touchNEUROLOGY touchNEUROLOGY
Multiple Sclerosis
Read Time: 12 mins

Symptomatic Treatment in Multiple Sclerosis

Copy Link
Published Online: Oct 26th 2013 US Neurology, 2013;9(1):35–40 DOI: http://doi.org/10.17925/USN.2013.09.01.35
Authors: Sandra Bigi, E Ann Yeh
Quick Links:
Abstract
Article
Article Information
Abstract:
Overview

Multiple sclerosis (MS) is a progressive and disabling neurodegenerative disease that primarily affects young adults. Despite significant therapeutic advances in the prevention of relapses, individuals with MS experience a variety of symptoms, most notably fatigue, spasticity, depression, gait and balance difficulties, and sexual dysfunction. These symptoms may interfere with activities of daily living and have a negative impact on quality of life. This review discusses treatment options for these symptoms.

Keywords

Multiple sclerosis, treatment, management, symptomatic, fatigue, depression, pain, urologic complications, sexual dysfunction

Article:

Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system, and is the leading cause of nontraumatic disability in young adults. Prevalence varies between 50 to 200 per 100,000 people.1–3 Despite recent therapeutic advances that have led to improvements in disease control in relapsing-remitting MS, the majority of individuals with MS experience significant symptomatic problems which may have an negative impact on quality of life.4 These symptomatic problems may include fatigue, pain, spasticity, depression, bowel and bladder dysfunction, balance and gait problems, or sexual dysfunction. Specific therapies targeting these symptoms may help to improve quality of life. This review aims to discuss these therapies.

Fatigue
Fatigue may affect up to 80 % of patients with MS.5–9 Multiple scales for fatigue in MS are currently in use, including the Fatigue Severity Scale (FSS),10 Modified Fatigue Impact Scale (MFIS),11 Visual Analogue Scale for Fatigue,12 the MS Fatigue Scale, and the Neurological Fatigue Index (NFI-MS).13 The pathophysiology of fatigue in MS is incompletely understood, but proinflammatory cytokines, lesion burden, axonal injury, and endocrine functions may play a causative role in fatigue.14,15 Functional magnetic resonance imaging (fMRI) studies have suggested greater activation in the motor-attentional network when performing motor tasks.16 In addition, structural brain changes have been found in patients with MS fatigue: atrophy of the sensorimotor cortex was greater in MS patients with fatigue than controls and MS patients without fatigue, despite no differences between MS patients in overall brain atrophy.17 Baseline disability (Expanded Disability Status Scale [EDSS]), mood, and pain predicts the presence of ongoing fatigue after one year.9,18 Exacerbating factors include heat and humidity.19–21

Pharmacologic and nonpharmacologic treatment options for MS-related fatigue have been well described. Agents that have been investigated for MS fatigue include: (1) amantadine, an agent whose specific mechanism of action in treating MS fatigue is unknown, but which has properties including contributing to the release of dopamine and norepinephrine in the brain, and weak antagonism of NMDA receptors; (2) modafinil, whose mechanism of action is unknown, but is thought to act by increasing hypothalamic histamine levels and increasing levels of extra-synaptic dopamine; and (3) stimulants (amphetamines), including methylphenyldate, pemoline, and lisdexamphetamine.

Two randomized controlled trials (RCTs) have been conducted on modafinil in MS fatigue. In one phase II study of 72 patients with MS, modafinil led to a significant improvement in fatigue compared with placebo at a dose of 200 mg/day, but not at 400 mg/day.22 Similarly, another study of 115 MS patients with fatigue did not show a significant difference between the use of 400 mg/day of modafinil and placebo. No information on response at 200 mg/day was available for this study.23

In a multicenter RCT, amantadine, pemoline, and placebo were compared in 93 MS patients with fatigue. In this study, amantidine-treated patients showed a significant decrease in fatigue as measured by the MS-Fatigue Scale in comparison to pemoline or placebo.24 Evidence for efficacy of stimulants in MS fatigue is limited. For example, methylphenidate leads to improvement of fatigue in patients with chronic fatigue syndrome,25 but no placebo-controlled trials have been conducted on its effect on MS fatigue. Lisdexamphetamine (70 mg/day) was evaluated in a placebo-controlled trial of 63 patients with MS, and although improvements were seen in processing speed and memory, no improvements were seen in fatigue.26

Nonpharmacologic strategies to improve fatigue include cooling, cognitivebehavioral therapy (CBT), and aerobic exercise. Cooling therapy, consisting of a cooling suit applied for 45 minutes two times a day, was found to have beneficial effects on fatigue and strength in an open-label series of six MS patients preselected for heat sensitivity.27 In a single-dose, double-blind randomized, sham-treatment controlled trial of cooling, 84 MS patients with heat sensitivity showed modest improvement in disability with ‘highdose’ cooling, as well as persistent improvements in fatigue as measured by daily recordings and the MFIS.28

In addition, CBT may improve fatigue. An RCT of 72 individuals with MS randomized to CBT or relaxation therapy (RT) showed improvements with both CBT and RT (effect size 3.03 [2.22–3.68] versus 1.83 [1.26–2.34]). The effect was persistent 6 months after treatment.29 More recently, internetbased CBT (MS Invigor8) has been investigated in 40 patients with MS fatigue who were randomized to either the intervention or standard therapy. A large treatment effect was seen in fatigue severity.30

Group exercise in any form may have a positive effect on MS fatigue. In a study of 99 MS patients randomized to group exercise or ‘wait list,’ significant improvements in the FSS were seen in patients randomized to ‘group exercise.’31 In another study comparing routine care (n=71) to physiotherapy-led exercise (n=80), yoga (n=77), or fitness instructorled exercise (n=86) in MS patients with fatigue, all interventions led to improvements in the MFIS in comparison with routine care.32 Other studies have shown similar results with yoga and exercise classes in comparison with controls.33 Group aquatic exercises have also been shown to lead to significant improvements in MS fatigue in comparison to routine care.34

Whether the effect of these exercises on fatigue is related to the group setting, or is due to the exercise itself is difficult to discern from the aforementioned studies. Several other studies focusing on specifically on aerobic training have evaluated its benefit on fatigue. Petajan et al. published a study of 54 patients who were randomized to a 12-week intervention of three 40-minute aerobic exercise sessions/week or no exercise, and although they found improvement in areas such as depression (see below), no effect was seen on fatigue in either group.35 However, in a case series of 21 MS patients with fatigue, elliptical training led to significant improvement in the FSS.36

