Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Nov 30.
Published in final edited form as: Nat Rev Clin Oncol. 2014 Aug 12;11(10):597–609. doi: 10.1038/nrclinonc.2014.127

Cancer-related fatigue: Mechanisms, risk factors, and treatments

Julienne E Bower 1,2,3,4
PMCID: PMC4664449  NIHMSID: NIHMS664147  PMID: 25113839

Abstract

Fatigue is one of the most common and distressing side effects of cancer and its treatment, and may persist for years after treatment completion in otherwise healthy survivors. Cancer-related fatigue causes disruption in all aspects of quality of life and may be a risk factor for reduced survival. The prevalence and course of fatigue in cancer patients has been well characterized, and there is growing understanding of underlying biological mechanisms. Inflammation has emerged as a key biological pathway for cancer-related fatigue, with studies documenting links between markers of inflammation and fatigue before, during, and particularly after treatment. There is considerable variability in the experience of cancer-related fatigue that is not explained by disease- or treatment-related characteristics, suggesting that host factors may play an important role in the development and persistence of this symptom. Indeed, longitudinal studies have begun to identify genetic, biological, psychosocial, and behavioral risk factors for cancer-related fatigue. Given the multi-factorial nature of cancer-related fatigue, a variety of intervention approaches have been examined in randomized controlled trials, including physical activity, psychosocial, mind-body, and pharmacological treatments. Although there is currently no gold standard for treating fatigue, several of these approaches have shown beneficial effects and can be recommended to patients. This report provides a state of the science review of mechanisms, risk factors, and interventions for cancer-related fatigue, with a focus on recent longitudinal studies and randomized trials that have targeted fatigued patients.

INTRODUCTION

Fatigue is now recognized as one of the most common and distressing side effects of cancer and its treatment1. Fatigue may be elevated before treatment onset and typically increases during cancer treatment, including treatment with radiation2, chemotherapy3, hormonal, and/or biological therapies4. Prevalence estimates of fatigue during treatment range from 25% to 99% depending on the patient population, type of treatment received, and method of assessment1, 5. In the majority of studies, 30% to 60% of patients report moderate to severe fatigue during treatment, which in some cases may lead to treatment discontinuation. Fatigue typically improves in the year after treatment completion, although a significant minority of patients continue to experience fatigue for months or years after successful treatment6, 7. Studies of long-term cancer survivors suggest that approximately one-quarter to one-third experience persistent fatigue for up to 10 years after cancer diagnosis8, 9. Fatigue has a negative impact on work, social relationships, mood, and daily activities and causes significant impairment in overall quality of life during and after treatment6, 1012. Fatigue may also be a predictor of shorter survival13, 14.

Patient reports suggest that cancer-related fatigue is more severe, more persistent, and more debilitating than “normal” fatigue caused by lack of sleep or overexertion and is not relieved by adequate sleep or rest15. Indeed, studies have confirmed that the intensity and duration of fatigue experienced by cancer patients and survivors is significantly greater than healthy controls and causes greater impairment in quality of life3, 10, 16, 17. Cancer-related fatigue is multi-dimensional and may have physical, mental, and emotional manifestations including generalized weakness, diminished concentration or attention, decreased motivation or interest to engage in usual activities, and emotional lability7 (Box 1). Although cancer-related fatigue shares some characteristics with depression, patients experience fatigue as a distinct and central symptom that impairs mood and functional abilities.

Box 1. Defining and assessing cancer-related fatigue.

Cancer-related fatigue (CRF) has been defined as a distressing, persistent, subjective sense of physical, emotional, and/or cognitive tiredness or exhaustion related to cancer and/or cancer treatment that is not proportional to recent activity and interferes with usual functioning. Fatigue is a subjective experience and patient self-report is the gold-standard method for assessing CRF. A number of measures have been developed to assess fatigue in the cancer context, which vary from single-item assessments of fatigue severity to multi-dimensional scales assessing different components of fatigue (severity, duration, interference; mental, physical, emotional fatigue)170. A common approach to assessing fatigue involves asking patients to rate their fatigue on a 0–10 scale, where mild fatigue is indicated as a score of 1 to 2, moderate fatigue as 4 to 6, and severe fatigue as 7 to 10171. In 1998, formal diagnostic criteria were proposed to define a clinical syndrome of cancer-related fatigue172. One of the advantages of the criteria is that they specifically ask patients whether the fatigue they are experiencing is a consequence of cancer or cancer therapy. In addition, the criteria attempt to distinguish CRF from fatigue that occurs secondary to depression. However, these criteria have not been widely used to identify patients with CRF or to direct intervention efforts173, and may be overly stringent.

Despite the prevalence and negative impact of cancer-related fatigue, this symptom is underreported by patients and undertreated by clinicians18. One of the barriers to the assessment and management of fatigue may be a lack of information about mechanisms underlying this symptom, risk factors, and effective treatments. This review will summarize recent work on the biological mechanisms that underlie cancer-related fatigue, focusing on inflammation as a key pathway. In addition, risk factors for fatigue will be examined, as growing evidence suggests that only certain patients are at risk for severe and persistent fatigue. The identification of potential risk factors has been facilitated by recent longitudinal studies assessing pre-treatment risk factors for on-treatment and post-treatment fatigue. Finally, interventions for cancer-related fatigue will be reviewed, including physical activity, psychosocial, mind-body, and pharmacologic approaches. The focus here is on randomized controlled trials that have specifically targeted fatigue and particularly those that have enrolled fatigued patients.

MECHANISMS FOR CANCER-RELATED FATIGUE

Fatigue in cancer patients is multi-factorial and may be influenced by a variety of demographic, medical, psychosocial, behavioral, and biological factors. In terms of demographic factors, marital status and income have been linked to cancer-related fatigue in some reports, with unmarried patients who have a lower household income reporting higher levels of fatigue6, 19. This suggests that contextual factors (e.g., absence of partner who can provide instrumental and emotional support) may influence the experience of this symptom. Other potential contributing factors include medical comorbidities, medications, nutritional issues, physical deconditioning, mood disturbance, and physical symptoms, among others20. However, fatigue often occurs in patients who are otherwise healthy and have few if any of these contributing factors, suggesting that other processes may also be at work. Of note, treatment-related factors (e.g., type of treatment, dose-intensity) are not consistently associated with fatigue, particularly in the post-treatment period.

A variety of biological mechanisms of CRF have been proposed and investigated over the past two decades21, 22. These include anemia, cytokine dysregulation, hypothalamic-pituitary-adrenal (HPA) axis dysregulation, five hydroxy tryptophan (5-HT) neurotransmitter dysregulation, and alterations in adenosine triphosphate and muscle metabolism, among others. To date, the mechanism that has garnered the most empirical attention and support is cytokine dyregulation, with a focus on pro-inflammatory cytokines.

Inflammation and cancer-related fatigue

The possibility that inflammatory processes may be involved in the etiology of cancer-related fatigue draws from basic research on neural-immune signaling. This body of work has demonstrated that peripheral inflammatory cytokines can signal the central nervous system to generate symptoms of fatigue and other behavioral changes via alterations in neural processes23, 24 (Box 2). In the cancer context, investigators have proposed that tumors and the treatments used to eradicate them can activate the pro-inflammatory cytokine network, leading to symptoms of fatigue via cytokine signaling in the central nervous system2527. In the pre-treatment period, the tumor itself may be a source for pro-inflammatory cytokines28, 29 while during treatment, cytokines may be produced in response to tissue damage from radiation or chemotherapy28, 30. The inflammatory response may persist well after treatment completion as the host tries to deal with persisting pathogenesis and alterations in homeostatis. Of note, factors other than cancer and its treatment can influence inflammatory activity, including psychological, behavioral, and biological risk factors.

Box 2. Neural immune signaling: Effects on fatigue and other behavioral symptoms.

Basic research on neuro-immune interactions has documented behavioral effects of peripheral immune activation that are mediated by pro-inflammatory cytokines23. Signals from the peripheral immune system are conveyed to the central nervous system through several routes, including direct neural activation via the afferent vagus nerve, transport of peripheral cytokines across the blood-brain barrier via carrier molecules, and interaction of circulating cytokines with brain cytokine receptors in areas that lack a functional blood-brain barrier (i.e., circumventricular organs) and with brain vascular endothelial cells that release second messages to stimulate cytokine production in the brain79. Cytokine signaling leads to changes in neural activity, physiological processes (e.g., fever), and behavior174. In animal models, injection or induction of pro-inflammatory cytokines leads to decreased motor activity (presumably a behavioral manifestation of fatigue) as well as reduced food and water intake, social withdrawal, anhedonia, and altered cognition. These behavioral changes have been collectively described as “sickness behavior” and are thought to represent a motivational shift designed to facilitate recovery and prevent the spread of infection79, 175. In humans, pharmacologic doses of cytokines given for treatment of cancer or hepatitis C are associated with significant increases in fatigue and other markers of sickness, including depressed mood and sleep disturbance176178. Experimental studies of cytokine induction in healthy individuals have documented similar effects, with subjects reporting increased fatigue following endotoxin administration that are correlated with elevations in circulating concentrations of pro-inflammatory cytokines179, 180. In observational studies, sub-clinical levels of inflammatory markers prospectively predict the development of fatigue in otherwise healthy individuals181, 182. Further, pharmacologic agents that block the pro-inflammatory cytokine TNF-α lead to reduced fatigue among individuals with inflammatory conditions162 and in pilot studies with cancer patients160. Together, this evidence provides a strong biological rationale for inflammation as a potential mechanism underlying cancer-related fatigue.

Here, we consider human studies that have examined links between inflammation and fatigue in patients before, during, and after cancer treatment. These studies have examined a range of inflammatory markers, including circulating concentrations of the pro-inflammatory cytokines IL-1β, TNF-α, and IL-6 and markers of their activity, including the IL-1 receptor antagonist (IL-1RA), the soluble TNF receptor (sTNFR), the soluble IL-6 receptor (sIL-6R), and C reactive protein (CRP). Alterations in other biological systems that have been linked to cancer-related fatigue will also be addressed.

Inflammation and fatigue prior to cancer treatment

A handful of studies have examined associations between inflammation and fatigue before treatment. In patients with newly diagnosed acute myelogenous leukemia or myelodysplastic syndrome, levels of several inflammatory markers were correlated with symptoms of fatigue31. Similar results have emerged in studies conducted with ovarian cancer patients assessed prior to surgery, which found a positive association between plasma concentrations of IL-6 and fatigue32, 33. On the other hand, a recent study of breast cancer patients assessed prior to surgery did not find elevated levels of CRP in those categorized as “fatigued”34. It is possible that small, localized breast tumors may not produce elevations in systemic cytokine concentrations that are sufficient to induce symptoms of fatigue. Another recent study conducted with breast cancer patients assessed prior to chemotherapy found that fatigue was associated with elevations in CRP35; however, the majority of patients in this study were assessed after surgery, which is known to elicit an inflammatory response.

Inflammation and fatigue during cancer treatment

Radiation therapy and chemotherapy are two of the most common types of cancer treatment, and both are associated with increases in fatigue36 and with elevations in certain inflammatory markers37, 38. Thus, investigators have hypothesized that activation of pro-inflammatory cytokines may contribute to fatigue during treatment. Early reports conducted with patients undergoing treatment were conflicting, possibly due to constraints of study methods (including use of non-standard measures to detect cytokine levels) and focus on cross-sectional associations between cytokine levels and fatigue3942. However, more recent reports using mixed model analyses to model changes over time have yielded more positive results. In a study of patients undergoing radiation therapy for early-stage breast or prostate cancer, we found that increases in serum levels of inflammatory markers CRP and IL-1 receptor antagonist were associated with increases in fatigue43. Similarly, among breast cancer patients undergoing chemotherapy, changes in IL-6 were associated with changes in fatigue over the course of treatment44. Wang and colleagues intensively examined sickness symptoms and inflammatory markers in patients undergoing combined radiation and chemotherapy therapy for locally advanced colorectal, esophageal, and non-small cell lung cancer45, 46. These investigators documented acute increases in markers of inflammation that were correlated with increases in fatigue and other prominent sickness symptoms. Similar effects were seen in a study of individuals undergoing allogeneic hematopoietic stem cell transplantation (which includes high-dose chemotherapy) for acute myelogenous leukemia and myelodysplastic syndrome47.

