Correspondence to Dr Ben Julian Agustin Palanca; [email protected]
STRENGTHS AND LIMITATIONS OF THIS STUDY
Real-time analysis of drug pharmacokinetics and high-density electroencephalographic (EEG) signals facilitate targeting of propofol to maximise induction of EEG slow waves.
At-home unattended longitudinal sleep studies, using wireless EEG headbands equipped with dry electrodes enables assessment of slow wave sleep enhancement.
While small in sample size, open label in design and lacking a control group, the study will lay the groundwork for a future phase II investigation.
Introduction
Depression and the need for novel therapeutics
Depression negatively impacts quality of life and imparts emotional, physical and economic consequences. Depressive symptoms, such as low mood and anhedonia, are associated with increased all-cause mortality and cardiovascular disease.1 The annual economic impact of depression in the USA exceeds US$90 billion.2 Of the 9 million Americans who require medication treatment for depression each year, nearly a third are considered treatment-resistant.2 Treatment-resistant depression (TRD), commonly defined as failure to achieve remission after adequate trials of two antidepressants, is a leading cause of disability,3 suicide4 5 and dementia in older adults.6–8 The prevalence of late-life TRD (LL-TRD) will likely increase as the population ages.9 10 Remission rates with conventional antidepressants are low in LL-TRD,11 and long-term outcomes are dismal, with high recurrence rates in spite of the currently available treatment options.12 13
Current pharmacotherapies for depression target one or more neurotransmitter systems as first-line options,14 with remission in 55%–81% of older adults with depression.15–17 Novel treatments targeting core pathophysiology are desperately needed for the large population of patients who do not benefit from traditional depression treatment strategies. While electroconvulsive therapy18 19 and transcranial magnetic stimulation20 may be viable options for some patients, there remains an unmet need for unique treatment modalities. A potential target for these therapies is sleep disruption,21 which contributes to refractory depression,22 impaired attention and executive dysfunction.23
Sleep, cognition and depression in the ageing brain
Sleep is critical for daily physical and mental restoration. It is composed of rapid eye movement (REM) and non-rapid eye movement (NREM) sleep, each with distinct physiological roles. Slow wave sleep (SWS)/NREM stage 3 (N3)24 is associated alleviation of neurohumoral stress responses.25 Several crucial processes occur during SWS, including synaptic remodelling and memory replay.26 These promote plasticity and memory consolidation.27 Accordingly, SWS enhancement can improve memory.28 29
SWS may be particularly important for maintaining cognitive ability as we age. Slow waves have been shown to regulate the chemical environment within the brain through the glymphatic system30 by stimulating cerebrospinal fluid waves during sleep.31 Proper functioning of this system may be protective against Alzheimer’s disease and related dementias by flushing out amyloid-beta30 32 and tau33 proteins. An inverse relationship exists between the deposition of dementia-associated proteins and measures of SWS in older adults.34 Thus, ensuring sufficient SWS may promote healthier brain ageing. Chronic SWS deficits may damage circuitry underlying the regulation of sleep and circadian rhythms, compounding the effects of poor sleep.
There is a well-established relationship between depression and sleep disturbances. SWS deficiency is a known risk factor for depression recurrence35 and suicidality.36 Numerous antidepressants are known to augment SWS, including lithium,37–39 trazodone,40 mirtazapine,41 sertraline,42 clomipramine,43 ketamine44 and agomelatine.45 SWS is characterised by large-amplitude electroencephalographic (EEG) oscillations known as delta waves in the 0.5–4 Hz frequency band (figure 1A). The squared amplitude, or power, in these slow waves can be expressed as the slow wave activity (SWA).46–48 In healthy individuals, SWA and SWS duration peak during the first N3 cycle of the night then diminish throughout subsequent cycles as REM sleep becomes more frequent. SWA can be viewed as a marker of synaptic plasticity.46–48
Figure 1. Electroencephalographic (EEG) slow waves and burst suppression evoked from a single participant. During natural sleep, the predominance of high amplitude EEG slow waves in the delta band (delta waves) define slow wave sleep (SWS)/stage 3 non-rapid eye movement sleep (N3) (A). EEG slow waves can also be induced during propofol general anaesthesia (B). With high doses of propofol, EEG burst suppression arises (C). Data were acquired from a 71-year-old woman during the pilot study. 97
The decline in SWA across successive cycles of SWS is thought to reflect dissipation of sleep pressure and restoration of synaptic homeostasis.49 Reduced nighttime SWA, particularly in the first cycle,43 50–54 appears to be a core feature of depression pathophysiology, regardless of age.55 The shift in peak SWA can be quantified by the delta sleep ratio (DSR), which is computed by dividing the SWA of the first N3 cycle by the SWA of the second N3 cycle.56 Ratios >1.5 are typically observed in patients without depression. Changes in sleep architecture accompany antidepressant response. DSR has been used as a marker of antidepressant responsiveness57 and as a risk factor for recurrence.43 56
Depression-related changes in SWS are often accompanied by abnormal REM sleep. The delay in SWS that is characteristic of depression corresponds to REM intrusion into early sleep cycles. Greater REM durations are also often observed in depressed patients. Restoring SWS to early cycles normalises sleep architecture, allowing REM to dominate later sleep cycles. Later REM onset and reduced REM duration are observed with antidepressant response.
Poor sleep is also associated with cognitive dysfunction in older adults,23 with important implications for depressed patients. LL-TRD is often accompanied by impairments in executive functions,58 59 such as set-shifting (cognitive flexibility) and working memory.60 Poor executive function is associated with inadequate response to oral antidepressant treatment.12 58 61–65 Slowed processing speed, reduced alertness and impaired executive function are also associated with depression and sleep disturbances.66 67 Despite the reciprocal relationships between depression, cognitive dysfunction and sleep disturbances in older adults,68 no studies have specifically targeted SWS to treat TRD and improve cognitive function in these patients.
Propofol modulates sleep and mood
Propofol, a commonly used anaesthetic, induces dose-dependent EEG patterns with varied resemblance to natural sleep.69 70 With doses sufficient to induce unconsciousness, EEG slow waves emerge71 72 (figure 1B) resembling those of N3/SWS70 (figure 1A). Even higher doses elicit burst suppression,73–75 an EEG pattern characterised by episodes of suppressed cortical activity punctuated by bursts of mixed-frequency activity not seen in natural sleep (figure 1C). The electrophysiological similarities between propofol sedation and SWS may underlie its ability to fulfil SWS deficits in rodents.76 Patients often report subjective feelings of restorative sleep after propofol infusions, motivating its use as a treatment for refractory insomnia.77 78 In adults with chronic refractory insomnia, five nightly 2-hour propofol infusions from 22:00 to midnight enhanced SWS at least 6 months after therapy.78 Propofol-related changes in sleep architecture have not yet been linked to antidepressant effects in humans.
While antidepressant effects of propofol have been described, the role of SWS enhancement remains unexplored. After propofol anaesthesia, euphoria,79 80 amorous behaviour81 82 and elation83 may last for several hours.84 An antidepressant effect appears to be dose-dependent.85 Subanaesthetic infusions targeting plasma concentrations of 0.9 µg/mL were associated with no improvements in mood,86 while 10 high-dose propofol infusions, targeting at least 15 min of EEG burst suppression, correlated with antidepressant responses lasting up to 3 months.87 In rodent models, propofol pretreatment augments the antidepressant effects of ketamine88 and moderate-dose electroconvulsive therapy.89 There are numerous potential mechanisms underlying propofol’s antidepressant action. While the primary molecular mechanism of action underlying its hypnotic effects is GABA (γ-aminobutyric acid) agonism,90 91 propofol has additional potentially contributory molecular actions, including NMDA (N-methyl-D-aspartic acid) antagonism92 and AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) agonism.93 94 Recent animal studies demonstrate that propofol blocks dopamine transporters in the nucleus accumbens, leading to increased dopamine within this structure and reduced anhedonia.95 Interestingly, the nucleus accumbens contains a core set of neurons that regulate SWS.96 Thus, further work is needed to investigate relationships between propofol, SWS and depression.
