Skip to main content
Chest logoLink to Chest
. 2015 Jul;148(1):262–273. doi: 10.1378/chest.14-1304

Central Disorders of Hypersomnolence

Focus on the Narcolepsies and Idiopathic Hypersomnia

Zeeshan Khan 1, Lynn Marie Trotti 1,
PMCID: PMC4694150  PMID: 26149554

Abstract

The central disorders of hypersomnolence are characterized by severe daytime sleepiness, which is present despite normal quality and timing of nocturnal sleep. Recent reclassification distinguishes three main subtypes: narcolepsy type 1, narcolepsy type 2, and idiopathic hypersomnia (IH), which are the focus of this review. Narcolepsy type 1 results from loss of hypothalamic hypocretin neurons, while the pathophysiology underlying narcolepsy type 2 and IH remains to be fully elucidated. Treatment of all three disorders focuses on the management of sleepiness, with additional treatment of cataplexy in those patients with narcolepsy type 1. Sleepiness can be treated with modafinil/armodafinil or sympathomimetic CNS stimulants, which have been shown to be beneficial in randomized controlled trials of narcolepsy and, quite recently, IH. In those patients with narcolepsy type 1, sodium oxybate is effective for the treatment of both sleepiness and cataplexy. Despite these treatments, there remains a subset of hypersomnolent patients with persistent sleepiness, in whom alternate therapies are needed. Emerging treatments for sleepiness include histamine H3 antagonists (eg, pitolisant) and possibly negative allosteric modulators of the gamma-aminobutyric acid-A receptor (eg, clarithromycin and flumazenil).


Sleepiness is a common experience, with the prevalence of excessive daytime sleepiness (EDS) occurring at least 3 d/wk ranging from 4% to 21%.1 Such sleepiness may be caused by medical conditions, sleep disorders, illicit and prescribed substances, work and family demands (including shift work), and insufficient sleep time. Insufficient sleep is a particularly common cause of EDS, as more than one-third of Americans are sleep deprived.2

This review focuses on the central disorders of hypersomnolence, a group of sleep disorders characterized by EDS in the absence of disrupted nocturnal sleep or circadian rhythm disorders. The first of these disorders to be comprehensively described was narcolepsy, dating back to a case published in 1880 by Jean Baptiste Gélineau of a 38-year-old wine merchant with > 200 sleep attacks per day.3 Idiopathic hypersomnia (IH) was then detailed by Bedrich Roth in a series of 642 patients seen over 30 years.4 The classification, diagnosis, and treatment of the central disorders of hypersomnolence have evolved considerably since these early descriptions and will be the focus of this review. Together, these hypersomnolence disorders account for substantial morbidity and impairments in quality of life.5-9

Clinical Features, Diagnosis, and Classification

EDS is the cardinal feature of the central disorders of hypersomnolence. It is defined as the “inability to stay awake and alert during major waking episodes of the day, resulting in periods of irrepressible need for sleep or unintended lapses into drowsiness or sleep.”10 It can be confused with fatigue, but while fatigue presents with a lack of energy without inadvertent or excessive sleep, sleepiness implies an increased propensity to sleep. The most recent version of the International Classification of Sleep Disorders, Third Edition (ICSD-3) subdivides the central disorders of hypersomnolence into eight categories (Table 1).10 While insufficient sleep syndrome (ie, sleepiness caused by short sleep times and cured by sleep extension) is categorized as one of the eight hypersomnolence syndromes, insufficient sleep time must be excluded for the other diagnoses. This can be accomplished using patient-completed sleep logs or actigraphic monitoring over a 1- to 2-week period. In this version of the ICSD-3, there are three persistent hypersomnolence disorders not associated with another illness or substance: narcolepsy type 1, narcolepsy type 2, and IH (Table 2).

TABLE 1 ] .

Central Disorders of Hypersomnolence10

Disorders
Narcolepsy type 1
Narcolepsy type 2
Idiopathic hypersomnia
Kleine-Levin syndrome
Hypersomnia due to a medical disorder
Hypersomnia due to a medication or substance
Hypersomnia associated with a psychiatric disorder
Insufficient sleep syndrome

TABLE 2 ] .

International Classification of Sleep Disorders, Third Edition, Diagnostic Criteria10

Narcolepsy Type 1a
Criteria A and B
Narcolepsy Type 2b
All Criteria A-E
Idiopathic Hypersomniac
All Criteria A-F
A. Daily periods of irrepressible need to sleep or daytime lapses into sleep, present for at least 3 mo A. Daily periods of irrepressible need to sleep or daytime lapses into sleep, present for at least 3 mo A. Daily periods of irrepressible need to sleep or daytime lapses into sleep, present for at least 3 mo
B. Either 1 or 2 or both B. Mean sleep latency ≤ 8 min and two or more SOREMPs on MSLT. REM within 15 min of sleep onset on the preceding nocturnal polysomnogram may replace one of the SOREMPs. B. Fewer than two SOREMPs on MSLT (or fewer than one if nocturnal REM latency was ≤ 15 min)
 1. Cataplexy and mean sleep latency ≤ 8 min and two or more SOREMPs on MSLT. REM within 15 min of sleep onset on the preceding nocturnal polysomnogram may replace one of the SOREMPs. C. No cataplexy C. No cataplexy
 2. Low CSF hypocretin-1 concentration (< 110 pg/mL or less than one-third of control values) D. CSF hypocretin-1 concentration has not been measured or CSF hypocretin-1 concentration is ≥ 110 pg/mL or greater than one-third of control values. D. Either 1 or 2 or both
E. The hypersomnolence and/or MSLT findings are not better explained by other causes.  1. Mean sleep latency ≤ 8 min on MSLT
 2. Total 24-h sleep time ≥ 660 min on 24-h polysomnographic monitoring or wrist actigraphy (averaged over ≥ 7 d)
E. Insufficient sleep syndrome is ruled out.
F. The hypersomnolence and/or MSLT findings are not better explained by other causes.

CSF = cerebral spinal fluid; MSLT = multiple sleep latency test; REM = rapid eye movement; SOREMP = sleep-onset rapid eye movement period.

a

Formerly narcolepsy with cataplexy.

b

Formerly narcolepsy without cataplexy.

c

Formerly idiopathic hypersomnia with long sleep time and without long sleep time.

The classic symptom tetrad for narcolepsy is EDS, cataplexy, sleep paralysis, and hallucinations at sleep onset or offset (ie, hypnagogic or hypnopompic hallucinations, respectively). Cataplexy is defined as the sudden loss of muscle tone in response to a strong emotion, most typically when hearing or telling a joke. However, < 10% of patients exhibit all symptoms initially and this may contribute to the average diagnostic delay of 10½ years.11 Cataplexy is present in 65% to 75% of individuals with narcolepsy.12-14 It is quite specific; only rarely will cataplexy or cataplexy-like episodes occur in other disorders, including Coffin-Lowry syndrome, Norrie disease, and Niemann-Pick disease type C.15-19 The presence or absence of cataplexy is a key distinguishing feature between the two types of narcolepsy, which are now recognized to be quite different entities despite their similar nomenclature. Cataplexy is present in narcolepsy type 1 (formerly known as narcolepsy with cataplexy) and absent in narcolepsy type 2 (formerly narcolepsy without cataplexy) (Table 2). Many patients with narcolepsy type 1 have fragmented nocturnal sleep, underscoring the fact that this type of narcolepsy reflects difficulty with state control.20 In essence, patients with narcolepsy type 1 have difficulty remaining awake when desired, but also, at times, with remaining asleep when it is alternately desired.

Cataplexy can also be conceptualized as a problem of state control, such that a feature of rapid eye movement (REM) sleep (ie, the paralysis, or atonia, that typically accompanies normal REM sleep) suddenly intrudes into wakefulness.20 REM behavior disorder, in which patients are able to act out dreams because their motor control during REM more closely resembles that of wakefulness (ie, they have a lack of paralysis, or lack of atonia), is another disorder of sleep-wake state control that is common in narcolepsy type 1.21 Sleep fragmentation may also be a common feature of narcolepsy type 2,22 but is very atypical for IH.

