128
Views
13
CrossRef citations to date
0
Altmetric
Review

Levodopa-induced dyskinesias in Parkinson’s disease: emerging treatments

&
Pages 1605-1617 | Published online: 22 Oct 2013

Abstract

Parkinson’s disease therapy is still focused on the use of L-3,4-dihydroxyphenylalanine (levodopa or L-dopa) for the symptomatic treatment of the main clinical features of the disease, despite intensive pharmacological research in the last few decades. However, regardless of its effectiveness, the long-term use of levodopa causes, in combination with disease progression, the development of motor complications termed levodopa-induced dyskinesias (LIDs). LIDs are the result of profound modifications in the functional organization of the basal ganglia circuitry, possibly related to the chronic and pulsatile stimulation of striatal dopaminergic receptors by levodopa. Hence, for decades the key feature of a potentially effective agent against LIDs has been its ability to ensure more continuous dopaminergic stimulation in the brain. The growing knowledge regarding the pathophysiology of LIDs and the increasing evidence on involvement of nondopaminergic systems raises the possibility of more promising therapeutic approaches in the future. In the current review, we focus on novel therapies for LIDs in Parkinson’s disease, based mainly on agents that interfere with glutamatergic, serotonergic, adenosine, adrenergic, and cholinergic neurotransmission that are currently in testing or clinical development.

Introduction

Parkinson’s disease (PD) is a common neurodegenerative disorder with a wide spectrum of clinical features, including motor symptoms, gait and balance disorders, and cognitive, emotional, and behavioral deficits. The cardinal signs of PD reflect a decline in striatal dopamine (DA) due to the degeneration of neurons arising from the pars compacta of the substantia nigra. The application of levodopa restores DAergic transmission deficiency and provides remarkable symptomatic relief to the vast majority of patients with PD, and remains the most efficacious agent available for PD treatment. However, soon after the first clinical introduction of levodopa as an antiparkinsonian agent by George Cotzias,Citation1 it became apparent that the beneficial effect of levodopa is not permanent, and that its long-term use could cause a behavioral and molecular sensitization such that each exposure to a direct or indirect stimulant of DAergic transmission influences the response to a subsequent stimulus, a procedure known as priming. This narrows the therapeutic window of levodopa, and a variety of motor problems that are extremely disabling for the patient, known as motor-response complications, can occur. One of the most discomforting and frequent features of motor-response complications is the emergence of abnormal and involuntary movements affecting mainly the facial muscles, but also the neck, upper and lower limbs, and body axis, termed levodopa-induced dyskinesia (LID).

Chorea and dystonia are the most frequent forms of LIDs, but ballismus and myoclonus can appear as well. The occurrence of LIDs is directly related to the plasma concentration of levodopa. Three main forms of LIDs have been characterized: 1) “peak-dose” dyskinesias are choreic movements related to high levodopa plasma concentrations; 2) diphasic on/off dyskinesias, which coincide with rising and decreasing plasma concentrations of levodopa and might include both chorea and dystonia; and 3) “off” dystonia, which is an often-painful dystonic posture, appears early in the morning or at night, and characterizes the unmedicated state where plasma levels of levodopa are very low.

Approximately 50% of patients with PD will experience LID roughly 4–5 years after initiation of levodopa treatment.Citation2,Citation3 However, the percentage of PD patients experiencing troublesome LIDs (the ones interfering with normal activities) and requiring some intervention is actually much lower than 50%. The presence of LID significantly worsens the quality of life of the patients.Citation4 Moreover, in order to improve LID or to prevent its occurrence, the dose of levodopa is reduced and additional, less effective medication is added to the therapy. This significantly affects the effective control of parkinsonian symptoms and increases health care costs.Citation5

The pathophysiology of LID

The pathophysiology of LID is still not fully understood. Dyskinetic animals appear to have profound alterations at the pre- and postsynaptic level of the neural network of basal ganglia. Although current notions attribute these alterations to several factors, such as aging-related neurodegeneration, neuronal plasticity of DAergic and non-DAergic systems, and glutamatergic overabundance, it seems that the progressive neuronal loss in substantia nigra and the pulsatile chronic DAergic stimulation from levodopa are the key players not only for the appearance but for the severity of LID as well.

The stimulation of DA receptors in a noncanonical, noncontinuous way due to the short half-life of levodopa is thought to induce secondary modifications in striatal medium spiny neurons.Citation6 These modifications include changes in the intracellular signal-transduction pathways, in the expression of genes, and in the synaptic efficacy of DA and other neurotransmitter receptors. The affected corticostriatal transmission and plasticity sensitize the striatum in a way that expression of LIDs is facilitated. Indeed, dyskinetic rats showed a loss of synaptic depotentiation in response to low-frequency synaptic stimulation in comparison to nondyskinetic rats.Citation7

Despite existing data suggesting the opposite,Citation8,Citation9 a growing body of evidence indicates that progressive DAergic degeneration in substantia nigra lowers the threshold required for LID to occur, thus amplifying the dyskinesiogenic effect of levodopa. Animals with nigrostriatal lesions were at a significantly higher risk of developing LIDs after levodopa administration when compared to less lesioned controls.Citation10 In addition, the percentage of DAergic loss in substantia nigra is positively correlated with the severity of levodopa-induced involuntary movements in a rat model.Citation11 These findings were indirectly confirmed by the finding that in dopamine-responsive dystonia, where in contrast to PD the nigrostriatal terminals are preserved, the chronic administration of levodopa is not related to the development of LID.Citation12 One model that could explain the close relation between nigrostriatal denervation and LID is based on the dysregulation of release and reuptake of DA from the synaptic cleft. In the normal brain as well as in the early phase of PD, where only minor nigrostriatal degeneration is observed, exogenously administered levodopa is incorporated in DAergic neurons, converted to DA, and released via vesicles in the synaptic cleft. The action of DA at the synapse is terminated by its clearance across the presynaptic membrane with the action of the DA-reuptake transporter. With disease progression and further DAergic cell loss, this machinery of DA conversion and regulation is severely compromised, and the availability of DA depends highly on the pharmacokinetic characteristics and the bioavailability of exogenous levodopa, which explains the occurrence of LIDs in relation to levodopa dosage. Moreover, other cells, such as serotonergic neurons, try to compensate for the DAergic loss by converting exogenous levodopa to DA. However, these cells are not equipped with the proper regulatory machinery at the presynaptic level, which results in a defective release and reuptake of synaptic DA, thus leading to an abnormal and irregular postsynaptic stimulation of the striatal DA receptors.Citation13Citation16

Emerging treatments for LID

Treating LID or preventing its appearance is an open challenge in the field of PD treatment. In the last few decades, the consensus that providing continuous DAergic stimulation would modulate the expression of LIDs led to the development of several therapeutic approaches that unfortunately were found to be only partially effective. These approaches were based on the use of DA agonists that reduced the risk of motor complications, but were proved to be less effective regarding the control of parkinsonian symptoms in comparison to levodopa, and most importantly were associated with serious adverse effects. The N-methyl-d-aspartate (NMDA) antagonist amantadine, despite being far from a perfect drug, is the only widely used antidyskinetic agent in everyday clinical practice (see details later in this paper). On the other hand, the introduction of Duodopa and deep-brain stimulation made possible the reduction of daily levodopa dosage and proved to be very beneficial, especially for patients with severe disabling LIDs.Citation17,Citation18 However, both interventions are invasive, restricted to specific selection criteria and certainly not side effect-free.

In recent years, great effort has been made to characterize non-DAergic mechanisms that contribute to LID expression. These include changes in glutamatergic, serotonergic, adenosine, adrenergic, and cholinergic neurotransmission. The pharmacological targeting of compounds related to those systems could reduce or prevent and delay the development of LIDs. Therefore, preclinical and clinical research is currently focused on developing and testing novel agents that influence both DAergic and non-DAergic transmission (for clinical trials see ).

Table 1 Clinical trials for the treatment of LIDs in PD

Glutamate-receptor antagonists

There are ample data suggesting an active involvement of glutamate receptors (GluRs) in the acute expression and development of LID. Although several mechanisms have been suggested, it seems that changes in thalamo- and corticostriatal glutamatergic transmission are of significant importance. The striatum receives massive cortical and thalamic glutamatergic inputs. In PD patients with LID, an increase in NMDA-, metabotropic Glu (mGlu)-, and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-receptor binding have been reported.Citation19 This might explain the loss of spines in medium spiny neurons, since DAergic neurons in the substantia nigra appear to be hypersensitive to glutamatergic action.Citation20 The Glu-mediated neuronal degeneration in turn facilitates the expression of LIDs after levodopa treatment, as mentioned earlier.

NMDA antagonists

Significant changes in the synaptic abundance of NMDA receptors have been found in the striata of dyskinetic animals.Citation21 Moreover, abnormal phosphorylation and synaptic redistribution of several NMDA-subunit receptors seem to play an important role in the expression of dyskinesias, and have been reported in the dyskinetic state.Citation22Citation24 Despite existing data at the preclinical level showing the antidyskinetic effect of several NMDA modulators, the results from clinical trials using NMDA antagonists for the treatment of LID have been so far not that exciting. This is mainly due to the limited therapeutic effect and the potentially serious adverse effects associated with a nonselective blockade. Therefore, no agent of this group, including amantadine, has been approved for the treatment of dyskinesia.

Amantadine, a low-affinity, noncompetitive NMDA-receptor antagonist, proved to be effective in reducing LIDs in preclinical and clinical settings, providing strong evidence for the involvement of the glutamatergic system in the pathophysiology of LIDs. Early animal studies showed that amantadine improved LIDs without significant loss of the levodopa-induced antiparkinsonian benefit.Citation25,Citation26 Later, a significant number of clinical trials confirmed these results in patients with PD.Citation27Citation30 However, other studies raised questions about the duration of the antidyskinetic effect of amantadine, and the results were controversial. Thomas et al reported loss of effect after 9 months,Citation31 whereas other studies reported long-term benefits from amantadine.Citation27,Citation32 Recently, it has been shown that treatment with amantadine prior to the use of levodopa neither delayed onset of LID nor reduced the incidence of LID.Citation33

Currently, there is an ongoing multicenter, randomized, double-blind, placebo-controlled study to evaluate the tolerability and efficacy of each of three dose levels of ADS-5102 oral capsules, an extended-release formulation of amantadine, dosed once daily for the treatment of LID in subjects with PD. The use of ADS-5102 is expected to improve safety and tolerability of amantadine via the stabilization of its plasma concentrations during the day and overnight (NCT01397422).Citation34

Other agents that block NMDA receptors have also been tested in PD patients with motor fluctuations. In randomized clinical studies, remacemide, an uncompetitive NMDA-receptor antagonist, and riluzole, an antiglutamatergic compound that can also inhibit NMDA receptors noncompetitively, showed no antidyskinetic effect,Citation35,Citation36 whereas dextromethorphan reduced dyskinesia by only 30%–40% without affecting the beneficial effect of levodopa.Citation37 Dextromethorphan is rapidly and extensively metabolized by hepatic cytochrome P450 (CYP)-2D6. Therefore, without blocking CYP2D6, plasma dextromethorphan levels in some recipients have been undetectably low.Citation38 A clinical trial to evaluate the efficacy, safety, and tolerability of AVP-923 capsules containing 45 mg dextromethorphan and 10 mg quinidine (a potent CYP2D6 inhibitor) compared to placebo for the treatment of LID in patients with PD is currently recruiting participants (NCT01767129).Citation39

Memantine is also a potent nonselective NMDA inhibitor, and its efficacy against LIDs has been tested in an early study, with no improvement reported.Citation40 However, a recent small double-blind, placebo-controlled pilot study in PD patients with axial symptoms and motor fluctuations showed that memantine slightly reduced LIDs.Citation41 Accordingly, the introduction of memantine to daily medication significantly improved LID in several patients with motor complications, whereas other antidyskinetic drugs failed to do so.Citation42,Citation43 Given the good tolerability and safety of memantine and the aforementioned positive reports, a clinical trial designed to evaluate the efficacy of memantine further for the treatment of LIDs would have been of significant value.

During the last few years, a number of studies have tested selective NMDA antagonists in the treatment of LIDs, though without impressive results so far. The NMDA-subunit NR2B-specific inhibitor CP-101,606 prevented the expression of levodopa-induced motor complication in a rat model of PDCitation44; however, another study using a different animal model failed to report its beneficial antidyskinetic effect.Citation45 CP-101,606 has been tested in a randomized, double-blind, placebo-controlled clinical trial and failed to improve parkinsonism, showing only mild antidyskinetic action. Moreover, tolerability issues have been raised, since dose-related side effects, such as amnesia and dissociation, have been reported.Citation46

Neu-120 is a highly potent and selective uncompetitive NMDA-receptor modulator aiming at reducing LID. It also inhibits monoamine oxidase B (MAO-B) and glycogen synthase kinase-3β activities in vitro, but does not interact with other receptors, transporters, or enzymes. Neu-120 has been tested in several experimental models of PD and a completed Phase I study (unpublished data, http://www.neurim.com/products), and currently a clinical trial is evaluating its efficacy in reducing LID in patients with advanced-phase idiopathic PD and motor-response complication (NCT00607451).Citation47

Metabotropic glutamate-receptor antagonists

mGlu receptors (mGluRs) are found abundantly in the basal ganglia, and they regulate neuronal excitability and synaptic functions. A significant number of preclinical studies have highlighted the involvement of mGluRs in the pathophysiology of LIDs. Pharmacological and neuroimaging studies failed to report an involvement of mGlu2/3 receptors in LID,Citation48,Citation49 and thus the mGluR5 subtype seems to play a key role in the treatment of LIDs.

mGlu5Rs interact strongly with NMDA receptors and amplify their currents. Antagonists of mGluR5 reduce overactivity of NMDA receptors and resulting overexcitability, both important factors in the expression of LIDs. Interestingly, several studies suggested a neuroprotective role of the mGluR5 blockade, possibly by delaying neuronal degeneration.Citation50,Citation51 The results from preclinical studies testing mGluR5 inhibitors against LIDs are exciting.Citation52Citation55 Perhaps the first study to report that mGluR5 antagonists may prove useful for the symptomatic treatment of LID was Dekundy et al. In this study, MTEP (((2-methyl-1,3-thiazol-4-yl)ethynyl) pyridine) alleviated LID (at 2.5 and 5 mg/kg) in 6-hydroxy-DA-lesioned rats.Citation56

mGluR5 antagonists have been recently tested in humans as well. AFQ056, a novel mGluR5 antagonist, reduced LIDs in an animal model without affecting the antiparkinsonian effect of levodopa.Citation57 In the clinical setting, two studies by Berg et al demonstrated that AFQ056 successfully alleviated established LIDs without worsening other parkinsonian features. Patients with moderate-to-severe LID (study 1) and severe LID (study 2) on stable DAergic therapy received 25–150 mg AFQ056 or placebo twice daily for 16 days. Using dyskinesia and involuntary-movement scales, both studies showed that AFQ056-treated patients exhibited a statistically significant clinical improvement in comparison to placebo-treated patients. Mild side effects, such as fatigue, psychiatric and gastrointestinal disorders, and most commonly dizziness, have been reported in both studies. Worsening of dyskinesia associated with stopping treatment was the most common serious adverse effect in both studies.Citation58 Another recent clinical trial tested AFQ056 in several doses versus placebo, and demonstrated that with 200 mg daily a significant antidyskinetic effect without worsening of motor symptoms. However, similar to previous studies, AFQ056 was associated with increased adverse effects, with dizziness, fatigue, hallucination, and dyskinesia being the most common.Citation59 A clinical trial to assess the long-term safety, tolerability, and efficacy of AFQ056 is ongoing (NCT01173731).Citation60

A randomized, double-blind, placebo-controlled clinical study on safety and tolerability of dipraglurant (AX48621), a negative allosteric modulator of mGluR5, was recently published. The secondary objectives of the study included the evaluation of the compound’s efficacy in reducing LID compared with placebo in patients with PD. Dipraglurant showed no adverse effect on levodopa efficacy, increased daily “on” time without dyskinesia, reduced daily “off” time, and improved dyskinesia. No safety or tolerability issues have been raised (NCT01336088).Citation61

AMPA-receptor antagonists

The promising antidyskinetic results from NMDA-receptor antagonism prompted many researchers to investigate other receptors of the glutamatergic system, such as the AMPA receptors, as potential contributors in the expression of LID. More evidence regarding the involvement of AMPA receptors came from early studies in primates, where talampanel, an AMPA-receptor antagonist, enhanced the antiparkinsonian effect and improved LIDs.Citation62 Several clinical studies (NCT00108667,Citation63 NCT00036296,Citation64 NCT00004576Citation65) evaluated the effect of talampanel in PD and LID, but so far no data have been published.

Recently, three clinical studies testing perampanel, a selective non-competitive AMPA-type Glu-receptor antagonist, have been published. All of them reported no significant changes in PD symptoms and dyskinesia in any perampanel group versus placebo.Citation66Citation68 To the best of our knowledge, no further clinical trials testing AMPA-receptor antagonists have been announced.

α2-Adrenergic-receptor antagonists

A significant number of studies have suggested an important role for α- and β-adrenergic receptors in the expression of the dyskinetic phenotype. Most of the studies have been focused on the α2-adrenergic-receptor antagonists idazoxan and fipamezole, and both have been found to be effective in reducing LID when administered in animals.Citation69Citation71

The data in the clinical setting, though, are somehow controversial. Idazoxan showed antidyskinetic properties in patients with LID,Citation72 but the same dosage in another trial proved to be ineffective and caused adverse effects.Citation73 Fipamezole was tested in a double-blind, randomized, placebo-controlled, dose-escalating 28-day study in patients with LID from the US and India, and was found to be effective only in the US subpopulation.Citation74 Two other studies to evaluate mainly safety and tolerability issues, as well as the maximum tolerated dosage in patients with PD who are receiving levodopa, have been conducted (NCT01140841,Citation75 NCT01149811Citation76). To our knowledge, no results have been published so far.

Adenosine A2A-receptor antagonist

Presynaptic A2A adenosine receptors are abundantly expressed in basal ganglia, and can modulate γ-aminobutyric acid (GABA)-ergic synaptic transmission by increasing the excitability of medium spiny neurons in the striatum and by enhancing GABA inhibition in globus pallidus projection neurons. On the other hand, DA in the intact caudate–putamen dampens this activity. Hence, in the DA-depleted caudate–putamen in PD, A2A-receptor antagonists could modulate striatopallidal output balance and alleviate parkinsonian symptoms by preventing the excessive activity of striatopallidal neurons.Citation77,Citation78

Additionally, A2A adenosine receptors participate in the planning and execution of movements, probably by influencing D2- and D1-receptor activity.Citation79 Similarly to Glu, adenosine-receptor activity is increased in the parkinsonian state, and this might inhibit motor activity.Citation80 The A2A adenosine-receptor antagonists modulate (probably via regulation of specific neuropeptides, such as dynorphin and enkephalin) the effects of chronic levodopa administration in synaptic plasticity and contribute possibly to the expression of LID.Citation81

This profound implication, despite the relative lack of solid data on the effectiveness of A2A adenosine receptor antagonists against LID in the preclinical setting, led several researchers to test adenosine A2A-receptor antagonists in patients with PD. Preladenant (Sch 420814), a potent and selective competitive antagonist of the A2A adenosine receptor, showed promising results in mouse models of movement disorders,Citation82,Citation83 but failed to influence LID positively in a Phase II, double-blind, randomized trial where “on” time troublesome or nontroublesome dyskinesia was included as a secondary outcome.Citation84 However, a significant increase in “off ” time has been reported. A subsequent long-term, multicenter, open-label safety and tolerability extension of that study in subjects with fluctuating PD has been recently published. The improvement in “on” and “off” times was sustained, but no antidyskinetic effects have been reported; on the contrary, the preladenant group showed increased dyskinesia rates.Citation85

Istradefylline, another adenosine A2A antagonist, proved to be mildly effective in relieving wearing-off fluctuations in PD patients. However, similarly to preladenant, regarding the troublesome and nontroublesome dyskinesia, the addition of the drug was associated with a slight increase in “on” time with dyskinesia, and dyskinesia was reported as an adverse event more commonly in istradefylline- than in placebo-treated groups.Citation86Citation89 In the most recent study, in 610 patients with PD and motor fluctuations, the primary outcome – reduction in “off ” time – was not met, but 40 mg istradefylline per day significantly improved motor score (Unified PD Rating Scale [UPDRS]). However, as previously mentioned, the most commonly reported drug-related adverse effect was dyskinesia (placebo, 2.5%; 20 mg/day istradefylline, 8.5%; 40 mg/day istradefylline, 6.4%).Citation88

Bara-Jimenez et al suggested in an early study that only after lowering the levodopa dose did istradefylline show a clear antidyskinetic effect,Citation90 yet more data and clinical trials are needed in order to further test this finding.

Recently, a double-blind, randomized, placebo-controlled study of the safety and efficacy of tozadenant (SYN115) in PD patients with “wearing-off” fluctuations, including evaluation of dyskinesia as a secondary outcome measure, was published. Patients in tozadenant-treated groups, especially those who received 120 mg and 180 mg, reported significant improvement in primary outcomes, without any effect on the expression of troublesome dyskinesias in comparison to placebo.Citation91

Nicotinic acetylcholine-receptor agonists

Nicotinic receptors have been also implicated in the expression of LID in PD. Nicotinic receptors colocalize with DA receptors,Citation92 and although the mechanism by which the activation of nicotinic receptors influences the expression of LIDs remains unknown, it is believed that this effect is mediated via regulation of DA release in striatum.Citation93

Nicotine, a nonspecific agonist of nicotinic receptors, is very effective in preventing the occurrence of LIDs and reducing established LIDs in several parkinsonian animal models.Citation94Citation97 However, all the preclinical results come from just one laboratory, and thus should be interpreted with caution. The antidyskinetic efficacy of nicotine has been rather disappointing in clinical settings. Nicotine is poorly tolerated, showing side effects related to the autonomic nervous system, possibly due to its nonspecificity towards critical nicotinic receptors.Citation98 Interestingly, SIB-1508Y, despite being selective for α4β2 nicotinic acetylcholine-receptor agonists, showed very low tolerability as well, and the randomized placebo-controlled study testing its efficacy in patients with PD had to be redesigned.Citation99

Pharmacological studies with nicotinic-receptor agonists have focused on their potential to improve parkinsonian symptoms, but none of them included the antidyskinetic effect as a primary outcome. Improvement in motor scales have been reported by Villafane et al,Citation100 while Vieregge et al, in a randomized double-blind study, suggested that transdermal nicotine as an add-on treatment is not effective for the treatment of PD symptoms.Citation101 Recently, a study where nicotine dehydrate bitartrate was given as an oral capsule instead of a transdermal patch was completed, but to date no data have been published (NCT00957918).Citation102

At the preclinical level, there is growing evidence that the nicotine-mediated improvement in LIDs involves specific receptors’ subunits.Citation103,Citation104 Therefore, in the future, drugs that act more selectively at specific subunits of nicotinic receptors could be an attractive approach for the treatment of LIDs.

Partial dopamine agonists

Partial DA agonists are pharmacological agents able to occupy DA receptors fully (mainly D2 or D3) without producing the maximum pharmacological response, as the full agonists do. This might stabilize the DAergic tone in the DA-depleted network, thus preventing the appearance of motor fluctuations and LID, especially when a mild antiparkinsonian effect is required.

