Symptom Control in Palliative Care—Part II: Cachexia

Transcription

Symptom Control in Palliative Care—Part II: Cachexia
Palliative Care Reviews
JOURNAL OF PALLIATIVE MEDICINE
Volume 9, Number 2, 2006
© Mary Ann Liebert, Inc.
Feature Editors: Robert M. Arnold and Solomon Liao
Symptom Control in Palliative Care—Part II:
Cachexia/Anorexia and Fatigue
EGIDIO DEL FABBRO, M.D., SHALINI DALAL, M.D., and EDUARDO BRUERA, M.D.
INTRODUCTION
I
we addressed general symptom assessment, constipation, and chronic nausea. This part discusses cachexia/anorexia and fatigue, and once again most of the available
research is cancer-related.
N THE FIRST PART
CACHEXIA/ANOREXIA
Cachexia may occur in up to 80% of patients
with advanced cancer.1 This complex metabolic
syndrome, characterized by profound loss of lean
body mass, is the main cause of death in 20% of
patients. Cachexia (involuntary loss of more than
10% of premorbid weight) is a marker for poor
prognosis2 and is associated with anorexia (loss
of appetite), asthenia and changes in body image.
Cancer cachexia tends to occur in patients with
solid tumors, in children, and in elderly patients.
Cachexia is also common in acquired immune deficiency syndrome (AIDS), chronic obstructive
pulmonary disease (COPD), congestive heart failure (CHF)3–6 and other chronic illnesses, such as
dementia, tuberculosis, malaria,7 chronic kidney
disease, liver disease,8 and rheumatoid arthritis.
MECHANISMS
Cachexia is caused by a complex interaction between tumor by-products (proteolysis inducing
factor, lipid mobilizing factor) and host cytokines
(including interleukin [IL]-1, IL-6, tumor necrosis
factor, interferon). Cachexia in experimental animal models can be relieved by specific cytokine
antagonists, such as monoclonal antibodies to IL6.9 The metabolic abnormalities are characterized
by synthesis of acute phase proteins in the liver
at the expense of muscle protein, proteolysis,
lipolysis, insulin resistance, decreased lipogenesis, elevated triglycerides and decreased high
density lipoproteins (Fig. 1). Neuroendocrine abnormalities and the production of anorexigenic10
compounds may further contribute to the development of cachexia. Nutritional supplementation
and appetite stimulation alone cannot overcome
the block in accretion of muscle mass. Similar
mechanisms for cachexia in CHF, chronic kidney
disease,11 COPD,12 and rheumatoid arthritis13
have been described.
ASSESSMENT
The patient’s subjective loss of appetite can be
assessed with a numerical rating scale such as the
Edmonton Symptom Assessment System (ESAS),
or one of the other tools previously described.
The clinical assessment includes a careful history
that is focused on nutritional issues and a physical examination. Body weight is the only test
commonly used outside the research setting.
Measuring mid upper arm circumference may
have prognostic value.14 Whole-body impedence
and electroconductivity is based on the principle
that lean tissue conducts electricity better than fat.
This has the potential for more widespread clinical use as the equipment becomes more easily
Department of Palliative Care and Rehabilitation Medicine, University of Texas M. D. Anderson Cancer Center,
Houston, Texas.
409
410
DEL FABBRO ET AL.
FIG. 1.
Metabolic abnormalities in primary cachexia.
available. For research and for follow-up, the effect of any intervention on related symptoms,
such as fatigue and dyspnea, would be useful, as
well as quality of life and functional assessments.
MANAGEMENT
The management of major contributors to this
syndrome, such as chronic nausea, constipation,
early satiety, taste alteration, dyspnea, deconditioning, and depression may result in significant
improvement. Established pharmacologic treatment currently comprises either megestrol or corticosteroids, which predominantly stimulate appetite, however will not reverse cachexia in most
patients. Weight gain may be necessary to alleviate distress associated with changes in body image. The other therapies described below may
delay or reverse the cachexia component, or alleviate the symptom burden without major effects
on body composition.
Pharmacologic treatment
Progestational agents. Megestrol acetate has
demonstrated dose dependent improvements
(starting at 160 mg/d) in appetite, fatigue, and
general well-being in more than 60% of patients.15–17 Approximately 480–800 mg/d is optimal for weight gain, but it is prudent to start at
a lower dose and titrate upward, since adverse
effects18 and expense are dose-related. Symptomatic improvement in appetite occurs in less than
1 week; however, weight gain may take several
weeks and it happens in less than one quarter of
treated patients. In a comparison study, megesterol acetate (MA; 800 mg/d) was found to have
fewer side effects than dexamethasone (0.75 mg
four times daily [qid]), with a trend toward better weight gain.19 The authors concluded that for
planned long-term use (weeks to months) it
might be preferable to choose megestrol acetate.
Consideration should also be given to cost (up to
20 times more expensive) and the higher risk for
deep vein thrombosis (DVI).
A recent review of the literature on MA20
showed that this drug was safe and effective in
patients with cancer and AIDS. Megestrol has
also been used in other noncancer conditions. A
randomized double-blinded placebo controlled 8
week trial21 of either megestrol 800 mg daily or
placebo in underweight patients with COPD
demonstrated significant weight gain (mostly fat
mass) in the megestrol group. Ventilation was
stimulated (PaCO2 decreased), but there was no
change in the 6-minute walk test. Side effects of
megestrol include edema, hyperglycemia, and elevated liver enzymes.
Medroxyprogesterone22 at doses of 1000 mg/d
(equivalent to megestrol 160 mg/d) has demonstrated significant improvements in appetite and
body weight.
Corticosteroids may stimulate appetite and
decrease nausea as well.23,24 Randomized clinical trials have demonstrated that compared to
placebo, corticosteroids can improve appetite and
food intake.25 The effects of corticosteroids on ap-
SYMPTOM CONTROL: CACHEXIA, ANOREXIA, FATIGUE
petite and food intake are usually limited to a
couple of weeks, and the side effects of these
drugs increase dramatically over time. Therefore,
corticosteroids should be considered most useful
for patients with a life expectancy of less than 6
weeks. Most authors have used prednisone doses
ranging from 20–40 mg, or dexamethasone at an
equivalent dosage.
