Next Article in Journal
Differences in Children and Adolescents with Depression before and after a Remediation Program: An Event-Related Potential Study
Previous Article in Journal
Optimizing and Predicting Antidepressant Efficacy in Patients with Major Depressive Disorder Using Multi-Omics Analysis and the Opade AI Prediction Tools
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Centralizing the Knowledge and Interpretation of Pain in Chemotherapy-Induced Peripheral Neuropathy: A Paradigm Shift towards Brain-Centric Approaches

by
Mário Cunha
1,
Isaura Tavares
1,2,* and
José Tiago Costa-Pereira
1,2,3
1
Department of Biomedicine, Unit of Experimental Biology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
2
I3S—Institute of Investigation and Innovation in Health, University of Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
3
Faculty of Nutrition and Food Sciences, University of Porto, Rua do Campo Alegre 823, 4150-180 Porto, Portugal
*
Author to whom correspondence should be addressed.
Brain Sci. 2024, 14(7), 659; https://doi.org/10.3390/brainsci14070659
Submission received: 14 May 2024 / Revised: 17 June 2024 / Accepted: 24 June 2024 / Published: 28 June 2024
(This article belongs to the Section Neuroscience of Pain)

Abstract

:
Chemotherapy-induced peripheral neuropathy (CIPN) is a side effect of cancer treatment, often linked with pain complaints. Patients report mechanical and thermal hypersensitivity that may emerge during chemotherapy treatment and may persist after cancer remission. Whereas the latter situation disturbs the quality of life, life itself may be endangered by the appearance of CIPN during cancer treatment. The causes of CIPN have almost entirely been ascribed to the neurotoxicity of chemotherapeutic drugs in the peripheral nervous system. However, the central consequences of peripheral neuropathy are starting to be unraveled, namely in the supraspinal pain modulatory system. Based on our interests and experience in the field, we undertook a review of the brain-centered alterations that may underpin pain in CIPN. The changes in the descending pain modulation in CIPN models along with the functional and connectivity abnormalities in the brain of CIPN patients are analyzed. A translational analysis of preclinical findings about descending pain regulation during CIPN is reviewed considering the main neurochemical systems (serotoninergic and noradrenergic) targeted in CIPN management in patients, namely by antidepressants. In conclusion, this review highlights the importance of studying supraspinal areas involved in descending pain modulation to understand the pathophysiology of CIPN, which will probably allow a more personalized and effective CIPN treatment in the future.

1. Introduction

Better methods in cancer detection and the increased success of cancer treatment have led to a growing number of patients undergoing chemotherapy and an increased number of cancer survivors. Among the challenges of cancer treatment, chemotherapy-induced peripheral neuropathy (CIPN) has emerged as a significant and debilitating side effect, which may occur during cancer treatment and persist after cancer remission [1,2]. The prevalence of CIPN after chemotherapy ranges from 30% to 68% with significant variability in severity among individuals [2]. In comparison with patients that do not develop CIPN, on average patients with CIPN represent an additional demand on health services since they have the need for 12 additional outpatient visits, 3 additional days in hospital, and incur an extra USD 17,000 in medical expenses [3]. This highlights the profound impact of CIPN on the patients’ physical, social, emotional, functional, financial, and occupational well-being. The probability of developing CIPN is related to the type of chemotherapeutic drug and treatment protocol, namely the cumulative doses [2,4,5]. CIPN results from peripheral nerve injury caused by chemotherapy drugs, since it has the ability to damage peripheral nerve fibers, especially those involved in sensory and autonomic functions [6]. Small-diameter nerve fibers, unmyelinated or thin myelinated fibers responsible for conveying nociceptive information, are particularly susceptible to chemotherapeutic agents [7]. While extensive research has been conducted on the impact of CIPN on peripheral nerves [8,9], recent studies have begun to explore the consequences of peripheral damage on the spinal cord and supraspinal structures, specifically those involved in top-down pain modulation.
The descending pain modulatory system is a network of intrinsically connected brain areas that exerts a bidirectional balance (inhibition or facilitation) of the nociceptive inputs arising from the spinal cord [10,11,12]. Despite the neurochemical complexity of the system, the neurotransmitters released at the spinal cord during top-down modulation are mainly monoamines and opioids [13]. The proper functioning of the descending pain modulatory systems is essential to maintain the balance between inhibition and facilitation. Neuroplastic changes in the descending modulatory pathways are well documented in both human and animal models of neuropathic pain [14]. These changes disrupt the balance exerted by descending modulatory circuits, enhancing facilitation over inhibition [15,16]. That imbalance has been proposed to account for the development and persistence of neuropathic pain, namely of traumatic origin [15,16]. The role of brain-mediated pain modulation has been much less addressed in CIPN; despite the dramatically growing prevalence, this is a condition due to the increased number of patients undergoing chemotherapy. In clinical settings, patients with CIPN are often treated with antidepressants which raise the levels of serotonin (5-HT)/noradrenaline (NA) in the central nervous system (CNS) [7,17]. However, the mechanisms underlying the use of those drugs in CIPN are just starting to be unraveled. Furthermore, advanced neuroimaging tools available for both preclinical and clinical models, are important translational tools [18]. Filling the information gap between preclinical and clinical studies offers an opportunity to fundamentally change our understanding of CIPN by investigating the role of the CNS in its pathogenesis and developing novel treatment approaches.
This narrative review provides an overview of the central mechanisms of CIPN involving descending pain modulation, which can leverage the approach to managing and potentially preventing this condition.

2. Clinical Syndrome

In recent years, the number of cancer patients is increasing due to improved detection and better treatment of malignant conditions [19]. Cancer is now viewed as a chronic condition. While longer survival is a positive shift, it presents new challenges. Cancer survivors often deal with treatment side effects, like fatigue, emotional distress, and pain, in their post-treatment phase [1].
Chemotherapy drugs can harm the nervous system and lead to various types of neuropathies, including sensory, motor, and autonomic, affecting both large and small nerve fibers [20]. CIPN is a medical condition that refers to the damage or dysfunction of the peripheral nerves caused by chemotherapy [21]. The prevalence and incidence of CIPN vary widely depending on several factors, including the specific chemotherapy drugs used, the cumulative dose administered, and the individual characteristics of the patients [22]. Nevertheless, the individual features which may prompt a cancer patient to be more prone to develop CIPN have not been investigated in detail, and, herein, prevention is still not possible. Symptoms of CIPN can be acute or chronic. Acutely, CIPN occurs in 68% of patients and, if severe enough, it can require a reduction in the dose of chemotherapy or even stopping it before completing the planned protocol [2,4]. The prevalence of CIPN is estimated to be 68% in the first month, 60% after 3 months, and 30% after 6 months [2,4]. Since specific chemotherapeutic agents (e.g., cisplatin, paclitaxel) remain in the body for a long period [23], some patients experience paradoxical worsening and/or intensification of symptoms even after finishing the chemotherapy protocol, a phenomenon known as “coasting” [24,25]. This condition increases the financial burden on healthcare systems, prevents patients from working, and has a huge impact on the quality of life of cancer survivors [3,22].
The clinical presentation of CIPN varies greatly according to the type of chemotherapeutic drug, along with its dose, but factors such as the duration of chemotherapy treatment and assessment method are also important [26]. Sensory symptoms are the most common, but motor weakness, autonomic dysfunction, and cranial nerve involvement may occur [2,27]. Patients often describe sensory symptoms such as tingling, numbness, pressure, and paresthesia induced by touching warm or cool objects in a symmetric “glove and stocking” neuropathy pattern [28]. Although pain is not always a presenting feature [29], painful sensations, including spontaneous burning, shooting or electric shock-like pain, and mechanical or thermal allodynia/hyperalgesia, frequently occur [30]. In more severe cases, vibration sense and proprioception may be lost, which can significantly impact daily functioning [24]. Motor symptoms, although less common than sensory symptoms, also have severe consequences. They typically consist of weakness in the extremities, difficulties with gait and balance, and impaired coordination and have a significant, although sometimes underestimated, influence on the patient’s quality of life. For instance, cancer patients who experience CIPN are at a three-fold higher risk of experiencing falls [31]. In severe cases, CIPN can lead to paresis and complete patient immobilization [32]. Fine motor difficulties are also reported and patients may encounter challenges with daily tasks like buttoning clothing [24]. Autonomic symptoms are infrequent and the main features are orthostatic hypotension, altered sexual or urinary function, and constipation [27,32].

3. Clinical Assessment

Clinically, there is no uniform approach or standard tools for CIPN assessment [33,34]. In the context of painful CIPN, screening questionnaires can be used as a standard method for assessing and diagnosing neuropathic pain [35,36]. Other specific questionnaires commonly utilized are the National Cancer Institute (NCI-CTC) and the Total Neuropathy Score (TNS) [4]. These assessments help classify CIPN into grades or stages and provide a basis for tracking symptom changes over time [34]. The European Organization for Research and Treatment of Cancer (EORT) published the EORT-CIPN 20 guidelines that have proven valuable in clinical practice and research settings, particularly in the context of large oncology clinical trials [34].
Nerve conduction studies and electromyography can be useful in diagnosing CIPN. Those parameters can be especially valuable in cases where clinical symptoms may not fully reflect the extent of nerve damage, offering a more in-depth evaluation [37]. Similarly to other neuropathies, conduction nerve studies are considered the gold standard for detecting and monitoring CIPN [38]. However, these methods are expensive and time consuming and thus have had limited use in clinical settings [39]. Therefore, current CIPN assessment strategies are not sensitive or practical for regular clinical monitoring, making it difficult to predict who is at high risk for harmful side effects or irreversible damage [40].

4. Management of CIPN

As in other clinical conditions, research in CIPN can be conducted with the goals of prevention and/or treatment. To date, there are no proven ways to prevent CIPN and effective treatments are limited. For many patients, when the signs of CIPN first appear, reduction of chemotherapy or even its cessation can be implemented [41], which may negatively affect the overall survival of cancer patients [42]. Determining whether CIPN can be mitigated or prevented without compromising life-saving chemotherapy is crucial. Currently, managing CIPN poses significant challenges. Firstly, predicting who will develop CIPN is difficult. Secondly, symptoms can emerge at any stage during chemotherapy. Thirdly, there are no approved drugs for the effective prevention of CIPN. Finally, while duloxetine is the only approved drug for treating CIPN, its effectiveness is moderate [41,43]. Curiously, despite the expectation that pain control from the brain would be a target in CIPN studies—given that variations in pain modulation may explain individual differences in pain susceptibility, and duloxetine is proposed to control pain by increasing 5-HT/NA-mediated effects in descending pain modulation—the brain has been largely disregarded in the study of CIPN.
While numerous clinical trials have explored various agents with the potential to modify the development of CIPN by targeting its underlying mechanisms, no sufficient evidence is available to recommend a specific pharmacologic intervention [41,44]. Instead, the guidelines of the American Society of Clinical Oncology (ASCO) and the European Society for Medical Oncology (ESMO) suggest regular assessment of CIPN development, with particular vigilance of high-risk patients, considering some predisposing factors, which may include the specific chemotherapy agents employed, cumulative dosages, and individual patient characteristics [41,44]. It is noteworthy that the latter remains understudied.
The current treatment options for CIPN are insufficient. Patients and healthcare providers face significant challenges in effectively managing this debilitating condition. Available treatments mainly focus on symptom relief, such as pain management, but do not address the underlying mechanisms of CIPN [45]. Additionally, these therapies have limited efficacy and come with their own side effects [45,46]. Although duloxetine is the only recommended agent against CIPN [43,47], tricyclic antidepressants and anti-seizure medications are also repurposed in the treatment of painful CIPN, based on established evidence and guidelines for managing different types of neuropathic pain [4,41]. However, ASCO currently makes no recommendations on these therapies, but ESMO supports their consideration for treatment [41,44].
Nonpharmacological treatments may also be useful but there is some disagreement regarding the effectiveness of these approaches. A recent systematic scoping review and expert consensus process offer clinical recommendations for preventing and treating CIPN through non-pharmacological interventions [48]. Exercise, acupuncture, massage therapy, and nutritional interventions are highlighted as potential interventions to alleviate CIPN [48,49]. Also, given the established advantages of exercise in managing chronic pain and cancer and considering its low risk of harm, promoting increased physical activity should be an integral component of CIPN management [50,51,52]. Furthermore, noteworthy work has been carried out in applying brain–computer interface techniques, such as neurofeedback, with promising results in the mitigation of CIPN symptoms [53,54].

5. Mechanisms of CIPN

Chemotherapy agents are designed to destroy cancer cells but can also damage healthy cells, including those at peripheral nerves [25]. There are six classes of chemotherapeutic agents most associated with CIPN: the platinum-based antineoplastics (particularly oxaliplatin [55] and cisplatin [56]), the vinca alkaloids (particularly vincristine [57]), the epothilones (ixabepilone [58,59]), the taxanes (paclitaxel [60], docetaxel [61]), the proteasome inhibitors (bortezomib [62]), and immunomodulatory drugs (thalidomide [63]).
Chemotherapeutic drugs damage the nervous system structures and cause CIPN using complex mechanisms. It is well established that higher doses of chemotherapeutic agents and the number of chemotherapy cycles increase the probabilities of nerve damage and manifestation of neuropathic symptoms [22].
Although the focus of this narrative review is not the peripheral mechanisms of CIPN, it should be mentioned that they are diverse and are under investigation both in animal models and in humans since they play a partial role in CIPN pathophysiology. Preclinical studies have provided valuable insights, with animal models playing a crucial role in identifying the specific peripheral nerve damage caused by the different chemotherapeutic agents. Collectively, these studies showed various mechanisms underlying the effects of chemotherapy on peripheral nerves including mitochondrial damage, neuroinflammation, increased neuronal excitability in neurons of the dorsal root ganglion (DRG), and disruption of the neuronal cytoskeletal architecture and axonal transport [25,64,65]. For a more comprehensive review, please refer to [66,67]. Research into the peripheral mechanisms of CIPN has yielded promising results in both clinical and preclinical studies. For example, the administration of acetyl-L-carnitine decreases mitochondrial dysfunctions and prevents paclitaxel-, oxaliplatin-, and bortezomib-induced peripheral neuropathy [68,69,70,71]. However, a randomized controlled trial enrolling 409 patients found that acetyl-l-carnitine aggravated CIPN [72,73]. A similar pattern was observed with pregabalin, a medication that effectively alleviated CIPN in rodent models by interacting with voltage-gated calcium ion channels [74,75], which are upregulated in DRG during CIPN [76,77]. However, a recent phase III randomized double-blind, placebo-controlled trial using oral gabapentin for the prevention of paclitaxel-induced CIPN showed that gabapentin may not be effective in the prevention of that condition [78]. Due to the lack of effective treatments for CIPN based on current understanding, research should shift to exploring new mechanisms. Understanding the mechanisms of CIPN can lead to the development of more effective methods for diagnosis, treatment, and prevention of this condition, ultimately reducing its significant impact on patients.
In addition to the damage of peripheral structures, neuroinflammation and/or neuroplastic changes at the CNS also account for painful CIPN [79,80]. Central sensitization, involving increases in responses of spinal cord and brain neurons, is believed to also play a pivotal role in the chronic pain and sensory abnormalities characteristic of CIPN. Understanding the mechanisms of CIPN can help us find new treatments to mitigate the impact that CIPN [4]. Taking this into account, the next sections will focus on changes in brain areas, namely those that are involved in descending pain modulation.

