Vous êtes sur la page 1sur 7

ORIGINAL ARTICLE

Impact of chronic rhinosinusitis on sleep: a controlled clinical study


Jeremiah A. Alt, MD, PhD1 , Vijay R. Ramakrishnan, MD2 , Michael P. Platt, MD3 ,
Rodney J. Schlosser, MD4 , Tina Storck, MS4 and Zachary M. Soler, MD, MSc4

Background: Earlier studies have suggested that pa- p = 0.006). On home sleep studies, patients with CRS were
tients with chronic rhinosinusitis (CRS) report worse sleep found to have an increased number of awakenings during
quality than population norms. What remains unknown is a night’s sleep (8.6 ± 4.8 vs 6.3 ± 3.0; p = 0.004), lower
whether these patients are actually experiencing measur- average overnight oxygen saturation (93.2 ± 2.6% vs 94.3
able changes in objective sleep parameters. The goal of this ± 2.1%; p = 0.042), increased rapid eye movement sleep
study was to prospectively evaluate objective sleep mea- (REMS) latency (93.0 ± 67.1 vs 66.7 ± 35.3; p = 0.016), and
sures in a cohort of patients with CRS. spent a greater portion of the night snoring at >40 dB (24.7
± 27.4% vs 14.6 ± 19.7%; p = 0.034). All differences except
Methods: A prospective, multi-institutional, case-control mean oxygen saturation remained significant aer control-
study was designed to compare patients with CRS to ling for baseline differences.
nondiseased controls. Sleep quality was measured using
the Pisburgh Sleep Quality Index (PSQI) and the Epworth Conclusion: Differences in both patient-reported and ob-
Sleepiness Scale (EpSS). Home sleep studies were admin- jective sleep measures exist between patients with CRS
istered across all subjects and a baery of objective sleep and controls. 
C 2018 ARS-AAOA, LLC.

measurements were recorded using a portable sleep diag-


nostic device. Regression models were used to control for Key Words:
any factors that differed across groups. chronic rhinosinusitis; disease severity; patient-reported
outcome measure
Results: A total of 108 subjects were enrolled across 4 in-
stitutions, including 52 patients with CRS and 56 controls.
Total PSQI scores were worse in patients with CRS when How to Cite this Article:
compared with controls (10.1 ± 4.3 vs 4.7 ± 2.5; p < 0.001). Alt JA, Ramakrishnan VR, Pla MP, et al. Impact of chronic
Similarly, daytime somnolence, as measured by the EpSS, rhinosinusitis on sleep: a controlled clinical study. Int
was greater in patients with CRS (9.1 ± 5.3 vs 6.5 ± 3.7; Forum Allergy Rhinol. 2019;9:16–22.

persists beyond 3 months.1, 2 The cardinal symptoms of


C hronic rhinosinusitis (CRS) impacts 5–10% of the
United States population and is characterized by on-
going mucosal inflammation of the paranasal sinuses that
CRS include nasal congestion, discolored nasal drainage,
facial pain/pressure, and smell loss, but a wide body of
evidence supports impacts that extend beyond just the
nose and sinuses. A number of studies have examined
1 Sinusand Skull Base Surgery Program, Division of the impact of CRS on sleep quality.3–7 From a quality-
Otolaryngology–Head and Neck Surgery, Department of Surgery, of-life (QOL) standpoint, sleep dysfunction is a specific
University of Utah, Salt Lake City, UT; 2 Department of Otolaryngology, domain of the widely utilized 22-item Sino-Nasal Out-
University of Colorado, Aurora, CO; 3 Department of Otolaryngology,
comes Test (SNOT-22) QOL instrument. Differences in
Boston University, Boston, MA; 4 Division of Rhinology and Sinus
Surgery, Department of Otolaryngology–Head & Neck Surgery, sleep scores contribute to baseline QOL deficits in pa-
Medical University of South Carolina, Charleston, SC tients with CRS and appear to influence the likelihood
Correspondence to: Zachary M. Soler, MD, MSc, Division of Rhinology and with which those patients choose to undergo sinus surgery.8
Sinus Surgery, Department of Otolaryngology–Head and Neck Surgery, A number of studies have explored patient-reported sleep
Medical University of South Carolina, 135 Rutledge Avenue, Charleston, SC
29425; e-mail: solerz@musc.edu quality in more detail, often utilizing the Pittsburgh Sleep
Funding sources for the study: American Academy of Otolaryngic Allergy;
Quality Index (PSQI).9 These studies universally found
American Rhinologic Society. that patients with CRS report sleep quality to be signif-
Received: 2 August 2018; Revised: 1 September 2018; Accepted: icantly worse than population norms, often on par with
5 September 2018 other chronic diseases and even sleep-specific disorders such
DOI: 10.1002/alr.22212
View this article online at wileyonlinelibrary.com. as obstructive sleep apnea or narcolepsy.6 Furthermore,