Inflammation and post-treatment fatigue in cancer survivors

Although fatigue typically abates in the year after cancer treatment, approximately 20–30% of cancer survivors report persistent fatigue that may last for 5–10 years post-treatment and beyond8. Our group has documented consistent alterations in the pro-inflammatory cytokine network among breast cancer survivors with persistent post-treatment fatigue, including elevations in circulating markers of inflammation48, 49 and elevated intracellular cytokine production by monocytes after LPS stimulation49, 50. We have more recently shown an association between fatigue and elevations in plasma levels of the soluble TNF receptor type II (sTNF-RII), a downstream marker of TNF activity, in breast cancer survivors within one month after treatment; this association was particularly strong among women treated with chemotherapy51.

These findings have been replicated in larger samples of breast cancer survivors. For example, Alexander et al. found significant elevations in CRP in breast cancer survivors who met stringent criteria for cancer-related fatigue (n = 60) relative to non-fatigued controls (n = 104)52. Mean levels of CRP were 3.91 mg/dL among the fatigued survivors (vs. 2.74 in the non-fatigued group), indicating low-grade inflammation. In a sample of 633 breast cancer survivors, higher CRP was associated with increased odds of being classified as fatigued, controlling for age, race, menopausal status, antidepressant/anxiolytic use, medical comorbidities, and BMI53. In a sample of 299 breast cancer survivors, Orre et al. found a positive association between CRP and fatigue that remained significant after controlling for age, BMI, depressive symptoms, sleep disturbance, medication use, and self-rated health54. This group has also documented a positive association between inflammatory markers and fatigue in long-term survivors of testicular cancer55. In one of the few longitudinal studies to examine associations between inflammation and fatigue following treatment completion, Schrepf et al. found that decreases in IL-6 were correlated with declines in fatigue in ovarian cancer patients in the year after treatment completion56.

Several recent studies have probed the molecular underpinnings of cancer-related fatigue by conducting genome-wide expression analyses on leukocytes from breast cancer survivors with persistent fatigue compared to non-fatigued survivors. A study conducted by our group focused on transcription of inflammation-related genes, particularly those responsive to the proinflammatory NF-κB transcription control pathway57. Results showed that breast cancer survivors with persistent fatigue showed increased expression of genes encoding proinflammatory cytokines and other mediators of immunologic activation. Further, promoter-based bioinformatic analyses indicated increased activity of proinflammatory NF-κB/Rel transcription factors in leukocytes from fatigued breast cancer survivors, which might structure the observed differences in the expression of inflammation-related genes. In contrast, an exploratory study by Landmark-Hoyvik et al. found that fatigued breast cancer survivors showed altered expression of genes involved in plasma or B cell pathways58. Gene expression profiling has also been used to identify gene transcripts associated with fatigue in prostate cancer patients, with some preliminary evidence for elevated expression of inflammation-related genes in fatigued patients59, 60.

Cellular immunity, latent viral reactivation, and fatigue

Cancer treatments can cause pronounced and prolonged alterations in the cellular immune system61, 62, which may underlie alterations in inflammatory activity and associated symptoms of fatigue. Our group has documented alterations in T cell populations and myeloid dendritic cells in breast cancer survivors with persistent fatigue that are correlated with inflammatory processes49, 63. Other groups have shown more global changes in the cellular immune system in relation to fatigue, including elevations in leukocyte numbers among fatigued breast cancer survivors52, 58, though these effects have not been consistently replicated64. One of the few longitudinal studies in this area found that elevated leukocyte counts in the post-treatment period predicted persistent fatigue over a 2–3 year follow-up in breast cancer survivors65.

Another potential explanation for elevated inflammatory processes and fatigue in cancer patients is reactivation of latent herpesviruses66, 67. A recent study conducted with breast cancer patients prior to treatment found that elevated cytomegalovirus (CMV) antibody titers were associated with a greater likelihood of being fatigued, as well as higher levels of CRP68. Cancer treatments such as chemotherapy promote viral reactivation and associated increases in inflammatory markers69 which may have long-term implications for immune regulation and recovery as well as fatigue and other behavioral symptoms.

Neuroendocrine alterations and cancer-related fatigue

Hypothalamic-pituitary-adrenal (HPA) axis dysregulation and fatigue

Alterations in the HPA axis have been proposed as a mechanism underlying cancer-related fatigue, either directly or through effects on inflammatory processes. The HPA axis is an important regulator of cytokine production and has potent anti-inflammatory effects70. These effects can occur via alterations in glucocorticoid production (including dysregulated circadian profiles) and/or decreased sensitivity of the glucocorticoid receptor (GR) to hormone ligation71. Preliminary evidence suggests alterations in both pathways among patients with cancer-related fatigue. In terms of cortisol production, breast cancer survivors with persistent fatigue show alterations in diurnal cortisol slope, with elevated levels of evening cortisol relative to non-fatigued controls72. Fatigued breast cancer survivors also demonstrate blunted cortisol responses to psychological stress73 that are correlated with elevations in stimulated cytokine production and may underlie elevated inflammatory activity50. However, studies have not shown alterations in total daily cortisol production or 24-hour urinary free cortisol in breast cancer survivors with post-treatment fatigue52, 72. In ovarian cancer patients, higher levels of evening cortisol and reduced cortisol variability are associated with fatigue before treatment onset74, and normalization of cortisol profiles in the following year is associated with reductions in fatigue56.

In terms of glucocorticoid receptor sensitivity, genome-wide transcriptional profiling of leukocytes from fatigued breast cancer survivors showed a marked down-regulation of genes with response elements for the glucocorticoid receptor, suggesting a state of functional GR resistance57. Reduced GR sensitivity may contribute to the tonic upregulation of NF-κB observed in fatigued survivors, consistent with studies linking GR desensitization to increased NF-κB activity in non-cancer populations75, 76.

Autonomic nervous system dysregulation and fatigue

Preliminary reports suggest that alterations in the autonomic nervous system may also be relevant for cancer-related fatigue. In a study of breast cancer survivors, fatigue was associated with elevated levels of norepinephrine (indicating increased sympathetic activity) and lower heart rate variability (indicating reduced parasympathetic activity), both at rest and in response to a psychological challenge77. We recently replicated the association between cancer-related fatigue and lower resting HRV in a sample of pre-menopausal breast cancer survivors, who are at particular risk for elevated fatigue78.

Like the HPA axis, the autonomic nervous system regulates immune and inflammatory processes79, which may mediate effects on cancer-related fatigue. In general, sympathetic nervous system activity is associated with increased inflammatory activity, whereas parasympathetic nervous system activity is associated with reduced inflammatory activity. However, inflammation did not mediate the association between low HRV and fatigue in our sample of pre-menopausal breast cancer survivors78, suggesting that other pathways may also be relevant.

Summary of biological mechanisms

Overall, results from studies conducted with cancer patients and survivors support the hypothesis that inflammatory processes contribute to fatigue during and particularly after treatment. The association between inflammation and fatigue has been documented primarily in breast cancer survivors, though similar effects have been observed in ovarian and testicular cancer survivors. Importantly, most of the studies in this area have controlled for potential biobehavioral confounds, including age and BMI, indicating that links between inflammation and fatigue are not driven by these factors. Findings are not entirely uniform, and associations have not been found in all patient groups80, for all aspects of fatigue55, 81, or for all inflammatory markers51, 54. Inconsistency across studies may be due to differences in the definition and assessment of cancer-related fatigue, disease- and treatment-related characteristics, and type (and quality) of immunologic assessments. Different components of the pro-inflammatory cytokine network may be associated with different aspects of fatigue, in different patient groups, at difference stages of the cancer trajectory. Thus, it is important to assess key components of the cytokine network, as well as key dimensions of fatigue, using valid and reliable measurement techniques. Of note, one of the most consistent findings in this literature is the link between CRP and post-treatment fatigue, perhaps because CRP is routinely assayed in many clinical laboratories (and thus may be more reliably measured than other markers of inflammation) and because acute effects of treatment have resolved at this time.

Studies have also documented associations between cancer-related fatigue and alterations in the immune and neuroendocrine system, including changes in leukocyte subsets, reactivation of latent herpesviruses, dysregulated cortisol rhythm, reduced glucocorticoid receptor sensitivity, and alterations in the autonomic nervous system. These systems are closely linked to inflammation and may influence fatigue by initiating or maintaining elevated inflammatory activity. In addition, changes in these systems may have direct effects on fatigue. At this point, it is unclear whether these alterations play a causal role in the development and persistence of cancer-related fatigue, as activity in these systems has typically been measured concurrently with fatigue. In addition, because most studies have focused on post-treatment survivors, it is unclear whether the alterations associated with fatigue were driven by cancer treatment (e.g., chemotherapy effects on the cellular immune system) or may have been present prior to cancer diagnosis and treatment. For example, a recent prospective study conducted with military personnel deployed to a war zone found that pre-deployment levels of GR sensitivity predicted the development of post-deployment fatigue82. Similarly, it is possible that pre-cancer alterations in GR sensitivity and other biological systems may serve as a risk factor for cancer-related fatigue, comparable to the risk factors discussed below. Prospective, longitudinal studies are required to determine the role of neuroendocrine and immune alterations in the onset and persistence of fatigue and the mechanisms through which this occurs.

RISK FACTORS FOR CANCER-RELATED FATIGUE

As noted previously, fatigue typically increases during cancer treatment and improves in the year after treatment completion. However, there is considerable variability in the experience of fatigue before, during, and after treatment19, 83, suggesting that certain individuals may be at particular risk for this disabling symptom. Of note, there is also variability in the inflammatory response to treatment, which is correlated with variability in fatigue (e.g.,43). Over the past several years, longitudinal studies have begun to examine risk factors for cancer-related fatigue, and particularly fatigue that persists for months or years after cancer treatment. Studies in this area have focused primarily on demographic, medical, behavioral, and psychosocial predictors, but genetic risk factors are of growing interest. Identification of these factors is important for advancing our understanding of this symptom and for improving identification and treatment of vulnerable patients. In this section, we review this growing literature and suggest pathways through which these factors may influence fatigue.

Genetic risk factors

Given growing evidence that inflammation plays a key role in the onset and persistence of cancer-related fatigue, investigators have begun to examine genetic factors that influence pro-inflammatory cytokine activity as potential risk factors for fatigue in the cancer setting. Most of these studies have used a candidate gene approach, focusing on single nucleotide polymorphisms (SNPs) in inflammation-related genes including IL1B, IL6, and TNF. There is preliminary evidence that variations in these genes are associated with cancer-related fatigue during and after treatment. In longitudinal studies with patients undergoing radiation therapy, polymorphisms in TNFA and IL6 were associated with elevated fatigue before, during, and for four months after treatment completion84, 85. Polymorphisms in TNFA and IL6 were also associated with increases in fatigue in a small longitudinal study of prostate cancer patients undergoing androgen deprivation therapy86.

Cross-sectional studies conducted with cancer populations have yielded similar results. In two large studies conducted with lung cancer patients, polymorphisms in IL8 were associated with increased fatigue before treatment onset87, while polymorphisms in IL1B and IL1RN were associated with post-treatment fatigue88. In studies conducted with breast cancer survivors, polymorphisms in TNFA, IL6, IL1B have been associated with elevated fatigue89, 90, although these findings have not been consistently replicated91. Of note, polymorphisms in inflammation-related genes have been linked to fatigue in other patient populations92, 93 and in cancer caregivers85, suggesting that inflammation-promoting genes may serve as a general risk factor for fatigue symptomatology.

Overall, research in this area supports the hypothesis that inflammatory processes are important for cancer-related fatigue and suggests that certain cytokine genetic variants may increase risk for this symptom. However, the majority of this work has been conducted in relatively small samples and requires replication. In addition, genome-wide scanning might help to identify other genetic risk factors for fatigue, related to inflammation or other systems21.

Psychological and biobehavioral risk factors

Pre-treatment fatigue

Across studies, the strongest and most consistent predictor of post-treatment fatigue is pre-treatment fatigue. Patients who report higher levels of fatigue before radiation and/or chemotherapy also report elevated fatigue immediately after treatment completion94, over the following year35, 95, 96, and up to 2.5 years later97. In studies that compared multiple predictors, pre-treatment fatigue emerged as one of the strongest, if not the strongest predictor of fatigue in the post-treatment period35, 95. Together, these findings suggest that whatever biological, psychological, or behavioral dysregulation contributes to cancer-related fatigue may be present before treatment onset.

