Abstract
Huntington’s disease (HD) is a neurodegenerative disorder characterized by motor, behavioral, and cognitive impairments and significant impacts on patient quality of life. This evidence-based review, conducted by the Korean Huntington Disease Society task force, systematically examines current pharmacological and nonpharmacological interventions for symptomatic management of HD. Following PRISMA guidelines, databases were searched for studies up to August 2022 that focused on 23 symptoms across four domains: motor, neuropsychological, cognition, and others. This review provides a comprehensive and systematic approach to the management of HD, highlighting the need for more high-quality clinical trials to develop robust evidence-based guidelines.
Keywords: Huntington’s disease, Review, Pharmacological, Non-pharmacological
GRAPHICAL ABSTRACT
INTRODUCTION
Huntington’s disease (HD) is a neurodegenerative disorder caused by an autosomal dominant trinucleotide repeat expansion in the huntingtin gene, which is located on chromosome 4p16.3. The prevalence rate is estimated to be approximately 2.7 per 100,000 worldwide; however, it has been consistently reported to be lower (0.4 cases per 100,000) in the Asian population [1]. A recent report suggested that the 10-year prevalence in South Korea is estimated at approximately 2.2 per 100,000 people on the basis of national health insurance system registration data [2].
The signs and symptoms of HD are known to manifest in a triad of motor disturbances, behavioral/psychiatric symptoms and cognitive impairment [3]. The mean age of onset is approximately 35–45 years, which is inversely correlated with the number of CAG repeats in the HD gene. Compared with other genetic disorders, HD has relatively greater penetrance [4]. Progressive neurological abnormalities tend to manifest at a younger age with successive generations, a phenomenon known as anticipation. The overall survival duration for HD patients is estimated to range from 15 to 25 years following the onset of motor symptoms, including in a study from South Korea [5,6]. Given that onset usually occurs in early middle age, the disease inevitably leads to a significant reduction in quality of life for patients and their families [7].
Despite more than a century of research since its initial description, HD remains an untimately fatal disease with no cure. Moreover, comprehensive documentation of symptomatic treatments targeted for troublesome motor, behavioral and cognitive symptoms is lacking. Importantly, the available treatment options are not equally accessible in different regions around the globe. This dearth of evidence may contribute to variations in care, which is predominantly reliant on clinical experience rather than scientific evidence. In a recent survey conducted by the Korean Huntington Disease Society (KHDS) task force, many physicians (36.7%) stated that the lack of treatment guidelines and evidence-based review of the available treatment options in South Korea is one of the huddles in managing HD patients (Supplementary Figure 1 in the online-only Data Supplement).
In recent years, there has been a surge in clinical trials and studies evaluating both pharmacological and nonpharmacological interventions for various clinical problems associated with HD. On the basis of this need, the KHDS task force for the treatment of HD was established, and this study was a systematic review of up-to-date evidence on the symptomatic management of HD, incorporating both pharmacological and nonpharmacological treatments.
MATERIALS & METHODS
The KHDS task force for the systematic review was organized in August 2022. The members of the task force were appointed by the executive committee of the KHDS. This systematic review follows the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines [8].
Search strategy
We searched PubMed, Web of Science, Embase, KoreaMed, Scopus and the Cochrane Review for papers that had been published before August 2022. We selected 22 symptoms that were classified into 4 major HD-related symptom domains: motor, neuropsychological, cognition, and others. The detailed list of 22 symptoms are Motor (Chorea, Dystonia, Swallowing difficulty, Gait and balance, Rigidity, Myoclonus and Bruxism), Behavior (Apathy, Depression, Obsession, Irritability, Akathisia, Anxiety, Suicidal idea or attempt and Sexual disorders), Cognition (Dementia, Hallucination and psychosis), and Others (Sleep, Pain, Weight loss, Hypersalivation and Respiratory dysfunction). The search terms were chosen on the basis of a list of symptoms and were customized depending on the symptoms (For example, suicidal idea, ALL = [huntington] OR ALL = [huntington’s] OR ALL = [huntington’s disease]) AND (ALL = [suicide] OR ALL = [suicidal] OR ALL = [suicidal ideation]). We included clinical trials, observational studies, case series and case reports for 22 symptoms that were published from 1994 to 2022. However, for chorea, we excluded single case reports. After the identification phase, we removed duplications, and the list of articles entered the screening phase. The titles and abstracts of the articles were manually reviewed for exclusion. The exclusion criteria included animal studies, nonmedical articles, and non-English articles. All the articles were subsequently sought for retrieval, and the reports that were unable to be retrieved online were excluded. Finally, reports were assessed for eligibility. In this phase, review articles and studies without reported outcomes were excluded. The flowcharts of each symptom were drawn with the PRISMA flowchart and are shown in Figure 1 and Supplementary Figures 2-23 (in the online-only Data Supplement). The availability of the treatment options in South Korea as of July 2024 is noted in Supplementary Figures 2-23 (in the online-only Data Supplement). Owing to the limited space for references, some of the references for Class U studies are provided in the Supplementary Material (in the online-only Data Supplement).
Efficacy evaluation
As we reviewed studies, including case series and case reports, we used the AAN classification of evidence [9]. From the criteria, we labeled every article with Classes I, II, III and IV as defined in the criteria. We then marked the level of evidence for each treatment option as levels A, B, C and U.
Method for reaching a consensus
The initial manuscript by the task committee was constructed and sent to the clinical experts of HD, who were chosen by the executive committee of the KHDS. Recommendations and expert opinions were obtained in two rounds and were met with consensus by the entire task force.
RESULTS
Motor symptoms
Chorea
Chorea, characterized by involuntary, irregular, random, and unsustained movements, is the most apparent and cardinal motor feature of HD, and the current consensus guidelines recommend that pharmacological interventions should be considered when the patient experiences functional discomfort or suffering from chorea [10-12].
Selective vesicular monoamine transporter 2 inhibitors
Tetrabenazine was assessed in a 12-week Class I randomized clinical trial as antichoreic therapy in 84 study patients [13]. This TETRA-HD study demonstrated that tetrabenazine effectively ameliorates chorea and is well tolerated [14]. Fifty HD patients treated with tetrabenazine had a significant reduction in the mean difference (±standard deviation) in the total maximal chorea score on the Unified Huntington’s Disease Rating Scale (UHDRS) compared with 34 placebo-treated patients (-5.0 ± 0.5 vs. -1.5 ± 0.7), with a treatment difference of -3.5 (95% confidence interval [CI] -5.2 to -1.9; p < 0.0001) [14]. In addition, a Class II randomized clinical study evaluating chorea after tetrabenazine withdrawal reported that the discontinuation of tetrabenazine was significantly related to reemergent chorea [15]. Clinicians might offer tetrabenazine to reduce chorea in HD patients (Level B), but there are safety concerns about the use of tetrabenazine, which can cause depression, parkinsonism, neuroleptic malignant syndrome and prolonged QT intervals [13-17]. When patients have poorly controlled depression or suicidality, tetrabenazine is contraindicated [13-19]. Recently, two Class I randomized controlled studies examined deutetrabenazine and valbenazine for chorea [20,21]. In a 12-week randomized controlled trial (RCT) involving 90 patients with HD, deutetrabenazine treatment resulted in a reduction of 4.4 units from the baseline in the UHDRS, the total maximal chorea score, compared with a reduction of 1.9 units on placebo treatment (treatment difference -2.5, 95% CI -3.7 to -1.3; p < 0.001) [20]. A 12-week, 128-subject KINECT-HD study revealed that UHDRS total maximal chorea scores significantly decreased by -4.6 in 64 patients receiving valbenazine vs. -1.4 for placebo (treatment difference -3.2, 95% CI -4.4 to -2.0; p < 0.0001) [21]. Both new-generation vesicular monoamine transporter 2 (VMAT2) inhibitors have demonstrated effectiveness on chorea and can be used as first-line antichoreic therapies in HD (Level B), particularly when patients are intolerant to tetrabenazine [19-22]. However, new-generation VMAT2 inhibitors are not available in South Korea as of January 2024.
Antipsychotic drugs
Antipsychotics are currently used as first-line monotherapy or second-line off-label alternatives to suppress HD chorea by a considerable number of respondents in several opinion surveys of HD experts [18,23-25]. Clozapine has shown little benefit in reducing HD chorea in Class II clinical trials [26] and single-arm observation studies [26] (Level U). In a small class III crossover trial, aripiprazole had a beneficial effect on China that was comparable to that of tetrabenazine [27]. Other atypical antipsychotic drugs (APDs), including olanzapine and risperidone, have been studied only in small uncontrolled clinical trials (Level U) [28-30]. However, longitudinal, observational data from an Enroll-HD multicenter study with 1,612 patients revealed that olanzapine and risperidone appeared to have antichoreic efficacy comparable to that of tetrabenazine [29,31]. Overall, although limited in evidence, several studies support the effect of APDs on choreic manifestations in patients with HD.
Other pharmacological agents
Amantadine is a weak, noncompetitive N-methyl-D-aspartate (NMDA) antagonist. For Huntington’s chorea, one Class II trial involving acute intravenous (IV) amantadine challenge followed by chronic oral amantadine for 1 year and another 2-week Class II crossover clinical trial involving oral amantadine showed that amantadine had a modest effect on reducing the incidence of chorea [32,33]. However, the other 2-week Class I crossover trial using oral amantadine showed contrasting results, with insignificant effects on objective measures, although some patients reported subjective improvement in China [34]. Taken together, the data are inconclusive in terms of supporting or refuting the regular use of oral amantadine for controlling chorea in HD patients [32-36] (conflicting Class I and II studies, Level U).
Apomorphine is a nonselective dopamine receptor agonist that has been used to attenuate motor complications in Parkinson’s disease (PD) patients [37]. One small class III clinical trial of acute administration of subcutaneous apomorphine revealed a favorable antichoreic effect [38]. Apomorphine resulted in a significant decrease in the chorea at rest (-50.00%, p < 0.01) and the chorea during voluntary movement score (-30.18%, p < 0.05) of the quantified neurological examination from the baseline, whereas no significant change was found after placebo [38]. Another 5-day Class II crossover controlled study involving continuous subcutaneous apomorphine injection (CSAI) also revealed a significant and sustained reduction in chorea in HD patients [39]. The abnormal involuntary movement scale score was significantly lower in patients receiving apomorphine injection (-34.4% ± 14.6%, p < 0.0001) than in those receiving placebo (-0.20% ± 3.7%) [39]. These data suggest that apomorphine may be effective for the short-term management of chorea in HD patients (Level C); however, the long-term antichoreic efficacy of apomorphine, including CSAI, is unknown [39,40].
Multiple RCTs of ethyl-eicosapentaenoic acid (ethyl-EPA) [41-44] and latrepirdine [45,46] have failed to show benefits in chorea patients with HD (Level A). Coenzymes Q10 [47,48], creatine [49-54], pepinemab [55] and PBT2 [56] were also not effective for chorea in HD (Level B). (-)-OSU6162 [57], AFQ-056 (mavoglurant) [58], atomoxetine [59], bupropion [60], cannabidiol oral spray [61], citalopram [62], donepezil [63,64], fluoxetine [65], lamotrigine [66], OPC-14117 [67], SBT-020 [68], selisistat [69], and nabilone [70,71] did not show antichoreic efficacy and are possibly ineffective for reducing chorea in HD (Level C). There are insufficient or conflicting data to draw recommendations on HD chorea about riluzole, cysteamine IONIS-HTTRx (tominersen), rivastigmine, minocycline, memantine, valproic acid, levetiracetam, ketamine, idebenone, nilotinib, proglumide, rilmenidine, triheptanoin, remacemide, pridopidine and cysteamine (Level U, Supplementary Material in the online-only Data Supplement). Data are also inadequate to support or refute cell-based therapy, including fetal cell transplantation and gene therapy using ciliary neurotrophic factor on chorea in HD (Level U, Supplementary Material in the online-only Data Supplement).
Physical therapy and dietary interventions
Physical therapies are possibly effective for reducing HD-associated chorea in two Class II clinical trials [72-74] (Level C). Additional Class III clinical trials and case reports further indicate that physical therapy and rehabilitation can improve chorea in HD patients [75-81]. However, while two longitudinal observational studies [82,83] have explored dietary interventions in HD patients, there is insufficient evidence to support dietary interventions for chorea (Level U).
Brain modulation therapy
Case-based studies in multisensory stimulation, transcranial magnetic stimulation (TMS), pallidal deep brain stimulation (DBS) and pallidothalamic tractomy have shown improvements in chorea in HD patients (Level U, Supplementary Material in the online-only Data Supplement). However, additional well-designed clinical trials are needed to confirm their effects.
