Вы находитесь на странице: 1из 11

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/323318626

Beyond symptomatic relief for chemotherapy-induced peripheral


neuropathy: Targeting the source

Article  in  Cancer · February 2018


DOI: 10.1002/cncr.31248

CITATIONS READS

4 221

4 authors, including:

Jiacheng Ma Annemieke Kavelaars


University of Texas MD Anderson Cancer Center University of Texas MD Anderson Cancer Center
14 PUBLICATIONS   127 CITATIONS    465 PUBLICATIONS   11,538 CITATIONS   

SEE PROFILE SEE PROFILE

Cobi Heijnen
University Medical Center Utrecht
358 PUBLICATIONS   10,328 CITATIONS   

SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Genomic mechanisms for the devastating impact of combat trauma on mental health View project

Chronic Pain and Depression View project

All content following this page was uploaded by Jiacheng Ma on 27 February 2018.

The user has requested enhancement of the downloaded file.


Review Article

Beyond Symptomatic Relief for Chemotherapy-Induced


Peripheral Neuropathy: Targeting the Source
Jiacheng Ma, PhD1; Annemieke Kavelaars, PhD1; Patrick M. Dougherty, PhD2; and Cobi J. Heijnen, PhD 1

Chemotherapy-induced peripheral neuropathy (CIPN) is a serious adverse side effect of many chemotherapeutic agents, affecting
>60% of patients with cancer. Moreover, CIPN persists long into survivorship in approximately 20% to 30% of these patients. To the
authors’ knowledge, no drugs have been approved to date by the US Food and Drug Administration to effectively manage
chemotherapy-induced neuropathic pain. The majority of the drugs tested for the management of CIPN aim at symptom relief, includ-
ing pain and paresthesia, yet are not very efficacious. The authors propose that there is a need to acquire a more thorough under-
standing of the etiology of CIPN so that effective, mechanism-based, disease-modifying interventions can be developed. It is
important to note that such interventions should not interfere with the antitumor effects of chemotherapy. Mitochondria are rod-
shaped cellular organelles that represent the powerhouses of the cell, in that they convert oxygen and nutrients into the cellular
energy “currency” adenosine triphosphate. In addition, mitochondria regulate cell death. Neuronal mitochondrial dysfunction and the
associated nitro-oxidative stress represent crucial final common pathways of CIPN. Herein, the authors discuss the potential to pre-
vent or reverse CIPN by protecting mitochondria and/or inhibiting nitro-oxidative stress with novel potential drugs, including the
mitochondrial protectant pifithrin-l, histone deacetylase 6 inhibitors, metformin, antioxidants, peroxynitrite decomposition catalysts,
and anti-inflammatory mediators including interleukin 10. This review hopefully will contribute toward bridging the gap between pre-
clinical research and the development of realistic novel therapeutic strategies to prevent or reverse the devastating neurotoxic effects
of chemotherapy on the (peripheral) nervous system. Cancer 2018;000:000-000. V C 2018 American Cancer Society.

KEYWORDS: cancer, chemotherapy, chemotherapy-induced peripheral neuropathy (CIPN), intervention, mitochondria, nitro-oxidative,
pain, symptom.

INTRODUCTION
In 2017, it is estimated that 1.7 million new cases of cancer will have been diagnosed in the United States alone. Despite
this grim statistic, the numbers of successful cancer treatments and cancer survivors have increased dramatically over the
last decade.1,2 However, managing cancer-related pain remains challenging. Cancer-related pain can be categorized either
as pain related to active tumor growth or as pain resulting from cancer treatment. The latter, when produced by chemo-
therapeutic agents, often manifests as chemotherapy-induced peripheral neuropathy (CIPN).3
CIPN is one of the most common adverse effects of cancer chemotherapy, affecting >60% of patients.3 A high inci-
dence of CIPN is associated with widely used chemotherapeutics, including platinum derivatives (cisplatin, oxaliplatin),
taxanes (paclitaxel, docetaxel), vinca alkaloids (vincristine, vinblastine), bortezomib, ixabepilone, and thalidomide.4,5
CIPN symptoms include persistent shooting, stabbing, or burning pain even in the absence of potentially painful stimuli,
and loss of sensation or “numbness.” These symptoms typically present in a “stocking-and-glove” distribution, starting in
the toes and fingers and spreading to the legs and arms as the disorder progresses.4 CIPN symptoms often cause significant
loss of functional abilities; moreover, CIPN represents a therapeutic challenge, given the limited treatment options and
the poor effectiveness of currently available therapies.5,6 CIPN not only severely affects quality of life, but also frequently
leads to chemotherapy dose reductions and even early cessation, thereby hampering treatment effectiveness. Several studies
with large subsets of patients with cancer have shown that CIPN does not resolve at the time of treatment completion, but
persists long into survivorship.3,7,8 Therefore, prevention or management of CIPN is key to the effective control of cancer
and to improvements in quality of life for patients with cancer and cancer survivors.
Current clinical strategies for CIPN aim primarily at symptom relief, with a focus on pain. Unfortunately, anticon-
vulsant and antidepressant drugs, which are first-line treatments for the symptomatic relief of neuropathic pain, have

Corresponding author: Cobi J. Heijnen, PhD, Division of Internal Medicine, Department of Symptom Research, The University of Texas MD Anderson Cancer Cen-
ter, 1515 Holcombe Blvd, Unit 384, Houston, TX 77030; cjheijnen@mdanderson.org
1
Neuroimmunology Laboratory, Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, Texas; 2Department of Pain
Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas

We thank Jeanie F. Woodruff, BS, ELS, for scientific editing.

DOI: 10.1002/cncr.31248, Received: November 17, 2017; Revised: December 21, 2017; Accepted: December 29, 2017, Published online Month 00, 2018 in
Wiley Online Library (wileyonlinelibrary.com)

