Вы находитесь на странице: 1из 95

Nutrition in

Gastrointestinal and
Liver Diseases

Editors
Christos Dervenis, Athens
Herbert Lochs, Berlin

12 figures, 18 tables, 2003

Basel 폷 Freiburg 폷 Paris 폷 London 폷 New York 폷


Bangalore 폷 Bangkok 폷 Singapore 폷 Tokyo 폷 Sydney
S. Karger Drug Dosage All rights reserved.
Medical and Scientific Publishers The authors and the publisher have exerted every effort to en- No part of this publication may be translated into other
sure that drug selection and dosage set forth in this text are in languages, reproduced or utilized in any form or by any means,
Basel 폷 Freiburg 폷 Paris 폷 London
accord with current recommendations and practice at the time electronic or mechanical, including photocopying, recording,
New York 폷 Bangalore 폷 Bangkok of publication. However, in view of ongoing research, changes microcopying, or by any information storage and retrieval
Singapore 폷 Tokyo 폷 Sydney in government regulations, and the constant flow of informa- system, without permission in writing from the publisher or, in
tion relating to drug therapy and drug reactions, the reader is the case of photocopying, direct payment of a specified fee to
urged to check the package insert for each drug for any change the Copyright Clearance Center (see ‘General Information’).
in indications and dosage and for added warnings and precau-
tions. This is particularly important when the recommended © Copyright 2003 by S. Karger AG,
agent is a new and/or infrequently employed drug. P.O. Box, CH–4009 Basel (Switzerland)
Printed in Switzerland on acid-free paper by
Reinhardt Druck, Basel
ISBN 3–8055–7657–9

Fax + 41 61 306 12 34
E-Mail karger@karger.ch
www.karger.com
Vol. 21, No. 3, 2003

Contents

196 Editorial 252 Sugar Intake, Taste Changes and Dental Health
Lochs, H. (Berlin); Dervenis, C. (Athens) in Crohn’s Disease
Schütz, T.; Drude, C.; Paulisch, E.; Lange, K.-P.; Lochs, H.
(Berlin)
Review Articles
258 Serum Mineral Levels in Children with Intestinal
198 What We Have Learned about Cachexia in Parasitic Infection
Gastrointestinal Cancer Olivares, J.L.; Fernández, R.; Fleta, J.; Rodríguez, G.; Clavel, A.
(Zaragoza)
Palesty, J.A.; Dudrick, S.J. (Waterbury, Conn.)
262 Impact of Body Mass Index on Fasting Blood Glucose
214 Nutritional Support in Acute Pancreatitis
Concentration among Helicobacter pylori Carriers
Avgerinos, C.; Delis, S.; Rizos, S.; Dervenis, C. (Athens)
Kyriazanos, I.D.; Sfiniadakis, I.; Dimakos, P.; Gizaris,V.;
220 Nutrition and Inflammatory Bowel Disease: Its Datsakis, K.; Dafnopoulou, A. (Athens)
Relation to Pathophysiology, Outcome and Therapy
266 Selenium Is Depleted in Crohn’s Disease on Enteral
Gassull, M.A. (Badalona)
Nutrition
228 Factors Enhancing Intestinal Adaptation after Kuroki, F.; Matsumoto, T.; Iida, M. (Fukuoka City)
Bowel Compensation
271 L-Carnitine in the Treatment of Mild or Moderate
Botsios, D.S.; Vasiliadis, K.D. (Thessaloniki)
Hepatic Encephalopathy
237 Helicobacter pylori Infection, Vitamin B12 and Malaguarnera, M.; Pistone, G.; Astuto, M.; Dell’Arte, S.;
Homocysteine. A Review Finocchiaro, G.; Lo Giudice, E.; Pennisi, G. (Catania)
Dierkes, J.; Ebert, M.; Malfertheiner, P.; Luley, C. (Magdeburg) 276 Fructose Breath Hydrogen Test – Is It Really
a Harmless Diagnostic Procedure?
Müller, P. (Leisnig/Leipzig); Meier, C.; Böhme, H.J.; Richter, T.
Original Papers (Leipzig)
245 Prevalence of Malnutrition in Hospitalized Medical 279 Screening for Iron Overload in the Turkish Population
Patients: Impact of Underlying Disease Barut, G.; Balci, H. (Istanbul); Bozdayi, M. (Ankara); Hatemi, I.;
Pirlich, M.; Schütz, T.; Kemps, M.; Luhman, N.; Ozcelik, D.; Senturk, H. (Istanbul)
Burmester, G.-R.; Baumann, G. (Berlin); Plauth, M. (Dessau);
Lübke, H.J.; Lochs, H. (Berlin)
286 Author Index and Subject Index

© 2003 S. Karger AG, Basel

Fax + 41 61 306 12 34 Access to full text and tables of contents,


E-Mail karger@karger.ch including tentative ones for forthcoming issues:
www.karger.com www.karger.com/ddi_issues
Editorial

Dig Dis 2003;21:196–197


DOI: 10.1159/000074105

Malnutrition – The Ignored Risk Factor


Herbert Lochs Christos Dervenis

Nutritional therapy, once a cornerstone of medical treatment, has and fat, or indices including dynamic parameters, like weight loss or
lost its attractivity in favor of drug treatment, molecular genetic a stress factor, do correlate better with the patients’ prognosis than
interventions or other high-tech therapies. As a consequence, data plain body weight or body mass index. From these studies, several
about nutritional status or dietary habits are missing in many patient nutritional indices have been developed which allow conclusions
charts. This development reflects the opinion of many doctors that about the prognosis and might therefore be used to define patients
nutrition does not greatly affect the course of diseases, and that nutri- with an indication for nutritional therapy.
tional intervention is laborious but much less effective than other The subjective Global Assessment (SGA) is one of the most wide-
forms of therapy. In view of this trend, it seems interesting to analyze ly used nutritional indices [1]. It consists of a patient history part
the importance of nutrition in different dieseases. (recent weight loss, changes in eating habits, gastrointestinal symp-
Of course, it is well accepted that nutrition is an important factor toms, physical fitness and stress factor) and a physical examination
for the therapy and prognosis in several diseases, such as diabetes (body weight, subcutaneous fat mass, muscle atrophy, edema). The
mellitus, hyperlipidemia, obesity or hemochromatosis. However, the subjective global assessment is easy to calculate and therefore conve-
most basic nutritional disturbance – malnutrition – is frequently nient to use in a clinical setting. A similar index (NRS 2002) has
ignored since it is considered as a complication of the disease process, recently been developed by the European Society of Parenteral and
with little bearing on the prognosis and little possibility for therapeut- Enteral Nutrition [2]. It also includes a patient history (weight loss,
ic intervention. However, an analysis of the literature reveals that reduced dietary intake) physical parameters (body mass index) and a
malnutrition is an independent risk factor in many disease processes disease severity factor. Like the SGA, it can be calculated quickly and
and that treatment of malnutrition can indeed improve the patients’ is therefore useful in the clinical situation. It is important to mention
prognosis. that malnutrition does not necessarily correlate with a low body
Such an analysis has to address several questions, mainly the mass. Changes in body composition with an increased fat mass and
prevalence and diagnosis of malnutrition and its impact on the decreased body cell mass pose a similar risk to the patient than overt
patients’ prognosis. It should set the stage for the papers in this issue malnutrition with reduced body mass, as has been shown in a recent
of Digestive Diseases, which deals with nutritional questions. investigation [3].

Prevalence and Diagnosis of Malnutrition in Impact of Malnutrition on Prognosis


Hospitalized Patients
Malnutrition has a profound impact on a patient’s prognosis.
An evaluation of the nutritional status of hospitalized patients in Malnourished patients have a higher risk of postoperative complica-
different countries shows a prevalence of malnutrition between 20 tions when compared to well-nourished patients and a longer hospi-
and 60% of admitted patients (table 1). The prevalence is not differ- tal stay [3–7]. Furthermore the long-term mortality of malnourished
ent among different countries; however, a subgroup analysis showed patients is much higher than that of comparable well-nourished
that geriatric patients as well as patients with malignant diseases car- patients [8]. This is particularly important since physicians in the
ry the highest risk of becoming malnourished. hospital usually see the patient only for 7–10 days during his/her hos-
The parameters used do diagnose malnutrition varied consider- pital stay. As has been shown in several studies, posthospital mortali-
ably in the different studies. In older studies, usually static parame- ty is significantly increased in malnourished patients. The fact that
ters of the nutritional status, like body mass index or anthropometric this complication of malnutrition is developing so late explains why
measurements, were used. However, later on, it became clear that hospital physicians are not more aware of malnutrition-associated
body composition reflecting the percentage of lean body mass, water risks.

© 2003 S. Karger AG, Basel Prof. Dr. Herbert Lochs


ABC 0257–2753/03/0213–0196$19.50/0 Universitätsklinikum Charité
Fax + 41 61 306 12 34 Medizinische Klinik mit Schwerpunkt Gastroenterologie, Hepatologie und Endokrinologie
E-Mail karger@karger.ch Accessible online at: Schumannstrasse 20/21, DE−10098 Berlin (Germany)
www.karger.com www.karger.com/ddi Tel. +49 30 450 514021, Fax +49 30 450 514923, E-Mail herbert.lochs@charite.de
Table 1. Prevalence of malnutrition in hospitalized patients prospectively calculated the expenses for 100 patients. Well-nour-
ished patients had a median stay of 10 days whereas malnourished
Authors n Country Disease Malnour- patients stayed for 16.6 days. The actual cost was USD 7,692 for the
ished, % well-nourished and USD 16,691 for the malnourished patients. On a
DRG basis, payment from well-nourished patients was USD 4,352
Bistrian et al. (1974) [12] 131 USA surgery 50 and USD 5,124 for malnourished patients. This clearly shows that
Bistrian et al. (1976) [13] 251 USA general medicine 44
malnutrition is not just a risk for the patient but also a financial risk
Hill et al. (1977) [14] 105 UK surgery 48
Weinsier et al. (1979) [15] 134 USA general medicine 48 for the hospital. A study by Braunschweig et al. [11] who followed the
Coats et al. (1993) [16] 228 USA general medicine 38 expenses for 404 patients who were hospitalized for more than 7 days
McWhirter and Pennington is especially interesting. Patients were divided into a reference group
(1994) [17] 500 UK multidisciplinary 40 which had not lost weight during their hospital stay and a group who
Cederholm et al. (1995) [6] 205 S geriatric 20 lost weight independently of their initial nutritional status. The con-
Naber et al. (1997) [5] 155 NL internal medicine 45–62
trol group generated an average expenditure of USD 28,631, whereas
Bruun et al. (1999) [18] 244 USA surgery 39
Edington et al. (2000) [19] 850 UK multidisciplinary 20
the group who lost weight generated an average expenditure of USD
Waitzberg et al. (2001) [20] 4,000 Brazil multidisciplinary 20 45,762.
Pirlich et al. (2003) [21] 803 Germany multidisciplinary 22 This issue of Digestive Diseases is devoted to nutritional prob-
lems. A number of papers deal with problems of malnutrition in dif-
ferent diseases. Besides the prevalence of malnutrition in hospital-
ized patients, questions concerning the impact of malnutrition in gas-
trointestinal cancer and acute pancreatitis as well as inflammatory
However, there are also economic impacts of malnutrition during bowel disease are discussed. Furthermore, nutritional therapy has
the hospital stay. Reilly et al. [9] calculated the patient costs of 771 proved to be effective not just as treatment of preexisting malnutri-
hospitalized patients in Internal Medicine and Surgery. They defined tion but also to influence disease processes, as has been shown in
malnourished patients on a history of weight loss, low albumin and inflammatory bowel disease, hepatic encephalopathy and other gas-
total lymphocyte count as well as low body mass index. The median trointestinal diseases. By raising their attention to nutritional prob-
expenditures for malnourished patients amounted to USD 11,217 lems, doctors will hopefully increasingly again include nutritional
versus USD 7,660 for well-nourished patients. Robinson et al. [10] therapy in patient care.

References

1 Detsky AS, McLaughlin JR, Baker JP, John- for patients undergoing gastrointestinal sur- 15 Weinsier RL, Hunker EM, Krumdieck CL,
ston N, Whittaker S, Mendelson RA, Jeejeeboy gery. JPEN J Parenter Enteral Nutr 1987;11: Butterworth CE Jr: Hospital malnutrition. A
KN: What is subjective global assessment of 440–446. prospective evaluation of general medical pa-
nutritional status? JPEN J Parenter Enteral 8 Pirlich M, Schütz T, Gastell S, Lochs H: Mal- tients during the course of hospitalization. Am
Nutr 1987;11:8–13. nutrition affects long-term prognosis in hospi- J Clin Nutr 1979;32:418–426.
2 Kondrup J, Allison SP, Elia M, Vellas B, Plauth talized patients. Gastroenterology 2002;122: 16 Coats KG, Morgan SL, Bartolucci AA, Wein-
M: ESPEN guidelines for nutrition screening 363. sier RL: Hospital-associated malnutrition: A
2002. Clin Nutr 2003;22:415–421. 9 Reilly JJ Jr, Hull SF, Albert N, Waller A, Brin- reevaluation 12 years later. J Am Diet Assoc
3 Kyle UG, Pirlich M, Morabia A, Lochs H, gardener S: Economic impact of malnutrition: 1993;93:27–33.
Schuets T, Pichard C: Abnormal body compo- A model system for hospitalized patients. 17 McWhirter JP, Pennington CR: Incidence and
sition is associated with increased length of JPEN J Parent Enteral Nutr 1988;12:371–376. recognition of malnutrition in hospital. BMJ
hospital stay: A controlled population study in 10 Robinson G, Goldstein M. Levine GM: Impact 1994;308:945–948.
Switzerland and Germany, submitted to Clini- of nutritional status on DRG length of stay. 18 Bruun LI, Bosaeus I, Bergstad I, Nygaard K:
cal Nutrition. JPEN J Parenter Enteral Nutr 1987;11:49–51. Prevalence of malnutrition in surgical patients:
4 Selberg O, Böttcher J, Tusch G, Pichlmayr R, 11 Braunschweig C, Gomez S, Sheean PM: Im- Evaluation of nutritional support and docu-
Henkel E, Müller MJ: Identification of high- pact of declines in nutritional status on out- mentation. Clin Nutr 1999;18:141–147.
and low-risk patients before liver transplanta- comes in adult patients hospitalized for more 19 Edinton J, Boorman J, Durrant ER, Perkins A,
tion: A prospective cohort study of nutritional than 7 days. J Am Diet Assoc 2000;100:1316– Giffin CV, James R, Thomson JM, Oldroyd
and metabolic parameters in 150 patients. He- 1322. JC, Smith JC, Torrance AD, Blackshaw V,
patology 1997;25:652–657. 12 Bistrian BR, Blackburn GL, Hallowell E, Hed- Green S, Hill CJ, Berry C, McKenzie C, Vicca
5 Naber TH, Schermer T, de Bree A, Nusteling dle R: Protein status of general surgical pa- N, Ward JE, Coles SJ: Prevalance of malnutri-
K, Eggink L, Kruimel JW, Bakkeren J, van tients. JAMA 1974;230:858–860. tion on admission to four hospitals in England.
Heereveld H, Katan MB: Prevalence of malnu- 13 Bistrian BR, Blackburn GL, Vitale J, Cochran The Malnutrition Prevalance Group. Clin Nutr
trition in nonsurgical hospitalized patients and D, Maylor J: Prevalence of malnutrition in gen- 2000;19:191–195.
its association with disease complications. Am eral medical patients. JAMA 1976;235:1567– 20 Waitzberg DL, Caiaffa WT, Correia MI: Hos-
J Clin Nutr 1987;66:1232–1239. 1570. pital malnutrition: The Brazilian National Sur-
6 Cederholm T, Jagren C, Hellstrom K: Outcome 14 Hill GL, Blackett RL, Pickford I, Burkinshaw vey (IBRANUTRI): A study of 4,000 patients.
of protein-energy malnutrition in elderly medi- L, Young GA, Warren JV, Schorath CJ, Mor- Nutrition 2001;17:573–580.
cal patients. Am J Med 1995;98:67–74. gan DB: Malnutrition in surgical patients. An 21 Pirlich M, Schütz T, Kemps M, Luhman N,
7 Detsky AS, Baker JP, O’Rourke K, Johnston N, unrecognized problem. Lancet 1977;i:689– Burmester GR, Baumann G, Plauth M, Lübke
Whitwell J, Mendelson RA, Jeejeeboy KN: 692. HJ, Lochs H: Prevalence of malnutrition in
Predicting nutrition-associated complications hospitalized medical patients: Impact of un-
derlying disease. Dig Dis 2003;21:245–252.

Editorial Dig Dis 2003;21:196–197 197


Review Article

Dig Dis 2003;21:198–213


DOI: 10.1159/000073337

What We Have Learned about Cachexia


in Gastrointestinal Cancer
J.A. Palesty S.J. Dudrick
St. Mary’s Hospital, Yale University Affiliate, Department of Surgery, Waterbury, Conn., USA

Key Words mately leads to death. Therapeutic interventions have


Cancer W Cachexia W Cancer cachexia syndrome W met with little success, and, regardless of tremendous
Malnutrition W Anorexia W Gastrointestinal cancer efforts throughout the decades, the exact nature of the
mediators responsible for cancer cachexia remain elu-
sive. The pathogenesis of cancer cachexia appears to be
Abstract related to proinflammatory cytokines, alterations in the
It is appreciated widely by clinicians that significant mal- neuroendocrine axis and tumor-derived catabolic fac-
nutrition accompanies malignant processes in approxi- tors. Despite trials of conventional and/or aggressive
mately 50% of patients and eventually leads to severe nutritional support by a myriad of feeding techniques,
wasting which accounts for approximately 30% of can- patients with cancer cachexia have failed to gain consis-
cer-related deaths overall, 30–50% of deaths in patients tent significant benefits in terms of weight gain, function-
with gastrointestinal tract cancers, and up to 80% of al ability, quality of life or survival. Additionally, attempts
deaths in patients with advanced pancreatic cancer. The to ameliorate the abnormal clinical and metabolic fea-
body wasting known as cancer cachexia is a complex tures of cancer cachexia with a variety of pharmacologic
syndrome characterized by progressive tissue depletion agents have met with only limited success. Either until
and decreased nutrient intake that is manifested clinical- cancer of the gastrointestinal tract can be cured or until it
ly as inexplicable, recalcitrant anorexia and inexorable is possible to identify the exact causes and mechanisms
host weight loss. Decreased nutritional intake, increased of the cancer cachexia syndrome, the most realistic and
metabolic expenditure and dysfunctional metabolic pro- practical options currently are directed toward minimiz-
cesses, including hormonal and cytokine-related abnor- ing adverse gastrointestinal side effects or complications
malities, all appear to play roles in the development of of the malignant process and/or therapy, as well as
cancer cachexia. Although this condition of advanced increasing appetite, food intake and nutrient utilization in
protein-calorie malnutrition, sometimes described as the an effort to enhance quality of life and improve survival.
cancer anorexia-cachexia syndrome, is not entirely un- Copyright © 2003 S. Karger AG, Basel

derstood, it appears to be multifactorial, is a major cause


of morbidity and mortality in cancer patients, and ulti-

© 2003 S. Karger AG, Basel Stanley J. Dudrick, MD, FACS


ABC 0257–2753/03/0213–0198$19.50/0 St. Mary’s Hospital, Yale University Affiliate
Fax + 41 61 306 12 34 Department of Surgery, 56 Franklin Street
E-Mail karger@karger.ch Accessible online at: Waterbury, CT 06706 (USA)
www.karger.com www.karger.com/ddi Tel. +1 203 5746479, Fax +1 203 5975877, E-Mail sdudrick@stmh.org
Introduction greater than 10% weight loss [6, 7]. In a prospective study
of 280 cancer patients, malnutrition was related predomi-
Cachexia results from the inexorable and recalcitrant nantly to tumor type and site, with stomach and esopha-
progression of nutritional deterioration, which occurs in geal cancer patients demonstrating significantly higher
approximately 50% of patients with malignant disorders, degrees of malnutrition compared with the other groups.
and in about 80% of patients with cancers involving the Moreover, as expected, malnutrition was shown to in-
upper gastrointestinal tract. It is among the most debili- crease in severity as the disease advanced [8]. In another
tating and life-threatening aspects of cancer, and results in study of 365 patients with gastrointestinal cancer, almost
body mass wasting that accounts for up to 30% of cancer- 50% were shown to be malnourished. The incidence of
related deaths [1]. Clinically, the cancer cachexia syn- malnutrition was related to the site of the disease, and the
drome is characterized primarily by anorexia, early satie- stage of the disease was identified as a predictor of weight
ty, wasting, weight loss, weakness, fatigue, poor mental loss, with over 50% of stage III patients manifesting mal-
and physical performance, decreased capacity for wound nutrition [9].
healing, impaired immunologic function, and a compro- Although nutritional status is usually evaluated by a
mised quality of life, none of which are resolved by forced combination of clinical assessment and anthropometric
nutrient intake [1, 2]. Diminished nutritional intake, tests, including body weight, skin-fold thickness and mid-
increased metabolic expenditure and disordered or mal- arm circumference, most clinicians rely on body weight as
functioning metabolic processes, including hormonal and the major measure of nutritional status, using usual adult
cytokine-related abnormalities, all appear to play roles in body weight as a point of reference [2].
the development of cancer cachexia [1, 3]. Although this Cachexia should be expected if an involuntary or unex-
condition of advanced protein-calorie malnutrition, fre- pected weight loss of greater than 5% has occurred within
quently referred to as the cancer anorexia-cachexia syn- a previous 6-month period, especially when combined
drome, is not entirely understood, it appears to be multi- with muscle wasting. A weight loss of 10% or more ordi-
factorial, it is a major cause of morbidity and mortality in narily indicates severe depletion, but can also be used as a
cancer patients, and it ultimately leads to death [3, 4]. starting criterion for the anorexia-cachexia syndrome in
The origin of the word cachexia is both descriptive and obese patients. Body compartment analysis has shown
incriminating. It is derived from the Greek words kakos, that patients with cachexia lose approximately equal
meaning bad, and hexis, meaning condition, habit or state amounts of fat and fat-free mass. Losses of fat-free mass
of being [2, 3]. occur primarily from skeletal muscle and reflect decreases
Cancer cachexia was first reported as a commonly both in cellular mass and intracellular potassium concen-
occurring syndrome more than 70 years ago, when an tration [2, 10]. Cancer patients with documented 5%
autopsy series of 500 cancer patients documented that the weight loss have a shorter median survival rate than
immediate cause of death in cancer patients was second- patients with stable weight, respond poorly to chemother-
ary to inanition in 114 (22%) of the patients, and that up apy, and experience increased toxicity to chemotherapy
to two-thirds of this cadre of patients exhibited some [2, 7, 11].
degree of cachexia [5]. Since then, the scope of malnutri- How does cancer cachexia compare with other condi-
tion in the cancer patient has been studied in a wide vari- tions associated with weight loss, such as the anorexia
ety of patient groups. In a series of 3,047 patients enrolled which may occur in uncomplicated starvation, or that fol-
in Eastern Cooperative Oncology Group (ECOG) chemo- lowing a major injury or accompanying sepsis? Usually,
therapy protocols and who were all assessed for weight the non-cancer patient with acute starvation initially de-
loss before initiating chemotherapy, survival was signifi- velops rapid breakdown of protein from lean body mass,
cantly lower in the patients who demonstrated weight loss amino acid mobilization, gluconeogenesis and increased
when compared with those who had not lost any weight excretion of nitrogen in the urine. These early responses
prior to starting chemotherapy [6, 7]. In this study group, to the stressful stimulus are altered subsequently by the
patients with breast cancer or sarcomas had the lowest fre- body to conserve protein and energy. Addition of a major
quency of weight loss (31–40%), patients with colon can- injury or sepsis to simple starvation, or to the presence of
cer had an intermediate frequency of weight loss (48– a tumor, compounds the situation by increasing the rate
61%), and patients with pancreatic or gastric cancer had and duration of the catabolic response, and the associated
the highest frequency of weight loss (83–87%), with about high rate of tissue breakdown may persist for quite some
one-third of these patients having presented initially with time [12]. The patient with cancer cachexia exhibits fea-

What We Have Learned about Cachexia in Dig Dis 2003;21:198–213 199


Gastrointestinal Cancer
Table 1. Nutritional problems associated with the development of tures of both starvation and injury. However, in the can-
cancer cachexia (adapted from Shils [14]) cer patient, not only is food intake usually inadequate, but
the presence of the malignant tumor may decrease the
1 Anorexia and early satiety with progressive weight loss and
undernutrition ability of the patient to regulate the response to starvation.
2 Altered taste or dysgeusia causing hypophagia, food aversion or Cancer cachexia is more likely to resemble the condition
altered food intake produced by a major injury or sepsis than cachexia sec-
3 Alterations in protein, carbohydrate and fat metabolism ondary to simple starvation [13].
4 Increased energy expenditure despite decreased body mass
In addition to the multiple effects of cancer on the
5 Impaired food intake secondary to:
A. Mechanical obstruction of the gastrointestinal tract at any nutritional status of the patient, the antineoplastic thera-
level pies currently in use can have adverse effects contributing
B. Intestinal dysmotility induced by various tumors to the cachexia of the patient. Understanding the multiple
6 Malabsorption secondary to: nutritional, metabolic and physiologic changes that can
A. Deficiency or inactivation of pancreatic digestive enzymes
occur provides the basis for evaluating the requirements
B. Deficiency or inactivation of bile salts
C. Failure of ingested food to mix and interact effectively with for nutritional support in various clinical situations and
digestive enzymes for planning and providing support as indicated. Al-
D. Intestinal fistulas – internal and/or external though a localized tumor may exert various initial sys-
E. Infiltration of the small bowel wall or lymphatics and temic and generalized effects in a patient, as the tumor
mesentery by malignant cells
grows and metastasizes, these effects often become even
F. Blind loop syndrome with associated bacterial overgrowth
G. Malnutrition-induced villous hypoplasia in the small more obvious as a result of the increased tumor burden
intestine and its local and distant invasion. Additionally, some
7 Protein-losing enteropathy malignant tumors can produce effects at a distance from
8 Metabolic abnormalities induced by tumor-derived eutopic the original tumor or its metastases, further compounding
hormones or peptides
malnutrition [14]. The nutritional problems associated
A. Hypoglycemia induced by insulin-secreting tumors
B. Hyperglycemia induced by islet glucagonoma or somato- with neoplastic disease which can cause or contribute to
statinoma the development of cancer cachexia are outlined in ta-
C. Hypercalcemia and hypophosphatemia with osteomalacia ble 1.
induced by parathyroid hormone-like polypeptides secreted
by some tumors
D. Anemia secondary to chronic blood loss and/or bone
marrow suppression Metabolic Pathophysiology of Cancer Cachexia
9 Electrolyte and fluid derangements secondary to: [3]
A. Persistent vomiting and intestinal obstruction or intra-
cranial tumors Inability to gain and/or maintain weight despite seem-
B. Intestinal fluid losses through fistulas or diarrhea
ingly adequate nutrient intake could be due to at least
C. Intestinal secretory abnormalities with hormone-secreting
tumors such as: these obvious possibilities: (1) increased metabolic needs
a) Carcinoid syndrome generated by the accentuated nutritional demands of the
b) Zollinger-Ellison syndrome (gastrinoma) semiautonomous tumor; (2) generalized increased energy
c) Verner-Morrison syndrome (VIPoma) expenditure in the cancer patient; (3) maladaptive metab-
d) Villous adenoma
olism, i.e., failure of the cancer patient’s host tissues to
e) Increased calcitonin
D. Inappropriate antidiuretic hormone (ADH) secretion utilize the nutrients available efficiently and effectively to
associated with specific tumors such as lung carcinomas satisfy the energy requirements of the body [3]. Although
E. Hyperadrenalism secondary to tumors producing cortico- malignant cells do have their own specific nutrient re-
tropins or corticosteroids quirements and do expend energy, it has been shown that
10 Miscellaneous organ dysfunction with nutritional complica-
this fact does not fully explain the cancer cachexia-
tions such as intractable gastric ulcers with gastrinomas, Fan-
coni’s syndrome with light-chain disease, or coma with brain anorexia syndrome in human cancer patients [15]. There-
tumors fore, it is important to differentiate data reported in ani-
11 Tumor products stimulating monocyte production of various mal tumors, which tend to represent a much larger pro-
interleukins portion of the total body weight of the organism, from
data reported with human tumors which generally repre-
sent a much smaller proportion of the weight of the host.

200 Dig Dis 2003;21:198–213 Palesty/Dudrick


Many studies of resting energy expenditure (REE) and Table 2. Abnormalities of carbohydrate, protein, and fat metabo-
basal energy expenditure have been performed to estab- lism associated with cancer cachexia (adapted from Shils [14])
lish whether cancer patients are hypermetabolic [15].
Carbohydrate metabolism
These studies have demonstrated that approximately Glucose intolerance
60% of cancer patients have abnormal REE, of which Insulin resistance
approximately 35% are hypometabolic, and 25% are hy- Abnormal insulin secretion
permetabolic. A confounding aspect of these studies is Impaired glucose clearance
Increased glucose production
that lean body mass, rather than total body mass, corre-
Increased glucose turnover
lated with alterations found in measured REE. Thus, can- Increased Cori cycle activity
cer patients, who ordinarily have early diminished lean
Protein metabolism
body mass, may be expected to have energy expenditure Increased whole body protein turnover
levels which are low for their total weight, and what Increased protein fractional synthesis rates in the liver
appears to be hypometabolism, may really represent a Decreased fractional synthesis rates in muscle
eumetabolic state. Despite this non-standard interpreta- Increased hepatic protein synthesis
tion of REE data in cachectic cancer patients, it appears Recalcitrant muscle protein breakdown
Decreased plasma levels of branched-chain amino acids
that although hypermetabolism of the host and/or tumor
plays a role in cancer cachexia, hypermetabolism does not Fat metabolism
Excess body fat depletion relative to body protein loss
suffice as a complete explanation of the cancer cachexia
Increased lipolysis
syndrome. Increased free fatty acids
For convenience and understanding, maladaptive me- Increased glycerol turnover
tabolism may be separated into metabolism of carbohy- Decreased lipogenesis
drate, and fat, which are obviously intimately interrelated Hyperlipidemia
Failure of glucose to suppress oxidation of free fatty acids
[16]. The most apparent precipitating event of maladap-
Decreased serum lipoprotein lipase activity despite normal
tive metabolism in the cachectic cancer patient is decreas- insulin availability
ing blood glucose or a tendency toward hypoglycemia,
which results both from hypermetabolism of the tumor
and the host tissues, together with decreased nutrient
intake related to anorexia hypophagia or the anatomic or
physiologic changes produced by malignant cells of var- cancer patient, increased rates of glycerol and free fatty
ious origins. In normal starving human beings in whom acid turnover occur which ordinarily does not occur dur-
decreased nutrient intake is the principal factor, an imme- ing adaptation in a non-tumor-bearing, otherwise healthy
diate increase in glucose production as well as in protein but starving individual. Moreover, in healthy patients, a
synthesis and breakdown occurs as a compensatory mech- glucose infusion suppresses lipolysis, but does not sup-
anism. This self-preserving adjustment can occur because press lipolysis in cancer patients. Ultimately, the cancer
the body responds with an increase in use of fat-derived patient depletes fat stores, develops hypertriglyceridemia
calories, primarily ketone bodies for energy production and manifests decreased lipoprotein lipase levels. This
[3]. However, in cancer patients, this conservation of glu- results in failure of production of free fatty acids and
coneogenesis does not occur, but rather both protein monoacylglycerol, and subsequently, decreased adipocyte
breakdown and lipolysis occur at increasing rates in the triglyceride synthesis, which depends upon free fatty acids
process of maintaining high rates of glucose synthesis. [3].
Accordingly, the role of insulin in the cancer patient A summary of the current status of the understanding
appears to become paramount at this point as a state of of the metabolic maladaptation of the cancer patient to
relative insulin resistance develops. The insulin resistance decreased nutrient intake and increased metabolic caloric
state, in turn, is characterized by a decreased uptake and needs of both the tumor and the host include: increased
use of glucose, predominantly in skeletal muscle, and a hepatic glucose production; failure of uptake of the gluco-
tendency toward gluconeogenesis and lipolysis [3]. The neogenic glucose by muscle, resulting in proteolysis and
lipolytic component of insulin resistance is manifested by production of glycogenic amino acids; uptake and use of
excess fatty acid oxidation which does not decrease even glucose by tumor cells with resulting lactate production
with increased fat or caloric intake. Additionally, as and occasional lactic acidosis; and increased lipolysis with
decreased nutrient intake and absorption occur in the production of fatty acid and glycerol, ultimately used for

What We Have Learned about Cachexia in Dig Dis 2003;21:198–213 201


Gastrointestinal Cancer
energy and further gluconeogenesis (table 2). Thus, the this adaptive feeding response, which is so impressive in
nutritional requirements of the tumor are selectively normal human beings [23–26].
favored over those of the host by these aberrant metabolic A hormone secreted by adipose tissue, leptin, is now
mandates in the cancer patient [3]. known to be an integral component of the mechanism for
body weight recognition [19, 27–34]. Because weight loss
causes leptin levels to fall in proportion to the loss of body
Pathogenic Mechanisms of Anorexia fat, it appears that leptin plays an important role in trigger-
ing the adaptive response to starvation [2]. The activity of
The intake of energy substrates is significantly reduced the hypothalamic orexigenic signals that stimulate feeding
among cancer patients who lose weight, and although and suppress energy expenditure are increased by low lep-
anorexia is unlikely to be responsible solely for the wast- tin levels in the brain, which also decrease the activity of
ing seen in cancer patients, it may be a substantial con- anorexigenic signals that suppress appetite and increase
tributing factor [3, 16, 17]. Cancer patients, especially energy expenditure [2, 23–26]. In experimental animals
those with gastrointestinal cancer, may frequently suffer that are fasted, most of the orexigenic signals are known to
from physical obstruction of the alimentary tract, pain, be up-regulated, suggesting that these signals play an
constipation, maldigestion, malabsorption, debility, or important role in facilitating the recovery of lost weight. It
the side effects of therapy, including opiates, radiothera- appears that neuropeptide Y (NPY) produced in the hypo-
py, or chemotherapy, any of which may lead to decreased thalamus binds to a receptor (Y-5) in adjacent hypotha-
food intake [19, 20]. Additionally, hypercalcemia associ- lamic cells, which in turn increases the response of addi-
ated with cancer is a fairly common condition which can tional NPY and stimulates the neuronal basis of appetite
cause nausea, vomiting and weight loss as well as other [3]. It has further been shown that leptin also binds to Y-5,
untoward side effects. Moreover, anorexia is an extremely inhibiting NPY activity and producing satiety [35]. Re-
distressing syndrome because appetite and the ability to cently, glucagon-like factor 1 and urocortin have also been
eat have been reported to be the most important factors in shown to be appetite suppressants [3, 36, 37].
the physical and psychologic aspects (especially depres- The cancer anorexia syndrome may result from circu-
sion) of a patient’s quality of life [12, 21]. However, no lating factors produced by the tumor or by the host in
clinical cause of reduced food intake is obvious in a large response to the tumor, and several cytokines have been
number of patients with cancer and cancer cachexia. The proposed as possible mediators of the cachectic process
early satiety which accompanies reduced appetite in ano- [19]. Elevated serum levels of tumor necrosis factor-·,
rectic cancer patients has been postulated to be caused by interleukin (IL)-1, and IL-6 have been found in some, but
the production of tumor cell factors that exert their effects not all cancer patients, and the serum levels of these cyto-
by acting on hypothalamic sensory cells [38]. Possible fac- kines appear to correlate with the progression of the
tors include the satietins which have been purified from tumor [38–40]. Interferon-·, interferon-Á and leukemia-
human plasma and found to consist of two extensively inhibitory factor are additional cytokines which have
glycosylated glycoproteins. The larger molecule, satie- been postulated to play a role in the etiology of cancer
tin D, has been shown to produce a long-lasting anorectic cachexia, but which have not been proven to be responsi-
effect when injected into rats, although its role in the ble solely for the induction of cachexia [38]. However,
development of anorexia has not been established. Anoth- chronic administration of these cytokines, either alone or
er possible cause of anorexia is the increased serotonergic in combination, is capable of reducing food intake and
activity within the central nervous system of patients with reproducing the cancer anorexia-cachexia syndrome [19,
cancer cachexia, attributed to the enhanced availability of 38–43]. These cytokines may produce long-term inhibi-
tryptophan to the brain. A close relationship between ele- tion of appetite and feeding by stimulating the production
vated plasma-free tryptophan and anorexia has been ob- and release of leptin and/or by mimicking the hypotha-
served in patients with cancer and reduced food intake lamic effect of excessive negative feedback signaling from
[22]. The fact that uptake of tryptophan into the brain is leptin, leading to the normal compensatory mechanisms
competitive with uptake of branched-chain amino acids for decreases in food intake and body weight [18, 19, 23–
has suggested the use of BCAA to decrease the incidence 29, 34, 39, 44–46]. Thus, the weight loss which occurs in
of anorexia [22]. Because weight loss is such a potent stim- cancer patients obviously differs considerably from that
ulus to food intake in healthy human beings, the persis- which occurs in simple starvation in otherwise healthy
tence of anorexia in cancer patients implies a failure of human beings.

202 Dig Dis 2003;21:198–213 Palesty/Dudrick


Melanocortins, a family of regulatory peptides, which tumor has the greatest affinity and which will result in
include adrenocorticotropin (ACTH) and the melano- further increased tumor growth. Investigations are cur-
cyte-stimulating hormones, have recently been reported rently underway to determine whether this proteoglycan
as potential contributory factors in anorexia and cachexia is widely present in cachectic human cancer patients and
[2, 47–49]. This group of peptides and their receptors are whether its pathophysiologic actions mimic those of the
important in memory, behavior and immunity, but also cancer anorexia-cachexia syndrome [3]. A potentially
help to regulate appetite and body temperature [19, 31– clinically useful feature of this research is the discovery of
33]. In animal studies, the melanocortin system remained an antagonist (eicosapentaenoic acid) of this proteoglycan
active during cancer-induced cachexia, whereas the nor- which could have a role in the treatment of the human
mal response to marked loss of body weight would have anorexia-cachexia syndrome [16, 53].
resulted in down-regulation of the anorexigenic melano- It is highly likely that multiple mediators rather than a
cortin-signaling system in order to conserve energy stores. single mediator probably account for the metabolic and
Moreover, blockade of the melanocortin receptor re- pathophysiologic abnormalities associated with the an-
versed the anorexia and cachexia in the animals, suggest- orexia-cachexia syndrome. It is possible, indeed likely,
ing a pathogenetic role for this system [19, 47–49]. that tumor cells produce multiple active peptides or glyco-
The specific roles and the relative importance of the peptides; that they also stimulate the induction of addi-
various factors in the cancer anorexia-cachexia syndrome tional cytokines or lymphokines from host immune cells;
remain to be clarified. It is presumed that anorexia is and that they stimulate abnormal hormonal responses to
caused by the same mediator or mediators that also pro- the metabolic stress induced by the malignancy. It is
duce the metabolic derangements of cancer cachexia unlikely that a single factor could account for the hyper-
itself. Use of antianorectic agents alone may improve metabolism, abnormal glucose metabolism, protein glu-
quality of life in the cancer patient, but may not solve the coneogenesis and insulin resistance, in addition to the
problem of anorexia and its associated morbidity and anorexia, anemia and fever that accompany the cancer
mortality in cancer patients [16]. anorexia-cachexia syndrome.
The role of hormones in the cancer anorexia-cachexia
syndrome must be considered because of the obvious role
that hormones play in the intermediary metabolism of Dysfunctional Gastrointestinal Manifestations
carbohydrates [50]. Insulin, epinephrine, ACTH, human
growth hormone, and insulin-like growth factor have all As a result of the malignant disease and/or its treat-
been suggested to have a role in the anorexia-cachexia ment, abnormalities can occur in the mouth or elsewhere
syndrome. During early starvation, decreased insulin lev- in the gastrointestinal tract and may interfere with inges-
els and increased glucagon and epinephrine result in acti- tion of food [19]. Aberrations of taste and smell have also
vation of cyclic adenosine monophosphate and of a pro- been documented in cancer patients [54, 55]. Altered
tein kinase that activates hormone-sensitive lipase. Fail- capacity to recognize and taste sweetness in foods occurs
ure of this normal mechanism may account for the weight in more than one-third of patients, whereas bitterness,
loss in cancer patients who have increased rates of glycer- sourness, and saltiness are less frequently encountered in
ol and free fatty acid turnover compared with starved cancer patients [56, 57]. Decreased threshold for recogni-
healthy patients [16]. tion of bitter taste correlates well with meat aversion, and
Recent studies have indicated the presence of an acidic other aversions to specific foods may be learned or devel-
peptide in animal tumors which appears to have lipolytic oped secondary to unpleasant experiences that coincide
properties that could lead to some of the characteristics of with exposure to a particular food [54]. This usually
the cancer cachexia syndrome [16, 51]. This factor ap- occurs in cancer patients in association with chemothera-
pears to be a proteoglycan which has been found in the py [58]. It appears that these changes in taste and smell
urine of murine species with tumors as well as in human correlate with decreased nutrient intake, poor response to
patients with cancer anorexia-cachexia syndrome involv- therapy, and tumor progression including metastasis [57].
ing weight loss of 11.4 kg [16, 52]. In animals, this proteo- The roles of other factors in the etiology of cachexia
glycan has been shown to mobilize free fatty acids from require clarification, including zinc deficiency, alterations
adipose tissue, and amino acids from muscle, which in brain neurotransmitters, and opioid peptides that af-
prompted the investigators to suggest that the role of these fect taste and nutrient selection [2, 39, 54, 58]. Direct
tumor products is to mobilize nutrients for which the involvement in the gastrointestinal tract or its accessory

What We Have Learned about Cachexia in Dig Dis 2003;21:198–213 203


Gastrointestinal Cancer
Table 3. Nutritional problems secondary to gastrointestinal cancer therapy (adapted from Shils [14])

I. Radiation therapy III. Pharmacologic therapy


A. Radiation of the oropharynx A. Corticosteroids
1. Impairment or destruction of taste; xerostomia and 1. Fluid and electrolyte imbalance
odynophagia; loss of teeth 2. Nitrogen and calcium losses
B. Radiation of lower neck and mediastinum 3. Hyperglycemia
1. Esophagitis with dysphagia B. Sex hormone analogue
2. Fibrosis with esophageal stricture 1. Fluid retention
C. Radiation of abdomen and pelvis 2. Nausea
1. Acute and chronic intestinal damage with diarrhea, 3. Glucocorticoid effects secondary to megestrol acetate
malabsorption, stenosis, obstruction, and fistula formation C. Immunotherapy
1. Tumor necrosis factor
II. Surgery therapy
a) Fluid retention
A. Radical resection of oropharyngeal structures
b) Hypotension
1. Chewing and swallowing difficulties
c) Nausea and vomiting
B. Esophagectomy
d) Diarrhea
1. Gastric stasis and hypochlorhydria secondary to vagotomy
2. Interleukin-2
2. Steatorrhea secondary to vagotomy
a) Hypotension
3. Diarrhea secondary to vagotomy
b) Fluid retention
4. Early satiety
c) Azotemia
5. Reflux and/or regurgitation
3. Interferons
C. Gastrectomy (total or near total)
a) Anorexia
1. Dumping syndrome
b) Nausea and vomiting
2. Malabsorption
c) Diarrhea
3. Achlorhydria and lack of intrinsic factor
d) Azotemia
4. Hypoglycemia
D. Cytotoxic chemotherapy
5. Early satiety
1. Nausea and vomiting
D. Intestinal resection
2. Mucositis and stomatitis
1. Jejunum
3. Diarrhea
a) Decreased efficiency of absorption of many nutrients
4. Abdominal pain or cramping
2. Ileum
5. Hepatotoxicity
a) Vitamin B12 deficiency
6. Gastrointestinal bleeding with anemia
b) Bile salt malabsorption with diarrhea or steatorrhea
7. Anorexia
c) Hyperoxaluria and renal stone formation
8. Weight loss
d) Calcium and magnesium depletion
9. Inhibition of protein synthesis
e) Fat and fat-soluble vitamin malabsorption
10. Inhibition of purine and pyrimidine synthesis
3. Massive bowel resection
11. Inhibition of DNA synthesis, replication and transcription
a) Life-threatening malabsorption (short bowel
12. Inhibition of various metabolic processes
syndrome)
b) Malnutrition
c) Metabolic acidosis
d) Dehydration
4. Ileostomy and/or colostomy
a) Complications of salt and water balance and
homeostasis digestive organs by tumors can cause problems with diges-
E. Blind loop syndrome tion and nutrient absorption, and subsequently lead to
1. Vitamin B12 malabsorption malnutrition and cachexia [2]. Odynophagia and/or dys-
F. Pancreatectomy
1. Malabsorption
phagia occur most commonly in patients with cancers of
2. Diabetes mellitus the head and neck and esophagus, and tumors elsewhere
in the gastrointestinal tract and the hepatobiliary tract are
often complicated by partial or total obstruction of the
digestive tract, causing nausea and/or vomiting [55]. Met-
astatic cancers from other systems to the gastrointestinal
tract can also cause, or contribute to, similar problems by
extrinsic pressure on the gastrointestinal tract structures.
Although satiety signals from the gastrointestinal tract
help to regulate appetite and food intake, early satiety is a

204 Dig Dis 2003;21:198–213 Palesty/Dudrick


characteristic of cachectic cancer patients, even without operative intravenous feeding should be limited to pa-
direct involvement of the gastrointestinal tract, and may tients unable to sustain lean body mass by oral or enteral
be associated with increased activity of proinflammatory feeding [69]. The authors stated further that intravenous
cytokines including IL-1ß, and centrocorticotropic-releas- feeding had not been documented to affect responses ei-
ing factor (CRF), which is a potent anorexigenic factor ther to therapy or survival favorably in patients receiving
[19, 60, 61]. Changes in gastrointestinal motility observed radiation therapy or chemotherapy. However, they also
during stress suggest that CRF may be involved as the affirmed that selecting patients for support with TPN
triggering signal [2]. CRF may also induce the delayed remains a matter of clinical judgment, and that when
gastric emptying and gastric stasis observed not only in therapy response rates and nutritional morbidity are high,
cancer patients, but in non-neoplastic conditions such as intravenous feeding should be instituted until the host can
infection and anorexia nervosa [2, 54, 62, 63]. These recover from the effects of antitumor therapy [69]. The
abnormal gastrointestinal effects may result in early satie- authors concluded additionally that there is clear evi-
ty and may have a negative influence on food intake. dence that in individual patients with cancer, intravenous
Another major cause of malnutrition leading to an- feeding can prevent death from starvation and decrease
orexia-cachexia syndrome occurs concomitantly with an- the morbidity of treatment, but only those patients un-
ticancer therapies [54, 56]. Nausea, vomiting, abdominal dergoing surgery for gastrointestinal tract neoplasia have
cramping, bloating, mucositis and paralytic ileus can all benefited significantly from the addition of adjuvant
accompany chemotherapy. Fluorouracil, adriamycin, intravenous feeding [69]. Another subsequent review con-
methotrexate and cisplatin are among the many antineo- cluded that chronically malnourished patients given nu-
plastic agents that may induce severe gastrointestinal tritional support often have restoration of a sense of well-
complications [64]. Because enterocytes are rapidly divid- being and become more physically functional; however,
ing cells, they are more susceptible to the cytotoxic effects whether this support results in long-term clinical benefits
of both chemotherapy and radiotherapy than most other is difficult to evaluate [70]. The authors stated also that it
cells in the body. Erosive lesions that can occur at various appears that routine application of nutritional support to
levels of the digestive tract, resulting in the impairment of all patients undergoing treatment for malignancy is not
food intake, digestion and nutrient absorption, can be justified, and that the primary indication for nutritional
caused by both chemotherapy and radiotherapy [2]. Con- support is for the malnourished patient undergoing a
sequences of cancer treatment predisposing to nutritional major operation for upper gastrointestinal malignancy,
problems are listed in table 3. and for the chemotherapy patient with severe gastrointes-
tinal dysfunction [70]. In another analysis of 18 trials in
children with cancer, the author acknowledged the clear
Nutritional Support with Cancer Cachexia benefits of nutritional support in patients undergoing a
bone marrow transplantation; however, there appeared to
Early studies promoted optimism that judicious enter- be little support for the routine aggressive nutritional sup-
al or parenteral nutritional support might overcome can- port in the non-surgical oncology patient [71]. Nonethe-
cer anorexia, and modulate or obviate malnutrition and less, the author concluded further that circumstances exist
cancer cachexia [19, 65–67]. However, the failure of in which aggressive nutritional support by any and all
aggressive nutritional support to increase lean body mass, routes should be provided, especially during prolonged
especially skeletal muscle mass, in the patient with cancer inability to eat, when nutrition is secondary to poor
cachexia has been disappointing. In a meta-analysis of 28 intake, when a nutrition support team is available to
randomized studies of cancer patients receiving total par- decrease related complications, and when the tumor
enteral nutrition (TPN), the use of TPN preoperatively in present is deemed likely to respond to antineoplastic
patients with gastrointestinal cancer helped to reduce treatment [71]. Additionally, parenteral nutrition may
major surgical complications and operative mortality sig- facilitate administration of complete chemoradiation
nificantly, but no significant benefit was shown on surviv- therapy dosages in esophageal cancer patients and may
al, tolerance of treatment, toxicity, or tumor response in have beneficial effects for certain patients with decreased
patients receiving chemotherapy and TPN [68]. A survey food intake because of mechanical obstruction of the gas-
analyzing results individually for patients receiving radia- trointestinal tract [72–74]. Home parenteral nutrition can
tion therapy and chemotherapy and for patients undergo- also be efficacious and rewarding for such patients [2]. If
ing major surgery, concluded that the routine use of pre- the gastrointestinal tract can be used for nutritional sup-

What We Have Learned about Cachexia in Dig Dis 2003;21:198–213 205


Gastrointestinal Cancer
port, enteral nutrition has the advantage of maintaining The best treatment for cancer cachexia, especially that
the integrity of the enterocytes, the mucosal barrier, and related to cancer of the gastrointestinal tract, is to cure the
immunologic function, as well as the advantage of having cancer. Unfortunately, this lofty goal remains an infre-
lower cost and fewer adverse side effects [2, 54, 74]. quent achievement currently among patients with solid
Some surveys of the effectiveness of ineffectiveness of tumors. Since a cure for cancer is still under investigation
nutritional support in cancer patients have emphasized and forthcoming, the best palliative or therapeutic meta-
survival as an important criterion [14]. On this matter, bolic options available are to stimulate increased nutri-
several points merit consideration. First, most surveys did tional intake and ameliorate or prevent the catabolism of
not include survival data, thus leaving only a relatively muscle and fat. This can be accomplished to varying
small number of patients in this category. Second, if degrees with a multitude of pharmacologic agents. How-
malignancies such as metastatic esophageal, stomach, co- ever, a goal as monumental as this would be much easier
lon and lung cancers do not respond well to any known to achieve if it were possible to identify the exact causes
antitumor treatments, why should nutritional support be and mechanisms of the cancer anorexia-cachexia syn-
expected to help such patients beyond delaying death and drome. Currently, only those elements of decreased food
perhaps improving the quality of life in some patients in intake directly related to the antineoplastic treatment,
whom starvation existed to varying degrees? Finally, the such as nausea, vomiting, mucositis and dysfunctional
clinician or investigator interested in the subject of effica- gastrointestinal motility, can be somewhat controlled or
cy of nutritional support in cancer cachexia should review modulated clinically. The most realistic and practical
the published meta-analysis and also the original trial options at this time are directed toward minimizing
reports together with other reviews that provide evidence adverse gastrointestinal side effects or complications and
of the value of many cases of improved nutrition for increasing appetite and nutritional intake in an effort to
appropriate patients with cancer cachexia [14, 75, 76]. improve quality of life. Several types of drugs which have
There is much more to be learned about the metabolism been commonly employed in attempts to improve appe-
of cancer patients with progressive disease, and such tite, food intake, sense of well-being and body weight gain,
knowledge will likely lead to improvements in nutritional include progestational drugs, corticosteroids and antisero-
support as more effective antineoplastic therapies are also tonergic drugs. Additionally, a newer group of drugs is
developed [14]. under investigation primarily because of their effects on
The effects of aggressive nutritional support on tumor tumor necrosis factor-·, or their effects on muscle metab-
growth and development have been difficult to delineate olism. These and other agents now present the possibility
in cancer patients and are still being debated [2, 77]. How- of confronting this recalcitrant syndrome from a different
ever, a clear benefit of nutritional support may be derived approach and an opportunity for combined therapeutic
in cancer patients with severe malnutrition who may endeavors to improve the quality of life of these patients
require surgery or may have an obstructing, but potential- [80].
ly responsive tumor to therapy [2, 55, 73, 78]. Experience
has also indicated that the cancer patient with a severely
dysfunctional alimentary tract resulting from radiation, Progestational Agents
chemotherapy, surgery or combination thereof, but who is
free of residual malignant disease, is entitled to proper In many patients who were originally receiving proges-
management by oral, enteral, and/or parenteral feeding tational drugs as part of a multimodal antineoplastic
support which can lead to a prolonged life of good quality treatment regimen, notable increases in appetite and sig-
[14]. Finally, controversy remains regarding the influence nificant body weight gain occurred. These empirically
of aggressive nutritional support on the quality of life of derived findings prompted controlled trials to study these
patients with advanced cancer, and it is very important to agents specifically regarding their ability to increase appe-
consider the risks, benefits and ethical aspects involved tite in patients with cancer anorexia-cachexia and perhaps
before beginning such a regimen. Physician attitudes, promote weight gain as well. Megestrol acetate (Megace)
patient age and prognosis, and family or patient’s percep- has been the most widely used and studied drug in this
tions often play important roles in the decision to admin- category, and its effects on increasing appetite, caloric
ister nutritional support, and these variables will continue intake and weight gain has been shown to be dose related,
as issues during the patient’s course [79]. in doses ranging from 160 to 1,600 mg, with an optimal
dosage of about 800 mg/day [80, 81]. Generally, patients