Summary
VMAT2 inhibitors (tetrabenazine, deutetrabenazine and valbenazine) are effective pharmacological options for treating chorea in HD. The use of apomorphine, amantadine and atypical antipsychotics requires careful consideration due to limited evidence. Nonpharmacological approaches, including exercise programs and physical therapy, may show potential benefits. The treatment evidence for chorea in HD patients is summarized in Table 1.
Table 1.
Effective |
No benefit |
Conflicting |
||||||||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
Class | I | II | III | IV | Class | I | II | III | IV | Class | I | II | III | IV |
Level A | Level U | |||||||||||||
Ethyl-EPA | 3 | 1 | Riluzole | 1 | 1 | 1 | ||||||||
Latrepirdine | 2 | Cysteamine | 1 | 2 | ||||||||||
Level B | Level B | Rivastigmine | 1 | 1 | ||||||||||
Tetrabenazine | 1 | 1 | 3 | Pepinemab | 1 | Amantadine | 4 | 1 | ||||||
Valbenazine | 1 | Coenzyme Q10 | 1 | 1 | Minocycline | 2 | 3 | |||||||
Deutetrabenazine | 1 | 1 | Creatinine | 1 | 2 | 1 | 2 | Clozapine | 1 | 1 | ||||
PBT2 | 1 | Remacemide | 2 | |||||||||||
Pridopidine | 3 | 1 | 1 | 3 | ||||||||||
Level C | Level C | |||||||||||||
Apomorphine | 1 | 1 | Bupripion | 1 | ||||||||||
Physical therapy | 4 | 3 | 3 | Cannabidiol oral spray | 1 | |||||||||
Citalopram | 1 | |||||||||||||
Donepezil | 1 | 1 | ||||||||||||
Fluoxetine | 1 | |||||||||||||
Lamotrigine | 1 | |||||||||||||
OPC-14117 | 1 | |||||||||||||
SBT-020 | 1 | |||||||||||||
Nabilone | 1 | |||||||||||||
(-)-OSU6162 | 1 | |||||||||||||
AFQ-056 | 1 | |||||||||||||
Atomoxetine | 1 |
Therapies with undetermined evidence (U) for effective or no benefit domain were excluded from this table.
EPA, eicosapentaenoic acid.
Korean HD expert comment
Although VMAT2 inhibitors are the most reliable treatment option, tetrabenazine is the only available drug in South Korea. However, fewer than 10% of HD patients in South Korea are prescribed this drug, possibly due to poor accessibility and high cost. 2 New-generation VMAT2 inhibitors (deutetrabenaine and valbenaine) have not yet been introduced in South Korea. Notably, deutetrabenazine is reported to be relatively safe in terms of QT prolongation (see related comment in Depression section) [84]. VMAT2 inhibitors should be cautiously prescribed for patients with severe depression or suicidal tendencies, as they may worsen these conditions. Atypical antipsychotics may be considered as a second-line off-label alternative treatment. The importance of exercise and physical therapies in HD patients has been overlooked, which warrants a well-designed clinical trial in South Korea.
Dystonia
Whereas chorea is the most widely recognized involuntary movement in HD, dystonia is also frequently reported. The majority of HD patients experience dystonia to some degree, whereas the dystonia of HD patients manifests as a variety of movements and postures that are not typical of what is most common in idiopathic child-onset torsion dystonia (e.g., foot inversion) or adult-onset focal dystonia (e.g., blepharospasm, torticollis, writer’s cramp), and patients are rarely aware of their dystonic movements or postures.
One Class II RCT demonstrated that deutetrabenazine is possibly effective in improving dystonia in patients with HD (Level C) [20], whereas other pharmaceutical management methods, including riluzole [85], fluoxetine [65], minocycline [86], and ethyl-EPA [42], are possibly ineffective (Level C). There is insufficient evidence to assess the efficacy of pridopidine and IV administration of amantadine and cannabinoids on dystonia in HD patients (Level U, Supplementary Material in the online-only Data Supplement). Data are inadequate for assessing the efficacy of surgical interventions, including pallidal DBS, internal globus pallidotomy, and fetal neural transplants (Level U, Supplementary Material in the online-only Data Supplement). There is also insufficient evidence to assess the efficacy of noninvasive brain stimulation, namely, direct current stimulation (Level U, Supplementary Material in the online-only Data Supplement).
Summary
Deutetrabenazine may be effective for dystonia in HD patients, but there is limited evidence supporting the use of other medications (Table 2).
Table 2.
Effective |
No benefit |
Conflicting |
||||||||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
Class | I | II | III | IV | Class | I | II | III | IV | Class | I | II | III | IV |
Dystonia | ||||||||||||||
Level C | Level C | Level U | ||||||||||||
Deutetrabenazine | 1 | Riluzole | 1 | 1 | Pridopidine | 2 | ||||||||
Fluxetine | 1 | |||||||||||||
Minocycline | 1 | |||||||||||||
Ethyl-EPA | 1 | |||||||||||||
Gait | ||||||||||||||
Level B | Level U | |||||||||||||
Pridopidine | 1 | Gpi DBS | 4 | |||||||||||
Level C | ||||||||||||||
Physical therapy | 1 | 1 | 12 |
Therapies with undetermined evidence (U) for effective or no benefit domain were excluded from this table.
EPA, eicosapentaenoic acid; Gpi, globus pallidus interna; DBS, deep brain stimulation.
Korean HD expert comment
Tetrabenazine may be considered, despite limited evidence, depending on its availability. In cases of troublesome dystonia, typical antidystonia medications (anticholinergics, clonazepam, baclofen, etc.) can be used in HD patients, although there is a lack of evidence. For severe dystonic spasms in young-onset patients with advanced stages of HD, gabapentin or pregabalin—alpha-2 delta ligands—may relieve symptoms despite the lack of sufficient evidence in HD. Notably, higher doses than those typically used for pain relief may be needed.
Gait and balance
Gait impairment in HD patients can be influenced by concomitant chorea or impaired balance. As a result, HD patients may experience frequent falls and become physically dependent. Physical therapy or exercise has been studied in the context of gait impairment in HD patients, with home-based exercise showing some improvement in an RCT and a single-blind controlled trial (Level C) [73,87]. Gait improvement after physical therapy or exercise was also observed in 11 out of 12 case series [75,88-97]. One study did not show improvement in gait after exercise, but this study only included short-duration exercise (20 minutes on a treadmill), which might have been insufficient [79].
In terms of pharmacological treatment, RCTs have been conducted using pridopidine, a dopaminergic stabilizer. While there was no significant change in the total motor score, subitem scores of gait and balance showed improved (Level B) [98]. Conventional neuroleptics generally improve chorea, but they do not have a beneficial effect on gait (Level U, Supplementary Material in the online-only Data Supplement). Gait and balance improvements were observed in some patients treated with tetrabenazine and olanzapine (Level U, Supplementary Material in the online-only Data Supplement). Two case reports revealed improved parkinsonism, including gait and balance, when levodopa and amantadine (Level U, Supplementary Material in the online-only Data Supplement) were used. Other case reports revealed improved gait or balance after the use of zotepine, bromocriptine, and caryolanemagnolol (Level U, Supplementary Material in the online-only Data Supplement).
There is insufficient evidence to assess the efficacy of DBS in improving gait in HD patients (Level U). In one study, improvements in gait and balance, along with chorea, were observed following DBS [99], whereas in three studies, there was no gait improvement after DBS [100-102]. After DBS, some patients experience worsened gait and bradykinesia as side effects [102].
Summary
Home-based exercise and pridopidine may be effective in reducing gait and balance impairment in HD patients. There is insufficient evidence to recommend the use of any medication to improve gait impairment in HD patients (Table 2).
Korean HD expert comment
In patients with severe chorea, particularly in the legs, antichoreic medications can help improve gait. In some patients with predominant parkinsonian symptoms, small-dose levodopa (mostly 450 mg/day or less) can help improve bradykinesia and short-step shuffling gait.
Parkinsonism
Parkinsonism can manifest in the later stages of adult-onset HD as the predominant akinetic rigidity or as the Westphal variant in juvenile cases [103]. Parkinsonism may also occur as drug-induced parkinsonism caused by medications such as tetrabenazine [104,105], neuroleptics [106] and levetiracetam [107]. Additionally, there have been reports of concomitant PD and HD [108].
There are only case-based reports that have assessed the efficacy of treatment in improving parkinsonism in HD patients. In adult cases, studies have reported that levodopa (200 mg/day to 600 mg/day) [109], amantadine, a combination of levodopa/amantadine and rasagiline improved parkinsonism in HD patients (Level U, Supplementary Material in the online-only Data Supplement). A case series showed that nabilone improved parkinsonism induced by tetrabenazine in HD patients [110]. In juvenile patients, five childhood HD patients with rigidity and bradykinesia, rigidity and hypokinesia improved when levodopa was used at doses ranging from 75 mg/day to 600 mg/day (Level U, Supplementary Material in the online-only Data Supplement). Additionally, a patient who did not respond to levodopa 150 mg/day showed improvements in rigidity and gait when treated with pramipexole ranging from 0.27 mg/day to 1.08 mg/day (Level U, Supplementary Material in the online-only Data Supplement). Pyridostigmine did not effectively improve rigidity (Level U, Supplementary Material in the online-only Data Supplement).
Summary
Antiparkinsonian medications, including levodopa, dopamine agonists, amantadine and/or rasagiline, may be beneficial for parkinsonism in HD patients.
Korean HD expert comment
In advanced cases with concomitant rigidity and dystonia, antidystonic medications can be first attempted, followed by small-dose levodopa. Other antiparkinsonian medications, including dopamine agonists, may be attempted in patients with parkinsonism.
Myoclonus
Myoclonus is a relatively uncommon feature of late-onset HD but can present with tremors. In the juvenile form of HD, nonepileptic myoclonus may present as a dominant symptom that may impair the functional capacity and activity of daily living. There are case series and reports showing benefits from valproate, clonazepam, or their combination (Level U, Supplementary Material in the online-only Data Supplement). Piracetam and haloperidol/valproate combinations have been reported in case reports showing some improvement in myoclonus (Level U, Supplementary Material in the online-only Data Supplement). These symptomatic medications improve the clinical outcome when they are used appropriately to balance risk and benefit, especially for elderly individuals, who are monitoring for potential somnolence and falls.
Summary
Valproate and clonazepam might be beneficial in relieving myoclonus in HD patients.
Korean HD expert comment
Because myoclonus is relatively rare in adult-onset HD, there is a lack of research on pharmacological treatments targeting myoclonus in HD. However, fast chorea can sometimes be mistaken for myoclonus. Recurrent sudden vomiting can be caused by myocloniform diaphragmatic hyperkinesia. Although evidence is limited, levetiracetam, valproic acid and clonazepam may be used to manage myoclonus symptoms.
Akathisia
Akathisia is defined as the inability to remain still and is a neuropsychiatric syndrome that is associated with psychomotor restlessness. Akathisia may be a side effect of antichoreic medications, including the VMAT2 inhibitor tetrabenazine [13,14,111,112] or deutetrabenazine [20,22,113]. Most cases are mild, with a prevalence ranging from 6%–14%. Cessation of responsible medications may result in symptom remission. There are reports of akathisia with a cariprazine trial [114] for mood and cognition and M6 nabilone [110] for the treatment of resistant motor symptoms in HD patients.
Summary
Akathisia may present as a side effect of antichoreic medications in HD. Cessation of responsible medications can lead to remission of akathisia.
Korean HD expert comment
Medication-induced akathisia can be closely monitored and controlled.
Behavioral symptoms
Apathy
Apathy is characterized by a quantifiable reduction in goal-directed behavior [115], which becomes prevalent in the advanced stages of HD. One RCT and 6 therapeutic intervention studies were included to evaluate the evidence of available therapies for apathy in HD patients [60,114,116-120]. One Class II RCT demonstrated that bupropion is possibly ineffective in improving apathy in patients with HD [60] (Level C), whereas other pharmaceutical management methods, including rasagiline, cariprazine, cannabinoids, and risperidone, also have insufficient evidence for managing apathy (Level U, Supplementary Material in the online-only Data Supplement). Data are inadequate for assessing the efficacy of contemporary dance practices (Level U) or time-restricted ketogenic diets (Level U, Supplementary Material in the online-only Data Supplement).
Summary
There is no available evidence-based therapy for managing apathy (Table 3).
