Nutritional Approach in Malnourished Surgical Patients, A Prospective
Randomized Study

Arch Surg. 2002;137:174-180

Author Information
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#aainfo>   Marco Braga,
MD; Luca Gianotti, MD, ScD; Luca Nespoli, MD; Giovanni Radaelli, PhD;
Valerio Di Carlo, MD
Hypothesis  Perioperative administration of a supplemented enteral formula
may decrease postoperative morbidity.
Design  Randomized clinical trial.
Setting  Department of surgery at a university hospital.
Patients  One hundred ninety-six registered malnourished patients (weight
loss 10%) who were candidates for major elective surgery for malignancy of
the gastrointestinal tract.
Intervention  After randomization (n = 150), one group received
postoperative enteral feeding with a standard diet within 12 hours of
surgery (control group; n = 50). Another group orally received 1 L/d for 7
consecutive days of a liquid diet enriched with arginine, omega-3 fatty
acids, and RNA (preoperative group; n = 50). After surgery, patients were
given the same standard enteral formula as the control group. A third group
orally received 1 L/d for 7 consecutive days of the enriched liquid diet.
After surgery, patients were given enteral feeding with the same enriched
formula (perioperative group; n = 50).
Main Outcome Measures  Postoperative complications and length of hospital
stay.
Results  The 3 groups were comparable for baseline demographics, biochemical
markers, comorbidity factors, and surgical variables. The intent-to-treat
analysis showed that the total number of patients with complications was 24
in the control group, 14 in the preoperative group, and 9 in the
perioperative group (P = .02, control group vs perioperative group).
Postoperative length of stay was significantly shorter in the preoperative
(13.2 days) and perioperative (12.0 days) groups than in the control group
(15.3 days) (P = .01 and P = .001, respectively, vs the control group).
Conclusion  Perioperative immunonutrition seems to be the best approach to
support malnourished patients with cancer.
Arch Surg. 2002;137:174-180
SOA1091
PROTEIN ENERGY malnutrition is recognized as an important risk factor for
the occurrence of postoperative complications. 1-3
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r1>  Thus, artificial
nutritional support has been proposed as an essential part of perioperative
care of malnourished surgical patients. 4-6
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r4>  Feeding enterally
rather than parenterally may improve outcome, particularly in malnourished
patients who undergo major surgery for a neoplasm. 7-12
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r7>
Moreover, administration of standard enteral diets supplemented with
arginine, omega-3 fatty acids, glutamine, and other key nutritional
substrates (immunonutrition) modulates immune and inflammatory responses and
gut function. 13 <http://archsurg.ama-assn.org/issues/v137n2/rfull/#r13> ,
14 <http://archsurg.ama-assn.org/issues/v137n2/rfull/#r14>
Results of 2 randomized, double-blind trials 15
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r15> , 16
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r16>  consistently
indicated that perioperative (before and after surgery) administration of
immunoenhancing diets could significantly improve several outcome variables,
but none of these studies were designed with separate randomization
according to the nutritional status of the patients. Results of a post-hoc
analysis 16 <http://archsurg.ama-assn.org/issues/v137n2/rfull/#r16>
suggested that the positive effects of immunonutrition were more pronounced
in a subgroup of malnourished patients. Another finding was that patients
receiving only preoperative immunonutrition, because of noncompliance with
postoperative immunonutrition, also had a significant reduction in
complications (M.B., L.G., G.R., et al, unpublished data, 1999). These data
suggested that the simple preoperative approach could be sufficient to
improve outcome.
Therefore, this study was designed with the primary end point of
establishing in a prospective fashion whether administration of
perioperative immunonutrition could reduce the rate of post-operative
complications and the length of postoperative hospital stay compared with
administration of standard enteral formulas in a homogeneous group of
patients defined a priori as malnourished. Furthermore, we evaluated, in the
policy of cost minimization, whether the simple preoperative administration
of immunonutrition could be as effective as the perioperative approach.