We propose using propofol as a therapeutic probe to enhance SWS in older adults with TRD. Proof-of-concept was provided by a pilot study in which two participants were treated with two propofol infusions.97 One patient demonstrated EEG slow waves and minimal burst suppression during infusions. SWS enhancement was noted on postinfusion nights. The patient subsequently exhibited an antidepressant response that was sustained for at least several months. Another patient did not exhibit slow waves during infusions, postinfusion SWS enhancement, nor antidepressant response. These cases demonstrated that propofol can enhance SWS and improve depression for select individuals, motivating a subsequent clinical trial. Below, we present the protocol for the ongoing open-label phase I study, which will evaluate propofol’s ability to induce EEG slow waves and augment SWS in patients with LL-TRD.
Methods and analysis
Study design overview
Slow Wave Induction by Propofol to Eliminate Depression (SWIPED) (ClinicalTrials.gov NCT04680910) is a phase I clinical trial assessing the safety and feasibility of propofol infusions for SWS enhancement in older adults with depression (figure 2). There is no randomisation in this open-label single-arm investigation. The study tests the hypothesis that targeted intravenous propofol infusions in patients with LL-TRD induce SWA during sedation and augment subsequent sleep SWA. This single-centre trial takes place at Washington University in St. Louis Medical Center. Participants are undergoing two propofol infusions, with each infusion maximising the expression of EEG slow waves. Preinfusion and postinfusion sleep EEG recordings and assessments of mood and cognitive function are used to evaluate the correlations between propofol infusions and changes in sleep architecture, mood and cognition. This protocol is based on the latest approved protocol (version 3.1.5., 30 January 2023).
Figure 2. Study timeline. 15 study participants will undergo two 2-hour propofol infusions, high-density EEG, serial at-home unattended sleep studies, cognitive assessments and evaluations of depression, anhedonia and suicidality. Participants will complete a sleep diary. A blood biobanking study is optional, as marked by asterisks. C-SSRS, Columbia Suicide Severity Rating Scale; EEG, electroencephalography; HD, high density; MADRS, Montgomery-Åsberg Depression Rating Scale; MoCA, Montreal Cognitive Assessment; NIH, National Institutes of Health; PHQ-9, Patient Health Questionnaire-9; PVT, Psychomotor Vigilance Test; SHAPS, Snaith-Hamilton Pleasure Scale.
Study participants
Inclusion and exclusion criteria
This trial enrolled 15 older adults with TRD, defined as persistent depression despite adequate trials of two oral antidepressants (table 1). Exclusion criteria were chosen to maximise safety during propofol infusions (eg, severe left ventricular systolic dysfunction) and to avoid factors that impact SWS (eg, heavy alcohol consumption and certain medications).
Table 1Study inclusion and exclusion criteria for enrolment
| Inclusion criteria | Exclusion criteria |
|
|
|
|
|
|
| |
| |
| |
| |
| |
| |
| |
| |
|
bpm, beats per minute; C-SSRS, Columbia Suicide Severity Rating Scale; LVEF, left ventricular ejection fraction; MoCA, Montreal Cognitive Assessment; NYHA, New York Heart Association; RV, right ventricular.
Recruitment, enrolment and retention
Participants are recruited from the Geriatric Depression Clinic at Washington University in St. Louis, prior clinical trials and social media. Patients are compensated based on the number of study procedures completed, up to US$305 in total. Initial study eligibility is determined via phone screening. The phone screening employs the Patient Health Questionnaire-9 (PHQ-9) to enrol patients who have at least moderate depression (PHQ-9 ≥10).98 Eligible participants undergo a comprehensive review of their medical history, a physical exam, sleep apnoea screening and a urine drug screen. In addition, they undergo the psychiatric and cognitive assessments below. Participants are assigned a study ID with confidentiality of personal health information.
Withdrawal and replacement
Participants may withdraw from the study at any time. The study team may withdraw a participant if there is significant non-compliance with study procedures or if study procedures are poorly tolerated. Patients requiring repeated boluses of ephedrine (total dose >50 mg) for persistent hypotension would be replaced after study session termination.
Participants withdrawn or discontinued before both propofol infusions have been completed would be replaced and would not count towards recruitment target or milestones.
Intervention
Propofol dosing
The Eleveld pharmacokinetic model is used to maximise the stability of propofol effect site concentration in the brain, based on the participant’s sex, age, height and weight.99 To precisely target EEG slow waves while maintaining cardiopulmonary stability, infusion rates range up to 200 µg/kg/min. When possible, real-time target-controlled infusion modelling is implemented. Real-time EEG monitoring during infusions enables dose titration to maximise the expression of EEG slow waves. These visualisations include raw time-series, time-frequency spectrograms and spectral power measures. The presence and amplitude of EEG slow waves and burst suppression guide changes in propofol infusion rates. Dose titration during the first infusion informs subsequent propofol dosing, enabling rapid, stable target engagement of EEG slow waves during the second infusion, administered 2–6 days later.
Infusion visit procedures
Participants fast prior to infusions according to American Society of Anesthesiologists (ASA) guidelines. ACE inhibitors and angiotensin receptor blockers are held on infusion days to mitigate the risk of hypotension. Otherwise, patients generally continue their usual medication regimens throughout the study period.
Infusions take place at the Washington University in St. Louis Clinical Trial Research Unit. After placement of a peripheral intravenous catheter, propofol is infused for 2 hours. Patients are continuously monitored by a board-certified anaesthesiologist. Standard monitors (blood pressure, pulse oximetry, ECG and CO2 capnography) are employed in accordance with ASA guidelines. Supplemental oxygen is administered to all patients. Chin lift/jaw thrust manoeuvres are used to alleviate airway obstruction. Glycopyrrolate, intravenous fluids, phenylephrine and ephedrine are used to treat haemodynamic disturbances. The anaesthetic protocol would be discontinued prematurely if the patient continues to exhibit profound bradycardia, hypotension or hypoxaemia, or sustained cardiac conduction abnormalities refractory to intervention. After each infusion, patients are monitored by a nurse until they meet standard criteria for discharge from the postanaesthesia care unit.100 Participants must be taken home by a responsible adult per institutional policy and are counselled not to work or drive until the next day. There are no provisions for ancillary or post-trial care.
Data collection
Safety outcomes
Adverse events are reviewed at each study visit as primary safety outcomes. The most common adverse events associated with propofol infusions are pain on injection, bradycardia, hypotension, hypoxaemia, hypercapnia, airway obstruction and apnoea. The description of the adverse experience includes the time of onset, duration, intensity, aetiology, relationship to the study drug (none, unlikely, possible, probable, highly probable) and any treatment required.
High-density EEG
Patients undergo 64-channel EEG recordings at enrolment, during propofol infusions and at the second follow-up visit. A MagStim system and 64-channel GSN nets (MagStim, Whitland, UK) are used. Elefix conductive paste (Nihon Kohden, Tokyo, Japan) is injected within the Ag/AgCl electrode sensors to keep impedances below 100 kOhms/electrode. Data are acquired with a Net Amps 400 amplifier and Net Station Software V.5.0. An Axis P3364LV network camera, synchronised to EEG recordings, provides video for post hoc analysis. Eyes open and eyes closed recording periods (10 min each) are obtained.
