Infection Control in The Intensive Care
Infection Control in The Intensive Care
Infection Control in The Intensive Care
Care Unit
H. K. F. van Saene L. Silvestri
M. A. de la Cal A. Gullo
Editors
Infection Control
in the Intensive
Care Unit
Third Edition
123
H. K. F. van Saene M. A. de la Cal
Institute of Aging and Chronic Diseases Department of Intensive Care Medicine
University of Liverpool Hospital Universitario de Getafe
Liverpool Getafe, Madrid
UK Spain
L. Silvestri A. Gullo
Department of Emergency and Unit of Department of Anesthesia
Anesthesia and Intensive Care and Intensive Care
Presidio Ospedaliero di Gorizia School of Medicine
Gorizia University Hospital Catania
Italy Catania
Italy
vii
Preface
ix
x Preface
We are grateful to Donatella Rizza, Catherine Mazars and Hilde Haala for the
their superb assistance. We hope that this third edition is instructive, and helpful in
your daily practice and that you enjoy it.
Part II Antimicrobials
6 Systemic Antibiotics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 67
A. R. De Gaudio, S. Rinaldi and C. Adembri
7 Systemic Antifungals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 99
C. J. Collins and Th. R. Rogers
xi
xii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 507
Contributors
xv
xvi Contributors
1.1 Introduction
R. E. Sarginson (&)
Paediatric Anaesthesia, Royal Liverpool Childrens NHS Trust,
Liverpool, Merseyside, UK
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 3
DOI: 10.1007/978-88-470-1601-9_1, Springer-Verlag Italia 2012
4 R. E. Sarginson et al.
Bone raised some important issues [58] for the terms sepsis and inflammation,
a debate that continues. Other interesting approaches in the fields of sepsis, sys-
temic inflammatory response, and multiple organ dysfunction are the use of
physiological state space concepts by Rixen et al. [9] and ideas from complex
adaptive system and network theory [1013]. A number of consensus confer-
ences have been held in recent years to seek agreement on definitions of infections
as they apply to patients in the intensive care unit (ICU) [14].
The glossary outlined here forms a basis for our clinical practice in various
aspects of intensive care infection and microbiology. We advocate definitions that
are usable in routine clinical practice and that emphasize the role of surveillance
samples in classifying the origins of infection.
1.2.1 Acquisition
Bloodstream infections (BSI) were classified into primary, secondary, and catheter
related by the International Consensus Forum on ICU infections in 2005 [14].
Debate continues over the number and type of cultures required to detect patho-
gens in the blood [15]. The clinical impact of BSI depends on the pathogenicity of
the invading microorganism, together with the nature and severity of the host
response (see Microorganisms, and Systemic inflammatory response syndrome
(SIRS), sepsis, and septic shock definitions).
1.2.5 Colonization
Microorganisms are present in body sites that are normally sterile, such as the
lower airways or bladder. Clinical features of infection are absent. Diagnostic
samples yield B1+ leukocytes per high power field (HPF) [19], and microbial
growth is \2+ or \105 CFU/ml.
1.2.6 Defense
1.2.7 Endemicity
Endemicity is defined as at least one new case per month having a diagnostic
sample positive for the outbreak strain. Endemicity can be interpreted as an
uncontrolled, ongoing outbreak.
1.2.8 Infection
Inflammatory markers are cells and proteins associated with the proinflammatory
process. These include C-reactive protein [20], procalcitonin [21, 22], tumor
necrosis factor alpha (TNF)-a, interleukin (IL)-1 and IL-6 [24], lymphocytes, and
neutrophils. The onset, magnitude, and duration of changes in these factors vary
with infection site and severity.
8 R. E. Sarginson et al.
ICU infection refers to secondary endogenous and exogenous infections, which are
infections due to organisms not carried by the patient at the time of ICU admission
and transmitted via hands of carers [25]. The term nosocomial (literally, related to
the hospital) is widely used but lacks a precise definition.
1.2.12 Isolation
Patients are nursed in separate cubicles or rooms, with strict hygiene measures,
including protective clothing and hand washing by the staff, to control transmis-
sion of microorganisms. These measures particularly apply to patients infected
with high-level pathogens or resistant microorganisms and those with impaired
immunity.
1.2.13 Microorganisms
1.2.14 Migration
Migration is the process whereby microorganisms carried in the throat and gut
move to colonize and possibly infect internal organs. Migration is promoted by
underlying chronic disease, some drugs, and invasive devices.
1.2.15 Outbreak
1.2.16 Overgrowth
1.2.17 Pneumonia
1.2.18 Resistance
1.2.19 Samples
1.2.19.1 Diagnostic
Diagnostic or clinical samples are taken from sites that are normally sterile in
order to diagnose infection or evaluate response to therapy. Samples are taken on
clinical indication only from blood, lower airways, CSF, urinary tract, wounds,
peritoneum, joints, sinuses, or conjunctiva.
1.2.19.2 Surveillance
Surveillance samples are taken from the oropharynx and rectum on admission and
subsequently at regular intervals (usually twice weekly). These specimens are
needed to:
evaluate the abnormal carriage level of potentially pathogenic microorganisms,
in particular, overgrowth;
assess the eradication of potential pathogens by enteral nonabsorbable anti-
microbial regimens used in SDD protocols;
detect the carriage of resistant strains.
Definitions for SIRS, sepsis, severe sepsis, and septic shock have been extensively
reviewed in recent years, particularly in relation to the inclusion criteria for
clinical trials [8, 40, 41]. Consensus definitions form categories based on cutoff
12 R. E. Sarginson et al.
1.2.21.2 Sepsis
Sepsis is defined as SIRS with a clear infectious etiology.
1.2.21.3 Septicemia
Septicemia is sepsis with a positive blood culture. In contrast, bacteremia
is defined as a positive blood culture in a patient exhibiting no clinical
symptoms.
1.2.22 Sinusitis
1.2.23 Tracheitis/Bronchitis
1.2.25 Transmission
Urinary tract infection is defined as infection of the urinary tract, most fre-
quently the bladder. The common clinical features of dysuria, suprapubic pain,
frequency, and urgency are often absent in the sedated ICU patient. The
diagnosis rests on a freshly voided catheter urine specimen or suprapubic
sample containing C105 bacteria or yeasts per milliliter of urine and C5
WBC/HPF.
14 R. E. Sarginson et al.
References
1. Feinstein AR (1967) Clinical judgment. Williams and Wilkins, Baltimore
2. Feinstein AR (1994) Clinical judgment revisited: the distraction of quantitative models. Ann
Intern Med 120:799805
3. Bonten M (1999) Controversies on diagnosis and prevention of ventilator-associated
pneumonia. Diagn Microbiol Infect Dis 34:199204
4. Toltzis P, Rosolowski B, Salvator A (2001) Etiology of fever and opportunities for reduction
of antibiotic use in a pediatric intensive care unit. Infect Control Hosp Epidemiol 22:499504
5. Bone RC (1991) Lets agree on terminology: definitions of sepsis. Crit Care Med 19:973976
6. Canadian Multiple Organ Failure Study Group (1991) Sepsisclarity of existing
terminologyor more confusion? Crit Care Med 19:996998
7. Bone RC, Grodzin CJ, Balk RA (1997) Sepsis: a new hypothesis for pathogenesis of the
disease process. Chest 112:235243
8. Levy MM, Fink MP, Marshall JC et al (2003) 2001 SCCM/ESICM/ACCP/ATS/SIS
international sepsis definitions conference. Crit Care Med 31:12501256
9. Rixen D, Siegel JH, Friedman HP (1996) Sepsis/SIRS, physiologic classification, severity
stratification, relation to cytokine elaboration and outcome prediction in post trauma critical
illness. J Trauma 41:581598
10. Seeley AJE, Christou NV (2000) Multiple organ dysfunction syndrome: exploring the
paradigm of complex non-linear systems. Crit Care Med 28:21932200
11. Toweill DL, Goldstein B (1998) Linear and nonlinear dynamics and the pathophysiology of
shock. New Horiz 6:155168
12. Aird WC (2002) Endothelial cell dynamics and complexity theory. Crit Care Med
30(suppl):S180S185
13. Strogatz SH (2001) Exploring complex networks. Nature 410:268276
14. Calandra T, Cohen J (2005) The international sepsis forum consensus conference on
definitions of infection in the intensive care unit. Crit Care Med 33(7):15381548
15. Lee A, Mirrett S, Reller LB et al (2007) Detection of bloodstream infections in adults: how
many blood cultures are needed? J Clin Microbiol 46(3):35463548
16. Smith TL, Nathan BR (2002) Central nervous system infections in the immune-competent
adult. Curr Treat Options Neurol 4:323332
17. Carrol ED, Thomson AP, Shears P et al (2000) Performance characteristics of the polymerase
chain reaction assay to confirm clinical meningococcal disease. Arch Dis Child 83:271273
18. Cursons RTM, Jeyerajah E, Sleigh JW (1999) The use of the polymerase chain reaction to
detect septicemia in critically ill patients. Crit Care Med 27:937940
19. ACourt CHD, Garrard CS, Crook D et al (1993) Microbiological lung surveillance in
mechanically ventilated patients, using non-directed bronchial lavage and quantitative
culture. Q J Med 86:635648
20. Reny JL, Vuagnat A, Ract C et al (2002) Diagnosis and follow up of infections in intensive
care patients: value of C-reactive protein compared with other clinical and biological
variables. Crit Care Med 30:529535
21. Claeys R, Vinken S, Spapen H et al (2002) Plasma procalcitonin and C-reactive protein in
acute septic shock: clinical and biological correlates. Crit Care Med 30:757762
1 Glossary of Terms and Definitions 15
44. Marik PE (2002) Definition of sepsis: not quite time to dump SIRS. Crit Care Med
30:706708
45. Zahorec R (2000) Definitions for the septic syndrome should be re-evaluated. Intensive Care
Med 26:1870
46. Lucy Lum Chai S (2005) Bloodstream infection in children. Pediatr Crit Care Med
6(Suppl):S42S44
Carriage, Colonization and Infection
2
L. Silvestri, H. K. F. van Saene
and J. J. M. van Saene
2.1 Introduction
Physiologically, internal organs such as lower airways and bladder, are sterile.