Cannabinoids. Dronabinol, a synthetic cannabinoid, is Food and Drug Administration (FDA) approved for anorexia related to AIDS and chemotherapy-induced nausea and emesis. In a study
of patients with AIDS-related cachexia, dronabinol showed significant improvement in nausea,
appetite, and mood but no weight gain.26 Although some studies found beneficial effects of
dronabinol on appetite in patients with advanced
cancer,27,28 the effects of dronabinol on cachexia
appear to be limited. In a more recent study comparing dronabinol with megesterol acetate, the
latter drug was found to be more beneficial as an
orexigenic agent.29 A combination of both drugs
was not better than MA alone. The use of dronabinol is also limited by undesirable central nervous side effects, such as sedation, confusion, and
perceptual disturbances.30
Thalidomide has immunomodulatory and antineoplastic effects and its use is being explored
in a variety of conditions.31 Recent trials in patients with advanced cancer have been encouraging. Thalidomide was found to improve appetite, nausea and well-being32 after 10 days
of treatment (100 mg daily) in patients with
advanced cancer. A recent randomized placebo
controlled trial33 of 50 patients with pancreatic
cancer demonstrated that a dose of 200 mg
daily for 24 weeks was well-tolerated and effective at attenuating loss of weight and lean body
mass. There was no significant difference in appetite symptom scores between thalidomide and
placebo. Two patients developed peripheral neuropathy that resolved on cessation of the drug.
Another open-label trial of thalidomide for 2
weeks in patients with esophageal cancer34 appeared to reverse loss of lean body mass, and although sedation was experienced by all patients,
daytime somnolence was transitory and disappeared after 2 or 3 days.
Thalidomide also appears to benefit patients
without cancer with cachexia. Patients with human immunodeficiency virus (HIV) with aph-
411
thous ulcers have demonstrated weight gain on
doses of 200 mg daily.35 Low-dose (100 mg)
thalidomide36 for 8 weeks in AIDS-associated
cachexia was also associated with significant
weight gain, half of which was fat-free mass. A
trial of thalidomide in patients with active pulmonary tuberculosis37 was well tolerated and resulted in significant weight gain and reduction of
tumor necrosis factor (TNF)-.
Testosterone and its derivatives, such as oxandrolone,38 have been studied in patients with
AIDS, neuromuscular disorders, and alcoholic
cirrhosis. A recent review of oxandrolone concluded that improvements in body composition
and muscle strength are significant in the majority of well-designed trials. Long-term safety ( 1
year) and dosage titration still need to be determined. A prospective open-label study39 demonstrated a preferential increase of lean body mass
in patients with COPD after 2 months of oxandrolone. There was an increase in 6 minute walking distance of more than 65 m in the majority of
patients. Eighteen percent of patients withdrew
because of side effects. Nandrolone has shown
similar benefits in patients with COPD40 and
AIDS, however, it can only be given parenterally
and this limits its use.
Other hormones, such as growth hormone,
have demonstrated increases in lean body mass
in patients with AIDS,41 and may have future applications in the syndrome of frailty42 in the elderly, but the cost is prohibitive. Ghrelin, a novel
growth hormone-releasing peptide isolated from
the stomach, may have future therapeutic potential for cachexia43 of different causes (cardiac or
cancer).
Melatonin may inhibit cancer cell growth, improve survival and decrease chemotherapy induced side effects. In two studies with a large
number of patients (total 1640), the frequency
of cachexia, asthenia, and thrombocytopenia was
significantly lower in patients treated with 20 mg
of melatonin at night.44 A randomized pilot trial
in patients with cancer of fish oil, melatonin, or
their combination did not induce major biochemical changes suggestive of a strong anticachectic effect, but may have produced a weightstabilizing effect.45 This well-tolerated drug
deserves further placebo-controlled trials.
-3 fish oil has few adverse effects and several
benefits, including an immunomodulatory action and an antidepressant effect. Despite its
412
promise,46 there was no nutritional or symptomatic benefit in two randomized double-blinded
controlled trials of advanced cancer patients.47,48
-Antagonists and agonists. The possible role of
the sympathetic nervous system in cardiac
cachexia was illustrated by a study49 that measured body weight, plasma norepinephrine, leptin, and insulin in both cachectic and noncachectic chronic heart failure patients. After 6 months
of -blocker therapy, patients with baseline
cachexia demonstrated significantly greater
weight gain, increase in leptin levels, and decrease in norepinephrine levels compared to noncachectic patients. The -agonist Clenbuterol has
an anabolic effect in normal and wasted muscles
of animals. A single double-blinded placebo controlled trial50 of healthy postoperative orthopedic
patients demonstrated increased muscle strength
in the group treated with clenbuterol.
Immunologic. A review51 of six clinical trials of
various monoclonal antibodies to IL-6 in the
treatment of cancer showed that the therapy was
well tolerated and decreased the cancer-related
symptoms of cachexia, pain, and fever. In most
patients C-reactive protein levels decreased below detectable limits. A recent trial52 of a low-cost
oral therapeutic HIV vaccine (V1) in patients with
AIDS prolonged survival and increased body
mass.
Psychotropics. Antidepressants, such as mirtazapine, and atypical antipsychotics, such as
olanzapine, may be useful for the management of
anorexia, nausea, and other symptoms in patients
with cancer.53 Tricyclics, such as amitriptylline,
may improve appetite but unfortunately have
side effects, especially in the elderly.
Nonpharmacologic treatment
Nutritional support and counseling. Those patients with a predominant starvation component
resulting from obstructive gastrointestinal tumors or painful mucositis as a result of head and
neck cancer treatment may benefit from nutritional interventions.
Artificial nutrition via the enteral or parenteral
routes is inappropriate for most patients with advanced cancer and may hinder the transition to
hospice. A meta-analysis54 of total parenteral nutrition (TPN) in patients with cancer undergoing
DEL FABBRO ET AL.
chemotherapy showed decreased survival and
increased susceptibility to infection. A review of
70 prospective randomized trials of enteral and
parenteral nutrition in cancer patients55 concluded that if there is any therapeutic benefit it is
small and confined to a small subset of patients.
In rare cases, home TPN can be associated with
long-term survival. A retrospective single institution study of home TPN56 identified 16 patients
with cancer over a 20-year period who survived
a year or longer. Most of the tumors were carcinoid and quality of life data were not assessed.
One recent randomized prospective study57 of
patients with primarily gastrointestinal tumors
suggested a trend toward increased survival in
patients who received TPN.
Counseling. Compassionate communication is required to address the benefits and burdens of nutrition and to reframe the condition from that of
“starving to death” to the more complex one of irreversible (usually) metabolic abnormalities. The
psychological component is also important to consider, because eating meals together is important
for social integration. Families of patients who are
able to eat should be advised that frequent small
meals may be more tolerable than three large meals
daily. Nutritional counseling has been reported to
improve nutritional intake in patients undergoing
chemotherapy but not to influence symptom distress or survival.58 In patients with advanced cancer, this increased nutritional intake may last for
only a few weeks.59 Unfortunately, there has been
only limited research on the value of nutritional
counseling in palliative care.
FATIGUE
Fatigue is one of the most common symptoms
encountered in palliative care patients.60,61 It has
been ranked as the longest lasting and most disruptive of symptoms,62 having the greatest impact on quality-of-life parameters.63 Fatigue, similar to other symptoms, is a subjective sensation,
which manifests with varying dimensions of impaired physical, cognitive, and affective functioning.64 The physical dimension is usually described as a perception of muscle weakness or
decreased energy. The cognitive and affective dimensions include difficulty in concentrating or
maintaining attention and lack of motivation or
interest in activities.
