• Open Access
    Review

    The role of physical activity against chemotherapy-induced peripheral neuropathy: a narrative review

    Daniele Diotti 1
    Luca Puce 1
    Laura Mori 1,2
    Carlo Trompetto 1,2
    Elena Saretti 1
    Cecilia Contenti 1
    Lucio Marinelli 1,2*

    Explor Neuroprot Ther. 2022;2:87–99 DOI: https://doi.org/10.37349/ent.2022.00020

    Received: January 27, 2022 Accepted: April 11, 2022 Published: April 28, 2022

    Academic Editor: Noureddin Nakhostin Ansari, Tehran University of Medical Sciences, Iran

    Abstract

    Several studies investigated the side effect of adjuvant cancer treatments, and different types of preventive techniques or treatments have been assessed. Chemotherapy-induced peripheral neuropathy (CIPN) is the most common neurological side effect. Exercise training has been widely studied as an adjuvant therapy to prevent CIPN and improve post-chemotherapy functional outcome and quality of life (QoL). This narrative review aims to summarize the data obtained from the latest studies about physical activity (PA) for the prevention and treatment of CIPN and associated QoL measures. Literature research was conducted to obtain studies including PA interventions for patients with CIPN. Ten studies met inclusion criteria and were therefore summarized and discussed, focusing on exercise type and functional outcome. It seems clear that, regardless of the type of exercise, PA plays a positive role in the treatment of CIPN, providing a significant symptom improvement. There has been no standardization of type, quantity, and intensity of PA administered to the subjects in the various studies probably due to a physiological difference between samples, grade of neuropathy, and difference among therapies.

    Keywords

    Chemotherapy-induced peripheral neuropathy, cancer, physical activity, treatment, prevention

    Introduction

    In recent years, there have been many progress in neoplastic disease treatment, both in terms of diagnosis, through the development of increasingly sensitive analysis tests, and their treatment, through the use of new drugs. Suffice it to say that the number of cancer survivors is rising constantly: for example, in the United States more than 16.9 million Americans (8.1 million males and 8.8 million females) with a history of cancer were alive on January 1st, 2019; this number is projected to reach more than 22.1 million by January 1st, 2030 [1].

    With the rise in survivors of different kinds of tumors, there is more and more evidence of drug side effects. Indeed, survivors often develop physical limitations or pathological conditions caused by the chemotherapy treatment, and very often, these side effects are long-lasting, well beyond the end of the chemotherapy treatment [2, 3]. Although very effective for cancer treatment, antineoplastic drugs often determine side effects that can be very serious and could affect the quality of life (QoL). Chemotherapy-induced peripheral neuropathy (CIPN) is among the most relevant complication affecting the peripheral nervous system. Chemotherapy agents can cause a variety of neuropathies involving large and small fibers, with demyelinating and axonal features, determining sensory, motor, or autonomic dysfunction [4]. Prevalence of neuropathies is high (19–85%) especially if predisposing factors are present, including age, preexistent neuropathy, renal or hepatic function impairment, exposures to other neurotoxic agents, paraneoplastic antibodies, and independent cancer-associated neuropathy [5].

    In the last few years, there has been an increase in the literature about the prevention and treatment of CIPN and the role of physical activity (PA) in managing the side effects of chemotherapy treatments.

    PA for cancer survivors is recommended according to the international guidelines proposed by the sports medicine associations, such as the American College of Sports Medicine (ACSM), American Heart Association (AHA), American Cancer Society (ACS), and US Department of Health and Human Services (US DHHS) [68]. All these guidelines are similar, with minor variations.

    In a concept paper published in 2017, a different approach has been proposed which takes into account the different characteristics of the European population compared to the American one, to provide a more suitable tool. These guidelines are not validated but adhere closely to the ACSM model [9].

    Although it seems clear how the studies recognize the PA as an important role in terms of re-education to movement, countering negative effects, and QoL in cancer survivors, there are few references to the specific problem of CIPN.

    This review aims to provide an overview of the role of PA as an adjunct preventative treatment for CIPN.

    Methods

    Pubmed and Scopus databases have been searched using various combinations of keywords related to CIPN and exercise and results were limited to manuscripts published in the last 20 years.

    Studies had to include a PA intervention on cancer patient survivors who developed CIPN. Regardless of the design chosen by the authors, studies had to include a pre and post evaluation in order to assess the effect of the PA on patients at least as regards improvement of CIPN, balance, or QoL.

    Further inclusion criteria were: English language, one or both genders from all races, all ages, and any diagnosis of cancer. Cross-sectional studies, case reports, published abstracts, dissertation materials, and conference presentations were not included.

    The studies considered most influential, have been inserted in a summary table (Table 1) where the types of PA administered, quantity, and the respective effects of the intervention have been underlined.

    CIPN

    From a clinical point of view, CIPN may appear with different symptoms, which are caused by the action of chemotherapeutic agents on nerves.

    These symptoms often involve sensory aspects, such as numbness, widespread pain, hypersensitivity to mechanical and cold stimuli, paresthesia, and loss of proprioception. CIPN could affect the QoL and increase the risk of falling as well [10].

    Sensory disturbances usually reflect the underlying length-dependent sensory neuropathy that progresses with a subacute course affecting distal limbs first and eventually progressing in a distal-proximal direction.

