Abstract
Objective:
To describe the known predictors and pathophysiological mechanisms of chronic painful chemotherapy-induced peripheral neuropathy (CIPN) in cancer survivors and the challenges in assessing and managing it.
Data Sources:
PubMed/Medline, CINAHL, Scopus, and PsycINFO.
Conclusion:
The research on chronic painful CIPN is limited. Additional research is needed to identify the predictors and pathophysiological mechanisms of chronic painful CIPN to inform the development of assessment tools and management options for this painful and possibly debilitating condition.
Implications for Nursing Practice:
Recognition of the predictors of chronic painful CIPN and proactive CIPN assessment and palliative management are important steps in reducing its impact on physical function and quality of life.
Keywords: chemotherapy-induced peripheral neuropathy, neuropathic pain, polyneuropathy, cancer, chronic pain, centralized pain
Chemotherapy-induced peripheral neuropathy (CIPN), often described as numbness, tingling, and neuropathic (burning, freezing, zapping, or shock-like) pain in the hands and feet, is experienced by 20% to 85% of individuals who receive neurotoxic chemotherapy.1 Common preclinical manifestations of CIPN, primarily a sensorimotor axonal neuropathy, are loss of vibration and temperature sensation, proprioception, and deep tendon reflexes. Individuals may also report difficulty with balance, impaired fine and gross motor skills, and muscle cramps and weakness. However, autonomic symptoms such as orthostatic hypotension, loss of bladder control, constipation, hearing loss, and difficulty obtaining an erection have also been reported.2,3 The effect of CIPN on individuals’ ability to perform activities such as buttoning a shirt, typing or writing, climbing stairs, and avoiding falls in turn affects their occupational and social role performance.4–8 Some individuals report a sense of isolation, a lost sense of purpose, and depression.9–11 Because no curative treatments for CIPN are known,12 it is a leading chemotherapy dose-limiting factor.13–16
Neuropathic pain caused by CIPN is generally chronic and often refractory to treatment. The chronic central nervous system (CNS) changes that underlie chronic painful CIPN may also increase an individual’s susceptibility to or exacerbate other centrally mediated symptoms (eg, fatigue, emotional distress, insomnia).17–21 While research on nonpainful CIPN is fairly abundant,12,22–31 few reviews focus specifically on chronic painful CIPN. The purpose of this review is to raise awareness of this under-recognized chronic pain condition by summarizing the known predictors, pathophysiological mechanisms, assessment methods, and treatments of chronic painful CIPN in cancer survivors.
CIPN Patterns and Pathophysiology
Table 1 lists the various classes of neurotoxic chemotherapy—proteasome inhibitors, platinums, taxanes, vinca alkaloids, and thalidomides—known to most commonly cause CIPN.2,3,14,29,32–44 Ifosfamide, epothilones, and topoisomerase inhibitors are excluded from the table because less evidence supports their association with high-grade CIPN. Table 1 also includes the specific CIPN-causing agents and the CIPN manifestations, pain incidence, and pain-causing mechanisms associated with each agent.
Table 1.
Drug Class | Specific Agents |
Prominent Manifestations |
Pain Incidence | Sensitization Mechanism |
---|---|---|---|---|
Proteasome inhibitors | Bortezomib |
*Neuropathic pain and allodynia32,33 Hypoesthesia Loss of fine motor function Cramps2,3 |
Up to 47%3,34 | TRPV, TRPA1, and NMDA receptor up-regulation Inflammation (cytokine release and macrophage infiltration) Altered enzyme activity (reductions in phosphoglycerate dehydrogenase and L-serine; protein kinase C → NMDA receptor upregulation and increased glutamate release) |
Platinums | Oxaliplatin, cisplatin, carboplatin | Acute cold hyperalgesia (oxaliplatin alone) *Sensory Loss of vibration sensation and sensory ataxia |
5%−50%14,33,36 | TRPV, TRPA1, and NMDA receptor upregulation Ion channel (sodium, calcium, and potassium) dysfunction |
Taxanes | Paclitaxel, docetaxel |
* Sensory14 Muscle weakness |
Up to 30%14 | TRPV, TRPA1, and NMDA receptor upregulation Ion channel (sodium, calcium, and potassium) dysfunction Inflammation (cytokine release and macrophage infiltration) |
Vinca alkaloids | Vincristine, vinblastine, vinorelbine, vindesine |
* Sensory and motor29,37,38 Autonomic |
11%−44%29,39–41 | Serotonin increase and channel upregulation |
Thalidomides | Thalidomide, lenalidomide |
*Autonomic42 *Hypoesthesia43,44 |
Uncommon44 | Inflammation (cytokine release, and macrophage infiltration) |
Indicates the most prominent manifestations. Sensory refers primarily to numbness and tingling.
Abbreviations: NMDA, N-methyl-D-aspartate; TRP, transient receptor potential.
Acute CIPN
Specific types of neurotoxic chemotherapy (ie, oxaliplatin and bortezomib) induce acute painful CIPN. In 85% to 95% of individuals, oxaliplatin causes reversible painful cold hypersensitivity in the face, throat, hands, and feet, and muscle cramps.36,45 Painful CIPN can manifest quickly, even before the third chemotherapy cycle, in up to 47% of individuals receiving bortezomib.46
With the exception of acute CIPN pain patterns, nonpainful manifestations of CIPN generally precede painful symptoms.45,47 Nonpainful numbness and tingling generally progress distally to proximally, affecting the toes and fingertips first, then advancing up the extremities.27,28, 48 Nonpainful CIPN is sometimes called cumulative CIPN because its severity and duration usually increase with each additional dose of neurotoxic chemotherapy.45,49 Even after completion of treatment, nonpainful and painful CIPN symptoms can develop or worsen in individuals who have received platinums49 and vinca alkaloids.50
Acute CIPN pathophysiology.
Various mechanisms underlying CIPN development have been proposed: primarily, disruption of neuron cell metabolism (mitochondrial51 and enzyme33,52 function) and ion channel function; alteration of gene and protein expression; upregulation of N-methyl-D-aspartate (NMDA) and transient receptor potential (TRP) receptors; and inflammation. Neuron dysfunction that leads to an increase in the neurotransmitters serotonin and glutamate may also facilitate the development of painful CIPN. These changes can contribute to oxidative stress53,54 and neuron hyperexcitability, demyelination, and apoptosis (cell death). The primary sites directly or indirectly affected by neurotoxic chemotherapy are the dorsal root ganglia, intraepidermal neurons, c-fiber sensory neuron axons and cell bodies, wide dynamic range neurons (WDRN) in the spinal cord, and the thalamus and hypothalamus.26,52,55–57 The dorsal root ganglia are collections of peripheral sensory neuron cell bodies near each spinal cord nerve root that relay sensory information. The sensory intraepidermal neurons include pain-signaling c-fibers that extend into the skin. The WDRN in the spinal cord dorsal horn and the thalamus in the brain process information from various painful and nonpainful sensory inputs and inhibitory signals, then relay information to appropriate areas of the brain. The mechanisms of acute nonpainful CIPN may differ based on the type of neurotoxic chemotherapy; however, acute CIPN may advance to chronic painful CIPN via shared mechanisms.
Chronic painful CIPN
Up to 40% of individuals who receive neurotoxic chemotherapy develop chronic painful CIPN,1,14,36,47 which has previously been defined as centralized pain caused by pathologic changes or disturbances in function of one or several nerves that persists (a) for at least 3 months or (b) after the visible somatic and/or nerve tissue has healed.58 The persistence of pain is generally understood to result from chemotherapy-induced neuronal changes (ie, sensitization) in the CNS.
Chronic painful CIPN pathophysiology.
Sensitization can result in increased peripheral and/or central neuron excitability—magnitude and duration of response to received pain signals—and constant or spontaneous neuron activation initiating in abnormal sites (outside the axon hillock) of the neuron. It manifests with allodynia (pain elicited by normally nonpainful, low-intensity stimuli), hyperalgesia (heightened pain-severity response to painful stimuli), dysesthesia (abnormal painful sensation, such as burning and pins-and-needles sensations), and continuous or shooting pain.59 Peripheral sensitization can cause persistent uncontrolled pain signaling to and sensitization of the WDRN and supportive (ie, satellite, Schwann, and glial) cells in the spinal cord dorsal horn, and in the thalamus and primary somatosensory cortex of the brain.26,60,61
Central sensitization may also result from direct chemotoxic damage62–64 and/or dysfunction of the CNS descending pain-modulating pathways.65–68 Very few studies have reported chemotherapy effects on descending pain-modulating pathways; however, emerging evidence suggests that analgesia through the descending pain-modulating pathway, particularly involving the lateral hypothalamus and orexinergic system, may be key in combatting CIPN pain.65–68 The longer CIPN goes unmanaged, the more central sensitization progresses; painful CIPN then becomes chronic.
