Skip to main content

Chemotherapy-induced peripheral neuropathy biomarkers: current updates, challenges, and potentials

Abstract

Chemotherapy-induced peripheral neuropathy (CIPN) is a debilitating condition of peripheral nerve because of chemotherapeutic agent exposure among cancer patients. Despite its relatively high prevalence, to date, there has been no specific or standardized diagnostic criteria for CIPN and establishing diagnosis can be burdensome with significant time and efforts. Therefore, there is a need for an accurate biomarker to aid in early and objective diagnosis of CIPN. Based on the relevant pathogenesis of CIPN, herein we discussed several potential biomarker candidates to be incorporated in the diagnosis of CIPN, ranging from bodily fluid-based biomarker such as neurotrophic factors and neurofilaments, genetic biomarker such as microRNAs, electrophysiologic biomarker such as quantitative sensory testing, and imaging biomarkers such as high-resolution ultrasound and magnetic resonance neurography. We also discussed the strengths and weaknesses of each biomarker type, and future directions to accelerate its translation into routine use in clinical practice.

Introduction

Chemotherapy-induced peripheral neuropathy (CIPN) is highly prevalent among cancer patients undergoing chemotherapy. In a meta-analysis, it has been reported to be as high as 68% during the first month of treatment [1]. CIPN can be physically disabling as it is marked by sensibility and motor function impairment, neuropathic pain, ataxia, and other disabilities of the affected individuals [2,3,4]. CIPN potentially reduce their quality of life and, possibly, their adherence to the subsequent chemotherapeutic regimens [5,6,7,8]. However, to date, there has been no specific or standardized diagnostic criteria for CIPN. Establishing CIPN diagnosis varies widely between centers and the clinical evaluation often incorporates exhausting and time-consuming questionnaires [9]. Unfortunately, there has been no reliable and universally accepted quantitative biomarker to aid in early and objective diagnosis of CIPN, although there has been some progress in several areas. Herein we discuss the relevant and contemporary pathomechanisms of CIPN, along with several potential biomarker candidates for CIPN and its future directions to translate it into routine clinical practice.

Molecular mechanisms of chemotherapy-induced peripheral neuropathy

CIPN occurs mainly via two major pathways, i.e., neuroinflammation and altered excitability of peripheral neurons. Platinum-based chemotherapeutic medications such as oxaliplatin, cisplatin, and carboplatin exert its adverse effects toward peripheral nerves in the similar way as it induces its main actions on tumor cells [10]. Chemotherapeutic agents primarily induce alterations of intracellular organelles, affect membrane receptors and ion channels, impairing intracellular homeostasis, signaling, and neurotransmission, all of which resulting in neuroinflammation, DNA damage, and axonal degeneration [10].

Chemotherapeutic agents induce mitochondrial damage, thus increasing the reactive oxygen species (ROS) production which leads to oxidative stress. Oxidative stress, in turn, induces cellular impairment and apoptosis by means of bioenergetic failure (via mitochondrial damage), depletion of intracellular antioxidants, biomolecular damage, microtubular damage, activation of ion channels, demyelination, neuroinflammation, and impaired mitophagy [10,11,12,13,14]. Furthermore, it has been found that dorsal root ganglion (DRG) was vulnerable to damage caused by chemotherapeutic agents because it is less protected by blood–nerve barrier [11, 15]. DRG exposure to platinum-based, vinca alkaloids, taxanes, and thalidomide chemotherapeutic agents have been associated with its damage [16,17,18,19].

ROS directly damages mitochondria and its function. ROS may activate cellular apoptotic pathways and increase the synthesis and secretion of pro-inflammatory cytokines and mediators, by which it accelerates mitochondrial damage [20,21,22,23]. Oftentimes, the resulting mitochondrial damage was irreversible. For instance, oxaliplatin and cisplatin was able to bind to mitochondrial DNA (mDNA). The binding, in turn, impairs mitochondrial DNA replication and transcription, thereby disrupting protein synthesis essential for cellular respiration and metabolism, thus causing mitochondrial and cellular dysfunction [10, 24]. This binding is irreparable since mitochondria does not possess DNA repair mechanism. Another example was mitochondrial dysfunction after vincristine exposure by means of altering mitochondrial Ca 2+ signaling or vacuolization of mitochondria after exposure to paclitaxel [10, 24, 25]. In addition, paclitaxel also damages mitochondria via several mechanisms, including disruption of mitochondrial iron homeostasis via perturbation of ferroptosis pathway and downregulation of FIS1, a gene responsible for mitochondrial fission to maintain cellular energy demands [26].

In addition, ROS was found to directly disrupt microtubules. For instance, taxane-based chemotherapeutic agent was found to bind with β-tubulin of microtubules, inducing over polymerization and impairing normal microtubular function [11, 27,28,29]. Microtubules are critical to axonal transport and impaired microtubular function may lead to peripheral nerve demyelination and neuronal apoptosis. Other chemotherapeutic agents were also linked to microtubular impairment, including vinca alkaloid which binds to tubulin and interferes with mitotic spindle [27, 28, 30]. Similar mechanism was also found on bortezomib exposure which increases microtubule-associated proteins, thus induces microtubular hyper-stabilization [11, 31] (Fig. 1).

Fig. 1
figure 1

Relevant pathomechanism of CIPN with respect to the development of suitable biomarker candidates (adapted from Starobova and Vetter [10])

Oxidative stress is also strongly associated with neuroinflammation. Many chemotherapeutic agents can cross the blood–brain or blood–nerve barrier and directly incite neuroinflammation through activation of a stream of pro-inflammatory cytokines and mediators. Chemotherapy-induced neuroinflammation affects central and peripheral nervous system and occurs through activation of both innate and adaptive immune system, including activation of satellite glial cells, Schwann cells, astrocytes, and microglia [32,33,34,35,36]. Exposure to chemotherapeutic agents were well-documented to cause increased levels of pro-inflammatory cytokines and mediators, including IL-6, IL-8, IL-1β, and TNF-α [37,38,39,40,41]. Aberrant pro-inflammatory state was also triggered by suppression of regulatory T (T-reg) cells, further exacerbating the inflammation [42, 43]. In addition, chemotherapeutic agents like vincristine were found to be able to activate CX3CR-macrophages, which leads to activate TRPA1, an ion channel responsible for pain sensation in humans. Vincristine and paclitaxel were also known to increased STAT3-CXCL12 gene promotor binding, inducing upregulation and binding of CXCL12 to CXCR4 which leads to increased chemotaxis and promoting inflammation [10].

The combination of oxidative stress and neuroinflammation also lead to axonal degeneration in the peripheral nervous system. Long-term administration of chemotherapeutic agents was found to damage large myelinated, small unmyelinated nerve fibers, as well as intra-epidermal nerve fibers (IENF). IENF are unmyelinated or thinly myelinated nociceptors responsible for sensation of pain arising from periphery. Exposure to chemotherapeutic agents have been shown to reduce IENF [44]. Although IENF density does not always correlate with duration and dose of chemotherapy, as well as the associated CIPN symptoms [45]. The variable outcomes of chemotherapeutic agents to IENF may be influenced by the methods of IENF quantification, demographics (including age and height of the corresponding innervated area), chemotherapy dose–response relationships, and the variable compensatory regenerative nerve response within the course of chemotherapy exposure [46], all of which had to be further delineated.