    Summary of the most relevant studies about CIPN and PA

    ReferenceNumber subjects, age, median (range), sex, IG–CGDiagnosisMethodsProtocolOutcome measurementResultsConclusions
    Kneis et al. [11]Pre-protocol subjects:
    N = 37
    IG: 18, female: 14, male: 4, age median 70 (44–82)
    CG: 19, female: 12, male: 7, age median 60 (46–75)
    Breast cancer, colorectal cancer, gynecological cancer, upper gastrointestinal cancer, NSCLC, non-Hodgkin’s lymphoma, multiple myelomaOne-on-one IG: 12 weeks twice/weekly (endurance + balance training)
    CG: active control group (endurance training)
    Endurance training:
    30 min stable bike moderate intensity (under the IAT)
    Balance training:
    3–8 exercises 3 repetitions each 20/30 s progressively increasing exercise difficulty (reducing the support surface, visual input, adding motor/cognitive tasks, and instability induction
    Functional performance
    CIPN symptoms and QoL Cardiorespiratory fitness
    Endurance training induced a reduction in sensory symptoms in both groups, while balance training additionally improved patients’ functional statusBoth exercises provide a clear and relevant benefit for patients with CIPN
    Streckman et al. [12]N = 61
    IG: 30
    CG: 31
    LymphomaIG: 36 weeks, 2 times per week (sensorimotor-, endurance- and strength training)
    CG: usual care
    Endurance training:
    Treadmill, bike-dynamometer 70–80% HRmax
    Sensorimotor:
    4 postural stabilization tasks progressively increase task difficulty
    Resistance training:
    4 exercises carried out at maximum force
    QoL (EORTC QLQ-C30 questionnaire)
    Movement coordination, endurance, strength, and therapy-induced side-effects
    Increase of balance in IG compared with CG. Static (P = 0.03), dynamic (P = 0.007), perturbed (P = 0.009), and bipedal (P = 0.006)Exercise, especially sensorimotor training provide benefit to cancer patients, ↑ QoL (emotional function, pain, constipation, diarrhea), ↓ side-effects, ↑ balance control, ↑ mobility
    Kleckner et al.[13]N = 355, age: 56 ± 11, 93% female
    IG: 170
    CG: 185
    Different types of cancer (mostly breast cancer)IG: 6 weeks, daily session (EXCAP©®)
    CG: usual care
    Endurance training:
    Low-to-moderate walking: 60–85% of HRR
    Strength training:
    Bands: RPE of 3–5 (1–10 scale)
    Effects of exercise on CIPN symptoms
    Factors that predict CIPN symptoms
    Factors that moderate effects of exercise on CIPN symptoms
    Reduction symptoms (on 10-point scale) hot/cold in hand/feet (-0.46 point) P = 0.045; reduction numbness and tingling (-0.42 point) P = 0.061
    More effect in older people (P = 0.086), male (P = 0.028), and breast cancer (0.076)
    Exercise appears to reduce CIPN symptoms in patients receiving taxane-, platinum-, or vinca alkaloid-based chemotherapy
    Vollmers et al. [14]N = 36
    IG: 17, age: 48.56 ± 11.94
    CG: 19, age: 52.39 ± 10.14
    Breast cancerIG: 2 times/week during chemotherapy and 6 weeks after the end (physical training and sensorimotor exercises)
    CG: received an instruction sheet with a PA program to do autonomously by the patients
    Not reportedBalance, upper and lower strength, QoL (EORTC and MFI questionnaires)No significant difference reported↑ Postural stability in IG
    Moderate strength exercises proved to prevent a loss of upper extremity strength
    ↑ In the decreased intensity of CIPN or possibly faster remission
    Zimmer et al. [15]N = 30
    IG: 17
    CG: 13
    Colorectal cancerIG: 8 weeks–two times/week (a combo of endurance and resistance training)
    CG: usual care (written standard recommendations to obtain physical fitness)
    Endurance training:
    Walking, bicycle ergometer, or cross-trainer
    Resistance training:
    Circuit training of bench press, lat pulldown, leg press, seated row, and abdominal exercise
    Trial outcome index (TOI)
    Balance
    Neuropathic symptoms remained stable in the IG over time, while CIPN significantly worsened in CG of 7.14 points (P = 0.077) of the FACT/GOG-NTX questionnaire
    IG significantly
    Improved strength and balance function
    ↓ CIPN symptoms, ↑ balance, ↑ strength, ↑ QoL
    Henke et al. [16]N = 29
    IG: 18
    CG: 11
    Lung cancer (NSCLC and SCLC)IG: 5 times/week endurance training and breathing technique; 2 times/week strength training
    CG: usual care
    Endurance training:
    Walking 55–70% of HRR
    Strength training:
    50% of RM
    Barthel index
    QoL (EORTC QLQ C-30/LC-13 questionnaire)
    Barthel index was significantly worse after the intervention in CG (P = 0.041); functional capacity increased more in IG (6MWT); an increased single score of EORTC QLQ C-30/LC-13 questionnaire↑ QoL (physical functioning, hemoptysis, pain in arms or shoulder, peripheral neuropathy, cognitive functioning)
    Training has a positive effect on the patient’s endurance and strength capacity
    Wonders et al. [17]N = 6 (finished the intervention), female subjectsBreast cancerIG: 10-week home-based program
    CG: not present
    Endurance training:
    Moderate-intensity 55–65% of their self-estimated HRR
    Increase gradually from 20 min (1st week) to 150 min (10th week) of exercise per week
    Resistance training:
    Not reported
    CIPN symptoms rate
    QoL (McGill QoL questionnaire)
    Significantly lowered levels of both troublesome and pain symptoms↓ CIPN symptoms
    ↑ QoL (level of troublesome symptoms related to peripheral neuropathy)
    Low adherence rate
    Difficulty getting subjects to complete the 10-week program
    Andersen Hammond et al. [18]N = 48
    IG: 22 mean age 56.3 ± 9.9
    CG: 26 mean age 53.0 ± 10.3
    Breast cancerIG: 3 times daily home-based exercise program; 5/10 min every session
    CG: usual care
    Nerve gliding exercise programEffects of exercise on CIPN symptoms
    QoL (DASH questionnaire)
    Strength
    Less pain for IG (P = 0.053) preservation of vibration (P = 0.001) and heat pain thresholds (left P = 0.021, right P = 0.039)
    Significant improvements in grip strength
    ↓ CIPN pain
    ↓ Pain pressure
    ↑ Grip dynamometry
    Van Waart et al. [19]N = 230
    IG: 153; HBI = 77, HIP = 76
    CG: 77
    Breast or colon cancerIG: divided into “Onco-Move program” and “OnTrack program
    CG: usual care
    Onco-Move:
    Home-based program, 30 min, 5 days/week, 12–14 Borg Scale, exercises not reported
    OnTrack program:
    Supervised 20 min, 2 days/week
    Endurance training:
    30 minutes, 50% to 80% of the maximal workload, exercises not reported
    Resistance training:
    2 series of 8 repetitions at 80% of the 1RM
    Cardiorespiratory fitness, upper muscle strength, lower muscle strength, fatigue, QoL (EORTC QLQ-C30 questionnaire), functioning in daily life, quality of sleep, return to work, psychological distress, self-reported PA level, chemotherapy regimen, dose, and adverse effects of chemotherapy, compliance with exercise programsIGs: less decline of cardiorespiratory fitness (P < 0.001), better physical functioning (P < 0.001), less pain (HBI P = 0.003, HIP P = 0.011)Both IGs provide a benefit for patients, especially the HIP group
    McCrary et al. [20]A single group for pre and post
    N = 29 mean age 61.6 (32–79)
    Female 21, male 8
    Breast cancer, colorectal cancer, ovarian cancer, endometrial cancer, appendix cancer, lymphoma, myeloma, urothelial, carcinomaIG: 8 weeks, 3 times weekly (resistance training, balance training, and endurance training)
    CG: not present
    Exercises performed at RPE of 13–15 (6–20 scale)Objective CIPN, patient-reported CIPN, patient-reported disability, QoL (SF-36 questionnaire), mobility, dynamic balance/leg strength, standing balance, sensoryneurophysiology, motor neurophysiologyDynamic balance, standing balance in eye-open conditions, mobility, and QoL were improved (P ≤ 0.05)
    Ameliorating objective CIPN and patient reported CIPN
    Significance increasing of:
    Objective CIPN, patient-reported CIPN, patient-reported disability, QoL, mobility
    Dynamic balance, stable surfacewith eyes open, unstable surface with eyes open
    Display full size