However, colonization of lower airways and bladder by potentially pathogenic
microorganisms (PPMs) is common in critically ill patients [1]. Colonization of
the internal organs generally follows impaired carriage defense of the digestive
tract, which promotes PPM carriage and overgrowth, and impaired defenses of the
host against colonization due to illness severity. Failure to clear colonizing
microorganisms from the internal organs invariably leads to high concentrations of
PPMs, predisposing to infection. The host mobilizes both humoral and cellular
defense systems to hinder the invading microorganisms. However, infection requires
not only invasion but severity of the underlying disease, which jeopardizes immu-
nocompetence. This chapter defines the concepts of carriage, colonization, and
infection and describes the host defense mechanisms against carriage, colonization,
and infection.
2.2 Definitions
Carriage is defined as the patients state in which the same strain is isolated from
at least two surveillance samples (saliva, gastric fluid, feces, throat, rectum) in
any concentration over a period of at least 1 week (Fig. 2.1). Overgrowth is
defined as the presence of C2+ or C105 colony forming units (CFU) per
L. Silvestri (&)
Department of Emergency, Unit of Anesthesia and Intensive Care,
Presidio Ospedaliero di Gorizia, Gorizia, Italy
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 17
DOI: 10.1007/978-88-470-1601-9_2, Springer-Verlag Italia 2012
18 L. Silvestri et al.
Acquisition
Carriage
Overgrowth
Colonization
Infection
Fig. 2.1 The slippery slope of the pathogenesis of infection in critically ill patients. Acquisition
develops if only one surveillance sample is positive for a potentially pathogenic microorganism
(PPM) that differs from the previous and following isolates. Acquisition refers to the transient
presence of a microorganism (usually in the oropharynx and gut), whereas carriage is a persistent
phenomenon. Carriage or carrier state is the patients state in which the same bacterial strain is
isolated from at least two surveillance samples (saliva, gastric fluid, feces, throat, rectum) in any
concentration over a period of at least 1 week. Overgrowth is defined as C105 colony-forming units
(CFU)/ml of saliva or gastric fluid or gram of feces and is nearly always present in the critically ill
intensive care unit (ICU) patient with impaired gut motility. Colonization is the presence of a PPM
in an internal organ that is normally sterile (e.g., lower airways, bladder) without inflammatory host
response. The diagnostic sample yields \105 CFU/ml of diagnostic sample. Infection is a
microbiologically proven clinical diagnosis of inflammation. Apart from the clinical signs of
infection, the diagnostic sample obtained from the internal organ contains C105 CFU/ml or is
positive in blood, cerebrospinal fluid, and pleural fluid. Surveillance samples are samples from body
sites where potentially pathogenic microorganisms are carried, such as digestive tract and skin
lesions (tracheotomy, wounds, pressure sores). A surveillance set comprises throat and rectal swabs
taken on admission and twice weekly thereafter, e.g., on Monday and Thursday. The purpose of
surveillance samples is to determine the microbiological endpoint of the level of PPM carriage.
Diagnostic samples are samples from internal organs that are normally sterile, such as lower
airways, blood, and bladder. The aim of diagnostic samples is clinical, i.e., to microbiologically
prove a diagnosis of inflammation, either generalized or local
2 Carriage, Colonization and Infection 19
2.3.1 Carriage
Secretions from internal organs, such as the lower airways, sinuses, middle ear,
lachrymal gland, and urinary tract, of healthy individuals are normally sterile.
Colonization of internal organs can occur with the two types of PPMs: normal,
including S. pneumoniae and H. influenzae; and abnormal, such as Klebsiella and
Pseudomonas spp. Three examples illustrate the concept of colonization followed
by infection:
1. Elderly people cared for in a nursing home carry S. pneumoniae and H. influenzae
in their oropharynx. During winter months, elderly people are at high risk of
developing the flu. The flu virus destroys the cilia and causes systemic
immunosuppression. Colonization of the lower airways with S. pneumoniae and
H. influenzae invariably occurs in this population during a flu epidemic. If these
patients do not receive a short course of commonly used antibiotics, colonization
of the lower airways often progresses to pneumonia associated with high
mortality rates. A similar pattern has been described for previously healthy
trauma patients admitted to the ICU [68].
2. COPD patients with a forced expiratory volume in 1 s (FEV1) \50% are
oropharyngeal carriers of both types of flora, including H. influenzae and
AGNB [9]. The severity of their underlying lung disease promotes colonization
of the lower airways with oral flora, including normal and abnormal bacteria.
The presence of bacteria in the lower airways, or colonization, is proinflam-
matory and may result in a range of important effects on the lung, including
activation of host defenses with release of inflammatory cytokines and sub-
sequent neutrophil recruitment, mucus hypersecretion, impaired mucociliary
clearance, and respiratory cell damage [10, 11, 12]. Bacterial colonization
of lower airways in COPD patients modulates the character and frequency
of exacerbations and is associated with greater airway inflammation and
accelerated decline in FEV1 [13]. An acute exacerbation of their underlying
condition may require intubation and ventilation in the ICU. The immediate
administration of an adequate antimicrobial that is active against H. influenzae
2 Carriage, Colonization and Infection 21
1. Phagocytic cells
2. Immunity
Six factors:
1. Anatomy 2. Physiology
3. Cilia 4. Mucus
5. IgA 6. Cell turnover
Seven factors:
1. Anatomy 2. Physiology
3. Cilia 4. Mucus
5. IgA 6. Cell turnover
7. Indigenous flora
Fig. 2.2 Three hurdles defending against carriage, colonization, and infection
7. Indigenous flora. These flora are primarily anaerobes and the indigenous
E. coli. Indigenous flora helps control carriage of acquired AGNBthe so-called
colonization resistance. In most cases, the beneficial functions of the indigenous
flora outweigh potentially harmful side effects. The microbiota provides diges-
tive functions, modulates host metabolism, and stimulates development of
lymphatic tissue and the mucosal immune system [5, 18]. Moreover, it can
efficiently limit gut infection by pathogenic bacteria. In fact [5, 18]:
normal flora acts as living wallpaper covering the mucosal receptor sites, thus
preventing AGNB adherence to those receptors;
anaerobes in high concentration require a huge quantities of nutrients,
resulting in AGNB starvation;
normal flora produces bacteriocidins that are bactericidal for AGNB, release
volatile fatty acids that create a growth-inhibiting environment, and are an
important source of energy for the gut epithelium (e.g., butyrate produced by
Faecalibacterium prausnitzii [19]);
presence of normal flora stimulates peristalsis;
indigenous flora stimulates host defenses; moreover,
deconjugation of bile salts by the indigenous flora is crucial for enterohepatic
circulation;
anaerobes produce b-lactamases that neutralize b-lactam antibiotics;
indigenous flora releases biopeptides, which play a role in gastroendocrine
metabolism, maintains water balance, promotes digestive tract motility, and
produces vitamins, including vitamin K, biotin, riboflavin, and folate [5].
[26, 27]. Fibronectin covers surface-cell receptors and thereby blocks attachment
of many microorganisms. The mucociliary blanket transports the invading
microorganisms out of the lung, and coughing aids this expulsion. In addition,
bronchial secretions contain various antimicrobial substances, such as lysozyme,
and defensins. Once the microorganism reaches the alveoli, the alveolar
macrophages and tissue histiocytes play an important role in protecting the host.
3. Cilia motility. In conjunction with mucus, cilia mechanically remove microor-
ganisms reaching its surface. Airway hygiene depends largely on mucociliary
clearance, which in turn depends upon movement of viscoelastic mucus along the
airway [28]. Aspirated or breathed material sticks to the mucus and is thus cleared
from the respiratory tract. Mucociliary clearance can be impaired by (a) genetic
defects, e.g., primary ciliary dyskinesia, cystic fibrosis; (b) secondary ciliary
dyskinesia due to artificial ventilation or toxins released by microorganisms
producing cytotoxic damage of epithelial cells (in this situation, microorganisms
may remain longer in the airways, causing colonization and infection);
(c) abnormal physicochemical properties of mucus, making it difficult to move it
along the airway. A persistent host inflammatory response driven by cytokines
fails to eliminate microorganisms and maintains the inflammatory process.
4. Secretory IgA. IgA in bronchial secretions coats microorganisms to prevent
adherence to mucosal cell receptors. Secretory IgA is the predominant
immunoglobulin present in the respiratory tract, nasal secretions, saliva, tears,
gastrointestinal fluids, and other mucous secretions. In addition, IgA can
neutralize toxin activity [29].
5. Mucosal cell turnover and desquamation. This process eliminates adherent
bacteria.
Similarly, six mechanisms are present to help prevent fecal PPMs from
colonizing the urinary tract [30] (Fig. 2.2):
1. Anatomical integrity. The bladder mucosa acts as a barrier to invading
microorganisms.
2. Intact physiology. Assists with clearing PPMs migrating from the rectal cavity
into the urethra and finally into the bladder [31]; extreme levels of osmolality,
high urea concentration, and low pH inhibit growth of some bacteria that cause
urinary tract infections;
3. Urinary flow. Mechanically removes PPMs unless they are capable of adhering
to epithelial cells in the urinary tract;
4. Mucus. covers the bladder mucosa.
5. Secretory IgA. Presence in mucus prevents adherence of fecal bacteria;
6. Mucosal cell turnover. Promotes elimination of PPMs already adhering to
bladder mucosal cells.
Colonizing microorganisms that are not eliminated from internal organs invariably
lead to a high concentration (C105) of PPMs, predisposing to invasion. The host
2 Carriage, Colonization and Infection 25
mobilizes both humoral and cellular defense systems to hinder the invading
microorganisms. However, infection requires both invasion and critical illness,
which jeopardize immunocompetence (Fig. 2.2).
There are two basic mechanisms of colonization and infection in ICU patients:
migration and translocation. Migration is the movement of live PPMs from one
place, e.g., throat and gut, where they are present in overgrowth, to another site, in
particular, normally sterile internal organs. Migration is the main mechanism
by which microorganisms may cause colonization/infection in ICU patients.
Migration of microorganisms in contaminated secretions from the oropharynx into
the lower airways within a few days of mechanical ventilation is considered to be
the most common route by which PPMs may enter the lung and cause colonization
and infection [22, 32, 33]. The severity of the underlying disease, which impairs
PPM clearance, is the main factor promoting colonization of the lower airways.