206 Dig Dis 2003;21:198–213 Palesty/Dudrick


are started on the lowest effective dosage (160 mg/day), shown to increase the sense of well-being and to alleviate
and the dose is increased according to the clinical re- anorexia and asthenia in cancer patients; however, their
sponse of the patient to the drug [80, 82]. effects diminish appreciably after 3–4 weeks. Although
Studies using medroxyprogesterone acetate have also several randomized, controlled clinical studies have dem-
been associated with a significant increase in appetite onstrated the symptomatic benefits of some of the differ-
together with increases in serum thyroid binding prealbu- ent corticosteroids, most reports have indicated a limited
min (transthyretin) and retinal binding protein without beneficial effect on appetite, food intake, sense of well-
any concomitant changes in body weight, functional per- being and performance for up to 4 weeks [2, 82, 98–103].
formance status, or anthropometric measurements [80, Corticosteroids also have a significant antinausea effect
83]. On the other hand, a controlled multicenter study and can improve asthenia and pain control in some
comparing medroxyprogesterone with a placebo demon- patients; however, no lasting positive effect on body
strated significantly approved appetite and body weight weight has been observed. Moreover, prolonged cortico-
(but not quality of life) in the experimental group versus steroid administration has a negative effect on protein
the control group in advanced-stage, non-hormone sensi- synthesis, thus neutralizing potential benefits of increased
tive cancer [80, 84]. appetite. Additionally, prolonged corticosteroid treat-
A combination of megestrol and prednisone was ment may result in weakness, delirium, osteoporosis and
shown to improve appetite significantly better, compared immunosuppression, compounding these adverse side ef-
with those patients given a placebo while receiving radia- fects, which are frequently already present in patients
tion therapy [85]. Moreover, megestrol was significantly with advanced cancer [2, 104]. Prednisolone and dexa-
better as an appetite stimulant than prednisone or fluoxy- methasone have been shown to improve appetite to a
mesterone [80]. The data comparing megestrol with dexa- greater extent than a placebo, and methylprednisolone
methasone as appetite stimulant remain to be clarified may improve quality of life; however, no compelling evi-
[80, 86]. dence exists to indicate the superiority of any one gluco-
The possible mechanisms of action of progestational corticoid in stimulating appetite [2, 20, 78, 100].
drugs is unclear, but may be related to their glucocorticoid Although the exact mechanism of action of these drugs
activity. Currently suggested explanations for their effec- on appetite is unclear, some studies indicate that the corti-
tiveness include stimulation of NPY, modulation of cal- costeroids inhibit prostaglandin metabolism and inhibit
cium channels in the ventromedial hypothalamus, and the synthesis or release of the cytokines IL-1 and TNF-·,
hindrance of the activity of cytokines, including TNF, which, in turn, theoretically decrease appetite through
IL-1 and IL-6 [80, 87–89]. their action on NPY, leptin, CRF and serotonin [2, 20, 78,
Concerns with the use of either megestrol or medroxy- 100]. At this time, no definitive evidence exists to support
progesterone are centered on the associated side effects an explanation for the exact mechanism of action of corti-
including induction of thromboses and thromboemboli, costeroids in stimulating appetite in cancer anorexia-
breakthrough uterine bleeding, hypertension, hypergly- cachexia patients.
cemia, peripheral edema, alopecia, Cushing syndrome,
adrenal suppression and adrenal insufficiency [80, 90–
93]. However, these adverse effects have been encoun- Anabolic Steroids
tered only rarely in most clinical trials [80, 94–97]. On the
other hand, caution must be taken in administering these Although anabolic steroids have been shown to be
drugs to patients with known thromboembolic disease, effective among athletes and other healthy people in gain-
heart disease or high risk for fluid retention [2, 78]. ing muscle mass, they have not been shown to achieve this
benefit in gastrointestinal cancer patients [80, 106]. On
the other hand, nandrolone has been shown to increase
Glucocorticoids weight gain in a group of lung cancer patients by increas-
ing protein synthesis via the androgen receptor. However,
Although appetite stimulants, including corticoste- this is the only study that has demonstrated any useful-
roids, have been of some value in patients with low ness of anabolic steroids in the treatment of cancer
nutrient intakes, they have not proven to be as useful in patients [2, 107].
patients with major metabolic abnormalities or cancer
anorexia-cachexia syndrome. Corticosteroids have been

What We Have Learned about Cachexia in Dig Dis 2003;21:198–213 207


Gastrointestinal Cancer
Antiserotonergic Drugs 3-month study [115]. In other studies in cachectic cancer
patients, the inclusion of eicosapentaenoic acid in the diet
Cyproheptadine is an antiserotonergic drug with anti- significantly increased weight gain, lean body mass, per-
histamine actions which have been useful in the symp- formance status and survival, the latter unexpected result
tomatic relief of rhinitis, conjunctivitis and pruritis asso- occurring in both the weight-losing and non-weight-losing
ciated with allergic reactions, which has also been useful groups of patients [116, 117].
as an appetite stimulant in anorexia nervosa [80]. Al- Gastroparesis and gastrointestinal dyskinesia are rec-
though cyproheptadine did not prevent progressive ognized complications of cancer cachexia which can cause
weight loss in patients with advanced cancer, it was suc- anorexia, nausea, early satiety and constipation, leading
cessful as a mild appetite stimulant in a randomized con- to impaired food intake [2, 118]. Because many patients
trolled study [2, 82, 103, 108]. Because evidence suggests with advanced cancer have delayed gastric emptying and
that anorexia may be mediated by increased serotonergic gastroparesis, prokinetic agents such as metoclopramide
activity in the brain, its blockade by cyproheptadine have been used in cancer patients to prevent or ameliorate
might mediate an antianorexia effect [2, 109, 110]. How- the emesis associated with chemotherapy and may alle-
ever, its primary side effect is sedation, which might limit viate anorexia and early satiety with minimal side effects
its usefulness in advanced cancer patients [80, 108]. [2, 54, 82, 119]. Other prokinetic agents such as domperi-
Granisetron and odansetron, which are often used as done and erythromycin require additional controlled ran-
antiemetics during chemotherapy, are additional seroto- domized studies to define their roles in treatment of can-
nin receptor inhibitors that have been used in the cancer cer cachexia patients [2, 63, 104].
anorexia-cachexia patient as antagonists to the satiating
effect of serotonin [2, 111]. Although these agents have
failed to promote weight gain in trials to date, they have Other Potentially Useful Drugs
been noted to improve the patients’ ability to enjoy food
[2, 112]. Additional investigations in cancer anorexia- Because hydrazine sulfate inhibits phosphoenol-pyru-
cachexia patients are required to define the role of these vate carboxykinase, an integral enzyme in gluconeogene-
and other antiserotonergic mediators in stimulating appe- sis, it was hypothesized that by interrupting the Cori cycle
tite. and normalizing carbohydrate metabolism, this agent
might be useful in improving appetite, caloric intake and
nutritional status in cachectic cancer patients [2, 20].
Metabolic and Physiologic Modulators However, several large studies showed that this drug has
no significant impact on weight gain or any other benefit
Branched-chain amino acids may modulate the pe- in patients with advanced colorectal cancer [120–122].
ripheral muscle proteolysis which occurs in cancer ca- Increased survival times, improved appetite and in-
chexia by providing protein-sparing substrate for both creases in caloric intake and serum albumin levels have
muscle metabolism and gluconeogenesis [2, 104]. When been reported with the use of hydrazine sulfate in some
used in TPN solutions, the branched-chain amino acids studies, but these favorable effects have not been con-
have been shown to improve protein accretion and albu- firmed by others [79]. Because of the lack of definitive
min synthesis while improving nitrogen balance [2, 113]. studies showing consistent efficacy, together with the sig-
Moreover, oral supplementation of branched-chain ami- nificant risk of neurotoxicity associated with its use, this
no acids has been used successfully to decrease the severi- drug has not gained popularity with oncologists [2, 80].
ty of anorexia in cancer patients, theoretically, by compet- Thalidomide has been shown to decrease the activity
ing with tryptophan, the precursor of brain serotonin, of TNF-· through cytokine modulation in cancer pa-
thus blocking increased hypothalamic activity of seroto- tients, in addition to improving insomnia, restlessness
nin [2, 110, 111]. and nausea in advanced cancer patients, while improving
Following studies in animals, which demonstrated that appetite and sense of well-being in the majority of patients
eicosapentaenoic acid inhibited lipolysis and muscle pro- studied [2, 80, 123]. Side effects of thalidomide include
tein degradation in a cachexia model, patients with pan- fever, skin rash and peripheral neuropathy in addition to
creatic cancer who received supplements of fish oil ex- its well-known teratogenic effects [80, 124, 125].
hibited decreased fatigue and a slight body weight gain Melatonin, a hormone produced by the pineal body,
as well as a reduction of acute-phase proteins during a has promoted increased weight gain and increased appe-

208 Dig Dis 2003;21:198–213 Palesty/Dudrick


tite in patients with advanced cancer in association with Non-steroidal anti-inflammatory drugs (NSAIDS) re-
its ability to decrease levels of circulating TNF-· [2, 80, duce circulating levels of IL-6, acute phase proteins and
126]. A study of 100 patients with metastatic carcinoma cortisol, which renders them useful in treating fever and
showed that weight loss of more than 10% was a much less pain in patients with advanced cancer [2]. In a series of
common finding among patients receiving melatonin colorectal cancer patients, ibuprofen was shown to nor-
than among the patients receiving a placebo [2, 80, 126]. malize protein kinetics in cachectic patients and stabilize
Pentoxifylline is a methylxanthine-derived phospho- weight, while reducing REE and increasing quality of life
diesterase inhibitor which has been shown to decrease in patients with pancreatic cancer [2, 143–146]. The main
TNF-· synthesis in cancer patients by decreasing gene limitation of this class of drugs is their adverse effect on
transcription [80, 127, 128]. However, its use in a con- the gastrointestinal tract; however, recent studies into the
trolled trial of cancer patients with solid tumors elicited efficacy of COX-2 inhibitors such as celecoxib and rofe-
no increase in appetite or body weight gain compared coxib, which appear to have fewer and less severe gas-
with the controls [80, 129]. trointestinal side effects, have indicated promising early
5-Deoxy-5-fluorouridine, a fluorinated pyrimidine nu- results in their use as alternatives to the older NSAIDS.
cleoside, is a cytostatic agent which has been shown to
decrease the progression of cachexia in an animal model
by inhibiting IL-6 and proteolysis-inducing factor [2, 130, Commentary
131]. Although this agent decreased the progression of
tumor growth, it did not demonstrate preservation of In this review of cachexia associated with gastrointesti-
body weight even though it ameliorated progressive nal cancer, we have attempted to summarize the more
weight loss and hyperglycemia while increasing the pro- common metabolic abnormalities in the cachectic cancer
duction acute phase proteins [2, 130]. patient, the theories proposed as explanations of these
Cannabinoids are well-known appetite stimulants phenomena and the nutritional and pharmacologic inter-
which can promote weight gain. Marijuana and its deriva- ventions currently in use for amelioration of the severe
tives, dronabinol, marinol and nabilone have shown their host tissue depletion caused by this wasting syndrome.
effectiveness as antiemetics in cancer patients especially The metabolic pathophysiology of cancer cachexia has
those receiving chemotherapy [2, 78, 102–104]. A num- been reviewed, discussed and summarized, and the cur-
ber of studies have demonstrated improved appetite after rent status of the understanding of the metabolic malad-
administration of cannabinoids; however, minimal im- aptation of the cancer patient to decreased nutrient intake
provement in overall body weight was achieved [2, 132, and increased metabolic needs of both the tumor and the
133]. Limitations of these agents as potential appetite host has been outlined. The nutritional problems associat-
stimulants include their adverse effects on the central ner- ed with the development of cancer cachexia have also
vous system such as somnolence, confusion and percep- been presented comprehensively with particular empha-
tual disturbances which may be augmented further in sis on the side effects and complications indigenous to
advanced cancer patients who often already exhibit some cancers of the gastrointestinal tract. Additionally, the
cognitive impairment [2, 80, 134, 135]. abnormalities of carbohydrate, protein and fat metabo-
ß2-Adrenergic receptor agonists have been shown to lism commonly associated with cancer cachexia have
prevent muscle protein wasting in tumor-bearing rats and been outlined and discussed from historical, current and
to increase muscle mass and function significantly in non- future perspectives. Moreover, the nutritional problems
tumor-bearing rats [80, 136–140]. Even without changing secondary to individual forms of antineoplastic therapy in
food intake or rate of tumor growth, a recent study dem- specific regions of the gastrointestinal tract have been
onstrated that salbutamol, salmeterol and clenbuterol had delineated extensively. The established and experimental
a positive effect on muscle mass in tumor-bearing rats nutritional and pharmacologic support options in the
[141]. Although studies in human beings are limited, this management of patients with cancer cachexia and ad-
class of drugs has been shown to improve muscle strength vanced gastrointestinal cancer have been described brief-
significantly following orthopedic surgery, even without ly in an attempt to update their status as therapeutic
the need for exercise. However, to date, no controlled adjuncts.
studies have been reported using these agents in cancer The perplexing problem of cancer cachexia and an-
patients [2, 142]. orexia is compounded in patients with malignancies of
the gastrointestinal tract because not only are the usual

What We Have Learned about Cachexia in Dig Dis 2003;21:198–213 209


Gastrointestinal Cancer
cancer-associated mediators of cachexia and anorexia ac- Attempts to overcome this grave physical and func-
tive in inducing weight loss, wasting and deterioration of tional handicap initially included parenteral nutritional
functional status, but, the normal means by which nu- supplementation and total parenteral nutrition; however,
trient intake is attained by the patient is compromised by lack of uniform success in achieving optimal nutritional
direct involvement of the gastrointestinal tract anywhere status, improvement of function, and increase in survival
from the mouth to the anus by tumor, thus interfering has been disappointing and challenging. Currently, the
with the normal mechanisms favoring appetite, gustatory most promising therapeutic possibilities are dependent
satisfaction, food intake, digestion, nutrient absorption upon the continuing development of combinations of
and substrate assimilation. This ‘double jeopardy’ does antineoplastic therapies, unique nutrient formulations,
not ordinarily occur with malignancies of other organ sys- and pharmacologic agents designed to ameliorate the met-
tems unless or until metastases compromise the gastroin- abolic derangements induced by the tumor and its prod-
testinal tract and its appendages secondarily. Thus, the ucts. Obviously, the ultimate answer to this vexing situa-
increased metabolic expenditure and requirements im- tion is to assault the biology of the neoplastic process at its
posed by the neoplastic process are less likely to be met by foundation and develop a cure for cancer.
the gastrointestinal tract alone. Furthermore, curative
and palliative therapies, including surgery, chemothera-
py, radiotherapy and immunotherapy alone or in combi- Acknowledgment
nation, impose an additional burden on an already crip-
The authors greatly appreciate the many significant contributions
pled system. of Joan Reeser in the preparation of the manuscript.

References

1 Toomey D, Redmond P, Bouchier-Hayes D: 10 Kotler DP: Cachexia. Ann Intern Med 2000; 21 Padilla GV: Psychological aspects of nutrition
Mechanisms mediating cancer cachexia. Can- 133:622–634. and cancer. Surg Clin North Am 1986;66:
cer 1995;76:2418–2426. 11 Rosenbaum K, Wang J, Peirson RN Jr et al: 1121–1135.
2 Inui A: Cancer anorexia-cachexia syndrome. Time-dependent variation in weight and body 22 Cangiano C, Laviano A, Meguid MM, Mulieri
CA Cancer J Clin 2002;52:72–91. composition in healthy adults. J Parenter En- M, Conversano L, Preziosa I, Rossi-Fanelli F:
3 Puccio M, Nathanson: The cancer cachexia teral Nutr 2000;24:52–55. Effects of administration of oral branched-
syndrome. Semin Oncol 1997;24:277–287. 12 Brennan M: Uncomplicated starvation versus chain amino acids on anorexia and caloric
4 DeWys W: Pathophysiology of cancer cachex- cancer cachexia. Cancer Res 1977;37:2359– intake in cancer patients. J Natl Cancer Inst
ia: Current understanding and areas of future 2364. 1996;88:550–552.
research. Cancer Res 1982;42(suppl):721–726. 13 Shamberger RJ: Nutrition and Cancer. New 23 Flier JS: Clinical review 94: What’s in a name?
5 Warren S: The immediate causes of death and York, Plenum Press, 1984, pp 353–366. In search of leptin’s physiologic role. J Clin
cancer. Am J Med Sci 1932;184:610–615. 14 Shils ME: Nutrition and Diet in Cancer Man- Endocrinol Metab 1998;83:1407–1413.
6 Harrison LE, Fong Y: Enteral nutrition in the agement; in Shils ME, Olson JA, Shike M (eds): 24 Schwartz MW, Dallman MF, Woods SC: Hy-
cancer patient; in Rombeau JL, Rolandelli RH Modern Nutrition in Health and Disease, ed 8. pothalamic response to starvation: Implica-
(eds): Clinical Nutrition Enteral and Tube Philadelphia, Lea & Febiger, 1994, pp 1317– tions for the study of wasting disorders. Am J
Feeding, ed 3. Philadelphia, Saunders, 1997, 1348. Physiol 1995;269:949–957.
pp 300–323. 15 Heber D, Tchekmedyian NS: Mechanisms of 25 Schwartz MW, Seeley RJ: Seminars in medi-
7 Dewys WD, Begg C, Lavin PT, Band PR, Ben- cancer cachexia. Contemp Oncol 1995, special cine of the Beth Israel Deaconess Medical Cen-
nett JM, Bertino JR, Cohen MH, Douglass HO issue, pp 6–10. ter. Neuroendocrine responses to starvation
Jr, Engstrom PF, Ezdinli EZ, Horton J, John- 16 Langstein HN, Norton JA: Mechanisms of can- and weight loss. N Engl J Med 1997;336:1802–
son GJ, Moertel CG, Oken MM, Perlia C, cer cachexia. Hematol Oncol Clin North Am 1811.
Rosenbaum C, Silverstein MN, Skeel RT, 1991;5:103–123. 26 Inui A: Feeding and body-weight regulation by
Sponzo RW, Tormey DC: Prognostic effect of 17 Staal-van den Brekel AJ, Schols AM, ten Velde hypothalamic neuropeptides – Mediation of
weight loss prior to chemotherapy in cancer GP, Buurman WA, Wouters EF: Analysis of the actions of leptin. Trends Neurosci 1999;22:
patients. Eastern Cooperative Oncology the energy balance in lung cancer patients. 62–67.
Group. Am J Med 1980;69:491–497. Cancer Res 1994;54:6430–6433. 27 Friedman JM, Halaas JL: Leptin and the regu-
8 Bozzetti F, Migliavacca S, Scotti A, Bonalumi 18 Wigmore SJ, Plester CE, Ross JA, Fearon KC: lation of body weight in mammals. Nature
MG, Scarpa D, Baticci F, Ammatuna M, Pupa Contribution of anorexia and hypermetabo- 1998;395:763–770.
A, Terno G, Sequeira C, Masserini C, Ema- lism to weight loss in anicteric patients with 28 Elmquist JK, Maratos-Flier E, Saper CB, Flier
nuelli H: Impact of cancer, type, site, stage and pancreatic cancer. Br J Surg 1997;84:196–197. JS: Unraveling the central nervous system
treatment on the nutritional status of patients. 19 Bruera E: ABC of palliative care. Anorexia, pathways underlying responses to leptin. Nat
Ann Surg 1982;196:170–179. cachexia and nutrition. BMJ 1997;315:1219– Neurosci 1998;1:445–450.
9 Meguid MM, Meguid V: Preoperative identifi- 1222. 29 Woods SC, Seeley RJ, Porte D Jr, Schwartz
cation of the surgical cancer patient in need of 20 Barber MD, Ross JA, Fearon KC: Cancer ca- MW: Signals that regulate food intake and
postoperative supportive total parenteral nutri- chexia. Surg Oncol 1999;8:133–141. energy homeostasis. Science 1998;280:1378–
tion. Cancer 1985;55(suppl):258–262. 1383.

210 Dig Dis 2003;21:198–213 Palesty/Dudrick


30 Bray GA, York DA: The Mona Lisa hypothesis 48 Wisse BE, Frayo RS, Schwartz MW, Cum- 67 Dudrick SJ, MacFadyen BV Jr, Souchon EA,
in the time of leptin. Recent Prog Horm Res mings DE: Reversal of cancer anorexia by Englert DM, Copeland EM: Parenteral nutri-
1998;53:95–117. blockade of central melanocortin receptors in tion techniques in cancer patients. Cancer Res
31 Kalra SP, Dube MG, Pu S, Xu B, Horvath TL, rats. Endocrinology 2001;142:3292–3301. 1977;37:2440–2450
Kalra PS: Interacting appetite-regulating path- 49 Lechan RM, Tatro JB: Hypothalamic melano- 68 Klein SM, Simes J, Blackburn GL: Total paren-
ways in the hypothalamic regulations of body cortin signaling in cachexia. Endocrinology teral nutrition and cancer clinical trials. Cancer
weight. Endocr Rev 1999;20:68–100. 2001;142:3288–3291. 1986;58:1378–1386.
32 Schwartz MW, Woods SC, Porte D Jr, Seeley 50 Bartlett DL, Charland SL, Torosian MH: Re- 69 Lowry SF, Brennan MF: Intravenous feeding
RJ, Baskin DG: Central nervous system con- versal of tumor associated hyperglucagonemia of the cancer patient; in Rombeau JL, Caldwell
trol of food intake. Nature 2000;404:661–671. as treatment for cancer cachexia. Surgery 1995; MD (eds): Parenteral Nutrition. Philadelphia,
33 Inui A: Transgenic approach to the study of 118:87–91. Saunders, 1986.
body weight regulation. Pharmacol Rev 2000; 51 Beck SA, Mulligan HD, Tisdale MJ: Lipolytic 70 Shike M, Brennan MF: Supportive care of the
52:35–61. factors associated with murine and human can- cancer patient; in DeVita VT, Hellman S, Ro-
34 Inui A: Transgenic study of energy homeostasis cer cachexia. J Natl Cancer Inst 1990;82:1922– senberg SA (eds): Principles and Practice of
equation: Implications and confounding in- 1926. Oncology, ed 3. Philadelphia, Lippincott, 1989.
fluences. FASEB J 2000;14:2158–2170. 52 Todorov P, Cariuk P, McDevitt T, Coles B, 71 Lipman TO: Clinical trials of nutritional sup-
35 Gerald C, Walker MW, Criscione L, Gustafson Fearon K, Tisdale M: Characterization of a port in cancer. Parenteral and enteral therapy.
EL, Batzl-Hartmann C, Smith KE, Vaysse P, cancer cachectic factor. Nature 1996;379:739– Hematol Oncol Clin North Am 1991;5:91–
Durkin MM, Laz TM, Linemeyer DL, Schaff- 742. 102.
hauser AO, Whitebread S, Hofbauer KG, Ta- 53 Beck SA, Smith KL, Tisdale MJ: Anti-cachec- 72 Sikora SS, Ribeiro U, Kane JM 3rd, Landre-
ber RI, Branchek TA, Weinshank RL: A recep- tic and anti-tumor effect of eicosapentaenoic neau RJ, Lembersky B, Posner MC: Role of
tor sub-type involved a neuropeptide-Y in- acid and its effect on protein turnover. Cancer nutrition support during induction chemora-
duced food intake. Nature 1996;382:168–171. Res 1991;51:6089–6093. diation therapy in esophageal cancer. J Parent-
36 Donaldson CJ, Sutton SW, Perrin, Corrigan 54 Nelson KA, Walsh D, Sheehan FA: The cancer er Enteral Nutr 1998;22:18–21.
AZ, Lewis KA, Rivier JE, Vaughan JM, Vale anorexia-cachexia syndrome. J Clin Oncol 73 Body JJ: Metabolic sequelae of cancers (exclud-
WW: Cloning and characterization of human 1994;12:213–225. ing bone marrow transplantation). Curr Opin
urocortin. Endocrinology 1996;137:2167– 55 Nitenberg G, Raynard B: Nutritional support Clin Nutr Metab Care 1999;2:339–344.
2170. of the cancer patient: issues and dilemmas. Crit 74 Body JJ: The syndrome of anorexia-cachexia.
37 Spina M, Merlo-Pich E, Chan RK, Basso AM, Rev Oncol Hematol 2000;34:137–168. Curr Opin Oncol 1999;11:255–260.
Rivier J, Vale W, Koob GF: Appetite suppress- 56 DeWys WD: Anorexia as a general effect of 75 Bozetti F: Effects of artificial nutrition on the
ing effect of urocortin, a CRF-related neuro- cancer. Cancer 1979;43(suppl):2013–2019. nutritional status of cancer patients. J Parenter
peptide. Science 1996;273:1561–1564. 57 DeWys WD, Walters K: Abnormalities of taste Enteral Nutr 1989;13:406–420.
38 Tisdale MJ: Biology of cachexia. J Natl Cancer sensation in cancer patients. Cancer 1975;36: 76 Chen MK, Souba WW, Copeland EM: Nutri-
Inst 1997;89:1763–1773. 1888–1896. tional support of the surgical oncology patient.
39 Inui A: Cancer anorexia-cachexia syndrome: 58 Bernstein IL: Learned taste aversions in chil- Hematol Oncol Clin North Am 1991;5:125–
Are neuropeptides the key? Cancer Res 1999; dren receiving chemotherapy. Science 1978; 145.
59:4493–4501. 200:1302–1303. 77 Miller M: Can reducing caloric intake also help
40 Moldawer LL, Rogy MA, Lowry SF: The role of 59 Glass MJ, Billington CJ, Levine AS: Opioids reduce cancer? J Natl Cancer Inst 1998;90:
cytokines in cancer cachexia. J Parenter Enter- and food intake: Disturbed functional neural 1766–1767.
al Nutr 1992;16(suppl):43–49. pathways? Neuropeptides 1999;33:360–368. 78 Nelson KA: The cancer anorexia-cachexia syn-
41 Noguchi Y, Yoshikawa T, Matsumoto A, Svan- 60 Fujimiya M, Inui A: Peptidergic regulation of drome. Semin Oncol 2000;27:64–68.
iger G, Gelin J: Are cytokines possible media- gastrointestinal motility in rodents. Peptides 79 Mercadante S: Parenteral versus enteral nutri-
tors of cancer cachexia? Surg Today 1996;26: 2000;21:11565–1582. tion in cancer patients: Indications and prac-
467–475. 61 Tache Y, Garrick T, Raybould H: Central ner- tice. Support Care Cancer 1998;6:85–93.
42 Matthys P, Billiau A: Cytokines and cachexia. vous system action of peptides to influence gas- 80 Gagnon B, Bruera E: A review of the drug treat-
Nutrition 1997;13:763–770. trointestinal motor function. Gastroenterology ment of cachexia associated with cancer. Drugs
43 Gelin J, Moldawer LL, Lonnroth C, Sherry B, 1990;98:517–528. 1998;55:675–688.
Chizzonite R, Lundholm K: Role of endoge- 62 Inui A, Okano H, Miyamoto M, Aoyama N, 81 Loprinzi CL, Michalak JC, Schaid DJ: Phase
nous tumor necrosis factor-· and interleukin-1 Uemoto M, Baba S, Kasuga MI: Delayed gas- three evaluation of four doses of megestrol ace-
for experimental tumor growth and the devel- tric emptying in bulimic patients. Lancet 1995; tate as therapy for patients with cancer anorex-
opment of cancer cachexia. Cancer Res 1991; 346:1240. ia and/or cachexia. J Clin Oncol 1993;11:762–
51:415–421. 63 Okano H, Inui A, Ueno N, Morimoto S, Ohmo- 767.
44 Moldawer LL, Copeland EM: Proinflammato- to A, Miyamoto M, Aoyama N, Nakajima Y, 82 Mantovani G, Maccio A, Massa E, Madeddu
ry cytokines, nutritional support and the ca- Baba S, Kasuga M: EM523l, a nonpeptide mo- C: Managing cancer-related anorexia/cachexia.
chexia syndrome: Interactions and therapeutic tilin agonist, stimulated gastric emptying and Drugs 2001;61:499–514.
options. Cancer 1997;79:1828–1839. pancreatic polypeptide secretion. Peptides 83 Downer S, Joel S, Allbright A, Plant H, Stubbs
45 Haslett PA: Anticytokine approaches to the 1996;17:895–900. L, Talbot D, Slevin M: A double-blind placebo-
treatment of anorexia and cachexia. Semin On- 64 Mitchell EP: Gastrointestinal toxicity of che- controlled trial of medroxyprogesterone acetate
col 1998;25(suppl):53–57. motherapeutic agents. Semin Oncol 1992;19: in cancer cachexia. Br J Cancer 1993;67:1102–
46 Mantovani G, Maccio A, Lai P, Massa E, 566–579. 1105.
Ghiani M, Santona MC: Cytokine involvement 65 Copeland EM, MacFadyen BV Jr, Lanzotti VJ, 84 Simons JP, Aaronson NK, Vansteenkiste JF,
in cancer-related anorexia/cachexia: Role of Dudrick SJ: Intravenous hyperalimentation as ten Velde GP, Muller MJ, Drenth BM, Erd-
megestrol acetate and medroxyprogesterone. an adjunct to cancer chemotherapy. Am J Surg kamp FL, Cobben EG, Schoon EJ, Smeets JB,
Semin Oncol 1998;25(suppl 6):45–52. 1975;129:167–173. Schouten HC, Demedts M, Hillen HF, Blijham
47 Marks DL, Ling N, Cone RD: Role of the cen- 66 Copeland EM, Souchon EA, MacFadyen BV GH, Wouters EF: Effects of medroxyprogeste-
tral melanocortin system in cachexia. Cancer Jr, Rapp MA, Dudrick SJ: Intravenous hyper- rone acetate on appetite, weight and quality of
Res 2001;61:1432–1438. alimentation as an adjunct to radiation thera- life in advanced stage non-hormone sensitive
py. Cancer 1977;39:609–616. cancer: A placebo-controlled multicenter
study. J Clin Oncol 1996;14:1077–1084.

What We Have Learned about Cachexia in Dig Dis 2003;21:198–213 211


Gastrointestinal Cancer
85 Fietkau R, Riepl M, Kettner H: Supportive use 100 Popiela T, Lucchi R, Giongo F: Methylpred- 115 Wigmore SJ, Ross JA, Falconer JS, Plester
of megestrol acetate in patients with head and nisolone as palliative therapy for female ter- CE, Tisdale MJ, Carter DC, Fearon KC: The
neck cancer during radio(chemo)therapy. Eur J minal cancer patients. Eur J Cancer Clin On- effect of polyunsaturated fatty acids on the
Cancer 1997;33:75–79. col 1989;25:1823–1829. progress of cachexia in patients with pan-
86 Loprinzi CL, Kugler J, Sloan J: Phase III ran- 101 Willox JC, Corr J, Shaw J, Richardson M, creatic cancer. Nutrition 1996;12(suppl):27–
domized comparison of megestrol acetate, Calman KC, Drennan M: Prednisolone as an 30.
dexamethasone and fluoxymesterone for the appetite stimulant in patients with cancer. Br 116 Barber MD, Ross JA, Voss AC, Tisdale MJ,
treatment of cancer anorexia/cachexia. Proc Med J (Clin Res Ed) 1984;288:27. Fearon KC: The effect of an oral nutritional
ASCO 1997;16:48A. 102 Gagnon B, Bruera E: A review of the drug supplement enriched with fish oil on weight
87 McCarthy WD, Crowder RE, Dryden S, Wil- treatment of cachexia associated with cancer. loss in patients with pancreatic cancer. Br J
liams G: Megestrol acetate stimulates food and Drugs 1998;55:675–688. Cancer 1999;81:80–86.
water intake in the rat: effects on regional hypo- 103 Fainsinger R: Pharmacological approach to 117 Gogos CA, Ginopoulos P, Salsa B, Apostoli-
thalamic neuropeptide Y concentrations. Eur J cancer anorexia and cachexia; in Bruera E, dou E, Zoumbos NC, Kalfarentzos F: Dietary
Pharmacol 1994;265:99–102. Higginson I (eds): Cachexia-Anorexia in Can- ˆ3 polyunsaturated fatty acids plus vitamin E
88 Costa AM, Spence KT, Plata-Salaman CR, cer Patients. Oxford, Oxford University restore immunodeficiency and prolong sur-
French-Mullen JM: Residual Ca2+ channel cur- Press, 1996, pp 128–140. vival for severely ill patients with generalized
rent modulation by megestrol acetate via a G- 104 Argiles JM, Meijsing SH, Pallares-Trujillo J, malignancy: A randomized control trial. Can-
protein-a s-subunit in rat hypothalamic neu- Guirao X, Lopez-Soriano FJ: Cancer cachex- cer 1998;82:395–402.
rones. J Physiol 1995;487:291–303. ia: A therapeutic approach. Med Res Rev 118 Periera J, Bruera E: Chronic nausea; in
89 Mantovani G, Maccio A, Esu S, Lai P, Santona 2001;21:83–101. Bruera E, Higginson I (eds): Cachexia-An-
MC, Massa E, Dessi D, Melis G, Del Giacco S: 105 Daly JM, Redmond HP, Gallagher H: Peri- orexia in Cancer Patients. Oxford, Oxford
Medroxyprogesterone acetate reduces the pro- operative nutrition in cancer patients. J Par- University Press, 1996, pp 23–27.
duction of cytokines and serotonin involved in enter Enteral Nutr 1992;16(suppl):100–105. 119 Davis MP, Dickerson D: Cachexia and an-
anorexia/cachexia and emesis by peripheral 106 Forbes GB: The effect if anabolic steroids on orexia: Cancer’s covert killer. Support Care
blood mononuclear cells of cancer patient. Bio- lean body mass: The dose-response curve. Cancer 2000;8:180–187.
chem Soc Trans 1997;25:296S. Metabolism 1985;34:571–573. 120 Loprinzi CL, Kuross SA, O’Fallon JR, Gesme
90 DeVita VT Jr, Hellman S, Rosenberg SA: Can- 107 Chlebowski RT, Herrold J, Ali I, Oktay E, DH Jr, Gerstner JB, Rospond RM, Cobau
cer Principles and Practice of Oncology, ed 5. Chlebowski JS, Ponce AT, Heber D, Block CD, Goldberg RM: Randomized placebo-
Philadelphia, Lippincott Raven, 1997. JB: Influence of nadrolone decanoate on controlled evaluation of hydrazine sulfate in
91 Bruera E, Fainsinger RL: Clinical management weight loss in advanced non-small cell lung patients with advanced colorectal cancer. J
of cachexia and anorexia; in Doyle D, Hanks cancer. Cancer 1986;58:183–186. Clin Oncol 1994;12:1121–1125.
G, MacDonald N (eds): Oxford Textbook of 108 Kardinal CG, Loprinzi CL, Schaid DJ, Hass 121 Loprinzi CL, Goldberg RM, Su JQ, Mailliard
Palliative Medicine. London, Oxford Medical, AC, Dose AM, Athmann LM, Mailliard JA, JA, Kuross SA, Maksymiuk AW, Kugler JW,
1993, pp 330–337. McCormack GW, Gerstner JB, Schray MF: A Jett JR, Ghosh C, Pfeifle DM: Placebo-con-
92 Steer KA, Kurtz AB, Honour JW: Megestrol- controlled trial of cyproheptadine in cancer trolled trial of hydrazine sulfate in patients
induced Cushing’s syndrome. Clin Endocrinol patients with anorexia and/or cachexia. Can- with newly diagnoses non-small-cell lung can-
1995;42:91–93. cer 1990;65:2657–2662. cer. J Clin Oncol 1994;12:1126–1129.
93 Leinung MC, Liporace R, Miller CH: Introduc- 109 Meguid MM, Fetissov SO, Varma M, Sato T, 122 Kosty MP, Fleishman SB, Herndon JE 2nd,
tion of adrenal suppression by megestrol ace- Zhang L, Laviano A, Rossi-Fanelli F: Hypo- Coughlin K, Kornblith AB, Scalzo A, Morris
tate in patients with AIDS. Ann Intern Med thalamic dopamine and serotonin in the regu- JC, Mortimer J, Green MR: Cisplatin, vin-
1995;122:843–845. lation of food intake. Nutrition 2000;16:843– blastine and hydrazine sulfate in advanced,
94 Bruera E, Macmillan K, Kuehn N, Hanson J, 857. non-small-cell lung cancer: A randomized,
MacDonald RN: A controlled trial of megestrol 110 Laviano A, Meguid MM, Yang ZJ, Gleason placebo-controlled, double-blind phase III
acetate on appetite, caloric intake, nutritional JR, Cangiano C, Fanelli FR: Cracking the rid- study of the Cancer and Leukemia Group B. J
status, and other symptoms in patients with dle of cancer anorexia. Nutrition 1996;12: Clin Oncol 1994;12:1113–1120.
advanced cancer. Cancer 1990;66:1279–1282. 706–710. 123 Bruera E, Neumann CM, Pituskin E, Calder
95 Loprinzi CL, Ellison NM, Schaid DJ, Krook 111 Moertel CG, Kvols LK, Rubin J: A study of K, Ball G, Hanson J: Thalidomide in patients
JE, Athmann LM, Dose AM, Mailliard JA, cyproheptadine in the treatment of metastatic with cachexia due to terminal cancer: Prelim-
Johnson PS, Ebbert LP, Geeraerts LH: Con- carcinoid tumor and the malignant carcinoid inary report. Ann Oncol 1999;10:857–859.
trolled trial of megestrol acetate for the treat- syndrome. Cancer 1991;67:33–36. 124 Reyes-Teran G, Sierra-Madero JG, Martinez
ment of cancer, anorexia and cachexia. J Natl 112 Edelman MJ, Gandara DR, Meyers FJ, Ishii del Cerro V, Arroyo-Figueroa H, Pasquetti A,
Cancer Inst 1990;82:1127–1132. R, O’Mahony M, Uhrich M, Lauder I, Hous- Calva JJ, Ruiz-Palacios GM: Effects of thali-
96 Heckmayr M, Gatzeneir U: Treatment of can- ton J, Gietzen DW: Serotonergic blockade in domide on HIV-associated wasting syn-
cer weight loss in patients with advanced lung the treatment of the cancer anorexia-cachexia drome: A randomized, double-blind, placebo-
cancer. Oncology 1992;49(suppl 2):32–34. syndrome. Cancer 1999;86:684–688. controlled clinical trial. AIDS 1996;10:1501–
97 Feliu J, Gonzalez-Baron M, Berracal A: Useful- 113 Tayek JA, Bistrian BR, Hehir DJ, Martin R, 1507.
ness of megestrol acetate in cancer cachexia Moldawer LL, Blackburn GL: Improved pro- 125 Haslett P, Tramontana J, Burroughs M,
and anorexia. J Clin Oncol 1992;15:436–440. tein kinetics and albumin syntheses by Hempstead M, Kaplan G: Adverse reactions
98 Moertel CG, Schutt AJ, Reitemeier RJ, Hahn branched chained amino acid-enriched total to thalidomide in patients infected with hu-
RG: Corticosteroid therapy of preterminal gas- parenteral nutrition in cancer cachexia. A man immunodeficiency virus. Clin Infect Dis
trointestinal cancer. Cancer 1974;33:1607– prospective randomized crossover trial. Can- 1997;24:1223–1227.
1609. cer 1986;58:147–157. 126 McMillan DC, O’Gorman P, Fearon KC,
99 Bruera E, Roca E, Cedaro L, Carraro S, Chacon 114 Cangiano C, Laviano A, Meguid MM, Mu- McArdle CS: A pilot study of megestrol ace-
R: Action of oral methylprednisolone in termi- lieri M, Conversano L, Preziosa I, Rossi- tate and ibuprofen in the treatment of cachex-
nal cancer patients: A randomized, double- Fanelli F: Effects of administration of oral ia in gastrointestinal cancer patients. Br J
blind study. Cancer Treat Rep 1985;69:751– branched-chain amino acids on anorexia and Cancer 1997;76:788–790.
754. caloric intake in cancer patients. J Natl Can-
cer Inst 1996;88:550–552.

212 Dig Dis 2003;21:198–213 Palesty/Dudrick


127 Dezube BJ, Sherman ML, Fridovich-Keil JL, 133 Plasse TF, Gorter RW, Krasnow SH, Lane M, 141 Carbo N, Lopez-Soriano J, Tarrago T, Gon-
Allen-Ryan J, Pardee AB: Down-regulation of Shepard KV, Wadleigh RG: Recent clinical zalez O, Llovera M, Lopez-Soriano FJ, Ar-
tumor necrosis factor expression by pentoxi- experience with dronabinol. Pharmacol Bio- giles JM: Comparative effects of ß2-adrener-
fylline in cancer patients: A pilot study. Can- chem Behav 1991;40:695–700. gic agonists on muscle waste associated with
cer Immunol Immunother 1993;36:57–60. 134 Beal JE, Olson R, Laubenstein L, Morales JO, tumour growth. Cancer Lett 1997;115:113–
128 Lissoni P, Ardizzoia A, Barni S, Pittalis S, Bellman P, Yangco B, Lefkowitz L, Plasse TF, 118.
Rossini F, Porta A, Tancini G: Characteriza- Shepard KV: Dronabinol as a treatment for 142 Maltin CA, Delday MI, Watson JS, Heys SD,
tion of cancer-related disseminated intravas- anorexia associated with weight loss in pa- Nevison IM, Ritchie IK, Gibson PH: Clenbu-
cular coagulation in relation to tumor necro- tients with AIDS. J Pain Symptom Manage terol, a ß-adrenoreceptor agonist, increases
sis factor-· blood concentrations: Possible 1995;10:89–97. relative muscle strength in orthopaedic pa-
therapeutic role of pentoxifylline. Tumori 135 Fainsinger RL, Tapper M, Bruera E: A per- tients. Clin Sci 1993;84:651–654.
1996;82:78–80. spective on the management of delirium in 143 McMillan DC, Leen E, Smith J, Sturgeon C,
129 Goldberg RM, Loprinzi CL, Mailliard JA, the terminally ill. J Palliat Care 1993;9:4–8. Preston T, Cooke TG, McArdle CS: Effect of
O’Fallon JR, Krook JE, Ghosh C, Hestorff 136 Costelli P, Garcia-Martinez C, Llovera M, extended ibuprofen administration on the
RD, Chong SF, Reuter NF, Shanahan TG: Carbo N, Lopez-Soriano FJ, Agell N, Tessi- acute phase protein response in colorectal
Pentoxifylline for treatment of cancer anorex- tore L, Baccino FM, Argiles JM: Muscle pro- cancer patients. Eur J Surg Oncol 1995;21:
ia and cachexia? A randomized, double-blind, tein waste in tumor-bearing rats is effectively 531–534.
placebo-controlled trial. J Clin Oncol 1995; antagonized by a ß2-adrenergic agonist (clen- 144 Preston T, Fearon KC, McMillan DC, Win-
13:2856–2859. buterol): Role of the ATP-ubiquitin-depen- stanley FP, Slater C, Shenkin A, Carter DC:
130 Tanaka Y, Eda H, Fujimoto K, Tanaka T, dent proteolytic pathway. J Clin Invest 1995; Effect of ibuprofen on the acute-phase re-
Ishikawa T, Ishitsuka H: Anticachectic activi- 95:2367–2372. sponse and protein metabolism in patients
ty of 5-deoxy-5-fluorouridine in a murine tu- 137 Chance WT, Cao LQ, Zhang FS, Foley-Nel- with cancer and weight loss. Br J Surg 1995;
mor cachexia model, colon 26 adenocarcino- son T, Fischer JE: Clenbuterol treatment in- 82:229–234.
ma. Cancer Res 1990;50:4528–4532. creases muscle mass and protein content of 145 Wigmore SJ, Falconer JS, Plester CE, Ross
131 Hussey HJ, Todorov PT, Field WN, Inagaki tumor-bearing rats maintained on total par- JA, Maingay JP, Carter DC, Fearon KC: Ibu-
N, Tanaka Y, Ishitsuka H, Tisdale MJ: Effect enteral nutrition. J Parenter Enteral Nutr profen reduces energy expenditure and acute-
of a fluorinated pyrimidine on cachexia and 1991;15:530–535. phase protein production in cancer patients
tumor growth in murine cachexia models: Re- 138 Rehfeldt C, Weikard R, Reichel K: The effect compared with placebo in pancreatic cancer
lationship with a proteolysis inducing factor. of the ß-adrenergic agonist clenbuterol on the patients. Br J Cancer 1995;72:185–188.
Br J Cancer 2000;83:56–62. growth of skeletal muscles of rats. Arch Tier- 146 McMillan DC, Wigmore SJ, Fearon KC,
132 Nelson K, Walsh D, Deeter P, Sheehan F: A ernähr 1994;45:333–344. O’Gorman P, Wright CE, McArdle CS: A pro-
phase II study of delta-9-tetrahydrocannabi- 139 Young OA, Watkins S, Oldham JM: The role spective randomized study of megesterol ace-
nol for appetite stimulation in cancer-associ- of insulin-like growth factor I in clenbuterol- tate and ibuprofen in gastrointestinal cancer
ated anorexia. J Palliat Care 1994;10:14–18. stimulated growth in growing lambs. J Anim patients with weight loss. Br J Cancer 1999;
Sci 1995;73:3069–3077. 79:495–500.
140 Zhou GH, Han ZK: Effects of dietary supple-
mentation of ß2-adrenergic agonist clenbute-
rol on carcass characteristics and some me-
tabolites in ducks. Br Poult Sci 1994;35:355–
361.

What We Have Learned about Cachexia in Dig Dis 2003;21:198–213 213


Gastrointestinal Cancer
Review Article

Dig Dis 2003;21:214–219


DOI: 10.1159/000073338

Nutritional Support in Acute


Pancreatitis
Costas Avgerinos Spiros Delis Spyros Rizos Christos Dervenis
1st Department of Surgery, Konstantopoulion, Agia Olga Hospital, Athens, Greece

Key Words Acute pancreatitis (AP) is a disease with a wide spec-


Acute pancreatitis W Gut barrier W Total parenteral trum of clinical courses, ranging from the mild form with
nutrition W Enteral nutrition minimum morbidity and almost zero mortality, to the
severe form with a high percentage of complications and a
high risk for a lethal outcome [1, 2]. The latter form is
Abstract characterized by various degrees of necrosis of pancreatic
Acute pancreatitis (AP), mainly the severe necrotizing parenchyma as well as local and systemic complications
type, results in extreme energy demands which might such as SIRS and MOF, representing a typical hypermeta-
lead, if prolonged, to severe malnutrition. Besides that, bolic septic model with high-energy consumption and
starving during AP contributes to gut barrier dysfunc- protein breakdown that leads to severe malnutrition. On
tion, the main cause of bacterial translocation and sep- the other hand, the mild form of the disease is much less
sis. The aim of nutritional support in AP is to prevent mal- aggressive against body metabolism and is usually self-
nutrition and protect the gut by maintaining mucosal limited. Patient’s oral intake starts within 3–7 days after
integrity. Traditionally, nutritional support during the disease onset, resulting in minor and usually easily revers-
acute phase of the disease has been provided through ible nutritional defects. As a result, nutritional support by
total parenteral nutrition (TPN) solutions. However, re- enteral feeding or total parenteral nutrition (TPN) should
cent animal and human studies have identified new pat- be one of the main therapeutic aims in AP, and nutrition-
terns of pancreatic secretion and hormonal stimulation al management should depend on the underlying pan-
during the course of AP, different from those assumed creatic disease form.
for years. Thus it has become feasible to use the natural
enteral route for nutrition with potential benefits com-
pared with TPN. Malnutrition in Acute Pancreatitis
Copyright © 2003 S. Karger AG, Basel

Malnutrition during the clinical course of AP is a result


of a combination of oral food intake restriction, increased
energy demands and often preexisting nutritional defects
(i.e. alcoholic malnutrition). All AP-pathogenetic factors

© 2003 S. Karger AG, Basel Christos Dervenis, MD


ABC 0257–2753/03/0213–0214$19.50/0 1st Department of Surgery, Konstantopoulion
Fax + 41 61 306 12 34 Agia Olga Hospital, 3–5 Ag. Olgas str.
E-Mail karger@karger.ch Accessible online at: GR–14233 Athens (Greece)
www.karger.com www.karger.com/ddi Tel. +30 1 277 546, Fax +30 1 279 3969, E-Mail chrisder@otenet.gr
meet at a starting point where a common pattern leading Under physiologic conditions the entry of nutrients
to the clinical presentation of the disease begins. This into the duodenum results in a burst of pancreaticobiliary
pathway involves activation of trypsinogen to trypsin digestive secretions that mix with the nutrients in order to
after which an activation cascade of various enzymes is be exposed to the small intestine mucosa. It has been
started leading to pancreatic and peripancreatic tissue shown that up to 70–80% of nutrients are absorbed from
autodigestion. At the same time, liberations of acute the lumen as proximal as the duodenojejunal junction
phase reactants resulted in a hemodynamic response simi- [12, 13]. The rest remains unabsorbed within the small
lar to that seen in sepsis syndrome (increased cardiac out- intestine. This physiologic malabsorption is believed to
put, decreased peripheral resistance and increased oxygen act as an energy and amino acid supply to the mucosa of
consumption). This is accompanied by alterations in the small and large intestine [14] with regard to fat, amino
carbohydrates, proteins and lipids metabolism. The dis- acids and carbohydrates in different proportions [14–
ease and its complications are also associated with the 17].
release of cytokines (IL-1, IL-6, IL-8, TNF, PAF), activa- Enteral feeding support in AP aims to avert negative
tion of the compliment and creation of arachidonic acid nitrogen balance, to protect gut barrier function and
metabolic products. These energy-consuming biochemi- therefore prevent secondary pancreatic infection. The
cal alterations in parallel with reduction of food intake rationale behind the concept of enteral feeding is that
result in a negative energy balance and a rapid loss of lean there is at least some evidence that it is important in re-
body mass which adversely affects the host’s defenses and storing and might prevent morphological changes in the
immune competence [3]. Non-suppressive gluconeogene- intestine associated with starvation. Lack of nutrients in
sis is observed in severe pancreatitis and net protein the gut lumen leads to loss of mucosal integrity as a result
catabolism can rise to 40 g nitrogen/day as opposed to of a decrease of mucosal thickness [18]. Enteral feeding
normal subjects where glucose supply cannot inhibit that can also reverse the reduction in hilus height occurring
phenomenon completely [4, 5]. In 80% with severe AP after TPN and theoretically can play an important role in
complicated by sepsis, a marked increase in resting energy the management of severe AP as it probably prevents sep-
expenditure is observed and using the Harris-Benedict sis and immune failure.
equation total energy expenditure is 1.49 times the pre-
dicted resting energy expenditure [6, 7].
Pathophysiology of Pancreas in Acute
Pancreatitis
Pathophysiology of the Gut in Acute
Pancreatitis For many years to put the pancreas at rest in AP was
considered as the best medical practice and the corner-
Contamination of pancreatic necrosis and consequent stone of therapeutic manipulations. It was stated that by
sepsis is the main cause of death in severe pancreatitis, that way exacerbation of pancreatic inflammation and
although in the early period of the disease, SIRS remains autodigestion would be avoided as the absence of content
the main fatal course [8]. Maintenance of gut integrity is in stomach and duodenum diminish further production
of high priority in severe AP in order to prevent pancreat- of pancreatic proteolytic enzymes [19]. The true clinical
ic necrosis contamination and preserve its immune func- significance of that belief has been recently disputed. The
tion. Bacterial translocation through the gut wall is the three phases of exocrine pancreatic secretion (cephalic,
prominently proposed mechanism for pancreatic necrosis gastric, intestinal) and the real pattern of pancreatic out-
contamination. put proved to be different in AP. Pancreatic exocrine
In normal subjects the gut which represents the largest secretion is severely reduced early after the onset of AP
immune organ of the human body acts as a physiologic and the cholecystokinin-stimulated secretion is abolished
barrier against bacteria and endotoxins. Secretory IgA at the time of maximal histological damage to the pan-
produced by gut lymphoid tissue prevents bacterial and creas [20].
viral adherence to the mucosa [9]. Experimental work in Several experimental and clinical trials have shown
rats has shown that gut barrier function in the small intes- that delivery of nutrients into the jejunum does not
tine is compromised very early after the onset of AP. This increase pancreatic secretion and is well tolerated with no
observation has also been documented in humans [10, increase in complications. More specifically, although ad-
11]. ministration of lipid in the duodenum is a strong stimula-

Nutritional Support in Acute Pancreatitis Dig Dis 2003;21:214–219 215


tory factor for pancreatic exocrine secretion, jejunal deliv- among patients with a persistent negative nitrogen bal-
ery of the same amount of lipids causes minimal pancreat- ance. A high incidence of catheter sepsis was also docu-
ic reaction [21–24]. Intravenous lipid infusion has minor mented.
effects as has been shown in human studies. Gastric or In 1991, Kalfarentzos et al. [39] divided 67 patients
duodenal protein or carbohydrate administration is also a with severe AP (13 Ranson criteria) into two groups of
strong stimulus for pancreatic secretion, while as expected early (within 72 h after admission) and late (after 72 h)
jejunal delivery of the same nutrients is harmless to the onset of TPN. They noted a significantly lower incidence
pancreas [25–31]. As there is no evidence that putting the of complications and mortality in the early group but a
pancreas at rest has any clinical benefit, this theory is high catheter sepsis incidence as well. Sax et al. [40] car-
strongly challenged nowadays [32] and enteral feeding has ried out the only randomized controlled trial on the
gained increasing acceptance for nutritional support in effects of early parenteral nutrition versus no nutritional
AP. support in patients with AP. The two groups of patients
(mean Ranson score 1) that received either conventional
therapy alone or conventional therapy with TPN had no
Energy Supply in Acute Pancreatitis significant differences in length of hospital stay, number
of days of oral intake and number of complications as well
According to the ESPEN guidelines [33], a hypocaloric as a ninefold increase in the incidence of catheter sepsis in
energy supply of 15–20 kcal/kg/day is more suitable dur- the TPN group.
ing the early catabolic stage of non-surgical patients with As a conclusion it can be stated that no strong level 1
MOF in AP. In non-severely ill patients, nutritional information regarding the role of TPN in severe AP exists
requirements can be resumed at approximately 25– and more trials are needed in order to show any benefit. A
35 kcal/kg/day with 1.2–1.5 g protein/kg/day. Carbohy- trend for improvement of morbidity and mortality
drates and lipids should be administered 3–6 and 2 g/kg/ though is delineated in patients with severe pancreatitis –
day respectively, but care should be taken to avoid high those with prolonged starvation and those who achieved a
blood glucose and triglyceride concentrations. positive nitrogen balance.