Table 3.
Effective |
No benefit |
||||||||
---|---|---|---|---|---|---|---|---|---|
Class | I | II | III | IV | Class | I | II | III | IV |
Apathy | |||||||||
Level C | |||||||||
Bupropione | 1 | ||||||||
Depression | |||||||||
Level B | |||||||||
Citalopram | 1 | ||||||||
Suicidal ideation | |||||||||
Level C | |||||||||
SRX246 | 1 | ||||||||
Laquinimod | 1 | ||||||||
Irritability | |||||||||
Level C | |||||||||
Aripiprazole | 1 | 2 | |||||||
Anxiety | |||||||||
Level C | |||||||||
Pridopidine | 1 |
Therapies with undetermined evidence (U) for effective or no benefit domain were excluded from this table. There were no treatments classified as having conflicting evidence in this category.
Korean HD expert comment
There are no treatments specifically effective for apathy in HD. In patients with unique cognitive features associated with apathy, cognitive enhancers such as choline esterase inhibitors may be used, as in other degenerative diseases. In some patients with concomitant depression, antidepressants may be beneficial. Clinicians may consider reducing the dosage of sedative medications in patients with HD with apathy [10].
Depression
Depression is a common psychiatric symptom in HD patients and may appear in any stage of the disease, even in the premanifest stage, and affects their ability to perform activities of daily living. One RCT reported a positive effect of citalopram (Level B) [62]. Fluoxetine was used to treat nondepressed HD patients and failed to improve functional capacity (Level U) [65]. Several case reports have applied venlafaxine, cariprazine and lamotrigine, which are effective in relieving symptoms of depression (Level U, Supplementary Material in the online-only Data Supplement). There are case series regarding electroconvulsive therapy (ECT) and TMS as a treatment for depression, which also have positive effects (Level U, Supplementary Material in the online-only Data Supplement). Finally, some studies have shown the benefit of physical therapy/rehabilitation as a treatment option for depression in HD patients (Level U, Supplementary Material in the online-only Data Supplement).
Summary
Citalopram is likely efficacious for relieving depression in HD patients, whereas there is limited evidence for the effectiveness of other medications, TMS, or physical therapy (Table 3).
Korean HD expert comment
Selective serotonin reuptake inhibitors (SSRIs) can be tried first for moderate to severe depression in HD. Combining SSRIs and antipsychotics requires caution, as it can lead to QT prolongation. Deutetrabenazine can be considered a viable option because of its relative safety regarding QT prolongation [84]. Nonpharmacological therapies such as TMS or transcrinial direct current stimulation (tDCS) are worth studying for depressive symptoms in HD patients.
Suicidal idea or attempt
The incidence of suicidal ideation is estimated to be as high as 19%, and the complete suicide rate in HD patients is reported to be as high as 13% [121,122]. Depression, anxiety, bipolar disorder, antidepressant or anxiolytic use, and a prior suicide attempt at baseline are associated with increased suicidality in HD patients [123]. There were randomized phase 2 clinical trials that showed insignificant results for suicidality with SRX246 (a vasopressin 1a receptor antagonist) [124] and liquinimod (Level C). There are several case report-based pharmacological interventions showing some benefit in terms of suicidality in HD patients treated with mirtazapine, lithium, and olanzapine (Level U, Supplementary Material in the online-only Data Supplement). Several case reports have shown improvements in suicidal ideation with ECT and psychotherapy (Level U, Supplementary Material in the online-only Data Supplement).
Summary
SRX246 and laquinimod did not reduce suicidal ideation. ECT and psychotherapy may reduce suicidal ideation or attempts (Table 3).
Korean HD expert comment
In HD, suicidal ideation can occur from the early stages of the disease and is associated with a higher mortality rate, necessitating close monitoring throughout the disease course.
Obsession
The treatment strategy for obsession in HD patients is similar to that for obsessive‒compulsive disorder (OCD) patients [125]. All of the studies targeting HD and obsession have been case reports. Cognitive behavioral therapy (CBT) improved OCD symptoms in one patient [126], and the use of SSRIs or tricyclic antidepressants such as fluoxetine, clomipramine, and sertraline also led to improvements in OCD symptoms (Level U, Supplementary Material in the online-only Data Supplement). Antipsychotics such as paliperidone reduced aggression, and the use of quetiapine and olanzapine improved OCD symptoms (Level U, Supplementary Material in the online-only Data Supplement).
Summary
There is insufficient evidence for the management of obsession in HD patients, with some case reports of improvement after the use of SSRIs, antipsychotics and CBTs.
Korean HD expert comment
Psychiatric symptoms, including obsession, are rarely observed in isolated symptoms but are commonly accompanied by some cognitive or motor impairment in HD patients, for which physicians have difficulty assessing severity.
Irritability
Irritability is a common and major nonmotor symptom that affects 38%–73% of HD patients. The irritable and aggressive behavior of medical staff, other patients and caregivers results in ineffective treatment for HD. Aripiprazole has been reported to improve irritability (Level C) [127-129]. A small double-blind RCT revealed that the combination of lithium and haloperidol was effective but not effective when these drugs were administered separately [130] (Level U). On the basis of case reports and series, buspirone, cannabinoids, olanzapine, risperidone, valproate, sertraline [131], quetiapine, and lamotrigine have shown positive effects. Conflicting results have been reported with propranolol (Level U, Supplementary Material in the online-only Data Supplement). No definite effect was found with SRX246 [132], donepezil [63] or ECT [133].
Summary
Aripiprazole and a combination of haloperidol and lithium may be effective for treating irritability in HD patients (Table 3).
Korean HD expert comment
Aripiprazole may be used to manage irritable behavior.
Anxiety
Anxiety is one of the commonly observed nonmotor symptoms in HD patients, although it has been underrecognized in clinical practice. In addition to available therapeutic options for anxiety in the general population (i.e., SSRIs, serotonin-norepinephrine reuptake inhibitors, or anxiolytics), one RCT and 5 therapeutic intervention studies were reviewed to evaluate the evidence of available therapies for anxiety in HD patients [81,116,134-137]. One Class II RCT revealed that pridopidine tended to improve anxiety symptoms, but the changes were insignificant compared with those associated with placebo [136] (Level C). There is insufficient evidence for olanzapine (Level U, Supplementary Material in the online-only Data Supplement). Multidisciplinary rehabilitation intervention and CBT also require further studies to establish sufficient evidence for managing anxiety in patients with HD (Level U, Supplementary Material in the online-only Data Supplement).
Summary
There is insufficient evidence for the management of anxiety in HD patients. Pridopidine and neuroleptics, multidisciplinary rehabilitation and CBT have provided limited evidence for their ability to improve anxiety (Table 3).
Korean HD expert comment
As with other psychiatric symptoms, anxiety is often accompanied by other psychiatric and neurologic symptoms in HD patients. Furthermore, psychiatric symptoms are also linked to neurodegeneration in HD patients, which can progress over time, and nonpharmacological interventions are worth further investigation.
Sexual disorders
The behavioral symptoms of HD often result in a prevalence of sexual disorders of up to 85% [138]. Most sexual disorders are hypoactive disorders that are not frequently managed. In the case of hypersexuality in HD, behaviors should be controlled. There is limited evidence with case reports for the use of haloperidol, leuprolide acetate, cyproterone and medroxyprogesterone (Level U, Supplementary Material in the online-only Data Supplement).
Summary
There is insufficient evidence to recommend any medication to manage hypersexuality in HD patients.
Korean HD expert comment
Sexual disorders in HD patients are often overlooked in neurologic clinics, but they need to be regularly monitored.
Cognition
Dementia
Cognitive impairment is one of the major nonmotor symptoms of HD, with behavioral and motor symptoms. The earliest change is characterized as psychomotor slowing, which may be reveled at the premanifest stage [139] and progresses over time. Executive and attention dysfunction is characteristic of HD, which may contribute to difficulty in multitasking [140]. Emotional processing and memory are also significantly impaired in HD patients.
However, the majority of clinical trials have failed to show a benefit in cognitive function in HD patients. Several RCTs with pridopidine have targeted the cognitive features of HD but have shown no effect [98,141,142] (Level A). A single RCT with donepezil [64] did not significantly improve cognition in HD patients (Level B). RCTs with citalopram [62], atomoxetine [59], fetal striatal grafting and cannabinoids (sativex) [61] failed to show any benefit (Level C). Studies with latrepirdine [45,46] and rivastigmine [143,144] have shown conflicting results (Level U). Several case series have shown that multidisciplinary rehabilitation/physical therapies [145,146] improve cognition or stabilize cognitive progression (Level U). Globus pallidus interna (GPi) DBS [147-149] showed nonsignificant or conflicting results for cognition in HD patients (Level U). One small randomized sham-controlled trial using tDCS revealed immediate improvement in working memory function in HD patients without confirmation of long-term benefit [150] (Level U). There was insufficient evidence for the use of cariprazine, memantine, a ketogenic diet and TMS (Level U, Supplementary Material in the online-only Data Supplement).
Summary
Pridopidine, latrepirdine, donepezil, rivastigmine and GPi DBS showed nonsignificant or conflicting results for managing dementia in HD patients (Table 4).
Table 4.
Effective |
No benefit |
Conflicting |
||||||||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
Class | I | II | III | IV | Class | I | II | III | IV | Class | I | II | III | IV |
Dementia | ||||||||||||||
Level A | Level U | |||||||||||||
Pridopidine | 3 | Latrepirdine | 2 | |||||||||||
Level B | Rivastigmine | 1 | 1 | |||||||||||
Donepezil | 1 | Gpi DBS | 1 | 3 | 4 | |||||||||
Level C | ||||||||||||||
Citalopram | 1 | |||||||||||||
Atomoxetine | 1 | |||||||||||||
Fetal striatal grafting | 1 | 1 | ||||||||||||
Cannabinoid (sativex) | 1 | |||||||||||||
Sleep | ||||||||||||||
Level C | ||||||||||||||
Pridopidine | 1 | |||||||||||||
Respiratory function | ||||||||||||||
Level C | ||||||||||||||
Home-based respiratory muscle training program | 1 |
Therapies with undetermined evidence (U) for effective or no benefit domain were excluded from this table.
Gpi, globus pallidus interna; DBS, deep brain stimulation.
Korean HD expert comment
Cognitive changes in HD patients may not be effectively measured with routine neuropsychological tests. While more studies are needed to elucidate the causes of cognitive impairment in HD patients, nonpharmacological interventions such as noninvasive stimulation, cognitive training or rehabilitation may be worth investigating.
Psychosis and hallucination
Psychotic symptoms, which impair the quality of life of patients and caregivers, are reported to occur in 3%–20% of HD patients [151]. In the juvenile form of HD, psychosis is reported in more than one-third of individuals, with visual hallucination being the most common symptom, followed by auditory hallucinations [152]. Thus, juvenile HD may be misdiagnosed as schizophrenia [153].
Antipsychotics, including risperidone, olanzapine, aripiprazole and quetiapine, have been reported to be effective on a case-report basis (Level U, Supplementary Material in the online-only Data Supplement). One case report revealed benefits with the addition of reboxetine to antipsychotics. Several studies have reported positive outcomes with clozapine, especially in patients refractory to other antipsychotics (Level U, Supplementary Material in the online-only Data Supplement). Clozapine treatment necessitates regular complete blood count monitoring due to the risk of agranulocytosis. In case reports and series, ECT could improve psychosis in HD patients, including those who are resistant to medication and those in urgent clinical settings (Level U, Supplementary Material in the online-only Data Supplement).
Summary
Antipsychotics and ECTs may reduce hallucinations and psychosis in HD patients.
Korean HD expert comment
First-line management of psychosis may involve the administration of antipsychotics. However, antipsychotic use is linked to increased mortality in the neurodegenerative population; thus, cautious prescriptions and close monitoring of adverse events are needed.
Other nonmotor symptoms
Sleep disturbance
Approximately two-thirds of patients with HD suffer from sleep disturbances with various causes and diverse clinical manifestations [10]. In addition to available therapeutic options (i.e., lifestyle modification and hypnotics) in the general population, one RCT and 2 therapeutic intervention studies were included to evaluate the evidence of therapies for sleep disturbance in HD patients [136,137,154]. One Class II RCT failed to show the efficacy of pridopidine for improving sleep disturbance in HD patients [136] (Level C). Quetiapine also has insufficient evidence for treating insomnia in patients with HD (Level U, Supplementary Material in the online-only Data Supplement). Multidisciplinary rehabilitation intervention also requires further studies to establish sufficient evidence for managing sleep disturbance in HD (Level U, Supplementary Material in the online-only Data Supplement). Although the relationship between sleep-disordered breathing and HD is not yet clear, one case report revealed a reduction in apnea and respiratory arousal after continuous positive airway pressure was used (Level U, Supplementary Material in the online-only Data Supplement).