413
SYMPTOM CONTROL: CACHEXIA, ANOREXIA, FATIGUE
Frequency rates as high as 96% for fatigue have
been reported in association with chemotherapy
and radiotherapy,65 and severe fatigue is almost
universal in patients receiving biological response modifiers, such as interferon- and IL2.66–68 In cancer survivors, fatigue has also been
reported as a disruptive symptom months or
years after cancer treatment ends.69–71 A qualitative study of patients with severe heart failure revealed difficulties with walking and extreme fatigue similar to that of patients living with
advanced cancer.72 In a study of 427 ambulatory
patients with AIDS,73 fatigue was found to be
present in 54%, and its presence correlated
strongly with the number of AIDS-related physical symptoms, anemia and pain. The patients
with fatigue were also observed to have relatively
poorer physical functioning and quality of life
and greater psychological distress than those
with no fatigue.
MECHANISMS
Multiple etiological factors may coexist and be
interrelated. The common contributors to fatigue in advanced cancer include the tumor, the
treatments and the proinflammatory cytokines
produced (interleukin-1, IL-6, and TNF-). Cytokines are the likely mediators of anemia, fever,
FIG. 2.
pain, cachexia, and depression. Comorbidities
such as congestive heart failure, kidney, and
liver failure exacerbate fatigue as do electrolyte
endocrine and mood disorders These factors are
shown in Figure 2.
ASSESSMENT
Fatigue is assessed much like any other symptom, namely by characterizing the severity, temporal features (onset, course, duration, and daily
pattern), exacerbating and relieving factors, associated distress, and impact on daily life,74 while
searching for contributing factors. Additional information is obtained from the history and physical examination and laboratory data. In palliative care practice, patients present with multiple
symptoms, which can be simultaneously assessed
by the ESAS. The presence of other symptoms
such as pain, sleep, depression, and anxiety, are
strongly correlated with fatigue.
A more rigorous method, used for research
purposes and validated for internal consistency,
Functional Assessment of Cancer Therapy-Fatigue Subscale (FACT-F).75 The Brief Fatigue Inventory (BFI) has also been used and validated
as a measure of fatigue in patients with cancer,76
as has the Memorial Symptom Assessment ScaleShort Form (MSAS-SF). A study of patient re-
Contributors to fatigue.
414
DEL FABBRO ET AL.
sponses to BFI, FACT-F, and the lack of energy
item yielded similar information for assessing fatigue.77
MANAGEMENT
Figure 3 shows an approach to management of
fatigue and is discussed below.
Specific measures
Anemia is frequent in patients with cancer and
may be secondary to bleeding, treatment-related
FIG. 3.
myelosuppression, chronic disease, and malnutrition. Severe anemia (hemoglobin 8 g/dL) is
known to cause profound fatigue; however, a
higher hemoglobin may also be associated with fatigue. Using a hemoglobin value of 12 g/dL or less
to define anemia, anemic cancer patients requiring
chemotherapy were found to have significantly
higher levels of fatigue and impaired physical and
functional well-being.78 There is a clear relationship
between anemia and fatigue in patients with cancer receiving chemotherapy,79 and treatment of
these patients with red blood cell transfusion
and/or erythropoetin alpha80 has shown to im-
Approach to fatigue management in palliative care.
SYMPTOM CONTROL: CACHEXIA, ANOREXIA, FATIGUE
prove quality of life and energy.81 Although therapy with erythropoetic agents is relatively safe, the
disadvantages include the cost and the delay of 4–8
weeks before an increase of 1–2 g/dL and symptom improvement occurs. Recent data suggest that
erythropoietin leads to an increase risk of deep venous thrombosis and may, in certain populations,
increase mortality. Moreover, data82 supporting
erythropoetin’s usefulness in patients not on chemotherapy are less clear. In a recent retrospective
study of 147 patients with cancer receiving palliative care, anemia was not found to be independently associated with fatigue. Because the number and severity of other comorbid conditions
increase with advancement of disease, the relative
contribution of anemia to fatigue likely decreases
progressively.83
Infection. Fatigue is frequently associated with infection. It may predate the episode of infection and
may outlast the infection by weeks or months.
Anxiety, depression, or other mood disturbances are common in patients with cancer.84
There is an association between depression and
fatigue in patients with cancer; however, a causal
relationship has not been established
Hypogonadism may also contribute to fatigue.
Much of the research on the role of hypogonadism in fatigue has been done on HIV-positive
men. Testosterone deficiency is associated with
loss of muscle mass, fatigue, reduced libido, and
anemia.85 In patients with HIV with testosterone
deficiency, treatment with androgenic anabolic
steroids, including testosterone and its derivatives, has been found to increase muscle mass,
energy levels, and libido.86–89 A randomized 10week trial of 47 patients with COPD with testosterone deficiency showed that testosterone plus
resistance training had additive benefits.90 Both
lean body mass and leg muscle strength increased
significantly. Recently published studies have
found a high prevalence of hypogonadism in patients with cancer91 and in patients receiving high
doses of opioids.92 In both these studies, the presence of hypogonadism was associated with fatigue. Therapeutic trials of androgen replacement
therapy in these patients for fatigue management
are currently in progress.
Pharmacologic
Medications such as MA and dexamethasone
may alleviate some of the symptoms of fatigue in
415
patients with advanced cancer.93,94 Corticosteroids have been shown to decrease fatigue via unknown mechanisms.95 The duration of benefit is
between 2–4 weeks, and treatment may be suitable for patients with a short life expectancy. Prolonged use may induce myopathy and infection,
thereby further contributing to fatigue. The usual
starting dose in most studies is prednisone 20–40
mg per day or its equivalent.
Psychostimulant medications have been
shown in studies to ameliorate fatigue in patients
with AIDS and multiple sclerosis.96,97 In a randomized double-blinded placebo-controlled trial
of ambulatory patients with HIV, two psychostimulants—pemoline and methylphenidate—
were shown to improve fatigue scores and quality of life. In patients with cancer, the most
commonly used psychostimulant is methylphenidate. Dextroamphetamine, pemoline, and
modafinil have also been used, although there
have been no controlled comparisons of any of
these drugs. Methylphenidate has been used in
cancer patients to treat opioid induced somnolence, reduce pain intensity, treat depression, and
improve cognition.98 Preliminary evidence suggests that psychostimulants may have a role in
fatigue management in patients with cancer.99
Clinical trials are in progress. Modafinil, which is
licensed for treatment of narcolepsy, is a novel
psychostimulant that has been shown to be effective for management of fatigue in multiple
sclerosis.100 Further research is warranted to evaluate its role in fatigue management in advanced
chronic disease.
Donepezil, a centrally acting acetylcholinesterase (AChE) inhibitor approved for use in
Alzheimer’s disease, may have a role in some patients with fatigue. Preliminary studies in patients with cancer have suggested at least shortterm benefit in treating opiate-related sedation,101
as well as improvement in fatigue scores.102
Randomized controlled trials are currently in
progress.