PATIENTS AND METHODS



This was a randomized clinical trial conducted by a single institution (San
Raffaele Hospital, Milan, Italy) between September 1, 1998, and December 31,
2000.
Inclusion and exclusion criteria are summarized in Table 1
<http://archsurg.ama-assn.org/issues/v137n2/fig_tab/soa1091_t1.html> .
Patients had to meet all 3 inclusion criteria to be registered. After
applying the exclusion criteria, patients were allocated by
computer-generated individual random numbers into 3 arms. Eligible patients
were required to sign a written informed consent form after the details of
the protocol were fully explained. The protocol was approved by the ethical
committee of San Raffaele Hospital.
Before surgery, one group drank 1 L of a supplemented liquid diet (Oral
Impact; Novartis Consumer Health, Bern, Switzerland) per day for 7
consecutive days. After surgery, patients continued to be fed enterally with
the same supplemented formula (perioperative group).
Before surgery, another group drank 1 L of a supplemented liquid diet (Oral
Impact) per day for 7 consecutive days. After surgery, patients were given a
standard enteral formula (preoperative group).
The third group received only postoperative enteral feeding with a standard
diet (control group).
The standard and enriched diets contained the same amounts of energy and
nitrogen ( Table 2
<http://archsurg.ama-assn.org/issues/v137n2/fig_tab/soa1091_t2.html> ).
Regardless the type of diet, in all 3 groups, postoperative enteral
nutrition was administered within 12 hours of surgery via a feeding catheter
jejunostomy or a nasojejunal feeding tube. Enteral flow was controlled by
using a peristaltic infusion pump. The initial rate of 10 mL/h was
progressively increased 20 mL/h per day until reaching the full nutritional
goal (28 kcal/kg per day). Enteral infusion was continued until patients
resumed adequate oral food intake (approximately 50% of the basal energy
requirement).
All patients were advised to consume regular food as desired before surgery.
In the preoperative and perioperative groups, presurgical oral
supplementation with the enriched formula was given as outpatient therapy.
Patients were also asked to register the daily amount of the preoperative
supplement consumed. Hospital admission was scheduled for all patients 2
days before surgery.
Eight days before surgery, the following baseline variables were determined
in all patients: body weight, degree of weight loss (with respect to usual
body weight in the previous 6 months), performance status according to
Karnofsky score, hemoglobin level, plasma level of total protein, albumin
level, retinol binding protein level, prealbumin level, total circulating
lymphocyte count, creatinine level, and arginine plasma level. The arginine
level was also determined 1 day before surgery. Comorbidity factors, as
measured by the American Society of Anesthesiologists score, were also
recorded in all patients.
Type of surgery, duration of surgery, operative blood loss, and rate and
amount of homologous blood transfused were registered. The decision to give
homologous blood was based on the perioperative hemoglobin level (<8 g/dL)
or the clinical condition of the patient.
Intestinal washout with an isosmotic solution (3 L) was carried out the day
before surgery in patients undergoing colorectal surgery. The evening before
and the morning of surgery, patients were also treated with enemas. These
patients received antibiotic prophylaxis in a single intravenous dose (2 g
of cefotetan disodium) 30 minutes before surgery. Candidates for gastric,
esophageal, or pancreatic surgery were treated with intestinal washout (1 L)
the day before surgery and with antibiotic prophylaxis in a single
intravenous dose (2 g of cefazolin sodium) 30 minutes before surgery. A
second dose of the antibiotic was given if the surgery lasted longer than 4
hours. Deep-vein thrombosis prophylaxis was carried out daily using
low-molecular-weight heparin (50 IU/kg).
Adverse effects of postoperative enteral feeding, such as abdominal cramping
and distension, diarrhea (defined as >3 liquid stools per day), and
vomiting, were evaluated in all patients, as was the time from canalization
to gas and bowel movement.
Patients were defined as nontolerant to postoperative feeding if they were
unable to tolerate a minimum diet infusion of 800 mL/d by postoperative day
4.
Trained members of the surgical staff who were not involved in the study
registered the postoperative complications (definitions are given in Table 3
<http://archsurg.ama-assn.org/issues/v137n2/fig_tab/soa1091_t3.html> ). They
also decided independently the day of hospital discharge, the indication,
and the duration of antibiotic therapy in the postoperative course and the
first day of oral food resumption. Total parenteral nutrition was given to
patients who were nontolerant of enteral nutrition.
Death, anastomotic leak, relaparotomy, complications requiring transfer of
the patient to the intensive care unit, and percutaneous drainage of a deep
abscess using interventional radiologic techniques were defined as major
complications. Follow-up for infectious and noninfectious complications was
carried out for 30 days after hospital discharge via office visits.
STATISTICAL ANALYSIS