Summary
There is insufficient evidence to recommend therapeutic interventions for sleep disturbance in HD patients (Table 4).
Korean HD expert comment
Disruptions in sleep structures and insomnia need to be systematically studied in HD patients, and more therapeutic trials are needed in the future.
Bruxism
Bruxism, which is characterized by grinding and clenching of the teeth, is a relatively uncommon presentation in HD. After the initial report by Tan et al. [155] in 2000, subsequent cases were reported [156]. The underlying phenomenology is understood as the form of oromandibular dystonia. Bruxism may not respond or worsen despite successful treatment of chorea. The symptoms may accompany dysphagia and worsen with neuroleptics. The only treatment option that is reported to relieve bruxism is botulinum toxin injection [155,157] (Level U). The main injection site was the bilateral masseter muscles, with a total of 60–100 units.
Summary
Botulinum toxin injections have been found to be helpful in improving bruxism.
Korean HD expert comment
Neuroleptics used for chorea can exacerbate bruxism, so if bruxism is severe, adjusting the causative medication may be necessary. Additionally, the use of botulinum toxin injections could be beneficial for treating bruxism.
Hypersalivation
Treatment for hypersalivation includes the use of topical scopolamine, oral glycopyrrolate, and botulinum toxin injections [158], but there have been no studies specifically targeting patients with HD. There were two cases in which cannabinoids improved hypersalivation, leading to the discontinuation of previously used botulinum toxin A injections (Level U) [118].
Summary
There is insufficient evidence to recommend any therapeutic options for the management of hypersalivation in HD patients.
Korean HD expert comment
This symptom is not common. Pathophysiology studies are needed for severe cases with hypersalivation.
Respiratory muscle function
In an RCT in which home-based respiratory muscle training was conducted for four months, the training group presented increased vital capacity and inspiratory and expiratory pressure (Level C) [159]. In a 12-week gym and home walking exercise program, an improvement in the respiratory exchange ratio was observed (Level U) [160].
Summary
Respiratory muscle training and/or exercise may be beneficial for respiratory function in HD patients (Table 4).
Korean HD expert comment
Further studies with respiratory muscle training and rehabilitation exercise are worth being conducted in HD.
Pain
Pain has not been well recognized in HD, with only a few reports described to date [161]. In a large global cohort study, the prevalence of pain was estimated to be 38% in the early stages, which increased as the disease progressed. Furthermore, 26%–29% of HD mutation carriers reported pain interference [162]. Pain in HD may originate from degenerative changes in the central nervous system (including the cortex, subcortex, and spinal cord), degeneration of the peripheral nervous system, and/or associated musculoskeletal problems [161]. However, because cognitive and emotional disturbances can affect the communication ability of HD patients, patients cannot describe their pain symptoms and are therefore often underestimated or neglected [10]. This systematic review did not find any studies reporting effective pain management strategies in HD patients.
Summary
There is no evidence to recommend any therapeutic options for the management of pain in HD patients.
Korean HD expert comment
Physicians need to be alert to pain in HD patients. It has also been suggested that sensitivity to pain and temperature is altered in HD. Even if there is no complaint of pain, physicians need to monitor whether there is any organic damage. Further studies on HD pain are needed.
Weight loss
Weight loss is common in patients with HD at all stages, even in patients who maintain a normal diet [163]. Studies have shown that body mass index (BMI) is significantly lower in individuals with HD than in healthy controls, which is observed from the early stages of the disease [164]. Progressive weight loss is correlated with the number of CAG repeats [165]. Severe weight loss adversely affects daily activities, leading to difficulties in walking, speaking, and swallowing [166]. Several mechanisms have been proposed for weight loss in HD, including an increased metabolic rate due to hypothalamic dysfunction, mitochondrial dysfunction, malnutrition resulting from swallowing difficulties, and increased energy expenditure due to hyperkinetic movements [167-170].
However, evidence from prospective studies on the symptomatic management of weight loss in HD patients is limited. A single-arm observational study indicated that nutritional support in HD patients did not alter BMI, although it did stabilize some anthropometric variables [82] (Level U). The use of mesoridazine did not result in weight gain; however, the use of antipsychotics, including risperidone, was associated with relatively high BMI among HD patients in a cross-sectional study (Level U, Supplementary Material in the online-only Data Supplement). Case series focusing on in-hospital rehabilitation for HD patients with low BMI have shown improvements in BMI and overall functional recovery (Level U, Supplementary Material in the online-only Data Supplement).
Summary
There is insufficient evidence for the management of weight loss in HD patients. Rehabilitation and nutritional support may help improve weight loss in HD patients.
Korean HD expert comment
Providing dietary support, including a high-protein, high-calorie diet, can help maintain the nutritional status and functional outcomes of HD patients. Timely decisions regarding the use of feeding tubes, such as L-tubes or PEG (percutaneous endoscopic gastrostomy), may be crucial in managing weight loss and preventing fatal malnutrition in HD patients.
Dysphagia or difficulty swallowing
Most patients with HD eventually suffer from dysphagia, and aspiration pneumonia is the leading cause of death [171]. One pilot RCT and 4 therapeutic intervention studies were reviewed to evaluate the evidence of available therapies for swallowing difficulty in HD patients [134,159,172-174]. The impact of pharmacological treatment (i.e., olanzapine) has rarely been studied (Level U). There is also insufficient evidence to assess the impact of rehabilitative strategies, including home-based respiratory muscle training programs, compensatory techniques, and speech therapy, as well as nursing interventions (Level U, Supplementary Material in the online-only Data Supplement).
Summary
There are no proven therapeutic strategies to improve dysphagia in HD patients.
Korean HD expert comment
Dysphagia should be assessed from the early stage of the disease to reduce the medical complications of HD. HD patients may develop parkinsonian features in the advanced stages of the disease. At this stage, patients easily develop dysphagia with continuing antipsychotic (such as haloperidol) treatment for their chorea, for whom discontinuation of the offending drug may improve dysphagia.
DISCUSSION
In this review, we systematically compiled clinical evidence pertaining to various motor and nonmotor symptoms of HD. Undoubtedly, HD manifests as an enormous spectrum of motor and nonmotor symptoms that significantly impact the quality of life of both patients and caregivers. While curative treatment for HD remains elusive, numerous studies have explored diverse approaches to provide care and comfort for HD patients.
The pharmacological management of motor symptoms, particularly chorea, has garnered support from well-designed RCTs employing VMAT2 inhibitors, which have demonstrated significant improvements in symptoms and overall quality of life. However, the majority of other motor and nonmotor symptoms lack Level A evidence supporting their clinical benefits. The uniqueness of the current systematic review is that we included open-label trials and case series that may show potential therapeutic benefits in HD patients, which have not been systematically examined in prior reviews. By adopting this approach, we revealed that nonpharmacological interventions such as physical therapy have shown some promise by highlighting potential benefits. From this standpoint, we emphasized studies with multidisciplinary approaches that integrate both pharmacological and nonpharmacological strategies in HD.
However, we found that there is a relative scarcity of well-designed double-blind RCTs, with the exceptions of studies on chorea, depression and dementia. The management of symptoms may have to rely on open-label studies, case series, or case reports. Consequently, management strategies for most symptoms lack robust clinical and statistical power. Alternatively, we included comments from Korean HD experts on each segment of this review, contributing practical insights to this systematic review.
Nonetheless, there is no doubt that well-designed RCTs will contribute to clarifying uncertainties and establishing robust evidence-based guidelines. The relative scarcity of well-designed clinical trials stems largely from the rarity of HD, hindering the feasibility of conducting double-blind placebo-controlled trials on large samples [10]. Therefore, establishing and sustaining HD cohorts on a regional basis, not in a single country, is crucial. Unfortunately, the creation of a multicenter international cohort in Asia has been challenging.
Recently, a novel class of drugs has been developed, including antisense oligonucleotides, small interfering RNAs, splicing modulators, and enzyme replacements, with some currently undergoing clinical trials [175]. Although no novel classes of drugs, including antisense oligonucleotides, have yet proven their clinical effectiveness, these new drugs target disease-modifying effects, in contrast with current therapeutics for symptomatic management of HD. Thus, these new drugs hold promise for the future treatment of HD and should be reviewed in a separate article in the future.
In summary, this systematic review by the task force of the KHDS addressed the currently available options for the symptomatic management of HD. We found that many of the neurologic symptoms of HD patients lack robust evidence for effective management, and there is limited accessibility to various treatment options in South Korea. Unfortunately, this review does not address potential curative or disease-modifying therapies that are currently under investigation. Ongoing and forthcoming clinical trials focused on symptomatic management and disease-modifying therapies should be incorporated into future evidence-based reviews. Finally, more clinical trials are needed in this region to establish comprehensive, evidence-based guidelines for the management of HD.
Acknowledgments
None
Footnotes
Conflicts of Interest
The authors have no financial conflicts of interest.
Funding Statement
None
Author Contributions
Conceptualization: Jung Hwan Shin, Jee-Young Lee. Data curation: Jung Hwan Shin, Hui-Jun Yang, Jong Hyun Ahn, Sungyang Jo, Seok Jong Chung. Formal analysis: Jung Hwan Shin. Funding acquisition: Jee-Young Lee. Investigation: all authors. Methodology: Jung Hwan Shin, Jee-Young Lee. Supervision: Jee-Young Lee, Hyun Sook Kim, Manho Kim. Validation: Jung Hwan Shin, Jee-Young Lee. Visualization: Jung Hwan Shin, Hui-Jun Yang, Jong Hyun Ahn, Sungyang Jo, Seok Jong Chung, Jee-Young Lee. Writing—original draft: Jung Hwan Shin, Hui-Jun Yang, Jong Hyun Ahn, Sungyang Jo, Seok Jong Chung, Jee-Young Lee. Writing—review & editing: all authors.
Supplementary Materials
The online-only Data Supplement is available with this article at https://doi.org/10.14802/jmd.24140.