5.1. The Brain of CIPN Patients

In the past few decades, brain-imaging studies revealed changes in brain areas in patients who have experienced prolonged pain, namely the limbic system, the part involved in behavioral and emotional responses [81,82]. Abnormalities in pain processing, as revealed by neuroimaging, rather than just peripheral nerve damage or inflammation, are believed to be significant contributors to other chronic pain conditions like osteoarthritis [83,84], trigeminal neuralgia [85], post-herpetic neuralgia [86], chronic low back pain [87], and diabetic neuropathy [88,89]. However, little is known about these mechanisms in CIPN. Despite differences in patient populations, study designs, and sample sizes, Table 1 presents the main findings of the only three neuroimaging studies that have assessed brain structure and function in patients with painful CIPN [90,91,92].
These studies applied fMRI to investigate brain activation patterns in response to chemotherapy treatments. Results reveal distinct patterns of brain activation associated with pain perception in CIPN patients, shedding light on the central mechanisms contributing to chronic neuropathic pain in this population. In 2015, Nudelman and his team conducted a study involving 45 women diagnosed with non-metastatic breast cancer, comparing brain scans before (baseline) and after (1 month and one year later) chemotherapy with women who had cancer but did not undergo chemotherapy [90]. At one-month post-chemotherapy, increased severity of CIPN was correlated with heightened perfusion in several brain regions, including the superior frontal gyrus, cingulate gyrus, left middle gyrus, and medial frontal gyrus. Additionally, at the one-year follow-up, an escalation in CIPN severity from the pre- to the one-month post-chemotherapy assessment was associated with increased perfusion in the left cingulate gyrus and left superior frontal gyrus. However, after one year, no significant associations were found between CIPN severity and brain perfusion [90]. Boland and colleagues also conducted a case-control study in which 24 participants, 12 with multiple myeloma (MM) and CIPN and 12 healthy volunteers, underwent fMRI during the application of noxious heat-pain stimulation [91]. In this study, patients with multiple myeloma exhibited greater activation in the left precuneus and lower activation in the right superior frontal gyrus for both foot and thigh compared to healthy volunteers. Also, activation in the left frontal operculum (near the insula) in response to heat-pain stimulation of the foot was associated with worse CIPN [91]. Recently, Seretny and collaborators conducted a prospective, multicenter cohort study using brain fMRI to study the development of chronic sensory/painful CIPN nine months after chemotherapy. A total of 12 out of 20 patients, classified as CIPN positive (CIPNþ), demonstrated robust activity in sensory, motor, attentional, and affective brain regions in response to punctate stimulation. Additionally, a region-of-interest analysis focusing on the periaqueductal grey (PAG) matter, hypothesized to be relevant for developing CIPNþ, showed significantly increased responses in CIPN-negative (CIPNe) compared to CIPNþ patients [92].
Collectively, the imaging studies in patients with CIPN indicate that this condition is accompanied by structural and functional changes in the brain. The causality relations cannot be established, namely due to the design of the studies and the lack of tools to perform neurochemical characterization of the involved brain regions.

5.2. Contribution of Preclinical Research

In this section, we will review the evidence obtained in animal models that may explain the role of pain modulation from the brain in the appearance of pain during CIPN. Although the features of CIPN models need to be critically reappraised [93], several studies focused on descending pain modulation, using the neurochemical systems targeted by duloxetine, namely 5-HT/NA. Recent imaging studies of CIPN models were performed [80,94,95,96], and this is an approach with potential translational perspectives.
The descending pain modulatory system is an intrinsic network of brain areas that exerts a bidirectional (inhibition/facilitation) control of the nociceptive transmission from the spinal cord [10,11,12]. The PAG plays a key role in top-down pain modulation. It has reciprocal projections with the prefrontal cortex (PFC), anterior cingulate cortex (ACC), and amygdala, along with several regions of the thalamus, hypothalamus [97,98,99], and brainstem regions, such as the locus coeruleus (LC), the rostral ventromedial medulla (RVM), and areas of the medullary reticular formation [100,101,102,103]. The PAG relays its descending input through the RVM since it does not send direct projections to the spinal cord [104]. The RVM is a brainstem center that exerts bidirectional control, leading to a balance of inhibition (antinociception) and facilitation (pronociception) of nociceptive transmission at the spinal cord [105]. The PAG and RVM also project to noradrenergic areas, namely LC, which project to the spinal cord [106].
It is noteworthy that descending pain modulation is also influenced by several corticolimbic regions. The connections between the ACC, PFC, and other mesolimbic areas, such as the ventral tegmental area (VTA) and nucleus accumbens (NAc), contribute to emotional, and cognitive control of pain perception and are involved in reward [107]. Describing these circuits and understanding their function in the context of neuropathic pain is now a focus of research across the field [108,109]. Figure 1 illustrates the brain areas that are affected by neuropathic pain in animal models treated with chemotherapy. There is still much to study, namely the connectivity between the brain areas involved in pain modulation.
The neurochemical modulation used in top-down circuits is complex [15], but the main neurotransmitters used are 5-HT, NA, and opioids. Though opioids display a key role in pain modulation and are used to treat painful conditions [13], this review will not focus on the role of opioids since they are not the first line of therapeutic approach to CIPN [44,110].

5.2.1. New Approaches in Preclinical Studies of CIPN: Imaging Studies Using MRI and Spectroscopy Analysis

The difficulties in studying neuronal activation in the brain of rodent models with neuropathic pain are increasing. Traditionally, methods such as behavioral tests, electrophysiology, pharmacology, and molecular biology approaches have been used to examine the plasticity of the descending pain modulation system. Furthermore, neuroimaging is becoming an important technique for investigating brain activity in animal models and identifying local variations associated with pathological conditions such as chronic pain [111,112]. Neuroimaging techniques applied for brain imaging in pain research include electroencephalography (EEG), fMRI, manganese-enhanced MRI (MEMRI), and positron emission tomography (PET) [111]. These approaches have enabled the study of activation patterns in the brain using chronic pain models. The brain regions most frequently activated during persistent pain include frontal cortex areas, the hippocampus, amygdala, basal ganglia, and NAc [113,114,115,116].
The first studies on animal models of CIPN utilizing MRI methods appeared quite recently (Table 2). Research using a particular fMRI method based on water diffusion found that paclitaxel-induced neuropathy causes alterations in brain regions associated with the emotional and affective aspects of pain at the early stage of polyneuropathy [94]. Our group recently performed imaging studies using MEMRI that demonstrated that paclitaxel treatment increases the neuronal activation of the hypothalamus and PAG at a late stage of CIPN [80]. Besides these brain functional alterations, ex vivo spectroscopy revealed a significant metabolic change in brain regions comprising pain modulation at early and late stages of paclitaxel-induced neuropathy [80]. These alterations in supraspinal metabolites suggest that supplementary processes involving glial cells may be occurring during paclitaxel-induced neuropathy. Similarly, preclinical studies with non-human primates demonstrated the use of fMRI in the investigation of CIPN. Oxaliplatin-treated macaques presented higher activation of the secondary somatosensory and insular cortex in response to cold stimuli, which were attenuated by 5-HT/NA reuptake inhibitor, duloxetine [95,96].
The functional significance of these changes in brain activity and metabolism needs to be checked, particularly the connectivity between these brain regions and the involvement of neuroinflammatory events. Furthermore, it is crucial to thoroughly examine the impact of each chemotherapeutic agent on the brain during the progression of polyneuropathy, at several timepoints. This may be achieved by utilizing multiparametric MRI techniques (e.g., fMRI, MEMRI, magnetization transfer imaging), which will offer valuable insights into the underlying processes of CIPN and may allow to develop protocols that may be used to approach CIPN in its early phases.

5.2.2. The Study of the Descending Pain Modulation during CIPN: Neurochemical Studies of the Serotoninergic and Noradrenergic Systems

Neuroplastic alterations in the descending modulatory pathways have been observed in both animal models of neuropathic pain and in humans [14]. These changes disrupt the balance between descending modulatory circuits promoting descending facilitation over inhibition, which may contribute to the persistence and installation of pain [15,16]. Clinically, patients with CIPN are treated with antidepressants, which boost monoamine levels [7]. Duloxetine, a 5-HT/NA reuptake inhibitor, is the only effective pharmacological treatment in alleviating pain during CIPN [117]. However, the exact mechanisms driving these changes in neurochemical systems remain largely unclear (Table 3).

Involvement of 5-HT in Descending Pain Modulation

The 5-HT, a monoaminergic neurotransmitter, is synthesized from tryptophan through the action of the enzyme tryptophan hydroxylase [129]. Spinal 5-HT can exert both pro- and antinociceptive effects, depending on the specific subtype of the 5-HT receptor [130,131,132]. It is widely recognized that activation of the spinal receptors 5-HT1A and 5-HT7, coupled to Gi/o protein, suppresses nociception, while the activation of the receptors 5-HT2, coupled to Gq/11 protein, and 5-HT3, linked to non-selective cationic channel, has the opposite effect [133]. The proper functioning of the descending serotoninergic pain system is essential for maintaining the balance between facilitation and inhibition in the modulation of nociceptive transmission. The data suggest that chronic pain alters the descending serotoninergic modulatory pathway, with an imbalance favoring facilitation, which might explain how chronic pain develops and is maintained [15,16]. The depletion of serotoninergic neurons in the RVM or serotoninergic pathways has been shown to reduce pain-like behaviors [134] and prevent sensory hypersensitivity in animal models of neuropathic pain [135]. Moreover, in several neuropathic pain models, the descending pain modulation system seems to adapt to persistent pain by enhancing the serotoninergic input to the spinal cord [136,137].
Our lab has developed pioneer work in what concerns paclitaxel-induced neuropathic pain. We showed that during CIPN, the RVM undergoes neuroplastic alterations including local increases in neuronal activation, particularly 5-HT neurons [118]. The increased activation of 5-HT neurons could be due to the activation of the p38MAPK pathway in the RVM [119]. Recently, new evidence about the activation of the RVM during CIPN was provided, namely by showing excitatory projections from PAG somatostatin neurons, which were proposed to activate RVM neurons, promoting descending pain facilitation [138].
Considering the overactivity of the serotoninergic RVM neurons, preclinical studies have shown an increased engagement of the descending pain modulation during CIPN [118,119]. Furthermore, paclitaxel-induced neuropathy is associated with increased levels of 5-HT in the spinal cord [118]. On the contrary, decreased levels of the spinal 5-HT decreased after oxaliplatin and cisplatin treatments [120,121]. Although we could conclude that other studies showed the involvement of 5-HT in descending pain, facilitation during CIPN may depend on the chemotherapeutic agent. Considering that there are some concerns about experimental reports in CIPN models [93,139], the studies need to be continued to fully understand the mechanisms subserving the descending serotoninergic pain modulation.
It is well established in several pain models that spinal 5-HT may induce pro-or antinociception effects depending on the target receptor subtype [133]. A study showed that the levels of 5-HT1A receptor mRNA were downregulated in the spinal dorsal horn after oxaliplatin treatment [122,123]. It was also reported, with greater evidence, that its mRNA level rose in the rodent spinal dorsal horn and that treatment with a 5-HT1A agonist inhibited the oxaliplatin-induced pain-like behaviors [123,140,141,142]. Curiously, no changes were reported in spinal 5-HT1A expression after paclitaxel and vincristine treatment [123]. Regarding 5-HT2 receptors, the information available about its role in chemotherapy-induced neuropathic pain is not very clear. In a model of oxaliplatin-induced neuropathy, a 5-HT2A receptor antagonist was shown to suppress the analgesic effects of neurotrophin on nociceptive behaviors [143]. Moreover, 5-HT2A receptor knock-out mice did not exhibit any vincristine-related painful phenotype, suggesting that the 5-HT2A receptor could be involved in vincristine-induced neuropathic pain. In addition to these modifications, vincristine treatment also triggered the overexpression of this receptor in the superficial layers of the spinal dorsal horn [124]. It is noteworthy that oxaliplatin also induced an increase in the 5-HT2C expression in the spinal cord and its antagonism produced antinociception [125].
The 5-HT3 receptor, which is the only ionotropic 5-HT receptor with excitatory functions, has a pronociceptive role in numerous models of chronic pain, including during CIPN [118,119,126,144,145]. Our group showed that spinal administration of 5-HT3A receptor antagonist induced antinociception in paclitaxel-treated animals [118]. Likewise, Liu et al. confirmed that the intrathecal injection of the 5-HT3A receptor antagonist partially reversed the paclitaxel-induced neuropathic pain [119]. Furthermore, our work showed that the antinociceptive effects of the 5-HT3A receptor antagonist are associated with the overexpression of this receptor in the superficial dorsal horn [118]. Moreover, recent work also showed that the spinal 5-HT3A receptor seems to be a perfect target for natural products, such as natural coumarin, to treat vincristine-induced neuropathic pain [126].
There is a clear gap of knowledge on the contribution of each 5-HT receptor in pain installation and management during CIPN. It will be critical to explore the participation of other l 5-HT receptors in the spinal cord during CIPN, as their role and expression seem to be dependent on the chemotherapeutic drug and the stage of CIPN.

Involvement of NA in Descending Pain Modulation

The biosynthesis of NA involves the enzymes tyrosine hydroxylase (TH) and dopamine-β-hydroxylase (DBH), which are frequently used as surrogates of noradrenergic pathways [146].
Descending noradrenergic projections originate from three brainstem clusters of noradrenergic neurons: the A6 (the LC and nucleus subcoeruleus), as well as the A5 and A7 noradrenergic cell groups [147,148,149,150,151,152]. These fibers release NA at the spinal cord, where it inhibits peripheral input and spinal dorsal horn neurons by activating α2-adrenergic receptors (α2-AR) [146].
Alpha-adrenergic receptors (AR) are divided into α1- (subtypes 1A, 1B, and 2D) and α2- (subtypes 2A and 2C) AR, which are part of the G protein-coupled receptor family [146,153]. In the DRG and spinal dorsal horn, the α1-AR are combined to phospholipase C via Gq protein or directly to calcium influx facilitating nociceptive transmission [154]. Otherwise, the α2-AR are combined to adenylcyclase via Gi protein, reducing the formation of cyclic AMP and calcium influx during action potential to inhibit neurotransmitter release [146]. These α2-AR are located in noradrenergic terminals (as autoreceptors), spinal neurons, central terminals of peripheral nerve fibers, and DRG neurons [146,155]. When NA is released from descending fibers, it produces analgesia by acting on distinct subtypes of spinal α2-AR. The α2A-AR subtype is highly expressed in descending noradrenergic fibers, and its activation inhibits NA release to spinal neurons [153]. This subtype is also found on central terminals of primary afferents that contain substance P and glutamate; its activation leads to presynaptic inhibition of these neurotransmitters, promoting antinociception [156]. The α2C-AR is located on DRG terminals and postsynaptically on spinal excitatory interneurons. Activation of postsynaptic α2C-AR inhibits transmission of the nociceptive signals to the supraspinal pain processing regions [157].
Chronic pain induced neuroplastic modifications at the descending noradrenergic modulation, which may promote the chronification of this condition [158,159].
It is well established that NA release at the spinal cord mainly comes from the pontine LC [146] and induces analgesia by activating spinal α2A-AR, which inhibits nociceptive transmission in the spinal dorsal horn [160].
Some studies have reported a diminished role of the noradrenergic system in animal models of neuropathic pain [158,161]. However, this view is not universally accepted, as other studies suggest that the descending noradrenergic system activity may increase to counteract the enhanced nociceptive input from damaged peripheral fibers in traumatic neuropathic pain models [162,163]. The impact of changes in descending noradrenergic pain modulation during CIPN is still largely unexplored (Table 3). Recent findings have shed some light on this, demonstrating that paclitaxel treatment increases TH levels in the LC [127]. Moreover, in paclitaxel-induced neuropathy, there is an engagement of descending noradrenergic inhibition. Reboxetine, a selective NA reuptake inhibitor, has been shown to produce antinociceptive effects in paclitaxel-treated rats [128]. This animal model also exhibited increased expression of DBH in the spinal cord [128]. Supporting those results, the increased levels of NA in the spinal cord were also reported in paclitaxel-induced neuropathy [127]. Noradrenaline inhibits the nociceptive transmission by activating spinal α2-AR located both pre- and postsynaptically [156,160,164]. The results of our investigation demonstrated that the behavioral effects of spinal injection of an α2-AR agonist or antagonist were significantly stronger in the paclitaxel group. But paclitaxel did not change α2-AR expression, suggesting an increased potency of the spinal α2-AR [128]. On the contrary, another study showed that paclitaxel induced an increase in spinal α1- and α2-AR gene expression [127]. Similarly, a study using oxaliplatin-injected animals indicated that spinal administration of NA yields an inhibitory effect in spinal hyperactivation and nociceptive behaviors [165].
As discussed for the descending serotonergic modulation of pain, it is necessary to better understand the descending noradrenergic pain modulation in CIPN, specifically the mechanistic differences and similarities that exist in different animal models, since the descending noradrenergic inhibitory controls seem to be crucial for pain relief by supraspinal drugs during CIPN [166].

