Chronic fatigue syndrome

Chronic fatigue syndrome (CFS), also called myalgic encephalomyelitis (ME) or ME/CFS, is a complex, debilitating, long-term medical condition. The causes and mechanisms of the disease are not fully understood.[12] Distinguishing core symptoms are lengthy exacerbations or flare-ups of the illness following ordinary minor physical or mental activity, known as post-exertional malaise (PEM);[13][14] greatly diminished capacity to accomplish tasks that were routine before the illness; and sleep disturbances.[13][15][1][5][2]:7 Orthostatic intolerance (difficulty sitting and standing upright) and cognitive dysfunction are also diagnostic. Frequently and variably, other common symptoms occur involving numerous body systems, and chronic pain is common.[15][16] The unexplained and often incapacitating fatigue in CFS is different from that caused by normal strenuous ongoing exertion, is not significantly relieved by rest, and is not due to a previous medical condition.[15] Diagnosis is based on the person's symptoms because no confirmed diagnostic test is available.[17]

Chronic fatigue syndrome
Other namesMyalgic encephalomyelitis/ chronic fatigue syndrome (ME/CFS),[1] myalgic encephalomyelitis (ME), post-viral fatigue syndrome (PVFS), chronic fatigue immune dysfunction syndrome (CFIDS), systemic exertion intolerance disease (SEID), others[2]:20
Chart of the symptoms of CFS according to various definitions
SpecialtyRheumatology, rehabilitation medicine, endocrinology, Infectious disease, neurology, immunology, internal medicine, paediatrics, other specialists in ME/CFS[3]
SymptomsWorsening of symptoms with activity, long-term fatigue, others[1]
Usual onset10 to 30 years old[4]
DurationOften for years[5]
CausesUnknown[1]
Risk factorsFemale sex, virus and bacterial infections, blood relatives with the illness, major injury, bodily response to severe stress and others[6][7]:1–2
Diagnostic methodBased on symptoms[1]
TreatmentSymptomatic[8][9]
PrevalenceAbout 0.68 to 1% globally[10][11]

Proposed mechanisms include biological, genetic, epigenetic, infectious, and physical or psychological stress affecting the biochemistry of the body.[6][18] Persons with CFS may recover or improve over time, but some will become severely affected and disabled for an extended period.[19] No therapies or medications are approved to treat the cause of the illness; treatment is aimed at alleviation of symptoms.[8][20] The CDC recommends pacing (personal activity management) to keep mental and physical activity from making symptoms worse.[8] Limited evidence suggests that rintatolimod, counseling,[21] and personalized activity management[20] helps improve some patients' functional abilities.

About 1% of primary-care patients have CFS; estimates of incidence vary widely because epidemiological studies define the illness dissimilarly.[11][17][10] It has been estimated that 836,000 to 2.5 million Americans and 250,000 to 1,250,000 people in the United Kingdom have CFS.[1][22] CFS occurs 1.5 to 2 times as often in women as in men.[11] It most commonly affects adults between ages 40 and 60 years;[23] it can occur at other ages, including childhood.[24] Other studies suggest that about 0.5% of children have CFS, and that it is more common in adolescents than in younger children.[2]:182[24] Chronic fatigue syndrome is a major cause of school absence.[2]:183 CFS reduces health, happiness, productivity, and can also cause socio-emotional disruptions such as loneliness and alienation.[25] However, there is controversy over many aspects of the disorder. Physicians, researchers, and patient advocates promote different names[26] and diagnostic criteria. Results of studies of proposed causes and treatments are often poor or contradictory.[27]

Signs and symptoms

The United States Centers for Disease Control and Prevention (CDC) recommends these criteria for diagnosis:[15]

  1. Greatly lowered ability to do activities that were usual before the illness. This drop in activity level occurs along with fatigue and must last six months or longer.
  2. Worsening of symptoms after physical or mental activity that would not have caused a problem before the illness. The amount of activity that might aggravate the illness is difficult for a person to predict, and the decline often presents 12 to 48 hours after the activity.[28] The 'relapse', or 'crash', may last days, weeks or longer. This is known as post-exertional malaise (PEM).
  3. Sleep problems; people may still feel weary after full nights of sleep, or may struggle to stay awake, fall asleep or stay asleep.

Additionally, one of the following symptoms must be present:[15]

  • Problems with thinking and memory (cognitive dysfunction, sometimes described as "brain fog")
  • While standing or sitting upright; lightheadedness, dizziness, weakness, fainting or vision changes may occur (orthostatic intolerance)

Other common symptoms

Many, but not all people with ME/CFS report:[15]

Increased sensitivity to sensory stimuli and pain have also been observed in CFS.[19][29]

The CDC recommends that people with symptoms of CFS consult a physician to rule out other illnesses, which may be treatable.[30]

Onset

The onset of CFS may be gradual or sudden.[2] When it begins suddenly, it often follows a period of infectious-like symptoms or a known infection, and between 20 and 80% of patients report an onset resembling an infection.[2]:158[31] When gradual, the illness may begin over the course of months or years.[32] Studies disagree as to which pattern is more common.[2]:158:181 CFS may also occur after physical trauma such as a car accident or surgery.[32]

Physical functioning

The functional capacity of individuals with CFS varies greatly.[33] Some persons with mild CFS lead relatively normal lives with vigilant energy management; persons that are severely ill may be totally bed-ridden and unable to care for themselves.[32] For the majority of persons with CFS, work, school, and family activities are significantly reduced for extended periods of time.[32][15] The severity of symptoms and disability is the same regardless of gender,[34] and many experience strongly disabling chronic pain.[35] Persons report critical reductions in levels of physical activity.[36] Also, a reduction in the complexity of activity has been observed.[37] Reported impairment is comparable to other fatiguing medical conditions[38] including late-stage AIDS,[39] lupus, rheumatoid arthritis, chronic obstructive pulmonary disease (COPD), and end-stage kidney disease.[40] CFS affects a person's functional status and well-being more than major medical conditions such as multiple sclerosis, congestive heart failure, or type II diabetes mellitus.[41][42]

Often, courses of remission and relapse of symptoms occur, which make the illness difficult to manage. Persons who feel better for a period may overextend their activities, and the result can be a worsening of their symptoms with a relapse of the illness.[28]

About 25% of people with CFS are house-bound or bed-ridden for long periods during their illness, often for decades.[2]:32[5] An estimated 75% are unable to work because of their illness.[43] More than half were on disability benefits or temporary sick leave, and less than a fifth worked full-time.[44] Children who become ill with CFS are a major cause of school absence.[2]:183

People with CFS have decreased scores on the SF-36 quality-of-life questionnaire, especially in the sub scales on vitality, physical functioning, general health, physical role, and social functioning; however, the sub scales for "role emotional" and mental health in CFS patients were consistent with or not substantially lower than healthy controls.[45]

Cognitive functioning

Cognitive dysfunction is one of the more disabling aspects of CFS due to its negative impact on occupational and social functioning. Fifty to eighty percent of persons with CFS are estimated to have serious problems with cognition.[46] Cognitive symptoms are mainly due to deficits in attention, memory, and reaction time. Measured cognitive abilities are found to be below projected normal values and likely to affect day-to-day activities; for example, increases in common mistakes, forgetting scheduled tasks, or having difficulty responding when spoken to are observed.[47]

Simple and complex information-processing speed and functions entailing working memory over long time periods are moderately to extensively impaired. These deficits are generally consistent with the patient's perceptions. Perceptual abilities, motor speed, language, reasoning, and intelligence do not appear to be significantly altered. When poorer health status was reported, a person's perception of their cognitive problems was frequently greater. Better physical functioning in people with CFS is associated with less visuoperceptual difficulty and fewer language-processing complaints.[47]

Inconsistencies of subjective and observed values of cognitive dysfunction reported across multiple studies are likely caused by a number of factors. Differences of research participants' cognitive abilities pre- and post-illness onset are naturally variable and are difficult to measure because of a lack of specialized analytical tools that can consistently quantify the specific cognitive difficulties in CFS.[47]

The frequency of neuropsychiatric and neuropsychological symptoms is increased in the population of persons with CFS; the understanding of why this occurs is unresolved. Various hypotheses have been advanced to try to explain the relationship between the cognitive symptoms and the illness. Some researchers believe psychiatric causes underlie or contribute to the illness, while other researchers believe the illness causes biochemical and sociological changes in people that produce the symptoms.[46]

Cause

The cause of CFS is unknown.[45] Genetic and physiological factors are thought to work together to precipitate and perpetuate the condition.[18] A 2016 report by the Institute of Medicine states that CFS is a biologically based illness, but that the biologic abnormalities are not sensitive or specific enough to be useful as a diagnosis.[45]

Because it may begin as an influenza-like illness with a sudden onset, various infectious causes have been proposed, but evidence is insufficient to support such causation.[48][2] Infections proposed include mononucleosis, Chlamydophila pneumoniae, human herpesvirus 6, and Lyme disease. Inflammation may be involved.[49] Often, the illness will follow a viral illness such as mononucleosis or gastroenteritis.[50]

Risk factors

All ages, ethnic groups, and income levels are susceptible to the illness. The CDC states that Caucasians may be diagnosed more frequently than other races in America,[5] but the illness is at least as prevalent among African Americans and Hispanics.[23] A 2009 meta-analysis showed that African Americans and Native Americans have a higher risk of CFS, though it specifically excluded other more common ethnicities worldwide, and it acknowledged that studies and data were limited.[51]

More women than men get CFS.[5] A large 2020 meta-analysis estimated that between 1.5 and 2.0 times more cases are women. The review acknowledged that different case definitions and diagnostic methods within datasets yielded a wide range of prevalence rates.[11] The CDC estimates CFS occurs up to four times more often in women than in men.[23] The illness can occur at any age, but has the highest prevalence in persons between the ages of 40 and 60.[23] CFS is less prevalent among children and adolescents than among adults.[24]

Blood relatives of those who have CFS appear to be more predisposed, implying that genetic factors may increase the risk of susceptibility to the illness.[14]

According to the CDC, "CFS is a biological illness, not a psychologic disorder", and those affected "are neither malingering nor seeking secondary gain".[52] The World Health Organization (WHO) classifies CFS as a neurological disease in the ICD-11 for Mortality and Morbidity Statistics (ICD-11).[53]

Viral and other infections

Post-viral fatigue syndrome (PVFS) or post-viral syndrome describes a type of chronic fatigue syndrome that occurs following a viral infection.[31] A recent review found Epstein–Barr virus (EBV) antibody activity to be higher in patients with CFS, and that a subset of patients with CFS were likely to have increased EBV activity compared to controls.[54] Viral infection is a significant risk factor for CFS, with one study finding 22% of people with EBV experience fatigue six months later, and 9% having strictly defined CFS.[55] A systematic review found that fatigue severity was the main predictor of prognosis in CFS, and did not identify psychological factors linked to prognosis.[56]

Another review found that risk factors for developing CFS after mononucleosis, dengue fever, or Q-fever included longer bed-rest during the illness, poorer pre-illness physical fitness, attributing symptoms to physical illness, belief that a long recovery time is needed, as well as pre-infection distress and fatigue.[57] The same review found biological factors such as CD4 and CD8 activation and liver inflammation are predictors of sub-acute fatigue but not CFS.[57] However, these findings are not generally accepted due to the use of the Oxford criteria in selecting patients. The CDC does not recognize attribution of symptoms as a risk factor.[6]

A study comparing diagnostic labels found that people labelled with ME had the worst prognosis, while those with PVFS had the best. Whether this is due to those with more severe or longer-lasting symptoms results in a label with the description of ME, or if being labelled with ME adversely causes a more severe or prolonged illness is unclear.[58]

Pathophysiology

Neurological

Brain imaging, comparing adolescents with CFS and healthy controls showing abnormal network activity in regions of the brain.

A range of neurological structural and functional abnormalities is found in people with CFS, including lowered metabolism at the brain stem and reduced blood flow to areas of the brain; these differences are consistent with neurological illness, but not depression or psychological illness.[7] The World Health Organization classes chronic fatigue syndrome as a central nervous system disease.[59]

Some neuroimaging studies have observed prefrontal and brainstem hypometabolism; however, sample size was limited.[60] Neuroimaging studies in persons with CFS have identified changes in brain structure and correlations with various symptoms. Results were not consistent across the neuroimaging brain structure studies, and more research is needed to resolve the discrepancies found between the disparate studies.[61][60]

Tentative evidence suggests a relationship between autonomic nervous system dysfunction and diseases such as CFS, fibromyalgia, irritable bowel syndrome, and interstitial cystitis. However, it is unknown if this relationship is causative.[62] Reviews of CFS literature have found autonomic abnormalities such as decreased sleep efficiency, increased sleep latency, decreased slow wave sleep, and abnormal heart rate response to tilt table tests, suggesting a role of the autonomic nervous system in CFS. However, these results were limited by inconsistency.[63][64][65]

Central sensitization, or increased sensitivity to sensory stimuli such as pain have been observed in CFS. Sensitivity to pain increases after exertion, which is opposite to the normal pattern.[29]

Immunological

Immunological abnormalities are frequently observed in those with CFS. Decreased NK cell activity is found more often in people with CFS and this correlates with severity of symptoms.[6][66] People with CFS have an abnormal response to exercise, including increased production of complement products, increased oxidative stress combined with decreased antioxidant response, and increased Interleukin 10, and TLR4, some of which correlates with symptom severity.[67] Increased levels of cytokines have been proposed to account for the decreased ATP production and increased lactate during exercise;[68][69] however, the elevations of cytokine levels are inconsistent in specific cytokine, albeit frequently found.[2][70] Similarities have been drawn between cancer and CFS with regard to abnormal intracellular immunological signaling. Abnormalities observed include hyperactivity of Ribonuclease L, a protein activated by IFN, and hyperactivity of NF-κB.[71]