The presence of the plastic endotracheal tube is invariably associated with mucosal
lesions, which further enhances colonization. Finally, progression toward infection
depends on the patients immune status or defense capacity.
Potential pathogens may also cause colonization and subsequent infection,
bypassing the stage of carriage and overgrowth, i.e., exogenous colonization/
infection. An example is a lower respiratory tract colonization/infection in a
tracheotomized patient due to microorganisms not previously carried in throat
and/or gut but directly introduced following breaches of hygiene [34].
Translocation (or transmural migration) was originally defined by Berg and
Owens [35] as the passage of viable bacteria from the gut through the epithelium
to the lamina propria and hence to mesenteric lymph nodes and possibly other
organs. This was subsequently modified by Alexander et al. [36] to refer to the
movement of viable and nonviable microorganisms or their toxic products across
an intact intestinal barrier. Tsujimoto et al. [37] recently proposed a radical
revision of the definition, which includes translocation of pathogen-associated
molecular patterns. In normally healthy people, GALT macrophages are generally
effective in killing intestinal microorganisms translocating from the gut. When gut
function is impairedas in the critically ill patienteither in the anatomically
intact gastrointestinal tract or in altered intestinal mucosa, bacterial translocation
can spread into the systemic bloodstream, leading to sepsis and multiple organ
failure [38]. Gut overgrowth of PPMs, in particular, in the terminal ileum, is
required for translocation [39]. The phenomenon of translocation has been
described in surgical patients [40], patients with pancreatitis [23] and neutropenia
[41], in surgical neonates and infants receiving parenteral nutrition [42], and in
patients requiring intensive care, including mechanical ventilation [43]. Critical
illness impacts three elements of the gut: (1) it alters cellular proliferation and
death in the epithelium [44, 45]; it has a profound effect on the number of cells in
26 L. Silvestri et al.
the mucosal immune system [46, 47]; (3) it changes the normal carrier state into
abnormal carriage, defined as the persistent presence of abnormal potential
pathogens in the oropharynx and/or gut [22, 32, 33].
2.6 Conclusions
Only a general well-being guarantees the efficacy of carriage defenses, which are
based on seven innate host factors that facilitate clearing abnormal AGNB from the
gut, maintain normal flora, and subsequently prevent colonization and infection of
internal organs. Most importantly, the shift from normal to abnormal flora in
individuals with an underlying disease is thought to depend on the severity of the
illness. The use of antimicrobials, which impair the microbial factor of the carriage
defense system, further promotes gut carriage and overgrowth of abnormal flora.
Oropharyngeal and gastrointestinal eradication of abnormal flora using enteral,
nonabsorbable antimicrobials polymyxin B/tobramycin and amphotericin B is the
most logical approach by which to control or minimize the risk of PPM overgrowth
in the digestive and control colonization and infection of internal organs [48].
References
1. Kerver AJH, Rommes JH, Mevissen-Verhage EAE et al (1987) Colonization and infection in
surgical intensive care patients: a prospective study. Intensive Care Med 13:347351
2. Sarginson RE, Taylor N, van Saene HKF (2001) Glossary of terms and definitions.
Curr Anaesth Crit Care 12:25
3. Pittet D, Monod M, Suter PM et al (1994) Candida colonization and subsequent infections in
critically ill surgical patients. Ann Surg 220:751758
4. Mobbs KJ, van Saene HKF, Sunderland D, Davies PDO (1999) Oropharyngeal Gram-
negative bacillary carriage. A survey of 120 healthy individuals. Chest 115:15701575
5. Rosseneu S, Rios G, Spronk PE, van Saene JJM (2005) Carriage. In: van Saene HKF,
Silvestri L, de la Cal MA (eds) Infection control in the intensive care unit, 2nd edn. Springer,
Milan, pp 1536
6. Sirvent JM, Torres A, Vidaur L et al (2000) Tracheal colonisation within 24 h of intubation
in patients with head trauma: risk factor for developing early-onset ventilator-associated
pneumonia. Intensive Care Med 26:13691372
7. Ewig S, Torres A, El-Ebiary M et al (1999) Bacterial colonization patterns in mechanically
ventilated patients with traumatic and medical injury. Incidence, risk factors and association
with ventilator-associated pneumonia. Am J Respir Crit Care Med 159:188198
8. Acquarolo A, Urli T, Perone G et al (2005) Antibiotic prophylaxis of early onset pneumonia
in critically ill comatose patients. A randomized study. Intensive Care Med 31:510516
9. Mobbs KJ, van Saene HKF, Sunderland D, Davies PDO (1999) Oropharyngeal Gram-
negative bacillary carriage in chronic obstructive pulmonary disease: relation to severity of
disease. Respir Med 93:540545
10. Yamamoto C, Yoneda T, Yoshikawa M et al (1997) Airway inflammation in COPD patients
assessed by sputum levels of interleukin-8. Chest 112:505510
11. Sethi S, Murphy TF (2001) Bacterial infection in chronic obstructive pulmonary disease in
2000: state of the art. Clin Microbiol Rev 14:336363
12. Hillman KM, Riordan T, OFarrel SM, Tabacqchali S (1982) Colonization of the gastric
content in critically ill patients. Crit Care Med 10:444447
2 Carriage, Colonization and Infection 27
13. Patel IS, Seemungal TA, Wilks M et al (2002) Relationship between bacterial colonisation
and the frequency, character, and severity of COPD exacerbations. Thorax 57:753754
14. Silvestri L, Lenhart FP, Fox MA (2001) Prevention of intensive care unit infections.
Curr Anaesth Crit Care 12:3440
15. Proctor RA (1987) Fibronectin: a brief overview of its structure function and physiology.
Rev Infect Dis 9:S317S312
16. Dal Nogare AR, Toews GB, Pierce AK (1987) Increased salivary elastase precedes Gram-
negative bacillary colonization in post-operative patients. Am Rev Respir Dis 135:671675
17. Mestesky J, Russel M, Elson CO (1999) Intestinal IgA, novel views on its function in the
defence of the largest mucosal surface. Gut 44:25
18. Barber S, Wolf-Dietrich H (2011) Mechanisms controlling pathogen colonization of the gut.
Curr Opin Microbiol 14:8291
19. Sokol H, Pigneur B, Watterlot L et al (2008) Faecalibacterium prausnitzii is an anti-
inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease
patients. Proc Natl Acad Sci U S A 105:1673116736
20. van Saene HKF, Damjanovic V, Alcock SR (2001) Basics in microbiology for the patient
requiring intensive care. Curr Anaesth Crit Care 12:617
21. Marshall JC, Christou NV, Meakins JL (1988) Small-bowel bacterial overgrowth and
systemic immuno-suppression in experimental peritonitis. Surgery 104:404411
22. van Uffelen R, van Saene HKF, Fidler V et al (1984) Oropharyngeal flora as a source of
colonizing the lower airways in patients on artificial ventilation. Intensive Care Med 10:
233237
23. Luiten EJT, Hop WCJ, Endtz HP et al (1988) Prognostic importance of Gram-negative
intestinal colonization preceding pancreatic infection in severe acute pancreatitis. Intensive
Care Med 24:438445
24. Oodstijk EAN, de Smet AMGA, Kesecioglu J, Bonten MJM, on behalf of the Dutch SOD-SDD
trialists group (2011) The role of intestinal colonization with Gram-negative bacteria as a source
for intensive care unit-acquired bacteremia. Crit Care Med 39:961966
25. Manson CM, Summer WR, Nelson S (1992) Pathophysiology of pulmonary defence
mechanisms. J Crit Care 7:4256
26. Peacock SJ, Foster TJ, Cameron BJ, Berend R (1999) Bacterial fibronectin-binding proteins
and endothelial cell surface fibronectin mediate adherence of Staphylococcus aureus to
resting human endothelial cells. Microbiology 145:34773486
27. Mongodin E, Bajolet O, Cutrona J et al (2002) Fibronectin-binding proteins of
Staphylococcus aureus are involved in adherence to human airway epithelium. Infect
Immun 70:620630
28. Cole P (2001) Pathophysiology and treatment of airway mucociliary clearance. Minerva
Anestesiol 67:206209
29. Hienzel FP (2000) Antibodies. In: Mandell GL, Bennett JE, Dolin R (eds) Mandell, Douglas
and Bennetts principles and practice of infectious diseases. Churchill Livingstone,
Philadelphia, pp 4567
30. Kass EH, Schneiderman LJ (1957) Entry of bacteria into the urinary tract of patients with
implying catheters. N Engl J Med 256:556557
31. Kunin CM, Evans C, Bartholomew D, Bates DG (2002) The antimicrobial defense
mechanism of the female urethra: a reassessment. J Urol 168:413419
32. Johanson WG Jr, Pierce AK, Sandford JP et al (1972) Nosocomial respiratory tract infections
with Gram-negative bacilli: the significance of colonization of the respiratory tract. Ann
Intern Med 77:701706
33. Estes RJ, Meduri GU (1995) The pathogenesis of ventilator associated pneumonia: I.
Mechanisms of bacterial trans-colonization and airway inoculation. Intensive Care Med
21:365383
34. Morar P, Makura Z, Jones A et al (2000) Topical antibiotics on tracheostoma prevent
exogenous colonization and infection of lower airways in children. Chest 117:513518
28 L. Silvestri et al.
35. Berg RD, Owens WE (1979) Inhibition of translocation of viable Escherichia coli from the
gastrointestinal tract of mice by bacterial antagonism. Infect Immun 25:820827
36. Alexander JW, Boyce ST, Babcock GF et al (1990) The process of microbial translocation.
Ann Surg 212:496510
37. Tsujimoto H, Ono S, Mochizuki H (2009) Role of translocation of pathogen-associated
molecular patterns in sepsis. Dig Surg 26:100109
38. Sganga G, van Saene HKF, Brisinda G, Castagneto M (2001) Bacterial translocation.
In: van Saene HKF, Sganga G, Silvestri L (eds) Infection in the critically ill: an ongoing
challenge. Springer, Milan, pp 3545
39. Husebye E (1995) Gastro-intestinal motility disorders and bacterial overgrowth. J Intern Med
237:419427
40. Kane TD, Wesley Alexander J, Johannigman JA (1998) The detection of microbial DNA in
the blood. A sensitive method for diagnosing bacteremia and/or bacterial translocation in
surgical patients. Ann Surg 227:19
41. Tancrede CH, Andremont AO (1985) Bacterial translocation and Gram-negative bacteremia
in patients with hematological malignancies. J Infect Dis 152:99103
42. van Saene HKF, Taylor N, Donnell SC et al (2003) Gut overgrowth with abnormal flora: the
missing link in parenteral nutrition-related sepsis in surgical neonates. Eur J Clin Nutr
57:548553
43. Feltis BA, Wells CL (2000) Does microbial translocation play a role in critical illness?
Curr Opin Crit Care 6:117122
44. Coopersmith CM, Stromberg PE, Davis CG et al (2003) Sepsis from Pseudomonas
aeruginosa pneumonia decreases intestinal proliferation and reduces gut epithelial cell cycle
arrest. Crit Care Med 39:16301637
45. Husain KD, Stromberg PE, Woolsey CA et al (2005) Mechanisms of decreased intestinal
epithelial proliferation and increased apoptosis in murine acute lung injury. Crit Care Med
33:23502357
46. Fukatsu K, Sakamoto S, Hara E et al (2005) Gut ischemiareperfusion affects gut mucosal
immunity: a possible mechanism for infectious complications after severe surgical insults.