Other Neurochemical Systems

Cannabinoids (CB) are compounds that possess pain-relieving qualities, and the endocannabinoid system appears to have an important function in the regulation of pain through descending modulation [167,168,169]. Multiple cannabinoid receptors have been identified, namely the CB1 cannabinoid (CB1) receptor and the CB2 cannabinoid (CB2) receptor. Both receptors are G protein-coupled receptors with inhibitory effects of adenylyl cyclase and certain calcium and potassium channels. They also activate the mitogen-activated protein kinase [170]. It is noteworthy that CB1 receptors can also activate or arrest various G-proteins, as well as form heterodimers with opioids or serotonin receptors [171,172,173]. These characteristics render CB receptors a compelling focus for therapeutic intervention. The CB1 receptors are expressed in several areas involved in pain modulation, such as neurons of the cerebral cortex, basal ganglia, amygdala, PAG, hypothalamus, brainstem medullary nuclei, and spinal cord [174], but they are also expressed by astrocytes [175]. The CB2 receptors are expressed in neurons of the brainstem, cerebellum, basal ganglia, PFC, and hippocampus [176], but also in microglia, astrocytes, and oligodendrocytes [177,178,179]. There are studies indicating that prolonged pain conditions lead to an overregulation of CB2 receptors in both the peripheral and CNS [180,181,182]. A recent publication showed that the peripheral CB2 receptor appears to have a role in suppressing paclitaxel-induced neuropathic pain [183]. Pain-relieving properties of cannabis in CIPN have been described. Combined or alone, cannabidiol (CBD) and tetrahydrocannabinol (THC) seem to be effective in attenuating pain-like behaviors in paclitaxel-, oxaliplatin- and vincristine-induced neuropathic pain [184]. It was also reported that the CB1 and/or CB2 receptor agonists also present analgesic effects in paclitaxel-, cisplatin- and vincristine-induced neuropathy [185,186,187]. Moreover, paclitaxel induced the expression of CB2 receptors in spinal dorsal horn microglia, which was attenuated by a selective CB2 agonist [188]. Furthermore, the interaction of CB receptors with other receptors, e.g., opioid receptors, has been described in CIPN with a potential role for therapeutic targets. Combined CB1 receptor and delta-opioid receptor agonists reversed the paclitaxel-induced hypersensitivity through the spinal upregulated CB1 receptor/delta-opioid receptor heteromers [189].
While there is ongoing research on the use of cannabis to treat CIPN, more extensive investigation is required to fully understand the processes of action of CB, as well as the synergistic interactions of CB receptors with other receptors, namely opioid receptors, and the specific alterations that take place in brain areas involved in endocannabinoid modulation.
The implication of the cholinergic system in changes in pain modulation during chronic pain was demonstrated with a loss of tonic spinal inhibition of nociceptive transmission in neuropathic pain models [190]. The cholinergic system involves two different types of receptors: the nicotinic acetylcholine receptor (nAChR) and muscarinic acetylcholine receptor (mAChR) [191].
The nAChRs, ionotropic receptors, present a wide range of subunits (α1–α10, β1–β4, γ, δ, and ε), which can combine to form different subtypes of this receptor [192]. The metabotropic mAChRs present distinctive types of receptors: m1AChR, m2AChR, m3AChR, m4AChR, and m5AChR. m1 AChR, m3AChR, and m5AChR are excitatory, and m2AChR and m4AChR are inhibitory [193]. The nAChRs and mAChRs regulate the pain modulation at the spinal cord and at supraspinal levels. For example, it is well established that nAChR may stimulate descending inhibitory pathways at the supraspinal areas, via spinal α2-Ars and 5-HT3 receptors [194]. For an extensive review, see [191].
A large body of evidence indicates that the cholinergic system may also play a role in the onset and progression of painful CIPN [195,196]. Pharmacological interventions that increased the quantity of acetylcholine (ACh) in the synaptic cleft, such as Ach inhibitors, inhibitors of Ach exocytosis (botulinum neurotoxin A), and a precursor of choline (citicoline), exhibited antinociceptive effects on mechanical and/or thermal hypersensitivity in rodent models of CIPN [195,196,197,198]. Furthermore, nicotine (nAChR agonist) also demonstrated analgesic effects and could prevent mechanical and thermal hypersensitivity in paclitaxel-treated rats [199]. In addition, other more specific nAChR agonists, namely compounds that bind to the α4β2 nAChRs seem to be more effective in the resolution of the mechanical hypersensitivity induced by vincristine and oxaliplatin [200,201]. Likewise, α7 nAChRs agonists presented an analgesic effect in pain-like behaviors in rodents treated with oxaliplatin and paclitaxel [202,203,204]. There is some evidence that systemic α9/α10 nAChRs antagonists present antinociceptive effects in both mechanical and/or thermal sensorial modalities in rodent models of oxaliplatin- and paclitaxel-induced neuropathy [205,206,207,208,209,210,211,212,213]. Surprisingly, m1AChRs agonists and antagonists reversed mechanical and thermal hypersensitivity induced by paclitaxel and oxaliplatin [214,215]. Noteworthy, spinal m2AChRs antagonist, methoctramine, reversed the analgesic effect of donepezil, a cholinesterase inhibitor, only in paclitaxel-treated animals [196].
Although the cholinergic system is well studied in pain modulation, the role of this neurochemical system in CIPN requires more investigation, particularly at the supraspinal level, to discover new therapeutic options.

Pharmacological Interventions

Given the change in pain modulatory areas and the significant role of the brain in the pathophysiology of CIPN, it is crucial to review brain interventions in animal models of CIPN to enhance our understanding of the pathology.
For several years, efforts have been made to evaluate the effects of specific agonists, antagonists, and repurposed drugs administered directly into the brain, to improve CIPN symptoms. Preclinical studies using paclitaxel-treated rats or mice have shown that the intracerebroventricular administration of selective kinin B1 and B2 receptor antagonists, gabapentin (voltage-gated calcium channel inhibitor), and a novel N-type voltage-gated calcium channel blocker resulted in analgesic effects for thermal and/or mechanical hypersensitivity [166,216,217].
Reasonably better studied are the effects of brain interventions in CIPN using oxaliplatin-induced neuropathy animal models. Oxaliplatin-treated macaques exhibited an improvement in cold hypersensitivity following the administration of the GABAA receptor agonist, muscimol, into the secondary somatosensorial cortex and insula [95]. Additionally, muscimol injection into the dorsolateral PAG induced analgesia in oxaliplatin-treated rats [218]. Local administration of interleukin 1β and 6, and tumor necrosis factor α antagonists in the same brainstem area also ameliorated CIPN symptoms [218]. Moreover, the injection of m2AChR agonist, oxotremorine, into the posterior insula reversed the mechanical allodynia in oxaliplatin-treated rats [195]. Similarly, triple monoamine reuptake inhibitors, a selective NA and 5-HT reuptake inhibitor, and a selective 5-HT reuptake inhibitor administrated directly into the ACC reversed different pain-like behaviors in oxaliplatin-treated mice [219].
Other brain interventions, specifically the intracerebroventricular administration of CDP-choline (which increases choline and Ach), a selective Gi/o protein inhibitor, a GIRK1 channel blocker, a PKC inhibitor, and orexin-A also induced analgesia in oxaliplatin-treated rodents [197,220,221,222,223].
While brain interventions hold promise for alleviating the effects of painful CIPN, further preclinical studies are necessary. These studies should involve other chemotherapy drugs and take advantage of PET-MRI neuroimaging techniques, which enable the correlation of drug-induced changes in metabolism and neurotransmission with structural and connectivity alterations. Additionally, although this section addresses pharmacological interventions with precise molecular targets, it should be noted that the role of natural compounds with therapeutic potential should not be discarded (reviewed in [224]) but it is not the focus of the current review.

6. Concluding Remarks and Future Perspectives

This narrative review proposes that understanding the central mechanisms of CIPN involving the brain’s role in pain processing and response to chemotherapy can leverage the approach to managing and potentially preventing this condition.
Preclinical studies, using animal models of CIPN, have attempted to contribute to a better understanding of the brain mechanisms underlying painful CIPN. Cutting-edge studies are starting to uncover functional and metabolic brain changes, namely in the pain modulation areas, in response to chemotherapy [80]. Furthermore, there is also a great effort to identify blood biomarkers for painful CIPN to use them in clinical practice [225]. These techniques have a good potential for translation to the patient’s treatment.
In clinical settings, future challenges can be overcome with this new perspective of the CIPN approach (Figure 2). The main points are:
  • Targeted Therapies: Understanding how the brain processes pain and responds to chemotherapy allows for developing treatments targeting these specific mechanisms. This means that medications and interventions can be tailored to the individual, addressing their unique pain processing and tolerance;
  • Reducing Side Effects: A personalized approach based on a patient’s brain responses can reduce the risk of CIPN. By selecting prophylactic treatments, it is possible to mitigate or prevent painful CIPN;
  • Improved Treatment Outcomes: The ability to reduce side effects based on an individual’s brain profile can improve treatment outcomes. By avoiding or minimizing CIPN, patients may be more likely to complete their prescribed chemotherapy regimens, leading to improved cancer treatment success;
  • Enhanced Quality of Life: CIPN can have a profound impact on a patient’s quality of life, as it often leads to chronic pain and limitations in daily activities. Personalized treatment that minimizes the risk of CIPN can contribute to a better quality of life during and after cancer treatment;
  • Reducing Healthcare Costs: Effective personalized treatment of CIPN can potentially reduce healthcare costs associated with treating CIPN-related complications, including pain management and rehabilitative care.
Shifting research focus on CIPN from the peripheral nerves to the CNS, particularly the brain, may reveal novel insights into the mechanisms of development and persistence of this neuropathic pain. Understanding the central mechanisms of CIPN could lead to more effective treatments and personalized approaches. Additionally, the personalized treatment of neuropathic pain has been widely discussed [226,227]. Adopting a multidimensional strategy in clinical settings that allows patient categorization based on central functional and structural biomarkers obtained from neuroimaging, together with clinical indicators such as patient-reported outcomes and sensory phenotyping seems to be the pathway for preventing and treatment of CIPN.
In conclusion, understanding the key role of the brain and personalizing treatment for painful CIPN based on individual brain responses offer the possibility of providing relief and improving the quality of life for cancer survivors.

