ArticlePDF Available

Abstract and Figures

There is an association between Alzheimer disease and sleep-disordered breathing. Donepezil is the drug most frequently used to treat cognitive symptoms in Alzheimer disease. This study evaluates the effects of donepezil on obstructive sleep apnea in patients with Alzheimer disease. Randomized, double-blind, placebo-controlled design. Twenty-three patients with mild-to-moderate Alzheimer disease and apnea-hypopnea index (AHI) > 5/h were allocated to two groups: donepezil treated (n = 11) and placebo treated (n = 12). Polysomnography and cognitive evaluation using Alzheimer disease assessment scale-cognitive (ADAS-cog) subscale were performed at baseline and after 3 months. Cognitive and sleep data were analyzed using analysis of variance. AHI and oxygen saturation improved significantly after donepezil treatment compared to baseline and placebo (p < 0.05). Rapid eye movement (REM) sleep duration increased after donepezil treatment (p < 0.05). ADAS-cog scores improved after donepezil treatment, although they did not correlate with REM sleep increase and sleep apnea improvement (p < 0.01). Donepezil treatment improved AHI and oxygen saturation in patients with Alzheimer disease. Treatment also increased REM sleep duration and reduced ADAS-cog scores. Trial registration: ClinicalTrials.gov Identifier: NCT00480870.
Content may be subject to copyright.
Donepezil Improves Obstructive Sleep
Apnea in Alzheimer Disease*
A Double-Blind, Placebo-Controlled Study
Walter Moraes, MD, PhD; Dalva Poyares, MD, PhD;
Lucia Sukys-Claudino, MD; Christian Guilleminault, MD, PhD;
and Sergio Tufik, MD, PhD
Background: There is an association between Alzheimer disease and sleep-disordered breathing.
Donepezil is the drug most frequently used to treat cognitive symptoms in Alzheimer disease.
This study evaluates the effects of donepezil on obstructive sleep apnea in patients with
Alzheimer disease.
Methods: Randomized, double-blind, placebo-controlled design. Twenty-three patients with
mild-to-moderate Alzheimer disease and apnea-hypopnea index (AHI) > 5/h were allocated to
two groups: donepezil treated (n 11) and placebo treated (n 12). Polysomnography and
cognitive evaluation using Alzheimer disease assessment scale-cognitive (ADAS-cog) subscale
were performed at baseline and after 3 months. Cognitive and sleep data were analyzed using
analysis of variance.
Results: AHI and oxygen saturation improved significantly after donepezil treatment compared to
baseline and placebo (p < 0.05). Rapid eye movement (REM) sleep duration increased after
donepezil treatment (p < 0.05). ADAS-cog scores improved after donepezil treatment, although
they did not correlate with REM sleep increase and sleep apnea improvement (p < 0.01).
Conclusions: Donepezil treatment improved AHI and oxygen saturation in patients with Alzhei-
mer disease. Treatment also increased REM sleep duration and reduced ADAS-cog scores.
Trial registration: ClinicalTrials.gov Identifier: NCT00480870. (CHEST 2008; 133:677– 683)
Key words: Alzheimer; donepezil; oxygen saturation; polysomnography; rapid eye movement sleep; sleep apnea
Abbreviations: AASM American Academy of Sleep Medicine; ADAS-cog Alzheimer disease assessment scale-
cognitive; AHI apnea-hypopnea index; ANOVA analysis of variance; BMI body mass index; CDR clinical dementia
rating; MMSE mini-mental state examination; OSA obstructive sleep apnea; OSA-AD obstructive sleep apnea-
Alzheimer disease; REM rapid eye movement; WASO wake after sleep onset
T
he association between Alzheimer disease and
sleep-disordered breathing has been frequently
studied.
1–5
The presence of apolipoprotein E epsilon
4 seems to be linked to the occurrence of both
conditions, suggesting a genetic basis.
3,6
Central-
acting cholinesterase inhibitors are the first primary
pharmacologic treatments approved for Alzheimer
disease, for which donepezil is the most frequently
used.
7
Multicenter studies
7–10
have found little tox-
icity, and its side effects (diarrhea, nausea, vomiting,
nightmares, among others) are mild and transient.
Donepezil is a reversible inhibitor of the acetyl-
cholinesterase enzyme, thus enhancing cholinergic
transmission.
11
Its half-life is approximately 70 h.
11
It
is excreted intact in the urine and metabolized into
four major metabolites, two of which are known to
be active.
11
Cholinergic activity also influences the upper air-
way opening via central and peripheral mecha-
nisms.
12–15
Decreased thalamic pontine cholinergic
projections may affect respiratory drive, leading to
both central and obstructive apnea at least in certain
degenerative conditions.
14,15
Since impairment of
cholinergic transmission is a putative pathophysio-
logic mechanism for Alzheimer disease, it is ex-
pected that cholinergic-related respiratory distur-
bances may occur.
7,8,16
However, donepezil has been
shown to augment rapid eye movement (REM) sleep
Original Research
SLEEP MEDICINE
www.chestjournal.org CHEST / 133/3/MARCH, 2008 677
in patients with Alzheimer disease, a sleep stage
during which sleep apnea events are more fre-
quent.
7,17–19
Such conflicting effects make it difficult to
know a priori whether improvement of sleep apnea is
likely to occur during donepezil treatment.
14,15
Mindful
of the above, the authors hypothesized the following:
(1) donepezil may influence respiratory variables dur-
ing sleep in patients with obstructive sleep apnea-
Alzheimer disease (OSA-AD), and (2) there is a corre-
lation between cognitive improvement and sleep
respiratory changes.
Materials and Methods
Population
Forty patients with mild-to-moderate Alzheimer disease were
consecutively recruited from geriatric and neurologic clinics at
the University Hospital of the Universidade Federal de Sa˜o
Paulo. Thirty of them had obstructive sleep apnea (OSA) and
presented with apnea-hypopnea index (AHI) 5/h according to
American Academy of Sleep Medicine (AASM) criteria.
