causative factor in CIPN induced by a number of cancer therapies. Thus, specific targeted molecules, such as APX2009 and APX3330, which improve the activity of apurinic/apyrimidinic endonuclease APE-1, involved in base excision repair, could be effective in preventing or reversing CIPNCitation45,Citation46. The results of clinical trials (APX3330 has been approved for phase 1 clinical investigations), and further in vitro and in vivo studies, could represent a significant step forward in the battle against CIPN.

Moreover, further extensive preclinical and clinical studies need to be conducted with the aim of completing the safety and the effectiveness profiles of certain substances, such as nutraceuticals, already tested. In case of complex herbal medicines more controlled investigations should better elucidate the pharmacokinetics and pharmacodynamics of the single compounds, also studying potential interactions of these agents with chemotherapies and adjuvants, as well as evaluating long term results.

On the other hand, research is currently aimed at testing new molecules. For instance, the endogenous lipid palmitoylethanolamide, alone or in combination with the anticonvulsant gabapentin, could be effective for this purpose, as a preclinical investigation showed that this strategy was able to reduce allodynia in a mouse paclitaxel model of CIPNCitation47, whereas several antioxidants, such as ethoxyquinCitation48 have been also tested.

At the moment very few options are available for CIPN prevention, which mainly consists of several proposed strategies, including the application of a stop-and-go regimen, cumulative dose reduction, or the use of lower dose intensities. The dearth of effective prevention strategies for CIPN represents a big lack in oncology as this side effect can lead to dose reduction of the chemotherapy agent, or possible cessation of treatment, which may have an adverse impact on cancer treatment and disease outcomes. On the other hand, although CIPN prevalence decreases with time, at 6 months 30% of patients continue to suffer from CIPN1. Thus, the elevated incidence of CIPN and the conspicuous number of cancer survivors calls for a rapid resolution of the phenomenon through the search for more effective prophylactic approaches.

Marco Cascella

Division of Anesthesia, Department of Anesthesia, Endoscopy and Cardiology Istituto Nazionale Tumori “Fondazione G. Pascale” – IRCCS, Naples, Italy

[email protected]

Transparency

Declaration of funding

This editorial was not funded.

Declaration of financial/other relationships

M.C. has disclosed that he has no significant relationships with or financial interests in any commercial companies related to this article.

CMRO peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Acknowledgments

I would like to express my sincere gratitude to Dr. Maria Rosaria Muzio for the immeasurable amount of support and guidance she has provided throughout this paper.

