You are on page 1of 7

International Journal of Cardiology 119 (2007) 83 – 89

www.elsevier.com/locate/ijcard

Anorexia in chronic obstructive pulmonary disease — Association


to cachexia and hormonal derangement
Friedrich Koehler a , Wolfram Doehner b,⁎, Soeren Hoernig c , Christian Witt c ,
Stefan D. Anker b,d , Matthias John c
a
Department of Cardiology, Charité University Hospital, Campus Mitte, Berlin, Germany
b
Division of Applied Cachexia Research, Department of Cardiology, Charité University Hospital, Campus Virchow, Berlin, Germany
c
Department of Pneumology, Charité University Hospital, Campus Mitte, Berlin, Germany
d
Clinical Cardiology, National Heart and Lung Institute, Imperial College, London, United Kingdom
Received 10 April 2006; accepted 15 July 2006
Available online 24 October 2006

Abstract

Background: In patients with chronic obstructive pulmonary disease (COPD) weight loss frequently occurs that may ultimately lead to
cachexia as a serious co-morbidity, indicating severely impaired functional capacity, health status and increased mortality. Increased energy
expenditure due to mechanic and metabolic inefficiency and systemic inflammation are determinants of a hypermetabolic state that is not
balanced by dietary intake. Anorexia may importantly contribute to weight loss in COPD, however, the association between immune and
hormonal derangement and altered appetite has not been studied in detail.
Aim: The aim of the present study was to investigate whether anorexia in COPD is related to inflammation and hormonal derangement in
association to weight loss.
Methods: We prospectively enrolled 103 consecutive patients with COPD (age 59.8 ± 1.3 years, 35% female, mean FEV1 38.3 ± 1.7%) in
comparison to healthy controls of similar age (n = 15).
Results: In 34 patients (33%) cachexia was diagnosed (weight loss N 7.5%, BMI ≤ 24 kg/m2). Cachectic COPD patients had lower BMI
(19.0 ± 0.5 vs 25.6 ± 0.7 kg/m2) and impaired lung function (FEV1 31 ± 2% vs 42 ± 2%, FVC 51 ± 3 vs 59 ± 3%, both p b 0.001). Inflammatory
immune activation (IL-6 and IL-6/IL-10 ratio) was significantly higher in cachectic COPD patients. Analysis of the extent of anorexia (visual
analogue scale) revealed that cachectic COPD patients had significantly decreased subjective desire to eat compared to non-cachectic patients
(3.5 ± 0.3 vs 6.3 ± 0.2, p b 0.001). Patients with COPD and cachexia showed evidence of acquired GH resistance (decreased IGF-1/GH ratio)
and insulin resistance (HOMA). Anorexia showed a direct correlation with the IGF-1/GH ratio (r = 0.34, p b 0.05) and was further related to
BMI and % weight loss (both p b 0.001).
Conclusion: In COPD anorexia relates to hormonal derangement and inflammatory immune activation. Anorexia contributes to development
of cachexia. The concept of appetite stimulating therapy emerges as a novel therapeutic option in cachectic COPD patients.
© 2006 Elsevier Ireland Ltd. All rights reserved.

Keywords: COPD; Anorexia; Cachexia; Catabolic/anabolic imbalance

Abbreviations: AIDS, Acquired immune deficiency syndrome; BMI, Body mass index; CHF, Chronic heart failure; cCOPD, Cachectic COPD; CLIA,
Chemiluminescence Immunoassay; COPD, Chronic obstructive pulmonary disease; DHEA, Dehydroepiandrosterone; ELISA, Enzyme-linked immunosorbent
assay; FEV1, Forced expiratory volume in 1 s; FVC, Forced ventilatory capacity; GH, Growth hormone; HOMA, Homeostatic model assessment; IGF-1, Insulin-
like growth factor-1; IL-6, Interleukin-6; Log, Logarithm; ncCOPD, Non-cachectic COPD; SEM, Standard error of mean; TNF-α, Tumor necrosis factor-α; IQR,
Interquartile range.
⁎ Corresponding author. Div of Applied Cachexia Research, Department of Cardiology, Charité Medical School, Campus Virchow Hospital, Augustenburger
Platz 1, 13353 Berlin, Germany. Tel.: +49 30 450 553 507; fax: +49 450 553951.
E-mail address: wolfram.doehner@charite.de (W. Doehner).