20
OSA
was not previously diagnosed or treated in these patients. There
were technical problems in the polysomnography of seven pa-
tients due to frequent patient-movement artifacts. The final
sample consisted of the remaining 23 patients who were ran-
domly allocated to two groups: donepezil treated (n 11) and
placebo treated (n 12).
The diagnosis of Alzheimer disease was based on the proba-
bility criteria of the Alzheimer’s Disease and Related Disorders
Association.
21
Patients were rated 1 and 2 (mild-to-moderate
level) on the Brazilian version of the clinical dementia rating
(CDR), and the more severe cases were excluded.
22
Potential
subjects were evaluated by history, physical examination, mini-
mental state examination (MMSE), Alzheimer disease assess-
ment scale-cognitive (ADAS-cog) subscale,
23
brain MRI, and
laboratory tests (hematologic evaluation, renal and liver func-
tions, vitamin B
12
, folic acid, thyroid hormones, fasting glycemia,
venereal disease research test, and urine sediment).
.
Exclusion
criteria were the presence of other causes of dementia; MRI
compatible with other etiology of dementia; and pulmonary, cardiac,
and other current severe medical or psychiatric diseases. No psy-
choactive drugs other than the experimental drug were taken during
the trial or the preceding 1-month period. Alcohol and sleep
medications were not permitted during the trial and the preceding
1-month period. Caregivers were instructed to take note of the use
of any nonpsychoactive drugs in daily sleep diaries.
Drugs and Administration
Tablets containing placebo were prepared and packed in the
same fashion as those with 5 mg of donepezil. Donepezil and
placebo were administered in a single dose at bedtime. The
dosage was one tablet per day in the first month and two tablets
per day for the next 2 months. After completion of the protocol,
patients were referred to our sleep clinic to have OSA treated.
Randomization
Each medication-containing box was packed by a standard
pharmacy service. Boxes were coded as A or B, indicating placebo
or donepezil (the signification of the codes was kept in a closed
envelope). A random number list with uniform distribution from
0 to 1 was generated using software (Statistica; Statsoft; Tulsa, OK).
Patients were consecutively allocated to two treatment groups
according to the random number list: 0.5 to group A and 0.5 to
group B. Researchers were blind to patient conditions when record-
ing and scoring parameters. Codes were opened and assigned to
each patient when statistical analysis was performed.
Polysomnography and Scoring
Patients were submitted to 2 nights of polysomnography for
habituation purposes, followed by a baseline recording before the
onset of treatment and a second recording after 3 months of
treatment. The minimum duration of polysomnography was 7 h.
Polysomnography was performed in the sleep laboratory of the
Psychobiology Department at Universidade Federal de Sa˜o Paulo
using a 32-channel device (Sonolab; Meditron; Sao Paulo, Brazil):
resolution, 256 Hz; 22 EEG; 2 electro-oculogram, 1 chin electro-
myogram, 1 leg electromyogram, 1 ECG, 1 tracheal microphone, 1
oronasal thermistor, 1 nasal pressure transducer (Pro-Tech Services;
Mukilteo, WI), 2 chest and abdominal effort sensors, and 1 pulse
oximeter (Nellcor; Pleasanton, CA). Two researchers scored the
recordings visually using Rechtshaffen and Kales and AASM criteria
for respiratory parameters and microarousals.
24–26
Apneas and hypopneas were scored according to AASM crite-
ria.
20
Variables analyzed were total sleep time, sleep efficiency
(sleep time/recording time 100), sleep latency (time from
lights off to sleep onset), REM sleep latency (time from sleep
onset to REM sleep onset), wake after sleep onset (WASO),
REM and non-REM sleep percentage, microarousal index (mi-
croarousals per hour), overall AHI, obstructive AHI, central AHI,
mixed AHI, REM AHI, non-REM AHI, average oxygen satura-
tion, and percentage of sleep time with oxygen saturation 90%.
Apnea-hypopnea improvement ratio was calculated was calcu-
lated as follows: AHI improvement initial AHI final AHI/
initial AHI. Caregivers answered a Portuguese language-modified
version of Stanford Sleep Disorders Questionnaire after polysom-
nography to rule out other major sleep disorders.
27
Psychometric Testing
The ADAS-cog subscale evaluates multiple cognitive functions
including word evocation, verbal fluency, understanding of sim-
ple commands, constructive praxis, ideational praxis, temporo-
spatial orientation, word recognition, verbal fluency, vocabulary,
and understanding. Scores range from 0 to 70, with higher scores
indicating more cognitive deterioration. The Brazilian version of
ADAS-cog
22
was applied before donepezil treatment and again
after 3 months.
*From the Psychobiology Department (Drs. Moraes, Poyares,
Sukys-Claudino, and Tufik), Universidade Federal de Sa˜o Paulo,
Sa˜o Paulo, Brazil; and Human Sleep Research Center (Dr.
Guilleminault), Department of Psychiatry and Behavioral Sci-
ence, School of Medicine, Stanford University, Palo Alto, CA.
This work was supported by Fundac¸a˜o de Amparo a` Pesquisa do
Estado de Sa˜o Paulo and Associac¸a˜o Fundo de Incentivo a`
Psicofarmacologia.
The authors have no conflicts of interest to disclose.
Manuscript received June 22, 2007; revision accepted December
1, 2007.
Reproduction of this article is prohibited without written permission
from the American College of Chest Physicians (www.chestjournal.
org/misc/reprints.shtml).
Correspondence to: Walter Moraes, MD, PhD, R. Manuel de
Paiva, 313-Sa˜o Paulo-SP, 04106-020, Brazil; e-mail: waltermoraes@
giro.com.br
DOI: 10.1378/chest.07-1446
678 Original Research
Ethics
Subjects or caregivers signed informed consent forms that ex-
plained possible side effects of donepezil and assured them that they
could leave the trial at any moment. All subjects were assured that
they would receive clinical treatment. The Ethics Committee at
Universidade Federal de Sa˜o Paulo authorized the study.
