Skip to main content
Journal of Clinical Sleep Medicine : JCSM : Official Publication of the American Academy of Sleep Medicine logoLink to Journal of Clinical Sleep Medicine : JCSM : Official Publication of the American Academy of Sleep Medicine
. 2023 Jun 1;19(6):1133–1144. doi: 10.5664/jcsm.10450

An overview of the effects of levodopa and dopaminergic agonists on sleep disorders in Parkinson’s disease

Amanda Scanga 1, Anne-Louise Lafontaine 2, Marta Kaminska 3,4,
PMCID: PMC10235717  PMID: 36716191

Abstract

Sleep disorders are among the most common nonmotor symptoms in Parkinson’s disease and are associated with reduced cognition and health-related quality of life. Disturbed sleep can often present in the prodromal or early stages of this neurodegenerative disease, rendering it crucial to manage and treat these symptoms. Levodopa and dopaminergic agonists are frequently prescribed to treat motor symptoms in Parkinson’s disease, and there is increasing interest in how these pharmacological agents affect sleep and their effect on concomitant sleep disturbances and disorders. In this review, we discuss the role of dopamine in regulating the sleep–wake state and the impact of neurodegeneration on sleep. We provide an overview of the effects of levodopa and dopaminergic agonists on sleep architecture, insomnia, excessive daytime sleepiness, sleep-disordered breathing, rapid eye movement sleep behavior disorder, and restless legs syndrome in Parkinson’s disease. Levodopa and dopaminergic drugs may have different effects, beneficial or adverse, depending on dosing, method of administration, and differential effects on the different dopamine receptors. Future research in this area should focus on elucidating the specific mechanisms by which these drugs affect sleep in order to better understand the pathophysiology of sleep disorders in Parkinson’s disease and aid in developing suitable therapies and treatment regimens.

Citation:

Scanga A, Lafontaine A-L, Kaminska M. An overview of the effects of levodopa and dopaminergic agonists on sleep disorders in Parkinson’s disease. J Clin Sleep Med. 2023;19(6):1133–1144.

Keywords: Parkinson’s disease, sleep disorders, levodopa, dopaminergic agonists

INTRODUCTION

Parkinson’s disease (PD) is the second-most-common neurodegenerative disorder1 and is characterized by progressive neurodegeneration. PD is most commonly known for its cardinal motor features resulting from dopamine depletion. However, nonmotor symptoms, including mood disorders, autonomic dysfunction, sensory deficits, and sleep disturbances are now recognized as major contributors to disease symptomatology and continue to gain research interest. Sleep disturbances in PD can manifest as changes in normal sleep architecture, insomnia and excessive daytime sleepiness (EDS), or specific sleep disorders including sleep-disordered breathing (SDB), rapid eye movement sleep behavior disorder (RBD), and restless legs syndrome (RLS).2 Sleep disturbances affect approximately 90% of patients with PD3 and have been linked to cognitive decline.4 Moreover, sleep problems negatively affect health-related quality of life, beginning during the early stages of PD.5 Thus, the management of these symptoms is relevant and necessary in order to reduce disease burden and improve patient well-being. This article reviews the effects of levodopa and dopaminergic agonists (DAs) on sleep disorders in PD and provides an overview of the impact of neurodegeneration on the development of sleep issues in PD.

REVIEW

Sleep disturbances in PD

Sleep disturbances are among the most common nonmotor symptoms.6,7 In PD, sleep disruptions are considered multifactorial and are influenced by factors including motor and nonmotor symptoms, autonomic dysfunction, circadian dysfunction, and iatrogenic insult.8 Sleep disorders commonly occur in the prodromal phase of PD, before the onset of motor symptoms, and tend to progress with advancing stages of PD.2 A primary cause is the progressive neurodegeneration of sleep/wake centers.9

The neuropathological hallmark of PD includes the degeneration of dopaminergic neurons in the substantia nigra pars compacta and dopaminergic depletion of the nigrostriatal pathways.10 Dopamine is a catecholamine involved primarily in arousal and promoting wakefulness, specifically through cortical activation and behavioral arousal.12 The ventral tegmental area and the mesencephalic substantia nigra pars compacta are the major dopaminergic nuclei.13 Dopaminergic cells in the ventral tegmental area and substantia nigra pars compacta have efferent and afferent projections to and from different brain structures involved in the control of sleep and wakefulness, including the dorsal raphe nucleus, the pedunculopontine and laterodorsal tegmental nuclei, the locus coeruleus, the lateral and posterior hypothalamus, the basal forebrain, and the thalamus.14 Dopaminergic neurons do not alter their mean firing rate across sleep–wake states15 but rather are thought to modify their tonic and phasic pattern of discharge in accordance with inputs from serotonergic, cholinergic, orexinergic, glutamatergic, and noradrenergic neurons in the abovementioned brain regions.13

Extranigral structures including in the Braak10 and Sandyk11 are important in the pathology of sleep disruptions in PD. In a postmortem study, PD patients with sleep disturbances exhibited greater pathological changes in specific regions of the brainstem and the hypothalamus than PD patients without sleep disturbances.16 Degeneration in hypothalamic regions such as the ventrolateral preoptic17,18 and median preoptic areas19 may lead to insomnia and sleep disturbances in PD.8 Neurons projecting from these regions innervate and inhibit major monoamine arousal systems leading to the generation and promotion of sleep. Thus, pathological alterations in the hypothalamus can alter sleep–wake states and circadian rhythms and decrease consolidated sleep17 in PD.20

While dopamine is one of many neurotransmitters playing a crucial role in the sleep–wake cycle, sleep disturbances can also result from degeneration of nondopaminergic pathways including serotonergic,21 noradrenergic,22 and cholinergic.23 Additionally, EDS in PD may in part be attributed to a deficiency in hypocretin-containing neurons.24 A loss of hypocretin neurons is involved in hypersomnolence and the pathogenesis of narcolepsy. In some instances, PD patients with EDS can experience sleep attacks, a narcolepsy-like symptom.25 Other specific neurological regions affected in PD include the noradrenergic locus coeruleus and subcoeruleus complex,22 which are involved in sleep–wake state regulation, particularly in promoting arousal and vigilance,26 with relevance to cognition.27 Locus coeruleus neurons are in turn affected by sleep disturbances.2830 In addition, these neurons have been found to be involved in the pathogenesis of RBD. Neuroimaging has shown decreased signal intensity in the abovementioned regions in patients with PD and RBD.31

The brainstem is home to important components of respiratory control, including the respiratory pacemaker and central chemoreceptor.32 Thus, SDB in PD may be attributed to the degeneration of neurons in specific brainstem structures involved in the control of ventilation.33

As per the Braak hypothesis, which suggests that neurodegeneration in PD begins in lower brainstem structures and follows an ascending pathway,15 brainstem regions could be among the first to be affected by neurodegeneration in PD,15 potentially acting as a predisposition to the development of sleep disorders. However, Braak’s hypothesis is likely not the only way by which this neurodegenerative process progresses. A postmortem study by Kalaitzakis et al found that lower brainstem structures were not affected in a subset of PD cases, indicating that different trigger sites and progression patterns of PD exist,34 which could also account for differences in sleep-related manifestations.

Dopamine receptors and sleep regulation in animal models

Much of what is known about the effects of the different dopaminergic receptors in sleep comes from animal data. In rats, administration of D1 and D2 receptor agonists induces wakefulness and reduces slow-wave sleep and rapid eye movement (REM) sleep.14 However, D2 receptors exhibit biphasic effects, such that a reduction in wakefulness and increases in slow-wave sleep and REM sleep have been observed with small doses of D2 receptor agonists, and the opposite has been observed with higher doses.14 Animal data have shown that levodopa and DAs facilitate sleep at lower doses but have the opposite effect at higher doses, through the biphasic action of presynaptic D2 receptors.35,36 Similarly, low doses of the D2 receptor agonist quinpirole decreased wakefulness and increased sleep by acting presynaptically, whereas higher doses promoted wakefulness by acting postsynaptically.37 In primate models, quinpirole was shown to increase sleep latency and wakefulness after sleep onset, exhibiting a deleterious effect on sleep parameters. The DA pramipexole, which has D2 and D3 receptor affinity, also exhibited a biphasic effect in rats with increased sleep at lower doses and greater alertness at higher doses.38 The D1 receptor agonist SKF38393 increased slow-wave sleep, reduced periods of wakefulness after sleep onset, albeit insignificantly, and restored levels of REM sleep to baseline values in a primate model of PD.39 This effect is contrary to what was previously found in other animal models.4042 Thus, further experimentation is required to elucidate the effects of the different dopaminergic receptors, particularly in humans.

Pharmacology of levodopa and DAs

Levodopa is the gold standard in the treatment of PD due to its effectiveness and ability to alleviate the hallmark motor symptoms. Levodopa is decarboxylated in the striatum to form dopamine, which stimulates dopaminergic receptors. This pharmacological agent has a half-life of 50 minutes; however, this can be increased to approximately 90 minutes when levodopa is administered with a decarboxylase inhibitor such as carbidopa.43 The combination of levodopa and carbidopa enables levodopa to cross the blood–brain barrier without being metabolized prematurely by the gastrointestinal system.44 Adverse gastrointestinal effects of levodopa are less frequent with the use of carbidopa.45 Adverse drug reactions occurring with advancing PD include motor fluctuations and symptoms recurring due to the “on–off” phenomenon. After taking levodopa, patients will be in the “on” phase when symptoms are alleviated, but due its short half-life its effects begin to wear off after a few hours and patients experience the “off” phase when symptoms can become unpredictably more severe. Common “on” complications include a delay in the onset of symptomatic relief or a less-powerful relief of symptoms than usual upon intake of medication.46 Levodopa-induced dyskinesia is a common but physiologically complex complication in PD,47 occurring in 3%48 to 94% of PD patients.49 Levodopa-induced dyskinesia occurs when dopamine concentrations are at their maximum in the brain, termed peak-dose dyskinesia, which occurs in a dose-dependent fashion47,50 and also depends on the methods of drug delivery.47

Long-acting levodopa (LALD) has a longer half-life than regular levodopa, maintaining a greater plasma concentration over a greater amount of time.51 LALD exhibits the same safety and tolerability as immediate-release levodopa but also has similar rates of motor complications. Its role as an alternative for immediate-release levodopa remains unclear due to its less-predictable absorption and effect.52

DAs are dopaminergic drugs that can be administered either in monotherapy or with levodopa, depending on the symptomatic profile of the individual patient.53 DAs act directly to continuously stimulate the central dopamine receptors54 in situations of insufficient endogenous dopamine such as PD. DAs can be divided into 2 groups: ergoline- and nonergoline-derived agonists. Common ergoline-class drugs are bromocriptine, cabergoline, pergolide, and lisuride,55 all now rarely prescribed in PD due to their severe adverse complications including cardiovascular,56 retroperitoneal,57 and pleuropulmonary58 fibrosis.54

Currently, the widely used nonergot-derived DAs include pramipexole, ropinirole, apomorphine, and rotigotine. These drugs bind with high affinity to the D2-like dopamine receptor family and modestly interact with the D1-like family.54 In contrast to levodopa, DAs are appreciated for their longer half-life and can be used to reduce motor fluctuations caused by levodopa therapy.55

Pramipexole is among the therapies with the longest half-life, 12 hours.55 It also binds with high affinity to D3 receptors found in the limbic system55 and is often prescribed for unipolar and bipolar depression, making it a treatment option for PD patients with mood disorders.59 Ropinirole is pharmacologically similar in many ways to pramipexole, although it has a shorter half-life of approximately 6 hours and exhibits a preferential affinity for D2 receptors. Adverse effects of both these drugs include orthostatic hypertension, dizziness, nausea, and somnolence.53

Unlike pramipexole and ropinirole, apomorphine and rotigotine are not administered orally. Apomorphine is among the shortest-acting DAs, with a half-life of 30–60 minutes, and is administered subcutaneously. It is used in cases where patients exhibit uncontrollable “off” episodes.55 Rotigotine is primarily a D2/D3 receptor agonist administered in the form of a daily transdermal patch. It has a biphasic half-life beginning with an initial distribution of 3 hours followed by 5 to 7 hours of continuous drug availability. Rotigotine is a valuable option for patients with issues adhering to their daily drug regimen.55 Apomorphine and rotigotine may cause reactions at the site of administration, and other adverse effects include dizziness and orthostatic hypertension.53 Treatment with DAs can also lead to the development of impulse control disorders.60

Sleep architecture and objective sleep quality

In PD, progressive alterations in architectural parameters of sleep structure have been evaluated and confirmed in numerous studies, including changes in sleep efficiency, total sleep time, sleep fragmentation, prolonged sleep latency, and a reduction in the percentage of REM sleep.6163 Architectural sleep changes advance progressively with disease duration,61 indicating that the neuropathological process of PD may play a pivotal role in structural sleep changes. When compared to healthy control patients, drug-naïve patients with PD present a significantly reduced sleep efficiency, a greater sleep latency, and decreased time in REM sleep, supporting the hypothesis that the neurodegenerative process of PD contributes to changes in sleep architecture.64

Conflicting evidence exists regarding the effect of levodopa and DAs on sleep architecture. In their early study, Nausieda et al65 concluded that sleep disruptions were associated with chronic antiparkinsonian therapy because of the increased prevalence of patient-reported sleep disturbances with longer treatment duration.65 Furthermore, the onset of antiparkinsonian treatment in early PD patients has been linked to a statistically significant increase in awakenings and decrease in stage 1 non-REM sleep evidenced by polysomnography (PSG) recordings.66 However, in some studies, PSG data exhibited no significant correlation with levodopa or DA dosage, suggesting that dopaminergic therapy does not result in sleep “destructuring.”61,67 Moreover, Ferreira et al found that in drug-naïve patients started on levodopa for 2 months prior to undergoing a sleep study, sleep efficiency improved from 75.4% to 86.4% (P = .012), with improvements in wakefulness after sleep onset and sleep latency noted as well.64 This was attributed to improved motor symptoms rather than direct changes in sleep architecture. Improvement of sleep fragmentation with dopaminergic treatment was previously shown to be related to normalization of muscle activity in sleep.68 A study assessing the effect of LALD on microstructural sleep parameters found that this preparation of levodopa had no significant effect on objective sleep, that is, LALD did not cause alterations in sleep architecture, but it also did not reverse sleep disturbances or rescue sleep structure.69

In a recent meta-analysis it was found that the levodopa equivalent daily dose (LEDD) contributed significantly to the heterogeneity of PSG changes in PD and that increased LEDD was associated with increased wakefulness after sleep onset and REM latency and decreased total sleep time.70 Self-reported and objective sleep parameters assessed by SCOPA-SLEEP questionnaire and PSG, respectively, worsened with increasing dosage of dopaminergic medication taken within 4 hours of bedtime.71 However, contradictory evidence was reported when the effects of levodopa and DAs were compared separately. While levodopa was not significantly associated with altered sleep macrostructure in a cohort of 351 PD patients, DAs were associated with more awakenings (P < .001) and decreased time spent in REM sleep (P = .012).72 However, rotigotine resulted in improved objective quality of sleep with a 10% increase in sleep efficiency, decreased wakefulness after sleep onset and sleep latency, and increased REM sleep compared to placebo.73 Although improved sleep through improved motor symptom control remains a possibility, the authors suggest a direct effect on D1 receptors,73 which have been shown to be involved in sleep mechanisms in a primate PD model.39

Overall, the effects of levodopa and DAs on sleep architecture appear to be variable in PD, depending on factors including PD disease duration, the dosage of medication used, and the specific pharmacodynamic profile of levodopa formulation or DA. As evidenced experimentally, the dose of levodopa and DAs is critical in both sleep promotion and sleep disruption. Thus, further experiments should focus on determining the optimal dose, formulation, and pharmacologic profile that can maintain consolidated sleep while alleviating motor symptoms.

Subjective sleep quality and insomnia

Insomnia, the most prevalent sleep disorder among PD patients,74 refers to difficulties in sleep initiation or sleep maintenance or early awakenings.75 Subjective sleep quality and insomnia are related. Patients with PD experiencing sleep dysfunction and insomnia report issues primarily regarding sleep maintenance and consolidation.76 Insomnia can be caused by other sleep disorders such as RLS, particularly for sleep-onset insomnia, and obstructive sleep apnea (OSA)77 as well as depression,78 pain, and nocturia. Patients experiencing insomnia often report daytime sleepiness79 that alters their ability to perform daily tasks.

Levodopa may affect subjective sleep quality.67 Increasing doses of levodopa administered chronically over time in PD negatively correlate with subjectively assessed sleep quality67,80,81 and sleep maintenance.67 The motor complications of levodopa can lead to a reduction in subjective sleep quality out of proportion to PSG-measured sleep architecture effects related, for example, to akinesia, cramps, dystonia, and pain related to “off” periods.67 However, LALD administered at bedtime appears to improve nocturnal akinesia and increase total sleep time but has no effect on sleep fragmentation, sleep initiation, and sleep maintenance. It can also reduce rigidity and promote sleep in some patients.52,82 Conversely, in cross-sectional analyses, Schaeffer et al80 found that LALD may not be adequate in reducing nocturnal akinesia and impaired subjective quality of sleep and results in increased perception of nocturnal akinesia.80 Whereas some patients may benefit from LALD to control nocturnal motor symptoms, others may not benefit sufficiently from it to experience better subjective sleep quality.

In a longitudinal study, patients with early PD who reported insomnia had a greater overall LEDD in comparison with those who did not experience sleep disturbances, indicating a potential correlation.74 Insomnia severity assessed in PD using the Insomnia Severity Index83 was not associated with LEDD.84 Therefore, although LEDD may be correlated with the presence of insomnia it may not necessarily correlate with insomnia severity. Moreover, sleep maintenance insomnia, in a cohort of 182 drug-naïve PD patients, increased by 50% from baseline after the introduction of DAs.75 Other investigators have established that treatment with DAs may be an independent risk factor for insomnia.78

Three randomized controlled trials observing the effects of prolonged-release ropinirole,85 rotigotine,86 and pramipexole,87 either together with levodopa or as monotherapy in early PD, reported insomnia as one of the most common adverse events experienced by participants in the groups receiving the intervention. However, a later randomized controlled trial testing rotigotine in a cohort of advanced PD patients reported no difference in reported insomnia between PD patients who received the intervention and those who did not.88 The difference in study population, early vs late PD, might account for different effects. In the RECOVER trial, rotigotine improved nocturnal PD symptoms assessed by the revised Parkinson’s disease sleep scale.89,90 Moreover, other randomized controlled trials using rotigotine and prolonged-release ropinirole showed an improvement in the general quality of self-reported sleep evidenced by significant improvements in Parkinson’s disease sleep scale scores.73,91,92

Insomnia is multifactorial, and PD drug regimens may differentially influence sleep depending on the balance of beneficial vs adverse effects in a given individual. Once motor disturbances are alleviated with levodopa and/or DAs, including throughout the night, patients may be more likely to experience fewer nocturnal disturbances, reducing symptoms of insomnia and ameliorating the perceived quality of sleep. However, motor fluctuations and other adverse effects may lead to poor sleep outcomes. Side effects can also include hallucinations, which are more prevalent in PD patients taking DAs,93 and may aggravate any insomnia experienced. A direct drug effect promoting alertness may also impair sleep.

In treating insomnia, nonpharmacological approaches should be favored, including optimization of sleep hygiene and cognitive behavioral therapy.94 Pharmacological treatment options are limited and associated with potential adverse effects (Table 1). Research to further our understanding of the specific etiologies contributing to the development of poor sleep quality and insomnia in individuals with PD could aid in developing targeted and personalized therapies that will leave patients with more consolidated sleep overall.

Table 1.

Summary of the pharmacological treatment options and effects of levodopa and dopaminergic agonists on sleep disorders in Parkinson’s disease.

Sleep Disorder Pharmacologic Treatments Overall Effect of Levodopa (Better or Worse) Overall Effect of DAs (Better or Worse)
Insomnia
  • Eszopiclone* (off label)94,176

  • Melatonin* (off label)94,176

Better Worse
Excessive daytime sleepiness
  • Modafinil (possibly useful)176

  • Caffeine (off label)176

  • Solriamfetol177

  • Sodium oxybate (off label)178

  • THN102 (investigational)179

Worse Worse
Sleep-disordered breathing
  • No pharmacological treatment recommended

Better (under investigation) No effect
REM sleep behavior disorder Worse Better
Restless legs syndrome
  • Alpha2-delta ligands180: Gabapentin enacarbil, pregabalin, gabapentin

  • Dopaminergic agonists180: Pramipexole, ropinirole, rotigotine

  • Opioids180: Prolonged-release oxycodone-naloxone

Better Better in short term but risk of augmentation (second-line treatment)†
Circadian rhythm disorders
  • Melatonin‡181,182 (off label)

  • Melatonin agonists (investigational)§183

  • REV-ERBα agonists (investigational)¶184

Worse185,186 Worse186

*Drugs with the greatest supporting evidence for treating insomnia in Parkinson’s disease. Other pharmacological agents such as zolpidem, ramelteon, trazodone, and doxepin may be useful but there is insufficient evidence. Safety issues may also arise such as risk of falls. †Doses of dopaminergic agonists should be kept low and other treatments should be considered before additional dopaminergic agonists are prescribed. ‡Melatonin has not been directly assessed as a modulator of the circadian rhythm in Parkinson’s disease. Rather, clinical trials have observed the effects of melatonin on sleep–wake cycle alterations. §Experiments performed in vivo in animal models only and assessed the effects of a melatonin agonist on age-related changes of the circadian rhythm. ¶Experiment performed in vivo in animal models only and assessed the ability of synthetic REV-ERBα agonists to regulate circadian behavior. Das = dopaminergic agonists, REM = rapid eye movement.

EDS

EDS is classified as a state of daytime hypersomnolence where daily, unintentional sleep episodes occur inappropriately.95 A recent meta-analysis found that based on 59 analyzed studies the estimated prevalence of EDS in PD was 35.1%, which represents a higher prevalence in PD than in the general population.96 EDS can pose a threat to the safety of patients, particularly while they are driving.97 The pathophysiology of EDS in PD is multifactorial, and evidence suggests that it can arise because of the nigrostriatal dopamine depletion occurring in the neurodegenerative process of PD,98 coexisting sleep disorders, nocturnal motor symptoms,99,100 and the use of ergot101 and nonergot DAs.102,103

Routine use of DAs in PD resulted in the observation of a severe adverse effect: sleep attacks. Eight PD patients on pramipexole or ropinirole reported falling asleep while driving without feeling any drowsiness or fatigue.104 The same effect of hypersomnolence while driving was reported in 3 other patients with PD on bromocriptine, lisuride, and piribedil, ergot DAs.101 Paus et al105 surveyed 2,952 PD patients regarding the occurrence of sleep attacks or sleep episodes and found that this phenomenon occurred with all DAs available on the market at the time and with levodopa monotherapy. The latter was associated with the lowest risk of developing sleep attacks, whereas the greatest risk was associated with combination therapy of levodopa and DAs. Treatment with DAs was one of the main determining factors of sleep attacks in this cohort.105 However, this finding has been challenged by other investigators arguing that neither pramipexole nor levodopa monotherapy makes patients with PD more susceptible to sleep attacks.106 Amara et al longitudinally assessed EDS in PD patients over 3 years and determined that at years 2 and 3 the total LEDD was significantly higher in PD patients with EDS, setting forth the possibility of a dose-dependent association between EDS and total LEDD.107 Notably, consistent evidence suggests that levodopa monotherapy confers less of a risk of somnolence in comparison with DAs alone and when compared with DA and levodopa combination therapy.108,109 However, levodopa may exert sedative effects contributing to EDS and sleep episodes in patients with PD.101 Other investigators have determined that the dosage of DAs is associated with EDS but the dose of levodopa is not.110 Although there is conflicting evidence regarding levodopa and increased somnolence, the potential sedative effects of levodopa and the sedative effects of DAs indicate that somnolence may occur due to a drug class effect.111 Bliwise et al112 have challenged this idea, however, suggesting that daytime alertness is differentially affected by DAs and levodopa: Increasing doses of DAs resulted in reduced daytime alertness whereas higher doses of levodopa increased daytime alertness, as measured using the Maintenance of Wakefulness Test.113 The authors thus proposed a divergent, dose-dependent effect of drug class on daytime alertness.112 This is different from animal data where higher doses of DA led to greater alertness, as discussed above. Differences may relate to lower dosages per kilogram used in the Bliwise study in humans compared to the animal studies, and possibly patient/PD-related factors.

EDS can also be attributed to the neurodegeneration occurring in PD. EDS is frequent in PD even before treatment initiation103 and associated with longer PD duration.96 Mean sleep latency from the Multiple Sleep Latency Test, an objective sleepiness measure,114 did not differ significantly between the rotigotine group and the placebo group in a randomized controlled trial during a 4-month period,115 suggesting this DA had no effect on EDS. EDS has been found to be related to Dopaminergic Nigrostriatal degeneration in PD, as assessed using dopamine transporter scanning.98 Recently, a positron emission tomography imaging study found reductions in hypothalamic D3 receptor availability to be associated with EDS, irrespective of LEDD.116 This is consistent with previous studies that found daytime sleepiness was not correlated with sleep disorders or with levodopa or DAs but rather appeared to be a consequence of the pathology of PD.117 It has been suggested that PD patients with greater neurodegeneration, such as has been identified in ascending arousal systems, may be more susceptible to the effect of dopaminergic therapy in promoting EDS.116

Insomnia and EDS are both associated with DAs.77 DAs, as well as levodopa, may promote insomnia in several ways. They increase the risk of hallucination, which can cause sleep disturbances.118,119 Motor fluctuation such as “off” periods related to medication can also lead to poor sleep. Moreover, as discussed above, DAs have a biphasic effect on D2 receptors such that higher doses may lead to insomnia whereas lower doses could exert a soporific effect.36,37,78 Both EDS and insomnia in PD can be caused by disturbed sleep from motor or other symptoms, including other sleep disorders, and neurodegeneration. Specific alterations in sleep/wake circuitry may result in different clinical manifestations. For example, hypothalamic D3 receptors appear to be involved in EDS but not insomnia.116 Consequently, the manifestations of insomnia and EDS in a given patient might result from the interaction of the specific neurodegeneration pattern and medication type, dose, and method of delivery.

SDB

SDB in PD can present in different forms, the most prevalent form being OSA, which affects between 20% and 60% of all PD patients.120122 It is suggested that OSA in PD can develop and worsen due to factors such as upper airway motor instability and impaired ventilatory control.32 Whether OSA is more prevalent in PD than in the general population remains a topic of debate. Nevertheless, OSA results in fragmented sleep and intermittent hypoxemia, which can exacerbate symptoms of poor sleep, EDS, and cognitive dysfunction, greatly impairing quality of life.32 In our group’s work and that of others, greater cognitive dysfunction was found in PD patients with OSA and greater cognitive impairment was positively associated with increasing OSA severity.123,124 Furthermore, treatment of OSA in PD using continuous positive airway pressure was found to improve nonmotor symptoms, reduce global cognitive dysfunction125 but not specific cognitive domains,124 and reduce daytime somnolence.122

The effect of levodopa and DAs on SDB in PD has not been well-described. Vincken et al reported a positive effect of levodopa on upper airway obstruction in 1 PD patient. Spirometry was performed on this patient before intake of levodopa, immediately after, and approximately 1 hour later. The flow rate increased substantially after levodopa treatment, in parallel with parkinsonian symptoms and dyspnea improvement. The authors thus showed that upper airway muscles are involved in disorders affecting motor activity, such as PD.126 In a cohort of 21 PD patients, 5 were found to have evidence of upper airway obstruction after levodopa withdrawal. Reintroduction of levodopa resulted in improvement in upper airway obstruction.127 However, airway obstruction may not ameliorate following levodopa treatment and may be due to comorbid chronic obstructive pulmonary disease.128 Additionally, it has been postulated that the DA apomorphine can improve dysfunction of the musculature in the airway, potentially reversing obstruction that could result in OSA.127,129 Interestingly, irregular and tachypneic breathing has been reported as a symptom of levodopa-induced dyskinesia in 2 PD patients. The authors postulated that, in some patients, respiratory dysfunction induced by levodopa may be a result of neurodegeneration of dopaminergic neurons in the respiratory control centers. Dopamine is involved in both the peripheral and central circuitry of respiration. Exogenous dopamine may result in denervation hypersensitivity of peripheral chemoreceptor dopaminergic neurons, causing levodopa-induced respiratory dyskinesia.130 Moreover, the degeneration of dopaminergic neurons in the brainstem that control breathing and compromised dopamine receptor function may also contribute to the development of levodopa-induced respiratory dysfunction.131

Higher LEDD is associated with less-severe OSA.132 A retrospective study conducted by Valko et al133 compared patients with PD with or without SDB. Patients in the group with PD and SDB were divided further into cohorts of central and obstructive SDB predominance. Here, AHI was calculated using overnight PSG, where central apnea was characterized by an absence of respiratory effort after the event and obstructive apnea was characterized by the presence of respiratory effort after the event. Patients with central SDB predominance (n = 7) had a greater overall LEDD than patients with obstructive SDB predominance (n = 50). Moreover, 100% of patients exhibiting central SDB were on dopaminergic treatment, compared to only 56% with obstructive SDB (P = .03). The authors suggested that central SDB predominance may be more likely to occur in patients on both levodopa and DAs; however, further studies with a larger sample size are required to confirm this finding. Additionally, Valko et al speculated that PD patients with central SDB may be predisposed to sleep attacks, considering EDS was highly prevalent in the central SDB group. Interestingly, in patients with PD and SDB treated with DAs, the authors observed a significant decrease in the AHI during REM sleep that occurred exclusively in this group. The authors attributed this lower REM SDB severity to the close interplay between the neurodegenerative processes affecting REM-sleep-related structures in the brainstem and the effect of DAs.133 While important observations were made in this study, the exact effect of DAs and levodopa on SDB in PD remains ambiguous and warrants further investigation, considering the impact of SDB on cognition, among other clinical variables.

Recently, our group found that patients with PD taking LALD before bedtime had a lower incidence of OSA events with a lower AHI and fewer respiratory arousal events compared to patients not taking LALD. These results suggest that LALD might be a potential treatment for OSA in some PD patients. Moreover, DAs were not significant predictors of OSA in this cohort and adjusting statistical models for LEDD did not alter results, suggesting that DAs do not affect OSA in PD.134 A randomized controlled trial assessing the effect of LALD on OSA in PD is ongoing.

RBD

RBD is characterized by the loss of muscle atonia during REM sleep accompanied by enactments of dreams with or without vocalizations,95 which can be bothersome for patients and family members. The incidence of RBD increases throughout disease progression,135 and it is estimated that up to 50% of PD patients will eventually be diagnosed with RBD.136,137 RBD may be a prodromal feature of PD. The loss of dopaminergic midbrain neurons is associated with idiopathic RBD. Consistent with the hypothesis that RBD precedes disorders involving nigrostriatal dopamine depletion such as PD,138 up to 82% of patients diagnosed with idiopathic RBD develop a neurodegenerative syndrome including parkinsonism after 12 years, on average, of follow-up.139

Patients with PD frequently experience psychosis, most commonly in the form of visual hallucinations140—a phenomenon that has been investigated with RBD in PD.141,142 RBD in PD is significantly related to the development of hallucinations. RBD and visual hallucinations likely have overlapping neuropathological mechanisms, including cholinergic dysfunction and degeneration.143 The latency with which psychosis in PD develops depends on differing clinical correlates. In early-onset psychosis, RBD was more frequently present than in patients with late-onset psychosis. Moreover, the development of hallucinations was found to be correlated with the dosage of DA therapy.141 Later onset of psychosis in particular correlated significantly with LEDD. Generally, as disease duration increases and PD progresses, there will be an increase in LEDD. Therefore, this correlation may be influenced more so by disease duration rather than by the actual drug effect.144

LEDD was found to be among the statistically significant clinical correlates of RBD when comparing patients with PD who either did or did not have RBD.145 Longer disease duration was also a significant clinical correlate, further contributing to the likelihood that the association of LEDD and RBD may be confounded by disease duration. A recent preliminary study146 has confirmed the finding that LEDD is associated with RBD symptoms. Meloni et al sought to evaluate how dopamine replacement therapy affects the presence of symptomatic RBD in a cohort of 250 patients with PD. Intriguingly, levodopa therapy was directly and positively associated with RBD severity, as measured with the RBD Screening Questionnaire, after adjusting for age and PD severity.146 Unlike levodopa, DA therapy was not associated with RBD Screening Questionnaire scores.146

The potential prodromal nature of RBD in PD further points toward an association with dopaminergic loss. Patients with idiopathic RBD appear to experience progressive nigrostriatal dopaminergic dysfunction.147 Hence, DAs have been evaluated in the treatment of RBD in patients with and without PD. In 10 patients with RBD only, pramipexole was highly efficacious, with 89% of these patients reporting that RBD symptoms were moderately reduced or completely resolved.148 These results were in line with other findings indicating that clinical manifestations of RBD decreased in both frequency and intensity,149 as well as the severity of RBD symptoms,150 after treatment with pramipexole. However, Kumru et al assessed the effect of pramipexole in a cohort of PD patients undergoing PSG and found that while parkinsonism improved in all patients the frequency and severity of RBD symptoms remained unchanged.151

Rotigotine may be beneficial for RBD symptoms in PD, as reported in an exploratory, open-label study assessing RBD-related symptoms in PD subjectively and objectively using the RBD Questionnaire – Hong Kong152 and PSG, respectively.153 The preliminary findings of this study present an improvement in the severity of RBD symptoms with rotigotine.

The presented studies on DAs in RBD are observational, lack a control group, and involve small sample sizes. Furthermore, studies evaluating idiopathic RBD148150 differ from the study of Kumru et al,151 in which secondary RBD (RBD present with another neurological disorder) was assessed. Idiopathic RBD and secondary RBD may present distinct stages of this disorder, potentially changing response to treatment.154

The potential improvement of RBD symptoms in PD with the administration of rotigotine suggests that a potential relationship exists between dopamine and the pathogenesis of RBD.153 Likewise, Meloni et al speculate that the lack of association between selective D2/D3 receptor agonists and RBD scores implicates the involvement of D1 receptor activation in the underlying pathophysiology of RBD in PD.146 Indeed, REM sleep is modulated by D1 receptor activation,40 a hallmark of rotigotine’s mechanism of action, as confirmed by in vivo modeling.155 Randomized controlled trials evaluating the effects of rotigotine in PD patients with RBD should be employed to confirm these observations,153 and further experimentation is required to clarify these neurological mechanisms.

RLS

RLS is a sleep-related movement disorder characterized by waking dysesthesia and an urge to move the legs that occurs at rest and primarily in the evening and at night, resulting in delayed sleep onset. These symptoms can be relieved by movement.95 They may be accompanied by periodic limb movements during sleep. RLS is commonly diagnosed in PD patients, with its prevalence ranging from 0–52%.156 RLS in PD is typically associated with an older age of onset, a lower rate of family history, and a lower periodic limb movement index on PSG.157

It is postulated that RLS is an early clinical feature of PD; however, recent evidence suggests that symptoms of RLS do not predate a new diagnosis of parkinsonism.158 Moreover, the pathogenesis of RLS in PD likely differs from that of idiopathic RLS.159 Dopamine dysregulation, not dopamine deficiency, may be the predominant mechanism underlying idiopathic RLS,160 differing from the hypodopaminergic state of PD. The lack of association between RLS and periodic limb movements in PD also suggests different pathophysiology in PD.161

Treatment of RLS should initially focus on correction of exacerbating factors, including iron deficiency.162 Pramipexole,163,164 ropinirole,165 and rotigotine166 are all approved for the treatment of RLS167 and have shown considerable efficacy and safety.168 However, long-term use of these drugs often leads to augmentation: the apparent worsening of RLS symptoms related to RLS treatment symptoms.169 Additionally, the risk of impulse control disorders with DAs also remains an issue in RLS treatment. Augmentation also often occurs with levodopa treatment.167 Thus, recent guidelines indicate that alpha2-delta ligands should be the first-line agents used to treat chronic persistent RLS and DAs should only be prescribed if alpha2-delta ligands are contraindicated.170

Maestri et al have reported that extended-release formulations of DAs may resolve the issue of augmentation due to their longer half-life.171 There appears to be an inverse relationship between drug half-life and the development of augmentation. In a cohort of 24 patients with RLS, augmentation was resolved after switching from immediate-release DA treatment to extended-release pramipexole.171 However, further investigation is required to determine if the risk of augmentation is reduced using extended-release DAs, including in PD.

The pharmacological treatment of RLS in PD aligns with that of idiopathic RLS. While RLS in PD can be treated by optimizing and adjusting dopaminergic drug regimens to alleviate symptoms, the same adverse effects exist as in idiopathic RLS.172 Small doses of pramipexole and ropinirole have shown great efficacy and tolerability. In the double-blind, placebo-controlled RECOVER trial, rotigotine showed a significant overall improvement of sleep complaints in PD (assessed by the revised Parkinson’s disease sleep scale), including on specific questions regarding RLS symptoms such as the urge to move arms and legs and restlessness felt in the arms and legs.90 In patients already on dopaminergic therapy, it has been proposed that the schedule of levodopa intake can be altered to optimize its effectiveness in alleviating RLS, with administration of an evening dose.172 In older PD patients with greater cognitive dysfunction, DAs can lead to confusion and hallucinations and, in rarer cases, impulse control issues.172 Levodopa could then be considered over DAs. Nonetheless, to reduce the risk of augmentation and avoid increasing what may be an already high LEDD for some PD patients, it is recommended that other treatment options be thoroughly discussed and considered, namely alpha2-delta ligands.173 Studies have shown that subthalamic nucleus deep brain stimulation can also reduce RLS symptoms in PD,174,175 although this requires further investigation.

CONCLUSIONS

Sleep disturbances and disorders are prevalent nonmotor symptoms warranting safe and efficacious treatments to prevent cognitive decline and improve quality of life. The development of sleep issues in PD is likely multifactorial and influenced by the complex interplay of neurodegeneration, disease progression, and iatrogenic insult. Levodopa and DAs continue to be effective in the treatment of motor symptoms, and preliminary studies suggest they may be beneficial for certain sleep disorders such as OSA and RBD. However, these drugs can pose a risk for developing sleep disturbances including daytime somnolence, a common adverse effect of DAs. Moreover, insomnia may also be an adverse effect associated with DA use. Insomnia can result from a multitude of adverse events in PD such as levodopa-induced dyskinesia and nocturnal motor complications and thus may be likely to improve when motor symptoms are alleviated by treatment. Overall, larger randomized controlled trials and longitudinal studies are necessary to evaluate the effects of levodopa and DAs on sleep and sleep issues in PD. Additional experimental evidence is required to elucidate the specific mechanisms of sleep disorders in PD, and, consequently, develop a greater understanding of how levodopa and dopaminergic drug therapy may influence these neuropathological mechanisms.

ABBREVIATIONS

DA,

dopaminergic agonist

EDS,

excessive daytime sleepiness

LALD,

long-acting levodopa

LEDD,

levodopa equivalent daily dose

OSA,

obstructive sleep apnea

PD,

Parkinson’s disease

PSG,

polysomnography

RBD,

rapid eye movement sleep behavior disorder

REM,

rapid eye movement

RLS,

restless legs syndrome

SDB,

sleep-disordered breathing

DISCLOSURE STATEMENT

All authors have seen and approved the manuscript. Work for this study was performed at the Respiratory Epidemiology and Clinical Research Unit, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada. A.-L.L. reports advisory board honoraria from Abbvie, Sunovion, Paladin Labs, and Merz and speaker honoraria from Sunovion and Paladin Labs. The other authors report no conflicts of interest.

REFERENCES

  • 1. Pringsheim T, Jette N, Frolkis A, Steeves TDL . The prevalence of Parkinson’s disease: a systematic review and meta-analysis . Mov Disord. 2014. ; 29 ( 13 ): 1583 – 1590 . [DOI] [PubMed] [Google Scholar]
  • 2. Stefani A, Högl B . Sleep disorders in Parkinson disease . Sleep Med Clin. 2021. ; 16 ( 2 ): 323 – 334 . [DOI] [PubMed] [Google Scholar]
  • 3. Factor SA, McAlarney T, Sanchez-Ramos JR, Weiner WJ . Sleep disorders and sleep effect in Parkinson’s disease . Mov Disord. 1990. ; 5 ( 4 ): 280 – 285 . [DOI] [PubMed] [Google Scholar]
  • 4. Zhang Y, Zhao JH, Huang DY, et al . Multiple comorbid sleep disorders adversely affect quality of life in Parkinson’s disease patients . NPJ Parkinsons Dis. 2020. ; 6 ( 1 ): 25 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Liguori C, De Franco V, Cerroni R, et al . Sleep problems affect quality of life in Parkinson’s disease along disease progression . Sleep Med. 2021. ; 81 : 307 – 311 . [DOI] [PubMed] [Google Scholar]
  • 6. Kurtis MM, Rodriguez-Blazquez C, Martinez-Martin P ; ELEP Group . Relationship between sleep disorders and other non-motor symptoms in Parkinson’s disease . Parkinsonism Relat Disord. 2013. ; 19 ( 12 ): 1152 – 1155 . [DOI] [PubMed] [Google Scholar]
  • 7. Ragab OA, Elheneedy YA, Bahnasy WS . Non-motor symptoms in newly diagnosed Parkinson’s disease patients . Egypt J Neurol Psychiat Neurosurg. 2019. ; 55 ( 1 ): 24 . [Google Scholar]
  • 8. Albers JA, Chand P, Anch AM . Multifactorial sleep disturbance in Parkinson’s disease . Sleep Med. 2017. ; 35 : 41 – 48 . [DOI] [PubMed] [Google Scholar]
  • 9. Diederich NJ, McIntyre DJ . Sleep disorders in Parkinson’s disease: many causes, few therapeutic options . J Neurol Sci. 2012. ; 314 ( 1-2 ): 12 – 19 . [DOI] [PubMed] [Google Scholar]
  • 10. Braak H, Del Tredici K, Rüb U, de Vos RAI, Jansen Steur ENH, Braak E . Staging of brain pathology related to sporadic Parkinson’s disease . Neurobiol Aging. 2003. ; 24 ( 2 ): 197 – 211 . [DOI] [PubMed] [Google Scholar]
  • 11. Sandyk R, Iacono RP, Bamford CR . The hypothalamus in Parkinson disease . Ital J Neurol Sci. 1987. ; 8 ( 3 ): 227 – 234 . [DOI] [PubMed] [Google Scholar]
  • 12. Jones BE . Arousal and sleep circuits . Neuropsychopharmacology. 2020. ; 45 ( 1 ): 6 – 20 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Dauvilliers Y, Tafti M, Landolt HP . Catechol-O-methyltransferase, dopamine, and sleep-wake regulation . Sleep Med Rev. 2015. ; 22 : 47 – 53 . [DOI] [PubMed] [Google Scholar]
  • 14. Moore RY, Bloom FE . Central catecholamine neuron systems: anatomy and physiology of the dopamine systems . Annu Rev Neurosci. 1978. ; 1 ( 1 ): 129 – 169 . [DOI] [PubMed] [Google Scholar]
  • 15. Monti JM, Jantos H . The roles of dopamine and serotonin, and of their receptors, in regulating sleep and waking . Prog Brain Res. 2008. ; 172 : 625 – 646 . [DOI] [PubMed] [Google Scholar]
  • 16. Kalaitzakis ME, Gentleman SM, Pearce RKB . Disturbed sleep in Parkinson’s disease: anatomical and pathological correlates . Neuropathol Appl Neurobiol. 2013. ; 39 ( 6 ): 644 – 653 . [DOI] [PubMed] [Google Scholar]
  • 17. Lu J, Greco MA, Shiromani P, Saper CB . Effect of lesions of the ventrolateral preoptic nucleus on NREM and REM sleep . J Neurosci. 2000. ; 20 ( 10 ): 3830 – 3842 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Sherin JE, Shiromani PJ, McCarley RW, Saper CB . Activation of ventrolateral preoptic neurons during sleep . Science. 1996. ; 271 ( 5246 ): 216 – 219 . [DOI] [PubMed] [Google Scholar]
  • 19. Gong H, McGinty D, Guzman-Marin R, Chew KT, Stewart D, Szymusiak R . Activation of c-fos in GABAergic neurones in the preoptic area during sleep and in response to sleep deprivation . J Physiol. 2004. ; 556 ( Pt 3 ): 935 – 946 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Videnovic A . Management of sleep disorders in Parkinson’s disease and multiple system atrophy . Mov Disord. 2017. ; 32 ( 5 ): 659 – 668 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Wilson H, Giordano B, Turkheimer FE, Chaudhuri KR, Politis M . Serotonergic dysregulation is linked to sleep problems in Parkinson’s disease . Neuroimage Clin. 2018. ; 18 : 630 – 637 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Doppler CEJ, Smit JAM, Hommelsen M, et al . Microsleep disturbances are associated with noradrenergic dysfunction in Parkinson’s disease . Sleep. 2021. ; 44 ( 8 ): zsab040 . [DOI] [PubMed] [Google Scholar]
  • 23. Kotagal V, Albin RL, Müller MLTM, et al . Symptoms of rapid eye movement sleep behavior disorder are associated with cholinergic denervation in Parkinson disease . Ann Neurol. 2012. ; 71 ( 4 ): 560 – 568 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Wienecke M, Werth E, Poryazova R, et al . Progressive dopamine and hypocretin deficiencies in Parkinson’s disease: is there an impact on sleep and wakefulness? J Sleep Res. 2012. ; 21 ( 6 ): 710 – 717 . [DOI] [PubMed] [Google Scholar]
  • 25. Thannickal TC, Lai YY, Siegel JM . Hypocretin (orexin) cell loss in Parkinson’s disease . Brain. 2007. ; 130 ( Pt 6 ): 1586 – 1595 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Aston-Jones G, Chiang C, Alexinsky T . Chapter 35 - Discharge of noradrenergic locus coeruleus neurons in behaving rats and monkeys suggests a role in vigilance . Prog Brain Res. 1991. ; 88 : 501 – 520 . [DOI] [PubMed] [Google Scholar]
  • 27. Aston-Jones G, Bloom FE . Activity of norepinephrine-containing locus coeruleus neurons in behaving rats anticipates fluctuations in the sleep-waking cycle . J Neurosci. 1981. ; 1 ( 8 ): 876 – 886 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Zhu Y, Fenik P, Zhan G, et al . Selective loss of catecholaminergic wake active neurons in a murine sleep apnea model . J Neurosci. 2007. ; 27 ( 37 ): 10060 – 10071 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Zhang J, Zhu Y, Zhan G, et al . Extended wakefulness: compromised metabolics in and degeneration of locus ceruleus neurons . J Neurosci. 2014. ; 34 ( 12 ): 4418 – 4431 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Zhu Y, Fenik P, Zhan G, Somach R, Xin R, Veasey S . Intermittent short sleep results in lasting sleep wake disturbances and degeneration of locus coeruleus and orexinergic neurons . Sleep. 2016. ; 39 ( 8 ): 1601 – 1611 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. García-Lorenzo D, Longo-Dos Santos C, Ewenczyk C, et al . The coeruleus/subcoeruleus complex in rapid eye movement sleep behaviour disorders in Parkinson’s disease . Brain. 2013. ; 136 ( Pt 7 ): 2120 – 2129 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Kaminska M, Lafontaine AL, Kimoff RJ . The interaction between obstructive sleep apnea and Parkinson’s disease: possible mechanisms and implications for cognitive function . Parkinsons Dis. 2015. ; 2015 : 849472 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Lajoie AC, Lafontaine AL, Kaminska M . The spectrum of sleep disorders in Parkinson disease: a review . Chest. 2021. ; 159 ( 2 ): 818 – 827 . [DOI] [PubMed] [Google Scholar]
  • 34. Kalaitzakis ME, Graeber MB, Gentleman SM, Pearce RKB . The dorsal motor nucleus of the vagus is not an obligatory trigger site of Parkinson’s disease: a critical analysis of α-synuclein staging . Neuropathol Appl Neurobiol. 2008. ; 34 ( 3 ): 284 – 295 . [DOI] [PubMed] [Google Scholar]
  • 35. Laloux C, Derambure P, Houdayer E, et al . Effect of dopaminergic substances on sleep/wakefulness in saline- and MPTP-treated mice . J Sleep Res. 2008. ; 17 ( 1 ): 101 – 110 . [DOI] [PubMed] [Google Scholar]
  • 36. Monti JM, Hawkins M, Jantos H, D’Angelo L, Fernández M . Biphasic effects of dopamine D-2 receptor agonists on sleep and wakefulness in the rat . Psychopharmacology (Berl). 1988. ; 95 ( 3 ): 395 – 400 . [DOI] [PubMed] [Google Scholar]
  • 37. Monti JM, Jantos H, Fernández M . Effects of the selective dopamine D-2 receptor agonist, quinpirole on sleep and wakefulness in the rat . Eur J Pharmacol. 1989. ; 169 ( 1 ): 61 – 66 . [DOI] [PubMed] [Google Scholar]
  • 38. Lagos P, Scorza C, Monti JM, et al . Effects of the D3 preferring dopamine agonist pramipexole on sleep and waking, locomotor activity and striatal dopamine release in rats . Eur Neuropsychopharmacol. 1998. ; 8 ( 2 ): 113 – 120 . [DOI] [PubMed] [Google Scholar]
  • 39. Hyacinthe C, Barraud Q, Tison F, Bezard E, Ghorayeb I . D1 receptor agonist improves sleep-wake parameters in experimental parkinsonism . Neurobiol Dis. 2014. ; 63 : 20 – 24 . [DOI] [PubMed] [Google Scholar]
  • 40. Trampus M, Ferri N, Monopoli A, Ongini E . The dopamine D1 receptor is involved in the regulation of REM sleep in the rat . Eur J Pharmacol. 1991. ; 194 ( 2–3 ): 189 – 194 . [DOI] [PubMed] [Google Scholar]
  • 41. Trampus M, Ferri N, Adami M, Ongini E . The dopamine D1 receptor agonists, A68930 and SKF 38393, induce arousal and suppress REM sleep in the rat . Eur J Pharmacol. 1993. ; 235 ( 1 ): 83 – 87 . [DOI] [PubMed] [Google Scholar]
  • 42. Monti JM, Fernández M, Jantos H . Sleep during acute dopamine D1 agonist SKF 38393 or D1 antagonist SCH 23390 administration in rats . Neuropsychopharmacology. 1990. ; 3 ( 3 ): 153 – 162 . [PubMed] [Google Scholar]
  • 43. National Center for Biotechnology Information . PubChem Compound Summary for CID 6047, Levodopa. https://pubchem.ncbi.nlm.nih.gov/compound/Levodopa . Accessed on February 4, 2022.
  • 44. Whitfield AC, Moore BT, Daniels RN . Classics in chemical neuroscience: levodopa . ACS Chem Neurosci. 2014. ; 5 ( 12 ): 1192 – 1197 . [DOI] [PubMed] [Google Scholar]
  • 45. Markham C, Diamond SG, Treciokas LJ . Carbidopa in Parkinson disease and in nausea and vomiting of levodopa . Arch Neurol. 1974. ; 31 ( 2 ): 128 – 133 . [DOI] [PubMed] [Google Scholar]
  • 46. Aradi SD, Hauser RA . Medical management and prevention of motor complications in Parkinson’s disease . Neurotherapeutics. 2020. ; 17 ( 4 ): 1339 – 1365 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Espay AJ, Morgante F, Merola A, et al . Levodopa-induced dyskinesia in Parkinson disease: current and evolving concepts . Ann Neurol. 2018. ; 84 ( 6 ): 797 – 811 . [DOI] [PubMed] [Google Scholar]
  • 48. Fahn S, Oakes D, Shoulson I, et al. Parkinson Study Group . Levodopa and the progression of Parkinson’s disease . N Engl J Med. 2004. ; 351 ( 24 ): 2498 – 2508 . [DOI] [PubMed] [Google Scholar]
  • 49. 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 ( 2 ): 190 – 199 . [DOI] [PubMed] [Google Scholar]
  • 50. Tran TN, Vo TNN, Frei K, Truong DD . Levodopa-induced dyskinesia: clinical features, incidence, and risk factors . J Neural Transm (Vienna). 2018. ; 125 ( 8 ): 1109 – 1117 . [DOI] [PubMed] [Google Scholar]
  • 51. Hammerstad JP, Woodward WR, Nutt JG, Gancher ST, Block GA, Cyhan G . Controlled release levodopa/carbidopa 25/100 (Sinemet CR 25/100): pharmacokinetics and clinical efficacy in untreated parkinsonian patients . Clin Neuropharmacol. 1994. ; 17 ( 5 ): 429 – 434 . [DOI] [PubMed] [Google Scholar]
  • 52. Fabbrini G, Di Stasio F, Bloise M, Berardelli A . Soluble and controlled-release preparations of levodopa: do we really need them? J Neurol. 2010. ; 257 ( Suppl 2 ): S292 – S297 . [DOI] [PubMed] [Google Scholar]
  • 53. Armstrong MJ, Okun MS . Diagnosis and treatment of Parkinson disease: a review . JAMA. 2020. ; 323 ( 6 ): 548 – 560 . [DOI] [PubMed] [Google Scholar]
  • 54. Blandini F, Armentero MT . Dopamine receptor agonists for Parkinson’s disease . Expert Opin Investig Drugs. 2014. ; 23 ( 3 ): 387 – 410 . [DOI] [PubMed] [Google Scholar]
  • 55. Borovac JA . Side effects of a dopamine agonist therapy for Parkinson’s disease: a mini-review of clinical pharmacology . Yale J Biol Med. 2016. ; 89 ( 1 ): 37 – 47 . [PMC free article] [PubMed] [Google Scholar]
  • 56. Antonini A, Poewe W . Fibrotic heart-valve reactions to dopamine-agonist treatment in Parkinson’s disease . Lancet Neurol. 2007. ; 6 ( 9 ): 826 – 829 . [DOI] [PubMed] [Google Scholar]
  • 57. Brasselet D, Chouchana L, Vial T, Damin-Pernik M, Lebrun-Vignes B . Drug-induced retroperitoneal fibrosis: a case/non-case study in the French PharmacoVigilance Database . Expert Opin Drug Saf. 2020. ; 19 ( 7 ): 903 – 914 . [DOI] [PubMed] [Google Scholar]
  • 58. Tintner R, Manian P, Gauthier P, Jankovic J . Pleuropulmonary fibrosis after long-term treatment with the dopamine agonist pergolide for Parkinson Disease . Arch Neurol. 2005. ; 62 ( 8 ): 1290 – 1295 . [DOI] [PubMed] [Google Scholar]
  • 59. Tundo A, de Filippis R, De Crescenzo F . Pramipexole in the treatment of unipolar and bipolar depression. A systematic review and meta-analysis . Acta Psychiatr Scand. 2019. ; 140 ( 2 ): 116 – 125 . [DOI] [PubMed] [Google Scholar]
  • 60. Grall-Bronnec M, Victorri-Vigneau C, Donnio Y, et al . Dopamine agonists and impulse control disorders: a complex association . Drug Saf. 2018. ; 41 ( 1 ): 19 – 75 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Diederich NJ, Vaillant M, Mancuso G, Lyen P, Tiete J . Progressive sleep ‘destructuring’ in Parkinson’s disease. A polysomnographic study in 46 patients . Sleep Med. 2005. ; 6 ( 4 ): 313 – 318 . [DOI] [PubMed] [Google Scholar]
  • 62. Martinez-Ramirez D, De Jesus S, Walz R, et al . A polysomnographic study of Parkinson’s disease sleep architecture . Parkinsons Dis. 2015. ; 2015 : 570375 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Yong MH, Fook-Chong S, Pavanni R, Lim LL, Tan EK . Case control polysomnographic studies of sleep disorders in Parkinson’s disease . PLoS One. 2011. ; 6 ( 7 ): e22511 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Ferreira T, Prabhakar S, Kharbanda PS . Sleep disturbances in drug naïve Parkinson’s disease (PD) patients and effect of levodopa on sleep . Ann Indian Acad Neurol. 2014. ; 17 ( 4 ): 416 – 419 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Nausieda PA, Weiner WJ, Kaplan LR, Weber S, Klawans HL . Sleep disruption in the course of chronic levodopa therapy: an early feature of the levodopa psychosis . Clin Neuropharmacol. 1982. ; 5 ( 2 ): 183 – 194 . [DOI] [PubMed] [Google Scholar]
  • 66. Brunner H, Wetter TC, Hogl B, Yassouridis A, Trenkwalder C, Friess E . Microstructure of the non-rapid eye movement sleep electroencephalogram in patients with newly diagnosed Parkinson’s disease: effects of dopaminergic treatment . Mov Disord. 2002. ; 17 ( 5 ): 928 – 933 . [DOI] [PubMed] [Google Scholar]
  • 67. Antczak JM, Rakowicz MJ, Banach M, et al . Negative influence of L-dopa on subjectively assessed sleep but not on nocturnal polysomnography in Parkinson’s disease . Pharmacol Rep. 2013. ; 65 ( 3 ): 614 – 623 . [DOI] [PubMed] [Google Scholar]
  • 68. Askenasy JJ, Yahr MD . Reversal of sleep disturbance in Parkinson’s disease by antiparkinsonian therapy: a preliminary study . Neurology. 1985. ; 35 ( 4 ): 527 – 532 . [DOI] [PubMed] [Google Scholar]
  • 69. Wailke S, Herzog J, Witt K, Deuschl G, Volkmann J . Effect of controlled-release levodopa on the microstructure of sleep in Parkinson’s disease . Eur J Neurol. 2011. ; 18 ( 4 ): 590 – 596 . [DOI] [PubMed] [Google Scholar]
  • 70. Zhang Y, Ren R, Sanford LD, et al . Sleep in Parkinson’s disease: a systematic review and meta-analysis of polysomnographic findings . Sleep Med Rev. 2020. ; 51 : 101281 . [DOI] [PubMed] [Google Scholar]
  • 71. Chahine LM, Daley J, Horn S, et al . Association between dopaminergic medications and nocturnal sleep in early-stage Parkinson’s disease . Parkinsonism Relat Disord. 2013. ; 19 ( 10 ): 859 – 863 . [DOI] [PubMed] [Google Scholar]
  • 72. Sixel-Döring F, Trautmann E, Mollenhauer B, Trenkwalder C . Age, drugs, or disease: what alters the macrostructure of sleep in Parkinson’s disease? Sleep Med. 2012. ; 13 ( 9 ): 1178 – 1183 . [DOI] [PubMed] [Google Scholar]
  • 73. Pierantozzi M, Placidi F, Liguori C, et al . Rotigotine may improve sleep architecture in Parkinson’s disease: a double-blind, randomized, placebo-controlled polysomnographic study . Sleep Med. 2016. ; 21 : 140 – 144 . [DOI] [PubMed] [Google Scholar]
  • 74. Xu Z, Anderson KN, Saffari SE, et al . Progression of sleep disturbances in Parkinson’s disease: a 5-year longitudinal study . J Neurol. 2021. ; 268 ( 1 ): 312 – 320 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75. Tholfsen LK, Larsen JP, Schulz J, Tysnes OB, Gjerstad MD . Changes in insomnia subtypes in early Parkinson disease . Neurology. 2017. ; 88 ( 4 ): 352 – 358 . [DOI] [PubMed] [Google Scholar]
  • 76. Ratti PL, Nègre-Pagès L, Pérez-Lloret S, et al . Subjective sleep dysfunction and insomnia symptoms in Parkinson’s disease: insights from a cross-sectional evaluation of the French CoPark cohort . Parkinsonism Relat Disord. 2015. ; 21 ( 11 ): 1323 – 1329 . [DOI] [PubMed] [Google Scholar]
  • 77. Chahine LM, Amara AW, Videnovic A . A systematic review of the literature on disorders of sleep and wakefulness in Parkinson’s disease from 2005 to 2015 . Sleep Med Rev. 2017. ; 35 : 33 – 50 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Zhu K, van Hilten JJ, Marinus J . The course of insomnia in Parkinson’s disease . Parkinsonism Relat Disord. 2016. ; 33 : 51 – 57 . [DOI] [PubMed] [Google Scholar]
  • 79. Mizrahi-Kliger AD, Feldmann LK, Kühn AA, Bergman H . Etiologies of insomnia in Parkinson’s disease - lessons from human studies and animal models . Exp Neurol. 2022. ; 350 : 113976 . [DOI] [PubMed] [Google Scholar]
  • 80. Schaeffer E, Vaterrodt T, Zaunbrecher L, et al . Effects of Levodopa on quality of sleep and nocturnal movements in Parkinson’s Disease . J Neurol. 2021. ; 268 ( 7 ): 2506 – 2514 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. Sweet RD, McDowell FH . Five years’ treatment of Parkinson’s disease with levodopa . Ann Intern Med. 1975. ; 83 ( 4 ): 456 – 463 . [DOI] [PubMed] [Google Scholar]
  • 82. Stocchi F, Barbato L, Nordera G, Berardelli A, Ruggieri S . Sleep disorders in Parkinson’s disease . J Neurol. 1998. ; 245 ( Suppl 1 ): S15 – S18 . [DOI] [PubMed] [Google Scholar]
  • 83. Bastien CH, Vallières A, Morin CM . Validation of the Insomnia Severity Index as an outcome measure for insomnia research . Sleep Med. 2001. ; 2 ( 4 ): 297 – 307 . [DOI] [PubMed] [Google Scholar]
  • 84. Chung S, Bohnen NI, Albin RL, Frey KA, Müller ML, Chervin RD . Insomnia and sleepiness in Parkinson disease: associations with symptoms and comorbidities . J Clin Sleep Med. 2013. ; 9 ( 11 ): 1131 – 1137 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Watts RL, Lyons KE, Pahwa R, et al. 228 Study Investigators . Onset of dyskinesia with adjunct ropinirole prolonged-release or additional levodopa in early Parkinson’s disease . Mov Disord. 2010. ; 25 ( 7 ): 858 – 866 . [DOI] [PubMed] [Google Scholar]
  • 86. Parkinson Study Group . A controlled trial of rotigotine monotherapy in early Parkinson’s disease . Arch Neurol. 2003. ; 60 ( 12 ): 1721 – 1728 . [DOI] [PubMed] [Google Scholar]
  • 87. Parkinson Study Group . Pramipexole in levodopa-treated Parkinson disease patients of African, Asian, and Hispanic heritage . Clin Neuropharmacol. 2007. ; 30 ( 2 ): 72 – 85 . [DOI] [PubMed] [Google Scholar]
  • 88. Nicholas AP, Borgohain R, Chaná P, et al. SP921 Study Investigators . A randomized study of rotigotine dose response on ‘off’ time in advanced Parkinson’s disease . J Parkinsons Dis. 2014. ; 4 ( 3 ): 361 – 373 . [DOI] [PubMed] [Google Scholar]
  • 89. Trenkwalder C, Kohnen R, Högl B, et al . Parkinson’s disease sleep scale--validation of the revised version PDSS-2 . Mov Disord. 2011. ; 26 ( 4 ): 644 – 652 . [DOI] [PubMed] [Google Scholar]
  • 90. Trenkwalder C, Kies B, Rudzinska M, et al. Recover Study Group . Rotigotine effects on early morning motor function and sleep in Parkinson’s disease: a double-blind, randomized, placebo-controlled study (RECOVER) . Mov Disord. 2011. ; 26 ( 1 ): 90 – 99 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Pagonabarraga J, Piñol G, Cardozo A, et al . Transdermal rotigotine improves sleep fragmentation in Parkinson’s disease: results of the multicenter, prospective SLEEP-FRAM Study . Parkinsons Dis. 2015. ; 2015 : 131508 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Ray Chaudhuri K, Martinez-Martin P, Rolfe KA, et al . Improvements in nocturnal symptoms with ropinirole prolonged release in patients with advanced Parkinson’s disease . Eur J Neurol. 2012. ; 19 ( 1 ): 105 – 113 . [DOI] [PubMed] [Google Scholar]
  • 93. Graham JM, Grünewald RA, Sagar HJ . Hallucinosis in idiopathic Parkinson’s disease . J Neurol Neurosurg Psychiatry. 1997. ; 63 ( 4 ): 434 – 440 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Loddo G, Calandra-Buonaura G, Sambati L, et al . The treatment of sleep disorders in Parkinson’s disease: from research to clinical practice . Front Neurol. 2017. ; 8 : 42 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Sateia MJ . International classification of sleep disorders-third edition: highlights and modifications . Chest. 2014. ; 146 ( 5 ): 1387 – 1394 . [DOI] [PubMed] [Google Scholar]
  • 96. Feng F, Cai Y, Hou Y, Ou R, Jiang Z, Shang H . Excessive daytime sleepiness in Parkinson’s disease: a systematic review and meta-analysis . Parkinsonism Relat Disord. 2021. ; 85 : 133 – 140 . [DOI] [PubMed] [Google Scholar]
  • 97. Meindorfner C, Körner Y, Möller JC, Stiasny-Kolster K, Oertel WH, Krüger HP . Driving in Parkinson’s disease: mobility, accidents, and sudden onset of sleep at the wheel . Mov Disord. 2005. ; 20 ( 7 ): 832 – 842 . [DOI] [PubMed] [Google Scholar]
  • 98. Happe S, Baier PC, Helmschmied K, Meller J, Tatsch K, Paulus W . Association of daytime sleepiness with nigrostriatal dopaminergic degeneration in early Parkinson’s disease . J Neurol. 2007. ; 254 ( 8 ): 1037 – 1043 . [DOI] [PubMed] [Google Scholar]
  • 99. Louter M, Munneke M, Bloem BR, Overeem S . Nocturnal hypokinesia and sleep quality in Parkinson’s disease . J Am Geriatr Soc. 2012. ; 60 ( 6 ): 1104 – 1108 . [DOI] [PubMed] [Google Scholar]
  • 100. Höglund A, Hagell P, Broman JE, et al . Associations between fluctuations in daytime sleepiness and motor and non-motor symptoms in Parkinson’s disease . Mov Disord Clin Pract (Hoboken). 2020. ; 8 ( 1 ): 44 – 50 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Ferreira JJ, Galitzky M, Montastruc JL, Rascol O . Sleep attacks and Parkinson’s disease treatment . Lancet. 2000. ; 355 ( 9212 ): 1333 – 1334 . [DOI] [PubMed] [Google Scholar]
  • 102. Gjerstad MD, Alves G, Wentzel-Larsen T, Aarsland D, Larsen JP . Excessive daytime sleepiness in Parkinson disease: is it the drugs or the disease? Neurology. 2006. ; 67 ( 5 ): 853 – 858 . [DOI] [PubMed] [Google Scholar]
  • 103. Tholfsen LK, Larsen JP, Schulz J, Tysnes OB, Gjerstad MD . Development of excessive daytime sleepiness in early Parkinson disease . Neurology. 2015. ; 85 ( 2 ): 162 – 168 . [DOI] [PubMed] [Google Scholar]
  • 104. Frucht S, Rogers JD, Greene PE, Gordon MF, Fahn S . Falling asleep at the wheel: motor vehicle mishaps in persons taking pramipexole and ropinirole . Neurology. 1999. ; 52 ( 9 ): 1908 – 1910 . [DOI] [PubMed] [Google Scholar]
  • 105. Paus S, Brecht HM, Köster J, Seeger G, Klockgether T, Wüllner U . Sleep attacks, daytime sleepiness, and dopamine agonists in Parkinson’s disease . Mov Disord. 2003. ; 18 ( 6 ): 659 – 667 . [DOI] [PubMed] [Google Scholar]
  • 106. Pal S, Bhattacharya KF, Agapito C, Chaudhuri KR . A study of excessive daytime sleepiness and its clinical significance in three groups of Parkinson’s disease patients taking pramipexole, cabergoline and levodopa mono and combination therapy . J Neural Transm (Vienna). 2001. ; 108 ( 1 ): 71 – 77 . [DOI] [PubMed] [Google Scholar]
  • 107. Amara AW, Chahine LM, Caspell-Garcia C, et al. Parkinson’s Progression Markers Initiative . Longitudinal assessment of excessive daytime sleepiness in early Parkinson’s disease . J Neurol Neurosurg Psychiatry. 2017. ; 88 ( 8 ): 653 – 662 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Valko PO, Waldvogel D, Weller M, Bassetti CL, Held U, Baumann CR . Fatigue and excessive daytime sleepiness in idiopathic Parkinson’s disease differently correlate with motor symptoms, depression and dopaminergic treatment . Eur J Neurol. 2010. ; 17 ( 12 ): 1428 – 1436 . [DOI] [PubMed] [Google Scholar]
  • 109. Etminan M, Samii A, Takkouche B, Rochon PA . Increased risk of somnolence with the new dopamine agonists in patients with Parkinson’s disease: a meta-analysis of randomised controlled trials . Drug Saf. 2001. ; 24 ( 11 ): 863 – 868 . [DOI] [PubMed] [Google Scholar]
  • 110. Zhu K, van Hilten JJ, Marinus J . Course and risk factors for excessive daytime sleepiness in Parkinson’s disease . Parkinsonism Relat Disord. 2016. ; 24 : 34 – 40 . [DOI] [PubMed] [Google Scholar]
  • 111. Contin M, Provini F, Martinelli P, et al . Excessive daytime sleepiness and levodopa in Parkinson’s disease: polygraphic, placebo-controlled monitoring . Clin Neuropharmacol. 2003. ; 26 ( 3 ): 115 – 118 . [DOI] [PubMed] [Google Scholar]
  • 112. Bliwise DL, Trotti LM, Wilson AG, et al . Daytime alertness in Parkinson’s disease: potentially dose-dependent, divergent effects by drug class . Mov Disord. 2012. ; 27 ( 9 ): 1118 – 1124 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Doghramji K, Mitler MM, Sangal RB, et al . A normative study of the maintenance of wakefulness test (MWT) . Electroencephalogr Clin Neurophysiol. 1997. ; 103 ( 5 ): 554 – 562 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Carskadon MA, Dement WC, Mitler MM, Roth T, Westbrook PR, Keenan S . Guidelines for the multiple sleep latency test (MSLT): a standard measure of sleepiness . Sleep. 1986. ; 9 ( 4 ): 519 – 524 . [DOI] [PubMed] [Google Scholar]
  • 115. Liguori C, Mercuri NB, Albanese M, Olivola E, Stefani A, Pierantozzi M . Daytime sleepiness may be an independent symptom unrelated to sleep quality in Parkinson’s disease . J Neurol. 2019. ; 266 ( 3 ): 636 – 641 . [DOI] [PubMed] [Google Scholar]
  • 116. Pagano G, Molloy S, Bain PG, et al . Sleep problems and hypothalamic dopamine D3 receptor availability in Parkinson disease . Neurology. 2016. ; 87 ( 23 ): 2451 – 2456 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Arnulf I, Konofal E, Merino-Andreu M, et al . Parkinson’s disease and sleepiness: an integral part of PD . Neurology. 2002. ; 58 ( 7 ): 1019 – 1024 . [DOI] [PubMed] [Google Scholar]
  • 118. Zhong M, Gu R, Zhu S, et al . Prevalence and risk factors for minor hallucinations in patients with Parkinson’s disease . Behav Neurol. 2021. ; 2021 : 3469706 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Powell A, Ireland C, Lewis SJG . Visual hallucinations and the role of medications in Parkinson’s disease: triggers, pathophysiology, and management . J Neuropsychiatry Clin Neurosci. 2020. ; 32 ( 4 ): 334 – 343 . [DOI] [PubMed] [Google Scholar]
  • 120. Maria B, Sophia S, Michalis M, et al . Sleep breathing disorders in patients with idiopathic Parkinson’s disease . Respir Med. 2003. ; 97 ( 10 ): 1151 – 1157 . [DOI] [PubMed] [Google Scholar]
  • 121. Cochen De Cock V, Abouda M, Leu S, et al . Is obstructive sleep apnea a problem in Parkinson’s disease? Sleep Med. 2010. ; 11 ( 3 ): 247 – 252 . [DOI] [PubMed] [Google Scholar]
  • 122. Neikrug AB, Liu L, Avanzino JA, et al . Continuous positive airway pressure improves sleep and daytime sleepiness in patients with Parkinson disease and sleep apnea . Sleep. 2014. ; 37 ( 1 ): 177 – 185 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Mery VP, Gros P, Lafontaine AL, et al . Reduced cognitive function in patients with Parkinson disease and obstructive sleep apnea . Neurology. 2017. ; 88 ( 12 ): 1120 – 1128 . [DOI] [PubMed] [Google Scholar]
  • 124. Harmell AL, Neikrug AB, Palmer BW, et al . Obstructive sleep apnea and cognition in Parkinson’s disease . Sleep Med. 2016. ; 21 : 28 – 34 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Kaminska M, Mery VP, Lafontaine AL, et al . Change in cognition and other non-motor symptoms with obstructive sleep apnea treatment in Parkinson disease . J Clin Sleep Med. 2018. ; 14 ( 5 ): 819 – 828 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Vincken WG, Darauay CM, Cosio MG . Reversibility of upper airway obstruction after levodopa therapy in Parkinson’s disease . Chest. 1989. ; 96 ( 1 ): 210 – 212 . [DOI] [PubMed] [Google Scholar]
  • 127. Herer B, Arnulf I, Housset B . Effects of levodopa on pulmonary function in Parkinson’s disease . Chest. 2001. ; 119 ( 2 ): 387 – 393 . [DOI] [PubMed] [Google Scholar]
  • 128. Obenour WH, Stevens PM, Cohen AA, McCutchen JJ . The causes of abnormal pulmonary function in Parkinson’s disease . Am Rev Respir Dis. 1972. ; 105 ( 3 ): 382 – 387 . [DOI] [PubMed] [Google Scholar]
  • 129. de Bruin PFC, de Bruin VMS, Lees AJ, Pride NB . Effects of treatment on airway dynamics and respiratory muscle strength in Parkinson’s disease . Am Rev Respir Dis. 1993. 148 ( 6_pt_1 ): 1576 – 1580 . [DOI] [PubMed] [Google Scholar]
  • 130. Serebrovskaya T, Karaban I, Mankovskaya I, Bernardi L, Passino C, Appenzeller O . Hypoxic ventilatory responses and gas exchange in patients with Parkinson’s disease . Respiration. 1998. ; 65 ( 1 ): 28 – 33 . [DOI] [PubMed] [Google Scholar]
  • 131. Rice JE, Antic R, Thompson PD . Disordered respiration as a levodopa-induced dyskinesia in Parkinson’s disease . Mov Disord. 2002. ; 17 ( 3 ): 524 – 527 . [DOI] [PubMed] [Google Scholar]
  • 132. Shen Y, Shen Y, Dong ZF, Pan PL, Shi HC, Liu CF . Obstructive sleep apnea in Parkinson’s disease: a study in 239 Chinese patients . Sleep Med. 2020. ; 67 : 237 – 243 . [DOI] [PubMed] [Google Scholar]
  • 133. Valko PO, Hauser S, Sommerauer M, Werth E, Baumann CR . Observations on sleep-disordered breathing in idiopathic Parkinson’s disease . PLoS One. 2014. ; 9 ( 6 ): e100828 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Gros P, Mery VP, Lafontaine AL, et al . Obstructive sleep apnea in Parkinson’s disease patients: effect of Sinemet CR taken at bedtime . Sleep Breath. 2016. ; 20 ( 1 ): 205 – 212 . [DOI] [PubMed] [Google Scholar]
  • 135. Bugalho P, Viana-Baptista M . REM sleep behavior disorder and motor dysfunction in Parkinson’s disease--a longitudinal study . Parkinsonism Relat Disord. 2013. ; 19 ( 12 ): 1084 – 1087 . [DOI] [PubMed] [Google Scholar]
  • 136. Sixel-Döring F, Trautmann E, Mollenhauer B, Trenkwalder C . Associated factors for REM sleep behavior disorder in Parkinson disease . Neurology. 2011. ; 77 ( 11 ): 1048 – 1054 . [DOI] [PubMed] [Google Scholar]
  • 137. Romenets SR, Gagnon JF, Latreille V, et al . Rapid eye movement sleep behavior disorder and subtypes of Parkinson’s disease . Mov Disord. 2012. ; 27 ( 8 ): 996 – 1003 . [DOI] [PubMed] [Google Scholar]
  • 138. Albin RL, Koeppe RA, Chervin RD, et al . Decreased striatal dopaminergic innervation in REM sleep behavior disorder . Neurology. 2000. ; 55 ( 9 ): 1410 – 1412 . [DOI] [PubMed] [Google Scholar]
  • 139. Iranzo A, Tolosa E, Gelpi E, et al . Neurodegenerative disease status and post-mortem pathology in idiopathic rapid-eye-movement sleep behaviour disorder: an observational cohort study . Lancet Neurol. 2013. ; 12 ( 5 ): 443 – 453 . [DOI] [PubMed] [Google Scholar]
  • 140. Fénelon G, Mahieux F, Huon R, Ziégler M . Hallucinations in Parkinson’s disease: prevalence, phenomenology and risk factors . Brain. 2000. ; 123 ( Pt 4 ): 733 – 745 . [DOI] [PubMed] [Google Scholar]
  • 141. Onofrj M, Thomas A, D’Andreamatteo G, et al . Incidence of RBD and hallucination in patients affected by Parkinson’s disease: 8-year follow-up . Neurol Sci. 2002. ; 23 ( Suppl 2 ): S91 – S94 . [DOI] [PubMed] [Google Scholar]
  • 142. Meral H, Aydemir T, Ozer F, et al . Relationship between visual hallucinations and REM sleep behavior disorder in patients with Parkinson’s disease . Clin Neurol Neurosurg. 2007. ; 109 ( 10 ): 862 – 867 . [DOI] [PubMed] [Google Scholar]
  • 143. Lenka A, Hegde S, Jhunjhunwala KR, Pal PK . Interactions of visual hallucinations, rapid eye movement sleep behavior disorder and cognitive impairment in Parkinson’s disease: a review . Parkinsonism Relat Disord. 2016. ; 22 : 1 – 8 . [DOI] [PubMed] [Google Scholar]
