Você está na página 1de 9

Curr Neurol Neurosci Rep (2013) 13:320

DOI 10.1007/s11910-012-0320-5

DEMYELINATING DISORDERS (DN BOURDETTE AND V YADAV, SECTION EDITORS)

Pain and Multiple Sclerosis: Pathophysiology and Treatment


Claudio Solaro & Erika Trabucco &
Michele Messmer Uccelli

Published online: 19 December 2012


# Springer Science+Business Media New York 2012

Abstract Pain is a common symptom in multiple sclerosis Introduction


(MS) and has recently been estimated to be experienced by
up to 75 % of patients. Pain can be present at any point in Multiple sclerosis (MS) is a chronic, demyelinating disease
the course of the disease and patients may experience pain of the central nervous system, with diagnosis occurring
from various causes simultaneously. Pain in MS can also be typically between 20 and 40 years of age, although it can
secondary to other symptoms, such as spasticity, fatigue, be diagnosed in children and in older adults. MS is charac-
and mood disorder. Of all drug use to treat MS symptoms, terized by various clinical courses, including a relapsing
treatment for pain accounts for nearly 30 % of total use. At remitting form which typically shifts to a progressive course
the same time, patients report low satisfaction with pain over time and a progressive form. Common symptoms
management. Pain affects quality of life and can influence include, among others, sensory impairment, extremity
a persons participation in family life and work and affect weakness, urinary dysfunction, visual impairment, fatigue,
mood. Most of the pain literature in the field of MS is based spasticity, incoordination, and pain [1]. Pain may be the
on open-label studies involving small numbers of subjects. most commonly treated symptom in MS, estimated to ac-
Placebo-controlled trials in severe pain syndromes such as count for 30 % of all symptomatic treatment [2]. A number
trigeminal neuralgia are unethical but for other types of MS- of drugs are used for managing symptoms in MS, some of
related pain conditions, placebo-controlled trials are ethical which have been approved for the disease, whereas most
and necessary to establish efficacy, particularly given the have been approved for other illnesses and have been found
well-documented placebo effect for various painful condi- to have some use in treatment of MS. There are no double-
tions This review discusses available data and emphasizes blind, parallel-arm, placebo-controlled trials, and most ther-
areas of pain research that require further attention apeutic approaches are based on clinical experience. Man-
agement of painful symptoms is important since pain has a
Keywords Antiepileptic drugs . Baclofen . Cannabinoids . significant impact on patients quality of life.
Carbamazepine . Central pain . Gabapentin . Lamotrigine .
Levetiracetam . Multiple sclerosis . Neuropathic pain Classification of Pain Syndromes
treatment . Opioid analgesics . Painful tonic spasms . Pain
treatment . Pregabalin . Topiramate . Tricyclic Pain is defined as an unpleasant sensory experience asso-
antidepressants . Trigeminal neuralgia treatment ciated with actual or potential tissue damage or described in
terms of such damage [3]. Treende et al. [4] classified pain
syndromes as nociceptive somatic/visceral and neuropathic
This article is part of the Topical Collection on Demyelinating
Disorders pain. Nociceptive pain occurs as an appropriate encoding of
noxious stimuli and represents a physiological response
C. Solaro (*) : E. Trabucco
transmitted to a conscious level when nociceptors in bone,
Head and Neck Department, Neurology Unit, ASL 3 Genovese,
Largo Rosso 3, muscle, or any body tissue are activated, warning the organ-
16153 Genoa, Italy ism of tissue damage, in turn eliciting coordinated reflexes
e-mail: csolaro@libero.it and behavioral responses. Neuropathic pain is typically
initiated by a primary lesion or dysfunction in the peripheral
M. Messmer Uccelli
Department of Research, Via Operai 40, or central nervous system, with no biological advantage
Genoa, Italy (such as warning), causing suffering and distress. Clinical
320, Page 2 of 9 Curr Neurol Neurosci Rep (2013) 13:320

hallmarks are burning, dysesthetic, piercing pain, painful revised on the basis of several recent articles [627]; see
responses to nonpainful stimuli (allodynia), and/or in- Table 1. Solaro et al. [18] reported, in a large epidemiolog-
creased pain sensation when noxious stimuli are applied ical study based on a face-to-face structured interview, that
(hyperalgesia) [4]. 43 % of MS patients experience at least one type of pain.
A 2008 report classified pain associated with MS in four The presence of pain correlated with disability, disease
categories: continuous central neuropathic pain; intermittent course, disease duration, and age but not with gender. In a
central neuropathic pain (i.e., trigeminal neuralgia, Lher- casecontrol study Svendsen et al. [17] evaluated 771 MS
mitte sign, glossopharyngeal neuralgia); musculoskeletal patients using a postal questionnaire and found a high prev-
pain (i.e., painful tonic spasms, pain secondary to spasticity, alence of pain in both MS patients and controls (79.4 % in
pain related to being wheelchair-bound), and mixed neuro- MS patients vs 74.7 % in controls), with no statistical
pathic and nonneuropathic pain (i.e., headache) [5]. difference between subjects and controls, although pain in
MS patients was severer and more often interfered with
Prevalence of Pain in MS daily activities.
The remainder of the article will discuss the major cate-
Previous estimates of the occurrence of pain in MS patients gories of pain in MS, providing an overview of pathophys-
ranged from 29 to 86 %, although these numbers have been iology and treatment.

