Abstract
Scientific evidence that acute, posttrauma sleep disturbances (eg, nightmares and insomnia) can contribute significantly to the pathogenesis of trauma-induced disorders is compelling. Sleep disturbances precipitating from trauma are uniquely predictive of daytime posttrauma symptom occurrence and severity, as well as subsequent onset of mental health disorders, including post–traumatic stress disorder. Conversely, adequate sleep during the acute posttrauma period is associated with reduced likelihood of adverse mental health outcomes. These findings, which are broadly consistent with what is known about the role of sleep in the regulation of emotion, suggest that the acute posttrauma period constitutes a “window of opportunity” during which treatment of sleep disturbances may be especially effective for preventing or mitigating progression of aberrant psychophysiological processes. At this point, the weight of the scientific evidence supporting this possibility warrants initiation of clinical trials to confirm the benefits of targeted prophylactic sleep enhancement, and to establish treatment guidelines as appropriate.
Citation:
Swift KM, Thomas CL, Balkin TJ, Lowery-Gionta EG, Matson LM. Acute sleep interventions as an avenue for treatment of trauma-associated disorders. J Clin Sleep Med. 2022;18(9):2291–2312.
Keywords: post–traumatic stress disorder (PTSD), acute stress disorder (ASD), acute stress reaction (ASR), pharmacological treatment, behavioral treatment, sleep disturbance
INTRODUCTION
While the factors that precipitate most mental health disorders are often ill-defined, traumatic stress–associated disorders, by definition, stem from exposure to traumatic events. Following trauma, most individuals acutely experience hyperarousal and emotional distress—symptoms that dissipate with time.1,2 However, for a subset of individuals, initially severe trauma-related symptoms (acute stress reaction, or ASR) may be followed by development of trauma-associated disorders such as acute stress disorder (ASD) and post–traumatic stress disorder (PTSD).3 Despite knowing the origin of trauma, the pathogenesis from traumatic event to disease state is not well understood. Prior work suggests that individuals with sleep disturbances are at greater risk for developing trauma-related symptoms, and that sleep disturbance is present throughout the development of traumatic stress–related disorders.4 Accordingly, sleep abnormalities may have predictive value for determining the risk of developing trauma-associated disorders. If so, then it is also possible that treatment of sleep disturbance following trauma exposure may prevent the onset or reduce the severity of trauma-related disorders.
The objectives of this review are to assess the evidence that peri-trauma sleep disruption contributes to the onset of trauma-associated disorders, and to determine the extent to which treatment of sleep disturbances potentially prevents or alleviates traumatic stress–associated symptoms. We review the myriad sleep changes that arise following trauma and persist in ASD and PTSD, as well as those occurring in the absence of a psychiatric diagnosis. Additionally, we examine evidence that disordered sleep is a predisposing factor for maladaptive responses to traumatic stress exposure. Finally, we discuss the possible utility of current and potential treatments for sleep disturbance if administered early following trauma exposure to alleviate psychiatric symptoms. Overall, we review the evidence pertinent to the possibility that early detection and treatment of sleep disturbance following trauma may prevent the development, or reduce the severity, of traumatic stress–related disorders, thereby promoting better long-term mental health outcomes.
TRAUMATIC STRESS AND TRAUMATIC STRESS–RELATED DISORDERS
The defining diagnostic criterion for ASD and PTSD is exposure to a precipitating traumatic event. A trauma is any event that imparts an actual or perceived threat of death, serious injury, or sexual violence.5 In the immediate aftermath of a traumatic event, some individuals present with an ASR. Symptoms of ASR include emotional distress, hyperarousal, trauma cue or context avoidance, and trouble sleeping.3,6,7 These symptoms are often short-lived, persisting for no more than 48 hours.3,8 Although ASR is not formally recognized by the DSM-5 (Diagnostic and Statistical Manual of Mental Disorders, fifth edition),5 it is included in the World Health Organization’s ICD-10 (International Classification of Diseases, 10th Revision).9 The relatively broad ICD-10 definition of ASR encompasses the DSM-5 definitions of both ASR and ASD, except for the qualifier that if symptoms persist for 3–30 days, ASD can be diagnosed. Individuals with ASD often present with heightened arousal or anxiety, a dissociative or detached state, trauma-related flashbacks, and sleep disruption.5,8 If posttrauma symptoms persist for longer than 1 month, PTSD can be diagnosed. PTSD symptoms are divided into 4 subclusters (intrusion, avoidance, cognition and mood changes, and alterations in arousal and reactivity), all of which must be present for a diagnosis.5 Symptoms within each subcluster vary in manifestation, resulting in a wide variety of symptom presentations. Symptoms of PTSD can persist for decades after a traumatic event; chronic PTSD is often a debilitating condition that is treatment-resistant and that rarely remits spontaneously.10
Although the trajectory from a traumatic event to ASR, ASD, and PTSD seems linear, this is not always the case. There is conflicting evidence as to whether ASR symptoms are predictive of later ASD and/or PTSD development.1,11–15 Likewise, it is unclear whether ASD reliably predicts PTSD. A review of 22 studies of ASD found that 50% of patients with ASD later met criteria for PTSD.16 However, individuals who later develop PTSD are not always diagnosed with ASD following trauma, so although ASR and ASD may precede PTSD, they do not always precede development of PTSD. It remains unclear if inconsistent relationships between ASR, ASD, and PTSD reflect diagnostic limitations or actual individual variations in the time course upon which symptoms develop. It is, therefore, perhaps more informative to assess specific symptoms that are predictive of trauma-related disorder development. An emerging body of literature suggests that peri-trauma sleep disturbances are predictive of trauma-related symptoms and the development of PTSD17 Therefore, understanding how sleep is altered by trauma exposure may help to identify at-risk individuals and possibly inform development of early sleep-enhancing treatment strategies to prophylactically reduce overall trauma-related symptomology and the pathogenesis of PTSD.
SLEEP DISTURBANCE AND THE EMERGENCE OF TRAUMATIC STRESS SYMPTOMS
Disordered sleep is a hallmark symptom of trauma-related disorders and is included in the diagnostic criteria for both ASD and PTSD.5 However, the pathophysiological processes by which posttrauma sleep disruption reduces adaptive responses to trauma are currently unknown. What is known is that sleep loss or disruption interferes with normal neurological and homeostatic processes and increases neuroinflammation in animals and β-amyloid deposition in humans.18–20 In fact, chronic sleep disruption has detrimental effects on human health in general,21 including on neural protective processes that mediate adaptation to physical and psychological stressors.22 Conversely, improving disrupted sleep can improve physical and mental health.23 Prior literature supports the role of sleep in reducing symptoms of PTSD and anxiety, possibly via improved regulation of neural pathways that modulate emotion.24 While this emotional dysregulation may manifest as simple moodiness in day-to-day life, following trauma it can potentially increase the severity of posttrauma symptoms.24 Additionally, disrupted sleep is intrinsically stressful.22 Accordingly, it is not surprising that disturbed sleep can synergistically compound other trauma-related effects and exacerbate mental health outcomes.25
Due to the unpredictability of traumatic stress exposure, few studies have quantified the acute impact of trauma on human sleep, and no study has utilized polysomnography to measure early posttrauma changes in sleep parameters. Patient self-reports indicate that ∼35% of trauma survivors experience nightmares and ∼58% experience sleep disturbances within 24 hours following a traumatic event, with nocturnal sleep disturbance remaining elevated during the following week.7,26 There is limited evidence from 2 actigraphy studies that captured objective measures of sleep-wake activity (ie, sleep duration, continuity, and timing) in small populations before and immediately after trauma (eg, missile attacks, earthquake).27,28 Patients experienced lower than typical sleep efficiency, abnormally extended sleep onset latency, and increased wake after sleep onset. However, these studies did not assess whether trauma imparted any lasting sleep changes or whether—and the extent to which—sleep parameters predicted or reflected posttrauma psychopathology.
Trauma can also produce lingering, deleterious changes to sleep regardless of whether ASD or PTSD is subsequently diagnosed.29,30 Individuals exposed to missile attacks during the Gulf War reported an increased prevalence of insomnia several months after the war.31 Persistent insomnia and sleep disturbances have also been reported in survivors of earthquakes and the 9/11 terrorist attacks.32–34 Early life trauma can result in sleep disturbances years afterward35,36 and is associated with increased sleep disturbance in adulthood.30 Similarly, a study of Holocaust survivors with and without traumatic stress diagnoses revealed self-reported sleep disturbances decades later, with the severity of sleep disturbance related to the amount of time imprisoned in concentration camps.37 It is thought that sleep disturbance initiated by a traumatic experience can be maintained by self-perpetuating factors that accompany insomnia, such as maladaptive thoughts and behavioral patterns surrounding sleep.38 Overall, the existing literature suggests that long-term trauma-induced sleep disturbance is sometimes, but not always, associated with diagnosable trauma-related mental health problems, irrespective of the trauma type, population, or duration.
The presence of chronic, trauma-related sleep disturbances in participants who do not meet criteria for a mental health disorder diagnosis has led to the proposal of a new parasomnia: trauma-associated sleep disorder (TSD).39 TSD is a putative parasomnia characterized by nightmares, disruptive nocturnal behavior (eg, thrashing or tossing), dream-enactment behavior, REM sleep without atonia, and enhanced sympathetic activity during sleep.40,41 Some of the symptoms of TSD are similar to those exhibited by patients with nightmare disorder and rapid eye movement (REM) sleep behavior disorder (RBD), but unlike these other disorders, TSD is characterized by dream-enactment behaviors and complex vocalizations during nightmares that are precipitated by a specific, identifiable traumatic event. Further work is warranted to better understand and validate TSD as a formal diagnosis, but the data cited in support of this putative diagnosis nevertheless serves as additional evidence that long-term sleep disturbance can result from exposure to traumatic events.
Although sleep disturbance is among the diagnostic criteria for ASD, few studies have examined how self-reported sleep is changed in ASD, and no studies have yet documented ASR- or ASD-associated changes with objectively measured sleep parameters (eg, polysomnographically). In 1 of the few studies in which ASD and sleep were both assessed, 91% of ASD patients reported difficulty sleeping and 73% reported nightmares.42 However, in this and other studies based on data collected from case reports of individuals admitted to hospitals following bodily injury, it is unclear whether, or the extent to which, the reported sleep disturbances were due to ASD or due to physical pain following traumatic injury.43–45 Despite a dearth of objective data on sleep and ASD, the previously described research suggesting that sleep loss exacerbates the stress response makes plausible the hypothesis that ASR and ASD constitute an early window during which sleep-promoting intervention might help prevent the development, or mitigate the long-term effects, of PTSD.
The long-term impact of trauma on sleep is evident from numerous studies documenting sleep disturbances in PTSD. While there is consensus that PTSD impacts sleep, there is less agreement regarding which sleep parameters are affected.46–48 Commonly self-reported sleep disturbances in PTSD patients include interrupted sleep, insomnia, and a general feeling that sleep is nonrestorative.49–52 Objective studies of PTSD’s impact on sleep vary by study, but some general themes are evident in the scientific literature: REM sleep is dysregulated in PTSD, with both increased and decreased amounts of REM having been reported.53–62 The dysregulation of REM sleep may be linked to the increased prevalence of trauma-related nightmares that are one of the hallmark symptoms of PTSD.51,52,63–65 Individuals with PTSD also tend to spend more time in, or exhibit more transitions to, light (stage N1) non-REM sleep at the expense of deep (stage N3) non-REM sleep.62,63,66–69 Total sleep time and sleep efficiency are decreased in PTSD patients—potentially due to an increased rate of arousals from sleep.53,54,56,59,63,66–68,70–75 Additionally, PTSD has a high comorbidity with obstructive sleep apnea (OSA).76 OSA can worsen existing trauma-related sleep disruptions and contribute to neuroendocrine and metabolic dysregulation commonly present in PTSD.76 As with other PTSD symptoms, the sleep-related symptoms of PTSD also vary by sex. Women with PTSD display an increased percentage of REM sleep as well as reduced total sleep time compared to women without PTSD.61,62 Men with PTSD display an increased percentage of N2 sleep and a reduced percentage of N3 sleep compared to healthy men. Compared to men with PTSD, women with PTSD have increased total sleep time but also increased wake after sleep onset. These sex-specific differences may partially explain the differential rates of PTSD in men (4%–5%) vs women (9%–14%) reported in the National Comorbidity Survey in 1990–1992.77 Overall, the breadth of literature regarding sleep disturbance characteristics in PTSD patients may inform the development of putative targets for intervention, whether at early or later time points following traumatic stress. For example, if a novel pharmacotherapy is observed to normalize REM sleep, based on the existing literature of sleep and PTSD, it might be hypothesized that this compound could then be used in a targeted way to improve both sleep and other trauma-related symptoms.
In addition to being a symptom of trauma exposure, sleep disturbances may increase the vulnerability to PTSD and/or be a marker of subthreshold PTSD. Overall, several studies have provided evidence that poor sleep in proximity to trauma may contribute to the pathogenesis of PTSD.4,73,78,79 Patients who reported sleep issues before experiencing a hurricane or bodily injury were at increased risk of developing PTSD, panic disorder, and other psychiatric symptoms.73,80 Similarly, studies of veterans have shown that sleep complaints prior to a combat deployment are associated with onset and severity of PTSD and depression symptoms during postdeployment, and these associations remain even when controlling for predeployment psychiatric symptoms.78,81,82 Sleep disturbances precipitating from a traumatic event also increase the risk of developing symptoms of PTSD. Studies of combat veterans have shown that postdeployment sleep disturbances are associated with symptoms of PTSD, depression, and substance abuse 3–12 months following deployment.83–85 Likewise, survivors of automobile accidents and political violence have demonstrated sleep disturbance in the aftermath of trauma that was associated with the development of PTSD symptoms up to a year later.86,87 Interestingly, while early sleep issues are predictive of later PTSD symptom severity, early PTSD symptoms are not associated with the later development of sleep disturbances—suggesting that sleep complaints following trauma may be an early marker for subsequent PTSD diagnosis.87 Although these studies provide a cohesive narrative regarding the impact of sleep disturbances on PTSD onset, their interpretation is limited by the use of self-reported sleep measures. Of course, the unpredictable occurrence of trauma limits the feasibility of recording objective measures of sleep prior to traumatic events. More feasible would be the collection of self-reported sleep measures combined with the objective sleep data from wearable devices across multiple time points in populations at high risk for exposure to trauma. Together, self-reported and objective data might be used in future studies to further elucidate the relationship between sleep disturbance and the pathogenesis of trauma-related disorders.
Despite limitations, the current literature suggests that: a) sleep disturbance arising from trauma is commonly present throughout the development of trauma-related disorders, and likely contributes to both the onset and maintenance of these disorders17; b) adequate sleep reduces the risk of developing PTSD and other mental health disorders23,88; and c) sleep disturbance (as might be measured using a variety of metrics such as those provided by commercial fitness trackers) occurring in close temporal proximity to a traumatic event is maladaptive and associated with poor mental health outcomes. Accordingly, it is reasonable to hypothesize that treatments which improve sleep continuity and quality, or that normalize sleep may be beneficial at all time points, regardless of the duration of the trauma-associated disorder. More work is needed to understand how specific changes in sleep exacerbate traumatic stress symptoms, particularly those changes acutely following a traumatic event, and to determine whether, and the extent to which, sleep phenotype is predictive of susceptibility to posttrauma psychopathology. Such knowledge would assist in the development of interventions that are tailored for individual sleep phenotypes.
SHOULD INDIVIDUALS SLEEP IMMEDIATELY FOLLOWING TRAUMATIC STRESS?
Although the scientific literature generally suggests that sleep promotes psychological resilience, there is also some evidence that sleep deprivation immediately following trauma may be adaptive and constitute a useful strategy for mitigating subsequent mental health problems. The use of sleep deprivation posttrauma has not yet been tested and validated as a treatment method at this time, as a number of clinical studies would be needed to test the effectiveness of this approach. Based mainly on findings from a broad scientific literature demonstrating that sleep facilitates the consolidation of memories,89,90 it has been hypothesized that posttrauma sleep loss should interfere with consolidation of the contextual fear and emotionally charged memories,91,92 thereby reducing their negative effects. This hypothesis has received some support. Two studies utilizing films to induce analog trauma found that sleep deprivation after viewing the films reduced generalized fear responses, intrusive memories, and scores on the Impact of Event Scale.93,94 Similar beneficial effects of sleep deprivation have been demonstrated in human and rodent studies, producing evidence that weakening the memory of the traumatic event reduces the subsequent impact on markers of stress.
