Benefits Balanced with Harm

Erythropoiesis Stimulating Factors (ESA)

for Fatigue

Erythropoiesis-stimulating factors (ESAs) such as erythropoetin and its synthetic alternatives (darbopoetin, erythropoetin alpha) are essential hormones that control red blood cell production, and are mainly used in the treatment of anemia. The U.S. Food and Drug Administration has issued several warnings related to the use of ESAs; related concerns included risk of increased tumor growth, decreased survival time and cardiovascular side effects. The effects of erythropoietin on fatigue and cognitive impairment have been examined in patients with cancer in the setting of anemia.

Systematic Review/Meta-Analysis

Bohlius, J., Tonia, T., Nuesch, E., Juni, P., Fey, M.F., Egger, M., & Bernhard, J. (2014). Effects of erythropoiesis-stimulating agents on fatigue- and anaemia-related symptoms in cancer patients: Systematic review and meta-analyses of published and unpublished data. British Journal of Cancer, 111, 33–45. 

Purpose

STUDY PURPOSE: To critically evaluate and quantify the effects of erythropoiesis-stimulating agents (ESAs) on quality of life (QoL) in patients with cancer; to examine the effects of ESAs on patient-rated fatigue and anamia-related symptoms; and to identify groups of patients who may benefit most from treatment with ESAs
 
TYPE OF STUDY: Systematic review and meta-analysis

Search Strategy

DATABASES USED: Updated literature searches from previous meta-analyses on ESAs (Bohlius et al., 2006a, 2006b, 2009a, 2009b) in MEDLINE, Embase, Cochrane Central Register of Controlled Trials, and databases of conference proceedings for the years 2008 to January 2011; screened reference lists of relevant meta-analyses and clinical trials registries (US Clinical Trials Registry; International Standard Randomised Controlled Trial Number); information from study protocols and reports obtained from ESA manufacturers (Amgen, Thousand Oaks, CA, USA; Johnson & Johnson, New Brunswick, NJ, USA; Hoffmann-La Roche, Basel, Switzerland) and clinical study groups for a previous IPD meta-analysis (Bohlius et al, 2009a, b); QoL results in clinical trials registries (http://clinicaltrials.gov/; http://www.isrctn.org/).
 
KEYWORDS: Epoetin or darbepoetin and QoL, fatigue, anemia-related symptoms (not explicitly stated)
 
INCLUSION CRITERIA: Randomized controlled trials (RCTs) that compared epoetin or darbepoetin with a placebo or best standard of care, which assessed fatigue and anaemia-related symptoms in patients with cancer receiving or not receiving anticancer treatment; studies that prospectively evaluated QoL using a validated or generally accepted instrument with a planned sample size of > 50 participants per study arm or 100 participants in total; trials using different types of iron supplementation were included and evaluated in stratified analysis.
 
EXCLUSION CRITERIA: Trials with high-dose myeloablative chemotherapy regimens followed by stem cell transplantation, trials in patients with myelodysplastic syndromes and acute leukemia, and trials using ESAs for short-term preoperative treatment

Literature Evaluated

TOTAL REFERENCES RETRIEVED = 304 (2,018 documents initially identified; 1,714 excluded by screening; 246 excluded; 58 studies evaluated)
 
EVALUATION METHOD AND COMMENTS ON LITERATURE USED: Four reviewers worked in pairs and independently determined study eligibility. Data on study characteristics, study quality, and outcomes were extracted by one reviewer and checked for accuracy by another. Published and unpublished literature were used. 