The remaining features of the narcolepsy tetrad, sleep paralysis and hallucinations, are common in patients with either type of narcolepsy, but sleep paralysis also occurs in healthy subjects (5%-40%)23 and these features do not reliably distinguish among the major hypersomnolence syndromes (Table 3).24-38 Patients with IH have EDS but never cataplexy, and have a clinical presentation more similar to those patients with narcolepsy type 2 than type 1. Sleep paralysis and hallucinations are variably present in IH. One-third to two-thirds of patients with IH experience what Roth described as “sleep drunkenness”: a prolonged state after awakening in which motor functions return before full awareness or there is partial return of both.10,32,39 Patients report great difficulty with awakening, requiring multiple alarms or specific procedures to awaken. Most patients with IH (75%) feel unrefreshed after naps, which are long.32 In contrast to narcolepsy type 1, high sleep efficiency (≥ 90%) and occasional spontaneous remission are seen in IH.26

TABLE 3 ] .

Clinical Features of the Narcolepsies and Idiopathic Hypersomnia

Feature Narcolepsy Type 1 Narcolepsy Type 2 Idiopathic Hypersomnia
Excessive daytime sleepiness Present Present Present
Cataplexy Generally present (cataplexy plus characteristic MSLT features, or hypocretin deficiency, are necessary for diagnosis) Absent (by definition) Absent (by definition)
Sleep paralysis Present in 69%a Present in 35%a Present in 20%a
Sleep hallucinations Present in 77%a Present in 42%a Present in 25%a
Tetrad of all four of the above symptoms Present in 42% (although not all present initially)24 Absent Absent
Fragmented nocturnal sleep Significantly lower sleep efficiency than narcolepsy without cataplexy25 or idiopathic hypersomnia26 May be common22 Not typical
REM sleep behavior disorder Present in 45%-61%27; significantly more PSG-measured REM sleep without atonia than in IH28 Significantly more PSG-measured REM sleep without atonia than in IH28 Rate of REM sleep behavior disorder not studied
Sleep drunkenness Rare, but occasionally reported26,29 May be common29 Common
Long nocturnal sleep times Present in 18% of patients with narcolepsy with or without cataplexy30 Present in 18% of patients with narcolepsy with or without cataplexy30 Common
Effect and duration of naps Refreshing, short Unrefreshing (compared with either patients with narcolepsy with cataplexy31 or normal control subjects32), long

IH = idiopathic hypersomnia. See Table 2 legend for expansion of other abbreviations.

a

Frequency estimates for sleep paralysis and hypnogogic hallucinations are compilations from case series reporting on at least two of the groups outlined in this table.29,33-38

Given these overlapping clinical features, the diagnosis of hypersomnolence disorders requires attention to both clinical presentation and sleep testing, especially the multiple sleep latency test (MSLT). The MSLT consists of five 20-min nap opportunities at 2-h intervals.40,41 The fifth nap opportunity is sometimes omitted in cases of narcolepsy where diagnostic criteria have been met after the first four naps.41 A polysomnogram immediately precedes the MSLT to ensure a sufficient amount of sleep (≥ 6 h) and to rule out other sleep disorders, and sleep logs and/or actigraphy are recommended the week before to document habitual sleep times and rule out insufficient sleep. All stimulants and REM-suppressing medications should be discontinued 2 weeks before the test, although in practice, this may sometimes be difficult. The two parameters of most interest are the mean sleep latency (MSL) and the number of sleep-onset REM periods (SOREMPs). The sleep latency is the first epoch of sleep (any stage), and the MSL is the mean across all naps. A SOREMP is the presence of at least one epoch of REM during a nap opportunity. The MSLT is a major factor in current classification of patients with hypersomnolence disorders, such that the number of SOREMPs determines whether a patient with a clinical syndrome of hypersomnolence is classified as having narcolepsy (if they have two or more SOREMPs) or if they have IH (if they have fewer than two SOREMPs) (Table 2).

The MSLT has long been the gold standard for the diagnosis of narcolepsy, but like most diagnostic modalities, is not without flaws. First, neither short MSL nor SOREMPs are specific. Up to 30% of the normal population may have a MSL ≤ 8 min, the current cutoff for the hypersomnolence disorders.42 Multiple SOREMPs can be seen in 3.9% to 9.5% of the general population,43,44 although retest reliability of this (and other) MSLT parameter is poor on a population level.45 Multiple SOREMPs are more common among shift workers and men.44,45 Smaller studies have suggested that multiple SOREMPs may be observed in OSA,46 Prader-Willi syndrome,47 Parkinson disease,48 and myotonic dystrophy.49 Second, the MSLT may not be adequately sensitive, especially for IH. The 8-min cutoff was determined for patients with narcolepsy and extended to IH for “simplicity,” without independent determination.50 This arbitrary cutoff misses 22% to 39% of subjects who otherwise meet clinical criteria for hypersomnia, and up to 71% of those hypersomnolent patients with long sleep times (> 600 min).51,52 Third, while MSLT test-retest reliability is high in patients with narcolepsy with cataplexy restudied within 3 weeks,53 in clinical practice, test-retest reliability of the MSLT in narcolepsy without cataplexy and IH is poor. More than one-half of subjects with these disorders are given a changed diagnosis on repeat testing.54

There are several reasons why the MSLT may not accurately capture hypersomnolence. First, the subjective experience of sleepiness (on the Epworth Sleepiness Scale [ESS]) correlates only modestly with MSL.55,56 Sleepiness is typically experienced as the inability to stay awake when desired, yet the MSLT measures “sleepability,” or the ability to fall asleep on command. These two constructs, while related, are clearly not identical. Furthermore, subjective sleepiness and MSL may not be equivalent because individuals can misperceive electrophysiologic sleep.57 Second, the MSLT is affected by fluctuating physiologic levels of arousal that are distinct from sleepiness.58 The MSLT is also affected by age: In patients with narcolepsy and cataplexy, older age correlates with higher MSL and fewer SOREMPs.59 For all these reasons, it is imperative to interpret the MSLT in clinical context. The ICSD-3 retained diagnostic criteria using MSL, but specified that MSL is best considered as a continuum, with scores < 5 min generally reflecting sleepiness and scores > 10 min generally not.10

There are several important differences when compared with the second edition of the ICSD. The condition of hypersomnolence, or the state of excessive sleepiness, is clearly distinguished from the specific syndrome of hypersomnia. Narcolepsy is now subdivided into narcolepsy type 1 (characterized by either cataplexy and typical MSLT findings or low cerebrospinal fluid [CSF] hypocretin levels) and narcolepsy type 2 (lacking both cataplexy and low CSF hypocretin levels) (Table 3). This departure from prior nomenclature (narcolepsy with and without cataplexy) emphasizes the pathogenic role of hypocretin deficiency. While the diagnosis of narcolepsy still relies on the presence of REM sleep occurring shortly after sleep onset during MSLT (ie, SOREMPs), new criteria allow a nocturnal REM latency ≤ 15 min to count toward the two or more SOREMPs needed. A REM latency ≤ 15 min on a nocturnal polysomnogram is highly specific for narcolepsy with hypocretin deficiency or cataplexy (95%-99%) but not sensitive (36%-58%).60

Additionally, the ICSD-3 no longer distinguishes between IH with and without long sleep time. While the MSLT remains important in the diagnostic framework for IH, there is now a non-MSLT criterion based on measured sleep time of 660 min over 24 h, either through continuous polysomnography (PSG) or actigraphy. This might be expected to better capture the group of patients with a clinical picture of IH but an MSL > 8 min. In patients with hypersomnia with a MSL < 8 min or a documented 24-h sleep time ≥ 660 min, sleep time during 24-h PSG well differentiated patients from control subjects (525 min [SD, ± 87] in control subjects and 695 ± 99 in patients with IH).51 However, in this study, 30 of 105 patients (29%) with suspected IH had both MSL > 8 and sleep time < 660. In a separate cohort of 98 subjects with hypersomnolence (excluding the two subjects with IH and habitual sleep time > 10 h), patients who had a clinical phenotype of IH were indistinguishable on 24-h continuous PSG, regardless of whether they had an MSL < 8 min or > 8 min, and the sum of average night sleep plus average naps did not exceed 660 min in either group (493.9 min in those with MSL > 8 min and 516.8 min in those with MSL < 8 min).61

Optimal diagnostic methods for IH require further study. Given the limited face validity of the MSLT for capturing sleepiness, it has been suggested that the maintenance of wakefulness test (MWT), in which patients are asked to remain awake, may be useful. However, at present, this is not validated for IH or narcolepsy diagnosis. Within the MSLT itself, a longer sustained sleep latency, defined as the latency to either three stage-1 non-REM epochs or a single epoch of any other sleep stage, has been proposed as a possible marker to differentiate IH from the narcolepsies, but further validation is needed.52 Other authors have found that transitions from N1 or wake directly into REM, without passing through N2, are very common in patients with narcolepsy type 1 (either during the MSLT or during the first REM period of the night), are completely absent in IH, and occur with an intermediate frequency in patients with narcolepsy type 2.62,63

The ICSD-3 contains diagnostic criteria for an additional five central hypersomnolence disorders (Table 1). The hypersomnolence syndromes that occur on a recurrent basis, rather than persistently, have now been consolidated into the single diagnosis of Kleine-Levin syndrome. This diagnosis requires at least two episodes of recurrent, time-limited hypersomnia (2 days to 5 weeks), associated with cognitive or perceptual dysfunction, disinhibition, or disordered eating, with return to normal baseline between events.