0167-5273/$ - see front matter © 2006 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.ijcard.2006.07.088
84 F. Koehler et al. / International Journal of Cardiology 119 (2007) 83–89

1. Introduction cording to the guidelines of the American Thoracic Society


[15]. All patients showed a FEV1 reversibility of less than
Chronic obstructive pulmonary disease (COPD) is an 15% in response to inhaled bronchodilators. At time of
inflammatory disease. In the chronic course of the disease, investigation, all patients were clinically stable without signs
the pathogenesis and clinical manifestations of COPD are of acute exacerbation. No patient had required hospital ad-
not restricted to pulmonary structural and functional im- mission or treatment change during the preceding 3 months.
pairment but rather have multi-systemic implications includ- Treatment was individually and included long acting β2
ing metabolic, hormonal and organ dysfunction such as in agonists (88%), ipratropium (20%), oxitropium bromide
skeletal muscle, heart, brain and skeleton [1]. Weight loss (30%), tiotropium bromide (11%), theophylline (69%) and/
due to tissue wasting is frequent in COPD and may ul- or inhaled steroids (32%) in varying combinations. Specific
timately lead to cachexia as a serious co-morbidity in ad- exclusion criteria were current or past diagnosis of allergic
vanced disease state. Cachexia in COPD – as in other asthma and a respiratory tract infection in the previous
chronic diseases – is associated with a greater susceptibility 3 months. Patients with cancer, thyroid disease, severe liver
to exacerbation of clinical symptoms, with severely im- disease and chronic heart failure were also excluded. Healthy
paired functional capacity [2], poor health status and quality subjects of similar age (n = 15) with no medical history
of life [3,4]. The loss of skeletal muscle tissue and, in served as control group.
particular, of respiratory muscle is associated with a loss of Body mass index (weight/height2) and weight loss were
power and endurance leading to a further mechanical assessed. Patients who reported to have had intentional
impairment of lung function with consecutive hypoxia and weight loss were excluded from the study. Patients where
hypercapnia [5]. This may contribute to progression of the divided into cachectic and non-cachectic subgroups. Ca-
disease. Accordingly, increasing evidence suggests that chexia was diagnosed when non-intentional, non-oedema-
weight loss and cachexia are independent prognostic factors tous weight loss of ≥ 7.5% of the previous normal weight
in COPD [6,7]. over a period of N 6 months was documented [16]. 34
Weight loss in COPD is a consequence of increased patients were diagnosed with cachexia (age 58 ± 2, 11 female
energy expenditure due to mechanic and metabolic ineffi- [32%]). The non-cachectic group consisted of 69 patients
ciency that is not balanced by adequately increased energy (age 61 ± 2, 25 female [36%], p N 0.2 vs cachectic patients).
supply. Inflammatory activation and hormonal derangements Appetite (the subjective desire to eat) was assessed using a
may further add to a hypermetabolic state and an overall visual analogue scale ranging from 0–10 (0 no appetite at all,
catabolic/anabolic imbalance leading to depletion of endog- 10 always very good appetite) based on the current eating
enous energy storages and eventually to structural tissue behaviour during 14 days prior to the study. The study was
degradation. approved by the Local Ethics Committee of the University
The regulation of energy expenditure in association to Hospital Charite, Humboldt University, Berlin. All patients
inflammatory activation and hormonal derangement in gave informed consent.
COPD patients has widely been studied [8–12]. Anorexia
may importantly contribute to weight loss in COPD. The 2.2. Pulmonary function test
therapeutic concept, however, of nutritional supplementa-
tion failed to show significant effects on anthropometric FVC and FEV1 were measured with standard spirometric
measures and functional capacity in these patients [13] and techniques (Masterlab, Jaeger, Wurzburg, Germany). All
mechanisms of non-response to nutritional therapy have values obtained were related to age and gender and expressed
been discussed [14]. In this context, the significance of as percentage of their predicted value.
alteration in appetite as a central regulator of food intake has
not been studied in detail. 2.3. Biochemical analyses
In the current study we prospectively assessed in a cohort
of COPD patients with and without weight loss the extent of In patients with and without cachexia, insulin-like growth
anorexia in relationship to alteration in body weight. The factor-1 (IGF-1), insulin and human growth hormone (GH)
specific association to hormonal and inflammatory para- were measured by radioimmunossay (Biochem Immunsys-
meters in order to identify potential factors that may in- tems, Freiburg, Germany, test kit sensitivity: 12 ng/ml for IGF-
fluence appetite in COPD is taken into account. 1, 1.0 μU/ml for insulin, 10 ng/ml for GH). The inflammatory
parameters, tumor necrosis factor alpha (TNFα, sensitivity
2. Methods 0.1 pg/ml), Interleukin-6 (IL-6, sensitivity 5 pg/ml) and were
analysed by ELISA technique using commercially available
2.1. Patient population kits (R and D Systems Inc., Minneapolis MN, USA). Cortisol,
testosterone and DHEA (Dihydroepiandrosterone) were
We recruited 103 consecutive patients (age 59.8 ± analysed by chemilumineszenz immunoassay (CLIA) (sensi-
1.3 years, 36 female [35%]) diagnosed with COPD by his- tivity: DHEA 0.054 μmol/l, cortisol 5,5 nmol/l, testosterone
tory, physical examination and pulmonary function test, ac- 0.35 nmol/l). Homeostasis model assessment (HOMA)
F. Koehler et al. / International Journal of Cardiology 119 (2007) 83–89 85