It was assumed that postoperative complications would occur in approximately
40% of eligible control patients. 7
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r7> , 16
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r16>  A reduction in the
incidence to 20% would indicate the efficacy of perioperative
immunonutrition treatment. With a targeted maximum of 50 individuals in each
group, we had approximately 80% power to detect such a reduction at alpha=
.05.
All patients were analyzed on an intent-to-treat basis. Descriptive results
are given as mean (SD) or number (percentage) of observations. One-way
analysis of variance and the Kruskal-Wallis test (for nonnormally
distributed data, ie, operative time, operative blood loss, length of stay,
resumption of oral food intake, duration of antibiotic therapy) were used to
compare continuous variables among groups. Post-hoc multiple comparisons
were performed using the Bonferroni correction. The chi2 test (with Yates
correction if there were cells with <10 observations) and the Fisher exact
test were used to compare discrete variables. All P values are 2-sided, and
significance was set at P<.05. A statistical software program (SPSS version
8.0 for Windows; SPSS Inc, Chicago, Ill) was used for statistical analysis.



RESULTS



During the study, 196 patients were observed: 46 were excluded and 150 were
eligible and entered by randomization into the control group (n = 50), the
preoperative group (n = 50), or the perioperative group (n = 50). All
eligible patients were analyzed on an intent-to-treat basis ( Figure 1
<http://archsurg.ama-assn.org/issues/v137n2/fig_tab/soa1091_f1.html> ).
The mean preoperative intake of the oral supplementation was 910 mL/d, with
no difference between the preoperative and perioperative groups. Table 4
<http://archsurg.ama-assn.org/issues/v137n2/fig_tab/soa1091_t4.html>  lists
the preoperative (day -8) characteristics and the comorbidity factors of the
eligible participants. The 3 study arms were well-balanced for all
variables. Mean weight loss was 13%. Mean serum levels of total protein,
albumin, prealbumin, and retinol binding protein were below the reference
range in all groups. The arginine plasma level was similar 8 days before
surgery in all groups, whereas 1 day before surgery the level was 1.15 mg/dL
in the control group, 1.64 mg/dL in the preoperative group, and 1.69 mg/dL
in the perioperative group (P = .03). The 3 groups were comparable for
surgical variables ( Table 5
<http://archsurg.ama-assn.org/issues/v137n2/fig_tab/soa1091_t5.html> ). Most
patients underwent upper gastrointestinal tract operations.
Outcome variables are given in Table 6
<http://archsurg.ama-assn.org/issues/v137n2/fig_tab/soa1091_t6.html> . The
total number of patients who developed postoperative complications was
significantly lower in the perioperative group than in the control group (P
= .02). Moreover, the number of patients with complications was 9 (18%) of
50 in the perioperative group vs 14 (28%) of 50 in the preoperative group
(50% relative reduction), but this difference did not reach statistical
significance (P = .34). Also, comparing the preoperative and control groups,
a substantial reduction in complications was observed (28% vs 42%; P = .21).
Mean length of postoperative hospital stay was significantly shorter in the
preoperative (P = .01) and perioperative (P = .001) groups than in the
control group.
Table 7 <http://archsurg.ama-assn.org/issues/v137n2/fig_tab/soa1091_t7.html>
gives the complications in each group. Considering infectious morbidity,
respiratory tract and wound infections were the most frequent, with a lower
incidence in the groups receiving immunonutrition than in controls. Among
major complications, anastomotic leak seemed to be the most frequent, with a
similar trend toward reduction in both groups receiving the enriched diet.
The duration of antibiotic therapy needed to treat postoperative infections
was 6.1 (1.7) days in the perioperative group vs 5.8 (3.5) days in the
preoperative group and 10.2 (6.3) days in the control group.
One hundred four patients (69%) did not experience any adverse effects
related to postoperative enteral nutrition. Abdominal cramps or distension
were observed in 29 patients (19%), diarrhea in 13 (9%), and vomiting in 4
(3%). The rate of gastrointestinal tract adverse effects was similar in the
3 groups. Because most adverse effects were controlled by the temporary
interruption or reduction of the jejunal infusion, overall the nutritional
goal was achieved in 138 patients (92%). Twelve patients (8%) had to be
switched to total parenteral nutrition for intolerance (5 patients in the
control group, 3 in the preoperative group, and 4 in the perioperative
group).
Recovery of bowel function as measured by canalization to gas and feces
occurred after 3.5 (1.7) and 5.2 (2.1) days, respectively, without any
substantial difference among groups. Patients recovered adequate oral food
intake 9.8 (5.4) days after surgery in the control group, 8.2 (2.3) days in
the perioperative group, and 8.4 (3.6) days in the preoperative group.
Between surgery and hospital discharge, patients lost an additional 3.1% of
their body weight. The value was similar among groups.