    IG: intervention group; CG: control group; HRmax: maximum heart rate; HRR: heart rate reserve; RM: maximum repetitions; RPE: rated perceived exertion; IAT: individual anaerobic threshold; 1RM: one repetition maximum; NSCLC: non-small cell lung cancer; SCLC: small-cell lung cancer; ↑: increase; ↓: decrease; 6MWT: 6-minute walking test; HBI: home-based intervention; HIP: high intensity program

    Symptoms could appear both while taking medication (e.g., typically using platinum-based drugs) [21], and even a long time after treatment, in a condition called “coasting” [22].

    It is very common for patients to develop tingling and numbness in the hands and feet during the first year of chemotherapy treatment follow-up. Symptoms in the lower limbs typically last longer than those in the upper limbs [23].

    The mechanisms of action and molecular target of these chemotherapeutic agents are diverse and include both DNA and microtubular targets to arrest cell division and induce cell death. The pathobiology of CIPN, irrespective of the causative agent, shares some important similarities. For example, sensory neurons are those most affected and longer axons are damaged first. Histopathological changes associated with CIPN commonly involve large, myelinated fibers and result in sensorimotor symptoms such as hypersensitivity to mechanical stimuli or distal weakness due to mechanisms that are not entirely understood [24, 25].

    There are six main substance groups that may damage peripheral sensory and motor fibers: platinum-based antineoplastics (particularly oxaliplatin and cisplatin), vinca alkaloids (particularly vincristine and vinblastine), epothilones (ixabepilone), taxanes (paclitaxel, docetaxel), proteasome inhibitors (bortezomib) and immunomodulatory drugs (thalidomide) [24].

    Antineoplastic drugs may induce CIPN with different timing and modalities. Platinum-based drugs have the highest incidence of neurotoxicity with a rate ranging from 70% to 100% of all treated patients [26]. Furthermore, oxaliplatin may induce neuropathy in the very early phase of the treatment [27]. The mechanism of action of platinum-based drugs develops damage to the nucleus inhibiting the DNA replication and mRNA transcription [24].