Predictors and Comorbidities of Painful CIPN
Research is now beginning to uncover the predictors of chronic painful CIPN. Some evidence suggests that individuals who have more severe CIPN during chemotherapy treatment35,49,69 experience preclinical sensory changes during chemotherapy (eg, thermal hyperalgesia)35 or have a pre-existing diagnosis of osteoarthritis69,70 may be at higher risk for developing chronic painful neuropathy following treatment with neurotoxic chemotherapy. In addition, being born premature, and having a lower income, a higher number of comorbidities, and/or back pain have also been shown to be associated with chronic painful CIPN.70 Evidence is mixed for the role of age,35,69–72 cumulative neurotoxic chemotherapy dose,14,35,69,70 diabetes,69,70 alcohol consumption,14,70 body mass index,14,70 and type of neurotoxic chemotherapy14,49,70,73 in the development of chronic painful CIPN. Indicators that have not been associated with chronic painful CIPN development include gender;35,70,71 educational,70 marital,70 and smoking14 status; and ethnicity.70,72 Finally, mindfulness has been linked to less severe chronic painful CIPN.71 Overall, the described potential predictors of chronic painful CIPN must be interpreted cautiously because the reviewed studies used varying measures of pain and/or CIPN and definitions of chronic neuropathy (eg, did not specifically measure pain separately).
Various chronic pain conditions are known to co-occur with other physical and psychological symptoms that contribute to reductions in physical function and/or quality of life.74–76 However, research on the comorbidities associated with chronic painful CIPN is particularly scant. Increased fatigue, insomnia,77 and anxiety and/or depression severity14,77 are commonly observed in patients with chronic painful CIPN. Most recently, Knoerl and colleagues18 examined the prevalence of the sleep impairment, pain, anxiety, depression, and low energy/fatigue (SPADE) symptom cluster and its association with pain-related interference in a sample of patients with chronic painful CIPN (n = 59). Frequencies of moderate to severe SPADE symptoms were characterized based on predefined cut-off scores on the 0 to 10 average pain intensity numerical rating scale and the Patient-Reported Outcome Measurement Information System (PROMIS)78 subscales. Participants frequently experienced moderate levels of average pain (67.8%), fatigue (62.7%), sleep-related impairment (69.5%), and anxiety (30.5%), while depression (10.2%) was less prevalent. Clusters of SPADE symptoms were common: 54.2% of participants experienced at least three SPADE symptoms concurrently. Further, the number of SPADE symptoms reported by participants (0–5) was moderately correlated with pain interference scores (r = 0.48).18 Additional research is needed to elucidate predictors of and symptoms that co-occur with chronic painful CIPN so that clinicians can identify those at greatest risk of developing painful CIPN. During chemotherapy, proactive referral for individualized therapy for CIPN-associated SPADE symptoms may help to mitigate the effects of chronic painful CIPN on physical function after completion of chemotherapy.
Assessment of Painful CIPN
Several clinical evaluation and patient-reported outcome (PRO) measures are available to assess painful CIPN in the clinical and research settings. Methods of clinical evaluation of painful CIPN include quantitative sensory testing (QST), skin biopsy, and several variants of the Total Neuropathy Score (ie, TNSc, TNSr, TNS-PV, Ped mTNS).79–82 QST methods utilize different sensory stimuli such as heat, cold, and pressure to test for pain sensitivity (ie, hyperalgesia and allodynia).83–85 Skin biopsy may be used to measure intraepidermal nerve fiber density (IENFD) to evaluate for reduced c-fiber (pain-sensing nerve) density, which may be associated with painful CIPN.86,87
Several PRO measures subjectively evaluate the incidence and severity of CIPN. Some examples of these measures are the Oxaliplatin-Associated Neurotoxicity Questionnaire,88 the European Organisation for Research and Treatment of Cancer-Quality of Life Questionnaire-CIPN 20-item scale (QLQ-CIPN20),89 and proposed variants of the QLQ-CIPN20.90
While measuring specific symptoms like numbness and tingling is vital to understanding the presentation of CIPN, many of these PRO measures do not operationalize painful CIPN using the word pain. To our knowledge, the following seven measures assess painful CIPN, among other symptoms: the Neuropathic Pain Scale for Chemotherapy-Induced Neuropathy (NPS-CIN),91 the QLQ-CIPN20,89 the Oxaliplatin-Associated Neurotoxicity Questionnaire,88 the Scale for Chemotherapy-Induced Long Term Neurotoxicity (SCIN),92 the Functional Assessment of Cancer Therapy-Taxane (FACT-Taxane),93 the Treatment-induced Neuropathy Assessment Scale (TNAS),94 and the Patient Neurotoxicity Questionnaire (PNQ).95 These scales ask only a few questions regarding painful CIPN, except the NPS-CIN, which includes six neuropathy-specific pain items.91 The QLQ-CIPN20, Oxaliplatin-Associated Neurotoxicity Questionnaire, and SCIN each include two questions about painful CIPN symptoms; the FACT-Taxane and TNAS each ask one question about pain. 88–90,92,94
Limitations of painful CIPN measurement
Both clinical evaluation methods and PRO measures of painful CIPN have limitations, whether from inadequate psychometric properties or poor feasibility. The TNS and its variants, QST, and skin biopsy are psychometrically strong measures; however, the feasibility of these methods within clinical and research settings is limited by equipment expense and time and testertraining requirements.84 Further, none of the clinical evaluation methods comprehensively assess the CIPN symptom experience because of their limited number or absence of subjective and pain-specific items.80,96–100 While many studies use clinician-graded scales like the National Cancer Institute Common Toxicity Criteria for Adverse Events (NCI-CTCAE)101 to quantify neuropathy severity, they do not evaluate painful CIPN. Thus, research studies often use CIPN PRO measures in addition to QST and/or the TNS.102–104
Given the dearth of pain-specific CIPN PRO measures, previous randomized controlled trials investigating treatments for painful CIPN have used both CIPN- and non-CIPN–specific pain PRO measures to operationalize pain outcomes. Some examples of pain measures commonly coupled with validated CIPN measures include the Brief Pain Inventory (BPI), the Numeric Rating Scale (NRS), and the Neuropathic Pain Symptom Inventory (NPSI).81,102,105–109
The lack of a comprehensive gold-standard measure of painful CIPN is detrimental to randomized controlled trials that attempt to evaluate various interventions for painful CIPN. Additionally, the use of multiple modes of CIPN assessment—clinical evaluation, PRO, and/or sensory function tests—can increase participant burden. Thus, future research is needed to develop comprehensive measurement tools that include questions about painful CIPN to improve intervention research testing prevention and management strategies for painful CIPN.
CIPN Management
No curative treatments for CIPN have been discovered. This section summarizes the strongest evidence supporting the use of pharmacologic and complementary treatments, as well as preliminary results from preclinical studies of promising new treatments.
