ArticlePDF AvailableLiterature Review

Emerging Trends in Understanding Chemotherapy-Induced Peripheral Neuropathy

Authors:

Abstract and Figures

Chemotherapy-induced peripheral neuropathy (CIPN) is a major concern in oncology practice given the increasing number of cancer survivors and the lack of effective treatment. The incidence of peripheral neuropathy depends upon the anticancer drug used, but is commonly under-reported in clinical trials. Several animal models have been developed in an attempt to better characterize the pathophysiological mechanisms underlying these CIPN and to find more specific treatments. Over the past two decades, three main trends have emerged from preclinical research on CIPN. There is a compelling body of evidence that neurotoxic anticancer drugs affect the peripheral sensory nerve by directly targeting the mitochondria and producing oxidative stress, by functionally impairing the ion channels and/or by triggering immunological mechanisms through the activation of satellite glial cells. These various neurotoxic events may account for the lack of effective treatment, as neuroprotection may probably only be achieved using a polytherapy that targets all of these mechanisms. The aim of this review is to describe the clinical features of CIPN and to summarize the recent trends in understanding its pathophysiology.
Content may be subject to copyright.
NEUROPATHIC PAIN (E EISENBERG, SECTION EDITOR)
Emerging Trends in Understanding Chemotherapy-Induced
Peripheral Neuropathy
Jérémy Ferrier & Vanessa Pereira & Jérome Busserolles &
Nicolas Authier & David Balayssac
#
Springer Science+Business Media New York 2013
Abstract Chemotherapy-induced peripheral neuropathy
(CIPN) is a major concern in oncology practice given the
increasing number of cancer survivors and the lack of effec-
tive treatment. The incidence of peripheral neuropathy de-
pends upon the anticancer drug used, but is commonly
under-reported in clinical trials. Several animal models have
been developed in an attempt to better characterize the path-
ophysiological mechanisms underlying these CIPN and to
find more specific treatments. Over the past two decades, three
main trends have emerged from preclinical research on CIPN.
There is a compelling body of evidence that neurotoxic anti-
cancer drugs affect the peripheral sensory nerve by directly
targeting the mitochondria and producing oxidative stress, by
functionally impairing the ion channels and/or by triggering
immunological mechanisms through the activation of satellite
glial cells. These various neurotoxic events may account for
the lack of effective treatment, as neuroprotection may prob-
ably only be achieved using a polytherapy that targets all of
these mechanisms. The aim of this review is to describe the
clinical features of CIPN and to summarize the recent trends in
understanding its pathophysiology.
Keywords Anticancer drugs
.
Neurotoxicity
.
Ion channels
.
Mitochondria
.
Oxidative stress
.
Glial cells
.
Neuropathic pain
Introduction
The number of cancer survivors is constantly increasing
in the western world, attesting to the therapeutic prog-
ress of improved cancer management and increased
survival rates. Consequently, taking into account the
long-term consequences of cancer treatments is of major
importance in oncology practice. Chemotherapy-induced
peripheral neuropathy (CIPN) is a major dose-limiting
adverse effect of many anticancer drugs such as plati-
num salts (cisplatin, carboplatin and oxaliplatin), spindle
poisons (taxanes and vinca alkaloids), bortezomib and
thalidomide [1]. Patients experience a combination of
sensory and motor symptoms that can be both painful
and painless, the m ost reported being numbness, loss of
balance, muscle weakness, tingling and burning pain [2].
The incidence, severity and persistence of these neuropa-
thies strongly depend on the anticancer drug involved. Clini-
cal assessment is challenging due to the lack of reliable,
standardized and validated tests that could help the physician
to identify the presence and severity of CIPN [3]. Subjective
methods, such as the Patient Neurotoxicity Questionnaire
(PNQ), have been developed that could help to identify pa-
tients at risk of developing a CIPN or to measure the patients
response to a treatment. A recent study has reported a very
significant difference between the physicians diagnosis of
CIPN, using a standardized neurological examination with
National Cancer InstituteCommon Terminology Criteria
(NCI-CTC) grading, and the patients self-reported intensity
and severity using the PNQ [4]. Hence, CIPN is very likely to
be under-reported by physicians and the severity and func-
tional impact on daily activities are often underestimated
regarding patients subjective experience.
This article is part of the Topical Collection on Neuropathic Pain
J. Ferrier
:
V. Pereira
:
J. Busserolles
:
N. Authier
:
D. Balayssac
Clermont Université, Université dAuvergne,
Pharmacologie fondamentale et clinique de la douleur,
63000 Clermont-Ferrand, France
J. Ferrier
:
V. Pereira
:
J. Busserolles
:
N. Authier
:
D. Balayssac
INSERM, U1107 NEURO-DOL, 63001 Clermont-Ferrand, France
N. Authier
:
D. Balayssac
CHU Clermont-Ferrand, 63000 Clermont-Ferrand, France
D. Balayssac (*)
Laboratory of Toxicology, Faculty of Pharmacy, 28, place Henri
Dunant, BP 38, 63000 Clermont-Ferrand cedex 01, France
e-mail: dbalayssac@chu-clermontferrand.fr
Curr Pain Headache Rep (2013) 17:364
DOI 10.1007/s11916-013-0364-5
Several therapeutic strategies have been tried in order to
prevent or overcome the neurotoxic effects of these com-
pounds, but so far without success. CIPN is currently treated
symptomatically, but no effective neuroprotective agent has
been reported. CIPN is considered to be resistant to most of
first-line treatments for neuropathic pain [5]. Dose reduction or
treatment discontinuation is the only recourse of the oncolo-
gists to limit the apparition of neuropathic symptoms. Howev-
er, these dosage adjustments may strongly affect the prognosis
of cancer remission. In addition, neurotoxicity of anticancer
drugs has an important negative impact on patients quality of
life and especially on daily living activities [6], eventually
leading to emotional distress [2]. Patients with CIPN are also
associated with a great economic burden, with considerably
higher healthcare costs than cancer patients without peripheral
neuropathy [7]. Hence, there is a strong need for improvement
in CIPN management and prevention, which requires a greater
understanding of its particular pathophysiology.
The molecular and cellular mechanisms leading to CIPN
have been extensively studied over the past two decades.
Animal models that mimic the clinical neuropathic symptoms
have been developed in an attempt to understand the integrat-
ed mechanisms of neurotoxicity and the molecular events
leading to CIPN [8]. However, the pathophysiology of these
neuropathies is still a matter of debate. Three main mecha-
nisms have emerged from preclinical studies: (1) mitotoxicity
and oxidative stress, (2) ion channel involvement, and (3)
inflammatory processes through the activation of glial
cells. This article will review and discuss the latest
findings on CIPN research and highlight the recent
advances in its therapeutic treatment.
Clinical Features
Platinum Salts
Cisplatin is used as a first or second line of treatment against a
wide range of solid tumors (lung, ovary, bladder , testicular,
head and neck, esophagus, stomach, colon, pancreas, melano-
ma, breast, prostate, mesothelioma, leiomyosarcoma and glio-
ma) [9]. Cisplatin-induced neuropathy is dependent on the
cumulative dose and generally appears after 400700 mg/m
2
[10]. About 28 % of patien ts develop symptomatic neuropa-
thies, of which 6 % suffer from incapacitating polyneuropathies
[11]. This CIPN is primarily sensory-based, with paresthesias,
sensory ataxia, loss of vibratory sensitivity and a decrease or
loss of tendon reflexes [10]. Proprioceptive alterations, muscu-
lar cramps and Lhermittes sign are described in severe cases of
neuropathy [12]. These symptoms are usually reversible after
discontinuation of treatment, but recovery is often very slow.
Oxaliplatin is widely used for the treatment of advanced
colorectal cancer. Neurotoxicity is the dose-limiting adverse
effect, with two components: an acute nerve hyperexcitability
and a chronic cumulative peripheral neuropathy. In more than
90 % of treated patients, oxaliplatin is responsible for sensory
symptoms including cold-induced dysesthesias in the hands
and the circumoral area, numbness and tingling in the extrem-
ities, muscle weakness and neuropathic pain [13, 14]. Muscle
spasms or cramps are often reported, sometimes described as
stiffness in the hands and feet or an inability to release the grip.
These symptoms are more likely to develop shortly after
chemotherapy (within hours or a couple of days of comple-
tion) and are usually self-limited, resolving in a few days.
However, they usually reoccur following subsequent admin-
istration and increase in both duration and severity. In addi-
tion, pharyngolaryngospasm syndrome, distinct from cold-
induced pharyngolaryngeal dysesthesias, can appear in less
than 1 % of patients within hours of oxaliplatin infusion and
without any sign of respiratory distress [15, 16].
W ith the repetition of chemotherapy cycles, 50 to 70 % of
patients develop a persistent peripheral neuropathy, manifesting
as a symmetric, distal, primarily sensory polyneuropathy char -
acterized by the persistence of paresthesias between the che-
motherapy cycles, numbness in the hands and feet and neuro-
pathic pain. In the most severe cases, patients may suffer from
sensory ataxia as well as a deep and superficial sensory loss,
eventually leading to functional impairment [17]. The severity
of this CIPN depends on both the cumulated dose received and
its intensity. The median time to onset of grade 3 neurotoxicity
is at the 10th cycle, namely a cumulative dose of 874 mg/m
2
[18]. Improvement of the neurological symptoms is observed
after discontinuation of treatment, with a median time to recov-
eryof13weeks[18]. However, a recent study has shown the
persistence of neuropathic symptoms 29 months after the last
chemotherapy cycle for almost 80 % of treated patients, seri-
ously challenging the reversibility of this neuropathy [19].
Moreover, peripheral neuropathy may develop and worsen
several months after cessation of chemotherapy [20]. This
phenomenon, known as coasting, is shared by all platinum
drugs and highlights the import ance of finding early clinical
markers of chronic neuropathy. Interestingly, the intensity of
acute thermal hypersensitivity (especially cold allodynia) is a
relevant clinical marker of early oxaliplatin neurotoxicity and
may predict an increased risk of developing chronic and
severe CIPN [21].