    Vinca alkaloids, used mainly to treat lung, brain, and bladder testicular cancer, may determine CIPN in up to 20% of treated patients [26]. The cytotoxic action of vinca alkaloids is provoked by its ability to bond the β-subunit of tubulin and inhibits microtubule formation [24].

    The ixabepilone is an analog of epothilone B used in breast cancer patients and belongs to a relatively new class of anti-cancer drugs that act as tubulin destabilizers effectively preventing divisions of cancer cells. Evidence from clinical trials showed that 65% of patients treated with ixabepilone demonstrated symptoms of neuropathy, therefore reflecting a high incidence of CIPN [26]. Taxanes are drugs used for different types of cancer: breast, ovarian, lung, pancreatic, and prostate cancer. In relation to the type of taxanes used, there is a variety in the incidence of CIPN ranging from 11% to 87% [26]. Proteasome inhibitors are drugs used against multiple myeloma and achieve their anticancer effects by inhibiting the activity of the proteasome of cancer cells. This leads to an accumulation of aberrant proteins, hence the arrest of the cell cycle and finally to apoptosis. These drugs determine a moderate (20–30%) incidence of CIPN [26].

    Immunomodulatory drugs such as thalidomide cause lower limb neuropathies estimated to affect 20% to 60% of all cancer patients treated with this drug [26].

    Long-term effects of CIPN consist of depression, insomnia, and worse QoL. Although some risk factors have been identified such as age, pre-existing neuropathy, nutrient deficiency, and concomitant drugs facilitating neurotoxicity, these are difficult to overcome and CIPN incidence is still very high [28].

    Literature on long-term effects is still very sparse, especially regarding the effects of thalidomide and bortezomib. In addition, there is ample literature regarding the effects of chemotherapy drugs but, only recently some studies have been considered the QoL of cancer patients with CIPN.

    Prevention and therapy

    Pharmacological intervention

    To date, drug therapies aimed at preventing CIPN have not produced encouraging results. Indeed, studies evaluating the effects of some drugs, including chemoprotective, anticonvulsants, antidepressants, and dietary supplements, did not yield any significant data [29].

    This is also confirmed by a more recent review, which assessed the effect of many drugs to prevent CIPN (carbamazepine, minocycline, nimodipine) and supplements (acetyl-L-carnitine, acetylcysteine, α-lipoic acid, amifostine, calcium, magnesium, diethyldithiocarbamate, glutathione, gosha-jinki-gan, omega-3 fatty acids, vitamins B and E) without demonstrating any positive results [30].

    Limitations in studies of these substances include small sample sizes, lack of placebo control groups, and in some cases, the study design was not blinded [31].

    In another recent review, Hu et al. [32] highlighted the effects of some preventive treatments aimed at blocking CIPN onset. To do this, they examined the mechanisms by which antineoplastic drugs cause a degeneration of the nervous system. Unlike the previous study, this review showed that glutathione and its precursor, N-acetylcysteine, could have positive effects to prevent neurotoxicity induced by platinum-based drugs.

    Non-pharmacological intervention

    There are studies supporting the hypothesis that the cryotherapy technique may have a decisive role in the prevention of CIPN. In a recent study, encouraging results were obtained about neuropathies induced by taxanes drugs (paclitaxel) [33]. A recently published review highlighted how there is a positive effect on the prevention of CIPN through the use of cryotherapy without finding any type of side effect [34]. However, many studies are still needed to confirm the efficacy of cryotherapy in the prevention of CIPN. Cryotherapy associated with taxane-based chemotherapy determined better sensory and motor scores on the Patient Neurotoxicity Questionnaire [35].

    Another non-pharmacological strategy derives from the use of surgical gloves which reduce blood flow in the most distal part of the fingers due to compression. The glove is worn before, during, and after chemotherapy treatment. This technique appeared effective in some studies related to taxane-induced CIPN [36]. Another recent double-blind study, however, seems to refute this hypothesis as there would be no significant differences in the incidence of CIPN between the hand compressed by the glove and the free hand [37].

    Another approach to treat CIPN-related pain is the “Scrambler therapy”, which involves a device providing non-invasive cutaneous electrical stimulation. The impulses are transmitted via surface electrodes placed above the pain region. Exploiting the spinal gating mechanisms, this therapy aims to replace “painful” with “non-painful” signals, with immediate pain reduction [38].

    Scrambler therapy proved more effective than transcutaneous electrical nerve stimulation (TENS) in CIPN patients [39]. However, further studies with placebo and double-blind groups still need to be conducted in order to confirm their efficacy [40].

    Effect of PA in subjects with CIPN

    The positive effects of PA in patients with CIPN have now been demonstrated by several studies. Indeed, it now seems well known how a resistance or endurance exercise program can significantly reduce the side effects of adjuvant therapies involving peripheral neuropathies [41].

    A study conducted on mice has shown that PA carried out beforehand at the beginning of adjuvant therapy with paclitaxel, could have a preventive effect on the onset of thermal hypoalgesia and prevent the reduction of the number of unmyelinated axons [42]. Furthermore, although the data were not significant, a reduction in the diameter of the axons was noted in the group that did not perform exercise.

    Apart from sensory deficits, antineoplastic drug cumulative dose, and cycle number, the risk of falls in persons with CIPN includes the severity of muscle weakness and performance status [43]. A recent review highlighted that PA always produces a positive effect on subjects, regardless of the type of exercise, dosage, and setting [44]. It was also pointed out that studies show a moderate/high risk of bias in addition to considering only an adult population. The same review also argues that, although endurance exercise appears to be very effective in counteracting the limiting effects of CIPN, no studies tested aerobic exercise alone to ameliorate CIPN.

