Curr Neurol Neurosci Rep (2014) 14:458 DOI 10.1007/s11910-014-0458-4

MOVEMENT DISORDERS (M OKUN, SECTION EDITOR)

Updates in Medical and Surgical Therapies for Tourette Syndrome Irene A. Malaty & Umer Akbar

# Springer Science+Business Media New York 2014

Abstract Tourette syndrome is a complex neurobehavioral disorder defined by multiple motor and at least 1 vocal tic, persisting over 1 year, waxing and waning in severity, and not explained by another condition. The condition may range from mild nuisance to debilitating and disabling in severity. Management includes counseling and reassurance, behavioral interventions, pharmacologic, and rarely, surgical interventions. Traditionally, alpha-2 agonists and dopamine receptor antagonists have been utilized. In addition, a number of different pharmacotherapies have been implemented in the search for improved management of tics with better tolerability. In rare, severely disabling cases, neuromodulation with deep brain stimulation may be indicated. Optimal brain targets and candidate selection are still in evolution. This article will review the evidence for current medical and surgical therapies with a focus on recent updates. Keywords Tourette . Tic . Treatment . Neuroleptic . Antipsychotic . Alpha-2 receptor agonist . Deep brain stimulation . Movement disorders

Introduction Tourette syndrome (TS) is a complex neuropsychiatric disorder characterized by multiple motors and phonic tics and often complicated by comorbid conditions which can be more disabling than the tics. A tic is a sudden, rapid, recurrent, nonrhythmic motor movement or vocalization. Motor tics This article is part of the Topical Collection on Movement Disorders I. A. Malaty (*) : U. Akbar Department of Neurology, University of Florida, UF Center for Movement Disorders and Neurorestoration, P.O. Box 100236, Gainesville, FL 32610, USA e-mail: [email protected]

can be simple, such as blinking, transient eye deviations, head nods or shrugs, or more complex sequences of patterned motor programs. Phonic or vocal tics are those which produce sound, most commonly sniffing or throat clearing, but potentially squeaks, hums, words, or phrases. These behaviors may initially mask as “normal” movements, allergies, or even seizures. Tics are distinguished in many cases by a premonitory urge or sensory phenomenon that provokes the action, and a sense of relief with completion of the motoric act. Some individuals describe palpable sensory experiences such as a muscle tension needing to be stretched, while others have a nondescript discomfort that is satisfied temporarily by performing the tic. There is typically a component of temporary suppressibility, but often at the cost of eventual rebound of the tics. Tics may be diminished during focused, directed attention and may be exacerbated by stress in some individuals, but tics characteristically wax and wane despite any impact of external factors. Coprophenomena (socially unacceptable vocalizations or gestures—coprolalia and copropraxia), though commonly considered defining of TS by the lay population, are actually rare and occur in a minority of individuals. A large international database of 3500 patients observed that 14 % had coprolalia [1]. “Malignant TS” is rare, and has been defined as TS requiring ≥2 emergency room visits or at least 1 hospitalization. Self-injurious tic behavior, injuries from tics, aggressive, or suicidal behaviors contribute. At 1 specialty referral center, only 5 % of patients met “malignant” criteria [2]. The TS eponym was coined by Charcot after his resident Georges Albert Édouard Brutus Gilles de la Tourette described 9 cases of individuals with typical symptoms in 1885 [3]. The Diagnostic and Statistical Manual of Mental Disorders (DSM)-IV criteria require onset in childhood (1 year) did not detect a single case [41]. Akathisia and neuroleptic malignant syndrome are additional risks. Appropriate counseling regarding risks, alternatives, and potential benefit is critical. Haloperidol Haloperidol is a butyrophenone [35]. It is the neuroleptic with the longest experience in TS, and 1 of the 2 FDA approved for TS in adults and children in the U.S. Haloperidol has been studied against pimozide and against pimozide and placebo (twice) in RCTs, as well as against clonidine, fluphenazine, and other agents. It consistently demonstrates efficacy in reducing tics in adults and children, but with fatigue, sedation, and extrapyramidal side effects [30••, 32••]. Haloperidol has demonstrated slight superiority to pimozide in efficacy but with lower tolerability and more serious side events in a double-blind placebo-controlled trial. Of note, dose ranges considered in that trial were wide (2–20 mg/day haloperidol, 2–48 mg/day pimozide), and not necessarily typical for those

458, Page 4 of 13

Curr Neurol Neurosci Rep (2014) 14:458

used in routine practice [32••]. Dosing more typically starts 0.25–0.5 mg daily and therapeutic range is 0.75–5 mg daily or divided bid [35••, 36]. Keeping dose to minimum necessary is advisable to reduce risk of adverse events.

gynecomastia has received media attention, although that risk is not unique to risperidone.

Pimozide

Quetiapine antagonizes D2 receptors but also 5HT1a and 5HT2, histamine H1, and alpha 1 and 2 adrenergic receptors. Small open label and retrospective pediatric studies have shown tic improvement using doses ranging approximately 50–300 mg [35••, 46, 47]. Olanzapine binds D1 and D2 receptors, 5HT 2a and 2c, D2, D4, and alpha1 adrenergic receptors [35••]. Tic reduction over placebo has been shown in open label pediatric studies and a double-blind cross over 52 week adult study vs low dose (2– 4 mg/day) pimozide [35••, 48]. More recently, an open label 6 week study of 12 youth demonstrated significant tic reduction and also improved ADHD symptoms, but in this short study mean weight gain was about 4 kg. Starting dose is 2.5 mg, with target range 5–10 mg total per day [30••, 35••].

Pimozide is a diphenylbutylpiperidine derivative that antagonizes D2 receptors [35••]. It has demonstrated efficacy for tic reduction against placebo and has been studied against haloperidol in multiple studies demonstrating equivalency, slight inferiority, or superiority in different studies. It has also been studied against risperidone with equivalent benefit and risk profile [30••, 31••, 32••]. Typical starting dose is 0.5–1 mg and therapeutic range is 2–4 mg (up to 10 mg) [35••, 36]. Prolonged QT interval is a particular risk, especially if paired with other medications that contribute further risk. It is advisable to obtain a baseline and postinduction EKG as risk of sudden cardiac death is reported [35••]. A Cochrane review for pimozide in TS has been published , including 162 patients in 6 trials (age 7–53), concluding it is an effective option but longer trials are needed [42]. Fluphenazine Fluphenazine antagonizes D1 and D2 dopamine receptors. In addition to smaller open label studies reporting efficacy with improved tolerability over haloperidol [43], a retrospective study of 268 patients with TS (ages 4–70) demonstrated marked to moderate improvement in 80.5 % of the patients, at an average optimal dose of roughly 3 mg daily (range 0.5– 12 mg). Drowsiness and fatigue were the most common side effects, with no reports of tardive dyskinesia (average 2.6 year duration; .01–16.8 years) [44•]. Starting dose range is 0.5– 1 mg, with therapeutic range 2–5 mg daily [35••]. QT prolongation is a risk with this medication as well, and narrow angle glaucoma is a contraindication. Atypical Neuroleptics Risperidone Risperidone is the most well-studied atypical neuroleptic for TS. It works at D2 receptors but also 5HT2, histamine H1, alpha 1 adrenergic, D3, and D4 receptors [35••]. It has demonstrated tic reduction against placebo and efficacy at least equal to pimozide in head to head trials. In the U.S. it has an additional indication for irritability associated with autism spectrum disorder. In 2012 a meta-analysis was published concluding efficacy similar to traditional agents [45•]. Starting dose is 0.25–0.5 mg with target range typically 2– 4 mg daily total [35••]. QT prolongation is a concern, as well as the other typical risks of neuroleptics. Recently