Total Parenteral Nutrition in Acute Pancreatitis Enteral Nutrition in Acute Pancreatitis

Traditionally, TPN has for many years been the only More recently, attention has been given to the possible
nutrient providing treatment in patients with AP and pro- role of the enteral route of delivering the necessary calo-
longed starvation. TPN achieves minimal exocrine pan- ries and nutrients. The rationale behind the concept of
creatic secretions as well as sufficient energy and protein enteral feeding is that there is at least some evidence that
provision. Since 1974, when Feller et al. [34] showed in an is important in restoring and might help in preventing
uncontrolled retrospective study a decrease in the mortal- morphological changes in the intestine associated with
ity rate of patients who received intravenous hyperali- starvation. As reported already, a lack of nutrients in the
mentation while suffering from AP, several other similar gut lumen leads to loss of mucosal integrity as a result of a
retrospective uncontrolled clinical trials have failed to decrease of mucosal thickness [32]. Enteral feeding also
reproduce the same results. On the contrary and despite can reverse the reduction in villus height occurring after
differences in the parameters of the trials, other authors TPN. In an experimental model, rats with pancreatitis
observed a higher incidence of catheter-related sepsis were infused with Ringer’s lactate solution for 48 h fol-
among TPN groups [35, 36] but no difference in total lowed by parenteral or enteral nutrition until the 7th day.
mortality [35–37]. Two prospective non-randomized Results showed lower endotoxin levels, a more prominent
trials have been published on that subject. villus height and T-cell levels in animals that received
In 1989, Sitzmann et al. [38] divided 73 patients with enteral nutrition compared with those that received TPN
AP of various severities into three groups depending on [41]. In theory, enteral nutrition could play an important
their ability to tolerate glucose-free, lipid-based and lipid- role in the treatment of severe AP, as it probably prevents
free nutrition. Within 15 days, most patients in all groups sepsis and immune failure.
achieved an improvement in nutritional status. A higher Since the mid-1980s, some clinical evidence from criti-
mortality rate was observed in the fat-free group as well as cally ill patients, especially those with severe injuries, has

216 Dig Dis 2003;21:214–219 Avgerinos/Delis/Rizos/Dervenis


shown that giving enteral nutrition very early could favor- study, patients with moderate and severe disease were
ably alter the outcome. Moore et al. [42] studied 32 included. Patients who received enteral feeding fared bet-
patients with severe trauma and compared early enteral ter after 7 days with respect to Apache II score and CRP
nutrition with no nutritional intervention. They found a levels compared with the TPN group. The authors also
statistically significant difference in septic morbidity (9 reported an increase in serum IgM endotoxin core anti-
vs. 29%). bodies in the TPN group which remained unchanged in
The first retrospective study for the effects of the enter- the EN group. The total antioxidant capacity was less in
al nutrition in patients with AP was published in 1973 by the former group. They concluded that patients on enteral
Voitk et al. [43], who demonstrated good tolerance and nutrition were exposed to lower endotoxin levels, which
even beneficial effects of an elemental diet delivered in was probably related to preserved host defense.
the stomach of 6 patients suffering from complicated pan- More recently, Abou-Assi and O’Keefe [52] demon-
creatitis. Kudsk et al. [44] and Parekh et al. [45] published strated earlier recovery, shorter hospital stay and shorter
their experiences on enteral feeding in retrospective stud- duration of nutritional support, better tolerance to restart-
ies, the former through feeding jejunostomies and the lat- ing oral feeding and much cheaper costs for nutrition in
er through gastric catheters. In general, they demonstrated the group of 17 enterally fed patients with AP compared
improvement of the nutritional status accompanied by with 16 patients who received TPN. Catheter-related sep-
successful resolution of complications, but in both studies sis and hyperglycemia necessitating insulin were signifi-
enteral nutrition was commenced after the acute phase of cantly more common in the TPN group but overall mor-
pancreatitis. Simpson et al. [46] in a similar study pub- tality was no different. Olah et al. [53] compared conven-
lished favorable results with nasoenteral feeding in 5 tional parenteral nutrition (48 patients) with early jejunal
patients with acute alcoholic pancreatitis, while Nakad et nutrition (41 patients) in 89 patients admitted with AP.
al. [47] in a prospective uncontrolled study described The rate of septic complications, need for surgery, multi-
their experience of early jejunal nutrition starting 36 h ple organ failure and death was higher in the TPN group
after admission. Recently, Eatock et al. [48] in a prospec- but differences were not statistically significant. Finally,
tive uncontrolled study revealed the positive effects of another randomized controlled trial was published by a
gastric delivered nutrients in 26 patients with severe AP. Scottish group [54] that studied the effect of early enteral
Overall, all of these papers have proved that enteral nutrition on markers of the inflammatory response in pre-
nutrition can be started in patients with AP since it is a dicted severe AP. Serum IL-6, tumor necrosis factor
safe method of nutritional support, it is well tolerated and receptor I and CRP were used as inflammatory markers.
causes no aggravation of the disease. On the contrary, Despite previous findings, the authors documented that
Powell et al. [49] published in 2001 the only randomized early enteral nutrition did not ameliorate the inflammato-
controlled study comparing 13 patients that received ry response in patients with severe AP compared with no
enteral nutrition within 72 h of disease onset with 14 oth- nutritional intervention.
ers treated conventionally (nil by mouth). It was shown in A randomized study is under way by our group to try
this study that early enteral nutrition did not ameliorate and identify the role of early enteral nutrition in severe
the inflammatory response as demonstrated by a serum AP compared with standard TPN in reducing the need for
concentration of inflammatory response markers (CRP, surgery in patients with predicted severe AP. We recently
IL-6, sTNFRI). Furthermore, abnormal intestinal perme- reported preliminary results (23 patients) where we
ability occurred more frequently in patients receiving showed that early enteral nutrition seems to reduce surgi-
enteral nutrition. cal interventions in the enteral nutrition group by reduc-
Recently, five randomized controlled studies were ing the incidence of sepsis (9 vs. 33%) [55].
published comparing enteral feeding (EN) with TPN. Kal- Although the current practice is to use nasojejunal
farentzos et al. [50] randomized 38 patients, all with tubes, placed endoscopically or under radiographic
severe AP, in two groups (EN vs. TPN). They found a screening, a recent study by the Glasgow group [48]
significant reduction in total, including septic complica- showed that nasogastric feeding is usually possible in
tions in the enteral feeding group. The cost was three severe AP. They reported that this practice is safe, well
times lower in EN than in TPN and they suggested the tolerated and without any sign of clinical or biochemical
preferable use of EN in all patients with severe disease. deterioration.
The other study was from the UK [51], and 34 patients Another question that needs to be answered is the pos-
were also randomized in two groups (EN vs. TPN). In that sible role of immune-enhanced enteral diets in severe AP

Nutritional Support in Acute Pancreatitis Dig Dis 2003;21:214–219 217


[56]. There are a number of reports, mainly in severely Conclusions
injured patients, dealing with this aspect [56–58]. A meta-
analysis [57] including 1,009 patients from 11 trials At present there is no definite evidence that artificial
showed that immune-modulated regimens resulted in a nutrition support – either total parenteral or enteral –
significant reduction of infectious complications and alters the outcome in most patients with AP, unless mal-
length of hospital stay, but with no effect on survival. nutrition is also a problem. Diagnosis of AP is not itself an
Only one study has dealt with the use of glutamine in AP, indication for instituting artificial nutrition, unless severi-
as a supplement in the standard TPN. They found that ty of the disease is the case. Enteral nutrition support is
glutamine improves leukocyte activity and reduces proin- safe, well tolerated and does not stimulate the pancreas
flammatory cytokine release in AP. No conclusions can be compared with parenteral feeding, and therefore should
drawn from these studies, but as it seems possible that be used preferably in the treatment or prevention of mal-
immune-enriched diets could play a role, further trials are nutrition and probably immunosuppression and infec-
needed to clarify this issue. tion, in patients with AP. Finally, larger, well-conducted
trials, recruiting only patients with severe AP and strati-
fying them for disease severity, nutritional status and eti-
ology of pancreatitis before randomization, are needed
before any conclusive statement on the benefits of nutri-
tional support on outcome can be made.

References

1 Wycoll DL: The management of severe acute 10 Andersson R, Yang X, Sun Z, Deng X, Soltesz 18 Johnson C, Kudsk K: Nutrition and intestinal
necrotizing pancreatitis: An evidence-based re- V, Ihse I: Effect of a platelet-activating factor mucosal immunity. Clin Nutr 1999;18:337–
view of the literature. Intensive Care Med antagonist on pancreatitis-associated gut bar- 344.
1999;25:146–156. rier dysfunction in rats. Pancreas 1998;17:107– 19 Wyncoll D: The management of severe acute
2 Dervenis C, Johnson CD, Bassi C et al: Diagno- 119. necrotizing pancreatitis: An evidence-based re-
sis, objective assessment of severity and man- 11 Ammori BJ, Leeder PC, King RF, Barclay GR, view of the literature. Intensive Care Med
agement of acute pancreatitis: Santorini Con- Martin IG, Larvin M, McMahon MJ: Early 1999;25:146–156.
sensus Conference. Int J Pancreatol 1999;25: increase in intestinal permeability in patients 20 Lobo D, Memon M, Allison S, Rowlands B:
195–210. with severe acute pancreatitis: Correlation with Evolution of nutritional support in acute pan-
3 Scolapio JS, Mahli-Chowla N, Ukleja A: Nutri- endotoxaemia, organ failure and death. J Gas- creatitis. Br J Surg 2000;87:695–707.
tion supplementation in patients with acute trointest Surg 1998;3:252–262. 21 Burns GP, Stein TA: Pancreatic enzyme secre-
and chronic pancreatitis. Gastroenterol Clin 12 Carriere F, Barrowman JA, Verger R, Lougier tion during intravenous fat infusion. J Parenter
North Am 1999;28:695–707. R: Secretion and contribution to lipolysis of Enteral Nutr 1987;11:60–62.
4 Eckerwall G, Andersson R: Early enteral nutri- gastric and pancreatic lipases during a test meal 22 Fried GM, Ogden WD, Rhea A, Greeley G,
tion in severe acute pancreatitis: A way of pro- in humans. Gastroenterology 1993;105:876– Thompson JC: Pancreatic protein secretion
viding nutrients, gut barrier protection, immu- 888. and gastrointestinal hormone release in re-
nomodulation or all of them? Scand J Gas- 13 Holtmann G, Kelly DG, Sternby B, DiMagno sponse to parenteral amino acids and lipids in
troenterol 2001;5:449–458. EP: Survival of human pancreatic enzymes dogs. Surgery 1982;92:902–905.
5 Heys S, Walker L, Smith I, Eremin O: Enteral during small bowel transit: Effect of nutrients, 23 Stabile BE, Borzatta M, Stubbs RS, Debas HT:
nutritional supplementation with key nutrients bile acids and enzymes. Am J Physiol 1997; Intravenous mixed amino acids and fats do not
in patients with critical illness and cancer. Ann 273:G553–G558. stimulate exocrine pancreatic secretion. Am J
Surg 1999;229:467–477. 14 Anderson IH, Levine AS, Levitt MD: Incom- Physiol 1984;246:G274–G280.
6 Dickerson RN, Vehe KL, Mullen JL, Feurer plete absorption of the carbohydrate in all-pur- 24 Edelmann K, Valenzuela JE: Effect of intrave-
ID: Resting energy expenditure in patients with pose wheat flour. N Engl J Med 1981;304:891– nous feeding on human pancreatic secretion.
pancreatitis. Crit Care Med 1991;19:484–490. 892. Gastroenterology 1983;85:1063–1068.
7 Bouffard YH, Delafosse BX, Annat GJ, Viale 15 Layer P, Go VL, Dimagno EP: Fate of pan- 25 Niederau C, Sonnenberg A, Erckenbrecht J:
JP, Bertrand OM, Motin JP: Energy expendi- creatic enzymes during small intestinal aboral Effects of intravenous infusion of amino acids,
ture during severe acute pancreatitis. J Paren- transit in humans. Am J Physiol 1986;251: fat, or glucose on unstimulated pancreatic se-
ter Enteral Nutr 1989;13:26–29. G475–G480. cretion in healthy humans. Dig Dis Sci 1985;
8 Schwarz M, Büchler M, Thomsen J et al: Pan- 16 Layer P, Zinsmeister AR, Dimagno EP: Effects 30:445–455.
creatic infection in acute pancreatitis: A fre- of decreasing intraluminal amylase activity on 26 Stabile BE, Debas HT: Intravenous vs. intradu-
quent finding. Digestion 1993;54:A119. starch digestion and postprandial gastrointesti- odenal amino acids, fats and glucose as stimu-
9 Underdown BJ, Schiff JM: Immunoglobulin A: nal function in humans. Gastroenterology lants of pancreatic secretion. Surg Forum 1981;
Strategic defence initiative at the mucosal sur- 1986;91:41–48. 32:224–226.
face. Annu Rev Immunol 1986;4:389–417. 17 Layer P, von der One MR, Holst JI, Jansen MJ: 27 Ragins H, Levenson SM, Singer R, Stamford
Altered postprandial motility in chronic pan- W, Seifner E: Intrajejunal administration of an
creatitis: Role of malabsorption. Gastroenter- elemental diet at neutral pH avoids pancreatic
ology 1997;112:1624–1634. stimulation. Studies in dog and man. Am J
Surg 1973;126:606–614.

218 Dig Dis 2003;21:214–219 Avgerinos/Delis/Rizos/Dervenis


28 Johnson LR, Schanbacker LM, Dudrick SJ, 39 Kalfarentzos FE, Karavias DD, Karatzas TM, 50 Kalfarentzos F, Kehagias J, Mead N, Kokkinis
Copeland EM: Effect of long-term parenteral Alevizatos BA, Androulakis LA: Total paren- K, Gogos C: Enteral nutrition is superior to
feeding on pancreatic secretion and serum se- teral nutrition in severe acute pancreatitis. J parenteral nutrition in severe acute pancreati-
cretin. Am J Physiol 1977;233:E524–E529. Am Coll Nutr 1991;10:156–164. tis: Results of a randomised prospective trial.
29 Grant JP, Davey-McCrae J, Snyder PJ: Effect 40 Sax AC, Warner BW, Talamini MA, Hamilton Br J Surg 1997;83:349–353.
of enteral nutrition on human pancreatic secre- FN, Bell RH Jr, Fischer JE: Early total paren- 51 McClave SA, Greene LM, Snider HL, Makk
tions. J Parenter Enteral Nutr 1987;11:302– teral nutrition in acute pancreatitis: Lack of LJ, Cheadle WG, Owens NA et al: Comparison
304. beneficial effects. Am J Surg 1987;153:117– of the safety of early enteral vs. parenteral
30 Vidon N, Hecketsveiler P, Butel J: Effect of 124. nutrition in mild acute pancreatitis. J Parenter
continuous jejunal perfusion of elemental and 41 Kotani J, Usami M, Nomura H: Enteral nutri- Enteral Nutr 1997;21:14–20.
complex nutritional solutions on pancreatic en- tion prevents bacterial translocation but does 52 Abou-Assi S, O’Keefe SJD: Nutrition support
zyme secretion in human subjects. Gut 1978; not improve survival in acute pancreatitis. during acute pancreatitis. Nutrition 2002;18:
19:194–198. Arch Surg 1999;134:287–292. 938–943.
31 Variyam EP, Fuller RK, Brown FM, Quallich 42 Moore F, Moore E, Kudsk K et al: Clinical ben- 53 Olah A, Pardavi G, Belagyi T, Nagy A, Issekutz
LG: Effect of parenteral amino acids on human efits of an immune enhancing diets for early A, Mohamed GE: Early nasojejunal feeding in
pancreatic exocrine secretion. Dig Dis Sci post-injury feeding. J Trauma 1994;37:607– acute pancreatitis is associated with a lower
1985;30:445–455. 615. complication rate. Nutrition 2002;18:259–
32 Saluja AK, Steer M: Pathophysiology of pan- 43 Voitk A, Brown RA, Echave V, McArdle AH, 262.
creatitis. Role of cytokines and other mediators Gurd FN, Thompson AG: Use of an elemental 54 Powell JJ, Murchison JT, Fearon KCH, Ross
of inflammation. Digestion 1999;60(suppl diet in the treatment of complicated pancreati- JA, Siriwardena AK: Randomized controlled
1):27–33. tis. Am J Surg 1973;125:223–227. trial of the effect of early enteral nutrition on
33 Meier R, Beglinger C, Layer P, Gullo L, Keim 44 Kudsk KA, Campbell SM, O’Brien T, Fuller R: markers of the inflammatory response in pre-
V, Laugier R, Friess H, Schweitzer M, Macfie Postoperative jejunal feeding following compli- dicted severe acute pancreatitis. Br J Surg
J, Espen Consensus Group: Espen guidelines cated pancreatitis. Nutr Clin Pract 1990;5:14– 2000;87:1375–1381.
on nutrition in acute pancreatitis. Clin Nutr 17. 55 Paraskeva C, Smailis D, Priovolos A, Sofianou
2002;21:173–183. 45 Parekh D, Lawson HH, Segal I: The role of K, Lytras D, Avgerinos C, Rizos S, Karagiannis
34 Feller JH, Brown RA, Toussaint GPM, total enteral nutrition in pancreatic disease. S J, Dervenis C: Early enteral nutrition reduces
Thompson AG: Changing methods in the treat- Afr J Surg 1993;31:57–61. the need for surgery in severe acute pancreati-
ment of severe pancreatitis. Am J Surg 1974; 46 Simpson WG, Marsano L, Gates L: Enteral tis. Pancreatology 2001;1:372.
127:196–201. nutritional support in acute alcoholic pancre- 56 Eckerwall G, Andersson R: Early enteral nutri-
35 Goodgame JT, Fischer JE: Parenteral nutrition atitis. J Am Coll Nutr 1995;14:662–665. tion in severe acute pancreatitis: A way of pro-
in the treatment of acute pancreatitis: Effects 47 Nakad A, Piessevaux H, Marot JC, Hoang P, viding nutrients, gut barrier protection, immu-
on complications and mortality. Ann Surg Geubel A, Van Steenberger W: Is early enteral nomodulation or all of them? Scand J Gas-
1977;186:651–658. nutrition in acute pancreatitis dangerous? troenterol 2001;5:449–458.
36 Grand JP, James S, Grabowski V, Trexler KM: About 20 patients fed by endoscopically placed 57 Heys S, Walker L, Smith I, Eremin O: Enteral
Total parenteral nutrition in pancreatic dis- nasogastrojejunal tube. Pancreas 1998;17:187– nutritional supplementation with key nutrients
ease. Ann Surg 1984;200:627–631. 193. in patients with critical illness and cancer. Ann
37 Robin AP, Campbell R, Palini CK, Liu K, 48 Eatock F, Brombacher G, Steven A, Imrie C, Surg 1999;229:467–477.
Donahue PE, Nyhus LM: Total parenteral nu- McKay C, Carter R: Nasogastric feeding in 58 Olah A, Belagyi T, Issekutz A, Gamal ME,
trition during acute pancreatitis: Clinical expe- severe acute pancreatitis may be practical and Bengmark S: Randomized clinical trial of spe-
rience with 156 patients. World J Surg 1990; safe. Int J Pancreatol 2000;28:25–31. cific lactobacillus and fibre supplement to early
14:572–579. 49 Powell JJ, Murchison JT, Fearon KC, Ross JA, enteral nutrition in patients with acute pancre-
38 Sitzmann JV, Steinborn PA, Zinner MJ, Came- Siriwardena AK: Randomized controlled trial atitis. Br J Surg 2002;89:1103–1107.
ron JN: Total parenteral nutrition and alter- of the effect of early enteral nutrition on mark-
nate energy substrates in treatment of severe ers of the inflammatory response in predicted
acute pancreatitis. Surg Gynecol Obstet 1989; severe acute pancreatitis. Br J Surg 2000;87:
168:311–317. 1357–1381.

Nutritional Support in Acute Pancreatitis Dig Dis 2003;21:214–219 219


Review Article

Dig Dis 2003;21:220–227


DOI: 10.1159/000073339

Nutrition and Inflammatory Bowel


Disease: Its Relation to
Pathophysiology, Outcome and Therapy
Miquel A. Gassull
Department of Gastroenterology and Hepatology, Hospital Universitari Germans Trias i Pujol, Badalona,
Catalonia, Spain

Key Words Several facts relate nutrition and inflammatory bowel


Ulcerative colitis W Crohn’s disease W Inflammatory bowel disease (IBD) and many features even support the exis-
disease W Malnutrition W Enteral nutrition W Dietary fat W tence of a bidirectional relationship between them. In this
Unabsorbable dietary carbohydrates W Colonic sense, a high prevalence of nutritional derangement
fermentation (macro- and micronutrients), both in ulcerative colitis
(UC) and Crohn’s disease (CD), has been reported [1–5].
In addition, qualitative differences in diet composition
Abstract have been suggested as being the cause of the low inci-
Nutritional deficiencies are frequent in patients with dence of IBD and other immune-based diseases in spe-
ulcerative colitis and Crohn’s disease, and negatively cific populations [6]. Finally, it has been shown that
influence the outcome of the disease. Growth retarda- changes in some components of the diet are able to down-
tion, osteopenia and thromboembolic phenomena are regulate the inflammatory response in vitro, in animals
some of the inflammatory bowel disease complications and in humans [7–14].
in which nutritional deficits are involved. Moreover, nu- In this paper the above-mentioned aspects will be
trients can play a role in the pathogenesis of the disease reviewed, with special emphasis on data relating to nutri-
and, in some cases, can be a primary therapeutic tool. tional deficiencies, IBD pathophysiology, clinical out-
Enteral nutrition has proven to play a therapeutic role in come and the changes of incidence in some geographical
Crohn’s disease. The nutrient(s) responsible for this ef- areas. Also the evidence about the role of nutritional
fect are not well identified but dietary fat appears to be a manipulation in inducing and maintaining clinical dis-
major factor. In ulcerative colitis, unabsorbable carbohy- ease remission will be discussed.
drates can modulate the intestinal microbial environ-
ment, thus contributing to improve colonic inflamma-
tion. Protein Energy Malnutrition and Micronutrient
Copyright © 2003 S. Karger AG, Basel Deficit in IBD: Their Effect on Disease Outcome

Various degrees of protein energy malnutrition, vita-


min, mineral and trace element deficiencies may develop
in UC and CD [1–5]. Anorexia has been blamed for many

© 2003 S. Karger AG, Basel Miquel A. Gassull MD, PhD, Assoc. Prof. Med.
ABC 0257–2753/03/0213–0220$19.50/0 Head, Department of Gastroenterology and Hepatology
Fax + 41 61 306 12 34 Hospital Universitari Germans Trias i Pujol, Carretera del Canyet s/n
E-Mail karger@karger.ch Accessible online at: ES–08916 Badalona, Catalonia (Spain)
www.karger.com www.karger.com/ddi Tel./Fax +34 93 4978951, E-Mail mgassull@ns.hugtip.scs.es
years as being one of the main causes of IBD-associated tional formulas may achieve both goals; this aspect will be
malnutrition, growth failure and sexual development re- dealt with later in this article.
tardation in children and adolescents. The cause of these Suboptimal levels of micronutrients are often found
derangements is multifactorial and related to the exis- both in children and adults with IBD, but except for iron
tence of active inflammation. In animals with experimen- and folate, it is unusual to discover symptoms attributable
tal colitis, this clinical situation has been reproduced and to these deficits [3–5]. In spite of this, subclinical deficits
found to be especially related to the release of IL-1 by the may have a pathophysiological significance since they
inflamed mucosa [13–15]. may favor self-perpetuation of the disease (because of
Other causes of malnutrition in IBD include a hyper- defects in the mechanisms of tissue repair), defective
metabolic state, due to inflammation, extensive intestinal defense against damage produced by oxygen free radical
involvement or surgical resection of the small intestine, and can facilitate lipid peroxidation [22, 23], even in clin-
interfering with nutrient absorption, or an increase of ically inactive or mildly active disease [24], as well as the
nutrient losses through the inflamed and ulcerated gut development of dysplasia in the intestinal mucosa [25].
(small and large bowel). In addition, the existence of fistu- However, the potential therapeutic role of antioxidant
las and/or strictures in the small intestine facilitates bacte- micronutrients, both in inducing disease remission and in
rial overgrowth. The excess of bacteria results in an the prevention of disease relapse, has scarcely been inves-
increased production of bacterial metabolites of nutrients tigated. Recently, it was shown that the administration of
through fermentation and hydroxylation processes. Some vitamins E and C for 4 weeks in mild to moderate cases of
of the products such as folate may have a beneficial effect, CD induced a significant reduction of oxidative stress as
however an excess of short-chain fatty acids and especial- measured by breath pentane and ethane output, plasma
ly fatty acid hydroxylation results in increased diarrhea lipid peroxides and F-2 isoprostane, suggesting increased
and hence nutrient wasting. Bacterial overgrowth also requirements of such micronutrients [26].
induces vitamin B12 deficiency. Finally, medical treat- One of the most risky situations in patients with acute
ment (therapeutic fasting, steroids, cholestyramine) [16, IBD is the appearance of thromboembolic complications,
17] and self- or medically-recommended dietary restric- related in part to the existence of nutritional imbalances,
tions may also contribute the development of nutritional resulting in excessive plasma homocysteine. Increased
deficiencies in IBD patients [10, 17]. homocysteine levels have proved to be thrombogenic and
Nutritional derangement may influence adult IBD out- an independent risk factor for cardiovascular disease [27,
come, since it may increase the need for surgical treat- 28] and deep-vein thrombosis [29]. This sulfur-containing
ment [1] and its complications [17]. However, it has been amino acid is formed during the demethylation of methi-
in children and adolescents where malnutrition, in its onine. Hyperhomocysteinemia has been reported to be
wider sense, together with steroid treatment (by IGF-1 prevalent in patients with IBD [28, 29] and related to both
suppression) have been blamed as inducing one of the low folate [30] and vitamin B12 [31] levels. However, only
most severe IBD complications in this population – linear a small amount of homocysteine is cleared via a remethy-
growth and sexual maturation failure [18–20]. Experi- lation pathway dependent on vitamin B12 and folate, but
ments in rats with colitis have shown the important most of the homocysteine is converted to cystothione by
impact of the excessive release of proinflammatory cyto- cystothione ß-synthase which is a vitamin B6-dependent
kines by the inflamed mucosa on growth retardation [21]. enzyme. Although low vitamin B6 levels were reported in
However, nutritional deterioration can also be blamed for IBD patients many years ago [3], it is only recently that
causing more than 50% of the delayed growth observed in they have been related to the high homocysteine levels in
these experimental animals, because it is associated with a IBD and the risk of thrombosis [32].
low IGF-1 synthesis. Feeding properly colitic rats partial- Osteopenia associated with IBD is an area of increas-
ly reversed the linear growth deficit, but it never reached ing concern, especially in CD [33–37]. Nutritional defi-
that of the control animals [21]. These studies strongly ciencies, inflammatory cytokines and treatment with cor-
support the fact that decreasing the inflammatory process ticosteroids have been incriminated in its pathogenesis
and feeding patients may adequately improve linear [38, 39]. The disease itself seems to exert a direct effect in
growth in the infant and adolescent IBD population. inducing osteopenia in these patients. This was shown in
These objectives could be reached by using concomitantly an in vitro experiment in which the serum of patients with
immunomodulatory therapy and nutritional support. CD impaired osteoblastic function [40]. In 36 new cases
There is also the possibility that especially designed nutri- of pediatric CD, a dual X-ray absorptiometry study dem-

Nutrition and Inflammatory Bowel Disease Dig Dis 2003;21:220–227 221


onstrated osteopenia and osteoporosis in 17 and 25% molecules, lymphocyte chemotactic activity and an in-
respectively [41]. A bone fracture rate as high as 40% has creased synthesis of eicosapentaenoic (n-3) acid-derived
been reported in a population-based cohort study [42]. It eicosanoids with attenuated proinflammatory activity [8,
is noteworthy that low bone mineral density has been 9, 54–57]. The hypothesis is supported both by in vitro
described in adult CD patient populations as distant as and in vivo studies [53–59]. Moreover, increases in the
Puerto Rico, The Netherlands and Korea [43–45]. In annual incidence in both UC and CD have been observed
some cases, this abnormality is detected in newly diag- in countries where qualitative changes in dietary fat were
nosed cases in both adults [45] and children [46]. Support- established because of public health reasons [60–62] or
ing the fact that inflammation is an important factor in when hit by western dietary influences as in Japan [63].
inducing osteopenia in IBD is that TNF-·, IL-1ß and IL- The major dietary changes in these countries were the
11 (proinflammatory cytokines) promote the secretion of substitution of butter by margarine in Sweden [62] and
a ligand able to stimulate receptors in osteoclast precur- the decrease in intake of fish while increasing that of meat
sors, inducing osteoclast maturation and bone resorption and seed oils in Japan [63]. These changes, as mentioned,
[47]. were followed by increases in the incidence of IBD in
Glucocorticoids, the gold standard treatment in IBD, these countries.
are associated with side effects including permanent dam-
age such as cataracts, growth failure, sexual development
retardation, and osteopenia. In fact, glucocorticoids are Is Diet a Factor in the Etiopathogenesis of IBD?
an important factor aggravating bone loss in IBD, because Does It Explain the Therapeutic Effect of
they decrease intestinal calcium absorption, increase re- Enteral Nutrition?
nal calcium excretion and induce parathyroid hormone
secretion. The result is decreased bone formation and Nutrients in the form of chemically and physicochemi-
enhanced bone resorption [46, 47]. These data should be cally complex food products are one of the main compo-
enough to restrict their use in the treatment of IBD, both nents of the intestinal environment. As such, the possibili-
as long-term therapy (even at low doses) and in patients ty that the food antigenic load may be one of the triggering
with frequent relapses (steroid-dependent) [48–51]. Bone factors initiating and perpetuating the abnormal intesti-
loss induced by steroid therapy is dose- and duration- nal inflammatory response has been frequently invocated.
dependent. In addition, growth and sexual development This has been the basis for using therapeutic fasting in
retardation in children and adolescents may be aggravat- acute UC and CD (bowel rest), amino acid-base elemental
ed by glucocorticoid treatment due to its suppressive diets (low antigenic power) for treating acute CD and
effect on IGF-1, which mediates the effect of growth hor- excluding the suspected offending food (exclusion diets)
mone on the growth of plate bones [21]. Then it is highly in the treatment of active or maintenance of remission in
recommendable in this population to use steroids only CD.
when strictly necessary, in short treatments, avoid re- Various studies have shown that withholding nutrients
peated treatments (use other immunosuppressors) and from the intestinal lumen does not influence disease out-
especially try nutrition as primary treatment. come [64–66], even in acute disease. Elemental diets have
shown to offer no advantage as compared to whole pro-
tein-based polymeric diets (higher antigenic load) in in-
Possible Role of Nutrition in the Incidence of ducing remission in active CD, at least in three meta-anal-
IBD in Some Geographical Areas yses summarizing the findings of several trials [67–69].
The effect of exclusion diets in inducing or maintaining
The low incidence of IBD and other immune-based remission is still doubtful, since the reintroduction of the
diseases in some communities like Greenland Eskimos, as offensive food frequently is not accompanied by disease
compared to western populations, has been attributed to relapse [70]. Recently, experimental work in vitro (human
differences in lifestyle and the type of diet [52, 53]. It has peripheral blood lymphocytes) and humans (skin and rec-
been hypothesized that components of the Eskimo’s diet tal mucosa) has tried to ascertain whether or not intestinal
(abundant in n-3 fatty acid-rich marine oils) produce epithelium and immune cells have an increased sensitivi-
changes in cell membrane composition (including that of ty to different components of the diet. Although peripher-
the immune-competent cells) [53]. This would result in a al blood lymphocytes and rectal mucosa are not, under
downregulated expression of certain cytokines, adhesion normal circumstances, exposed to intact food, the results

222 Dig Dis 2003;21:220–227 Gassull


showed increased sensitivity in CD compared to healthy one. In this sense, a recent paper showed that the use in an
individuals [71, 72]. The true value in etiopathogenic and enteral formula diet of an artificially made mixture with a
therapeutic terms of these findings needs to be further highly predominant (70%) synthetic fatty acid, did not
explored. obtain any therapeutic effect as compared to one using a
Several years ago, Gonzalez-Huix et al. [73] first sug- natural source [74]. This fact also raises the question of
gested that fat composition of the diet, rather than the the role of the non-fatty acid component of the lipid
nitrogen source, may explain why enteral nutrition may sources, since many compounds are found, at least in
have an anti-inflammatory effect in active CD. This was some oil sources that may have, through their antioxidant
based on the fact that in a randomized clinical trial a poly- effect, immune regulatory properties [85, 86]. This possi-
meric enteral diet (higher antigenic load than elemental) bility has to be further explored.
containing 30% of the calories as lipids (65% olive oil, An interesting but seldom mentioned aspect is the
10% milk fat and 25% MCT) was as effective as 1 mg/kg potential anti-inflammatory role of MCT. These com-
b.w. of prednisolone. These authors hypothesized that the pounds have always been regarded as energy providers in
key factor for the therapeutic effectiveness of enteral malabsorptive states or easy sources of energy for mal-
nutrition in CD was the amount (elemental low fat) or the nourished individuals. However, it has been shown exper-
type of fat used in the design of the formula diet. This imentally [84, 87–89] that MCT modulates the expression
hypothesis has been reinforced in a recent paper [74]. In of adhesion molecules and cytokines and the metabolism
the literature a great deal of data supports the immuno- of linoleic acid (precursor of potent proinflammatory
modulatory role of dietary fat. The effect that fatty acids eicosanoids). Also, a polymeric enteral diet containing
have from different sources in modifying cell membrane high amounts of MCT is as effective as a very low fat ele-
composition and hence the influence on the function of mental diet in inducing remission in active CD [90]. In
the proteins embedded in it, has already been mentioned. fact, when the composition of the diets reported as highly
The consequences of this are relevant in terms not only on effective in inducing remission in active CD are carefully
the type of eicosanoids and other second messengers syn- analyzed, it can be observed that those with a higher
thesized [9], but also in the cell signaling system, influenc- remission rate are those with very low fat content and
ing the activity of nuclear transcription factors and hence those where one third of the fat source was MCT [73,
the genes expressing proteins involved in the inflammato- 91].
ry process (cytokines, adhesion molecules, iNOS, etc.) A further aspect related to dietary fat is its influence in
[75–83]. It is worth emphasizing that usually there is a the cell cycle. A feature in CD is the resistance of T lym-
tendency, especially by clinical nutritionists, in using the phocytes to the natural apoptotic process, leading to its
words fat and fatty acids synonymously. Mammals do not accumulation in the bowel wall and hence contributing to
eat single fatty acids in the diet but different fat sources perpetuate the inflammatory process [92]. Some lipid
with mixtures of fatty acids. The predominance of one or sources (fish oil or olive oil, but not corn oil) induce apop-
some of them give to the fat source not only its physico- tosis in intestinal cell lines cultures by, at least, inhibiting
chemical properties, but also its nutritional and metabolic the expression of the antiapoptotic mitochondrial protein
value. In this sense it is worth mentioning that different Bcl-2 [93]. This may be a further mechanism explaining
fat sources with different predominant fatty acids have why some fat sources show a positive therapeutic effect in
different effects on the expression of different cytokines in active CD while others do not.
an elegant experimental setting in mice [84]. It may not be To summarize this part, it can be said that although T
the effect of a single fatty acid but the combination of sev- lymphocytes and rectal mucosa have shown sensitization
eral that exerts the immunomodulatory action. An exam- to some food products, the therapeutic effect of exclusion
ple of this possibility is fish oil, which has been made syn- diets is still not well established and there is no difference
onymous of a fat source rich in n-3 eicosapentanoic (EPA) in the rate of remission achieved by whole protein or ami-
and docosahexaenoic acid (DHA). In fact, fish oil con- no acid formula diets when used as primary treatment in
tains no more than 25% of EPA and DHA together, the CD.
larger component being saturated fatty acids (LCT + Nutrients as components of cell structures: Changes in
MCT) 45%, with 16% of monounsaturated and 10% of dietary components can influence numerous cell func-
n-6 fatty acids [84]. It is then plausible that the anti- tions and the cell cycle. Fat is, up to now, the best studied
inflammatory effect would be related more to an adequate nutrient and has been shown to influence immune cell
mixture of fatty acids (fat source) than to a single response, although many other aspects of these com-

Nutrition and Inflammatory Bowel Disease Dig Dis 2003;21:220–227 223


pounds need to be explored. So far, changes in the compo- and butyrate) are involved in the maintenance of colonic
sition of dietary fat have been shown to be able to structure (butyrate is the preferred fuel for the colonocyte)
influence disease outcome in CD and some data relate and immune homeostasis. The first consequence of short-
major qualitative changes in fat intake in geographically chain fatty acid synthesis is the decrease of intraluminal
distant populations (Sweden and Japan) with an increase pH in the colon [95]. This fact favors the development of
in the incidence of CD, suggesting a possible role as trigger Lactobacilli, Bifidobacteria strains (probiotics) and a de-
in genetically susceptible individuals. Little is known crease of those of Clostridia, Bacteroides, Escherichia coli.
about the potential effect of changes in dietary fat on This is known as the ‘prebiotic effect’ effect of fiber.
intestinal bacterial populations. As mentioned, short-chain fatty acid, especially buty-
rate, is oxidized by the colonic mucosa and used mainly as
an energy provider. In recent years it has been postulated
Nutrition and Ulcerative Colitis that butyrate oxidation is incomplete or defective in both
UC and pouchitis, suggesting that this defect may be
Up to now in this review, the relationship between implicated in the pathogenesis of both conditions [96–
nutrition and UC has seldom been mentioned, except for 103]. Rectally administered 13C-labelled butyrate was sig-
its capacity of inducing nutritional derangement. When nificantly less oxidized by active UC patients than by
dealing with the role of enteral nutrition, the effect of dif- healthy controls, whereas patients with inactive disease
ferent dietary fat composition in primary therapy, its showed a trend towards a lower oxidation than healthy
influence in disease etiopathogenesis and incidence, subjects [104]. These findings in non-active UC are sup-
mostly CD has been mentioned. In fact the enteral formu- ported by other studies [105].
la diets used as primary treatment in CD have proven not Recent work suggests that in active UC there is an
to be effective in UC, although improve patients’ nutri- excess of SH2 synthesis by sulfate-reducing bacteria in the
tional status and prevent complications related to surgery, colon [103]. This excessive SH2 contributes to the ineffi-
when compared to parenteral nutrition [94]. One of the cient butyrate oxidation in the colonic mucosa [100]. In
different possible explanations is that while CD is an patients with proctitis refractory to treatment with topical
antiapoptotic disease, UC is not [92]. Moreover, there are steroids and 5-aminosalicylic acid (5-ASA), the nutrition-
major differences in the etiopathogenesis of both diseases, al approach, by intrarectally administering butyrate as
as far as the environmental factors are concerned. The maintenance treatment for such a type of UC, has pro-
most obvious is the effect of smoking, being protective in duced significantly positive results as compared to place-
UC and inductor in CD. More is known about the rela- bo [99, 106]. Moreover, the relapse rate of quiescent UC
tionship between the intestinal environment and the co- treated with soluble dietary fiber (Plantago ovata seeds),
lonic mucosa. Finally, the concentration of bacteria in the substrate precursor for the production of butyrate in the
colon is much higher than that in the small intestine and, colon (prebiotic), was not different than that of patients
even though endogenous bacteria play a crucial role in the given 5-ASA at the usual maintenance doses [107], al-
pathogenesis of CD, little is known about the relationship though there was a non-significant trend towards a lower
between small intestinal bacteria and nutrients present in relapse rate in those patients on combined therapy. Inter-
the intestinal lumen in such a condition. estingly enough, patients on treatment with this fiber
The colon is highly dependent of the environment in preparation increased butyrate production in the colon
its lumen to maintain its structure and function. Almost (as measured by stool analysis) as compared to those on
80% of the energy required for such a task is obtained 5-ASA alone [107]. This nutritional approach in the main-
from nutrients reaching the large bowel. Several sub- tenance therapy of UC supports the role of butyrate in the
strates enter the colon every day, these include non- pathogenesis of the UC.
absorbed carbohydrates, proteins and a few others. In However, butyrate is not only an energy provider, but a
short, anaerobic bacteria deal with these substrates true anti-inflammatory substance, since it prevents the
through a fermentation process producing branched- activation of the nuclear transcription factor NFÎB and
chain fatty acids from proteins and short-chain fatty acids through this mechanism prevents the expression of cyto-
plus a large volume gas (H2 and CO2) from carbohydrates. kines and other molecules involved in the inflammatory
The gas volume is reduced by the action of methanogenic process [108–110]. This is achieved by the inhibition of
and sulfate-reducing bacteria producing CH4 and SH2 the phosphorylation or degradation of the inhibitory pro-
respectively. Short-chain fatty acids (acetate, propionate tein IÎB that holds NFÎB in the cell cytoplasm. Butyrate

224 Dig Dis 2003;21:220–227 Gassull


would be a dietary factor regulating the inflammatory that of CD, with a more clear interaction in the intestinal
process from the intestinal lumen. Its therapeutic possibil- ecosystem in the former. Thus a nutritional therapeutic
ities as primary therapy in active UC are currently under approach may also be possible in UC. The interest in the
investigation. Its anti-inflammatory properties, together knowledge of the possible role of nutritional imbalances
with its prebiotic action, strongly suggest the possibility of in IBD relies on its potential usefulness of nutritional
using a combined prebiotic-probiotic approach (symbiot- manipulation as a preventive measure in genetically sus-
ic) in the treatment of UC and pouchitis. ceptible individuals and as its use as a true biological ther-
The UC nutritional approach both in terms of patho- apy, free of side effects, in certain groups or subgroups of
genesis and therapy seems to follow a different route than patients with IBD.

References

1 Gassull MA, Abad A, Cabré E, González-Huix 11 Morris GP, Beck PL, Herridge MS, Depew 21 Ballinger A, Azooz O, El-Haj Y, Poole S, Farth-
F, Giné JJ, Dolz C: Enteral tube feeding in WT, Szewczuk MR, Wallace JL: Hapten-in- ing MJG: Growth failure occurs through a de-
inflammatory bowel disease. Gut 1986; duced model of chronic inflammation and ul- crease in insulin-like growth factor, which is
27(suppl):76–80. ceration of the rat colon. Gastroenterology independent of undernutrition in the rat model
2 Cabré E, Fernández-Bañares F, Esteve M, Gas- 1989;96:795–803. of colitis. Gut 2000;46:1–6.
sull MA: Micronutrients in inflammatory bow- 12 McHugh KJ, Weingarten HP, Keenan C, Wal- 22 Miralles-Barrachina O, Savoye G, Belmonte-
el disease. J Clin Nutr Gastroenterol 1989;4: lace J, Collins SM: On the suppression of food Zalar L, Hochain P, Ducrotté P, Hecketsweiler
100–102. intake in experimental models of colitis in the B, Lerebours E, Déchelotte P: Low levels of glu-
3 Fernández-Bañares F, Abad A, Xiol X, Giné rat. Am J Physiol 1993;264:R871–R876. tathione in endoscopic biopsies of patients with
JJ, Dolz C, Cabré E, Esteve M, González-Huix 13 McHugh K, Castonguay TW, Collins SM, Crohn’s colitis: The role of malnutrition. Clin
F, Gassull MA: Vitamin status in patients with Weingarten HP: Characterization of suppres- Nutr 1999;18:313–317.
inflammatory bowel disease. Am J Gastroen- sion of food intake following acute colonic 23 Fernández-Bañares F: Papel de los metabolitos
terol 1989;84:744–748. inflammation in the rat. Am J Physiol 1993; de oxı́geno reactivos en la patogenia de la enfer-
4 Fernández-Bañares F, Mingorance MD, Esteve 265:R1001–R1005. medad inflamatoria intestinal. Gastroenterol
M, Cabré E, Lachica M, Abad A, Gil A, Hum- 14 Otterness IG, Seymour PA, Golden HW, Rey- Hepatol 1995;18:526–532.
bert P, Boix J, Gassull MA: Serum zinc, copper nolds JA, Daumy GO: The effects of contin- 24 Wendland B, Aghdassi E, Tam C et al: Lipid
and selenium levels in inflammatory bowel dis- uous administration of interleukin-1 in the rat. peroxidation and plasma antioxidant micronu-
ease. Am J Gastroenterol 1990;85:1584–1589. Physiol Behav 1989;43:797–804. trients in Crohn’s disease. Am J Clin Nutr
5 Abad A, Fernández-Bañares F, Cabré E, Gil A, 15 Ballinger A, El-Haj T, Perrett D, Turvill J, 2001;74:259–264.
Esteve M, González-Huix F, Xiol X, Gassull Obeid O, Dryden S, Williams G, Farthing 25 Lashner BA, Provencher KS, Seidner DL, Kne-
MA: The effect of total enteral tube feeding on MJG: The role of medial hypothalamic sero- sebeck A, Brzezinski A: The effect of folic acid
the vitamin status of malnourished patients tonin in the suppression of feeding in a rat supplementation on the risk for cancer or dys-
with inflammatory bowel disease. Int J Vitam model of colitis. Gastroenterology 2000;118: plasia in ulcerative colitis. Gastroenterology
Nutr Res 1988;58:428–435. 544–553. 1997;112:29–32.
6 Kromann N, Green A: Epidemiological studies 16 Gassull MA, Cabré E, Vilar L, Montserrat A: 26 Aghdassi E, Wendland E, Steinhard H et al:
in the Upernavik district, Greenland. Acta Nivel de ingesta alimentaria y su posible papel Antioxidant vitamin supplementation in
Med Scand 1980;208:401–406. en el desarrollo de malnutrición calórico-pro- Crohn’s disease decreases oxidative stress: A
7 O’Morain C, Segal AW, Levi AJ: Elemental teica en pacientes gastroenterológicos hospital- randomized controlled trial. Am J Gastroen-
diets as primary treatment of acute Crohn’s izados. Med Clin (Barc) 1985;85:85–87. terol 2003;98:348–353.
disease. Br Med J 1984;228:1859–1862. 17 Gassull MA, Fernández Bañares F: Nutrition 27 Graham IM, Daly LE, Refsum HM et al: Plas-
8 De Caterina, R, Cybulsky MI, Clinton SK, in inflammatory bowel disease; in Payne- ma homocysteine as a risk factor for vascular
Gimbrone M, Libby P: The ˆ-3 fatty acid James J, Grimble G, Silk DBA (eds): Artificial disease. The European Concerted Action Pro-
docosahexanoate reduces cytokine-induced ex- Nutrition Support in Clinical Practice, ed 2. ject. JAMA 1997;277:1775–1781.
pression of proatherogenic and proinflamma- London, Greenwich Medical Media, 2001, pp 28 Nygard O, Nordrehaug JE, Refsum H et al:
tory proteins in human endothelial cells. Arter- 555–573. Plasma homocysteine levels and mortality in
ioscler Thromb 1994;14:1829–1836. 18 González-Huix F, Fernández Bañares F, Es- patients with coronary artery disease. N Engl J
9 Lee T, Hoover R, Williams J, Sperling R, Rav- teve M, Abad A, Cabré E, Acero D, Figa M, Med 1997;337:230–236.
alese J III, Spur B, Robinson D, Corey E, Lewis Guilera M, Humbert P, De León R, Gassull 29 Den Heijer M, Koster T, Blom HJ et al: Hyper-
R, Austen K: Effect of dietary enrichment with MA: Enteral versus parenteral nutrition as ad- homocysteinemia as a risk factor for deep-vein
eicosapentaenoic and docosahexaenoic acids junct therapy in acute ulcerative colitis. Am J thrombosis. N Engl J Med 1996;334:759–762.
on in vivo neutrophil and monocyte leuco- Gastroenterol 1993;88:227–232. 30 Chowers Y, Sela BA, Holland R et al: Increased
triene generation and neutrophil function. N 19 Winter HS: Special consideration in pediatric levels of homocysteine in patients with Crohn’s
Engl J Med 1985;312:1217–1224. inflammatory bowel disease; in Targan SR, disease are related to folate levels. Am J Gas-
10 Gassull MA, Fernández Bañares F, Cabré E, Shanahan F (eds): Inflammatory Bowel Dis- troenterol 2000;95:3498–3502.
Esteve M: Enteral nutrition in inflammatory ease. From Bench to Bedside. Baltimore, Wil- 31 Romagnuolo J, Fedorak RN, Dias VC: Hyper-
bowel disease; in Rombeau JL, Rolandelli RH liams & Wilkins, 1994, pp 701–710. homocysteinemia and inflammatory bowel dis-
(eds): Clinical Nutrition. Enteral and Tube 20 Griffiths AM, Nguyen P, Smith C, MacMillan ease: Prevalence and predictors in a cross-sec-
Feeding, ed 3. Philadelphia, Saunders, 1997, H, Sherman PM: Growth and clinical course of tional study. Am J Gastroenterol 2001;96:
pp 403–416. children with Crohn’s disease. Gut 1993;34: 2143–2149.
939–943.