REFERENCES
- 1.Medina A, Mahjoub Y, Shaver L, Pringsheim T. Prevalence and incidence of Huntington’s disease: an updated systematic review and meta-analysis. Mov Disord. 2022;37:2327–2335. doi: 10.1002/mds.29228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Lee CY, Ro JS, Jung H, Kim M, Jeon B, Lee JY. Increased 10-year prevalence of Huntington’s disease in South Korea: an analysis of medical expenditure through the national healthcare system. J Clin Neurol. 2023;19:147–155. doi: 10.3988/jcn.2022.0212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Paulson HL, Albin RL. Huntington’s disease: clinical features and routes to trherapy. In: Lo DC, Hughes RE. Neurobiology of Huntington’s disease: applications to drug discovery. Boca Raton: CRC Press. 2011;1-28. [Google Scholar]
- 4.Jiang A, Handley RR, Lehnert K, Snell RG. From pathogenesis to therapeutics: a review of 150 years of Huntington’s disease research. Int J Mol Sci. 2023;24:13021. doi: 10.3390/ijms241613021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Kim HJ, Shin CW, Jeon B, Park H. Survival of Korean Huntington’s disease patients. J Mov Disord. 2016;9:166–170. doi: 10.14802/jmd.16022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Rodrigues FB, Abreu D, Damásio J, Goncalves N, Correia-Guedes L, Coelho M, et al. Survival, mortality, causes and places of death in a European Huntington’s disease prospective cohort. Mov Disord Clin Pract. 2017;4:737–742. doi: 10.1002/mdc3.12502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Ready RE, Mathews M, Leserman A, Paulsen JS. Patient and caregiver quality of life in Huntington’s disease. Mov Disord. 2008;23:721–726. doi: 10.1002/mds.21920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. Rev Esp Cardiol (Engl Ed) 2021;74:790–799. doi: 10.1016/j.rec.2021.07.010. [DOI] [PubMed] [Google Scholar]
- 9.French J, Gronseth G. Lost in a jungle of evidence: we need a compass. Neurology. 2008;71:1634–1638. doi: 10.1212/01.wnl.0000336533.19610.1b. [DOI] [PubMed] [Google Scholar]
- 10.Bachoud-Lévi AC, Ferreira J, Massart R, Youssov K, Rosser A, Busse M, et al. International guidelines for the treatment of Huntington’s disease. Front Neurol. 2019;10:710. doi: 10.3389/fneur.2019.00710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Ferreira JJ, Rodrigues FB, Duarte GS, Mestre TA, Bachoud-Levi AC, Bentivoglio AR, et al. An MDS evidence-based review on treatments for Huntington’s disease. Mov Disord. 2022;37:25–35. doi: 10.1002/mds.28855. [DOI] [PubMed] [Google Scholar]
- 12.Coppen EM, Roos RA. Current pharmacological approaches to reduce chorea in Huntington’s disease. Drugs. 2017;77:29–46. doi: 10.1007/s40265-016-0670-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Ondo WG, Tintner R, Thomas M, Jankovic J. Tetrabenazine treatment for Huntington’s disease-associated chorea. Clin Neuropharmacol. 2002;25:300–302. doi: 10.1097/00002826-200211000-00003. [DOI] [PubMed] [Google Scholar]
- 14.Huntington Study Group Tetrabenazine as antichorea therapy in Huntington disease: a randomized controlled trial. Neurology. 2006;66:366–372. doi: 10.1212/01.wnl.0000198586.85250.13. [DOI] [PubMed] [Google Scholar]
- 15.Frank S, Ondo W, Fahn S, Hunter C, Oakes D, Plumb S, et al. A study of chorea after tetrabenazine withdrawal in patients with Huntington disease. Clin Neuropharmacol. 2008;31:127–133. doi: 10.1097/WNF.0b013e3180ca77ea. [DOI] [PubMed] [Google Scholar]
- 16.Kenney C, Hunter C, Davidson A, Jankovic J. Short-term effects of tetrabenazine on chorea associated with Huntington’s disease. Mov Disord. 2007;22:10–13. doi: 10.1002/mds.21161. [DOI] [PubMed] [Google Scholar]
- 17.Frank S. Tetrabenazine as anti-chorea therapy in Huntington disease: an open-label continuation study. Huntington Study Group/TETRA-HD Investigators. BMC Neurol. 2009;9:62. doi: 10.1186/1471-2377-9-62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Reilmann R. Pharmacological treatment of chorea in Huntington’s disease-good clinical practice versus evidence-based guideline. Mov Disord. 2013;28:1030–1033. doi: 10.1002/mds.25500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Szpisjak L, Salamon A, Zadori D, Klivenyi P, Vecsei L. Selecting dopamine depleters for hyperkinetic movement disorders: how do we choose? Expert Opin Pharmacother. 2020;21:1–4. doi: 10.1080/14656566.2019.1685980. [DOI] [PubMed] [Google Scholar]
- 20.Huntington Study Group Effect of deutetrabenazine on chorea among patients with Huntington disease: a randomized clinical trial. JAMA. 2016;316:40–50. doi: 10.1001/jama.2016.8655. [DOI] [PubMed] [Google Scholar]
- 21.Furr Stimming E, Claassen DO, Kayson E, Goldstein J, Mehanna R, Zhang H, et al. Safety and efficacy of valbenazine for the treatment of chorea associated with Huntington’s disease (KINECT-HD): a phase 3, randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2023;22:494–504. doi: 10.1016/S1474-4422(23)00127-8. [DOI] [PubMed] [Google Scholar]
- 22.Frank S, Testa C, Edmondson MC, Goldstein J, Kayson E, Leavitt BR, et al. The safety of deutetrabenazine for chorea in Huntington disease: an open-label extension study. CNS Drugs. 2022;36:1207–1216. doi: 10.1007/s40263-022-00956-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Killoran A, Biglan KM. Current therapeutic options for Huntington’s disease: good clinical practice versus evidence-based approaches? Mov Disord. 2014;29:1404–1413. doi: 10.1002/mds.26014. [DOI] [PubMed] [Google Scholar]
- 24.Burgunder JM, Guttman M, Perlman S, Goodman N, van Kammen DP, Goodman L. An international survey-based algorithm for the pharmacologic treatment of chorea in Huntington’s disease. PLoS Curr. 2011;3:RRN1260. doi: 10.1371/currents.RRN1260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Priller J, Ecker D, Landwehrmeyer B, Craufurd D. A Europe-wide assessment of current medication choices in Huntington’s disease. Mov Disord. 2008;23:1788. doi: 10.1002/mds.22188. [DOI] [PubMed] [Google Scholar]
- 26.Bonuccelli U, Ceravolo R, Maremmani C, Nuti A, Rossi G, Muratorio A. Clozapine in Huntington’s chorea. Neurology. 1994;44:821–823. doi: 10.1212/wnl.44.5.821. [DOI] [PubMed] [Google Scholar]
- 27.Brusa L, Orlacchio A, Moschella V, Iani C, Bernardi G, Mercuri NB. Treatment of the symptoms of Huntington’s disease: preliminary results comparing aripiprazole and tetrabenazine. Mov Disord. 2009;24:126–129. doi: 10.1002/mds.22376. [DOI] [PubMed] [Google Scholar]
- 28.van Vugt JP, Siesling S, Vergeer M, van der Velde EA, Roos RA. Clozapine versus placebo in Huntington’s disease: a double blind randomised comparative study. J Neurol Neurosurg Psychiatry. 1997;63:35–39. doi: 10.1136/jnnp.63.1.35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Schultz JL, Kamholz JA, Nopoulos PC, Killoran A. Comparing risperidone and olanzapine to tetrabenazine for the management of chorea in Huntington disease: an analysis from the enroll-HD database. Mov Disord Clin Pract. 2019;6:132–138. doi: 10.1002/mdc3.12706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Bonelli RM, Mahnert FA, Niederwieser G. Olanzapine for Huntington’s disease: an open label study. Clin Neuropharmacol. 2002;25:263–265. doi: 10.1097/00002826-200209000-00007. [DOI] [PubMed] [Google Scholar]
- 31.Harris KL, Kuan WL, Mason SL, Barker RA. Antidopaminergic treatment is associated with reduced chorea and irritability but impaired cognition in Huntington’s disease (enroll-HD) J Neurol Neurosurg Psychiatry. 2020;91:622–630. doi: 10.1136/jnnp-2019-322038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Verhagen Metman L, Morris MJ, Farmer C, Gillespie M, Mosby K, Wuu J, et al. Huntington’s disease: a randomized, controlled trial using the NMDA-antagonist amantadine. Neurology. 2002;59:694–699. doi: 10.1212/wnl.59.5.694. [DOI] [PubMed] [Google Scholar]
- 33.Lucetti C, Del Dotto P, Gambaccini G, Dell’ Agnello G, Bernardini S, Rossi G, et al. IV amantadine improves chorea in Huntington’s disease: an acute randomized, controlled study. Neurology. 2003;60:1995–1997. doi: 10.1212/01.wnl.0000068165.07883.64. [DOI] [PubMed] [Google Scholar]
- 34.O’Suilleabhain P, Dewey RB., Jr A randomized trial of amantadine in Huntington disease. Arch Neurol. 2003;60:996–998. doi: 10.1001/archneur.60.7.996. [DOI] [PubMed] [Google Scholar]
- 35.Lucetti C, Gambaccini G, Bernardini S, Dell’Agnello G, Petrozzi L, Rossi G, et al. Amantadine in Huntington’s disease: open-label video-blinded study. Neurol Sci. 2002;23(Suppl 2):S83–S84. doi: 10.1007/s100720200081. [DOI] [PubMed] [Google Scholar]
- 36.Heckmann JM, Legg P, Sklar D, Fine J, Bryer A, Kies B. IV amantadine improves chorea in Huntington’s disease: an acute randomized, controlled study. Neurology. 2004;63:597–598. doi: 10.1212/wnl.63.3.597. [DOI] [PubMed] [Google Scholar]
- 37.Jenner P, Katzenschlager R. Apomorphine - pharmacological properties and clinical trials in Parkinson’s disease. Parkinsonism Relat Disord. 2016;33(Suppl 1):S13–S21. doi: 10.1016/j.parkreldis.2016.12.003. [DOI] [PubMed] [Google Scholar]
- 38.Albanese A, Cassetta E, Carretta D, Bentivoglio AR, Tonali P. Acute challenge with apomorphine in Huntington’s disease: a double-blind study. Clin Neuropharmacol. 1995;18:427–434. doi: 10.1097/00002826-199510000-00005. [DOI] [PubMed] [Google Scholar]
- 39.Vitale C, Marconi S, Di Maio L, De Michele G, Longo K, Bonavita V, et al. Short-term continuous infusion of apomorphine hydrochloride for treatment of Huntington’s chorea: a double blind, randomized crossover trial. Mov Disord. 2007;22:2359–2364. doi: 10.1002/mds.21718. [DOI] [PubMed] [Google Scholar]
- 40.Auffret M, Drapier S, Vérin M. The many faces of apomorphine: lessons from the past and challenges for the future. Drugs R D. 2018;18:91–107. doi: 10.1007/s40268-018-0230-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Puri BK, Leavitt BR, Hayden MR, Ross CA, Rosenblatt A, Greenamyre JT, et al. Ethyl-EPA in Huntington disease: a double-blind, randomized, placebo-controlled trial. Neurology. 2005;65:286–292. doi: 10.1212/01.wnl.0000169025.09670.6d. [DOI] [PubMed] [Google Scholar]
- 42.Huntington Study Group TREND-HD Investigators Randomized controlled trial of ethyl-eicosapentaenoic acid in Huntington disease: the TREND-HD study. Arch Neurol. 2008;65:1582–1589. doi: 10.1001/archneur.65.12.1582. [DOI] [PubMed] [Google Scholar]
- 43.Ferreira JJ, Rosser A, Craufurd D, Squitieri F, Mallard N, Landwehrmeyer B. Ethyl-eicosapentaenoic acid treatment in Huntington’s disease: a placebo-controlled clinical trial. Mov Disord. 2015;30:1426–1429. doi: 10.1002/mds.26308. [DOI] [PubMed] [Google Scholar]
- 44.Vaddadi KS, Soosai E, Chiu E, Dingjan P. A randomised, placebo-controlled, double blind study of treatment of Huntington’s disease with unsaturated fatty acids. Neuroreport. 2002;13:29–33. doi: 10.1097/00001756-200201210-00011. [DOI] [PubMed] [Google Scholar]