Cancer Month 00, 2018 1


Review Article

reportedly had only limited success9; only the serotonin- mitochondria with compromised functionality.21 Vincris-
norepinephrine reuptake inhibitors duloxetine and venlafax- tine and bortezomib also dysregulate mitochondrial Ca21
ine have been shown to ameliorate chemotherapy-induced signaling, leading to defective energy production.22-24
(oxaliplatin or paclitaxel) neuropathic pain in phase 3 Persistent mitochondrial dysfunction generates
placebo-controlled randomized clinical trials.10,11 Other nitro-oxidative stress (described below), which causes
agents, including the Ca21 channel blockers gabapentin and nitro-oxidative damage to mitochondrial DNA and pro-
pregabalin and the Na1 channel blockers carbamazepine teins, further aggravating mitochondrial damage and lead-
and oxcarbazepine, either failed in randomized trials or ing to energy deficiency in sensory neurons.25 Consistent
demonstrated effectiveness only in small, non–placebo- with the mitotoxicity theory of CIPN, swollen and vacuo-
controlled trials.12-17 The development of novel treatments lated mitochondria appear in peripheral nerves and dorsal
to prevent or reverse CIPN is a pressing need that will root ganglion (DRG) neuronal cell bodies in rodent mod-
require an in-depth understanding of the underlying els of peripheral neuropathy induced by paclitaxel,26,27
mechanisms. cisplatin,28 oxaliplatin,29 bortezomib,24 or vincristine.24
Herein, we review current knowledge regarding the These abnormalities in mitochondrial morphology are
pathophysiological mechanisms underlying CIPN, with a accompanied by deficiencies in mitochondrial bioenerget-
focus on mitochondrial dysfunction, nitro-oxidative ics in DRG neurons and peripheral nerves in these models
stress, and neuroinflammation. We also describe potential of CIPN.28,30-32 More specifically, impairments in mito-
novel avenues for the development of mechanism-based chondrial basal respiration, maximal respiration capacity,
therapeutic strategies for the prevention and treatment of and ATP production are observed.28,30-32
CIPN. A major concern in the development of novel ther- Chemotherapy-induced mitochondrial dysfunction
apeutic strategies is that such agents might interfere with and the concomitant energy deficits in sensory neurons
tumor control by the chemotherapeutics. An optimal are believed to lead to the failure of these cells to sustain
solution would be to identify agents that protect against normal cellular activities, and ultimately to the symptoms
CIPN while enhancing tumor control. Therefore, we of CIPN. For example, the formation of intraepidermal
have focused on candidate agents that have the potential nerve fibers (IENFs) requires nerve fibers originating
to meet both goals. from subepidermal nerve fiber bundles to traverse the epi-
dermal basal lamina and branch within the epidermis,
Targeting Neuronal Mitochondrial Dysfunction
which is a highly bioenergetically demanding process.19,30
Mitochondria are double-membrane–bound organelles
In CIPN, the lack of a sufficient energy supply has been
that function as a cell’s “power plant,” converting
proposed to result in the inability of IENFs to sprout
nutrients into the cellular energy “currency” adenosine tri-
within the epidermis19,30 and thus to loss of IENFs, a clin-
phosphate (ATP), primarily through oxidative phosphor-
ical diagnostic marker of CIPN.33 Mitochondrial deficits
ylation. In neuronal cells, mitochondrial oxidative
also are likely to lead to dysfunction of the Na1 /K1
phosphorylation produces >90% of the ATP required for
exchanger, a major energy consumer in neuronal cells.34
maintaining normal cellular activities. Failure of the mito-
Dysfunction of the Na1 /K1 exchanger disrupts the elec-
chondria to synthesize adequate amounts of ATP for neu-
trochemical gradient across the cellular membrane,
ronal activities that require high energy, such as action
thereby facilitating spontaneous discharges in sensory
potential generation, synaptic transmission, and axonal
neurons,35 which is another characteristic of CIPN that
transport, is thought to be one of the prime mechanisms
may well be responsible for the spontaneous shooting and
underlying CIPN.18,19
burning pain that many patients report.
A wide array of chemotherapeutic agents that induce
CIPN cause mitochondrial dysfunction in peripheral sen- Preventing mitochondrial damage by targeting
sory neurons, despite marked differences in the mecha- mitochondrial p53
nisms via which they exert their antitumor effects. For Given the fundamental role of mitochondrial dysfunction
example, cisplatin and oxaliplatin intercalate mitochon- in the peripheral nervous system in the pathogenesis of
drial DNA, leading to failure in mitochondrial protein CIPN, agents that protect mitochondria may represent
synthesis and to mitochondrial dysfunction.20 Paclitaxel good candidates for disease prevention.
induces opening of the mitochondrial permeability transi- Mitochondrial translocation of the tumor suppres-
tion pore, which evokes mitochondrial membrane depo- sor molecule p53 has been identified as an early event
larization and Ca21 release, resulting in vacuolated leading to mitochondrial abnormalities in models of

2 Cancer Month 00, 2018


Novel Therapeutic Strategies for CIPN/Ma et al

neurological disorders.36,37 Recent studies have indicated inhibitor with cisplatin prevented cisplatin-induced neu-
that chemotherapeutic agents trigger a similar damage ropathic pain.
pathway. For example, we demonstrated that cisplatin It is interesting to note that HDAC6 inhibition also
treatment rapidly increases mitochondrial levels of p53 in has disease-modifying properties. Administration of the
DRG neurons, followed by a reduction in mitochondrial HDAC6 inhibitor ACY-1083 after the completion of
membrane potential and ultimately by the disruption of chemotherapy, when CIPN had fully developed, reversed
mitochondrial integrity and function.28 It is interesting to the mitochondrial bioenergetic deficits in distal peripheral
note that we found that the mitochondrial protectant nerves and DRG neurons.30 Consequently, HDAC6
pifithrin-l, which prevents mitochondrial accumulation inhibition reversed all symptoms of CIPN, including
of p53, also prevented paclitaxel-induced and cisplatin- mechanical allodynia, spontaneous pain, and sensory loss.
induced neuropathic pain and sensory loss and averted the HDAC6 inhibitors also stimulated the regrowth of
loss of IENFs in mouse models of CIPN.27,28 Pifithrin-l IENFs, presumably through promoting the distribution
not only prevented CIPN symptoms but also protected of bioenergetically functional mitochondria to nerve ter-
against loss of mitochondrial membrane potential, abnor- minals in which the energy-consuming processes take
malities in mitochondrial morphology, and functional place.30 It is important to note that both the preventive
mitochondrial deficiencies in DRG neurons.27,28 It is and restorative effects of HDAC6 inhibition were main-
important to note that because pifithrin-l targets the tained for months in mice after the termination of treat-
p53-mitochondrial pathway without affecting the tran- ment with the HDAC6 inhibitor, highlighting its disease-
scriptional functions of p53,38 it does not negatively affect modifying effects.30
the tumor-killing effects of chemotherapy.27,37 Rather, in Because HDAC6 inhibitors already are in clinical
vivo studies have shown that pifithrin-l can itself exert trials testing their potential capacity to increase the effi-
antitumor effects.39 In vitro, the coadministration of cacy of chemotherapy, they also are of interest as treat-
pifithrin-l with paclitaxel or cisplatin enhanced tumor ment of the neurotoxic side effects of cancer therapy.41,42
cell killing.27,40 Taken together, these data suggest that Additional clinical trials that include the evaluation of
the coadministration of pifithrin-l with chemotherapeu- CIPN parameters in patients treated with a combination
tics may represent a promising novel approach with which of HDAC6 inhibitors and paclitaxel or cisplatin as a sec-
to improve cancer treatment while protecting against the ondary outcome also currently are underway (Clinical-
development of CIPN.27,28 Trials.gov identifiers NCT02632071, NCT02661815,
and NCT02856568).