Nerve growth factor

Nerve growth factor (NGF) is a neurotrophic factor which promotes growth and survival of peripheral sensory nerve cells, sympathetic nerves, as well as the functional integrity of the cholinergic neurons in the central nervous system (CNS) [47, 48]. The seminal study by De Santis and colleagues [49]. had discovered lower circulating NGF levels among subjects receiving platinum-based and taxane-based chemotherapy, by which some patients had undetectable NGF levels after prolonged treatment. Furthermore, the study also found that NGF levels decrement correlated with the severity of chemo-induced toxicity. Similarly, several studies also confirmed these findings. Subjects who received chemotherapy were generally found to have lower NGF levels when compared to control. This comprises those with hematological malignancies who had received bortezomib, thalidomide, and vincristine, or those with multiple myeloma who had received bortezomib [50, 51]. In a study involving women with cervical cancer receiving paclitaxel and cisplatin, with or without ifosfamide, NGF levels had a significant negative correlation with the severity of CIPN [52]. In addition, a clinical study involving 60 subjects with colorectal, gastric, or lung cancer receiving taxane, platinum or bortezomib chemotherapy demonstrated increased NGF levels among those with painful CIPN [53]. Moreover, those subjects did not demonstrate significant loss of intra-epidermal nerve fiber density, suggesting a weak correlation between structural nerve fiber changes and NGF levels. However, the study did show a different NGF trajectory level (high vs. low in the previously mentioned studies), partly of which could be explained by the inclusion of mixed chemotherapeutic agents used (including oxaliplatin), different measurement time points (putting coasting phenomenon into consideration), and different criteria for establishing CIPN severity.

In addition to functioning as a screening or diagnostic tool for CIPN, NGF levels were also reported to be higher after vitamin B administration among subjects who suffered from peripheral neuropathy after bortezomib therapy when compared with control groups [51], suggesting its effectiveness as a biomarker for monitoring treatment success and perhaps, prognosis.

The possible mechanistic interaction of NGF and CIPN was perhaps due to its high affinity to NGF receptor, tropomyosin-related kinase receptor A (TrkA) which are richly expressed in the dorsal root ganglion (DRG) neurons, which in turn able to regulate the impact of chemotherapy-induced neuronal toxicity [9, 54]. Nevertheless, the pragmatic use of NGF as a blood-based biomarker for CIPN requires further studies, particularly those which attempt to delineate the mechanistic and temporal relationships between the two, with a precise and standardized diagnostic criteria of CIPN, along with its progression.

Brain-derived neurotrophic factor

Brain-derived neurotrophic factor (BDNF) is also an NGF that possesses a central role in the development, maintenance, and repair of the CNS and the peripheral nervous system (PNS). BDNF is synthesized exclusively in humans and specifically binds to tropomyosin-related kinase receptor B (TrkB) and nonselective low-affinity TNF-α-related p75 (p75 NTR) neurotrophin receptor. There are few clinical studies evaluating the relationship of BDNF and CIPN. One study evaluated 91 multiple myeloma patients receiving bortezomib and/or thalidomide and found a significant correlation between BDNF levels and the severity of polyneuropathy and BDNF levels also determined chemotherapeutic treatment responses. Interestingly, this study provided a diagnostic sensitivity and specificity of BDNF levels for diagnosing CIPN (76 and 71%, respectively) [55]. Another study involved 45 multiple myeloma patients receiving vincristine or bortezomib and have their BDNF levels and single nucleotide polymorphism (SNP) for valine-to-methionine substitution at codon 66 (Val66Met) checked, along with an established diagnosis of CIPN via Total Neuropathy Score (TNSr) and FACT-GOG-NTx [56]. The study found that low pre-treatment serum BDNF levels significantly correlated with CIPN development and vice versa, and that development of CIPN was associated with depression among subjects with Met/Met genotypes. Interestingly, authors believed that Val66Met did not affect BDNF signaling but impaired cellular distribution and dysregulation of BDNF [56, 57]. Furthermore, platelet’s BDNF secretion was thought to be impaired by chemotherapeutic agent such as bortezomib, thus reducing its capacity to repair the damaged peripheral nerves [58, 59]. In addition, the administration of pro-neurotrophic agent such as 2-pentadecyl-2-oxazoline of palmitoylethanolamide (PEA-OXA) to rats with oxaliplatin-induced peripheral neuropathy was able to reduced hyperactivation of glial cells, increased production of pro-inflammatory cytokines in the dorsal horn of spinal cord, and upregulation of neurotrophic factors (including BDNF) in the DRG [60]. The authors believed that increased BDNF levels was associated with structural as well as functional improvements in CIPN. Hence, BDNF levels can potentially be used as a screening tool, prognostication, and to monitor disease progression and therapeutic benefits.

However, again, the potential use of BDNF as a blood-based biomarker for CIPN warrants further studies as there was a discrepant association between BDNF levels and the severity of CIPN symptoms between two study groups [55, 56]. One author explained several underlying reasons for this discrepancy, comprising different cancer-dependent BDNF secretion levels (particularly those of hematologic vs. non-hematologic malignancies), different criteria used for the diagnosis of CIPN, and the absence of any objective measurements to address CIPN, such as quantitative sensory testing (QST) or alternatively, by using small-fiber testing using Q-sense device [61, 62]. An obscure temporal and mechanistic relationships between BDNF levels and the natural history of CIPN itself may preclude its definitive and routine use in CIPN, be it as a screening tool, prognostication, or to monitor disease progression and therapeutic benefits.

Neurofilament proteins

Neurofilaments (NF) are intermediate filaments primarily expressed by large, myelinated axons and will be secreted into the CSF during neuron or synaptic degeneration. NF can be classified into three categories according to its molecular weight, i.e., light (NF-L), medium, or heavy chain (NF-H). NF levels can be detected on ultra-low concentration in the blood or corresponding injured tissue, thus ensuring its sensitivity [9]. NF-L has been studied in animal model, that is using rat model exposed to vincristine. NF-L levels of rats exposed to vincristine demonstrated a fourfold increase as opposed to controls, in parallel with signs of axonopathy and IENF loss [63]. Another study has also confirmed these findings, wherein exposure to cisplatin and paclitaxel increased NF-L levels and that its increment was associated with severity of morphological and functional alteration of axonal structure, suggesting its reliability in detecting CIPN and linear association between CIPN signs and symptoms as well as its morphological changes [64]. In addition, NF-L has also been studied among human patients receiving oxaliplatin and monitored for oxaliplatin-induced peripheral neuropathy (OAIPN). NF-L levels were significantly higher among those with more severe OAIPN and that its levels markedly decrease 6 months after cessation of chemotherapy [65]. Another study also observed NF-L levels to increase linearly with CIPN severity and that its levels are markedly lower among asymptomatic individuals as opposed to those with active symptoms, indicating the dynamic and linear association of NF-L levels with disease progression and severity, thus highlighting the usefulness of NF-L to detect early CIPN and monitor the ongoing neuroaxonal injury [66]. However, it was reported that NF-L had poor accuracy in defining CIPN according to one clinical study involving breast cancer patients treated with chemotherapy [67]. The authors suggested that CIPN involves a more distal portion of peripheral nerves rich in dendrites as opposed to axons. Regardless, further studies are required to fully elucidate NF roles and natural responses in the event of CIPN (Fig. 2).

