Subido por nosellama

Fibromialgia: Actualizacion de características clínicas

Anuncio
REVIEWS
­ ibromyalgia: an update on clinical
F
characteristics, aetiopathogenesis
and treatment
Piercarlo Sarzi-Puttini
1
✉, Valeria Giorgi
, Daniela Marotto
1
2
and Fabiola Atzeni3
Abstract | Fibromyalgia is characterized by chronic widespread pain, fatigue, sleep disturbances
and functional symptoms. The etiopathogenesis, diagnostic criteria and classification criteria of
fibromyalgia are still debated and, consequently, so are the strategies for treating this condition.
Fibromyalgia is the third most frequent musculoskeletal condition, and its prevalence increases
with age. However, although diagnosis has improved with the evolution of more accurate diag­
nostic criteria, a considerable proportion of physicians still fail to recognize the syndrome.
Many factors contribute to the development of fibromyalgia in a unique manner: genetic pre­
disposition, personal experiences, emotional–cognitive factors, the mind–body relationship and
a biopsychological ability to cope with stress. The multiple components of the pathogenesis
and maintenance of the condition necessitate a multi-modal treatment approach. Individually
tailored treatment is an important consideration, with the increasing recognition that different
fibromyalgia subgroups exist with different clinical characteristics. Consequently, although an
evidence-based approach to fibromyalgia management is always desirable, the approach of
physicians is inevitably empirical, and must have the aim of creating a strong alliance with the
patient and formulating shared, realistic treatment goals.
Biopsychosocial model
of medicine
An interdisciplinary model
commonly used in the field of
chronic pain that incorporates
the interactions among bio­
logical factors (such as physiopathological factors), psychosocial factors (that is, emotional
factors, such as distress or fear)
and behavioural factors.
1
Rheumatology Unit, ASST
Fatebenefratelli-Sacco, Luigi
Sacco University Hospital,
Milan, Italy.
2
Rheumatology Unit, ATS
Sardegna, Paolo Dettori
Hospital, Tempio Pausania,
Italy.
3
Rheumatology Unit,
Department of Internal and
Experimental Medicine,
University of Messina,
Messina, Italy.
✉e-mail: piercarlo.
[email protected]
https://doi.org/10.1038/
s41584-020-00506-w
Fibromyalgia or fibromyalgia syndrome is one of the
most common causes of chronic widespread pain
(CWP), but, although pain is its main and distinguishing feature, fibromyalgia is characterized by a complex
polysymptomatology that also comprises fatigue, sleep
disturbances and functional symptoms (that is, medical
symptoms not explained by structural or pathologically
defined causes). Fibromyalgia is quite a common condition in the general population1,2; however, no consistently effective treatments are yet available owing to a
lack of consensus regarding fibromyalgia diagnostic
and classification criteria and, especially, regarding
fibromyalgia aetiopathogenesis. Indeed, fibromyalgia has proven to be a mysterious syndrome and is an
interesting condition as far as philosophy of medicine
is concerned, because it falls outside the mechanistic
definition of disease3.
In this Review, we provide a comprehensive, critical
overview on the burden, diagnosis and treatment of
fibromyalgia, considering the latest research, guidelines
and clinical experience. We describe clinical aspects of
this syndrome, including the different diagnostic criteria developed over time. We also bring together various
hypotheses of fibromyalgia aetiopathogenesis, keeping
in mind the biopsychosocial model of medicine and the
Nature Reviews | Rheumatology
complex mind–body relationship. In particular, we
herein hypothesize that chronic pain and fibromyalgia
might rise both from a bottom-up (body periphery to
central nervous system) and a top-down (central nervous system to body periphery) mechanism, so that a
psychological pathogenic process (for example, trauma
or stress) can coexist with, but is not necessary for, a
physical pathogenic process (for example, an inflammatory or degenerative process). Finally, we discuss
fibromyalgia treatment, delving into the most effective
and the latest, most promising treatment strategies,
keeping in mind the importance of an individualized,
patient-centred perspective. We try to provide a novel
and practical management workflow for physicians,
based on clinical expertise and the latest EULAR criteria
for managing fibromyalgia4, to be used in their everyday
clinical practice.
Epidemiology
The reported prevalence of fibromyalgia varies depending on the diagnostic criteria used to define this condition. Studies using the 1990 ACR criteria have recorded
prevalence rates that range from 0.4% (Greece) to 8.8%
(Turkey), with a mean estimated global prevalence of
2.7%. The average worldwide female to male ratio for
Reviews
Key points
• Fibromyalgia is a fairly common syndrome in the general population, reaching a
prevalence of 2–3% worldwide.
• The complex polysymptomatology of fibromyalgia comprises not only chronic
widespread pain, fatigue and sleep alterations but also autonomic disturbances,
cognitive dysfunction, hypersensitivity to external stimuli, somatic symptoms and
psychiatric disorders.
• Owing to the subjectivity of the symptoms and the lack of biomarkers, diagnosis is
exquisitely clinical, and diagnostic criteria are constantly evolving; early diagnosis
and prevention are still elusive goals.
• Fibromyalgia severity and progression or improvement can be evaluated by means
of a plethora of composite tests.
• Fibromyalgia pathogenesis is not fully understood; hypotheses state that genetic
predisposition, stressful life events, peripheral (inflammatory) and central
(cognitive–emotional) mechanisms interplay to create pain dysperception owing
to neuromorphological modifications (‘nociplastic pain’).
• Treatment should be multimodal and built on four pillars (patient education; fitness;
pharmacotherapy; and psychotherapy); the approach should be individualized,
symptom-based and stepwise, establishing shared goals with the patient.
Paraesthesia
A qualitative alteration of
the sensitivity of the skin
(which can be an abnormal
sensation of pricking, tingling
and numbness).
Fibro-fog
A symptom of fibromyalgia
involving an inability to think
clearly or difficulties in
concentrating.
Raynaud phenomenon
A condition that causes
decreased blood flow to
the extremities (such as the
fingers, toes, ears and nose)
due to vasospasm; such
spasms occur in response to
cold, stress or emotional
upset.
fibromyalgia is 3:1 (ref.1). One study of five European
countries (France, Portugal, Spain, Germany and Italy)5
estimated a general population prevalence of 4.7%.
However, these prevalence rates might vary by as much
as four times when considering subsequent criteria
sets2. Examples of the prevalence of fibromyalgia estimated by different studies for various countries1,5–16 are
shown in Fig. 1.
Fibromyalgia is the third most common musculoskeletal condition in terms of prevalence, after lumbar
pain and osteoarthritis17. Prevalence is proportional to
the age of the population, peaking at 50–60 years old11.
However, these prevalence estimates might be inaccurate: discrepancies exist between administrative data
(that is, rates reported by patients) and epidemiological
data (that is, data based on a diagnosis made by the physician)18, as a substantial proportion of physicians still
fail to recognize the syndrome.
The generally poor quality of life of patients with
fibromyalgia is reflected by the massive health-care costs
of patients, who frequently seek medical attention. The
annual number of consultations required is almost double that of healthy individuals19, and total health-care
costs are estimated to be three times higher for patients
with fibromyalgia than for other individuals, as assessed
by comparing costs among patients with those of a random population sample20. Indirect societal costs are also
high, mainly because of lost working productivity21,22: one
study showed that 24.3% of the patients involved in the
study stopped working 5 years after fibromyalgia onset23.
Clinical features and diagnosis
The symptoms of fibromyalgia
Fibromyalgia is a complex, chronic pain condition that
primarily (but not only) involves the musculoskeletal
system. Unlike other rheumatic diseases, fibromyalgia
does not manifest by means of visible clinical signs: a
physical examination only reveals greater sensitivity
to pressure at some specific points (‘tender points’),
although these regions tend to be more tender than other
regions in most individuals, regardless of whether they
have fibromyalgia24. Fibromyalgia symptomatology is
summarized in Fig. 2. In this section, we provide a brief
description of each symptom.
Cardinal features. In fibromyalgia, pain can affect the
whole body from head to toe. Patients with fibromyalgia use a plethora of pain descriptors, and their pain is
often described as being similar to neuropathic pain25:
20–30% of patients report paraesthesia in the limbs,
hands or trunk, which is commonly described as a tingling sensation or pins-and-needles26. The type, location
and severity of pain depends on a number of modulating
factors, the most important of which are working activities, comorbidities (such as obesity27) and variations in
temperature28,29. Physical or mental stress is also a known
factor associated with worsening pain22,30.
The other two most frequent symptoms of fibromyalgia are fatigue and sleep disturbances31–33. Fatigue might
be physical or mental. The degree of fatigue varies widely
from mild tiredness to a state of exhaustion similar to
that experienced during viral diseases such as influenza.
Sleeping problems include any type of insomnia or frequent awakenings. Non-restorative sleep is especially
preponderant and, even if the quality and duration of
sleep is normal, patients with fibromyalgia often report
the feeling of not having had enough rest31,32.
Other common features. Cognitive dysfunction (especially ‘fibro-fog’) and memory deficits are among the
more severe symptoms of patients with fibromyalgia33.
Depression, anxiety, pain or sleeping problems can all
have a negative effect on cognitive symptoms, but they
do not entirely explain all the cognitive symptoms of
patients with fibromyalgia34.
Patients with fibromyalgia often complain about
many other clinical symptoms involving almost all
organs and systems, the severity of which varies from
patient to patient and within each patient during the
syndrome course35. Idiopathic, regional pain syndromes
are particularly common. Headache with or without a
history of migraine is very frequent, and the reverse
is also true, with fibromyalgia being frequent among
individuals who have episodic migraines36. Dyspepsia,
abdominal pain and alternating constipation and diarrhoea are also common symptoms, and might be part of
a full-blown irritable bowel syndrome37. Many patients
experience genitourinary disorders (such as urinary
urgency in the absence of urinary tract infections38,
dysmenorrhoea or vulvar vestibulitis, which leads to
difficulties in sexual intercourse39). Another frequent
symptom is stiffness33,40, although morning stiffness does
not usually exceed 60 min.
Autonomic disturbances manifest in all body areas
and correlate with the severity of the condition41,42.
Patients can report a subjective feeling of dry mouth
(xerostomia) and eyes (xerophthalmia), blurred vision
and photophobia, and Raynaud phenomenon 43. Over
30% of patients develop lower limb discomfort and a
need to move their legs continuously (restless legs syndrome)44–46. Patients with fibromyalgia often report a
feeling of instability or staggering as well, especially after
standing upright for prolonged periods47.
www.nature.com/nrrheum
Reviews
Canada 3.3%
Denmark 0.7%
France, Italy,
Germany, Spain,
Portugal 2.9–4.7%
Japan 2.1%
Italy 3.6%
USA 6.4%
Turkey 8.8%
Hong Kong 0.8%
Lebanon 1%
Tunisia 9.3%
Israel 2–2.6%
Brazil 2.5%
Criteria and/or questionnaire
Country or region
Study
Total prevalence (%)
1990 ACR
Hong Kong
Scudds et al. (2006)13
0.8
Denmark
Prescott et al. (1993)15
0.7
Italy
Salaffi et al. (2005)
3.6
Turkey
Turhanoglu et al. (2008)8
8.8
The 2010 ACR criteria
16
Japan
Nakamura et al. (2014)
2.1
USA
Vincent et al. (2013)12
6.4
Israel
Ablin et al. (2012)
LFESSQ-4: 2.6
LFESSQ-6: 2.0
France, Italy, Germany,
Spain and Portugal
Branco et al. (2009)5
LFESSQ-4: 4.7
LFESSQ-6: 2.9
COPCORD
Brazil
Rodrigues Senna et al. (2004)10
2.5
LFESSQ and the 1990 ACR criteria
Canada
White et al. (1999)11
3.3
Tunisia*
Guermazi et al. (2008)
9.3
Lebanon
Chaaya et al. (2011)
1
World
Queiroz (2013)1
2.7
LFESSQ
COPCORD and the 1990 ACR criteria
7
14
9
6
Fig. 1 | estimated prevalence of fibromyalgia in different regions using different diagnostic criteria or questionnaires.
The prevalence of fibromyalgia has been estimated in different regions worldwide using various diagnostic criteria and
questionnaires, such as the London Fibromyalgia Epidemiology Study Screening Questionnaire (LFESSQ; shown in light
green), the Community Oriented Program for the Control of Rheumatic Diseases (COPCORD; shown in dark green), the
ACR 1990 classification criteria (shown in blue) and the ACR 2010 diagnosis criteria (shown in red). It should be noted
that direct comparisons of the prevalence in different regions cannot be made owing to the use of different assessment
methodologies. *Individuals with a positive screening test were invited for examination to confirm or exclude the presence
of fibromyalgia by applying the 1999 ACR criteria.
Psychologically, patients with fibromyalgia are
characterized by a preponderant negative affect, that
is, the presence of negative emotions associated with
a generalized distress state48. This state of psychological suffering can accompany full-blown psychiatric
disorders, which are frequent in patients with fibromyalgia and can notably affect the lives of the patients
and even the severity of the syndrome49. The lifetime
prevalence of anxiety disorders in patients with fibromyalgia is 60%, and depression is observed in 14–36%
of patients compared with 6.6% of healthy individuals50.
In a Danish population of patients with fibromyalgia, the
risk of suicide was ten times higher than in the general
Nature Reviews | Rheumatology
population51, which was confirmed by a subsequent systematic review52. However, depressive symptoms are not
reported more frequently for patients with fibromyalgia
than for patients with other painful conditions such as
rheumatoid arthritis or cancer, and might be related to
maladaptive coping with psychological distress30.