Quetiapine

Aripiprazole Aripiprazole is a D2 receptor antagonist as well as a partial agonist of D2 receptors and a 5-HT1-A and 5-HT2-A antagonist [36]. It has recently gained favor as an atypical neuroleptic that may be better tolerated but adequately potent to treat TS, and evidence is mounting to support its use. An open label study of 28 consecutive TS patients with comorbid ADHD found that motor and vocal tics improved by 42.5 % and 47.9 %, respectively, using YGTSS. In that study, attention (ADHD-Rating Scale IV) also improved by 22.5 % [49]. A retrospective review of 100 cases at 1 specialty center reported that patients (27 +/–11.5 years) had been treated with doses (5–45 mg daily). Eighty-two had significant benefit in tic severity and 48 patients followed to 1 year had lasting benefit, although 31 % discontinued for side effects or lack of efficacy [50]. Enduring benefit was demonstrated in another study of 20 adult patients treated with a mean dose of 11.8 mg daily for up to 56 months [51]. Most recently, a 10-week multicenter RCT with 61 patients (6–18 y old) demonstrated improvement in total tic score on YGTSS and TS CGIS. Aripiprazole was well-tolerated. Mean weight gain was 1.6 kg compared with 0.2 kg on placebo [52•]. In a multicenter controlled trial of 195 children with TS in China, aripiprazole led to tic reduction of 29 points on YGTSS, equivalent to the comparator, the D2 and D3 receptor blocker tiapride [53]. Ziprasidone Ziprasidone is a D2 receptor antagonist that also binds 5HT2, alpha 1 adrenergic, and H1 histamine receptors. A pilot study involving 28 pediatric patients demonstrated efficacy over

Curr Neurol Neurosci Rep (2014) 14:458

Page 5 of 13, 458

Dopamine Depletor

have compared topiramate with haloperidol (12) or tiapride (2) with similar benefit [30••, 60•]. Duration was variable (days to 1 year) as have been the doses utilized (1–9 mg/kg/ day for children, 50–200 day for adults) and outcome measures, but benefit did occur. A retrospective study of 41 patients treated at a single center found that average efficacy was “moderate improvement” using a global impression of response (none/worse, mild, moderate, or marked) [61]. A RCT of 29 patients (children and adults) demonstrated improved total tic score on YGTSS of 14.3 vs 5.0 on placebo with mean daily dose 118 mg [62]. Drowsiness, loss of appetite and weight, and cognitive side effects occurred. Paresthesias, slight increase in risk of kidney stones and glaucoma need to be discussed when prescribing. This agent may be a consideration for patients who have experienced excessive weight gain on neuroleptics, or those with comorbid migraine or seizure disorders.

Tetrabenazine

GABAergic Agents

Tetrabenazine works by reversibly inhibiting human vesicular monoamine transporter type 2 (VMAT) and decreasing presynaptic monoamine uptake into vesicles, depleting monoamine stores. It also has mild postsynaptic dopamine receptor antagonism [35••]. Evidence for tic reduction comes from several open label and retrospective studies. Retrospective reviews have suggested that 77 %–80 % of patients had improvement in tics [35••, 56, 57]. Starting dose is 12.5 mg daily, with slow titration to efficacy (50–150 mg/day). Poor CYP2D6 metabolizers may encounter benefit or side effects at lower doses, and CYP2D6 genotyping may be considered particularly when titrating above 50 mg/day. A recent study, however, determined that extensive vs intermediate metabolizers had similar benefit and risk of adverse events, but faster metabolizers take longer to titrate to efficacy [58]. Induction or worsening of depression needs to be monitored, as well as risk of somnolence, hypotension, gastrointestinal upset, and emergence of parkinsonism, or other extrapyramidal syndromes. Weight gain is much less than with neuroleptics; In 1 pediatric study, 0.8 lb/mo were gained vs 1.7 lb/mo over approximately a 2-year study [59].

Clonazepam

placebo and tolerability at mean dose of 28.2 mg (5–40 mg) [54]. QT prolongation is an important risk. Ecopipam Ecopipam is a promising agent with a novel mechanism of selective D1 receptor antagonism. It is proposed that this mechanism might impact the excitatory direct pathway in the basal ganglia, whereas D2 modulation theoretically targets faulty inhibition of inhibitory pathways. In an open label 8-week study of 18 adults, YGTSS total tic scores, motor and vocal tics, and impairment were reduced with no serious adverse events and no impact on weight. Sedation, fatigue, insomnia, and somnolence were the most common side effects [55•].

Other Agents Topiramate Topiramate is an anticonvulsant that inhibits voltage gated sodium channels and has GABAergic as well as glutamate antagonistic effects [35••]. A recent meta-analysis (2013) concluded that quality of evidence was insufficient to support topiramate as a tic-suppression agent [60•]. However, 14 trials

Clonazepam is a benzodiazepine and GABA-A receptor agonist, as well as alpha-2 adrenergic agonist with case reports and small open label trials demonstrating benefit against placebo and clonidine, at doses up to 6 mg daily [35••, 63–65]. Anxiolysis may be a secondary benefit, and this agent may be a choice for anxious patients with tics. Starting dose is 0.25–0.5 mg daily and target 0.5–4 mg/day divided bid or tid [35••]. Sedation, cognitive clouding, hypotension, ataxia, paradoxical agitation, and risk for addiction/dependence must be considered. Baclofen Baclofen is a GABA-B receptor agonist and was studied in a small (n=10) 4-week pediatric (8–14years) double-blind placebo-controlled crossover trial at 60 mg/day. YGTSS total score had nonsignificant improvement relative to placebo (14.7 points) and significant improvement in Clinical Global impression Severity score [66]. In an open label study of 264 children (6–18years), there were significant decreases in motor and vocal tics in 250 patients at 30 mg/day [67]. Somnolence, constipation, and nausea were among common side effects reported. Withdrawal seizures or psychosis can occur with abrupt cessation. Dose ranges typically employed are 10–60 mg daily starting 5–10 mg tid and titrating as needed [30••, 35••]. Dopamine Agonists The selective nonergot ropinirole 0.25–0.5 mg bid had shown tic reduction in a small open-label trial of 15 children [68], and pergolide has been shown to improve tic severity in 2 trials [69,

458, Page 6 of 13

70], but the latter was withdrawn from the market in the U.S. due to valvular heart disease risk. Subsequently, a multicenter randomized placebo-controlled trial involving 63 children and adolescents found no benefit of pramipexole for tics, but some potential impact on ADHD based on DuPaul scores [71].

Curr Neurol Neurosci Rep (2014) 14:458

Levetiracetam Levetiracetam has been compared with clonidine in a double-blind, crossover study [83] and had no positive impact on tics (YGTSS total tic score, total score, or CGIS behavioral measures) [84].

Cannabinoids Other Agents A Cochrane review exists for Delta-9-tetrahyrdrocannabinol (Delta 9-THC) including 28 participants and 2 studies (1 double-blind crossover study and 1 double-blind parallel group study), doses of 5–10 mg/day. There were small improvements reported in some of the outcome measures used. Demonstrable benefit did not balance risk [72–74]. Nicotinic Agents Nicotine Nicotine, an agonist of nicotinic acetylcholine receptors, has been shown to potentiate the benefit of neuroleptics in reducing tics in open-label studies and in controlled trials of both nicotine gum and patches [75, 76]. Although initially there was suggestion that benefit could last up to weeks [77], double-blind placebo controlled trial of a single transdermal dose of nicotine failed to reach significant improvement in primary outcomes of tics. Commonly, nausea, vomiting, or headache were side effects [32••, 77]. Mecamylamine Mecamylamine is a nonselective antagonist of nicotinic acetylcholine receptors that failed to show efficacy over placebo in a randomized double-blind trial of 61 pediatric patients (only 38 completed the study) [78]. Botulinum Toxin Botulinum Toxin inhibits acetylcholine release at the neuromuscular junction injection by being internalized at the presynaptic membrane, cleaving a SNARE protein called SNAP 25, and preventing docking of acetylcholine vesicles. Botulinum toxin injection leads to temporary relaxation of muscles and has been studied as a means to reduce motor and vocal tics in open label studies [79–81]. One RCT demonstrated reduced number of tics/minute and reduced urge to tic [82]. Potential side effects are dependent on injection site, but include ptosis, dysphagia, weakness, hypophonia, and evolution of tics into other manifestations. The role is likely limited to focal and particularly bothersome tics, but may be 1 more tool in the treatment arsenal [82].