Crit Care Med 34:182187
47. Osterberg J, Ljungdahl M, Haglund U (2006) Influence of cyclooxygenase inhibitors on gut
immune cell distribution and apoptosis rate in experimental sepsis. Shock 25:147154
48. van Saene HKF (2008) The history of SDD. In: an der Voort PHJ, van Saene HKF (eds)
Selective digestive decontamination in intensive care medicine. Springer, Milan, pp 135
Classification of Microorganisms
According to Their Pathogenicity 3
M. A. de la Cal, E. Cerda, A. Abella
and P. Garcia-Hierro
3.1 Introduction
M. A. de la Cal (&)
Department of Intensive Care Medicine,
Hospital Universitario de Getafe, Getafe, Spain
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 29
DOI: 10.1007/978-88-470-1601-9_3, Springer-Verlag Italia 2012
30 M. A. de la Cal et al.
Term Definition
Pathogenicity The ability of microorganisms to induce disease, which may be assessed by
disease-carriage ratios
Virulencea The severity of the disease induced by microorganisms. In epidemiological
studies virulence may be assessed by mortality or morbidity rates and the
degree of communicability
Reservoir The place where the organism maintains its presence, metabolizes, and
replicates
Source The place from which the infectious agent passes to the host. In some cases,
the reservoir and the source are the same, but not always
Infection A microbiologically proven clinical diagnosis of inflammation
Carriageb Permanent (minimally 1 week) presence of the same strain in any
concentration in body sites normally not sterile (oropharynx, external nares,
gut, vagina, skin)
Abnormal carrier The abnormal carrier state exists when the isolated microorganism is not a
state constituent of normal flora (i.e., aerobic Gram-negative bacilli and
methicillin-resistant Staphylococcus aureus) [3]
Colonizationb The presence of microorganisms in an internal organ that is normally sterile
(e.g., lower airways, bladder). The diagnostic sample yields less than a
predetermined level of CFU/ml of diagnostic sample [3]
CFU colony-forming units
a
Some authors [4] consider virulence a synonym of pathogenicity
b
Some authors [2] define colonization as the permanent presence of a micro-organism in or on a
host without clinical expression. Carrier state is the condition of an individual colonized with a
specific organism. These definitions do not take the sterility of colonized sites in normal indi-
viduals into consideration
3.2.1 Habitat
It is estimated that the human body consists of approximately 1013 cells, and hosts
10141015 individual microorganisms [1]. These microorganisms can be divided into
two groups: those that usually remain constant in their normal habitat (indigenous
flora), and those that are accidentally acquired and that, after adherence to epithelial
or mucosal surfaces, have to compete with other microorganisms and host defenses.
The final outcome could be clearance or colonization of the new organisms.
Body areas that usually harbor microorganisms (Tables 3.2 and 3.3) [11] are
skin, mouth, nasopharynx, oropharynx and tonsils, large intestine and lower ileum,
external genitalia, anterior urethra, vagina, skin, and external ear. Nevertheless, the
various anatomical sites suitable for microbiological habitats display overlapping
boundaries and are subject to variation. Temporary habitats include larynx, tra-
chea, bronchi, accessory nasal sinuses, esophagus, stomach and upper portions of
the small intestine, and distal areas of the male and female genital organs [12].
Permanent colonization is often found in patients with some risks factors, i.e.,
chronic bronchitis [13].
32 M. A. de la Cal et al.
3.2.2 Flora
The range of this index is 01. The highest IPI found was for Pseudomonas
spp. (0.38). Other potential pathogens isolated had an IPI \0.1 (Enterobacter spp.
0.08; S. aureus 0.06; Klebsiella spp. 0.05; E. coli 0.05; S. epidermidis 0.03;
Enterococcus spp. 0). This index provides useful information about the relative
pathogenicity of different microorganisms in a specific population and could be
used to design antibiotic policies, both prophylactic and therapeutic, in selected
groups of patients in whom microbiological surveillance could be indicated
(e.g., burn, severe trauma patients).
There are inherent limitations related to the small number of studied patients
and the small number of infections (i.e., S. aureus: one infection over 17 colo-
nizations), which can give an unreliable estimation of the IPI. Furthermore, the
authors of the study suggested that the results should be interpreted taking into
account technical aspects (definitions used; sites chosen for surveillance; micro-
biological techniques; result interpretation) and population characteristics, because
IPI does not differentiate between the organisms intrinsic pathogenicity and other
factors (host and environment) that allow their expression. The extreme alterations
in host defense mechanisms in immunosuppressed patients is a good example of
the different pathogenicity of microorganisms depending on the specific type of
systemic immunosuppression, i.e., neutropenia or cellular (T lymphocyte) immune
defect [17].
In general, the classification of microorganisms according to their pathogenicity
is based on scales with few categories. Isenberg and DAmato [12] classify
organisms as commonly involved, occasionally involved, and rarely involved in
disease production. Murray et al. [3] classify the pathogenicity of organisms as
high, potential, and low. Categories in both classifications are not always equiv-
alent. We found that the Murray et al. [3] classification (Table 3.4) is useful in ICU
practice because it is best adapted to flora isolated in ICU patients and is more
discriminatory between organisms of interest in the ICU. Another possible
advantage is that this classification integrates other concepts of clinical epidemi-
ology, such as community, or normal; and hospital, or abnormal, flora.
the invasive management group and 25.8% in the clinical management group, i.e.,
a difference of 9.6%. This survival benefit can be explained by the fact that
significantly more patients who did not undergo bronchoscopy received early,
inadequate antimicrobial therapy (one patient died in the invasive group versus 24
in the control group; p \ 0.001). The survival benefit was only transient, as the
difference in mortality was no longer significant at day 28 (p = 0.10). Only a few
of the evaluable studies adjusted mortality data for appropriate antimicrobial
treatment, underlying disease, and illness severity.
Three studies in patients with positive blood cultures and that adjusted for
appropriate antibiotic treatment are available [3133]. Only one study in 106
patients reports a significantly higher mortality rate due to vancomycin-resistant
enterococci (VRE), with an OR of 4.0 (1.213.3). The other two studies in a total
of 467 patients failed to show a mortality rate difference.
One study in 135 patients compared mortality rates in patients with infections
due to piperacillin-resistant P. aeruginosa and patients with infections due to
piperacillin-sensitive P. aeruginosa [34]. Mortality data were not adjusted for
immediate, adequate antimicrobial therapy, as there was no difference in crude
mortality. There are no data on Acinetobacter mortality, whether sensitive or
resistant.
These data show that the association of antimicrobial resistance with mortality
rate has not yet been appropriately evaluated. Evidence supports the concept that
antibiotic resistance does not contribute to mortality.
3.5 Conclusions
References
1. Isenberg HD (1988) Pathogenicity and virulence: another view. Clin Microbiol Rev 1:4053
2. Brachman PS (1992) Epidemiology of nosocomial infections. In: Bennett JV, Brachman PS
(eds) Hospital infections, 3rd edn. Little Brown, Boston, pp 320
3. Murray AE, Mostafa SM, van Saene HKF (1991) Essentials in clinical microbiology. In:
Stoutenbeek CP, van Saene HKF (eds) Infection and the anaesthetist, vol 5. Bailliere Tindall,
London, pp 126
4. McCloskey RV (1979) Microbial virulence factors. In: Mandell GL, Douglas RG, Bennett IE
(eds) Principles and practice of infectious diseases, vol 1, 1st edn. Wiley, New York, pp 311
5. Pfaller MA, Herwald LA (1988) Laboratory, clinical, and epidemiological aspects of
coagulase-negative staphylococci. Clin Microbiol Rev 1:281299
6. Freeman I, Platt R, Sidebottom DG et al (1987) Coagulase-negative staphylococcal
bacteremia in the changing neonatal intensive care unit population. JAMA 258:25482552
7. Kerver AIH, Rommes IH, Mevissen-Verhage EAE et al (1987) Colonization and infection in
surgical intensive care patients. Intensive Care Med 13:347351
8. Leonard EM, van Saene HKF, Shears P, Walker I, Tam PKH (1990) Pathogenesis of
colonization and infection in a neonatal surgical unit. Crit Care Med 18:264269
9. van Saene HKF, Stoutenbeek CP, Zandstra DF, Gilberston A, Murray A, Hart CA (1987)
Nosocomial infections in severely traumatized patient: magnitude of problem, pathogenesis,
prevention and therapy. Acta Anaesthesiol Belg 38:347356
10. van Saene HKF, Damjanovic V, Murray AE, de la Cal MA (1996) How to classify infections
in intensive care unitsthe carrier state, a criterion whose time has come? J Hosp Infect
33:112
11. Tramont EC (1979) General or nonspecific host defense mechanisms. In: Mandell GL,
Douglas RG, Bennett IE (eds) Principles and practice of infectious diseases, vol 1, 1st edn.