Spindle Poisons
Vinca Alkaloids
Vincristine is a major anticancer drug in hematology and
pediatrics (sarcoma) but represents the most neurotoxic among
the Vinca alkaloids [22]. Around 50 % of patients experience
sensory-motor peripheral neuropathies [23]. Vincristine neuro-
toxicity is cumulative and dose-dependent [24]. Symptoms
364, Page 2 of 9 Curr Pain Headache Rep (2013) 17:364
appear after a cumulative dose of 12 mg while chemotherapy
should be stopped after a cumulative dose of 3050 mg [25,
26]. Neuropathy includes numbness and tingling of the hands
and feet with paresthesias and dysesthesias, and a loss of deep
tendon reflexes. The most severe cases can be associated with
distal muscle weakness. Autonomic disorders can also be
found in more than a third of patients [24]. More rarely,
patients develop eye movement disturbances and paralysis of
the vocal cords [24]. After treatment discontinuation, the re-
versal of neurological symptoms is usually slow, taking several
months [25].
Taxanes
Paclitaxel is approved for the treatment of various tumors
(ovary, breast, head and neck, and lung) [27]. It is responsible
for sensory neuropathy which may begin as early as 24
72 hours after the administration of a single high dose and
affects 5978 % of patients [23, 28]. The neuropathy is
dependent on the cumulative dose (>1,400 mg/m
2
), the dose
magnitude during each cycle (>200 mg/m
2
)andtheperfusion
duration (short duration) [10, 29]. This CIPN is associated
with paresthesia, numbness, tingling and burning, and me-
chanical and cold allodynia. Patients describe the symptoms
as a stocking-and-glove distribution that affects the feet and
the toes [28]. Perioral numbness has also been reported [30].
Loss of tendon reflexes, vibration sensation and propriocep-
tion can also be observed [28]. More rarely, motor symptoms
such as mild distal weakness with myalgia are found, affecting
toeextensormuscles[31, 32].
As with paclitaxel, docetaxel is mainly used for the treat-
ment of solid tumors (breast, lung, stomach and androgen-
independent prostate cancer) [33]. Docetaxel-induced neurop-
athy is mainly sensory and correlated to cumulative
dose. Severe neuropathies may appear after a cumula-
tive dose of 600 mg/m
2
[34] and can be associated with motor
impairment [35]. However, compared to paclitaxel, docetaxel-
induced neuropathy is less frequent (19 %; grade 3/4) [36],
with only mild sensory symptoms that reverse spontaneously
after treatment discontinuation [34].
Thalidomide
Thalidomide is used in the treatment of multiple mye-
loma [37]. About 40 % of patients experience neuropa-
thy and this proportion increases to 100 % after
7 months of thalidomide therapy [38, 39]. This CIPN
is characterized by sensory neuropathy associated with
paresthesia, tingling, dysesthesia and a slight loss of
tactile sensation at the extremities of the limbs [39].
The sev erity of the neuropathy is pr obably dose-
dependen t but definitive correlation with doses or treatment
durations has yet to be clarified [40, 41].
Bortezomib
Bortezomib is also approved for the treatment of multiple
myeloma [42]. Neurotoxicity, generally occurring within the
first chemotherapy courses, is one of the most non-
hematologic al and dose-limiting toxicities of bortezomib [22].
Pain is the most prominent symptom in neuropathic patients
[43] and is reported in approximately 50 % of previously
untreated patients and 81 % of previously treated patients
[44]. In the majority of cases, the neuropathy is reversible [45].
Pathophysiology: Cellular and Subcellular Targets
In the past two decades, the development of in vitro and
in vivo models has provided valuable tools for the study of
the pathogenesis of CIPN [8]. Nowadays, it is well known that
one anticancer drug may act on various subcellular targets of
peripheral sensory nerves, and that one mechanism of neuro-
toxicity may be shared by several chemotherapeutic agents,
independent of their antitumor properties. Hence, there is still
no consensus on the molecular mechanisms leading to the
development of CIPN. The main neurotoxic mechanisms of
anticancer drugs identified so far are summarized in Fig. 1.
Toxicokinetics of Anticancer Drugs
Recent work has suggested a specific relationship between the
toxicokinetics of anticancer drugs and their neurotoxicity,
suggesting that several membrane transporters could contrib-
ute to the uptake of the anticancer drugs by dorsal root ganglia
(DRG) and nerves [46, 47, 48]. Platinum salts are substrates
for ATP-Binding Cassette (ABC) proteins (ABCC1, 2 and 4),
solute carrier (SLC) proteins (SLC22A, 31A and 47A) and
ATPase membrane proteins (ATP7A and 7B), which means
that these drug carriers can influence the influx or efflux of
platinum salts across cell membranes, and consequently their
cellular uptake [48]. For example, copper transporters may be
associated with the neurotoxicity of platinum drugs in rats [49,
50]. Ctr1 and ATP7A transporters were overexpressed in rat
DRG with a neu ron specific pa ttern; large-sized neurons
expressed rCtr1 and medium- or small-sized neurons expressed
ATP 7A [ 49, 50]. What
s more, in treated rats, equitoxic doses
of oxaliplatin, cisplatin or carboplatin caused a selective toxic-
ity in rCtr1-expressing DRG neurons, but not in ATP7A ex-
pressing neurons [49]. The organic cation/carnitine transporters
rOctn1 (SLC22A4) and rOctn2 (SLC22A5) can mediate
oxaliplatin neurotoxicity and are strongly expressed in rat
DRG [51]. Inversely, low-level ABCC2 (Multidrug Resistance
Protein 2 [MRP2]) gene expressionwasfoundinDRGcom-
pared to the brain and spinal cord of the rat, which could
facilitate peripheral neurotoxicity with cisplatin by decreasing
the platinum salt efflux out of cells [47]. The same observation
Curr Pain Headache Rep (2013) 17:364 Page 3 of 9, 364
hasbeenmadeforVinca alkaloids and taxanes, which are
substrates of ABCB1 (P-glycoprotein [P-gp]). In the rats,
ABCB1 genes were less expressed in the DRG compared to
the brain and spinal cord, and the activity of the efflux proteins
(measured by the incorporation of a radioactive substrate,
99m
Tc-sestamibi) was lower in the peripheral n ervous system
(PNS) (DRG and sciatic nerve) compared to the central nervous
system (CNS) (brain and spinal cord). These results suggest
that the PNS would be less protected by the blood nerve barrier
than the CNS would be with the blood brain barrier, explaining
the susceptibility of the PNS to neurotoxic anticanc er drugs [46,
47]. In agreement with this suggestion, DRG concentrations of
platinum salts or paclitaxel are close to those achievable in
tumor tissue, while much lower concentrations can be
detected in the CNS [5254]. However, more experiments
(histology, pharmacology and toxicology) will be required to
advance our understanding of these cellular transportation
mechanisms.
Platinum Salts
Oxaliplatin-induced acute neurotoxicity has been described as
a channelopathy, involving voltage-gated sodium and potassi-
um channels. Oxaliplatin would shift the voltage dependence
of both Na
+
and K
+
channels toward more negative voltages
(the channels become activated by lower positive charges),
thus leading to a transient nerve hyperexcitability [5559]. As
first hypothesized by Grolleau et al. (2001), oxaliplatin-
associated acute neurologic disorders have long been attributed
to oxalate, an oxaliplatin metabolite, which could induce a
transient and non-functional disruption of voltage-gated ion
channels by chelating intracellular calcium ions in neurons
[57]. However, recent evidence questions this hypothesis. First-
ly, intracellular calcium concentrations have been shown to be
unaffected following acute exposure to oxaliplatin [60]. Sec-
ondly, the concentrations of oxaliplatin used in these studies
were sometimes very high (up to 500 μM), while it has been
shown that the max imum plasmatic concentration of
oxaliplatin after a dose of 85 mg/m
2
infused over 2 hours
(FOLFOX regimen) is about 5 μM[61]. Oxalate is known to
be responsible for renal and neurological damage caused by the
formation of calcium oxalate crystals, as in ethylene glycol
poisoning [62]. It is very likely that an oxalate concentration
sufficiently high to influence the voltage dependency of sodi-
um channels would also cause a strong renal toxicity associated
with hypocalcaemia, which is rarely observed in patients re-
ceiving oxaliplatin [15]. Similarly, although ethylene glycol
toxicity is associated with neuromuscular symptoms that re-
semble those induced by oxaliplatin, cold-induced dysesthesia
has never been documented in any case of ethylene glycol
intoxication [62]. Recent evidence suggests that oxaliplatin
has a specific effect on voltage-gated channels [63], although
the exact mechanism of action remains unknown. Dimitrov
and Dimitrova (2011) recently provided a new potential mech-
anism for oxaliplatin-induced acute nerve hyperexcitability that
involves the impairment of fast potassium channel functioning
in myelinated axon internodes, forming internodal sources of
after-discharges in response to a saltatory action potential [64].
Fig. 1 Schematic representation
of the main cellular and
subcellular actors involved in the
pathogenesis of CIPN at the
dorsal root ganglia level.
Although neurotoxic anticancer
drugs display different
pharmacological mechanisms,
several neurotoxic mechanisms
(i.e., neuronal targets) are shared
by various chemotherapeutic
agents from different classes
364, Page 4 of 9 Curr Pain Headache Rep (2013) 17:364
Oxaliplatin-induced chronic neuropathy resembles that of
the other platinum compounds, cisplatin and carboplatin. In
the same way, animal studies have demonstrated similar mor-
phological changes in nervous tissues [65]. Cisplatin was
responsible of nucleolar alterations (rather than nuclear ones)
and a disorganization of ribosomes, with a shrinkage of the
Nissl substance and an increase in neurofilaments. The somat-
ic, nuclear and nucleolar size of DRG neurons showed a
significant and dose dependent cellular atrophy without obvi-
ous neuronal loss [66]. In contrast, satellite cells were less
altered than neurons [66]. Pathological changes in the periph-
eral nerves were very mild in comparison to neuron body cells
[67]. Platinum salts have a strong affinity for the DNA of
DRG cells and the neurotoxicity is believed to result from the
effect of their alkylating properties on the DNA [53, 68, 69].