Pain
Pain accounts for almost one-third (30 %) of symptomatic therapy that is prescribed for MS patients37 and is experienced by almost half (43 %) of MS patients.38 Pain associated with MS may be classified into four categories: continuous central neuropathic pain, intermittent central neuropathic pain, musculoskeletal pain and mixed neuropathic, and nonneuropathic pain.39

Continuous central neuropathic pain, which may described as a burning sensation or deep, aching pain, occurs in about 40–50 % of MS patients.40,41

Although central neuropathic pain is thought to be due to demyelinating lesions in areas of pain perception, a study comparing MS patients with and without pain did not show a correlation between pain and lesion localization.42 Antiepileptic medications (e.g. carbamazepine, lamotrigine, gabapentin, pregabalin, and levetiracetam), tricyclic antidepressants, opioids, intrathecal baclofen, anesthetics, and cannabinoids have been described for MS-related pain in this category. Although several studies have shown antiepileptic drugs to be effective against pain, the rate of discontinuation of these medications due to adverse side effects is high.43,44 Tricyclic antidepressants such as clomipramine, amitryptiline, and nortryptiline are frequently used in the treatment of central pain, although guidelines regarding optimal dose scheduling are lacking.45

The highest level of evidence for pharmacologic intervention in MS pain is in the use of cannabinoids. A double-blind, placebo-controlled crossover trial (n=24) showed a modest decrease in pain using an oral agent, delta-9-tetrahydrocannabinol dronabinol (maximum 10 mg/ day).46 Another large RCT whose primary endpoint was improvement in spasticity scores, but had change in pain as a secondary endpoint (n=630), showed cannabinoids (oral cannabis extract [CE], n=211, delta9- tetrahydrocannabinol [D9THC], n=206, and placebo, n=213) to be more effective than placebo for pain in MS.47

Results of studies of oromucosal tetrahydrocannabinol/cannabidol (THC/CBE) in MS pain have been mixed. Researchers found oromucosal THC/CBE (n=66) to be efficacious for pain in MS in a five-week RCT,48 and further, found sustained effect in an open-label extension study (n=28).49

However, another large, randomized, open-label controlled trial of THC/ CBE oromucosal spray versus placebo as add-on therapy after failure with other pain medications (n=339), showed no difference between the treatment group and placebo after 14 weeks of treatment. On the other hand, statistically significant differences favouring the THC/CBE group were seen in time-to-treatment failure, as well as the secondary endpoints of pain upon withdrawal of medication and sleep quality.50 It is possible that the lack of effect seen in the second trial was due to patient selection, as only patients who had failed conventional pain therapies participated in this trial.

Intermittent central neuropathic pain frequently occurs in the form of trigeminal neuralgia (1.9–6.3 %) in MS patients.38,51 Descriptions of successful treatment of this entity in MS using anticonvulsants, such as topiramate,52 gabapentin,53 lamotrigine,54 and carbamazepine55 have been published, although no randomized-controlled studies have been performed on these interventions in the MS population. Multiple surgical interventions for medically refractory cases have been described, including gamma knife surgery, percutaneous balloon compression, microvascular decompression, and percutaneous radiofrequency rhizotomy.56 A recent meta-analysis suggests little difference in acute pain control related to these procedures, but higher recurrence rate for percutaneous balloon compression.56

Musculoskeletal pain in MS is usually related to painful tonic spasms that occur in the context of spasticity or immobility. These spasms usually happen at night, affect the lower limbs and may be elicited by sensory stimuli.38 Treatment of spasticity-related pain should be oriented toward treatment of the underlying spasticity (see ‘spasticity’ below).

Spasticity
Spasticity occurs in 60 % of MS patients.57 It may manifest as gait disturbance, and affects the lower extremities to a greater extent than the upper extremities in most MS patients. Demyelinating lesions and axonal loss cause upper motor neuron dysfunction, leading to spasticity.58 Painful spasms may result from tonic contractions of both agonists and antagonists due to insufficient descending inhibition at the segmental level of the spinal cord.59

Nonpharmacologic options, such as stretching, may control mild spasticity,60 but the effect of stretching on moderate to severe spasticity is less clear. Medications used in the treatment of spasticity influence the GABAergic system (baclofen, gabapentin, benzodiazepines), the α2 adrenergic system (tizanidine), and calcium release in the muscles (dantrolene).61 Several RCTs have been performed evaluating tizanidine and baclofen in MS spasticity,62–64 the largest of which suggests functional benefit in the majority of patients (80 and 76 %, respectively), with a good tolerability profile at doses up to 24 mg (tizanidine) and 60 mg (baclofen).62 Caution is indicated in discontinuing baclofen, as immediate withdrawal can lead to encephalopathy and seizures.

Several reports suggesting benefit of dantrolene sodium for the treatment of spasticity in MS are available,65–68 but concern regarding its effects on the liver limit its use. Diazepam has been evaluated in an open-label crossover trial comparing it with dantrolene,69 and a double-blinded trial comparing it with baclofen.70 In both studies, diazapem was equally effective to the other agent. Sedation was noted to be a frequent side effect.

In a placebo-controlled, double-masked, crossover trial, gabapentin (900 mg TID) was found to improve impairment related to spasticity without worsening concentration and fatigue.71 It may therefore be considered as an alternative to the abovementioned agents. Finally, cannabinoid extract may be of mild to moderate benefit in MS spasticity. In the large RCT noted above comparing CE and D9THC to placebo, Zajicek et al. found improvements the Ashworth scale in both treatment groups in comparison to placebo (CE versus placebo, 0.32 (95 % confidence interval [CI] –1.04 to 1.67); D9THC versus placebo, 0.94 (-0.44 to 2.31). Patient-reported improvement in spasticity was 61 % and 60 % in the treatment groups, respectively, in comparison to 45 % in the placebo group. In another large RCT, Zajicek et al. compared oral E (n=144) to placebo (n=135), and found it to be effective for spasticity in approximately 29 % of patients in comparison with almost 16 % of placebo-treated patients.72 Importantly, there are concerns regarding the long-term effects of CE on cognition and behavior.73,74

Intrathecal baclofen, in the form of a baclofen pump, is a well-established therapeutic modality for the treatment of patients with spinal-cord related spasticity.75 It was reported to be effective for the management of pain due to spasticity in a case series of four MS patients with spinal cord lesions.76 A retrospective study of its use in 64 patients with MS spasticity suggests benefit in improving comfort in nonambulatory patients with severe spasticity.77 Another retrospective analysis of its long-term use in MS spasticity suggests tolerability and effectiveness that may last up to 12 years.78 Finally, botulinum toxin has been shown to be effective compared with placebo in MS-related spasticity, but it is only indicated in focal spasticity.79,80