COMMENT



Malnutrition in patients who are candidates for major surgery is a
considerable problem for the surgeon because it represents a risk factor for
postoperative morbidity and mortality. 1-3
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r1>  Cancer-bearing
patients may have additional protein energy depletion due to the occurrence
of cachexia. 17 <http://archsurg.ama-assn.org/issues/v137n2/rfull/#r17>
Despite extensive research in the field of clinical nutrition, 18
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r18>  indisputable
results on which to base rational nutritional support in malnourished
surgical patients are sparse.
A recent consensus statement 18
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r18>  concluded that
malnourished patients are candidates for preoperative artificial nutritional
support. This support could reduce the rate of postoperative complications
by approximately 10%. These general conclusions have several potential
flaws: (1) in the trials analyzed, malnutrition was defined by a multitude
of different scores and threshold values for weight loss and serum protein;
(2) the study design was rarely a priori dedicated to malnourished patients;
(3) enrolled patients were often heterogeneous for the primary diagnosis;
(4) the quantity of calories was, in most cases, excessive compared with
current standards; and (5) the type of nutrients and the presence of lipids
were frequently uncontrolled. These considerations made the comparison among
trial results puzzling and the conclusions difficult to be applied in
clinical practice. Moreover, nutritional support, with the suggested
modalities, requires preoperative hospitalization for at least 7 to 10 days,
with an obvious increase in sanitary costs. Another reason for skepticism is
that severe malnutrition (the only condition that benefits from preoperative
parenteral feeding) 19
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r19>  is often an
indirect sign of advanced neoplastic disease (unresectable tumor, metastatic
spread, peritoneal carcinosis, etc), which usually makes patients unsuitable
for a surgical approach. Therefore, the cost-effectiveness of this
nutritional treatment may be limited. 20
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r20>
Results of recent studies strongly suggest that in malnourished patients,
postoperative enteral feeding with standard feeds should be preferred to
parenteral nutrition. In fact, surgical patients fed enterally had an
improved outcome compared with patients treated parenterally, 7-12
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r7>  and the
nutrition-related sanitary costs are substantially reduced when using the
enteral route. 7 <http://archsurg.ama-assn.org/issues/v137n2/rfull/#r7> ,
21-23 <http://archsurg.ama-assn.org/issues/v137n2/rfull/#r21>  Thus, we
believe that postoperative enteral nutrition with a standard diet could be
considered the reference treatment. In the present study, we did not include
an unfed group because it was considered unethical to restrain such patients
from any support.
In the past decade, research in clinical nutrition focused on the addition
of key substrates (arginine, omega-3 fatty acids, glutamine, RNA, etc) to
standard formulas. Two meta-analyses 24
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r24> , 25
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r25>  showed that in
surgical patients, postoperative use of enteral immunonutrition seems to
improve outcome compared with standard formulas. Nevertheless, by analyzing
singularly the major studies, 26-30
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r26>  the results were
conflicting. In fact, the positive findings reported by Daly et al 26
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r26> , 27
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r27>  were partially
confirmed by others. 28-30
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r28>  Postoperative
treatment is conceptually limited because the amount of key substrates given
in the first days after surgery is little and therefore may be insufficient
for an efficacious modulation of the immune and inflammatory responses after
surgery. 31-34 <http://archsurg.ama-assn.org/issues/v137n2/rfull/#r31>  As
any other substance with supposed pharmacological action, immunonutrients
should reach suitable tissue and plasma concentrations to be active. Thus, a
different approach was tested to anticipate the provision of immunonutrients
before surgery to obtain high levels at the time of surgical stress, when
the depression of the host response is maximal.
Phase 2 clinical trials 35-37
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r35>  showed that
perioperative treatment (before and after surgery) with an enteral diet
enriched with arginine, omega-3 fatty acids, and RNA significantly prevented
early postoperative impairment of the host defense mechanisms, controlled
the overwhelming inflammatory reaction, improved intestinal microperfusion
and postoperative gut mucosal oxygen metabolism, and modulated the metabolic
response favoring the synthesis of constitutive proteins instead of
acute-phase proteins.
The impact of perioperative administration of immunoenhancing diets was
tested subsequently in phase 3 double-blind trials to address whether the
advantages listed in the previous paragraph on surrogate end points could
translate into true outcome benefits. Senkal and colleagues 15
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r15>  showed that