Preventing or reversing neuronal mitochondrial Preventing CIPN by targeting mitochondrial


abnormalities and CIPN by inhibiting histone pathways with metformin
deacetylase 6 Recent preclinical studies have suggested that metformin,
Pharmacological targeting of histone deacetylase 6 a drug approved by the US Food and Drug Administra-
(HDAC6) is another potential therapeutic strategy for tion (FDA) for glycemic control in patients with type 2
CIPN that targets the mitochondrial pathway and has diabetes, alleviates neuropathic and inflammatory pain in
potential antitumor effects.30,41,42 HDAC6 is a class IIB various models.46 We demonstrated that metformin also
histone deacetylase that has limited effect on histones and prevents cisplatin-induced neuropathic pain, numbness,
acts mainly in the cytosol, in which it deacetylates tubulin, and loss of IENFs.47 Although the exact mechanisms
heat shock protein 90, and many other substrates. The underlying these protective effects need to be delineated,
neuroprotective effects of HDAC6 inhibitors have been it is likely that metformin acts via modulation of
demonstrated in models of neurological disorders, includ- mitochondria-dependent cellular metabolism. Metfor-
ing Alzheimer disease and Charcot-Marie-Tooth dis- min’s beneficial effects on energy metabolism include
ease.43-45 The neuroprotective effects of HDAC6 enhanced activity of carnitine palmitoyltransferase 1, an
inhibitors in these models are associated with improved essential component of mitochondrial fatty acid oxida-
mitochondrial axonal transport and reduced oxidative tion.48 Metformin also promoted restoration of mito-
damage.43-45 We recently demonstrated that HDAC6 chondrial membrane potential and reversal of mutation-
inhibitors prevent cisplatin-induced deficits in axonal associated alterations in energy metabolism in a model of
mitochondrial transport in primary cultures of DRG neu- Parkinson disease49 and contributed toward the restora-
rons.30 In vivo, the coadministration of an HDAC6 tion of cellular energy by activating adenosine

Cancer Month 00, 2018 3


Review Article

monophosphate-activated protein kinase.50 Our prelimi- prevented mitochondrial damage and the development
nary data have indicated that the coadministration of met- of neuropathic pain in response to paclitaxel, oxaliplatin,
formin with cisplatin prevents the neuronal and bortezomib.24,61-64 The free radical scavenger phenyl
mitochondrial dysfunction that results from cisplatin N-tert-butylnitrone and the superoxide dismutase
treatment in a mouse model of CIPN. It is interesting to mimetic TEMPOL ameliorated mechanical allodynia in
note that epidemiological studies also have demonstrated rats treated with paclitaxel.65,66 Mitochondria-targeted
an association between metformin use and a decreased antioxidants, such as mitoVitE and melatonin, also
occurrence of cancer,51 suggesting that metformin may attenuated paclitaxel-induced neuropathic pain.67,68
have anticancer effects. In addition, metformin has been However, despite positive results from preclinical studies,
shown to promote tumor cell death in vitro.52-54 There- results from clinical studies are less satisfying. Clinical
fore, further exploration of metformin as a potential 2- trials using nutraceuticals with antioxidant properties,
pronged approach that targets both tumor growth and including vitamin E, acetyl-L-carnitine, glutamine, gluta-
CIPN is warranted. thione, vitamin B6, omega-3 fatty acids, and a-lipoic
acid, have reported controversial results.69,70 For exam-
Contribution of Nitro-Oxidative Stress to CIPN ple, acetyl-L-carnitine, despite demonstrating great
Downstream of mitochondrial dysfunction, oxidative promise in initial clinical studies,71 was not found to be
stress is another important common pathway that could effective in later clinical trials and even exacerbated
be targeted to prevent or reverse CIPN. Mitochondria are CIPN in some patients.72,73 Likewise, in a recent study
the most important endogenous source of reactive oxygen of 243 patients treated with a-lipoic acid while receiving
species (ROS). Whereas the amount of ROS produced is platinum chemotherapy, the agent demonstrated no pro-
tightly regulated in intact mitochondria, chemotherapy- tective effects.74 The failure of these antioxidants to alle-
induced mitochondrial damage leads to dysregulation of viate CIPN might be attributed to their rapidly
ROS production and the generation of oxidative stress.55 exhausted antioxidant activity. Moreover, the majority of
The peripheral nervous system is especially susceptible to antioxidants that have been tested clinically were chosen
chemotherapy-induced oxidative damage, due to its high for their availability and safety profiles rather than their
content of phospholipids, mitochondria-rich axoplasm, strong antioxidant properties. An alternative route to
and weak cellular antioxidant defenses.56 Evidence of the external antioxidant supplementation would be to boost
oxidative modification of lipids and proteins in the sciatic endogenous antioxidant responses. The transcription fac-
nerve and spinal cord has been reported in a rat model of tor peroxisome proliferator-activated receptor c (PPAR-c)
oxaliplatin-induced painful neuropathy.57 Increased ROS has emerged as a central hub in endogenous responses
levels in these tissues also were observed during the early against oxidative stress.75 Upregulation of key antioxidant
development of paclitaxel-induced neuropathic pain.58 enzymes, including MnSOD and glutathione peroxidase,
Nitrosative stress has emerged as an additional strong and enhanced mitochondrial energy metabolism have
contributor to chemotherapy-induced neuropathic pain. been observed at the time of PPAR-c activation.76,77 It is
Nitric oxide rapidly reacts with superoxide to form reactive important to note that the neuroprotective effects of
nitrogen species, mainly peroxynitrite, a highly reactive oxi- PPAR-c agonists have been reported in various neurologi-
dant under physiological conditions.59 Peroxynitrite inacti- cal disorders, including nerve injury-induced neuropathic
vates mitochondrial manganese superoxide dismutase pain and trigeminal neuropathic pain.78,79 Given that
(MnSOD), the primary antioxidant enzyme that keeps cel- PPAR-c agonists including pioglitazone and rosiglitazone
lular superoxide levels under control, thereby generating a are approved by the FDA for type 2 diabetes, these agents
feed-forward loop of nitro-oxidative stress and mitochon- may be worth investigating for their therapeutic potential
drial dysfunction.60 CIPN induced by paclitaxel, oxalipla- in CIPN.
tin, and bortezomib is associated with the nitration and Conversely, therapeutic agents targeting peroxyni-
inactivation of MnSOD, accompanied by mitochondrial trite may be an appealing therapeutic approach. Unlike
bioenergetic deficits in peripheral sensory axons.32 ROS, whose existence at normal physiological concentra-
Supporting the pathogenic role of nitro-oxidative tions is necessary for synaptic plasticity and cognitive
stress, experimental strategies targeting ROS and peroxy- function,80 to the best of our knowledge peroxynitrite has
nitrite prevent the development of CIPN in various no known beneficial effects. Peroxynitrite decomposition
rodent models. For example, antioxidants such as acetyl- catalysts have been shown to block the development
L-carnitine and a-lipoic acid are reported to have of neuropathic pain induced by paclitaxel, oxaliplatin, and