Nonpharmacologic approaches
A multidisciplinary approach to management
benefits patients with advanced cancer and other
disease such as CHF.103
Patients with heart failure have a wide array of
options104 should standard medical therapy (diuretics, angiotensin-converting enzyme [ACE]inhibitors, beta blockers, aldosterone antagonists,
416
digoxin) fail to improve fatigue and functional
capacity. However, more invasive emerging
therapies, such as ventricular assist devices or resynchronization pacing should be carefully discussed with the primary team before implementation, because of their considerable impact on
complexity of care and hospital discharge.
Patient education. Patients with symptoms of fatigue need to be counseled about the nature of
symptoms, likely etiology of their fatigue, options
for management, and the expected outcome. This
will help patients and their families set realistic
expectations. As the disease progresses, patients
will need to adapt to progressive limitations in
physical function and activity. Patient preferences for information and willingness to participate in exercise and other treatment options
should be ascertained.
Exercise programs. In patients with cancer, muscle abnormalities have been found even when lean
body mass is constant and caloric intake normal.
Excessive amounts of lactate have been found in
tumor-free muscle tissues and it is unclear whether
lactate is part of the pathogenic mechanism or consequent to it.105 Tumor-free muscle from tumorbearing animals show alterations in the activity of
various enzymes, distribution of isoenzymes, and
synthesis and breakdown of myofibrillar and sarcoplasmic proteins.106 Patients with breast cancer
who underwent electrical stimulation of the abductor pollicis muscle via the ulnar nerve were
found to have impaired maximal strength, decreased relaxation velocity, and increased fatigue
compared to normal controls.107 In addition, medications such as corticosteroids and cyclosporine
may contribute to myopathy.108 Deconditioning
resulting from inactivity and prolonged bed rest
results in loss of muscle mass and reduced cardiac
output. This in turn decreases endurance and activities of daily living.109,110 There is good evidence111 to support the effectiveness of exercise in
patients with fatigue.112–117 Exercise improves fatigue through an adaptive cardiorespiratory response, maintenance of muscle mass, improvement in mood, and quality of sleep. Several studies
have shown that endurance exercise training improves fatigue and physical performance in patients with cancer undergoing chemotherapy or in
those who have undergone bone marrow or stem
cell transplantation.118–120 Patients with multiple
sclerosis121 who underwent an 8-week progressive
DEL FABBRO ET AL.
resistance training program reported decreased fatigue and disability. There are also similarities122
between COPD and CHF with regard to muscle
dysfunction and the impact of exercise.
Although there is abundance of data to support exercise, selection of the most appropriate
program for patients with various types of advanced disease and different levels of performance status are not well defined. Any exercise
program should be initiated gradually.
Psychosocial interventions. Patients should be
counseled about stress management and methods
to deal with depression and anxiety, which are
commonly associated with fatigue. Clinical studies testing interventions to reduce stress and increase psychosocial support in cancer patients
have shown reduction in fatigue levels, although
in many of these studies fatigue was a secondary
endpoint.123–126
Nutrition and hydration. Patients may have decreased oral intake due to a variety of causes related to their illness or treatment. Referral to a nutritionist for dietary planning may be indicated if
decreased intake is judged to be one of the causes
of fatigue.
Future treatment approaches
Several promising approaches to fatigue management have been identified. Cytokine antagonists may be exploitable in combating the components of cancer-related fatigue and may also
inhibit tumor growth.127 Pentoxifylline and
bradykinin antagonists inhibit the release of cytokines, while others, such as COX-2 inhibitors,
nonselective COX inhibitors, -melanocytes–
stimulating hormone, and monoclonal antibodies,128 inhibit cytokine action. Thalidomide has
been shown to improve the sensation of well-being in patients with cancer cachexia, and may also
be helpful in cancer-related fatigue.129 Other potential treatment approaches that warrant research include human growth hormone, and Lcarnitine130 supplementation.
REFERENCES
1. Ma G, Alexander HR: Prevalence and pathophysiology of cancer cachexia. In Bruera E, Portenoy RK
(eds): Topics in Palliative Care. New York: Oxford
University Press, 1998, pp. 91–129.
SYMPTOM CONTROL: CACHEXIA, ANOREXIA, FATIGUE
2. DeWys WD, Begg D, Lavin PT: Prognostic effect of
weight loss prior to chemotherapy in cancer patients.
Am J Med 1980;69:491–499.
3. Skeletal muscle dysfunction in chronic obstructive
pulmonary disease. A statement of the American
Thoracic Society and European Respiratory Society.
Am J Respir Crit Care Med 1999;159:1–40.
4. Debigare R, Cote CH, Maltais F: Peripheral muscle
wasting in chronic obstructive pulmonary disease.
Clinical relevance and mechanisms. Am J Respir Crit
Care Med 2001;164:1712–1717.
5. Nahlen BL, Chu SY, Nwanyanwu OC, Berkelmon
RL, Martinez SA, Rullon JV: HIV wasting syndrome
in the United States. AIDS 1993;7:183–188.
6. Anker SD, Coats AJ: Cardiac cachexia: A syndrome
with impaired survival and immune and neuroendocrine activation. Chest 1999;115:836–847.
7. Onwuamaegbu ME: Cachexia in malaria and heart
failure: therapeutic considerations in clinical practice. Postgrad Med J 2004;80:642–649.
8. Roongpisuthipong C, Sobhonslidsuk A, Nantiruj K,
Songchitsomboon S: Nutritional assessment in various
stages of liver cirrhosis. Nutrition 2001;17:761–765.
9. Zaki MH, Nemeth JA, Trikha M: CNTO 328 a monoclonal antibody to IL-6 inhibits human tumor-induced cachexia in nude mice. Int J Cancer 2004
;111:592–595.
10. Mantovani G, Maccio A, Madeddu C, Massa A: Cancer-related cachexia and oxidative stress:beyond current therapeutic options. Expert Rev Anticancer Ther
2003;3:381–392.
11. Mak RH, Cheung W, Cone RD, Marks DL: Orexigenic and anorexigenic mechanisms in the control of
nutrition in chronic kidney disease. Pediatr Nephrol
2005;20:427–431.
12. Berry J, Baum C: Reversal of COPD-associated
weight loss. Drugs 2004;64:1041–1052.
13. Rall LC, Roubenoff R: Rheumatoid cachexia: Metabolic abnormalities,mechanisms and interventions.
Rheumatology 2004;43:1219–1223.
14. Ferrigno D, Buccheri G: Anthropometric measurements in non-small cell lung cancer. Supp Care Cancer 2001;9:522–527.
15. Loprinzi CL, Micholok JC, Schaid DJ, Mailliard JA,
Athmann LM, Goldberg RM, Tschetter LK, Hatfield
AK, Morton RF: Phase III evaluation of four doses
of megestrol acetate as therapy for patients with cancer anorexia and/or cachexia. J Clin Oncol
1993;11:762–767.
