ArticlePDF Available

Pain and depression are associated with both physical and mental fatigue independently of comorbidities and medications in primary Sjögren's syndrome

BMJ
RMD Open
Authors:

Abstract

Objectives To report on fatigue in patients from the United Kingdom Primary Sjögren’s Syndrome Registry identifying factors associated with fatigue and robust to assignable causes such as comorbidities and medications associated with drowsiness. Methods From our cohort (n=608) we identified those with comorbidities associated with fatigue, and those taking medications associated with drowsiness. We constructed dummy variables, permitting the contribution of these potentially assignable causes of fatigue to be assessed. Using multiple regression analysis, we modelled the relationship between Profile of Fatigue and Discomfort (PROFAD) physical and mental fatigue scores and potentially related variables. Results Pain, depression, and daytime sleepiness scores were closely associated with both physical and mental fatigue (all p≤0.0001). In addition, dryness was strongly associated with physical fatigue (p≤0.0001). These effects were observed even after adjustment for co-morbidities associated with fatigue or medications associated with drowsiness. Conclusions These findings support further research and clinical interventions targeting pain, dryness, depression and sleep to improve fatigue in PSS patients.
1
Hackett KL, etal. RMD Open 2019;5:e000885. doi:10.1136/rmdopen-2018-000885
ORIGINAL ARTICLE
Pain and depression are associated with
both physical and mental fatigue
independently of comorbidities and
medications in primary
Sjögren’s syndrome
Kate L Hackett,1 Kristen Davies,2 Jessica Tarn,3 Rebecca Bragg,3 Ben Hargreaves,3
Samira Miyamoto,3,4 Peter McMeekin,1 Sheryl Mitchell,5 Simon Bowman,6
Elizabeth J Price,7 Colin Pease,8 Paul Emery,9,10 Jacqueline Andrews,10
Peter Lanyon, 11 John Hunter,12 Monica Gupta,12 Michele Bombardieri,13
Nurhan Sutcliffe,14,15 Costantino Pitzalis,16 John McLaren,17 Annie Cooper,18
Marian Regan,19 Ian Giles,20 David Isenberg,20 Saravan Vadivelu,21 David Coady,22
Bhaskar Dasgupta,23 Neil McHugh,24 Steven Young-Min,25 Robert Moots, 26
Nagui Gendi,27 Mohammed Akil,28 Bridget Grifths,29 Dennis W Lendrem, 3,30
Wan-Fai Ng,30,31 On behalf of the UK primary Sjögren’s Syndrome Registry
To cite: HackettKL, DaviesK,
TarnJ, etal. Pain and
depression are associated with
both physical and mental fatigue
independently of comorbidities
and medications in primary
Sjögren’s syndrome. RMD Open
2019;5:e000885. doi:10.1136/
rmdopen-2018-000885
Prepublication history and
additional material for this
paper are available online. To
view these les, please visit
the journal online (http:// dx. doi.
org/ 10. 1136/ rmdopen- 2018-
000885).
KLH and KD are joint rst
authors.
Received 18 December 2018
Revised 20 March 2019
Accepted 27 March 2019
For numbered afliations see
end of article.
Correspondence to
Dr Wan-Fai Ng;
wan- fai. ng@ newcastle. ac. uk
Sjögren syndrome
© Author(s) (or their
employer(s)) 2019. Re-use
permitted under CC BY-NC. No
commercial re-use. See rights
and permissions. Published
by BMJ.
ABSTRACT
Objectives To report on fatigue in patients from the
United Kingdom primary Sjögren’s syndrome (pSS) registry
identifying factors associated with fatigue and robust to
assignable causes such as comorbidities and medications
associated with drowsiness.
Methods From our cohort (n = 608), we identied those
with comorbidities associated with fatigue, and those
taking medications associated with drowsiness. We
constructed dummy variables, permitting the contribution
of these potentially assignable causes of fatigue to be
assessed. Using multiple regression analysis, we modelled
the relationship between Prole of Fatigue and Discomfort
physical and mental fatigue scores and potentially related
variables.
Results Pain, depression and daytime sleepiness
scores were closely associated with both physical and
mental fatigue (all p ≤ 0.0001). In addition, dryness was
strongly associated with physical fatigue (p ≤ 0.0001).
These effects were observed even after adjustment for
comorbidities associated with fatigue or medications
associated with drowsiness.
Conclusions These ndings support further research and
clinical interventions targeting pain, dryness, depression
and sleep to improve fatigue in patients with pSS.
This nding is robust to both the effect of other
comorbidities associated with fatigue and medications
associated with drowsiness.
INTRODUCTION
Primary Sjögren’s syndrome (pSS) is a
chronic, systemic autoimmune disease
characterised by functional impairment of
the lachrymal and salivary glands. The disease
can result in systemic involvement, which
Key messages
What is already known about this subject?
Patients with primary Sjögren’s syndrome (pSS) of-
ten report fatigue as the symptom most in need of
improvement.
What does this study add?
This study identies pain, depression and daytime
sleepiness scores as those factors most closely as-
sociated with both physical and mental fatigue in
pSS.
This nding is robust to both the effect of other co-
morbidities associated with fatigue and medications
associated with drowsiness.
How might this impact on clinical practice?
Interventions directed at managing fatigue might
be expected to have a signicant impact on the pa-
tient’s quality of life.
These ndings support an interdisciplinary clinical
assessment with:
an initial medication review,
assessment for comorbidities associated with
fatigue,
screening for anxiety and depression,
assessment for undiagnosed sleep disorders,
pain management interventions and
interventions targeting fatigue management
directly.
on 25 April 2019 by guest. Protected by copyright.http://rmdopen.bmj.com/RMD Open: first published as 10.1136/rmdopen-2018-000885 on 24 April 2019. Downloaded from
2Hackett KL, etal. RMD Open 2019;5:e000885. doi:10.1136/rmdopen-2018-000885
RMD OpenRMD OpenRMD Open
may affect any organ, resulting in manifestations such
as vasculitis, neuropathy, as well as skin, lung and kidney
involvement.1 Patients with pSS often report fatigue as an
important symptom in need of management.2 Defined as
an overwhelming sense of tiredness, lack of energy and a feeling
of exhaustion,3 4 fatigue is associated with poor health5 and
functional impairment.6 Previous studies have shown that
physical fatigue measured with the somatic component
of the Profile of Fatigue scale (PROFAD)7 is more preva-
lent and severe in patients with pSS than mental fatigue
as measured with the mental component of the same
scale.7–9 Unsurprisingly, patients with pSS also report
higher daytime sleepiness scores.9
For some patients with pSS, fatigue is likely to have an
assignable cause other than pSS. For example, they may
be taking medications associated with drowsiness, or have
comorbidities associated with fatigue—including hypo-
thyroidism, depression, obesity, coeliac disease, diabetes
and anaemia.
While there have been several previous studies exam-
ining the relationship between pSS, fatigue and other
factors, these were conducted with lower numbers of
patients and without regard to confounding medications
or comorbidities. Furthermore, few studies have used the
PROFAD which is a fatigue measure developed specifi-
cally for pSS.7
The objective of this paper is to report on the fatigue
symptoms of a large cohort of 608 patients diagnosed with
pSS defined using the American European Consensus
Group (AECG) classification criteria and to identify
factors associated with fatigue, and robust to assignable
causes such as comorbidities and medications associated
with fatigue.
METHODS
Design
The United Kingdom primary Sjögren’s syndrome
registry (UKPSSR, www. sjogrensregistry. org) is a proto-
col-driven, multicentre, cross-sectional study using stand-
ardised patient-reported measures and physician-gen-
erated assessments of patients with pSS from across the
UK.10 All patients are participants in the UKPSSR and
fulfil the AECG classification criteria. Note that fibromy-
algia was an explicit exclusion criterion for the UKPSSR.
Informed consent was obtained from all participants
according to the principles of the Helsinki Declaration.
Research ethics approval was granted by the North West
Research Ethics Committee in the UK. All clinical and
laboratory data were collected prospectively using a stand-
ardised pro-forma at the time of recruitment as previously
described.10 At the time of analysis, 639 patients had been
recruited to UKPSSR and complete data were available
for 608 patients.
Fatigue assessment
The Profile of Fatigue and Discomfort (PROFAD)7 was
designed specifically for patients with pSS. It includes
six questions which assess somatic and mental fatigue
on an eight-point (0–7) scale. An average is taken for
each domain score with a score of above 2.0 and 1.8
being considered significant for the somatic fatigue and
mental fatigue domains, respectively.7 8 The PROFAD
also includes a 10 cm visual analogue score of fatigue,
which provides a score of 0 for absent to 100 for worst
imaginable perceived fatigue levels.
Pain, depression and other assessments of patients with pSS
The following patient-reported outcome measures are
collected for all UKPSSR pSS subjects: Epworth Sleepi-
ness Scale (EPWORTH), Hospital Anxiety and Depres-
sion Scale (HADs—anxiety and depression), Euro-
pean League Against Rheumatism (EULAR) Sjögren's
syndrome Patient Reported Index (ESSPRI) (measure
of overall symptom burden and includes a 1–10 pain
score), EULAR Sicca Score (ESS) and EULAR Sjögren’s
Syndrome Disease Activity Index (ESSDAI).
All UKPSSR participants are asked to indicate which
symptom they perceive as being the most in need of
improvement (physical fatigue, mental fatigue, dryness
or pain). At the time of recruitment, the presence of
anti-Ro and/or anti-La antibodies are recorded, as well
as levels of systemic inflammation including C-reactive
protein and erythrocyte sedimentation rate. Additionally,
body mass index (BMI) and co-morbidities and medica-
tions are recorded for each participant.
We captured the effect of multiple drugs and comorbid-
ities using a comorbidity and polypharmacy score (CPS).
This score is obtained by combining the number of comor-
bidities and the number of prescribed medications.11 To
identify potentially assignable causes for fatigue in patients
with pSS, we analysed our cohort for existing comorbidities
and medication status. We classified the following comor-
bidities as conditions potentially associated with fatigue
independently of pSS: hypothyroidism, diabetes mellitus
(insulin-dependent diabetes mellitus (IDDM) and non-in-
sulin-dependent diabetes mellitus (NIDDM)), coeliac
disease, anaemia (haemoglobin <10), a diagnosis of clinical
depression and severe obesity (BMI 40).12 13 In addition,
patients whose fatigue might be assignable to medications
linked with drowsiness (eg, antidepressants, antipsychotics,
opioids, etc) were identified—see online supplementary
table S1. Using these data, we created dummy variables for
each patient to adjust for medications associated with drows-
iness (DROWSY Medications) or comorbidities associated
with fatigue (COMORBID).