Fig. 2
figure 2

Multiple candidate biomarkers potentially used to aid screening, diagnosis, to monitor disease progression, treatment response, and prognostication of CIPN [49, 55, 65, 68,69,70]

MicroRNAs

MicroRNAs (miRNAs) are endogenous, single-stranded small RNA with length of approximately 22 nucleotides which function as antisense RNA to regulate target genes post-transcriptionally [71]. The association between miRNAs and genes are somewhat minute but significant as a single gene can be simultaneously targeted by several miRNAs, whereas a single miRNA may target multiple genes [72]. MiRNAs consist of a highly complex network with their gene targets [71]. MiRNAs’ active role has been found in both physiological as well as pathological processes, including cancer [73]. Many studies have confirmed the extent of miRNAs involvement in various aspects of cancer, including its proliferation, differentiation, apoptosis, metabolism, invasiveness, metastatic preponderance, and resistance to chemotherapy [68].

The potential use of miRNAs for the diagnosis of CIPN was studied. An in vivo animal model with paclitaxel-induced CIPN demonstrated significantly increased levels of miR-124 (i.e., 5- and 10-fold on day 8 and 16, respectively) [74]. The increased miR-124 expression levels were associated with cold allodynia and axonal degeneration of DRG and sciatic nerve, indicating its significant role in the pathogenesis of CIPN. Indeed, miR-124 is one of the many miRNAs associated with neuronal cell activity, wherein its plasma levels increment was reported in CNS injury, such as stroke [75]. In addition, miR-124 was also shown to regulate microglial function and that miRNA-124-3p was able to attenuate allodynia and thermal hyperalgesia by means of regulating Egr1 expression in the DRG and dorsal horn of spinal cord [76, 77].

There was one clinical study involving 84 patients with severe and mild CIPN after receiving paclitaxel for breast cancer [78]. There were 15 miRNAs with |fold change| > 0.5, of which miR-451a demonstrated the highest fold change, although statistically insignificant (P = 0.103). MiR-451a is associated with metabolism of paclitaxel, i.e., regulating the expression of drug transporter protein (P-glycoprotein), thus plays a central role in resistance to chemotherapy. Furthermore, there were 14 combinations of three miRNAs ranging in accuracy of 50 to 78.6%. However, the potential use of miRNAs as CIPN biomarker is rather a long shot due to multiple reasons. Firstly, most of the identified miRNAs did not have high fold change, not to mention its statistical insignificance. This is reasonable since most of them do not possess direct mechanistic association with CIPN. For instance, among the 14 miRNAs, one of them was responsible for angiogenesis and another was useful in distinguishing early-stage pancreatic cancers, a somewhat typical loose association and rather generic role of many miRNAs. Secondly, validation of significant miRNAs requires a large pool of sample size and should have enough discriminatory powers with respect to demographics, health and disease status, and other confounding factors [71]. Thirdly, there should be a clear establishment of different types of miRNAs involved within the natural history continuum of CIPN. For instance, there were roughly 25 different miRNAs clustering in early, progressing, and late non-small cell lung cancer at diagnosis, treatment selection, and its prognosis [71]. The last criterion is critically important to ensure the accurate and prudent use of miRNA as a screening tool, prognostication, or to monitor disease progression and therapeutic benefits.

Various attempts have been made to bolster the diagnostic power of miRNAs toward certain diseases. For instance, there is an ongoing trend to combine several miRNAs or incorporation with other classic biomarkers. Others utilized bioinformatics or computational approach to better sort and calculate the complex array of miRNAs and its associated target genes. In fact, bioinformatics approach was used to address significant genes involved in platinum-induced neuropathy and combined taxane–platinum-induced neuropathy according to its number of connections.

Quantitative sensory testing (QST)

QST is a standardized psychophysical test to measure multiple sensations, including vibration sensation, mechanical detection thresholds, and thermal (cold and heat) detection thresholds [79]. QST has the upper hand of being non-invasive, objective, and yields personalized results for each individual. QST is also able to detect subclinical pathological changes that might otherwise be regarded as normal when tested using a more conventional electrodiagnostic approach, such as nerve conduction studies [80]. The latter is particularly relevant in the case of small-fiber neuropathy, including CIPN, primarily in the early phase of the disorder [81, 82]. There were numerous studies employing QST for the purpose of CIPN detection. For instance, one study evaluated two clinical trials involving solid tumor survivors had found that those with moderate-to-severe CIPN had markedly impaired tactile, vibratory, and thermal thresholds compared to patients without CIPN, and that patient-reported outcome was correlated with QST results [69]. Another study evaluated the impact of oxaliplatin on peripheral neuropathy with QST and found prolonged pegboard test completion time, elevated bumps detection threshold, increased touch detection threshold, and increased warm detection threshold among those with increasing cumulative dose of oxaliplatin [83]. The diagnostic profiles of each QST component varies considerably, depending on the type of chemotherapy used, measurement time point, and the methods employed for QST. For instance, vibration detection threshold measured among subjects receiving oxaliplatin to detect clinically significant neuropathy 6 months after treatment demonstrated sensitivity and specificity of 76% and 53%, respectively [84]. Another study which evaluated the impact of oxaliplatin and cisplatin treatment among colorectal cancer patients using QST found that a change of −0.05 °C of cold detection threshold had a sensitivity and specificity of 92.3% and 64.9% in detecting CIPN after 6 months of chemotherapeutic treatment, whereas a change of −0.85 °C of heat detection threshold yielded a sensitivity and specificity of 64.3% and 70%, respectively [85].

In addition, QST can also be used to distinguish different types of CIPN, mainly neuropathic vs. non-neuropathic pain. For instance, one study evaluated the sensory phenotypes of taxane-induced peripheral neuropathy (TIPN) among breast cancer patients [86]. Those with neuropathic pain symptoms had normalized heat pain thresholds when examined using QST, thus indicating retained C-fiber and TRPV1 function. Recently, one study successfully utilized diode laser fiber-type selective stimulator (DLss) to selectively stimulate C and Aδ fibers, as determined by C/Aδ ratio, to detect painful CIPN on its earliest phase [87]. The resulting classification based on CIPN symptoms may potentially be used to monitor disease progression, determining the temporal relationship between symptoms with pathological process of CIPN in various time points, and to monitor treatment response and prognosis. In fact, several studies had incorporated QST profiles with patients’ response to medications for various types of neuropathic pain [88, 89].

However, the use of QST may be used with caution due to the wide normal range (thus increasing the false-negative rate), highly variable measurement methods, requires full patient cooperation and attention (inattentive individuals may increase erroneous results), as well as inability to distinguish malingering cases [90, 91]. Considering CIPN, there should be a well-defined diagnostic criterion, both in terms of clinical symptoms and QST thresholds for each of its component.