Table 1 Studies of the prevalence of pain in multiple sclerosis (MS)

Study N (men/women) Selection method Data collection Prevalence (%)

Clifford and Trotter [6] 317 (not available) Consecutive outpatients Chart review 29
Vermote et al. [7] 83 (not available) Rehabilitation inpatients McGill Pain 54
Questionnaire
Kassirer and 28, of whom 84 % Long-term outpatients Specific questionnaire 75
Osterberg [8] were wheelchair
users (26/2)
Moulin et al. [9] 159 (51/108) Outpatients Specific questionnaire 64
plus interview
Stenager et al. [10] 117 (52/65) Random sample of inpatients Specific questionnaire 65
Warnell [11] 364 (115/249) Outpatients Specific questionnaire 64
Archibald et al. [12] 85 (13/72) Outpatients Specific questionnaire 64
Indaco et al. [13] 141 (61/80) Outpatients Structured interview 57 (excluding headache)
Stenager et al. [14] 49 (22/27) Follow-up of younger Structured interview 86
individuals from a 1991
study
Beiske et al. [15] 142 (47/95) Outpatients Interview 73.9 (pain and/or
sensory complaints)
Ehde et al. [16] 442 (111/331) MS society member database Postal questionnaire 44
Svendsen et al. [17] 627 with MS Casecontrol study Postal questionnaire 79.4 (patients with MS
(214/413) vs vs 74.7 % in controls)
487 controls
Solaro et al. [18] 1,672 (520/1,152) Random sample of outpatients Structured interview 42.8
sterberg et al. [19] 364 (124/240) Single-center registry Postal questionnaire, 57.5
telephone interview,
in-person interview
plus examination
Ehde et al. [20] 180 (40/140) Study pool Postal questionnaire 66
Hadjimichael 9,115 (2,188/6,927) National patient registry Postal questionnaire 74.5
et al. [21]
Piwko et al. [22] 297 (68/229) Population-based Postal questionnaire 71
Khan and Pallant [23] 94 (26/68) Community-based Semistructured interview 67
Boneschi et al. [24] 428 (161/267) Outpatients Semi-structured interview 39.8
Douglas et al. [25] 219 (40/179) Community-based Structured interview 75
Hirsh et al. [26] 2,994 (390/2,604) MS population base Postal questionnaire 91
Seixas et al. [27] 85 (62/23) MS population base Interview 34
Curr Neurol Neurosci Rep (2013) 13:320 Page 3 of 9, 320

Continuous Central Neuropathic Pain Prevalence of Central Neuropathic Pain

Pathophysiology A number of studies have reported central neuropathic pain


to be among the commonest pain syndrome in MS, with a
Although important advances have been made in changes prevalence of nearly 50 % [18]. The classic signs of central
related to peripheral nervous system injury, the pathophys- pain such as hyperalgesia and allodynia have been reported
iological mechanisms of central neuropathic pain is poorly in 38 % of MS patients, although this frequency may seem
defined. MS pain is thought to be a type of central pain lower in clinical practice, perhaps in part due to the diffi-
due to demyelinating lesions in areas involved in pain culty of patients to describe pain [17]. Approximately 40 %
perception. of patients describe a constant and burning sensation usually
Experimental autoimmune encephalomyelitis (EAE) is involving the lower limbs, more frequently distally than
an animal model of MS that is inducible in various rodent proximally. Thirty percent describe this pain as a deep or
strains by immunization with several myelin antigens, is muscular aching [15].
widely used in order to better understand the pathophysio- A recent study evaluated the location of lesions in MS
logical features of the disease as well as the efficacy of patients with or without chronic pain using brain and spinal
medication. However, there are few reports in the literature cord MRI. Thirteen MS patients with chronic pain and ten
specifically studying pain in EAE. The availability of an MS patients without pain were evaluated. There was no
animal model could be an important step for understanding association between pain and the site of demyelination
the mechanism underlying pain in MS and an opportunity to (spinothalamic tract, dorsal columnmedial lemniscus, dor-
test pharmacological therapies. Aicher et al. [28] used pro- solateral funiculus, grey substance, thalamus, or capsula
teolipid protein to induce active immunization (chronic-re- interna) [33].
lapsing form) and proteolipid-protein-specific splenocytes
for passive induction in SJL mice. Nociceptive testing was Treatment of Central Neuropathic Pain
done with withdrawal latencies to a radiant stimulus. In both
models, hypoalgesia peaked prior to the peak of motor The treatment includes tricyclic antidepressants, antiepilep-
deficits, whereas during the chronic phase of the disease, tic medications, intrathecally administered baclofen, opioid
the mice developed hyperalgesia, thus suggesting that EAE analgesics, anesthetic and antiarrhythmic agents, and
may be a useful model for pain. In a chronic-relapsing cannabinoids.
model of EAE, Olechowski et al. [29] studied pain sensi-
tivity and found a significant decrease in elicited pain be- Tricyclic Antidepressants
havior. This behavior was found to involve the
glutamatergic system, suggesting a potential mechanism Tricyclic antidepressants such as amitriptyline, nortriptyline,
underlying neuropathic pain. Thibault et al. [30] character- and clomipramine have historically been considered first-
ized sensory abnormalities including thermal and mechani- line drugs for treatment of central pain. They act at the
cal hyperalgesia and showed a partial effect of medications synaptic level, increasing serotoninergic and noradrenergic
(gabapentin, duloxetine, and tramadol) in two EAE models. transmission, as well as on sodium channels. Adverse
Recently, it has been hypothesized that MS is an acquired effects include drowsiness, dry mouth, constipation, urinary
channelopathy [31]. Sodium channel Nav1.8, whose expres- retention, and hypotension, limiting their use in MS patients
sion is normally restricted to the peripheral nervous system, [34]. There are no available recommendations as to optimal
is present in cerebellar Purkinje cells in a mouse model of dose scheduling.
MS. The ectopic expression of Nav1.8 contributes to symp-
tom development in the model. Dysregulated sodium chan- Antiepileptic Medications
nel expression on sensory fibers can lead to a functional
change in axonal conduction [32], contributing to neuro- Antiepileptic medications are also used in treating central
pathic pain. Although the theory is intriguing and several neuropathic pain associated with MS. Solaro et al. [35]
hypotheses can be made regarding the mechanisms of action evaluated the frequency of antiepileptic medication use
of sodium channel blockers such as antiepileptic drugs (i.e., and reported adverse effects in a cohort of MS patients.
cerbamazepine) on pain in MS, no data are available to draw Carbamazepine is commonly used for treatment of neuro-
definitive conclusions. pathic pain in MS, although often with inadequate results
Although the field has gained insights into the patho- [35]. It has a significantly higher incidence of adverse
physiological mechanisms of neuropathic pain, particularly effects, with a high rate of discontinuation at relatively low
in animal models, their replication in human subjects is doses, as compared with gabapentin and lamotrigine. These
unknown. adverse effects include drowsiness, vertigo, hypertension,
320, Page 4 of 9 Curr Neurol Neurosci Rep (2013) 13:320