Statistical analysis
Multiple regression analysis was used to model the relation-
ships between physical and mental fatigue and candidate
variables, including age, sex, pain, dryness, anxiety, depres-
sion, daytime sleepiness scores, BMI, symptom years, drowsy
medications (DROWSY) and comorbidities (Anaemia
COMORBID, Hypothyroidism COMORBID, IDDM
COMORBID, NIDDM COMORBID, Obesity COMORBID,
Depression COMORBID, Coeliac COMORBID). Causation
on 25 April 2019 by guest. Protected by copyright.http://rmdopen.bmj.com/RMD Open: first published as 10.1136/rmdopen-2018-000885 on 24 April 2019. Downloaded from
3
Hackett KL, etal. RMD Open 2019;5:e000885. doi:10.1136/rmdopen-2018-000885
Sjögren syndromeSjögren syndromeSjögren syndrome
Table 1 Numbers of patients presenting with
comorbidities. While 170 patients presented with one and
22 presented with two or more comorbidities, most patients
(416) presented with no comorbidities
Comorbidities
Number of
patients
None 416
THYROID 114
COELIAC 16
DEPRESSION 16
DIABETES 10
OBESITY 8
THYROID+OBESITY 8
ANAEMIA 6
THYROID+DEPRESSION 3
THYROID+DIABETES 3
DIABETES+OBESITY 2
THYROID+ANAEMIA 2
DIABETES+DEPRESSION 1
ANAEMIA+COELIAC 1
THYROID+COELIAC 1
THYROID+DIABETES+OBESITY 1
Total 608
COMORBIDITIES Key: THYROID=hypothyroidism,
COELIAC=coeliac disease, DEPRESSION=clinical
depression diagnosis, DIABETES=IDDM or NIDDM diabetes,
OBESITY=obesity, ANAEMIA=anaemia.
cannot be inferred from such models: they can only hope to
highlight those variables associated with fatigue after adjust-
ment for the presence of other covariates. The assumptions
underlying the multiple regression analysis were tested by
lack-of-fit testing and inspection of residuals. Statistical anal-
yses were performed using SAS JMP (V.13) Statistical Data
Visualisation Software. Statistical significance was set at a p
value of 0.05.
RESULTS
The median age of our cohort (n=608) was 64 years
(IQR: 53–71). The median number of symptom years for
patients was 10 years (IQR: 5–17). Most UKPSSR patients
experienced dryness as measured by objective measures
of dryness such as Salivary Flow and Schirmer’s Test.
The median score on the ESSPRI scale was 5.67 (IQR:
3.67–7.00) and the EULAR Dryness Scale (ESS) was 6
(IQR: 4–8). The median disease activity score (ESSDAI)
for the cohort was 3 (IQR: 1–7). While more than half
of patients received one or more medications to manage
their dryness symptoms, 40% of patients ranked fatigue
or mental fatigue—rather than dryness—as the symptom
‘most in need of improvement’. This is similar to the
numbers ranking dryness as the most in need of improve-
ment (45%), and rather more than those ranking pain as
the symptom most in need of improvement (15%).
Comorbidities and medications associated with drowsiness
In addition to meeting AECG classification criteria for
Sjogren’s syndrome, patients in the UKPSSR cohort are
an older cohort presenting with relatively complex health
needs. The median CPS for the cohort was 6 (IQR: 3–9).
Almost one-third of patients with pSS presented with
one or more fatigue-related comorbidities. The most
commonly reported comorbidity was hypothyroidism,
see table 1.
In addition, 130 patients with pSS were prescribed a
number of different drugs associated with drowsiness.
The most common drowsiness-associated medications
included antidepressants, such as citalopram (18%),
amitriptyline (12%) and fluoxetine (11%), or analgesics
such as tramadol (13%). Some patients took two or more
drowsiness-associated medications, see table 2.
For information, summary statistics are reported sepa-
rately for four separate subsets of patients: (a) patients
with comorbidities associated with drowsiness, (b)
patients with medications associated with drowsiness, (c)
patients with both comorbidities and medications associ-
ated with drowsiness and (d) patients with neither comor-
bidities nor medications associated with drowsiness, see
online supplementary table S2. Note that most patients
presented with neither comorbidities nor medications
associated with drowsiness. While apparent differences
between subsets must be interpreted with caution—see
online supplementary appendix—not surprisingly, there
were statistically significant differences in both physical
and mental PROFAD fatigue scores for patients with
comorbidities associated with fatigue or medications asso-
ciated with drowsiness (Kruskal-Wallis test, p<0.0001).
Models of fatigue
In order to identify variables associated with physical and
mental fatigue, we ran multiple regression models sepa-
rately for PROFAD physical and PROFAD mental fatigue
scores. The models included the candidate variables: age,
sex, symptom years, anti-Ro or anti-La positivity, pain,
dryness, Epworth Sleepiness Scale and BMI. In addition,
we included dummy variables for comorbidities associ-
ated with drowsiness—anaemia, obesity, hypothyroidism,
IDDM, NIDDM, coeliac disease and a diagnosis of clinical
depression (labelled respectively, Anaemia COMORBID,
Obesity COMORBID, Hypothyroidism COMORBID, IDD
COMORBID, NIDD COMORBID, Coeliac COMORBID and
Depression COMORBID). The dummy variable DROWSY
Medications captured those patients on medications associ-
ated with drowsiness. A list of medications associated with
drowsiness is given in the online supplementary appendix.
In addition, the model included anxiety and depression
scores from the HADS. Given the large number of candi-
date variables, we used a Benjamini-Hochberg correction to
adjust the false discovery rate (p value=0.05).14 Further statis-
tical details are given in the online supplementary file. Full
multiple regression equations for PROFAD physical and
on 25 April 2019 by guest. Protected by copyright.http://rmdopen.bmj.com/RMD Open: first published as 10.1136/rmdopen-2018-000885 on 24 April 2019. Downloaded from
4Hackett KL, etal. RMD Open 2019;5:e000885. doi:10.1136/rmdopen-2018-000885
RMD OpenRMD OpenRMD Open
Table 2 Numbers of patients prescribed drowsy
medications. While 108 patients took one and 22 took two
or more drowsy medications, most patients (478) took no
drowsy medications. Of those taking one or more drowsy
medications, citalopram, tramadol, amitriptyline, uoxetine
and zopiclone were the most frequently reported
Drowsy medications
Number of
patients
None 478
CITALOPRAM 23
TRAMADOL 17
AMITRIPTYLINE 16
FLUOXETINE 14
ZOPICLONE 4
DIAZEPAM 3
DOSULEPIN 3
GABAPENTIN 3
PAROXETINE 3
BACLOFEN 2
DULOXETINE 2
IMIPRAMINE 2
MIRTAZAPINE 2
SERTRALINE 2
TEMAZEPAM 2
ZOLPIDEM 2
CARBAMAZEPINE 1
CLOMIPRAMINE 1
CLONAZEPAM 1
LAMOTRIGINE 1
LEVETIRACETAM 1
NORTRIPTYLINE 1
RISPERIDONE 1
SODIUM VALPROATE 1
CARBAMAZAPINE ESCITALOPRAM AMITRIPTYLINE 1
DIAZEPAM DOSULEPIN 1
DULOXETINE AMITRIPTYLINE CITALOPRAM 1
ESCITALOPRAM DIAZEPAM 1
GABAPENTIN FLUOXETINE 1
LAMOTRIGINE CITALOPRAM 1
TOPIRAMATE MIRTAZAPINE AMITRIPTYLINE 1
TRAMADOL AMITRIPTYLINE 1
TRAMADOL CITALOPRAM 3
TRAMADOL DOSULEPIN 1
TRAMADOL FLUOXETINE DIAZEPAM 1
TRAMADOL GABAPENTIN 1
TRAMADOL SERTRALINE LITHIUM CHLORPROMAZINE
TEMAZEPAM
1
TRAMADOL ZOPICLONE 1
VENLAFAXINE ROPINIROLE QUETIAPINE 1
ZOLPIDEM SERTRALINE 1
ZOPICLONE AMITRIPTYLINE 1
ZOPICLONE FLUOXETINE 1
ZOPICLONE LAMOTRIGINE FLUOXETINE 1
ZOPICLONE OLANZAPINE GABAPENTIN DIAZEPAM 1
Total 608
Figure 1 Summary multiple regression charts for (A)
PROFAD physical fatigue and (B) PROFAD mental fatigue
scores. Log worth values indicate the relative importance
of each variable in the model. The broken, blue, vertical
reference line is for Log worth=2.0 equivalent to an false
discovery rate-adjusted p value of 0.01. Note that both pain
and depression are closely associated with both physical and
mental fatigue. See text. BMI, body mass index; PROFAD,
Prole of Fatigue and Discomfort.
PROFAD mental scores are presented in online supplemen-
tary table 3. These analyses are summarised in figure 1. Note
that while there was a weak association between ESSDAI
disease activity scores and PROFAD fatigue scores, this was
not robust to the inclusion of other covariates, see online
supplementary appendix.
Interestingly, pain and depression scores are correlated
with both PROFAD physical fatigue and PROFAD mental
fatigue scores. The physical fatigue model identified pain,
depression, dryness and scores on the Epworth daytime
sleepiness scale as the most important variables associ-
ated with PROFAD physical fatigue scores (all p0.0001).
The mental fatigue model identified pain, depression
and scores on the Epworth daytime sleepiness scale as the
three most important variables associated with PROFAD
mental fatigue scores (all p0.0001).
The strong association of depression on the HADS
with both physical and mental fatigue prompted us to
repeat the analysis with depression COMORBID recoded
to include a new category—those patients with a HADS
score >8 or a depression score >8. Such patients warrant
further assessment for depression and may represent
a subset of those patients with currently undiagnosed
depression. Repeating the analysis, even after adjustment
for potential undiagnosed depression, the relationships
of pain, depression, dryness and Epworth daytime sleep-
iness scores remained statistically significant (p<0.0001).
None of the comorbidities associated with fatigue had
a statistically significant relationship with either PROFAD
physical or PROFAD mental scores. However, patients
prescribed medications associated with drowsiness had
higher PROFAD physical fatigue scores (p=0.0001).
on 25 April 2019 by guest. Protected by copyright.http://rmdopen.bmj.com/RMD Open: first published as 10.1136/rmdopen-2018-000885 on 24 April 2019. Downloaded from
5
Hackett KL, etal. RMD Open 2019;5:e000885. doi:10.1136/rmdopen-2018-000885
Sjögren syndromeSjögren syndromeSjögren syndrome
DISCUSSION
Fatigue is important to patients with pSS
This is the largest study to date investigating fatigue in
patients with pSS. Our study confirms previous observations
that fatigue is an important symptom in need of improve-
ment.15 The main objective was to report on fatigue in
patients with pSS after separating out effects attributable to
assignable causes such as comorbidities and medications.
Our data suggest that while patients with pSS may present
with complex comorbidities and medications associated
with fatigue, these account for a relatively small proportion
of the variation in fatigue symptoms.