When plat inum adducts exceed the DNA-repair c apacity,
neurons undergo cell death through apoptotic mechanisms.
More precisely, cisplatin is able to induce cell cycle re-entry in
postmitotic neurons, thus triggering apoptosis through cell
cycle checkpoint signaling [70]. Recently, the formation of
platinum adducts on mitochondrial DNA following cisplatin
exposure has been demonstrated in vitro and in vivo. Mito-
chondrial toxicity could also represent an important patho-
physiological basis for platinum salt neurotoxicity [71], as
well as satellite glial cell activation in DRG [72].
Several studies in animal models of CIPN have pointed out
the involvement of ion channels in the pathogenesis of neu-
ropathic pain. In particular, transient receptor potential (TRP)
channels have been extensively studied for their role in tem-
perature perception and mechanosensation, two commonly
altered features in patients with CIPN. Oxaliplatin-induced
acute and chronic sensory disorders have been associated with
sensitization of TRP V1 and TRPA1 in cultured rat DRG
neurons, potentially accounting for hot and cold hypersensi-
tivity, respectively [73]. TRPM8 was found to be up-regulated
in mice DRG at day 3 after a single oxaliplatin administration,
while capsazepine (a non-selective TRPM8 channel blocker)
significantly decreased cold allodynia [74]. More recently,
Descoeur et al.(2011)demonstratedanimpairedexpression
profile of several ion channels in mice DRG following a single
oxaliplatin injection: a decreased e xpression of TREK-1,
TRAAK and Kv1.1 and an up-regulation of Nav1.8, TRPA1
and HNC1 (hyperpolarization-activated cyclic nucleotide-
gated 1) mRNA. Ivabradine, a non-selective HCN inhibitor,
succes sfully reduced oxaliplatin-evoked co ld a llodynia in
mice [75]. Oxaliplatin-induced acute and chronic sensory
disorders have been associated with sensitization of TRPV1
and TRPA1 in cultured rat DRG neurons, potentially account-
ing for hot and cold hypersensitivity, respectively [73]. In
mice lacking TRPA1, Nassini et al. (2011) demonstrated that
oxaliplatin- and cisplatin-induced mechanical allodynia
were absent and reduced, respectively [76]. Additionally,
cisplatin-induced mechanical hypersensitivity has been
associated with an increased expression of TRPV2, P2X3
and ASIC3 channels in DRG neurons [77].
Spindle Poisons
The mechanism of action of taxanes and Vinca alkaloids
involves, respectively, excessive stabilization and inhibition
of mitotic spindle microtubule formation. Although DRG
cells are non-proliferative, differentiated neurons, microtu-
bules are essential for the transport of proteins from the cell
body into and down the length of the axon. By impairing
microtubule formation, the spindle poisons disrupt the axo-
plasmic transport which eventually leads to neuronal death.
Longer axons are more vulnerable to axonal transport disrup-
tion which explains the stocking-and glove pattern of the
resulting neuropathy, affecting primarily the lower limbs.
Nerve sections from paclitaxel or docetaxel treated animals
showed an axonopathy with axonal degeneration and col-
lapsed or fragmented myelin sheaths, when examined by light
microscopy [78]. Electron microscopic examination of pe-
ripheral nerves showed primary axonal degeneration, but also
the presence of Schwann cells with condensed chromatin and
nucleolus-like formations or enlarged cytoplasmic organ-
elles [78
]. In skin biopsies of rat foot pads, intra-epidermal
nerve fiber density was decreased and correlated with the
neurophysiological assessment [79]. Paclitaxel induces mito-
chondrial abnormalities in peripheral nerves by impairing
Complex I and Complex IImediated respiration and ATP
production [80], manifesting as an increase incidence of swol-
len and vacuolated mitochondria in sensory axons [81]but
without affecting the motor nerves [82]. This mitochondrial
impairment results in the production of reactive oxygen spe-
cies (ROS) reinforced by a reduction of the antioxidant de-
fenses in peripheral neurons, suggesting a strong involvement
of oxidative stress in paclitaxel-induced neuropathy [83]. The
pharmacological inhibition of ROS by a non-specific scaven-
ger prevented mechanical hypersensitivity and attenuated
established paclitaxel-induced pain [84]. The involvement of
glial cells in the pathogenesis of paclitaxel-induced neuropa-
thy has also been demonstrated. In rodents, paclitaxel treat-
ment induces an activation of satellite glial cells in the DRG
[72] and spinal cord [80], and a loss of intraepidermal nerve
fibers associated with the activation of Langerhans cells [85].
An accumulation of macrophages within the DRG has also
been described following paclitaxel treatment in rats [86]. A
marked reduction of DRG nerve blood supply by a direct
toxicity on the endothelial cells of the vasa nervorum has also
been demonstrated in animals [87]. Finally, a role for TRP
receptors in the pathogenesis of paclitaxel-associated pain
symptoms has been shown in animals. Paclitaxel-induced pain
hypersensitivity was strongly reduced by selective antagonists
of TRPV1, TRPV4 and TRPA1 (depending on stimulus mo-
dality) [88]aswellasinmicelackingTRPV4[89].
Curr Pain Headache Rep (2013) 17:364 Page 5 of 9, 364
The pathophysiology of Vinca alkaloids induced peripheral
neuropathy remains poorly understood. In animals, vincristine
induces an axonopathy with Wallerian-like degeneration of
myelinated and unmyelinated fibers correlated with neuropath-
ic pain symptoms [65]. In a rat model, this CIPN has been
associated with an increase of calcium levels and oxidative
stress in peripheral nerves, probably mediated by a mitochon-
drialimpairment[90]. An important role of inflammatory me-
diators has also been linked to vincristine-indu ced neuropath y.
An increased number of Langerhans cells and a loss of intra-
epidermal nerve fibers have also been observed in the skin of
vincristine-treated animals [85], as well as an increased number
of macrophages in the DRG [91]. TRPV4 has also been found
to be involved in the develop ment of sensory disorders follow-
ing vincristine treatment [89]. Finally, abnormal serotonin neu-
rotransmission may be involved in the development of neuro-
pathic pain following vincristine administration, as evidenced
by an increased expression of 5HT2A receptors in the superfi-
cial layers of the dorsal horn spinal cord and in the small- and
medium-sized DRG cells [92]. Mice lacking the serotonin
transporter 5HTT are also less sensitive to vincristine-induced
neuropathic pain but not to neuroto xicity [91].
Bortezomib
Bortezomib-induced neurotoxicity has been correlated with
morphological changes in DRG, affecting predominantly the
Schwann and satellite cells [93]. Mitochondrial toxicity and
endoplasmic reticulum stress represent the two main re-
sponses in Schwann cells following bortezomib treatment,
leading to pathological adaptive responses such as demyelin-
ation and macrophage recruitment [94]. Inhibition of tran-
scription, transport and cytoplasmic translation of mRNAs in
DRG neurons, caused by the accumulation of ubiquitin-
conjugated proteins, has also been reported [95]. Recent find-
ings suggest that bortezomib neurotoxicity may also be inde-
pendent from its proteasome inhibition properties [96]. This
CIPN may be caused by an indirect increase in microtubules
stabilization due to an increased expression of microtubule-
associated proteins [97]
Thalidomide
The toxicodynamic effects of thalidomide on per ipheral
nerves remains poorly elucidated. Neurotoxicity may result
from the inhibition of NF-κB leading to a dysregulation of
neurotrophin sensitivity and to impaired nerve growth factor-
mediated neuron survival [98]. However, this hypothesis is yet
to be demonstrated. Interestingly, thalidomide has been shown
to induce microvascular damage at the vasa nervorum level
due to its antiangiogenic properties. Subsequent decreased
nerve blood flow in DRG could represent a core pathological
mechanism of neurotoxicity [87]. Further studies are needed
to understand the pathogenesis of thalidomide-associated neu-
ropathy, especially in animals.
Therapeutic Strategies
Two therapeutic strategies have been proposed to manage
CIPN, with symptomatic and preventive treatments [99]. The
current pharmacotherapies used to treat CIPN involve tricyclic
antidepressants (amitriptyline, nortriptyline) and anticonvul-
sants (gabapentin, valproic acid, lamotrigine). However these
drugs have limited to no efficacy in the treatment of CIPN and
are associated with important adverse drug ef fects that limit
their chronic use [99]. A recent and robust clinical trial has
demonstrated the efficacy of duloxetine (5 weeks of treatment)
in reducing pain symptoms in patients suffering from
paclitaxel- or oxaliplatin-induced neuropathies compared to
placebo [100••]. This is the first phase III study demonstrating
a beneficial effect in the treatment of CIPN. However , although
duloxetine has been proven to be superior to placebo with
regards to pain relief, its efficacy remains relative (59 % of
patients reported a pain reduction versus 38 % in the placebo
group). New therapeutic opportunities have emerged for the
treatment of CIPN with the use of cannabinoids, which must be
assessed in large scale trials [101].
The use of neuroprotectants, according to the profile of
anticancer drug cytotoxicity (e.g., amifostine/platinum salts
and neurotrophic factors/taxanes), has also revealed a very
weak efficacy in limiting or preventing CIPN [102]. In this
context, the nutraceuticals (e.g., vitamin E, vitamin B6, mag-
nesium, calcium, acetyl-L-carnitine, glutamine, glutathione,
n-acetyl cysteine, omega-3 fatty acids, alpha lipoic acid), as
alternative strategies to pharmacotherapy, have given promis-
ing early results in the treatment or the prevention of CIPN
(for review see [103]).