However, such studies may not be adequately representative of real-world traumatic experiences. The contextual fear associated with a real-world trauma in which there is a perceived threat to life likely produces a much more salient fear association than that produced by laboratory contextual fear tasks, which are limited to using stimuli that are no more than mildly aversive. Similarly, it is difficult to draw direct comparisons between laboratory tasks evaluating emotional memory formation and traumatic memory formation. Emotional memory-based laboratory tasks commonly require participants to identify and recall information about contrasting neutral and negatively charged stimuli. While these tasks are useful for evaluating the formation of memories with an emotional valence, the extent to which findings from these studies are generalizable to real-world traumatic experiences—which almost by definition involve exposure to actual danger and/or significant injury and pain—is likely limited. Also, laboratory studies invariably utilize healthy participants who are not in a state of posttrauma hyperarousal. Following actual trauma, heightened sympathetic activity, particularly noradrenergic activity, may promote hyperconsolidation of highly salient traumatic experiences95—making subsequent sleep-dependent memory consolidation processes less critical for establishing the traumatic memory in the short term, and contributing to the persistence of these memories even years after the trauma occurred. Because it is not clear that memory consolidation processes for real-world trauma are qualitatively comparable to memories produced by exposure to relatively mild stressors in laboratory settings, and because ethical considerations preclude exposing study participants to truly traumatic events (eg, exposing participants to actual physical danger), generalization from such studies is problematic, and the potential real-world utility of posttrauma sleep deprivation as a therapeutic intervention remains uncertain.
Even more problematic for the hypothesis that sleep deprivation may be beneficial is the finding from prior studies that sleep deprivation following fear learning and analog trauma fails to improve psychological outcomes. Sleep deprivation after viewing a trauma film increased or had no effect on the number of intrusive memories.96,97 Prior research has also shown that while positive or neutral memories are susceptible to sleep deprivation, negative memories are relatively resistant.98 Additionally, sleep deprivation following cued fear learning impairs the ability to recall extinction of the fear memory, a deficit that is similar to what is thought to occur in PTSD,99 and probably contributes to the persistence of this disorder. Conversely, some studies in both rodents and humans have shown that adequate sleep following trauma exposure promotes better outcomes.88,97 For example, optogenetically enhancing sleep in rats following a model of fear trauma (exposure followed by extinction) improves recall of extinction to fear.100 In humans, sleep after viewing a traumatic film reduces the subsequent number of intrusive memories and level of distress.97 In fact, studies have shown that improving sleep reduces mental health symptoms across a variety of conditions (particularly in those with comorbid sleep disruption).23 It is, therefore, reasonable to hypothesize that sleep enhancement immediately following traumatic stress exposure reduces, rather than exacerbates, negative mental health outcomes.
Several prior studies have indeed found that adequate sleep following trauma exposure is associated with better mental health outcomes,88,97 whereas sleep disturbance correlates positively with overall symptom severity in trauma-related disorders. Patients with ASD or PTSD often present with sleep-related complaints, although these complaints are not always recognized as being trauma-related—especially when nightmares are not a primary complaint. A number of studies have found that the severity of PTSD symptoms varies as a function of the severity of sleep disturbance,101 and treatment of sleep disturbance often improves other PTSD symptoms.102,103 Therefore, addressing sleep disturbances, especially during the early posttrauma phase, may reduce trauma-related symptoms and prevent or mitigate PTSD pathogenesis. Despite the evidence-based logic behind this approach, few studies have evaluated treatments for acute sleep disturbances following trauma exposure.
INTERVENTIONS TO TREAT SLEEP DISTRBANCE AND TRAUMATIC STRESS–RELATED SYMPTOMS
Current guidelines for treatment of some aspects of sleep disturbance in PTSD exist, but they do not specifically address adverse sleep changes early after trauma.104–106 For posttrauma symptoms persisting beyond 2 days, existing guidelines recommend a brief 4- to 5-session trauma-focused cognitive behavioral therapy with additional tailored management of other acute symptoms (eg, pain, rage, hyperarousal, anger, etc).107 Several guidelines also recommend the use of psychological first aid in the early aftermath of trauma.107,108 Though psychological debriefing has been utilized in the immediate aftermath of trauma, existing treatment guidelines do not recommend its use due to mixed evidence supporting its efficacy.108,109 Tailored treatments for acute sleep disturbances, including relaxation techniques and sleep hygiene education, are also recommended with the caveat that there is little evidence for their effectiveness in the acute phase.108 In the clinical setting, sleep-related treatments for PTSD may be administered by primary care providers, behavioral health providers, or sleep medicine physicians. The approaches taken by these professionals vary according to their training. Generally, providers make efforts to normalize sleep. Clinicians, regardless of specialty, are familiar with sleep hygiene practices, which include obtaining 7–8 hours of sleep, setting structured wake and sleep times, making the bedroom environment optimal for sleep, and avoiding activities or substances that may impact sleep. However, sleep hygiene changes as a stand-alone treatment for insomnia and sleep disturbances has not been shown to be effective.110 Patients who have experienced trauma may need a multidisciplinary approach to improve outcomes.
Therapies used to treat sleep in PTSD are largely unevaluated in addressing sleep disturbances arising early after trauma. We assess current nonpharmacologic and pharmacologic treatment recommendations for PTSD and gauge their potential applicability for sleep-specific symptoms in the acute period (immediately to 30 days) following trauma (see Table 1). Ideally, any of the discussed treatments could be initiated in the days following trauma in individuals demonstrating sleep disturbance to both regulate sleep and support recovery from the traumatic exposure. As there is very little existing evidence collected during the acute period after trauma exposure, evidence from clinical trials would be needed to support early use of both behavioral and pharmacologic treatments.
Table 1.
Modality | Therapeutic Agent | Indications | Benefits | Contraindications/Risks | Pitfalls | Sleep Effects | Clinical Trials |
---|---|---|---|---|---|---|---|
Medications | Selective serotonin reuptake inhibitors (sertraline, paroxetine) | PTSD | Improvement in daytime PTSD symptoms |
|
|
|
|
Medications | Serotonin norepinephrine reuptake inhibitors (venlafaxine) | PTSD | Improvement in daytime PTSD symptoms |
|
|
|
|
Medications | Serotonin antagonist and reuptake inhibitors (trazodone, nefazodone) |
|
|
|
Mixed results on objective measures of sleep disturbance in PTSD |
|
NCT03668041 |
Medications | Atypical antipsychotics (quetiapine, olanzapine, risperidone) | Off-label use for PTSD associated nightmares and sleep disturbances |
|
|
|
|
|
Medications | α-1 Adrenergic receptor antagonist (prazosin) | Off-label use for nocturnal symptoms of PTSD (ie, nightmares and hyperarousal) |
|
Orthostatic hypotension |
|
May improve sleep quality in patients with elevated sympathetic activity | NCT03997864 |
Medications | α-2 Adrenergic receptor agonist (clonidine) | Off-label use for nocturnal symptoms of PTSD | Decreases nightmare frequency and improves PTSD-related sleep disturbances in patients who do not response to prazosin | Orthostatic hypotension | Suppresses REM sleep |
|
NCT04877093 |
Medications | Benzodiazepine γ-aminobutyric-A receptor agonists |
|
Improves anxiety and insomnia associated with PTSD |
|
|
|
|
Medications | Non-benzodiazepine Z-drug γ-aminobutyric-A receptor agonists (zolpidem, zopiclone, and eszopiclone) | Sleep onset or sleep maintenance insomnia | Improved sleep with less risk of abuse and dependence than benzodiazepines |
|
Mixed evidence to support use in trauma-exposed populations |
|
|
Medications | Dual orexin receptor antagonists (suvorexant, lemborexant) | Sleep maintenance insomnia | Improved sleep with less risk of abuse and dependence than benzodiazepines. Do not suppress REM sleep. |
|
No evidence in trauma-exposed populations |
|
NCT02704754, NCT03642028, NCT02849548 |
Medications | Synthetic cannabinoid (nabilone) | Experimental use for PTSD and insomnia |
|
|
|
May promote sleep by activation of cannabinoid type 1 receptors | |
Medications | NMDA (N-methyl-d-aspartic acid) receptor antagonist (ketamine) |
|
|
|
Mixed evidence for use in trauma-exposed populations | Beneficial effect on mood may be linked to its ability to alter sleep-wake behavior and the amplitude of circadian systems—potentially by modulating expression of circadian rhythm genes254,257 | NCT04032301, NCT04889664, NCT04771767 |
Medication-assisted therapy | β-Adrenergic receptor antagonist (propranolol) | Experimental use for acute posttrauma treatment to prevent development of PTSD | May prevent development of PTSD posttrauma due to administration before or after memory reactivation sessions in the context of exposure-based therapy |
|
Mixed evidence for use in trauma-exposed populations |
|
NCT03752918 |
Medication-assisted therapy | 3,4 Methylenedioxy methamphetamine (MDMA)–assisted psychotherapy | Experimental use for PTSD that targets memory consolidation and fear response which has been associated with improved self-reported sleep |
|
|
May increase bruxism, anxiety, and jitteriness, which could potentially cause sleep disturbance | Improved self-reported sleep | |
Therapy | Cognitive Behavioral Therapy for insomnia | Sleep onset or sleep maintenance insomnia | Long-term sleep improvements compared to pharmacotherapy alone and improvement of overall PTSD severity | Contraindicated in patients with bipolar disorder, epilepsy, and those at high risk for falls. |
|
Improved sleep onset latency, WASO, and sleep efficiency | |
Therapy | Brief behavioral therapy for insomnia (BBTi) | Sleep onset or sleep maintenance insomnia |
|
Contraindicated in patients with bipolar disorder, epilepsy, and those at high risk for falls. | No evidence for trauma-exposed populations |
|
|
Therapy | Imagery rehearsal therapy (IRT) | PTSD-related nightmares |
|
Contraindicated in patients with cognitive deficits, severe mental illness, and high levels of anxiety, stress, or avoidance | Limited evidence for use in acute stress disorder | Decreased nightmares and hyperarousal leading to improved sleep outcomes | |
Therapy | Prolonged exposure (PE) therapy | PTSD and ASD |
|
Contraindicated if imminent threat of suicidal or homicidal behavior, recent (past 3 months) serious self-injurious behavior, and current psychosis |
|
Effects on objective measure of sleep are unknown. | |
Therapy | Cognitive processing therapy (CPT) | PTSD | Cognitive intervention which assists trauma survivors to contextualize and process the highly negative affect surrounding a traumatic event in an adaptive manner | Contraindicated in psychotic patients, in the presence of substance dependence, if severe dissociative reactions or panic attacks, if imminent risk of self-harm, and if the patient is in abusive relationship |
|
Effects on objective measure of sleep are unknown. | |
Therapy | Eye movement desensitization and reprocessing (EMDR) | PTSD | Exposure-based psychotherapy that involves the recollection of traumatic events while focusing on external stimuli (lateral eye movements) in order to decouple the emotional saliency from the recall of traumatic events |
|
|
Effects on objective measure of sleep are unknown. | |
Therapy | Accelerated resolution therapy (ART) | PTSD |
|
|
Limited evidence supporting use for trauma-related sleep disturbances | Effects on objective measure of sleep are unknown. | |
Technologies | Transcranial magnetic stimulation (TMS) | PTSD | Utilizes electromagnetic currents on the skull to improve daytime PTSD symptom severity |
|
No evidence for acute stress disorder | May improve sleep quality through promoting slow wave sleep and REM sleep and inhibiting a hyperarousal state in the cerebral cortex |
Behavioral and nonpharmacologic therapies
Cognitive behavioral therapy for insomnia (CBTi) is a first-line recommended treatment for insomnia because, compared to pharmacotherapy alone, it actually produces superior, long-term improvements in sleep.104,111 A meta-analysis of randomized controlled trials of CBTi for sleep disturbances in PTSD revealed that these improvements include shorter sleep onset latency, reduced wake after sleep onset, and improved sleep efficiency, concomitant with reductions in in daytime PTSD symptom severity.112 Thus, although pharmacotherapy is superior for providing relief from insomnia acutely, CBTi provides more sustained relief.113,114 The downsides to CBTi are: a) that it requires more effort by the patient, and b) it can take several weeks for benefits to manifest.115 While this is less of a concern in treating PTSD, the increased length of treatment limits the usefulness of CBTi for treatment of acute sleep disturbances resulting from trauma.
Nevertheless, there may be a role for short courses of CBTi (2–12 weeks), possibly as adjunctive therapies, for the treatment of acute, posttrauma sleep disturbance,112 since they have also been found to be effective at reducing insomnia in PTSD patients and have relatively low attrition rates.112 Stimulus control and sleep restriction, 2 components of CBTi, have been identified as evidence-based, stand-alone treatments for insomnia and may require less time to implement, although they may not have the durability of CBTi in the long-term outcomes.116–118 Brief behavioral therapy for insomnia (BBTi) is a manualized 4-session treatment module that focuses on stimulus control and sleep restriction. It was designed for medical providers who did not have the time to implement CBTi and is associated with improvement of sleep efficiency both at early and late follow-up.119 Limited research exists for the use of BBTi in the setting of acute trauma or PTSD.120
It should be noted that if a patient is undergoing exposure-related treatments for PTSD, hyperarousal and nightmares may initially, albeit temporarily, be exacerbated by these treatments.106 For patients with nightmare-related issues, providers will often prescribe a form of cognitive behavioral therapy called imagery rehearsal therapy (IRT), which focuses on imagery rescripting and imaginal exposure.121 Other techniques include systematic desensitization and progressive muscle relaxation to assist patients with sleep initiation, especially in patients with prominent hypervigilance symptoms. These nonpharmacologic techniques are generally paired with pharmacotherapy addressing hypervigilance and sleep complaints, such as prazosin or clonidine.106 One study identified a reduction in nightmares at 6 months follow-up after 3 sessions of IRT (two 3-hour sessions 1 week apart with a 1-hour follow-up 3 weeks later), whereas other studies demonstrated sustained benefit after a single IRT session.122,123 Therefore, is it possible that IRT can be implemented early after trauma exposure with long-term effects on sleep outcomes.
Prolonged exposure (PE) therapy is a cognitive therapy designed to treat symptoms of PTSD. PE effectively reduces daytime symptoms of PTSD and is a recommended first-line treatment for PTSD.124 Immediate treatment with PE within 12 hours of trauma exposure reduces the severity of posttrauma symptoms at 4 weeks and 12 weeks.125 Similarly, early treatment of ASD patients with PE reduces the likelihood of later developing PTSD.126 However, the effects of PE on trauma-related sleep disturbances are mixed. One study found that PE improves self-reported sleep quality in PTSD with sustained improvements for at least 1 year,127 while another found that PE does not improve sleep in PTSD.128 Further, preexisting sleep disturbances decrease the efficacy of PE to improve symptoms of PTSD.129,130 While PE is effective when initiated acutely following trauma to prevent PTSD, it does not have rapid effects, and additional studies are necessary to determine whether PE is effective for treating acute trauma-related sleep disturbance.
Cognitive processing therapy (CPT) is a cognitive intervention that assists trauma survivors in addressing and contextualizing the highly negative affect surrounding a traumatic event, thereby allowing them to process the event in an adaptive manner. CPT is effective at treating symptoms of PTSD and trauma exposure; however, studies assessing the efficacy of CPT to address PTSD-related sleep disturbances have produced mixed results.131–134 Limited evidence suggests that augmenting CPT with a sleep-directed therapy improves sleep disturbances and overall PTSD symptoms, but sleep improvements are not attributed to CPT.135,136 Additionally, during the treatment course with CPT, hyperarousal may precede improvement in certain PTSD symptom domains.132 Hyperarousal may impair the individual’s ability to fall or stay asleep, which may initiate or perpetuate current sleep problems. Therefore, CPT is highly manualized and is more widely available clinically, but current studies do not support its use in addressing trauma-related sleep disturbance. Additional evidence is needed to make definitive statements regarding the use of CPT for both acute trauma and sleep disturbance.
Eye movement desensitization and reprocessing (EMDR) is an exposure-based psychotherapy that involves the recollection of traumatic events while focusing on external stimuli. During EMDR, the patient is directed through a series of lateral eye movements that allow the patient to decouple the emotional saliency from the recall of traumatic events. It is hypothesized that the eye movements associated with EMDR mimic the eye movements in REM sleep, allowing for the adaptive processing of traumatic memories.137,138 However, 1 randomized clinical trial demonstrated that the PTSD symptoms reduction was a result of an external focus of attention, regardless of whether eye movements were implemented or eyes were fixed on a nonmoving hand.139 EMDR has been shown to be equivalent to, if not superior to, cognitive behavioral therapy for the treatment of PTSD.140 EMDR takes about the same number of sessions as PE for combat-associated trauma. It is therefore not a rapid treatment, but it could be administered acutely following a traumatic event. Accelerated resolution therapy (ART) is an adapted form of EMDR for PTSD that typically requires 1–5 one-hour sessions over 3 weeks, and it has been shown to be effective with large effect sizes as measured on the PTSD Checklist for DSM-5.141,142 Some preliminary evidence suggests that administration of one 45- to 60-minute session within 96 hours of a traumatic event results in a significant reduction of symptoms in patients with ASRs.143 More research to investigate the efficacy of ART as a treatment for acute traumatic stress symptoms and sleep disturbance is warranted.