Nerve conduction studies

The use of conventional nerve conduction studies (NCS) has generally been limited by low sensitivity in the case of small-fiber neuropathies such as CIPN. It is obviously due to the relative nature of the small nerve diameter, non-myelinated, with miniscule axonal dimension resulting in too low of conduction velocities and amplitudes to be able to be detected by the NCS device [92]. However, NCS when applied appropriately, may yield important cues to nerve damage associated with CIPN, albeit with varying degree of significance. Several studies had found early SNAP decrease preceded CIPN among asymptomatic individuals with prior exposure to chemotherapy [93,94,95]. In fact, one observational study using NCS confirmed that a more severe damage was found among sensory than motor nerves among CIPN patients at different stages of chemotherapy, although it had a low association with clinical symptoms [96]. Another study investigated the impact of platinum and taxane-based chemotherapy toward CIPN using NCS and found markedly lower amplitude of SNAP and CMAP, with platinum-based chemotherapy caused a more profound CMAP decrement of ulnar and tibial nerve [97]. However, when combined with clinical symptoms (i.e., using total Neuropathy Score), the model did not show statistical significance. Moreover, one study from Japan took even further steps that it used point-of-care NCS, a relatively quick and easy-to-use device to do NCS which found significant decrease of SNAP and preservation of conduction velocities as CIPN progressed [98], further confirmed axonal degeneration resulting from chemotherapy exposure. The potential of NCS in detecting CIPN is thus worth revisiting and might be combined with other related modalities such as QST to yield better diagnostic sensitivity.

Imaging biomarkers

Neuroimaging can potentially be used as a biomarker for CIPN. One can measure the cross-sectional area (CSA) of the affected nerve by using high-resolution ultrasound (HRUS) [99]. One study evaluated the effect of taxane chemotherapy against the CSA of tibial motor nerve, median motor nerve, and sural sensory nerve and compared the results with healthy controls. The study also examined the intra-epidermal nerve fiber density (IENFD) of the corresponding patients. It was found on HRUS that sural nerve of those who had received chemotherapy was 1.2 mm2 smaller than healthy controls, and that decreased nerve CSA was associated with decreased IENFD by a ratio of 1 mm2: 2.1 nerve/mm [70]. On the contrary, one study which evaluated the impact of oxaliplatin, irinotecan, and 5-fluorouracil (FOLFIRINOX) had found an increased CSA of tibial and fibular nerve using HRUS, and that there were no correlations between CSA size, electrophysiologic findings, or clinical severity [100]. The increased CSA at entrapment sites may indicate oxaliplatin-induced nerve susceptibility to mechanical damage, even when there was no neurophysiologic evidence of entrapment [101].

Magnetic resonance neurography (MRN) had also been evaluated as a potential neuroimaging biomarker for CIPN. It has been previously used as a biomarker for other peripheral neuropathies, including diabetic neuropathy and HIV-associated neuropathy [102]. There was a significant DRG hypertrophy in oxaliplatin-induced peripheral neuropathy patients as measured using MRN and that it correlated with the sensory neuropathy [103]. Recently, diffusion tensor imaging (DTI) was employed for early diagnosis of CIPN, demonstrating correlation between fractional anisotropy (FA) and apparent diffusion coefficient (ADC) at medial and left plantar nerve [104]. The evidence for MRN is robust on proof-of-concept ground, although its integration into clinical practice requires further studies.

Artificial intelligence and machine learning

Artificial intelligence (AI) has been extensively incorporated and studied for the advancements of diagnosis and treatment of various disorders. In the field of cancer, AI has been proven useful to aide chemotherapeutic drug research and development (e.g., identifying new drug compounds or improving drug delivery effectiveness and its permeability), predicting treatment response, as well as assisting in clinical decision support system [105]. Recently, AI also has shown promise in the detection and prevention of CIPN prior to drug exposure. For example, one study used machine learning-based quantitative-structure toxicity relationship models, i.e., computational models to detect chemotherapeutic drug toxicity (i.e., CIPN) among novel antineoplastic drug candidates with satisfactory results, and the system even able to stratify drugs’ risk (high, medium, low) in inducing CIPN [106]. Another study used machine learning using input from electronic health records data and compared three algorithms to develop prediction models and found that logistic regression yielded the highest and sufficient AUC (0.62–0.83) compared to decision tree and artificial neural network [107].

Conclusions

Given the account of the relatively high prevalence of CIPN and absence of any predictive biomarker for the purpose of early diagnosis, prognostication, or to monitor disease progression and therapeutic benefits, the existence of such biomarkers is regarded highly necessary. There are several problems, at least in our sight, that needs to be addressed. Firstly, there should be a standardized definition and diagnostic criteria for CIPN, whereby there are a plethora of discrepancies in defining CIPN, including the use of multiple patient- or examiner-based questionnaires, lack of nerve conduction study protocols for CIPN and the resulting underutilization of the examination thereof. Inability to define CIPN universally has been proven to be problematic as it encourages varying research methods and, thus varying outcomes, resulting in inconclusive findings.

Secondly, there should be a clear temporal and mechanistic relationships between various chemotherapeutic agents in inducing CIPN, along with the candidate biomarker profiles during the natural course of CIPN. For instance, there has not been a clear pathomechanism regarding the coasting phenomenon induced by platinum-based chemotherapy, or the contradictory rise and fall of nerve growth factors, including NGF and BNDF, along the course of CIPN and that by artificially replacing these growth factors (e.g., NGF) during the depletion phase was proven to be ineffective in ameliorating CIPN or worse, even exacerbated the pain severity, or the variable impacts of multiple chemotherapeutic agents with its corresponding dose–response relationship toward IENFD, and that NGF levels do not correlate with IENFD in spite of both reflects nerve degeneration and regeneration phases. These first two factors needed to be addressed simultaneously by conducting longitudinal clinical studies which attempt to incorporate and characterize potential biomarker candidates along with the natural history of CIPN which was preceded by a confirmed diagnosis using universally accepted diagnostic criteria. In fact, one longitudinal clinical multicenter study called Genetics and Inflammatory Markers for CIPN (GENIE) is attempting to solve this issue by incorporating genomic, metabolites, DNA methylation, gene expression, cytokines, and chemokines before, during, and 12 months after taxane treatment among 400 patients with breast cancer. The combined biomarker monitoring results will be compared with multiple CIPN assessment tools, including CTCAE, EORTC CIP20, BPI-SF, and PROMIS. Given the broad spectrum nature of the study, it hopefully may be able to capture the natural history of CIPN along with association to multiple biomarkers of each stage more robustly. Moreover, the authors employed machine learning to detect non-linear relationships among variables, thus reinforcing the diagnostic and prognostic sensitivity of the study [108] (Fig. 3).

Fig. 3
figure 3

Several critical key points needed to be addressed to better diagnose and treat CIPN

Diagnosing CIPN requires a multidimensional approach, by which it incorporates the subjective symptoms, clinical signs (e.g., reduced vibratory perception or the presence of motor weakness), supporting electrodiagnostic findings, and, potentially, quantitative biomarkers. CIPN diagnosis also requires a thorough understanding of its natural history and phases, thus encouraging one to put CIPN into a disease continuum. The diagnostic approach, therefore, should follow the path of those neurodegenerative diseases, like Alzheimer’s disease (AD) for example, and less of toward disease with simple diagnostic cut-off using blood-based biomarker, such as diabetes mellitus, wherein diagnostic confirmation, monitoring treatment response, as well as prognostication can be done using a clear cut-off point of plasma glucose levels. In AD, the currently accepted diagnostic criteria established by National Institute on Aging (NIA) and the Alzheimer’s Association (AA) has already incorporated biomarkers including neuroimaging-based (MRI or PET scan [amyloid, Tau, or FDG]) and bodily fluid-based (CSF beta-amyloid levels), as well as the classification of AD diagnosis based on disease continuum, from preclinical to dementia due to AD [109, 110]. Although the corresponding diagnostic criteria is reserved to facilitate research, there is an ongoing trend to incorporate it into routine clinical practice [111]. Such an approach has the upper hand to study the natural history of AD in a complete picture, the temporal and mechanistic relationships of biomarker candidates along AD natural course, standardizing clinical study outcomes, from biomarkers to clinical trials, and potentially accelerating the findings of disease-modifying drugs with clinical significance. Given the dramatic progress in AD research in recent years, for instance, marked by the first ever approval of disease-modifying monoclonal antibodies, aducanumab for the treatment of AD by FDA [109], regardless of its controversy [112], it is prudent to assume that one of which may be contributed by the utilization of NIA-AA AD diagnostic criteria. In our opinion, the same approach should also be applied to CIPN.