16 International Forum of Allergy & Rhinology, Vol. 9, No. 1, January 2019


Impact of CRS on sleep

patient-reported sleep quality scores have been shown to accompanying patients to otolaryngology appointments
improve after both medical and surgical treatment of CRS, at the same institution, such that both cases and controls
suggesting that the sleep dysfunction experienced by pa- derived from the same source population. Theoretically,
tients is related to their underlying CRS.3, 6, 7 this could include bed partners of patients with CRS,
Although patient-reported sleep quality is clearly worse although specific relationships and sleeping arrangements
in patients with CRS, what remains unknown is whether were not queried. All subjects provided written informed
these patients are actually experiencing measurable, objec- consent and the study protocol was approved by the
tive changes in sleep. From a mechanistic standpoint, it institutional review boards at all contributing sites.
is certainly plausible that sinonasal inflammation related to Demographic information and history of comorbidities,
CRS may disturb sleep physiology. Simplistically, this could such as asthma, aspirin intolerance, allergies, and depres-
be related to nasal airway blockage, altering the balance of sion, was collected using standardized questionnaires for all
nasal and oral airflow and contributing to snoring and/or participants. Tobacco and alcohol intake was recorded as
sleep apnea. Alternatively, it remains possible that rhino- cigarettes/day and drinks/week, respectively. Allergic rhini-
genic symptoms, such as nasal congestion and postnasal tis was established for those who carried a physician’s di-
drainage, impact sleep quality by interfering with the sleep agnosis and had confirmation with previous positive ob-
cycle via frequent awakenings. Inflammatory cytokines as- jective testing. Height and weight data were obtained for
sociated with CRS could also directly impact sleep via neu- each individual and converted into body mass index (BMI).
roimmune signaling.10 Last, it is possible that measurable, Current use of antihistamines, antidepressants, anxiolytics,
objective changes in sleep are not occurring, despite patient- and antipsychotics was recorded. Any subject with a pre-
reported deficits in sleep quality. viously diagnosed primary sleep disorder (obstructive sleep
It is important to understand whether CRS patients truly apnea, narcolepsy, insomnia) was excluded from the study.
suffer from objective sleep dysfunction so we may under- Control subjects were excluded if they reported a history of
stand the potential for significant psychosocial and health CRS or recurrent sinus infections, or met symptoms criteria
implications. The goal of this study was to prospectively for CRS according to EPOS12.11
evaluate objective sleep measures in a cohort of patients
with CRS, comparing findings with those from a control Measures of CRS
population. We hypothesized that patients with CRS would
Patients and controls completed the SNOT-22, which re-
have alterations in objective measures associated with the
quires patients to rate 22 symptom-related items on a scale
sleep cycle. Secondary goals were to evaluate differences
ranging from 0 (no problem) to 5 (problem is as bad as
in subgroups defined by polyp status and control for re-
it can be).12 Nasal endoscopy was used to categorize each
lated factors more common in CRS patients, which may
patient into CRS with polyps (CRSwNP) and CRS with-
confound the analysis.
out polyps (CRSsNP) and was also scored using the Lund-
Kennedy (LK) staging system.13 Computed tomography
Patients and methods (CT) scans of patients with CRS were scored using the
Lund-Mackay (LM) scoring method.14
This study was implemented as a prospective, multi-
institutional, case-control study. Adults (ࣙ18 years) with
medically refractory CRS were enrolled from tertiary rhi- Evaluation of patient-reported sleep quality,
nology clinics at 4 academic centers: the Medical Univer- fatigue, daytime somnolence, depression, and pain
sity of South Carolina (MUSC; Charleston, SC); the Uni- All study participants completed the PSQI, a self-reported
versity of Utah (UU; Salt Lake City, Utah); the University questionnaire measuring sleep quality and disturbance over
of Colorado (CU; Aurora, Colorado); and Boston Univer- the preceding 1-month period.9 The PSQI yields 7 compo-
sity (BU; Boston, Massachusetts). The presence of CRS was nent or subdomain scores: subjective sleep quality; sleep
diagnosed using criteria from the the European Position Pa- latency; sleep duration; sleep efficiency; sleep disturbance;
per on Rhinosinusitis and Nasal Polyps 2012 (EPOS12).11 sleep medication usage; and daytime dysfunction. Subdo-
Those patients who were considered surgical candidates main component scores (range, 0–3) are assessed using an
were offered the option to undergo ESS after failing ini- openly available scoring algorithm and added to obtain
tial attempts at appropriate medical treatment. The ex- a total score (range, 0–21), with higher scores indicating
tent of attempted medical treatments varied, but to be in- worse sleep quality. The level of fatigue experienced by
cluded all patients must have failed initial medical man- the subject was assessed using the Fatigue Severity Scale
agement consisting of at least one course of oral steroid, (FSS).15, 16 The FSS is a valid, reliable, and internally consis-
broad-spectrum, or culture-directed antibiotics for at least tent instrument that measures fatigue over the last 4 weeks
14 days, and a minimum 3 weeks of topical steroid sprays. (range, 1–7), with higher scores representing more fatigue.
To be eligible, patients with CRS must have been off oral The Epworth Sleepiness Scale (EpSS) was utilized to mea-
steroids for ࣙ1 month before enrollment. sure a person’s general level of daytime sleepiness, giving
Control subjects (ࣙ18 years) without any history a sleep propensity score ranging from 0 to 24.17 The po-
of sinusitis were recruited from among individuals tential presence of depression was determined using the