Statistical Analysis
One-way analysis of variance (ANOVA) was used to compare
all variables for donepezil and placebo groups during the baseline
recording night. Polysomnographic and cognitive data at baseline
and after 3 months of treatment were analyzed using two-way
ANOVA for repeated measures with treatment group and treat-
ment time as the main factors and time/treatment interaction
effect followed by Bonferroni test, with p 0.01 comparing data.
The Spearman correlation test was used to assess possible
correlations between cognitive, body mass index (BMI) and sleep
respiratory parameters in the donepezil-treated group.
Results
General
Table 1 shows that there were no significant
differences in age, gender, BMI, MMSE, and CDR
scores between the donepezil and placebo groups
before treatment. There was no significant correla-
tion between AHI improvement ratio and BMI. MRI
scan showed brain atrophy. Results of other labora-
tory tests were within normal range.
Adverse Effects
Mild and transitory side effects involving nausea
and headache occurred in three patients receiving
donepezil. There were no reports of nightmares or
worsening of sleep when caregivers were questioned.
Sleep Polysomnographic Variables
REM sleep percentage increased after 3 months
of donepezil treatment, as seen in Table 2. There was
a significant improvement in the following respira-
tory parameters after 3 months of donepezil treat-
ment: AHI, obstructive AHI, mixed AHI, lowest
oxygen saturation, and time spent with oxygen satu-
ration 90% (Table 2). Central AHI did not im-
prove significantly after donepezil treatment (Table
2, Fig 1). There was also a significant reduction in
non-REM AHI after donepezil treatment (interac-
tion factor F[1,21] 5.39, p 0.03) [Table 2]. Al-
though there was a trend toward reduction of REM
AHI, it was not significant (interaction factor
F[1,21] 3.47 p 0.07) [Table 2]. Microarousal
index decreased significantly after donepezil treat-
ment due to a reduction in respiratory-related mi-
croarousals (Table 2). Nine donepezil-treated pa-
tients (81%) had improvement of AHI (Fig 1). Six
patients (54%) spent 1% sleep time with oxygen
saturation 90% before donepezil treatment (Fig
1). Five of these patients (83%) improved after
donepezil treatment (Fig 1). There were no signifi-
cant differences between donepezil and placebo
groups in the following polysomnographic parame-
ters: total sleep time, sleep efficiency, sleep latency,
REM sleep latency, WASO, and percentage of non-
REM sleep stages. There was no significant differ-
ence in percentage of time spent in supine position
within and between groups (Table 2).
Psychometric Variables
ADAS-cog scores significantly decreased after 3
months of donepezil treatment (Table 2). Correlations
between ADAS-cog scores, AHI, average oxygen satu-
ration, time of oxygen saturation spent 90%, mi-
croarousal index, and other polysomnographic variables
did not reach statistical significance in the donepezil-
treated group before and after therapy.
Discussion
The main finding of the present study was the
significant improvement in AHI and oxygen satura-
tion of OSA-AD patients treated with donepezil. No
previous polysomnographic studies on the effect of
donepezil on sleep apnea in Alzheimer disease have
been undertaken. As expected, donepezil was well
tolerated and no major side effects occurred.
10,11
No
evidence of worsening of sleep or nightmares was
found in this study. This is in accordance with the
Table 1—Sociodemographic Data*
Variables Donepezil-Treated Group (n 11) Placebo-Treated Group (n 12) p Value†
Age, yr 76.8 6.2 (68 to 86) 72.6 11.0 (62 to 87) 0.27
Male/female gender 3/8 5/7 0.49
BMI, kg/m
2
26.3 4.8 (17.2 to 32.0) 26.6 4.1 (17.0 to 32.1) 0.85
MMSE 19 3.6 (13 to 26) 17.2 7.8 (6 to 27) 0.50
CDR 1.3 0.5 1.3 0.5 0.76
*Data are presented as mean SD (range) or No.
†p 0.05, ANOVA.
www.chestjournal.org CHEST / 133/3/MARCH, 2008 679
findings of other authors
11
who have studied larger
samples in general practice. Acetylcholinesterase
inhibitors elevate central and peripheral acetylcho-
line levels. However, the distribution of donepezil
varies between different brain and peripheral tis-
sues.
28
An increase in REM sleep proportion after
donepezil treatment was an expected finding.
7
In contrast to the prolific literature on physical
and surgical treatments for sleep apnea, there is a
dearth of effective pharmacologic approaches.
29–32
Most drugs previously tested for this purpose acted
on monoaminergic and adenosinergic systems and
showed unsuccessful or ambiguous results.
29–32
Hed-
ner and colleagues
12
published the only previous
report on a cholinergic pharmacologic treatment in
humans using an IV infusion of the anticholinester-
ase drug physostigmine in sleep-disordered breath-
ing patients, improving both obstructive and central
respiratory events mostly during REM sleep. How-
ever, that study
12
focused on the acute effect of a
drug that does not sustain plasma levels during the
entire night. We have selected a cholinergic drug
already approved by many regulatory agencies for
the treatment of Alzheimer disease and that has been
extensively studied and shown to be safe and phar-
macologically stable.
10
Taking advantage of the char-
acteristics of this drug, we were able to show that the
beneficial respiratory effect is maintained after long-
term use, possibly adding to its therapeutic spec-
trum. In contrast to this previous report,
12
we found
that overall AHI improvement was due to a reduc-
tion in obstructive and mixed respiratory events. In
addition, we did not find a significant improvement
in AHI during REM sleep, although there was a
nonsignificant trend toward reduction. These results
suggest that AHI improvement after donepezil treat-
ment is related to cholinergic mechanisms involved
in obstructive and mixed apnea and is not specifically
dependent on REM AHI reduction. OSA-AD pa-
tients showed different individual responses to done-
pezil. We found no parameter that could predict
individual treatment outcome. Future studies are
needed to explain individual differences in response
to cholinergic drugs for OSA treatment. As expected,
there was a high degree of variability in respiratory
parameters of the control group and no clear ten-
dency to worsening.