Diagnostic criteria
Ongoing research has so far led to the publication of
at least five different sets of classification and diagnostic criteria for fibromyalgia over the past 30 years or so
(a summary is shown in Table 1). The earliest criteria
sets described fibromyalgia as a CWP disorder with
Reviews
various associated symptoms53, but the 1990 ACR classification criteria43 only considered CWP (defined as
pain on the left and right sides of the body, above and
below the waist, and axial skeletal (cervical or thoracic
spine, anterior chest or low back) pain), and tenderness
(defined as pain upon the palpation of ≥11 out of 18 tender point sites), and did not include other symptoms or
exclusion criteria. However, the requirement of a tender
point examination (which is examiner dependent and
intrinsically intra-individually and inter-individually
variable) made the 1990 ACR criteria impractical for
use in a clinical setting. The subsequent 2010 and 2011
ACR criteria54,55 changed the definition of fibromyalgia
to that of a multi-symptom disorder and removed the
tender point examination as a diagnostic requirement;
however, although the criteria returned to considering
the associated symptoms as important, there was perhaps
too little emphasis on the core symptom of chronic pain.
The 2016 revisions to the 2010/2011 ACR diagnostic
criteria56 highlighted the concept of ‘generalized pain’,
which also lies at the heart of the ACTTION-APS Pain
Taxonomy diagnostic criteria published in 2018 (ref.40).
Psychiatric symptoms
• Anxiety
• Depression
• Post-traumatic stress
disorder
In developing these criteria, the Fibromyalgia Working
Group concentrated on generalized pain (defined as
multi-site pain), sleeping problems and fatigue, but
also considered other supportive diagnostic features
such as cognitive disturbances, tenderness to the touch,
musculoskeletal stiffness and environmental sensitivity
(for example, sensitivity to cold, light or noise) with the
aim of providing more practical criteria.
The central problem and barrier to fibromyalgia
diagnosis is a lack of biomarkers. Researchers over the
past 5 years have investigated new molecules that might
help diagnosis and monitoring (including microRNA,
and proteome and metabolome analysis), but, although
the results have been promising, this area of research is
still in its infancy57.
In brief, the diagnosis of fibromyalgia is exquisitely
clinical. A physical examination is not diagnostically useful because of its poor validity and poor reproducibility18,
but is essential for excluding other diseases that might
explain the presence of pain and fatigue. Fibromyalgia has
no pathognomonic feature, and so diagnostic clues have
to be collected by means of thorough history taking18.
Cognitive dysfunctions
• Concentration
difficulties
• Memory deficits
Sleep disturbances
• Insomnia
• Frequent awakening
• Non-restoring sleep
Autonomic disturbances
• Blurred vision, photophobia
and xerophthalmia
• Feeling of instability
• Xerostomia
• Variations in responses
to cold at the extremities
(including Raynaud
phenomenon)
• Orthostatic hypotension
Pain
• Generalized
(head-to-toes)
• Described in terms
of neuropathic pain,
paraesthesias
Fatigue
• Physical
• Mental
Regional pain syndromes
• Migraine or headache
• Stomach ache or dyspepsia
• Abdominal pain or
irritable bowel syndrome
• Dysmenorrhoea
• Vulvodynia
• Dysuria
Hypersensitivity to external
stimuli
• Hypersensitivity to light,
odours and sounds
• Chemical sensitivity
Stiffness
• Morning stiffness not
exceeding 60 min
Cardinal features
Other common features
Fig. 2 | Principal fibromyalgia symptoms. Fibromyalgia has a complex symptomatology. Symptoms can be are divided in
two groups: cardinal features (shown in pink), which include the most characteristic fibromyalgia symptoms that are
pivotal for a diagnosis according to the latest criteria, and other common features (shown in in grey).
www.nature.com/nrrheum
Reviews
Table 1 | the evolving classification and diagnostic criteria for fibromyalgia
Criteria set
measures of pain
ACR 1990
classification
criteria
Widespread pain noted as pain in all
four quadrants (both the left and right
side of the body, above and below the
waist); plus axial skeletal pain (pain in
the cervical spine or anterior chest or
thoracic spine or low back)
Use of WPI: a 0–19 count of the body
ACR 2010
regions reported as painful by the
preliminary
diagnostic criteria patient over the past weeka
tender
points
Ref.
Widespread pain and at least 11
tender points for at least 3 months
43
No
Various symptoms included
in an SSS, a score of the sum
of severity of three symptoms
(fatigue, waking unrefreshed,
cognitive symptoms) plus
somatic symptoms in general (on
a 0–12 scale)
WPI ≥7 and SSS ≥5; or WPI 3–6
and SSS ≥9
55
Various symptoms included
in an SSS, a score of the sum
of severity of three symptoms
(fatigue, waking unrefreshed,
cognitive symptoms) plus
the sum of the number of the
following symptoms occurring
during the previous 6 months:
headaches, pain or cramps in the
lower abdomen and depression
(on a 0–12 scale)
WPI ≥7 and SSS ≥5; or WPI 3–6
and SSS ≥9
Various symptoms included
in an SSS, a score of the sum
of severity of three symptoms
(fatigue, waking unrefreshed,
cognitive symptoms) plus
the sum of the number of the
following symptoms occurring
during the previous 6 months:
headaches, pain or cramps in the
lower abdomen and depression
WPI ≥7 and SSS ≥5; or WPI 4–6
and SSS ≥9
Moderate to severe sleep
problems or moderate to severe
fatigue
MSP ≥6
No
2016 revisions to
the 2010/2011
ACR fibromyalgia
diagnostic criteria
No
Use of WPI: a 0–19 count of the body
regions reported as painful by the
patient over the past weeka
Use of MSP: a 0–9 count of the
AAPT core
diagnostic criteria number body sites reported as painful
(the sites consisting of the head, right
for fibromyalgia
arm, left arm, chest, abdomen, upper
back and spine, lower back and spine
(including buttocks), left leg and
right leg)
Diagnosis or classification
Yes (≥11 None included
out of
18)
ACR 2011
Use of WPI: a 0–19 count of the body
modifications
regions reported as painful by the
of the ACR
patient over the past weeka
preliminary
diagnostic criteria
(designed for
epidemiological
and clinical
studies, and not
for clinical
diagnosis)b
Generalized pain defined as pain in
at least 4 out of 5 regions (left upper
region, right upper region, left
lower region, right lower region and
axial region). Pain in the jaw, chest
and abdomen are not evaluated as
part of the generalized pain definition
associated symptoms
No
Symptoms present at a similar
level for at least 3 months
The patient does not have a
disorder that would otherwise
explain the pain
54
Symptoms present at a similar
level for at least 3 months
The patient does not have a
disorder that would otherwise
sufficiently explain the painc
The criteria also include a
fibromyalgia severity score (the
sum of WPI plus SSS), which is
a quantitative measurement of
fibromyalgia severity
56
The presence of generalized pain
Symptoms have been present at a
similar level for at least 3 months
A diagnosis of fibromyalgia is valid
irrespective of other diagnoses
and does not exclude the
presence of other illnesses
40
Moderate to severe sleep
problems or fatigue
Symptoms have been present for
at least 3 months
AAPT, ACTTION-American Pain Society Pain Taxonomy; MSP, multisite pain; SSS, Symptom Severity Score; WPI, Widespread Pain Index. aRegions assessed by the
WPI: left shoulder girdle, right shoulder girdle, left hip (buttock or trochanter), right hip (buttock or trochanter), left jaw, right jaw, upper back, lower back, left
upper arm, right upper arm, left upper leg, right upper leg, chest, neck, abdomen, left lower arm, right lower arm, left lower leg and right lower leg. bThis
modification enabled the use of these criteria in epidemiological and clinical studies without the requirement for an examiner (but should not be used for
self-diagnosis). cNote that the 2011 criteria are based on the possibility of self-administration of the questionnaires.
Screening tools
Some routine screening tools have been developed to
help general practitioners to identify those patients who
are most at risk of developing fibromyalgia. These tools
include the Fibromyalgia Rapid Screening Tool, which
consists of six general questions58, and the FibroDetect
test59, which covers pain and all fibromyalgia-influenced
domains, as well as the patient’s attitude and history.
The Simple Fibromyalgia Screening Questionnaire was
also validated as a useful screening tool in 2019 (ref.60).
General practitioners could take advantage of these tools
to detect patients with or even at risk of fibromyalgia, so
that they can refer the patient to a specialist.
Prevention, as well as very early fibromyalgia diagnosis, remains an elusive goal, mainly owing to the lack of
Nature Reviews | Rheumatology
established risk factors (see the section on hypothetical
pathogenic mechanisms). In addition, insufficient data
are available on the effect of early diagnosis on clinical
progression; nonetheless, early recognition could enable
the commencement of non-pharmacological approaches,
such as psychotherapy or physical reconditioning, at an
early stage and prevent the need for pharmacological
treatments, therefore limiting adverse effects.
Patient assessment
The assessment of fibromyalgia should be holistic and
not only consider all of the symptoms experienced by
patients but also alleviating or aggravating factors and
the effect of fibromyalgia on everyday life, functional
status and working ability. This approach is crucial for
Reviews
establishing treatment goals that are shared by patients
and their physicians.
The presence and severity of fibromyalgia symptoms
(such as sleep disturbances, fatigue, cognitive and somatic
symptoms) in the general population usually follow a
bell-shaped curve61. This variation means that the diagnosis of the syndrome is completely arbitrary and involves
the dichotomization of a continuum often referred to
as ‘fibromyalgianess’62, a scale that can essentially be
interpreted as the likelihood of having fibromyalgia63,64.
Using various scales to assess fibromyalgia in individual
patients is important not only to discover the degree of
‘fibromyalgianess’ or severity of the condition but also
to establish a baseline against which improvement can be
assessed during follow-up. These assessments need to
be reliable, easy to use, and validated in clinical practice,
but, most importantly, they should take into account the
multidimensional nature of chronic pain65.
Fibromyalgia and CWP can be assessed using vali­
dated single and composite tests66. The most widely
used tests include the Fibromyalgia Impact Question­
naire (FIQ)67 and its revised version (FIQR)68,69, the
Fibromyalgia Assessment Status (FAS)70,71, the Fibro­
myalgia Survey Criteria (FSC)72 and the Patient Health
Questionnaire 15 (PHQ15)73. Various studies have
hypothesized the level of fibromyalgia activity that can
be considered remission for each questionnaire (<39 for
the FIQ, <12 for the FSC and <5 for the PHQ15)67,70,73,
but the target of clinical improvement should be an
improvement in function from the patient’s point of view.
Nociplastic pain
A clinical definition of
pain arising from altered
nociception, despite no
evidence of tissue damage
causing the activation of
nociceptors or evidence
of disease or lesions of the
somatosensory system causing
the pain.
Central sensitization
A neurophysiological process
of pain amplification in the
central nervous system; this
process occurs physiologically
after injuries to elicit a
protective behaviour and
maximize the healing process.
Hyperalgesia
A condition in which a painful
stimulus is perceived as being
even more painful.
Allodynia
A condition in which a normal
stimulus is perceived as being
painful.
Temporal summation
The perception of repetitive
noxious stimulation as being
increasingly painful.
Hypothetical pathogenic mechanisms
Pain
An important symptom of fibromyalgia is chronic
widespread musculoskeletal pain. Generally speaking,
pain can be divided into three categories: nociceptive,
neuropathic and nociplastic pain74. Physiologically, pain
functions as an alarm system that warns the body of the
presence of a potentially harmful situation, known as
‘nociceptive pain’. In some situations, pain loses its function as an alarm signal, such as when pain persists after
the end of the original stimulus or when pain is started by
a stimulus that is completely innocuous. Such pain can
be caused by real damage to the nervous system, known
as ‘neuropathic pain’, or by mostly reversible modifications to the nervous system, known as ‘nociplastic pain’.
In the latter case, the changes increase the sensitivity
of the control system that usually decides which stimuli should be interpreted as painful and which should
not. This type of pain is in line with the description of
fibromyalgia as part of the nosological group of central
sensitivity syndromes75. Clinically, fibromyalgia has
many of the features of central sensitization (also known
as central hyperactivation)76,77: hyperalgesia, allodynia78,
temporal summation79,80 and hypersensitivity to various
external stimuli such as sounds or lights81–83.
Nociplastic pain in fibromyalgia
Over the past 20 years, researchers have identified neurobiological features that correlate with fibromyalgia nociplastic pain84. Emerging evidence suggest that diffuse
pain processing in the brain is altered in fibromyalgia,
as indicated by increased activation in areas of the brain
dedicated to pain (that is, patients with fibromyalgia
require less pressure than healthy individuals to show the
same level of brain activity)85–89, altered connectivity90 and
a reduction in brain activity associated with visual cues
that signal an imminent painful stimulus (pain anticipation) or its imminent end (anticipatory analgesia)91.
Various studies have shown that the functional activation
and connectivity of endogenous pain inhibitory signals
are altered in patients with fibromyalgia (meaning that
there is an imbalance between the various nociceptive
and anti-nociceptive systems)92–95. Furthermore, patients
with fibromyalgia have less grey matter in the cortical
and subcortical areas involved in processing nociceptive
stimuli, particularly at the level of the cingulate cortex, frontal orbit and insula96, than healthy individuals,
although whether these alterations causally precede the
experience of CWP and hypersensitivity is unclear.
This imbalance between the nociceptive and
anti-nociceptive systems also subsists at a microscopic
level. Increased levels of substance P (a neurotransmitter
that mediates pain facilitation, especially temporal summation) have been detected in the cerebrospinal fluid of
patients with fibromyalgia compared with that in healthy
individuals97. Patients with fibromyalgia also have a
lower μ-opioid receptor availability in regions of the
brain involved in pain modulation (including the nucleus
accumbens, the amygdala and the dorsal cingulate) and
higher levels of opioids in the cerebrospinal fluid than
healthy individuals98,99. In keeping with the hypoactivity of the descending analgesic pathways during fibromyalgia, the levels of noradrenergic and serotoninergic
neurotransmitters in the biological fluid of patients with
fibromyalgia are lower than in healthy individuals100,101,
and brain dopaminergic activity during painful stimulation is attenuated102,103. Finally, hypersensitivity might be
mirrored by an excess of excitatory neurotransmitters in
brain areas important for pain modulation104,105.