Author Contributions

M.C., I.T. and J.T.C.-P. prepared the original draft. I.T. and J.T.C.-P. conceptualized, supervised, reviewed, and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research was performed in the grant “Cátedra de Medicina da Dor” ascribed by Fundaҫão Grunenthal to the Faculty of Medicine of the University of Porto.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Since this is a comprehensive review, the availability of the data is not an issue to be considered.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. NCI. National Cancer Institute—Side Effects of Cancer Treatment. Available online: http://evs.nci.nih.gov/ftp1/CTCAE/CTCAE_4.03/CTCAE_4.03_2010-06-14_QuickReference_8.5x11.pdf (accessed on 31 October 2023).
  2. Seretny, M.; Currie, G.L.; Sena, E.S.; Ramnarine, S.; Grant, R.; MacLeod, M.R.; Colvin, L.A.; Fallon, M. Incidence, prevalence, and predictors of chemotherapy-induced peripheral neuropathy: A systematic review and meta-analysis. Pain 2014, 155, 2461–2470. [Google Scholar] [CrossRef] [PubMed]
  3. Pike, C.T.; Birnbaum, H.G.; Muehlenbein, C.E.; Pohl, G.M.; Natale, R.B. Healthcare costs and workloss burden of patients with chemotherapy-associated peripheral neuropathy in breast, ovarian, head and neck, and nonsmall cell lung cancer. Chemother. Res. Pract. 2012, 2012, 913848. [Google Scholar] [CrossRef] [PubMed]
  4. Colvin, L.A. Chemotherapy-induced peripheral neuropathy: Where are we now? Pain 2019, 160 (Suppl. S1), S1–S10. [Google Scholar] [CrossRef] [PubMed]
  5. Fallon, M.T. Neuropathic pain in cancer. Br. J. Anaesth. 2013, 111, 105–111. [Google Scholar] [CrossRef]
  6. Cavaletti, G.; Marmiroli, P. Chemotherapy-induced peripheral neurotoxicity. Curr. Opin. Neurol. 2015, 28, 500–507. [Google Scholar] [CrossRef] [PubMed]
  7. Sisignano, M.; Baron, R.; Scholich, K.; Geisslinger, G. Mechanism-based treatment for chemotherapy-induced peripheral neuropathic pain. Nat. Rev. Neurol. 2014, 10, 694–707. [Google Scholar] [CrossRef] [PubMed]
  8. Boyette-Davis, J.A.; Hou, S.; Abdi, S.; Dougherty, P.M. An updated understanding of the mechanisms involved in chemotherapy-induced neuropathy. Pain Manag. 2018, 8, 363–375. [Google Scholar] [CrossRef] [PubMed]
  9. Ma, J.; Kavelaars, A.; Dougherty, P.M.; Heijnen, C.J. Beyond symptomatic relief for chemotherapy-induced peripheral neuropathy: Targeting the source. Cancer 2018, 124, 2289–2298. [Google Scholar] [CrossRef] [PubMed]
  10. Heinricher, M.M.; Tavares, I.; Leith, J.L.; Lumb, B.M. Descending control of nociception: Specificity, recruitment and plasticity. Brain Res. Rev. 2009, 60, 214–225. [Google Scholar] [CrossRef] [PubMed]
  11. Ossipov, M.H.; Dussor, G.O.; Porreca, F. Central modulation of pain. J. Clin. Investig. 2010, 120, 3779–3787. [Google Scholar] [CrossRef] [PubMed]
  12. Bannister, K.; Dickenson, A.H. What do monoamines do in pain modulation? Curr. Opin. Support. Palliat. Care 2016, 10, 143–148. [Google Scholar] [CrossRef] [PubMed]
  13. Tavares, I.; Costa-Pereira, J.T.; Martins, I. Monoaminergic and Opioidergic Modulation of Brainstem Circuits: New Insights Into the Clinical Challenges of Pain Treatment? Front. Pain Res. 2021, 2, 696515. [Google Scholar] [CrossRef] [PubMed]
  14. Ossipov, M.H.; Morimura, K.; Porreca, F. Descending pain modulation and chronification of pain. Curr. Opin. Support. Palliat. Care 2014, 8, 143–151. [Google Scholar] [CrossRef] [PubMed]
  15. Millan, M.J. Descending control of pain. Prog. Neurobiol. 2002, 66, 355–474. [Google Scholar] [CrossRef] [PubMed]
  16. Tracey, I.; Mantyh, P.W. The cerebral signature for pain perception and its modulation. Neuron 2007, 55, 377–391. [Google Scholar] [CrossRef] [PubMed]
  17. Mezzanotte, J.N.; Grimm, M.; Shinde, N.V.; Nolan, T.; Worthen-Chaudhari, L.; Williams, N.O.; Lustberg, M.B. Updates in the Treatment of Chemotherapy-Induced Peripheral Neuropathy. Curr. Treat. Options Oncol. 2022, 23, 29–42. [Google Scholar] [CrossRef] [PubMed]
  18. Tracey, I. Neuroimaging mechanisms in pain: From discovery to translation. Pain 2017, 158 (Suppl. S1), S115–S122. [Google Scholar] [CrossRef] [PubMed]
  19. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer statistics, 2022. CA Cancer J. Clin. 2022, 72, 7–33. [Google Scholar] [CrossRef] [PubMed]
  20. Cioroiu, C.; Weimer, L.H. Update on Chemotherapy-Induced Peripheral Neuropathy. Curr. Neurol. Neurosci. Rep. 2017, 17, 47. [Google Scholar] [CrossRef] [PubMed]
  21. Maihöfner, C.; Diel, I.; Tesch, H.; Quandel, T.; Baron, R. Chemotherapy-induced peripheral neuropathy (CIPN): Current therapies and topical treatment option with high-concentration capsaicin. Support. Care Cancer 2021, 29, 4223–4238. [Google Scholar] [CrossRef]
  22. Staff, N.P.; Grisold, A.; Grisold, W.; Windebank, A.J. Chemotherapy-induced peripheral neuropathy: A current review. Ann. Neurol. 2017, 81, 772–781. [Google Scholar] [CrossRef] [PubMed]
  23. Sprauten, M.; Darrah, T.H.; Peterson, D.R.; Campbell, M.E.; Hannigan, R.E.; Cvancarova, M.; Beard, C.; Haugnes, H.S.; Fosså, S.D.; Oldenburg, J.; et al. Impact of long-term serum platinum concentrations on neuro- and ototoxicity in Cisplatin-treated survivors of testicular cancer. J. Clin. Oncol. 2012, 30, 300–307. [Google Scholar] [CrossRef] [PubMed]
  24. Flatters, S.J.L.; Dougherty, P.M.; Colvin, L.A. Clinical and preclinical perspectives on Chemotherapy-Induced Peripheral Neuropathy (CIPN): A narrative review. Br. J. Anaesth. 2017, 119, 737–749. [Google Scholar] [CrossRef] [PubMed]
  25. 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. [Google Scholar] [CrossRef] [PubMed]
  26. Park, S.B.; Goldstein, D.; Krishnan, A.V.; Lin, C.S.; Friedlander, M.L.; Cassidy, J.; Koltzenburg, M.; Kiernan, M.C. Chemotherapy-induced peripheral neurotoxicity: A critical analysis. CA Cancer J. Clin. 2013, 63, 419–437. [Google Scholar] [CrossRef] [PubMed]
  27. Zhang, S. Chemotherapy-induced peripheral neuropathy and rehabilitation: A review. Semin. Oncol. 2021, 48, 193–207. [Google Scholar] [CrossRef] [PubMed]
  28. Hu, L.Y.; Mi, W.L.; Wu, G.C.; Wang, Y.Q.; Mao-Ying, Q.L. Prevention and Treatment for Chemotherapy-Induced Peripheral Neuropathy: Therapies Based on CIPN Mechanisms. Curr. Neuropharmacol. 2019, 17, 184–196. [Google Scholar] [CrossRef]
  29. Grisold, W.; Cavaletti, G.; Windebank, A.J. Peripheral neuropathies from chemotherapeutics and targeted agents: Diagnosis, treatment, and prevention. Neuro Oncol. 2012, 14 (Suppl. S4), iv45–iv54. [Google Scholar] [CrossRef] [PubMed]
  30. Bernhardson, B.M.; Tishelman, C.; Rutqvist, L.E. Chemosensory changes experienced by patients undergoing cancer chemotherapy: A qualitative interview study. J. Pain Symptom Manag. 2007, 34, 403–412. [Google Scholar] [CrossRef] [PubMed]
  31. Kolb, N.A.; Smith, A.G.; Singleton, J.R.; Beck, S.L.; Stoddard, G.J.; Brown, S.; Mooney, K. The Association of Chemotherapy-Induced Peripheral Neuropathy Symptoms and the Risk of Falling. JAMA Neurol. 2016, 73, 860–866. [Google Scholar] [CrossRef] [PubMed]
  32. Mols, F.; van de Poll-Franse, L.V.; Vreugdenhil, G.; Beijers, A.J.; Kieffer, J.M.; Aaronson, N.K.; Husson, O. Reference data of the European Organisation for Research and Treatment of Cancer (EORTC) QLQ-CIPN20 Questionnaire in the general Dutch population. Eur. J. Cancer 2016, 69, 28–38. [Google Scholar] [CrossRef] [PubMed]
  33. Gewandter, J.S.; Brell, J.; Cavaletti, G.; Dougherty, P.M.; Evans, S.; Howie, L.; McDermott, M.P.; O’Mara, A.; Smith, A.G.; Dastros-Pitei, D.; et al. Trial designs for chemotherapy-induced peripheral neuropathy prevention: ACTTION recommendations. Neurology 2018, 91, 403–413. [Google Scholar] [CrossRef] [PubMed]
  34. Cavaletti, G.; Cornblath, D.R.; Merkies, I.S.J.; Postma, T.J.; Rossi, E.; Frigeni, B.; Alberti, P.; Bruna, J.; Velasco, R.; Argyriou, A.A.; et al. The chemotherapy-induced peripheral neuropathy outcome measures standardization study: From consensus to the first validity and reliability findings. Ann. Oncol. 2013, 24, 454–462. [Google Scholar] [CrossRef] [PubMed]
  35. Jones, R.C., 3rd; Backonja, M.M. Review of neuropathic pain screening and assessment tools. Curr. Pain Headache Rep. 2013, 17, 363. [Google Scholar] [CrossRef]
  36. Haanpää, M.; Attal, N.; Backonja, M.; Baron, R.; Bennett, M.; Bouhassira, D.; Cruccu, G.; Hansson, P.; Haythornthwaite, J.A.; Iannetti, G.D.; et al. NeuPSIG guidelines on neuropathic pain assessment. Pain 2011, 152, 14–27. [Google Scholar] [CrossRef] [PubMed]
  37. Argyriou, A.A.; Park, S.B.; Islam, B.; Tamburin, S.; Velasco, R.; Alberti, P.; Bruna, J.; Psimaras, D.; Cavaletti, G.; Cornblath, D.R. Neurophysiological, nerve imaging and other techniques to assess chemotherapy-induced peripheral neurotoxicity in the clinical and research settings. J. Neurol. Neurosurg. Psychiatry 2019, 90, 1361–1369. [Google Scholar] [CrossRef] [PubMed]
  38. Fuglsang-Frederiksen, A.; Pugdahl, K. Current status on electrodiagnostic standards and guidelines in neuromuscular disorders. Clin. Neurophysiol. 2011, 122, 440–455. [Google Scholar] [CrossRef] [PubMed]
  39. Ibrahim, E.Y.; Ehrlich, B.E. Prevention of chemotherapy-induced peripheral neuropathy: A review of recent findings. Crit. Rev. Oncol. Hematol. 2020, 145, 102831. [Google Scholar] [CrossRef]
  40. McCrary, J.M.; Goldstein, D.; Boyle, F.; Cox, K.; Grimison, P.; Kiernan, M.C.; Krishnan, A.V.; Lewis, C.R.; Webber, K.; Baron-Hay, S.; et al. Optimal clinical assessment strategies for chemotherapy-induced peripheral neuropathy (CIPN): A systematic review and Delphi survey. Support. Care Cancer 2017, 25, 3485–3493. [Google Scholar] [CrossRef] [PubMed]
  41. Loprinzi, C.L.; Lacchetti, C.; Bleeker, J.; Cavaletti, G.; Chauhan, C.; Hertz, D.L.; Kelley, M.R.; Lavino, A.; Lustberg, M.B.; Paice, J.A.; et al. Prevention and Management of Chemotherapy-Induced Peripheral Neuropathy in Survivors of Adult Cancers: ASCO Guideline Update. J. Clin. Oncol. 2020, 38, 3325–3348. [Google Scholar] [CrossRef] [PubMed]
  42. Dorsey, S.G.; Kleckner, I.R.; Barton, D.; Mustian, K.; O’Mara, A.; St Germain, D.; Cavaletti, G.; Danhauer, S.C.; Hershman, D.L.; Hohmann, A.G.; et al. The National Cancer Institute Clinical Trials Planning Meeting for Prevention and Treatment of Chemotherapy-Induced Peripheral Neuropathy. J. Natl. Cancer Inst. 2019, 111, 531–537. [Google Scholar] [CrossRef] [PubMed]
  43. Smith, E.M.; Pang, H.; Cirrincione, C.; Fleishman, S.; Paskett, E.D.; Ahles, T.; Bressler, L.R.; Fadul, C.E.; Knox, C.; Le-Lindqwister, N.; et al. Effect of duloxetine on pain, function, and quality of life among patients with chemotherapy-induced painful peripheral neuropathy: A randomized clinical trial. JAMA 2013, 309, 1359–1367. [Google Scholar] [CrossRef] [PubMed]
  44. Jordan, B.; Margulies, A.; Cardoso, F.; Cavaletti, G.; Haugnes, H.S.; Jahn, P.; Le Rhun, E.; Preusser, M.; Scotté, F.; Taphoorn, M.J.B.; et al. Systemic anticancer therapy-induced peripheral and central neurotoxicity: ESMO-EONS-EANO Clinical Practice Guidelines for diagnosis, prevention, treatment and follow-up. Ann. Oncol. 2020, 31, 1306–1319. [Google Scholar] [CrossRef]
  45. Stillman, M.; Cata, J.P. Management of chemotherapy-induced peripheral neuropathy. Curr. Pain Headache Rep. 2006, 10, 279–287. [Google Scholar] [CrossRef]
  46. D’Souza, R.S.; Alvarez, G.A.M.; Dombovy-Johnson, M.; Eller, J.; Abd-Elsayed, A. Evidence-Based Treatment of Pain in Chemotherapy-Induced Peripheral Neuropathy. Curr. Pain Headache Rep. 2023, 27, 99–116. [Google Scholar] [CrossRef] [PubMed]
  47. Farshchian, N.; Alavi, A.; Heydarheydari, S.; Moradian, N. Comparative study of the effects of venlafaxine and duloxetine on chemotherapy-induced peripheral neuropathy. Cancer Chemother. Pharmacol. 2018, 82, 787–793. [Google Scholar] [CrossRef] [PubMed]
  48. Klafke, N.; Bossert, J.; Kröger, B.; Neuberger, P.; Heyder, U.; Layer, M.; Winkler, M.; Idler, C.; Kaschdailewitsch, E.; Heine, R.; et al. Prevention and Treatment of Chemotherapy-Induced Peripheral Neuropathy (CIPN) with Non-Pharmacological Interventions: Clinical Recommendations from a Systematic Scoping Review and an Expert Consensus Process. Med. Sci. 2023, 11, 15. [Google Scholar] [CrossRef] [PubMed]
  49. Papadopoulou, M.; Stamou, M.; Bakalidou, D.; Moschovos, C.; Zouvelou, V.; Zis, P.; Tzartos, J.; Chroni, E.; Michopoulos, I.; Tsivgoulis, G. Non-pharmacological Interventions on Pain and Quality of Life in Chemotherapy Induced Polyneuropathy: Systematic Review and Meta-Analysis. In Vivo 2023, 37, 47–56. [Google Scholar] [CrossRef] [PubMed]
  50. Geneen, L.J.; Moore, R.A.; Clarke, C.; Martin, D.; Colvin, L.A.; Smith, B.H. Physical activity and exercise for chronic pain in adults: An overview of Cochrane Reviews. Cochrane Database Syst. Rev. 2017, 1, Cd011279. [Google Scholar] [CrossRef]
  51. Huang, Y.; Tan, T.; Liu, L.; Yan, Z.; Deng, Y.; Li, G.; Li, M.; Xiong, J. Exercise for reducing chemotherapy-induced peripheral neuropathy: A systematic review and meta-analysis of randomized controlled trials. Front. Neurol. 2023, 14, 1252259. [Google Scholar] [CrossRef]
