Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Short and Long Term Outcome of Bilateral Pallidal Stimulation in Chorea-Acanthocytosis

  • Marie Miquel ,

    Contributed equally to this work with: Marie Miquel, Umberto Spampinato

    Affiliations Service de Neurologie, CHU Bordeaux, Bordeaux, France, Service de Neurologie, CH François Mitterrand, Pau, France

  • Umberto Spampinato ,

    Contributed equally to this work with: Marie Miquel, Umberto Spampinato

    umberto.spampinato@inserm.fr

    Affiliations Service de Neurologie, CHU Bordeaux, Bordeaux, France, Univ.Bordeaux-INSERM U862, Neurocentre Magendie, Bordeaux, France

  • Chrystelle Latxague,

    Affiliation Service de Neurologie, CHU Bordeaux, Bordeaux, France

  • Iciar Aviles-Olmos,

    Affiliation Unit of Functional Neurosurgery, Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London, United Kingdom

  • Benedikt Bader,

    Affiliation Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, Munich, Germany

  • Kelly Bertram,

    Affiliations Neurosciences, Alfred Hospital, Commercial Road, Melbourne, Victoria, Australia, Van Cleef Roet Centre for Nervous Diseases, Monash University, Melbourne, Victoria, Australia

  • Kailash Bhatia,

    Affiliation Sobell Department of Motor Neuroscience and Movement Disorders, University College London, Institute of Neurology, Queen Square, London, United Kingdom

  • Pierre Burbaud,

    Affiliation Service de Neurophysiologie Clinique, CHU Bordeaux, Bordeaux, France

  • Lothar Burghaus,

    Affiliation Department of Neurology, University Hospital Cologne, Cologne, Germany

  • Jin Whan Cho,

    Affiliation Department of Neurology, Samsung Medical Centre, Sungkyunkwan University, School of Medicine, Seoul, Korea

  • Emmanuel Cuny,

    Affiliation Service de Neurochirurgie, CHU Bordeaux, Bordeaux, France

  • Adrian Danek,

    Affiliation Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität München, Munich, Germany

  • Thomas Foltynie,

    Affiliation Unit of Functional Neurosurgery, Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London, United Kingdom

  • Pedro J. Garcia Ruiz,

    Affiliation Department of Neurology, Fundacion Jimenez Diaz, Madrid, Spain

  • Santiago Giménez-Roldán,

    Affiliation Servicio de Neurología, Hospital General Universitario Gregorio Marañón, Madrid, España

  • Dominique Guehl,

    Affiliation Service de Neurophysiologie Clinique, CHU Bordeaux, Bordeaux, France

  • Jorge Guridi,

    Affiliation Neurology and Neurosurgical Department, Clinica Universidad de Navarra, Pamplona, Spain

  • Marwan Hariz,

    Affiliation Unit of Functional Neurosurgery, Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London, United Kingdom

  • Paul Jarman,

    Affiliation National Hospital of Neurology and Neurosurgery, Queen Square, London, United Kingdom

  • Zinovia Maria Kefalopoulou,

    Affiliation Unit of Functional Neurosurgery, Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London, United Kingdom

  • Patricia Limousin,

    Affiliation Unit of Functional Neurosurgery, Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London, United Kingdom

  • Nir Lipsman,

    Affiliation Division of Neurosurgery, Toronto Western Hospital, University of Toronto, University Health Network, Toronto, Ontario, Canada

  • Andres M. Lozano,

    Affiliation Division of Neurosurgery, Toronto Western Hospital, University of Toronto, University Health Network, Toronto, Ontario, Canada

  • Elena Moro,

    Affiliations Morton and Gloria Shulman Movement Disorders Centre, Toronto Western Hospital, University of Toronto, University Health Network, Toronto, Ontario, Canada, Movement Disorders Center, Department of Psychiatry and Neurology, University Hospital Centre of Grenoble, Grenoble, France

  • Dhita Ngy,

    Affiliations Department of Neurology, Mount Sinai School of Medicine, New York, New York, United States of America, Avicenna Medical Center, New York, New York, United States of America

  • Maria Cruz Rodriguez-Oroz,

    Affiliation Department of Neurology, University Hospital Donostia, Neuroscience Unit, BioDonostia Research Institute, San Sebastian, Spain

  • Huifang Shang,

    Affiliation Department of Neurology, West China Hospital, Sichuan University, Chengdu, Sichuan, China

  • Hyeeun Shin,

    Affiliation Department of Neurology, Samsung Medical Centre, Sungkyunkwan University, School of Medicine, Seoul, Korea

  • Ruth H. Walker,

    Affiliations Department of Neurology, Mount Sinai School of Medicine, New York, New York, United States of America, Department of Neurology, James J. Peters Veterans Affairs Medical Center, Bronx, New York, United States of America

  • Fusako Yokochi,

    Affiliation Department of Neurology, Tokyo Metropolitan Neurological Hospital, Fuchu, Tokyo, Japan

  • Ludvic Zrinzo,

    Affiliation Unit of Functional Neurosurgery, Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London, United Kingdom

  •  [ ... ],
  • François Tison

    Affiliations Service de Neurologie, CHU Bordeaux, Bordeaux, France, Université Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France, CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France

  • [ view all ]
  • [ view less ]

Abstract

Background

Chorea-acanthocytosis (ChAc) is a neuroacanthocytosis syndrome presenting with severe movement disorders poorly responsive to drug therapy. Case reports suggest that bilateral deep brain stimulation (DBS) of the ventro-postero-lateral internal globus pallidus (GPi) may benefit these patients. To explore this issue, the present multicentre (n=12) retrospective study collected the short and long term outcome of 15 patients who underwent DBS.

Methods

Data were collected in a standardized way 2-6 months preoperatively, 1-5 months (early) and 6 months or more (late) after surgery at the last follow-up visit (mean follow-up: 29.5 months).