33,34
Besides respiratory effects,
donepezil increased REM sleep, and decreased mi-
croarousal index and ADAS-cog scores after 3
months of donepezil treatment, confirming findings
of previous research.
7,17,18
Hedner et al
35
reported a
similar finding in non-Alzheimer OSA patients in
abstract form.
Table 2—Cognitive and Polysomnographic Findings*
Variables
Donepezil-Treated
Group (n 11)
Placebo-Treated Group (n 12)
Interaction
p ValueBaseline Third Month Baseline Third Month
Total sleep time, min 307.7 90.9 312.9 106.8 296.9 61.6 295.6 81.3 0.81
Sleep efficiency, % 76.6 18.0 73.4 23.4 74.2 12.7 74.0 17.0 0.56
Sleep latency, min 35.6 86.6 41.9 86.0 14.5 21.5 16.9 19.1 0.63
REM sleep latency, min 125.7 80.7 138.7 136.4 129.5 79.4 133.7 89.4 0.87
WASO, min 91.9 74.4 114.8 111.1 104.4 56.7 100.9 64.9 0.23
Stage 1, % 9.0 7.7 9.1 11.2 14.4 11.3 14.8 13.0 0.90
Stage 2, % 54.2 16.7 49.0 14.8 54.5 13.5 52.7 12.8 0.53
Slow-wave sleep, % 30.3 16.6 30.5 14.8 22.0 15.3 21.8 10.5 0.95
REM sleep % 7.2 4.3 15.9 11.1 11.8 6.6 9.7 5.9 0.022†
Time spent in supine position, % 42.7 17.1 41.7 17.2 39.2 24.7 36.1 26.4 0.63
Microarousal index 26.3 12.6 15.7 8.3 23.9 19.7 23.8 21.0 0.021†
Nonrespiratory microarousal index 15.5 14.8 10.9 4.7 12.3 10.8 11.8 8.8 0.33
Respiratory microarousal index 11.0 10.4 5.1 7.1 11.6 15.9 12.0 8.2 0.001†
AHI, /h 20.0 15.9 9.9 11.5 23.2 26.4 22.9 28.8 0.035†
REM AHI, /h 24.7 19.4 7.6 7.9 35.4 31.5 39.0 25.6 0.077
Obstructive AHI, /h 19.4 15.4 9.2 10.3 22.7 25.8 21.3 26.7 0.047†
Central AHI, /h 0.1 0.2 0.6 1.9 0.1 0.4 0.3 0.6 0.56
Mixed AHI, /h 0.4 0.8 0.1 0.2 0.3 0.8 1.3 2.3 0.033†
Average oxygen saturation, % 91.5 6.1 94.0 2.0 94.0 2.5 93.5 3.1 0.12
Lowest oxygen saturation, % 80.8 8.3 85.7 6.3 83.0 11.1 81.9 10.9 0.002†
Desaturation events, No. 15.4 18.2 5.8 8.7 31.7 44.3 30.4 61.3 0.36
Time with oxygen saturation 90%, % 13.4 17.4 3.7 4.8 8.5 14.7 11.0 20.1 0.017†
ADAS-cog score 34.5 15.8 29.7 15.7 29.3 17.3 31.8 18.5 0.004†
*Data are presented as mean SD.
†p 0.05, ANOVA.
680
Original Research
As previously mentioned, it was expected that the
REM sleep increase would be associated to the
worsening of OSA because respiratory events are
more frequent during this sleep stage.
12,19
However,
cholinergic drugs display more complex effects on
respiration. Available evidence demonstrates that
cholinergic stimulation has potent excitatory effects
on medullary respiratory neurons and respiratory
motoneurones.
14
It also affects the central chemo-
sensitive drive of the respiratory control system.
14
Upper airway dilating muscle activity is characterized
by an early peaking pattern that serves to dilate the
upper airway at the time when the greatest negative
intraluminal pressure is generated by contraction of the
chest wall muscles and diaphragm.
36
An experimental
study
36
showed an increase in hypoglossus nerve activ-
ity with consequent opening of the upper airway fol-
lowing carbachol pontine injection in cats. Physostig-
mine administered to cholinergic neurons located in
the rostral ventrolateral medulla of anesthetized
vagotomized and artificially ventilated cats resulted
in elongation of the hypoglossal-to-phrenic nerve
firing interval resulting in improved oxygenation.
37
It
also antagonizes respiratory depression induced by
fentanyl in rats.
38
Furthermore, physostigmine stimu-
lates saliva production, resulting in reduced superficial
tension that increases upper airway stability, which may
represent another mechanism for improving OSA.
39,40
Figure 1. AHI before (1) and after (2) donepezil (top left, A) or placebo (top right, B) treatment, and
time spent with oxygen saturation 90% before (1) and after (2) donepezil (bottom left, C) or placebo
(bottom right, D) treatment. An outlier (Subject 12) is not presented (top right, B) due to scale: AHI
was 81 events/h at entry and 87 events/h after placebo.
www.chestjournal.org CHEST / 133/3/MARCH, 2008 681
Cholinergic stimulation of the respiratory center and
the carotid body increases sensitivity to hypoxia improv-
ing the chemoreflex response.
12,14,15,41
Such effects
may contribute to the cholinergic-induced improve-
ment of the peripheral and central components of
respiratory regulation during sleep.
12,14,15,37–42
In our
study, most of the reduction of the microarousal index
in the donepezil group was attributable to a significant
decrease in respiratory-related microarousals, even
though cholinergic drugs may elicit arousal.
43,44
Donepezil-related cognitive improvement measured
by ADAS-cog subscale did not correlate with respira-
tory parameters, suggesting independence between
cognition and sleep respiratory symptoms in patients
with Alzheimer disease.
1
However, further clarification
is required as to whether or not OSA contributes to
cognitive deterioration in Alzheimer disease.
3–5
In summary our results support the concept that
cholinergic transmission is involved in the patho-
genesis of OSA in Alzheimer disease, suggesting a
possible target for pharmacologic intervention.
