Skip to main content
Erschienen in: World Journal of Surgery 4/2014

Open Access 01.04.2014

Perioperative Immunonutrition in Surgical Cancer Patients: A Summary of a Decade of Research

verfasst von: Stanislaw Klek, Piotr Szybinski, Kinga Szczepanek

Erschienen in: World Journal of Surgery | Ausgabe 4/2014

Abstract

Background

Immunonutrition is assumed to enhance immune system function. In surgical patients, it is supposed to reduce postoperative complications. However, results of recent clinical trials have been puzzling and have not supported this theory.

Aim

The aim of our study was to evaluate the value of enteral and parenteral postoperative immunonutrition.

Methods

After initial evaluation of 969 patients, the intent-to-treat analysis included 776 patients (female 407, male 466, mean age 61.1 years) undergoing gastric or pancreatic resections between 2001 and 2009. All patients were randomly assigned after surgery to one of the following groups: standard enteral nutrition (SEN), immunomodulating enteral nutrition (IMEN), standard parenteral nutrition (SPN), or immunomodulating parenteral nutrition (IMPN). All malnourished patients received preoperative parenteral nutrition. Number and type of postoperative complications, length of hospitalization (length of stay [LOS]), and vital organ function were assessed.

Results

No statistically significant differences were observed in well-nourished patients, during either enteral or parenteral intervention, independent of the type of intervention (standard or immunomodulating). However, analysis of the malnourished group revealed the positive impact of enteral immunonutrition on reduction of postoperative complications (28.3 vs. 39.2 %, respectively; p = 0.043) and LOS (17.1 and 13.1 days, respectively; p < 0.05) compared with a standard enteral diet. The cross-analysis of SEN, IMEN, SPN, and IMPN was insignificant.

Conclusions

The type of postoperative nutrition was of no importance in well-nourished patients. However, in malnourished patients, enteral immunonutrition helped to improve treatment outcome. These findings suggest its use as a method of choice during the postoperative period.
Hinweise
Clinical Trials Database – number: NCT00576940.

Introduction

Only a few factors may influence results of surgery to such an extent as malnutrition. It complicates wound healing, increases the rate of postoperative infections, and lengthens hospital stay. These outcomes are consequences of the destruction of immune function, amplifying the response to stress and organ dysfunction [1].
Nutritional therapy has been used in the postoperative period for over 100 years, since Kausch administered intravenous glucose solution to help his patient’s recovery [2]. However, the history of nutritional support as we know it nowadays began with the invention of parenteral nutrition by Dudrick et al. [3]. Intravenous admixtures proved effective not only in maintaining health status, but also in safeguarding growth. This medical approach irreversibly changed the perception of perioperative care. However, Buzby et al. [4] and the Veteran Affairs Trial [5] highlighted the consequences of hyperalimentation in well-nourished patients, indicating a reduction in complications of up to 20 %, but only in malnourished patients. These studies clarified the perspective of perioperative care and switched a proportion of nutritional intervention to the more physiological, less expensive, and safer enteral feeding [4, 5].
It took another decade to set the criteria for the selection of the proper feeding route. Nowadays, leading scientific societies, ASPEN (American Society for Parenteral and Enteral Nutrition) and ESPEN (European Society for Clinical Nutrition and Metabolism), agree that enteral nutrition should be used as the method of choice in perioperative treatment [6, 7]. Further studies concentrated on pharmaconutrition: formulas that can be used not only to deliver basic nutrients, but also to influence vital organs and systems to improve the outcome of therapies. One form of pharmaconutrition, aiming at improvement of immune function, was called immunomodulating or immunostimulating. These diets, both parenteral and enteral, included amino acids (arginine and glutamine), lipids (omega-3 fatty acids), micronutrients (vitamins C and E), and nucleotides. Soon after their implementation, some authors observed their encouraging influence on the outcome of surgery, which raised new hope for surgical patients [810]. Controversy soon began, and the actual value of immunomodulating formulas for surgical and critically ill patients was examined. The positive effects of immunodiets observed in experimental models were often denied by clinical results, far more important for clinicists [11]. In contradiction to Braga et al. [8]. or Gianotti et al. [9], Senkal et al. [10]. and Lobo et al. [11]. revealed that enteral immunodiets bore no advantage over standard enteral nutrition (SEN) when a peptide diet was used; other authors noticed analogous results. Furthermore, most research performed in well-nourished individuals failed to demonstrate the quantifiable efficacy of immunomodulating diets [1113].
These outcomes were difficult to match because of the heterogeneity of study populations, study designs, sample quantities, and systematic approaches.
To completely address these uncertainties and to verify the value of immunonutrition in surgical patients, randomized well-designed trials were conducted in a tertiary surgical center between 2001 and 2009. Results of these trials were partially published in medical journals, but have never been presented as a summary [1215].

Methods

Study design

We conducted a randomized, not blinded (due to obvious differences in enteral and parenteral routes), controlled study in order to evaluate the impact of enteral and parenteral immunonutrition on postoperative complications in surgical patients. The study was conducted in the tertiary surgical center—the 1st Department of General and Oncology Surgery, Jagiellonian University School of Medicine, Cracow, Poland—and was performed between January 2001 and December 2009.
The research was planned to test the hypothesis that immunonutrition would decrease the occurrence of surgical and non-surgical complications after gastrointestinal (GI) surgery. The secondary objectives included evaluation of effects of nutritional intervention on morbidity and mortality, length of hospital stay (LOS), and vital organ function.

Patient characteristics and inclusion/exclusion criteria

A total of 969 patients were initially assessed for participation in the study; 96 patients were unable to meet inclusion criteria and were excluded after initial assessment. The intent-to-treat (ITT) analysis of 776 patients (female 407, male 466, mean age 61.1 years) who underwent gastrectomy (subtotal or total resection) or pancreatoduodenectomy (subtotal or total) with lymph node excision were enrolled in the trial. The inclusion criteria included good overall status (Karnofsky >80, Eastern Cooperative Oncology Group [ECOG] grade 0 or 1); the absence of cancer dissemination or severe associated diseases (cardiac, pulmonary, renal, liver failure, chronic obstructive pulmonary disease [COPD], coronary aortic bypass graft, etc.); no history of known aversions or intolerance to analyzed substances. Patients with metastatic or unresectable cancer, who were pregnant or in poor general health (Karnofsky <80, ECOG > 1), with recent history of severe heart, lung, kidney, or liver failure, with history of allergies or drug intolerance were excluded. Malnutrition was defined as either of the following: unintentional weight loss of at least 10–15 % within 3–6 months before admission or body mass index (BMI) <18 kg/m2. Respective groups of patients were comparable with each other in terms of sex, age, type of surgery, BMI, weight loss, serum albumin, and total lymphocyte count (TLC) on admission and blood transfusion.