Pain, depression and dryness
Fatigue symptoms are important to patients with pSS and
there is an association between fatigue and other symp-
toms such as pain and depression.16 17 Howard Tripp et
al18 identified pain and depression scores, in association
with the pro-inflammatory cytokines inducible protein
10 and interferon gamma, as the two symptoms best
predicting fatigue groups.18
Pain is associated with both mental and physical
fatigue.19 The origin of pain in pSS is unclear, although
research points towards neuropathic pain being the
most common type of pain in pSS, followed by nocicep-
tive pain.16 The relationship between pain and fatigue
is reported in other conditions including rheumatoid
arthritis (RA), systemic lupus erythematosus and anky-
losing spondylitis (AS).20–22 Previous research using
anti-tumour necrosis factor inhibitors in RA suggests that
the reductions in reported fatigue were attributable to a
reduction in pain, rather than direct effects on fatigue
itself23 and pain may be a mediator of fatigue. Pain can be
modified through medications which have an association
with drowsiness such as amitriptyline and gabapentin.
This contribution gives a possible explanation as to why
drowsy medications do not have a major role in predicting
mental fatigue.
Interestingly, we found that a diagnosis of clinical depres-
sion was not associated with higher fatigue. It is possible
that those patients with a pre-existing depression diagnosis
have been successfully treated for their depression and
consequently are less likely to experience fatigue. However,
less than 5% of our cohort had a clinical diagnosis of
depression—rather less than that for other rheumatic
diseases24—and yet more than half presented with HADS
scores suggesting they be screened for depression or were
treated with antidepressants. If depression were under-re-
ported, then this might lead to an apparent association
between depression scores and fatigue. However, the rela-
tionship between depression scores and fatigue was robust
to the introduction of an additional classification capturing
those patients with a potentially undiagnosed depression.
Dryness is one of the characteristic symptoms of pSS
and we observed a strong relationship between dryness
symptoms and physical fatigue. Dryness may be directly
related to fatigue, or it may act as an indicator of either
severity or disease activity. In a recent qualitative study
of pSS fatigue, patients described experiencing ‘ocular
fatigue’, where they experienced tiredness in their eyes,
which for many, related to their ocular dryness symp-
toms such as feeling gritty or sore.25 High dryness scores
may simply indicate that the disease is highly active, or
needs to be more closely managed in the clinic. Dryness
symptoms may be an indicator of autonomic dysfunction,
and fatigue may be a result of autonomic dysfunction.
In addition, dryness symptoms may be associated with
changes in nocturnal behaviour—see below.
Sleep
The Epworth Sleepiness Scale can be used as a screening
tool to identify patients who potentially have obstructive
sleep apnoea and other primary sleep disorders.26 We
observed that the scores on the Epworth scale were associ-
ated both with physical and mental fatigue. This finding is
similar to a recent study in RA, which found that both phys-
ical and mental fatigue were associated with poor sleep.27
While there are limitations to the Epworth scale, this does
suggest that patients with pSS be screened for sleep prob-
lems. This could include incorporating information from a
caregiver, partner or others, using other sleep instruments,
polysomnography and other tests with referral to special-
ists in sleep disorders. A recent review has identified that a
range of sleep disturbances, including night awakenings, are
common in patients with pSS.28 Troublesome sicca symp-
toms, pain and nocturia have been identified as symptoms
which can potentially disturb sleep causing a reduction in
sleep quality.28 Identification of specific sleep disturbances
in this patient group is essential, and a combination of objec-
tive and subjective measures is required to identify specific
primary sleep disorders,29 30 thereby ensuring that patients
have access to appropriate interventions. Treatment of sleep
disturbances in other rheumatic diseases has resulted in a
reduction of pain, fatigue and depression.31
RECOMMENDATIONS
Our data confirm the importance of fatigue symptoms—
both mental and physical—to patients with pSS and
permit identification of factors contributing to fatigue
including other comorbidities and medications associ-
ated with drowsiness. Given the wide variety of potential
factors contributing to fatigue, we support the view that
a multidisciplinary approach is essential for the clinical
management of fatigue in pSS.32
We observe that many patients with pSS are taking
multiple medications and we recommend a medica-
tion review be undertaken to identify drowsy medica-
tions which could be contributing to fatigue. If these
medications are discontinued, then a review should
be arranged and if the fatigue does not improve,
then treatment of comorbidities should be consid-
ered. However, as pain is a major contributor of both
mental and physical fatigue, the contribution of
some pain-modifying medications may be beneficial,
despite their association with drowsiness.
on 25 April 2019 by guest. Protected by copyright.http://rmdopen.bmj.com/RMD Open: first published as 10.1136/rmdopen-2018-000885 on 24 April 2019. Downloaded from
6Hackett KL, etal. RMD Open 2019;5:e000885. doi:10.1136/rmdopen-2018-000885
RMD OpenRMD OpenRMD Open
Patients with pSS should be tested for common comor-
bidities which can contribute to fatigue, including
anaemia or hypothyroidism and offered appropriate
treatments.
We recommend screening for depression and anxiety
and offering patients appropriate interventions to
address these symptoms. Non-pharmacological inter-
ventions (such as talking therapies) may reduce the
need for antidepressants and anti-anxiolytics, many of
which are associated with drowsiness.
We recommend that patients undergo a more detailed
sleep assessment in order to screen for a primary
sleep disorder. Patients identified with conditions
such as obstructive sleep apnoea should be offered
assignable causes such as Continuous Positive Airway
Pressure (CPAP) treatment.33 Interventions such as
cognitive behavioural therapy for insomnia (CBT-I)
are a first-line treatment for insomnia associated with
other medical conditions34 and may prove beneficial
to patients with pSS.28
Patients with pSS should be offered appropriate pain
management interventions. If pain is associated with
poor sleep, then CBT-I with a pain adjunct has been
suggested as a feasible treatment.35
Finally, in the absence of good evidence to support
effective drug treatments, fatigue interventions in
pSS might focus on a multidisciplinary approach
incorporating activity management, graded exercise/
activity and CBT.32
There are limitations to this study. While the PROFAD
system is an established and well-validated tool, there is still
a need for a good objective measure of fatigue. Though a
sample population of 608 patients gives some assurance that
this study is a reasonable cross section of patients in the UK,
it would be useful to have comparative data in the future
from other cohorts. While there was no evidence of biases
arising from missing data, we note that data were incom-
plete for some measures and these are noted in the relevant
tables of summary statistics. In addition, this was an obser-
vational study using cross-sectional data. We cannot infer
causality and are only able to report associations. Prediction
models do not imply causation.
CONCLUSIONS
Our data support that of others in recognising the impor-
tance of fatigue in the clinical management of patients
with pSS.3 5 36–40 Furthermore, our analysis permits the
identification of contributing factors—such as comor-
bidities and drowsiness-associated medications. In addi-
tion, we identified multiple factors associated with both
physical and mental fatigue in pSS. Most notable of these
are pain scores measured on the ESSPRI and depres-
sion scores measured on the HADS. These associations
are robust and observed in patients with pSS even after
adjustment for assignable causes such as other comor-
bidities and use of medications associated with drowsi-
ness. Interventions directed at managing fatigue might
be expected to have a significant impact on the patient’s
quality of life. We recommend that the management of
fatigue in pSS be multidisciplinary, and personalised to
each patient depending on potential contributors to
what is a debilitating and complex symptom.
Author afliations
1Department of Life & Health Sciences, Northumbria University Department of
Public Health and Wellbeing, Newcastle upon Tyne, UK
2Musculoskeletal Research Group, Newcastle University Faculty of Medical
Sciences, Newcastle upon Tyne, UK
3Musculoskeletal Research Group, Newcastle University Faculty of Medical
Sciences, Newcastle upon Tyne, UK
4Departamento de Educação Integrada em Saúde, Universidade Federal do Espirito
Santo, Vitoria, Brazil
5Musculoskeletal Services, Newcastle Upon Tyne Hospitals NHS Foundation Trust,
Newcastle Upon Tyne, UK
6Rheumatology Research Group, University of Birmingham, Birmingham, UK
7Rheumatology, Great Western Hospitals NHS Foundation Trust, Swindon, UK
8Rheumatology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
9Rheumatology, University of Leeds, Leeds Institute of Rheumatic and
Musculoskeletal Medicine, Leeds, UK
10Musculoskeletal, NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds,
UK
11Rheumatology, Nottingham University Hospitals NHS Trust, Nottingham, UK
12Gartnavel General Hospital, Glasgow, UK
13Experimental Medicine and Rheumatology, Queen Mary University of London,
London, UK
14Barts Health NHS Trust, London, UK
15Rheumatology, Barts and The London School of Medicine and Dentistry, London,
UK
16Experimental Medicine and Rheumatology, William Harvey Research Institute
Experimental Medicine and Musculoskeletal Sciences, London, UK
17NHS Fife, Kirkcaldy, UK
18Royal Hampshire County Hospital, Winchester, UK
19Royal Derby Hospital, Derby, UK
20Centre for Rheumatology, University College London, London, UK
21Queen Elizabeth Hospital, Gateshead, UK
22City Hospitals Sunderland NHS Foundation Trust, Sunderland, UK
23Department of Rheumatology, Southend University Hospital NHS Foundation Trust,
Westcliff-on-Sea, UK
24Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
25Rheumatology, Portsmouth Hospitals NHS Trust, Portsmouth, UK
26Rheumatology, Aintree University Hospitals, Liverpool, UK
27Department of Rheumatology, Basildon Hospital, Basildon, UK
28Department of Rheumatology, Shefeld Teaching Hospitals NHS Foundation Trust,
Shefeld, UK
29Musculoskeletal Services, Newcastle Upon Tyne Hospitals NHS Foundation Trust,
Newcastle Upon Tyne, UK
30Musculoskeletal Ageing, NIHR Newcastle Biomedical Research Centre, Newcastle
upon Tyne, UK
31Musculoskeletal Research Group, Newcastle University Faculty of Medical
Sciences, Newcastle upon Tyne, UK
Acknowledgements We thank all patients and volunteers who joined the UKPSSR
cohort.
Collaborators See Appendix 1 for the full list of members.
Contributors SJB, BG, W-FN conceived the UKPSSR study. Study coordination
and data collection for the UKPSSR was coordinated by SM. Data collection
was performed by SM, SJB, EP, CTP, PE, PL, JH, MG, MB, NS, CP, JMcL, AC, MR,
IG, DI, SV, DC, BD, NMcH, SY-M, RM, NG, MA, BG and W-FN. BH performed data
management and data processing. DWL advised and KLH, KD, JRT, STM and RB
performed the analyses. The paper was drafted by KLH, KD, DWL and W-FN. All
authors reviewed and approved the nal manuscript.