Availability of data and materials

Not applicable.

Abbreviations

CIPN:

Chemotherapy-induced peripheral neuropathy

microRNAs:

Micro ribonucleic acids

QST:

Quantitative sensory testing

DNA:

Deoxyribonucleic acid

ROS:

Reactive oxygen species

DRG:

Dorsal root ganglion

mDNA:

Mitochondrial DNA

IL:

Interleukin

TNF-α:

Tumor necrosis factor alpha

T-reg:

Regulatory T

CX3CR:

Chemokine receptor type 3

TRPA1:

Transient receptor potential cation channel, subfamily A, member 1

CXCL12:

C-X-C motif chemokine ligand 12

CXCR4:

Chemokine receptor type 4

IENF:

Intra-epidermal nerve fiber

NGF:

Nerve growth factor

CNS:

Central nervous system

TrkA:

Tropomyosin-related kinase receptor A

BDNF:

Brain-derived neurotrophic factor

PNS:

Peripheral nervous system

TrkB:

Tropomyosin-related kinase receptor B

p75 NTR:

Low-affinity TNF-α-related p75

SNP:

Single nucleotide polymorphism

TNSr:

Total Neuropathy Score

PEA:

Palmitoylethanolamide

NF:

Neurofilament

OAIPN:

Oxaliplatin-induced peripheral neuropathy

CSA:

Cross-sectional area

HRUS:

High-resolution ultrasound

IENFD:

Intra-epidermal nerve fiber density

MRN:

Magnetic resonance neurography

ADC:

Apparent diffusion coefficient

AD:

Alzheimer’s disease

References

  1. Seretny M, et al. Incidence, prevalence, and predictors of chemotherapy-induced peripheral neuropathy: a systematic review and meta-analysis. Pain. 2014;155:2461–70.

    Article  PubMed  Google Scholar 

  2. ZajÄ…czkowska R, et al. Mechanisms of chemotherapy-induced peripheral neuropathy. Int J Mol Sci. 2019;20:1451.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Brown TJ, Sedhom R, Gupta A. Chemotherapy-induced peripheral neuropathy. JAMA Oncol. 2019;5:750.

    Article  PubMed  Google Scholar 

  4. Kolb NA, et al. The association of chemotherapy-induced peripheral neuropathy symptoms and the risk of falling. JAMA Neurol. 2016;73:860–6.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Beijers A, Mols F, Dercksen W, Driessen C, Vreugdenhil G. Chemotherapy-induced peripheral neuropathy and impact on quality of life 6 months after treatment with chemotherapy. J Community Support Oncol. 2014;12:401–6.

    Article  CAS  PubMed  Google Scholar 

  6. Mols F, Beijers T, Vreugdenhil G, van de Poll-Franse L. Chemotherapy-induced peripheral neuropathy and its association with quality of life: a systematic review. J Multinatl Assoc Support Care Cancer. 2014;22:2261–9.

    Article  Google Scholar 

  7. Loprinzi CL, et al. Prevention and management of chemotherapy-induced peripheral neuropathy in survivors of adult cancers: ASCO guideline update. J Clin Oncol. 2020;38:3325–48.

    Article  PubMed  Google Scholar 

  8. Bonhof CS, Trompetter HR, Vreugdenhil G, van de Poll-Franse LV, Mols F. Painful and non-painful chemotherapy-induced peripheral neuropathy and quality of life in colorectal cancer survivors: results from the population-based PROFILES registry. Support Care Cancer. 2020;28:5933–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Meregalli C, Bonomo R, Cavaletti G, Carozzi VA. Blood molecular biomarkers for chemotherapy-induced peripheral neuropathy: from preclinical models to clinical practice. Neurosci Lett. 2021;749: 135739.

    Article  CAS  PubMed  Google Scholar 

  10. Starobova H, Vetter I. Pathophysiology of chemotherapy-induced peripheral neuropathy. Front Mol Neurosci. 2017;10:174.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Eldridge S, Guo L, Hamre J. A comparative review of chemotherapy-induced peripheral neuropathy in in vivo and in vitro models. Toxicol Pathol. 2020;48:190–201.

    Article  PubMed  Google Scholar 

  12. Ferrier J, Pereira V, Busserolles J, Authier N, Balayssac D. Emerging trends in understanding chemotherapy-induced peripheral neuropathy. Curr Pain Headache Rep. 2013;17:364.

    Article  PubMed  Google Scholar 

  13. Naik AK, et al. Role of oxidative stress in pathophysiology of peripheral neuropathy and modulation by N-acetyl-L-cysteine in rats. Eur J Pain Lond Engl. 2006;10:573–9.

    Article  CAS  Google Scholar 

  14. Yowtak J, et al. Reactive oxygen species contribute to neuropathic pain by reducing spinal GABA release. Pain. 2011;152:844–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Allen DT, Kiernan JA. Permeation of proteins from the blood into peripheral nerves and ganglia. Neuroscience. 1994;59:755–64.

    Article  CAS  PubMed  Google Scholar 

  16. Gorgun MF, Zhuo M, Englander EW. Cisplatin toxicity in dorsal root ganglion neurons is relieved by meclizine via diminution of mitochondrial compromise and improved clearance of DNA damage. Mol Neurobiol. 2017;54:7883–95.

    Article  CAS  PubMed  Google Scholar 

  17. Addington J, Freimer M. Chemotherapy-induced peripheral neuropathy: an update on the current understanding. F1000Res. 2016. https://doi.org/10.12688/f1000research.8053.1.

    Article  PubMed  PubMed Central  Google Scholar 

  18. Zhang H, et al. Dorsal root ganglion infiltration by macrophages contributes to paclitaxel chemotherapy-induced peripheral neuropathy. J Pain. 2016;17:775–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kesserwani H. Death and rebirth of the thalidomide molecule: a case of thalidomide-induced sensory neuropathy. Cureus. 2021;13: e13140.

    PubMed  PubMed Central  Google Scholar 

  20. di Penta A, et al. Oxidative stress and proinflammatory cytokines contribute to demyelination and axonal damage in a cerebellar culture model of neuroinflammation. PLoS ONE. 2013;8: e54722.

    Article  PubMed  PubMed Central  Google Scholar 

  21. Solleiro-Villavicencio H, Rivas-Arancibia S. Effect of chronic oxidative stress on neuroinflammatory response mediated by CD4+T cells in neurodegenerative diseases. Front Cell Neurosci. 2018;12:114.

