Primary Hypersomnia

Back

Background

In 1966, William Dement proposed that patients with excessive daytime sleepiness, but without cataplexy, sleep paralysis, or sleep-onset rapid eye movement (REM), should not be considered narcoleptic.[3] In 1972, Roth et al described a type of hypersomnia with sleep drunkenness that consists of difficulty coming to complete wakefulness, confusion, disorientation, poor motor coordination, and slowness, accompanied by deep and prolonged sleep.[4] The abrupt sleep attacks seen in classic narcolepsy are not present in this disorder.

Primary hypersomnia, idiopathic hypersomnia (ICSD-3), and hypersomnolence disorder (DSM-5) refer to a central disorder of hypersomnia. The diagnostic criteria have changed over time and specifics differ depending on the organization. Similarities from all, however, include daily periods of irresistible sleep or daytime lapses into sleep, absence of cataplexy, and that the hypersomnolence is not better accounted for by either insufficient sleep or by another sleep disorder. In comparison, narcolepsy is characterized by well-defined clinical, polysomnographic, and immunogenetic features.[5, 6]

Classification

In the literature, 3 possible subgroups of idiopathic CNS hypersomnia have been suggested.

Subgroup I

These patients have a positive family history, and associated clinical symptoms suggest dysfunction of the autonomic nervous system. These symptoms include headache, syncope, orthostatic hypotension, and peripheral vasoconstriction (cold hands and feet).

Subgroup II

This group includes patients who had a viral infection associated with neurologic symptoms, such as Guillain-Barré syndrome, infectious mononucleosis, or atypical viral pneumonia. Even after their infectious disease resolves, these patients continue to require significantly more nocturnal sleep and continue to feel very tired.

Although initially these patients are fatigued, they subsequently have difficulty differentiating fatigue from sleepiness. To fight tiredness, these patients nap and eventually present with complaints of excessive daytime sleepiness. Analysis of cerebral spinal fluid demonstrates moderate lymphocytosis (30-50 cells/µL with mild to moderate elevation in protein).

Subgroup III

These patients do not have a positive family or viral infection history, and the cause of the disorder truly is idiopathic.

Diagnostic criteria (DSM-5 and ICSD-3)

The specific DSM-5 criteria for hypersomnolence disorder are as follows:

In addition, hypersomnolence disorder is specified by duration: acute (less than 1 month), subacute (1–3 months), persistent (more than 3 months); and by the severity based on degree of difficulty maintaining daytime alertness: mild (1–2 days a week), moderate (3–4 days a week), severe (5–7 days a week).

The American Sleep Disorders Association’s International Classification of Sleep Disorders, Third Edition (ICSD-3) has redefined the criteria of idiopathic hypersomnia to include varied presentations under the same diagnosis as opposed to distinguishing two separate forms (with and without long sleep time) that were characteristic of the ICSD-2 definition.  

ICSD-3 classifies “Central disorders of hypersomolence” into ‘primary’ and ‘secondary’ groupings, with narcolepsy type 1 (NT1), narcolepsy type 2 (NT2), idiopathic hypersomnia (IH), and Kline-Levin syndrome (KLS) making up the primary disorders. The secondary disorders include hypersomnia due to a medical or psychiatric disorder, due to a drug or substance, and lastly, insufficient sleep syndrome (ISS).

ISCD-3 Central Disorders of Hypersomnolence



View Table

See Table

Recurrent primary hypersomnia

Kleine-Levin syndrome (KLS) is a rare disorder that starts during adolescence and has a male gender preference. The patients have recurrent episodes of hypersomnia, which are often associated with compulsive overeating and hypersexuality.[9] The periods of hypersomnia occur for days to weeks at a time and recur several times a year. In between the symptomatic periods, the patients have normal sleep requirements and do not have excessive daytime sleepiness. Some patients may develop symptoms of irritability, impulsive behavior, depersonalization, hallucinations, depression, and confusion. The etiology of this disorder is not known, but genetic factors are believed to contribute, citing 2% to 5% of cases are of multiplex family origin.[70, 73, 95, 96, 97, 98] Metabolic, inflammatory, and autoimmune etiologies are suspected, though not yet confirmed.[10, 11, 62]

The disorder mainly affects males (68%). The median age of onset is 15 years (range, 4–82 years; 81% during the second decade), and the syndrome may last up to 8 years. The episodes recur every 3–4 months and may last up to 10 days, but they may last longer in women. (See Epidemiology.)

KLS may be precipitated by infections (72%), alcohol consumption (23%), sleep deprivation (22%), unusual stress (20%), physical exertion (19%), traveling (10%), head trauma (9%), and marijuana use (6%).  Symptoms of infection-triggered KLS generally occur shortly after onset of fever (3 to 5 days).[63, 64, 65, 66]

ISCD-3 diagnostic criteria for KLS are as follows:[2]

  1. The patient experiences at least two recurrent episodes of excessive sleepiness of 2 days to 5 weeks duration.
  2. Episodes recur usually more than once a year and at least once every 18 months.
  3. The patient exhibits normal alertness, cognitive function, behavior, and mood between episodes.
  4. The patient must demonstrate at least one of the following during episodes:  cognitive dysfunction, altered perception, eating disorder (anorexia or hyperphagia), disinhibited behavior (such as hypersexuality).
  5. The hypersomnia and related symptoms are not better explained by the other sleep disorders, other medical neurologic, or psychiatric disorders (especially bipolar disorder), or use of drugs or medications.

Characteristic symptoms of KLS include the following:[11, 64, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92]

Sleep symptoms:

Cognitive changes:

Neuropsychiatric symptoms:

Common symptoms:

Less common symptoms:

Clinical subtypes

KLS may be mild, moderate, or severe. When episodes consistently occur with a temporal relation to menstruation, it is referred to as subtype “menstrual/menstruation-related hypersomnia” and is extremely rare (18 known cases worldwide).[69, 93, 94, 99, 100, 101]   Menstrual-related hypersomnia is diagnosed when excessive daytime sleepiness occurs on a periodic basis over a few days preceding menstruation.[12] It is assumed that the symptoms follow hormonal changes, but the etiology of the syndrome, as well as its prevalence and course, are virtually unknown.

The ICSD-3 classifies KLS as a recurrent hypersomnia. DSM-5 refers to the recurrent hypersomnia as seen in KLS as an “other specified hypersomnolence disorder, brief-duration hypersomnolence."[60, 61]

Etiology

Hypersomnolence is an idiopathic disorder. Although head injury or viral infections can cause a disorder resembling primary hypersomnia, the true causes for most cases remain unknown. No genetic, environmental, or other predisposition has been identified.[6]  Rye et al postulated that there is a naturally occurring “somnogen” in the CSF of those with hypersomnolence that potentiated the inhibitory effects of GABAA in an in vitro setting.[48]

Excessive daytime sleepiness has been described in a subset of patients following viral illnesses such as Guillain-Barré syndrome, hepatitis, mononucleosis, and atypical viral pneumonia. Familial cases associated with HLA-Cw2, -Cw3, -DR5, -DR11, -DQ1, and –DQ3 genotypes have also been reported, and it is known that there are overlapping features found in both idiopathic hypersomnia and narcolepsy, though no consistent findings are agreed upon. HLA typing does not currently play a role in diagnosis of idiopathic hypersomnia.[13, 36, 40, 41, 42, 63] However, the majority of patients diagnosed with idiopathic hypersomnia have neither a positive family history nor a past medical history of viral illnesses.