Depression
Depression occurs in approximately 50 % of MS patients, three times higher than in the general population.7,81,82 Depression may have a negative impact on cognitive function, relationships, treatment adherence, and quality of life.83–86 Pharmacologic and nonpharmacologic strategies have been evaluated in the treatment of depression in MS patients.87

Several studies of MS-related depression have suggested benefit of fluvoxamine (200 mg),88 sertraline,89 and fluoxetine.90 As for nonpharmacologic interventions, CBT has shown promising results. In a study comparing CBT to supportive expressive therapy and sertraline, CBT sertraline showed equal efficacy. Both were superior to supportive expressive therapy. The effect was sustained after a time period of 6 months.91

Exercise has been reported to have a positive effect on mood in MS. In the 1996 randomized trial of exercise versus no exercise by Petjan et al. discussed above (n=54), significant improvements were seen in the exercise group in depression and anger scores after 5 and 10 weeks of intervention.35 In addition, in one study of female MS patients (n=25), exercise had immediate effects on total mood disturbance, with greater effect seen in patients with high levels of baseline anxiety.92 Progressive resistance training (i.e. strength training), improved mood in a RCT (control group [n=15] or treatment group [n=16]). This benefit was sustained 12 weeks after the intervention was completed.93 Finally, a mindfulnessbased intervention has been shown to be effective for depression when compared with placebo in a study of 67 MS patients.94

Bladder Dysfunction
Over half of patients with MS will experience bladder dysfunction at some point during their disease course due to damage to central autonomic pathways.87 Bladder dysfunction can be related to urinary frequency or urgency, urinary retention or a combination of both due to dyssynergia of the detrusor and sphincter muscles. Untreated, bladder dysfunction leads to complications in over 50 % of cases. These complications include urinary tract infections, urolithiasis, hydronephrosis, and kidney failure.95,96 Nonpharmacologic, pharmacologic, and surgical options can be offered for bladder dysfunction in MS.

Nonpharmacologic bladder rehabilitation programs include a selection or combination of behavioral treatment, pelvic floor muscle training, electromyography biofeedback, and neuromuscular electrical stimulation.97 A study assessing pelvic floor muscle training alone showed a decrease in storage and voiding symptoms and improvement in quality of life in MS patients with bladder dysfunction using standardized questionnaires.98 Another study reported effectiveness of an individual bladder rehabilitation program in a RCT assessing bladder impairment, activity limitation, and quality of life.99 In the event that incomplete voiding and high post-void residuals occur, regular clean intermittent self-catheterization may prevent further complications.100

Anticholinergics are major pharmacologic interventions used in the treatment of bladder dysfunction in MS.101 They reduce urinary frequency, urgency, and incontinence. The side-effect profile includes dry mouth, constipation, cognitive problems, and nausea, leading to a high rate of discontinuation of therapy. According to one study, after 6 months, fewer than 30 % of MS patients continue to be on treatment.102 A valid option for some patients is once-a-day treatment in combination with self-catheterization to reduce urologic complications and reduce incontinence.103 Desmopressin has a positive effect on urine volume and urinary frequency during the first 6 hours after treatment and may be offered on occasion for social occasions or travel.104

Surgical options include bladder augmentation, sacral neuromodulation, and botulinum toxin injections. Augmentation cytoplasty has been shown to be effective in the treatment of refractory urgency, urinary incontinence, or detrusor overactivity.105 Sacral neuromodulation may be used in patients with MS-related bladder dysfunction. A case series of four patients with MS-bladder dysfunction reported improvement in leakage from an average of four episodes/day to 0.3/day, with two of these patients completely dry for 24 hours.106 Long-term follow up (average 43 months, range 7–72 months) of nine women who received this procedure for detrusor hyperreflexia showed sustained benefit in all patients.107

Botulinum toxin injections may be used to treat detrusor-sphincter dyssynergia. The effect lasts approximately 6–12 months, and over 70 % of patients treated successfully with botulinum toxin are able to use reduced anticholinergic therapy. On the other hand, caution is warranted, as botulinim toxin injections may lead to increased post-void residual and therefore increase the need for self-catheterization.108–110

Bowel dysfunction in MS consists of constipation, bowel urgency, or incontinence and can be highly embarrassing for the patient. The pathophysiology of bowel dysfunction in MS patients is not fully understood, but is thought to be due to dysfunctional extrinsic autonomic control of bowel function.111 Bowel dysfunction may worsen due to medications used for other MS symptoms, such as anticholinergics, antidepressants, or medications used for spasticity.111 A conservative approach is recommended as a first step in MS-related constipation. Dietary modifications are recommended, including increasing fluid and fiber intake. In more severe cases, laxatives such as lactulose syrup or polyethylene glycol may be used, as can enemas, but they carry the risk of dependence.112 Patients resistant to standard therapies may benefit from bowel biofeedback therapy.113 Approximately 50 % of the patients evaluated in a study of bowel biofeedback therapy showed improvement in standardized bowel and depression scores.113

Sexual Dysfunction
Sexual dysfunction is common in women and men with MS, but patients may be hesitant to discuss it with their physicians. It may affect patients at any time during the disease course. Female complaints may include decreased vaginal lubrication and loss of libido, whereas men often suffer from erectile and ejaculatory dysfunction. Both genders have been reported to have anorgasmia.114–116 The first step in treatment of sexual dysfunction in MS is to identify potentially aggravating therapies such as antidepressants, anticonvulsants, or anticholinergics. If possible, adjustment in these medications should be attempted. Lubrication can be improved by using synthetic lubricants or phosphodiesterase inhibitors in women.112 Sildenafil has been shown in randomized placebo-controlled trials to lead to significant improvement of erectile dysfunction in male MS patients.117,118 Sildenafil has been shown to improve quality of life in MS in one study,117 whereas another study did not demonstrate significant differences in quality of life compared with placebo.118

Gait Abnormalities—Balance and Falls
Mobility and balance are important areas of concern in the MS population. Multiple cross-sectional studies have shown that approximately half of patients with MS experience falls.119,120 Of MS patients with falls, 79 % will experience recurrent falls.119 Much attention has focused on prevention of falls in this population; risk factors include older age, greater disability, use of an assistive device, decreased walking coordination/endurance, and poor balance.119