Epidemiology

The prevalence of narcolepsy with cataplexy is 0.025% to 0.05%.12,64 Globally, the prevalence varies from highest in Japan (0.16%) to lowest in Israel (0.0002%).65 The age of onset in clinical populations appears to be bimodal, with the first peak at 15 years and the second at 35 years,66 although a population-based study demonstrated a single large peak between ages 10 and 19, with gradual tapering off with increasing age.12 There are no population-based prevalence estimates for IH using the second edition of ICSD or ICSD-3 classifications,1 so prevalence estimates are extrapolations from sleep disorders clinics. These estimates of the relative frequency of IH to narcolepsy with cataplexy vary substantially, from 1:10 to greater than 1:1.25,26,29,34,52 This may reflect differing referral patterns, but makes it difficult to conclusively estimate IH prevalence. The age of onset of IH symptoms ranges from the late teens to the mid 30s.67 Unlike narcolepsy, spontaneous remission has been reported in 14% to 25% of patients with IH.51

Pathophysiology

The neuropeptide hypocretin (also called orexin) was first identified in 1998.68,69 Hypocretin is produced in the lateral hypothalamus and is involved in the regulation of feeding, stress response, reward, and the autonomic nervous system.70 Hypocretin is vital for the regulation of the sleep-wake cycle by its influence on the histaminergic, nonadrenergic, serotonergic, and cholinergic systems.71 CSF hypocretin-1 levels are reduced in the majority (90%-95%) of subjects with narcolepsy and typical cataplexy.72 While loss of hypocretin neurons is also seen in 10% to 30% of cases of narcolepsy without cataplexy, most patients with narcolepsy without cataplexy have normal hypocretin levels.22

The loss of hypocretin and development of narcolepsy type 1 involves both genetic and environmental factors, likely resulting from an autoimmune attack on hypocretin neurons in genetically susceptible individuals. The clear genetic predisposition is seen in the 10 to 40 times higher risk of narcolepsy in first-degree relatives of patients. Human leukocyte antigen (HLA) DQB1*06:02 is present in > 85% to 95% of patients with typical cataplexy but is not specific, as it is also present in 40% of cases of narcolepsy without cataplexy and 24% of non-sleepy control subjects.13 Genomewide association studies of narcolepsy have identified several risk alleles in additional genes involved with immune system functioning, including the T-cell receptor α locus (responsible for antigen recognition),73 P2RY11 (receptor expressed in CD8+ cells),74 cathepsin H (antigen processing and presentation on major histocompatibility complex molecules),75 and TNFSF4/OX40L (costimulatory factor for T-cell activation).75

Despite this apparent genetic predisposition to narcolepsy, concordance rates in identical twins are only 25% to 31%,76 implicating substantial environmental or stochastic factors. The occurrence of narcolepsy onset is seasonal (most frequent in April) in China, implicating a variable exposure, possibly infectious.77 Narcolepsy incidence increased threefold to fourfold after the 2009-2010 H1N1 pandemic in China,77 and particular versions of the adjuvanted H1N1 vaccine were associated with narcolepsy onset.78 The increase in narcolepsy incidence after H1N1 vaccination in Europe ranged from a rate ratio of 1.9 (95% CI, 1.1-3.1) in Denmark to 7.5 (95% CI, 5.2-10.7) in Sweden, in the age group 5 to 19 years.79 Other infections might trigger narcolepsy, as suggested by the presence of antistreptococcal antibodies in 65% of patients with narcolepsy within 1 year of disease onset (compared with 26% in age-matched control subjects),80 and the observation that narcolepsy is 5.4 times more common in those individuals who are HLA DQB1*0602 positive with physician-diagnosed streptococcal infection than in individuals who are DQB1*0602 positive without childhood streptococcal infection.81 The combination of HLA association, genetic polymorphisms in immune genes, and apparent triggering of disease by infection or vaccination all suggest an autoimmune basis for hypocretin-deficient narcolepsy, but this has yet to be conclusively demonstrated.82

The pathophysiologies of narcolepsy type 2 and IH are not yet known. A familial component has been proposed, as a family history of EDS is common in patients with IH, more so than in patients with narcolepsy with cataplexy.5,25,83 The HLA DQB1*0602 allele implicated in narcolepsy has been shown to be increased in IH patients in some,35 but not all,34,51 investigations. Japanese subjects with “essential hypersomnia” (defined as the presence of excessive sleepiness in the absence of cataplexy or a condition such as sleep apnea that explains the sleepiness, which appears to be inclusive of the current ICSD-3 entities of narcolepsy type 2 and IH) also have increased positivity for the haplotype DRB1*1501-DQB1*0602.84 An increased frequency of three HLA alleles in linkage disequilibrium—Cw2, DR5, and B27—and a decreased frequency of DRB1*11 have been reported in IH, but not across all studies.51,85,86 A single genomewide association study of Japanese subjects with essential hypersomnia identified risk alleles in three genes: NCKAP5, SPRED1, and CRAT.87 A single nucleotide polymorphism between CPT1B and CHKB, known to be overrepresented in patients with narcolepsy plus cataplexy,88 is also more common in patients with essential hypersomnia than control subjects.84 The role of these genes in predisposing or causing hypersomnolence remains to be determined.

Patients with IH may have low CSF histamine levels,89 although this was not replicated.36 Patients with IH may have higher total serum IgG levels than control subjects (in contrast to patients with narcolepsy with cataplexy who have lower total levels).90 The authors speculated that this finding, as well as the distribution of IgG subclasses in patients with IH, might be related to sleep-associated cytokine production in patients with IH.90

Rye et al9 studied CSF of 32 hypersomnolent patients and found a gain of function within the gamma-aminobutyric acid-A (GABAA) system (ie, the presence of a positive allosteric modulator of GABAA receptors in patients more than control subjects). The effect on GABAA receptors could be reversed in vitro with flumazenil, a negative allosteric modulator of GABAA receptors, and IV flumazenil improved subjective sleepiness and measured vigilance in patients.9 Case reports of two patients treated successfully with sublingual/transdermal9 or subcutaneous91 flumazenil over weeks to years further support the hypothesis that abnormal GABAA receptor activity may be contributing to sleepiness, but further work is needed to establish the possible role of the GABA system in hypersomnolence.

Treatment

In patients with hypersomnolence disorders, the goal of treatment is to curtail daytime sleepiness. In patients with narcolepsy type 1, treatment of cataplexy is also often desired. Nonpharmacologic measures may be helpful in reducing sleepiness in some patients. In particular, for patients with narcolepsy, the combination of scheduled naps and a regular bedtime may reduce daytime sleep time, especially in those patients who remain sleepy despite stimulant medication.92 However, scheduled naps alone appear insufficient to control sleepiness as monotherapy (ie, without wake-promoting medication).93 In patients with idiopathic hypersomnia, in whom naps tend to be long and unrefreshing, scheduling of naps as a treatment strategy tends to be less successful. Support for patient and family through organized patient advocacy and support groups is often reported by patients to be helpful in providing information and in combatting the sometimes negative public perception of people who are sleepy. Patients with narcolepsy, especially with cataplexy, are more likely than normal control subjects or patients with IH to be overweight or obese,94-96 and so management of this comorbidity can be an important part of the treatment plan. OSA is also present in approximately 25% to 30% of patients with narcolepsy, although data are mixed regarding the potential benefit of CPAP on sleepiness in this group.97-99 Patients with central disorders of hypersomnolence are at increased risk of motor vehicle accidents,100,101 and counseling about this risk and the need to avoid driving while sleepy is very important. Regulations regarding driving with sleep disorders vary by state.