patients receiving enteral immunonutrition had significantly fewer
infections than the control group (14 vs 27). We 16
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r16>  reported that in
the intent-to-treat analysis, the overall rate of postoperative infectious
complications was 14% in the supplemented group and 30% in the control group
(P = .009), and length of hospital stay was also shorter in the supplemented
group (P = .01). Furthermore, our data suggested that perioperative
immunonutrition is efficacious regardless of the baseline nutritional status
of the patients. In fact, the rate of complication in well-nourished
patients was 10% (6/63) in the supplemented group vs 27% (14/68) in the
control group (P = .05). In a subgroup of malnourished patients, the rate of
complication was 14% (3/22) in the supplemented group vs 39% (7/18) in the
control group (P = .05).
Yet, these studies were not originally designed to randomize patients by
their nutritional status. To overcome this bias, we designed a prospective
study to investigate the effect of perioperative immunonutrition on outcome
in patients a priori homogeneous for nutritional status. In the present
trial, we also included a group receiving only preoperative immunonutrition
because previous results (M.B., L.G., G.R., et al, unpublished data, 1999)
suggested that patients receiving only preoperative immunonutrition because
noncompliant with postoperative immunonutrition had a significant reduction
in complications. This suggested that the simple preoperative approach could
be sufficient to improve outcome. Moreover, in the light of
cost-minimization policy, the restriction to preoperative treatment only may
save approximately $90 to $180 per treated patient. 15
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r15> , 38
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r38>
The results of the present trial are consistent with those of the previous
subset analysis and confirm that perioperative treatment with
immunonutrition may significantly decrease the postoperative morbidity rate
in malnourished patients. In contrast, the expected equivalence between
perioperative and preoperative immunonutrition was not achieved. This may be
explained by the fact that malnourished patients, beside having energy and
nitrogen needs, have marked impairment of the immune response, and, thus,
prolonged and increased administration of key nutrients is required. This
speculation is consistent with recent data 39
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r39>  suggesting instead
that in well-nourished patients, the simple provision of immunonutrients
before surgery was sufficient to improve outcome.
The present data showed a clear trend on the incidence of postoperative
complications according to the type of nutrition strategy. In particular, a
progressive effect was noted, with perioperative immunonutrition giving the
best outcome, postoperative standard formula the worst results, and
preoperative immunonutrition intermediate results. Thus, a dose-response
effect of key nutrients on the occurrence of complications may be
hypothesized.
The 2 groups supplemented with the enriched formula received more energy and
proteins than the control group before surgery. However, the better outcome
in the groups receiving immunonutrition should not be attributed to the
differences in preoperative energy and nitrogen intake compared with the
control group because it has been shown previously that when isoenergetic
and isonitrogenous preoperative load was used, the results were still in
favor of immunonutrition. 15
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r15> , 16
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r16>  This allows us to
speculate that the key factor for improved outcome was the provision of
immune-enhancing substrates rather than the simple provision of energy and
proteins.
In the present study, the incidence of infection was not significantly
different among groups. Yet, patients receiving the supplemented diet
required a shorter duration of antibiotic therapy to treat infectious
complications. These data may be interpreted as the result of the
therapeutic combination of drugs and a more efficient host response on the
process of bacterial clearance. Also, it may be hypothesized that the
reduction in the postoperative infectious morbidity rate in the supplemented
groups is related to improved gut oxygen metabolism and tension already
reported in patients perioperatively supplemented with immunonutrition. 35
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r35>  This may result in
advantages on intestinal barrier function and subsequent decreased bacterial
translocation that, coupled with malnutrition, may become clinically
relevant. 40 <http://archsurg.ama-assn.org/issues/v137n2/rfull/#r40>
This study was not designed to address treatment and complication costs.
Nevertheless, the health care resources saved by the marked reduction in the
morbidity rate by using perioperative immunonutrition may largely outnumber
the additional costs of the supplemented diet. 15
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r15> , 38
<http://archsurg.ama-assn.org/issues/v137n2/rfull/#r38>
In conclusion, the present trial is the first, to our knowledge, to report
that in a selected population of malnourished patients with cancer
undergoing major elective surgery, the administration of key substrates was
beneficial to outcome. Administration of the supplemented diet before and
after surgery seemed to be the best strategy to reduce complications and
length of hospital stay.



