Abstract
Paclitaxel (Brand name Taxol) is widely used in the treatment of common cancers like breast, ovarian and lung cancer. Although highly effective in blocking tumor progression, paclitaxel also causes peripheral neuropathy as a side effect in 60–70% of chemotherapy patients. Recent efforts by numerous labs have aimed at defining the underlying mechanisms of paclitaxel-induced peripheral neuropathy (PIPN). In vitro models using rodent dorsal root ganglion neurons, human induced pluripotent stem cells, and rodent in vivo models have revealed a number of molecular pathways affected by paclitaxel within axons of sensory neurons and within other cell types, such as the immune system and peripheral glia, as well skin. These studies revealed that paclitaxel induces altered calcium signaling, neuropeptide and growth factor release, mitochondrial damage and reactive oxygen species formation, and can activate ion channels that mediate responses to extracellular cues. Recent studies also suggest a role for the matrix-metalloproteinase 13 (MMP-13) in mediating neuropathy. These diverse changes may be secondary to paclitaxel-induced microtubule transport impairment. Human genetic studies, although still limited, also highlight the involvement of cytoskeletal changes in PIPN. Newly identified molecular targets resulting from these studies could provide the basis for the development of therapies with which to either prevent or reverse paclitaxel-induced peripheral neuropathy in chemotherapy patients.
Keywords: Paclitaxel-induced peripheral neuropathy, small fiber neuropathy, intraepidermal nerve endings, inflammation, reactive oxygen species, human genetic studies, neuropathic pain
1. Introduction
The taxanes are microtubule stabilizing agents that provide great benefits in treating a large number of cancers, including breast, ovarian and lung cancers. Therefore, a large cohort of cancer patients is treated with paclitaxel or other taxanes each year. Unfortunately, neuropathy is a major and dose-limiting side effect of treatment with taxanes. It has been estimated that over half the patients treated with the most commonly used taxane, paclitaxel, experience symptoms; and a subset develop long-lasting, irreversible peripheral neuropathy due to the treatment (Nyrop et al., 2019). The symptoms of neuropathy include pain, tingling, cold-sensitivity and numbness that typically presents in a stocking glove distribution. Many patients experience allodynia, so that normal stimuli such as light touch are experienced as severe pain. As these symptoms dramatically interfere with quality of life (Miaskowski et al., 2018), multiple recent studies have focused on understanding the mechanisms for paclitaxel-induced peripheral neuropathy and how this condition can be prevented or treated (Flatters et al., 2017; Fukuda et al., 2017).
Diverse approaches have been taken to investigate paclitaxel-induced peripheral neuropathy, including in vitro studies investigating the direct effects of paclitaxel on rodent peripheral sensory neurons of the dorsal root ganglia (DRG) and DRGs derived from human induced pluripotent stem cells, in vivo studies using rodent models, and genetic approaches in humans (Fukuda et al., 2017; Gornstein and Schwarz, 2014; Smith et al., 2016). These have highlighted several features of the disorder. The initial effects of paclitaxel appear to reflect on-target changes in the axonal microtubules of sensory neurons. Subsequent changes have been found in intracellular Ca++ regulation, axonal transport, mitochondrial function, and neuropeptide secretion (Bobylev et al., 2015; Chiba et al., 2016) (Li et al., 2019; Li et al., 2018; Pease-Raissi et al., 2017). Interactions within the peripheral nervous system among these neurons and surrounding Schwann cells, satellite glial cells, immune cells and specialized intradermal mechanoreceptor cells, such as Merkel cells, epidermal keratinocytes, as well as the interactions of the central axon of DRG sensory neurons with astrocytes and microglia may all contribute to the progression of the disease (Sekiguchi et al., 2018; Zhang et al., 2008). The complex interplay of all of these processes is also affected by genetic predisposition of the individual patients.
Here we review recent studies using diverse approaches to understand the pathogenesis of paclitaxel-induced peripheral neuropathy (PIPN) (Figure 1). To date, none of these approaches have yielded new clinical methods for preventing or treating this debilitating consequence of chemotherapy. However, integration of these multiple approaches will ultimately yield insights regarding the mechanisms that contribute to this neuropathy, and so will provide new techniques for preventing or treating this devastating, iatrogenic condition.
Fig. 1. Mechanisms of paclitaxel pathogenesis leading to axon degeneration and painful neuropathy based on mammalian studies.

Isolated rodent DRG neurons and patient-derived induced pluripotent stem cells have revealed acute and chronic changes in calcium signaling in vitro. In vivo studies using rodent models have shown various effects directly in sensory neurons and indirectly in skin cells, immune cells and glia. Genetic studies using patient data identified polymorphisms and mutations in various pathways associated with increased risk for paclitaxel-induced peripheral neuropathy (PIPN). Clinical trials for antioxidants have so far failed to prevent or reverse paclitaxel-neurotoxicity.
2. In vitro methods to model neuron-autonomous mechanisms for PIPN
Several in vitro models, using cultures of primary sensory neurons or neuron-like cell lines have been utilized to identify the mechanisms by which paclitaxel alters neuronal function and structure. While these in vitro systems are limited in recapitulating the interactions of neurons within their in vivo microenvironment, they do allow for direct manipulation of protein expression and function via pharmacological and genetic tools to interrogate intracellular signaling pathways altered by paclitaxel within the neurons.
Paclitaxel-induced changes in neuronal sensitivity and excitability have been assessed using assays for neurotransmitter release, changes in intracellular calcium levels, and electrophysiological techniques. In primary cultures derived from rodent dorsal root ganglion neurons, exposure of the cultures to paclitaxel causes time- and concentration-dependent alterations in the basal and stimulated release of two neuropeptides, calcitonin-gene related peptide (CGRP) and substance P (SP). Acute exposure of neuronal cultures to paclitaxel (10nM - 10μM for 10–30 minutes) directly stimulates the release of CGRP and SP in a manner dependent on the activation of protein kinase C (PKC) (He and Wang, 2015; Miyano et al., 2009). A possible mechanism underlying the activation of PKC and neuropeptide release is a transient increase in intracellular calcium, an effect observed after acute treatment of both cultured sensory neurons and neuroblastoma cells with paclitaxel (Boehmerle et al., 2006; Miyano et al., 2009). Putative mechanisms for paclitaxel to acutely enhance intracellular calcium dynamics and neuropeptide release include direct activation of the TLR4 by paclitaxel (Li et al., 2015) and an indirect activation of intracellular calcium release by paclitaxel binding to the neuronal calcium sensor-1 (Boehmerle et al., 2006). In contrast to the acute effects of paclitaxel, chronic exposure of cultures to paclitaxel (300nM - 1μM, for ≥ 3 days) diminishes the release of CGRP stimulated by TRPV1 or TRPA1 agonists (Pittman et al., 2014), which occurs concurrent with a diminution in stimulated calcium release from intracellular stores (Boehmerle et al., 2007). Proposed mechanisms by which extended exposure of neuronal cultures to paclitaxel leads to desensitization of peptide release include disruption of axonal transport and subsequent impairment of the movement of organelles and/or calcium dysregulation (Boehmerle et al., 2007; Shemesh and Spira, 2010; Theiss and Meller, 2000), and the loss of PKC activation (Darby et al., 2017).
Because mechanisms for paclitaxel-induced changes in neuronal sensitivity may differ between rodents and humans, research on human primary neurons and neuron-like cells induced from human pluripotent stem cells has surged in recent years. Some mechanisms for paclitaxel-induced changes identified in rodents have been validated in human neurons, such as a role for paclitaxel to enhance sensitivity of the TRPV1 through activation of TLR4 (Li et al., 2015); however, fundamental differences between human and rodent DRGs also have been identified. Ru Rong Ji’s laboratory has found differential sodium channel expression in human and mouse DRG: human DRG predominantly express Nav1.7, whereas mouse DRG predominantly express Nav1.8. Furthermore, exposure of paclitaxel to cultures derived from human DRG neurons elicits an increase in the expression of Nav1.7, but not that of Nav1.8 (Chang et al., 2018). Another approach to studying the mechanisms of paclitaxel-induced neurotoxicity in cells derived from humans has been to reprogram fibroblasts or embryonic stem cells into sensory neuron-like cells, called induced pluripotent stem cells (IPSCs) (Chambers et al., 2012; Wainger et al., 2015). If these IPSC neurons closely recapitulate the gene expression and cellular functions of sensory neurons, they may provide human-derived cells suitable for in vitro mechanistic studies. A distinct advantage of using patient-derived precursor cells is to recapitulate genotype differences between patient populations in vitro, potentially allowing for a correlation between patient-specific genotypes with peripheral neuron sensitivity and/or response to injury. To date, the studies using IPSC neurons are mostly limited to using cellular morphology as an endpoint of neurotoxicity. However, one study examined the effects of paclitaxel on the mean firing rate of the IPSC neurons and observed a slight decrease in the firing rate of cells following a 48-hour exposure in the absence of any paclitaxel-induced cell death (Wing et al., 2017).
Patients treated with paclitaxel have a decrease in the density of nerve fibers that innervate the epidermis of the hands and feet (Boyette-Davis et al., 2013), and it is likely that this “dying back” or retraction of nerve endings underlies some of the symptoms of PIPN. A similar length-dependent decrease in intraepidermal innervation is apparent in rodents treated with paclitaxel (Boyette-Davis et al., 2011; Jin et al., 2008; Ko et al., 2014; Siau et al., 2006). The effects of paclitaxel on sensory neuron morphology have been examined in DRG explants and dissociated neuronal cultures derived from both embryonic and adult rodents and, dependent upon the timing of drug administration to the cultures, paclitaxel can either inhibit outgrowth or promote degeneration of existing neurites (Gracias et al., 2011; Malgrange et al., 1994; Melli et al., 2006; Pittman et al., 2016; Yang et al., 2009b). Studies from Ahmet Hoke’s laboratory used a compartmentalized microfluidic culture platform to determine whether paclitaxel acts on discrete regions of embryonic neurons to elicit changes in the cellular morphology. Paclitaxel exposure to the axonal compartment caused a reduction in neurite length; however, no changes in neurite length were observed when paclitaxel was applied to the soma (Yang et al., 2009a). These studies suggest that the primary site of paclitaxel uptake or site of action is along the axons and at the peripheral terminals of sensory neurons. Not surprisingly, some of the identified mechanisms by which paclitaxel alters cell morphology are similar to those which underlie functional changes in vitro and in vivo, including activation of the TLR4 (Ustinova et al., 2013), modulation of calcium dynamics (Pease-Raissi et al., 2017), and the production of ROS/RNS (Duggett et al., 2016; Janes et al., 2013; Melli et al., 2008).
