Abstract
This is the fourth yearly article in the Tourette Syndrome Research Highlights series, summarizing research from 2017 relevant to Tourette syndrome and other tic disorders. The authors briefly summarize reports they consider most important or interesting. The highlights from 2018 article is being drafted on the Authorea online authoring platform, and readers are encouraged to add references or give feedback on our selections using the comments feature on that page. After the calendar year ends, the article is submitted as the annual update for the Tics collection on F1000Research.
Keywords: Tourette syndrome, tic disorders, review, natural history, etiology, pathophysiology
Introduction
This article is meant to disseminate recent scientific progress on Gilles de la Tourette Syndrome (TS).
Methods
We searched PubMed from time to time using the search strategy “("Tic Disorders"[MeSH] OR Tourette NOT Tourette[AU]) AND 2017[PDAT] NOT 1950:2016[PDAT]”. On 06 July 2018 this search returned 212 citations. Colleagues also recommended articles, and we attended medical conferences. We selected material to be discussed in this review subjectively, guided by our judgment of possible future impact on the field.
Results
A number of tic experts contributed to a review article on TS ( Robertson et al., 2017).
Phenomenology and natural history
Schaefer et al. (2017) describe 16 people with TS who had experienced a clinical remission or marked improvement of more than 1 year’s duration, followed by symptomatic worsening as adults, leading them again to seek treatment. On average the “latent period” (the absence or substantial reduction in tics) had lasted 16 years. Seven of them had worse tics when returning for care than they recalled as children. New substance use was reported as a trigger for exacerbation in 5 patients. This report strengthens evidence that even long-lasting symptomatic improvement in tic disorders (TDs) may not always be permanent, and that in fact the typical course of TS “is one of occasional recurrences of mild tics throughout adult life” ( Bruun & Budman, 1997); see also ( Black et al., 2016; Shapiro et al., 1988, p. 188; Stárková, 1990; Singer, 2006).
Regarding natural course and history, the fate of non-tic symptoms in TS has remained less well explored. A large Danish study reported follow-up data 6 years after enrolling 314 children and teenagers with TS, assessing tics and comorbidities (mainly obsessive–compulsive disorder (OCD) and Attention-Deficit/Hyperactivity Disorder (ADHD); N=226 at follow-up) ( Groth et al., 2017). Most patients’ tics improved over time, but almost a quarter of those over age 16 still had severe tics, and only a sixth had no tics. The severity of OCD and ADHD declined significantly during adolescence, suggesting a shift towards so-called “pure” TS with age. Furthermore, the authors expected tic-related impairment to improve with an age-related decline in tic frequency and severity, but surprisingly the impairment score did not reflect the improvement in tics.
A report on 606 patients with a movement disorder starting in childhood produced an estimate for tic onset of 7.4 ± 3.8 years with a mean delay to diagnosis of 9.9 ± 11 years ( Bäumer et al., 2016).
Epidemiology. New and important findings this year involve the previously unappreciated risk of death in TDs. Meier et al. (2017) demonstrated that mortality rates are elevated in TS and other TDs, with or without comorbidities. In a very large epidemiological study, TDs in adults were associated with a four-fold higher risk of suicide, with the risk not explained by other psychiatric illness such as major depression ( Fernández de la Cruz et al., 2017b). These researchers analyzed 7736 TS/chronic tic disorder (CTD) cases in the Swedish National Patient Register over a 44-year period (1969–2013) and compared them with control subjects from the general population. An increased risk of both suicide and attempted suicide was observed in TS/CTD patients, which was not solely dependent on psychiatric comorbidities. Tics that persisted beyond young adulthood significantly predicted completed suicide. Thus, TS/CTD is a serious medical condition that requires careful monitoring of suicidality. The same group also found a 7- to 10-fold higher risk of completed suicide in people with OCD, even after controlling for comorbid diagnoses ( Fernández de la Cruz et al., 2017a).
Martino et al. (2017c) review screening instruments and rating scales for TDs; see also ( Augustine et al., 2017; Martino & Pringsheim, 2017).
Transient environmental effects on tic severity. A study of 45 children with TS supported the typical antecedent–behavior–consequence behavioral psychology model ( Eaton et al., 2017). Specifically, consequences of tics, “such as receiving accommodations or attention from others,” explained significantly more variance in tic severity than did the child’s level of separation anxiety, though the latter was also a significant factor. This study provides supportive evidence for the approach taken by “CBIT-Jr,” a behavior therapy designed for younger children with TS ( Piacentini et al., 2015).
Other. Non-tic symptoms in TS are reviewed by Martino et al. (2017b). Lee et al. (2017) used Taiwan’s National Health Insurance Research Database to compare 1124 newly diagnosed TS patients to controls in a 1:3 match. Sleep disorders were twice as common in TS, and remained significantly higher in TS after accounting for anxiety disorders, which were the comorbid conditions associated with the highest risk. In a sample of 811 TS subjects recruited for a genetics study, hair pulling (3.8%) and skin picking (13.0%) disorders by DSM-5 were surprisingly common ( Greenberg et al., 2018).
Autism spectrum disorders (ASD) comprise an underexplored comorbidity of TD. There are few epidemiological studies on the subject but the prevalence of ASD in children with TDs is estimated at 20% ( Khalifa & von Knorring, 2006). In a large study including patients with TD (n=535) and their family members (n=234), Darrow et al. (2017a) used the Social Responsiveness Scale Second Edition (SRS) to characterize ASD symptoms, and compared them to historical ASD samples. SRS scores in participants with TD were similar to those observed in other clinical samples but lower than in ASD samples. This is mostly but not entirely explained by elevations in the RRB (restricted interests and repetitive behaviors) subscale, which may be indicating tics rather than other stereotypic movements. The presence of OCD was associated with higher scores on the social cognition and RRB subscales. Complex tics and OCD symptoms (repetitive behaviors) can also be hard to discriminate from core ASD symptoms, especially those related to social communication.
Etiology
A national database study found that parents of children with chronic tics had significantly higher rates of psychiatric illness: more than twice as high in mothers and 39% higher in fathers ( Leivonen et al., 2017). The maternal risk, which included a range of psychopathology, was significantly higher than the paternal risk, which comprised primarily OCD and anxiety disorders. Further work will be needed to clarify whether results reflect maternal-specific environmental risks, genetic risks, factors related to parental care-seeking, or (to a modest extent, given the typical ages of onset for various parental disorders identified) parental stress.