  52. Cao, A.; Cartmel, B.; Li, F.Y.; Gottlieb, L.T.; Harrigan, M.; Ligibel, J.A.; Gogoi, R.; Schwartz, P.E.; Esserman, D.A.; Irwin, M.L.; et al. Effect of Exercise on Chemotherapy-Induced Peripheral Neuropathy Among Patients Treated for Ovarian Cancer: A Secondary Analysis of a Randomized Clinical Trial. JAMA Netw. Open 2023, 6, e2326463. [Google Scholar] [CrossRef]
  53. Prinsloo, S.; Novy, D.; Driver, L.; Lyle, R.; Ramondetta, L.; Eng, C.; Lopez, G.; Li, Y.; Cohen, L. The Long-Term Impact of Neurofeedback on Symptom Burden and Interference in Patients With Chronic Chemotherapy-Induced Neuropathy: Analysis of a Randomized Controlled Trial. J. Pain Symptom Manag. 2018, 55, 1276–1285. [Google Scholar] [CrossRef] [PubMed]
  54. Prinsloo, S.; Novy, D.; Driver, L.; Lyle, R.; Ramondetta, L.; Eng, C.; McQuade, J.; Lopez, G.; Cohen, L. Randomized controlled trial of neurofeedback on chemotherapy-induced peripheral neuropathy: A pilot study. Cancer 2017, 123, 1989–1997. [Google Scholar] [CrossRef] [PubMed]
  55. Kang, L.; Tian, Y.; Xu, S.; Chen, H. Oxaliplatin-induced peripheral neuropathy: Clinical features, mechanisms, prevention and treatment. J. Neurol. 2021, 268, 3269–3282. [Google Scholar] [CrossRef] [PubMed]
  56. Zhang, X.; Trendowski, M.R.; Wilkinson, E.; Shahbazi, M.; Dinh, P.C.; Shuey, M.M.; Feldman, D.R.; Hamilton, R.J.; Vaughn, D.J.; Fung, C.; et al. Pharmacogenomics of cisplatin-induced neurotoxicities: Hearing loss, tinnitus, and peripheral sensory neuropathy. Cancer Med. 2022, 11, 2801–2816. [Google Scholar] [CrossRef] [PubMed]
  57. Triarico, S.; Romano, A.; Attinà, G.; Capozza, M.A.; Maurizi, P.; Mastrangelo, S.; Ruggiero, A. Vincristine-Induced Peripheral Neuropathy (VIPN) in Pediatric Tumors: Mechanisms, Risk Factors, Strategies of Prevention and Treatment. Int. J. Mol. Sci. 2021, 22, 4112. [Google Scholar] [CrossRef] [PubMed]
  58. Vahdat, L.T.; Thomas, E.S.; Roché, H.H.; Hortobagyi, G.N.; Sparano, J.A.; Yelle, L.; Fornier, M.N.; Martín, M.; Bunnell, C.A.; Mukhopadhyay, P.; et al. Ixabepilone-associated peripheral neuropathy: Data from across the phase II and III clinical trials. Support. Care Cancer 2012, 20, 2661–2668. [Google Scholar] [CrossRef] [PubMed]
  59. Tamburin, S.; Park, S.B.; Alberti, P.; Demichelis, C.; Schenone, A.; Argyriou, A.A. Taxane and epothilone-induced peripheral neurotoxicity: From pathogenesis to treatment. J. Peripher. Nerv. Syst. 2019, 24 (Suppl. S2), S40–S51. [Google Scholar] [CrossRef] [PubMed]
  60. Staff, N.P.; Fehrenbacher, J.C.; Caillaud, M.; Damaj, M.I.; Segal, R.A.; Rieger, S. Pathogenesis of paclitaxel-induced peripheral neuropathy: A current review of in vitro and in vivo findings using rodent and human model systems. Exp. Neurol. 2020, 324, 113121. [Google Scholar] [CrossRef] [PubMed]
  61. Cheng, H.L.; Molassiotis, A.; Leung, A.K.T.; Wong, K.H. Docetaxel-Induced Peripheral Neuropathy in Breast Cancer Patients Treated with Adjuvant or Neo-Adjuvant Chemotherapy. Breast Care 2021, 16, 269–275. [Google Scholar] [CrossRef] [PubMed]
  62. Yamamoto, S.; Egashira, N. Pathological Mechanisms of Bortezomib-Induced Peripheral Neuropathy. Int. J. Mol. Sci. 2021, 22, 888. [Google Scholar] [CrossRef] [PubMed]
  63. Cundari, S.; Cavaletti, G. Thalidomide chemotherapy-induced peripheral neuropathy: Actual status and new perspectives with thalidomide analogues derivatives. Mini Rev. Med. Chem. 2009, 9, 760–768. [Google Scholar] [CrossRef] [PubMed]
  64. 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. [Google Scholar] [CrossRef] [PubMed]
  65. Ollodart, J.; Steele, L.R.; Romero-Sandoval, E.A.; Strowd, R.E.; Shiozawa, Y. Contributions of neuroimmune interactions to chemotherapy-induced peripheral neuropathy development and its prevention/therapy. Biochem. Pharmacol. 2024, 222, 116070. [Google Scholar] [CrossRef] [PubMed]
  66. Chen, X.; Gan, Y.; Au, N.P.B.; Ma, C.H.E. Current understanding of the molecular mechanisms of chemotherapy-induced peripheral neuropathy. Front. Mol. Neurosci. 2024, 17, 1345811. [Google Scholar] [CrossRef] [PubMed]
  67. Kacem, H.; Cimini, A.; d’Angelo, M.; Castelli, V. Molecular and Cellular Involvement in CIPN. Biomedicines 2024, 12, 751. [Google Scholar] [CrossRef] [PubMed]
  68. Zheng, H.; Xiao, W.H.; Bennett, G.J. Mitotoxicity and bortezomib-induced chronic painful peripheral neuropathy. Exp. Neurol. 2012, 238, 225–234. [Google Scholar] [CrossRef] [PubMed]
  69. Zheng, H.; Xiao, W.H.; Bennett, G.J. Functional deficits in peripheral nerve mitochondria in rats with paclitaxel- and oxaliplatin-evoked painful peripheral neuropathy. Exp. Neurol. 2011, 232, 154–161. [Google Scholar] [CrossRef] [PubMed]
  70. Xiao, W.H.; Bennett, G.J. Effects of mitochondrial poisons on the neuropathic pain produced by the chemotherapeutic agents, paclitaxel and oxaliplatin. Pain 2012, 153, 704–709. [Google Scholar] [CrossRef]
  71. Xiao, W.H.; Zheng, H.; Bennett, G.J. Characterization of oxaliplatin-induced chronic painful peripheral neuropathy in the rat and comparison with the neuropathy induced by paclitaxel. Neuroscience 2012, 203, 194–206. [Google Scholar] [CrossRef]
  72. De Grandis, D. Acetyl-L-carnitine for the treatment of chemotherapy-induced peripheral neuropathy: A short review. CNS Drugs 2007, 21 (Suppl. S1), 39–43; discussion 36–45. [Google Scholar] [CrossRef] [PubMed]
  73. Hershman, D.L.; Unger, J.M.; Crew, K.D.; Minasian, L.M.; Awad, D.; Moinpour, C.M.; Hansen, L.; Lew, D.L.; Greenlee, H.; Fehrenbacher, L.; et al. Randomized double-blind placebo-controlled trial of acetyl-L-carnitine for the prevention of taxane-induced neuropathy in women undergoing adjuvant breast cancer therapy. J. Clin. Oncol. 2013, 31, 2627–2633. [Google Scholar] [CrossRef] [PubMed]
  74. Ohsawa, M.; Otake, S.; Murakami, T.; Yamamoto, S.; Makino, T.; Ono, H. Gabapentin prevents oxaliplatin-induced mechanical hyperalgesia in mice. J. Pharmacol. Sci. 2014, 125, 292–299. [Google Scholar] [CrossRef] [PubMed]
  75. Kato, N.; Tateishi, K.; Tsubaki, M.; Takeda, T.; Matsumoto, M.; Tsurushima, K.; Ishizaka, T.; Nishida, S. Gabapentin and Duloxetine Prevent Oxaliplatin- and Paclitaxel-Induced Peripheral Neuropathy by Inhibiting Extracellular Signal-Regulated Kinase 1/2 (ERK1/2) Phosphorylation in Spinal Cords of Mice. Pharmaceuticals 2020, 14, 30. [Google Scholar] [CrossRef] [PubMed]
  76. Kawakami, K.; Chiba, T.; Katagiri, N.; Saduka, M.; Abe, K.; Utsunomiya, I.; Hama, T.; Taguchi, K. Paclitaxel increases high voltage-dependent calcium channel current in dorsal root ganglion neurons of the rat. J. Pharmacol. Sci. 2012, 120, 187–195. [Google Scholar] [CrossRef] [PubMed]
  77. Yamamoto, K.; Tsuboi, M.; Kambe, T.; Abe, K.; Nakatani, Y.; Kawakami, K.; Utsunomiya, I.; Taguchi, K. Oxaliplatin administration increases expression of the voltage-dependent calcium channel α2δ-1 subunit in the rat spinal cord. J. Pharmacol. Sci. 2016, 130, 117–122. [Google Scholar] [CrossRef]
  78. Pandey, P.; Kumar, A.; Pushpam, D.; Khurana, S.; Malik, P.S.; Gogia, A.; Arunmozhimaran, E.; Singh, M.B.; Chandran, D.S.; Batra, A. Randomized double-blind, placebo-controlled study of oral gabapentin for prevention of neuropathy in patients receiving paclitaxel. Trials 2023, 24, 79. [Google Scholar] [CrossRef] [PubMed]
  79. Fumagalli, G.; Monza, L.; Cavaletti, G.; Rigolio, R.; Meregalli, C. Neuroinflammatory Process Involved in Different Preclinical Models of Chemotherapy-Induced Peripheral Neuropathy. Front. Immunol. 2020, 11, 626687. [Google Scholar] [CrossRef] [PubMed]
  80. Costa-Pereira, J.T.; Oliveira, R.; Guadilla, I.; Guillen, M.J.; Tavares, I.; Lopez-Larrubia, P. Neuroimaging uncovers neuronal and metabolic changes in pain modulatory brain areas in a rat model of chemotherapy-induced neuropathy—MEMRI and ex vivo spectroscopy studies. Brain Res. Bull. 2023, 192, 12–20. [Google Scholar] [CrossRef] [PubMed]
  81. Wood, J.; Stein, C.; Gaveriaux-Ruff, C. The Oxford Handbook of the Neurobiology of Pain; Oxford University Press: Oxford, UK, 2020. [Google Scholar] [CrossRef]
  82. Odling-Smee, L. Chronic pain can be treated—So why are millions still suffering? Nature 2023, 615, 782–786. [Google Scholar] [CrossRef] [PubMed]
  83. Barroso, J.; Branco, P.; Pinto-Ramos, J.; Vigotsky, A.D.; Reis, A.M.; Schnitzer, T.J.; Galhardo, V.; Apkarian, A.V. Subcortical brain anatomy as a potential biomarker of persistent pain after total knee replacement in osteoarthritis. Pain 2023, 164, 2306–2315. [Google Scholar] [CrossRef] [PubMed]
  84. Barroso, J.; Wakaizumi, K.; Reis, A.M.; Baliki, M.; Schnitzer, T.J.; Galhardo, V.; Apkarian, A.V. Reorganization of functional brain network architecture in chronic osteoarthritis pain. Hum. Brain Mapp. 2021, 42, 1206–1222. [Google Scholar] [CrossRef] [PubMed]
  85. Zhang, C.; Hu, H.; Das, S.K.; Yang, M.J.; Li, B.; Li, Y.; Xu, X.X.; Yang, H.F. Structural and Functional Brain Abnormalities in Trigeminal Neuralgia: A Systematic Review. J. Oral. Facial Pain Headache 2020, 34, 222–235. [Google Scholar] [CrossRef] [PubMed]
  86. Tang, Y.; Wang, M.; Zheng, T.; Xiao, Y.; Wang, S.; Han, F.; Chen, G. Structural and functional brain abnormalities in postherpetic neuralgia: A systematic review of neuroimaging studies. Brain Res. 2021, 1752, 147219. [Google Scholar] [CrossRef]
  87. Medrano-Escalada, Y.; Plaza-Manzano, G.; Fernández-de-Las-Peñas, C.; Valera-Calero, J.A. Structural, Functional and Neurochemical Cortical Brain Changes Associated with Chronic Low Back Pain. Tomography 2022, 8, 2153–2163. [Google Scholar] [CrossRef] [PubMed]
  88. Chao, C.C.; Tseng, M.T.; Hsieh, P.C.; Lin, C.J.; Huang, S.L.; Hsieh, S.T.; Chiang, M.C. Brain Mechanisms of Pain and Dysautonomia in Diabetic Neuropathy: Connectivity Changes in Thalamus and Hypothalamus. J. Clin. Endocrinol. Metab. 2022, 107, e1167–e1180. [Google Scholar] [CrossRef] [PubMed]
  89. Fischer, T.Z.; Waxman, S.G. Neuropathic pain in diabetes--evidence for a central mechanism. Nat. Rev. Neurol. 2010, 6, 462–466. [Google Scholar] [CrossRef] [PubMed]
  90. Nudelman, K.N.; McDonald, B.C.; Wang, Y.; Smith, D.J.; West, J.D.; O’Neill, D.P.; Zanville, N.R.; Champion, V.L.; Schneider, B.P.; Saykin, A.J. Cerebral Perfusion and Gray Matter Changes Associated With Chemotherapy-Induced Peripheral Neuropathy. J. Clin. Oncol. 2016, 34, 677–683. [Google Scholar] [CrossRef] [PubMed]
  91. Boland, E.G.; Selvarajah, D.; Hunter, M.; Ezaydi, Y.; Tesfaye, S.; Ahmedzai, S.H.; Snowden, J.A.; Wilkinson, I.D. Central pain processing in chronic chemotherapy-induced peripheral neuropathy: A functional magnetic resonance imaging study. PLoS ONE 2014, 9, e96474. [Google Scholar] [CrossRef] [PubMed]
  92. Seretny, M.; Romaniuk, L.; Whalley, H.; Sladdin, K.; Lawrie, S.; Warnaby, C.E.; Roberts, N.; Colvin, L.; Tracey, I.; Fallon, M. Neuroimaging reveals a potential brain-based pre-existing mechanism that confers vulnerability to development of chronic painful chemotherapy-induced peripheral neuropathy. Br. J. Anaesth. 2023, 130, 83–93. [Google Scholar] [CrossRef]
  93. Bacalhau, C.; Costa-Pereira, J.T.; Tavares, I. Preclinical research in paclitaxel-induced neuropathic pain: A systematic review. Front. Vet. Sci. 2023, 10, 1264668. [Google Scholar] [CrossRef] [PubMed]
  94. Ferris, C.F.; Nodine, S.; Pottala, T.; Cai, X.; Knox, T.M.; Fofana, F.H.; Kim, S.; Kulkarni, P.; Crystal, J.D.; Hohmann, A.G. Alterations in brain neurocircuitry following treatment with the chemotherapeutic agent paclitaxel in rats. Neurobiol. Pain 2019, 6, 100034. [Google Scholar] [CrossRef] [PubMed]
  95. Nagasaka, K.; Yamanaka, K.; Ogawa, S.; Takamatsu, H.; Higo, N. Brain activity changes in a macaque model of oxaliplatin-induced neuropathic cold hypersensitivity. Sci. Rep. 2017, 7, 4305. [Google Scholar] [CrossRef] [PubMed]
  96. Shidahara, Y.; Natsume, T.; Awaga, Y.; Ogawa, S.; Yamoto, K.; Okamoto, S.; Hama, A.; Hayashi, I.; Takamatsu, H.; Magata, Y. Distinguishing analgesic drugs from non-analgesic drugs based on brain activation in macaques with oxaliplatin-induced neuropathic pain. Neuropharmacology 2019, 149, 204–211. [Google Scholar] [CrossRef] [PubMed]
  97. Rizvi, T.A.; Ennis, M.; Behbehani, M.M.; Shipley, M.T. Connections between the central nucleus of the amygdala and the midbrain periaqueductal gray: Topography and reciprocity. J. Comp. Neurol. 1991, 303, 121–131. [Google Scholar] [CrossRef]
  98. Bandler, R.; Keay, K.A. Columnar organization in the midbrain periaqueductal gray and the integration of emotional expression. Prog. Brain Res. 1996, 107, 285–300. [Google Scholar] [CrossRef] [PubMed]
  99. Floyd, N.S.; Price, J.L.; Ferry, A.T.; Keay, K.A.; Bandler, R. Orbitomedial prefrontal cortical projections to distinct longitudinal columns of the periaqueductal gray in the rat. J. Comp. Neurol. 2000, 422, 556–578. [Google Scholar] [CrossRef]
  100. Ennis, M.; Behbehani, M.; Shipley, M.T.; Van Bockstaele, E.J.; Aston-Jones, G. Projections from the periaqueductal gray to the rostromedial pericoerulear region and nucleus locus coeruleus: Anatomic and physiologic studies. J. Comp. Neurol. 1991, 306, 480–494. [Google Scholar] [CrossRef]
