Abstract
Purpose of review
Sleep disorders are among the most challenging non-motor features of Parkinson’s disease (PD) and significantly affect quality of life. Research in this field has gained recent interest among clinicians and scientists and is rapidly evolving. This review is dedicated to sleep and circadian dysfunction associated with PD.
Recent findings
Most primary sleep disorders may co-exist with PD; majority of these disorders have unique features when expressed in the PD population.
Summary
We discuss the specific considerations related to the common sleep problems in Parkinson’s disease including insomnia, rapid eye movement sleep behavior disorder, restless legs syndrome, sleep disordered breathing, excessive daytime sleepiness and circadian rhythm disorders. Within each of these sleep disorders, we present updated definitions, epidemiology, etiology, diagnosis, clinical implications and management. Furthermore, areas of potential interest for further research are outlined.
Keywords: Parkinson’s disease, sleep disorders, sleepiness, circadian rhythm disorders, non-motor symptoms
INTRODUCTION
Sleep disruption was recognized in the initial description of Parkinson’s disease (PD) two centuries ago (1). Disrupted sleep and excessive daytime sleepiness are among the most common non-motor manifestations of PD. These symptoms negatively affect the quality of life, and may have significant implications for morbidity and safety in the PD population. Despite its common presence, sleep disruption is frequently under-reported by patients and under-recognized by health care providers. Treatment may be quite challenging due to limited treatment options and its potential side effects. In this article, we present an overview of the most common sleep and circadian disorders associated with PD.
1) INSOMNIA
Definition
The International Classification of sleep disorders (ICSD-3) has defined insomnia as the difficulty of initiating sleep, maintaining sleep or waking earlier than desired (2). The symptoms should occur at least 3 nights per week for at least 3 months and cause significant personal distress, interference with personal functioning in daily living (2–4). In patients with dementia, insomnia can manifest as the resistance to go to bed on appropriate schedule or the difficulty sleeping without caregiver intervention (2).
Epidemiology
Insomnia is thought to be the most common sleep disorder in PD and its prevalence ranges from 27% to 80%, part of the variability being due to differences in diagnostic criteria (5, 6). Sleep maintenance insomnia, including frequent awakenings/sleep fragmentation, is the most prevalent sleep disturbance in PD patients (81%) (5, 7). Difficulty falling asleep (18%) and early awakenings (40%) are also encountered (5, 6).
Etiology
The factors associated with insomnia in PD are multiple (8). Motor symptoms that emerge overnight causing sleep fragmentation include tremor, dyskinesia, nighttime cramps, dystonia (9). Overnight non-motor symptoms that negatively affect sleep include nocturia, autonomic dysfunction and psychiatric symptoms (9, 10). Comorbid primary sleep disorders such as obstructive sleep apnea (OSA), rapid eye movement sleep behavior disorder (RBD), restless legs syndrome (RLS) or periodic limb movements of sleep (PLMS) contribute to sleep fragmentation as well (11). Circadian dysfunction is another potential factor that may contribute to insomnia, and will be discussed separately in this review. PD-associated neurodegenerative process itself negatively impacts regulation of sleep due to changes within sleep regulatory networks. Finally, dopaminergic medications also may have negative impact on sleep continuity. Its effects on sleep are influenced by the dose and activation of specific dopaminergic receptors. Low doses of dopaminergic medications act on pre-synaptic D2 receptors, facilitating sleep and reducing wakefulness (9, 12, 13). In contrast, higher doses of dopaminergic medication act at post-synaptic D1 and D2 receptors, inhibiting sleep and increasing insomnia (9, 10, 14). Overall, chronic insomnia appears to increase with the duration of PD (5, 15).
Diagnosis
A detailed sleep history is necessary as an initial step in diagnosing insomnia in PD. Several instruments have been developed to assess sleep quality. Some of these instruments are not PD specific such as the Pittsburgh Sleep Quality Index (PSQI) or the Insomnia Severity Index (ISI) (5, 16). Other instruments are validated in the PD population. The newest version of the PD Sleep Scale (PDSS-2) addresses sleep initiation, sleep maintenance and daytime sleepiness (17). PDSS correlates well with sleep fragmentation on PSG (18). The Scale for Outcomes in PD (SCOPA) is a two subscales questionnaire that include a nighttime scale addressing sleep initiation, sleep fragmentation, sleep efficiency, sleep duration and early wakening, and a daytime scale for alertness (19). Polysomnography (PSG) is not indicated for the diagnosis of primary insomnia, both in the general population and in PD, as the diagnosis is based on the clinical diagnostic criteria. PSG, however, is indicated when co-existing sleep disorders associated with PD are suspected, such as sleep-disordered breathing, RLS, PLMS or RBD. Actigraphy is another objective tool that has gained interest due to its simplicity of use and ability to monitor rest-activity cycles over prolonged period of time. (20). It is however unable to differentiate activity due to wakefulness versus to other PD sleep disturbances such as RBD or OSA (20).
Clinical implications
Good sleep quality is of essence of PD patients. This has been long recognized as “sleep benefit in PD”, which emphasizes improved performance and responsiveness to dopaminergic medications after a good night of sleep. Poor motor function, mostly axial symptoms as well as OFF periods (i.e. time when medication is not working optimally (21)), have been associated with poor sleep in PD, including reduced total sleep time and sleep efficiency (18, 22, 23). Insomnia is also associated with autonomic dysfunction, such as gastrointestinal, urinary, pupilomotor and thermoregulatory dysfunction (5, 24, 25), as well as with depression and lower scores on cognitive tests in PD (26, 27). However, associations between insomnia dementia in PD are less clear (28), and this topic deserves more attention in future clinical investigations. Poor health-related quality of life and daytime sleepiness are other consequences of insomnia associated with PD (5, 29).
Management
The management of insomnia starts by correctly identifying its causes. Dopaminergic medications may have a role in improving sleep quality. In the context of nighttime motor dysfunction, controlled release formulation of levodopa/carbidopa may be considered, although its chronic effects on sleep architecture require further study (23, 30). Dopamine agonists have garnered recent interest in treating sleep disorders in PD. Ropinirole improved the PDSS score in a double blind study in PD patients with motor fluctuations (31). Trenkwalder et al. suggest that 24 hour transdermal rotigotine can improve sleep by improving nighttime PD symptoms such as limb restlessness, pain, cramps and early morning symptoms such as fatigue and mood (17). Another trial suggests rotigotine can increase sleep efficiency and reduce wakefulness after sleep onset and sleep latency (32). Treatment of underlying behavioral, psychiatric and sleep disorders is an important step in improving sleep duration and continuity in a well selected group of PD patients. Non-pharmacologic options such as cognitive behavioral therapy (CBT), utilized in general sleep medicine, have been explored in the PD population. Rios Romenets et al. compared 6 weeks of CBT with placebo, and reported an improvement in Insomnia Severity Index as well as examiner-reported clinical global impression of change, but not in the SCOPA score (33). Yang and Petrini studied 4 weeks of CBT found an improvement in PDSS score and sleep diary measures (onset latency, wake after sleep onset, total sleep time, time in bed and sleep efficiency) (34). Circadian based interventions, such as light therapy, are increasingly being proposed as a treatment intervention not only for sleep dysfunction but also for other non-motor manifestations of the disease.
2) EXCESSIVE DAYTIME SLEEPINESS
Definition
Excessive daytime sleepiness (EDS) is a symptom defined as the difficulty to stay alert and awake during the major waking periods of the day, culminating in unintended lapses into sleep (35). It differs from hypersomnia, being a diagnosis in the ICSD-3, for which EDS is the most important symptom (2). Sleep attacks are described as sudden “irresistible, overwhelming sleepiness without awareness of falling asleep”, with or without preceding signs (e.g. yawning, closing of the eyelids, rapid decrease of mental alertness etc.) (36). Sleep attacks are commonly associated with dopaminergic therapy, especially to exposure to dopamine agonists (37). While sudden onset sleep or sleep attacks represent a spectrum of EDS associated with PD, it is not required to establish the diagnosis of EDS.
Epidemiology
EDS is frequent in PD and its prevalence is thought to range from 20 to 60% (8, 38–41). Subjective complaint of EDS does not correlate well to objective measures of EDS (42). The variability in prevalence is attributable to multiple definitions and cut-off scores for EDS, differences in tools used to measure EDS, variability of medication regimens and comorbidities (42). The prevalence of sleep attacks ranges between 6.6% and 43% (36, 43, 44). Sleep attacks are associated with nonergoline dopamine agonists, male sex, increased Epworth Sleepiness Scale score (ESS), advanced PD severity (44). A higher dose of dopamine agonists, or higher levodopa equivalent doses rather than the specific type of dopaminergic medication was an effective predictor of EDS in PD (45, 46).
Etiology
The factors associated with EDS are multiple. SPECT based studies suggest that PD-related neurodegeneration plays a role in EDS (47). The hypocretin system, a key regulator of the sleep-wake cycle, is affected by PD specific neurodegeneration (48, 49). Fronczek et al. reported reduced levels of hypocretin-1 for 40% in prefrontal cortex and 25% in ventricular CSF (49). Concomitant sleep disorders commonly prevalent in PD, such as RLS (50, 51), OSA (46, 52) and RBD (53, 54), may contribute to EDS. EDS has been associated with a higher AHI in PD, and treatment of OSA with CPAP is associated with reduced objective EDS in PD (52). Studies also reported other predictors of EDS such as age (39), body mass index (BMI) (42), duration and severity of PD (45, 46) and non-motor symptoms such as pain (42). Several pivotal trials of levodopa and dopamine agonists revealed somnolence as a side effect in 15% –35% PD patients. EDS is most common in patients treated with a combination therapy of levodopa and dopamine agonist, followed by dopamine agonist monotherapy, and levodopa monotherapy (36, 55). Circadian dysfunction was recently reported as a potential culprit of EDS in PD (56).
Diagnosis
The gold standard for EDS diagnosis is the Multiple Sleep Latency Test (MSLT) (57, 58). Tirunahari et al. proposed the introduction of microsleep determination during an MSLT as a more sensitive (42.9%) and specific test (63.6%) for EDS compared to the standard MSLT. The Epworth Sleepiness Scale (ESS) is a self-administered questionnaire often used in clinical practice to screen for EDS (59). It is thought to successfully distinguish normal subjects from OSA, narcolepsy and idiopathic hypersomnia patients (59). The correlation between ESS and the sleep latency on MSLT remains a topic of debate (59, 60). At present, both MSLT and ESS are been used to assess EDS in the PD population (24, 52, 61–63).
Clinical implications
EDS in PD is associated with decreased health related quality of life (64, 65), worse cognitive impairment (66), worse motor and non-motor symptoms as well as increased medication wearing off (67, 68). In PD, depressive symptoms correlate significantly with EDS (27, 63). In patients with idiopathic RBD, EDS may be a marker of alpha-synuclein related neurodegeneration (69). EDS in PD, in the context of daytime sleep attacks, RBD, nocturnal insomnia, hallucinations and depression is reminiscent of narcolepsy. As described above, several studies have found a decrease in hypocretin levels in PD. However, the clinical significance and contribution of hypocretin to a PD related narcolepsy-like syndrome is still not fully elucidated (70).