Funding The UKPSSR received grant support from the UK Medical Research
Council (G0800629 to WFN, SJB, BG), British Sjögren’s Syndrome Association
and Newcastle University. This work also received infra-structure support from
the National Institute for Health Research Newcastle Biomedical Research Centre
on 25 April 2019 by guest. Protected by copyright.http://rmdopen.bmj.com/RMD Open: first published as 10.1136/rmdopen-2018-000885 on 24 April 2019. Downloaded from
7
Hackett KL, etal. RMD Open 2019;5:e000885. doi:10.1136/rmdopen-2018-000885
Sjögren syndromeSjögren syndromeSjögren syndrome
based at Newcastle Hospitals NHS Foundation Trust and Newcastle University. SB
was part funded by the Birmingham Biomedical Research Centre. KH was in receipt
of Newcastle upon Tyne NHS Foundation Trust NIHR Research Capability Funding.
Competing interests SJB has provided consultancy services in the area of
Sjögren’s syndrome for the following companies: Celgene, Eli Lilly, Glenmark, GSK,
Medimmune, Novartis, Ono, Pzer, Takeda, UCB. PE has undertaken clinical trials
and provided expert advice to Pzer, MSD, Abbvie, BMS, UCB, Roche, Novartis,
Samsung, Sandoz and Lilly. WFN has undertaken clinical trials and provided
consultancy or expert advice in the area of Sjögren’s syndrome to the following
companies: GlaxoSmithKline, MedImmune, Novartis, UCB, Abbvie, Roche, Eli Lilly,
Takeda, Resolves Therapeutics, and Nascient.
Patient consent for publication Obtained.
Provenance and peer review Not commissioned; externally peer reviewed.
Data sharing statement Data are available upon reasonable request.
Open access This is an open access article distributed in accordance with the
Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which
permits others to distribute, remix, adapt, build upon this work non-commercially,
and license their derivative works on different terms, provided the original work is
properly cited, appropriate credit is given, any changes made indicated, and the
use is non-commercial. See: http:// creativecommons. org/ licenses/ by- nc/ 4. 0/.
RefeRences
1. Seror R, Theander E, Brun JG, etal. Validation of EULAR primary
Sjögren's syndrome disease activity (ESSDAI) and patient indexes
(ESSPRI). Ann Rheum Dis 2015;74:859–66.
2. Arends S, Meiners PM, Moerman RV, etal. Physical fatigue
characterises patient experience of primary Sjögren's syndrome.
Clin Exp Rheumatol 2017;35:255–61.
3. WF N, Bowman SJ. Primary Sjogren's syndrome: too dry and too
Tired. Rheumatology 2010;49:844–53.
4. Shen J, Barbera J, Shapiro CM. Distinguishing sleepiness and
fatigue: focus on denition and measurement. Sleep Med Rev
2006;10:63–76.
5. Segal B, Bowman SJ, Fox PC, etal. Primary Sjögren's syndrome:
health experiences and predictors of health quality among patients
in the United States. Health and Quality of Life Outcomes 2009;7.
6. Hackett KL, Newton JL, Frith J, etal. Impaired functional status in
primary Sjögren's syndrome. Arthritis Care Res 2012;64:1760–4.
7. Bowman SJ, Booth DA, Platts RG, etal. Measurement of fatigue and
discomfort in primary Sjögren's syndrome using a new questionnaire
tool. Rheumatology 2004;43:758–64.
8. Segal B, Thomas W, Rogers T, etal. Prevalence, severity, and
predictors of fatigue in subjects with primary Sjögren's syndrome.
Arthritis Rheum 2008;59:1780–7.
9. Theander L, Strombeck B, Mandl T, etal. Sleepiness or fatigue?
Can we detect treatable causes of tiredness in primary Sjogren's
syndrome? Rheumatology 2010;49:1177–83.
10. WF N, Bowman SJ, Grifths B. United Kingdom primary Sjogren's
syndrome Registry-a United effort to tackle an orphan rheumatic
disease. Rheumatology 2011;50:32–9.
11. Evans DC, Gerlach AT, Christy JM, etal. Pre-injury polypharmacy
as a predictor of outcomes in trauma patients. Int J Crit Illn Inj Sci
2011;1:104–9.
12. Fukuda K, Straus SE, Hickie I, etal. The chronic fatigue syndrome:
a comprehensive approach to its denition and study. International
chronic fatigue syndrome Study Group. Ann Intern Med
1994;121:953–9.
13. Reeves WC, Lloyd A, Vernon SD, etal. Identication of ambiguities
in the 1994 chronic fatigue syndrome research case denition and
recommendations for resolution. BMC Health Serv Res 2003;3.
14. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a
practical and powerful approach to multiple testing. J Roy Stat Soc
B Met 1995;57:289–300.
15. Hackett KL, Deane KHO, Newton JL, etal. Identifying priority
intervention targets to improve functional ability and participation
in primary Sjögren’s syndrome patients: A group concept mapping
study. Arthritis Care Res 2018;70:1064–73.
16. Giacomelli C, Talarico R, Baldini C, etal. Pain in Sjögren’s syndrome.
Reumatismo 2014;66:39–43.
17. Koh JH, Kwok SK, Lee J, etal. Pain, xerostomia, and younger age
are major determinants of fatigue in Korean patients with primary
Sjogren's syndrome: a cohort study. Scand J Rheumatol 2016:1–7.
18. Howard Tripp N, Tarn J, Natasari A, etal. Fatigue in primary
Sjögren's syndrome is associated with lower levels of
proinammatory cytokines. RMD Open 2016;2:e000282.
19. Segal BM, Pogatchnik B, Henn L, etal. Pain severity and
neuropathic pain symptoms in primary Sjögren's syndrome: a
comparison study of seropositive and seronegative Sjögren's
syndrome patients. Arthritis Care Res 2013;65:1291–8.
20. Hammoudeh M, Zack DJ, Li W, etal. Associations between
inammation, nocturnal back pain and fatigue in ankylosing
spondylitis and improvements with etanercept therapy. J Int Med
Res 2013;41:1150–9.
21. Waldheim E, Elkan A-C, Pettersson S, etal. Health-related quality
of life, fatigue and mood in patients with SLE and high levels of pain
compared to controls and patients with low levels of pain. Lupus
2013;22:1118–27.
22. van Dartel SAA, Repping-Wuts JWJ, van Hoogmoed D, etal.
Association between fatigue and pain in rheumatoid arthritis: does
pain precede fatigue or does fatigue precede pain? Arthritis Care
Res 2013;65:862–9.
23. Druce KL, Jones GT, Macfarlane GJ, etal. Improvements in
fatigue following anti-TNF therapy do not reect reductions in
inammation and are instead a downstream consequence of pain
improvements: results from the british Society for rheumatology
biologics register for rheumatoid arthritis. Rheumatology
2015;54(suppl_1).
24. Matcham F, Rayner L, Steer S, etal. The prevalence of depression
in rheumatoid arthritis: a systematic review and meta-analysis.
Rheumatology 2013;52:2136–48.
25. Stack RJ, Southworth S, Fisher BA, etal. A qualitative exploration of
physical, mental and ocular fatigue in patients with primary Sjögren's
syndrome. Plos One 2017;12:e0187272.
26. Løppenthin K, Esbensen BA, Jennum P, etal. Sleep quality and
correlates of poor sleep in patients with rheumatoid arthritis. Clin
Rheumatol 2015;34:2029–39.
27. Rosenthal LD, Dolan DC. The Epworth Sleepiness Scale in
the identication of obstructive sleep apnea. J Nerv Ment Dis
2008;196:429–31.
28. Hackett KL, Gotts ZM, Ellis J, etal. An investigation into
the prevalence of sleep disturbances in primary Sjögren’s
syndrome: a systematic review of the literature. Rheumatology
2017;56:570–80.
29. Epstein LJ, Kristo D, Strollo PJ, etal. Clinical guideline for the
evaluation, management and long-term care of obstructive sleep
apnea in adults. J Clin Sleep Med 2009;5;3.
30. Zhang L, Zhao Z-X. Objective and subjective measures for sleep
disorders. Neurosci Bull 2007;23:236–40.
31. Martínez MP, Miró E, Sánchez AI, etal. Cognitive-behavioral therapy
for insomnia and sleep hygiene in bromyalgia: a randomized
controlled trial. J Behav Med 2014;37:683–97.
32. Price E, Rauz S, Sutcliffe N, etal. BSR and BHPR Guideline for
the Management of Adults with Primary Sjögren’s Syndrome.
Rheumatology 2017;56:e24–48.
33. National Institute for Health and Clinical Excellence (NICE).
Continuous positive airway pressure for the treatment of obstructive
sleep apnoea/hypopnoea syndrome. London: National Institute for
Health and Care Excellence, 2008.
34. Smith MT, Huang MI, Manber R. Cognitive behavior therapy for
chronic insomnia occurring within the context of medical and
psychiatric disorders. Clin Psychol Rev 2005;25:559–92.
35. Finan PH, Buenaver LF, Coryell VT, etal. Cognitive-behavioral
therapy for comorbid insomnia and chronic pain. Sleep Med Clin
2014;9:261–74.
36. Bowman SJ, Booth DA, Platts RG. Measurement of fatigue and
discomfort in primary Sjögren's syndrome using a new questionnaire
tool. Rheumatology 2004;43:758–64.
37. Frith J, Newton J. Fatigue Impact Scale. Occup Med
2010;60:159–59.
38. Lendrem D, Mitchell S, McMeekin P, etal. Health-related utility
values of patients with primary Sjögren's syndrome and its
predictors. Ann Rheum Dis 2014;73.
39. Newton JL, Jones DEJ. Making sense of fatigue. Occup Med
2010;60:326–9.
40. Westhoff G, Dörner T, Zink A. Fatigue and depression predict
physician visits and work disability in women with primary
Sjogren's syndrome: results from a cohort study. Rheumatology
2012;51:262–9.
on 25 April 2019 by guest. Protected by copyright.http://rmdopen.bmj.com/RMD Open: first published as 10.1136/rmdopen-2018-000885 on 24 April 2019. Downloaded from
... These extra-glandular manifestations, including persistent fatigue, widespread joint and muscular systems. These extra-glandular manifestations, including persistent fatigue, widespr joint and muscular pain, and cognitive impairments, not always aligning with serol indicators, significantly contribute to a diminished quality of life [6]. ...
... Nevertheless, Hacket et al. outlined that individuals diagnosed with pSS demonstrate a reduced capacity to perform a variety of daily tasks compared to age-and gender-matched individuals in good health [6]. ...
... These limitations also correspond to a decrease in the health-related quality of life, as a significant correlation was found between the quality of life and some physical symptoms such as pain or dryness, and the somatic aspects such as fatigue and depression. They also described an association between the CRP (C-reactive protein) and functional disability, indicating the contribution of inflammatory status to functional capacity [6]. ...