In an elegant series of experiments, investigators demonstrated that paclitaxel’s effects on microtubule axonal transport decrease the local translation of a Bcl2 family member, bclw, in cultures of embryonic sensory neurons. Loss of axonal bclw expression dysregulates calcium flux at the endoplasmic reticulum and can lead to activation of calcium-dependent calpains that subsequently induce axonal degeneration (Pease-Raissi et al., 2017). Based upon some early in vivo work, whereby NGF administration alleviated the neurotoxic effects of paclitaxel administration to rodents (Apfel et al., 1991), investigators endeavored to overcome the degenerative effects of chemotherapeutics by increasing growth factors in the in vitro culture media (Chen et al., 2017; Konings et al., 1994; Malgrange et al., 1994; Pittman et al., 2016). Although a protective effect of NGF was observed in embryonic neuronal explants treated with paclitaxel (Konings et al., 1994), these findings were not replicated in dissociated neurons derived from adult rats (Pittman et al., 2016), thus there might be a contribution of development in the protective effects of NGF on neuronal morphology. Other neuroprotective agents that have been found to inhibit or counteract the morphological changes of neurons induced by paclitaxel or docetaxel include insulin-like growth factor-1 (Chen et al., 2017), minoxidil (Chen et al., 2017), erythropoietin (Maggioni et al., 2010; Melli et al., 2006), and pirenzepine (Calcutt et al., 2017). Interestingly, one study combined a functional output to the morphological effects of paclitaxel in vitro. In this work, paclitaxel-induced changes in the release of CGRP from adult sensory neurons was inhibited by the addition of nerve growth factor to the culture media; however, this growth factor addition did not protect against the neurodegenerative effects of paclitaxel in decreasing neurite length in the same cultures (Pittman et al., 2016), suggesting that the mechanisms for neuronal sensitivity and neurite growth could be independent. Further studies are warranted to determine whether these growth factors abrogate the effects of paclitaxel or whether they mask the symptoms because of their growth-promoting effects. To assess the effects of paclitaxel on human cells, IPSC neurons have been exposed to paclitaxel and total neurite outgrowth has been shown to decrease with increasing concentrations of paclitaxel. As in rodent models, studies are ongoing to test the ability of putative neuroprotective drugs to reverse the degenerative effects of paclitaxel on neurite outgrowth (Wing et al., 2017). In summary, neuronal cultures provide a reduced system to study the effects of paclitaxel on neuronal signaling and provide clear endpoints to assess neuroprotection by putative therapies. Using these systems, several neuron-autonomous mechanisms by which paclitaxel alters neuronal function and morphology have been identified, providing novel targets for pharmacological intervention.
3. In vivo pathophysiology of taxanes
Several animal studies explored the mechanisms involved in paclitaxel-induced peripheral neuropathy (PIPN) in vivo. Some non-mammalian models have also been used to understand the mechanisms behind CIPN, such as Zebrafish or Drosophila models, and these are described in detail in another review within this Special Issue. We will therefore focus our review on rodent studies. Paclitaxel is typically administered to mice and rats through intravenous or intraperitoneal injection. The pathophysiological processes induced by paclitaxel include inflammation, oxidative stress, loss of primarily epidermal nerve fibers, and alterations of mitochondrial function and excitability of peripheral neurons. Various cell types in the peripheral and central nervous system have been shown to be affected by paclitaxel treatment in these models, including dorsal root ganglion neurons, Schwann cells, satellite glial cells, microglia, epidermis and spinal astrocytes. Different mechanisms leading to paclitaxel neurotoxicity in rodent models are summarized below:
3.1. Peripheral neurons
It is well established that neuropathic pain induced by paclitaxel treatment is accompanied by damage of small fiber sensory axons. Many publications report the degeneration of fine intra-epidermal nociceptive fibers, as well as axonal demyelination due to the loss of contact between axolemma and Schwann cells, and also a loss of Meissner’s corpuscles, the sensory end-organ for one mechanosensitive neuron subtypes (Beh et al., 2019; Bennett et al., 2011; Boehmerle et al., 2014; Li et al., 2014; Pevida et al., 2013; Wozniak et al., 2018). Nociceptive symptoms observed in animals reflect alterations of electrophysiological parameters as well as the degeneration of nerve endings (Boehmerle et al., 2014; Boyette-Davis et al., 2011). Recently, factors, such as Notch1, phosphorylated Janus kinase 2 (p-JAK2), phosphorylated signal transducer and activator of transcription 3 (p-STAT3) have been implicated in neuropathic pain induced by paclitaxel treatment (Al-Massri et al., 2018, 2019). Paclitaxel alters the electrophysiological properties of peripheral neurons in vivo, and the effect is dependent upon the modified expression of diverse receptors and voltage-gated ion channels within dorsal root ganglion neurons, including increased expression of the calcium channels, Cav3.2, Cav2.3, Cav2.2, a decreased expression of the potassium channels, K2P and Kv7, and the upregulation of sodium channel Nav1.7, all of which results in a gain-of-function (Li et al., 2019; Li et al., 2018; Li et al., 2017; Okubo et al., 2011; Shan et al., 2019; Zhang and Dougherty, 2014). Other receptors involved in paclitaxel-induced neuropathic pain in rats include glutamatergic receptors. Paclitaxel induces a tonic activation of presynaptic NMDA receptors, and increases the expression of the voltage-gated calcium channel subunit, α2δ−1, and the NMDA receptor subunits, GluN1, GluN2A, and GluN2B (Chen et al., 2019).
As paclitaxel directly targets microtubules and prevents depolymerization, it is not surprising that microtubule dysfunction is prevalent in axons following treatment with paclitaxel (Flatters and Bennett, 2006; Xiao et al., 2011; Zheng et al., 2011). It is generally accepted from various in vitro and in vivo studies that paclitaxel impedes fast axonal transport along microtubule tracks (Bobylev et al., 2015; Malacrida et al., 2019; Shemesh and Spira, 2010). While paclitaxel treatment does not directly lead to damage of mitochondrial DNA in neurons (Boyette-Davis et al., 2018), swollen and vacuolated mitochondria in myelinated and unmyelinated sensory nerve fibers have been observed (Flatters and Bennett, 2006), as well as changes in mitochondrial membrane potential and subsequent calcium release (Mironov et al., 2005; Yilmaz and Gold, 2016; Yilmaz et al., 2017). Mitochondrial damage induced by paclitaxel further leads to the formation of reactive oxygen species, resulting in intra-axonal oxidative stress, as indicated by numerous studies (Miao et al., 2018; Singh et al., 2019; Sun et al., 2019; Zhao et al., 2019). For instance, it was found that lipid peroxidation was augmented and glutathione levels increased, whereas expression of the antioxidant enzyme, superoxide dismutase, was decreased in the sciatic nerve and DRG neurons following paclitaxel treatment (Miao et al., 2018; Sun et al., 2019). Treatment of mice with paclitaxel resulted in impaired Nrf2-antioxidant response element (Nrf2-ARE) function and decreased Nrf2-responsive gene (HO-1, γ-GCLC, and NQO1) expression, associated with neuropathic pain (Zhao et al., 2019). Whether the increase in oxidative stress contributes to degeneration of nerve endings has, however, not been directly tested in rodents. However, a recent study demonstrated in zebrafish that axons do not show increased oxidative stress upon paclitaxel treatment despite that axonal mitochondria are vacuolated (Cirrincione et al., 2019). Intriguingly, rather than a role for intrinsic effects of paclitaxel in axon degeneration, epidermally expressed MMP-13 appears to play a role in neuropathy of rats and mice injected with paclitaxel. MMP-13 was previously shown to mediate axon degeneration in the zebrafish model since it is upregulated upon paclitaxel treatment and its pharmacological inhibition prevented axon degeneration (Lisse et al., 2016); yet vacuolation of axonal mitochondria still occurs in inhibitor treated zebrafish. Pharmacological inhibition of MMP-13 either by intraperitoneal injection or upon topical administration to the paw pad of rats also prevented paclitaxel-induced neuropathy (mechanical and cold hypersensitivity), suggesting that MMP-13 dependent mechanisms are conserved. Taken together, evidence points to a multitude of mechanisms leading to paclitaxel-induced axon damage, including inflammation, microtubule aggregation and transport defects, and mitochondrial changes. Whether these phenotypes are underlying axon degeneration remains to be tested in vivo. New molecular players originally identified in zebrafish and validated in mammals, such as MMP-13, could serve as new targets in the treatment of paclitaxel-induced peripheral neuropathy.
3.2. Immune cells and glia
Paclitaxel treatment induces inflammation, with numerous pro-inflammatory chemokines released from either dorsal root ganglion neurons or associated cells. Receptor-mediated signaling by these chemokines can cause neuropathic pain. For example, the chemokine CXCL8 (otherwise known as IL-8) and its receptors CXCR1/2 are upregulated in rat dorsal root ganglion neurons. Importantly inhibition of CXCL8/CXCR1/2 signaling reduced paclitaxel-induced nociception and alleviated mechanical and cold hypersensitivity in a rat model (Brandolini et al., 2017). More recently, it was shown that upregulation of another chemokine, CXCL1, and its receptor CXCR2 and subsequent activation of PI3Kγ contributes to the development and maintenance of paclitaxel-induced mechanical hypersensitivity in mice (Manjavachi et al., 2019). Paclitaxel treatment of mice can cause accumulation of macrophages in the sciatic nerve, and upregulation of the chemokine receptor, CXCR4, and RAGE (Receptor for Advanced Glycation Endproducts) in dorsal root ganglia. Intriguingly, antagonists for CXCR4 and RAGE can prevent paclitaxel-induced allodynia (Sekiguchi et al., 2018). Another chemokine, MCP-1/CCL-2 (Monocyte Chemoattractant Protein 1) and its cognate receptor CCR2 may also play a role in paclitaxel-induced inflammation and macrophage migration, as increased MCP-1 expression was observed in small nociceptive dorsal root ganglion neurons and in spinal astrocytes in rats treated with paclitaxel (Zhang et al., 2013). This chemokine may stimulate CCR2 which is expressed in large and medium-sized myelinated neurons. Direct addition of MCP-1 to dorsal root ganglion neurons led to calcium spikes selectively in CCR2-positive medium-sized neurons, and injection of anti-MCP-1 antibodies and CCR2 antisense oligonucleotides reduced intra-epidermal nerve fiber loss and mechanical hypersensitivity (Zhang et al., 2016). Similarly, previous studies showed that paclitaxel induces an inflammatory response in dorsal root ganglion neurons by activating the Toll-like receptor 4 (TLR4) pathway and increasing MCP-1 expression (Byrd-Leifer et al., 2001) (Li et al., 2014). Taken together, chemokine signaling plays a critical part in the crosstalk among different dorsal root ganglion neuron subpopulations and so contributes to neuropathic pain and allodynia. Cytokine-dependent nuclear translocation of nuclear factor-κB (NFκB), which regulates nerve growth factor, promotes paclitaxel-induced mechanical, thermal and cold hypersensitivity in outbred ICR mice (Meng et al., 2019). In this study, nuclear NF-κB translocation and peripheral nerve injury could be blocked by inhibiting the activation of p38 phosphorylation via duloxetine.