Aripiprazole, a partial agonist of D2 DA and 5-hydroxytryptamine1A (5-HT1A) receptors with known anti-psychotic action, was well tolerated and reduced significantly the intensity and the frequency of LID in PD patients, while other antidyskinetic treatments failed to do so.Citation105 Similarly, preclinical data revealed the antidyskinetic profile of another partial DA D2 agonist, pardoprunox.Citation106 At the clinical level, pardoprunox was assessed regarding UPDRS motor scoreCitation107 as well as “on” time without troublesome dyskinesias,Citation108 and was found to be effective in comparison to placebo. Doses up to 18 mg/day were well tolerated.Citation109

Aplindore has also been tested in patients with PD, but no data have been published yet (NCT00623324).Citation110

Monoamine oxidase-B inhibitors

The pharmacological action of MAO-B inhibitors is the blockade of the enzymatic metabolism of DA in the brain, thus increasing DA concentrations. Based on the idea that an adjunct therapy to levodopa could delay the occurrence of LID, several studies have investigated the effect of MAO-B inhibitors in LID. Waters et al showed that in PD patients who were experiencing motor fluctuations under treatment with levodopa, the time in a dyskinesia-free state was significantly increased in the selegiline patients compared with the placebo group.Citation111 Similarly, rasagiline increased daily “on” time without troublesome dyskinesia when used as an add-on therapy in patients with PD and motor fluctuations.Citation112

Safinamide, a novel MAO-B and Glu-release inhibitor, reduced LID and increased the duration of the antiparkinsonian response of levodopa in parkinsonian animals.Citation113 Several other clinical studies have investigated the effect of safinamide in patients with early PD without motor fluctuation. In a recent placebo-controlled, double-blind trial, a large cohort of patients with mid- to late-stage idiopathic PD with “wearing-off” symptoms and LIDs under optimal treatment received safinamide (50 mg or 100 mg) or placebo as an add-on therapy. In the 50 mg safinamide group, patients showed significant increases in “on” time without worsening of LIDs, whereas in patients who had severe dyskinesia (Dyskinesia Rating Scale >4) and received 100 mg safinamide, both motor fluctuations and LIDs were improved.Citation114

On the other hand, the addition of an MAO-B inhibitor increases the availability of DA at the nigrostriatal synapse, and this could lead to an increased occurrence of LIDs. Shoulson et al tested individuals with PD, initially treated with deprenyl (selegiline) who in the progress of the disease had required levodopa. Continuing deprenyl was associated with increased incidences of dyskinesia in comparison to individuals who changed to a matching placebo under double-blind conditions.Citation115 The meta-analysis performed by Talati et al, which included the Shoulson et al study, confirmed that coadministration of selegiline with levodopa increases the incidence of LID, despite the decreased need for levodopa.Citation116

5-HT agonists

There is growing evidence that implicates the serotonergic system in the pathophysiology of LID;Citation16,Citation117 however, the exact mechanism is still unknown. As mentioned above, it has been suggested that DAergic degeneration causes the raphestriatal serotonergic system to convert exogenous levodopa to DA that is released and acts as a “false neurotransmitter,” which in the absence of autoregulatory mechanisms proper to DAergic transmission determines the expression of LIDs. However, in the clinical setting tandospirone failed to induce a clear antidyskinetic effect, and in addition it may worsen the antiparkinsonian effect of levodopa.Citation118 Similarly, sari-zotan, a compound with full 5-HT1A-agonist properties and additional high affinity for D3 and D4 receptors, despite initial data showing promising antidyskinetic effects,Citation119 failed to improve dyskinesia significantly in two later double-blind, placebo-controlled, multicenter, multinational trials designed to assess its efficacy and safety in PD patients suffering from LIDs.Citation120Citation122

Piclozotan, a 5-HT1A-receptor agonist, significantly reduced levodopa-induced forelimb hyperkinesias and improved motor complications in a rat model of advanced PD. A pilot study with primary outcome measure the “on” time without dyskinesia, testing also adverse effects and pharmacokinetic data of the drug, has been recently completed (NCT00623363).Citation123 No preliminary data are available so far.

Bezard et al tested the effects of eltoprazine, a mixed 5-HT1A/1B-receptor agonist, in LIDs in two experimental animal models of PD. The compound proved to be very effective in reducing LID, and was found to potentiate synergistically the antidyskinetic action of amantadine. However the administration of eltoprazine was associated with a worsening of the parkinsonian symptoms.Citation124 The study was based on previous data showing that a lesion in the serotonergic system or the administration of a 5-HT1A and a 5-HT1B agonist (and especially their coadministration) resulted in a spectacular decrease of LID. These intriguing results found recent confirmation in a clinical trial testing the antidyskinetic action of eltoprazine; however, to date no publication is available. According to the press release of the study, eltoprazine exhibited a statistically significant reduction in LID at the 5 mg dose (P=0.0007) and the 7.5 mg dose (P=0.0467), while no tolerability and safety issues have been raised (http://www.psychogenics.com/pdf/Positive_Effi-cacy_Data_in_Levodopa_Induced_Dyskinesia.pdf).Citation125

Other treatments

A therapeutic class of agents that has been intensively tested for the efficacy against LIDs despite having a very different pharmacological profile is anticonvulsants. One of the very first studies to test the antidyskinetic effects of anticonvulsants came from Price et al, who tested sodium valproate for the treatment of LIDs in a double-blind crossover trial with matched placebo and found no significant effect.Citation126 In the next two decades, several other antiepileptic agents were tested regarding the effectiveness against motor fluctuations in PD. Gabapentin failed to improve LIDs.Citation127 Zonisamide significantly improved PD symptoms, but the positive effect on LID was not consistently documented (improvement only in the 50 mg group, but not in the 25 mg and 100 mg groups versus placebo has been reported).Citation128 Similarly, levetiracetam, despite existing data suggesting the opposite,Citation129 seems to have only mild antidyskinetic effectsCitation130,Citation131 in a wide range of doses. In an MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine)-lesioned marmoset model of PD, treatment with topiramate, a Glu antagonist, significantly improved LID without affecting the antiparkinsonian action of levodopa, thus making topiramate a potentially attractive therapeutic approach for LIDs. However, the only clinical crossover trial to assess the antidyskinetic properties of topiramate in patients with PD and LID was terminated due to slow recruitment, and no data have been published to date (NCT00296959).Citation132 Dopidines are a new pharmacological class of agents that act primarily on DA D2 receptors in a dual way (agonist or antagonist), and therefore they are known as DA stabilizers. Pridopidine, the most widely used compound, was found to be effective as a psychomotor stabilizer, and in clinical studies improved motor function in patients with Huntington’s disease.

Based on unpublished preclinical data and positive Phase I results (https://newsclient.omxgroup.com/cdsPublic/viewDisclosure.action?disclosureId=368187&messageI d=439581), a clinical study has been initiated, with safety and tolerability being the primary outcome and the effect of ACR325 (odopidine) on established LIDs the secondary outcome of the study. The trial has been completed, but no published data are available (NCT01023282).Citation133

Conclusion

The management of LID has proved to be a challenging issue in the field of PD treatment. The appearance of LID is not only an unfortunate emergence in the course of PD, having serious negative effects on the quality of the patient’s life, but also limits the use of levodopa, the most effective agent so far for the symptomatic control of the disease.

Levodopa induces priming actions due to chronic pulsatile DAergic stimulation, and this is the reason that research interest has been focused, in the last few decades, on agents that could provide more continuous DAergic stimulation. However, this strategy appeared to have several limitations, and the control of LID remained an unsolved problem. Recent findings regarding the pathophysiology of LIDs turned scientific interest towards non-DAergic pathways as possible targets for modulating the emergence of LIDs. The results from numerous neuroimaging and pharmacological studies in animals plus postmortem findings were in some cases impressive, and a number of compounds have been further tested in humans. Several studies reported positive results, most notably those using mGluR antagonists and partial DA agonists. However, the majority of the pharmacological agents failed to confirm previously reported safety, tolerability, and effectiveness, a typical example being the antagonists of NMDA and AMPA Glu receptors.

Given the effectiveness (though limited) of amantadine against LID, the positive preclinical data, and the undisputed involvement of NMDA receptors in the pathophysiology of levodopa-induced motor complications, NMDA-receptor antagonists entered the clinical setting as a very promising group of agents for the treatment of LIDs. However, the classical agents that block NMDA receptors in a nonspecific way (remacemide and dextromethorphan) proved to be ineffective in improving LIDs. Similarly, the highly promising results from selected AMPA-receptor antagonists were not confirmed in the clinical setting. The reason for this discrepancy is unknown. Given the structural differences and the degree of complexity between human and rodent brains, one might expect differences in effectiveness and the adverse effects. Moreover, the high prevalence of adverse effects might be associated with the abundance of NMDA receptors in other brain areas, which are not directly involved in motor functions in PD. However, a significant number of clinical trials were based on preclinical data on primates, where this obvious explanation would lose much of its strength. The time the animals were killed during the priming phase (“on” or “off” state) and the lack of a behavioral correlate could also explain discrepancies between clinical data and postmortem findings in animal models.Citation134

NMDA receptors consist of several receptor subunits. Studies in experimental models demonstrated that the expression and development of LIDs are associated with changes in selected NMDA subunits, mostly NR2A and NR2B, with the ratio between these two in the molecular composition of NMDA receptors playing a crucial role in the occurrence of LIDs (reviewed in Huot et al).Citation135 So far, the results from clinical trials using selective NMDA-receptor antagonists, despite positive preclinical data, have been limited and not that exciting. Differences between toxin-induced parkinsonism in animals and idiopathic PD, as well as differences in the doses of the administered agents, could only partially explain this discrepancy. In the future, determining the specific role of each receptor subunit in the establishment of LID could provide additional information concerning its pathophysiology and facilitate the development of an agent with better selectivity and improved safety.

In conclusion, no truly innovative therapies have come to light in recent years, and treatment options against LIDs remain limited. Maybe what is missing are more sophisticated agents capable of acting in a specific way on several pathways involved in the expression of LID, thus causing a synergistic antidyskinetic effect. However, the recent widening of research focus on non-DAergic pathways in itself is an innovative step towards novel therapeutic approaches, which will hopefully soon lead to more fruitful results regarding this disabling effect of levodopa treatment.

Disclosure

No sources of funding were used to assist in the preparation of this review. The authors have no conflicts of interest that are directly relevant to the content of this review.

References

  • CotziasGCL-Dopa for ParkinsonismN Engl J Med1968278116305637779
  • AhlskogJEMuenterMDFrequency of levodopa-related dyskinesias and motor fluctuations as estimated from the cumulative literatureMov Disord200116344845811391738
  • FahnSThe spectrum of levodopa-induced dyskinesiasAnn Neurol2000474 Suppl 1S2S9 discussion S9–S1110762127
  • ChapuisSOuchchaneLMetzOGerbaudLDurifFImpact of the motor complications of Parkinson’s disease on the quality of lifeMov Disord200520222423015384126
  • MaurelFLilliuHLe PenCSocial and economic cost of L-dopa-induced dyskinesias in patients with Parkinson’s diseaseRev Neurol (Paris)20011575507514 French11438770
  • CalabresiPDi FilippoMGhiglieriVTambascoNPicconiBLevodopa-induced dyskinesias in patients with Parkinson’s disease: filling the bench-to-bedside gapLancet Neurol20109111106111720880751
  • PicconiBCentonzeDHåkanssonKLoss of bidirectional striatal synaptic plasticity in L-DOPA-induced dyskinesiaNat Neurosci20036550150612665799
  • GuigoniCDoveroSAubertILevodopa-induced dyskinesia in MPTP-treated macaques is not dependent on the extent and pattern of nigrostrial lesioningEur J Neurosci200522128328716029219
  • LinazasoroGVan BlercomNBergaretxeAIñakiFMLabordaERuiz OrtegaJALevodopa-induced dyskinesias in Parkinson disease are independent of the extent of striatal dopaminergic denervation: a pharmacological and SPECT studyClin Neuropharmacol200932632632919855268
  • Di MonteDAMcCormackAPetzingerGJansonAMQuikMLangstonWJRelationship among nigrostriatal denervation, parkinsonism, and dyskinesias in the MPTP primate modelMov Disord200015345946610830409
  • PailléVBrachetPDamierPRole of nigral lesion in the genesis of dyskinesias in a rat model of Parkinson’s diseaseNeuroreport200415356156415094523
  • de la Fuente-FernándezRDrug-induced motor complications in dopa-responsive dystonia: implications for the pathogenesis of dyskinesias and motor fluctuationsClin Neuropharmacol199922421621910442251
  • CenciMALundbladMPost- versus presynaptic plasticity in L-dopa-induced dyskinesiaJ Neurochem200699238139216942598
  • CartaMCarlssonTKirikDBjörklundADopamine released from 5-HT terminals is the cause of L-dopa-induced dyskinesia in parkinsonian ratsBrain2007130Pt 71819183317452372
  • BlandiniFArmenteroMTNew pharmacological avenues for the treatment of L-dopa-induced dyskinesias in Parkinson’s disease: targeting glutamate and adenosine receptorsExp Opin Investig Drugs2012212153168
  • KannariKYamatoHShenHTomiyamaMSudaTMatsunagaMActivation of 5-HT(1A) but not 5-HT(1B) receptors attenuates an increase in extracellular dopamine derived from exogenously administered L-dopa in the striatum with nigrostriatal denervationJ Neurochem20017651346135311238719
  • AnnicADevosDSeguyDDujardinKDestéeADefebvreLContinuous dopaminergic stimulation by Duodopa in advanced Parkinson’s disease: efficacy and safetyRev Neurol (Paris)20091658–9718727 French19150100
  • DeuschlGSchade-BrittingerCKrackPA randomized trial of deep-brain stimulation for Parkinson’s diseaseN Engl J Med2006355989690816943402
  • SmithYRajuDNandaBPareJFGalvanAWichmannTThe thalamostriatal systems: anatomical and functional organization in normal and parkinsonian statesBrain Res Bull2009782–3606818805468
  • BlandiniFAn update on the potential role of excitotoxicity in the pathogenesis of Parkinson’s diseaseFunct Neurol2010252657120923603
  • HallettPJDunahAWRavenscroftPAlterations of striatal NMDA receptor subunits associated with the development of dyskinesia in the MPTP-lesioned primate model of Parkinson’s diseaseNeuropharmacology200548450351615755478
  • GardoniFPicconiBGhiglieriVA critical interaction between NR2B and MAGUK in L-dopa induced dyskinesiaJ Neurosci200626112914292216540568
  • OhJDRussellDSVaughanCLChaseTNEnhanced tyrosine phosphorylation of striatal NMDA receptor subunits: effect of dopaminergic denervation and L-dopa administrationBrain Res199881311501599824689
  • NapolitanoMPicconiBCentonzeDBernardiGCalabresiPGulinoAL-dopa treatment of parkinsonian rats changes the expression of Src, Lyn and PKC kinasesNeurosci Lett2006398321121416529858
  • BlanchetPJKonitsiotisSChaseTNAmantadine reduces levodopa-induced dyskinesias in parkinsonian monkeysMov Disord19981357988029756148
  • LundbladMUsielloACartaMHåkanssonKFisoneGCenciMAPharmacological validation of a mouse model of l-dopa-induced dyskinesiaExp Neurol20051941667515899244
  • MetmanLVDel DottoPLePooleKKonitsiotisSFangJChaseTNAmantadine for levodopa-induced dyskinesias: a 1-year follow-up studyArch Neurol199956111383138610555659
  • LugingerEWenningGKBöschSPoeweWBeneficial effects of amantadine on L-dopa-induced dyskinesias in Parkinson’s diseaseMov Disord200015587387811009193
  • SnowBJMacdonaldLMcAuleyDWallisWThe effect of amantadine on levodopa-induced dyskinesias in Parkinson’s disease: a double-blind, placebo-controlled studyClin Neuropharmacol2000232828510803797
  • Del DottoPPaveseNGambacciniGIntravenous amantadine improves levadopa-induced dyskinesias: an acute double-blind placebo-controlled studyMov Disord200116351552011391748
  • ThomasAIaconoDLucianoALArmellinoKDi IorioAOnofrjMDuration of amantadine benefit on dyskinesia of severe Parkinson’s diseaseJ Neurol Neurosurg Psychiatry200475114114314707325
  • WolfESeppiKKatzenschlagerRLong-term antidyskinetic efficacy of amantadine in Parkinson’s diseaseMov Disord201025101357136320198649
  • JahangirvandARajputAEarly use of amantadine to prevent or delay onset of levodopainduced dyskinesia in Parkinson’s diseaseMov Disord201328Suppl 1S207
  • PahwaRTannerCMHauserRARandomized trial of extended release amantadine in Parkinson’s disease patients with levodopa-induced dyskinesia (EASED study)Mov Disord201328Suppl 1S158
  • Parkinson Study GroupEvaluation of dyskinesias in a pilot, randomized, placebo-controlled trial of remacemide in advanced Parkinson diseaseArch Neurol200158101660166811594926
  • Bara-JimenezWDimitrovaTDSherzaiAAksuMChaseTNGlutamate release inhibition ineffective in levodopa-induced motor complicationsMov Disord20062191380138316758479
  • Verhagen MetmanLDel DottoPNatteRvan den MunckhofPChaseTNDextromethorphan improves levodopa-induced dyskinesias in Parkinson’s diseaseNeurology19985112032069674803
  • ZhangYBrittoMRValderhaugKLWedlundPJSmithRADextromethorphan: enhancing its systemic availability by way of low-dose quinidine-mediated inhibition of cytochrome P4502D6Clin Pharmacol Ther19925166476551611804
  • Avanir PharmaceuticalsSafety and Efficacy of AVP-923 in the Treatment of Levodopa-induced Dyskinesia in Parkinson’s Disease Patients (LID in PD)ClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2013 [updated July 29, 2013]. Available from: http://clinicaltrials.gov/show/NCT01767129. NLM identifier: NCT01767129Accessed September 6, 2013
  • MerelloMNouzeillesMICammarotaALeiguardaREffect of memantine (NMDA antagonist) on Parkinson’s disease: a double-blind crossover randomized studyClin Neuropharmacol199922527327610516877
  • MoreauCDelvalATiffreauVMemantine for axial signs in Parkinson’s disease: a randomised, double-blind, placebo-controlled pilot studyJ Neurol Neurosurg Psychiatry201384555255523077087
  • VaraneseSHowardJDi RoccoANMDA antagonist memantine improves levodopa-induced dyskinesias and “on-off” phenomena in Parkinson’s diseaseMov Disord201025450851020014061
  • VidalEIFukushimaFBValleAPVillas BoasPJUnexpected improvement in levodopa-induced dyskinesia and on-off phenomena after introduction of memantine for treatment of Parkinson’s disease dementiaJ Am Geriatr Soc201361117017223311565
  • WessellRHAhmedSMMennitiFSDunbarGLChaseTNOhJDNR2B selective NMDA receptor antagonist CP-101,606 prevents levodopa-induced motor response alterations in hemi-parkinsonian ratsNeuropharmacology200447218419415223297
  • NashJERavenscroftPMcGuireSCrossmanARMennitiFSBrotchieJMThe NR2B-selective NMDA receptor antagonist CP-101,606 exacerbates L-dopa-induced dyskinesia and provides mild potentiation of anti-parkinsonian effects of L-dopa in the MPTP-lesioned marmoset model of Parkinson’s diseaseExp Neurol2004188247147915246846
  • NuttJGGunzlerSAKirchhoffTEffects of a NR2B selective NMDA glutamate antagonist, CP-101,606, on dyskinesia and ParkinsonismMov Disord200823131860186618759356
  • Neurim Pharmaceuticals LtdSafety, Tolerability, PK and PD Study of Neu-120 in the Treatment of Levodopa-induced DyskinesiaClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2008 [updated June 9, 2011]. Available from: http://clinicaltrials.gov/ct2/show/NCT00607451?term=NCT00607451&rank=1. NLM identifier: NCT00607451Accessed September 6, 2013
  • PicconiBPisaniACentonzeDStriatal metabotropic glutamate receptor function following experimental parkinsonism and chronic levodopa treatmentBrain2002125Pt 122635264512429591
  • SamadiPRajputACalonFMetabotropic glutamate receptor II in the brains of Parkinsonian patientsJ Neuropathol Exp Neurol200968437438219287314
  • MasilamoniGJBogenpohlJWAlagilleDMetabotropic glutamate receptor 5 antagonist protects dopaminergic and noradrenergic neurons from degeneration in MPTP-treated monkeysBrain2011134Pt 72057207321705423