International Forum of Allergy & Rhinology, Vol. 9, No. 1, January 2019 17


Alt et al.

Patient Health Questionnaire-2 (PHQ2), which measures TABLE 1. Characteristics of the study population
frequency of depressed mood and anhedonia over the pre-
ceding 2 weeks, with higher scores representing depressed CRS (n = 52) Control (n = 56) p value
mood.18
Demographics
Age 45.6 (14.1) 43.9 (15.1) 0.564
Evaluation of objective sleep measures
All subjects were given a WatchPAT device for home sleep Sex Male 32 (61.5%) 24 (42.9%) 0.052
testing (Itamar Medical, Inc, Franklin, MA). A dedicated Female 20 (38.5%) 32 (57.1%)
study coordinator gave detailed instruction on how to ad-
Race White 42 (80.8%) 48 (85.7%) 0.701
minister the home sleep test and subjects were encour-
aged to choose a night where they follow their typical Black 9 (17.3%) 6 (10.7%)
nightly routine. WatchPAT is a United States Food and Other 1 (1.9%) 2 (3.6%)
Drug Administration–approved, wrist-worn device with 2
BMI 28.9 (6.6) 26.3 (4.9) 0.021
sensors, which attach to the ipsilateral fingers, and an in-
tegrated snoring sensor, which allows measurement of pe- Exposures
ripheral arterial tone (PAT), pulse rate, oxygen saturation, Cigarettes/day 0.3 (1.5) 0.5 (2.9) 0.603
actigraphy, and snoring.19–21 Measurements for Respira-
tory Disturbance Index (RDI) and Apnea-Hypopnea Index Alcoholic drinks/week 2.8 (5.7) 2.3 (3.2) 0.591
(AHI) was done by simultaneously measuring PAT, heart Comorbidities
rate, and oxygen saturation. There have been numerous Asthma No 33 (63.5%) 56 (100.0%) <0.001
direct comparisons to formal in-office polysomnography
(PSG), showing excellent correlation between WatchPAT Yes 19 (36.5%) 0 (0.0%)
RDI/AHI and in-office PSG (r = 0.82-0.89).20–23 Measures Allergic rhinitis No 24 (46.2%) 48 (85.7%) <0.001
of sleep architecture were recorded, including sleep latency,
Yes 28 (53.9%) 8 (14.3%)
total sleep time, and awakenings, as well the percentage of
time in stages of rapid eye movement (REM) and non- Immunodeficiency No 51 (98.1%) 56 (100.0%) 0.481
REM (NREM) sleep. Direct comparisons of WatchPAT to Yes 1 (1.9%) 0 (0.0%)
formal in-office PSG have shown excellent epoch-by-epoch
AERD No 49 (94.2%) 55 (98.2%) 0.350
agreements ranging from 80% to 90%.19, 24, 25 Last, the
percentage of sleep spent snoring at >40 decibels (dB) was Yes 3 (5.8%) 1 (1.8%)
recorded. GERD No 42 (80.8%) 51 (91.1%) 0.122
Yes 10 (19.2%) 5 (8.9%)
Statistical analysis
Depression (MD diagnosis) No 43 (82.7%) 51 (91.1%) 0.195
All data recorded were collected on standardized forms
and entered into a REDCap database using double data- Yes 9 (17.3%) 5 (8.9%)
entry processes to ensure data integrity. Data from each Migraine No 41 (78.9%) 50 (89.3%) 0.137
center were then merged and analysis was performed us-
Yes 11 (21.2%) 6 (10.7%)
ing SPSS version 24.0 (IBM Corporation, Armonk, NY).
Demographic information, comorbidities, patient-reported Pain disorder No 49 (94.2%) 53 (94.6%) 1.000
metrics, and objective sleep parameters were assessed using Yes 3 (5.8%) 3 (5.4%)
descriptive statistics. Continuous variables were compared
between 2 cases and controls using the independent t test Medications
or Mann-Whitney U test. Categorical variables were com- Antihistamines (oral) No 30 (60.0%) 50 (92.6%) <0.001
pared between 2 groups using the Pearson’s chi-square test
Yes 20 (40.0%) 4 (7.4%)
or Fisher’s exact test. Linear, Poisson, and beta-regression
models were created to evaluate for statistical differences Antidepressants No 42 (82.4%) 46 (86.8%) 0.530
while controlling for confounding factors. Resulting regres- Yes 9 (17.7%) 7 (13.2%)
sion coefficients (β) and standard errors (SEs) were used to
Anxiolytics No 50 (98.0%) 53 (98.2%) 1.000
estimate linear changes.
Yes 1 (2.0%) 1 (1.9%)
Antipsychotics No 51 (100.0%) 54 (100.0%) NA
Results
*
Data expressed as mean (standard deviation) or as number (%)
A total of 108 subjects were enrolled across 4 institutions, AERD = aspirin-exacerbated respiratory disease; BMI = body mass index;
including 52 patients with CRS and 56 controls (MUSC GERD = gastroesophageal reflux disease; NA = not applicable.
= 41, UU = 30, CU = 21, BU = 16; Table 1). There
were no differences in age, gender, or race across groups.