Wiley, NewYork, pp 1321
12. Isenberg HD, DAmato RF (1990) Indigenous and pathogenic micro-organisms of humans.
In: Mandell GL, Douglas RG, Bennett IE (eds) Principles and practice of infectious diseases,
vol 1, 3rd edn. Churchill Livingstone, New York, pp 214
13. Jordan GW, Wong GA, Hoeprich PB (1976) Bacteriology of the lower respiratory tract as
determined by fiberoptic bronchoscopy and transtracheal aspiration. J Infect Dis 134:428435
14. van der Waaij D (1992) Selective gastrointestinal decontamination: history of recognition
and measurement of colonization of the digestive tract as an introduction to selective
gastrointestinal decontamination. Epidemiol Infect 109:315326
15. Mackowiak PA, Martin RM, Smith LW (1979) The role of bacterial interference in the
increased prevalence of oropharyngeal Gram-negative bacilli among alcoholics and diabetics.
Am Rev Respir Dis 120:289593
16. Leonard EM, van Saene HKF, Stoutenbeek CP, Walker I, Tam PKH (1990) An intrinsic
pathogenicity index for micro-organisms causing infection in a neonatal surgical unit. Microb
Ecol Health Dis 3:151157
17. Shelhamer IH, Toews GB, Masur H et al (1992) Respiratory disease in the immuno-
suppressed patient. Ann Intern Med 117:415443
18. Alvarez-Lerma F (1996) Modification of empiric antibiotic treatment in patients with
pneumonia acquired in the intensive care unit. ICU-acquired pneumonia study group.
Intensive Care Med 22:387394
19. Luna CM, Vujacich P, Niederman MS et al (1997) Impact of BAL data on the therapy and
outcome of ventilator-associated pneumonia. Chest 111:676685
3 Classification of Microorganisms According to Their Pathogenicity 39
20. Iregui M, Ward S, Sherman G, Fraser VJ, Kollef MH (2002) Clinical importance of delays in
the initiation of appropriate antibiotic treatment for ventilator associated pneumonia. Chest
122:262268
21. Valles J, Rello J, Ochagavia A, Garnacho J, Alcala MA (2003) Community-acquired
bloodstream infection in critically ill adult patients: impact of shock and inappropriate
antibiotic therapy on survival. Chest 123:16151624
22. Fagon JY, Chastre J, Wolff M et al (2000) Invasive and noninvasive strategies for
management of suspected ventilator associated pneumonia. A randomized trial. Ann Intern
Med 132:621630
23. Cosgrove SE, Sakoulas G, Perencevich EN, Schwaber MJ, Karchmer AW, Carmeli Y (2003)
Comparison of mortality associated with methicillin-resistant and methicillin-susceptible
Staphylococcus aureus bacteremia: a meta-analysis. Clin Infect Dis 36:5359
24. Conterno LO, Wey SB, Castelo A (1998) Risk factors for mortality in Staphylococcus aureus
bacteremia. Infect Control Hosp Epidemiol 19:3237
25. Romero-Vivas J, Rubio M, Fernndez C, Picazo JJ (1995) Mortality associated with
nosocomial bacteremia due to methicillin-resistant Staphylococcus aureus. Clin Infect Dis
21:14171423
26. Harbarth S, Rutschmann O, Sudre P, Pittet D (1998) Impact of methicillin resistance on the
outcome of patients with bacteremia caused by Staphylococcus aureus. Arch Intern Med
158:182189
27. Soriano A, Martnez JA, Mensa J et al (2000) Pathogenic significance of methicillin
resistance for patients with Staphylococcus aureus bacteremia. Clin Infect Dis 30:368373
28. Mylotte JM, Tayara A (2000) Staphylococcus aureus bacteremia: predictors of 30-day
mortality in a large cohort. Clin Infect Dis 31:11701174
29. Topeli A, Unal S, Akalin HE (2000) Risk factors influencing clinical outcome in
Staphylococcus aureus bacteraemia in a Turkish University Hospital. Int J Antimicrob
Agents 141:5763
30. Gonzlez C, Rubio M, Romero-Vivas J, Gonzlez M, Picazo JJ (1999) Bacteremic
pneumonia due to Staphylococcus aureus: a comparison of disease caused by methicillin-
resistant and methicillin-susceptible organisms. Clin Infect Dis 29:11711177
31. Garbutt JM, Ventrapragada M, Littenberg B, Mundy LM (2000) Association between
resistance to vancomycin and death in cases of Enterococcus faecium bacteremia. Clin Infect
Dis 30:466472
32. Vergis EN, Hayden MK, Chow JW et al (2001) Determinants of vancomycin resistance and
mortality rates in enterococcal bacteremia. A prospective multicenter study. Ann Intern Med
135:484492
33. Lodise TP, McKinnon PS, Tam VH, Rybak MJ (2002) Clinical outcomes for patients with
bacteremia caused by vancomycin-resistant enterococcus in a level 1 trauma center. Clin
Infect Dis 34:922929
34. Trouillet JL, Vuagnat A, Combes A, Kassis N, Chastre J, Gibert C (2002) Pseudomonas
aeruginosa ventilator-associated pneumonia: comparison of episodes due to piperacillin-
resistant versus piperacillin-susceptible organisms. Clin Infect Dis 34:10471054
35. Alverdy JC, Laughlin RS, Wu L (2003) Influence of the critically ill state on host-pathogen
interactions within the intestine: gut-derived sepsis redefined. Crit Care Med 31:598607
36. Relman DA, Falkow S (1990) A molecular perspective of microbial pathogenicicty. In:
Mandell GL, Douglas RG, Bennett IE (eds) Principles and practice of infectious diseases, vol
1, 3rd edn. Churchill Livingstone, New York, pp 2532
37. Emori TG, Gaynes RG (1993) An overview of nosocomial infections, including the role of
the microbiology laboratory. Clin Microbiol Rev 6:428442
38. Neish AS (2002) The gut microflora and intestinal epithelial cells: a continuing dialogue.
Microbes Infect 4:309317
39. Guarner F, Malagelada JR (2003) Gutflora in health and disease. Lancet 361:512519
40 M. A. de la Cal et al.
40. DAgata EMC, Venkataran L, De Girolami P, Burke P, Eliopoulos GM, Karchmer AW,
Samore MH (1999) Colonisation with broad-spectrum cephalosporin-resistant Gram-negative
bacilli in intensive care units during a non-outbreak period: prevalence, risk factors and rate
of infection. Crit Care Med 27:10901095
41. Safdar N, Maki DG (2002) The commonality of risk factors for nosocomial colonization and
infection with antimicrobial-resistant Staphylococcus aureus, Enterococcus Gram-negative
bacilli, Clostridium difficile and Candida. Ann Intern Med 136:834844
Classification of ICU Infections
4
L. Silvestri, H. K. F. van Saene
and A. J. Petros
4.1 Introduction
L. Silvestri (&)
Department of Emergency, Unit of Anesthesia and Intensive Care,
Presidio Ospedaliero di Gorizia, Gorizia, Italy
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 41
DOI: 10.1007/978-88-470-1601-9_4, Springer-Verlag Italia 2012
42 L. Silvestri et al.
The traditional approach has been challenged by the carrier state concept [2].
Carriage or carrier state exists when the same strain is isolated from at least two
consecutive surveillance samples (e.g., throat and rectal swabs) from an ICU
patient, at any concentration, over a period of at least 1 week [23]. A surveillance
set comprises throat and rectal swabs taken on admission and twice weekly
afterward (e.g., Monday and Thursday). Diagnostic or clinical samples are samples
from internal organs that are normally sterile, such as lower airways, blood,
44 L. Silvestri et al.
bladder and skin lesions and are only taken on clinical indication with the aim of
microbiologically proving a diagnosis of inflammation, either generalized or local
[23]. Knowledge of the carrier state, together with diagnostic cultures, allows the
distinction between the three types of infection occurring in the ICU [2, 24]
(Table 4.1):
1. Primary endogenous infections are the most frequent infections in the ICU;
the incidence varies between 50% and 85%, depending on the population
studied and their degree of immunosuppression. They are caused by both
normal and abnormal PPMs imported into the ICU by the patient in the
admission flora. These episodes of infection generally occur early, during
the first week of ICU stay. S. pneumoniae, H. influenzae and S. aureus are
the etiological agents in previously healthy individuals requiring intensive
care following an acute event, such as (surgical) trauma, pancreatitis, acute
hepatic failure and burns. Abnormal AGNB can cause primary endogenous
infections in patients with previous chronic underlying disease, such as
severe chronic obstructive pulmonary disease, following acute deterioration
of the underlying disease. Adequate parenteral antibiotics given immediately
on admission to the ICU reduce the incidence of primary endogenous
infection.
2. Secondary endogenous infections are invariably caused by one or more of eight
abnormal AGNB as well as methicillin-resistant S. aureus (MRSA), accounting
for one-third of all ICU infections. This type of infection, in general, occurs
after 1 week in the ICU. These PPMs are first acquired in the oropharynx and
subsequently in the gut. The topical application of nonabsorbable antimicro-
bials polymyxin E/tobramycin/amphotericin B has been shown to control
secondary endogenous infection.
3. Exogenous infections are caused by abnormal hospital PPMs (15%) and may
occur at any time during the patients stay in the ICU. Typical examples are
Acinetobacter lower airway infection following the use of contaminated
4 Classification of ICU Infections 45
Endogenous Exogenous
infection
infection
Yes No
Primary
endogenous
Secondary
infection
endogenous
infection
Fig. 4.1 Flowchart for classifying infections in the ICU using knowledge of the carrier state
(PPM potentially pathogenic microorganism)
Evidence that infections occurring on or at a specific time after ICU admission are
attributable solely to microorganisms transmitted via careers handsand hence
acquired during the ICU stayis limited. Classifications based on time have been
developed following the common experience of specific incubation times associated
with highly pathogenic microorganisms due to their high intrinsic pathogenicity
(or virulence) [25]. However, patients requiring intensive care develop infections with
PPMs, including MRSA and AGNB and with low-level pathogens such as CNS and
enterococci, due to the severity of their illness and associated immunosuppression
[26]. The severity of illness rather than microorganism virulence is the most important
factor for the conversion of the normal into the abnormal carrier state and will
determine the time at which a potential or a low-level pathogen will cause infection.