Author/Article Information


From the Department of Surgery, San Raffaele University (Drs Braga,
Gianotti, Nespoli, and Di Carlo), and the Department of Informative Systems
and Statistics, University of Milan (Dr Radaelli), Milan, Italy.

Corresponding author and reprints: Luca Gianotti, MD, ScD, Department of
Surgery (Chirurgia 3) San Gerardo Hospital, Via Donizetti 106, 20052 Monza
(MI), Italy (e-mail: [log in to unmask]
<mailto:[log in to unmask]> ).
The diets were provided by Novartis Consumer Health.




REFERENCES



1. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr1>
Mullen JL.
Consequences of malnutrition in the surgical patient.
Surg Clin North Am.
1981;61:465-487.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
6789472>
2. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr2>
Gianotti L, Braga M, Radaelli G, et al.
Lack of improvement of prognostic performance of weight loss when combined
with other nutritional parameters.
Nutrition.
1995;11:12-16.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
7749237>
3. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr3>
Mora RJF.
Malnutrition: organ and functional consequences.
Word J Surg.
1999;23:530-535.
4. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr4>
ASPEN Board of Directors.
Guidelines and standards.
JPEN J Parenter Enteral Nutr.
1995;19:1-2.
5. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr5>
Società Italiana di Nutrizione Parenterale ed Enterale.
Linee guida per l'impiego della nutrizione artificiale nei pazienti adulti
ospedalizzati.
Riv Ital Nutr Parenter Enteral.
1995;13(suppl 2):S2-S7.
6. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr6>
French-Speaking Society for Parenteral and Enteral Nutrition.
Perioperative artificial nutrition in elective adult surgery.
Clin Nutr.
1996;15:223-229.
7. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr7>
Braga M, Gianotti L, Gentilini O, Parisi V, Salis C, Di Carlo V.
Early postoperative enteral nutrition improves gut oxygenation and reduces
costs compared with total parenteral nutrition.
Crit Care Med.
2001;29:242-248.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
11246300>
8. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr8>
Bozzetti F, Braga M, Gianotti L, et al.
Postoperative enteral versus parenteral nutrition in malnourished patients
with gastrointestinal cancer: a randomized multicenter trial.
Lancet.
2001;358:1487-1492.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
11705560>
9. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr9>
Beier-Holgersen R, Boesby S.
Influence of postoperative enteral nutrition on postsurgical infections.
Gut.
1996;39:833-835.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
9038665>
10. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr10>
Sand J, Luostarinen M, Matikainen M.
Enteral or parenteral feeding after total gastrectomy: prospective
randomized pilot study.
Eur J Surg.
1997;163:761-766.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
9373227>
11. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr11>
Carr CS, Ling E, Boulos P, Singer M.
Randomised trial of safety and efficacy of immediate postoperative enteral
feeding in patients undergoing gastrointestinal resection.
BMJ.
1996;312:869-871.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
8611872>
12. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr12>
Moore FA, Feliciano DV, Andrassy RJ, et al.
Early enteral feeding, compared with parenteral, reduces postoperative
septic complications: the results of a meta-analysis.
Ann Surg.
1992;216:172-183.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
1386982>
13. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr13>
Gianotti L, Gentilini O, Braga M.
Enteral nutrition with supplemented arginine, glutamine, RNA, and omega-3
fatty acids.
Curr Opt Crit Care.
1998;4:263-268.
14. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr14>
Hall JC, Heel K, McCauley R.
Glutamine.
Br J Surg.
1996;83:305-312.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
8665180>
15. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr15>
Senkal M, Zumtobel V, Bauer K-H, et al.
Outcome and cost-effectiveness of perioperative enteral immunonutrition in