Genetics. A large mixed genetic sample yielded two heritable collections of symptoms that cross diagnostic boundaries, here named symmetry (including some other obsessions and compulsions) and disinhibition (including complex verbal tics) ( Darrow et al., 2017b). Whole exome sequencing from over 500 trios identified a clear excess of likely gene-disrupting de novo mutations, and 4 risk genes that were altered by different mutations in multiple probands ( Willsey et al., 2017). Another report described whole exome sequencing in a 3-generation family with TS, using induced pluripotent stem cells converted into neuronal cell types and assessing protein expression levels ( Sun et al., 2018). The PNKD gene product was expressed at a lower rate in family members with TS or OCD.
Pathophysiology
Roger Albin wrote a very thoughtful review of TS as “a disorder of the social decision-making network” ( Albin, 2018). Beste & Münchau (2018) describe tics in the context of the Theory of Event Coding, which describes the brain’s bidirectional pairing of stimulus and movement. They note the salience of urges to most tic patients, and the fact that most tics are individually relatively normal movements, but occur repeatedly and out of context. Similarly, Shafer et al. (2017) focus on the strong connections between movement and sensory function to propose a theory that encompasses both the development of stereotypies as part of typical development and their persistence in maladaptive forms.
Animal models. A special issue on animal models of TS appeared in the Journal of Neuroscience Methods ( Bortolato & di Giovanni, 2017). Articles reviewed gait and sensorimotor function in the D1CT-7 mouse model of TS ( Fowler et al., 2017), stress mediating the timing of abnormal movements in animal models ( Godar & Bortolato, 2017), and chemogenetic and optogenetic models ( Burton, 2017). Deer mice have behavior that has been discussed as a natural model of OCD ( Wolmarans et al., 2018).
Recently postnatal ablation of the TrkB receptor in cells expressing parvalbumin was shown to produce dramatic changes in cortex and cerebellum and “profound hyperactivity, stereotypies, motor deficits and learning/memory defects” ( Xenos et al., 2017). This result may help explain why autopsies in TS show a lower number of parvalbumin-containing interneurons in the striatum, but this model also produces much more substantial neuroanatomical changes than are seen in TS.
A gene identified in human OCD, slc1a1, which codes an excitatory amino acid transporter, was altered in mice to prevent its expression and function ( Zike et al., 2017). The loss of this protein resulted in mice with reduced extracellular dopamine concentrations and reduced movement and stereotypic behavior after challenge with amphetamine or a dopamine D 1 receptor agonist. Restoring the gene’s expression in the midbrain, but not in the striatum, partially rescued the exogenous dopamine-induced stereotypies. This research is important for its direct links to human illness and its anatomical specificity, and lends additional support to testing dopamine D 1 antagonists in TS (see Medication section, below).
New insights from computational modelling. Using the neurophysiological data obtained from a TS animal model of pharmacological striatal disinhibition, Caligiore et al. (2017) proposed a computational model of the basal ganglia-cerebellar-thalamo-cortical system to address the mechanisms of motor tic generation in TS. Overall, the model suggested that interplay between dopaminergic signal and cortical activity triggered the occurrence of a tic and was able to predict the number of tics generated when striatal dopamine increases and when the cortex is externally stimulated. Maia & Conceição (2017) further discussed the role of tonic and phasic dopamine in tics learning and expression. Based on the existing literature of habit formation and reinforcement learning in TS, the authors proposed a model of tics as exaggerated and persistent motor habits reinforced by aberrant, increased phasic dopamine responses. According to this model, tonic dopamine release would serve to amplify the tendency to execute learned tics. The authors also proposed the mechanism of antipsychotics’ action on tics: increased activity of indirect pathway due to antipsychotic administration could result in tic reduction, but at the same time potentially also could increase the propensity for reinforcing tics due to plasticity in the indirect pathway. In contrast, the authors also suggested that low-dose dopamine agonists could decrease both phasic and tonic dopamine and thus reduce both tic learning and tic expression. Both of these reports assume increasing tics with increasing dopamine concentrations, an assumption that seems to contradict observations that tics do not worsen with exogenous levodopa ( Black & Mink, 2000; Gordon et al., 2013) nor improve with development of Parkinson disease ( Kumar & Lang, 1997; Martinez-Torres et al., 2009; Shale et al., 1986). Furthermore, low-dose pergolide in children and adolescents with TS that reduced tic severity suppressed prolactin rather than increasing it, consistent with overall enhancement of dopamine transmission, at least in the hypothalamic-pituitary dopaminergic pathway ( Gilbert et al., 2000a; Gilbert et al., 2000b).
Based largely on available functional anatomical studies, Conceição et al. (2017) also proposed a computational model of premonitory urges in TS. According to this model, premonitory urges and in particular their termination, like termination of other aversive stimuli, might elicit positive prediction errors, supported by phasic dopamine release that would then reinforce tics. The insula may play a central role in aversive feeling associated with premonitory urges and their learned negative value. The insula might send this information via direct or indirect projections to dopamine neurons, which might use it for calculation of the positive prediction errors that occur with termination of the premonitory urge. In short, the authors provide a more detailed neurobiological explanation for the classic model that premonitory urges may strengthen tics through negative reinforcement.
Cognitive function and decision-making in TS. Morand-Beaulieu et al. (2017a) provide an updated and exhaustive review of neuropsychological aspects of TS (compare Eddy et al. (2009)). The review highlighted the slight alteration of social cognition as well as more frequent learning difficulties and disabilities in children with TS. Recent data also seem to confirm the deficit in executive function in TS as indexed by poor performance in continuous performance test and Stroop tests. Interestingly, using longitudinal evaluation of executive function in children with TS, Yaniv et al. (2018) showed that adults with TS showed response inhibition deficits, that tic reduction over time was significantly associated with development of response inhibition, and that former TS patients whose tics had remitted performed as well as, or on some tests better than, healthy control subjects. In contrast, the attentional and memory capacity seems to be impacted by comorbid symptoms more than TS per se. Sample size (n=122) or the healthy control group may explain different results obtained by Abramovitch et al. (2017), who studied treatment response in TS. They concluded that “the finding that significant change in symptom severity of TS/CTD patients is not associated with impairment or change in inhibitory control regardless of treatment type suggests that inhibitory control [as measured by the tests selected] may not be a clinically relevant facet of these disorders in adults.” Given these contradictory results, Morand-Beaulieu et al. (2017b) review the puzzling question of inhibitory control in TS in a recent meta-analysis, and find larger inhibitory deficits in TS + ADHD patients, but this deficit was also present in “pure” TS. This deficit in TS was most prominent in verbal responses, was associated with tics severity as assessed with Yale Global Tic Severity Scale-Total Tics Score (YGTSS-TTS) and was larger in studies that included medicated TS patients.