There are multiple US Food and Drug Administration (FDA) approved medications for narcolepsy and none for IH; medications for narcolepsy are frequently extended to off-label use in IH. Modafinil, a non-amphetamine, wakefulness-promoting agent, is considered standard therapy for EDS in narcolepsy by the American Academy of Sleep Medicine (AASM) (Table 4).93 A meta-analysis of 1,054 patients with narcolepsy demonstrated that modafinil (200-600 mg/d) improved EDS relative to placebo, decreasing ESS by 2.73 points (95% CI, −3.39 to −2.08), increasing MSL on MSLT by 1.11 min (95% CI, 0.55-1.66), and increasing MSL on the MWT by 2.82 min (95% CI, 2.4-3.24).102 Although both once-daily (morning) dosing and bid (morning and midday) dosing have been evaluated in randomized controlled trials (RCTs), split-dose regimens of modafinil taken bid appear more effective at controlling symptoms into the evening than single morning dosing.103-105 Despite published use of dosages up to 600 mg/d,102,106 the FDA-listed maximum dose in adults is 400 mg. Armodafinil, the longer half-life enantiomer of racemic modafinil, also significantly increases MSL on MWT compared with placebo in narcolepsy.107 Armodafinil is typically dosed once daily (in the morning). Modafinil and armodafinil are also FDA-approved for the treatment of EDS in OSA syndrome and shift-work sleep disorder. Use of modafinil for IH has been based, until recently, on expert consensus,93 but appears to have similar treatment benefit in IH and narcolepsy with cataplexy in clinical use (ESS change, −2.6 ± 5.1 in IH vs −3 ± 5.1 in narcolepsy).108 In the first published RCT for EDS to include patients with IH, modafinil enhanced driving performance, increased sleep latency on MWT, and decreased subjective sleepiness compared with placebo.109 Advantages of modafinil/armodafinil over traditional stimulants include low abuse potential and a generally better side effect profile. They are, however, associated with headache, nausea, and anxiety, which sometimes abate over time. Postmarketing data revealed rare cases of serious or life-threatening rash (Stevens-Johnson syndrome, toxic epidermal necrolysis, and drug rash with eosinophilia and systemic symptoms).110 Although these medications are not FDA approved for pediatric use, they are used off-label in this population.106 Their interaction with oral contraception (decreasing contraception efficacy) is important to consider in women of childbearing potential.

TABLE 4 ] .

Current and Future Treatment Options for Narcolepsy and Idiopathic Hypersomnia

Medication Disorder AASM Recommendationa or Level of Evidence if No Recommendation
For treatment of daytime sleepiness
 Modafinil Narcolepsy Narcolepsy: standard
IH IH: option, but RCT published subsequent to recommendation
 Armodafinil Narcolepsy See recommendation for modafinil
IH
 Sodium oxybate Narcolepsy Standard (for both sleepiness and cataplexy)
 Amphetamine, methamphetamine, dextroamphetamine, methylphenidate Narcolepsy Narcolepsy: guideline
IH IH: option
 Ritanserin (not available in United States) Narcolepsy Option
 Selegiline Narcolepsy Option (for both sleepiness and cataplexy)
 Pitolisant (not available in the United States) Narcolepsy Narcolepsy: RCT published subsequent to recommendation (RCT to evaluate effect on cataplexy is ongoing)
IH IH: clinical case series
 Clarithromycin Narcolepsy type 2 Clinical case series (RCT results pending)
IH Clinical case series (RCT results pending)
 Levothyroxine IH (with long sleep time) Clinical case series
For treatment of cataplexy
 Sodium oxybate Narcolepsy Standard (for both sleepiness and cataplexy)
 Venlafaxine, SSRIs, tricyclic antidepressants, reboxetine (not available in the United States) Narcolepsy Guideline
 Selegiline Narcolepsy Option (for both sleepiness and cataplexy)

AASM = American Academy of Sleep Medicine; RCT = randomized controlled trial; SSRI = selective serotonin reuptake inhibitor. See Table 2 legend for expansion of other abbreviation.

a

AASM recommendations follow these criteria: “Standard” refers to an accepted treatment reflecting high-quality evidence (highest recommendation); “guideline” refers a treatment supported by level 2 or substantial level 3 evidence (middle level of recommendation); “option” refers to a treatment with conflicting (or inconclusive) evidence or expert opinion (lowest level of recommendation).93

Sympathomimetic stimulants such as methylphenidate and dextroamphetamine are also effective for daytime sleepiness, but do have possible adverse psychiatric and cardiovascular effects.111 Amphetamines have been used for narcolepsy since 1935,112 and do increase MSL (from 4.3 to 9.3 min) and decrease errors on driving simulations (from 2.53% to 0.33%).113 Methylphenidate relieves subjective sleepiness and improves the ability to stay awake on the MWT.114 These agents have been given a guideline recommendation for the treatment of EDS by the AASM.93 Clinical series suggest that wake-promoting medications are successful in 62% to 83% of subjects with IH,26,67,85 with a clinically challenging subgroup remaining refractory to these standard treatments.

Sodium oxybate, the sodium salt of γ-hydroxybutryrate, is considered a standard therapy for EDS, cataplexy, and disrupted sleep in narcolepsy by the AASM.93 In meta-analysis, it was superior to placebo in reducing mean weekly cataplexy attacks by 8.5 (95% CI, −15.3 to −1.6), increasing MWT latency by 5.18 min (95% CI, 2.59-7.78), and reducing sleep attacks by 9.65 (95% CI, −17.72 to −1.59).115 Modafinil and sodium oxybate may have additive effects on EDS.116 The nightly divided dose of sodium oxybate can be burdensome for patients. Because of abuse potential and possible adverse effects (ie, deep sedation, respiratory depression), sodium oxybate is dispensed through a central pharmacy after thorough patient education.

Cataplexy can also be treated with REM-suppressing antidepressants, which are often used as first-line agents, although evidence is limited.117 Tricyclic antidepressants have been used for several decades,118 but may have adverse anticholinergic effects. Serotonin-norepinephrine reuptake inhibitors and selective serotonin reuptake inhibitors (SSRIs) may also control cataplexy. Venlafaxine is a preferred cataplexy treatment, considering its benefit to risk ratio.119,120 Tricyclics, SSRIs, and venlafaxine may also treat sleep paralysis and hypnagogic hallucinations.93

The treatment of the central disorders of hypersomnolence during pregnancy is complicated by a lack of available evidence regarding medication safety. The majority of agents are in FDA pregnancy category C, meaning either an absence of both human and animal safety data or evidence for harm in animal studies but absent human studies. Although no published guidelines exist for the treatment of narcolepsy during pregnancy, Thorpy et al121 summarized the animal and human data on these medications, which may be helpful to clinicians in guiding therapy decisions. Pregnancy registries, which compile data from women exposed to specific medications during pregnancy, are currently available for modafinil and armodafinil, and the FDA maintains an updated list of all medications for which such registries exist.122

Novel therapies are under development for narcolepsy and IH. Hypocretin agonists administered via the intranasal route had have some limited success to date.123 Pitolisant, a histamine-3 receptor inverse agonist, stimulates histamine release and promotes wakefulness. In a randomized trial of 95 patients with narcolepsy, it was superior to placebo in reducing ESS and increasing MWT latencies, although noninferiority to modafinil could not be demonstrated.124 Pitolisant has been used with some success in patients with treatment-refractory IH,125 but is not currently available in the United States. Based on the suspected autoimmune pathophysiology of type 1 narcolepsy, individual patients have been given IV immunoglobulin. Pooling data from these published cases, Knudsen et al126 proposed that IV immunoglobulin may be useful for cataplexy and sleepiness in patients whose disease duration at the time of treatment is ≤ 9 months, although they tempered their conclusion, due to lack of a placebo-controlled trial.

Based on the findings that hypersomnolent patients demonstrate a positive allosteric modulator of GABAA receptors in their CSF,9 and that clarithromycin is a negative allosteric modulator of GABAA receptors,127 Trotti et al128 reported the clinical use of clarithromycin in 53 subjects with central hypersomnolence disorders (without cataplexy). Sixty-four percent of these subjects, who had failed an average of 2.6 prior wake-promoting medications, reported improved EDS. Full results from a randomized, placebo-controlled trial of clarithromycin for hypersomnolence are pending publication, but data from the study, published in abstract form, confirmed a significant benefit on subjective sleepiness (a four-point greater reduction in the ESS with clarithromycin than with placebo).129 Low-dose levothyroxine was beneficial in a small series of patients with IH with normal thyroid function.130 Bupropion decreases sleepiness associated with depression131 and, in our experience, may be helpful adjunct therapy in hypersomnolent patients even in the absence of depression.