    Article  PubMed  PubMed Central  Google Scholar 

  22. He J, Zhu G, Wang G, Zhang F. Oxidative stress and neuroinflammation potentiate each other to promote progression of dopamine neurodegeneration. Oxid Med Cell Longev. 2020;2020:1–12.

    Google Scholar 

  23. Areti A, Yerra VG, Naidu V, Kumar A. Oxidative stress and nerve damage: role in chemotherapy induced peripheral neuropathy. Redox Biol. 2014;2:289–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Canta A, Pozzi E, Carozzi VA. Mitochondrial dysfunction in chemotherapy-induced peripheral neuropathy (CIPN). Toxics. 2015;3:198–223.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Chen X-J, Wang L, Song X-Y. Mitoquinone alleviates vincristine-induced neuropathic pain through inhibiting oxidative stress and apoptosis via the improvement of mitochondrial dysfunction. Biomed Pharmacother. 2020;125: 110003.

    Article  CAS  PubMed  Google Scholar 

  26. Kober KM, et al. Expression of mitochondrial dysfunction-related genes and pathways in paclitaxel-induced peripheral neuropathy in breast cancer survivors. Mol Pain. 2018;14:1744806918816462.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Mukhtar E, Adhami VM, Mukhtar H. Targeting microtubules by natural agents for cancer therapy. Mol Cancer Ther. 2014;13:275–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Malacrida A, Meregalli C, Rodriguez-Menendez V, Nicolini G. Chemotherapy-induced peripheral neuropathy and changes in cytoskeleton. Int J Mol Sci. 2019;20:E2287.

    Article  Google Scholar 

  29. LaPointe NE, et al. Effects of eribulin, vincristine, paclitaxel and ixabepilone on fast axonal transport and kinesin-1 driven microtubule gliding: implications for chemotherapy-induced peripheral neuropathy. Neurotoxicology. 2013;37:231–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Triarico S, et al. Vincristine-induced peripheral neuropathy (VIPN) in pediatric Tumors: mechanisms, risk factors, strategies of prevention and treatment. Int J Mol Sci. 2021;22:4112.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Staff NP, et al. Bortezomib alters microtubule polymerization and axonal transport in rat dorsal root ganglion neurons. Neurotoxicology. 2013;39:124–31.

    Article  CAS  PubMed  Google Scholar 

  32. Warwick RA, Hanani M. The contribution of satellite glial cells to chemotherapy-induced neuropathic pain. Eur J Pain Lond Engl. 2013;17:571–80.

    Article  CAS  Google Scholar 

  33. Leo M, Schmitt L-I, Kutritz A, Kleinschnitz C, Hagenacker T. Cisplatin-induced activation and functional modulation of satellite glial cells lead to cytokine-mediated modulation of sensory neuron excitability. Exp Neurol. 2021;341: 113695.

    Article  CAS  PubMed  Google Scholar 

  34. Ji X-T, et al. Spinal astrocytic activation contributes to mechanical allodynia in a rat chemotherapy-induced neuropathic pain model. PLoS ONE. 2013;8: e60733.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Liu X, Tonello R, Ling Y, Gao Y-J, Berta T. Paclitaxel-activated astrocytes produce mechanical allodynia in mice by releasing tumor necrosis factor-α and stromal-derived cell factor 1. J Neuroinflammation. 2019;16:209.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Lee JH, Kim W. The role of satellite glial cells, astrocytes, and microglia in oxaliplatin-induced neuropathic pain. Biomedicines. 2020;8:E324.

    Article  Google Scholar 

  37. Wang X-M, Lehky TJ, Brell JM, Dorsey SG. Discovering cytokines as targets for chemotherapy-induced painful peripheral neuropathy. Cytokine. 2012;59:3–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Brandolini L, d’Angelo M, Antonosante A, Allegretti M, Cimini A. Chemokine signaling in chemotherapy-induced neuropathic pain. Int J Mol Sci. 2019;20:E2904.

    Article  Google Scholar 

  39. Fumagalli G, Monza L, Cavaletti G, Rigolio R, Meregalli C. Neuroinflammatory process involved in different preclinical models of chemotherapy-induced peripheral neuropathy. Front Immunol. 2021;11: 626687.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Lees JG, et al. Immune-mediated processes implicated in chemotherapy-induced peripheral neuropathy. Eur J Cancer. 2017;73:22–9.

    Article  CAS  PubMed  Google Scholar 

  41. Starkweather A. Increased interleukin-6 activity associated with painful chemotherapy-induced peripheral neuropathy in women after breast cancer treatment. Nurs Res Pract. 2010;2010:1–9.

    Article  Google Scholar 

  42. Makker PGS, et al. Characterisation of immune and neuroinflammatory changes associated with chemotherapy-induced peripheral neuropathy. PLoS ONE. 2017;12: e0170814.

    Article  PubMed  PubMed Central  Google Scholar 

  43. Zhang L, et al. Differential impairment of regulatory T cells rather than effector T cells by paclitaxel-based chemotherapy. Clin Immunol. 2008;129:219–29.

    Article  CAS  PubMed  Google Scholar 

  44. Boyette-Davis J, Xin W, Zhang H, Dougherty PM. Intraepidermal nerve fiber loss corresponds to the development of taxol-induced hyperalgesia and can be prevented by treatment with minocycline. Pain. 2011;152:308–13.

    Article  CAS  PubMed  Google Scholar 

  45. Koskinen MJ, et al. Intraepidermal nerve fibre density in cancer patients receiving adjuvant chemotherapy. Anticancer Res. 2011;31:4413–6.

    CAS  PubMed  Google Scholar 

  46. Mangus LM, Rao DB, Ebenezer GJ. Intraepidermal nerve fiber analysis in human patients and animal models of peripheral neuropathy: a comparative review. Toxicol Pathol. 2020;48:59–70.

    Article  PubMed  Google Scholar 

  47. Aloe L, Rocco M, Balzamino B, Micera A. Nerve growth factor: a focus on neuroscience and therapy. Curr Neuropharmacol. 2015;13:294–303.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Aloe L, Rocco ML, Bianchi P, Manni L. Nerve growth factor: from the early discoveries to the potential clinical use. J Transl Med. 2012;10:239.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. De Santis S, et al. Patients treated with antitumor drugs displaying neurological deficits are characterized by a low circulating level of nerve growth factor. J Am Assoc Cancer Res. 2000;6:90–5.

    Google Scholar 

  50. Youk J, et al. Depletion of nerve growth factor in chemotherapy-induced peripheral neuropathy associated with hematologic malignancies. PLoS ONE. 2017;12: e0183491.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Yan M, et al. The effect of rat nerve growth factor combined with vitamin B on peripheral neuropathy in multiple myeloma patients. Hematology. 2020;25:264–9.

    Article  CAS  PubMed  Google Scholar 

  52. Cavaletti G, et al. Early predictors of peripheral neurotoxicity in cisplatin and paclitaxel combination chemotherapy. Ann Oncol. 2004;15:1439–42.

    Article  CAS  PubMed  Google Scholar 

  53. Velasco R, et al. Neuropathic pain and nerve growth factor in chemotherapy-induced peripheral neuropathy: prospective clinical-pathological study. J Pain Symptom Manage. 2017;54:815–25.