patients undergoing elective upper gastrointestinal tract surgery.
Arch Surg.
1999;134:1309-1316.
ABSTRACT <http://archsurg.ama-assn.org/issues/v134n12/abs/ssi9025.html>   |
FULL TEXT <http://archsurg.ama-assn.org/issues/v134n12/rfull/ssi9025.html>
|   PDF <http://archsurg.ama-assn.org/issues/v134n12/rpdf/ssi9025.pdf>   |
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
10593328>
16. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr16>
Braga M, Gianotti L, Radaelli G, et al.
Perioperative immunonutrition in patients undergoing cancer surgery: results
of a randomized double-blind phase 3 trial.
Arch Surg.
1999;134:428-433.
ABSTRACT <http://archsurg.ama-assn.org/issues/v134n4/abs/ssi8001.html>   |
FULL TEXT <http://archsurg.ama-assn.org/issues/v134n4/rfull/ssi8001.html>
|   PDF <http://archsurg.ama-assn.org/issues/v134n4/rpdf/ssi8001.pdf>   |
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
10199318>
17. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr17>
Gianotti L, Gentilini O, Braga M.
Nutrition in surgical oncology.
In: Mason JB, Nitenberg G, eds. Cancer & Nutrition: Prevention and
Treatment. Basel, Switzerland: S Karger AG; 2000:239-254.
18. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr18>
Klein S, Kinney J, Jeejeebhoy K, et al.
Nutritional support in clinical practice: review of the published data and
recommendation for future research directions.
JPEN J Parenter Enteral Nutr.
1997;21:133-156.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
9168367>
19. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr19>
Veteran Affairs Total Parenteral Nutrition Cooperative Study Group.
Perioperative total parenteral nutrition in surgical patients.
N Engl J Med.
1991;325:525-532.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
1906987>
20. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr20>
Eisenberg JM, Glick HA, Buzby GP, Kinosian B, Williford WO.
Does perioperative total parenteral nutrition reduce medical costs?
JPEN J Parenter Enteral Nutr.
1993;17:199-200.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
8505823>
21. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr21>
Salis C, Paccagnella A, Vannucci A, Faraguna D, Michieletto F.
Italian multicentric study of the resources employed in artificial nutrition
in hospitalized patients.
Riv Ital Nutr Parenter Enteral.
1998;16:14-24.
22. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr22>
Wicks C, Somasuydaram S, Bjarnason I, et al.
Comparison of enteral feeding and total parenteral nutrition after liver
transplantation.
Lancet.
1994;344:837-840.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
7916398>
23. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr23>
Hedberg AM, Lairson DR, Aday LA, et al.
Economic implications of an early postoperative enteral feeding protocol.
J Am Diet Assoc.
1999;99:802-807.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
10405677>
24. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr24>
Beale RJ, Bryg DJ, Bihari DJ.
Immunonutrition in the critically ill: a systematic review of clinical
outcome.
Crit Care Med.
1999;27:2799-2805.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
10628629>
25. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr25>
Heys S, Walker LG, Smith I, et al.
Enteral nutrition supplementation with key nutrients in patients with
critical illness and cancer: a meta-analysis of randomized controlled
clinical trials.
Ann Surg.
1999;229:467-477.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
10203078>
26. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr26>
Daly JM, Lieberman MD, Goldfine J, et al.
Enteral nutrition with supplemental arginine, RNA, and omega 3 fatty acids
in patients after operation: immunologic, metabolic and clinical outcome.
Surgery.
1992;112:56-67.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
1377838>
27. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr27>
Daly JM, Weintraub FN, Shou J, Rosato EF, Lucia M.
Enteral nutrition during multimodality therapy in upper gastrointestinal
cancer patients.
Ann Surg.
1995;221:327-338.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
7726669>
28. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr28>
Senkal M, Mumme A, Eickhoff U, et al.
Early postoperative immunonutrition: clinical outcome and cost-comparison
analysis in surgical patients.
Crit Care Med.
1997;25:1489-1496.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
9295822>
29. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr29>
Gianotti L, Braga M, Vignali A, et al.
Effect of route of delivery and formulation of postoperative nutritional
support in patients undergoing major operations for malignant neoplasm.
Arch Surg.
1997;132:1222-1230.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
9366716>
30. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr30>
Heslin MJ, Latkany L, Leung D, et al.
A prospective randomized trial of early enteral feeding after resection of
upper gastrointestinal malignancy.
Ann Surg.
1997;226:567-580.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
9351723>
31. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr31>
Braga M, Vignali A, Gianotti L, Cestari A, Profili M, Di Carlo V.
Immune and nutritional effects of early enteral nutrition after major
abdominal operations.
Eur J Surg.
1996;162:105-112.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
8639722>
32. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr32>
Kemen M, Senkal M, Homann HH, et al.
Early postoperative enteral nutrition with arginine, omega-3 fatty acids and
ribonucleic acid–supplemented diet versus placebo in cancer patients: an
immunologic evaluation of Impact.
Crit Care Med.
1995;23:652-659.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
7536138>
33. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr33>
Senkal M, Kemen M, Homann HH, et al.
Modulation of postoperative immune response by enteral nutrition with a diet
enriched with arginine, RNA, and omega-3 fatty acids in patients with upper
gastrointestinal cancer.
Eur J Surg.
1995;161:115-122.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
7539633>
34. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr34>
Cerra FB, Lehmann S, Kostantinites N, et al.
Improvement in immune function in ICU patients by enteral nutrition
supplemented with arginine, RNA, and menhaden oil is independent of nitrogen
balance.
Nutrition.
1991;7:193-199.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
1724941>
35. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr35>
Braga M, Gianotti L, Cestari A, et al.
Gut function and immune and inflammatory responses in patients
perioperatively fed with supplemented formulas.
Arch Surg.
1996;131:1257-1265.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
8956766>
36. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr36>
Gianotti L, Braga M, Fortis C, et al.
A prospective randomized clinical trial on perioperative feeding with an
arginine, omega-3 fatty acids, and RNA-enriched enteral diet: effect on host
response and nutritional status.
JPEN J Parenter Enteral Nutr.
1999;23:314-320.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
10574478>
37. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr37>
Wachtler P, Axel R, König W, Bauer H, Kemen M, Köller M.
Influence of a pre-operative enteral supplementation on functional
activities of peripheral leukocytes from patients with major surgery.
Clin Nutr.
1995;14:275-282.
38. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr38>
Gianotti L, Braga M, Frei A, Greiner R, Di Carlo V.
Health care resources consumed to treat postoperative infections: cost
saving by perioperative immunonutrition.
Shock.
2000;14:325-330.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
11028551>
39. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr39>
Gianotti L, Braga M, Radaelli G, Nespoli L, Di Carlo V.
A prospective randomized trial on pre-operative immunonutrition in
well-nourished patients undergoing major surgery for gastrointestinal cancer
[abstract].
Br J Surg.
2001;88:1142.
40. <http://archsurg.ama-assn.org/issues/v137n2/rfull/#rr40>
Deitch EA, Winterton J, Berg RD.
The gut as a portal of entry for bacteremia: role of protein malnutrition.
Ann Surg.
1987;205:681-692.
MEDLINE
<http://www.ncbi.nlm.nih.gov/htbin-post/Entrez/query?db=m&form=6&Dopt=r&uid=
3592811>


Edward E. Rylander, M.D.
Diplomat American Board of Family Practice.
Diplomat American Board of Palliative Medicine.