Nutrition and Inflammatory Bowel Disease Dig Dis 2003;21:220–227 225


32 Salveni S, Cattaneo M, Vecchi M et al: Low 46 Thearle M, Horlick M, Bilezikian JP, Levy J, 64 Lochs H, Meryn S, Marosi L, Ferenci P, Hort-
vitamin B6 plasma levels, a risk factor for Gertner JM, Levine LS, Harbison M, Berdon nag H: Has bowel rest a beneficial effect in the
thrombosis, in inflammatory bowel disease: W, Oberfield SE: Osteoporosis: An unusual treatment of Crohn’s disease? Clin Nutr 1983;
Role of inflammation and correlation with presentation of childhood Crohn’s disease. J 2:61–64.
acute phase reactants. Am J Gastroenterol Clin Endocrinol Metab 2000;85:2122–2126. 65 Dickinson RJ, Ashton MG, Axon ATR, Smith
2003:98:112–117. 47 Roux S, Orcel P: Bone loss: Factors that regu- RC, Yeung CK, Hill GL: Controlled trial of
33 Silvennoinen JA, Karttunen TJ, Niemelae SE, late osteoclast differentiation. An update. Ar- intravenous hyperalimentation and total bowel
Manelius JJ, Lehtola JK: A controlled study of thritis Res 2000;2:451–456. rest as an adjunct to routine treatment of acute
bone mineral density in patients with inflam- 48 Canalis E: Mechanisms of glucocorticoid ac- colitis. Gastroenterology 1980;79:1199–1204.
matory bowel disease. Gut 1995;37:71–76. tion in bone: Implications to corticosteroid- 66 McIntyre PB, Powel-Tuck J, Wood SR et al:
34 Bjarnason I, MacPherson A, Mackintosh C, induced osteoporosis. J Clin Endocrinol Metab Controlled trial of bowel rest in the treatment
Buxton-Thomas M, Forgacs I, Moniz C: Re- 1996;81:3441–3447. of severe acute colitis. Gut 1986;27:481–485.
duced bone density in patients with inflamma- 49 Hodsman AB, Toogood JH, Jennings B, Fraher 67 Fernandez-Bañares F, Cabré E, Esteve-Comas
tory bowel disease. Gut 1997;40:228–233. LJ, Baskerville JC: Differential effect of in- M, Gassull MA: How effective is enteral nutri-
35 Jahnsen J, Falch JA, Aadland E, Mowinckel P: haled budesonide and oral prednisone in serum tion in inducing remission in active Crohn’s
Bone mineral density is reduced in patients osteocalcin. J Clin Endocrinol Metab 1991;72: disease? A meta-analysis of the randomized
with Crohn’s disease but not in patients with 530–540. clinical trials. J Parenter Enteral Nutr 1995;19:
ulcerative colitis: A population-based study. 50 Schoon EJ, Wolffenbuttel BHR, Stockbrügger 356–364.
Gut 1997;40:313–319. RW: Osteoporosis as a risk in inflammatory 68 Griffiths AM, Ohlsson A, Sherman PM, Su-
36 Martin JP, Tonge KA, Bohnsle U, Jacyna MR, bowel disease. Drugs Today 1999;35(suppl therland LR: Meta-analysis of enteral nutrition
Levi J: Bone mineral density in patients with A):17–28. as a primary treatment of active Crohn’s dis-
inflammatory bowel disease. Eur J Gastroen- 51 Adinoff AD, Hollister R: Steroid-induced frac- ease. Gastroenterology 1995;108:1056–1067.
terol Hepatol 1999;11:537–541. tures and bone loss in patients with asthma. N 69 Mesori A, Trallori G, D’Albasio G, Milla M,
37 Cowan FJ, Warner JT, Dunstan FD, Evans Engl J Med 1983;309:265–268. Vannozzi G, Pacini F: Defined formula diets
WD, Gregory JW, Jenkins HR: Inflammatory 52 Kromann N, Green A: Epidemiological studies versus steroids in the treatment of active
bowel disease and predisposition to osteopenia in the Upernavik district, Greenland. Acta Crohn’s disease. A meta-analysis. Scand J Gas-
Arch Dis Child 1997;76:325–329. Med Scand 1980;208:401–406. troenterol 1996;31:267–272.
38 Semeao E, Jawad AF, Stoufer NO, Babette SZ, 53 Bang H, Dyerberg J, Hjorne N: The composi- 70 Riordan AM, Hunter LJO, Cowman RE et al:
Piccoli DA, Stalling VA: Risk factors for low tion of food consumed by Greenland Eskimos. Treatment of active Crohn’s disease by exclu-
bone mineral density in children and young Acta Med Scand 1976;200:69–73. sion diet: East Anglian multicentre controlled
adults with Crohn’s disease. J Pediatr 1999; 54 Sanderson P, Calder PC: Dietary fish oil di- trial. Lancet 1993;342:1131–1134.
135:593–600. minishes lymphocyte adhesion to macrophage 71 Van Den Bogaerde J, Kamm MA, Knight SC:
39 Issenman RM: Bone mineral metabolism in and endothelial dell monolayers. Immunology Immune sensitization to food, yeast and bacte-
pediatric inflammatory bowel disease. In- 1998;94:79–87. ria in Crohn’s disease. Aliment Pharmacol
flamm Bowel Dis 1999;5:192–199. 55 De Caterina R, Libby P: Control of endothelial Ther 2001;15:1647–1653.
40 Hymans JS, Syzga N, Kreutzer DL, Justinich leucocyte adhesion molecules by fatty acids. 72 Van Den Bogaerde J, Cahill J, Emmanuel AV,
CJ, Gronowicz GA: Alterations in bone metab- Lipids 1996;31(suppl):57–63. Vaizey CJ, Talbot IC, Knight SC, Kamm MA:
olism in children with inflammatory bowel dis- 56 Yeh SL, Chang KY, Huang C, Chen WJ: Effect Gut mucosal response to food antigens in
ease: An in vitro study. J Pediatr Gastroenterol of n-3 and n-6 fatty acids on plasma eicosanoid Crohn’s disease. Aliment Pharmacol Ther
Nutr 1996;24:289–295. and liver antioxidant enzymes in rats receiving 2002;11:1903–1915.
41 Sylvester FA, Hymans JS, Justinich C, Zeiter total parenteral nutrition. Nutrition 1997;13: 73 Gonzalez-Huix F, de Leon R, Fernandez-Ba-
D, Davis P, Harris K, Hawker G, Griffiths 32–36. ñares F, Esteve M, Cabré E, Acero D, Abad-
AM: Osteopenia is frequent in children with 57 Miles EA, Calder PC: Modulation of immune Lacruz A, Figa M, Guilera M, Planas R, Gas-
Crohn’s disease at diagnosis (abstract). J Pe- function by dietary fatty acids. Proc Nutr Soc sull MA: Polymeric enteral diets as primary
diatr Gastroenterol Nutr 1999;29:A524. 1998;57:277–292. treatment of active Crohn’s disease: A prospec-
42 Bernstein CN, Blanchard JF, Leslie W, Wadja 58 Enders S: Messengers and mediators: Interac- tive steroid-controlled trial. Gut 1993;34:778–
A, Yu BN: The incidence of fracture among tions among lipids, eicosanoids and cytokines. 782.
patients with inflammatory bowel disease. A Am J Clin Nutr 1993;57(suppl):798–800. 74 Gassull MA, Fernandez-Bañares F, Cabré E et
population-based cohort study. Ann Intern 59 Calder PC: n-3 polyunsaturated fatty acids and al: Fat composition may be the clue to explain
Med 2000;133:795–799. immune cell function. Adv Enzyme Regul the primary therapeutic effect of enteral nutri-
43 Chinea B, Rosa A, Ohariz JJ, Ramirez M, Had- 1997;37:197–237. tion in Crohn’s disease. Results of a double-
dick L, Perez C, Torres EA: Osteopenia in 60 Guthy E: Morbus Crohn und Nahrungsfette: blind, randomised, multicenter European
Puerto Ricans with Crohn’s disease. P R Hypothese zur Ätiologie der Enteritis regio- study. Gut 2002;51:164–168.
Health Sci J 2000;19:329–333. nalis. Dtsch Med Wochenschr 1982;107:71– 75 Sanderson P, Calder PC: Dietary fish oil di-
44 Scoon EJ, van Nunnen AB, Wouters RS, Stock- 73. minishes lymphocyte adhesion to macrophage
brugger RW, Russel MG: Osteopenia and os- 61 Guthy E: Ätiologie des Morbus Crohn: Was and endothelial dell monolayers. Immunology
teoporosis in Crohn’s disease: Prevalence in a spricht für Fette als mögliche Ursache. Dtsch 1998;94:79–87.
Dutch population-based cohort. Scand J Gas- Med Wochenschr 1983;108:1729–1733. 76 De Caterina R, Libby P: Control of endothelial
troenterol Suppl 2000;232:43–47. 62 Nordenval B, Brostrom O, Hellers G: Entzünd- leucocyte adhesion molecules by fatty acids.
45 Lee SH, Kim HJ, Yang SK, Kim WH, Joo YS, liche Darmkrankheiten und Nahrungsfette. Lipids 1996;31(suppl):57–63.
Dong SH, Kim BH, Lee JI, Chang YW, Chang Dtsch Med Wochenschr 1982;107:1900. 77 Yeh SL, Chang KY, Huang C, Chen WJ: Effect
R: Decreased trabecular bone mineral density 63 Shoda R, Matsueda K, Yamato S, Umeda N: of n-3 and n-6 fatty acids on plasma eicosanoid
in newly diagnosed inflammatory bowel dis- Epidemiologic analysis of Crohn’s disease in and liver antioxidant enzymes in rats receiving
ease patients in Korea. J Gastroenterol Hepatol Japan: Increased dietary intake of n-6 polyun- total parenteral nutrition. Nutrition 1997;13:
2000;15:512–518. saturated fatty acids and animal protein relates 32–36.
to the increase of Crohn’s disease in Japan. Am 78 Miles EA, Calder PC: Modulation of immune
J Clin Nutr 1996;63:741–745. function by dietary fatty acids. Proc Nutr Soc
1998;57:277–292.

226 Dig Dis 2003;21:220–227 Gassull


79 Enders S: Messengers and mediators: Interac- 90 Sakurai T, Matsui T, Yal T et al: Short-term 102 Roediger WEW, Duncan A, Kapanidis O,
tions among lipids, eicosanoids and cytokines. efficacy of enteral nutrition in the treatment Millard S: Sulphide impairment of butyrate
Am J Clin Nutr 1993;57(suppl):798–800. of active Crohn’s disease: A randomized-con- oxidation in rat colonocytes: A biochemical
80 Calder PC: n-3 polyunsaturated fatty acids and trolled trial comparing nutrient formulas. J basis for ulcerative colitis? Clin Sci 1993;85:
immune cell function. Adv Enzyme Regul Parenter Enteral Nutr 2002,26:98–103. 623–627.
1997;37:197–237. 91 Middleton SJ, Rucker JT, Kirby GA, Riordan 103 Pitcher MCL, Beatty ER, Cummings JH: The
81 Chen LH, Zhao Y: Eicosapentaenoic acid sup- AM, Hunter JO: Long-chain triglycerides re- contribution of sulphate bacteria and 5-ami-
presses lipopolysaccharide-induced tumor ne- duce the efficacy of enteral feeds in patients nosalycilic acid to faecal sulphide in patients
crosis factor-· expression by decreasing activa- with active Crohn’s disease. Clin Nutr 1995; with ulcerative colitis. Gut 2000;46:64–72.
tion of activator protein-1. Am J Clin Nutr 14:229–236. 104 Den Hond E, Hiele M, Evenepoel P, Peeters
2002;75(suppl):366. 92 Ina K, Itoh J, Fukushima K, Kusugami K et M, Ghoos Y, Rutgeerts P: In vivo butyrate
82 Karsten S, Schäfer G, Schauder P: Cytokine al: Resistance of Crohn’s disease T cells to metabolism and colonic permeability in ex-
production and DNA synthesis by human pe- multiple apoptotic signals is associated with tensive ulcerative colitis. Gastroenterology
ripheral lymphocytes in response to palmitic, Bcl-2/Bax mucosal imbalance. J Immunol 1998;115:584–590.
stearic, oleic and linoleic acid. J Cell Physiol 1999;163:1081–1090. 105 Simpson EJ, Chapman MAS, Dawson J, Ber-
1994;161:15–22. 93 Llor X, Pons E, Roca A, Alvarez M et al: The ry D, Macdonald IA, Colen A: In vivo mea-
83 Fürst P, Kuhn KS: Fish oil emulsions: What effects of fish oil, olive oil, oleic acid, and lin- surement of colonic butyrate metabolism in
benefits can they bring? Clin Nutr 2000;19:7– oleic acid on coloreatal neoplastic processes. patients with quiescent ulcerative colitis. Gut
14. Clin Nutr 2003;22:71–79. 2000;46:73–77.
84 Sadeghi S, Wallace FA, Calder PC: Dietary lip- 94 González-Huix F, Fernández Bañares F, Es- 106 Scheppach W, Bartram HP, Richter F, Muell-
ids modify cytokine response to bacterial lipo- teve M, Abad A, Cabré E, Acero D, Figa M, er JG, Greinwald R, Tauschel HD, Gierend
polysaccharide in mice. Immunology 1999;96: Guilera M, Humbert P, De León R, Gassull M, Weber A, Hegemann D, Kubetzko W,
404–410. MA: Enteral versus parenteral nutrition as Rabast U, Schuetz E, Raedsch R, Britsch R,
85 Owen RW, Giacosa A, Hull WE, Haubner R, adjunct therapy in acute ulcerative colitis. Rehmann IH, Otto P, Judmaier G, Press AG,
Spiegelhalder B, Bartsch H: The antioxidant/ Am J Gastroenterol 1993;88:227–232. Woerdehoff D, Mlitz H, Stein J, Schmidt C:
anticancer potential of phenolic compounds 95 Govers MJAP, Gannon NJ, Dunshea FR, Treatment of distal ulcerative colitis with
isolated from olive oil. Eur J Cancer 2000;36: Gibson PR, Muir JG: Wheat bran affects the short-chain fatty acid enemas – A placebo-
1235–1237. site of fermentation of resistant starch and controlled trial. Dig Dis Sci 1996;41:2254–
86 Mangas-Cruz MA, Fernandez A, Albi T, Guin- luminal indexes related to colon cancer risk: 2259.
da A, Relimpio F, Lanzon A, Pereira JL, Ser- A study in pigs. Gut 1999;45:840–847. 107 Fernández Bañares F, Hinojosa J, Gomollón
rera JL, Montilla C, Astorga R, Garcia-Luna 96 Rombeau JL, Kripke SA: Metabolic effects of F, Sánchez-Lombraña JL, Rodrı́guez-Abascal
PP: Effects of minor constituents (non-glycer- short-chain fatty acids. J Parenter Enteral J, Gassull MA, Spanish Group for the Study
ide compounds) of virgin olive oil on plasma Nutr 1990;14(suppl):181–185. of Crohn’s Disease and Ulcerative Colitis:
lipid concentrations in male Wistar rats. Clin 97 Royall D, Wolever TMS, Jeejeebhoy KN: Randomized clinical trial of Plantago ovata
Nutr 2001;20:211–215. Clinical significance of colonic fermentation. (dietary fibre) as compared to mesalamine in
87 Wanten GJ, Geijtenbeek TBH, Raymakers Am J Gastroenterol 1990;85:1307–1312. maintaining remission in ulcerative colitis.
RAP, van Kooyk Y, Roos D, Jansen JBMJ, 98 Scheppach W: Effects of short-chain fatty Am J Gastroenterol 1999;94:427–433.
Naber AHJ: Medium-chain triglyceride-con- acids on gut morphology and function. Gut 108 Inan MS, Rasoulpour RJ, Yin L, Hubbard
taining emulsions increase human neutrophil 1994;35:S35–S38. AK, Rosenberg DW, Giardina C: The lumi-
ß2 integrin expression, adhesion and degranu- 99 Vernia P, Cittadini M, Caprilli R, Torsoli A: nal short-chain fatty acid butyrate modulates
lation. J Parenter Enteral Nutr 2000;24:228– Topical treatment of refractory distal ulcer- NF-ÎB activity in a human colonic epithelial
233. ative colitis with 5-ASA and sodium butyrate. cell line. Gastroenterology 2000;118:724–
88 Andoh A, Takaya H, Araki Y, Tsujikawa T, Dig Dis Sci 1995;40:305–307. 734.
Fujiyama Y, Bamba T: Medium- and long- 100 Chapman MAS, Grahn MF, Boyle MA, Hut- 109 Wächterhaushäuser A, Loitsch SM, Stein J:
chain fatty acids differentially modulate inter- ton M, Rogers J, Williams NS: Butyrate oxi- PPAR-Á is selectively upregulated in Caco-2
leukin-8 secretion in human fetal intestinal epi- dation is impaired in the colonic mucosa of cells by butyrate. Biochem Biophys Res Com-
thelial cells. J Nutr 2000;130:2636–2640. sufferers of quiescent ulcerative colitis. Gut mun 2000;272:380–385.
89 Rodriguez-Palmero M, Kiss S, Fink M, Dem- 1994;35:73–76. 110 Segain JP, Raingeard de la Blétière D, Bour-
melmair H, Szabo I, Koletzko B: Effects of 101 Roediger WEW, Duncan A, Kapanidis O, reille A, Leray V, Gervois N, Rosales C, Fer-
medium-chain triglycerides on metabolism of Millard S: Reducing sulfur compounds of the rier L, Bonnet C, Blottiere HM, Galmiche JP:
linoleic acid in preterm infants. Clin Nutr colon impair colonocyte nutrition: Implica- Butyrate inhibits inflammatory responses
1999;18(suppl 1):207A. tions for ulcerative colitis. Gastroenterology through NFÎB inhibition: Implications for
1993;104:802–809. Crohn’s disease. Gut 2000;47:397–403.

Nutrition and Inflammatory Bowel Disease Dig Dis 2003;21:220–227 227


Review Article

Dig Dis 2003;21:228–236


DOI: 10.1159/000073340

Factors Enhancing Intestinal Adaptation


after Bowel Compensation
D.S. Botsios K.D. Vasiliadis
4th Surgical Clinic, Aristotle University of Thessaloniki, Thessaloniki, Greece

Key Words trition, in a shorter period of time, which reduce the rate
Gut adaptation W Intestinal failure W Growth factors of adverse effects caused by artificial nutrition and im-
prove quality of life.
Copyright © 2003 S. Karger AG, Basel

Abstract
Intestinal failure (IF) refers to the condition in which cer-
tain causes lead to derangements in nutrient absorption Introduction
capacity. Gut adaptation occurs in response to IF and it is
both morphologic and physiologic in nature and can be Intestinal failure (IF) refers to the condition in which
mediated by growth factors and nutrients. Our paper the functioning bowel is insufficient to allow for the
reviews certain trophic growth factors that have impor- absorption of an adequate amount of nutrients. Consider-
tant interactions relevant for intestinal growth, function able progress in the understanding and treatment of IF
and adaptation. Data Source: The literature was re- has been made over the last years. Fleming and Reming-
viewed (data from both animal and human studies) and ton [1] defined IF as ‘the reduction in functioning gut
certain trophic factors that modulate intestinal adapta- mass below the amount necessary for adequate digestion
tion are summarized. The factors reviewed are: epider- and absorption of food’. According to this definition, IF
mal growth factor, insulin-like growth factor I and II, can be present in patients with a normal, yet dysfunction-
transforming growth factor · and ß, neurotensin, inter- al, length of bowel.
leukin-11, glucagon-like peptide-2, keratinocyte growth IF may be divided into two types: the first is character-
factor, human growth hormone, short-chain fatty acids, ized by an absolute reduction in normally functioning gut
and glutamine. Conclusions: Growth factors augment mass (short bowel syndrome – SBS), and the second fea-
intestinal proliferation, diminish programmed apoptosis, tured by an intestine with extensive lesions or functional
and modulate the adaptive process. They also have the insufficiency (intestinal dysfunction). In addition, IF can
potential to improve nutrient absorption in some bowel be complete (total small bowel enterectomy) or partial
disease. The enhancement of gut adaptation may allow (partial resection). The condition can be acute and tempo-
patients to transition of parenteral/enteral to normal nu- rary, as seen with recoverable motility disorders such as

© 2003 S. Karger AG, Basel D. Botsios, Assoc. Prof. Surg.


ABC 0257–2753/03/0213–0228$19.50/0 Kaukasou 64
Fax + 41 61 306 12 34 GR–55133 Thessaloniki (Greece)
E-Mail karger@karger.ch Accessible online at: Tel. +30 2392051560, Fax +30 2310358000
www.karger.com www.karger.com/ddi E-Mail keva@med.auth.gr
ileus and obstruction, or chronic and permanent. Al- not recognized previously. These are problems related to
though a wide spectrum of conditions can be associated (a) intravenous catheter care, (b) supply of micronutrients
with IF, four principal underlying causes can be identified and (c) the occurrence of irreversible hepatic failure.
– these are (a) the SBS, (b) total parenchymal bowel dis-
ease (e.g. Crohn’s disease), (c) motility disorders, such as
visceral myopathy and chronic intestinal obstruction, and Small Bowel Adaptation in Intestinal Failure
(d) small bowel fistulation which leads to premature loss
of enteric content. The major resulting nutritional disor- Where IF is the result of the SBS, it is the hope that
ders are starvation and dehydration, but loss of body mass compensatory mechanisms may reduce dependence on
is frequently made worse by catabolism from associated PN. Several mechanisms are available: (a) Spontaneous
sepsis [2]. adaptation, which results from cellular hyperplasia, vil-
SBS is the best-known form of IF. It usually results lous enlargement, intestinal lengthening and dilatation,
from extensive small bowel resection for the conse- altered motility, increased absorptive capacity and in-
quences of superior mesenteric artery or vein thrombosis creased activity of brush-border enzymes. Alimentary
or volvulus. SBS can also result from repeated resection of secretions, ingested nutrients and hormones stimulate the
small bowel strictures due to Crohn’s disease. In addition, changes. (b) Bowel-lengthening procedures: over the past
SBS is associated with many aftereffects, such as gastric 16 years, Bianchi has pioneered bowel-lengthening proce-
hypersecretion, gallstone formation, renal calculi, liver dures in neonates and infants with SBS [5]. (c) Reserved
disease and metabolic bone disease. Chronic motility dis- small bowel segments: segmental bowel reversal in pa-
orders, such as those characterized, as intestinal pseudo- tients with SBS aims to slow intestinal transit time, there-
obstruction or visceral myopathy, are rare causes of IF. by theoretically enhancing absorption. (d) Intestinal
Regarding survival following massive bowel resection transplantation, and (e) Pharmacological stimulation of
in humans, a review made by Wilmore [3] documented adaptation-growth factors and other modulators.
that in infants with an intact ileocecal valve, none with
small intestinal length !15 cm survived, while, in infants
receiving ileocecal resection, this length is extended to Growth Factors and Other Modulators
40 cm. However, patients with an even shorter length of
remaining intestine can nowadays be successfully weaned A number of specific factors that enhance the prolifera-
from parenteral nutrition (PN), if certain nutrients are tion response of the enterocyte have been identified and
administered in combination. The lower limit of remain- their effect was demonstrated mainly in animal studies.
ing gut length compatible with successful weaning from
enteral feeding is estimated to be ^30 cm in children. In Epidermal Growth Factor (EGF)
adults, on the other hand, the extent of compensatory EGF, extracted from the mucous gland and Brunner’s
hypertrophy is becoming very limited (the lower limit of glands, is a low-molecular-weight polypeptide that is
remaining intestinal length is said to be at least 50–70 cm) known to enhance the growth of the intestinal mucosa. It
[4]. has been shown to be involved in the maturation of a
Recovery of intestinal function can be expected from number of developing tissues. Several studies have dem-
efficient use of certain factors stimulating regeneration onstrated the trophic effects of EGF on intestinal mucosa
and hypertrophy of the remnant intestinal mucosa, in in a variety of experimental models [6–11]. Some of the
addition to prolonged nutritional management with en- effects of exogenous EGF on the gastrointestinal mucosa
teral and parenteral nutritional regimens. It should be are notably similar to the compensatory changes observed
noted that both SBS and IF are associated with the loss of after massive small bowel (SB) resection [12–14]. EGF
the gut barrier function. receptors are present throughout the intestinal tract and
Resolution of IF can occur spontaneously by the pro- located on both basolateral and brush-border mem-
cess of intestinal adaptation, a process that is stimulated branes.
by enteral nutrition, or in the case of intestinal fistula can In experimental studies continuous administration of
be resolved surgically. In those cases where resolution the EGF resulted in significant increases in body weight,
does not take place, recourse has to be made to permanent intestinal weight (SB and colon), SB length, and ileal
PN. The increased use of long-term total parenteral nutri- mucosal mass following massive SB resection [15]. In
tion (TPN) revealed numerous adverse effects that were addition, the mucosal DNA and protein contents in-

Factors Enhancing Intestinal Adaptation Dig Dis 2003;21:228–236 229


after Bowel Compensation
creased after EGF administration, suggesting an augmen- Insulin-Like Growth Factors I and II (IGF-I and II)
tation of the usual mucosal hyperplasia observed after IGF-I and II are growth factors similar in structure to
massive SB resection. insulin, which is primarily involved in the regulation of
Goodlad et al. documented that EGF administration normal growth and development. The major source of cir-
increased colon weight, which resulted from intestinal culating IGF-I and II is the liver, and their effects are
epithelial proliferation. In addition, they observed an more direct than that of growth hormone (GH). Several
increase in pancreatic weight after EGF administration, studies have demonstrated the trophic effects of IGF-I
which raised the possibility that EGF mediates some of its and II on intestinal mucosa in a variety of experimental
trophic effects through increases in pancreatic growth or models [24].
secretion [16]. IGF-I is produced in the liver in response to GH, and
In normal mature intestinal mucosa, administration of leads to an increase in crypt cell production rate, with a
EGF has been reported to increase intestinal weight, lac- consequent increase in villus high and nutrient absorptive
tase-specific activity, net calcium transport [8], and galac- capability [25, 26]. IGF-I enhances DNA and protein syn-
tose and glycine absorption [17]. Similar trophic effects of thesis in intestinal crypt cells in vitro, maintains intestinal
EGF on atrophic mucosa (caused by lack of enteral nutri- integrity, and enhances intestinal mucosal adaptation af-
tion while on TPN [7, 18, 19] or by administration of an ter intestinal resection. Localization of IGF receptors in
elemental diet) have also been reported [10]. Chaet et al. the intestinal epithelium suggests a functional role of
showed in 1994 that administration of EGF after massive IGF-I in intestinal epithelial cell growth and differentia-
SB loss can by of nutritional benefit through its enhance- tion and thus a possible role of IGF-I in gut repair [27].
ment of the normal post-resection intestinal response A recent experimental study demonstrated that admin-
[15]. istration of IGF-I enhanced epithelial restitution after
In a series of studies, Thomson et al. [9, 20–22] showed intestinal mucosal injury. In this model, increased thymi-
that EGF stimulates neomucosal growth on serosal dine uptake, cell migration and increased TGF-ß mRNA
patches created in the rabbit intestine. In their studies, expression were noted in intestinal epithelial cells [27].
maximal stimulation was achieved through intravenous Sigalet and Martin [28] showed that IGF-I could im-
[10] administration of EGF. EGF given enterally does not prove weight gain in a rat model of severe bowel syn-
stimulate cell proliferation in the colon, whereas intrave- drome (SBS). This improvement in weight gain was asso-
nous recombinant EGF reverses the marked intestinal ciated with an increase in nutrient transport at the cellular
hypoplasia characteristically found in TPN-fed rats. In a level and variable increases in villus size. IGF-I has the
study of EGF administration after massive SB resection, greatest effect on weight gain, which should be the guide
Read et al. [23] reported increased intestinal net weight to further studies regarding the mechanisms underlying
and sucrase activity after 7 days of oral EGF administra- effects. IGF-I probably acts directly by improving nu-
tion. Furthermore, EGF upregulates electrolytes and nu- trient absorption at the enterocyte level or by an effect on
trient absorption in the small bowel and thus mediates the adaptation. According to other similar studies [12, 29,
absorptive capacities of various nutrients. 30], it is assumed that an increase in villus height accom-
Read et al. [23], in an experimental study, adminis- panies any significant improvement in nutrient absorp-
trated EGF orally and documented that there were no tion characteristics. It is possible that IGF-I is acting
positive synergistic effects of EGF and intestinal resec- through endocrine pathways and only indirectly affecting
tion, nor did EGF fully compensate for the lack of luminal nutrient transport characteristics.
contents. Because post-resection adaptation may begin Although markedly beneficial effects of IGF-I and II
immediately after resection and continue for several have been observed in animal studies, its clinical effects
weeks [12], further studies are needed to evaluate EGF’s on protein metabolism and gut integrity have not yet been
effect on the residual SB after massive resection, when clearly demonstrated and further analysis is required.
administrated throughout the period of expected adapta-
tion. Although EGF had multiple trophic effects in experi- Transforming Growth Factor (TGF)-· and ß
mental models, EGF’s safety and utility for human thera- TGF-· is a 5.5-kDa protein containing 50-amino-acid
py are presently under research. residues that share 35% structural homology with EGF, a
common membrane receptor with a similar spectrum of
biological activity [31]. TGF-· has been reported to stim-
ulate proliferation of a wide range of epidermal and epi-

230 Dig Dis 2003;21:228–236 Botsios/Vasiliadis


thelial cells and is important in intestinal cell proliferation Neurotensin
[32]. In a recent study, Falcone et al. [33] have shown that Neurotensin is a tridecapeptide originally isolated
in a mouse model of SBS, normal adaptation resulted in from bovine hypothalamus and subsequently found to be
30% increase in ileal TGF-· mRNA levels by post-opera- widely distributed in the gastrointestinal tract. Neuroten-
tive day 3. Additionally, it has been shown that exogenous sin-like immunoreactivity has been localized to a specific
TGF-· given within the first 3 days significantly en- mucosal endocrine cell type (N-cell) that is found in high-
hanced enterocyte proliferation and stimulated intestinal est density in the ileum [42]. Neurotensin-like immunore-
adaptation. activity in plasma increases two- to threefold after meals
TGFs were isolated for the first time in 1978 from cul- [43]. Thus, potential physiological actions of neurotensin
ture medium conditioned by Rous sarcoma virus-trans- on gastric and pancreatic secretion, gastrointestinal motil-
formed fibroblasts [34] and include two proteins, now ity, and gastrointestinal growth may be mediated by a hor-
designated TGF-· and TGF-ß. Both proteins have a dif- monal mechanism with release of neurotensin from N
ferent structure and a different spectrum of biological cells of the intestinal mucosa, as well as paracrine and
activity. Since its isolation from transformed cell lines, neurocrine mechanisms involving local release from N
TGF-· has been identified in a wide spectrum of epithe- cells and nerve fibers.
lial cells, including gastrointestinal epithelium [35]. Neurotensin increases contractile activity of the colon
TGF-· has recently been found to promote proliferation [44] and disrupts the interdigestive myoelectric complex
in established cell lines and to exert a trophic effect on of the small intestine [45]. Neurotensin also increases
intact gastric, intestinal and colonic mucosa. TGF-· is intestinal blood flow and capillary permeability [42] and
considered important in epithelial cell proliferation in stimulates net fluid secretion by small intestine in vitro
digestive tissues during the developmental period [36], in [47].
the promotion of cell migration and motivation of fluid Wood et al. [48] showed that neurotensin had trophic
and electrolyte transport in the enterocyte [37], in the effects on the small intestine in rats. Neurotensin pro-
inhibition of gastric acid secretion and promotion of gas- duced increases in weight, DNA and protein content of
tric ulcer healing [38] and gastric mucosal repair [39] and the small intestine. There were also increases in total con-
in intestinal mucosal repair [40] following acute epithelial tent of three brush-border enzymes measured in this
injury. study, namely sucrase, maltase and leucine aminopepti-
Several lines of evidence have implicated TGF-· as a dase. The potential action of neurotensin on small intesti-
potential modulator of intestinal adaptation in an animal nal growth is not due solely to an endocrine mechanism.
model of SBS. High doses of neurotensin may result in high concentra-
In a recent study the potential effects of exogenous tions within the intestine, similar to those expected after
TGF-· as a gut-trophic agent after small bowel resection local release of neurotensin from endocrine cells or neu-
in the rat was investigated. It was shown that exogenous rons. By this reasoning, neurotensin could exert paracrine
TGF-· given at high doses (75 Ìg/kg/day) stimulated or neurocrine effects on intestinal growth.
intestinal adaptation in the remaining gut. This was evi- It is not yet established whether neurotensin acts
dent from increased bowel and mucosal weights, mucosal directly on the small intestine or indirectly through effects
DNA and protein, villus height and crypt depth. Exoge- on other growth factors. Neurotensin has been reported to
nous TGF-· has been showed to accelerate the prolifera- stimulate release of thyrotropin in rats [49] and insulin
tion of intestinal cells. In addition, the administration of and glucagon in dogs [50]. Other similar studies showed
TGF-· reduced cell death via apoptosis. Both increased that neurotensin prevents atrophy of the intestinal muco-
cell production and decreased cell death may indicate an sa associated with TPN and promotes the release of secre-
adaptive mechanism to increase enterocyte mass follow- tory IgA and IgM into the bile [51].
ing TGF-· administration [41]. These observations sug-
gest that TGF-· may have clinical utility for the patient Interleukin-11 (IL-11)
with SBS. IL-11, a 19-kDa bone marrow-derived growth factor, is
TGF-ß inhibits cell proliferation in vitro and in vivo. a multifunctional peptide that uses the gp130 receptor
No clinical studies have been performed to demonstrate common subunit for receptor function [52]. It was first
clinical efficacy. cloned in 1990 from an immortalized primate bone mar-
row stromal cell line, PU-34 [53]. Its hematopoietic
effects include increases in peripheral blood neutrophils

Factors Enhancing Intestinal Adaptation Dig Dis 2003;21:228–236 231


after Bowel Compensation
and platelets, and stimulation of erythroid elements, me- expression of KGF receptor and KGF messenger RNA
gakaryocytes, and B-cell differentiation in vivo and in (mRNA) has been detected throughout the gastrointesti-
vitro [54]. IL-11 also improves survival rate and decreases nal tract [69]. Recent studies support a role for KGF in
bacterial translocation in burned mice [55]. Recent stud- tissue repair [70, 71]. In inflammatory bowel disease,
ies suggest that IL-11 enhances a small intestine mucosal increased expression of KGF has been described in the
mass after massive small bowel resection [56, 57]. How- mucosa, suggesting a potential role for KGF in mediating
ever, these studies did not determine if there were changes mucosal repair mechanisms [72, 73].
in the function at the remnant small intestine. The current The role for KGF as an endogenous mediator of
findings show that exogenous administration of IL-11 growth and differentiation in the gastrointestinal tract is
increases mucosal mass, as noted by enhanced DNA con- supported by the isolation of mRNA for KGF and its
tent, in a dose-dependent manner. These data corroborate receptor in both the small and large intestine of rodents
the results of previously described in vivo studies and fur- [69, 74].
ther strengthen the assertion that IL-11 has a trophic Recently it has been demonstrated that exogenous full-
effect on intestinal epithelial cells. The systemic infusion length KGF significantly reduces the severity and extent
IL-11 had significantly enhanced galactose and glycine of mucosal injury in different experimental models of
absorption. [58]. colitis in rats and mice [75, 76]. KGF induces prolifera-
tion of colonic epithelium (or alternatively decreased
Glucagon-Like Peptide-2 (GLP-2) apoptosis) and quantitatively increases the mucin content
GLP-2 is a 33-amino-acid peptide known to enhance [75, 77]. KGF has also been demonstrated to increase the
mucosal integrity in normal intestine and in intestine that number of goblet cells as well as mucin production
has undergone adaptation after massive small bowel re- throughout the gastrointestinal tract of rats and mice [69,
section (MSBR) [59]. GLP-2 is liberated from the car- 75, 76]. Housley et al. [69] showed that KGF enhanced
boxy-terminus of proglucagon in small and large intestine the growth of the intestinal mucosa. An animal study by
L cells by the action of tissue-specific proteases. GLP-2 Estivarez et al. [78] showed that KGF inhibits atrophy of
belongs to a specific class of compounds referred to as pro- the duodenal and ileal mucosa induced by fasting. John-
glucagon-derived peptides (PGDPs), products of the post- son et al. [79] evaluated the effects of KGF on intestinal
translational processing of proglucagon. A role for PGDPs adaptation after resection of 85% of the small intestine. It
as intestinal hyperplasia and hypertrophy were noted in 2 was concluded that KGF enhances gut growth, differenti-
patients with PGDP-secreting tumors [60, 61]. It was ation and gene regulation during adaptation in rat small
shown subsequently that nude mice carrying subcuta- intestine after massive resection. The clinical relevance
neous PGDP-producing tumors had a significant increase was that KGF might be beneficial in the management of
in small intestinal weight [62]. The PGDP responsible for patients undergoing massive intestinal resection. In a
the intestinal epithelial proliferation in these mice was recent experimental study in mice, Yang et al. [80]
identified as GLP-2. Subsequent studies confirmed that showed that KGF administration improved epithelial ab-
the naturally occurring form of GLP-2 induces growth in sorptive function and stimulated intestinal proliferation
normal intestine [63–65]. after SBS. They concluded that KGF improved intestinal
Drucker et al. [66] further showed that a synthetic, pro- adaptation after small bowel syndrome and may have
tease-resistant form of GLP-2 was a potent agent for clinical applicability.
increasing small and large intestine mass in normal rats.
Findings in other similar studies showed that GLP-2 Human Growth Hormone (hGH)
increases mucosal mass and absorptive function in intes- hGH belongs to anabolic hormones. In 1974, Wilmore
tine after MSBR [67]. In experiments on massive bowel et al. [81] first reported the clinical effects of hGH. They
resection or ischemia/reperfusion injury, administration reported that administration of hGH to burn patients
of GLP-2 significantly increased DNA and protein con- resulted in an improvement of nitrogen and potassium
tent of the intestinal mucosa [59]. However, such effects balance and enhanced wound healing. Based on initial
have not been confirmed in humans. observations showing GH can increase nutrient and elec-
trolyte absorption in hypophysectemized animals [82],
Keratinocyte Growth Factor (KGF) Byrne et al. [83] reported in a nonrandomized, open study
KGF, synthesized and secreted by stromal fibroblasts, that GH and dietary modification improved nutrient
is expressed predominantly in the dermis [68]. However, absorption in patients with small bowel syndrome. Subse-

232 Dig Dis 2003;21:228–236 Botsios/Vasiliadis


quent studies using more rigorous techniques have shown SCFAs are a logical mediator of trophic gastrointestinal
no effect on nutrient absorption with GH treatment in hormones (i.e., PGDPs) and ileal adaptation because
human patients [84, 85], whereas a variety of animal stud- they are produced distally within the gastrointestinal
ies have shown conflicting results [86–88]. tract in response to malabsorbed substrate. Many studies
Kissmeyer-Nielsen et al. [89] administrated GH to have established a strong relation between cellular prolif-
TPN patients with gastrointestinal diseases and reported eration and elevated amounts of proglucagon mRNA and
its protein-sparing effect, a reduced incidence of infec- PGDPs [97, 98, 102–104]. Taylor et al. [100] reported
tion, and a decrease in malaise. Concerning the proliferat- that after intestinal resection, proglucagon expression in-
ing effect of GH on intestinal mucosa, a number of animal creased threefold, peaking 2 days after surgery and de-
studies have demonstrated its beneficial effect on struc- clining thereafter. In a recent study it was shown that
tural maintenance, adaptation and wound healing [90]. If SCFA-supplemented TPN increased proglucagon mRNA
GH does affect the adaptive process, it is likely this effect abundance [95].
is mediated through IGF-I [24]. Tappenden et al. [105] showed that SCFA-supple-
Sigalet and Martin [28], in their experimental study, mented TPN rapidly upregulated jejunal GLUT-2
documented that GH administration improved weight mRNA (glucose transporter 2) and ileal GLUT-2 abun-
gain which resulted as a consequence in nutrient absorp- dance and ileal proglucagon mRNA. Within the intestinal
tion characteristics [12, 29, 30]. Their experimental evi- resection model, SCFAs may exert their trophic effect by
dence raises the hypothesis that GH is acting indirectly extending the duration of increased proglucagon expres-
through endocrine pathways, enhancing nutrient trans- sion.
port. Gu et al. showed, in a rat model, that GH enhanced
small bowel adaptation. This result indicated that appli- Glutamine (Gln)
cation of GH could alleviate mucosal atrophy of the rem- Gln is the most abundant nonessential amino acid in
nant small intestine caused by PN and improve intestinal the body. The most important function of Gln lies in its
adaptation significantly in short bowel rats [91]. metabolic role as a nitrogen carrier between major organs.
Gln prevents the degeneration of muscle protein, main-
Short-Chain Fatty Acids (SCFAs) tains body protein levels at the time of surgical stress and
SCFAs are formed in the gastrointestinal tract of mam- is involved in the maintenance of intestinal epithelial cell
mals by microbial fermentation of carbohydrates, and function as an energy source. A number of studies suggest
readily absorbed by intestinal and colonic mucosa, and that Gln is conditionally essential for gut metabolism,
are trophic to intestinal mucosa. Acetate, propinate and structure and function in the stressed animal [106, 107].
butyrate account for about 85% of SCFAs and are pro- Gln has therefore been postulated to be of crucial impor-
duced in a nearly constant molar ratio of 60:25:15 [92]. tance for the integrity and growth of small intestinal
One week of SCFA supplementation has been shown to mucosa [108, 109]. Wiren et al. [110] were not able to
retard TPN-associated atrophy in rats with intact bowels show better adaptation using Gln supplementation to the
[93] and after 80% intestinal resection [94]. Recent stud- diet compared to chow feeding, whereas several studies
ies have reported that SCFA-supplemented TPN en- confirmed that a dipeptive alanyl-Gln-enriched TPN so-
hanced both structural [95] and functional [96] adapta- lution could increase the growth of the intestinal mucosa
tion to 80% intestinal resection as early as 3 days after in animals with intestinal atrophy due to TPN [111] and
surgery. massive bowel resection [112]. Dipeptive alanyl-Gln-
SCFAs may influence small intestinal mucosal prolif- enriched TPN solution could also have a protective effect
eration by stimulating secretion of PGDPs. Intestinal on the mucus covering the surface of the intestinal muco-
proglucagon mRNA abundance and plasma concentra- sa [113].
tions of PGDPs are strongly correlated with cellular pro- For the clinical efficacy of Gln, several studies have
liferation during intestinal adaptation [97–100]. Precise been performed demonstrating conflicting results [114–
physiologic roles for each of the PGDPs continue to be 116]. Specifically, concerning the effect of Gln on intesti-
elucidated, however, it was shown recently that GLP-2 nal function, Wilmore et al. [117] treated 300 short bowel
modulates basolateral membrane glucose transport in patients with a combination of GH, Gln and high carbo-
rats [101]. Other studies showed that SCFA-supple- hydrate diet, and reported that 60% of the patients could
mented TPN increases proglucagon gene expression 3 be weaned from TPN, in 30% TPN could be reduced, and
and 7 days after intestinal resection [95]. Physiologically, the remaining 10% still required the same quantity of

Factors Enhancing Intestinal Adaptation Dig Dis 2003;21:228–236 233


after Bowel Compensation
TPN. This result shows the importance of such a combi- Conclusions
nation therapy in the adaptive process of SBS.
On the contrary, Scolapio et al. [84] reported no Growth factors augment intestinal proliferation, di-
improvements in small bowel morphology (villus height, minish programmed apoptosis, and modulate the adap-
crypt depth), stool losses and macronutrient absorption in tive process. They also have the potential to improve
response to active treatment with GH, Gln and high-car- nutrient absorption in some bowel diseases. The enhance-
bohydrate, low-fat diet when compared with placebo car- ment of gut adaptation may allow the patient’s transition
bohydrate in a randomized, crossover study in 8 patients from parenteral/enteral to normal nutrition in a shorter
with well-established SBS. Additionally, Mandir and period of time, which reduces the rate of adverse effects
Goodlad [118] also found there was no effect of Gln sup- caused by artificial nutrition and improves quality of life.
plementation on mitotic activity in the small intestine. These data confirm that gut-trophic agent administration
Alavi et al. [119] have also demonstrated that enteral may be of benefit to patients with IF during adaptation. In
Gln does not promote adaptation either alone or in com- addition, bowel adaptation enhancement could be useful
bination with hepatocyte growth factor in resected rats. in the therapeutic management of patients with malab-
However, further clinical studies need to be conducted to sorption syndromes. Although beneficial effects of these
confirm the efficacy of Gln-enriched TPN formulas in the trophic agents have been observed in human and animal
treatment of intestinal failure. studies, further research and analysis is required for their
clinical applicability.

References

1 Fleming CR, Remington M: Intestinal failure. 11 Ulshen MH, Lyn-Cook LE, Raasch RH: Effects 20 Thompson JS, Saxena SK, Sharp JG: Effect of
Clin Surg Int 1981;2:219. of intraluminal epidermal growth factor on the duration of infusion of urogastrone on in-
2 Irving M: Intestinal failure. J Gastroenterol mucosal proliferation in the small intestine of testinal regeneration in rabbits. Cell Tissue Ki-
Hepatol 2000;15:G26–G29. adults rats. Gastroenterology 1986;91:1134– net 1989;22:303–309.
3 Wilmore DW: Factor correlating with a suc- 1140. 21 Thompson JS, Sharp JG, Saxena SK: Stimula-
cessful outcome following extensive intestinal 12 Dowling RH, Booth CC: Structural and func- tion of neomucosal growth by systemic urogas-
resection in newborn infants. J Pediatr 1972; tional changes following small intestinal resec- trone. J Surg Res 1987;42:402–410.
80:88–95. tion in the rat. Clin Sci 1967;32:139–149. 22 Thomson JS: Epidermal growth factor and the
4 Okada A: Signaling factors for gut adaptation. 13 Hanson WR: Proliferation and morphological short bowel syndrome. J Parenteral Enteral
Nestle Nutr Workshop Ser Clin Perform Pro- adaptation of the intestine to experimental re- Nutr 1999;23:S113–S116.
gramme 2002;7:169–178, discussion 178–181. section. Scand J Gastroenterol 1982;17:11–20. 23 Read LC, Ford WDA, Filsell OH et al: Is orally-
5 Jeppesen PB, Mortensen PB: Intestinal failure 14 Booth CC, Evans KT, Menzies T et al: Intesti- derived epidermal growth factor beneficial fol-
defined by measurements of intestinal energy nal hypertrophy following partial resection of lowing premature birth or intestinal resection?
and wet weight absorption. Gut 2000;46:701. the small bowel in the rat. Br J Surg 1959;46: Endocr Exp 1986;20:199–207.
6 Al-Nafussi Al, Wright NA: The effect of epider- 403–410. 24 Le Roith D: Insulin-like growth factors. Mass
mal growth factor on cell proliferation of the 15 Goodlad RA, Savage AP, Lenton W et al: Does Med Soc 1997;336:633–639.
gastrointestinal mucosa in rodents. Virchows resection enhance the response of the intestine 25 Lemmey AB, Matine AA, Read LC et al: IGF-1
Arch B Cell Pathol 1982;40:63–69. to urogastrone-epidermal growth factor in the and the truncated analogue des-(1–3) IGF-1
7 Goodlad RA, Wilson TJG, Lenton W et al: rat? Clin Sci 1988;75:121–126. enhance growth in rats after gut resection. Am J
Urogastrone-epidermal growth factor is trophic 16 Schwartz MZ, Storazuk RB: Influence of epi- Physiol 1991;260:E213–E219.
to the epithelium of parenterally fed rats. Ex- dermal growth factor on intestinal function in 26 Steeb CB, Trahair JF, Tomas FM et al: Pro-
perientia 1985;41:1161–163. the rat: Comparison of systemic infusion ver- longed administration of IGF peptides en-
8 Oka Y, Ghishan FK, Greene HL et al: Effect of sus luminal perfusion. Am J Surg 1988;155:18– hances growth of gastrointestinal tissues in nor-
mouse epidermal growth factor/urogastrone on 22. mal rats. Am J Physiol 1993;266:G1090–
the functional maturation of rat intestine. En- 17 Goodlad RA, Raja KB, Peters TJ et al: Effects G1098.
docrinology 1983;112:940–944. of urogastrone-epidermal growth factor on in- 27 Chen K, Inoue M, Wasa M, Azuma T, Fukuza-
9 Thomson JS, Saxena SK, Greaton C et al: The testinal brush border enzymes and mitotic ac- wa M, Kamata S, Okada A: Insulin-like growth
effect of the route of delivery of urogastrone on tivity. Gut 1991;32:994–998. factor-I stimulates intestinal epithelial cell pro-
intestinal regeneration. Surgery 1989;106:45– 18 Goodlad RA, Wilson TJG, Lenton W et al: liferation and migration. Surg Forum 1997;48:
51. Intravenous but not intragastric urogastrone- 716–718.
10 Tsujikawa T, Bamba T, Hosada S: The trophic EGF to the intestine of parenterally fed rats. 28 Sigalet DL, Martin GR: Hormonal therapy for
effect of epidermal growth factor on morpho- Gut 1987;28:573–582. short bowel syndrome. J Pediatr Surg 2000;35:
logical changes and polyamine metabolism in 19 Chaet MS, Arya G, Ziegler MM, Warner BW: 360–364.
the small intestine of rats. Gastroenterol Jpn Epidermal growth factor enhances intestinal 29 Williamson R: Intestinal adaptation: Structur-
1990;25:328–334. adaptation after massive small bowel resection. al, functional and cytokinetic changes. N Engl J
J Pediatr Surg 1994;29:1035–1039. Med 1978;298:144, 1393–1402, 1450.
30 Thomson JS: Can the intestine adapt to a
changing environment? Gastroenterology
1997;113:1402–1412.

234 Dig Dis 2003;21:228–236 Botsios/Vasiliadis


31 Derynck R: Transforming growth factor-·. Cell 48 Wood JG, Hoang HD, Bussjaeger LJ, Solomon 64 Tsai C, Hill M, Asa SL et al: Intestinal growth
1988;54:593. TE: Neurotensin stimulates growth of small promoting properties of glucagon-like peptide-
32 Alison MR, Sarraf CE: The role of growth fac- intestine in rats. Am J Physiol 1988;255:813– 2 in mice. Am J Physiol 1997;273:E77–E84.
tors in intestinal cell proliferation. Cell Biol Int 817. 65 Fischer KD, Dhanvantari S, Drucker DJ et al:
1994;18:1. 49 Folkers K, Chang D, Humphries J, Carraway Intestinal growth is associated with elevated
33 Falcone RA, Stern LE, Kemp CJ, Erwin CR, R, Leeman SE, Bowers CY: Synthesis and ac- levels of glucagon-like peptide-2 in diabetic
Warner BW: Intestinal adaptation occurs inde- tivities of neurotensin and its acid and amine rats. Am J Physiol 1997;273:E815–E820.
pendent of transforming growth factor-·. J Pe- analogs: Possible natural occurrence of Gln4- 66 Drucker DJ, DeForest L, Brubaker PL: Intesti-
diatr Surg 2000;35:365. neurotensin. Proc Natl Acad Sci USA 1976;73: nal response to growth factors administered
34 Delarco JE, Torado GJ: Growth factors from 3833–3837 alone or in combination with human [Gly2]glu-
murine sarcoma virus-transformed cells. Proc 50 Ukai M, Inoue I, Itatsu T: Effect of somatosta- cagon-like peptide 2. Am J Physiol 1997;273:
Natl Acad Sci USA 1978;75:4001. tin on neurotensin-induced glucagon release G1252–G1262.
35 Malden LT, Novak U, Burgess AW: Expression and hyperglycemia. Endocrinology 1977;100: 67 Kato Y, Alavi K, Yu D et al: Glucagon-like
of transforming growth factor-· messenger in 1284–1286. peptide-2 enhances intestinal function follow-
the normal and neoplastic gastrointestinal 51 Burke D, Alverdy J, Aoys E, Agarwal A, Moss ing massive small bowel resection. Gastroen-
tract. Int J Cancer 1989;43:380. G: The effect of neurotension and epidermal terology 1998;114:A1152.
36 Hormi K, Lehy T: Transforming growth factor- growth factor on TPN induced IgA deficiency. 68 Aaronson SA, Bottaro DP, Miki T, Ron D,
· in vivo stimulates epithelial cell proliferation Surg Forum 1990;41:70–72. Finch PW, Fleming TP et al: Keratinocyte
in digestive tissues of suckling rats. Gut 1996; 52 Du XX, Williams DA: Interleukin-11: Review growth factor. A fibroblast growth factor family
39:532. of molecular, cell biology and clinical use. member with unusual target cell specificity.
37 Barnard JA, Polk WH, Moses HL, Coffey RJ: Blood 1997;89:3897–3908. Ann NY Acad Sci 1991;638:62–77.
Production of transforming growth factor-· by 53 Du XX, Williams DA: Interleukin-11: A multi- 69 Housley RM, Morris CF, Boyle W, Ring B,
normal small intestine. Am J Physiol 1991;261: functional growth factor derived from the he- Biltz R, Tarpley JE et al: Keratinocyte growth
C994. matopoietic microenvironment. Blood 1994; factor induces proliferation of hepatocytes and
38 Konturek SJ, Brzozowski T, Maka J, Dembins- 83:2023–2030. epithelial cells throughout the rat gastrointesti-
ki A, Slomiany A, Slomiany BL: Transforming 54 Du XX, Neben T, Goldman S: Effects of nal tract. J Clin Invest 1994;94:1764–1777.
growth factor-· and epidermal growth factor in recombinant human interleukin-11 on hemato- 70 Werner S, Peters KG, Longaker MT, Fuller-
protection and healing of gastric mucosal inju- poietic reconstitution in transplant mice: Ac- Pace F, Banda MJ, Williams LT: Large induc-
ry. Scand J Gastroenterol 1992;27:649. celeration of recovery of peripheral blood neu- tion of keratinocyte growth factor expression in
39 Romano M, Polk WH, Awad JA, Arteaga CL, trophils and platelets. Blood 1993;81:27–34. the dermis during wound healing. Proc Natl
Nanney LB, Wargovich MJ, Kraus ER, Boland 55 Schindel D, Maze R, Liu, Q et al: Interleukin- Acad Sci USA 1992;89:6896–6900.
CR, Coffey RJ: Transforming growth factor-· 11 improves survival and reduces bacterial 71 Staiano-Coico L, Krueger JG, Rubin JS, D’limi
protection against drug-induced injury to the translocation and bone marrow suppression in S, Vallat VP, Valentino L et al: Human kerati-
rat gastric mucosa in vivo. J Clin Invest 1992; burned mice. J Pediatr Surg 1997;32:312–315. nocyte growth factor effects in a porcine model
90:2409. 56 Liu Q, Du XX, Schindel DT et al: Trophic of epidermal wound healing. J Exp Med 1993;
40 Dignass AU, Stow JL, Babyatsky MW: Acute effects of interleukin-11 in rats with experi- 178:865–878.
epithelial injury in the rat small intestine in mental short bowel syndrome. J Pediatr Surg 72 Brauchle M, Madlener M, Wagner AD, Anger-
vivo is associated with expanded expression of 1996;31:1047–1051. meyer K, Lauer U, Hofschneider PH et al: Ker-
transforming growth factor-· and ß. Gut 1996; 57 Fiore NF, Ledniczky G, Liu Q et al: Compari- atinocyte growth factor is highly over expressed
38:291. son of interleukin-11 and epidermal growth in inflammatory bowel disease. Am J Pathol
41 Sukhotnik I, Yakirevich E, Coran AG, Siplo- factor on residual small intestine after massive 1996;149:521–529.
vich L, Krausz M, Hirsh M, Sabo E, Shiloni E: small bowel resection. J Pediatr Surg 1998;33: 73 Finch PW, Pricolo V, Wu A, Finkelstein SD:
Effect of transforming growth factor-· on intes- 24–29. Increased expression of keratinocyte growth
tinal adaptation in a rat model of short bowel 58 Alavi K, Prasad R, Lundgren K, Schwartz MZ: factor messenger RNA associated with inflam-
syndrome. J Surg Res 2002;108:235–242. Interleukin-11 enhances small intestine ab- matory bowel disease. Gastroenterology 1996;
42 Helmstaedter V, Taugner CH, Feurle GE, sorptive function and mucosal mass after intes- 110:441–451.
Forssmann WG: Localization of neurotensin- tinal adaptation. J Pediatr Surg 2000;35:371– 74 Finch PW, Rubin JS, Miki T, Ron D, Aaronson
immunoreactive cells in the small intestine of 374. SA: Human KGF is FGF-related with proper-
man and various mammals. Histochemistry 59 Prasad R, Alavi K, Schwartz MZ: Glucagon- ties of a paracrine effector of epithelial cell
1977;53:35–41. like peptide-2 analogue enhances intestinal growth. Science 1989;245:752–755.
43 Holst-Pedersen J, Fahrenkrug J: Neurotensin- mucosal mass after ischemia and reperfusion. J 75 Zeeh JM, Proccacino F, Hoffmann P, Auker-
like immunoreactivities in human plasma: Pediatr Surg 2000;35:357–359. man SL, McRoberts JA, Soltani S et al: Kerati-
Feeding responses and metabolism. Peptides 60 Gleeson MH, Bloom SR, Polak JM et al: Endo- nocyte growth factor ameliorates mucosal inju-
1986;7:15–20. crine tumour in kidney affecting small bowel ry in an experimental model of colitis in rats.
44 Thor K, Rosell S: Neurotensin increases co- structure, motility and absorptive function. Gastroenterology 1996;110:1077–1083.
lonic motility. Gastroenterology 1986;90:27– Gut 1971;12:773–782. 76 Egger B, Proccacino F, Sarosi I, Tolmos J,
31. 61 Stevens FM, Flanagan RW, O’Gorman D et al: Büchler MW, Eysselein VE: Keratinocyte
45 Andersson S, Rosell S, Hjelmquist V, Chang D, Glucagonoma syndrome demonstrating giant growth factor ameliorates dextran sodium sul-
Folkers K: Inhibition of gastric and intestinal duodenal villi. Gut 1984;25:784–791. fate colitis in mice. Dig Dis Sci 1999;44:836–
motor activity in dogs by [Gln4]-neurotensin. 62 Drucker DJ, Ehrlich P, Asa SL et al: Induction 844.
Acta Physiol Scand 1977;100:231–235. of intestinal epithelial proliferation by gluca- 77 Egger B, Tolmos J, Proccacino F, Sarosi I,
46 Harper SL, Barrowman JA, Kvietys PR, Gran- gon-like peptide 2. Proc Natl Acad Sci USA Friess H, Büchler MW et al: Keratinocyte
ger DN: Effect of neurotensin on intestinal cap- 1996;93:7911–7916. growth factor promotes healing of left sided
illary permeability and blood flow. Am J Physi- 63 Brubaker PL, Izzo A, Hill M et al: Intestinal colon anastomoses. Am J Surg 1998;176:18–
ol 1984;247:G161–G166. function in mice with small bowel growth in- 24.
47 Mitchenere P, Adrian TE, Kirk RM, Bloom duced by glucagon-like peptide-2. Am J Physiol
SR: Effect of gut regulatory peptides on intesti- 1997;272:E1050–E1058.
nal luminal fluid in the rat. Life Sci 1981;29:
1563–1570.