Improvements in balance have been explored using computer-based interventions, such as Wii Fit©, but the results of a randomized study (n=84) have suggested no difference between controls and those randomized to Wii Fit©, in the timed-up-and-go, a measure of balance and mobility.121 Group exercise classes, such as group kick-boxing, may be of benefit. One open-label pilot study of patients with MS with mild to moderate disability (n=15) demonstrated improvements in gait speed and some measures of balance after a 5-week group kick-box intervention.122 Finally, assistance in proprioceptive input may be of benefit: preliminary data on MS patients (n=15) has suggested that the use of kinesio-taping on the ankle may improve postural control.123

In another study, MS patients (n=40) and controls (n=12) were randomized to receive a six-week visuo-proprioceptive feedback training program; improvements were seen in walking speed and fall risk.124

Gait training, in the form of robot-assisted gait training (n=15), has been found to be equivalent to conventional walking treatment (n=17) in a group of MS patients with severe motor impairment (EDSS 5-7).125 Importantly, neither of these interventions had a lasting effect: patients returned to baseline 3 months after the intervention.125 This underlines the need for ongoing rehabilitative efforts in this population.

One pharmacologic intervention has been approved by the US Food and Drug Administration (FDA) for the improvement of walking speed in MS, 4-aminopyridine (dalfampridine; Fampyra©). This therapy has been evaluated in a phase III, randomized, double-blind, controlled trial (n=229 on dalfampridine 10 mg BID, n=72, on placebo) and shown to result in a greater proportion of patients responding to therapy than in the placebo group (35 % versus 8 %), as well as a 25 % increase in walking speed in responders.126 A second phase III, double-blind RCT (intervention n=120, placebo n=119) showed similar results (responders 43 % versus nonresponders 9 %; improvement in walking speed in responders 24.9 %).127

Summary
Patients with MS suffer from many comorbidities, including fatigue, depression, pain, bowel and bladder dysfunction, sexual dysfunction, and balance and gait difficulties. A variety of therapies targeted specifically at these symptoms have been evaluated, and in some cases, have shown efficacy in ameliorating these symptoms. As these pharmacologic or nonpharmacologic interventions have the potential to improve the quality of life of patients with MS, they should be considered and offered to MS patients on a regular basis.

Article Information:
Disclosure

Sandra Bigi, MD, has received an educational grant from the Swiss Multiple Sclerosis Society, the Bangerter-Rhyner Foundation, and Biogen-Dompe. E Ann Yeh, MD, has no conflicts of interest to declare.

Correspondence

E Ann Yeh, MD, Hospital for Sick Children, Division of Neurology, 555 University Avenue, Toronto, ON M5G 1X8, Canada. E: ann.yeh@sickkids.ca
An erratum to this article can be found below.