A recent study highlighted expression dynamics of circadian clock genes in dermal fibroblasts, in which 10 patients with idiopathic hypersomnia were compared to healthy controls. They found the rhythmically expressed BMAL1, PER1, and PER2 were expressed less in cells from idiopathic hypersomnia patients over two circadian periods, and that the overall BMAL1 expression was reduced significantly.[43, 63]

In experimental animal studies, destruction of the nonadrenergic neurons of the rostral third of the locus ceruleus complex has produced hypersomnia. While trauma has been associated with excessive daytime sleepiness in a case series, cerebrospinal fluid (CSF) analysis for specific neurotransmitter metabolites did not differentiate patients with posttraumatic excessive daytime sleepiness from patients with narcolepsy or other patients with excessive daytime sleepiness.[14] Injury to the adrenergic neurons at the bundle of isthmus has led to hypersomnia associated with a proportional increase of both NREM and REM sleep.[15]

Montplaisir et al found decreased dopamine and indoleacetic acid in both narcolepsy and idiopathic hypersomnia patients.[44, 63]

Faull et al discovered dopaminergic dysregulation in narcolepsy and noradrenergic dysregulation in idiopathic hypersomnia.[21]  

This evidence suggests the possibility of aminergic arousal system dysfunction in idiopathic hypersomnia. Feline studies have shown hypersomnia and monoamine dysregulation can be induced reproducibly via lesioning of the ascending noradrenergic pathways.[45, 63]

Evidence suggests that a dopamine system dysfunction may occur in narcolepsy, while a similar malfunction of the norepinephrine system may occur in idiopathic hypersomnia. Decreased CSF histamine levels have been reported in primary hypersomnia, as well as in narcolepsy, but not in non-CNS hypersomnias, suggesting that histamine may be an indicator of a central (versus a peripheral) origin for hypersomnias.[16]

A major advance in the understanding of the pathology of narcolepsy, a disorder closely related to primary hypersomnia, was made after the discovery of narcolepsy-associated genes in animals; ie, genes involved in the pathology of the hypocretin/orexin ligand and its receptor.[17, 18] Low CSF concentrations of hypocretin-1 and hypocretin-2 in HLA DQB1*0602 were also found in primary hypersomnia, and a generalized defect in hcrt-2 transmission may be present in this disorder. As hypocretin peptides excite the histaminergic system by the hypocretin receptor 2,[19] hypocretin deficiency may result in excessive daytime sleepiness via decreased histaminergic function.[16]

Epidemiology

Occurrence in the United States

While the rates of excessive daytime sleepiness complaints in the general population are between 0.5-5% of adults (in surveys without a specific consideration of causes/diagnoses), idiopathic hypersomnia is diagnosed in about 5-10% of individuals who are self referred to a sleep clinic with a chief complaint of daytime sleepiness.[1] A precise estimation of idiopathic hypersomnia prevalence is complicated by a lack of clear biologic markers or unambiguous diagnostic criteria.

A study by Ohayon et al suggested that excessive sleepiness is more prevalent than previously estimated. The study found that with 27.8% of 15,929 individuals from 15 US states reported excessive sleepiness. Even when using restrictive criteria of frequency at least 3 times per week for at least 3 months despite normal sleep duration, the prevalence was 4.7%.[20]

A recent large series found that idiopathic hypersomnia represented about 1% of 6000 patients seen in sleep centers—given that idiopathic hypersomnia is believed to be 60% as prevalent as narcolepsy, this raises the question of diagnostic accuracy.[37, 63]

Sex-, age- and family-related demographics

Gender ratio for hypersomnolence is unknown, though female predominance was found in some but not all of these studies.[35, 36, 37, 38]   Approximately 33%–66% of idiopathic hypersomnia cases appear to be familial.[63]

 

As with narcolepsy and Klein-Levin syndrome, onset of hypersomnolence is most common during adolescence and rare in people older than 30 years. The diagnosis of idiopathic hypersomnia is complicated by the fact that differentiating between excessive versus long sleep or normal versus abnormal wakefulness is often difficult in this population.

Prognosis

After a typical onset between the ages of 15 and 30 years, untreated hypersomnolence presents a chronic but stable course. Idiopathic hypersomnia is a lifelong disorder, believed to have no tendency to remit spontaneously, though a few studies have reported up to 25% of patients that carry the idiopathic hypersomnia diagnosis demonstrate spontaneous improvement in excessive daytime somnolence. This seemingly conflicting data again raises the question of diagnostic accuracy. 

Consequences of this disease are mostly social and professional in nature, sharing a psychosocial burden that is similar to narcolepsy.[57, 58, 59, 63]    

Daytime sleepiness can lead to depression. Of note, in children, daytime sleepiness can present as hyperactivity.[1]

 

Patient Education

While treating patients with hypersomnolence, the patient's close family should be involved in the overall education and decision-making process.

Because these disorders may lead to marriage breakdown, extensive counseling for the patient's partners, educating them about the symptomatology and treatment options, must be part of a comprehensive management plan.

Patients often need significant support because they are at risk of being misunderstood as being incompetent or slothful. Therefore, education of relatives, friends, and colleagues helps the patient to function much better with this incurable disease.

For patient education information, see the Sleep Disorders Center, as well as Disorders That Disrupt Sleep (Parasomnias), the Hypersomnia Foundation, and Narcolepsy.

Medline Plus/National Institutes of Health (NIH) provides concise and to-the-point summaries of the diagnosis and recommendations for patients and families dealing with primary hypersomnia and Kleine-Levine syndrome.

The Mayo clinic offers an additional, more comprehensive patient resource on idiopathic/primary hypersomnia.

History

The diagnostic criteria of idiopathic hypersomnia in ICSD-3 includes both objective testing parameters and clinical criteria, which are similar to DSM-5 criteria:[2, 60, 61]

Additional characteristics

The most typical referral is for the polysymptomatic form of primary hypersomnia and is characterized by the following:[5, 21]

These patients do not feel refreshed following naps and, therefore, fight sleepiness as long as they are able. Patients are difficult to awaken from sleep or naps.

Some patients complain of headaches, fainting episodes, orthostatic hypotension, and peripheral vascular complaints of Raynaud phenomenon. Rarely, hypnagogic hallucinations and sleep paralysis may be observed. During long periods of drowsiness, patients might develop automatic behavior, during which they act in a semicontrolled fashion. (During automatic behavior episodes, these patients may endanger themselves through risk of injury.)

In patients with the recurrent form (i.e., Kleine-Levine syndrome), hypersomnia occurs for days to weeks several times a year. In between, patients do not have excessive daytime sleepiness. Some patients may develop symptoms of irritability, hypersexuality, hyperphagia, impulsive behavior, depersonalization, hallucinations, depression, and disorientation.

Physical Examination

The patient may appear overtired or even fall asleep in the physician’s office. The rest of the physical examination, however, will not reveal any particular features suggesting a diagnosis of hypersomnolence.

The physical examination goal is to exclude alternate diagnoses. A diagnosis of obstructive sleep apnea rather than hypersomnolence should be considered for a patient presenting with hypersomnia associated with central obesity, micrognathia or retrognathia, macroglossia, crowded oropharynx, nasal obstruction, and tonsillar enlargement.