Other agents have been explored, including case reports of tic remission with clozaril (including 1 patient with comorbid schizophreniform disorder) [85–87]. Risk of agranulocytosis and minimal data make this less appealing. Atomoxetine was shown to improve both attention and tics (total YGTSS score) over placebo and reached statistical significance at the endpoint (though not at interval time points where placebo improved as well) [88]. Metoclopramide showed 55 % total tic reduction in an open label study of 10 patients, and 39 % reduction (vs 13 % on placebo) in an 8-week RCT of 27 young patients with chronic tic or TS [89, 90]. Longer term studies with close safety monitoring were recommended.

Deep Brain Stimulation Brief Historical Perspective Given the shortcomings in efficacy and risks of chronic therapy with current pharmacologic options, there has been a hunger for more aggressive and innovative therapies to treat severe TS. The similarity of tics to other extrapyramidal hyperkinetic movements led to investigation of surgical approaches. In 1970, Hassler and Dieckman [91] reported 70 %–100 % improvement in tics of 3 patients with ablation of thalamic targets. Ablative procedures over the next 2 decades were retrospectively studied and intolerable adverse effects were reported in a majority of patients [92], leading to a halt of further expansion of this approach. With the success of DBS in modulating other basal ganglia disorders, the appeal of a modifiable treatment intervention was recognized. In 1999, Vandewalle et al [93] introduced an alternative surgical method using stimulation, rather than ablation, at the junction of the centromedial parafascicular complex (CM-Pf), ventro-oralis nucleus (Vo) and substantia periventricularis (Spv) on the basis of Hassler and Dieckman’s success in lesioning the same focus. This represented the dawn of a new era in the surgical management of medication-refractory TS in the late 1990s. Subsequently interest blossomed in alternative targets including other foci in the thalamus, globus pallidus and ventral striatum.

Curr Neurol Neurosci Rep (2014) 14:458

Brief Review of the Evidence After the initial report of successful tic improvement postDBS of the thalamus, the body of evidence has edged forward by trickles of case series (primarily single-case reports) and small clinical trials, culminating in approximately 100 reported cases thus far. Tic improvement of varying degrees has been reported after DBS and several cases have also reported improvement in the associated comorbidities. Since these outcomes varied by target, we briefly discuss the results achieved with each target. Tic Outcomes Thalamic targets are most commonly chosen and have yielded tic improvement (as measured by YGTSS) ranging from 24 % to 90 % [93, 94••, 95–100]. Tics in the first 3 reported TS patients improved by 72 %–90 % after DBS [93, 100]. In a prospective, double-blind, cross-over trial of 5 patients, Maciunas et al [98] reported a 53 % and 70 % improvement in motor and vocal tics during the randomized phase, respectively, but the benefit decreased to 40 % improvement in motor tics and 21 % worsening in vocal tics during the open label, 3-month follow-up. A single case by Bajwa et al [96]. improved by 66 %. Ackermans et al [94••] reported a 49 % reduction in tics of 6 patients in a doubleblind RCT. A case-series of 18 patients [101] with DBS implantation 2 mm anterior to the traditional thalamic target demonstrated 24 %–79 % tic improvement. Fifteen of the same 18 patients maintained 52 % tic reduction after 2 years [102]. Vernaleken et al [103] targeted the dorsomedial (DM) thalamus which resulted in a 36 % improvement in a single-patient. Mean tic reduction of 70 % was observed at 1-year post-DBS in 3 patients implanted more posteriorly, in the center of the CM-Pf complex [104•]. Most recently, Okun et al [105•] compared the classic continuous stimulation paradigm with scheduled intermittent stimulation in 5 patients and found mean reduction in tics of 19 %. Although the 50 % reduction endpoint criterion was not met, scheduled stimulation was found to be safe and feasible. With the exception of an unsuccessful outcome in a mentally retarded patient [106], DBS of the posteroventral (PV) or anteromedial (AM) globus pallidus internus (GPi) has resulted in 37 %–96 % improvement [106–117]. Martinez-Fernandez et al [114] reported 2 patients implanted in each of these distinct targets of the GPi, and 1 in both targets. The anteromedial target yielded a greater improvement than posteroventral GPi (54 % vs 37 %). Massano et al [117] reported a 60 % tic reduction in a 14-year-old boy and

Page 7 of 13, 458

Shahed et al [111] demonstrated 84 % tic improvement in a 16-year-old boy. Welter et al [115] studied patients who were implanted bilaterally in the CM-Pf and GPi (4 leads) and stimulated each nucleus independently, as well as simultaneously, and found improvement of 78 % with GPi stimulation, 45 % with CM-Pf stimulation and 60 % with simultaneous stimulation of both targets. In response to urgent DBS implantation for cervical cord compression caused by morbid TS, tic reduction of 55 % was observed at 1-year follow-up in a recent case-report [116]. Improvement of 61 %–96 % was seen with stimulation of the globus pallidus externa (GPe) in a caseseries of 7 patients [118]. One patient reported worsening of tics by 17 % with the ALIC/NA target [119], while other cases demonstrated an improvement of 25 %–82 % [120–125]. A parkinsonian patient implanted DBS in the STN reported 97 % coincident improvement of co-existing tics [126]. In summary, the majority of the DBS cases reported to date have documented significant tic reduction. The European Society for the Study of Tourette Syndrome (TSSTS) published guidelines in 2011 after review of 69 cases, 59 (93.7 %) of which reported significant improvement in tics [127••]. However, it must be remembered that reported literature has a bias for successful results, while unsuccessful outcomes may not be eagerly reported. Our search has yielded at least 3 cases [98, 106, 119] in which tics (motor, vocal, or both) were unchanged or worsened after DBS. Additionally, adapting and reintegrating even after tic improvement can be difficult for patients with longstanding social and psychological strain. A suicide attempt could not be avoided in a 42-year-old male who experienced a major depressive episode 4 years after successful DBS therapy [122].

Associated Comorbidity Outcomes Follow-up assessment of associated comorbid disorders, like OCD (measured by YBOCS), ADHD, and selfinjurious behavior (SIB), was reported in some, but not all, of the studies. Using thalamic targets, Maciucna et al [98] reported a trend for improvement in mood, anxiety, and OCD symptoms. Bajwa et al [96] demonstrated a 76 % improvement in OCD in their case-report. The double-blind RCT of 6 patients reported no significant change in comorbidities [94••]. In the long-term follow-up of the first 3 TS patients [93, 100] and a large-case series [101], a subjective improvement in associated comorbidities, including compulsions and mood was noted. The 5 patients reported by Okun et al

458, Page 8 of 13

[105•] had no improvement in depression, mania, OCD, or QOL measures. In the GPi targets, Houeto et al [113] reported cessation of SIB and Shahed et al [111] reported improvement of 69 % in OCD symptoms and 65 % in QOL. A 14-year-old boy experienced significant improvement in OCD, mood, and QOL 2years after DBS of anteromedial GPi [117]. Cases reported of ALIC/NA DBS demonstrated 8 %–64 % improvement in associated comorbidities, including OCD improvement of 64 % and SIB reduction in one [121], 50 % OCD improvement and complete cessation of SIB up to 36 months postimplantation in another [123] and subjective improvement in OCD and SIB in a third [125]. Complications DBS for TS has proven to be relatively safe. Of the 3 major complications reported, 2 resulted from hematomas at tips of the electrodes: 1 patient [109] experienced a permanent but mild reduction in rapidity of alternating hand movements, and the other [128] suffered vertical gaze palsy which improved over the following 6 months. Bilateral subcortical hematomas were observed post-DBS in a patient with coagulopathy [97]. In studying their DBS implantations, Servello et al [129•] noticed a higher rate of postoperative infections of extracranial cables and generator pockets in TS patients compared with other movement disorders (18 % vs 3.7 %). Stimulationrelation adverse effects are as varied as the targets used and respective outcomes. Fatigue, apathy, and lethargy have been reported occasionally with several targets [94••, 95, 99, 100, 109, 112, 120]. Transient visual disturbances [94••, 101], and less commonly, sexual problems [95, 100, 115] have also been reported. Presurgical Assessment The assessment for DBS candidacy in TS should be a team approach, as with other movement disorders. After confirming a diagnosis, a comprehensive assessment by an experienced multidisciplinary team including a neurologist, psychiatrist, neuropsychologist, and a functional neurosurgeon is essential to a successful outcome. A thorough assessment of the tics and associated behavioral comorbidities should be performed with direct observation and video-recording. The impairment of QOL should be primarily by tics [95, 127••], since comorbidities may not respond. Recommendations for presurgical assessment have been detailed in several published guidelines including the Tourette Syndrome Association (American) [130], a Dutch-Flemish group [95], an Italian group [131], and most recently by the European Society for the Study of Tourette Syndrome (ESSTS) [127••] and the UK group [132]. The