18 International Forum of Allergy & Rhinology, Vol. 9, No. 1, January 2019


Impact of CRS on sleep

TABLE 2. Patient-reported sleep quality, fatigue, TABLE 3. Objective sleep measures from home sleep test
and daytime somnolence
Cohort N Mean (SD) p value
Cohort N Mean (SD) p value
Respiratory disturbance Index CRS 48 19.9 (14.9) 0.516
PSQI Total CRS 50 10.1 (4.3) <0.001
Control 52 18.1 (12.4)
Control 55 4.7 (2.5)
Apnea-Hypopnea Index CRS 48 14.4 (15.2) 0.431
FSS Total CRS 48 3.9 (1.5) <0.001
Control 52 12.2 (13.0)
Control 55 2.5 (1.3)
Average overnight oxygen CRS 48 93.3 (2.6) 0.042a
EpSS Total CRS 49 9.1 (5.3) 0.006 saturation
Control 55 6.5 (3.8) Control 52 94.3 (2.1)
PHQ-2 Total CRS 50 1.8 (1.7) <0.001 Mean pulse rate CRS 50 68.3 (11.0) 0.112
Control 55 0.4 (0.7) Control 52 64.9 (10.3)

SD = standard deviation; PSQI = Pittsburgh Sleep Quality Index; FSS = Fa- Sleep latency (minutes) CRS 51 22.3 (12.9) 0.285
tigue Severity Scale; EpSS = Epworth Sleepiness Scale; PHQ-2 = Patient Health
Questionnaire-2; MPQ-SF = McGill Pain Questionnaire short form. Control 52 19.9 (8.7)
SD = standard deviation.
REM latency (minutes) CRS 50 93.0 (67.1) 0.016a
Control 52 66.7 (35.3)
Patients with CRS had more asthma and allergic rhinitis
Number of wakes CRS 50 8.6 (4.8) 0.004a
compared with controls (p < 0.001). Average BMI was
higher in those with CRS when compared with controls Control 52 6.3 (3.0)
(28.9 ± 6.6 vs 26.3 ± 4.9; p = 0.021). As expected, SNOT- Total sleep time (minutes) CRS 51 367.9 (72.5) 0.786
22 scores were significantly higher in CRS patients than in
controls (54.0 ± 19.8 vs 10.3 ± 9.0; p < 0.001). Patients Control 52 364.0 (72.8)
with CRS were evenly split between CRSwNP (n = 27; Percent of sleep while snoring CRS 51 24.7 (27.4) 0.034a
52%) and CRSsNP (n = 25; 48%), with an average LM at >40 dB
CT score of 12.8 ± 6.0 and LK endoscopy score of 6.5 ± Control 52 14.6 (19.7)
3.3.
Patient-reported outcomes metrics were significantly dif- REM percent CRS 49 25.5 (11.3) 0.936
ferent between cases with CRS and controls (Table 2). As Control 52 25.3 (6.5)
expected, PSQI scores were worse in patients with CRS Light sleep percent CRS 49 56.0 (11.4) 0.798
compared with controls (10.1 ± 4.3 vs 4.7 ± 2.5; p <
0.001). There were also differences in EpSS, with greater Control 52 56.5 (10.2)
daytime somnolence in patients with CRS (9.1 ± 5.3 vs 6.5 Deep sleep percent CRS 49 19.1 (6.1) 0.956
± 3.7; p = 0.006). Fatigue (FSS) and depression (PHQ-
Control 52 19.2 (9.4)
2) scores were also significantly worse in CRS patients
vs controls (p < 0.001 for all). No difference in patient- CRS = chronic rhinosinusitus; REM = rapid eye movement; SD = standard devi-
ation.
reported metrics were seen between CRSsNP and CRSwNP a
Statistically significant (p < 0.05).
subgroups (p > 0.08 for all).
Differences were seen between cases and controls for a
number of objective sleep parameters (Table 3). Patients were seen in objective sleep parameters between CRS
with CRS were found to have an increased number of subgroups.
awakenings during a night’s sleep compared with controls Linear regression was then used to control for factors that
(8.6 ± 4.8 vs 6.3 ± 3.0; p = 0.004), as well as a lower may differentially impact sleep in CRS compared with con-
average overnight oxygen saturation (93.2 ± 2.6% vs 94.3 trols (Tables 4 and 5). In this study, those with CRS were
± 2.1%; p = 0.042). Increased REM sleep latency was also more likely to be male and have higher BMI than controls, 2
seen in patients with CRS when compared with controls factors that could potentially impact sleep in these groups
(93.0 ± 67.1 vs 66.7 ± 35.3; p = 0.016). Last, patients with and confound outcomes. However, PSQI, FSS, and ESS
CRS spent a greater portion of the night snoring at >40 dB remained worse in CRS patients compared with controls
(24.7 ± 27.4% vs 14.6 ± 19.7%; p = 0.034). No differ- in regression models adjusted for gender and BMI. Simi-
ences were seen between groups for total sleep time, AHI, larly, sleep latency and number of awakenings remained
RDI, or percentage of time in discrete sleep stages. When significantly increased after adjusting for gender and BMI,
comparing CRS subgroups, REM latency was increased in whereas average overnight oxygen saturation and percent-
those with CRSsNP when compared with CRSwNP (104.3 age of the night snoring at >40 dB no longer reached
± 48.2 vs 80.7 ± 22.1; p = 0.032). No other differences statistical significance. Last, in patients with CRS, no