Recently a mechanistic link between complement, a key component of the innate immune system, and paclitaxel-induced peripheral neuropathy has been found (Xu et al., 2018). Using CRISPR/Cas9 in rats, a major component of this system, C3, was knocked out to study its involvement in paclitaxel-induced neuropathy. Rats lacking C3 showed improved sensitivity to touch and reduced loss of intradermal nerve fibers. Furthermore in vitro studies using the neuronal cell line PC12 revealed that paclitaxel treatment enhances C3 activation and this contributes to cytotoxicity.
Increasing evidence indicates that non-neuronal cells, including microglia and astrocytes of the CNS, are also involved in the initiation and resolution of pain. The role of microglia in paclitaxel-induced peripheral neuropathy has therefore been the subject to a few recent studies. It was shown that paclitaxel treatment induces microglia dysregulation in the dorsal horn of the spinal cord and increases the expression of cannabinoid type 2 receptor (CB2), and of the chemokines, CCL2 and interleukin (IL-6, IL-4 and IL-10) (Ha et al., 2019; Pevida et al., 2013; Wu et al., 2019). More recently, there has been a growing interest in cannabinoid receptors in the treatment of neuropathic pain. For example, by modulating microglia these receptors can have analgesic properties and reduce pain associated with paclitaxel-induced neuropathy (Burgos et al., 2012; Wu et al., 2019). Paclitaxel has further been shown to induce neuropathic pain by activation of spinal astrocytes, which causes the secretion of pro-inflammatory cytokines, i.e. tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) and IL-6 (Ba et al., 2018; Burgos et al., 2012).
Other glia cells, such as satellite glial cells that support and surround DRG neuronal cell bodies, have been shown to play an additional role in paclitaxel-induced peripheral neuropathy. For example, paclitaxel treatment leads to increased gap junction coupling among satellite glial cells, which may contribute to pain sensation (Warwick and Hanani, 2013). It has been suggested that activation of TLR4 in satellite glial cells stimulates the release of TNF-α and subsequent inflammation, TRP channel activation, and pain sensation (Boehmerle et al., 2018; Duggett et al., 2016; Materazzi et al., 2012; Wu et al., 2015). Interestingly, TRPA1 and TRPV4 have also been implicated in paclitaxel-induced cold hypersensitivity (Alessandri-Haber et al., 2004) Satellite glial cells were further shown to upregulate angiotensin II type 1 receptor (AT1R) following paclitaxel treatment, which increases the expression of inflammatory cytokines, such as IL-1β and TNF-α in the nearby dorsal root ganglion neurons (Kim et al., 2019).
3.3. Pathogenesis of other taxanes leading to peripheral neuropathy
Along with paclitaxel, also docetaxel and cabazitaxel belong to the family of taxanes that are used in oncology and which can cause peripheral neuropathy, although not as severe as the effects induced by paclitaxel treatment. However, few studies explored their mechanisms in this condition. Docetaxel binds to tubulin with greater affinity than paclitaxel and causes cell cycle arrest in the S rather than G2-M phase junction as is the case for paclitaxel (Dorr, 1997). Nonetheless, paclitaxel is more commonly associated with the development of CIPN in cancer patients (Chon et al., 2009; Hilkens and ven den Bent, 1997). In animal studies, docetaxel is typically administered to mice and rats through intravenous or intraperitoneal injection. However, few studies have described docetaxel-induced peripheral neuropathies. A decrease in nerve conduction velocity was observed for intravenous injection of docetaxel at doses between 5 and 12.5 mg/kg in rats (Persohn et al., 2005). Nevertheless, no significant changes in sciatic nerve morphology (number of fibers and demyelinization) have been reported (Persohn et al., 2005). Another study showed a decrease in nerve conduction velocity, intraepidermal fiber numbers and myelin protein expression in a docetaxel rat model (10mg/kg, i.v., 4 weeks) (Roglio et al., 2009). More recently, there have been reports of the involvement of TRPA1 receptors (through upward regulation) in the development of mechanical allodynia induced by docetaxel (Huang et al., 2017). Cabazitaxel is a dimethyl derivative of docetaxel with equivalent efficacy to docetaxel for stabilizing microtubules in vitro. Clinical studies suggest that the incidence and severity of peripheral neuropathy may be lower with cabazitaxel compared with other taxanes (de Bono et al., 2010). However, to our knowledge, no animal studies explored the mechanisms involved in peripheral neuropathies and neuropathic pain induced by cabazitaxel.
4. Human genetic studies provide insight into clinical paclitaxel neurotoxicity
Human genetic studies have provided pathomechanistic insight into taxane-induced peripheral neuropathy. The genetic study design takes advantage of an important clinical fact: for a given taxane exposure, neuropathy can have dramatically variable severity or may not occur at all. By comparing genotypes in patients with varying CIPN phenotypes, these studies have uncovered specific genes and pathways that may underlie CIPN susceptibility. There are two broad approaches that are employed in these genetic studies: targeted or unbiased. In the targeted approach, specific genes that are hypothesized to contribute to CIPN susceptibility are investigated (i.e. neuropathy or metabolism genes), whereas in the unbiased studies, genome wide associations are investigated that may uncover unexpected pathways.
A review of available human genetic studies of taxane-induced neuropathy susceptibility on PubMed revealed 30 studies dating back to 2006, which includes both targeted (Abraham et al., 2014; Apellaniz-Ruiz et al., 2015; Apellaniz-Ruiz et al., 2017; Beutler et al., 2014; Boora et al., 2016; Boora et al., 2015; de Graan et al., 2013; Dou et al., 2017; Eckhoff et al., 2015; Hertz et al., 2014; Hertz et al., 2013; Kulkarni et al., 2015; Leandro-Garcia et al., 2012; Lee et al., 2015a; Leskela et al., 2011; McWhinney-Glass et al., 2013; Mir et al., 2009; Park et al., 2017; Park et al., 2014; Sissung et al., 2006; Sucheston et al., 2011; Tanabe et al., 2017) and unbiased (Baldwin et al., 2012; Bergmann et al., 2013; Hertz et al., 2016; Leandro-Garcia et al., 2013; Schneider et al., 2016; Schneider et al., 2015; Sucheston-Campbell et al., 2018; Wheeler et al., 2013) approaches. Polymorphisms and mutations in genes that have been associated with altered risk of taxane neuropathy include genes in structural and cytoskeletal pathways, such as LIMK2 (Dou et al., 2017; Leandro-Garcia et al., 2013), MAPT (Park et al., 2017), SBF2 (Schneider et al., 2016), PRX (Beutler et al., 2014), TUBB2A (Abraham et al., 2014; Leandro-Garcia et al., 2012), TECTA (Bergmann et al., 2013), FGD4 (Baldwin et al., 2012), in neural developmental pathways, including GSK3B (Park et al., 2017; Park et al., 2014), EPHA4–6 (Apellaniz-Ruiz et al., 2017; Boora et al., 2016; Leandro-Garcia et al., 2013), BDNF (Azoulay et al., 2015), ARGHEF10 (Beutler et al., 2014; Boora et al., 2015), GPR177 (Schneider et al., 2015), SOX10 (McWhinney-Glass et al., 2013), in cell damage and apoptosis, including BCL2 (McWhinney-Glass et al., 2013), RWDD3 (Bergmann et al., 2013), FANCD2 (Sucheston et al., 2011), and in drug metabolism, including ABCB1 (Abraham et al., 2014; Boora et al., 2016; Sissung et al., 2006; Tanabe et al., 2017), CYP genes (Apellaniz-Ruiz et al., 2015; de Graan et al., 2013; Hertz et al., 2014; Hertz et al., 2013; Lee et al., 2015b; Leskela et al., 2011), GSTP1 (Eckhoff et al., 2015; Mir et al., 2009), in addition to several genes that when mutated can cause Charcot-Marie Tooth Disease and other neurodegenerative diseases (Baldwin et al., 2012; Beutler et al., 2014; Boora et al., 2015; Hertz et al., 2016; Schneider et al., 2016). While these studies have provided mechanistic insights to genes and pathways that might influence taxane-induced peripheral neuropathy, many of these genetic association studies have been fraught with methodological problems and challenges with reproducibility. Varied methods of genetic analysis, underpowered studies, lack of validated cohorts, neuropathy phenotyping, and concomitant neurotoxic chemotherapies (often platinum-based compounds) have unfortunately prevented wide applicability of these studies (Argyriou et al., 2017; Cavaletti et al., 2011). A recent review suggested that ABCB1, TUBB2A, GPR177, and EPHA4–6 were the genes that had the strongest data supporting their role in taxane-induced neuropathy susceptibility and should be validated further (Argyriou et al., 2017).
Attempts at prevention of taxane-induced neuropathy or its treatment have been disappointing in human clinical trials. The only clear success in a large randomized clinical trial was the demonstration that duloxetine provides a modest improvement of pain in patients with established CIPN (Smith et al., 2013). All other studies were either negative or showed benefits in small cohorts that have not been reproduced (Majithia et al., 2016). Failed prevention trials have focused on diffuse neuroprotection and have employed agents that are cytoprotective, antioxidants and improve mitochondria function (Amifostine, glutathione, vitamin E, acetyl-L-carnitine). It is hoped that more targeted prevention strategies that focus on unique aspects of CIPN (drug transporters, axonal degeneration, neuroinflammation) will be able to prevent CIPN while still maintaining taxanes’ tumor-killing properties.
In all the human CIPN studies described above, it is critical that researchers are able to accurately determine the presence of a given patient’s CIPN and its severity. In the complicated clinical setting of a patient with cancer, this is less straightforward than it may initially seem. There has been intensive research into the methodology of quantitating CIPN in patients, which broadly can be divided into clinician-reported outcomes (ClinROs) and patient reported outcomes (PROs). ClinROs may include very blunt instruments such as the Common Terminology Criteria for Adverse Events (CTCAE) or more sensitive assessments such as, quantitative neurological examinations, neurophysiology, or nerve pathology (Argyriou et al., 2019). PROs in CIPN have become more widely used and are typically questionnaires related to symptoms (sensory, motor and autonomic) (Griffith et al., 2010) or functionality (e.g. activity of daily living) (Binda et al., 2015). Many of the above cited studies used poorly sensitive or specific CIPN tools such as the CTCAE, which raises concerns about the reliability of their findings. Fortunately, the more sensitive CIPN measurements, often a combination of ClinROs and PROs, are now routinely being incorporated into human studies of CIPN (Gewandter et al., 2018).
