Skip to main content
F1000Research logoLink to F1000Research
. 2020 Mar 11;9:F1000 Faculty Rev-177. [Version 1] doi: 10.12688/f1000research.21625.1

Recent advances in understanding chemotherapy-induced peripheral neuropathy

Richard Gordon-Williams 1,a, Paul Farquhar-Smith 1
PMCID: PMC7076330  PMID: 32201575

Abstract

Chemotherapy-induced peripheral neuropathy (CIPN) is a common cause of pain and poor quality of life for those undergoing treatment for cancer and those surviving cancer. Many advances have been made in the pre-clinical science; despite this, these findings have not been translated into novel preventative measures and treatments for CIPN. This review aims to give an update on the pre-clinical science, preventative measures, assessment and treatment of CIPN.

Keywords: Chemotherapy, Neuropathy, Pain, Survivorship

Introduction

The last decade has heralded improvements in cancer survival 1. However, persistent effects following the treatment of cancer can lead to pain and an impaired quality of life long after treatment has finished or cancer has been cured 2. Chemotherapy-induced peripheral neuropathy (CIPN) is one of those effects that can lead to a continuing symptom burden after treatment 3.

CIPN is characterised by the classic “glove and stocking” distribution of symptoms. After chemotherapy, 68% of patients have painful neuropathy at 6 months, improving to 33% at 1 year 4. Although different chemotherapies have variable characteristics, symptoms tend to be predominantly sensory. Sensory toxicity is the predominant feature as dorsal root ganglion (DRG), containing the sensory cell bodies, have a fenestrated endothelium that is more permeable than that found in the spinal cord, where the motor cell bodies lie. Sensory features are characterised by so-called “positive” and “negative” symptoms. “Negative” symptoms include numbness, loss of vibration sense, proprioception and deep tendon reflexes, whereas paraesthesia, dysaesthesia, and cold and mechanical hypersensitivity are referred to as “positive” symptoms.

The development of pain is also a common reason for dose reduction 4, 5, which may have implications for oncological outcome 6. The situation is further complicated by the effect of “coasting”, whereby the development of pain is delayed until after stopping the chemotherapy.

Despite advances in cancer treatment and survival, we still have much to learn about CIPN. It is important to recognise that CIPN is a heterogeneous population; it may be acute, such as the neuropathy commonly experienced with oxaliplatin, or chronic, lasting well beyond the end of treatment. Although there may be some overlap in features, it is likely that the underlying pathophysiology, clinical features and therefore its management differ substantially. Furthermore, not all CIPN is considered painful. This review will focus on the mechanisms but also deliberate on clinical features and treatment of chronic painful CIPN.

Animal models

Animal models of CIPN have increased understanding of the pathophysiology of CIPN, yet a recent meta-analysis highlights problems with the current models and may help deliver more robust and valid models 7. For example, how do studies assessing short-term pain behaviours in animals without tumour burden model chronic CIPN? Pre-clinical studies often focus on the gain-of-function symptoms rather than the loss of function (for example, numbness) more common with chronic CIPN. Misrepresentation of the sexes is evident; 83% of animals used were male. Newer models have addressed some of these criticisms. Griffiths et al. describe a paclitaxel model of CIPN for 28 days with ethologically relevant behavioural tests that better mirror the clinical picture 8. Non-human primate models may be more similar to the human condition 9 but, owing to ethical and pragmatic issues, are not a feasible alternative to rodents.

Mechanisms

The main classes of chemotherapeutics that cause neuropathy include the platinum-based anti-cancer therapies (oxaliplatin and cisplatin), vinca alkaloids (vincristine and vinblastine), taxanes (paclitaxel and docetaxel), proteasome inhibitors (bortezomib) and immunomodulatory drugs (thalidomide). These classes have differing anti-neoplastic mechanisms and likely different mechanisms for neuropathy. Evidence suggests that a number of mechanisms are shared between classes of chemotherapeutics, and most studies investigate the taxanes and the platinums. Currently, these mechanisms can be broadly separated into mitochondrial dysfunction and oxidative stress, microtubule disruption, neuroinflammation and immunological processes, and ion channel dysregulation.

Mitochondrial and oxidative stress

Bioenergetic pathways, predominantly via the oxidation of glucose through the Krebs cycle within the mitochondria, are responsible for the generation of ATP. Chemotherapeutics commonly target nucleolar DNA and may also affect mitochondrial DNA. Indeed, targeting mitochondrial DNA as a principal therapy is an area of ongoing research 10. Whereas nucleolar DNA has well-established repair mechanisms, mitochondria do not. Flatters and Bennett showed that paclitaxel treatment in rats led to swollen vacuolated mitochondria that followed the course of pain-like behaviours for almost 3 months 11. Mitochondrial dysfunction within sensory neurones has also been demonstrated by other chemotherapeutics 1215. Krukowski et al. found that cisplatin-induced mechanical allodynia is associated with mitochondrial damage in DRG but that the loss of intra-epidermal nerve fibres (IENFs), seen in patients with CIPN, is related to bioenergetic deficits in peripheral nerves 16. Gregg et al. found that post-mortem platinum concentrations in patients who received platinum chemotherapy were highest in DRGs and demonstrated a linear relationship between DRG levels and cumulative dose 17, and levels were higher in patients with neuropathy. Animal data suggest a dose-dependent accumulation within the mitochondria of DRG neurones 18. Recently, gene expression analysis further supported mitochondrial dysfunction in patients who develop CIPN. Kober et al. 19 found that breast cancer patients who develop neuropathy after paclitaxel demonstrate differential expression in a number of pathways implicated in mitochondrial dysfunction, including oxidative stress 20. Additionally, genetic polymorphisms in anti-oxidant pathways have been associated with an increased incidence of CIPN 21.

Numerous animal studies indicate that chemotherapy worsens oxidative stress 22, 23. Furthermore, anti-oxidants prevent the development of mitochondrial dysfunction, IENF loss and pain-like behaviours in animal models 24, 25.

The anti-oxidant alpha-lipoic acid reduces neuropathy in patients with diabetes and also animal models of CIPN 26. Concurrent administration of alpha-lipoic acid reduces neuropathic symptoms secondary to bortezomib with less alteration to chemotherapy regimen secondary to adverse events 27. However, despite the neuroprotective effects of anti-oxidants in vitro studies 28, there is little clinical evidence for other nutraceutical anti-oxidants in the prevention of CIPN 29. Recently, however, a phase I trial showed that calmangafodipir, a manganese superoxide dismutase mimic that aids reactive oxygen species (ROS) degradation, reduces acute and chronic CIPN after oxaliplatin in patients 30 without affecting response to chemotherapy and life expectancy. Metformin can also reduce neuropathic behaviours via a reduction in oxidative stress 13, 31, 32. Metformin treatment in 40 patients receiving oxaliplatin reduced National Cancer Institute–Common Terminology Criteria for Adverse Events (NCI-CTCAE) grade 2 and 3 neuropathy with a moderate reduction in neurotoxicity score and a modest reduction in pain 33.

Mitochondria play a key role not only in ROS regulation but in numerous other cellular processes, including calcium buffering, apoptosis and energy production via oxidative phosphorylation. Duggett et al. have shown that whereas basal respiration and ATP turnover were unaffected in DRG mitochondria of paclitaxel treated rats, maximal respiration and spare reserve capacity were greatly reduced at peak pain behaviour 12. This indicates a reduced ability of these neurones to respond to stress, and the authors postulated that a switch to glycolysis could be an adaptive mechanism to reduce harmful ROS production.

Schwann cells play a crucial role in the regrowth of peripheral axons after injury; however, Nishida et al. found that accumulation of platinum compounds within Schwann cells was much lower than that in peripheral nerves and DRG 18. Conversely, Imai et al. suggested that in vitro platinum compounds cause mitochondrial dysfunction in Schwann cells at drug concentrations lower than those required to induce neurotoxicity 34, suggesting a greater role for mitochondrial dysfunction in Schwann cells in CIPN.

In animal models, treatment with pifithrin-μ, a molecule that suppresses mitochondrial damage, improves mitochondrial morphology, bioenergetics and IENF density while reducing pain behaviours 14, 35. Combined with evidence that it may act synergistically with the anti-cancer mechanisms of chemotherapeutics 35, 36, pifithrin-μ represents an exciting prospect in cancer care.

Glia and neuroinflammation

Glia are key in maintaining homeostasis and immunity in the central nervous system in both health and disease. In models of non-chemotherapy-induced neuropathy, microglia have been found to play an integral role in the development of the pain state 37, 38. Oxaliplatin-treated rats displayed persistent mechanical allodynia, sensory deficits and decreased density of IENFs 39. Hu et al. showed a persistent activation of spinal cord microglia through strengthening of triggering receptor expressed on myeloid cells 2 (TREM2) signalling and demonstrated that either inhibiting microglia with minocycline or interrupting TREM2 signalling improved pain-like behaviours and IENF density 40. Furthermore, an agonist at the CB2 cannabinoid receptor, colocalised with spinal microglia, inhibited microgliosis and pain behaviours in an animal model of paclitaxel-induced neuropathy 41.