Article
Full-text available
Background: Primary Sjögren’s syndrome (pSS) is a complex autoimmune disorder characterized by organ-specific symptoms in the salivary and lacrimal glands, as well as systemic manifestations. Fatigue, a prominent aspect, significantly influences the overall quality of life for individuals with pSS. Methods: This review seeks to evaluate the impact of fatigue by exploring its consequences, potential causes, and effects on physical and psychological well-being, while also investigating its management strategies. Following the “Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA)” guidelines, our systematic literature review involved a five-step algorithm. Initially identifying 78 articles in reputable international medical databases, we applied eligibility criteria and removed duplicates, resulting in 19 articles for qualitative synthesis. Results: This review delves into the predictive factors for heightened fatigue in pSS, encompassing rheumatoid factor levels, erythrocyte sedimentation rate, and immunoglobulin G levels. Sleep disturbances, specifically nighttime pain and nocturia, emerged as determinants of persistent daytime fatigue. Cognitive impairment in pSS involves deteriorations in global memory, executive functioning, and attentional resources. Furthermore, functional limitations in pSS impact patients’ quality of life. Conclusions: The significance of fatigue in pSS, its consequences, and profound influence on the quality of life necessitate further research for a more comprehensive understanding of this complex issue.
... Fatigue [8] and work disability [7] correlate poorly with serological disease activity. On the other hand, fatigue correlates more strongly with symptoms such as dryness, [9] daytime sleepiness, [9] pain, [8,9] cognitive dysfunction [10] comorbid depression. [1,11] Autonomic nervous system (ANS) dysregulation is estimated to occur in 55% of people with SS, significantly more than age-and sex-matched controls. ...
... Fatigue [8] and work disability [7] correlate poorly with serological disease activity. On the other hand, fatigue correlates more strongly with symptoms such as dryness, [9] daytime sleepiness, [9] pain, [8,9] cognitive dysfunction [10] comorbid depression. [1,11] Autonomic nervous system (ANS) dysregulation is estimated to occur in 55% of people with SS, significantly more than age-and sex-matched controls. ...
... Fatigue [8] and work disability [7] correlate poorly with serological disease activity. On the other hand, fatigue correlates more strongly with symptoms such as dryness, [9] daytime sleepiness, [9] pain, [8,9] cognitive dysfunction [10] comorbid depression. [1,11] Autonomic nervous system (ANS) dysregulation is estimated to occur in 55% of people with SS, significantly more than age-and sex-matched controls. ...
Article
Full-text available
Objectives Sjögren’s syndrome (SS) includes many extra-glandular symptoms such as fatigue, pain, sleepiness and depression, which impact on quality of life (QoL). These symptoms also influence each other and could be linked by autonomic nervous system (ANS) dysregulation. Our aim was to model the role of putative predictive variables, including depression in the relationships between ANS function, fatigue, and QoL in SS. Methods Cross-sectional analysis of self-reported data from the multicentre UK primary SS registry. The Composite Autonomic Symptom Scale (COMPASS) was used to assess autonomic function, the Hospital Anxiety and Depression Scale (HADS) to assess anxiety and depression and the EuroQol-5 Dimension (EQ-5D) to assess QoL. Validated scales were used for other clinical variables. Using multiple regression analysis and structural equation modelling (SEM), we investigated how the QoL of people with SS is impacted by the direct and indirect effects of fatigue, sleepiness, depression, symptom burden and ANS function, and their interactions. Results Data was obtained for 1046 people with SS, 56% COMPASS completers. Symptoms of ANS dysregulation were common. Participants with ANS dysregulation had more severe depression, anxiety, dryness, fatigue, pain, sleepiness and QoL (P < 0.01 for all). Depression, anxiety, dryness, and pain were independent predictors of ANS function in the multiple regression model (P < 0.05 for all). ANS function could not be included in the SEM. The SEM model had good fit to the data (comparative fit index = 0.998) and showed that, in people with SS, depression mediates the effects of pain, fatigue and sleepiness on QoL. Conclusion Our results show that diagnosing and treating depression in people with SS could have direct positive impact on QoL, and significantly ameliorate the impact of fatigue and pain.
... The pathophysiology of SS-related fatigue is multifactorial and not yet fully elucidated (16). It has been previously associated with several psychological and physiological factors, which include sleep disturbances (17)(18)(19), dysfunction of the autonomic nervous system (20,21), depression (6,8,19,(22)(23)(24)(25), dysfunctional or alexithymic psychological profile (26), anxiety (24), pain (19,27), neuroticism and fibromyalgia (6,28). SS related fatigue has been also associated with lower levels of proinflammatory cytokines (22), as well as with higher heat shock protein levels, through their signaling in the central nervous system (29). ...
... The pathophysiology of SS-related fatigue is multifactorial and not yet fully elucidated (16). It has been previously associated with several psychological and physiological factors, which include sleep disturbances (17)(18)(19), dysfunction of the autonomic nervous system (20,21), depression (6,8,19,(22)(23)(24)(25), dysfunctional or alexithymic psychological profile (26), anxiety (24), pain (19,27), neuroticism and fibromyalgia (6,28). SS related fatigue has been also associated with lower levels of proinflammatory cytokines (22), as well as with higher heat shock protein levels, through their signaling in the central nervous system (29). ...
... The pathophysiology of SS-related fatigue is multifactorial and not yet fully elucidated (16). It has been previously associated with several psychological and physiological factors, which include sleep disturbances (17)(18)(19), dysfunction of the autonomic nervous system (20,21), depression (6,8,19,(22)(23)(24)(25), dysfunctional or alexithymic psychological profile (26), anxiety (24), pain (19,27), neuroticism and fibromyalgia (6,28). SS related fatigue has been also associated with lower levels of proinflammatory cytokines (22), as well as with higher heat shock protein levels, through their signaling in the central nervous system (29). ...
Article
Full-text available
Background/Purpose Primary Sjögren’s Syndrome (SS) is characterized by B lymphocyte hyperactivity with B cell activating factor (BAFF) acting as an important regulator. Single Nucleotide Polymorphisms (SNPs) of the BAFF gene have been implicated in the pathogenesis of several autoimmune diseases characterized by heightened fatigue levels, including primary SS. We aimed to explore potential associations between BAFF SNPs and fatigue status of primary SS patients. Methods Fatigue status was assessed in 199 consecutive primary SS patients (Greek cohort) using the Functional Assessment of Chronic Illness Therapy-Fatigue (FACIT-F) scale. Clinical, histological, laboratory, psychometric and personality data were also collected. DNA extracted from peripheral blood of all patients underwent evaluation for the presence of five BAFF SNPs (rs9514827, rs1041569, rs9514828, rs1224141, rs12583006) by PCR. To confirm our findings, an independent replicative cohort of 62 primary SS patients (Dutch cohort) was implemented. Finally, 52 multiple sclerosis (MS) patients were served as disease controls (MS cohort). Analysis of BAFF SNPs in association with fatigue levels was performed by the online platforms SNPStats and SHEsis and the SPSS 26 and Graph Pad Prism 8.00 software. Results TT genotype of the rs9514828 BAFF polymorphism was significantly less frequent in the fatigued primary SS patients of the Greek cohort compared to the non-fatigued (14.1% vs 33.3%). The corresponding ORs [95%CI] in the dominant and overdominant models were 0.33 [0.15-0.72], p=0.003 and 0.42 [0.23-0.78], p=0.005 respectively. The association remained significant after adjustment for the variables contributing to fatigue in the univariate analysis (OR [95% CI]: 0.3 [0.1-0.9], p=0.026). Accordingly, in the Dutch cohort, there was a trend of lower mental fatigue among patients carrying the TT rs9514828 BAFF genotype compared to their CC counterparts (4.1 ± 2.4 vs 6.0 ± 2.2 respectively, p=0.06). The rs9514828 BAFF SNP was not significantly associated with fatigue in the MS cohort. Conclusions We report a novel association between genetic makeup and primary SS-associated fatigue with the rs9514828 TT genotype decreasing the likelihood of fatigue development among these patients. These findings need validation in multi-center studies.
... The association between fatigue and autonomic dysfunction also deserve more investigations. Fatigue is often described as the most disabling symptom of pSS (3,60) and has been shown elsewhere to be associated with autonomic dysfunction using multiple measuring tools in pSS (27,28,30,41,42) and other conditions such as chronic fatigue syndrome and PBC (61). The biological basis for fatigue is still unclear in pSS but immune dysregulation have been suggested (62,63). ...
... The biological basis for fatigue is still unclear in pSS but immune dysregulation have been suggested (62,63). In this regard, the existence of cross-talk between the ANS and the immune system raises the possibility that autonomic dysfunction may contribute to fatigue (60). ...
Article
Full-text available
Primary Sjögren’s syndrome (pSS) is an autoimmune disease which primarily affects the exocrine glands, but can also affect other organs, including the nervous system. Many studies have reported evidence of autonomic nervous system (ANS) dysfunction in pSS which may contribute to a wide range of symptoms and functional burden. Symptoms of ANS dysfunction are common and widespread among patients with pSS and are associated with other features of the disease, particularly fatigue. Accumulating data on the inter-relationship between the ANS and the immune system via the vagus nerve have been reported. Vagus nerve stimulation (VNS) has also been associated with improvement in fatigue in patients with pSS. Taken together, these data suggest that the ANS may be a potential treatment target for pSS, in particularly those with fatigue being a predominant symptom. Future research to dissect the link between the ANS, immune dysregulation and clinical manifestations in pSS and to evaluate the potential of VNS as a therapy for pSS is warranted.
... On the other hand, pain, depression, and daytime sleepiness scores were closely associated with both physical and mental fatigue in pSS, with effects observed even after adjustment for comorbidities associated with fatigue or medications associated with drowsiness (20). ...
... Fatigue (and pain) is an important predictor of poor QoL regardless of age, schooling, marital status, work disability, fibromyalgia and disease activity (11). Fatigue in pSS is multifactorial and is clinically related to sleep and mood disorders (20). On the other hand, it is not associated with systemic disease activity (11,17) and has an inverse correlation with pro-inflammatory cytokines (18,19). ...
Article
Full-text available
Sjögren’s syndrome (SS) is an autoimmune disease affecting the salivary and lacrimal glands. Symptoms range from dryness to severe extra-glandular disease involving manifestations in the skin, lungs, nervous system, and kidney. Fatigue occurs in 70% of patients, characterizing primary SS (pSS) and significantly impacting the patient’s quality of life. There are some generic and specific instruments used to measure fatigue in SS. The mechanisms involved with fatigue in SS are still poorly understood, but it appears fatigue signaling pathways are more associated with cell protection and defense than with pro-inflammatory pathways. There are no established pharmacological treatment options for fatigue in pSS. So far, exercise and neuromodulation techniques have shown positive effects on fatigue in pSS. This study briefly reviews fatigue in pSS, with special attention to outcome measures, biomarkers, and possible treatment options.