International Forum of Allergy & Rhinology, Vol. 9, No. 1, January 2019 19


Alt et al.

TABLE 4. Linear regressions of CRS and outcomes adjusting for gendera

Unstandardized coefficients 95% CI for β

Model β SE t p value Lower bound Upper bound

PSQI CRS 5.574 0.682 8.169 <0.001 4.221 6.928


Genderc 1.232 0.682 1.807 0.074 −0.120 2.585
FSS CRS 1.449 0.278 5.211 <0.001 0.897 2.001
Gender 0.130 0.277 0.470 0.639 −0.420 0.681
ESS CRS 2.556 0.907 2.819 0.006 0.758 4.355
Gender −0.093 0.906 −0.102 0.919 −1.890 1.704
PHQ-2 d
CRS 1.511 0.241 e
39.281 (1) <0.001 1.038 1.983
Gender 0.157 0.194 e
0.661 (1) 0.416 −0.222 0.537
Average overnight oxygen saturation CRS −0.814 0.480 −1.695 0.093 −1.766 0.139
Gender 0.820 0.480 1.708 0.091 −0.133 1.772
REM latency CRS 25.574 10.775 2.373 0.020 4.194 46.953
Gender −4.593 10.775 −0.426 0.671 −25.973 16.786
Number of wakes CRS 2.377 0.807 2.947 0.004 0.777 3.977
Gender 0.130 0.807 0.161 0.872 −1.471 1.731
Percent of sleep while CRS 0.372 0.208 1.79 0.076 −0.039 0.784
snoring at >40 dB f
Gender 0.040 0.207 0.200 0.846 −0.370 0.451
a
Significant CRS variables indicate that those with CRS have worse scores than those without for that particular outcome, after adjusting for gender.
b
Statistically significant (p < 0.05).
c
Males are the reference for gender.
d
Poisson regression.
e
Wald chi-square (degrees of freedom).
f
Beta regression.
CI = confidence interval; CRS = chronic rhinosinusitus; EpSS = Epworth Sleepiness Scale; FSS = Fatigue Severity Scale; PHQ-2 = Patient Health Questionnaire-2; PSQI
= Pittsburgh Sleep Quality Index; REM = rapid eye movement.