Results

Motor severity, assessed by the Unified Huntington’s Disease Rating Scale-Motor Score, UHDRS-MS), was significantly reduced at both early and late post-surgery time points (mean improvement 54.3% and 44.1%, respectively). Functional capacity (UHDRS-Functional Capacity Score) was also significantly improved at both post-surgery time points (mean 75.5% and 73.3%, respectively), whereas incapacity (UHDRS-Independence Score) improvement reached significance at early post-surgery only (mean 37.3%). Long term significant improvement of motor symptom severity (≥20 % from baseline) was observed in 61.5 % of the patients. Chorea and dystonia improved, whereas effects on dysarthria and swallowing were variable. Parkinsonism did not improve. Linear regression analysis showed that preoperative motor severity predicted motor improvement at both post-surgery time points. The most serious adverse event was device infection and cerebral abscess, and one patient died suddenly of unclear cause, 4 years after surgery.

Conclusion

This study shows that bilateral DBS of the GPi effectively reduces the severity of drug-resistant hyperkinetic movement disorders such as present in ChAc.

Introduction

Chorea-acanthocytosis (ChAc) is an autosomal recessive disease due to mutations of the VPS13A gene encoding for chorein [1]. It belongs to the neuroacanthocytosis syndromes, a heterogeneous group of genetically defined conditions, exhibiting neurological and neuropsychiatric disorders with red blood cell acanthocytosis [1,2]. ChAc is very rare, and typically presents as an adult-onset progressive disorder resembling Huntington's disease (HD) [1,2]. ChAc has however a slower progression, and displays some more or less specific clinical features such as tongue- and lip-biting, self-mutilations, seizures and neuromuscular manifestations but no specific neuropathological features besides prominent striatal degeneration [2,3].

There is no specific treatment for ChAc and the most frequent and disabling motor disorders, chorea and dystonia, are poorly controlled by conventional symptomatic drug therapy [2]. This has prompted neurosurgical treatment attempts, such as stereotactic brain lesions [4,5], and, more recently, deep brain stimulation (DBS) surgery of the internal globus pallidus (GPi) [6-8] following reported success in the management of other hyperkinetic movement disorders [9-12]. However, only single case or small series have been reported heterogeneously and in the short term, which likely bias the results towards favourable outcomes.

We performed a worldwide multicentre retrospective and cross-sectional study to gather all currently available data on all ChAc patients treated by GPi DBS and to provide a standardized, long-term comprehensive review of the clinical and functional outcome of these patients. In addition, to help guide GPi DBS surgery decision making in ChAc patients we aimed at identifying factors corresponding with favourable outcomes.

Clinical Materials and Methods

A total of 12 centres were contacted to participate in this retrospective study.

Case selection

Movement disorders and neurosurgical tertiary centres were identified and contacted on the basis of scientific publications (original research and review articles published between 1997 and November 2012) in Medline and PubMed databases (search terms: deep brain stimulation, DBS, high-frequency stimulation, pallidal stimulation, and neuroacanthocytosis, chorea, chorea-acanthocytosis, ChAc), as well as of meeting abstracts. The support association for neuroacanthocytosis patients (www.naadvocacy.org), and the diagnostic reference centre and database in Munich, Germany, also collaborated for patient identification, and announcements were published at the Movement Disorders Society (MDS) meeting 2011 and MDS website.

Patients

Selected patients were diagnosed with molecularly proven or clinically probable ChAc (Table 1), and treated with GPi DBS regardless of surgery outcome. In the case of previously published cases, authors were asked to provide additional updated long term information at last patient visit using a standardized data collection sheet. Recruitment was conducted from February 2011 to November 2012.

PatientAcanthocytes (%)CK*Chorein in Western blot of red cells membranesVPS13A mutationsMRI (atrophy)
120x4absentNDCa, Pu, Cx
240x4absentNDCa
310NDpresent (2 blots in 4 years)present, o.m.Ca
4x2.6absentpresent, c.h.nl
525x1.1absentpresent, HCa, Pu, Cx; striatal T2 hyper-intensity
631x2.1absentpresent, HCa, Cx
7NDNDNDpresent, c.h.nl
820x1.6NDpresent, c.h.Ca heads, Pu with T2 hyper-intensity
910x18.8NDpresent, Hnl
10NDsubnormal low expressionNDnl
1120x4.2NDpresent, c.h.Ca heads, subcortical.
12NDabsentNDCa heads, Pu T2 hyper-intensity
137x13.2absentNDCa, lentiform nuclei with T2 hyper-intensity
146x2.2NDNDCa, Pu, T2 hyper-intensity
156NDNDNDCa, Pu, T2 hyper-intensity

Table 1. Basic diagnostic data in 15 patients with chorea.

CK = creatine kinase; ↑ elevated; * relative increase with respect to normal value for each centre laboratory; o.m. = one mutation known; c.h. = compound heterozygous; H = homozygous; Ca = Caudate; Cx = Cortical; Pu = Putamen; nl = normal; ND = not done.
CSV
Download CSV

Data acquisition, protocol and surgery outcome measures

All centres were sent a standardized data collection sheet, prepared according to the guidelines for reporting results from clinical DBS studies in Parkinson’s disease [13].

Data were collected 2-6 months preoperatively (PREOP), at 1-5 months postoperatively (early post-operative: EPOP), and 6 months or more after surgery (last outcome reporting: LOR) (see Table S1).

Change in the severity of the motor disorder was measured by the Unified Huntington’s Disease Rating Scale-Motor Score (UHDRS-MS), and in functional status by the UHDRS-Independence Score (UHDRS-IS) and the UHDRS-Functional Capacity Score (UHDRS-FCS) [14]. In accordance with previous studies [9,15] an improvement equal or superior to 20% of the baseline score was considered as clinically relevant.

Data analysis

Data obtained are presented as the mean ± SEM values for each scale administered and at each time point: PREOP, EPOP and LOR. Statistical analysis was carried out using Statistica 8.0 for Windows (Statsoft, Maison-Alfort, France). Data were first analyzed by the Kolmogorov-Smirnov Test for normal distribution (p > 0.05). Thereafter, the effect of DBS upon each variable (UHDRS-MS, UHDRS-IS, UHDRS-FCS) was assessed by a multifactor analysis of variance (ANOVA) with time as the within-subjects factor. When ANOVA results were significant (p<0.05), the post hoc Bonferroni test was applied to allow adequate multiple comparisons between groups.

For each variable, ANOVA was performed using only those cases (11 patients) for which values were available at all three time points (Table 2). Individual values available for each patient were also entered and plotted at each time point. Data available at EPOP and/or LOR time points were also expressed as the percentage of PREOP values (100%).

PREOPEPOPLOR
UHDRS-MS131311
UHDRS-FCS141411
UHDRS-IS141411
BFMDRS111
MMSE973
Adverse eventsNA1515
Clinical status/outcome by symptoms151414
Drug treatment 14-14

Table 2. Number of patients with exploitable data.

PREOP: preoperative; EPOP: early post-operative; LOR: last outcome report; UHDRS-MS: Unified Huntington’s Disease Rating Scale-Motor Score; UHDRS-FCS: UHDRS-Functional-Capacity Score; UHDRS-IS: UHDRS-Independence Score; BFMDRS-M: Burke-Fahn-Marsden Dystonia Rating Scale-Motor part [16]; MMSE: Mini Mental State Examination [17]; NA: not applicable.
CSV
Download CSV

A linear regression analysis was performed to identify preoperative factors (age at surgery, disease duration and severity) that could predict DBS-induced improvement of motor and/or functional impairment at EPOP and/or LOR assessments. Analysis was performed using cases for which postoperative UHDRS values were available at either EPOP and/or LOR evaluations (see Table 2). “Improvement” was considered for each patient as the difference between UHDRS scores at PREOP and EPOP or LOR time points. P-values < 0.05 were considered significant; no adjustment was made for multiple testing.

Ethics

The study was conducted after approval from the local Ethics Committee of the University Hospital of Bordeaux, and was carried out in accordance with the ethical standards laid down in the 1964 Declaration of Helsinki. All patients or a family member gave their written informed consent prior to their inclusion in the study, and data were entered into a database in an anonymous format in accordance with the personal data protection laws in France (CNIL).

Results

Eleven out of the 12 centres (see Appendix S1) provided data. Fifteen patients were included in the data analysis but data for one patient (patient 11) were extracted only from a previous publication, and UHDRS scores were missing. Table 2 indicates the number of patients available for analysis at each time point. Twelve of the 15 cases have already been published in either scientific article format (n=9) or in congress posters (n=3) (for individual references see Table 3).

Patient numberGenderAge at onset of symptoms (years)Age at time of diagnosis (years)Age at surgery (years)Duration of disease (years)LOR after surgery (months)
1 [18]F252633860
2F313438736
3F353741636
4 [19,20]M484949141
5 [21]M2946481924
6 [21]M303232236
7 [22]M222431939
8 [23]F353839413
9 [24,25]M242932884
10F25293276
11 [26]M35 38383NA*
12 [8]M304554245
13 [8]M323943113
14 [7]M174040239
15 [7]M1830301221
Mean ± SD5 F (33%) 10 M (67%)29.1 ± 7.835.7 ± 7.538.7 ± 7.39.6 ± 7.229.5 ± 23

Table 3. Patient characteristics.

F = female, M = male; LOR = last outcome report; NA = not available; * electrodes were removed 3 weeks after surgery.
CSV
Download CSV

Patient clinical features

Patient characteristics are summarized in Table 3. Most of the patients presented with chorea (n=14, 93%) and/or dystonia (n=12, 80%), frequently associated with trunk spasms (n=12, 80%) and/or head drops/chorea (n=10, 67%), leading to recurrent face and head trauma. Most patients had gait and balance impairment. All patients had dysarthria, severe in 6. Orofacial dyskinesias and tics were present in 12 patients, 10 displayed lip-, tongue- or cheek-biting. Swallowing problems were present in 12 patients. Five patients had epilepsy, effectively treated in all. Psychiatric and behavioural disturbances, mainly depression and obsessive-compulsive behaviour, were present in 11 patients. Cognition was reported as slightly impaired in 6 patients, but extensive neuropsychological evaluations were lacking.

Surgery was indicated to reduce disabling motor symptoms such as chorea, dystonia, trunk spasms, falls, gait impairment (7 patients), self-mutilating and biting behaviours (6 patients) or head banging induced by trunk spasms or head drops (4 patients), feeding dystonia (3 patients) and recurrent belching (2 patients).

Outcome measures of global efficacy

Figure 1 displays the effect of DBS on UHDRS motor scores at EPOP and LOR time points compared to baseline. Mean UHDRS-MS (Figure 1A) was significantly reduced at both time points after surgery. Mean proportional improvement of UHDRS-MS compared to baseline was +54.3% and +44.1% for EPOP and LOR, respectively. Figures 1B and 1C show individual outcomes on UHDRS-MS scores and changes compared to PREOP values. All 13 patients showed an improvement of 20% or more at EPOP, whereas at LOR, only 8 of these 13 patients (61.5 %), for whom UHDRS-MS was available, showed a significant improvement.

thumbnail
Figure 1. Effect of deep brain stimulation on motor impairment as assessed by the Unified Huntington’s Disease Rating Scale-Motor Score (UHDRS-MS).

Bar histograms (panel A) represent means ± SEM (n= 11 patients). Line plots (panels B, C) show individual values per patient plotted at each of three time points: 2-6 months preoperatively (PREOP), 1-5 months postoperatively (early post-operative: EPOP), and 6 months or more after surgery (last outcome reporting: LOR). Individual data curves are shown in panel B, whereas panel C depicts percentage changes at EPOP and LOR with PREOP values set to 100% to make improvements and deteriorations easier to distinguish. Scores of patient 7 correspond to the Burke-Fahn-Marsden Dystonia Rating Scale-Motor part (BFMDRS-M) [16]. **p<0.01, ***p<0.001 versus PREOP time point (Bonferroni test after ANOVA: F(2, 20)= 15.11, p< 0.001, n= 11).

https://doi.org/10.1371/journal.pone.0079241.g001

DBS effects on the patients’ functional status, as evaluated by UHDRS-IS and UHDRS-FCS, are displayed in Figure 2. Mean UHDRS-IS was improved at both time points after surgery (EPOP: +37.3%, LOR: +22%), but reached significance only at EPOP (Figure 2A). Ten out of 14 patients (71.4 %) for whom UHDRS-IS was available at EPOP showed a significant improvement, which was still observed at LOR for 6 of them (Figure 2B, 2C). Mean UHDRS-FCS was significantly improved at both time points post-surgery (EPOP: +75.5%; LOR: +73.3%; Figure 2D). Eleven out of 14 patients (78.6 %) for whom UHDRS-IS was available at EPOP showed a clinically relevant improvement; which was still observed at LOR for 6 of them (Figure 2E, 2F).

thumbnail
Figure 2. Effect of deep brain stimulation on independence and functional impairment as assessed by the Unified Huntington’s Disease Rating Scale-Independence Score (UHDRS-IS) and UHDRS-Functional Capacity Score (UHDRS-FCS), respectively.

Bar histograms (panels A, D) represent means ± SEM (n= 11 patients). Line plots (panels B, C, E, F) show individual values per patient plotted at each of three time points: 2-6 months preoperatively (PREOP), 1-5 months postoperatively (early post-operative: EPOP), and 6 months or more after surgery (last outcome reporting: LOR). Individual data curves are shown in panels B and E, whereas panels C and F depict percentage changes at EPOP and LOR with PREOP values set to 100% to make improvements and deteriorations easier to distinguish. *p<0.05 versus the corresponding PREOP time point (Bonferroni test after ANOVA: F(2, 20)= 4.72, p< 0.05, n= 11, and F(2, 20)= 5.94, p< 0.01, n= 11, for UHDRS-IS and UHDRS-FCS, respectively).

https://doi.org/10.1371/journal.pone.0079241.g002

Outcome by symptoms

Chorea and dystonia were the most improved symptoms in a majority of patients. In terms of chorea, 13 out of 14 patients were improved at EPOP, observed within hours or days after surgery. At LOR, some relapse of choreic movements was observed in 3 patients (1, 4 and 15 at 24, 11 and 21 months, respectively), while the effect was maintained in others. Trunk spasms improvement was marked and maximal at EPOP for 11 out of 12 patients. Patients 3 and 9 experienced slower improvement of limb dystonia, with the maximum effect observed at LOR.

There was improvement of gait in patients with chorea and dystonia, but this was not significant in patients with pre-operative parkinsonism (4 out of 6). Except for patient 15, all patients who suffered from orofacial movements and self-mutilation improved. DBS had little effect on dysarthria, only 2 improved at EPOP, one of whom was found worse at LOR; one patient (patient 9) presented complete anarthria after surgery which improved after withdrawal of stimulation. Feeding status was available at LOR only in 9 patients and 6 had improved. Insufficient information was available to allow conclusions upon cognitive or neuropsychiatric and behavioural disorders. Mini Mental State Examination [17] was available in 9 patients before surgery (mean score = 26.8; range = 19-30), in 7 patients at EPOP (mean score = 27.3; range = 25-30), and in 3 patients at LOR (mean score = 26.7; range = 26-27). After surgery, antidepressant treatment remained unchanged in 6 patients. Mood improved for 3 patients, but no change was reported for the others. Four patients (patients 2, 7, 10 and 11) had preoperative compulsive behaviours: after surgery a marked and sustained (48 months) improvement was observed in one patient (patient 7).

Relationship between preoperative status and surgery outcome

No significant relationship was found between age at surgery or disease duration and postoperative improvement on either motor or functional scales. Preoperative motor severity in UHDRS-MS predicted improvement in motor function at both EPOP (Figure 3A) and LOR time points (Figure 3B). Motor improvement was associated with an improvement in functional status (UHDRS-FCS) at EPOP and at LOR (Figure 3C, D); but not in the dependency score at both time points (UHDRS-IS).

thumbnail
Figure 3. Regression analysis between preoperative (PREOP) severity of motor impairment, as assessed by the Unified Huntington’s Disease Rating Scale-Motor Score (UHDRS-MS), and motor severity improvement at early and late post-operative time points (EPOP, panel A, r = 0.66, r2 = 0.44, p< 0.05, n=13; LOR, panel B, r = 0.67, r2 = 0.45, p< 0.05, n=11); and motor severity improvement versus functional improvement, as assessed by the UHDRS-Functional Capacity Score (UHDRS-FCS), at EPOP (panel C, r = 0.63, r2 = 0.40, p< 0.05, n=13) and LOR (panel D, r = 0.67, r2 = 0.45, p< 0.05, n=11) time points.

For each patient, motor and functional improvements correspond to the difference between appropriate UHDRS scores at PREOP and EPOP or LOR time points.

https://doi.org/10.1371/journal.pone.0079241.g003

Surgery: implantation, target coordinates and stimulation parameters

Surgical procedure details and individual electrode coordinates as per the Schaltenbrand and Wahren atlas [27] are reported in Tables S2 and S3, respectively. Mean and range of stimulation parameters at per-operative, EPOP and LOR assessments are displayed in Table 4; individual optimal stimulation settings are given in Supporting Information, Table S4.

Parameterintra-OP (n=14)EPOP (n=14)LOR (n=12)
Pulse frequency (Hz)102.5 (40-185)114.3 (40-185)128.8 (40-185)
Pulse width (μs)90.0 (60-210)96.3 (60-180)104.0 (60-150)
Pulse amplitude (V)2.8 (1.6-4.5)2.9 (2-4.5)3.4 (2-4.8)

Table 4. Mean stimulation parameters.

Data= mean (range); intra-OP= intra-operative; EPOP= early post-operative; LOR= last outcome report.
CSV
Download CSV

The pulse frequency applied at EPOP was high in 10 patients (130-185 Hz), and low in 4 (40-60 Hz). Stimulus duration, frequency and amplitude tended to increase at LOR. No patient initially treated with high frequency was switched to low frequency for further improvement; only one patient initially treated with low frequency was switched to high frequency stimulation at LOR (patient 9). At EPOP, stimulation was single monopolar for 10 patients and double monopolar for 4 patients. Most patients were treated via ventral contacts. At LOR, the stimulation field tended to be larger and more dorsal, 4 patients were still stimulated by single monopolar contact, and 6 by double or triple monopolar contacts.

Movement disorders medication

Before surgery, all patients took at least one drug for their movement disorder (neuroleptics, tetrabenazine, benzodiazepines, amantadine, anticholinergic drugs (mean number taken: 3 ± 1, mean number tried: 4 ± 2). At LOR, medication was reduced in 7 patients (50%) and had remained unchanged in 3 patients (21%), but drug number had increased in 4 patients (28.6%). In only one patient (patient 9) medication was discontinued for 2 years after surgery.

DBS adverse events

Data on adverse events were available for all 15 patients.

Surgery-related adverse events.

Patient 9 had a misplaced right electrode requiring a new surgery. Two months after surgery, patient 13 had a device infection leading to implanted pulse generator (IPG) removal, thereby causing severe rebound of abnormal movements requiring sedation in the intensive care unit. Ten days later, while the infection was apparently resolved, a new IPG was implanted but subsequently the patient developed a cerebral abscess at the sites of the left electrode tracks, which prompted the complete removal of the whole system. One year later, a new right GPi DBS has had a positive effect on left-sided symptoms.

Stimulation-related adverse events.

Patient 1 displayed a worsening of trunk spasms at 40-50Hz stimulation frequency and with the use of the ventral contact he experienced gait freezing that disappeared with stimulation at more dorsal contacts but at the cost of worsening hyperkinesias and trunk spasms. Patient 4, at EPOP period, complained of abnormal face and mouth pulling over 60 Hz stimulation. One month after surgery he displayed increased tongue movements at 50Hz stimulation using bilateral double monopolar settings (C+1-2-), thereby requiring an adjustment towards bilateral single monopolar (C+1-) stimulation. Patient 9, at EPOP time point, exhibited worsening of chorea and dysarthria, with palilalia and drooling at 130Hz, 40Hz stimulation was effective on choreic movements. At LOR assessment, again under 130Hz stimulation, abnormal movements were well-controlled but clinical examination revealed mild parkinsonism and complete anarthria. Rigidity and bradykinesia slightly decreased 2 hours after stopping the stimulation, and the patient was able to pronounce few simple words. Patient 11 experienced increased chorea and side effects (facial hemispasm, visual disturbances and vegetative changes) whatever the bipolar stimulation parameters used at high frequency. Low frequency stimulation led to deterioration of speech and gait [26]. Patient 14 had an exacerbation of both chorea and dystonia at high-frequency (130 Hz) stimulation. Patient 15 complained of blurred vision during the per-operative macro-stimulation on the lowest plots and using high amplitude.

Device-related adverse events.

Two years after surgery, patient 2 experienced a “Twiddler syndrome” with lead rupture that required the replacement of the extension wires, IPG and both electrodes [28]. Device infection in patient 13 was discussed above.

Patient-related adverse events.

At LOR, patient 4 requested to have the stimulator switched “off” because of suspected stimulation-related increased involuntary tongue protrusion. This, however, resulted in a further increase of tongue thrusting and DBS subsequently was again turned “on”, with settings unchanged. This patient suddenly died at home at age 53, 4 years after surgery and 1 year after LOR. An autopsy was not performed and the cause of death remained undetermined.

Discussion

The present long-term multicentre study of 15 cases of ChAc treated with bilateral DBS of the GPi provides evidence that stimulation of the postero-ventrolateral "sensorimotor" portion of the GPi may help to manage severe, drug-resistant movement disorders, particularly chorea and dystonia. In the 11 patients, with two postoperative standardized evaluations, a significant long term improvement of motor symptom severity (improvement ≥ 20% in UHDRS-MS) was observed for 61.5 % at a mean follow-up of 2.5 years. Because duration of follow-up was variable, we constructed individual outcome curves that demonstrated that most patients remained above the pre-operative level, even though there was a tendency for motor deterioration over time. Gait impairment, orofacial movements and tics, self-mutilation, head drops and trunk spasms were DBS-sensitive symptoms, while dysarthria and feeding problems were less DBS-responsive and had a more variable outcome. Limb, face and trunk chorea improved early after surgery and responded immediately to switching the stimulation “on” and relapsed abruptly when switching “off”. This response resembles that observed in HD [10,29,30] and in McLeod Syndrome [5,25].

An early improvement was observed for phasic/mobile dystonia (debilitating trunk and limb spasms, head drops with injuries), as described in other conditions including primary generalized dystonia (PGD) [11,31,32]. Conversely, improvement of tonic dystonia was slower and occurred after 6 months of stimulation for patients 3 and 9, also resembling observations made in PGD [31,33].

As in other primary choreic syndromes [10,29] we observed a tendency for symptom deterioration and/or occurrence of parkinsonism in the longer term. This may be related to disease progression and/or to DBS itself, possibly by activation of inhibitory pallidal outputs to the motor thalamus [34]. Although GPi-DBS is an effective treatment for motor symptoms in Parkinson’s disease (PD), worsening of akinesia and gate deterioration was also observed in GPi-DBS-treated PD patients depending on the stimulated area within the GPi [34].

Dysarthria occurrence or worsening could also be related to disease progression or to DBS adverse effect, as observed in one of our patients (patient 9). GPi-DBS efficacy on swallowing was highly variable. Improvement could be seen when swallowing impairment was mainly due to feeding dystonia, orolingual movement disorders or trunk spasms. Otherwise, swallowing problems tended to deteriorate along with axial signs and parkinsonism.

At LOR assessment, drugs used to control the movement disorders were reduced in 71% of patients (10/14), thus supporting the assumption that the effect upon movement disorders can reasonably be attributed to DBS rather than to drug regime changes. Only one patient did not benefit from strictly monopolar DBS. His disease was very advanced (17 years disease duration) as well as drug-resistant (11 drugs had been tried).

Motor improvement translated into an improvement of functional capacity both at early and late post-operative observations. The patients showed significant functional improvement of 75.5% (UHDRS-FCS) at EPOP and LOR. Independence, as assessed by the UHDRS-IS, was significantly improved only at EPOP, even if there was also a trend for improvement at LOR. Thus, improvement in motor signs and functional capacity did not fully translate to improvement of dependency at the long-term. This could be due to the sensitivity and/or the psychometric characteristics of the subscale used and/or the low number of patients under study. Alternatively, DBS, while improving motor symptoms and function, may be ineffective with respect to dependency in patients severely affected by advanced disease. This issue remains to be addressed in prospective studies using also quality of life measures. The cognitive outcome of ChAc patients with GPi DBS was inconclusive with the limited data in hand.

There was no correlation between the patients´ demographics or disease duration and outcome. However, there was a significant relation between pre-operative severity of motor signs (UHDRS-MS) and improvement of motor signs.

The UHDRS scale was used as previously proposed for neurodegeneration with brain iron accumulation (NBIA) in an attempt to standardize the data available [15] but, as in NBIA, the UHDRS has not been validated in ChAc. Although the disease spectrum is very similar to HD, the scale may have missed characteristic ChAc symptoms such as orofacial movement disorders and self-mutilations, feeding dystonia, trunk spasms, head drops, breathing dystonia and recurrent belching. Formal validation of the UHDRS in ChAc or design of a ChAc specific scale is needed but difficult to perform due to the very small number of patients involved.

Implanted devices and stimulation parameter settings varied among centres. However, optimal stimulation parameter settings mostly used higher frequency (mean = 102.5 Hz) and monopolar stimulation of lower contacts with a mean amplitude of 2.8 volts. There was a tendency to gradually increase frequency and amplitude along with the use of more dorsal contacts at the latest follow-up [35,36]. This may be due to disease progression or, alternatively, to an attempt to control increased bradykinesia over time [37].

The risk/benefit ratio appears acceptable in the present case series. Although the limited number of patients prevents definitive conclusions, adverse events were roughly similar to those observed in HD, PD, NBIA or dystonia case series [10,15,30,31,38,39]. Misplacement of one electrode requiring new surgery, Twiddler syndrome and device infection with brain abscess were the most serious surgery-related adverse events. In the 5 patients with preoperative epilepsy, there was no worsening of epilepsy or increase in antiepileptic drug dosage, and no de novo seizures after surgery in patients without epilepsy. These findings, although observed in few patients, suggest that epilepsy may not be a contraindication or a serious concern for GPi-DBS in ChAc, but further studies are needed to address this point. Indeed, a recent case reported possible intraoperative seizures and also post-operative intracranial hematoma in a 32-years-old patient with non-molecularly proven ChAc [40]. Two years later, the patient underwent a second DBS operation leading to a marked improvement of his dystonia, chorea and overall quality of life 2 and 8 months postoperatively [40].

Finally, although our results are positive and encouraging, it is important to note that our study presents some limitations due to its retrospective and multi-centre nature leading to some lack of homogeneity in patients’ assessment and data collection, despite of our efforts of standardization. Also, some data were missing (see Table 2), and in two patients (Table 1, patients 14 and 15) the diagnosis of ChAc was only “highly probable” in the absence of screening for Chorein blot or VPS13A mutations [7].

In conclusion, our study of 15 patients collected worldwide, is in favour of long-term efficacy and relative safety of the bilateral GPi-DBS for severe, drug-resistant hyperkinetic movement disorders (chorea, dystonia and/or orofacial movements and self-mutilations) in ChAc. DBS provides long-term motor and functional improvement but has less effect on long-term dependency. Also, as reported previously in NBIA patients [15] preoperative severity is predictive of a good response to bilateral GPi-DBS. Our study emphasizes the need to improve prospective data collection in international frameworks; gathering clinical characteristics of ChAc patients and their long term monitoring is essential to further evaluate interventions such as DBS in this debilitating disease.

Supporting Information

Table S1.

Time-schedule of the data collected during the study.

https://doi.org/10.1371/journal.pone.0079241.s002

(DOCX)

Acknowledgments

The authors would like to thank Ginger and Glenn Irvine and the Advocacy for Neuroacanthocytosis Patients (www.naadvocacy.org) for their help in identifying the patients, and Miss Adeline Cathala for statistical assistance. Western blot analysis for chorein was performed by Carol Dobson-Stone in the lab of Anthony Monaco at the Wellcome Trust Centre for Human Genetics, University of Oxford, UK, and by Benedikt Bader and Gertrud Kwiatkowski in the lab of Hans Kretzschmar at the Center for Neuropathology and Prion Research at Ludwig-Maximilians-Universität Munich, Germany.

Author Contributions

Conceived and designed the experiments: FT CL US. Performed the experiments: MM US CL IA-O BB K. Bertram K. Bhatia PB LB JWC EC AD TF PJGR SG-R DG JG MH PJ Z-MK PL NL AML EM DN MCR-O H. Shang H. Shin RHW FY LZ FT. Analyzed the data: MM US FT. Contributed reagents/materials/analysis tools: MM US CL IA-O BB K. Bertram K. Bhatia PB LB JWC EC AD TF PJGR SG-R DG JG MH PJ Z-MK PL NL AML EM DN MCR-O H. Shang H. Shin RHW FY LZ FT. Wrote the manuscript: MM US FT. Made critical revision for important intellectual content and gave final approval of the version to be published: MM US CL IA-O BB K. Bertram K. Bhatia PB LB JWC EC AD TF PJGR SG-R DG JG MH PJ Z-MK PL NL AML EM DN MCR-O H. Shang H. Shin RHW FY LZ FT.

References

  1. 1. Danek A, Jung HH, Melone MA, Rampoldi L, Broccoli V et al. (2005) Neuroacanthocytosis: new developments in a neglected group of dementing disorders. J Neurol Sci 229-230: 171-186. doi:https://doi.org/10.1016/j.jns.2004.11.024. PubMed: 15760637.
  2. 2. Jung HH, Danek A, Walker RH (2011) Neuroacanthocytosis syndromes. Orphanet J Rare Dis 6: 68. doi:https://doi.org/10.1186/1750-1172-6-68. PubMed: 22027213.
  3. 3. Walterfang M, Looi JC, Styner M, Walker RH, Danek A et al. (2011) Shape alterations in the striatum in chorea-acanthocytosis. Psychiatry Res 192: 29-36. doi:https://doi.org/10.1016/j.pscychresns.2010.10.006. PubMed: 21377843.
  4. 4. Fujimoto Y, Isozaki E, Yokochi F, Yamakawa K, Takahashi H et al. (1997) A case of chorea-acanthocytosis successfully treated with posteroventral pallidotomy. Rinsho Shinkeigaku 37: 891-894. PubMed: 9490899.
  5. 5. Yokochi F, Burbaud P (2008) Neurosurgery for Neuroacanthocytosis. In: RH WalkerS. SaïkiA. Danek. Neuroacanthocytosis Syndromes, vol II. Springer, Berlin. pp 255-269.
  6. 6. Edwards TC, Zrinzo L, Limousin P, Foltynie T (2012) Deep brain stimulation in the treatment of chorea. Mov Disord 27: 357-363. doi:https://doi.org/10.1002/mds.23967. PubMed: 21997283.
  7. 7. Li P, Huang R, Song W, Ji J, Burgunder JM et al. (2012) Deep brain stimulation of the globus pallidus internal improves symptoms of chorea-acanthocytosis. Neurol Sci 33: 269-274. doi:https://doi.org/10.1007/s10072-011-0741-y. PubMed: 21863267.
  8. 8. Kefalopoulou Z, Zrinzo L, Aviles-Olmos I, Bhatia K, Jarman P et al. (2013) Deep brain stimulation as a treatment for chorea-acanthocytosis. J Neurol 260: 303-305. doi:https://doi.org/10.1007/s00415-012-6714-0. PubMed: 23086180.
  9. 9. Vidailhet M, Yelnik J, Lagrange C, Fraix V, Grabli D et al. (2009) Bilateral pallidal deep brain stimulation for the treatment of patients with dystonia-choreoathetosis cerebral palsy: a prospective pilot study. Lancet Neurol 8: 709-717. doi:https://doi.org/10.1016/S1474-4422(09)70151-6. PubMed: 19576854.
  10. 10. Kang GA, Heath S, Rothlind J, Starr PA (2011) Long-term follow-up of pallidal deep brain stimulation in two cases of Huntington's disease. J Neurol Neurosurg Psychiatry 82: 272-277. doi:https://doi.org/10.1136/jnnp.2009.202903. PubMed: 20974647.
  11. 11. Lyons MK (2011) Deep brain stimulation: current and future clinical applications. Mayo Clin Proc 86: 662-672. doi:https://doi.org/10.4065/mcp.2011.0045. PubMed: 21646303.
  12. 12. Lim BC, Ki CS, Cho A, Hwang H, Kim KJ et al. (2012) Pantothenate kinase-associated neurodegeneration in Korea: recurrent R440P mutation in PANK2 and outcome of deep brain stimulation. Eur J Neurol 19: 556-561. doi:https://doi.org/10.1111/j.1468-1331.2011.03589.x. PubMed: 22103354.
  13. 13. Vitek JL, Lyons KE, Bakay R, Benabid AL, Deuschl G et al. (2010) Standard guidelines for publication of deep brain stimulation studies in Parkinson's disease (Guide4DBS-PD). Mov Disord 25: 1530-1537. doi:https://doi.org/10.1002/mds.23151. PubMed: 20544809.
  14. 14. Study Huntington Group (1996) Unified Huntington's Disease Rating Scale: reliability and consistency. Mov Disord 11: 136-142 doi:https://doi.org/10.1002/mds.870110204. PubMed: 8684382.
  15. 15. Timmermann L, Pauls KA, Wieland K, Jech R, Kurlemann G et al. (2010) Dystonia in neurodegeneration with brain iron accumulation: outcome of bilateral pallidal stimulation. Brain 133: 701-712. doi:https://doi.org/10.1093/brain/awq022. PubMed: 20207700.
  16. 16. Burke RE, Fahn S, Marsden CD, Bressman SB, Moskowitz C et al. (1985) Validity and reliability of a rating scale for the primary torsion dystonias. Neurology 35: 73-77. doi:https://doi.org/10.1212/WNL.35.1.73. PubMed: 3966004.
  17. 17. Folstein MF, Folstein SE, McHugh PR (1975) "Mini-mental state". A practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res 12: 189-198. doi:https://doi.org/10.1016/0022-3956(75)90026-6. PubMed: 1202204.
  18. 18. Ruiz PJ, Ayerbe J, Bader B, Danek A, Sainz MJ et al. (2009) Deep brain stimulation in chorea acanthocytosis. Mov Disord 24: 1546-1547. doi:https://doi.org/10.1002/mds.22592. PubMed: 19425062.
  19. 19. Gan JJ GF, Alterman RL, Cheung T, Gora-Stahlberg G, Tagliati M (2011) Long-term benefit of pallidal stimulation in a case of chorea-acanthocytosis [abstract] Neurology 76 (Suppl 4): A590 doi:https://doi.org/10.1212/WNL.0b013e31820d8b61.
  20. 20. Walker RH, Schulz VP, Tikhonova IR, Mahajan MC, Mane S et al. (2012) Genetic diagnosis of neuroacanthocytosis disorders using exome sequencing. Mov Disord 27: 539-543. doi:https://doi.org/10.1002/mds.24020. PubMed: 22038564.
  21. 21. Yokochi F, Kimura K, Okiyama R, Taniguchi M, Yokochi M (2011) Surgical treatment for neuroacanthocytosis [abstract] Mov Disord 26 (Suppl 2): S96-S97.
  22. 22. Schneider SA, Lang AE, Moro E, Bader B, Danek A et al. (2010) Characteristic head drops and axial extension in advanced chorea-acanthocytosis. Mov Disord 25: 1487-1491. doi:https://doi.org/10.1002/mds.23052. PubMed: 20544815.
  23. 23. Shin H, Ki CS, Cho AR, Lee JI, Ahn JY et al. (2012) Globus pallidus interna deep brain stimulation improves chorea and functional status in a patient with chorea-acanthocytosis. Stereotact Funct Neurosurg 90: 273-277. doi:https://doi.org/10.1159/000338216. PubMed: 22777538.
  24. 24. Sibon I, Ghorayeb I, Arné P, Tison F (2004) Distressing belching and neuroacanthocytosis. Mov Disord 19: 856-859. doi:https://doi.org/10.1002/mds.20091. PubMed: 15254955.
  25. 25. Guehl D, Cuny E, Tison F, Benazzouz A, Bardinet E et al. (2007) Deep brain pallidal stimulation for movement disorders in neuroacanthocytosis. Neurology 68: 160-161. doi:https://doi.org/10.1212/01.wnl.0000250536.81426.1a. PubMed: 17210902.
  26. 26. Wihl G, Volkmann J, Allert N, Lehrke R, Sturm V et al. (2001) Deep brain stimulation of the internal pallidum did not improve chorea in a patient with neuro-acanthocytosis. Mov Disord 16: 572-575. doi:https://doi.org/10.1002/mds.1109. PubMed: 11391763.
  27. 27. Schaltenbrand G, Wahren W (1977) Atlas for Stereotaxy of the Human Brain. Thieme, Stuttgart.
  28. 28. Guridi J, Rodriguez-Oroz MC, Alegre M, Obeso JA (2012) Hardware complications in deep brain stimulation: electrode impedance and loss of clinical benefit. Parkinsonism Relat Disord 18: 765-769. doi:https://doi.org/10.1016/j.parkreldis.2012.03.014. PubMed: 22522071.
  29. 29. Moro E, Lang AE, Strafella AP, Poon YY, Arango PM et al. (2004) Bilateral globus pallidus stimulation for Huntington's disease. Ann Neurol 56: 290-294. doi:https://doi.org/10.1002/ana.20183. PubMed: 15293283.
  30. 30. Biolsi B, Cif L, Fertit HE, Robles SG, Coubes P (2008) Long-term follow-up of Huntington disease treated by bilateral deep brain stimulation of the internal globus pallidus. J Neurosurg 109: 130-132. doi:https://doi.org/10.3171/JNS/2008/109/7/0130. PubMed: 18590443.
  31. 31. Vidailhet M, Vercueil L, Houeto JL, Krystkowiak P, Lagrange C et al. (2007) Bilateral, pallidal, deep-brain stimulation in primary generalised dystonia: a prospective 3 year follow-up study. Lancet Neurol 6: 223-229. doi:https://doi.org/10.1016/S1474-4422(07)70035-2. PubMed: 17303528.
  32. 32. Volkmann J, Wolters A, Kupsch A, Müller J, Kühn AA et al. (2012) Pallidal deep brain stimulation in patients with primary generalised or segmental dystonia: 5-year follow-up of a randomised trial. Lancet Neurol 11: 1029-1038. doi:https://doi.org/10.1016/S1474-4422(12)70257-0. PubMed: 23123071.
  33. 33. Isaias IU, Alterman RL, Tagliati M (2009) Deep brain stimulation for primary generalized dystonia: long-term outcomes. Arch Neurol 66: 465-470. doi:https://doi.org/10.1001/archneurol.2009.20. PubMed: 19364931.
  34. 34. Yelnik J, Damier P, Bejjani BP, Francois C, Gervais D et al. (2000) Functional mapping of the human globus pallidus: contrasting effect of stimulation in the internal and external pallidum in Parkinson's disease. Neuroscience 101: 77-87. doi:https://doi.org/10.1016/S0306-4522(00)00364-X. PubMed: 11068138.
  35. 35. Moro E, Esselink RJ, Xie J, Hommel M, Benabid AL et al. (2002) The impact on Parkinson's disease of electrical parameter settings in STN stimulation. Neurology 59: 706-713. doi:https://doi.org/10.1212/WNL.59.5.706. PubMed: 12221161.
  36. 36. Koss AM, Alterman RL, Tagliati M, Shils JL (2005) Calculating total electrical energy delivered by deep brain stimulation systems. Ann Neurol 58: 168-169. doi:https://doi.org/10.1002/ana.20526. PubMed: 15984018.
  37. 37. Bejjani BP, Damier P, Arnulf I, Papadopoulos S, Bonnet AM et al. (1998) Deep brain stimulation in Parkinson's disease: opposite effects of stimulation in the pallidum. Mov Disord 13: 969-970. doi:https://doi.org/10.1002/mds.870130618. PubMed: 9827624.
  38. 38. Rodriguez-Oroz MC, Obeso JA, Lang AE, Houeto JL, Pollak P et al. (2005) Bilateral deep brain stimulation in Parkinson's disease: a multicentre study with 4 years follow-up. Brain 128: 2240-2249. doi:https://doi.org/10.1093/brain/awh571. PubMed: 15975946.
  39. 39. Odekerken VJ, van Laar T, Staal MJ, Mosch A, Hoffmann CF et al. (2013) Subthalamic nucleus versus globus pallidus bilateral deep brain stimulation for advanced Parkinson's disease (NSTAPS study): a randomised controlled trial. Lancet Neurol 12: 37-44. doi:https://doi.org/10.1016/S1474-4422(12)70264-8. PubMed: 23168021.
  40. 40. Lim TT, Fernandez HH, Cooper S, Wilson KM, Machado AG (2013) Successful DBS Surgery with Intraoperative MRI on a Difficult Neuroacanthocytosis Case. Neurosurgery 73: E184-E188. doi:https://doi.org/10.1227/01.neu.0000429852.45073.73. PubMed: 23615095.