Factors Enhancing Intestinal Adaptation Dig Dis 2003;21:228–236 235


after Bowel Compensation
78 Estivarez CF, Jonas CR, Gu LH, Diaz EE, Wal- 93 Koruda MJ, Rolandeli RH, Bliss DZ, Hast- 107 Winmueller HG, Spaeth AE: Identification of
lace TM, Pascal RR, Farrell CL, Ziegler TR: ings J, Rombeau JL, Settle RG: Parenteral ketone bodies and glutamine as the major
Gut-trophic effects of keratinocyte growth fac- nutrition supplemented with short-chain fat- respiratory fuel in vivo for post-absorptive rat
tor in rat small intestine and colon during ty acids: Effects on the small-bowel mucosa in small intestine. J Biol Chem 1978;1078:69–
enteral refeeding. J Parenter Enteral Nutr normal rats. Am J Clin Nutr 1990;51:685– 76.
1998;22:259–267. 689. 108 Wiren M, Permert J, Skullman S, Wang F,
79 Johnson WF, DiPalma CR, Ziegler TR, Scully 94 Koruda MJ, Rolandeli RH, Settle RG, Zim- Larsson J: Intraluminal glutamine refeeding
S, Farrell CL: Keratinocyte growth factor en- maro DM, Rombeau JL: Effect of parenteral supports mucosal growth in rat jejunum.
hances early gut adaptation in a rat model of nutrition supplemented with short chain fatty Transplant Proc 1994;26:1460–1463.
short bowel syndrome. Vet Surg 2000;29:17– acids on adaptation to massive small bowel 109 Barber AE, Jones WG, Minei JP et al: Gluta-
27. resection. Gastroenterology 1988;95:715– mine or fiber supplementation of a defined
80 Yang H, Wildhaber BE, Teitelbaum DH: Har- 720. formula diet: Impact on bacterial transloca-
ry M. Vars Research Award: Keratinocyte 95 Tappenden KA, Thomson ABR, Wild GE, tion, tissue composition and response to en-
growth factor improves epithelial function af- McBurney MI: Short-chain fatty acids in- dotoxin. J Parenter Enteral Nutr 1990;14:
ter massive small bowel resection. J Parenter crease proglucagon and ornithine decarboxyl- 335–343.
Enteral Nutr 2003;27:198–206. ase messenger RNAs following intestinal re- 110 Wiren M, Magnusson KE, Larsson J: Enteral
81 Wilmore DW, Moylan JA Jr, Bristow BF, Ma- section in rats. J Parenter Enteral Nutr 1996; glutamine increases growth and absorptive
son AD Jr, Pruitt BA Jr: Anabolic effects of 20:357–362. capacity of intestinal mucosa in the malnour-
human growth hormone and high caloric feed- 96 Tappenden KA, Thomson ABR, Wild GE, ished rat. Scand J Gastroenterol 1995;30:
ings following thermal injury. Surg Gynecol McBurney MI: Short-chain fatty acid supple- 146–152.
Obstet 1974;138:875–884. mented total parenteral nutrition enhances 111 Tamada H, Nezu R, Imamura I, Takagi Y,
82 Mainoya JR: Influence of ovine growth hor- intestinal function following massive small Okada A: The dipeptide alanyl glutamine pre-
mone on water and NaCl absorption by the rat bowel resection. Gastroenterology 1997;112: vents intestinal mucosal atrophy in parenter-
proximal jejunum and distal ileum. Comp Bio- 792–802. ally fed rats. J Parenter Enteral Nutr 1992;16:
chem Physiol 1982;71A:477–479. 97 Bloom SR, Polak JM: The hormonal pattern 110–116.
83 Byrne TA, Persinger RL, Young LS et al: A new of intestinal adaptation: A major role for ente- 112 Tamada H, Nezu R, Matsuo Y, Imamura I,
treatment for patients with short bowel syn- roglucagon. Scand J Gastroenterol Suppl Takagi Y, Okada A: Alanyl glutamine-en-
drome. Ann Surg 1995;222:243–255. 1982;14:93–103. riched total parenteral nutrition restores in-
84 Scolapio JS, Calilleri M, Gleming CR et al: 98 Sagor GR, Ghatei MA, Al-Mukhtar MYT, testinal adaptation after either proximal or
Effect of growth hormone, glutamine and diet Wright NA, Bloom SR: Evidence for a hu- distal massive resection in rats. J Parenter
on adaptation in short bowel syndrome: A ran- moral mechanism after small intestinal resec- Enteral Nutr 1993;17:236–242.
domized, controlled study. Gastroenterology tion: exclusion of gastrin but not enterogluca- 113 Khan J, Liboshi Y, Cui L, Wasa M, Sando K,
1997;113:1074–1081. gon. Gastroenterology 1983;84:902–906. Takagi Y, Okada A: Alanyl-glutamine-sup-
85 Park JHY, Vanderhoof JA: Growth hormone 99 Rountree DB, Ulshen MH, Selub S et al: plemented parenteral nutrition increases lu-
did not enhance mucosal hyperplasia after Nutrient independent increases in progluca- minal mucus gel and decreases permeability
small bowel resection. Scand J Gastroenterol gon and ornithine decarboxylase messenger in the rat small intestine. J Parenter Enteral
1996;31:349–354. RNAs after jejunoileal resection. Gastroen- Nutr 1999;23:24–31.
86 Vanderhoof JA, Kollmann KA, Griffin S et al: terology 1992;103:462–468. 114 Ziegler TR, Young LS, Banfell K, Shelings M,
Growth hormone and glutamine do not stimu- 100 Taylor RG, Verity K, Fuller PJ: Ileal glucagon Hortos K, Bye R, Morrow FD, Jacobs DO,
late intestinal adaptation following massive gene expression: Ontogeny and response to Smith RJ, Antin JH, Wilmore DD: Clinical
small bowel resection in the rat. J Pediatr Gas- massive small bowel resection. Gastroenterol- and metabolic efficiency of glutamine-supple-
troenterol Nutr 1997;25:327–331. ogy 1990;99:724–729. mented parenteral nutrition after bone mar-
87 Shulman DI, Hu CS, Duckett G et al: Effects of 101 Cheeseman CI, Tsang R: The effect of gastric row transplantation. A randomized double-
short-term growth hormone therapy in rats un- inhibitory polypeptide and glucagons like blind, control study. Ann Intern Med 1992;
dergoing 75% small intestinal resection. J Pe- peptides on intestinal hexose transport. Am J 116:821–828.
diatr Gastroenterol Nutr 1992;14:3–11. Physiol 1996;261:G477–G482. 115 Mochizuki H, Saito H, Tashiro T, Tahara Y,
88 Benhamou PH, Canarelli JP, Richard C et al: 102 Dowling RH: Small bowel resection and its Okada A: A clinical study on the effect of ala-
Human recombinant growth hormone in- regulation. Scand J Gastroenterol Suppl nyl-glutamine supplemented TPN in gastrec-
creases small bowel lengthening after massive 1982;17:53–74. tomized patients – Early II study (abstract). J
small bowel resection in piglets. J Pediatr Surg 103 Gornacz GE, Al-Mukhtar MYT, Ghatei MA, Parenter Enteral Nutr 1999;23(suppl):160.
1997;32:1332–1336. Sagor GR, Wright NA, Bloom SR: Pattern of 116 Griffiths RD, Jones C, Palmer A: A six-
89 Kissmeyer-Nielsen P, Jensen MB, Laurberg S: cell proliferation and enteroglucagon re- month outcome of critically ill patients given
Perioperative growth hormone treatment and sponse following small bowel resection in the glutamine-supplemented parenteral nutri-
functional outcome after major abdominal sur- rat. Digestion 1984;29:65–72. tion. Nutrition 1997;13:295–302.
gery. A randomized double-blind, controlled 104 Al-Mukhtar MYT, Sagor GR, Ghati MA, 117 Wilmore DW, Lacey JM, Soultanakis RP,
study. Ann Surg 1999;229:298–302. Bloom SR, Wright NA: The role of pancreati- Bosch RL, Burne TA: Factors predicting a
90 Gomez de Segura IA, Aguilera MJ, Codesal J, co-biliary secretions in intestinal adaptation successful outcome after pharmacologic bow-
De Miguel E: Administration of growth hor- after resection, and its relationship to plasma el compensation. Ann Surg 1997;226:288–
mone enhances the intestinal adaptive re- enteroglucagon. Br J Surg 1983;70:398–400. 292.
sponse after resection of small intestine in rats. 105 Tappenden KA, Drozdowski LA, Thomson 118 Mandir N, Goodlad RA: The effects of gluta-
Rev Esp Enferm Dig 1995;87:288–293. AB, McBarney MI: Short-chain fatty acid- mine on intestinal cell proliferation in paren-
91 Cummings MC, Winterford CM, Walker NI: supplemented total parenteral nutrition alters terally fed rats. Gut 1999;44:608–614.
Apoptosis. Am J Surg Pathol 1997;21:88–101. intestinal structure, glucose transporter-2 119 Alavi K, Kato Y, Yu D, Schwartz MZ: Enter-
92 Cummings JH: Colonic absorption: The im- mRNA and protein, and proglucagon mRNA al glutamine does not enhance the effects of
portance of short chain fatty acids in man. abundance in normal rats. Am J Clin Nutr hepatocyte growth factor in short bowel syn-
Scand J Gastroenterol 1984;19(suppl 93):89– 1998;68:118–125. drome. J Pediatr Surg 1998;33:1666–1669.
99. 106 Souba WW: Intestinal glutamine metabolism
and nutrition. J Nutr Biochem 1993;4:2–9.

236 Dig Dis 2003;21:228–236 Botsios/Vasiliadis


Review Article

Dig Dis 2003;21:237–244


DOI: 10.1159/000073341

Helicobacter pylori Infection,


Vitamin B12 and Homocysteine
A Review

Jutta Dierkes a Matthias Ebert b Peter Malfertheiner b Claus Luley a


a Institute
of Clinical Chemistry and Biochemistry, and b Department of Gastroenterology and Hepatology,
University Hospital Magdeburg, Magdeburg, Germany

Key Words Introduction


Helicobacter pylori infection W Vitamin B12 deficiency W
Homocysteine W Pernicious anemia The aetiology of atrophic gastritis and gastric cancer
has been rewritten since the detection of Helicobacter
pylori during the 1980s. It is now known that the major
Abstract cause of atrophic gastritis is an infection with H. pylori,
It has been suggested that there is an association be- which normally occurs in early childhood and persists
tween Helicobacter pylori infection, reduced cobalamin lifelong if left untreated. Prevalence rates of H. pylori
absorption and cobalamin status and, consequently, ele- infection vary by age, country of origin, socioeconomic
vated homocysteine levels. This would offer an explana- status and are typically about 20–50% in healthy adults in
tion why H. pylori infection is associated with coronary Western Europe and 80% in adults in economically less
heart disease. To date, more than 25 studies have been developed countries [1]. Although the infection will cause
published that either deal with H. pylori infection and gastric inflammation in virtually all infected subjects, the
homocysteine, H. pylori infection and cobalamin status, majority of infected subjects will remain asymptomatic,
or both. The design of these studies differs widely in while others develop atrophic gastritis and subsequently
terms of definition of H. pylori status, measuring cobal- gastric ulcer which can further develop to gastric cancer
amin status, selection of study cohorts and geographical [2].
study areas. Therefore, results are fairly inconclusive at One severe consequence of atrophic gastritis is the
present and do not suggest a major role of H. pylori malabsorption of cobalamin (vitamin B12), which is fre-
infection in the development of cobalamin deficiency quent in the elderly due to hypo- or achlorhydria with sub-
and elevated homocysteine levels. sequent bacterial overgrowth, and reduced production
Copyright © 2003 S. Karger AG, Basel and secretion of intrinsic factor [3]. It has been suggested
that H. pylori infection may play an important role in the
reduction of acid production, reduced intrinsic factor
secretion and therefore the development of vitamin B12
deficiency. However, development of vitamin B12 defi-

© 2003 S. Karger AG, Basel Dr. Jutta Dierkes


ABC 0257–2753/03/0213–0237$19.50/0 Institute of Clinical Chemistry and Biochemistry
Fax + 41 61 306 12 34 Leipziger Strasse 44, DE–39120 Magdeburg (Germany)
E-Mail karger@karger.ch Accessible online at: Tel. +49 391 6713900, Fax +49 391 6713902
www.karger.com www.karger.com/ddi E-Mail jutta.dierkes@medizin.uni-magdeburg.de
Table 1. Studies on H. pylori and cobalamin, pernicious anemia and homocysteine

Group (first author) Patients Effect with H. pylori

Cobalamin
Van Asselt, 1998 [21] Mild cobalamin deficiency in healthy elderly No association
Pilott, 1996 [22] Serum cobalamin and folate in elderly No association
Cenerelli, 2002 [23] Serum cobalamin and folate in diabetes mellitus No association
type 2
Bloemenkamp, 2001 [24] Serum cobalamin and folate women with No association
peripheral artery disease
Shuval-Sudai, 2003 [25] Serum cobalamin in outpatients Higher frequency of low serum cobalamin in H.pylori-
positive patients
Tamura, 2002 [26] Serum cobalamin and folate in patients with Lower serum cobalamin and folate in patient with
coronary artery disease H. pylori
Carmel, 2001 [28] Food cobalamin malabsorption in patients with Lower food cobalamin absorption in H.pylori-positive
low cobalamin patients
Serin, 2002 [29] Gastric histology and serum cobalamin Serum cobalamin related to atrophy and
inflammation and H.pylori density
Pernicious anemia
Fong, 1991 [11] Pernicious anemia, case-control study No association
Haruma, 1995 [12] Pernicious anemia, case-control study No association
Marignani, 1999 [13] Patients with macrocytic anemia No association
Annibale, 2001 [14] Pernicious anemia No association
Andres, 2003 [15] Cobalamin-deficient elderly No association
Annibale, 2000 [16] Pernicious anemia Frequency of H. pylori-positive patients: 60%
Kaptan, 2000 [17] Vitamin B12-related anemia Frequency of H. pylori-positive patients: 56%
Avcu, 2001 [18] Vitamin B12 deficiency Frequency of H. pylori-positive patients: 57%
Ma, 1994 [19] Pernicious anemia Frequency of H.pylori-positive patients: 83%
Homocysteine
Saxena, 1997 [30] Patients No association
Leung, 2001 [31] Dyspeptic patients No association
Yoshino, 2002 [32] Case-control H. pylori-positive vs. -negative No association
Bloemenkamp, 2001 [24] Women with peripheral artery disease and controls No association
Tamura, 2002 [26] Patients with coronary artery disease Higher homocysteine in H. pylori-positive patients
Whincup, 2000 [33] Prospective study on CVD Higher homocysteine in H. pylori-positive patients
Cenerelli, 2002 [23] Patients with diabetes mellitus type 2 and controls Higher homocysteine in H. pylori-positive patients
Effect of eradication therapy
Leung, 2001 [31] Dyspeptic patients No change in homocysteine
Kaptan, 2000 [17] Patients with low vitamin B12 and anemia Increase in serum cobalamin and improvement in
haematological variables
Avcu, 2001 [18] Vitamin B12 deficiency anemia Increase in serum cobalamin and improvement in
haematological variables
Serin, 2002 [29] Low serum cobalamin, no anemia Increase in serum cobalamin

ciency occurs slowly due to the low requirement (2 Ìg/ deficiency which are often overlooked are psychiatric and
day), the enterohepatic cycle of cobalamin and the liver neurodegenerative changes [4]. Diagnosis of vitamin B12
stores of the vitamin that have been built up during life deficiency is usually made by low serum cobalamin con-
and that are about 2–3 mg of cobalamin by the age of 60 centrations which, however, has an only low diagnostic
[3]. sensitivity [5]. Other diagnostic tools to detect vitamin
The classical sign of vitamin B12 deficiency is megalo- B12 deficiency are the measurement of specific metabo-
blastic anaemia which, however, occurs in only 50% of lites such as homocysteine and methylmalonic acid [6, 7].
vitamin B12-deficient subjects. Other signs of vitamin B12 With the exception of homocysteine, these diagnostic

238 Dig Dis 2003;21:237–244 Dierkes/Ebert/Malfertheiner/Luley


tools require specialized equipment and are not offered anaemia were older than those with microcytic anaemia,
routinely. had lower vitamin B12 levels and higher gastrin levels, but
Lack of intrinsic factor causing pernicious anaemia is only 2 of 44 patients were H. pylori-positive in contrast to
classically diagnosed by the Schilling test. This test, how- 22 of 36 patients with microcytic anaemia.
ever, will not detect malabsorption of food-bound cobal- Annibale et al. [14] investigated 150 patients with atro-
amin, since free, crystalline vitamin B12 is used during the phic body gastritis. Patients were grouped according to
Schilling test. Therefore, another test, the egg yolk cobal- their H. pylori status and the prevalence of pernicious
amin absorption test (EYCAT) has been introduced for anaemia. 37 patients were H. pylori-negative by both
the diagnosis of malabsorption of food-bound cobalamin serology and histology; 79 patients were negative by his-
[8]. However, this test is not widely used. tology but positive by serology and 34 patients were posi-
The interest in the association of H. pylori infection tive by both serology and histology. The lowest frequency
with cobalamin status is further triggered by the associa- of pernicious anaemia was found in patients being posi-
tion of low cobalamin status with hyperhomocysteinaem- tive by serology and histology (12%), an intermediate fre-
ia which is a widely acknowledged risk factor of athero- quency was found in the group with negative histology but
sclerosis [9]. The association between H. pylori – low positive serology (46%) and the highest frequency of per-
cobalamin – elevated homocysteine offers a metabolic nicious anaemia was found in the group negative for H.
explanation why H. pylori infection is associated with pylori (76%).
increased risk for cardiovascular disease, especially coro- Andres et al. [15] investigated the H. pylori infection
nary heart disease [10]. status in 60 elderly patients who had been selected by
This review summarizes the present knowledge of the cobalamin deficiency due to food-cobalamin malabsorp-
association of H. pylori, cobalamin and homocysteine tion and who presented with polyneuropathy (35%), de-
concentrations. It considers studies published in English mentia and confusion (30%) and anaemia (27%). Gastric
until April 2003 (table 1). atrophy was documented in 19 out of 32 investigated
patients, but infection with H. pylori was documented in
only 1 of the patients.
Helicobacter and Pernicious Anaemia The above cited studies suggest that the hypothesis
that H. pylori is a causative factor in pernicious anaemia
If H. pylori indeed causes cobalamin deficiency, an does not hold true and that H. pylori infection is even pro-
association of H. pylori with pernicious anaemia could tective in the development of cobalamin-associated
also be expected. This issue was investigated by a number anaemia. However, some other studies report that pa-
of studies in patients with pernicious anaemia which, tients with pernicious anaemia have H. pylori infection
however, could not prove this hypothesis and even rates comparable to the normal population.
showed a lower prevalence of H. pylori infection in Annibale et al. [16] reported that 49 of 81 patients with
patients with pernicious anaemia. pernicious anaemia were positive for H. pylori infection
Fong et al. [11] studied the frequency of H. pylori infec- (60%), although only 8 showed a positive histological H.
tion in 28 patients with pernicious anaemia and 28 age- pylori status and 41 were positive by serological investiga-
matched control subjects (mean age 59 B 14 years). Diag- tion. A Turkish study revealed comparable results [17].
nosis of H. pylori infection was made by gastric biopsy They investigated 138 patients with vitamin B12 deficien-
and serology. Positive biopsies were found in 3 of 28 cy and anaemia which was not due to intrinsic factor defi-
patients with pernicious anaemia (11%) in contrast to 20 ciency or gastrectomy. Diagnosis of H. pylori infection
of 28 control subjects (71%). A similar result was reported was confirmed by rapid urease testing and gastric biopsies
by Haruma et al. [12], who investigated 24 patients with in 77 of 138 patients (56%). Another study from Turkey
pernicious anaemia and 24 age-matched control subjects. [18] reported that among 421 patients with vitamin B12
H. pylori status was diagnosed by biopsies and IgG anti- deficiency, H. pylori infection was detected in 241 of them
bodies. No patient with pernicious anaemia but 67% of (57%). A small Swedish study [19] involving 30 patients
control subjects were H. pylori-positive. A low frequency with pernicious anaemia reported that 25 of them had
of H. pylori infection in patients with macrocytic anaemia antibodies against H. pylori (83%).
in comparison to microcytic anaemia was also reported by Thus, so far, studies revealed heterogeneous data on
Marignani et al. [13]. H. pylori status was diagnosed by the association of H. pylori infection and cobalamin-relat-
biopsy and IgG antibodies. Patients with macrocytic ed anaemia. However, it has been suggested that the low

H. pylori Infection, Vitamin B12 and Dig Dis 2003;21:237–244 239


Homocysteine
frequency of H. pylori infection in total gastric atrophy min and folate in 102 elderly. H. pylori antibodies were
can be explained by the fact that infected subjects become found in 84% of the elderly.
Helicobacter-negative as a result of the virtual absence of Cenerelli et al. [23] investigated patients with diabetes
acid production which inhibits survival of H. pylori [20]. mellitus type 2 and controls and found no effect of H.
This would mean that the low frequency of H. pylori pylori infection on the level of cobalamin. Even higher
infection reported by some of the studies reflects a false- cobalamin levels were reported by Bloemenkamp et al.
negative result. As all studies on this issue had a cross- [24] in H. pylori-positive subjects in a study including
sectional design, a definitive answer cannot be given at women with peripheral artery disease and controls. Since
present. Longitudinal studies, however, are difficult to the latter two studies also investigated homocysteine, they
perform and are ethically questionable. are described in detail below.
On the other hand, a significant association of H. pylori
infection and prevalence of low cobalamin concentrations
Helicobacter and Subclinical Cobalamin was reported by Shuval-Sudai and Granot [25]. 133
Deficiency patients were investigated in Israel for H. pylori infection
and cobalamin and folate status. Patients were enrolled in
Vitamin B12 deficiency is rare in young adults and has a community primary care clinic, were between 18 and 90
an increasing prevalence in the elderly. Development of years of age, and were eligible for the study if they had no
vitamin B12 requires years and depends mainly on pres- history of H. pylori eradication, previous gastrointestinal
ence of achlorhydria and on whether intrinsic factor is disease or surgery, vegetarian diet or multivitamin use. H.
present or absent, since the intrinsic factor is necessary to pylori status was assessed by IgG antibodies. A signifi-
maintain the enterohepatic circle of cobalamin. Thus, cantly higher frequency of low cobalamin levels (cut-off
hypo- and achlorhydria which compromise food-cobal- !250 pg/ml) was found in H. pylori-positive patients (n =
amin absorption must be present for long time before 96). Similar results were obtained with the lower cut-off
vitamin B12 deficiency occurs. Furthermore, subclinical level of 180 or 145 pg/ml. Interestingly, lower folate levels
vitamin B12 deficiency is difficult to diagnose as haemato- were observed in the H. pylori-positive patients.
logical changes are rare in this stage. Therefore, it is main- A similar result was reported by Tamura et al. [26] who
ly diagnosed by low serum cobalamin or metabolic investigated 93 patients with coronary artery disease and
changes as elevated concentrations of the metabolites observed both lower cobalamin and folate levels in pa-
homocysteine or methylmalonic acid. Studies have also tients positive for H. pylori. H. pylori infection was diag-
investigated whether food-cobalamin malabsorption is as- nosed by biopsies. In this study, homocysteine was also
sociated with H. pylori infection and whether the degree investigated (see below).
of gastric atrophy can be linked to cobalamin deficiency. Reasons for differences in cobalamin status due to H.
Van Asselt et al. [21] investigated the determinants of pylori infection may be that H. pylori infection affects the
cobalamin deficiency in healthy elderly, but did not find a absorption of either food-cobalamin or free cobalamin
statistically significant association of H. pylori infection which can be studied employing the EYCAT test or the
with mild cobalamin deficiency. The study included 105 Schilling test, respectively. In particular, food-cobalamin
healthy elderly Dutch subjects (age range 74–80 years) malabsorption has been investigated with respect to H.
who were classified according to cobalamin and methyl- pylori infection.
malonic acid concentrations as ‘cobalamin-deficient’ (n = The contribution of different factors to food-cobala-
25), ‘possibly cobalamin-deficient’ (n = 53) or ‘not cobal- min malabsorption was studied in a small group of 19 vol-
amin-deficient’ (n = 27). H. pylori IgG antibodies were unteers by Cohen et al. [27]. Volunteers had been selected
found in 54, 68 and 44% of the respective groups. Thus, it on the basis of age and their cobalamin levels to ensure a
was concluded that both H. pylori infection and mild wide range of deficient and replete subjects. Food-cobal-
cobalamin deficiency was common in this cohort, but H. amin absorption was tested using the EYCAT. H. pylori
pylori infection did not contribute significantly to cobal- status was determined by biopsies and 13C-urea breath
amin deficiency. tests. Gastric function was investigated by basal and pen-
Also, other studies did not find a difference in cobal- tagastrin-stimulated gastric aspirates, and gastric inflam-
amin levels between H pylori-positive and -negative pa- mation and atrophy were scored on scales ranging from 0
tients. Pilott et al. [22] reported no effect of H. pylori to 3 and 1 to 4, respectively. Six of the 19 patients had
infection on nutritional indices including serum cobala- severe food cobalamin malabsorption. Three of them also

240 Dig Dis 2003;21:237–244 Dierkes/Ebert/Malfertheiner/Luley


had H. pylori infection and antral gastritis while the other with hyperhomocysteinaemia. A further link may exist
3 patients did not have H. pylori infection but corpus atro- due to the observed effect of H. pylori on folate status.
phy with achlorhydria. Both vitamin B12 and folate status are important for
The issue whether H. pylori infection affects food-cobal- maintenance of low homocysteine concentrations. Pres-
amin malabsorption was further studied by Carmel et al. ent evidence suggests that elevated homocysteine concen-
[28]. Data of 202 subjects who underwent the EYCAT test trations are independently associated with increased risk
in their clinic, among them 43 healthy volunteers and 159 for cardiovascular disease. Thus, hyperhomocysteinaem-
patients with low cobalamin levels, were analysed retro- ia in patients with H. pylori infection may be one mecha-
spectively. H. pylori infection was confirmed either by the nism by which H. pylori is associated with coronary heart
presence of IgG antibodies or the 13C-urea breath test. disease.
Determination of H. pylori status was done in 167 subjects However, an association of H. pylori infection and
and in 133 subjects, serum pepsinogen I and II were mea- homocysteine concentrations was not reported in a num-
sured, while serum gastrin and parietal cell antibodies were ber of studies. Saxena et al. [30] investigated homocys-
assayed in 158 subjects. Severe food-cobalamin malabsorp- teine in 220 subjects (mean age 66 B 10 years). They
tion was found in 24% of the patients with low cobalamin reported no differences in homocysteine levels in patients
levels and in 9% of the healthy volunteers. The mean excre- with positive serology for H. pylori (n = 122) compared to
tion rate of radiolabelled cobalamin during the EYCAT those with no antibodies against H. pylori. The result
was significantly lower in H. pylori-positive subjects than in remained the same after analyzing men and women sepa-
H. pylori-negative subjects (2.26 B 1.90% vs. 3.43 B rately. Interestingly, the values reported by the authors
2.22%). H. pylori infection was associated with presence of are remarkably high with a mean homocysteine value of
severe food-cobalamin malabsorption (78% H. pylori-posi- 22.7 B 11 Ìmol/l.
tive subjects vs. 50% H. pylori-positive subjects in mild A similar result was reported by Leung et al. [31] who
food-cobalamin malabsorption and 44% H. pylori-positive investigated the association between H. pylori and homo-
subjects with normal absorption). In a multivariate regres- cysteine in 49 patients with dyspepsia referred for endos-
sion analysis using data of 136 subjects, ethnic origin, age, copy. Patients were relatively young (median age 43
H. pylori status, and serum gastrin contributed indepen- years), and 37 were found to be H. pylori-positive by gas-
dently to the EYCAT result. tric biopsies. There was no difference in homocysteine in
Thus, present studies support the view that H. pylori infected compared to non-infected subjects and no asso-
infection interferes with food-cobalamin absorption, al- ciation of homocysteine to gastric histology or H. pylori
though results by Cohen et al. [27] suggest that different density.
mechanisms may be involved. Further studies investi- A Japanese study [32] included 45 healthy middle-
gated whether cobalamin status is related to gastric atro- aged subjects infected with H. pylori and 45 age-matched
phy or inflammation. Serin et al. [29] selected 145 pa- controls without infection. The mean age was 39 years in
tients from a gastroenterological clinic who did not show both groups. H. pylori infection was diagnosed by 13C-
signs of atrophy, erosions, or ulcers. Biopsy specimen of urea breath test and serology. No difference was found in
the gastric antrum and corpus were taken and analysed plasma homocysteine between patients and controls.
for signs of inflammation, gastritis and H. pylori density. None of these studies investigated cobalamin or folate sta-
Vitamin B12 was measured in serum. Histopathological tus.
scores for both antral and corpus H. pylori density and No difference in homocysteine according to H. pylori
inflammation were significantly inversely associated with status was found in 150 women with peripheral artery dis-
serum vitamin B12 levels. In multivariate analyses, only ease and 412 control women [24]. H. pylori was diagnosed
H. pylori density was associated with vitamin B12 levels. by positive IgG antibody test. The frequency of H. pylori
infection in PAD patients was 42% in comparison to 27%
in control subjects, resulting in an odds ratio for PAD of
Is Helicobacter Infection Related to 2.0 (1.3–2.9). Homocysteine was significantly higher in
Hyperhomocysteinaemia? patients than in controls, however, adjustment for homo-
cysteine did not alter substantially the association be-
Although the role for H. pylori in the development of tween H. pylori and PAD. In this study, folate and vita-
cobalamin deficiency is not clear, it offers an attractive min B12 levels were also measured. No difference was
explanation why H. pylori infection could be associated found in homocysteine and folate levels between H. pylo-

H. pylori Infection, Vitamin B12 and Dig Dis 2003;21:237–244 241


Homocysteine
ri-positive and -negative patients and even higher cobal- change in homocysteine concentration after eradication
amin levels were measured in H. pylori-positive subjects. (10.5 vs. 10.2 Ìmol/l).
The authors conclude that homocysteine cannot explain A number of studies reported a spontaneous improve-
the association between H. pylori infection and athero- ment of vitamin B12 status after H. pylori eradication,
sclerosis. It has to be noted that the blood sample for anal- without specific treatment with cobalamin. Serin et al.
yses was taken between 1998 and 2000, while the diagno- [29] investigated 65 of 145 patients after eradication ther-
sis of PAD was made between 1990 and 1999. apy. At baseline, 145 patients with no gastric atrophy, ero-
In contrast to the above-mentioned reports on lack of sion or ulcers were included. Patients were then given an
association between H. pylori and homocysteine concen- eradication therapy and 65 of the patients were re-ana-
trations, other investigators reported significantly higher lyzed 2–3 months later. Baseline cobalamin levels were
levels of homocysteine in H. pylori-positive patients. Ta- indicative for cobalamin deficiency although no anaemia
mura et al. [26] studied homocysteine, related vitamins was present. In 33 of these 65 patients, eradication thera-
and H. pylori infection in 93 Japanese patients who py was successful. The authors observed a significant
underwent coronary arteriography. The mean age of the increase in vitamin B12 after eradication therapy with a
patients was 64 B 9 years, and 57 of them were found to more pronounced increase in those with complete eradi-
be H. pylori-positive as diagnosed by stomach biopsies. cation.
Significantly lower cobalamin and folate levels and higher Avcu et al. [18] investigated 108 H. pylori-infected
levels of homocysteine were found in H. pylori-positive patients where H. pylori was also present in dental plaques
patients than in H. pylori-negative patients. Homocys- and who suffered from vitamin B12 deficiency and from
teine was weakly, but significantly related to the atrophic signs of anaemia. Mean patient age was 44 years. After
score of the stomach of the patients. eradication therapy, gastric biopsies were repeated in 61
Whincup et al. [33] reported higher homocysteine lev- patients and confirmed the successful eradication thera-
els both in H. pylori-positive patients with coronary heart py. Haematological parameters (red blood cell count, hae-
disease and in healthy controls which have been prospec- moglobin, haematocrit and MCV) improved significantly
tively investigated in the British Regional Heart Study. as well as vitamin B12 levels which increased from 71 B
However, the effect of H. pylori infection was, although 11 to 293 B 11 pg/ml. Mean follow-up time was 15 B 1
significantly, not very strong and increased the homocys- months and no treatment for vitamin B12 deficiency was
teine modestly. No vitamin levels were reported in this given.
study. A similar effect of eradication therapy was observed by
Cenerelli et al. [23] investigated homocysteine in pa- Kaptan et al. [17]. Eradication therapy was given to 77
tients with diabetes type 2 and healthy controls (n = 30 patients with a positive H. pylori biopsy who were also
and 43, respectively) and found no difference between anaemic and had low cobalamin levels. In 31 patients, H.
patients and controls. However, subjects with H. pylori pylori eradication therapy was successful and the cobal-
infection showed significantly higher homocysteine con- amin level and the blood count were re-investigated after
centrations. The diagnosis of H. pylori was made by 13C- a mean follow-up time of 42 B 19 months. There was a
urea breath test. There was no difference in folate or significant improvement in haematocrit, and MCV, and a
cobalamin between H. pylori-positive and -negative sub- significant increase in serum cobalamin at re-investiga-
jects. tion (63 B 30 to 223 B 38 pmol/l). Patients with unsuc-
cessful eradication therapy received cyanocobalamin
therapy, but results were not reported.
Does H. pylori Eradication Affect Cobalamin
Status and Homocysteine?
Discussion
There are a few studies that investigate the effect of H.
pylori eradication therapy on homocysteine or cobalamin Although the hypothesis on the association of H. pylori
levels. The only study that investigated homocysteine was infection, cobalamin and homocysteine status is clear and
reported by Leung et al. [31] who treated 37 dyspeptic intriguing, the results of the clinical studies on this asso-
patients. Eradication was confirmed by a negative 13C- ciation revealed highly disparate results and do not clearly
urea breath test. After 24 weeks, homocysteine was mea- support the hypothesis. On the other hand, there is evi-
sured again in these patients. There was no significant dence that cobalamin status improves after H. pylori erad-

242 Dig Dis 2003;21:237–244 Dierkes/Ebert/Malfertheiner/Luley


ication suggesting a causal effect of H. pylori in the devel- teine concentrations. Lower folate concentrations in H.
opment of cobalamin deficiency. pylori-positive patients have been shown in two of four
The different results on H. pylori and cobalamin status studies that also reported folate concentrations. Dimin-
may depend on study design, selection of patients and ished folate absorption has been shown in patients with
variables measured. Cobalamin status in the studies var- atrophic gastritis [35], but the effect of H. pylori infection
ies widely with pernicious anaemia as the ‘tip of the ice- has not been investigated in particular.
berg’ in cobalamin deficiency, and milder forms of cobal- In conclusion, clinical studies do not show a uniform
amin deficiency that are not always clearly defined. It has association of H. pylori infection with cobalamin or folate
to be considered that many patients with cobalamin mal- deficiency or elevated homocysteine concentrations.
absorption do not develop pernicious anaemia at all or Therefore, routine determination of cobalamin or homo-
only after decades. H. pylori infection obviously is associ- cysteine in H. pylori-infected patients as a means of pre-
ated with food-cobalamin malabsorption, but the associa- vention of deficiency or with respect to atherosclerotic
tion with cobalamin deficiency due to intrinsic factor risk cannot be recommended at present. On the other
deficiency was not demonstrated. Furthermore, the low hand, the studies do suggest that H. pylori infection has a
prevalence of H. pylori infection in pernicious anaemia is role in atrophic gastritis and food-cobalamin malabsorp-
a striking phenomenon that may be explained by the pos- tion. Subgroups of infected patients are obviously at risk
sibility that infected subjects become Helicobacter-nega- of developing cobalamin deficiency and hyperhomocys-
tive as a result of the virtual absence of acid production teinaemia, who are not identified at present. Before a con-
which inhibits survival of H. pylori [20]. cise recommendation can be given, more data in patient
Most studies investigated the effect of H. pylori on groups with a well-defined cobalamin status are needed.
serum cobalamin levels. However, serum cobalamin has
been shown to have limited sensitivity to detect deficien-
cy since many patients still have low-to-normal serum Acknowledgement
concentrations [6]. Instead, diagnosis of subclinical cobal-
M. Ebert is supported by the Heisenberg-Programme of the DFG
amin deficiency should be based on metabolic markers
(Eb 187/5-1).
like methylmalonic acid or homocysteine [5]. Most stud-
ies, however, did not use these additional diagnostic
tools.
How can the increase of vitamin B12 after eradication
therapy be explained? The only study that also measured
homocysteine after eradication did not find an effect of
eradication on homocysteine [31]. Three studies reported
on vitamin B12 response to eradication therapy. None of
these studies reported individual data and in each case, a
substantial portion of patients was missed during follow-
up. Thus, it can be questioned whether the effect may be
influenced by regression to the mean or selection bias.
However, it is possible that there is a real effect since
reversal of food-cobalamin malabsorption in atrophic gas-
tritis after tetracycline therapy was reported earlier [34].
A likely mechanism is the reduction of bacteria in the
stomach and small intestine in hypo- or achlorhydria.
This finding supports the view that H. pylori infection
may also play a role in food-cobalamin malabsorption.
In addition to the association of H. pylori to cobala-
min, studies on the association of H. pylori to homocys-
teine revealed also inconsistent results. A significant ef-
fect of H. pylori infection on plasma homocysteine was
found in three of six studies. Besides cobalamin deficien-
cy, folate deficiency is also associated to higher homocys-

H. pylori Infection, Vitamin B12 and Dig Dis 2003;21:237–244 243


Homocysteine
References

1 Suerbaum S, Michetti P: Helicobacter pylori 14 Annibale B, Negrini R, Caruana P, Lahner E, 25 Shuval-Sudai O, Granot E: An association be-
infection. N Engl J Med 2002;347:1175–1186. Grossi C, Bordi C, Delle Fave G: Two-thirds of tween Helicobacter pylori infection and serum
2 Parsonnet J, Friedman GD, Vandersteen DP, atrophic body gastritis patients have evidence vitamin B12 levels in healthy adults. J Clin Gas-
Chang Y, Vogelman JH, Orentreich N, Sibley of Helicobacter pylori infection. Helicobacter troenterol 2003;36:130–133.
RK: Helicobacter pylori infection and the risk 2001;6:225–233. 26 Tamura A, Fujioka T, Nasu M: Relation of
of gastric carcinoma. N Engl J Med. 1991; 15 Andres E, Noel E, Kaltenbach G, Perrin AE, Helicobacter pylori infection to plasma vitamin
325(16):1127–1131. Vinzio S, Goichot B, Schlienger JL, Blickle JF: B12, folic acid and homocysteine levels in pa-
3 Carmel R: Cobalamin, the stomach and aging. Vitamin B12 deficiency with normal Schilling tients who underwent diagnostic coronary arte-
Am J Clin Nutr 1997;66:750–759. test or non-dissociation of vitamin B12 and its riography. Am J Gastroenterol 2002;97:861–
4 Nexo E, Hansen M, Rasmussen K, Lindgren A, carrier proteins in elderly patients. A study of 866.
Grasbeck R: How to diagnose cobalamin defi- 60 patients (in French). Rev Méd Interne 2003; 27 Cohen H, Weinstein WM, Carmel R: Hetero-
ciency. Scand J Clin Lab Invest Suppl 1994; 24:218–223. geneity of gastric histology and function in food
219:61–76. 16 Annibale B, Lahner E, Bordi C, Martino G, cobalamin malabsorption: Absence of atrophic
5 Stabler SP, Lindenbaum J, Allen RH: The use Caruana P, Grossi C, Negrini R, Delle Fave G: gastritis and achlorhydria in some patients
of homocysteine and other metabolites in the Role of Helicobacter pylori infection in perni- with severe malabsorption. Gut 2000;47:638–
specific diagnosis of vitamin B12 deficiency. J cious anaemia. Dig Liver Dis 2000;32:756– 645.
Nutr 1996;126(suppl):1266–1272. 762. 28 Carmel R, Aurangzeb I, Qian D: Associations
6 Bolann BJ, Solli JD, Schneede J, Grottum KA, 17 Kaptan K, Beyan C, Ural AU, Cetin T, Avcu F, of food-cobalamin malabsorption with ethnic
Loraas A, Stokkeland M, Stallemo A, Schjoth Gulsen M, Finci R, Yalcin A: Helicobacter origin, age, Helicobacter pylori infection, and
A, Bie RB, Refsum H, Ueland PM: Evaluation pylori – Is it a novel causative agent in vitamin serum markers of gastritis. Am J Gastroenterol
of indicators of cobalamin deficiency defined B12 deficiency? Arch Intern Med 2000;160: 2001;96:63–70.
as cobalamin-induced reduction in increased 1349–1353. 29 Serin E, Gumurdulu Y, Ozer B, Kayaselcuk F,
serum methylmalonic acid. Clin Chem 2000; 18 Avcu N, Avcu F, Beyan C, Ural AU, Kaptan K, Yilmaz U, Kocak R: Impact of Helicobacter
46:1744–1750. Ozyurt M, Nevruz O, Yalcin A: The relation- pylori on the development of vitamin B12 defi-
7 Holleland G, Schneede J, Ueland PM, Lund ship between gastric-oral Helicobacter pylori ciency in the absence of gastric atrophy. Helico-
PK, Refsum H, Sandberg S: Cobalamin defi- and oral hygiene in patients with vitamin B12- bacter 2002;7:337–341.
ciency in general practice. Assessment of the deficiency anemia. Oral Surg Oral Med Oral 30 Saxena V, Markus H, Swaminathan S, Mendall
diagnostic utility and cost-benefit analysis of Pathol Oral Radiol Endod 2001;92:166–169. ME: Hyperhomocysteinaemia, Helicobacter
methylmalonic acid determination in relation 19 Ma JY, Borch K, Sjostrand SE, Janzon L, pylori and coronary heart disease. Heart 1997;
to current diagnostic strategies. Clin Chem Mardh S: Positive correlation between H,K- 78:524.
1999;45:189–198. adenosine triphosphatase autoantibodies and 31 Leung WK, Ma PK, Choi PC, Ching JY, Ng
8 Doscherholmen A, Ripley D, Chang S, Silvis Helicobacter pylori antibodies in patients with AC, Poon P, Woo KS, Sung JJ: Correlation
SE: Influence of age and stomach function on pernicious anaemia. Scand J Gastroenterol between Helicobacter pylori infection, gastric
serum vitamin B12 concentration. Scand J Gas- 1994;29:961–965. inflammation and serum homocysteine con-
troenterol 1977;12:313–319. 20 Kuipers EJ, Klinkenberg-Knol EC, Vanden- centration. Helicobacter 2001;6:146–150.
9 Graham IM, O’Callaghan P: Vitamins, homo- broucke-Grauls CM, Appelmelk BJ, Schenk 32 Yoshino N, Adachi K, Takashima T, Miyaoka
cysteine and cardiovascular risk. Cardiovasc BE, Meuwissen SG: Role of Helicobacter pylori Y, Yuki T, Ishihara S, Kinoshita Y: Helicobac-
Drugs Ther 2002;16:383–389. in the pathogenesis of atrophic gastritis. Scand ter pylori infection does not affect the serum
10 Pellicano R, Oliaro E, Gandolfo N, Aruta E, J Gastroenterol Suppl 1997;223:28–34. level of homocysteine. Am J Gastroenterol
Mangiardi L, Orzan F, Bergerone S, Rizzetto 21 Van Asselt DZ, de Groot LC, van Staveren 2002;97:2927–2928.
M, Ponzetto A: Ischemic cardiovascular dis- WA, Blom HJ, Wevers RA, Biemond I, Hoef- 33 Whincup P, Danesh J, Walker M, Lennon L,
ease and Helicobacter pylori. Where is the link? nagels WH: Role of cobalamin intake and atro- Thomson A, Appleby P, Hawkey C, Atherton J:
J Cardiovasc Surg (Torino) 2000;41:829–833. phic gastritis in mild cobalamin deficiency in Prospective study of potentially virulent strains
11 Fong TL, Dooley CP, Dehesa M, Cohen H, older Dutch subjects. Am J Clin Nutr 1998;68: of Helicobacter pylori and coronary heart dis-
Carmel R, Fitzgibbons PL, Perez-Perez GI, 328–334. ease in middle-aged men. Circulation 2000;
Blaser MJ: Helicobacter pylori infection in per- 22 Pilott A, Fabrello R, Franceschi M, Scagnelli 101:1647–1652.
nicious anemia: A prospective controlled M, Soffiati F, Di Mario F, Fortunato A, Valerio 34 Suter PM, Golner BB, Goldin BR, Morrow FD,
study. Gastroenterology 1991;100:328–332. G: Helicobacter pylori infection in asymptom- Russell RM: Reversal of protein-bound vita-
12 Haruma K, Komoto K, Kawaguchi H, Okamo- atic elderly subjects living at home or in a nurs- min B12 malabsorption with antibiotics in atro-
to S, Yoshihara M, Sumii K, Kajiyama G: Per- ing home: Effects on gastric function and nutri- phic gastritis. Gastroenterology 1991;101:
nicious anaemia and Helicobacter pylori infec- tional status. Age Aging 1996;25:245–249. 1039–1045.
tion in Japan: Evaluation in a country with a 23 Cenerelli S, Bonazzi P, Galeazzi R, Testa I, 35 Russell RM, Krasinski SD, Samloff IM, Jacob
high prevalence of infection. Am J Gastroen- Bonfigli AR, Sirolla C, Giunta S, Galeazzi L, RA, Hartz SC, Brovender SR: Folic acid mal-
terol 1995;90:1107–1110. Fumelli D, Testa R: Helicobacter pylori masks absorption in atrophic gastritis. Possible com-
13 Marignani M, Delle Fave G, Mecarocci S, Bor- differences in homocysteine plasma levels be- pensation by bacterial folate synthesis. Gastro-
di C, Angeletti S, D’Ambra G, Aprile MR, Cor- tween controls and type 2 diabetic patients. Eur enterology 1986;91:1476–1482.
leto VD, Monarca B, Annibale B: High preva- J Clin Invest 2002;32:158–162.
lence of atrophic body gastritis in patients with 24 Bloemenkamp DG, Mali WP, Tanis BC, Ro-
unexplained microcytic and macrocytic sendaal FR, van den Bosch MA, Kemmeren
anaemia: a prospective screening study. Am J JM, Algra A, Visseren FL, van der Graaf Y:
Gastroenterol 1999;94:766–772. The relation between Helicobacter pylori and
atherosclerosis cannot be explained by a high
homocysteine concentration. Eur J Clin Invest
2002;32:549–555.

244 Dig Dis 2003;21:237–244 Dierkes/Ebert/Malfertheiner/Luley


Original Paper

Dig Dis 2003;21:245–251


DOI: 10.1159/000073342

Prevalence of Malnutrition in
Hospitalized Medical Patients: Impact of
Underlying Disease
Matthias Pirlich a Tatjana Schütz a Martin Kemps a Niklas Luhman a
Gerd-Rüdiger Burmester b Gert Baumann c Mathias Plauth d
Heinrich Josef Lübke e Herbert Lochs a
a Medizinische Klinik mit Schwerpunkt Gastroenterologie, Hepatologie und Endokrinologie, b Rheumatologie und

klinische Immunologie, c Kardiologie, Pulmonologie und Angiologie, Universitätsklinikum Charité,


Humboldt-Universität zu Berlin; d Klinik für Innere Medizin, Städtisches Klinikum Dessau und e Medizinische Klinik I,
Krankenhaus Zehlendorf, Berlin, Deutschland

Key Words diseases. Patients with gastrointestinal diseases, how-


Malnutrition, prevalence W Malnourished hospitalized ever, were not more frequently malnourished than other
patients W Subjective global assessment W medical patients (28.8 vs. 22.0%). Malnourished patients
Gastrointestinal diseases W Malnutrition, diagnoses were significantly older (70.0 B 13.6 vs. 58.3 B 15.6
years, p ! 0.0001) and had a 40% longer hospital stay
(13.1 B 8.1 vs. 9.3 B 6.8 days, p ! 0.0001) than well-
Abstract nourished patients. Conclusions: Patients with malig-
Background/Aims: Malnutrition is common among hos- nancies, inflammatory bowel disease, chronic heart fail-
pitalized patients. We investigated whether certain dis- ure and benign lung diseases need special attention due
eases predispose more frequently for malnutrition than to the high prevalence of malnutrition.
others. Methods: Nutritional state was assessed by clini- Copyright © 2003 S. Karger AG, Basel

cal scores, anthropometry and bioimpedance analysis in


502 consecutively admitted patients in the departments
of internal medicine in two hospitals in Berlin (n = 300, Introduction
university hospital; n = 202, district hospital). The preva-
lence of malnutrition was compared in patient groups Malnutrition is a frequent finding in hospitalized pa-
with a different diagnosis. Results: Malnutrition was tients [1–7]. Data on the prevalence of malnutrition in
present in 24.2% of all patients. A clear association these patients, however, differ considerably according to
between diagnoses and malnutrition was found: the the population investigated and the definitions used.
prevalence of malnutrition was significantly higher in From a number of studies performed during the last 30
malignant than in non-malignant diseases (50.9 vs. years, predominantly in the US and UK, it can be esti-
21.0%, p ! 0.0001). High prevalence rates 1 30% were mated that about 20–50% of all medical and surgical
observed in subgroups of patients with inflammatory patients admitted to hospitals show signs of malnutrition
bowel diseases, chronic heart failure and benign lung [8]. Malnutrition has been found to be associated with

© 2003 S. Karger AG, Basel Dr. med. Matthias Pirlich


ABC 0257–2753/03/0213–0245$19.50/0 Medizinische Klinik und Poliklinik
Fax + 41 61 306 12 34 Universitätsklinikum Charité, Humboldt-Universität zu Berlin
E-Mail karger@karger.ch Accessible online at: DE–10098 Berlin (Germany)
www.karger.com www.karger.com/ddi Tel. +49 30 450 514006, Fax +49 30 450 514923, E-Mail matthias.pirlich@charite.de
Table 1. Participating specialities Table 2. Distribution of diagnoses

n Gender, m/f Age (mean B SD) Diagnoses n %

University hospital Gastrointestinal diseases


Gastroenterology 100 48/52 56.9B16.3 Chronic liver disease 45 9.0
Cardiology 100 74/26 62.0B12.0 Malignancies of liver, pancreas or bile ducts 22 4.4
Rheumatology 100 37/63 52.2B15.5 Gastrointestinal malignancies 17 3.4
Community hospital Gastrointestinal bleeding 16 3.2
Gastroenterology 101 38/63 65.0B15.7 Benign pancreatic or biliary diseases 13 2.6
Cardiology 101 39/62 69.6B14.4 Inflammatory bowel disease 10 2.0
All patients 502 236/266 61.2B16.0 Reflux disease 10 2.0
Other gastroenterology 8 1.6
Gastroenteritis (without salmonella infection) 7 1.4
Other benign intestinal diseases (colonic polyps,
IBS, diverticulosis) 8 1.6
increased morbidity [2, 4], with prolonged hospital stay at All gastrointestinal diseases 156 31.1
substantial extra cost of health care [7, 9, 10], and with
Other internal diseases
increased mortality especially in elderly patients [3, 5,
Coronary heart disease 85 16.9
11]. The association between malnutrition and poor sur- Arrhythmia 40 8.0
vival is especially established in cancer patients [7, 8, 12]. Rheumatoid arthritis 25 5.0
However, despite an increasing number of studies in oth- Heart failure 24 4.8
er countries, the influence of diagnoses on the develop- Systemic infection 20 4.0
Valvular defect 16 3.2
ment of malnutrition in medical patients is unclear. It has
Vasculitis/myositis 15 3.0
been suggested that patients with digestive diseases are at Extraintestinal malignancies 15 3.0
higher risk of developing malnutrition than patients with Systemic lupus erythematodes 13 2.6
other internal diseases [4, 7]. Cerebral ischemia 13 2.6
The primary objectives of this study were (1) to esti- All other diseases 13 2.6
Benign lung diseases 13 2.6
mate the overall prevalence of malnutrition among medi-
Other rheumatology 12 2.4
cal patients and (2) to evaluate the impact of diagnoses on Other cardiology 11 2.2
the prevalence of malnutrition. The study was performed Dermatosclerosis 9 1.8
in two hospitals with different settings (one university Diabetes mellitus 9 1.8
hospital and one community hospital) in the city of Ber- Urology/kidney diseases 7 1.4
Other collagenoses 6 1.2
lin, Germany.
All other internal diseases 346 68.9

Patients and Methods

Patients
The study protocol was approved by the Ethics Committee of the For further classification of patients according to the different
Universitätsklinikum Charité. The nutritional state of 502 patients diseases, the main diagnoses determined at the time of discharge or
consecutively admitted to different specialities of internal medicine referral from the speciality were used. Since there was an overlap of
was studied in two different hospitals: 300 patients were included in diagnoses between the different specialities, the data analysis of the
the University Hospital Charité (specialities: gastroenterology (n = nutritional state was not performed according to the speciality but to
100), cardiology (n = 100), and rheumatology (n = 100)), and 202 the main diagnoses of the patients. The distribution of diagnoses is
patients were included in the District Hospital of Zehlendorf (spe- given in table 2. 156 patients (31.1%) had diseases of the gastrointes-
cialities: gastroenterology (n = 101) and cardiology (n = 101)). tinal tract, and 346 patients (68.9% of all patients) had other internal
Patients were considered eligible for entry if they were over the diseases.
age of 18, were assumed to stay longer than 2 days, and were willing
and able to give written informed consent. Patients admitted to day Assessment of Nutritional State
care units or for observation after endoscopic or other invasive treat- Two investigators (N.L. and M.K) were trained by the principle
ment and those admitted to intensive care units were excluded. The investigator (M.P.) in performing the nutritional assessment. The
number of patients in each speciality, the distribution of gender and nutritional state of the patients was assessed on the day of hospital
the mean age are given in table 1. Patients in the community hospital admission according to clinical scores, anthropometric measure-
were slightly older than patients in the university hospital. ments, and to the results of bioelectrical impedance analysis. The
subjective global assessment was used as the main criterion for the
classification of malnutrition.