bradycardia, rash, neutropenia, and abnormal liver function [44]. Both patients were older than 60 years and were
[36]. Discontinuation of carbamazepine, even at low doses, undergoing therapy with medication acting on the central
is often necessary [35]. It was recently reported that patients nervous system (benzodiazepine, baclofen, and/or trama-
with MS and those with epilepsy have a similar occurrence dol). We suggest a careful evaluation of possible drug inter-
of cutaneous rash (17 % vs 13 %) when using carbamaze- actions particularly in older patients when pregabalin is
pine [37]. Cianchetti et al. [38] performed an open, add-on prescribed.
study with 21 subjects with different types of neuropathic Antiepileptic drugs appear to be at least partially effective
pain, including 15 with central neuropathic pain. Lamotri- for treatment of central neuropathic pain associated with
gine was added to current pain therapy up to a dosage of MS, although with a lack of rigorous, MS-specific clinical
400 mg daily. In two of 15 individuals with MS treated with trials their full potential remains unknown.
a daily dose of 400 mg lamotrigine as an add-on therapy, Unfortunately, many patients never advance to the regi-
pain was successfully treated. Six of 15 patients reported men necessary for pain relief because of intolerable adverse
some improvement. Improvement was sustained from effects [35, 36, 41].
4 months to 1 year. No relevant adverse effects were
reported. One randomized, double-blind, placebo- Intrathecally Administered Baclofen
controlled, cross-over study that included 12 patients with
MS reached a maximum lamotrigine dosage of 400 mg daily Two studies have looked at intrathecal medications for treat-
and found no difference between the lamotrigine and place- ing pain in MS. Four patients with spinal lesions experi-
bo groups for the change in mean pain intensity [39]. Gaba- enced markedly reduced central neuropathic pain with
pentin for treatment of pain was evaluated in an open-label 50 g of baclofen delivered intrathecally [45]. Intrathecally
study of 25 patients with MS. Fifteen of 22 subjects reported administered baclofen (51,200 g/day) and intrathecally
excellent to moderate relief of pain at a dosage of 600 mg administered morphine (2109,500 g/day) reduced pain
daily. Pain in 15 patients was either improved or resolved. in nine subjects with MS without serious adverse effects
Adverse events occurred in 50 % of the study participants, [46]. Generally, support for the intrathecal administration of
five of whom discontinued use of gabapentin because of baclofen in treating neuropathic pain in MS remains
somnolence and dyspepsia [40]. A further case was reported insufficient.
in which gabapentin at a dosage of 900 mg daily resulted in
dramatic improvement in dysesthetic limb pain, without no Opioid Analgesics
adverse effects. Levetiracetam was used in 20 MS patients
with central neuropathic pain in a randomized, single-blind, Only one nonrandomized, single-blind, placebo-controlled
placebo-controlled study at a maximum dosage of 3,000 mg study of 14 subjects with MS-related central neuropathic
daily. The 12 patients receiving active treatment reported a pain used intravenously administered morphine at a median
significant reduction in pain compared with the placebo dose of 0.67 mg/kg body weight [47]. Four patients reported
group. Adverse events occurred in eight treated patients that pain was reduced by more than 50 % after treatment.
versus five patients receiving placebo, with one individual Clearly, insufficient evidence exists for the use of morphine
discontinuing treatment owing to somnolence [41]. A recent for neuropathic pain in MS.
double-blind, placebo-controlled study found no effect of
levetiracetam (3,000 mg/day, 6-week treatment periods) in Cannabinoids
nonselected patients with central pain in MS, but an effect in
subgroups with specific pain symptoms was reported. There Cannabinoids, unlike most other drugs, have been rigorous-
were no differences in ratings of pain relief, total pain ly assessed for the treatment of pain in MS. Four clinical
intensity, or any other outcomes in the total sample (n0 trials have been reported in the literature. In one trial, the
27), but there was significant pain reduction, increased pain synthetic cannabinoid dronabinol improved pain at a daily
relief, and/or more positive pain relief with levetiracetam dose of 10 mg compared with placebo [48]. A second, 5-
than with placebo in patients with intense pain or without week trial of an oromucosal spray form of the cannabis
touch-evoked pain [42]. In one open-label pilot study of extract 9-tetrahydrocannabinol (9-THC) reported benefit
pregabalin, an anticonvulsant agent, 16 subjects with MS with regard to pain intensity in the treatment group, al-
and paroxysmal pain reached a mean treatment dosage of though treated patients experienced deficits in long-term
154 mg daily [43]. Pregabalin was effective in nine patients, memory storage [49]. Sixty-three patients of the 64 who
but three patients discontinued treatment owing to adverse completed the original randomized study continued in an
effects, specifically dizziness in one patient and general open-label extension trial for up to 2 years to determine
malaise in two patients. A subsequent report on two patients long-term tolerability and the effectiveness of 9-THC. In
described delirium at a low dose of pregabalin (75 mg daily) 28 patients who completed the 2-year follow-up, the mean
Curr Neurol Neurosci Rep (2013) 13:320 Page 5 of 9, 320

