Polysomnographic assessment of suvorexant in patients with probable Alzheimer's disease dementia and insomnia: a randomized trial

W Joseph Herring, Paulette Ceesay, Ellen Snyder, Donald Bliwise, Kerry Budd, Jill Hutzelmann, Joanne Stevens, Christopher Lines, David Michelson, W Joseph Herring, Paulette Ceesay, Ellen Snyder, Donald Bliwise, Kerry Budd, Jill Hutzelmann, Joanne Stevens, Christopher Lines, David Michelson

Abstract

Introduction: We evaluated the clinical profile of the orexin receptor antagonist suvorexant for treating insomnia in patients with mild-to-moderate probable Alzheimer's disease (AD) dementia.

Methods: Randomized, double-blind, 4-week trial of suvorexant 10 mg (could be increased to 20 mg based on clinical response) or placebo in patients who met clinical diagnostic criteria for both probable AD dementia and insomnia. Sleep was assessed by overnight polysomnography in a sleep laboratory. The primary endpoint was change-from-baseline in polysomnography-derived total sleep time (TST) at week 4.

Results: Of 285 participants randomized (suvorexant, N = 142; placebo, N = 143), 277 (97%) completed the trial (suvorexant, N = 136; placebo, N = 141). At week 4, the model-based least squares mean improvement-from-baseline in TST was 73 minutes for suvorexant and 45 minutes for placebo; (difference = 28 minutes [95% confidence interval 11-45], p < 0.01). Somnolence was reported in 4.2% of suvorexant-treated patients and 1.4% of placebo-treated patients.

Discussion: Suvorexant improved TST in patients with probable AD dementia and insomnia.

Trial registration: ClinicalTrials.gov NCT02750306.

Keywords: Alzheimer's disease; insomnia; randomized clinical trial; suvorexant.

Conflict of interest statement

W.J.H., P.C., E.S., K.B., J.H., J.S., C.L., and D.M. are current or former employees of Merck Sharp & Dohme Corp. (MSD), a subsidiary of Merck & Co., Inc., Kenilworth, NJ, USA, and they own or owned stock options in Merck & Co., Inc., Kenilworth, NJ, USA. D.B. has acted as a consultant for Merck & Co., Inc. (Kenilworth, NJ USA), Jazz, Eisai, and Ferring.

© 2020 The Authors. Alzheimer's & Dementia published by Wiley Periodicals, Inc. on behalf of Alzheimer's Association.

Figures

Figure 1
Figure 1
Trial design. Polysomnography (PSG) was recorded overnight (8‐hours duration) in a sleep laboratory. The Mini‐Mental State Exam and Digit Symbol test were administered the morning after PSG recording nights. The patient's partner completed an e‐diary of the patient's sleep each morning throughout the treatment period, and the Sleep Disorders Inventory weekly in the e‐diary starting at visit 4. Partners rated their own sleep using the single‐item Sleep Quality Scale at visits 4, 5, and 6. The Neuropsychiatric Inventory was completed by an interview with the partner and scored by a qualified trained rater at visits 4 and 6. Clinicians completed the Clinical Global Impression–Severity for insomnia at visits 4, 5, and 6
Figure 2
Figure 2
Patient disposition. aThere were 43 reasons for exclusion, and an individual patient could be excluded for multiple reasons. The main reasons for exclusions were the following: underlying pathology of sleep identified during the screening polysomnography (PSG) night, N = 116; Mini‐Mental State Exam outside the 12 to 26 range, N = 19; total sleep time (TST) >6.5 hours during the screening PSG night, N = 19; and mean TST over the screening and baseline PSG nights >6 hours, N = 15. bThe number of patients in the full‐analysis‐set for the primary endpoint of change from baseline in TST at week 4. In the suvorexant group, seven patients were excluded due to missing PSG data. In the placebo group, two patients were excluded due to missing PSG data and two patients were excluded due to Good Clinical Practice noncompliance issues at one site
Figure 3
Figure 3
Change from baseline in total sleep time (TST) and wake after persistent sleep onset (WASO) by thirds of the night (first, middle, and last) with baseline values in minutes
Figure 4
Figure 4
Subgroup analyses based on patient characteristics at baseline: point estimates and 95% confidence intervals for the difference between suvorexant and placebo in change from baseline in total sleep time (TST; minutes) at week 4. aRegions were North America, Europe, and Other. The confidence interval for Europe was not calculated because there were fewer than 20 participants per treatment group. Abbreviations: AHI, Apnea/Hypopnea Index (number of apneas or hypopneas per hour assessed during PSG); APOE ɛ4, apolipoprotein ɛ4 gene variant; MMSE, Mini‐Mental State Exam