16. Bruera E, Macmillan K, Kuehn N, Hanson J, MacDonald RN: A controlled trial of megestrol acetate
on appetite, caloric intake, nutritional status, and
other symptoms in patients with advanced cancer.
Cancer 1990;66:1279–1282.
17. Bruera E, Ernst S, Hagen N, Spachynski K, Belzile
M, Hanson J, Summers N, Brown B, Dulude H, Gallant G: Effectiveness of megestrol acetate in patients
with advanced cancer: A randomized, double-blind,
crossover study. Cancer Prev Control 1998;2:74–78.
18. Kornblith AB, Hollis DR, Zuckerman E, Lyss AP,
19.
20.
21.
22.
23.
24.
25.
26.
27.
28.
29.
30.
417
Canellos GP, Cooper MR, Herndon JE, Phillips CA,
Abrams J, Aisner J: Effect of megestrol acetate on quality of life in a dose-response trial in women with advanced breast cancer. J Clin Oncol 1993;11:2081–2089.
Loprinzi CL, Kugler JW, Sloan JA, Mailliard JA,
Krock JE, Wilwerding MB, Rowland KM, Camoriano JK, Novotny PJ: Randomized comparison of
megestrol acetate versus dexamethasone versus fluoxymesterone for the treatment of cancer anorexia/
cachexia. J Clin Oncol 1999;17:3299–3306.
Pascual Lopez A, Roque i Figuls M, Urrutia Cuchi,
Berenstein EG, Almenar Pasies B, Balcells Alegre M,
Herdman M: Systematic review of megestrol acetate
in the treatment of anorexia-cachexia syndrome. J
Pain Symptom Manage 2004;27:360–369.
Weisberg J, Wanger J, Olson J, Streit B, Fogarty C,
Martin T, Casaburi R: Megestrol acetate stimulates
weight gain and ventilation in underweight COPD
patients. Chest 2002;121:1070–1078.
Simons JPFHA, Aaronson NK, Vansteenkiste JF,
ten Velde GP, Muller MJ, Drenth BM, Erdkamp FL,
Cobben EG, Schoon EJ, Smeets JB, Schouten HC,
Demedts M, Hillen HF, Blijham GH, Wouters EF:
Effect of medroxyprogesterone acetate on appetite,
weight and quality of life in advanced-stage nonhormone-sensitive cancer: a placebo controlled
multicenter study. J Clin Onc 1996;14:1077–1084.
Popiela T, Lucchi R, Giongo F: Methylprednisolone
as an appetite stimulant in patients with cancer. Eur
J Cancer Clin Oncol 1989;25:1823–1829.
Willox J, Corr J, Shaw J, Richardson M, Calman KC,
Drennan M: Prednisolone as an appetite stimulant
in patients with cancer. Br Med J 1984;27:288–290.
Bruera E, Roca E, Cedaro l, Carraro S, Chacon R: Action of oral methylprednisolone in terminal cancer
patients: A prospective randomized double blind
study. Cancer Treat Rep 1985;69:751–754.
Beal JE, Olson R, Laubenstein L, Morales JO, Bellman P, Yangco B, Lefkowitz L, Plasse TF, Shepard
KV: Dronabinol as a treatment for anorexia associated with weight loss in patients with AIDS. J Pain
Symptom Manage 1995;10:89–97.
Walsh D, Nelson KA, Mahmoud FA: Established and
potential therapeutic applications of cannabinoids in
oncology. Support Care Cancer 2003;11:137–143.
Lane M, Vogel CL, Ferguson J, Kranow S, Saiers JL,
Hamm J, Salva K, Wiernik PH, Holroyde CP, Hammill S: Dronabinol and prochlorperazine in combination for treatment of cancer chemotherapy-induced nausea and vomiting. J Pain Symptom
Manage 1991;6:352–359.
Jatoi A, Windschitl HE, Loprinzi CL, Sloan JA,
Dakhil SR, Mailliard JA, Pundaleeka S, Kardinal CG,
Fitch TR, Krook JE, Novotny PJ, Christensen B:
Dronabinol versus megestrol acetate versus combination therapy for cancer-associated anorexia: A
North Central Cancer Treatment Group study. J Clin
Oncol 2002;20:567–573.
Beal JE, Olson R, Lefkowitz L, Laubenstein L, Bellman P, Yangco B, Morales JO, Murphy R, Powderly
418
31.
32.
33.
34.
35.
36.
37.
38.
39.
40.
41.
42.
43.
DEL FABBRO ET AL.
W, Plasse TF, Mosdell KW, Shepard KV: Long–term
efficacy and safety of Dronabinol for AIDS-associated anorexia. J Pain Symp Manage 1997;14:7–14.
Peuckmann V, Fisch M, Bruera E: Potential novel
uses of thalidomide. Focus on palliative care. Drugs
2000;60:273–292.
Bruera E, Neumann CM, Pituskin E, Calder K, Ball
G, Hanson J: Thalidomide in patients with cachexia
due to terminal cancer: preliminary report. Ann Oncol 1999;10:857–859.
Gordon JN, Trebble TM, Ellis RD, Duncan HD, Johns
T, Goggin PM: Thalidomide in the treatment of cancer cachexia:a randomized placebo controlled trial.
Gut 2005;54:540–545.
Khan ZH, Simpson EJ, Cole AT: Oesophageal
cancer and cachexia:the effect of short–term treatment with thalidomide on weight loss and lean
body mass. Aliment Pharmacol Ther 2003;17:
677–682.
Jacobson JM, Greenspan JS, Spritzler J, Ketter N, Fahey JL, Jackson JB, Fox L, Chernoff M, Wu AW,
MacPhail L, Vasquez GJ, Wohl DA: Thalidomide for
the treatment of oral aphthous ulcers in patients
with HIV infection. N Engl J Med 1997;336:1487–
1493.
Kaplan G, Thomas S, Fierer DS, Mulligan K, Haslett
PA, Fessel WJ, Smith LG, Kook KA, Stirling D,
Schambelan M: Thalidomide for the treatment of
AIDS-associated wasting. AIDS Res Hum Retroviruses 2000;16:1345–1355.
Tramontana JM, Utaipat U, Molloy A, Akarasewi P,
Burroughs M, Makonkawkeyoon S, Johnson B,
Klausner JC, Ram W, Kaplan G: Thalidomide treatment reduces tumor necrosis factor alpha production and enhances weight gain in patients with pulmonary tuberculosis. Mol Med 1995;1:384–397.
Orr R, Fiatarone Singh M: The anabolic androgenic
steroid oxandrolone in the treatment of wasting and
catabolic disorders:review of efficacy and safety.
Drugs 2004;64:725–750.
Yeh SS, DeGuzman B, Kramer T: Reversal of COPDassociated weight loss using the anabolic agent
drolone. Chest 2002;122:421–428.
Schols AM, Soeters R, Mostert R, Pluymers RJ,
Wouters EF: Physiologic effects of nutritional support and anabolic steroids in patients with COPD. A
placebo-controlled randomized trial. Am J Resp Crit
Care Med 1995;152(4 Pt 1):1268–1274.
Schambelan M, Mulligan K, Grunfeld C, Daar ES,
LaMarca A, Kotler DP, Wang J, Bozzette SA, Breitmeyer JB: Recombinant human growth hormone in
patients with HIV-associated wasting. A randomized placebo controlled trial. Serostim Study group.
Ann Intern Med 1996;125:873–882.
Vanitallie TB: Frailty in the elderly: Contributions of
sarcopenia and visceral protein depletion. Metabolism 2003;52:22–26.
Nagaya N, Kangawa K: Ghrelin improves left ventricular dysfunction and cardiac cachexia in heart
failure. Curr Opin Pharmacol 2003;3:146–151.
44. Lissoni P: Is there a role for melatonin in supportive
care? Supp Care Cancer 2002;10:110–116.
45. Persson C, Glimelius B, Ronalid J, Nygren P: Impact
of fish oil and melatonin on cachexia in patients with
advanced gastrointestinal cancer: A randomized pilot study. Nutrition 2005;21:170–178.
46. Barber MD, Ross JA, Voss AC, Tisdale MJ, Fedron
KC: The effect of an oral nutritional supplement enriched with fish oil on weight loss in patients with
pancreatic cancer. Br J Cancer 1999;81:80–86.
47. Bruera E, Strasser F, Palmer JL, Willey J, Calder K,
Amyotte G, Baracos V: Effect of fish oil on appetite
and other symptoms in patients with advanced cancer and anorexia/cachexia:a double blind placebo
controlled study. J Clin Oncol 2003;21:129–134.
48. Jatoi A, Rowland K, Loprinzi CL, Sloan JA, Dakhil
SR, MacDonald N, Gagnon B, Novotny PJ, Mailliard
JA, Bushey TI, Nair S, Christensen B: An eicosapentaenoic acid supplement versus megestrol acetate
versus both for patients with cancer-associated wasting. J Clin Oncol 2004;22:2469–2476.
49. Hryniewicz K, Androne AS, Hudaihed A, Katz SD:
Partial reversal of cachexia by beta adrenergic receptor blocker therapy in patients with chronic heart
failure. J Card Fail 2003;9:464–468.
50. Maltin CA, Delday MI, Watson JS, Heys SD, Nevison IM, Ritchie IK, Gibson PH: Clenbuterol a beta
adrenoreceptor agonist, increases relative muscle
strength in orthopaedic patients. Clin Sci 1993;84:
651–654.
51. Trikha M, Corringham R, Klein B, Rossi JF: Targeted
anti-interleukin-6 monoclonal antibody therapy for
cancer: A review of the rationale and clinical evidence. Clin Cancer Res 2003;9:4653–4665.
52. Jirathitikal V, Metadilogkul O, Bourinbaiar AS: Increased body weight and improved quality of life in
AIDS patients following V1 Immunitor administration. Eur J Clin Nutr 2004;58:110–115.
53. Davis MP, Khawam E, Pozuelo L, Lagman R: Management of symptoms associated with advanced
cancer: Olanzapine and mirtazapine. A WHO project. Expert Rev Anticancer Ther 2002;2:365–376.
54. McGeer AJ, Detsky AS, O’Rourke K: Parenteral nutrition in cancer patients undergoing chemotherapy
:a meta analysis. Nutrition 1990;6:233–240.
55. Klein S, Koretz RL: Nutrition support in patients
with cancer: What do the data really show? Nutr Clin
Pract. 1994;9:91–100.
56. Hoda D, Jatoi A, Burnes J, Loprinzi C, Kelly D:
Should patients with advanced, incurable cancers
ever be sent home with total parenteral nutrition?
Cancer 2005;103:863–868.
57. Lundholm K, Daneryd P, Bosaeus I, Korner U, Lindholm E: Palliative nutritional intervention in addition to cyclooxygenase and erythropoietin treatment
for patients with malignant disease: Effects on survival, metabolism, and function. Cancer 2004;100:
1967–1977.
58. Ovesen L, Allingstrup L, Hannibal J, Mortensen EL,
Hansen OP: Effect of dietary counseling on food in-
SYMPTOM CONTROL: CACHEXIA, ANOREXIA, FATIGUE
59.
60.
61.
62.
63.
64.
65.
66.
67.
68.
69.
70.
71.
72.
73.
take, body weight, response rate, survival, and quality of life in cancer patients undergoing chemotherapy: A prospective, randomized study. J Clin Oncol
1993;11:2043–2049.
Bruera E, MacDonald RN: Nutrition in cancer patients: An update and review of our experience. Issues in symptom control. 3. J Pain Symptom Manage 1998;3:133–140.
Portenoy RK: Cancer-related fatigue: An immense
problem. Oncologist 2000;5:350–352.
Jenkins CA, Schulz M, Hanson J, Bruera E: Demographic, symptom, and medication profiles of cancer patients seen by a palliative care consult team in
a tertiary referral hospital. J Pain Symptom Manage
2000;19:174–184.
Curt GA, Breitbart W, Cella D, Groopman JE, Hornings SJ, Itri LM, Johnson DH, Miaskowski C, Scherr
SL, Portenoy RK, Vogelzang NJ: Impact of cancerrelated fatigue on the lives of patients: New findings
from the Fatigue Coalition. Oncologist 2000;5:353–
360.
Asbury FD, Findaly H, Reynolds B, McKerracher K:
Canadian survey of cancer patients’ experiences: Are
their needs being met? J Pain Symptom Manage
1998;16:298–306.
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.
Irvine DM, Vincent L, Bubela N, Thompson L, Graydon L: A critical appraisal of the research literature
investigating fatigue in the individual with cancer.
Cancer Nurs 1991;14:188–199.
Piper BF, Rieger PT, Brophy L, Haeuber D, Hood LE,
Lyver A, Sharp E: Recent advances in the management of biotherapy-related side effects: fatigue. Oncol Nurs Forum 1989;16(Suppl 6):27–34.
Skalla K, Rieger P: Fatigue. In: Rieger PT (ed): Biotherapy: Comprehensive Review. Boston: Jones &
Bartlett, 1995, pp. 221–242.
Dean GE, Spears L, Ferrell B, Quan WD, Groshon S,
Mitchell MS: Fatigue in patients with cancer receiving interferon alpha. Cancer Pract 1995;3:164–171.
Bower JE, Ganz PA, Desmond KA, Rowland JH,
Meyerowitz BE, Belin TR: Fatigue in breast cancer
survivors: Occurrence, correlates, and impact on
quality of life. J Clin Oncol 2000;18:743–753.