... It has been shown that, for patients with SS, QoL is impacted by the dry syndrome, fatigue, and pain (representing the perceptual sphere) (9,37), as in our study with a high prevalence of self-reported nonspecific symptoms. Hackett et al were able to highlight links between fatigue and pain in patients with primary SS (38). Taken together, these results lead to the notion of a psychosomatic approach which designates an interdisciplinary medical field exploring the relationships among social, psychological processes, pathological physical manifestations, whether they are functional or organic. ...
Article
Objectives: Sjögren's syndrome (SS) is an autoimmune disease with an impact on quality of life (QoL). The aim of patient education (PE) is to improve patients' QoL. The main objective of this study was to describe the medico-psycho-social characteristics defining the six spheres of an allosteric educational model in order to characterise clusters of patients with SS and intentionality for patients to participate in a programme of patient education. Methods: A self-administered questionnaire was proposed to 408 patients with SS followed in the internal medicine department of the University Hospital of Lille, France with the aim of assessing the six spheres of the allosteric model: intentional, perceptual, affective, cognitive, infra-cognitive and meta-cognitive. Sub objectives were to determine factors that can influence intentionality to participate in a PE programme and to determine, using cluster analysis, similar characteristics of patients with SS. Results: 127 patients (31%) agreed to participate and were included in the study; 96% were women and the median age was 51 years (± 14.5). They mostly reported dry syndrome and fatigue. They had a good knowledge of SS. They presented anxiety symptoms. They mainly had problem-centred coping strategies, internal locus of control and low self-esteem. SS had an impact on their social interactions. Considering intentionality to participate in a PE programme, patients were significantly younger, had a shorter duration of the disease, more frequently had disabled status, reported more fatigue, more self-reported symptoms and a poorer QoL. Two clusters of patients could be individualised, with one group including 75 (59%) patients presenting a higher global impact of the disease, including a more severe impairment for the scores of the perceptual, emotional and infra-cognitive spheres, worse physical QoL, and a higher intentionality to participate in a PE programme. Conclusions: Our study described an SS population in terms of the different spheres of an allosteric model applicable to the practice of PE. A cluster of patients appeared to present more impact of the disease and more intentionality to participate in a programme of PE. There was no difference between the two groups in terms of the cognitive sphere (i.e., knowledge of the disease), thus indicating that motivation to participate in a PE programme is influenced by non-cognitive factors. Considering intentionality to participate in a PE programme, duration disease, age of the patient and QoL should be more considered to propose to patients to participate in a PE programme. Use of the allosteric model appears promising for future research in PE.
... PDQ in multiple sclerosis 33 ) and seemingly unrelated (e.g. dryness in Sjögren's syndrome 34 ) symptoms. ...
Article
Full-text available
Background Autonomic nervous system (ANS) dysregulation might be relevant to the pathophysiology of fatigue and cognitive impairment in depression and perhaps should be considered when making prescribing decisions. Aims To determine the relationship of self-reported ANS symptoms with fatigue, cognition and prescribed medication in people with a diagnosis of depression, in comparators without depression but with other mental health, neurodevelopmental or neurode- generative disorders (active controls) and in healthy controls. Method Cross-sectional analysis of an opportunistic sample from England. Self-reported data were collected on demographics, diagnosis, medication, ANS symptoms (Composite Autonomic Symptom Scale-31, COMPASS-31) and fatigue (Visual Analogue Scale for Fatigue, VAS-F). A subsample completed cognitive tests (THINC-it), including the subjective Perceived Deficits Questionnaire five-item version (PDQ-5). Spearman’s correlation and mediation models were used to explore the relationship between COMPASS-31, VAS-F and PDQ-5 scores. Results Data were obtained for 3345 participants, 22% with depression. The depression group had significantly (P < 0.01) more severe autonomic dysregulation as measured by COMPASS-31 scores (median 30) than active (median 23) and healthy controls (median 10). The depression group had significantly higher symptom severity (P < 0.01) than both control groups on the VAS-F and PDQ-5. Overall, there was a significantly positive correlation (P < 0.01) between COMPASS-31, VAS-F scores (Spearman’s rho rs = 0.44) and PDQ-5 scores (rs = 0.56). COMPASS-31 scores mediated greater symptom severity on the VAS-F and PDQ-5 for those with depression. COMPASS-31 scores remained significantly different between the depression group and both control groups independently of medication. Conclusions People with a diagnosis of depression report worse fatigue and cognition than active and healthy comparators; this appears to be mediated by ANS dysregulation.
... 1 Studies in the literature on predictors of fatigue in PSS provide evidence that emotional state, fibromyalgia, pain, sleep disturbance, and dryness are associated with fatigue. [2][3][4][5][6] The associations of disease activity and inflammation with fatigue in PSS patients are somewhat complex. Evidence to date shows a limited association between disease activity and fatigue in PSS patients, and in addition, B cell-acting drugs, such as rituximab, also have limited beneficial effects on fatigue. ...
Article
Full-text available
Objectives The study aimed to evaluate the level of fatigue and the relationship between mood, pain, fibromyalgia, insomnia, disease activity, and dryness with fatigue in primary Sjögren’s syndrome (PSS) patients. Patients and methods In this case-control study, the participants were recruited between January 2021 and July 2021. Functional Assessment of Chronic Illness Therapy Fatigue (FACIT-F), pain DETECT questionnaire, Beck Depression Inventory (BDI), Beck Anxiety Inventory (BAI), Insomnia Severity Index (ISI) were administered to 50 PSS patients (48 females, 2 males; mean age: 48.9±10.8 years; median age: 47 years; range, 29 to 71 years) and 60 healthy controls (HCs; 57 females, 3 males; mean age: 49.8±8.4 years, median age: 52 years; range, 32 to 72 years). In addition, EULAR Sjögren’s syndrome disease activity index (ESSPRI), EULAR Sjögren’s Syndrome Patient Reported Index (ESSDAI), pain thresholds, Schirmer tests, and whole unstimulated salivary flow rate measurements were determined in PSS patients. Independent predictors of fatigue (fatigue subscale scores <30.5) were investigated by logistic regression analysis. Results The frequency of fatigue in PSS patients and HCs was 54.0% and 8.3%, respectively. The rates of mood disturbance (BDI ≥11) in PSS patients with and without fatigue were 70.4% and 13.1%, respectively. BDI (Rho=-0.804), BAI (Rho=-0.586), ISI (Rho=-0.483), and ESSDAI (Rho=-0.345) were negatively correlated with the fatigue subscale score. Depression [Odds ratio (OR): 1.214, confidence interval (CI): 1.007-1.463], fibromyalgia (OR: 21.674, CI: 1.470-319.469), disease activity (OR: 1.440; CI: 1.005-2.065), and insomnia (OR: 1.223, CI: 1.003-1.4922) were identified as independent predictors of fatigue in PSS patients. It was determined that BD alone could predict fatigue by 84% in PSS patients. Conclusion Depression can be a prominent predictor of fatigue in PSS patients. There is a need for studies evaluating the effect of antidepressant treatment approaches on fatigue accompanied by mood disturbance in PSS patients.
... Many factors can contribute to the co-occurrence of depression and inflammatory rheumatic diseases, such as common genetic risk factors and shared biological pathways, as well as the influence of social, behavioural and psychological factors 144 . Depression is also strongly linked with fatigue in inflammatory rheumatic diseases 21,28,35,47,49,81,145 as well as in non-inflammatory diseases 146 . Notably, 'marked tiredness' is one of the classification criteria for depression 147 . ...
Article
Fatigue is a complex phenomenon and an important health concern for many people with chronic inflammatory rheumatic diseases, such as rheumatoid arthritis, psoriatic arthritis, primary Sjögren syndrome and systemic lupus erythematosus. Although some clinical trials have shown the benefits of cognitive behavioural therapy in fatigue management, the effect of this approach is relatively modest, and no curative treatment has been identified. The pathogenesis of fatigue remains unclear. Despite many challenges and limitations, a growing body of research points to roles for the immune system, the central and autonomic nervous systems and the neuroendocrine system in the induction and maintenance of fatigue in chronic diseases. New insights indicate that sleep, genetic susceptibility, metabolic disturbances and other biological and physiological mechanisms contribute to fatigue. Furthermore, understanding of the relationships between psychosocial factors and fatigue is increasing. However, the interrelationships between these diverse mechanisms and fatigue remain poorly defined. In this Review, we outline various biological, physiological and psychosocial determinants of fatigue in inflammatory rheumatic diseases, and propose mechanistic and conceptual models of fatigue to summarize current understanding, stimulate debate and develop further research ideas.
Article
Full-text available
Background: What baseline predictors would be involved in mortality in people with primary Sjögren syndrome (SjS) remains uncertain. This study aimed to investigate the baseline characteristics collected at the time of diagnosis of SjS associated with mortality and to identify mortality risk factors for all-cause death and deaths related to systemic SjS activity measured by the ESSDAI score. Methods: In this international, real-world, retrospective, cohort study, we retrospectively collected data from 27 countries on mortality and causes of death from the Big Data Sjögren Registry. Inclusion criteria consisted of fulfilling 2002/2016 SjS classification criteria, and exclusion criteria included chronic HCV/HIV infections and associated systemic autoimmune diseases. A statistical approach based on a directed acyclic graph was used, with all-cause and Sjögren-related mortality as primary endpoints. The key determinants that defined the disease phenotype at diagnosis (glandular, systemic, and immunological) were analysed as independent variables. Findings: Between January 1st, 2014 and December 31, 2023, data from 11,372 patients with primary SjS (93.5% women, 78.4% classified as White, mean age at diagnosis of 51.1 years) included in the Registry were analysed. 876 (7.7%) deaths were recorded after a mean follow-up of 8.6 years (SD 7.12). Univariate analysis of prognostic factors for all-cause death identified eight Sjögren-related variables (ocular and oral tests, salivary biopsy, ESSDAI, ANA, anti-Ro, anti-La, and cryoglobulins). The multivariate CPH model adjusted for these variables and the epidemiological features showed that DAS-ESSDAI (high vs no high: HR = 1.68; 95% CI, 1.27-2.22) and cryoglobulins (positive vs negative: HR = 1.72; 95% CI, 1.22-2.42) were independent predictors of all-cause death. Of the 640 deaths with available information detailing the specific cause of death, 14% were due to systemic SjS. Univariate analysis of prognostic factors for Sjögren-cause death identified five Sjögren-related variables (oral tests, clinESSDAI, DAS-ESSDAI, ANA, and cryoglobulins). The multivariate competing risks CPH model adjusted for these variables and the epidemiological features showed that oral tests (abnormal vs normal results: HR = 1.38; 95% CI, 1.01-1.87), DAS-ESSDAI (high vs no high: HR = 1.55; 95% CI, 1.22-1.96) and cryoglobulins (positive vs negative: HR = 1.52; 95% CI, 1.16-2) were independent predictors of SjS-related death. Interpretation: The key mortality risk factors at the time of SjS diagnosis were positive cryoglobulins and a high systemic activity scored using the ESSDAI, conferring a 2-times increased risk of all-cause and SjS-related death. ESSDAI measurement and cryoglobulin testing should be considered mandatory when an individual is diagnosed with SjS. Funding: Novartis.