Identifying the cause of these nociplastic alterations is
difficult, but what is clear is that fibromyalgia is unlikely
to have a single aetiology. Genetic background seems to
have a fundamental role, as patients with fibromyalgia
often report a family history of chronic pain and studies have identified notable familial clustering of fibromyalgia or muscle tenderness106–108, as well as various
polymorphisms in genes of the nociceptive pathway
that are associated with fibromyalgia109,110. In addition
to this genetic substrate, a variety of other peripheral
and central mechanisms also have a role. The relative
contribution and relationship among these pathogenic
mechanisms is represented in Fig. 3.
Peripheral mechanisms
Painful stimuli coming from the periphery might initiate or reinforce the nociplastic process, and the fact
that some of these peripheral sources of pain could
originate from the joints might explain both the higher
prevalence of fibromyalgia among patients with rheumatic diseases111,112 and the beneficial effects of extensive
treatment of rheumatic conditions such as osteoarthritis
on fibromyalgia symptoms113. In addition to peripheral
sources of pain (such as joint inflammation), alterations
www.nature.com/nrrheum
Reviews
Central nervous system
• Activation of pain areas
• Altered brain connectivity
• ↓ Pain inhibitory signals and
paradoxical stimulation
• ↓ Noradrenaline, 5HT,
dopamine and opioid
receptors
• ↑ Substance P and excitatory
neurotransmitters (such as
glutamate)
• Low resilience
• Maladaptive stress coping
• Sleep alterations
• Depression and anxiety
• Autonomic alterations
• Genetic factors
Top
down
Bottom
up
Body periphery (sensory neurons, joints, viscera and immune cells)
• Neuroinflammation
• Small fibre neuropathy
• Peripheral nociceptive stimuli
or any chronic painful disease
• Genetic factors
Peripheral sensitization
(↓ nociceptive threshold)
Nociplastic alterations
Pathogenic mechanisms
Fig. 3 | hypothesized interplay between potential pathogenic mechanisms and
nociplastic alterations in fibromyalgia. The aetiology of fibromyalgia and the underlying
cause of fibromyalgia-related nociplastic alterations are not fully understood. Interplay
between various mechanisms, including genetic predisposition, stressful life events and
peripheral (inflammatory) and central (cognitive–emotional) mechanisms, are thought to
lead to neuromorphological modifications (‘nociplastic pain’) and pain dysperception. This
figure illustrates the potential interplay between various pathogenic mechanisms (shown
in grey boxes) and major nociplastic alterations (shown in red boxes). These pathogenic
mechanisms influence nociplastic alterations in a causal fashion, but the opposite is also
thought to be true (for example, heightened pain perception negatively influences sleep).
A reciprocal aetiopathogenic relationship might also occur between the central nervous
system and the periphery of the body, which occurs in a both bottom-up and top-down
fashion: the former is mainly inflammatory and algesic, whereas the latter is mainly
psychological and cognitive-emotional. 5HT, 5-hydroxytryptamine.
Dysaesthesia
An unpleasant abnormal
sensation (that can be
spontaneous or evoked)
that is usually associated with
irritation or injury to a sensory
nerve or nerve root.
Small fibre neuropathy
Damage to small myelinated
(type Aδ) nerve fibres or
unmyelinated C peripheral
nerve fibres; these small
somatic fibres have sensory
functions including thermal
perception and nociception.
Dysautonomia
An umbrella term used to
describe several different
medical conditions that cause
a malfunction of the autonomic
nervous system.
Catastrophizing
An exaggerated amplification
of emotional aspects that leads
individuals to consider pain
terrible and intolerable.
an association between fibromyalgia and traumas or
abuse121–123. Patients with fibromyalgia might indeed
have reduced levels of resilience and effective coping
strategies30. This low resilience is strikingly reflected by
the low heart rate variability (HRV) of these patients
(HRV is a powerful indicator of sympathetic versus para­
sympathetic activation of the autonomic nervous sys­
tem in response to environmental demands)124–126. An
interesting hypothesis is that the sympathetic autonomic
nervous system is hyper-active but also hypo-reactive
in fibromyalgia, blunting the response to stressors.
Altered activation of the autonomic nervous system
(dysautonomia) could be the cause of many fibromyalgia
symptoms, such as balance disturbances and episodes
of low blood pressure127. Low HRV is associated with
neuromorphological alterations128 that are also present
in patients with fibromyalgia, including the presence of
low-density grey matter in the cingulate cortex128,129. In
addition, a low level of resilience is associated with an
increased probability of developing post-traumatic stress
disorder, anxiety or mood disorders, which are very prevalent in the fibromyalgia population130. As the development of resilience-based strategies is an important factor
in treating such disorders131,132, implementing resilience
and coping strategies might be a promising means of
treating fibromyalgia and chronic pain in general133.
in the peripheral nervous system could also be involved.
Researchers have attempted to explain fibromyalgia
dysaesthesia in terms of small fibre dysfunction, and
a number of studies have identified the presence of
small fibre neuropathy (fibre loss and reduced axonal
diameter) in patients with fibromyalgia114, although
this finding might not be specific to fibromyalgia115.
Moreover, an emerging hypothesis is that immune system activation is capable of modulating the excitability of
nociceptive pathways116 as a result of what has been called
neuro-inflammation. This hypothesis was first postulated on the basis of the detection of pro-inflammatory
substances and organ-specific and non-specific autoantibodies in the serum of patients with fibromyalgia117,118.
Some researchers have long argued that infections might
trigger fibromyalgia, as fibromyalgia is more prevalent
among individuals infected with hepatitis C virus, HIV
or Borrelia burgdorferi, although the findings are not
convincing119,120.
Cognitive factors. Far from being a solely sensory experience, pain is a mental state that necessarily involves
educational, social and cognitive factors, in line with
the increasingly recognized biopsychosocial model of
medicine134. Maladaptive coping styles when facing
adverse situations (for example, a low level of selfefficacy, hypervigilance to pain stimuli, avoidance and
catastrophizing ) can dysfunctionally modulate pain
and affect the intensity of subjective pain and a patient’s
general health, as well as increasing activation in painrelated areas of the brain135–139. This mechanism can be
referred to as cognitive–emotional sensitization to pain.
An additional complication is that patients with
fibromyalgia more frequently have psychological alterations that might escalate to full-blown psychiatric
disorders140. Depression is highly prevalent in patients
with fibromyalgia, but is also a common denominator
of other chronic painful conditions141. Determining
whether these alterations come with the condition,
precede the condition, or are secondary to the condition, can be difficult. The relationship between pain and
depression seems to be bidirectional: chronic depression
can induce central sensitization and thus lower the nociceptive threshold, and chronic pain can be associated
with mood changes that can lead to a depressive state142.
Moreover, among the different symptoms of depression
(affective, cognitive and somatic symptoms), the somatic
symptoms often overlap with the physical symptoms of
many chronic dysfunctional pain syndromes (headache,
low back pain and visceral pain)143.
Central mechanisms
Coping with stress. Many patients with fibromyalgia
associate stressors with the onset and exacerbations of
their condition33, and multiple studies have reported
Sleep. The bilateral connection between fibromyalgia
and psychological alterations might also be true for sleep
alterations144. CWP disrupts sleep in a vicious circle that
involves the autonomic nervous system145, but the quality
Nature Reviews | Rheumatology
Reviews
of sleep is also causally related to pain severity146,147.
A 1975 study was the first to show that people with fibrositis (an old term for fibromyalgia) experience objective
sleep disturbances and that the same symptoms can be
induced in sleep-deprived healthy individuals148. Since
this initial study, clinical trials have shown that improving the quality of sleep by means of pharmacological or
non-pharmacological treatment can reduce pain and
fatigue in patients with fibromyalgia144. Furthermore,
some evidence suggests the existence of a bidirectional
relationship between sleep disturbances and anxiety
or depression149, and data from a large populationbased study in Norway suggest that poor sleep quality
predisposes adolescents to mental illnesses150.
ability to cope with stress. In this sense, fibromyalgia
can be seen as a condition that represents a mind–
body hyper-connection, rather than a mind–body
disconnection3. Consequently, fibromyalgia treatment
needs to be holistic and comprehensive. Indeed, the
therapeutic approach to managing patients with fibromyalgia is characterized by integrated and multidisciplinary interventions152. In this section, we describe
the various interventions available for the management of fibromyalgia. We propose that the treatment
of fibromyalgia can be divided into four pillars: patient
education, fitness, pharmacological treatment and
psychotherapy (Fig. 4). Our suggested treatment strategy, shown in Fig. 4, takes into account not only the
latest EULAR recommendations for fibromyalgia
management4 but also real-life clinical experience and
realistic patient expectations and goals. Indeed, we suggest starting pharmacological treatment straightaway,
mainly because patients are usually diagnosed years
after symptom onset153.
Treatment
Many factors contribute to the development of fibromyalgia in a unique manner61: genetic predisposition,
personal experiences, emotional–cognitive factors,
mind–body relationship 151 and a biopsychological
Diagnosis of fibromyalgia
Patient
education
CBT, hypnosis
and/or relaxation
techniques
(according to
the patient’s needs)
Antidepressant
(duloxetine or
milnacipram) or
anticonvulsant
and
analgesic
(paracetamol)
Physical activity,
weight loss and
a nutritional
programme
Lack of efficacy (patient reassessment)
CBT, hypnosis
and/or relaxation
techniques
(according to
the patient’s needs)
New modalities
(such as
hyperbaric
oxygen therapy or
neurostimulation)
A different
antidepressant or
anticonvulsant
and/or
analgesic and/or
muscle relaxant
Physical activity,
weight loss and
a nutritional
programme
Any type of
complementary
intervention that
is useful for the
patient (such as
acupuncture
or TENS)
Lack of efficacy (patient reassessment)
CBT, hypnosis
and/or relaxation
techniques
(according to
the patient’s needs)
Patient education
Patient education and fitness
Cannabinoids
or weak opioids
(tramadol)
Psychotherapy
Additional options
Physical activity,
weight loss and
a nutritional
programme
Pharmacotherapy
Fig. 4 | Proposed treatment strategy for fibromyalgia. Our proposed therapeutic flowchart for managing patients
with fibromyalgia is shown, which was built on the basis of scientific literature (discussed in the main text), the latest
EULAR recommendations4 and real-life clinical experience. The main difference between this flowchart and the EULAR
recommendations is that herein we start with both pharmacological and non-pharmacological treatments simultaneously
(the EULAR recommendations has a sequential workflow, rather than a parallel workflow, that begins with nonpharmacological treatment) and follow the ‘four pillar concept’ described in the text (patient education (grey),
psychotherapy (dark green), pharmacotherapy (light green) and fitness (blue)). The main idea is that all treatment pillars
should be applied from the beginning of fibromyalgia management, and that if there is a lack of efficacy of one approach
(mainly the pharmacological therapy), the treatment approach should be modified according to the patient’s needs.
This scheme should not be rigidly applied in clinical practice, but rather should always be individualized according to
patients’ needs and preferences. CBT, cognitive-behavioural therapy; TENS, transcutaneous electric nervous stimulation.
www.nature.com/nrrheum
Reviews
Patient education
An important step in managing patients with fibromyalgia
is ensuring that the patients understand their illness before
they are prescribed any medications154–157. It is crucial to
reassure patients that fibromyalgia is a real pathological
condition and to legitimize their suffering, making it clear
that, although disabling, the condition is not progressive and is not due to peripheral tissue damage. Patients
should also be told that they will have a predominant role
in fibromyalgia management, and should develop their
own particular techniques and approaches to maximize
their quality of life. This approach is paradigmatic of
the ‘self-management’ approach that should be used in the
case of any chronic condition. Moreover, as stress, mood
and sleep disturbances have an important role in fibromyalgia, patients should be encouraged to learn good
sleep hygiene and relaxation techniques, and take part in
formal stress reduction programmes, including psychiatric consultations if necessary. Importantly, patients can
be encouraged to continue non-pharmacological measures on the basis of their individual needs as long as the
interventions do not cause any harm. Pharmacological
treatment might be helpful in relieving some symptoms,
but patients rarely improve substantially without adopting
these core self-management strategies154,158.
Fitness
The latest EULAR recommendations on fibromyalgia management stress the importance of first using
non-pharmacological measures in fibromyalgia management, but the only ‘strong’ recommendation is in favour
of exercise4. As in the case of other chronic conditions,
fitness is pivotal and should involve weight loss, aerobic
and strengthening exercises as well as dietary modifications, all of which are important disease-modifying
factors4. Weight loss improves posture and well-being,
and decreases both obesity-induced inflammation and
peripheral nociceptive inputs159. Aerobic exercise is
strongly recommended as it can improve pain and physical function in patients with fibromyalgia160, although
the commencement of training can be difficult for some
patients because of deconditioning and psychological
factors161. The recommended optimal cardiovascular
fitness training consists of a minimum of 20 min of aero­
bic exercise three times a week4. Regarding nutrition,
although patients with fibromyalgia might have a higher
rate of nutritional deficiencies or incorrect dietary regimens than the general population162, no precise diet or
vitamin integration is recommended, as no clear data are
available on the correct nutritional protocol163.
Pharmacological treatment
Pharmacotherapy should be aimed at analgesia in
a mechanism-oriented fashion164. In line with this
approach, centrally acting medications can be effective in fibromyalgia, particularly antidepressants
and anticonvulsants155, which increase the presence
of pain-inhibitory neurotransmitters by facilitating
descending pathways and decreasing dorsal horn sensitization, or decreasing systemic hyperexcitability155.