Fossa SD, Dahl AA, Loge JH: Fatigue, anxiety and
depression in longterm survivors of testicular cancer. J Clin Oncol 2003;21:1249–1254.
Servaes P, Verhagen S, Schreuder HW, Veth RP,
Bleijenberg G: Fatigue after treatment for malignant
and benign bone and soft tissue tumors. J Pain Symp
Manage 2003;26:1113–1122.
Horne G, Payne S: Removing the boundaries: Palliative care for patients with heart failure. Palliat
Med 2004;18:291–296.
Breitbart W, McDonald MV, Rosenfeld B, Monkman
ND, Passik S: Fatigue in ambulatory AIDS patients.
J Pain Symptom Manage 1998;15:157–167.
419
74. Portenoy RK, Itri LM: Cancer-related fatigue. Guidelines for evaluation and management. Oncologist
1999;4:1–10.
75. Cella DF, Tulsky DS, Gray G, Sarafian B, Linn E,
Bonomi A, Silberman M, Yellen SB, Winicour P,
Brannon J: The functional assessment of cancer therapy scale: Development and validation of the general measure. J Clin Oncol 1993;11:570–579.
76. Mendoza TR, Wang XS, Cleeland CS, Morrissey M,
Johnson BA, Wendt JK, Huber SL: The rapid assessment of fatigue severity in cancer patients. Cancer.
1999;85:1186–1196.
77. Hwang SS, Chang VT, Kasimis BS: A comparison of
three fatigue measures in veterans with cancer. Cancer Invest 2003;21:363–373.
78. Cella D: The Functional Assessment of Cancer Therapy-Anemia (FACT-An) Scale: A new tool for the assessment of outcomes of cancer anemia and fatigue.
Semin Hematol 1997;34:13–19.
79. Cella D, Kallich J, McDermott A, Xu X: The longitudinal relationship of hemoglobin, fatigue and quality of life in anemic cancer patients: Results from five
randomized clinical trials. Ann Oncol 2004;15:
979–986.
80. Turner R, Anglin P, Burkes R, Couture F, Evans W,
Goss G, Grimshaw R, Melosky B, Paterson A, Quirt
I: Epoetin alfa in cancer patients: Evidence-based
guidelines. J Pain Symptom Manage 2001;22:954–
965.
81. Glaspy J, Bukowski R, Steinberg D, Taylor C,
Tchekmedyian S, Vadhan-Raj S: Impact of therapy
with epoetin alfa on clinical outcomes in patients
with nonmyeloid malignancies during cancer chemotherapy in community oncology practice. Procrit
Study Group. J Clin Oncol 1997;15:1218–1234.
82. Henke M, Laszig R, Rube C, Schafer U, Haase KD,
Schilcher B, Mose S, Beer K, Burger U, Daugherty C,
Frommhold H: Erythropoietin to treat head and neck
cancer patients with anaemia undergoing radiotherapy: Randomized, double-blinded placebo-controlled trial. Lancet 2003;362:1255–1260.
83. Bruera E: EAPC Meeting: Aahen 2005.
84. Valentine AD, Meyers CA: Cognitive and mood disturbance as causes and symptoms of fatigue in cancer patients. Cancer 2001;15(Suppl. 6):1694–1698.
85. Basaria S, Wahlstrom JT, Bobs AS: Clinical review
138: Anabolic-androgenic steroid therapy in the
treatment of chronic diseases. J Clin Endocrinol
Metab 2001;86:5108–5117.
86. Snyder PJ, Peachey H, Berlin JA, Hannoush P, Haddad G, Dlewati A, Santanna J, Loh L, Lenrow DA,
Holmes J, Kapoor SC, Atkinson LE, Strom BL: Effects of testosterone replacement in hypogonadal
men. J Clin Endocrinol Metab 2000;85:2670–2677.
87. Bhasin S, Storer TW, Berman N, Yarasheski KE, Clevenger B, Phillips J, Lee WP, Bunnell TJ, Casaburi R:
Testosterone replacement increases fat-free mass
and muscle size in hypogonadal men. J Clin Endocrinol Metab 1997;82:407–413.
88. Bhasin S, Storer TW, Asbel-Sethi N, Kilbourne A,
420
89.
90.
91.
92.
93.
94.
95.
96.
97.
98.
99.
100.
DEL FABBRO ET AL.
Hays R, Sinha-Hikim, Shen R, Arver S, Beall G: Effects of testosterone replacement with a nongenital, transdermal system, Androderm, in human immunodeficiency virus-infected men with low
testosterone levels. J Clin Endocrinol Metab 1998;
83:3155–3162.
Grinspoon S, Corcoran C, Askari H, Schoenfeld D,
Wolf L, Burrows B, Walsh M, Hayden D, Parlman
K, Anderson E, Basgoz N, Klibanski A: Effects of
androgen administration in men with the AIDS
wasting syndrome. A randomized, double-blind,
placebo-controlled trail. Ann Intern Med 1998;129:
18–26.
Casaburi R, Bhasin S, Cosantino L: Effects of testosterone and resistance training in men with COPD.
Am J Respir Crit Care Med 2004;170:870–878.
Shafqat A, Einhorn LH, Hanna N, Sledge GW,
Hanna A, Juliar B, Monahan P, Bhatia S: Screening
studies for fatigue and laboratory correlates in cancer patients undergoing treatment. Ann Oncol
2005;16:1545–1550.
Rajagopal A, Vassilopoulou-Sellin R, Palmer JL,
Kaur G, Bruera E: Symptomatic hypogonadism in
male survivors of cancer with chronic exposure to
opioids. Cancer 2004;100:851–858.
Bruera E, Roca E, Cedaro L, Carraro S, Chacon R:
Action of oral methylprednisolone in terminal cancer patients: a prospective randomized double-blind
study. Cancer Treat Rep 1985;69:751–754.
Bruera E, Ersnt S, Hagen N, Spachynski K, Belzile
M, Hanson J, Summers N, Brown B, Dulude H, Gallant G: Effectiveness of megestrol acetate in patients
with advanced cancer: A randomized, double-blind,
crossover study. Cancer Prev Control 1998;2:74–78.
Bruera E, Roca E, Cedaro L, Carraro S, Chacon R:
Action of oral methylprednisolone in terminal cancer patients: A prospective randomized doubleblind study. Cancer Treat Rep 1985;69:751–754.
Breitbart W, Rosenfeld B, Kaim M, Funesti-Esch J:
A randomized, double-blind, placebo-controlled
trial of psychostimulants for the treatment of fatigue in ambulatory patients with human immunodeficiency virus disease. Arch Intern Med 2001;
161:411–420.
Krupp LB, Coyle PK, Doscher C, Miller A, Cross AH,
Jandorf L, Halper J, Johnson B, Morgante L, Grimson R: Fatigue therapy in multiple sclerosis: A double-blind, randomized, parallel trial of amantadine,
pemoline and placebo. Neurology 1995;45:1956–
1961.