Larger placebo-controlled studies are needed to
confirm these results.
ACKNOWLEDGMENT: The authors thank Dr. Robert Skomro
for his comments.
References
1 Hoch CC, Reynolds CF III, Nebes RD, et al. Clinical
significance of sleep-disordered breathing in Alzheimer’s
disease: preliminary data. J Am Geriatr Soc 1989; 37:138–144
2 Reynolds CF III, Kupfer DJ, Taska LS, et al. Sleep apnea in
Alzheimer’s dementia: correlation with mental deterioration.
J Clin Psychiatry 1985; 46:257–261
3 Bliwise DL. Sleep apnea, APOE4 and Alzheimer’s disease: 20
years and counting? J Psychosom Res 2002; 53:539–546
4 Bliwise DL. Sleep disorders in Alzheimer’s disease and other
dementias. Clin Cornerstone 2004; 6(suppl 1A):S16 –S28
5 Abrams B. Add Alzheimer’s to the list of sleep apnea
consequences. Med Hypotheses 2005; 65:1201–1202
6 Kadotani H, Kadotani T, Young T, et al. Association between
apolipoprotein E epsilon4 and sleep-disordered breathing in
adults. JAMA 2001; 285:2888 –2890
7 Moraes WAS, Poyares DR, Guilleminault C, et al. The effect
of donepezil on sleep and REM sleep EEG in patients with
Alzheimer disease: a double-blind placebo-controlled study.
Sleep 2006; 29:199 –205
8 Pratt RD, Perdomo CA, Surick IW, et al. Donepazil: tolera-
bility and safety in Alzheimer’s disease. Int J Clin Pract 2002;
56:710–717
9 Rogers SL, Friedhoff LT. The efficacy and safety of donepezil
in patients with Alzheimer’s disease: results of a US multi-
centre, randomized, double-blind, placebo-controlled trial.
Dementia 1996; 7:293–303
10 Dunn NR, Pearce GL, Shakir AW. Adverse effects associated
with the use of donepezil in general practice in England.
J Psychopharmacol 2000; 14:406 408
11 Davis KL. Current and experimental therapeutics of Alzhei-
mer’s disease. In: Davis KL, Charney D, Coyle JT, et al, eds.
Neuropsychopharmacology: the fifth generation of progress.
Philadelphia, PA: Lippincott, Williams and Wilkins, 2002;
1243–1252
12 Hedner J, Kraiczi H, Peker Y, et al. Reduction of sleep-
disordered breathing after physostigmine. Am J Respir Crit
Care Med 2003; 168:1246 –1251
13 O’Donnell CP, Schwartz AR, Smith PL. Upper airway col-
lapsibility: the importance of gender and adiposity. Am J
Respir Crit Care Med 2000; 162:1606 –1607
14 Bellingham MC, Ireland MF. Contribution of cholinergic
systems to state dependent modulation of respiratory control.
Respir Physiol Neurobiol 2002; 131:135–144
15 Gilman S, Chervin RD, Koeppe RA, et al. Obstructive sleep
apnea is related to a thalamic cholinergic deficit in MSA.
Neurology 2003; 61:35–39
16 Brun A, Englund E. Regional pattern of degeneration in
Alzheimer’s disease: neuronal loss and histopathological grad-
ing. Histopathology 1981; 5:459 –564
17 Kanbayashi T, Sugiyama T, Aizawa R, et al. Effects of
donepezil (Aricept) on the rapid eye movement sleep of
normal subjects: psychiatry Clin Neurosci 2002; 56:307–308
18 Schredl M, Weber B, Leins ML, et al. Donepezil-induced
REM sleep augmentation enhances memory performance in
elderly, healthy persons. Exp Gerontol 2001; 36:353–361
19 Wiegand L, Zwillich CW, Wiegand D, et al. Changes in upper
airway muscle activation and ventilation during phasic REM
sleep in normal men. J Appl Physiol 1991; 71:488497
20 AASM Task Force report. Sleep-related breathing disorders
in adults: recommendations for syndrome definition and
measurement techniques in clinical research. Sleep 1999;
22:667–690
21 McKhann G, Drachman D, Folstein M, et al. Clinical
diagnosis of Alzheimer’s disease: report of the NINCDS-
ADRDA Work Group under the auspices of Department of
Health and Human Services Task Force on Alzheimer’s
Disease. Neurology 1984; 34:939 –944
22 Bertolucci PHF, Okamoto IV, Brucki SMD, et al. Applica-
bility of the CERAD Neuropsychological Battery to Brazilian
elderly. Arq Neuropsiquiatr 2001; 59:532–536
23 Schulz RR, Siviero MO. Bertolucci PHF. The cognitive
subscale of the Alzheimer’s Disease Assessment Scale in a
Brazilian sample. Braz J Med Biol Res 2001; 34:1295–1302
24 Rechtschaffen A, Kales A. A manual of standardized termi-
nology, techniques and scoring system for sleep states of
human subjects. Washington, DC: US Government Printing
Office, 1968
25 American Sleep Disorders Association. EEG arousals: scoring
rules and examples: a preliminary report from Sleep Disor-
ders Atlas Task Force of the American Sleep. Disorders
Association. Sleep 1992; 15:173–184
26 ASDA Atlas Task Force. Recording and scoring leg move-
ments. Sleep 1993; 16:748 –759
27 Braz S, Neumann BG, Tufik S. Avaliac¸a˜o dos distu´rbios do
sono: elaborac¸a˜o e validac¸a˜o de um questiona´rio. Revista da
ABP-APAL (Revista Brasileira de Psiquiatria) 1987; 9:9–14
28 Kasa P, Papp H, Kasa P Jr, et al. Donepezil dose-dependently
inhibits acetylcholinesterase activity in various areas and in
the presynaptic cholinergic and the postsynaptic cholinocep-
tive enzyme-positive structures in the human and rat brain.