Randomization and allocation of patients

After tumor resection, individuals who met the eligibility norms were intraoperatively allocated to either of four groups using sealed envelopes containing computer-generated distribution numbers. A 2 × 2 factorial scheme was used with the subsequent groups: SEN, immunomodulating enteral nutrition (IMEN), standard parenteral nutrition (SPN), and immunomodulating parenteral nutrition (IMPN) in two parts of the research. In the third part, in which enteral intervention was assessed, patients were randomized into SEN or IMEN groups. The CONSORT (Consolidated Standards of Reporting Trials) diagram (Fig. 1) shows the flow of participants through the study.

Clinical management

In contrast with well-nourished patients, who underwent surgery without preoperative nutritional support, all malnourished patients received intravenous nutrition the fortnight before surgery. Protein and energy demands were calculated using the nitrogen to body weight (b.w.) ratio (0.15 g N/kg b.w.) and the non-protein energy proportion (Q = 150 kcal/g N). The 10 % amino acid solutions, 10–40 % glucose, and 10–20 % lipid emulsions, trace elements (Aminoplasmal, Glucose and Lipofundin MCT/LCT B and Tracutil, B Braun, Germany), vitamins (Cernevit, Baxter, USA), and electrolyte solutions were used to prepare all-in-one bags at the pharmacy. The central intravenous catheter was implanted in the subclavian or jugular vein before the onset of therapy. The tip location was confirmed by chest X-ray. The same type of intravenous admixture as given preoperatively was provided to each patient during the postoperative period up to postoperative day 7, or longer in case of complications.
The selection of parenteral instead of enteral feeding during the preoperative period, which has been endorsed for many years, was the consequence of the absence of those guidelines at the time of study preparation (2001) and the wide acceptance of that kind of perioperative approach in local surgical units (Polish national standards).
During surgery (gastric or pancreatic resection), an enteral feeding tube (Flocare Nutricia Ltd., 140 cm length) was inserted into the first intestinal loop, 15–20 cm below the nethermost anastomosis. The surgical team included at least two skilled general and oncological surgeons, and the anesthesiology team comprised four people.
Preceding surgery, BMI, weight loss, full blood count with TLC, albumin and pre-albumin concentration, liver and kidney tests were assessed.
On postoperative day 1, 3, and 8, the following assessments were made: full blood count with TLC, serum albumin and pre-albumin concentrations, liver and renal function, quantity of diet administered, and tolerance. Assessments were performed by physicians and nurses.
The energy and protein requirements during the postoperative period were calculated using the same method as during the preoperative intervention. Enteral feeding was started 6 h after the procedure, with 5 % glucose solution at the rate of 20 ml per h for the first 12 h, followed by infusion of Peptisorb (SEN group; Nutricia Ltd., Poland) or Reconvan® (IMEN group; Fresenius Kabi, Poland) at the rate of 20 ml/h on day 1, 50 ml/h on day 2, 75 ml/h on day 3, and 100 ml/h thereafter. The whole intervention lasted 1 week. Diet ingredients are presented in Table 1. Infusion devices were used to administerr the diet for 20–22 h, with a 2–4 h rest period. An oligopeptide, isocaloric diet was selected as a control because of previous high tolerance in the small intestine. Infusion pumps were used to guarantee volume and speed control.
Table 1
Composition of enteral diets (per 100 ml/100 kcal)
Ingredient
Peptisorb (standard diet)
Stresson (immunodiet)
Reconvan® (immunodiet)
Energy (kcal)
100
125
100
Amino acids (g)
4.0
7.5
5.5
Carbohydrates (g)
17.6
14.5
18.0
 Polysaccharides (g)
14.6
13.3
13.3
 Sugars (g)
1.7
0.7
0.7
 Lactose (g)
0.1
<0.025
0.15
Total fat (g)
1.7
4.1
3.3
 Saturated (g)
1.0
2.6
3.3
 MCT (g)
0.8
1.5
1.2
Fibre (g)
0
0.1
0
Electrolytes
   
 Sodium mg (mmol)
100 (4.3)
134
138
 Potassium mg (mmol)
150 (3.8)
263
207
 Chloride mg (mmol)
125 (3.5)
139
141
 Calcium mg (mmol)
80 (2.0)
67
80 (2.0)
 Phosphorus (mg)
72 (2.3)
67
60
 Magnesium (mg)
23 (0.9)
28
25
 Iron (mg)
1.6
1.0
1.33
 Zinc (mg)
1.2
1.0
1.2
 Copper (mcg)
180
338
133
 Manganese (mg)
0.33
0.63
0.63
 Fluoride (mg)
0.10
0.13
0.27
 Molybdenum (mcg)
10.0
13
10.0
 Selenium (mcg)
5.7
14
6.7
 Chromium (mcg)
6.7
8.9
6.7
 Iodine (mcg)
13
17
13.3
Vitamins
   
 Vitamin A (mcg RE)
82
91
70
 Vitamin D (mcg)
0.70
0.88
0.88
 Vitamin E (mg α-TE)
1.3
1.3
1.0
 Vitamin K (mcg)
5.3
6.6
6.7
 Thiamine (mg)
0.15
0.19
0.2
 Riboflavin (mg)
0.16
0.31
0.16
 Niacin (mg NE)
1.8
2.3
1.6
 Pantothenic acid (mg)
0.53
0.66
0.47
 Vitamin B6 (mg)
0.17
0.39
0.16
 Folate (mcg)
27
50
27
 Vitamin B12 (mcg)
0.21
0.4
0.27
 Biotin (mcg)
4.0
5.0
5.0
 Vitamin C (mg)
10.0
25
6.7
 Choline (mg)
37
46
26.7
 Taurine (mg)
10
13
13
 Glutamine (g/l)
10.1
10.2
 Arginine (g/l)
7.2
6.7

Primary objective (primary endpoint)