Overall, the clinical assessment of therapeutic strategies
suffers from the lack of harmonization among outcome mea-
sures (qualitative or quantitative) and the standardization of
CIPN assessment procedures (objective v ersus subjective
evaluation) [103]. Consequently, for a validated evidenced
based medicinal treatment, therapeutic strategies for the man-
agement of CIPN must be assessed in large scale randomized
controlled trials.
Conclusion
As highlighted in this review, CIPN still represents a real
problem for the therapeutic management of cancer patients
as it compromises both the oncological prognosis and the
patients quality of life. The number of affected patients each
year is probably very high. In clinical trials, CIPN is often
under-rated and under-reported by clinicians due to the lack of
364, Page 6 of 9 Curr Pain Headache Rep (2013) 17:364
consensus about its evaluation. A lot of effort has been made
to characterize the cellular and subcellular mechanisms un-
derlying the neurotoxicity of anticancer drugs. Actual knowl-
edge suggests that neurotoxicity result from several molecular
events in peripheral nerves that may account for the failure of
investigated drugs to prevent CIPN. Indeed, it is highly likely
that only a combination of several treatments that target the
highlighted mechanisms will be successful in providing
neuroprotection.
Compliance with Ethics Guidelines
Conflict of Interest Dr. rémy Ferrier reported no potential conflicts
of interest relevant to this article.
Dr. Vanessa Pereira reported no potential conflicts of interest relevant
to this article.
Dr. Jérome Busserolles reported no potential conflicts of interest
relevant to this article.
Dr. Nicolas Authier reported no potential conflicts of interest relevant
to this article.
Dr . David Balayssac reported receiving a grant from Ligue contre le
cancer.
Human and Animal Rights and Informed Consent This article does
not contain any studies with human or animal subjects performed by any
of the authors.
References
Papers of particular interest, published recently, have been
highlighted as:
Of importance
•• Of major importance
1. Balayssac D, Ferrier J, Descoeur J, et al. Chemotherapy-induced
peripheral neuropathies: from clinical relevance to preclinical evi-
dence. Expert Opin Drug Saf. 2011;10(3):40717.
2. Tofthagen C. Patient perceptions associated with chemotherapy-
induced peripheral neuropathy. Clin J Oncol Nurs. 2010;14(3):E228.
3. Cleeland CS, Farrar JT, Hausheer FH. Assessment of cancer-related
neuropathy and neuropathic pain. Oncologist. 2010;15 Suppl 2:138.
4. Shimozuma K, Ohashi Y, Takeuchi A, et al. Feasibility and validity
of the Patient Neurotoxicity Questionnaire during taxane chemo-
therapy in a phase III randomized trial in patients with breast cancer:
N-SAS BC 02. Support Care Cancer . 2009;17(12):148391.
5. Dworkin RH, O'Connor AB, Audette J, et al. Recommendations for
the pharmacological management of neuropathic pain: an overview
and literature update. Mayo Clin Proc. 2010;85(3 Suppl):S3S14.
6. Driessen CM, de Kleine-Bolt KM, Vingerhoets AJ, et al. Assessing
the impact of chemotherapy-induced peripheral neurotoxicity on the
quality of life of cancer patients: the introduction of a new measure.
Support Care Cancer. 2011;20(4):87781.
7. Pike CT, Birnbaum HG, Muehlenbein CE, et al. Healthcare costs and
workloss burden of patients with chemotherapy-associated peripheral
neuropathy in breast, ovarian, head and neck, and nonsmall cell lung
cancer. Chemother Res Pract. 2012;2012:913848.
8. Authier N, B alayssac D, Marchand F, et al. Animal models
of chemotherapy-evoked painful peripheral neuropathies.
Neurotherapeutics. 2009;6(4 ):6209.
9. Boulikas T, Vougiouka M. Recent clinical trials using cisplatin,
carboplatin and their combination chemotherapy drugs (review).
Oncol Rep. 2004;11(3):55995.
10. Chaudhry V, Chaudhry M, Crawford TO, et al. Toxic neu-
ropathy in patients with pre-existing neuropathy. Neurology.
2003;60(2): 33740.
11. Strumberg D, Brugge S, Korn MW, et al. Evaluation of long-
term toxi city in pa tients after cispl atin-base d chemotherapy
for non-seminomatous testicular cancer . Ann Oncol. 2002;13(2):229
36.
12. Siegal T, Haim N. Cisplatin-induced peripheral neuropathy. Fre-
quent off-therapy deterioration, demyelinating syndromes, and
muscle cramps. Cancer. 1990;66(6):111723.
13. Raymond E, Chaney SG, Taamma A, Cvitkovic E. Oxaliplatin:
a rev iew of p reclinical and clini cal studies. Ann Onc ol.
1998;9(10):105371.
14. Raymond E, Faivre S, Woynarowski JM, Chaney SG. Oxaliplatin:
mechanism of action and antineoplastic activity. Semin Oncol.
1998;25(2 Suppl 5):412.
15. Haller DG. Safety of oxaliplatin in the treatment of colorectal
cancer. Oncology (Williston Park). 2000;14(12 Suppl 11):1520.
16. de Gramont A, Figer A, Seymour M, et al. Leucovorin and fluoro-
uracil with or without oxaliplatin as first-line treatment in advanced
colorectal cancer. J Clin Oncol. 2000;18(16):293847.
17. Tofthagen C, McAllister RD, McMillan SC. Peripheral neuropathy
in patients with colorectal cancer receiving oxaliplatin. Clin J Oncol
Nurs. 2011;15(2):1828.
18. Grothey A. Oxaliplatin-safety profile: neurotoxicity. Semin Oncol.
2003;30(4 Suppl 15):513.
19. Park SB, Lin CS, Krishnan AV, et al. Long-term neuropathy after
oxaliplatin treatment: challenging the dictum of reversibility. On-
cologist. 2011;16(5):70816.
20. Tofthagen C. Surviving chemotherapy for colon cancer and living
with the consequences. J Palliat Med. 2010;13(11):138991.
21. Attal N, Bouhassira D, Gautron M, et al. Thermal hyperalgesia as a
marker of oxaliplatin neurotoxicity: a prospective quantified senso-
ry assessment study. Pain. 2009;144(3):24552.
22. Windebank AJ, Grisold W. Chemotherapy-induced neuropathy. J
Peripher Nerv Syst. 2008;13(1):2746.
23. Wilkes G. Peripheral neuropathy related to chemotherapy. Semin
Oncol Nurs. 2007;23(3):16273.
24. Quasthoff S, Hartung HP. Chemotherapy-induced peripheral neu-
ropathy. J Neurol. 2002;249(1):917.
25. Legha SS. Vincristine neurotoxicity. Pathophysiology and manage-
ment. Med Toxicol. 1986;1(6):4217.
26. Verstappen CC, Koeppen S, Heimans JJ, et al. Dose-related
vincristine-induced peripheral neuropathy with unexpected off-
therapy worsening. Neurology. 2005;64(6):10767.
27. Mekhail TM, Markman M. Paclitaxel in cancer therapy. Expert
Opin Pharmacother. 2002;3(6):75566.
28. Rowinsky EK, Chaudhry V, Cornblath DR, Donehower RC. Neu-
rotoxicity of taxol. J Natl Cancer Inst Monogr. 1993;15:10715.
29. Smith RE, Brown AM, Mamounas EP, et al. Randomized trial of 3-
hour versus 24-hour infusion of high-dose paclitaxel in patients with
metastatic or locally advanced breast cancer: National Surgical
Adjuvant Breast and Bowel Project Protocol B-26. J Clin Oncol.
1999;17(11):340311.
30. Wiernik PH, Schwartz EL, Strauman JJ, et al. Phase I clinical and
pharmacokinetic study of taxol. Cancer Res. 1987;47(9):248693.
31. Berger T, Malayeri R, Doppelbauer A, et al. Neurological monitor-
ing of neurotoxicity induced by paclitaxel/cisplatin chemotherapy.
Eur J Cancer. 1997;33(9):13939.
32. Chaudhry V, Rowins ky EK, Sartorius SE, et al. Peripheral
neuropathy from taxol a nd ci splatin combination chemother-
apy: clinical and electrophysiological studies. Ann Neurol.
1994;35(3): 30411.
Curr Pain Headache Rep (2013) 17:364 Page 7 of 9, 364
33. Engels FK, Verweij J. Docetaxel administration schedule: from
fever to tears? a review of randomised studies. Eur J Cancer.
2005;41(8):111726.
34. Hilkens PH, Verweij J, Stoter G, et al. Peripheral neurotoxicity
induced by docetaxel. Neurology. 1996;46(1):1048.
35. Freilich RJ, Balmaceda C, Seidman AD, et al. Motor neuropathy
due to docetaxel and paclitaxel. Neurology. 1996;47(1):1158.
36. Lee JJ, Swain SM. Peripheral neuropathy induced by microtubule-
stabilizing agents. J Clin Oncol. 2006;24(10):163342.
37. Teo SK, Stirling DI, Zeldis JB. Thalidomide as a novel therapeutic
agent: new uses for an old product. Drug Discov Today.
2005;10(2):10714.
38. Corso A, Zappasodi P, Barbarano L, et al. Long-term outcome in
relapsed and refractory multiple myeloma treated with thalidomide.
Balancing efficacy and side-effects. Leuk Res. 2009;33(9):e1459.
39. Plasmati R, Pastorelli F, Cavo M, et al. Neuropathy in multiple
myeloma treated with thalidomide: a prospective study. Neurology.
2007;69(6):57381.
40. Kocer B, Sucak G, Kuruoglu R, et al. Clinical and electrophysio-
logical evaluation of patients with thalidomide-induced neuropathy.
Acta Neurol Belg. 2009;109(2):1206.
41. Zara G, Ermani M, Rondinone R, et al. Thalidomide and sensory
neurotoxicity: a neurophysiological study. J Neurol Neurosurg Psy-
chiatry. 2008;79(11):125861.