Depression

Depression is of particular interest as a risk factor for cancer-related fatigue, as fatigue and depression are strongly correlated in cancer populations98. The association between these two constructs is complex; fatigue is a symptom of depression, but may also precipitate depressed mood due to interference with social, occupational, and leisure activities. Rather than trying to disentangle causality, it may be more informative to examine whether mood disturbance predicts the onset and persistence of fatigue and can thus be used to identify vulnerable patients. Indeed, there is evidence from several longitudinal studies that pre-treatment depression and anxiety predict cancer-related fatigue before, during and after treatment65, 83, 94, 95, 97, 99. Of note, the majority of these studies did not control for pre-treatment fatigue, and thus the independent contribution of depression over and above pre-existing fatigue is not entirely clear. A history of major depressive disorder (and treatment for mental problems prior to cancer diagnosis) also predicted post-treatment fatigue in several reports65, 100, with effects observed up to 42 months after treatment completion101. Thus, patients with a history of mental illness and those with elevated distress in acute stage of cancer diagnosis and treatment onset appear to be at risk for persistent post-treatment fatigue.

Sleep disturbance

Like depressed mood, sleep disturbance is closely correlated with fatigue in cancer populations, and investigators have hypothesized that sleep problems may contribute to daytime symptoms of fatigue102. Indeed, studies conducted with breast and prostate cancer patients undergoing radiation therapy have shown that pre-treatment sleep disturbance is associated with higher levels of fatigue before, during, and for up to 6 months after treatment completion83, 99. In patients with gynecologic cancers beginning chemotherapy, higher levels of sleep disturbance (assessed objectively using actigraphy) predicted earlier subsequent peaks in fatigue103. Of note, fatigue predicted subsequent elevations in depressed mood in this study, suggesting a cascade effect among these symptoms in the early stages of cancer treatment. Together, these reports suggest that sleep disturbance may be a risk factor for cancer-related fatigue, although additional research in the post-treatment period is needed. Studies of cancer survivors have shown that fatigue can persist even when patients report getting adequate sleep, indicating that other factors contribute to fatigue maintenance over time.

Physical activity, physical deconditioning, and body mass index

Physical inactivity is correlated with cancer-related fatigue; patients who are more fatigued typically report lower levels of physical activity104, 105. Lack of physical activity may lead to physical deconditioning, which makes everyday tasks more challenging and potentially contributes to the development and persistence of fatigue. Indeed, cancer survivors with post-treatment fatigue show decreased cardiorespiratory fitness106. However, few studies have examined the temporal association between activity, deconditioning, and fatigue, making it difficult to determine causality. There is evidence from longitudinal studies that lower levels of physical activity after treatment completion predict persistent fatigue in breast cancer survivors19, 107, although elevated fatigue during treatment may have preceded (and precipitated) lower physical activity in these reports. In either case, low levels of physical activity and associated decreases in cardiorespiratory fitness may play an important role in the development and/or persistence of cancer-related fatigue. Elevated body mass index (BMI) has also been linked with fatigue, and a longitudinal study of women with early-stage breast cancer found that BMI was one of the key predictors of fatigue at 619 and 42 months post-treatment101. Body mass index also predicted persistent fatigue in a longitudinal study of post-treatment breast cancer survivors, above and beyond other risk factors65.

Coping and appraisal

Psychological responses to cancer diagnosis and treatment can also influence fatigue symptoms. In particular, the tendency to “catastrophize”, or engage in negative self-statements and thoughts regarding fatigue (e.g., I begin thinking of all the possible bad things that could go wrong in association with the fatigue; I tell myself I don’t think I can bear the fatigue any longer) was associated with higher levels of fatigue during108 and for up to 42 months after treatment100, 101 in research with breast cancer patients. Indeed, catastrophizing was one of the strongest predictors of persistent elevations in fatigue in these reports. Similarly, patients who expect to experience fatigue are more likely to report elevated fatigue after cancer surgery109. Thus, patients’ negative expectations and coping strategies early in the cancer trajectory appear to put them at increased risk for post-treatment fatigue.

Other psychosocial risk factors

Emerging evidence has identified other psychological risk factors for cancer-related fatigue. Exposure to childhood stress, including experiences of abuse and neglect, is associated with elevated fatigue in cross-sectional studies of breast cancer survivors110, 111. These findings are consistent with research conducted in non-cancer populations showing that early life stress is associated with increased risk for fatigue112114. Loneliness is also associated with elevated fatigue in cancer survivors (and older adults) and predicts increases in fatigue over time115.

Summary and mechanisms

A growing number of longitudinal studies have identified risk factors for fatigue during and after cancer treatment. These include genetic risk factors (SNPs in inflammation-related genes), psychosocial factors (pre-treatment fatigue, depression, and sleep disturbance, dysfunctional coping and appraisal processes, loneliness, early life stress) and biobehavioral factors (physical inactivity, elevated body mass index). Many of these factors are associated with inflammatory processes, including depression, sleep disturbance, physical inactivity, body mass index, early life stress, and loneliness. Individuals with these risk factors may already have elevated inflammatory activity at the time of diagnosis, increasing risk for pre-treatment fatigue. In addition, these factors may increase the inflammatory response to diagnosis and treatment. Indeed, in experimental studies conducted with non-cancer samples, individuals with a history of depression and early life stress show an exaggerated inflammatory response to psychosocial challenge116, 117. The mechanisms through which these and other risk factors influence fatigue are an important topic for future research.

It may also be useful to distinguish between factors that increase risk for fatigue during treatment (precipitating factors) and those that lead to its persistence in the post-treatment period (perpetuating factors)96. To date, studies have primarily focused on the period during and immediately after treatment, or in the years after treatment completion. Longitudinal studies that follow patients from pre-treatment into the survivorship period will illuminate which factors are most important for acute and more persistent fatigue. This will help to identify appropriate targets for intervention at different stages of the cancer trajectory.

TREATMENTS FOR CANCER-RELATED FATIGUE

A diverse range of treatment approaches have been used to address cancer-related fatigue during and after cancer treatment. Indeed, a recent review of the literature indicated that more than 170 intervention studies that included fatigue as a primary or secondary outcome have been conducted in patients with cancer20. These include physical activity, psychosocial, mind-body, and pharmacological interventions. Perhaps because the etiology of cancer-related fatigue is multi-factorial and still poorly understood, there is currently no “gold standard” for treatment of this symptom. Still, a number of these approaches have been shown to be beneficial in reducing cancer-related fatigue, as reviewed below.

Exercise

There are a large and growing number of randomized controlled trials of exercise as a treatment for cancer-related fatigue. One recent meta-analysis of this literature identified 56 randomized controlled trials that investigated the effects of exercise on cancer-related fatigue118. Results from this meta-analysis indicated that exercise was more effective than control in reducing fatigue, with a mean effect size of −0.27. These findings are similar to other recent meta-analyses of exercise interventions for cancer-related fatigue which have yielded effect sizes in the range of −0.30 to −0.38119123, suggesting a moderate effect. Beneficial effects of exercise on fatigue have been observed in trials conducted with patients during and after treatment, indicating that exercise can be helpful at different stages of the disease trajectory. During treatment, exercise may buffer treatment-related increases in fatigue, whereas exercise may reduce fatigue in patients after treatment completion121.

What forms of exercise are particularly beneficial for fatigue? Results from the meta-analyses indicate that aerobic exercise regimens are associated with significant reductions in cancer-related fatigue118, 121. More mixed effects are seen for resistance exercise118, 122, 124. A number of different aerobic exercise regimens have shown beneficial effects on fatigue, ranging from home-based programs125 to supervised, laboratory-based programs126. Guidelines from the American College of Sports Medicine (ACSM) recommend that cancer patients and survivors engage in at least 150 minutes of moderate intensity aerobic activity each week, consistent with recommendations for the general population127. Exercise trials conducted with cancer patients often begin with more modest levels of physical activity that increase in dose and intensity over time125. ACSM guidelines further recommend that exercise should be tailored to the individual cancer survivor to account for exercise tolerance and specific diagnosis, and that patients be closely monitored to safely progress exercise intensity and avoid injury.

One important limitation of the literature on exercise for cancer-related fatigue is the lack of studies that have specifically targeted fatigued patients. These trials have typically not enrolled patients who endorse fatigue, but instead have taken all patients who meet other eligibility criteria. Thus, it is unclear whether these interventions will be feasible or effective for patients with more severe fatigue. Indeed, fatigue may be a significant barrier to participation in exercise interventions, particularly among cancer survivors128. For these patients, other strategies may be more appropriate.

Psychosocial interventions

There is a large literature on psychosocial interventions for cancer patients and survivors129, and many of these trials have included measures of fatigue. Meta-analyses of psychosocial intervention trials that included fatigue as a primary or secondary outcome have shown reductions in fatigue relative to control, with effect sizes ranging from −0.10 to −0.30, suggesting a small to moderate effect130132. The more modest effect sizes seen in these trials relative to physical activity interventions may be due to the fact that most were focused on reducing stress and improving general quality of life and did not include fatigue as a primary focus or outcome. Here, we review randomized controlled trials of psychosocial interventions that had a more explicit focus on cancer-related fatigue, including those that enrolled fatigued patients.

Several interventions have targeted fatigue among patients undergoing cancer treatment. In one study, breast cancer patients commencing chemotherapy received a 3-session individualized fatigue education and support program delivered in the clinic and by phone133. The intervention buffered the acute increase in fatigue observed in control group participants undergoing treatment, although this effect did not persist. Another trial conducted with a mixed sample of cancer patients undergoing chemotherapy found that a 3-session individualized intervention focusing on fatigue-related thoughts and behavior led to greater reductions in fatigue one month after treatment completion than usual care134. A cognitive-behavioral approach combined with hypnosis also showed beneficial effects on fatigue among breast cancer patients undergoing radiation therapy; specifically, the intervention buffered the increase in fatigue observed in controls135.

Psychoeducational interventions conducted in the post-treatment period have also demonstrated beneficial effects on fatigue. The Moving Beyond Cancer Trial, a multi-center, randomized controlled trial for breast cancer patients who had recently completed treatment, found that a brief psychoeducational video that included information on fatigue (as well as modeling of physical activity) led to significant improvements in fatigue relative to control136. Similarly, a brief group-based psychoeducational intervention for breast cancer survivors that also included physical activity led to significant improvements in fatigue137.

To date, only two psychosocial intervention studies have used fatigue as an entry criteria for trial participation. Both were conducted with cancer survivors who reported moderate to severe fatigue. Gielissen and colleagues randomized 112 fatigued cancer survivors to individual cognitive-behavioral therapy or wait-list control138. The therapy focused on perpetuating factors for persistent fatigue, including dysfunctional cognitions concerning fatigue, poor coping, fear of recurrence, dysregulation of sleep and activity patterns, and low social support. They found a significant decrease in fatigue in the intervention group relative to controls that was maintained over a long-term (1–4 year) follow-up139. Yun et al. randomized 273 fatigued cancer survivors to a 12-week, web-based, individually tailored program based on National Comprehensive Cancer Network (NCCN) fatigue guidelines140. This program provided information on cancer-related fatigue as well as energy conservation, physical activity, sleep hygiene, distress management, nutrition, and pain control. Results showed a significant decrease in fatigue in the intervention group relative to controls.

Overall, these studies suggest that educating patients about cancer-related fatigue and providing them with cognitive and behavioral strategies to manage fatigue symptoms (including physical activity) can have beneficial effects on fatigue, both during and after treatment. Preliminary evidence also indicates that more intensive interventions targeting post-treatment fatigue, both in-person and web-based, may be effective for fatigued cancer survivors.