246 Dig Dis 2003;21:245–251 Pirlich/Schütz/Kemps/Luhman/Burmester/


Baumann/Plauth/Lübke/Lochs
Subjective Global Assessment (SGA) Table 3. Clinical characterization of the study population
The SGA was established by Detsky et al. [13] and relies primari-
ly on physical signs of malnutrition (loss of subcutaneous fat or mus- Gastrointestinal Other internal
cle mass, edema, ascites) and the patient’s history regarding weight diseases (n = 156) diseases (n = 346)
loss, dietary intake, gastrointestinal symptoms, functional capacity,
and the disease and its relation to nutritional requirements. Each Gender, m/f (%) 71/85 (45.5/54.5) 165/181 (47.7/52.3)
patient was classified as either well nourished (SGA A), moderately Age, years 60.3B16.3 61.6B15.9
or suspected of being malnourished (SGA B) and severely malnour- Height, m 1.68B0.09 1.69B0.09
ished (SGA C). The SGA requires only a few minutes by a trained Weight, kg 69.5B15.8** 74.4B15.0
clinician. Its validity to indicate malnutrition-associated risks of BMI, kg/m2 24.6B4.5* 25.9B4.6
poor outcome has been proven in a number of studies [4, 5, 7, 12, 14]. Length of stay, days1 9.7B7.6 (n = 139) 9.8B6.7 (n = 299)
Since subgroups of patients classified as SGA C were too small, com-
parative analyses were performed between malnourished patients * p ! 0.01; ** p ! 0.001.
classified as either SGA B or C and well-nourished patients classified 1 64/502 (12.7%) patients were transferred and are therefore not
as SGA A. included.

Nutritional Risk Index (NRI)


The NRI was developed by Buzby et al. [15] and is based on the
serum albumin concentration and the ratio of actual to usual weight:
NRI = 1.489 ! serum albumin (g/l) ! 41.7 ! (present weight/usual Statistical Analysis
weight). Calculated values 1 100 indicate good nutritional state, All data are given as mean B SD. Comparison of mean values
97.5–100 mild malnourishment, 83.5 to ! 97.5 moderate malnour- between two groups was performed by Mann-Whitney U-test, and
ishment, and ! 83.5 severe malnourishment. In our analysis, we used differences in frequencies were compared by ¯2 test. p ! 0.05 was
values ! 97.5 for classification of malnutrition. considered to be significant.

Bioelectrical Impedance Analysis (BIA)


BIA was performed by the whole-body tetrapolar contact elec- Results
trode approach applying an alternating electric current of 800 ÌA at
50 kHz (BIA 2000-M, Data Input GmbH, Frankfurt am Main, Ger-
many). Two pairs of current-introducing and voltage-sensing elec- Age and gender distribution as well as body height
trodes were attached to the dorsum of hand and foot of the dominant were not different between patients with digestive dis-
side of the body [16]. Resistance (R), reactance (Xc) and the phase eases and patients with other internal diseases (table 3).
angle (·) were measured. All impedance measurements were taken
Body weight and, thus, body mass index (BMI) were sig-
under standardized conditions [17]. Patients with implanted cardiac
pacemaker or defibrillator were excluded from BIA measurements nificantly lower in patients with digestive than in patients
because of possible interactions with even small electrical currents. with other internal diseases (p ! 0.01). The average length
Total body water was calculated as 0.69 ! H2/R + 0.8 [16] and fat of hospital stay was not different between both patient
free mass = total body water/0.732. Body cell mass (BCM) was calcu- groups.
lated [18] as BCM = fat free mass ! 0.29 ! ln (·). According to
Assessment of malnutrition by the SGA demonstrated
Süttmann [19], a BCM ! 30% of body weight was considered as an
indicator of protein malnutrition. that almost every fourth patient was malnourished (ta-
ble 4). A similar result was obtained by using the NRI or the
Anthropometry body cell mass (!30% of body weight) as criteria for malnu-
Body height was measured without shoes to the nearest 0.5 cm trition. Other parameters frequently used for the diagnosis
with a stadiometer. Weight was measured using calibrated Seca chair
of malnutrition such as BMI, upper arm anthropometry or
scales and compared with the body weight 6 months prior to admis-
sion to calculate weight loss. Midarm circumference was measured albumin resulted in lower incidence values of malnutrition
with a tape measure. Triceps skinfold thickness was measured using a ranging from 3.8% for BMI to 14.5% for albumin.
Lange caliper (Holtain Ltd, Crymych, Dyfed, UK). The average of The incidence of malnutrition as defined by SGA was
three measurements at each site was used for the calculation of arm not significantly different between patients with gastroin-
muscle area and arm fat area according to Gurney and Jelliffe [20].
testinal diseases and other medical patients. In contrast,
Measured arm muscle and arm fat area were compared with refer-
ence data [21], and values below the 10th percentile were considered the NRI indicated a significantly higher prevalence of
as an indicator of malnutrition. malnutrition among patients with digestive diseases when
compared with the other patients. Since the NRI includes
Albumin serum albumin, it is not surprising that low serum albu-
Serum albumin was measured in the clinical chemistry laboratory
min levels were also more frequently observed in gastroin-
by an automated analyzer using standard procedures, and values
! 3.5 g/dl were considered as indicators of impaired protein synthe- testinal patients. Regarding BMI and weight loss 110%
sis. during the last 6 months prior to admission, there were no

Hospital Malnutrition Dig Dis 2003;21:245–251 247


Table 4. Prevalence of malnutrition in all patients and in subgroups of patients with gastrointestinal vs. other internal
diseases (% values in parentheses)

All patients Gastrointestinal Other internal


(n = 502) diseases (n = 156) diseases (n = 346)

Nutritional scores
SGA B+C 121/501 (24.2) 45/156 (28.8) 76/345 (22.0) ns
Nutritional risk index ! 97.5 107/435 (24.6) 52/138 (37.7) 55/297 (18.5) p ! 0.001
Albumin ! 3.5 g/dl 63/434 (14.5) 34/138 (24.6) 29/296 (9.8) p ! 0.001
Anthropometry
BMI ! 18.5 kg/m2 19/501 (3.8) 8/156 (5.1) 11/345 (3.2) ns
Weight loss 1 10% body weight 48/501 (9.6) 17/156 (10.9) 31/345 (9.0) ns
AMA ! 10th percentile 51/496 (10.3) 23/153 (15.0) 28/343 (8.2) p = 0.020
AFA ! 10th percentile 55/495 (11.1) 22/153 (14.4) 33/342 (9.6) ns
Impedance analysis
BCM ! 30% body weight 123/453 (27.2) 43/147 (29.3) 80/306 (26.1) ns

SGA = Subjective global assessment; AMA = arm muscle area; AFA = arm fat area; BCM = body cell mass.
Statistics: ¯2 test, gastrointestinal diseases compared to other diseases.

were classified as SGA B or C than patients with non-


***
60 malignant diseases (fig. 1). Patients with malignancies
Prevalence of malnutrition (%)

were then divided into three subgroups: malignancies of


50
liver, pancreas or bile ducts (n = 22), malignancies of the
40 gastrointestinal tract (n = 17), and extraintestinal malig-
nancies (n = 15). According to the SGA, the highest preva-
30
lence of malnutrition of 63.6% was observed in patients
20 with malignancies of the liver, pancreas or bile ducts,
while patients with gastrointestinal or extraintestinal ma-
10
lignancies had a similar prevalence of 41.2 and 40.0%,
0 respectively (fig. 2).
nt nt al rs Among patients with benign diseases (table 5) the high-
na es na es tin es he
l g
i as ig te
s s ot est prevalence of malnutrition was observed in patients
a
m ise al as in sea
- m ise tro di
d n d s with chronic inflammatory bowel disease (40%), chronic
no ga
heart failure (37.5%), benign lung diseases (30.8%),
chronic liver diseases (28.9%) and rheumatoid arthritis
Fig. 1. Prevalence of malnutrition as diagnosed by SGA in malignant (28%). In contrast, a rather low percentage of patients
(n = 54) vs. non-malignant (n = 448) diseases, and in gastrointestinal with benign digestive diseases was classified as malnour-
diseases vs. other internal diseases; *** p ! 0.0001.
ished (ranging from 10 to 15.4%).
Finally, all patients were classified into two groups
with either good nutritional state or malnutrition accord-
differences observed between gastrointestinal patients ing to SGA (table 6). There were no differences between
and other medical patients. Regarding the results of upper both groups regarding the gender distribution. However,
arm anthropometry, we found that significantly more malnourished patients were significantly older than well-
patients with gastrointestinal disease had a low arm mus- nourished patients. Malnutrition was also associated with
cle area than the other patients, but frequencies for arm a significantly higher length of hospital stay. Further anal-
fat area were not significantly different. ysis demonstrated that NRI, albumin, BMI and parame-
When all patients were classified according to malig- ters of body composition obtained either by anthropome-
nant (n = 54) and non-malignant disease (n = 448), we try or BIA all yielded significantly lower values in patients
found that significantly more patients with malignancies with malnutrition than in well-nourished subjects.

248 Dig Dis 2003;21:245–251 Pirlich/Schütz/Kemps/Luhman/Burmester/


Baumann/Plauth/Lübke/Lochs
Table 5. Frequency of malnutrition in
selected benign diseases Total SGA B+C SGA B+C, %

Inflammatory bowel disease 10 4 40.0


Heart failure 24 9 37.5
Benign lung diseases 13 4 30.8
Chronic liver disease 45 13 28.9
Rheumatoid arthritis 25 7 28.0
Valvular defect 16 4 25.0
Systemic lupus erythematodes 13 3 23.1
Cerebral ischemia 13 3 23.1
Arrhythmia 40 9 22.5
Systemic infection 20 4 20.0
Vasculitis/myositis 15 3 20.0
Coronary heart disease 85 14 16.5
Benign pancreatic or biliary diseases 13 2 15.4
Gastrointestinal bleeding 16 2 12.5
Other benign gastrointestinal diseases 16 2 12.5
Reflux disease 10 1 10.0
Other cardiology 11 1 9.1
All other diseases 13 1 7.7
Other rheumatology 12 0 0

Note: This table includes only disease groups with n 6 10.

Discussion Table 6. Clinical characteristics and body composition in patients


with SGA A compared to SGA B+C
In this study the nutritional state of 502 hospitalized
SGA A SGA B+C p value
medical patients with a wide variety of disease states was (n = 380) (n = 121)
prospectively assessed. Using the SGA as the main criteri-
on, we found that almost every fourth patient admitted to Age, years 58.3B15.6 70.0B13.6 0.000
hospital was malnourished. This prevalence rate appears Gender, m/f 184/196 52/69 ns
Nutritional score
to be high, especially if one considers the fact that over-
Nutritional risk index 105.8B7.6 93.6B9.9 0.000
but not undernutrition is the main nutrition-related Albumin, g/d l4.3B0.5 3.7B0.6 0.000
health problem in Germany, indicated by a high and Anthropometry
obviously increasing average BMI in the healthy popula- BMI, kg/m2 26.6B4.3 22.1B3.6 0.000
tion, especially in the age group over 50 years [22]. Body weight, kg 76.2B14.8 62.21B12.0 0.000
AMA, mm2 5,584B1,499 4,421B1,121 0.000
Prevalence rates for malnutrition from other countries
AFA, mm2 2,829B1,320 1,650B854 0.000
reported during the last 15 years ranged from 20% [3, 6] Impedance analysis
up to 62% [4]. The majority of studies on medical BCM, kg 26.1B6.4 19.3B4.7 0.000
patients, however, reported malnutrition rates around Outcome
40% [1, 2, 5, 7, 9]. However, there are no representative LOS, days 9.3B6.8 13.1B8.1 0.000
LOS, median (range) 7.0 (2–55) 12.0 (3–49)
data from Germany. Therefore, we cannot answer the
question whether or not the nutritional state of hospital- SGA = Subjective global assessment; AMA = arm muscle area;
ized patients in Germany might have changed along with AFA = arm fat area; BCM = body cell mass; LOS = length of hospital
changes of medical care or the demographic transition stay.
during the last decades.
Differences in prevalence data on malnutrition among
different studies not only depend on the population select-
ed or on the institutional setting, but also on the different
diagnostic criteria used for the definition [11]. This was
highlighted in a very detailed study on 155 non-surgical

Hospital Malnutrition Dig Dis 2003;21:245–251 249


Malignancies: liver, pancreas, bile ducts n=22 63.6
Extraintestinal malignancies n=15 42.9
Gastrointestinal malignancies n=17 41.2
Inflammatory bowel disease n=10 40.0
Heart failure n=24 37.5
Benign lung diseases n=13 30.8
Chronic liver disease n=45 28.9
Rheumatoid arthritis n=25 28.0
Cerebral ischemia n=13 23.1
Systemic infection n=20 20.0

Benign pancreatic or biliary disease n=13 15.4

0 10 20 30 40 50 60 70
Prevalence of malnutrition: SGA B+C (%)
Fig. 2. Prevalence of malnutrition in select-
ed diagnoses.

patients applying four different established clinical scores diseases of the digestive system than to some other medi-
to the same patients [4]. The authors found that depend- cal conditions. This assumption appears plausible, since
ing on the instrument used for diagnosis, between 40 and impaired digestion per se can be expected to influence the
62% of their patients were classified as malnourished. nutritional state. In fact, Naber et al. [4] found that 61%
The question, however, which score or which diagnostic of their gastrointestinal patients but only 30% of other
instrument might provide the best description of the medical patients were malnourished. ln the study of
nutritional state, is still open [23]. Waitzberg et al. [7], 61% of all patients with gastrointesti-
We decided to choose the SGA as the primary diagnos- nal disorders were also found to be malnourished while on
tic criterion in our study because this score is simple, inex- average 48% of the patients were found to be malnour-
pensive, non-invasive and can be performed at the bed- ished.
side in a very short period of time. The SGA has also been In contrast, we found a much lower frequency of mal-
proven to be of prognostic relevance in a number of differ- nutrition of 29% in gastrointestinal patients. Further-
ent clinical settings, especially indicating an increased more, our gastrointestinal patients as a whole group were
mortality of patients with internal diseases [4, 5, 12, 14, not different from patients with other internal diseases, so
24]. In our study, the SGA yielded results similar to the that our data obviously do not support the assumption of
NRI, and patients who were classified as malnourished patients with digestive diseases being at higher risk for
according to SGA also had significantly impaired body malnutrition. To better understand possible reasons for
composition and plasma albumin compared to patients these contradictory results, we performed more detailed
classified as well nourished. We also found that malnour- analyses in subgroups of patients. Not unexpectedly, we
ished patients were on average 12 years older and had a found that the malnutrition rate in patients with malig-
40% longer hospital stay than well-nourished patients. nancies was more than twice as high as in benign medical
This is in accordance with studies in other countries [7, 9] conditions. The highest prevalence of 64% was seen
and further supports the assumptions that increasing age among patients with malignancies of the liver, pancreas or
is a risk factor for the development of malnutrition [11], bile ducts. A subgroup analysis of benign diseases demon-
and that malnutrition, on the other hand, is associated strated high prevalence rates of 130% in patients with
with an impaired clinical course and extra costs of health inflammatory bowel disease, heart failure and benign lung
care [4, 7–9]. disease (fig. 2). In contrast, benign pancreatic or biliary
Two more recent studies from the Netherlands [4] and diseases, gastrointestinal bleeding, reflux disease or other
from Brazil [7] also using the SGA for diagnosis suggested benign digestive diseases were not associated with higher
that malnutrition might show a stronger association to malnutrition rates, but – compared with other medical

250 Dig Dis 2003;21:245–251 Pirlich/Schütz/Kemps/Luhman/Burmester/


Baumann/Plauth/Lübke/Lochs
conditions – demonstrated rather under average preva- to hospitalization but more frequently directed to ambu-
lence values. latory treatment. Therefore, one can expect that in the
In the cited studies from the Netherlands [4] and Brazil next years the prevalence of malnourished hospitalized
[7], the gastrointestinal patients were not subclassified. patients will increase in our country. Moreover, our data
Therefore, we can only speculate that the higher malnutri- show that some patient groups like malignancies, inflam-
tion rates of gastrointestinal patients in these studies com- matory bowel disease, chronic heart failure, or benign pul-
pared with our results might be caused by the inclusion of monary diseases have a specifically high prevalence of
more severely sick patients with inflammatory bowel dis- malnutrition. In these patient groups, special care has to
eases, more patients with maldigestion or malabsorption, be taken in the evaluation of the nutritional state and
or by inclusion of a higher percentage of malignancies of nutritional support. It is, therefore, important to improve
the digestive system. the awareness of physicians, nurses and other profession-
From previous studies we know that malnutrition in als for this problem by better education in nutritional
hospitalized patients is frequently underestimated by the assessment.
medical staff [2], and even simple diagnostic procedures
such as the body weight are not performed in a relevant
number if not in the majority of patients [2, 7]. Since the Acknowledgement
health system in Germany is currently under high eco-
Dr. M. Pirlich was supported by the Else Kröner-Fresenius-Stif-
nomic pressure it can be expected that in the future more
tung, Bad Homburg, Germany.
sick patients will be hospitalized and a number of patients
included in the present study will no longer be subjected

References

1 Coats KG, Morgan SL, Bartolucci AA, Wein- 10 Tucker HN, Miguel SG: Cost containment 18 Pirlich M, Schütz T, Spachos T, Ertl S, Weiss
sier RL: Hospital-associated malnutrition: A through nutrition intervention. Nutr Rev 1996; M-L, Lochs H, Plauth M: Bioelectrical impe-
reevaluation 12 years later. J Am Diet Assoc 54:111–121. dance analysis is a useful bedside technique to
1993;93:27–33. 11 Pirlich M, Lochs H: Nutrition in the elderly. assess malnutrition in cirrhotic patients with
2 McWhirter JP, Pennington CR: Incidence and Best Pract Res Clin Gastroenterol 2001;15: and without ascites. Hepatology 2000;32:
recognition of malnutrition in hospital. BMJ 869–884. 1208–1215.
1994;308:945–948. 12 Persson C, Sjöden PO, Glimelius B: The Swed- 19 Süttmann U, Ockenga J, Selberg O, Hooge-
3 Cederholm T, Jägrén C, Hellström K: Outcome ish version of the patient-generated global as- straat L, Deicher H, Müller MJ: Incidence and
of protein-energy malnutrition in elderly medi- sessment of nutritional status: Gastrointestinal prognostic value of malnutrition and wasting in
cal patients. Am J Med 1995;98:67–74. vs. urological cancers. Clin Nutr 1999;18:71– human immunodeficiency virus-infected out-
4 Naber THJ, Schermer T, de Bree A, Nusteling 77. patients. J Acquir Immune Defic Syndr Hum
K, Eggink L, Kruimel JW, Bakkeren J, van 13 Detsky AS, McLaughlin JR, Baker JP, John- Retrovirol 1995;8:239–246.
Heereveld H, Katan MB: Prevalence of malnu- ston N, Whittaker S, Mendelson RA, Jeejeeb- 20 Gurney JM, Jelliffe DB: Arm anthropometry in
trition in nonsurgical hospitalized patients and hoy KN: What is subjective global assessment nutritional assessment: nomogram for rapid
its association with disease complications. Am of nutritional status? J Parenter Enteral Nutr calculation of muscle circumference and cross-
J Clin Nutr 1997;66:1232–1239. 1987;11:8–13. sectional muscle and fat mass. Am J Nutr 1973;
5 Sacks GS, Dearman K, Replogle WH, Cora 14 Lawson JA, Lazarus R, Kelly JJ: Prevalence 26:912–915.
VL, Meeks M, Canada T: Use of subjective and prognostic significance of malnutrition in 21 Frisancho AR: New norms of upper limb fat
global assessment to identify nutrition-associ- chronic renal insufficiency. J Ren Nutr 2001; and muscle areas for assessment of nutritional
ated complications and death in geriatric long- 11:16–22. status. Am J Clin Nutr 1981;34:2540–2545.
term care facility residents. J Am Coll Nutr 15 Buzby GP, Knox LS, Crosby LO, Eisenber JM, 22 Pirlich M, Schwenk A, Müller M, Ockenga J,
2000;19:570–577. Haakenson C, McNeal GE, Page CP, Peterson Schmidt S, Schütz T, Selberg O, Volkert D:
6 Malnutrition Prevalence Group: Prevalence of OL, Reinhardt GF, Williford W: Study proto- Leitlinie enterale Ernährung, Ernährungssta-
malnutrition on admission to four hospitals in col: A randomized clinical trial of total paren- tus. Akt Ernähr Med 2003;28(suppl 1):10–25.
England. Clin Nutr 2000;19:191–195. teral nutrition in malnourished surgical pa- 23 Klein S, Kinney J, Jeejeebhoy K, Alpers D,
7 Waitzberg DL, Caiaffa WT, Correia MITD: tients. Am J Clin Nutr 1988;47:357–365. Hellerstein M, Murray M, Twomey P: Nutri-
Hospital malnutrition: the Brazilian National 16 Kushner RF, Schoeller DA: Estimation of total tion support in clinical practice: Review of
Survey (IBRANUTRI): A study of 4,000 pa- body water by bioelectrical impedance analy- published data and recommendations for fu-
tients. Nutrition 2001;17:573–580. sis. Am J Clin Nutr 1986;44:417–424. ture research directions. Am J Clin Nutr 1997;
8 Pirlich M, Luhmann N, Schütz T, Plauth M, 17 Pirlich M, Biering H, Gerl H, Ventz M, 66:683–706.
Lochs H: Mangelernährung bei Klinikpatien- Schmidt B, Ertl S, Lochs H: Loss of body cell 24 Pirlich M, Schütz T, Gastell S, Lochs H: Mal-
ten: Diagnostik und klinische Bedeutung. Akt mass in Cushing’s syndrome: Effect of treat- nutrition affects long-term prognosis in hospi-
Ernähr Med 1999;24:260–266. ment. J Clin Endocrinol Metab 2002;87:1078– talized patients. Gastroenterology 2002;122
9 Robinson G, Goldstein M, Levine GM: Impact 1084. (suppl 1):A363.
of nutritional status on DRG length of stay. J
Parenter Enteral Nutr 1987;11:49–51.

Hospital Malnutrition Dig Dis 2003;21:245–251 251


Original Paper

Dig Dis 2003;21:252–257


DOI: 10.1159/000073343

Sugar Intake, Taste Changes and Dental Health


in Crohn’s Disease
Tatjana Schütz a Clemens Drude b Erika Paulisch b Klaus-Peter Lange b
Herbert Lochs a
a Medizinische Klinik mit Schwerpunkt Gastroenterologie, Hepatologie und Endokrinologie, Universitätsklinikum

Charité Campus Mitte, Berlin und b Zentrum für Zahnmedizin, Abteilung für zahnärztliche Prothetik und
Alterszahnmedizin, Universitätsklinikum Charité Campus-Virchow, Berlin, Deutschland

Key Words ar consumption and insufficient oral hygiene seem to


Crohn’s disease W Taste changes W Sugar intake W Zinc W cause the higher caries prevalence. Obviously, patients
Dental caries with CD belong to a high-risk group, and preventive mea-
sures should be taken early in the course of the disease.
Copyright © 2003 S. Karger AG, Basel

Abstract
Background: An increased intake of sucrose has been Introduction
reported in patients with Crohn’s disease (CD). Since
subclinical zinc deficiency reduces taste perception for Malnutrition is frequently observed in patients with
sweet, we investigated taste perception, sucrose intake Crohn’s disease (CD) and manifests itself in weight loss
and plasma zinc levels as well as dental status in CD and nutrient deficiencies [1]. Special importance is at-
patients. Methods: Carbohydrate intake and plasma zinc tached to zinc and its role in taste perception. In this con-
levels were assessed in 24 CD patients and 24 age- text, Henkin et al. [2] and Solomons et al. [3] described
matched controls (Con). Taste threshold for sucrose, oral that reducing zinc triggers a reversible hypogeusia.
hygiene and caries prevalence were evaluated. Results: In the 1970s, studies on the dietary habits of CD
In CD a higher sucrose intake (CD 107.1 B 27.7 vs. Con patients drew attention to higher intakes of carbohy-
71.9 B 13.7 g/day; p ! 0.001), a higher taste threshold for drates, sugar or added sugar before or after the onset of
sweet (CD 7.31 vs. Con 2.91 g/l; p ! 0.001) and lower plas- the disease compared to patients with ulcerative colitis [4]
ma zinc levels (CD 11.5 B 1.5 vs. Con 13.5 B 2.0 Ìmol/l; or healthy controls [5–11]. As one possible cause for the
p ! 0.001) were found. API was poor (CD 85.4 B 23.6, Con increased sugar consumption, an impaired gustatory
31.8 B 24.1, p ! 0.001) and correlated with sucrose intake function for the sweet taste has been discussed. However,
(p ! 0.01). Caries prevalence was increased in patients the hypothesis that an excessive intake of refined carbohy-
with longer disease (1 3 years) (DMFT index: 13 years drates may be the result of a higher taste threshold for
15.6 B 5.7 vs. ! 3 years 9.5 B 4.3; p ! 0.05). Conclusion: sweet was refuted by three studies [12–14]. A higher
Dental status in CD patients is poor. Both increased sug- threshold for sweet was only observed in patients with

© 2003 S. Karger AG, Basel Dr. Tatjana Schütz


ABC 0257–2753/03/0213–0252$19.50/0 Universitätsklinikum Charité
Fax + 41 61 306 12 34 Medizinische Klinik mit Schwerpunkt Gastroenterologie, Hepatologie und Endokrinologie
E-Mail karger@karger.ch Accessible online at: Schumannstrasse 20/21, DE–10098 Berlin (Germany)
www.karger.com www.karger.com/ddi Tel. +49 30 450 514059, Fax +49 30 450 514923, E-Mail elke-tatjana.schuetz@charite.de
active disease (Crohn’s disease activity index – CDAI Table 1. Characteristics of CD patients and healthy controls (mean
1200) and patients with CD for more than 2 years [15]. B SD)
Studies on dental status imply that CD patients have a
CD patients Controls
high caries frequency and activity [16, 17]. The reason for
the increased caries prevalence is unclear but as the most Number 24 24
likely link the dietary regimen can be suggested. Gender, m/f 10/14 12/12
In this overlapping field of gastroenterology, otorhino- Age, years 36.2B15.0 35.4B10.8
Smoker/non-smoker 13/11 12/12
laryngology and dentistry this interdisciplinary study was
Crohn’s disease activity index 153B98 –
conducted to examine the association between serum zinc Disease duration, years 5.0B3.9 –
concentrations and the taste threshold for sweet, as well as
the intake of sucrose and carbohydrates and its effect on
the risk of caries and caries prevalence in CD patients
compared to healthy controls.
Table 2. Mono-, di- and polysaccharide intake (mean B SD) in CD
patients and healthy controls
Methods
CD patients Controls
Patients
Twenty-four CD diagnosed by generally accepted criteria [18] Monosaccharides, g/day 43B15 46B14
and 24 age-matched healthy subjects participated in the study (ta- Glucose, g/day 20B7 20B5
ble 1). Disease activity was assessed according to the CDAI [19]. Dis- Disaccharides, g/day 122B32*** 87B16
ease was active (CDAI 1 150) in 50% of the patients. The duration of Sucrose, g/day 107B28*** 72B74
disease ranged between 1 and 18 years with a median of 4 years. Polysaccharides, g/day 195B27* 202B21
Eleven patients were current smokers. Patients received standard Total carbohydrates, g/day 367B64 332B70
medical treatment including corticosteroids (n = 11), mesalazine (n =
17), azathioprine (n = 2), and metronidazole (n = 1) as mono- or * p ! 0.05; *** p ! 0.001.
combination therapy. Two patients were irregularly supplemented
with vitamin A (n = 1) or zinc (n = 1). Patients with severe secondary
diseases, otorhinolaryngologic and neurologic disorders, or diseases
of the oral cavity as well as patients taking medication that influences
taste perception (diuretics, antidiabetics, antihypertensives, anti-
rheumatics, psychoactive drugs) were excluded from the study. The
study protocol was approved by the local ethical committee and all typical site for the perception of the sweet taste, and the subjects were
patients and healthy subjects gave their written informed consent. asked to identify the test stimulus. After each tasting the subjects
expectorated the samples and rinsed their mouth with water. Taste
Procedure threshold was defined as the lowest concentration which the subject
Sucrose and carbohydrate intake of the present daily diet were correctly identified.
assessed by a food-frequency questionnaire in which the intake of 64 Plasma zinc concentration: Metal-free plastic syringes and con-
food items and 9 beverages was documented in three categories: regu- tainers (Sarstedt, Nümbrecht, Germany) were used to collect blood
larly (once to several times a day), rarely (once a week to once a for the zinc measurement. Zinc and other laboratory parameters (al-
month), and never. Standard portion sizes were applied to the food bumin, hematocrit) were determined by standard methods.
categories and glucose and carbohydrate content was then calculated Dental and oral health: The prevalence of caries was assessed by
using the EBIS software (Forschungszentrum für Ernährung in Prä- determining the percentage of decayed, missing and filled teeth
vention und Therapie, Hohenheim GmbH, Stuttgart, Germany), (DMFT index) [22]. Low values indicate good dental health. Oral
which is based on the German Food and Nutrition Database (BLS, hygiene was assessed by using the API index (approximal plaque
version II.1). index), which is calculated as the percentage of contaminated ap-
Taste threshold: Olfactometric and gustometric testing was per- proximal sites [23]. Analogous to the DMFT index, low values point
formed in the morning at least 1 h after breakfast by a standard pro- to sufficient oral hygiene.
cedure [20]. Shortly, subjects were asked to restrain from eating,
smoking and using toothpaste during that period. Prior to the gus- Statistics
tometric measurements, anosmia was excluded by testing the smell Results are given as mean B SD. Means were compared by
stimuli camphor, vanillin, dichlorethane, menthol and formic acid in Mann-Whitney U-test and frequencies by the ¯2 test with p ! 0.05 as
different concentration ranges using the sniff-bottle technique [20]. level of significance. All calculations were performed by the comput-
The taste threshold for sweet was quantitatively determined with 8 er software SPSS (version 10.0, SPSS Inc., Chicago, Ill., USA).
sucrose solutions in graded concentrations (1–256 g/l in deionized
water) using the pipette method [21]. 0.5 ml of the solutions in
ascending order was given drop by drop on the tip of the tongue, the

Sugar Intake in Crohn’s Disease Dig Dis 2003;21:252–257 253


60 CD Controls
200 **

50
Sucrose intake (g/day)

***

Number of subjects (%)


40
***
100

30
***

20

0
Controls CD CD CD 10
(n = 24) (n = 6) (n = 12) (n = 6)
<3 years 3-6 years >6 years
0
1 2 4 8 16 32
Fig. 1. Sucrose intake in healthy controls and CD patients in groups Sucrose (g/l)
according to length of disease. Box plots with horizontal bars indicat-
ing median values, boxes indicating the 25th centiles, error bars indi-
cating the 95% confidence interval and [ indicating values outside Fig. 2. Proportion of CD patients compared to healthy controls, who
the 95th centile. ** p ! 0.01. detected the tested sucrose concentration. *** p ! 0.001.

Results tion, was excluded in the study population. Taste thresh-


old for sweet was significantly higher in CD patients com-
Eating Habits, Sucrose and Carbohydrate Intake. Eat- pared to healthy controls (7.31 vs. 2.91 g/l, p ! 0.001). In
ing habits differed between patients and controls. Signifi- contrast to 58.3% of the control subjects, none of the CD
cantly more CD patients abstained from alcoholic bever- patients were able to recognize the two lowest sucrose con-
ages (wine: 50.0 vs. 12.5%; beer: 62.5 vs. 25.0%; p ! 0.01) centrations (fig. 2). There was no difference in taste detec-
and dried fruits (83.3 vs. 50%, p ! 0.05) compared to tion comparing patients with active (CDAI 1150) to inac-
healthy controls. Total carbohydrate intake was not dif- tive (CDAI !150) disease. There was no correlation
ferent between CD patients and healthy controls (table 2). between the taste threshold for sweet and sucrose intake
CD patients, however, consumed 40% more disaccha- in either group.
rides than healthy controls, which was due to a signifi- Plasma Zinc Concentration. Since plasma zinc is bound
cantly higher sucrose intake (p ! 0.001) (fig. 1). This dif- to albumin, we also measured albumin concentrations.
ference remained significant when sucrose intake was cal- Plasma zinc concentrations significantly correlated with
culated per kilogram body weight (CD 1.72 B 0.62 vs. albumin concentrations in both CD patients and controls
control 1.03 B 0.26 g/kg b.w.; p ! 0.001). Surprisingly, (CD: r2 = 0.150, control: r2 = 0.052, both p ! 0.001). The
patients with shorter disease duration had a significantly plasma zinc values of 5 CD patients (20.8%) and 2 con-
higher sucrose intake than patients with a longer lasting trol subjects (8.3%) were below the normal range (10.6–
disease (fig. 1). The higher sucrose intake was due to a 17.9 Ìmol/l). The mean value for the group of CD patients
more frequent consumption of sweet-tasting food prod- was significantly lower than that of the healthy controls
ucts, such as pudding (25 vs. 0%, p ! 0.01), honey and jam (11.5 B 1.5 vs. 13.5 B 2.0 Ìmol/l; p ! 0.001) (fig. 3). Nei-
(70.8 vs. 37.5%, p ! 0.05), chocolate (58.3 vs. 16.7%, p ! ther in CD patients nor in healthy controls were plasma
0.01), and cake (50.0 vs. 12.5%, p ! 0.01). zinc levels correlated with sucrose intake.
Olfactometry and Gustometry. All subjects could smell Dental and Oral Health. Dental health was impaired in
at least one concentration level of each odorant so that CD patients compared to controls. However, this became
anosmia, which would negatively influence taste percep- apparent only with longer lasting disease with an in-

254 Dig Dis 2003;21:252–257 Schütz/Drude/Paulisch/Lange/Lochs


30 *
20 ***
Plasma zinc concentration (µmol/l)

18 *

DMFT-index (%)
16 20

14

12
10
10

0
6
Controls CD CD CD
Controls CD
( = 24) (n = 6) (n = 12) (n = 6)
(n = 24) (n = 24)
<3 years 3-6 years >6 years

Fig. 3. Plasma zinc concentration in healthy controls and CD Fig. 4. Prevalence of caries (DMFT index) in healthy controls and in
patients. Box plots as explained in figure 1. *** p ! 0.001. CD patients in groups according to length of disease. Box plots as
explained in figure 1. * p ! 0.05.

creased DMFT index indicating higher caries prevalence gustin/carbonic anhydrase IV, which is decreased in pa-
(fig. 4). Oral hygiene was insufficient in CD patients with tients with hypogeusia and dysgeusia [2]. It seems to exert
a significantly higher API index compared to controls its action as a trophic factor on the taste bud stem cells
(CD: 85.5 B 23.6% vs. control: 31.8 B 24.1%, p ! 0.001). and so promotes growth and development of taste buds.
The API index was significantly correlated to sucrose So far there have been no data on gustin/carboanhydrase
intake in both groups (CD: r2 = 0.0104; control: IV concentrations in saliva of CD patients with impaired
r2 = 0.0059; p ! 0.01). 25% of CD patients vs. 4% of con- taste acuity. Moreover, a possible influence of medication
trols did not regularly go to the dentist’s. on gustatory function must be assumed.
Our data do in fact demonstrate higher sugar intake,
higher caries prevalence and lower zinc plasma levels in
Discussion CD patients. However, these changes appear not to be
related to each other. The taste threshold for sweet was
Several studies have shown a higher sugar intake in CD also significantly higher in CD patients than healthy con-
patients when compared to the healthy population even trols. Only Lederer et al. [15] could also measure a signifi-
before the onset of the disease [4–11]. The reason for this cantly higher taste threshold for sweet in patients with
dietary habit is unclear. However, a correlation with the active disease (CDAI 1200), whereas other studies only
decreased zinc status was suspected since zinc is an reported a significantly higher threshold for salt taste [13]
important trace element for the taste perception sweet [2, and for acid taste [14]. Solomons et al. [3] found a signifi-
3]. Furthermore, a higher caries prevalence would be cantly lower overall taste detection score, which com-
expected in CD patients as a consequence of the higher prised all four basic taste modalities, whereas Kasper et al.
sugar consumption. [12] could not detect any differences.
Zinc deficiency has frequently been described in CD The question has to be raised why a correlation be-
patients [3, 24, 25], and alterations in taste acuity have tween low plasma zinc levels and increased taste thresh-
been observed in these patients [13, 14, 24]. The mecha- old for sweet could not have been found. This might be
nism of chemosensory dysfunction is not well understood due to the fact that plasma zinc levels do not necessarily
but the key role in the influence of zinc on taste perception reflect the zinc status but could rather be dependent on
is attributed to the zinc-dependent parotid saliva enzyme inflammatory processes. This might, however, not ex-

Sugar Intake in Crohn’s Disease Dig Dis 2003;21:252–257 255


plain our results since there was no difference in the taste tobacilli [26], which may result from high sucrose intake
threshold between patients with active disease versus in combination with insufficient oral hygiene. Factors of
patients with quiescent disease. It would be interesting to the development of dental caries resulting from CD per se
investigate the effect of zinc supplementation on the taste such as altered tooth structure [17], salivary flow rate [26]
threshold in CD patients and consequently on sugar and salivary constituency in regard to antimicrobial pro-
intake. To our knowledge this has not yet been adequately teins [27] have not been proven.
investigated. The poor dental status was correlated to insufficient
The higher sugar intake was not accompanied by a oral hygiene and the lack of regular visits to the dentist’s
higher total carbohydrate intake in our study but was rath- in CD patients. This clearly indicates that CD patients are
er due to the consumption of sweets. This is very well in a risk group for poor dental health, and therefore mea-
line with former studies, using different methods to evalu- sures should be taken early in the course of the disease to
ate dietary habits of CD patients [4–11]. This specific eat- avoid deterioration. Since eating habits are difficult to
ing of sweets makes it unlikely that CD patients substitute influence, training for dental hygiene and specific atten-
carbohydrates for a possible fat intolerance since it would tion to regular dentist visits should be taken in CD
be expected that the intake of all carbohydrates would be patients.
increased.
One consequence of high sugar intake is an increase in
the incidence of caries. Dental caries is a multifactorial Conclusion
disease in which the effects of the two main initiating fac-
tors dental plaque and fermentable carbohydrates are The dental status in CD patients was found to be poor.
modified by various endogenic and exogenic factors. CD The taste threshold for sweet was increased in CD pa-
patients are reported to have a higher incidence of dental tients; an association to plasma zinc concentrations and
caries than healthy controls [16, 17]. In our study the sucrose intake, however, could not be proven. Both in-
extent of caries increased with the duration of disease. creased sugar consumption and insufficient oral hygiene
This could be due to long-lasting high sugar intake but seem to cause the higher caries prevalence. Obviously,
also to nutritional deficiencies like calcium and fluoride CD patients belong to a high-risk group, and preventive
deficiency. A further explanation could be high levels of measures should be taken early in the course of the dis-
cariogenic bacteria in saliva, such as streptococci and lac- ease.

References

1 Geerling BJ, Badart-Smook A, Stockbrügger 7 Silkoff K, Hallak A, Yegena L, Rozen P, May- 12 Kasper H, Sommer H, Wild M: Ge-
RW, Brummer RJ: Comprehensive nutritional berry JF, Rhodes J, Newcombe RG: Consump- schmacksschwellen bei Kranken mit Enteritis
status in patients with long-standing Crohn’s tion of refined carbohydrate by patients with regionalis (Morbus Crohn). Akt Ernähr Med
disease currently in remission. Am J Clin Nutr Crohn’s disease in Tel-Aviv-Yafo. Postgrad 1980;5:196–198.
1998;67:919–926. Med J 1980;56:842–846. 13 Tiomny E, Horwitz C, Graff E, Rozen P, Gilat
2 Henkin RI, Martin BM, Agarwal RP: Efficacy 8 Mayberry JF, Rhodes J, Allan R, Newcombe T: Serum zinc and taste acuity in Tel-Aviv
of exogenous oral zinc treatment of patients RG, Regan GM, Chamberlain LM, Wragg KG: patients with inflammatory bowel disease. Am
with carbonic anhydrase IV deficiency. Am J Diet in Crohn’s disease. Two studies of current J Gastroenterol 1982;77:101–104.
Med Sci 1999;318:392–405. and previous habits in newly diagnosed pa- 14 Penny WJ, Mayberry JF, Aggett PJ, Gilbert JO,
3 Solomons NW, Rosenber IH, Sandstead HH tients. Dig Dis Sci 1981;26:444–448. Newcombe RG, Rhodes J: Relationship be-
Vo-Khactu KP: Zinc deficiency in Crohn’s dis- 9 Järnerot G, Järnmark I, Nilsson K: Consump- tween trace elements, sugar consumption and
ease. Digestion 1977;16:87–95. tion of refined sugar by patients with Crohn’s taste in Crohn’s disease. Gut 1983;24:288–292.
4 Mayberry JF, Rhodes J, Newcombe RG: In- disease, ulcerative colitis, or irritable bowel 15 Lederer PC, Cidlinsky K, Kobal G, Lux G:
creased sugar consumption in Crohn’s disease. syndrome. Scand J Gastroenterol 1983;18: Geschmacksstörungen bei M. Crohn Patien-
Digestion 1980;20:323–326. 999–1002. ten. Elektrogustometrie und chemische Ge-
5 Martini GA, Brandes JW: Increased consump- 10 Tragnone A, Valpiani D, Miglio F, Elmi G, schmacksprüfung. Z Gastroenterol 1985;23:
tion of refined carbohydrates in patients with Bazzocchi G, Pipitone E, Lanfranchi GA: Di- 470.
Crohn’s disease. Klin Wochenschr 1976;54: etary habits as risk factors for inflammatory 16 Sundh B, Hultén L: Oral status in patients with
367–371. bowel disease. Eur J Gastroenterol Hepatol Crohn’s disease. Acta Chir Scand 1982;148:
6 Kasper H, Sommer H: Dietary fiber and nu- 1995;7:47–51. 531–534.
trient intake in Crohn’s disease. Am J Clin 11 Reif S, Klein I, Lubin F, Farbstein M, Hallak
Nutr 1979;32:1898–1901. A, Gilat T: Pre-illness dietary factors in inflam-
matory bowel disease. Gut 1997;40:754–760.

256 Dig Dis 2003;21:252–257 Schütz/Drude/Paulisch/Lange/Lochs


17 Rooney T: Dental caries prevalence in patients 20 Anonymous: Mitteilungen der Gesellschaft: 25 Ainley CC, Cason J, Carlsson LK, Slavin BM,
with Crohn’s disease. Oral Surg 1984;57:623– Empfehlungen zur Untersuchung des Riech- Thompson RP: Zinc status in inflammatory
624. und Schmeckvermögens. HNO Prax 1980;5: bowel disease. Clin Sci 1988;75:277–283.
18 Malchow H, EWE K, Brandes JW, Goebell 62–67. 26 Sundh B, Emilson CG: Salivary and microbial
HM, Ehms S, Sommer H, Jesdinsky H: Euro- 21 Rollin H: Funktionsprüfung und Störungen des conditions and dental health in patients with
pean Cooperative Crohn’s Disease Study Geschmackssinnes. Arch Otorhinolaryngol Crohn’s disease: A 3-year study. Oral Surg Oral
(ECCDS): Results of drug treatment. Gastroen- 1975;210:165–207. Med Oral Pathol 1989;67:286–290.
terology 1984;86:249–266. 22 WHO Oral Health Survey, ed 2. Geneva, 27 Sundh B, Johansson I, Emilson CG, Nordgren
19 Best WR, Becktel JM, Singleton W: Develop- WHO, 1977. S, Birkhed D: Salivary antimicrobial proteins
ment of a Crohn’s disease activity index. Na- 23 Lange DE: Die gezielte Vorbehandlung vor der in patients with Crohn’s disease. Oral Surg Oral
tional Cooperative Crohn’s Disease Study. systemischen Paradontolbehandlung. Zahn- Med Oral Pathol 1993;76:564–569.
Gastroenterology 1976;70:439–444. ärztl Welt/Reform 1975;8:366.
24 McClain C, Soutor C, Zieve L: Zinc deficiency:
A complication of Crohn’s disease. Gastroen-
terology 1980;78:272–279.

Sugar Intake in Crohn’s Disease Dig Dis 2003;21:252–257 257


Original Paper

Dig Dis 2003;21:258–261


DOI: 10.1159/000073344

Serum Mineral Levels in Children with


Intestinal Parasitic Infection
José L. Olivares a Ramona Fernández a Jesús Fleta a Gerardo Rodrı́guez a
Antonio Clavel b
a Departmentof Paediatrics and b Microbiology and Parasitology ‘Lozano Blesa’ Hospital, University of Zaragoza,
School of Medicine, Zaragoza, Spain

Key Words Introduction


Copper W Zinc W Magnesium W Enterobius vermicularis W
Giardia lamblia Childhood is probably the most demanding period of
life for meeting the body’s nutritional demands. Minerals
perform important functions in children’s growth and
Abstract development. The recognition that trace minerals regu-
Parasitic infections are highly prevalent in the general late key metabolic pathways, modulate the immune re-
population. A relation between a parasitic infection and sponse, and suppress the incidence of various disease
absorption of minerals is not an easy task. Serum levels states serves to emphasize their direct importance in
of copper, zinc and magnesium were prospectively mea- health maintenance [1]. Intestinal parasitosis remains an
sured in 64 children with intestinal parasitic infection. important public health concern because of the high fre-
Thirty-nine children with Enterobius vermicularis were quency reached in several countries as well as its nutri-
treated with pyrantel pamoate and 25 children with Giar- tional consequences. Although childhood copper, zinc
dia lamblia with tinidazole and metronidazole. Three and magnesium deficiencies are rather unusual, recent
months after treatment, significant differences in serum studies suggest that several diseases (malnutrition, intesti-
copper, zinc and magnesium were seen in patients with nal malabsorption syndromes and intestinal parasitosis)
E. vermicularis infection, and in serum magnesium lev- may produce them. Early diagnosis and treatment of these
els in patients with G. lamblia. Although the pathogenic disorders are very important to obtain optimal levels of
mechanism is not clear, these findings could reflect a growth, development, immune response and intellectual
deficiency related to malabsorption due to mucous affec- capacity [2–4].
tion. Early detection and treatment of intestinal parasito- The aim of this study was to compare serum copper,
sis could avoid these serum mineral deficiencies. zinc and magnesium levels in children with Enterobius
Copyright © 2003 S. Karger AG, Basel vermicularis and Giardia lamblia intestinal infection, be-
fore and after treatment without infection.

© 2003 S. Karger AG, Basel Prof. José L. Olivares López


ABC 0257–2753/03/0213–0258$19.50/0 Departamento de Pediatrı́a
Fax + 41 61 306 12 34 Facultad de Medicina, Universidad de Zaragoza
E-Mail karger@karger.ch Accessible online at: C/. Domingo Miral s/n, ES–50009 Zaragoza (Spain)
www.karger.com www.karger.com/ddi Tel. +34 976 761724/761725, Fax +34 976 761726, E-Mail olivares@posta.unizar.es
Table 1. Anthropometric measures and serum mineral levels of Table 2. Anthropometric measures and serum mineral levels of
patients with E. vermicularis at diagnosis and after treatment (mean patients with G. lamblia at diagnosis and after treatment (mean B
B SD) SD)

Diagnosis After treatment Diagnosis After treatment

Weight-for-age Z-score – 0.65B1.37 – 0.32B1.16* Weight-for-age Z-score –0.11B1.25 0.34B1.38


Body mass index, kg/m2 17.28B2.31 17.60B2.41** Body mass index, kg/m2 16.88B2.50 17.42B2.62**
Copper, Ìg/dl 116.34B18.62 123.08B24.29* Copper, Ìg/dl 132.17B29.23 132.34B26.69
Zinc, Ìg/dl 91.32B10.90 97.07B13.12** Zinc, Ìg/dl 92.03B13.42 92.12B12.63
Magnesium, mg/dl 1.65B0.11 1.70B0.16* Magnesium, mg/dl 1.64B0.12 1.69B0.13*

Differences between paired sets of time, diagnosis – after treat- Differences between paired sets of time, diagnosis – after treat-
ment, using Student’s test. ment, using Student’s test.
* ! 0.05; ** ! 0.01. * p ! 0.05; ** p ! 0.01.