An underlying rheumatologic disease, such as active rheumatoid arthritis or osteoarthritis, may cause daytime hyperoxia and sleepiness associated with poor nighttime sleep due to pain. Prior head trauma sequela or a current brain tumor can leave their specific mark on the neurologic examination.

Specific findings may suggest a degenerative neurologic condition (e.g., Parkinson or Huntington disease), endocrine dysfunction (e.g., hypothyroidism), viral and bacterial infections (e.g., hypersomnia secondary to viral encephalitis), pulmonary disease with secondary sleep-related breathing difficulties (e.g., chronic bronchitis) or musculoskeletal disorders (e.g., rheumatoid arthritis, fibromyalgia).

An overweight patient with primary hypersomnia should be assessed for underlying endocrine problems and sleep apnea.

If psychomotor retardation or agitation is noted, mental disorders with secondary sleep disturbance need to be ruled out before making a diagnosis of hypersomnolence.

Mental status examination

Major depressive disorder commonly presents with decreased energy and tiredness and atypical depression presents with hypersomnia. Similarly, speech that is soft in quality, with a decreased rate of production and an increased latency of answers, might indicate excessive tiredness, but it may also indicate depression.

Mood might be “down," tired,” or even “depressed,” with mood congruent and a decreased range of affect. If this is the case, the meaning of the words needs to be carefully qualified. Is it that the patient has depression (ie, major depressive disorder) or is it that he/she feels down as a reaction to oversleeping and decreased ability to function?

Thought processes should be coherent and goal directed. While suicidal ideation is not typical for hypersomnolence, because of the overlap between sleeping and affective disorders, standard questioning about the presence of suicidal and homicidal ideation is recommended.

Insight and judgment are most times good. Mild cognitive changes in the domains of attention, concentration and short-term memory are occasionally present, but most often the cognitive examination should not reveal any significant deficits.

Recurrent hypersomnia

On the neurologic examination, patients with Klein-Levin syndrome may present with a number of nonspecific findings including nystagmus, dysarthria, and generalized hyperreflexia.

For secondary Kleine-Levin syndrome, patients tend to be older and have more frequent and longer episodes, but they present with clinical symptoms and treatment responses similar to those of primary cases.[11]

Approach Considerations

Hypersomnolence is a diagnosis of exclusion. Other causes of excessive daytime somnolence should be ruled out before a diagnosis of hypersomnolence is made.

Patients should receive a complete blood count (CBC), screening biochemistry tests, and thyroid-stimulating hormone tests to exclude common physical disorders that may present with complaints of excessive tiredness, often expressed as excessive sleepiness by patients. A drug screen is indicated if substance-induced sleep disorder needs to be ruled out.

As excessive sleepiness is essentially a self-reported, subjective complaint, a number of tests have been created with the goal of increasing the data collection validity and reliability. Commonly used scales for a quantitative, systematic assessment of excessive sleepiness are the Epworth Sleepiness Scale and the Stanford Sleepiness Scale. While helpful, these scales remain essentially subjective in nature, which raises questions about the characteristics of sleepiness as assessed by subjective methods (e.g., the Epworth and Stanford sleepiness scales) versus objective ones (e.g., polysomnography and the Multiple Sleep Latency Test).[24, 25]

Polysomnography and Multiple Sleep Latency Test

Complete in-laboratory polysomnography (PSG) studies are essential to exclude other sleep disorders, particularly sleep breathing disorder, periodic limb movement disorder, and narcolepsy. Nocturnal PSG findings in hypersomnolence include a short sleep latency, absence of arousals or awakenings, normal distribution of REM and NREM sleep, and normal to prolonged sleep duration.[26, 27]

A PSG study completion is required prior to the Multiple Sleep Latency Test to objectively characterize preceding sleep and uncover potential causes of sleep fragmentation. The PSG must have confirmed at least 6 hours of sleep for the Multiple Sleep Latency Test results to be considered in diagnosing hypersomnolence. Typical PSG findings in idiopathic hypersomnia include shortened sleep latency, increased sleep efficiency (often >90%), and increased slow wave sleep[3, 10, 12, 43, 63]   It should be noted that these are nonspecific findings and could be present in behaviorally induced insufficient sleep syndrome (BIISS).[63]  Some studies report increased frequency of sleep spindles either throughout the sleep period or at the beginning and end of the night in idiopathic hypersomnia.[8, 35]  It should be noted that SOREMPs are rare in idiopathic hypersomnia (IH). Indices for arousal, apnea-hypopnea, and periodic limb movements (PLM) are generally less than 5 to 10 per hour. Other studies have a belief that PLM in a patient with excessive daytime sleepiness who does not have a diagnosis of restless legs syndrome (RLS) should not preclude giving a diagnosis of IH—occasionally higher indices are seen.[10, 12, 44, 45, 63]   

Sleep latency on the Multiple Sleep Latency Test is short, with a mean sleep latency time < 8 minutes.[9, 10, 12]  SOREMPs can occur in approximately 3–4% of naps but by definition will not occur more than once. This is contrasted with narcolepsy, where sleep-onset REM periods (the occurrence of REM sleep within 20 minutes of sleep onset) are observed in two or more naps.

Breathing-related sleep disturbances and frequent limb movements disrupting sleep are not present.

The following PSG features are required for the diagnosis of hypersomnolence:

The Multiple Sleep Latency Test is performed to evaluate the presence of pathologic sleepiness. The subject is studied during 5 daytime naps taken 2 hours apart. According to 2 studies, the mean Multiple Sleep Latency Test score in hypersomnolence is slightly higher than the score in narcolepsy. The mean Multiple Sleep Latency Test score was found to be 6.5 ± 3.2 minutes for idiopathic hypersomnolence versus 3.3 ± 3.3 minutes for narcolepsy. Narcolepsy is excluded by the absence of sleep-onset REM periods on the 5-nap Multiple Sleep Latency Test.

Electroencephalography

In recurrent primary hypersomnia (ie, Kleine-Levin syndrome), routine electroencephalographic studies performed during hypersomnia show a general slowing of the background rhythm and paroxysmal bursts of theta activity. Nocturnal PSG shows prolonged sleep duration and decreased sleep latency (< 10 min). In addition, sleep-onset REM has been reported during symptomatic periods. (See the images below.)[1, 8, 9, 10, 12]



View Image

Primary hypersomnia. Polysomnographic study demonstrates apnea (absence of carbon dioxide fluctuation indicating no flow), chest wall paradox, abrupt ....



View Image

Primary hypersomnia. In contrast to obstructive sleep apnea, mixed apnea shows absence of respiratory efforts in the first segment of the apnea.



View Image

Primary hypersomnia. Periodic limb movements show intermittent leg electromyogram activity accompanied by electroencephalogram arousals.

Approach Considerations

Severe idiopathic hypersomnolence is a disabling problem that often leads to permanent unemployment and responds poorly to medical treatment.[9, 8] Moreover, because the underlying cause of idiopathic hypersomnolence is unknown, treatment remains symptomatic in nature.