Pharmacologic interventions: clinical studies
Duloxetine is the only drug recommended by the American Society of Clinical Oncology to treat established, painful CIPN caused by oxaliplatin or paclitaxel.81,110 This recommendation is based on a systematic review of 48 randomized controlled trials that were designed to test 22 different pharmacologic interventions.12 seven81,105,106,111–114 of the 48 studies included in the review specifically tested drug interventions for painful CIPN (ie, duloxetine,81 nortriptyline,113 gabapentin,106 amitriptyline,114 lamotrigine,105 or topical amitriptyline ketamine and baclofen combinations 111,112); only one of the seven studies revealed an effective treatment: duloxetine.81 However, it should be noted that, in the other six pain studies, none of which demonstrated an effect, limitations because of suboptimal pain measurement, lack of control for confounding factors, high attrition, and low statistical power could have resulted in an inability to detect truly effective treatments.115
The study that provides evidence of duloxetine efficacy for painful CIPN was a double-blind, randomized, placebo-controlled, crossover trial exploring whether duloxetine 60 mg taken orally once daily would decrease CIPN pain severity (primary aim) following paclitaxel or oxaliplatin treatment.81 Participants (N = 231) had > grade-1 sensory CIPN (per the NCI-CTCAE v4.0) and an average CIPN neuropathic pain score ≥ 4 (0–10 scale) for ≥ 3 months after chemotherapy. Patients were randomized to receive either placebo or duloxetine 60 mg daily. Individuals receiving duloxetine had a larger mean decrease in average pain score than did those in the placebo group (P = .003) (es = 0.513). Further, 33% and 21% experienced a clinically significant reduction in pain score: 30% and 50%, respectively. Side effects were generally mild and occurred infrequently. Duloxetine- and placebo-related fatigue (7%/5%), insomnia (5%/7%), and nausea (5%/3%) were the most commonly reported adverse events. Also, a secondary data analysis suggested that duloxetine worked better for pain induced by oxaliplatin than by paclitaxel.9
Given that chronic painful CIPN is a centralized pain condition, it is not surprising that a centrally acting drug (ie, duloxetine) is effective. Serotonin (5-HT) and norepinephrine (NE) dual reuptake inhibitors (SNRI) such as duloxetine work by increasing the amounts of key pain-inhibiting neurotransmitters—5-HT and NE—within the CNS descending pain-modulating pathways.116 These neurotransmitters suppress the transmission of painful stimuli arising from the periphery by inhibiting input to the WDRN in the spinal cord.117–119 Duloxetine exerts its analgesic action by blocking serotonin (SLC6A4) and norepinephrine (SLC6A2) transporters that are responsible for reuptake in the pre-/post-synaptic cleft.120
Drugs that target peripheral nerve function would not be expected to have a significant effect on chronic centralized CIPN pain. However, preliminary evidence suggests that peripherally acting drugs may in fact work to prevent chronic CIPN pain if administered before neurotoxic chemotherapy begins. Specifically, in addition to its central effects in the CNS/spinal cord, duloxetine exhibits a local effect by blocking Nav1.7 sodium channel currents,121,122 thereby inhibiting the transmission of spontaneous nerve impulses from the periphery to the CNS. Thus, drugs that target peripheral mechanisms could subsequently prevent central sensitization and chronic painful CIPN. Because the effect of peripherally acting drugs to prevent chronic painful CIPN has not yet been demonstrated, more well-designed clinical studies are needed in this area.
Given that duloxetine is the only treatment option recommended for painful CIPN, clinicians often prescribe drugs with efficacy in other neuropathic pain conditions (eg, tricyclic antidepressants, venlafaxine, gabapentin, pregabalin). This practice is cautiously supported by American Society of Clinical Oncology and other experts because duloxetine may be ineffective, contraindicated, or poorly tolerated in some patients.12,123,124 Although opioids are frequently used to treat neuropathic pain,125–127 they should not be used as first-line treatment for chronic CIPN pain because of the risk of addiction, overdose-associated mortality,128 and opioid-induced hyperalgesia—a condition whereby opioids make pain worse.129
Several other promising pharmacologic agents might be effective for painful CIPN but require further testing. Cannabis has demonstrated efficacy for neuropathic pain from peripheral neuropathy caused by HIV and diabetes,130–136 but cannot be recommended for CIPN because of a lack of studies for this specific indication. Small quasi-experimental studies provide preliminary evidence that high-dose 8% capsaicin patches137 and intravenous lidocaine138 may ameliorate painful CIPN. However, larger studies with control group comparisons are needed to confirm or refute these preliminary findings.
Natural products and complementary treatments
Some patients will not benefit from pharmacological treatments because of lack of efficacy (eg, 41% experienced no effect from duloxetine) and drug interactions.139–144 For these patients, non-drug treatment options are sorely needed. Two systematic literature reviews reveal that several natural products and complementary therapy interventions have been tested for CIPN.12,145 Based on the findings of these systematic reviews, the following treatments cannot be recommended to treat painful CIPN: vitamin E, glutamate, goshajinkigan, acetyl-L-carnitine, alpha-lipoic acid, and omega-3 fatty acids. Studies of these products either were significantly biased because of small sample sizes and poor outcome measures, or showed detrimental (in the case of acetyl-L-carnitine) or insignificant effects on CIPN.12,145 Furthermore, pain was not the primary outcome in any of the studies.
Several experimental and quasi-experimental studies have tested complementary treatments for painful CIPN, such as acupuncture 108,109,146 electrocutaneous nerve stimulation (scrambler therapy)147–149, and exercise 150–152. Results from studies of acupuncture are mixed, and only quasi-experimental scrambler studies provide preliminary evidence of efficacy. Although exercise interventions have been tested to reduce CIPN, their effects on painful CIPN as the primary outcome have not been adequately explored. Given the threats to the external and internal validity of these studies—small samples, lack of blinded placebo control comparisons, and high drop-out rates—none of these complementary treatments can be recommended to treat CIPN pain.
Cognitive behavioral therapy (CBT), a complementary therapy with demonstrated efficacy for chronic pain conditions (eg, fibromyalgia, migraine headaches, arthritis)153 and cancer pain,154–156 often leads to larger improvements in pain-related outcomes than does pharmacologic therapy.157 The CBT approach involves educational strategies that teach about causes and treatment of pain, modify inaccurate beliefs about pain control, and provide practical approaches for improving problem-solving and coping. One small randomized wait-list controlled pilot study provides preliminary evidence that CBT—specifically, a self-guided online cognitive and behaviorally based pain management intervention —may be effective to reduce chronic CIPN pain severity.158 Patients (N = 60) with chronic CIPN pain rated as ≥ 4 on a 0–10 numeric rating scale were randomized either to an 8-week Web-based CBT intervention or a wait-list control. The Web-based CBT program included self-guided modules about late effects, patient/provider communication, exercise, sleep, goal setting, activity pacing, strategies to encourage participation in enjoyable activities, and peripheral neuropathy management. Individuals who received the CBT intervention had significantly greater improvements in averaged 7-day pain scores when compared with the wait-list controls (P = .046, d = .58). The main study limitations were lack of blinding and high attrition (22%). Despite these limitations, the positive findings justify the need for a larger, placebo-controlled trial of CBT for painful CIPN.
Preclinical studies of CIPN preventive interventions
Although no evidence has emerged from well-designed randomized controlled trials that anything prevents CIPN, painful or otherwise, several preclinical studies provide information that could lead to new treatments in the future. Researchers at the University of Michigan explored the effect of duloxetine to prevent oxaliplatin-induced hyperalgesia in male and female rats. When compared with vehicle/water-treated animals, duloxetine-treated animals demonstrated less hyperalgesia following 5 days of oxaliplatin treatment (unpublished). These preliminary findings provide the foundation for a pending multi-site National Cancer Institute-funded Phase 2/3 trial testing the comparative effectiveness of 30 mg or 60 mg daily duloxetine versus placebo as prevention for painful and nonpainful CIPN.
Several preclinical studies also suggest potential benefits of other agents, such as enzymes, nutritional supplements, opioid agonists, and neuropeptides, for painful CIPN. One exciting new approach to preventing painful CIPN involves the use of poly ADP-ribose polymerase (PARP) inhibitors. PARP—a nuclear enzyme found in nerve cells—plays a role in DNA damage. Although the precise mechanisms of action are not fully understood, PARP inhibitors can turn off DNA-damaging processes, such as oxidative stress, and thus might prevent painful neuropathy caused by neurotoxic drugs.159 This premise has been supported in a preclinical study of vincristine-induced neuropathy: three different PARP inhibiters attenuated mechanical allodynia in vincristine-treated male mice.160 Studies have also evaluated another enzyme, histone deacetylase 6 (HDAC6), a microtubule-associated deacetylase that is involved in α-tubulin-dependent intracellular mitochondrial transport in sensory nerves.161 Because HDAC6 inhibitors improve nerve function in animals with peripheral neuropathy caused by Charcot-Marie-Tooth disease,162 these novel agents might help to prevent CIPN. This idea is supported by a recent study showing that a HDAC6 inhibitor, ACY-1083, was more effective than a control vehicle to mitigate mechanical allodynia, spontaneous pain, and numbness in cisplatin-treated male rodents.161 Flavonoids—compounds found in fruits and vegetables—have been tested in animal models of oxaliplatin- and cisplatin-induced neuropathy.163,164 In a recent study, several dose levels of the flavonoid 6-Methoxyflavone (6-MF) were compared with a control vehicle and gabapentin.164 After receiving 4 weekly cisplatin injections, 6-MF–treated male rats demonstrated significantly less cisplatin-induced mechanical allodynia than animals receiving the control vehicle. While the antinociceptive effects of the highest 6-MF dose and gabapentin were similar, only gabapentin caused motor dysfunction. Another preclinical study showed that cebranopadol, an opioid receptor agonist that binds to many different types of opioid receptors in the peripheral and CNS, decreased cold-induced allodynia in male mice following oxaliplatin administration.165 Orexin A, an endogenous neuropeptide that facilitates descending pain modulation in the CNS, was compared with duloxetine and a control vehicle in another preclinical study of oxaliplatin-induced neuropathy.68 One group of male mice was injected with Orexin A into the cerebral ventricles; another group of male mice was injected with duloxetine into the peritoneum. Mechanical allodynia tests suggested that orexin may be more effective than duloxetine in preventing oxaliplatin-induced painful neuropathy. These highlighted studies, and many others not described here, provide hope for new treatments that might prevent or attenuate painful CIPN for millions of cancer survivors.