positive correlation with asthma, allergy, or antihistamine mechanisms for sleep disturbance in CRS. However, one
usage was seen for any of the objective sleep variables that can hypothesize mechanisms by which CRS may result in
differed between CRS and controls. alterations of sleep parameters. Perhaps the most apparent
is increased snoring, which is likely related to nasal con-
gestion, increased nasal resistance, decreased nasal airflow,
Discussion and increased mouth breathing during sleep. The fact that
Findings from this study support a growing body of evi- CRS patients awaken more frequently during the night also
dence showing that patients with CRS have poor subjective makes intuitive sense and mirrors common patient com-
sleep quality compared to similar subjects without CRS. plaints. Nasal congestion, postnasal drainage, and cough-
Furthermore, patients in this study also reported increased ing at night are common in CRS and each could explain
fatigue and daytime somnolence as compared to those with- the increased awakenings seen in CRS compared with con-
out CRS. What has remained unknown is the degree to trols. Furthermore, the high levels of sleep fragmentation,
which these patient-reported symptoms reflect measurable, due to the recurrent awakenings we observed in patients
objective sleep changes or are a manifestation of disease with CRS may explain why patients with CRS are report-
unrelated to actual sleep physiology. Findings from home ing nonrestorative sleep (PSQI, FSS, EpSS), even with ob-
sleep tests suggest that discernible changes in objective sleep served normal total sleep times. The other consistent finding
are present in patients with CRS as compared with nondis- in this study was increased REMS latency compared with
eased controls. These data provide further evidence that controls, but this finding is harder to explain. Depression
the pathophysiologic impacts of CRS extend beyond just can impact REMS latency, but usually this shortens rather
the nose and sinuses and that sleep dysfunction in patients than prolongs latency.26 Primary sleep disorders and psy-
with CRS is a major consequence of ongoing disease. choactive medications can impact REMS latency, but no
The cross-sectional, case-control nature of this study does difference was seen in AHI/RDI between cases and con-
not allow for direct insights regarding causality or specific trols, and subjects with other primary sleep disorders or

20 International Forum of Allergy & Rhinology, Vol. 9, No. 1, January 2019


Impact of CRS on sleep

TABLE 5. Linear regressions of CRS and outcomes adjusting for BMIa

Unstandardized coefficients 95% CI for β

Model β SE t p value Lower bound Upper bound

PSQI CRS 5.138 0.695 7.389 <0.001b 3.759 6.517


BMI 0.086 0.059 1.469 0.145 −0.030 0.203
FSS CRS 1.411 0.283 4.983 <0.001b 0.849 1.973
BMI 0.007 0.024 0.279 0.781 −0.041 0.054
ESS CRS 2.154 0.902 2.389 0.019b 0.366 3.943
BMI 0.151 0.077 1.962 0.053 −0.002 0.304
PHQ-2 c
CRS 1.422 0.243 10.224 (1) d
<0.001 b
0.946 1.899
BMI 0.019 0.015 1.678 (1)d
0.195 −0.010 0.047
Aveeage overnight oxygen saturation CRS −0.734 0.485 −1.515 0.133 −1.696 0.228
BMI −0.081 0.042 −1.950 0.054 −0.164 0.001
b
REM latency CRS 25.125 11.031 2.278 0.025 3.237 47.014
BMI 0.393 0.945 0.415 0.679 −1.483 2.268
b
Number of wakes CRS 1.579 0.762 2.071 0.041 0.066 3.091
b
BMI 0.259 0.066 3.906 0.000 0.127 0.390
Percent of sleep while snoring at >40 CRS 0.207 0.204 1.01 0.315 −0.200 0.614
dBe
BMI 0.059 0.018 3.33 0.001 0.024 0.094
a
Significant CRS variables indicate that those with CRS have worse scores than those without for that particular outcome, after adjusting for BMI.
b
Statistically significant (p < 0.05).
c
Poisson regression.
d
Wald chi-square (degrees of freedom).
e
Beta regression.
BMI = body mass index; CI =confidence interval; CRS = chronic rhinosinusitus; EpSS = Epworth Sleepiness Scale; FSS = Fatigue Severity Scale; PHQ-2 = Patient Health
Questionnaire-2; PSQI = Pittsburgh Sleep Quality Index; REM = rapid eye movement.

taking medications known to impact sleep were excluded tainly, the role of any specific inflammatory cytokine in
from the study. Future investigations using the Multiple sleep dysfunction in CRS is speculative and future studies
Sleep Latency Test (MSLT) could shed further light on this will need to be specifically designed to explore hypothetical
finding, as the MSLT has been used to distinguish hyper- mechanisms.
somnia, narcolepsy, and behavior-induced inadequate sleep The main limitation of this study is its case-control de-
syndrome.27 sign. Although patients and controls were enrolled irrespec-
Refractory CRS is a disease characterized by chronic in- tive of any preconceived notion of sleep quality, one can
flammation and much work has explored local and systemic never be sure that unintentional selection bias did not occur.
inflammatory cytokine profiles.28 On a local level, sinonasal Significant effort was made to assess measures that could
inflammation results in nasal congestion, drainage, and confound the analysis based on preexisting knowledge,
coughing, which could impact sleep as hypothesized earlier. including age, gender, BMI, tobacco/alcohol use, medical
However, circulating inflammatory cytokines could impact comorbidities, and medication usage. There were major
sleep directly, independent of nasal congestion or mucus baseline differences between the CRS and control groups.
production. Studies have shown that fatigue and feelings Some of these differences were expected, such as increased
of sleepiness can be elicited by injection of exogenous cy- asthma and allergic rhinitis in the CRS group. The main
tokines and thus it remains possible that some degree of unexpected difference was that patients with CRS had in-
patient-reported sleep quality and fatigue is a direct conse- creased BMI compared with controls. It remains unknown
quence of the inflammatory, cytokine milieu characteristic whether increased BMI seen in cases is a real consequence of
of CRS.10 These same cytokines and downstream media- CRS, or simply a random association that could confound
tors also appear to play some role in sleep regulation and outcomes.33 Regardless, differences in patient-reported
maintenance of proper sleep architecture during health and (PSQI, FSS, EpSS) and objective (awakenings, REM
disease.10, 29, 30 Some studies have also observed alterations latency, snoring) sleep measures persisted when controlling
in REM latency related to systemic inflammation.31, 32 Cer- for BMI, suggesting that differences in BMI could not fully