Despite these findings, astrogliosis rather than microgliosis is thought to be of greater importance to the development of CIPN 42, 43, while in some models, astrocyte inhibition with minocycline prevented the development of pain-like behaviours. But how would astrocyte activation lead to the development of CIPN? One proposed mechanism in a rat model of oxaliplatin-induced painful neuropathy is dysregulation of spinal adenosine kinase expression in astrocytes 44. This may lead to activation of NRLP3/interleukin 1 beta (NRLP3/IL1β) pathway, promoting dorsal horn neuronal excitability with concurrent suppression of the anti-inflammatory IL-10 system, leading to central sensitisation and pain behaviours 44, 45. Importantly, restoration of adenosine signalling with an A3AR adenosine receptor agonist prevents the development of both astrocytosis and pain behaviours 45. Another mechanism proposed in rodent models is through the alteration of sphingolipid signalling within astrocytes in the superficial layers of the dorsal horn of the spinal cord, an area concerned with nociceptive transmission 46, 47. Maladapted sphingolipid metabolism, through direct bortezomib effects and increased IL-1β, may increase glutamatergic transmission and consequently nociceptive transmission and pain behaviours 47.

Reasons for the discrepancies in the role glia play in CIPN remain unclear but the discrepancies may be due to variations in chemotherapy, species, time point and sex studied. Pain phenotype differs greatly between male and female patients, and the pathophysiology in animal models is also sex-dependent 48. In animal models of bortezomib-induced peripheral neuropathy, modulation of sphingolipid signalling attenuates pain behaviours in male but not female rodents 47. Additional examples of sexual dimorphism are found in paclitaxel-induced peripheral neuropathy, and Toll-like receptor 9 (TLR9) expression in macrophages infiltrating DRG plays a role in the development of pathophysiological changes and behaviours in male mice but not females 49. Macrophage infiltration into DRG and peripheral nerves has been seen in a number of animal models of CIPN, and as with other models of neuropathic pain, activation of TLR4 seems to be crucial 5052.

Clinically, minocycline treatment reduced only the acute pain syndrome associated with paclitaxel infusion but not the development of chronic CIPN 53. Additionally, in another phase 2 trial, minocycline failed to prevent oxaliplatin-induced peripheral neuropathy 54. Despite previous pre-clinical trials indicating minocycline’s efficacy at inhibiting astrocyte activation and pain behaviours, its actions have been ascribed predominantly to inhibition of microglia and not astrocytes 55, 56. Given the differential role that microglia may have in CIPN, minocycline’s lack of clinical efficacy may be of no surprise and neuroinflammation still represents a worthy area for continued research in the prevention of CIPN. Fingolimod, a drug used in the treatment of multiple sclerosis, downregulates the S1PR1 receptor found on astrocytes. Antagonism of this receptor has been shown to reverse immunochemical and behavioural changes in rodent models 47. This presents the exciting prospect of a potentially new mechanistic target with a readily available therapeutic agent; however, additional trials are required to assess both its effects on CIPN and importantly tumour activity.

Ion channels

Pre-clinical studies have highlighted many chemotherapy-induced changes in ion channel expression, possibly driving behavioural changes in other neuropathic pain states 57.

Changes in sodium channel expression and their sensitisation increase spontaneous neuronal firing and decrease activation threshold 58, mechanisms possibly analogous to the allodynia, hyperalgesia and paroxysmal sensations of CIPN. In patients, sodium channel dysfunction is found in acute oxaliplatin toxicity 59, and sodium channel polymorphisms may have a causal role in the development of acute and possibly chronic CIPN 60. Furthermore, Na v1.7 channel has been found to be similarly upregulated in nociceptive neurones in both a rat model and patients with chronic paclitaxel-induced peripheral neuropathy 61. Although dysregulation of other sodium channels is seen in pre-clinical studies of CIPN 62, the clinical efficacy of sodium channel blockers has been disappointing 63.

Potassium channel dysregulation is present in animal models of CIPN 64. Acutely, oxaliplatin leads to the down-regulation of potassium channels in animal models 62, and Poupon et al. found that treatment with a riluzole (a potassium channel activator) prevents the development of persistent CIPN in mice 65. A phase 2 randomised controlled trial (RCT) investigating the efficacy of riluzole in the prevention of CIPN is under way 66. Transient receptor potential (TRP) channels are critical in temperature transduction. Oxaliplatin treatment leads to an increased expression of TRPA1, TRPV1 and TRPM8 in sensory neurones 67. Interestingly, suppression of TREK-1 and TRAAK potassium channels (and an increase in pro-excitatory Na v1.8 and HCN ion channels) is found on neurones expressing TRPM8, a receptor responsive to cold 62. This may present a mechanism through which menthol provides symptomatic relief and oxaliplatin produces cold hypersensitivity acutely.

Although calcium channel modulation has shown promise in animal models of CIPN 68, 69, no direct calcium channel blockers are in clinical use for neuropathic pain. Cisplatin causes an increase in the calcium channel alpha-2-delta subunit, the target of gabapentinoids 70, and both topical and systemic treatment with gabapentinoids have been found to be beneficial in rat models of CIPN 71, 72. Despite this, treatment with pregabalin for 3 days before and after each cycle of oxaliplatin failed to prevent CIPN in patients 73.

Clinical features

Risk factors

There are many potential predictors in the development of CIPN, including patient-related factors, such as increased age, pre-existing neuropathy, smoking status, and impaired renal function, and chemotherapy-related factors, such as type of chemotherapy, cumulative chemotherapy dose, concurrent chemotherapy treatment, and duration of infusion 7476. Certain cancers may cause a subclinical neuropathy which may predispose patients to CIPN and worsen outcomes 77.

Genetic markers have been implicated in chemotherapy-related toxicity, and a number of genome-wide association studies have looked at polymorphisms associated with CIPN. A number of polymorphisms have been identified, none of which (at present) has sufficient prognostic value to be of use in the clinical context 78. Argyriou et al. 78 called for improved methodology and more standardised diagnostic and severity grading to better inform future studies.

Assessment of CIPN

Despite challenges in prevention and treatment, assessment for CIPN should occur before, during and after chemotherapy. Assessment should include (1) diagnosis (including possible differential diagnoses), (2) severity (including functional impairment) and (3) time course of symptoms and relationship to chemotherapy.

Diagnosis of CIPN requires a full history and examination ( Table 1). Within the history, it is important to determine pre-existing risk factors for neuropathy such as diabetes, vitamin deficiency, alcohol use and previous chemotherapy. Blood tests, including a full blood count, comprehensive metabolic profile, measurement of erythrocyte sedimentation rate, fasting blood glucose, vitamin B 12, and thyroid-stimulating hormone levels, should be considered to help rule out other causative or contributory causes for neuropathy.

Table 1. Key elements in history and examination.

History Examination
     •    Details of chemotherapy regimen

     •    Number of cycles, dose and cumulative dose

     •    Onset of symptoms in relation to chemotherapy

     •    “Coasting” assessment (neuropathy occurring or
worsening after chemotherapy cessation)

     •    Evidence of change over time (better or worse)

Symptoms

     •    Distribution (hands, feet or more proximal)

     •    Numbness, paraesthesia, pain, spontaneous or evoked

     •    Motor or sympathetic dysfunction

     •    Functionality and interference on activities
Sensation

     •    Light touch

     •    Pinprick or painful stimulus

     •    Vibration sense

     •    Cold/hot sensation

Other

     •    Deep tendon reflexes

     •    Motor power

     •    Balance

Painful CIPN is a subset that may benefit from further characterisation with multidimensional pain assessment tools. The McGill Pain Questionnaire (MPQ) and the Brief Pain Inventory (BPI) have been validated for use in cancer pain 79 and although they both assess sensory aspects, including severity, the BPI also assesses impact on function. Likewise, screening tools may aid in the assessment of neuropathic pain. Two such tools, the Leeds Assessment of Neuropathic Symptoms and Signs (LANSS) and Douleur Neuropathique 4 (DN4) 80, 81, have good sensitivity and specificity in cancer pain but are not validated in CIPN. Furthermore, although chemotherapy is a common treatment for childhood cancers and the subsequent neuropathy may differ in phenotype from that of adults 82, 83, there are few validated tools for the assessment of CIPN in children. One such score is the paediatric-modified total neuropathy score (ped-mTNS), which has been validated in a small group of children undergoing vincristine or cisplatin chemotherapy for leukaemia 84; however, the 2008 Pediatric Initiative on Methods, Measurement, and Pain Assessment in Clinical Trials (PedIMMPACT) called for the development of reliable and valid tools for use in children 85.

Numerous tools have been developed for the assessment of CIPN; however, there is notable inter-observer variation between these scales. Clinical rated scales such as the Ajani scale, World Health Organization, Eastern Cooperative Oncology Group neuropathy scale, and NCI-Common Toxicity Criteria (NCI-CTC) have limited assessment of pain 86 and may not truly reflect the incidence of adverse neuropathy, leading to inappropriate treatment reduction or cessation. Furthermore, clinician-rated neuropathy scales underestimate the severity of CIPN when compared with patient-reported measures 87. In a recent systematic review, Haryani et al. suggested that, owing to their psychometric properties and practicality, the Functional Assessment of Cancer Therapy/Gynecologic Oncology Group-Neurotoxicity (Fact/GOG-Ntx) and total neuropathy score (TNS) (see below) were the most appropriate assessment tools available 88. Yet other studies are contradictory; this is in line with a recent DELPHI survey which showed there is little consensus amongst clinicians 89.

Fact/GOG-Ntx was developed for assessing the impact of neuropathy on quality of life after chemotherapy for gynaecological cancer and consists of questions on physical, social, emotional and functional wellbeing with an additional 11-question neurotoxicity subscale. This subscale has been used independently and demonstrates good sensitivity in diagnosis and responsiveness to treatment 90 and has been validated in other non-gynaecological, non-cisplatin/paclitaxel patients. Further shortening the neurotoxic subscale to four sensory questions maintains the validity and sensitivity while reducing the burden of patient questions 91.