At-home overnight sleep EEG recordings
Participants conduct at-home sleep EEG recordings using the third-generation Dreem device (Beacon Biosignals, Boston, Massachusetts, USA), a wearable, wireless EEG headband with dry electrodes positioned over frontal (F7, F8 and Fpz) and occipital (O1 and O2) locations.101 To maximise patient compliance and EEG signal quality, device usage and adjustment for proper fit are demonstrated during enrolment. Participants conduct at least two overnight recordings in the week prior to their first propofol infusion, to account for acclimation to device wear and user errors. They provide overnight recordings on the nights of the infusions. After the second propofol infusion, participants will record up to five additional sleep studies over 3 weeks. Participants use a portable wireless hotspot to allow for remote data quality monitoring of at-home sleep recordings.
Psychiatric assessments
Depression severity is evaluated at enrolment using the Montgomery-Åsberg Depression Rating Scale (MADRS).102 In addition, baseline suicidality and anhedonia are assessed on enrolment via the Columbia Suicide Severity Rating Scale (C-SSRS)103 and Snaith-Hamilton Pleasure Scale (SHAPS),104 respectively. The MADRS, C-SSRS and SHAPS questionnaires are repeated approximately 1, 3 and 10 weeks after the second propofol infusion (figure 2). The Feeling Scale105 are used during infusion visits to gauge participants’ affect within an hour before propofol administration and approximately 30 min after return of consciousness.
Cognitive assessments
The Montreal Cognitive Assessment (MoCA)106 is administered at enrolment and approximately 3 weeks after the second propofol infusion (figure 2). An alternate version of the MoCA is used for the post-treatment in-person assessment to prevent confounding due to recall of test items.
To examine cognitive function, feasibility of including the National Institutes of Health (NIH) Toolbox Cognition Battery65 and Psychomotor Vigilance Test (PVT)107 are assessed. Baseline testing occurs at enrolment, with follow-up at approximately 3 weeks after the second propofol infusion (figure 2). The NIH Toolbox Cognition Battery is a computer-based instrument that assesses fluid and crystallised cognition through the following five component tests (and tested cognitive domains): The Flanker Inhibitory Control Test (executive function-sustained attention and inhibitory control), the Dimensional Change Card Sort test (executive function-cognitive flexibility), the List Sorting Working Memory Test (working memory), the Picture Sequence Memory Test (episodic memory) and the Pattern Comparison Processing Test (processing speed). The PVT is used to assess vigilance/alertness in the context of poor sleep and psychiatric illness and can be administered on touchscreen devices.108 It requires the participant to respond as quickly as possible and to maintain sustained vigilance during random intertrial intervals of several seconds. The task generates a right skewed distribution in reaction time (RT), that can be assessed in terms of lapses (outliers) as well as overall metrics of speed (the reciprocal of RT). With sleep deprivation, there is an increased number of lapses (RT >500 ms).109
Assessment of circadian rhythms
Circadian rhythms are assessed over the course of the study. The Morningness-Eveningness Questionnaire110 is administered at enrolment to determine each participant’s baseline chronotype. Participants maintain a daily sleep diary throughout the study period to allow confirmation of bedtimes. Feasibility of 3 weeks of wrist actigraphy is being assessed.
Peripheral blood biomarkers
Optional blood sampling occurs at both infusion visits and at the follow-up visit approximately 3 weeks after the second infusion. Sampling on infusion visits occurs twice, before propofol administration and approximately 1 hour after infusion start. Following centrifugation, plasma samples are banked. Potential future approaches include proteomic analysis with the Olink Neurology 384 Platform, exploration of inflammatory cytokines (eg, interleukin (IL)-6, IL-8) and markers axonal injury (neurofilament light chain), genotyping of sleep/circadian/neuroinflammation loci (eg, adenosine deaminase or adenosine receptor typing) or transmitter receptors (eg, dopamine, P2X7R). Samples are also processed for storage in PAXGene RNA tubes for future differential gene expression and gene set enrichment analyses. Gene expression data may also be used to assess circadian health.111
Data analysis
EEG preprocessing
Deidentified data from overnight sleep recordings are preprocessed by the Dreem manufacturer and imported into MATLAB analysis platform. Through custom-written scripts for MATLAB (Mathworks, Natick, Massachusetts, USA), raw data are down-sampled to 250 Hz and frequency bandpass filtered at 0.5–50 Hz using EEGLAB,112 an open-source analysis toolbox.
High-density EEG data are preprocessed in EEGLAB.112 Data are low-pass filtered at 40 Hz prior to decimation to 125 Hz. A 0.1–0.6 bandstop filter is applied to remove respiratory artefact prior to high pass filtering at 0.5 Hz.34
Sleep scoring
European data format records are imported into Respironics Sleepware G3 software (V.3.7.1, Philips Respironics, Murrysville, Pennsylvania, USA) and evaluated by American Academy of Sleep Medicine (AASM) certified sleep technologists. Recordings are scored manually in 30 s epochs using modified AASM scoring rules previously developed for single frontal EEG analyses113 and described recently.113 114 The sleep stages are aligned with quantitative EEG analytical measures. N2 and N3 (sleep slow waves present in >20% epoch) epochs are used in quantitation of EEG metrics below. Eye movements in frontal EEG allow scoring of REM sleep epochs. Staff are blinded to clinical/cognitive outcomes during sleep staging and EEG quantitative analyses.
SWA and DSR
We quantify the expression of EEG slow waves during propofol infusions and N2 and N3 sleep. Multitaper methods are used for power spectral analysis using the MATLAB Chronux toolbox.115 Spectral estimates are based on 6-second non-overlapping time windows, time-bandwidth product of 3 and five tapers. Distributions of power in the 20–30 Hz and 1–4.5 Hz bands are used in a semiautomated fashion to reject artifactual epochs.34 SWA is calculated at 1 min intervals as the total EEG 0.5–4 Hz power/minute during propofol treatment (sedation SWA) and during overnight SWS (sleep SWA). Average SWA for each night is quantified during the first and second N2/N3 cycles, using custom-written MATLAB scripts.116 Secondary analyses rely on the DSR for each night of sleep, which is computed by dividing the average SWA of the first cycle by the average of the second cycle.97
Burst suppression
Burst suppression has previously been investigated as a potential marker or mediator of the antidepressant effects of propofol87 and electroconvulsive therapy. Total burst suppression duration for each infusion is quantified from infusion EEG data using a previously described voltage-based algorithm.97 117 118
Blinding
Masking of total drug infused and infusion rates is applied to patients and those performing depression, behavioural and cognitive assessments.
Endpoints
Table 2 lists the primary, secondary and tertiary endpoints. This trial primarily assesses the safety and feasibility of administering intravenous propofol infusions for SWS enhancement. Secondary endpoints include changes in suicidality, cognitive ability and sleep architecture. Tertiary endpoints assess the feasibility of acquiring propofol-associated changes in circadian rhythms, psychomotor vigilance, fluid cognition, affect, anhedonia and other depression symptoms.
Table 2Primary, secondary and tertiary endpoints of the phase I trial
| Endpoints | Outcomes | Measures |
| Primary | Safety | Adverse events (AEs) |
| Induction of EEG slow waves | Slow wave activity (SWA) during propofol infusions | |
| Enhancement of sleep slow waves | Preinfusion versus postinfusion sleep SWA | |
| Secondary | Sleep architecture | N3 and REM duration and percentage of total sleep time |
| Sleep SWA | Delta sleep ratio | |
| Cognitive ability | Montreal Cognitive Assessments | |
| Suicidality | Columbia-Suicide Severity Rating Scale | |
| Tertiary | Depression severity | Montgomery-Åsberg Depression Rating Scale |
| Anhedonia | Snaith-Hamilton Pleasure Scale | |
| Affect | Feeling Scale | |
| Cognitive ability | Psychomotor Vigilance Task NIH Toolbox Cognition Battery | |
| Circadian rhythms | Morningness-Eveningness Questionnaire Sleep diary | |
| Neuroinflammation | Molecular markers (optional) |
EEG, Electroencephalographic; N3, Non-REM Stage 3; REM, rapid eye movement; SWA, Slow wave activity.