  101. Basbaum, A.I.; Fields, H.L. Endogenous pain control systems: Brainstem spinal pathways and endorphin circuitry. Annu. Rev. Neurosci. 1984, 7, 309–338. [Google Scholar] [CrossRef] [PubMed]
  102. Almeida, A.; Cobos, A.; Tavares, I.; Lima, D. Brain afferents to the medullary dorsal reticular nucleus: A retrograde and anterograde tracing study in the rat. Eur. J. Neurosci. 2002, 16, 81–95. [Google Scholar] [CrossRef]
  103. Cobos, A.; Lima, D.; Almeida, A.; Tavares, I. Brain afferents to the lateral caudal ventrolateral medulla: A retrograde and anterograde tracing study in the rat. Neuroscience 2003, 120, 485–498. [Google Scholar] [CrossRef]
  104. Behbehani, M.M.; Fields, H.L. Evidence that an excitatory connection between the periaqueductal gray and nucleus raphe magnus mediates stimulation produced analgesia. Brain Res. 1979, 170, 85–93. [Google Scholar] [CrossRef]
  105. Porreca, F.; Ossipov, M.H.; Gebhart, G.F. Chronic pain and medullary descending facilitation. Trends Neurosci. 2002, 25, 319–325. [Google Scholar] [CrossRef]
  106. Bajic, D.; Proudfit, H.K. Projections of neurons in the periaqueductal gray to pontine and medullary catecholamine cell groups involved in the modulation of nociception. J. Comp. Neurol. 1999, 405, 359–379. [Google Scholar] [CrossRef]
  107. Porreca, F.; Navratilova, E. Reward, motivation, and emotion of pain and its relief. Pain 2017, 158 (Suppl. S1), S43–S49. [Google Scholar] [CrossRef]
  108. Song, Q.; Wei, A.; Xu, H.; Gu, Y.; Jiang, Y.; Dong, N.; Zheng, C.; Wang, Q.; Gao, M.; Sun, S.; et al. An ACC-VTA-ACC positive-feedback loop mediates the persistence of neuropathic pain and emotional consequences. Nat. Neurosci. 2024, 27, 272–285. [Google Scholar] [CrossRef]
  109. Juarez-Salinas, D.L.; Braz, J.M.; Etlin, A.; Gee, S.; Sohal, V.; Basbaum, A.I. GABAergic cell transplants in the anterior cingulate cortex reduce neuropathic pain aversiveness. Brain 2019, 142, 2655–2669. [Google Scholar] [CrossRef]
  110. Finnerup, N.B.; Attal, N.; Haroutounian, S.; McNicol, E.; Baron, R.; Dworkin, R.H.; Gilron, I.; Haanpaa, M.; Hansson, P.; Jensen, T.S.; et al. Pharmacotherapy for neuropathic pain in adults: A systematic review and meta-analysis. Lancet Neurol. 2015, 14, 162–173. [Google Scholar] [CrossRef]
  111. Da Silva, J.T.; Seminowicz, D.A. Neuroimaging of pain in animal models: A review of recent literature. Pain Rep. 2019, 4, e732. [Google Scholar] [CrossRef]
  112. Thompson, S.J.; Bushnell, M.C. Rodent functional and anatomical imaging of pain. Neurosci. Lett. 2012, 520, 131–139. [Google Scholar] [CrossRef]
  113. Morris, L.S.; Sprenger, C.; Koda, K.; de la Mora, D.M.; Yamada, T.; Mano, H.; Kashiwagi, Y.; Yoshioka, Y.; Morioka, Y.; Seymour, B. Anterior cingulate cortex connectivity is associated with suppression of behaviour in a rat model of chronic pain. Brain Neurosci. Adv. 2018, 2, 2398212818779646. [Google Scholar] [CrossRef]
  114. Chang, P.C.; Centeno, M.V.; Procissi, D.; Baria, A.; Apkarian, A.V. Brain activity for tactile allodynia: A longitudinal awake rat functional magnetic resonance imaging study tracking emergence of neuropathic pain. Pain 2017, 158, 488–497. [Google Scholar] [CrossRef]
  115. Jeong, K.Y.; Kang, J.H. Investigation of spinal nerve ligation-mediated functional activation of the rat brain using manganese-enhanced MRI. Exp. Anim. 2018, 67, 23–29. [Google Scholar] [CrossRef]
  116. Onishi, O.; Ikoma, K.; Oda, R.; Yamazaki, T.; Fujiwara, H.; Yamada, S.; Tanaka, M.; Kubo, T. Sequential variation in brain functional magnetic resonance imaging after peripheral nerve injury: A rat study. Neurosci. Lett. 2018, 673, 150–156. [Google Scholar] [CrossRef]
  117. Yang, Y.H.; Lin, J.K.; Chen, W.S.; Lin, T.C.; Yang, S.H.; Jiang, J.K.; Chang, S.C.; Lan, Y.T.; Lin, C.C.; Yen, C.C.; et al. Duloxetine improves oxaliplatin-induced neuropathy in patients with colorectal cancer: An open-label pilot study. Support. Care Cancer 2012, 20, 1491–1497. [Google Scholar] [CrossRef]
  118. Costa-Pereira, J.T.; Serrao, P.; Martins, I.; Tavares, I. Serotoninergic pain modulation from the rostral ventromedial medulla (RVM) in chemotherapy-induced neuropathy: The role of spinal 5-HT3 receptors. Eur. J. Neurosci. 2020, 51, 1756–1769. [Google Scholar] [CrossRef]
  119. Liu, X.; Wang, G.; Ai, G.; Xu, X.; Niu, X.; Zhang, M. Selective Ablation of Descending Serotonin from the Rostral Ventromedial Medulla Unmasks Its Pro-Nociceptive Role in Chemotherapy-Induced Painful Neuropathy. J. Pain Res. 2020, 13, 3081–3094. [Google Scholar] [CrossRef]
  120. Gang, J.; Park, K.T.; Kim, S.; Kim, W. Involvement of the Spinal Serotonergic System in the Analgesic Effect of [6]-Shogaol in Oxaliplatin-Induced Neuropathic Pain in Mice. Pharmaceuticals 2023, 16, 1465. [Google Scholar] [CrossRef]
  121. Kim, Y.O.; Song, J.A.; Kim, W.M.; Yoon, M.H. Antiallodynic Effect of Intrathecal Korean Red Ginseng in Cisplatin-Induced Neuropathic Pain Rats. Pharmacology 2020, 105, 173–180. [Google Scholar] [CrossRef]
  122. Lee, J.H.; Min, D.; Lee, D.; Kim, W. Zingiber officinale Roscoe Rhizomes Attenuate Oxaliplatin-Induced Neuropathic Pain in Mice. Molecules 2021, 26, 548. [Google Scholar] [CrossRef]
  123. Andoh, T.; Sakamoto, A.; Kuraishi, Y. Effects of xaliproden, a 5-HT(1)A agonist, on mechanical allodynia caused by chemotherapeutic agents in mice. Eur. J. Pharmacol. 2013, 721, 231–236. [Google Scholar] [CrossRef]
  124. Thibault, K.; Van Steenwinckel, J.; Brisorgueil, M.J.; Fischer, J.; Hamon, M.; Calvino, B.; Conrath, M. Serotonin 5-HT2A receptor involvement and Fos expression at the spinal level in vincristine-induced neuropathy in the rat. Pain 2008, 140, 305–322. [Google Scholar] [CrossRef]
  125. Chenaf, C.; Chapuy, E.; Libert, F.; Marchand, F.; Courteix, C.; Bertrand, M.; Gabriel, C.; Mocaer, E.; Eschalier, A.; Authier, N. Agomelatine: A new opportunity to reduce neuropathic pain-preclinical evidence. Pain 2017, 158, 149–160. [Google Scholar] [CrossRef]
  126. Usman, M.; Malik, H.; Tokhi, A.; Arif, M.; Huma, Z.; Rauf, K.; Sewell, R.D.E. 5,7-Dimethoxycoumarin ameliorates vincristine induced neuropathic pain: Potential role of 5HT(3) receptors and monoamines. Front. Pharmacol. 2023, 14, 1213763. [Google Scholar] [CrossRef]
  127. Park, K.T.; Kim, S.; Choi, I.; Han, I.H.; Bae, H.; Kim, W. The involvement of the noradrenergic system in the antinociceptive effect of cucurbitacin D on mice with paclitaxel-induced neuropathic pain. Front. Pharmacol. 2022, 13, 1055264. [Google Scholar] [CrossRef]
  128. Costa-Pereira, J.T.; Ribeiro, J.; Martins, I.; Tavares, I. Role of Spinal Cord alpha(2)-Adrenoreceptors in Noradrenergic Inhibition of Nociceptive Transmission during Chemotherapy-Induced Peripheral Neuropathy. Front. Neurosci. 2019, 13, 1413. [Google Scholar] [CrossRef]
  129. Messing, R.B.; Lytle, L.D. Serotonin-containing neurons: Their possible role in pain and analgesia. Pain 1977, 4, 1–21. [Google Scholar] [CrossRef]
  130. Dogrul, A.; Ossipov, M.H.; Porreca, F. Differential mediation of descending pain facilitation and inhibition by spinal 5HT-3 and 5HT-7 receptors. Brain Res. 2009, 1280, 52–59. [Google Scholar] [CrossRef]
  131. Viisanen, H.; Pertovaara, A. Roles of the rostroventromedial medulla and the spinal 5-HT(1A) receptor in descending antinociception induced by motor cortex stimulation in the neuropathic rat. Neurosci. Lett. 2010, 476, 133–137. [Google Scholar] [CrossRef]
  132. Rahman, W.; Bannister, K.; Bee, L.A.; Dickenson, A.H. A pronociceptive role for the 5-HT2 receptor on spinal nociceptive transmission: An in vivo electrophysiological study in the rat. Brain Res. 2011, 1382, 29–36. [Google Scholar] [CrossRef]
  133. Bardoni, R. Serotonergic Modulation of Nociceptive Circuits in Spinal Cord Dorsal Horn. Curr. Neuropharmacol. 2019, 17, 1133–1145. [Google Scholar] [CrossRef]
  134. Wei, F.; Dubner, R.; Zou, S.; Ren, K.; Bai, G.; Wei, D.; Guo, W. Molecular depletion of descending serotonin unmasks its novel facilitatory role in the development of persistent pain. J. Neurosci. 2010, 30, 8624–8636. [Google Scholar] [CrossRef]
  135. Rahman, W.; Suzuki, R.; Webber, M.; Hunt, S.P.; Dickenson, A.H. Depletion of endogenous spinal 5-HT attenuates the behavioural hypersensitivity to mechanical and cooling stimuli induced by spinal nerve ligation. Pain 2006, 123, 264–274. [Google Scholar] [CrossRef]
  136. Satoh, O.; Omote, K. Roles of monoaminergic, glycinergic and GABAergic inhibitory systems in the spinal cord in rats with peripheral mononeuropathy. Brain Res. 1996, 728, 27–36. [Google Scholar] [CrossRef]
  137. Morgado, C.; Silva, L.; Pereira-Terra, P.; Tavares, I. Changes in serotoninergic and noradrenergic descending pain pathways during painful diabetic neuropathy: The preventive action of IGF1. Neurobiol. Dis. 2011, 43, 275–284. [Google Scholar] [CrossRef]
  138. Zhang, Y.; Huang, X.; Xin, W.J.; He, S.; Deng, J.; Ruan, X. Somatostatin Neurons from Periaqueductal Gray to Medulla Facilitate Neuropathic Pain in Male Mice. J. Pain 2023, 24, 1020–1029. [Google Scholar] [CrossRef]
  139. White, D.; Abdulla, M.; Park, S.B.; Goldstein, D.; Moalem-Taylor, G.; Lees, J.G. Targeting translation: A review of preclinical animal models in the development of treatments for chemotherapy-induced peripheral neuropathy. J. Peripher. Nerv. Syst. 2023, 28, 179–190. [Google Scholar] [CrossRef]
  140. Andoh, T.; Sakamoto, A.; Kuraishi, Y. 5-HT1A receptor agonists, xaliproden and tandospirone, inhibit the increase in the number of cutaneous mast cells involved in the exacerbation of mechanical allodynia in oxaliplatin-treated mice. J. Pharmacol. Sci. 2016, 131, 284–287. [Google Scholar] [CrossRef]
  141. Salat, K.; Kolaczkowski, M.; Furgala, A.; Rojek, A.; Sniecikowska, J.; Varney, M.A.; Newman-Tancredi, A. Antinociceptive, antiallodynic and antihyperalgesic effects of the 5-HT(1A) receptor selective agonist, NLX-112 in mouse models of pain. Neuropharmacology 2017, 125, 181–188. [Google Scholar] [CrossRef]
  142. Rapacz, A.; Obniska, J.; Koczurkiewicz, P.; Wojcik-Pszczola, K.; Siwek, A.; Grybos, A.; Rybka, S.; Karcz, A.; Pekala, E.; Filipek, B. Antiallodynic and antihyperalgesic activity of new 3,3-diphenyl-propionamides with anticonvulsant activity in models of pain in mice. Eur. J. Pharmacol. 2018, 821, 39–48. [Google Scholar] [CrossRef]
  143. Masuguchi, K.; Watanabe, H.; Kawashiri, T.; Ushio, S.; Ozawa, N.; Morita, H.; Oishi, R.; Egashira, N. Neurotropin(R) relieves oxaliplatin-induced neuropathy via Gi protein-coupled receptors in the monoaminergic descending pain inhibitory system. Life Sci. 2014, 98, 49–54. [Google Scholar] [CrossRef]
  144. Zeitz, K.P.; Guy, N.; Malmberg, A.B.; Dirajlal, S.; Martin, W.J.; Sun, L.; Bonhaus, D.W.; Stucky, C.L.; Julius, D.; Basbaum, A.I. The 5-HT3 subtype of serotonin receptor contributes to nociceptive processing via a novel subset of myelinated and unmyelinated nociceptors. J. Neurosci. 2002, 22, 1010–1019. [Google Scholar] [CrossRef]
  145. Guo, W.; Miyoshi, K.; Dubner, R.; Gu, M.; Li, M.; Liu, J.; Yang, J.; Zou, S.; Ren, K.; Noguchi, K.; et al. Spinal 5-HT3 receptors mediate descending facilitation and contribute to behavioral hypersensitivity via a reciprocal neuron-glial signaling cascade. Mol. Pain 2014, 10, 35. [Google Scholar] [CrossRef]
  146. Pertovaara, A. Noradrenergic pain modulation. Prog. Neurobiol. 2006, 80, 53–83. [Google Scholar] [CrossRef]
  147. Clark, F.M.; Proudfit, H.K. The projection of noradrenergic neurons in the A7 catecholamine cell group to the spinal cord in the rat demonstrated by anterograde tracing combined with immunocytochemistry. Brain Res. 1991, 547, 279–288. [Google Scholar] [CrossRef]
  148. Clark, F.M.; Proudfit, H.K. The projections of noradrenergic neurons in the A5 catecholamine cell group to the spinal cord in the rat: Anatomical evidence that A5 neurons modulate nociception. Brain Res. 1993, 616, 200–210. [Google Scholar] [CrossRef]
  149. Fritschy, J.M.; Lyons, W.E.; Mullen, C.A.; Kosofsky, B.E.; Molliver, M.E.; Grzanna, R. Distribution of locus coeruleus axons in the rat spinal cord: A combined anterograde transport and immunohistochemical study. Brain Res. 1987, 437, 176–180. [Google Scholar] [CrossRef]
  150. Tavares, I.; Lima, D.; Coimbra, A. The ventrolateral medulla of the rat is connected with the spinal cord dorsal horn by an indirect descending pathway relayed in the A5 noradrenergic cell group. J. Comp. Neurol. 1996, 374, 84–95. [Google Scholar] [CrossRef]
  151. Westlund, K.N.; Bowker, R.M.; Ziegler, M.G.; Coulter, J.D. Noradrenergic projections to the spinal cord of the rat. Brain Res. 1983, 263, 15–31. [Google Scholar] [CrossRef]
  152. Kwiat, G.C.; Basbaum, A.I. The origin of brainstem noradrenergic and serotonergic projections to the spinal cord dorsal horn in the rat. Somatosens. Mot. Res. 1992, 9, 157–173. [Google Scholar] [CrossRef]
  153. Fairbanks, C.A.; Stone, L.S.; Wilcox, G.L. Pharmacological profiles of alpha 2 adrenergic receptor agonists identified using genetically altered mice and isobolographic analysis. Pharmacol. Ther. 2009, 123, 224–238. [Google Scholar] [CrossRef] [PubMed]