Salvador et al. (2017) study decisional capacities in TS and specifically the ability to learn from the outcomes of alternative courses of action (known as counterfactual learning). Unmedicated patients with TS showed normal performance on this task, whereas alteration was found in TS patients treated with the dopamine D2-like receptor (D2R) partial agonist aripiprazole, suggesting that modulating D2Rs may impair certain aspects of human reinforcement learning.
The Committee on Research of the American Neuropsychiatric Association published a systematic review on the neurobiology of the premonitory urge in TS ( Cavanna et al., 2017).
Electrophysiology. Brandt et al. (2017) reported on enhanced multi-component behavior in TS, which was also reflected in a smaller P3 event-related potential measured by EEG and potentially related to chronic tic control in these patients. An EEG study during simulated driving found that “mind wandering” may be quantifiable using EEG measures of alpha power and the P3a component of an auditory event-related potential ( Baldwin et al., 2017). Since mind wandering is a defining feature of ADD, in addition to being ubiquitous during repetitive tasks, these measures may prove useful in studying ADHD phenomenology in TDs.
Neuroimaging studies. Polyanska & colleagues. (2017) provide a meta-analysis of task-based neuroimaging reports in TS. Adults with TS show some impairment in lateralized sequential finger tapping movements, and this impairment was compared to fractional anisotropy of white matter tracts connecting primary motor cortex (M1) to M1 and supplementary motor area (SMA) to SMA ( Martino et al., 2017a).
Previous in vivo MRI studies of basal ganglia volume and shape in TS have produced differing results (reviewed in Greene et al. (2017)). A study of 47 children age 8-12 with TS, and controls with or without ADHD, found no significant group differences ( Forde et al., 2017).
A PET study of the microglial activating marker TSPO in OCD found elevated TSPO concentrations in dorsal caudate, orbitofrontal cortex, thalamus, ventral striatum, dorsal putamen, and anterior cingulate cortex ( Attwells et al., 2017). Concentrations in patients were about 1/3 higher than in controls. This report implicates low-level brain inflammation in the pathophysiology of OCD.
Clinical and neuropsychological studies. A study using the alternating serial reaction time task found no impairment of procedural learning in TS or ADHD ( Takács et al., 2017). This result is surprising, given that habit learning on a “weather prediction” task is slower in TS ( Kéri et al., 2002; Marsh et al., 2004), but may indicate that the two tasks engage different learning systems, a conclusion supported by the generally normal cognitive function in TS.
Münchau and colleagues extended their previous work on echopraxia (imitation) to children with TS ( Brandt et al., 2017). Participants were asked to lift either their index or pinky finger when prompted by an auditory tone; simultaneously they were shown a compatible or incompatible visual stimulus. Children with TS were slower overall but thereby gained less interference from the incompatible stimulus. The authors conclude that these results suggest that children with TS may employ “different or additional inhibition strategies” than children without tics. Again, on this task children with TS show superior, not deficient, action inhibition.
Treatment
Ganos et al. (2017) review treatments for TDs in children.
Psychological interventions. Houghton et al. (2017) analyzed their existing data to test the longstanding theory that habituation to premonitory urges during adequate periods of tic suppression is the mechanism by which behavior therapies for tic disorders exerted their beneficial effects. In two previous randomized trials, 126 children and adolescents and 122 adults with TS or a persistent TD had been assigned to Comprehensive Behavioral Intervention for Tics (CBIT) or to education and supportive therapy. The prediction was that urge severity would decrease in CBIT responders. Surprisingly, however, children showed no significant reduction in premonitory urges with treatment, even though CBIT was quite effective in the child study, and declines in urges in adults had no association with clinical improvement or group assignment. The authors conclude that habituation cannot be the underlying process by which CBIT exerts its beneficial effects. This important result warrants further study.
Another report from the same data set examined what clinical features at baseline predicted improvement in tics during treatment ( Sukhodolsky et al., 2017). Importantly, those treated with CBIT improved regardless of medication status, while in the control (supportive therapy) group, tics improved only in those taking medication for tics. Also importantly for the understanding of behavior therapy in TS and patient selection, other psychiatric symptoms, age, sex, family functioning, and expectation of improvement had no significant effects on benefit from therapy. And contrary to some opinions, patients with worse tic severity had significantly better improvement in tics with CBIT. Anxiety disorders and, surprisingly, more severe premonitory urges predicted less improvement.
O’Connor and colleagues published a book describing their combined psychotherapeutic approach to tics ( O’Connor et al., 2017).
Several groups reported efforts to increase dissemination and use of behavior therapy for tics. An internet-based, therapist-guided behavior therapy for tics is being studied in the BiP-TIC project ( Karlsson, 2016). The TicHelper.com internet-based CBIT program went live in late 2017 and received a positive review ( Conelea & Wellen, 2017).
Medication. A group of international experts provided a status report and recommendations for using brain imaging for the rational development of novel psychopharmacological interventions ( Suhara et al., 2017). As an example, a fascinating study in pediatric ADHD showed that fMRI response to a cognitive task (Go/No-Go) strongly predicted better clinical response to methylphenidate than to atomoxetine ( Schulz et al., 2017). As the authors conclude, “These data do not yet translate directly to the clinical setting, but the approach is potentially important for informing future research and illustrates that it may be possible to predict differential treatment response using a biomarker-driven approach.”
Initial results from the first randomized, controlled trial (RCT) with a dopamine D 1 receptor antagonist in pediatric TS were released by the sponsor in January, 2017 ( Chipkin, 2017). These new results supported the positive results from a pilot study in adults with TS ( Gilbert et al., 2014), and suggest a novel treatment mechanism for tics.
In April, the US FDA approved the presynaptic dopamine depleting agent valbenazine (Ingrezza®) for treatment of tardive dyskinesia ( Neurocrine Biosciences, Inc. 2017b). TS is a likely off-label use for the drug, as the company has been conducting studies in children and adults with TS ( ClinicalTrials.gov). The FDA designated valbenazine an orphan drug for pediatric patients with TS ( Neurocrine Biosciences, Inc. 2017a). Another VMAT2 inhibitor, tetrabenazine, has been used for some time in the treatment of TS ( Marsden, 1973; Sweet et al., 1974; Jankovic, 2016). A related compound, deutetrabenazine ( Paton, 2017), showed initial positive results in TS ( Jankovic et al., 2016), and the FDA approved it for treatment of tardive dyskinesia in August ( Business Wire, 2017).