Management
EDS is a multifactorial disorder, and therefore the first step is to identify its potential causes and develop treatment plan accordingly. Safety for driving is one of major topics that should also be systematically addressed during the clinical assessment of EDS. There has been recent interest in non-pharmacological options for EDS management. Light therapy, typically used for psychiatric and primary circadian sleep disorders, may be an effective non-pharmacological intervention for improving EDS in PD (71). A recent randomized placebo controlled trial showed significant improvement in ESS score, sleep fragmentation and sleep quality after 14 days of bright light therapy (72). A careful review of the current dopaminergic medication regimen is necessary with possible dose reduction or change of dopaminergic medications. Pharmacological treatment options can also be considered. The efficiency of modafinil for EDS in PD remains somewhat controversial. Although some studies have shown improvement in EDS with modafinil (100–200 milligrams per day), others have not (73–75). Stimulants may be beneficial for EDS but are not frequently prescribed in the PD population due to potential side effects. Finally, one open label trial of sodium oxybate (3–9 g in split doses), a drug used in treatment of narcolepsy, showed improvement of fatigue and daytime sleepiness in PD patients (76). Further research should focus on alternative pharmacological options to improve EDS in PD.
3) SLEEP-DISORDERED BREATHING
Definition
Obstructive sleep apnea (OSA) is the most common type of sleep-disordered breathing (SDB). OSA is defined as the cessation of breathing caused by repetitive transient pharyngeal airway collapse (77). Central and mixed apneas are less prevalent forms of SDB in the PD population.
Epidemiology
The prevalence of OSA in PD varies from 20–60% (24, 78–80). This wide range is likely due to methodological differences across studies including differences in scoring respiratory events (15, 81). Several recent studies that employed well-selected controls revealed similar prevalence of OSA in PD and general population (82, 83). Interestingly, three studies have suggested an increased risk of PD among patients with OSA with hazard ratios (HR) of 1.37, 1.84 and 2.26, respectively (84–86). Although the risks are higher, the absolute numbers remain low, and thus OSA might overall provide a minor risk for subsequent development of PD (84).
Etiology
Upper airway obstruction (UAO), pulmonary dysfunction and autonomic dysfunction have been described as common causes of OSA in PD (87, 88). Increased upper airway resistance in PD may be related to bradykinesia and chest wall rigidity. Several studies reported UAO being responsive to levodopa (89–91).
Diagnosis
The gold-standard for OSA diagnosis is polysomnography (PSG) (92). It is however expensive and inconvenient, as it requires a complex and expensive technical setting. Recently, there has been growing interests in alternatives, such as the level III portable monitoring (PM) at home (93, 94). Gros et al. suggested that PM had an acceptable specificity for moderate or severe OSA in PD patients (81), although the failure rate for study completion was higher and the signal quality poorer. PD patients can have central apnea, hypopneas with EEG arousals without desaturations, but also other comorbidities such as RBD or silent REM sleep without atonia etc. In these context, PSG remains the best option and should be favored as the diagnostic test of choice when possible. Other available tools such as the STOP-BANG and Berlin questionnaires (95, 96) have been used for OSA screening. The STOP-BANG in the PD population has a sensitivity of up to 80% for severe OSA with apnea-hypopnea index (AHI) ≥30/h, but the specificity is as low as 20% (97). The Berlin questionnaire appears to have higher specificity, that diminishes with OSA severity (100% for AHI ≥5/h, 53% for AHI≥30/h)(97).
Clinical implications
SDB has similar implications for overall health in PD compared with the general population. OSA has been linked with cognitive dysfunction in PD. Two studies reported lower scores on the Mini-Mental Status Exam (MMSE) and the Montreal Cognitive Assessment (MoCA) among PD patients with co-existent OSA compared with non-OSA patients (98, 99). Underlying mechanisms remain unclear, with intermittent hypoxemia (IH), systemic inflammation and/or hormonal imbalance being speculated as potential culprits (100). IH is thought to lead to oxidative and neural injury (101, 102), possibly via disruption of the blood-brain barrier (103). Hoth et al. found that high hypoxemia subjects performed better on learning and immediate recall than low hypoxemia subjects, suggesting a possible protective effect (104).
Management
Continuous positive airway pressure (CPAP) remains the main treatment modality for OSA in PD, similarly to the general population. Neikrug et al. reported that CPAP is well tolerated by PD patients with mild to moderate disease (52). The effect of CPAP beyond the improvement of OSA has also been explored. CPAP appears to improve attention and vigilance in the general population with OSA (105), but the effect on executive function and memory remains debatable (105, 106). Harmell et al. did not find an improvement on cognition in PD patients after 3 and 6 weeks of CPAP therapy. In another study however, MoCA score improved by 1.6±1.9 points (p=0.043) after 6 months of CPAP (107). Further research should investigate which domains of cognition are likely to improve by CPAP in PD patients.
CPAP can be difficult to tolerate among PD patients. There has been growing interest in alternative treatment modalities for OSA. Gros et al. reported improvements of OSA in PD patients with Sinemet CR 25/100, one to two tablets administered at bedtime (108). This finding possibly suggests that UAO are responsive to levodopa and are involved both in OSA and in PD pathophysiology. The interaction between OSA and PD certainly needs to be further explored.
4) RESTLESS LEGS SYNDROME
Definition
RLS has been defined by the ICSD-3 as an “unpleasant sensation in the legs at night or difficulty in initiating sleep”, usually accompanied or caused by an “disagreeable sensations” inside the calves that are often associated with general aches and pains in the legs and “relieved by movement of the limbs” (2, 109–111).
Epidemiology
The prevalence of RLS in PD ranges between 0 to 52.3%. Some studies found a higher prevalence of RLS in PD (51, 112, 113), especially in drug naïve patients (114, 115) that increased during the course of PD (114), while other studies have not (50, 116). The observed variability is controversial, as the IRLSSG diagnostic criteria have not been formally validated in PD.
Etiology
The etiology of RLS and RLS associated with PD is not fully elucidated. One of the possible mechanisms underlying RLS is central dopaminergic dysfunction. RLS is clinically responsive to dopaminergic agents and previous studies in rats found a favorable response of 6-hydroxydopamine injections in dopaminergic diencephalic spinal neurons (117). A study by Rios Romenets et al. found an increased prevalence of RLS in PD patients using domperidone, suggesting a potential contribution of dopaminergic neurons outside the blood brain barrier (118). Whether RLS and PD have common pathophysiology is debatable. Several studies found differences on SPECT imaging between idiopathic RLS and PD associated RLS, suggesting different pathophysiologic mechanisms (119, 120).
Diagnosis
The diagnosis of RLS is based on the diagnostic criteria defined in the International Classification of Sleep Disorders, third edition (2). The International RLS Study Group (IRLSSG) Rating Scale (121), has been recently updated in 2014 (111). The scale has been primarily been used in research settings and can be useful for the assessment of RLS severity. Moving forward, it will be important to validate the IRLSSG criteria in PD patients and possibly investigate what are the predisposing factors of RLS in PD (122). De Cock et al. have suggested the use of the suggested immobilization test (SIT) to differentiate PD with RLS from PD without RLS (123).
Clinical implications
PD patients with RLS tend to have higher age at PD onset as well as more advanced PD, more prominent limb parkinsonism especially tremor, depression, anxiety, autonomic dysfunction and worse nutritional status (114, 115, 124). They also tend to have delayed sleep onset, daytime sleepiness and overall poor sleep quality (114, 125, 126). RLS diagnosis in PD can be challenging, and therefore a thorough clinical interview is necessary to help differentiate between RLS and PD-specific mimickers or RLS. These include akathisia, motor restlessness, nocturnal leg restlessness, and sensory complains frequently reported by PD patients (122, 127).
Management
Evidence-based recommendations for moderate to severe RLS treatment suggest dopamine agonists including pramipexole (128–130), rotigotine (131, 132), ropinirole (133, 134) as well as gabapentin enacarbil (135–138), pregabalin (134, 136, 139) and IV ferric carboxymaltose use. To our knowledge, none have been studied in a randomized clinical trial in PD. Although dopamine agonists are initially efficacious for RLS, long term treatment can result in decreased tolerability and efficacy. It can also lead to augmentation, corresponding to increased symptoms severity after initial response to treatment (140). In the context of the latter, dopamine agonists may eventually need to be discontinued and replaced with other agents such as gabapentin, pregabalin or opiods. Allen et al. compared pregabalin versus pramipexole in a 12-week double blind clinical trial. Participants treated with pregabalin showed significantly less augmentation compared with pramipexole treated individuals (2.1% vs 7.1%, p=0.001) with long-term treatment (134). Opioids may be very effective in relieving both motor and sensory RLS symptoms (141). A double blind randomized placebo controlled trial by Trenkwalder et al. suggested that prolonged release oxycodone-naloxone is efficacious for treatment of severe RLS refractory to first-line treatment options (142). Several studies suggested that subthalamic nucleus DBS can possibly improve RLS in PD (143, 144).
5) CIRCADIAN RHYTHM DISORDERS
Circadian rhythm sleep disorders are disturbances of sleep-wake rhythm due to alterations of the circadian timing system or to a misalignment between the timing of the endogenous circadian rhythm and the sleep-wake times required by social schedules. Circadian rhythm disorders have not been systematically studied in the PD population. The prevalence of these disorders in PD is unknown. Emerging evidence suggests significant modifications of circadian system in PD (145). These include, for example, changes in firing neuronal pattern of SCN neurons in animal models of PD, as well as change in circadian amplitude (56, 146). Dopamine and circadian system have bidirectional modulatory relationships (147). Both motor and non-motor manifestations of PD exhibit strong diurnal oscillations that may be influenced by circadian function. Hormonal and molecular markers of the circadian system, such as melatonin and clock genes, have lower amplitude of their circadian rhythms in PD patients compared with healthy controls (145, 148, 149). This blunting of circadian amplitude may be reflective of weakened alerting circadian signaling that may contribute to EDS in PD (56). Circadian-based interventions, such as supplemental light exposure (light therapy), has favorable effects on sleep-wake cycles, mood, and motor manifestations of PD (72, 150, 151). Light therapy is easily administered, widely available and well-tolerated intervention that holds significant promise as a novel therapeutic approach not only for disturbed sleep-wake cycles in PD, but also for other disease manifestations. In summary, evidence available to date has positioned circadian system as a novel diagnostic and therapeutic target in PD.
6) REM SLEEP BEHAVIOR DISORDER
Definition
Rapid eye movement sleep behavior disorder (RBD) is a parasomnia characterized by enactment of dreams associated with loss of physiologic muscle atonia during REM sleep (152).