Neuroscience 2000; 101:89 –100
29 Loube DI. Treatment algorithm for OSA. Chest 1997; 111:
528–529
30 Henderson JH II, Strollo PJ Jr. Medical management of
obstructive sleep apnea. Prog Cardiovasc Dis 1999; 41:377–
386
31 Smith I, Lasserson TJ, Wright J. Drug therapy for obstructive
sleep apnoea in adults. Cochrane Database Syst Rev 2006;
2:CD003002
32 Abad VC, Guilleminault C. Pharmacological management of
sleep apnoea. Expert Opin Pharmacother 2006; 7:11–23
682 Original Research
33 Bliwise DL, Benkert RE, Ingham RH. Factors associated
with nightly variability in sleep-disordered breathing in the
elderly. Chest 1991; 100:973–976
34 Pendlebury ST, Pe´pin JL, Veale D, et al. Natural evolution of
moderate sleep apnoea syndrome: significant progression
over a mean of 17 months. Thorax 1997; 52:872–878
35 Hedner J, Kraiczi H, Peker Y, et al. Reduction of sleep apnea
after the orally available cholinesterase inhibitor donepezil
[abstract]. Sleep Med 2005; 6:S54 –S55
36 Haxhiu MA, Cherniack NS, Mitra J, et al. Nonvagal
modulation of hypoglossal neural activity. Respiration
1992; 59:65–71
37 Willette RN, Doorley BM, Sapru HN. Activation of cholin-
ergic mechanisms in the medulla oblongata reverse intrave-
nous opioid-induced respiratory depression. J Pharmacol Exp
Ther 1987; 240:352–358
38 Liu X, Sood S, Liu H, et al. Opposing muscarinic and
nicotinic modulation of hypoglossal motor output to genio-
glossus muscle in rats in vivo. J Physiol 2005; 565:965–980
39 Jokic R, Klimaszewski A, Mink J, et al. Surface tension forces
in sleep apnea: the role of a soft tissue lubricant: a random-
ized double-blind, placebo-controlled trial. Am J Respir Crit
Care Med 1998; 157:1522–1525
40 Hedner E, Birkhed D, Hedner J, et al. Stimulation of minor
salivary glands by intraoral treatment with the cholinesterase
inhibitor physostigmine in man. Eur J Oral Sci 2001; 109:
371–374
41 Fitzgerald RS, Shirahata M, Wang HY. Acetylcholine is
released from in vitro cat carotid bodies during hypoxic
stimulation. Adv Exp Med Biol 2000; 475:485–494
42 Kubin L, Kimura H, Tojima H, et al. Suppression of hypo-
glossal motoneurons during the carbachol-induced atonia of
REM sleep is not caused by fast synaptic inhibition. Brain Res
1993; 611:300 –312
43 Martin SE, Brander PE, Deary IJ, et al. The effect of
clustered versus regular sleep fragmentation on daytime
function. J Sleep Res 1999; 8:305–311
44 Phillips AJ, Robinson PA. A quantitative model of sleep-wake
dynamics based on the physiology of the brainstem ascending
arousal system. J Biol Rhythms 2007; 22:167–179
www.chestjournal.org CHEST / 133/3/MARCH, 2008 683
... 25,26 Arousal threshold Eszopiclone, zolpidem, and zopiclone were studied and showed no difference in AHI from placebo. 27 [38][39][40] Risk of bias assessment ...
Article
Full-text available
Objective Summarize the evidence on drug therapies for obstructive sleep apnea. Methods The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were followed. PubMed, Embase, Scopus, Web of Science, SciELO, LILACS, Scopus, Cochrane Central Register of Controlled Trials, and ClinicalTrials.gov were searched on February 17th, 2023. A search strategy retrieved randomized clinical trials comparing the Apnea-Hypopnea Index (AHI) in pharmacotherapies. Studies were selected and data was extracted by two authors independently. The risk of bias was assessed using the Cochrane Risk of Bias tool. RevMan 5.4. was used for data synthesis. Results 4930 articles were obtained, 68 met inclusion criteria, and 29 studies (involving 11 drugs) were combined in a meta-analysis. Atomoxetine plus oxybutynin vs placebo in AHI mean difference of -7.71 (-10.59, -4.83) [Fixed, 95 % CI, I2 = 50 %, overall effect: Z = 5.25, p < 0.001]. Donepezil vs placebo in AHI mean difference of -8.56 (-15.78, -1.33) [Fixed, 95 % CI, I2 = 21 %, overall effect: Z = 2.32, p = 0.02]. Sodium oxybate vs placebo in AHI mean difference of -5.50 (-9.28, -1.73) [Fixed, 95 % CI, I2 = 32 %, overall effect: Z = 2.86, p = 0.004]. Trazodone vs placebo in AHI mean difference of -12.75 (-21.30, -4.19) [Fixed, 95 % CI, I2 = 0 %, overall effect: Z = 2.92, p = 0.003]. Conclusion The combination of noradrenergic and antimuscarinic drugs shows promising results. Identifying endotypes may be the key to future drug therapies for obstructive sleep apnea. Moreover, studies with longer follow-up assessing the safety and sustained effects of these treatments are needed. PROSPERO registration number CRD42022362639.
... The selection process is described in Fig. 1. Finally, 29 studies were included in the review (Supplement Table S1): 29 studies [26e53] included AHI data, see Table S2, 4 studies [31,32,40,44] contained ODI data, see Table S3, 12 studies [28,30e32,36,40,44e46,48,50,53] reported mean saturation (Table S4) and 15 studies [26,28e30,32,34,37,39,41,42,45,46,49,50] ESS data (Table S5). ...