    Article  PubMed  Google Scholar 

  54. Fischer SJ, Podratz JL, Windebank AJ. Nerve growth factor rescue of cisplatin neurotoxicity is mediated through the high affinity receptor: studies in PC12 cells and p75 null mouse dorsal root ganglia. Neurosci Lett. 2001;308:1–4.

    Article  CAS  PubMed  Google Scholar 

  55. Szudy-Szczyrek A, et al. Serum brain-derived neurotrophic factor (BDNF) concentration predicts polyneuropathy and overall survival in multiple myeloma patients. Br J Haematol. 2020;191:77–89.

    Article  CAS  PubMed  Google Scholar 

  56. Azoulay D, Giryes S, Nasser R, Sharon R, Horowitz NA. Prediction of chemotherapy-induced peripheral neuropathy in patients with lymphoma and myeloma: the roles of brain-derived neurotropic factor protein levels and a gene polymorphism. J Clin Neurol. 2019;15:511.

    Article  PubMed  PubMed Central  Google Scholar 

  57. Egan MF, et al. The BDNF val66met polymorphism affects activity-dependent secretion of BDNF and human memory and hippocampal function. Cell. 2003;112:257–69.

    Article  CAS  PubMed  Google Scholar 

  58. Azoulay D, et al. Bortezomib-induced peripheral neuropathy is related to altered levels of brain-derived neurotrophic factor in the peripheral blood of patients with multiple myeloma. Br J Haematol. 2014;164:454–6.

    Article  CAS  PubMed  Google Scholar 

  59. Fujimura H, et al. Brain-derived neurotrophic factor is stored in human platelets and released by agonist stimulation. Thromb Haemost. 2002;87:728–34.

    Article  CAS  PubMed  Google Scholar 

  60. Campolo M, et al. PEA-OXA mitigates oxaliplatin-induced painful neuropathy through NF-κB/Nrf-2 axis. Int J Mol Sci. 2021;22:3927.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Azoulay D, Horowitz NA. Brain-derived neurotrophic factor as a potential biomarker of chemotherapy-induced peripheral neuropathy and prognosis in haematological malignancies; what we have learned, the challenges and a need for global standardization. Br J Haematol. 2020;191:17–8.

    Article  PubMed  Google Scholar 

  62. Hovaguimian A, Gibbons CH. Diagnosis and treatment of pain in small-fiber neuropathy. Curr Pain Headache Rep. 2011;15:193–200.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Meregalli C, et al. Neurofilament light chain as disease biomarker in a rodent model of chemotherapy induced peripheral neuropathy. Exp Neurol. 2018;307:129–32.

    Article  CAS  PubMed  Google Scholar 

  64. Meregalli C, et al. Neurofilament light chain: a specific serum biomarker of axonal damage severity in rat models of chemotherapy-induced peripheral neurotoxicity. Arch Toxicol. 2020;94:2517–22.

    Article  CAS  PubMed  Google Scholar 

  65. Kim S-H, et al. Serum neurofilament light chain levels as a biomarker of neuroaxonal injury and severity of oxaliplatin-induced peripheral neuropathy. Sci Rep. 2020;10:7995.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Burgess BL, Cho E, Honigberg L. Neurofilament light as a predictive biomarker of unresolved chemotherapy-induced peripheral neuropathy in subjects receiving paclitaxel and carboplatin. Sci Rep. 2022;12:15593.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Sumitani M, et al. Poor efficacy of the phosphorylated high-molecular-weight neurofilament heavy subunit serum level, a biomarker of axonal damage, as a marker of chemotherapy-induced peripheral neuropathy. Biomed Rep. 2016;4:758–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Cui M, et al. Circulating microRNAs in cancer: potential and challenge. Front Genet. 2019;10:626.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Zhi WI, et al. Characterization of chemotherapy-induced peripheral neuropathy using patient-reported outcomes and quantitative sensory testing. Breast Cancer Res Treat. 2021;186:761–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Lycan TW, et al. Neuromuscular ultrasound for taxane peripheral neuropathy in breast cancer. Muscle Nerve. 2020;61:587–94.

    Article  PubMed  Google Scholar 

  71. Wang H, Peng R, Wang J, Qin Z, Xue L. Circulating microRNAs as potential cancer biomarkers: the advantage and disadvantage. Clin Epigenetics. 2018;10:59.

    Article  PubMed  PubMed Central  Google Scholar 

  72. Xu P, et al. A systematic way to infer the regulation relations of miRNAs on target genes and critical miRNAs in cancers. Front Genet. 2020;11:278.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Bracken CP, Scott HS, Goodall GJ. A network-biology perspective of microRNA function and dysfunction in cancer. Nat Rev Genet. 2016;17:719–32.

    Article  CAS  PubMed  Google Scholar 

  74. Peng Q, et al. Circulating microRNA and automated motion analysis as novel methods of assessing chemotherapy-induced peripheral neuropathy in mice. PLoS ONE. 2019;14: e0210995.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Wang Z, et al. Plasma miR-124 Is a Promising candidate biomarker for human intracerebral hemorrhage stroke. Mol Neurobiol. 2018;55:5879–88.

    Article  CAS  PubMed  Google Scholar 

  76. Zhao J, He Z, Wang J. MicroRNA-124: a key player in microglia-mediated inflammation in neurological diseases. Front Cell Neurosci. 2021;15: 771898.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Jiang M, et al. MicroRNA-124-3p attenuates the development of nerve injury–induced neuropathic pain by targeting early growth response 1 in the dorsal root ganglia and spinal dorsal horn. J Neurochem. 2021;158:928–42.

    Article  CAS  PubMed  Google Scholar 

  78. Noda-Narita S, et al. Peripheral neuropathy from paclitaxel: risk prediction by serum microRNAs. BMJ Support Palliat. 2020. https://doi.org/10.1136/bmjspcare-2019-001900.

    Article  Google Scholar 

  79. Velasco R, Alemany M, Villagrán M, Argyriou AA. Predictive biomarkers of oxaliplatin-induced peripheral neurotoxicity. J Pers Med. 2021;11:669.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Reddy SM, et al. Quantitative sensory testing at baseline and during cycle 1 oxaliplatin infusion detects subclinical peripheral neuropathy and predicts clinically overt chronic neuropathy in gastrointestinal malignancies. Clin Colorectal Cancer. 2016;15:37–46.

    Article  PubMed  Google Scholar 

  81. Scott K, Simmons Z, Kothari MJ. A comparison of quantitative sensory testing with skin biopsy in small fiber neuropathy. J Clin Neuromuscul Dis. 2003;4:129–32.

    Article  PubMed  Google Scholar 

  82. Ridehalgh C, Sandy-Hindmarch OP, Schmid AB. Validity of clinical small-fiber sensory testing to detect small-nerve fiber degeneration. J Orthop Sports Phys Ther. 2018;48:767–74.

    Article  PubMed  Google Scholar 

  83. de Carvalho Barbosa M, et al. A quantitative sensory analysis of peripheral neuropathy in colorectal cancer and its exacerbation by oxaliplatin chemotherapy. Cancer Res. 2014;74:5955–62.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Krøigård T, et al. Oxaliplatin neuropathy: predictive values of skin biopsy, QST and nerve conduction. J Neuromuscul Dis. 2021;8:679–88.

    Article  PubMed  Google Scholar 

  85. Delmotte J, et al. Is quantitative sensory testing helpful in the management of oxaliplatin neuropathy? a two-year clinical study. Cancer Treat Res Commun. 2018;17:31–6.