    In fact, a randomized controlled trial study by Kneis et al. [11] developed a protocol in which the control group performed only aerobic activity, and it was then compared to the intervention group where balance exercises were added. In this study, both groups benefited from their respective PA program in terms of neuropathy-related symptoms, suggesting that even endurance exercises alone can be effective in counteracting CIPN [11].

    Due to the physiological diversity of the subject’s conditions, antineoplastic drug type and dose, and the difficulty of quantifying the degree of CIPN, the studies that have dealt with this topic show many differences both in type and quantity of exercises carried out. Although the benefit of a PA intervention is well established, it is not easy to find a standardization of the exercises to propose.

    In a randomized study, Courneya et al. [45] showed how a higher intensity activity can be more effective than moderate activity for younger, normal weight, and more fit subjects [45]. This implies that individuals require different doses of PA depending on their baseline conditions.

    Vollmers et al. [14] structured an intervention where subjects exercised twice a week during the course of chemotherapy and for six weeks thereafter. Although there are no details on the sensorimotor exercises performed, an increase in postural stability was detected in the intervention group [14].

    Some authors have chosen an activity throughout the week, even every day [13, 14] while others have administered PA two or three times per week [14, 15]. However, all authors obtained encouraging results from their respective training protocols.

    The methodology for carrying out the protocol is also at the discretion of the authors. Some prescribe exercises in a gym or in a specialized clinic, instead, other authors have the subjects carry out the training program independently at home [13, 17, 18]. Subjects are instructed about how to perform the exercises and, in some cases, also about effort perception in order to remain within predefined parameters avoiding excessive or insufficient training.

    A mixed methodology was also used one group carried out the activity in the gym at a higher intensity (OnTrack: a moderate- to high-intensity, combined supervised resistance and aerobic exercise program), while another group carried out a more moderate home training program called “Onco-Move”: a low-intensity, home-based PA program. Although both programs produced positive outcomes, the group that performed higher intensity training reached better results [19].

    Another mixed methodology was developed by McCrary et al. [20] in which a single group alternated exercises in a gym, assisted by an operator, and exercises carried out independently at their own home. Also, in this case, there was an improvement in the symptoms of CIPN as well as static and dynamic balance [20]. The Table 1 shows a summary of the studies assessing the effects of PA in relation to the type of exercise carried out.

    It seems clear that there is no standardized methodology to evaluate the effects of PA in terms of methods of administering the exercises: type of activity, duration, and intensity.

    However, it seems clear that, regardless of the method and exercises are chosen, the effects are always ameliorative compared to the control groups that do not carry out any type of activity. Notably, it would be useful to further investigate the effects of aerobic exercise alone as already proposed by Kanzawa-Lee et al. [44].

    Effect of PA in individuals with CIPN on QoL

    Paralleling evidence of a positive role of PA on the physical condition (e.g., strength or balance) of subjects with CIPN, there is much literature that deals with how an exercise program can improve the QoL of individuals during and after chemotherapy treatment.

    Regarding the assessment of the degree of the CIPN, it is not always easy to have objective data as it is necessary to rely on very different assessment tools.

    In a recent review, Park et al. [46] identified 41 assessment tools and, among other results, underlined that most of them (41.5%) are based on patient-reported outcomes (PROs) questionnaires highlighting how there is increasing attention to the subjective evaluation of symptoms.

    However, there is always a discrepancy when using clinical assessment tools and PRO, especially in the intermediate grades of CIPN. It would be advisable to consider the results of clinical tests together with PRO in order to improve the quality of the evaluation of CIPN [47].

    Among subjects diagnosed with cancer undergoing chemotherapy, there are common aspects that can affect their QoL such as depression, anxiety, and lower self-esteem. Physical issues, on the other hand, include fatigue, reduced cardiovascular and respiratory efficiency, muscle weakness and atrophy, pain, difficulty sleeping, and nausea [48].

    In a recent review, it was reported how PA improved QoL in 4 out of 8 studies that considered the topic [44]. Furthermore, Mols et al. [49] indirectly demonstrated that neuropathies induced by chemotherapy drugs are inversely proportional to the PA performed weekly: they showed that statistically significant and clinically relevant worse scores on almost all EORTC QLQ-C30 [50] subscales were reported by those not meeting the PA guideline compared to those who did meet the guideline, regardless of CIPN symptoms.

    Streckmann et al. [12] developed a training protocol for patients with lymphoma, assessing the effects of QoL as the first endpoint. They found a significant improvement in the comparison of the intervention group. In this case, the activity was carried out twice a week for 36 weeks and training sessions lasted about an hour [12].

    Some aspects of QoL such as global health status, social functioning, physical functioning, role functioning, and fatigue, significantly improved in the intervention groups with resistance training and sensorimotor exercise training protocols compared to the usual care. In addition, there was an increase in fear of falling in the usual care group, but not in the resistance training and sensorimotor exercise groups [51].

    These data have been also confirmed by a review that investigated the effects of physical exercise on fatigue (using the multidimensional fatigue inventory), highlighting how subjects benefited from a physical exercise program as regards general fatigue, physical fatigue, reduced activity, and reduced motivation [52].

