Levodopa is the most effective agent to alleviate motor dysfunction in Parkinson’s disease but its long-term use is associated with the development of dyskinesias. Although the pathogenic processes behind the development of levodopa-induced dyskinesias are still being elucidated, it appears that chronic administration of this short-lived agent results in nonphysiologic pulsatile stimulation of striatal neurons and abnormal firing patterns in the basal ganglia. Dyskinesias have been associated with decreased quality of life, and a number of methodologies to evaluate severity of dyskinesias are now available. Strategies to avoid, reduce, or eliminate dyskinesias include providing more continuous dopaminergic stimulation, administering an antidyskinetic agent, and surgery. Several new compounds that may provide an antidyskinetic effect are also under investigation.

Parkinson’s disease (PD) is a degenerative disorder characterized by the loss of substantia nigra pars compacta dopaminergic neurons and the subsequent loss of dopaminergic input to the striatum. As the degenerative process evolves, dopamine replacement therapy becomes necessary to help alleviate motor dysfunction. Four decades after its introduction, levodopa remains the most effective agent to improve parkinsonian motor symptoms, but chronic use is associated with the emergence of motor complications that include response fluctuations and dyskinesias. This has raised the question of how best to use levodopa in the management of PD.

The pathophysiologic mechanisms that underlie motor complications are not completely understood, but substantial support has been garnered for the idea that nonphysiologic pulsatile stimulation of striatal neurons contributes to a disturbance of basal ganglia homeostasis [1, 2]. Motor complications include motor fluctuations, defined as a loss of clinical benefit before the next levodopa dose (i.e., effect wearing off), and abnormal involuntary movements, termed dyskinesias. With current treatment, motor fluctuations usually precede dyskinesias [3]. Notably, the development of one is a risk factor for the development of the other [3].

Dyskinesias are usually choreiform but may resemble dystonia, myoclonus, or other movement disorders [4]. Peak-dose dyskinesias are the most common type of dyskinesia. They occur during peaks of levodopa-derived dopamine in the brain, when the patient is otherwise experiencing a beneficial response (the ‘on’ state) [4]. Peak-dose dyskinesias worsen with increases in dopaminergic therapy and lessen with reductions in dopaminergic therapy. Some patients exhibit diphasic dyskinesia or dyskinesia–improvement–dyskinesia, which occurs when levodopa-derived dopamine concentrations are increasing or decreasing and the patient is turning ‘on’ and ‘off’ [4]. Diphasic dyskinesias are usually more dystonic and preferentially involve the lower extremities compared to peak-dose dyskinesias.

When patients experience motor fluctuations without dyskinesias, it is usually a relatively simple matter to smooth out the clinical response. Levodopa doses can be given closer together, or adjunctive medications that reduce ‘off’ time can be added. However, once a patient has motor fluctuations and peak-dose dyskinesias, it becomes difficult to smooth the clinical response. Increases in dopaminergic treatment then increase dyskinesias, and decreases in dopaminergic treatment reduce dyskinesias but worsen parkinsonian motor symptoms. Although for many patients dyskinesias are not disabling, they create a barrier to adequate treatment of fluctuations and parkinsonian symptoms. This review provides an update on dyskinesias; specifically, their etiology, underlying mechanisms of development, and impact on quality of life (QoL). It also considers pharmacologic and surgical strategies to reduce their development and frequency.

Levodopa

The underlying pathophysiologic mechanisms responsible for the development of levodopa-induced dyskinesias are not fully understood, though both severity of the dopamine neuron loss and chronic intermittent administration of a drug with a short half-life, such as levodopa, are of critical importance (fig. 1) [1, 2, 5]. Under normal physiologic conditions, presynaptic dopaminergic neurons in nigrostriatal pathways fire tonically, resulting in a relatively continuous release of dopamine that causes a steady stimulation of postsynaptic striatal receptors [6, 7]. As PD advances, the loss of striatal dopaminergic neurons and terminals leads to a diminished ability to form, store, and regulate the release of dopamine, and the remaining receptors rely on the availability of exogenous dopaminomimetic agents [7, 8]. The short half-life (approximately 90 min) of levodopa, when administered with a peripheral dopa decarboxylase inhibitor such as carbidopa, causes intermittent (pulsatile) stimulation of striatal dopaminergic receptors [1, 7, 9]. This chronic pulsatile stimulation alters cell signals in striatal dopaminergic spiny neurons, causing potentiation of GABAergic efferents, particularly N-methyl-D-aspartate (NMDA) glutamate receptors [6, 7, 10]. Glutamate-mediated striatal sensitization, which results in nonphysiologic stimulation, triggers a cascade of events involving the induction of certain intracellular messengers and proteins, leading to abnormal basal ganglia firing patterns that result in dyskinesias [1, 9]. Thus, the proposal that continuous dopaminergic stimulation (CDS) leads to fewer motor complications has been the rationale for many new therapeutic strategies and interventions.

Fig. 1

Steps in the genesis of levodopa-induced motor complications [5].

Fig. 1

Steps in the genesis of levodopa-induced motor complications [5].

Close modal

The overall duration of levodopa treatment [11, 12], disease severity [3, 11, 13], age of onset [12,14,15,16,17], levodopa dosage [3,17,18,19], and initial levodopa dosage [12, 14] are the most significant risk factors for the development of dyskinesias. Of these, the most influential factor associated with levodopa-induced dyskinesias is the duration of levodopa therapy [11, 12]. Dyskinesias occur in approximately 30% of patients by 5 years of treatment [14] and in 59–100% of patients by 10 years [12, 14, 20]. With respect to disease severity at the time levodopa is introduced, a review of the literature [13] showed that median dyskinesia frequency was 50% by 5–6 months of treatment when pre-levodopa era patients first received the medication but that it is slightly less than 40% by 4–6 years of treatment among levodopa era patients. The mean duration of disease was 6–10 years in the pre-levodopa era, in contrast to 2–3 years in modern-era patients [13]. Age of onset is also an important risk factor, with the 5-year incidence of dyskinesia greater than 50% when PD develops in patients 40–59 years of age and 16% when it develops in patients after age 70 [15]. The ELLDOPA (Earlier vs. Later Levodopa Therapy in Parkinson Disease) study showed the importance of levodopa dosage as a risk factor for dyskinesia. In this study, patients with early PD who were randomly assigned to levodopa 200 mg three times daily experienced significantly more dyskinesia than those who received a lower dose or placebo [18, 19]. Of note is the fact that motor fluctuations and dyskinesias appear to be interrelated because the presence of one is associated with the earlier development of the other [3].

Other Agents

Although more commonly associated with levodopa, dyskinesias can also occur with dopamine agonist monotherapy [21,22,23]. The development of dyskinesia in some patients treated with dopamine agonists that have relatively long half-lives (ropinirole, 6 h; pramipexole, 8 h) or very long half-lives (cabergoline, 68 h) suggests that, to some extent, dopamine stimulation can cause dyskinesias even when provided in a continuous fashion.

Other medications can cause or worsen dyskinesias in patients already taking levodopa. In patients who are already exhibiting levodopa-induced dyskinesia, adjunctive medication that increases dopaminergic stimulation commonly increases dyskinesia. In addition, in some patients who have been treated with levodopa but in whom dyskinesia is not yet manifest, dyskinesia may emerge when the levodopa dose is increased or when adjunctive medications are introduced. In these cases, it is thought that the patient has already developed a lowered threshold for dyskinesia, which then becomes apparent when dopamine stimulation is increased. New or worsening dyskinesia has been reported in levodopa-treated patients when adjunctive medications are added, including dopamine agonists [24,25,26], the catechol-O-methyltransferase (COMT) inhibitors entacapone [27, 28] and tolcapone [29], and the monoamine oxidase type B inhibitors selegiline [30, 31] and rasagiline [32].

Quality of Life Scales

Some scales used to evaluate dyskinesia depend on a rater assessment at a single point in time, while others gather historical information provided by the patient or caregiver over a period of time [33]. Some scales are dyskinesia specific, whereas others include an evaluation of dyskinesia as part of a multidimensional patient assessment.

The Unified Parkinson’s Disease Rating Scale has proven to be reliable and valid [34], and it is the most widely used rating scale for measuring severity of parkinsonian symptoms in clinical research and practice [35]. It includes a patient- or caregiver-provided estimate of dyskinesia duration during the waking day (0, 1–25, 26–50, 51–75 or 76–100%) for the past week and a rating of its severity (not disabling, mildly disabling, moderately disabling, severely disabling or completely disabling) [36]. In clinical trials, dyskinesia has been considered present if the duration item response is at least 1–25% of the waking day [23]. The major limitation of the dyskinesia items of this rating scale is that they are rough estimates based on patient recall.

The Abnormal Involuntary Movement Scale [37] was originally developed to assess tardive dyskinesia, but it is also used to assess patients with PD and possible dyskinesia in clinical trials. It is a 12-item, clinician-administered scale that assesses the presence and severity of involuntary movements of the face, mouth, tongue, extremities, and trunk. Body parts are rated on a scale of 0–4, with 0 indicating no abnormal movement and 4 indicating severe symptoms [37]. It is familiar to clinicians, can be completed quickly, and can be used in routine clinical settings and clinical trials. Its major limitation is that it captures dyskinesia at one moment in time and does not assess functional impairment [36]. A modified version for use in patients with PD is commonly used in clinical trials [38, 39].

The Goetz Dyskinesia Rating Scale [40] is a physician- or study coordinator-completed scale in which the rater assesses severity and functional disability associated with the patient’s dyskinesias while observing the patient performing specific tasks. Its main advantage is the documentation of the functional impairment caused by dyskinesias at the time of assessment [36], and it has good interrater and intrarater reliability [40]. On the other hand, its disadvantages include only evaluating function over a short time period (minutes), and the office examination may not reflect the patient’s condition at home. In addition, completion of the scale is somewhat labor intensive and time consuming [36].

The Unified Dyskinesia Rating Scale is under development and is projected to be introduced, together with instructions and teaching tools, by early 2008. Its goal is to eliminate the use of disparate scales, relying on various clinical assessments in clinical research, and to increase the robustness of trials evaluating dyskinesia treatments.

A PD-specific home diary to assess functional status in patients with motor fluctuations and dyskinesia has been validated for use in clinical trials [41, 42]. The diary asks patients to indicate their predominant status over half-hour intervals based on the following categories: ‘asleep’; ‘off’; ‘on’ without dyskinesia; ‘on’ with nontroublesome dyskinesia, and ‘on’ with troublesome dyskinesia [42]. Time ‘on’ without troublesome dyskinesia was demonstrated to most strongly correlate with a patient’s perceived duration of a good response through the day and is an important outcome variable [42]. In recent years, home diaries have been routinely used in clinical trials investigating new treatments for PD. The major advantage of the diary is that it allows evaluation of the patient’s status through the course of the day in the patient’s own environment. It also allows the patient to determine whether the dyskinesia experienced during any time period is troublesome (interferes with activity or causes meaningful discomfort). A major limitation of the diary is that patients must be adequately trained to provide accurate responses and, despite this training, they may still make errors such as confusing the ‘off’ with tremor state and the ‘on’ with dyskinesia state. In addition, diaries are subject to patient compliance and recall.

Clinical Perspectives

QoL for PD patients is more impaired than it is for healthy elderly people [43] and for patients with chronic conditions [44]. Reduced QoL significantly correlates with disease progression [16, 45, 46] and duration [16]. Although it is apparent that PD patients have reduced QoL, the impact of dyskinesias on QoL remains controversial. An observational study of patients in 3 European countries found that dyskinesias do have a significant impact on the QoL of PD patients [47]. In addition, dyskinesias have been reported to adversely affect various dimensions of QoL – including mobility [48], activities of daily living [48, 49], stigma [48], communications [48, 49], and bodily discomfort [49] – as assessed by the PDQ-39 Parkinson’s Disease Questionnaire. Increasing severity of dyskinesias is also associated with increasing depression [47]. Other studies have shown that dyskinesias can seriously interfere with the performance of activities of daily living [50], ambulation, and balance [51], and that they can be associated with increased falls [52] and unintended weight loss [53]. On the other hand, other investigators have reported that dyskinesias do not have a significant impact on QoL [16, 35, 54] and may not pose a major concern for patients [14, 55, 56]. Differences in QoL study results may be attributed to variability in methodology and to the patient populations studied.

In the Sydney Multicenter Study, Hely et al. [55] found that 94% of PD patients experienced dyskinesias by 15 years, and 46% considered their dyskinesia disabling. Conversely, 54% did not consider their dyskinesias disabling, and in only 12% were dyskinesias rated severe. Similarly, a clinical review of patients treated with levodopa for 5–10 years showed that only 12% had clinically significant dyskinesias [56]. More recently, a population-based study [14] revealed that dyskinesias can be expected to develop in nearly 60% of patients at 10 years and that in 43% they would be severe enough to necessitate medication adjustments. However, in only 12% could dyskinesia not be controlled with medication adjustments [14]. Nonetheless, it is unclear what effects these medication adjustments had on potential worsening of motor fluctuations and parkinsonian motor features. Dyskinesias have also been associated with significant increases in health care costs [47, 57], and strategies to delay the onset of dyskinesias or to alleviate them can significantly reduce health care expenditure.

From a clinical perspective, patients may have mild dyskinesias without apparent adverse impact on QoL. In fact, most patients prefer mild dyskinesias to being ‘off’ when they may feel immobile and constricted. However, marked dyskinesias can cause functional impairment and patient discomfort. Therefore, the effect of dyskinesias on QoL may very much depend on the severity of the dyskinesias. In addition, it must be recognized that the presence of dyskinesias limits the ability of the physician to increase PD medications to reduce ‘off’ time. Therefore, the actual impact of dyskinesias may come not only from disability caused by dyskinesia itself but also from motor fluctuations and ‘off’ time that cannot be adequately treated as a result.

Restoration of striatal dopaminergic stimulation is the goal in the treatment of parkinsonian motor symptoms. Levodopa provides the greatest benefit for treating parkinsonian motor dysfunction, but because its use is associated with the development of motor complications, one of the great unmet needs for the treatment of PD is a medication that will match the efficacy of levodopa but not cause motor complications. Until such a medication is available, it is useful to identify treatment strategies that can provide adequate efficacy while minimizing motor complications.

The short half-life of levodopa and the resultant pulsatile dopaminergic stimulation appear at least in part to be responsible for the development of motor complications [1, 6, 7, 9, 10]. Therefore, CDS may delay the onset of dyskinesias in early disease and alleviate dyskinesias in advanced disease.

Early Disease: Avoiding the Development of Dyskinesia

Early disease strategies include administering a dopamine agonist as initial therapy, adding levodopa when the agonist alone is no longer able to adequately control parkinsonian symptoms, and using methods to deliver levodopa in a more continuous fashion.

Initiating Therapy with an Agonist and Adding Levodopa when Necessary

Animal data suggest that pulsatile stimulation with short-acting agents is the driving force in the genesis of dyskinesias [58]. Conversely, these short-acting agents do not induce dyskinesias when given in a continuous fashion [58]. In drug-naive, MPTP-lesioned monkeys, the administration of longer-acting dopamine agonists results in significantly less dyskinesia than does levodopa [59, 60]. However, once a long-acting agonist is administered to animals already primed to exhibit dyskinesias with levodopa, the resultant dyskinesias are comparable to those seen in the levodopa group [59]. An MPTP-marmoset study [60] evaluating the combination of ropinirole, a long-acting agonist, plus levodopa showed that the levodopa-dominant group (mostly levodopa) had increasingly intense dyskinesias as the study progressed, whereas the ropinirole-dominant combination produced no greater intensity of dyskinesias than was produced by ropinirole alone.

Clinical studies randomly assigning patients to initial treatment with a dopamine agonist or levodopa have shown a lower risk for dyskinesias in the agonist-treated groups (agonists studied have included pramipexole [22, 61], ropinirole [23, 62], bromocriptine [63, 64], pergolide [65], and cabergoline [21]). Retrospective analyses have demonstrated that once levodopa is added, the rate of development of dyskinesias is the same regardless of whether or not the patient was already taking a dopamine agonist [62, 66]. Therefore, it appears that the benefit of initial treatment with a dopamine agonist in lowering the incidence of dyskinesias is related to the ability of the agonist to delay the need for levodopa [62, 66].

Delivering Levodopa in a More Continuous Fashion

Optimizing levodopa therapy by adding an agent that can prolong its half-life and deliver it in a less pulsatile manner is another promising way to achieve CDS. Levodopa is metabolized peripherally by aromatic amino acid decarboxylase and COMT. The combination of levodopa/carbidopa plus the COMT inhibitor entacapone can reduce the peripheral conversion of levodopa and extend the levodopa half-life to 2.5 h [67]. In MPTP-treated monkeys, Jenner [68] showed that coadministration of the same dose of levodopa 4 times a day with entacapone improved parkinsonian motor response and caused less dyskinesia than treatment with levodopa alone. Peak dose dyskinesia scores and dyskinesia duration were decreased [68]. Rat studies showed similar results [69], supporting the theory that reducing pulsatile delivery of levodopa leads to fewer motor complications. Clinical trials are now under way in which patients in need of levodopa therapy are randomly assigned to treatment with levodopa and carbidopa, or levodopa, carbidopa and entacapone. The aim is to determine whether the introduction of entacapone when levodopa and carbidopa are first administered will lower the rate of dyskinesia onset.

It was previously hoped that sustained-release levodopa and carbidopa might be able to decrease the rate of dyskinesia onset. However, a double-blind, multicenter study comparing immediate-release carbidopa-levodopa twice daily (b.i.d.) with controlled-release carbidopa-levodopa therapy b.i.d. in patients with PD proved otherwise and showed no statistical difference in the incidence of dyskinesia between the 2 treatment groups during the first 5 years of treatment [70]. However, the dosing of controlled-release carbidopa-levodopa b.i.d. might have been insufficient to achieve continuous stimulation.

Advanced Disease: Reducing or Eliminating Established Dyskinesias

PD patients eventually need levodopa to help alleviate parkinsonian motor features. When dyskinesia emerges, one strategy is to dose levodopa so that it peaks just below the dyskinesia threshold and administer it frequently enough to avoid wearing off. This typically amounts to administering levodopa in smaller doses more often, which may be inconvenient and result in reduced compliance. At the extreme, liquid levodopa (which is not commercially available but can be easily prepared [71]) can be used to deliver very small doses of levodopa very frequently (usually every hour) [71, 72]; however, patients usually find this very inconvenient.

Another strategy is to use higher doses of a dopamine agonist to reduce both the total daily levodopa dose and its frequency [73] or to gradually substitute a dopamine agonist for levodopa [72]. Unfortunately, these strategies rarely work and typically reduce dyskinesias at the expense of less satisfactory control of parkinsonian symptoms.

Alternative routes of levodopa administration, such as intraintestinal [39, 74, 75] or intravenous infusion [76], provide more sustained levodopa delivery and reduce motor complications. Stocchi et al. [39] demonstrated significant reductions in ‘off’ time and dyskinesia after 6 months of intraintestinal levodopa infusion. Infusion was associated with continuously elevated levodopa concentrations that avoided wide fluctuations and especially low troughs [39]. This suggests that the benefits for dyskinesia may not be attributed to maintaining levodopa levels just below the dyskinesia threshold. Rather, by providing high concentrations that avoid levodopa troughs, desensitization may be induced over time. Continuous intraduodenal levodopa infusion has been evaluated in small trials [74, 75, 77, 78] and warrants further study. It may be an option for patients with marked fluctuations and dyskinesia in whom deep-brain stimulation (DBS) is contraindicated or not possible due to advanced age, or it may provide an alternative to DBS. Continuous subcutaneous infusion of apomorphine has also been used in Europe for almost 2 decades [51,79,80,81,82,83,84].

As the therapeutic window for levodopa narrows and the balance between alleviating dyskinesias without compromising motor status becomes more difficult, amantadine (200–400 mg/day, in 2–4 divided doses) can be considered [72]. Amantadine, a noncompetitive NMDA antagonist, appears to have antidyskinetic effects, and several studies have assessed its efficacy and safety [85,86,87,88,89,90,91,92]. Some studies suggest that the benefits of amantadine persist for only a few weeks [90] or months [87], but others suggest that amantadine has a more long-lasting effect [91]. An adequate trial of amantadine may be appropriate before surgery for severe dyskinesias is considered [91].

Clozapine is an atypical antipsychotic that has been assessed for the treatment of drug-induced psychosis in PD. It may also be effective in decreasing dyskinesias [72], and a few studies have focused on its antidyskinetic effect [93, 94]. However, the hematologic monitoring necessary with clozapine can be burdensome for both the patient and the physician and can limit its usefulness.

Many other available drug therapies have been evaluated for the treatment of dyskinesias, but nearly all studies were small and had other methodologic limitations [95]. Other noncompetitive NMDA antagonists, such as dextromethorphan [96, 97] and remacemide, have been studied. The latter was studied in a larger trial and was found to be safe and well tolerated but did not show any demonstrable anti-dyskinetic benefit [36]. Topiramate, an AMPA glutamate antagonist, significantly reduced levodopa-induced dyskinesias in the MPTP-lesioned marmoset model of PD [98]. In a small open-label study, the antiepileptic drug levetiracetam significantly improved the percentage of the awake day ‘on’ without dyskinesia or with nontroublesome dyskinesia but was frequently associated with somnolence [99]. Two ongoing randomized, double-blind, placebo-controlled studies in Europe are assessing the use of levetiracetam in the management of levodopa-induced dyskinesias. Buspirone, a 5-HT1A receptor partial agonist [100, 101], fluoxetine, a serotonin reuptake inhibitor [102], and idazoxan, an alpha-2 antagonist [103], may reduce dyskinesia without worsening parkinsonism. Mirtazapine – a 5-HT1A agonist, a 5-HT2 antagonist, and an alpha-2 antagonist – was moderately effective in reducing levodopa-induced dyskinesia, alone or together with amantadine [104]. A variety of novel compounds, such as adenosine A2A antagonists [105,106,107] and serotoninergic agonists [108,109,110], are also under investigation.

Diphasic dyskinesias are difficult to treat [72]. In theory, maintaining continuous high levels of dopaminergic stimulation should avoid the turning ‘off’ and turning ‘on’ phases of the levodopa cycle in which these dyskinesias occur. However, this is usually only partially successful, and using higher levodopa doses may encourage the development of peak dose dyskinesias and other levodopa-induced adverse effects. Amantadine and clozapine are only partially effective. Patients with severe, medically refractory peak dose dyskinesias or diphasic dyskinesias should be considered for DBS [72].

Abnormal basal ganglia output neurons communicate inappropriate information to cortical and brain stem motor regions [111], and surgical procedures are thought to act by interrupting these abnormal neuron firing patterns [112]. Surgical approaches can be highly effective in reducing dyskinesias. Dyskinesias and motor fluctuations are the main reasons for surgery in patients with PD [73]. Patients with PD who may benefit from surgery include those who have substantial dyskinesias unresponsive to medication adjustments, are levodopa responsive, do not have dementia, and do not have neuropsychiatric impairment [95]. DBS is the most frequently performed surgery for PD in North America [95]. In patients with advanced PD, DBS of the globus pallidus interna (GPi) or the subthalamic nucleus (STN) has been shown to reduce dyskinesia severity by up to 89% [113, 114] and to reduce the duration of dyskinesias by 86% [115]. Long-term follow-up studies in patients who underwent STN DBS showed sustained improvements in dyskinesia 4 years [116,117,118] and 5 years after surgery [119]. STN DBS is better than medical management alone [120]. It provides significant improvement in parkinsonian motor features and allows a reduction of dyskinesias, in part through the subsequent reduction of levodopa [121, 122]. Evidence indicates that GPi DBS has a greater direct effect on reducing dyskinesia [122], and studies directly comparing STN and GPi DBS are underway.

DBS is associated with relatively low rates of serious intraoperative and perioperative complications (e.g., ischemic stroke, subdural hematoma, intracranial hemorrhage, infection) [113, 114]. Adverse effects can be transient (e.g., mild delirium, anxiety, hallucinations, cognitive changes) [113, 114 ]or persistent (e.g., cognitive impairment, hemiparesis, aphasia, hallucinations, hypomania or mania) [117, 118].

Levodopa continues to be the mainstay of medical therapy for PD, but it is associated with the emergence of dyskinesias. Because of its short half-life, its administration leads to pulsatile stimulation of striatal neurons with resultant intracellular events that are expressed clinically as dyskinesias. Risk factors for dyskinesias are disease duration, duration of levodopa therapy, levodopa dose, and age. The emergence of dyskinesias has been associated with decreased QoL and increased health care costs.

One strategy to address levodopa-induced dyskinesias is to attempt to provide CDS to delay the onset of dyskinesias in early disease and to alleviate dyskinesias in advanced disease. Strategies in early disease include using initial treatment with a dopamine agonist and adding levodopa later when parkinsonian motor symptoms progress, and delivering levodopa in a more continuous manner. Controlling troublesome dyskinesias once they emerge is usually difficult. As the therapeutic window for levodopa narrows, decreasing levodopa to alleviate dyskinesias often compromises motor function. An attempt can be made to administer smaller levodopa doses more frequently. Amantadine and clozapine can also be considered. Other nondopaminergic drugs have been shown to reduce dyskinesias in small trials, and some are undergoing evaluation. Surgery should be considered for patients with medically intractable motor fluctuations and dyskinesias. STN DBS is the most commonly performed surgical procedure. It improves parkinsonian symptoms, reduces response fluctuations, and allows for significant reductions of levodopa dose, which in turn alleviates dyskinesias.

1.
Olanow W, Schapira AH, Rascol O: Continuous dopamine-receptor stimulation in early Parkinson’s disease. Trends Neurosci 2000;23:S117–S126.
2.
Nutt JG, Obeso JA, Stocchi F: Continuous dopamine-receptor stimulation in advanced Parkinson’s disease. Trends Neurosci 2000;23:S109–S115.
3.
Hauser RA, McDermott MP, Messing S, for the Parkinson Study Group: Factors associated with the development of motor fluctuations and dyskinesias in Parkinson disease. Arch Neurol 2006;63:1756–1760.
4.
Koller WC, Tse W: Unmet medical needs in Parkinson’s disease. Neurology 2004;62:S1–S8.
5.
Thanvi BR, Lo TC: Long term motor complications of levodopa: clinical features, mechanisms, and management strategies. Postgrad Med J 2004;80:452–458.
6.
Chase TN: The significance of continuous dopaminergic stimulation in the treatment of Parkinson’s disease. Drugs 1998;55:1–9.
7.
Chase TN: Levodopa therapy: consequences of the nonphysiologic replacement of dopamine. Neurology 1998;50:S17–S25.
8.
Obeso JA, Rodriguez-Oroz MC, Chana P, Lera G, Rodriguez M, Olanow CW: The evolution and origin of motor complications in Parkinson’s disease. Neurology 2000;55:S13–S20.
9.
Obeso JA, Rodriguez-Oroz MC, Rodriguez M, DeLong MR, Olanow CW: Pathophysiology of levodopa-induced dyskinesias in Parkinson’s disease: problems with the current model. Ann Neurol 2000;47:S22–S32.
10.
Oh JD, Russell DS, Vaughan CL, Chase TN: Enhanced tyrosine phosphorylation of striatal NMDA receptor subunits: effect of dopaminergic denervation and L-DOPA administration. Brain Res 1998;813:150–159.
11.
Schrag A, Quinn N: Dyskinesias and motor fluctuations in Parkinson’s disease: a community-based study. Brain 2000;123:2297–2305.
12.
Grandas F, Galiano ML, Tabernero C: Risk factors for levodopa-induced dyskinesias in Parkinson’s disease. J Neurol 1999;246:1127–1133.
13.
Ahlskog JE, Muenter MD: Frequency of levodopa-related dyskinesias and motor fluctuations as estimated from the cumulative literature. Mov Disord 2001;16:448–458.
14.
Van Gerpen JA, Kumar N, Bower JH, Weigand S, Ahlskog JE: Levodopa-associated dyskinesia risk among Parkinson disease patients in Olmsted County, Minnesota, 1976–1990. Arch Neurol 2006;63:205–209.
15.
Kumar N, Van Gerpen JA, Bower JH, Ahlskog JE: Levodopa-dyskinesia incidence by age of Parkinson’s disease onset. Mov Disord 2005;20:342–344.
16.
Zach M, Friedman A, Slawek J, Derejko M: Quality of life in Polish patients with long-lasting Parkinson’s disease. Mov Disord 2004;19:667–672.
17.
Blanchet PJ, Allard P, Grégoire L, Tardif F, Bédard PJ: Risk factors for peak dose dyskinesia in 100 levodopa-treated parkinsonian patients. Can J Neurol Sci 1996;23:189–193.
18.
The Parkinson Study Group: Levodopa and the progression of Parkinson’s disease. N Engl J Med 2004;351:2498–2508.
19.
Fahn S; the Parkinson Study Group: Does levodopa slow or hasten the rate of progression of Parkinson’s disease? J Neurol 2005;252:IV37–IV42.
20.
Colosimo C, De Michele M: Motor fluctuations in Parkinson’s disease: pathophysiology and treatment. Eur J Neurol 1999;6:1–21.
21.
Bracco F, Battaglia A, Chouza C, Dupont E, Gershanik O, Marti Masso JF, Montastruc JL; PKDS009 Study Group: The long-acting dopamine receptor agonist cabergoline in early Parkinson’s disease: final results of a 5-year, double-blind, levodopa-controlled study. CNS Drugs 2004;18:733–746.
22.
The Parkinson Study Group: Pramipexole versus levodopa as initial treatment for Parkinson disease: a 4-year randomized controlled trial. Arch Neurol 2004;61:1044–1053.
23.
Rascol O, Brooks DJ, Korczyn AD, De Deyn PP, Clarke CE, Lang AE; for the 056 Study Group: A five-year study of the incidence of dyskinesia in patients with early Parkinson’s disease who were treated with ropinirole or levodopa. N Engl J Med 2000;342:1484–1491.
24.
Luquin MR, Scipioni O, Vaamonde J, Gershanik O, Obeso JA: Levodopa-induced dyskinesias in Parkinson’s disease: clinical and pharmacological classification. Mov Disord 1992;7:117–124.
25.
Lieberman A, Olanow CW, Sethi K, Swanson P, Waters CH, Fahn S, Hurtig H, Yahr M: A multicenter trial of ropinirole as adjunct treatment for parkinson’s disease: ropinirole study group. Neurology 1998;51:1057–1062.
26.
Olanow CW, Fahn S, Muenter MD, Klawans H, Hurtig H, Stern MB, Shoulson I, Kurlan R, Grimes JD, Jankovic J, Hoehn M, Markham CH, Duvoisin R, Reinmuth O, Leonard HA, Ahlskog E, Feldman R, Hershey L, Yahr M: A multicenter double-blind placebo-controlled trial of pergolide as an adjunct to Sinemet in Parkinson’s disease. Mov Disord 1994;9:40–47.
27.
Brooks DJ, Sagar H; the UK-Irish Entacapone Study Group: Entacapone is beneficial in both fluctuating and non-fluctuating patients with Parkinson’s disease: a randomised, placebo controlled, double blind, six month study. J Neurol Neurosurg Psychiatry 2003;74:1071–1079.
28.
Olanow CW, Kieburtz K, Stern M, Watts R, Langston JW, Guarnieri M, Hubble J; for the US01 Study Team: Double-blind, placebo-controlled study of entacapone in levodopa-treated patients with stable Parkinson disease. Arch Neurol 2004;61:1563–1568.
29.
Waters CH, Kurth M, Bailey P, Shulman LM, LeWitt P, Dorflinger E, Deptula D, Pedder S: Tolcapone in stable Parkinson’s disease: efficacy and safety of long-term treatment: the Tolcapone Stable Study Group. Neurology 1997;49:665–671.
30.
Quinn N: Drug treatment of Parkinson’s disease. BMJ 1995;310:575–579.
31.
Shoulson I, Oakes D, Fahn S, Lang A, Langston JW, LeWitt P, Olanow CW, Penney JB, Tanner C, Kieburtz K, Rudolph A; The Parkinson Study Group: Impact of sustained deprenyl (selegiline) in levodopa-treated Parkinson’s disease: a randomized placebo-controlled extension of the deprenyl and tocopherol antioxidative therapy of parkinsonism trial. Ann Neurol 2002;51:604–612.
32.
The Parkinson Study Group: A randomized placebo-controlled trial of rasagiline in levodopa-treated patients with Parkinson disease and motor fluctuations: the PRESTO study. Arch Neurol 2005;62:241–248.
33.
Marras C, Lang AE: Measuring motor complications in clinical trials for early Parkinson’s disease. J Neurol Neurosurg Psychiatry 2003;74:143–146.
34.
Movement Disorder Society Task Force on Rating Scales for Parkinson’s Disease: The Unified Parkinson’s Disease Rating Scale (UPDRS): status and recommendations. Mov Disord 2003;18:738–750.
35.
Siderowf A, McDermott M, Kieburtz K, Blindauer K, Plumb S, Shoulson I; the Parkinson Study Group: Test-retest reliability of the unified Parkinson’s disease rating scale in patients with early Parkinson’s disease: results from a multicenter clinical trial. Mov Disord 2002;17:758–763.
36.
Evaluation of dyskinesias in a pilot, randomized, placebo-controlled trial of remacemide in advanced Parkinson disease. Arch Neurol 2001;58:1660–1668.
37.
Guy W: ECDEU Assessment Manual for Psychopharmacology (Abnormal Involuntary Movement Scale [AIMS]). Rockville, National Institute of Mental Health, 1976, pp 534–537.
38.
Stocchi F, Ruggieri S, Vacca L, Olanow CW: Prospective randomized trial of lisuride infusion versus oral levodopa in patients with Parkinson’s disease. Brain 2002;125:2058–2066.
39.
Stocchi F, Vacca L, Ruggieri S, Olanow CW: Intermittent versus continuous levodopa administration in patients with advanced Parkinson disease: a clinical and pharmacokinetic study. Arch Neurol 2005;62:905–910.
40.
Goetz CG, Stebbins GT, Shale HM, Lang AE, Chernik DA, Chmura TA, Ahlskog JE, Dorflinger EE: Utility of an objective dyskinesia rating scale for Parkinson’s disease: inter- and intrarater reliability assessment. Mov Disord 1994;9:390–394.
41.
Hauser RA, Friedlander J, Zesiewicz TA, Adler CH, Seeberger LC, O’Brien CF, Molho ES, Factor SA: A home diary to assess functional status in patients with Parkinson’s disease with motor fluctuations and dyskinesia. Clin Neuropharmacol 2000;23:75–81.
42.
Hauser RA, Deckers F, Lehert P: Parkinson’s disease home diary: further validation and implications for clinical trials. Mov Disord 2004;19:1409–1413.
43.
Karlsen KH, Larsen JP, Tandberg E, Maeland JG: Influence of clinical and demographic variables on quality of life in patients with Parkinson’s disease. J Neurol Neurosurg Psychiatry 1999;66:431–435.
44.
Gage H, Hendricks A, Zhang S, Kazis L: The relative health related quality of life of veterans with Parkinson’s disease. J Neurol Neurosurg Psychiatry 2003;74:163–169.
45.
Schrag A, Selai C, Jahanshahi M, Quinn NP: The EQ-5D – a generic quality of life measure – is a useful instrument to measure quality of life in patients with Parkinson’s disease. J Neurol Neurosurg Psychiatry 2000;69:67–73.
46.
Karlsen KH, Tandberg E, Arsland D, Larsen JP: Health related quality of life in Parkinson’s disease: a prospective longitudinal study. J Neurol Neurosurg Psychiatry 2000;69:584–589.
47.
Pechevis M, Clarke CE, Vieregge P, Khoshnood B, Deschaseaux-Voinet C, Berdeaux G, Ziegler M; on behalf of the Trial Study Group: Effects of dyskinesias in Parkinson’s disease on quality of life and health-related costs: a prospective European study. Eur J Neurol 2005;12:956–963.
48.
Chapuis S, Ouchchane L, Metz O, Gerbaud L, Durif F: Impact of the motor complications of Parkinson’s disease on the quality of life. Mov Disord 2005;20:224–230.
49.
Damiano AM, McGrath MM, Willian MK, Snyder CF, LeWitt PA, Reyes PF, Richter RR, Means ED: Evaluation of a measurement strategy for Parkinson’s disease: assessing patient health-related quality of life. Qual Life Res 2000;9:87–100.
50.
Miyasaki JM, Martin W, Suchowersky O, Weiner WJ, Lang AE: Practice parameter: initiation of treatment for Parkinson’s disease: an evidence-based review: report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology 2002;58:11–17.
51.
Colzi A, Turner K, Lees AJ: Continuous subcutaneous waking day apomorphine in the long term treatment of levodopa induced interdose dyskinesias in Parkinson’s disease. J Neurol Neurosurg Psychiatry 1998;64:573–576.
52.
Ashburn A, Stack E, Pickering RM, Ward CD: A community-dwelling sample of people with Parkinson’s disease: characteristics of fallers and non-fallers. Age Ageing 2001;30:47–52.
53.
Kempster PA, Wahlqvist ML: Dietary factors in the management of Parkinson’s disease. Nutr Rev 1994;52:51–58.
54.
Marras C, Lang A, Krahn M, Tomlinson G, Naglie G; the Parkinson Study Group: Quality of life in early Parkinson’s disease: impact of dyskinesias and motor fluctuations. Mov Disord 2004;19:22–28.
55.
Hely MA, Morris JGL, Reid WGJ, Trafficante R: Sydney Multicenter study of Parkinson’s disease: non-L-dopa-responsive problems dominate at 15 years. Mov Disord 2005;20:190–199.
56.
Hiner BC, Earnhart M: Ropinirole as compared with levodopa in Parkinson’s disease. N Engl J Med 2000;343:884.
57.
Dodel RC, Berger K, Oertel WH: Health-related quality of life and healthcare utilisation in patients with Parkinson’s disease: impact of motor fluctuations and dyskinesias. Pharmacoeconomics 2001;19:1013–1038.
58.
Blanchet PJ, Calon F, Morissette M, Goulet M, Grondin R, Levesque D, Bedard PJ, Di Paolo T: Regulation of dopamine receptors and motor behavior following pulsatile and continuous dopaminergic replacement strategies in the MPTP primate model. Adv Neurol 2001;86:337–344.
59.
Pearce RK, Banerji T, Jenner P, Marsden CD: De novo administration of ropinirole and bromocriptine induces less dyskinesia than L-dopa in the MPTP-treated marmoset. Mov Disord 1998;13:234–241.
60.
Maratos EC, Jackson MJ, Pearce RK, Jenner P: Antiparkinsonian activity and dyskinesia risk of ropinirole and L-DOPA combination therapy in drug naive MPTP-lesioned common marmosets (Callithrix jacchus). Mov Disord 2001;16:631–641.
61.
Parkinson Study Group: Pramipexole versus levodopa as initial treatment for Parkinson disease: a randomized controlled trial. JAMA 2000;284:1931–1938.
62.
Rascol O, Brooks DJ, Korczyn AD, De Deyn PP, Clarke CE, Lang AE, Abdalla M; the 056 Study Group: Development of dyskinesias in a 5-year trial of ropinirole and L-dopa. Mov Disord 2006;21:1844–1850.
63.
Hely MA, Morris JG, Reid WG, O’Sullivan DJ, Williamson PM, Rail D, Broe GA, Margrie S: The Sydney Multicentre Study of Parkinson’s Disease: a randomised, prospective five year study comparing low dose bromocriptine with low dose levodopa-carbidopa. J Neurol Neurosurg Psychiatry 1994;57:903–910.
64.
Lees AJ, Katzenschlager R, Head J, Ben- Shlomo Y; on behalf of the Parkinson’s Disease Research Group of the United Kingdom: Ten-year follow-up of three different initial treatments in de-novo PD: a randomized trial. Neurology 2001;57:1687–1694.
65.
Oertel WH, Wolters E, Sampaio C, Gimenez-Roldan S, Bergamasco B, Dujardin M, Grosset DG, Arnold G, Leenders KL, Hundemer HP, Lledo A, Wood A, Frewer P, Schwarz J: Pergolide versus levodopa monotherapy in early Parkinson’s disease patients: The PELMOPET study. Mov Disord 2006;21:343–353.
66.
Constantinescu R, Romer M, McDermott MP, Kamp C, Kieburtz K. Impact of pramipexole on the onset of levodopa-related dyskinesias. Mov Disord 2007;22:1317–1319.
67.
Nutt JG, Woodward WR, Beckner RM, Stone CK, Berggren K, Carter JH, Gancher ST, Hammerstad JP, Gordin A: Effect of peripheral catechol-O-methyltransferase inhibition on the pharmacokinetics and pharmacodynamics of levodopa in parkinsonian patients. Neurology 1994;44:913–919.
68.
Jenner P: Avoidance of dyskinesia: preclinical evidence for continuous dopaminergic stimulation. Neurology 2004;62:S47–S55.
69.
Marin C, Aguilar E, Obeso JA: Coadministration of entacapone with levodopa attenuates the severity of dyskinesias in hemiparkinsonian rats. Mov Disord 2006;21:646–653.
70.
Koller WC, Hutton JT, Tolosa E, Capilldeo R; the Carbidopa/Levodopa Study Group: Immediate-release and controlled-release carbidopa/levodopa in PD: a 5-year randomized multicenter study. Neurology 1999;53:1012–1019.
71.
Kurth MC, Tetrud JW, Irwin I, Lyness WH, Langston JW: Oral levodopa/carbidopa solution versus tablets in Parkinson’s patients with severe fluctuations: a pilot study. Neurology 1993;43:1036–1039.
72.
Nirenberg MJ, Fahn S: The role of levodopa and catechol-o-methyltransferase inhibitors; in Schapira AHV, Olanow CW (eds): Principles of Treatment in Parkinson’s Disease. Philadelphia, Butterworth Heinemann Elsevier, 2005, pp 3–16.
73.
Obeso JA, Rodriguez-Oroz MC, Zamarbide I: Clinical features, pathophysiology, and management of motor complications in Parkinson’s disease; in Schapira AHV, Olanow CW (eds): Principles of Treatment in Parkinson’s Disease. Philadelphia, Butterworth Heinemann Elsevier, 2005, pp 99–112.
74.
Nyholm D, Askmark H, Gomes-Trolin C, Knutson T, Lennernas H, Nystrom C, Aquilonius SM: Optimizing levodopa pharmacokinetics: intestinal infusion versus oral sustained-release tablets. Clin Neuropharmacol 2003;26:156–163.
75.
Nyholm D, Nilsson Remahl AIM, Dizdar N, Constantinescu R, Holmberg B, Jansson R, Aquilonius S-M, Askmark H: Duodenal levodopa infusion monotherapy versus oral polypharmacy in advanced Parkinson disease. Neurology 2005;64:216–223.
76.
Mouradian MM, Heuser IJ, Baronti F, Chase TN: Modification of central dopaminergic mechanisms by continuous levodopa therapy for advanced Parkinson’s disease. Ann Neurol 1990;27:18–23.
77.
Nilsson D, Hansson LE, Johansson K, Nystrom C, Paalzow L, Aquilonius SM: Long-term intraduodenal infusion of a water based levodopa-carbidopa dispersion in very advanced Parkinson’s disease. Acta Neurol Scand 1998;97:175–183.
78.
Kurth MC, Tetrud JW, Tanner CM, Irwin I, Stebbins GT, Goetz CG, Langston JW: Double-blind, placebo-controlled, crossover study of duodenal infusion of levodopa/carbidopa in Parkinson’s disease patients with ‘on-off’ fluctuations. Neurology 1993;43:1698–1703.
79.
Obeso JA, Grandas F, Vaamonde J, Rosario LM, Martinez-Lage JM: Apomorphine infusion for motor fluctuations in Parkinson’s disease. Lancet 1987;1:1376–1377.
80.
Pollak P, Champay AS, Hommel M, Perret JE, Benabid AL: Subcutaneous apomorphine in Parkinson’s disease. J Neurol Neurosurg Psychiatry 1989;52:544.
81.
Frankel JP, Lees AJ, Kempster PA, Stern GM: Subcutaneous apomorphine in the treatment of Parkinson’s disease. J Neurol Neurosurg Psychiatry 1990;53:96–101.
82.
Poewe W, Kleedorfer B, Wagner M, Bosch S, Schelosky L: Continuous subcutaneous apomorphine infusions for fluctuating Parkinson’s disease: long-term follow-up in 18 patients. Adv Neurol 1993;60:656–659.
83.
Hughes AJ, Bishop S, Kleedorfer B, Turjanski N, Fernandez W, Lees AJ, Stern GM: Subcutaneous apomorphine in Parkinson’s disease: response to chronic administration for up to five years. Mov Disord 1993;8:165–170.
84.
Pietz K, Hagell P, Odin P: Subcutaneous apomorphine in late stage Parkinson’s disease: a long term follow up. J Neurol Neurosurg Psychiatry 1998;65:709–716.
85.
Snow BJ, Macdonald L, Mcauley D, Wallis W: The effect of amantadine on levodopa-induced dyskinesias in Parkinson’s disease: a double-blind, placebo-controlled study. Clin Neuropharmacol 2000;23:82–85.
86.
da Silva-Junior FP, Braga-Neto P, Sueli Monte F, de Bruin VM: Amantadine reduces the duration of levodopa-induced dyskinesia: a randomized, double-blind, placebo-controlled study. Parkinsonism Related Disord 2005;11:449–452.
87.
Thomas A, Iacono D, Luciano AL, Armellino K, Di Iorio A, Onofrj M: Duration of amantadine benefit on dyskinesia of severe Parkinson’s disease. J Neurol Neurosurg Psychiatry 2004;75:141–143.
88.
Paci C, Thomas A, Onofrj M: Amantadine for dyskinesia in patients affected by severe Parkinson’s disease. Neurol Sci 2001;22:75–76.
89.
Del Dotto P, Pavese N, Gambaccini G, Bernardini S, Metman LV, Chase TN, Bonuccelli U: Intravenous amantadine improves levadopa-induced dyskinesias: an acute double-blind placebo-controlled study. Mov Disord 2001;16:515–520.
90.
Luginger E, Wenning GK, Bosch S, Poewe W: Beneficial effects of amantadine on L-dopa-induced dyskinesias in Parkinson’s disease. Mov Disord 2000;15:873–878.
91.
Metman LV, Del Dotto P, LePoole K, Konitsiotis S, Fang J, Chase TN: Amantadine for levodopa-induced dyskinesias: a 1-year follow-up study. Arch Neurol 1999;56:1383–1386.
92.
Verhagen Metman L, Del Dotto P, van den Munckhof P, Fang J, Mouradian MM, Chase TN: Amantadine as treatment for dyskinesias and motor fluctuations in Parkinson’s disease. Neurology 1998;50:1323–1326.
93.
Durif F, Debilly B, Galitzky M, Morand D, Viallet F, Borg M, Thobois S, Broussolle E, Rascol O: Clozapine improves dyskinesias in Parkinson disease: a double-blind, placebo-controlled study. Neurology 2004;62:381–388.
94.
Pierelli F, Adipietro A, Soldati G, Fattapposta F, Pozzessere G, Scoppetta C: Low dosage clozapine effects on L-dopa induced dyskinesias in parkinsonian patients. Acta Neurol Scand 1998;97:295–299.
95.
Pahwa R, Factor SA, Lyons KE, Ondo WG, Gronseth G, Bronte-Stewart H, Hallett M, Miyasaki J, Stevens J, Weiner WJ: Practice parameter: treatment of Parkinson disease with motor fluctuations and dyskinesia (an evidence-based review): report of the Quality Standards Subcommittee of the American Academy of Neurology. Neurology 2006;66:983–995.
96.
Verhagen Metman L, Blanchet PJ, van den Munckhof P, Del Dotto P, Natte R, Chase TN: A trial of dextromethorphan in parkinsonian patients with motor response complications. Mov Disord 1998;13:414–417.
97.
Metman LV, Del Dotto P, Natte R, van den Munckhof P, Chase TN: Dextromethorphan improves levodopa-induced dyskinesias in Parkinson’s disease. Neurology 1998;51:203–206.
98.
Silverdale MA, Nicholson SL, Crossman AR, Brotchie JM: Topiramate reduces levodopa-induced dyskinesia in the MPTP-lesioned marmoset model of Parkinson’s disease. Mov Disord 2005;20:403–409.
99.
Zesiewicz TA, Sullivan KL, Maldonado JL, Tatum WO, Hauser RA: Open-label pilot study of levetiracetam (Keppra) for the treatment of levodopa-induced dyskinesias in Parkinson’s disease. Mov Disord 2005;20:1205–1209.
100.
Bonifati V, Fabrizio E, Cipriani R, Vanacore N, Meco G: Buspirone in levodopa-induced dyskinesias. Clin Neuropharmacol 1994;17:73–82.
101.
Kleedorfer B, Lees AJ, Stern GM: Buspirone in the treatment of levodopa induced dyskinesias. J Neurol Neurosurg Psychiatry 1991;54:376–377.
102.
Durif F, Vidailhet M, Bonnet AM, Blin J, Agid Y: Levodopa-induced dyskinesias are improved by fluoxetine. Neurology 1995;45:1855–1858.
103.
Rascol O, Arnulf I, Peyro-Saint Paul H, Brefel-Courbon C, Vidailhet M, Thalamas C, Bonnet AM, Descombes S, Bejjani B, Fabre N, Montastruc JL, Agid Y: Idazoxan, an alpha-2 antagonist, and L-DOPA-induced dyskinesias in patients with Parkinson’s disease. Mov Disord 2001;16:708–713.
104.
Meco G, Fabrizio E, Di Rezze S, Alessandri A, Pratesi L: Mirtazapine in L-dopa-induced dyskinesias. Clin Neuropharmacol 2003;26:179–181.
105.
Jenner P: Istradefylline, a novel adenosine A2A receptor antagonist, for the treatment of Parkinson’s disease. Exp Opin Invest Drugs 2005;14:729–738.
106.
Hauser RA, Hubble JP, Truong DD, and the Istradefylline US-001 Study Group: Randomized trial of the adenosine A2A receptor antagonist istradefylline in advanced PD. Neurology 2003;61:297–303.
107.
Bara-Jimenez W, Sherzai A, Dimitrova T, Favit A, Bibbiani F, Gillespie M, Morris MJ, Mouradian MM, Chase TN: Adenosine A2A receptor antagonist treatment of Parkinson’s disease. Neurology 2003;61:293–296.
108.
Bara-Jimenez W, Bibbiani F, Morris MJ, Dimitrova T, Sherzai A, Mouradian MM, Chase TN: Effects of serotonin 5-HT1A agonist in advanced Parkinson’s disease. Mov Disord 2005;20:932–936.
109.
Olanow CW, Damier P, Goetz CG, Mueller T, Nutt J, Rascol O, Serbanescu A, Deckers F, Russ H: Multicenter, open-label, trial of sarizotan in Parkinson disease patients with levodopa-induced dyskinesias (the SPLENDID Study). Clin Neuropharmacol 2004;27:58–62.
110.
Kannari K, Kurahashi K, Tomiyama M, Maeda T, Arai A, Baba M, Suda T, Matsunaga M: Tandospirone citrate, a selective 5-HT1A agonist, alleviates L-DOPA-induced dyskinesia in patients with Parkinson’s disease [in Japanese]. No To Shinkei 2002;54:133–137.
111.
DeLong MR: Primate models of movement disorders of basal ganglia origin. Trends Neurosci 1990;13:281–285.
112.
McIntyre CC, Savasta M, Walter BL, Vitek JL: How does deep brain stimulation work? Present understanding and future questions. J Clin Neurophysiol 2004;21:40–50.
113.
Anderson VC, Burchiel KJ, Hogarth P, Favre J, Hammerstad JP: Pallidal versus subthalamic nucleus deep brain stimulation in Parkinson disease. Arch Neurol 2005;62:554–560.
114.
Ford B, Winfield L, Pullman SL, Frucht SJ, Du Y, Greene P, Cheringal JH, Yu Q, Cote LJ, Fahn S, McKhann GM II, Goodman RR: Subthalamic nucleus stimulation in advanced Parkinson’s disease: blinded assessments at one year follow up. J Neurol Neurosurg Psychiatry 2004;75:1255–1259.
115.
Ostergaard K, Sunde N, Dupont E: Effects of bilateral stimulation of the subthalamic nucleus in patients with severe Parkinson’s disease and motor fluctuations. Mov Disord 2002;17:693–700.
116.
Ostergaard K, Aa Sunde N: Evolution of Parkinson’s disease during 4 years of bilateral deep brain stimulation of the subthalamic nucleus. Mov Disord 2006;21:624–631.
117.
Rodriguez-Oroz MC, Zamarbide I, Guridi J, Palmero MR, Obeso JA: Efficacy of deep brain stimulation of the subthalamic nucleus in Parkinson’s disease 4 years after surgery: double blind and open label evaluation. J Neurol Neurosurg Psychiatry 2004;75:1382–1385.
118.
Visser-Vandewalle V, van der Linden C, Temel Y, Celik H, Ackermans L, Spincemaille G, Caemaert J: Long-term effects of bilateral subthalamic nucleus stimulation in advanced Parkinson disease: a four year follow-up study. Parkinsonism Related Disord 2005;11:157–165.
119.
Krack P, Batir A, Van Blercom N, Chabardes S, Fraix V, Ardouin C, Koudsie A, Limousin PD, Benazzouz A, LeBas JF, Benabid A-L, Pollak P: Five-year follow-up of bilateral stimulation of the subthalamic nucleus in advanced Parkinson’s disease. N Engl J Med 2003;349:1925–1934.
120.
Deuschl G, Schade-Brittinger C, Krack P, Volkmann J, Schafer H, Botzel K, Daniels C, Deutschlander A, Dillmann U, Eisner W, Gruber D, Hamel W, Herzog J, Hilker R, Klebe S, Kloss M, Koy J, Krause M, Kupsch A, Lorenz D, Lorenzl S, Mehdorn HM, Moringlane JR, Oertel W, Pinsker MO, Reichmann H, Reuss A, Schneider G-H, Schnitzler A, Steude U, Sturm V, Timmermann L, Tronnier V, Trottenberg T, Wojtecki L, Wolf E, Poewe W, Voges J; for the German Parkinson Study Group, Neurostimulation Section: A randomized trial of deep-brain stimulation for Parkinson’s disease. N Engl J Med 2006;355:896–908.
121.
Moro E, Scerrati M, Romito LM, Roselli R, Tonali P, Albanese A: Chronic subthalamic nucleus stimulation reduces medication requirements in Parkinson’s disease. Neurology 1999;53:85–90.
122.
Vingerhoets FJG, Villemure J-G, Temperli P, Pollo C, Pralong E, Ghika J: Subthalamic DBS replaces levodopa in Parkinson’s disease: two-year follow-up. Neurology 2002;58:396–401.
Copyright / Drug Dosage / Disclaimer
Copyright: All rights reserved. No part of this publication may be translated into other languages, reproduced or utilized in any form or by any means, electronic or mechanical, including photocopying, recording, microcopying, or by any information storage and retrieval system, without permission in writing from the publisher.
Drug Dosage: The authors and the publisher have exerted every effort to ensure that drug selection and dosage set forth in this text are in accord with current recommendations and practice at the time of publication. However, in view of ongoing research, changes in government regulations, and the constant flow of information relating to drug therapy and drug reactions, the reader is urged to check the package insert for each drug for any changes in indications and dosage and for added warnings and precautions. This is particularly important when the recommended agent is a new and/or infrequently employed drug.
Disclaimer: The statements, opinions and data contained in this publication are solely those of the individual authors and contributors and not of the publishers and the editor(s). The appearance of advertisements or/and product references in the publication is not a warranty, endorsement, or approval of the products or services advertised or of their effectiveness, quality or safety. The publisher and the editor(s) disclaim responsibility for any injury to persons or property resulting from any ideas, methods, instructions or products referred to in the content or advertisements.