Conclusions

Over the last 2 decades, there have been major advances in understanding the neurobiology of hypersomnolence. This is especially true for narcolepsy type 1, which now appears to result from a genetic predisposition interacting with environmental factors to trigger loss of hypocretin-containing neurons. Several genetic, immunologic, and biochemical abnormalities have been identified in subjects with narcolepsy type 2 and/or IH, although the full pathophysiology remains to be elucidated. Of the abnormalities documented to date, the CSF constituent that leads to excess GABAA receptor potentiation might have the most immediate treatment implications, but additional work is needed before GABA receptor modulators are considered for routine hypersomnolence treatment. Current diagnostic criteria are helpful but imperfect, and more sensitive and specific tests are needed. Effective treatments are available for many patients, but a treatment-refractory subgroup remains. Further research is essential to understand the biology and optimal management of these disorders.

Acknowledgments

Financial/nonfinancial disclosures: The authors have reported to CHEST that no potential conflicts of interest exist with any companies/organizations whose products or services may be discussed in this article.

Role of sponsors: The sponsor had no role in the design of the study, the collection and analysis of the data, or the preparation of the manuscript.

ABBREVIATIONS

AASM

American Academy of Sleep Medicine

CSF

cerebrospinal fluid

EDS

excessive daytime sleepiness

ESS

Epworth Sleepiness Scale

FDA

US Food and Drug Administration

GABA

gamma-aminobutyric acid

HLA

human leukocyte antigen

ICSD-3

International Classification of Sleep Disorders, Third Edition

IH

idiopathic hypersomnia

MSL

mean sleep latency

MSLT

multiple sleep latency test

MWT

maintenance of wakefulness test

PSG

polysomnography

RCT

randomized controlled trial

REM

rapid eye movement

SOREMP

sleep-onset rapid eye movement period

SSRI

selective serotonin reuptake inhibitor

Footnotes

FUNDING/SUPPORT: This study was supported in part by the National Institutes of Neurological Disorders and Stroke [Grant K23 NS083748 to Dr Trotti].

Reproduction of this article is prohibited without written permission from the American College of Chest Physicians. See online for more details.