    Conclusions

    We could not find standardized protocols indicating which type of PA is better for subjects with CIPN, and no uniformity in the modality, quantity, or intensity of the exercises.

    Perhaps this is because samples are always different and not homogeneous as regards age, medical therapy, and entry physical condition.

    More randomized controlled trials are needed to assess the efficacy of specific exercise interventions in order to confirm the efficacy of a single type of PA and provide information to the clinicians about which kind of exercise is preferred to manage CIPN symptoms.

    It seems that aerobic PA plays a decisive role against CIPN symptoms since aerobic exercise-containing interventions led to significant CIPN benefits [44].

    Since endurance PA determined encouraging results in all studies, it is reasonable to think that this type of PA is to be preferred in the management of neuropathies. Walking may also be useful to improve dynamic balance.

    As already suggested by Kanzawa-Lee et al. [44], it would be advisable to develop more protocols that use this training modality to better understand its value.

    However, it seems clear that PA plays a decisive role in significantly reducing the side effects of chemotherapy compared to physical inactivity or usual care. The improvement effect of PA and a better QoL is a consequence of the improvement of specific training aspects (strength, balance, endurance). Exercise seems the best, non-invasive, and side-effect-free practice as an adjuvant treatment of CIPN.

    Furthermore, in most of the studies, PA at home or supported by an operator in specialized structures (labs or gym) is the preferred modality. Future studies could consider PA performed in groups rather than individually in order to evaluate the positive effects on mood and motivation.

    Abbreviations

    CIPN:

    chemotherapy-induced peripheral neuropathy

    PA:

    physical activity

    PROs:

    patient-reported outcomes

    QoL:

    quality of life

    Declarations

    Author contributions

    DD designed the study and wrote the first draft of the manuscript; ES and CC contributed to the study conception and design; L Marinelli, LP, L Mori and CT revised the manuscript. All authors read and approved the submitted version.

    Conflicts of interest

    The authors declare that they have no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    Not applicable.

    Copyright

    © The Author(s) 2022.