numerical rating score for pain in the last week of the nerve entry zone in the pons. The presence of a plaque in
randomized trial was 2.9 (scale range 0.08.0) compared the proximal part of the trigeminal root was reported in an
with 3.8 in the placebo group. Ninety-two percent of the 63 MS patient who underwent a rhizotomy for treatment of
patients experienced a least one adverse event. A third, 15- trigeminal neuralgia [57]. Mixed results have been reported
week trial included 630 individuals with stable MS experi- using MRI studies demonstrating multiple causes. A study
encing spasticity. Pain was assessed as a secondary out- reported enhancement on T1-weighted images and hyperin-
come, along with a number of other symptoms, using tense lesions on T2-weighted images of the cisternal portion
subjective scales [50]. Although most patients in the treat- of the nerve or at the root entry zone in eight of 275 scans,
ment arm reported improvement in pain, 20 % reported although trigeminal neuralgia was not present [58]. In a
worsening while receiving treatment with cannabinoids. study of 74 Japanese subjects studied retrospectively, five
Finally, 9-THC and cannabidiol administered sublingually (6.8 %) showed a T1-hypointense and T2-hyperintense,
were compared with a placebo in a consecutive series of nonenhanced, linear lesion in the pons, in the intramedullary
double-blind, randomized, placebo-controlled, single- portion of the nerve that was associated with different sen-
subject, crossover trials. The design consisted of 2-week sory symptoms. Only one subject had trigeminal neuralgia
treatment phases and a subsequent double-blind phase of [59]. A further study of six MS patients with trigeminal
6 weeks duration. neuralgia demonstrated that all subjects had a plaque in the
Although active treatment improved pain, as rated on a pons, whereas no vascular compression was observed [60].
visual analogue scale, the study findings were negative The role of neurovascular compromise in trigeminal neural-
owing to a large placebo effect [51]. gia has also been proposed. Meaney et al. [61] found a
There are more robust data for cannabinoids than for any plaque in the pons on MRI in one of seven patients, whereas
other drugs for treating neuropathic pain in MS, although no vascular compression of the nerve by an artery at the root
head-to-head trial comparing them with other classic pain entry zone was demonstrated in five subjects. A further
treatments has been conducted. study reported nerve compression in 23 of 35 cases (60 %)
[62], whereas T2-weighted imaging demonstrated a brain-
stem lesion in 26 of 35 subjects (74 %). Trigeminal evoked
Intermittent Central Neuropathic Pain potentials, infrequently used for MS patients with trigeminal
neuralgia, can help to indicate the site of lesions, as demon-
Trigeminal Neuralgia strated in two studies in which there were abnormal findings
for five of five patients and 11 of 13 patients [63, 64].
Trigeminal neuralgia is likely the most widely recognized
neuropathic pain syndrome in MS. Prevalence ranges from Treatment of Trigeminal Neuralgia
1.9 % to 6.3 % [18, 19, 52]. Trigeminal neuralgia is char-
acterized by paroxysmal, episodic facial pain, occurring in Although relatively uncommon in MS patients, trigeminal
the sensory trigeminal area, often triggered by external neuralgia is extremely painful, with a relevant impact on
stimuli [53]. A 2010 report, however, found no relationship quality of life [52]. Treatment of trigeminal neuralgia pri-
between trigeminal lesions and clinical symptoms, despite marily consists of antiepileptic medications acting on
extensive involvement of the entire trigeminal complex in voltage-dependent sodium channels, such as carbamazepine
some cases [54]. The clinical features are the same as those and lamotrigine. Although there have been no randomized
of the idiopathic form, with an absence of sensory loss and clinical trials, reports from small, open-label studies on
the presence of trigger points, described by patients as an various medications are available: for cabamazepine [65],
electrical discharge. The first branch is rarely involved lamotrogine [66], gabapentin [67], topiramate [68], combi-
alone, and there is involvement of the second and/or the nation therapy [69], and misoprostol [70]. Carbamazepine is
third branches in approximately 90 % of cases [55]. Atyp- relatively effective in treating trigeminal neuralgia but may
ical facial pain, often confused with trigeminal neuralgia, is result in adverse effects, particularly causing reversible
described as subcontinuous pain, with the absence of trigger worsening of MS symptoms [35, 65]. Lamotrigine was
points and a general sensory deficit in the trigeminal nerve evaluated in three small studies, one involving lamotrigine
area. In this type of pain the lesion is likely located along the in combination with gabapentin [69, 71]. It was effective in
lemniscus lateralis pathway [56]. 22 of 23 subjects when used individually and in five of five
subjects in combination with gabapentin [67, 72]. Gabapen-
Pathophysiology of Trigeminal Neuralgia tin was described in two small studies in a total of 13
subjects. Complete resolution of pain at low doses was
Trigeminal neuralgia is likely due to an electric ectopic achieved in 11 of 13 subjects. Topiramate was assessed in
discharge caused by a plaque at the trigeminal neuralgia two small studies of eight subjects, of which seven reported
320, Page 6 of 9 Curr Neurol Neurosci Rep (2013) 13:320

resolution of pain at relatively low doses [68, 73]. Finally, that most commonly occurs in the lower extremities, rarely
two studies of misoprostol, a prostaglandin E1 analogue, in the upper limbs, more often at night, typically triggered
demonstrated efficacy in nine of 25 subjects [70, 74]. by sensory stimuli [18]. The origin of pain in painful tonic
In summary, there are insufficient data to make evidence- spasms is due to ectopic impulses resulting from demyelin-
based treatment decisions. ation and axonal damage determining a painful sensation at
the level of the muscle [5].
Surgical Interventions for Treatment of Trigeminal Anecdotally, medications such as baclofen, benzodiaze-
Neuralgia pines, gabapentin, and carbamazepine are used for treating
painful tonic spasms. In an open-label study, gabapentin was
Glycerol injection [7577], radiofrequency lesioning [78], reported to be effective at a mean dosage of 487 mg daily in
and radiosurgery [79, 80] are procedures that ablate the 22 of 24 individuals with MS who had painful tonic spasms,
retrogasserian ganglion in order to interrupt the trigeminal of whom three experienced mild adverse effects [84]. In four
pathway. These procedures have been used in MS patients of seven patients, tiagabine, a selective inhibitor of the -
with trigeminal neuralgia who have not responded to phar- aminobutyric acid transporter, was effective in relieving
macological treatment. These procedures cause nerve dam- pain at a daily dose of 12.8 mg. Two subjects dropped out
age that may lead to adverse effects including hypoesthesia/ of the open-label study owing to adverse events that includ-
hyperesthesia, decreased corneal reflex, transitory mastica- ed drowsiness, dizziness, weakness, and nausea [85]. Final-
tory weakness, and hearing loss, although these effects are ly, in an open-label study in which subjects were evaluated
uncommon. Microvascular decompression of the trigeminal using polymyography, botulinum toxin was effective in
root may be effective in carefully selected subjects, although reducing painful tonic spasms in four of five individuals.
recurrence of pain is relevant, suggesting that combined Treatment was maintained for at least 90 days, with no
mechanisms other than nerve compression have a role in adverse events reported [86]. In summary, although painful
the generation of trigeminal neuralgia in MS [81]. tonic spasms have been successfully treated in some cases,
A lack of long-term benefit and the occurrence of poten- the small number of patients and the open-label design
tially serious adverse effects dictates that surgical interven- prevent any conclusions as to the most effective treatment.
tions should be reserved for critical nonresponders to
medications. Pain Secondary to Spasticity