4 Cancer Month 00, 2018


Novel Therapeutic Strategies for CIPN/Ma et al

bortezomib in rats, through attenuating chemotherapeutic- produce proinflammatory cytokines that can lower the acti-
induced decreases in mitochondrial ATP production.32,81 vation threshold of spinal and DRG neurons and lead to
The protective effects were exerted without compromising hyperexcitability and spontaneous discharges, thereby con-
the antitumor activity of the chemotherapeutic agents.32,81 tributing to pain.94,95 In response to these inflammatory
stimuli, glial cells (including Schwann cells and satellite
Contribution of Neuroinflammation to CIPN cells in the peripheral nervous system and astrocytes in the
Although the widely accepted concept is that chemothera- spinal cord) undergo phenotypic changes and secrete more
peutics are immunosuppressive, recent studies have shown proinflammatory cytokines and chemokines, further
that they actually can enhance the immune response to enhancing neuronal excitability and pain hypersensitiv-
assist T-cell–mediated killing of tumor cells.82,83 In addi- ity.84,96,97 Blockade of proinflammatory signaling through
tion, increased levels of circulating proinflammatory cyto- interleukin 1 receptor antagonist (IL-1ra) and antitumor
kines have been reported in patients receiving necrosis factor a (TNF-a) have been shown to attenuate
chemotherapy.84 It is well known that local or systemic CIPN symptoms in rodents.96,98 In addition, depleting
inflammation and the production of proinflammatory macrophages through the administration of clodronate, or
cytokines can induce pain. Collectively, these findings blocking macrophage infiltration by the intrathecal admin-
suggest that neuroinflammation (ie, inflammation in the istration of CCL2 or CX3CL1 neutralizing antibody, has
peripheral or central nervous tissues) is one of the major been reported to prevent CIPN in multiple models.92,99
contributors to CIPN. Neuroinflammation is character- Minocycline, a broad-spectrum antibiotic that
ized by infiltration of immune cells, activation of glial inhibits macrophage/microglia activation and proinflam-
cells, and the production of inflammatory mediators in matory cytokine production, prevented oxaliplatin-
the nervous system. It is interesting to note that the effects induced and bortezomib-induced mechanical hyperalgesia
from chemotherapeutic drugs on innate and adaptive in rodents.99,100 A recent pilot clinical study demonstrated
immune responses, along with glial cell activation, have significant reductions in the daily average pain score with
been reported in rodent studies of CIPN.84 the use of minocycline, although it did not fully prevent
In response to chemotherapy-induced damage, intra- paclitaxel-induced neuropathy.101 Further studies are
cellular molecules known as damage-associated molecular needed to examine the usefulness and safety of minocycline
patterns, such as mitochondrial DNA and high-mobility as a preventive intervention for CIPN.
group box 1, are released and activate the receptor for Pharmacological modulation of pathways that con-
advanced glycation end product (RAGE) and toll-like trol neuroinflammation can mitigate chemotherapy-
receptors (TLRs) on neurons and immune cells infiltrating induced neuropathic pain. For example, rodent studies
the DRG, leading to neuroinflammation.85,86 It has been have shown that orally available A3 adenosine receptor
proposed that chemotherapeutics such as paclitaxel may
(A3AR) agonists protect against CIPN. These A3AR ago-
directly activate TLRs. In support of a role for TLRs in
nists also mitigated spinal neuroinflammatory processes,
CIPN, it has been shown that coadministration of a TLR4
including astrocyte activation and increases in IL-1b and
antagonist with paclitaxel prevented the development of
TNF-a, and blocked neuropathic pain caused by pacli-
mechanical allodynia in rats.87 Similarly, genetic ablation
taxel, oxaliplatin, and bortezomib without antagonizing
of either TLR4 or TLR3 is reported to protect mice from
their antitumor effects.97,102 Fingolimod, an FDA-
developing hyperalgesia after treatment with cisplatin.88 It
approved immunomodulatory drug for the treatment of
is interesting to note that, despite previous studies sugges-
multiple sclerosis, also prevented neuroinflammation and
ting that spinal cord TLR4 mediates mechanical hypersen-
ameliorated neuropathic pain induced by paclitaxel and
sitivity in inflammatory and injury-induced neuropathic
oxaliplatin without having a negative impact on their anti-
pain models only in male mice,89 the development of
cancer effects.103 This drug targets the sphingosine-1-
(TLR4-dependent) cisplatin-induced mechanical allodynia
phosphate receptor, thereby antagonizing chemotherapy-
was observed to be present in both male and female
induced activation of nuclear factor jB (NF-jB) and
mice.90 To the best of our knowledge, it remains unclear if
enhanced formation of proinflammatory cytokines.103
such sex dimorphism exists in humans with CIPN.
Chemotherapy-induced infiltration of macrophages Contribution of Neuroimmune Pathways Toward
into the DRG is mediated via the chemoattractants C-C Resolution of CIPN
motif chemokine 2 (CCL2) and C-X3-C motif chemokine In contrast to the pain-promoting effects of proinflamma-
ligand 1 (CX3CL1).87,91-93 Infiltrating macrophages tory cytokines such as TNF-a and IL-1b, evidence has

Cancer Month 00, 2018 5


Review Article

Figure 1. Schematic overview of mechanisms underlying chemotherapy-induced peripheral neuropathy and mechanism-based
disease interventions. CCL2 indicates C-C motif chemokine 2; CX3CL1, C-X3-C motif chemokine ligand 1; HDAC6, histone deacety-
lase 6; IL-10, interleukin 10; IENF, intraepidermal nerve fiber; RNS, reactive nitrogen species; ROS, reactive oxygen species; SNRIs,
serotonin-norepinephrine reuptake inhibitors.

suggested that regulatory T cells (Tregs) and cytokines associated with paclitaxel treatment.96,108 In vitro, IL-10
such as IL-10 are needed to recover from CIPN. For suppressed paclitaxel-induced spontaneous discharges in
example, reduced numbers of Tregs have been reported in DRG neurons.108
models of paclitaxel-induced and oxaliplatin-induced It is interesting to note that we recently found that
pain.104,105 In this regard, boosting the endogenous anti- spontaneous resolution of CIPN depends on endogenous
inflammatory state might serve as another route to pain IL-10 signaling; blocking IL-10 signaling through the
resolution in patients with CIPN. Several studies have intrathecal administration of a neutralizing antibody
demonstrated that boosting the Treg subpopulation delayed recovery from CIPN.108 Consistently, IL-10–
through the intrathecal adoptive transfer of Tregs or the deficient mice did not recover from CIPN. These findings
administration of the Treg-enhancing bee venom-derived indicate that IL-10 signaling is part of the natural resolu-
phospholipase A2 reduces chemotherapy-induced neuro- tion pathway for CIPN, and further underscores the ther-
pathic pain in rodents.104,106 With respect to regulatory apeutic potential of IL-10 therapy to reverse CIPN. One
cytokines, oxaliplatin-induced mechanohypersensitivity is concern is that enhancing IL-10 levels might suppress the
associated with reduced levels of the anti-inflammatory immune response to the tumor; however, evidence has
cytokines IL-10 and IL-4 in the spinal dorsal horn.107 suggested that IL-10 signaling actually may prevent tumor
Conversely, intrathecal IL-10 gene therapy or the intra- formation and contribute toward the antitumor effects of
thecal administration of recombinant IL-10 has been CD81 T cells. Specifically, mice deficient in IL-10 signal-
shown to progressively reverse the allodynic state ing develop tumors spontaneously, and treatment with

6 Cancer Month 00, 2018


Novel Therapeutic Strategies for CIPN/Ma et al

Figure 2. Site of action of chemotherapeutics and mechanism-based treatments for chemotherapy-induced peripheral neuropa-
thy. CCL2 indicates C-C motif chemokine 2; CX3CL1, C-X3-C motif chemokine ligand 1; DRG, dorsal root ganglion; HDAC6, histone
deacetylase 6; IENF, intraepidermal nerve fiber; IL-1b, interleukin-1 beta; IL-10, interleukin 10; SNRI, serotonin-norepinephrine reup-
take inhibitors; TNF-a, tumor necrosis factor a.