Sample Characteristics

FINAL NUMBER STUDIES INCLUDED = 37 
 
TOTAL PATIENTS INCLUDED IN REVIEW = 10,581
 
SAMPLE RANGE ACROSS STUDIES = 45–1,379
 
KEY SAMPLE CHARACTERISTICS: Solid and hematologic malignancies; primarily receiving chemotherapy; primarily advanced disease; other demographics (e.g., age, gender, ethnicity) not stated

Phase of Care and Clinical Applications

PHASE OF CARE: Active antitumor treatment
 
APPLICATIONS: Elder care, palliative care

Results

ESAs provide a small but clinically important improvement in anaemia-related symptoms (Functional Assessment of Cancer Therapy–Anemia [FACT-An]), which was confirmed when the analysis was restricted to placebo-controlled RCTs in patients receiving chemotherapy. For fatigue-related symptoms (Functional Assessment of Cancer Therapy–Fatigue [FACT-F]), the overall effect did not reach the threshold for a clinically important difference (CID). For FACT-F, there was some evidence that treatment effects were above the threshold for a CID in RCTs in patients receiving chemotherapy with hemoglobin levels below 12 g/dl–1 at baseline and in RCTs stopping ESAs at hemoglobin levels above 13 g/dl–1.
 
However, these findings for FACT-F were not confirmed when we restricted the analysis to placebo-controlled RCTs in patients receiving chemotherapy.

Conclusions

Societies in the United States and Europe continue to have different recommendations regarding the use of ESAs during cancer treatment; for QoL in palliative settings, the harmful effects of ESAs should be balanced against potential benefits. There is no evidence to support their use in managing cancer-related fatigue.

Limitations

Greater than 90% of the studies were funded by pharmaceutical companies.

Nursing Implications

For patients receiving palliative care, there may be a role for ESAs in reducing transfusion use; there does not appear to be any role for ESAs in managing cancer-related fatigue.

Print

Eton, D.T., & Cella, D. (2011). Do erythropoietic-stimulating agents relieve fatigue? A review of reviews. Cancer Treatment and Research, 157, 181–194. 

Purpose

STUDY PURPOSE: To review findings of 10 systematic reviews of clinical trials of erythropoietic-stimulating agents (ESAs) in the five-year period spanning 2004–2008.

TYPE OF STUDY: Systematic review

Search Strategy

DATABASES USED: Sources of articles not described in the article.
 
KEYWORDS: Not described
 
INCLUSION CRITERIA: Published from 2004–2008 
 
EXCLUSION CRITERIA: Not described

Literature Evaluated

TOTAL REFERENCES RETRIEVED: 10

Sample Characteristics

  • FINAL NUMBER OF STUDIES INCLUDED = 10
  • TOTAL PATIENTS INCLUDED IN REVIEW =  86,374
  • SAMPLE RANGE ACROSS STUDIES: 1,949–21,378
  • KEY SAMPLE CHARACTERISTICS: Inference that all patients included in reviews were patients with cancer, but report did not actually specify

Phase of Care and Clinical Applications

PHASE OF CARE: Active treatment

Results

10 reviews of effects of ESA treatment on symptoms and quality of life (QOL) were reviewed.

Conclusions

  • (From authors) Overall evidence from studies seem to support  a symptom and QOL benefit from ESA treatment, which is unlikely to be due to chance alone.
  • (From reviewer) Quality of studies presented in review, variability of instruments and ESAs used to measure variables of interest, missing data, and lack of description of sample populations in studies limits sufficient evidence for empirical benefit from ESA treatment in patients with cancer

Limitations

  • Focus of review was on patient-reported symptoms and QOL outcomes versus all risks (safety, survival, thrombosis) and benefits (transfusion use, hematologic response) of ESA use.
  • Not all of the studies reviewed were placebo controlled or double blinded.
  • Increased risk of placebo effect when outcome of interest is patient self-report.
  • Many studies failed to report metrics needed for a more detailed analysis.
  • Substantial loss of data and lack of documentation of how missing data were handled.
  • Timing and conditions under which symptoms and QOL were measured were not described in many studies.

Nursing Implications

  • Focus future studies on description of conditions for the safe use of ESAs.
  • Determine empirically derived cutoff above which ESA therapy is contraindicated.
  • Conduct detailed analysis of secondary data sources (meta-analysis).
  • Need for inclusion of patient perspective of benefits and harms of ESA therapy.
  • Development of patient education materials about relative risks and benefits of ESA use or lack thereof.
Print

Minton, O., Richardson, A., Sharpe, M., Hotopf, M., & Stone, P. (2010). Drug therapy for the management of cancer-related fatigue. Cochrane Database of Systematic Reviews, 7, CD006704.

Purpose

To evaluate the effectiveness of pharmacologic interventions used for fatigue in patients with cancer

Search Strategy

Databases searched were PaPaS, Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, EMBASE, CINAHL, Dissertation Abstracts International (DAI), metaRegister of Controlled Trials (mRCT) (January 2007–October 2009). Journals searched were British Journal of Cancer, Journal of Clinical Oncology, Journal of Pain and Symptom Management, and Journal of Palliative Medicine. The reference lists of all articles were checked for additional studies. Conference abstracts also were searched.

Search keywords were neoplasms, bone marrow transplantation, cancer, carcinoma, tumour, adenocarcinoma, leukemia, lymphoma, malignant, radiotherapy, fatigue, tired, weary, weariness, exhausted, lack or loss or lost energy or vigor, apathy or lassitude or lethargy, or feeling drained, sleepy, or sluggish.

Studies were included in the review if they

  • Assessed drug therapy for the management of cancer-related fatigue (CRF) compared to placebo, usual care, or a nonpharmacologic intervention.
  • Were randomized, controlled trials (single-blind and open-label were allowed).
  • Included adult patients with a clinical diagnosis of cancer.

Literature Evaluated

This review was an update of a previous review. The updated search retrieved 647 additional references. Of those, six additional studies met the inclusion criteria. The final sample of studies included was 31.

Sample Characteristics

The review included 7,104 participants who received a drug intervention for CRF.

Results

Psychostimulants

  • Four trials examined methylphenidate, and one used dexamphetamine. These included 426 patients total.
  • Evidence existed of a significant effect on fatigue with methylphenidate over placebo, and evidence supported the use of psychostimulants in the treatment of CRF.
  • The standardized mean difference was positive, with a small effect and narrow confidence interval (CI) (total mean difference = –0.28; 95% CI [-0.48, -0.09]; Z = 2.83; p = 0.005).
  • Fatigue was measured with the Functional Assessment of Cancer Therapy-Fatigue (FACT-F) in all studies.

Erythropoietin and Darbepoetin

  • Eleven studies were combined in total and demonstrated a positive effect. The weighted mean difference of studies using the FACT-F outcome measure in erythropoietin gave a score of 4.33, which was a clinically significant difference. The conclusion was limited to patients with anemia who were undergoing chemotherapy. Greater improvement was more likely in those with lower hemoglobin levels.
  • In placebo-controlled trials of darbepoetin, the mean difference using the FACT-F score was -1.96, which was less than the minimally clinical significant difference.
  • Combined analysis for both agents gave a mean difference score of 3.75, which was clinically significant. 
  • Erythropoietin and darbepoetin cannot be recommended because of adverse events associated with these drugs.

Antidepressants/Paroxetine

  • Two studies using paroxetine and a trial using sertraline were analyzed. Analysis showed no benefit for the treatment of CRF.

Progestational Steroids

  • In studies that could be combined, no evidence existed to support continued use for the treatment of fatigue.
  • The clinical significance of results of ibandronate were unclear.
  • One study of etanercept during chemotherapy had statistically significant results, but the study had a small sample size and poor design. It was suggested that additional trials be conducted.
  • One study of donepezil showed no benefit over placebo.

Conclusions

Four trials of methylphenidate provided evidence for use that was supportive but associated with a small effect size in a dose of 10–20 mg per day. Serious adverse events were minimal; however, clinicians need to review contraindications before prescribing. Additional large-scale trials were suggested using methylphenidate to further evaluate use in CRF. Erythropoietin and darbepoetin can no longer be recommended for CRF because of increased adverse events associated with these drugs. No current evidence exists to support the use of steroids.

Limitations

  • Reviewers found major limitations in the reporting of trials and multiple methods of measuring outcomes.
  • Some outcomes in trials were not reported due to extensive missing data.
  • These findings point to the need for improved research reporting to meet Consolidated Standards of Reporting Trials (CONSORT) guidelines and the benefit that could be derived from use of consistent methods of measuring outcomes.
Print

Research Evidence Summaries

Auerbach, M., Silberstein, P. T., Webb, R. T., Averyanova, S., Ciuleanu, T. E., Shao, J., & Bridges, K. (2010). Darbepoetin alfa 300 or 500 µg once every 3 weeks with or without intravenous iron in patients with chemotherapy-induced anemia. American Journal of Hematology, 85, 655–663.

Study Purpose

To evaluate the safety and efficacy of darbepoetin alfa (DA) administration every three weeks at fixed doses with or without intravenous (IV) iron.

Intervention Characteristics/Basic Study Process

The study used 1:1:1:1 randomization to one of four groups, stratified by platinum versus nonplatinum. The primary endpoint was hemoglobin 11 g/dL or greater, with no iron deficiency. The secondary endpoint was incidence of transfusions and change in Functional Assessment of Cancer Therapy-Fatigue (FACT-F) score from baseline to end of study. DA was administered every three weeks, and dosages were reduced according to hemoglobin levels. Weekly, 400 µg of IV iron were given, and dosages were reduced as warranted based on ferritin levels. Patients were assigned to one of four groups with or without iron with DA at either 300 µg or 500 µg.

Sample Characteristics

  • The study included 128 patients (78 females, 40 males).
  • Age ranged from 27 to 97 years. Mean ages for the four arms were 61.7, 54.5, 64.5, and 61.8 years.
  • Patients had nonmyeloid malignancies.
  • Hemoglobin 10 or less
  • Patients had more than eight weeks of planned chemotherapy.

Setting

  • Multisite
  • North America and Europe (Romania and Russia)

Measurement Instruments/Methods

FACT-F

Results

DA at 300 µg and 500 µg given every three weeks demonstrated similar benefits, and added IV iron improved treatment response. There were larger increases in hemoglobin level, and increase in hemoglobin level occurred earlier. The proportions of patients with clinically significant increases in FACT-F score were 100% for the DA 300 µg group, 64% for the 500 µg DA group, 66% for those not receiving iron, and 100% for those receiving iron. No evidence existed of a statistically significant interaction between DA dose received and IV iron usage.

Conclusions

Fatigue and hemoglobin levels improved in patients with anemia receiving DA with and without iron.

Limitations

  • No appropriate control group was used.
  • Several anaphylactoid reactions to IV iron shed light on a safety issue.
  • Efficacy results were pooled and summarized because no evidence was observed of a statistically significant interaction between DA and IV iron usage.

Nursing Implications

DA cannot be recommended at any dose for cancer-related fatigue.

Print

Hoskin, P. J., Robinson, M., Slevin, N., Morgan, D., Harrington, K., & Gaffney, C. (2009). Effect of epoetin alfa on survival and cancer treatment-related anemia and fatigue in patients receiving radical radiotherapy with curative intent for head and neck cancer. Journal of Clinical Oncology, 27, 5751–5756.

Study Purpose

To evaluate the effect of epoetin alfa on local disease-free survival (DFS) (primary endpoint), overall survival, and cancer treatment-related anemia and fatigue (secondary endpoint).

Intervention Characteristics/Basic Study Process

Patients were randomized to one of the following three groups:

  • Radiation and epoetin alfa 10,0000 units subcutaneously (subq) if hemoglobin (Hgb) was less than 15 g/dL
  • Radiation and epoetin alfa 4,000 units subq if Hgb was less than 12.5 g/dL
  • Radiation alone.

Sample Characteristics

  • The study included 300 patients (232 men, 68 women); 111 completed the study.
  • Age ranged from 35 to 88 years.
  • Patients were diagnosed with squamous cell head and neck cancer, stages I to III.
  • Patients had a Hgb level less than 15 g/dL.

Setting

  • Multisite
  • United Kingdom

Study Design

The study was a phase III, randomized, controlled trial.

Measurement Instruments/Methods

  • Functional Assessment of Cancer Therapy-Anemia (FACT-An)
  • FACT-Head and Neck

Results

Hgb increased from baseline with epoetin alfa. Median duration of local DFS (primary endpoint) was not statistically significant (hazard ratio = 1.04). The groups did not differ significantly in terms of DFS, overall survival, tumor outcomes, or cancer-related anemia or fatigue.

Conclusions

The addition of epoetin alfa did not affect survival, tumor outcomes, anemia, or fatigue positively or negatively in the head and neck cancer population.

Limitations

  • The study had low accrual.
  • The degree of diagnostic severity, combined with the effects of radiation, may have minimized the potential benefit of epoetin alfa.

Nursing Implications

Epoetin alfa cannot be recommended for the treatment of cancer-related fatigue in the head and neck cancer population.

Print

Pronzato, P., Cortesi, E., van der Rijt, C. C., Bols, A., Moreno-Nogueira, J. A., de Oliveira, C. F., . . . EPO-INT-47 Study Group. (2010). Epoetin alfa improves anemia and anemia-related, patient-reported outcomes in patients with breast cancer receiving myelotoxic chemotherapy: results of a European, multicenter, randomized, controlled trial. Oncologist, 15, 935–943.

Study Purpose

To evaluate the effects of treatment with epoetin alfa on anemia-related, patient-reported outcomes in women with breast cancer receiving myelotoxic chemotherapy.

Intervention Characteristics/Basic Study Process

Women with a hemoglobin level of 12 g/dL or less were randomized 1:1 to receive epoetin alfa (10,000 IU three times weekly) or best standard care during chemotherapy.

Sample Characteristics

  • The study included 166 females (81 in the epoetin alfa group, 85 in the best supportive care group).
  • Mean patient age ranged from 53.3 to 54.3 years (mean range 27–77). 
  • Patients with breast cancer had a planned 12-week administration of chemotherapy.
  • Stage IV was the most common stage.

Setting

  • Multisite
  • Outpatient
  • Six European countries

Study Design

The study was a randomized, controlled phase IIIb trial.

Measurement Instruments/Methods

  • Functional Assessment of Cancer Therapy-Anemia (FACT-An)
  • Cancer Linear Analog Scale (CLAS) to assess patient-related outcomes

Results

On the FACT-An nonfatigue subscale, change was significantly better (p = 0.008) in the epoetin alfa group than in the best supportive care group. CLAS assessment showed that energy, ability to perform activities of daily living, and overall quality of life were significantly better (p = 0.007). No difference existed between groups in tumor response of survival. A significant increase was noted in transfusions in the best supportive care group (p = 0.048).

Conclusions

Epoetin alfa was well tolerated. Early administration in the treatment course attenuated deterioration in anemia-related, patient-reported outcomes.

Limitations

  • No actual direct fatigue measurement was used. 
  • A visual analog scale (VAS) was used for related measures, but no recognized definitive methods of measurement were used.

Nursing Implications

New U.S. Food and Drug Administration (FDA) guidelines exist for the administration of epoetin alfa.

Print

Guideline / Expert Opinion

National Comprehensive Cancer Network. (2011). NCCN Clinical Practice Guidelines in Oncology: Cancer-Related Fatigue. Version 1.2011.

Purpose & Patient Population

To ensure that all cancer patients with fatigue were identified and treated promptly and effectively.  These guidelines included recommended standards of care for assessment and management of fatigue in children, adolescents, and adults with cancer.

Type of Resource/Evidence-Based Process

The guidelines were evidence- and consensus-based. The guidelines were multidisciplinary, and all recommendations were category 2A unless otherwise stated.

Results Provided in the Reference

The guidelines provided several algorithms for assessment and management based on age group, level of self-reported fatigue, and phase of treatment.

Guidelines & Recommendations

Screening

  • All patients with cancer should be screened for the presence or absence of fatigue at regular intervals as a vital sign.
    • Age older than 12 years:  Screen on a 0-to-10 scale or as none, mild, moderate, or severe.
    • Age 7 to 12 years:   Use 1-to-5 scale (1 = no fatigue and 5 = worst).
    • Age 5 to 6 years:  Screen using “tired” or “not tired.”

Focused Evaluation of Fatigue

  • A focused history and assessment of contributing factors should be performed when screening indicates moderate to severe fatigue.
    • Age older than 12 years:  score of 4 to 10
    • Age 7 to 12 years:  score of 3 to 5
    • Age 5 to 6 years:  “tired”
  • Focused history should
    • Rule out recurrence or progression of cancer
    • Include a review of systems
    • Include an in depth fatigue history, including onset and patterns, associated/alleviating factors, and interference with function.
  • Assessment of treatable contributing factors, such as
    • Other related symptoms
    • Anemia
    • Sleep disturbance
    • Medication and side effects
    • Comorbidities
    • Activity and fitness level.

Management and Interventions

  • Active Treatment
    • Education and counseling regarding known patterns of fatigue and reassurance that treatment-related fatigue is not necessarily indicative of progression of disease.
    • General management strategies to include self-monitoring, energy conservation techniques, and use of distraction
    • Nonpharmacologic interventions to include activity enhancement, physically based therapies (such as massage), psychosocial interventions, nutritional consultation, and cognitive behavioral therapy for sleep
    • Pharmacologic interventions to include consider psychostimulants, treatment of anemia as indicated, and consideration of mediation for sleep
  • Posttreatment
    • Education and counseling about known fatigue patterns and self-monitoring of fatigue levels
    • General management and nonpharmacological and pharmacological interventions as for active treatment above
  • End of Life
    • Education and counseling about known fatigue patterns and as an expected end of life symptom
    • General strategies as per active treatment and post treatment
    • Nonpharmacologic interventions to include activity enhancement, psychosocial interventions, and nutrition consultation
    • Pharmacologic interventions as per active and post treatment

Within activity enhancement information, the guideline cites several synthesized reviews regarding the use of exercise and concludes that

  • Improvement in fatigue was not noted with all diagnoses.
  • It is reasonable to encourage all patients to engage in a moderate level of physical activity during and after cancer treatment.
  • Referral to exercise specialists or physical therapy should be triggered by
    • Patients with comorbid conditions, such as chronic obstructive pulmonary disease or cardiac disease
    • Recent major surgery
    • Specific functional or anatomical deficits
    • Substantial deconditioning.
  • Exercise should be used with caution in patients with
    • Bone metastases
    • Immunosuppression or neutropenia
    • Thrombocytopenia
    • Anemia
    • Fever or active infection
    • Limitations due to other illnesses.

Because fatigue is a subjective experience, it was recommended that assessment should use patient self-reports and other sources of data.

Several barriers were identified related to effective treatment for fatigue.  Due to barriers, it was stated that screening for fatigue needs to be emphasized.  Rescreening was emphasized because fatigue may exist beyond the period of active treatment.

Factors identified as potential causative agents that should be specifically assessed were outlined.  These factors were pain, emotional distress, sleep disturbance, anemia, nutrition, activity level, medication side effects, and other comorbidities.

It was noted that fatigue often occurs as part of a symptom cluster, often with sleep disturbance, emotional distress, or pain, so that assessment of these problems and institution of effective treatment is essential.

The importance of comprehensive assessment, including review of all current medications and noncancer comorbidities, was identified.  For example, it was noted that there can be thyroid dysfunction after radiation therapy for various cancers or use of biological and that hypogonadism can be associated with fatigue.

Limitations

  • The majority of studies regarding the impact of exercise on fatigue were performed in patients with limited types of cancer, and findings may not be applicable to all types of patients.  In addition, the timing and amount of exercise for various groups are not clear.  There are also few longitudinal studies examining fatigue in long-term disease-free survivors, although fatigue can be a long-term or late effect.
  • Although the guideline was structured according to phase of treatment, recommended interventions did not vary according to phase of treatment.  There were minimal differences in recommended content of education and counseling.
  • There was little evidence regarding effective management of fatigue in end of life care.
  • There was no discussion of prevention related to fatigue.
Print