Lhermitte Sign Pain in MS can be secondary to spasticity and successful


spasticity management reduces pain.
Lhermitte sign is reported by 40 % of patients with MS, and
although it is quite common, it is not exclusive to the Pain Related to Being Wheelchair-Bound
disease. Lhermitte sign is described as a painful electric-
like sensation that runs down the back and into the limbs, Patients with a significant level of impairment may require
elicited by flexing the neck. Treatment with low doses of technical aids for mobility, and many may remain seated in a
carbamazepine is rarely requested by patients and is typi- wheelchair for extended periods of time. In these patients,
cally reserved for persistent cases [82]. pain may be due to incorrect posture, improper use of
technical aids, and prolonged wheelchair use [87].
Glossopharyngeal Neuralgia It is important that patients with mobility assistance
needs are evaluated for appropriate interventions. Pharma-
Glossopharyngeal neuralgia is rare in MS. Pain is intense cological treatment should consist of anti-inflammatory
and occurs in the posterior pharynx, tonsillar fossa, and base drugs or opioids, although specific studies in MS are not
of the tongue. One report on three individuals with MS currently available.
found a daily mean dose of 633 mg of carbamazepine to
be successful in reducing pain [83].
Conclusions and Future Directions

Musculoskeletal Pain Recommendations for the treatment of pain in subjects with


MS based on rigorous scientific methods are unavailable
Painful Tonic Spasms and so clinicians base treatment decisions on clinical expe-
rience [88]. The only medications studied in large clinical
Painful tonic spasms occur in approximately 11 % of sub- trials are cannabinoids, although no head-to-head studies
jects with MS and are described as cramping, pulling pain with other medications for pain associated with MS are
Curr Neurol Neurosci Rep (2013) 13:320 Page 7 of 9, 320

currently available. Data suggest that cannabinoids may References


be useful for treating pain in MS, although the safety
profile remains a matter of discussion [89]. Only five Papers of particular interest, published recently, have been
randomized clinical trials on central neuropathic pain highlighted as:
are available in the literature but the sample sizes were Of importance
small [39, 41, 42, 48, 49], and no head-to-head trials Of major importance
have been reported. Research efforts should exploit
what is known about those drugs that have shown
potential in open-label studies for various pain syn- 1. Paty DW, Ebers GC. Multiple sclerosis 135191. Philadelphia:
dromes related to MS, such as central neuropathic pain F.A. Davis Company; 1999. p. 13591.
2. Brichetto G, Messmer Uccelli M, Mancardi GL, Solaro C.
treated with lamotrigine, gabapentin, and levetiracetam,
Symptomatic medication use in multiple sclerosis. Mult Scler.
trigeminal neuralgia treated individually or in combina- 2003;9(5):45860.
tion with lamotrigine and gabapentin, and painful tonic 3. "Part III: pain terms, a current list with definitions and notes on
spasms treated with gabapentin and botulinum toxin. It usage" (pp 209214) Classification of chronic pain, second edi-
tion, IASP Task Force on Taxonomy, edited by H. Merskey and N.
is also difficult to identify pharmaco-naive patients,
Bogduk, IASP Press, Seattle; 1994. p. 20914.
particularly in trigeminal neuralgia. Further, for ethical 4. Treede RD, et al. Neuropathic pain: redefinition and a grading
reasons it is not possible to design placebo studies in system for clinical and research purposes. Neurology.
severe pain syndromes, which would essentially deny a 2008;70:16305.
5. OConnor AB, Schwid SR, Herrmann DN, Markman JD, Dworkin
subject of pain relief when a potentially effective med- RH. Pain associated with multiple sclerosis: systematic review and
ication is available. This type of effort would only be proposed classification. Pain. 2008;137:96111.
possible through multicenter trials that would ensure 6. Clifford DB, Trotter JL. Pain in multiple sclerosis. Arch Neurol.
adequate subject samples. 1984;41:12702.
7. Vermote R, Ketalaer P, Carltonn H. Pain in multiple sclerosis
MS does not occur in isolation, and comorbidities are
patients: a prospective study using the McGill pain questionnaire.
common, particularly in the aging segment of the MS Clin Neurol Neurosurg. 1986;88:8793.
population. Clinicians should be aware that pain may 8. Kassirer MR, Osterberg DH. Pain in chronic multiple sclerosis. J
have various origins and can be unrelated to MS [90]. Pain Symptom Manage. 1987;2:957.
9. Moulin DE, Foley KM, Ebers GC. Pain syndromes in multiple
Further, patients with MS may have difficulty describing
sclerosis. Neurology. 1988;38:18304.
their pain experience, and pain scales are biased by a 10. Stenager E, Knutsen L, Jensen K. Acute and chronic pain syn-
number of factors, including psychological comorbidity dromes in multiple sclerosis. Acta Neurol Scand. 1991;84:197
(i.e., depression), unrealistic expectations related to 200.
11. Warnell P. The pain experience of a multiple sclerosis population: a
treatment, and inadequate sleep [91, 92]. A recent study
descriptive study. Axon. 1991;13:268.
used focus groups to assess how subjects describe their 12. Archibald CJ, et al. Pain prevalence, severity and impact in a
pain experience [93]. MS patients in particular used clinical sample of multiple sclerosis patients. Pain. 1994;58:8993.
various symbolic descriptors to express pain quality 13. Indaco A, Iachetta C, Nappi C, Socci L, Carrieri PB. Chronic and
acute pain syndromes in patients with multiple sclerosis. Acta
not found on the frequently used McGill Pain Question-
Neurol. 1994;16:97102.
naire, suggesting that pain assessment is complex and 14. Stenager E, Knudsen L, Jensen K. Acute and chronic pain syn-
should be based on patient experience [93, 94]. dromes in multiple sclerosis. Ital J Neurol Sci. 1995;16:62932.
Recommendations for effective pain management in MS 15. Beiske AG, Pedersen ED, Czujko B, Myhr KM. Pain and sensory
complaints in multiple sclerosis. Eur J Neurol. 2004;1:47982.
will depend on the development of multiple-arm, random-
16. Ehde DM, et al. Chronic pain in a large community sample of
ized clinical trials, with adequate numbers of subjects with persons with multiple sclerosis. Mult Scler. 2003;6:60511.
specific pain characteristics [88]. In the meantime, it is 17. Svendsen KB, et al. Pain in patients with multiple sclerosis: a
important for clinicians to evaluate the patient for pain at population-based study. Arch Neurol. 2003;60:108994.
18. Solaro C, et al. The prevalence of pain in multiple sclerosis. A
each visit, keeping in mind that the same patient can expe-
multicenter cross-sectional study. Neurology. 2004;63:91921.
rience different types of pain concurrently, thus making 19. sterberg A, Boivie J, Thomas KA. Central pain in multiple
differential diagnosis important. Finally, patients with MS sclerosis-prevalence and clinical characteristics. Eur J Pain.
are at higher risk of adverse effects from pain medications 2005;9:53142.
20. Ehde DM, Osborne TL, Hanley MA, Jensen MP, Kraft GH. The
than the general population and frequently experience
scope and nature of pain in persons with multiple sclerosis. Mult
comorbidities that can distort the clinical assessment, par- Scler. 2006;12:62938.
ticularly depression and fatigue. 21. Hadjimichael O, Kerns RD, Rizzo MA, Cutter G, Vollmer T.
Persistent pain and uncomfortable sensations in persons with mul-
tiple sclerosis. Pain. 2007;127:3541.
22. Piwko C, et al. Pain due to multiple sclerosis: analysis of the
Disclosure No potential conflicts of interest relevant to this article prevalence and economic burden in Canada. Pain Res Manag.
were reported. 2007;12:25965.
320, Page 8 of 9 Curr Neurol Neurosci Rep (2013) 13:320