Received

2013-05-23T00:00:00

References

  1. Noseworthy JH, Lucchinetti C, Rodriguez M, Weinshenker BG, Multiple sclerosis, N Engl J Med, 2000;343:938–52.
  2. Pugliatti M, Rosati G, Carton H, et al., The epidemiology of multiple sclerosis in Europe, Eur J Neurol, 2006;13:700–722.
  3. Vukusic S, Van Bockstael V, Gosselin S, Confavreux C, Regional variations in the prevalence of multiple sclerosis in French farmers, J Neurol Neurosurg Psychiatry, 2007;78:707–9.
  4. Tullman MJ, A review of current and emerging therapeutic strategies in multiple sclerosis, Am J Manag Care, 2013;19:s21– 27.
  5. Krupp L, Fatigue is intrinsic to multiple sclerosis (MS) and is the most commonly reported symptom of the disease, Mult Scler, 2006;12:367–68.
  6. Lerdal A, Celius EG, Krupp L, Dahl AA. A prospective study of patterns of fatigue in multiple sclerosis, Eur J Neurol, 2007;14:1338-1343.
  7. Parrish JB, Weinstock-Guttman B, Smerbeck A, et al., Fatigue and Depression in Children With Demyelinating Disorders, J Child Neurol, 2013;28(6):710–15.
  8. Barak Y, Achiron A, Cognitive fatigue in multiple sclerosis: findings from a two-wave screening project, J Neurol Sci, 2006;245:73–6.
  9. Leocani L, Colombo B, Comi G, Physiopathology of fatigue in multiple sclerosis, Neurol Sci, 2008;29 Suppl. 2:S241–3.
  10. Krupp LB, LaRocca NG, Muir-Nash J, Steinberg AD, The fatigue severity scale. Application to patients with multiple sclerosis and systemic lupus erythematosus, Arch Neurol, 1989;46:1121– 3.
  11. Kos D, Kerckhofs E, Carrea I, et al., Evaluation of the Modified Fatigue Impact Scale in four different European countries, Mult Scler, 2005;11:76–80.
  12. Kos D, Nagels G, D’Hooghe MB, et al., A rapid screening tool for fatigue impact in multiple sclerosis, BMC Neurol, 2006;6:27.
  13. Mills RJ, Young CA, Pallant JF, Tennant A, Development of a patient reported outcome scale for fatigue in multiple sclerosis: The Neurological Fatigue Index (NFI-MS), Health Qual Life Outcomes, 2010;8:22.
  14. Heesen C, Nawrath L, Reich C, et al., Fatigue in multiple sclerosis: an example of cytokine mediated sickness behaviour?, J Neurol Neurosurg Psychiatry, 2006;77:34–9.
  15. Flachenecker P, Bihler I, Weber F, et al., Cytokine mRNA expression in patients with multiple sclerosis and fatigue, Mult Scler, 2004;10:165–9.
  16. Specogna I, Casagrande F, Lorusso A, et al., Functional MRI during the execution of a motor task in patients with multiple sclerosis and fatigue, La Radiologia Medica, 2012;117:1398– 1407.
  17. Riccitelli G, Rocca MA, Forn C, et al., Voxelwise assessment of the regional distribution of damage in the brains of patients with multiple sclerosis and fatigue, AJNR Am J Neuroradiol, 2011;32:874–9.
  18. Patrick E, Christodoulou C, Krupp LB, Longitudinal correlates of fatigue in multiple sclerosis, Mult Scler, 2009;15:258–61.
  19. Leavitt VM, Sumowski JF, Chiaravalloti N, Deluca J, Warmer outdoor temperature is associated with worse cognitive status in multiple sclerosis, Neurology, 2012;78:964–8.
  20. Bol Y, Smolders J, Duits A, et al., Fatigue and heat sensitivity in patients with multiple sclerosis, Acta Neurol Scand, 2012;126:384–89.
  21. Bakshi R, Fatigue associated with multiple sclerosis: diagnosis, impact and management, Mult Scler, 2003;9:219–27.
  22. Rammohan KW, Rosenberg JH, Lynn DJ, et al., Efficacy and safety of modafinil (Provigil) for the treatment of fatigue in multiple sclerosis: a two centre phase 2 study, J Neurol Neurosurg Psychiatry, 2002;72:179–83.
  23. Stankoff B, Waubant E, Confavreux C, et al., Modafinil for fatigue in MS: a randomized placebo-controlled double-blind study, Neurology, 2005;64:1139–43.
  24. Krupp LB, Coyle PK, Doscher C, et al., Fatigue therapy in multiple sclerosis: results of a double-blind, randomized, parallel trial of amantadine, pemoline, and placebo, Neurology, 1995;45:1956– 61.
  25. Blockmans D, Persoons P, Van Houdenhove B, Bobbaers H, Does methylphenidate reduce the symptoms of chronic fatigue syndrome?, Am J Med, 2006;119:167 e123–30.
  26. Morrow SA, Smerbeck A, Patrick K, et al., Lisdexamfetamine dimesylate improves processing speed and memory in cognitively impaired MS patients: a phase II study, J Neurol, 2013;260:489–97.
  27. Capello E, Gardella M, Leandri M, et al., Lowering body temperature with a cooling suit as symptomatic treatment for thermosensitive multiple sclerosis patients, Ital J Neurol Sci, 1995;16:533-539.
  28. Schwid SR, Tyler CM, Scheid EA, et al., Cognitive fatigue during a test requiring sustained attention: a pilot study, Mult Scler, 2003;9:503–8.
  29. van Kessel K, Moss-Morris R, Willoughby E, et al., A randomized controlled trial of cognitive behavior therapy for multiple sclerosis fatigue, Psychosom Med, 2008;70:205–13.
  30. Moss-Morris R, McCrone P, Yardley L, et al., A pilot randomised controlled trial of an Internet-based cognitive behavioural therapy self-management programme (MS Invigor8) for multiple sclerosis fatigue, Behav Res Ther, 2012;50:415–21.
  31. Tarakci E, Yeldan I, Huseyinsinoglu BE, et al., Group exercise training for balance, functional status, spasticity, fatigue and quality of life in multiple sclerosis: a randomized controlled trial, Clin Rehabil, 2013 [Epub ahead of print].
  32. Garrett M, Hogan N, Larkin A, et al., Exercise in the community for people with minimal gait impairment due to MS: an assessor-blind randomized controlled trial, Mult Scler, 2013;19(6):782–9.
  33. Oken BS, Kishiyama S, Zajdel D, et al., Randomized controlled trial of yoga and exercise in multiple sclerosis, Neurology, 2004;62:2058–64.
  34. Kargarfard M, Etemadifar M, Baker P, et al., Effect of aquatic exercise training on fatigue and health-related quality of life in patients with multiple sclerosis, Arch Phys Med Rehabil, 2012;93:1701–8.
  35. Petajan JH, Gappmaier E, White AT, et al., Impact of aerobic training on fitness and quality of life in multiple sclerosis, Ann Neurol, 1996;39:432–41.
  36. Huisinga JM, Filipi ML, Stergiou N, Elliptical exercise improves fatigue ratings and quality of life in patients with multiple sclerosis, J Rehabil Res Dev, 2011;48:881–90.
  37. Brichetto G, Messmer Uccelli M, Mancardi GL, Solaro C, Symptomatic medication use in multiple sclerosis, Mult Scler, 2003;9:458–60.
  38. Solaro C, Brichetto G, Amato MP, et al., The prevalence of pain in multiple sclerosis: a multicenter cross-sectional study, Neurology, 2004;63:919–21.
  39. O’Connor AB, Schwid SR, Herrmann DN, et al., Pain associated with multiple sclerosis: systematic review and proposed classification, Pain, 2008;137:96–111.
  40. Beiske AG, Pedersen ED, Czujko B, Myhr KM, Pain and sensory complaints in multiple sclerosis, Eur J Neurol, 2004;11:479–82.
  41. Svendsen KB, Jensen TS, Overvad K, et al., Pain in patients with multiple sclerosis: a population-based study, Arch Neurol, 2003;60:1089–94.
  42. Svendsen KB, Sorensen L, Jensen TS, et al., MRI of the central nervous system in MS patients with and without pain, Eur J Pain, 2011;15:395–401.
  43. Solaro C, Brichetto G, Battaglia MA, et al., Antiepileptic medications in multiple sclerosis: adverse effects in a threeyear follow-up study. Neurol Sci 2005;25:307-310.