Endocrine

Evidence points to abnormalities in the hypothalamic-pituitary-adrenal axis (HPA axis) in some, but not all, persons with CFS, which may include slightly low cortisol levels,[72] a decrease in the variation of cortisol levels throughout the day, decreased responsiveness of the HPA axis, and a high serotonergic state, which can be considered to be a "HPA axis phenotype" that is also present in some other conditions, including post-traumatic stress disorder and some autoimmune conditions.[73] It is unclear whether or not decreased cortisol levels of the HPA axis plays a primary role as a cause of CFS,[74][75][76] or has a secondary role in the continuation or worsening of symptoms later in the illness.[77] In most healthy adults, the cortisol awakening response shows an increase in cortisol levels averaging 50% in the first half-hour after waking. In people with CFS, this increase apparently is significantly less, but methods of measuring cortisol levels vary, so this is not certain.[78]

Autoimmunity

Autoimmunity has been proposed to be a factor in CFS, but there are only a few relevant findings so far. There are a subset of patients with increased B cell activity and autoantibodies, possibly as a result of decreased NK cell regulation or viral mimicry.[79] In 2015, a large German study found 29% of ME/CFS patients had elevated autoantibodies to M3 and M4 muscarinic acetylcholine receptors as well as to ß2 adrenergic receptors.[80][81][82] A 2016 Australian study found that ME/CFS patients had significantly greater numbers of single nucleotide polymorphisms associated with the gene encoding for M3 muscarinic acetylcholine receptors.[83]

Energy metabolism

Studies have observed mitochondrial abnormalities in cellular energy production, but recent focus has concentrated on secondary effects that may result in aberrant mitochondrial function because inherent problems with the mitochondria structure or genetics have not been replicated.[84]

Diagnosis

Could You Have MECFS? from US Centers for Disease Control

No characteristic laboratory abnormalities are approved to diagnose CFS; while physical abnormalities can be found, no single finding is considered sufficient for diagnosis.[85][7] Blood, urine, and other tests are used to rule out other conditions that could be responsible for the symptoms.[86][87][2] The CDC states that a medical history should be taken and a mental and physical examination should be done to aid diagnosis.[86]

Diagnostic tools

The CDC recommends considering the questionnaires and tools described in the 2015 Institute of Medicine report, which include:[88]

  • The Chalder Fatigue Scale
  • Multidimensional Fatigue Inventory
  • Fisk Fatigue Impact Scale
  • The Krupp Fatigue Severity Scale
  • DePaul Symptom Questionnaire
  • CDC Symptom Inventory for CFS
  • Work and Social Adjustment Scale (WSAS)
  • SF-36 / RAND-36[2]:270

A two-day cardiopulmonary exercise test (CPET) is not necessary for diagnosis, although lower readings on the second day may be helpful in supporting a claim for social security disability. A two-day CPET cannot be used to rule out chronic fatigue syndrome.[2]:216

Definitions

Notable definitions include:[89]

  • Centers for Disease Control and Prevention (CDC) definition (1994),[90] the most widely used clinical and research description of CFS,[18] is also called the Fukuda definition and is a revision of the Holmes or CDC 1988 scoring system.[91] The 1994 criteria require the presence of four or more symptoms beyond fatigue, while the 1988 criteria require six to eight.[92]
  • The ME/CFS 2003 Canadian Clinical working definition[93] states: "A patient with ME/CFS will meet the criteria for fatigue, post-exertional malaise and/or fatigue, sleep dysfunction, and pain; have two or more neurological/cognitive manifestations and one or more symptoms from two of the categories of autonomic, neuroendocrine, and immune manifestations; and the illness persists for at least 6 months".
  • The Myalgic Encephalomyelitis International Consensus Criteria (ICC) published in 2011 is based on the Canadian working definition and has an accompanying primer for clinicians[94][7] The ICC does not have a six months waiting time for diagnosis. The ICC requires post-exertional neuroimmune exhaustion (PENE) which has similarities with post-exertional malaise, plus at least three neurological symptoms, at least one immune or gastrointestinal or genitourinary symptom, and at least one energy metabolism or ion transportation symptom. Unrefreshing sleep or sleep dysfunction, headaches or other pain, and problems with thinking or memory, and sensory or movement symptoms are all required under the neurological symptoms criterion.[94] According to the ICC, patients with post-exertional neuroimmune exhaustion but only partially meet the criteria should be given the diagnosis of atypical myalgic encephalomyelitis.[7]
  • The 2015 definition by the National Academy of Medicine (then referred to as the "Institute of Medicine") is not a definition of exclusion (differential diagnosis is still required).[2] "Diagnosis requires that the patient have the following three symptoms: 1) A substantial reduction or impairment in the ability to engage in pre-illness levels of occupational, educational, social, or personal activities, that persists for more than 6 months and is accompanied by fatigue, which is often profound, is of new or definite onset (not lifelong), is not the result of ongoing excessive exertion, and is not substantially alleviated by rest, and 2) post-exertional malaise* 3) Unrefreshing sleep*; At least one of the two following manifestations is also required: 1) Cognitive impairment* 2) Orthostatic intolerance" and notes that "*Frequency and severity of symptoms should be assessed. The diagnosis of ME/CFS should be questioned if patients do not have these symptoms at least half the time with moderate, substantial, or severe intensity."[2]

Clinical practice guidelines are generally based on case descriptions, with the aim of improving diagnosis, management and treatment. An example is the ME/CFS guideline for the National Health Services in England and Wales, updated in 2021.[95] Other guidance can be found at the New York Department of Health.[96]

Differential diagnosis

Certain medical conditions can cause chronic fatigue and must be ruled out before a diagnosis of CFS can be given. Hypothyroidism, anemia,[97] coeliac disease (that can occur without gastrointestinal symptoms),[98] diabetes and certain psychiatric disorders are a few of the diseases that must be ruled out if the patient presents with appropriate symptoms.[95][90][97] Other diseases, listed by the Centers for Disease Control and Prevention, include infectious diseases (such as Epstein–Barr virus, influenza, HIV infection, tuberculosis, Lyme disease), neuroendocrine diseases (such as thyroiditis, Addison's disease, adrenal insufficiency, Cushing's disease), hematologic diseases (such as occult malignancy, lymphoma), rheumatologic diseases (such as fibromyalgia, polymyalgia rheumatica, Sjögren's syndrome, lupus, giant-cell arteritis, polymyositis, dermatomyositis), psychiatric diseases (such as bipolar disorder, schizophrenia, delusional disorders, dementia, anorexia/bulimia nervosa), neuropsychologic diseases (such as obstructive sleep apnea, parkinsonism, multiple sclerosis), and others (such as nasal obstruction from allergies, sinusitis, anatomic obstruction, autoimmune diseases, cancer, chronic hepatitis, some chronic illness, alcohol or other substance abuse, pharmacologic side effects, heavy metal exposure and toxicity, marked body weight fluctuation).[97] Ehlers–Danlos syndromes (EDS) may also have similar symptoms.[99] Medications can also cause side effects that mimic symptoms of CFS.[30]

Persons with fibromyalgia (FM, or fibromyalgia syndrome, FMS), like those with CFS, have muscle pain, severe fatigue and sleep disturbances. The presence of allodynia (abnormal pain responses to mild stimulation) and of extensive tender points in specific locations differentiates FM from CFS, although the two diseases often co-occur.[100]

Depressive symptoms, if seen in CFS, may be differentially diagnosed from primary depression by the absence of anhedonia, decreased motivation, and guilt; and the presence of somatic symptoms such as sore throat, swollen lymph nodes, and exercise intolerance with post exertional exacerbation of symptoms.[97]

Management

There is no approved pharmacological treatment, therapy or cure for CFS,[8][95] although various drugs have been or are being investigated.[101] A 2014 report prepared by the Agency for Healthcare Research and Quality stated that there are wide variations in patient management, that many receive a multifaceted approach to treatment, and that no medications have been approved by the US Food and Drug Administration (FDA) for the treatment of ME/CFS, although several have been used off label. The report concluded that although counseling and graded exercise therapy (GET) have shown some benefits, these interventions have not been studied fully enough to recommend them for all persons affected. The report expressed concern that GET appears to be associated with worsening symptoms in some.[102] The CDC no longer recommends these interventions, and there is some evidence of patient harm.[103][104]

The CDC guide for the management of CFS states that while there is no cure, a number of methods might improve symptoms.[8] Treatment strategies for sleep problems; pain; depression, stress, and anxiety; dizziness and lightheadedness (orthostatic intolerance); and memory and concentration problems are enumerated. Other useful topics that patients and doctors might discuss include carefully monitoring and managing activity to avoid worsening of symptoms, counseling to cope with the impact the illness may have on quality of life, proper nutrition and nutritional supplements that may support better health, and complementary therapies that might help increase energy or decrease pain.[8]

The United Kingdom's National Institute for Health and Clinical Excellence (NICE) 2021 guideline directed toward healthcare professionals and patients, specifies the need for shared decision-making between patients and medical care teams and acknowledges the reality and impact of the condition and the symptoms. The NICE guideline covers diagnosis, illness management, and aspects of symptom management: diet, medications, coexisting conditions, flare-ups, and energy management. The guideline recognized symptoms of severe ME/CFS may be misunderstood as neglect or abuse and recommends assessment for safeguarding of persons suspected of having ME/CFS be evaluated by professionals with experience and understanding of the illness. The guideline states that GET is not an appropriate treatment for ME/CFS. CBT might be offered to help a person manage the difficulties of dealing with a chronic illness, not to cure the illness.[95]

Prior to publication of the NICE 2021 guideline, Andrew Goddard, president of the Royal College of Physicians, stated there was concern NICE did not adequately consider the experts' support and evidence of the benefits of GET and CBT, and urged they be included in the guideline. Various ME/CFS patient groups disputed the benefits of the therapies and stated that GET can make the illness more severe.[105][106]

Comorbid conditions can occur in CFS that may interact with and exacerbate the symptoms of CFS. Appropriate medical intervention for these conditions may be beneficial. The most commonly diagnosed include fibromyalgia, irritable bowel syndrome, depression, anxiety, allergies, and chemical sensitivities.[107]

Pacing

Pacing, or activity management, is an illness management strategy based on the observation that symptoms tend to increase following mental or physical exertion,[8] and was recommended for CFS in the 1980s.[108] It is now commonly used as a management strategy in chronic illnesses and in chronic pain.[109]

Its two forms are symptom-contingent pacing, in which the decision to stop (and rest or change an activity) is determined by self-awareness of an exacerbation of symptoms, and time-contingent pacing, which is determined by a set schedule of activities that a patient estimates he or she is able to complete without triggering post-exertional malaise (PEM). Thus, the principle behind pacing for CFS is to avoid overexertion and an exacerbation of symptoms. It is not aimed at treating the illness as a whole. Those whose illness appears stable may gradually increase activity and exercise levels, but according to the principle of pacing, must rest or reduce their activity levels if it becomes clear that they have exceeded their limits.[108][20] Use of a heart-rate monitor with pacing to monitor and manage activity levels is recommended by a number of patient groups,[110] and the CDC considers it useful for some individuals to help avoid post-exertional malaise.[8]

Energy envelope theory

Energy envelope theory, considered to be consistent with pacing, is a management strategy suggested in the 2011 international consensus criteria for ME, which refers to using an "energy bank budget". Energy envelope theory was devised by psychologist Leonard Jason, who previously had CFS.[111] Energy envelope theory states that patients should stay within, and avoid pushing through, the envelope of energy available to them, so as to reduce the post-exertional malaise "payback" caused by overexertion. This may help them make "modest gains" in physical functioning.[112][113] Several studies have found energy envelope theory to be a helpful management strategy, noting that it reduces symptoms and may increase the level of functioning in CFS.[114][115][113] Energy envelope theory does not recommend unilaterally increasing or decreasing activity and is not intended as a therapy or cure for CFS.[114] It has been promoted by various patient groups.[116][117] Some patient groups recommend using a heart rate monitor to increase awareness of exertion and enable patients to stay within their aerobic threshold envelope.[118][119] Despite a number of studies showing positive results for energy envelope theory, randomized controlled trials are lacking.

Exercise

Stretching, movement therapies, and toning exercises are recommended for pain in patients with CFS, and pain medication is also suggested. In many chronic illnesses, aerobic exercise is beneficial, but in chronic fatigue syndrome, the CDC does not recommend it. The CDC states:[8]

Any activity or exercise plan for people with ME/CFS needs to be carefully designed with input from each patient. While vigorous aerobic exercise can be beneficial for many chronic illnesses, patients with ME/CFS do not tolerate such exercise routines. Standard exercise recommendations for healthy people can be harmful for patients with ME/CFS. However, it is important that patients with ME/CFS undertake activities that they can tolerate...