represents a marker of insulin sensitivity and is defined as


insulin × glucose/22.5 [17]. The ratio of IGF-1/GH was
calculated as an estimate of GH resistance with a lower IGF-
1/GH ratio indicating presence of FH resistance. Further ratios
between catabolic and anabolic hormones and cytokines were
calculated to characterise metabolic imbalance.

2.4. Blood sampling

Blood samples from an anticubital vein were collected


under standardised conditions in the morning, between 9 and
10 am, after a fasting period of ≥ 12 h and after resting in a
supine position for at least 15 min. Samples were immedi-
ately processed using a cooled centrifuge and aliquots were
stored at − 70 °C until analysis.

2.5. Statistical analysis

Results are given as mean ± SEM. To analyse relationships


between variables, Student's t-test and simple regression
analyses were performed (StatView 4.5, Abacus Concepts
Inc., Berkley CA, USA). Parameters that are not normally
distributed were log-transformed to allow a parametric sta-
tistical approach. Median and inter-quartile range (IQR] are
reported for these variables. A p-value of b0.05 was con-
Fig. 1. A: Appetite score (visual analogue scale) in COPD patients with and
sidered to be significant. If blood results were below the limit
without cachexia in comparison to healthy control subjects. B: Weight loss
of detectability of a test, the lower limit of detection was in COPD patients with and without cachexia.
recorded.

3. Results p b 0.05). Age, height and drug therapy were similar between
subgroups (p N 0.2).
3.1. Patient characteristics
3.2. Analysis of anorexia
The main clinical characteristics of the study population
are presented in Table 1. COPD Patients and control subjects The analysis of the extent of anorexia using the visual
were similar for age and global parameters of body com- appetite score revealed that COPD patients in comparison to
position but patients had impaired respiratory function healthy controls had a 36% lower appetite (p = 0.001). Ca-
parameters. Comparing patient subgroups of cachectic and chectic COPD patients showed a further significant reduc-
non-cachectic patients, COPD patients with cachexia showed tion of appetite compared to non-cachectic patients (− 44%,
further decreased respiratory (FEV1, FVC, and FEV1/FVC; all p b 0.001; Fig. 1A). Observed weight loss in the patient