Herein, we discuss commonly prescribed drugs
for fibromyalgia that have the most consistent and
Nature Reviews | Rheumatology
largest amount of evidence. These drugs, along with
their associated adverse effects, are listed in Table 2.
Antidepressants. Systematic literature reviews and
meta-analyses suggest that the antidepressant amitriptyline is quite effective at treating fibromyalgia, especially for reducing pain and fatigue165,166, although most
of the studies reviewed were old and had methodological limitations167. The mean number of patients needed
to treat to achieve a 30% pain reduction was four4,165.
Interestingly, amitriptyline was found to also have a
moderate effect on sleep and a slight effect on fatigue165.
Both duloxetine and milnacipran have proved to be
more effective than placebo in treating fibromyalgia
pain and are FDA-approved for fibromyalgia, although
the incremental benefit is small and these drugs do not
have any effect on other fibromyalgia symptoms168. Data
from one systematic review169 indicate that, in the case
of duloxetine, the number needed to treat is eight169
and, importantly, that this drug improves pain severity
regardless of the presence of a comorbid major depressive disorder. However, adverse effects can lead to dropouts, which have ranged from 9% to 23% in short-term
studies, and from 11.4% to 27.2% in long-term studies167,
but these adverse effects can be limited by using a slow
dose-titration approach. It should also be noted that the
results of a randomized controlled trial of milnacipran170
were unfavourable in terms of pain modulation, global
pain, mechanical and thermal thresholds, allodynia,
cognition and tolerance.
Anticonvulsants. Anticonvulsants have been extensively investigated for fibromyalgia treatment171. Among
the gabapentinoids, the benefits of gabapentin are
uncertain172, whereas the results of various meta-analyses
suggest that pregabalin is effective and safe for some
patients with fibromyalgia173–175. Pregabalin is currently
the only anticonvulsant that has been approved by the
FDA for fibromyalgia, although adverse effects are
frequent, particularly dizziness171.
Muscle relaxants. Cyclobenzaprine is structurally related
to tricyclic antidepressants but is approved as a muscle
relaxant; this drug improves pain and the quality of life
(especially sleep) of patients with fibromyalgia, but not
fatigue176. Tizanidine is an α2 receptor agonist that has
anxiolytic, analgesic and sedative properties177, has been
used for the treatment of myofascial pain disorders178
and can be of help in fibromyalgia179.
Analgesic drugs. The role of opioids in the treatment
of fibromyalgia is limited180. Patients with fibromyalgia have altered endogenous opioid activity, with little
opioid receptor availability but high concentrations
of opioid peptides in biological fluids98,99, which might
explain why opioids are generally not very effective and
naltrexone (an opioid receptor antagonist that also has
antagonist effects on non-opioid receptors and can have
neuroprotective and analgesic effects) was hypothesized
to be of some benefit181,182. Therefore, opioids are generally avoided, not least because of their unfavourable
risk-to-benefit profile183. The only opioid that has proved
Reviews
Table 2 | Commonly prescribed drugs for fibromyalgia treatment and their adverse effects
Drug
Class of drug
FDa-approved adverse effects169,174,184,187,203,239–242
drugs for
fibromyalgia
Antidepressants
Duloxetine
SNRI
Milnacipran
SNRI
Yes243
Yes246
Amitriptyline
No
Tricyclic antidepressant
Anticonvulsants
Pregabalin
GABAergic drug
Gabapentin
GABAergic drug
Muscle relaxants
Cyclobenzaprine Serotoninergic muscle
relaxant
Tizanidine
α2 receptor agonist
Analgesic drugs
Tramadol
Paracetamol
Yes247
No
Sedation, dizziness, vertigo, asthenia, nausea and
weight gain
No
Nausea, palpitations, headache, fatigue,
xerostomia, constipation and serotonin syndromea
Dizziness, asthenia, xerostomia, vomiting,
constipation, liver test abnormalities, bradycardia,
hypotension and blurred vision
No
Weak opioid and SNRI
No
Analgesic and antipyretic
drug
No
Hypnotic drugs
Zolpidem
GABAergic and
non-benzodiazepine
hypnotic drug
Antipsychotic drugs
Quetiapine
Atypical antipsychotic drug
Cannabis or cannabinoids
Nabilone
Pure cannabinoid
(tetrahydrocannabinol)
Cannabis
Phytopharmaceutical
(different concentrations of
tetrahydrocannabinol and
cannabidiol)
Nausea, palpitations, headache, fatigue,
tachycardia, insomnia, xerostomia, constipation
and serotonin syndromea(Refs244,245)
Xerostomia, constipation, weight gain, urinary
retention, sedation and serotonin syndromea
Constipation, nausea, vomiting, dizziness, fatigue,
headache, itching and xerostomia
Nausea, vomit, constipation and liver disease
No
Dizziness, headache, somnolence, confusion,
agitation, abdominal pain, constipation and
xerostomia
No
Somnolence, headache, dizziness, extrapyramidal
symptoms, weight gain, dyslipidaemia,
hyperglycaemia, xerostomia, vomiting and nausea,
and constipation
No
Drowsiness, dizziness, nausea, xerostomia,
confusion, anxiety and tachycardia
Drowsiness, dizziness, nausea, xerostomia, blurred
vision, increased/decreased appetite, vertigo,
tachycardia and hypotension
No
All these drugs target neurotransmitters, and their classification is based on the disease for which they were initially approved (for
example, antidepressants for depression). The adverse effects of anticonvulsants are dose dependent, whereas the adverse effects
of antidepressants depend on the metabolism of the individual. Treatment with a combination of antidepressants should be
avoided owing to the risk of serotonin syndrome. SNRI, serotonin–norepinephrine reuptake inhibitor. aA potentially
life-threatening syndrome characterized by the combination of mental status alteration (such as agitation, anxiety, disorientation
and excitement), neuromuscular hyperactivity (such as tremors, hyperreflexia, muscle rigidity and clonus) and autonomic
hyperactivity (such as vomiting, diarrhoea, hypertension and tachycardia, and mydriasis).
to be effective in patients with fibromyalgia is tramadol, alone or combined with paracetamol4. Tramadol
functions as a weak agonist of µ-opioid receptors and
as a serotonin–norepinephrine reuptake inhibitor
(SNRI). Substantial evidence suggests that traditional
analgesic drugs such as paracetamol and non-steroidal
anti-inflammatory drugs are not effective in treating
fibromyalgia184, but these drugs are fundamental for
treating concomitant peripheral forms of pain such as
osteoarthritic pain because peripheral nociceptive inputs
can promote central sensitization185.
Hypnotic and antipsychotic drugs. Benzodiazepines and
other hypnotic drugs, such as zolpidem, can be used
in the short term to improve sleep, but tend not to be
efficacious for fibromyalgia pain4,186.
Quetiapine has been so far the most frequently studied antipsychotic drug for fibromyalgia. A Cochrane
review187 suggested that this drug shows some benefit
in treating fibromyalgia-related pain, sleep problems,
depression and anxiety, but, owing to the low quality of
evidence of the trials, this drug should only be taken
for a short period of time for fibromyalgia treatment.
Interestingly, one trial comparing amitriptyline with
quetiapine showed no difference between the two drugs
in terms of their ability to reduce various symptoms in
patients with fibromyalgia, including pain, fatigue, sleep
problems, anxiety and depression188.
Cannabis and cannabinoids. The cannabis plant is very
different from pure, synthetic cannabinoids, insofar
as it contains about 100 different active cannabinoids,
www.nature.com/nrrheum
Reviews
among which tetrahydrocannabinol (THC) and cannabidiol (CBD) are the most relevant and most frequently studied. Most trials have investigated nabilone,
a semi-synthetic THC analogue that is ten times more
potent than THC itself189. One Cochrane review190 does
not recommend the use of nabilone to treat fibromyalgia. The proper use of pure cannabinoids, if any, is still to
be defined. By contrast, cannabis has proved to be moderately effective in the treatment of a number of chronic
non-cancer pain conditions191–193, and for this reason it
was hypothesized that it could be beneficial for fibromyalgia as well194–196. The effectiveness of preparations
with different THC to cannabidiol ratios is still under
investigation197–199, and well-conducted randomized
clinical trials are still needed; however, a US National
Pain Report survey of the efficacy of three drugs duly
approved for fibromyalgia in comparison with that of
cannabis had 1,300 respondents and proved favourable
towards cannabis200.
Drug combinations and sequences. In brief, there is no
gold standard pharmacological treatment for fibromyalgia. Maximum doses of a single drug are rarely used
because of safety concerns201. Moreover, single drugs
tend to have a clinically relevant effect in fewer than
half of the treated patients202. Therefore, a combination
of drugs is usually preferred using a patient-centred,
symptom-based stepwise approach202 that has inevitably prevented any specific recommendation concerning which type of combined treatment should be
used203. However, empirical evidence, and our own
experience, suggest that treatment should be started
with an SNRI antidepressant, followed by one of the
anticonvulsants for patients who respond inadequately
or cannot tolerate antidepressants. An SNRI or an anticonvulsant could be beneficial for patients with severe
fatigue, depression, or a severe sleep disturbance. The
efficacy and safety of combinations of anticonvulsants
and antidepressants have been investigated in various
studies204–206.
Psychotherapy
Cognitive-behavioural therapy is the most widely
studied and practised psychotherapy for fibromyalgia.
This approach is aimed at helping patients to identify
condition-related maladaptive thoughts to develop
effective coping strategies and behaviour. Developing
effective coping strategies is particularly important in
a condition such as fibromyalgia because dysfunctional
pain modulation is a fundamental factor in exacerbating
and protracting pain (as discussed earlier). In a systematic review207, the investigators concluded that patients
who received cognitive-behavioural therapy (including
acceptance-based cognitive-behavioural therapy) might
have shown greater improvements in pain, physical
functioning and mood than patients receiving usual care,
on the waiting list, or being treated with other active,
non-pharmacological methods. Cognitive-behavioural
therapy might be particularly useful in patients with
fibromyalgia as this intervention teaches effective coping strategies that can be used on a long-term basis,
which is very useful in the case of a chronic condition.
Nature Reviews | Rheumatology
Investigations along these lines have begun in the past
year, and one non-controlled study found promising
results 1 year after the start of treatment208. Ensuring
greater access to treatment (perhaps by means of the
Internet) will probably be the next step209.
Other non-pharmacological treatments
Non-pharmacological treatments include a wide range
of interventions that are usually referred to as ‘complementary’ or ‘alternative’ therapies. A 2014 meta-analysis
suggested that the magnitude of the multidimensional
effect of these approaches can exceed that of pharmacological treatments for fibromyalgia210. However, the
benefit of these inventions is still an area of controversy, as the study designs are often weak and the quality of the evidence is usually low. Nevertheless, various
non-pharmacological therapies are included in the
EULAR recommendations for the management of fibromyalgia, and non-pharmacological treatments might at
least be considered as adjunctive, if not the core, treatment for many patients4. Non-pharmacological or alternative measures can be introduced depending on the
limitations of cost, availability and patient preference.
Some of the most frequently used non-pharmacological
treatments are briefly described below.
Spa therapy. Spa therapy consists of multiple methods
that are based on the curative effects of thermal water
and include balneotherapy, mud packs and hydrotherapy. These approaches have been used empirically since
ancient times to treat a wide range of conditions211.
If available and affordable to the patient, hot thermal
baths are an option for some patients and are particularly popular in many European countries. Hot thermal
baths are thought to improve various fibromyalgia symptoms (for example, they have been shown to moderately
decrease pain, improve the patients’ health-related quality of life212–214 and also have a small effect on mood)212.
Balneotherapy might even be considered as a first-line
treatment together with patient education and aerobic
exercise215: the mechanism of action of this approach
is still a matter of discussion but probably involves
an interplay of many hormonal, inflammatory and
cognitive-emotional factors216.
Tai chi, qigong and yoga. Tai chi, qigong and yoga are
forms of alternative exercises or ‘meditative movement therapies’ that have been increasingly adopted by
patients with fibromyalgia. These exercises are based on
physical movement integrated with mental relaxation
and breathing techniques, and two meta-analyses217,218
have indicated that these approaches can be efficacious
and safe in treating fibromyalgia. The results suggested
that the fibromyalgia symptoms of sleep, fatigue, depression, pain and the quality of life all improved, although
the poor quality of the studies did not enable any definite conclusions to be drawn. The effect of tai chi has
also been investigated in another meta-analysis219, which
found that this approach had a notable positive effect on
many aspects of the patients’ lives, and that tai chi could
become a promising alternative to conventional exercise
by possibly attracting less compliant patients220.
Reviews
Mindfulness. Mindfulness is based on the principle of
the non-judgemental acceptance of one’s condition,
thoughts and suffering. This approach is different from
cognitive-behavioural therapy insofar as it does not
address any particular maladaptive behaviour or thought,
but rather endorses a general view of coping with difficulties. The difference between these two techniques
was investigated in a randomized controlled trial, the
results of which suggested that mindfulness techniques
were more effective than cognitive-behavioural therapy
in improving various symptoms of fibromyalgia221.
Recognizing that nothing is intrinsically positive or
negative might be particularly helpful for patients with
fibromyalgia whose condition has a preponderant catastrophizing or negative emotional component, as mindfulness and acceptance-based interventions seem to have a
small to moderate effect on many aspects of the syndrome,
including pain, depression, anxiety, sleep and the quality
of life222,223. However, although the results achieved so far
are promising, the effects are still uncertain because of the
poor quality of evidence provided by the individual studies. An interesting new approach called acceptance and
commitment therapy can be seen as an intermediate form
between cognitive-behavioural therapy and mindfulness.
Two studies224,225 found that acceptance and commitment
therapy can lead to a greater improvement in functional
status in patients with fibromyalgia compared with usual
care and pharmacological treatment, thus confirming the
importance of the mind–body connection.