  44. Rossi S, Mataluni G, Codeca C, et al., Effects of levetiracetam on chronic pain in multiple sclerosis: results of a pilot, randomized, placebo-controlled study, Eur J Neurol, 2009;16:360–66.
  45. Saarto T, Wiffen PJ, Antidepressants for neuropathic pain, Cochrane Database Syst Rev, 2007;CD005454.
  46. Svendsen KB, Jensen TS, Bach FW, Does the cannabinoid dronabinol reduce central pain in multiple sclerosis? Randomised double blind placebo controlled crossover trial, BMJ, 2004;329:253.
  47. Zajicek J, Fox P, Sanders H, et al., Cannabinoids for treatment of spasticity and other symptoms related to multiple sclerosis (CAMS study): multicentre randomised placebo-controlled trial, Lancet, 2003;362:1517–26.
  48. Rog DJ, Nurmikko TJ, Friede T, Young CA, Randomized, controlled trial of cannabis-based medicine in central pain in multiple sclerosis, Neurology, 2005;65:812–19.
  49. Rog DJ, Nurmikko TJ, Young CA, Oromucosal delta9- tetrahydrocannabinol/cannabidiol for neuropathic pain associated with multiple sclerosis: an uncontrolled, open-label, 2-year extension trial, Clin Ther, 2007;29:2068–79.
  50. Langford RM, Mares J, Novotna A, et al., A double-blind, randomized, placebo-controlled, parallel-group study of THC/ CBD oromucosal spray in combination with the existing treatment regimen, in the relief of central neuropathic pain in patients with multiple sclerosis, J Neurol, 2013;260:984–97.
  51. Hooge JP, Redekop WK. Trigeminal neuralgia in multiple sclerosis, Neurology, 1995;45:1294–6.
  52. Zvartau-Hind M, Din MU, Gilani A, et al., Topiramate relieves refractory trigeminal neuralgia in MS patients, Neurology, 2000;55:1587–8.
  53. Khan OA, Gabapentin relieves trigeminal neuralgia in multiple sclerosis patients, Neurology, 1998;51:611–14.
  54. Leandri M, Lundardi G, Inglese M, et al., Lamotrigine in trigeminal neuralgia secondary to multiple sclerosis, J Neurol, 2000;247:556–8.
  55. Espir ML, Millac P, Treatment of paroxysmal disorders in multiple sclerosis with carbamazepine (Tegretol), J Neurol Neurosurg Psychiatry, 1970;33:528–31.
  56. Montano N, Papacci F, Cioni B, et al., What is the best treatment of drug-resistant trigeminal neuralgia in patients affected by multiple sclerosis? A literature analysis of surgical procedures, Clin Neurol Neurosurg, 2013;115:567–72.
  57. Novotna A, Mares J, Ratcliffe S, et al., A randomized, doubleblind, placebo-controlled, parallel-group, enriched-design study of nabiximols* (Sativex((R)) ), as add-on therapy, in subjects with refractory spasticity caused by multiple sclerosis, Eur J Neurol, 2011;18:1122–31.
  58. Pandyan AD, Gregoric M, Barnes MP, et al., Spasticity: clinical perceptions, neurological realities and meaningful measurement, Disabil Rehabil, 2005;27:2–6.
  59. Kheder A, Nair KP, Spasticity: pathophysiology, evaluation and management, Pract Neurol, 2012;12:289–98.
  60. Smania N, Picelli A, Munari D, et al., Rehabilitation procedures in the management of spasticity, Eur J Phys Rehabil Med, 2010;46:423–38.
  61. Shakespeare DT, Boggild M, Young C, Anti-spasticity agents for multiple sclerosis, Cochrane Database Syst Rev, 2003;CD001332.
  62. Eyssette M, Rohmer F, Serratrice G, et al., Multi-centre, double-blind trial of a novel antispastic agent, tizanidine, in spasticity associated with multiple sclerosis, Curr Med Res Opin, 1988;10:699–708.
  63. Smolenski C, Muff S, Smolenski-Kautz S, A double-blind comparative trial of new muscle relaxant, tizanidine (DS 103- 282), and baclofen in the treatment of chronic spasticity in multiple sclerosis, Curr Med Res Opin, 1981;7:374–83.
  64. Bass B, Weinshenker B, Rice GP, et al., Tizanidine versus baclofen in the treatment of spasticity in patients with multiple sclerosis, Can J Neurol Sci, 1988;15:15–ß19.
  65. Horne ML, Letter: Treatment of spasticity in multiple sclerosis with dantrolene, JAMA, 1976;235:251.
  66. Tolosa ES, Soll RW, Loewenson RB, Letter: Treatment of spasticity in multiple sclerosis with dantrolene, JAMA, 1975;233:1046.
  67. Ladd H, Oist C, Jonsson B, The effect of Dantrium on spasticity in multiple sclerosis, Acta Neurol Scand, 1974;50:397–408.
  68. Gelenberg AJ, Poskanzer DC, The effect of dantrolene sodium on spasticity in multiple sclerosis, Neurology, 1973;23:1313–15.
  69. Schmidt RT, Lee RH, Spehlmann R. Comparison of dantrolene sodium and diazepam in the treatment of spasticity, J Neurol Neurosurg Psychiatry, 1976;39:350–56.
  70. From A, Heltberg A, A double-blind trial with baclofen (Lioresal) and diazepam in spasticity due to multiple sclerosis, Acta Neurol Scand, 1975;51:158–66.
  71. Cutter NC, Scott DD, Johnson JC, Whiteneck G, Gabapentin effect on spasticity in multiple sclerosis: a placebo-controlled, randomized trial, Arch Phys Med Rehabil, 2000;81:164–9.
  72. Zajicek JP, Hobart JC, Slade A, et al., Multiple sclerosis and extract of cannabis: results of the MUSEC trial, J Neurol Neurosurg Psychiatry, 2012;83:1125–32.
  73. Killestein J, Hoogervorst EL, Reif M, et al., Safety, tolerability, and efficacy of orally administered cannabinoids in MS, Neurology, 2002;58:1404–7.
  74. Sastre-Garriga J, Vila C, Clissold S, Montalban X, THC and CBD oromucosal spray (Sativex(R)) in the management of spasticity associated with multiple sclerosis, Expert Rev Neurother, 2011;11:627–37.
  75. Penn RD, Kroin JS, Intrathecal baclofen alleviates spinal cord spasticity, Lancet, 1984;1:1078.
  76. Herman RM, D’Luzansky SC, Ippolito R, Intrathecal baclofen suppresses central pain in patients with spinal lesions. A pilot study, Clin J Pain, 1992;8:338–45.
  77. Ben Smail D, Peskine A, Roche N, et al., Intrathecal baclofen for treatment of spasticity of multiple sclerosis patients, Mult Scler, 2006;12:101–3.
  78. Rekand T, Gronning M, Treatment of spasticity related to multiple sclerosis with intrathecal baclofen: a long-term follow-up, J Rehabil Med, 2011;43:511–14.
  79. Snow BJ, Tsui JK, Bhatt MH, et al., Treatment of spasticity with botulinum toxin: a double-blind study, Ann Neurol, 1990;28:512– 15.
  80. Grazko MA, Polo KB, Jabbari B, Botulinum toxin A for spasticity, muscle spasms, and rigidity, Neurology, 1995;45:712–17.
  81. Minden SL, Schiffer RB, Affective disorders in multiple sclerosis. Review and recommendations for clinical research, Arch Neurol, 1990;47:98–104.
  82. Kessler RC, McGonagle KA, Zhao S, et al., Lifetime and 12-month prevalence of DSM-III-R psychiatric disorders in the United States. Results from the National Comorbidity Survey, Arch Gen Psychiatry, 1994;51:8–19.
  83. D’Alisa S, Miscio G, Baudo S, et al., Depression is the main determinant of quality of life in multiple sclerosis: a classification-regression (CART) study, Disabil Rehabil, 2006;28:307–14.
  84. Arnett PA, Higginson CI, Voss WD, et al., Depression in multiple sclerosis: relationship to working memory capacity, Neuropsychology, 1999;13:546–56.
  85. Maybury CP, Brewin CR, Social relationships, knowledge and adjustment to multiple sclerosis, J Neurol Neurosurg Psychiatry, 1984;47:372–6.
  86. Mohr DC, Goodkin DE, Gatto N, Van der Wende J, Depression, coping and level of neurological impairment in multiple sclerosis, Mult Scler, 1997;3:254–8.
  87. The Goldman Consensus statement on depression in multiple sclerosis, Mult Scler, 2005;11:328–37.