recombinant IL-10 stimulates the cytotoxicity of tumor- as pifithrin-l, HDAC6 inhibitors, and metformin, as
resident CD81 T cells and promotes long-lasting antitu- well as antioxidants and peroxynitrite decomposition cata-
mor immunity.109 Therefore, reinforcing IL-10 signaling lysts as prophylactic treatment of CIPN.24,27,30,47,61-68,110
through gene therapy or recombinant IL-10 also may be a It is interesting to note that HDAC6 inhibitors were found
promising approach that could simultaneously stimulate to be effective in both preventing and reversing CIPN,30
antitumor immunity and reverse CIPN. with reported synergistic effects on tumor killing in combi-
nation with chemotherapy.41,42 Given these unique fea-
Conclusions and Future Perspectives tures, HDAC6 inhibition may be a preferred treatment
Clinically, various analgesics have been tested in an option for patients already receiving chemotherapy.
attempt to control neuropathic pain induced by chemo- Conversely, “repurposing” metformin has the advantage of
therapy, with only serotonin-norepinephrine reuptake using an inexpensive drug with a well-known safety profile
inhibitors and Ca21 and Na1 channel blockers demon- to actively prevent CIPN, provided it is administered
strating limited success.10,11,13,15,16 before the first chemotherapy session. Because metformin
Although the exact mechanisms remain elusive, a already is being tested in clinical trials for potential antican-
central role for mitochondrial dysfunction and nitro- cer effects, the rapid evaluation of its preventive effects in
oxidative stress in the pathogenesis of CIPN has been CIPN should be achievable.111 Fingolimod, A3AR
identified (Fig. 1). Preclinical data have demonstrated agonists, and minocycline are candidates for targeting
great promise for compounds targeting this pathway, such the proinflammatory pathways that contribute to

Cancer Month 00, 2018 7


Review Article

CIPN.97,99,100,107 IL-10 gene therapy or recombinant IL- 13. Tsavaris N, Kopterides P, Kosmas C, et al. Gabapentin monotherapy
for the treatment of chemotherapy-induced neuropathic pain: a pilot
10 treatment may represent a more efficacious approach to study. Pain Med. 2008;9:1209-1216.
promote recovery from CIPN in both patients with cancer 14. Rao RD, Michalak JC, Sloan JA, et al; North Central Cancer Treat-
ment Group. Efficacy of gabapentin in the management of
and cancer survivors because it engages natural pain resolu- chemotherapy-induced peripheral neuropathy: a phase 3 randomized,
tion pathways and therefore is likely to have true disease- double-blind, placebo-controlled, crossover trial (N00C3). Cancer.
modifying effects.96,108 2007;110:2110-2118.
15. Saif MW, Syrigos K, Kaley K, Isufi I. Role of pregabalin in treat-
Preclinical studies have identified novel potential ment of oxaliplatin-induced sensory neuropathy. Anticancer Res.
targets for mechanism-based preventive and curative 2010;30:2927-2933.
16. Argyriou AA, Chroni E, Polychronopoulos P, et al. Efficacy of
interventions for CIPN (Fig. 2). Clinical trials currently oxcarbazepine for prophylaxis against cumulative oxaliplatin-induced
are underway to focus not only on symptom relief, but neuropathy. Neurology. 2006;67:2253-2255.
17. Wilson RH, Lehky T, Thomas RR, Quinn MG, Floeter MK, Grem
also on mechanism-based preventive and curative JL. Acute oxaliplatin-induced peripheral nerve hyperexcitability.
interventions. J Clin Oncol. 2002;20:1767-1774.
18. Pareyson D, Piscosquito G, Moroni I, Salsano E, Zeviani M.
Peripheral neuropathy in mitochondrial disorders. Lancet Neurol.
FUNDING SUPPORT 2013;12:1011-1024.
Supported by National Institutes of Health grants R21CA183736, 19. Bennett GJ, Doyle T, Salvemini D. Mitotoxicity in distal symmetrical
R01NS073939, and R01CA208371; Regenacy Pharmaceuticals; sensory peripheral neuropathies. Nat Rev Neurol. 2014;10:326-336.
20. Podratz JL, Knight AM, Ta LE, et al. Cisplatin induced mitochon-
and The University of Texas MD Anderson Cancer Center Knowl-
drial DNA damage in dorsal root ganglion neurons. Neurobiol Dis.
edge Gap funding. 2011;41:661-668.
21. Kidd JF, Pilkington MF, Schell MJ, et al. Paclitaxel affects cytosolic
CONFLICT OF INTEREST DISCLOSURES calcium signals by opening the mitochondrial permeability transition
pore. J Biol Chem. 2002;277:6504-6510.
Annemieke Kavelaars and Cobi J. Heijnen have received partial 22. Tari C, Fournier N, Briand C, Ducet G, Crevat A. Action of vinca
funding from Regenacy Pharmaceuticals for work performed as alkaloides on calcium movements through mitochondrial membrane.