References

    1. Zhao QF, Tan L, Wang HF, et al. The prevalence of neuropsychiatric symptoms in Alzheimer's disease: systematic review and meta‐analysis. J Affect Disord. 2016;190:264‐271.
    1. Xie L, Kang H, Xu Q, et al. Sleep drives metabolite clearance from the adult brain. Science. 2013;342(6156):373‐377.
    1. Mander BA, Winer JR, Jagust WJ, Walker MP. Sleep: a novel mechanistic pathway, biomarker, and treatment target in the pathology of Alzheimer's disease? Trends Neurosci. 2016;39(8):552‐566.
    1. Krause AJ, Simon EB, Mander BA, et al. The sleep‐deprived human brain. Nat Rev Neurosci. 2017;18(7):404‐418.
    1. Holth J, Patel T, Holtzman DM. Sleep in Alzheimer's disease ‐ beyond amyloid. Neurobiol Sleep Circadian Rhythms. 2017;2:4‐14.
    1. Shokri‐Kojori E, Wang GJ, Wiers CE, et al. beta‐Amyloid accumulation in the human brain after one night of sleep deprivation. Proc Natl Acad Sci U S A. 2018;115(17):4483‐4488.
    1. 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(474).
    1. Holth JK, Fritschi SK, Wang C, et al. The sleep‐wake cycle regulates brain interstitial fluid tau in mice and CSF tau in humans. Science. 2019;363(6429):880‐884.
    1. Macedo AC, Balouch S, Tabet N. Is sleep disruption a risk factor for Alzheimer's disease? J Alzheimers Dis. 2017;58(4):993‐1002.
    1. Asayama K, Yamadera H, Ito T, Suzuki H, Kudo Y, Endo S. Double blind study of melatonin effects on the sleep‐wake rhythm, cognitive and non‐cognitive functions in Alzheimer type dementia. J Nippon Med Sch. 2003;70(4):334‐341.
    1. Gehrman PR, Connor DJ, Martin JL, Shochat T, Corey‐Bloom J, Ancoli‐Israel S. Melatonin fails to improve sleep or agitation in double‐blind randomized placebo‐controlled trial of institutionalized patients with Alzheimer disease. Am J Geriatr Psychiatry. 2009;17(2):166‐169.
    1. Serfaty M, Kennell‐Webb S, Warner J, Blizard R, Raven P. Double blind randomised placebo controlled trial of low dose melatonin for sleep disorders in dementia. Int J Geriatr Psychiatry. 2002;17(12):1120‐1127.
    1. Singer C, Tractenberg RE, Kaye J, et al. A multicenter, placebo‐controlled trial of melatonin for sleep disturbance in Alzheimer's disease. Sleep. 2003;26(7):893‐901.
    1. Meguro K, Meguro M, Tanaka Y, Akanuma K, Yamaguchi K, Itoh M. Risperidone is effective for wandering and disturbed sleep/wake patterns in Alzheimer's disease. J Geriatr Psychiatry Neurol. 2004;17(2):61‐67.
    1. Onor ML, Saina M, Trevisiol M, Cristante T, Aguglia E. Clinical experience with risperidone in the treatment of behavioral and psychological symptoms of dementia. Prog Neuropsychopharmacol Biol Psychiatry. 2007;31(1):205‐209.
    1. Yin Y, Liu Y, Zhuang J, et al. Low‐dose atypical antipsychotic risperidone improves the 5‐year outcome in Alzheimer's disease Patients with sleep disturbances. Pharmacology. 2015;96(3‐4):155‐162.