5. Conclusions
Of the patients given paclitaxel for cancer treatment, about 60–70% suffer from paclitaxel-induced peripheral neuropathy (Seretny et al., 2014). Paclitaxel-induced peripheral neuropathy, among other chemotherapy-induced peripheral neuropathies, is a major reason for early termination of chemotherapy. Although many groups have begun to uncover mechanisms by which paclitaxel causes neuropathy, there are currently no effective clinical treatments available for this condition. Several questions are being addressed, including (i) What are the direct effects of paclitaxel on sensory axons? (ii) What type of damage causes specific symptoms? and (iii) Are the neurons or other cell types connect to neurons the primary targets of paclitaxel leading to neuropathy? In addition, it will be important to address whether alternative mechanisms, besides microtubule damage, which is the primary effect in cancer treatment, could cause axon degeneration. Addressing these questions is likely to enable the development of new treatments.
Microtubule stabilization has been analyzed as a potential cause of axonal degeneration, but most of the knowledge in this respect has been derived from in vitro analyses. An important observation in vitro and in vivo is that mitochondrial damage associated with oxidative stress also contributes to nerve damage, and given findings in the cancer field, which show that mitochondria can be directly targeted by paclitaxel and damaged (André et al., 2002; Kidd et al., 2002; Varbiro et al., 2001), it is possible that mitochondrial damage is a primary cause of axon degeneration through the formation of reactive oxygen species. Intriguing and consistent with this idea are findings that in some human trials, antioxidants such as Amifostine, acetyl-L-carnitine, β-carotene and α-tocopherol are beneficial to patients. Nevertheless, clinical trials with these agents have also revealed a great variability among between individual patients, which has prevented their widespread clinical use despite FDA approval (Block and Gyllenhaal, 2005; Maestri et al., 2005). The difficulty in using antioxidant therapies in addition to cancer treatment is that interference with reactive oxygen species may exert widespread toxicities. For instance, cells depend on low-level reactive oxygen species whereas their overproduction in diseases like cancer, or due to chemotherapy treatment, causes cellular damage. Thus, finding the proper balance of antioxidants during treatment will be important but is inherently difficult. Intravenous injections of the antioxidant and anti-inflammatory agent, Amifostine, for instance, led to a wide variety of systemic toxicities like hypotension, nausea, vomiting, and hypocalcemia (Block and Gyllenhaal, 2005). On the other hand, many, but not all, of these toxicities were eliminated upon subcutaneous administration. This further highlights the complexity of oxidation and proper delivery of these agents into inherently different tissues and/or cell types. Targeted approaches, such as those that manipulate the function of downstream or upstream factors of reactive oxygen species will likely be much more controllable since these approaches target one or few specific pathways. Efforts in the private sector have been under way to target, for instance, mitochondria damage induced by paclitaxel treatment to reduce the formation of reactive oxygen species.
Given a relatively broad understanding of potential mechanisms implicated in paclitaxel-induced peripheral neuropathy, the mechanisms identified thus far need to be carefully dissected in pre-clinical models in order to determine whether any of those might be useful for the development of treatments. Future efforts need to be directed at validating this mechanistic data in humans. One approach could be, for example, to compare data across species and with data in humans, such as patient databases. Conservation of mechanisms across multiple species including non-mammalian and mammalian vertebrates or invertebrates has a higher chance to also be conserved in humans. Prior to clinical trials, some of the findings could be further validated using patient-derived induced pluripotent stem cells, for example. Generating new data sets from patient samples derived either from tissue storage resources or clinical trials could also be useful. Various efforts are currently under way, such as within the Toxic Neuropathy Consortium, to collect patient data across the globe for comparative purposes, which will be available to clinicians and scientists.
Additional efforts should be made to develop two classes of treatments, one class comprising preventative therapies, and those that can reverse the symptoms in those patients already suffering from the condition. Ideally one therapy shows efficacy toward both. These therapies could either target pathways inducing axon de- and regeneration or mitigate or prevent pain symptoms without opioid-based drugs. To date, most efforts concentrate on preventing painful neuropathies, which could be related to the increased efforts related to pain research. Some studies using non-mammalian in vivo models have in contrast been quite successful in their identification of new candidate targeting axon de- and regeneration pathways, which are also discussed in this Special issue.
Highlights.
Inflammation, ion channel function and perturbed calcium homeostasis contribute to paclitaxel-induced neuropathic pain
Reactive oxygen species formation and altered microtubule function are common in animal models of paclitaxel-induced peripheral neuropathy
Genetic studies in patients with paclitaxel-induced peripheral neuropathy have revealed genes that are also altered in animal models and could represent new targets in the treatment of this condition
Acknowledgments
We thank the Peripheral Nerve Society and Toxic Neuropathy Consortium for their support.
Funding sources
N.P.S. received financial support from the National Cancer Institute/NIH [R01CA 211887 (PI)], [R01 CA 215973 sub-award], Minnesota Regenerative Medicine Partnership: RMM 11215 CT002, Target ALS, ALS Association. J.C.F. received financial support from the National Cancer Institute/NIH [CA205166 and CA231267]. M.I.D. was supported by the National Institutes of Health (NIH) [grants R01CA206028-01, R01CA221260 and R01 CA219637]. R.A.S. received funding for this work from the National Institutes of Health (NIH) [grant RO1CA205255]. S.R. was supported by the National Cancer Institute (NIH) [grant 5RO1CA215973] and a CTSI-Pilot Award (University of Miami).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflict of interest
The authors declare no conflicts of interest.
References
- Abraham JE, Guo Q, Dorling L, Tyrer J, Ingle S, Hardy R, Vallier AL, Hiller L, Burns R, Jones L, Bowden SJ, Dunn JA, Poole CJ, Caldas C, Pharoah PP, Earl HM, 2014. Replication of genetic polymorphisms reported to be associated with taxane-related sensory neuropathy in patients with early breast cancer treated with Paclitaxel. Clinical cancer research: an official journal of the American Association for Cancer Research 20, 2466–2475. [DOI] [PubMed] [Google Scholar]
- Al-Massri KF, Ahmed LA, El-Abhar HS, 2018. Pregabalin and lacosamide ameliorate paclitaxel-induced peripheral neuropathy via inhibition of JAK/STAT signaling pathway and Notch-1 receptor. Neurochemistry International 120, 164–171. [DOI] [PubMed] [Google Scholar]
- Al-Massri KF, Ahmed LA, El-Abhar HS, 2019. Mesenchymal stem cells therapy enhances the efficacy of pregabalin and prevents its motor impairment in paclitaxel-induced neuropathy in rats: Role of Notch1 receptor and JAK/STAT signaling pathway. Behavioural Brain Research 360, 303–311. [DOI] [PubMed] [Google Scholar]
- Alessandri-Haber N, Dina OA, Yeh JJ, Parada CA, Reichling DB, Levine JD, 2004. Transient receptor potential vanilloid 4 is essential in chemotherapy-induced neuropathic pain in the rat. J Neurosci 24, 4444–4452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- André N, Carré M, Brasseur G, Pourroy B, Kovacic H, Briand C, Braguer D, 2002. Paclitaxel targets mitochondria upstream of caspase activation in intact human neuroblastoma cells. FEBS Lett 532, 256–260. [DOI] [PubMed] [Google Scholar]
- Apellaniz-Ruiz M, Lee MY, Sanchez-Barroso L, Gutierrez-Gutierrez G, Calvo I, Garcia-Estevez L, Sereno M, Garcia-Donas J, Castelo B, Guerra E, Leandro-Garcia LJ, Cascon A, Johansson I, Robledo M, Ingelman-Sundberg M, Rodriguez-Antona C, 2015. Whole-exome sequencing reveals defective CYP3A4 variants predictive of paclitaxel dose-limiting neuropathy. Clinical cancer research: an official journal of the American Association for Cancer Research 21, 322–328. [DOI] [PubMed] [Google Scholar]
- Apellaniz-Ruiz M, Tejero H, Inglada-Perez L, Sanchez-Barroso L, Gutierrez-Gutierrez G, Calvo I, Castelo B, Redondo A, Garcia-Donas J, Romero-Laorden N, Sereno M, Merino M, Curras-Freixes M, Montero-Conde C, Mancikova V, Avall-Lundqvist E, Green H, Al-Shahrour F, Cascon A, Robledo M, Rodriguez-Antona C, 2017. Targeted Sequencing Reveals Low-Frequency Variants in EPHA Genes as Markers of Paclitaxel-Induced Peripheral Neuropathy. Clinical cancer research: an official journal of the American Association for Cancer Research 23, 1227–1235. [DOI] [PubMed] [Google Scholar]
- Apfel SC, Lipton RB, Arezzo JC, Kessler JA, 1991. Nerve growth factor prevents toxic neuropathy in mice. Ann Neurol 29, 87–90. [DOI] [PubMed] [Google Scholar]
- Argyriou AA, Bruna J, Genazzani AA, Cavaletti G, 2017. Chemotherapy-induced peripheral neurotoxicity: management informed by pharmacogenetics. Nat Rev Neurol 13, 492–504. [DOI] [PubMed] [Google Scholar]
- Argyriou AA, Park SB, Islam B, Tamburin S, Velasco R, Alberti P, Bruna J, Psimaras D, Cavaletti G, Cornblath DR, (TNC), T.N.C., 2019. Neurophysiological, nerve imaging and other techniques to assess chemotherapy-induced peripheral neurotoxicity in the clinical and research settings. J Neurol Neurosurg Psychiatry. [DOI] [PubMed] [Google Scholar]
- Azoulay D, Leibovici A, Sharoni R, Shaoul E, Gross B, Braester A, Goldberg H, 2015. Association between Met-BDNF allele and vulnerability to paclitaxel-induced peripheral neuropathy. Breast Cancer Res Treat 153, 703–704. [DOI] [PubMed] [Google Scholar]
- Ba X, Wang J, Zhou S, Luo X, Peng Y, Yang S, Hao Y, Jin G, 2018. Cinobufacini protects against paclitaxel-induced peripheral neuropathic pain and suppresses TRPV1 up-regulation and spinal astrocyte activation in rats. Biomedicine & Pharmacotherapy = Biomedecine & Pharmacotherapie 108, 76–84. [DOI] [PubMed] [Google Scholar]
- Baldwin RM, Owzar K, Zembutsu H, Chhibber A, Kubo M, Jiang C, Watson D, Eclov RJ, Mefford J, McLeod HL, Friedman PN, Hudis CA, Winer EP, Jorgenson EM, Witte JS, Shulman LN, Nakamura Y, Ratain MJ, Kroetz DL, 2012. A genome-wide association study identifies novel loci for paclitaxel-induced sensory peripheral neuropathy in CALGB 40101. Clinical cancer research: an official journal of the American Association for Cancer Research 18, 5099–5109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beh ST, Kuo Y-M, Chang W-SW, Wilder-Smith E, Tsao C-H, Tsai C-H, Chen L-T, Liao L-D, 2019. Preventive hypothermia as a neuroprotective strategy for paclitaxel-induced peripheral neuropathy. Pain. [DOI] [PubMed] [Google Scholar]
- Bennett GJ, Liu GK, Xiao WH, Jin HW, Siau C, 2011. Terminal arbor degeneration--a novel lesion produced by the antineoplastic agent paclitaxel. Eur J Neurosci 33, 1667–1676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bergmann TK, Vach W, Feddersen S, Eckhoff L, Green H, Herrstedt J, Brosen K, 2013. GWAS-based association between RWDD3 and TECTA variants and paclitaxel induced neuropathy could not be confirmed in Scandinavian ovarian cancer patients. Acta Oncol 52, 871–874. [DOI] [PubMed] [Google Scholar]
- Beutler AS, Kulkarni AA, Kanwar R, Klein CJ, Therneau TM, Qin R, Banck MS, Boora GK, Ruddy KJ, Wu Y, Smalley RL, Cunningham JM, Le-Lindqwister NA, Beyerlein P, Schroth GP, Windebank AJ, Zuchner S, Loprinzi CL, 2014. Sequencing of Charcot-Marie-Tooth disease genes in a toxic polyneuropathy. Annals of neurology 76, 727–737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Binda D, Cavaletti G, Cornblath DR, Merkies IS, group C.-P.s., 2015. Rasch-Transformed Total Neuropathy Score clinical version (RT-TNSc(©)) in patients with chemotherapy-induced peripheral neuropathy. J Peripher Nerv Syst 20, 328–332. [DOI] [PubMed] [Google Scholar]
- Block KI, Gyllenhaal C, 2005. Commentary: the pharmacological antioxidant amifostine -- implications of recent research for integrative cancer care. Integr Cancer Ther 4, 329–351. [DOI] [PubMed] [Google Scholar]
- Bobylev I, Joshi AR, Barham M, Ritter C, Neiss WF, Höke A, Lehmann HC, 2015. Paclitaxel inhibits mRNA transport in axons. Neurobiol Dis 82, 321–331. [DOI] [PubMed] [Google Scholar]
- Boehmerle W, Huehnchen P, Lee SLL, Harms C, Endres M, 2018. TRPV4 inhibition prevents paclitaxel-induced neurotoxicity in preclinical models. Experimental Neurology 306, 64–75. [DOI] [PubMed] [Google Scholar]
- Boehmerle W, Huehnchen P, Peruzzaro S, Balkaya M, Endres M, 2014. Electrophysiological, behavioral and histological characterization of paclitaxel, cisplatin, vincristine and bortezomib-induced neuropathy in C57Bl/6 mice. Sci Rep 4, 6370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boehmerle W, Splittgerber U, Lazarus MB, McKenzie KM, Johnston DG, Austin DJ, Ehrlich BE, 2006. Paclitaxel induces calcium oscillations via an inositol 1,4,5-trisphosphate receptor and neuronal calcium sensor 1-dependent mechanism. Proceedings of the National Academy of Sciences of the United States of America 103, 18356–18361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boehmerle W, Zhang K, Sivula M, Heidrich FM, Lee Y, Jordt SE, Ehrlich BE, 2007. Chronic exposure to paclitaxel diminishes phosphoinositide signaling by calpain-mediated neuronal calcium sensor-1 degradation. Proceedings of the National Academy of Sciences of the United States of America 104, 11103–11108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boora GK, Kanwar R, Kulkarni AA, Abyzov A, Sloan J, Ruddy KJ, Banck MS, Loprinzi CL, Beutler AS, 2016. Testing of candidate single nucleotide variants associated with paclitaxel neuropathy in the trial NCCTG N08C1 (Alliance). Cancer Med 5, 631–639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boora GK, Kulkarni AA, Kanwar R, Beyerlein P, Qin R, Banck MS, Ruddy KJ, Pleticha J, Lynch CA, Behrens RJ, Zuchner S, Loprinzi CL, Beutler AS, 2015. Association of the Charcot-Marie-Tooth disease gene ARHGEF10 with paclitaxel induced peripheral neuropathy in NCCTG N08CA (Alliance). Journal of the neurological sciences 357, 35–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boyette-Davis J, Xin W, Zhang H, Dougherty PM, 2011. Intraepidermal nerve fiber loss corresponds to the development of taxol-induced hyperalgesia and can be prevented by treatment with minocycline. Pain 152, 308–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boyette-Davis JA, Cata JP, Driver LC, Novy DM, Bruel BM, Mooring DL, Wendelschafer-Crabb G, Kennedy WR, Dougherty PM, 2013. Persistent chemoneuropathy in patients receiving the plant alkaloids paclitaxel and vincristine. Cancer chemotherapy and pharmacology 71, 619–626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boyette-Davis JA, Hou S, Abdi S, Dougherty PM, 2018. An updated understanding of the mechanisms involved in chemotherapy-induced neuropathy. Pain Manag 8, 363–375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brandolini L, Benedetti E, Ruffini PA, Russo R, Cristiano L, Antonosante A, d’Angelo M, Castelli V, Giordano A, Allegretti M, Cimini A, 2017. CXCR1/2 pathways in paclitaxel-induced neuropathic pain. Oncotarget 8, 23188–23201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burgos E, Gómez-Nicola D, Pascual D, Martín MI, Nieto-Sampedro M, Goicoechea C, 2012. Cannabinoid agonist WIN 55,212–2 prevents the development of paclitaxel-induced peripheral neuropathy in rats. Possible involvement of spinal glial cells. European Journal of Pharmacology 682, 62–72. [DOI] [PubMed] [Google Scholar]
- Byrd-Leifer CA, Block EF, Takeda K, Akira S, Ding A, 2001. The role of MyD88 and TLR4 in the LPS-mimetic activity of Taxol. European Journal of Immunology 31, 2448–2457. [DOI] [PubMed] [Google Scholar]
- Calcutt NA, Smith DR, Frizzi K, Sabbir MG, Chowdhury SK, Mixcoatl-Zecuatl T, Saleh A, Muttalib N, Van der Ploeg R, Ochoa J, Gopaul A, Tessler L, Wess J, Jolivalt CG, Fernyhough P, 2017. Selective antagonism of muscarinic receptors is neuroprotective in peripheral neuropathy. J Clin Invest 127, 608–622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cavaletti G, Alberti P, Marmiroli P, 2011. Chemotherapy-induced peripheral neurotoxicity in the era of pharmacogenomics. Lancet Oncol 12, 1151–1161. [DOI] [PubMed] [Google Scholar]
- Chambers SM, Qi Y, Mica Y, Lee G, Zhang X-J, Niu L, Bilsland J, Cao L, Stevens E, Whiting P, Shi S-H, Studer L, 2012. Combined small-molecule inhibition accelerates developmental timing and converts human pluripotent stem cells into nociceptors. Nature Biotechnology 30, 715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang W, Berta T, Kim YH, Lee S, Lee SY, Ji RR, 2018. Expression and Role of Voltage-Gated Sodium Channels in Human Dorsal Root Ganglion Neurons with Special Focus on Nav1.7, Species Differences, and Regulation by Paclitaxel. Neurosci Bull 34, 4–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen C, Bai X, Bi Y, Liu G, Li H, Liu Z, Liu H, 2017. Insulin-like growth factor-1 attenuates apoptosis and protects neurochemical phenotypes of dorsal root ganglion neurons with paclitaxel-induced neurotoxicity in vitro. Nutr Neurosci 20, 89–102. [DOI] [PubMed] [Google Scholar]
- Chen Y, Chen S-R, Chen H, Zhang J, Pan H-L, 2019. Increased α2δ−1-NMDA receptor coupling potentiates glutamatergic input to spinal dorsal horn neurons in chemotherapy-induced neuropathic pain. Journal of Neurochemistry 148, 252–274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chiba T, Oka Y, Kambe T, Koizumi N, Abe K, Kawakami K, Utsunomiya I, Taguchi K, 2016. Paclitaxel-induced peripheral neuropathy increases substance P release in rat spinal cord. Eur J Pharmacol 770, 46–51. [DOI] [PubMed] [Google Scholar]
- Chon HJ, Rha SY, Im CK, Kim C, Hong MH, Kim HR, An JR, Noh SH, Chung HC, Jeung HC, 2009. Docetaxel versus paclitaxel combined with 5-FU and leucovorin in advanced gastric cancer: combined analysis of two phase II trials. Cancer Res Treat 41, 196–204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cirrincione AM, Pellegrini AD, Dominy JR, Benjamin ME, Utkina-Sosunova I, Lotti F, Jergova S, Sagen J, Rieger S, 2019. Paclitaxel-induced peripheral neuropathy is caused by epidermal ROS and mitochondrial damage through conserved MMP-13 activation. bioRxiv, 743419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Darby LM, Meng H, Fehrenbacher JC, 2017. Paclitaxel inhibits the activity and membrane localization of PKCalpha and PKCbetaI/II to elicit a decrease in stimulated calcitonin gene-related peptide release from cultured sensory neurons. Mol Cell Neurosci 82, 105–117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Graan AJ, Elens L, Sprowl JA, Sparreboom A, Friberg LE, van der Holt B, de Raaf PJ, de Bruijn P, Engels FK, Eskens FA, Wiemer EA, Verweij J, Mathijssen RH, van Schaik RH, 2013. CYP3A4*22 genotype and systemic exposure affect paclitaxel-induced neurotoxicity. Clinical cancer research: an official journal of the American Association for Cancer Research 19, 3316–3324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dorr RT, 1997. Pharmacology of the taxanes. Pharmacotherapy 17, 96S–104S. [PubMed] [Google Scholar]