42. Jackson G, Einsele H, Moreau P, Miguel JS. Bortezomib, a novel
proteasome inhibitor, in the treatment of hematologic malignancies.
Cancer Treat Rev. 2005;31(8):591602.
43. Rampen AJ, Jongen JL, van Heuvel I, et al. Bortezomib-induced
polyneuropathy. Neth J Med. 2013;71(3):12833.
44. Corso A, Mangiacavalli S, Varettoni M, et al. Borte zomib-
induced peripheral neuropathy in multiple myeloma: a com-
parison between previously treated and untreated patients.
Leuk Res. 2010;34(4):4714.
45. El-Cheikh J, Stoppa AM, Bouabdallah R, et al. Features and risk
factors of peripheral neuropathy during treatment with bortezomib
for advanced multiple myeloma. Clin Lymphoma Myeloma.
2008;8(3):14652.
46. Balayssac D, Cayre A, Authier N, et al. Patterns of P-glycoprotein
activity in the nervous system during vincristine-induced neuropa-
thy in rats. J Peripher Nerv Syst. 2005;10(3):30110.
47. Balayssac D, Cayre A, Authier N, et al. Involvement of the multi-
drug resistance transporters in cisplatin-induced neuropathy in rats.
Comparison with the chronic constriction injury model and
monoarthritic rats. Eur J Pharmacol. 2006;544(13):4957.
48. Liu JJ, Lu J, McKeage MJ. Membrane transporters as determinants
of the pharmacology of platinum anticancer drugs. Curr Cancer
Drug Targets. 2012;12(8):96286. This review gives a very good
up-to-date on the involvement of membrane transporters in plati-
num salts neurotoxicity.
49. Ip V, Liu JJ, Mercer JF, McKeage MJ. Differential expression of
ATP7A, ATP7B and CTR1 in adult rat dorsal root ganglion tissue.
Mol Pain. 2010;6:53.
50. Liu JJ, Jamieson SM, S ubrama niam J, et al. Neuron al ex-
pression of copper transporter 1 in rat dorsal root ganglia:
association with platinum neurotoxicity. Cancer Chemother
Pharmacol. 2009;64(4):847 56.
51. Jong NN, Nakanishi T, Liu JJ, et al. Oxaliplatin transport mediated
by organic cation/carnitine transporters OCTN1 and OCTN2 in
overexpressing human embryonic kidney 293 cells and rat dorsal
root ganglion neurons. J Pharmacol Exp Ther. 2011;338(2):53747.
52. Cavaletti G, Tredici G, Pizzini G, Minoia A. Tissue platinum con-
centrations and cisplatin schedules. Lancet. 1990;336(8721):1003.
53. Gregg RW, Molepo JM, Monpetit VJ, et al. Cisplatin neurotoxicity:
the relationship between dosage, time, and platinum concentration
in neurologic tissues, and morphologic evidence of toxicity. J Clin
Oncol. 1992;10(5):795803.
54. Thompson SW, Davis LE, Kornfeld M, et al. Cisplatin neuropathy.
Clinical, electrophysiologic, morphologic, and toxicologic studies.
Cancer. 1984;54(7):126975.
55. Adelsberger H, Quasthoff S, Grosskreutz J, et al. The chemothera-
peutic oxaliplatin alters voltage-gated Na(+) channel kinetics on rat
sensory neurons. Eur J Pharmacol. 2000;406(1):2532.
56. Benoit E, Brienza S, Dubois JM. Oxaliplatin, an anticancer agent
that affects both Na+ and K+ channels in frog peripheral myelinated
axons. Gen Physiol Biophys. 2006;25(3):26376.
57. Grolleau F, Gamelin L, Boisdron-Celle M, et al. A possible expla-
nation for a neurotoxic effect of the anticancer agent oxaliplatin on
neuronal voltage-gated sodium channels. J Neurophysiol.
2001;85(5):22937.
58. Kagiava A, Tsingotjidou A , Emmanouilides C , Theophilidis
G. The effects of oxaliplatin, an anticancer drug, on potas-
sium cha nne ls of the p eripheral myeli nated nerve fibres of
the adult rat. Neurotoxicology. 2008;29(6):11006.
59. Webster RG, Brain KL, Wilson RH, et al. Oxaliplatin induces
hyperexcitability at motor and autonomic neuromuscular junctions
through effects on voltage-gated sodium channels. Br J Pharmacol.
2005;146(7):102739.
60. Schulze C, McGowan M, Jordt SE, Ehrlich BE. Prolonged
oxaliplatin exposure alters intracellular calcium signaling: a new
mechanism to explain oxaliplatin-associated peripheral neuropathy.
Clin Colorectal Cancer. 2011;10(2):12633.
61. Graham MA, Lockwood GF, Greenslade D, et al. Clinical pharma-
cokinetics of oxaliplatin: a critical review. Clin Cancer Res.
2000;6(4):120518.
62. Fraser AD. Clinical toxicologic implications of ethylene glycol and
glycolic acid poisoning. Ther Drug Monit. 2002;24(2):2328.
63. Broomand A, Jerremalm E, Yachnin J, et al. Oxaliplatin neurotox-
icityno general ion channel surface-charge effect. J Negat Results
Biomed. 2009;8:2.
64. Dimitrov AG, Dimitrova NA. A possible link of oxaliplatin-
induced neuropathy with potassium channel deficit. Muscle Nerve.
2011;45(3):40311. This study provides a new potential mechanism
for oxaliplatin-induced acute neurotoxicity involving potassium
channels.
65. Marmiroli P, Nicolini G, Miloso M, et al. The fundamental role of
morphology in experimental neurotoxicology: the example of
chemotherapy-induced peripheral neurotoxicity. Ital J Anat
Embryol. 2012;117(2):7597. This review gives a very good over-
view of morphology (nervous lesions) in animal models of CIPN.
66. Tredici G, Braga M, Nicolini G, et al. Effect of recombinant human
nerve growth factor on cisplatin neurotoxicity in rats. Exp Neurol.
1999;159(2):5518.
67. Cavaletti G, Tredici G, Marmiroli P, et al. Morphometric
study of the sensory neuron and peripheral nerve chan ges
induced by chronic cisplatin (DDP ) admini stration in rats.
Acta Neuropathol. 1992;84 (4):364 71.
68. McDonald ES, Randon KR, Knight A, Windebank AJ. Cisplatin
preferentially binds to DNA in dorsal root ganglion neurons in vitro
and in vivo: a potential mechanism for neurotoxicity. Neurobiol Dis.
2005;18(2):30513.
69. Ta LE, Espeset L, Podratz J, Windebank AJ. Neurotoxicity of
oxaliplatin and cisplatin for dorsal root ganglion neurons correlates
with platinum-DNA binding. Neurotoxicology. 2006;27(6):9921002.
70. Gill JS, Windebank AJ. Cisplatin-induced apoptosis in rat dorsal
root ganglion neurons is associated with attempted entry into the cell
cycle. J Clin Invest. 1998;101(12):284250.
71. Podratz JL, Knight AM, Ta LE, et al. Cisplatin induced mitochon-
drial DNA damage in dorsal root ganglion neurons. Neurobiol Dis.
2011;41(3):6618.
72. Warwick RA, Hana ni M. The contribution of satellite glial
cells to chemotherapy-induced neuropathic pain. Eur J Pain .
2013;17(4): 57180.
364, Page 8 of 9 Curr Pain Headache Rep (2013) 17:364
73. Anand U, Otto WR, Anand P. Sensitization of capsaicin and icilin
responses in oxaliplatin treated adult rat DRG neurons. Mol Pain.
2010;6:82.
74. Gauchan P, Andoh T, Kato A, Kuraishi Y. Involvement of increased
expression of transient receptor potential melastatin 8 in oxaliplatin-
induced cold allodynia in mice. Neurosci Lett. 2009;458(2):935.
75. Descoeur J, Pereira V, Pizzoccaro A, et al. Oxaliplatin-induced cold
hypersensitivity is due to remodelling of ion channel expression in
nociceptors. EMBO Mol Med. 2011;3(5):26678.
76. Nassini R, Gees M, Harrison S, et al. Oxaliplatin elicits mechanical
and cold allodynia in rodents via TRPA1 receptor stimulation. Pain.
2011;152(7):162131.
77. Hori K, Ozaki N, Suzuki S, Sugiura Y. Upregulations of P2X(3) and
ASIC3 involve in hyperalgesia induced by cisplatin administration
in rats. Pain. 2010;149(2):393405.
78. Persohn E, Canta A, Schoepfer S, et al. Morphological and mor-
phometric analysis of paclitaxel and docetaxel-induced peripheral
neuropathy in rats. Eur J Cancer. 2005;41(10):14606.
79. Lauria G, Lombardi R, Borgna M, et al. Intraepidermal nerve fiber
density in rat foot pad: neuropathologic-neurophysiologic correla-
tion. J Peripher Nerv Syst. 2005;10(2):2028.
80. Zheng H, Xiao WH, Bennett GJ. Functional deficits in peripheral
nerve mitochondria in rats with paclitaxel- and oxaliplatin-evoked
painful peripheral neuropathy. Exp Neurol. 2011;232(2):15461.
81. Flatters SJ, Bennett GJ. Studies of peripheral sensory nerves in
paclitaxel-induced painful peripheral neuropathy: evidence for mi-
tochondrial dysfunction. Pain. 2006;122(3):24557.
82. Xiao WH, Zheng H, Zheng FY, et al. Mitochondrial abnormality in
sensory, but not motor, axons in paclitaxel-evoked painful periph-
eral neuropathy in the rat. Neuroscience. 2011;199:4619.
83. Barriere DA, Rieusset J, Chanteranne D, et al. Paclitaxel therapy
potentiates cold hyperalgesia in streptozotocin-induced diabetic rats
through enhanced mitochondrial reactive oxygen species produc-
tion and TRPA1 sensitization. Pain. 2012;153(3):55361.