There are a variety of stimulation technologies that have been assessed for their potential utility to treat PTSD, including deep-brain stimulation, vagus nerve stimulation, transcranial direct current stimulation, and transcranial magnetic stimulation (TMS).144 Notably, TMS has shown potential efficacy for PTSD symptomology, and there is mechanistic evidence to suggest it may regulate sleep disturbance. TMS utilizes electromagnetic currents that are placed on the skull and reach the neurons of the cortex, causing depolarization and forcing an action potential.145 TMS has the potential to modulate aberrant maladaptive circuits and is US Food and Drug Administration (FDA)-approved for other neuropsychiatric conditions, including major depressive disorder and obsessive compulsive disorder.146 Preliminary studies, have demonstrated reduced Pittsburgh Quality Index scores in patients with insomnia.147 It is hypothesized that repetitive TMS may improve sleep quality through promoting slow wave sleep and REM sleep, inhibiting a hyperarousal state in the cerebral cortex, altering sleep-related hormones and metabolic activity, and increasing hippocampal neurogenesis.147–150 In the treatment of PTSD patients, medium effect sizes have been observed, particularly when high frequency stimulation was applied over the right dorsolateral prefrontal cortex.151–155 No studies have assessed the efficacy of TMS in an acute trauma setting, but the potential benefits, in terms of promoting sleep and reducing trauma-related symptoms, are considerable.
Overall, few studies have assessed the efficacy of behavioral and nonpharmacologic therapies to treat sleep disturbances acutely posttrauma. While prior studies support the use of CPT, PE, and EMDR to address daytime symptoms of PTSD, the existing evidence does not support their use to address acute sleep disturbances. CBTi is the only behavioral therapy discussed that is specifically designed to treat sleep disturbances. Notably, sleep restriction is a component of CBTi, and its use posttrauma should be carefully considered, as the current literature does not support the use of sleep restriction acutely posttrauma to prevent the consolidation of a traumatic experience. However, once sleep disturbances have set in posttrauma, a targeted sleep restriction under the guidance of a clinician to consolidate nocturnal sleep periods for CBTi would likely prove beneficial. Another consideration of behavioral and nonpharmacological treatments is time. The time course required to produce therapeutic benefits would likely exceed the “acute phase” posttrauma. The combination of pharmacologic therapy to address acute symptoms alongside a nonpharmacologic or behavioral therapy to provide more sustained relief would likely be advantageous. Providing other posttrauma symptoms do not limit which therapies can be applied (eg, exposure therapies should not be initiated in ASD patients expressing severe dissociation or avoidance), initiating these therapies as soon as possible is recommended to target sleep disturbance associated with acute trauma exposure.
Pharmacologic therapies
Several pharmacological guidelines have been established to treat sleep-related symptoms of PTSD, while limited pharmacologic guidelines exist for ASD and for very early acute symptoms (within the first 72 hours).104,106,107 It is likely that most pharmacotherapies recommended for PTSD will be ineffective at treating symptoms of acute trauma, especially trauma-related sleep disturbances, as several treatments recommended for PTSD have the potential to cause sleep disturbance or require a time course too long to be effective. Conversely, several treatments are not recommended for use in PTSD although they are effective at treating acute trauma-related sleep disturbances. We examine current treatment guidelines for PTSD to gauge their effectiveness for addressing trauma-related sleep disturbances (see Table 1).
Selective serotonin reuptake inhibitors (SSRIs) are a class of antidepressants used to treat of a variety of mental health conditions. The SSRIs sertraline and paroxetine are first-line, FDA-approved treatments for daytime PTSD symptoms.156–159 However, SSRIs generally suppress REM sleep and increase REM latency.160 SSRIs also impact NREM sleep by increasing light stage sleep, nighttime arousals, and wake after sleep onset. Additionally, adverse effects of SSRIs may further exacerbate insomnia, including restless legs syndrome, periodic nocturnal limb movements, REM sleep without atonia (RSWA), and, less commonly, RBD.161–163 RSWA is defined as increased tonic or phasic motor tone identified on electromyography channels during REM sleep, whereas RBD is characterized by both RSWA and dream-enacting behavior. Studies have reported as high as a 10-fold risk of RSWA with antidepressant use, and there is increased likelihood as high as 1.9 times the odds of developing RBD.163,164 The mechanism behind the SSRI induction of RSWA and RBD appears to be linked to the complex double-switch model of REM-on and REM-off neurons on the brainstem.165 Some have suggested that SSRIs may just unmask an underlying condition rather than cause RBD because withdrawal of the medication does not always result in resolution of the symptoms.166,167 As previously discussed, PTSD has been associated with what has been proposed to be a new parasomnia called TSD that resembles RBD (except that onset of TSD is clearly precipitated by a traumatic event), and it is unknown whether SSRIs unmask or contribute to the development of this sleep disturbance.
Additionally, several case reports have linked SSRI use to restless legs syndrome, which may be misattributed as anxiety or hyperarousal prior to sleep and thus prolong sleep latency.168,169 However, evidence to support the connection has been mixed.163,170–172 Previous studies have identified that SSRIs induce or worsen periodic limb movements during sleep, which in some patients can increase arousals and contribute to poor sleep quality.173–175 Although SSRIs may take 4–6 weeks to reach full therapeutic effect for PTSD-related symptoms, the effects on REM sleep are most significant early in treatment and diminish over time.176 Therefore, if given for acute trauma victims, SSRIs may impact sleep even before patients experience other trauma-related symptom relief. However, it should be noted that most studies that examine the influence of SSRIs on sleep occur in the context of depressed patients. Depression decreases REM latency and increases REM density during the first sleep cycles, so it is hypothesized that improvement of depression may also be related to REM sleep deprivation.177 REM sleep appears to be dysregulated in PTSD, and evidence suggests that multiple sleep phenotypes occur, with some patients having increased REM sleep and others having decreased REM sleep. It may be that SSRIs impact sleep differently in the setting of acute trauma due to individual variability. However, as SSRIs acutely disrupt sleep and require weeks to reach full therapeutic potency, it is unlikely that they would provide benefit for treating acute sleep disturbance.
Serotonin and norepinephrine reuptake inhibitors (SNRIs) are another class of antidepressants used to treat PTSD.178 The SNRI venlafaxine is used to treat daytime symptoms of PTSD, and like SSRIs, venlafaxine can exacerbate insomnia, increase REM latency, and suppress REM sleep.179 Additionally, SNRIs have been associated with the same sleep disturbances as SSRIs, including restless legs syndrome, periodic limb movements, RSWA, and RBD.163,164,175 In the case of RSWA and RBD, norepinephrine suppresses REM sleep by activating the REM-OFF cells in the locus coeruleus, while at the same time, norepinephrine is required for the activation of REM sleep. This REM sleep deprivation state impacts brain excitability, which is believed to lead to motor activation in the context of REM sleep initiation. Moreover, venlafaxine is ineffective at treating nightmares in PTSD and, like SSRIs, requires several weeks to reach full therapeutic effect.178 Although useful for treating daytime symptoms of PTSD, caution should be used in prescribing both SSRIs and SNRIs in acutely traumatized individuals prior to a PTSD diagnosis. Additional research would be needed to evaluate if normalization of REM could be achieved with SNRI use in acutely traumatized individuals demonstrating increased levels of REM sleep.
Serotonin antagonists and reuptake inhibitors (SARIs) are used to treat depression and anxiety but are more commonly prescribed off-label as a treatment for insomnia due to their sedative effects via antagonism of histamine receptors.180 SARIs are weak inhibitors of serotonin reuptake but are also strong antagonists of postsynaptic 5-hydroxytryptamine subfamily 2 (5-HT2) receptors which promotes sleep continuity.181,182 Studies of the SARIs trazodone and nefazodone in PTSD patients show improved sleep and reduced frequency of nightmares.183–190 However, the effect of nefazodone on objective measures of sleep disturbances in PTSD have been mixed,186,188 which contributes to why trazodone is commonly used to treat sleep disturbances in PTSD.191 While both SARIs are rapidly effective, nefazodone has a risk of hepatotoxicity, and long-term off-label use should be avoided.192 In comparison, trazodone is safer for longer duration of use and helps maintain sleep by decreasing arousals.193 However, in some individuals, trazodone may precipitate increased anxiety symptoms and trazodone’s active metabolite, meta-chlorophenylpiperazine (m-CPP), may cause panic symptoms in a minority of patients with existing mental health conditions.194 Trazodone should be considered as a potential treatment for acute trauma-related sleep disturbances, due to its widely accepted safety and ability to improve sleep continuity. A clinical trial comparing trazodone to other sleep aids in treating sleep disturbances in currently underway (Identifier: NCT03668041), but studies evaluating the utility of trazadone for improving sleep disturbance in acutely traumatized individuals are also necessary.
Atypical antipsychotics are FDA-approved to manage symptoms of schizophrenia but are also used off-label to treat symptoms of ASD and treatment-resistant PTSD.195 Antipsychotics as a drug class are heterogeneous, with some agents being more activating and others possessing more sedating effects.196 With respect to sleep, atypical antipsychotics like quetiapine, olanzapine, and risperidone generally cause mild sedation, which aids in normalizing sleep in trauma-related disorders. Studies of the atypical antipsychotics risperidone, quetiapine, and olanzapine in patients with PTSD have found decreased nightmares and sleep disturbances, typically within 1–4 days of initial administration.195,197–204 Reductions in sleep disturbances are correlated with improvements in overall PTSD symptoms, suggesting that improved sleep from atypical antipsychotics can benefit daytime PTSD symptoms. Atypical antipsychotics are acutely effective and are, therefore, used following trauma exposure and in ASD.205 Studies of risperidone in acutely traumatized patients have found decreased hyperarousal with fewer sleep disturbances and nightmares within 48 hours of administration.44,45 While atypical antipsychotics are effective acutely, chronic use can cause significant metabolic dysregulation and increase the risk of myocardial infarction.206,207 Thus, they may be helpful for managing sleep disruptions and traumatic stress–related symptoms acutely posttrauma but may not be suitable for managing persistent sleep disturbances or PTSD symptoms over long time periods.
The α-1 adrenergic receptor antagonist prazosin is an antihypertensive drug that has been lauded as an off-label treatment for nocturnal symptoms of PTSD. Prazosin is nonsedating and does not improve sleep disturbances in healthy individuals or daytime symptoms in those with PTSD.208 However, in PTSD, where nocturnal central sympathetic activity is elevated,209 prazosin improves sleep disturbances.210–212 Early small-scale studies found that prazosin is particularly effective at alleviating PTSD-related nightmares regardless of age or trauma type.213–220 However, a recent large-scale phase 3 study found that prazosin was ineffective for treating nightmares and sleep disturbances in PTSD.208 As a result, prazosin has been downgraded from a first-line treatment for trauma-related nightmares.104 Prazosin has been effective at relieving sleep disturbances in putative trauma-associated sleep disorder patients, suggesting that it may have wide applicability for treating trauma-related sleep issues.40 While prazosin has a rapid onset, is generally well tolerated, and is safe to use in conjunction with other therapies, it requires titration up to a tolerable level (often greater than 10 mg/day) to target nightmare symptoms due to the fact that postural hypotension has been associated with rapid titration of this medication.210 Studies typically utilize a flexible titration schedule, and in clinical practice, it can take weeks to reach a therapeutic effect. A slow titration may limit the utility of prazosin in some patients with acute trauma exposure. Clinical trials are currently underway (Identifier: NCT03997864), but more research is needed to understand prazosin’s potential efficacy for relieving sleep disturbances acutely after trauma. Given its well established safety profile, prazosin may be a reasonable treatment option for early treatment of sleep disturbances following trauma.
The α-2 adrenergic receptor agonist clonidine is used to treat hypertension but has also been used to treat sleep and behavioral disruptions in attention-deficit hyperactivity disorder due to its mild sedative effects.221 Clonidine improves overall PTSD symptoms but has primarily been used to address nightmares and sleep disruptions in PTSD in patients who do not respond to prazosin.222,223 In PTSD, clonidine decreases nightmare frequency and improves sleep quality and sleep latency.222,224–226 However, clonidine suppresses REM sleep in healthy individuals and in individuals with PTSD,227 which could exacerbate existing REM changes resulting from trauma. A clinical trial is currently underway (Identifier: NCT04877093) to assess the use of low dose clonidine in assessing daytime and night symptoms of PTSD, but further research is necessary to evaluate the use of clonidine to improve sleep disturbance acutely following trauma.
The β-adrenergic receptor antagonist propranolol has also been proposed as an acute posttrauma treatment to prevent the development of PTSD.228 However, studies have failed to replicate initial findings suggesting propranolol administered acutely posttrauma prevents the development of PTSD symptoms.229–231 Propranolol has a beneficial effect when administered before or after memory reactivation sessions, which suggests that more research should examine the timing of propranolol administration in the memory reconsolidation process.232,233 These results may indicate that propranolol has some utility when used in combination with PE or other exposure-based therapy methods. When considering effects on sleep, propranolol has little therapeutic benefit for sleep disturbances and can worsen symptoms of insomnia in patients with existing sleep issues.234 Adverse effects of propranolol can include vivid dreams and nightmares,235 and at higher dosages propranolol can suppress REM sleep.236 Moreover, β-blockers like propranolol decrease the secretion of melatonin, which can precipitate sleep disruptions and contribute to circadian rhythm abnormalities.237,238 Although propranolol may provide relief for daytime trauma-related anxiety, there is not currently sufficient evidence to suggest its use to treat sleep disturbance acutely following trauma.
Benzodiazepines are γ-aminobutyric acid (GABA)-A receptor agonists widely used to treat symptoms of anxiety and insomnia as they tend to decrease sleep latency and improve sleep continuity. Despite seeming well suited to address hyperarousal and sleep disturbances following trauma, benzodiazepines are not recommended for use in individuals with PTSD due to their abuse potential and tendency to worsen aggression, depression, and psychotherapy outcomes.239 Additionally, benzodiazepines are ineffective at alleviating trauma-related sleep disturbances and general symptoms of PTSD. Furthermore, there is some evidence to suggest that benzodiazepines worsen PTSD symptoms and when used acutely following trauma may increase the risk of PTSD developing.239,240 Benzodiazepines generally increase NREM stage 2 sleep and spindle activity while suppressing slow-wave sleep (stage 3) and REM sleep.241 As slow wave sleep and REM sleep are important for memory consolidation and other cognitive functions,89 it is unclear how these sleep changes may uniquely impact victims of acute trauma, especially in conjunction with other therapies that seek to process recent memories. Due to lack of efficacy, increased risk, and high potential for abuse, benzodiazepines are not recommended for sleep disturbances in acutely traumatized individuals.
The nonbenzodiazepine Z-drugs (zolpidem, zopiclone, and eszopiclone) are positive allosteric modulators of the GABA-A receptor and are used primarily to treat insomnia by decreasing sleep latency and improving sleep quality without significant changes to sleep architecture.242 Although the Z-drugs have fewer adverse side effects and reduced risk of abuse compared to benzodiazepines, few studies have demonstrated that they mitigate sleep issues in PTSD. Prolonged use of zolpidem, especially at high doses, is also associated with increased incidence of PTSD and poorer health outcomes in military service members243—although prolonged use of sleep aids in general suggests there may be chronic underlying sleep issues, making it difficult to parse the reason for this effect. Studies of eszopiclone in treating sleep disturbances in PTSD have had mixed results.242,244 Overall, it is unclear whether Z-drugs have potential for alleviating acute trauma-related sleep disturbances, but the limited, available research does not support use following acute traumatic stress. More research should be undertaken to understand the utility and longer-term outcomes of using Z-drugs to treat sleep disturbances associated with trauma.
Hypothalamic orexins are responsible for modulating fear learning, arousal, and sleep-waking behavior.245 Dual orexin receptor antagonists (DORAs) have been touted as a superior treatment for insomnia as they do not suppress REM sleep like benzodiazepines and Z-drugs.246 The DORA suvorexant is efficacious in treating insomnia and does not produce as severe adverse side effects or risk of abuse as benzodiazepines.246 No evidence exists evaluating the effectiveness of suvorexant to treat sleep disturbances in PTSD or acutely following trauma, although multiple clinical trials are underway (Identifier: NCT02704754, NCT03642028, NCT02849548). However, there is evidence from animal models to suggest that orexin receptor antagonists alleviate traumatic stress–related effects as well as support extinction of fear memory.247,248 Reports of suvorexant inducing nightmares (likely due to its potentiating effect on REM sleep) may limit its utility in treating sleep disturbances following trauma and raises concerns for those with RBD.246,249 Alternatively, as REM sleep is commonly dysregulated in PTSD, suvorexant may be able to support increases in REM in populations that experience suppressed REM resulting from trauma. Due to the current lack of results evaluating suvorexant for use in trauma-related disorders, further studies of suvorexant in acutely traumatized individuals and those with PTSD are needed to assess its therapeutic potential, though the ongoing clinical trials may provide additional insight regarding suvorexant utility.
Nabilone is a synthetic cannabinoid used to treat chemotherapy-induced nausea and chronic pain.250 Nabilone also improves pain-related sleep disturbances in patients with chronic pain.251 In PTSD, nabilone overall symptom severity and reduces the frequency of trauma-related nightmares and increases sleep length.252 These improvements occur rapidly and are sustained out to 9 weeks with no withdrawal symptoms reported.253 Nabilone’s ability to improve sleep in chronic pain patients may be particularly beneficial for addressing sleep disturbances posttrauma as pain can occur from trauma-related injury. Increased pain is also predictive of PTSD diagnosis; therefore, nabilone may have a dual benefit in preventing later diagnosis.254 Moreover, nabilone’s effectiveness at reducing nightmare frequency makes it a potential alternative for patients who cannot take prazosin. Cannabinoids generally promote sleep by activation of cannabinoid type 1 receptors, although their action on the sleep-wake circuit is not well understood.255 As such, future research should evaluate nabilone and other cannabinoids for their ability to regulate sleep disturbance acutely following trauma, especially in cases of injury where pain may disrupt normal sleep.
Ketamine is an NMDA (N-methyl-d-aspartic acid) receptor antagonist is an FDA-approved therapy for treatment-resistant depression and anesthesia256,257 and has also been used to treat sleep disturbances in individuals with PTSD.258,259 Recent research suggests that ketamine’s beneficial effect on mood may be linked to its ability to alter sleep-wake behavior and the amplitude of circadian systems—potentially by modulating expression of circadian rhythm genes.257,260 While rapidly effective, the administration of ketamine following acute trauma exposure has yielded mixed results. Two studies found that ketamine administration acutely following trauma increased symptoms of ASD and likelihood of developing PTSD,261,262 2 studies found no change in outcomes, and 1 study found a decreased incidence of PTSD.263–266 Multiple clinical trials are underway, which will assist in elucidating the impact of ketamine on PTSD symptoms (Identifiers: NCT04032301, NCT04889664, NCT04771767). Given these conflicting findings, additional research is necessary to gauge the therapeutic potential of ketamine before it can be considered a viable treatment for sleep disturbance acutely following trauma.
3,4-Methylenedioxymethamphetamine (MDMA)-assisted psychotherapy is an experimental treatment that targets memory consolidation and fear response in chronic PTSD patients.267 As part of this treatment strategy, therapists ask patients to recall specific traumatic memories after taking MDMA, a triple monoamine reuptake inhibitor that promotes release of multiple neurotransmitters, including dopamine, serotonin, and norepinephrine as well as oxytocin, cortisol, prolactin, and vasopressin.268 It is believed that MDMA enhances the positive effects of therapy by increasing the ability of the patient to tolerate negative emotions associated with recalling traumatic events, which decreases patients dropping out and improves treatment success.267 Preliminary studies support its use in PTSD patients, including clinically significant reductions in Clinician-Administered PTSD Scale scores and improved self-reported sleep.268,269 However, in some patients, it may increase bruxism, anxiety, and jitteriness, which could potentially contribute to further sleep disturbance.269,270 Additionally, MDMA is a known drug of abuse and a few incidences of increased suicidal ideation have been observed as part of these clinical trials267; therefore, caution should be taken in administering it outside of a strongly controlled psychotherapeutic environment. Multiple clinical trials are ongoing to investigate the use of MDMA in PTSD treatment, with a more limited number assessing its efficacy in treating PTSD-related sleep disturbances (Identifier: NCT03752918). Additional evidence is needed to understand if this treatment would be effective acutely following trauma for both sleep disturbance and other trauma-related symptoms, though MDMA would likely not be a first-line treatment for acute disturbance.
Overall, there is a paucity of research on early treatment for both sleep disturbance and trauma-related symptoms following an acute traumatic stress event. Based on mechanistic understanding of pharmacologics and results from clinical trials for PTSD or trauma-related sleep disturbance, caution should be taken with SSRIs and SNRIs in the setting of acute trauma as they can cause sleep disturbance early before reaching full therapeutic effect. On the other hand, there is some evidence that SSRIs and SNRIs drugs could be used in a targeted fashion by clinicians to decrease REM sleep, which could be undertaken in select individuals if specific sleep characteristics were identified early by clinicians. The evidence for use of adrenergic pharmacotherapies is mixed; therefore, additional inquiry is needed to understand their potential utility for sleep disturbance acutely following trauma. Generally, the existing, albeit limited, evidence for GABAergic modulators suggests minimal utility for trauma-related sleep disturbance. However, there is some early evidence supporting the use of SARIs, atypical antipsychotics, dual orexin receptor antagonists, and cannabinoids to quickly restore trauma-disturbed sleep. Clinical trials for early implementation of pharmacologic treatment of trauma-related sleep disturbance are warranted and based on what is known about the trajectory of sleep and trauma following a traumatic stress event. Such pharmacologics have potential to prevent long-term trauma-related symptoms.
DISCUSSION AND FUTURE DIRECTIONS
Currently, little is known about how sleep is altered acutely following real-life trauma. Only 2 actigraphy studies have quantified objective changes to sleep parameters within 48 hours of trauma,27,28 and no studies have characterized objective sleep parameters in ASD. This constitutes a critical knowledge gap. Sleep studies in ASD sufferers may be especially informative, since they could potentially reveal whether, and the extent to which, sleep disturbance predicts the likelihood that initial trauma-related symptoms will persist and become chronic. This is a reasonable hypothesis because it is known that sleep facilitates emotional and declarative memory consolidation, and REM sleep is thought to aid in the proper contextualization of traumatic experiences.24 Thus, the characterization of early posttrauma sleep disturbances may be critical for understanding the initial formation of trauma-related symptoms as well as the pathogenesis of chronic trauma-related disorders.
Prior studies provide compelling evidence that peri-trauma sleep disturbance increases the risk of developing PTSD. However, the pathophysiological processes by which sleep disturbance contributes to the development of PTSD are unclear. Do posttrauma sleep disturbances independently contribute to PTSD development, or do they reflect trauma-induced changes in neural circuitry that increase susceptibility to PTSD? How and why does sleep disturbance prior to trauma increase vulnerability to PTSD in otherwise neuro-typical and psychologically healthy individuals? Would improving the sleep of poor sleepers increase their resilience to trauma? Further studies examining the effects of sleep and sleep loss in preclinical models of trauma exposure will likely aid in elucidating the mechanisms by which sleep mediates risk of developing trauma-related disorders.
Several nonpharmacologic and pharmacologic interventions have been evaluated as treatments for sleep disturbance in PTSD, but few studies have tested the same drugs acutely posttrauma or in ASD. Of those few studies that have evaluated the utility of various interventions for improving sleep in ASD, small sample sizes and/or lack of proper controls limit interpretations relevant to clinical practice. Clearly, well powered and well controlled studies are necessary to accurately gauge each intervention’s effectiveness for improving sleep and reducing traumatic stress symptoms. Furthermore, it is suggested that new therapies be developed that are specifically designed to treat trauma-related disorders—a process that will be made easier via an improved understanding of the pathophysiology of trauma-induced symptoms and disorders. Mechanistic preclinical studies investigating trauma-induced neurological changes and their time course could identify novel targets for pharmacological interventions. In fact, it is possible that multiple treatments will need to be developed since the trauma-related pathophysiology may evolve and cascade over time—eg, as exposure to trauma persists and different adaptive mechanisms are serially recruited and exhausted.
Although more work is needed to definitively establish whether, and the extent to which, a reciprocal cause-effect relationship between sleep disturbance and stress-related pathologies exists, the current evidence suggesting the likelihood of such a relationship is substantial and, in our opinion, compelling. At the very least, better sleep—both before and after traumatic events—is associated with better outcomes. Accordingly, it is suggested that the time has come to aggressively study the potential utility of improving sleep characteristics that are disordered during the acute posttrauma phase, with an ultimate aim of developing evidence-based sleep-enhancing treatment guidelines for ASR/ASD and potential prevention of PTSD.
DISCLOSURE STATEMENT
All authors have seen and approved the final manuscript. This work was supported by the US Army Military Operational Medicine Research Program. Material has been reviewed by the Walter Reed Army Institute of Research. There is no objection to its presentation and/publication. The opinions or assertions contained herein are the private views of the authors and are not to be construed as official or reflecting the view of the Department of the Army or the Department of Defense. All authors declare no conflict of interests defined as any financial interests or connections, direct or indirect, or other situations that might raise the question of bias in the work reported or the conclusions, implications, or opinions within the manuscript.
ACKNOWLEDGMENTS
The authors thank LTC Vincent F. Capaldi, M.D. for his insights and clinical input on sleep disturbance and traumatic stress exposure.
ABBREVIATIONS
- ASD
acute stress disorder
- ASR
acute stress reaction
- CBTi
cognitive behavioral therapy for insomnia
- CPT
cognitive processing therapy
- EMDR
eye movement desensitization and reprocessing
- FDA
US Food and Drug Administration
- GABA
γ-aminobutyric acid
- IRT
imagery rehearsal therapy
- MDMA
3,4 methylenedioxymethamphetamine
- PE
prolonged exposure
- PTSD
post–traumatic stress disorder
- RBD
REM sleep behavior disorder
- REM
rapid eye movement
- RSWA
REM sleep without atonia
- SARI
serotonin antagonist and reuptake inhibitor
- SNRI
serotonin and norepinephrine reuptake inhibitor
- SSRI
selective serotonin reuptake inhibitor
- TMS
transcranial magnetic stimulation
- TSD
trauma-associated sleep disorder
REFERENCES
- 1. Yehuda R , McFarlane AC , Shalev AY . Predicting the development of posttraumatic stress disorder from the acute response to a traumatic event . Biol Psychiatry. 1998. ; 44 ( 12 ): 1305 – 1313 . [DOI] [PubMed] [Google Scholar]
- 2. McFarlane AC , Papay P . Multiple diagnoses in posttraumatic stress disorder in the victims of a natural disaster . J Nerv Ment Dis. 1992. ; 180 ( 8 ): 498 – 504 . [DOI] [PubMed] [Google Scholar]
- 3. Isserlin L , Zerach G , Solomon Z . Acute stress responses: a review and synthesis of ASD, ASR, and CSR . Am J Orthopsychiatry. 2008. ; 78 ( 4 ): 423 – 429 . [DOI] [PubMed] [Google Scholar]
- 4. Spoormaker VI , Montgomery P . Disturbed sleep in post-traumatic stress disorder: secondary symptom or core feature? Sleep Med Rev. 2008. ; 12 ( 3 ): 169 – 184 . [DOI] [PubMed] [Google Scholar]
- 5. American Psychiatric Association . Diagnostic and Statistical Manual of Mental Disorders . 5th ed . Washington, DC: : APA Publishing; ; 2013. . [Google Scholar]
- 6. Bryant RA . Early predictors of posttraumatic stress disorder . Biol Psychiatry. 2003. ; 53 ( 9 ): 789 – 795 . [DOI] [PubMed] [Google Scholar]
- 7. Weisaeth L . Acute posttraumatic stress: nonacceptance of early intervention . J Clin Psychiatry. 2001. ; 62 ( Suppl 17 ): 35 – 40 . [PubMed] [Google Scholar]
- 8. Bryant RA , Friedman MJ , Spiegel D , Ursano R , Strain J . A review of acute stress disorder in DSM-5 . Depress Anxiety. 2011. ; 28 ( 9 ): 802 – 817 . [DOI] [PubMed] [Google Scholar]
- 9. World Health Organization . ICD-10: International Statistical Classification of Diseases and Related Health Problems, 10th Revision. Geneva, Switzerland: : World Health Organization; ; 2004. . [Google Scholar]
- 10. van Gelderen MJ , Nijdam MJ , Vermetten E . An innovative framework for delivering psychotherapy to patients with treatment-resistant posttraumatic stress disorder: rationale for interactive motion-assisted therapy . Front Psychiatry. 2018. ; 9 : 176 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Soldatos CR , Paparrigopoulos TJ , Pappa DA , Christodoulou GN . Early post-traumatic stress disorder in relation to acute stress reaction: an ICD-10 study among help seekers following an earthquake . Psychiatry Res. 2006. ; 143 ( 2–3 ): 245 – 253 . [DOI] [PubMed] [Google Scholar]
- 12. Shalev AY , Sahar T , Freedman S , et al . A prospective study of heart rate response following trauma and the subsequent development of posttraumatic stress disorder . Arch Gen Psychiatry. 1998. ; 55 ( 6 ): 553 – 559 . [DOI] [PubMed] [Google Scholar]
- 13. Koren D , Arnon I , Klein E . Acute stress response and posttraumatic stress disorder in traffic accident victims: a one-year prospective, follow-up study . Am J Psychiatry. 1999. ; 156 ( 3 ): 367 – 373 . [DOI] [PubMed] [Google Scholar]
- 14. McFarlane AC , Atchison M , Yehuda R . The acute stress response following motor vehicle accidents and its relation to PTSD . Ann N Y Acad Sci. 1997. ; 821 ( 1 Psychobiology ): 437 – 441 . [DOI] [PubMed] [Google Scholar]
- 15. Shalev AY , Freedman S , Peri T , et al . Prospective study of posttraumatic stress disorder and depression following trauma . Am J Psychiatry. 1998. ; 155 ( 5 ): 630 – 637 . [DOI] [PubMed] [Google Scholar]
- 16. Bryant RA . Acute stress disorder as a predictor of posttraumatic stress disorder: a systematic review . J Clin Psychiatry. 2011. ; 72 ( 2 ): 233 – 239 . [DOI] [PubMed] [Google Scholar]