    References

    Miller KD, Nogueira L, Mariotto AB, Rowland JH, Yabroff KR, Alfano CM, et al. Cancer treatment and survivorship statistics, 2019. CA Cancer J Clin. 2019;69:36385. [DOI] [PubMed]
    Glare PA, Davies PS, Finlay E, Gulati A, Lemanne D, Moryl N, et al. Pain in cancer survivors. J Clin Oncol. 2014;32:173947. [DOI] [PubMed] [PMC]
    Seretny M, Currie GL, Sena ES, Ramnarine S, Grant R, MacLeod MR, et al. Incidence, prevalence, and predictors of chemotherapy-induced peripheral neuropathy: a systematic review and meta-analysis. Pain. 2014;155:246170. [DOI] [PubMed]
    Cioroiu C, Weimer LH. Update on chemotherapy-induced peripheral neuropathy. Curr Neurol Neurosci Rep. 2017;17:47. [DOI] [PubMed]
    Zajączkowska R, Kocot-Kępska M, Leppert W, Wrzosek A, Mika J, Wordliczek J. Mechanisms of chemotherapy-induced peripheral neuropathy. Int J Mol Sci. 2019;20:1451. [DOI] [PubMed] [PMC]
    Haskell WL, Lee IM, Pate RR, Powell KE, Blair SN, Franklin BA, et al. Physical activity and public health: updated recommendation for adults from the American College of Sports Medicine and the American Heart Association. Med Sci Sports Exerc. 2007;39:142334. [DOI] [PubMed]
    Doyle C, Kushi LH, Byers T, Courneya KS, Demark-Wahnefried W, Grant B, et al.; 2006 Nutrition, Physical Activity and Cancer Survivorship Advisory Committee; American Cancer Society. Nutrition and physical activity during and after cancer treatment: an American Cancer Society guide for informed choices. CA Cancer J Clin. 2006;56:32353. [DOI] [PubMed]
    Physical Activities Guidelines Advisory Committee. Physical activity guidelines advisory committee report, 2008 [Internet]. Washington (DC): U.S. Department of Health and Human Services; [cited 2022 Jan 26]. Available from: https://health.gov/sites/default/files/2019-10/CommitteeReport_7.pdf
    Stefani L, Galanti G, Klika R. Clinical implementation of exercise guidelines for cancer patients: adaptation of ACSM’s guidelines to the Italian model. J Funct Morphol Kinesiol. 2017;2:4. [DOI]
    Kolb NA, Smith AG, Singleton JR, Beck SL, Stoddard GJ, Brown S, et al. The association of chemotherapy-induced peripheral neuropathy symptoms and the risk of falling. JAMA Neurol. 2016;73:8606. [DOI] [PubMed] [PMC]
    Kneis S, Wehrle A, Müller J, Maurer C, Ihorst G, Gollhofer A, et al. It’s never too late—balance and endurance training improves functional performance, quality of life, and alleviates neuropathic symptoms in cancer survivors suffering from chemotherapy-induced peripheral neuropathy: results of a randomized controlled trial. BMC Cancer. 2019;19:414. [DOI] [PubMed] [PMC]
    Streckmann F, Kneis S, Leifert JA, Baumann FT, Kleber M, Ihorst G, et al. Exercise program improves therapy-related side-effects and quality of life in lymphoma patients undergoing therapy. Ann Oncol. 2014;25:4939. [DOI] [PubMed]
    Kleckner IR, Kamen C, Gewandter JS, Mohile NA, Heckler CE, Culakova E, et al. Effects of exercise during chemotherapy on chemotherapy-induced peripheral neuropathy: a multicenter, randomized controlled trial. Support Care Cancer. 2018;26:101928. [DOI] [PubMed] [PMC]
    Vollmers PL, Mundhenke C, Maass N, Bauerschlag D, Kratzenstein S, Röcken C, et al. Evaluation of the effects of sensorimotor exercise on physical and psychological parameters in breast cancer patients undergoing neurotoxic chemotherapy. J Cancer Res Clin Oncol. 2018;144:178592. [DOI] [PubMed]
    Zimmer P, Trebing S, Timmers-Trebing U, Schenk A, Paust R, Bloch W, 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;26:61524. [DOI] [PubMed]
    Henke CC, Cabri J, Fricke L, Pankow W, Kandilakis G, Feyer PC, et al. Strength and endurance training in the treatment of lung cancer patients in stages IIIA/IIIB/IV. Support Care Cancer. 2014;22:95101. [DOI] [PubMed]
    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. Health Psychol Res. 2013;1:e28. [DOI] [PubMed] [PMC]
    Andersen Hammond E, Pitz M, Steinfeld K, Lambert P, Shay B. An exploratory randomized trial of physical therapy for the treatment of chemotherapy-induced peripheral neuropathy. Neurorehabil Neural Repair. 2020;34:23546. [DOI] [PubMed]
    van Waart H, Stuiver MM, van Harten WH, Geleijn E, Kieffer JM, Buffart LM, 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;33:191827. [DOI] [PubMed]
    McCrary JM, Goldstein D, Sandler CX, Barry BK, Marthick M, Timmins HC, et al. Exercise-based rehabilitation for cancer survivors with chemotherapy-induced peripheral neuropathy. Support Care Cancer. 2019;27:384957. [DOI] [PubMed]
    Wilson RH, Lehky T, Thomas RR, Quinn MG, Floeter MK, Grem JL. Acute oxaliplatin-induced peripheral nerve hyperexcitability. J Clin Oncol. 2002;20:176774. [DOI] [PubMed]
    Argyriou AA, Bruna J, Marmiroli P, Cavaletti G. Chemotherapy-induced peripheral neurotoxicity (CIPN): an update. Crit Rev Oncol Hematol. 2012;82:5177. [DOI] [PubMed]
    Bonhof CS, van de Poll-Franse LV, Wasowicz DK, Beerepoot LV, Vreugdenhil G, Mols F. The course of peripheral neuropathy and its association with health-related quality of life among colorectal cancer patients. J Cancer Surviv. 2021;15:190200. [DOI] [PubMed] [PMC]
    Starobova H, Vetter I. Pathophysiology of chemotherapy-induced peripheral neuropathy. Front Mol Neurosci. 2017;10:174. [DOI] [PubMed] [PMC]
    Ibrahim EY, Ehrlich BE. Prevention of chemotherapy-induced peripheral neuropathy: a review of recent findings. Crit Rev Oncol Hematol. 2020;145:102831. [DOI] [PubMed] [PMC]
    Banach M, Juranek JK, Zygulska AL. Chemotherapy-induced neuropathies-a growing problem for patients and health care providers. Brain Behav. 2016;7:e00558. [DOI] [PubMed] [PMC]
    Argyriou AA, Cavaletti G, Briani C, Velasco R, Bruna J, Campagnolo M, et al. Clinical pattern and associations of oxaliplatin acute neurotoxicity: a prospective study in 170 patients with colorectal cancer. Cancer. 2013;119:43844. [DOI] [PubMed]