Data management and statistical analysis
Data will be stored in REDCap and laboratory servers. The sample size was determined using pilot study data (two pretreatment and two post-treatment sleep recordings from one participant).97 We used a mean N3 duration (minutes) of 1.75 (pretreatment) and 10 (post-treatment) and a combined SD of 4.19. For paired measurements within participants, these estimates would only lead to a sample size of 6. This first participant had a >400% augmentation of her baseline N3 duration, which may be greater compared with others. Furthermore, we expect a 20% attrition such that targeting 15 participants seems compatible with a convenience sample size for gauging safety, feasibility and persistence of postintervention sleep SWA promotion. Descriptive statistics and regression analyses will be performed.
Patient and public involvement
Feedback from the National Institute of Mental Health (NIMH) Data and Monitoring Safety Board-D (DSMB-D) members and study participants guide the execution of this investigation and the planned phase II study. After the conclusion of the trial, the study team will provide participants with a summary of study outcomes.
Data sharing
We will share our data with the National Sleep Research Resource within 3 years after completion of the study. Furthermore, we will share our data on the NIMH Data Archive.
Discussion
The SWIPED study aims to enhance SWS with propofol as a novel approach towards depression treatment in older adults. In contrast to current depression treatments, which largely target monoaminergic neurotransmitter systems, we hope to engage endogenous neural circuitry to restore normal sleep architecture, thereby addressing a core pathophysiological feature of the illness. This is similar to efforts with other novel antidepressants such as the neurosteroid brexanolone,119 which is thought to correct pathophysiology associated with post-partum depression.
Phase I of the SWIPED study will assess the safety and feasibility of using propofol to generate slow waves during infusions and enhance subsequent SWS. Pending the results of this investigation, a future phase II clinical trial would assess the association of SWS enhancement with cognitive and clinical outcomes. Phase I will progress to a phase II study if: (1) less than 5% of serious adverse events are directly attributable to propofol infusions, (2) EEG slow waves are present for the majority of the infusion time in at least 60% of participants and (3) total sleep SWA is augmented in at least 40% of participants who complete the study. Phase II will entail a randomised placebo-controlled trial of 70 participants. We anticipate that this approach will further elucidate the relationships between depression, cognitive dysfunction and sleep disturbances and yield new avenues for treating TRD.
Key limitations of this study include its small sample size (n=15) and lack of control groups. We will not be able to compare propofol treatment to (1) no treatment or (2) other methods of SWS enhancement (eg, non-pharmacological enhancement). An intervention that strictly enhances SWS without potential independent mechanisms of antidepressant effect would more incisively address our hypothesis regarding the effect of SWS enhancement on depression. In addition, the findings may be challenging to generalise, given the advanced age of the study participants and the numerous exclusion criteria employed.
Ethics and dissemination
The study design and procedures are guided and approved by the NIMH DSMB-D, which convenes thrice per year. A board-certified anaesthesiologist with no professional conflicts of interest is serving as a clinical site monitor. Their recommendations for improving study conduct have been integrated. The study has been approved by the Washington University Human Research Protection Office (WU HRPO). Any changes to the study protocol are formally approved by both the NIMH DSMB-D and WU HRPO prior to implementation. Full informed consent is obtained from all enrolled participants by trained research staff (online supplemental file 1). This document has been drafted to comply with the Standard Protocol Items: Recommendations for Interventional Trials checklist (online supplemental file 2).
The findings of this study will be disseminated via peer-reviewed publications, presentations at scientific conferences and mass media. In addition, data will be stored in the NIMH data archive, which shares deidentified data with qualified researchers and provides summary data to the public. EEG data will be shared via the National Sleep Research Resource within 3 years of study completion.
We appreciate the ongoing assistance and feedback provided by study participants and nursing staff in the WU Clinical Trials Research Unit. Contributions from Alec Hester were appreciated. We thank Sherri McKinnon, Sara Fisher, C. Ryan King, Ashley Kennedy and the NIMH DSMB-D for regulatory oversight.
Ethics statements
Patient consent for publication
Not applicable.
Collaborators SWIPED Study Team: Thomas Nguyen, Elliott Kraenzle, Matthew Robeck, Aris Perez, Jennifer Wulfers, Robby Greenspan, Hilary Simon, Julia Schweiger, Adam Koenig, Olivia Foster, Alyssa K Labonte, Orlandrea Hyche, Allyson Quigley, Tiffany Janssen-Scott, Sriram Zassenhaus, Sofia Salvatore and Jay Patel.
Contributors RLR: Drafting of manuscript and review of final manuscript. MG: Revision and review of final manuscript. MK: Revision and review of final manuscript. SC: Conceptualisation, funding and review of final manuscript. NBF: Conceptualisation, funding and review of final manuscript. NL: Conceptualisation and review of final manuscript. BPL: Conceptualisation, funding and review of final manuscript. CFR: Conceptualisation, funding and review of final manuscript. EL: Conceptualisation, funding and review of final manuscript. BJAP: Conceptualisation, funding, drafting of manuscript and review of final manuscript. SWIPED Study Team: Conceptualisation, drafting of manuscript and review of final manuscript.
Funding This work was supported by the Washington University Center for Perioperative Mental Health grant number P50MH122351 from the National Institute of Mental Health (NIMH) of the National Institutes of Health (NIH), NIMH grant U01MH128483, NIMH grant K01MH128663 and NIH grant UL1TR002345.
Disclaimer The content is solely the responsibility of the authors and does not necessarily represent the official view of the NIH.
Competing interests BJAP has a patent on control of anaesthetic state modulation and an agreement with Elemind on the use of non-pharmacological potentiation of EEG slow waves. EL reports consulting fees from Merck, Boehringer-Ingelheim, Pritikin ICR, IngenioRx and Prodeo, grant funding from Janssen, the COVID Early Treatment Fund and FastGrants, and a patent pending on sigma-1 receptor agonists for COVID-19. BPL reports consulting fees from Eisai, Eli Lilly, OrbiMed and the GLG Group. He has also received research funding from Eisai and serves on the Scientific Advisory Board for Beacon Biosignals and a Data Safety Monitoring Board for Eli Lilly. RLR and MK have no conflicts to disclose concerning any product mentioned or concept discussed in this article. MK, BPL, EL and BJAP have received funding from the National Institutes of Health.
Patient and public involvement Patients and/or the public were involved in the design, or conduct, or reporting, or dissemination plans of this research. Refer to the Methods section for further details.
Provenance and peer review Not commissioned; peer reviewed for ethical and funding approval prior to submission.
Supplemental material This content has been supplied by the author(s). It has not been vetted by BMJ Publishing Group Limited (BMJ) and may not have been peer-reviewed. Any opinions or recommendations discussed are solely those of the author(s) and are not endorsed by BMJ. BMJ disclaims all liability and responsibility arising from any reliance placed on the content. Where the content includes any translated material, BMJ does not warrant the accuracy and reliability of the translations (including but not limited to local regulations, clinical guidelines, terminology, drug names and drug dosages), and is not responsible for any error and/or omissions arising from translation and adaptation or otherwise.
1 Rajan S, McKee M, Rangarajan S, et al. Association of symptoms of depression with cardiovascular disease and mortality in Low-, middle-, and high-income countries. JAMA Psychiatry 2020; 77: 1052–63. doi:10.1001/jamapsychiatry.2020.1351
2 Zhdanava M, Pilon D, Ghelerter I, et al. The prevalence and national burden of treatment-resistant depression and major depressive disorder in the United States. J Clin Psychiatry 2021; 82: 20m13699. doi:10.4088/JCP.20m13699
3 Lenze EJ, Rogers JC, Martire LM, et al. The Association of late-life depression and anxiety with physical disability: a review of the literature and prospectus for future research. Am J Geriatr Psychiatry 2001; 9: 113–35.