part of the current study and have a patent pending (US App. No. Pharmacol Res Commun. 1986;18:519-528.
15 of 170,335). 23. Joseph EK, Levine JD. Mitochondrial electron transport in models
of neuropathic and inflammatory pain. Pain. 2006;121:105-114.
24. Zheng H, Xiao WH, Bennett GJ. Mitotoxicity and bortezomib-induced
REFERENCES chronic painful peripheral neuropathy. Exp Neurol. 2012;238:225-234.
1. Henley SJ, Singh SD, King J, Wilson RJ, O’Neil ME, Ryerson AB. 25. Areti A, Yerra VG, Naidu V, Kumar A. Oxidative stress and nerve
Invasive cancer incidence and survival-United States, 2013. MMWR damage: role in chemotherapy induced peripheral neuropathy. Redox
Morb Mortal Wkly Rep. 2017;66:69-75. Biol. 2014;2:289-295.
2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J 26. Flatters SJ, Bennett GJ. Studies of peripheral sensory nerves in
Clin. 2017;67:7-30. paclitaxel-induced painful peripheral neuropathy: evidence for mito-
3. Marmiroli P, Scuteri A, Cornblath DR, Cavaletti G. Pain in chondrial dysfunction. Pain. 2006;122:245-257.
chemotherapy-induced peripheral neurotoxicity. J Peripher Nerv Syst. 27. Krukowski K, Nijboer CH, Huo X, Kavelaars A, Heijnen CJ. Pre-
2017;22:156-161. vention of chemotherapy-induced peripheral neuropathy by the
4. Wolf S, Barton D, Kottschade L, Grothey A, Loprinzi C. Chemo- small-molecule inhibitor pifithrin-mu. Pain. 2015;156:2184-2192.
therapy-induced peripheral neuropathy: prevention and treatment 28. Maj MA, Ma J, Krukowski KN, Kavelaars A, Heijnen CJ. Inhibition
strategies. Eur J Cancer. 2008;44:1507-1515. of mitochondrial p53 accumulation by PFT-mu prevents cisplatin-
5. Brewer JR, Morrison G, Dolan ME, Fleming GF. Chemotherapy- induced peripheral neuropathy. Front Mol Neurosci. 2017;10:108.
induced peripheral neuropathy: current status and progress. Gynecol 29. Xiao WH, Bennett GJ. Effects of mitochondrial poisons on the neu-
Oncol. 2016;140:176-183. ropathic pain produced by the chemotherapeutic agents, paclitaxel
6. Fallon MT. Neuropathic pain in cancer. Br J Anaesth. 2013;111: and oxaliplatin. Pain. 2012;153:704-709.
105-111. 30. Krukowski K, Ma J, Golonzhka O, et al. HDAC6 inhibition effec-
7. Seretny M, Currie GL, Sena ES, et al. Incidence, prevalence, and tively reverses chemotherapy-induced peripheral neuropathy. Pain.
predictors of chemotherapy-induced peripheral neuropathy: a system- 2017;158:1126-1137.
atic review and meta-analysis. Pain. 2014;155:2461-2470. 31. Zheng H, Xiao WH, Bennett GJ. Functional deficits in peripheral
8. Miltenburg NC, Boogerd W. Chemotherapy-induced neuropathy: a nerve mitochondria in rats with paclitaxel- and oxaliplatin-evoked
comprehensive survey. Cancer Treat Rev. 2014;40:872-882. painful peripheral neuropathy. Exp Neurol. 2011;232:154-161.
9. Pachman DR, Watson JC, Loprinzi CL. Therapeutic strategies for 32. Janes K, Doyle T, Bryant L, et al. Bioenergetic deficits in peripheral
cancer treatment related peripheral neuropathies. Curr Treat Options nerve sensory axons during chemotherapy-induced neuropathic pain
Oncol. 2014;15:567-580. resulting from peroxynitrite-mediated post-translational nitration of
10. Smith EM, Pang H, Cirrincione C, et al; Alliance for Clinical Trials mitochondrial superoxide dismutase. Pain. 2013;154:2432-2440.
in Oncology. Effect of duloxetine on pain, function, and quality of 33. Pachman DR, Barton DL, Watson JC, Loprinzi CL. Chemotherapy-
life among patients with chemotherapy-induced painful peripheral induced peripheral neuropathy: prevention and treatment. Clin Phar-
neuropathy: a randomized clinical trial. JAMA. 2013;309:1359- macol Ther. 2011;90:377-387.
1367. 34. de Lores Arnaiz GR, Ordieres MG. Brain Na(1), K(1)-ATPase
11. Durand JP, Deplanque G, Montheil V, et al. Efficacy of venlafaxine activity in aging and disease. Int J Biomed Sci. 2014;10:85-102.
for the prevention and relief of oxaliplatin-induced acute neurotoxic- 35. Nasu S, Misawa S, Nakaseko C, et al. Bortezomib-induced neuropa-
ity: results of EFFOX, a randomized, double-blind, placebo-con- thy: axonal membrane depolarization precedes development of neu-
trolled phase III trial. Ann Oncol. 2012;23:200-205. ropathy. Clin Neurophysiol. 2014;125:381-387.
12. Dworkin RH, O’Connor AB, Audette J, et al. Recommendations for 36. Nijboer CH, Heijnen CJ, van der Kooij MA, et al. Targeting the
the pharmacological management of neuropathic pain: an overview p53 pathway to protect the neonatal ischemic brain. Ann Neurol.
and literature update. Mayo Clin Proc. 2010;85(suppl 3):S3-S14. 2011;70:255-264.