Curr Neurol Neurosci Rep (2014) 14:458

variations in these guidelines highlight the lack of uniformity in the practice of DBS in TS. Experts agree that the diagnosis of TS should rely on the DSM IV criteria or assessment by an experienced clinician. Two groups [95, 132] suggest the additional use of the Diagnostic Confidence Index. A total score of ≥35/50 on the YGTSS meets the symptom severity criteria for all groups [127••, 130–132] except 1 [95], which uses the cutoff of >20. Most groups specify that severity criteria should be met for at least 12 months [127••, 130, 132] and that tics should be the primary symptom affecting QOL (not comorbidities) [95, 127••]. Presence of significant functional and social impairment is unanimously a criterion. The criteria for treatmentrefractoriness vary. Partial or no response, or intolerance to 3 medications is generally considered refractory [95, 127••, 130, 132]. Whether the 3 medications must be from different classes is debated. A thorough assessment and optimal treatment of comorbid conditions is essential to establish DBS candidacy. Implementation of behavioral therapy, or at least evaluation for its suitability, is emphasized. Comprehensive evaluation of tics and comorbid symptoms should include validated measures for tic severity, such as the YGTSS, an OCD scale (YaleBrown Obsessive-Compulsive Scale), an anxiety scale (such as State-Trait Anxiety Inventory), and depression index (Beck Depression Inventory). Other associated symptoms, including ADHD, anger control, and conduct disorders, can be detected by a detailed interview and the use of DSM-IV criteria checklist. The TS-quality of life scale (TS-QOL) [133] is a disease-specific measure incorporating 4 domains of QOL: psychological, physical (ADLs), OCD, and cognitive. Since spontaneous regression of symptoms in TS is common and predicting which patients will improve is difficult, determining an optimal age for intervention is challenging. DBS surgery at a young age might expose a patient to unnecessary risks, particularly when remission may have been the natural course, while delaying surgery for an otherwise qualified candidate might cause unnecessary strain on QOL and patient’s psychology, hindering learning, social integration, emotional growth, and productivity [134, 135]. Groups with earlier guidelines [95, 130] suggested age for DBS >25 years but more recent publications have suggested lower cutoffs (>18 years [127••, 131], >20 years [132]). Increasing experience demonstrating safety and feasibility of DBS in TS has likely contributed to the expansion of the criteria to younger patients. Rare cases have been reported in even younger patients: a 14-year-old [117], 2 16-year-old [106, 111], and 2 17-year-old [104•] boys. Since DBS in TS is still considered experimental, surgery for pediatric patients should be considered on a case-by-case basis with involvement of the young patient and perhaps a member of the ethics board.

Curr Neurol Neurosci Rep (2014) 14:458

Page 9 of 13, 458

Fig. 1 Potential therapeutic targets of DBS for TS. Figure is not drawn to scale. ALIC anterior limb internal capsule

Target Selection Until now, 9 targets have been used, individually or in combination, for the treatment of TS: 4 within the thalamus (CM-Pf complex [93, 94••, 96–98, 100, 105•], 2 mm anterior [101] or 4 mm posterior [104•], DM nucleus [103]), 2 within the GPi (posteroventral [106–111, 114] and anteromedial [112–117]), GPe [118], NA/ALIC [99, 119–121, 123–125], and STN [126] (Fig. 1). Targets within the thalamus have been used most frequently, followed by GPi, NA/ALIC, GPe and STN in decreasing frequency. Judging the superiority of any 1 target over another is challenging since the inclusionary criteria and outcomes vary throughout the literature. However, it appears that although improvement in tics has been reported with all targets, improvement in associated comorbidities is more frequently reported with thalamic targets. As more data becomes

Table 1 Factors to consider before DBS in Tourette Syndrome • Diagnosis confirmation • Tic severity and associated disability • Medication refractoriness • Adequateness of therapeutic trials- duration of therapies and titration or intolerance • Age • Reasonable expectations Red flags • Secondary tics • Severe psychiatric comorbidities • Anticipated poor compliance • Poor access to follow-up care for programming and battery surveillance • Significant brain imaging abnormalities • Compulsive picking- risk for hardware infection • General surgical contraindications-cardiovascular, pulmonary, or hematologic disorders

available potential approach comes in

about the efficacy of each target as well as distinct side effect profiles, a more tailored can be recommended to achieve better outindividual patients.

Other Considerations As with DBS in other movement disorders, the risks, benefits and alternative therapies must be discussed with each candidate. The potential of failed DBS surgery and possibility of encountering adverse effects or worsening symptoms must be effectively communicated. Presurgical counseling and setting appropriate expectations is critical to achieving an optimal surgical outcome. Patient or caregiver noncompliance can result in a failed surgery, and thus, should be considered a contraindication for DBS. Other potential contraindications include abnormalities on brain MRI, poor general medical condition, and uncontrolled severe psychiatric disorder (Table 1). Compulsive picking in particular has to be carefully considered as a potential risk for infection. In some cases, chronic social isolation and environmental aspects remain destructive even after improvement of tics. In these scenarios, a case-bycase approach with the help of a multidisciplinary team should be utilized.

Conclusions Tourette syndrome is a complex and fascinating neuropsychiatric disorder. Medical and surgical therapies, when indicated, may improve quality of life but with risks of side effects that must be weighed against impact of tics on well-being. Research is desperately needed to continue to advance the care of individuals with TS.

458, Page 10 of 13 Compliance with Ethics Guidelines Conflict of Interest Umer Akbar declares no conflict of interest. Irene A. Malaty has received speaker honoraria and paid travel expenses from the Tourette Syndrome Association for participation in TSA International Consortium for Genetics study. Human and Animal Rights and Informed Consent This article does not contain any studies with human or animal subjects performed by any of the authors.

References Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance 1.

2. 3. 4. 5.

6.

7.

8.

9.

10.

11. 12.

13. 14.

15.

Freeman RD, Fast DK, Burd L, Kerbeshian J, Robertson MM, Sandor P. An international perspective on Tourette syndrome: selected findings from 3,500 individuals in 22 countries. Dev Med Child Neurol. 2000;42(7):436–47. Cheung MY, Shahed J, Jankovic J. Malignant Tourette syndrome. Mov Disord Off J Mov Disord Soc. 2007;22(12):1743–50. Kramer H, Daniels C. Pioneers of movement disorders: Georges Gilles de la Tourette. J Neural Transm. 2004;111(6):691–701. American Psychiatric Association. Electronic DSM-IV-TR plus. Washington, D.C.: American Psychiatric Association; 2000. Roessner V, Ludolph AG, Muller-Vahl K, Neuner I, Rothenberger A, Woitecki K, et al. DSM-5 Kommentar: Tourette Syndrom und andere Tic-Storungen im DSM-5. Z Kinder Jugendpsychiatrie Psychother. 2014;42(2):129–34. Robertson MM. The prevalence and epidemiology of Gilles de la Tourette syndrome. Part 1: the epidemiological and prevalence studies. J Psychosom Res. 2008;65(5):461–72. Knight T, Steeves T, Day L, Lowerison M, Jette N, Pringsheim T. Prevalence of tic disorders: a systematic review and meta-analysis. Pediatr Neurol. 2012;47(2):77–90. Leckman JF, Zhang H, Vitale A, Lahnin F, Lynch K, Bondi C, et al. Course of tic severity in Tourette syndrome: the first two decades. Pediatrics. 1998;102(1 Pt 1):14–9. Kraft JT, Dalsgaard S, Obel C, Thomsen PH, Henriksen TB, Scahill L. Prevalence and clinical correlates of tic disorders in a community sample of school-age children. Eur Child Adolesc Psychiatr. 2012;21(1):5–13. Bloch MH, Peterson BS, Scahill L, Otka J, Katsovich L, Zhang H, et al. Adulthood outcome of tic and obsessive-compulsive symptom severity in children with Tourette syndrome. Arch Pediatr Adolesc Med. 2006;160(1):65–9. Leckman JF. Tourette's syndrome. Lancet. 2002;360(9345):1577– 86. Mink JW. Neurobiology of basal ganglia and Tourette syndrome: basal ganglia circuits and thalamocortical outputs. Adv Neurol. 2006;99:89–98. Frey KA, Albin RL. Neuroimaging of Tourette syndrome. J Child Neurol. 2006;21(8):672–7. Singer HS, Minzer K. Neurobiology of Tourette's syndrome: concepts of neuroanatomic localization and neurochemical abnormalities. Brain Dev. 2003;25 Suppl 1:S70–84. Rickards H. Functional neuroimaging in Tourette syndrome. J Psychosom Res. 2009;67(6):575–84.