Article
Full-text available
Objective: New drug treatments are under development in obstructive sleep apnea (OSA). The placebo effect is well recognized in various conditions, but its relevance in OSA is debated. In the current study we determined the influence of a placebo effect in studies of drug therapy in OSA. Methods: A systematic review and meta-analysis (PROSPERO CRD42021229410) with searches in MEDLINE, Scopus, Web of Science and Cochrane CENTRAL from inception to 2021-01-19. Inclusion criteria were (i) RCTs of adults with OSA, (ii) drug intervention with placebo baseline and follow-up sleep study (iii) outcomes: apnea hypopnea index (AHI), mean oxygen saturation (mSaO2), oxygen desaturation index (ODI) and/or Epworth Sleepiness Scale (ESS). Risk-of-bias was assessed with Cochrane RoB 2. Results: 7436 articles were identified and 29 studies included (n = 413). Studies were generally small (median n = 14), with 78% men, baseline AHI range 9-74 events/h and treatment duration range 1-120 days. Meta-analyses were conducted for main outcomes. Mean change of the primary outcome, AHI, was -0.84 (95% CI -2.98 to 1.30); mSaO2 and ODI estimations were also non-significant. ESS showed a trend towards a reduction of -1 unit. Subgroup analysis did not show significant differences. Risk-of-bias assessment indicated mostly low risk but studies were small with wide confidence intervals. Conclusions: In this meta-analysis we did not identify systematic placebo effects on the AHI, ODI or mSaO2 while ESS score showed a trend for a small reduction. These results have an impact on the design and interpretation of drug trials in OSA.
... Notably, three randomized, double-blind, placebo-controlled human studies have shown that OSA severity can be reduced by administration of acetylcholinesterase inhibitors [49][50][51] . Nevertheless, the improvements were variable, likely reflecting the multi-factorial nature of OSA and the varying contributions of the four major endotypes operating within and between patients: pharyngeal anatomy, pharyngeal dilator muscle activity, arousal threshold and loop gain [52][53][54][55] . ...
Article
Full-text available
Successful cholinergic-noradrenergic pharmacotherapy for obstructive sleep apnea (OSA) is thought to be due to effects at the hypoglossal motor nucleus (HMN). Clinical efficacy varies with muscarinic-receptor (MR) subtype affinities. We hypothesized that oxybutynin (cholinergic agent in successful OSA pharmacotherapy) is an effective MR antagonist at the HMN and characterized its efficacy with other antagonists. We recorded tongue muscle activity of isoflurane anesthetized rats (121 males and 60 females, 7–13 per group across 13 protocols) in response to HMN microperfusion with MR antagonists with and without: (i) eserine-induced increased endogenous acetylcholine at the HMN and (ii) muscarine. Eserine-induced increased acetylcholine decreased tongue motor activity (p < 0.001) with lesser cholinergic suppression in females versus males (p = 0.017). Motor suppression was significantly attenuated by the MR antagonists atropine, oxybutynin, and omadacycline (MR2 antagonist), each p < 0.001, with similar residual activity between agents (p ≥ 0.089) suggesting similar efficacy at the HMN. Sex differences remained with atropine and oxybutynin (p < 0.001 to 0.05) but not omadacycline (p = 0.722). Muscarine at the HMN also decreased motor activity (p < 0.001) but this was not sex-specific (p = 0.849). These findings have translational relevance to antimuscarinic agents in OSA pharmacotherapy and understanding potential sex differences in HMN suppression with increased endogenous acetylcholine related to sparing nicotinic excitation.
Article
Introduction: This systematic review and meta-analysis evaluates the evidence from randomized controlled trials (RCTs) involving pharmacological interventions for improving sleep in people with Alzheimer's disease (AD). Methods: A systematic literature search in eight databases from January 2000 to July 2023 focusing on RCTs that compared a pharmacological intervention with a placebo for enhancing sleep in people with AD. The authors registered the study protocol at Prospero, followed the PRISMA guidelines, and produced the pooled estimates using random-effect or IVhet models. Results: Eight different interventions and 29 different sleep outcomes were examined in 14 RCTs included in this review. Eszopiclone positively affected sleep efficiency, as did orexin antagonists. However, there was no difference when melatonin was used. The interventions demonstrated low discontinuation rates and a few adverse drug reactions. Conclusion: Although melatonin was the most investigated intervention, the evidence for its efficacy is inconclusive. On the other hand, trazodone and orexin receptor antagonists showed promising results; however, more RCTs are needed for definite answers.
Article
Full-text available
Due to worldwide demographic change, the number of older persons in the population is increasing. Aging is accompanied by changes of sleep structure, deposition of beta-amyloid (Aß) and tau proteins and vascular changes and can turn into mild cognitive impairment (MCI) as well as dementia. Sleep disorders are discussed both as a risk factor for and as a consequence of MCI/dementia. Cross-sectional and longitudinal population-based as well as case–control studies revealed sleep disorders, especially sleep-disorderded breathing (SDB) and excessive or insufficient sleep durations, as risk factors for all-cause MCI/dementia. Regarding different dementia types, SDB was especially associated with vascular dementia while insomnia/insufficient sleep was related to an increased risk of Alzheimer’s disease (AD). Scarce and still inconsistent evidence suggests that therapy of sleep disorders, especially continuous positive airway pressure (CPAP) in SDB, can improve cognition in patients with sleep disorders with and without comorbid dementia and delay onset of MCI/dementia in patients with sleep disorders without previous cognitive impairment. Regarding potential pathomechanisms via which sleep disorders lead to MCI/dementia, disturbed sleep, chronic sleep deficit and SDB can impair glymphatic clearance of beta-amyloid (Aß) and tau which lead to amyloid deposition and tau aggregation resulting in changes of brain structures responsible for cognition. Orexins are discussed to modulate sleep and Aß pathology. Their diurnal fluctuation is suppressed by sleep fragmentation and the expression suppressed at the point of hippocampal atrophy, contributing to the progression of dementia. Additionally, sleep disorders can lead to an increased vascular risk profile and vascular changes such as inflammation, endothelial dysfunction and atherosclerosis which can foster neurodegenerative pathology. There is ample evidence indicating that changes of sleep structure in aging persons can lead to dementia and also evidence that therapy of sleep disorder can improve cognition. Therefore, sleep disorders should be identified and treated early.