  154. Summers, R.J.; McMartin, L.R. Adrenoceptors and their second messenger systems. J. Neurochem. 1993, 60, 10–23. [Google Scholar] [CrossRef] [PubMed]
  155. Llorca-Torralba, M.; Borges, G.; Neto, F.; Mico, J.A.; Berrocoso, E. Noradrenergic Locus Coeruleus pathways in pain modulation. Neuroscience 2016, 338, 93–113. [Google Scholar] [CrossRef] [PubMed]
  156. Stone, L.S.; Broberger, C.; Vulchanova, L.; Wilcox, G.L.; Hokfelt, T.; Riedl, M.S.; Elde, R. Differential distribution of alpha2A and alpha2C adrenergic receptor immunoreactivity in the rat spinal cord. J. Neurosci. 1998, 18, 5928–5937. [Google Scholar] [CrossRef] [PubMed]
  157. Olave, M.J.; Maxwell, D.J. Axon terminals possessing the alpha 2c-adrenergic receptor in the rat dorsal horn are predominantly excitatory. Brain Res. 2003, 965, 269–273. [Google Scholar] [CrossRef] [PubMed]
  158. Viisanen, H.; Pertovaara, A. Influence of peripheral nerve injury on response properties of locus coeruleus neurons and coeruleospinal antinociception in the rat. Neuroscience 2007, 146, 1785–1794. [Google Scholar] [CrossRef] [PubMed]
  159. Wei, H.; Pertovaara, A. Regulation of neuropathic hypersensitivity by alpha(2)-adrenoceptors in the pontine A7 cell group. Basic. Clin. Pharmacol. Toxicol. 2013, 112, 90–95. [Google Scholar] [CrossRef] [PubMed]
  160. Pertovaara, A. The noradrenergic pain regulation system: A potential target for pain therapy. Eur. J. Pharmacol. 2013, 716, 2–7. [Google Scholar] [CrossRef] [PubMed]
  161. Rahman, W.; D’Mello, R.; Dickenson, A.H. Peripheral nerve injury-induced changes in spinal alpha(2)-adrenoceptor-mediated modulation of mechanically evoked dorsal horn neuronal responses. J. Pain 2008, 9, 350–359. [Google Scholar] [CrossRef] [PubMed]
  162. Bantel, C.; Eisenach, J.C.; Duflo, F.; Tobin, J.R.; Childers, S.R. Spinal nerve ligation increases alpha2-adrenergic receptor G-protein coupling in the spinal cord. Brain Res. 2005, 1038, 76–82. [Google Scholar] [CrossRef] [PubMed]
  163. Ma, W.; Eisenach, J.C. Chronic constriction injury of sciatic nerve induces the up-regulation of descending inhibitory noradrenergic innervation to the lumbar dorsal horn of mice. Brain Res. 2003, 970, 110–118. [Google Scholar] [CrossRef] [PubMed]
  164. Kawasaki, Y.; Kumamoto, E.; Furue, H.; Yoshimura, M. Alpha 2 adrenoceptor-mediated presynaptic inhibition of primary afferent glutamatergic transmission in rat substantia gelatinosa neurons. Anesthesiology 2003, 98, 682–689. [Google Scholar] [CrossRef] [PubMed]
  165. Choi, S.; Yamada, A.; Kim, W.; Kim, S.K.; Furue, H. Noradrenergic inhibition of spinal hyperexcitation elicited by cutaneous cold stimuli in rats with oxaliplatin-induced allodynia: Electrophysiological and behavioral assessments. J. Physiol. Sci. 2017, 67, 431–438. [Google Scholar] [CrossRef] [PubMed]
  166. Juarez-Salinas, D.L.; Braz, J.M.; Hamel, K.A.; Basbaum, A.I. Pain relief by supraspinal gabapentin requires descending noradrenergic inhibitory controls. Pain Rep. 2018, 3, e659. [Google Scholar] [CrossRef] [PubMed]
  167. de Novellis, V.; Mariani, L.; Palazzo, E.; Vita, D.; Marabese, I.; Scafuro, M.; Rossi, F.; Maione, S. Periaqueductal grey CB1 cannabinoid and metabotropic glutamate subtype 5 receptors modulate changes in rostral ventromedial medulla neuronal activities induced by subcutaneous formalin in the rat. Neuroscience 2005, 134, 269–281. [Google Scholar] [CrossRef] [PubMed]
  168. Blanton, H.L.; Brelsfoard, J.; DeTurk, N.; Pruitt, K.; Narasimhan, M.; Morgan, D.J.; Guindon, J. Cannabinoids: Current and Future Options to Treat Chronic and Chemotherapy-Induced Neuropathic Pain. Drugs 2019, 79, 969–995. [Google Scholar] [CrossRef] [PubMed]
  169. Burston, J.J.; Woodhams, S.G. Endocannabinoid system and pain: An introduction. Proc. Nutr. Soc. 2014, 73, 106–117. [Google Scholar] [CrossRef] [PubMed]
  170. Howlett, A.C. The cannabinoid receptors. Prostaglandins Other Lipid Mediat. 2002, 68–69, 619–631. [Google Scholar] [CrossRef] [PubMed]
  171. Vasquez, C.; Lewis, D.L. The CB1 cannabinoid receptor can sequester G-proteins, making them unavailable to couple to other receptors. J. Neurosci. 1999, 19, 9271–9280. [Google Scholar] [CrossRef] [PubMed]
  172. Rozenfeld, R.; Bushlin, I.; Gomes, I.; Tzavaras, N.; Gupta, A.; Neves, S.; Battini, L.; Gusella, G.L.; Lachmann, A.; Ma’ayan, A.; et al. Receptor heteromerization expands the repertoire of cannabinoid signaling in rodent neurons. PLoS ONE 2012, 7, e29239. [Google Scholar] [CrossRef] [PubMed]
  173. Vinals, X.; Moreno, E.; Lanfumey, L.; Cordomi, A.; Pastor, A.; de La Torre, R.; Gasperini, P.; Navarro, G.; Howell, L.A.; Pardo, L.; et al. Cognitive Impairment Induced by Delta9-tetrahydrocannabinol Occurs through Heteromers between Cannabinoid CB1 and Serotonin 5-HT2A Receptors. PLoS Biol. 2015, 13, e1002194. [Google Scholar] [CrossRef] [PubMed]
  174. Haspula, D.; Clark, M.A. Cannabinoid Receptors: An Update on Cell Signaling, Pathophysiological Roles and Therapeutic Opportunities in Neurological, Cardiovascular, and Inflammatory Diseases. Int. J. Mol. Sci. 2020, 21, 7693. [Google Scholar] [CrossRef] [PubMed]
  175. Navarrete, M.; Araque, A. Endocannabinoids mediate neuron-astrocyte communication. Neuron 2008, 57, 883–893. [Google Scholar] [CrossRef] [PubMed]
  176. Navarro, G.; Morales, P.; Rodriguez-Cueto, C.; Fernandez-Ruiz, J.; Jagerovic, N.; Franco, R. Targeting Cannabinoid CB2 Receptors in the Central Nervous System. Medicinal Chemistry Approaches with Focus on Neurodegenerative Disorders. Front. Neurosci. 2016, 10, 406. [Google Scholar] [CrossRef]
  177. Stella, N. Cannabinoid and cannabinoid-like receptors in microglia, astrocytes, and astrocytomas. Glia 2010, 58, 1017–1030. [Google Scholar] [CrossRef] [PubMed]
  178. Cabral, G.A.; Ferreira, G.A.; Jamerson, M.J. Endocannabinoids and the Immune System in Health and Disease. Handb. Exp. Pharmacol. 2015, 231, 185–211. [Google Scholar] [CrossRef] [PubMed]
  179. Riquelme-Sandoval, A.; de Sa-Ferreira, C.O.; Miyakoshi, L.M.; Hedin-Pereira, C. New Insights Into Peptide Cannabinoids: Structure, Biosynthesis and Signaling. Front. Pharmacol. 2020, 11, 596572. [Google Scholar] [CrossRef] [PubMed]
  180. Guindon, J.; Hohmann, A.G. Cannabinoid CB2 receptors: A therapeutic target for the treatment of inflammatory and neuropathic pain. Br. J. Pharmacol. 2008, 153, 319–334. [Google Scholar] [CrossRef]
  181. Ghosh, K.; Zhang, G.F.; Chen, H.; Chen, S.R.; Pan, H.L. Cannabinoid CB2 receptors are upregulated via bivalent histone modifications and control primary afferent input to the spinal cord in neuropathic pain. J. Biol. Chem. 2022, 298, 101999. [Google Scholar] [CrossRef] [PubMed]
  182. Zhang, J.; Hoffert, C.; Vu, H.K.; Groblewski, T.; Ahmad, S.; O’Donnell, D. Induction of CB2 receptor expression in the rat spinal cord of neuropathic but not inflammatory chronic pain models. Eur. J. Neurosci. 2003, 17, 2750–2754. [Google Scholar] [CrossRef] [PubMed]
  183. Lin, X.; Xu, Z.; Carey, L.; Romero, J.; Makriyannis, A.; Hillard, C.J.; Ruggiero, E.; Dockum, M.; Houk, G.; Mackie, K.; et al. A peripheral CB2 cannabinoid receptor mechanism suppresses chemotherapy-induced peripheral neuropathy: Evidence from a CB2 reporter mouse. Pain 2022, 163, 834–851. [Google Scholar] [CrossRef] [PubMed]
  184. King, K.M.; Myers, A.M.; Soroka-Monzo, A.J.; Tuma, R.F.; Tallarida, R.J.; Walker, E.A.; Ward, S.J. Single and combined effects of Delta(9)-tetrahydrocannabinol and cannabidiol in a mouse model of chemotherapy-induced neuropathic pain. Br. J. Pharmacol. 2017, 174, 2832–2841. [Google Scholar] [CrossRef] [PubMed]
  185. Deng, L.; Guindon, J.; Vemuri, V.K.; Thakur, G.A.; White, F.A.; Makriyannis, A.; Hohmann, A.G. The maintenance of cisplatin- and paclitaxel-induced mechanical and cold allodynia is suppressed by cannabinoid CB(2) receptor activation and independent of CXCR4 signaling in models of chemotherapy-induced peripheral neuropathy. Mol. Pain 2012, 8, 71. [Google Scholar] [CrossRef] [PubMed]
  186. Pascual, D.; Goicoechea, C.; Suardiaz, M.; Martin, M.I. A cannabinoid agonist, WIN 55,212-2, reduces neuropathic nociception induced by paclitaxel in rats. Pain 2005, 118, 23–34. [Google Scholar] [CrossRef] [PubMed]
  187. Rahn, E.J.; Makriyannis, A.; Hohmann, A.G. Activation of cannabinoid CB1 and CB2 receptors suppresses neuropathic nociception evoked by the chemotherapeutic agent vincristine in rats. Br. J. Pharmacol. 2007, 152, 765–777. [Google Scholar] [CrossRef] [PubMed]
  188. Wu, J.; Hocevar, M.; Bie, B.; Foss, J.F.; Naguib, M. Cannabinoid Type 2 Receptor System Modulates Paclitaxel-Induced Microglial Dysregulation and Central Sensitization in Rats. J. Pain 2019, 20, 501–514. [Google Scholar] [CrossRef] [PubMed]
  189. Sierra, S.; Gupta, A.; Gomes, I.; Fowkes, M.; Ram, A.; Bobeck, E.N.; Devi, L.A. Targeting Cannabinoid 1 and Delta Opioid Receptor Heteromers Alleviates Chemotherapy-Induced Neuropathic Pain. ACS Pharmacol. Transl. Sci. 2019, 2, 219–229. [Google Scholar] [CrossRef] [PubMed]
  190. Matsumoto, M.; Xie, W.; Inoue, M.; Ueda, H. Evidence for the tonic inhibition of spinal pain by nicotinic cholinergic transmission through primary afferents. Mol. Pain 2007, 3, 41. [Google Scholar] [CrossRef] [PubMed]
  191. Naser, P.V.; Kuner, R. Molecular, Cellular and Circuit Basis of Cholinergic Modulation of Pain. Neuroscience 2018, 387, 135–148. [Google Scholar] [CrossRef] [PubMed]
  192. Hone, A.J.; McIntosh, J.M. Nicotinic acetylcholine receptors: Therapeutic targets for novel ligands to treat pain and inflammation. Pharmacol. Res. 2023, 190, 106715. [Google Scholar] [CrossRef] [PubMed]
  193. Picciotto, M.R.; Higley, M.J.; Mineur, Y.S. Acetylcholine as a neuromodulator: Cholinergic signaling shapes nervous system function and behavior. Neuron 2012, 76, 116–129. [Google Scholar] [CrossRef]
  194. Iwamoto, E.T.; Marion, L. Adrenergic, serotonergic and cholinergic components of nicotinic antinociception in rats. J. Pharmacol. Exp. Ther. 1993, 265, 777–789. [Google Scholar] [PubMed]
  195. Ferrier, J.; Bayet-Robert, M.; Dalmann, R.; El Guerrab, A.; Aissouni, Y.; Graveron-Demilly, D.; Chalus, M.; Pinguet, J.; Eschalier, A.; Richard, D.; et al. Cholinergic Neurotransmission in the Posterior Insular Cortex Is Altered in Preclinical Models of Neuropathic Pain: Key Role of Muscarinic M2 Receptors in Donepezil-Induced Antinociception. J. Neurosci. 2015, 35, 16418–16430. [Google Scholar] [CrossRef] [PubMed]
  196. Selvy, M.; Mattevi, C.; Dalbos, C.; Aissouni, Y.; Chapuy, E.; Martin, P.Y.; Collin, A.; Richard, D.; Dumontet, C.; Busserolles, J.; et al. Analgesic and preventive effects of donepezil in animal models of chemotherapy-induced peripheral neuropathy: Involvement of spinal muscarinic acetylcholine M2 receptors. Biomed. Pharmacother. 2022, 149, 112915. [Google Scholar] [CrossRef] [PubMed]
  197. Kanat, O.; Bagdas, D.; Ozboluk, H.Y.; Gurun, M.S. Preclinical evidence for the antihyperalgesic activity of CDP-choline in oxaliplatin-induced neuropathic pain. J. BUON 2013, 18, 1012–1018. [Google Scholar] [PubMed]
  198. Favre-Guilmard, C.; Auguet, M.; Chabrier, P.E. Different antinociceptive effects of botulinum toxin type A in inflammatory and peripheral polyneuropathic rat models. Eur. J. Pharmacol. 2009, 617, 48–53. [Google Scholar] [CrossRef] [PubMed]
  199. Kyte, S.L.; Toma, W.; Bagdas, D.; Meade, J.A.; Schurman, L.D.; Lichtman, A.H.; Chen, Z.J.; Del Fabbro, E.; Fang, X.; Bigbee, J.W.; et al. Nicotine Prevents and Reverses Paclitaxel-Induced Mechanical Allodynia in a Mouse Model of CIPN. J. Pharmacol. Exp. Ther. 2018, 364, 110–119. [Google Scholar] [CrossRef] [PubMed]
  200. Lynch, J.J., 3rd; Wade, C.L.; Mikusa, J.P.; Decker, M.W.; Honore, P. ABT-594 (a nicotinic acetylcholine agonist): Anti-allodynia in a rat chemotherapy-induced pain model. Eur. J. Pharmacol. 2005, 509, 43–48. [Google Scholar] [CrossRef] [PubMed]
  201. Yoon, H.; Kim, M.J.; Yoon, I.; Li, D.X.; Bae, H.; Kim, S.K. Nicotinic Acetylcholine Receptors Mediate the Suppressive Effect of an Injection of Diluted Bee Venom into the GV3 Acupoint on Oxaliplatin-Induced Neuropathic Cold Allodynia in Rats. Biol. Pharm. Bull. 2015, 38, 710–714. [Google Scholar] [CrossRef] [PubMed]
  202. Arias, H.R.; Ghelardini, C.; Lucarini, E.; Tae, H.S.; Yousuf, A.; Marcovich, I.; Manetti, D.; Romanelli, M.N.; Elgoyhen, A.B.; Adams, D.J.; et al. (E)-3-Furan-2-yl-N-p-tolyl-acrylamide and its Derivative DM489 Decrease Neuropathic Pain in Mice Predominantly by alpha7 Nicotinic Acetylcholine Receptor Potentiation. ACS Chem. Neurosci. 2020, 11, 3603–3614. [Google Scholar] [CrossRef] [PubMed]
  203. Di Cesare Mannelli, L.; Pacini, A.; Matera, C.; Zanardelli, M.; Mello, T.; De Amici, M.; Dallanoce, C.; Ghelardini, C. Involvement of alpha7 nAChR subtype in rat oxaliplatin-induced neuropathy: Effects of selective activation. Neuropharmacology 2014, 79, 37–48. [Google Scholar] [CrossRef]