References

  • 1.Ohayon MM. From wakefulness to excessive sleepiness: what we know and still need to know. Sleep Med Rev. 2008;12(2):129-141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Centers for Disease Control and Prevention (CDC). Unhealthy sleep-related behaviors—12 states, 2009. MMWR Morb Mortal Wkly Rep. 2011;60(8):233-238. [PubMed] [Google Scholar]
  • 3.Schenck CH, Bassetti CL, Arnulf I, Mignot E. English translations of the first clinical reports on narcolepsy and cataplexy by Westphal and Gélineau in the late 19th century, with commentary. J Clin Sleep Med. 2007;3(3):301-311. [PMC free article] [PubMed] [Google Scholar]
  • 4.Roth B. Narcolepsy and hypersomnia: review and classification of 642 personally observed cases. Schweiz Arch Neurol Neurochir Psychiatr. 1976;119(1):31-41. [PubMed] [Google Scholar]
  • 5.Billiard M, Dauvilliers Y. Idiopathic hypersomnia. Sleep Med Rev. 2001;5(5):349-358. [DOI] [PubMed] [Google Scholar]
  • 6.Ozaki A, Inoue Y, Hayashida K, et al. Quality of life in patients with narcolepsy with cataplexy, narcolepsy without cataplexy, and idiopathic hypersomnia without long sleep time: comparison between patients on psychostimulants, drug-naïve patients and the general Japanese population. Sleep Med. 2012;13(2):200-206. [DOI] [PubMed] [Google Scholar]
  • 7.Ozaki A, Inoue Y, Nakajima T, et al. Health-related quality of life among drug-naïve patients with narcolepsy with cataplexy, narcolepsy without cataplexy, and idiopathic hypersomnia without long sleep time. J Clin Sleep Med. 2008;4(6):572-578. [PMC free article] [PubMed] [Google Scholar]
  • 8.Bayon V, Léger D, Philip P. Socio-professional handicap and accidental risk in patients with hypersomnias of central origin. Sleep Med Rev. 2009;13(6):421-426. [DOI] [PubMed] [Google Scholar]
  • 9.Rye DB, Bliwise DL, Parker K, et al. Modulation of vigilance in the primary hypersomnias by endogenous enhancement of GABAA receptors. Sci Transl Med. 2012;4(161):161ra151. [DOI] [PubMed] [Google Scholar]
  • 10.American Academy of Sleep Medicine. International classification of sleep disorders: diagnostic and coding manual. 3rd ed. Westchester, IL: American Academy of Sleep Medicine; 2014. [Google Scholar]
  • 11.Morrish E, King MA, Smith IE, Shneerson JM. Factors associated with a delay in the diagnosis of narcolepsy. Sleep Med. 2004;5(1):37-41. [DOI] [PubMed] [Google Scholar]
  • 12.Silber MH, Krahn LE, Olson EJ, Pankratz VS. The epidemiology of narcolepsy in Olmsted County, Minnesota: a population-based study. Sleep. 2002;25(2):197-202. [DOI] [PubMed] [Google Scholar]
  • 13.Mignot E, Hayduk R, Black J, Grumet FC, Guilleminault C. HLA DQB1*0602 is associated with cataplexy in 509 narcoleptic patients. Sleep. 1997;20(11):1012-1020. [PubMed] [Google Scholar]
  • 14.Guilleminault C, Mignot E, Partinen M. Controversies in the diagnosis of narcolepsy. Sleep. 1994;17(suppl 8):S1-S6. [DOI] [PubMed] [Google Scholar]
  • 15.Nelson GB, Hahn JS. Stimulus-induced drop episodes in Coffin-Lowry syndrome. Pediatrics. 2003;111(3):e197-e202. [DOI] [PubMed] [Google Scholar]
  • 16.Vossler DG, Wyler AR, Wilkus RJ, Gardner-Walker G, Vlcek BW. Cataplexy and monoamine oxidase deficiency in Norrie disease. Neurology. 1996;46(5):1258-1261. [DOI] [PubMed] [Google Scholar]
  • 17.Kanbayashi T, Abe M, Fujimoto S, et al. Hypocretin deficiency in Niemann-Pick type C with cataplexy. Neuropediatrics. 2003;34(1):52-53. [DOI] [PubMed] [Google Scholar]
  • 18.Smit LS, Lammers GJ, Catsman-Berrevoets CE. Cataplexy leading to the diagnosis of Niemann-Pick disease type C. Pediatr Neurol. 2006;35(1):82-84. [DOI] [PubMed] [Google Scholar]
  • 19.Vankova J, Stepanova I, Jech R, et al. Sleep disturbances and hypocretin deficiency in Niemann-Pick disease type C. Sleep. 2003;26(4):427-430. [DOI] [PubMed] [Google Scholar]
  • 20.Saper CB, Cano G, Scammell TE. Homeostatic, circadian, and emotional regulation of sleep. J Comp Neurol. 2005;493(1):92-98. [DOI] [PubMed] [Google Scholar]
  • 21.Nightingale S, Orgill JC, Ebrahim IO, de Lacy SF, Agrawal S, Williams AJ. The association between narcolepsy and REM behavior disorder (RBD). Sleep Med. 2005;6(3):253-258. [DOI] [PubMed] [Google Scholar]
  • 22.Baumann CR, Mignot E, Lammers GJ, et al. Challenges in diagnosing narcolepsy without cataplexy: a consensus statement. Sleep. 2014;37(6):1035-1042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Bell CC, Dixie-Bell DD, Thompson B. Further studies on the prevalence of isolated sleep paralysis in black subjects. J Natl Med Assoc. 1986;78(7):649-659. [PMC free article] [PubMed] [Google Scholar]
  • 24.Luca G, Haba-Rubio J, Dauvilliers Y, et al. ; European Narcolepsy Network. Clinical, polysomnographic and genome-wide association analyses of narcolepsy with cataplexy: a European Narcolepsy Network study. J Sleep Res. 2013;22(5):482-495. [DOI] [PubMed] [Google Scholar]
  • 25.Takei Y, Komada Y, Namba K, et al. Differences in findings of nocturnal polysomnography and multiple sleep latency test between narcolepsy and idiopathic hypersomnia. Clin Neurophysiol. 2012;123(1):137-141. [DOI] [PubMed] [Google Scholar]
  • 26.Anderson KN, Pilsworth S, Sharples LD, Smith IE, Shneerson JM. Idiopathic hypersomnia: a study of 77 cases. Sleep. 2007;30(10):1274-1281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Dauvilliers Y, Jennum P, Plazzi G. Rapid eye movement sleep behavior disorder and rapid eye movement sleep without atonia in narcolepsy. Sleep Med. 2013;14(8):775-781. [DOI] [PubMed] [Google Scholar]
  • 28.DelRosso LM, Chesson AL, Jr, Hoque R. Characterization of REM sleep without atonia in patients with narcolepsy and idiopathic hypersomnia using AASM scoring manual criteria. J Clin Sleep Med. 2013;9(7):675-680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Bassetti C, Gugger M, Bischof M, et al. The narcoleptic borderland: a multimodal diagnostic approach including cerebrospinal fluid levels of hypocretin-1 (orexin A). Sleep Med. 2003;4(1):7-12. [DOI] [PubMed] [Google Scholar]
  • 30.Vernet C, Arnulf I. Narcolepsy with long sleep time: a specific entity? Sleep. 2009;32(9):1229-1235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Bruck D, Parkes JD. A comparison of idiopathic hypersomnia and narcolepsy-cataplexy using self report measures and sleep diary data. J Neurol Neurosurg Psychiatry. 1996;60(5):576-578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Vernet C, Leu-Semenescu S, Buzare MA, Arnulf I. Subjective symptoms in idiopathic hypersomnia: beyond excessive sleepiness. J Sleep Res. 2010;19(4):525-534. [DOI] [PubMed] [Google Scholar]
  • 33.Kanbayashi T, Inoue Y, Chiba S, et al. CSF hypocretin-1 (orexin A) concentrations in narcolepsy with and without cataplexy and idiopathic hypersomnia. J Sleep Res. 2002;11(1):91-93. [DOI] [PubMed] [Google Scholar]
  • 34.Sasai T, Inoue Y, Komada Y, Sugiura T, Matsushima E. Comparison of clinical characteristics among narcolepsy with and without cataplexy and idiopathic hypersomnia without long sleep time, focusing on HLA-DRB1(*)1501/DQB1(*)0602 finding. Sleep Med. 2009;10(9):961-966. [DOI] [PubMed] [Google Scholar]
  • 35.Coelho FM, Pradella-Hallinan M, Predazzoli Neto M, Bittencourt LR, Tufik S. Prevalence of the HLA-DQB1*0602 allele in narcolepsy and idiopathic hypersomnia patients seen at a sleep disorders outpatient unit in São Paulo. Rev Bras Psiquiatr. 2009;31(1):10-14. [DOI] [PubMed] [Google Scholar]
  • 36.Dauvilliers Y, Delallée N, Jaussent I, et al. Normal cerebrospinal fluid histamine and tele-methylhistamine levels in hypersomnia conditions. Sleep. 2012;35(10):1359-1366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Dauvilliers Y, Baumann CR, Carlander B, et al. CSF hypocretin-1 levels in narcolepsy, Kleine-Levin syndrome, and other hypersomnias and neurologcial conditions. J Neurol Neurosurg Psychiatry. 2003;74(12):1667-1673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Heier MS, Evsiukova T, Vilming S, Gjerstad MD, Schrader H, Gautvik K. CSF hypocretin-1 levels and clinical profiles in narcolepsy and idiopathic CNS hypersomnia and Norway. Sleep. 2007;30(8):969-973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Roth B, Nevsimalova S, Rechtschaffen A. Hypersomnia with “sleep drunkenness.” Arch Gen Psychiatry. 1972;26(5):456-462. [DOI] [PubMed] [Google Scholar]
  • 40.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]
  • 41.Littner MR, Kushida C, Wise M, et al. ; Standards of Practice Committee of the American Academy of Sleep Medicine. Practice parameters for clinical use of the multiple sleep latency test and the maintenance of wakefulness test. Sleep. 2005;28(1):113-121. [DOI] [PubMed] [Google Scholar]
  • 42.American Academy of Sleep Medicine. International classification of sleep disorders: diagnostic and coding manual. 2nd ed. Westchester, IL: American Academy of Sleep Medicine; 2005. [Google Scholar]
  • 43.Singh M, Drake CL, Roth T. The prevalence of multiple sleep-onset REM periods in a population-based sample. Sleep. 2006;29(7):890-895. [DOI] [PubMed] [Google Scholar]