Aripiprazole has become a drug of choice in treating tics and comorbidities in TS over the past decade. However, large scale trials have been missing. Moreover, aripiprazole is not marketed for children and adolescents in many countries, regardless of the indication. Sallee et al. (2017) report on a phase 3, randomized, double-blind, placebo-controlled trial in 133 pediatric patients randomized in a 1:1:1 ratio to low-dose aripiprazole (5 mg/day if <50 kg; 10 mg/day if ≥50 kg), high-dose aripiprazole (10 mg/day if <50 kg; 20 mg/day if ≥50 kg), or placebo for 8 weeks. The primary efficacy endpoint was mean change from baseline to week 8 in the YGTSS-TTS. The Clinical Global Impression-Tourette’s Syndrome improvement score was also evaluated. High-dose aripiprazole was more effective than low-dose aripiprazole, and both were superior to placebo. Importantly, tolerance was overall good and no serious adverse events or deaths occurred, indicating that oral aripiprazole is a safe and effective treatment for tics in children and adolescents. These results provide important reassurance to clinicians who have been using aripiprazole for TS for years now. The placebo response rate (for the Clinical Global Impression scale), though half the response rate in the active treatment groups, was nevertheless surprisingly high (38%).
A small ( N=34) RCT of guanfacine showed no meaningful difference in effects on tic ratings or clinical impressions of improvement between the drug and placebo groups ( Murphy et al., 2017). This result is important and surprising, given that adrenergic α2 agonists have been seen as first-line treatment for TS, especially in TS patients with ADHD ( Hollis et al., 2016). Both these studies show how important RCTs are to clinical care in TS. Speaking of RCTs and guanfacine, a press release reported that extended-release guanfacine showed superiority to placebo in adults with ADHD (discussed here). The importance of this report comes primarily from the fact that data on ADHD treatments are scarcer in adults than in children.
Cannabinoids for TS are increasingly being studied; a brief summary of some of this work appears in Black (2017).
Neurosurgery. A fascinating study demonstrated in mice that interfering electrical “beats” (similar to the beats one hears when tuning one instrument to another) can be used to steer neuron activation to focal sites in the brain without surgical electrode implantation ( Grossman et al., 2017). Much work remains to be done to demonstrate feasibility, safety and efficacy in humans, but this approach potentially could lead to noninvasive, focal brain stimulation.
Welter et al. (2017) performed a randomized, double-blind, controlled trial of deep brain stimulation (DBS) of the posterior and anterior internal globus pallidus for severe TS. The design was very similar to that reported previously by Kefalopoulou et al. (2015), and so were the results. The primary endpoint was the difference in YGTSS score between the beginning and end of the 3 month double-blind period. No significant differences between groups were noted in YGTSS score change between the beginning and the end of the 3 month double-blind period, despite a slight improvement in the stimulated condition. However, after the end of the open stimulation period and further parameter adjustment, results become significant compared to baseline. Also, when turning the stimulators off in a blinded fashion, YGTSS scores increased again to reach near baseline levels. Overall, this study is most important in terms of optimal study design. The stimulator programming period preceding the blinded phases was likely too short and the parameters suboptimal (a choice intended to reduce unblinding). Future studies will need to consider these results carefully.
A London center reported an analysis of GPi DBS data, looking for the “sweet spot” for DBS for tic improvement ( Akbarian-Tefaghi et al., 2017). They report that “a region within the ventral limbic GPi, specifically on the medial medullary lamina in the pallidum at the level of the AC-PC, was significantly associated with improved tics but not mood or OCB outcome.” Two patients with DBS to the fields of Forel experienced a good outcome ( Neudorfer et al., 2017).
TS patients undergoing DBS, independent of the surgical target, usually require high stimulation parameters, leading to short (2–3 years) battery life, meaning frequent and costly battery replacements. Michael Okun and colleagues showed proof of the principle that triggered rather than continuous DBS may be helpful ( Molina et al., 2017). In a single patient with medically refractory TS, a spectral feature in the 5- to 15-Hz band was used as the control signal from bilateral leads in the centromedian-parafascicular (Cm-Pf) region of the thalamus. Significant tic improvement compared to baseline was observed 12 months after the procedure, similar to that obtained with continuous DBS, and resulted in a 63% improvement in the neurostimulator’s projected mean battery life. These so called closed-loop systems are gaining traction in various CNS diseases where DBS is applied, and this case report paves the way in TS.
A report from the DBS group in the Netherlands called attention to side effects over the course of treatment in TS patients with thalamic DBS ( Smeets et al., 2018). A subthalamic nucleus (STN) DBS study in OCD reminds us that DBS can cause side effects; STN stimulation at higher voltages caused chorea-ballismus ( Mulders et al., 2017).
Tics, family and society
Quality of life is lower in parents of children with TS and is more related to factors other than tic severity ( Jalenques et al., 2017). Stewart et al. (2015) previously reported similar results. These studies emphasize the need to assess and treat symptoms other than tics in TS patients, and to care for the whole patient.
Conclusions
2017 has again seen a rise in publications on TS, reflecting the increased interest this field receives, both from clinicians and researchers devoted to this disorder but also from adjoining fields, given the substantial psychopathology associated with TDs.
One important question raised is the natural course of TS and the rate of (non-)remission. Recent data suggest that the optimistic outlook prevalent with regard to tic severity when gliding from adolescence into adulthood might not be completely justified. Here, large longitudinal studies are warranted, and also to better understand the prognostic factors associated with tic remission or persistence. The fate of comorbidities also needs to be better understood, even though data have begun to emerge. We also still face important delays in the diagnosis of TS, and we need to deal with the recently demonstrated increased suicide risk in this condition, which underlines that TDs are far from benign.
Of note, there is a rise in computational models of TS, which provide important leads to understanding its pathophysiology and will likely fuel more targeted treatment approaches for tics. In the therapy field, 2017 has mainly seen studies centered on well-known pharmacological targets such as the dopaminergic system; however, new antidopaminergic medications were studied or came to the market in 2017, and increasing data supports existing off-label prescription practices. Behavioral therapy continues to emerge as a main pillar of tic treatment, with an improved understanding of its mechanisms and response factors. In the surgical field, deep brain stimulation for severe TS cases continues to draw interest; in particular, target choice, length of stimulator programming for optimal outcome, and closed-loop systems are under active investigation.
Overall, the field is active and burgeoning. Breakthroughs are to be expected in the upcoming years, especially with regard to large-scale efforts in the field.