Hypnosis. In the past 3 years, hypnosis has captured the
interest of the scientific community as an increasing
number of studies have shown its efficacy in targeting
chronic pain226. One systematic review from 2017 highlighted the potential of hypnosis as a possible treatment
for patients with fibromyalgia as this approach not only
improved pain and sleeping problems at the end of the
sessions (across a study duration ranging between 8, 12,
14 and 26 weeks), but also after 3 months of follow-up227.
However, more methodologically valid studies are
needed, as the quality of evidence is still poor.
Acupuncture. Acupuncture is frequently sought by
patients with fibromyalgia and is (albeit weakly) recommended by EULAR because of the moderate quality of the
evidence in the literature4. Two meta-analyses228,229 have
underlined its efficacy in improving stiffness and pain,
although whether and how acupuncture differs in terms
of efficacy from sham (random) acupuncture is unclear.
Other modalities. Physical-agent modalities use different forms of energy such as thermal energy (for example
thermotherapy and cryotherapy) and electric energy (for
example electrotherapy) that are passively administered
to patients. In 2018, a meta-analysis214 provided evidence that transcutaneous electrical nerve stimulation,
electromagnetic therapy and, most importantly, thermal
therapy have positive effects on pain and the quality of
life (as measured using the FIQ) of patients with fibromyalgia, although the overall quality of the evidence
was poor. Researchers have speculated that these effects
are caused by changes in local inflammatory reactions,
pain thresholds and perceptions214. Various studies
have also investigated hyperbaric oxygen therapy230,231
and neurostimulation232. Hyperbaric oxygen therapy
facilitates oxygen delivery to the peripheral tissues by
increasing the partial pressure of oxygen in the arterial
system, decreasing the expression of pro-inflammatory
mediators233. Neurostimulation, operated by means of
electrical or magnetic currents (transcranial electrical stimulation and transcranial magnetic stimulation,
respectively) at the level of the primary motor cortex,
seems to be a promising treatment232. A 2019 structured
review234 suggested that direct current stimulation is
effective in modulating fibromyalgia pain, although the
data suggested that it has less effect on cognitive and
affective symptoms, and the length of time that the
effects lasted was unclear. Notably though, one study
suggested that the reduction in pain and other symptoms of fibromyalgia (fatigue and quality of life-related
aspects) induced by monthly transcranial magnetic
stimulation could be maintained for at least 6 months235.
Conclusions
The exponential number of clinical and other research
studies on fibromyalgia in the rheumatic field and other
biomedical fields reflects the interest recently aroused by
fibromyalgia, even though the real nature of this condition has not yet been completely clarified. After a century
of unsuccessful research aimed at identifying alterations
in the structures in which pain is perceived (the skin,
muscles and tendons), the most interesting findings
from the past 2–3 years concern the mechanisms underlying pain perception and the body’s response to stressful situations. Techniques such as functional MRI of the
brain have shown that chronic pain is related to changes
in the sensitivity to and processing of stimuli throughout the nociceptive system89, and are beginning to reveal
the neurophysiological signature of fibromyalgia236.
The finding of induced peripheral receptor sensitization in situations of psychological stress supports the
hypothesis that chronic pain is a result of interactions
between neurophysiological factors (neuroplasticity)
and socio-environmental stressors, and reflects the
increasingly recognized importance of the biopsychosocial model of medicine in relation to fibromyalgia and
chronic pain conditions in general237. The multiple components of the pathogenesis and maintenance of the syndrome mean that multi-modal treatment is necessary,
and non-pharmacological approaches can have a pivotal
role4. Within this framework, the concept of an individually tailored treatment is preponderant in fibromyalgia.
Therefore, it is difficult to interpret the results of randomized controlled trials that enrol a random sample
of the fibromyalgia population and measure average
treatment effectiveness, and such results might actually
be misleading given the potential existence of various
subgroups of patients with fibromyalgia with different
clinical characteristics. Being patient-centred, the therapeutic approach will inevitably be empirical238, although
evidence-based, and should adopt therapeutic goals that
are shared between the patient and the physician.
Published online xx xx xxxx
www.nature.com/nrrheum
Reviews
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
21.
22.
23.
24.
25.
26.
Queiroz, L. P. Worldwide epidemiology of fibromyalgia.
Curr. Pain. Headache Rep. 17, 356 (2013).
Jones, G. T. et al. The prevalence of fibromyalgia in
the general population: a comparison of the American
College of Rheumatology 1990, 2010, and modified
2010 classification criteria. Arthritis Rheumatol. 67,
568–575 (2015).
Perrot, S. Fibromyalgia: a misconnection in a
multiconnected world? Eur. J. Pain. 23, 866–873
(2019).
Macfarlane, G. J. et al. EULAR revised recommendations
for the management of fibromyalgia. Ann. Rheum. Dis.
76, 318–328 (2017).
Branco, J. C. et al. Prevalence of fibromyalgia: a survey
in five European countries. Semin. Arthritis Rheum.
39, 448–453 (2009).
Chaaya, M. et al. High burden of rheumatic diseases
in Lebanon: a COPCORD study. Int. J. Rheum. Dis. 15,
136–143 (2011).
Nakamura, I. et al. An epidemiologic internet survey
of fibromyalgia and chronic pain in Japan. Arthritis
Care Res. 66, 1093–1101 (2014).
Turhanoglu, A. D. et al. The epidemiological aspects
of fibromyalgia syndrome in adults living in Turkey:
a population based study. J. Musculoskelet. Pain 16,
141–147 (2008).
Guermazi, M. et al. [Fibromyalgia prevalence in
Tunisia]. Tunis. Med. 86, 806–811 (2008).
Rodrigues Senna, E. et al. Prevalence of rheumatic
diseases in Brazil: a study using the COPCORD
approach. J. Rheumatol. 31, 594–597 (2004).
White, K. P., Speechley, M., Harth, M. & Ostbye, T.
The London fibromyalgia epidemiology study: the
prevalence of fibromyalgia syndrome in London,
Ontario. J. Rheumatol. 26, 1570–1576 (1999).
Vincent, A. et al. Prevalence of fibromyalgia:
a population-based study in Olmsted County,
Minnesota, utilizing the Rochester epidemiology
project. Arthritis Care Res. 65, 786–792 (2013).
Scudds, R. A., Li, E. K. M. & Scudds, R. J. The
prevalence of fibromyalgia syndrome in Chinese
people in Hong Kong. J. Musculoskelet. Pain 14,
3–11 (2006).
Ablin, J. N. et al. Prevalence of fibromyalgia in the
Israeli population: a population-based study to estimate
the prevalence of fibromyalgia in the Israeli population
using the London Fibromyalgia Epidemiology Study
Screening Questionnaire (LFESSQ). Clin Exp Rheumatol
30, 39–43 (2012).
Prescott, E. et al. Fibromyalgia in the adult danish
population. I. A prevalence study. Scand. J. Rheumatol.
22, 233–237 (1993).
Salaffi, F. et al. Prevalence of musculoskeletal
conditions in an Italian population sample: Results
of a regional community-based study. I. The MAPPING
study. Clin. Exp. Rheumatol. 23, 819–828 (2005).
Spaeth, M. Epidemiology, costs, and the economic
burden of fibromyalgia. Arthritis Res. Ther. 11, 2–3
(2009).
Häuser, W., Sarzi-Puttini, P. & Fitzcharles, M. A.
Fibromyalgia syndrome: under-, over- and misdiagnosis.
Clin. Exp. Rheumatol. 37, 90–97 (2019).
Lachaine, J., Beauchemin, C. & Landry, P.-A.
Clinical and economic characteristics of patients with
fibromyalgia syndrome. Clin. J. Pain 26, 284–290
(2010).
Berger, A., Dukes, E., Martin, S., Edelsberg, J. &
Oster, G. Characteristics and healthcare costs of
patients with fibromyalgia syndrome. Int. J. Clin. Pract.
61, 1498–1508 (2007).
Knight, T. et al. Health-resource use and costs
associated with fibromyalgia in France, Germany,
and the United States. Clinicoecon. Outcomes Res. 5,
171–180 (2013).
Lacasse, A., Bourgault, P. & Choinière, M. Fibromyalgiarelated costs and loss of productivity: a substantial
societal burden. BMC Musculoskelet. Disord. 17, 168
(2016).
Guymer, E. K., Littlejohn, G. O., Brand, C. K. &
Kwiatek, R. A. Fibromyalgia onset has a high impact
on work ability in Australians. Intern. Med. J. 46,
1069–1074 (2016).
Gracely, R. H., Grant, M. A. & Giesecke, T. Evoked
pain measures in fibromyalgia. Best Pract. Res.
Clin. Rheumatol. 17, 593–609 (2003).
Koroschetz, J. et al. Fibromyalgia and neuropathic
pain — differences and similarities. A comparison of
3057 patients with diabetic painful neuropathy and
fibromyalgia. BMC Neurol. 11, 55 (2011).
Rehm, S. E. et al. A cross-sectional survey of
3035 patients with fibromyalgia: subgroups
of patients with typical comorbidities and sensory
Nature Reviews | Rheumatology
27.
28.
29.
30.
31.
32.
33.
34.
35.
36.
37.
38.
39.
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
51.
symptom profiles. Rheumatology 49, 1146–1152
(2010).
Rossi, A. et al. Fibromyalgia and nutrition: what news?
Clin. Exp. Rheumatol. 33, S117–S125 (2015).
Bossema, E. R., Van Middendorp, H., Jacobs, J. W. G.,
Bijlsma, J. W. J. & Geenen, R. Influence of weather on
daily symptoms of pain and fatigue in female patients
with fibromyalgia: a multilevel regression analysis.
Arthritis Care Res. 65, 1019–1025 (2013).
Staud, R., Robinson, M. E., Weyl, E. E. & Price, D. D.
Pain variability in fibromyalgia is related to activity
and rest: role of peripheral tissue impulse input.
J. Pain 11, 1376–1383 (2010).
Casale, R. et al. Fibromyalgia and the concept
of resilience. Clin. Exp. Rheumatol. 37, 105–113
(2019).
Sandıkçı, S. C. & Özbalkan, Z. Fatigue in rheumatic
diseases. Eur. J. Rheumatol. 2, 109–113 (2015).
Kleinman, L. et al. Assessment of sleep in patients
with fibromyalgia: qualitative development of the
fibromyalgia sleep diary. Health Qual. Life Outcomes
12, 111 (2014).
Bennett, R. M., Jones, J., Turk, D. C., Russell, I. J.
& Matallana, L. An internet survey of 2,596 people
with fibromyalgia. BMC Musculoskelet. Disord. 8, 27
(2007).
Glass, J. M. Review of cognitive dysfunction in
fibromyalgia: a convergence on working memory
and attentional control impairments. Rheum. Dis.
Clin. North Am. 35, 299–311 (2009).
Walitt, B. et al. The longitudinal outcome of
fibromyalgia: a study of 1555 patients. J. Rheumatol.
38, 2238–2246 (2011).
Ifergane, G., Buskila, D., Simiseshvely, N., Zeev, K.
& Cohen, H. Prevalence of fibromyalgia syndrome in
migraine patients. Cephalalgia 26, 451–456 (2006).
Mathieu, N. [Somatic comorbidities in irritable bowel
syndrome: fibromyalgia, chronic fatigue syndrome,
and interstitial cystitis]. Gastroenterol. Clin. Biol. 33,
S17–S25 (2009).
Nickel, J. C. et al. Interstitial cystitis/painful bladder
syndrome and associated medical conditions with an
emphasis on irritable bowel syndrome, fibromyalgia
and chronic fatigue syndrome. J. Urol. 184,
1358–1363 (2010).
Kalichman, L. Association between fibromyalgia and
sexual dysfunction in women. Clin. Rheumatol. 28,
365–369 (2009).
Arnold, L. M. et al. AAPT diagnostic criteria for
fibromyalgia. J. Pain 20, 611–628 (2018).
Solano, C. et al. Autonomic dysfunction in fibromyalgia
assessed by the composite autonomic symptoms
scale (COMPASS). J. Clin. Rheumatol. 15, 172–176
(2009).
Vincent, A. et al. A report of the autonomic symptom
profile in patients with fibromyalgia. J. Clin. Rheumatol.
20, 106–108 (2014).
Wolfe, F. et al. The American College of Rheumatology
1990 Criteria for the Classification of Fibromyalgia.
Report of the Multicenter Criteria Committee.
Arthritis Rheum. 33, 160–172 (1990).
Yunus, M. B. & Aldag, J. C. Restless legs syndrome
and leg cramps in fibromyalgia syndrome: a controlled
study. BMJ 312, 1339 (1996).
Viola-Saltzman, M., Watson, N. F., Bogart, A.,
Goldberg, J. & Buchwald, D. High prevalence
of restless legs syndrome among patients with
fibromyalgia: a controlled cross-sectional study.
J. Clin. Sleep Med. 6, 423–427 (2010).
Stehlik, R., Arvidsson, L. & Ulfberg, J. Restless legs
syndrome is common among female patients with
fibromyalgia. Eur. Neurol. 61, 107–111 (2009).
Jones, K. D., Horak, F. B., Winters-Stone, K.,
Irvine, J. M. & Bennett, R. M. Fibromyalgia is associated
with impaired balance and falls. J. Clin. Rheumatol. 15,
16–21 (2009).
Galvez-Sánchez, C. M., Duschek, S. & Del Paso, G. A. R.
Psychological impact of fibromyalgia: current
perspectives. Psychol. Res. Behav. Manag. 12,
117–127 (2019).
González, E., Elorza, J. & Failde, I. Fibromyalgia and
psychiatric comorbidity: their effect on the quality
of life patients. Actas Esp. Psiquiatr. 38, 295–300
(2010).
Kessler, R. C. et al. The epidemiology of major
depressive disorder: results from the National
Comorbidity Survey Replication (NCS-R). JAMA 289,
3095–3105 (2003).