- Dou XL, Mai YL, Sun Z, Wang YY, Shao YJ, Cheng YJ, Zhou N, Luo F, Zhang B, Bai CM, Ma SQ, 2017. Single Nucleotide Polymorphisms of Paclitaxel-induced Peripheral Sensory Neuropathy in Chinese Han Population. Zhongguo yi xue ke xue yuan xue bao. Acta Academiae Medicinae Sinicae 39, 593–601. [DOI] [PubMed] [Google Scholar]
- Duggett NA, Griffiths LA, McKenna OE, de Santis V, Yongsanguanchai N, Mokori EB, Flatters SJ, 2016. Oxidative stress in the development, maintenanceand resolution of paclitaxel-induced painful neuropathy. Neuroscience 333, 13–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eckhoff L, Feddersen S, Knoop AS, Ewertz M, Bergmann TK, 2015. Docetaxel-induced neuropathy: a pharmacogenetic case-control study of 150 women with early-stage breast cancer. Acta Oncol 54, 530–537. [DOI] [PubMed] [Google Scholar]
- Flatters SJ, Bennett GJ, 2006. Studies of peripheral sensory nerves in paclitaxel-induced painful peripheral neuropathy: evidence for mitochondrial dysfunction. Pain 122, 245–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Flatters SJL, Dougherty PM, Colvin LA, 2017. Clinical and preclinical perspectives on Chemotherapy-Induced Peripheral Neuropathy (CIPN): a narrative review. Br J Anaesth 119, 737–749. [DOI] [PubMed] [Google Scholar]
- Fukuda Y, Li Y, Segal RA, 2017. A Mechanistic Understanding of Axon Degeneration in Chemotherapy-Induced Peripheral Neuropathy. Front Neurosci 11, 481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gewandter JS, Brell J, Cavaletti G, Dougherty PM, Evans S, Howie L, McDermott MP, O’Mara A, Smith AG, Dastros-Pitei D, Gauthier LR, Haroutounian S, Jarpe M, Katz NP, Loprinzi C, Richardson P, Lavoie-Smith EM, Wen PY, Turk DC, Dworkin RH, Freeman R, 2018. Trial designs for chemotherapy-induced peripheral neuropathy prevention: ACTTION recommendations. Neurology 91, 403–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gornstein E, Schwarz TL, 2014. The paradox of paclitaxel neurotoxicity: Mechanisms and unanswered questions. Neuropharmacology 76 Pt A, 175–183. [DOI] [PubMed] [Google Scholar]
- Gracias NG, Cummins TR, Kelley MR, Basile DP, Iqbal T, Vasko MR, 2011. Vasodilatation in the rat dorsal hindpaw induced by activation of sensory neurons is reduced by paclitaxel. Neurotoxicology 32, 140–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Griffith KA, Merkies IS, Hill EE, Cornblath DR, 2010. Measures of chemotherapy-induced peripheral neuropathy: a systematic review of psychometric properties. J Peripher Nerv Syst 15, 314–325. [DOI] [PubMed] [Google Scholar]
- Ha J-W, You M-J, Park H-S, Kim JW, Kwon M-S, 2019. Differential effect of LPS and paclitaxel on microglial functional phenotypes and circulating cytokines: the possible role of CX3CR1 and IL-4/10 in blocking persistent inflammation. Archives of Pharmacal Research 42, 359–368. [DOI] [PubMed] [Google Scholar]
- He Y, Wang ZJ, 2015. Nociceptor Beta II, Delta, and Epsilon Isoforms of PKC Differentially Mediate Paclitaxel-Induced Spontaneous and Evoked Pain. J Neurosci 35, 4614–4625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hertz DL, Owzar K, Lessans S, Wing C, Jiang C, Kelly WK, Patel J, Halabi S, Furukawa Y, Wheeler HE, Sibley AB, Lassiter C, Weisman L, Watson D, Krens SD, Mulkey F, Renn CL, Small EJ, Febbo PG, Shterev I, Kroetz DL, Friedman PN, Mahoney JF, Carducci MA, Kelley MJ, Nakamura Y, Kubo M, Dorsey SG, Dolan ME, Morris MJ, Ratain MJ, McLeod HL, 2016. Pharmacogenetic Discovery in CALGB (Alliance) 90401 and Mechanistic Validation of a VAC14 Polymorphism that Increases Risk of Docetaxel-Induced Neuropathy. Clinical cancer research: an official journal of the American Association for Cancer Research 22, 4890–4900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hertz DL, Roy S, Jack J, Motsinger-Reif AA, Drobish A, Clark LS, Carey LA, Dees EC, McLeod HL, 2014. Genetic heterogeneity beyond CYP2C8*3 does not explain differential sensitivity to paclitaxel-induced neuropathy. Breast Cancer Res Treat 145, 245–254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hertz DL, Roy S, Motsinger-Reif AA, Drobish A, Clark LS, McLeod HL, Carey LA, Dees EC, 2013. CYP2C8*3 increases risk of neuropathy in breast cancer patients treated with paclitaxel. Ann Oncol 24, 1472–1478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hilkens PH, ven den Bent MJ, 1997. Chemotherapy-induced peripheral neuropathy. J Peripher Nerv Syst 2, 350–361. [PubMed] [Google Scholar]
- Janes K, Doyle T, Bryant L, Esposito E, Cuzzocrea S, Ryerse J, Bennett GJ, Salvemini D, 2013. Bioenergetic deficits in peripheral nerve sensory axons during chemotherapy-induced neuropathic pain resulting from peroxynitrite-mediated post-translational nitration of mitochondrial superoxide dismutase. Pain 154, 2432–2440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jin HW, Flatters SJ, Xiao WH, Mulhern HL, Bennett GJ, 2008. Prevention of paclitaxel-evoked painful peripheral neuropathy by acetyl-L-carnitine: effects on axonal mitochondria, sensory nerve fiber terminal arbors, and cutaneous Langerhans cells. Experimental neurology 210, 229–237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kidd JF, Pilkington MF, Schell MJ, Fogarty KE, Skepper JN, Taylor CW, Thorn P, 2002. Paclitaxel affects cytosolic calcium signals by opening the mitochondrial permeability transition pore. J Biol Chem 277, 6504–6510. [DOI] [PubMed] [Google Scholar]
- Kim E, Hwang S-H, Kim H-K, Abdi S, Kim HK, 2019. Losartan, an Angiotensin II Type 1 Receptor Antagonist, Alleviates Mechanical Hyperalgesia in a Rat Model of Chemotherapy-Induced Neuropathic Pain by Inhibiting Inflammatory Cytokines in the Dorsal Root Ganglia. Molecular Neurobiology. [DOI] [PubMed] [Google Scholar]
- Ko MH, Hu ME, Hsieh YL, Lan CT, Tseng TJ, 2014. Peptidergic intraepidermal nerve fibers in the skin contribute to the neuropathic pain in paclitaxel-induced peripheral neuropathy. Neuropeptides 48, 109–117. [DOI] [PubMed] [Google Scholar]
- Konings PN, Makkink WK, van Delft AM, Ruigt GS, 1994. Reversal by NGF of cytostatic drug-induced reduction of neurite outgrowth in rat dorsal root ganglia in vitro. Brain Res 640, 195–204. [DOI] [PubMed] [Google Scholar]
- Kulkarni AA, Boora G, Kanwar R, Ruddy KJ, Banck MS, Le-Lindqwister N, Therneau TM, Loprinzi CL, Beutler AS, 2015. RWDD3 and TECTA variants not linked to paclitaxel induced peripheral neuropathy in North American trial Alliance N08C1. Acta Oncol 54, 1227–1229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leandro-Garcia LJ, Inglada-Perez L, Pita G, Hjerpe E, Leskela S, Jara C, Mielgo X, Gonzalez-Neira A, Robledo M, Avall-Lundqvist E, Green H, Rodriguez-Antona C, 2013. Genome-wide association study identifies ephrin type A receptors implicated in paclitaxel induced peripheral sensory neuropathy. Journal of medical genetics 50, 599–605. [DOI] [PubMed] [Google Scholar]
- Leandro-Garcia LJ, Leskela S, Jara C, Green H, Avall-Lundqvist E, Wheeler HE, Dolan ME, Inglada-Perez L, Maliszewska A, de Cubas AA, Comino-Mendez I, Mancikova V, Cascon A, Robledo M, Rodriguez-Antona C, 2012. Regulatory polymorphisms in beta-tubulin IIa are associated with paclitaxel-induced peripheral neuropathy. Clinical cancer research: an official journal of the American Association for Cancer Research 18, 4441–4448. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee CC, Perkins BA, Kayaniyil S, Harris SB, Retnakaran R, Gerstein HC, Zinman B, Hanley AJ, 2015a. Peripheral Neuropathy and Nerve Dysfunction in Individuals at High Risk for Type 2 Diabetes: The PROMISE Cohort. Diabetes care 38, 793–800. [DOI] [PubMed] [Google Scholar]
- Lee MY, Apellaniz-Ruiz M, Johansson I, Vikingsson S, Bergmann TK, Brosen K, Green H, Rodriguez-Antona C, Ingelman-Sundberg M, 2015b. Role of cytochrome P450 2C8*3 (CYP2C8*3) in paclitaxel metabolism and paclitaxel-induced neurotoxicity. Pharmacogenomics 16, 929–937. [DOI] [PubMed] [Google Scholar]
- Leskela S, Jara C, Leandro-Garcia LJ, Martinez A, Garcia-Donas J, Hernando S, Hurtado A, Vicario JC, Montero-Conde C, Landa I, Lopez-Jimenez E, Cascon A, Milne RL, Robledo M, Rodriguez-Antona C, 2011. Polymorphisms in cytochromes P450 2C8 and 3A5 are associated with paclitaxel neurotoxicity. Pharmacogenomics J 11, 121–129. [DOI] [PubMed] [Google Scholar]