4 Wolkowitz OM, Reus VI, Mellon SH. Of sound mind and body: depression, disease, and accelerated aging. Dialogues Clin Neurosci 2011; 13: 25–39. doi:10.31887/DCNS.2011.13.1/owolkowitz
5 Szanto K, Lenze EJ, Waern M, et al. Research to reduce the suicide rate among older adults: methodology roadblocks and promising paradigms. Psychiatr Serv 2013; 64: 586–9. doi:10.1176/appi.ps.003582012
6 Ownby RL, Crocco E, Acevedo A, et al. Depression and risk for Alzheimer disease: systematic review, meta-analysis, and Metaregression analysis. Arch Gen Psychiatry 2006; 63: 530–8. doi:10.1001/archpsyc.63.5.530
7 Byers AL, Yaffe K. Depression and risk of developing dementia. Nat Rev Neurol 2011; 7: 323–31. doi:10.1038/nrneurol.2011.60
8 Diniz BS, Butters MA, Albert SM, et al. Late-life depression and risk of vascular dementia and Alzheimer’s disease: systematic review and meta-analysis of community-based cohort studies. Br J Psychiatry 2013; 202: 329–35. doi:10.1192/bjp.bp.112.118307
9 Thomas L, Mulsant BH, Solano FX, et al. Response speed and rate of remission in primary and specialty care of elderly patients with depression. Am J Geriatr Psychiatry 2002; 10: 583–91.
10 Mulsant BH, Blumberger DM, Ismail Z, et al. A systematic approach to pharmacotherapy for geriatric major depression. Clin Geriatr Med 2014; 30: 517–34. doi:10.1016/j.cger.2014.05.002
11 Lenze EJ, Mulsant BH, Roose SP, et al. Antidepressant augmentation versus switch in treatment-resistant geriatric depression. N Engl J Med 2023; 388: 1067–79. doi:10.1056/NEJMoa2204462
12 Deng Y, McQuoid DR, Potter GG, et al. Predictors of recurrence in remitted late-life depression. Depress Anxiety 2018; 35: 658–67. doi:10.1002/da.22772
13 Rush AJ, Warden D, Wisniewski SR, et al. STAR*D: revising conventional wisdom. CNS Drugs 2009; 23: 627–47. doi:10.2165/00023210-200923080-00001
14 Gabriel FC, de Melo DO, Fráguas R, et al. Pharmacological treatment of depression: a systematic review comparing clinical practice guideline recommendations. PLOS ONE 2020; 15: e0231700. doi:10.1371/journal.pone.0231700
15 Allard P, Gram L, Timdahl K, et al. Efficacy and tolerability of venlafaxine in geriatric outpatients with major depression: a double-blind, randomised 6-month comparative trial with citalopram. Int J Geriatr Psychiatry 2004; 19: 1123–30. doi:10.1002/gps.1190
16 Raskin J, Wiltse CG, Siegal A, et al. Efficacy of duloxetine on cognition, depression, and pain in elderly patients with major depressive disorder: an 8-week, double-blind, placebo-controlled trial. Am J Psychiatry 2007; 164: 900–9. doi:10.1176/ajp.2007.164.6.900
17 Schatzberg A, Roose S. A double-blind, placebo-controlled study of venlafaxine and fluoxetine in geriatric outpatients with major depression. Am J Geriatr Psychiatry 2006; 14: 361–70. doi:10.1097/01.JGP.0000194645.70869.3b
18 Kellner CH, Husain MM, Knapp RG, et al. Right unilateral Ultrabrief pulse ECT in geriatric depression: phase 1 of the PRIDE study. AJP 2016; 173: 1101–9. doi:10.1176/appi.ajp.2016.15081101
19 Kellner CH, Husain MM, Knapp RG, et al. A novel strategy for continuation ECT in geriatric depression: phase 2 of the PRIDE study. AJP 2016; 173: 1110–8. doi:10.1176/appi.ajp.2016.16010118
20 Conelea CA, Philip NS, Yip AG, et al. Transcranial magnetic stimulation for treatment-resistant depression: naturalistic treatment outcomes for younger versus older patients. J Affect Disord 2017; 217: 42–7. doi:10.1016/j.jad.2017.03.063
21 Murphy MJ, Peterson MJ. Sleep disturbances in depression. Sleep Med Clin 2015; 10: 17–23. doi:10.1016/j.jsmc.2014.11.009
22 Schröder CM, O’Hara R. Depression and obstructive sleep apnea (OSA). Ann Gen Psychiatry 2005; 4: 13. doi:10.1186/1744-859X-4-13
23 Holanda Júnior FWN, Almondes KM de. Sleep and executive functions in older adults: a systematic review. Dement Neuropsychol 2016; 10: 185–97. doi:10.1590/S1980-5764-2016DN1003004
24 Berry R, Gamaldo CE, Harding SM, et al. The AASM Manual for Scoring of Sleep and Associated Events: Rules, Terminology and Technical Specifications, Version 2.3. American Academy of Sleep Medicine, 2015.
25 Tasali E, Leproult R, Ehrmann DA, et al. Slow-wave sleep and the risk of type 2 diabetes in humans. Proc Natl Acad Sci U S A 2008; 105: 1044–9. doi:10.1073/pnas.0706446105
26 Mölle M, Yeshenko O, Marshall L, et al. Hippocampal sharp wave-ripples linked to slow oscillations in rat slow-wave sleep. J Neurophysiol 2006; 96: 62–70. doi:10.1152/jn.00014.2006
27 Rasch B, Büchel C, Gais S, et al. Odor cues during slow-wave sleep prompt declarative memory consolidation. Science 2007; 315: 1426–9. doi:10.1126/science.1138581
28 Marshall L, Mölle M, Hallschmid M, et al. Transcranial direct current stimulation during sleep improves declarative memory. J Neurosci 2004; 24: 9985–92. doi:10.1523/JNEUROSCI.2725-04.2004
29 Marshall L, Helgadóttir H, Mölle M, et al. Boosting slow oscillations during sleep potentiates memory. Nature 2006; 444: 610–3. doi:10.1038/nature05278
30 Iliff JJ, Wang M, Liao Y, et al. A Paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci Transl Med 2012; 4: 147ra111. doi:10.1126/scitranslmed.3003748
31 Fultz NE, Bonmassar G, Setsompop K, et al. Coupled electrophysiological, hemodynamic, and cerebrospinal fluid oscillations in human sleep. Science 2019; 366: 628–31. doi:10.1126/science.aax5440
32 Peng W, Achariyar TM, Li B, et al. Suppression of glymphatic fluid transport in a mouse model of Alzheimer’s disease. Neurobiol Dis 2016; 93: 215–25. doi:10.1016/j.nbd.2016.05.015
33 Iliff JJ, Chen MJ, Plog BA, et al. Impairment of glymphatic pathway function promotes Tau pathology after traumatic brain injury. J Neurosci 2014; 34: 16180–93. doi:10.1523/JNEUROSCI.3020-14.2014
34 Lucey BP, McCullough A, Landsness EC, et al. Reduced non-rapid eye movement sleep is associated with Tau pathology in early Alzheimer’s disease. Sci Transl Med 2019; 11: eaau6550. doi:10.1126/scitranslmed.aau6550
35 Buysse DJ, Frank E, Lowe KK, et al. Electroencephalographic sleep correlates of episode and vulnerability to recurrence in depression. Biol Psychiatry 1997; 41: 406–18. doi:10.1016/S0006-3223(96)00041-8
36 Zeoli I, Lanquart J-P, Wacquier B, et al. Polysomnographic markers of suicidal ideation in untreated unipolar major depressed individuals. Int J Psychophysiol 2021; 166: 19–24. doi:10.1016/j.ijpsycho.2021.05.001
37 Kupfer DJ, Reynolds CF, Weiss BL. Lithium carbonate and sleep in affective disorders. Arch Gen Psychiatry 1974; 30: 79. doi:10.1001/archpsyc.1974.01760070061009
38 Billiard M. Lithium carbonate: effects on sleep patterns of normal and depressed subjects and its use in sleep-wake pathology. Pharmacopsychiatry 1987; 20: 195–6. doi:10.1055/s-2007-1017102
39 Friston KJ, Sharpley AL, Solomon RA, et al. Lithium increases slow wave sleep: possible mediation by brain 5-Ht2 receptors Psychopharmacology (Berl) 1989; 98: 139–40. doi:10.1007/BF00442020
40 Montgomery I, Oswald I, Morgan K, et al. Trazodone enhances sleep in subjective quality but not in objective duration. Br J Clin Pharmacol 1983; 16: 139–44. doi:10.1111/j.1365-2125.1983.tb04977.x
41 Doghramji K, Jangro WC. Adverse effects of psychotropic medications on sleep. Psychiatr Clin North Am 2016; 39: 487–502. doi:10.1016/j.psc.2016.04.009
42 Jindal RD, Friedman ES, Berman SR, et al. Effects of sertraline on sleep architecture in patients with depression. J Clin Psychopharmacol 2003; 23: 540–8. doi:10.1097/01.jcp.0000095345.32154.9a
43 Ehlers CL, Havstad JW, Kupfer DJ. Estimation of the time course of slow-wave sleep over the night in depressed patients: effects of Clomipramine and clinical response. Biol Psychiatry 1996; 39: 171–81. doi:10.1016/0006-3223(95)00139-5
44 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: 301–11. doi:10.1017/S1461145712000545
45 Quera Salva M-A, Vanier B, Laredo J, et al. Major depressive disorder, sleep EEG and agomelatine: an open-label study. Int J Neuropsychopharmacol 2007; 10: 691–6. doi:10.1017/S1461145707007754
46 Esser SK, Hill SL, Tononi G. Sleep homeostasis and cortical synchronization: I. Modeling the effects of synaptic strength on sleep slow waves. Sleep 2007; 30: 1617–30. doi:10.1093/sleep/30.12.1617
47 Riedner BA, Vyazovskiy VV, Huber R, et al. Sleep homeostasis and cortical synchronization: III. A high-density EEG study of sleep slow waves in humans. Sleep 2007; 30: 1643–57. doi:10.1093/sleep/30.12.1643
48 Vyazovskiy VV, Riedner BA, Cirelli C, et al. Sleep homeostasis and cortical synchronization: II. A local field potential study of sleep slow waves in the rat. Sleep 2007; 30: 1631–42. doi:10.1093/sleep/30.12.1631
49 Achermann P, Dijk DJ, Brunner DP, et al. A model of human sleep homeostasis based on EEG slow-wave activity: quantitative comparison of data and Simulations. Brain Res Bull 1993; 31: 97–113. doi:10.1016/0361-9230(93)90016-5
50 Ganguli R, Reynolds CF, Kupfer DJ. Electroencephalographic sleep in young, never-medicated schizophrenics. A comparison with delusional and nondelusional depressives and with healthy controls. Arch Gen Psychiatry 1987; 44: 36–44. doi:10.1001/archpsyc.1987.01800130038006
51 Kupfer DJ, Ulrich RF, Coble PA, et al. Application of automated REM and slow wave sleep analysis: II. testing the assumptions of the two-process model of sleep regulation in normal and depressed subjects. Psychiatry Res 1984; 13: 335–43. doi:10.1016/0165-1781(84)90081-7
52 Reynolds CF, Kupfer DJ. Sleep research in affective illness: state of the art circa 1987. Sleep 1987; 10: 199–215. doi:10.1093/sleep/10.3.199
53 Gillin JC, Duncan W, Pettigrew KD, et al. Successful separation of depressed, normal, and Insomniac subjects by EEG sleep data. Arch Gen Psychiatry 1979; 36: 85–90. doi:10.1001/archpsyc.1979.01780010091010
54 Benca RM, Obermeyer WH, Thisted RA, et al. Sleep and psychiatric disorders. A meta-analysis [ discussion 669-670 ]. Arch Gen Psychiatry 1992; 49: 651–68;. doi:10.1001/archpsyc.1992.01820080059010
55 Gillin JC, Duncan WC, Murphy DL, et al. Age-related changes in sleep in depressed and normal subjects. Psychiatry Research 1981; 4: 73–8. doi:10.1016/0165-1781(81)90010-X
56 Kupfer DJ, Frank E, McEachran AB, et al. Delta sleep ratio. A biological correlate of early recurrence in unipolar affective disorder. Arch Gen Psychiatry 1990; 47: 1100–5. doi:10.1001/archpsyc.1990.01810240020004
57 Duncan WC, Selter J, Brutsche N, et al. Baseline delta sleep ratio predicts acute ketamine mood response in major depressive disorder. J Affect Disord 2013; 145: 115–9. doi:10.1016/j.jad.2012.05.042
58 Cristancho P, Lenze EJ, Dixon D, et al. Executive function predicts antidepressant treatment noncompletion in late-life depression. J Clin Psychiatry 2018; 79: 16m11371. doi:10.4088/JCP.16m11371
59 Snyder HR. Major depressive disorder is associated with broad impairments on neuropsychological measures of executive function: a meta-analysis and review. Psychol Bull 2013; 139: 81–132. doi:10.1037/a0028727
60 Diamond A. Executive functions. Annu Rev Psychol 2013; 64: 135–68. doi:10.1146/annurev-psych-113011-143750
61 Burdick DJ, Rosenblatt A, Samus QM, et al. Predictors of functional impairment in residents of assisted-living facilities: the Maryland assisted living study. J Gerontol A Biol Sci Med Sci 2005; 60: 258–64. doi:10.1093/gerona/60.2.258
62 Galvin JE, Roe CM, Coats MA, et al. Patient’s rating of cognitive ability: using the Ad8, a brief informant interview, as a self-rating tool to detect dementia. Arch Neurol 2007; 64: 725–30. doi:10.1001/archneur.64.5.725
63 Kaneriya SH, Robbins-Welty GA, Smagula SF, et al. Predictors and Moderators of remission with aripiprazole augmentation in treatment-resistant late-life depression: an analysis of the IRL-grey randomized clinical trial. JAMA Psychiatry 2016; 73: 329–36. doi:10.1001/jamapsychiatry.2015.3447
64 Löwe B, Unützer J, Callahan CM, et al. Monitoring depression treatment outcomes with the patient health questionnaire-9. Med Care 2004; 42: 1194–201. doi:10.1097/00005650-200412000-00006
65 Weintraub S, Dikmen SS, Heaton RK, et al. Cognition assessment using the NIH toolbox. Neurology 2013; 80: S54–64. doi:10.1212/WNL.0b013e3182872ded
66 Plante DT, Hagen EW, Ravelo LA, et al. Impaired neurobehavioral alertness quantified by the psychomotor vigilance task is associated with depression in the Wisconsin sleep cohort study. Sleep Med 2020; 67: 66–70. doi:10.1016/j.sleep.2019.11.1248
67 Yun C-H, Kim H, Lee SK, et al. Daytime sleepiness associated with poor sustained attention in middle and late adulthood. Sleep Med 2015; 16: 143–51. doi:10.1016/j.sleep.2014.07.028
68 Plante DT. The evolving nexus of sleep and depression. Am J Psychiatry 2021; 178: 896–902. doi:10.1176/appi.ajp.2021.21080821
69 Purdon PL, Sampson A, Pavone KJ, et al. Clinical electroencephalography for anesthesiologists: part I: background and basic signatures. Anesthesiology 2015; 123: 937–60. doi:10.1097/ALN.0000000000000841
70 Murphy M, Bruno M-A, Riedner BA, et al. Propofol anesthesia and sleep: a high-density EEG study. Sleep 2011; 34: 283–91A. doi:10.1093/sleep/34.3.283
71 Mhuircheartaigh RN, Rosenorn-Lanng D, Wise R, et al. Cortical and subcortical connectivity changes during decreasing levels of consciousness in humans: a functional magnetic resonance imaging study using propofol. J Neurosci 2010; 30: 9095–102. doi:10.1523/JNEUROSCI.5516-09.2010
72 Ní Mhuircheartaigh R, Warnaby C, Rogers R, et al. Slow-wave activity saturation and thalamocortical isolation during propofol anesthesia in humans. Sci Transl Med 2013; 5: 208ra148. doi:10.1126/scitranslmed.3006007
73 Jones KG, Lybbert C, Euler MJ, et al. Diversity of electroencephalographic patterns during propofol-induced burst suppression. Front Syst Neurosci 2023; 17: 1172856. doi:10.3389/fnsys.2023.1172856
74 Ching S, Purdon PL, Vijayan S, et al. A neurophysiological-metabolic model for burst suppression. Proc Natl Acad Sci U S A 2012; 109: 3095–100. doi:10.1073/pnas.1121461109
75 Kenny JD, Westover MB, Ching S, et al. Propofol and sevoflurane induce distinct burst suppression patterns in rats. Front Syst Neurosci 2014; 8: 237. doi:10.3389/fnsys.2014.00237
76 Tung A, Bergmann BM, Herrera S, et al. Recovery from sleep deprivation occurs during propofol anesthesia. Anesthesiology 2004; 100: 1419–26. doi:10.1097/00000542-200406000-00014
77 Rabelo FAW, Braga A, Küpper DS, et al. Propofol-induced sleep: polysomnographic evaluation of patients with obstructive sleep apnea and controls. Otolaryngol Head Neck Surg 2010; 142: 218–24. doi:10.1016/j.otohns.2009.11.002
78 Xu Z, Jiang X, Li W, et al. Propofol-induced sleep: efficacy and safety in patients with refractory chronic primary insomnia. Cell Biochem Biophys 2011; 60: 161–6. doi:10.1007/s12013-010-9135-7
79 O’Toole DP, Milligan KR, Howe JP, et al. A comparison of propofol and methohexitone as induction agents for day case isoflurane anaesthesia. Anaesthesia 1987; 42: 373–6. doi:10.1111/j.1365-2044.1987.tb03977.x
80 McDonald NJ, Mannion D, Lee P, et al. Mood evaluation and outpatient anaesthesia. A comparison between propofol and thiopentone. Anaesthesia 1988; 43 Suppl: 68–9. doi:10.1111/j.1365-2044.1988.tb09075.x
81 Canaday BR. Amorous, disinhibited behavior associated with propofol. Clin Pharm 1993; 12: 449–51.
82 Kent EA, Bacon DR, Harrison P, et al. Sexual illusions and propofol sedation. Anesthesiology 1992; 77: 1037–8. doi:10.1097/00000542-199211000-00030
83 Kim JH, Byun H, Kim JH. Abuse potential of propofol used for sedation in gastric endoscopy and its correlation with subject characteristics. Korean J Anesthesiol 2013; 65: 403–9. doi:10.4097/kjae.2013.65.5.403
84 D’Haese J, Camu F, Dekeyzer PJ, et al. Propofol and methohexitone anaesthesia: effects on the profile of mood state. Eur J Anaesthesiol 1994; 11: 359–63.
85 Tadler SC, Jones KG, Lybbert C, et al. Propofol for treatment resistant depression: a randomized controlled trial. medRxiv 2023.: 2023.09.12.23294678. doi:10.1101/2023.09.12.23294678
86 Whitehead C, Sanders LD, Oldroyd G, et al. The subjective effects of low-dose propofol. A double-blind study to evaluate dimensions of sedation and consciousness with low-dose propofol. Anaesthesia 1994; 49: 490–6. doi:10.1111/j.1365-2044.1994.tb03518.x
87 Mickey BJ, White AT, Arp AM, et al. Propofol for treatment-resistant depression: a pilot study. Int J Neuropsychopharmacol 2018; 21: 1079–89. doi:10.1093/ijnp/pyy085
88 Wang X, Yang Y, Zhou X, et al. Propofol pretreatment increases antidepressant-like effects induced by acute administration of ketamine in rats receiving forced swimming test. Psychiatry Res 2011; 185: 248–53. doi:10.1016/j.psychres.2010.04.046
89 Luo J, Min S, Wei K, et al. Propofol interacts with stimulus intensities of electroconvulsive shock to regulate behavior and hippocampal BDNF in a rat model of depression. Psychiatry Res 2012; 198: 300–6. doi:10.1016/j.psychres.2011.09.010
90 Peduto VA, Concas A, Santoro G, et al. Biochemical and electrophysiologic evidence that propofol enhances GABAergic transmission in the rat brain. Anesthesiology 1991; 75: 1000–9. doi:10.1097/00000542-199112000-00012
91 Irifune M, Takarada T, Shimizu Y, et al. Propofol-induced anesthesia in mice is mediated by gamma-aminobutyric acid-A and excitatory amino acid receptors. Anesth Analg 2003; 97: 424–9. doi:10.1213/01.ANE.0000059742.62646.40
92 Orser BA, Bertlik M, Wang LY, et al. Inhibition by propofol (2,6 Di-Isopropylphenol) of the N-methyl-D-aspartate subtype of glutamate receptor in cultured hippocampal Neurones. Br J Pharmacol 1995; 116: 1761–8. doi:10.1111/j.1476-5381.1995.tb16660.x
93 Mao L-M, Hastings JM, Fibuch EE, et al. Propofol selectively alters Glua1 AMPA receptor phosphorylation in the hippocampus but not prefrontal cortex in young and aged mice. Eur J Pharmacol 2014; 738: 237–44. doi:10.1016/j.ejphar.2014.05.053
94 Krampfl K, Cordes AL, Schlesinger F, et al. Effects of propofol on recombinant AMPA receptor channels. Eur J Pharmacol 2005; 511: 1–7. doi:10.1016/j.ejphar.2005.02.012
95 Zhu X-N, Li J, Qiu G-L, et al. Propofol exerts anti-anhedonia effects via inhibiting the dopamine transporter. Neuron 2023; 111: 1626–36. doi:10.1016/j.neuron.2023.02.017
96 Oishi Y, Xu Q, Wang L, et al. Slow-wave sleep is controlled by a subset of nucleus accumbens core neurons in mice. Nat Commun 2017; 8: 734. doi:10.1038/s41467-017-00781-4
97 Rios RL, Kafashan M, Hyche O, et al. Targeting slow wave sleep deficiency in late-life depression: a case series with propofol. Am J Geriatr Psychiatry 2023; 31: 643–52. doi:10.1016/j.jagp.2023.03.009
98 Kroenke K, Spitzer RL, Williams JB. The PHQ-9: validity of a brief depression severity measure. J Gen Intern Med 2001; 16: 606–13. doi:10.1046/j.1525-1497.2001.016009606.x
99 Eleveld DJ, Colin P, Absalom AR, et al. Pharmacokinetic-pharmacodynamic model for propofol for broad application in anaesthesia and sedation. Br J Anaesth 2018; 120: 942–59. doi:10.1016/j.bja.2018.01.018
100 Aldrete JA. Modifications to the postanesthesia score for use in ambulatory surgery. J Perianesth Nurs 1998; 13: 148–55. doi:10.1016/s1089-9472(98)80044-0
101 Kafashan MM, Hyche O, Nguyen T, et al. Perioperative sleep in geriatric cardiac surgical patients: a feasibility study using a wireless wearable device. Br J Anaesth 2021; 126: e205–8. doi:10.1016/j.bja.2021.02.018
102 Montgomery SA, Asberg M. A new depression scale designed to be sensitive to change. Br J Psychiatry 1979; 134: 382–9. doi:10.1192/bjp.134.4.382
103 Posner K, Brown GK, Stanley B, et al. The Columbia-suicide severity rating scale: initial validity and internal consistency findings from three multisite studies with adolescents and adults. Am J Psychiatry 2011; 168: 1266–77. doi:10.1176/appi.ajp.2011.10111704
104 Snaith RP, Hamilton M, Morley S, et al. A scale for the assessment of Hedonic tone the Snaith-Hamilton pleasure scale. Br J Psychiatry 1995; 167: 99–103. doi:10.1192/bjp.167.1.99
105 Hardy CJ, Rejeski WJ. Not what, but how one feels: the measurement of affect during exercise. J Sport Exerc Psychol 1989; 11: 304–17. doi:10.1123/jsep.11.3.304
106 Nasreddine ZS, Phillips NA, Bédirian V, et al. The Montreal Cognitive Assessment, MoCA: a brief screening tool for mild cognitive impairment. J Am Geriatr Soc 2005; 53: 695–9. doi:10.1111/j.1532-5415.2005.53221.x
107 Dinges DF, Powell JW. Microcomputer analyses of performance on a portable simple visual RT task during sustained operations. Beh Res Meth Instr Comp 1985; 17: 652–5. doi:10.3758/BF03200977
108 Arsintescu L, Kato KH, Cravalho PF, et al. Validation of a touchscreen psychomotor vigilance task. Accid Anal Prev 2019; 126: 173–6. doi:10.1016/j.aap.2017.11.041
109 Doran SM, Van Dongen HP, Dinges DF. Sustained attention performance during sleep deprivation: evidence of state instability. Arch Ital Biol 2001; 139: 253–67.