According to the pathogenesis of ICU-acquired infections, PPM acquisition is
followed by carriage and overgrowth of that microorganism before colonization
and infection of an internal organ occurs. Undoubtedly, this process takes more
than 24 days. Accordingly, a lower respiratory tract infection due to a PPM
already carried in the throat and/or gut on admission and that develops in a
ventilated trauma patient after 3, 4 or even 10 days from ICU admission, cannot
be considered as ICU acquired. The concept of early- and late-onset infection,
mainly pneumonia, remains accepted in general [21, 27]. Moreover, early-onset
nosocomial pneumonia is believed to be due primarily to normal flora, such as
H. influenzae, MSSA and S. pneumoniae, whereas late-onset nosocomial pneu-
monia is mainly caused by higher-level antibiotic-resistant AGNBs, such as
P. aeruginosa, Acinetobacter species, or MRSA [28]. These statements are not
generalizable to all ICU populations; patients with chronic underlying diseases, such
as diabetes, alcoholism, chronic obstructive pulmonary disease and liver disease,
may carry abnormal flora on ICU admission, including AGNB and/or MRSA, in the
throat and gut. This is why some authors found no difference in microorganisms
between early- and late-onset infections [28]. Moreover, it is unknown what is the
best cut-off time by which to separate early- from late-onset pneumonia, as it is
unknown how long it takes to develop pneumonia after aspiration of microorgan-
isms [27]. Therefore, many clinicians, in prolonging the cutoff time, implicitly
recognized the inaccuracy of the time classification method to determine whether an
infection was imported or was really nosocomial [13, 14, 28].
In contrast, knowledge of the carrier state at the time of admission and throughout
the ICU stay is indispensable in distinguishing infections due to imported PPMs
(i.e., primary endogenous) from infections due to bacteria acquired on the unit
4 Classification of ICU Infections 47
Table 4.2 Classification of ICU infections using the 48-h time cutoff compared with carrier-state criteria
4.5 Conclusions
Most ICU infections are due to microorganisms carried by the patient on admis-
sion to the unit. The difference in philosophy between the traditionalists and those
who advocate the carriage-state method for classifying ICU infections is that the
former focus on preventing transmission of all microorganisms via careers hands
in order to control all Gram-positive and Gram-negative infections occurring after
2 days of ICU stay. However, we believe that ICU patients may benefit from an
infection control program that includes surveillance of both carriage and infection.
In detecting abnormal carriage and overgrowth, surveillance cultures are indis-
pensable for identifying a subset of patients at high risk of infection. Awareness of
carriage in long-stay patients can provide more insights into the epidemiology of
infection. The true nosocomial infection problem (i.e., secondary endogenous and
exogenous infections) is detected easily and early. A regular audit of patients with
nosocomial infections only may be useful, as the combination of secondary
endogenous and exogenous infections may highlight a transmission problem in
the ICU.
References
1. Spencer RC (1996) Definitions of nosocomial infections: surveillance of nosocomial
infections. Baillieres Clin Infect Dis 3:237252
2. van Saene HKF, Damjanovic V, Murray AE et al (1996) How to classify infections in
intensive care unitsthe carrier state, a criterion whose time has come? J Hosp Infect
33:112
3. Park DR (2005) The microbiology of ventilator associated pneumonia. Respir Care
50:742763
4. Garner JS, Jarvis WR, Emori TG et al (1988) CDC definitions for nosocomial infections,
1988. Am J Infect Control 16:128140
5. Vincent J-L, Bihari DJ, Suter PM et al (1995) The prevalence of nosocomial infection in
intensive care units in Europe. JAMA 274:639644
6. McGowan JE, Barnes MW, Finland M (1975) Bacteremia at Boston city hospital: occurrence
and mortality during 12 selected years (19651972), with special reference to hospital-
acquired cases. J Infect Dis 132:316335
7. Potgieter PD, Hammond JMJ (1992) Etiology and diagnosis of pneumonia requiring
ICU-admission. Chest 101:199203
50 L. Silvestri et al.
29. Murray AE, Chambers JJ, van Saene HKF (1998) Infections in patients requiring ventilation
in intensive care: application of a new classification. Clin Microb Infect 4:94102
30. Silvestri L, Monti-Bragadin C, Milanese M et al (1999) Are most ICU infections really
nosocomial? a prospective observational cohort study in mechanically ventilated patients.
J Hosp Infect 42:125133
31. Petros AJ, OConnell M, Roberts C et al (2001) Systemic antibiotics fail to clear
multiresistant Klebsiella from a pediatric intensive care unit. Chest 119:862866
32. de la Cal MA, Cerda E, Garcia-Hierro P et al (2001) Pneumonia in severe burns: a
classification according to the concept of the carrier state. Chest 119:11601165
33. Silvestri L, Sarginson RE, Hughes J et al (2002) Most nosocomial pneumonias are not due to
nosocomial bacteria in ventilated patients: prospective evaluation of the accuracy of the 48 h
time cut-off using carriage as the gold standard. Anaesth Intensive Care 30:275282
34. Sarginson R, 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
32:839847
35. Silvestri L, Petros AJ, Sarginson RE et al (2005) Handwashing in the intensive care unit: a
big measure-with modest effects. J Hosp Infect 59:175179
Gut Microbiology: Surveillance
Samples for Detecting the Abnormal 5
Carrier State in Overgrowth
5.1 Introduction
Critical illness impacts all organ systems, such as lungs, heart and gut. The gut also
includes the vast living microbial tissue of the indigenous, mainly anaerobic, flora.
This enormous bacterial tissue is embedded in the mucous layer and covers the
inner wall of the gut. Amongst the aerobic Gram-negative bacilli (AGNB), only the
indigenous Escherichia coli is carried by healthy people in the gut. Critical illness
converts the normal carrier state of E. coli into carriage of abnormal AGNB,
including Klebsiella, Enterobacter and Pseudomonas species [1], and methicillin-
resistant Staphylococcus aureus (MRSA) [2]. It is hypothesised that receptors for
AGNB and MRSA are constitutively expressed on the mucosal lining but are
covered by a protective layer of fibronectin in the healthy mucosa. Significantly
increased levels of salivary elastase have been shown to precede AGNB carriage in
the oropharynx in post-operative patients and the elderly [3, 4]. It is probable that in
individuals with both acute and chronic underlying illness, activated macrophages
release elastase into mucosal secretions, thereby denuding the protective fibronectin
layer. It is thought that this possible mechanism is a deleterious consequence of
the inflammatory response encountered during and after illness. Critical illness
profoundly impacts body flora in two ways: it induces qualitative changes from
normal to abnormal flora [1, 2], as well as quantitative changes from low- to
high-grade carriage or gut overgrowth defined as C2+ or C105 potential pathogens
per millilitre of saliva and/or gram of faeces [5]. Overgrowth concentrations of both
normal and abnormal flora in surveillance samples are frequently found on
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 53
DOI: 10.1007/978-88-470-1601-9_5, Springer-Verlag Italia 2012
54 H. K. F. van Saene et al.
admission to the intensive care unit (ICU), accounting for the large percentage of
primary endogenous infections [6, 7] (Table 4.1). Abnormal flora is often acquired
during treatment in the ICU. Acquisition invariably leads to abnormal carriage due
to critical illness. Most iatrogenic interventions in the patient requiring intensive
care, including mechanical ventilation, promote quantitative changes from
low- to high-grade carriage or overgrowth. Gut protection using H2 antagonists and
antimicrobials are commonly applied in the critically ill. H2 antagonists increase
gastric pH, thereby impairing the gastric acidity barrier [8]. Antimicrobials that are
active against the indigenous, mainly anaerobic, flora, which are excreted via
bile into the gut, may disturb gut ecology [9]. Integrity of both physiology and flora
is essential for the individuals defence against AGNB carriage. Impairment of
these two factors promotes overgrowth of abnormal, potentially pathogenic,
microorganisms (PPM), such as AGNB in concentrations of C2+ or C105 colony
forming units (CFU) per millilitre or gram of faeces.
Gut overgrowth of abnormal flora is not only a marker of critical illness, but it
harms the patient, as it is a disease in itself. In addition, gut overgrowth of
abnormal flora has a major epidemiological impact on the other patients in the ICU
as well as on the ICU environment.
The higher the salivary and faecal concentrations of AGNB and MRSA, the higher
the possibility of PPM transmission via carers hands [14, 15]. PPM acquisition
invariably leads to carriage, as the critically ill are unable to clear the acquired
AGNB and MRSA. Carriers of abnormal bacteria in overgrowth shed these
microorganisms into the environment and determine the contamination level of the
inanimate environment, including beds, tables, telephones and floors [16, 17].
5 Gut Microbiology 55
5.4 Definitions
Surveillance samples are defined as samples obtained from body sites where PPM
may potentially be carried, i.e. the digestive tract, comprising oropharyngeal and
rectal cavities [18]. Surveillance cultures should be distinguished from surface and
diagnostic samples.
Surface samples are taken from the skin, such as axilla, groin and umbilicus, and from
the nose, eye and ear. They do not belong to a surveillance sampling protocol because
positive surface swabs merely reflect the oropharyngeal and rectal carrier states.
Diagnostic samples are from internal organs that are normally sterile, such as lower
airways, blood, bladder, and skin lesions. They are only taken on clinical indication.
The endpoint of diagnostic samples is clinical, as they aim to prove microbiologically
a clinical diagnosis of inflammation, both generalised and/or local.
Only the most critically ill patients require intensive microbiological monitoring
using surveillance samples to detect the abnormal carrier state of AGNB and
MRSA in overgrowth concentrations. Due to the severity of their illness, these
patients require intensive care, including mechanical ventilation, for a minimum of
56 H. K. F. van Saene et al.
A surveillance programme for this type of patient includes samples from both
oropharynx and gut. Potential pathogens carried in the throat and gut cause pneu-
monia and septicaemia, respectively [19]. These two serious infections are respon-
sible for a high rate of mortality. Potential pathogens present in overgrowth in the
throat and gut are implicated in transmission via the hands of carers, in particular, in
outbreak situations. A throat and rectal swab are taken to detect oropharyngeal
and gut carriage of AGNB and MRSA. Rectal swabs must be coated with stool.
As MRSA has an affinity for the skin, skin is sampled only if lesions are present.
5.6.3 When?
Surveillance cultures allow the intensive care specialist to distinguish normal from
abnormal carrier state, overgrowth from low-level carriage and endogenous from
exogenous infections in combination with diagnostic samples.
5 Gut Microbiology 57
1*
1st quadrant
+1: > 103 CFU/g or ml
2*
3rd quadrant
+3: > 107 CFU/g or ml
Knowledge of the carrier state at the time of admission (primary carriage) and
subsequently during treatment in the ICU (secondary or super carriage) is crucial
for managing infection in the unit.