Some limited evidence postulates that a CSF “somnogen” potentiates GABAA inhibitory effects and may increase excessive daytime sleepiness (EDS). Flumazenil is thought to competitively antagonize activity at the benzodiazepine recognition site, though it does not impact all GABA-ergic neurons. In a small study, 7 hypersomnolent patients had improved vigilance with flumazenil.[31]  Another study from a single sleep center reported subjective improvement in EDS in 53 idiopathic hypersomnia (IH) patients that were refractory to traditional psychostimulants, who were given clarithromycin, an antibiotic that has GABAA inhibitory properties. Long-term treatment, however, has potential side effects of antibiotic resistance, GI problems, and infections.[88]

The American Academy of Sleep Medicine practice parameters state that successful treatment of hypersomnia of central origin requires an accurate diagnosis, individual tailoring of therapy to produce maximum possible return of function, and regular follow-up to monitor response to therapy.

Modafinil, armodafinil,sodium oxybate, amphetamine, methamphetamine, dextroamphetamine, methylphenidate, and selegiline are effective treatments for excessive sleepiness associated with narcolepsy and primary hypersomnias. Scheduled naps can be beneficial to combat sleepiness in these patients.[28]

Behavioral approaches and sleep hygiene techniques are recommended, although they have little overall positive impact on this disease.

Consultations

The diagnosis of hypersomnolence is made after excluding neurologic, pulmonary, and psychiatric disorders known to cause excessive sleepiness. Therefore, if an underlying cause is suggested, appropriate consultations with a neurologist, pulmonologist, and psychiatrist should be obtained.

Activity

Caution is recommended in activities in which hypersomnolence may be hazardous.

Pharmacologic Therapy

Medications that have been used in the treatment of this disorder include tricyclic antidepressants (TCAs), monoamine oxidase inhibitors (MAOIs), clonidine, levodopa, bromocriptine, amantadine, methysergide, pemoline (as of October 2005, this is no longer available in the United States; risk of liver toxicity outweighs benefits), and modafinil. (Patients develop tolerance to their medications; exercise caution in prescribing drugs.)

Therapy for idiopathic hypersomnolence involves maintaining the patient on a daily use of stimulants. The drug dose is titrated so that the patient stays alert during the day, but adverse effects should be avoided.

Methylphenidate (Ritalin), mazindol (withdrawn from the US market in 2001), and dextroamphetamine are the most commonly prescribed medications.

Pitolisant, a wake-promoting agent that increases CNS histamine via blocking presynaptic H3 reuptake, appears promising, but is still in the new drug application (NDA) process in the United States pending FDA approval.[63]

Modafinil and armodafinil

Modafinil, a 1:1 racemic mixture of (R)-(-) and (S)-(-) enantiomers, and armodafinil, the isolated (R)-(-) enantiomer, have proved clinically useful in the treatment of narcolepsy and other causes of excessive daytime sleepiness, such as idiopathic hypersomnolence.[29] It is a psychostimulant that enhances wakefulness and vigilance, but its pharmacologic profile is notably different from the amphetamines, methylphenidate, or cocaine. Modafinil is less likely to produce side effects such as jitteriness, anxiety, or excess locomotor activity or to lead to a hypersomnolent rebound effect. Modafinil, with its 1:1 racemic mixture has an estimated elimination half-life of 4 hours for the S-enantiomer and 15 hours for the R-enantiomer, functionally producing an estimated 9–14 hours half-life. Given the enantiomeric differences, armodafinil has a longer elimination half-life of around 10–15 hours.[3, 30, 63]

The mechanism of action of modafinil is not fully understood. Modafinil induces wakefulness in part by its action in the anterior hypothalamus. Its dopamine-releasing action in the nucleus accumbens is weak and dose dependent; the likelihood of a euphoric response, and, therefore, the abuse potential and tolerance, is small.

Modafinil has central alpha 1-adrenergic agonist effects (ie, it directly stimulates the receptors). Modafinil inhibits the reuptake of noradrenaline by the noradrenergic terminals on sleep-promoting neurons of the ventrolateral preoptic nucleus (VLPO). More significant, perhaps, is its ability to increase excitatory glutaminergic transmission and reduce local gamma-aminobutyric acid (GABA)–ergic transmission, thereby diminishing GABA(A) receptor signaling on the mesolimbic dopamine terminals.[30, 31]

Physician Legal Responsibilities

Physicians have a legal responsibility to know which medical conditions may impede driving ability, to diagnose these conditions in their patients, and to discuss the implications of these conditions.

The requirement to report unfit drivers varies among different jurisdictions, and interpretations of the law vary among the courts. Therefore, a physician’s risk of liability is unclear. Physicians may face legal action by their patients if they fail to counsel the patients on the dangers of driving associated with certain medications or medical conditions.

Physicians’ legal responsibilities to report patients with certain medical conditions, when required by law, override their ethical responsibilities to keep patients’ medical information confidential.

Medication Summary

Patients often require drug therapy to treat daytime hypersomnolence. Prior to initiating therapy with stimulants, clearly establish a diagnosis and consider potential for abuse.[8, 23]

Modafinil, a wake-promoting agent, is approved for treatment of excessive sleepiness associated with narcolepsy, obstructive sleep apnea-hypopnea syndrome (OSAHS), and shift-work sleep disorder (SWSD).[30, 31] The studies have shown significant benefits on various objective measures and subjective estimates of excessive sleepiness.

The clinical efficacy of modafinil, combined with its improved safety over CNS stimulants, has made it the most prescribed medication for the treatment of excessive sleepiness associated with narcolepsy. Unlike many other medications used for excessive sleepiness, modafinil is not known to be abused. The most common adverse event reported in clinical studies was headaches; most were transient and mild to moderate in severity. Modafinil also has the potential for interactions with other drugs metabolized via cytochrome P450 enzyme pathways.

For Kleine-Levin syndrome, somnolence can decrease with stimulants (mainly amphetamines), while neuroleptics and antidepressants are of poor benefit. Lithium, rather than carbamazepine or other antiepileptics, was found to have a higher success rate for stopping relapses.[32]

Modafinil (Provigil)

Clinical Context:  Modafinil, (1:1 racemic mixture of R- and S-modafinil) improves wakefulness, though the mechanism of action remains unclear. It may act at the dopamine receptor (DAT), inhibiting dopamine reuptake and may exert stimulant effects by decreasing GABA-medicated neurotransmission. It has wake-promoting actions similar to those of sympathomimetic agents, though no direct action on dopamine receptors themselves; wakefulness is not attenuated by known dopamine antagonists.

Armodafinil (Nuvigil)

Clinical Context:  Armodafinil is an R-enantiomer of modafinil (mixture of R- and S-enantiomers). It elicits wake-promoting actions similar to those of sympathomimetic agents, although its pharmacologic profile is not identical to sympathomimetic amines and has no direct action on dopamine receptors themselves.  Wakefulness is not attenuated by known dopamine antagonists. It may act at the dopamine receptor (DAT), inhibiting dopamine reuptake and may exert stimulant effects by decreasing GABA-medicated neurotransmission. It is not a direct- or indirect-acting dopamine receptor agonist. Armodafinil is indicated for the improvement of wakefulness in individuals with excessive sleepiness associated with narcolepsy, obstructive sleep apnea-hypopnea syndrome (OSAHS), or shift-work sleep disorder.

Methylphenidate (Ritalin, Daytrana, Methylin, Concerta)

Clinical Context:  Methylphenidate is used for symptomatic management of primary hypersomnolence whenever the patient needs to be alert or engages in activities in which hypersomnolence may be hazardous. The drug blocks the reuptake mechanism of dopaminergic neurons. Methylphenidate stimulates the cerebral cortex and subcortical structures.