110 Horne JA, Ostberg O. A self-assessment questionnaire to determine morningness-eveningness in human circadian rhythms. Int J Chronobiol 1976; 4: 97–110.
111 Braun R, Kath WL, Iwanaszko M, et al. Universal method for robust detection of circadian state from gene expression. Proc Natl Acad Sci U S A 2018; 115: E9247–56. doi:10.1073/pnas.1800314115
112 Delorme A, Makeig S. EEGLAB: an open source Toolbox for analysis of single-trial EEG Dynamics including independent component analysis. J Neurosci Methods 2004; 134: 9–21. doi:10.1016/j.jneumeth.2003.10.009
113 Lucey BP, Mcleland JS, Toedebusch CD, et al. Comparison of a single-channel EEG sleep study to Polysomnography. J Sleep Res 2016; 25: 625–35. doi:10.1111/jsr.12417
114 Smith SK, Nguyen T, Labonte AK, et al. Protocol for the prognosticating delirium recovery outcomes using wakefulness and sleep electroencephalography (P-DROWS-E) study: a prospective observational study of delirium in elderly cardiac surgical patients. BMJ Open 2020; 10: e044295. doi:10.1136/bmjopen-2020-044295
115 Mitra PP, Bokil H. Observed Brain Dynamics. 1st edn. 381. Oxford University Press, 2008.
116 Latta F, Leproult R, Tasali E, et al. Sex differences in Delta and alpha EEG activities in healthy older adults. Sleep 2005; 28: 1525–34. doi:10.1093/sleep/28.12.1525
117 Hickman LB, Hogan RE, Labonte AK, et al. Voltage-based automated detection of postictal generalized electroencephalographic suppression: algorithm development and validation. Clin Neurophysiol 2020; 131: 2817–25. doi:10.1016/j.clinph.2020.08.015
118 Kafashan M, Brian Hickman L, Labonte AK, et al. Quiescence during burst suppression and postictal generalized EEG suppression are distinct patterns of activity. Clin Neurophysiol 2022; 142: 125–32. doi:10.1016/j.clinph.2022.07.493
119 Lüscher B, Möhler H. Brexanolone, a Neurosteroid antidepressant, Vindicates the GABAergic deficit hypothesis of depression and may foster resilience. F1000Res 2019; 8: F1000 Faculty Rev-751. doi:10.12688/f1000research.18758.1
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
© 2024 Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ. http://creativecommons.org/licenses/by-nc/4.0/ This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/ . Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
Introduction
Late-life treatment-resistant depression (LL-TRD) is common and increases risk for accelerated ageing and cognitive decline. Impaired sleep is common in LL-TRD and is a risk factor for cognitive decline. Slow wave sleep (SWS) has been implicated in key processes including synaptic plasticity and memory. A deficiency in SWS may be a core component of depression pathophysiology. The anaesthetic propofol can induce electroencephalographic (EEG) slow waves that resemble SWS. Propofol may enhance SWS and oral antidepressant therapy, but relationships are unclear. We hypothesise that propofol infusions will enhance SWS and improve depression in older adults with LL-TRD. This hypothesis has been supported by a recent small case series.
Methods and analysis
SWIPED (Slow Wave Induction by Propofol to Eliminate Depression) phase I is an ongoing open-label, single-arm trial that assesses the safety and feasibility of using propofol to enhance SWS in older adults with LL-TRD. The study is enrolling 15 English-speaking adults over age 60 with LL-TRD. Participants will receive two propofol infusions 2–6 days apart. Propofol infusions are individually titrated to maximise the expression of EEG slow waves. Preinfusion and postinfusion sleep architecture are evaluated through at-home overnight EEG recordings acquired using a wireless headband equipped with dry electrodes. Sleep EEG recordings are scored manually. Key EEG measures include sleep slow wave activity, SWS duration and delta sleep ratio. Longitudinal changes in depression, suicidality and anhedonia are assessed. Assessments are performed prior to the first infusion and up to 10 weeks after the second infusion. Cognitive ability is assessed at enrolment and approximately 3 weeks after the second infusion.
Ethics and dissemination
The study was approved by the Washington University Human Research Protection Office. Recruitment began in November 2022. Dissemination plans include presentations at scientific conferences, peer-reviewed publications and mass media. Positive results will lead to a larger phase II randomised placebo-controlled trial.
Trial registration number
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Details
; Kafashan, MohammadMehdi 2 ; ShiNung Ching 3 ; Farber, Nuri B 4 ; Lin, Nan 5 ; Lucey, Brendan P 6 ; Reynolds, Charles F 7 ; Lenze, Eric J 8 ; Ben Julian Agustin Palanca 9
1 Department of Anesthesiology, Washington University School of Medicine in St. Louis, St Louis, Missouri, USA
2 Department of Anesthesiology, Washington University School of Medicine in St. Louis, St Louis, Missouri, USA; Center on Biological Rhythms and Sleep, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
3 Department of Electrical & Systems Engineering, Washington University in St. Louis, St Louis, Missouri, USA
4 Department of Psychiatry, Washington University School of Medicine in St. Louis, St Louis, Missouri, USA
5 Department of Biostatistics and Data Science, Washington University in St Louis, St Louis, Missouri, USA
6 Center on Biological Rhythms and Sleep, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA; Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
7 Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
8 Department of Anesthesiology, Washington University School of Medicine in St. Louis, St Louis, Missouri, USA; Department of Psychiatry, Washington University School of Medicine in St. Louis, St Louis, Missouri, USA
9 Department of Anesthesiology, Washington University School of Medicine in St. Louis, St Louis, Missouri, USA; Center on Biological Rhythms and Sleep, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA; Department of Psychiatry, Washington University School of Medicine in St. Louis, St Louis, Missouri, USA; Division of Biology and Biomedical Sciences, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri, USA