84. Fidanboylu M, Griffiths LA, Flatters SJ. Global inhibition of reac-
tive oxygen species (ROS) inhibits paclitaxel-induced painful pe-
ripheral neuropathy. PLoS One. 2011;6(9):e25212.
85. Siau C, Xiao W, Bennett GJ. Paclitaxel- and vincristine-evoked
painful peripheral neuropathies: loss of epidermal innervation and
activation of Langerhans cells. Exp Neurol. 2006;201(2):50714.
86. Nishida K, Kuchiiwa S, Oiso S, et al. Up-regulation of matrix
metalloproteinase-3 in the dorsal root ganglion of rats with
paclitaxel-induced neuropathy. Cancer Sci. 2008;99(8):161825.
87. Kirchmair R, Tietz AB, Panagiotou E, et al. Therapeutic angiogen-
esis inhibits or rescues chemotherapy-induced peripheral neuropa-
thy: taxol- and thalidomide-induced injury of vasa nervorum is
ameliorated by VEGF. Mol Ther. 2007;15(1):6975.
88. Chen Y, Yang C, Wang ZJ. Proteinase-act ivated re ceptor 2
sensitizes transient receptor potential vanilloid 1, transient
receptor potential vanilloid 4, and transient receptor potential
ankyrin 1 in paclitaxel-induced neuropathic pain. Neurosci-
ence. 2011;193:44051.
89. Alessandri-Haber N, Dina OA, Joseph EK, et al. Interaction of
transient receptor potential vanilloid 4, integrin, and SRC tyrosine
kinase in mechanical hyperalgesia. J Neurosci. 2008;28(5):104657.
90. Muthuraman A, Jaggi AS, Singh N, Singh D. Ameliorative effects
of amiloride and pralidoxime in chronic constriction injury and
vincristine induced painful neuropathy in rats. Eur J Pharmacol.
2008;587(13):10411.
91. Hansen N, Uceyler N, Palm F, et al. Serotonin transporter deficiency
protects mice from mechanical allodynia and heat hyperalgesia in
vincristine neuropathy. Neurosci Lett. 2011;495(2):937.
92. Thibault K, Van Steenwinckel J, Brisorgueil MJ, et al. Serotonin 5-
HT2A receptor involvement and Fos expression at the spinal level in
vincristine-induced neuropathy in the rat. Pain. 2008;140(2):30522.
93. Meregalli C, Canta A, Carozzi VA, et al. Bortezomib-induced
painful neuropathy in rats: a behavioral, neurophysiological and
pathological study in rats. Eur J Pain. 2009;14(4):343
50.
94. Shin YK, Jang SY, Lee HK, et al. Pathological adaptive responses of
Schwann cells to endoplasmic reticulum stress in bortezomib-
induced peripheral neuropathy. Glia. 2010;58(16):196176.
95. Casafont I, Berciano MT, Lafarga M. Bortezomib induces the
formation of nuclear poly(A) RNA granules enriched in Sam68
and PABPN1 in sensory gangli a neurons. Neurotox Res.
2009;17(2):16778.
96. Arastu-Kapur S, Anderl JL, Kraus M, et al. Nonproteasomal targets
of the proteasome inhibitors bortezomib and carfilzomib: a link to
clinical adverse events. Clin Cancer Res. 2011;17(9):273443.
97. Poruchynsky MS, Sackett DL, Robey RW, et al. Proteasome inhib-
itors increase tubulin polymerization and stabilization in tissue
culture cells: a possible mechanism contributing to peripheral neu-
ropathy and cellular toxicity following proteasome inhibition. Cell
Cycle. 2008;7(7):9409.
98. Briani C, Zara G, Rondinone R, et al. Thalidomide neurotoxicity:
prospective study in patients with lupus erythematosus. Neurology.
2004;62(12):228890.
99. Kaley TJ, Deangelis LM. Therapy of chemotherapy-induced pe-
ripheral neuropathy. Br J Haematol. 2009;145(1):314.
100. •• Smith EM, Pang H, Cirrincione C, et al. Effect of duloxetine on
pain, f unction, and quality of life among patients with
chemotherapy-induced painful peripheral neuropathy: a randomized
clinical trial. JAMA. 2013;309(13):135967. This study presents a
robustand well designed trial on the duloxetine efficacy in CIPN.
101. Lynch ME, Cesar-Rittenberg P, Hohmann AG. A double-blind,
placebo-controlled, crossover pilot trial with extension using an oral
mucosal cannabinoid extract for treatment of chemotherapy-
induced neuropathic pain. J Pain Symptom Manag. 2013.
102. Argyriou AA, Bruna J, Marmiroli P, Cavaletti G. Chemotherapy-
induced peripheral neurotoxicity (CIPN): an update. Crit Rev Oncol
Hematol. 2012;82(1):5177.
103. Schloss JM, Colosimo M, Airey C, et al. Nutraceuticals and chemo-
therapy induced peripheral neuropathy (CIPN): a systematic review.
Clin Nutr. 2013. This review pr esents a good overview on the alter-
native strategies to prevent/tr eat CIPN with the use of nutraceuticals.
Curr Pain Headache Rep (2013) 17:364 Page 9 of 9, 364
... Chemotherapy-induced peripheral neuropathy (CIPN) is an adverse consequence of a wide variety of commonly used anticancer agents [1][2][3][4][5][6][7][8][9] and there are no gold standard therapeutics recommended for the prevention or treatment of CIPN [10]. CIPN frequently leads to dose reduction or discontinuation of therapy [4,11,12]. ...
... CIPN is a debilitating adverse effect with a prevalence ranging from 19% to over 85% [8] and caused by a spectrum of classes of widely used anticancer therapeutics including platinum-based agents, microtubule disruptors (taxanes and vinca alkaloids), proteasome, and angiogenesis inhibitors (Table 1) [1][2][3][4][5][7][8][9][23][24][25]. Clinically, CIPN symptoms may be acute, worsen with cumulative drug dosing, or emerge late during the course of treatment, even long after cessation of treatment [18]. ...
... Central to the transport of proteins from the nerve cell body, down the length of the axon are microtubules [3]. A commonly used class of anticancer agents, taxanes, are microtubule binding agents, which produce polymerization that interferes with normal microtubule dynamics linked to disruption of axonal transport [24,33,34]. ...
Article
Full-text available
Chemotherapy-induced peripheral neuropathy (CIPN) is widely recognized as a potentially severe toxicity that often leads to dose reduction or discontinuation of cancer treatment. Symptoms may persist despite discontinuation of chemotherapy and quality of life can be severely compromised. The clinical symptoms of CIPN, and the cellular and molecular targets involved in CIPN, are just as diverse as the wide variety of anticancer agents that cause peripheral neurotoxicity. There is an urgent need for extensive molecular and functional investigations aimed at understanding the mechanisms of CIPN. Furthermore, a reliable human cell culture system that recapitulates the diversity of neuronal modalities found in vivo and the pathophysiological changes that underlie CIPN would serve to advance the understanding of the pathogenesis of CIPN. The demonstration of experimental reproducibility in a human peripheral neuronal cell system will increase confidence that such an in vitro model is clinically useful, ultimately resulting in deeper exploration for the prevention and treatment of CIPN. Herein, we review current in vitro models with a focus on key characteristics and attributes desirable for an ideal human cell culture model relevant for CIPN investigations.
... In fact, unlike the central nervous system, DRG are devoid of an efficient blood-nerve barrier and are directly exposed to the toxic effects of chemotherapy drugs. Until now, the mechanisms proposed for chemotherapy-induced neurotoxicity include mitochondrial dysfunction, neuronal apoptosis, increased oxidative stress, and altered ionic transport in the somata of DRG sensory neurons as well as in their axons along peripheral nerves [34,36,37]. Moreover, considering the chemotherapy-specific mechanisms of action on cancer cells, differences in the mechanisms of neurotoxicity can be also suspected. ...
... In fact, unlike the central nervous system, DRG are devoid of an efficient blood-nerve barrier and are directly exposed to the toxic effects of chemotherapy drugs. Until now, the mechanisms proposed for chemotherapyinduced neurotoxicity include mitochondrial dysfunction, neuronal apoptosis, increased oxidative stress, and altered ionic transport in the somata of DRG sensory neurons as well as in their axons along peripheral nerves [34,36,37]. Moreover, considering the chemotherapyspecific mechanisms of action on cancer cells, differences in the mechanisms of neurotoxicity can be also suspected. ...
Article
Full-text available
Chemotherapy-induced peripheral neurotoxicity is one of the most common dose-limiting toxicities of several widely used anticancer drugs such as platinum derivatives (cisplatin) and taxanes (paclitaxel). Several molecular mechanisms related to the onset of neurotoxicity have already been proposed, most of them having the sensory neurons of the dorsal root ganglia (DRG) and the peripheral nerve fibers as principal targets. In this study we explore chemotherapy-induced peripheral neurotoxicity beyond the neuronocentric view, investigating the changes induced by paclitaxel (PTX) and cisplatin (CDDP) on satellite glial cells (SGC) in the DRG and their crosstalk. Rats were chronically treated with PTX (10 mg/Kg, 1qwx4) or CDDP (2 mg/Kg 2qwx4) or respective vehicles. Morpho-functional analyses were performed to verify the features of drug-induced peripheral neurotoxicity. Qualitative and quantitative immunohistochemistry, 3D immunofluorescence, immunoblotting, and transmission electron microscopy analyses were also performed to detect alterations in SGCs and their interconnections. We demonstrated that PTX, but not CDDP, produces a strong activation of SGCs in the DRG, by altering their interconnections and their physical contact with sensory neurons. SGCs may act as principal actors in PTX-induced peripheral neurotoxicity, paving the way for the identification of new druggable targets for the treatment and prevention of chemotherapy-induced peripheral neurotoxicity.