The primary objective of this study was to assess the influence of immunomodulating nutrition on postoperative complications in surgical patients. The ratio of postoperative complications was nominated as the primary outcome measure, with the hypothesis that the routine use of immunodiets in the postoperative period reduces the number of infectious and surgical complications. Definitions of complications are presented in Table 2.
Table 2
Definitions of complications
Complication
Definition
Wound infection
Purulent exudate in the wound with positive bacterial culture
Abdominal abscess
Collection of pus confirmed by percutaneous drainage or at reoperation
Pneumonia
Clinical signs of pneumonia and/or radiographic evidence (both required to diagnose) or positive culture of tracheal aspirate or blood or brushing
Urinary tract infection
Clinical symptoms and the presence of bacteria in urine (>100,000 colony-forming units/ml)
Bacteremia
Positive blood culture
Infection of venous catheter
Local signs of inflammation, and/or the isolation of pathogen organisms in culture
Sepsis
Fever >38 °C or hypotension (<90 mm Hg systolic BP) or oliguria (<20 ml/h) along with positive blood culture
Wound dehiscence
Any dehiscence of the fascia >3 cm
Bleeding
Necessity of blood transfusion (≥2 U)
Anastomotic leak
Positive dye-swallow or contrast-swallow test
Respiratory tract failure
Presence of dyspnoea and respiratory rate >35 breaths/min or PaO2 <70 mm Hg
Circulatory insufficiency
Unstable BP requiring use of extra fluids or cardiac stimulants
Renal failure
Necessity of haemodialysis
Hepatic dysfunction
Increased serum bilirubin (2–3 times above baseline) or hepatic enzyme level (3–4 times above baseline)
Pancreatic fistula
Drain output of any measurable volume of fluid on or after postoperative day 3 with an amylase content greater than 3 times the serum amylase activity
Delayed gastric emptying
Necessity for nasogastric suction for ≥8 days after surgery
BP blood pressure

Secondary objectives (secondary endpoints)

The secondary objectives included LOS, immune system function (clinical observations and TLC), assessment of liver and renal function, and treatment compliance. Furthermore, operational time, intra-operative blood loss, blood transfusions, and the necessity for re-operation were recorded. Post-operative mortality was defined as any fatal outcome within 31 days after hospitalization. The length of postoperative stay was number of days from the date of operation until the date of discharge. Albumin solutions were not used as standard treatment.

Sample size and statistical analysis

The sample size was calculated using SamplePower™, version 16–19 (SPSS Inc., Chicago, IL, USA). A general estimation was made for each study: the total rate of complications after upper GI surgery described by previous studies was approximately 40 %. To detect a 50 % reduction triggered by immune enteral nutrition, more than 82 patients should be randomized to each of the two related arms (alpha = 0.05 two-sided, power = 0.80). We assumed a drop-out rate of 15 %; therefore, 200 patients were needed. The figures were investigated on an ITT basis using SPSS v.14 software. The differences in proportions amid groups were assessed using the Chi squared test, and Yates correction was implemented if any of the probable incidences were <5. Continuous data were studied using the Mann–Whitney U test. Differences at p < 0.05 were considered statistically significant.
The Ethics Committee of Jagiellonian University sanctioned the study (KBET/91/L/2004). It was not possible to obtain approval in 2001, as the Ethics Committee did not participate in such activities beforehand. Patients were registered by one of two investigators (SK, KS). Written approval was acquired from each participant before acceptance. The study was carried out following the universal ethical endorsements stated in the Helsinki Declaration and was recorded in the Clinical Trials Database (NCT00576940).

Role of sources of funding

The study was performed with no outside sponsorship.

Results

Participants were adequately matched for age, sex, weight loss, BMI, type of operation (stomach/pancreas resection), TLC, and serum albumin (indicators of nutritional state). The number of patients who were operated on for gastric and pancreatic neoplasm, as well as patients within both groups, was comparable. The ratio of resection types (sub/total excision) did not differ between groups.
During preoperative intravenous feeding in malnourished patients, each study group received a comparable level of energy, proteins, and micronutrients.

Postoperative follow-up

There were no noteworthy dissimilarities between the enteral groups in the tube feeding delivery, either in the malnourished or the well-nourished group. For the purpose of this research, the observation was completed on postoperative day 8, after a full 7 days of enteral feeding, but the mean length of intervention in the SEN and IMEN group was 8.4 (±1.2) days and 8.6 (±1.4) days. The mean interval of intravenous nutrition was 7.9 (0.8) days for SPN and 8.1 (1.0) days for IMPN.
Compliance was similar amid malnourished patient groups, protocol violation due to the full dose of diet not being delivered was the reason for premature cessation in eight patients (SEN-1, IMEN-2), which accounted for <1 % of all patients. The planned delivery reached 80 % of those originally prescribed. The average quantity of blood units was 1.7 in SEN and 1.6 in IMEN, which was not significant (p = 0.42).
Detailed analyses of postoperative complications are presented in Tables 3, 4, 5 and 6.
Table 3
Postoperative complications among study groups (part 1: 2 × 2 randomization in malnourished patients)
Variable
SEN and SPN (N = 84)
IMEN and IMPN (N = 83)
OR (95 % CI)
p value
PN (N = 83)
EN (N = 84)
OR (95 % CI)
p value
Rate of infectious complications
23 (27)
20 (24)
0.842 (0.420–1.687)
0.627
19 (23)
24 (29)
1.347 (0.671–2.706)
0.401
Overall morbidity
33 (39)
25 (30)
0.666 (0.351–1.265)
0.214
29 (35)
29 (35)
0.982 (0.519–1.857)
0.995
Morbidity (30 days post-surgery)
37 (41)
28 (33)
0.655 (0.353–1.345)
0.216
32 (38)
32 (38)
0.983 (0.499–1.878)
0.991
Mortality
3 (4)
0 (0)
0.882 (0.473–1.643)
0.692
1 (1)
2 (2)
1.078 (0.579–2.008)
0.812
Mortality (30 days post-surgery)
4 (5)
1 (1)
0.789 (0.541–1.742)
0.691
2 (2)
3 (3)
1.112 (0.611–1.997)
0.828
Hospital stay, days [median (IQR)]
9 (9–14)
9 (9–12)
0.835
9 (9–13)
9 (9–12)
0.415
Data are presented as N (%) unless otherwise indicated
EN enteral nutrition, IMEN immunomodulating enteral nutrition, IMPN immunomodulating parenteral nutrition, IQR interquartile range, OR odds ratio, PN parenteral nutrition, SEN standard enteral nutrition, SPN standard parenteral nutrition
Table 4
Postoperative complications among study groups (part 2: enteral nutrition in malnourished patients)
Type of complication
SEN (N = 153)
IMEN (N = 152)
p value*
Infectious complications
60 (39.22)
43 (28.29)
0.04366
 Pneumonia
45 (29.41)
33 (21.71)
0.12322
 Urinary tract infection
15 (9.80)
11 (7.24)
0.42213
 Surgical wound infection
27 (17.65)
12 (7.89)
0.01077
 Intra-abdominal abscess
10 (6.54)
5 (3.29)
0.18988
 Bacteremia
11 (7.19)
2 (1.32)
0.01112
 Sepsis
2 (1.31)
4 (2.63)
0.40498
Other complications
   