Article
Full-text available
Obstructive sleep apnea (O.S.A.), also referred to as obstructive sleep hypopnea – is a sleep disorder that involvescessation or significant decrease in airflow in the presence of breathing effort. It is the most common type of sleep-disorderedbreathing. It is characterized by episodes of total or partial airway collapse and decreased oxygen saturation or sleep arousal. Asa result of this disturbance, sleep is interrupted and non-restorative. It significantly raises morbidity and mortality in both developedand developing countries. This review aims to review relevant studies on obstructive sleep apnea. The method used is acomputerized search method, the pathophysiology, clinical presentation, complications, diagnostic procedures, and therapeuticchoices from the literature. The effects of O.S.A. on cardiovascular health, mental disease, quality of life, and driving safety aresevere. This course of action highlights the interprofessional team's involvement in treating obstructive sleep apnea, itspathophysiology, and its causes.
Article
Introduction: Alzheimer's disease (AD) is one of the most common neurodegenerative disorders among the older population. Sleep disruption and circadian rhythm disorders often develop in AD patients, and many experience sleeping difficulties requiring pharmacological and non-pharmacological interventions. Areas covered: This review appraised the evidence from clinical studies on various pharmacological and non-pharmacological therapies for sleep disturbances in AD patients and proposed an algorithm to manage sleep disturbances in this population of patients. Expert opinion: Non-pharmacological interventions are generally preferred as the first-line approach to improve sleep-related symptoms in AD due to their favorable safety profile. However, when non-pharmacological interventions alone are insufficient, a range of pharmacological agents can be considered. Trazodone and melatonin are commonly used as adjunctive therapies, while Z-drugs including zopiclone and zolpidem are specifically employed to treat insomnia in patients with late-onset AD. Furthermore, a newer class of agents known as dual orexin receptor antagonists has emerged and gained approval for improving sleep onset and maintenance in AD patients.
Article
Full-text available
Chronische Atemwegserkrankungen stellen eine weltweite Epidemie dar. Ihre Inzidenz und Prävalenz in der Weltbevölkerung nimmt zu, und insbesondere bei älteren Menschen treten sie häufig in Verbindung mit anderen Krankheiten auf, was häufig zu einen Status hoher klinischer Komplexität. Fachärzte für Neurologie, Innere Medizin und Pneumologen sollten sich des gemeinsamen Hintergrunds dieser Erkrankungen bewusst sein, um den komorbiden Zustand des Patienten richtig zu behandeln und die Therapie unter Berücksichtigung möglicher Überschneidungen zu optimieren. In dieser Übersichtsarbeit konzentrieren wir uns auf die Beziehungen zwischen chronischen Atemwegserkrankungen und chronischen neurodegenerativen Erkrankungen auf verschiedenen Ebenen; wir untersuchen die gemeinsamen Risikofaktoren und die Wechselwirkungen zwischen den Erkrankungen, die Indikationen für die Untersuchung der Atemfunktion bei neurodegenerativen Erkrankungen und die Wechselwirkungen zwischen Pathologie und Pathologie sowie zwischen Medikamenten und Pathologie bei Patienten, die sowohl von chronischen neurologischen als auch von respiratorischen Krankheiten betroffen sind.
Article
Context Apolipoprotein E ∊4 (ApoE ∊4) is a well-known risk factor for Alzheimer disease and cardiovascular disease. Sleep-disordered breathing occurs in Alzheimer disease patients and increases risks for cardiovascular disease. Complex interactions among sleep, brain pathology, and cardiovascular disease may occur in ApoE ∊4 carriers.Objective To study whether genetic variation at the level of ApoE is associated with sleep-disordered breathing or sleep abnormalities in the general population.Design, Setting, and Participants Ongoing longitudinal cohort study of sleep disorders at a US university beginning in 1989, providing a population-based probability sample of 791 middle-aged adults (mean [SD] age, 49 [8] years; range, 32-68 years).Main Outcome Measure Nocturnal polysomnography to evaluate apnea-hypopnea index.Results The probability of moderate-to-severe sleep-disordered breathing (apnea-hypopnea index ≥15%) was significantly higher in participants with ∊4, independent of age, sex, body mass index, and ethnicity (12.0% vs 7.0%; P = .003). Mean (SEM) apnea-hypopnea index was also significantly higher in participants with ApoE ∊4 (6.5 [0.6] vs 4.8 [0.3]; P = .01). These effects increased with the number of ApoE ∊4 alleles carried.Conclusions A significant portion of sleep-disordered breathing is associated with ApoE ∊4 in the general population.
Article
Upper airway dilating muscle activity is characterized by an early-peaking pattern which serves to dilate or stiffen the upper airway at the time when the greatest negative intraluminal pressure is generated by contraction of chest wall muscles. This pattern has been attributed to phasic afferent inputs from pulmonary stretch receptors. The present study examines the hypothesis that nonvagal factors may also influence the discharge pattern and coordination of upper airway and chest wall muscle activities. Therefore, in anesthetized, paralyzed, vagotomized and artificially ventilated cats, we examined the effects of changes in respiratory drive produced by activation of cholinergic and GA-BAergic (gamma-aminobutyric acid) receptors at the ventrolateral aspects of the medulla oblongata on phasic intrabreath discharge patterns of hypoglossal and phrenic nerves. Cholinergic agents (acetylcholine, carbachol, methacholine, physostigmine) applied directly to chemoreceptive areas on the ventral medullary surface increased hypoglossal activity, and in addition converted inspiratory discharge from an augmenting to a decrementing pattern of activity. The reverse effect on the discharge pattern of hypoglossal activity was observed with a decrease in respiratory drive. While the amplitude of the phrenic nerve discharge was also affected by these interventions, the augmenting discharge pattern of phrenic nerve activity did not change. These results suggest that the early peaking pattern of hypoglossal nerve discharge in vagotomized cats also depends on the level of respiratory drive, and is not solely dependent on vagal afferent inputs. In addition, the data suggest that structures near the ventral surface of the medulla are influential in shaping the pattern of hypoglossal nerve activity and maintaining balanced activity of upper airway and chest wall muscles.