... As remission rates have increased dramatically over recent decades, the incidence of chemotherapy-induced neurotoxicity (CIPN) has seen a significant increase (Argyriou et al. 2014;Cavaletti 2014). It is thought that anywhere between 30% and 80% of patients treated with an antineoplastic agent will go on to develop a peripheral neuropathy (Ferrier et al. 2013). The time to onset of CIPN is heterogeneous and can manifest in acute and chronic forms; for example, cisplatin predominantly leads to chronic neurological effects (Argyriou et al. 2014) while oxaliplatin demonstrates toxic neurological effects in both the acute and chronic settings (Argyriou et al. 2014). ...
... Vincristine has the ability to alter mitochondrial function and cause dysregulation of calcium homeostasis, leading to increased neuronal excitability, glial cell dysfunction and activation of apoptosis (Starobova and Vetter 2017;Kerckhove et al. 2017). Mitochondrial damage is the cause of an overproduction of reactive oxygen species with increased oxidative stress and activation of astrocytes in the spinal cord (Ferrier et al. 2013). This activation of glial cells is mediated via upregulation of IL-1β which increases phosphorylation of NMDA receptors of spinal cord cells, resulting in both increased neuronal excitability and increased transmission of pain, which underlie the symptoms of vincristine-induced peripheral neuropathy (Kerckhove et al. 2017). ...
Article
Full-text available
In the treatment of cancer, patients that receive anti-cancer drugs such as Vincristine develop peripheral neuropathic pain. Scyphocephalione A is a new bioactive compound isolated from Scyphocephalium ochocoa (Myristicaceae), a medicinal plant traditionally used in African countries. Recently, an in vitro study has shown its anti-inflammatory and cytotoxic activities on MCF-7 cell line of mammary carcinoma. The purpose of the present study was to assess the in vitro anti-inflammatory and in vivo anti-nociceptive activities of Scyphocephalione A. In vitro tests were carried out on cyclooxygenase and 5-lipoxygenase activities, and on protein denaturation; while in vivo tests were performed on acute and chronic pain models. It was noticed that Scyphocephalione A (1000 µg/ml), inhibits proteins denaturation, cyclooxygenase and 5-lipoxygenase activities respectively by 74.21%, 75.80% and 64.43%. The dose 50 mg/kg of Scyphocephalione A, inhibits acetic acid (63.43%, p < 0.001) and formalin (42.12%, p < 0.001) within first phase and 67.53% (p < 0.001) within second phase)-induced pains. At the same dose, Scyphocephalione A significantly inhibited mechanical and heat hyperalgesia, as well as cold allodynia induced by vincristine. In addition, the compound restored haematological, biochemical and oxidative stress parameters which were altered following Vincristine administration. These results suggest that Scyphocephalione A is endowed with anti-inflammatory potential and antinociceptive properties. Therefore, Scyphocephalione A can be classified as a promising molecule for the management of peripheral neuropathic pain triggered by anti-cancer drug. Graphical abstract
... Chemotherapy-induced peripheral neuropathy (CIPN) is a frequent complication of common anticancer therapies [1]. CIPN represents a debilitating side-effect arising from the administration of chemotherapeutic agents and its incidence is growing exponentially [2]. The chemotherapy drugs that can origin CIPN include vinca alkaloids, taxanes, and, above all, platinum analogues. ...
Article
Full-text available
Chemotherapy-induced neuropathy is a common, dose-dependent adverse effect of several antineoplastics, such as oxaliplatin (L-OHP). The aim of the present work was to evaluate the potential beneficial effects of 2-pentadecyl-2-oxazoline (PEA-OXA) in a murine model of oxaliplatin-induced peripheral neuropathy (OIPN). OIPN was induced by an intraperitoneally injection of L-OHP in rats on five consecutive days (D0–4) for a final cumulative dose of 10 mg/kg. PEA-OXA and ultramicronized palmitoylethanolamide (PEAum), both 10 mg/kg, were given orally 15–20 min prior (L-OHP) and sacrifice was made on day 25. Our results demonstrated that PEA-OXA, more than PEAum, reduced the development of hypersensitivity in rats; this was associated with the reduction in hyperactivation of glia cells and the increased production of proinflammatory cytokines in the dorsal horn of the spinal cord, accompanied by an upregulation of neurotrophic factors in the dorsal root ganglia (DRG). Moreover, we showed that PEA-OXA reduced L-OHP damage via a reduction in NF-κB pathway activation and a modulation of Nrf-2 pathways. Our findings identify PEA-OXA as a therapeutic target in chemotherapy-induced painful neuropathy, through the biomolecular signaling NF-κB/Nrf-2 axis, thanks to its abilities to counteract L-OHP damage. Therefore, we can consider PEA-OXA as a promising adjunct to chemotherapy to reduce chronic pain in patients.
... The pathogenesis of CIPN is largely unknown, and this incomplete knowledge is a main reason for the absence of effective neuroprotection strategies while maintaining chemotherapy drug anticancer activities (2)(3)(4)(5). Several classes of anticancer drugs with different antineoplastic mechanisms can induce CIPN (6)(7)(8). However, sensory impairment is the predominant adverse effect associated with each class, suggesting the existence of a common mechanism of pathogenesis (2). ...
Book
Full-text available
12th International Medicine and Health Sciences Researches Congress
Article
Full-text available
INTRODUCTION Chemotherapy Induced Peripheral Neuropathy (CIPN) is a serious adverse effect of drugs like paclitaxel, vincristine, oxaliplatin and bortezomib [1]. The debilitating and painful symptoms like spontaneous pain, allodynia and hyperalgesia result in noncompliance of chemotherapy schedule by cancer patients. The management of CIPN is mostly unsatisfactory. A variety of drugs like anticonvulsants, antidepressants, opioids, local anaesthetics and antioxidants are employed to treat CIPN [2] however, with only partial success. Moreover, the above drugs also produce various adverse effects confounding the treatment paradigm. Thus, there is an urgent requirement for newer agents to overcome CIPN. Such new drugs per se should be free from serious adverse effects to ensure proper adherence to chemotherapeutic schedule. Though the aetiopathogenesis of CIPN is complex, extensive preclinical research provides evidences for three main neuronal toxic effects of cancer chemotherapeutic drugs. These drugs may target mitochondria and cause severe oxidative stress, produce functional impairment of ion channels and trigger inflammatory mechanisms through the activation of glial cells [3]. Flavone compounds are naturally occurring antioxidants [4] and the anti-inflammatory activity of many flavone derivatives has been documented [5,6]. Considering these beneficial effects, a few flavonoids like myricetin, quercetin and rutin have been investigated and found to be effective in animal models of neuropathy [7,8]. Recently, 6-methoxyflavone has been reported to suppress the manifestations of cisplatin induced peripheral neuropathy in rodents [9]. Flavone and its monohydroxy derivatives have been found to possess significant antinociceptive effect in mice [10] and potent anti-inflammatory effect in rats [5]. However, these compounds have not been investigated for their effect on CIPN. Hence, the main aim of the present study was to investigate the effect of flavone and a few structurally related monohydroxy flavones (5-hydroxy flavone, 6-hydroxy flavone and 7-hydroxy flavone) for their potential effect in CIPN induced by two most commonly used chemotherapeutic drugs vincristine and oxaliplatin in mice.
Article
Peripheral neuropathy is a common disorder that results from nerve damage in the periphery. The degeneration of sensory axon terminals leads to changes or loss of sensory functions, often manifesting as debilitating pain, weakness, numbness, tingling, and disability. The pathogenesis of most peripheral neuropathies remains to be fully elucidated. Cumulative evidence from both early and recent studies indicates that tubulin damage may provide a common underlying mechanism of axonal injury in various peripheral neuropathies. In particular, tubulin post-translational modifications have been recently implicated in both toxic and inherited forms of peripheral neuropathy through regulation of axonal transport and mitochondria dynamics. This knowledge forms a new area of investigation with the potential for developing therapeutic strategies to prevent or delay peripheral neuropathy by restoring tubulin homeostasis.
Article
Full-text available
Purpose The objectives of this project were to assess the current situation and management of cancer-related neuropathic pain (CRNP) in Spain and to provide specific recommendations for the assessment, diagnosis and treatment of CRNP using a Delphi methodology. Methods This was a qualitative study that followed a Delphi methodology using a questionnaire with 56 statements that were grouped into 5 areas related to CRNP: prevalence and impact, pathophysiology, assessment and diagnosis, specific syndromes, treatment, and multidisciplinary approach. Based on the responses, the scientific committee prepared an algorithm and a recommended pathway for the management of CRNP. Results Seventy-nine physicians attended the meeting and completed the questionnaire. Consensus was reached for all statements relating to the prevalence and impact of CRNP. However, the perceptions of specialists from palliative care of the frequency and impact of CRNP differed from those of other specialists. A high degree of consensus was reached for all statements concerning the assessment and diagnosis of CRNP. Regarding specific syndromes, the only statement with a lack of consensus was that on the frequency of NP in patients undergoing radiotherapy. There were some disagreements regarding the multidisciplinary approach and referral criteria for the management of NP. Conclusion Our results show a large degree of agreement on the assessment, diagnosis and treatment of cancer-related neuropathic pain among the specialists involved in its management. There were, however, some disagreements regarding the multidisciplinary approach and referral criteria for the management of neuropathic pain.
Article
Full-text available
Chemotherapy induced peripheral neuropathy [CIPN] is a common significant and debilitating side effect resulting from the administration of neurotoxic chemotherapeutic agents. These pharmaco-chemotherapeutics can include taxanes, vinca alkaloids and others. Moderate to severe CIPN significantly decreases the quality of life and physical abilities of cancer patients and current pharmacotherapy for CIPN e.g. Amifostine and antidepressants have had limited efficacy and may themselves induce adverse side effects. To determine the potential use of nutraceuticals i.e. vitamin E, acetyl-L-carnitine, glutamine, glutathione, vitamin B6, omega-3 fatty acids, magnesium, calcium, alpha lipoic acid and n-acetyl cysteine as adjuvants in cancer treatments a systematic literature review was conducted. Revised clinical studies comprised of randomized clinical trials that investigated the anti-CIPN effect of nutraceuticals as the adjuvant intervention in patients administered chemotherapy. Twenty-four studies were assessed on methodological quality and limitations identified. Studies were mixed in their recommendations for nutraceuticals. Currently no agent has shown solid beneficial evidence to be recommended for the treatment or prophylaxis of CIPN. The standard of care for CIPN includes dose reduction and/or discontinuation of chemotherapy treatment. The management of CIPN remains an important challenge and future studies are warranted before recommendations for the use of supplements can be made.