International Forum of Allergy & Rhinology, Vol. 9, No. 1, January 2019 21


Alt et al.

explain the association between CRS and poor sleep qual- our findings in a separate, unrelated cohort with a wider
ity. The difference in average overnight oxygen saturation range of disease severity and BMI. In addition, one may ex-
was no longer significant when controlling for BMI, sug- plore whether objective sleep parameters change with dis-
gesting either BMI was confounding this relationship or the ease exacerbations and therapies, findings that could pro-
study was not powered to perform this secondary analysis. vide further evidence of causality.
Certainly, with a case-control study design, one can never
fully rule out that unmeasured or unknown residual con-
founding was present, and thus confirmatory studies are
Conclusion
needed. Patients with CRS report increased sleep dysfunction, fa-
Strengths of the study include its prospective, multi- tigue, and daytime somnolence when compared with con-
institutional design and exploration of both patient- trol subjects. In addition, subjects with CRS have increased
reported and objective sleep measures. However, the study snoring, more awakenings, and longer REM sleep latency,
took place in tertiary rhinology clinics and included even after controlling for confounding factors. Further
patients severe enough to consider surgery; thus, it is un- study is needed to confirm these findings in additional co-
clear whether these findings are generalizable across all pa- horts and to determine whether objective measures of sleep
tients and settings. A logical next step would be to confirm dysfunction improve after treatment.