  88. Benedetti F, Campori E, Colombo C, Smeraldi E, Fluvoxamine treatment of major depression associated with multiple sclerosis, J Neuropsychiatry Clin Neurosci, 2004;16:364–6.
  89. Mohr DC, Boudewyn AC, Goodkin DE, et al., Comparative outcomes for individual cognitive-behavior therapy, supportiveexpressive group psychotherapy, and sertraline for the treatment of depression in multiple sclerosis, J Consult Clin Psychol, 2001;69:942–9.
  90. Flax JW, Gray J, Herbert J, Effect of fluoxetine on patients with multiple sclerosis, Am J Psychiatry, 1991;148:1603.
  91. Mohr DC, Boudewyn AC, Goodkin DE, et al., Comparative outcomes for individual cognitive-behavior therapy, supportiveexpressive group psychotherapy, and sertraline for the treatment of depression in multiple sclerosis, J Consult Clin Psychol, 2001;69:942–9.
  92. Petruzzello SJ, Snook EM, Gliottoni RC, Motl RW, Anxiety and mood changes associated with acute cycling in persons with multiple sclerosis, Anxiety Stress Coping, 2009;22:297–307.
  93. Dalgas U, Stenager E, Jakobsen J, et al., Fatigue, mood and quality of life improve in MS patients after progressive resistance training, Mult Scler, 2010;16:480–90.
  94. Grossman I, Miller A, Multiple sclerosis pharmacogenetics: personalized approach towards tailored therapeutics, Epma J, 2010;1:317–27.
  95. Mahfouz W, Corcos J, Management of detrusor external sphincter dyssynergia in neurogenic bladder, Eur J Phys Rehabil Med, 2011;47:639-50.
  96. Bacsu CD, Chan L, Tse V, Diagnosing detrusor sphincter dyssynergia in the neurological patient, BJU Int, 2012;109 (Suppl. 3):31–4.
  97. Lucio AC, Campos RM, Perissinotto MC, et al., Pelvic floor muscle training in the treatment of lower urinary tract dysfunction in women with multiple sclerosis, Neurourol Urodyn, 2010;29:1410–13.
  98. Lucio AC, Perissinoto MC, Natalin RA, et al., A comparative study of pelvic floor muscle training in women with multiple sclerosis: its impact on lower urinary tract symptoms and quality of life, Clinics (Sao Paulo), 2011;66:1563–8.
  99. Khan F, Pallant JF, Pallant JI, et al., A randomised controlled trial: outcomes of bladder rehabilitation in persons with multiple sclerosis, J Neurol Neurosurg Psychiatry, 2010;81:1033–8.
  100. Tubaro A, Puccini F, De Nunzio C, et al., The treatment of lower urinary tract symptoms in patients with multiple sclerosis: a systematic review, Curr Urol Rep, 2012;13:335–42.
  101. Fowler CJ, van Kerrebroeck PE, Nordenbo A, Van Poppel H, Treatment of lower urinary tract dysfunction in patients with multiple sclerosis. Committee of the European Study Group of SUDIMS (Sexual and Urological Disorders in Multiple Sclerosis), J Neurol Neurosurg Psychiatry, 1992;55:986–9.
  102. Howell OW, Reeves CA, Nicholas R, et al., Meningeal inflammation is widespread and linked to cortical pathology in multiple sclerosis, Brain, 2011;134:2755–71.
  103. Chancellor MB, Anderson RU, Boone TB, Pharmacotherapy for neurogenic detrusor overactivity, Am J Phys Med Rehabil, 2006;85:536–45.
  104. Hoverd PA, Fowler CJ, Desmopressin in the treatment of daytime urinary frequency in patients with multiple sclerosis, J Neurol Neurosurg Psychiatry, 1998;65:778–80.
  105. Zachoval R, Pitha J, Medova E, et al., Augmentation cystoplasty in patients with multiple sclerosis, Urol Int, 2003;70:21–26; discussion 26.
  106. Ruud Bosch JL, Groen J, Treatment of refractory urge urinary incontinence with sacral spinal nerve stimulation in multiple sclerosis patients, Lancet, 1996;348:717–19.
  107. Chartier-Kastler EJ, Ruud Bosch JL, Perrigot M, et al., Long-term results of sacral nerve stimulation (S3) for the treatment of neurogenic refractory urge incontinence related to detrusor hyperreflexia, J Urol, 2000;164:1476–80.
  108. Kalsi V, Gonzales G, Popat R, et al., Botulinum injections for the treatment of bladder symptoms of multiple sclerosis, Ann Neurol, 2007;62:452–7.
  109. Habek M, Karni A, Balash Y, Gurevich T, The place of the botulinum toxin in the management of multiple sclerosis, Clin Neurol Neurosurg, 2010;112:592–6.
  110. Deffontaines-Rufin S, Weil M, Verollet D, et al.,Botulinum toxin A for the treatment of neurogenic detrusor overactivity in multiple sclerosis patients, Int Braz J Urol, 2011;37:642–8.
  111. Wiesel PH, Norton C, Glickman S, Kamm MA, Pathophysiology and management of bowel dysfunction in multiple sclerosis, Eur J Gastroenterol Hepatol, 2001;13:441–8.
  112. Markowitz C, Symptomatic therapy of multiple sclerosis, Continuum (Minneap Minn), 2010;16:90–104.
  113. Preziosi G, Raptis DA, Storrie J, et al., Bowel biofeedback treatment in patients with multiple sclerosis and bowel symptoms, Dis Colon Rectum, 2011;54:1114–21.
  114. Zorzon M, Zivadinov R, Bosco A, et al., Sexual dysfunction in multiple sclerosis: a case-control study. I. Frequency and comparison of groups, Mult Scler, 1999;5:418–27.
  115. Demirkiran M, Sarica Y, Uguz S, et al., Multiple sclerosis patients with and without sexual dysfunction: are there any differences?, Mult Scler, 2006;12:209–14.
  116. Barak Y, Achiron A, Elizur A, et al., Sexual dysfunction in relapsing-remitting multiple sclerosis: magnetic resonance imaging, clinical, and psychological correlates, J Psychiatry Neurosci, 1996;21:255–8.
  117. Fowler CJ, Miller JR, Sharief MK, et al., A double blind, randomised study of sildenafil citrate for erectile dysfunction in men with multiple sclerosis, J Neurol Neurosurg Psychiatry, 2005;76:700–5.
  118. Safarinejad MR, Evaluation of the safety and efficacy of sildenafil citrate for erectile dysfunction in men with multiple sclerosis: a double-blind, placebo controlled, randomized study, J Urol, 2009;181:252–8.
  119. Sosnoff JJ, Socie MJ, Boes MK, et al., Mobility, balance and falls in persons with multiple sclerosis, PloS One, 2011;6:e28021.
  120. Coote S, Hogan N, Franklin S, Falls in people with multiple sclerosis who use a walking aid: prevalence, factors, and effect of strength and balance interventions, Arch Phys Med Rehabil, 2013;94:616–21.
  121. Nilsagard YE, Forsberg AS, von Koch L, Balance exercise for persons with multiple sclerosis using Wii games: a randomised, controlled multi-centre study, Mult Scler, 2013;19:209–16.
  122. Jackson K, Edginton-Bigelow K, Cooper C, Merriman H, A group kickboxing program for balance, mobility, and quality of life in individuals with multiple sclerosis: a pilot study, J Neurol Phys Ther, 2012;36:131–7.
  123. Cortesi M, Cattaneo D, Jonsdottir J, Effect of kinesio taping on standing balance in subjects with multiple sclerosis: A pilot study\m{1}, NeuroRehabilitation, 2011;28:365–72.
  124. Prosperini L, Leonardi L, De Carli P, et al., Visuo-proprioceptive training reduces risk of falls in patients with multiple sclerosis, Mult Scler, 2010;16:491–9.
  125. Schwartz I, Sajin A, Moreh E, et al., Robot-assisted gait training in multiple sclerosis patients: a randomized trial, Mult Scler, 2012;18:881–90.
  126. Goodman AD, Brown TR, Krupp LB, et al., Sustained-release oral fampridine in multiple sclerosis: a randomised, double-blind, controlled trial, Lancet, 2009;373:732–8.
  127. Goodman AD, Brown TR, Edwards KR, et al., A phase 3 trial of extended release oral dalfampridine in multiple sclerosis, Ann Neurol, 2010;68:494–502.