    Kerckhove N, Collin A, Condé 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] [PubMed] [PMC]
    Hershman DL, Lacchetti C, Dworkin RH, Lavoie Smith EM, Bleeker J, Cavaletti G, et al.; American Society of Clinical Oncology. 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;32:194167. [DOI] [PubMed]
    Jordan B, Jahn F, Sauer S, Jordan K. Prevention and management of chemotherapy-induced polyneuropathy. Breast Care (Basel). 2019;14:7984. [DOI] [PubMed] [PMC]
    Brewer JR, Morrison G, Dolan ME, Fleming GF. Chemotherapy-induced peripheral neuropathy: current status and progress. Gynecol Oncol. 2016;140:17683. [DOI] [PubMed] [PMC]
    Hu LY, Mi WL, Wu GC, Wang YQ, Mao-Ying QL. Prevention and treatment for chemotherapy-induced peripheral neuropathy: therapies based on CIPN mechanisms. Curr Neuropharmacol. 2019;17:18496. [DOI] [PubMed] [PMC]
    Hanai A, Ishiguro H, Sozu T, Tsuda M, Yano I, Nakagawa T, et al. Effects of cryotherapy on objective and subjective symptoms of paclitaxel-induced neuropathy: prospective self-controlled trial. J Natl Cancer Inst. 2018;110:1418. [DOI] [PubMed] [PMC]
    Bailey AG, Brown JN, Hammond JM. Cryotherapy for the prevention of chemotherapy-induced peripheral neuropathy: a systematic review. J Oncol Pharm Pract. 2021;27:15664. [DOI] [PubMed]
    Shigematsu H, Kimura Y, Itagaki T, Yasui D. Preventive effect of cryotherapy on persistent chemotherapy-induced peripheral neuropathy after completion of weekly paclitaxel therapy for breast cancer. Version: 1. [Preprint]. [cited 2022 Jan 26]. Available from: https://europepmc.org/article/PPR/PPR338488#impact
    Tsuyuki S, Senda N, Kanng Y, Yamaguchi A, Yoshibayashi H, Kikawa Y, et al. Evaluation of the effect of compression therapy using surgical gloves on nanoparticle albumin-bound paclitaxel-induced peripheral neuropathy: a phase II multicenter study by the Kamigata Breast Cancer Study Group. Breast Cancer Res Treat. 2016;160:617. [DOI] [PubMed]
    Kotani H, Terada M, Mori M, Horisawa N, Sugino K, Kataoka A, et al. Compression therapy using surgical gloves does not prevent paclitaxel-induced peripheral neuropathy: results from a double-blind phase 2 trial. BMC Cancer. 2021;21:548. [DOI] [PubMed] [PMC]
    Pachman DR, Weisbrod BL, Seisler DK, Barton DL, Fee-Schroeder KC, Smith TJ, et al. Pilot evaluation of scrambler therapy for the treatment of chemotherapy-induced peripheral neuropathy. Support Care Cancer. 2015;23:94351. [DOI] [PubMed] [PMC]
    Loprinzi C, Le-Rademacher JG, Majithia N, McMurray RP, O’Neill CR, Bendel MA, et al. Scrambler therapy for chemotherapy neuropathy: a randomized phase II pilot trial. Support Care Cancer. 2020;28:118397. [DOI] [PubMed]
    Li Y, Lustberg MB, Hu S. Emerging pharmacological and non-pharmacological therapeutics for prevention and treatment of chemotherapy-induced peripheral neuropathy. Cancers (Basel). 2021;13:766. [DOI] [PubMed] [PMC]
    Furmaniak AC, Menig M, Markes MH. Exercise for women receiving adjuvant therapy for breast cancer. Cochrane Database Syst Rev. 2016;9:CD005001. [DOI] [PubMed] [PMC]
    Park JS, Kim S, Hoke A. An exercise regimen prevents development paclitaxel induced peripheral neuropathy in a mouse model. J Peripher Nerv Syst. 2015;20:714. [DOI] [PubMed]
    Tofthagen C, Overcash J, Kip K. Falls in persons with chemotherapy-induced peripheral neuropathy. Support Care Cancer. 2012;20:5839. [DOI] [PubMed] [PMC]
    Kanzawa-Lee GA, Larson JL, Resnicow K, Smith EML. Exercise effects on chemotherapy-induced peripheral neuropathy: a comprehensive integrative review. Cancer Nurs. 2020;43:E17285. [DOI] [PubMed]
    Courneya KS, McKenzie DC, Mackey JR, Gelmon K, Friedenreich CM, Yasui Y, et al. Subgroup effects in a randomised trial of different types and doses of exercise during breast cancer chemotherapy. Br J Cancer. 2014;111:171825. [DOI] [PubMed] [PMC]
    Park SB, Alberti P, Kolb NA, Gewandter JS, Schenone A, Argyriou AA. Overview and critical revision of clinical assessment tools in chemotherapy-induced peripheral neurotoxicity. J Peripher Nerv Syst. 2019;24:S1325. [DOI] [PubMed]
    Alberti P, Rossi E, Cornblath DR, Merkies IS, Postma TJ, Frigeni B, et al.; CI-PeriNomS Group. Physician-assessed and patient-reported outcome measures in chemotherapy-induced sensory peripheral neurotoxicity: two sides of the same coin. Ann Oncol. 2014;25:25764. [DOI] [PubMed] [PMC]
    Brown JK, Byers T, Doyle C, Coumeya KS, Demark-Wahnefried W, Kushi LH, et al.; American Cancer Society. Nutrition and physical activity during and after cancer treatment: an American Cancer Society guide for informed choices. CA Cancer J Clin. 2003;53:26891. [DOI] [PubMed]
    Mols F, Beijers AJ, Vreugdenhil G, Verhulst A, Schep G, Husson O. Chemotherapy-induced peripheral neuropathy, physical activity and health-related quality of life among colorectal cancer survivors from the PROFILES registry. J Cancer Surviv. 2015;9:51222. [DOI] [PubMed]
    Aaronson NK, Ahmedzai S, Bergman B, Bullinger M, Cull A, Duez NJ, et al. The European organization for research and treatment of cancer QLQ-C30: a quality-of-life instrument for use in international clinical trials in oncology. J Natl Cancer Inst. 1993;85:36576. [DOI] [PubMed]
    Müller J, Weiler M, Schneeweiss A, Haag GM, Steindorf K, Wick W, et al. Preventive effect of sensorimotor exercise and resistance training on chemotherapy-induced peripheral neuropathy: a randomised-controlled trial. Br J Cancer. 2021;125:95565. [DOI] [PubMed] [PMC]
    van Vulpen JK, Peeters PH, Velthuis MJ, van der Wall E, May AM. Effects of physical exercise during adjuvant breast cancer treatment on physical and psychosocial dimensions of cancer-related fatigue: a meta-analysis. Maturitas. 2016;85:10411. [DOI] [PubMed]