Table 5.2 Carriage classification of severe infections of lower airways and blood
bacteria in only 5% of these patients [23]. A recent study from Trieste [7] reports
resistance figures of 8% in diagnostic cultures compared with 22% in surveillance
cultures. This difference is highly likely to be due to the observation that overgrowth
in the throat and gut is necessary to make diagnostic cultures positive [24].
However, information provided by surveillance cultures of throat and rectum
enables the intensivist to implement isolation and to reinforce hygiene measures
as soon as possible following admission. Two recent studies show that MRSA
and ceftazidime-resistant AGNB were identified in 23.8% and 52.1% of patients,
respectively, within the first 72 h of admission to the ICU [6, 25].
Recent studies using surveillance cultures of throat and rectum to detect carrier
state demonstrate that only infections occurring after 1 week of ICU stay are
due to microbes transmitted via the hands of health-care workers [2630]. The
incidence varies between 15% and 45% depending on illness severity. Microor-
ganisms related to the ICU environment are first acquired in the oropharynx. In the
critically ill, oropharyngeal acquisition invariably leads to secondary carriage.
The subsequent build up to digestive tract overgrowth, which can then result in
colonisation of normally sterile internal organs, takes a few days. Finally, it is
the degree of immunosuppression of the ICU patient that determines the day of
colonization, leading to an established secondary endogenous or super infection.
The other type of ICU infection is the exogenous infection [3133] due to breaches
of hygiene. Causative bacteria are also acquired in the unit but are never present in
patient throat and/or gut flora. For example, long-stay patients, particularly those
who receive a tracheostomy in respiratory units, are at high risk of exogenous
lower-airway infections. Purulent lower-airway secretions yield a microorganism
60 H. K. F. van Saene et al.
that has never been previously carried by the patient in the digestive tract flora, or
indeed in their oropharynx. Although both the tracheostomy and the oropharynx
are equally accessible for bacterial entry, the tracheotomy tends to be the entry site
for bacteria that colonise/infect the lower airways.
However, primary endogenous infections cause the major infection problems,
and the microorganisms involved do not bear any relation to ICU ecology
[34, 35]. A recent study compared the traditional 48 h cutoff and the criterion
of the carrier state and found that the time cutoff significantly overestimated the
magnitude of the nosocomial problem [30]. This approach to the carrier state
may be more useful for interhospital comparison, as only infections due to
microorganisms acquired in the different units are compared, independent of
illness severity.
In identifying the right population with primary endogenous infections,
classification using the carrier state avoids blaming staff for all infections
occurring after 48 h for which they are not responsible. Knowledge of carrier
status thus prevents fruitless investigation of apparent cross-infection episodes.
Secondly, without surveillance samples, exogenous infections are impossible to
recognise, at least at an early stage when only diagnostic samples such as
tracheal aspirate, urine and blood have been tested. Finally, knowledge of the
carrier state using surveillance cultures on admission and twice weekly is an
effective strategy for early identification of carriers of multidrug-resistant
microorganismsincluding AGNB such as A. baumannii [7, 36], MRSA [6, 24]
and vancomycin-resistant enterococci [37]both on admission and during ICU
stay. Surveillance cultures, in particular of the oropharynx, that become positive
for a PPM during ICU stay reveal ongoing transmission and an impending outbreak
long before the diagnostic samples yield the outbreak strain [38]. This surveillance
strategy optimises targeted infection control interventionsincluding (1) hand
hygiene, (2) isolation, (3) personal protective equipment and (4) care of patient
equipmentto control transmission from one patient-carrier to another patient via
carers hands.
Most infection surveillance programmes include all patients admitted to the ICU,
whether they stay a few days or 2 weeks [39, 40]. Including a large number of
relatively short-stay patients with a low risk of infection tends to dilute total rates
of infection by increasing the size of the denominator. However, whereas low
percentages look good to the hospital manager, they do not allow room for
improvement, i.e. detecting a significant reduction in infection rate following the
introduction of an intervention [39]. We believe that critically ill patients benefit
from a surveillance programme of both infection and of carriage [41, 42], in
particular, in combination with selective decontamination of the digestive tract
(SDD) [4345].
5 Gut Microbiology 61
References
1. Johanson WG, Pierce AK, Sanford JP (1969) Changing pharyngeal bacterial flora of
hospitalized patients. Emergence of Gram-negative bacilli. New Engl J Med 281:11371140
2. Chang FY, Singh N, Gayowski T et al (1998) Staphylococcus aureus nasal colonization in
patients with cirrhosis: prospective assessment of association with infection. Infect Control
Hosp Epidemiol 19:328332
3. Dal Nogare AR, Toews GB, Pierce AK (1987) Increased salivary elastase precedes Gram-
negative bacillary colonization in post-operative patients. Am Rev Respir Dis 135:671675
4. Palmer LB, Albulak K, Fields S et al (2001) Oral clearance and pathogenic oropharyngeal
colonization in the elderly. Am J Respir Crit Care Med 164:464468
5. van Saene HKF, Damjanovic V, Murray AE, de la Cal MA (1996) How to classify infections in
intensive care unitsthe carrier state, a criterion whose time has come? J Hosp Infect 33:112
6. Viviani M, van Saene HKF, Dezzoni R et al (2005) Control of imported and acquired
methicillin-resistant Staphylococcus aureus [MRSA] in mechanically ventilated patients: a
dose response study of oral vancomycin to reduce absolute carriage and infection. Anaesth
Intensive Care 33:361372
7. Viviani M, van Saene HK, Pisa F et al (2010) The role of admission surveillance cultures in
patients requiring prolonged mechanical ventilation in the intensive care unit. Anaesth
Intensive Care 38:325335
8. Hillman KM, Riordan T, OFarrell SM, Tabaqchali S (1982) Colonization of the gastric
contents in critically ill patients. Crit Care Med 10:444447
9. Vollaard EJ, Clasener HAL (1994) Colonization resistance. Antimicrob Agents Chemother
38:409414
10. Deitch EA, Xu DZ, Qi L, Berg RD (1993) Bacterial translocation from the gut impairs
systemic immunity. Surgery 104:269276
11. Baue AE (1993) The role of the gut in the development of multiple organ dysfunction in
cardiothoracic patients. Ann Thoracic Surg 55:822829
12. van Uffelen R, van Saene HKF, Fidler V et al (1984) Oropharyngeal flora as a source of
bacteria colonizing the lower airways in patients on artificial ventilation. Intensive Care Med
10:233237
13. van Saene HKF, Taylor N, Damjanovic V, Sarginson RE (2008) Microbial gut overgrowth
guarantees increased spontaneous mutation leading to polyclonality and antibiotic resistance
in the critically ill. Curr Drug Targ 9:419421
14. Riley TV, Webb SAR, Cadwallader H et al (1996) Outbreak of gentamicin-resistant
Acinetobacter baumanii in an intensive care unit: clinical, epidemiological and micro-
biological features. Pathology 28:359363
15. Lin MY, Hayden MK (2010) Methicillin-resistant Staphylococcus aureus and vancomycin-
resistant enterococcus: recognition and prevention in intensive care units. Crit Care Med
38(suppl 8):S335S344
16. Go ES, Urban C, Burns J et al (1994) Clinical and molecular epidemiology of Acinetobacter
infections sensitive only to polymyxin B and sulbactam. Lancet 344:13291332
17. Bocher S, Skov RL, Knudsen MA et al (2010) The search and destroy strategy prevents
spread and long-term carriage of MRSA: results from follow-up screening of a large ST22
(E-MRSA) 15 outbreak in Denmark. Clin Microb Infect 16:14271434
18. Damjanovic V, van Saene HKF, Weindling AM (1994) The multiple value of surveillance
cultures: an alternative view. J Hosp Infect 28:7178
19. Silvestri L, van Saene HKF, Zandstra DF et al (2010) Selective decontamination of the
digestive tract reduces multiple organ failure and mortality in critically ill patients:
systematic review of randomized controlled trials. Crit Care Med 38:13701376
20. van Saene HKF, Damjanovic V, Alcock SR (2001) Basics in microbiology for the patient
requiring intensive care. Curr Anaesth Crit Care 12:617
62 H. K. F. van Saene et al.
21. Mobbs KJ, van Saene HKF, Sunderland D, Davies PDO (1999) Oropharyngeal Gram-
negative bacillary carriage in chronic obstructive pulmonary disease: relation to severity of
disease. Respir Med 93:540545
22. Silvestri L, Petros AJ, Sarginson RE et al (2005) Handwashing in the intensive care unit: a
big measure with modest effects. J Hosp Infect 59:172179
23. DAgata EM, Venkataraman L, DeGirolami P et al (1999) Colonization with broad-spectrum
cephalosporin-resistant gram-negative bacilli in intensive care units during a nonoutbreak
period: prevalence, risk factors, and rate of infection. Crit Care Med 27:10901095
24. de la Cal MA, Cerda E, van Saene HKF et al (2004) Effectiveness and safety of enteral
vancomycin to control endemicity of methicillin-resistant Staphylococcus aureus in a
medical/surgical intensive care unit. J Hosp Infect 56:175183
25. Toltzis P, Yamashita T, Vilt L et al (1997) Colonization with antibiotic-resistant Gram-
negative organisms in a pediatric intensive care unit. Crit Care Med 25:538544
26. Murray AE, Chambers JJ, van Saene HKF (1998) Infections in patients requiring ventilation
in intensive care: application of a new classification. Clin Microbiol Infect 4:94102
27. Silvestri L, Monti Bragadin C, Milanese M et al (1999) Are most ICU-infections really
nosocomial? A prospective observational cohort study in mechanically ventilated patients.
J Hosp Infect 42:125133
28. Petros AJ, OConnell M, Roberts C et al (2001) Systemic antibiotics fail to clear multi-
drug-resistant Klebsiella from a pediatric ICU. Chest 119:862866
29. de la Cal MA, Cerda E, Garcia-Hierro P et al (2001) Pneumonia in patients with severe burns.