Data availability
No data are associated with this article.
Funding Statement
This work was supported in part by the U.S. National Institutes of Health, grant R01 MH104030.
The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
[version 1; referees: 3 approved]
References
- Abramovitch A, Hallion LS, Reese HE, et al. : Neurocognitive Predictors of Treatment Response to Randomized Treatment in Adults with Tic Disorders. Prog Neuropsychopharmacol Biol Psychiatry. 2017;74:9–14. 10.1016/j.pnpbp.2016.11.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Akbarian-Tefaghi L, Akram H, Johansson J, et al. : Refining the Deep Brain Stimulation Target within the Limbic Globus Pallidus Internus for Tourette Syndrome. Stereotact Funct Neurosurg. 2017;95(4):251–58. 10.1159/000478273 [DOI] [PubMed] [Google Scholar]
- Albin RL: Tourette Syndrome: a Disorder of the Social Decision-Making Network. Brain. 2018;141(2):332–47. 10.1093/brain/awx204 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Attwells S, Setiawan E, Wilson AA, et al. : Inflammation in the Neurocircuitry of Obsessive-Compulsive Disorder. JAMA Psychiatry. 2017;74(8):833–840. 10.1001/jamapsychiatry.2017.1567 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Augustine EF, Adams HR, Mink JW: Screening Tools for Tic Disorders-Focus on Development or Implementation? Mov Disord. 2017;32(6):946. 10.1002/mds.26981 [DOI] [PubMed] [Google Scholar]
- Baldwin CL, Roberts DM, Barragan D, et al. : Detecting and Quantifying Mind Wandering during Simulated Driving. Front Hum Neurosci.Frontiers Media SA:2017;11:406. 10.3389/fnhum.2017.00406 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bäumer T, Sajin V, Münchau A: Childhood-Onset Movement Disorders: A Clinical Series of 606 Cases. Mov Disord Clin Pract.Wiley-Blackwell:2016;4(3):437–40. 10.1002/mdc3.12399 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beste C, Münchau A: Tics and Tourette Syndrome - Surplus of Actions Rather than Disorder? Mov Disord. 2018;33(2):238–42. 10.1002/mds.27244 [DOI] [PubMed] [Google Scholar]
- Black KJ: Smoke gets in your tics. Authorea.(updated 16 Jul 2018). 10.22541/au.149308896.61629534 [DOI] [Google Scholar]
- Black KJ, Black ER, Greene DJ, et al. : Provisional Tic Disorder: What to tell parents when their child first starts ticcing [version 1; referees: 3 approved]. F1000Res. 2016;5:696. 10.12688/f1000research.8428.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Black KJ, Mink JW: Response to levodopa challenge in Tourette syndrome. Mov Disord. 2000;15(6):1194–1198. [DOI] [PubMed] [Google Scholar]
- Bortolato M, Di Giovanni G: Editorial on Special Issue: Animal Models of Tourette Syndrome. J Neurosci Methods. 2017;292:1. 10.1016/j.jneumeth.2017.10.008 [DOI] [PubMed] [Google Scholar]
- Brandt VC, Moczydlowski A, Jonas M, et al. : Imitation inhibition in children with Tourette syndrome. J Neuropsychol. 2017. 10.1111/jnp.12132 [DOI] [PubMed] [Google Scholar]
- Brandt VC, Stock AK, Münchau A, et al. : Evidence for enhanced multi-component behaviour in Tourette syndrome - an EEG study. Sci Rep. 2017;7(1): 7722. 10.1038/s41598-017-08158-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bruun RD, Budman CL: The course and prognosis of Tourette syndrome. Neurol Clin. 1997;15(2):291–98. 10.1016/S0733-8619(05)70313-3 [DOI] [PubMed] [Google Scholar]
- Burton FH: Back to the Future: Circuit-testing TS & OCD. J Neurosci Methods. 2017;292:2–11. 10.1016/j.jneumeth.2017.07.025 [DOI] [PubMed] [Google Scholar]
- Business Wire: “Teva Announces FDA Approval of AUSTEDO ® (Deutetrabenazine) Tablets for the Treatment of Tardive Dyskinesia in Adults”.2017. Reference Source [Google Scholar]
- Caligiore D, Mannella F, Arbib MA, et al. : Dysfunctions of the basal ganglia-cerebellar-thalamo-cortical system produce motor tics in Tourette syndrome. PLoS Comput Biol. 2017;13(3):e1005395. 10.1371/journal.pcbi.1005395 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cavanna AE, Black KJ, Hallett M, et al. : Neurobiology of the Premonitory Urge in Tourette’s Syndrome: Pathophysiology and Treatment Implications. J Neuropsychiatry Clin Neurosci. 2017;29(2):95–104. 10.1176/appi.neuropsych.16070141 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chipkin RE: Psyadon Announces Positive Results from Phase 2b Clinical Study of Ecopipam for the Treatment of Tourette’s Syndrome in Children.2017. Reference Source [Google Scholar]
- Conceição VA, Dias Â, Farinha AC, et al. : Premonitory urges and tics in Tourette syndrome: computational mechanisms and neural correlates. Curr Opin Neurobiol. 2017;46:187–99. 10.1016/j.conb.2017.08.009 [DOI] [PubMed] [Google Scholar]
- Conelea CA, Wellen BCM: Tic Treatment Goes Tech: A Review of TicHelper.com. Cogn Behav Pract. 2017;24(3):374–81. 10.1016/j.cbpra.2017.01.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Darrow SM, Grados M, Sandor P, et al. : Autism Spectrum Symptoms in a Tourette’s Disorder Sample. J Am Acad Child Adolesc Psychiatry. 2017a;56(7):610–617.e1. 10.1016/j.jaac.2017.05.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Darrow SM, Hirschtritt ME, Davis LK, et al. : Identification of Two Heritable Cross-Disorder Endophenotypes for Tourette Syndrome. Am J Psychiatry. 2017b;174(4):387–96. 10.1176/appi.ajp.2016.16020240 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eaton CK, Jones AM, Gutierrez-Colina AM, et al. : The Influence of Environmental Consequences and Internalizing Symptoms on Children’s Tic Severity. Child Psychiatry Hum Dev. 2017;48(2):327–34. 10.1007/s10578-016-0644-5 [DOI] [PubMed] [Google Scholar]