Epidemiology
The prevalence of RBD in PD patients is variable and ranges from 4 to 70% (153–156), whereas it is estimated 0.5–2% in the general population (157). RBD is considered an early feature of synucleinopathies. Multiple studies have found a conversion to either PD, dementia with Lewy bodies (DLB) or multiple system atrophy (MSA) in more than 80% of idiopathic RBD patients (158–160). Moreover, Boeve et al. found α-synuclein deposition in 94% patients with a neurodegenerative disorder and RBD (161). In longitudinal studies, idiopathic RBD precedes onset of parkinsonism by a median time of 13 years (162), up to 50 years (163), with an annual risk of developing a neurodegenerative disease of approximately 9% (164). RBD is a predictor of future cognitive decline in PD, and is more prevalent in PD patients with dementia compared with than PD patients without cognitive deficits (165–168).
Etiology
Unless triggered by pharmacological agents such as antidepressants, RBD is thought to be related to dysfunction within pontomedullary centers that regulate REM sleep including the locus coeruleus/subcoeruleus complex (169, 170). Development of RBD prior to the onset of cardinal motor symptoms of PD is reflective of synuclein-related neurodegenerative process that stems from low levels of neuroaxis and spreads through brainstem regions that are critical in regulation of REM sleep (1, 171).
Diagnosis
The gold-standard for RBD diagnosis is polysomnography (PSG) that demonstrates REM sleep without atonia (RSWA). Video analysis of events is also useful, but often unpredictable, as events are either subtle or infrequent (172). Polysomnographic EMG analysis of RSWA has good night-to-night stability, and usually a single night is sufficient to establish presence of RSWA (172–174). Several questionnaires have been developed as the screening tool for RBD (167, 175–181). Sensitivity of these instruments ranged from 74–96% and specificity from 56 to 92%. Although those questionnaires had excellent sensitivity, specificity of some was affected by the inability of the questionnaire to differentiate RBD from non-REM parasomnias, sleep disordered breathing, and seizures. When applied outside of the context of a strict validation protocol, these instruments may have less robust capacity for detecting RBD. Some of these instruments have been tested in the PD population, such as the REM behavior disorder screening questionnaire (RBDSQ), the REM Sleep Behavior Disorder Single-Question Screen (RBD1Q) and the Mayo Sleep Questionnaire which respectively showed a sensitivity of 74.2–85.4% and specificity of 43.2–96.3% for the RBDSQ (175, 182), a sensitivity of 93–100% and specificity of 48–68% for the RBD1Q (183), and finally a sensitivity of 90.3–100% and specificity of 36–85.2% for the Mayo Sleep Questionnaire (178, 182, 183). Moreover, the Hong-Kong RBD questionnaire and the Innsbruck RBD Inventory have been developed but not been validated for PD (176, 177).
Clinical implications
RBD phenotype is commonly described as “acting out dreams”. Symptoms encompass variable degree of involuntary vocal and motor behaviors on a background of action-packed and usually distressing dreams. This symptom constellation predisposes patients and their bed partners to significant injuries. Polysomnography-confirmed idiopathic RBD is considered one of the most specific and most predictive markers of prodromal PD, and is included in diagnostic criteria for prodromal PD according to the Movement Disorder Society criteria (184). This provides exciting platform for ongoing research on early biomarkers and impacts the development strategies for neuroprotective therapies. Moreover, RBD in PD is associated with differences in clinical phenotype, such as worse motor and non-motor symptoms, increased falls, more prominent dyskinesias, more hallucinations, worse cognition and overall quality of life (185–187). These differences warrant for screening and timely diagnosis of these common motor and non-motor manifestations of PD in patients with co-existent RBD.
Management
The initial step in the management of RBD is education of patients and their bed partners. This will help maximize safety within a sleep environment, which is a critical step in the management of RBD (188). Multiple studies recommend the use of clonazepam and melatonin as level B recommendations (152, 188–191). A placebo-controlled cross-over trial of melatonin in 8 male patients with mild RBD revealed complete or partial resolution of dream enactment behaviors in 7/8 patients (190). Although these two agents have not been compared, melatonin appears to be better tolerated and has fewer side effects than clonazepam (191). A combination of clonazepam and melatonin are sometimes needed to provide sufficient symptomatic effect. Pramipexole has shown some benefit in improving clinical events in a small cohort of patients, but has not been routinely used in clinical practice (192). Monoamine oxidase inhibitors, tricyclic antidepressants, serotonergic synaptic reuptake inhibitors, and noradrenergic antagonists should be avoided, as are thought to induce or aggravate RBD (189). Moving forward, future studies should design randomized controlled studies to better assess the efficacy of clonazepam and melatonin.
CONCLUSION
Sleep disorders are common in PD, and available treatment options are limited. Many of these sleep disorders have specific differences when co-expressed with PD compared with the general population. Insomnia is frequent in PD, is associated with other multiple PD sleep comorbidities and leads to sleep fragmentation that is the most common sleep disturbance in PD. EDS associated with PD can present as sleep attacks in context of dopaminergic therapy, and in certain cases in a narcolepsy-like syndrome. Safety implications, such as driving, should be therefore addressed systematically. Sleep-disordered breathing presents in somewhat different phenotype in PD compared with the general population. Obesity is less prevalent and less associated with SDB in the PD population, and central and mixed apneas more common. Circadian dysfunction has been an under-appreciated problem in PD and has started to gain more attention. Both motor and non-motor features of PD have strong diurnal oscillations that may be influenced by the circadian function. RLS may pose diagnostic challenges in PD patients, as there are numerous RLS mimickers. Moreover, RLS can be masked by dopaminergic medications used for treatment of PD symptoms. Long-term dopamine agonist use for RLS can lead to augmentation that is quite challenging to treat. RBD is linked to the premotor phase of PD and is gaining interest for disease modifying clinical trials. As evidenced in this review, there is a great opportunity and need to develop novel treatment approaches for impaired sleep and alertness in PD. Improvements in sleep may translate into beneficial effects on many other manifestations of PD. Further advancements of sleep therapeutics in PD will depend on advancement of the current understanding of pathophysiology of sleep and circadian disruption in PD. This will require an organized effort of international scientific community dedicated to this research direction.
Acknowledgments
We would like acknowledge research support form NIH/MINDS. Grant numbers: R01NS099055 and K23NS07228
Footnotes
CONFLICT OF INTERESTS
Priti Gros declares no conflict of interest.
Aleksandar Videnovic reports grants from NIH/NINDS during the conduct of the study.
Human and Animal Rights and Informed Consent
This article does not contain any studies with human or animal subjects performed by any of the authors.
References
*Important Reference
- 1.Braak H, Del Tredici K, Rub U, de Vos RA, Jansen Steur EN, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24(2):197–211. doi: 10.1016/s0197-4580(02)00065-9. [DOI] [PubMed] [Google Scholar]
- 2.Medicine AAoS. The International Classification of Sleep Disorders: Diagnostic and Coding Manual. 3. American Academy of Sleep Medicine; Darien, IL: 2014. Rev. ed. [Google Scholar]
- 3.WHO. International Classification of Diseases (ICD) 10. WHO; Geneva, Switzerland: 1994. [Google Scholar]
- 4.Association AP. Diagnostic and statistical manual of mental disorders. 5. D.A.P.A; Washington: 2013. [Google Scholar]
- 5.Ylikoski A, Martikainen K, Sieminski M, Partinen M. Parkinson’s disease and insomnia. Neurol Sci. 2015;36(11):2003–10. doi: 10.1007/s10072-015-2288-9. [DOI] [PubMed] [Google Scholar]
- 6.Loddo G, Calandra-Buonaura G, Sambati L, Giannini G, Cecere A, Cortelli P, et al. The Treatment of Sleep Disorders in Parkinson’s Disease: From Research to Clinical Practice. Front Neurol. 2017;8:42. doi: 10.3389/fneur.2017.00042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Tandberg E, Larsen JP, Karlsen K. A community-based study of sleep disorders in patients with Parkinson’s disease. Mov Disord. 1998;13(6):895–9. doi: 10.1002/mds.870130606. [DOI] [PubMed] [Google Scholar]
- 8.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. 2016 doi: 10.1016/j.smrv.2016.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Zhu K, van Hilten JJ, Marinus J. The course of insomnia in Parkinson’s disease. Parkinsonism Relat Disord. 2016;33:51–7. doi: 10.1016/j.parkreldis.2016.09.010. [DOI] [PubMed] [Google Scholar]
- 10.Tholfsen LK, Larsen JP, Schulz J, Tysnes OB, Gjerstad MD. Changes in insomnia subtypes in early Parkinson disease. Neurology. 2017;88(4):352–8. doi: 10.1212/WNL.0000000000003540. [DOI] [PubMed] [Google Scholar]