  • VernonACPalmerSDatlaKPZbarskyVCroucherMJDexterDTNeuroprotective effects of metabotropic glutamate receptor ligands in a 6-hydroxydopamine rodent model of Parkinson’s diseaseEur J Neurosci20052271799180616197521
  • MelaFMartiMDekundyADanyszWMorariMCenciMAAntagonism of metabotropic glutamate receptor type 5 attenuates l-dopa-induced dyskinesia and its molecular and neurochemical correlates in a rat model of Parkinson’s diseaseJ Neurochem2007101248349717359492
  • RylanderDRecchiaAMelaFDekundyADanyszWCenciMAPharmacological modulation of glutamate transmission in a rat model of L-dopa-induced dyskinesia: effects on motor behavior and striatal nuclear signalingJ Pharmacol Exp Ther2009330122723519357321
  • JohnstonTHFoxSHMcIldowieMJPiggottMJBrotchieJMReduction of L-dopa-induced dyskinesia by the selective metabotropic glutamate receptor 5 antagonist 3-[(2-methyl-1,3-thiazol-4-yl)ethynyl] pyridine in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned macaque model of Parkinson’s diseaseJ Pharmacol Exp Ther2010333386587320231306
  • LevandisGBazziniEArmenteroMTNappiGBlandiniFSystemic administration of an mGluR5 antagonist, but not unilateral subthalamic lesion, counteracts l-dopa-induced dyskinesias in a rodent model of Parkinson’s diseaseNeurobiol Dis200829116116817933546
  • DekundyAPietraszekMSchaeferDCenciMADanyszWEffects of group I metabotropic glutamate receptors blockade in experimental models of Parkinson’s diseaseBrain Res Bull200669331832616564428
  • GrégoireLMorinNOuattaraBThe acute antiparkinsonian and antidyskinetic effect of AFQ056, a novel metabotropic glutamate receptor type 5 antagonist, in L-dopa-treated parkinsonian monkeysParkinsonism Relat Disord201117427027621315648
  • BergDGodauJTrenkwalderCAFQ056 treatment of levodopa-induced dyskinesias: results of 2 randomized controlled trialsMov Disord20112671243125021484867
  • StocchiFRascolODesteeAAFQ056 in Parkinson patients with levodopa-induced dyskinesia: 13-week, randomized, dose-finding studyMov Disord EpubJuly122013
  • Novartis PharmaceuticalsOpen Label, Safety, Tolerability and Efficacy of AFQ056 in Parkinson’s Patients With L-dopa Induced DyskinesiasClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2010 [updated December 6, 2012]. Available from: http://clinicaltrials.gov/ct2/show/NCT01173731?term=NCT01173731&rank=1. NLM identifier: NCT01173731Accessed September 6, 2013
  • TisonFDurifFChristopheJSafety, tolerability and anti-dyskinetic efficacy of dipraglurant, a novel mGluR5 negative allosteric modulator (NAM) in Parkinson’s disease (PD) patients with levodopa-induced dyskinesiaPoster presented at: 65th American Academy of Neurology Annual MeetingMarch 16–23, 2013San Diego, CA, USA
  • KonitsiotisSBlanchetPJVerhagenLLamersEChaseTNAMPA receptor blockade improves levodopa-induced dyskinesia in MPTP monkeysNeurology20005481589159510762498
  • National Institute of Neurological Disorders and Stroke (NINDS)Talampanel to Treat Parkinson’s DiseaseClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2005 [updated March 3, 2008]. Available from: http://clinicaltrials.gov/ct2/show/NCT00108667?term=NCT00108667&rank=1. NLM identifier: NCT00108667Accessed September 6, 2013
  • Teva Pharmaceutical IndustriesEffects of Talampanel on Patients With Advanced Parkinson’s DiseaseClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2002 [updated April 11, 2011]. Available from: http://clinicaltrials.gov/ct2/show/NCT00036296?term=NCT00036296&rank=1Accessed September 6, 2013
  • National Institute of Neurological Disorders and Stroke (NINDS)Study of LY300164 for the Treatment of Parkinson’s DiseaseClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2000 [updated March 3, 2008]. Available from: http://clinicaltrials.gov/ct2/show/NCT00004576?term=NCT00004576&rank=1Accessed September 6, 2013
  • EggertKSquillacoteDBaronePSafety and efficacy of perampanel in advanced Parkinson’s disease: a randomized, placebo-controlled studyMov Disord201025789690520461807
  • LeesAFahnSEggertKMPerampanel, an AMPA antagonist, found to have no benefit in reducing “off” time in Parkinson’s diseaseMov Disord201227228428822161845
  • RascolOBaronePBehariMPerampanel in Parkinson disease fluctuations: a double-blind randomized trial with placebo and entacaponeClin Neuropharmacol2012351152022222634
  • SavolaJMHillMEngstromMFipamezole (JP-1730) is a potent alpha2 adrenergic receptor antagonist that reduces levodopa-induced dyskinesia in the MPTP-lesioned primate model of Parkinson’s diseaseMov Disord200318887288312889076
  • HenryBFoxSHPeggsDCrossmanARBrotchieJMThe alpha2-adrenergic receptor antagonist idazoxan reduces dyskinesia and enhances anti-parkinsonian actions of L-dopa in the MPTP-lesioned primate model of Parkinson’s diseaseMov Disord199914574475310495035
  • GrondinRHadj TaharADoanVDLadurePBedardPJNoradrenoceptor antagonism with idazoxan improves L-dopa-induced dyskinesias in MPTP monkeysNaunyn Schmiedebergs Arch Pharmacol2000361218118610685874
  • RascolOArnulfIPeyro-Saint PaulHIdazoxan, an alpha-2 antagonist, and L-dopa-induced dyskinesias in patients with Parkinson’s diseaseMov Disord200116470871311481696
  • MansonAJIakovidouELeesAJIdazoxan is ineffective for levodopa-induced dyskinesias in Parkinson’s diseaseMov Disord200015233633710752589
  • LewittPAHauserRALuMRandomized clinical trial of fipamezole for dyskinesia in Parkinson disease (FJORD study)Neurology201279216316922744665
  • Valeant Pharmaceuticals International, IncA Study of Safety and Tolerability of Fipamezole in Adult Subjects With Parkinson’s Disease Who Are Receiving LevodopaClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2010 [updated August 30, 2011]. Available from: http://clinicaltrials.gov/ct2/show/NCT01140841?term=NCT01140841&rank=1. NLM identifier: NCT01140841Accessed September 6, 2013
  • Valeant Pharmaceuticals International, IncA Study Comparing the Safety and Tolerability of Two Doses of Fipamezole in Adult Patients With Parkinson’s DiseaseClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2010 [updated June 20, 2012]. Available from: http://clinicaltrials.gov/ct2/show/NCT01149811?term=NCT01149811&rank=1. NLM identifier: NCT01149811Accessed September 6, 2013
  • HickeyPStacyMAdenosine A2A antagonists in Parkinson’s disease: what’s next?Curr Neurol Neurosci Rep201212437638522585137
  • MoriAShindouTModulation of GABAergic transmission in the striatopallidal system by adenosine A2A receptors: a potential mechanism for the antiparkinsonian effects of A2A antagonistsNeurology20036111 Suppl 6S44S4814663009
  • MorelliMDi PaoloTWardasJCalonFXiaoDSchwarzschildMARole of adenosine A2A receptors in parkinsonian motor impairment and l-dopa-induced motor complicationsProg Neurobiol200783529330917826884
  • CalonFDridiMHornykiewiczOBedardPJRajputAHDi PaoloTIncreased adenosine A2A receptors in the brain of Parkinson’s disease patients with dyskinesiasBrain2004127Pt 51075108415033896
  • CartaARPinnaACauliOMorelliMDifferential regulation of GAD67, enkephalin and dynorphin mRNAs by chronic-intermittent L-dopa and A2A receptor blockade plus L-dopa in dopamine-denervated ratsSynapse200244316617411954048
  • HodgsonRABertorelliRVartyGBCharacterization of the potent and highly selective A2A receptor antagonists preladenant and SCH 412348 [7-[2-[4–2,4-difluorophenyl]-1-piperazinyl]ethyl]-2-(2-furanyl)-7H-pyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidin-5-amine] in rodent models of movement disorders and depressionJ Pharmacol Exp Ther2009330129430319332567
  • HodgsonRABedardPJVartyGBPreladenant, a selective A(2A) receptor antagonist, is active in primate models of movement disordersExp Neurol2010225238439020655910
  • HauserRACantillonMPourcherEPreladenant in patients with Parkinson’s disease and motor fluctuations: a phase 2, double-blind, randomised trialLancet Neurol201110322122921315654
  • FactorSAWolskiKTogasakiDMLong-term safety and efficacy of preladenant in subjects with fluctuating Parkinson’s diseaseMov Disord201328681782023589371
  • HauserRAShulmanLMTrugmanJMStudy of istradefylline in patients with Parkinson’s disease on levodopa with motor fluctuationsMov Disord200823152177218518831530
  • LeWittPAGuttmanMTetrudJWAdenosine A2A receptor antagonist istradefylline (KW-6002) reduces “off” time in Parkinson’s disease: a double-blind, randomized, multicenter clinical trial (6002-US-005)Ann Neurol200863329530218306243
  • PourcherEFernandezHHStacyMMoriABalleriniRChaikinPIstradefylline for Parkinson’s disease patients experiencing motor fluctuations: results of the KW-6002-US-018 studyParkinsonism Relat Disord201218217818422000279
  • MizunoYHasegawaKKondoTKunoSYamamotoMClinical efficacy of istradefylline (KW-6002) in Parkinson’s disease: a randomized, controlled studyMov Disord201025101437144320629136
  • Bara-JimenezWSherzaiADimitrovaTAdenosine A(2A) receptor antagonist treatment of Parkinson’s diseaseNeurology200361329329612913186
  • HauserRAOlanowCWKieburtzKA phase 2, placebo-controlled, randomized, double-blind trial of tozadenant (SYN-115) in patients with Parkinson’s disease with wearing-off fluctuations on levodopaMov Disord201328Suppl 1S158