References
1. Orlandi RR, Kingdom TT, Hwang PH, et al. Inter- 12. Hopkins C, Gillett S, Slack R, Lund VJ, Browne JP. synchronized home polysomnography. Sleep.
national Consensus Statement on Allergy and Rhi- Psychometric validity of the 22-item Sinonasal Out- 2006;29:367–374.
nology: Rhinosinusitis. Int Forum Allergy Rhinol. come Test. Clin Otolaryngol. 2009;34:447–454. 24. Herscovici S, Pe’er A, Papyan S, Lavie P. Detecting
2016;6(suppl 1):S22–S209. 13. Lund VJ, Kennedy DW. Staging for rhinosinusi- REM sleep from the finger: an automatic REM sleep
2. Adams PF, Heyman KM, Vickerie JL. Summary tis. Otolaryngol Head Neck Surg. 1997;117(suppl): algorithm based on peripheral arterial tone (PAT) and
health statistics for the U.S. population: National S35–40. actigraphy. Physiol Meas. 2007;28:129–140.
Health Interview Survey, 2008. Vital Health Stat 10. 14. Lund VJ, Mackay IS. Staging in rhinosinusitus. Rhi- 25. Bresler M, Sheffy K, Pillar G, Preiszler M, Herscovici
2009;(243):1–104. nology. 1993;31:183–184. S. Differentiating between light and deep sleep stages
3. Alt JA, Ramakrishnan VR, Platt MP, et al. Sleep qual- 15. Ferentinos P, Kontaxakis V, Havaki-Kontaxaki B, using an ambulatory device based on peripheral arte-
ity outcomes after medical and surgical management Dikeos D, Lykouras L. Psychometric evaluation of the rial tonometry. Physiol Meas. 2008;29:571–584.
of chronic rhinosinusitis. Int Forum Allergy Rhinol. Fatigue Severity Scale in patients with major depres- 26. Palagini L, Baglioni C, Ciapparelli A, Gemignani A,
2017;7:113–118. sion. Qual Life Res. 2011;20:457–465. Riemann D. REM sleep dysregulation in depression:
4. Alt JA, Smith TL, Mace JC, Soler ZM. Sleep quality 16. Krupp LB, LaRocca NG, Muir-Nash J, Steinberg AD. state of the art. Sleep Med Rev. 2013;17:377–390.
and disease severity in patients with chronic rhinosi- The fatigue severity scale. Application to patients with 27. Marti I, Valko PO, Khatami R, Bassetti CL,
nusitis. Laryngoscope. 2013;123:2364–2370. multiple sclerosis and systemic lupus erythematosus. Baumann CR. Multiple sleep latency measures in nar-
5. Alt JA, Smith TL, Schlosser RJ, Mace JC, Soler ZM. Arch Neurol. 1989;46:1121–1123. colepsy and behaviourally induced insufficient sleep
Sleep and quality of life improvements after endo- 17. Johns MW. Reliability and factor analysis of the Ep- syndrome. Sleep Med. 2009;10:1146–1150.
scopic sinus surgery in patients with chronic rhinosi- worth Sleepiness Scale. Sleep. 1992;15:376–381. 28. Tomassen P, Vandeplas G, Van Zele T, et al. In-
nusitis. Int Forum Allergy Rhinol. 2014;4:693–701. flammatory endotypes of chronic rhinosinusitis based
18. Kroenke K, Spitzer RL, Williams JB. The Patient
6. Sukato DC, Abramowitz JM, Boruk M, Goldstein NA, Health Questionnaire-2: validity of a two-item depres- on cluster analysis of biomarkers. J Allergy Clin Im-
Rosenfeld RM. Endoscopic sinus surgery improves sion screener. Med Care. 2003;41:1284–1292. munol. 2016;137:1449–1456.e1444.
sleep quality in chronic rhinosinusitis: a systematic re- 29. Krueger JM, Clinton JM, Winters BD, et al. Involve-
view and meta-analysis. Otolaryngol Head Neck Surg. 19. Hedner J, Pillar G, Pittman SD, Zou D, Grote L, White
DP. A novel adaptive wrist actigraphy algorithm for ment of cytokines in slow wave sleep. Prog Brain Res.
2018;158:249–256. 2011;193:39–47.
sleep-wake assessment in sleep apnea patients. Sleep.
7. Rotenberg BW, Pang KP. The impact of sinus 2004;27:1560–1566. 30. Alt JA, Bohnet S, Taishi P, et al. Influenza virus-
surgery on sleep outcomes. Int Forum Allergy Rhinol. induced glucocorticoid and hypothalamic and lung
2015;5:329–332. 20. Pittman SD, Ayas NT, MacDonald MM, Malhotra A,
Fogel RB, White DP. Using a wrist-worn device based cytokine mRNA responses in dwarf lit/lit mice. Brain
8. DeConde AS, Mace JC, Bodner T, et al. SNOT-22 on peripheral arterial tonometry to diagnose obstruc- Behav Immunol. 2007;21:60–67.
quality of life domains differentially predict treatment tive sleep apnea: in-laboratory and ambulatory vali- 31. Cui XY, Yang G, Cui SY, et al. Sleep patterns deteri-
modality selection in chronic rhinosinusitis. Int Forum dation. Sleep. 2004;27:923–933. orate over time in chronic corticosterone-treated rats.
Allergy Rhinol. 2014;4:972–979. Neurosci Lett. 2018;682:74–78.
21. Bar A, Pillar G, Dvir I, Sheffy J, Schnall RP, Lavie
9. Buysse DJ, Reynolds CF 3rd, Monk TH, Berman SR, P. Evaluation of a portable device based on peripheral 32. Berson SR, Klimczak J, Prezio EA, Hu S, Abraham
Kupfer DJ. The Pittsburgh Sleep Quality Index: a new arterial tone for unattended home sleep studies. Chest. M. Clinical associations between allergies and rapid
instrument for psychiatric practice and research. Psy- 2003;123:695–703. eye movement sleep disturbances. Int Forum Allergy
chiatry Res. 1989;28:193–213. Rhinol. 2018;8:817–824.
22. Ayas NT, Pittman S, MacDonald M, White DP. As-
10. Krueger JM. The role of cytokines in sleep regulation. sessment of a wrist-worn device in the detection of 33. Bhattacharyya N. Associations between obesity and
Curr Pharm Des. 2008;14:3408–3416. obstructive sleep apnea. Sleep Med. 2003; 4:435–442. inflammatory sinonasal disorders. Laryngoscope.
11. Fokkens WJ, Lund VJ, Mullol J, et al. EPOS 2012: 23. Zou D, Grote L, Peker Y, Lindblad U, Hedner J. 2013;123:1840–1844.
European position paper on rhinosinusitis and nasal Validation a portable monitoring device for sleep
polyps 2012. A summary for otorhinolaryngologists. apnea diagnosis in a population based cohort using
Rhinology. 2012; 50:1–12.

22 International Forum of Allergy & Rhinology, Vol. 9, No. 1, January 2019

Vous aimerez peut-être aussi