Table 1
Clinical status and lung function test of cachectic and non-cachectic COPD patients in comparison to healthy controls
Healthy controls All COPD p, controls vs all ncCOPD p, ncCOPD vs cCOPD p, controls vs p, cCOPD
n = 15 n = 103 COPD n = 69 controls n = 34 cCOPD vs ncCOPD
Age, years 54.5 ± 1.4 59.8 ± 1.3 0.1 60.7 ± 1.6 0.08 58.0 ± 2.1 0.4 0.3
Weight, kg 71.4 ± 2.8 66.9 ± 1.7 0.3 72.8 ± 2.1 0.8 55.0 ± 1.7 b0.001 b0.001
Height, cm 169 ± 3.0 169 ± 1.0 0.9 169 ± 1.1 0.9 170 ± 1.7 0.7 0.5
Body mass index, 25.2 ± 1.1 23.4 ± 0.6 0.3 25.6 ± 0.7 0.8 19.0 ± 0.5 b0.001 b0.001
kg/m2
Weight loss, % 0.0 ± 0.0 5.2 ± 0.8 b0.02 0.3 ± 0.2 0.8 15.4 ± 1.2 b0.001 b0.001
Appetite 8.5 ± 0.1 5.4 ± 0.2 0.001 6.3 ± 0.2 b0.001 3.5 ± 0.3 b0.001 b0.001
FEV1, % 106.0 ± 3.2 38.3 ± 1.7 b0.001 42.0 ± 2.2 b0.001 30.7 ± 2.3 b0.001 b0.002
FVC, % 102.9 ± 2.5 56.1 ± 1.9 b0.001 58.7 ± 2.5 b0.001 50.5 ± 2.8 b0.001 b0.02
FEV1 / FVC, % 102.9 ± 1.7 60.7 ± 1.6 b0.001 65.0 ± 2.0 b0.001 52.0 ± 2.1 b0.001 b0.001
cCOPD indicates cachectic and ncCOPD non-cachectic COPD patients.
Data are mean ± SEM.
86 F. Koehler et al. / International Journal of Cardiology 119 (2007) 83–89

Table 2
Hormonal and inflammatory parameters of cachectic and non-cachectic COPD Patients
Healthy controls All COPD patients p, controls ncCOPD p, ncCOPD cCOPD p, controls p, cCOPD
N = 15 vs COPD vs controls vs cCOPD vs ncCOPD
GH, μg/ml 383 ± 213 1154 ± 174 n=80 b0.001 912 ± 158 n=54 b0.005 1658 ± 412 n=26 b0.001 0.16
IGF-1, ng/ml 132 ± 11 130 ± 7 n=75 0.9 133 ± 9 n=53 0.9 118 ± 11 n=22 0.5 0.3
Glucose (mg/dl) 94 ± 5.0 122 ± 3.9 b0.01 125 ± 4.8 b0.1 115 ± 5.9 0.058 0.2
Insulin, μIU/ml 24.8 ± 6.6 36.1 ± 4.4 n=80 0.3 40.3 ± 5.6 n=55 0.1 26.9 ± 6.7 n=25 0.8 b0.02
HOMA 6.1 ± 1.6 10.4 ± 1.3 n=80 0.18 12.2 ± 1.7 n=55 0.06 6.4 ± 1.9 n=25 0.9 b0.05
Cortisol, nmol/l 216.8 ± 27.4 399.1 ± 25.3 n=95 b0.01 391.0 ± 32.8 n=63 =0.01 415.1 ± 39.1 n=32 b0.01 0.7
Testosterone, nmol/l 5.13 ± 1.41 8.55 ± 1.0 n=80 0.2 8.0 ± 1.2 n=53 0.3 9.72 ± 1.8 n=27 0.1 0.4
DHEA, μmol/l n.a. 11.08 ± 2.83 n=44 12.41 ± 4.32 n=26 9.17 ± 3.05 n=18 0.6
TNF-α, pg/ml n.a. 4.8 ± 0.7 n=76 3.9 ± 0.6 n=50 6.5 ± 1.5 n=26 0.06
IL-6, pg/ml 2.0 ± 0.0 12.24 ± 2.4 n=76 b0.001 8.26 ± 1.51 n=49 b0.001 19.5 ± 6.0 n=27 b0.001 b0.05
IL-10, pg/ml 5.0 ± 0.0 4.35 ± 0.42 n=66 b0.05 4.45 ± 0.40 n=44 0.2 4.15 ± 1.0 n=22 b0.01 0.1
cCOPD indicates cachectic and ncCOPD non-cachectic COPD patients.
Data are shown as mean ± SEM.