 Wound dehiscence
8 (5.23)
2 (1.32)
0.05502
 Pancreatic fistula
10 (6.54)
4 (2.63)
0.10329
 Intestinal fistula
8 (5.23)
4 (2.63)
0.24340
 Duodenal fistula
1 (0.65)
2 (1.32)
0.55793
 Biliary fistula
2 (1.31)
3 (1.97)
0.64672
 Abdominal fluid collection
2 (1.31)
3 (1.97)
0.64672
 Delayed gastric emptying
13 (8.50)
8 (5.26)
0.26479
 Acute pancreatitis
1 (0.65)
2 (1.32)
0.55793
 Intestinal obstruction
2 (1.31)
2 (1.32)
0.99473
 Peritonitis
1 (0.65)
1 (0.66)
0.99629
 Pulmonary embolism
2 (1.31)
1 (0.66)
0.56563
 Heart failure
3 (1.96)
2 (1.32)
0.65738
 Respiratory failure
7 (4.58)
5 (3.29)
0.56362
 Liver failure
1 (0.65)
0 (0.00)
0.31810
 Renal failure
0 (0.00)
3 (1.97)
0.08075
 Neurological
1 (0.65)
1 (0.66)
0.99629
 Peripheral veins thrombosis
2 (1.31)
1 (0.66)
0.56563
 GI bleeding
1 (0.65)
1 (0.66)
0.99629
 Abdominal bleeding
2 (1.31)
2 (1.32)
0.99473
Mortality
9 (5.88)
2 (1.32)
0.03247
Overall morbidity
72 (47.06)
51 (33.55)
0.01621
All data are presented as N (%) unless otherwise indicated
GI gastrointestinal, IMEN immunomodulating enteral nutrition, SEN standard enteral nutrition
Table 5
Postoperative complications among study groups (part 3: 2 × 2 randomization in well-nourished patients)
Variable
Standard nutrition (N = 102)
Immunonutrition (N = 103)
OR (95 % CI)
p value
PN (N = 100)
EN (N = 105)
OR (95 % CI)
p value
Rate of infectious complications
28 (27)
25 (24)
0.81 (0.43–1.50)
0.498
25 (25)
28 (27)
1.14 (0.61–2.14)
0.672
Overall morbidity
36 (35)
37 (36)
1.08 (0.60–1.93)
0.804
35 (35)
38 (36)
0.85 (0.48–1.50)
0.577
Mortality
2 (2)
2 (2)
0.99 (0.14–7.17)
0.992
2 (2)
2 (2)
0.95 (0.13–6.89)
0.960
Hospital stay (days, mean)
12.8
12.5
–0.32 (–1.62 to 2.26)
0.746
12.9
12.4
–0.43 (–2.31 to 1.46)
0.656
Data are presented as N (%) unless otherwise indicated
EN enteral nutrition, OR odds ratio, PN parenteral nutrition
Table 6
Postoperative complications among study groups (part 2: enteral nutrition in well-nourished patients)
Type of complication
SEN (N = 91)
IMEN (N = 92)
p value
Infectious complications
   
 Pneumonia
15 (16.4)
13 (14.1)
>0.05
 Urinary tract infection
1 (1.1)
2 (2.1)
>0.05
 Surgical wound infection
5 (5.5)
4 (4.2)
>0.05
 Abscess formation
2 (2.2)
2 (2.1)
>0.05
Surgical complications
   
 Evisceration
0
1 (1.1)
>0.05
 Pancreatic fistula
1 (1.1)
0
>0.05
 Duodenal fistula
1 (1.1)
1 (1.1)
>0.05
 Jejunal fistula
2 (2.2)
1 (1.1)
>0.05
 Biliary fistula
0
1 (1.1)
>0.05
 Surgical complications overall
4 (4.4)
4 (4.4)
>0.05
General complications
   
 Pulmonary thrombosis
0
0
>0.05
 Myocardial infarct
0
1 (1.1)
>0.05
 Peripheral vein thrombosis
0
0
>0.05
 Neurological complications
0
0
>0.05
 Fatal outcome
1 (1.1)
1 (1.1)
>0.05
Complications overall (patients)
21 (23.1)
23 (25.2)
>0.05
Uncomplicated postoperative period (patients)
70 (76.9)
69 (75)
>0.05
Data are presented as N (%) unless otherwise indicated
IMEN immunomodulating enteral nutrition, SEN standard enteral nutrition
The LOS differed between the two study groups and extended up to 17.1 days (standard deviation [SD] 12.2) in the SEN group and 13.1 days (SD 13.8) in the IMEN group (p = 0.006) in malnourished patients. In the same group, there were significant dissimilarities in infectious complications, which occurred in 60 patients (39.2 %) in the SEN group and 43 (28.3 %) in the IMEN group (p = 0.04). Differences were also detected in morbidity (47.1 vs 33.5 %; p = 0.01) and mortality (5.9 vs 1.3 %; p = 0.03).
In well-nourished patients, the median LOS was 12.4 (SD 5.9) days in the SEN group and 12.9 (SD 8.0) days in the IMEN group (p = 0.42). Complications were detected in 21 patients (23.1 %) in the SEN group and 23 (25.2 %) in the IMEN group (p > 0.05). Four (4.4 %) patients in the SEN group and four (4.4 %) in the IMEN group had surgical complications (p > 0.05).
Blood transfusions were necessary in 12 well-nourished patients in the SEN group and 11 in the IMEN group; the median numbers of transfused units were 2.5 in SEN and 2 in IMEN (interquartile range [IQR] 1–3.5 and 1–5, respectively).
In these patients, LOS was similar in both groups: 12.4 (SD 5.9) days in the SEN and 12.9 (SD 8.0) days in the IMEN (p = 0.42) groups. Complications were noted in 21 (23.1 %) patients in the SEN and 23 (25.2 %) in the IMEN groups. Infective complications were detected in 23 patients in the SEN group and 21 in the IMEN group.
Well-nourished patients administered parenteral and enteral nutrition had a morbidity rate of 36 %; the occurrence of specific complications was similar among all groups (Table 4). Infectious complications were detected in 28 of 102 patients on standard diets and in 25 of 103 patients receiving immuno-formula (odds ratio [OR] 0.81; 95 % CI 0.43–1.50) (Table 5). Furthermore, there were no dissimilarities amid infectious complications between those receiving enteral nutrition (25 of 100 patients) and those receiving parenteral formulas (28 of 105, OR 1.14, 95 % CI 0.61–2.14). Neither the immunodiet nor enteral nutrition affected secondary outcome measures, including morbidity, mortality, and LOS.
Serum pre-albumin, albumin, and TLC levels were secondary endpoints. The first two were used to assess visceral protein synthesis and the restoration of nutritional status. However, the study did not indicate any differences among groups, as demonstrated in Table 6. Significant differences were found only in TLC on postoperative day 3, when the mean number of lymphocytes was higher in the IMEN than the SEN group (p = 0.011), which was not reflected in postoperative clinical course.
No differences were recorded in hepatic and renal function. These were assessed by clinical status and laboratory tests. The mean concentrations of aspartate aminotransferase (SGOT), alanine aminotransferase (SGTP), blood urea nitrogen (BUN), and creatinine concentrations did not differ.

Discussion

Modern surgery tries to reduce the rate of postoperative complications by concentrating more on technical aspects and less on metabolic aspects. The latter probably represents the only hope for improvement in the discipline, as there are not many opportunities for further improvement from the technical point of view. It is a holistic approach that gives hope for improvement. Nowadays, the multimodal approach to perioperative care should include analgesia, physiotherapy, aseptics, antiseptics, anticoagulants, infusion therapy, nutritional support, and many other therapeutic options. Nutritional intervention matters the most, as the worsening of nutritional status has been acknowledged as a crucial factor influencing surgical outcomes [1].
The place of pre- and postoperative nutrition is no longer in question; particularly since it has been confirmed that, in severely malnourished individuals scheduled for major GI surgery, it was advantageous to postpone surgery for up to 10–14 days and to administer nutritional support, preferably with enteral diets [6]. From the surgical point of view, it was not enough to stop there; for over 10 years the focus has been on understanding immunologic and inflammatory responses, so as to enhance host defense mechanisms and improve clinical course. These activities led to the idea of immunonutrition, a type of pharmaconutrition that has been described as nutritional intervention, not only able to provide essential nutrients to maintain basic organ functions, but also to augment the immune system [16]. The use of various biochemical agents, such as non-essential (glutamine, arginine) or sulfur-containing amino acids, omega-3-polyunsaturated fatty acids, nucleotides, and anti-oxidants (free radical scavengers), administered simultaneously, in few, some or alone, was assumed to alter the host immune response [16, 17].
Several clinical trials and meta-analyses have described the beneficial effect of perioperative administration of an enteral or parenteral formula containing immune-ingredients on the outcome of surgery, independent of nutritional state [1820]. Benefits included reduction of postoperative complications and shortening of LOS in both surgical and critically ill patients [2025]. These results were independent of age, particularly when patients received admixtures pre-operatively [2628]. Beneficial effects were also detected at the sub-clinical level: immunonutrition led to an increase in immune function due to an increase in TLC, CD4 levels, immunoglobulin (Ig)-G levels, and decrease in interleukin (IL)-6 concentrations, and the inversion of the correlation between IL-6 and prealbumin concentrations after surgery [8, 9, 21]. Senkal et al. [10]. observed beneficial effects of immunotherapy and even better cost effectiveness during the late phase of recovery (defined as the time period after postoperative day 5); at that time, the effect of immunodiets was incontrovertible.
However, criticism came with some studies on immunonutrition that were not able to demonstrate reduction of either overall mortality or morbidity, failed to prove benefits of immunonutrition, and indicated no reduction in complications or LOS [12, 2831]. Some authors observed only a reduction in infectious complications without any cost-effectiveness benefits, particularly in well-nourished patients [30, 31]. Likewise, only some trials confirmed that such formulas might lower the ratio of infectious complications, and a few even suggested that immunonutrition could increase the risk of death in the critically ill [32].
There are several explanations for these inconsistencies. Most important is the question of study group: numerous studies in which immune-intervention presented no clinical effect were undertaken in well-nourished patients, while trials indicating a decrease in complications involved moderately or severely malnourished individuals [3335]. It was obvious that any type of surgical intervention in malnourished patients would be beneficial, therefore results from malnourished patients in mixed study populations would overbalance the lack of positive results in well-nourished patients, as proven by the fresh meta-analysis of 13 randomized, controlled trials including 1,269 patients that demonstrated that perioperative immunonutrition in GI surgical patients reduced rates of postoperative infection (OR 0.41, 95 % CI 0.30–0.54), shortened LOS, and increased several markers of immune function [4]. However, nearly all of these trials comprised patients with and without malnutrition, and the percentage of malnourished patients in some of them reached nearly 60 [68].
Furthermore, the heterogeneity of definitions used in clinical studies to define simple concepts was confusing. Kudsk et al. [33] and Lobo et al. [11]. specified these inconsistencies: various definitions of malnutrition and co-morbidities, imprecise timing, and route of administration; various durations of therapy; uncontrolled execution of the nutritional intervention; and occurrence of nutrition support-related complications [7, 36].
Another point is that many patients undergoing upper GI surgery are at fairly low risk of fatal outcome after elective procedures, in contrast to critically ill patients. Therefore, ingredients such as arginine, which may be helpful in surgical patients, can be unsafe in the latter, because the high arginine content drives nitric oxide assembly [33].
Hence, the configuration of enteral diet plays a vital role. Studies in which Impact® (Novartis) was the tested substance showed benefits, even in well-nourished patients, as validated by Daly et al. [34] and Waitzberg et al. [27, 35]. It is important to bear in mind that Impact® has a specific composition: the amount of arginine in Impact® is twice as low as that in, for instance, Reconvan® (Fresenius Kabi). Furthermore, it contains no glutamine, while the concentration of this amino acid is quite significant in Reconvan® and other enteral immunodiets. Finally, in contrast with other diets, it also contains nucleotides.
The timing of the intervention represents another perilous issue. During the postoperative period, the patient goes through contrasting stages: systemic inflammatory response (SIRS) and compensatory anti-inflammatory response (CARS), which hamper maintenance of homeostasis [9]. Thus, the same substance delivered pre-operatively may have a useful effect since inflammatory processes during these periods are dissimilar. It is also easier to accomplish a nutritional plan before than after surgery [7].
The current study was designed to confirm the hypothesis that the treatment of choice, preoperative enteral nutrition enhanced with immune ingredients, can reduce the rate of infectious complications in surgical cancer patients, who represent one of the most challenging groups of patients. Study groups were perfectly homogenous in terms of baseline characteristics, type and timing of intervention, and nutritional status. We observed that, in well-nourished patients, it was the nutritional intervention itself, not its characteristics, that mattered the most. The median postoperative LOS was 12.4 (SD 5.9) days in the SEN group and 12.9 (SD 8.0) days in the IMEN group (p = 0.42). Infectious complications were observed in 21 patients (23.1 %) in the SEN group and 23 (25.2 %) in the IMEN group (p > 0.05). The rest of the study participants did not differ.
Conversely, in malnourished patients undergoing GI surgery, it was possible to demonstrate the positive effect of IMEN on treatment outcome. The most significant clinical parameters varied markedly in favor of immunonutrition. LOS was shorter: 17.1 days in the SEN group versus 13.1 in the IMEN group; the overall morbidity (47.1 vs. 33.5 %), mortality (5.9 vs. 1.3 %), and infectious complications (39.2 in SEN vs. 28.3 % in IMEN) were reduced.
Neither diet influenced hepatic and renal function, visceral protein production, or immune system recovery. The difference observed in TLC on day 3 was too slight to have been considered clinically important.
These results reinforced the value of immunonutrition confirmed previously in the preoperative period by Braga et al. [8. and Gianotti et al. [9, 20], and in the postoperative period by Zheng et al. [21] and Heyland et al. [22]. They also support the concept that the administration of arginine- and nucleotide-rich, glutamine-free enteral diets could be advantageous in malnourished and even in some well-nourished patients. However, well-nourished patients are unlikely to gain from this management during the postoperative period.

Acknowledgments

Stanislaw Klek was the coordinator of the research, responsible for the concept, and contributed to the design, data analysis, interpretation, and writing of the manuscript. Piotr Szybinski and Kinga Szczepanek contributed to the writing of the manuscript, data analysis, statistical analysis, and data interpretation.

Conflict of interest

None declared.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.

Unsere Produktempfehlungen

Die Chirurgie

Print-Titel

Das Abo mit mehr Tiefe

Mit der Zeitschrift Die Chirurgie erhalten Sie zusätzlich Online-Zugriff auf weitere 43 chirurgische Fachzeitschriften, CME-Fortbildungen, Webinare, Vorbereitungskursen zur Facharztprüfung und die digitale Enzyklopädie e.Medpedia.

Bis 30. April 2024 bestellen und im ersten Jahr nur 199 € zahlen!

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Literatur
1.
Zurück zum Zitat Studley HO (1936) Percentage of weight loss: a basic indicator of surgical risk in patients with chronic peptic ulcer. JAMA 106:458–460CrossRef Studley HO (1936) Percentage of weight loss: a basic indicator of surgical risk in patients with chronic peptic ulcer. JAMA 106:458–460CrossRef
2.
Zurück zum Zitat Kausch W (1911) Uber intravenose und subcutane Ernharung mit Traubenzuker. Deutsch Medizin Wochen 37:8CrossRef Kausch W (1911) Uber intravenose und subcutane Ernharung mit Traubenzuker. Deutsch Medizin Wochen 37:8CrossRef
3.
Zurück zum Zitat Dudrick SJ, Wilmore DW, Vars HM et al (1968) Long-term total parenteral nutrition with growth, development, and positive nitrogen balance. Surgery 64:134–142PubMed Dudrick SJ, Wilmore DW, Vars HM et al (1968) Long-term total parenteral nutrition with growth, development, and positive nitrogen balance. Surgery 64:134–142PubMed
4.
Zurück zum Zitat Buzby GP, Knox LS, Crosby LO et al (1988) Study protocol: a randomized clinical trial of total parenteral nutrition in malnourished surgical patients. Am J Clin Nutr 47(2 Suppl):366–381PubMed Buzby GP, Knox LS, Crosby LO et al (1988) Study protocol: a randomized clinical trial of total parenteral nutrition in malnourished surgical patients. Am J Clin Nutr 47(2 Suppl):366–381PubMed
5.
Zurück zum Zitat The Veterans Affairs Total Parenteral Nutrition Cooperative Study Group (1991) Perioperative total parenteral nutrition in surgical patients. N Engl J Med 325:525–532CrossRef The Veterans Affairs Total Parenteral Nutrition Cooperative Study Group (1991) Perioperative total parenteral nutrition in surgical patients. N Engl J Med 325:525–532CrossRef
6.
Zurück zum Zitat Weimann A, Braga M, Harsanyi L et al (2006) ESPEN Guidelines on enteral nutrition: surgery including organ transplantation. Clin Nutr 25(2):224–244PubMedCrossRef Weimann A, Braga M, Harsanyi L et al (2006) ESPEN Guidelines on enteral nutrition: surgery including organ transplantation. Clin Nutr 25(2):224–244PubMedCrossRef
7.
Zurück zum Zitat ASPEN Board of Directors and the Clinical Guidelines Task Force (2002) Guidelines for the use of parenteral and enteral nutrition in adult and pediatric patients. JPEN J Parenter Enteral Nutr 26(1 Suppl):1SA–138SA ASPEN Board of Directors and the Clinical Guidelines Task Force (2002) Guidelines for the use of parenteral and enteral nutrition in adult and pediatric patients. JPEN J Parenter Enteral Nutr 26(1 Suppl):1SA–138SA
8.
Zurück zum Zitat Braga M, Gianotti L, Nespoli L et al (2002) Nutritional approach in malnourished surgical patients: a prospective randomized study. Arch Surg 137(2):174–180PubMedCrossRef Braga M, Gianotti L, Nespoli L et al (2002) Nutritional approach in malnourished surgical patients: a prospective randomized study. Arch Surg 137(2):174–180PubMedCrossRef
9.
Zurück zum Zitat Gianotti L, Braga M, Nespoli L et al (2002) A randomized controlled trial of preoperative oral supplementation with a specialized diet in patients with gastrointestinal cancer. Gastroenterology 122:1763–1770PubMedCrossRef Gianotti L, Braga M, Nespoli L et al (2002) A randomized controlled trial of preoperative oral supplementation with a specialized diet in patients with gastrointestinal cancer. Gastroenterology 122:1763–1770PubMedCrossRef
10.
Zurück zum Zitat Senkal M, Zumtobel V, Bauer KH et al (1999) Outcome and cost-effectiveness of perioperative enteral immunonutrition in patients undergoing elective upper gastrointestinal tract surgery: a prospective randomized study. Arch Surg 134:1309–1316PubMedCrossRef Senkal M, Zumtobel V, Bauer KH et al (1999) Outcome and cost-effectiveness of perioperative enteral immunonutrition in patients undergoing elective upper gastrointestinal tract surgery: a prospective randomized study. Arch Surg 134:1309–1316PubMedCrossRef
11.
Zurück zum Zitat Lobo DN, Williams RN, Welch NT et al (2006) Early postoperative jejunostomy feeding with an immune modulating diet in patients undergoing resectional surgery for upper gastrointestinal cancer: a prospective, randomized, controlled, double-blind study. Clin Nutr 25:716–726PubMedCrossRef Lobo DN, Williams RN, Welch NT et al (2006) Early postoperative jejunostomy feeding with an immune modulating diet in patients undergoing resectional surgery for upper gastrointestinal cancer: a prospective, randomized, controlled, double-blind study. Clin Nutr 25:716–726PubMedCrossRef
12.
Zurück zum Zitat Klek S, Kulig J, Sierzega M et al (2008) The impact of immunostimulating nutrition on infectious complications after upper gastrointestinal surgery: a prospective, randomized, clinical trial. Ann Surg 248:212–220PubMedCrossRef Klek S, Kulig J, Sierzega M et al (2008) The impact of immunostimulating nutrition on infectious complications after upper gastrointestinal surgery: a prospective, randomized, clinical trial. Ann Surg 248:212–220PubMedCrossRef
13.
Zurück zum Zitat Klek S, Kulig J, Sierzega M et al (2008) Standard and immunomodulating enteral nutrition in patients after extended gastrointestinal surgery: a prospective, randomized, controlled clinical trial. Clin Nutr 27(4):504–512PubMedCrossRef Klek S, Kulig J, Sierzega M et al (2008) Standard and immunomodulating enteral nutrition in patients after extended gastrointestinal surgery: a prospective, randomized, controlled clinical trial. Clin Nutr 27(4):504–512PubMedCrossRef
14.
Zurück zum Zitat Klek S, Sierzega M, Szybinski P (2011) Perioperative nutrition in malnourished surgical cancer patients: a prospective, randomized, controlled clinical trial. Clin Nutr 30(6):708–713PubMedCrossRef Klek S, Sierzega M, Szybinski P (2011) Perioperative nutrition in malnourished surgical cancer patients: a prospective, randomized, controlled clinical trial. Clin Nutr 30(6):708–713PubMedCrossRef
15.
Zurück zum Zitat Klek S, Sierzega M, Szybinski P et al (2011) The immunomodulating enteral nutrition in malnourished surgical patients: a prospective, randomized, double-blind clinical trial. Clin Nutr 30(3):282–288PubMedCrossRef Klek S, Sierzega M, Szybinski P et al (2011) The immunomodulating enteral nutrition in malnourished surgical patients: a prospective, randomized, double-blind clinical trial. Clin Nutr 30(3):282–288PubMedCrossRef
16.
Zurück zum Zitat Turczynowski W, Szczepanik AM, Klek S (2000) Nutritional therapy and the immune system. Przeg Lek 57:36–40 Turczynowski W, Szczepanik AM, Klek S (2000) Nutritional therapy and the immune system. Przeg Lek 57:36–40
17.
Zurück zum Zitat Chen DW, Wei Fei Z, Zhang YC et al (2005) Role of enteral immunonutrition in patients with gastric carcinoma undergoing major surgery. Asian J Surg 28:121–124PubMedCrossRef Chen DW, Wei Fei Z, Zhang YC et al (2005) Role of enteral immunonutrition in patients with gastric carcinoma undergoing major surgery. Asian J Surg 28:121–124PubMedCrossRef
18.
Zurück zum Zitat Klek S, Kulig J, Szczepanik AM et al (2005) The clinical value of parenteral immunonutrition in surgical patients. Acta Chir Belg 105:175–179PubMed Klek S, Kulig J, Szczepanik AM et al (2005) The clinical value of parenteral immunonutrition in surgical patients. Acta Chir Belg 105:175–179PubMed
19.
Zurück zum Zitat Bozzetti F, Gianotti L, Braga M et al (2007) Postoperative complications in gastrointestinal cancer patients: the joint role of the nutritional status and the nutritional support. Clin Nutr 26:698–709PubMedCrossRef Bozzetti F, Gianotti L, Braga M et al (2007) Postoperative complications in gastrointestinal cancer patients: the joint role of the nutritional status and the nutritional support. Clin Nutr 26:698–709PubMedCrossRef
20.
Zurück zum Zitat Gianotti L (2006) Nutrition and infections. Surg Infect 7(Suppl 2):S29–S32 Gianotti L (2006) Nutrition and infections. Surg Infect 7(Suppl 2):S29–S32
21.
Zurück zum Zitat Zheng Y, Li F, Qi B et al (2007) Application of perioperative immunonutrition for gastrointestinal surgery: a meta-analysis of randomized controlled trials. Asia Pac J Clin Nutr 16(Suppl 1):253–257PubMed Zheng Y, Li F, Qi B et al (2007) Application of perioperative immunonutrition for gastrointestinal surgery: a meta-analysis of randomized controlled trials. Asia Pac J Clin Nutr 16(Suppl 1):253–257PubMed
22.
Zurück zum Zitat Heyland DK, Novak F, Drover JW et al (2001) Should immunonutrition become routine in critically ill patients? A systematic review of the evidence. JAMA 286(8):944–953PubMedCrossRef Heyland DK, Novak F, Drover JW et al (2001) Should immunonutrition become routine in critically ill patients? A systematic review of the evidence. JAMA 286(8):944–953PubMedCrossRef
23.
Zurück zum Zitat Horie H, Okada M, Kojima M et al (2006) Favorable effects of preoperative enteral immunonutrition on a surgical site infection in patients with colorectal cancer without malnutrition. Surg Today 36:1063–1068PubMedCrossRef Horie H, Okada M, Kojima M et al (2006) Favorable effects of preoperative enteral immunonutrition on a surgical site infection in patients with colorectal cancer without malnutrition. Surg Today 36:1063–1068PubMedCrossRef
26.
Zurück zum Zitat Wang ZD, Peng JS, Chen S et al (2006) Effects of perioperative enteral immunonutrition on nutritional status, immunity and inflammatory response of elderly patients. Zhonghua Yi Xue Za Zhi 86:1410–1413PubMed Wang ZD, Peng JS, Chen S et al (2006) Effects of perioperative enteral immunonutrition on nutritional status, immunity and inflammatory response of elderly patients. Zhonghua Yi Xue Za Zhi 86:1410–1413PubMed
28.
Zurück zum Zitat Giger U, Büchler M, Farhadi J et al (2007) Preoperative immunonutrition suppresses perioperative inflammatory response in patients with major abdominal surgery: a randomized controlled pilot study. Ann Surg Oncol 14:2798–2806PubMedCrossRef Giger U, Büchler M, Farhadi J et al (2007) Preoperative immunonutrition suppresses perioperative inflammatory response in patients with major abdominal surgery: a randomized controlled pilot study. Ann Surg Oncol 14:2798–2806PubMedCrossRef
29.
Zurück zum Zitat Satinsky I, Mittak M, Foltys A et al (2005) Comparison various types of artificial nutrition on postoperative complications after major surgery. Rozhl Chir 84:134–141PubMed Satinsky I, Mittak M, Foltys A et al (2005) Comparison various types of artificial nutrition on postoperative complications after major surgery. Rozhl Chir 84:134–141PubMed
30.
Zurück zum Zitat Alivizatos V, Athanasopoulos P, Makris N et al (2005) Early postoperative glutamine-supplemented parenteral nutrition versus enteral immunonutrition in cancer patients undergoing major gastrointestinal surgery. J Buon 10:119–122PubMed Alivizatos V, Athanasopoulos P, Makris N et al (2005) Early postoperative glutamine-supplemented parenteral nutrition versus enteral immunonutrition in cancer patients undergoing major gastrointestinal surgery. J Buon 10:119–122PubMed
31.
Zurück zum Zitat Helminen H, Raitanen M, Kellosalo J (2007) Immunonutrition in elective gastrointestinal surgery patients. Scand J Surg 96:46–50PubMed Helminen H, Raitanen M, Kellosalo J (2007) Immunonutrition in elective gastrointestinal surgery patients. Scand J Surg 96:46–50PubMed
32.
Zurück zum Zitat Calder PC (2007) Immunonutrition in surgical and critically ill patients. Br J Nutr 98(Suppl 1):S133–S139PubMed Calder PC (2007) Immunonutrition in surgical and critically ill patients. Br J Nutr 98(Suppl 1):S133–S139PubMed
33.
Zurück zum Zitat Kudsk KA, Tolley EA, DeWitt RC et al (2003) Preoperative albumin and surgical site identify surgical risk for major postoperative complications. JPEN J Parenter Enteral Nutr 27(1):1–9PubMedCrossRef Kudsk KA, Tolley EA, DeWitt RC et al (2003) Preoperative albumin and surgical site identify surgical risk for major postoperative complications. JPEN J Parenter Enteral Nutr 27(1):1–9PubMedCrossRef
34.
Zurück zum Zitat Daly JM, Lieberman MD, Goldfine J et al (1992) Enteral nutrition with supplemental arginine, RNA, and omega-3 fatty acids in patients after operation: immunologic, metabolic, and clinical outcome. Surgery 112:56–67PubMed Daly JM, Lieberman MD, Goldfine J et al (1992) Enteral nutrition with supplemental arginine, RNA, and omega-3 fatty acids in patients after operation: immunologic, metabolic, and clinical outcome. Surgery 112:56–67PubMed
35.
Zurück zum Zitat Heslin MJ, Latkany L, Leung D et al (1997) A prospective, randomized trial of early enteral feeding after resection of upper gastrointestinal malignancy. Ann Surg 226(4):567–577PubMedCentralPubMedCrossRef Heslin MJ, Latkany L, Leung D et al (1997) A prospective, randomized trial of early enteral feeding after resection of upper gastrointestinal malignancy. Ann Surg 226(4):567–577PubMedCentralPubMedCrossRef
36.
Zurück zum Zitat No authors listed (2001) Consensus recommendations from the US summit on immune-enhancing enteral therapy. JPEN J Parenter Enteral Nutr 25(2 Suppl):S61–S63CrossRef No authors listed (2001) Consensus recommendations from the US summit on immune-enhancing enteral therapy. JPEN J Parenter Enteral Nutr 25(2 Suppl):S61–S63CrossRef
Metadaten
Titel
Perioperative Immunonutrition in Surgical Cancer Patients: A Summary of a Decade of Research
verfasst von
Stanislaw Klek
Piotr Szybinski
Kinga Szczepanek
Publikationsdatum
01.04.2014
Verlag
Springer US
Erschienen in
World Journal of Surgery / Ausgabe 4/2014
Print ISSN: 0364-2313
Elektronische ISSN: 1432-2323
DOI
https://doi.org/10.1007/s00268-013-2323-z

Weitere Artikel der Ausgabe 4/2014

World Journal of Surgery 4/2014 Zur Ausgabe

Update Chirurgie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.

S3-Leitlinie „Diagnostik und Therapie des Karpaltunnelsyndroms“

Karpaltunnelsyndrom BDC Leitlinien Webinare
CME: 2 Punkte

Das Karpaltunnelsyndrom ist die häufigste Kompressionsneuropathie peripherer Nerven. Obwohl die Anamnese mit dem nächtlichen Einschlafen der Hand (Brachialgia parästhetica nocturna) sehr typisch ist, ist eine klinisch-neurologische Untersuchung und Elektroneurografie in manchen Fällen auch eine Neurosonografie erforderlich. Im Anfangsstadium sind konservative Maßnahmen (Handgelenksschiene, Ergotherapie) empfehlenswert. Bei nicht Ansprechen der konservativen Therapie oder Auftreten von neurologischen Ausfällen ist eine Dekompression des N. medianus am Karpaltunnel indiziert.

Prof. Dr. med. Gregor Antoniadis
Berufsverband der Deutschen Chirurgie e.V.

S2e-Leitlinie „Distale Radiusfraktur“

Radiusfraktur BDC Leitlinien Webinare
CME: 2 Punkte

Das Webinar beschäftigt sich mit Fragen und Antworten zu Diagnostik und Klassifikation sowie Möglichkeiten des Ausschlusses von Zusatzverletzungen. Die Referenten erläutern, welche Frakturen konservativ behandelt werden können und wie. Das Webinar beantwortet die Frage nach aktuellen operativen Therapiekonzepten: Welcher Zugang, welches Osteosynthesematerial? Auf was muss bei der Nachbehandlung der distalen Radiusfraktur geachtet werden?

PD Dr. med. Oliver Pieske
Dr. med. Benjamin Meyknecht
Berufsverband der Deutschen Chirurgie e.V.

S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“

Appendizitis BDC Leitlinien Webinare
CME: 2 Punkte

Inhalte des Webinars zur S1-Leitlinie „Empfehlungen zur Therapie der akuten Appendizitis bei Erwachsenen“ sind die Darstellung des Projektes und des Erstellungswegs zur S1-Leitlinie, die Erläuterung der klinischen Relevanz der Klassifikation EAES 2015, die wissenschaftliche Begründung der wichtigsten Empfehlungen und die Darstellung stadiengerechter Therapieoptionen.

Dr. med. Mihailo Andric
Berufsverband der Deutschen Chirurgie e.V.