- Eddy CM, Rizzo R, Cavanna AE: Neuropsychological aspects of Tourette syndrome: a review. J Psychosom Res. 2009;67(6):503–13. 10.1016/j.jpsychores.2009.08.001 [DOI] [PubMed] [Google Scholar]
- Fernández de la Cruz L, Rydell M, Runeson B, et al. : Suicide in obsessive-compulsive disorder: a population-based study of 36 788 Swedish patients. Mol Psychiatry. 2017a;22(11):1626–1632. 10.1038/mp.2016.115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fernández de la Cruz L, Rydell M, Runeson B, et al. : Suicide in Tourette’s and Chronic Tic Disorders. Biol Psychiatry. 2017b;82(2):111–18. 10.1016/j.biopsych.2016.08.023 [DOI] [PubMed] [Google Scholar]
- Forde NJ, Zwiers MP, Naaijen J, et al. : Basal ganglia structure in Tourette’s disorder and/or attention-deficit/hyperactivity disorder. Mov Disord. 2017;32(4):601–4. 10.1002/mds.26849 [DOI] [PubMed] [Google Scholar]
- Fowler SC, Mosher LJ, Godar SC, et al. : Assessment of gait and sensorimotor deficits in the D1CT-7 mouse model of Tourette syndrome. J Neurosci Methods. 2017;292:37–44. 10.1016/j.jneumeth.2017.01.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ganos C, Martino D, Pringsheim T: Tics in the Pediatric Population: Pragmatic Management. Mov Disord Clin Pract. 2017;4(2):160–72. 10.1002/mdc3.12428 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gilbert DL, Budman CL, Singer HS, et al. : A D1 receptor antagonist, ecopipam, for treatment of tics in Tourette syndrome. Clin Neuropharmacol. 2014;37(1):26–30. 10.1097/WNF.0000000000000017 [DOI] [PubMed] [Google Scholar]
- Gilbert DL, Sethuraman G, Sine L, et al. : Tourette’s syndrome improvement with pergolide in a randomized, double-blind, Crossover trial. Neurology. 2000a;54(6):1310–15. 10.1212/WNL.54.6.1310 [DOI] [PubMed] [Google Scholar]
- Gilbert DL, Salleee FR, Sine L, et al. : Behavioral and Hormonal Effects of Low-Dose Pergolide in Children and Adolescents with Gilles De La Tourette’s Syndrome. Curr Ther Res Clin Exp. 2000b;61(6):378–87. 10.1016/S0011-393X(00)80007-7 [DOI] [Google Scholar]
- Godar SC, Bortolato M: What makes you tic? Translational approaches to study the role of stress and contextual triggers in Tourette syndrome. Neurosci Biobehav Rev. 2017;76(Pt A):123–33. 10.1016/j.neubiorev.2016.10.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gordon M, Cologne SE, Hartlein JM, et al. : A Pilot Study of Levodopa for Treatment of Tics in Children and Adults. In F1000Posters2013;4:668 Reference Source [Google Scholar]
- Greenberg E, Tung ES, Gauvin C, et al. : Prevalence and predictors of hair pulling disorder and excoriation disorder in Tourette syndrome. Eur Child Adolesc Psychiatry. 2018;27(5):569–579. 10.1007/s00787-017-1074-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- Greene DJ, Williams Iii AC, Koller JM, et al. : Brain structure in pediatric Tourette syndrome. Mol Psychiatry. 2017;22(7):972–80. 10.1038/mp.2016.194 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grossman N, Bono D, Dedic N, et al. : Noninvasive Deep Brain Stimulation via Temporally Interfering Electric Fields. Cell. 2017;169(6):1029–41.e16. 10.1016/j.cell.2017.05.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Groth C, Mol Debes N, Rask CU, et al. : Course of Tourette Syndrome and Comorbidities in a Large Prospective Clinical Study. J Am Acad Child Adolesc Psychiatry. 2017;56(4):304–12. 10.1016/j.jaac.2017.01.010 [DOI] [PubMed] [Google Scholar]
- Hollis C, Pennant M, Cuenca J, et al. : Clinical effectiveness and patient perspectives of different treatment strategies for tics in children and adolescents with Tourette syndrome: a systematic review and qualitative analysis. Health Technol Assess. 2016;20(4):1–450, vii–viii. 10.3310/hta20040 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Houghton DC, Capriotti MR, Scahill LD, et al. : Investigating Habituation to Premonitory Urges in Behavior Therapy for Tic Disorders. Behav Ther. 2017;48(6):834–46. 10.1016/j.beth.2017.08.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jalenques I, Auclair C, Morand D, et al. : Health-related quality of life, anxiety and depression in parents of adolescents with Gilles de la Tourette syndrome: a controlled study. Eur Child Adolesc Psychiatry. 2017;26(5):603–17. 10.1007/s00787-016-0923-5 [DOI] [PubMed] [Google Scholar]
- Jankovic J, Jimenez-Shahed J, Budman C, et al. : Deutetrabenazine in Tics Associated with Tourette Syndrome. Tremor Other Hyperkinet Mov (N Y). 2016;6:422. 10.7916/D8M32W3H [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jankovic J: Dopamine depleters in the treatment of hyperkinetic movement disorders. Expert Opin Pharmacother. 2016;17(18):2461–70. 10.1080/14656566.2016.1258063 [DOI] [PubMed] [Google Scholar]
- Karlsson V: Using the internet to treat children with tics. Karolinska Institutet; previously published in Medicinsk Vetenskap number 3. ( https://issuu.com/karolinska_institutet/docs/mv_nr_3_2016). Accessed: 2018-07-11. (Archived by WebCite® at http://www.webcitation.org/70pzwqN2o).2016. Reference Source [Google Scholar]
- Kefalopoulou Z, Zrinzo L, Jahanshahi M, et al. : Bilateral globus pallidus stimulation for severe Tourette’s syndrome: a double-blind, randomised crossover trial. Lancet Neurol. 2015;14(6):595–605. 10.1016/S1474-4422(15)00008-3 [DOI] [PubMed] [Google Scholar]
- Kéri S, Szlobodnyik C, Benedek G, et al. : Probabilistic classification learning in Tourette syndrome. Neuropsychologia. 2002;40(8):1356–62. 10.1016/S0028-3932(01)00210-X [DOI] [PubMed] [Google Scholar]
- Khalifa N, von Knorring AL: Psychopathology in a Swedish population of school children with tic disorders. J Am Acad Child Adolesc Psychiatry. 2006;45(11):1346–53. 10.1097/01.chi.0000251210.98749.83 [DOI] [PubMed] [Google Scholar]