- Li L, Li J, Zuo Y, Dang D, Frost JA, Yang Q, 2019. Activation of KCNQ Channels Prevents Paclitaxel-Induced Peripheral Neuropathy and Associated Neuropathic Pain. The Journal of Pain: Official Journal of the American Pain Society 20, 528–539. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y, Adamek P, Zhang H, Tatsui CE, Rhines LD, Mrozkova P, Li Q, Kosturakis AK, Cassidy RM, Harrison DS, Cata JP, Sapire K, Zhang H, Kennamer-Chapman RM, Jawad AB, Ghetti A, Yan J, Palecek J, Dougherty PM, 2015. The Cancer Chemotherapeutic Paclitaxel Increases Human and Rodent Sensory Neuron Responses to TRPV1 by Activation of TLR4. J Neurosci 35, 13487–13500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y, North RY, Rhines LD, Tatsui CE, Rao G, Edwards DD, Cassidy RM, Harrison DS, Johansson CA, Zhang H, Dougherty PM, 2018. DRG Voltage-Gated Sodium Channel 1.7 Is Upregulated in Paclitaxel-Induced Neuropathy in Rats and in Humans with Neuropathic Pain. The Journal of Neuroscience: The Official Journal of the Society for Neuroscience 38, 1124–1136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y, Tatsui CE, Rhines LD, North RY, Harrison DS, Cassidy RM, Johansson CA, Kosturakis AK, Edwards DD, Zhang H, Dougherty PM, 2017. Dorsal root ganglion neurons become hyperexcitable and increase expression of voltage-gated T-type calcium channels (Cav3.2) in paclitaxel-induced peripheral neuropathy. Pain 158, 417–429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y, Zhang H, Zhang H, Kosturakis AK, Jawad AB, Dougherty PM, 2014. Toll-like receptor 4 signaling contributes to Paclitaxel-induced peripheral neuropathy. The Journal of Pain: Official Journal of the American Pain Society 15, 712–725. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lisse TS, Middleton LJ, Pellegrini AD, Martin PB, Spaulding EL, Lopes O, Brochu EA, Carter EV, Waldron A, Rieger S, 2016. Paclitaxel-induced epithelial damage and ectopic MMP-13 expression promotes neurotoxicity in zebrafish. Proc Natl Acad Sci USA 113, E2189–2198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maestri A, De Pasquale Ceratti A, Cundari S, Zanna C, Cortesi E, Crinò L, 2005. A pilot study on the effect of acetyl-L-carnitine in paclitaxel- and cisplatin-induced peripheral neuropathy. Tumori 91, 135–138. [DOI] [PubMed] [Google Scholar]
- Maggioni D, Nicolini G, Chiorazzi A, Meregalli C, Cavaletti G, Tredici G, 2010. Different effects of erythropoietin in cisplatin- and docetaxel-induced neurotoxicity: an in vitro study. Journal of neuroscience research 88, 3171–3179. [DOI] [PubMed] [Google Scholar]
- Majithia N, Temkin SM, Ruddy KJ, Beutler AS, Hershman DL, Loprinzi CL, 2016. National Cancer Institute-supported chemotherapy-induced peripheral neuropathy trials: outcomes and lessons. Support Care Cancer 24, 1439–1447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Malacrida A, Meregalli C, Rodriguez-Menendez V, Nicolini G, 2019. Chemotherapy-Induced Peripheral Neuropathy and Changes in Cytoskeleton. International Journal of Molecular Sciences 20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Malgrange B, Delree P, Rigo JM, Baron H, Moonen G, 1994. Image analysis of neuritic regeneration by adult rat dorsal root ganglion neurons in culture: quantification of the neurotoxicity of anticancer agents and of its prevention by nerve growth factor or basic fibroblast growth factor but not brain-derived neurotrophic factor or neurotrophin-3. Journal of neuroscience methods 53, 111–122. [DOI] [PubMed] [Google Scholar]
- Manjavachi MN, Passos GF, Trevisan G, Araújo SB, Pontes JP, Fernandes ES, Costa R, Calixto JB, 2019. Spinal blockage of CXCL1 and its receptor CXCR2 inhibits paclitaxel-induced peripheral neuropathy in mice. Neuropharmacology 151, 136–143. [DOI] [PubMed] [Google Scholar]
- Materazzi S, Fusi C, Benemei S, Pedretti P, Patacchini R, Nilius B, Prenen J, Creminon C, Geppetti P, Nassini R, 2012. TRPA1 and TRPV4 mediate paclitaxel-induced peripheral neuropathy in mice via a glutathione-sensitive mechanism. Pflugers Archiv: European Journal of Physiology 463, 561–569. [DOI] [PubMed] [Google Scholar]
- McWhinney-Glass S, Winham SJ, Hertz DL, Yen Revollo J, Paul J, He Y, Brown R, Motsinger-Reif AA, McLeod HL, Scottish Gynaecological Clinical Trials, G., 2013. Cumulative genetic risk predicts platinum/taxane-induced neurotoxicity. Clinical cancer research: an official journal of the American Association for Cancer Research 19, 5769–5776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Melli G, Jack C, Lambrinos GL, Ringkamp M, Hoke A, 2006. Erythropoietin protects sensory axons against paclitaxel-induced distal degeneration. Neurobiology of disease 24, 525–530. [DOI] [PubMed] [Google Scholar]
- Melli G, Taiana M, Camozzi F, Triolo D, Podini P, Quattrini A, Taroni F, Lauria G, 2008. Alpha-lipoic acid prevents mitochondrial damage and neurotoxicity in experimental chemotherapy neuropathy. Experimental neurology 214, 276–284. [DOI] [PubMed] [Google Scholar]
- Miao H, Xu J, Xu D, Ma X, Zhao X, Liu L, 2018. Nociceptive behavior induced by chemotherapeutic paclitaxel and beneficial role of antioxidative pathways. Physiological Research. [DOI] [PubMed] [Google Scholar]
- Miaskowski C, Mastick J, Paul SM, Abrams G, Cheung S, Sabes JH, Kober KM, Schumacher M, Conley YP, Topp K, Smoot B, Mausisa G, Mazor M, Wallhagen M, Levine JD, 2018. Impact of chemotherapy-induced neurotoxicities on adult cancer survivors’ symptom burden and quality of life. J Cancer Surviv 12, 234–245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mir O, Alexandre J, Tran A, Durand JP, Pons G, Treluyer JM, Goldwasser F, 2009. Relationship between GSTP1 Ile(105)Val polymorphism and docetaxel-induced peripheral neuropathy: clinical evidence of a role of oxidative stress in taxane toxicity. Ann Oncol 20, 736–740. [DOI] [PubMed] [Google Scholar]
- Mironov SL, Ivannikov MV, Johansson M, 2005. [Ca2+]i signaling between mitochondria and endoplasmic reticulum in neurons is regulated by microtubules. From mitochondrial permeability transition pore to Ca2+-induced Ca2+ release. J Biol Chem 280, 715–721. [DOI] [PubMed] [Google Scholar]
- Miyano K, Tang HB, Nakamura Y, Morioka N, Inoue A, Nakata Y, 2009. Paclitaxel and vinorelbine, evoked the release of substance P from cultured rat dorsal root ganglion cells through different PKC isoform-sensitive ion channels. Neuropharmacology 57, 25–32. [DOI] [PubMed] [Google Scholar]
- Nyrop KA, Deal AM, Shachar SS, Basch E, Reeve BB, Choi SK, Lee JT, Wood WA, Anders CK, Carey LA, Dees EC, Jolly TA, Reeder-Hayes KE, Kimmick GG, Karuturi MS, Reinbolt RE, Speca JC, Muss HB, 2019. Patient-Reported Toxicities During Chemotherapy Regimens in Current Clinical Practice for Early Breast Cancer. Oncologist 24, 762–771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Okubo K, Takahashi T, Sekiguchi F, Kanaoka D, Matsunami M, Ohkubo T, Yamazaki J, Fukushima N, Yoshida S, Kawabata A, 2011. Inhibition of T-type calcium channels and hydrogen sulfide-forming enzyme reverses paclitaxel-evoked neuropathic hyperalgesia in rats. Neuroscience 188, 148–156. [DOI] [PubMed] [Google Scholar]
- Park SB, Kwok JB, Asher R, Lee CK, Beale P, Selle F, Friedlander M, 2017. Clinical and genetic predictors of paclitaxel neurotoxicity based on patient- versus clinician-reported incidence and severity of neurotoxicity in the ICON7 trial. Ann Oncol 28, 2733–2740. [DOI] [PubMed] [Google Scholar]
- Park SB, Kwok JB, Loy CT, Friedlander ML, Lin CS, Krishnan AV, Lewis CR, Kiernan MC, 2014. Paclitaxel-induced neuropathy: potential association of MAPT and GSK3B genotypes. BMC cancer 14, 993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pease-Raissi SE, Pazyra-Murphy MF, Li Y, Wachter F, Fukuda Y, Fenstermacher SJ, Barclay LA, Bird GH, Walensky LD, Segal RA, 2017. Paclitaxel Reduces Axonal Bclw to Initiate IP3R1-Dependent Axon Degeneration. Neuron 96, 373–386 e376. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pevida M, Lastra A, Hidalgo A, Baamonde A, Menéndez L, 2013. Spinal CCL2 and microglial activation are involved in paclitaxel-evoked cold hyperalgesia. Brain Research Bulletin 95, 21–27. [DOI] [PubMed] [Google Scholar]
- Pittman SK, Gracias NG, Fehrenbacher JC, 2016. Nerve growth factor alters microtubule targeting agent-induced neurotransmitter release but not MTA-induced neurite retraction in sensory neurons. Exp Neurol 279, 104–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pittman SK, Gracias NG, Vasko MR, Fehrenbacher JC, 2014. Paclitaxel alters the evoked release of calcitonin gene-related peptide from rat sensory neurons in culture. Experimental neurology 253, 146–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schneider BP, Lai D, Shen F, Jiang G, Radovich M, Li L, Gardner L, Miller KD, O’Neill A, Sparano JA, Xue G, Foroud T, Sledge GW Jr., 2016. Charcot-Marie-Tooth gene, SBF2, associated with taxane-induced peripheral neuropathy in African Americans. Oncotarget 7, 82244–82253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schneider BP, Li L, Radovich M, Shen F, Miller KD, Flockhart DA, Jiang G, Vance G, Gardner L, Vatta M, Bai S, Lai D, Koller D, Zhao F, O’Neill A, Smith ML, Railey E, White C, Partridge A, Sparano J, Davidson NE, Foroud T, Sledge GW Jr., 2015. Genome-Wide Association Studies for Taxane-Induced Peripheral Neuropathy in ECOG-5103 and ECOG-1199. Clinical cancer research: an official journal of the American Association for Cancer Research 21, 5082–5091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sekiguchi F, Domoto R, Nakashima K, Yamasoba D, Yamanishi H, Tsubota M, Wake H, Nishibori M, Kawabata A, 2018. Paclitaxel-induced HMGB1 release from macrophages and its implication for peripheral neuropathy in mice: Evidence for a neuroimmune crosstalk. Neuropharmacology 141, 201–213. [DOI] [PubMed] [Google Scholar]
- Seretny M, Currie GL, Sena ES, Ramnarine S, Grant R, MacLeod MR, Colvin LA, Fallon M, 2014. Incidence, prevalence, and predictors of chemotherapy-induced peripheral neuropathy: A systematic review and meta-analysis. Pain 155, 2461–2470. [DOI] [PubMed] [Google Scholar]
- Shan Z, Cai S, Yu J, Zhang Z, Vallecillo TGM, Serafini MJ, Thomas AM, Pham NYN, Bellampalli SS, Moutal A, Zhou Y, Xu G-B, Xu Y-M, Luo S, Patek M, Streicher JM, Gunatilaka AAL, Khanna R, 2019. Reversal of Peripheral Neuropathic Pain by the Small-Molecule Natural Product Physalin F via Block of CaV2.3 (R-Type) and CaV2.2 (N-Type) Voltage-Gated Calcium Channels. ACS chemical neuroscience. [DOI] [PubMed] [Google Scholar]