    Article  CAS  PubMed  Google Scholar 

  86. Hammond EA, Pitz M, Lambert P, Shay B. Quantitative sensory profiles of upper extremity chemotherapy induced peripheral neuropathy: are there differences in sensory profiles for neuropathic versus nociceptive pain? Can J Pain. 2019;3:169–77.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Nemenov MI, et al. Characterization of patients with and without painful peripheral neuropathy after receiving neurotoxic chemotherapy: traditional quantitative sensory testing vs C-fiber and Aδ-fiber selective diode laser stimulation. J Pain. 2021. https://doi.org/10.1016/j.jpain.2021.11.011.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Demant DT, et al. The effect of oxcarbazepine in peripheral neuropathic pain depends on pain phenotype: a randomised, double-blind, placebo-controlled phenotype-stratified study. Pain. 2014;155:2263–73.

    Article  CAS  PubMed  Google Scholar 

  89. Baron R, et al. Peripheral neuropathic pain: a mechanism-related organizing principle based on sensory profiles. Pain. 2017;158:261–72.

    Article  PubMed  Google Scholar 

  90. Sommer C. Psychophysics: quantitative sensory testing in the diagnostic work-up of small fiber neuropathy. In: Hsieh ST, Anand P, Gibbons CH, Sommer C, editors. Small fiber neuropathy and related syndromes: pain and neurodegeneration. Singapore: Springer Singapore; 2019. p. 33–42.

    Chapter  Google Scholar 

  91. Shy ME, et al. Quantitative sensory testing: report of the therapeutics and technology assessment subcommittee of the american academy of neurology. Neurology. 2003;60:898–904.

    Article  CAS  PubMed  Google Scholar 

  92. Timmins HC, et al. Quantification of small fiber neuropathy in chemotherapy-treated patients. J Pain. 2020;21:44–58.

    Article  CAS  PubMed  Google Scholar 

  93. Velasco R, et al. Early predictors of oxaliplatin-induced cumulative neuropathy in colorectal cancer patients. J Neurol Neurosurg Psychiatry. 2014;85:392–8.

    Article  PubMed  Google Scholar 

  94. McHugh JC, Tryfonopoulos D, Fennelly D, Crown J, Connolly S. Electroclinical biomarkers of early peripheral neurotoxicity from oxaliplatin. Eur J Cancer Care (Engl). 2012;21:782–9.

    Article  CAS  PubMed  Google Scholar 

  95. Argyriou AA, et al. Peripheral neuropathy induced by administration of cisplatin- and paclitaxel-based chemotherapy. Could it be predicted? Support. J Multinatl Assoc Support Care Cancer. 2005;13:647–51.

    Article  Google Scholar 

  96. Wang M, Bandla A, Sundar R, Molassiotis A. The phenotype and value of nerve conduction studies in measuring chemotherapy-induced peripheral neuropathy: a secondary analysis of pooled data. Eur J Oncol Nurs Off J Eur Oncol Nurs Soc. 2022;60: 102196.

    Article  Google Scholar 

  97. Myftiu B, et al. Chemotherapy-induced peripheral neuropathy (CIPN) in patients receiving 4–6 cycles of platinum-based and Taxane-based chemotherapy: a prospective, single-center study from Kosovo. Med Sci Monit Int Med J Exp Clin Res. 2022;28: e937856.

    CAS  Google Scholar 

  98. Matsuoka A, et al. Quantitative assessment of chemotherapy-induced peripheral neurotoxicity using a point-of-care nerve conduction device. Cancer Sci. 2016;107:1453–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Alberti P. A review of novel biomarkers and imaging techniques for assessing the severity of chemotherapy-induced peripheral neuropathy. Expert Opin Drug Metab Toxicol. 2020;16:1147–58.

    Article  CAS  PubMed  Google Scholar 

  100. Pitarokoili K, et al. Prospective study of the clinical, electrophysiologic, and sonographic characteristics of oxaliplatin-induced neuropathy. J Neuroimaging. 2019;29:133–9.

    Article  PubMed  Google Scholar 

  101. Briani C, et al. Ultrasound assessment of oxaliplatin-induced neuropathy and correlations with neurophysiologic findings. Eur J Neurol. 2013;20:188–92.

    Article  CAS  PubMed  Google Scholar 

  102. Evans MC, et al. Magnetic resonance imaging as a biomarker in diabetic and HIV-associated peripheral neuropathy: a systematic review-based narrative. Front Neurosci. 2021;15: 727311.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Apostolidis L, et al. Morphological correlates of oxaliplatin induced peripheral neuropathy assessed by magnetic resonance neurography. J Clin Oncol. 2017;35:10092–10092.

    Article  Google Scholar 

  104. Gimber LH, et al. Diffusion tensor imaging of the ankle as a possible predictor of chemotherapy induced peripheral neuropathy: pilot study. Curr Probl Diagn Radiol. 2019;48:121–6.

    Article  PubMed  Google Scholar 

  105. Zhou J, Zeng ZY, Li L. Progress of artificial intelligence in gynecological malignant Tumors. Cancer Manag Res. 2020;12:12823–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Bloomingdale P, Mager DE. Machine learning models for the prediction of chemotherapy-induced peripheral neuropathy. Pharm Res. 2019;36:35.

    Article  PubMed  Google Scholar 

  107. On J, Park H-A, Yoo S. Development of a prediction models for chemotherapy-induced adverse drug reactions: a retrospective observational study using electronic health records. Eur J Oncol Nurs. 2022;56: 102066.

    Article  PubMed  Google Scholar 

  108. Sharma A, et al. A multimodal approach to discover biomarkers for taxane-induced peripheral neuropathy (TIPN): a Study protocol. Technol Cancer Res Treat. 2022;21:15330338221127168.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Jack CR, et al. NIA-AA research framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dement J Alzheimers Assoc. 2018;14:535–62.

    Article  Google Scholar 

  110. Jagust WJ. The changing definition of Alzheimer’s disease. Lancet Neurol. 2021;20:414–5.

    Article  PubMed  Google Scholar 

  111. Porsteinsson AP, Isaacson RS, Knox S, Sabbagh MN, Rubino I. Diagnosis of early Alzheimer’s disease: clinical practice in 2021. J Prev Alzheimers Dis. 2021. https://doi.org/10.14283/jpad.2021.23.

    Article  PubMed  Google Scholar 

  112. Alexander GC, et al. Revisiting FDA approval of aducanumab. N Engl J Med. 2021;385:769–71.

    Article  CAS  PubMed  Google Scholar 

Download references

Funding

This study did not receive any funding.

Author information

Authors and Affiliations

Authors

Contributions

A.S. and I.P.E.W. drafted the manuscript. I.P.E.W. proofread the final draft of the manuscript, A.S. formatted the manuscript in accordance to The Egyptian Journal of Neurology, Psychiatry, and Neurosurgery style.

Corresponding author

Correspondence to Andreas Soejitno.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

All authors had no competing of interest to disclose.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Soejitno, A., Widyadharma, I.P.E. Chemotherapy-induced peripheral neuropathy biomarkers: current updates, challenges, and potentials. Egypt J Neurol Psychiatry Neurosurg 60, 92 (2024). https://doi.org/10.1186/s41983-024-00869-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s41983-024-00869-x

Keywords