Dreyer, L., Kendall, S., Danneskiold-Samsøe, B.,
Bartels, E. M. & Bliddal, H. Mortality in a cohort of
Danish patients with fibromyalgia: Increased frequency
of suicide. Arthritis Rheum. 62, 3101–3108 (2010).
52. Gill, H. et al. The prevalence of suicidal behaviour in
fibromyalgia patients. Prog. Neuropsychopharmacol.
Biol. Psychiatry https://doi.org/10.1016/j.pnpbp.
2020.110078 (2020).
53. Bennett, R. M. Fibrositis: misnomer for a common
rheumatic disorder. West. J. Med. 134, 405–413
(1981).
54. Wolfe, F. et al. Fibromyalgia criteria and severity scales
for clinical and epidemiological studies: a modification
of the ACR preliminary diagnostic criteria for
fibromyalgia. J. Rheumatol. 38, 1113–1122 (2011).
55. Wolfe, F. et al. The American College of Rheumatology
preliminary diagnostic criteria for fibromyalgia and
measurement of symptom severity. Arthritis Care Res.
62, 600–610 (2010).
56. Wolfe, F. et al. 2016 Revisions to the 2010/2011
fibromyalgia diagnostic criteria. Semin. Arthritis
Rheum. 46, 319–329 (2016).
57. Atzeni, F. et al. One year in review 2019: fibromyalgia.
Clin. Exp. Rheumatol. 37, S3–S10 (2019).
58. Perrot, S., Bouhassira, D. & Fermanian, J. Development
and validation of the fibromyalgia rapid screening tool
(FiRST). Pain 150, 250–256 (2010).
59. Baron, R. et al. Improving the primary care physicians’
decision making for fibromyalgia in clinical practice:
development and validation of the fibromyalgia
detection (FibroDetect®) screening tool. Health Qual.
Life Outcomes 12, 128 (2014).
60. Salaffi, F. et al. Development and validation of the
SImple FIbromyalgia Screening questionnaire for
improving the recognition of fibromyalgia in daily
practice. Clin. Exp. Rheumatol. 38, 9–16 (2019).
61. Yunus, M. B. Central sensitivity syndromes: a new
paradigm and group nosology for fibromyalgia and
overlapping conditions, and the related issue of
disease versus illness. Semin. Arthritis Rheum. 37,
339–352 (2008).
62. Wolfe, F. Fibromyalgianess. Arthritis Care Res. 61,
715–716 (2009).
63. Salaffi, F., Di Carlo, M., Arcà, S. & Galeazzi, M.
Categorisation of disease severity states in fibromyalgia:
a first step to support decision-making in health care
policy. Clin. Exp. Rheumatol. 36, 1074–1081 (2018).
64. Davis, F. et al. Characterizing classes of fibromyalgia
within the continuum of central sensitization
syndrome. J. Pain Res. 11, 2551–2560 (2018).
65. Ballantyne, J. C. & Sullivan, M. D. Intensity of chronic
pain — the wrong metric? N. Engl. J. Med. 373,
2098–2099 (2015).
66. Salaffi, F., Sarzi-Puttini, P., Ciapetti, A. & Atzeni, F.
Clinimetric evaluations of patients with chronic
widespread pain. Best Pract. Res. Clin. Rheumatol.
25, 249–270 (2011).
67. Burckhardt, C. S., Clark, S. R. & Bennett, R. M. The
fibromyalgia impact questionnaire: development and
validation. J. Rheumatol. 18, 728–733 (1991).
68. Bennett, R. M. et al. The Revised Fibromyalgia Impact
Questionnaire (FIQR): validation and psychometric
properties. Arthritis Res. Ther. 11, R120 (2009).
69. Salaffi, F. et al. Psychometric characteristics of the
Italian version of the revised Fibromyalgia Impact
Questionnaire using classical test theory and Rasch
analysis. Clin. Exp. Rheumatol. 31, S41–S49 (2013).
70. Salaffi, F. et al. Development and validation of the
self-administered Fibromyalgia Assessment Status:
a disease-specific composite measure for evaluating
treatment effect. Arthritis Res. Ther. 11, R125
(2009).
71. Iannuccelli, C. et al. Psychometric properties of
the Fibromyalgia Assessment Status (FAS) index:
a national web-based study of fibromyalgia. Clin. Exp.
Rheumatol. 29, S49–S54 (2011).
72. Häuser, W. et al. Validation of the fibromyalgia
survey questionnaire within a cross-sectional survey.
PLoS ONE 7, e37504 (2012).
73. Häuser, W., Brähler, E., Wolfe, F. & Henningsen, P.
Patient Health Questionnaire 15 as a generic measure
of severity in fibromyalgia syndrome: surveys with
patients of three different settings. J. Psychosom. Res.
76, 307–311 (2014).
74. Kosek, E. et al. Do we need a third mechanistic
descriptor for chronic pain states? Pain 157,
1382–1386 (2016).
75. Yunus, M. B. Central sensitivity syndromes: a unified
concept for fibromyalgia and other similar maladies.
J. Indian Rheumatol. Assoc. 8, 27–33 (2000).
76. Borchers, A. T. & Gershwin, M. E. Fibromyalgia: a
critical and comprehensive review. Clin. Rev. Allergy
Immunol. 49, 100–151 (2015).
77. Ceko, M., Bushnell, M. C. & Gracely, R. H. Neurobiology
underlying fibromyalgia symptoms. Pain Res. Treat.
https://doi.org/10.1155/2012/585419 (2012).
Reviews
78. Desmeules, J. A. et al. Neurophysiologic evidence for
a central sensitization in patients with fibromyalgia.
Arthritis Rheum. 48, 1420–1429 (2003).
79. Sörensen, J., Graven-Nielsen, T., Henriksson, K. G.,
Bengtsson, M. & Arendt-Nielsen, L. Hyperexcitability
in fibromyalgia. J. Rheumatol. 25, 152–155 (1998).
80. Staud, R., Vierck, C. J., Cannon, R. L., Mauderli, A. P.
& Price, D. D. Abnormal sensitization and temporal
summation of second pain (wind-up) in patients with
fibromyalgia syndrome. Pain 91, 165–175 (2001).
81. McDermid, A. J., Rollman, G. B. & McCain, G. A.
Generalized hypervigilance in fibromyalgia: evidence of
perceptual amplification. Pain 66, 133–144 (1996).
82. Geisser, M. E. et al. A psychophysical study of auditory
and pressure sensitivity in patients with fibromyalgia
and healthy controls. J. Pain 9, 417–422 (2008).
83. Martenson, M. E. et al. A possible neural mechanism
for photosensitivity in chronic pain. Pain 157,
868–878 (2016).
84. Harris, R. E. & Clauw, D. J. How do we know that the
pain in fibromyalgia is “Real”? Curr. Pain Headache
Rep. 10, 403–407 (2006).
85. Gracely, R. H., Petzke, F., Wolf, J. M. & Clauw, D. J.
Functional magnetic resonance imaging evidence
of augmented pain processing in fibromyalgia.
Arthritis Rheum. 46, 1333–1343 (2002).
86. Cook, D. B. et al. Functional imaging of pain in
patients with primary fibromyalgia. J. Rheumatol. 31,
364–378 (2004).
87. Burgmer, M. et al. Altered brain activity during pain
processing in fibromyalgia. Neuroimage 44, 502–508
(2009).
88. Jensen, K. B. et al. Evidence of dysfunctional pain
inhibition in fibromyalgia reflected in rACC during
provoked pain. Pain 144, 95–100 (2009).
89. Dehghan, M. et al. Coordinate-based (ALE)
meta-analysis of brain activation in patients with
fibromyalgia. Hum. Brain Mapp. 37, 1749–1758
(2016).
90. Napadow, V. et al. Intrinsic brain connectivity in
fibromyalgia is associated with chronic pain intensity.
Arthritis Rheum. 62, 2545–2555 (2010).
91. Pujol, J. et al. Mapping brain response to pain in
fibromyalgia patients using temporal analysis of fMRI.
PLoS ONE 4, e5224 (2009).
92. Jensen, K. B. et al. Patients with fibromyalgia display
less functional connectivity in the brain’s pain
inhibitory network. Mol. Pain 8, 32 (2012).
93. Julien, N., Goffaux, P., Arsenault, P. & Marchand, S.
Widespread pain in fibromyalgia is related to a deficit
of endogenous pain inhibition. Pain 114, 295–302
(2005).
94. Vierck, C. J. et al. The effect of maximal exercise
on temporal summation of second pain (windup)
in patients with fibromyalgia syndrome. J. Pain 2,
334–344 (2001).
95. Normand, E. et al. Pain inhibition is deficient in chronic
widespread pain but normal in major depressive
disorder. J. Clin. Psychiatry 72, 219–224 (2011).
96. Lutz, J. et al. White and gray matter abnormalities
in the brain of patients with fibromyalgia: a diffusiontensor and volumetric imaging study. Arthritis Rheum.
58, 3960–3969 (2008).
97. Russell, I. J. et al. Elevated cerebrospinal fluid levels
of substance P in patients with the fibromyalgia
syndrome. Arthritis Rheum. 37, 1593–1601
(1994).
98. Harris, R. E. et al. Decreased central mu-opioid
receptor availability in fibromyalgia. J. Neurosci. 27,
10000–10006 (2007).
99. Baraniuk, J. N., Whalen, G., Cunningham, J. &
Clauw, D. J. Cerebrospinal fluid levels of opioid
peptides in fibromyalgia and chronic low back pain.
BMC Musculoskelet. Disord. 5, 48 (2004).
100. Russell, I. J., Vaeroy, H., Javors, M. & Nyberg, F.
Cerebrospinal fluid biogenic amine metabolites in
fibromyalgia/fibrositis syndrome and rheumatoid
arthritis. Arthritis Rheum. 35, 550–556 (1992).
101. Yunus, M. B., Dailey, J. W., Aldag, J. C., Masi, A. T.
& Jobe, P. C. Plasma tryptophan and other amino
acids in primary fibromyalgia: a controlled study.
J. Rheumatol. 19, 90–94 (1992).
102. Wood, P. B. et al. Fibromyalgia patients show an
abnormal dopamine response to pain. Eur. J. Neurosci.
25, 3576–3582 (2007).
103. Wood, P. B. et al. Reduced presynaptic dopamine
activity in fibromyalgia syndrome demonstrated with
positron emission tomography: a pilot study. J. Pain 8,
51–58 (2007).
104. Harris, R. E. Elevated excitatory neurotransmitter
levels in the fibromyalgia brain. Arthritis Res. Ther. 12,
141 (2010).
105. Foerster, B. R. et al. Reduced insular γ-aminobutyric
acid in fibromyalgia. Arthritis Rheum. 64, 579–583
(2012).
106. Arnold, L. M. et al. Family study of fibromyalgia.
Arthritis Rheum. 50, 944–952 (2004).
107. Stormorken, H. & Brosstad, F. Fibromyalgia: family
clustering and sensory urgency with early onset
indicate genetic predisposition and thus a “true”
disease. Scand. J. Rheumatol. 21, 207 (1992).
108. Buskila, D., Neumann, L., Hazanov, I. & Carmi, R.
Familial aggregation in the fibromyalgia syndrome.
Semin. Arthritis Rheum. 26, 605–611 (1996).
109. Ablin, J. N. & Buskila, D. Update on the genetics
of the fibromyalgia syndrome. Best Pract. Res.
Clin. Rheumatol. 29, 20–28 (2015).
110. Smith, S. B. et al. Large candidate gene association
study reveals genetic risk factors and therapeutic
targets for fibromyalgia. Arthritis Rheum. 64,
584–593 (2012).
111. Haliloglu, S., Carlioglu, A., Akdeniz, D., Karaaslan, Y.
& Kosar, A. Fibromyalgia in patients with other
rheumatic diseases: prevalence and relationship with
disease activity. Rheumatol. Int. 34, 1275–1280
(2014).
112. Fan, A. et al. Frequency of concomitant fibromyalgia in
rheumatic diseases: monocentric study of 691 patients.
Semin. Arthritis Rheum. 47, 129–132 (2017).
113. Affaitati, G. et al. Effects of treatment of peripheral
pain generators in fibromyalgia patients. Eur. J. Pain
15, 61–69 (2011).
114. Doppler, K., Rittner, H. L., Deckart, M. & Sommer, C.
Reduced dermal nerve fiber diameter in skin biopsies
of patients with fibromyalgia. Pain 156, 2319–2325
(2015).
115. Clauw, D. J. What is the meaning of “small fiber
neuropathy” in fibromyalgia? Pain 156, 2115–2116
(2015).
116. Ren, K. & Dubner, R. Interactions between the
immune and nervous systems in pain. Nat. Med. 16,
1267–1276 (2010).
117. Sarzi-Puttini, P. et al. Anti-polymer antibodies are
correlated with pain and fatigue severity in patients
with fibromyalgia syndrome. Autoimmunity 41, 74–79
(2008).
118. Bazzichi, L. et al. Association between thyroid
autoimmunity and fibromyalgic disease severity.
Clin. Rheumatol. 26, 2115–2120 (2007).
119. Cassisi, G., Sarzi-Puttini, P. & Cazzola, M. Chronic
widespread pain and fibromyalgia: could there be
some relationships with infections and vaccinations?
Clin. Exp. Rheumatol. 29, S118–S126 (2011).
120. Wormser, G. P. et al. Long-term assessment of
fibromyalgia in patients with culture-confirmed
Lyme disease. Arthritis Rheumatol. 67, 837–839
(2015).
121. Häuser, W., Kosseva, M., Üceyler, N., Klose, P. &
Sommer, C. Emotional, physical, and sexual abuse
in fibromyalgia syndrome: a systematic review with
meta-analysis. Arthritis Care Res. 63, 808–820
(2011).
122. Häuser, W. et al. Self-reported childhood maltreatment,
lifelong traumatic events and mental disorders in
fibromyalgia syndrome: a comparison of US and
German outpatients. Clin. Exp. Rheumatol. 33,
S86–S92 (2015).
123. Paras, M. L. et al. Sexual abuse and lifetime diagnosis
of somatic disorders. JAMA 302, 550 (2009).
124. Meeus, M. et al. Heart rate variability in patients
with fibromyalgia and patients with chronic fatigue
syndrome: a systematic review. Semin. Arthritis Rheum.
43, 279–287 (2013).
125. Furlan, R. et al. Abnormalities of cardiovascular
neural control and reduced orthostatic tolerance in
patients with primary fibromyalgia. J. Rheumatol. 32,
1787–1793 (2005).
126. Martínez-Martínez, L. A., Mora, T., Vargas, A.,
Fuentes-Iniestra, M. & Martínez-Lavín, M. Sympathetic
nervous system dysfunction in fibromyalgia, chronic
fatigue syndrome, irritable bowel syndrome, and
interstitial cystitis: a review of case-control studies.
J. Clin. Rheumatol. 20, 146–150 (2014).
127. Martínez-Lavín, M. & Hermosillo, A. G. Autonomic
nervous system dysfunction may explain the
multisystem features of fibromyalgia. Semin. Arthritis
Rheum. 29, 197–199 (2000).
128. Carnevali, L., Koenig, J., Sgoifo, A. & Ottaviani, C.
Autonomic and brain morphological predictors of
stress resilience. Front. Neurosci. 12, 228 (2018).
129. Rodriguez-Raecke, R., Niemeier, A., Ihle, K., Ruether, W.
& May, A. Brain gray matter decrease in chronic pain is
the consequence and not the cause of pain. J. Neurosci.
29, 13746–13750 (2009).
130. Tan, A. C., Jaaniste, T. & Champion, D. Chronic
widespread pain and fibromyalgia syndrome:
life-course risk markers in young people. Pain Res.
Manag. https://doi.org/10.1155/2019/6584753
(2019).
131. Thompson, R. W., Arnkoff, D. B. & Glass, C. R.
Conceptualizing mindfulness and acceptance as
components of psychological resilience to trauma.
Trauma Violence Abuse 12, 220–235 (2011).
132. Bennett, J. M., Rohleder, N. & Sturmberg, J. P.
Biopsychosocial approach to understanding
resilience: Stress habituation and where to intervene.
J. Eval. Clin. Pract. 24, 1339–1346 (2018).
133. Hassett, A. L. & Finan, P. H. The role of resilience in
the clinical management of chronic pain. Curr. Pain.
Headache Rep. 20, 39 (2016).
134. Engel, G. The need for a new medical model: a
challenge for biomedicine. Science 196, 129–136
(1977).
135. Gracely, R. H. et al. Pain catastrophizing and neural
responses to pain among persons with fibromyalgia.
Brain 127, 835–843 (2004).
136. Ellingson, L. D., Stegner, A. J., Schwabacher, I. J.,
Lindheimer, J. B. & Cook, D. B. Catastrophizing
interferes with cognitive modulation of pain in
women with fibromyalgia. Pain Med. 19, 2408–2422
(2018).
137. Geisser, M. E. et al. Perception of noxious and
innocuous heat stimulation among healthy women
and women with fibromyalgia: association with
mood, somatic focus, and catastrophizing. Pain 102,
243–250 (2003).
138. Lami, M. J., Martínez, M. P., Miró, E., Sánchez, A. I.
& Guzmán, M. A. Catastrophizing, acceptance, and
coping as mediators between pain and emotional
distress and disability in fibromyalgia. J. Clin. Psychol.
Med. Settings 25, 80–92 (2018).
139. Broadbent, P., Liossi, C. & Schoth, D. E. Attentional
bias to somatosensory stimuli in chronic pain patients:
a systematic review and meta-analysis. Pain https://
doi.org/10.1097/j.pain.0000000000002040
(2020).
140. Alciati, A. et al. Childhood adversities in patients with
fibromyalgia: are they related to comorbid lifetime
major depression? Clin. Exp. Rheumatol. 35, 112–118
(2017).
141. Epstein, S. A. et al. Psychiatric disorders in patients
with fibromyalgia. Psychosomatics 40, 57–63
(1999).
142. Alciati, A., Sgiarovello, P., Atzeni, F. & Sarzi-Puttini, P.
Psychiatric problems in fibromyalgia: clinical and
neurobiological links between mood disorders and
fibromyalgia. Reumatismo 64, 268–274 (2012).
143. Sarzi-Puttini, P. et al. Dysfunctional syndromes
and fibromyalgia: a 2012 critical digest. Clin. Exp.
Rheumatol. 30, 143–151 (2012).
144. Choy, E. H. S. The role of sleep in pain and
fibromyalgia. Nat. Rev. Rheumatol. 11, 513–520
(2015).
145. Rizzi, M. et al. Influence of autonomic nervous system
dysfunction in the genesis of sleep disorders in
fibromyalgia patients. Clin. Exp. Rheumatol. 35,
74–80 (2017).
146. Lentz, M. J., Landis, C. A., Rothermel, J. & Shaver, J. L.
Effects of selective slow wave sleep disruption on
musculoskeletal pain and fatigue in middle aged
women. J. Rheumatol. 26, 1586–1592 (1999).
147. Smith, M. T., Edwards, R. R., McCann, U. D. &
Haythornthwaite, J. A. The effects of sleep deprivation
on pain inhibition and spontaneous pain in women.
Sleep 30, 494–505 (2007).
148. Moldofsky, H., Scarisbrick, P., England, R. & Smythe, H.
Musculosketal symptoms and non-REM sleep
disturbance in patients with “fibrositis syndrome”
and healthy subjects. Psychosom. Med. 37, 341–351
(1975).
149. Yalinay Dikmen, P., Yavuz, B. G. & Aydinlar, E. I. The
relationships between migraine, depression, anxiety,
stress, and sleep disturbances. Acta Neurol. Belg.
115, 117–122 (2015).
150. Sivertsen, B., Harvey, A. G., Pallesen, S. & Hysing, M.
Mental health problems in adolescents with delayed
sleep phase: results from a large population-based
study in Norway. J. Sleep. Res. 24, 11–18 (2015).
151. Haase, L. et al. When the brain does not adequately
feel the body: links between low resilience and
interoception. Biol. Psychol. 113, 37–45 (2016).
152. Giusti, E. M., Castelnuovo, G., & Molinari, E.
Differences in multidisciplinary and interdisciplinary
treatment programs for fibromyalgia: a mapping
review. Pain Res. Manag. https://doi.org/10.1155/
2017/7261468 (2017).
www.nature.com/nrrheum
Reviews
153. Gendelman, O. et al. Time to diagnosis of fibromyalgia
and factors associated with delayed diagnosis in
primary care. Best Pract. Res. Clin. Rheumatol. 32,
489–499 (­20­18­).
154. García-Ríos, M. C. et al. Effectiveness of health
education in patients with fibromyalgia: a systematic
review. Eur. J. Phys. Rehabil. Med. 55, 301–313
(2019).
155. Clauw, D. J. Fibromyalgia: a clinical review. JAMA 311,
1547–1555 (2014).
156. Häuser, W. & Fitzcharles, M. A. Facts and myths
pertaining to fibromyalgia. Dialogues Clin. Neurosci.
20, 53–62 (2018).
157. Fitzcharles, M. A., Ste-Marie, P. A. & Pereira, J. X.
Fibromyalgia: evolving concepts over the past 2
decades. Can. Med. Assoc. J. 185, 645–651 (2013).
158. Pearson, J. et al. Fibromyalgia self-management:
mapping the behaviour change techniques used in a
practice-based programme. Musculoskelet. Care 18,
372–382 (2020).
159. Schrepf, A. et al. Improvement in the spatial
distribution of pain, somatic symptoms, and
depression after a weight loss intervention. J. Pain 18,
1542–1550 (2017).
160. Busch, A. J., Barber, K. A. R., Overend, T. J.,
Peloso, P. M. J. & Schachter, C. L. Exercise for treating
fibromyalgia syndrome. Cochrane Database Syst. Rev.
17, CD003786 (2007).
161. O’Dwyer, T., Maguire, S., Mockler, D., Durcan, L. &
Wilson, F. Behaviour change interventions targeting
physical activity in adults with fibromyalgia: a
systematic review. Rheumatol. Int. 39, 805–817
(2019).
162. Bjørklund, G., Dadar, M., Chirumbolo, S. & Aaseth, J.
Fibromyalgia and nutrition: therapeutic possibilities?
Biomed. Pharmacother. 103, 531–538 (2018).
163. Pagliai, G., Giangrandi, I., Dinu, M., Sofi, F. &
Colombini, B. Nutritional Interventions in the
management of fibromyalgia syndrome. Nutrients 12,
2525 (2020).
164. Clauw, D. J. Diagnosing and treating chronic
musculoskeletal pain based on the underlying
mechanism(s). Best Pract. Res. Clin. Rheumatol. 29,
6–19 (2015).
165. Häuser, W., Petzke, F., Üçeyler, N. & Sommer, C.
Comparative efficacy and acceptability of amitriptyline,
duloxetine and milnacipran in fibromyalgia syndrome:
a systematic review with meta-analysis. Rheumatology
50, 532–543 (2011).
166. Häuser, W., Wolfe, F., Tölle, T., Uçeyler, N. & Sommer, C.
The role of antidepressants in the management of
fibromyalgia syndrome: a systematic review and
meta-analysis. CNS Drugs 26, 297–307 (2012).
167. Calandre, E. P., Rico-Villademoros, F. & Slim, M.
An update on pharmacotherapy for the treatment
of fibromyalgia. Expert. Opin. Pharmacother. 16,
1347–1368 (2015).
168. Häuser, W., Urrútia, G., Tort, S., Üçeyler, N. & Walitt, B.
Serotonin and noradrenaline reuptake inhibitors
(SNRIs) for fibromyalgia syndrome. Cochrane Database
Syst. Rev. 31, CD010292 (2013).
169. Lunn, M. P., Hughes, R. A. & Wiffen, P. J. Duloxetine
for treating painful neuropathy, chronic pain or
fibromyalgia. Cochrane Database Syst. Rev. 3,
CD007115 (2014).
170. Pickering, G. et al. Milnacipran poorly modulates pain
in patients suffering from fibromyalgia: a randomized
double-blind controlled study. Drug Des. Devel. Ther.
12, 2485–2496 (2018).
171. Üçeyler, N., Sommer, C., Walitt, B. & Häuser, W.
Anticonvulsants for fibromyalgia. Cochrane Database
Syst. Rev. 16, CD010782 (2013).
172. Cooper, T. E., Derry, S., Wiffen, P. J. & Moore, R. A.
Gabapentin for fibromyalgia pain in adults. Cochrane
Database Syst. Rev. 1, CD012188 (2017).
173. Straube, S., Derry, S., Moore, R. A. & McQuay, H. J.
Pregabalin in fibromyalgia: meta-analysis of efficacy
and safety from company clinical trial reports.
Rheumatology 49, 706–715 (2010).
174. Derry, S. et al. Pregabalin for pain in fibromyalgia in
adults. Cochrane Database Syst. Rev. 9, CD011790
(2016).
175. Alciati, A. et al. Controlled-release pregabalin in the
treatment of fibromyalgia. Expert. Rev. Neurother. 18,
617–623 (2018).
176. Tofferi, J. K., Jackson, J. L. & O’Malley, P. G. Treatment
of fibromyalgia with cyclobenzaprine: a meta-analysis.
Arthritis Rheum. 51, 9–13 (2004).
177. Giovannitti, J. A., Thoms, S. M. & Crawford, J. J.
Alpha-2 adrenergic receptor agonists: a review of
current clinical applications. Anesth. Prog. 62, 31–38
(2015).
Nature Reviews | Rheumatology
178. Malanga, G. A., Gwyn, M. W., Smith, R. & Miller, D.
Tizanidine is effective in the treatment of myofascial
pain syndrome. Pain Physician 5, 422–432 (2002).
179. See, S. & Ginzburg, R. Choosing a skeletal muscle
relaxant. Am. Fam. Physician 78, 365 (2008).
180. Littlejohn, G. O., Guymer, E. K. & Ngian, G.-S. Is there
a role for opioids in the treatment of fibromyalgia?
Pain Manag. 6, 347–355 (2016).
181. Younger, J., Noor, N., McCue, R. & MacKey, S.
Low-dose naltrexone for the treatment of fibromyalgia:
findings of a small, randomized, double-blind,
placebo-controlled, counterbalanced, crossover
trial assessing daily pain levels. Arthritis Rheum. 65,
529–538 (2013).
182. Kim, P. S. & Fishman, M. A. Low-dose naltrexone for
chronic pain: update and systemic review. Curr. Pain
Headache Rep. 24, 64 (2020).
183. Painter, J. T. & Crofford, L. J. Chronic opioid use
in fibromyalgia syndrome: a clinical review. J. Clin.
Rheumatol. 19, 72–77 (2013).
184. Cazzola, M., Sarzi-Puttini, P., Buskila, D. & Atzeni, F.
[Pharmacological treatment of fibromyalgia].
Reumatismo 59, 280–291 (2007).
185. Clauw, D. J. & Hassett, A. L. The role of centralised
pain in osteoarthritis. Clin. Exp. Rheumatol. 35,
S79–S84 (2017).
186. Moldofsky, H., Lue, F. A., Mously, C., Roth-Schechter, B.
& Reynolds, W. J. The effect of zolpidem in patients
with fibromyalgia: a dose ranging, double blind,
placebo controlled, modified crossover study.