- 45.Kieburtz K, McDermott MP, Voss TS, Corey-Bloom J, Deuel LM, Dorsey ER, et al. A randomized, placebo-controlled trial of latrepirdine in Huntington disease. Arch Neurol. 2010;67:154–160. doi: 10.1001/archneurol.2009.334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.HORIZON Investigators of the Huntington Study Group and European Huntington’s Disease Network A randomized, double-blind, placebo-controlled study of latrepirdine in patients with mild to moderate Huntington disease. JAMA Neurol. 2013;70:25–33. doi: 10.1001/2013.jamaneurol.382. [DOI] [PubMed] [Google Scholar]
- 47.McGarry A, McDermott M, Kieburtz K, de Blieck EA, Beal F, Marder K, et al. A randomized, double-blind, placebo-controlled trial of coenzyme Q10 in Huntington disease. Neurology. 2017;88:152–159. doi: 10.1212/WNL.0000000000003478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Huntington Study Group A randomized, placebo-controlled trial of coenzyme Q10 and remacemide in Huntington’s disease. Neurology. 2001;57:397–404. doi: 10.1212/wnl.57.3.397. [DOI] [PubMed] [Google Scholar]
- 49.Tabrizi SJ, Blamire AM, Manners DN, Rajagopalan B, Styles P, Schapira AH, et al. Creatine therapy for Huntington’s disease: clinical and MRS findings in a 1-year pilot study. Neurology. 2003;61:141–142. doi: 10.1212/01.wnl.0000070186.97463.a7. [DOI] [PubMed] [Google Scholar]
- 50.Verbessem P, Lemiere J, Eijnde BO, Swinnen S, Vanhees L, Van Leemputte M, et al. Creatine supplementation in Huntington’s disease: a placebo-controlled pilot trial. Neurology. 2003;61:925–930. doi: 10.1212/01.wnl.0000090629.40891.4b. [DOI] [PubMed] [Google Scholar]
- 51.Bender A, Auer DP, Merl T, Reilmann R, Saemann P, Yassouridis A, et al. Creatine supplementation lowers brain glutamate levels in Huntington’s disease. J Neurol. 2005;252:36–41. doi: 10.1007/s00415-005-0595-4. [DOI] [PubMed] [Google Scholar]
- 52.Tabrizi SJ, Blamire AM, Manners DN, Rajagopalan B, Styles P, Schapira AH, et al. High-dose creatine therapy for Huntington disease: a 2-year clinical and MRS study. Neurology. 2005;64:1655–1656. doi: 10.1212/01.WNL.0000160388.96242.77. [DOI] [PubMed] [Google Scholar]
- 53.Hersch SM, Gevorkian S, Marder K, Moskowitz C, Feigin A, Cox M, et al. Creatine in Huntington disease is safe, tolerable, bioavailable in brain and reduces serum 8OH2’dG. Neurology. 2006;66:250–252. doi: 10.1212/01.wnl.0000194318.74946.b6. [DOI] [PubMed] [Google Scholar]
- 54.Hersch SM, Schifitto G, Oakes D, Bredlau AL, Meyers CM, Nahin R, et al. The CREST-E study of creatine for Huntington disease: a randomized controlled trial. Neurology. 2017;89:594–601. doi: 10.1212/WNL.0000000000004209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Feigin A, Evans EE, Fisher TL, Leonard JE, Smith ES, Reader A, et al. Pepinemab antibody blockade of SEMA4D in early Huntington’s disease: a randomized, placebo-controlled, phase 2 trial. Nat Med. 2022;28:2183–2193. doi: 10.1038/s41591-022-01919-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Huntington Study Group Reach2HD Investigators Safety, tolerability, and efficacy of PBT2 in Huntington’s disease: a phase 2, randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2015;14:39–47. doi: 10.1016/S1474-4422(14)70262-5. [DOI] [PubMed] [Google Scholar]
- 57.Kloberg A, Constantinescu R, Nilsson MK, Carlsson ML, Carlsson A, Wahlström J, et al. Tolerability and efficacy of the monoaminergic stabilizer (-)-OSU6162 (PNU-96391A) in Huntington’s disease: a double-blind cross-over study. Acta Neuropsychiatr. 2014;26:298–306. doi: 10.1017/neu.2014.16. [DOI] [PubMed] [Google Scholar]
- 58.Reilmann R, Rouzade-Dominguez ML, Saft C, Süssmuth SD, Priller J, Rosser A, et al. A randomized, placebo-controlled trial of AFQ056 for the treatment of chorea in Huntington’s disease. Mov Disord. 2015;30:427–431. doi: 10.1002/mds.26174. [DOI] [PubMed] [Google Scholar]
- 59.Beglinger LJ, Adams WH, Paulson H, Fiedorowicz JG, Langbehn DR, Duff K, et al. Randomized controlled trial of atomoxetine for cognitive dysfunction in early Huntington disease. J Clin Psychopharmacol. 2009;29:484–487. doi: 10.1097/JCP.0b013e3181b2ac0a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Gelderblom H, Wüstenberg T, McLean T, Mütze L, Fischer W, Saft C, et al. Bupropion for the treatment of apathy in Huntington’s disease: a multicenter, randomised, double-blind, placebo-controlled, prospective crossover trial. PLoS One. 2017;12:e0173872. doi: 10.1371/journal.pone.0173872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.López-Sendón Moreno JL, García Caldentey J, Trigo Cubillo P, Ruiz Romero C, García Ribas G, Alonso Arias MA, et al. A double-blind, randomized, cross-over, placebo-controlled, pilot trial with Sativex in Huntington’s disease. J Neurol. 2016;263:1390–1400. doi: 10.1007/s00415-016-8145-9. [DOI] [PubMed] [Google Scholar]
- 62.Beglinger LJ, Adams WH, Langbehn D, Fiedorowicz JG, Jorge R, Biglan K, et al. Results of the citalopram to enhance cognition in Huntington disease trial. Mov Disord. 2014;29:401–405. doi: 10.1002/mds.25750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Fernandez HH, Friedman JH, Grace J, Beason-Hazen S. Donepezil for Huntington’s disease. Mov Disord. 2000;15:173–176. doi: 10.1002/1531-8257(200001)15:1<173::aid-mds1032>3.0.co;2-t. [DOI] [PubMed] [Google Scholar]
- 64.Cubo E, Shannon KM, Tracy D, Jaglin JA, Bernard BA, Wuu J, et al. Effect of donepezil on motor and cognitive function in Huntington disease. Neurology. 2006;67:1268–1271. doi: 10.1212/01.wnl.0000238106.10423.00. [DOI] [PubMed] [Google Scholar]
- 65.Como PG, Rubin AJ, O’Brien CF, Lawler K, Hickey C, Rubin AE, et al. A controlled trial of fluoxetine in nondepressed patients with Huntington’s disease. Mov Disord. 1997;12:397–401. doi: 10.1002/mds.870120319. [DOI] [PubMed] [Google Scholar]
- 66.Kremer B, Clark CM, Almqvist EW, Raymond LA, Graf P, Jacova C, et al. Influence of lamotrigine on progression of early Huntington disease: a randomized clinical trial. Neurology. 1999;53:1000–1011. doi: 10.1212/wnl.53.5.1000. [DOI] [PubMed] [Google Scholar]
- 67.Huntington Study Group Safety and tolerability of the free-radical scavenger OPC-14117 in Huntington’s disease. Neurology. 1998;50:1366–1373. doi: 10.1212/wnl.50.5.1366. [DOI] [PubMed] [Google Scholar]
- 68.van Diemen MPJ, Hart EP, Abbruscato A, Mead L, van Beelen I, Bergheanu SC, et al. Safety, pharmacokinetics and pharmacodynamics of SBT-020 in patients with early stage Huntington’s disease, a 2-part study. Br J Clin Pharmacol. 2021;87:2290–2302. doi: 10.1111/bcp.14656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Süssmuth SD, Haider S, Landwehrmeyer GB, Farmer R, Frost C, Tripepi G, et al. An exploratory double-blind, randomized clinical trial with selisistat, a SirT1 inhibitor, in patients with Huntington’s disease. Br J Clin Pharmacol. 2015;79:465–476. doi: 10.1111/bcp.12512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Müller-Vahl KR, Schneider U, Emrich HM. Nabilone increases choreatic movements in Huntington’s disease. Mov Disord. 1999;14:1038–1040. doi: 10.1002/1531-8257(199911)14:6<1038::aid-mds1024>3.0.co;2-7. [DOI] [PubMed] [Google Scholar]
- 71.Curtis A, Mitchell I, Patel S, Ives N, Rickards H. A pilot study using nabilone for symptomatic treatment in Huntington’s disease. Mov Disord. 2009;24:2254–2259. doi: 10.1002/mds.22809. [DOI] [PubMed] [Google Scholar]
- 72.Busse M, Quinn L, Debono K, Jones K, Collett J, Playle R, et al. A randomized feasibility study of a 12-week community-based exercise program for people with Huntington’s disease. J Neurol Phys Ther. 2013;37:149–158. doi: 10.1097/NPT.0000000000000016. [DOI] [PubMed] [Google Scholar]
- 73.Khalil H, Quinn L, van Deursen R, Dawes H, Playle R, Rosser A, et al. What effect does a structured home-based exercise programme have on people with Huntington’s disease? A randomized, controlled pilot study. Clin Rehabil. 2013;27:646–658. doi: 10.1177/0269215512473762. [DOI] [PubMed] [Google Scholar]
- 74.Thompson JA, Cruickshank TM, Penailillo LE, Lee JW, Newton RU, Barker RA, et al. The effects of multidisciplinary rehabilitation in patients with early-to-middle-stage Huntington’s disease: a pilot study. Eur J Neurol. 2013;20:1325–1329. doi: 10.1111/ene.12053. [DOI] [PubMed] [Google Scholar]
- 75.Bohlen S, Ekwall C, Hellström K, Vesterlin H, Björnefur M, Wiklund L, et al. Physical therapy in Huntington’s disease--toward objective assessments? Eur J Neurol. 2013;20:389–393. doi: 10.1111/j.1468-1331.2012.03760.x. [DOI] [PubMed] [Google Scholar]
- 76.Quinn L, Debono K, Dawes H, Rosser AE, Nemeth AH, Rickards H, et al. Task-specific training in Huntington disease: a randomized controlled feasibility trial. Phys Ther. 2014;94:1555–1568. doi: 10.2522/ptj.20140123. [DOI] [PubMed] [Google Scholar]
- 77.Quinn L, Hamana K, Kelson M, Dawes H, Collett J, Townson J, et al. A randomized, controlled trial of a multi-modal exercise intervention in Huntington’s disease. Parkinsonism Relat Disord. 2016;31:46–52. doi: 10.1016/j.parkreldis.2016.06.023. [DOI] [PubMed] [Google Scholar]
- 78.Busse M, Quinn L, Drew C, Kelson M, Trubey R, McEwan K, et al. Physical activity self-management and coaching compared to social interaction in Huntington disease: results from the ENGAGE-HD randomized, controlled pilot feasibility trial. Phys Ther. 2017;97:625–639. doi: 10.1093/ptj/pzx031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Kegelmeyer DA, Kostyk SK, Fritz NE, Scharre DW, Young GS, Tan Y, et al. Immediate effects of treadmill walking in individuals with Lewy body dementia and Huntington’s disease. Gait Posture. 2021;86:186–191. doi: 10.1016/j.gaitpost.2021.03.016. [DOI] [PubMed] [Google Scholar]
- 80.Quinn L, Playle R, Drew CJG, Taiyari K, Williams-Thomas R, Muratori LM, et al. Physical activity and exercise outcomes in Huntington’s disease (PACE-HD): results of a 12-month trial-within-cohort feasibility study of a physical activity intervention in people with Huntington’s disease. Parkinsonism Relat Disord. 2022;101:75–89. doi: 10.1016/j.parkreldis.2022.06.013. [DOI] [PubMed] [Google Scholar]
- 81.Ringqvist K, Borg K, Möller MC. Tolerability and psychological effects of a multimodal day-care rehabilitation program for persons with Huntington’s disease. J Rehabil Med. 2021;53:jrm00143. doi: 10.2340/16501977-2748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Trejo A, Boll MC, Alonso ME, Ochoa A, Velásquez L. Use of oral nutritional supplements in patients with Huntington’s disease. Nutrition. 2005;21:889–894. doi: 10.1016/j.nut.2004.12.012. [DOI] [PubMed] [Google Scholar]
- 83.Aganzo M, Montojo MT, López de Las Hazas MC, Martínez-Descals A, Ricote-Vila M, Sanz R, et al. Customized dietary intervention avoids unintentional weight loss and modulates circulating miRNAs footprint in Huntington’s disease. Mol Nutr Food Res. 2018;62:e1800619. doi: 10.1002/mnfr.201800619. [DOI] [PubMed] [Google Scholar]
- 84.Schneider F, Darpo B, Loupe PS, Xue H, Knebel H, Gutierrez M, et al. Evaluation of deutetrabenazine’s potential to delay cardiac repolarization using concentration-QTc analysis. Clin Pharmacol Drug Dev. 2023;12:94–106. doi: 10.1002/cpdd.1161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Huntington Study Group Dosage effects of riluzole in Huntington’s disease: a multicenter placebo-controlled study. Neurology. 2003;61:1551–1556. doi: 10.1212/01.wnl.0000096019.71649.2b. [DOI] [PubMed] [Google Scholar]
- 86.Huntington Study Group Minocycline safety and tolerability in Huntington disease. Neurology. 2004;63:547–549. doi: 10.1212/01.wnl.0000133403.30559.ff. [DOI] [PubMed] [Google Scholar]
- 87.Kloos AD, Fritz NE, Kostyk SK, Young GS, Kegelmeyer DA. Video game play (dance dance revolution) as a potential exercise therapy in Huntington’s disease: a controlled clinical trial. Clin Rehabil. 2013;27:972–982. doi: 10.1177/0269215513487235. [DOI] [PubMed] [Google Scholar]
- 88.Ciancarelli I, Tozzi Ciancarelli MG, Carolei A. Effectiveness of intensive neurorehabilitation in patients with Huntington’s disease. Eur J Phys Rehabil Med. 2013;49:189–195. [PubMed] [Google Scholar]
- 89.Brabcová L, Roth J, Ulmanová O, Horáček O, Kolářová M, Božková H, et al. K26 specific in-patient rehabilitation improves postural and gait instability in Huntington’s disease. J Neurol Neurosurg Psychiatry. 2016;87(Suppl 1):A88. [Google Scholar]
- 90.Brabcová L, Roth J, Ulmanová O, Rusz J, Klempíř J, Horáček O, et al. Inpatient multidisciplinary rehabilitation programme for postural and gait stability in Huntington’s disease – a pilot study. Cesk Slov Neurol N. 2019;82:301–308. [Google Scholar]
- 91.Mirek E, Filip M, Banaszkiewicz K, Rudzińska M, Szymura J, Pasiut S, et al. The effects of physiotherapy with PNF concept on gait and balance of patients with Huntington’s disease - pilot study. Neurol Neurochir Pol. 2015;49:354–357. doi: 10.1016/j.pjnns.2015.09.002. [DOI] [PubMed] [Google Scholar]
- 92.Fritz NE, Busse M, Jones K, Khalil H, Quinn L. A classification system to guide physical therapy management in Huntington disease: a case series. J Neurol Phys Ther. 2017;41:156–163. doi: 10.1097/NPT.0000000000000188. [DOI] [PubMed] [Google Scholar]
- 93.Mirek E, Filip M, Chwała W, Szymura J, Pasiut S, Banaszkiewicz K, et al. The influence of motor ability rehabilitation on temporal-spatial parameters of gait in Huntington’s disease patients on the basis of a three-dimensional motion analysis system: an experimental trial. Neurol Neurochir Pol. 2018;52:575–580. doi: 10.1016/j.pjnns.2018.02.001. [DOI] [PubMed] [Google Scholar]
- 94.Clark D, Danzl MM, Ulanowski E. Development of a community-based exercise program for people diagnosed and at-risk for Huntington’s disease: a clinical report. Physiother Theory Pract. 2016;32:232–239. doi: 10.3109/09593985.2015.1110738. [DOI] [PubMed] [Google Scholar]
- 95.Capato TTC, Haddad MS, Piemonte MEP, Barbosa ER. Multisensory cues could be effective to improve balance and gait in Huntingtons disease? Mov Disord. 2015;30(suppl 1):S529. [Google Scholar]
- 96.Capato T, Haddad M, Piemonte ME, Barbosa ER. A proposal for a physiotherapy programme to improve gait, balance and functional independence in Huntington’s disease. Mov Disord. 2012;27(suppl 1):S52. [Google Scholar]
- 97.Capato TTC, Cury RG, Tornai J, Fonoff ET, Guimarães R, Jacobsen MT, et al. Use of objective outcomes measures to verify the effects of ICF-based gait treatment in Huntington’s disease patient on globus pallidus deep brain stimulation: a case report. Front Rehabil Sci. 2022;3:849333. doi: 10.3389/fresc.2022.849333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.de Yebenes JG, Landwehrmeyer B, Squitieri F, Reilmann R, Rosser A, Barker RA, et al. Pridopidine for the treatment of motor function in patients with Huntington’s disease (MermaiHD): a phase 3, randomised, double-blind, placebo-controlled trial. Lancet Neurol. 2011;10:1049–1057. doi: 10.1016/S1474-4422(11)70233-2. [DOI] [PubMed] [Google Scholar]
- 99.Capato TTC, Cury RG, Gimaaes R, Fonoff ET, Haddad MS. Functional motor outcomes after globus pallidus internus deep brain stimulation in Huntington’s disease - a case report. Mov Disord. 2016;31(suppl 2):S47–S48. [Google Scholar]
- 100.Cislaghi G, Capiluppi E, Saleh C, Romano L, Servello D, Mariani C, et al. Bilateral globus pallidus stimulation in Westphal variant of Huntington disease. Neuromodulation. 2014;17:502–505. doi: 10.1111/ner.12098. [DOI] [PubMed] [Google Scholar]
- 101.Kang GA, Heath S, Rothlind J, Starr PA. Long-term follow-up of pallidal deep brain stimulation in two cases of Huntington’s disease. J Neurol Neurosurg Psychiatry. 2011;82:272–277. doi: 10.1136/jnnp.2009.202903. [DOI] [PubMed] [Google Scholar]
- 102.Gruber D, Kuhn AA, Schoenecker T, Kopp UA, Kivi A, Huebl J, et al. Quadruple deep brain stimulation in Huntington’s disease, targeting pallidum and subthalamic nucleus: case report and review of the literature. J Neural Transm (Vienna) 2014;121:1303–1312. doi: 10.1007/s00702-014-1201-7. [DOI] [PubMed] [Google Scholar]
- 103.Reuter I, Hu MT, Andrews TC, Brooks DJ, Clough C, Chaudhuri KR. Late onset levodopa responsive Huntington’s disease with minimal chorea masquerading as Parkinson plus syndrome. J Neurol Neurosurg Psychiatry. 2000;68:238–241. doi: 10.1136/jnnp.68.2.238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Moss JH, Stewart DE. Iatrogenic parkinsonism in Huntington’s chorea. Can J Psychiatry. 1986;31:865–866. doi: 10.1177/070674378603100916. [DOI] [PubMed] [Google Scholar]
- 105.Kenney C, Hunter C, Jankovic J. Long-term tolerability of tetrabenazine in the treatment of hyperkinetic movement disorders. Mov Disord. 2007;22:193–197. doi: 10.1002/mds.21222. [DOI] [PubMed] [Google Scholar]
- 106.Shternberg EI, Smulevich AB. [Experience in the use of neuroleptic substances in chronic progressive chorea (Huntington’s chorea)] Zh Nevropatol Psikhiatr Im S S Korsakova. 1963;63:96–102. Russian. [PubMed] [Google Scholar]
- 107.Zesiewicz TA, Sanchez-Ramos J, Sullivan KL, Hauser RA. Levetiracetam-induced parkinsonism in a Huntington disease patient. Clin Neuropharmacol. 2005;28:188–190. doi: 10.1097/01.wnf.0000169732.00690.32. [DOI] [PubMed] [Google Scholar]
- 108.Hadden R, Sung V. A movement disorders paradox: case report of a patient with gene-positive Huntington’s disease who presented with DaTScan positive and levodopa responsive Parkinson’s disease. Neurology. 2015;84(14 supplement):P3.031. [Google Scholar]
- 109.Racette BA, Perlmutter JS. Levodopa responsive parkinsonism in an adult with Huntington’s disease. J Neurol Neurosurg Psychiatry. 1998;65:577–579. doi: 10.1136/jnnp.65.4.577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Heim B, Bajaj S, De Marzi R, Mangesius S, Djamshidian A, Poewe W, et al. Nabilone in Huntington’s disease: a case series of five patients. J Neurol Neurosurg Psychiatry. 2016;87:A103. [Google Scholar]
- 111.Clarence-Smith K, Shen V, Hunter C, Jankovic J. Concomitant use of antidepressants and neuroleptics with tetrabenazine during treatment of Huntington’s disease (HD) Neurology. 2012;78(suppl 1):P06.039. [Google Scholar]
- 112.Ondo W, Arif C, Williams B, Podel K. Tetrabenazine: impact on impulsivity scales in Huntington’s disease. Neurotherapeutics. 2020;17(Suppl 1):S36. [Google Scholar]
- 113.Gordon M, Testa C, Whaley J, Gross N, Chen M, Alexander J. Long-term efficacy and safety of deutetrabenazine (DTBZ) in patients with chorea associated with Huntington’s disease (HD) Mov Disord Clin Pract. 2022;9(suppl 1):S22–S23. [Google Scholar]
- 114.Molnar MJ, Molnar V, Fedor M, Csehi R, Acsai K, Borsos B, et al. Improving mood and cognitive symptoms in Huntington’s disease with cariprazine treatment. Front Psychiatry. 2022;12:825532. doi: 10.3389/fpsyt.2021.825532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Levy R, Czernecki V. Apathy and the basal ganglia. J Neurol. 2006;253(Suppl 7):vii54–vii61. doi: 10.1007/s00415-006-7012-5. [DOI] [PubMed] [Google Scholar]
- 116.Duff K, Beglinger LJ, O’Rourke ME, Nopoulos P, Paulson HL, Paulsen JS. Risperidone and the treatment of psychiatric, motor, and cognitive symptoms in Huntington’s disease. Ann Clin Psychiatry. 2008;20:1–3. doi: 10.1080/10401230701844802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Pérez-Pérez J, Martínez-Horta S, Pagonabarraga J, Carceller M, Kulisevsky J. Rasagiline for the treatment of parkinsonism in Huntington’s disease. Parkinsonism Relat Disord. 2015;21:340–342. doi: 10.1016/j.parkreldis.2015.01.002. [DOI] [PubMed] [Google Scholar]
- 118.Saft C, von Hein SM, Lücke T, Thiels C, Peball M, Djamshidian A, et al. Cannabinoids for treatment of dystonia in Huntington’s disease. J Huntingtons Dis. 2018;7:167–173. doi: 10.3233/JHD-170283. [DOI] [PubMed] [Google Scholar]
- 119.Trinkler I, Chéhère P, Salgues J, Monin ML, Tezenas du Montcel S, Khani S, et al. Contemporary dance practice improves motor function and body representation in Huntington’s disease: a pilot study. J Huntingtons Dis. 2019;8:97–110. doi: 10.3233/JHD-180315. [DOI] [PubMed] [Google Scholar]
- 120.Phillips MCL, McManus EJ, Brinkhuis M, Romero-Ferrando B. Time-restricted ketogenic diet in Huntington’s disease: a case study. Front Behav Neurosci. 2022;16:931636. doi: 10.3389/fnbeh.2022.931636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Cummings JL. Behavioral and psychiatric symptoms associated with Huntington’s disease. Adv Neurol. 1995;65:179–186. [PubMed] [Google Scholar]
- 122.Wetzel HH, Gehl CR, Dellefave-Castillo L, Schiffman JF, Shannon KM, Paulsen JS, et al. Suicidal ideation in Huntington disease: the role of comorbidity. Psychiatry Res. 2011;188:372–376. doi: 10.1016/j.psychres.2011.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.McGarry A, McDermott MP, Kieburtz K, Fung WLA, McCusker E, de Blieck EA, et al. Risk factors for suicidality in Huntington disease in the context of a randomized clinical trial: an analysis of the 2CARE Study. Neurotherapeutics. 2018;15:1188. [Google Scholar]
- 124.Brownstein MJ, Simon NG, Long JD, Yankey J, Maibach HT, Cudkowicz M, et al. Safety and tolerability of SRX246, a vasopressin 1a antagonist, in irritable Huntington’s disease patients—a randomized phase 2 clinical trial. J Clin Med. 2020;9:3682. doi: 10.3390/jcm9113682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Anderson K, Craufurd D, Edmondson MC, Goodman N, Groves M, van Duijn E, et al. An international survey-based algorithm for the pharmacologic treatment of obsessive-compulsive behaviors in Huntington’s disease. PLoS Curr. 2011;3:RRN1261. doi: 10.1371/currents.RRN1261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Meyer M, Barreault L, Frismand S, Hingray C. Benefit of cognitive behavioral therapy for post-traumatic stress disorder and obsessive-compulsive disorders in Huntington’s disease: a case report. Neurocase. 2022;28:188–193. doi: 10.1080/13554794.2022.2051562. [DOI] [PubMed] [Google Scholar]
- 127.Testa CM, Jones K, Slobodnikova Z, Jones SR, Wood-Siverio C, Factor SA, et al. The safety and effectiveness of aripiprazole for treating irritability and aggression in Huntington disease: a double-blind placebo-controlled pilot study. Neurotherapeutics. 2016;13:249–250. [Google Scholar]
- 128.Lin WC, Chou YH. Aripiprazole effects on psychosis and chorea in a patient with Huntington’s disease. Am J Psychiatry. 2008;165:1207–1208. doi: 10.1176/appi.ajp.2008.08040503. [DOI] [PubMed] [Google Scholar]
- 129.Ciammola A, Sassone J, Colciago C, Mencacci NE, Poletti B, Ciarmiello A, et al. Aripiprazole in the treatment of Huntington’s disease: a case series. Neuropsychiatr Dis Treat. 2009;5:1–4. [PMC free article] [PubMed] [Google Scholar]
- 130.Leonard DP, Kidson MA, Brown JG, Shannon PJ, Taryan S. A double blind trial of lithium carbonate and haloperidol in Huntington’s chorea. Aust N Z J Psychiatry. 1975;9:115–118. doi: 10.3109/00048677509159834. [DOI] [PubMed] [Google Scholar]
- 131.Ranen NG, Lipsey JR, Treisman G, Ross CA. Sertraline in the treatment of severe aggressiveness in Huntington’s disease. J Neuropsychiatry Clin Neurosci. 1996;8:338–340. doi: 10.1176/jnp.8.3.338. [DOI] [PubMed] [Google Scholar]
- 132.Raulerson S, Berg S, Costigan M, Hersch S, Anderson K, Long J, et al. Safety and tolerability of SRX246 in irritable/aggressive subjects with Huntington’s disease (stair): a phase II exploratory clinical trial. Neurotherapeutics. 2018;15:236. [Google Scholar]
- 133.Ranen NG, Peyser CE, Folstein SE. ECT as a treatment for depression in Huntington’s disease. J Neuropsychiatry Clin Neurosci. 1994;6:154–159. doi: 10.1176/jnp.6.2.154. [DOI] [PubMed] [Google Scholar]
- 134.Squitieri F, Cannella M, Porcellini A, Brusa L, Simonelli M, Ruggieri S. Short-term effects of olanzapine in Huntington disease. Neuropsychiatry Neuropsychol Behav Neurol. 2001;14:69–72. [PubMed] [Google Scholar]
- 135.Silver A. Cognitive-behavioural therapy with a Huntington’s gene positive patient. Patient Educ Couns. 2003;49:133–138. doi: 10.1016/s0738-3991(02)00070-8. [DOI] [PubMed] [Google Scholar]
- 136.Lundin A, Dietrichs E, Haghighi S, Göller ML, Heiberg A, Loutfi G, et al. Efficacy and safety of the dopaminergic stabilizer pridopidine (ACR16) in patients with Huntington’s disease. Clin Neuropharmacol. 2010;33:260–264. doi: 10.1097/WNF.0b013e3181ebb285. [DOI] [PubMed] [Google Scholar]
- 137.Bartlett DM, Dominguez D JF, Lazar AS, Kordsachia CC, Rankin TJ, Lo J, et al. Multidisciplinary rehabilitation reduces hypothalamic grey matter volume loss in individuals with preclinical Huntington’s disease: a nine-month pilot study. J Neurol Sci. 2020;408:116522. doi: 10.1016/j.jns.2019.116522. [DOI] [PubMed] [Google Scholar]
- 138.Jhanjee A, Anand KS, Bajaj BK. Hypersexual features in Huntington’s disease. Singapore Med J. 2011;52:e131–e133. [PubMed] [Google Scholar]
- 139.Foroud T, Siemers E, Kleindorfer D, Bill DJ, Hodes ME, Norton JA, et al. Cognitive scores in carriers of Huntington’s disease gene compared to noncarriers. Ann Neurol. 1995;37:657–664. doi: 10.1002/ana.410370516. [DOI] [PubMed] [Google Scholar]
- 140.Snowden JS. The neuropsychology of Huntington’s disease. Arch Clin Neuropsychol. 2017;32:876–887. doi: 10.1093/arclin/acx086. [DOI] [PubMed] [Google Scholar]
- 141.McGarry A, Kieburtz K, Abler V, Grachev ID, Gandhi S, Auinger P, et al. Safety and exploratory efficacy at 36 months in Open-HART, an open-label extension study of pridopidine in Huntington’s disease. J Huntingtons Dis. 2017;6:189–199. doi: 10.3233/JHD-170241. [DOI] [PubMed] [Google Scholar]
- 142.Reilmann R, McGarry A, Grachev ID, Savola JM, Borowsky B, Eyal E, et al. Safety and efficacy of pridopidine in patients with Huntington’s disease (PRIDE-HD): a phase 2, randomised, placebo-controlled, multicentre, dose-ranging study. Lancet Neurol. 2019;18:165–176. doi: 10.1016/S1474-4422(18)30391-0. [DOI] [PubMed] [Google Scholar]
- 143.Sešok S, Bolle N, Kobal J, Bucik V, Vodušek DB. Cognitive function in early clinical phase huntington disease after rivastigmine treatment. Psychiatr Danub. 2014;26:239–248. [PubMed] [Google Scholar]
- 144.de Tommaso M, Difruscolo O, Sciruicchio V, Specchio N, Livrea P. Two years’ follow-up of rivastigmine treatment in Huntington disease. Clin Neuropharmacol. 2007;30:43–46. doi: 10.1097/01.wnf.0000240945.44370.f0. [DOI] [PubMed] [Google Scholar]
- 145.Cruickshank TM, Thompson JA, Domínguez D JF, Reyes AP, Bynevelt M, Georgiou-Karistianis N, et al. The effect of multidisciplinary rehabilitation on brain structure and cognition in Huntington’s disease: an exploratory study. Brain Behav. 2015;5:e00312. doi: 10.1002/brb3.312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Piira A, van Walsem MR, Mikalsen G, Nilsen KH, Knutsen S, Frich JC. Effects of a one year intensive multidisciplinary rehabilitation program for patients with Huntington’s disease: a prospective intervention study. PLoS Curr. 2013;5:ecurrents.hd.9504af71e0d1f87830c25c394be47027. doi: 10.1371/currents.hd.9504af71e0d1f87830c25c394be47027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Vesper J, Groiss S, Schnitzler A, Wojtecki L. A prospective trial for pallidal deep brain stimulation in Huntington’s disease. J Neurol Neurosurg Psychiatry. 2016;87:A103–A104. [Google Scholar]
- 148.Zittel S, Tadic V, Moll CKE, Bäumer T, Fellbrich A, Gulberti A, et al. Prospective evaluation of globus pallidus internus deep brain stimulation in Huntington’s disease. Parkinsonism Relat Disord. 2018;51:96–100. doi: 10.1016/j.parkreldis.2018.02.030. [DOI] [PubMed] [Google Scholar]
- 149.Gonzalez V, Cif L, Biolsi B, Garcia-Ptacek S, Seychelles A, Sanrey E, et al. Deep brain stimulation for Huntington’s disease: long-term results of a prospective open-label study. J Neurosurg. 2014;121:114–122. doi: 10.3171/2014.2.JNS131722. [DOI] [PubMed] [Google Scholar]
- 150.Eddy CM, Shapiro K, Clouter A, Hansen PC, Rickards HE. Transcranial direct current stimulation can enhance working memory in Huntington’s disease. Prog Neuropsychopharmacol Biol Psychiatry. 2017;77:75–82. doi: 10.1016/j.pnpbp.2017.04.002. [DOI] [PubMed] [Google Scholar]
- 151.Jaini A, Yomtoob J, Yeh C, Bega D. Understanding HD psychosis: an analysis from the ENROLL-HD database. Tremor Other Hyperkinet Mov (N Y) 2020;10:16. doi: 10.5334/tohm.395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Moser AD, Epping E, Espe-Pfeifer P, Martin E, Zhorne L, Mathews K, et al. A survey-based study identifies common but unrecognized symptoms in a large series of juvenile Huntington’s disease. Neurodegener Dis Manag. 2017;7:307–315. doi: 10.2217/nmt-2017-0019. [DOI] [PubMed] [Google Scholar]
- 153.Mustafa FA. Misdiagnosis of Huntington’s disease. Lancet Psychiatry. 2017;4:21. doi: 10.1016/S2215-0366(16)30355-8. [DOI] [PubMed] [Google Scholar]
- 154.Alpay M, Koroshetz WJ. Quetiapine in the treatment of behavioral disturbances in patients with Huntington’s disease. Psychosomatics. 2006;47:70–72. doi: 10.1176/appi.psy.47.1.70. [DOI] [PubMed] [Google Scholar]
- 155.Tan EK, Jankovic J, Ondo W. Bruxism in Huntington’s disease. Mov Disord. 2000;15:171–173. doi: 10.1002/1531-8257(200001)15:1<171::aid-mds1031>3.0.co;2-y. [DOI] [PubMed] [Google Scholar]
- 156.Louis ED, Tampone E. Bruxism in Huntington’s disease. Mov Disord. 2001;16:785. doi: 10.1002/mds.1129. [DOI] [PubMed] [Google Scholar]
- 157.Nash MC, Ferrell RB, Lombardo MA, Williams RB. Treatment of bruxism in Huntington’s disease with botulinum toxin. J Neuropsychiatry Clin Neurosci. 2004;16:381–382. doi: 10.1176/jnp.16.3.381-a. [DOI] [PubMed] [Google Scholar]
- 158.Lakraj AA, Moghimi N, Jabbari B. Sialorrhea: anatomy, pathophysiology and treatment with emphasis on the role of botulinum toxins. Toxins (Basel) 2013;5:1010–1031. doi: 10.3390/toxins5051010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Reyes A, Cruickshank T, Nosaka K, Ziman M. Respiratory muscle training on pulmonary and swallowing function in patients with Huntington’s disease: a pilot randomised controlled trial. Clin Rehabil. 2015;29:961–973. doi: 10.1177/0269215514564087. [DOI] [PubMed] [Google Scholar]
- 160.Dawes H, Collett J, Debono K, Quinn L, Jones K, Kelson MJ, et al. Exercise testing and training in people with Huntington’s disease. Clin Rehabil. 2015;29:196–206. doi: 10.1177/0269215514540921. [DOI] [PubMed] [Google Scholar]
- 161.Li J, Wang Y, Yang R, Ma W, Yan J, Li Y, et al. Pain in Huntington’s disease and its potential mechanisms. Front Aging Neurosci. 2023;15:1190563. doi: 10.3389/fnagi.2023.1190563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Sprenger GP, Roos RAC, van Zwet E, Reijntjes RH, Achterberg WP, de Bot ST. The prevalence of pain in Huntington’s disease in a large worldwide cohort. Parkinsonism Relat Disord. 2021;89:73–78. doi: 10.1016/j.parkreldis.2021.06.015. [DOI] [PubMed] [Google Scholar]
- 163.Robbins AO, Ho AK, Barker RA. Weight changes in Huntington’s disease. Eur J Neurol. 2006;13:e7. doi: 10.1111/j.1468-1331.2006.01319.x. [DOI] [PubMed] [Google Scholar]
- 164.Djoussé L, Knowlton B, Cupples LA, Marder K, Shoulson I, Myers RH. Weight loss in early stage of Huntington’s disease. Neurology. 2002;59:1325–1330. doi: 10.1212/01.wnl.0000031791.10922.cf. [DOI] [PubMed] [Google Scholar]
- 165.Aziz NA, van der Burg JM, Landwehrmeyer GB, Brundin P, Stijnen T, Roos RA. Weight loss in Huntington disease increases with higher CAG repeat number. Neurology. 2008;71:1506–1513. doi: 10.1212/01.wnl.0000334276.09729.0e. [DOI] [PubMed] [Google Scholar]
- 166.Brotherton A, Campos L, Rowell A, Zoia V, Simpson SA, Rae D. Nutritional management of individuals with Huntington’s disease: nutritional guidelines. Neurodegener Dis Manag. 2012;2:33–43. [Google Scholar]
- 167.Dubinsky JM. Towards an understanding of energy impairment in Huntington’s disease brain. J Huntingtons Dis. 2017;6:267–302. doi: 10.3233/JHD-170264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Chen CM. Mitochondrial dysfunction, metabolic deficits, and increased oxidative stress in Huntington’s disease. Chang Gung Med J. 2011;34:135–152. [PubMed] [Google Scholar]
- 169.Hamilton JM, Wolfson T, Peavy GM, Jacobson MW, Corey-Bloom J, et al. Rate and correlates of weight change in Huntington’s disease. J Neurol Neurosurg Psychiatry. 2004;75:209–212. doi: 10.1136/jnnp.2003.017822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Mortimer J, Zingales-Browne R, Drazinic C. Severe jaw dystonia causing weight loss in a young adult with Huntington’s disease relieved by tizanidine. Neurotherapeutics. 2014;11:217. [Google Scholar]
- 171.Pizzorni N, Pirola F, Ciammola A, Schindler A. Management of dysphagia in Huntington’s disease: a descriptive review. Neurol Sci. 2020;41:1405–1417. doi: 10.1007/s10072-020-04265-0. [DOI] [PubMed] [Google Scholar]
- 172.Hunt VP, Walker FO. Dysphagia in Huntington’s disease. J Neurosci Nurs. 1989;21:92–95. [PubMed] [Google Scholar]
- 173.Kagel MC, Leopold NA. Dysphagia in Huntington’s disease: a 16-year retrospective. Dysphagia. 1992;7:106–114. doi: 10.1007/BF02493441. [DOI] [PubMed] [Google Scholar]
- 174.Giddens CL, Coleman AE, Adams CM. A home program of speech therapy in Huntington’s disease. J Med Speech Lang Pathol. 2010;18:1–11. [Google Scholar]
- 175.Sampaio C. Huntington disease - update on ongoing therapeutic developments and a look toward the future. Parkinsonism Relat Disord. 2024;122:106049. doi: 10.1016/j.parkreldis.2024.106049. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.