Dextroamphetamine (Procentra, Dexedrine, Zenzedi)

Clinical Context:  This agent increases the amount of circulating dopamine and norepinephrine in the cerebral cortex by blocking the reuptake of norepinephrine or dopamine from the synapse.

Dextroamphetamine and amphetamine mixtures (Mydayis, Adderall XR)

Clinical Context:  This agent produces CNS and respiratory stimulation. The CNS effect may occur in the cerebral cortex and reticular activating system. Dextroamphetamine-amphetamine mixture may have a direct effect on alpha- and beta-receptor sites in the peripheral system and may also release stores of norepinephrine in adrenergic nerve terminals. The mixture contains various salts of amphetamine and dextroamphetamine. It is available as 5-, 7.5-, 10-, 12.5-, 15-, 20-, and 30mg scored tablets.

Amphetamine (Evekeo)

Clinical Context:  Amphetamines are noncatecholamine sympathomimetic amines that promote release of catecholamines from their storage sites in the presynaptic nerve terminals. Amphetamine 1:1 racemic mixture of dextroamphetamine and levoamphetamine (immediate release only) has an FDA indication for promoting wakefulness in narcolepsy.

Class Summary

These agents have wake-promoting activities.

What is the difference between primary hypersomnia and narcolepsy?What are the subgroups of idiopathic CNS hypersomnia?What are the DSM-5 diagnostic criteria for hypersomnolence disorder?What are the ASDA diagnostic criteria for idiopathic hypersomnia?How does the ICSD-3 classify central disorders of hypersomnolence?What is Kleine-Levin syndrome (KLS)?What are the subtypes of Kleine-Levin syndrome (KLS)?What causes primary hypersomnia?What is the prevalence of primary hypersomnia?What are the sexual predilections of primary hypersomnia?At what age is primary hypersomnia usually diagnosed?What is the prognosis of primary hypersomnia?What is included in patient education about primary hypersomnia?What are the ICSD-3 diagnostic criteria for idiopathic hypersomnia?Which clinical history findings are characteristic of primary hypersomnia?Which physical findings are characteristic of primary hypersomnia?Which mental status exam (MSE) findings are characteristic of primary hypersomnia?Which neurologic exam findings are characteristic of Kleine-Levin syndrome (KLS)?What should be considered prior to diagnosing primary hypersomnia?How is primary hypersomnia differentiated from behaviorally induced insufficient sleep syndrome (BIISS)?How is primary hypersomnia differentiated from normal sleep variation?How is primary hypersomnia differentiated from chronic insufficient sleep?How is primary hypersomnia differentiated from substance-induced sleep disorder, hypersomnia type?How is primary hypersomnia differentiated from upper airway resistance syndrome (breathing-related sleep disorder)?How is primary hypersomnia differentiated from posttraumatic hypersomnia?How is primary hypersomnia differentiated from other neurologic disorders?How is primary hypersomnia differentiated from hypersomnia due to other general medical conditions?How is primary hypersomnia differentiated from a major depressive disorder?How is primary hypersomnia differentiated from narcolepsy?How is primary hypersomnia differentiated from circadian rhythm sleep disorders?How is primary hypersomnia differentiated from dysthymia?How is primary hypersomnia differentiated from chronic fatigue syndrome?What are the differential diagnoses for Primary Hypersomnia?How is primary hypersomnia diagnosed?What is the role of PSG and MSLT in the workup of primary hypersomnia?What is the role of electroencephalography in the workup of primary hypersomnia?How is primary hypersomnia treated?Which specialist consultations are beneficial to patients with primary hypersomnia?Which activity modifications should be taken by patients with primary hypersomnia?Which medications are used in the treatment of primary hypersomnia?What is the role of modafinil and armodafinil in the treatment of primary hypersomnia?What are the physician&#39;s legal responsibilities to report patients with hypersomnia?What is the role of medications in the treatment of primary hypersomnia?Which medications in the drug class Stimulants are used in the treatment of Primary Hypersomnia?

Author

Adrian Preda, MD, Professor of Clinical Psychiatry and Human Behavior, University of California, Irvine, School of Medicine

Disclosure: Nothing to disclose.

Coauthor(s)

David R Bowman, MD, Psychiatrist, Deputy Chief, Joint Behavioral Health Clinic, Multi-Disciplinary Outpatient Clinic, Medical Review Officer, Clinical Practice Guidelines Champion, External Behavioral Health Consultant, Department of Behavioral Health, Moncrief Army Health Clinic

Disclosure: Nothing to disclose.

Specialty Editors

Francisco Talavera, PharmD, PhD, Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Received salary from Medscape for employment. for: Medscape.

Chief Editor

Ana Hategan, MD, FRCPC, Associate Clinical Professor, Department of Psychiatry and Behavioral Neurosciences, Division of Geriatric Psychiatry, McMaster University School of Medicine; Geriatric Psychiatrist, St Joseph's Health Care Hamilton, Canada

Disclosure: Book royalties and/or honoraria for articles from American Psychiatric Publishing, Springer, and Current Psychiatry.

Acknowledgements

Jennifer S Morse, MD Associate Medical Director, Optum Health

Jennifer S Morse, MD is a member of the following medical societies: Academy of Psychosomatic Medicine, Aerospace Medical Association, and American Psychiatric Association

Disclosure: Nothing to disclose.

Francisco Talavera, PharmD, PhD Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Medscape Salary Employment