J. Rheumatol. 23, 529–533 (1996).
187. Walitt, B., Klose, P., Üçeyler, N., Phillips, T. & Häuser, W.
Antipsychotics for fibromyalgia in adults. Cochrane
Database Syst. Rev. 2016, CD011804 (2016).
188. Calandre, E. P. et al. Quetiapine extended-release
(Seroquel-XR) versus amitriptyline monotherapy for
treating patients with fibromyalgia: a 16-week,
randomized, flexible-dose, open-label trial.
Psychopharmacology 231, 2525–2531 (2014).
189. Strouse, T. B. Cannabinoids in medical practice.
Cannabis Cannabinoid Res. 1, 38–43 (2016).
190. Walitt, B., Klose, P., Ma, F., Phillips, T. & Häuser, W.
Cannabinoids for fibromyalgia (Review). Cochrane
Database Syst. Rev. 7, CD011694 (2016).
191. Farrell, M. et al. Cannabis and cannabinoids for the
treatment of people with chronic noncancer pain
conditions. Pain 159, 1932–1954 (2018).
192. Aviram, J. & Samuelly-Leichtag, G. Efficacy of
cannabis-based medicines for pain management:
a systematic review and meta- analysis of randomized
controlled trials. Pain Physician 20, E755–E796
(2017).
193. Hill, K. P., Palastro, M. D., Johnson, B. & Ditre, J. W.
Cannabis and pain: a clinical review. Cannabis
Cannabinoid Res. 2, 96–104 (2017).
194. Fiz, J., Duràn, M., Capellà, D., Carbonell, J. & Farré, M.
Cannabis use in patients with fibromyalgia: effect on
symptoms relief and health-related quality of life.
PLoS ONE 6, e18440 (2011).
195. Sarzi-Puttini, P. et al. Medical cannabis and
cannabinoids in rheumatology: where are we now?
Expert Rev. Clin. Immunol. 15, 1019–1032 (2019).
196. Habib, G. & Artul, S. Medical cannabis for the
treatment of fibromyalgia. J. Clin. Rheumatol. 24,
255–258 (2018).
197. van de Donk, T. et al. An experimental randomized
study on the analgesic effects of pharmaceutical-grade
cannabis in chronic pain patients with fibromyalgia.
Pain 160, 860–869 (2019).
198. Yassin, M., Oron, A. & Robinson, D. Effect of adding
medical cannabis to analgesic treatment in patients
with low back pain related to fibromyalgia: an
observational cross-over single centre study.
Clin. Exp. Rheumatol. 37, 13–20 (2019).
199. Giorgi, V. et al. Adding medical cannabis to standard
analgesic treatment for fibromyalgia: a prospective
observational study. Clin. Exp. Rheumatol. 38, 53–59
(2020).
200. National Pain Report. Marijuana rated most effective
for treating fibromyalgia, http://nationalpainreport.
com/marijuana-rated-most-effective-for-treatingfibromyalgia-8823638.html (2014).
201. Rathore, F. A. & Afridi, A. Is combination
pharmacotherapy effective for management of
fibromyalgia in adults? — a Cochrane Review
summary with commentary. J. Musculoskelet.
Neuronal Interact. 20, 297–300 (2020).
202. Boomershine, C. S. & Crofford, L. J. A symptombased approach to pharmacologic management of
fibromyalgia. Nat. Rev. Rheumatol. 5, 191–199 (2009).
203. Thorpe, J., Shum, B., Ra, M., Pj, W. & Gilron, I.
Combination pharmacotherapy for the treatment of
fibromyalgia in adults (Review). Cochrane Database
Syst. Rev. 2018, CD010585 (2018).
204. Mease, P. J. et al. Milnacipran combined with
pregabalin in fibromyalgia: a randomized, open-label
study evaluating the safety and efficacy of adding
milnacipran in patients with incomplete response
to pregabalin. Ther. Adv. Musculoskelet. Dis. 5,
113–126 (2013).
205. Gilron, I. et al. Combination of pregabalin with
duloxetine for fibromyalgia. Pain 157, 1532–1540
(2016).
206. Arnold, L. M. et al. Efficacy and safety of pregabalin in
patients with fibromyalgia and comorbid depression
taking concurrent antidepressant medication: a
randomized, placebo-controlled study. J. Rheumatol.
42, 1237–1244 (2015).
207. Bernardy, K., Klose, P., Welsch, P. & Häuser, W.
Efficacy, acceptability and safety of cognitive
behavioural therapies in fibromyalgia syndrome - A
systematic review and meta-analysis of randomized
controlled trials. Eur. J. Pain 22, 242–260 (2018).
208. Jacobs, H. et al. The impact of a group-based
multidisciplinary rehabilitation program on the quality
of life in patients with fibromyalgia: results from the
QUALIFIBRO Study. J. Clin. Rheumatol. https://doi.org/
10.1097/RHU.0000000000001120 (2019).
209. Bernardy, K., Klose, P., Welsch, P. & Häuser, W.
Efficacy, acceptability and safety of Internet-delivered
psychological therapies for fibromyalgia syndrome:
a systematic review and meta-analysis of randomized
controlled trials. Eur. J. Pain 23, 3–14 (2019).
210. Perrot, S. & Russell, I. J. More ubiquitous effects
from non-pharmacologic than from pharmacologic
treatments for fibromyalgia syndrome: a meta-analysis
examining six core symptoms. Eur. J. Pain 18,
1067–1080 (2014).
211. Gálvez, I., Torres-Piles, S. & Ortega-Rincón, E.
Balneotherapy, immune system, and stress response:
a hormetic strategy? Int. J. Mol. Sci. 19, 1687 (2018).
212. Naumann, J. & Sadaghiani, C. Therapeutic benefit of
balneotherapy and hydrotherapy in the management
of fibromyalgia syndrome: a qualitative systematic
review and meta-analysis of randomized controlled
trials. Arthritis Res. Ther. 16, R141 (2014).
213. Fioravanti, A. et al. Is balneotherapy effective for
fibromyalgia? Results from a 6-month double-blind
randomized clinical trial. Clin. Rheumatol. 37,
2203–2212 (2018).
214. Honda Y. et al. Effects of physical-agent pain relief
modalities for fibromyalgia patients: a systematic
review and meta-analysis of randomized controlled
trials. Pain Res. Manag. https://doi.org/10.1155/
2018/2930632 (2018).
215. Kurt, E. E., Koçak, F. A., Erdem, H. R., Tuncay, F. &
Kelez, F. Which non-pharmacological treatment is
more effective on clinical parameters in patients with
fibromyalgia: balneotherapy or aerobic exercise?
Arch. Rheumatol. 31, 162–169 (2016).
216. Guidelli, G. M., Tenti, S., de Nobili, E. & Fioravanti, A.
Fibromyalgia syndrome and spa therapy: myth or
reality? Clin. Med. Insights Arthritis Musculoskelet.
Disord. 5, 19–26 (2012).
217. Langhorst, J., Klose, P., Dobos, G. J., Bernardy, K. &
Häuser, W. Efficacy and safety of meditative movement
therapies in fibromyalgia syndrome: a systematic
review and meta-analysis of randomized controlled
trials. Rheumatol. Int. 33, 193–207 (2013).
218. Mist, S. D., Firestone, K. A. & Jones, K. D.
Complementary and alternative exercise for
fibromyalgia: a meta-analysis. J. Pain Res. 6,
247–260 (2013).
219. Cheng, C.-A. et al. Effectiveness of Tai Chi on
fibromyalgia patients: a meta-analysis of randomized
controlled trials. Complement. Ther. Med. 46, 1–8
(2019).
220. Wang, C. et al. Effect of tai chi versus aerobic
exercise for fibromyalgia: comparative effectiveness
randomized controlled trial. BMJ 360, k851 (2018).
221. Van Gordon, W., Shonin, E., Dunn, T. J.,
Garcia-Campayo, J. & Griffiths, M. D. Meditation
awareness training for the treatment of fibromyalgia
syndrome: a randomized controlled trial. Br. J. Health
Psychol. 22, 186–206 (2017).
222. Haugmark, T., Hagen, K. B., Smedslund, G. &
Zangi, H. A. Mindfulness- and acceptance-based
interventions for patients with fibromyalgia — a
systematic review and meta-analyses. PLoS One 14,
e0221897 (2019).
223. Lauche, R., Cramer, H., Dobos, G., Langhorst, J. &
Schmidt, S. A systematic review and meta-analysis of
mindfulness-based stress reduction for the fibromyalgia
syndrome. J. Psychosom. Res. 75, 500–510 (2013).
Reviews
224. Luciano, J. V. et al. Effectiveness of group acceptance
and commitment therapy for fibromyalgia: A 6-month
randomized controlled trial (EFFIGACT study). Pain
155, 693–702 (2014).
225. Luciano, J. V. et al. Cost-utility of group acceptance
and commitment therapy for fibromyalgia versus
recommended Drugs: an economic analysis alongside
a 6-month randomized controlled trial conducted
in Spain (EFFIGACT Study). J. Pain 18, 868–880
(2017).
226. Jensen, M. P. Hypnosis for chronic pain management:
a new hope. Pain 146, 235–237 (2009).
227. Zech, N., Hansen, E., Bernardy, K. & Häuser, W.
Efficacy, acceptability and safety of guided imagery/
hypnosis in fibromyalgia — a systematic review
and meta-analysis of randomized controlled trials.
Eur. J. Pain 21, 217–227 (2017).
228. Deare, J. C. et al. Acupuncture for treating
fibromyalgia. Cochrane Database Syst. Rev. 2013,
CD007070 (2013).
229. Yang, B. et al. Efficacy of acupuncture on fibromyalgia
syndrome: a meta-analysis. J. Tradit. Chin. Med. 34,
381–391 (2014).
230. Atzeni, F. et al. Hyperbaric oxygen treatment of
fibromyalgia: a prospective observational clinical
study. Clin. Exp. Rheumatol. 37, 63–69 (2019).
231. Efrati, S. et al. Hyperbaric oxygen therapy can
diminish fibromyalgia syndrome — prospective
clinical trial. PLoS ONE 10, e0127012 (2015).
232. Moisset, X., Lanteri-Minet, M. & Fontaine, D.
Neurostimulation methods in the treatment of chronic
pain. J. Neural Transm. 127, 673–686 (2020).
233. Atzeni, F. et al. Hyperbaric oxygen therapy in
fibromyalgia and the diseases involving the central
nervous system. Clin. Exp. Rheumatol. 38, S94–S98
(2020).
234. Brighina, F. et al. Brain modulation by electric currents
in fibromyalgia: a structured review on non-invasive
approach with transcranial electrical stimulation.
Front. Hum. Neurosci. 11, 13–40 (2019).
235. Mhalla, A. et al. Long-term maintenance of the
analgesic effects of transcranial magnetic stimulation
in fibromyalgia. Pain 152, 1478–1485 (2011).
236. López-Solà, M. et al. Towards a neurophysiological
signature for fibromyalgia. Pain 158, 34–47 (2017).
237. Clauw, D. J., Essex, M. N., Pitman, V. & Jones, K. D.
Reframing chronic pain as a disease, not a symptom:
rationale and implications for pain management.
Postgrad. Med. 131, 185–198 (2019).
238. Bennett, R. Fibromyalgia: shining a light on
fibromyalgia treatment. Nat. Rev. Rheumatol. 12,
568–569 (2016).
239. Arnold, L. M. Duloxetine and other antidepressants in
the treatment of patients with fibromyalgia. Pain Med.
8, S63–S74 (2007).
240. Tzellos, T. G. et al. Gabapentin and pregabalin in the
treatment of fibromyalgia: a systematic review and
a meta-analysis. J. Clin. Pharm. Ther. 35, 639–656
(2010).
241. Üçeyler, N., Sommer, C., Walitt, B. & Häuser, W.
Anticonvulsants for fibromyalgia. Cochrane Database
Syst. Rev. 2017, 2–5 (2017).
242. Sarzi-Puttini, P. et al. Cannabinoids in the treatment of
rheumatic diseases: pros and cons. Autoimmun. Rev.
18, 102409 (2019).
243. Food and Drug Administration. Drug approval
package: Cymbalta (duloxetine hydrochloride), 20, 30,
and 60 mg capsules. FDA https://www.accessdata.fda.
gov/drugsatfda_docs/nda/2010/022516_cymbalta_
tocEDT.cfm (2010).
244. IBM Micromedex. Duloxetine (Oral Route). Mayo
Foundation for Medical Education and Research
(MFMER). https://www.mayoclinic.org/drugssupplements/duloxetine-oral-route/side-effects/
drg-20067247 (2020).
245. IBM Micromedex. Milnacipran (Oral Route).
Mayo Foundation for Medical Education and
Research (MFMER)2. https://www.mayoclinic.org/
drugs-supplements/milnacipran-oral-route/side-effects/
drg-20072479 (2020).
246. U.S. Food and Drug Administration. Drug approval
package: Savella (Milnacipran HCI) Tablets. FDA
https://www.accessdata.fda.gov/drugsatfda_docs/
nda/2009/022256s000TOC.cfm (2009).
247. U.S. Food and Drug Administration. Drug approval
package: Lyrica (pregabalin) Oral Solution 20 mg/ml.
FDA https://www.accessdata.fda.gov/drugsatfda_docs/
nda/2010/022488_lyrica_toc.cfm (2010).
Author contributions
P.S.-P., V.G. and D.M. wrote the article. P.S.-P. and F.A. substantially contributed to discussion of content. P.S.-P., V.G.,
D.M. and F.A. researched data for the article and reviewed
and edited the manuscript before submission.
Competing interests
The authors declare no competing interests.
Peer review information
Nature Reviews Rheumatology thanks M.-A. Fitzcharles,
G. Littlejohn and the other, anonymous, reviewer(s) for their
contribution to the peer review of this work.
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional
claims in published maps and institutional affiliations.
© Springer Nature Limited 2020
www.nature.com/nrrheum
Descargar