Counseling

The CDC states that counseling may help patients cope with pain caused by CFS, and that talking with a professional counselor or therapist may help people to more effectively manage the symptoms that affect their quality of daily life.[8]

Nutrition

A proper diet is a significant contributor to the health of any individual. Medical consultation about diet and supplements is recommended for persons with CFS.[8] Persons with CFS may benefit from a balanced diet and properly supervised administration of nutritional support if deficiencies are detected by medical testing. Risks of nutritional supplements include interactions with prescribed medications.[120][8]

Treatment

Cognitive behavioral therapy

NICE indicates CBT might be offered to help cope with the difficulty of dealing with the symptoms of ME/CFS, but should not be intended to be curative.[95] The rationale behind the use of CBT to change beliefs about the illness is disputed.[103] The CDC states that speaking with a therapist may help people cope with the illness.[8]

A 2015 National Institutes of Health report concluded that while counseling and behavior therapies could produce benefits for some people, they may not yield improvement in quality of life, and because of this limitation such therapies should not be considered as a primary treatment, but rather should be used only as one component of a broader approach.[121] This same report stated that although counseling approaches have shown benefit in some measures of fatigue, function and overall improvement, these approaches have been inadequately studied in subgroups of the wider CFS patient population. Further concern was expressed that reporting of negative effects experienced by patients receiving counseling and behavior therapies had been poor.[102] A report by the Institute of Medicine published in 2015 states that it is unclear whether CBT helps to improve cognitive impairments experienced by patients.[2]:265

A 2014 systematic review reported that there was only limited evidence that patients increased levels of physical activity after receiving CBT. The authors concluded that, as this finding is contrary to the cognitive behavioural model of CFS, patients receiving CBT were adapting to the illness rather than recovering from it.[122] In a letter published online in the Lancet in 2016, Dr Charles Shepherd, medical advisor to the MEA, expressed the view that the contention between patients and researchers lay in "a flawed model of causation that takes no account of the heterogeneity of both clinical presentations and disease pathways that come under the umbrella diagnosis of ME/CFS".[123]

Patient organisations have rebuffed the use of CBT as a treatment for CFS to alter illness beliefs.[104][124] The ME Association (MEA) recommended in 2015, based on the results of an opinion survey of 493 patients who had received CBT treatment in the UK, CBT in its current form should not be used as a primary intervention for people with CFS.[125] In 2019, a large UK survey of people with ME/CFS reported that CBT was ineffective for more than half of respondents.[126]

Graded exercise therapy

Recommendation for treatment using graded exercise therapy (GET) was removed from NICE's updated Guidelines for Diagnosis and Management of ME/CFS in October 2021. It was removed due to low quality evidence regarding benefit, with the guidelines now stating that clinicians should not prescribe "any programme that ... uses fixed incremental increases in physical activity or exercise, for example, graded exercise therapy." [20]

Previously, the National Institutes of Health concluded that while GET could produce benefits, it may not yield improvement in quality of life, and because of this limitation GET should not be considered as a primary treatment. It was recommended to be used only as one component of a broader approach. It noted that a focus on exercise programs had discouraged patient participation in other types of physical activity due to concerns of increased symptoms.[121] An addendum stated, if studies based on the Oxford criteria were excluded, there would be insufficient evidence of the effectiveness of GET.[104]

A 2019 updated Cochrane review stated that, exercise therapy probably has a positive effect on fatigue in adults, and slightly improves sleep, but the long-term effects are unknown and has limited relevance to current definitions of ME/CFS.[127][9] Cochrane started re-evaluating the effects of exercise therapies in chronic fatigue syndrome in 2020.[9][128]

Patient organisations have long criticised the use of exercise therapy, most notably GET, as a treatment for CFS.[124] Based on an opinion survey of patients who had received GET, in 2015 the ME Association concluded, GET in its current delivered form should not be recommended as a primary intervention for persons with CFS.[125]

Adaptive pacing therapy

APT, not to be confused with pacing,[129] is a therapy rather than a management strategy.[130] APT is based on the idea that CFS involves a person only having a limited amount of available energy, and using this energy wisely will mean the "limited energy will increase gradually".[130]:5 A large clinical trial known as the PACE trial found APT was no more effective than usual care or specialized medical care.[131] The PACE trial generated much criticism due to the broad Oxford criteria patient selection, the standards of outcome effectiveness were lowered during the study and that re-analysis of the data did not support the magnitude of improvements initially reported.[132]

Unlike pacing, APT is based on the cognitive behavioral model of chronic fatigue syndrome and involves increasing activity levels, which it states may temporarily increase symptoms.[133] In APT, the patient first establishes a baseline level of activity, which can be carried out consistently without any post-exertional malaise ("crashes"). APT states that persons should plan to increase their activity, as able. However, APT also requires patients to restrict their activity level to only 70% of what they feel able to do, while also warning against too much rest.[130] This has been described as contradictory, and Jason states that in comparison with pacing, this 70% limit restricts the activities that patients are capable of and results in a lower level of functioning.[129] Jason and Goudsmit, who first described pacing and the energy envelope theory for CFS, have both criticized APT for being inconsistent with the principles of pacing and highlighted significant differences.[129] APT was promoted by Action for ME, the patient charity involved in the PACE trial, until 2019.[133]

Rintatolimod

Rintatolimod is a double-stranded RNA drug developed to modulate an antiviral immune reaction through activation of toll-like receptor 3. In several clinical trials of CFS, the treatment has shown a reduction in symptoms, but improvements were not sustained after discontinuation.[134] Evidence supporting the use of rintatolimod is deemed low to moderate.[21] The US FDA has denied commercial approval, called a new drug application, citing several deficiencies and gaps in safety data in the trials, and concluded that the available evidence is insufficient to demonstrate its safety or efficacy in CFS.[135][136] Rintatolimod has been approved for marketing and treatment for persons with CFS in Argentina,[137] and in 2019, FDA regulatory requirements were met for exportation of rintatolimod to the country.[138] Rintatolimod is currently in an experimental trial in the USA to treat both ME/CFS and Long Covid.[139]

Prognosis

Information on the prognosis of CFS is limited, and the course of the illness is variable.[140] According to the NICE guideliene, CFS "varies in long-term outlook from person to person."[141] Complete recovery, partial improvement, and worsening are all possible.[140] Symptoms generally fluctuate over days, weeks, or longer periods, and some people may experience periods of remission.[141] Overall, "many will need to adapt to living with ME/CFS."[141] Some people who improve need to manage their activities in order to stay improved.[140] Children and teenagers are more likely to recover or improve than adults.[140][141]

A 2005 systematic review found that for untreated CFS, "the median full recovery rate was 5% (range 0–31%) and the median proportion of patients who improved during follow-up was 39.5% (range 8–63%)," and that 8 to 30% of patients were able to return to work. Age at onset, a longer duration of follow-up, less fatigue severity at baseline, and other factors were occasionally, but non consistently, related to outcome.[142] Another review found that children have a better prognosis than adults, with 54–94% having recovered by follow-up, compared to less than 10% of adults returning to pre-illness levels of functioning.[143]

Epidemiology

Incidence rates by age and sex, from a 2014 study in Norway

The prevalence rates for CFS/ME vary widely depending on "case definitions and diagnostic methods".[11] Based on the 1994 CDC diagnostic criteria, the global prevalence rate for CFS is 0.89%.[11] In comparison, the prevalence rate for the stricter criteria, such as the 1988 CDC "Holmes" criteria for CFS and the 2003 Canadian criteria for ME (both of which, for example, exclude patients with psychiatric diagnoses), produce an incidence rate of only 0.17%.[11] For an example of how these rates impact a nation: the CDC website notes that between 836,000 and 2.5 million Americans have ME/CFS, "but most remain undiagnosed".[1]

Females are diagnosed about 1.5 to 2.0 times more often with CFS than males.[11] An estimated 0.5% of children have CFS, and more adolescents are affected with the illness than younger children.[2]:182[24]

The incidence rate according to age has two peaks, one at 10–19 and another at 30–39 years. The effect is seen both in female and in male data, but is more pronounce in females. It was suggested that this occurs because these age groups may be more vulnerable to CFS.[144][4][145] The rate of prevalence is highest between ages 40 and 60.[45][146]

History

Myalgic encephalomyelitis

  • From 1934 onwards, outbreaks of a previously unknown illness began to be recorded by doctors.[147][148] Initially considered to be occurrences of poliomyelitis, the illness was subsequently referred to as "epidemic neuromyasthenia".[148]
  • In the 1950s, the term "benign myalgic encephalomyelitis" was used in relation to a comparable outbreak at the Royal Free Hospital in London.[149] The descriptions of each outbreak were varied, but included symptoms of malaise, tender lymph nodes, sore throat, pain, and signs of encephalomyelitis.[150] The cause of the condition was not identified, although it appeared to be infectious, and the term "benign myalgic encephalomyelitis" was chosen to reflect the lack of mortality, the severe muscular pains, symptoms suggesting damage to the nervous system, and to the presumed inflammatory nature of the disorder. Björn Sigurðsson disapproved of the name, stating that the illness is rarely benign, does not always cause muscle pain, and is possibly never encephalomyelitic.[147] The syndrome appeared in sporadic as well as epidemic cases.[151]
  • In 1969, benign myalgic encephalomyelitis appeared as an entry to the International Classification of Diseases under Diseases of the nervous system.[152]
  • In 1986, Ramsay published the first diagnostic criteria for ME, in which the condition was characterized by: 1) muscle fatiguability in which, even after minimal physical effort, three or more days elapse before full muscle power is restored; 2) extraordinary variability or fluctuation of symptoms, even in the course of one day; and 3) chronicity.[153]
  • By 1988, the continued work of Ramsay had demonstrated that, although the disease rarely resulted in mortality, it was often severely disabling.[2]:28–29 Because of this, Ramsay proposed that the prefix "benign" be dropped.[149][154][155]

Chronic fatigue syndrome

  • In the mid-1980s, two large outbreaks of an illness that resembled mononucleosis drew national attention in the United States. Located in Nevada and New York, the outbreaks involved an illness characterized by "chronic or recurrent debilitating fatigue, and various combinations of other symptoms, including a sore throat, lymph node pain and tenderness, headache, myalgia, and arthralgias". An initial link to the Epstein-Barr virus had the illness acquire the name "chronic Epstein-Barr virus syndrome".[2]:29[91]
  • In 1987, the CDC convened a working group to reach a consensus on the clinical features of the illness. The working group concluded that CFS was not new, and that the many different names given to it previously reflected widely differing concepts of the illness's cause and epidemiology.[156] The CDC working group chose "chronic fatigue syndrome" as a more neutral and inclusive name for the illness, but noted that "myalgic encephalomyelitis" was widely accepted in other parts of the world.[91]
  • In 1988, the first definition of CFS was published. Although the cause of the illness remained unknown, several attempts were made to update this definition, most notably in 1994.[90]
  • The most widely referenced diagnostic criteria and definition of CFS for research and clinical purposes were published in 1994 by the CDC.[58]
  • In 2006, the CDC commenced a national program to educate the American public and health-care professionals about CFS.[157]

Other medical terms

A range of both theorised and confirmed medical entities and naming conventions have appeared historically in the medical literature dealing with ME and CFS. These include:

  • Epidemic neuromyasthenia was a term used for outbreaks with symptoms resembling poliomyelitis.[147][158]
  • Iceland disease and Akureyri disease were synonymous terms used for an outbreak of fatigue symptoms in Iceland.[159]
  • Low natural killer syndrome, a term used mainly in Japan, reflected research showing diminished in vitro activity of natural killer cells isolated from patients.[160][161]
  • Neurasthenia has been proposed as an historical diagnosis that occupied a similar medical and cultural space to CFS.[162]
  • Royal Free disease was named after the historically significant outbreak in 1955 at the Royal Free Hospital used as an informal synonym for "benign myalgic encephalomyelitis".[163]
  • Tapanui flu was a term commonly used in New Zealand, deriving from the name of a town, Tapanui, where numerous people had the syndrome.[164]

Society and culture

Presentation of a petition to the National Assembly for Wales relating to ME support in South East Wales

Naming

Many names have been proposed for the illness. Currently, the most commonly used are "chronic fatigue syndrome", "myalgic encephalomyelitis", and the umbrella term "ME/CFS". Reaching consensus on a name is challenging because the cause and pathology remain unknown.[2]:29–30

The term "chronic fatigue syndrome" has been criticized by some patients as being both stigmatizing and trivializing, and which in turn prevents the illness from being seen as a serious health problem that deserves appropriate research.[165] While many patients prefer "myalgic encephalomyelitis", which they believe better reflects the medical nature of the illness,[153][166] there is resistance amongst some clinicians toward the use of myalgic encephalomyelitis on the grounds that the inflammation of the central nervous system (myelitis) implied by the term has not been demonstrated.[167][168]

A 2015 report from the Institute of Medicine recommended the illness be renamed "systemic exertion intolerance disease", (SEID), and suggested new diagnostic criteria, proposing post-exertional malaise, (PEM), impaired function, and sleep problems are core symptoms of ME/CFS. Additionally, they described cognitive impairment and orthostatic intolerance as distinguishing symptoms from other fatiguing illnesses.[2][169][170]

Economic impact

Reynolds et al. (2004)[171] estimated that the illness caused about $20,000 per person with CFS in lost productivity, which totals to $9.1 billion per year in the United States.[172]:29 This is comparable to other chronic illnesses that extract some of the biggest medical and socioeconomic costs.[173] Direct healthcare costs are estimated at between $9 and $14 billion annually in the US alone.[43] A 2008 study[174] calculated that the total annual cost burden of ME/CFS to society in the US was extensive, and could approach $24.0 billion.[175] A 2017 estimate for the annual economic burden in the United Kingdom from ME/CFS was 3.3 billion pounds sterling.[14]

Awareness day

12 May is designated as ME/CFS International Awareness Day.[176] The day is observed so that stakeholders have an occasion to improve the knowledge of "the public, policymakers, and health-care professionals about the symptoms, diagnosis, and treatment of ME/CFS, as well as the need for a better understanding of this complex illness."[177] It was chosen because it is the birthday of Florence Nightingale, who had an illness appearing similar to ME/CFS or fibromyalgia.[176][178]

Doctor–patient relations

Some in the medical community do not recognize CFS as a real condition, nor does agreement exist on its prevalence.[179][180][181] There has been much disagreement over proposed causes, diagnosis, and treatment of the illness.[182][183][184][185][186] This uncertainty can significantly affect doctor-patient relations. A 2006 survey of GPs in southwest England found that despite more than two-thirds of them accepting CFS/ME as a recognizable clinical entity, nearly half did not feel confident with making the diagnosis and/or treating the disease. Three other key factors that were significantly, positively associated with GPs' attitudes were knowing someone socially with CFS/ME, being male, and seeing more patients with the condition in the last year.[187]

From the patient perspective, one 1997 study found that 77% of individuals with CFS reported negative experiences with health-care providers.[41] In a more recent metaanalysis of qualitative studies, a major theme identified in patient discourses was that they felt severely ill, yet were blamed and dismissed.[188] A study of themes in patient newsgroup postings noted key themes relating to denial of social recognition of suffering and feelings of being accused of "simply faking it". Another theme that emerged strongly was that achieving diagnosis and acknowledgement requires tremendous amounts of "hard work" by patients.[181][189]

Blood donation

In 2010, several national blood banks adopted measures to discourage or prohibit individuals diagnosed with CFS from donating blood, based on concern following the 2009 claim of a link,[190] between CFS and a retrovirus which was subsequently shown to be unfounded. Organizations adopting these or similar measures included the Canadian Blood Services,[191] the New Zealand Blood Service,[192] the Australian Red Cross Blood Service[193] and the American Association of Blood Banks,[194] In November 2010, the UK National Blood Service introduced a permanent deferral of donation from ME/CFS patients based on the potential harm to those patients that may result from their giving blood.[195] Donation policy in the UK now states, "The condition is relapsing by nature and donation may make symptoms worse, or provoke a relapse in an affected individual."[196]