Rozans M, Dreisbach A, Lertora JJ, Kahn MJ: Palliative uses of methylphenidate in patients with cancer: A review. J Clin Oncol 2002;20:335–339.
Bruera E, Driver L, Barnes EA, Willey J, Shen L,
Palmer JL, Escalante C: Patient-controlled methylphenidate for the management of fatigue in patients
with advanced cancer: A preliminary report. J Clin
Oncol 2003;21:4439–4443.
Zifko UA: Management of fatigue in patients with
multiple sclerosis. Drugs 2004;64:1295–1304.
101. Slatkin NE, Rhiner M: Treatment of opiate-related
sedation: Utility of the cholinesterase inhibitors. J
Support Oncol 2003;1:53–63.
102. Bruera E, Strasser F, Shen L, Palmer J, Willey J, Driver L, Burton A: The effect of donepezil on sedation and other symptoms in patients receiving opioids for cancer pain: A pilot study. J Pain Symptom
Manage 2003;26:1049–1054.
103. Phillips C, Wright S, Kern D, Singa RM, Shepperd S,
Rubin HR: Comprehensive discharge planning with
post discharge support for older patients with congestive heart failure:a meta-analysis. JAMA 2004;
291:1358–1367.
104. Hauptman P, Havranek E: Integrating palliative care
into heart failure care. Arch Intern Med 2005;
165:374–378.
105. Holroyde CP, Axelrod RS, Skutches CL, Haff AC,
Paul P, Reichard GA: Lactate metabolism in patients
with metastatic colorectal cancer. Cancer Res 1979;
39:4900–4904.
106. Warmolts JR, Peter K, Lewis RJ, Engel WK: Type II
muscle fibre atrophy (II-atrophy: An early systemic
effect of cancer. Neurology 1975;2:374.
107. Bruera E, Brenneis C, Michaud M, Jackson PI, MacDonald RN: Muscle electrophysiology in patients
with advanced breast cancer. J Natl Cancer Inst
1988;80:282–285.
108. Tirdel GB, Girgis R, Fishman RS, Theodore J: Metabolic myopathy as a cuase of the exercise limitation
in lung transplant recipients. J Heart Lung Transplant 1998;17:1231–1237.
109. Barnes EA, Bruera E: Fatigue in patients with advanced cancer: A review. Int J Gynecol Cancer
2002;12:424–428.
110. Germain P, Guell A, Marini JF: Muscle strength during bedrest with and without muscle exercise as a
countermeasure. Eur J Appl Physiol 1995;71:342–348.
111. Lucia A, Earnest C, Perez M: Cancer–related fatigue:
Can exercise physiology assist oncologists? Lancet
Oncol 2003;4:616–625.
112. Mock V: Fatigue management: Evidence and guidelines for practice. Cancer 2001;92:1699–1707.
113. Ahlberg K, Ekman T, Gaston-Johansson F, Mock V:
Assessment and management of cancer related fatigue in adults. Lancet 2003;362:640–650.
114. Dimeo FC: Effects of exercise on cancer-related fatigue. Cancer 2001;15(Suppl 6):1689–1693.
115. Courneya KS, Friedenreich CM: Physical exercise
and quality of life following cancer diagnosis:
A literature review. Ann Behav Med 1999;21:
171–179.
116. Schwartz AL: Daily fatigue patterns and effects of
exercise in women with breast cancer. Cancer Pract
2000;8:16–24.
117. Mock V, Pickett M, Ropka M, Muscari Lin E, Stewart KJ, Rhodes V, McDaniel R, Grimm PM, Krumm
S, McCorkle R: Fatigue, physical functioning, emotional distress, and quality of life outcomes of a walking intervention during breast cancer treatment.
Cancer Pract 2001;9:119–127.
SYMPTOM CONTROL: CACHEXIA, ANOREXIA, FATIGUE
118. Dimeo FC, Stieglitz RD, Novelli-Fischer U, Fetscher
S, Keul J: Effects of physical activity on the fatigue
and psychologic status of cancer patients during chemotherapy. Cancer. 1999;85:2273–2277.
119. Dimeo F, Fetscher S, Lange W, Mertelsmann R, Keul
J: Effects of aerobic exercise on the physical performance and incidence of treatment-related complications after high-dose chemotherapy. Blood 1997;90:
3390–3394.
120. Dimeo F, Bertz H, Finke J, Fetscher S, Mertelsmann
R, Keul J: An aerobic exercise program for patients
with haematological malignancies after bone marrow transplantation. Bone Marrow Transplant
1996;18:1157–1160.
121. White LJ, McCoy SC, Castellano V, Gutlerrez G,
Stevens JE, Walter G, Vandenborne K: Resistance
training improves strength and functional capacity
in persons with multiple sclerosis. Mult Scler
2004;10:668–674.
122. Troosters T, Gosselink R, Decramer M: COPD and
CHF: Two muscle diseases? J Cardiopulmon Rehabil 2004;24:137–145.
123. Fawzy NW: A psychoeducational nursing intervention to enhance coping and affective state in newly
diagnosed malignant melanoma patients. Cancer
Nurs 1995;18:427–438.
124. Spiegel D, Bloom JR, Yalom ID: Group Support for
metastatic cancer patients: A randomized prospective outcome study. Arch Gen Psychiatry 1981;
38:527–533.
125. Fawzy FI, Cousins N, Fawzy NW, Kemeny M,
Elashoff R, Morton D: A structured psychiatric intervention for cancer patients. Changes over time in
126.
127.
128.
129.
130.
421
methods of coping and affective disturbance. Arch
Gen Psychiatry 1990;47:720–725.
Jacobsen PB, Meade CD, Stein KD, et al: Efficacy and
costs of two forms of stress management training for
cancer patients undergoing chemotherapy. J Clin
Oncol 2002;20:2851–2862.
Kurzrock R: The role of cytokines in cancer-related
fatigue. Cancer 2001;92:1684–1688.
Burks TF: New agents for the treatment of cancerrelated fatigue. Cancer 2001;15(Suppl 6):1714–1718.
Bruera E, Neumann CM, Pitudkin E, Calder K, Ball
G, Hanson J: Thalidomide in patients with cachexia
due to terminal cancer: Preliminary report. Ann Oncol 1999;10:857–959.
Cruciani RA, Dvorkin E, Homel P, Culliney B, Malamud S, Shaiova L, Fleishman S, Lapin J, Klein E,
Lesage P, Portenoy R, Esteban-Cruciani N: L-carnitine supplementation for the treatment of fatigue
and depressed mood in cancer patients with carnitine deficiency: A preliminary analysis. Ann NY
Acad Sci 2004;1033:168–176.
Address reprint requests to:
Eduardo Bruera, M.D.
Department of Palliative
and Rehabilitation Medicine
University of Texas M. D. Anderson Cancer Center
Box 008
1515 Holcombe Boulevard
Houston, TX 77030
E-mail: [email protected]