In conclusion, limited high-level evidence supports the use of anything other than duloxetine for the treatment of established painful CIPN, and numerous clinical trials have failed to identify effective preventive treatments. However, preclinical studies provide evidence for promising new agents that may be examined in future clinical studies. Rigorous placebo-controlled studies of natural products and complementary interventions are also needed. Our understanding of the pathophysiological mechanisms underlying painful CIPN is limited but expanding. As our knowledge of CIPN pain pathophysiology increases, future mechanism-targeted interventions will emerge.
Conclusion
Impact and implications for the nursing practice
Distressing CIPN often leads to reductions in potentially life-saving chemotherapy treatment. Most patients receive inadequate education about CIPN before initiating chemotherapy and have difficulty describing their symptoms when they manifest. 4,82,166 Additionally, clinicians often lack the time, knowledge, and resources for adequate assessment of CIPN.167 Although individuals may adjust to CIPN, it might never resolve. While no curative treatments for CIPN are known, several nursing and pharmacologic interventions may help to manage CIPN and lessen resulting functional limitations. Duloxetine is the only drug that is recommended for the palliative treatment of chronic painful CIPN. However, treatments (eg, nortriptyline, gabapentin, amitriptyline) for other chronic pain conditions that exhibit chronic painful CIPN-like manifestations and pathophysiological mechanisms may also be recommended. Further research will help to identify which of these chronic pain treatments are effective for chronic painful CIPN.
Ultimately, nurses play a vital role in assessing for early signs and existing manifestations of CIPN, providing CIPN education, and assisting patients with CIPN management. Nurses can advocate and suggest referrals to CIPN and co-occurring symptom management resources for patients. Finally, nurses can provide emotional support to individuals who are experiencing the scary, painful, isolating, and functionally debilitating manifestations of CIPN.
Future directions in research
Further research is needed to explore the specific mechanisms and predictors of chronic painful CIPN and to evaluate interventions to prevent and treat each type of CIPN. Several classes of neurotoxic agents may induce CIPN through varying mechanisms; thus, the intervention must be tailored to the type of CIPN. Many of the more than 75 trials that have tested CIPN interventions were limited by poor CIPN measurement and lack of intervention specificity to the type of CIPN studied. Most importantly, few studies have focused specifically on interventions for chronic painful CIPN. Additional studies of nonpharmacologic interventions and preclinical studies of pharmacologic agents and supplements are needed to inform larger and more rigorous clinical research testing mechanism-specific interventions for chronic painful CIPN.
Acknowledgments
Funding: This work was supported by the Rita & Alex Hillman Foundation Predoctoral Fellowship (to G.A.K.-L.); American Cancer Society Denny Hoelzer Sentinel Technologies Doctoral Scholarship in Cancer Nursing (grant no. DSCN-17-082-01-SCN); and the National Institute of Nursing Research, T32 (grant no. NR016914).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflicts of interest: Robert Knoerl reports a consulting honorarium from System Analytic. The other authors have no conflicts of interest to disclose.
References
- 1.Wang XS, Shi Q, Dougherty PM, et al. Prechemotherapy touch sensation deficits predict oxaliplatin-induced neuropathy in patients with colorectal cancer. Oncology. 2016;90:127–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Cavaletti G, Jakubowiak AJ. Peripheral neuropathy during bortezomib treatment of multiple myeloma: a review of recent studies. Leuk Lymphoma. 2010;51:1178–1187. [DOI] [PubMed] [Google Scholar]
- 3.Argyriou AA, Cavaletti G, Bruna J, Kyritsis AP, Kalofonos HP. Bortezomib-induced peripheral neurotoxicity: An update. Arch Toxicol. 2014;88:1669–1679. [DOI] [PubMed] [Google Scholar]
- 4.Bakitas MA. Background noise: the experience of chemotherapy-induced peripheral neuropathy. Nurs Res. 2007;56:323–331. [DOI] [PubMed] [Google Scholar]
- 5.Monfort SM, Pan X, Patrick R, et al. Natural history of postural instability in breast cancer patients treated with taxane-based chemotherapy: a pilot study. Gait Posture. 2016;48:237–242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Speck RM, DeMichele A, Farrar JT, et al. Scope of symptoms and self-management strategies for chemotherapy-induced peripheral neuropathy in breast cancer patients. Support Care Cancer. 2012;20:2433–2439. [DOI] [PubMed] [Google Scholar]
- 7.Tofthagen CS. Surviving chemotherapy for colon cancer and living with the consequences. J Palliat Med. 2010;13:1389–1391. [DOI] [PubMed] [Google Scholar]
- 8.Tofthagen CS. Patient perceptions associated with chemotherapy-induced peripheral neuropathy. Clin J Oncol Nurs. 2010;14:E22–8. [DOI] [PubMed] [Google Scholar]
- 9.Exposito Vizcaino S, Casanova-Molla J, Escoda L, Galan S, Miro J. Neuropathic pain in cancer patients treated with bortezomib. Neurologia. 2018;33:28–34. [DOI] [PubMed] [Google Scholar]
- 10.Tofthagen CS, McAllister RD, McMillan SC. Peripheral neuropathy in patients with colorectal cancer receiving oxaliplatin. Clin J Oncol Nurs. 2011;15:182–188. [DOI] [PubMed] [Google Scholar]
- 11.Zanville NR, Nudelman KNH, Smith DJ, et al. Evaluating the impact of chemotherapy-induced peripheral neuropathy symptoms (CIPN-sx) on perceived ability to work in breast cancer survivors during the first year post-treatment. Support Care Cancer. 2016;24:4779–4789. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.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;32:1941–1967. [DOI] [PubMed] [Google Scholar]
- 13.Speck RM, Sammel MD, Farrar JT, et al. Impact of chemotherapy-induced peripheral neuropathy on treatment delivery in nonmetastatic breast cancer. J Oncol Pract. 2013;9:e234–240. [DOI] [PubMed] [Google Scholar]
- 14.Ventzel L, Jensen AB, Jensen AR, Jensen TS, Finnerup NB. Chemotherapy-induced pain and neuropathy: a prospective study in patients treated with adjuvant oxaliplatin or docetaxel. Pain. 2016;157:560–568. [DOI] [PubMed] [Google Scholar]
- 15.Beijers AJM, Mols F, Tjan-Heijnen VCG, Faber CG, van de Poll-Franse L V, Vreugdenhil G. Peripheral neuropathy in colorectal cancer survivors: the influence of oxaliplatin administration. Results from the population-based PROFILES registry. Acta Oncol (Madr). 2015;54:463–469. [DOI] [PubMed] [Google Scholar]
- 16.Liu D, Fischer C, Petriccione M, Goldman D, Riedel E, De Braganca K. Assessing the relationship between vincristine use and outcomes in a retrospective cohort of pediatric patients with average risk medulloblastomas. Neuro Oncol. 2016;18(suppl 6):vi161. [Google Scholar]
- 17.Finan PH, Smith MT. The comorbidity of insomnia, chronic pain, and depression: dopamine as a putative mechanism. Sleep Med Rev. 2013;17:173–183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Knoerl R, Chornoby Z, Smith EML. Estimating the frequency, severity, and clustering of SPADE symptoms in chronic painful chemotherapy-induced peripheral neuropathy. Pain Manag Nurs. 2018;19:354–365. [DOI] [PubMed] [Google Scholar]
- 19.Nijs J, Loggia ML, Polli A, et al. Sleep disturbances and severe stress as glial activators: key targets for treating central sensitization in chronic pain patients? Expert Opin Ther Targets. 2017;21:817–826. [DOI] [PubMed] [Google Scholar]
- 20.Pachman DR, Barton DL, Swetz KM, Loprinzi CL. Troublesome symptoms in cancer survivors: fatigue, insomnia, neuropathy, and pain. J Clin Oncol. 2012;30:3687–3696. [DOI] [PubMed] [Google Scholar]
- 21.Sachs-Ericsson NJ, Sheffler JL, Stanley IH, Piazza JR, Preacher KJ. When emotional pain becomes physical: adverse childhood experiences, pain, and the role of mood and anxiety disorders. J Clin Psychol. 2017;73:1403–1428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Schneider BP, Hershman DL, Loprinzi C. Symptoms: chemotherapy-induced peripheral neuropathy. Adv Exp Med Biol. 2015;862:77–87. [DOI] [PubMed] [Google Scholar]
- 23.Jaggi AS, Singh N. Mechanisms in cancer-chemotherapeutic drugs-induced peripheral neuropathy. Toxicology. 2012;291:1–9. [DOI] [PubMed] [Google Scholar]
- 24.Boyette-Davis JA, Walters ET, Dougherty PM. Mechanisms involved in the development of chemotherapy-induced neuropathy. Pain Manag. 2015;5:285–296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Staff NP, Grisold A, Grisold W, Windebank AJ. Chemotherapy-induced peripheral neuropathy: a current review. Ann Neurol. 2017;81:772–781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Carozzi VA, Canta A, Chiorazzi A. Chemotherapy-induced peripheral neuropathy: what do we know about mechanisms? Neurosci Lett. 2015;596:90–107. [DOI] [PubMed] [Google Scholar]
- 27.Fukuda Y, Li Y, Segal RA. A mechanistic understanding of axon degeneration in chemotherapy-induced peripheral neuropathy. Front Neurosci. 2017;11:481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Starobova H, Vetter I. Pathophysiology of chemotherapy-induced peripheral neuropathy. Front Mol Neurosci. 2017;10:174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kandula T, Park SB, Cohn RJ, Krishnan AV, Farrar MA. Pediatric chemotherapy induced peripheral neuropathy: a systematic review of current knowledge. Cancer Treat Rev. 2016;50:118–128. [DOI] [PubMed] [Google Scholar]
- 30.Park SB, Goldstein D, Krishnan AV, et al. Chemotherapy-induced peripheral neurotoxicity: a critical analysis. Cancer J Clin. 2013;63:419–437. [DOI] [PubMed] [Google Scholar]
- 31.Kerckhove N, Collin A, Conde S, Chaleteix C, Pezet D, Balayssac D. Long-term effects, pathophysiological mechanisms, and risk factors of chemotherapy-induced peripheral neuropathies: a comprehensive literature review. Front Pharmacol. 2017;8:86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Badros A, Goloubeva O, Dalal JS, et al. Neurotoxicity of bortezomib therapy in multiple myeloma: a single-center experience and review of the literature. Cancer. 2007;110:1042–1049. [DOI] [PubMed] [Google Scholar]
- 33.Broyl A, Corthals SL, Jongen JL, et al. Mechanisms of peripheral neuropathy associated with bortezomib and vincristine in patients with newly diagnosed multiple myeloma: a prospective analysis of data from the HOVON-65/GMMG-HD4 trial. Lancet Oncol. 2010;11:1057–1065. [DOI] [PubMed] [Google Scholar]
- 34.Kropff M, Vogel M, Kreter A, et al. Bortezomib and low-dose dexamethasone with or without continuous low-dose oral cyclophosphamide for primary refractory or relapsed multiple myeloma: final results of a national multicenter randomized controlled phase III study. Blood. 2017;96:1857–1866 [DOI] [PubMed] [Google Scholar]
- 35.Attal N, Bouhassira D, Gautron M, et al. Thermal hyperalgesia as a marker of oxaliplatin neurotoxicity: a prospective quantified sensory assessment study. Pain. 2009;144:245–252. [DOI] [PubMed] [Google Scholar]
- 36.Brozou V, Vadalouca A, Zis P. Pain in platin-induced neuropathies: a systematic review and meta-analysis. Pain Ther. 2018;7:105–119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Postma TJ, Benard BA, Huijgens PC, Ossenkoppele GJ, Heimans JJ. Long-term effects of vincristine on the peripheral nervous system. J Neurooncol. 1993;15:23–27. [DOI] [PubMed] [Google Scholar]
- 38.Casey EB, Jellife AM, Le Quesne PM, Millett YL. Vincristine neuropathy. Clinical and electrophysiological observations. Brain. 1973;96:69–86. [DOI] [PubMed] [Google Scholar]
- 39.Anghelescu DL, Faughnan LG, Jeha S, et al. Neuropathic pain during treatment for childhood acute lymphoblastic leukemia. Pediatr Blood Cancer. 2011;57:1147–1153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Lavoie Smith EM, Li L, Chiang C, et al. Patterns and severity of vincristine-induced peripheral neuropathy in children with acute lymphoblastic leukemia. J Peripher Nerv Syst. 2015;20:37–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Lieber S, Blankenburg M, Apel K, et al. Small-fiber neuropathy and pain sensitization in survivors of pediatric acute lymphoblastic leukemia. Eur J Paediatr Neurol. 2018;22:457–469. [DOI] [PubMed] [Google Scholar]
- 42.Glasmacher A, Hahn C, Hoffmann F, et al. A systematic review of phase-II trials of thalidomide monotherapy in patients with relapsed or refractory multiple myeloma. Br J Haematol. 2006;132:584–593. [DOI] [PubMed] [Google Scholar]
- 43.Cavaletti G, Beronio A, Reni L, et al. Thalidomide sensory neurotoxicity: a clinical and neurophysiologic study. Neurology. 2004;62:2291–2293. [DOI] [PubMed] [Google Scholar]
- 44.Briani C, Zara G, Rondinone R, et al. Thalidomide neurotoxicity: prospective study in patients with lupus erythematosus. Neurology. 2004;62:2288–2290. [DOI] [PubMed] [Google Scholar]
- 45.Pachman DR, Qin R, Seisler DK, et al. Clinical course of oxaliplatin-induced neuropathy: results from the randomized phase III trial N08CB (Alliance). J Clin Oncol. 2015;33:3416–3422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Richardson PG, Briemberg H, Jagannath S, et al. Frequency, characteristics, and reversibility of peripheral neuropathy during treatment of advanced multiple myeloma with bortezomib. J Clin Oncol. 2006;24:3113–3120. [DOI] [PubMed] [Google Scholar]
- 47.Wolf SL, Barton DL, Qin R, et al. The relationship between numbness, tingling, and shooting/burning pain in patients with chemotherapy-induced peripheral neuropathy (CIPN) as measured by the EORTC QLQ-CIPN20 instrument, N06CA. Support Care Cancer. 2012;20:625–632. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Quasthoff S, Hartung HP. Chemotherapy-induced peripheral neuropathy. J Neurol. 2002;249:9–17. [DOI] [PubMed] [Google Scholar]
- 49.Pachman DR, Qin R, Seisler D, et al. Comparison of oxaliplatin and paclitaxel-induced neuropathy (Alliance A151505). Support Care Cancer. 2016;24:5059–5068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Verstappen CCP, Koeppen S, Heimans JJ, et al. Dose-related vincristine-induced peripheral neuropathy with unexpected off-therapy worsening. Neurology. 2005;64:1076–1077. [DOI] [PubMed] [Google Scholar]
- 51.Toyama S, Shimoyama N, Ishida Y, Koyasu T, Szeto HH, Shimoyama M. Characterization of acute and chronic neuropathies induced by oxaliplatin in mice and differential effects of a novel mitochondria-targeted antioxidant on the neuropathies. Anesthesiology. 2014;120:459–473. [DOI] [PubMed] [Google Scholar]
- 52.Kiya T, Kawamata T, Namiki A, Yamakage M. Role of satellite cell-derived L-serine in the dorsal root ganglion in paclitaxel-induced painful peripheral neuropathy. Neuroscience. 2011;174:190–199. [DOI] [PubMed] [Google Scholar]
- 53.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;49:2557–2563. [DOI] [PubMed] [Google Scholar]
- 54.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Carozzi VA, Renn CL, Bardini M, et al. Bortezomib-induced painful peripheral neuropathy: An electrophysiological, behavioral, morphological and mechanistic study in the mouse. PLoS One. 2013;8:e72995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Quartu M, Carozzi VA, Dorsey SG, et al. Bortezomib treatment produces nocifensive behavior and changes in the expression of TRPV1, CGRP, and substance P in the rat DRG, spinal cord, and sciatic nerve. Biomed Res Int. 2014;2014:180428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Renn CL, Carozzi VA, Rhee P, Gallop D, Dorsey SG, Cavaletti G. Multimodal assessment of painful peripheral neuropathy induced by chronic oxaliplatin-based chemotherapy in mice. Mol Pain. 2011;7:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.International Association for the Study of Pain. IASP Terminology. 2014; Available at: https://www.iasp-pain.org/Education/Content.aspx?ItemNumber=1698#Sensitization. (Accessed December 1, 2018).