- 11.Dhawan V, Dhoat S, Williams AJ, Dimarco A, Pal S, Forbes A, et al. The range and nature of sleep dysfunction in untreated Parkinson’s disease (PD). A comparative controlled clinical study using the Parkinson’s disease sleep scale and selective polysomnography. J Neurol Sci. 2006;248(1–2):158–62. doi: 10.1016/j.jns.2006.05.004. [DOI] [PubMed] [Google Scholar]
- 12.Chahine LM, Daley J, Horn S, Duda JE, Colcher A, Hurtig H, et al. Association between dopaminergic medications and nocturnal sleep in early-stage Parkinson’s disease. Parkinsonism Relat Disord. 2013;19(10):859–63. doi: 10.1016/j.parkreldis.2013.05.009. [DOI] [PubMed] [Google Scholar]
- 13.Monti JM, Hawkins M, Jantos H, D’Angelo L, Fernandez M. Biphasic effects of dopamine D-2 receptor agonists on sleep and wakefulness in the rat. Psychopharmacology. 1988;95(3):395–400. doi: 10.1007/BF00181955. [DOI] [PubMed] [Google Scholar]
- 14.Cianchetti C, Masala C, Mangoni A, Gessa GL. Suppression of REM and delta sleep by apomorphine in man: a dopamine mimetic effect. Psychopharmacology. 1980;67(1):61–5. doi: 10.1007/BF00427596. [DOI] [PubMed] [Google Scholar]
- 15.Diederich NJ, Vaillant M, Leischen M, Mancuso G, Golinval S, Nati R, et al. Sleep apnea syndrome in Parkinson’s disease. A case-control study in 49 patients. Mov Disord. 2005;20(11):1413–8. doi: 10.1002/mds.20624. [DOI] [PubMed] [Google Scholar]
- 16.Mollayeva T, Thurairajah P, Burton K, Mollayeva S, Shapiro CM, Colantonio A. The Pittsburgh sleep quality index as a screening tool for sleep dysfunction in clinical and non-clinical samples: A systematic review and meta-analysis. Sleep Med Rev. 2016;25:52–73. doi: 10.1016/j.smrv.2015.01.009. [DOI] [PubMed] [Google Scholar]
- 17.Trenkwalder C, Kohnen R, Hogl B, Metta V, Sixel-Doring F, Frauscher B, et al. Parkinson’s disease sleep scale--validation of the revised version PDSS-2. Mov Disord. 2011;26(4):644–52. doi: 10.1002/mds.23476. [DOI] [PubMed] [Google Scholar]
- 18.Norlinah MI, Afidah KN, Noradina AT, Shamsul AS, Hamidon BB, Sahathevan R, et al. Sleep disturbances in Malaysian patients with Parkinson’s disease using polysomnography and PDSS. Parkinsonism Relat Disord. 2009;15(9):670–4. doi: 10.1016/j.parkreldis.2009.02.012. [DOI] [PubMed] [Google Scholar]
- 19.Marinus J, Visser M, van Hilten JJ, Lammers GJ, Stiggelbout AM. Assessment of sleep and sleepiness in Parkinson disease. Sleep. 2003;26(8):1049–54. doi: 10.1093/sleep/26.8.1049. [DOI] [PubMed] [Google Scholar]
- 20.Stavitsky K, Saurman JL, McNamara P, Cronin-Golomb A. Sleep in Parkinson’s disease: a comparison of actigraphy and subjective measures. Parkinsonism Relat Disord. 2010;16(4):280–3. doi: 10.1016/j.parkreldis.2010.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Postuma RB, Berg D, Stern M, Poewe W, Olanow CW, Oertel W, et al. MDS clinical diagnostic criteria for Parkinson’s disease. Mov Disord. 2015;30(12):1591–601. doi: 10.1002/mds.26424. [DOI] [PubMed] [Google Scholar]
- 22.Gomez-Esteban JC, Zarranz JJ, Lezcano E, Velasco F, Ciordia R, Rouco I, et al. Sleep complaints and their relation with drug treatment in patients suffering from Parkinson’s disease. Mov Disord. 2006;21(7):983–8. doi: 10.1002/mds.20874. [DOI] [PubMed] [Google Scholar]
- 23.Stocchi F, Barbato L, Nordera G, Berardelli A, Ruggieri S. Sleep disorders in Parkinson’s disease. J Neurol. 1998;245(Suppl 1):S15–8. doi: 10.1007/pl00007731. [DOI] [PubMed] [Google Scholar]
- 24.Chung S, Bohnen NI, Albin RL, Frey KA, Muller ML, Chervin RD. Insomnia and sleepiness in Parkinson disease: associations with symptoms and comorbidities. J Clin Sleep Med. 2013;9(11):1131–7. doi: 10.5664/jcsm.3150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Tijero B, Somme J, Gomez-Esteban JC, Berganzo K, Adhikari I, Lezcano E, et al. Relationship between sleep and dysautonomic symptoms assessed by self-report scales. Mov Disord. 2011;26(10):1967–8. doi: 10.1002/mds.23761. [DOI] [PubMed] [Google Scholar]
- 26.Erro R, Santangelo G, Picillo M, Vitale C, Amboni M, Longo K, et al. Link between non-motor symptoms and cognitive dysfunctions in de novo, drug-naive PD patients. J Neurol. 2012;259(9):1808–13. doi: 10.1007/s00415-011-6407-0. [DOI] [PubMed] [Google Scholar]
- 27.Suzuki K, Miyamoto M, Miyamoto T, Okuma Y, Hattori N, Kamei S, et al. Correlation between depressive symptoms and nocturnal disturbances in Japanese patients with Parkinson’s disease. Parkinsonism Relat Disord. 2009;15(1):15–9. doi: 10.1016/j.parkreldis.2008.02.002. [DOI] [PubMed] [Google Scholar]
- 28.Anang JB, Gagnon JF, Bertrand JA, Romenets SR, Latreille V, Panisset M, et al. Predictors of dementia in Parkinson disease: a prospective cohort study. Neurology. 2014;83(14):1253–60. doi: 10.1212/WNL.0000000000000842. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Forsaa EB, Larsen JP, Wentzel-Larsen T, Herlofson K, Alves G. Predictors and course of health-related quality of life in Parkinson’s disease. Mov Disord. 2008;23(10):1420–7. doi: 10.1002/mds.22121. [DOI] [PubMed] [Google Scholar]
- 30.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):590–6. doi: 10.1111/j.1468-1331.2010.03213.x. [DOI] [PubMed] [Google Scholar]
- 31.Ray Chaudhuri K, Martinez-Martin P, Rolfe KA, Cooper J, Rockett CB, Giorgi L, et al. Improvements in nocturnal symptoms with ropinirole prolonged release in patients with advanced Parkinson’s disease. Eur J Neurol. 2012;19(1):105–13. doi: 10.1111/j.1468-1331.2011.03442.x. [DOI] [PubMed] [Google Scholar]
- 32.Pierantozzi M, Placidi F, Liguori C, Albanese M, Imbriani P, Marciani MG, et al. Rotigotine may improve sleep architecture in Parkinson’s disease: a double-blind, randomized, placebo-controlled polysomnographic study. Sleep Med. 2016;21:140–4. doi: 10.1016/j.sleep.2016.01.016. [DOI] [PubMed] [Google Scholar]
- 33.Rios Romenets S, Creti L, Fichten C, Bailes S, Libman E, Pelletier A, et al. Doxepin and cognitive behavioural therapy for insomnia in patients with Parkinson’s disease -- a randomized study. Parkinsonism Relat Disord. 2013;19(7):670–5. doi: 10.1016/j.parkreldis.2013.03.003. [DOI] [PubMed] [Google Scholar]
- 34.Yang H, Petrini M. Effect of cognitive behavior therapy on sleep disorder in Parkinson’s disease in China: a pilot study. Nursing & health sciences. 2012;14(4):458–63. doi: 10.1111/j.1442-2018.2012.00711.x. [DOI] [PubMed] [Google Scholar]
- 35.Thorpy MJ. Classification of sleep disorders. Neurotherapeutics. 2012;9(4):687–701. doi: 10.1007/s13311-012-0145-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Paus S, Brecht HM, Koster J, Seeger G, Klockgether T, Wullner U. Sleep attacks, daytime sleepiness, and dopamine agonists in Parkinson’s disease. Mov Disord. 2003;18(6):659–67. doi: 10.1002/mds.10417. [DOI] [PubMed] [Google Scholar]
- 37.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):1908–10. doi: 10.1212/wnl.52.9.1908. [DOI] [PubMed] [Google Scholar]
- 38.Bjornara KA, Dietrichs E, Toft M. Clinical features associated with sleep disturbances in Parkinson’s disease. Clinical neurology and neurosurgery. 2014;124:37–43. doi: 10.1016/j.clineuro.2014.06.027. [DOI] [PubMed] [Google Scholar]
- 39.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: 10.1371/journal.pone.0022511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Verbaan D, van Rooden SM, Visser M, Marinus J, van Hilten JJ. Nighttime sleep problems and daytime sleepiness in Parkinson’s disease. Mov Disord. 2008;23(1):35–41. doi: 10.1002/mds.21727. [DOI] [PubMed] [Google Scholar]
- 41.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):853–8. doi: 10.1212/01.wnl.0000233980.25978.9d. [DOI] [PubMed] [Google Scholar]
- 42.Cochen De Cock V, Bayard S, Jaussent I, Charif M, Grini M, Langenier MC, et al. Daytime sleepiness in Parkinson’s disease: a reappraisal. PLoS One. 2014;9(9):e107278. doi: 10.1371/journal.pone.0107278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Tavora DG, de Bruin VM, Lopes Gama R, Lopes EM, Jorge IF, de Bruin PF. The nature of excessive sleepiness and sudden sleep onset in Parkinsons disease. Sleep Sci. 2014;7(1):13–8. doi: 10.1016/j.slsci.2014.07.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Korner Y, Meindorfner C, Moller JC, Stiasny-Kolster K, Haja D, Cassel W, et al. Predictors of sudden onset of sleep in Parkinson’s disease. Mov Disord. 2004;19(11):1298–305. doi: 10.1002/mds.20163. [DOI] [PubMed] [Google Scholar]
- 45.Tan EK, Lum SY, Wong MC. Restless legs syndrome in Parkinson’s disease. J Neurol Sci. 2002;196(1–2):33–6. doi: 10.1016/s0022-510x(02)00020-5. [DOI] [PubMed] [Google Scholar]
- 46.Poryazova R, Benninger D, Waldvogel D, Bassetti CL. Excessive daytime sleepiness in Parkinson’s disease: characteristics and determinants. Eur Neurol. 2010;63(3):129–35. doi: 10.1159/000276402. [DOI] [PubMed] [Google Scholar]
- 47.Matsui H, Nishinaka K, Oda M, Hara N, Komatsu K, Kubori T, et al. Excessive daytime sleepiness in Parkinson disease: a SPECT study. Sleep. 2006;29(7):917–20. doi: 10.1093/sleep/29.7.917. [DOI] [PubMed] [Google Scholar]
- 48.Thannickal TC, Lai YY, Siegel JM. Hypocretin (orexin) cell loss in Parkinson’s disease. Brain. 2007;130(Pt 6):1586–95. doi: 10.1093/brain/awm097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Fronczek R, Overeem S, Lee SY, Hegeman IM, van Pelt J, van Duinen SG, et al. Hypocretin (orexin) loss in Parkinson’s disease. Brain. 2007;130(Pt 6):1577–85. doi: 10.1093/brain/awm090. [DOI] [PubMed] [Google Scholar]
- 50.Verbaan D, van Rooden SM, van Hilten JJ, Rijsman RM. Prevalence and clinical profile of restless legs syndrome in Parkinson’s disease. Mov Disord. 2010;25(13):2142–7. doi: 10.1002/mds.23241. [DOI] [PubMed] [Google Scholar]