Mind-body interventions

There is considerable interest in mind-body approaches among cancer patients, and a growing number of randomized trials have evaluated the efficacy of mind-body interventions for improving health and well-being in this population141143. We focus here on studies that used fatigue as an entry criteria for study participation, including trials of acupuncture, mindfulness meditation, yoga, and biofield therapy. Three acupuncture trials have targeted cancer survivors with moderate to severe post-chemotherapy fatigue. The largest of these trials randomized 302 patients to 6 weeks of acupuncture or usual care, and saw significant improvement in fatigue in the acupuncture group144. These findings are consistent with an earlier pilot study conducted by this group that saw beneficial effects of acupuncture relative to real or sham acupressure on post-chemotherapy fatigue145. However, in a trial that compared acupuncture to sham acupuncture for cancer survivors with post-chemotherapy fatigue, no group differences were observed146.

Building on a growing literature on the beneficial effects of mindfulness meditation, Van der Lee and colleagues randomly assigned 100 cancer survivors with severe fatigue to a 9-week program of mindfulness-based cognitive therapy or wait-list control147. The intervention was designed to help patients become aware of and inhibit potentially maladaptive automatic responses, including feelings, thoughts, and behaviors, and focused specifically on cancer-related fatigue. Patients randomized to the intervention group showed significant reductions in fatigue at post-treatment that were maintained over a 6-month follow-up. Our group conducted an Iyengar-based yoga intervention for breast cancer survivors with persistent fatigue148. The 12-week intervention specifically targeted fatigue and included postures believed to be efficacious for improving this symptom, including restorative poses, passive inversions, and passive backbends. This specialized yoga program led to significant improvements in fatigue relative to the health education control condition and also had beneficial effects on inflammatory activity149. Finally, in a study evaluating the efficacy of biofield therapy for cancer-related fatigue, Jain and colleagues randomized breast cancer survivors with fatigue to a 4-week program of biofield healing, mock healing, or wait-list control150. Both biofield healing and mock healing led to significant reductions in fatigue relative to control.

The literature on mind-body interventions for cancer-related fatigue is still quite small, but preliminary findings suggest that certain approaches may be beneficial for survivors with persistent fatigue, including mindfulness, yoga, and acupuncture. Of note, several studies that compared “real” to “sham” approaches did not find differential effects on fatigue (both were helpful)146, 150, highlighting the importance of including active control conditions in these trials. The same criticism could be applied to psychosocial interventions and physical activity interventions, which typically do not include active control groups. It is also important to note that the interventions showing positive effects were specifically designed to target fatigue, and non-specific approaches may be less effective151.

Pharmacologic interventions

A number of pharmacologic treatments have been evaluated for the treatment of cancer-related fatigue. A meta-analysis of this literature published in 2008 included 27 randomized controlled trials, including hematopoietic growth factors (14 studies), progestational steroids (4 studies), methylphenidate (a psychostimulant; 2 studies), and paroxetine (an antidepressant; 2 studies), among others152. The hematopoietic growth factor trials were all conducted with anemic patients, the majority of whom were undergoing chemotherapy. In general, treatment with hematopoietic agents led to improvements in fatigue caused by chemotherapy-induced anemia (effect size for erythropoietin = −0.30; effect size for darbepoetin = −0.13). Methylphenidate also led to greater reductions in fatigue than placebo (effect size = −0.30), but progestational steroids and paroxetine did not. Another anti-depressant, sertraline, had no beneficial effect on fatigue in patients with advanced cancer who were neither fatigued nor depressed153. A recent trial of dexamethasone for patients with advanced stage cancer who reported moderate to severe symptoms of cancer-related fatigue showed significant improvements in fatigue and quality of life154.

An updated meta-analysis included 5 randomized controlled psychostimulant trials, most of which were conducted among patients with advanced disease and used methylphenidate155. Overall, results suggested that psychostimulants were more effective than placebo in improving fatigue (effect size = −0.28), although only one of the five studies yielded a statistically significant treatment effect156. Two recent studies conducted with larger samples of patients showed no benefit for methylphenidate vs. placebo for improving fatigue157, 158, although in sub-group analyses methylphenidate did appear to be effective for patients with severe fatigue and those with advanced disease158. There is also interest in a nonamphetamine-based stimulant, the wakefulness agent modafinil, as a potential treatment for cancer-related fatigue. One large multicenter trial of patients undergoing chemotherapy found beneficial effects of modafinil among patients who reported severe fatigue at baseline, but not among those with mild or moderate fatigue159.

Based on research suggesting an inflammatory basis for cancer-related fatigue, a handful of small Phase II trials have used anti-cytokine agents to treat fatigue in patients with advanced cancer. In a study conducted by Monk and colleagues, patients undergoing dose-intensive chemotherapy who received etanercept (a TNF-decoy receptor) reported significantly less fatigue than those receiving chemotherapy alone160. A small non-randomized study also showed some benefit for infliximab (an anti-TNF antibody) on fatigue in the palliative care setting161. Beneficial effects of anti-TNF agents on fatigue have also been observed among patients with inflammatory conditions, including psoriasis162 and depression163. Although there are ongoing trials of other anti-inflammatories for cancer-related fatigue, the effectiveness of other agents (e.g., minocycline) has not been determined.

Despite interest in supplements to treat fatigue, very few controlled trials have examined the efficacy of these agents in cancer patients. One large, multisite trial examined the effect of L-cartinine for patients with fatigue, most of whom were undergoing treatment164. There was no evidence that 4 weeks of L-carnitine was more effective than placebo in improving fatigue; instead, fatigue improved in both the treatment and control groups. In contrast, a large multisite trial of American ginseng for patients with cancer-related fatigue did find beneficial effects, particularly among patients undergoing active cancer treatment165.

Overall, this literature suggests that hematopoietic agents may be effective in improving fatigue that occurs secondary to chemotherapy-induced anemia. However, because most fatigued patients are not anemic, these agents are unlikely to be useful for the majority of patients with cancer-related fatigue, particularly in the post-treatment period. Among the other agents tested to date, methylphenidate appears to be the most promising, although results are quite mixed and two recent trials did not find beneficial effects on fatigue. Because these studies have primarily focused on patients with advanced cancer, there is limited evidence for the use of psychostimulants in the management of fatigue in patients who are disease-free following active treatment. Of note, selective serotonin reuptake inhibitor (SSRI) antidepressants do not appear to have beneficial effects on cancer-related fatigue, supporting the distinction between fatigue and depression in cancer patients and suggesting that fatigue is not solely a side effect of depression. American ginseng and dexamethasone may hold promise for treating cancer-related fatigue but more research on these agents are needed.

Mechanisms for intervention effects

The literature reviewed above suggests that a variety of different intervention approaches may be useful for cancer-related fatigue, including physical activity, psycho-education, cognitive-behavioral, and mind-body approaches. These interventions have different targets and may work through different mechanism, including cognitive, behavioral, and biological mechanisms. For example, cognitive approaches to treating cancer-related fatigue specifically target maladaptive thoughts about fatigue, including catastrophizing138. Given that catastrophizing predicts more severe and persistent fatigue symptoms in cancer patients19, reducing the use of this coping mechanism may be one of the “active ingredients” that promotes reductions in fatigue. Even more physical approaches may work by changing thoughts and beliefs about fatigue; for example, patients felt more confident about their ability to manage fatigue after learning certain yoga postures148, which might lead to reductions in fatigue symptoms.

Biological mechanisms for intervention effects are also possible, including changes in inflammatory processes. Individuals who are more physically active show lower inflammatory activity166; thus, interventions that increase physical activity (and potentially reduce BMI) may influence fatigue by reducing inflammation. Of note, these interventions may also improve fatigue by improving cardiorespiratory fitness. Mind-body and psychosocial approaches may also work by reducing inflammatory activity. We have shown that a targeted yoga program for fatigued breast cancer survivors was not only effective in reducing fatigue but also led to reductions in NF-kB signaling, a key regulator of inflammatory activity149. Similar effects on inflammatory signaling were observed in a recent trial of mindfulness meditation for older adults167. Cognitive-behavioral stress management for breast cancer patients also leads to reductions in pro-inflammatory signaling168, though the effects of cognitive behavioral therapy for cancer-related fatigue on inflammation have not been examined.

CONCLUSIONS

Fatigue is one of the common and distressing side effects of cancer treatment and can persist for months or years after treatment completion. Cancer-related fatigue may be influenced by multiple factors, including demographic, medical, cognitive/emotional, behavioral, and biological factors. In particular, growing evidence suggests an inflammatory basis for cancer-related fatigue, and studies have documented an association between elevated inflammatory processes and fatigue in patients before, during, and after treatment. The evidence linking inflammation and fatigue in cancer survivors is particularly strong, with consistent findings emerging from large, well-controlled studies of breast cancer survivors. Other biological processes that may influence fatigue include alterations in the neuroendocrine and immune systems, which are closely tied to inflammatory activity.

There is considerable variability in the experience of fatigue before, during, and after treatment, indicating that some patients may be particularly vulnerable to this symptom. Longitudinal studies have begun to illuminate risk factors for cancer-related fatigue, including depression, sleep disturbance, physical inactivity, and dysfunctional expectations and beliefs about fatigue. In addition, preliminary evidence indicates that variations in inflammation-related genes may increase risk for fatigue, suggesting a genetic contribution. Of note, the variability in fatigue is not closely tied to cancer treatment; patients who receive similar types of treatment may experience very different levels of fatigue, particularly in the post-treatment period.

A variety of different intervention approaches have been used to treat cancer-related fatigue. Physical activity is among the most promising approaches, and randomized controlled trials have documented beneficial effects of exercise during and after treatment. However, because these trials have not specifically focused on fatigued patients (i.e., presence of fatigue was not used as an inclusion criteria), the feasibility and efficacy of physical activity for patients with moderate to severe fatigue is unclear. Other psychosocial and mind-body interventions have targeted fatigued patients and shown beneficial effects. These include cognitive behavioral approaches, mindfulness, yoga, and acupuncture. Despite interest in psychostimulants such as methylphenidate, the evidence for these agents is quite mixed and recent guidelines do not recommend their use in post-treatment survivors169.

After two decades of research on cancer-related fatigue, we have a good understanding of the characteristics, prevalence, and course of this symptom and are beginning to elucidate mechanisms, risk factors, and effective treatments. We also have a growing appreciation of the complexity of this symptom, which shows significant inter-individual variability in its severity and expression. To advance our understanding of cancer-related fatigue, and particularly the variability in its experience and expression, the next generation of research must address a few key questions: Who is at risk for fatigue, and why? What are the mechanisms that underlie fatigue during and after treatment? To answer these questions, longitudinal studies are required that track patients before, during, and after treatment and include comprehensive assessment of biobehavioral risk factors. Together with appropriate statistical techniques (e.g., multilevel modeling, latent growth mixture modeling), this longitudinal approach will facilitate the identification of distinct trajectories of fatigue, and associated risk factors. These studies should also include in-depth assessment of underlying mechanisms, which can be used to direct intervention efforts; this is particularly important if the risk factors themselves are not amenable to intervention (e.g., genetic risk factors). Further, determination of factors that influence fatigue onset vs. persistence may be helpful in determining which type of interventions may be most helpful during vs. after treatment. Studies should also examine the co-occurrence of fatigue and related symptoms to elucidate the complex interactions between them, including depression and sleep disturbance. Finally, the degree to which cancer-related fatigue differs from normal age-related fatigue (and fatigue in other contexts) merits focused attention. Cancer and its treatment may accelerate age-related changes in inflammation, aerobic capacity, and other physiological processes, which may contribute to fatigue; thus, the fatigued cancer patient may look biologically “older” and potentially at greater risk for premature conditions of aging. There may also be different contributing factors for fatigue in older vs. younger patients, with implications for treatment.

The identification of underlying mechanisms should guide the development of targeted, individualized interventions for cancer-related fatigue, similar to current individualized approaches to cancer therapy. For example, patients whose fatigue appears to be primarily driven by dysfunctional coping strategies (e.g., catastrophizing) may be more responsive to cognitive-behavioral therapy approaches. In contrast, those whose fatigue is primarily driven by inflammatory activity may be more responsive to anti-inflammatory therapies (either behavioral or pharmacological). The importance of targeting the treatment to the underlying mechanism was illustrated in a recent trial evaluating the effect of the TNF antagonist infliximab for patients with treatment-resistant depression163. Results showed that infliximab was only effective for patients with elevated inflammatory markers at baseline. Similarly, anti-inflammatory approaches may be most efficacious for fatigued patients who show evidence of elevated inflammatory activity. Of note, even patients with more biologically driven fatigue (if there is such a group) may have developed dysfunctional cognitions and behaviors about their fatigue that are amenable to cognitive-behavioral intervention. Understanding the complexity of cancer-related fatigue, and using that understanding to identify vulnerable individuals and develop targeted, individualized interventions, is critical for reducing the burden of this symptom and improving quality of life and well-being in cancer patients and survivors.