23. Khan F, Pallant J. Chronic pain in multiple sclerosis: prevalence, 42. Falah M, Madsen C, Holbech JV, Sindrup SH. A randomized,
characteristics and impact on quality of life in an Australian placebo-controlled trial of levetiracetam in central pain in multiple
community cohort. J Pain. 2007;8:61423. sclerosis. Eur J Pain. 2011;15322149. This study is a randomized,
24. Boneschi M, et al. Lifetime and actual prevalence of pain and double-blind, placebo-controlled, cross-over trial with levetirace-
headache in multiple sclerosis. Mult Scler. 2008;14:51421. tam 3,000 mg/day versus placebo. There were no differences in the
25. Douglas C, Wollin J, Windsor C. Illness and demographic corre- ratings of pain relief but an effect in patients with lancinating pain
lates of chronic pain among a community-based sample of people or without touch-evoked pain was observed.
with multiple sclerosis. Arch Phys Med Rehabil. 2008;89:1923 43. Solaro C, Boemker M, Tanganelli P. Pregabalin for treating parox-
32. ysmal symptoms in multiple sclerosis: a pilot study. J Neurol.
26. Hirsh AT, Turner AP, Ehde DM, Haselkorn JK. Prevalence and 2009;256:17734.
impact of pain in multiple sclerosis: physical and psychological 44. Solaro C, Tanganelli P. Acute delirium in patients with multiple
contributors. Arch Phys Med Rehabil. 2009;90:64651. sclerosis treated with pregabalin. Clin Neuropharmacol.
27. Seixa D, Vasco Galhardo MJ, Guimares J, Lima L. Pain in 2009;32:2367.
Portuguese patients with multiple sclerosis. Front Neurol. 45. Herman RM, DLuzansky SC, Ippolito R. Intrathecal baclofen
2011;2:20. suppresses central pain in patients with spinal lesions. A pilot
28. Aicher SA, Silverman MB, Winkler CW, Bebo Jr BF. study. Clin J Pain. 1992;8:33845.
Hyperalgesia in an animal model of multiple sclerosis. Pain. 46. Sadiq S, Poopatana C. Intrathecal basclofen and morphine in
2004;110:56070. multiple sclerosis patients with severe pain and spasticity. J
29. Olechowski CJ, Truong JJ, Kerr BJ. Neuropathic pain behaviours Neurol. 2007;254:14645.
in a chronic-relapsing model of experimental autoimmune enceph- 47. Kalman S, sterbergn A, Srensenn J, Boivie J, Bertler A.
alomyelitis (EAE). Pain. 2009;141(12):15664. This article Morphine responsiveness in a group of well-defined multiple
reports the dysregulation of glutamate transport function in ab- sclerosis patients: a study with i.v. morphine. Eur J Pain.
normal pain behavior in response to persistent noxious stimulation 2002;6:6980.
in EAE mice. 48. Svendsen KB, Jensen TS, Bach FW. Does the cannabinoid drona-
30. Thibault K, Calvino B, Pezet S. Characterisation of sensory binol reduce central pain in multiple sclerosis? Randomised double
abnormalities observed in an animal model of multiple sclerosis: blind placebo controlled crossover trial. BMJ. 2004;329:253.
a behavioural and pharmacological study. Eur J Pain. 2011;15 49. Rog DJ, Nurmikko TJ, Young CA. Oromucosal delta9-
(3):231.e1231.e16. This article reports the clinical signs linked tetrahydrocannabinol/ cannabidiol for neuropathic pain associated
to pain in two models of EAE related to time-course changes in with multiple sclerosis: an uncontrolled, open-label, 2-year exten-
disease representing a model of sensory abnormalities in MS. sion trial. Clin Ther. 2007;29:206879.
Thibault et al. also reported pain relief with medications such as 50. Zajicek J, et al. Cannabinoids for treatment of spasticity and other
gabapentin, duloxetine, and tramadol. symptoms related to multiple sclerosis (CAMS study): multicentre
31. Waxman SG. Acquired channelopathies in nerve injury and MS. randomised placebo-controlled trial. Lancet. 2003;362:151726.
Neurology. 2001;56(12):16217. 26. 51. Wade DT, Makela P, Robson P, House H, Bateman C. Do cannabis-