Curr Neurol Neurosci Rep (2014) 14:458 16.

Shapiro AK, Shapiro E. Treatment of Gilles de la Tourette's Syndrome with haloperidol. British J Psychiatr J Ment Sci. 1968;114(508):345–50. 17. Paschou P. The genetic basis of Gilles de la Tourette Syndrome. Neurosci Biobehav Rev. 2013;37(6):1026–39. 18. Scharf JM, Yu D, Mathews CA, Neale BM, Stewart SE, Fagerness JA, et al. Genome-wide association study of Tourette's syndrome. Mol Psychiatry. 2013;18(6):721–8. 19. Abelson JF, Kwan KY, O'Roak BJ, Baek DY, Stillman AA, Morgan TM, et al. Sequence variants in SLITRK1 are associated with Tourette's syndrome. Science. 2005;310(5746):317–20. 20.• Ercan-Sencicek AG, Stillman AA, Ghosh AK, Bilguvar K, O'Roak BJ, Mason CE, et al. L-histidine decarboxylase and Tourette's syndrome. N Engl J Med. 2010;362(20):1901–8. In a 2-generation pedigree, the authors identified a novel mutation in the gene encoding L-histidine decarboxylase, the enzyme for the ratelimiting step in histamine synthesis. This brought to light a potential role of histamine transmission in TS pathogenesis. 21. Lei J, Deng X, Zhang J, Su L, Xu H, Liang H, et al. Mutation screening of the HDC gene in Chinese Han patients with Tourette syndrome. Am J Med Genet Part B, Neuropsychiatr Genet Off Publ Int Soc Psychiatr Genet. 2012;159B(1):72–6. 22. Karagiannidis I, Dehning S, Sandor P, Tarnok Z, Rizzo R, Wolanczyk T, et al. Support of the histaminergic hypothesis in Tourette syndrome: association of the histamine decarboxylase gene in a large sample of families. J Med Genet. 2013;50(11): 760–4. 23.• Castellan Baldan L, Williams KA, Gallezot JD, Pogorelov V, Rapanelli M, Crowley M, et al. Histidine decarboxylase deficiency causes tourette syndrome: parallel findings in humans and mice. Neuron. 2014;81(1):77–90. HDC gene knockout mice developed ticlike stereotypies that responded to haloperidol, and other abnormalities of dopamine transmission potentially relevant to TS. 24. Krauss JK, Jankovic J. Severe motor tics causing cervical myelopathy in Tourette's syndrome. Movement disorders : official journal of the Movement Disord Soc. 1996;11(5):563–6. 25. Lehman LL, Gilbert DL, Leach JL, Wu SW, Standridge SM. Vertebral artery dissection leading to stroke caused by violent neck tics of Tourette syndrome. Neurology. 2011;77(18):1706–8. 26. Piacentini J, Woods DW, Scahill L, Wilhelm S, Peterson AL, Chang S, et al. Behavior therapy for children with Tourette disorder: a randomized controlled trial. JAMA. 2010;303(19):1929–37. 27. Wilhelm S, Peterson AL, Piacentini J, Woods DW, Deckersbach T, Sukhodolsky DG, et al. Randomized trial of behavior therapy for adults with Tourette syndrome. Arch Gen Psychiatry. 2012;69(8):795–803. 28. Scahill L, Woods DW, Himle MB, Peterson AL, Wilhelm S, Piacentini JC, et al. Current controversies on the role of behavior therapy in Tourette syndrome. Mov Disord Off J Mov Disord Soc. 2013;28(9):1179–83. 29.• McGuire JF, Piacentini J, Brennan EA, Lewin AB, Murphy TK, Small BJ, et al. A meta-analysis of behavior therapy for Tourette Syndrome. J Psychiatr Res. 2014;50:106–12. This meta-analysis identified 8 RCTs involving 438 participants. Medium to large effect sizes were reported, with a number needed to treat of 3. The authors concluded that benefit of behavioral therapy approximated that of anti-psychotics in meta-analyses. 30.•• Pringsheim T, Doja A, Gorman D, McKinlay D, Day L, Billinghurst L, et al. Canadian guidelines for the evidence-based treatment of tic disorders: pharmacotherapy. Can J Psychiatry. 2012;57(3):133–43. This is a concise, comprehensive review of literature for treatment in TS. 31.•• Waldon K, Hill J, Termine C, Balottin U, Cavanna AE. Trials of pharmacological interventions for Tourette syndrome: a

Curr Neurol Neurosci Rep (2014) 14:458 systematic review. Behav Neurol. 2013;26(4):265–73. This article has quick reference tables for trials done in TS. 32.•• Roessner V, Plessen KJ, Rothenberger A, Ludolph AG, Rizzo R, Skov L, et al. European clinical guidelines for Tourette syndrome and other tic disorders. Part II: pharmacological treatment. Eur Child Adolesc Psychiatr. 2011;20(4):173–96. This is a comprehensive review of the literature in pharmacotherapy for TS. 33. Du YS, Li HF, Vance A, Zhong YQ, Jiao FY, Wang HM, et al. Randomized double-blind multicentre placebo-controlled clinical trial of the clonidine adhesive patch for the treatment of tic disorders. Aust New Zealand Jf Psychiatry. 2008;42(9):807–13. 34. Group TsSS. Treatment of ADHD in children with tics: a randomized controlled trial. Neurology. 2002;58(4):527–36. 35.•• Singer HS. Treatment of tics and tourette syndrome. Curr Treat Options Neurol. 2010;12(6):539–61. This is a good clinically focused paper describing a rational approach to treatment of tics in TS. 36. Hartmann A, Worbe Y. Pharmacological treatment of Gilles de la Tourette syndrome. Neurosci Biobehav Rev. 2013;37(6):1157–61. 37. Scahill L, Chappell PB, Kim YS, Schultz RT, Katsovich L, Shepherd E, et al. A placebo-controlled study of guanfacine in the treatment of children with tic disorders and attention deficit hyperactivity disorder. Am J Psychiatry. 2001;158(7):1067–74. 38. Chappell PB, Riddle MA, Scahill L, Lynch KA, Schultz R, Arnsten A, et al. Guanfacine treatment of comorbid attentiondeficit hyperactivity disorder and Tourette's syndrome: preliminary clinical experience. J Am Acad Child Adolesc Psychiatry. 1995;34(9):1140–6. 39. Boon-yasidhi V, Kim YS, Scahill L. An open-label, prospective study of guanfacine in children with ADHD and tic disorders. J Med Assoc Thail = Chotmaihet thangphaet. 2005;88 Suppl 8: S156–62. 40. Cummings DD, Singer HS, Krieger M, Miller TL, Mahone EM. Neuropsychiatric effects of guanfacine in children with mild tourette syndrome: a pilot study. Clin Neuropharmacol. 2002;25(6):325–32. 41. Muller-Vahl KR, Krueger D. Does Tourette syndrome prevent tardive dyskinesia? Mov Disord Off J Mov Disord Soc. 2011;26(13):2442–3. 42. Pringsheim T, Marras C. Pimozide for tics in Tourette's syndrome. The Cochrane database of systematic reviews. 2009;2, CD006996. 43. Singer HS, Gammon K, Quaskey S. Haloperidol, fluphenazine and clonidine in Tourette syndrome: controversies in treatment. Pediatr Neurosci. 1985;12(2):71–4. 44.• Wijemanne S, Wu LJ, Jankovic J. Long-term efficacy and safety of fluphenazine in patients with Tourette syndrome. Mov Disord Off J Mov Disord Soc. 2014;29(1):126–30. This retrospective review of 268 patients treated with fluphenazine at 1 center reported tic benefit in 80.5%with no cases of tardive dyskinesia. 45.• Cheng W, Lin L, Guo S. [A Meta-analysis of the effectiveness of risperidone versus traditional agents for Tourette's syndrome]. Zhong nan da xue xue bao Yi xue ban = Journal of Central South University Medical sciences. 2012;37(4):359–65. Data from 11 RCTs and 741 participants found risperidone to be as effective as traditional agents for TS. 46. Mukaddes NM, Abali O. Quetiapine treatment of children and adolescents with Tourette's disorder. J Child Adolesc Psychopharmacol. 2003;13(3):295–9. 47. Copur M, Arpaci B, Demir T, Narin H. Clinical effectiveness of quetiapine in children and adolescents with Tourette's syndrome : a retrospective case-note survey. Clin Drug Investig. 2007;27(2): 123–30. 48. Onofrj M, Paci C, D'Andreamatteo G, Toma L. Olanzapine in severe Gilles de la Tourette syndrome: a 52-week double-blind