    1. Scoralick FM, Louzada LL, Quintas JL, Naves JO, Camargos EF, Nobrega OT. Mirtazapine does not improve sleep disorders in Alzheimer's disease: results from a double‐blind, placebo‐controlled pilot study. Psychogeriatrics. 2017;17(2):89‐96.
    1. Lopez‐Pousa S, Garre‐Olmo J, Vilalta‐Franch J, Turon‐Estrada A, Pericot‐Nierga I. Trazodone for Alzheimer's disease: a naturalistic follow‐up study. Arch Gerontol Geriatr. 2008;47(2):207‐215.
    1. Camargos EF, Louzada LL, Quintas JL, Naves JO, Louzada FM, Nobrega OT. Trazodone improves sleep parameters in Alzheimer disease patients: a randomized, double‐blind, and placebo‐controlled study. Am J Geriatr Psychiatry. 2014;22(12):1565‐1574.
    1. Salami O, Lyketsos C, Rao V. Treatment of sleep disturbance in Alzheimer's dementia. Int J Geriatr Psychiatry. 2011;26(8):771‐782.
    1. Ellul J, Archer N, Foy CM, et al. The effects of commonly prescribed drugs in patients with Alzheimer's disease on the rate of deterioration. J Neurol Neurosurg Psychiatry. 2007;78(3):233‐239.
    1. Elmstahl S, Stenberg I, Annerstedt L, Ingvad B. Behavioral disturbances and pharmacological treatment of patients with dementia in family caregiving: a 2‐year follow‐up. Int Psychogeriatr. 1998;10(3):239‐252.
    1. Cox CD, Breslin MJ, Whitman DB, et al. Discovery of the dual orexin receptor antagonist [(7R)‐4‐(5‐chloro‐1,3‐benzoxazol‐2‐yl)‐7‐methyl‐1,4‐diazepan‐1‐yl][5‐methyl‐2‐(2H ‐1,2,3‐triazol‐2‐yl)phenyl]methanone (MK‐4305) for the treatment of insomnia. J Med Chem. 2010;53(14):5320‐5332.
    1. Winrow CJ, Gotter AL, Cox CD, et al. Promotion of sleep by suvorexant‐a novel dual orexin receptor antagonist. J Neurogenet. 2011;25(1‐2):52‐61.
    1. Herring WJ, Connor KM, Ivgy‐May N, et al. Suvorexant in patients with insomnia: results from two 3‐month randomized controlled clinical trials. Biol Psychiatry. 2016;79(2):136‐148.
    1. Herring WJ, Connor KM, Snyder E, et al. Suvorexant in patients with insomnia: pooled analyses of three‐month data from phase‐3 randomized controlled clinical trials. J Clin Sleep Med. 2016;12(9):1215‐1225.
    1. Herring WJ, Connor KM, Snyder E, et al. Suvorexant in elderly patients with insomnia: pooled analyses of data from phase III randomized controlled clinical trials. Am J Geriatr Psychiatry. 2017;25(7):791‐802.
    1. Herring WJ, Snyder E, Budd K, et al. Orexin receptor antagonism for treatment of insomnia: a randomized clinical trial of suvorexant. Neurology. 2012;79(23):2265‐2274.
    1. Michelson D, Snyder E, Paradis E, et al. Safety and efficacy of suvorexant during 1‐year treatment of insomnia with subsequent abrupt treatment discontinuation: a phase 3 randomised, double‐blind, placebo‐controlled trial. Lancet Neurol. 2014;13(5):461‐471.
    1. Fronczek R, van Geest S, Frolich M, et al. Hypocretin (orexin) loss in Alzheimer's disease. Neurobiol Aging. 2012;33(8):1642‐1650.
    1. Liguori C, Chiaravalloti A, Nuccetelli M, et al. Hypothalamic dysfunction is related to sleep impairment and CSF biomarkers in Alzheimer's disease. J Neurol. 2017;264(11):2215‐2223.