8 Cancer Month 00, 2018


Novel Therapeutic Strategies for CIPN/Ma et al

37. Chiu GS, Maj MA, Rizvi S, et al. Pifithrin-mu prevents cisplatin- 59. Beckman JS, Beckman TW, Chen J, Marshall PA, Freeman BA.
induced chemobrain by preserving neuronal mitochondrial function. Apparent hydroxyl radical production by peroxynitrite: implications
Cancer Res. 2017;77:742-752. for endothelial injury from nitric oxide and superoxide. Proc Natl
38. Strom E, Sathe S, Komarov PG, et al. Small-molecule inhibitor of Acad Sci U S A. 1990;87:1620-1624.
p53 binding to mitochondria protects mice from gamma radiation. 60. Macmillan-Crow LA, Cruthirds DL. Invited review: manganese
Nat Chem Biol. 2006;2:474-479. superoxide dismutase in disease. Free Radic Res. 2001;34:325-336.
39. Kaiser M, Kuhnl A, Reins J, et al. Antileukemic activity of the HSP70 61. Flatters SJ, Xiao WH, Bennett GJ. Acetyl-L-carnitine prevents and
inhibitor pifithrin-mu in acute leukemia. Blood Cancer J. 2011;1:e28. reduces paclitaxel-induced painful peripheral neuropathy. Neurosci
40. McKeon AM, Egan A, Chandanshive J, McMahon H, Griffith DM. Lett. 2006;397:219-223.
Novel improved synthesis of HSP70 inhibitor, pifithrin-mu. In vitro 62. Xiao WH, Bennett GJ. Chemotherapy-evoked neuropathic pain:
synergy quantification of pifithrin-mu combined with Pt drugs in abnormal spontaneous discharge in A-fiber and C-fiber primary
prostate and colorectal cancer cells. Molecules. 2016;21(7). pii: E949. afferent neurons and its suppression by acetyl-L-carnitine. Pain.
41. Mishima Y, Santo L, Eda H, et al. Ricolinostat (ACY-1215) induced 2008;135:262-270.
inhibition of aggresome formation accelerates carfilzomib-induced 63. Ghirardi O, Lo Giudice P, Pisano C, et al. Acetyl-L-carnitine pre-
multiple myeloma cell death. Br J Haematol. 2015;169:423-434. vents and reverts experimental chronic neurotoxicity induced by oxa-
42. Santo L, Hideshima T, Kung AL, et al. Preclinical activity, pharma- liplatin, without altering its antitumor properties. Anticancer Res.
codynamic, and pharmacokinetic properties of a selective HDAC6 2005;25:2681-2687.
inhibitor, ACY-1215, in combination with bortezomib in multiple 64. Melli G, Taiana M, Camozzi F, et al. Alpha-lipoic acid prevents
myeloma. Blood. 2012;119:2579-2589. mitochondrial damage and neurotoxicity in experimental chemother-
43. Chen S, Owens GC, Makarenkova H, Edelman DB. HDAC6 regu- apy neuropathy. Exp Neurol. 2008;214:276-284.
lates mitochondrial transport in hippocampal neurons. PLoS One. 65. Kim HK, Zhang YP, Gwak YS, Abdi S. Phenyl N-tert-butylnitrone, a
2010;5:e10848. free radical scavenger, reduces mechanical allodynia in chemotherapy-
44. d’Ydewalle C, Krishnan J, Chiheb DM, et al. HDAC6 inhibitors induced neuropathic pain in rats. Anesthesiology. 2010;112:432-439.
reverse axonal loss in a mouse model of mutant HSPB1-induced 66. Kim HK, Hwang SH, Abdi S. Tempol ameliorates and prevents
Charcot-Marie-Tooth disease. Nat Med. 2011;17:968-974. mechanical hyperalgesia in a rat model of chemotherapy-induced
45. Choi H, Kim HJ, Kim J, et al. Increased acetylation of peroxire- neuropathic pain. Front Pharmacol. 2016;7:532.
doxin1 by HDAC6 inhibition leads to recovery of Ab-induced 67. McCormick B, Lowes DA, Colvin L, Torsney C, Galley HF. Mito-
impaired axonal transport. Mol Neurodegener. 2017;12:23. VitE, a mitochondria-targeted antioxidant, limits paclitaxel-induced
46. Price TJ, Das V, Dussor G. Adenosine monophosphate-activated oxidative stress and mitochondrial damage in vitro, and paclitaxel-
protein kinase (AMPK) activators for the prevention, treatment and induced mechanical hypersensitivity in a rat pain model. Br J
potential reversal of pathological pain. Curr Drug Targets. 2016;17: Anaesth. 2016;117:659-666.
908-920. 68. Galley HF, McCormick B, Wilson KL, Lowes DA, Colvin L,
47. Mao-Ying QL, Kavelaars A, Krukowski K, et al. The anti-diabetic Torsney C. Melatonin limits paclitaxel-induced mitochondrial dys-
drug metformin protects against chemotherapy-induced peripheral function in vitro and protects against paclitaxel-induced neuropathic
neuropathy in a mouse model. PLoS One. 2014;9:e100701. pain in the rat. J Pineal Res. 2017;63(4).
48. Tokubuchi I, Tajiri Y, Iwata S, et al. Beneficial effects of metformin 69. Schloss JM, Colosimo M, Airey C, Masci PP, Linnane AW, Vitetta
on energy metabolism and visceral fat volume through a possible L. Nutraceuticals and chemotherapy induced peripheral neuropathy
mechanism of fatty acid oxidation in human subjects and rats. PLoS (CIPN): a systematic review. Clin Nutr. 2013;32:888-893.
One. 2017;12:e0171293. 70. Sisignano M, Baron R, Scholich K, Geisslinger G. Mechanism-based
49. Fitzgerald JC, Zimprich A, Carvajal Berrio DA, et al. Metformin treatment for chemotherapy-induced peripheral neuropathic pain.
reverses TRAP1 mutation-associated alterations in mitochondrial Nat Rev Neurol. 2014;10:694-707.
function in Parkinson’s disease. Brain. 2017;140:2444-2459. 71. Maestri A, De Pasquale Ceratti A, Cundari S, Zanna C, Cortesi E,
50. Zhou G, Myers R, Li Y, et al. Role of AMP-activated protein kinase Crino L. A pilot study on the effect of acetyl-L-carnitine in pacli-
in mechanism of metformin action. J Clin Invest. 2001;108:1167- taxel- and cisplatin-induced peripheral neuropathy. Tumori. 2005;
1174. 91:135-138.
51. Kasznicki J, Sliwinska A, Drzewoski J. Metformin in cancer preven- 72. Callander N, Markovina S, Eickhoff J, et al. Acetyl-L-carnitine
tion and therapy. Ann Transl Med. 2014;2:57. (ALCAR) for the prevention of chemotherapy-induced peripheral
52. Hirsch HA, Iliopoulos D, Struhl K. Metformin inhibits the inflam- neuropathy in patients with relapsed or refractory multiple myeloma
matory response associated with cellular transformation and cancer treated with bortezomib, doxorubicin and low-dose dexamethasone:
stem cell growth. Proc Natl Acad Sci U S A. 2013;110:972-977. a study from the Wisconsin Oncology Network. Cancer Chemother
53. Cufi S, Corominas-Faja B, Vazquez-Martin A, et al. Metformin- Pharmacol. 2014;74:875-882.
induced preferential killing of breast cancer initiating 73. Hershman DL, Unger JM, Crew KD, et al. Randomized double-
CD441CD24-/low cells is sufficient to overcome primary resistance blind placebo-controlled trial of acetyl-L-carnitine for the prevention
to trastuzumab in HER21 human breast cancer xenografts. Oncotar- of taxane-induced neuropathy in women undergoing adjuvant breast
get. 2012;3:395-398. cancer therapy. J Clin Oncol. 2013;31:2627-2633.
54. Chen G, Xu S, Renko K, Derwahl M. Metformin inhibits growth 74. Guo Y, Jones D, Palmer JL, et al. Oral alpha-lipoic acid to prevent
of thyroid carcinoma cells, suppresses self-renewal of derived cancer chemotherapy-induced peripheral neuropathy: a randomized, double-
stem cells, and potentiates the effect of chemotherapeutic agents. blind, placebo-controlled trial. Support Care Cancer. 2014;22:1223-1231.
J Clin Endocrinol Metab. 2012;97:E510-E520. 75. Polvani S, Tarocchi M, Galli A. PPARg and oxidative stress: con(-
55. Brand MD, Affourtit C, Esteves TC, et al. Mitochondrial superox- beta) catenating NRF2 and FOXO. PPAR Res. 2012;2012:641087.
ide: production, biological effects, and activation of uncoupling pro- 76. Yu X, Shao XG, Sun H, et al. Activation of cerebral peroxisome
teins. Free Radic Biol Med. 2004;37:755-767. proliferator-activated receptors gamma exerts neuroprotection by
56. Low PA, Nickander KK, Tritschler HJ. The roles of oxidative stress inhibiting oxidative stress following pilocarpine-induced status epi-
and antioxidant treatment in experimental diabetic neuropathy. Dia- lepticus.. Brain Res. 2008;1200:146-158.
betes. 1997;46(suppl 2):S38-S42. 77. Chung SS, Kim M, Youn BS, et al. Glutathione peroxidase 3 mediates
57. Di Cesare Mannelli L, Zanardelli M, Failli P, Ghelardini C. Oxali- the antioxidant effect of peroxisome proliferator-activated receptor
platin-induced neuropathy: oxidative stress as pathological mecha- gamma in human skeletal muscle cells. Mol Cell Biol. 2009;29:20-30.
nism. Protective effect of silibinin. J Pain. 2012;13:276-284. 78. Churi SB, Abdel-Aleem OS, Tumber KK, Scuderi-Porter H, Taylor
58. Duggett NA, Griffiths LA, McKenna OE, et al. Oxidative stress in BK. Intrathecal rosiglitazone acts at peroxisome proliferator-activated
the development, maintenance and resolution of paclitaxel-induced receptor-gamma to rapidly inhibit neuropathic pain in rats.. J Pain.
painful neuropathy. Neuroscience. 2016;333:13-26. 2008;9:639-649.