population was significantly more severe than in control (cortisol, DHEA, testosterone) were not different between
subjects (p b 0.02), however, this was driven by a significant cachectic and non-cachectic COPD patients.
weight loss (− 10.3 ± 1.0 kg) in the cachectic population only Cachectic COPD patients presented with increased ratios
(− 15.4%; vs controls: p b 0.001) while no significant weight of IL-6/IGF-1 (p b 0.05), IL-6/insulin (p b 0.01), IL-6/IL-10
reduction (0.2 ± 0.1 kg) was seen in non-cachectic patients (p b 0.05), and TNF/insulin (p b 0.01) compared to non-
(− 0.3%; vs controls: p N 0.5; Fig. 1B). This accords with a cachectic COPD patients.
decreased BMI in cachectic vs non-cachectic COPD patients
(19.0 ± 0.5 vs 25.6 ± 0.7, p b 0.001) whereas BMI was not 3.4. Relation between appetite and catabolic/anabolic
different between ncCOPD and controls (p N 0.5). imbalance

3.3. Hormonal imbalance and inflammatory parameters In the total patient population of COPD patients
significant correlations between appetite and parameters of
Data on hormonal balance in controls vs patients and in body composition were revealed (appetite vs BMI [r = 0.48,
cachectic/non-cachectic patient subgroups are shown in p b 0.0001] and vs weight loss [r = − 0.58, p b 0.0001,
Table 2. Hormonal derangement was observed in COPD n = 103]; (Fig. 2). Furthermore, significant correlations
patients compared to controls but metabolic balance was between appetite and measures of catabolic anabolic
further impaired in cachectic vs non-cachectic COPD. Indi- imbalance were observed as indicated by the IGF-1/GH
cation of acquired GH resistance was seen in COPD patients ratio (r = 0.34, p b 0.005, n = 75), the IL-6/insulin ratio (r =
compared to controls and was most pronounced in cachectic − 0.46, p b 0.0001, n = 73), TNF/insulin ratio (r = − 0.33,
COPD. GH levels were highest in cachectic COPD (p b 0.05 p = 0.01, n = 54) and IL-6/IGF-1 ratio (r = −0.29, p b 0.02,
for ANOVA, Table 2) with no significant differences in IGF- n = 69; Fig. 3).
1 levels. Accordingly, the IGF-1/GH ratio was lowest in
cachectic COPD (p b 0.002 for ANOVA, Table 3). 4. Discussion
Non-cachectic COPD Patients presented elevated levels
of fasting insulin. HOMA insulin sensitivity was similar in This study demonstrates a significant prevalence of
cachectic COPD compared to controls but impaired in non- cachexia occurring in COPD and shows that anorexia is
cachectic COPD (Table 2). Plasma levels of steroid hormones frequently present in patients with COPD and cachexia. The