Article
Full-text available
Introduction Primary Sjögren’s Syndrome (pSS) affects exocrine glands such as those producing the tear film, leading to dry and painful eyes, but is also associated with fatigue. The experience of fatigue in pSS, and its relationship with sicca symptoms, is poorly understood. Methods Twenty people diagnosed with pSS were recruited to participate in a semi-structured qualitative interview about their symptoms experience. Interviews were audio-recorded, transcribed verbatim and analysed using thematic analysis. Results People with pSS described physical tiredness, mental fatigue and ocular fatigue. Mental fatigue was characterised by difficulties in attention, particularly, the ability to follow conversations and short-term memory problems. Participants linked their experience of fatigue to feeling of depression, frustration, irritation and anxiety, and therefore, fatigue was suggested to have had a large impact on their psychological well-being. People with pSS also described a range of ocular symptoms including pain, dryness, and itching, which were compounded by fatigue. For some, eye fatigue was pervasive, and daily activities involving the eyes such as reading, using the computer and driving were impaired. In some cases, the level of ocular discomfort was so severe it prevented sleep, which in turn impacted on general fatigue levels. Conclusions People with pSS experience fatigue in a range of ways; physical, mental and ocular fatigue were described. Fatigue was suggested to exacerbate other ocular symptoms, posed serious physical limitations and caused psychological distress. Further research into the nature of fatigue and ocular symptoms in pSS is required.
Article
Full-text available
BSR and BHPR Guideline for the Management of Adults with Primary Sjögren’s Syndrome Elizabeth Price1, Saaeha Rauz2, Anwar Tappuni3, Nurhan Sutcliffe4, Katie L. Hackett 5, Francesca Barone6, Guido Granata6, Wan-Fai Ng5, Benjamin A. Fisher6, Michele Bombardieri7, Elisa Astorri7, Ben Empson8, Genevieve Larkin9, Bridget Crampton10 and Simon Bowman11 on behalf of the BSR and BHPR Standards, Guideline and Audit Working Group 1Department of Rheumatology, Great Western Hospital NHS Foundation Trust; 2Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, and Birmingham and Midland Eye Centre, UK; 3Queen Mary, University of London; 4Department of Rheumatology, Barts Health NHS Trust; 5Institute of Cellular Medicine, Faculty of Medical Sciences, Newcastle University, UK and Newcastle upon Tyne Hospitals NHS Foundation Trust, UK; 6Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, UK and Department of Rheumatology, Queen Elizabeth Hospital, Birmingham, UK; 7Centre for Experimental Medicine and Rheumatology, Queen Mary, University of London; 8Modality partnership, The Laurie Pike Health Centre, Birmingham; 9Kings College Hospital, London; 10British Sjögren’s Syndrome Association, Birmingham, UK; 11Queen Elizabeth Hospital, Birmingham, UK Correspondence to: Elizabeth Price, Great Western Hospital NHS Foundation Trust, Swindon, SN3 6BB, UK. E-mail: Elizabeth.price@gwh.nhs.uk Key words: Sjögren’s, guideline, management, sicca Executive Summary Scope and Purpose Background Primary Sjögren’s syndrome (pSS) is a chronic, immune-mediated, condition (1) typically presenting in women in their 5th or 6th decade, although up to 10% of cases occur in men. The prevalence in women in the UK is 0.1-0.4% (2). Patients characteristically present with dryness of the eyes and mouth but systemic features are common and B-cell lymphoma affects 5-10% (3, 4). Objective of Guideline This document aims to provide a pragmatic, practical guideline for the management of adults with pSS. Target audience The target audience includes rheumatologists, general physicians, general practitioners, specialist nurses and other specialists (e.g. ophthalmologists, dental practitioners and Ear, Nose and Throat Specialists). Areas NOT covered This guideline does not cover the detailed management of patients with secondary Sjögren’s or patients with Lymphoma who should be managed in conjunction with oncologists and haematologists. The management of children is not specifically covered although is similar to that for adults with special emphasis on good dental care and hygiene. Rigor of Development & limitations A search was undertaken for all relevant evidence from 1990 to January 2016. The level of evidence was graded from Ia through to IV and A through to D. See appendix 1 for full details of search criteria and Level of Evidence (LOE) definitions. A Strength of Agreement (SOA) was calculated for each recommendation and the results expressed as an SOA score (0-10) with percentage of respondents scoring the recommendation ≥7 in brackets. Recommendations were only included where the mean SOA was ≥7 and >75% respondents scored ≥7. The Guideline Eyes I. Conservation of tears by humidification and avoidance of systemic medication that exacerbates dryness LOE Ib/A; SOA 8.9 (97%) II. Stimulate meibomian gland secretion daily using warm compresses, commercially available eye bags or other devices and perform lid hygiene if required. LOE III/C; SOA 8.9 (93.3%) III. If persistent meibomian gland inflammation and blepharitis treatment with doxycycline 50mg od for a minimum of 3 months may be effective. LOE IV/D; SOA8.83 (92.9%) IV. Early referral to an ophthalmologist for review and consideration of insertion of punctual plugs or cauterisation. LOE II/B; SOA 9.27 (100%) V. Recommend liposomal sprays to reduce evaporative tear loss and replace the meibomian gland layer LOE III/C; SOA 8.29 (92.8%) VI. Start with simple lubricating drops using the most cost-effective options (see Table 1 for preparations) LOE IV/D; SOA 9.3 (93.3%) VII. Avoid preservative containing preparations LOE I/A; SOA 9.21 (92.9%) VIII. Refer patients with severe dry eye, not responding to conventional treatment, to specialist commissioned centres for consideration of serum eye drops. LOE IIb/B; SOA 9.5 (100%) IX. Low dose steroid containing eye drops for short term use under ophthalmic supervision only LOE Ib/A; SOA 9.64 (100%) X. Avoid long term use of topical steroids LOE Ib/A; SOA 9.64 (100%) XI. Ciclosporin eye drops or ointments under ophthalmic supervision for chronic inflammation LOE Ib/A; SOA 9.38 (100%) XII. Use topical NSAIDs with caution under ophthalmic supervision only LOE IIb/B; SOA 9.86 (100%) XIII. A trial of Pilocarpine 5mg once daily increasing stepwise to 5mg qds is recommended for patients with significant sicca symptoms. LOE IIb/B; SOA 9.08 (92.3%) XIV. Prescribe mucolytic eye drops to patients with mucous threads or ocular surface filaments LOE III/C; SOA 9.38 (100%) XV. Refer patients with severe dry eye and associated blepharospasm to a specialist centre for consideration of Botulinum toxin treatment LOE III/C; SOA 9.17 (100%) XVI. Refer patients with severe dry eye and corneal ulceration, not responding to conventional treatment, to specialist centre for consideration of ‘bandage’ contact lenses or corneal grafting. LOE III/C; SOA 9.15 (92.9%) Mouth I. Advise excellent oral hygiene, limit sugar intake and avoid food and drinks other than plain water between meals and from 1 hour before bedtime and through the night LOE IV/D; SOA 9.8 (100%) II. Assessment by an Oral Medicine specialist and/or regular visits to a general dental practitioner LOE IV/D; SOA 9.8 (100%) III. Avoid acidic and sugar containing products in dentate patients. LOE IV/D SOA 9.87 (100%) IV. Humidify the environment LOE III/C; SOA 9.13 (100%) V. Brush teeth at least twice daily (but not immediately after eating) including before bed using a pea sized amount of high fluoride toothpaste and use fluoride containing oral gel on teeth twice daily LOE I/A; SOA 9.8 (100%) VI. Alcohol free chlorhexidene mouth wash twice daily for maximum of 2 weeks every 3 months can help prevent gum disease LOE IV/D; SOA 9.0 (100%) VII. Use fluoride containing mouth wash, gel or spray as required for symptomatic relief. LOE III/C; SOA 9.43 (92.8%) VIII. Chew xylitol containing sugar free gum. LOE IIb/B; SOA 9.67 (100%) IX. A trial of Pilocarpine 5mg once daily increasing stepwise to 5mg qds if significant sicca symptoms. LOE IIb/B; SOA 9.77 (100%) Treatment of oral Candida I. Ssimple candida infection (visible white plaques) - oral nystatin liquid 1ml five times daily for 7 days. Repeat for 1 week in 8 if frequent recurrence. LOE IV/D; SOA 9.86 (100%) II. Erythematous infection (red, raw tongue or oral cavity) - fluconazole 50mg od for 10 days. LOE IV/D; SOA 9.86 (100%) III. Treat angular cheilitis with miconazole topically for 2 weeks. LOE IV/D; SOA 9.86 (100%) Management of Salivary Gland enlargement I. Consider baseline ultrasound to assess for active inflammation, infection and stones. LOE IV/D; SOA 9.7 (100%) II. If acute inflammation, in the absence of infection and stones, consider short course of oral prednisolone or intra-muscular Depomedrone. LOE IV/D; SOA 9.01 (100%) III. Massaging the glands reduces inflammation in chronically inflamed glands LOE IV/D; SOA 9.31 (92.8%) Systemic dryness I. A trial of Pilocarpine 5mg once daily increasing stepwise to 5mg qds if systemic dryness. LOE IV/D; SOA 9.08 (92.3%) II. Anon-hormonal vaginal moisturiser +/-topical oestrogen if vaginal dryness. LOE I/A; SOA 9.8 (100%) Treatment of Systemic Disease Non-pharmacological interventions for systemic disease I. Advise a graded exercise programme for fatigue. LOE IIb/B; SOA 9.33 (93.3%) II. Provide written information and details of appropriate support groups and on-line resources LOE IV/D; SOA 9.8 (100%) Pharmacological treatment for systemic disease I. Hydroxychloroquine (6mg/kg) for those with skin, joint disease or fatigue LOE IIa/B; SOA 9.64 (100%) II. Ciclosporin A may be helpful in patients with significant joint involvement LOE III/C; SOA 8.58 (85.7%) III. Azathioprine may be considered in patients with systemic complications. LOE III/C; SOA 9.09 (100%) IV. Methotrexate is useful for patients with an associated inflammatory arthritis. LOE IV/D SOA 9.54 (100%) V. Mycophenolate may be considered in patients with systemic complications. LOE III/C; SOA 9.1 (100%) Corticosteroids I. Intermittent short courses of oral or intramuscular steroid for systemic flares and significant organ manifestations with or without additional immunosuppressive treatment. LOE III/C; SOA 9.2 (100%) II. Low dose oral prednisolone for persistent constitutional symptoms in patients with inadequate response to other immunosuppresants. LOE IIb/B; SOA 8.92 (100%) Cyclophosphamide I. Cyclophosphamide (usually in combination with steroids) for patients with organ threatening systemic complications. LOE III/C; SOA 9.09 (100%) Rituximab I. Rituximab for specialist use in patients with significant systemic manifestations refractory to other immunosuppresives and those with lymphoma, Immune thrombocytopaenia, vasculitic neuropathy or cryoglobulinaemia. LOE IIb/B; SOA 9.43 (100%) Intravenous Immunoglobulins I. Immunoglobulin treatment for Sjögren’s associated myositis and neuropathies if the patient has failed to respond to treatment with conventional immunosuppression. LOE III/C; SOA 9.43 (100%) Colchicine I. Colchicine as