Page 11 of 13, 458

49.

50.

51.

52.•

53.

54.

55.•

56. 57.

58.

59.

60.•

61. 62.

63.

64. 65.

66.

cross-over study vs. low-dose pimozide. J Neurol. 2000;247(6): 443–6. Masi G, Gagliano A, Siracusano R, Berloffa S, Calarese T, Ilardo G, et al. Aripiprazole in children with Tourette's disorder and comorbid attention-deficit/hyperactivity disorder: a 12-week, openlabel, preliminary study. J Child Adolesc Psychopharmacol. 2012;22(2):120–5. Wenzel C, Kleimann A, Bokemeyer S, Muller-Vahl KR. Aripiprazole for the treatment of Tourette syndrome: a case series of 100 patients. J Clin Psychopharmacol. 2012;32(4):548–50. Neuner I, Nordt C, Schneider F, Kawohl W. Effectiveness of aripiprazole in the treatment of adult Tourette patients up to 56 months. Human Psychopharmacol. 2012;27(4):364–9. Yoo HK, Joung YS, Lee JS, Song DH, Lee YS, Kim JW, et al. A multicenter, randomized, double-blind, placebo-controlled study of aripiprazole in children and adolescents with Tourette's disorder. J Clin Psychiatr. 2013;74(8):e772–80. This RCT demonstrated efficacy of aripiprazole in treating TS. Liu ZS, Chen YH, Zhong YQ, Zou LP, Wang H, Sun D, et al. [A multicenter controlled study on aripiprazole treatment for children with Tourette syndrome in China]. Zhonghua er ke za zhi Chinese J Pediatr. 2011;49(8):572–6. Sallee FR, Kurlan R, Goetz CG, Singer H, Scahill L, Law G, et al. Ziprasidone treatment of children and adolescents with Tourette's syndrome: a pilot study. J Am Acad Child Adolesc Psychiatry. 2000;39(3):292–9. Gilbert DL, Budman CL, Singer HS, Kurlan R, Chipkin RE. A D1 receptor antagonist, ecopipam, for treatment of tics in Tourette syndrome. Clin Neuropharmacol. 2014;37(1):26–30. This 8-week, open-label paper reported tic improvement using a novel mechanism of action, D1 receptor antagonism. Kenney C, Jankovic J. Tetrabenazine in the treatment of hyperkinetic movement disorders. Expert Rev Neurother. 2006;6(1):7–17. Porta M, Sassi M, Cavallazzi M, Fornari M, Brambilla A, Servello D. Tourette's syndrome and role of tetrabenazine: review and personal experience. Clin Drug Investig. 2008;28(7):443–59.. Mehanna R, Hunter C, Davidson A, Jimenez-Shahed J, Jankovic J. Analysis of CYP2D6 genotype and response to tetrabenazine. Mov Disord Off J Mov Disord Soc. 2013;28(2):210–5. Ondo WG, Jong D, Davis A. Comparison of weight gain in treatments for Tourette syndrome: tetrabenazine versus neuroleptic drugs. J Child Neurol. 2008;23(4):435–7. Yang CS, Zhang LL, Zeng LN, Huang L, Liu YT. Topiramate for Tourette's syndrome in children: a meta-analysis. Pediatr Neurol. 2013;49(5):344–50. Including 9 trials and 668 patients, benefit similar to haloperidol or tiapride. Quality of trials was poor, leading to reservations against endorsing topiramate despite optimism. Kuo SH, Jimenez-Shahed J. Topiramate in treatment of tourette syndrome. Clin Neuropharmacol. 2010;33(1):32–4. Jankovic J, Jimenez-Shahed J, Brown LW. A randomised, doubleblind, placebo-controlled study of topiramate in the treatment of Tourette syndrome. J Neurol Neurosurg Psychiatry. 2010;81(1): 70–3. Gonce M, Barbeau A. Seven cases of Gilles de la tourette's syndrome: partial relief with clonazepam: a pilot study. Canadian J Neurological Sci Le journal canadien des sciences neurologiques. 1977;4(4):279–83. Kaim B. A case of Gilles de la Tourette's syndrome treated with clonazepam. Brain Res Bull. 1983;11(2):213–4. Merikangas JR, Merikangas KR, Kopp U, Hanin I. Blood choline and response to clonazepam and haloperidol in Tourette's syndrome. Acta Psychiatr Scand. 1985;72(4):395–9. Singer HS, Wendlandt J, Krieger M, Giuliano J. Baclofen treatment in Tourette syndrome: a double-blind, placebo-controlled, crossover trial. Neurology. 2001;56(5):599–604.

458, Page 12 of 13 67.