Table 3
Ratios of catabolic and anabolic hormones
Healthy controls n = 15 All COPD patients p, controls ncCOPD p, ncCOPD cCOPD p, controls p, cCOPD
vs all COPD vs controls vs cCOPD vs ncCOPD
IGF-1/GH 1560 ± 342 543 ± 74 n=73 b0.001 570 ± 85 n=53 b0.01 474 ± 154 n=20 b0.001 0.09
IL-6/IGF-1 0.017 ± 0.001 0.116 ± 0.027 n=69 b0.001 0.074 ± 0.014 n=47 0.3 0.206 ± 0.077 n=22 b0.01 0.01
IL-6/insulin 0.17 ± 0.03 1.26 ± 0.36 n=73 0.17 0.60 ± 0.18 n=55 0.6 2.60 ± 0.98 n=25 b0.01 b0.01
IL-6/IL-10 0.4 ± 0.0 5.91 ± 1.69 n=62 b0.001 3.62 ± 1.25 n=41 b0.001 10.4 ± 4.27n=21 b0.001 b0.02
TNF/IGF-1 n.a. 0.048 ± 0.007 n=50 0.037 ± 0.006 n=34 0.074 ± 0.015 n=16 b0.01
TNF/Insulin n.a. 0.29 ± 0.038 n=54 0.24 ± 0.47 n=36 0.39 ± 0.06 n=18 b0.01
cCOPD indicates cachectic and ncCOPD non-cachectic COPD patients.
Data are shown as mean ± SEM.
F. Koehler et al. / International Journal of Cardiology 119 (2007) 83–89 87

strength and endurance, decreased diaphragmatic size [22]


with increased work of breathing, and predisposition to death
due to heart failure play an important role triggering weight
loss in COPD. The lean tissue mass in COPD patients is
influenced by a number of interacting factors such as aging,
medication, tissue hypoxia and inactivity [23]. Therefore the
development of cachexia in COPD is a multifactorial process
including tissue hypoxia, systemic inflammation, physical
inactivity, neurohormonal activation and hypermetabolism
[24,25].

Fig. 2. The relationship of the appetite score (visual analogue scale) to


clinical/humoral parameters in COPD: A: Appetite vs % weight loss
(n = 103), B: Appetite vs BMI (n = 103), C: Appetite vs IGF-1/GH ratio
(n = 75).

degree of appetite in COPD patients is related to body mass


index, inflammation and catabolic/anabolic imbalance.
The pathogenesis and clinical manifestations of COPD
are not restricted to pulmonary inflammation and airway
remodelling [18]. Involuntary weight loss occurs frequently
in a significant number of patients, particularly in those
Fig. 3. The relationship of the appetite score (visual analogue scale) to ratios
suffering from emphysema [19,20], and presents an of catabolic and anabolic parameters in COPD. A: Appetite vs IL-6/insulin
independent risk factor of impaired prognosis [21]. Several ratio (n = 73), B: Appetite vs TNF/insulin ratio (n = 54), C: Appetite vs IL-6/
studies have shown that decreased respiratory muscle IGF-1 ratio (n = 69).
88 F. Koehler et al. / International Journal of Cardiology 119 (2007) 83–89