References

  • 1.Lawrence DP, Kupelnick B, Miller K, Devine D, Lau J. Evidence report on the occurrence, assessment, and treatment of fatigue in cancer patients. J Natl Cancer Inst Monogr. 2004:40–50. doi: 10.1093/jncimonographs/lgh027. [DOI] [PubMed] [Google Scholar]
  • 2.Hickok JT, et al. Frequency, severity, clinical course, and correlates of fatigue in 372 patients during 5 weeks of radiotherapy for cancer. Cancer. 2005;104:1772–1778. doi: 10.1002/cncr.21364. [DOI] [PubMed] [Google Scholar]
  • 3.Jacobsen PB, et al. Fatigue in women receiving adjuvant chemotherapy for breast cancer: characteristics, course, and correlates. J Pain Symptom Manage. 1999;18:233–242. doi: 10.1016/s0885-3924(99)00082-2. [DOI] [PubMed] [Google Scholar]
  • 4.Phillips K, et al. Quality of life outcomes in patients with chronic myeloid leukemia treated with tyrosine kinase inhibitors: a controlled comparison. 2013;21:1097–1103. doi: 10.1007/s00520-012-1630-5. [DOI] [PubMed] [Google Scholar]
  • 5.Servaes P, Verhagen C, Bleijenberg G. Fatigue in cancer patients during and after treatment: prevalence, correlates and interventions. Eur J Cancer. 2002;38:27–43. doi: 10.1016/s0959-8049(01)00332-x. [DOI] [PubMed] [Google Scholar]
  • 6.Bower JE, et al. Fatigue in breast cancer survivors: occurrence, correlates, and impact on quality of life. J Clin Oncol. 2000;18:743–753. doi: 10.1200/JCO.2000.18.4.743. [DOI] [PubMed] [Google Scholar]
  • 7.Cella D, Davis K, Breitbart W, Curt G. Cancer-related fatigue: prevalence of proposed diagnostic criteria in a United States sample of cancer survivors. J Clin Oncol. 2001;19:3385–3391. doi: 10.1200/JCO.2001.19.14.3385. [DOI] [PubMed] [Google Scholar]
  • 8.Bower JE, et al. Fatigue in long-term breast carcinoma survivors: a longitudinal investigation. Cancer. 2006;106:751–758. doi: 10.1002/cncr.21671. [DOI] [PubMed] [Google Scholar]
  • 9.Servaes P, Gielissen MF, Verhagen S, Bleijenberg G. The course of severe fatigue in disease-free breast cancer patients: a longitudinal study. Psychooncology. 2006;16:787–795. doi: 10.1002/pon.1120. [DOI] [PubMed] [Google Scholar]
  • 10.Andrykowski MA, Curran SL, Lightner R. Off-treatment fatigue in breast cancer survivors: a controlled comparison. J Behav Med. 1998;21:1–18. doi: 10.1023/a:1018700303959. [DOI] [PubMed] [Google Scholar]
  • 11.Broeckel JA, Jacobsen PB, Horton J, Balducci L, Lyman GH. Characteristics and correlates of fatigue after adjuvant chemotherapy for breast cancer. J Clin Oncol. 1998;16:1689–1696. doi: 10.1200/JCO.1998.16.5.1689. [DOI] [PubMed] [Google Scholar]
  • 12.Curt GA, et al. Impact of cancer-related fatigue on the lives of patients: new findings from the Fatigue Coalition. Oncologist. 2000;5:353–360. doi: 10.1634/theoncologist.5-5-353. [DOI] [PubMed] [Google Scholar]
  • 13.Groenvold M, et al. Psychological distress and fatigue predicted recurrence and survival in primary breast cancer patients. Breast Cancer Res Treat. 2007;105:209–219. doi: 10.1007/s10549-006-9447-x. [DOI] [PubMed] [Google Scholar]
  • 14.Quinten C, et al. Patient self-reports of symptoms and clinician ratings as predictors of overall cancer survival. J Natl Cancer Inst. 2011;103:1851–1858. doi: 10.1093/jnci/djr485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Poulson MJ. Not just tired. J Clin Oncol. 2001;19:4180–4181. doi: 10.1200/JCO.2001.19.21.4180. [DOI] [PubMed] [Google Scholar]
  • 16.Cella D, Lai JS, Chang CH, Peterman A, Slavin M. Fatigue in cancer patients compared with fatigue in the general United States population. Cancer. 2002;94:528–538. doi: 10.1002/cncr.10245. [DOI] [PubMed] [Google Scholar]
  • 17.Forlenza MJ, Hall P, Lichtenstein P, Evengard B, Sullivan PF. Epidemiology of cancer-related fatigue in the Swedish twin registry. Cancer. 2005;104:2022–2031. doi: 10.1002/cncr.21373. [DOI] [PubMed] [Google Scholar]
  • 18.Vogelzang NJ, et al. Patient, caregiver, and oncologist perceptions of cancer-related fatigue: results of a tripart assessment survey. The Fatigue Coalition. Semin Hematol. 1997;34:4–12. [PubMed] [Google Scholar]
  • 19.Donovan KA, Small BJ, Andrykowski MA, Munster P, Jacobsen PB. Utility of a cognitive-behavioral model to predict fatigue following breast cancer treatment. Health Psychol. 2007;26:464–472. doi: 10.1037/0278-6133.26.4.464. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Mitchell SA. Cancer-related fatigue: state of the science. PM R. 2010;2:364–383. doi: 10.1016/j.pmrj.2010.03.024. [DOI] [PubMed] [Google Scholar]
  • 21.Barsevick A, Frost M, Zwinderman A, Hall P, Halyard M. I’m so tired: biological and genetic mechanisms of cancer-related fatigue. Qual Life Res. 2010;19:1419–1427. doi: 10.1007/s11136-010-9757-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Morrow GR, Andrews PL, Hickok JT, Roscoe JA, Matteson S. Fatigue associated with cancer and its treatment. Support Care Cancer. 2002;10:389–398. doi: 10.1007/s005200100293. [DOI] [PubMed] [Google Scholar]
  • 23.Dantzer R, O’Connor JC, Freund GG, Johnson RW, Kelley KW. From inflammation to sickness and depression: when the immune system subjugates the brain. Nat Rev Neurosci. 2008;9:46–56. doi: 10.1038/nrn2297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Haroon E, Raison CL, Miller AH. Psychoneuroimmunology meets neuropsychopharmacology: translational implications of the impact of inflammation on behavior. Neuropsychopharmacology. 2012;37:137–162. doi: 10.1038/npp.2011.205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Miller AH, Ancoli-Israel S, Bower JE, Capuron L, Irwin MR. Neuroendocrine-immune mechanisms of behavioral comorbidities in patients with cancer. J Clin Oncol. 2008;26:971–982. doi: 10.1200/JCO.2007.10.7805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Seruga B, Zhang H, Bernstein LJ, Tannock IF. Cytokines and their relationship to the symptoms and outcome of cancer. Nat Rev Cancer. 2008;8:887–899. doi: 10.1038/nrc2507. [DOI] [PubMed] [Google Scholar]
  • 27.Cleeland CS, et al. Are the symptoms of cancer and cancer treatment due to a shared biologic mechanism? A cytokine-immunologic model of cancer symptoms. Cancer. 2003;97:2919–2925. doi: 10.1002/cncr.11382. [DOI] [PubMed] [Google Scholar]
  • 28.Aggarwal BB, Vijayalekshmi RV, Sung B. Targeting inflammatory pathways for prevention and therapy of cancer: short-term friend, long-term foe. Clin Cancer Res. 2009;15:425–430. doi: 10.1158/1078-0432.CCR-08-0149. [DOI] [PubMed] [Google Scholar]
  • 29.Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420:860–867. doi: 10.1038/nature01322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Stone HB, Coleman CN, Anscher MS, McBride WH. Effects of radiation on normal tissue: consequences and mechanisms. The Lancet Oncology. 2003;4:529–536. doi: 10.1016/s1470-2045(03)01191-4. [DOI] [PubMed] [Google Scholar]
  • 31.Meyers CA, Albitar M, Estey E. Cognitive impairment, fatigue, and cytokine levels in patients with acute myelogenous leukemia or myelodysplastic syndrome. Cancer. 2005;104:788–793. doi: 10.1002/cncr.21234. [DOI] [PubMed] [Google Scholar]
  • 32.Clevenger L, et al. Sleep disturbance, cytokines, and fatigue in women with ovarian cancer. Brain Behav Immun. 2012 doi: 10.1016/j.bbi.2012.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Lutgendorf SK, et al. Interleukin-6, cortisol, and depressive symptoms in ovarian cancer patients. J Clin Oncol. 2008;26:4820–4827. doi: 10.1200/JCO.2007.14.1978. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Fagundes CP, et al. Fatigue and herpesvirus latency in women newly diagnosed with breast cancer. Brain Behav Immun. 2012;26:394–400. doi: 10.1016/j.bbi.2011.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Pertl MM, et al. C-reactive protein predicts fatigue independently of depression in breast cancer patients prior to chemotherapy. Brain Behav Immun. 2013;34:108–119. doi: 10.1016/j.bbi.2013.07.177. [DOI] [PubMed] [Google Scholar]
  • 36.Donovan KA, et al. Course of fatigue in women receiving chemotherapy and/or radiotherapy for early stage breast cancer. J Pain Symptom Manage. 2004;28:373–380. doi: 10.1016/j.jpainsymman.2004.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Arpin D, et al. Early variations of circulating interleukin-6 and interleukin-10 levels during thoracic radiotherapy are predictive for radiation pneumonitis. J Clin Oncol. 2005;23:8748–8756. doi: 10.1200/JCO.2005.01.7145. [DOI] [PubMed] [Google Scholar]
  • 38.Mills PJ, et al. The effects of standard anthracycline-based chemotherapy on soluble ICAM-1 and vascular endothelial growth factor levels in breast cancer. Clin Cancer Res. 2004;10:4998–5003. doi: 10.1158/1078-0432.CCR-0734-04. [DOI] [PubMed] [Google Scholar]
  • 39.Greenberg DB, Gray JL, Mannix CM, Eisenthal S, Carey M. Treatment-related fatigue and serum interleukin-1 levels in patients during external beam irradiation for prostate cancer. J Pain Symptom Manage. 1993;8:196–200. doi: 10.1016/0885-3924(93)90127-h. [DOI] [PubMed] [Google Scholar]
  • 40.Wratten C, et al. Fatigue during breast radiotherapy and its relationship to biological factors. Int J Radiat Oncol Biol Phys. 2004;59:160–167. doi: 10.1016/j.ijrobp.2003.10.008. [DOI] [PubMed] [Google Scholar]
  • 41.Geinitz H, et al. Fatigue, serum cytokine levels, and blood cell counts during radiotherapy of patients with breast cancer. Int J Radiat Oncol Biol Phys. 2001;51:691–698. doi: 10.1016/s0360-3016(01)01657-1. [DOI] [PubMed] [Google Scholar]
  • 42.Ahlberg K, Ekman T, Gaston-Johansson F. Levels of fatigue compared to levels of cytokines and hemoglobin during pelvic radiotherapy: a pilot study. Biol Res Nurs. 2004;5:203–210. doi: 10.1177/1099800403259500. [DOI] [PubMed] [Google Scholar]
  • 43.Bower JE, et al. Inflammatory biomarkers and fatigue during radiation therapy for breast and prostate cancer. Clin Cancer Res. 2009;15:5534–5540. doi: 10.1158/1078-0432.CCR-08-2584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Liu L, et al. Fatigue and sleep quality are associated with changes in inflammatory markers in breast cancer patients undergoing chemotherapy. Brain, Behavior, and Immunity. 2012;26:706–713. doi: 10.1016/j.bbi.2012.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Wang XS, et al. Inflammatory cytokines are associated with the development of symptom burden in patients with NSCLC undergoing concurrent chemoradiation therapy. Brain Behav Immun. 2010;24:968–974. doi: 10.1016/j.bbi.2010.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Wang XS, et al. Serum sTNF-R1, IL-6, and the development of fatigue in patients with gastrointestinal cancer undergoing chemoradiation therapy. Brain Behav Immun. 2012 doi: 10.1016/j.bbi.2011.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Wang XS, et al. Serum interleukin-6 predicts the development of multiple symptoms at nadir of allogeneic hematopoietic stem cell transplantation. Cancer. 2008;113:2102–2109. doi: 10.1002/cncr.23820. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Bower JE, Ganz PA, Aziz N, Fahey JL. Fatigue and proinflammatory cytokine activity in breast cancer survivors. Psychosom Med. 2002;64:604–611. doi: 10.1097/00006842-200207000-00010. [DOI] [PubMed] [Google Scholar]