References

  1. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5th edition. Washington, DC: American Psychiatric Association;
  2. American Academy of Sleep Medicine. International Classification of Sleep Disorders. Third Edition (ICSD-3). Westchester, IL: American Academy of Sleep Medicine; 2014.
  3. Dement W, Rechtschaffen A, Gulevich G. The nature of the narcoleptic sleep attack. Neurology. 1966 Jan. 16(1):18-33. [View Abstract]
  4. Roth B, Nevsimalova S, Rechtschaffen A. Hypersomnia with "sleep drunkenness". Arch Gen Psychiatry. 1972 May. 26(5):456-62. [View Abstract]
  5. Roehrs T, Zorick F, Sicklesteel J. Excessive daytime sleepiness associated with insufficient sleep. Sleep. 1983. 6(4):319-25. [View Abstract]
  6. Billiard M. Diagnosis of narcolepsy and idiopathic hypersomnia. An update based on the International classification of sleep disorders, 2nd edition. Sleep Med Rev. 2007 Oct. 11(5):377-88. [View Abstract]
  7. Bassetti C, Pelayo R, Guilleminault C. Idiopathic Hypersomnia. Kryger MH, Roth T, Dement WC. Principles and Practices of Sleep Medicine. 4th Edition. Philadelphia, PA: Elsevier; 2005. 791-800.
  8. Roth T. Introduction: narcolepsy and excessive daytime sleepiness: from the bench to the bedside. J Clin Psychiatry. 2007. 68 Suppl 13:4. [View Abstract]
  9. Guilleminault C. Disorders of excessive sleepiness. Ann Clin Res. 1985. 17(5):209-19. [View Abstract]
  10. Roth B. Narcolepsy and hypersomnia: review and classification of 642 personally observed cases. Schweiz Arch Neurol Neurochir Psychiatr. 1976. 119(1):31-41. [View Abstract]
  11. Arnulf I, Zeitzer JM, File J, et al. Kleine-Levin syndrome: a systematic review of 186 cases in the literature. Brain. 2005 Dec. 128(Pt 12):2763-76. [View Abstract]
  12. Billiard M, Guilleminault C, Dement WC. A menstruation-linked periodic hypersomnia: Kleine-Levin syndrome or new clinical entity?. Neurology. 1975. 25:436-443. [View Abstract]
  13. Montplaisir J, Poirier G. HLA in disorders of excessive sleepiness without cataplexy in Canada. Honda Y, Juti T. HLA in Narcolepsy. Berlin, Germany: Springer-Verlag; 1988. 186-190.
  14. Guilleminault C, Faull KF, Miles L. Posttraumatic excessive daytime sleepiness: a review of 20 patients. Neurology. 1983 Dec. 33(12):1584-9. [View Abstract]
  15. Montplaisir J, de Champlain J, Young SN. Narcolepsy and idiopathic hypersomnia: biogenic amines and related compounds in CSF. Neurology. 1982 Nov. 32(11):1299-302. [View Abstract]
  16. Kanbayashi T, Kodama T, Kondo H, Satoh S, Inoue Y, Chiba S, et al. CSF histamine contents in narcolepsy, idiopathic hypersomnia and obstructive sleep apnea syndrome. Sleep. 2009 Feb 1. 32(2):181-7. [View Abstract]
  17. Nishino S, Okuro M, Kotorii N, Anegawa E, Ishimaru Y, Matsumura M, et al. Hypocretin/orexin and narcolepsy: new basic and clinical insights. Acta Physiol (Oxf). 2009 Jun 25. [View Abstract]
  18. Nishino S, Kanbayashi T. Symptomatic narcolepsy, cataplexy and hypersomnia, and their implications in the hypothalamic hypocretin/orexin system. Sleep Med Rev. 2005 Aug. 9(4):269-310. [View Abstract]
  19. Yamanaka A, Tsujino N, Funahashi H, Honda K, Guan JL, Wang QP, et al. Orexins activate histaminergic neurons via the orexin 2 receptor. Biochem Biophys Res Commun. 2002. 290:1237-45. [View Abstract]
  20. Ohayon MM, Dauvilliers Y, Reynolds CF 3rd. Operational Definitions and Algorithms for Excessive Sleepiness in the General Population: Implications for DSM-5 Nosology. Arch Gen Psychiatry. 2012 Jan. 69(1):71-9. [View Abstract]
  21. Guilleminault C, Faull KF. Sleepiness in nonnarcoleptic, non-sleep apneic EDS patients: the idiopathic CNS hypersomnolence. Sleep. 1982. 5 Suppl 2:S175-81. [View Abstract]
  22. Bassetti C, Gugger M, Bischof M. The narcoleptic borderland: a multimodal diagnostic approach including cerebrospinal fluid levels of hypocretin-1 (orexin A). Sleep Med. 2003 Jan. 4(1):7-12. [View Abstract]
  23. Ohayon MM. From wakefulness to excessive sleepiness: what we know and still need to know. Sleep Med Rev. 2008 Apr. 12(2):129-41. [View Abstract]
  24. Sangal RB; Mitler MM; Sangal JM. Subjective sleepiness ratings (Epworth sleepiness scale) do not reflect the same parameter of sleepiness as objective sleepiness (maintenance of wakefulness test) in patients with narcolepsy. Clin Neurophysiol. Dec 1999. (110)12:2131-5. [View Abstract]
  25. Sangal RB, Sangal JM, Belisle C. Subjective and objective indices of sleepiness (ESS and MWT) are not equally useful in patients with sleep apnea. Clin Electroencephalogr. 1999 Apr. 30(2):73-5. [View Abstract]
  26. Rechtschaffen A, Roth B. Nocturnal sleep of hypersomniacs. Act Nerv Super (Praha). 1969. 11(3):229-33. [View Abstract]
  27. Anderson KN, Pilsworth S, Sharples LD, Smith IE, Shneerson JM. Idiopathic hypersomnia: a study of 77 cases. Sleep. 2007 Oct 1. 30(10):1274-81. [View Abstract]
  28. [Guideline] Morgenthaler TI, Kapur VK, Brown T, Swick TJ, Alessi C, Aurora RN, et al. Practice parameters for the treatment of narcolepsy and other hypersomnias of central origin. Sleep. 2007 Dec 1. 30(12):1705-11. [View Abstract]
  29. Ballon JS, Feifel D. A systematic review of modafinil: Potential clinical uses and mechanisms of action. J Clin Psychiatry. 2006 Apr. 67(4):554-66. [View Abstract]
  30. Valentino RM, Foldvary-Schaefer N. Modafinil in the treatment of excessive daytime sleepiness. Cleve Clin J Med. 2007 Aug. 74(8):561-6, 568-71. [View Abstract]
  31. Schwartz JR. Modafinil: new indications for wake promotion. Expert Opin Pharmacother. 2005 Jan. 6(1):115-29. [View Abstract]
  32. Poppe M, Friebel D, Reuner U, Todt H, Koch R, Heubner G. The Kleine-Levin syndrome - effects of treatment with lithium. Neuropediatrics. 2003. 34:113-9. [View Abstract]
  33. Roth B, Nevsimalova S, Rechtschaffen A. Hypersomnia with "sleep drunkenness". Arch Gen Psychiatry. 1972 May. 26 (5):456-62. [View Abstract]
  34. Billiard M, Dauvilliers Y. Idiopathic Hypersomnia. Sleep Med Rev. 2001 Oct. 5 (5):349-358. [View Abstract]
  35. Vernet C, Arnulf I. Idiopathic hypersomnia with and without long sleep time: a controlled series of 75 patients. Sleep. 2009 Jun. 32 (6):753-9. [View Abstract]
  36. Bassetti C, Aldrich MS. Idiopathic hypersomnia. A series of 42 patients. Brain. 1997 Aug. 120 ( Pt 8):1423-35. [View Abstract]
  37. Anderson KN, Pilsworth S, Sharples LD, Smith IE, Shneerson JM. Idiopathic hypersomnia: a study of 77 cases. Sleep. 2007 Oct. 30 (10):1274-81. [View Abstract]
  38. 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 May. 60 (5):576-8. [View Abstract]
  39. Bassetti C, Gugger M, Bischof M, Mathis J, Sturzenegger C, Werth E, et al. The narcoleptic borderland: a multimodal diagnostic approach including cerebrospinal fluid levels of hypocretin-1 (orexin A). Sleep Med. 2003 Jan. 4 (1):7-12. [View Abstract]
  40. 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):153-8. [View Abstract]
  41. Billiard M. Idiopathic hypersomnia. Neurol Clin. 1996 Aug. 14 (3):573-82. [View Abstract]
  42. Honda Y, Honda M. [Idiopathic hypersomnia--review of literatures and clinical experiences on 16 Japanese cases]. Nihon Rinsho. 1998 Feb. 56 (2):371-5. [View Abstract]
  43. Lippert J, Halfter H, Heidbreder A, Röhr D, Gess B, Boentert M, et al. Altered dynamics in the circadian oscillation of clock genes in dermal fibroblasts of patients suffering from idiopathic hypersomnia. PLoS One. 2014. 9 (1):e85255. [View Abstract]
  44. Montplaisir J, de Champlain J, Young SN, Missala K, Sourkes TL, Walsh J, et al. Narcolepsy and idiopthic hypersomnia: biogenic amines and related compounds in CSF. Neurology. 1982 Nov. 32 (11):1299-302. [View Abstract]
  45. Petitjean F, Jouvet M. [Hypersomnia and increase of cerebral 5-hydroxyindoleacetic acid due to isthmic lesions in the cat]. C R Seances Soc Biol Fil. 1970. 164 (11):2288-93. [View Abstract]
  46. Mignot E, Lammers GJ, Ripley B, Okun M, Nevsimalova S, Overeem S, et al. The role of cerebrospinal fluid hypocretin measurement in the diagnosis of narcolepsy and other hypersomnias. Arch Neurol. 2002 Oct. 59 (10):1553-62. [View Abstract]
  47. Dauvilliers Y, Baumann CR, Carlander B, Bischof M, Blatter T, Lecendreux M, et al. CSF hypocretin-1 levels in narcolepsy, Kleine-Levin syndrome, and other hypersomnias and neurological conditions. J Neurol Neurosurg Psychiatry. 2003 Dec. 74 (12):1667-73. [View Abstract]
  48. Rye DB, Bliwise DL, Parker K, Trotti LM, Saini P, Fairley J, et al. Modulation of vigilance in the primary hypersomnias by endogenous enhancement of GABAA receptors. Sci Transl Med. 2012 Nov 21. 4 (161):161ra151. [View Abstract]
  49. Bové A, Culebras A, Moore JT, Westlake RE. Relationship between sleep spindles and hypersomnia. Sleep. 1994 Aug. 17 (5):449-55. [View Abstract]
  50. Baker TL, Guilleminault C, Nino-Murcia G, Dement WC. Comparative polysomnographic study of narcolepsy and idiopathic central nervous system hypersomnia. Sleep. 1986. 9 (1 Pt 2):232-42. [View Abstract]