Patients and Methods Parents were fully informed about the aims of the study and
signed a consent form for participation. The study protocol was
We studied 64 Spanish children (34 boys, 30 girls) from Aragón, reviewed and approved by the Ethical Research Committee of the
Northeast Spain, who were diagnosed as having one unique parasite ‘Lozano Blesa’ Zaragoza University Hospital (Spain).
infection; 39 of these 64 cases were infected by E. vermicularis and 25
by G. lamblia. Fifty-one patients came from an urban area (Zarago-
za) and 13 from rural areas. Most of these children were of a medium Results
socioeconomic status. Ages at diagnosis ranged from 10 months to 15
years (mean 9.71 B 3.63 years) for E. vermicularis and 5.82 B 3.31
years for G. lamblia. Abdominal pain, acute diarrhea, anorexia, anal Table 1 shows measurements of patients with E. vermi-
itch and fever were the most frequent symptoms. cularis intestinal infection at diagnosis and 3 months after
Identification of E. vermicularis was carried out by the Graham treatment, without infection. After treatment we ob-
technique [5]. For G. lamblia identification, concentration of fecal served a significant improvement in weight for age Z-
stool was performed by the method described by Ritchie [6], using
ether instead of acetyl acetate [7, 8]. Copper, zinc and magnesium
score (p ! 0.05), body mass index (BMI) (p ! 0.01), serum
levels were assessed by atomic absorption spectrophotometer (model copper (p ! 0.05), zinc (p ! 0.01) and magnesium (p !
Video 11E, Thermo Jarrel Ash). Serum copper and zinc levels were 0.05) concentrations. Table 2 shows measurements of pa-
previously diluted to a 1/5 proportion and magnesium to a 1/100. tients with G. lamblia infection at diagnosis and 3 months
The method obtains directly measurements of mineral concentration after treatment. We also observed a significant improve-
[9]. Children were evaluated twice, first at diagnosis and on a second
follow-up visit 3 months after the treatment and without active infec-
ment in BMI (p ! 0.01) and serum magnesium (p ! 0.05)
tion. levels after treatment but not in weight-for-age Z-score
Pyrantel pamoate (10 mg/kg/day, two doses separated by 2 weeks) (p = 0.083), copper (p = 0.963) and zinc (p = 0.996) lev-
was the treatment for E. vermicularis. Tinidazole (50 mg/kg/day, two els.
doses, separated by 2 weeks) was used to treat G. lamblia-infected Differences found in patients with E. vermicularis
children and, when G. lamblia parasitation persisted after this treat-
ment, metronidazole (25 mg/kg/ day, 7 days) was employed [10].
intestinal infection compared with the G. lamblia group
Anthropometric measures and serum copper, zinc and magne- were only significant for copper levels at diagnosis (116.3
sium concentrations were determined 3 months after treatment, B 18.6 vs. 132.1 B 29.2 Ìg/dl respectively; p = 0.01) but
when patients were asymptomatic and stools were not infected. not after treatment 3 months later (123.1 B 24.3 vs. 132.3
Anthropometric data were compared with international standards B 26.7 Ìg/dl respectively; p = 0.157). The rest of the vari-
(Anthro database, WHO). Fecal stool samples were collected after
the completion of treatment to verify that there was no intestinal
ables did not show significant differences.
parasitic infection.
Kolmogorov-Smirnov (Lilliefors modification) was applied to
assess normality of each variable. All variables were found to be nor- Discussion
mally distributed (Kolmogorov-Smirnov: Z 1 0.05) and then a para-
metric test was performed. Differences between variables at diagno-
sis and after treatment were examined using Student’s t test. Statisti- The physiologic role and dietary needs for minerals in
cal programs SPSS for Windows 11.5 (SPSS Inc.) were employed. the nutrition of the children are known with varying
Statistical significance was defined as a p ! 0.05. degrees of certainty, and only limited information exists

Serum Minerals in Intestinal Parasitic Dig Dis 2003;21:258–261 259


Infection
on the bioavailability of these elements in different foods. revealed that giardiasis increased the serum copper levels
Copper, zinc and magnesium are components of some cel- and decreased the zinc and iron levels. In our study in
lular enzymes, participate in several immune processes children with parasitic G. lamblia intestinal infection, we
and they play an important role in the resistance to free have observed a significant increase in serum magnesium
radical damage by stabilizing the cellular membrane [1]. levels 3 months after treatment. There are no significant
Absorption of copper in the small intestine has both differences in serum copper or zinc levels after therapy.
active and passive components that do not appear to be The pathogenic mechanism of these serum mineral
dramatically influenced by the form in which the copper changes observed in intestinal parasitic infections is not
is presented. Inside the mucosal cells, copper can asso- clear. Abel et al. [15] pointed out the possible roles of pro-
ciate with metallothionein. Fructose and sucrose accen- tein kinase A (PKA) in cell motility and excystation of the
tuate the deficiency signs induced by a diet low in copper, early diverging eukaryote G. lamblia. Kinetic analysis of
compared with the effect of starch and glucose. Although the recombinant PKA showed that ATP and magnesium
the mechanisms and control of zinc are no completely are preferred in this enzymatic process. Otherwise, Cragg
understood, intestinal absorption involves uptake by the et al. [16] showed that a novel zinc-regulated human zinc
intestinal cell, movement through the mucosal cell, trans- transporter, hZTL1 (human ZnT-like transporter 1), is
fer to the portal circulation, and secretion of endogenous located in the enterocyte apical membrane. Localization,
zinc back into the intestinal cell. Zinc supply and tissue regulatory properties and function of hZTL1 indicate a
reserves are major factors in the homeostatic control of role in regulating the absorption of nutrients.
zinc absorption by regulation of mucosal cell absorption. In conclusion, this study shows significant differences
Magnesium absorption occurs mainly in the ileum and in serum copper, zinc and magnesium levels in patients
colon by passive diffusion, pulling of solvent and active with E. vermicularis intestinal infection, and in serum
transport. Vitamin D increases magnesium absorption magnesium concentrations in patients with G. lamblia, 3
but the presence of phytates decreases it [1, 2]. months after treatment. Although the pathogenic mecha-
The presence of gastrointestinal parasites in children is nism is not clear, persistent small intestinal damage is a
negatively associated with anthropometric characteris- matter of importance in such children [17]. There is clear
tics, physical work capacity, blood hemoglobin levels and evidence that zinc and other minerals are involved in the
nutritional status [2, 11]. Pegelow et al. [4], in a study recovery of small intestinal mucosa after injury. Early
developed in 8- to 10-year-old children with E. vermicu- detection and treatment for each case of intestinal parasit-
laris infection and other parasitic infestations, from ten ic infection could avoid these mineral deficiencies.
schools of the rural districts of Sukaraja, West Java,
Indonesia, concluded that their nutritional status was
characterized by anemia (13%) and a stunting prevalence Acknowledgment
of 51%. In our study, after 3 months of treatment, we have
The authors thank Prof. Tomás Martı́nez, Department of Statis-
observed a significant increase in weight for age Z-score
tics, School of Medicine, University of Zaragoza.
and BMI in E. vermicularis group, and in BMI of children
with G. lamblia infection.
Koltas et al. [12], in children with enterobiosis, showed
that mean levels of serum copper, zinc and magnesium
were significantly lower in the infected group than in the
control group. In our study we have observed a significant
increase in serum concentrations of copper, magnesium
and, especially of zinc, 3 months after treatment with tini-
dazole or metronidazole.
Karakas et al. [13], in children with giardiasis or ame-
biasis, showed that serum zinc levels were significantly
decreased when compared to controls. After metronida-
zole therapy, a significant increase in zinc levels was
observed. There was no significant difference in serum
copper levels between patients and controls before thera-
py. Among G. lamblia-infected children, Ertan et al. [14]

260 Dig Dis 2003;21:258–261 Olivares/Fernández/Fleta/Rodrı́guez/Clavel


References

1 Milner JA: Trace minerals in the nutrition of 7 Young K, Bullock S, Melvin D: Ethyl acetate as 13 Karakas Z, Demirel N, Tarakcioglu M, Mete
children. J Pediatr 1990;117:147–155. a substitute for diethyl ether in the formalin- N: Serum zinc and copper levels in southeast-
2 Stephenson LS, Lathan MC, Ottesen EA: Mal- ether sedimentation techniques. J Clin Micro- ern Turkish children with giardiasis or amebia-
nutrition and parasitic helminth infections. Pa- biol 1979;10:852–853. sis. Biol Trace Elem Res 2001;82:11–18.
rasitology 2000;121(suppl):23–38. 8 Erdman DD: Clinical comparison of ethyl ace- 14 Ertan P, Yereli K, Kurt O, Balcioglu IC, Onag
3 Bahader S, Ali G, Shaalan A, Khalil H, Khalil tate and diethyl in the formalin-ether sedimen- A: Serological levels of zinc, copper and iron
N: Effects of Enterobius vermicularis infection tation techniques. J Clin Microbiol 1981;14: elements among Giardia lamblia-infected chil-
on intelligence quotient and anthropometric 483–485. dren in Turkey. Pediatr Int 2002;44:286–288.
measurements of Egyptian rural children. J 9 Dawson JB, Walker BE: Direct determination 15 Abel ES, Davids BJ, Robles LD, Loflin CE,
Egypt Soc Parasitol 1995;25:183–194. of zinc in wholes blood plasma and urine by Gillin FD, Chakrabarti R: Possible roles of pro-
4 Pegelow K, Gross R, Pietrzik K, Lukito W, atomic absorption spectrometry. Clin Chim tein kinase A in cell motility and excystation of
Richards AL, Fryauff D: Parasitological and Acta 1969;26:465–475. the early diverging eukaryote Giardia lamblia.
nutritional situation of school children in the 10 Hill DR: Giardiasis. Issues in diagnosis and J Biol Chem 2001;276:10320–10329.
Sukaraja district, West Java, Indonesia. South- management. Infect Dis Clin North Am 1993; 16 Cragg RA, Christie GR, Phillips SR, Russi
east Asian J Trop Med Public Health 1997;28: 7:503–525. RM, Kury S, Mathers C, Taylor PM, Ford D: A
173–190. 11 Wilson WM, Dufour DL, Staten LK, Barac- novel zinc-regulated human zinc transporter,
5 Graham CF: A device for the diagnosis of Ente- Nieto M, Reina JC Spurr GB: Gastrointestinal hZTL1, is localized to the enterocyte apical
robius vermicularis. Am J Trop Med 1941;21: parasitic infection, anthropometrics, nutrition- membrane. J Biol Chem 2002;277:22789–
159–161. al status and physical work capacity in Colom- 22797.
6 Ritchie LS: An ether sedimentation technique bian boys. Am J Human Biol 1999;11:763– 17 Walker-Smitth JA: Post-infection diarrhea.
for routine stool examinations. Bull US Army 771. Curr Opin Infect Dis 2001:14;567–571.
Med Dept 1948;8:326. 12 Koltas IS, Ozcan K, Tanner L, Aksungur P:
Serum copper, zinc and magnesium levels in
children with enterobiosis. J Trace Elem Med
Biol 1997;11:49–52.

Serum Minerals in Intestinal Parasitic Dig Dis 2003;21:258–261 261


Infection
Original Paper

Dig Dis 2003;21:262–265


DOI: 10.1159/000073345

Impact of Body Mass Index on Fasting


Blood Glucose Concentration
among Helicobacter pylori Carriers
Ioannis D. Kyriazanos a Ioannis Sfiniadakis b Panagiotis Dimakos c
Vasilios Gizaris d Konstantinos Datsakis e Aggeliki Dafnopoulou f
a 2nd
Department of Surgery and Departments of b Pathology, c Endocrinology, d Immunology and e Gastroenterology,
Naval Hospital of Athens, and f Department of Radiology, P. Faliron Medical Center, Naval Hospital of Athens,
Athens, Greece

Key Words sons with abnormal oral glucose tolerance curve test
Helicobacter pylori W Fasting blood glucose levels W were excluded. Results: Among Hp-positive individuals,
Obesity W Body mass index obese persons presented with a significantly lower mean
blood glucose level than non-obese persons. Obese Hp-
contaminated participants had significantly lower mean
Abstract fasting blood glucose concentrations as well as a signifi-
Background/Aims: Despite the fact that Helicobacter py- cantly smaller percentage of participants with abnormal
lori (Hp) is regarded as a major gastroduodenal patho- elevated blood glucose levels than obese participants
gen, it has recently been suggested to be an important negative to Hp infection. Conclusions: Our data suggest
factor for non-gastroenterologic conditions such as dia- that obesity in combination with Hp infection may induce
betes mellitus. Accordingly, it seems that Hp infection an enhanced response to insulin leading to reduced fast-
may have implications in glycemic control and in fasting ing blood glucose levels, among Hp-positive obese per-
plasma glucose concentrations. As overnutrition and sons in comparison to Hp-positive lean persons.
obesity are directly related to impaired glucose toler- Copyright © 2003 S. Karger AG, Basel

ance, the aim of the present study was to determine


whether Hp infection leads to alterations in fasting plas-
ma glucose concentrations of Hp carriers and especially Introduction
in relation to their body mass index. Methods: Serum
was obtained from 224 young, male navy recruits. An Despite the fact that Helicobacter pylori (Hp) is re-
enzyme-linked immunosorbent assay to detect Hp-spe- garded as a major gastroduodenal pathogen etiologically
cific IgG serum antibodies as well as gastroscopy along linked with duodenal and gastric disease, it has recently
with biopsy was used to identify the infected individuals. been suggested to be an important factor for non-gas-
Serum levels of glucose, urea, creatinine and uric ac- troenterologic conditions such as coronary heart disease
id were also determined. Non-fasting subjects and per- and diabetes mellitus [1]. Although the relationship be-

© 2003 S. Karger AG, Basel Ioannis D. Kyriazanos, MD


ABC 0257–2753/03/0213–0262$19.50/0 22, Thetidos str. P. Faliro
Fax + 41 61 306 12 34 GR–17561 Athens (Greece)
E-Mail karger@karger.ch Accessible online at: Tel. +30 2109845327, Fax +30 2107710671
www.karger.com www.karger.com/ddi E-Mail medkyri@yahoo.com
tween diabetes mellitus and Hp remains controversial, it Obesity Definition
seems that Hp infection may have implications in gly- More specifically by calculating the BMI, we created two distinct
groups: the group of overweight individuals with BMI 625 kg/m2,
cemic control [2]. Furthermore, Hp infection raises basal
and the lean group with BMI ! 25 kg/m2. The cut-off point of ! 25 or
and meal-stimulated serum gastrin concentrations and 625 kg/m2 was proposed in the first federal obesity clinical guide-
lowers iron stores, which may in turn reduce fasting plas- lines released by NHLBI and NIDDK in 1998 as the most acceptable
ma glucose concentrations [3]. classification for obesity [5].
Overnutrition and obesity, due to their well-known
Statistical Analyses
complications including heart disease and diabetes melli- To examine whether the Hp-affected members differed from the
tus, are directly related to impaired glucose tolerance. non-affected members for each study variable, ¯2, Fischer’s exact and
Although we have already reported that obesity and Student’s t tests were used from the SPSS statistical package version
increased BMI cannot be considered as predisposing fac- 8 (SPSS, Chicago, Ill., USA, 1997).
tors for Hp contamination [4], it could be possible that the
combination of Hp infection and obesity can facilitate
Hp-related alterations of plasma glucose concentrations. Results
Accordingly, the aim of the present study was to deter-
mine whether Hp infection leads to alterations in fasting Among several biochemical parameters, fasting blood
plasma glucose concentrations of Hp carriers and in rela- glucose revealed interesting results. Blood glucose levels
tion to their body mass index (BMI). were not significantly different between either obese or
non-obese participants (104.67 B 11.6 vs. 104.4 B 11.7,
p = 0.86), nor between Hp-positive and Hp-negative
Materials and Methods members (103.5 B 8.3 vs. 104.9 B 12.6, p = 0.79).
Abnormal elevated fasting blood glucose levels (nor-
Subjects
mal range 70–110 mg/dl) were detected in 52 (23.2%) out
The study population consisted of 224 healthy male Hellenic
navy recruits (median age 22.84 years old, range 20–30). All the sub- of 224 recruits and the number of abnormal blood glucose
jects were Caucasians and came from different regions of the coun- carriers was comparable between obese and non-obese
try. They were randomly selected out of 600 males inducted into the (28/137, 20.4% vs. 24/87, 27.6%, p = 0.25) as well as
Hellenic Navy at the naval base of Salamis. A blood sample was col- between Hp-positive and Hp-negative (17/61, 27.8% vs.
lected in February 2000, during their induction. After centrifugation,
the serum sample was split into three aliquots, which were frozen at
35/163, 21.4%, p = 0.72) groups.
–70 ° C and thawed once, before analysis. Subsequently and by stratifying the participants in two
groups according to their Hp status (positive or negative),
Determination of Biochemical Parameter Concentrations and we further evaluated the relation between fasting blood
Hp Status glucose levels and BMI for each group separately. Among
Titers of Hp-specific IgG antibodies, as well as serum levels of
glucose, urea, creatinine and uric acid were determined in the
163 uninfected participants (Hp-negative group) there
Department of Immunology of Naval Hospital of Athens. Individu- was no significant difference between obese and non-
als with abnormally increased blood glucose concentrations under- obese persons either in the mean blood glucose levels
went an oral glucose tolerance curve test. Persons with abnormal test (105.8 B 12.7 vs. 103.3 B 12.7, 95% CI = –1.5 to 6, p =
results as well as non-fasting subjects were excluded from the study. 0.22) or in the number of individuals with abnormal
For the serological determination of the specific antibody against Hp,
an enzyme-linked immunosorbent assay was used (Hp IgG Elisa Kit,
blood glucose levels (23/102, 22.5% vs. 12/61, 19.7%, p =
Hycor Biomedical GmbH, Kassel, Germany; inter-assay coefficient 0.85). On the contrary, among 61 Hp-positive individu-
of variation: 7%, intra-assay coefficient of variation: 4.3%, sensitivi- als, obese persons presented with a significantly lower
ty: 0.1 IU/ml). Levels of IgG were categorized as seropositive for Hp mean blood glucose level than non-obese persons (101.2
according to an OD titer of IgG antibodies to Hp of 1 0.5. The normal B 6.4 vs. 106.9 B 8.6, 95% CI = – 9.5 to –1.8, p = 0.005).
range for blood glucose was 60–110 mg/dl, for serum urea 10–40 mg/
dl, for creatinine ! 1.2 mg/dl and for uric acid up to 5.3 mg/dl. All
Furthermore, a significantly higher number of non-obese
seropositive subjects underwent endoscopy in the Department of individuals had blood glucose levels above the normal
Gastroenterology at the Naval Hospital of Athens. Along with the range (1110 mg/dl), in comparison to obese members in
examination, two biopsy samples of the antrum and one of the cor- the Hp-positive group of participants (12/26, 46.1% vs.
pus were obtained from each of the individuals, for the verification of 5/35, 14.2%, p = 0.009).
Hp infection. Subjects with both sero-surveillance- and biopsy-posi-
tive results were finally considered as Hp-contaminated individuals.
Among obese participants, Hp-positive individuals
had a significantly lower mean blood glucose level (101.2
B 6.4 vs. 105.8 B 12.7, 95% CI = 1.2–7.9, p = 0.007) as

Fasting Blood Glucose Concentrations and Dig Dis 2003;21:262–265 263


H. pylori Infection
well as a significantly smaller percentage of members with The identification and sequencing of the mouse obese
abnormal elevated fasting blood glucose concentrations (ob) gene by Friedman’s group in 1994 opened important
(10.7% vs. 29.4%, p = 0.032) than obese Hp-negative per- new avenues in obesity research. The ob gene encodes the
sons. The same was not true among lean individuals. protein leptin, which is produced in the adipocytes,
secreted into the blood and informs the brain about the
size of the fat mass. It is known that plasma leptin highly
Discussion correlates to body fat content and BMI, with obese per-
sons having increased leptin levels [15]. Accumulating
Hp colonization of the human stomach produces an evidence suggests that the role of leptin is much broader
innate host immune response, which finally stimulates G than that of an anti-obesity hormone; leptin exerts several
cells in production of gastrin and gastric acid [6, 7]. other metabolic effects on peripheral tissue including
Hypergastrinemia becomes more enhanced by the ago- modification of the insulin action. Supraphysiologic lev-
nistic action of the microorganism to the H3 histamine els of insulin markedly increase circulating leptin levels,
receptors and its inhibitory effect to CCK and somatosta- while administration of leptin improves insulin resistance
tin secretion that normally diminish gastrin release [8]. in mice and insulin-resistance has been associated with
The increased levels of gastrin and the diminished soma- increased leptin levels [16, 17]. Although the mechanism
tostatin release, in combination with others hormones col- underlying the changes of leptin induced by insulin and
lectively known as the ‘enteroinsular axis’, could poten- vice versa remains to be studied in more detail, these data
tate insulin secretion, increase plasma insulin levels and demonstrate a significant relationship between leptin and
as a result diminish fasting blood glucose levels [9], as insulin.
somatostatin normally has an inhibitory effect on insulin Insulin and corticosteroids are obvious candidates sig-
release. Additionally, insulin can directly stimulate epi- naling fat cells for leptin production. Thus, the increased
nephrine release, which may induce hypoglycemia [10]. leptin expression observed in obesity could result from
Accordingly, Hp infection can affect glucose metabolism chronic hyperinsulinemia and increased cortisol turnover
and Hp-positive individuals might have higher insulin [18] as insulin plasma levels in obese individuals are ele-
and lower glucose plasma levels in comparison to Hp-neg- vated compared to those in normal subjects [19, 20]. The
ative persons. It is already identified that the inflammato- fact that in normoglycemic obese persons, leptin levels are
ry reaction, which Hp infection produces, could be delete- higher than the leptin levels in obese persons with dia-
rious for the control of glucemia, especially for patients betes, can support the option that normally, leptin levels
with diabetes, as well as that Hp gastritis may contribute are negatively correlated with fasting glucose levels [14,
to postprandial hypoglycemia due to hyperinsulinemia 21, 22].
[11–13]. Peach and Barnett [3] reported that Hp infection Taking all these points into consideration and in accor-
may lead to a lower fasting plasma glucose concentration dance with our results, it could be possible that in obese
among women. persons, stimulated sympathetic activity, hyperleptinem-
In the present study we could not identify any signifi- ia and its related hyperinsulinemia, can be combined with
cant difference in fasting blood glucose levels between the increment of gastrin, reduction of somatostatin and
Hp-positive and Hp-negative participants. Hp infection increased histamine resulting from Hp infection, with
demonstrated its role in controlling fasting blood glucose finally further hyperinsulinemia. This combined incre-
concentrations only when it was combined with obesity. ment of insulin might be a reason for the lower fasting
Among Hp-infected individuals, obese persons presented serum glucose values that we recognized in obese than in
with a significantly lower mean blood glucose level than lean Hp-positive persons. Alterations of glucose metabo-
non-obese persons, further supporting the finding that lism in Hp colonization need further investigation.
obese Hp-positive individuals had a significantly lower
mean blood glucose level than obese Hp-negative persons.
Among Hp non-infected individuals, the mean serum glu-
cose level was higher (even not statistically significant) for
obese than lean persons as it is well known that obesity
can be accompanied by alterations in glucose metabolism
[14].

264 Dig Dis 2003;21:262–265 Kyriazanos/Sfiniadakis/Dimakos/Gizaris/


Datsakis/Dafnopoulou
References

1 Senturk O, Canturk Z, Cetinarslan B, Ercin C, 7 Rose S: Gastrointestinal and Hepatobilliary 15 Janeckowa R: The role of leptin in human
Hulagu S, Canturk NZ: Prevalence and com- Pathophysiology. Philadelphia, Fence Creek physiology and pathophysiology. Physiol Res
parison of five different diagnostic methods for Publishing, 1998, pp 161–172. 2001;50:443–459.
Helicobacter pylori in diabetic patients. Endocr 8 Calam J, Gibbons A, Haley Z, Bliss P, Arebi N: 16 Mantzoros CS: The role of leptin in human
Res 2001;27:179–189. How does Helicobacter pylori cause mucosal obesity and disease: A review of current evi-
2 De Luis DA, Cordero JM, Caballero C, Boixe- damage? Its effect on acid and gastrin physiolo- dence. Ann Intern Med 1999;130:671–680.
da D, Aller C, Canton R, De la Calle H: Effect gy. Gastroenterology 1997;113:S43–S49. 17 Wolf G, Chen S, Han DC, Ziyadeh FN: Leptin
of the treatment of Helicobacter pylori infec- 9 Acbay O, Celik AF, Gundogdu S: Does Helico- and renal disease. Am J Kidney Dis 2002;39:
tion on gastric emptying and its influence on bacter pylori-induced gastritis enhance food- 1–11.
the glycaemic control in type 1 diabetes melli- stimulated insulin release? Dig Dis Sci 1996; 18 Fried SK, Ricci MR, Russell CD, Laferrere B:
tus. Diabetes Res Clin Pract 2001;52:1–9. 41:1327–1331. Regulation of leptin production in humans. J
3 Peach HG, Barnett NE: Helicobacter pylori in- 10 Landsberg L, Krieger DR: Obesity, metabolism Nutr 2000;130:3127S–3131S.
fection and fasting plasma glucose concentra- and the sympathetic nervous system. Am J 19 Rasmussen MH, Hvidberg A, Juul A, Main
tion. J Clin Pathol 2001;54:446–449. Hypertens 1989;2:125S–132S. KM, Gotfredsen A, Skakkebaek NE, Hilsted J:
4 Kyriazanos ID, Sfiniadakis I, Gizaris V, Houn- 11 Acbay O, Celik AF, Kadioglu P, Goksel S, Massive weight loss restores 24-hour growth
tis P, Hatziveis K, Dafnopoulou A, Datsakis K: Gundogdu S: Helicobacter pylori-induced gas- hormone release profiles and serum insulin-
The incidence of Helicobacter pylori infection tritis may contribute to occurrence of postpran- like growth factor-I levels in obese subjects. J
is not increased among obese young individu- dial symptomatic hypoglycemia. Dig Dis Sci Clin Endocrinol Metab 1995;80:2446.
als in Greece. J Clin Gastroenterol 2002;34: 1999;44:1837–1842. 20 Larsson H, Elmstahl S, Ahren B: Plasma leptin
541–546. 12 Perdichizzi G, Bottari M, Pallio S, Fera MT, levels correlate to islet function independently
5 NIH: Clinical Guidelines on the Identification, Carbone M, Barresi G: Gastric infection by of body fat in postmenopausal women. Dia-
Evaluation and Treatment of Overweight and Helicobacter pylori and antral gastritis in hy- betes 1996;45:1580–1584.
Obesity in Adults. Bethesda, NIH, 1998. perglycemic obese and in diabetic subjects. 21 Sorensen IT, Echwald MS, Holm JC: Leptin in
www.nhlbi.nih.gov/guidelines New Microbiol 1996;19:149–154. obesity. BMJ 1996;313:953–954.
6 Van Den Brink GR, ten Kate FJ, Ponsioen CY, 13 Begue RE, Mirza A, Compton T, Gomez R, 22 Clement K, Lahlou N, Ruiz J, Hager J, Boug-
Rive MM, Tytgat GN, van Deventer SJ, Pep- Vargas A: Helicobacter pylori infection and in- neres P, Basdevant A, Guy-Grand B, Froguel
pelenbosch MP: Expression and activation of sulin requirement among children with type 1 P: Association of poorly controlled diabetes
the NF-ÎB in the antrum of the human stom- diabetes mellitus. Pediatrics 1999;103:e83. with low serum leptin in morbid obesity. Int J
ach. J Immunol 2000;164:3353–3359. 14 Zimmet P, Hodge A, Nicolson M, Staten M, de Obes Relat Metab Disord 1997;21:556–561.
Courten M, Moore J, Morawiecki A, Lubina J,
Collier G, Alberti G, Dowse G: Serum leptin
concentration, obesity and insulin resistance in
Western Samoans: Cross-sectional study. BMJ
1996;313:965–969.

Fasting Blood Glucose Concentrations and Dig Dis 2003;21:262–265 265


H. pylori Infection
Original Paper

Dig Dis 2003;21:266–270


DOI: 10.1159/000073346

Selenium Is Depleted in Crohn’s Disease


on Enteral Nutrition
Fumitoshi Kuroki Takayuki Matsumoto Mitsuo Iida
Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University,
Fukuoka City, Japan

Key Words lated with the Crohn’s disease activity index. Conclusion:
Selenium W Crohn’s disease W Enteral nutrition These findings suggest that patients with CD on enteral
nutrition are at risk for selenium deficiency and that even
patients without enteral nutrition may develop selenium
Abstract deficiency at the active phase of the disease.
Backgroud/Aims: Selenium is an important trace ele- Copyright © 2003 S. Karger AG, Basel

ment and its deficiency has been reported to be associat-


ed with cardiomyopathy or gastrointestinal cancer. The
aim of this study is to clarify the selenium status in Introduction
Crohn’s disease (CD) on enteral nutrition. Methods: We
measured serum selenium concentrations in 53 patients Selenium is an essential trace element which is a com-
with CD and compared them with those in 21 healthy ponent of enzyme glutathione peroxidase. The enzyme
controls. Twenty-nine patients were under the treatment protects membrane structures by detoxifying oxygen spe-
by enteral nutrition (EN group), and the remaining 24 cies. Because selenium is contained in various foods, defi-
patients were free from formulated enteral nutrition ciency of the trace mineral rarely occurs, except in sub-
(non-EN group). Results: While the serum selenium con- jects who reside in areas where the soil contains a low
centration in the non-EN group was not decreased when amount of selenium and for patients on long-term paren-
compared to controls, the value in the EN group was sig- teral nutrition [1, 2].
nificantly lower than those in the non-EN group and in Because patients with Crohn’s disease (CD) usually
controls. Clinical manifestations of selenium deficiency have various nutritional disturbances, parenteral nutri-
were found in a patient on exclusive enteral nutrition. In tion or enteral nutrition (EN) are applied for the manage-
the EN group, the serum selenium concentration ment of malnutrition. In addition, EN has been reported
showed an inverse correlation with the duration and the to be one of the primary therapies for active CD [3, 4] and
daily dose of enteral nutrition. In the non-EN group, the widely accepted for prolongation of remission in children
serum selenium concentrations were inversely corre- and adolescents [5]. On such occasions, subnormal seleni-

© 2003 S. Karger AG, Basel Fumitoshi Kuroki


ABC 0257–2753/03/0213–0266$19.50/0 Department of Medicine and Clinical Science
Fax + 41 61 306 12 34 Graduate School of Medical Sciences, Kyushu University
E-Mail karger@karger.ch Accessible online at: 3-1-1 Maidashi, Higashiku, Fukuoka City 812-8582 (Japan)
www.karger.com www.karger.com/ddi Tel. +81 982 550505, Fax +81 982 550500, E-Mail fkuro60@f3.dion.ne.jp
Table 1. Details of the patients’ clinical characteristics at the time of investigation

Controls Patients
(n = 21)
EN group (n = 29) non-EN group (n = 24)

Age, years 22–46 (30.0B6.3) 16–77 (29.8B11.7) 16–53 (30.6B10.4) NSa


Sex, m/f 14/7 25/4 18/6 NSa
Body mass index 15.8–22.7 (20.0B2.0) 14.8–24.2 (19.2B2.2) 15.1–24.3 (19.2B2.5) NSa
Time intervalc 0.2–18 (7.1B5.6) 0.1–28 (6.6B7.1) NSa
CD activity index 13–377 (127B98) 15–414 (129B106) NSa
Bowel involvement
Small bowel only 10 11
Large bowel only 2 5 NSb
Small and large bowel 17 8
History of intestinal resection 9 5 NSb
Current medical treatments
Salazosulfapyridine 10 2
Prednisolone 1 2
None 18 22

Values are expressed as the range (mean B SD).


a Comparison among the EN group, non-EN group, and controls.
b Comparison between the EN group and non-EN group.
c Time interval from onset of symptoms to diagnosis, in years.

um status in CD patients on total parenteral nutrition has been ingesting both. Among the EN group, 22 had been ingesting
already been reported [6, 7]. However, there have been a ordinary foods, but 3 had been on exclusive EN without any food
intake. The duration of EN ranged from 0.1 to 5.3 years (1.5 B 1.3
few reports concerning the selenium status in CD patients
years, mean B SD), and the daily dose of EN ranged from 500 to
on EN [8, 9]. The aim of this study is to investigate the 2,300 kcal (1,384 B 483 kcal).
selenium status in CD patients with a reference to EN. Twenty-one normal healthy volunteers participated in this study.
They were randomly selected from our laboratory staff, according to
the following criteria: (1) completely free from previous or present
illness, (2) normal physical examination and blood chemistry, and
Subjects and Methods (3) consumption of ordinary Japanese food. Neither the CD patients
nor controls had been supplied with trace minerals at the time of
Subjects selenium measurement. Informed consent was obtained from each
Fifty-three patients with an established diagnosis of CD and 21 patient and control prior to blood sampling.
healthy control subjects were enrolled for the present study. The diag-
nosis of CD was based on confirmation of clinical, radiographic, or Disease Activity
endoscopic findings and histologic features characteristic of this dis- Activity of CD at the time of investigation was calculated, accord-
ease. None of the patients had steatorrhea, which is a major clinical ing to the formula described as Crohn’s disease activity index (CDAI)
manifestation of malabsorption. [10]. A cut-off value of CDAI 150 was used to divide each patient
CD patients were divided into two groups – EN and non-EN. EN into active (CDAI 1 150) or inactive (CDAI ^ 150) disease.
was used for primary therapy for active disease and for prolongation
of remission. The EN group was composed of 29 patients who had Measurement of Serum Selenium Concentration
been treated by EN. The non-EN group consisted of 24 patients who After an overnight fast, blood samples were obtained from each
had been ingesting ordinary Japanese foods without any formulated patient and control. Blood was centrifuged for separation of serum.
diet. Ten of them had not been treated by any medical and nutrition- After the initial reduction of the serum sample and treatment with
al treatments, and the others had rejected EN. Details of the patients’ the palladium modifier, the concentrations of selenium were deter-
clinical characteristics at the time of investigation are shown in mined by graphite-furnace atomic absorption spectrometry using
table 1. In the EN group, 7 patients had been ingesting elemental diet Model AA-40G atomic absorption spectrophotometer (Varian Cana-
(Elental, Roussel Morishita, Osaka, Japan), 7 had been ingesting da Inc., Georgetown, Ont., Canada) [11]. In addition, hematocrit val-
polymeric diet (Ensure Liquid, Dainabot, Osaka, Japan), and 15 had ue, erythrocyte sedimentation rate, serum albumin and C-reactive

Selenium and Crohn’s Disease Dig Dis 2003;21:266–270 267


Fig. 2. a Thumbnails of the patient prior to selenite supplementa-
tion. The nails are rough and wavy. b The rough and wavy changes of
the nails have improved after selenite supplementation.

Fig. 1. Comparison of serum selenium concentrations among the EN lar, the serum selenium concentration in 3 patients with
group, non-EN group, and controls. exclusive EN showed extremely low values (0.6, 0.7 and
2.9 Ìg/dl, respectively). One of them had been clinically
diagnosed as having selenium deficiency because of im-
paired serum glutathione peroxidase activity, cardiac dys-
function, fingernail deformities, and subsequent im-
protein were measured and body mass index (BMI) was calculated as
provement of clinical symptoms by selenite supplement
follows: weight (kg)/height (m)2.
(fig. 2). In each CD group, the serum selenium concentra-
Statistical Analysis tion was not different according to the site of the disease
Results are expressed as mean B SD. Comparisons among the and to prior history of the intestinal resection.
EN group, non-EN group and the controls were performed by one- The CDAI ranged from 13 to 414 (128 B 101). As
way analysis of variance with ‘a posterior’ Scheffé test. Correlation
indicated in table 1, the CDAI was not different between
coefficients (r) between serum selenium concentrations and various
parameters in each CD group were calculated by regression analysis. the EN and non-EN group. The serum selenium concen-
Probabilities ! 0.05 were considered significant. tration was not different between active CD and inactive
CD in each group.
When possible, the correlation between serum seleni-
Results um concentration and CDAI was further investigated, the
selenium concentration was inversely correlated with
Age, gender and BMI values were not different among CDAI in the non-EN group (r = –0.513, p ! 0.01) (fig. 3),
the two CD groups and the controls (table 1). Serum albu- but such a significant correlation was not found in the EN
min concentration in the CD groups showed lower values group. In contrast, the serum selenium concentration in
than the controls (p ! 0.0001), but the value was not dif- the EN group correlated with daily dose (r = –0.374, p !
ferent between the two CD groups (EN group, 3.9 B 0.05) and duration of EN (r = –0.433, p ! 0.05) with sta-
0.6 mg/dl; non-EN group, 3.8 B 0.6 mg/dl; controls, 4.8 tistical significance (fig. 4). In both EN and non-EN
B 0.2 mg/dl). groups, the serum selenium concentration did not show
any significant correlation with serum albumin levels,
Comparison of Selenium Status BMI, C-reactive protein or erythrocyte sedimentation
The selenium status of the CD patients and controls rate values.
are shown in figure 1. The serum selenium concentration
in the EN group (7.1 B 3.5 Ìg/dl) was significantly lower
than in the non-EN group (10.2 B 2.0 Ìg/dl, p ! 0.0001)
and in controls (11.8 B 1.4 Ìg/dl, p ! 0.0001). In particu-

268 Dig Dis 2003;21:266–270 Kuroki/Matsumoto/Iida


Fig. 3. Correlation between serum selenium concentration and
CDAI in the non-EN group (r = –0.513, p ! 0.01).

Discussion

Cardiomyopathy, weakness in proximal muscle, eleva-


tion of liver transaminase and creatine kinase activities,
and nail changes are the major clinical manifestations of
selenium deficiency [12]. In addition, selenium depletion
has been known to be associated with an increased risk for
gastrointestinal and prostatic cancer [13]. Because cardio-
myopathy in selenium deficiency is irreversible and fatal
[2], early recognition of the condition seems to be inevita-
ble. In clinical situations, selenium status has generally Fig. 4. Correlation between serum selenium concentration and daily
been assessed by serum or erythrocyte glutathione peroxi- dose of EN (a) (r = –0.374, p ! 0.05) or duration of EN (b) (r =
–0.433, p ! 0.05) in the EN group.
dase activities. Because a significant correlation between
serum selenium concentration and glutathione peroxi-
dase activity has been established, the former applied for
the present investigation has been regarded to be appro-
priate for the assessment of selenium status [14].
CD has been known to be accompanied by various patients with severe disease may develop profound seleni-
nutritional disturbances. Low selenium concentrations in um deficiency.
active CD have been shown in previous investigations Parenteral nutrition and EN are established strategies
[14–19], and insufficient oral intake and/or malabsorp- for the management of CD [1–9, 14]. These nutritional
tion have been considered to be the main cause of low therapies contribute to the improvement of nutritional
selenium levels. In this study, the selenium concentration status, intestinal lesions and disease activity [3, 4]. In par-
in the non-EN group was not different from controls. The ticular, EN has been accepted to prolong the remission of
insignificant difference in selenium concentration can be CD in children and in adolescents [5]. Because most of the
explained by the inclusion of inactive or mild disease. enteral formulas prevailing in Japan or in the USA do not
However, since the selenium concentration showed in- contain sufficient selenium [20–22], patients on EN are at
verse correlation with CDAI in the non-EN group, CD high risk of selenium deficiency. In this study, the serum

Selenium and Crohn’s Disease Dig Dis 2003;21:266–270 269


selenium concentration in the EN group was significantly EN showed an extraordinarily low value, and a patient
lower than in the non-EN group and controls. Because had actually developed clinical manifestations of seleni-
there were no differences in clinical or nutritional param- um deficiency. In addition, the selenium concentration
eters other than the diet between the two CD groups, was even low in patients who ingested both EN and ordi-
insufficient selenium content in the enteral formulas nary food. Because the selenium levels were inversely cor-
seems to be the major cause for decreased selenium con- related with the dose and the duration of EN, it would be
centration in the EN group. necessary to take account of selenium deficiency in CD
It has been reported that Elental and Ensure Liquid patients under a prolonged period of EN even if ordinary
used as the formula of EN for our patients contain 1.6 and food is not prohibited.
7.3 Ìg/1,000 kcal of selenium respectively [22]. Because In conclusion, this study indicated that selenium defi-
the recommended dietary allowance of selenium has been ciency possibly occurs in patients with CD, especially in
established to be 0.87 Ìg/kg/day by the US Research those on EN. While selenium supplement seems to be nec-
Council [23], selenium intake with the formula of EN essary for a certain proportion of CD patients, the optimal
alone would be much lower than the recommended value. dose in consideration of possible toxic effects should fur-
In fact, selenium concentrations in 3 cases on exclusive ther be elucidated.

References

1 Van Riji AM, Thomson CD, Mckenzie JM, 8 Fernández-Bañares F, Mingorance MD, Esteve 16 Loeschke K, König A, Haeberlin SH, Lux F:
Robinson MF: Selenium deficiency in total M, Cabré E, Lachica M, Abad-Lacrus A, Gil A, Low blood selenium concentration in Crohn
parenteral nutrition. Am J Clin Nutr 1979;32: Humbert P, Boix J, Gassull MA: Serum zinc, disease. Ann Intern Med 1987;106:908.
2076–2085. copper, and selenium levels in inflammatory 17 Hinks LJ, Inwards KD, Lloyd B, Clayton B:
2 Fleming CR, Lie JT, McCall JT, OBrien JF, bowel disease: Effect of total enteral nutrition Reduced concentrations of selenium in mild
Baillie EE, Thistle JL: Selenium deficiency and on trace element status. Am J Gastroenterol Crohn’s disease. J Clin Pathol 1988;41:198–
fatal cardiomyopathy in a patients on home 1990;85:1584–1589. 201.
parenteral nutrition. Gastroenterology 1982; 9 Thomas AG, Miller V, Shenkin A, Fell GS, 18 Rannen T, Ladefoged K, Hylender E, Hegnhøt
83:689–693. Taylor F: Selenium and glutathione peroxidase J, Jarnum S: Selenium status in patients with
3 González-Huix F, de León R, Fernández-Ba- status in paediatric health and gastrointestinal Crohn’s disease. Am J Clin Nutr 1992;56:933–
ñares F, Esteve M, Cabré E, Acero D, Abad- disease. J Pediatr Gastroenterol Nutr 1994;19: 937.
Lacruz A, Figa M, Guilera M, Planas R, Gas- 213–219. 19 Geerling BJ, Badart-Smook A, Stockbrügger
sull MA: Polymeric enteral diet as primary 10 Best WR, Becktel JM, Singleton JW, Kern F: RW, Brummer RJM: Comprehensive nutri-
treatment of active Crohn’s disease: A prospec- Development of a Crohn’s disease activity in- tional status in patients with long-standing
tive steroid controlled trial. Gut 1993;34:778– dex. Gastroenterology 1976;70:439–444. Crohn disease currently in remission. Am J
782. 11 Jacobson BE, Lockitch G: Direct determina- Clin Nutr 1998;67:919–926.
4 Hiwatashi N: Enteral nutrition for Crohn’s dis- tion of selenium in serum by graphite-furnace 20 Zabel NL, Harland J, Gormican A, Ganther
ease in Japan. Dis Colon Rectum 1997;40: atomic absorption spectrometry with deuter- HE: Selenium content of commercial formula
S48–S53. ium background correction and a reduced pal- diets. Am J Clin Nutr 1978;31:850–858.
5 Wilschanski M, Sherman P, Pencharz P, Davis ladium modifier: Age-specific reference ranges. 21 Martin RF, Young VR, Janghorbani M: Seleni-
L, Corey M, Griffiths A: Supplementary enter- Clin Chem 1988;34:709–714. um content of enteral formulas. J Parenter En-
al nutrition maintains remission in paediatric 12 Lockitch G: Selenium: Clinical significance teral Nutr 1986;10:213–215.
Crohn’s disease. Gut 1996;38:543–548. and analytical concepts. Crit Rev Clin Lab Sci 22 Tanaka T, Higashi A, Matsuda I, Suzuki I, Asa-
6 Fleming CR, McCall JT, O’Brien JF, Forsman 1989;27:483–541. kawa M: Selenium content of Japanese enteral
RW, Ilstrup DM, Petz J: Selenium status in 13 Willett WC, Morris JS, Pressel S: Prediagnostic formulas (Engl abstract). J Jpn Soc Nutr Food
patients receiving home parenteral nutrition. J serum selenium and risk of cancer. Lancet Sci 1995;48:147–50.
Parenter Enteral Nutr 1984;8:258–262. 1983;ii:130–134. 23 Food and Nutritional Board, National Re-
7 Jacobson S, Plantin LO: Concentration of sele- 14 Rannem T, Ladefoged K, Hylander E, Hegnhøj search Council: Trace Elements. Recom-
nium in plasma and erythrocytes during paren- J, Staun M: Selenium depletion in patients with mended Dietary Allowances, ed 10. Washing-
teral nutrition in Crohn’s disease. Gut 1985;26: gastrointestinal disease: Are there any predic- ton, National Academy Press, 1989, pp 217–
50–54. tive factors? Scand J Gastroenterol 1998;33: 224.
1057–1061.
15 Penny WJ, Mayberry JF, Aggett PJ, Gilbert JO,
Newcombe RG, Rhodes J: Relationship be-
tween trace elements, sugar, consumption and
taste in Crohn’s disease. Gut 1983;24:288–
292.

270 Dig Dis 2003;21:266–270 Kuroki/Matsumoto/Iida


Original Paper

Dig Dis 2003;21:271–275


DOI: 10.1159/000073347

L-Carnitine in the Treatment of Mild or


Moderate Hepatic Encephalopathy
Mariano Malaguarnera a Giovanni Pistone a Marinella Astuto c
Simona Dell’Arte a Giovanna Finocchiaro a Emilia Lo Giudice a
Giovanni Pennisi b
a Dipartimento di Scienze della Senescenza, Urologiche e Neurologiche, Università degli Studi di Catania, e
b Dipartimenti di Neuroscienze e c Specialità Medico-chirurgiche Sez. Anestesiologia, Università di Catania, Italia

Key Words Introduction


Hepatic encephalopathy W L-Carnitine treatment
Hepatic encephalopathy (HE) is one of the major com-
plications of cirrhosis. Clinical grading is based on im-
Abstract paired mental function, neurological disorders and al-
Hepatic encephalopathy (HE) is one of the major compli- tered states of consciousness, to stupor and coma. Patho-
cations of cirrhosis. Experimental and clinical findings genesis of this common syndrome remains partially un-
observed in liver, muscle and brain have provided new known. Hyperammoniemia, due to the several mecha-
insights into the ammonia mechanism of action. L-Carni- nisms proposed, is one of the main pathogenetic factors in
tine (LC), inducing ureagenesis, may decrease blood and the development of HE, as shown by the association of
brain ammonia levels. 120 patients meeting inclusion increased serum ammonia levels with impairment of
criteria were randomized either to a treatment for 60 mental function in patients with liver failure. Experimen-
days with LC or placebo (2 g twice a day). Previous stud- tal and clinical findings observed in liver, muscle and
ies have reported a significant protective effect of LC in brain have provided new insights into the ammonia
mice and rats, which is associated with a significant mechanism of action. In fact, these organs play a funda-
reduction of blood and brain ammonia concentration, mental role in the removal of ammonia in cases of hyper-
suggesting an action of LC either at peripheral or central ammoniemia [1, 2].
sites. Results of our study show a protective effect of LC A decrease in the muscle mass is a jeopardizing factor
in ammonia-precipitated encephalopathy in cirrhotic pa- for the development of encephalopathy [3]. Muscle mass
tients. Either in subjects with HE 1 or 2 we observed a loss is also associated with a decreased ability to change
significant reduction at day 30 and more markedly at day ammonia in glutamine. All morphological and functional
60 of treatment. A significant therapeutic effect of LC was characteristics of the brain in experimental models and
also observed in the NCT-A, which is an accepted and humans rule out an initial involvement of neurons in HE,
reliable psychometric test for the assessment of mental the astrocytes being the first target of the disease [4]. As
function in cirrhotic patients with HE. showed by Ullian et al. [5], these cells play a key role in the
Copyright © 2003 S. Karger AG, Basel correct development and function of neurons. In HE, no

© 2003 S. Karger AG, Basel Mariano Malaguarnera, AP


ABC 0257–2753/03/0213–0271$19.50/0 Viale Andrea Doria, 69
Fax + 41 61 306 12 34 IT–95126 Catania (Italy)
E-Mail karger@karger.ch Accessible online at: Tel. +39 095 7262008, Fax +39 095 7262011
www.karger.com www.karger.com/ddi E-Mail malaguar@mbox.unict.it
morphological neuronal alterations are detected, changes ry; (3) patient with other neurological disease and metabolic disor-
in astrocyte characteristics being present (Alzheimer type ders such as alcoholism, diabetes mellitus, unbalanced heart failure
and/or respiratory failure or end-stage renal disease; (4) severe HE
II cells) [6]. The majority of therapeutic measures current-
(mental state grade 3–4); (5) administration of anti-HE medications
ly in use are therefore aimed at reduction of serum ammo- such as neomycin, lactulose, lactitol, branched-chain amino acids;
nia levels, by decreasing enteric ammonia production [7]. (6) any additional precipitating factors such as high protein intake
Carnitine is a natural substance involved in regulating (additional high-protein meals), constipation or intake of psycho-
substrate flux and energy balance across cell membranes. stimulants, sedatives, antidepressants, benzodiazepines, or benzodi-
azepine antagonists (flumazenil), and (7) patients with fever, sepsis
It is a cofactor for the shuttle mechanism whereby long-
or shock were also excluded to avoid variations caused by body tem-
chain fatty acids are transformed into L-acetylcarnitine perature.
metabolites and may be carried out into mitochondria in
order to be part of ß-oxidation [8]. Furthermore, carnitine
and the carnitine acetyltransferase are involved in the fol- Study Design
lowing reaction: acetyl-coenzyme A + carnitine = acetyl-
carnitine + coenzyme A. This reaction modulates the Patients meeting inclusion criteria were randomized in a double-
blind fashion to either a treatment for 60 days with LC (2 g twice a
intracellular concentration of coenzyme A and acetyl- day) (group A) or placebo (which were identical in appearance and
CoA. By this reaction, carnitine produces free CoA for packaging) (group B). Randomization was based on a computer-gen-
other metabolic reactions and reduces the ratio of acetyl- erated list. The consumption of oral LC at a dose of 4 g/day divided
CoA to CoA. This reduction stimulates the activity of into two doses (2 g in the morning and 2 g in the evening) for 60 days
pyruvate dehydrogenase and, thus, enhances the oxida- met the 100% of compliance. A minimum of 210 pills (each pill 1 g)
was considered as good compliance. Concomitant medications
tive use of glucose. (whose administration continued throughout the study) included
O’Connor et al. [9] showed a protective effect of L- diuretics (36 of group A and 30 of group B) and ß-blockers (27 of
carnitine (LC) administration to mice 1 h before a lethal group A and 25 of group B).
injection of ammonium acetate. Therrien et al. [10] All study series were subdivided into groups belonging to HE 1 or
reported that LC in the portocaval shunted rat prevented HE 2 according to the initial HE grade (West Haven criteria). Group
A was composed of patients with initial HE 1 (LC 35 patients, place-
not only the development of severe encephalopathy, but bo 33 patients); group B with initial HE 2 (LC 25 patients, placebo 27
also reduced mortality. It was suggested that LC, inducing patients). The effectiveness of therapy was compared and evaluated
ureagenesis, may decrease blood and brain ammonia lev- separately in the different subgroups.
els. In order to assess the clinical efficacy of LC in the The groups were homogeneous with regard to anamnestic and
treatment of HE, a randomized, double-blind, placebo- diagnostic criteria. Differences in the composition of the two groups
with respect to precipitant factors may be minimized, because the
controlled study with oral administration in cirrhotic patient population was well defined by inclusion and exclusion crite-
patients with hyperammoniemia and chronic symptoms ria. Any pathological, clinical or laboratory findings observed during
has been carried out. the study were monitored and documented until their normaliza-
The aim of our study was to evaluate in a practice- tion.
adapted design the influence of LC using the number con-
NCT-A
nection test A (NCT-A), mental conditions and ammonia The NCT-A measures cognitive and motor ability, and consists of
effects. a combination of number series on a sheet with best celerity possible.
A decrease in the time employed to complete the NCT-A reflects an
improvement of the neurological function.

Patients and Methods Venous Ammonia Concentration


The blood ammonia levels were evaluated by the enzymatic
120 randomly selected patients (9 alcoholics, 33 hepatitis B virus determination using glutamate dehydrogenase in a rapid and inter-
infected, 63 hepatitis C virus infected, 15 cryptogenetic cirrhosis) ference-free photometric determination (340 nm) of NH +4 in native
met the following inclusion criteria and were enrolled in the study:
blood plasma according to the De Fonseca-Wollheim method [12].
(1) chronic hepatitis with spontaneous manifest HE (mental state
The blood sample was immediately taken after withdrawal to the lab-
grade 1 or 2 according to the West Haven criteria) and an NCT-A
oratory for immediate (within 15 min) determination of NH4 +4.
performance time 1 30 s; (2) hyperammoniemia (venous ammonia
concentration 1 50 mmol/l), and (3) cooperative, hospitalized adult Hepatic Encephalopathy
patients with liver cirrhosis diagnosed by clinical, histological and Encephalopathy grade was diagnosed on the basis of the evalua-
ultrasonographic findings. tion of consciousness, intellectual functions, behavior and neuromus-
Exclusion criteria were the following: (1) major complications of cular functions according to the West Haven criteria introduced by
portal hypertension, such as gastrointestinal blood loss, hepatorenal Conn and Liebenthal [13, 14].
syndrome or bacterial peritonitis; (2) acute superimposed liver inju-

272 Dig Dis 2003;21:271–275 Malaguarnera/Pistone/Astuto/Dell’Arte/


Finocchiaro/Lo Giudice/Pennisi
Table 1. Baseline characteristics of patients
Parameter Carnitine group (A) Placebo group (B)

Male/female 40/20 38/22


Age 51.7B11.8 52.4B10.4
Cirrhosis etiology
Alcohol 4 5
Post-hepatitis B 16 17
Post-hepatitis C 32 31
Cryptogenetic 8 7
Child-Pugh class
A 29 30
B 32 31
C 9 9
Prothrombin time, % 60.4B6.7 61.2B6.9
Serum albumin level, g/dl 2.7B0.6 2.8B0.5
Serum bilirubin level, mg/dl 3.8B1.4 3.6B1.5
Serum alanine aminotransferase level, IU/l 128B61 120B63
Blood urea nitrogen, mg/dl 44B10 43B12
Serum creatinine level, mg/dl 1B0.15 0.98B0.14
Natriemia, mEq/l 133B3.4 132B4.7

Safety Parameters differences in serum NH +4 levels. In group HE 1, after 30


Safety parameters included blood tests (hemoglobin, hematocrit, days of treatment, p was 0.003 (CI 6.37–30.43), being
white and red blood cell count, platelet count) and liver function tests
0.000 (CI 27.82–48.98) after 60 days compared to base-
(alanine aminotransferase, aspartate aminotransferase, Á-glutamyl-
transpeptidase, cholinesterase activity, serum bilirubin, serum albu- line values and 0.000 (CI 11.14–28.86) compared to day
min concentrations, prothrombin time and partial thromboplastin 30. In group HE 2, p was 0.000 (CI 15.76–40.04) after 30
time) on days 0, 30 and 60. days, being 0.000 (CI 32.96–53.24) after 60 days com-
pared to baseline values and 0.001 (CI 6.01–24.39) com-
Statistical Analysis
pared to day 30 of treatment respectively. With reference
Descriptive statistics were prepared from the study sample and
results were expressed as means B SD. Statistical significance in con- to NCT-A, in group HE 1 we observed a p of 0.000 (CI
tingency tables was evaluated using ¯2 test and Fisher’s exact test. 6.23–19.17) after 30 days and 0.000 (CI 17.78–30.62)
Student’s test for unpaired data, one-way ANOVA and Mann-Whit- after 60 days compared to baseline values as well as 0.000
ney rank sum test were used for comparisons of continuous variables. (CI 5.15–17.85) with respect to day 30 of treatment
Statistical analyses were performed using appropriate tests for re-
respectively. In group HE 2 we found a p of 0.000 (CI
peated measures as well as by controlling for multiple comparisons
with correction of the Duncan procedure. The study protocol was 7.91–21.89) after 30 days and 0.000 (CI 20.20–34.20)
received and approved by the Institutional Review Board of the Hos- after 60 days compared to baseline values and 0.000 (CI
pital following the guidelines of the 1975 Declaration of Helsinki. 5.63–18.97) with respect to day 30 of treatment.