References

  • Seretny M, Currie GL, Sena, et al. Incidence, prevalence, and predictors of chemotherapy-induced peripheral neuropathy: a systematic review and meta-analysis. Pain 2014;155:2461-70
  • Park S, Goldstein D, Krishnan, et al. Chemotherapy-induced peripheral neurotoxicity: a critical analysis. CA Cancer J Clin 2013;63:419-37
  • Smith EM, Cohen JA, Pett MA, et al. The reliability and validity of a modified total neuropathy score-reduced and neuropathic pain severity items when used to measure chemotherapy-induced peripheral neuropathy in patients receiving taxanes and platinums. Cancer Nurs 2010;33:173-83
  • Cavaletti G, Marmiroli P. Chemotherapy-induced peripheral neurotoxicity. Curr Opin Neurol 2015;28:500-7
  • Younes A, Gopal AK, Smith SE, et al. Results of a pivotal phase II study of brentuximab vedotin for patients with relapsed or refractory Hodgkin’s lymphoma. J Clin Oncol 2012;30:2183-9
  • Avivi I, Chakrabarti S, Kottaridis P, et al. Neurological complications following alemtuzumab-based reduced-intensity allogeneic transplantation. Bone Marrow Transplant 2004;34:137-42
  • International Collaborative Ovarian NeoPlasm Group. Paclitaxel plus carboplatin versus standard chemotherapy with either single-agent carboplatin or cyclophosphamide, doxorubicin, and cisplatin in women with ovarian cancer: the ICON3 randomised trial. Lancet 2002;360:505-15
  • Denlinger CS, Barsevick AM. Challenges of colorectal cancer survivorship. J Natl Compr Canc Netw 2009;7:883-93
  • Tanay MA, Armes J, Ream E. The experience of chemotherapy-induced peripheral neuropathy in adult cancer patients: a qualitative thematic synthesis. Eur J Cancer Care (Engl) 2016: published online 20 January 2016, doi: 10.1111/ecc.12443
  • Richardson PG, Sonneveld P, Schuster MW, et al. Reversibility of symptomatic peripheral neuropathy with bortezomib in the phase III APEX trial in relapsed multiple myeloma: impact of a dose-modification guideline. Br J Haematol 2009;144:895-903
  • Argyriou AA, Bruna J, Marmiroli P, et al. Chemotherapy-induced peripheral neurotoxicity (CIPN): an update. Crit Rev Oncol Hematol 2012;82:51-77
  • Hershman DL, Lacchetti C, Dworkin RH, et al. Prevention and management of chemotherapy-induced peripheral neuropathy in survivors of adult cancers: American Society of Clinical Oncology clinical practice guideline. J Clin Oncol 2014;32:1941-67
  • Albers JW, Chaudhry V, Cavaletti G, et al. Interventions for preventing neuropathy caused by cisplatin and related compounds. Cochrane Database Syst Rev 2014;3:CD005228
  • Schmidinger M, Budinsky AC, Wenzel C, et al. Glutathione in the prevention of cisplatin induced toxicities. A prospectively randomized pilot trial in patients with head and neck cancer and non small cell lung cancer. Wien Klin Wochenschr 2000;112:617-23
  • Hilpert F, Stähle A, Tomé O, et al. Neuroprotection with amifostine in the first-line treatment of advanced ovarian cancer with carboplatin/paclitaxel-based chemotherapy – a double-blind, placebo-controlled, randomized phase II study from the Arbeitsgemeinschaft Gynäkologische Onkologoie (AGO) Ovarian Cancer Study Group. Support Care Cancer 2005;13:797-805
  • Lin PC, Lee MY, Wang WS, et al. N-acetylcysteine has neuroprotective effects against oxaliplatin-based adjuvant chemotherapy in colon cancer patients: preliminary data. Support Care Cancer 2006;14:484-7
  • Cassidy J, Paul J, Soukop M, et al. Clinical trials of nimodipine as a potential neuroprotector in ovarian cancer patients treated with cisplatin. Cancer Chemother Pharmacol 1998;41:161-6
  • Amara S. Oral glutamine for the prevention of chemotherapy-induced peripheral neuropathy. Ann Pharmacother 2008;42:1481-5
  • Hershman DL, Unge JM, Crew KD, et al. Randomized double-blind placebo-controlled trial of acetyl-L-carnitine for the prevention of taxane-induced neuropathy in women undergoing adjuvant breast cancer therapy. J Clin Oncol 2013;31:2627-33
  • Kottschade LA, Sloan JA, Mazurczak MA, et al. The use of vitamin E for the prevention of chemotherapy-induced peripheral neuropathy: results of a randomized phase III clinical trial. Support Care Cancer 2011;19:1769-77
  • Ghoreishi Z, Esfahani A, Djazayeri A, et al. Omega-3 fatty acids are protective against paclitaxel-induced peripheral neuropathy: a randomized double-blind placebo controlled trial. BMC Cancer 2012;1:355
  • Arrieta Ó, Hernández-Pedro N, Fernández-González-Aragón MC. Retinoic acid reduces chemotherapy-induced neuropathy in an animal model and patients with lung cancer. Neurology 2011;77:987-95