  51. Nicolas A, Lespérance P, Montplaisir J. Is excessive daytime sleepiness with periodic leg movements during sleep a specific diagnostic category?. Eur Neurol. 1998 Jul. 40 (1):22-6. [View Abstract]
  52. Montplaisir J, Fantini L. Idiopathic hypersomnia: a diagnostic dilemma. A commentary of "Idiopathic hypersomnia" (M. Billiard and Y. Dauvilliers). Sleep Med Rev. 2001 Oct. 5 (5):361-362. [View Abstract]
  53. Guilleminault C, Stoohs R, Clerk A, Cetel M, Maistros P. A cause of excessive daytime sleepiness. The upper airway resistance syndrome. Chest. 1993 Sep. 104 (3):781-7. [View Abstract]
  54. Roehrs T, Zorick F, Sicklesteel J, Wittig R, Roth T. Excessive daytime sleepiness associated with insufficient sleep. Sleep. 1983. 6 (4):319-25. [View Abstract]
  55. Roehrs TA, Roth T. Chronic insufficient sleep and its recovery. Sleep Med. 2003 Jan. 4 (1):5-6. [View Abstract]
  56. Trotti LM, Saini P, Freeman AA, Bliwise DL, García PS, Jenkins A, et al. Improvement in daytime sleepiness with clarithromycin in patients with GABA-related hypersomnia: Clinical experience. J Psychopharmacol. 2014 Jul. 28 (7):697-702. [View Abstract]
  57. Broughton R et al. The socio-economic effects of idiopathic hypersomnia-comparison with controls and with compound narcoleptics. Sleep. 1978. 229-33.
  58. Dauvilliers Y, Paquereau J, Bastuji H, Drouot X, Weil JS, Viot-Blanc V. Psychological health in central hypersomnias: the French Harmony study. J Neurol Neurosurg Psychiatry. 2009 Jun. 80 (6):636-41. [View Abstract]
  59. Vernet C, Leu-Semenescu S, Buzare MA, Arnulf I. Subjective symptoms in idiopathic hypersomnia: beyond excessive sleepiness. J Sleep Res. 2010 Dec. 19 (4):525-34. [View Abstract]
  60. American Academy of Sleep Medicine. International classification of Sleep Disorders. 3rd edition. Darien, IL: American Academy of Sleep Medicine.;
  61. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. Fifth edition. Arlington, VA: American Psychiatric Association; 2013.
  62. Arnulf I, Rico TJ, Mignot E. Diagnosis, disease course, and management of patients with Kleine-Levin syndrome. Lancet Neurol. 2012 Oct. 11 (10):918-28. [View Abstract]
  63. Kryger MH, Roth T, Dement WC. Principles and Practice of Sleep Medicine. 6th edition. Philadelphia, PA: Elsevier; 2017.
  64. Arnulf I, Lin L, Gadoth N, File J, Lecendreux M, Franco P, et al. Kleine-Levin syndrome: a systematic study of 108 patients. Ann Neurol. 2008 Apr. 63 (4):482-93. [View Abstract]
  65. Salter MS, White PD. A variant of the Kleine-Levin syndrome precipitated by both Epstein-Barr and varicella-zoster virus infections. Biol Psychiatry. 1993 Mar 1. 33 (5):388-90. [View Abstract]
  66. GALLINEK A. The Kleine-Levin syndrome: hypersomnia, bulimia, and abnormal mental states. World Neurol. 1962 Mar. 3:235-43. [View Abstract]
  67. Kas A, Lavault S, Habert MO, Arnulf I. Feeling unreal: a functional imaging study in patients with Kleine-Levin syndrome. Brain. 2014 Jul. 137 (Pt 7):2077-87. [View Abstract]
  68. A Brierre de Boismont. Des Hallucinations: Ou Histoire Raisonnée des Apparitions, des Visions, des Songes, de l'Extase, des Rêves, du Magnetisme Et du Somnambulisme. Paris: Germer Baillière; 1862.
  69. Billiard M, Jaussent I, Dauvilliers Y, Besset A. Recurrent hypersomnia: a review of 339 cases. Sleep Med Rev. 2011 Aug. 15 (4):247-57. [View Abstract]
  70. Arnulf I, Lin L, Gadoth N, File J, Lecendreux M, Franco P, et al. Kleine-Levin syndrome: a systematic study of 108 patients. Ann Neurol. 2008 Apr. 63 (4):482-93. [View Abstract]
  71. Gadoth N, Kesler A, Vainstein G, Peled R, Lavie P. Clinical and polysomnographic characteristics of 34 patients with Kleine-Levin syndrome. J Sleep Res. 2001 Dec. 10 (4):337-41. [View Abstract]
  72. Huang YS, Guilleminault C, Lin KL, Hwang FM, Liu FY, Kung YP. Relationship between Kleine-Levin syndrome and upper respiratory infection in Taiwan. Sleep. 2012 Jan 1. 35 (1):123-9. [View Abstract]
  73. Lavault S, Golmard JL, Groos E, Brion A, Dauvilliers Y, Lecendreux M, et al. Kleine-Levin syndrome in 120 patients: differential diagnosis and long episodes. Ann Neurol. 2015 Mar. 77 (3):529-40. [View Abstract]
  74. Arnulf I, Zeitzer JM, File J, Farber N, Mignot E. Kleine-Levin syndrome: a systematic review of 186 cases in the literature. Brain. 2005 Dec. 128 (Pt 12):2763-76. [View Abstract]
  75. GALLINEK A. The Kleine-Levin syndrome: hypersomnia, bulimia, and abnormal mental states. World Neurol. 1962 Mar. 3:235-43. [View Abstract]
  76. Shukla GD, Bajpai HS, Mishra DN. Kleine-levin syndrome: a case report from India. Br J Psychiatry. 1982 Jul. 141:97-8. [View Abstract]
  77. Landtblom AM, Dige N, Schwerdt K, Säfström P, Granérus G. Short-term memory dysfunction in Kleine-Levin syndrome. Acta Neurol Scand. 2003 Nov. 108 (5):363-7. [View Abstract]
  78. CRITCHLEY M. Periodic hypersomnia and megaphagia in adolescent males. Brain. 1962 Dec. 85:627-56. [View Abstract]
  79. American Sleep Disorders Association. The international classification of sleep disorders: Diagnosis and coding manual. Chicago, IL: American Academy of Sleep Medicine; 1990.
  80. American Academy of Sleep Medicine. The international classification of sleep disorders—revised. Chicago, IL: American Academy of Sleep Medicine; 2005.
  81. Rosenow F, Kotagal P, Cohen BH, Green C, Wyllie E. Multiple sleep latency test and polysomnography in diagnosing Kleine-Levin syndrome and periodic hypersomnia. J Clin Neurophysiol. 2000 Sep. 17 (5):519-22. [View Abstract]
  82. Katz JD, Ropper AH. Familial Kleine-Levin syndrome: two siblings with unusually long hypersomnic spells. Arch Neurol. 2002 Dec. 59 (12):1959-61. [View Abstract]
  83. Portilla P, Durand E, Chalvon A, Habert M, Navelet Y, Prigent A, et al. [SPECT-identified hypoperfusion of the left temporomesial structures in a Kleine-Levin syndrome]. Rev Neurol (Paris). 2002 May. 158 (5 Pt 1):593-5. [View Abstract]
  84. Arnulf I, Lecendreux M, Franco P, Dauvilliers Y. [Kleine-Levin syndrome: state of the art]. Rev Neurol (Paris). 2008 Aug-Sep. 164 (8-9):658-68. [View Abstract]
  85. Schenck CH, Arnulf I, Mahowald MW. Sleep and sex: what can go wrong? A review of the literature on sleep related disorders and abnormal sexual behaviors and experiences. Sleep. 2007 Jun. 30 (6):683-702. [View Abstract]
  86. Malhotra S, Das MK, Gupta N, Muralidharan R. A clinical study of Kleine-Levin syndrome with evidence for hypothalamic-pituitary axis dysfunction. Biol Psychiatry. 1997 Aug 15. 42 (4):299-301. [View Abstract]
  87. VLACH V. [Periodic somnolence, bulimia and mental disorder (Kleine-Levin syndrome)]. Cesk Neurol. 1962 Nov. 25:401-5. [View Abstract]
  88. Domzał-Stryga A, Emeryk-Szajewska B, Kowalski J. [A case of hypersomnia resembling Kleine-Levin syndrome]. Neurol Neurochir Pol. 1986 Mar-Apr. 20 (2):158-60. [View Abstract]
  89. Manni R, Martinetti M, Ratti MT, Tartara A. Electrophysiological and immunogenetic findings in recurrent monosymptomatic-type hypersomnia: a study of two unrelated Italian cases. Acta Neurol Scand. 1993 Oct. 88 (4):293-5. [View Abstract]
  90. Hegarty A, Merriam AE. Autonomic events in Kleine-Levin syndrome. Am J Psychiatry. 1990 Jul. 147 (7):951-2. [View Abstract]
  91. Koerber RK, Torkelson R, Haven G, Donaldson J, Cohen SM, Case M. Increased cerebrospinal fluid 5-hydroxytryptamine and 5-hydroxyindoleacetic acid in Kleine-Levin syndrome. Neurology. 1984 Dec. 34 (12):1597-600. [View Abstract]
  92. Fukunishi I, Hosokawa K. A female case with the Kleine-Levin syndrome and its physiopathologic aspects. Jpn J Psychiatry Neurol. 1989 Mar. 43 (1):45-9. [View Abstract]
  93. Kleine W. Periodische schlafsucht. Monatsschr Psychiatr Neurol. 1925. 285-320.
  94. Smolík P, Roth B. Kleine-Levin syndrome ethiopathogenesis and treatment. Acta Univ Carol Med Monogr. 1988. 128:5-94. [View Abstract]
  95. Abe K. Lithium prophylaxis of periodic hypersomnia. Br J Psychiatry. 1977 Mar. 130:312-3. [View Abstract]
  96. Itokawa K, Fukui M, Ninomiya M, Yamamoto T, Imabayashi E, Tamura N, et al. Gabapentin for Kleine-Levin syndrome. Intern Med. 2009. 48 (13):1183-5. [View Abstract]
  97. Peraita-Adrados R, Vicario JL, Tafti M, García de León M, Billiard M. Monozygotic twins affected with Kleine-Levin syndrome. Sleep. 2012 May 1. 35 (5):595-6. [View Abstract]
  98. Ueno T, Fukuhara A, Ikegami A, Ohishi F, Kume K. Monozygotic twins concordant for Kleine-Levin syndrome. BMC Neurol. 2012 May 30. 12:31. [View Abstract]
  99. Billiard M, Guilleminault C, Dement WC. A menstruation-linked periodic hypersomnia. Kleine-Levin syndrome or new clinical entity?. Neurology. 1975 May. 25 (5):436-43. [View Abstract]
  100. Rocamora R, Gil-Nagel A, Franch O, Vela-Bueno A. Familial recurrent hypersomnia: two siblings with Kleine-Levin syndrome and menstrual-related hypersomnia. J Child Neurol. 2010 Nov. 25 (11):1408-10. [View Abstract]
  101. Sachs C, Persson HE, Hagenfeldt K. Menstruation-related periodic hypersomnia: a case study with successful treatment. Neurology. 1982 Dec. 32 (12):1376-9. [View Abstract]