  • 44.Mignot E, Lin L, Finn L, et al. Correlates of sleep-onset REM periods during the Multiple Sleep Latency Test in community adults. Brain. 2006;129(pt 6):1609-1623. [DOI] [PubMed] [Google Scholar]
  • 45.Goldbart A, Peppard P, Finn L, et al. Narcolepsy and predictors of positive MSLTs in the Wisconsin Sleep Cohort. Sleep. 2014;37(6):1043-1051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Chervin RD, Aldrich MS. Sleep onset REM periods during multiple sleep latency tests in patients evaluated for sleep apnea. Am J Respir Crit Care Med. 2000;161(2 pt 1):426-431. [DOI] [PubMed] [Google Scholar]
  • 47.Helbing-Zwanenburg B, Kamphuisen HA, Mourtazaev MS. The origin of excessive daytime sleepiness in the Prader-Willi syndrome. J Intellect Disabil Res. 1993;37(pt 6):533-541. [DOI] [PubMed] [Google Scholar]
  • 48.Rye DB, Bliwise DL, Dihenia B, Gurecki P. FAST TRACK: daytime sleepiness in Parkinson’s disease. J Sleep Res. 2000;9(1):63-69. [DOI] [PubMed] [Google Scholar]
  • 49.Yu H, Laberge L, Jaussent I, et al. Daytime sleepiness and REM sleep characteristics in myotonic dystrophy: a case-control study. Sleep. 2011;34(2):165-170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Billiard M. Diagnosis of narcolepsy and idiopathic hypersomnia. An update based on the International classification of sleep disorders, 2nd edition. Sleep Med Rev. 2007;11:377-388. [DOI] [PubMed] [Google Scholar]
  • 51.Vernet C, Arnulf I. Idiopathic hypersomnia with and without long sleep time: a controlled series of 75 patients. Sleep. 2009;32(6):753-759. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Pizza F, Vandi S, Detto S, et al. Different sleep onset criteria at the multiple sleep latency test (MSLT): an additional marker to differentiate central nervous system (CNS) hypersomnias. J Sleep Res. 2011;20(1 pt 2):250-256. [DOI] [PubMed] [Google Scholar]
  • 53.Folkerts M, Rosenthal L, Roehrs T, et al. The reliability of the diagnostic features in patients with narcolepsy. Biol Psychiatry. 1996;40(3):208-214. [DOI] [PubMed] [Google Scholar]
  • 54.Trotti LM, Staab BA, Rye DB. Test-retest reliability of the multiple sleep latency test in narcolepsy without cataplexy and idiopathic hypersomnia. J Clin Sleep Med. 2013;9(8):789-795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Chervin RD, Aldrich MS, Pickett R, Guilleminault C. Comparison of the results of the Epworth Sleepiness Scale and the Multiple Sleep Latency Test. J Psychosom Res. 1997;42(2):145-155. [DOI] [PubMed] [Google Scholar]
  • 56.Olson LG, Cole MF, Ambrogetti A. Correlations among Epworth Sleepiness Scale scores, multiple sleep latency tests and psychological symptoms. J Sleep Res. 1998;7(4):248-253. [DOI] [PubMed] [Google Scholar]
  • 57.Weigand D, Michael L, Schulz H. When sleep is perceived as wakefulness: an experimental study on state perception during physiological sleep. J Sleep Res. 2007;16(4):346-353. [DOI] [PubMed] [Google Scholar]
  • 58.Bonnet MH. ACNS clinical controversy: MSLT and MWT have limited clinical utility. J Clin Neurophysiol. 2006;23(1):50-58. [DOI] [PubMed] [Google Scholar]
  • 59.Dauvilliers Y, Gosselin A, Paquet J, Touchon J, Billiard M, Montplaisir J. Effect of age on MSLT results in patients with narcolepsy-cataplexy. Neurology. 2004;62(1):46-50. [DOI] [PubMed] [Google Scholar]
  • 60.Andlauer O, Moore H, Jouhier L, et al. Nocturnal rapid eye movement sleep latency for identifying patients with narcolepsy/hypocretin deficiency. JAMA Neurol. 2013;70(7):891-902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Pizza F, Moghadam KK, Vandi S, et al. Daytime continuous polysomnography predicts MSLT results in hypersomnias of central origin. J Sleep Res. 2013;22(1):32-40. [DOI] [PubMed] [Google Scholar]
  • 62.Drakatos P, Kosky CA, Higgins SE, Muza RT, Williams AJ, Leschziner GD. First rapid eye movement sleep periods and sleep-onset rapid eye movement periods in sleep-stage sequencing of hypersomnias. Sleep Med. 2013;14(9):897-901. [DOI] [PubMed] [Google Scholar]
  • 63.Drakatos P, Suri A, Higgins SE, et al. Sleep stage sequence analysis of sleep onset REM periods in the hypersomnias. J Neurol Neurosurg Psychiatry. 2013;84(2):223-227. [DOI] [PubMed] [Google Scholar]
  • 64.Longstreth WT, Jr, Koepsell TD, Ton TG, Hendrickson AF, van Belle G. The epidemiology of narcolepsy. Sleep. 2007;30(1):13-26. [DOI] [PubMed] [Google Scholar]
  • 65.Dauvilliers Y, Arnulf I, Mignot E. Narcolepsy with cataplexy. Lancet. 2007;369(9560):499-511. [DOI] [PubMed] [Google Scholar]
  • 66.Dauvilliers Y, Montplaisir J, Molinari N, et al. Age at onset of narcolepsy in two large populations of patients in France and Quebec. Neurology. 2001;57(11):2029-2033. [DOI] [PubMed] [Google Scholar]
  • 67.Ali M, Auger RR, Slocumb NL, Morgenthaler TI. Idiopathic hypersomnia: clinical features and response to treatment. J Clin Sleep Med. 2009;5(6):562-568. [PMC free article] [PubMed] [Google Scholar]
  • 68.de Lecea L, Kilduff TS, Peyron C, et al. The hypocretins: hypothalamus-specific peptides with neuroexcitatory activity. Proc Natl Acad Sci USA. 1998;95(1):322-327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Sakurai T, Amemiya A, Ishii M, et al. Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior. Cell. 1998;92(4):573-585. [DOI] [PubMed] [Google Scholar]
  • 70.Inutsuka A, Yamanaka A. The physiological role of orexin/hypocretin neurons in the regulation of sleep/wakefulness and neuroendocrine functions. Front Endocrinol (Lausanne). 2013;4:18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Mieda M, Tsujino N, Sakurai T. Differential roles of orexin receptors in the regulation of sleep/wakefulness. Front Endocrinol (Lausanne). 2013;4:57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Bourgin P, Zeitzer JM, Mignot E. CSF hypocretin-1 assessment in sleep and neurological disorders. Lancet Neurol. 2008;7(7):649-662. [DOI] [PubMed] [Google Scholar]
  • 73.Hallmayer J, Faraco J, Lin L, et al. Narcolepsy is strongly associated with the T-cell receptor alpha locus. Nat Genet. 2009;41(6):708-711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Kornum BR, Kawashima M, Faraco J, et al. Common variants in P2RY11 are associated with narcolepsy. Nat Genet. 2011;43(1):66-71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Faraco J, Lin L, Kornum BR, et al. ImmunoChip study implicates antigen presentation to T cells in narcolepsy. PLoS Genet. 2013;9(2):e1003270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Mignot E. Genetic and familial aspects of narcolepsy. Neurology. 1998;50(2 suppl 1):S16-S22. [DOI] [PubMed] [Google Scholar]
  • 77.Han F, Lin L, Warby SC, et al. Narcolepsy onset is seasonal and increased following the 2009 H1N1 pandemic in China. Ann Neurol. 2011;70(3):410-417. [DOI] [PubMed] [Google Scholar]
  • 78.Dauvilliers Y, Montplaisir J, Cochen V, et al. Post-H1N1 narcolepsy-cataplexy. Sleep. 2010;33(11):1428-1430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Wijnans L, Lecomte C, de Vries C, et al. The incidence of narcolepsy in Europe: before, during, and after the influenza A(H1N1)pdm09 pandemic and vaccination campaigns. Vaccine. 2013;31(8):1246-1254. [DOI] [PubMed] [Google Scholar]
  • 80.Aran A, Lin L, Nevsimalova S, et al. Elevated anti-streptococcal antibodies in patients with recent narcolepsy onset. Sleep. 2009;32(8):979-983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Koepsell TD, Longstreth WT, Ton TG. Medical exposures in youth and the frequency of narcolepsy with cataplexy: a population-based case-control study in genetically predisposed people. J Sleep Res. 2010;19(1 pt 1):80-86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Mahlios J, De la Herrán-Arita AK, Mignot E. The autoimmune basis of narcolepsy. Curr Opin Neurobiol. 2013;23(5):767-773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Billiard M, Merle C, Carlander B, Ondze B, Alvarez D, Besset A. Idiopathic hypersomnia. Psychiatry Clin Neurosci. 1998;52(2):125-129. [DOI] [PubMed] [Google Scholar]
  • 84.Miyagawa T, Honda M, Kawashima M, et al. Polymorphism located between CPT1B and CHKB, and HLA-DRB1*1501-DQB1*0602 haplotype confer susceptibility to CNS hypersomnias (essential hypersomnia). PLoS ONE. 2009;4(4):e5394. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Bassetti C, Aldrich MS. Idiopathic hypersomnia. A series of 42 patients. Brain. 1997;120(pt 8):1423-1435. [DOI] [PubMed] [Google Scholar]
  • 86.Poirier G, Montplaisir J, Décary F, Momège D, Lebrun A. HLA antigens in narcolepsy and idiopathic central nervous system hypersomnolence. Sleep. 1986;9(1 pt 2):153-158. [DOI] [PubMed] [Google Scholar]
  • 87.Khor SS, Miyagawa T, Toyoda H, et al. Genome-wide association study of HLA-DQB1*06:02 negative essential hypersomnia. PeerJ. 2013;1:e66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Miyagawa T, Kawashima M, Nishida N, et al. Variant between CPT1B and CHKB associated with susceptibility to narcolepsy. Nat Genet. 2008;40(11):1324-1328. [DOI] [PubMed] [Google Scholar]