The Patient Neurotoxicity Questionnaire (PNQ) evaluates sensory, motor and functional components of neuropathy with good sensitivity to change over time and showed improved reporting of CIPN when compared with clinician reporting tools such as the NCI-CTC (see above). Importantly, the PNQ assesses the impact of neuropathy on 22 activities (such as fastening buttons or typing) that are not assessed with other tools, thus representing a more holistic patient-centred assessment of neuropathy.

The TNS is an eight-item score of patient report of neuropathic symptoms, examination findings to pinprick, vibration and deep tendon reflexes and nerve conduction studies (NCSs). A shortened version without the electrophysiological factors has been validated: the TNSc (clinical version of the TNS) 92; both have been shown to be more sensitive to CIPN changes than NCI-CTC and comparable in changes to quality-of-life measures 93. Importantly, TNS delivers both clinician- and patient-rated components. Quality-of-life measures are commonly not assessed in many CIPN tools. The European Organization of Research and Treatment of Cancer (EORTC), 20-item quality-of-life questionnaire, is sensitive to changes in quality of life secondary to CIPN 94.

Investigations

There has been a great deal of interest in phenotyping CIPN by using minimally invasive tools such as NCSs, quantitative sensory testing (QST) and IENF density. It seems sensible that underlying mechanisms may translate to differing patterns of neuronal loss and therefore differences in functional deficits, yet in practice this theory is not robust. Traditionally, CIPN has been characterised as a predominant sensory neuropathy effecting large myelinated fibre function, and nerve biopsies from patients with cisplatin- and paclitaxel-induced neuropathy show a loss of large fibres with axonal atrophy and secondary demyelination 95, 96.

Platinum chemotherapeutics cause neuronal cycle arrest within the DRG and therefore likely cause a neuronopathy (also referred to as ganglionopathy) and anterograde neuronal degeneration. On NCS, this would manifest as non-length-dependent neuropathy affecting both the proximal and distal neurone. In contrast, chemotherapeutics interfering with mitochondrial or microtubule function impair axonal transportation giving a length-dependent axonal polyneuropathy, leading to a die back of intraepidermal nerve fibres. However, owing to poor correlation with clinical symptoms, NCSs cannot be routinely recommended. Furthermore, NCSs assess predominantly large-fibre function, missing small-fibre changes that may occur with painful CIPN.

Owing to its ability to assess large- and small-fibre types, QST may be of use in the phenotyping of neuropathic pain 97 and therefore has been proposed as a useful tool in CIPN 98. In patients with paclitaxel-induced peripheral neuropathy, the reduction in light touch and vibration detection thresholds seen in hands and feet supports the mechanism of paclitaxel causing a distal neuropathy predominantly effecting the large, non-nociceptive neurones 99. Additionally, some report that thermal detection thresholds and pinprick detection are minimally affected, indicating that small-fibre function is preserved in this group. This is in contrast to findings in patients with vincristine and bortezomib-induced neuropathy, with some studies reporting changes in pinprick perception and warm detection thresholds suggesting small, nociceptive fibre dysfunction in this group of patients 100, 101.

Pre-existing QST sensory deficits increase the risk of developing CIPN 102; in some cases, cancer itself may be responsible for QST changes 103. Although the QST sensory profile may differ between agents 104, 105, QST profiles for painful and painless CIPN may be similar 106 and changes in QST may occur later than symptoms develop 107. Furthermore, QST requires expertise and time and consequently is not commonly used in routine clinical practice for the evaluation of CIPN.

Skin punch biopsy can inform the diagnosis of small-fibre neuropathies. In CIPN, similar to other small-fibre neuropathies, IENF loss is observed 108. Taking comparative distal thigh and distal leg punch biopsies can help differentiate between a length-dependent neuropathy or a neuronopathy; however, evaluating CIPN using IENF densities has been found to be unreliable; there is a large overlap between different chemotherapeutics, and results conflict with other assessment tools 109111. Furthermore, although punch biopsy can be repeated, it is time-consuming and invasive and IENF density has been found to be a poor correlate of pain 112.

Other techniques for assessing neuropathy have yet to be fully validated. Nevertheless, simple bedside measures such as vibration sense, light touch and pinprick have good validity in the measurement of neuropathy 113.

Prevention

Reducing regional perfusion (cryotherapy) may reduce CIPN; cooling gloves and stockings have been shown to reduce the risk of desquamation and nail changes associated with chemotherapy. Of the three published trials, only one showed benefit 114. Owing to poorly tolerated treatment or a greater-than-expected control group response, the other studies were negative 115117.

In 2014, the American Society of Clinical Oncology evaluated 42 studies while developing guidelines on the prevention of CIPN 63. Owing to a lack of high-quality data, they were unable to make any recommendations and encouraged additional research.

Treatment

Pharmacological

RCT evidence of treatments in CIPN suggested that duloxetine is the only anti-neuropathic agent with evidence of benefit 118. Many CIPN RCTs fail to meet the IMMPACT guidelines for outcome measures in clinical trials 119, 120. Nevertheless, a recent comparative study showed that venlafaxine and duloxetine reduced pain in established CIPN 121. Careful phenotyping may help, as demonstrated with the improved efficacy of oxcarbazepine in the “irritable nociceptor” subgroup 122. Phenotyping patients for biological and psychosocial characteristics may give additional insight 122125.

Topical treatments are an attractive option for the management of CIPN. A small non-randomised study of topical menthol in 52 patients showed improved BPI scores 126, and combination therapy with baclofen, amitriptyline and ketamine showed an improvement on some of the EORTC QLQ-CIPN-20 measures 127. Topical 8% capsaicin patch application following CIPN has been shown to improve continuous pain, neuropathic pain symptoms, and patient global impression of change 128. This treatment has also been found to improve IENF density, suggesting underlying disease modification 128.

There is increasing enthusiasm for the use of cannabinoids in the treatment of many chronic pain states. Agonism at CB1 and CB2 receptors has shown analgesia in rodent models of CIPN 129133 but these findings have not translated into evidence of clinical efficacy. One published pilot study of nabiximols (THC:CBD mix) in 15 patients with CIPN 134 showed no significant improvement in pain, but a 2-point decrease over placebo was seen in five patients classified as “responders” 134.

Without specific evidence for CIPN, clinicians extrapolate treatments from other neuropathic pain states 135. Interestingly, strong opioids have some of the best “numbers needed to treat” (NNTs) for neuropathic pain (NNT 4.3, 95% confidence interval 3.4–5.8) 135. Some clinicians may advocate the use of opioids in CIPN, however with increasing survivorship amongst patients with cancer, the possible benefits of opioids should be continually weighed up against the risk of long-term opioid therapy 2.

Non-pharmacological

Neuromodulation has shown promise in various neuropathic pain states 136. A number of case reports indicate that neuromodulation may help refractory CIPN 137, 138, but RCT data are lacking.

A recent study found that the use of wireless transcutaneous electrical nerve stimulation significantly improved some measures of CIPN, including pain, numbness and tingling 139. Furthermore, scrambler therapy (a novel transcutaneous neurostimulation technique) has been postulated as a potential treatment 140 but was no more effective than sham therapy in a recent RCT 141.

Acupuncture

A Cochrane Review of the efficacy of acupuncture in the treatment of cancer pain showed insufficient evidence of its efficacy 142. Since then, a number of trials of acupuncture in CIPN have demonstrated improvements in several domains 143145. A systematic review concluded that there was insufficient evidence to recommend acupuncture for the treatment of CIPN 146, although low risk of harm and possible benefit may allow its pragmatic use in painful CIPN.

Physical therapy

Exercise has been shown to improve a number of facets that contribute to morbidity associated with CIPN, including balance and strength 147, 148, numbness, tingling, and hot and cold sensations 149. One study found that, on analysis of the quality-of-life data, exercise had a moderate effect on pain in patients undergoing chemotherapy; however, this was not limited to CIPN 150.

Psychological therapy

Psychological factors have been shown to play a role in both the initiation and maintenance of a number of chronic pain states 151. The activity of duloxetine, via enhancing descending inhibitory pathways, suggests that alteration of mood may play a role. In favour of this viewpoint, a study of 111 patients who received treatment for breast cancer found that pre-existing anxiety and pre-therapy numbness were the only factors to predict CIPN eight months later 152. Knoerl et al. found that an eight-week web-based cognitive behavioural programme led to modest improvements in worst pain with no differences in mean pain 153. It was hypothesised that this would be due to improvements in fatigue, anxiety, sleep-related factors, or depression; however, a follow-up analysis was unable to substantiate these findings 154.

Future directions

Pre-clinical studies have shown that antagonism of the sigma 1 receptor (present on mitochondrial endoplasmic reticulum) is able to reduce mitochondrial structural changes and pain behaviours that occur in CIPN. A phase II clinical trial found that sigma 1 antagonist treatment during FOLFOX chemotherapy diminished cold hypersensitivity, reduced the dropout rate and allowed a higher cumulative dose of oxaliplatin 155. Although the long-term pain outcomes are not known, this highlights a pathway for potential therapeutics that could improve CIPN.

Summary

Despite an ever-expanding body of literature behind the pathophysiology and treatment of CIPN, new treatment options are still limited, and a proportion of patients continue to have difficulty controlling symptoms causing a significant impact on quality of life. Guided by the pre-clinical literature, novel targets that may help prevent CIPN are beginning to emerge. However, with continual advancements in chemotherapeutic agents with novel mechanisms, it is important that ongoing development of treatments for CIPN continue.

Editorial Note on the Review Process

F1000 Faculty Reviews are commissioned from members of the prestigious F1000 Faculty and are edited as a service to readers. In order to make these reviews as comprehensive and accessible as possible, the referees provide input before publication and only the final, revised version is published. The referees who approved the final version are listed with their names and affiliations but without their reports on earlier versions (any comments will already have been addressed in the published version).

The referees who approved this article are:

  • Peter M. Grace, Department of Symptom Research, University of Texas MD Anderson Cancer Center, Houston, TX, USA

  • Irina Vetter, Centre for Pain Research, Institute for Molecular Bioscience, University of Queensland, Saint Lucia, QLD, Australia

Funding Statement

The author(s) declared that no grants were involved in supporting this work.

[version 1; peer review: 2 approved]

References

  • 1. Arnold M, Rutherford MJ, Bardot A, et al. : Progress in cancer survival, mortality, and incidence in seven high-income countries 1995-2014 (ICBP SURVMARK-2): a population-based study. Lancet Oncol. 2019;20(11):1493–505. 10.1016/S1470-2045(19)30456-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Brown M, Farquhar-Smith P: Pain in cancer survivors; filling in the gaps. Br J Anaesth. 2017;119(4):723–36. 10.1093/bja/aex202 [DOI] [PubMed] [Google Scholar]
  • 3. Mols F, Beijers T, Vreugdenhil G, et al. : Chemotherapy-induced peripheral neuropathy and its association with quality of life: a systematic review. Support Care Cancer. 2014;22(8):2261–9. 10.1007/s00520-014-2255-7 [DOI] [PubMed] [Google Scholar]
  • 4. Seretny M, Currie GL, Sena ES, et al. : Incidence, prevalence, and predictors of chemotherapy-induced peripheral neuropathy: A systematic review and meta-analysis. Pain. 2014;155(12):2461–70. 10.1016/j.pain.2014.09.020 [DOI] [PubMed] [Google Scholar]
  • 5. Gutiérrez-Gutiérrez G, Sereno M, Miralles A, et al. : Chemotherapy-induced peripheral neuropathy: clinical features, diagnosis, prevention and treatment strategies. Clin Transl Oncol. 2010;12(2):81–91. 10.1007/S12094-010-0474-z [DOI] [PubMed] [Google Scholar]
  • 6. Bhatnagar B, Gilmore S, Goloubeva O, et al. : Chemotherapy dose reduction due to chemotherapy induced peripheral neuropathy in breast cancer patients receiving chemotherapy in the neoadjuvant or adjuvant settings: a single-center experience. SpringerPlus. 2014;3:366. 10.1186/2193-1801-3-366 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Currie GL, Angel-Scott HN, Colvin L, et al. : Animal models of chemotherapy-induced peripheral neuropathy: A machine-assisted systematic review and meta-analysis. PLoS Biol. 2019;17(5):e3000243. 10.1371/journal.pbio.3000243 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Griffiths LA, Duggett NA, Pitcher AL, et al. : Evoked and Ongoing Pain-Like Behaviours in a Rat Model of Paclitaxel-Induced Peripheral Neuropathy. Pain Res Manag. 2018;2018: 8217613. 10.1155/2018/8217613 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Hama A, Natsume T, Ogawa S, et al. : Gaps in Understanding Mechanism and Lack of Treatments: Potential Use of a Nonhuman Primate Model of Oxaliplatin-Induced Neuropathic Pain. Pain Res Manag. 2018;2018: 1630709. 10.1155/2018/1630709 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Wisnovsky SP, Wilson JJ, Radford RJ, et al. : Targeting mitochondrial DNA with a platinum-based anticancer agent. Chem Biol. 2013;20(11):1323–8. 10.1016/j.chembiol.2013.08.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Flatters SJL, Bennett GJ: Studies of peripheral sensory nerves in paclitaxel-induced painful peripheral neuropathy: evidence for mitochondrial dysfunction. Pain. 2006;122(3):245–57. 10.1016/j.pain.2006.01.037 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Duggett NA, Griffiths LA, Flatters SJL: Paclitaxel-induced painful neuropathy is associated with changes in mitochondrial bioenergetics, glycolysis, and an energy deficit in dorsal root ganglia neurons. Pain. 2017;158(8):1499–508. 10.1097/j.pain.0000000000000939 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Ludman T, Melemedjian OK: Bortezomib and metformin opposingly regulate the expression of hypoxia-inducible factor alpha and the consequent development of chemotherapy-induced painful peripheral neuropathy. Mol Pain. 2019;15: 174480691985004. 10.1177/1744806919850043 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Maj MA, Ma J, Krukowski KN, et al. : Inhibition of Mitochondrial p53 Accumulation by PFT-μ Prevents Cisplatin-Induced Peripheral Neuropathy. Front Mol Neurosci. 2017;10:108. 10.3389/fnmol.2017.00108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Xiao WH, Zheng H, Zheng FY, et al. : Mitochondrial abnormality in sensory, but not motor, axons in paclitaxel-evoked painful peripheral neuropathy in the rat. Neuroscience. 2011;199:461–9. 10.1016/j.neuroscience.2011.10.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Krukowski K, Ma J, Golonzhka O, et al. : HDAC6 inhibition effectively reverses chemotherapy-induced peripheral neuropathy. Pain. 2017;158(6):1126–37. 10.1097/j.pain.0000000000000893 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Gregg RW, Molepo JM, Monpetit VJ, et al. : Cisplatin neurotoxicity: the relationship between dosage, time, and platinum concentration in neurologic tissues, and morphologic evidence of toxicity. J Clin Oncol. 1992;10(5):795–803. 10.1200/JCO.1992.10.5.795 [DOI] [PubMed] [Google Scholar]
  • 18. Nishida K, Takeuchi K, Hosoda A, et al. : Ergothioneine ameliorates oxaliplatin-induced peripheral neuropathy in rats. Life Sci. 2018;207:516–24. 10.1016/j.lfs.2018.07.006 [DOI] [PubMed] [Google Scholar]
  • 19. Kober KM, Olshen A, Conley YP, et al. : Expression of mitochondrial dysfunction-related genes and pathways in paclitaxel-induced peripheral neuropathy in breast cancer survivors. Mol Pain. 2018;14: 174480691881646. 10.1177/1744806918816462 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Ziegler D, Hanefeld M, Ruhnau KJ, et al. : Treatment of symptomatic diabetic peripheral neuropathy with the anti-oxidant alpha-lipoic acid. A 3-week multicentre randomized controlled trial (ALADIN Study). Diabetologia. 1995;38(12):1425–33. 10.1007/bf00400603 [DOI] [PubMed] [Google Scholar]
  • 21. Katayanagi S, Katsumata K, Mori Y, et al. : GSTP1 as a potential predictive factor for adverse events associated with platinum-based antitumor agent-induced peripheral neuropathy. Oncol Lett. 2019;17(3):2897–2904. 10.3892/ol.2019.9907 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Ma J, Kavelaars A, Dougherty PM, et al. : Beyond symptomatic relief for chemotherapy-induced peripheral neuropathy: Targeting the source. Cancer. 2018;124(11):2289–98. 10.1002/cncr.31248 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Shim HS, Bae C, Wang J, et al. : Peripheral and central oxidative stress in chemotherapy-induced neuropathic pain. Mol Pain. 2019;15: 1744806919840098. 10.1177/1744806919840098 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Janes K, Doyle T, Bryant L, et al. : Bioenergetic deficits in peripheral nerve sensory axons during chemotherapy-induced neuropathic pain resulting from peroxynitrite-mediated post-translational nitration of mitochondrial superoxide dismutase. Pain. 2013;154(11):2432–40. 10.1016/j.pain.2013.07.032 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Jin HW, Flatters SJ, Xiao WH, et al. : Prevention of paclitaxel-evoked painful peripheral neuropathy by acetyl-L-carnitine: effects on axonal mitochondria, sensory nerve fiber terminal arbors, and cutaneous Langerhans cells. Exp Neurol. 2008;210(1):229–37. 10.1016/j.expneurol.2007.11.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Papanas N, Ziegler D: Efficacy of α-lipoic acid in diabetic neuropathy. Expert Opin Pharmacother. 2014;15(18):2721–31. 10.1517/14656566.2014.972935 [DOI] [PubMed] [Google Scholar]
  • 27. Maschio M, Zarabla A, Maialetti A, et al. : Prevention of Bortezomib-Related Peripheral Neuropathy With Docosahexaenoic Acid and α-Lipoic Acid in Patients With Multiple Myeloma: Preliminary Data. Integr Cancer Ther. 2018;17(4):1115–24. 10.1177/1534735418803758 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Popović J, Klajn A, Paunesku T, et al. : Neuroprotective Role of Selected Antioxidant Agents in Preventing Cisplatin-Induced Damage of Human Neurons In Vitro. Cell Mol Neurobiol. 2019;39(5):619–36. 10.1007/s10571-019-00667-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Schloss JM, Colosimo M, Airey C, et al. : Nutraceuticals and chemotherapy induced peripheral neuropathy (CIPN): a systematic review. Clin Nutr. 2013;32(6):888–93. 10.1016/j.clnu.2013.04.007 [DOI] [PubMed] [Google Scholar]
  • 30. Glimelius B, Manojlovic N, Pfeiffer P, et al. : Persistent prevention of oxaliplatin-induced peripheral neuropathy using calmangafodipir (PledOx ®): a placebo-controlled randomised phase II study (PLIANT). Acta Oncol. 2018;57(3):393–402. 10.1080/0284186X.2017.1398836 [DOI] [PubMed] [Google Scholar]
  • 31. Chakraborty A, Chowdhury S, Bhattacharyya M: Effect of metformin on oxidative stress, nitrosative stress and inflammatory biomarkers in type 2 diabetes patients. Diabetes Res Clin Pract. 2011;93(1):56–62. 10.1016/j.diabres.2010.11.030 [DOI] [PubMed] [Google Scholar]
  • 32. Hou X, Song J, Li XN, et al. : Metformin reduces intracellular reactive oxygen species levels by upregulating expression of the antioxidant thioredoxin via the AMPK-FOXO3 pathway. Biochem Biophys Res Commun. 2010;396(2):199–205. 10.1016/j.bbrc.2010.04.017 [DOI] [PubMed] [Google Scholar]
  • 33. El-Fatatry BM, Ibrahim OM, Hussien FZ, et al. : Role of metformin in oxaliplatin-induced peripheral neuropathy in patients with stage III colorectal cancer: randomized, controlled study. Int J Colorectal Dis. 2018;33(12):1675–83. 10.1007/s00384-018-3104-9 [DOI] [PubMed] [Google Scholar]
  • 34. Imai S, Koyanagi M, Azimi Z, et al. : Taxanes and platinum derivatives impair Schwann cells via distinct mechanisms. Sci Rep. 2017;7(1):5947. 10.1038/s41598-017-05784-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Krukowski K, Nijboer CH, Huo X, et al. : Prevention of chemotherapy-induced peripheral neuropathy by the small-molecule inhibitor pifithrin-μ. Pain. 2015;156(11):2184–92. 10.1097/j.pain.0000000000000290 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. McKeon A, Egan A, Chandanshive J, et al. : Novel Improved Synthesis of HSP70 Inhibitor, Pifithrin-μ. In Vitro Synergy Quantification of Pifithrin-μ Combined with Pt Drugs in Prostate and Colorectal Cancer Cells. Molecules. 2016;21(7): pii: E949. 10.3390/molecules21070949 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Zhao H, Alam A, Chen Q, et al. : The role of microglia in the pathobiology of neuropathic pain development: what do we know? Br J Anaesth. 2017;118(4):504–16. 10.1093/bja/aex006 [DOI] [PubMed] [Google Scholar]
  • 38. Sayo A, Konishi H, Kobayashi M, et al. : GPR34 in spinal microglia exacerbates neuropathic pain in mice. J Neuroinflammation. 2019;16(1):82. 10.1186/s12974-019-1458-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Di Cesare Mannelli L, Pacini A, Micheli L, et al. : Glial role in oxaliplatin-induced neuropathic pain. Exp Neurol. 2014;261:22–33. 10.1016/j.expneurol.2014.06.016 [DOI] [PubMed] [Google Scholar]
  • 40. Hu LY, Zhou Y, Cui WQ, et al. : Triggering receptor expressed on myeloid cells 2 (TREM2) dependent microglial activation promotes cisplatin-induced peripheral neuropathy in mice. Brain Behav Immun. 2018;68:132–45. 10.1016/j.bbi.2017.10.011 [DOI] [PubMed] [Google Scholar]
  • 41. Wu J, Hocevar M, Bie B, et al. : Cannabinoid Type 2 Receptor System Modulates Paclitaxel-Induced Microglial Dysregulation and Central Sensitization in Rats. J Pain. 2019;20(5):501–14. 10.1016/j.jpain.2018.10.007 [DOI] [PubMed] [Google Scholar]
  • 42. Zhang H, Yoon SY, Zhang H, et al. : Evidence that spinal astrocytes but not microglia contribute to the pathogenesis of Paclitaxel-induced painful neuropathy. J Pain. 2012;13(3):293–303. 10.1016/j.jpain.2011.12.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Robinson CR, Zhang H, Dougherty PM: Astrocytes, but not microglia, are activated in oxaliplatin and bortezomib-induced peripheral neuropathy in the rat. Neuroscience. 2014;274:308–17. 10.1016/j.neuroscience.2014.05.051 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Janes K, Wahlman C, Little JW, et al. : Spinal neuroimmune activation is independent of T-cell infiltration and attenuated by A3 adenosine receptor agonists in a model of oxaliplatin-induced peripheral neuropathy. Brain Behav Immun. 2015;44:91–9. 10.1016/j.bbi.2014.08.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Wahlman C, Doyle TM, Little JW, et al. : Chemotherapy-induced pain is promoted by enhanced spinal adenosine kinase levels through astrocyte-dependent mechanisms. Pain. 2018;159(6):1025–34. 10.1097/j.pain.0000000000001177 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Janes K, Little JW, Li C, et al. : The development and maintenance of paclitaxel-induced neuropathic pain require activation of the sphingosine 1-phosphate receptor subtype 1. J Biol Chem. 2014;289(30):21082–97. 10.1074/jbc.M114.569574 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Stockstill K, Doyle TM, Yan X, et al. : Dysregulation of sphingolipid metabolism contributes to bortezomib-induced neuropathic pain. J Exp Med. 2018;215(5):1301–13. 10.1084/jem.20170584 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Mapplebeck JCS, Dalgarno R, Tu Y, et al. : Microglial P2X4R-evoked pain hypersensitivity is sexually dimorphic in rats. Pain. 2018;159(9):1752–1763. [DOI] [PubMed] [Google Scholar]
  • 49. Luo X, Huh Y, Bang S, et al. : Macrophage Toll-like Receptor 9 Contributes to Chemotherapy-Induced Neuropathic Pain in Male Mice. J Neurosci. 2019;39(35):6848–64. 10.1523/JNEUROSCI.3257-18.2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Zhang H, Li Y, de Carvalho-Barbosa M, et al. : Dorsal Root Ganglion Infiltration by Macrophages Contributes to Paclitaxel Chemotherapy-Induced Peripheral Neuropathy. J Pain. 2016;17(7):775–86. 10.1016/j.jpain.2016.02.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Li Y, Zhang H, Zhang H, et al. : Toll-like receptor 4 signaling contributes to Paclitaxel-induced peripheral neuropathy. J Pain. 2014;15(7):712–25. 10.1016/j.jpain.2014.04.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Liu CC, Lu N, Cui Y, et al. : Prevention of paclitaxel-induced allodynia by minocycline: Effect on loss of peripheral nerve fibers and infiltration of macrophages in rats. Mol Pain. 2010;6:76. 10.1186/1744-8069-6-76 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Pachman DR, Dockter T, Zekan PJ, et al. : A pilot study of minocycline for the prevention of paclitaxel-associated neuropathy: ACCRU study RU221408I. Support Care Cancer. 2017;25(11):3407–16. 10.1007/s00520-017-3760-2 [DOI] [PubMed] [Google Scholar]
  • 54. Wang XS, Shi Q, Bhadkamkar NA, et al. : Minocycline for Symptom Reduction During Oxaliplatin-Based Chemotherapy for Colorectal Cancer: A Phase II Randomized Clinical Trial. J Pain Symptom Manage. 2019;58(4):662–71. 10.1016/j.jpainsymman.2019.06.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Tikka T, Fiebich BL, Goldsteins G, et al. : Minocycline, a tetracycline derivative, is neuroprotective against excitotoxicity by inhibiting activation and proliferation of microglia. J Neurosci. 2001;21(8):2580–8. 10.1523/JNEUROSCI.21-08-02580.2001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Yrjänheikki J, Tikka T, Keinänen R, et al. : A tetracycline derivative, minocycline, reduces inflammation and protects against focal cerebral ischemia with a wide therapeutic window. Proc Natl Acad Sci U S A. 1999;96(23):13496–500. 10.1073/pnas.96.23.13496 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Aromolaran KA, Goldstein PA: Ion channels and neuronal hyperexcitability in chemotherapy-induced peripheral neuropathy; cause and effect? Mol Pain. 2017;13:174480691771469. 10.1177/1744806917714693 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Bennett DL, Clark AJ, Huang J, et al. : The Role of Voltage-Gated Sodium Channels in Pain Signaling. Physiol Rev. 2019;99(2):1079–151. 10.1152/physrev.00052.2017 [DOI] [PubMed] [Google Scholar]
  • 59. Heide R, Bostock H, Ventzel L, et al. : Axonal excitability changes and acute symptoms of oxaliplatin treatment: In vivo evidence for slowed sodium channel inactivation. Clin Neurophysiol. 2018;129(3):694–706. 10.1016/j.clinph.2017.11.015 [DOI] [PubMed] [Google Scholar]
  • 60. Argyriou AA, Cavaletti G, Antonacopoulou A, et al. : Voltage-gated sodium channel polymorphisms play a pivotal role in the development of oxaliplatin-induced peripheral neurotoxicity: results from a prospective multicenter study. Cancer. 2013;119(9):3570–7. 10.1002/cncr.28234 [DOI] [PubMed] [Google Scholar]
  • 61. Li Y, North RY, Rhines LD, et al. : DRG Voltage-Gated Sodium Channel 1.7 Is Upregulated in Paclitaxel-Induced Neuropathy in Rats and in Humans with Neuropathic Pain. J Neurosci. 2018;38(5):1124–36. 10.1523/JNEUROSCI.0899-17.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Descoeur J, Pereira V, Pizzoccaro A, et al. : Oxaliplatin-induced cold hypersensitivity is due to remodelling of ion channel expression in nociceptors. EMBO Mol Med. 2011;3(5):266–78. 10.1002/emmm.201100134 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. 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(18):1941–67. 10.1200/JCO.2013.54.0914 [DOI] [PubMed] [Google Scholar]
  • 64. Viatchenko-Karpinski V, Ling J, Gu JG: Down-regulation of Kv4.3 channels and a-type K + currents in V2 trigeminal ganglion neurons of rats following oxaliplatin treatment. Mol Pain. 2018;14(14):1744806917750995. 10.1177/1744806917750995 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Poupon L, Lamoine S, Pereira V, et al. : Targeting the TREK-1 potassium channel via riluzole to eliminate the neuropathic and depressive-like effects of oxaliplatin. Neuropharmacology. 2018;140:43–61. 10.1016/j.neuropharm.2018.07.026 [DOI] [PubMed] [Google Scholar]
  • 66. Kerckhove N, Busserolles J, Stanbury T, et al. : Effectiveness assessment of riluzole in the prevention of oxaliplatin-induced peripheral neuropathy: RILUZOX-01: protocol of a randomised, parallel, controlled, double-blind and multicentre study by the UNICANCER-AFSOS Supportive Care intergroup. BMJ Open. 2019;9(6):e027770. 10.1136/bmjopen-2018-027770 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Chukyo A, Chiba T, Kambe T, et al. : Oxaliplatin-induced changes in expression of transient receptor potential channels in the dorsal root ganglion as a neuropathic mechanism for cold hypersensitivity. Neuropeptides. 2018;67:95–101. 10.1016/j.npep.2017.12.002 [DOI] [PubMed] [Google Scholar]
  • 68. Rigo FK, Dalmolin GD, Trevisan G, et al. : Effect of ω-conotoxin MVIIA and Phα1β on paclitaxel-induced acute and chronic pain. Pharmacol Biochem Behav. 2013;114–115:16–22. 10.1016/j.pbb.2013.10.014 [DOI] [PubMed] [Google Scholar]
  • 69. Flatters SJ, Bennett GJ: Ethosuximide reverses paclitaxel- and vincristine-induced painful peripheral neuropathy. Pain. 2004;109(1–2):150–61. 10.1016/j.pain.2004.01.029 [DOI] [PubMed] [Google Scholar]
  • 70. Dolphin AC: Calcium channel auxiliary α 2δ and β subunits: trafficking and one step beyond. Nat Rev Neurosci. 2012;13(8):542–55. 10.1038/nrn3311 [DOI] [PubMed] [Google Scholar]
  • 71. Han FY, Kuo A, Nicholson JR, et al. : Comparative analgesic efficacy of pregabalin administered according to either a prevention protocol or an intervention protocol in rats with cisplatin-induced peripheral neuropathy. Clin Exp Pharmacol Physiol. 2018;45(10):1067–75. 10.1111/1440-1681.12971 [DOI] [PubMed] [Google Scholar]
  • 72. Shahid M, Subhan F, Ahmad N, et al. : Efficacy of a topical gabapentin gel in a cisplatin paradigm of chemotherapy-induced peripheral neuropathy. BMC Pharmacol Toxicol. 2019;20(1):51. 10.1186/s40360-019-0329-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. de Andrade DC, Jacobsen Teixeira M, Galhardoni R, et al. : Pregabalin for the Prevention of Oxaliplatin-Induced Painful Neuropathy: A Randomized, Double-Blind Trial. Oncologist. 2017;22(10):1154–e105. 10.1634/theoncologist.2017-0235 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Hershman DL, Till C, Wright JD, et al. : Comorbidities and Risk of Chemotherapy-Induced Peripheral Neuropathy Among Participants 65 Years or Older in Southwest Oncology Group Clinical Trials. J Clin Oncol. 2016;34(25):3014–22. 10.1200/JCO.2015.66.2346 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Uwah AN, Ackler J, Leighton JC, Jr, et al. : The effect of diabetes on oxaliplatin-induced peripheral neuropathy. Clin Colorectal Cancer. 2012;11(4):275–9. 10.1016/j.clcc.2012.05.002 [DOI] [PubMed] [Google Scholar]
  • 76. Vincenzi B, Frezza AM, Schiavon G, et al. : Identification of clinical predictive factors of oxaliplatin-induced chronic peripheral neuropathy in colorectal cancer patients treated with adjuvant Folfox IV. Support Care Cancer. 2013;21(5):1313–9. 10.1007/s00520-012-1667-5 [DOI] [PubMed] [Google Scholar]
  • 77. Argyriou AA, Iconomou G, Kalofonos HP: Bortezomib-induced peripheral neuropathy in multiple myeloma: a comprehensive review of the literature. Blood. 2008;112(5):1593–9. 10.1182/blood-2008-04-149385 [DOI] [PubMed] [Google Scholar]
  • 78. Argyriou AA, Bruna J, Genazzani AA, et al. : Chemotherapy-induced peripheral neurotoxicity: management informed by pharmacogenetics. Nat Rev Neurol. 2017;13(8):492–504. 10.1038/nrneurol.2017.88 [DOI] [PubMed] [Google Scholar]
  • 79. Caraceni A, Cherny N, Fainsinger R, et al. : Pain measurement tools and methods in clinical research in palliative care: recommendations of an Expert Working Group of the European Association of Palliative Care. J Pain Symptom Manage. 2002;23(3):239–55. 10.1016/s0885-3924(01)00409-2 [DOI] [PubMed] [Google Scholar]
  • 80. Bennett M: The LANSS Pain Scale: the Leeds assessment of neuropathic symptoms and signs. Pain. 2001;92(1–2):147–57. 10.1016/s0304-3959(00)00482-6 [DOI] [PubMed] [Google Scholar]
  • 81. Bouhassira D, Attal N, Alchaar H, et al. : Comparison of pain syndromes associated with nervous or somatic lesions and development of a new neuropathic pain diagnostic questionnaire (DN4). Pain. 2005;114(1–2):29–36. 10.1016/j.pain.2004.12.010 [DOI] [PubMed] [Google Scholar]
  • 82. Sandier SG, Tobin W, Henderson ES: Vincristine-induced neuropathy. A clinical study of fifty leukemic patients. Neurology. 1969;19(4):367. 10.1212/wnl.19.4.367 [DOI] [PubMed] [Google Scholar]
  • 83. Kandula T, Park SB, Cohn RJ, et al. : Pediatric chemotherapy induced peripheral neuropathy: A systematic review of current knowledge. Cancer Treat Rev. 2016;50:118–28. 10.1016/j.ctrv.2016.09.005 [DOI] [PubMed] [Google Scholar]
  • 84. Lavoie Smith EM, Li L, Hutchinson RJ, et al. : Measuring vincristine-induced peripheral neuropathy in children with acute lymphoblastic leukemia. Cancer Nurs. 2013;36(5):E49–E60. 10.1097/NCC.0b013e318299ad23 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. McGrath PJ, Walco GA, Turk DC, et al. : Core outcome domains and measures for pediatric acute and chronic/recurrent pain clinical trials: PedIMMPACT recommendations. J Pain. 2008;9(9):771–83. 10.1016/j.jpain.2008.04.007 [DOI] [PubMed] [Google Scholar]
  • 86. Postma TJ, Heimans JJ, Muller MJ, et al. : Pitfalls in grading severity of chemotherapy-induced peripheral neuropathy. Ann Oncol. 1998;9(7):739–44. 10.1023/a:1008344507482 [DOI] [PubMed] [Google Scholar]
  • 87. Shimozuma K, Ohashi Y, Takeuchi A, et al. : Feasibility and validity of the Patient Neurotoxicity Questionnaire during taxane chemotherapy in a phase III randomized trial in patients with breast cancer: N-SAS BC 02. Support Care Cancer. 2009;17(12):1483–91. 10.1007/s00520-009-0613-7 [DOI] [PubMed] [Google Scholar]
  • 88. Haryani H, Fetzer SJ, Wu CL, et al. : Chemotherapy-Induced Peripheral Neuropathy Assessment Tools: A Systematic Review. Oncol Nurs Forum. 2017;44(3):E111–E123. 10.1188/17.ONF.E111-E123 [DOI] [PubMed] [Google Scholar]
  • 89. McCrary JM, Goldstein D, Boyle F, et al. : Optimal clinical assessment strategies for chemotherapy-induced peripheral neuropathy (CIPN): a systematic review and Delphi survey. Support Care Cancer. 2017;25(11):3485–93. 10.1007/s00520-017-3772-y [DOI] [PubMed] [Google Scholar]
  • 90. Calhoun EA, Welshman EE, Chang CH, et al. : Psychometric evaluation of the Functional Assessment of Cancer Therapy/Gynecologic Oncology Group-Neurotoxicity (Fact/GOG-Ntx) questionnaire for patients receiving systemic chemotherapy. Int J Gynecol Cancer. 2003;13(6):741–8. [DOI] [PubMed] [Google Scholar]
  • 91. Huang HQ, Brady MF, Cella D, et al. : Validation and reduction of FACT/GOG-Ntx subscale for platinum/paclitaxel-induced neurologic symptoms: A gynecologic oncology group study. Int J Gynecol Cancer. 2007;17(2):387–93. 10.1111/j.1525-1438.2007.00794.x [DOI] [PubMed] [Google Scholar]
  • 92. Cavaletti G, Frigeni B, Lanzani F, et al. : The Total Neuropathy Score as an assessment tool for grading the course of chemotherapy-induced peripheral neurotoxicity: Comparison with the National Cancer Institute-Common Toxicity Scale. J Peripher Nerv Syst. 2007;12(3):210–5. 10.1111/j.1529-8027.2007.00141.x [DOI] [PubMed] [Google Scholar]
  • 93. Cavaletti G, Cornblath DR, Merkies IS, et al. : The chemotherapy-induced peripheral neuropathy outcome measures standardization study: from consensus to the first validity and reliability findings. Ann Oncol. 2013;24(2):454–62. 10.1093/annonc/mds329 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Postma TJ, Aaronson NK, Heimans JJ, et al. : The development of an EORTC quality of life questionnaire to assess chemotherapy-induced peripheral neuropathy: The QLQ-CIPN20. Eur J Cancer. 2005;41(8):1135–9. 10.1016/j.ejca.2005.02.012 [DOI] [PubMed] [Google Scholar]
  • 95. Thompson SW, Davis LE, Kornfeld M, et al. : Cisplatin neuropathy. Clinical, electrophysiologic, morphologic, and toxicologic studies. Cancer. 1984;54(7):1269–75. [DOI] [PubMed] [Google Scholar]
  • 96. Rowinsky EK, Chaudhry V, Cornblath DR, et al. : Neurotoxicity of Taxol. J Natl Cancer Inst Monogr. 1993; (15):107–15. [PubMed] [Google Scholar]
  • 97. Maier C, Baron R, Tölle TR, et al. : Quantitative sensory testing in the German Research Network on Neuropathic Pain (DFNS): somatosensory abnormalities in 1236 patients with different neuropathic pain syndromes. Pain. 2010;150(3):439–50. 10.1016/j.pain.2010.05.002 [DOI] [PubMed] [Google Scholar]
  • 98. Zhi WI, Chen P, Kwon A, et al. : Chemotherapy-induced peripheral neuropathy (CIPN) in breast cancer survivors: a comparison of patient-reported outcomes and quantitative sensory testing. Breast Cancer Res Treat. 2019;178(3):587–95. 10.1007/s10549-019-05416-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Dougherty PM, Cata JP, Cordella JV, et al. : Taxol-induced sensory disturbance is characterized by preferential impairment of myelinated fiber function in cancer patients. Pain. 2004;109(1–2):132–42. 10.1016/j.pain.2004.01.021 [DOI] [PubMed] [Google Scholar]
  • 100. Cata JP, Weng HR, Burton AW, et al. : Quantitative sensory findings in patients with bortezomib-induced pain. J Pain. 2007;8(4):296–306. 10.1016/j.jpain.2006.09.014 [DOI] [PubMed] [Google Scholar]
  • 101. Dougherty PM, Cata JP, Burton AW, et al. : Dysfunction in multiple primary afferent fiber subtypes revealed by quantitative sensory testing in patients with chronic vincristine-induced pain. J Pain Symptom Manage. 2007;33(2):166–79. 10.1016/j.jpainsymman.2006.08.006 [DOI] [PubMed] [Google Scholar]
  • 102. Wang XS, Shi Q, Dougherty PM, et al. : Prechemotherapy Touch Sensation Deficits Predict Oxaliplatin-Induced Neuropathy in Patients with Colorectal Cancer. Oncology. 2016;90(3):127–35. 10.1159/000443377 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Roldan CJ, Johnson C, Lee SO, et al. : Subclinical Peripheral Neuropathy in Patients with Head and Neck Cancer: A Quantitative Sensory Testing (QST) Study. Pain Physician. 2018;21(4):E419–E427. 10.36076/ppj.2018.4.E419 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Ventzel L, Madsen CS, Karlsson P, et al. : Chronic Pain and Neuropathy Following Adjuvant Chemotherapy. Pain Med. 2018;19(9):1813–24. 10.1093/pm/pnx231 [DOI] [PubMed] [Google Scholar]
  • 105. Forstenpointner J, Oberlojer VC, Naleschinski D, et al. : A-Fibers Mediate Cold Hyperalgesia in Patients with Oxaliplatin-Induced Neuropathy. Pain Pract. 2018;18(6):758–67. 10.1111/papr.12670 [DOI] [PubMed] [Google Scholar]
  • 106. Geber C, Breimhorst M, Burbach B, et al. : Pain in chemotherapy-induced neuropathy–more than neuropathic? Pain. 2013;154(12):2877–87. 10.1016/j.pain.2013.08.028 [DOI] [PubMed] [Google Scholar]
  • 107. Delmotte JB, Beaussier H, Auzeil N, et al. : Is quantitative sensory testing helpful in the management of oxaliplatin neuropathy? a two-year clinical study. Cancer Treat Res Commun. 2018;17:31–6. 10.1016/j.ctarc.2018.10.002 [DOI] [PubMed] [Google Scholar]
  • 108. Mangus LM, Rao DB, Ebenezer GJ: Intraepidermal Nerve Fiber Analysis in Human Patients and Animal Models of Peripheral Neuropathy: A Comparative Review. Toxicol Pathol. 2020;48(1):59–70. 10.1177/0192623319855969 [DOI] [PubMed] [Google Scholar]
  • 109. Krøigård T, Svendsen TK, Wirenfeldt M, et al. : Early changes in tests of peripheral nerve function during oxaliplatin treatment and their correlation with chemotherapy-induced polyneuropathy symptoms and signs. Eur J Neurol. 2020;27(1):68–76. 10.1111/ene.14035 [DOI] [PubMed] [Google Scholar]
  • 110. Burakgazi AZ, Messersmith W, Vaidya D, et al. : Longitudinal assessment of oxaliplatin-induced neuropathy. Neurology. 2011;77(10):980–6. 10.1212/WNL.0b013e31822cfc59 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Richardson PG, Xie W, Mitsiades C, et al. : Single-agent bortezomib in previously untreated multiple myeloma: efficacy, characterization of peripheral neuropathy, and molecular correlations with response and neuropathy. J Clin Oncol. 2009;27(21):3518–25. 10.1200/JCO.2008.18.3087 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Koskinen MJ, Kautio AL, Haanpää ML, et al. : Intraepidermal nerve fibre density in cancer patients receiving adjuvant chemotherapy. Anticancer Res. 2011;31(12):4413–6. [PubMed] [Google Scholar]
  • 113. Griffith KA, Dorsey SG, Renn CL, et al. : Correspondence between neurophysiological and clinical measurements of chemotherapy-induced peripheral neuropathy: secondary analysis of data from the CI-PeriNomS study. J Peripher Nerv Syst. 2014;19(2):127–35. 10.1111/jns5.12064 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Hanai A, Ishiguro H, Sozu T, et al. : Effects of Cryotherapy on Objective and Subjective Symptoms of Paclitaxel-Induced Neuropathy: Prospective Self-Controlled Trial. J Natl Cancer Inst. 2018;110(2):141–8. 10.1093/jnci/djx178 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Griffiths C, Kwon N, Beaumont JL, et al. : Cold therapy to prevent paclitaxel-induced peripheral neuropathy. Support Care Cancer. 2018;26(10):3461–9. 10.1007/s00520-018-4199-9 [DOI] [PubMed] [Google Scholar]
  • 116. Ruddy KJ, Le-Rademacher J, Lacouture ME, et al. : Randomized controlled trial of cryotherapy to prevent paclitaxel-induced peripheral neuropathy (RU221511I); an ACCRU trial. Breast. 2019;48:89–97. 10.1016/j.breast.2019.09.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Sundar R, Bandla A, Tan SS, et al. : Limb Hypothermia for Preventing Paclitaxel-Induced Peripheral Neuropathy in Breast Cancer Patients: A Pilot Study. Front Oncol. 2017;6:274. 10.3389/fonc.2016.00274 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Smith EM, Pang H, Cirrincione C, et al. : Effect of duloxetine on pain, function, and quality of life among patients with chemotherapy-induced painful peripheral neuropathy: a randomized clinical trial. JAMA. 2013;309(13):1359. 10.1001/jama.2013.2813 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Gewandter JS, Dworkin RH, Finnerup NB, et al. : Painful chemotherapy-induced peripheral neuropathy: lack of treatment efficacy or the wrong clinical trial methodology? Pain. 2017;158(1):30–3. 10.1097/j.pain.0000000000000653 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Dworkin RH, Turk DC, Farrar JT, et al. : Core outcome measures for chronic pain clinical trials: IMMPACT recommendations. Pain. 2005;113(1–2):9–19. 10.1016/j.pain.2004.09.012 [DOI] [PubMed] [Google Scholar]
  • 121. Farshchian N, Alavi A, Heydarheydari S, et al. : Comparative study of the effects of venlafaxine and duloxetine on chemotherapy-induced peripheral neuropathy. Cancer Chemother Pharmacol. 2018;82(5):787–93. 10.1007/s00280-018-3664-y [DOI] [PubMed] [Google Scholar]
  • 122. Demant DT, Lund K, Vollert J, et al. : The effect of oxcarbazepine in peripheral neuropathic pain depends on pain phenotype: a randomised, double-blind, placebo-controlled phenotype-stratified study. Pain. 2014;155(11):2263–73. 10.1016/j.pain.2014.08.014 [DOI] [PubMed] [Google Scholar]
  • 123. Attal N, Rouaud J, Brasseur L, et al. : Systemic lidocaine in pain due to peripheral nerve injury and predictors of response. Neurology. 2004;62(2):218–25. 10.1212/01.wnl.0000103237.62009.77 [DOI] [PubMed] [Google Scholar]
  • 124. Finnerup NB, Sindrup SH, Bach FW, et al. : Lamotrigine in spinal cord injury pain: a randomized controlled trial. Pain. 2002;96(3):375–83. 10.1016/s0304-3959(01)00484-5 [DOI] [PubMed] [Google Scholar]
  • 125. Mainka T, Malewicz NM, Baron R, et al. : Presence of hyperalgesia predicts analgesic efficacy of topically applied capsaicin 8% in patients with peripheral neuropathic pain. Eur J Pain. 2016;20(1):116–29. 10.1002/ejp.703 [DOI] [PubMed] [Google Scholar]