- Shemesh OA, Spira ME, 2010. Paclitaxel induces axonal microtubules polar reconfiguration and impaired organelle transport: implications for the pathogenesis of paclitaxel-induced polyneuropathy. Acta neuropathologica 119, 235–248. [DOI] [PubMed] [Google Scholar]
- Siau C, Xiao W, Bennett GJ, 2006. Paclitaxel- and vincristine-evoked painful peripheral neuropathies: loss of epidermal innervation and activation of Langerhans cells. Experimental neurology 201, 507–514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Singh J, Saha L, Singh N, Kumari P, Bhatia A, Chakrabarti A, 2019. Study of nuclear factor-2 erythroid related factor-2 activator, berberine, in paclitaxel induced peripheral neuropathy pain model in rats. The Journal of Pharmacy and Pharmacology 71, 797–805. [DOI] [PubMed] [Google Scholar]
- Sissung TM, Mross K, Steinberg SM, Behringer D, Figg WD, Sparreboom A, Mielke S, 2006. Association of ABCB1 genotypes with paclitaxel-mediated peripheral neuropathy and neutropenia. Eur J Cancer 42, 2893–2896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith EM, Pang H, Cirrincione C, Fleishman S, Paskett ED, Ahles T, Bressler LR, Fadul CE, Knox C, Le-Lindqwister N, Gilman PB, Shapiro CL, 2013. Effect of duloxetine on pain, function, and quality of life among patients with chemotherapy-induced painful peripheral neuropathy: a randomized clinical trial. Jama 309, 1359–1367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith JA, Slusher BS, Wozniak KM, Farah MH, Smiyun G, Wilson L, Feinstein S, Jordan MA, 2016. Structural Basis for Induction of Peripheral Neuropathy by Microtubule-Targeting Cancer Drugs. Cancer Res 76, 5115–5123. [DOI] [PubMed] [Google Scholar]
- Sucheston LE, Zhao H, Yao S, Zirpoli G, Liu S, Barlow WE, Moore HC, Thomas Budd G, Hershman DL, Davis W, Ciupak GL, Stewart JA, Isaacs C, Hobday TJ, Salim M, Hortobagyi GN, Gralow JR, Livingston RB, Albain KS, Hayes DF, Ambrosone CB, 2011. Genetic predictors of taxane-induced neurotoxicity in a SWOG phase III intergroup adjuvant breast cancer treatment trial (S0221). Breast Cancer Res Treat 130, 993–1002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sucheston-Campbell LE, Clay-Gilmour AI, Barlow WE, Budd GT, Stram DO, Haiman CA, Sheng X, Yan L, Zirpoli G, Yao S, Jiang C, Owzar K, Hershman D, Albain KS, Hayes DF, Moore HC, Hobday TJ, Stewart JA, Rizvi A, Isaacs C, Salim M, Gralow JR, Hortobagyi GN, Livingston RB, Kroetz DL, Ambrosone CB, 2018. Genome-wide meta-analyses identifies novel taxane-induced peripheral neuropathy-associated loci. Pharmacogenet Genomics 28, 49–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun H, Guo X, Wang Z, Wang P, Zhang Z, Dong J, Zhuang R, Zhou Y, Ma G, Cai W, 2019. Alphalipoic Acid Prevents Oxidative Stress and Peripheral Neuropathy in Nab-Paclitaxel-Treated Rats through the Nrf2 Signalling Pathway. Oxidative Medicine and Cellular Longevity 2019, 3142732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tanabe Y, Shimizu C, Hamada A, Hashimoto K, Ikeda K, Nishizawa D, Hasegawa J, Shimomura A, Ozaki Y, Tamura N, Yamamoto H, Yunokawa M, Yonemori K, Takano T, Kawabata H, Tamura K, Fujiwara Y, 2017. Paclitaxel-induced sensory peripheral neuropathy is associated with an ABCB1 single nucleotide polymorphism and older age in Japanese. Cancer chemotherapy and pharmacology 79, 1179–1186. [DOI] [PubMed] [Google Scholar]
- Theiss C, Meller K, 2000. Taxol impairs anterograde axonal transport of microinjected horseradish peroxidase in dorsal root ganglia neurons in vitro. Cell and tissue research 299, 213–224. [DOI] [PubMed] [Google Scholar]
- Ustinova EE, Shurin GV, Gutkin DW, Shurin MR, 2013. The role of TLR4 in the paclitaxel effects on neuronal growth in vitro. PLoS One 8, e56886. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Varbiro G, Veres B, Gallyas F, Sumegi B, 2001. Direct effect of Taxol on free radical formation and mitochondrial permeability transition. Free Radic Biol Med 31, 548–558. [DOI] [PubMed] [Google Scholar]
- Wainger BJ, Buttermore ED, Oliveira JT, Mellin C, Lee S, Saber WA, Wang AJ, Ichida JK, Chiu IM, Barrett L, Huebner EA, Bilgin C, Tsujimoto N, Brenneis C, Kapur K, Rubin LL, Eggan K, Woolf CJ, 2015. Modeling pain in vitro using nociceptor neurons reprogrammed from fibroblasts. Nat Neurosci 18, 17–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Warwick RA, Hanani M, 2013. The contribution of satellite glial cells to chemotherapy-induced neuropathic pain. European Journal of Pain (London, England) 17, 571–580. [DOI] [PubMed] [Google Scholar]
- Wheeler HE, Gamazon ER, Wing C, Njiaju UO, Njoku C, Baldwin RM, Owzar K, Jiang C, Watson D, Shterev I, Kubo M, Zembutsu H, Winer EP, Hudis CA, Shulman LN, Nakamura Y, Ratain MJ, Kroetz DL, Cancer, Leukemia Group, B., Cox NJ, Dolan ME, 2013. Integration of cell line and clinical trial genome-wide analyses supports a polygenic architecture of Paclitaxel-induced sensory peripheral neuropathy. Clinical cancer research: an official journal of the American Association for Cancer Research 19, 491–499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wing C, Komatsu M, Delaney SM, Krause M, Wheeler HE, Dolan ME, 2017. Application of stem cell derived neuronal cells to evaluate neurotoxic chemotherapy. Stem Cell Res 22, 79–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wozniak KM, Vornov JJ, Wu Y, Liu Y, Carozzi VA, Rodriguez-Menendez V, Ballarini E, Alberti P, Pozzi E, Semperboni S, Cook BM, Littlefield BA, Nomoto K, Condon K, Eckley S, DesJardins C, Wilson L, Jordan MA, Feinstein SC, Cavaletti G, Polydefkis M, Slusher BS, 2018. Peripheral neuropathy induced by microtubule-targeted chemotherapies: insights into acute injury and long-term recovery. Cancer research 78, 817–829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu J, Hocevar M, Bie B, Foss JF, Naguib M, 2019. Cannabinoid Type 2 Receptor System Modulates Paclitaxel-Induced Microglial Dysregulation and Central Sensitization in Rats. The Journal of Pain: Official Journal of the American Pain Society 20, 501–514. [DOI] [PubMed] [Google Scholar]
- Wu Z, Wang S, Wu I, Mata M, Fink DJ, 2015. Activation of TLR-4 to produce tumour necrosis factor-α in neuropathic pain caused by paclitaxel. European Journal of Pain (London, England) 19, 889–898. [DOI] [PubMed] [Google Scholar]
- Xiao WH, Zheng H, Zheng FY, Nuydens R, Meert TF, Bennett GJ, 2011. Mitochondrial abnormality in sensory, but not motor, axons in paclitaxel-evoked painful peripheral neuropathy in the rat. Neuroscience 199, 461–469. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu J, Zhang L, Xie M, Li Y, Huang P, Saunders TL, Fox DA, Rosenquist R, Lin F, 2018. Role of Complement in a Rat Model of Paclitaxel-Induced Peripheral Neuropathy. Journal of Immunology (Baltimore, Md.: 1950) 200, 4094–4101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang IH, Siddique R, Hosmane S, Thakor N, Hoke A, 2009a. Compartmentalized microfluidic culture platform to study mechanism of paclitaxel-induced axonal degeneration. Experimental neurology 218, 124–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang IH, Siddique R, Hosmane S, Thakor N, Höke A, 2009b. Compartmentalized microfluidic culture platform to study mechanism of paclitaxel-induced axonal degeneration. Experimental neurology 218, 124–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yilmaz E, Gold MS, 2016. Paclitaxel-induced increase in NCX activity in subpopulations of nociceptive afferents: A protective mechanism against chemotherapy-induced peripheral neuropathy? Cell Calcium 60, 25–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yilmaz E, Watkins SC, Gold MS, 2017. Paclitaxel-induced increase in mitochondrial volume mediates dysregulation of intracellular Ca2+ in putative nociceptive glabrous skin neurons from the rat. Cell Calcium 62, 16–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang H, Boyette-Davis JA, Kosturakis AK, Li Y, Yoon S-Y, Walters ET, Dougherty PM, 2013. Induction of monocyte chemoattractant protein-1 (MCP-1) and its receptor CCR2 in primary sensory neurons contributes to paclitaxel-induced peripheral neuropathy. The Journal of Pain: Official Journal of the American Pain Society 14, 1031–1044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang H, Dougherty PM, 2014. Enhanced excitability of primary sensory neurons and altered gene expression of neuronal ion channels in dorsal root ganglion in paclitaxel-induced peripheral neuropathy. Anesthesiology 120, 1463–1475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang H, Li Y, de Carvalho-Barbosa M, Kavelaars A, Heijnen CJ, Albrecht PJ, Dougherty PM, 2016. Dorsal Root Ganglion Infiltration by Macrophages Contributes to Paclitaxel Chemotherapy-Induced Peripheral Neuropathy. The Journal of Pain: Official Journal of the American Pain Society 17, 775–786. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang W, Bianchi L, Lee WH, Wang Y, Israel S, Driscoll M, 2008. Intersubunit interactions between mutant DEG/ENaCs induce synthetic neurotoxicity. Cell Death Differ 15, 1794–1803. [DOI] [PubMed] [Google Scholar]
- Zhao X, Liu L, Wang Y, Wang G, Zhao Y, Zhang Y, 2019. Electroacupuncture enhances antioxidative signal pathway and attenuates neuropathic pain induced by chemotherapeutic paclitaxel. Physiological Research. [DOI] [PubMed] [Google Scholar]
- Zheng H, Xiao WH, Bennett GJ, 2011. Functional deficits in peripheral nerve mitochondria in rats with paclitaxel- and oxaliplatin-evoked painful peripheral neuropathy. Exp Neurol 232, 154–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