  • 89.Kanbayashi T, Kodama T, Kondo H, et al. CSF histamine contents in narcolepsy, idiopathic hypersomnia and obstructive sleep apnea syndrome. Sleep. 2009;32(2):181-187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Tanaka S, Honda M. IgG abnormality in narcolepsy and idiopathic hypersomnia. PLoS ONE. 2010;5(3):e9555. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Kelty E, Martyn V, O’Neil G, Hulse G. Use of subcutaneous flumazenil preparations for the treatment of idiopathic hypersomnia: a case report. J Psychopharmacol. 2014;28(7):703-706. [DOI] [PubMed] [Google Scholar]
  • 92.Rogers AE, Aldrich MS, Lin X. A comparison of three different sleep schedules for reducing daytime sleepiness in narcolepsy. Sleep. 2001;24(4):385-391. [DOI] [PubMed] [Google Scholar]
  • 93.Morgenthaler TI, Kapur VK, Brown T, et al. ; Standards of Practice Committee of the American Academy of Sleep Medicine. Practice parameters for the treatment of narcolepsy and other hypersomnias of central origin. Sleep. 2007;30(12):1705-1711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Kok SW, Overeem S, Visscher TL, et al. Hypocretin deficiency in narcoleptic humans is associated with abdominal obesity. Obes Res. 2003;11(9):1147-1154. [DOI] [PubMed] [Google Scholar]
  • 95.Kotagal S, Krahn LE, Slocumb N. A putative link between childhood narcolepsy and obesity. Sleep Med. 2004;5(2):147-150. [DOI] [PubMed] [Google Scholar]
  • 96.Sonka K, Kemlink D, Busková J, et al. Obesity accompanies narcolepsy with cataplexy but not narcolepsy without cataplexy. Neuroendocrinol Lett. 2010;31(5):631-634. [PubMed] [Google Scholar]
  • 97.Sansa G, Iranzo A, Santamaria J. Obstructive sleep apnea in narcolepsy. Sleep Med. 2010;11(1):93-95. [DOI] [PubMed] [Google Scholar]
  • 98.Pataka AD, Frangulyan RR, Mackay TW, Douglas NJ, Riha RL. Narcolepsy and sleep-disordered breathing. Eur J Neurol. 2012;19(5):696-702. [DOI] [PubMed] [Google Scholar]
  • 99.Pizza F, Tartarotti S, Poryazova R, Baumann CR, Bassetti CL. Sleep-disordered breathing and periodic limb movements in narcolepsy with cataplexy: a systematic analysis of 35 consecutive patients. Eur Neurol. 2013;70(1-2):22-26. [DOI] [PubMed] [Google Scholar]
  • 100.Aldrich MS. Automobile accidents in patients with sleep disorders. Sleep. 1989;12(6):487-494. [DOI] [PubMed] [Google Scholar]
  • 101.Philip P, Sagaspe P, Lagarde E, et al. Sleep disorders and accidental risk in a large group of regular registered highway drivers. Sleep Med. 2010;11(10):973-979. [DOI] [PubMed] [Google Scholar]
  • 102.Golicki D, Bala MM, Niewada M, Wierzbicka A. Modafinil for narcolepsy: systematic review and meta-analysis. Med Sci Monit. 2010;16(8):RA177-RA186. [PubMed] [Google Scholar]
  • 103.Broughton RJ, Fleming JA, George CF, et al. Randomized, double-blind, placebo-controlled crossover trial of modafinil in the treatment of excessive daytime sleepiness in narcolepsy. Neurology. 1997;49(2):444-451. [DOI] [PubMed] [Google Scholar]
  • 104.US Modafinil in Narcolepsy Multicenter Study Group. Randomized trial of modafinil for the treatment of pathological somnolence in narcolepsy. Ann Neurol. 1998;43(1):88-97. [DOI] [PubMed] [Google Scholar]
  • 105.Schwartz JR, Feldman NT, Bogan RK. Dose effects of modafinil in sustaining wakefulness in narcolepsy patients with residual evening sleepiness. J Neuropsychiatry Clin Neurosci. 2005;17(3):405-412. [DOI] [PubMed] [Google Scholar]
  • 106.Lecendreux M, Bruni O, Franco P, et al. Clinical experience suggests that modafinil is an effective and safe treatment for paediatric narcolepsy. J Sleep Res. 2012;21(4):481-483. [DOI] [PubMed] [Google Scholar]
  • 107.Harsh JR, Hayduk R, Rosenberg R, et al. The efficacy and safety of armodafinil as treatment for adults with excessive sleepiness associated with narcolepsy. Curr Med Res Opin. 2006;22(4):761-774. [DOI] [PubMed] [Google Scholar]
  • 108.Lavault S, Dauvilliers Y, Drouot X, et al. Benefit and risk of modafinil in idiopathic hypersomnia vs. narcolepsy with cataplexy. Sleep Med. 2011;12(6):550-556. [DOI] [PubMed] [Google Scholar]
  • 109.Philip P, Chaufton C, Taillard J, et al. Modafinil improves real driving performance in patients with hypersomnia: a randomized double-blind placebo-controlled crossover clinical trial. Sleep. 2014;37(3):483-487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.US Food and Drug Administration. Modafinil safety alert, 2007. Food and Drug Administration website. http://www.fda.gov/Safety/MedWatch/SafetyInformation/SafetyAlertsforHumanMedicalProducts/ucm152701.htm. Accessed November 4, 2014.
  • 111.Auger RR, Goodman SH, Silber MH, Krahn LE, Pankratz VS, Slocumb NL. Risks of high-dose stimulants in the treatment of disorders of excessive somnolence: a case-control study. Sleep. 2005;28(6):667-672. [DOI] [PubMed] [Google Scholar]
  • 112.Heal DJ, Smith SL, Gosden J, Nutt DJ. Amphetamine, past and present—a pharmacological and clinical perspective. J Psychopharmacol. 2013;27(6):479-496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Mitler MM, Hajdukovic R, Erman MK. Treatment of narcolepsy with methamphetamine. Sleep. 1993;16(4):306-317. [PMC free article] [PubMed] [Google Scholar]
  • 114.Mitler MM, Hajdukovic R, Erman M, Koziol JA. Narcolepsy. J Clin Neurophysiol. 1990;7(1):93-118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Alshaikh MK, Tricco AC, Tashkandi M, Mamdani M, Straus SE, BaHammam AS. Sodium oxybate for narcolepsy with cataplexy: systematic review and meta-analysis. J Clin Sleep Med. 2012;8(4):451-458. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Black J, Houghton WC. Sodium oxybate improves excessive daytime sleepiness in narcolepsy. Sleep. 2006;29(7):939-946. [DOI] [PubMed] [Google Scholar]
  • 117.Vignatelli L, D’Alessandro R, Candelise L. Antidepressant drugs for narcolepsy. Cochrane Database Syst Rev. 2008;(1):CD003724. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Akimoto H, Honda Y, Takahashi Y. Pharmacotherapy in narcolepsy. Dis Nerv Syst. 1960;21:704-706. [PubMed] [Google Scholar]
  • 119.Mignot EJ. A practical guide to the therapy of narcolepsy and hypersomnia syndromes. Neurotherapeutics. 2012;9(4):739-752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Lopez R, Dauvilliers Y. Pharmacotherapy options for cataplexy. Expert Opin Pharmacother. 2013;14(7):895-903. [DOI] [PubMed] [Google Scholar]
  • 121.Thorpy M, Zhao CG, Dauvilliers Y. Management of narcolepsy during pregnancy. Sleep Med. 2013;14(4):367-376. [DOI] [PubMed] [Google Scholar]
  • 122.US Food and Drug Administration. List of pregnancy exposure registries. Food and Drug Administration website. http://www.fda.gov/ScienceResearch/SpecialTopics/WomensHealthResearch/ucm134848.htm#Specific_Medical_Products. Accessed August 20, 2014.
  • 123.Baier PC, Hallschmid M, Seeck-Hirschner M, et al. Effects of intranasal hypocretin-1 (orexin A) on sleep in narcolepsy with cataplexy. Sleep Med. 2011;12(10):941-946. [DOI] [PubMed] [Google Scholar]
  • 124.Dauvilliers Y, Bassetti C, Lammers GJ, et al. ; HARMONY I study group. Pitolisant versus placebo or modafinil in patients with narcolepsy: a double-blind, randomised trial. Lancet Neurol. 2013;12(11):1068-1075. [DOI] [PubMed] [Google Scholar]
  • 125.Leu-Semenescu S, Nittur N, Golmard J-L, Arnulf I. Effects of pitolisant, a histamine H3 inverse agonist, in drug-resistant idiopathic and symptomatic hypersomnia: a chart review. Sleep Med. 2014;15(6):681-687. [DOI] [PubMed] [Google Scholar]
  • 126.Knudsen S, Mikkelsen JD, Bang B, Gammeltoft S, Jennum PJ. Intravenous immunoglobulin treatment and screening for hypocretin neuron-specific autoantibodies in recent onset childhood narcolepsy with cataplexy. Neuropediatrics. 2010;41(5):217-222. [DOI] [PubMed] [Google Scholar]
  • 127.Garcia PS, Jenkins A. Inhibition of the GABA(A) receptor by a macrolide but not by a lincosamide antibiotic. Proceedings of the 2009 Annual Meeting of the American Society of Anesthesiologists. October 17-21, 2009; New Orlean, LA; abstract number A1385. American Society of Anesthesiologists website. http://www.asaabstracts.com/strands/asaabstracts/abstract.htm;jsessionid=A3851932B5D3E948A03003B172D5E649?year=2009&index=10&absnum=477. Accessed May 29, 2014.
  • 128.Trotti LM, Saini P, Freeman AA, et al. Improvement in daytime sleepiness with clarithromycin in patients with GABA-related hypersomnia: clinical experience. J Psychopharmacol. 2013;28(7):697-702. [DOI] [PubMed] [Google Scholar]
  • 129.Trotti LM, Stout AK, Saini P, et al. Clarithromycin reduces sleepiness and improves vigilance in patients with central nervous system hypersomnias. Sleep. 2012;35(abstract supplement):A276-A277. [Google Scholar]
  • 130.Shinno H, Inami Y, Inagaki T, et al. Successful treatment with levothyroxine for idiopathic hypersomnia patients with subclinical hypothyroidism. Gen Hosp Psychiatry. 2009;31(2):190-193. [DOI] [PubMed] [Google Scholar]
  • 131.Cooper JA, Tucker VL, Papakostas GI. Resolution of sleepiness and fatigue: a comparison of bupropion and selective serotonin reuptake inhibitors in subjects with major depressive disorder achieving remission at doses approved in the European Union. J Psychopharmacol. 2014;28(2):118-124. [DOI] [PubMed] [Google Scholar]

Articles from Chest are provided here courtesy of American College of Chest Physicians

RESOURCES