Cancer Month 00, 2018 9


Review Article

79. Lyons DN, Zhang L, Danaher RJ, Miller CS, Westlund KN. 96. Ledeboer A, Jekich BM, Sloane EM, et al. Intrathecal interleukin-10
PPARg agonists attenuate trigeminal neuropathic pain. Clin J Pain. gene therapy attenuates paclitaxel-induced mechanical allodynia and
2017;33:1071-1080. proinflammatory cytokine expression in dorsal root ganglia in rats.
80. Massaad CA, Klann E. Reactive oxygen species in the regulation of Brain Behav Immun. 2007;21:686-698.
synaptic plasticity and memory. Antioxid Redox Signal. 2011;14: 97. Janes K, Wahlman C, Little JW, et al. Spinal neuroimmune activa-
2013-2054. tion is independent of T-cell infiltration and attenuated by A3 aden-
81. Doyle T, Chen Z, Muscoli C, et al. Targeting the overproduction of osine receptor agonists in a model of oxaliplatin-induced peripheral
peroxynitrite for the prevention and reversal of paclitaxel-induced neuropathy. Brain Behav Immun. 2015;44:91-99.
neuropathic pain. J Neurosci. 2012;32:6149-6160. 98. Ale A, Bruna J, Morell M, et al. Treatment with anti-TNF alpha
82. Apetoh L, Ghiringhelli F, Tesniere A, et al. Toll-like receptor 4- protects against the neuropathy induced by the proteasome inhibitor
dependent contribution of the immune system to anticancer chemo- bortezomib in a mouse model. Exp Neurol. 2014;253:165-173.
therapy and radiotherapy. Nat Med. 2007;13:1050-1059. 99. Boyette-Davis J, Dougherty PM. Protection against oxaliplatin-
83. Hato SV, de Vries IJ, Lesterhuis WJ. STATing the importance of induced mechanical hyperalgesia and intraepidermal nerve fiber loss
immune modulation by platinum chemotherapeutics. Oncoimmunol- by minocycline. Exp Neurol. 2011;229:353-357.
ogy. 2012;1:234-236. 100. Robinson CR, Zhang H, Dougherty PM. Astrocytes, but not
84. Lees JG, Makker PG, Tonkin RS, et al. Immune-mediated processes microglia, are activated in oxaliplatin and bortezomib-induced
implicated in chemotherapy-induced peripheral neuropathy. Eur J peripheral neuropathy in the rat. Neuroscience. 2014;274:308-317.
Cancer. 2017;73:22-29. 101. Pachman DR, Dockter T, Zekan PJ, et al. A pilot study of mino-
85. Vichaya EG, Chiu GS, Krukowski K, et al. Mechanisms of cycline for the prevention of paclitaxel-associated neuropathy:
chemotherapy-induced behavioral toxicities. Front Neurosci. 2015;9: ACCRU study RU221408I. Support Care Cancer. 2017;25:3407-
131. 3416.
86. Nishida T, Tsubota M, Kawaishi Y, et al. Involvement of high 102. Chen Z, Janes K, Chen C, et al. Controlling murine and rat
mobility group box 1 in the development and maintenance of chronic pain through A3 adenosine receptor activation. FASEB J.
chemotherapy-induced peripheral neuropathy in rats. Toxicology.
2012;26:1855-1865.
2016;365:48-58.
103. Janes K, Little JW, Li C, et al. The development and maintenance
87. Li Y, Zhang H, Zhang H, Kosturakis AK, Jawad AB, Dougherty
of paclitaxel-induced neuropathic pain require activation of the
PM. Toll-like receptor 4 signaling contributes to paclitaxel-induced
sphingosine 1-phosphate receptor subtype 1. J Biol Chem. 2014;
peripheral neuropathy. J Pain. 2014;15:712-725.
289:21082-21097.
88. Park HJ, Stokes JA, Corr M, Yaksh TL. Toll-like receptor signaling
104. Liu XJ, Zhang Y, Liu T, et al. Nociceptive neurons regulate innate
regulates cisplatin-induced mechanical allodynia in mice. Cancer
and adaptive immunity and neuropathic pain through MyD88
Chemother Pharmacol. 2014;73:25-34.
89. Sorge RE, LaCroix-Fralish ML, Tuttle AH, et al. Spinal cord Toll- adapter. Cell Res. 2014;24:1374-1377.
like receptor 4 mediates inflammatory and neuropathic hypersensitiv- 105. Makker PG, Duffy SS, Lees JG, et al. Characterisation of immune
ity in male but not female mice. J Neurosci. 2011;31:15450-15454. and neuroinflammatory changes associated with chemotherapy-
90. Woller SA, Corr M, Yaksh TL. Differences in cisplatin-induced induced peripheral neuropathy. PLoS One. 2017;12:e0170814.
mechanical allodynia in male and female mice. Eur J Pain. 2015;19: 106. Li D, Kim W, Shin D, Jung Y, Bae H, Kim SK. Preventive effects
1476-1485. of bee venom derived phospholipase A2 on oxaliplatin-induced neu-
91. Zhang H, Boyette-Davis JA, Kosturakis AK, et al. Induction of ropathic pain in mice. Toxins (Basel). 2016;8(1). pii: E27.
monocyte chemoattractant protein-1 (MCP-1) and its receptor 107. Janes K, Esposito E, Doyle T, et al. A3 adenosine receptor agonist
CCR2 in primary sensory neurons contributes to paclitaxel-induced prevents the development of paclitaxel-induced neuropathic pain by
peripheral neuropathy. J Pain. 2013;14:1031-1044. modulating spinal glial-restricted redox-dependent signaling path-
92. Huang ZZ, Li D, Liu CC, et al. CX3CL1-mediated macrophage ways. Pain. 2014;155:2560-2567.
activation contributed to paclitaxel-induced DRG neuronal apoptosis 108. Krukowski K, Eijkelkamp N, Laumet G, et al. CD81 T cells and
and painful peripheral neuropathy. Brain Behav Immun. 2014;40: endogenous IL-10 are required for resolution of chemotherapy-
155-165. induced neuropathic pain. J Neurosci. 2016;36:11074-11083.
93. Old EA, Nadkarni S, Grist J, et al. Monocytes expressing CX3CR1 109. Oft M. IL-10: master switch from tumor-promoting inflammation
orchestrate the development of vincristine-induced pain. J Clin to antitumor immunity. Cancer Immunol Res. 2014;2:194-199.
Invest. 2014;124:2023-2036. 110. Fidanboylu M, Griffiths LA, Flatters SJ. Global inhibition of reac-
94. Sorkin LS, Xiao WH, Wagner R, Myers RR. Tumour necrosis tive oxygen species (ROS) inhibits paclitaxel-induced painful
factor-alpha induces ectopic activity in nociceptive primary afferent peripheral neuropathy. PLoS One. 2011;6:e25212.
fibres. Neuroscience. 1997;81:255-262. 111. Troncone M, Cargnelli SM, Villani LA, et al. Targeting metabo-
95. Ozaktay AC, Kallakuri S, Takebayashi T, et al. Effects of lism and AMP-activated kinase with metformin to sensitize non-
interleukin-1 beta, interleukin-6, and tumor necrosis factor on sensi- small cell lung cancer (NSCLC) to cytotoxic therapy: translational
tivity of dorsal root ganglion and peripheral receptive fields in rats. biology and rationale for current clinical trials. Oncotarget. 2017;8:
Eur Spine J. 2006;15:1529-1537. 57733-57754.

10 Cancer Month 00, 2018

View publication stats

Вам также может понравиться