  • 144. Lenka A, George L, Arumugham SS, et al . Predictors of onset of psychosis in patients with Parkinson’s disease: who gets it early? Parkinsonism Relat Disord. 2017. ; 44 : 91 – 94 . [DOI] [PubMed] [Google Scholar]
  • 145. Kim YE, Jeon BS, Yang HJ, et al . REM sleep behavior disorder: association with motor complications and impulse control disorders in Parkinson’s disease . Parkinsonism Relat Disord. 2014. ; 20 ( 10 ): 1081 – 1084 . [DOI] [PubMed] [Google Scholar]
  • 146. Meloni M, Bortolato M, Cannas A, et al . Association between dopaminergic medications and REM sleep behavior disorder in Parkinson’s disease: a preliminary cohort study . J Neurol. 2020. ; 267 ( 10 ): 2926 – 2931 . [DOI] [PubMed] [Google Scholar]
  • 147. Iranzo A, Valldeoriola F, Lomeña F, et al . Serial dopamine transporter imaging of nigrostriatal function in patients with idiopathic rapid-eye-movement sleep behaviour disorder: a prospective study . Lancet Neurol. 2011. ; 10 ( 9 ): 797 – 805 . [DOI] [PubMed] [Google Scholar]
  • 148. Schmidt MH, Koshal VB, Schmidt HS . Use of pramipexole in REM sleep behavior disorder: results from a case series . Sleep Med. 2006. ; 7 ( 5 ): 418 – 423 . [DOI] [PubMed] [Google Scholar]
  • 149. Fantini ML, Gagnon JF, Filipini D, Montplaisir J . The effects of pramipexole in REM sleep behavior disorder . Neurology. 2003. ; 61 ( 10 ): 1418 – 1420 . [DOI] [PubMed] [Google Scholar]
  • 150. Sasai T, Inoue Y, Matsuura M . Effectiveness of pramipexole, a dopamine agonist, on rapid eye movement sleep behavior disorder . Tohoku J Exp Med. 2012. ; 226 ( 3 ): 177 – 181 . [DOI] [PubMed] [Google Scholar]
  • 151. Kumru H, Iranzo A, Carrasco E, et al . Lack of effects of pramipexole on REM sleep behavior disorder in Parkinson disease . Sleep. 2008. ; 31 ( 10 ): 1418 – 1421 . [PMC free article] [PubMed] [Google Scholar]
  • 152. Li SX, Wing YK, Lam SP, et al . Validation of a new REM sleep behavior disorder questionnaire (RBDQ-HK) . Sleep Med. 2010. ; 11 ( 1 ): 43 – 48 . [DOI] [PubMed] [Google Scholar]
  • 153. Wang Y, Yang Y, Wu H, Lan D, Chen Y, Zhao Z . Effects of rotigotine on REM sleep behavior disorder in Parkinson disease . J Clin Sleep Med. 2016. ; 12 ( 10 ): 1403 – 1409 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154. Tan SM, Wan YM . Pramipexole in the treatment of REM sleep behaviour disorder: a critical review . Psychiatry Res. 2016. ; 243 : 365 – 372 . [DOI] [PubMed] [Google Scholar]
  • 155. Fenu S, Espa E, Pisanu A, Di Chiara G . In vivo dopamine agonist properties of rotigotine: role of D1 and D2 receptors . Eur J Pharmacol. 2016. ; 788 : 183 – 191 . [DOI] [PubMed] [Google Scholar]
  • 156. Moccia M, Erro R, Picillo M, et al . A four-year longitudinal study on restless legs syndrome in Parkinson disease . Sleep. 2016. ; 39 ( 2 ): 405 – 412 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Nomura T, Inoue Y, Nakashima K . Clinical characteristics of restless legs syndrome in patients with Parkinson’s disease . J Neurol Sci. 2006. ; 250 ( 1-2 ): 39 – 44 . [DOI] [PubMed] [Google Scholar]
  • 158. Zolfaghari S, Yao CW, Wolfson C, Pelletier A, Postuma RB . Sleep disorders and future diagnosis of parkinsonism: a prospective study using the Canadian Longitudinal Study on Aging . J Parkinsons Dis. 2022. ; 12 ( 1 ): 257 – 266 . [DOI] [PubMed] [Google Scholar]
  • 159. Wong JC, Li Y, Schwarzschild MA, Ascherio A, Gao X . Restless legs syndrome: an early clinical feature of Parkinson disease in men . Sleep. 2014. ; 37 ( 2 ): 369 – 372 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160. Gossard TR, Trotti LM, Videnovic A, St Louis EK . Restless legs syndrome: contemporary diagnosis and treatment . Neurotherapeutics. 2021. ; 18 ( 1 ): 140 – 155 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161. Bliwise DL, Karroum EG, Greer SA, Factor SA, Trotti LM . Restless legs symptoms and periodic leg movements in sleep among patients with Parkinson’s disease . J Parkinsons Dis. 2022. ; 12 ( 4 ): 1339 – 1344 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162. Garcia-Borreguero D, Silber MH, Winkelman JW, et al . Guidelines for the first-line treatment of restless legs syndrome/Willis-Ekbom disease, prevention and treatment of dopaminergic augmentation: a combined task force of the IRLSSG, EURLSSG, and the RLS-foundation . Sleep Med. 2016. ; 21 : 1 – 11 . [DOI] [PubMed] [Google Scholar]
  • 163. Partinen M, Hirvonen K, Jama L, et al . Efficacy and safety of pramipexole in idiopathic restless legs syndrome: a polysomnographic dose-finding study--the PRELUDE study . Sleep Med. 2006. ; 7 ( 5 ): 407 – 417 . [DOI] [PubMed] [Google Scholar]
  • 164. Oertel WH, Stiasny-Kolster K, Bergtholdt B, et al. Pramipexole RLS Study Group . Efficacy of pramipexole in restless legs syndrome: a six-week, multicenter, randomized, double-blind study (effect-RLS study) . Mov Disord. 2007. ; 22 ( 2 ): 213 – 219 . [DOI] [PubMed] [Google Scholar]
  • 165. Adler CH, Hauser RA, Sethi K, et al . Ropinirole for restless legs syndrome: a placebo-controlled crossover trial . Neurology. 2004. ; 62 ( 8 ): 1405 – 1407 . [DOI] [PubMed] [Google Scholar]
  • 166. Trenkwalder C, Beneš H, Poewe W, et al. SP790 Study Group . Efficacy of rotigotine for treatment of moderate-to-severe restless legs syndrome: a randomised, double-blind, placebo-controlled trial . Lancet Neurol. 2008. ; 7 ( 7 ): 595 – 604 . [DOI] [PubMed] [Google Scholar]
  • 167. de Biase S, Merlino G, Lorenzut S, Valente M, Gigli GL . ADMET considerations when prescribing novel therapeutics to treat restless legs syndrome . Expert Opin Drug Metab Toxicol. 2014. ; 10 ( 10 ): 1365 – 1380 . [DOI] [PubMed] [Google Scholar]
  • 168. Zhou X, Du J, Liang Y, et al . The efficacy and safety of pharmacological treatments for restless legs syndrome: systemic review and network meta-analysis . Front Neurosci. 2021. ; 15 : 751643 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169. García-Borreguero D, Allen RP, Kohnen R, et al. International Restless Legs Syndrome Study Group . Diagnostic standards for dopaminergic augmentation of restless legs syndrome: report from a World Association of Sleep Medicine-International Restless Legs Syndrome Study Group consensus conference at the Max Planck Institute . Sleep Med. 2007. ; 8 ( 5 ): 520 – 530 . [DOI] [PubMed] [Google Scholar]
  • 170. Silber MH, Buchfuhrer MJ, Earley CJ, Koo BB, Manconi M, Winkelman JW ; Scientific and Medical Advisory Board of the Restless Legs Syndrome Foundation . The management of restless legs syndrome: an updated algorithm . Mayo Clin Proc. 2021. ; 96 ( 7 ): 1921 – 1937 . [DOI] [PubMed] [Google Scholar]
  • 171. Maestri M, Fulda S, Ferini-Strambi L, et al . Polysomnographic record and successful management of augmentation in restless legs syndrome/Willis-Ekbom disease . Sleep Med. 2014. ; 15 ( 5 ): 570 – 575 . [DOI] [PubMed] [Google Scholar]
  • 172. Cochen De Cock V . Therapies for restless legs in Parkinson’s disease . Curr Treat Options Neurol. 2019. ; 21 ( 11 ): 56 . [DOI] [PubMed] [Google Scholar]
  • 173. Schütz L, Sixel-Döring F, Hermann W . Management of sleep disturbances in Parkinson’s disease [published online ahead of print 2022 Jul 30]. J Parkinsons Dis. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174. Driver-Dunckley E, Evidente VGH, Adler CH, et al . Restless legs syndrome in Parkinson’s disease patients may improve with subthalamic stimulation . Mov Disord. 2006. ; 21 ( 8 ): 1287 – 1289 . [DOI] [PubMed] [Google Scholar]
  • 175. Chahine LM, Ahmed A, Sun Z . Effects of STN DBS for Parkinson’s disease on restless legs syndrome and other sleep-related measures . Parkinsonism Relat Disord. 2011. ; 17 ( 3 ): 208 – 211 . [DOI] [PubMed] [Google Scholar]
  • 176. Seppi K, Ray Chaudhuri K, Coelho M, et al. the collaborators of the Parkinson’s Disease Update on Non-Motor Symptoms Study Group on behalf of the Movement Disorders Society Evidence-Based Medicine Committee . Update on treatments for nonmotor symptoms of Parkinson’s disease-an evidence-based medicine review . Mov Disord. 2019. ; 34 ( 2 ): 180 – 198 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Videnovic A, Amara AW, Comella C, et al . Solriamfetol for excessive daytime sleepiness in Parkinson’s disease: phase 2 proof-of-concept trial . Mov Disord. 2021. ; 36 ( 10 ): 2408 – 2412 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178. Büchele F, Hackius M, Schreglmann SR, et al . Sodium oxybate for excessive daytime sleepiness and sleep disturbance in Parkinson disease: a randomized clinical trial . JAMA Neurol. 2018. ; 75 ( 1 ): 114 – 118 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179. Corvol JC, Azulay JP, Bosse B, et al. THN102-202 Study Investigators . THN 102 for excessive daytime sleepiness associated with Parkinson’s disease: a phase 2a trial . Mov Disord. 2022. ; 37 ( 2 ): 410 – 415 . [DOI] [PubMed] [Google Scholar]
  • 180. Trenkwalder C, Allen R, Högl B, et al . Comorbidities, treatment, and pathophysiology in restless legs syndrome . Lancet Neurol. 2018. ; 17 ( 11 ): 994 – 1005 . [DOI] [PubMed] [Google Scholar]
  • 181. Medeiros CAM, Carvalhedo de Bruin PF, Lopes LA, Magalhães MC, de Lourdes Seabra M, de Bruin VM . Effect of exogenous melatonin on sleep and motor dysfunction in Parkinson’s disease. A randomized, double blind, placebo-controlled study . J Neurol. 2007. ; 254 ( 4 ): 459 – 464 . [DOI] [PubMed] [Google Scholar]
  • 182. Dowling GA, Mastick J, Colling E, Carter JH, Singer CM, Aminoff MJ . Melatonin for sleep disturbances in Parkinson’s disease . Sleep Med. 2005. ; 6 ( 5 ): 459 – 466 . [DOI] [PubMed] [Google Scholar]
  • 183. Van Reeth O, Weibel L, Olivares E, Maccari S, Mocaer E, Turek FW . Melatonin or a melatonin agonist corrects age-related changes in circadian response to environmental stimulus . Am J Physiol Regul Integr Comp Physiol. 2001. ; 280 ( 5 ): R1582 – R1591 . [DOI] [PubMed] [Google Scholar]
  • 184. Solt LA, Wang Y, Banerjee S, et al . Regulation of circadian behaviour and metabolism by synthetic REV-ERB agonists . Nature. 2012. ; 485 ( 7396 ): 62 – 68 . [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185. Fertl E, Auff E, Doppelbauer A, Waldhauser F . Circadian secretion pattern of melatonin in de novo parkinsonian patients: evidence for phase-shifting properties of l-dopa . J Neural Transm Park Dis Dement Sect. 1993. ; 5 ( 3 ): 227 – 234 . [DOI] [PubMed] [Google Scholar]
  • 186. Bolitho SJ, Naismith SL, Rajaratnam SMW, et al . Disturbances in melatonin secretion and circadian sleep-wake regulation in Parkinson disease . Sleep Med. 2014. ; 15 ( 3 ): 342 – 347 . [DOI] [PubMed] [Google Scholar]

Articles from Journal of Clinical Sleep Medicine : JCSM : Official Publication of the American Academy of Sleep Medicine are provided here courtesy of American Academy of Sleep Medicine

RESOURCES