  • Loprinzi CL, Qin R, Dakhil SR, et al. Phase III randomized, placebo-controlled, double-blind study of intravenous calcium and magnesium to prevent oxaliplatin-induced sensory neurotoxicity (N08CB/Alliance). J Clin Oncol 2014;32:997-1005
  • Al Moundhri MS, Al-Salam S, Al Mahrouqee A, et al. The effect of curcumin on oxaliplatin and cisplatin neurotoxicity in rats. J Med Toxicol 2013;9:25-33
  • Cheng XL, Liu HQ, Wang Q, et al. Chemotherapy-induced peripheral neurotoxicity and complementary and alternative medicines: progress and perspective. Front Pharmacol 2015;6:234
  • Park HJ, Lee HG, Kim YS, et al. Ginkgo biloba extract attenuates hyperalgesia in a rat model of vincristine-induced peripheral neuropathy. Anesth Analg 2012;115:1228-33
  • Lee JS, Kim YT, Jeon EK, et al. Effect of green tea extracts on oxaliplatin-induced peripheral neuropathy in rats. BMC Complement Altern Med 2012;12:124
  • Kaur G, Jaggi AS, Singh N. Exploring the potential effect of Ocimum sanctum in vincristine-induced neuropathic pain in rats. J Brachial Plex Peripher Nerve Inj 2010;5:3
  • Muthuraman A, Singh N, Jaggi AS. Protective effect of Acorus calamus L. in rat model of vincristine induced painful neuropathy: an evidence of anti-inflammatory and anti-oxidative activity. Food Chem Toxicol 2011;49:2557-63
  • Abad ANA, Nouri MHK, Tavakkolia F. Effect of Salvia officinalis hydroalcoholic extract on vincristine-induced neuropathy in mice. Chin J Natural Med 2011;9:354-8
  • Oki E, Emi Y, Kojima H, et al. Preventive effect of Goshajinkigan on peripheral neurotoxicity of FOLFOX therapy (GENIUS trial): a placebo-controlled, double-blind, randomized phase III study. Int J Clin Oncol 2015;20:767-75
  • Schröder S, Beckmann K, Franconi G, et al. Can medical herbs stimulate regeneration or neuroprotection and treat neuropathic pain in chemotherapy-induced peripheral neuropathy? Evid Based Complement Alternat Med 2013;2013:423713
  • Shinde SS, Seisler D, Soori G, et al. Can pregabalin prevent paclitaxel-associated neuropathy? An ACCRU pilot trial. Support Care Cancer 2016;24:547-53
  • Durand JP, Deplanque G, Montheil V, et al. Efficacy of venlafaxine for the prevention and relief of oxaliplatin-induced acute neurotoxicity: results of EFFOX, a randomized, double-blind, placebo-controlled phase III trial. Ann Oncol 2012;23:200-5
  • Argyriou AA, Chroni E, Polychronopoulos P, et al. Efficacy of oxcarbazepine for prophylaxis against cumulative oxaliplatin-induced neuropathy. Neurology 2006;67:2253-5
  • Tournigand C, Cervantes A, Figer A, et al. OPTIMOX1: a randomized study of FOLFOX4 or FOLFOX7 with oxaliplatin in a stop-and-Go fashion in advanced colorectal cancer – a GERCOR study. J Clin Oncol 2006;24:394-400
  • Brzeziński K. Chemotherapy-induced polyneuropathy. Part I. Pathophysiology. Contemp Oncol (Pozn) 2012;16:72-8
  • Weickhardt A, Wells K, Messersmith W. Oxaliplatin-induced neuropathy in colorectal cancer. J Oncol 2011;2011:201593
  • Ta LE, Espeset L, Podratz J, et al. Neurotoxicity of oxaliplatin and cisplatin for dorsal root ganglion neurons correlates with platinum-DNA binding. Neurotoxicology 2006;27:992-1002
  • Swain SM, Arezzo JC. Neuropathy associated with microtubule inhibitors: diagnosis, incidence, and management. Clin Adv Hematol Oncol 2008;6:455-67
  • Cavaletti G, Gilardini A, Canta A, et al. Bortezomib-induced peripheral neurotoxicity: a neurophysiological and pathological study in the rat. Exp Neurol 2007;204:317-25
  • Cata JP, Weng HR, Burton AW, et al. Quantitative sensory findings in patients with bortezomib-induced pain. J Pain 2007;8:296-306
  • Schneider BP, Li L, Radovich M, et al. Genome-wide association studies for taxane-induced peripheral neuropathy in ECOG-5103 and ECOG-1199. Clin Cancer Res 2015;21:5082-91
  • Reyes-Gibby CC, Wang J, Yeung S-CJ, et al. Informative gene network for chemotherapy-induced peripheral neuropathy. BioData Mining 2015;8:24
  • Kelley MR, Wikel JH, Guo C, et al. Identification and Characterization of New Chemical Entities Targeting Apurinic/Apyrimidinic Endonuclease 1 for the Prevention of Chemotherapy-Induced Peripheral Neuropathy. J Pharmacol Exp Ther 2016;359:300-309
  • Fishel ML, Jiang Y, Rajeshkumar NV, et al. Impact of APE1/Ref-1 redox inhibition on pancreatic tumor growth. Mol Cancer Ther 2011;10:1698-708
  • Donvito G, Wilkerson JL, Damaj MI, et al. Palmitoylethanolamide Reverses Paclitaxel-Induced Allodynia in Mice. J Pharmacol Exp Ther 2016;359:310-318
  • Zhu J, Carozzi VA, Reed N, et al. Ethoxyquin provides neuroprotection against cisplatin-induced neurotoxicity. Sci Rep 2016;6:28861

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.