- 59.Baron R Neuropathic pain: a clinical perspective. Handb Exp Pharmacol. 2009;:3–30. [DOI] [PubMed] [Google Scholar]
- 60.Gilron I, Baron R, Jensen T. Neuropathic pain: principles of diagnosis and treatment. Mayo Clin Proc. 2015;90:532–545. [DOI] [PubMed] [Google Scholar]
- 61.Norcini M, Vivoli E, Galeotti N, Bianchi E, Bartolini A, Ghelardini C. Supraspinal role of protein kinase C in oxaliplatin-induced neuropathy in rat. Pain. 2009;146:141–147. [DOI] [PubMed] [Google Scholar]
- 62.Cata JP, Weng HR, Chen JH, Dougherty PM. Altered discharges of spinal wide dynamic range neurons and down-regulation of glutamate transporter expression in rats with paclitaxel-induced hyperalgesia. Neuroscience. 2006;138:329–338. [DOI] [PubMed] [Google Scholar]
- 63.Peddi PF, Peddi S, Santos ES, Morgensztern D. Central nervous system toxicities of chemotherapeutic agents. Expert Rev Anticancer Ther. 2014;14:857–863. [DOI] [PubMed] [Google Scholar]
- 64.Peters CM, Jimenez-Andrade JM, Kuskowski MA, Ghilardi JR, Mantyh PW. An evolving cellular pathology occurs in dorsal root ganglia, peripheral nerve and spinal cord following intravenous administration of paclitaxel in the rat. Brain Res. 2007;1168:46–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Kozachik SL, Page GG. A hyperresponsive HPA axis may confer resilience against persistent paclitaxel-induced mechanical hypersensitivity. Biol Res Nurs. 2016;18:290–298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Holden JE, Wagner MA, Reeves BL. Anatomical evidence for lateral hypothalamic innervation of the pontine A7 catecholamine cell group in rat. Neurosci Lett. 2018;668:80–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Wardach J, Wagner M, Jeong Y, Holden JE. Lateral hypothalamic stimulation reduces hyperalgesia through spinally descending Orexin-A neurons in neuropathic pain. West J Nurs Res. 2016;38:292–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Toyama S, Shimoyama N, Shimoyama M. The analgesic effect of orexin-A in a murine model of chemotherapy-induced neuropathic pain. Neuropeptides. 2017;61:95–100. [DOI] [PubMed] [Google Scholar]
- 69.Reyes-Gibby CC, Morrow PK, Buzdar A, Shete S. Chemotherapy-induced peripheral neuropathy as a predictor of neuropathic pain in breast cancer patients previously treated with paclitaxel. J Pain. 2009;10:1146–1150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Miaskowski C, Mastick J, Paul SM, et al. Chemotherapy-induced neuropathy in cancer survivors. J Pain Symptom Manage. 2017;54:204–218.e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Poulin PA, Romanow HC, Rahbari N, et al. The relationship between mindfulness, pain intensity, pain catastrophizing, depression, and quality of life among cancer survivors living with chronic neuropathic pain. Support Care Cancer. 2016;24:4167–4175. [DOI] [PubMed] [Google Scholar]
- 72.Bulls HW, Hoogland AI, Kennedy B, et al. A longitudinal examination of associations between age and chemotherapy-induced peripheral neuropathy in patients with gynecologic cancer. Gynecol Oncol. 2019;152:310–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Ventzel L, Madsen CS, Karlsson P, et al. Chronic pain and neuropathy following adjuvant chemotherapy. Pain Med. 2018;19:1813–1824. [DOI] [PubMed] [Google Scholar]
- 74.Davis LL, Kroenke K, Monahan P, Kean J, Stump TE. The SPADE symptom cluster in primary care patients with chronic pain. Clin J Pain. 2016;32:388–393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Dueñas M, Ojeda B, Salazar A, Mico JA, Failde I. A review of chronic pain impact on patients, their social environment and the health care system. J Pain Res. 2016;9:457–467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Edwards RR, Dworkin RH, Sullivan MD, Turk DC, Wasan AD. The role of psychosocial processes in the development and maintenance of chronic pain. J Pain. 2016;17:T70–T92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Smith EML, Pang H, Ye C, et al. Predictors of duloxetine response in patients with oxaliplatin-induced painful chemotherapy-induced peripheral neuropathy (CIPN): a secondary analysis of randomised controlled trial - CALGB/alliance 170601. Eur J Cancer Care (Engl). 2017;26:e12421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Cella D, Yount S, Rothrock N, et al. The Patient-Reported Outcomes Measurement Information System (PROMIS): progress of an NIH Roadmap cooperative group during its first two years. Med Care. 2007;45(Suppl 1):S3–S11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Gilchrist LS, Tanner L, Hooke MC. Measuring chemotherapy-induced peripheral neuropathy in children: Development of the Ped-mTNS and pilot study results. Rehabil Oncol. 2009;27 :7–15. [Google Scholar]
- 80.Lavoie Smith EM, Li L, Hutchinson RJ, et al. Measuring vincristine-induced peripheral neuropathy in children with acute lymphoblastic leukemia. Cancer Nurs. 2013;36:E49–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Smith EML, 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;309:1359–1367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Smith EML, Beck SL, Cohen J. The Total Neuropathy Score: a tool for measuring chemotherapy-induced peripheral neuropathy. Oncol Nurs Forum. 2008;35:96–102. [DOI] [PubMed] [Google Scholar]
- 83.Landmann G, Berger MF, Stockinger L, Opsommer E. Quantitative sensory testing: a comprehensive protocol for clinical trials. Spinal Cord. 2017;55:575–582. [DOI] [PubMed] [Google Scholar]
- 84.Rolke R, Magerl W, Campbell KA, et al. Quantitative sensory testing: a comprehensive protocol for clinical trials. Eur J Pain. 2006;10:77–88. [DOI] [PubMed] [Google Scholar]
- 85.Addington J, Freimer M. Chemotherapy-induced peripheral neuropathy: an update on the current understanding. F1000Research. 2016;5:eCollection 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Boyette-Davis JA, Cata JP, Driver LC, et al. Persistent chemoneuropathy in patients receiving the plant alkaloids paclitaxel and vincristine. Cancer Chemother Pharmacol. 2013;71:619–626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Siau C, Xiao W, Bennett GJ. Paclitaxel- and vincristine-evoked painful peripheral neuropathies: loss of epidermal innervation and activation of Langerhans cells. Exp Neurol. 2006;201:507–514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Leonard GD, Wright MA, Quinn MG, et al. Survey of oxaliplatin-associated neurotoxicity using an interview-based questionnaire in patients with metastatic colorectal cancer. BMC Cancer. 2005;5:116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.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;41:1135–1139. [DOI] [PubMed] [Google Scholar]
- 90.Smith EML, Knoerl R, Yang JJ, Kanzawa-Lee G, Lee D, Bridges CM. In search of a gold standard patient-reported outcome measure for use in chemotherapy-induced peripheral neuropathy clinical trials. Cancer Control. 2018;25:1073274818756608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Lavoie Smith EM, Cohen JA, Pett MA, Beck SL. The validity of neuropathy and neuropathic pain measures in patients with cancer receiving taxanes and platinums. Oncol Nurs Forum. 2011;38:133–142. [DOI] [PubMed] [Google Scholar]
- 92.Oldenburg J, Fosså SD, Dahl AA. Scale for chemotherapy-induced long-term neurotoxicity (SCIN): psychometrics, validation, and findings in a large sample of testicular cancer survivors. Qual Life Res. 2006;15:791–800. [DOI] [PubMed] [Google Scholar]
- 93.Cella D, Peterman A, Hudgens S, Webster K, Socinski MA. Measuring the side effects of taxane therapy in oncology: the Functional Assesment of Cancer Therapy-taxane (FACT-taxane). Cancer. 2003;98:822–831. [DOI] [PubMed] [Google Scholar]
- 94.Mendoza TR, Wang XS, Williams LA, et al. Measuring therapy-induced peripheral neuropathy: preliminary development and validation of the Treatment-Induced Neuropathy Assessment Scale. J Pain. 2015;16:1032–1043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.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;17:1483–1491. [DOI] [PubMed] [Google Scholar]
- 96.Cavaletti G, Bogliun G, Marzorati L, et al. Grading of chemotherapy-induced peripheral neurotoxicity using the Total Neuropathy Scale. Neurology. 2003;61:1297–1300. [DOI] [PubMed] [Google Scholar]
- 97.Cavaletti G, Jann S, Pace A, et al. Multi-center assessment of the Total Neuropathy Score for chemotherapy-induced peripheral neurotoxicity. J Peripher Nerv Syst. 2006;11:135–141. [DOI] [PubMed] [Google Scholar]
- 98.Wampler MA, Miaskowski C, Byl N, Rugo H, Topp KS. The modified Total Neuropathy Score: a clinically feasible and valid measure of taxane-induced peripheral neuropathy in women with breast cancer. J Support Oncol. 2006;4:W9–W16. [Google Scholar]
- 99.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;12:210–215. [DOI] [PubMed] [Google Scholar]
- 100.Smith EML, Cohen JA, Pett MA, Beck SL. 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;33:173–183. [DOI] [PubMed] [Google Scholar]
- 101.Dueck AC, Mendoza TR, Mitchell SA, et al. Validity and reliability of the US National Cancer Institute’s patient-reported outcomes version of the Common Terminology Criteria for Adverse Events (PRO-CTCAE). JAMA Oncol. 2015;1:1051–1059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Griffiths C, Kwon N, Beaumont JL, Paice JA. Cold therapy to prevent paclitaxel-induced peripheral neuropathy. Support Care Cancer. 2018;26:3461–3469. [DOI] [PubMed] [Google Scholar]
- 103.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;15:178–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Schonsteiner SS, Bauder Missbach H, Benner A, et al. A randomized exploratory phase 2 study in patients with chemotherapy-related peripheral neuropathy evaluating whole-body vibration training as adjunct to an integrated program including massage, passive mobilization and physical exercises. Exp Hematol Oncol. 2017;6:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Rao R, Flynn P, Sloan J, et al. Efficacy of lamotrigine in the management of chemotherapy-induced peripheral neuropathy: a phase 3 randomized, double-blind, placebo-controlled trial, N01C3. Cancer. 2008;112:2802–2808. [DOI] [PubMed] [Google Scholar]
- 106.Rao RD, Michalak JC, Sloan JA, et al. Efficacy of gabapentin in the management of chemotherapy-induced peripheral neuropathy: A phase 3 randomized, double-blind, placebo-controlled, crossover trial (N00C3). Cancer. 2007;110:2110–2118. [DOI] [PubMed] [Google Scholar]
- 107.Prinsloo S, Novy D, Driver L, et al. Randomized controlled trial of neurofeedback on chemotherapy-induced peripheral neuropathy: a pilot study. Cancer. 2017;123:1989–bortezomib-induced1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Greenlee H, Crew KD, Capodice J, et al. Randomized sham-controlled pilot trial of weekly electro-acupuncture for the prevention of taxane-induced peripheral neuropathy in women with early stage breast cancer. Breast Cancer Res Treat. 2016;156:453–464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Garcia MK, Cohen L, Guo Y, et al. Electroacupuncture for thalidomide/bortezomib-induced peripheral neuropathy in multiple myeloma: a feasibility study. J Hematol Oncol. 2014;7:41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.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;32:1941–1967. [DOI] [PubMed] [Google Scholar]
- 111.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;19:833–841. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Gewandter JS, Mohile SG, Heckler CE, et al. A phase III randomized, placebo-controlled study of topical amitriptyline and ketamine for chemotherapy-induced peripheral neuropathy (CIPN): A University of Rochester CCOP study of 462 cancer survivors. Support Care Cancer. 2014;22:1807–1814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Hammack JE, Michalak JC, Loprinzi CL, et al. Phase III evaluation of nortriptyline for alleviation of symptoms of cis-platinum-induced peripheral neuropathy. Pain. 2002;98:195–203. [DOI] [PubMed] [Google Scholar]
- 114.Kautio AL, Haanpaa M, Leminen A, Kalso E, Kautiainen H, Saarto T. Amitriptyline in the prevention of chemotherapy-induced neuropathic symptoms. Anticancer Res. 2009;29:2601–2606. [PubMed] [Google Scholar]
- 115.Gewandter JS, Dworkin RH, Finnerup NB, Mohile NA. Painful chemotherapy-induced peripheral neuropathy: lack of treatment efficacy or the wrong clinical trial methodology? Pain. 2017;158:30–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Dugan SE, Fuller MA. Duloxetine: A dual reuptake inhibitor. Ann Pharmacother. 2004;38:2078–2085. [DOI] [PubMed] [Google Scholar]
- 117.Delgado PL. Common pathways of depression and pain. J Clin Psychiatry. 2004;65(Suppl 12): 16–19. [PubMed] [Google Scholar]
- 118.Mochizucki D Serotonin and noradrenaline reuptake inhibitors in animal models of pain. Hum Psychopharmacol. 2004;19(Suppl 1):S15–19. [DOI] [PubMed] [Google Scholar]
- 119.Willis WD, Westlund KN. Neuroanatomy of the pain system and of the pathways that modulate pain. J Clin Neurophysiol. 1997;14:2–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Bymaster FP, Dreshfield-Ahmad LJ, Threlkeld PG, et al. Comparative affinity of duloxetine and venlafaxine for serotonin and norepinephrine transporters in vitro and in vivo, human serotonin receptor subtypes, and other neuronal receptors. Neuropsychopharmacology. 2001;25:871–880. [DOI] [PubMed] [Google Scholar]
- 121.Wang C-F, Russell G, Strichartz GR, Wang G-K. The local and systemic actions of duloxetine in allodynia and hyperalgesia using a rat skin incision pain model. Anesth Analg. 2015;121:532–544. [DOI] [PubMed] [Google Scholar]
- 122.Wang S-Y, Calderon J, Kuo Wang G. Block of neuronal Na+ channels by antidepressant duloxetine in a state-dependent manner. Anesthesiology. 2010;113:655–665. [DOI] [PubMed] [Google Scholar]
- 123.Majithia N, Temkin SM, Ruddy KJ, Beutler AS, Hershman DL, Loprinzi CL. National Cancer Institute-supported chemotherapy-induced peripheral neuropathy trials: outcomes and lessons. Support Care Cancer. 2016;24:1439–1447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Pachman DR, Watson JC, Lustberg MB, et al. Management options for established chemotherapy-induced peripheral neuropathy. Support Care Cancer. 2014;22:2281–2295. [DOI] [PubMed] [Google Scholar]
- 125.Dworkin RH, Backonja M, Rowbotham MC, et al. Advances in neuropathic pain: diagnosis, mechanisms, and treatment recommendations. Arch Neurol. 2003;60:1524–1534. [DOI] [PubMed] [Google Scholar]
- 126.Dworkin RH, O’Connor AB, Audette J, et al. Recommendations for the pharmacological management of neuropathic pain: an overview and literature update. Mayo Clin Proc. 2010;85(Suppl 3):S3–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Patil PR, Wolfe J, Said Q, Thomas J, Martin BC. Opioid use in the management of diabetic peripheral neuropathy (dpn) in a large commercially insured population. Clin J Pain. 2015;31:414–424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Gomes T, Tadrous M, Mamdani MM, Paterson JM, Juurlink DN. The burden of opioid-related mortality in the United States. JAMA Netw Open. 2018;1:e180217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Roeckel LA, Le Coz GM, Gavériaux-Ruff C, Simonin F. Opioid-induced hyperalgesia: cellular and molecular mechanisms. Neuroscience. 2016;338:160–182. [DOI] [PubMed] [Google Scholar]
- 130.Abrams DI, Jay CA, Shade SB, et al. Cannabis in painful HIV-associated sensory neuropathy: a randomized placebo-controlled trial. Neurology. 2007;68:515–521. [DOI] [PubMed] [Google Scholar]
- 131.Ellis RJ, Toperoff W, Vaida F, et al. Smoked medicinal cannabis for neuropathic pain in HIV: a randomized, crossover clinical trial. Neuropsychopharmacology. 2009;34:672–680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Hill KP. Medical marijuana for treatment of chronic pain and other medical and psychiatric problems: a clinical review. JAMA. 2015;313:2474–2483. [DOI] [PubMed] [Google Scholar]
- 133.Lee G, Grovey B, Furnish T, Wallace M. Medical cannabis for neuropathic pain. Curr Pain Headache Rep. 2018;22:8. [DOI] [PubMed] [Google Scholar]
- 134.Wallace MS, Marcotte TD, Umlauf A, Gouaux B, Atkinson JH. Efficacy of inhaled cannabis on painful diabetic neuropathy. J Pain. 2015;16:616–627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Wilsey B, Marcotte T, Tsodikov A, et al. A randomized, placebo-controlled, crossover trial of cannabis cigarettes in neuropathic pain. J Pain. 2008;9:506–521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Ware MA, Wang T, Shapiro S, et al. Smoked cannabis for chronic neuropathic pain: a randomized controlled trial. CMAJ. 2010;182:E694–701. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Filipczak-Bryniarska I, Krzyzewski RM, Kucharz J, et al. High-dose 8% capsaicin patch in treatment of chemotherapy-induced peripheral neuropathy: single-center experience. Med Oncol. 2017;34:1–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Heuvel SASV Den, Wal SEIV Wal Smedes LA, et al. Intravenous lidocaine: old-school drug, new purpose-reduction of intractable pain in patients with chemotherapy induced peripheral neuropathy. Pain Res Manag. 2017;2017:8053474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Caraci F, Crupi R, Drago F, Spina E. Metabolic drug interactions between antidepressants and anticancer drugs: focus on selective serotonin reuptake inhibitors and hypericum extract. Curr Drug Metab. 2011;12:570–577. [DOI] [PubMed] [Google Scholar]
- 140.Glueck CJ, Khalil Q, Winiarska M, Wang P. Interaction of duloxetine and warfarin causing severe elevation of international normalized ratio. JAMA. 2006;295:1517–1518. [DOI] [PubMed] [Google Scholar]
- 141.Jin Y, Desta Z, Stearns V, et al. CYP2D6 genotype, antidepressant use, and tamoxifen metabolism during adjuvant breast cancer treatment. J Natl Cancer Inst. 2005;97:30–39. h [DOI] [PubMed] [Google Scholar]
- 142.Patroneva A, Connolly SM, Fatato P, et al. An assessment of drug-drug interactions: the effect of desvenlafaxine and duloxetine on the pharmacokinetics of the CYP2D6 probe desipramine in healthy subjects. Drug Metab Dispos. 2008;36:2484–2491. [DOI] [PubMed] [Google Scholar]
- 143.Skinner MH, Kuan HY, Pan A, et al. Duloxetine is both an inhibitor and a substrate of cytochrome P4502D6 in healthy volunteers. Clin Pharmacol Ther. 2003;73:170–177. [DOI] [PubMed] [Google Scholar]
- 144.Spina E, Trifiro G, Caraci F. Clinically significant drug interactions with newer antidepressants. CNS Drugs. 2012;26:39–67. [DOI] [PubMed] [Google Scholar]
- 145.Brami C, Bao T, Deng G. Natural products and complementary therapies for chemotherapy-induced peripheral neuropathy: A systematic review. Crit Rev Oncol Hematol. 2016;98:325–334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Bao T, Goloubeva O, Pelser C, et al. A pilot study of acupuncture in treating bortezomib-induced peripheral neuropathy in patients with multiple myeloma. Integr Cancer Ther. 2014;13:396–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Pachman DR, Weisbrod BL, Seisler DK, et al. Pilot evaluation of Scrambler therapy for the treatment of chemotherapy-induced peripheral neuropathy. Support Care Cancer. 2015;23:943–951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Smith TJ, Coyne PJ, Parker GL, Dodson P, Ramakrishnan V. Pilot trial of a patient-specific cutaneous electrostimulation device (MC5-A Calmare®) for chemotherapy-induced peripheral neuropathy. J Pain Symptom Manag. 2010;40:883–891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Coyne PJ, Wan W, Dodson P, Swainey C, Smith TJ. A trial of scrambler therapy in the treatment of cancer pain syndromes and chronic chemotherapy-induced peripheral neuropathy. J Pain Palliat Care Pharmacother. 2013;27:359–364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Wonders KY, Whisler G, Loy H, Holt B, Bohachek K, Wise R. Ten weeks of home-based exercise attenuates symptoms of chemotherapy-induced peripheral neuropathy in breast cancer patients. Heal Psychol Res. 2013;1:28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Streckmann F, Kneis S, Leifert JA, et al. Exercise program improves therapy-related side-effects and quality of life in lymphoma patients undergoing therapy. Ann Oncol. 2014;25:493–499. [DOI] [PubMed] [Google Scholar]
- 152.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. [DOI] [PubMed] [Google Scholar]
- 153.Ehde DM, Dillworth TM, Turner JA. Cognitive-behavioral therapy for individuals with chronic pain: efficacy, innovations, and directions for research. Am Psychol. 2014;69:153–166. [DOI] [PubMed] [Google Scholar]
- 154.Dalton JA, Keefe FJ, Carlson J, Youngblood R. Tailoring cognitive-behavioral treatment for cancer pain. Pain Manag Nurs. 2004;5:3–18. [DOI] [PubMed] [Google Scholar]
- 155.Knoerl R, Lavoie Smith EM, Weisberg J. Chronic pain and cognitive behavioral therapy: an integrative review. West J Nurs Res. 2016;38:596–628. [DOI] [PubMed] [Google Scholar]
- 156.Tatrow K, Montgomery GH. Cognitive behavioral therapy techniques for distress and pain in breast cancer patients: a meta-analysis. J Behav Med. 2006;29:17–27. [DOI] [PubMed] [Google Scholar]
- 157.Turk DC. Clinical effectiveness and cost-effectiveness of treatments for patients with chronic pain. Clin J Pain. 2002;18:355–365. [DOI] [PubMed] [Google Scholar]
- 158.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;19:382–394. [DOI] [PubMed] [Google Scholar]
- 159.Drel VR, Pacher P, Vareniuk I, et al. A peroxynitrite decomposition catalyst counteracts sensory neuropathy in streptozotocin-diabetic mice. Eur J Pharmacol. 2007;569:48–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Brederson JD, Joshi SK, Browman KE, et al. PARP inhibitors attenuate chemotherapy-induced painful neuropathy. J Peripher Nerv Syst. 2012;17:324–330. [DOI] [PubMed] [Google Scholar]
- 161.Krukowski K, Ma J, Golonzhka O, et al. HDAC6 inhibition effectively reverses chemotherapy-induced peripheral neuropathy. Pain. 2017;158:1126–1137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.D’Ydewalle C, Krishnan J, Chiheb DM, et al. HDAC6 inhibitors reverse axonal loss in a mouse model of mutant HSPB1-induced Charcot-Marie-Tooth disease. Nat Med. 2011;17:968–974. [DOI] [PubMed] [Google Scholar]
- 163.Azevedo MI, Pereira AF, Nogueira RB, et al. The antioxidant effects of the flavonoids rutin and quercetin inhibit oxaliplatin-induced chronic painful peripheral neuropathy. Mol Pain. 2013;9:1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Shahid M, Subhan F, Ahmad N, Sewell RDE. The flavonoid 6-methoxyflavone allays cisplatin-induced neuropathic allodynia and hypoalgesia. Biomed Pharmacother. 2017;95:1725–1733. [DOI] [PubMed] [Google Scholar]
- 165.Salat K, Furgala A, Salat R. Evaluation of cebranopadol, a dually acting nociceptin/orphanin FQ and opioid receptor agonist in mouse models of acute, tonic, and chemotherapy-induced neuropathic pain. Inflammopharmacology. 2018;26:361–374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Given CW, Given BA. Symptom management and psychosocial outcomes following cancer. Semin Oncol. 2013;40:774–783. [DOI] [PubMed] [Google Scholar]
- 167.Smith EML, Campbell G, Tofthagen C, et al. Nursing knowledge, practice patterns, and learning preferences regarding chemotherapy-induced peripheral neuropathy. Oncol Nurs Forum. 2014;41:669–679. [DOI] [PubMed] [Google Scholar]