  • 126. Fallon MT, Storey DJ, Krishan A, et al. : Cancer treatment-related neuropathic pain: proof of concept study with menthol--a TRPM8 agonist. Support Care Cancer. 2015;23(9):2769–77. 10.1007/s00520-015-2642-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Barton DL, Wos EJ, Qin R, et al. : A double-blind, placebo-controlled trial of a topical treatment for chemotherapy-induced peripheral neuropathy: NCCTG trial N06CA. Support Care Cancer. 2011;19(6):833–41. 10.1007/s00520-010-0911-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Anand P, Elsafa E, Privitera R, et al. : Rational treatment of chemotherapy-induced peripheral neuropathy with capsaicin 8% patch: from pain relief towards disease modification. J Pain Res. 2019;12:2039–52. 10.2147/JPR.S213912 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Mulpuri Y, Marty VN, Munier JJ, et al. : Synthetic peripherally-restricted cannabinoid suppresses chemotherapy-induced peripheral neuropathy pain symptoms by CB1 receptor activation. Neuropharmacology. 2018;139:85–97. 10.1016/j.neuropharm.2018.07.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Brenneman DE, Kinney WA, Ward SJ: Knockdown siRNA Targeting the Mitochondrial Sodium-Calcium Exchanger-1 Inhibits the Protective Effects of Two Cannabinoids Against Acute Paclitaxel Toxicity. J Mol Neurosci. 2019;68(4):603–19. 10.1007/s12031-019-01321-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Sierra S, Gupta A, Gomes I, et al. : Targeting Cannabinoid 1 and Delta Opioid Receptor Heteromers Alleviates Chemotherapy-Induced Neuropathic Pain. ACS Pharmacol Transl Sci. 2019;2(4):219–29. 10.1021/acsptsci.9b00008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. King KM, Myers AM, Soroka-Monzo AJ, et al. : Single and combined effects of Δ 9 -tetrahydrocannabinol and cannabidiol in a mouse model of chemotherapy-induced neuropathic pain. Br J Pharmacol. 2017;174(17):2832–41. 10.1111/bph.13887 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Blanton HL, Brelsfoard J, DeTurk N, et al. : Cannabinoids: Current and Future Options to Treat Chronic and Chemotherapy-Induced Neuropathic Pain. Drugs. 2019;79(9):969–95. 10.1007/s40265-019-01132-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Lynch ME, Cesar-Rittenberg P, Hohmann AG: A double-blind, placebo-controlled, crossover pilot trial with extension using an oral mucosal cannabinoid extract for treatment of chemotherapy-induced neuropathic pain. J Pain Symptom Manage. 2014;47(1):166–73. 10.1016/j.jpainsymman.2013.02.018 [DOI] [PubMed] [Google Scholar]
  • 135. Finnerup NB, Attal N, Haroutounian S, et al. : Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis. Lancet Neurol. 2015;14(2):162–73. 10.1016/S1474-4422(14)70251-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Hofmeister M, Memedovich A, Brown S, et al. : Effectiveness of Neurostimulation Technologies for the Management of Chronic Pain: A Systematic Review. Neuromodulation. 2020;23(2):150–157. 10.1111/ner.13020 [DOI] [PubMed] [Google Scholar]
  • 137. Groenen PS, van Helmond N, Chapman KB: Chemotherapy-Induced Peripheral Neuropathy Treated with Dorsal Root Ganglion Stimulation. Pain Med. 2019;20(4):857–9. 10.1093/pm/pny209 [DOI] [PubMed] [Google Scholar]
  • 138. Abd-Elsayed A, Schiavoni N, Sachdeva H: Efficacy of spinal cord stimulators in treating peripheral neuropathy: a case series. J Clin Anesth. 2016;28:74–7. 10.1016/j.jclinane.2015.08.011 [DOI] [PubMed] [Google Scholar]
  • 139. Gewandter JS, Chaudari J, Ibegbu C, et al. : Wireless transcutaneous electrical nerve stimulation device for chemotherapy-induced peripheral neuropathy: an open-label feasibility study. Support Care Cancer. 2019;27(5):1765–74. 10.1007/s00520-018-4424-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Loprinzi C, Le-Rademacher JG, Majithia N, et al. : Scrambler therapy for chemotherapy neuropathy: a randomized phase II pilot trial. Support Care Cancer. 2020;28(3):1183–97. 10.1007/s00520-019-04881-3 [DOI] [PubMed] [Google Scholar]
  • 141. Smith TJ, Razzak AR, Blackford AL, et al. : A Pilot Randomized Sham-Controlled Trial of MC5-A Scrambler Therapy in the Treatment of Chronic Chemotherapy-Induced Peripheral Neuropathy (CIPN). J Palliat Care. 2020;35(1):53–8. 10.1177/0825859719827589 [DOI] [PubMed] [Google Scholar]
  • 142. Paley CA, Johnson MI, Tashani OA, et al. : Acupuncture for cancer pain in adults. Cochrane Database Syst Rev. 2015; (10):CD007753. 10.1002/14651858.CD007753.pub3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143. Jeong YJ, Kwak MA, Seo JC, et al. : Acupuncture for the Treatment of Taxane-Induced Peripheral Neuropathy in Breast Cancer Patients: A Pilot Trial. Evid Based Complement Alternat Med. 2018;2018:1–11. 10.1155/2018/5367014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Zhi WI, Ingram E, Li SQ, et al. : Acupuncture for Bortezomib-Induced Peripheral Neuropathy: Not Just for Pain. Integr Cancer Ther. 2018;17(4):1079–86. 10.1177/1534735418788667 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145. Molassiotis A, Suen LKP, Cheng HL, et al. : A Randomized Assessor-Blinded Wait-List-Controlled Trial to Assess the Effectiveness of Acupuncture in the Management of Chemotherapy-Induced Peripheral Neuropathy. Integr Cancer Ther. 2019;18: 1534735419836501. 10.1177/1534735419836501 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Li K, Giustini D, Seely D: A systematic review of acupuncture for chemotherapy-induced peripheral neuropathy. Curr Oncol. 2019;26(2):e147–e154. 10.3747/co.26.4261 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Zimmer P, Trebing S, Timmers-Trebing U, et al. : Eight-week, multimodal exercise counteracts a progress of chemotherapy-induced peripheral neuropathy and improves balance and strength in metastasized colorectal cancer patients: a randomized controlled trial. Support Care Cancer. 2018;26(2):615–24. 10.1007/s00520-017-3875-5 [DOI] [PubMed] [Google Scholar]
  • 148. Duregon F, Vendramin B, Bullo V, et al. : Effects of exercise on cancer patients suffering chemotherapy-induced peripheral neuropathy undergoing treatment: A systematic review. Crit Rev Oncol Hematol. 2018;121:90–100. 10.1016/j.critrevonc.2017.11.002 [DOI] [PubMed] [Google Scholar]
  • 149. Kleckner IR, Kamen C, Gewandter JS, et al. : Effects of exercise during chemotherapy on chemotherapy-induced peripheral neuropathy: a multicenter, randomized controlled trial. Support Care Cancer. 2018;26(4):1019–28. 10.1007/s00520-017-4013-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150. van Waart H, Stuiver MM, van Harten WH, et al. : Effect of Low-Intensity Physical Activity and Moderate- to High-Intensity Physical Exercise During Adjuvant Chemotherapy on Physical Fitness, Fatigue, and Chemotherapy Completion Rates: Results of the PACES Randomized Clinical Trial. J Clin Oncol. 2015;33(17):1918–27. 10.1200/JCO.2014.59.1081 [DOI] [PubMed] [Google Scholar]
  • 151. Mundal I, Gråwe RW, Bjørngaard JH, et al. : Psychosocial factors and risk of chronic widespread pain: an 11-year follow-up study--the HUNT study. Pain. 2014;155(8):1555–61. 10.1016/j.pain.2014.04.033 [DOI] [PubMed] [Google Scholar]
  • 152. Lee KM, Jung D, Hwang H, et al. : Pre-treatment anxiety is associated with persistent chemotherapy-induced peripheral neuropathy in women treated with neoadjuvant chemotherapy for breast cancer. J Psychosom Res. 2018;108:14–9. 10.1016/j.jpsychores.2018.02.012 [DOI] [PubMed] [Google Scholar]
  • 153. Knoerl R, Smith EML, Barton DL, et al. : Self-Guided Online Cognitive Behavioral Strategies for Chemotherapy-Induced Peripheral Neuropathy: A Multicenter, Pilot, Randomized, Wait-List Controlled Trial. J Pain. 2018;19(4):382–94. 10.1016/j.jpain.2017.11.009 [DOI] [PubMed] [Google Scholar]
  • 154. Knoerl R, Barton DL, Holden JE, et al. : Potential mediators of improvement in painful chemotherapy-induced peripheral neuropathy via a web-based cognitive behavioural intervention. Can Oncol Nurs J. 2018;28(3):178–83. 10.5737/23688076283178183 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155. Bruna J, Videla S, Argyriou AA, et al. : Efficacy of a Novel Sigma-1 Receptor Antagonist for Oxaliplatin-Induced Neuropathy: A Randomized, Double-Blind, Placebo-Controlled Phase IIa Clinical Trial. Neurotherapeutics. 2018;15(1):178–89. 10.1007/s13311-017-0572-5 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from F1000Research are provided here courtesy of F1000 Research Ltd

RESOURCES