- 51.Bhalsing K, Suresh K, Muthane UB, Pal PK. Prevalence and profile of Restless Legs Syndrome in Parkinson’s disease and other neurodegenerative disorders: a case-control study. Parkinsonism Relat Disord. 2013;19(4):426–30. doi: 10.1016/j.parkreldis.2012.12.005. [DOI] [PubMed] [Google Scholar]
- 52.Neikrug AB, Liu L, Avanzino JA, Maglione JE, Natarajan L, Bradley L, et al. Continuous positive airway pressure improves sleep and daytime sleepiness in patients with Parkinson disease and sleep apnea. Sleep. 2014;37(1):177–85. doi: 10.5665/sleep.3332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Rolinski M, Szewczyk-Krolikowski K, Tomlinson PR, Nithi K, Talbot K, Ben-Shlomo Y, et al. REM sleep behaviour disorder is associated with worse quality of life and other non-motor features in early Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2014;85(5):560–6. doi: 10.1136/jnnp-2013-306104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Poryazova R, Oberholzer M, Baumann CR, Bassetti CL. REM sleep behavior disorder in Parkinson’s disease: a questionnaire-based survey. J Clin Sleep Med. 2013;9(1):55–9a. doi: 10.5664/jcsm.2340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Arnulf I, Leu-Semenescu S. Sleepiness in Parkinson’s disease. Parkinsonism Relat Disord. 2009;15(Suppl 3):S101–4. doi: 10.1016/S1353-8020(09)70792-8. [DOI] [PubMed] [Google Scholar]
- 56.Videnovic A, Noble C, Reid KJ, Peng J, Turek FW, Marconi A, et al. Circadian melatonin rhythm and excessive daytime sleepiness in Parkinson disease. JAMA Neurol. 2014;71(4):463–9. doi: 10.1001/jamaneurol.2013.6239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Chervin RD. Sleepiness, fatigue, tiredness, and lack of energy in obstructive sleep apnea. Chest. 2000;118(2):372–9. doi: 10.1378/chest.118.2.372. [DOI] [PubMed] [Google Scholar]
- 58.Thorpy MJ. The clinical use of the Multiple Sleep Latency Test. The Standards of Practice Committee of the American Sleep Disorders Association. Sleep. 1992;15(3):268–76. doi: 10.1093/sleep/15.3.268. [DOI] [PubMed] [Google Scholar]
- 59.Johns MW. A new method for measuring daytime sleepiness: the Epworth sleepiness scale. Sleep. 1991;14(6):540–5. doi: 10.1093/sleep/14.6.540. [DOI] [PubMed] [Google Scholar]
- 60.Cai SJ, Chen R, Zhang YL, Xiong KP, Lian YX, Li J, et al. Correlation of Epworth Sleepiness Scale with multiple sleep latency test and its diagnostic accuracy in assessing excessive daytime sleepiness in patients with obstructive sleep apnea hypopnea syndrome. Chin Med J (Engl) 2013;126(17):3245–50. [PubMed] [Google Scholar]
- 61.Ataide M, Franco CM, Lins OG. Daytime sleepiness and Parkinson’s disease: the contribution of the multiple sleep latency test. Sleep Disord. 2014;2014:767181. doi: 10.1155/2014/767181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Mery VP, Gros P, Lafontaine AL, Robinson A, Benedetti A, Kimoff RJ, et al. Reduced Cognitive Function in Patients with Parkinson’s disease and Obstructive Sleep Apnea. Neurology. 2017 doi: 10.1212/WNL.0000000000003738. Accepted for publication on January 3rd 2017. [DOI] [PubMed] [Google Scholar]
- 63.Gong Y, Liu CF. An analysis of clinical characteristics and factors in Parkinson’s disease patients with excessive daytime sleepiness. Zhonghua Nei Ke Za Zhi. 2016;55(7):515–9. doi: 10.3760/cma.j.issn.0578-1426.2016.07.006. [DOI] [PubMed] [Google Scholar]
- 64.Pandey S, Bajaj BK, Wadhwa A, Anand KS. Impact of sleep quality on the quality of life of patients with Parkinson’s disease: a questionnaire based study. Clinical neurology and neurosurgery. 2016;148:29–34. doi: 10.1016/j.clineuro.2016.06.014. [DOI] [PubMed] [Google Scholar]
- 65.Qin Z, Zhang L, Sun F, Fang X, Meng C, Tanner C, et al. Health related quality of life in early Parkinson’s disease: impact of motor and non-motor symptoms, results from Chinese levodopa exposed cohort. Parkinsonism Relat Disord. 2009;15(10):767–71. doi: 10.1016/j.parkreldis.2009.05.011. [DOI] [PubMed] [Google Scholar]
- 66.Goldman JG, Stebbins GT, Leung V, Tilley BC, Goetz CG. Relationships among cognitive impairment, sleep, and fatigue in Parkinson’s disease using the MDS-UPDRS. Parkinsonism Relat Disord. 2014;20(11):1135–9. doi: 10.1016/j.parkreldis.2014.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Perez Lloret S, Rossi M, Merello M, Rascol O, Cardinali DP. Nonmotor symptoms groups in Parkinson’s disease patients: results of a pilot, exploratory study. Parkinsons Dis. 2011;2011:473579. doi: 10.4061/2011/473579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Verbaan D, Marinus J, Visser M, van Rooden SM, Stiggelbout AM, van Hilten JJ. Patient-reported autonomic symptoms in Parkinson disease. Neurology. 2007;69(4):333–41. doi: 10.1212/01.wnl.0000266593.50534.e8. [DOI] [PubMed] [Google Scholar]
- 69.Zhou J, Zhang J, Lam SP, Chan JW, Mok V, Chan A, et al. Excessive Daytime Sleepiness Predicts Neurodegeneration in Idiopathic REM Sleep Behavior Disorder. Sleep. 2017 doi: 10.1093/sleep/zsx041. [DOI] [PubMed] [Google Scholar]
- 70.Baumann CR, Scammell TE, Bassetti CL. Parkinson’s disease, sleepiness and hypocretin/orexin. Brain. 2008;131(Pt 3):e91. doi: 10.1093/brain/awm220. [DOI] [PubMed] [Google Scholar]
- 71.van Maanen A, Meijer AM, van der Heijden KB, Oort FJ. The effects of light therapy on sleep problems: A systematic review and meta-analysis. Sleep Med Rev. 2016;29:52–62. doi: 10.1016/j.smrv.2015.08.009. [DOI] [PubMed] [Google Scholar]
- 72*.Videnovic A, Klerman EB, Wang W, Marconi A, Kuhta T, Zee PC. Timed Light Therapy for Sleep and Daytime Sleepiness Associated With Parkinson Disease: A Randomized Clinical Trial. JAMA Neurol. 2017 doi: 10.1001/jamaneurol.2016.5192. This article is recent trial that assessed safety and efficacy of light therapy on excessive daytime sleepiness in Parkinson’s disease patients. It highlights non-invasive and effective way to manage impaired alertness and poor sleep in Parkinson’s disease. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Ondo WG, Fayle R, Atassi F, Jankovic J. Modafinil for daytime somnolence in Parkinson’s disease: double blind, placebo controlled parallel trial. J Neurol Neurosurg Psychiatry. 2005;76(12):1636–9. doi: 10.1136/jnnp.2005.065870. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Adler CH, Caviness JN, Hentz JG, Lind M, Tiede J. Randomized trial of modafinil for treating subjective daytime sleepiness in patients with Parkinson’s disease. Mov Disord. 2003;18(3):287–93. doi: 10.1002/mds.10390. [DOI] [PubMed] [Google Scholar]
- 75.Hogl B, Saletu M, Brandauer E, Glatzl S, Frauscher B, Seppi K, et al. Modafinil for the treatment of daytime sleepiness in Parkinson’s disease: a double-blind, randomized, crossover, placebo-controlled polygraphic trial. Sleep. 2002;25(8):905–9. [PubMed] [Google Scholar]
- 76.Ondo WG, Perkins T, Swick T, Hull KL, Jr, Jimenez JE, Garris TS, et al. Sodium oxybate for excessive daytime sleepiness in Parkinson disease: an open-label polysomnographic study. Arch Neurol. 2008;65(10):1337–40. doi: 10.1001/archneur.65.10.1337. [DOI] [PubMed] [Google Scholar]
- 77.Ramirez JM, Garcia AJ, 3rd, Anderson TM, Koschnitzky JE, Peng YJ, Kumar GK, et al. Central and peripheral factors contributing to obstructive sleep apneas. Respir Physiol Neurobiol. 2013;189(2):344–53. doi: 10.1016/j.resp.2013.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Neikrug AB, Maglione JE, Liu L, Natarajan L, Avanzino JA, Corey-Bloom J, et al. Effects of sleep disorders on the non-motor symptoms of Parkinson disease. J Clin Sleep Med. 2013;9(11):1119–29. doi: 10.5664/jcsm.3148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.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: 10.1371/journal.pone.0100828. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Beland SG, Postuma RB, Latreille V, Bertrand JA, Panisset M, Chouinard S, et al. Observational Study of the Relation between Parkinson’s Disease and Sleep Apnea. J Parkinsons Dis. 2015;5(4):805–11. doi: 10.3233/JPD-150602. [DOI] [PubMed] [Google Scholar]
- 81.Gros P, Mery VP, Lafontaine AL, Robinson A, Benedetti A, Kimoff RJ, et al. Diagnosis of Obstructive Sleep Apnea in Parkinson’s Disease Patients: Is Unattended Portable Monitoring a Suitable Tool? Parkinsons Dis. 2015;2015:258418. doi: 10.1155/2015/258418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Trotti LM, Bliwise DL. No increased risk of obstructive sleep apnea in Parkinson’s disease. Mov Disord. 2010;25(13):2246–9. doi: 10.1002/mds.23231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Cochen De Cock V, Abouda M, Leu S, Oudiette D, Roze E, Vidailhet M, et al. Is obstructive sleep apnea a problem in Parkinson’s disease? Sleep Med. 2010;11(3):247–52. doi: 10.1016/j.sleep.2009.05.008. [DOI] [PubMed] [Google Scholar]
- 84.Yeh NC, Tien KJ, Yang CM, Wang JJ, Weng SF. Increased Risk of Parkinson’s Disease in Patients With Obstructive Sleep Apnea: A Population-Based, Propensity Score-Matched, Longitudinal Follow-Up Study. Medicine (Baltimore) 2016;95(2):e2293. doi: 10.1097/MD.0000000000002293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Chen JC, Tsai TY, Li CY, Hwang JH. Obstructive sleep apnea and risk of Parkinson’s disease: a population-based cohort study. J Sleep Res. 2015;24(4):432–7. doi: 10.1111/jsr.12289. [DOI] [PubMed] [Google Scholar]
- 86.Sheu JJ, Lee HC, Lin HC, Kao LT, Chung SD. A 5-Year Follow-up Study on the Relationship between Obstructive Sleep Apnea and Parkinson Disease. J Clin Sleep Med. 2015;11(12):1403–8. doi: 10.5664/jcsm.5274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Hovestadt A, Bogaard JM, Meerwaldt JD, van der Meche FG, Stigt J. Pulmonary function in Parkinson’s disease. J Neurol Neurosurg Psychiatry. 1989;52(3):329–33. doi: 10.1136/jnnp.52.3.329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Vincken WG, Gauthier SG, Dollfuss RE, Hanson RE, Darauay CM, Cosio MG. Involvement of upper-airway muscles in extrapyramidal disorders. A cause of airflow limitation. N Engl J Med. 1984;311(7):438–42. doi: 10.1056/NEJM198408163110704. [DOI] [PubMed] [Google Scholar]