- 17. Neylan TC , Kessler RC , Ressler KJ , et al . Prior sleep problems and adverse post-traumatic neuropsychiatric sequelae of motor vehicle collision in the AURORA study . Sleep. 2021. ; 44 ( 3 ): zsaa2001 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Shokri-Kojori E , Wang GJ , Wiers CE , et al . β-Amyloid accumulation in the human brain after one night of sleep deprivation . Proc Natl Acad Sci USA. 2018. ; 115 ( 17 ): 4483 – 4488 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Mullington JM , Simpson NS , Meier-Ewert HK , Haack M . Sleep loss and inflammation . Best Pract Res Clin Endocrinol Metab. 2010. ; 24 ( 5 ): 775 – 784 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Manchanda S , Singh H , Kaur T , Kaur G . Low-grade neuroinflammation due to chronic sleep deprivation results in anxiety and learning and memory impairments . Mol Cell Biochem. 2018. ; 449 ( 1-2 ): 63 – 72 . [DOI] [PubMed] [Google Scholar]
- 21. Banks S , Dinges DF . Behavioral and physiological consequences of sleep restriction . J Clin Sleep Med. 2007. ; 3 ( 5 ): 519 – 528 . [PMC free article] [PubMed] [Google Scholar]
- 22. Meerlo P , Sgoifo A , Suchecki D . Restricted and disrupted sleep: effects on autonomic function, neuroendocrine stress systems and stress responsivity . Sleep Med Rev. 2008. ; 12 ( 3 ): 197 – 210 . [DOI] [PubMed] [Google Scholar]
- 23. Freeman D , Sheaves B , Goodwin GM , et al . The effects of improving sleep on mental health (OASIS): a randomised controlled trial with mediation analysis . Lancet Psychiatry. 2017. ; 4 ( 10 ): 749 – 758 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Goldstein AN , Walker MP . The role of sleep in emotional brain function . Annu Rev Clin Psychol. 2014. ; 10 : 679 – 708 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Minkel JD , Banks S , Htaik O , et al . Sleep deprivation and stressors: evidence for elevated negative affect in response to mild stressors when sleep deprived . Emotion. 2012. ; 12 ( 5 ): 1015 – 1020 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Wood JM , Bootzin RR , Rosenhan D , Nolen-Hoeksema S , Jourden F . Effects of the 1989 San Francisco earthquake on frequency and content of nightmares . J Abnorm Psychol. 1992. ; 101 ( 2 ): 219 – 224 . [DOI] [PubMed] [Google Scholar]
- 27. Mizuno K , Okamoto-Mizuno K . Actigraphically evaluated sleep on the days surrounding the Great East Japan Earthquake . Nat Hazards. 2014. ; 72 ( 2 ): 969 – 981 . [Google Scholar]
- 28. Lavie P , Carmeli A , Mevorach L , Liberman N . Sleeping under the threat of the Scud: war-related environmental insomnia . Isr J Med Sci. 1991. ; 27 ( 11-12 ): 681 – 686 . [PubMed] [Google Scholar]
- 29. Sinha SS . Trauma-induced insomnia: a novel model for trauma and sleep research . Sleep Med Rev. 2016. ; 25 : 74 – 83 . [DOI] [PubMed] [Google Scholar]
- 30. Thordardottir EB , Hansdottir I , Valdimarsdottir UA , Shipherd JC , Resnick H , Gudmundsdottir B . The manifestations of sleep disturbances 16 years post-trauma . Sleep. 2016. ; 39 ( 8 ): 1551 – 1554 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Askenasy JJM , Lewin I . The impact of missile warfare on self-reported sleep quality. Part 1 . Sleep. 1996. ; 19 ( 1 ): 47 – 51 . [DOI] [PubMed] [Google Scholar]
- 32. Varela E , Koustouki V , Davos CH , Eleni K . Psychological consequences among adults following the 1999 earthquake in Athens, Greece . Disasters. 2008. ; 32 ( 2 ): 280 – 291 . [DOI] [PubMed] [Google Scholar]
- 33. Galea S , Resnick H , Ahern J , et al . Posttraumatic stress disorder in Manhattan, New York City, after the September 11th terrorist attacks . J Urban Health. 2002. ; 79 ( 3 ): 340 – 353 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. McMillen JC , North CS , Smith EM . What parts of PTSD are normal: intrusion, avoidance, or arousal? Data from the Northridge, California, earthquake . J Trauma Stress. 2000. ; 13 ( 1 ): 57 – 75 . [DOI] [PubMed] [Google Scholar]
- 35. McWhorter KL , Parks CG , D’Aloisio AA , Rojo-Wissar DM , Sandler DP , Jackson CL . Traumatic childhood experiences and multiple dimensions of poor sleep among adult women . Sleep. 2019. ; 42 ( 8 ): zsz108 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Glod CA , Teicher MH , Hartman CR , Harakal T . Increased nocturnal activity and impaired sleep maintenance in abused children . J Am Acad Child Adolesc Psychiatry. 1997. ; 36 ( 9 ): 1236 – 1243 . [DOI] [PubMed] [Google Scholar]
- 37. Rosen J , Reynolds CF III , Yeager AL , Houck PR , Hurwitz LF . Sleep disturbances in survivors of the Nazi Holocaust . Am J Psychiatry. 1991. ; 148 ( 1 ): 62 – 66 . [DOI] [PubMed] [Google Scholar]
- 38. Buysse DJ . Insomnia . JAMA. 2013. ; 309 ( 7 ): 706 – 716 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Mysliwiec V , O’Reilly B , Polchinski J , Kwon HP , Germain A , Roth BJ . Trauma associated sleep disorder: a proposed parasomnia encompassing disruptive nocturnal behaviors, nightmares, and REM without atonia in trauma survivors . J Clin Sleep Med. 2014. ; 10 ( 10 ): 1143 – 1148 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Mysliwiec V , Brock MS , Creamer JL , O’Reilly BM , Germain A , Roth BJ . Trauma associated sleep disorder: a parasomnia induced by trauma . Sleep Med Rev. 2018. ; 37 : 94 – 104 . [DOI] [PubMed] [Google Scholar]
- 41. Feemster JC , Smith KL , McCarter SJ , St Louis EK . Trauma-associated sleep disorder: a posttraumatic stress/REM sleep behavior disorder mash-up? J Clin Sleep Med. 2019. ; 15 ( 2 ): 345 – 349 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Harvey AG , Bryant RA . Acute stress disorder after mild traumatic brain injury . J Nerv Ment Dis. 1998. ; 186 ( 6 ): 333 – 337 . [DOI] [PubMed] [Google Scholar]
- 43. Smith MT , Klick B , Kozachik S , et al . Sleep onset insomnia symptoms during hospitalization for major burn injury predict chronic pain . Pain. 2008. ; 138 ( 3 ): 497 – 506 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Stanovic JK , James KA , Vandevere CA . The effectiveness of risperidone on acute stress symptoms in adult burn patients: a preliminary retrospective pilot study . J Burn Care Rehabil. 2001. ; 22 ( 3 ): 210 – 213 . [DOI] [PubMed] [Google Scholar]
- 45. Meighen KG , Hines LA , Lagges AM . Risperidone treatment of preschool children with thermal burns and acute stress disorder . J Child Adolesc Psychopharmacol. 2007. ; 17 ( 2 ): 223 – 232 . [DOI] [PubMed] [Google Scholar]
- 46. Hurwitz TD , Mahowald MW , Kuskowski M , Engdahl BE . Polysomnographic sleep is not clinically impaired in Vietnam combat veterans with chronic posttraumatic stress disorder . Biol Psychiatry. 1998. ; 44 ( 10 ): 1066 – 1073 . [DOI] [PubMed] [Google Scholar]
- 47. Cohen DJ , Begley A , Alman JJ , et al . Quantitative electroencephalography during rapid eye movement (REM) and non-REM sleep in combat-exposed veterans with and without post-traumatic stress disorder . J Sleep Res. 2013. ; 22 ( 1 ): 76 – 82 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Fisher AJ , Woodward SH . Cardiac stability at differing levels of temporal analysis in panic disorder, post-traumatic stress disorder, and healthy controls . Psychophysiology. 2014. ; 51 ( 1 ): 80 – 87 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Grossman ES , Hoffman Y , Bodner E , et al . Psychological effects following the Iran nuclear deal: Iranian nuclear threat salience moderates the relationship between PTSD symptoms and sleep problems . Psychiatry Res. 2016. ; 243 : 292 – 294 . [DOI] [PubMed] [Google Scholar]
- 50. Lind MJ , Brown E , Farrell-Carnahan L , et al . Sleep disturbances in OEF/OIF/OND veterans: associations with PTSD, personality, and coping . J Clin Sleep Med. 2017. ; 13 ( 2 ): 291 – 299 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. van der Kolk B , Blitz R , Burr W , Sherry S , Hartmann E . Nightmares and trauma: a comparison of nightmares after combat with lifelong nightmares in veterans . Am J Psychiatry. 1984. ; 141 ( 2 ): 187 – 190 . [DOI] [PubMed] [Google Scholar]
- 52. Inman DJ , Silver SM , Doghramji K . Sleep disturbances in post-traumatic stress disorder: a comparison with non-PTSD insomnia . J Trauma Stress. 1990. ; 3 ( 3 ): 429 – 437 . [Google Scholar]
- 53. Lavie P , Hefez A , Halperin G , Enoch D . Long-term effects of traumatic war-related events on sleep . Am J Psychiatry. 1979. ; 136 ( 2 ): 175 – 178 . [DOI] [PubMed] [Google Scholar]
- 54. Hefez A , Metz L , Lavie P . Long-term effects of extreme situational stress on sleep and dreaming . Am J Psychiatry. 1987. ; 144 ( 3 ): 344 – 347 . [DOI] [PubMed] [Google Scholar]
- 55. Engdahl BE , Eberly RE , Hurwitz TD , Mahowald MW , Blake J . Sleep in a community sample of elderly war veterans with and without posttraumatic stress disorder . Biol Psychiatry. 2000. ; 47 ( 6 ): 520 – 525 . [DOI] [PubMed] [Google Scholar]
- 56. Mellman TA , Nolan B , Hebding J , Kulick-Bell R , Dominguez R . A polysomnographic comparison of veterans with combat-related PTSD, depressed men, and non-ill controls . Sleep. 1997. ; 20 ( 1 ): 46 – 51 . [DOI] [PubMed] [Google Scholar]
- 57. Ross RJ , Ball WA , Dinges DF , et al . Rapid eye movement sleep disturbance in posttraumatic stress disorder . Biol Psychiatry. 1994. ; 35 ( 3 ): 195 – 202 . [DOI] [PubMed] [Google Scholar]
- 58. Ross RJ , Ball WA , Sanford LD , et al . Rapid eye movement sleep changes during the adaptation night in combat veterans with posttraumatic stress disorder . Biol Psychiatry. 1999. ; 45 ( 7 ): 938 – 941 . [DOI] [PubMed] [Google Scholar]
- 59. Lipinska M , Timol R , Kaminer D , Thomas KGF . Disrupted rapid eye movement sleep predicts poor declarative memory performance in post-traumatic stress disorder . J Sleep Res. 2014. ; 23 ( 3 ): 311 – 317 . [DOI] [PubMed] [Google Scholar]
- 60. Kobayashi I , Lavela J , Bell K , Mellman TA . The impact of posttraumatic stress disorder versus resilience on nocturnal autonomic nervous system activity as functions of sleep stage and time of sleep . Physiol Behav. 2016. ; 164 ( Pt A ): 11 – 18 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Kobayashi I , Mellman TA . Gender differences in sleep during the aftermath of trauma and the development of posttraumatic stress disorder . Behav Sleep Med. 2012. ; 10 ( 3 ): 180 – 190 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Richards A , Metzler TJ , Ruoff LM , et al . Sex differences in objective measures of sleep in post-traumatic stress disorder and healthy control subjects . J Sleep Res. 2013. ; 22 : 679 – 687 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Mellman TA , Kulick-Bell R , Ashlock LE , Nolan B . Sleep events among veterans with combat-related posttraumatic stress disorder . Am J Psychiatry. 1995. ; 152 ( 1 ): 110 – 115 . [DOI] [PubMed] [Google Scholar]
- 64. Woodward SH , Arsenault NJ , Murray C , Bliwise DL . Laboratory sleep correlates of nightmare complaint in PTSD inpatients . Biol Psychiatry. 2000. ; 48 ( 11 ): 1081 – 1087 . [DOI] [PubMed] [Google Scholar]
- 65. Neylan TC , Marmar CR , Metzler TJ , et al . Sleep disturbances in the Vietnam generation: findings from a nationally representative sample of male Vietnam veterans . Am J Psychiatry. 1998. ; 155 ( 7 ): 929 – 933 . [DOI] [PubMed] [Google Scholar]
- 66. Wallace DM , Shafazand S , Ramos AR , et al . Insomnia characteristics and clinical correlates in Operation Enduring Freedom/Operation Iraqi Freedom veterans with post-traumatic stress disorder and mild traumatic brain injury: an exploratory study . Sleep Med. 2011. ; 12 ( 9 ): 850 – 859 . [DOI] [PubMed] [Google Scholar]
- 67. Breslau N , Roth T , Burduvali E , Kapke A , Schultz L , Roehrs T . Sleep in posttraumatic stress disorder: a community-based polysomnographic study . Ann Clin Psychiatry. 2004. ; 61 : 508 – 516 . [DOI] [PubMed] [Google Scholar]
- 68. Glaubman H , Mikulincer M , Porat A , Wasserman O , Birger M . Sleep of chronic post-traumatic patients . J Trauma Stress. 1990. ; 3 ( 2 ): 255 – 263 . [Google Scholar]
- 69. Rao MN , Chau A , Madden E , et al . Hyperinsulinemic response to oral glucose challenge in individuals with posttraumatic stress disorder . Psychoneuroendocrinology. 2014. ; 49 : 171 – 181 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Klein E , Koren D , Arnon I , Lavie P . Sleep complaints are not corroborated by objective sleep measures in post-traumatic stress disorder: a 1-year prospective study in survivors of motor vehicle crashes . J Sleep Res. 2003. ; 12 ( 1 ): 35 – 41 . [DOI] [PubMed] [Google Scholar]
- 71. Capaldi VF II , Guerrero ML , Killgore WDS . Sleep disruptions among returning combat veterans from Iraq and Afghanistan . Mil Med. 2011. ; 176 ( 8 ): 879 – 888 . [DOI] [PubMed] [Google Scholar]
- 72. van Liempt S , Vermetten E , Lentjes E , Arends J , Westenberg H . Decreased nocturnal growth hormone secretion and sleep fragmentation in combat-related posttraumatic stress disorder; potential predictors of impaired memory consolidation . Psychoneuroendocrinology. 2011. ; 36 ( 9 ): 1361 – 1369 . [DOI] [PubMed] [Google Scholar]
- 73. Mellman TA , David D , Kulick-Bell R , Hebding J , Nolan B . Sleep disturbance and its relationship to psychiatric morbidity after Hurricane Andrew . Am J Psychiatry. 1995. ; 152 ( 11 ): 1659 – 1663 . [DOI] [PubMed] [Google Scholar]
- 74. Straus LD , Drummond SPA , Nappi CM , Jenkins MM , Norman SB . Sleep variability in military-related PTSD: a comparison to primary insomnia and healthy controls . J Trauma Stress. 2015. ; 28 ( 1 ): 8 – 16 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Dow BM , Kelsoe JR Jr ., Gillin JC . Sleep and dreams in Vietnam PTSD and depression . Biol Psychiatry. 1996. ; 39 ( 1 ): 42 – 50 . [DOI] [PubMed] [Google Scholar]
- 76. Gupta MA , Simpson FC . Obstructive sleep apnea and psychiatric disorders: a systematic review . J Clin Sleep Med. 2015. ; 11 ( 2 ): 165 – 175 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Kessler RC , Sonnega A , Bromet E , Hughes M , Nelson CB . Posttraumatic stress disorder in the National Comorbidity Survey . Arch Gen Psychiatry. 1995. ; 52 ( 12 ): 1048 – 1060 . [DOI] [PubMed] [Google Scholar]
- 78. Koffel E , Polusny MA , Arbisi PA , Erbes CR . Pre-deployment daytime and nighttime sleep complaints as predictors of post-deployment PTSD and depression in National Guard troops . J Anxiety Disord. 2013. ; 27 ( 5 ): 512 – 519 . [DOI] [PubMed] [Google Scholar]
- 79. Kramer M , Kinney L . Sleep patterns in trauma victims with disturbed dreaming . Psychiatr J Univ Ott. 1988. ; 13 ( 1 ): 12 – 16 . [PubMed] [Google Scholar]
- 80. Bryant RA , Creamer M , O’Donnell M , Silove D , McFarlane AC . Sleep disturbance immediately prior to trauma predicts subsequent psychiatric disorder . Sleep. 2010. ; 33 ( 1 ): 69 – 74 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Gehrman P , Seelig AD , Jacobson IG , et al . Predeployment Sleep Duration and Insomnia Symptoms as Risk Factors for New-Onset Mental Health Disorders Following Military Deployment . Sleep. 2013. ; 36 ( 7 ): 1009 – 1018 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Wang HE , Campbell-Sills L , Kessler RC , et al . Pre-deployment insomnia is associated with post-deployment post-traumatic stress disorder and suicidal ideation in US Army soldiers . Sleep. 2019. ; 42 ( 2 ): 1 – 9 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Wright KM , Britt TW , Bliese PD , Adler AB . Insomnia severity, combat exposure and mental health outcomes . Stress Health. 2011. ; 27 ( 4 ): 325 – 333 . [Google Scholar]
- 84. Wright KM , Britt TW , Bliese PD , Adler AB , Picchioni D , Moore D . Insomnia as predictor versus outcome of PTSD and depression among Iraq combat veterans . J Clin Psychol. 2011. ; 67 ( 12 ): 1240 – 1258 . [DOI] [PubMed] [Google Scholar]
- 85. Osgood JM , Finan PH , Hinman SJ , So CJ , Quartana PJ . Combat exposure, post-traumatic stress symptoms, and health-related behaviors: the role of sleep continuity and duration . Sleep. 2019. ; 42 ( 3 ): 1 – 11 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Koren D , Arnon I , Lavie P , Klein E . Sleep complaints as early predictors of posttraumatic stress disorder: a 1-year prospective study of injured survivors of motor vehicle accidents . Am J Psychiatry. 2002. ; 159 ( 5 ): 855 – 857 . [DOI] [PubMed] [Google Scholar]
- 87. Gerhart JI , Hall BJ , Russ EU , Canetti D , Hobfoll SE . Sleep disturbances predict later trauma-related distress: cross-panel investigation amidst violent turmoil . Health Psychol. 2014. ; 33 ( 4 ): 365 – 372 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Porcheret K , Iyadurai L , Bonsall MB , et al . Sleep and intrusive memories immediately after a traumatic event in emergency department patients . Sleep. 2020. ; 43 ( 8 ): zsaa033 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Rasch B , Born J . About sleep’s role in memory . Physiol Rev. 2013. ; 93 ( 2 ): 681 – 766 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Stickgold R , Walker MP . Sleep-dependent memory triage: evolving generalization through selective processing . Nat Neurosci. 2013. ; 16 ( 2 ): 139 – 145 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Graves LA , Heller EA , Pack AI , Abel T . Sleep deprivation selectively impairs memory consolidation for contextual fear conditioning . Learn Mem. 2003. ; 10 ( 3 ): 168 – 176 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Nishida M , Pearsall J , Buckner RL , Walker MP . REM sleep, prefrontal theta, and the consolidation of human emotional memory . Cereb Cortex. 2009. ; 19 ( 5 ): 1158 – 1166 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Porcheret K , Holmes EA , Goodwin GM , Foster RG , Wulff K . Psychological effect of an analogue traumatic event reduced by sleep deprivation . Sleep. 2015. ; 38 ( 7 ): 1017 – 1025 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Kuriyama K , Soshi T , Kim Y . Sleep deprivation facilitates extinction of implicit fear generalization and physiological response to fear . Biol Psychiatry. 2010. ; 68 ( 11 ): 991 – 998 . [DOI] [PubMed] [Google Scholar]
- 95. Southwick SM , Bremner JD , Rasmusson A , Morgan CA III , Arnsten A , Charney DS . Role of norepinephrine in the pathophysiology and treatment of posttraumatic stress disorder . Biol Psychiatry. 1999. ; 46 ( 9 ): 1192 – 1204 . [DOI] [PubMed] [Google Scholar]
- 96. Porcheret K , van Heugten-van der Kloet D , Goodwin GM , Foster RG , Wulff K , Holmes EA . Investigation of the impact of total sleep deprivation at home on the number of intrusive memories to an analogue trauma . Transl Psychiatry. 2019. ; 9 : 104 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Kleim B , Wysokowsky J , Schmid N , Seifritz E , Rasch B . Effects of sleep after experimental trauma on intrusive emotional memories . Sleep. 2016. ; 39 ( 12 ): 2125 – 2132 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Sterpenich V , Albouy G , Boly M , et al . Sleep-related hippocampo-cortical interplay during emotional memory recollection . PLoS Biol. 2007. ; 5 ( 11 ): e282 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Milad MR , Pitman RK , Ellis CB , et al . Neurobiological basis of failure to recall extinction memory in posttraumatic stress disorder . Biol Psychiatry. 2009. ; 66 : 1075 – 1082 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Davis CJ , Vanderheyden WM . Optogenetic sleep enhancement improves fear-associated memory processing following trauma exposure in rats . Sci Rep. 2020. ; 10 ( 1 ): 18025 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Cox RC , Tuck BM , Olatunji BO . Sleep disturbance in posttraumatic stress disorder: epiphenomenon or causal factor? Curr Psychiatry Rep. 2017. ; 19 ( 4 ): 22 . [DOI] [PubMed] [Google Scholar]
- 102. Lommen MJJ , Grey N , Clark DM , Wild J , Stott R , Ehlers A . Sleep and treatment outcome in posttraumatic stress disorder: results from an effectiveness study . Depress Anxiety. 2016. ; 33 ( 7 ): 575 – 583 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Rusch HL , Guardado P , Baxter T , Mysliwiec V , Gill JM . Improved sleep quality is associated with reductions in depression and PTSD arousal symptoms and increases in IGF-1 concentrations . J Clin Sleep Med. 2015. ; 11 ( 6 ): 615 – 623 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Morgenthaler TI , Auerbach S , Casey KR , et al . Position paper for the treatment of nightmare disorder in adults: an American academy of sleep medicine position paper . J Clin Sleep Med. 2018. ; 14 ( 6 ): 1041 – 1055 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Ursano RJ , Bell C , Eth S , et al . Practice guideline for the treatment of patients with acute stress disorder and posttraumatic stress disorder . Am J Psychiatry. 2004. ; 161 ( 11 Suppl ): 3 – 31. [PubMed] [Google Scholar]
- 106. Aurora RN , Zak RS , Auerbach SH , et al . American Academy of Sleep Medicine . Best practice guide for the treatment of nightmare disorder in adults . J Clin Sleep Med. 2010. ; 6 ( 4 ): 389 – 401 . [PMC free article] [PubMed] [Google Scholar]
- 107. Nash WP , Watson PJ . Review of VA/DOD Clinical Practice Guideline on management of acute stress and interventions to prevent posttraumatic stress disorder . J Rehabil Res Dev. 2012. ; 49 ( 5 ): 637 – 648 . [DOI] [PubMed] [Google Scholar]
- 108. World Health Organization . Guidelines for the Management of Conditions Specifically Related to Stress; 2013. . https://www.who.int/publications/i/item/guidelines-for-the-management-of-conditions-that-are-specifically-related-to-stress; Accessed June 15, 2022. [PubMed]
- 109. Forbes D , Wolfgang B , Cooper J , Creamer M , Barton D . Post-traumatic stress disorder–best practice GP guidelines . Aust Fam Physician. 2009. ; 38 ( 3 ): 106 – 111 . [PubMed] [Google Scholar]
- 110. Chung K-F , Lee C-T , Yeung W-F , Chan M-S , Chung EW , Lin W-L . Sleep hygiene education as a treatment of insomnia: a systematic review and meta-analysis . Fam Pract. 2018. ; 35 ( 4 ): 365 – 375 . [DOI] [PubMed] [Google Scholar]
- 111. Jacobs GD , Pace-Schott EF , Stickgold R , Otto MW . Cognitive behavior therapy and pharmacotherapy for insomnia: a randomized controlled trial and direct comparison . Arch Intern Med. 2004. ; 164 ( 17 ): 1888 – 1896 . [DOI] [PubMed] [Google Scholar]
- 112. Ho FYY , Chan CS , Tang KNS . Cognitive-behavioral therapy for sleep disturbances in treating posttraumatic stress disorder symptoms: a meta-analysis of randomized controlled trials . Clin Psychol Rev. 2016. ; 43 : 90 – 102 . [DOI] [PubMed] [Google Scholar]
- 113. Mitchell MD , Gehrman P , Perlis M , Umscheid CA . Comparative effectiveness of cognitive behavioral therapy for insomnia: a systematic review . BMC Fam Pract. 2012. ; 13 ( 1 ): 40 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Lee DJ , Schnitzlein CW , Wolf JP , Vythilingam M , Rasmusson AM , Hoge CW . Psychotherapy vs pharmacotherapy for posttraumatic stress disorder: systemic review and meta-analyses to determine first-line treatments . Depress Anxiety. 2016. ; 33 ( 9 ): 792 – 806 . [DOI] [PubMed] [Google Scholar]
- 115. Pigeon WR , Crean HF , Cerulli C , Gallegos AM , Bishop TM , Heffner KL . A randomized clinical trial of cognitive-behavioral therapy for insomnia to augment posttraumatic stress disorder treatment in survivors of interpersonal violence . Psychother Psychosom. 2022. ; 91 ( 1 ): 50 – 62 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Morin CM . Cognitive-behavioral approaches to the treatment of insomnia . J Clin Psychiatry. 2004. ; 65 ( Suppl 16 ): 33 – 40 . [PubMed] [Google Scholar]
- 117. Spielman AJ , Saskin P , Thorpy MJ . Treatment of chronic insomnia by restriction of time in bed . Sleep. 1987. ; 10 ( 1 ): 45 – 56 . [PubMed] [Google Scholar]
- 118. Trauer JM , Qian MY , Doyle JS , Rajaratnam SMW , Cunnington D . Cognitive behavioral therapy for chronic insomnia: a systematic review and meta-analysis . Ann Intern Med. 2015. ; 163 ( 3 ): 191 – 204 . [DOI] [PubMed] [Google Scholar]
- 119. Kwon M , Wang J , Wilding G , Dickerson SS , Dean GE . Brief Behavioral Treatment for insomnia: a meta-analysis . Behav Sleep Med. 2021. : 1 – 21 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Short NA , Zvolensky MJ , Schmidt NB . A pilot randomized clinical trial of Brief Behavioral Treatment for Insomnia to reduce problematic cannabis use among trauma-exposed young adults . J Subst Abuse Treat. 2021. ; 131 : 108537 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Yücel DE , van Emmerik AAP , Souama C , Lancee J . Comparative efficacy of imagery rehearsal therapy and prazosin in the treatment of trauma-related nightmares in adults: a meta-analysis of randomized controlled trials . Sleep Med Rev. 2020. ; 50 : 101248 . [DOI] [PubMed] [Google Scholar]
- 122. Krakow B , Hollifield M , Johnston L , et al . Imagery rehearsal therapy for chronic nightmares in sexual assault survivors with posttraumatic stress disorder: a randomized controlled trial . JAMA. 2001. ; 286 ( 5 ): 537 – 545 . [DOI] [PubMed] [Google Scholar]
- 123. Kellner R , Neidhardt J , Krakow B , Pathak D . Changes in chronic nightmares after one session of desensitization or rehearsal instructions . Am J Psychiatry. 1992. ; 149 ( 5 ): 659 – 663 . [DOI] [PubMed] [Google Scholar]
- 124. Rauch SAM , Eftekhari A , Ruzek JI . Review of exposure therapy: a gold standard for PTSD treatment . J Rehabil Res Dev. 2012. ; 49 ( 5 ): 679 – 687 . [DOI] [PubMed] [Google Scholar]
- 125. Rothbaum BO , Kearns MC , Price M , et al . Early intervention may prevent the development of posttraumatic stress disorder: a randomized pilot civilian study with modified prolonged exposure . Biol Psychiatry. 2012. ; 72 ( 11 ): 957 – 963 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Bryant RA , Sackville T , Dang ST , Moulds M , Guthrie R . Treating acute stress disorder: an evaluation of cognitive behavior therapy and supportive counseling techniques . Am J Psychiatry. 1999. ; 156 ( 11 ): 1780 – 1786 . [DOI] [PubMed] [Google Scholar]
- 127. Gutner CA , Casement MD , Stavitsky Gilbert K , Resick PA . Change in sleep symptoms across cognitive processing therapy and prolonged exposure: a longitudinal perspective . Behav Res Ther. 2013. ; 51 ( 12 ): 817 – 822 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Walters EM , Jenkins MM , Nappi CM , et al . The impact of prolonged exposure on sleep and enhancing treatment outcomes with evidence-based sleep interventions: a pilot study . Psychol Trauma. 2020. ; 12 ( 2 ): 175 – 185 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. López CM , Lancaster CL , Gros DF , Acierno R . Residual sleep problems predict reduced response to prolonged exposure among veterans with PTSD . J Psychopathol Behav Assess. 2017. ; 39 ( 4 ): 755 – 763 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Reist C , Gory A , Hollifield M . Sleep-disordered breathing impact on efficacy of prolonged exposure therapy for posttraumatic stress disorder . J Trauma Stress. 2017. ; 30 ( 2 ): 186 – 189 . [DOI] [PubMed] [Google Scholar]
- 131. Haynes PL , Skobic I , Epstein DR , et al . Cognitive processing therapy for posttraumatic stress disorder is associated with negligible change in subjective and objective sleep . Behav Sleep Med. 2020. ; 18 ( 6 ): 809 – 819 . [DOI] [PubMed] [Google Scholar]
- 132. Holder N , Holliday R , Wiblin J , Surís A . A preliminary examination of the effect of cognitive processing therapy on sleep disturbance among veterans with military sexual trauma-related posttraumatic stress disorder . Traumatol Tallahass Fla. 2019. ; 25 ( 4 ): 316 – 323 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Haynes PL, Emert SE, Epstein D, Perkins S, Parthasarathy S, Wilcox J. The Effect of Sleep Disorders, Sedating Medications, and Depression on Cognitive Processing Therapy Outcomes: a Fuzzy Set Qualitative Comparative Analysis. J Trauma Stress. 2017;30(6):635–645. Epub 2017 Nov 21. PMID: 29160555. [DOI] [PubMed]
- 134. Galovski TE , Monson C , Bruce SE , Resick PA . Does cognitive-behavioral therapy for PTSD improve perceived health and sleep impairment? J Trauma Stress. 2009. ; 22 ( 3 ): 197 – 204 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Galovski TE , Harik JM , Blain LM , Elwood L , Gloth C , Fletcher TD . Augmenting cognitive processing therapy to improve sleep impairment in PTSD: a randomized controlled trial . J Consult Clin Psychol. 2016. ; 84 ( 2 ): 167 – 177 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Arditte Hall KA , Werner KB , Griffin MG , Galovski TE . The effects of cognitive processing therapy + hypnosis on objective sleep quality in women with posttraumatic stress disorder . Psychol Trauma. 2021. ; 13 ( 6 ): 652 – 656 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Landin-Romero R , Moreno-Alcazar A , Pagani M , Amann BL . How does eye movement desensitization and reprocessing therapy work? A systematic review on suggested mechanisms of action . Front Psychol. 2018. ; 9 : 1395 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Stickgold R . EMDR: a putative neurobiological mechanism of action . J Clin Psychol. 2002. ; 58 ( 1 ): 61 – 75 . [DOI] [PubMed] [Google Scholar]
- 139. Sack M , Zehl S , Otti A , et al . A Comparison of dual attention, eye movements, and exposure only during eye movement desensitization and reprocessing for posttraumatic stress disorder: results from a randomized clinical trial . Psychother Psychosom. 2016. ; 85 ( 6 ): 357 – 365 . [DOI] [PubMed] [Google Scholar]
- 140. Chen L , Zhang G , Hu M , Liang X . Eye movement desensitization and reprocessing versus cognitive-behavioral therapy for adult posttraumatic stress disorder: systematic review and meta-analysis . J Nerv Ment Dis. 2015. ; 203 ( 6 ): 443 – 451 . [DOI] [PubMed] [Google Scholar]
- 141. Kip KE , Diamond DM . Clinical, empirical, and theoretical rationale for selection of accelerated resolution therapy for treatment of post-traumatic stress disorder in VA and DoD facilities . Mil Med. 2018. ; 183 ( 9-10 ): e314 – e321 . [DOI] [PubMed] [Google Scholar]
- 142. Kip KE , Elk CA , Sullivan KL , et al . Brief treatment of symptoms of post-traumatic stress disorder (PTSD) by use of accelerated resolution therapy (ART(®)) . Behav Sci (Basel). 2012. ; 2 ( 2 ): 115 – 134 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Toukolehto OT , Waits WM , Preece DM , Samsey KM . Accelerated Resolution Therapy-Based Intervention in the Treatment of Acute Stress Reactions During Deployed Military Operations . Mil Med. 2020. ; 185 ( 3-4 ): 356 – 362 . [DOI] [PubMed] [Google Scholar]
- 144. Marin MF , Camprodon JA , Dougherty DD , Milad MR . Device-based brain stimulation to augment fear extinction: implications for PTSD treatment and beyond . Depress Anxiety. 2014. ; 31 ( 4 ): 269 – 278 . [DOI] [PubMed] [Google Scholar]
- 145. Camprodon JA , Pascual-Leone A . Multimodal applications of transcranial magnetic stimulation for circuit-based psychiatry . JAMA Psychiatry. 2016. ; 73 ( 4 ): 407 – 408 . [DOI] [PubMed] [Google Scholar]
- 146. Huerta PT , Volpe BT . Transcranial magnetic stimulation, synaptic plasticity and network oscillations. 2009 . J NeuroEngineering Rehabil. 2009. ; 6 : 7 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Sun N , He Y , Wang Z , Zou W , Liu X . The effect of repetitive transcranial magnetic stimulation for insomnia: a systematic review and meta-analysis . Sleep Med. 2021. ; 77 : 226 – 237 . [DOI] [PubMed] [Google Scholar]
- 148. Jiang C-G , Zhang T , Yue F-G , Yi M-L , Gao D . Efficacy of repetitive transcranial magnetic stimulation in the treatment of patients with chronic primary insomnia . Cell Biochem Biophys. 2013. ; 67 : 169 – 173 . [DOI] [PubMed] [Google Scholar]
- 149. Strafella AP , Âs T , Paus Ï , Fraraccio M , Dagher A . Striatal dopamine release induced by repetitive transcranial magnetic stimulation of the human motor cortex . Brain. 2003. ; 126 ( 12 ): 2609 – 2615 . [DOI] [PubMed] [Google Scholar]
- 150. Riemann D , Spiegelhalder K , Feige B , et al . The hyperarousal model of insomnia: a review of the concept and its evidence . Sleep Med Rev. 2010. ; 14 ( 1 ): 19 – 31 . [DOI] [PubMed] [Google Scholar]
- 151. Watts BV , Landon B , Groft A , Young-Xu Y . A sham controlled study of repetitive transcranial magnetic stimulation for posttraumatic stress disorder . Brain Stimul. 2012. ; 5 ( 1 ): 38 – 43 . [DOI] [PubMed] [Google Scholar]
- 152. Nam DH , Pae CU , Chae JH . Low-frequency, repetitive transcranial magnetic stimulation for the treatment of patients with posttraumatic stress disorder: a double-blind, sham-controlled study . Clin Psychopharmacol Neurosci. 2013. ; 11 ( 2 ): 96 – 102 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Isserles M , Shalev AY , Roth Y , et al . Effectiveness of deep transcranial magnetic stimulation combined with a brief exposure procedure in post-traumatic stress disorder–a pilot study . Brain Stimul. 2013. ; 6 ( 3 ): 377 – 383 . [DOI] [PubMed] [Google Scholar]
- 154. Cohen H , Kaplan Z , Kotler M , Kouperman I , Moisa R , Nimrod Grisaru B . Repetitive transcranial magnetic stimulation of the right dorsolateral prefrontal cortex in posttraumatic stress disorder: a double-blind, placebo-controlled study . Am J Psychiatry. 2004. ; 161 ( 3 ): 515 – 524 . [DOI] [PubMed] [Google Scholar]
- 155. Boggio PS , Valasek CA , Campanhã C , et al . Non-invasive brain stimulation to assess and modulate neuroplasticity in Alzheimer’s disease . Neuropsychol Rehabil. 2011. ; 21 ( 5 ): 703 – 716 . [DOI] [PubMed] [Google Scholar]
- 156. Brady K , Pearlstein T , Asnis GM , et al . Efficacy and safety of sertraline treatment of posttraumatic stress disorder: a randomized controlled trial . JAMA. 2000. ; 283 ( 14 ): 1837 – 1844 . [DOI] [PubMed] [Google Scholar]
- 157. Davidson JRT , Rothbaum BO , van der Kolk BA , Sikes CR , Farfel GM . Multicenter, double-blind comparison of sertraline and placebo in the treatment of posttraumatic stress disorder . Arch Gen Psychiatry. 2001. ; 58 ( 5 ): 485 – 492 . [DOI] [PubMed] [Google Scholar]
- 158. Marshall RD , Beebe KL , Oldham M , Zaninelli R . Efficacy and safety of paroxetine treatment for chronic PTSD: a fixed-dose, placebo-controlled study . Am J Psychiatry. 2001. ; 158 ( 12 ): 1982 – 1988 . [DOI] [PubMed] [Google Scholar]
- 159. Alexander W . Pharmacotherapy for post-traumatic stress disorder in combat veterans: focus on antidepressants and atypical antipsychotic agents . P&T. 2012. ; 37 ( 1 ): 32 – 38 . [PMC free article] [PubMed] [Google Scholar]
- 160. Brownlow JA , Harb GC , Ross RJ . Treatment of sleep disturbances in post-traumatic stress disorder: a review of the literature . Curr Psychiatry Rep. 2015. ; 17 ( 6 ): 41 . [DOI] [PubMed] [Google Scholar]
- 161. Schenck H , Mahowald MW , Kim W , O’connor TA , Hurwitz D . Prominent eye movements during NREM sleep and REM sleep behavior disorder associated with fluoxetine treatment of depression and obsessive-compulsive disorder . Sleep. 1992. ; 15 ( 3 ): 226 – 235 . [DOI] [PubMed] [Google Scholar]
- 162. Sheyner I , Khan S , Stewart JT . A case of selective serotonin reuptake inhibitor-induced rapid eye movement behavior disorder . J Am Geriatr Soc. 2010. ; 58 ( 7 ): 1421 – 1422 . [DOI] [PubMed] [Google Scholar]
- 163. Hoque R , Chesson AL Jr . Pharmacologically induced/exacerbated restless legs syndrome, periodic limb movements of sleep, and REM behavior disorder/REM sleep without atonia: literature review, qualitative scoring, and comparative analysis . J Clin Sleep Med. 2010. ; 6 ( 1 ): 79 – 83 . [PMC free article] [PubMed] [Google Scholar]
- 164. Lee K , Baron K , Soca R , Attarian H . The Prevalence and characteristics of REM sleep without atonia (RSWA) in patients taking antidepressants . J Clin Sleep Med. 2016. ; 12 ( 3 ): 351 – 355 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Peever J , Luppi PH , Montplaisir J . Breakdown in REM sleep circuitry underlies REM sleep behavior disorder . Trends Neurosci. 2014. ; 37 ( 5 ): 279 – 288 . [DOI] [PubMed] [Google Scholar]
- 166. Postuma RB , Lanfranchi PA , Blais H , Gagnon JF , Montplaisir JY . Cardiac autonomic dysfunction in idiopathic REM sleep behavior disorder . Mov Disord. 2010. ; 25 ( 14 ): 2304 – 2310 . [DOI] [PubMed] [Google Scholar]
- 167. Lam SP , Zhang J , Tsoh J , et al . REM sleep behavior disorder in psychiatric populations . J Clin Psychiatry. 2010. ; 71 ( 8 ): 1101 – 1103 . [DOI] [PubMed] [Google Scholar]
- 168. Rottach KG , Schaner BM , Kirch MH , et al . Restless legs syndrome as side effect of second generation antidepressants . J Psychiatr Res. 2008. ; 43 ( 1 ): 70 – 75 . [DOI] [PubMed] [Google Scholar]
- 169. Page RL II , Ruscin JM , Bainbridge JL , Brieke AA . Restless legs syndrome induced by escitalopram: case report and review of the literature . Pharmacotherapy. 2008. ; 28 ( 2 ): 271 – 280 . [DOI] [PubMed] [Google Scholar]
- 170. Bailey AL , Makela EH , Asberg K . Selective serotonin reuptake inhibitor/serotonin-norepinephrine reuptake inhibitor use as a predictor of a diagnosis of restless legs syndrome . J Psychiatr Pract. 2016. ; 22 ( 4 ): 263 – 269 . [DOI] [PubMed] [Google Scholar]
- 171. Dunvald AD , Pilsgaard Henriksen D , Hallas J , Marie Hougaard Christensen M , Christian Lund L . Selective serotonin reuptake inhibitors and the risk of restless legs syndrome: a symmetry analysis . Eur J Clin Pharmacol. 2020. ; 76 : 719 – 722 . [DOI] [PubMed] [Google Scholar]
- 172. Kolla BP , Mansukhani MP , Bostwick JM . The influence of antidepressants on restless legs syndrome and periodic limb movements: a systematic review . Sleep Med Rev. 2018. ; 38 : 131 – 140 . [DOI] [PubMed] [Google Scholar]
- 173. Zhang B , Hao Y , Jia F , et al . Sertraline and periodic limb movements during sleep: an 8-week open-label study in depressed patients with insomnia . Sleep Med. 2013. ; 14 ( 12 ): 1405 – 1412 . [DOI] [PubMed] [Google Scholar]
- 174. Armitage R , Trivedi M , Rush AJ . Fluoxetine and oculomotor activity during sleep in depressed patients . Neuropsychopharmacology. 1995. ; 12 ( 2 ): 159 – 165 . [DOI] [PubMed] [Google Scholar]
- 175. Yang C , White DP , Winkelman JW . Antidepressants and periodic leg movements of sleep . Biol Psychiatry. 2005. ; 58 ( 6 ): 510 – 514 . [DOI] [PubMed] [Google Scholar]
- 176. Wilson S , Argyropoulos S . Antidepressants and sleep: a qualitative review of the literature . Drugs. 2005. ; 65 ( 7 ): 927 – 947 . [DOI] [PubMed] [Google Scholar]
- 177. Palagini L , Baglioni C , Ciapparelli A , Gemignani A , Riemann D . REM sleep dysregulation in depression: state of the art . Sleep Med Rev. 2013. ; 17 ( 5 ): 377 – 390 . [DOI] [PubMed] [Google Scholar]
- 178. Stein DJ , Pedersen R , Rothbaum BO , et al . Onset of activity and time to response on individual CAPS-SX17 items in patients treated for post-traumatic stress disorder with venlafaxine ER: a pooled analysis . Int J Neuropsychopharmacol. 2009. ; 12 ( 1 ): 23 – 31 . [DOI] [PubMed] [Google Scholar]
- 179. Salín-Pascual RJ , Galicia-Polo L , Drucker-Colín R . Sleep changes after 4 consecutive days of venlafaxine administration in normal volunteers . J Clin Psychiatry. 1997. ; 58 ( 8 ): 348 – 350 . [DOI] [PubMed] [Google Scholar]
- 180. Stahl SM . Mechanism of action of trazodone: a multifunctional drug . CNS Spectr. 2009. ; 14 ( 10 ): 536 – 546 . [DOI] [PubMed] [Google Scholar]
- 181. Yamadera H , Nakamura S , Suzuki H , Endo S . Effects of trazodone hydrochloride and imipramine on polysomnography in healthy subjects . Psychiatry Clin Neurosci. 1998. ; 52 ( 4 ): 439 – 443 . [DOI] [PubMed] [Google Scholar]
- 182. Argyropoulos SV , Wilson SJ . Sleep disturbances in depression and the effects of antidepressants . Int Rev Psychiatry. 2005. ; 17 ( 4 ): 237 – 245 . [DOI] [PubMed] [Google Scholar]
- 183. Ashford JW , Miller TW . Effects of trazodone on sleep in patients diagnosed with Post-Traumatic Stress Disorder (PTSD) . J Contemp Psychother. 1996. ; 26 ( 3 ): 221 – 233 . [Google Scholar]
- 184. Warner MD , Dorn MR , Peabody CA . Survey on the usefulness of trazodone in patients with PTSD with insomnia or nightmares . Pharmacopsychiatry. 2001. ; 34 ( 4 ): 128 – 131 . [DOI] [PubMed] [Google Scholar]
- 185. Hertzberg MA , Feldman ME , Beckham JC , Davidson JRT . Trial of trazodone for posttraumatic stress disorder using a multiple baseline group design . J Clin Psychopharmacol. 1996. ; 16 ( 4 ): 294 – 298 . [DOI] [PubMed] [Google Scholar]
- 186. Neylan TC , Lenoci M , Maglione ML , et al . The effect of nefazodone on subjective and objective sleep quality in posttraumatic stress disorder . J Clin Psychiatry. 2003. ; 64 ( 4 ): 445 – 450 . [DOI] [PubMed] [Google Scholar]
- 187. Zisook S , Chentsova-Dutton YE , Smith-Vaniz A , et al . Nefazodone in patients with treatment-refractory posttraumatic stress disorder . J Clin Psychiatry. 2000. ; 61 ( 3 ): 203 – 208 . [DOI] [PubMed] [Google Scholar]
- 188. Gillin JC , Smith-Vaniz A , Schnierow B , et al . An open-label, 12-week clinical and sleep EEG study of nefazodone in chronic combat-related posttraumatic stress disorder . J Clin Psychiatry. 2001. ; 62 ( 10 ): 789 – 796 . [DOI] [PubMed] [Google Scholar]
- 189. Hidalgo R , Hertzberg MA , Mellman T , et al . Nefazodone in post-traumatic stress disorder: results from six open-label trials . Int Clin Psychopharmacol. 1999. ; 14 ( 2 ): 61 – 68 . [DOI] [PubMed] [Google Scholar]
- 190. Davidson JRT , Weisler RH , Malik ML , Connor KM . Treatment of posttraumatic stress disorder with nefazodone . Int Clin Psychopharmacol. 1998. ; 13 ( 3 ): 111 – 113 . [DOI] [PubMed] [Google Scholar]
- 191. Friedman MJ . PTSD: pharmacotherapeutic approaches . Focus J Lifelong Learn Psych. 2013. ; 11 ( 3 ): 315 – 320 . [Google Scholar]
- 192. Stewart DE . Hepatic adverse reactions associated with nefazodone . Can J Psychiatry. 2002. ; 47 ( 4 ): 375 – 377 . [DOI] [PubMed] [Google Scholar]
- 193. Yi X yan , Ni S fen , Ghadami MR , et al . Trazodone for the treatment of insomnia: a meta-analysis of randomized placebo-controlled trials . Sleep Med. 2018. ; 45 : 25 – 32 . [DOI] [PubMed] [Google Scholar]
- 194. Van Veen JF , Van der Wee NJA , Fiselier J , Van Vliet IM , Westenberg HGM . Behavioural effects of rapid intravenous administration of meta-chlorophenylpiperazine (m-CPP) in patients with generalized social anxiety disorder, panic disorder and healthy controls . Eur Neuropsychopharmacol. 2007. ; 17 ( 10 ): 637 – 642 . [DOI] [PubMed] [Google Scholar]
- 195. Rothbaum BO , Killeen TK , Davidson JRT , Brady KT , Connor KM , Heekin MH . Placebo-controlled trial of risperidone augmentation for selective serotonin reuptake inhibitor-resistant civilian posttraumatic stress disorder . J Clin Psychiatry. 2008. ; 69 ( 4 ): 520 – 525 . [DOI] [PubMed] [Google Scholar]
- 196. Citrome L . Activating and sedating adverse effects of second-generation antipsychotics in the treatment of schizophrenia and major depressive disorder: absolute risk increase and number needed to harm . J Clin Psychopharmacol. 2017. ; 37 ( 2 ): 138 – 147 . [DOI] [PubMed] [Google Scholar]
- 197. Detweiler MB , Khachiyants N , Detweiler JG , Ali R , Kim KY . Risperidone for post-traumatic combat nightmares: a report of four cases . Consult Pharm. 2011. ; 26 ( 12 ): 920 – 928 . [DOI] [PubMed] [Google Scholar]
- 198. Leyba CM , Wampler TP . Risperidone in PTSD . Psychiatr Serv. 1998. ; 49 ( 2 ): 245 – 246 . [DOI] [PubMed] [Google Scholar]
- 199. Gandotra K , Jaskiw GE , Wilson B , Konicki PE , Rosenberg CE , Strohl KP . Low-dose risperidone diminishes the intensity and frequency of nightmares in post-traumatic stress disorder . Sleep. 2019. ; 42 ( 10 ): zsz144 . [DOI] [PubMed] [Google Scholar]
- 200. Jakovljević M , Šagud M , Mihaljević-Peles A . Olanzapine in the treatment-resistant, combat-related PTSD–a series of case reports . Acta Psychiatr Scand. 2003. ; 107 ( 5 ): 394 – 396, discussion 396 . [DOI] [PubMed] [Google Scholar]
- 201. States JH , St Dennis CD . Chronic sleep disruption and the reexperiencing cluster of posttraumatic stress disorder symptoms are improved by olanzapine: brief review of the literature and a case-based series . Prim Care Companion J Clin Psychiatry. 2003. ; 5 ( 2 ): 74 – 79 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202. David D , De Faria L , Mellman TA . Adjunctive risperidone treatment and sleep symptoms in combat veterans with chronic PTSD . Depress Anxiety. 2006. ; 23 ( 8 ): 489 – 491 . [DOI] [PubMed] [Google Scholar]
- 203. Stein MB , Kline NA , Matloff JL . Adjunctive olanzapine for SSRI-resistant combat-related PTSD: a double-blind, placebo-controlled study . Am J Psychiatry. 2002. ; 159 ( 10 ): 1777 – 1779 . [DOI] [PubMed] [Google Scholar]
- 204. Villarreal G , Hamner MB , Cañive JM , et al . Efficacy of quetiapine monotherapy in posttraumatic stress disorder: a randomized, placebo-controlled trial . Am J Psychiatry. 2016. ; 173 ( 12 ): 1205 – 1212 . [DOI] [PubMed] [Google Scholar]
- 205. Eidelman I , Seedat S , Stein DJ . Risperidone in the treatment of acute stress disorder in physically traumatized in-patients . Depress Anxiety. 2000. ; 11 ( 4 ): 187 – 188 . [DOI] [PubMed] [Google Scholar]
- 206. Nasrallah HA . Atypical antipsychotic-induced metabolic side effects: insights from receptor-binding profiles . Mol Psychiatry. 2008. ; 13 ( 1 ): 27 – 35 . [DOI] [PubMed] [Google Scholar]
- 207. Yu ZH , Jiang HY , Shao L , Zhou YY , Shi HY , Ruan B . Use of antipsychotics and risk of myocardial infarction: a systematic review and meta-analysis . Br J Clin Pharmacol. 2016. ; 82 ( 3 ): 624 – 632 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208. Raskind MA , Peskind ER , Chow B , et al . Trial of prazosin for post-traumatic stress disorder in military veterans . N Engl J Med. 2018. ; 378 ( 6 ): 507 – 517 . [DOI] [PubMed] [Google Scholar]
- 209. Mellman TA , Kumar A , Kulick-Bell R , Kumar M , Nolan B . Nocturnal/daytime urine noradrenergic measures and sleep in combat-related PTSD . Biol Psychiatry. 1995. ; 38 ( 3 ): 174 – 179 . [DOI] [PubMed] [Google Scholar]
- 210. Kung S , Espinel Z , Lapid MI . Treatment of nightmares with prazosin: a systematic review . Mayo Clin Proc. 2012. ; 87 ( 9 ): 890 – 900 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211. Writer BW , Meyer EG , Schillerstrom JE . Prazosin for military combat-related PTSD nightmares: a critical review . J Neuropsychiatry Clin Neurosci. 2014. ; 26 ( 1 ): 24 – 33 . [DOI] [PubMed] [Google Scholar]
- 212. Singh B , Hughes AJ , Mehta G , Erwin PJ , Parsaik AK . Efficacy of prazosin in posttraumatic stress disorder: a systematic review and meta-analysis . Prim Care Companion CNS Disord. 2016. ; 18 ( 4 ): 18 . [DOI] [PubMed] [Google Scholar]
- 213. Raskind MA , Peterson K , Williams T , et al . A trial of prazosin for combat trauma PTSD with nightmares in active-duty soldiers returned from Iraq and Afghanistan . Am J Psychiatry. 2013. ; 170 ( 9 ): 1003 – 1010 . [DOI] [PubMed] [Google Scholar]
- 214. Raskind MA , Peskind ER , Hoff DJ , et al . A parallel group placebo controlled study of prazosin for trauma nightmares and sleep disturbance in combat veterans with post-traumatic stress disorder . Biol Psychiatry. 2007. ; 61 ( 8 ): 928 – 934 . [DOI] [PubMed] [Google Scholar]
- 215. Raskind MA , Peskind ER , Kanter ED , et al . Reduction of nightmares and other PTSD symptoms in combat veterans by prazosin: a placebo-controlled study . Am J Psychiatry. 2003. ; 160 ( 2 ): 371 – 373 . [DOI] [PubMed] [Google Scholar]
- 216. Germain A , Richardson R , Moul DE , et al . Placebo-controlled comparison of prazosin and cognitive-behavioral treatments for sleep disturbances in US Military Veterans . J Psychosom Res. 2012. ; 72 ( 2 ): 89 – 96 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217. Taylor F , Raskind MA . The α1-adrenergic antagonist prazosin improves sleep and nightmares in civilian trauma posttraumatic stress disorder . J Clin Psychopharmacol. 2002. ; 22 ( 1 ): 82 – 85 . [DOI] [PubMed] [Google Scholar]
- 218. Taylor FB , Martin P , Thompson C , et al . Prazosin effects on objective sleep measures and clinical symptoms in civilian trauma posttraumatic stress disorder: a placebo-controlled study . Biol Psychiatry. 2008. ; 63 ( 6 ): 629 – 632 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219. Raskind MA , Dobie DJ , Kanter ED , Petrie EC , Thompson CE , Peskind ER . The α1-adrenergic antagonist prazosin ameliorates combat trauma nightmares in veterans with posttraumatic stress disorder: a report of 4 cases . J Clin Psychiatry. 2000. ; 61 ( 2 ): 129 – 133 . [DOI] [PubMed] [Google Scholar]
- 220. Peskind ER , Bonner LT , Hoff DJ , Raskind MA . Prazosin reduces trauma-related nightmares in older men with chronic posttraumatic stress disorder . J Geriatr Psychiatry Neurol. 2003. ; 16 ( 3 ): 165 – 171 . [DOI] [PubMed] [Google Scholar]
- 221. Prince JB , Wilens TE , Biederman J , Spencer TJ , Wozniak JR . Clonidine for sleep disturbances associated with attention-deficit hyperactivity disorder: a systematic chart review of 62 cases . J Am Acad Child Adolesc Psychiatry. 1996. ; 35 ( 5 ): 599 – 605 . [DOI] [PubMed] [Google Scholar]
- 222. Alao A , Selvarajah J , Razi S . The use of clonidine in the treatment of nightmares among patients with co-morbid PTSD and traumatic brain injury . Int J Psychiatry Med. 2012. ; 44 ( 2 ): 165 – 169 . [DOI] [PubMed] [Google Scholar]
- 223. Burek GA , Waite MR , Heslin K , Liewen AK , Yaqub TM , Larsen SE . Low-dose clonidine in veterans with Posttraumatic stress disorder . J Psychiatr Res. 2021. ; 137 : 480 – 485 . [DOI] [PubMed] [Google Scholar]
- 224. Porter DM , Bell CC . The use of clonidine in post-traumatic stress disorder . J Natl Med Assoc. 1999. ; 91 ( 8 ): 475 – 477 . [PMC free article] [PubMed] [Google Scholar]
- 225. Kinzie JD , Leung P . Clonidine in Cambodian patients with posttraumatic stress disorder . J Nerv Ment Dis. 1989. ; 177 ( 9 ): 546 – 550 . [DOI] [PubMed] [Google Scholar]
- 226. Ziegenhorn AA , Roepke S , Schommer NC , et al . Clonidine improves hyperarousal in borderline personality disorder with or without comorbid posttraumatic stress disorder: a randomized, double-blind, placebo-controlled trial . J Clin Psychopharmacol. 2009. ; 29 ( 2 ): 170 – 173 . [DOI] [PubMed] [Google Scholar]
- 227. Gentili A , Godschalk MF , Gheorghiu D , Nelson K , Julius DA , Mulligan T . Effect of clonidine and yohimbine on sleep in healthy men: a double-blind, randomized, controlled trial . Eur J Clin Pharmacol. 1996. ; 50 ( 6 ): 463 – 465 . [DOI] [PubMed] [Google Scholar]
- 228. Vaiva G , Ducrocq F , Jezequel K , et al . Immediate treatment with propranolol decreases posttraumatic stress disorder two months after trauma . Biol Psychiatry. 2003. ; 54 ( 9 ): 947 – 949 . [DOI] [PubMed] [Google Scholar]
- 229. McGhee LL , Maani CV , Garza TH , Desocio PA , Gaylord KM , Black IH . The effect of propranolol on posttraumatic stress disorder in burned service members . J Burn Care Res. 2009. ; 30 ( 1 ): 92 – 97 . [DOI] [PubMed] [Google Scholar]
- 230. Hoge EA , Worthington JJ , Nagurney JT , et al . Effect of acute posttrauma propranolol on PTSD outcome and physiological responses during script-driven imagery . CNS Neurosci Ther. 2012. ; 18 ( 1 ): 21 – 27 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 231. Argolo FC , Cavalcanti-Ribeiro P , Netto LR , Quarantini LC . Prevention of posttraumatic stress disorder with propranolol: a meta-analytic review . J Psychosom Res. 2015. ; 79 ( 2 ): 89 – 93 . [DOI] [PubMed] [Google Scholar]
- 232. Kindt M , van Emmerik A . New avenues for treating emotional memory disorders: towards a reconsolidation intervention for posttraumatic stress disorder . Ther Adv Psychopharmacol. 2016. ; 6 ( 4 ): 283 – 295 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Brunet A , Saumier D , Liu A , Streiner DL , Tremblay J , Pitman RK . Reduction of PTSD symptoms with pre-reactivation propranolol therapy: a randomized controlled trial . Am J Psychiatry. 2018. ; 175 ( 5 ): 427 – 433 . [DOI] [PubMed] [Google Scholar]
- 234. Danjou P , Puech A , Warot D , Benoit JF . Lack of sleep-inducing properties of propranolol (80 mg) in chronic insomniacs previously treated by common hypnotic medications . Int Clin Psychopharmacol. 1987. ; 2 ( 2 ): 135 – 140 . [DOI] [PubMed] [Google Scholar]
- 235. Bernabeu-Wittel J , Narváez-Moreno B , de la Torre-García JM , et al . Oral nadolol for children with infantile hemangiomas and sleep disturbances with oral propranolol . Pediatr Dermatol. 2015. ; 32 ( 6 ): 853 – 857 . [DOI] [PubMed] [Google Scholar]
- 236. Betts TA , Alford C . β-blockers and sleep: a controlled trial . Eur J Clin Pharmacol. 1985. 28 , S1, Suppl : 65 – 68 . [DOI] [PubMed] [Google Scholar]
- 237. Stoschitzky K , Sakotnik A , Lercher P , et al . Influence of beta-blockers on melatonin release . Eur J Clin Pharmacol. 1999. ; 55 ( 2 ): 111 – 115 . [DOI] [PubMed] [Google Scholar]
- 238. Rothbaum BO , Price M , Jovanovic T , et al . A randomized, double-blind evaluation of D-cycloserine or alprazolam combined with virtual reality exposure therapy for posttraumatic stress disorder in Iraq and Afghanistan war veterans . Am J Psychiatry. 2014. ; 171 ( 6 ): 640 – 648 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239. Guina J , Rossetter SR , DeRhodes BJ , Nahhas RW , Welton RS . Benzodiazepines for PTSD: a systematic review and meta-analysis . J Psychiatr Pract. 2015. ; 21 ( 4 ): 281 – 303 . [DOI] [PubMed] [Google Scholar]
- 240. Mellman TA , Bustamante V , David D , Fins AI . Hypnotic medication in the aftermath of trauma . J Clin Psychiatry. 2002. ; 63 ( 12 ): 1183 – 1184 . [DOI] [PubMed] [Google Scholar]
- 241. Borbély AA , Mattmann P , Loepfe M , Strauch I , Lehmann D . Effect of benzodiazepine hypnotics on all-night sleep EEG spectra . Hum Neurobiol. 1985. ; 4 ( 3 ): 189 – 194 . [PubMed] [Google Scholar]
- 242. Pollack MH , Hoge EA , Worthington JJ , et al et al. Eszopiclone for the treatment of posttraumatic stress disorder and associated insomnia: a randomized, double-blind, placebo-controlled trial . J Clin Psychiatry. 2011. ; 72 ( 7 ): 892 – 897 . [DOI] [PubMed] [Google Scholar]
- 243. Shayegani R , Song K , Amuan ME , Jaramillo CA , Eapen BC , Pugh MJ . Patterns of zolpidem use among Iraq and Afghanistan veterans: a retrospective cohort analysis . PLoS One. 2018. ; 13 ( 1 ): e0190022 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244. Dowd SM , Zalta AK , Burgess HJ , Adkins EC , Valdespino-Hayden Z , Pollack MH . Double-blind randomized controlled study of the efficacy, safety and tolerability of eszopiclone vs placebo for the treatment of patients with post-traumatic stress disorder and insomnia . World J Psychiatry. 2020. ; 10 ( 3 ): 21 – 28 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245. Flores Á , Saravia R , Maldonado R , Berrendero F . Orexins and fear: implications for the treatment of anxiety disorders . Trends Neurosci. 2015. ; 38 ( 9 ): 550 – 559 . [DOI] [PubMed] [Google Scholar]
- 246. Kuriyama A , Tabata H . Suvorexant for the treatment of primary insomnia: a systematic review and meta-analysis . Sleep Med Rev. 2017. ; 35 : 1 – 7 . [DOI] [PubMed] [Google Scholar]
- 247. Prajapati SK , Krishnamurthy S . Non-selective orexin-receptor antagonist attenuates stress-re-stress-induced core PTSD-like symptoms in rats: behavioural and neurochemical analyses . Behav Brain Res. 2021. ; 399 : 113015 . [DOI] [PubMed] [Google Scholar]
- 248. Salehabadi S , Abrari K , Elahdadi Salmani M , Nasiri M , Lashkarbolouki T . Investigating the role of the amygdala orexin receptor 1 in memory acquisition and extinction in a rat model of PTSD . Behav Brain Res. 2020. ; 384 : 112455 . [DOI] [PubMed] [Google Scholar]
- 249. Tabata H , Kuriyama A , Yamao F , Hiroshi K , Shindo K . Suvorexant for the treatment of primary insomnia: a systematic review and meta-analysis . J Clin Sleep Med. 2017. ; 35 : 1-7 . [DOI] [PubMed] [Google Scholar]
- 250. Ware MA , Daeninck P , Maida V . A review of nabilone in the treatment of chemotherapy-induced nausea and vomiting . Ther Clin Risk Manag. 2008. ; 4 ( 1 ): 99 – 107 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251. Babson KA , Sottile J , Morabito D . Cannabis, cannabinoids, and sleep: a review of the literature . Curr Psychiatry Rep. 2017. ; 19 ( 4 ): 23 . [DOI] [PubMed] [Google Scholar]
- 252. Cameron C , Watson D , Robinson J . Use of a synthetic cannabinoid in a correctional population for posttraumatic stress disorder-related insomnia and nightmares, chronic pain, harm reduction, and other indications: a retrospective evaluation . J Clin Psychopharmacol. 2014. ; 34 ( 5 ): 559 – 564 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253. Jetly R , Heber A , Fraser G , Boisvert D . The efficacy of nabilone, a synthetic cannabinoid, in the treatment of PTSD-associated nightmares: a preliminary randomized, double-blind, placebo-controlled cross-over design study . Psychoneuroendocrinology. 2015. ; 51 : 585 – 588 . [DOI] [PubMed] [Google Scholar]
- 254. Norman SB , Stein MB , Dimsdale JE , Hoyt DB . Pain in the aftermath of trauma is a risk factor for post-traumatic stress disorder . Psychol Med. 2008. ; 38 ( 4 ): 533 – 542 . [DOI] [PubMed] [Google Scholar]
- 255. Kesner AJ , Lovinger DM . Cannabinoids, endocannabinoids and sleep . Front Mol Neurosci. 2020. ; 13 : 125 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256. Murrough JW , Perez AM , Pillemer S , et al . Rapid and longer-term antidepressant effects of repeated ketamine infusions in treatment-resistant major depression . Biol Psychiatry. 2013. ; 74 ( 4 ): 250 – 256 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257. Orozco-Solis R , Montellier E , Aguilar-Arnal L , et al . A circadian genomic signature common to ketamine and sleep deprivation in the anterior cingulate cortex . Biol Psychiatry. 2017. ; 82 ( 5 ): 351 – 360 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258. Feder A , Parides MK , Murrough JW , et al . Efficacy of intravenous ketamine for treatment of chronic posttraumatic stress disorder: a randomized clinical trial . JAMA Psychiatry. 2014. ; 71 ( 6 ): 681 – 688 . [DOI] [PubMed] [Google Scholar]
- 259. D’Andrea D , Andrew Sewell R . Transient resolution of treatment-resistant posttraumatic stress disorder following ketamine infusion . Biol Psychiatry. 2013. ; 74 ( 9 ): e13 – e14 . [DOI] [PubMed] [Google Scholar]
- 260. Duncan WC , Sarasso S , Ferrarelli F , et al . Concomitant BDNF and sleep slow wave changes indicate ketamine-induced plasticity in major depressive disorder . Int J Neuropsychopharmacol. 2013. ; 16 ( 2 ): 301 – 311 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261. Schönenberg M , Reichwald U , Domes G , Badke A , Hautzinger M . Effects of peritraumatic ketamine medication on early and sustained posttraumatic stress symptoms in moderately injured accident victims . Psychopharmacology (Berl). 2005. ; 182 ( 3 ): 420 – 425 . [DOI] [PubMed] [Google Scholar]
- 262. Schönenberg M , Reichwald U , Domes G , Badke A , Hautzinger M . Ketamine aggravates symptoms of acute stress disorder in a naturalistic sample of accident victims . J Psychopharmacol. 2008. ; 22 ( 5 ): 493 – 497 . [DOI] [PubMed] [Google Scholar]
- 263. McGhee LL , Maani CV , Garza TH , Gaylord KM , Black IH . The correlation between ketamine and posttraumatic stress disorder in burned service members . J Trauma. 2008. 64 ( 2 ): S195 – S199 , Discussion S197-S198. [DOI] [PubMed] [Google Scholar]
- 264. McGhee LL , Maani CV , Garza TH , Slater TM , Petz LN , Fowler M . The intraoperative administration of ketamine to burned U.S. service members does not increase the incidence of post-traumatic stress disorder . Mil Med. 2014. ; 179 ( suppl_8 ): 41 – 46 . [DOI] [PubMed] [Google Scholar]
- 265. Zeng MC , Niciu MJ , Luckenbaugh DA , et al . Acute stress symptoms do not worsen in posttraumatic stress disorder and abuse with a single subanesthetic dose of ketamine . Biol Psychiatry. 2013. ; 73 ( 12 ): e37 – e38 . [DOI] [PubMed] [Google Scholar]
- 266. Mion G , Le Masson J , Granier C , Hoffmann C . A retrospective study of ketamine administration and the development of acute or post-traumatic stress disorder in 274 war-wounded soldiers . Anaesthesia. 2017. ; 72 ( 12 ): 1476 – 1483 . [DOI] [PubMed] [Google Scholar]
- 267. Mitchell JM , Bogenschutz M , Lilienstein A , et al . MDMA-assisted therapy for severe PTSD: a randomized, double-blind, placebo-controlled phase 3 study . Nat Med. 2021. ; 27 ( 6 ): 1025 – 1033 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268. Tedesco S , Gajaram G , Chida S , et al . The efficacy of MDMA (3,4-Methylenedioxymethamphetamine) for post-traumatic stress disorder in humans: a systematic review and meta-analysis . Cureus. 2021. ; 13 ( 5 ): e15070 . [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269. Smith KW , Sicignano DJ , Hernandez AV , White CM . MDMA-assisted psychotherapy for treatment of posttraumatic stress disorder: a systematic review with meta-analysis . J Clin Pharmacol. 2022. ; 62 ( 4 ): 463 – 471 . [DOI] [PubMed] [Google Scholar]
- 270. Baylen CA , Rosenberg H . A review of the acute subjective effects of MDMA/ecstasy . Addiction. 2006. ; 101 ( 7 ): 933 – 947 . [DOI] [PubMed] [Google Scholar]