  • WonnacottSPresynaptic nicotinic ACh receptorsTrends Neurosci199720292989023878
  • ZhouFMLiangYDaniJAEndogenous nicotinic cholinergic activity regulates dopamine release in the striatumNat Neurosci20014121224122911713470
  • BordiaTCamposCHuangLQuikMContinuous and intermittent nicotine treatment reduces L-3,4-dihydroxyphenylalanine (L-dopa)-induced dyskinesias in a rat model of Parkinson’s diseaseJ Pharmacol Exp Ther2008327123924718650244
  • QuikMCoxHParameswaranNO’LearyKLangstonJWDi MonteDNicotine reduces levodopa-induced dyskinesias in lesioned monkeysAnn Neurol200762658859617960553
  • QuikMMallelaAChinMMcIntoshJMPerezXABordiaTNicotine-mediated improvement in L-dopa-induced dyskinesias in MPTP-lesioned monkeys is dependent on dopamine nerve terminal functionNeurobiol Dis201350304123009753
  • QuikMMallelaALyJZhangDNicotine reduces established levodopa-induced dyskinesias in a monkey model of Parkinson’s diseaseMov Disord EpubJuly82013
  • LemaySChouinardSBlanchetPLack of efficacy of a nicotine transdermal treatment on motor and cognitive deficits in Parkinson’s diseaseProg Neuropsychopharmacol Biol Psychiatry2004281313914687854
  • Parkinson Study GroupRandomized placebo-controlled study of the nicotinic agonist SIB-1508Y in Parkinson diseaseNeurology200666340841016476941
  • VillafaneGCesaroPRiallandAChronic high dose transdermal nicotine in Parkinson’s disease: an open trialEur J Neurol200714121313131617941858
  • ViereggeASiebererMJacobsHHagenahJMViereggePTransdermal nicotine in PD: a randomized, double-blind, placebo-controlled studyNeurology20015761032103511571330
  • Neuraltus Pharmaceuticals IncStudy of NP002 in Subjects With Idiopathic Parkinson’s Disease to Treat Dyskinesias Due to Levodopa TherapyClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2009 [updated September 26, 2011]. Available from: http://clinicaltrials.gov/ct2/show/NCT00957918?term=NCT00957918&rank=1. NLM identifier: NCT00957918Accessed September 6, 2013
  • BordiaTCamposCMcIntoshJMQuikMNicotinic receptor-mediated reduction in L-dopa-induced dyskinesias may occur via desensitizationJ Pharmacol Exp Ther2010333392993820200117
  • HuangLZCamposCLyJIvy CarrollFQuikMNicotinic receptor agonists decrease L-dopa-induced dyskinesias most effectively in partially lesioned parkinsonian ratsNeuropharmacology201160686186821232546
  • MecoGStirpePEditoFAripiprazole in L-dopa-induced dyskinesias: a one-year open-label pilot studyJ Neural Transm2009116788188419533295
  • Tayarani-BinazirKJacksonMJRoseSMcCrearyACJennerPThe partial dopamine agonist pardoprunox (SLV308) administered in combination with l-dopa improves efficacy and decreases dyskinesia in MPTP treated common marmosetsExp Neurol2010226232032720843474
  • BronzovaJSampaioCHauserRADouble-blind study of pardoprunox, a new partial dopamine agonist, in early Parkinson’s diseaseMov Disord201025673874620198713
  • RascolOBronzovaJHauserRAPardoprunox as adjunct therapy to levodopa in patients with Parkinson’s disease experiencing motor fluctuations: results of a double-blind, randomized, placebo-controlled, trialParkinsonism Relat Disord201218437037622316635
  • HauserRABronzovaJSampaioCSafety and tolerability of pardoprunox, a new partial dopamine agonist, in a randomized, controlled study of patients with advanced Parkinson’s diseaseEur Neurol2009621404819407454
  • Ligand PharmaceuticalsThe Effects of Aplindore on the Treatment of Signs and Symptoms of Parkinson’s DiseaseClinicalTrials gov [website on the Internet]Bethesda, MDUS National Library of Medicine2008 [updated September 29, 2011]. Available from: http://clinicaltrials.gov/ct2/show/NCT00623324?term=NCT00623324&rank=1. NLM identifier: NCT00623324Accessed September 6, 2013
  • WatersCHSethiKDHauserRAMolhoEBertoniJMZydis selegiline reduces off time in Parkinson’s disease patients with motor fluctuations: a 3-month, randomized, placebo-controlled studyMov Disord200419442643215077240
  • RascolOBrooksDJMelamedERasagiline as an adjunct to levodopa in patients with Parkinson’s disease and motor fluctuations (LARGO, Lasting Effect in Adjunct Therapy with Rasagiline Given Once Daily, study): a randomised, double-blind, parallel-group trialLancet2005365946394795415766996
  • GrégoireLJourdainVATownsendMRoachADi PaoloTSafinamide reduces dyskinesias and prolongs l-DOPA antiparkinsonian effect in parkinsonian monkeysParkinsonism Relat Disord201319550851423402994
  • BorgohainRSzaszJStanzionePFirst 2-year, controlled study to assess safinamide as add-on to levodopa in Parkinson’s disease with motor fluctuationsMov Disord201126Suppl 2S120
  • ShoulsonIOakesDFahnSImpact of sustained deprenyl (selegiline) in levodopa-treated Parkinson’s disease: a randomized placebo-controlled extension of the deprenyl and tocopherol antioxidative therapy of parkinsonism trialAnn Neurol200251560461212112107
  • TalatiRReinhartKBakerWWhiteCMColemanCIPharmacologic treatment of advanced Parkinson’s disease: a meta-analysis of COMT inhibitors and MAO-B inhibitorsParkinsonism Relat Disord200915750050519167259
  • TomiyamaMKimuraTMaedaTKannariKMatsunagaMBabaMA serotonin 5-HT1A receptor agonist prevents behavioral sensitization to L-dopa in a rodent model of Parkinson’s diseaseNeurosci Res200552218519415893579
  • KannariKKurahashiKTomiyamaMTandospirone citrate, a selective 5-HT1A agonist, alleviates L-DOPA-induced dyskinesia in patients with Parkinson’s diseaseNo To Shinkei200254213313711889759
  • OlanowCWDamierPGoetzCGMulticenter, open-label, trial of sarizotan in Parkinson disease patients with levodopa-induced dyskinesias (the SPLENDID Study)Clin Neuropharmacol2004272586215252265
  • GoetzCGLaskaEHickingCPlacebo influences on dyskinesia in Parkinson’s diseaseMov Disord200823570070718175337
  • RascolODamierPGoetzCGA large phase III study to evaluate the safety and efficacy of sarizotan in the treatment of L-dopa-induced dyskinesia associated with Parkinson’s disease: the Paddy-1 studyMov Disord200621Suppl 15S492S493
  • MüllerTOlanowCWNuttJThe PADDY-2 study: the evaluation of sarizotan for treatment-associated dyskinesia in Parkinson’s disease patientsMov Disord200621Suppl 15S591
  • Asubio Pharmaceuticals IncPreliminary Study of Piclozotan in Patients With Motor Complications Associated With Parkinson’s DiseaseClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2008 [updated April 4, 2011]. Available from: http://clinicaltrials.gov/ct2/show/NCT00623363?term=NCT00623363&rank=1. NLM identifier: NCT00623363Accessed September 6, 2013
  • BezardETronciEPioliEYStudy of the antidyskinetic effect of eltoprazine in animal models of levodopa-induced dyskinesiaMov Disord20132881088109623389842
  • NeuroSearch initiates a clinical study with ACR325 for the treatment of dyskinesias (involuntary movements) in Parkinson’s disease [webpage on the Internet]CopenhagenNeuroSearch A/S2009 Available from https://newsclient.omxgroup.com/cdsPublic/viewDisclosure.action?disclosureId=368187&messageId=439581Accessed September 26, 2013
  • PricePAParkesJDMarsdenCDSodium valproate in the treatment of levodopa-induced dyskinesiaJ Neurol Neurosurg Psychiatry1978418702706355601
  • Van BlercomNLasaAVergerKMasramonXSastreVMLinazasoroGEffects of gabapentin on the motor response to levodopa: a double-blind, placebo-controlled, crossover study in patients with complicated Parkinson diseaseClin Neuropharmacol200427312412815190235
  • MurataMHasegawaKKanazawaIZonisamide improves motor function in Parkinson disease: a randomized, double-blind studyNeurology2007681455017200492
  • WongKKAltyJEGoyAGRaghavSReutensDCKempsterPAA randomized, double-blind, placebo-controlled trial of levetiracetam for dyskinesia in Parkinson’s diseaseMov Disord20112681552155521538524
  • StathisPKonitsiotisSTagarisGPetersonDLevetiracetam for the management of levodopa-induced dyskinesias in Parkinson’s diseaseMov Disord201126226427021412833
  • WolzMLohleMStreckerKLevetiracetam for levodopa-induced dyskinesia in Parkinson’s disease: a randomized, double-blind, placebo-controlled trialJ Neural Transm2010117111279128620803300
  • University Health Network, TorontoTopiramate as a Treatment for Levodopa-Induced Dyskinesia in Parkinson’s DiseaseClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2006 [updated September 19, 2007]. Available from: http://clinicaltrials.gov/ct2/show/NCT00296959?term=NCT00296959&rank=1. NLM identifier: NCT00296959Accessed September 6, 2013
  • NeuroSearch A/SSafety and Tolerability Study Evaluating ACR325 in Parkinson’s Disease PatientsClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2009 [updated November 1, 2011]. Available from: http://clinicaltrials.gov/ct2/show/NCT01023282?term=NCT01023282&rank=1. NLM identifier: NCT01023282Accessed September 6, 2013
  • HuotPBrotchieJM5-HT(1A) receptor stimulation and L-dopa-induced dyskinesia in Parkinson’s disease: bridging the gap between serotonergic and glutamatergic mechanismsExp Neurol2011231219519821819982
  • HuotPJohnstonTHKoprichJBFoxSHBrotchieJMThe pharmacology of L-dopa-induced dyskinesia in Parkinson’s diseasePharmacol Rev201365117122223319549