  • 49.Collado-Hidalgo A, Bower JE, Ganz PA, Cole SW, Irwin MR. Inflammatory biomarkers for persistent fatigue in breast cancer survivors. Clin Cancer Res. 2006;12:2759–2766. doi: 10.1158/1078-0432.CCR-05-2398. [DOI] [PubMed] [Google Scholar]
  • 50.Bower JE, et al. Inflammatory responses to psychological stress in fatigued breast cancer survivors: relationship to glucocorticoids. Brain Behav Immun. 2007;21:251–258. doi: 10.1016/j.bbi.2006.08.001. [DOI] [PubMed] [Google Scholar]
  • 51.Bower JE, et al. Inflammation and behavioral symptoms after breast cancer treatment: do fatigue, depression, and sleep disturbance share a common underlying mechanism? J Clin Oncol. 2011;29:3517–3522. doi: 10.1200/JCO.2011.36.1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Alexander S, Minton O, Andrews P, Stone P. A comparison of the characteristics of disease-free breast cancer survivors with or without cancer-related fatigue syndrome. European Journal of Cancer. 2009;45:384–392. doi: 10.1016/j.ejca.2008.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Alfano CM, et al. Fatigue, Inflammation, and -ë-3 and -ë-6 Fatty Acid Intake Among Breast Cancer Survivors. Journal of Clinical Oncology. 2012 doi: 10.1200/JCO.2011.36.4109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Orre IJ, et al. Higher levels of fatigue are associated with higher CRP levels in disease-free breast cancer survivors. Journal of Psychosomatic Research. 2011;71:136–141. doi: 10.1016/j.jpsychores.2011.04.003. [DOI] [PubMed] [Google Scholar]
  • 55.Orre IJ, et al. Levels of circulating interleukin-1 receptor antagonist and C-reactive protein in long-term survivors of testicular cancer with chronic cancer-related fatigue. Brain, Behavior, and Immunity. 2009;23:868–874. doi: 10.1016/j.bbi.2009.04.003. [DOI] [PubMed] [Google Scholar]
  • 56.Schrepf A, et al. Cortisol and inflammatory processes in ovarian cancer patients following primary treatment: relationships with depression, fatigue, and disability. Brain Behav Immun. 2013;30(Suppl):S126–S134. doi: 10.1016/j.bbi.2012.07.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Bower JE, Ganz PA, Irwin MR, Arevalo JM, Cole SW. Fatigue and gene expression in human leukocytes: increased NF-kappaB and decreased glucocorticoid signaling in breast cancer survivors with persistent fatigue. Brain Behav Immun. 2011;25:147–150. doi: 10.1016/j.bbi.2010.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Landmark-Hoyvik H, et al. Alterations of gene expression in blood cells associated with chronic fatigue in breast cancer survivors. Pharmacogenomics J. 2009;9:333–340. doi: 10.1038/tpj.2009.27. [DOI] [PubMed] [Google Scholar]
  • 59.Light KC, et al. Differing leukocyte gene expression profiles associated with fatigue in patients with prostate cancer versus chronic fatigue syndrome. Psychoneuroendocrinology. 2013;38:2983–2995. doi: 10.1016/j.psyneuen.2013.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Saligan LN, et al. Upregulation of alpha-synuclein during localized radiation therapy signals the association of cancer-related fatigue with the activation of inflammatory and neuroprotective pathways. Brain Behav Immun. 2013;27:63–70. doi: 10.1016/j.bbi.2012.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Rotstein S, Blomgren H, Petrini B, Wasserman J, Baral E. Long term effects on the immune system following local radiation therapy for breast cancer. I. Cellular composition of the peripheral blood lymphocyte population. Int J Radiat Oncol Biol Phys. 1985;11:921–925. doi: 10.1016/0360-3016(85)90114-2. [DOI] [PubMed] [Google Scholar]
  • 62.Solomayer EF, et al. Influence of adjuvant hormone therapy and chemotherapy on the immune system analysed in the bone marrow of patients with breast cancer. Clin Cancer Res. 2003;9:174–180. [PubMed] [Google Scholar]
  • 63.Bower JE, Ganz PA, Aziz N, Fahey JL, Cole SW. T-cell homeostasis in breast cancer survivors with persistent fatigue. J Natl Cancer Inst. 2003;95:1165–1168. doi: 10.1093/jnci/djg0019. [DOI] [PubMed] [Google Scholar]
  • 64.Minton O, Stone PC. A comparison of cognitive function, sleep and activity levels in disease-free breast cancer patients with or without cancer-related fatigue syndrome. BMJ Support Palliat Care. 2013;2:231–238. doi: 10.1136/bmjspcare-2011-000172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Reinertsen K, et al. Predictors and course of chronic fatigue in long-term breast cancer survivors. Journal of Cancer Survivorship. 2010;4:405–414. doi: 10.1007/s11764-010-0145-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Glaser R, et al. Stress-associated changes in the steady-state expression of latent Epstein-Barr virus: implications for chronic fatigue syndrome and cancer. Brain Behav Immun. 2005;19:91–103. doi: 10.1016/j.bbi.2004.09.001. [DOI] [PubMed] [Google Scholar]
  • 67.Nazmi A, et al. The influence of persistent pathogens on circulating levels of inflammatory markers: a cross-sectional analysis from the Multi-Ethnic Study of Atherosclerosis. BMC Public Health. 2010;10:706. doi: 10.1186/1471-2458-10-706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Fagundes CP, Lindgren ME, Shapiro CL, Kiecolt-Glaser JK. Child maltreatment and breast cancer survivors: social support makes a difference for quality of life, fatigue and cancer stress. Eur J Cancer. 2012;48:728–736. doi: 10.1016/j.ejca.2011.06.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Kuo CP, et al. Detection of cytomegalovirus reactivation in cancer patients receiving chemotherapy. Clinical Microbiology and Infection. 2008;14:221–227. doi: 10.1111/j.1469-0691.2007.01895.x. [DOI] [PubMed] [Google Scholar]
  • 70.McEwen BS, et al. The role of adrenocorticoids as modulators of immune function in health and disease: neural, endocrine and immune interactions. Brain Res Brain Res Rev. 1997;23:79–133. doi: 10.1016/s0165-0173(96)00012-4. [DOI] [PubMed] [Google Scholar]
  • 71.Raison CL, Miller AH. When not enough is too much: the role of insufficient glucocorticoid signaling in the pathophysiology of stress-related disorders. Am J Psychiatry. 2003;160:1554–1565. doi: 10.1176/appi.ajp.160.9.1554. [DOI] [PubMed] [Google Scholar]
  • 72.Bower JE, et al. Diurnal cortisol rhythm and fatigue in breast cancer survivors. Psychoneuroendocrinology. 2005;30:92–100. doi: 10.1016/j.psyneuen.2004.06.003. [DOI] [PubMed] [Google Scholar]
  • 73.Bower JE, Ganz PA, Aziz N. Altered cortisol response to psychologic stress in breast cancer survivors with persistent fatigue. Psychosom Med. 2005;67:277–280. doi: 10.1097/01.psy.0000155666.55034.c6. [DOI] [PubMed] [Google Scholar]
  • 74.Weinrib AZ, et al. Diurnal cortisol dysregulation, functional disability, and depression in women with ovarian cancer. Cancer. 2010 doi: 10.1002/cncr.25299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Cole SW, et al. Social regulation of gene expression in human leukocytes. Genome Biol. 2007;8:R189. doi: 10.1186/gb-2007-8-9-r189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Miller GE, et al. A functional genomic fingerprint of chronic stress in humans: blunted glucocorticoid and increased NF-kappaB signaling. Biol Psychiatry. 2008;64:266–272. doi: 10.1016/j.biopsych.2008.03.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Fagundes CP, et al. Sympathetic and parasympathetic activity in cancer-related fatigue: more evidence for a physiological substrate in cancer survivors. Psychoneuroendocrinology. 2011;36:1137–1147. doi: 10.1016/j.psyneuen.2011.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Crosswell AD, Lockwood K, Bower JE. Low heart rate variability and cancer-related fatigue in breast cancer survivors. Psychoneuroendocrinology. 2014 doi: 10.1016/j.psyneuen.2014.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Irwin MR, Cole SW. Reciprocal regulation of the neural and innate immune systems. Nat Rev Immunol. 2011;11:625–632. doi: 10.1038/nri3042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Dimeo F, et al. Physical performance, depression, immune status and fatigue in patients with hematological malignancies after treatment. Ann Oncol. 2004;15:1237–1242. doi: 10.1093/annonc/mdh314. [DOI] [PubMed] [Google Scholar]
  • 81.de Raaf PJ, et al. Inflammation and fatigue dimensions in advanced cancer patients and cancer survivors: an explorative study. Cancer. 2012;118:6005–6011. doi: 10.1002/cncr.27613. [DOI] [PubMed] [Google Scholar]
  • 82.van Zuiden M, et al. Glucocorticoid sensitivity of leukocytes predicts PTSD, depressive and fatigue symptoms after military deployment: A prospective study. Psychoneuroendocrinology. 2012;37:1822–1836. doi: 10.1016/j.psyneuen.2012.03.018. [DOI] [PubMed] [Google Scholar]
  • 83.Dhruva A, et al. Trajectories of fatigue in patients with breast cancer before, during, and after radiation therapy. Cancer Nurs. 2010;33:201–212. doi: 10.1097/NCC.0b013e3181c75f2a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Aouizerat BE, et al. Preliminary evidence of a genetic association between tumor necrosis factor alpha and the severity of sleep disturbance and morning fatigue. Biol Res Nurs. 2009;11:27–41. doi: 10.1177/1099800409333871. [DOI] [PubMed] [Google Scholar]
  • 85.Miaskowski C, et al. Preliminary Evidence of an Association Between a Functional Interleukin-6 Polymorphism and Fatigue and Sleep Disturbance in Oncology Patients and Their Family Caregivers. J Pain Symptom Manage. 2010 doi: 10.1016/j.jpainsymman.2009.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Jim HS, et al. Genetic predictors of fatigue in prostate cancer patients treated with androgen deprivation therapy: Preliminary findings. Brain Behav Immun. 2012 doi: 10.1016/j.bbi.2012.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Reyes-Gibby CC, et al. Genetic variations in interleukin-8 and interleukin-10 are associated with pain, depressed mood, and fatigue in lung cancer patients. J Pain Symptom Manage. 2013;46:161–172. doi: 10.1016/j.jpainsymman.2012.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Rausch SM, et al. Relationship between cytokine gene single nucleotide polymorphisms and symptom burden and quality of life in lung cancer survivors. Cancer. 2010;116:4103–4113. doi: 10.1002/cncr.25255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Collado-Hidalgo A, Bower JE, Ganz PA, Irwin MR, Cole SW. Cytokine gene polymorphisms and fatigue in breast cancer survivors: Early findings. Brain Behav Immun. 2008 doi: 10.1016/j.bbi.2008.05.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Bower JE, et al. Cytokine genetic variations and fatigue among patients with breast cancer. J Clin Oncol. 2013;31:1656–1661. doi: 10.1200/JCO.2012.46.2143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Reinertsen KV, et al. Fatigued breast cancer survivors and gene polymorphisms in the inflammatory pathway. Brain, Behavior, and Immunity. 2011;25:1376–1383. doi: 10.1016/j.bbi.2011.04.001. [DOI] [PubMed] [Google Scholar]
  • 92.Carlo-Stella N, et al. A first study of cytokine genomic polymorphisms in CFS: Positive association of TNF-857 and IFNgamma 874 rare alleles. Clin Exp Rheumatol. 2006;24:179–182. [PubMed] [Google Scholar]