Controversy

Much contention has arisen over the cause, pathophysiology,[197] nomenclature,[198] and diagnostic criteria of CFS.[182][183] Historically, many professionals within the medical community were unfamiliar with CFS, or did not recognize it as a real condition; nor did agreement exist on its prevalence or seriousness.[180][181][199] Some people with CFS reject any psychological component.[200]

In 1970, two British psychiatrists, McEvedy and Beard, reviewed the case notes of 15 outbreaks of benign myalgic encephalomyelitis and concluded that it was caused by mass hysteria on the part of patients, or altered medical perception of the attending physicians.[201] Their conclusions were based on previous studies that found many normal physical test results, a lack of a discernible cause, and a higher prevalence of the illness in females. Consequently, the authors recommended that the disease should be renamed "myalgia nervosa". This perspective was rejected in a series of case studies by Dr. Melvin Ramsay and other staff of the Royal Free Hospital, the center of a significant outbreak.[202] The psychological hypothesis posed by McEvedy and Beard created great controversy, and convinced a generation of health professionals in the UK that this could be a plausible explanation for the condition, resulting in neglect by many medical specialties.[167] The specialty that did take a major interest in the illness was psychiatry.[203]

Because of the controversy, sociologists hypothesized that stresses of modern living might be a cause of the illness, while some in the media used the term "Yuppie flu" and called it a disease of the middle class. People with disabilities from CFS were often not believed and were accused of being malingerers.[203] The November 1990 issue of Newsweek ran a cover story on CFS, which although supportive of an organic cause of the illness, also featured the term 'yuppie flu', reflecting the stereotype that CFS mainly affected yuppies. The implication was that CFS is a form of burnout. The term 'yuppie flu' is considered offensive by both patients and clinicians.[204][205]

In 2009, the journal Science[190] published a study that identified the XMRV retrovirus in a population of people with CFS. Other studies failed to reproduce this finding,[206][207][208] and in 2011, the editor of Science formally retracted its XMRV paper[209] while the Proceedings of the National Academy of Sciences similarly retracted a 2010 paper which had appeared to support the finding of a connection between XMRV and CFS.[210]

United Kingdom

The lack of research funding and the funding bias towards biopsychosocial studies and against biomedical studies has been highlighted a number of times by patient groups and a number of UK politicians.[211] A parliamentary inquiry by an ad hoc group of parliamentarians in the United Kingdom, set up and chaired by former MP, Dr Ian Gibson, called the Group on Scientific Research into CFS/ME,[212]:169–86[213] was addressed by a government minister claiming that few good biomedical research proposals have been submitted to the Medical Research Council (MRC) in contrast to those for psychosocial research. They were also told by other scientists of proposals that have been rejected, with claims of bias against biomedical research. The MRC confirmed to the group that from April 2003 to November 2006, it has turned down 10 biomedical applications relating to CFS/ME and funded five applications relating to CFS/ME, mostly in the psychiatric/psychosocial domain.[213]

In 2008, the MRC set up an expert group to consider how the MRC might encourage new high-quality research into CFS/ME and partnerships between researchers already working on CFS/ME and those in associated areas. It currently lists CFS/ME with a highlight notice, inviting researchers to develop high-quality research proposals for funding.[214] In February 2010, the All-Party Parliamentary Group on ME (APPG on ME) produced a legacy paper, which welcomed the recent MRC initiative, but felt that far too much emphasis in the past had been on psychological research, with insufficient attention to biomedical research, and that further biomedical research must be undertaken to help discover a cause and more effective forms of management for this disease.[215]

Controversy surrounds psychologically oriented models of the disease and behavioral treatments conducted in the UK.[216]

United States

In 1998, $13 million for CFS research was found to have been redirected or improperly accounted for by the United States CDC, and officials at the agency misled Congress about the irregularities. The agency stated that they needed the funds to respond to other public-health emergencies. The director of a US national patient advocacy group charged the CDC had a bias against studying the disease. The CDC pledged to improve their practices and to restore the $13 million to CFS research over three years.[217]

On 29 October 2015, the National Institutes of Health declared its intent to increase research on ME/CFS. The NIH Clinical Center was to study individuals with ME/CFS, and the National Institute of Neurological Disorders and Stroke would lead the Trans-NIH ME/CFS Research Working Group as part of a multi-institute research effort.[218]

Notable cases

In 1989, The Golden Girls (1985–1992) featured chronic fatigue syndrome in a two-episode arc, "Sick and Tired: Part 1" and "Part 2", in which protagonist Dorothy Zbornak, portrayed by Bea Arthur, after a lengthy battle with her doctors in an effort to find a diagnosis for her symptoms, is finally diagnosed with CFS.[219] American author Ann Bannon had CFS.[220] Laura Hillenbrand, author of the popular book Seabiscuit, has struggled with CFS since age 19.[221][222]

Research

The different case definitions used to research the illness influence the types of patients selected for studies,[85] and research also suggests subtypes of patients may exist within a heterogeneous population.[172][223][224][225] In one of the definitions, symptoms are accepted that may suggest a psychiatric disorder, while others specifically exclude primary psychiatric disorders.[89] The lack of a single, unifying case definition was criticized in the Institute of Medicine's 2015 report for "creating an unclear picture of the symptoms and signs of the disorder" and "complicating comparisons of the results" (study results).[2]:72

More robust diagnostic methods are being investigated with the aim of identifying unique biomarkers that may be used in clinical testing. In 2019, two different papers were published proposing blood-based biomarkers for CFS.[226][227] One found that blood cells of CFS patients could be distinguished from healthy controls by their response to hyperosmotic stress.[228] Another found that the red blood cells of CFS patients were stiffer, and thus less able to deform in order to pass through capilliaries.[229]