Further Resources

Share this Article
Related Content In Multiple Sclerosis
  • Copied to clipboard!
    accredited arrow-down-editablearrow-downarrow_leftarrow-right-bluearrow-right-dark-bluearrow-right-greenarrow-right-greyarrow-right-orangearrow-right-whitearrow-right-bluearrow-up-orangeavatarcalendarchevron-down consultant-pathologist-nurseconsultant-pathologistcrosscrossdownloademailexclaimationfeedbackfiltergraph-arrowinterviewslinkmdt_iconmenumore_dots nurse-consultantpadlock patient-advocate-pathologistpatient-consultantpatientperson pharmacist-nurseplay_buttonplay-colour-tmcplay-colourAsset 1podcastprinter scenerysearch share single-doctor social_facebooksocial_googleplussocial_instagramsocial_linkedin_altsocial_linkedin_altsocial_pinterestlogo-twitter-glyph-32social_youtubeshape-star (1)tick-bluetick-orangetick-red tick-whiteticktimetranscriptup-arrowwebinar Sponsored Department Location NEW TMM Corporate Services Icons-07NEW TMM Corporate Services Icons-08NEW TMM Corporate Services Icons-09NEW TMM Corporate Services Icons-10NEW TMM Corporate Services Icons-11NEW TMM Corporate Services Icons-12Salary £ TMM-Corp-Site-Icons-01TMM-Corp-Site-Icons-02TMM-Corp-Site-Icons-03TMM-Corp-Site-Icons-04TMM-Corp-Site-Icons-05TMM-Corp-Site-Icons-06TMM-Corp-Site-Icons-07TMM-Corp-Site-Icons-08TMM-Corp-Site-Icons-09TMM-Corp-Site-Icons-10TMM-Corp-Site-Icons-11TMM-Corp-Site-Icons-12TMM-Corp-Site-Icons-13TMM-Corp-Site-Icons-14TMM-Corp-Site-Icons-15TMM-Corp-Site-Icons-16TMM-Corp-Site-Icons-17TMM-Corp-Site-Icons-18TMM-Corp-Site-Icons-19TMM-Corp-Site-Icons-20TMM-Corp-Site-Icons-21TMM-Corp-Site-Icons-22TMM-Corp-Site-Icons-23TMM-Corp-Site-Icons-24TMM-Corp-Site-Icons-25TMM-Corp-Site-Icons-26TMM-Corp-Site-Icons-27TMM-Corp-Site-Icons-28TMM-Corp-Site-Icons-29TMM-Corp-Site-Icons-30TMM-Corp-Site-Icons-31TMM-Corp-Site-Icons-32TMM-Corp-Site-Icons-33TMM-Corp-Site-Icons-34TMM-Corp-Site-Icons-35TMM-Corp-Site-Icons-36TMM-Corp-Site-Icons-37TMM-Corp-Site-Icons-38TMM-Corp-Site-Icons-39TMM-Corp-Site-Icons-40TMM-Corp-Site-Icons-41TMM-Corp-Site-Icons-42TMM-Corp-Site-Icons-43TMM-Corp-Site-Icons-44TMM-Corp-Site-Icons-45TMM-Corp-Site-Icons-46TMM-Corp-Site-Icons-47TMM-Corp-Site-Icons-48TMM-Corp-Site-Icons-49TMM-Corp-Site-Icons-50TMM-Corp-Site-Icons-51TMM-Corp-Site-Icons-52TMM-Corp-Site-Icons-53TMM-Corp-Site-Icons-54TMM-Corp-Site-Icons-55TMM-Corp-Site-Icons-56TMM-Corp-Site-Icons-57TMM-Corp-Site-Icons-58TMM-Corp-Site-Icons-59TMM-Corp-Site-Icons-60TMM-Corp-Site-Icons-61TMM-Corp-Site-Icons-62TMM-Corp-Site-Icons-63TMM-Corp-Site-Icons-64TMM-Corp-Site-Icons-65TMM-Corp-Site-Icons-66TMM-Corp-Site-Icons-67TMM-Corp-Site-Icons-68TMM-Corp-Site-Icons-69TMM-Corp-Site-Icons-70TMM-Corp-Site-Icons-71TMM-Corp-Site-Icons-72