Primary hypersomnia. Polysomnographic study demonstrates apnea (absence of carbon dioxide fluctuation indicating no flow), chest wall paradox, abrupt increase in tidal volume at the end of apnea, and oxygen desaturation. All of these features are consistent with obstructive sleep apnea.

Primary hypersomnia. In contrast to obstructive sleep apnea, mixed apnea shows absence of respiratory efforts in the first segment of the apnea.

Primary hypersomnia. Periodic limb movements show intermittent leg electromyogram activity accompanied by electroencephalogram arousals.

Primary hypersomnia. Polysomnographic study demonstrates apnea (absence of carbon dioxide fluctuation indicating no flow), chest wall paradox, abrupt increase in tidal volume at the end of apnea, and oxygen desaturation. All of these features are consistent with obstructive sleep apnea.

Primary hypersomnia. In contrast to obstructive sleep apnea, mixed apnea shows absence of respiratory efforts in the first segment of the apnea.

Primary hypersomnia. Periodic limb movements show intermittent leg electromyogram activity accompanied by electroencephalogram arousals.

PrimarySecondary
Narcolepsy type 1 (NT1)



Narcolepsy type 2 (NT2)



Hypersomnia due to a medical condition



Hypersomnia due to a psychiatric condition



Idiopathic hypersomnia (IH)Hypersomnia due to a drug or substance
Kleine-Levin syndrome (KLS)Insufficient sleep syndrome (ISS)