Further hypotheses for the development of cachexia in- resistance and proinflammatory immune activation. Reduced
clude inadequate dietary intake, increased resting energy appetite is part of the catabolic/anabolic imbalance in the
expenditure and diet induced thermogenesis [2,3]. Anorexia process of wasting in COPD. More pathophysiological
with loss of appetite might be one reason for an inadequate studies are needed to confirm the role of anorexia in the
food intake [2]. The results of the present study demonstrate pathogenesis of pulmonary cachexia and to develop therapies
that appetite is low in general in COPD patients and espe- that impact on this systemic manifestation of COPD.
cially in those who are cachectic. The appetite was quantified
by a visual analogue scale that assesses the subjective desire References
to eat. This test is widely used for assessment of appetite and
has been evaluated as a reliable method with good re- [1] Wouters EF, Creutzberg EC, Schols AM. Systemic effects in COPD.
producibility and feasibility [26,27]. In another study, we Chest May 2002;121(5 Suppl):127S–30S.
have applied the same test to healthy subjects (mean appetite [2] Engelen MP, Schols AM, Does JD, Wouters EF. Skeletal muscle
weakness is associated with wasting of extremity fat-free mass but not
score 8) and chronic heart failure patients with and without
with airflow obstruction in patients with chronic obstructive
cachexia (mean appetite score 6.5 and 7.5, respectively pulmonary disease. Am J Clin Nutr 2000;71:733–8.
(Anker et al, unpublished observation). The results of the [3] Shoup R, Dalsky G, Warner S, et al. Body composition and health-
present study show that appetite is reduced by 45% in ca- related quality of life in patients with obstructive airways disease. Eur
chectic vs non-cachectic COPD patients (p b 0.001). Strong Respir J 1997;10:1576–8150.
[4] Mostert R, Goris A, Weling-Scheepers C, Wouters EF, Schols AM.
correlations of the appetite score with body mass index,
Tissue depletion and health related quality of life in patients with
weight loss, and measurements of GH resistance, catabolic/ chronic obstructive pulmonary disease. Respir Med Sep 2000;94
anabolic imbalance and immune activation were observed. (9):859–67.
These data suggest that impaired appetite might be involved [5] Palange P, Forte S, Onorati P, et al. Effect of reduced body weight on
in the complex regulation of body composition in chronic muscle aerobic capacity in patients with COPD. Chest 1998;114:12–8.
[6] Wilson DO, Rogers RM, Wright EC, Anthonisen NR. Body weight in
inflammatory disease and the development of cachexia.
chronic obstructive pulmonary disease. The National Institutes of
The observed relationships are in line with the pathophys- Health Intermittent Positive-Pressure Breathing Trial. Am Rev Respir
iologic understanding of cachexia in COPD, resulting not Dis 1989;139:1435–8.
only from increased energy expenditure but also from reduced [7] Schols AM, Broekhuizen R, Weling-Scheepers CA, Wouters EF. Body
dietary intake due to loss of appetite. These data indicate that composition and mortality in chronic obstructive pulmonary disease.
Am J Clin Nutr Jul 2005;82(1):53–9.
treatment of anorexia itself may be beneficial in COPD
[8] Baarends EM, Schols AM, Westerterp KR, Wouters EF. Total daily
cachexia. The use of appetite stimulants (like megastrol energy expenditure relative to resting energy expenditure in clinically
acetate [28]) has been shown to be beneficial in cancer stable patients with COPD. Thorax 1997;52:780–5.
cachexia and AIDS cachexia. This could be tested in COPD as [9] Goldstein S, Askanazi J, Weissman C, Thomashow B, Kinney JM.
well. Energy expenditure in patients with chronic obstructive pulmonary
disease. Chest 1987;91:222–4.
Furthermore, our findings support the hypothesis that the
[10] Schols AM, Fredrix EW, Soeters PB, Westerterp KR, Wouters EF.
protein catabolic state in pulmonary cachexia is related to an Resting energy expenditure in patients with chronic obstructive
increased host inflammatory response and a hormonal pulmonary disease. Am J Clin Nutr 1991;54:983–7.
derangement. This phenomenon may be due to a hypoxia [11] Schols AM, Soeters PB, Mostert R, Saris WH, Wouters EF. Energy
related alteration in intermediary metabolism shifting the balance in chronic obstructive pulmonary disease. Am Rev Respir Dis
1991;143:1248–52.
body composition towards wasting of fat free mass.
[12] Sridhar MK, Carter R, Lean ME, Banham SW. Resting energy
Finally, increased levels of growth hormone were de- expenditure and nutritional state of patients with increased oxygen cost
tected in cachectic COPD patients indicating a growth hor- of breathing due to emphysema, scoliosis and thoracoplasty. Thorax
mone resistance. This is suggestive of presence of acquired 1994;49:781–5.
GH resistance, which is also seen in CHF patients [29]. [13] Ferreira IM, Brooks D, Lacasse Y, Goldstein RS, White J. Nutritional
supplementation for stable chronic obstructive pulmonary disease.
Anabolic therapy with higher doses of GH should therefore
Cochrane Database Syst Rev 2005(2) [CD000998].
be tested in cachectic COPD patients [30]. Alternatively, [14] Creutzberg EC, Schols AM, Weling-Scheepers CA, Buurman WA,
novel therapeutic options such as the growth hormone Wouters EF. Characterization of nonresponse to high caloric oral
releasing peptide ghrelin might provide additional benefit to nutritional therapy in depleted patients with chronic obstructive
those patients with severely impaired body composition. pulmonary disease. Am J Respir Crit Care Med 2000;161:745–52.
[15] Standards for the diagnosis and care of patients with chronic
Preliminary data from an open label pilot study show pro-
obstructive pulmonary disease. American Thoracic Society. Am J
mising results as muscle wasting, functional capacity, and Respir Crit Care Med 1995;152:S77–S121.
sympathetic status have all been improved following a [16] Anker SD, Ponikowski P, Varney S, et al. Wasting as independent risk
3 week iv administration of ghrelin in seven cachectic COPD factor for mortality in chronic heart failure. Lancet 1997;349:1050–3.
patients [31]. Further studies are required to verify whether [17] Matthews DR, Hosker JP, Rudenski AS, Naylor BA, Treacher DF,
Turner RC. Homeostasis model assessment: insulin resistance and
this concept may indeed translate into meaningful benefit for
beta-cell function from fasting plasma glucose and insulin concentra-
cachectic COPD patients. tions in man. Diabetologia 1985;28:412–9.
In summary, cachexia in COPD is a multifactorial disease [18] Wouters EFM, Creutzberg EC, Schols AMWJ. Systemic effects in
process characterised by loss of appetite, growth hormone COPD. Chest 2002;121:127S–30S.
F. Koehler et al. / International Journal of Cardiology 119 (2007) 83–89 89