- 89.Herer B, Arnulf I, Housset B. Effects of levodopa on pulmonary function in Parkinson’s disease. Chest. 2001;119(2):387–93. doi: 10.1378/chest.119.2.387. [DOI] [PubMed] [Google Scholar]
- 90.Vincken WG, Darauay CM, Cosio MG. Reversibility of upper airway obstruction after levodopa therapy in Parkinson’s disease. Chest. 1989;96(1):210–2. doi: 10.1378/chest.96.1.210. [DOI] [PubMed] [Google Scholar]
- 91.Tsai CC, Wu MN, Liou LM, Chang YP. Levodopa reverse stridor and prevent subsequent endotracheal intubation in Parkinson disease patients with bilateral vocal cord palsy: A case report. Medicine (Baltimore) 2016;95(50):e5559. doi: 10.1097/MD.0000000000005559. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Iber C, Ancoli-Israel S, Chesson AL, Quran SF. The AASM Manual for the Scoring of Sleep and Associated Events: Rules, Terminology, and Technical Specifications. 1. Westchester, IL: American Academy of Sleep Medicine; 2007. [Google Scholar]
- 93.Collop NA, Anderson WM, Boehlecke B, Claman D, Goldberg R, Gottlieb DJ, et al. Clinical guidelines for the use of unattended portable monitors in the diagnosis of obstructive sleep apnea in adult patients. Portable Monitoring Task Force of the American Academy of Sleep Medicine. J Clin Sleep Med. 2007;3(7):737–47. [PMC free article] [PubMed] [Google Scholar]
- 94.El Shayeb M, Topfer LA, Stafinski T, Pawluk L, Menon D. Diagnostic accuracy of level 3 portable sleep tests versus level 1 polysomnography for sleep-disordered breathing: a systematic review and meta-analysis. Cmaj. 2014;186(1):E25–51. doi: 10.1503/cmaj.130952. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Boynton G, Vahabzadeh A, Hammoud S, Ruzicka DL, Chervin RD. Validation of the STOP-BANG Questionnaire among Patients Referred for Suspected Obstructive Sleep Apnea. J Sleep Disord Treat Care. 2013;2(4) doi: 10.4172/2325-9639.1000121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Netzer NC, Stoohs RA, Netzer CM, Clark K, Strohl KP. Using the Berlin Questionnaire to identify patients at risk for the sleep apnea syndrome. Ann Intern Med. 1999;131(7):485–91. doi: 10.7326/0003-4819-131-7-199910050-00002. [DOI] [PubMed] [Google Scholar]
- 97.Gros P, Mery VP, Lafontaine AL, Robinson A, Benedetti A, Kimoff RJ, et al. Validation of Two Questionnaires for Obstructive Sleep Apnea Screening in Parkinson’s Disease Patients [abstract] Movement Disorders. 2014;29(Suppl 1):788. [Google Scholar]
- 98.Harmell AL, Neikrug AB, Palmer BW, Avanzino JA, Liu L, Maglione JE, et al. Obstructive Sleep Apnea and Cognition in Parkinson’s disease. Sleep Med. 2016;21:28–34. doi: 10.1016/j.sleep.2016.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99*.Mery VP, Gros P, Lafontaine AL, Robinson A, Benedetti A, Kimoff RJ, et al. Reduced cognitive function in patients with Parkinson disease and obstructive sleep apnea. Neurology. 2017;88(12):1120–8. doi: 10.1212/WNL.0000000000003738. This article suggests obstructive sleep apnea may be a reversible contributor to several non-motor symptoms in Parkinson’s disease (PD) including sleepiness and cognitive dysfunction. It highlights the clinical importance of OSA in PD and supports further CPAP trial as a potential interesting treatment for cognitive decline in this population. [DOI] [PubMed] [Google Scholar]
- 100.Lal C, Strange C, Bachman D. Neurocognitive impairment in obstructive sleep apnea. Chest. 2012;141(6):1601–10. doi: 10.1378/chest.11-2214. [DOI] [PubMed] [Google Scholar]
- 101.Zhan G, Fenik P, Pratico D, Veasey SC. Inducible nitric oxide synthase in long-term intermittent hypoxia: hypersomnolence and brain injury. Am J Respir Crit Care Med. 2005;171(12):1414–20. doi: 10.1164/rccm.200411-1564OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Li Y, Veasey SC. Neurobiology and neuropathophysiology of obstructive sleep apnea. Neuromolecular Med. 2012;14(3):168–79. doi: 10.1007/s12017-011-8165-7. [DOI] [PubMed] [Google Scholar]
- 103.Lim DC, Pack AI. Obstructive sleep apnea and cognitive impairment: addressing the blood-brain barrier. Sleep Med Rev. 2014;18(1):35–48. doi: 10.1016/j.smrv.2012.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Hoth KF, Zimmerman ME, Meschede KA, Arnedt JT, Aloia MS. Obstructive sleep apnea: impact of hypoxemia on memory. Sleep Breath. 2013;17(2):811–7. doi: 10.1007/s11325-012-0769-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Kylstra WA, Aaronson JA, Hofman WF, Schmand BA. Neuropsychological functioning after CPAP treatment in obstructive sleep apnea: a meta-analysis. Sleep Med Rev. 2013;17(5):341–7. doi: 10.1016/j.smrv.2012.09.002. [DOI] [PubMed] [Google Scholar]
- 106.Kushida CA, Nichols DA, Holmes TH, Quan SF, Walsh JK, Gottlieb DJ, et al. Effects of continuous positive airway pressure on neurocognitive function in obstructive sleep apnea patients: The Apnea Positive Pressure Long-term Efficacy Study (APPLES) Sleep. 2012;35(12):1593–602. doi: 10.5665/sleep.2226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Mery VP, Lafontaine AL, Robinson A, Benedetti A, Kimoff RJ, Kaminska M. Treatment of Obstructive Sleep Apnea with Continuous Positive Airway Pressure Improves Non-motor Symptoms in Parkinson’s disease Patients [abstract] Journal of Parkinson’s Disease. 2013;3(Supplement 1) Article 109. [Google Scholar]
- 108.Gros P, Mery VP, Lafontaine AL, Robinson A, Benedetti A, Kimoff RJ, et al. Obstructive sleep apnea in Parkinson’s disease patients: effect of Sinemet CR taken at bedtime. Sleep Breath. 2016;20(1):205–12. doi: 10.1007/s11325-015-1208-9. [DOI] [PubMed] [Google Scholar]
- 109.Allen RP, Picchietti D, Hening WA, Trenkwalder C, Walters AS, Montplaisi J. Restless legs syndrome: diagnostic criteria, special considerations, and epidemiology. A report from the restless legs syndrome diagnosis and epidemiology workshop at the National Institutes of Health. Sleep Med. 2003;4(2):101–19. doi: 10.1016/s1389-9457(03)00010-8. [DOI] [PubMed] [Google Scholar]
- 110.Walters AS. Toward a better definition of the restless legs syndrome. The International Restless Legs Syndrome Study Group. Mov Disord. 1995;10(5):634–42. doi: 10.1002/mds.870100517. [DOI] [PubMed] [Google Scholar]
- 111.Allen RP, Picchietti DL, Garcia-Borreguero D, Ondo WG, Walters AS, Winkelman JW, et al. Restless legs syndrome/Willis-Ekbom disease diagnostic criteria: updated International Restless Legs Syndrome Study Group (IRLSSG) consensus criteria--history, rationale, description, and significance. Sleep Med. 2014;15(8):860–73. doi: 10.1016/j.sleep.2014.03.025. [DOI] [PubMed] [Google Scholar]
- 112.Rijsman RM, Schoolderman LF, Rundervoort RS, Louter M. Restless legs syndrome in Parkinson’s disease. Parkinsonism Relat Disord. 2014;20(Suppl 1):S5–9. doi: 10.1016/S1353-8020(13)70004-X. [DOI] [PubMed] [Google Scholar]
- 113.Peralta CM, Frauscher B, Seppi K, Wolf E, Wenning GK, Hogl B, et al. Restless legs syndrome in Parkinson’s disease. Mov Disord. 2009;24(14):2076–80. doi: 10.1002/mds.22694. [DOI] [PubMed] [Google Scholar]
- 114.Moccia M, Erro R, Picillo M, Santangelo G, Spina E, Allocca R, et al. A Four-Year Longitudinal Study on Restless Legs Syndrome in Parkinson Disease. Sleep. 2016;39(2):405–12. doi: 10.5665/sleep.5452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Shin HY, Youn J, Yoon WT, Kim JS, Cho JW. Restless legs syndrome in Korean patients with drug-naive Parkinson’s disease: a nation-wide study. Parkinsonism Relat Disord. 2013;19(3):355–8. doi: 10.1016/j.parkreldis.2012.09.009. [DOI] [PubMed] [Google Scholar]
- 116.Suzuki K, Miyamoto M, Miyamoto T, Tatsumoto M, Watanabe Y, Suzuki S, et al. Nocturnal disturbances and restlessness in Parkinson’s disease: using the Japanese version of the Parkinson’s disease sleep scale-2. J Neurol Sci. 2012;318(1–2):76–81. doi: 10.1016/j.jns.2012.03.022. [DOI] [PubMed] [Google Scholar]
- 117.Ondo WG, He Y, Rajasekaran S, Le WD. Clinical correlates of 6-hydroxydopamine injections into A11 dopaminergic neurons in rats: a possible model for restless legs syndrome. Mov Disord. 2000;15(1):154–8. doi: 10.1002/1531-8257(200001)15:1<154::aid-mds1025>3.0.co;2-q. [DOI] [PubMed] [Google Scholar]
- 118.Rios Romenets S, Dauvilliers Y, Cochen De Cock V, Carlander B, Bayard S, Galatas C, et al. Restless legs syndrome outside the blood-brain barrier--exacerbation by domperidone in Parkinson’s disease. Parkinsonism Relat Disord. 2013;19(1):92–4. doi: 10.1016/j.parkreldis.2012.07.019. [DOI] [PubMed] [Google Scholar]
- 119.Kwon DY, Seo WK, Yoon HK, Park MH, Koh SB, Park KW. Transcranial brain sonography in Parkinson’s disease with restless legs syndrome. Mov Disord. 2010;25(10):1373–8. doi: 10.1002/mds.23066. [DOI] [PubMed] [Google Scholar]
- 120.Ryu JH, Lee MS, Baik JS. Sonographic abnormalities in idiopathic restless legs syndrome (RLS) and RLS in Parkinson’s disease. Parkinsonism Relat Disord. 2011;17(3):201–3. doi: 10.1016/j.parkreldis.2010.11.014. [DOI] [PubMed] [Google Scholar]
- 121.Walters AS, LeBrocq C, Dhar A, Hening W, Rosen R, Allen RP, et al. Validation of the International Restless Legs Syndrome Study Group rating scale for restless legs syndrome. Sleep Med. 2003;4(2):121–32. doi: 10.1016/s1389-9457(02)00258-7. [DOI] [PubMed] [Google Scholar]
- 122.Moller JC, Unger M, Stiasny-Kolster K, Oertel WH. Restless Legs Syndrome (RLS) and Parkinson’s disease (PD)-related disorders or different entities? J Neurol Sci. 2010;289(1–2):135–7. doi: 10.1016/j.jns.2009.08.035. [DOI] [PubMed] [Google Scholar]
- 123.De Cock VC, Bayard S, Yu H, Grini M, Carlander B, Postuma R, et al. Suggested immobilization test for diagnosis of restless legs syndrome in Parkinson’s disease. Mov Disord. 2012;27(6):743–9. doi: 10.1002/mds.24969. [DOI] [PubMed] [Google Scholar]
- 124.Fereshtehnejad SM, Shafieesabet M, Shahidi GA, Delbari A, Lokk J. Restless legs syndrome in patients with Parkinson’s disease: a comparative study on prevalence, clinical characteristics, quality of life and nutritional status. Acta Neurol Scand. 2015;131(4):211–8. doi: 10.1111/ane.12307. [DOI] [PubMed] [Google Scholar]
- 125.Krishnan PR, Bhatia M, Behari M. Restless legs syndrome in Parkinson’s disease: a case-controlled study. Mov Disord. 2003;18(2):181–5. doi: 10.1002/mds.10307. [DOI] [PubMed] [Google Scholar]
- 126.Nomura T, Inoue Y, Miyake M, Yasui K, Nakashima K. Prevalence and clinical characteristics of restless legs syndrome in Japanese patients with Parkinson’s disease. Mov Disord. 2006;21(3):380–4. doi: 10.1002/mds.20734. [DOI] [PubMed] [Google Scholar]
- 127.Gjerstad MD, Tysnes OB, Larsen JP. Increased risk of leg motor restlessness but not RLS in early Parkinson disease. Neurology. 2011;77(22):1941–6. doi: 10.1212/WNL.0b013e31823a0cc8. [DOI] [PubMed] [Google Scholar]
- 128.Winkelman JW, Sethi KD, Kushida CA, Becker PM, Koester J, Cappola JJ, et al. Efficacy and safety of pramipexole in restless legs syndrome. Neurology. 2006;67(6):1034–9. doi: 10.1212/01.wnl.0000231513.23919.a1. [DOI] [PubMed] [Google Scholar]
- 129.Ma JF, Wan Q, Hu XY, Sun SG, Wang WZ, Zhao ZX, et al. Efficacy and safety of pramipexole in chinese patients with restless legs syndrome: results from a multi-center, randomized, double-blind, placebo-controlled trial. Sleep Med. 2012;13(1):58–63. doi: 10.1016/j.sleep.2011.03.021. [DOI] [PubMed] [Google Scholar]
- 130.Zhang J, Liu B, Zheng Y, Chu T, Yang Z. Pramipexole for Chinese people with primary restless legs syndrome: a 12-week multicenter, randomized, double-blind study. Sleep Med. 2015;16(1):181–5. doi: 10.1016/j.sleep.2014.09.015. [DOI] [PubMed] [Google Scholar]
- 131.Stiasny-Kolster K, Kohnen R, Schollmayer E, Moller JC, Oertel WH. Patch application of the dopamine agonist rotigotine to patients with moderate to advanced stages of restless legs syndrome: a double-blind, placebo-controlled pilot study. Mov Disord. 2004;19(12):1432–8. doi: 10.1002/mds.20251. [DOI] [PubMed] [Google Scholar]
- 132.Oertel WH, Benes H, Garcia-Borreguero D, Hogl B, Poewe W, Montagna P, et al. Rotigotine transdermal patch in moderate to severe idiopathic restless legs syndrome: a randomized, placebo-controlled polysomnographic study. Sleep Med. 2010;11(9):848–56. doi: 10.1016/j.sleep.2010.02.014. [DOI] [PubMed] [Google Scholar]
- 133.Walters AS, Ondo WG, Dreykluft T, Grunstein R, Lee D, Sethi K. Ropinirole is effective in the treatment of restless legs syndrome. TREAT RLS 2: a 12-week, double-blind, randomized, parallel-group, placebo-controlled study. Mov Disord. 2004;19(12):1414–23. doi: 10.1002/mds.20257. [DOI] [PubMed] [Google Scholar]
- 134.Allen RP, Chen C, Garcia-Borreguero D, Polo O, DuBrava S, Miceli J, et al. Comparison of pregabalin with pramipexole for restless legs syndrome. N Engl J Med. 2014;370(7):621–31. doi: 10.1056/NEJMoa1303646. [DOI] [PubMed] [Google Scholar]
- 135.Kushida CA, Walters AS, Becker P, Thein SG, Perkins AT, Roth T, et al. A randomized, double-blind, placebo-controlled, crossover study of XP13512/GSK1838262 in the treatment of patients with primary restless legs syndrome. Sleep. 2009;32(2):159–68. doi: 10.1093/sleep/32.2.159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Walters AS, Ondo WG, Kushida CA, Becker PM, Ellenbogen AL, Canafax DM, et al. Gabapentin enacarbil in restless legs syndrome: a phase 2b, 2-week, randomized, double-blind, placebo-controlled trial. Clin Neuropharmacol. 2009;32(6):311–20. doi: 10.1097/WNF.0b013e3181b3ab16. [DOI] [PubMed] [Google Scholar]
- 137.Kushida CA, Becker PM, Ellenbogen AL, Canafax DM, Barrett RW. Randomized, double-blind, placebo-controlled study of XP13512/GSK1838262 in patients with RLS. Neurology. 2009;72(5):439–46. doi: 10.1212/01.wnl.0000341770.91926.cc. [DOI] [PubMed] [Google Scholar]
- 138.Lee DO, Ziman RB, Perkins AT, Poceta JS, Walters AS, Barrett RW. A randomized, double-blind, placebo-controlled study to assess the efficacy and tolerability of gabapentin enacarbil in subjects with restless legs syndrome. J Clin Sleep Med. 2011;7(3):282–92. doi: 10.5664/JCSM.1074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Garcia-Borreguero D, Patrick J, DuBrava S, Becker PM, Lankford A, Chen C, et al. Pregabalin versus pramipexole: effects on sleep disturbance in restless legs syndrome. Sleep. 2014;37(4):635–43. doi: 10.5665/sleep.3558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Garcia-Borreguero D, Allen RP, Benes H, Earley C, Happe S, Hogl B, et al. Augmentation as a treatment complication of restless legs syndrome: concept and management. Mov Disord. 2007;22(Suppl 18):S476–84. doi: 10.1002/mds.21610. [DOI] [PubMed] [Google Scholar]
- 141.Trenkwalder C, Zieglgansberger W, Ahmedzai SH, Hogl B. Pain, opioids, and sleep: implications for restless legs syndrome treatment. Sleep Med. 2017;31:78–85. doi: 10.1016/j.sleep.2016.09.017. [DOI] [PubMed] [Google Scholar]
- 142.Trenkwalder C, Benes H, Grote L, Garcia-Borreguero D, Hogl B, Hopp M, et al. Prolonged release oxycodone-naloxone for treatment of severe restless legs syndrome after failure of previous treatment: a double-blind, randomised, placebo-controlled trial with an open-label extension. Lancet Neurol. 2013;12(12):1141–50. doi: 10.1016/S1474-4422(13)70239-4. [DOI] [PubMed] [Google Scholar]
- 143.Driver-Dunckley E, Evidente VG, Adler CH, Hillman R, Hernandez J, Fletcher G, et al. Restless legs syndrome in Parkinson’s disease patients may improve with subthalamic stimulation. Mov Disord. 2006;21(8):1287–9. doi: 10.1002/mds.20911. [DOI] [PubMed] [Google Scholar]
- 144.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):208–11. doi: 10.1016/j.parkreldis.2010.11.017. [DOI] [PubMed] [Google Scholar]
- 145.Videnovic A, Lazar AS, Barker RA, Overeem S. ‘The clocks that time us’--circadian rhythms in neurodegenerative disorders. Nature reviews Neurology. 2014;10(12):683–93. doi: 10.1038/nrneurol.2014.206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Kudo T, Loh DH, Truong D, Wu Y, Colwell CS. Circadian dysfunction in a mouse model of Parkinson’s disease. Experimental neurology. 2011;232(1):66–75. doi: 10.1016/j.expneurol.2011.08.003. [DOI] [PubMed] [Google Scholar]
- 147.Golombek DA, Rosenstein RE. Physiology of circadian entrainment. Physiological reviews. 2010;90(3):1063–102. doi: 10.1152/physrev.00009.2009. [DOI] [PubMed] [Google Scholar]
- 148.Bolitho SJ, Naismith SL, Rajaratnam SM, Grunstein RR, Hodges JR, Terpening Z, et al. Disturbances in melatonin secretion and circadian sleep-wake regulation in Parkinson disease. Sleep Med. 2014;15(3):342–7. doi: 10.1016/j.sleep.2013.10.016. [DOI] [PubMed] [Google Scholar]
- 149.Breen DP, Vuono R, Nawarathna U, Fisher K, Shneerson JM, Reddy AB, et al. Sleep and circadian rhythm regulation in early Parkinson disease. JAMA Neurol. 2014;71(5):589–95. doi: 10.1001/jamaneurol.2014.65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Paus S, Schmitz-Hubsch T, Wullner U, Vogel A, Klockgether T, Abele M. Bright light therapy in Parkinson’s disease: a pilot study. Mov Disord. 2007;22(10):1495–8. doi: 10.1002/mds.21542. [DOI] [PubMed] [Google Scholar]
- 151.Willis GL, Turner EJ. Primary and secondary features of Parkinson’s disease improve with strategic exposure to bright light: a case series study. Chronobiol Int. 2007;24(3):521–37. doi: 10.1080/07420520701420717. [DOI] [PubMed] [Google Scholar]
- 152.Schenck CH, Bundlie SR, Ettinger MG, Mahowald MW. Chronic behavioral disorders of human REM sleep: a new category of parasomnia. Sleep. 1986;9(2):293–308. doi: 10.1093/sleep/9.2.293. [DOI] [PubMed] [Google Scholar]
- 153.Scaglione C, Vignatelli L, Plazzi G, Marchese R, Negrotti A, Rizzo G, et al. REM sleep behaviour disorder in Parkinson’s disease: a questionnaire-based study. Neurol Sci. 2005;25(6):316–21. doi: 10.1007/s10072-004-0364-7. [DOI] [PubMed] [Google Scholar]
- 154.Wu YH, Liao YC, Chen YH, Chang MH, Lin CH. Risk of premotor symptoms in patients with newly diagnosed PD: a nationwide, population-based, case-control study in Taiwan. PLoS One. 2015;10(6):e0130282. doi: 10.1371/journal.pone.0130282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Chaudhuri KR, Martinez-Martin P, Schapira AH, Stocchi F, Sethi K, Odin P, et al. International multicenter pilot study of the first comprehensive self-completed nonmotor symptoms questionnaire for Parkinson’s disease: the NMSQuest study. Mov Disord. 2006;21(7):916–23. doi: 10.1002/mds.20844. [DOI] [PubMed] [Google Scholar]
- 156.Sixel-Doring F, Trautmann E, Mollenhauer B, Trenkwalder C. Rapid eye movement sleep behavioral events: a new marker for neurodegeneration in early Parkinson disease? Sleep. 2014;37(3):431–8. doi: 10.5665/sleep.3468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Kang SH, Yoon IY, Lee SD, Han JW, Kim TH, Kim KW. REM sleep behavior disorder in the Korean elderly population: prevalence and clinical characteristics. Sleep. 2013;36(8):1147–52. doi: 10.5665/sleep.2874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Schenck CH, Boeve BF, Mahowald MW. Delayed emergence of a parkinsonian disorder or dementia in 81% of older men initially diagnosed with idiopathic rapid eye movement sleep behavior disorder: a 16-year update on a previously reported series. Sleep Med. 2013;14(8):744–8. doi: 10.1016/j.sleep.2012.10.009. [DOI] [PubMed] [Google Scholar]