adjunctive treatment if cutaneous manifestations or pericarditis not responding to other treatments. LOE III/C; SOA 8.45 (85.7%) Dapsone I. Dapsone as adjunctive treatment if cutaneous manifestations not responding to hydroxychloroquine. LOE III/C; SOA 8.8 (100%) Topical Tacrolimus I. Topical Tacrolimus as adjunctive treatment if cutaneous manifestations not responding to hydroxychloroquine. LOE III/C; SOA 8.75 (100%) Treatments NOT recommended I. Leflunomide and Penicillamine are not routinely recommended for Primary Sjögren’s Syndrome. Level of evidence III/C; SOA 9.8 (100%) II. Belimumab and Abatacept are not currently recommended although they merit further study. Level of evidence IIb/B & III/C; SOA 9.46 (92.8%) III. Anti-TNF, IFN alpha and Anakinra therapies are not recommended for treatment of Primary Sjögren’s Syndrome Level of evidence Ib/A; SOA 9.38 (85.7%) IV. Tociluzimab and Efalizuab are not recommended for the treatment of Primary Sjögren’s Syndrome. Level of evidence III/C; SOA 9.82 (100%) V. Dehydroepiandrosterone (DHEA) substitution treatment is not recommended in Primary Sjögren’s Syndrome. Level of evidence Ib/A; SOA 9.45 (100%) Management of Pregnancy I. Consider low dose aspirin to improve placental implantation LOE IIb/B; SOA 8.27 (92.8%) II. Monitor with serial US if anti Ro and/or La positive and consider referral to specialist centre LOE IIb/B; SOA 9.71 (100%) III. Review all medication in pregnancy. Hydroxychloroquine may be continued throughout pregnancy and breastfeeding LOE I/A; SOA 7.91 (85.7%) Assessment & Management of Lymphoma I. Review high risk patients regularly and warn patients to report firm, painless glandular swelling that doesn’t settle LOA IV/D; SOA 9.86 (100%) II. Investigate suspicious lesions with local US, biopsy and CT chest, abdomen and pelvis for staging LOA III/C; SOA9.77 (100%) Conclusion pSS is a chronic, debilitating condition that warrants effective management. All patients should be counselled and offered topical management for sicca symptoms. Systemic treatment should be considered early in those with constitutional symptoms. Reference 1. Fisher BA, Brown RM, Bowman SJ, Barone F. A review of salivary gland histopathology in primary Sjögren's syndrome with a focus on its potential as a clinical trials biomarker. Ann Rheum Dis. 2015;74(9):1645-50. 2. Bowman SJ, Ibrahim GH, Holmes G, Hamburger J, Ainsworth JR. Estimating the prevalence among Caucasian women of primary Sjogren's syndrome in two general practices in Birmingham, UK. Scandinavian journal of rheumatology. 2004;33(1):39-43. 3. Ramos-Casals M, Solans R, Rosas J, Camps MT, Gil A, Del Pino-Montes J, et al. Primary Sjogren syndrome in Spain: clinical and immunologic expression in 1010 patients. Medicine. 2008;87(4):210-9. 4. Ramos-Casals M, Brito-Zeron P, Solans R, Camps MT, Casanovas A, Sopena B, et al. Systemic involvement in primary Sjogren's syndrome evaluated by the EULAR-SS disease activity index: analysis of 921 Spanish patients (GEAS-SS Registry). Rheumatology (Oxford, England). 2014;53(2):321-31.
Article
Full-text available
Objectives: To identify whether sleep disturbances are more prevalent in primary SS (pSS) patients compared with the general population and to recognize which specific sleep symptoms are particularly problematic in this population. Methods: Electronic searches of the literature were conducted in PubMed, Medline (Ovid), Embase (Ovid), PsychINFO (Ovid) and Web of Science and the search strategy registered a priori Titles and abstracts were reviewed by two authors independently against a set of prespecified inclusion/exclusion criteria, reference lists were examined and a narrative synthesis of the included articles was conducted. Results: Eight whole-text papers containing nine separate studies met the inclusion criteria and were included in the narrative analysis. Few of these studies met all of the quality assessment criteria. The studies used a range of self-reported measures and objective measures, including polysomnography. Mixed evidence was obtained for some of the individual sleep outcomes, but overall compared with controls, pSS patients reported greater subjective sleep disturbances and daytime somnolence and demonstrated more night awakenings and pre-existing obstructive sleep apnoea. Conclusions: A range of sleep disturbances are commonly reported in pSS patients. Further polysomnography studies are recommended to confirm the increased prevalence of night awakenings and obstructive sleep apnoea in this patient group. pSS patients with excessive daytime somnolence should be screened for co-morbid sleep disorders and treated appropriately. Interventions targeted at sleep difficulties in pSS, such as cognitive behavioural therapy for insomnia and nocturnal humidification devices, have the potential to improve quality of life in this patient group and warrant further investigation.
Article
Full-text available
OBJECTIVES: This article reports relationships between serum cytokine levels and patient-reported levels of fatigue, in the chronic immunological condition primary Sjögren's syndrome (pSS). METHODS: Blood levels of 24 cytokines were measured in 159 patients with pSS from the United Kingdom Primary Sjögren's Syndrome Registry and 28 healthy non-fatigued controls. Differences between cytokines in cases and controls were evaluated using Wilcoxon test. Patient-reported scores for fatigue were evaluated, classified according to severity and compared with cytokine levels using analysis of variance. Logistic regression was used to determine the most important predictors of fatigue levels. RESULTS: 14 cytokines were significantly higher in patients with pSS (n=159) compared to non-fatigued healthy controls (n=28). While serum levels were elevated in patients with pSS compared to healthy controls, unexpectedly, the levels of 4 proinflammatory cytokines-interferon-γ-induced protein-10 (IP-10) (p=0.019), tumour necrosis factor-α (p=0.046), lymphotoxin-α (p=0.034) and interferon-γ (IFN-γ) (p=0.022)-were inversely related to patient-reported levels of fatigue. A regression model predicting fatigue levels in pSS based on cytokine levels, disease-specific and clinical parameters, as well as anxiety, pain and depression, revealed IP-10, IFN-γ (both inversely), pain and depression (both positively) as the most important predictors of fatigue. This model correctly predicts fatigue levels with reasonable (67%) accuracy. CONCLUSIONS: Cytokines, pain and depression appear to be the most powerful predictors of fatigue in pSS. Our data challenge the notion that proinflammatory cytokines directly mediate fatigue in chronic immunological conditions. Instead, we hypothesise that mechanisms regulating inflammatory responses may be important.
Article
Full-text available
Objectives This article reports relationships between serum cytokine levels and patient-reported levels of fatigue, in the chronic immunological condition primary Sjögren's syndrome (pSS). Methods Blood levels of 24 cytokines were measured in 159 patients with pSS from the United Kingdom Primary Sjögren's Syndrome Registry and 28 healthy non-fatigued controls. Differences between cytokines in cases and controls were evaluated using Wilcoxon test. Patient-reported scores for fatigue were evaluated, classified according to severity and compared with cytokine levels using analysis of variance. Logistic regression was used to determine the most important predictors of fatigue levels. Results 14 cytokines were significantly higher in patients with pSS (n=159) compared to non-fatigued healthy controls (n=28). While serum levels were elevated in patients with pSS compared to healthy controls, unexpectedly, the levels of 4 proinflammatory cytokines—interferon-γ-induced protein-10 (IP-10) (p=0.019), tumour necrosis factor-α (p=0.046), lymphotoxin-α (p=0.034) and interferon-γ (IFN-γ) (p=0.022)—were inversely related to patient-reported levels of fatigue. A regression model predicting fatigue levels in pSS based on cytokine levels, disease-specific and clinical parameters, as well as anxiety, pain and depression, revealed IP-10, IFN-γ (both inversely), pain and depression (both positively) as the most important predictors of fatigue. This model correctly predicts fatigue levels with reasonable (67%) accuracy. Conclusions Cytokines, pain and depression appear to be the most powerful predictors of fatigue in pSS. Our data challenge the notion that proinflammatory cytokines directly mediate fatigue in chronic immunological conditions. Instead, we hypothesise that mechanisms regulating inflammatory responses may be important.
Article
The common approach to the multiplicity problem calls for controlling the familywise error rate (FWER). This approach, though, has faults, and we point out a few. A different approach to problems of multiple significance testing is presented. It calls for controlling the expected proportion of falsely rejected hypotheses — the false discovery rate. This error rate is equivalent to the FWER when all hypotheses are true but is smaller otherwise. Therefore, in problems where the control of the false discovery rate rather than that of the FWER is desired, there is potential for a gain in power. A simple sequential Bonferronitype procedure is proved to control the false discovery rate for independent test statistics, and a simulation study shows that the gain in power is substantial. The use of the new procedure and the appropriateness of the criterion are illustrated with examples.
Article
Objectives: Besides ocular and oral dryness, fatigue is a major symptom in patients with primary Sjögren's syndrome (pSS). Our aim was to investigate the importance of fatigue in relation to other symptoms experienced as well as to evaluate the effect of rituximab treatment on fatigue in pSS patients with active disease. Methods: This analysis was based on data from our open-label rituximab study in 28 pSS patients. Symptoms of dryness, physical fatigue, pain, and mental fatigue were scored on 0-10 scales (according to ESSPRI). Systemic disease activity was assessed with ESSDAI. Results: At baseline, 24 (86%) patients rated physical fatigue as the complaint most eligible for improvement (median importance of 10), followed by pain, dryness, and mental fatigue. After rituximab treatment, physical fatigue showed maximum improvement of 2.5 points and 31% in median values at group level, and 10 (36%) patients reached physical fatigue score<5 representing patient-acceptable symptom state (PASS). In comparison, systemic disease activity improved 5.5 points and 73% at group level, and 22 (79%) patients reached ESSDAI<5 representing low disease activity. GEE analysis over time revealed that physical fatigue was significantly associated with absolute number of B cells, dryness and mental fatigue, but not with ESSDAI, IgG levels and IgM-RF. Conclusions: Physical fatigue characterises patient experience of pSS. Rituximab treatment resulted in significant improvement of patient-reported symptoms. However, the large majority of patients still experienced physical fatigue at an unsatisfactory level, above the cut-off value for PASS. Therefore, attention for optimal management of this prominent symptom is warrante.