  • 93.Piraino B, Vollmer-Conna U, Lloyd AR. Genetic associations of fatigue and other symptom domains of the acute sickness response to infection. Brain, Behavior, and Immunity. 2012 doi: 10.1016/j.bbi.2011.12.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Stone P, Richards M, A’Hern R, Hardy J. Fatigue in patients with cancers of the breast or prostate undergoing radical radiotherapy. J Pain Symptom Manage. 2001;22:1007–1015. doi: 10.1016/s0885-3924(01)00361-x. [DOI] [PubMed] [Google Scholar]
  • 95.Goldstein D, et al. Cancer-related fatigue in women with breast cancer: outcomes of a 5-year prospective cohort study. J Clin Oncol. 2012;30:1805–1812. doi: 10.1200/JCO.2011.34.6148. [DOI] [PubMed] [Google Scholar]
  • 96.Goedendorp MM, Gielissen MF, Verhagen CA, Bleijenberg G. Development of fatigue in cancer survivors: a prospective follow-up study from diagnosis into the year after treatment. J Pain Symptom Manage. 2013;45:213–222. doi: 10.1016/j.jpainsymman.2012.02.009. [DOI] [PubMed] [Google Scholar]
  • 97.Geinitz H, et al. Fatigue in patients with adjuvant radiation therapy for breast cancer: long-term follow-up. J Cancer Res Clin Oncol. 2004;130:327–333. doi: 10.1007/s00432-003-0540-9. [DOI] [PubMed] [Google Scholar]
  • 98.Jacobsen PB, Donovan KA, Weitzner MA. Distinguishing fatigue and depression in patients with cancer. Semin Clin Neuropsychiatry. 2003;8:229–240. [PubMed] [Google Scholar]
  • 99.Miaskowski C, et al. Trajectories of Fatigue in Men with Prostate Cancer Before, During, and After Radiation Therapy. Journal of Pain and Symptom Management. 2008;35:632–643. doi: 10.1016/j.jpainsymman.2007.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Andrykowski MA, Schmidt JE, Salsman JM, Beacham AO, Jacobsen PB. Use of a case definition approach to identify cancer-related fatigue in women undergoing adjuvant therapy for breast cancer. J Clin Oncol. 2005;23:6613–6622. doi: 10.1200/JCO.2005.07.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Andrykowski MA, Donovan KA, Laronga C, Jacobsen PB. Prevalence, predictors, and characteristics of off-treatment fatigue in breast cancer survivors. Cancer. 2010 doi: 10.1002/cncr.25294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Ancoli-Israel S, Moore PJ, Jones V. The relationship between fatigue and sleep in cancer patients: a review. Eur J Cancer Care (Engl) 2001;10:245–255. doi: 10.1046/j.1365-2354.2001.00263.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Jim HS, et al. Lagged relationships among sleep disturbance, fatigue, and depressed mood during chemotherapy. Health Psychol. 2013;32:768–774. doi: 10.1037/a0031322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Berger AM. Patterns of fatigue and activity and rest during adjuvant breast cancer chemotherapy. Oncol Nurs Forum. 1998;25:51–62. [PubMed] [Google Scholar]
  • 105.Winters-Stone KM, Bennett JA, Nail L, Schwartz A. Strength, physical activity, and age predict fatigue in older breast cancer survivors. Oncol Nurs Forum. 2008;35:815–821. doi: 10.1188/08.ONF.815-821. [DOI] [PubMed] [Google Scholar]
  • 106.Neil SE, Klika RJ, Garland SJ, McKenzie DC, Campbell KL. Cardiorespiratory and neuromuscular deconditioning in fatigued and non-fatigued breast cancer survivors. 2013;21:873–881. doi: 10.1007/s00520-012-1600-y. [DOI] [PubMed] [Google Scholar]
  • 107.Alfano CM, et al. Physical activity, long-term symptoms, and physical health-related quality of life among breast cancer survivors: a prospective analysis. J Cancer Surviv. 2007;1:116–128. doi: 10.1007/s11764-007-0014-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Jacobsen PB, Andrykowski MA, Thors CL. Relationship of catastrophizing to fatigue among women receiving treatment for breast cancer. J Consult Clin Psychol. 2004;72:355–361. doi: 10.1037/0022-006X.72.2.355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Montgomery GH, Schnur JB, Erblich J, Diefenbach MA, Bovbjerg DH. Presurgery psychological factors predict pain, nausea, and fatigue one week after breast cancer surgery. J Pain Symptom Manage. 2010;39:1043–1052. doi: 10.1016/j.jpainsymman.2009.11.318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Bower JE, Crosswell AD, Slavich GM. Childhood Adversity and Cumulative Life Stress: Risk Factors for Cancer-Related Fatigue. Clin Psychol Sci. 2014;2 doi: 10.1177/2167702613496243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Fagundes CP, Lindgren ME, Shapiro CL, Kiecolt-Glaser JK. Child maltreatment and breast cancer survivors: Social support makes a difference for quality of life, fatigue and cancer stress. Eur J Cancer. 2011 doi: 10.1016/j.ejca.2011.06.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Heim C, et al. Childhood trauma and risk for chronic fatigue syndrome: association with neuroendocrine dysfunction. Arch Gen Psychiatry. 2009;66:72–80. doi: 10.1001/archgenpsychiatry.2008.508. [DOI] [PubMed] [Google Scholar]
  • 113.Heim C, et al. Early adverse experience and risk for chronic fatigue syndrome: results from a population-based study. Arch Gen Psychiatry. 2006;63:1258–1266. doi: 10.1001/archpsyc.63.11.1258. [DOI] [PubMed] [Google Scholar]
  • 114.McCauley J, et al. Clinical characteristics of women with a history of childhood abuse: unhealed wounds. JAMA. 1997;277:1362–1368. [PubMed] [Google Scholar]
  • 115.Jaremka LM, et al. Pain, Depression, and Fatigue: Loneliness as a Longitudinal Risk Factor. Health Psychol. 2013 doi: 10.1037/a0034012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Pace TW, et al. Increased stress-induced inflammatory responses in male patients with major depression and increased early life stress. Am J Psychiatry. 2006;163:1630–1633. doi: 10.1176/ajp.2006.163.9.1630. [DOI] [PubMed] [Google Scholar]
  • 117.Carpenter LL, et al. Association between plasma IL-6 response to acute stress and early-life adversity in healthy adults. Neuropsychopharmacology. 2010;35:2617–2623. doi: 10.1038/npp.2010.159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Cramp F, Byron-Daniel J. Exercise for the management of cancer-related fatigue in adults. Cochrane Database Syst Rev. 2012;11:CD006145. doi: 10.1002/14651858.CD006145.pub3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Mishra SI, et al. Exercise interventions on health-related quality of life for cancer survivors. Cochrane Database Syst Rev. 2012;8:CD007566. doi: 10.1002/14651858.CD007566.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Mishra SI, et al. Exercise interventions on health-related quality of life for people with cancer during active treatment. Cochrane Database Syst Rev. 2012;8:CD008465. doi: 10.1002/14651858.CD008465.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Puetz TW, Herring MP. Differential effects of exercise on cancer-related fatigue during and following treatment: a meta-analysis. Am J Prev Med. 2012;43:e1–e24. doi: 10.1016/j.amepre.2012.04.027. [DOI] [PubMed] [Google Scholar]
  • 122.Brown JC, et al. Efficacy of exercise interventions in modulating cancer-related fatigue among adult cancer survivors: a meta-analysis. Cancer Epidemiol Biomarkers Prev. 2011;20:123–133. doi: 10.1158/1055-9965.EPI-10-0988. [DOI] [PubMed] [Google Scholar]
  • 123.Speck RM, Courneya KS, Masse LC, Duval S, Schmitz KH. An update of controlled physical activity trials in cancer survivors: a systematic review and meta-analysis. J Cancer Surviv. 2010;4:87–100. doi: 10.1007/s11764-009-0110-5. [DOI] [PubMed] [Google Scholar]
  • 124.Strasser B, Steindorf K, Wiskemann J, Ulrich CM. Impact of resistance training in cancer survivors: a meta-analysis. Med Sci Sports Exerc. 2013;45:2080–2090. doi: 10.1249/MSS.0b013e31829a3b63. [DOI] [PubMed] [Google Scholar]
  • 125.Pinto BM, Frierson GM, Rabin C, Trunzo JJ, Marcus BH. Home-based physical activity intervention for breast cancer patients. J Clin Oncol. 2005;23:3577–3587. doi: 10.1200/JCO.2005.03.080. [DOI] [PubMed] [Google Scholar]
  • 126.Courneya KS, et al. Randomized controlled trial of the effects of aerobic exercise on physical functioning and quality of life in lymphoma patients. J Clin Oncol. 2009;27:4605–4612. doi: 10.1200/JCO.2008.20.0634. [DOI] [PubMed] [Google Scholar]
  • 127.Schmitz KH, et al. American College of Sports Medicine roundtable on exercise guidelines for cancer survivors. Med Sci Sports Exerc. 2010;42:1409–1426. doi: 10.1249/MSS.0b013e3181e0c112. [DOI] [PubMed] [Google Scholar]
  • 128.Courneya KS, et al. Three independent factors predicted adherence in a randomized controlled trial of resistance exercise training among prostate cancer survivors. J Clin Epidemiol. 2004;57:571–579. doi: 10.1016/j.jclinepi.2003.11.010. [DOI] [PubMed] [Google Scholar]
  • 129.Moyer A, Sohl SJ, Knapp-Oliver SK, Schneider S. Characteristics and methodological quality of 25 years of research investigating psychosocial interventions for cancer patients. Cancer Treatment Reviews. 2009;35:475–484. doi: 10.1016/j.ctrv.2009.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Jacobsen PB, Donovan KA, Vadaparampil ST, Small BJ. Systematic review and meta-analysis of psychological and activity-based interventions for cancer-related fatigue. Health Psychol. 2007;26:660–667. doi: 10.1037/0278-6133.26.6.660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Kangas M, Bovbjerg DH, Montgomery GH. Cancer-related fatigue: a systematic and meta-analytic review of non-pharmacological therapies for cancer patients. Psychol Bull. 2008;134:700–741. doi: 10.1037/a0012825. [DOI] [PubMed] [Google Scholar]
  • 132.Goedendorp MM, Gielissen MF, Verhagen CA, Bleijenberg G. Psychosocial interventions for reducing fatigue during cancer treatment in adults. Cochrane Database Syst Rev. 2009:CD006953. doi: 10.1002/14651858.CD006953.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Yates P, et al. Randomized controlled trial of an educational intervention for managing fatigue in women receiving adjuvant chemotherapy for early-stage breast cancer. J Clin Oncol. 2005;23:6027–6036. doi: 10.1200/JCO.2005.01.271. [DOI] [PubMed] [Google Scholar]
  • 134.Armes J, Chalder T, Addington-Hall J, Richardson A, Hotopf M. A randomized controlled trial to evaluate the effectiveness of a brief, behaviorally oriented intervention for cancer-related fatigue. Cancer. 2007;110:1385–1395. doi: 10.1002/cncr.22923. [DOI] [PubMed] [Google Scholar]
  • 135.Montgomery GH, et al. Fatigue during breast cancer radiotherapy: an initial randomized study of cognitive-behavioral therapy plus hypnosis. Health Psychol. 2009;28:317–322. doi: 10.1037/a0013582. [DOI] [PubMed] [Google Scholar]
  • 136.Stanton AL, et al. Outcomes from the Moving Beyond Cancer psychoeducational, randomized, controlled trial with breast cancer patients. J Clin Oncol. 2005;23:6009–6018. doi: 10.1200/JCO.2005.09.101. [DOI] [PubMed] [Google Scholar]
  • 137.Fillion L, et al. A brief intervention for fatigue management in breast cancer survivors. Cancer Nurs. 2008;31:145–159. doi: 10.1097/01.NCC.0000305698.97625.95. [DOI] [PubMed] [Google Scholar]