- 159*.Iranzo A, Tolosa E, Gelpi E, Molinuevo JL, Valldeoriola F, Serradell M, 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):443–53. doi: 10.1016/S1474-4422(13)70056-5. This study by Iranzo et al. suggest that most patients with idiopathic REM behavior disorder will eventually develop a Lewy Body disorder, with a prevalence increasing with longer follow-up. Patients with no definite diagnosis at the end of the study showed short-term risk for developing PD or DLB, such as decreased striatal dopamine transporter binding on imaging. This supports the notion that iRBD patients are interesting candidates for prodromal phase of disease studies, neuroprotective or disease modifying drugs trials. [DOI] [PubMed] [Google Scholar]
- 160.Iranzo A, Fernandez-Arcos A, Tolosa E, Serradell M, Molinuevo JL, Valldeoriola F, et al. Neurodegenerative disorder risk in idiopathic REM sleep behavior disorder: study in 174 patients. PLoS One. 2014;9(2):e89741. doi: 10.1371/journal.pone.0089741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Boeve BF, Silber MH, Ferman TJ, Lin SC, Benarroch EE, Schmeichel AM, et al. Clinicopathologic correlations in 172 cases of rapid eye movement sleep behavior disorder with or without a coexisting neurologic disorder. Sleep Med. 2013;14(8):754–62. doi: 10.1016/j.sleep.2012.10.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Postuma RB, Berg D. Advances in markers of prodromal Parkinson disease. Nature reviews Neurology. 2016;12(11):622–34. doi: 10.1038/nrneurol.2016.152. [DOI] [PubMed] [Google Scholar]
- 163.Claassen DO, Josephs KA, Ahlskog JE, Silber MH, Tippmann-Peikert M, Boeve BF. REM sleep behavior disorder preceding other aspects of synucleinopathies by up to half a century. Neurology. 2010;75(6):494–9. doi: 10.1212/WNL.0b013e3181ec7fac. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Postuma RB, Iranzo A, Hogl B, Arnulf I, Ferini-Strambi L, Manni R, et al. Risk factors for neurodegeneration in idiopathic rapid eye movement sleep behavior disorder: a multicenter study. Ann Neurol. 2015;77(5):830–9. doi: 10.1002/ana.24385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Chahine LM, Xie SX, Simuni T, Tran B, Postuma R, Amara A, et al. Longitudinal changes in cognition in early Parkinson’s disease patients with REM sleep behavior disorder. Parkinsonism Relat Disord. 2016;27:102–6. doi: 10.1016/j.parkreldis.2016.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Fantini ML, Farini E, Ortelli P, Zucconi M, Manconi M, Cappa S, et al. Longitudinal study of cognitive function in idiopathic REM sleep behavior disorder. Sleep. 2011;34(5):619–25. [PMC free article] [PubMed] [Google Scholar]
- 167.Postuma RB, Bertrand JA, Montplaisir J, Desjardins C, Vendette M, Rios Romenets S, et al. Rapid eye movement sleep behavior disorder and risk of dementia in Parkinson’s disease: a prospective study. Mov Disord. 2012;27(6):720–6. doi: 10.1002/mds.24939. [DOI] [PubMed] [Google Scholar]
- 168.Anang JB, Nomura T, Romenets SR, Nakashima K, Gagnon JF, Postuma RB. Dementia Predictors in Parkinson Disease: A Validation Study. J Parkinsons Dis. 2017;7(1):159–62. doi: 10.3233/JPD-160925. [DOI] [PubMed] [Google Scholar]
- 169.Garcia-Lorenzo D, Longo-Dos Santos C, Ewenczyk C, Leu-Semenescu S, Gallea C, Quattrocchi G, et al. The coeruleus/subcoeruleus complex in rapid eye movement sleep behaviour disorders in Parkinson’s disease. Brain. 2013;136(Pt 7):2120–9. doi: 10.1093/brain/awt152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Boeve BF, Silber MH, Saper CB, Ferman TJ, Dickson DW, Parisi JE, et al. Pathophysiology of REM sleep behaviour disorder and relevance to neurodegenerative disease. Brain. 2007;130(Pt 11):2770–88. doi: 10.1093/brain/awm056. [DOI] [PubMed] [Google Scholar]
- 171.Al-Qassabi A, Fereshtehnejad SM, Postuma RB. Sleep Disturbances in the Prodromal Stage of Parkinson Disease. Curr Treat Options Neurol. 2017;19(6):22. doi: 10.1007/s11940-017-0458-1. [DOI] [PubMed] [Google Scholar]
- 172.Hogl B, Stefani A. REM sleep behavior disorder (RBD): Update on diagnosis and treatment. Somnologie: Schlafforschung und Schlafmedizin = Somnology: sleep research and sleep medicine. 2017;21(Suppl 1):1–8. doi: 10.1007/s11818-016-0048-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Ferri R, Marelli S, Cosentino FI, Rundo F, Ferini-Strambi L, Zucconi M. Night-to-night variability of automatic quantitative parameters of the chin EMG amplitude (Atonia Index) in REM sleep behavior disorder. J Clin Sleep Med. 2013;9(3):253–8. doi: 10.5664/jcsm.2490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Zhang J, Lam SP, Ho CK, Li AM, Tsoh J, Mok V, et al. Diagnosis of REM sleep behavior disorder by video-polysomnographic study: is one night enough? Sleep. 2008;31(8):1179–85. [PMC free article] [PubMed] [Google Scholar]
- 175.Stiasny-Kolster K, Mayer G, Schafer S, Moller JC, Heinzel-Gutenbrunner M, Oertel WH. The REM sleep behavior disorder screening questionnaire--a new diagnostic instrument. Mov Disord. 2007;22(16):2386–93. doi: 10.1002/mds.21740. [DOI] [PubMed] [Google Scholar]
- 176.Li SX, Wing YK, Lam SP, Zhang J, Yu MW, Ho CK, et al. Validation of a new REM sleep behavior disorder questionnaire (RBDQ-HK) Sleep Med. 2010;11(1):43–8. doi: 10.1016/j.sleep.2009.06.008. [DOI] [PubMed] [Google Scholar]
- 177.Frauscher B, Ehrmann L, Zamarian L, Auer F, Mitterling T, Gabelia D, et al. Validation of the Innsbruck REM sleep behavior disorder inventory. Mov Disord. 2012;27(13):1673–8. doi: 10.1002/mds.25223. [DOI] [PubMed] [Google Scholar]
- 178.Boeve BF, Molano JR, Ferman TJ, Lin SC, Bieniek K, Tippmann-Peikert M, et al. Validation of the Mayo Sleep Questionnaire to screen for REM sleep behavior disorder in a community-based sample. J Clin Sleep Med. 2013;9(5):475–80. doi: 10.5664/jcsm.2670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Boeve BF, Molano JR, Ferman TJ, Smith GE, Lin SC, Bieniek K, et al. Validation of the Mayo Sleep Questionnaire to screen for REM sleep behavior disorder in an aging and dementia cohort. Sleep Med. 2011;12(5):445–53. doi: 10.1016/j.sleep.2010.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Stiasny-Kolster K, Sixel-Doring F, Trenkwalder C, Heinzel-Gutenbrunner M, Seppi K, Poewe W, et al. Diagnostic value of the REM sleep behavior disorder screening questionnaire in Parkinson’s disease. Sleep Med. 2015;16(1):186–9. doi: 10.1016/j.sleep.2014.08.014. [DOI] [PubMed] [Google Scholar]
- 181.Nomura T, Inoue Y, Kagimura T, Uemura Y, Nakashima K. Utility of the REM sleep behavior disorder screening questionnaire (RBDSQ) in Parkinson’s disease patients. Sleep Med. 2011;12(7):711–3. doi: 10.1016/j.sleep.2011.01.015. [DOI] [PubMed] [Google Scholar]
- 182.Chahine LM, Daley J, Horn S, Colcher A, Hurtig H, Cantor C, et al. Questionnaire-based diagnosis of REM sleep behavior disorder in Parkinson’s disease. Mov Disord. 2013;28(8):1146–9. doi: 10.1002/mds.25438. [DOI] [PubMed] [Google Scholar]
- 183.Bolitho SJ, Naismith SL, Terpening Z, Grunstein RR, Melehan K, Yee BJ, et al. Investigating rapid eye movement sleep without atonia in Parkinson’s disease using the rapid eye movement sleep behavior disorder screening questionnaire. Mov Disord. 2014;29(6):736–42. doi: 10.1002/mds.25832. [DOI] [PubMed] [Google Scholar]
- 184.Berg D, Postuma RB, Adler CH, Bloem BR, Chan P, Dubois B, et al. MDS research criteria for prodromal Parkinson’s disease. Mov Disord. 2015;30(12):1600–11. doi: 10.1002/mds.26431. [DOI] [PubMed] [Google Scholar]
- 185.Gong Y, Xiong KP, Mao CJ, Shen Y, Hu WD, Huang JY, et al. Clinical manifestations of Parkinson disease and the onset of rapid eye movement sleep behavior disorder. Sleep Med. 2014;15(6):647–53. doi: 10.1016/j.sleep.2013.12.021. [DOI] [PubMed] [Google Scholar]
- 186.Postuma RB, Gagnon JF, Vendette M, Charland K, Montplaisir J. Manifestations of Parkinson disease differ in association with REM sleep behavior disorder. Mov Disord. 2008;23(12):1665–72. doi: 10.1002/mds.22099. [DOI] [PubMed] [Google Scholar]
- 187.Vendette M, Gagnon JF, Decary A, Massicotte-Marquez J, Postuma RB, Doyon J, et al. REM sleep behavior disorder predicts cognitive impairment in Parkinson disease without dementia. Neurology. 2007;69(19):1843–9. doi: 10.1212/01.wnl.0000278114.14096.74. [DOI] [PubMed] [Google Scholar]
- 188.Aurora RN, Zak RS, Maganti RK, Auerbach SH, Casey KR, Chowdhuri S, et al. Best practice guide for the treatment of REM sleep behavior disorder (RBD) J Clin Sleep Med. 2010;6(1):85–95. [PMC free article] [PubMed] [Google Scholar]
- 189.Gagnon JF, Postuma RB, Montplaisir J. Update on the pharmacology of REM sleep behavior disorder. Neurology. 2006;67(5):742–7. doi: 10.1212/01.wnl.0000233926.47469.73. [DOI] [PubMed] [Google Scholar]
- 190.Kunz D, Mahlberg R. A two-part, double-blind, placebo-controlled trial of exogenous melatonin in REM sleep behaviour disorder. J Sleep Res. 2010;19(4):591–6. doi: 10.1111/j.1365-2869.2010.00848.x. [DOI] [PubMed] [Google Scholar]
- 191.McCarter SJ, Boswell CL, St Louis EK, Dueffert LG, Slocumb N, Boeve BF, et al. Treatment outcomes in REM sleep behavior disorder. Sleep Med. 2013;14(3):237–42. doi: 10.1016/j.sleep.2012.09.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 192.Fantini ML, Gagnon JF, Filipini D, Montplaisir J. The effects of pramipexole in REM sleep behavior disorder. Neurology. 2003;61(10):1418–20. doi: 10.1212/wnl.61.10.1418. [DOI] [PubMed] [Google Scholar]