  204. Toma, W.; Kyte, S.L.; Bagdas, D.; Jackson, A.; Meade, J.A.; Rahman, F.; Chen, Z.J.; Del Fabbro, E.; Cantwell, L.; Kulkarni, A.; et al. The alpha7 nicotinic receptor silent agonist R-47 prevents and reverses paclitaxel-induced peripheral neuropathy in mice without tolerance or altering nicotine reward and withdrawal. Exp. Neurol. 2019, 320, 113010. [Google Scholar] [CrossRef] [PubMed]
  205. Christensen, S.B.; Hone, A.J.; Roux, I.; Kniazeff, J.; Pin, J.P.; Upert, G.; Servent, D.; Glowatzki, E.; McIntosh, J.M. RgIA4 Potently Blocks Mouse alpha9alpha10 nAChRs and Provides Long Lasting Protection against Oxaliplatin-Induced Cold Allodynia. Front. Cell Neurosci. 2017, 11, 219. [Google Scholar] [CrossRef]
  206. Dyachenko, I.A.; Palikova, Y.A.; Palikov, V.A.; Korolkova, Y.V.; Kazakov, V.A.; Egorova, N.S.; Garifulina, A.I.; Utkin, Y.N.; Tsetlin, V.I.; Kryukova, E.V. alpha-Conotoxin RgIA and oligoarginine R8 in the mice model alleviate long-term oxaliplatin induced neuropathy. Biochimie 2022, 194, 127–136. [Google Scholar] [CrossRef]
  207. Gajewiak, J.; Christensen, S.B.; Dowell, C.; Hararah, F.; Fisher, F.; Huynh, P.N.; Olivera, B.M.; McIntosh, J.M. Selective Penicillamine Substitution Enables Development of a Potent Analgesic Peptide that Acts through a Non-Opioid-Based Mechanism. J. Med. Chem. 2021, 64, 9271–9278. [Google Scholar] [CrossRef] [PubMed]
  208. Huynh, P.N.; Giuvelis, D.; Christensen, S.; Tucker, K.L.; McIntosh, J.M. RgIA4 Accelerates Recovery from Paclitaxel-Induced Neuropathic Pain in Rats. Mar. Drugs 2019, 18, 12. [Google Scholar] [CrossRef] [PubMed]
  209. Pacini, A.; Micheli, L.; Maresca, M.; Branca, J.J.; McIntosh, J.M.; Ghelardini, C.; Di Cesare Mannelli, L. The alpha9alpha10 nicotinic receptor antagonist alpha-conotoxin RgIA prevents neuropathic pain induced by oxaliplatin treatment. Exp. Neurol. 2016, 282, 37–48. [Google Scholar] [CrossRef]
  210. Romero, H.K.; Christensen, S.B.; Di Cesare Mannelli, L.; Gajewiak, J.; Ramachandra, R.; Elmslie, K.S.; Vetter, D.E.; Ghelardini, C.; Iadonato, S.P.; Mercado, J.L.; et al. Inhibition of alpha9alpha10 nicotinic acetylcholine receptors prevents chemotherapy-induced neuropathic pain. Proc. Natl. Acad. Sci. USA 2017, 114, E1825–E1832. [Google Scholar] [CrossRef]
  211. Li, Z.; Han, X.; Hong, X.; Li, X.; Gao, J.; Zhang, H.; Zheng, A. Lyophilization Serves as an Effective Strategy for Drug Development of the alpha9alpha10 Nicotinic Acetylcholine Receptor Antagonist alpha-Conotoxin GeXIVA[1,2]. Mar. Drugs 2021, 19, 121. [Google Scholar] [CrossRef] [PubMed]
  212. Wala, E.P.; Crooks, P.A.; McIntosh, J.M.; Holtman, J.R., Jr. Novel small molecule alpha9alpha10 nicotinic receptor antagonist prevents and reverses chemotherapy-evoked neuropathic pain in rats. Anesth. Analg. 2012, 115, 713–720. [Google Scholar] [CrossRef] [PubMed]
  213. Wang, H.; Li, X.; Zhangsun, D.; Yu, G.; Su, R.; Luo, S. The alpha9alpha10 Nicotinic Acetylcholine Receptor Antagonist alphaO-Conotoxin GeXIVA[1,2] Alleviates and Reverses Chemotherapy-Induced Neuropathic Pain. Mar. Drugs 2019, 17, 265. [Google Scholar] [CrossRef] [PubMed]
  214. Calcutt, N.A.; Smith, D.R.; Frizzi, K.; Sabbir, M.G.; Chowdhury, S.K.; Mixcoatl-Zecuatl, T.; Saleh, A.; Muttalib, N.; Van der Ploeg, R.; Ochoa, J.; et al. Selective antagonism of muscarinic receptors is neuroprotective in peripheral neuropathy. J. Clin. Investig. 2017, 127, 608–622. [Google Scholar] [CrossRef] [PubMed]
  215. Wood, M.W.; Martino, G.; Coupal, M.; Lindberg, M.; Schroeder, P.; Santhakumar, V.; Valiquette, M.; Sandin, J.; Widzowski, D.; Laird, J. Broad analgesic activity of a novel, selective M1 agonist. Neuropharmacology 2017, 123, 233–241. [Google Scholar] [CrossRef] [PubMed]
  216. Costa, R.; Motta, E.M.; Dutra, R.C.; Manjavachi, M.N.; Bento, A.F.; Malinsky, F.R.; Pesquero, J.B.; Calixto, J.B. Anti-nociceptive effect of kinin B(1) and B(2) receptor antagonists on peripheral neuropathy induced by paclitaxel in mice. Br. J. Pharmacol. 2011, 164, 681–693. [Google Scholar] [CrossRef] [PubMed]
  217. Cavalli, J.; de Assis, P.M.; Cristina Dalazen Goncalves, E.; Daniele Bobermin, L.; Quincozes-Santos, A.; Raposo, N.R.B.; Gomez, M.V.; Dutra, R.C. Systemic, Intrathecal, and Intracerebroventricular Antihyperalgesic Effects of the Calcium Channel Blocker CTK 01512-2 Toxin in Persistent Pain Models. Mol. Neurobiol. 2022, 59, 4436–4452. [Google Scholar] [CrossRef]
  218. Xu, D.; Zhao, H.; Gao, H.; Zhao, H.; Liu, D.; Li, J. Participation of pro-inflammatory cytokines in neuropathic pain evoked by chemotherapeutic oxaliplatin via central GABAergic pathway. Mol. Pain 2018, 14, 1744806918783535. [Google Scholar] [CrossRef]
  219. Hache, G.; Guiard, B.P.; Nguyen, T.H.; Quesseveur, G.; Gardier, A.M.; Peters, D.; Munro, G.; Coudore, F. Antinociceptive activity of the new triple reuptake inhibitor NS18283 in a mouse model of chemotherapy-induced neuropathic pain. Eur. J. Pain 2015, 19, 322–333. [Google Scholar] [CrossRef] [PubMed]
  220. Kanbara, T.; Nakamura, A.; Shibasaki, M.; Mori, T.; Suzuki, T.; Sakaguchi, G.; Kanemasa, T. Morphine and oxycodone, but not fentanyl, exhibit antinociceptive effects mediated by G-protein inwardly rectifying potassium (GIRK) channels in an oxaliplatin-induced neuropathy rat model. Neurosci. Lett. 2014, 580, 119–124. [Google Scholar] [CrossRef] [PubMed]
  221. Kanbara, T.; Nakamura, A.; Takasu, K.; Ogawa, K.; Shibasaki, M.; Mori, T.; Suzuki, T.; Hasegawa, M.; Sakaguchi, G.; Kanemasa, T. The contribution of Gi/o protein to opioid antinociception in an oxaliplatin-induced neuropathy rat model. J. Pharmacol. Sci. 2014, 126, 264–273. [Google Scholar] [CrossRef] [PubMed]
  222. Norcini, M.; Vivoli, E.; Galeotti, N.; Bianchi, E.; Bartolini, A.; Ghelardini, C. Supraspinal role of protein kinase C in oxaliplatin-induced neuropathy in rat. Pain 2009, 146, 141–147. [Google Scholar] [CrossRef] [PubMed]
  223. Toyama, S.; Shimoyama, N.; Shimoyama, M. The analgesic effect of orexin-A in a murine model of chemotherapy-induced neuropathic pain. Neuropeptides 2017, 61, 95–100. [Google Scholar] [CrossRef] [PubMed]
  224. Chung, G.; Kim, S.K. Therapeutics for Chemotherapy-Induced Peripheral Neuropathy: Approaches with Natural Compounds from Traditional Eastern Medicine. Pharmaceutics 2022, 14, 1407. [Google Scholar] [CrossRef] [PubMed]
  225. Balayssac, D.; Durif, J.; Lambert, C.; Dalbos, C.; Chapuy, E.; Etienne, M.; Demiot, C.; Busserolles, J.; Martin, V.; Sapin, V. Exploring Serum Biomarkers for Neuropathic Pain in Rat Models of Chemotherapy-Induced Peripheral Neuropathy: A Comparative Pilot Study with Oxaliplatin, Paclitaxel, Bortezomib, and Vincristine. Toxics 2023, 11, 1004. [Google Scholar] [CrossRef] [PubMed]
  226. Attal, N.; Bouhassira, D.; Colvin, L. Advances and challenges in neuropathic pain: A narrative review and future directions. Br. J. Anaesth. 2023, 131, 79–92. [Google Scholar] [CrossRef] [PubMed]
  227. Bouhassira, D.; Attal, N. Personalized treatment of neuropathic pain: Where are we now? Eur. J. Pain 2023, 27, 1084–1098. [Google Scholar] [CrossRef]
Figure 1. Brain mechanisms accounting for painful CIPN, according mainly to the results of preclinical studies. An increase in the activity of several areas leads to an imbalance of top-down pain modulation, towards facilitation, namely with increased release of -5-HT and NA at the spinal cord. The involvement of relevant circuits, such as the reward system, is under investigation. Abbreviations: Hypot: hypothalamus; LC: locus coeruleus; NAc: nucleus accumbens; PAG: periaqueductal grey; PFC: prefrontal cortex; RVM: rostroventromedial medulla; VTA: ventral tegmental area; purple arrow: descending serotoninergic pain modulation; orange arrow: descending noradrenergic pain modulation; different color lines: conections between brain areas; the symbol “?” shows neuronal circuits in which the effects of CIPN is not fully established.
Figure 1. Brain mechanisms accounting for painful CIPN, according mainly to the results of preclinical studies. An increase in the activity of several areas leads to an imbalance of top-down pain modulation, towards facilitation, namely with increased release of -5-HT and NA at the spinal cord. The involvement of relevant circuits, such as the reward system, is under investigation. Abbreviations: Hypot: hypothalamus; LC: locus coeruleus; NAc: nucleus accumbens; PAG: periaqueductal grey; PFC: prefrontal cortex; RVM: rostroventromedial medulla; VTA: ventral tegmental area; purple arrow: descending serotoninergic pain modulation; orange arrow: descending noradrenergic pain modulation; different color lines: conections between brain areas; the symbol “?” shows neuronal circuits in which the effects of CIPN is not fully established.
Brainsci 14 00659 g001
Figure 2. Brain’s role in CIPN and personalized chemotherapy.
Figure 2. Brain’s role in CIPN and personalized chemotherapy.
Brainsci 14 00659 g002
Table 1. Main findings of the three clinical human studies that have assessed brain plasticity using functional magnetic resonance (fMRI) in CIPN.
Table 1. Main findings of the three clinical human studies that have assessed brain plasticity using functional magnetic resonance (fMRI) in CIPN.
Study DesignType of ChemotherapyEffects of CIPN in Brain Ref.
Longitudinal study:
24 women with chemotherapy,
23 women no chemotherapy
Combinations of paclitaxel, docetaxel, carboplatin, and cisplatin ↑ perfusion in the CG and SFG [90]
Case-control study:
12 patients CIPN
12 healthy volunteers
Bortezomib, thalidomide, or vincristine↑ activation in the precuneus
↓ activation in the SFG
Activation in the FO associated with worse CIPN.
[91]
Prospective, multicenter cohort study:
20 patients
Bortezomib, oxaliplatin, paclitaxel, docetaxel, cisplatin Prior chemotherapy (punctate stimuli):
↑ activity in insula, somatosensory cortex, thalamus and cerebellum in CIPNþ.
↑ activity of PAG in CIPNe
[92]
CG: cingulate gyrus; FO: frontal operculum; SFG: superior frontal gyrus; CIPNþ: CIPN positive; CIPNe: CIPN negative; ↑ increase; ↓ decrease.
Table 2. Main findings of the preclinical studies that have assessed brain plasticity using MRI techniques in CIPN.
Table 2. Main findings of the preclinical studies that have assessed brain plasticity using MRI techniques in CIPN.
Neuroimaging ApproachSpecies (Sex)CIPN ModelMain ResultsRef.
DW imaging—quantitative anisotropyRats (males)PaclitaxelReorganization of gray matter in the PFC, amygdala, hippocampus, hypothalamus and striatum/NAc[94]
Rs functional connectivityRats (males)PaclitaxelAltered connections to the PAG [94]
MEMRIRats (males)Paclitaxel↑ activation of hypothalamus and PAG[80]
Ex vivo spectroscopyRats (males)PaclitaxelEarly CIPN:
↑ NAA levels in PFC
↑ NAA and lactate levels in hypothalamus
Late CIPN:
↓ NAA levels in PFC
↑ taurine levels in PFC
[80]
fMRINon-human primatesOxaliplatin↑ activation of SSC and Insula [95,96]
DW: diffusion weight; PAG: periaqueductal gray matter; PFC: prefrontal cortex; NAA: N-acetyl-aspartate; NAc: nucleus accumbens; SSC: secondary somatosensorial cortex; ↑ increase; ↓ decrease.
Table 3. Main findings of the preclinical studies that have assessed descending serotoninergic and noradrenergic pain modulation during CIPN.
Table 3. Main findings of the preclinical studies that have assessed descending serotoninergic and noradrenergic pain modulation during CIPN.
Neurotransmitter SystemCIPN ModelCNS RegionMain ResultsRef.
SerotoninergicPaclitaxelRVM↑ 5-HT neuron activation [118]
SC↑ 5-HT levels
↑ 5-HT3 receptors
PaclitaxelRVM↑ 5-HT neuron activation[119]
OxaliplatinSC↓ 5-HT levels[120]
CisplatinSC↓ 5-HT levels[121]
OxaliplatinSC↓ 5-HT1A receptors[122,123]
Paclitaxel
Vincristine
SC↔ 5-HT1A receptors[123]
VincristineSC↑ 5-HT2A receptors[124]
OxaliplatinSC↑ 5-HT2C receptors[125]
VincristineFC
Striatum
Hippocampus
↑ 5-HT levels[126]
NoradrenergicPaclitaxelLC↑ TH expression[127]
SC↑ NA levels
↑ α1-AR receptors
↑ α2-AR receptors
SC↑ DBH expression
↑ α2-AR receptor potency
[128]
5-HT: serotonin; AR: adrenoreceptor; DBH: dopamine-β-hydroxylase; FC: frontal cortex; LC: locus coeruleus; NA: noradrenaline; RVM: rostroventromedial medulla; SC: spinal cord; TH: tyrosine hydroxylase; ↑ increase; ↓ decrease; ↔ unchanged.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Cunha, M.; Tavares, I.; Costa-Pereira, J.T. Centralizing the Knowledge and Interpretation of Pain in Chemotherapy-Induced Peripheral Neuropathy: A Paradigm Shift towards Brain-Centric Approaches. Brain Sci. 2024, 14, 659. https://doi.org/10.3390/brainsci14070659

AMA Style

Cunha M, Tavares I, Costa-Pereira JT. Centralizing the Knowledge and Interpretation of Pain in Chemotherapy-Induced Peripheral Neuropathy: A Paradigm Shift towards Brain-Centric Approaches. Brain Sciences. 2024; 14(7):659. https://doi.org/10.3390/brainsci14070659

Chicago/Turabian Style

Cunha, Mário, Isaura Tavares, and José Tiago Costa-Pereira. 2024. "Centralizing the Knowledge and Interpretation of Pain in Chemotherapy-Induced Peripheral Neuropathy: A Paradigm Shift towards Brain-Centric Approaches" Brain Sciences 14, no. 7: 659. https://doi.org/10.3390/brainsci14070659

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop