Skip Navigation
Skip to contents

JMD : Journal of Movement Disorders

OPEN ACCESS
SEARCH
Search

Articles

Page Path
HOME > J Mov Disord > Volume 13(1); 2020 > Article
Brief communication
Rescue Levodopa/Carbidopa Intestinal Gel for Secondary Deep Brain Stimulation Failure
Juan Miguel Pilar Bautista1,2orcid, Genko Oyama1orcid, Maierdanjiang Nuermaimaiti1orcid, Satoko Sekimoto1orcid, Fuyuko Sasaki1orcid, Taku Hatano1orcid, Kenya Nishioka1orcid, Masanobu Ito3orcid, Atsushi Umemura4,5orcid, Yuji Ishibashi6orcid, Yasushi Shimo1,5orcid, Nobutaka Hattori1,5orcid
Journal of Movement Disorders 2020;13(1):57-61.
DOI: https://doi.org/10.14802/jmd.19051
Published online: January 31, 2020

1Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan

2Department of Neurology, St. Luke’s Medical Center, Quezon City, Philippines

3Department of Psychiatry, Juntendo University School of Medicine, Tokyo, Japan

4Department of Neurosurgery, Juntendo University School of Medicine, Tokyo, Japan

5Department of Research and Therapeutics for Movement Disorders, Juntendo University School of Medicine, Tokyo, Japan

6Department of Gastroenterology and Minimally Invasive Surgery, Juntendo University School of Medicine, Tokyo, Japan

Corresponding author: Genko Oyama, MD, PhD, FAAN Department of Neurology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan / Tel: +81-3-3813-3111 / Fax: +81-3-5800-0547 / E-mail: g_oyama@juntendo.ac.jp
• Received: June 16, 2019   • Revised: October 13, 2019   • Accepted: November 12, 2019

Copyright © 2020 The Korean Movement Disorder Society

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

  • 5,768 Views
  • 154 Download
  • 6 Web of Science
  • 6 Crossref
  • Objective
    The long-term efficacy of deep brain stimulation (DBS) for motor fluctuations in advanced Parkinson’s disease (PD) has been well established; however, motor fluctuations may recur over time despite multiple adjustments of DBS settings and medications.
  • Methods
    We conducted a retrospective chart review of three patients for whom levodopa-carbidopa intestinal gel (LCIG) was additionally administered as a rescue therapy for secondary DBS failure due to the recurrence of motor fluctuations.
  • Results
    The three patients had advanced PD with a disease duration of 14–19 years, and had undergone DBS for motor fluctuations refractory to standard medical management. LCIG was administered to the patients because of symptom recurrence years after DBS and provided complementary effects in all patients.
  • Conclusion
    The cases presented here show that rescue LCIG therapy may be a complementary treatment option for patients with post-DBS advanced PD who have a recurrence of troublesome motor complications.
Device-aided therapies such as deep brain stimulation (DBS) and levodopa-carbidopa intestinal gel (LCIG) are treatment options for patients with advanced Parkinson’s disease (PD) who present with motor fluctuations refractory to standard medical approaches. The major targets of DBS for PD are the subthalamic nucleus (STN) and globus pallidus internus (GPi) [1]. The longterm efficacy of DBS has been shown in several studies; however, “secondary DBS failure”, which represents a worsening of symptoms or a loss of response after initial improvement, has been reported [2,3]. Motor fluctuations may recur over 5 to 12 years, and dyskinesia might reappear within 1 year despite multiple adjustments of DBS settings and medications [4-7]. In these patients, the addition of LCIG, which has a different mechanism of action from DBS, might be beneficial in controlling motor fluctuation recurrence. Here, we describe our experience in 3 of 40 patients who underwent LCIG therapy between 2016 and 2018 at our institute, for whom LCIG was added as a rescue therapy after secondary failure of DBS treatment based on motor complication recurrence.
Case 1
A 71-year-old woman with PD had undergone bilateral STN DBS implantation at another institute because of wearing off at age 63. The left DBS lead was revised at our institute at age 65 for insufficient control of PD motor fluctuations because of a suboptimally placed lead. This resulted in good control of motor symptoms until age 70, when she was readmitted for the recurrence of motor complications and evaluation for possible LCIG therapy initiation.
In accordance with the standardized troubleshooting strategy at our institute [8], the Unified Parkinson’s Disease Rating Scale (part III) was used in assessments under four different conditions (off-medication/off-stimulation, off-medication/on-stimulation, on-medication/off-stimulation, and on-medication/on-stimulation). The evaluation revealed an excellent response to intravenous levodopa infusion (52.5% improvement in on-medication/off-stimulation) and a good response to DBS (35.0% in off-medication/on-stimulation), suggesting that the DBS setting was adequate. The patient was already taking levodopa/carbidopa tablets (10/10 mg half tablet five times per day) along with a combination of levodopa/carbidopa/entacapone tablets (50/5/100 mg five times per day) and long-acting pramipexole (1.5 mg once per day). However, owing to the persistence of motor complications, including peak-dose dyskinesia; fluctuations in nonmotor symptoms, such as anxiety and hallucinations; and mild impairment on the Frontal Assessment Battery (FAB: 15/18), although the Mini-Mental State Examination (MMSE) score was preserved (30/30), she decided to begin treatment with LCIG. LCIG immediately improved her off state but caused dyskinesia on her left side, which was improved after reducing the DBS setting for the right STN. With this change, motor and nonmotor complications were successfully controlled (Table 1). Her hallucinations improved after the dopamine agonist dose was reduced. She has been receiving DBS with adjunct LCIG therapy for 8 months.
Case 2
A 64-year-old woman who was diagnosed with PD at age 47 initially presented with left-leg rigidity and gait disturbance. She had a good response to the standard oral medications until wearing-off symptoms occurred at age 53. Owing to the worsening of motor fluctuations and peak-dose dyskinesia with excellent levodopa response (off medication, 40 points and on medication, 24 points; 40% improvement), she underwent bilateral STN DBS at age 58. The inclusion criteria for STN DBS in our institute were previously published elsewhere [9]. The motor fluctuations responded well to DBS, and DBS settings and medications were easily adjusted according to the symptoms. However, her motor fluctuations and dyskinesia gradually worsened, with freezing of gait, postural instability, and frequent falls, at age 63. On admission, she had been taking levodopa/carbidopa/entacapone tablets (50/5/100 mg) with additional entacapone tablets (100 mg) 11 times per day, as well as cabergoline (3 mg), pramipexole (4.125 mg), and zonisamide (25 mg). The four-condition test revealed an excellent response to intravenous levodopa infusion (37.5%) and a moderate response to DBS (27.0%; Table 1). Further contact screening and trials to optimize the DBS settings offered few additional benefits because of dysarthria. Mild impairment on the FAB (15/18) was also noted, although the MMSE score was preserved (28/30). She then agreed to add LCIG to her treatment, which improved the wearing-off and motor fluctuations but caused right-leg dyskinesia, which was easily managed by reducing left DBS stimulation. Other motor symptoms, mostly axial, such as freezing of gait and postural instability, remained the same after starting LCIG. The patient was on DBS-LCIG for 1 year, with reduced oral dopaminergic medications and stable motor symptoms.
Case 3
The patient was a 72-year-old woman diagnosed with PD at age 61; she initially presented with left-dominant tremor, rigidity, and mild depressive symptoms 2 years before diagnosis. The administration of levodopa, pramipexole, and an antidepressant improved her symptoms. She experienced wearing off, peakdose dyskinesia, and off-state dystonia in her lower limbs at age 70. She was taking various oral medications at this time, including half a tablet of levodopa/carbidopa (100/10 mg) with a tablet of entacapone 100 mg 6 times a day, and amantadine 100 mg once a day. Because her dyskinesia and off-state dystonia gradually worsened and she already had a mild decline in her FAB score (15/18), although the MMSE score was preserved (28/30), she underwent bilateral GPi DBS implantation. Her dyskinesia and dystonia dramatically improved; however, motor fluctuations, postural instability, and freezing of gait developed within 1 year after surgery. On admission, the four-condition test revealed a good motor response to intravenous levodopa infusion in addition to DBS (Table 1). Despite the reevaluation with contact screening and trials to optimize DBS settings, no further improvement was attained other than dyskinesia suppression. LCIG therapy was then started, which markedly improved the wearing-off symptoms without inducing dyskinesia. However, freezing of gait did not improve with the addition of duodenal levodopa infusion. Although LCIG therapy provided a better outcome, it was discontinued after 1 month owing to constraints at the nursing home where she was admitted because of her family circumstances.
We presented three cases of advanced-stage PD in patients who previously underwent DBS and benefited from the addition of LCIG therapy. All patients developed wearing-off symptoms after long-term treatment with DBS and manifested excellent benefits after the addition of LCIG therapy. In the patients who initially underwent bilateral STN DBS, LCIG therapy initiation caused unilateral dyskinesia, which was addressed by reducing contralateral STN stimulation. By contrast, the patient with bilateral GPi DBS tolerated increased LCIG doses without developing dyskinesia. In all three patients, multiple oral levodopa doses were replaced with LCIG. Our experience in these cases may indicate that DBS and LCIG can have complementary actions in reducing motor complications.
Adjunct LCIG therapy has two benefits in patients who previously underwent DBS. First, adding LCIG therapy can address the recurrence of motor fluctuations by continuous infusion of levodopa, thereby maintaining a more constant striatal dopamine concentration. In most cases, disease progression would involve the addition of medications and increased dosages, which may result in the recurrence of motor fluctuations in 7–10 years [4,5]. Klostermann et al. [10] reported two cases involving the addition of LCIG therapy in patients with long-term post-STN DBS that resulted in a 60–80% reduction in off-state time. In addition, they reported that hallucinations were reduced after replacing the dopamine agonist. Kumar et al. [11] reported seven cases of additional therapy with LCIG for post-DBS patients with different targets, including the STN, GPi, and pedunculopontine nucleus, and they reported that LCIG therapy reduced motor fluctuations regardless of the target. Thus, adding LCIG therapy can be considered a rescue treatment for the recurrence of motor fluctuations in cases of secondary failure in patients receiving DBS for a long time.
Second, DBS and LCIG, which have different mechanisms of action, can have complementary effects when used in combination. As shown in the cases presented here, LCIG therapy can improve wearing-off symptoms by achieving continuous drug delivery and dopaminergic stimulation. By contrast, DBS can separately manage unilateral symptoms by modulating basal ganglia circuits affected by dopaminergic deficits. Regidor et al. [12] conducted an open-label pilot study to compare a group treated with LCIG and bilateral STN DBS with a group treated with only LCIG. They found that among 19 patients who started double therapy, nine decided to continue both treatments [12]. Thus, in selected cases, LCIG with DBS can reduce motor complications. In the same study, five patients were successfully treated with maintenance LCIG monotherapy without replacing the implantable pulse generator after battery depletion. Moreover, five patients discontinued LCIG therapy and returned to DBS monotherapy because of adverse events caused by LCIG. This indicates that the initial selection of device-aided therapy should be carried out carefully. Patients with poor axial symptoms and older age may be more suitable for LCIG than DBS as an initial treatment [12].
Last, DBS on a second target has also been described as an option for the secondary failure of DBS on an original target [10]. The three cases we described herein included patients who had already developed a mild decline in frontal lobe function or a comorbid psychiatric condition prior to LCIG therapy; therefore, we were hesitant to perform additional DBS surgery. Thus, additional therapy with LCIG instead of rescue DBS or revision surgery may be preferable in cases with cognitive or psychiatric risk in the later stages of disease.
In summary, our cases show that rescue LCIG therapy may be a complementary treatment option for post-DBS patients with the recurrence of troublesome motor complications. While LCIG therapy may control motor fluctuations, it may also cause an asymmetricity of symptoms, such as dyskinesia, which can be controlled by adjusting the stimulation settings. However, such findings should be confirmed in a randomized controlled study with quality of life as the main outcome. Last, it is important that the initial selection of device-aided therapy should be carefully evaluated in consideration of various factors, including support and sustainability.

Ethical Compliance Statement

The authors confirm that institutional review board approval was not required for this work. We confirm that we have read the Journal’s position on issues regarding ethical publication and affirm that this work is consistent with the ethical guidelines.

Conflict of Interest

The authors have no financial conflicts of interest.

Financial Disclosures for the Past 12 Months

Dr. Oyama received speaker honoraria from Medtronic, Boston Scientific, Otsuka Pharmaceutical, Sumitomo Dainippon Pharma, Novartis Pharma, MSD, Nihon Medi-Physics, FP Pharmaceutical Corporation, Kyowa Hakko Kirin, and AbbVie, Inc. Dr. Shimo received speaker honoraria from Medtronic; Boston Scientific; Novartis Pharma; Nihon Medi-Physics; Otsuka Pharmaceutical; Sumitomo Dainippon Pharma, Co.; and Kyowa Hakko Kirin, Co. Dr. Hatano received honoraria from GSK K.K.; Nippon Boehringer Ingelheim, Co., Ltd.; FP Pharmaceutical, Co.; Otsuka Pharmaceutical, Co., Ltd.; Dai-nippon Sumitomo Pharma Co., Ltd.; Novartis Pharma K.K.; Nihon Medi-Physics Co., Ltd.; Kyowa Hakko Kirin Co., Ltd.; and Nihon Medi-Physics Co., Ltd. Dr. Umemura received honoraria from Boston Scientific Japan, Medtronic Japan, Novartis Pharma, and AbbVie, Inc. Dr. Hattori received honoraria from AbbVie GK; Alexion Pharmaceuticals; FP Pharmaceutical, Co.; Otsuka Pharmaceutical, Co., Ltd.; Kyowa Hakko Kirin Co., Ltd.; Daiichi Sankyo, Co.; Dai-Nippon Sumitomo Pharma, Co., Ltd.; Takeda Pharmaceutical, Co., Ltd.; Nippon Boehringer Ingelheim, Co., Ltd.; Medtronic, Inc.; MSD K.K.; Novartis Pharma K.K.; Pfizer Japan, Inc.; Boston Scientific, Co.; Mylan N.V.; Medtronic, Inc.; and Lund Beck Japan and donations from Astellas Pharma, Inc.; FP Pharmaceutical, Co.; Daiichi Sankyo, Co.; Takeda Pharmaceutical, Co., Ltd.; and Dai-Nippon Sumitomo Pharma, Co., Ltd. Drs. Bautista, Sekimoto, Sasaki, Ishibashi, Nishioka, and Ito has nothing to disclose. Mr. Nuermaimaiti has nothing to disclose.

Author Contributions

Conceptualization: Genko Oyama and Juan Miguel Pilar Bautista. Data curation: Juan Miguel Pilar Bautista and Maierdanjiang Nuermaimaiti. Investigation: Juan Miguel Pilar Bautista and Genko Oyama. Methodology: Genko Oyama. Supervision: Genko Oyama. Writing—original draft: Juan Miguel Pilar Bautista. Writing—review & editing: All authors.

Dr. Oyama was funded by grants from the Japan Society for the Promotion of Science, Grant-in-Aid for Young Scientists (B), Grant-in-Aid for Scientific Research (C), and Novartis Pharma. Dr. Shimo was funded by Novartis Pharma and Kyowa Hakko Kirin, Co. Dr. Hatano received research grants from a Grant-in-Aid for Scientific Research from Dai-nippon Sumitomo Pharma Co., Ltd., and Takeda Pharmaceutical, Co., Ltd. Dr. Umemura received funding as a consultant at Boston Scientific Japan, Medtronic Japan, and AbbVie, Inc.
Dr. Hattori received funding as a consultant for Dai-Nippon Sumitomo Pharma Co., Ltd., and as an advisory board member of Otsuka Pharmaceutical, Co., Ltd.; Takeda Pharmaceutical Co., Ltd.; Kyowa Hakko Kirin Co., Ltd.; Sanofi K.K.; and Dai-Nippon Sumitomo Pharma Co., Ltd. Drs. Bautista, Sekimoto, Sasaki, Ishibashi, Nishioka, and Ito has nothing to disclose. Mr. Nuermaimaiti has nothing to disclose.
Table 1.
Summary of cases
Case 1 (70 year old, female) Case 2 (65 year old, female) Case 3 (73 year old, female)
Age at onset (years) 56 46 59
Age at DBS (years) 63 (L STN was revised at age 65) 58 70
Age at LCIG (years) 71 64 72
DBS target Bilateral STN Bilateral STN Bilateral GPi
Reason for DBS Wearing off and dyskinesia Wearing off and dyskinesia Wearing off, dyskinesia, and off-dystonia
Reason for LCIG Wearing off, dyskinesia, and hallucination due to DA Wearing off and dyskinesia Wearing off
Four-condition test*
 Off-medication/Off-stim 40 48 87
 Off-medication/On-stim 26 (-35.0%) 35 (-27.0%) 80 (-8.1%)
 On-medication/Off-stim 19 (-52.5%) 30 (-37.5%) 65 (-25.3%)
 On-medication/On-stim 15 (-62.5%) 28 (-41.2%) 54 (-37.9%)
UPDRS part III scores (on-medication/on-stim) 19/15 39/33 43/33
UPDRS part IV scores 8/7 8/2 NA
LEDD (mg) 652.5/960 1,327.5/1,246 1,430/1,688.5
LCIG settings
 Morning dose (mL) 8.0 6.8 13.0
 Continuous dose (mL/h) 2.5 3.0 4.0
 Extra doses (mL) 1.0 1.0 1.0
DBS settings (pre-LCIG) L STN1: 1(−)C(+), 2.6 mA, 60 µs, 125 Hz L STN: 2(−)C(+), 2.6 mA, 60 µs, 130 Hz L GPi: 2(−)C(+), 3.2 mA, 90 µs, 130 Hz
L STN2: 2(−)C(+), 2.4 mA, 60 µs, 125 Hz R STN1: 1(−)C(+), 2.3 mA, 60 µs, 125 Hz R GPi1: 3(−)C(+), 3.1 mA, 60 µs, 125 Hz
R STN1: 1(−)C(+), 2.5 mA, 60 µs, 125 Hz R STN2: 2(−)C(+), 2.5 mA, 60 µs, 125 Hz R GPi2: 2(−)1(+), 3.0 mA, 60 µs, 125 Hz
R STN2: 2(−)C(+), 2.0 mA, 60 µs, 125 Hz
DBS settings (post-LCIG) L STN1: 1(−)C(+), 2.6 mA, 60 µs, 125 Hz L STN: 2(−)C(+), 1.9 mA, 60 µs, 200 Hz L GPi: 2(−)C(+), 2.9 mA, 90 µs, 130 Hz
L STN2: 2(−)C(+), 2.4 mA, 60 µs, 125 Hz R STN1: 1(−)C(+), 2.3 mA, 60 µs, 125 Hz R GPi1: 3(−)C(+), 3.2 mA, 60 µs, 125 Hz
R STN1: 1(−)C(+), 2.3 mA, 60 µs, 125 Hz R STN2: 2(−)C(+), 2.5 mA, 60 µs, 125 Hz R GPi2: 2(−)1(+), 3.3 mA, 60 µs, 125 Hz
R STN2: 2(−)C(+), 2.0 mA, 60 µs, 125 Hz
Oral medications (pre-LCIG) Levodopa/carbidopa 100/10 mg (1/2 tablet ×5), levodopa/carbidopa/entacapone 50/5/100 (×5), pramipexole 2 mg, and selegiline 5 mg Levodopa/carbidopa/entacapone 50/10/100 mg (×11), entacapone 100 mg (×11), cabergoline 3 mg, pramipexole 4.125 mg, zonisamide 25 mg, droxidopa 600 mg, donepezil 10 mg, and clonazepam 0.5 mg Levodopa/carbidopa (100/10 mg) 1,100 mg (6×, q3h), entacapone 600 mg (6×, q3h), amantadine 100 mg, and donepezil 10 mg
Oral medications (post-LCIG) Donepezil 5 mg, memantine 20 mg, quetiapine 50 mg, clonazepam 0.5 mg, and etizolam 0.5 mg Pramipexole LA 1.5 mg, donepezil 10 mg, and droxidopa 300 mg Rotigotine 13.5/24 h (overnight) and donepezil 10 mg
Outcomes 1. Motor fluctuations improved with LCIG 1. Motor fluctuations improved with LCIG 1. Motor fluctuations improved with LCIG
2. Hallucination improved with the discontinuation of DA 2. Right-sided dyskinesia improved with DBS adjustment on the left side 2. Dyskinesia improved by increasing bilateral GPi stim
3. Left-sided dyskinesia improved with DBS adjustment on the right side 3. LCIG discontinued after a month because of inability to maintain use

* UPDRS part III score (% improvement),

Pre-LCIG/post-LCIG.

DBS: deep brain stimulation, STN: subthalamic nucleus, LCIG: levodopa-carbidopa intestinal gel, GPi: globus pallidus internus, LA: long-acting, DA: dopamine agonist, UPDRS: Unified Parkinson’s Disease Rating Scale, NA: not available, LEDD: levodopa equivalent daily dose, stim: stimulation, R: right, L: left.

  • 1. Okun MS. Deep-brain stimulation for Parkinson’s disease. N Engl J Med 2012;367:1529–1538.ArticlePubMed
  • 2. Allert N, Lehrke R, Sturm V, Volkmann J. Secondary failure after ten years of pallidal neurostimulation in a patient with advanced Parkinson’s disease. J Neural Transm (Vienna) 2010;117:349–351.ArticlePubMedPDF
  • 3. Pauls KAM, Krauss JK, Kämpfer CE, Kühn AA, Schrader C, Südmeyer M, et al. Causes of failure of pallidal deep brain stimulation in cases with pre-operative diagnosis of isolated dystonia. Parkinsonism Relat Disord 2017;43:38–48.ArticlePubMed
  • 4. Krishnan S, Prasad S, Pisharady KK, Sarma G, Sarma SP, Kishore A. The decade after subthalamic stimulation in advanced Parkinson’s disease: a balancing act. Neurol India 2016;64:81–89.ArticlePubMed
  • 5. Nakajima A, Oyama G, Jo T, Shimo Y, Umemura A, Nakajima M, et al. Rescue pallidal stimulation for diphasic and stimulation-induced dyskinesia after successful subthalamic stimulation for Parkinson’s disease. Neurol Clin Neurosci 2017;5:127–128.Article
  • 6. Matias CM, Silva D, Machado AG, Cooper SE. “Rescue” of bilateral subthalamic stimulation by bilateral pallidal stimulation: case report. J Neurosurg 2016;124:417–421.ArticlePubMed
  • 7. Sriram A, Foote KD, Oyama G, Kwak J, Zeilman PR, Okun MS. Brittle dyskinesia following STN but not GPi deep brain stimulation. Tremor Other Hyperkinet Mov (N Y) 2014;4:242.ArticlePubMedPMC
  • 8. Oyama G, Shimo Y, Umemura A, Nishikawa N, Nakajima A, Takayuki J, et al. Troubleshooting in hospitalized Parkinson’s disease patients with a history of deep brain stimulation of the subthalamic nucleus. Neurol Clin Neurosci 2014;2:188–192.Article
  • 9. Oyama G, Shimo Y, Natori S, Nakajima M, Ishii H, Arai H, et al. Acute effects of bilateral subthalamic stimulation on decision-making in Parkinson’s disease. Parkinsonism Relat Disord 2011;17:189–193.ArticlePubMed
  • 10. Klostermann F, Jugel C, Marzinzik F. Jejunal levodopa infusion in longterm DBS patients with Parkinson’s disease. Mov Disord 2011;26:2298–2299.ArticlePubMed
  • 11. Kumar N, Murgai A, Naranian T, Jog M, Fasano A. Levodopa-carbidopa intestinal gel therapy after deep brain stimulation. Mov Disord 2018;33:334–335.ArticlePubMed
  • 12. Regidor I, Benita V, Del Álamo de Pedro M, Ley L, Martinez Castrillo JC. Duodenal levodopa infusion for long-term deep brain stimulationrefractory symptoms in advanced Parkinson disease. Clin Neuropharmacol 2017;40:103–107.ArticlePubMed

Figure & Data

References

    Citations

    Citations to this article as recorded by  
    • Nationwide Retrospective Analysis of Combinations of Advanced Therapies in Patients With Parkinson Disease
      Dominik Pürner, Mohammad Hormozi, Daniel Weiß, Michael T. Barbe, Hannah Jergas, Tino Prell, Eileen Gülke, Monika Pötter-Nerger, Björn Falkenburger, Lisa Klingelhöfer, Pia K. Gutsmiedl, Bernhard Haslinger, Angela M. Jochim, Andreas Wolff, Nils Schröter, Mi
      Neurology.2023;[Epub]     CrossRef
    • Combining Device‐Aided Therapies in Parkinson's Disease: A Case Series and a Literature Review
      Iro Boura, Nikolaos Haliasos, Ιrene‐Areti Giannopoulou, Dimitrios Karabetsos, Cleanthe Spanaki
      Movement Disorders Clinical Practice.2021; 8(5): 750.     CrossRef
    • Combined and Sequential Treatment with Deep Brain Stimulation and Continuous Intrajejunal Levodopa Infusion for Parkinson’s Disease
      Daniël van Poppelen, Annelie N.M. Tromp, Rob M.A. de Bie, Joke M. Dijk
      Journal of Personalized Medicine.2021; 11(6): 547.     CrossRef
    • Personalised Advanced Therapies in Parkinson’s Disease: The Role of Non-Motor Symptoms Profile
      Valentina Leta, Haidar S. Dafsari, Anna Sauerbier, Vinod Metta, Nataliya Titova, Lars Timmermann, Keyoumars Ashkan, Michael Samuel, Eero Pekkonen, Per Odin, Angelo Antonini, Pablo Martinez-Martin, Miriam Parry, Daniel J. van Wamelen, K. Ray Chaudhuri
      Journal of Personalized Medicine.2021; 11(8): 773.     CrossRef
    • Parkinson’s Kinetigraph in the Selection of Levodopa-Carbidopa Intestinal Gel for Motor Fluctuations Refractory to Deep Brain Stimulation
      Yassine Noui, Monty Adam Silverdale, Julian Evans, Lucy Partington-Smith, Christopher Kobylecki
      Journal of Movement Disorders.2021; 14(3): 239.     CrossRef
    • The Choice Between Advanced Therapies for Parkinson’s Disease Patients: Why, What, and When?
      Joke M. Dijk, Alberto J. Espay, Regina Katzenschlager, Rob M.A. de Bie, Bastiaan R. Bloem, Patrik Brundin
      Journal of Parkinson's Disease.2020; 10(s1): S65.     CrossRef

    Comments on this article

    Add a comment

    JMD : Journal of Movement Disorders