See also

References

  1. "Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) | CDC". www.cdc.gov. 13 April 2020. Archived from the original on 22 August 2020. Retrieved 20 May 2020.
  2. Committee on the Diagnostic Criteria for Myalgic Encephalomyelitis/Chronic Fatigue Syndrome; Board on the Health of Select Populations; Institute of Medicine (10 February 2015). Beyond Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: Redefining an Illness (PDF). PMID 25695122. Archived (PDF) from the original on 20 January 2017. Retrieved 28 July 2020.
  3. "Recommendations - Myalgic encephalomyelitis (or encephalopathy)/chronic fatigue syndrome: diagnosis and management - Guidance". NICE. 29 October 2021. Archived from the original on 29 December 2021. Retrieved 15 January 2022.
  4. Crawley E (2017). "Pediatric chronic fatigue syndrome: current perspectives". Pediatric Health, Medicine and Therapeutics. 9: 27–33. doi:10.2147/PHMT.S126253. PMC 5919160. PMID 29722371.
  5. "What is ME/CFS? | Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) | CDC". www.cdc.gov. 12 July 2018. Archived from the original on 17 August 2020. Retrieved 21 May 2020.
  6. "Possible Causes | Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) | CDC". www.cdc.gov. 15 May 2019. Archived from the original on 22 August 2020. Retrieved 20 May 2020.
  7. Carruthers BM, van de Sande MI, De Meirleir KL, Klimas NG, Broderick G, Mitchell T, et al. (International Consensus Panel) (2012). "Myalgic Encephalomyelitis – Adult & Paediatric: International Consensus Primer for Medical Practitioners" (PDF). Archived from the original (PDF) on 10 July 2020.
  8. "Treatment of ME/CFS | Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) | CDC". www.cdc.gov. 19 November 2019. Archived from the original on 20 March 2021. Retrieved 22 May 2020. This article incorporates text from this source, which is in the public domain.
  9. Cochrane (21 May 2020). "Publication of Cochrane Review: 'Exercise therapy for chronic fatigue syndrome'". www.cochrane.org. Archived from the original on 17 June 2020. Retrieved 24 May 2020. It now places more emphasis on the limited applicability of the evidence to definitions of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) used in the included studies, the long-term effects of exercise on symptoms of fatigue, and acknowledges the limitations of the evidence about harms that may occur.
  10. Sandler CX, Lloyd AR (May 2020). "Chronic fatigue syndrome: progress and possibilities". The Medical Journal of Australia. 212 (9): 428–433. doi:10.5694/mja2.50553. PMID 32248536. S2CID 214810583.
  11. Lim EJ, Ahn YC, Jang ES, Lee SW, Lee SH, Son CG (February 2020). "Systematic review and meta-analysis of the prevalence of chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME)". Journal of Translational Medicine. 18 (1): 100. doi:10.1186/s12967-020-02269-0. PMC 7038594. PMID 32093722.
  12. "Etiology and Pathophysiology | Presentation and Clinical Course | Healthcare Providers | Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) | CDC". Cdc.gov. 12 July 2018. Archived from the original on 18 July 2018. Retrieved 8 March 2022.
  13. "Information for Healthcare Providers | CDC". www.cdc.gov. 13 April 2020. Archived from the original on 9 August 2020. Retrieved 17 June 2020.
  14. Dibble JJ, McGrath SJ, Ponting CP (September 2020). "Genetic risk factors of ME/CFS: a critical review". Human Molecular Genetics. 29 (R1): R117–R124. doi:10.1093/hmg/ddaa169. PMC 7530519. PMID 32744306.
  15. "Symptoms of ME/CFS | Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) | CDC". www.cdc.gov. 19 November 2019. Archived from the original on 22 August 2020. Retrieved 20 May 2020.
  16. "Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) | CDC". www.cdc.gov. 13 April 2020. Archived from the original on 22 August 2020. Retrieved 20 May 2020.
  17. Estévez-López F, Mudie K, Wang-Steverding X, Bakken IJ, Ivanovs A, Castro-Marrero J, et al. (May 2020). "Systematic Review of the Epidemiological Burden of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome Across Europe: Current Evidence and EUROMENE Research Recommendations for Epidemiology". Journal of Clinical Medicine. MDPI AG. 9 (5): 1557. doi:10.3390/jcm9051557. PMC 7290765. PMID 32455633.
  18. Afari N, Buchwald D (February 2003). "Chronic fatigue syndrome: a review". The American Journal of Psychiatry. 160 (2): 221–36. doi:10.1176/appi.ajp.160.2.221. PMID 12562565. S2CID 8210151.
  19. "Severely Affected Patients – Clinical Care of Patients – Healthcare Providers – Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS)". CDC. 19 November 2019. Archived from the original on 11 December 2020. Retrieved 30 November 2020.
  20. "Recommendations - Myalgic encephalomyelitis (or encephalopathy)/chronic fatigue syndrome: diagnosis and management - Guidance". NICE. 29 October 2021. Archived from the original on 29 December 2021. Retrieved 15 January 2022.
  21. Smith ME, Haney E, McDonagh M, Pappas M, Daeges M, Wasson N, Fu R, Nelson HD (June 2015). "Treatment of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: A Systematic Review for a National Institutes of Health Pathways to Prevention Workshop". Annals of Internal Medicine (Systematic review). 162 (12): 841–50. doi:10.7326/M15-0114. PMID 26075755.
  22. "Annex 1: Epidemiology of CFS/ME". UK Department of Health. 6 January 2012. Archived from the original on 6 January 2012. Retrieved 28 July 2017.
  23. "Epidemiology | Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) | CDC". www.cdc.gov. 12 July 2018. Archived from the original on 6 June 2020. Retrieved 24 May 2020.
  24. "ME/CFS in Children | Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) | CDC". www.cdc.gov. 15 May 2019. Archived from the original on 28 July 2020. Retrieved 24 May 2020. ME/CFS is often thought of as a problem in adults, but children (both adolescents and younger children) can also get ME/CFS.
  25. Boulazreg S, Rokach A (October 2020). "The Lonely, Isolating, and Alienating Implications of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome". Healthcare. 8 (4): 413. doi:10.3390/healthcare8040413. PMC 7711762. PMID 33092097.
  26. Ottati VC (2002). The social psychology of politics. New York: Kluwer Academic/Plenum. pp. 159–60. ISBN 978-0-306-46723-3. Archived from the original on 14 February 2021. Retrieved 11 August 2009.
  27. Price JR, Mitchell E, Tidy E, Hunot V (July 2008). Price JR (ed.). "Cognitive behaviour therapy for chronic fatigue syndrome in adults". The Cochrane Database of Systematic Reviews. 2021 (3): CD001027. doi:10.1002/14651858.CD001027.pub2. PMC 7028002. PMID 18646067.
  28. "Treating the Most Disruptive Symptoms First and Preventing Worsening of Symptoms | CDC". www.cdc.gov. 19 November 2019. Archived from the original on 7 August 2020. Retrieved 19 August 2020.
  29. Nijs J, Meeus M, Van Oosterwijck J, Ickmans K, Moorkens G, Hans G, De Clerck LS (February 2012). "In the mind or in the brain? Scientific evidence for central sensitisation in chronic fatigue syndrome". European Journal of Clinical Investigation. 42 (2): 203–12. doi:10.1111/j.1365-2362.2011.02575.x. PMID 21793823. S2CID 13926525.
  30. "CDC – Chronic Fatigue Syndrome (CFS) – Diagnosis". Cdc.gov. Archived from the original on 5 August 2012. Retrieved 22 July 2012.
  31. O'Boyle S, Nacul L, Nacul FE, Mudie K, Kingdon CC, Cliff JM, Clark TG, Dockrell HM, Lacerda EM (2021). "A Natural History of Disease Framework for Improving the Prevention, Management, and Research on Post-viral Fatigue Syndrome and Other Forms of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome". Frontiers in Medicine. 8: 688159. doi:10.3389/fmed.2021.688159. PMC 8835111. PMID 35155455.
  32. "Presentation and Clinical Course of ME/CFS | Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) | CDC". www.cdc.gov. 19 November 2019. Archived from the original on 28 July 2020. Retrieved 11 July 2020.
  33. Vanness JM, Snell CR, Strayer DR, Dempsey L, Stevens SR (June 2003). "Subclassifying chronic fatigue syndrome through exercise testing". Medicine and Science in Sports and Exercise. 35 (6): 908–13. doi:10.1249/01.MSS.0000069510.58763.E8. PMID 12783037. S2CID 9448181.
  34. Ho-Yen DO, McNamara I (August 1991). "General practitioners' experience of the chronic fatigue syndrome". The British Journal of General Practice. 41 (349): 324–26. PMC 1371754. PMID 1777276.
  35. Meeus M, Nijs J, Meirleir KD (May 2007). "Chronic musculoskeletal pain in patients with the chronic fatigue syndrome: a systematic review". European Journal of Pain. 11 (4): 377–86. doi:10.1016/j.ejpain.2006.06.005. PMID 16843021. S2CID 21414690.
  36. McCully KK, Sisto SA, Natelson BH (January 1996). "Use of exercise for treatment of chronic fatigue syndrome". Sports Medicine. 21 (1): 35–48. doi:10.2165/00007256-199621010-00004. PMID 8771284. S2CID 239650.
  37. Burton C, Knoop H, Popovic N, Sharpe M, Bleijenberg G (June 2009). "Reduced complexity of activity patterns in patients with chronic fatigue syndrome: a case control study". BioPsychoSocial Medicine. 3 (1): 7. doi:10.1186/1751-0759-3-7. PMC 2697171. PMID 19490619.
  38. Solomon L, Nisenbaum R, Reyes M, Papanicolaou DA, Reeves WC (October 2003). "Functional status of persons with chronic fatigue syndrome in the Wichita, Kansas, population". Health and Quality of Life Outcomes. 1 (1): 48. doi:10.1186/1477-7525-1-48. PMC 239865. PMID 14577835.
  39. Johnson H (1996). Osler's Web: Inside the Labyrinth of the Chronic Fatigue Syndrome Epidemic (1st ed.). New York: Crown Publishers. pp. 364–65. ISBN 978-0-517-70353-3. Congressional testimony of Mark, Loveless, MD, May 12, 1995
  40. "Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS)". Centers for Disease Control and Prevention. 13 April 2020. Archived from the original on 26 June 2020. Retrieved 1 June 2020.
  41. Anderson JS, Ferrans CE (June 1997). "The quality of life of persons with chronic fatigue syndrome". The Journal of Nervous and Mental Disease. 185 (6): 359–67. doi:10.1097/00005053-199706000-00001. PMID 9205421.
  42. Komaroff AL, Fagioli LR, Doolittle TH, Gandek B, Gleit MA, Guerriero RT, Kornish RJ, Ware NC, Ware JE, Bates DW (September 1996). "Health status in patients with chronic fatigue syndrome and in general population and disease comparison groups". The American Journal of Medicine. 101 (3): 281–90. doi:10.1016/S0002-9343(96)00174-X. PMID 8873490.
  43. "Chronic Fatigue Syndrome: Advancing Research and Clinical Education". Centers for Disease Control and Prevention. 28 February 2018. Archived from the original on 28 July 2020. Retrieved 26 May 2020.
  44. Ross SD, Estok RP, Frame D, Stone LR, Ludensky V, Levine CB (May 2004). "Disability and chronic fatigue syndrome: a focus on function". Archives of Internal Medicine. 164 (10): 1098–107. doi:10.1001/archinte.164.10.1098. PMID 15159267.
  45. Unger ER, Lin JS, Brimmer DJ, Lapp CW, Komaroff AL, Nath A, Laird S, Iskander J (December 2016). "CDC Grand Rounds: Chronic Fatigue Syndrome – Advancing Research and Clinical Education" (PDF). MMWR. Morbidity and Mortality Weekly Report. 65 (50–51): 1434–38. doi:10.15585/mmwr.mm655051a4. PMID 28033311. Archived (PDF) from the original on 6 January 2017. Retrieved 5 January 2017. The highest prevalence of illness is in persons aged 40–50 years...
  46. Christley Y, Duffy T, Everall IP, Martin CR (April 2013). "The neuropsychiatric and neuropsychological features of chronic fatigue syndrome: revisiting the enigma". Current Psychiatry Reports. 15 (4): 353. doi:10.1007/s11920-013-0353-8. PMID 23440559. S2CID 25790262.
  47. Cvejic E, Birch RC, Vollmer-Conna U (May 2016). "Cognitive Dysfunction in Chronic Fatigue Syndrome: a Review of Recent Evidence". Current Rheumatology Reports. Springer Science and Business Media LLC. 18 (5): 24. doi:10.1007/s11926-016-0577-9. PMID 27032787. S2CID 38748839.
  48. Rasa S, Nora-Krukle Z, Henning N, Eliassen E, Shikova E, Harrer T, Scheibenbogen C, Murovska M, Prusty BK (October 2018). "Chronic viral infections in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS)". J Transl Med. 16 (1): 268. doi:10.1186/s12967-018-1644-y. PMC 6167797. PMID 30285773.
  49. Gerwyn M, Maes M (January 2017). "Mechanisms Explaining Muscle Fatigue and Muscle Pain in Patients with Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS): a Review of Recent Findings". Current Rheumatology Reports. 19 (1): 1. doi:10.1007/s11926-017-0628-x. PMID 28116577. S2CID 11823204.
  50. "Chronic fatigue syndrome (Tapanui flu)". www.southerncross.co.nz Southern Cross NZ. March 2020. Archived from the original on 30 January 2021. Retrieved 3 February 2021.
  51. Dinos S, Khoshaba B, Ashby D, White PD, Nazroo J, Wessely S, Bhui KS (December 2009). "A systematic review of chronic fatigue, its syndromes and ethnicity: prevalence, severity, co-morbidity and coping". International Journal of Epidemiology. 38 (6): 1554–70. doi:10.1093/ije/dyp147. PMID 19349479.
  52. "Myalgic Encephalomyelitis/Chronic Fatigue Syndrome – Etiology and Pathophysiology". 10 July 2018. Archived from the original on 18 July 2018. Retrieved 18 July 2018.
  53. "Mortality and Morbidity Statistics". ICD-11. Archived from the original on 1 August 2018. Retrieved 9 January 2021.
  54. Eriksen W (16 August 2018). "ME/CFS, case definition, and serological response to Epstein–Barr virus. A systematic literature review". Fatigue: Biomedicine, Health & Behavior. 6 (4): 220–34. doi:10.1080/21641846.2018.1503125. S2CID 80898744. The levels of antibodies to EBV in ME/CFS patients differed from those in controls in 14 studies. The differences in EBV serology that were revealed, were almost exclusively signs that may indicate higher EBV activity in the patient group. The serological differences between patients and controls were seen in the two studies in which ME/CFS was defined using the Canadian criteria, in 5 of the 9 studies using the Holmes criteria, in 1 of the 2 studies using modified Holmes criteria, in 2 of the 6 studies using the Fukuda criteria, and in 4 of the 7 studies using less known criteria. The single study using the Oxford criteria showed no difference between cases and controls. Conclusions: There seems to be increased EBV activity in subset(s) of ME/CFS patients.
  55. Cleare AJ (March 2004). "The HPA axis and the genesis of chronic fatigue syndrome". Trends in Endocrinology and Metabolism. 15 (2): 55–59. doi:10.1016/j.tem.2003.12.002. PMID 15036250. S2CID 1353041.
  56. Jason LA, Porter N, Brown M, Anderson V, Brown A, Hunnell J, Lerch A (2009). "CFS: A Review of Epidemiology and Natural History Studies". Bulletin of the IACFS/ME. 17 (3): 88–106. PMC 3021257. PMID 21243091.
  57. Hulme K, Hudson JL, Rojczyk P, Little P, Moss-Morris R (August 2017). "Biopsychosocial risk factors of persistent fatigue after acute infection: A systematic review to inform interventions". Journal of Psychosomatic Research. 99: 120–129. doi:10.1016/j.jpsychores.2017.06.013. PMID 28712416. Retrieved 26 May 2020.
  58. Brurberg KG, Fønhus MS, Larun L, Flottorp S, Malterud K (February 2014). "Case definitions for chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME): a systematic review". BMJ Open. 4 (2): e003973. doi:10.1136/bmjopen-2013-003973. PMC 3918975. PMID 24508851.
  59. "ICD-11 – Mortality and Morbidity Statistics". icd.who.int. Archived from the original on 1 August 2018. Retrieved 20 May 2020. Diseases of the nervous system
  60. Maksoud R, du Preez S, Eaton-Fitch N, Thapaliya K, Barnden L, Cabanas H, Staines D, Marshall-Gradisnik S (2020). "A systematic review of neurological impairments in myalgic encephalomyelitis/ chronic fatigue syndrome using neuroimaging techniques". PLOS ONE. 15 (4): e0232475. Bibcode:2020PLoSO..1532475M. doi:10.1371/journal.pone.0232475. PMC 7192498. PMID 32353033.
  61. Jason LA, Zinn ML, Zinn MA (2 February 2017). "Myalgic Encephalomyelitis: Symptoms and Biomarkers". Current Neuropharmacology. 13 (5): 701–34. doi:10.2174/1570159X13666150928105725. PMC 4761639. PMID 26411464. Decreased frontal grey matter
  62. Martínez-Martínez LA, Mora T, Vargas A, Fuentes-Iniestra M, Martínez-Lavín M (April 2014). "Sympathetic nervous system dysfunction in fibromyalgia, chronic fatigue syndrome, irritable bowel syndrome, and interstitial cystitis: a review of case-control studies". Journal of Clinical Rheumatology. 20 (3): 146–50. doi:10.1097/RHU.0000000000000089. PMID 24662556. S2CID 23799955.
  63. Jackson ML, Bruck D (December 2012). "Sleep abnormalities in chronic fatigue syndrome/myalgic encephalomyelitis: a review". Journal of Clinical Sleep Medicine. 8 (6): 719–28. doi:10.5664/jcsm.2276. PMC 3501671. PMID 23243408.
  64. Tanaka M, Tajima S, Mizuno K, Ishii A, Konishi Y, Miike T, Watanabe Y (November 2015). "Frontier studies on fatigue, autonomic nerve dysfunction, and sleep-rhythm disorder". The Journal of Physiological Sciences. 65 (6): 483–98. doi:10.1007/s12576-015-0399-y. PMC 4621713. PMID 26420687.
  65. Van Cauwenbergh D, Nijs J, Kos D, Van Weijnen L, Struyf F, Meeus M (May 2014). "Malfunctioning of the autonomic nervous system in patients with chronic fatigue syndrome: a systematic literature review". European Journal of Clinical Investigation. 44 (5): 516–26. doi:10.1111/eci.12256. PMID 24601948. S2CID 9722415.
  66. Lapp CW (16 February 2016). "Chronic Fatigue Syndrome: Advancing Research and Clinical Education" (PDF). CDC Public Health Grand Rounds. Centers for Disease Control and Prevention. Archived (PDF) from the original on 18 October 2020. Retrieved 26 May 2020.
  67. Nijs J, Nees A, Paul L, De Kooning M, Ickmans K, Meeus M, Van Oosterwijck J (2014). "Altered immune response to exercise in patients with chronic fatigue syndrome/myalgic encephalomyelitis: a systematic literature review". Exercise Immunology Review. 20: 94–116. PMID 24974723.
  68. Armstrong CW, McGregor NR, Butt HL, Gooley PR (2014). "Metabolism in chronic fatigue syndrome". Advances in Clinical Chemistry. 66: 121–172. doi:10.1016/B978-0-12-801401-1.00005-0. ISBN 978-0-12-801401-1. PMID 25344988.
  69. Morris G, Anderson G, Galecki P, Berk M, Maes M (March 2013). "A narrative review on the similarities and dissimilarities between myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) and sickness behavior". BMC Medicine. 11: 64. doi:10.1186/1741-7015-11-64. PMC 3751187. PMID 23497361.
  70. Griffith JP, Zarrouf FA (2008). "A systematic review of chronic fatigue syndrome: don't assume it's depression". Primary Care Companion to the Journal of Clinical Psychiatry. 10 (2): 120–128. doi:10.4088/pcc.v10n0206. PMC 2292451. PMID 18458765.
  71. Meeus M, Mistiaen W, Lambrecht L, Nijs J (November 2009). "Immunological similarities between cancer and chronic fatigue syndrome: the common link to fatigue?". Anticancer Research. 29 (11): 4717–4726. PMID 20032425.
  72. Silverman MN, Heim CM, Nater UM, Marques AH, Sternberg EM (May 2010). "Neuroendocrine and immune contributors to fatigue". PM & R. 2 (5): 338–46. doi:10.1016/j.pmrj.2010.04.008. PMC 2933136. PMID 20656615.
  73. Morris G, Anderson G, Maes M (November 2017). "Hypothalamic-Pituitary-Adrenal Hypofunction in Myalgic Encephalomyelitis (ME)/Chronic Fatigue Syndrome (CFS) as a Consequence of Activated Immune-Inflammatory and Oxidative and Nitrosative Pathways". Molecular Neurobiology. 54 (9): 6806–19. doi:10.1007/s12035-016-0170-2. PMID 27766535. S2CID 3524276.
  74. Cho HJ, Skowera A, Cleare A, Wessely S (January 2006). "Chronic fatigue syndrome: an update focusing on phenomenology and pathophysiology". Current Opinion in Psychiatry. 19 (1): 67–73. doi:10.1097/01.yco.0000194370.40062.b0. PMID 16612182. S2CID 12815707.
  75. Papadopoulos AS, Cleare AJ (September 2011). "Hypothalamic-pituitary-adrenal axis dysfunction in chronic fatigue syndrome". Nature Reviews. Endocrinology. 8 (1): 22–32. doi:10.1038/nrendo.2011.153. PMID 21946893. S2CID 22176725.
  76. Tak LM, Cleare AJ, Ormel J, Manoharan A, Kok IC, Wessely S, Rosmalen JG (May 2011). "Meta-analysis and meta-regression of hypothalamic-pituitary-adrenal axis activity in functional somatic disorders". Biological Psychology. 87 (2): 183–94. doi:10.1016/j.biopsycho.2011.02.002. PMID 21315796. S2CID 206108463.
  77. Van Den Eede F, Moorkens G, Van Houdenhove B, Cosyns P, Claes SJ (2007). "Hypothalamic-pituitary-adrenal axis function in chronic fatigue syndrome". Neuropsychobiology. 55 (2): 112–20. CiteSeerX 10.1.1.626.9632. doi:10.1159/000104468. PMID 17596739. S2CID 14956850.
  78. Powell DJ, Liossi C, Moss-Morris R, Schlotz W (November 2013). "Unstimulated cortisol secretory activity in everyday life and its relationship with fatigue and chronic fatigue syndrome: a systematic review and subset meta-analysis". Psychoneuroendocrinology. 38 (11): 2405–22. doi:10.1016/j.psyneuen.2013.07.004. PMID 23916911.
  79. Morris G, Berk M, Galecki P, Maes M (April 2014). "The emerging role of autoimmunity in myalgic encephalomyelitis/chronic fatigue syndrome (ME/cfs)". Molecular Neurobiology. 49 (2): 741–756. doi:10.1007/s12035-013-8553-0. hdl:11343/219795. PMID 24068616. S2CID 13185036.
  80. Loebel M, Grabowski P, Heidecke H, Bauer S, Hanitsch LG, Wittke K, et al. (February 2016). "Antibodies to β adrenergic and muscarinic cholinergic receptors in patients with Chronic Fatigue Syndrome". Brain, Behavior, and Immunity. 52: 32–39. doi:10.1016/j.bbi.2015.09.013. PMID 26399744.
  81. Sotzny F, Blanco J, Capelli E, Castro-Marrero J, Steiner S, Murovska M, Scheibenbogen C (June 2018). "Myalgic Encephalomyelitis/Chronic Fatigue Syndrome - Evidence for an autoimmune disease". Autoimmunity Reviews. 17 (6): 601–609. doi:10.1016/j.autrev.2018.01.009. PMID 29635081.
  82. Wirth K, Scheibenbogen C (June 2020). "A Unifying Hypothesis of the Pathophysiology of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS): Recognitions from the finding of autoantibodies against ß2-adrenergic receptors". Autoimmunity Reviews. 19 (6): 102527. doi:10.1016/j.autrev.2020.102527. PMID 32247028.
  83. Marshall-Gradisnik S, Smith P, Nilius B, Staines DR (1 January 2015). "Examination of Single Nucleotide Polymorphisms in Acetylcholine Receptors in Chronic Fatigue Syndrome Patients". Immunology and Immunogenetics Insights. 7: III.S25105. doi:10.4137/III.S25105. ISSN 1178-6345.
  84. Holden S, Maksoud R, Eaton-Fitch N, Cabanas H, Staines D, Marshall-Gradisnik S (July 2020). "A systematic review of mitochondrial abnormalities in myalgic encephalomyelitis/chronic fatigue syndrome/systemic exertion intolerance disease". Journal of Translational Medicine. 18 (1): 290. doi:10.1186/s12967-020-02452-3. PMC 7392668. PMID 32727475.
  85. Reeves WC, Lloyd A, Vernon SD, Klimas N, Jason LA, Bleijenberg G, Evengard B, White PD, Nisenbaum R, Unger ER (December 2003). "Identification of ambiguities in the 1994 chronic fatigue syndrome research case definition and recommendations for resolution". BMC Health Services Research. 3 (1): 25. doi:10.1186/1472-6963-3-25. PMC 317472. PMID 14702202.
  86. "Diagnosis of ME/CFS | Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) | CDC". 15 May 2019. Archived from the original on 28 July 2020. Retrieved 23 May 2020.
  87. Bansal AS (July 2016). "Investigating unexplained fatigue in general practice with a particular focus on CFS/ME". BMC Family Practice. 17 (81): 81. doi:10.1186/s12875-016-0493-0. PMC 4950776. PMID 27436349.
  88. Institute of Medicine (2015). "Beyond Myalgic Encephalomyelitis/Chronic Fatigue Syndrome - Redefining an Illness. Report for Clinicians" (PDF). Institute of Medicine. Archived (PDF) from the original on 8 March 2022. Retrieved 26 March 2022.
  89. Wyller VB (2007). "The chronic fatigue syndrome – an update". Acta Neurologica Scandinavica. Supplementum. 187: 7–14. doi:10.1111/j.1600-0404.2007.00840.x. PMID 17419822. S2CID 11247547.
  90. Fukuda K, Straus SE, Hickie I, Sharpe MC, Dobbins JG, Komaroff A (December 1994). "The chronic fatigue syndrome: a comprehensive approach to its definition and study. International Chronic Fatigue Syndrome Study Group". Annals of Internal Medicine. 121 (12): 953–59. doi:10.7326/0003-4819-121-12-199412150-00009. PMID 7978722. S2CID 510735.
  91. Holmes GP, Kaplan JE, Gantz NM, Komaroff AL, Schonberger LB, Straus SE, Jones JF, Dubois RE, Cunningham-Rundles C, Pahwa S (March 1988). "Chronic fatigue syndrome: a working case definition". Annals of Internal Medicine. 108 (3): 387–89. doi:10.7326/0003-4819-108-3-387. PMID 2829679.
  92. Guideline 53: Chronic fatigue syndrome/myalgic encephalomyelitis (or encephalopathy). London: National Institute for Health and Clinical Excellence. 2007. ISBN 978-1-84629-453-2. Archived from the original on 1 June 2014. Retrieved 3 September 2007.
  93. Carruthers BM, Jain AK, De Meirleir KL, Peterson DL, Klimas NG, Lerner AM, et al. (4 December 2011). "Myalgic Encephalomyelitis/Chronic Fatigue Syndrome". Journal of Chronic Fatigue Syndrome. 11 (1): 7–115. doi:10.1300/J092v11n01_02.
  94. Carruthers BM, van de Sande MI, De Meirleir KL, Klimas NG, Broderick G, Mitchell T, et al. (October 2011). "Myalgic encephalomyelitis: International Consensus Criteria". Journal of Internal Medicine. 270 (4): 327–38. doi:10.1111/j.1365-2796.2011.02428.x. PMC 3427890. PMID 21777306.
  95. "Recommendations - Myalgic encephalomyelitis (or encephalopathy)/chronic fatigue syndrome: diagnosis and management - Guidance". NICE. 29 October 2021. Archived from the original on 29 December 2021. Retrieved 15 January 2022.
  96. "Myalgic Encephalomyelitis ("Chronic Fatigue Syndrome")". NY DOH diseases and conditions. April 2018. Archived from the original on 18 April 2018. Retrieved 21 April 2018.
  97. Craig T, Kakumanu S (March 2002). "Chronic fatigue syndrome: evaluation and treatment". American Family Physician. 65 (6): 1083–90. PMID 11925084. Archived from the original on 26 September 2007. Retrieved 25 January 2008.
  98. Logan AC, Wong C (October 2001). "Chronic fatigue syndrome: oxidative stress and dietary modifications" (PDF). Alternative Medicine Review. 6 (5): 450–59. PMID 11703165. Archived from the original (PDF) on 7 November 2016. Retrieved 12 November 2015. Finally, recent evidence suggests celiac disease can present with neurological symptoms in the absence of gastrointestinal symptoms; therefore, celiac disease should be included in the differential diagnosis of CFS.
  99. Hakim A, De Wandele I, O'Callaghan C, Pocinki A, Rowe P (March 2017). "Chronic fatigue in Ehlers-Danlos syndrome-Hypermobile type". American Journal of Medical Genetics. Part C, Seminars in Medical Genetics. 175 (1): 175–180. doi:10.1002/ajmg.c.31542. PMID 28186393. Archived from the original on 11 December 2019. Retrieved 11 December 2019.
  100. Bradley LA, McKendree-Smith NL, Alarcón GS (2000). "Pain complaints in patients with fibromyalgia versus chronic fatigue syndrome". Current Review of Pain. 4 (2): 148–57. doi:10.1007/s11916-000-0050-2. PMID 10998728. S2CID 2555977.
  101. Smith ME, Nelson HD, Haney E, Pappas M, Daeges M, Wasson N, McDonagh M (December 2014). "Diagnosis and Treatment of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome". Evidence Report/Technology Assessment (Evidence Reports/Technology Assessments, No. 219 ed.). Agency for Healthcare Research and Quality (US) (219): 1–433. doi:10.23970/AHRQEPCERTA219. PMID 30313001. Archived from the original on 1 February 2016. Retrieved 22 January 2016.
  102. Smith ME, Nelson HD, Haney E, Pappas M, Daeges M, Wasson N, McDonagh M (December 2014). "Diagnosis and Treatment of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome". Evidence Report/Technology Assessment (Evidence Reports/Technology Assessments, No. 219 ed.). Agency for Healthcare Research and Quality (US) (219): 1–433. doi:10.23970/AHRQEPCERTA219. PMID 30313001. Archived from the original on 1 February 2016. Retrieved 22 January 2016.
  103. Vink M, Vink-Niese A (1 July 2018). "Graded exercise therapy for myalgic encephalomyelitis/chronic fatigue syndrome is not effective and unsafe. Re-analysis of a Cochrane review". Health Psychology Open. 5 (2): 2055102918805187. doi:10.1177/2055102918805187. PMC 6176540. PMID 30305916. The analysis of the 2017 Cochrane review reveals flaws, which means that contrary to its findings, there is no evidence that graded exercise therapy is effective. Because of the failure to report harms adequately in the trials covered by the review, it cannot be said that graded exercise therapy is safe. The analysis of the objective outcomes in the trials provides sufficient evidence to conclude that graded exercise therapy is an ineffective treatment for myalgic encephalomyelitis/chronic fatigue syndrome... The analysis of the 2017 Cochrane review reveals flaws, which means that contrary to its findings, there is no evidence that graded exercise therapy is effective. Because of the failure to report harms adequately in the trials covered by the review, it cannot be said that graded exercise therapy is safe. The analysis of the objective outcomes in the trials provides sufficient evidence to conclude that graded exercise therapy is an ineffective treatment for myalgic encephalomyelitis/chronic fatigue syndrome.
  104. Smith ME, Nelson HD, Haney E, Pappas M, Daeges M, Wasson N, McDonagh M (July 2016). "July 2016 Addendum. In Diagnosis and Treatment of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome 2014 Dec". Evidence Report/Technology Assessment. Agency for Healthcare Research and Quality (US): 1–433. Archived from the original on 1 November 2020. Retrieved 15 April 2017. The results are consistent across trials with improvement in function, fatigue, and global improvement and provided moderate strength of evidence for improved function (4 trials, n=607) and global improvement (3 trials, n=539), low strength of evidence for reduced fatigue (4 trials, n=607) and decreased work impairment (1 trial, n=480), and insufficient evidence for improved quality of life (no trials)
  105. "Outrage at chronic fatigue syndrome advice update pause". BBC News. 17 August 2021. Archived from the original on 17 August 2021. Retrieved 18 August 2021.
  106. Grover N (17 August 2021). "UK health standards body delays new ME guidance in therapy row". The Guardian. Archived from the original on 19 August 2021. Retrieved 19 August 2021.
  107. "Comorbid Conditions | Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) | CDC". www.cdc.gov. 12 July 2018. Archived from the original on 7 June 2020. Retrieved 29 May 2020.
  108. Goudsmit EM, Nijs J, Jason LA, Wallman KE (19 December 2011). "Pacing as a strategy to improve energy management in myalgic encephalomyelitis/chronic fatigue syndrome: a consensus document". Disability and Rehabilitation. 34 (13): 1140–47. doi:10.3109/09638288.2011.635746. PMID 22181560. S2CID 22457926. Archived from the original on 28 July 2020. Retrieved 23 May 2020.
  109. Nielson WR, Jensen MP, Karsdorp PA, Vlaeyen JW (May 2013). "Activity pacing in chronic pain: concepts, evidence, and future directions". The Clinical Journal of Pain. 29 (5): 461–68. doi:10.1097/AJP.0b013e3182608561. PMID 23247005. S2CID 28709499.
  110. "ME Association Summary Review: Assessing PEM (Post-exertional Malaise)" (PDF). ME Association. 2019. Archived (PDF) from the original on 28 July 2020. Retrieved 23 May 2020.
  111. Jason LA, Melrose H, Lerman A, Burroughs V, Lewis K, King CP, Frankenberry EL (January 1999). "Managing chronic fatigue syndrome: overview and case study". AAOHN Journal. 47 (1): 17–21. doi:10.1177/216507999904700104. PMID 10205371.
  112. Jason LA, Brown M, Brown A, Evans M, Flores S, Grant-Holler E, Sunnquist M (January 2013). "Energy Conservation/Envelope Theory Interventions to Help Patients with Myalgic Encephalomyelitis/Chronic Fatigue Syndrome". Fatigue. 1 (1–2): 27–42. doi:10.1080/21641846.2012.733602. PMC 3596172. PMID 23504301.
  113. O'connor K, Sunnquist M, Nicholson L, Jason LA, Newton JL, Strand EB (March 2019). "Energy envelope maintenance among patients with myalgic encephalomyelitis and chronic fatigue syndrome: Implications of limited energy reserves". Chronic Illness. 15 (1): 51–60. doi:10.1177/1742395317746470. PMC 5750135. PMID 29231037.
  114. Jason L, Muldowney K, Torres-Harding S (May 2008). "The Energy Envelope Theory and myalgic encephalomyelitis/chronic fatigue syndrome". AAOHN Journal. 56 (5): 189–95. doi:10.3928/08910162-20080501-06. PMID 18578185. S2CID 25558691.
  115. Brown M, Khorana N, Jason LA (March 2011). "The role of changes in activity as a function of perceived available and expended energy in nonpharmacological treatment outcomes for ME/CFS". Journal of Clinical Psychology. 67 (3): 253–60. doi:10.1002/jclp.20744. PMC 3164291. PMID 21254053.
  116. Campbell B (Winter 2009). "Managing your energy envelope" (PDF). The CFIDS Chronicle: 28–31. Archived (PDF) from the original on 27 September 2020. Retrieved 23 May 2020.
  117. Emerge Australia (November 2019). "Pacing". Archived from the original on 17 July 2022.
  118. Steefel L (2011). What Nurses Know...Chronic Fatigue Syndrome. Demos Medical Publishing. pp. 54–55. ISBN 978-1-61705-028-2. Archived from the original on 31 May 2021. Retrieved 9 November 2020.
  119. Campbell B (14 November 2009). "Pacing by Numbers: using your heart rate to stay inside the energy envelope". ME/CFS South Australia Inc. Archived from the original on 25 March 2020. Retrieved 23 May 2020.
  120. Castro-Marrero J, Sáez-Francàs N, Santillo D, Alegre J (March 2017). "Treatment and management of chronic fatigue syndrome/myalgic encephalomyelitis: all roads lead to Rome". Br. J. Pharmacol. 174 (5): 345–69. doi:10.1111/bph.13702. PMC 5301046. PMID 28052319.
  121. Green CR, Cowan P, Elk R, O'Neil KM, Rasmussen AL (June 2015). "National Institutes of Health Pathways to Prevention Workshop: Advancing the Research on Myalgic Encephalomyelitis/Chronic Fatigue Syndrome". Annals of Internal Medicine. 162 (12): 860–65. doi:10.7326/M15-0338. PMID 26075757.
  122. Adamowicz JL, Caikauskaite I, Friedberg F (November 2014). "Defining recovery in chronic fatigue syndrome: a critical review". Quality of Life Research. 23 (9): 2407–16. doi:10.1007/s11136-014-0705-9. PMID 24791749. S2CID 13609292.
  123. Shepherd C (February 2016). "Patient reaction to the PACE trial". The Lancet. Psychiatry. 3 (2): e7–8. doi:10.1016/S2215-0366(15)00546-5. PMID 26795759.
  124. Clark C, Buchwald D, MacIntyre A, Sharpe M, Wessely S (January 2002). "Chronic fatigue syndrome: a step towards agreement". Lancet. 359 (9301): 97–98. doi:10.1016/S0140-6736(02)07336-1. PMID 11809249. S2CID 38526912.
  125. The ME Association. "No decisions about me without me" (PDF). ME Association. The ME Association. Archived (PDF) from the original on 6 November 2015. Retrieved 20 January 2016.
  126. "Forward-ME and Oxford Brookes University announces results of patient survey on CBT and GET in ME/CFS". ME Association. April 2019. Archived from the original on 9 May 2020. Retrieved 26 May 2020.
  127. Larun L, Brurberg KG, Odgaard-Jensen J, Price JR (October 2019). "Exercise therapy for chronic fatigue syndrome". The Cochrane Database of Systematic Reviews. 10 (3): CD003200. doi:10.1002/14651858.CD003200.pub8. PMC 6953363. PMID 31577366.
  128. Elgot J (18 October 2015), "Chronic fatigue patients criticise study that says exercise can help", The Guardian, archived from the original on 21 June 2018, retrieved 20 June 2018
  129. Jason LA (August 2017). "The PACE trial missteps on pacing and patient selection". Journal of Health Psychology. 22 (9): 1141–45. doi:10.1177/1359105317695801. PMID 28805518.
  130. Cox D, Ludlam S, Mason L, Wagner S, Sharpe M, et al. (PACE Trial Management Group) (November 2004). "Manual for Participants Adaptive Pacing Therapy (APT) for CFS/ME" (PDF). Wolfson Institute | Queen Mary University of London. Archived (PDF) from the original on 28 July 2020. Retrieved 23 May 2020.
  131. White PD, Goldsmith KA, Johnson AL, Potts L, Walwyn R, DeCesare JC, et al. (March 2011). "Comparison of adaptive pacing therapy, cognitive behaviour therapy, graded exercise therapy, and specialist medical care for chronic fatigue syndrome (PACE): a randomised trial". Lancet. 377 (9768): 823–36. doi:10.1016/S0140-6736(11)60096-2. PMC 3065633. PMID 21334061.
  132. Wormgoor ME, Rodenburg SC (January 2021). "The evidence base for physiotherapy in myalgic encephalomyelitis/chronic fatigue syndrome when considering post-exertional malaise: a systematic review and narrative synthesis". Journal of Translational Medicine. 19 (1): 1. doi:10.1186/s12967-020-02683-4. PMC 7780213. PMID 33397399.
  133. "Pacing for people with ME" (PDF). Action for ME. 2013. Archived from the original (PDF) on 5 November 2018.
  134. Richman S, Morris MC, Broderick G, Craddock TJ, Klimas NG, Fletcher MA (May 2019). "Pharmaceutical Interventions in Chronic Fatigue Syndrome: A Literature-based Commentary". Clin Ther. 41 (5): 798–805. doi:10.1016/j.clinthera.2019.02.011. PMC 6543846. PMID 30871727.
  135. Center for Drug Evaluation and Research. "FDA Response Letter Regarding Approval of Ampligen for ME/CFS". www.fda.gov. Archived from the original on 23 October 2016. Retrieved 12 June 2018.
  136. Barclay L (5 February 2013). "FDA Nixes Rintatolimod for Chronic Fatigue Syndrome". Medscape. Archived from the original on 20 June 2015. Retrieved 18 January 2017.
  137. Agrawal S, Kandimalla ER (February 2019). "Chapter 14: Synthetic agonists of Toll-like receptors and therapeutic applications.". In Agrawal S, Gait MJ (eds.). Advances in Nucleic Acid Therapeutics. pp. 306–338 (310). ISBN 978-1-78801-732-9. Archived from the original on 14 May 2022. Retrieved 20 October 2021. 14.2: Agonists of TLR3
  138. "Rintatolimod for severe Chronic Fatigue Syndrome". fda.gov. 19 September 2019. Archived from the original on 29 June 2020. Retrieved 26 May 2020.
  139. "AIM ImmunoTech reports positive Phase III Ampligen data in long Covid". 29 July 2022.
  140. "Prognosis | Presentation and Clinical Course | Healthcare Providers | Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) | CDC". www.cdc.gov. 23 July 2019. Retrieved 15 July 2022.
  141. "Recommendations | Myalgic encephalomyelitis (or encephalopathy)/chronic fatigue syndrome: diagnosis and management | Guidance | NICE". www.nice.org.uk. Retrieved 15 July 2022.
  142. Cairns R, Hotopf M (January 2005). "A systematic review describing the prognosis of chronic fatigue syndrome". Occupational Medicine. 55 (1): 20–31. doi:10.1093/occmed/kqi013. PMID 15699087.
  143. Joyce J, Hotopf M, Wessely S (March 1997). "The prognosis of chronic fatigue and chronic fatigue syndrome: a systematic review". QJM. 90 (3): 223–33. doi:10.1093/qjmed/90.3.223. PMID 9093600.
  144. Collard SS, Murphy J (September 2020). "Management of chronic fatigue syndrome/myalgic encephalomyelitis in a pediatric population: A scoping review". Journal of Child Health Care. 24 (3): 411–431. doi:10.1177/1367493519864747. PMC 7863118. PMID 31379194.
  145. Bakken IJ, Tveito K, Gunnes N, Ghaderi S, Stoltenberg C, Trogstad L, et al. (October 2014). "Two age peaks in the incidence of chronic fatigue syndrome/myalgic encephalomyelitis: a population-based registry study from Norway 2008-2012". BMC Medicine. 12: 167. doi:10.1186/s12916-014-0167-5. PMC 4189623. PMID 25274261.
  146. "Epidemiology | Presentation and Clinical Course | Healthcare Providers | Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) | CDC". www.cdc.gov. 27 April 2021. Retrieved 16 July 2022.
  147. Acheson ED (April 1959). "The clinical syndrome variously called benign myalgic encephalomyelitis, Iceland disease and epidemic neuromyasthenia". The American Journal of Medicine. 26 (4): 569–95. CiteSeerX 10.1.1.534.4761. doi:10.1016/0002-9343(59)90280-3. PMID 13637100.
  148. Parish JG (November 1978). "Early outbreaks of 'epidemic neuromyasthenia'". Postgraduate Medical Journal. 54 (637): 711–17. doi:10.1136/pgmj.54.637.711. PMC 2425322. PMID 370810.
  149. Wojcik W, Armstrong D, Kanaan R (June 2011). "Chronic fatigue syndrome: labels, meanings and consequences". Journal of Psychosomatic Research. 70 (6): 500–04. doi:10.1016/j.jpsychores.2011.02.002. PMID 21624573.
  150. Lancet. Public health (1955). "Outbreak at the royal free". The Lancet. 266 (6885): 351–52. doi:10.1016/s0140-6736(55)92344-8.
  151. Price JL (April 1961). "Myalgic encephalomyelitis". Lancet. 1 (7180): 737–38. doi:10.1016/s0140-6736(61)92893-8. PMC 1836797. PMID 13737972.
  152. International Classification of Diseases. Vol. I. World Health Organization. 1969. pp. 158, (vol 2, pp. 173).
  153. Ramsay AM (1988). Myalgic encephalomyelitis and postviral fatigue states (Second ed.).
  154. Ramsey AM, David AS, Wessely S, Pelosi AJ, Dowsett EG (July 1988). "Myalgic encephalomyelitis, or what?". Lancet. 2 (8602): 100–101. doi:10.1016/s0140-6736(88)90028-1. PMID 2898668. S2CID 24860444.
  155. Ramsay AM, Dowsett EG, Dadswell JV, Lyle WH, Parish JG (May 1977). "Icelandic disease (benign myalgic encephalomyelitis or Royal Free disease)". British Medical Journal. 1 (6072): 1350. doi:10.1136/bmj.1.6072.1350-a. PMC 1607215. PMID 861618.
  156. Straus SE (1991). "History of chronic fatigue syndrome". Reviews of Infectious Diseases. 13 (Suppl. 1): S2–7. doi:10.1093/clinids/13.supplement_1.s2. PMID 2020800.
  157. "Press Briefing Transcripts". Centers for Disease Control and Prevention. 3 November 2006. Archived from the original on 15 October 2013. Retrieved 12 October 2013.
  158. Shelokov A, Habel K, Verder E, Welsh W (August 1957). "Epidemic neuromyasthenia; an outbreak of poliomyelitislike illness in student nurses". The New England Journal of Medicine. 257 (8): 345–55. doi:10.1056/NEJM195708222570801. PMID 13464938.
  159. Blattner RJ (October 1956). "Benign myalgic encephalomyelitis (Akureyri disease, Iceland disease)". The Journal of Pediatrics. 49 (4): 504–06. doi:10.1016/S0022-3476(56)80241-2. PMID 13358047.
  160. Straus SE, ed. (1994). Chronic Fatigue Syndrome. New York, Basel, Hong Kong: Marcel Dekker Inc. p. 227. ISBN 978-0-8247-9187-2.
  161. Aoki T, Usuda Y, Miyakoshi H, Tamura K, Herberman RB (1987). "Low natural killer syndrome: clinical and immunologic features". Natural Immunity and Cell Growth Regulation. 6 (3): 116–28. PMID 2442602.
  162. Wessely S (October 1991). "History of postviral fatigue syndrome". British Medical Bulletin. 47 (4): 919–41. doi:10.1093/oxfordjournals.bmb.a072521. PMID 1794091. S2CID 12964461.
  163. Ramsay AM (1986). Postviral Fatigue Syndrome. The saga of Royal Free disease. London: Gower. ISBN 978-0-906923-96-2.
  164. Simpson LO (October 1991). "Myalgic encephalomyelitis". Journal of the Royal Society of Medicine. 84 (10): 633. PMC 1295578. PMID 1744860.
  165. Jason LA, Richman JA (2008). "How science can stigmatize: The case of chronic fatigue syndrome". Journal of Chronic Fatigue Syndrome. 14 (4): 85–103. doi:10.1080/10573320802092146.
  166. Jason LA, Holbert C, Torres-Harding S, Taylor RR (2004). "Stigma and the term chronic fatigue syndrome". Journal of Disability Policy Studies. 14 (4): 222–28. CiteSeerX 10.1.1.486.4577. doi:10.1177/10442073040140040401. S2CID 72397898.
  167. Evengård B, Schacterle RS, Komaroff AL (November 1999). "Chronic fatigue syndrome: new insights and old ignorance". Journal of Internal Medicine. 246 (5): 455–69. doi:10.1046/j.1365-2796.1999.00513.x. PMID 10583715. S2CID 34123925.
  168. Chronic fatigue syndrome; Report of a joint working group of the Royal Colleges of Physicians, Psychiatrists and General Practitioners. London: Royal Colleges of Physicians, Psychiatrists and General Practitioners. 1996. ISBN 978-1-86016-046-2.
  169. Tuller D (10 February 2015). "Chronic Fatigue Syndrome Gets a New Name". well.blogs.nytimes.com. Archived from the original on 11 February 2021. Retrieved 3 February 2021.
  170. "NIH PEM subgroup CDE draft recommendations" (PDF). NIH Common Data Elements project. December 2017. Archived (PDF) from the original on 3 February 2018. Retrieved 21 April 2018.
  171. Reynolds KJ, Vernon SD, Bouchery E, Reeves WC (June 2004). "The economic impact of chronic fatigue syndrome". Cost Effectiveness and Resource Allocation. 2 (1): 4. doi:10.1186/1478-7547-2-4. PMC 449736. PMID 15210053.
  172. Jason LA, Corradi K, Torres-Harding S, Taylor RR, King C (March 2005). "Chronic fatigue syndrome: the need for subtypes". Neuropsychology Review. 15 (1): 29–58. doi:10.1007/s11065-005-3588-2. PMID 15929497. S2CID 8153255.
  173. Avellaneda Fernández A, Pérez Martín A, Izquierdo Martínez M, Arruti Bustillo M, Barbado Hernández FJ, de la Cruz Labrado J, Díaz-Delgado Peñas R, Gutiérrez Rivas E, Palacín Delgado C, Rivera Redondo J, Ramón Giménez JR (October 2009). "Chronic fatigue syndrome: aetiology, diagnosis and treatment". BMC Psychiatry. 9: S1. doi:10.1186/1471-244X-9-S1-S1. PMC 2766938. PMID 19857242.
  174. Jason LA, Benton MC, Valentine L, Johnson A, Torres-Harding S (April 2008). "The economic impact of ME/CFS: individual and societal costs". Dynamic Medicine. 7: 6. doi:10.1186/1476-5918-7-6. PMC 2324078. PMID 18397528.
  175. Broderick G, Craddock TJ (March 2013). "Systems biology of complex symptom profiles: capturing interactivity across behavior, brain and immune regulation". Brain, Behavior, and Immunity. 29: 1–8. doi:10.1016/j.bbi.2012.09.008. PMC 3554865. PMID 23022717.
  176. "ME/Chronic Fatigue Syndrome Awareness Day". Centers for Disease Control and Prevention. 12 May 2017. Archived from the original on 29 July 2017. Retrieved 12 July 2017.
  177. Lee N. "Dr. Nancy Lee on International Myalgic Encephalomyelitis/Chronic Fatigue Syndrome Awareness Day". US Department of Health & Human Services. Archived from the original on 8 July 2012. Retrieved 12 October 2013.
  178. Young DA (23 December 1995). "Florence Nightingale's fever". BMJ. 311 (7021): 1697–700. doi:10.1136/bmj.311.7021.1697. PMC 2539100. PMID 8541764.
  179. Feilden T (29 July 2011). "'Torrent of abuse' hindering ME research". BBC News. BBC. Archived from the original on 31 July 2011. Retrieved 31 July 2011.
  180. Wallace PG (October 1991). "Post-viral fatigue syndrome. Epidemiology: a critical review". British Medical Bulletin. 47 (4): 942–51. doi:10.1093/oxfordjournals.bmb.a072522. PMID 1794092.
  181. Mounstephen A, Sharpe M (May 1997). "Chronic fatigue syndrome and occupational health". Occupational Medicine. 47 (4): 217–27. doi:10.1093/occmed/47.4.217. PMID 9231495.
  182. Hooge J (1992). "Chronic fatigue syndrome: cause, controversy and care". British Journal of Nursing. 1 (9): 440–41, 443, 445–46. doi:10.12968/bjon.1992.1.9.440. PMID 1446147.
  183. Sharpe M (September 1996). "Chronic fatigue syndrome". The Psychiatric Clinics of North America. 19 (3): 549–73. doi:10.1016/S0193-953X(05)70305-1. PMID 8856816.
  184. Denz-Penhey H, Murdoch JC (April 1993). "General practitioners acceptance of the validity of chronic fatigue syndrome as a diagnosis". The New Zealand Medical Journal. 106 (953): 122–24. PMID 8474729.
  185. Greenlee JE, Rose JW (2000). "Controversies in neurological infectious diseases". Seminars in Neurology. 20 (3): 375–86. doi:10.1055/s-2000-9429. PMID 11051301.
  186. Horton-Salway M (December 2007). "The ME Bandwagon and other labels: constructing the genuine case in talk about a controversial illness" (PDF). The British Journal of Social Psychology (Submitted manuscript). 46 (Pt 4): 895–914. doi:10.1348/014466607X173456. PMID 17535450. Archived (PDF) from the original on 19 July 2018. Retrieved 7 November 2018.
  187. Bowen J, Pheby D, Charlett A, McNulty C (August 2005). "Chronic Fatigue Syndrome: a survey of GPs' attitudes and knowledge". Family Practice. 22 (4): 389–93. doi:10.1093/fampra/cmi019. PMID 15805128.
  188. Larun L, Malterud K (December 2007). "Identity and coping experiences in Chronic Fatigue Syndrome: a synthesis of qualitative studies". Patient Education and Counseling. 69 (1–3): 20–28. doi:10.1016/j.pec.2007.06.008. hdl:1956/5105. PMID 17698311.
  189. Dumit J (February 2006). "Illnesses you have to fight to get: facts as forces in uncertain, emergent illnesses". Social Science & Medicine. 62 (3): 577–90. doi:10.1016/j.socscimed.2005.06.018. PMID 16085344.
  190. Lombardi VC, Ruscetti FW, Das Gupta J, Pfost MA, Hagen KS, Peterson DL, Ruscetti SK, Bagni RK, Petrow-Sadowski C, Gold B, Dean M, Silverman RH, Mikovits JA (October 2009). "Detection of an infectious retrovirus, XMRV, in blood cells of patients with chronic fatigue syndrome". Science. 326 (5952): 585–89. Bibcode:2009Sci...326..585L. doi:10.1126/science.1179052. PMC 3073172. PMID 19815723. (Retracted, see doi:10.1126/science.334.6063.1636-a)
  191. "No blood from chronic fatigue donors: agency". CBC. 7 April 2010. Archived from the original on 11 April 2010. Retrieved 25 June 2010.
  192. Atkinson K (21 April 2010). "Chronic Fatigue Set To Disqualify Blood Donors". Voxy.co.nz. Archived from the original on 24 April 2010. Retrieved 25 June 2010.
  193. "Blood Service updates CFS donor policy". Australian Red Cross Blood Service. Archived from the original on 14 October 2013. Retrieved 7 July 2013.
  194. "Recommendation on Chronic Fatigue Syndrome and Blood Donation". American Association of Blood Banks. 18 June 2010. Archived from the original on 25 June 2010. Retrieved 25 June 2010.
  195. NHS Blood and Transplant (5 November 2010). "ME/CFS sufferers permanently deferred from giving blood". Archived from the original on 28 November 2015. Retrieved 9 October 2011.
  196. NHS Blood and Transplant. "Chronic Fatigue Syndrome". Archived from the original on 11 February 2015. Retrieved 11 February 2015.
  197. Hempel S, Chambers D, Bagnall AM, Forbes C (July 2008). "Risk factors for chronic fatigue syndrome/myalgic encephalomyelitis: a systematic scoping review of multiple predictor studies". Psychological Medicine. 38 (7): 915–26. doi:10.1017/S0033291707001602. PMID 17892624. S2CID 20277129.
  198. Tuller D (30 May 2008). "Chronic Fatigue Syndrome No Longer Seen as 'Yuppie Flu'". The New York Times. Archived from the original on 17 March 2015. Retrieved 29 June 2015.
  199. Jason LA, Richman JA, Friedberg F, Wagner L, Taylor R, Jordan KM (September 1997). "Politics, science, and the emergence of a new disease. The case of chronic fatigue syndrome". The American Psychologist. 52 (9): 973–83. doi:10.1037/0003-066X.52.9.973. PMID 9301342.
  200. "CFS/ME – The illness and the controversy | Science Media Centre". www.sciencemediacentre.org. Science Media Centre. Archived from the original on 24 August 2018. Retrieved 26 August 2018.
  201. McEvedy CP, Beard AW (January 1970). "Concept of benign myalgic encephalomyelitis". British Medical Journal. 1 (5687): 11–15. doi:10.1136/bmj.1.5687.11. PMC 1700895. PMID 5411596.
  202. "An Outbreak of Encephalomyelitis in the Royal Free Hospital Group, London, in 1955". British Medical Journal. 2 (5050): 895–904. 19 October 1957. ISSN 0007-1447. PMC 1962472. PMID 13472002.
  203. Speight N (2013). "Myalgic encephalomyelitis/chronic fatigue syndrome: Review of history, clinical features, and controversies". Saudi Journal of Medicine & Medical Sciences. 1 (1): 11–13. doi:10.4103/1658-631x.112905.
  204. Cowley G, Hager M, Joseph N (12 November 1990), "Chronic Fatigue Syndrome", Newsweek: Cover Story, archived from the original on 28 July 2020, retrieved 24 July 2020
  205. Frumkin H, Packard RM, Brown P, Berkelman RL (2004). Emerging illnesses and society: negotiating the public health agenda. Baltimore: Johns Hopkins University Press. p. 156. ISBN 978-0-8018-7942-5.
  206. Erlwein O, Kaye S, McClure MO, Weber J, Wills G, Collier D, Wessely S, Cleare A (January 2010). Nixon DF (ed.). "Failure to detect the novel retrovirus XMRV in chronic fatigue syndrome". PLOS ONE. 5 (1): e8519. Bibcode:2010PLoSO...5.8519E. doi:10.1371/journal.pone.0008519. PMC 2795199. PMID 20066031.
  207. Groom HC, Boucherit VC, Makinson K, Randal E, Baptista S, Hagan S, Gow JW, Mattes FM, Breuer J, Kerr JR, Stoye JP, Bishop KN (February 2010). "Absence of xenotropic murine leukaemia virus-related virus in UK patients with chronic fatigue syndrome". Retrovirology. 7 (1): 10. doi:10.1186/1742-4690-7-10. PMC 2839973. PMID 20156349.
  208. van Kuppeveld FJ, de Jong AS, Lanke KH, Verhaegh GW, Melchers WJ, Swanink CM, Bleijenberg G, Netea MG, Galama JM, van der Meer JW (February 2010). "Prevalence of xenotropic murine leukaemia virus-related virus in patients with chronic fatigue syndrome in the Netherlands: retrospective analysis of samples from an established cohort". BMJ. 340: c1018. doi:10.1136/bmj.c1018. PMC 2829122. PMID 20185493.
  209. Alberts B (December 2011). "Retraction". Science. 334 (6063): 1636. Bibcode:2011Sci...334.1636A. doi:10.1126/science.334.6063.1636-a. PMID 22194552.
  210. Lo SC, Pripuzova N, Li B, Komaroff AL, Hung GC, Wang R, Alter HJ (January 2012). "Retraction for Lo et al., Detection of MLV-related virus gene sequences in blood of patients with chronic fatigue syndrome and healthy blood donors". Proceedings of the National Academy of Sciences of the United States of America. 109 (1): 346. Bibcode:2012PNAS..109..346.. doi:10.1073/pnas.1119641109. PMC 3252929. PMID 22203980.
  211. "Parliamentary Briefing: Appropriate ME treatment". parliament.uk. 22 January 2019. Archived from the original on 28 July 2020. Retrieved 27 May 2020.
  212. Chronic fatigue syndrome/myalgic encephalomyelitis (or encephalopathy): diagnosis and management. London: National Institute for Health and Clinical Excellence. 22 August 2007. Archived from the original on 27 January 2021. Retrieved 3 February 2021.
  213. "ME_Inquiry_Report" (PDF). Erythos.com. Archived (PDF) from the original on 10 July 2011. Retrieved 28 January 2011.
  214. "Chronic Fatigue Syndrome/Myalgic Encephalomyelitis". MRC.ac.uk. Archived from the original on 6 January 2011. Retrieved 28 January 2011.
  215. "APPGME.org.uk" (PDF). Archived (PDF) from the original on 11 August 2011. Retrieved 28 January 2011.
  216. Cohen J (27 October 2015). "Criticism mounts of a long, controversial chronic fatigue study". Science. doi:10.1126/science.aad4784.
  217. Dove A (August 2000). "GAO reports on CFS funding controversy". Nature Medicine. 6 (8): 846. doi:10.1038/78579. PMID 10932206. S2CID 1431198.
  218. "NIH takes action to bolster research on Myalgic Encephalomyelitis/Chronic Fatigue Syndrome". 29 October 2015. Archived from the original on 31 May 2021. Retrieved 29 October 2015.
  219. "Golden Girls, Chronic Fatigue Syndrome, and the Legacies of Hysteria". Nursing Clio. 25 September 2018. Archived from the original on 28 July 2020. Retrieved 14 November 2019.
  220. Cain P (2007). "Ann Bannon". Leading the parade: Conversations with America's most influential lesbians and gay men. Scarecrow Press, Inc. pp. 155–63. ISBN 978-0-8108-5913-5.
  221. Hylton WS (18 December 2014). "The Unbreakable Laura Hillenbrand". The New York Times. ISSN 0362-4331. Archived from the original on 4 March 2020. Retrieved 27 June 2020.
  222. Parker-Pope T (4 February 2011). "An Author Escapes From Chronic Fatigue Syndrome". Well. Archived from the original on 4 July 2020. Retrieved 27 June 2020.
  223. Whistler T, Unger ER, Nisenbaum R, Vernon SD (December 2003). "Integration of gene expression, clinical, and epidemiologic data to characterize Chronic Fatigue Syndrome". Journal of Translational Medicine. 1 (1): 10. doi:10.1186/1479-5876-1-10. PMC 305360. PMID 14641939.
  224. Kennedy G, Abbot NC, Spence V, Underwood C, Belch JJ (February 2004). "The specificity of the CDC-1994 criteria for chronic fatigue syndrome: comparison of health status in three groups of patients who fulfill the criteria". Annals of Epidemiology. 14 (2): 95–100. doi:10.1016/j.annepidem.2003.10.004. PMID 15018881.
  225. Aslakson E, Vollmer-Conna U, White PD (April 2006). "The validity of an empirical delineation of heterogeneity in chronic unexplained fatigue". Pharmacogenomics. 7 (3): 365–73. doi:10.2217/14622416.7.3.365. PMID 16610947.
  226. Esfandyarpour R, Kashi A, Nemat-Gorgani M, Wilhelmy J, Davis RW (May 2019). "A nanoelectronics-blood-based diagnostic biomarker for myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS)". Proceedings of the National Academy of Sciences of the United States of America. 116 (21): 10250–10257. Bibcode:2019PNAS..11610250E. doi:10.1073/pnas.1901274116. PMC 6535016. PMID 31036648.
  227. Saha AK, Schmidt BR, Wilhelmy J, Nguyen V, Abugherir A, Do JK, et al. (1 January 2019). "Red blood cell deformability is diminished in patients with Chronic Fatigue Syndrome". Clinical Hemorheology and Microcirculation. 71 (1): 113–116. doi:10.3233/CH-180469. PMC 6398549. PMID 30594919.
  228. Wirth KJ, Scheibenbogen C (April 2021). "Pathophysiology of skeletal muscle disturbances in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS)". Journal of Translational Medicine. 19 (1): 162. doi:10.1186/s12967-021-02833-2. PMC 8058748. PMID 33882940.
  229. Kim DY, Lee JS, Son CG (October 2020). "Systematic Review of Primary Outcome Measurements for Chronic Fatigue Syndrome/Myalgic Encephalomyelitis (CFS/ME) in Randomized Controlled Trials". Journal of Clinical Medicine. 9 (11): 3463. doi:10.3390/jcm9113463. PMC 7692998. PMID 33126460.
This article is issued from Wikipedia. The text is licensed under Creative Commons - Attribution - Sharealike. Additional terms may apply for the media files.