Article
Full-text available
There are no known effective treatments for painful chemotherapy-induced peripheral neuropathy. To determine the effect of duloxetine, 60 mg daily, on average pain severity. Randomized, double-blind, placebo-controlled crossover trial at 8 National Cancer Institute (NCI)-funded cooperative research networks that enrolled 231 patients who were 25 years or older being treated at community and academic settings between April 2008 and March 2011. Study follow-up was completed July 2012. Stratified by chemotherapeutic drug and comorbid pain risk, patients were randomized to receive either duloxetine followed by placebo or placebo followed by duloxetine. Eligibility required that patients have grade 1 or higher sensory neuropathy according to the NCI Common Terminology Criteria for Adverse Events and at least 4 on a scale of 0 to 10, representing average chemotherapy-induced pain, after paclitaxel, other taxane, or oxaliplatin treatment. The initial treatment consisted of taking 1 capsule daily of either 30 mg of duloxetine or placebo for the first week and 2 capsules of either 30 mg of duloxetine or placebo daily for 4 additional weeks. The primary hypothesis was that duloxetine would be more effective than placebo in decreasing chemotherapy-induced peripheral neuropathic pain. Pain severity was assessed using the Brief Pain Inventory-Short Form "average pain" item with 0 representing no pain and 10 representing as bad as can be imagined. Individuals receiving duloxetine as their initial 5-week treatment reported a mean decrease in average pain of 1.06 (95% CI, 0.72-1.40) vs 0.34 (95% CI, 0.01-0.66) among those who received placebo (P = .003; effect size, 0.513). The observed mean difference in the average pain score between duloxetine and placebo was 0.73 (95% CI, 0.26-1.20). Fifty-nine percent of those initially receiving duloxetine vs 38% of those initially receiving placebo reported decreased pain of any amount. CONCLUSION AND RELEVANCE: Among patients with painful chemotherapy-induced peripheral neuropathy, the use of duloxetine compared with placebo for 5 weeks resulted in a greater reduction in pain. clinicaltrials.gov Identifier: NCT00489411.
Article
Full-text available
The peripheral nervous system is a frequent target of toxic agents. The accurate identification of the sites of neurotoxic action through the morphological characterization of reliable in vivo models or in vitro systems can give fundamental clues when investigating the pathogenesis and interpreting the clinical features of drug-induced neuropathy. The morphological approach has been used to investigate almost all the anticancer drugs able to induce chemotherapy-induced peripheral neurotoxicity, i.e. platinum drugs, antitubulins and proteasome inhibitors. No models have ever been described for thalidomide. This review demonstrates that any pathogenetic study on chemotherapy-induced peripheral neurotoxicity must be based on solid morphological observations obtained in reliable animal and in vitro models. This is particularly true in this setting, since the availability of tissues of human origin is extremely limited. In fact, peripheral (generally sural) nerve biopsies are never required for diagnostic purposes in chemotherapy-treated cancer patients, and their use for a purely scientific aim, although potentially very informative, is not ethical. Moreover, several neurotoxic drugs target the dorsal root ganglia neurons, and it is very difficult to obtain high-quality specimens even from early autopsies. It is, therefore, our opinion that an extensive morphological assessment of the in vitro and in vivo effect of any potentially neurotoxic antineoplastic drugs, as well as of neuroprotectant agents, should be taken into consideration right from the earliest stages of their development.
Article
PURPOSE: In a previous study of treatment for advanced colorectal cancer, the LV5FU2 regimen, comprising leucovorin (LV) plus bolus and infusional fluorouracil (5FU) every 2 weeks, was superior to the standard North Central Cancer Treatment Group/Mayo Clinic 5-day bolus 5FU/LV regimen. This phase III study investigated the effect of combining oxaliplatin with LV5FU2, with progression-free survival as the primary end point. PATIENTS AND METHODS: Four hundred twenty previously untreated patients with measurable disease were randomized to receive a 2-hour infusion of LV (200 mg/m²/d) followed by a 5FU bolus (400 mg/m²/d) and 22-hour infusion (600 mg/m²/d) for 2 consecutive days every 2 weeks, either alone or together with oxaliplatin 85 mg/m² as a 2-hour infusion on day 1. RESULTS: Patients allocated to oxaliplatin plus LV5FU2 had significantly longer progression-free survival (median, 9.0 v 6.2 months; P = .0003) and better response rate (50.7% v 22.3%; P = .0001) when compared with the control arm. The improvement in overall survival did not reach significance (median, 16.2 v 14.7 months; P = .12). LV5FU2 plus oxaliplatin gave higher frequencies of National Cancer Institute common toxicity criteria grade 3/4 neutropenia (41.7% v 5.3% of patients), grade 3/4 diarrhea (11.9% v 5.3%), and grade 3 neurosensory toxicity (18.2% v 0%), but this did not result in impairment of quality of life (QoL). Survival without disease progression or deterioration in global health status was longer in patients allocated to oxaliplatin treatment (P = .004). CONCLUSION: The LV5FU2-oxaliplatin combination seems beneficial as first-line therapy in advanced colorectal cancer, demonstrating a prolonged progression-free survival with acceptable tolerability and maintenance of QoL.
Article
PURPOSETo identify the major sites of platinum accumulation within neural tissues after treatment with cisplatin and to determine the relationship between cumulative dosage, time, and the development of histopathological and clinical neurotoxicity.PATIENTS AND METHODS Twenty-one patients treated antemortem with cisplatin had neural tissue harvested at autopsy. Neural tissues were assayed for platinum and examined for histopathologic evidence of neurotoxicity. The relationship between histopathologic neurotoxicity and various pharmacologic parameters was analyzed.RESULTSTissue platinum levels were found to be highest in the dorsal root ganglia and lowest in tissue protected by the blood-brain barrier. For peripheral nerve, dorsal root, and dorsal root ganglia, a linear relationship was observed between platinum levels and cumulative dose. Platinum levels in neural tissue were not observed to decrease with time. Histopathologic toxicity closely matched an index of exposure to platinum (cumulative dose and lo...
Article
Vincristine is an antineoplastic drug with a broad spectrum of activity against haematological malignancies and childhood sarcomas. Besides useful activity, it lacks the usual emetic and myelotoxicity of anticancer drugs, but its use is limited by neurotoxicity. The neurotoxicity commonly manifests as reduced motility of the intestines resulting in constipation, and peripheral neuropathy which is predominantly sensory in nature. The early symptoms include numbness and tingling of hands and feet which is accompanied by loss of deep tendon reflexes. In its more severe form, muscle weakness develops which is more marked in distal muscles of the hands and feet. Other manifestations of neurotoxicity include ocular palsies, hoarseness of voice, and autonomic neuropathy in the form of postural hypotension and atony of the urinary bladder. The neurotoxicity is dose related and cumulative with repeated dosage such that the drug therapy has to be stopped after a cumulative dose of 30 to 50mg. The neurotoxicity is usually reversible on interruption of the therapy, but the recovery is slow and takes several months. There are no specific antidotes which have established usefulness against neurotoxicity.
Article
Context: Neuropathic pain caused by chemotherapy limits dosing and duration of potentially life-saving anti-cancer treatment and impairs quality of life. Chemotherapeutic neuropathy responds poorly to conventional treatments, and there is an urgent medical need for new treatments. Recent preclinical studies demonstrate that cannabinoid agonists suppress established chemotherapy-evoked neuropathy. Objectives: This was a pilot trial to begin to investigate a currently available cannabinoid agent, nabiximols (oral mucosal spray containing cannabinoids), in the treatment of chemotherapy-induced neuropathic pain. Methods: A randomized, placebo-controlled crossover pilot study was done in 16 patients with established chemotherapy-induced neuropathic pain. A 0-10 point numeric rating scale for pain intensity (NRS-PI) was used as the primary outcome measure. Results: When examining the whole group, there was no statistically significant difference between the treatment and the placebo groups on the NRS-PI. A responder analysis demonstrated that there were five participants who reported a two-point or greater reduction in pain that trended toward statistical significance and the number needed to treat was five. Conclusion: Chemotherapy-induced neuropathic pain is particularly resistant to currently available treatments. This pilot trial found a number needed to treat of five and an average decrease of 2.6 on an 11-point NRS-PI in five "responders" (as compared with a decrease of 0.6 with placebo) and supports that it is worthwhile to study nabiximols in a full randomized, placebo-controlled trial of chemotherapy-induced neuropathic pain.
Article
Background: Peripheral neuropathy is a frequent side effect of bortezomib chemotherapy. Relatively little is known about the clinical characteristics of this neuropathy, especially with respect to pain. Our aim was to describe the clinical characteristics and course of bortezomib-induced polyneuropathy. Methods: This is a retrospective cohort study of 39 patients diagnosed with bortezomib-induced polyneuropathy. Results: Pain is the most prominent symptom and 14 of 39 patients suffered from severe pain. More than 50% of our patients used analgesics due to moderate or severe pain. We found no correlation between severity of symptoms of bortezomib-induced polyneuropathy and cumulative dose or dose intensity of bortezomib. Nerve conduction studies did not correlate well with symptom severity. Dose reduction or discontinuation of treatment reduced severity in most cases. Conclusion: Painful polyneuropathy is a frequent, dose-limiting side effect of bortezomib with a relatively good prognosis. Careful neurological monitoring of symptoms and timely dose adjustment is important.