  • 138.Gielissen MF, Verhagen S, Witjes F, Bleijenberg G. Effects of cognitive behavior therapy in severely fatigued disease-free cancer patients compared with patients waiting for cognitive behavior therapy: a randomized controlled trial. J Clin Oncol. 2006;24:4882–4887. doi: 10.1200/JCO.2006.06.8270. [DOI] [PubMed] [Google Scholar]
  • 139.Gielissen MF, Verhagen CA, Bleijenberg G. Cognitive behaviour therapy for fatigued cancer survivors: long-term follow-up. Br J Cancer. 2007;97:612–618. doi: 10.1038/sj.bjc.6603899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Yun YH, et al. Web-based tailored education program for disease-free cancer survivors with cancer-related fatigue: a randomized controlled trial. J Clin Oncol. 2012;30:1296–1303. doi: 10.1200/JCO.2011.37.2979. [DOI] [PubMed] [Google Scholar]
  • 141.Ledesma D, Kumano H. Mindfulness-based stress reduction and cancer: a meta-analysis. Psychooncology. 2009;18:571–579. doi: 10.1002/pon.1400. [DOI] [PubMed] [Google Scholar]
  • 142.Zainal NZ, Booth S, Huppert FA. The efficacy of mindfulness-based stress reduction on mental health of breast cancer patients: a meta-analysis. Psychooncology. 2013;22:1457–1465. doi: 10.1002/pon.3171. [DOI] [PubMed] [Google Scholar]
  • 143.Lin KY, Hu YT, Chang KJ, Lin HF, Tsauo JY. Effects of yoga on psychological health, quality of life, and physical health of patients with cancer: a meta-analysis. Evid Based Complement Alternat Med. 2011;2011:659876. doi: 10.1155/2011/659876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Molassiotis A, et al. Acupuncture for cancer-related fatigue in patients with breast cancer: a pragmatic randomized controlled trial. J Clin Oncol. 2012;30:4470–4476. doi: 10.1200/JCO.2012.41.6222. [DOI] [PubMed] [Google Scholar]
  • 145.Molassiotis A, Sylt P, Diggins H. The management of cancer-related fatigue after chemotherapy with acupuncture and acupressure: a randomised controlled trial. Complement Ther Med. 2007;15:228–237. doi: 10.1016/j.ctim.2006.09.009. [DOI] [PubMed] [Google Scholar]
  • 146.Deng G, et al. Acupuncture for the treatment of post-chemotherapy chronic fatigue: a randomized, blinded, sham-controlled trial. 2013;21:1735–1741. doi: 10.1007/s00520-013-1720-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.van der Lee ML, Garssen B. Mindfulness-based cognitive therapy reduces chronic cancer-related fatigue: a treatment study. Psychooncology. 2012;21:264–272. doi: 10.1002/pon.1890. [DOI] [PubMed] [Google Scholar]
  • 148.Bower JE, et al. Yoga for persistent fatigue in breast cancer survivors: a randomized controlled trial. Cancer. 2012;118:3766–3775. doi: 10.1002/cncr.26702. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Bower JE, et al. Yoga reduces inflammatory signaling in fatigued breast cancer survivors: A randomized controlled trial. Psychoneuroendocrinology. 2014 doi: 10.1016/j.psyneuen.2014.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Jain S, et al. Complementary medicine for fatigue and cortisol variability in breast cancer survivors: A Randomized Controlled Trial. Cancer. 2011 doi: 10.1002/cncr.26345. [DOI] [PubMed] [Google Scholar]
  • 151.Chandwani KD, et al. Yoga improves quality of life and benefit finding in women undergoing radiotherapy for breast cancer. J Soc Integr Oncol. 2010;8:43–55. [PubMed] [Google Scholar]
  • 152.Minton O, Richardson A, Sharpe M, Hotopf M, Stone P. A systematic review and meta-analysis of the pharmacological treatment of cancer-related fatigue. J Natl Cancer Inst. 2008;100:1155–1166. doi: 10.1093/jnci/djn250. [DOI] [PubMed] [Google Scholar]
  • 153.Stockler MR, et al. Effect of sertraline on symptoms and survival in patients with advanced cancer, but without major depression: a placebo-controlled double-blind randomised trial. Lancet Oncol. 2007;8:603–612. doi: 10.1016/S1470-2045(07)70148-1. [DOI] [PubMed] [Google Scholar]
  • 154.Yennurajalingam S, et al. Reduction of cancer-related fatigue with dexamethasone: a double-blind, randomized, placebo-controlled trial in patients with advanced cancer. J Clin Oncol. 2013;31:3076–82. doi: 10.1200/JCO.2012.44.4661. [DOI] [PubMed] [Google Scholar]
  • 155.Minton O, Richardson A, Sharpe M, Hotopf M, Stone PC. Psychostimulants for the Management of Cancer-Related Fatigue: A Systematic Review and Meta-Analysis. J Pain Symptom Manage. 2011;41:761–767. doi: 10.1016/j.jpainsymman.2010.06.020. [DOI] [PubMed] [Google Scholar]
  • 156.Lower EE, et al. Efficacy of dexmethylphenidate for the treatment of fatigue after cancer chemotherapy: a randomized clinical trial. J Pain Symptom Manage. 2009;38:650–662. doi: 10.1016/j.jpainsymman.2009.03.011. [DOI] [PubMed] [Google Scholar]
  • 157.Bruera E, et al. Methylphenidate and/or a nursing telephone intervention for fatigue in patients with advanced cancer: a randomized, placebo-controlled, phase II trial. J Clin Oncol. 2013;31:2421–2427. doi: 10.1200/JCO.2012.45.3696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Moraska AR, et al. Phase III, randomized, double-blind, placebo-controlled study of long-acting methylphenidate for cancer-related fatigue: North Central Cancer Treatment Group NCCTG-N05C7 trial. J Clin Oncol. 2010;28:3673–3679. doi: 10.1200/JCO.2010.28.1444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Jean-Pierre P, et al. A phase 3 randomized, placebo-controlled, double-blind, clinical trial of the effect of modafinil on cancer-related fatigue among 631 patients receiving chemotherapy: a University of Rochester Cancer Center Community Clinical Oncology Program Research base study. Cancer. 2010;116:3513–3520. doi: 10.1002/cncr.25083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Monk JP, et al. Assessment of tumor necrosis factor alpha blockade as an intervention to improve tolerability of dose-intensive chemotherapy in cancer patients. J Clin Oncol. 2006;24:1852–1859. doi: 10.1200/JCO.2005.04.2838. [DOI] [PubMed] [Google Scholar]
  • 161.Tookman AJ, Jones CL, DeWitte M, Lodge PJ. Fatigue in patients with advanced cancer: a pilot study of an intervention with infliximab. 2008;16:1131–1140. doi: 10.1007/s00520-008-0429-x. [DOI] [PubMed] [Google Scholar]
  • 162.Tyring S, et al. Etanercept and clinical outcomes, fatigue, and depression in psoriasis: double-blind placebo-controlled randomised phase III trial. The Lancet. 2006;367:29–35. doi: 10.1016/S0140-6736(05)67763-X. [DOI] [PubMed] [Google Scholar]
  • 163.Raison CL, et al. A randomized controlled trial of the tumor necrosis factor antagonist infliximab for treatment-resistant depression: the role of baseline inflammatory biomarkers. JAMA Psychiatry. 2013;70:31–41. doi: 10.1001/2013.jamapsychiatry.4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Cruciani RA, et al. L-carnitine supplementation for the management of fatigue in patients with cancer: an eastern cooperative oncology group phase III, randomized, double-blind, placebo-controlled trial. J Clin Oncol. 2012;30:3864–3869. doi: 10.1200/JCO.2011.40.2180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Barton DL, et al. Wisconsin Ginseng (Panax quinquefolius) to improve cancer-related fatigue: a randomized, double-blind trial, N07C2. J Natl Cancer Inst. 2013;105:1230–1238. doi: 10.1093/jnci/djt181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Plaisance EP, Grandjean PW. Physical activity and high-sensitivity C-reactive protein. Sports Med. 2006;36:443–458. doi: 10.2165/00007256-200636050-00006. [DOI] [PubMed] [Google Scholar]
  • 167.Creswell JD, et al. Mindfulness-Based Stress Reduction training reduces loneliness and pro-inflammatory gene expression in older adults: a small randomized controlled trial. Brain Behav Immun. 2012;26:1095–1101. doi: 10.1016/j.bbi.2012.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Antoni MH, et al. Cognitive-behavioral stress management reverses anxiety-related leukocyte transcriptional dynamics. Biol Psychiatry. 2012;71:366–372. doi: 10.1016/j.biopsych.2011.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Bower JE, et al. Screening, Assessment and Management of Fatigue in Adult Survivors of Cancer: An American Society of Clinical Oncology Clinical Practice Guideline Adaptation. J Clin Oncol. 2014 doi: 10.1200/JCO.2013.53.4495. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Minton O, Stone P. A systematic review of the scales used for the measurement of cancer-related fatigue (CRF) Annals of Oncology. 2009;20:17–25. doi: 10.1093/annonc/mdn537. [DOI] [PubMed] [Google Scholar]
  • 171.Mendoza TR, et al. The rapid assessment of fatigue severity in cancer patients: use of the Brief Fatigue Inventory. Cancer. 1999;85:1186–1196. doi: 10.1002/(sici)1097-0142(19990301)85:5<1186::aid-cncr24>3.0.co;2-n. [DOI] [PubMed] [Google Scholar]
  • 172.Cella D, Peterman A, Passik S, Jacobsen P, Breitbart W. Progress toward guidelines for the management of fatigue. Oncology (Huntingt) 1998;12:369–377. [PubMed] [Google Scholar]
  • 173.Donovan KA, McGinty HL, Jacobsen PB. A systematic review of research using the diagnostic criteria for cancer-related fatigue. Psychooncology. 2013;22:737–744. doi: 10.1002/pon.3085. [DOI] [PubMed] [Google Scholar]
  • 174.Miller AH, Haroon E, Raison CL, Felger JC. CYTOKINE TARGETS IN THE BRAIN: IMPACT ON NEUROTRANSMITTERS AND NEUROCIRCUITS. Depression and Anxiety. 2013;30:297–306. doi: 10.1002/da.22084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Dantzer R, Kelley KW. Twenty years of research on cytokine-induced sickness behavior. Brain Behav Immun. 2007;21:153–160. doi: 10.1016/j.bbi.2006.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Capuron L, Ravaud A, Dantzer R. Early depressive symptoms in cancer patients receiving interleukin 2 and/or interferon alfa-2b therapy. J Clin Oncol. 2000;18:2143–2151. doi: 10.1200/JCO.2000.18.10.2143. [DOI] [PubMed] [Google Scholar]
  • 177.Kirkwood J. Cancer immunotherapy: the interferon-alpha experience. Semin Oncol. 2002;29:18–26. doi: 10.1053/sonc.2002.33078. [DOI] [PubMed] [Google Scholar]
  • 178.Valentine AD, Meyers CA, Kling MA, Richelson E, Hauser P. Mood and cognitive side effects of interferon-alpha therapy. Semin Oncol. 1998;25:39–47. [PubMed] [Google Scholar]
  • 179.Reichenberg A, et al. Cytokine-associated emotional and cognitive disturbances in humans. Arch Gen Psychiatry. 2001;58:445–452. doi: 10.1001/archpsyc.58.5.445. [DOI] [PubMed] [Google Scholar]
  • 180.Spath-Schwalbe E, et al. Acute effects of recombinant human interleukin-6 on endocrine and central nervous sleep functions in healthy men. J Clin Endocrinol Metab. 1998;83:1573–1579. doi: 10.1210/jcem.83.5.4795. [DOI] [PubMed] [Google Scholar]
  • 181.Cho HJ, Kivimaki M, Bower JE, Irwin MR. Association of C-reactive protein and interleukin-6 with new-onset fatigue in the Whitehall II prospective cohort study. Psychol Med. 2013;43:1773–1783. doi: 10.1017/S0033291712002437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Cho HJ, Seeman TE, Bower JE, Kiefe CI, Irwin MR. Prospective Association Between C-Reactive Protein and Fatigue in the Coronary Artery Risk Development in Young Adults Study. Biol Psychiatry. 2009 doi: 10.1016/j.biopsych.2009.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES