• Non ci sono risultati.

Prevention of Infection Using Selective Decontami-nation of the Digestive Tract

N/A
N/A
Protected

Academic year: 2021

Condividi "Prevention of Infection Using Selective Decontami-nation of the Digestive Tract"

Copied!
17
0
0

Testo completo

(1)

Prevention of Infection Using Selective Decontami- nation of the Digestive Tract

L. S

ILVESTRI

, S. K

ERR

, A. G

ULLO

Rationale of Infection Prevention by Selective Decontami- nation of the Digestive Tract

The rationale for preventing infections using selective decontamination of the digestive tract (SDD) is based on the observation that 15 potentially pathogenic micro-organisms (PPMs) cause the three types of infection in the intensive care unit (ICU). The majority of ICU infections are “early” primary endogenous (Table 1). These require immediate adequate parenteral antibiotics in order to control incubating [1, 2] or treat established infections on admission to the ICU. Enteral antibiotics are administered into the throat and gut on admission to the unit to control acquisition, secondary carriage, and subsequent secondary endogenous infections that occur late, in general after 1 week of treatment on the ICU. Strict implementation of hygiene measures is required to control exogenous infection that may occur at any time during the treatment on the ICU. Finally, surveillance cultures of throat and rectum are an integral part of the strategy of SDD to mon- itor the efficacy of the protocol.

Table 1.Classification of infection occurring in the intensive care unit (ICU) using carria- ge (PPM potentially pathogenic micro-organism, AGNB aerobic Gram-negative bacilli, MRSA methicillin-resistant Staphylococcus aureus)

Type of infection PPM Timing Frequency (%)

Primary endogenous 6 normal and “Early” within About 55%

8 abnormal AGNB the 1st week MRSA

Secondary endogenous 8 abnormal AGNB “Late” after About 30%

MRSA the 1st week

Exogenous 8 abnormal AGNB Any time during About 15%

MRSA treatment on ICU

(2)

What Is SDD?

SDD is a maneuver designed to convert the “abnormal” carrier state (see Chapter 2) into the “normal” carrier state using non-absorbable antimicrobials.

The practice of SDD has four fundamental features termed the classical Stoutenbeek’s tetralogy [3, 4] (Fig. 1, Table 2).

Fig. 1.The Stoutenbeek’s tetralogy of selective decontamination of the digestive tract (SDD) and the type of infection prevented/controlled by each component. (PPM potentially patho- genic microorganism, ICU intensive care unit) (from Silvestri L, Lenhart FP, Fox MA (2001) Prevention of intensive care unit infections. Curr Anaesth Crit Care 12:35-40, with permission)

(3)

1. enteral antimicrobials given throughout the treatment on ICU, in combina- tion with

2. parenteral antibiotics given immediately on admission for 4 days 3. hand hygiene throughout the treatment on ICU

4. surveillance cultures of throat and rectum on admission and twice weekly thereafter

This strategy selectively targets the 15 PPMs and the high-level pathogens, such as Streptococcus pyogenes, which contribute to mortality. By design, SDD does not target low-level pathogens, including anaerobes, viridans streptococ- ci, enterococci, and coagulase-negative staphylococci as, in general, they only cause morbidity. The most-important feature of SDD is the enteral administra- tion of non-absorbable polymyxin E/tobramycin to eradicate the abnormal aer- obic Gram-negative bacilli (AGNB). This results in decontamination of the digestive tract. Critically ill patients are unable to clear these pathogens due to their underlying disease. Intestinal overgrowth with AGNB causes systemic immunoparalysis [4]. Enteral polymyxin E/tobramycin promotes recovery of systemic immunity [4] and prevention or eradication of abnormal AGNB in the throat and gut, and effectively controls aspiration and translocation of these

Table 2.Full four component protocol of SDD

Target PPM and antimicrobials Total daily dose (4x daily)

<5 years 5–12 years >12 years 1. enteral antimicrobials

A. oropharynx

AGNB: polymyxin E 2 g of 2% paste/gel

with tobramycin

Yeasts: amphotericin B, 2 g of 2% paste/gel

or nystatin

MRSA: vancomycin 2 g of 4% paste/gel

B. Gut

AGNB: polymyxin E (mg) 100 200 400

with tobramycin (mg) 80 160 320

Yeasts: amphotericin B (mg), 500 1,000 2,000

or nystatin (units) 2x106 4x106 8x106

MRSA: vancomycin (mg) 20-40/Kg 20-40/Kg 500/2,000

2. parenteral antimicrobials

cefotaxime (mg) 150/Kg 200/Kg 4,000

3. hygiene

4. surveillance cultures of throat and rectum on admission, Monday, Thursday

PPM, potentially pathogenic micro-oganism; AGNB, aerobic Gram-negative bacilli;

MRSA, methicillin-resistant Staphyloccoccus aureus

(4)

micro-organisms into the lower airways and blood, respectively. Enteral antimi- crobials are effective in the control of secondary endogenous infections.

However, the use of enteral antibiotics does not affect primary endogenous and exogenous infections. The second component is the immediate administration of an adequate parenteral antimicrobial to control primary endogenous pneu- monia and septicemia. Cefotaxime has been used in most randomized trials to cover both “community” and “hospital” pathogens. In adding enteral to par- enteral antibiotics, the original pre 1980s antibiotics remain useful, without the development of antimicrobial resistance (Chapter 28). Thirdly, high standards of hygiene are indispensable for reducing hand contamination and subsequent transmission from external sources. Finally, surveillance samples of the throat and rectum are taken on admission and twice weekly thereafter. These are an integral component of the SDD protocol. Knowledge of the carrier state allows the compliance with and efficacy of this prophylactic protocol to be monitored.

Effectiveness of SDD

In total, 54 randomized controlled trials (RCTs) have been undertaken over 20 years of clinical research (Table 3) [5–58]. SDD was actively researched during the 1990s and after a short interval there has been a revival in new randomized

Table 3.General characteristics of the 54 randomized trials (A, amphotericin B; Ami, amikacin; Cipro, ciprofloxacin; G, gentamicin; I, intestine; Nali, nalixidic; Neo, neomycin;

Net, netilmicin; Nor, norfloxacin; Ny, nystatin; O, oropharynx; Oflox, ofloxacin; P, polymyx- in; Pip, piperacillin; T, tobramycin; Van, vancomycin)

Enteral

Author Patients Parenteral AGNB Yeasts S. aureus Site

Abele-Horn et al. [5] Trauma Cefotaxime PT A O, -

Aerdts et al. [6] Mixed Cefotaxime P Nor A O, I

Arnow et al. [7] Liver Cefotaxime/ PG Ny O, I

transplantation ampicillin (2 arms)

Barret et al. [8] Pediatric, Pip/ami/ PT A -, I

burns van (2 arms)

Bergmans et al. [9] Mixed Antibiotics (40%) PG -- Van O, - (2 arms)

Bion et al. [10] Liver Cefotaxime/ PT A O, I

transplantation ampicillin (2 arms) (2 arms)

Blair et al. [11] Mixed Cefotaxime PT A O, I

Boland et al. [12] Trauma Cefotaxime PT Ny O, I

➝ Cont.

(5)

Enteral

Author Patients Parenteral AGNB Yeasts S. aureus Site

Bouter et al. [13] Cardiac Flucloxacillin P Neo -- -, I (2 arms)

Brun-Buisson Mixed -- P -- -,I

et al. [14] Neo

Nali

de la Cal et al. [17] Burns Cefotaxime PT A O, I

Cerra et al. [15] Mixed -- Nor Ny -, I

Cockerill et al. [18] Mixed Cefotaxime PG Ny O, I

Ferrer et al. [19] Respiratory Cefotaxime PT A O, I

(2 arms)

Finch et al. [20] Mixed Cefotaxime PG A O, I

Flaherty et al. [21] Cardiac Cefazolin PG Ny O, I

(2 arms)

Gastinne et al. [22] Mixed Antibiotics PT A O,I

(65%)(2 arms)

Gaussorges Mixed Antibiotics PG A Van -, I

et al. [23] (2 arms)

Georges et al. [24] Trauma Amoxycillin P Net A O, I

+clav (2 arms)

Hammond Respiratory Cefotaxime PT A O, I

et al. [25] (2 arms)

Hellinger et al. [26] Liver Ceftizoxime PG Ny O, I

transplantation (2 arms) (2 arms)

Jacobs et al. [27] Neurosurgical Cefotaxime PT A O, I

de Jonge et al. [16] Mixed Cefotaxime PT A O, I

Kerver et al. [28] Mixed Cefotaxime PT A O, I

Korinek et al. [29] Neurosurgical -- PT A Van O,I

Krueger et al. [30] Surgical, Ciprofloxacin PG -- Van O, I trauma

Laggner et al. [31] Mixed Amoxycillin+clav G A O, -

(70%) (2 arms) (2 arms)

➝ Table 3. Cont.

➝ Cont.

(6)

Enteral

Author Patients Parenteral AGNB Yeasts S. aureus Site

Lingnau et al. [32] Trauma Ciprofloxacin PT A O, I

(3 arms) P Cipro A O, I

Luiten et al. [33] Pancreatitis Cefotaxime P Nor A O, I

Martinez-Pellus Cardiac -- PT A -, I

et al. [34]

Martinez-Pellus Cardiac -- PT A -, I

et al. [35]

Palomar et al. [36] Mixed Cefotaxime PT A O, I

Pneumatikos Trauma -- PT A O, -

et al. [37]

Pugin et al. [38] Surgical, -- P Neo -- Van O, -

trauma

Quinio et al. [39] Trauma Cefazolin (38%) PG A O, I

(2 arms)

Rayes et al. [40] Liver Ceftriaxone/ PT A -,I

transplant metronidazole (2 arms)

Rocha et al. [41] Medical, Cefotaxime PT A O, I

trauma

Rodriguez-Roldan Mixed -- PT/Net A O, -

et al. [42]

Rolando et al. [43] Liver failure Cefuroxime PT A O, I

Rolando et al. [44] Liver failure Ceftazidime/ PT A O, I

flucloxacillin (2 arms)

(2 arms)

Ruza et al. [45] Pediatric -- PT Ny -, I

Sanchez-Garcia Mixed Ceftriaxone PG A O, I

et al. [46]

Schardey et al. [47] Gastrectomy Cefotaxime PT A Van -, I (2 arms)

➝ Cont.

➝ Table 3. cont.

(7)

SDD trials (Fig. 2). The randomized trials not only differ in combining enteral and/or parenteral antimicrobials, they also show large variations in the antimi- crobials used. The classical Stoutenbeek’s tetralogy has been evaluated in 17 RCTs [11, 16, 17, 19, 25, 27, 28, 36, 41, 43, 48-51, 54, 56, 58]. Infection was signif- icantly reduced in all but 12 studies. In those 12 negative studies the reduction was not significant [8, 14, 19, 22, 25, 26, 31, 32, 44, 54, 55, 58]. Only 2 studies showed a significant reduction in mortality following an intention-to-treat basis [16, 30]. The first study included 527 patients, and the reduction in mor- tality was significant with a relative risk of 0.69 [95% confidence interval (CI) 0.51–0.95]. The second study included 924 patients; both ICU [odds ratio (OR) 0.60, 95% CI 0.42–0.82] and hospital (OR 0.71, 95% CI 0.53–0.94) mortality were significantly reduced. In order to quantify the impact of SDD on infection and mortality meta-analysis is required.

➝ Table 3. cont.

Enteral

Author Patients Parenteral AGNB Yeasts S. aureus Site

Smith et al. [48] Pediatric, Cefotaxime/ PT A O, I

liver transplant ampicillin (2 arms)

Stoutenbeek Trauma Cefotaxime PT A O, I

et al. [49] (2 arms)

Stoutenbeek Trauma Cefotaxime PT A O, I

et al. [50]

Tetteroo et al. [51] Esophagectomy Cefotaxime PT A O, I

Ulrich et al. [52] Mixed Trimethoprim P Nor A O, I

Unertl et al. [53] Neurosurgical -- PG A O, I

Verwaest et al. [54] Mixed Cefotaxime PT A O, I

Ofloxacin Oflox A

Wiener et al. [55] Mixed -- PG Ny O, I

Winter et al. [56] Mixed Ceftazidime PT A O, I

Zobel et al. [57] Pediatric, Cefotaxime PG A O, I

Cardiac

Zwaveling et al. [58] Liver Cefotaxime/ PT A O, I

transplantation tobramycin (2 arms)

(8)

Meta-Analyses of Randomized Trials of SDD

Nine meta-analyses have been performed on RCTs only (Table 4) [59–67]. The

most rigorous meta-analysis [66] showed that SDD reduces the OR for lower

airway infections to 0.35 (95% CI 0.29–0.41) and mortality to 0.78 (95% CI

0.68–0.89), with a 6% overall mortality reduction from 30% to 24%. In surgical

patients the benefit is even higher, with the risk of lower airway infections

reduced by 81% (OR 0.19, 95% CI 0.15–0.26), bloodstream infections reduced

by 49% (OR 0.51, 95% CI 0.34–0.75), and mortality reduced by 30% (OR 0.70,

95% CI 0.52–0.93) [63]. The reason for this difference in improvement is that

surgical patients are in general younger and do not suffer from chronic under-

lying diseases, whereas medical patients are older and often have incurable con-

ditions such as diabetes, chronic obstructive pulmonary disease, and heart fail-

ure. In the 53rd and latest SDD trial the randomization unit was the ICU and

not the patient, as in all previous trials [16]. This Dutch study of 934 patients is

the largest single study yet undertaken. The primary endpoint was mortality as

opposed to infectious morbidity, and the risk of mortality was significantly

reduced to 0.6 (95% CI 0.4–0.8) in the unit where SDD was administered to all

patients. In contrast, the patient was the “randomization unit” in the previous

52 trials; therefore, half the population was not decontaminated. However, the

likelihood is high that those control patients, albeit not receiving SDD, will still

benefit from the intervention as concurrent patients were subjected to a lower

risk of microbial acquisition and carriage, acquired infection, and infection-

related mortality. This dilution risk due to the control group being present with

decontaminated patients at the same time in the same unit is termed “contam-

Fig. 2.Fifty-four randomized trials of SDD from 1987 to 2005

(9)

Table 4.Nine meta-analyses of randomized trials of SDD

Author Year Studies Patients Endpoints Results

(n) (n)

SDD trialists’ 1993 22 4,142 Odds ratio

group [59] Mortality 0.90 (0.79–1.04)

Parenteral and enteral 0.80 (0.67–0.97) antibiotics

Only enteral antibiotics 1.07 (0.86–1.32)

Pneumonia 0.37 (0.31–0.43)

Parenteral and enteral 0.33 (0.27–0.40) antibiotics

Only enteral antibiotics 0.43 (0.33–0.56)

Kollef [60] 1994 16 2,270 Difference in risk

Mortality 0.019

(-0.016 to 0.054)

Pneumonia 0.145

(0.016–0.174)

Heyland et al. [61] 1994 24 3,312 Relative risk

Mortality 0.87 (0.79–0.97)

Parenteral and enteral 0.81 (0.71–0.95) antibiotics

Only enteral antibiotics 1.00 (0.83–1.19)

Pneumonia 0.46 (0.39–0.56)

Parenteral and enteral 0.48 (0.39–0.60) antibiotics

Only enteral antibiotics 0.49 (0.32–0.59)

D’Amico et al. [62]1998 33 5,727 Odds ratio

Mortality

Parenteral and enteral 0.80 (0.69–0.93) antibiotics

Only enteral antibiotics 1.01 (0.84–1.22) Pneumonia

Parenteral and enteral 0.35 (0.29–0.41) antibiotics

Only enteral antibiotics 0.56 (0.46–0.68) cont.

(10)

Author Year Studies Patients Endpoints Results

(n) (n)

Nathens 1999 11 Not Odds ratio

et al. [63] mentioned

Mortality

Surgical patients 0.70 (0.52–0.93) Parenteral and enteral 0.60 (0.41–0.88) antibiotics

Only enteral antibiotics 0.86 (0.59–1.45) Non-surgical patients 0.91 (0.71–1.18) Parenteral and enteral 0.75 (0.53–1.06) antibiotics

Only enteral antibiotics 1.14 (0.77–1.68) Pneumonia

Surgical patients 0.19 (0.15–0.26) Non-surgical patients 0.45 (0.33–0.62)

Redman 2001 Not Not Odds ratio

et al. [64] mentioned mentioned

Pneumonia

Parenteral and enteral 0.31 (0.20–0.46) antibiotics

Only enteral antibiotics 0.40 (0.29–0.55)

Silvestri 2003 42 6,263 Odds Ratio

et al. [65] Fungal carriage 0.32 (0.19-0.53)

Fungal infections 0.31 (0.19-0.51)

Fungemia 0.49 (0.13-1.95)

Liberati 2004 36 6,922 Odds ratio

et al.[66] Mortality

Parenteral & enteral antibiotics 0.78 (0.68-0.89) Only enteral antibiotics 0.97 (0.81-1.16) Pneumonia

Parenteral & enteral antibiotics 0.35 (0.29-0.41) Only enteral antibiotics 0.52 (0.43-0.63)

Safdar 2004 4 259 Relative risk

et al.[67] (Liver Infection 0.88 (0.73-1.09)

transpl.) Gram negative infection 0.16 (0.07-0.37)

Mortality 0.82 (0.22-2.45)

➝ Table 4 cont.

(11)

ination bias”. The design of the latest trial has avoided this type of bias and may explain the highest reported mortality reduction to date. Translating this infor- mation into pragmatic terms, only 5 ICU patients need to be treated with SDD to prevent 1 case of pneumonia, and 21 ICU patients need to be treated to pre- vent 1 death [66]. The number needed to be treated to prevent 1 death in the lat- est study of de Jonge et al. is 12 [16].

Conclusions and Future Research

SDD is the only evidence-based maneuver that prevents infection and mortali- ty in the critically ill. The target micro-organisms of SDD include the “normal”

PPMs including Streptococcus pneumoniae and methicillin-sensitive

Staphylococcus aureus (MSSA) and the “abnormal” AGNB, including Klebsiella, Acinetobacter, and Pseudomonas species. Methicillin-resistant S. aureus

(MRSA), by design, is not covered by the original protocol of SDD, and, hence, seven randomized trials conducted in ICU where MRSA was endemic at the time of the study, showed a trend towards higher MRSA infection rates in patients receiving SDD [17, 19, 22, 25, 32, 54, 55]. These observations suggest that the parenteral and enteral antimicrobials of the SDD protocol, i.e., cefo- taxime, polymyxin, tobramycin, and amphotericin B, select and promote MRSA.

Under these circumstances, SDD requires the addition of oropharyngeal and intestinal vancomycin. Two RCTs show that the addition of vancomycin to SDD is an effective and safe maneuver [68, 69].

The Cochrane Library meta-analysis [66] - the only one that includes the first ever RCT on antimicrobial resistance [16] - reports that SDD does not lead to resistance amongst AGNB but, even better, the addition of enteral polymyxin/tobramycin to the parenteral antimicrobials reduces resistance com- pared with the parenteral antibiotics only. This is in line with a previous RCT demonstrating that enteral antimicrobials control extended spectrum β-lacta- mase producing Klebsiella [14]. Reports claiming resistance are invariably of low level evidence including before-after studies [70]. SDD implemented in two American ICUs with endemic vancomycin resistant enterococci (VRE) did not lead to an increased number of VRE infections [7, 26]. VRE did not emerge in any of the RCTs using enteral vancomycin [9, 23, 29, 30, 38, 47]. Antimicrobial resist- ance, being a long-term issue, has been evaluated in eight SDD studies monitor- ing antimicrobial resistance between 2 and 7 years, and bacterial resistance with SDD has not been a clinical problem [71-78].

A proper analysis of cost-effectiveness has not been performed. A Spanish

Working Party has undertaken a cost-effectiveness analysis of the 54 RCTs and

the nine meta-analyses using the decision tree analytic model [79, 80].

(12)

References

1. Sirvent JM, Torres A, Vidaur L, Armengol J, de Battle J, Bonet A (2000) Tracheal colo- nisation within 24 h of intubation in patients with head trauma: risk factor for deve- loping early-onset ventilator-associated pneumonia. Intensive Care Med 26:1369–1372 2. Sirvent JM, Torres A, El-Ebiary M, Castro P, de Battle J, Bonet A (1997) Protective effect of intravenously administered cefuroxime against nosocomial pneumonia in patients with structural coma. Am J Respir Crit Care Med 155:1729–1734

3. Stoutenbeek CP, van Saene HKF, Miranda DR, Zandstra DF (1984) The effect of selec- tive decontamination of the digestive tract on colonization and infection rate in mul- tiple trauma patients. Intensive Care Med 10:185–192

4. van Saene HKF, Petros AJ, Ramsay G, Baxby D (2003) All great truths are iconoclastic:

selective decontamination of the digestive tract moves from heresy to level 1 truth.

Intensive Care Med 29:677–690

5. Abele-Horn M, Dauber A, Bauernfeind A, Russwurm W, Seyfarth-Metzger I, Gleich P, Ruckdeschel G (1997) Decrease in nosocomial pneumonia in ventilated patients by selective oropharyngeal decontamination (SOD). Intensive Care Med 23:1878–1895 6. Aerdts SJA, van Dalen R, Clasener HAL, Festen J, van Lier HJJ, Vollaard EJ (1991)

Antibiotic prophylaxis of respiratory tract infection in mechanically ventilated patients. A prospective, blinded, randomized trial of the effect of a novel regimen.

Chest 100:783–791

7. Arnow PA, Caradang GC, Zabner R, Irwin ME (1996) Randomized controlled trial of selective decontamination for prevention of infections following liver transplantation.

Clin Infect Dis 22:997–1003

8. Barret JP, Jeschke MG, Herndon DN (2001) Selective decontamination of the digestive tract on severely burned pediatric patients. Burns 27:439–445

9. Bergmans DCJJ, Bonten MJM, Gaillard CA et al (2001) Prevention of ventilator-asso- ciated pneumonia by oral decontamination. A prospective, randomized, double-blind, placebo-controlled study. Am J Respir Crit Care Med 164:382–388

10. Bion JF, Badger I, Crosby HA et al (1994) Selective decontamination of the digestive tract reduces Gram-negative pulmonary colonization but not systemic endotoxemia in patients undergoing elective liver transplantation. Crit Care Med 22:40–49 11. Blair P, Rowlands BJ, Lowry K, Webb H, Armstrong P, Smilie J (1991) Selective decon-

tamination of the digestive tract: a stratified, randomized, prospective study in a mixed intensive care unit. Surgery 110:303–310

12. Boland JP, Sadler DL, Stewart W, Wood DJ, Zerick W, Snodgrass KR (1991) Reduction of nosocomial respiratory tract infections in the multiple trauma patients requiring mechanical ventilation by selective parenteral and enteral antisepsis regimen (SPEAR) in the intensive care (abstract). Abstracts of the 17th Congress of Chemotherapy, Berlin, nn. 0465

13. Bouter H, Schippers EF, Luelmo SAG, Versteegh MIM, Ros P, Guiot HFL, Frolich M, van Dissel JT (2002) No effect of preoperative selective gut decontamination on endotoxe- mia and cytokine activation during cardiopulmonary bypass: a randomized, placebo- controlled study. Crit Care Med 30:38–43

14. Brun-Buisson C, Legrand P, Rauss A et al (1989) Intestinal decontamination for con- trol of nosocomial multiresistant gram-negative bacilli. Study of an outbreak in an intensive care unit. Ann Intern Med 110:873–881

15. Cerra FB, Maddaus MA, Dunn DL, Wells CL, Kostantinides NN, Lehman SL, Mann HJ (1992) Selective gut decontamination reduces nosocomial infections and length of stay but not mortality or organ failure in surgical intensive care unit patients. Arch

(13)

Surg 127:163–169

16. de Jonge E, Schultz M, Spanjaard L, Bossuyt PPM, Vroom B, Dankert J (2003) Effects of selective decontamination of the digestive tract on mortality and acquisition of resi- stant bacteria in intensive care: a randomised controlled trial. Lancet 362:1011–1016 17. Cal MA de la, Cerda E, Garcia-Hierro P et al (2002) Survival benefit in severely burned

patients receiving selective decontamination of the digestive tract: a randomised, pla- cebo controlled, double blind trial. Med Intens 26:152

18. Cockerill FR, Muller SR, Anhalt JP et al (1992) Prevention of infection in critically ill patients by selective decontamination of the digestive tract. Ann Intern Med 117:545–553

19. Ferrer M, Torres A, Gonzalez J et al (1994) Utility of selective decontamination in mechanically ventilated patients. Ann Intern Med 120:389-395

20. Finch RG, Tomlinson P, Holliday M, Sole K, Stack C, Rocker G (1991) Selective decon- tamination of the digestive tract (SDD) in the prevention of secondary sepsis in a medical/surgical intensive care unit (abstract). Abstracts of the 17th Congress of Chemotherapy, Berlin, no. 0471

21. Flaherty J, Nathan C, Kabins SA, Weinstein RA (1990) Pilot trial of selective deconta- mination for prevention of bacterial infection in an intensive care unit. J Infect Dis 162:1393–1397

22. Gastinne H, Wolff M, Delatour F, Faurisson F, Chevret S, for the French Study Group on selective decontamination of the digestive tract (1992) A controlled trial in inten- sive care units of selective decontamination of the digestive tract with nonabsorbable antibiotics. N Engl J Med 326:594–599

23. Gaussorges P, Salord F, Sirodot M, Tigaud S, Cagnin S, Gerard M, Robert D (1991) Efficacité de la décontamination digestive sur la survenue des bacteriemies nosoco- miales chez les patients sous ventilation mécanique et recevant des betamimetiques.

Rean Soins Intens Med Urg 7:169–174

24. Georges B, Mazerolles M, Decun J-F et al (1994) Decontamination digestive selective:

resultats d’une etude chez le polytraumatise. Rean Urg 3:621–627

25. Hammond JMJ, Potgieter PD, Saunders GL, Forder AA (1992) Double-blind study of selective decontamination of the digestive tract in intensive care. Lancet 340:5–9 26. Hellinger WC, Yao JD, Alvarez S et al (2002) A randomized, prospective, double blin-

ded evaluation of selective bowel decontamination in liver transplantation.

Transplantation 73:1904–1909

27. Jacobs S, Foweraker JE, Roberts SE (1992) Effectiveness of selective decontamination of the digestive tract (SDD) in an ICU with a policy encouraging a low gastric pH. Clin Intensive Med 3:52–58

28. Kerver AJH, Rommes JH, Mevissen-Verhage EAE, Hulstaert PF, Vos A, Verhoef J, Wittebol P (1988) Prevention of colonization and infection in critically ill patients: a prospective randomized study. Crit Care Med 16:1087–1093

29. Korinek AM, Laisne MJ, Nicolas MH, Raskine L, Deroin V, Sanson-Lepors MJ (1993) Selective decontamination of the digestive tract in neurosurgical intensive care unit patients: a double-blind, randomized, placebo-controlled study. Crit Care Med 21:14661473

30. Krueger WA, Lenhart F-P, Neeser G et al (2002) Influence of combined intravenous and topical antibiotic prophylaxis on the incidence of infections, organ dysfunctions, and mortality in critically ill surgical patients. A prospective, stratified, randomized, dou- ble-blind, placebo-controlled clinical trial. Am J Respir Crit Care Med 166:1029–1037 31. Laggner AN, Tryba M, Georgopulos A et al (1994) Oropharyngeal decontamination

with gentamicin for long-stay ventilated patients on stress ulcer prophylaxis with sucralfate? Wien Klin Wochenschr 106:15–19

(14)

32. Lingnau W, Berger J, Javorsky F, Lejeune P, Mutz N, Benzer H (1997) Selective intesti- nal decontamination in multiple trauma patients: prospective, controlled trial. J Trauma 42:687–694

33. Luiten EJT, Hop WCJ, Lange JF, Bruining HA (1995) Controlled clinical trial of selecti- ve decontamination for the treatment of severe acute pancreatitis. Ann Surg 222:57–65 34. Martinez-Pellus AE, Merino P, Bru M et al (1993) Can selective digestive decontami- nation avoid the endotoxemia and cytokine activation promoted by cardiopulmonary bypass? Crit Care Med 21:1684–1691

35. Martinez-Pellus AE, Merino P, Bru M, Canovas J, Seller G, Sapina J, Fuentes T, Moro J (1997) Endogenous endotoxemia of intestinal origin during cardiopulmonary bypass.

Role of type of flow and protective effect of selective digestive decontamination.

Intensive Care Med 23:1251–1257

36. Palomar M, Alvarez-Lerma F, Jorda R, Bermejo B, and the Catalan study group of nosocomial pneumonia prevention (1997) Prevention of nosocomial infection in mechanically ventilated patients: selective digestive decontamination versus sucralfa- te. Clin Intensive Care 8:228–235

37. Pneumatikos I, Koulouras V, Nathanail C, Goe D, Nakos G (2002) Selective decontami- nation of subglottic area in mechanically ventilated patients with multiple trauma.

Intensive Care Med 28:432–437

38. Pugin J, Auckenthaler R, Lew DP, Suter PM (1991) Oropharyngeal decontamination decreases incidence of ventilator-associated pneumonia. A randomized, placebo-con- trolled, double-blind clinical trial. JAMA 265:2704–2710

39. Quinio B, Albanese J, Bues-Charbit M, Viavind X, Martin C (1996) Selective deconta- mination of the digestive tract in multiple trauma patients. A prospective double- blind, randomized, placebo-controlled study. Chest 109:765–772

40. Rayes N, Seehofer D, Hansen S, Boucsein K, Muller AR, Serke S et al (2002) Early ente- ral supply of Lactobacillus and fiber versus selective bowel decontamination: a con- trolled trial in liver transplant recipients. Transplantation 74:123-128

41. Rocha LA, Martin MJ, Pita S, Paz J, Seco C, Margusino L, Villanueva R, Duran MT (1993) Prevention of nosocomial infection in critically ill patients by selective decon- tamination of the digestive tract. A randomized, double-blind, placebo controlled study. Intensive Care Med 18:398–404

42. Rodriguez-Roldan JM, Altuna-Cuesta A, Lopez A, Carrillo A, Garcia J, Leon J Martinez- Pellus AJ (1990) Prevention of nosocomial lung infection in ventilated patients: use of an antimicrobial pharyngeal nonabsorbable paste. Crit Care Med 18:1239–1242 43. Rolando N, Gimson A, Wade J, Philpott-Howard J, Casewell M, Williams R (1993)

Prospective controlled trial of selective parenteral and enteral antimicrobial regimen in fulminant liver failure. Hepatology 17:196–201

44. Rolando N, Wade JJ, Stangou A, Gimson AES, Wendon J, Philpott-Howard J, Casewell MW, Williams R (1996) Prospective study comparing the efficacy of prophylactic parenteral antimicrobials with or without enteral decontamination, in patients with acute liver failure. Liver Transplant Surg 2:8–13

45. Ruza F, Alvarado F, Herruzo R, Delgado MA, Garcia S, Dorao P, Godel F (1998) Prevention of nosocomial infection in a pediatric intensive care unit (PICU) through the use of selective digestive decontamination. Eur J Epidemiol 14:719–727

46. Sanchez Garcia M, Cambronero Galache JA, Lopez Diaz Jet al (1998) Effectiveness and cost of selective decontamination of the digestive tract in critically ill intubated patients. A randomized, double-blind, placebo-controlled, multicenter trial. Am J Respir Crit Care Med 158:908–916

47. Schardey HM, Joosten U, Finke U et al (1997) The prevention of anastomotic leakage after total gastrectomy with local decontamination. A prospective, randomized, dou-

(15)

ble-blind, placebo-controlled, multicenter trial. Ann Surg 225:172–180

48. Smith SD, Jackson RJ, Hannakan CJ, Wadowsky RM, Tzakis AG, Rowe MI (1993) Selective decontamination in pediatric liver transplants. A randomized prospective study. Transplantation 55:1306–1309

49. Stoutenbeek CP, van Saene HKF, Zandstra DF (1996) Prevention of multiple organ fai- lure by selective decontamination of the digestive tract in multiple trauma patients. In:

Faist E, Baue AE, Schildberg FW (eds) The immune consequences of trauma, shock and sepsis—mechanisms and therapeutic approach. Pabst Science Publishers, Lengerich, pp 1055–1066

50. Stoutenbeek CP, van Saene HKF, Little RA, Whitehead A, for the working group on selective decontamination of the digestive tract (2005) The effect of selective deconta- mination of the digestive tract on mortality in multiple trauma patients. Intensive Care Med (in press)

51. Tetteroo GWM, Wagenvoort JHT, Castelein A, Tilanus HW, Ince C, Bruining HA (1990) Selective decontamination to reduce gram-negative colonisation and infections after oesophageal resection. Lancet 335:704–707

52. Ulrich C, Harinck-de Weerd JE, Bakker NC, Jacz K, Doornbos L, de Ridder VA (1989) Selective decontamination of the digestive tract with norfloxacin in the prevention of ICU-acquired infections: a prospective randomized study. Intensive Care Med 15:424–431

53. Unertl K, Ruckdeschel G, Selbmann HK, Jensen U, Forst H, Lenhart FP, Peter K (1987) Prevention of colonization and respiratory infections in long-term ventilated patients by local antimicrobial prophylaxis. Intensive Care Med 13:106–113

54. Verwaest C, Verhaegen J, Ferdinande P, Schetz M, van den Berghe G, Verbist L, Lauwers P (1997) Randomized, controlled trial of selective digestive decontamination in 600 mechanically ventilated patients in a multidisciplinary intensive care unit. Crit Care Med 25:63–71

55. Wiener J, Itokazu G, Nathan C, Kabins SA, Weinstein RA (1995) A randomized, double- blind, placebo-controlled trial of selective digestive decontamination in a medical- surgical intensive care unit. Clin Infect Dis 20:861–867

56. Winter R, Humphreys H, Pick A, MacGowan AP, Willatts SM, Speller DCE (1992) A controlled trial of selective decontamination of the digestive tract in intensive care and its effect on nosocomial infection. J Antimicrob Chemother 30:73–87

57. Zobel G, Kutting M, Grubbauer H-M, Semmelrock H-J, Thiel W (1991) Reduction of colonization and infection rate during pediatric intensive care by selective deconta- mination of the digestive tract. Crit Care Med 19:1242–1246

58. Zwaveling JH, Maring JK, Klompmaker IJ et al (2002) Selective decontamination of the digestive tract to prevent postoperative infection: a randomized placebo-controlled trial in liver transplant patients. Crit Care Med 30:1204–1209

59 Selective Decontamination of the Digestive Tract Trialists’ Collaborative Group (1993) Meta-analysis of randomised controlled trials of selective decontamination of the digestive tract. BMJ 307:525–532

60. Kollef M (1994) The role of selective digestive tract decontamination on mortality and respiratory tract infections. A meta-analysis. Chest 105:1101–1108

61. Heyland DK, Cook DJ, Jaeschke R, Griffith L, Lee HN, Guyatt GH (1994) Selective decontamination of the digestive tract: an overview. Chest 105:1221–1229

62. D’Amico R, Pifferi S, Leonetti C, Torri V, Tinazzi A, Liberati A (1998) Effectiveness of antibiotic prophylaxis in critically ill adult patients: systematic review of randomized controlled trials. BMJ 316:1275–1285

63. Nathens AB, Marshall JC (1999) Selective decontamination of the digestive tract in surgical patients. A systematic review of the evidence. Arch Surg 134:170–176

(16)

64. Redman R, Ludington E, Crocker M, Wittes J, Bellm L, Carlet J and the VAP Advisory Group (2001) Analysis of respiratory and non-respiratory infections in published trials of selective digestive decontamination. Intensive Care Med 27 [Suppl]:S285 65. Silvestri I, Milanese M, Gregori D, van Saene HKF (2003) Impact of SDD on fungal car-

riage and infection: a meta-analysis. Intensive Care Med 29[Suppl 1]:S128

66. Liberati A, D’Amico R, Pifferi S, Torri V, Brazzi L (2004) Antibiotic prophylaxis to redu- ce respiratory tract infections and mortality in adults receiving intensive care. Cochrane Review. In: The Cochrane Library, Issue 1. John Wiley and Sons, Chichester, UK 67. Safdar N, Said A, Lucey MR (2004) The role of selective digestive decontamination for

reducing infection in patients undergoing liver transplantation: a systematic review and meta-analysis. Liver Transpl 10:817-827

68. Sanchez M, Mir N, Canton R et al (1997) The effect of topical vancomycin on acquisi- tion, carriage and infection with methicillin-resistant Staphylococcus aureus in criti- cally ill patients. A double-blind, randomized, placebo controlled study. 37th ICAAC, Toronto, Canada. Abstract J-119:310

69. Silvestri L, van Saene HKF, Milanese M et al (2004) Prevention of MRSA pneumonia by oral vancomycin decontamination: a randomised trial. Eur Respir J 23:921-926 70. Baines P, van Saene H (2004) Selective decontamination of the digestive tract and pre-

vention of ventilator-associated pneumonia. Ann Intern Med 141:577-578

71. Hammond MJ, Potgieter PD (1995) Long-term effects of selective decontamination on antimicrobial resistance. Crit Care Med 23:637-645

72. Stoutenbeek CP, van Saene HKF, Zandstra DF (1987) The effect of oral non-absorba- ble antibiotics on the emergence of resistant bacteria in patients in an intensive care unit. J Antimicrob Chemother 19:513-520

73. Lingnau W, Berger J, Javorsky F et al (1998) Changing bacterial ecology during a five- year period of selective intestinal decontamination. J Hosp Infect 39:195-206 74. Sarginson RE, Taylor N, Reilly N et al (2004) Infection in prolonged pediatric critical

illness: A prospective four-year study based on knowledge of the carrier state. Crit Care Med 32:839-847

75. Leone M, Albanese J, Antonini F et al (2003) Long-term (6-year) effect of selective digestive decontamination on antimicrobial resistance in intensive care, multiple- trauma patients. Crit Care Med 31:2090-2095

76. Tetteroo GW, Wagenvoort JH, Bruining HA (1994) Bacteriology of selective deconta- mination: efficacy and rebound colonization. J Antimicrob Chemother 34:139-148 77. Silvestri L, Milanese M, Oblach L, Fontana F, Gregori D, Guerra R, van Saene HKF

(2002) Enteral vancomycin to control methicillin-resistant Staphylococcus aureus out- break in mechanically ventilated patients. Am J Infect Control 30:391–399

78. de la Cal MA, Cerdà E, van Saene HKF, Garcia-Hierro P, Negro E, Parra ML, Arias S, Ballestreros D (2003) Effectiveness and safety of enteral vancomycin to control ende- micity of methicillin-resistant Staphylococcus aureus in a medical/surgical intensive care unit. J Hosp Infect 56:175–183

79. Chaix C, Durand-Zaleski I, Alberti C, Brun-Buisson C (1999) Control of endemic methicillin-resistant Staphylococcus aureus. A cost-benefit analysis in an intensive care unit. JAMA 282:1745–1751

80. Veenstra DL, Saint S, Sullivan SD (1999) Cost-effectiveness of antiseptic-impregnated central venous catheters for the prevention of catheter-related bloodstream infection.

JAMA 11:282:554–560

(17)

SECTION FOUR

I NFECTIONS ON ICU

Riferimenti

Documenti correlati

So, the number of examinations seems a crucial is- sue in every training program but we have to consider the learning curve and its slope. Little data is available regarding

ABSTRACT The clinical course of intensive care unit (ICU) patients may be complicated by a large spectrum of lower respiratory tract infections (LRTI), defined by

In the case of inverse substitution the left-context replaces a substitution node in the elementary tree; in the case of inverse adjoining from the left and inverse adjoining from

Jonge EJ de, Schultz M, Spanjaard L et al (2003) Effects of selective decontamination of the digestive tract on mortality and acquisition of resistant bacteria in intensive care:

[r]

Annual scores from the ®rst axis of Principal com- ponents analysis of Common Bird Census index of 29 farmland bird

The corresponding interatomic distances indicated in Figure S9 were used as initial values for three separate Ce-O and Ce-C shells during EXAFS fitting.. Coordination numbers were

La seconda parte dovrebbe avere un contenuto puntuale in quanto ha lo scopo di rendere certo lo status di figlio legalmente acquisito, ovvero la filiazione stabilita nello Stato