- Kumar R, Lang AE: Coexistence of tics and parkinsonism: evidence for non-dopaminergic mechanisms in tic pathogenesis. Neurology. 1997;49(6):1699–1701. 10.1212/WNL.49.6.1699 [DOI] [PubMed] [Google Scholar]
- Lee WT, Huang HL, Wong LC, et al. : Tourette Syndrome as an Independent Risk Factor for Subsequent Sleep Disorders in Children: A Nationwide Population-Based Case-Control Study. Sleep. 2017;40(3):zsw072. 10.1093/sleep/zsw072 [DOI] [PubMed] [Google Scholar]
- Leivonen S, Scharf JM, Mathews CA, et al. : Parental Psychopathology and Tourette Syndrome/Chronic Tic Disorder in Offspring: A Nationwide Case-Control Study. J Am Acad Child Adolesc Psychiatry. 2017;56(4):297–303.e4. 10.1016/j.jaac.2017.01.009 [DOI] [PubMed] [Google Scholar]
- Maia TV, Conceição VA: The Roles of Phasic and Tonic Dopamine in Tic Learning and Expression. Biol Psychiatry. 2017;82(6):401–12. 10.1016/j.biopsych.2017.05.025 [DOI] [PubMed] [Google Scholar]
- Marsden CD: Drug treatment of diseases characterized by abnormal movements. Proc R Soc Med. 1973;66(9):871–73. [PMC free article] [PubMed] [Google Scholar]
- Marsh R, Alexander GM, Packard MG, et al. : Habit learning in Tourette syndrome: a translational neuroscience approach to a developmental psychopathology. Arch Gen Psychiatry. 2004;61(12):1259–68. 10.1001/archpsyc.61.12.1259 [DOI] [PubMed] [Google Scholar]
- Martinez-Torres I, Hariz MI, Zrinzo L, et al. : Improvement of tics after subthalamic nucleus deep brain stimulation. Neurology. 2009;72(20):1787–1789. 10.1212/WNL.0b013e3181a60a0c [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martino D, Pringsheim TM: Reply to "Screening tools for tic disorders - Focus on development or implementation?" Mov Disord. 2017;32(6):947. 10.1002/mds.26986 [DOI] [PubMed] [Google Scholar]
- Martino D, Delorme C, Pelosin E, et al. : Abnormal lateralization of fine motor actions in Tourette syndrome persists into adulthood. PLoS One. 2017a;12(7):e0180812. 10.1371/journal.pone.0180812 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martino D, Ganos C, Pringsheim TM: Tourette Syndrome and Chronic Tic Disorders: The Clinical Spectrum Beyond Tics. Int Rev Neurobiol. 2017b;134:1461–90. 10.1016/bs.irn.2017.05.006 [DOI] [PubMed] [Google Scholar]
- Martino D, Pringsheim TM, Cavanna AE, et al. : Systematic review of severity scales and screening instruments for tics: Critique and recommendations. Mov Disord. 2017c;32(3):467–473. 10.1002/mds.26891 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Meier SM, Dalsgaard S, Mortensen PB, et al. : Mortality risk in a nationwide cohort of individuals with tic disorders and with tourette syndrome. Mov Disord. 2017;32(4):605–9. 10.1002/mds.26939 [DOI] [PubMed] [Google Scholar]
- Molina R, Okun MS, Shute JB, et al. : Report of a patient undergoing chronic responsive deep brain stimulation for Tourette syndrome: proof of concept. J Neurosurg. 2017;1–7. 10.3171/2017.6.JNS17626 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morand-Beaulieu S, Leclerc JB, Valois P, et al. : A Review of the Neuropsychological Dimensions of Tourette Syndrome. Brain Sci. 2017a;7(8): pii: E106. 10.3390/brainsci7080106 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morand-Beaulieu S, Grot S, Lavoie J, et al. : The puzzling question of inhibitory control in Tourette syndrome: A meta-analysis. Neurosci Biobehav Rev. 2017b;80:240–62. 10.1016/j.neubiorev.2017.05.006 [DOI] [PubMed] [Google Scholar]
- Mulders AEP, Leentjens AFG, Schruers K, et al. : Choreatic Side Effects of Deep Brain Stimulation of the Anteromedial Subthalamic Nucleus for Treatment-Resistant Obsessive-Compulsive Disorder. World Neurosurg. 2017;104:1048.e9–1048.e13. 10.1016/j.wneu.2017.05.067 [DOI] [PubMed] [Google Scholar]
- Murphy TK, Fernandez TV, Coffey BJ, et al. : Extended-Release Guanfacine Does Not Show a Large Effect on Tic Severity in Children with Chronic Tic Disorders. J Child Adolesc Psychopharmacol. 2017;27(9):762–70. 10.1089/cap.2017.0024 [DOI] [PubMed] [Google Scholar]
- Neudorfer C, El Majdoub F, Hunsche S, et al. : Deep Brain Stimulation of the H Fields of Forel Alleviates Tics in Tourette Syndrome. Front Hum Neurosci. 2017;11:308. 10.3389/fnhum.2017.00308 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Neurocrine Biosciences Inc: Neurocrine Granted FDA Orphan Drug Designation for Valbenazine for the Treatment of Pediatric Patients with Tourette Syndrome.2017a. Reference Source [Google Scholar]
- Neurocrine Biosciences Inc: Press Release: Neurocrine Announces FDA Approval of INGREZZA (Valbenazine) Capsules as the First and Only Approved Treatment for Adults with Tardive Dyskinesia (TD).2017b. Reference Source [Google Scholar]
- O’Connor KP, Lavoie ME, Schoendorff B: Managing Tic and Habit Disorders: A Cognitive Psychophysiological Approach with Acceptance Strategies. Hoboken, NJ: Wiley-Blackwell.2017. Reference Source [Google Scholar]
- Paton DM: Deutetrabenazine: Treatment of hyperkinetic aspects of Huntington's disease, tardive dyskinesia and Tourette syndrome. Drugs Today (Barc). 2017;53(2):89–102. 10.1358/dot.2017.53.2.2589164 [DOI] [PubMed] [Google Scholar]
- Piacentini J, Bennett S, Capriotti M, et al. : CBIT-Jr: BEhavior Therapy for Chronic Tics in 5-8 Year Olds. Conference Proceedings In 1st World Congress on Tourette Syndrome and Tic Disorders2015;182– 83(Poster P101). 10.3389/978-2-88919-669-2 [DOI] [Google Scholar]
- Polyanska L, Critchley HD, Rae CL: Centrality of prefrontal and motor preparation cortices to Tourette Syndrome revealed by meta-analysis of task-based neuroimaging studies. Neuroimage Clin. 2017;16:257–67. 10.1016/j.nicl.2017.08.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robertson MM, Eapen V, Singer HS, et al. : Gilles de la Tourette syndrome. Nat Rev Dis Primers. 2017;3: 16097. 10.1038/nrdp.2016.97 [DOI] [PubMed] [Google Scholar]
- Sallee F, Kohegyi E, Zhao J, et al. : Randomized, Double-Blind, Placebo-Controlled Trial Demonstrates the Efficacy and Safety of Oral Aripiprazole for the Treatment of Tourette’s Disorder in Children and Adolescents. J Child Adolesc Psychopharmacol. 2017;27(9):771–781. 10.1089/cap.2016.0026 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Salvador A, Worbe Y, Delorme C, et al. : Specific effect of a dopamine partial agonist on counterfactual learning: evidence from Gilles de la Tourette syndrome. Sci Rep. 2017;7(1): 6292. 10.1038/s41598-017-06547-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schaefer SM, Chow CA, Louis ED, et al. : Tic Exacerbation in Adults with Tourette Syndrome: A Case Series. Tremor Other Hyperkinet Mov (N Y). 2017;7:450. 10.7916/D8FF3Z1Q [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schulz KP, Bédard AV, Fan J, et al. : Striatal Activation Predicts Differential Therapeutic Responses to Methylphenidate and Atomoxetine. J Am Acad Child Adolesc Psychiatry. 2017;56(7):602–9.e2. 10.1016/j.jaac.2017.04.005 [DOI] [PubMed] [Google Scholar]
- Shafer RL, Newell KM, Lewis MH, et al. : A Cohesive Framework for Motor Stereotypy in Typical and Atypical Development: The Role of Sensorimotor Integration. Front Integr Neurosci.Frontiers Media SA.2017;11:19. 10.3389/fnint.2017.00019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shale H, Fahn S, Mayeux R: Tics in a patient with Parkinson’s disease. Mov Disord. 1986;1(1):79–83. 10.1002/mds.870010111 [DOI] [PubMed] [Google Scholar]
- Shapiro AK, Shapiro E, Young JG, et al. : Gilles De La Tourette Syndrome. 2nd ed.Raven Press.1988. Reference Source [Google Scholar]
- Singer HS: Discussing outcome in Tourette syndrome. Arch Pediatr Adolesc Med. 2006;160(1):103–5. 10.1001/archpedi.160.1.103 [DOI] [PubMed] [Google Scholar]
- Smeets AYJM, Duits AA, Leentjens AFG, et al. : Thalamic Deep Brain Stimulation for Refractory Tourette Syndrome: Clinical Evidence for Increasing Disbalance of Therapeutic Effects and Side Effects at Long-Term Follow-Up. Neuromodulation. 2018;21(2):197–202. 10.1111/ner.12556 [DOI] [PubMed] [Google Scholar]
- Stárková L: [Tics in Childhood]. Cesk Psychiatr. 1990;86(5):304–10. [PubMed] [Google Scholar]
- Stewart SB, Greene DJ, Lessov-Schlaggar CN, et al. : Clinical correlates of parenting stress in children with Tourette syndrome and in typically developing children. J Pediatr.Elsevier BV:2015;166(5):1297–1302.e3. 10.1016/j.jpeds.2015.01.041 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Suhara T, Chaki S, Kimura H, et al. : Strategies for Utilizing Neuroimaging Biomarkers in CNS Drug Discovery and Development: CINP/JSNP WOrking GRoup Report. Int J Neuropsychopharmacol. 2017;20(4):285–94. 10.1093/ijnp/pyw111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sukhodolsky DG, Woods DW, Piacentini J, et al. : Moderators and predictors of response to behavior therapy for tics in Tourette syndrome. Neurology. 2017;88(11):1029–36. 10.1212/WNL.0000000000003710 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun N, Nasello C, Deng L, et al. : The PNKD gene is associated with Tourette Disorder or Tic disorder in a multiplex family. Mol Psychiatry. 2018;23(6):1487–1495. 10.1038/mp.2017.179 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sweet RD, Bruun R, Shapiro E, et al. : Presynaptic catecholamine antagonists as treatment for Tourette syndrome. Effects of alpha methyl para tyrosine and tetrabenazine. Arch Gen Psychiatry. 1974;31(6):857–61. 10.1001/archpsyc.1974.01760180095012 [DOI] [PubMed] [Google Scholar]
- Takács Á, Shilon Y, Janacsek K, et al. : Procedural learning in Tourette syndrome, ADHD, and comorbid Tourette-ADHD: Evidence from a probabilistic sequence learning task. Brain Cogn. 2017;117:33–40. 10.1016/j.bandc.2017.06.009 [DOI] [PubMed] [Google Scholar]
- Welter ML, Houeto JL, Thobois S, et al. : Anterior pallidal deep brain stimulation for Tourette's syndrome: a randomised, double-blind, controlled trial. Lancet Neurol. 2017;16(8):610–19. 10.1016/S1474-4422(17)30160-6 [DOI] [PubMed] [Google Scholar]
- Willsey AJ, Fernandez TV, Yu D, et al. : De Novo Coding Variants Are Strongly Associated with Tourette Disorder. Neuron. 2017;94(3):486–99.e9. 10.1016/j.neuron.2017.04.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wolmarans W, Scheepers IM, Stein DJ, et al. : Peromyscus maniculatus bairdii as a naturalistic mammalian model of obsessive-compulsive disorder: current status and future challenges. Metab Brain Dis. 2018;33(2):443–55. 10.1007/s11011-017-0161-7 [DOI] [PubMed] [Google Scholar]
- Xenos D, Kamceva M, Tomasi S, et al. : Loss of TrkB Signaling in Parvalbumin-Expressing Basket Cells Results in Network Activity Disruption and Abnormal Behavior. Cereb Cortex. 2017;1–15. 10.1093/cercor/bhx173 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yaniv A, Benaroya-Milshtein N, Steinberg T, et al. : Executive control development in Tourette syndrome and its role in tic reduction. Psychiatry Res. 2018;262:527–35. 10.1016/j.psychres.2017.09.038 [DOI] [PubMed] [Google Scholar]
- Zike ID, Chohan MO, Kopelman JM, et al. : OCD candidate gene SLC1A1/EAAT3 impacts basal ganglia-mediated activity and stereotypic behavior. Proc Natl Acad Sci U S A. 2017;114(22):5719–24. 10.1073/pnas.1701736114 [DOI] [PMC free article] [PubMed] [Google Scholar]