Awaad Y. Tics in Tourette syndrome: new treatment options. J Child Neurol. 1999;14(5):316–9. 68. Anca MH, Giladi N, Korczyn AD. Ropinirole in Gilles de la Tourette syndrome. Neurology. 2004;62(9):1626–7. 69. Gilbert DL, Sethuraman G, Sine L, Peters S, Sallee FR. Tourette's syndrome improvement with pergolide in a randomized, doubleblind, crossover trial. Neurology. 2000;54(6):1310–5. 70. Gilbert DL, Dure L, Sethuraman G, Raab D, Lane J, Sallee FR. Tic reduction with pergolide in a randomized controlled trial in children. Neurology. 2003;60(4):606–11. 71. Kurlan R, Crespi G, Coffey B, Mueller-Vahl K, Koval S, Wunderlich G, et al. A multicenter randomized placebo-controlled clinical trial of pramipexole for Tourette's syndrome. Mov Disord Off J Mov Disord Soc. 2012;27(6):775–8. 72. Curtis A, Clarke CE, Rickards HE. Cannabinoids for Tourette's Syndrome. The Cochrane database of systematic reviews. 2009;4, CD006565. 73. Muller-Vahl KR, Schneider U, Prevedel H, Theloe K, Kolbe H, Daldrup T, et al. Delta 9-tetrahydrocannabinol (THC) is effective in the treatment of tics in Tourette syndrome: a 6-week randomized trial. J Clin Psychiatr. 2003;64(4):459–65. 74. Muller-Vahl KR, Schneider U, Koblenz A, Jobges M, Kolbe H, Daldrup T, et al. Treatment of Tourette's syndrome with Delta 9tetrahydrocannabinol (THC): a randomized crossover trial. Pharmacopsychiatry. 2002;35(2):57–61. 75. Silver AA, Shytle RD, Philipp MK, Wilkinson BJ, McConville B, Sanberg PR. Transdermal nicotine and haloperidol in Tourette's disorder: a double-blind placebo-controlled study. J Clin Psychiatr. 2001;62(9):707–14. 76. McConville BJ, Sanberg PR, Fogelson MH, King J, Cirino P, Parker KW, et al. The effects of nicotine plus haloperidol compared to nicotine only and placebo nicotine only in reducing tic severity and frequency in Tourette's disorder. Biol Psychiatry. 1992;31(8):832–40. 77. Howson AL, Batth S, Ilivitsky V, Boisjoli A, Jaworski M, Mahoney C, et al. Clinical and attentional effects of acute nicotine treatment in Tourette's syndrome. European Psychiatr J Assoc European Psychiatr. 2004;19(2):102–12. 78. Silver AA, Shytle RD, Sheehan KH, Sheehan DV, Ramos A, Sanberg PR. Multicenter, double-blind, placebo-controlled study of mecamylamine monotherapy for Tourette's disorder. J Am Acad Child Adolesc Psychiatry. 2001;40(9):1103–10. 79. Scott BL, Jankovic J, Donovan DT. Botulinum toxin injection into vocal cord in the treatment of malignant coprolalia associated with Tourette's syndrome. Mov Disord Off J Mov Disord Soc. 1996;11(4):431–3. 80. Kwak CH, Hanna PA, Jankovic J. Botulinum toxin in the treatment of tics. Arch Neurol. 2000;57(8):1190–3. 81. Porta M, Maggioni G, Ottaviani F, Schindler A. Treatment of phonic tics in patients with Tourette's syndrome using botulinum toxin type A. Neurologic Sci Off J Italian Neurologic Soc Italian Soc Clin Neurophysiol. 2004;24(6):420–3. 82. Marras C, Andrews D, Sime E, Lang AE. Botulinum toxin for simple motor tics: a randomized, double-blind, controlled clinical trial. Neurology. 2001;56(5):605–10. 83. Hedderick EF, Morris CM, Singer HS. Double-blind, crossover study of clonidine and levetiracetam in Tourette syndrome. Pediatr Neurol. 2009;40(6):420–5. 84. Smith-Hicks CL, Bridges DD, Paynter NP, Singer HS. A double blind randomized placebo control trial of levetiracetam in Tourette syndrome. Mov Disord Off J Mov Disord Soc. 2007;22(12): 1764–70. 85. Adeloye A, Alhamad A. Gilles de la Tourette syndrome treated effectively with Clozapine. Neurosciences. 2004;9(2):129. 86. Alhamad AM. Gilles de la Tourette syndrome treated effectively with clozapine. Neurosciences. 2003;8(3):198–200.

Curr Neurol Neurosci Rep (2014) 14:458 87. 88.

89.

90.

91.

92.

93.

94.••

95.

96.

97.

98.

99.

100.

101.

102.

103.

Schmider J, Hoff P. Clozapine in Tourette's syndrome. J Clin Psychopharmacol. 1998;18(1):88–9. Spencer TJ, Sallee FR, Gilbert DL, Dunn DW, McCracken JT, Coffey BJ, et al. Atomoxetine treatment of ADHD in children with comorbid Tourette syndrome. J Atten Disord. 2008;11(4):470–81. Acosta MT, Castellanos FX. Use of the "Inverse Neuroleptic" metoclopramide in Tourette syndrome: an open case series. J Child Adolesc Psychopharmacol. 2004;14(1):123–8. Nicolson R, Craven-Thuss B, Smith J, McKinlay BD, Castellanos FX. A randomized, double-blind, placebo-controlled trial of metoclopramide for the treatment of Tourette's disorder. J Am Acad Child Adolesc Psychiatry. 2005;44(7):640–6. Hassler R, Dieckmann G. [Stereotaxic treatment of tics and inarticulate cries or coprolalia considered as motor obsessional phenomena in Gilles de la Tourette's disease]. Rev Neurol. 1970;123(2):89–100. Babel TB, Warnke PC, Ostertag CB. Immediate and long term outcome after infrathalamic and thalamic lesioning for intractable Tourette's syndrome. J Neurol Neurosurg Psychiatry. 2001;70(5): 666–71. Vandewalle V, van der Linden C, Groenewegen HJ, Caemaert J. Stereotactic treatment of Gilles de la Tourette syndrome by high frequency stimulation of thalamus. Lancet. 1999;353(9154):724. Ackermans L, Duits A, van der Linden C, Tijssen M, Schruers K, Temel Y, et al. Double-blind clinical trial of thalamic stimulation in patients with Tourette syndrome. Brain J Neurol. 2011;134(Pt 3): 832–44. This double-blind cross-over study of 8 patients demonstrated a 37 % reduction in tics (on YGTSS) and no change in comorbid conditions. The improvement was sustained at 1 year (49 % reduction). Ackermans L, Temel Y, Cath D, van der Linden C, Bruggeman R, Kleijer M, et al. Deep brain stimulation in Tourette's syndrome: two targets? Mov Disord Off J Mov Disord Soc. 2006;21(5):709– 13. Bajwa RJ, de Lotbiniere AJ, King RA, Jabbari B, Quatrano S, Kunze K, et al. Deep brain stimulation in Tourette's syndrome. Mov Disord Off J Mov Disord Soc. 2007;22(9):1346–50. Idris Z, Ghani AR, Mar W, Bhaskar S, Wan Hassan WN, Tharakan J, et al. Intracerebral haematomas after deep brain stimulation surgery in a patient with Tourette syndrome and low factor XIIIA activity. J Clin Neurosci Off J Neurosurgical Soc Australasia. 2010;17(10):1343–4. Maciunas RJ, Maddux BN, Riley DE, Whitney CM, Schoenberg MR, Ogrocki PJ, et al. Prospective randomized double-blind trial of bilateral thalamic deep brain stimulation in adults with Tourette syndrome. J Neurosurg. 2007;107(5):1004–14. Shields DC, Cheng ML, Flaherty AW, Gale JT, Eskandar EN. Microelectrode-guided deep brain stimulation for Tourette syndrome: within-subject comparison of different stimulation sites. Stereotact Funct Neurosurg. 2008;86(2):87–91. Visser-Vandewalle V, Temel Y, Boon P, Vreeling F, Colle H, Hoogland G, et al. Chronic bilateral thalamic stimulation: a new therapeutic approach in intractable Tourette syndrome. Report of three cases. J Neurosurg. 2003;99(6):1094–100. Servello D, Porta M, Sassi M, Brambilla A, Robertson MM. Deep brain stimulation in 18 patients with severe Gilles de la Tourette syndrome refractory to treatment: the surgery and stimulation. J Neurol Neurosurg Psychiatry. 2008;79(2):136–42. Porta M, Brambilla A, Cavanna AE, Servello D, Sassi M, Rickards H, et al. Thalamic deep brain stimulation for treatmentrefractory Tourette syndrome: two-year outcome. Neurology. 2009;73(17):1375–80. Vernaleken I, Kuhn J, Lenartz D, Raptis M, Huff W, Janouschek H, et al. Bithalamical deep brain stimulation in tourette syndrome is associated with reduction in dopaminergic transmission. Biol Psychiatry. 2009;66(10):e15–7.

Curr Neurol Neurosci Rep (2014) 14:458 104.• Savica R, Stead M, Mack KJ, Lee KH, Klassen BT. Deep brain stimulation in tourette syndrome: a description of 3 patients with excellent outcome. Mayo Clin Proc. 2012;87(1):59–62. Authors report 3 patients implanted DBS more centrally in the CM-Pf complex (posterior to the traditional thalamic target) and demonstrated 60 %–80 % improvement at 1 year. 105.• Okun MS, Foote KD, Wu SS, Ward HE, Bowers D, Rodriguez RL, et al. A trial of scheduled deep brain stimulation for Tourette syndrome: moving away from continuous deep brain stimulation paradigms. JAMA Neurol. 2013;70(1):85–94. Using a newer device with scheduled stimulation, rather than constant, 5 patients were implanted. Safety and feasibility of intermittent stimulation was established but the 50%tic improvement criterion was not met. 106. Dueck A, Wolters A, Wunsch K, Bohne-Suraj S, Mueller JU, Haessler F, et al. Deep brain stimulation of globus pallidus internus in a 16-year-old boy with severe tourette syndrome and mental retardation. Neuropediatrics. 2009;40(5):239–42. 107. Van der Linden C, Colle H, Vandewalle V, Alessi G, Rijckaert D, De Waele L. Successful treatment of tics with bilateral internal pallidum (GPI) stimulation in a 27-year-old male patient with Gilles de la Tourette's syndrome. Mov Disord Off J Mov Disord Soc. 2002;17:S341. 108. Dehning S, Mehrkens JH, Muller N, Botzel K. Therapy-refractory Tourette syndrome: beneficial outcome with globus pallidus internus deep brain stimulation. Mov Disord Off J Mov Disord Soc. 2008;23(9):1300–2. 109. Diederich NJ, Kalteis K, Stamenkovic M, Pieri V, Alesch F. Efficient internal pallidal stimulation in Gilles de la Tourette syndrome: a case report. Mov Disord Off J Mov Disord Soc. 2005;20(11):1496–9. 110. Gallagher CL, Garell PC, Montgomery Jr EB. Hemi tics and deep brain stimulation. Neurology. 2006;66(3):E12. 111. Shahed J, Poysky J, Kenney C, Simpson R, Jankovic J. GPi deep brain stimulation for Tourette syndrome improves tics and psychiatric comorbidities. Neurology. 2007;68(2):159–60. 112. Foltynie T, Martinez-Torres I, Zrinzo L, Joyce E, Cavanna A, Jahanshahi M, et al. Improvement in vocal and motor tics following DBS of motor GPI for Tourette syndrome, not accompanied by subjective improvement in quality of life—a case report. Mov Disord. 2009;24:S497–8. 113. Houeto JL, Karachi C, Mallet L, Pillon B, Yelnik J, Mesnage V, et al. Tourette's syndrome and deep brain stimulation. J Neurol Neurosurg Psychiatry. 2005;76(7):992–5. 114. Martinez-Fernandez R, Zrinzo L, Aviles-Olmos I, Hariz M, Martinez-Torres I, Joyce E, et al. Deep brain stimulation for Gilles de la Tourette syndrome: a case series targeting subregions of the globus pallidus internus. Mov Disord Off J Mov Disord Soc. 2011;26(10):1922–30. 115. Welter ML, Mallet L, Houeto JL, Karachi C, Czernecki V, Cornu P, et al. Internal pallidal and thalamic stimulation in patients with Tourette syndrome. Arch Neurol. 2008;65(7):952–7. 116. Huasen B, McCreary R, Evans J, Potter G, Silverdale M. Cervical myelopathy secondary to Tourette's syndrome managed by urgent deep brain stimulation. Mov Disord Off J Mov Disord Soc. 2014;29(4):452–3. 117. Massano J, Sousa C, Foltynie T, Zrinzo L, Hariz M, Vaz R. Successful pallidal deep brain stimulation in 15-year-old with Tourette syndrome: 2-year follow-up. J Neurol. 2013;260(9): 2417–9. 118. Vilela Filho O, Ragazzo P, Souza J, Silva D, Oliveira P, Piedmonte F, et al. Bilateral GPE-DBS for Tourette syndrome: a double-blind prospective controlled study of seven patients. ASSFN (American Society for Stereotactic and Functional Neurosurgery) Abstract 2010 biennial meeting Bridging the Future of Neurosurgery; New York, NY 2010.

Page 13 of 13, 458 119.

Burdick A, Foote KD, Goodman W, Ward HE, Ricciuti N, Murphy T, et al. Lack of benefit of accumbens/capsular deep brain stimulation in a patient with both tics and obsessive-compulsive disorder. Neurocase. 2010;16(4):321–30. 120. Flaherty AW, Williams ZM, Amirnovin R, Kasper E, Rauch SL, Cosgrove GR, et al. Deep brain stimulation of the anterior internal capsule for the treatment of Tourette syndrome: technical case report. Neurosurgery. 2005; (4 Suppl):E403; discussion E. 121. Kuhn J, Lenartz D, Mai JK, Huff W, Lee SH, Koulousakis A, et al. Deep brain stimulation of the nucleus accumbens and the internal capsule in therapeutically refractory Tourette-syndrome. J Neurol. 2007;254(7):963–5. 122. Neuner I, Halfter S, Wollenweber F, Podoll K, Schneider F. Nucleus accumbens deep brain stimulation did not prevent suicide attempt in tourette syndrome. Biol Psychiatry. 2010;68(4):e19–20. 123. Neuner I, Podoll K, Lenartz D, Sturm V, Schneider F. Deep brain stimulation in the nucleus accumbens for intractable Tourette's syndrome: follow-up report of 36 months. Biol Psychiatry. 2009;65(4):e5–6. 124. Servello D, Sassi M, Brambilla A, Porta M, Haq I, Foote KD, et al. De novo and rescue DBS leads for refractory Tourette syndrome patients with severe comorbid OCD: a multiple case report. J Neurol. 2009;256(9):1533–9. 125. Zabek M, Sobstyl M, Koziara H, Dzierzecki S. Deep brain stimulation of the right nucleus accumbens in a patient with Tourette syndrome. Case report. Neurol Neurochir Pol. 2008;42(6):554–9. 126. Martinez-Torres I, Hariz MI, Zrinzo L, Foltynie T, Limousin P. Improvement of tics after subthalamic nucleus deep brain stimulation. Neurology. 2009;72(20):1787–9. 127.•• Muller-Vahl KR, Cath DC, Cavanna AE, Dehning S, Porta M, Robertson MM, et al. European clinical guidelines for Tourette syndrome and other tic disorders. Part IV: deep brain stimulation. Eur Child Adolesc Psychiatr. 2011;20(4):209–17. This article reviews the 69 cases reported in the literature until 2010 and provides guidelines for patient and target selection, as well as other considerations for DBS in TS. 128. Ackermans L, Temel Y, Bauer NJ, Visser-Vandewalle V, DutchFlemish Tourette Surgery Study G. Vertical gaze palsy after thalamic stimulation for Tourette syndrome: case report. Neurosurgery. 2007;61(5):E1100. 129.• Servello D, Sassi M, Gaeta M, Ricci C, Porta M. Tourette syndrome (TS) bears a higher rate of inflammatory complications at the implanted hardware in deep brain stimulation (DBS). Acta Neurochir. 2011;153(3):629–32. 130. Mink JW, Walkup J, Frey KA, Como P, Cath D, Delong MR, et al. Patient selection and assessment recommendations for deep brain stimulation in Tourette syndrome. Mov Disord Off J Mov Disord Soc. 2006;21(11):1831–8. 131. Porta M, Servello D, Sassi M, Brambilla A, Defendi S, Priori A, et al. Issues related to deep brain stimulation for treatment-refractory Tourette's syndrome. Eur Neurol. 2009;62(5):264–73. 132. Cavanna AE, Eddy CM, Mitchell R, Pall H, Mitchell I, Zrinzo L, et al. An approach to deep brain stimulation for severe treatmentrefractory Tourette syndrome: the UK perspective. Br J Neurosurg. 2011;25(1):38–44. 133. Cavanna AE, Schrag A, Morley D, Orth M, Robertson MM, Joyce E, et al. The Gilles de la Tourette syndrome-quality of life scale (GTS-QOL): development and validation. Neurology. 2008;71(18):1410–6. 134. Eddy CM, Cavanna AE, Gulisano M, Agodi A, Barchitta M, Cali P, et al. Clinical correlates of quality of life in Tourette syndrome. Mov Disord Off J Mov Disord Soc. 2011;26(4):735–8. 135. Eddy CM, Rizzo R, Gulisano M, Agodi A, Barchitta M, Cali P, et al. Quality of life in young people with Tourette syndrome: a controlled study. J Neurol. 2011;258(2):291–301.

Updates in medical and surgical therapies for Tourette syndrome.

Tourette syndrome is a complex neurobehavioral disorder defined by multiple motor and at least 1 vocal tic, persisting over 1 year, waxing and waning ...
443KB Sizes 0 Downloads 3 Views