[19] Donahoe M, Rogers RM, Cottrell JJ. Is loss of body weight in chronic [27] Hollen PJ, Gralla RJ, Kris MG, McCoy S, Donaldson GW, Moinpour
obstructive pulmonary disease patients with emphysema secondary to CM. A comparison of visual analogue and numerical rating scale
low tissue oxygenation? Respiration 1992;59(Suppl 2):33–9. formats for the Lung Cancer Symptom Scale (LCSS): does format
[20] Sridhar MK. Why do patients with emphysema lose weight? Lancet affect patient ratings of symptoms and quality of life? Qual Life Res
1995;345:1190–1. 2005;14:837–40.
[21] Schols AM. Pulmonary cachexia. Int J Cardiol 2002;85:101–10. [28] Karcic E, Philpot C, Morley JE. Treating malnutrition with megestrol
[22] Arora NS, Rochester DF. Effect of body weight and muscularity on acetate: literature review and review of our experience. J Nutr Health
human diaphragm muscle mass, thickness, and area. J Appl Physiol Aging 2002;6:191–200.
1982;52:64–70. [29] Cicoira M, Zanolla L, Rossi A, et al. Failure of aldosterone suppression
[23] Donahoe M. Nutritional support in advanced lung disease. The despite angiotensin-converting enzyme (ACE) inhibitor administration
pulmonary cachexia syndrome. Clin Chest Med 1997;18:547–61. in chronic heart failure is associated with ACE DD genotype. J Am
[24] Congleton J. The pulmonary cachexia syndrome: aspects of energy Coll Cardiol 2001;37:1808–12.
balance. Proc Nutr Soc 1999;58:321–8. [30] Cicoira M, Kalra PR, Anker SD. Growth hormone resistance in chronic
[25] Agust AG, Gari PG, Sauleda J, Busquets X. Weight loss in chronic heart failure and its therapeutic implications. J Card Fail 2003;9:219–26.
obstructive pulmonary disease. Mechanisms and implications. Pulm [31] Nagaya N, Itoh T, Murakami S, et al. Treatment of cachexia with
Pharmacol Ther 2002;15:425–32. ghrelin in patients with COPD. Chest 2005;128:1187–93.
[26] Flint A, Raben A, Blundell JE, Astrup A. Reproducibility, power and
validity of visual analogue scales in assessment of appetite sensations in
single test meal studies. Int J Obes Relat Metab Disord 2000;24:38–48.

You might also like