    1. Liguori C. Orexin and Alzheimer's disease. Curr Top Behav Neurosci. 2017;33:305‐322.
    1. Gabelle A, Jaussent I, Hirtz C, et al. Cerebrospinal fluid levels of orexin‐A and histamine, and sleep profile within the Alzheimer process. Neurobiol Aging. 2017;53:59‐66.
    1. Liguori C, Nuccetelli M, Izzi F, et al. Rapid eye movement sleep disruption and sleep fragmentation are associated with increased orexin‐A cerebrospinal‐fluid levels in mild cognitive impairment due to Alzheimer's disease. Neurobiol Aging. 2016;40:120‐126.
    1. McKhann GM, Knopman DS, Chertkow H, et al. The diagnosis of dementia due to Alzheimer's disease: recommendations from the National Institute on Aging‐Alzheimer's Association workgroups on diagnostic guidelines for Alzheimer's disease. Alzheimers Dement. 2011;7(3):263‐269.
    1. American Psychiatric Association . Diagnostic and Statistical Manual of Mental Disorders. 5th ed. Arlington, VA: American Psychiatric Association; 2013.
    1. Folstein MF, Folstein SE, McHugh PR. "Mini‐mental state". A practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res. 1975;12(3):189‐198.
    1. Busner J, Targum SD. The clinical global impressions scale: applying a research tool in clinical practice. Psychiatry (Edgmont). 2007;4(7):28‐37.
    1. Silber MH, Ancoli‐Israel S, Bonnet MH, et al. The visual scoring of sleep in adults. J Clin Sleep Med. 2007;3(2):121‐131.
    1. Berry RB BR, Gamaldo CE, Harding SM, Lloyd RM, Marcus CL and Vaughn BV for the American Academy of Sleep Medicine . The AASM Manual for the Scoring of Sleep and Associated Events: Rules, Terminology and Technical Specifications, Version 2.2. Darien, Illinois: American Academy of Sleep Medicine; 2015.
    1. Tractenberg RE, Singer CM, Cummings JL, Thal LJ. The sleep disorders Inventory: an instrument for studies of sleep disturbance in persons with Alzheimer's disease. J Sleep Res. 2003;12(4):331‐337.
    1. Jaeger J. Digit Symbol Substitution Test: The case for sensitivity over specificity in neuropsychological testing. J Clin Psychopharmacol. 2018;38(5):513‐519.
    1. Cummings JL, Mega M, Gray K, Rosenberg‐Thompson S, Carusi DA, Gornbein J. The neuropsychiatric inventory: comprehensive assessment of psychopathology in dementia. Neurology. 1994;44(12):2308‐2314.
    1. Sateia MJ, Buysse DJ, Krystal AD, Neubauer DN, Heald JL. Clinical practice guideline for the pharmacologic treatment of chronic insomnia in adults: an American Academy of Sleep Medicine Clinical Practice Guideline. J Clin Sleep Med. 2017;13(2):307‐349.
    1. Snyder E, Ma J, Svetnik V, et al. Effects of suvorexant on sleep architecture and power spectral profile in patients with insomnia: analysis of pooled phase 3 data. Sleep Med. 2016;19:93‐100.
    1. Siemers ER, Sundell KL, Carlson C, et al. Phase 3 solanezumab trials: secondary outcomes in mild Alzheimer's disease patients. Alzheimers Dement. 2016;12(2):110‐120.

Source: PubMed

3
Se inscrever