32. Waxman SG, Dib-Hajj S, Cummins TR, Black JA. Sodium chan- based medicinal extracts have general or specific effects on symp-
nels and pain. Proc Natl Acad Sci USA. 1999;96(14):76359. 6. toms in multiple sclerosis? A double-blind, randomized, placebo-
33. Svendsen KB, Srensen L, Jensen TS, Hansen HJ, Bach FW. controlled study on 160 patients. Mult Scler. 2004;10:43441.
MRI of the central nervous system in MS patients with and without 52. Hooge JP, Redekop WK. Trigeminal neuralgia in multiple sclero-
pain. Eur J Pain. 2011;15(4):395401. This study compares 13 sis. Neurology. 1995;45:12946.
subjects with MS with chronic pain and ten patients without pain. 53. Love S, Coakham HB. Trigeminal neuralgia: pathology and path-
There was no association between chronic pain and the site of ogenesis. Brain. 2001;124:234760.
demyelinating lesions. 54. Mills RJ, Young CA, Smith ET. Central trigeminal involvement
34. Saarto T, Wiffen PJ. Antidepressants for neuropathic pain. in multiple sclerosis using high-resolution MRI at 3T. Br J Radiol.
Cochrane Database of Syst Rev. 2007; Issue 4. Art. No.: 2010;83:4938. The objective of this study was to determine the
CD005454 doi:10.1002/14651858.CD005454.pub2. prevalence of trigeminal lesions using high-resolution MRI at 3T.
35. Solaro C, Brichetto G, Battaglia MA, Messmer Uccelli M, Forty-seven patients with MS with a high prevalence of detectable
Mancardi GL. Antiepileptic medications in multiple sclerosis: trigeminal abnormalities were studied. MRI results did not corre-
adverse effects in a three-year follow-up study. Neurol Sci. spond to trigeminal symptoms.
2005;25:30710. 55. Cruccu G, et al. Trigeminal neuralgia and pain related to mul-
36. Killian JM, Fromm GH. Carbamazepine in the treatment of neu- tiple sclerosis. Pain. 2009;143:18691. MRI scans were performed
ralgia: use and side effects. Arch Neurol. 2001;19:12936. on 130 patients with MS: 50 with trigeminal neuralgia, 30 with
37. Shirzadi M, Alvestad S, Hovdal H, et al. Comparison of carbama- trigeminal sensory disturbances other than trigeminal neuralgia
zepine rash in multiple sclerosis and epilepsy. Acta Neurol Scand. and 50 without pain. Cruccu et al. concluded that the most likely
2012;125(1):603. cause of MS-related trigeminal neuralgia is a pontine plaque
38. Cianchetti C, Zuddas A, Randazzo AP, Perra L, Marrosu MG. damaging the primary afferents. Nevertheless, in some patients a
Lamotrigine adjunctive therapy in painful phenomena in MS: neurovascular contact may act as a concurring mechanism.
preliminary observations. Neurology. 1999;53:433. 56. Hutchins LG, Harnsberger HR, Jacobs JM, Apfelbaum RI.
39. Breuer B, et al. Randomized double-blind, placebo-controlled two Trigeminal neuralgia (tic douloureux): MR imaging assessment.
period crossover pilot trial of lamotrigine in patients with central Radiology. 1990;175:83741.
pain due to multiple sclerosis. Clin Ther. 2007;29:202230. 57. Lazar ML, Kirkpatrick JB. Trigeminal neuralgia and multiple
40. Houtchens MK, Richert JR, Sami A, Rose JW. Open label gaba- sclerosis: demonstration of the plaque in an operative case.
pentin treatment for pain in multiple sclerosis. Mult Scler. Neurosurgery. 1979;5:7117.
1997;3:2503. 58. Da Silva CJ, Da Rocha AJ, Mendes MF, Maja Jr ACM, Braga FT,
41. Rossi S, et al. Effects of Levetiracetam on chronic pain in multiple Tilbery CP. Trigeminal involvement in multiple sclerosis: magnetic
sclerosis: results of a randomized placebo-controlled study. Eur J resonance imaging findings with clinical correlation in a series of
Neurol. 2009;16:3606. patients. Mult Scler. 2005;11:2825.
Curr Neurol Neurosci Rep (2013) 13:320 Page 9 of 9, 320

59. Nakashima I, Fujihara K, Kimpara T, Okita N, Takase S, Itoyama 76. Kondziolka D, Lunsford LD, Bissonette DJ. Long-term results
Y. Linear pontine trigeminal root lesions in multiple sclerosis: after glycerol rhizotomy for multiple sclerosis-related trigeminal
clinical and magnetic resonance imaging studies in 5 cases. Arch neuralgia. Can J Neurol Sci. 1994;21:13740.
Neurol. 2001;58:1014. 77. Berk C, Constantoyannis C, Honey CR. The treatment of trigem-
60. Gass A, Kitchen N, MacManus DG, Moseley IF, Hennerici MG, inal neuralgia in patients with multiple sclerosis using percutane-
Miller DH. Trigeminal neuralgia in patients with multiple sclero- ous radiofrequency rhizotomy. Can J Neurol Sci. 2003;30:2203.
sis: lesion localization with magnetic resonance imaging. 78. Broggi G, Franzini A. Radiofrequency trigeminal rhizotomy in
Neurology. 1997;49(4):11424. treatment of symptomatic non-neoplastic facial pain. J
61. Meaney JFM, Watt JMG, Eldridge PR, Whitehouse GH, Wells Neurosurg. 1982;57:4836.
JCD, Miles JB. Association between trigeminal neuralgia and 79. Rogers CL, et al. Gamma knife radiosurgery for trigeminal neu-
multiple sclerosis: role of magnetic resonance imaging. J Neurol ralgia associated with multiple sclerosis. J Neurosurg. 2002;97
Neurosurg Psychiatry. 1995;59:2539. Suppl 5:52932.
62. Broggi G, Ferroli P, Franzini A, Nazzi V, Farina L, La Mantia L, 80. Huang E, et al. Gamma knife radiosurgery for treatment of trigem-
Milanese C. Operative findings and outcomes of microvascular inal neuralgia in multiple sclerosis patients. Stereotact Funct
decompression for trigeminal neuralgia in 35 patients affected by Neurosurg. 2002;79:4450.
multiple sclerosis. Neurosurgery. 2004;55(4):8308. discussion 81. Broggi G, et al. Role of microvascular decompression in trigeminal
8389. neuralgia and multiple sclerosis. Lancet. 1999;354:18789.
63. Cruccu G, Leandri M, Feliciani M, Manfredi M. Idiopathic and 82. Al-Araji AH, Oger J. Reappraisal of Lhermittes sign in multiple
symptomatic trigeminal pain. J Neurol Neurosurg Psychiatry. sclerosis. Mult Scler. 2005;11:398402.
1990;53(12):103442. 83. Minagar A, Sheremata WA. Glossopharyngeal neuralgia and MS.
64. Bergamaschi R, Romani A, Versino M, Callieco R, Gaspari D, Neurology. 2000;54:136870.
Citterio A, Cosi V. Usefulness of trigeminal somatosensory evoked 84. Solaro C, Uccelli MM, Guglieri P, Uccelli A, Mancardi GL.
potentials to detect subclinical trigeminal impairment in multiple Gabapentin is effective in treating nocturnal painful spasms in
sclerosis patients. Acta Neurol Scand. 1994;89:4124. multiple sclerosis. Mult Scler. 2000;6:1923.
65. Ramsaransing G, Zwanikken C, De Keyser J. Worsening of symp- 85. Solaro C, Tanganelli P. Tiagabine for treating painful tonic spasms
toms of multiple sclerosis associated with carbamazepine. BMJ. in multiple sclerosis: a pilot study. J Neurol Neurosurg Psychiatry.
2000;320:1113. 2004;75:341.
66. Leandri M, et al. Lamotrigine in trigeminal neuralgia secondary to 86. Restivo DA, Tinazzi M, Patti F, Palmeri A, Maimone D.
multiple sclerosis. J Neurol. 2000;247:5568. Botulinum toxin treatment of painful tonic spasms in multiple
67. Solaro C, et al. An open-label trial of gabapentin treatment of sclerosis. Neurology. 2003;61:71920.
paroxysmal symptoms in multiple sclerosis patients. Neurology. 87. Thompson AJ. Symptomatic treatment in multiple sclerosis. Curr
1998;51:60911. Opin Neurol. 1998;11:3059.
68. Solaro C, Uccelli MM, Brichetto G, Gasperini C, Mancardi GL. 88. Solaro C, Messmer Uccelli M. Management of pain in multiple
Topiramate relieves idiopathic and symptomatic trigeminal neural- sclerosis: a pharmacological approach. Nat Rev Neurol.
gia. J Pain Symptom Manage. 2001;21:3678. 2011;7:51927.
69. Solaro C, Uccelli MM, Uccelli A, Leandri M, Mancardi GL. Low- 89. Lienau FS, Fllgraf H, Moser A, Feuerstein TJ. Why do cannabi-
dose gabapentin combined with either lamotrigine or carbamaze- noids not show consistent effects as analgesic drugs in multiple
pine can be useful therapies for trigeminal neuralgia in multiple sclerosis? Eur J Neurol. 2007;14:11629.
sclerosis. Eur Neurol. 2000;44:458. 90. Marrie RA, Horwitz RI, Cutter G, Tyry T, Vollmer T. Association
70. DMKG Study Group. Misoprostol in the treatment of trigeminal between comorbidity and clinical characteristics of MS. Acta
neuralgia associated with multiple sclerosis. J Neurol. 2003;250 Neurol Scand. 2010;29:13541.
(542545). 91. Haythornthwaite JA, Benrud-Larsen LM. Psychological aspects of
71. Lunardi G, et al. Clinical effectiveness of lamotrigine and plasma neuropathic pain. Clin J Pain. 2000;16:S1015.
levels in essential and symptomatic trigeminal neuralgia. 92. Duarte R. In: Kanner R, editor. Pain management secrets, vol. 6.
Neurology. 1997;48:17147. Philadelphia: Hanley & Belfus; 2003.
72. Khan OA. Gabapentin relieves trigeminal neuralgia in multiple 93. Saverino A, Solaro C. Pain in individuals with multiple sclerosis,
sclerosis patients. Neurology. 1998;51:6114. knee prosthesis, and post-herpetic neuralgia: learning from focus
73. Zvartau-Hind M, Din MU, Gilan A, Lisak RP, Khan OA. group patients' experience. Clin J Pain. 2012;28(4):3008. Unique,
Topiramate relieves refractory trigeminal neuralgia in MS patients. spontaneous descriptors, possibly disease-specific, emerged in
Neurology. 2000;55:15878. various focus groups of subjects with MS and other diseases. All
74. Reder AT, Arnason B. Trigeminal neuralgia in multiple sclerosis disease groups used descriptors not included in the McGill Pain
relieved by a prostaglandin E analogue. Neurology. 1995;45:1097 Questionnaire, underlining the need for assessment measures of
100. pain based on individual perception.
75. Linderoth B, Hakanson S. Paroxysmal facial pain in disseminated 94. Vermote R, Ketalaer P, Carlton H. Pain in multiple sclerosis
sclerosis treated by retrogasserian glycerol injection. Acta Neurol patients: a prospective study using the McGill Pain
Scand. 1989;80:3416. Questionnaire. Clin Neurol Neurosurg. 1986;88:8793.

Você também pode gostar