Placebo Treatment
Results No statistical difference was found in these patients at
days 30 and 60 with respect to baseline and day 30 values
Baseline Values respectively in both groups. In fact, for fasting serum NH +4
The two groups were homogeneous for demographic levels, in group HE 1 we found a p of 0.477 after 30 days
characteristic, etiology, casting of disease and Child-Pugh (CI –7.63 to 16.23), after 60 days a p of 0.419 (CI –7.21 to
grade. Serum NH +4 fasting concentrations were not signifi- 17.21) with respect to baseline values and 0.905 com-
cantly different before the treatment. NCT-A did not pared to values of day 30 of treatment (CI –10.87 to
show significant differences at baseline. 12.27).
In group HE 2 we found a p of 0.396 (CI –7.01 to
L-Carnitine Treatment 17.61) after 30 days and of 0.303 (CI –5.95 to 18.95) after
The patients treated with carnitine either in group HE 60 days with respect to baseline and 0.832 (CI –10.01 to
1 or in group HE 2 showed fasting statistical significant 12.41) with respect to values of day 30 respectively. For

L-Carnitine in the Treatment of Mild or Dig Dis 2003;21:271–275 273


Moderate HE
Table 2. Comparison between evaluated parameters of the two groups (days of treatment)

Carnitine group Placebo group


0 days 30 days 60 days 0 days 30 days 60 days

NH +4 fasting HE 1 80.2B36.9 61.8B29.2 41.8B18.7 80.7B34.7 76.4B31.2 75.7B32.8


HE 2 88.7B35.6 60.8B31.4 45.6B17.5 86.7B37.2 81.4B30.6 80.2B31.4
NCT-A HE 1 62.8B18.1 50.1B17.7 38.6B17.4 60.7B20.3 58.7B19.4 56.7B20.7
HE 2 66.4B20.2 51.5B18.4 39.2B18.5 65.8B21.4 65.1B19.7 63.2B23.1

NCT-A, in group HE 1 we found a p of 0.582 after 30 days Discussion


(CI –5.18 to 9.18) and of 0.287 (CI –3.41 to 11.41) after
60 days compared to baseline values and 0.586 (CI –5.25 Several pathogenetic mechanisms have been suggested
to 9.25) with respect to values of day 30. Following the to explain the onset and progression of HE. The most
same sequence of the previously listed parameters, in ancient hypothesis is linked to the neurotoxicity of ammo-
group HE 2 we found a p of 0.852 (CI –6.74 to 8.14) after nia. The majority of blood NH +4 amount results from mus-
30 days, 0.524 (CI –5.45 to 10.65) after 60 days with cle protein catabolism at intestinal level; the remnant is
respect to basal conditions and 0.629 (CI –5.86 to 9.66) produced by the action of colic bacteria on the natrium
compared to day 30 of administration. present in digested foods. Ammonia is vehicled to the liv-
er throughout the portal flux and is normally eliminated
Comparison between Treatments as urea. The liver damage or the presence of portosys-
In group HE 1 for NH +4 at day 30 of administration, the temic shunts increase its serum levels [15, 16]. The
patients treated with carnitine showed a response statisti- exceeding ammonia is eliminated from the blood by
cally significant with respect to placebo (p = 0.009 CI transforming of glutamate into glutamine in skeletal mus-
–25.252 to –3.68); in group HE 2, at day 30 of administra- cle as well as central nervous system. Neurotoxicity of
tion p was 0.000 (CI –31.81 to –9.39). For NCT-A, in ammonia is probably due to a direct action on neurons,
group HE 1 at day 30 of administration a statistical differ- because the reduction of glutamate and the increase of
ence was found with p 0.012 (CI –15.31 to –1.89), while glutamine may induce a swelling of the astrocytes [16].
in group HE 2 at day 30 of administration p was 0.000 (CI Previous studies have reported a significant protective
–20.49 to –6.71) in favor of carnitine patients. At day 60 effect of LC in mice and rats, which is associated with a
for NH +4 in group HE 1, carnitine-treated patients showed significant reduction of blood and brain ammonia con-
a better significant response than placebo patients, p was centration [9, 17], suggesting an action of LC either at
0.000 (CI –43.55 to –24.25); in group HE 2, p was 0.000 peripheral or central sites. In the study by O’Connor et al.
(CI –43.73 to –25.41) in favor of carnitine. About NCT- [9], blood urea concentrations were significantly in-
A, at day 60 the response was strikingly in favor of carni- creased and inversely related to the lowering of blood
tine-treated patients: in group HE 1, p was 0.000 (CI ammonia after one carnitine administration to normal
–25.01 to –11.19), while in group HE 2, p was 0.000 (CI mice to whom toxic doses of ammonia were adminis-
–31.57 to –16.43). tered. This fact suggests that LC’s effect in these animals
was caused, at least partially, by an action on hepatic urea
Adverse Events synthesis. Results of our study show a protective effect of
Both LC and placebo were well tolerated in 100% of LC in ammonia-precipitated encephalopathy in cirrhotic
patients. In the group treated with LC, 1 patient com- patients. Either in subjects with HE 1 or HE 2, we
plained of nausea, 2 of slight headache and 2 of abdomi- observed a significant reduction at day 30 and more
nal pain. In placebo group, 2 patients complained of diar- markedly at day 60 of treatment. A significant therapeutic
rhea and 1 of moderate headache. Nobody withdrew from effect of carnitine was also observed in the NCT-A, which
the planned treatment. is an accepted and reliable psychometric test for the
assessment of mental function in cirrhotic patients with
HE [13, 18]. LC crosses the hematoencephalic barrier

274 Dig Dis 2003;21:271–275 Malaguarnera/Pistone/Astuto/Dell’Arte/


Finocchiaro/Lo Giudice/Pennisi
slowly (brain uptake index 5.5%) but in spite of this fact, mals. The possible beneficial effect of carnitine may be
its amount in the brain is relatively large [19]. In the study related to an improved pyruvate oxidation, Krebs cycle
of Therrien et al. [20], the protective effect of LC was and flux through glutamate dehydrogenase. The latter
accompanied by a significant attenuation of the increased could then explain the lowering of blood ammonia levels
cerebrospinal fluid and brain alanine as well as cerebro- that follows LC administration. In conclusion, this study
spinal fluid lactate content, caused by ammonium acetate demonstrates a clinically significant effect of LC on men-
administration. This fact suggests that mitochondria res- tal conditions and ammonia levels in patients with mild
piration is at least partially restored in LC-treated ani- or moderate HE.

References

1 Basile AS, Jones EA: Ammonia and GABAer- 8 Bremer J: The role of carnitine in cell metabo- 15 Rikkers L, Jenko P, Rudman D, Freides D:
gic neurotransmission: Interrelated factors in lism; in De Simone C, Famularo G (eds): Car- Subclinical hepatic encephalopathy: Detection
the pathogenesis of hepatic encephalopathy. nitine Today. Heidelberg, Springer, 1997, pp prevalence and relationship to nitrogen metab-
Hepatology 1997;25:103–105. 1–37. olism. Gastroenterology 1978;75:462–469.
2 Lockwood AH, Yap EWH, Wong WH: Cere- 9 O’Connor JE, Costell M, Grisolia S: Pre- 16 Gilberstadt SJ, Gilberstadt H, Zieve L, Buegel
bral ammonia metabolism in patients with se- vention of ammonia toxicity by L-carnitine: B, Collier RO, McClain CJ: Psychomotor per-
vere liver disease and minimal hepatic enceph- Metabolic changes in brain. Neurochem Res formance defects in cirrhotic patients without
alopathy. J Cereb Blood Flow Metab 1991;11: 1984;9:563–570. overt encephalopathy. Arch Intern Med 1980;
337–341. 10 Therrien G, Rose C, Butterworth J, Butter- 140:519–521.
3 Tarter RE, Panzak G, Switala J, Lu S, Simke- worth RF: Protective effect of L-carnitine in 17 Matsouka M, Igsu H, Kohriyama K, Inoue N:
vitz H, Van Thiel D: Isokinetic muscle strength ammonia-precipitated encephalopathy in the Suppression of neurotoxicity of ammonia by L-
and its association with neuropsychological ca- portocaval shunted rat. Hepatology 1997;25: carnitine. Brain Res 1991;567:328–331.
pacity in cirrhotic alcoholics. Alcohol Clin Exp 551–556. 18 Atterbury CE, Maddrey WC, Conn HO: Neo-
Res 1997;21:191–196. 11 Reitan RM, Woegson D: The Halstead-Reitan. mycin-sorbitol and lactulose in the treatment
4 Haussinger D, Kircheis G, Fischer R, Schiess Neuropsycological test battery: theory and clin- of acute portal-systemic encephalopathy. Dig
F, Von Dahl S: Hepatic encephalopathy in ical interpretation. Tucson, Neuropsychology Dis 1978;23:398–408.
chronic liver disease: A clinical manifestation Press, 1993. 19 Hearn TJ, Coleman AE, Lai JCK, Griffith OW,
of astrocyte swelling and low-grade cerebral 12 De Fonseca-Wollheim F: Direkte Plasmaam- Cooper AJL: Effect of orally administered L-
edema? J Hepatol 2000;32:1035–1038. moniakbestimmung ohne Enteiweissung. J carnitine on blood ammonia and L-carnitine
5 Ullian EM, Sepperstein SK, Christopherson Clin Chem Clin Biochem 1973;11:421–431. concentrations in portocaval-shunted rats. He-
KS, Barres BA: Control of synapse number by 13 Conn HO, Liebertahl MM: The Hepatic Coma patology 1989;10:822–828.
glia. Science 2001;291:657–661. Syndrome and Lactulose. Baltimore, Williams 20 Therrien G, Giguere JF, Butterworth RF: In-
6 Noremberg MD: Astrocyte-ammonia interac- & Wilkins, 1979, pp 1–121. creased cerebrospinal fluid lactate reflects dete-
tions in hepatic encephalopathy. Semin Liver 14 Conn HO: Trail-making and number connec- rioration of neurological status in experimental
Dis 1996;16:245–253. tion tests in the assessment of mental state in portal-systemic encephalopathy. Metab Brain
7 Conn HO, Leevy CM, Vlahovic ZR, et al: portal systemic encephalopathy. Am J Dig Dis Dis 1991;6:231–238.
Comparison of lactulose and neomycin in the 1977;22:541–550.
treatment of chronic portal systemic encepha-
lopathy. Gastroenterology 1997;72:573–583.

L-Carnitine in the Treatment of Mild or Dig Dis 2003;21:271–275 275


Moderate HE
Original Paper

Dig Dis 2003;21:276–278


DOI: 10.1159/000073348

Fructose Breath Hydrogen Test –


Is It Really a Harmless Diagnostic
Procedure?
P. Müller a, b C. Meier b H.J. Böhme c T. Richter b, d
a HELIOS Hospital, Leisnig; b Children’s Hospital, University of Leipzig; c Institute of Biochemistry,
University of Leipzig, and d St. Georg Hospital, Leipzig, Germany

Key Words Introduction


Fructose malabsorption W Hereditary fructose
intolerance W Fructose breath hydrogen test Patients with chronic abdominal pain, enteritis and
meteorism are frequent clients in clinical and ambulatory
practice. One possible cause of these complaints are indi-
Abstract vidual distinctions of the intestinal capacity of absorption
Usage of hydrogen breath tests has become one of the for fructose, which is estimated in adults between 0.07
standard procedures in diagnosing chronic unspecific and 0.7 g/kg body weight (b.w.) [1]. If this individual
abdominal pain. These tests are said to be of sufficient intestinal maximum of absorption for fructose is ex-
specificity and sensitivity, are easily done, non-invasive ceeded, fructose will be metabolized by bacterial fermen-
and are more often practiced in outpatients. A 13-year- tation and typical signs and symptoms of an isolated fruc-
old boy is reported with chronic unspecific abdominal tose malabsorption develop [2]. The prevalence of this
pain and growth retardation and so far misdiagnosed clinical entity increases with increasing use of high fruc-
hereditary fructose intolerance (HFI), who developed life- tose corn syrup as sweetener in processed food. The cur-
threatening adverse effects during the fructose breath rent view in the literature is that fructose is transported
hydrogen test. It is concluded that the possibility of HFI across the brush-border membrane by GLUT-5, a mem-
should be excluded first by a carefully explored dietary ber of the facilitative fructose transporter family [3].
history before the fructose breath test is performed However, screening for mutations in the coding region of
under medical supervision. If there is any suspicion of the GLUT-5 gene was without success and the conclusion
HFI, a molecular genetic analysis should be preferred. of the study was that isolated fructose malabsorption does
Copyright © 2003 S. Karger AG, Basel not result from the expression of mutant GLUT-5 protein
[4]. Moreover, it has recently been shown that in addition
to GLUT-5, GLUT-2 also contributes to the fructose
transport across the brush-border membrane – at least in
rats [5]. Thus, isolated fructose malabsorption still awaits

© 2003 S. Karger AG, Basel Peter Müller, MD


ABC 0257–2753/03/0213–0276$19.50/0 Department of Pediatrics, HELIOS Hospital Leisnig
Fax + 41 61 306 12 34 Colditzer Strasse 48
E-Mail karger@karger.ch Accessible online at: DE–04703 Leisnig (Germany)
www.karger.com www.karger.com/ddi Tel. +49 34321 8310, Fax +49 34321 8111, E-Mail pmueller@leisnig.helios-kliniken.de
an explanation. Hydrogen breath tests are known as suffi- Discussion
cient methods to detect intestinal carbohydrate malab-
sorptions. An increase of exhaled hydrogen of 120 ppm At present, the content of fructose as the naturally
after oral application of fructose (1 g/kg b.w., maximum sweetest occurring sugar in foodstuffs is enormous in
25 g) points to fructose malabsorption [6, 7]. Although developed industrial countries. The estimated daily con-
this test seems to be simple and safe, evaluations in sumption of sugar was 4.5 g per capita in England in 1700,
infants and children showed problems when measure- in 1800 already 37 g was consumed and in 1968 this had
ment results had to be interpreted and when the predic- risen to 140 g per capita daily [11]. These remarkable
tive value and consecutive indications of this breath test changes in nutritional habits are one of the common
were proven [8–10]. causes of chronic unspecific abdominal pain in children
We report on an adverse effect during a hydrogen (toddler’s diarrhea) due to fructose malabsorption. Beside
breath test with fructose in a schoolboy who suffered from other influences of environment and stress, this dysfunc-
unknown inherited fructose intolerance. Such a case has tion is also a frequent cause of chronic enteritis, meteo-
not been described so far and therefore, it is not advisable rism and irritable bowel syndrome in adults, especially in
to perform a hydrogen-exhaled breath test after oral fruc- cases where sorbitol-containing food for diabetics is con-
tose loading without relevant indications. sumed [12]. If the fructose malabsorption is recognized as
the cause of complaints, the dietary management with
supplementation of equimolar amounts of glucose will
Case Report immediately lead to recovery [13]. Affected children have
no growth retardation. In contrast, poor growth and fail-
A growth-retarded 13-year-old boy who had suffered from ab-
ure to thrive are typical signs of patients with autosomal
dominal pain for several years visited for gastroenterological consul-
tation. At the time, a partial growth hormone deficiency was treated recessively inherited fructose intolerance (HFI) after
with growth hormone. The family history was unremarkable regard- chronic fructose intoxication, which is pathogenetically
ing gastrointestinal diseases. Parents were of normal heights, but a distinguished from intestinal absorption disturbances
sister who was 2 years older was also growth-retarded. The patient’s [14]. HFI was first described as a clinical entity in 1956
dietary history was suspicious with aversion to fruits, small amounts
[15]. The molecular basis of this disease is a deficiency of
of sugar, but a considerable amount of milk products. In contrast, his
sister tolerated fructose-containing foods very well. The boy’s appe- the enzyme aldolase B (EC 4.1.2.13) in liver, intestine and
tite was good, stool was normal and reduction of weight or efficiency kidney [16]. After exposition to fructose, dose-dependent
was not observed. Clinical investigation, especially of the abdomen, cytosolic accumulations of fructose-1-phosphate occur
showed normal findings. The liver was palpable 1 cm below the ribs. which inhibit liver phosphorylase A. Glycogenolysis and
A fructose breath hydrogen test was performed to confirm the sus-
glycolysis are interrupted and the resulting hypoglycemia
pected fructose malabsorption. 60 min after oral fructose load with
1.5 g/kg b.w., the patient became indisposed with tachycardia (110/ is resistant to glucagon. Hypophosphatemia, which inten-
min), hypotonia (105/70 mm Hg), cold-sweaty skin, disturbed con- sifies the inhibition of liver phosphorylase A, is another
sciousness and vomiting. Measurement of blood glucose revealed a consequence of sequestration of phosphate as fructose-1-
hypoglycemia with 2.2 mmol/l and a metabolic acidosis (pH 7.32, phosphate. Intracellular deficiency of ATP and phosphate
base excess –7.5 mmol/l). Signs and symptoms immediately normal-
lead to hyperuricemia and hypermagnesemia, respective-
ized after intravenous injection of 10 g glucose. Further investiga-
tions revealed elevated liver enzyme activities of GPT (1.10 Ìmol/ ly [14]. At present, 21 mutations within the human aldo-
s-l) and GOT (1.68 Ìmol/s-l) and an increased lactate concentration lase B gene have been published, of these, 15 are single-
(3.3 mmol/l), while anorganic phosphate was slightly reduced to base substitutions. In Germany, 93% of HFI alleles are
1.41 mmol/l. Uric acid in serum was found to be in the normal range. one of three common mutations: A149P, A174D and
These findings pointed to hereditary fructose intolerance. Mutation-
N334K [17, 18]. Hypoglycemia, metabolic acidosis, hypo-
al analysis revealed compound heterozygosity of the patient for the
two common mutations A149P in exon 5 and the mutation N334K phosphatemia, hyperuricemia and hypermagnesemia are
in exon 9 of the aldolase B gene and therefore, no liver biopsy for the the laboratory hallmarks. Several of these findings
determination of the enzyme activity of aldolase B was needed. The pointed to the diagnosis in our case. To the best of our
patient was treated with a diet low in fructose and sucrose (maximum knowledge, this is the first report with casual diagnosis of
15 g fructose/day) with the results of physical wellness and catch-up
HFI by means of the fructose breath hydrogen test. This
growth without hormone therapy.
possibility has to be taken into account when a hydrogen
breath test is intended to be performed because an oral
fructose load with 1 up to 2 g/kg b.w. (we give a maximum
of 25 g) is considerably high. This amount of fructose is

Fructose Breath Hydrogen Test – Really a Dig Dis 2003;21:276–278 277


Harmless Diagnostic Procedure?
even higher than the dose used for an oral fructose loading under domestic conditions is risky and therefore, we rec-
test, which is meanwhile considered as an obsolete proce- ommend to perform this procedure under medical super-
dure to diagnose HFI [19]. Patients with HFI who are not vision and blood glucose monitoring (0, 30 and 60 min),
recognized within the first year of life very often develop for instance at a pediatric day-ward. In order to prevent
aversions against all sweet foods. This habit becomes such incidents as in our case, we recommend the follow-
gradually accepted by the parents, and the diet will be ing diagnostic work-up: If there are any doubts regarding
adapted by preparations that avoid symptoms like vomit- fructose tolerance after exploring the dietary history, a
ing and/or abdominal pain. For these reasons, it was PCR-based mutational analysis of the aldolase B gene
reported several times that patients with unrecognized should be performed because the prevalence of HFI car-
HFI were diagnosed during childhood or even during riers in Caucasians is about 1:50 [17]. If no relevant muta-
adulthood [12, 20]. Moreover, tragedies of iatrogenic tion can be detected, the fructose breath hydrogen test is
deaths related to medicinal use of fructose-based intrave- indicated to exclude fructose malabsorption. But it should
nous solutions are known [21]. As a consequence, before be pointed out that the absence of hydrogen-producing
an oral fructose load test is performed, it is necessary to bacteria in intestine can lead to false-negative results.
get exact information about the subject’s nutritional hab- Therefore, it is advisable to perform a following breath
its. HFI is probable when an avoidance to fruits or sweets test with lactulose which is not absorbed by the small bow-
is reported. The mutational analysis is then a feasible el. In dependence of hydrogen production, we investigate
first-line investigation because in 93% of the German HFI an intestine biopsy and measure the enzyme activity of
patients there is a prevalence of three mutations, which sucrase. In cases with normal enzyme activities, other
can easily be detected [18]. Considering the propagan- causes of chronic abdominal pain than fructose-induced
dized practicability of the fructose hydrogen breath test, disturbances have to be examined.
especially in outpatients, we think that the fructose load

References

1 Ramussen JJ, Gudmand-Hoyer E: Absorption 7 Bond, JH, Levitt MD: Investigation of small 14 Steinmann B, Gitzelmann R, van den Berghe
capacity of fructose in healthy adults. Compari- bowel transit time in man utilizing pulmonary G: Disorders of fructose metabolism; in Scriver
son with sucrose and its constituent monosac- hydrogen (H2) measurements. J Lab Clin Med CR, Beaudet AL, Sly WS, Valle D (eds): The
charides. Gut 1986;27:1161–1168. 1975;85:546–555. Metabolic and Molecular Bases of Inherited
2 Ledochowski M, Widner B, Bair H, Probst T, 8 Romagnuolu J, Schiller D, Bailey RJ: Using Disease, ed 8. New York, McGraw-Hill, 2001,
Fuchs D: Fructose- and sorbitol-reduced diet breath tests wisely in a gastroenterology prac- pp 1489–1520.
improves mood and gastrointestinal distur- tice: An evidence-based review of indications 15 Chambers RA, Pratt RTC: Idiosyncrasy to
bances in fructose malabsorbers. Scand J Gas- and pitfalls in interpretation. Am J Gastroen- fructose. Lancet 1956;271:340.
troenterol 2002;35:1113–1126. terol 2002;97:1113–1126. 16 Hers HG, Joassin G: An anomaly in hepatic
3 Burant CF, Takeda J, Brot-Laroche E, Bell GI, 9 Hoekstra JH: Fructose breath hydrogen test in aldolase during fructose intolerance. Enzymol
Davidson NO: Fructose transporter in human infants with chronic non-specific diarrhoea. Biol Clin 1961;1:4–14.
spermatozoa and small intestine is GLUT-5. J Eur J Pediatr 1995;154:362–364. 17 Tolan DR: Molecular basis of hereditary fruc-
Biol Chem 1992;267:14523–14526. 10 Götze H, Mahdi A: Role of fructose malabsorp- tose intolerance: Mutations and polymor-
4 Wassermann D, Hoekstra JH, Tolia V, Taylor tion in dysfunctional bowel disorders. Mo- phisms in the human aldolase B gene. Hum
CJ, Kirschner S, Takeda J, Bell GI, Taub R, natsschr Kinderheilkd 1992;140:814–817. Mutat 1995;6:210–218.
Rand EB: Molecular analysis of the fructose 11 Cox TM: Fructose intolerance: Diet and inheri- 18 Müller P, Koppelt B, Böhme HJ, Bührdel P:
transporter gene (GLUT-5) in isolated fructose tance. Proc Nutr Soc 1991;50:305–309. Detection of DNA mutations in German pa-
malabsorption. J Clin Invest 1996;98:2398– 12 Wössmann W, Wiemann J, Körber F, Görtner tients with hereditary fructose intolerance.
2402. L: Hereditäre Fruktoseintoleranz (HFI) als Ur- Amino Acids 1997;12:389.
5 Helliwell, PA, Richardson M, Affleck J, Kel- sache einer isolierten Á-GT-Erhöhung bei ei- 19 Koppelt B, Pöge AP, Müller P, Bührdel P,
lett, GL: Regulation of GLUT-5, GLUT-2 and nem 5-jährigen Jungen mit Hepatomegalie. Böhme HJ: Molecular biological investigation
intestinal brush-border fructose absorption by Klin Pädiatr 2000;212:108–109. in a family with hereditary fructose intolerance
the extracellular signal-regulated kinase, p38 13 Kneepkens CMF, Vonk RJ, Fernandes J: In- and diagnostic consequences. Monatsschr Kin-
mitogen-activated kinase and phosphatidylino- complete intestinal absorption of fructose. derheilkd 1996;144:383–386.
sitol 3-kinase intracellular signalling pathways: Arch Dis Child 1984;59:735–738. 20 Burmeister LA, Valdivia T, Nuttall FQ: Adult
Implications for adaptation to diabetes. Bio- hereditary fructose intolerance. Arch Intern
chem J 2000;50:163–169. Med 1991;151:773–776.
6 Barnes G, McKellan W, Lawrence S: Detection 21 Cox TM: Iatrogenic deaths in hereditary fruc-
of fructose malabsorption by breath hydrogen tose intolerance. Arch Dis Child 1993;69:413–
test in a child with diarrhea. J Pediatr 1983; 415.
103:575–577.

278 Dig Dis 2003;21:276–278 Müller/Meier/Böhme/Richter


Dig Dis 2003;21:279–285
DOI: 10.1159/000073985

Screening for Iron Overload in the


Turkish Population
Gultekin Barut a Huriye Balci b Mithat Bozdayi d Ibrahim Hatemi a
Dervis Ozcelik c Hakan Senturk a
a Department of Internal Medicine, b Central Research Laboratory, c Department of Biophysics,
Cerrahpasa Medical Faculty, University of Istanbul, Istanbul, and d Institute of Hepatology, Ankara Medical Faculty,
University of Ankara, Ankara, Turkey

Key Words mination of TS after an overnight fast was performed in


Hereditary hemochromatosis W HFE gene mutation W 135 subjects. In 26 subjects, the TS was 650% in the fast-
Epidemiology W Iron overload, prevalence ing state. HFE mutation and serum ferritin levels were
measured in these 26 subjects. Eleven subject (10 male, 1
female) were heterozygote and 1 male subject was ho-
Abstract mozygote in reference to H63D. C282Y mutation was not
Background/Aims: Hereditary hemochromatosis (HH), found. Four of these 26 subjects (all males, aged 23, 24,
the most common autosomal recessive disease in the 40, 49) had increased serum ferritin levels and liver biop-
white population, is characterized by excessive gastroin- sy was performed. In 1 male (aged 49) who was hetero-
testinal absorption of iron and loading of parenchymal zygote for H63D genotype with a serum ferritin level of
organs. HFE mutations of C282Y and H63D are largely 645 ng/ml, iron overload in liver tissue was shown by
responsible for HH in populations of Celtic ancestry. histology as well as atomic absorption spectrophotome-
Although many screening studies related to HH have try. Conclusion: The prevalence of hemochromatosis in
been done in Northern Europe, the USA and Australia, as the Turkish population is much lower in comparison to
yet, no such study has been published on Turkey. In this populations of Celtic ancestry and C282Y mutation is
study we aimed to screen the Turkish population for iron non-existent.
overload. Methods: Random samples were obtained Copyright © 2003 S. Karger AG, Basel

from 4,633 healthy adults (3,827 male, 806 female, mean


age B SD 35 B 8 years, range 14–76) for the measure-
ment of transferrin saturation (TS). Measurements were Introduction
repeated after an overnight fast in the subjects whose
initial TS was 650%. Serum ferritin levels and C282Y Hereditary hemochromatosis (HH) is the most com-
and H63D gene mutations were studied in cases when mon autosomal recessive disease in the white population
fasting TS was 650%. In cases where the serum ferritin [1–3]. Typical clinical symptoms do not develop until
level was 1 200 ng/ml with or without HFE mutations, liv- very late stages of the disease at which time treatment is
er biopsy was performed for histological evaluation and not rewarding. The value of screening studies largely by
determination of iron content. Results: In 158 subjects, means of measurement of transferrin saturation (TS) was
TS was 650% in the non-fasting state. A second deter- evaluated in several populations [1–10]. Mutation of HFE

© 2003 S. Karger AG, Basel Hakan Senturk, MD, Professor in Medicine


ABC 0257–2753/03/0213–0279$19.50/0 Cihangir, Akyol cad. No:18-5 Makbul apt
Fax + 41 61 306 12 34 TR–80060 Istanbul (Turkey)
E-Mail karger@karger.ch Accessible online at: Tel./Fax +90 212 5299565
www.karger.com www.karger.com/ddi E-Mail drhakansenturk@yahoo.com
Fig. 1. Frequency distribution of transferrin
saturation in 4,633 participants.

gene, is largely responsible for this disease in populations of Feder et al. Serum ferritin levels were measured by an immunoas-
of Celtic origin [6, 11]. To date, 37 allelic variants of the say system by chemiluminescent immunoassay method in 26 sub-
jects who had 650% fasting TS and were analyzed for HFE gene
HFE gene have been reported – C282Y and H63D are the
mutations.
most common. The gene frequency is 5% in the Anglo- Percutaneous needle biopsy of the liver were performed in 4 sub-
Saxon population. Homozygote HH prevalence is be- jects who had a fasting TS of 650% with a serum ferritin level
tween 0.3 and 0.5%, and the heterozygote carrier rate is 1 200 ng/ml. The tissue specimens were sent for routine histological
approximately 10% in European populations [1–5, 12, examination with Prussian blue staining. Liver iron content was
measured by atomic absorption spectrophotometry and hepatic iron
13]. As yet, no such screening study has been published on index (HII) was calculated by dividing liver iron concentration in
Turkey. HH is known as an inherently rare disease in this Ìmol Fe/g dry liver weight with subject’s age in years.
country. In this study we aimed to screen the Turkish pop-
ulation for iron overload.

Results

Methods
The mean TS was 29% for males and 25% for females
We included 4,633 healthy subjects (3,827 men, 806 women; (fig. 1). In 158 subjects (142 men and 16 women; mean
mean age B SD 35 B 8, range 14–76) who were employees of Istan- age B SD 36 B 8 years, range 17–52) who constituted
bul’s gas distribution, and water and sewage companies. Ten millili- 3.4% of total screened population, TS was 650% in the
ters of venous blood was obtained from non-fasting volunteers. non-fasting state. A second determination of TS after an
Serum was separated by centrifugation and frozen at –70 ° C until
analysis. Serum iron levels and total iron binding capacity (TIBC) overnight fast was performed in 135 of 158 subjects.
were measured by a standard colorimetric method on an automated Twenty-three subjects either could not be re-contacted or
analyzer (model Au-800, Olympus). Serum TS was calculated as denied further examination. In 26 subjects (24 men and 2
serum iron/TIBC ! 100. In cases where the TS value was 650%, the women; mean age B SD 34 B 8 years, range 22–52), who
participant was re-contacted to obtain a second sample in the fasting
constituted 0.6% of total screened population and 19% of
state and measurement of TS was repeated.
Fifteen-milliliter whole-blood samples were obtained from 26 re-evaluated population, the TS was 650% in the fasting
subjects whose fasting TS was 650%. These blood samples were state. Analysis of HFE mutations was done in these 26
immediately transported to the laboratory within 24 h at +4 ° C for subjects (fig. 2). C282Y mutation was not detected. Ho-
analysis of HFE gene mutations. DNA was prepared from these mozygote H63D mutation was found in only 1 male sub-
whole-blood samples by a method reported elsewhere [14]. The two
ject who had a normal serum ferritin level. Heterozygote
HFE mutations were screened with PCR assay followed by restric-
tion-enzyme digestion with Rsa1 for the C282Y mutation and Bcl1 H63D mutation was found in 10 males and 1 female.
for H63D mutation. PCR amplification of the regions containing Serum ferritin levels were also measured in 26 subjects. In
these two HFE mutations was performed with the primer sequences 4, the serum ferritin level was 1200 ng/ml, while in 1 of

280 Dig Dis 2003;21:279–285 Barut/Balci/Bozdayi/Hatemi/Ozcelik/


Senturk
Fig. 2. The algorithm of screening.

these 4 subjects, the serum ferritin level was 645 ng/ml; in This subject had heterozygote H63D genotype. Remain-
3 other subjects, it was !300 ng/ml (table 1) – all 4 were ing three subjects did not have either increased liver iron
males. In the subject with a serum ferritin level of content or staining. The HII of these 3 subjects was !1.9
645 ng/ml, an increased liver iron content (133.05 Ìmol/g (table 1). Only 1 of these 3 subjects had heterozygote
and HII = 2.71) and intensive iron staining were found. H63D genotype (HII = 0.67) (table 1).

Hereditary Hemochromatosis in Turkey Dig Dis 2003;21:279–285 281


Table 1. Laboratory features of participants whose phenotypes and since it is inherited autosomal recessively. However, the
genotypes were analyzed prevalence of HH and iron overload was higher in males
Hepatic iron Hepatic Serum Transferrin Mutation Mutation Age
than females in previous screening studies which had
concentration iron ferritin saturation C282Y H63D years been based on the phenotyping expression of the disease
Ìmol/g index ng/ml % [4, 6]. This can be explained by decreased clinical expres-
85.0 51.2 –/– –/– 34 sion resulting from menstrual blood loss and pregnancy in
37.4 51.4 –/– –/– 28 women. Many epidemiologic studies about HH and iron
37.27 1.55 238.5 90.6 –/– –/– 24 overload have been reported from different countries and
57.2 66.8 –/– –/+ 39 populations in the last 15 years. The prevalence of iron
39.9 52.0 –/– –/+ 27
overload resulting from HH was found to be 0.36% in
100.2 65.3 –/– –/– 40
36.3 59.8 –/– –/– 34 Australia where the population is predominantly com-
33.6 55.8 –/– –/+ 37 posed of whites of European descent [2]. The prevalence
67.2 64.9 –/– –/+ 29 of iron overload attributable to HH was reported as 0.40,
47.8 51.2 –/– –/+ 29 0.42 and 0.45% from different ethnic populations of the
25.20 1.09 200.8 52.3 –/– –/– 23
USA [6–8]. The prevalence was lower in blacks [3, 6, 15].
116.6 51.0 –/– –/– 27
53.1 65.2 –/– –/+ 31 Different approaches have been proposed for screening
42.9 51.4 –/– +/+ 25 HH and iron overload. The choice of screening method de-
27.8 52.4 –/– –/– 29 pends on whether one is seeking to detect all homozygotes
17.1 52.9 –/– –/– 42 or only those with iron overload [13]. As screening tests, TS,
69.6 60.1 –/– –/– 28
unsaturated iron binding capacity, serum ferritin level and
190.0 55.1 –/– –/+ 52
161.3 71.9 –/– –/– 42 HFE gene analysis were used in combination or alone.
86.6 67.9 –/– –/+ 40 Because of its ease of application, low cost, high sensi-
26.81 0.67 285.4 58.8 –/– –/+ 40 tivity and acceptable specificity, TS was the initial screen-
70.9 63.8 –/– –/+ 36 ing test in most of the previous studies [1–10]. The prob-
146.3 55.5 –/– –/– 29
lems with TS are related to its ill-defined threshold value
133.05 2.71 645.2 52.5 –/– –/+ 49
68.7 56.5 –/– –/+ 34 as well as low positive predictivity. The threshold value in
103.0 64.8 –/– –/– 30 previous studies has ranged from 45 to 70% [1, 5–8, 13,
15–17]. The rate of false positivity was intended to be
reduced with repeated measurements of TS after an over-
night fast in the subjects who had higher initial non-fast-
ing TS. Lower threshold TS values have been used for
Discussion females compared to males in previous studies. It has
been reported that a TS of 60% or more, identifies nearly
Turkey bridges the Middle East, Middle Asia and all homozygotes with iron loading, whereas a TS of 50%
Europe and has a population of over 60 million. The cur- identifies nearly all homozygotes regardless of sex or iron
rent Turkish population is a mixture of people who loading [13]. We set the threshold value to 50% to detect
migrated from Middle Asia (now Mongolia) around 1071 all homozygotes regardless of sex.
AD and the indigenous population of Anatolia. Over 95% A combination of determination of iron status with
of the population is Muslim and the rate of alcohol con- phenotyping methods associated with DNA testing have
sumption is much lower in comparison to European coun- the best sensitivity and specificity to detect risk from iron
tries and the USA. overload [7]. We used a strategy which was reported as
This study was performed in Istanbul and consisted of worthy of consideration in a previous study and started
4,633 workers from sewage and gas distribution compa- with determination of TS followed by an HFE gene analy-
nies. Istanbul is Turkey’s melting pot and most of the sis in subjects with elevated fasting TS [7].
workers have come from different parts of Anatolia in the In the four screening programs which were performed
last decade. Therefore, this population represents not only in the USA, the TS was elevated on initial testing in 2.5–
Istanbul but Turkey in general. 5.8% of the several screened populations [8]. The ratio
HH is the most common autosomal recessively inher- was 6.2% in the Edwards’ study [4]. The prevalence of an
ited disease in the white population [1–3]. The prevalence elevated initial TS rate of 3.4% in our screened popula-
of HH is supposed to be equal in both males and females, tion was similar to these studies.

282 Dig Dis 2003;21:279–285 Barut/Balci/Bozdayi/Hatemi/Ozcelik/


Senturk
A second determination of TS after an overnight fast small proportion of patients with iron loading [16]. The
was performed in 135 of 158 subjects with elevated initial average prevalences of homozygote and heterozygote
TS. All drop-outs appeared at this stage of screening. The C282Y mutations were reported to be 0.4 and 9.2%
compliance rate for a second determination of TS after an respectively from different European countries [16]. Al-
overnight fast was calculated as 85.4% (135/158). The though C282Y homozygosity has not been reported,
same rate in Phatak’s study, Edwards’ study and in the C282Y heterozygosity has been found to be 1–3% in
other two United States’ studies were 80.2% (747/932), Southern or Eastern Europe populations [16]. The highest
67.6% (465/688), 85.0% (1,147/1,349) and 87% (47/53) prevalence of heterozygote C282Y mutation has been
respectively [4, 6, 8]. In 26 subjects (24 men and 2 wom- reported as 24.8% from Ireland [16]. The prevalence of
en), who constitute 19.2% (26/135) of our re-evaluated homozygosity for C282Y mutation is 0.5% and the preva-
population, the TS was 650% in the fasting state in our lence of heterozygosity is 9% in North America [16].
study. The rates of elevated fasting TS in Edwards’ study, C282Y homozygosity was not found in the Asian-Indian
in Kaiser Permanente Medical Group’s study and in Pha- subcontinent, African/Middle Eastern and Australasian
tak’s study were 24.5% (114/465), 23% (73/318) and populations, and the frequency of C282Y heterozygosity
10.9% (82/747) (the threshold value for TS was 55% in was very low (0–5%) [16]. The prevalences of compound
this study), respectively [4, 6, 8]. These rates are also simi- heterozygote (C282Y/H63D) and homozygote H63D
lar to rates of the present study. have been found to be approximately 2% in European
In our study, 15.4% (4/26) of the subjects with elevated populations [16]. The prevalences of compound heterozy-
fasting TS had 6200 ng/ml serum ferritin levels. The val- gotes (C282Y/H63D), homozygote H63D and heterozy-
ue was 50.2% (128/255) in Phatak’s study [6]. In our pop- gote H63D were reported 2.5, 2.1 and 23% respectively in
ulation, most of the subjects with elevated fasting TS did North American populations [16]. The prevalence of het-
not have increased serum ferritin levels which is an erozygote H63D was 15–22% in European populations
important marker for liver iron content [6]. [9, 16]. Another study showed that only 64% of patients
Liver biopsy was performed in 4 subjects who had with hemochromatosis in Italy were homozygotes for the
increased serum ferritin. Iron overload was detected in C282Y mutation [19]. The study performed in Hong
only 1 subject in our study. In conclusion, the prevalence Kong showed that the C282Y mutation was not found in
of iron overload was 0.021% in our population. The prev- Chinese patients with hepatic iron overload disease [20].
alence of iron overload was reported as 0.36% from Aus- In a review by Lucotte [11] concerning 20 European pop-
tralia and 0.4, 0.42 and 0.45% from different ethnic popu- ulations, the highest C282Y allele frequency (6.88%) was
lations of the USA [2, 6–8]. observed in residual Celtic populations in the UK and
The candidate gene leading to hemochromatosis was France. This study showed that C282Y allele frequencies
first discovered in 1996 by Feder et al. on the short arm of were distributed among a decreasing line from north to
chromosome 6, and was termed the ‘HFE gene’ [18]. This south of Europe [11]. The lowest C282Y allele frequencies
HFE gene mutation is largely responsible for HH and were observed in Italians (1.00%), Ashkenazi Jews
related iron overload in populations of Celtic origin [6, (1.31%) and French Basques (1.63%) [11]. These data
11]. The interest was focused on C282Y and H63D muta- suggest that HFE gene mutation is probably of Celtic ori-
tions. The C282Y mutation results from G to A transition gin.
at nucleotide 845 of the HFE gene (845G→A) that pro- In our study, analysis of HFE mutations was done in
duces a substitution of cysteine for a tyrosine at amino 26 subjects whose fasting TS values were 650%. C282Y
acid position 282 in the protein product [16]. In the mutation was not detected. Homozygote H63D mutation
H63D mutation, a G replaces C at nucleotide 187 of the was found in only 1 subject, and heterozygote H63D
gene (187C→G), causing aspartate to substitute for histi- mutation was found in 11 subjects. The subject who had
dine at amino acid position 63 in the HFE protein [16]. In homozygote H63D mutation did not have any clinical
the last 10 years, studies performed in populations of symptom for hemochromatosis. In this subject, there was
largely Celtic descent showed that 60–100% of patients no clinical expression of HH except fasting TS (fasting
with typical HH were homozygotes for C282Y mutation TS: 51.3%) and the serum ferritin level was within normal
[3, 7, 9, 12, 15–17, 19]. In the general population, com- ranges (serum ferritin: 42.9 ng/ml). Our data are insuffi-
pound heterozygote (C282Y/H63D) and homozygote cient to comment on the prevalence of HFE mutations in
H63D genotypes occur more frequently than homozygote the general population of this country.
C282Y, but these genotypes are responsible for only a

Hereditary Hemochromatosis in Turkey Dig Dis 2003;21:279–285 283


In previous studies, different populations were chosen hemochromatosis as compared to a matched normal popu-
for screening hemochromatosis – blood donors [1, 4, 17], lation [12]. The risk of death from liver cancer in a patient
hospital inpatients [3, 5], primary care patients [6], bank with hemochromatosis is more than 100 times that of the
and insurance company employees [2], employees of matched healthy subjects [4]. There is a long asymptomatic
health maintenance organizations [7]. The advantages of phase in HH. The fact that hemochromatosis has a high
using blood donors for screening were expounded in the prevalence, a potential to cause substantial morbidity and
screening study of Adams’ group [1, 17]: (1) exclusion of mortality, a long asymptomatic phase and prevention of
persons with conditions such as hepatitis B and C and disease-associated complications by means of early diag-
sometimes elevated liver enzyme values reduce the num- nosis and treatment such as phlebotomy, make screening
ber of false-positive screening tests; (2) blood has already studies a cost-effective strategy [1, 13].
been obtained for other reasons; (3) systems are well Recent studies have reported that hemochromatosis
established for collection and distribution of samples, and screening based on TS testing is cost-effective [7]. The
(4) donors represent a young asymptomatic population cost analysis study of Adams and Valberg [17], which was
that would benefit from the early detection of hemochro- one of the largest comparing genotyping and phenotyping
matosis. In spite of all these advantages, there are some models, showed that an optimal strategy for screening
disadvantages of using blood donors, for example; the includes initial testing for iron overload (phenotyping)
false negative results for measurement of TS and serum with confirmatory genetic testing, or initial genetic testing
ferritin may occur because regular donation may cause if the test is less than USD 28 for each person screened. In
false low iron stores [6], subjects with elevated liver this study, it was reported that the cost of detecting one
enzyme concentrations and sideroblastic anemia who homozygote with potential life-threatening illness by ini-
may have HFE gene mutation are excluded from the tial testing for iron overload (phenotyping) with confirma-
blood donor pool. Hospital inpatients are, by nature, tory genetic testing was USD 2,711 [17]. For cost analysis,
selected groups, therefore they do not represent the gener- 10,000 blood donors were used and the prevalence of HH
al population. The other screening populations are less was 0.3% [17]. Adams and Valberg [17] concluded that
selective, but the actual prevalence of disease in the gener- the cost of detection of one homozygote with HH by ini-
al population cannot be detected with these groups be- tial testing for iron overload (phenotyping) with confirma-
cause they may not contain a sufficient enough number of tory genetic testing can be higher in a population with a
subjects. Increasing the number of screened subjects lower prevalence of the disease.
makes the investigation difficult due of logistic and eco- The prevalence of iron overload in our study was
nomical reasons. However, we believe that our screened 0.021%. Homozygote C282Y mutation was not found
population was sufficiently large enough to represent a and the prevalence of homozygote H63D mutation was
general population. 0.021%. The cost of HFE gene mutation analysis for each
The serologic tests of hepatitis B and C were done, and person was USD 75 in our study. The total cost of the
none of the HFE-gene-analyzed subjects with 650% fast- study was calculated approximately USD 50,000 by using
ing TS had HBsAg or anti-HCV. Most of the subjects in cost analysis. This total cost was equal to the cost of detec-
our study are teetotalers and none consumed 180 g/day tion in the only subject with iron overload. However, it
alcohol. seems that such a screening is not cost-effective in the
The age range in this study was 14–76 years. The mini- Turkish population; the rationality of prevention of one
mum age in Edwards’, Phatak’s and Olynyk’s studies were case of cirrhosis with eventual decompensation necessi-
17, 18 and 20 years, respectively [4, 6, 9]. The maximum tating liver transplantation or death at a cost of USD
age in this study was similar to the maximum age of pre- 50,000 should be further scrutinized.
vious ones. The mean age was 35.7 in males and 32.2 in In conclusion, the prevalence of HH and iron overload
females in our study. It was 37.5 in males and 34.7 in in the Turkish population is much lower in comparison to
females in Edwards’ study [4]. The major limitation of North European populations of Celtic ancestry and
our study was that males predominated (83%). C282Y mutation as a cause of HH is non-existent.
Life-threatening clinical manifestations, such as liver
failure, heart failure and diabetes mellitus, usually occur
Acknowledgement
after a latent period of 40–60 years in subjects with iron
overload [1]. The risk of death as a direct consequence of This work was supported by the Research Fund of The University
liver cirrhosis is significantly increased in patients with of Istanbul, Project No. T-825/07032000.

284 Dig Dis 2003;21:279–285 Barut/Balci/Bozdayi/Hatemi/Ozcelik/


Senturk
References

1 Adams PC, Gregor JC, Kertesz AE, Valberg 7 McDonnell SM, Hover A, Gloe D, Ou CY, 14 Boretto J, Jouanolle AM, Yaouaq J, et al: Ano-
LS: Screening blood donors for hereditary he- Cougswell ME, Grummer L: Population-based nymous markers located on chromosome 6 in
mochromatosis: Decision analysis model based screening for hemochromatosis using pheno- the HLA-A class I region: Allelic distribution in
on a 30-year database. Gastroenterology 1995; typic and DNA testing among employees of genetic haemochromatosis. Hum Genet 1992;
109:177–188. health maintenance organizations in Spring- 89:33–36.
2 McLaren CE, McLachlan GJ, Halliday JW, field, Missouri. Am J Med 1999;107:30–37. 15 Cogswell ME, McDonnell SM, Khoury MJ,
Webb SI, Leggett BA, Jazwinska EC, Crawford 8 McDonnell SM, Phatak PD, Felitti V, Hover Franks AL, Burke W, Brittenham G: Iron over-
DHG, Gordeug VR, McLaren GD, Powell LW: A, McLaren GD: Screening for hemochroma- load, public health and genetics: Evaluating the
Distribution of transferrin saturation in an tosis in primary care settings. Ann Intern evidence for hemochromatosis screening. Ann
Australian population: Relevance to the early Med1998;129:962–970. Intern Med 1998;129:971–979.
diagnosis of hemochromatosis. Gastroenterol- 9 Olynyk JK, Cullen DJ, Aquilia S, Rossi E, 16 Hanson HE, Imperatore G, Burke W: HFE
ogy 1998;114:543–549. Summerville L, Powell LW: A population- gene and hereditary hemochromatosis. HuGE
3 Beutler E, Felitti V, Gelbart T, Ho N: The based study of the clinical expression of the Rev IPDAS #549 (March 12, 2001).
effect of HFE genotypes on measurements of hemochromatosis gene. N Engl J Med 1999; 17 Adams PC, Valberg LS: Screening blood do-
iron overload in patients attending a health 341:718–724. nors for hereditary hemochromatosis: Decision
appraisal clinic. Ann Intern Med 2000;133: 10 McLaren CE, Gordeug VR, Looker AC, Has- analysis model comparing genotyping to phe-
329–337. selblad V, Edwards CQ, Griffen LM, Kushner notyping. Am J Gastroenterol 1999;94:1593–
4 Edwards CQ, Griffen LM, Goldgar D, Drum- JP, Brittenham GM: Prevalence of heterozy- 1600.
mond C, Skolnick MH, Kushner JP: Preva- gotes for hemochromatosis in the white popula- 18 Feder JN, Gnirke A, Thomas W, et al: A novel
lence of hemochromatosis among 11,065 pre- tion of the United States. Blood 1995;86:2021– MHC class-I like gene is mutated in patients
sumably healthy blood donors. N Engl J Med 2027. with hereditary hemochromatosis. Nat Genet
1988;21:1355–1362. 11 Lucotte G: Celtic origin of the C282Y mutation 1996;13:399–408.
5 Balan V, Baldus W, Fairbanks V, Michels V, of hemochromatosis. Blood Cells Mol Dis 19 Pietrangelo A, Montosi G, Totaro A, Garuti C,
Burrit M, Klee G: Screening for hemochroma- 1998;24:433–438. Conte D, Cassanelli S, Fraquelli M, Sardini C,
tosis: A cost-effectiveness study based on 12 Niederau C, Erhard A, Haussinger D, Stroh- Vasta F, Gasparini P: Hereditary hemochro-
12,258 patients. Gastroenterology 1994;107: meyer G: Haemochromatosis and liver. J He- matosis in adults without pathogenic muta-
453–459. patol 1999;30:6–11. tions in the hemochromatosis gene. N Engl J
6 Phatak PD, Sham RL, Raubertas RF, Dunni- 13 Edwards CQ, Kushner JP: Screening for hemo- Med 1999;341:725–732.
gan K, O’Leary MT, Braggins C, Cappuccio chromatosis. N Engl J Med 1993;328:1616– 20 Tsui WM, Lam PW, Lee KC, Ma KF, Chan
JD: Prevalence of hereditary hemochromatosis 1620. YK, Wong MW, Yip SP, Wong CS, Chow AS,
in 16,031 primary care patients. Ann Intern Lo ST: The C282Y mutation of the HFE gene
Med 1998;129:954–961. is not found in Chinese haemochromatotic pa-
tients: Multicentre retrospective study. Hong
Kong Med J 2000;6:153–158.

Hereditary Hemochromatosis in Turkey Dig Dis 2003;21:279–285 285


Author Index Vol. 21, No. 3, 2003

Astuto, M. 271 Ebert, M. 237 Matsumoto, T. 266


Avgerinos, C. 214 Fernández, R. 258 Meier, C. 276
Balci, H. 279 Finocchiaro, G. 271 Müller, P. 276
Barut, G. 279 Fleta, J. 258 Olivares, J.L. 258
Baumann, G. 245 Gassull, M.A. 220 Ozcelik, D. 279
Böhme, H.J. 276 Gizaris, V. 262 Palesty, J.A. 198
Bozdayi, M. 279 Hatemi, I. 279 Paulisch, E. 252
Botsios, D.S. 228 Iida, M. 266 Pennisi, G. 271
Burmester, G.-R. 245 Kemps, M. 245 Pirlich, M. 245
Clavel, A. 258 Kuroki, F. 266 Pistone, G. 271
Dafnopoulou, A. 262 Kyriazanos, I.D. 262 Plauth, M. 245
Datsakis, K. 262 Lange, K.-P. 252 Richter, T. 276
Delis, S. 214 Lo Giudice, E. 271 Rizos, S. 214
Dell’Arte, S. 271 Lochs, H. 196, 245, 252 Rodrı́guez, G. 258
Dervenis, C. 196, 214 Lübke, H.J. 245 Schütz, T. 245, 252
Dierkes, J. 237 Luhman, N. 245 Senturk, H. 279
Dimakos, P. 262 Luley, C. 237 Sfiniadakis, I. 262
Drude, C. 252 Malaguarnera, M. 271 Vasiliadis, K.D. 228
Dudrick, S.J. 198 Malfertheiner, P. 237

Subject Index Vol. 21, No. 3, 2003

Acute pancreatitis 214 Gastrointestinal cancer 198 Malnourished hospitalized patients 245
Anorexia 198 – diseases 245 Malnutrition 198
Body mass index 262 Giardia lamblia 258 –, diagnoses 245
Cachexia 198 Growth factors 228 –, prevalence 245
Cancer 198 Gut adaptation 228 Obesity 262
– cachexia syndrome 198 – barrier 214 Pernicious anemia 237
L-Carnitine treatment 271 Helicobacter pylori 262 Selenium 266
Copper 258 – infection 237 Subjective global assessment 245
Crohn’s disease 252, 266 Hepatic encephalopathy 271 Sugar intake 252
Dental caries 252 Hereditary fructose intolerance 276 Taste changes 252
Enteral nutrition 214, 266 – hemochromatosis 279 Total parenteral nutrition 214
Enterobius vermicularis 258 HFE gene mutation 279 Vitamin B12 deficiency 237
Epidemiology 279 Homocysteine 237 Zinc 252, 258
Fasting blood glucose levels 262 Intestinal failure 228
Fructose breath hydrogen test 276 Iron overload, prevalence 279
– malabsorption 276 Magnesium 258

© 2003 S. Karger AG, Basel


ABC
Fax + 41 61 306 12 34
E-Mail karger@karger.ch Accessible online at:
www.karger.com www.karger.com/ddi

Вам также может понравиться