A classification according to the concept of the carrier state. Chest 119:11601165
30. Silvestri L, Sarginson RE, Hughes J et al (2002) Most nosocomial pneumonias are not due to
nosocomial bacteria in ventilated patients. Evaluation of the accuracy of the 48 h time cut-off
using carriage as the gold standard. Anaesth Intensive Care 30:275282
31. Hammond JMJ, Potgieter PD, Saunders GL et al (1992) Double blind study of selective
decontamination of the digestive tract in intensive care. Lancet 340:59
32. Morar P, Singh V, Makura Z et al (2002) Differing pathways of lower airway colonization
and infection according to mode of ventilation (endotracheal versus tracheostomy). Arch
Otolaryngol Head Neck Surg 128:10611066
33. Morar P, Makura Z, Jones AS et al (2000) Topical antibiotics on tracheostoma prevents
exogenous colonization and infection of lower airways in children. Chest 117:513518
34. Stoutenbeek CP (1989) The role of systemic antibiotic prophylaxis in infection prevention in
intensive care by SDD. Infection 17:418421
35. Sirvent JM, Torres A, El-Ebiary M et al (1997) Protective effect of intravenously
administered cefuroxime against nosocomial pneumonia in patients with structural coma.
Am J Respir Crit Care Med 155:17291734
36. Corbella X, Pujol M, Ayats J et al (1996) Relevance of digestive tract colonization in
the epidemiology of nosocomial infections due to multiresistant Acinetobacter baumannii.
Clin Infect Dis 23:329334
37. Hendrix CW, Hammond JMJ, Swoboda SM et al (2001) Surveillance strategies and impact of
vancomycin-resistant enterococcal colonization and infection in critically ill patients. Ann
Surg 233:259265
38. Chetchotisakd P, Phelps CL, Hartstein AI (1994) Assessment of bacterial cross-transmission
as a cause of infections in patients in intensive care units. Clin Infect Dis 18:929937
39. Kollef MH, Sherman G, Ward S, Fraser VJ (1999) Inadequate antimicrobial treatment of
infections. Chest 115:462474
40. Richards MJ, Edwards JR, Culver DH et al (1999) Nosocomial infections in medical
intensive care units in the United States. Crit Care Med 27:887892
41. Langer M, Carretto E, Haeusler EA (2001) Infection control in ICU: back (forward) to
surveillance samples? Intensive Care Med 27:15611563
42. Silvestri L, van Saene HKF (2002) Surveillance of carriage. Minerva Anestesiol 68(Suppl 1):
S179S182
5 Gut Microbiology 63
43. de Jonge E, Schultz MJ, Spanjaard L et al (2003) Effects of selective decontamination of the
digestive tract on mortality and acquisition of resistant bacteria in intensive care: a
randomised controlled trial. Lancet 362:10111016
44. de Smet AM, Kluytmans JA, Cooper BS et al (2009) Decontamination of the digestive tract
and oropharynx in ICU patients. N Engl J Med 360:2031
45. Liberati A, DAmico R, Pifferi S et al (2009) Antibiotic prophylaxis to reduce respiratory
tract infections and mortality in adults receiving intensive care. Cochrane Database Syst Rev
CD000022
Part II
Antimicrobials
Systemic Antibiotics
6
A. R. De Gaudio, S. Rinaldi and C. Adembri
6.1 Introduction
Systemic antibiotics remain the main causative therapy for critically ill patients
with infection. This chapter aims to provide a clinical review of the antibiotics
available for systemic administration in the intensive care unit (ICU). Pharma-
cological and microbiological factors that affect antimicrobial administration
regimen in the critical patient are also discussed.
As new drugs have been developed to overcome antimicrobial resistance,
bacteria acquired new kinds of resistance in the endless war for survival. The
opportunity of using appropriate pharmacokinetic/pharmacodynamic (PK/PD)
parameters to optimize dosing in order not only to cure patients but also to reduce
the spreading of resistance is therefore becoming imperative, as there are now
fewer new therapeutic options than in the past.
Bacteria have the capacity to adapt to a wide range of conditions. Any
strategy aimed at destroying bacterial flora has resulted in a dramatic failure. It is
likely that in the future, the definitive answer to infectious diseases will stand not
only on the development of new antibiotics but also in immunomodulating
strategies. The goal of the intensivist challenged with an infectious disease will
include turning the relationship between bacterial flora and the host from
infection back to symbiosis.
C. Adembri (&)
Department of Medical and Surgical Critical Care,
Section of Anesthesiology and Intensive Care, University of Florence,
Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 67
DOI: 10.1007/978-88-470-1601-9_6, Springer-Verlag Italia 2012
68 A. R. De Gaudio et al.
Antimicrobial activity is the result of many variables that fit together in the clinical
setting just as in a puzzle (Table 6.1), as it depends on the PD properties of the
antibiotic, the specific bacterial strain, the infection site, and the setting in which
the antimicrobial drug challenges the microorganism [13]. In the ICU, infections
are often caused by multiresistant strains and develop in patients who have
coexistent multiple organ dysfunction and impaired immune function, a clinical
setting in which optimal antimicrobial therapy (in terms of both activity spectrum
and administration dose and modality) is essential to improve outcome.
One of the main goals of antimicrobial therapy is that the antimicrobial reaches
and remains in the site of infection in a sufficient concentration and for a sufficient
time. Regarding the pharmacological action of antimicrobials (that is, killing
activity), this may be concentration dependent or time dependent [4]. Concen-
tration-dependent antibiotics kill bacteria at a greater rate and to a greater extent
with increasing antibiotic concentrations, whereas time-dependent antibiotics kill
bacteria at the same rate and to the same extent once an appropriate concentration
threshold has been achieved. Aminoglycosides, fluoroquinolones, clarithromycin,
azalides, ketolides, and metronidazole are considered concentration-dependent
antimicrobial drugs, whereas glycopeptides, clindamycin, natural macrolides,
b-lactams, linezolid, and quinupristin-dalfopristin are considered time-dependent
drugs. These differences in PD activity should result in different dosing regimens
for time- and concentration-dependent antibiotics.
Besides these PD considerations, the bactericidal activity of antimicrobial agent
is influenced by several factors correlated with infection site, including antibiotic
diffusion, local pH, bacterial load, phase of bacterial growth, and oxygen tension.
In recent years, antibiotic penetration into the infection site has become one of the
main factors that should be taken into account when choosing an antibiotic regi-
men. Linezolid and fluoroquinolones penetrate well into the lungs, and their use in
nosocomial pneumonia is associated with a significant clinical success rate.
Vancomycin has a poor lung penetration, and many clinical failures of this drug in
pneumonia due to strains sensitive to this antimicrobial may derive from its poor
lung disposition.
Several studies regarding the inoculum effect considered the effect of the bacterial
load on the minimal inhibitory concentration (MIC) of several antibiotics. Standard
laboratory inoculum for MIC determination is around 105 CFU/ml. The presence of
the inoculum effect is defined as an eightfold or greater increase in MIC on testing,
with an inoculum as high as 107108 CFU/ml, with higher inoculum better reflecting
most clinical settings. Aztreonam, piperacillin, cefotaxime, and cefoxitin show a
significant inoculum effect against susceptible strains. A significant inoculum effect
is associated with b-lactams with prevalent inhibition of penicillin-binding protein
3(PBP3) because, in the setting of a high inoculum, bacteria proliferation slows and
they produce less PBP3. Moreover, the inoculum effect is present when a bacterium
produces an enzyme able to destroy the tested antibiotic, because the enzymes
6 Systemic Antibiotics 69
released from the dead cells inactivate the antibiotic, which is why the combination
with b-lactamase inhibitors may prevent the inoculum effect observed with many
b-lactams. Among the clinical conditions mimicking a high inoculum are endocar-
ditis, meningitis, septic arthritis, osteomyelitis, abscesses, and deep-seated infec-
tions. In these conditions, antibiotics without an inoculum effect have been
demonstrated to be more effective.
Regarding the bactericidal activity of antibiotics, another important factor is the
presence or absence of a postantibiotic effect (PAE). PAE is the ability of an
antimicrobial drug to exert a persistent inhibitory effect on microorganism growth
after the drug has been completely removed. Usually, antibiotics with a main time-
dependent activity lack a significant PAE, although differences among the different
classes do exist. b-lactams, for example, show a significant PAE only against
Gram-positive organisms, although carbapenems may have a sustained PAE
against aerobic Gram-negative bacilli (AGNB). Agents that interfere with protein
or DNA synthesis, such as fluoroquinolones and aminoglycosides, usually show a
sustained PAE, mainly against AGNB. The PAE is unique to the pathogen and is
generally longer in vivo than in vitro. For example, PAE duration for aminogly-
cosides ranges from 0.5 to 8 h depending on the bacterial strain, the MIC,
the duration of exposure, and the relative concentration of the aminoglycoside.
The PAE may decrease with multiple dosing [13].
6.3.1 Aminoglycosides
6.3.2 b-Lactams
6.3.3 Glycopeptides
6.3.4 Fluoroquinolones
6.3.5 Linezolid
Linezolid is quite lipophilic, and it distributes widely into tissues and is mostly
metabolized hepatically before being cleared renally. No dose adjustment seems to
be necessary in renal or hepatic dysfunction. The killing characteristic is time
dependent, being optimal when the time above MIC is [4080%. In critical
patients, linezolid shows an increased Vd and CL, requiring appropriate dose
adjustment.
6.3.6 Tigecycline
6.3.7 Daptomycin
6.3.8 Colistin
carbapenems, and that are not inhibited by clavulanic acid. They can be either
chromosomal or plasmid encoded. Group 2 includes b-lactamases susceptible to
inhibition due to clavulanic acid, and they are divided in six subgroups. Subgroup
2a includes staphylococcal and enterococcal penicillinases. Subgroup 2b includes
broad spectrum b-lactamases of AGNB strains but also extended spectrum
b-lactamases. Subgroup 2c includes carbenicillin hydrolyzing b-lactamases.
Subgroup 2d includes cloxacillin hydrolyzing enzymes. Subgroup 2e includes
cephalosporinases. Subgroup 2f includes b-lactamases active on carbapenems.
Group 3 includes metallo-beta-lactamases with activity against carbapenems and
all b-lactam classes except monobactams. Group 4 includes the other b-lactamases
not included in the previous groups [12].
Extended spectrum b-lactamases (ESBL) are b-lactamase able to hydrolyze
the extended-spectrum cephalosporins, including third-generation compounds.
Antibiotics susceptible to ESBL include cefotaxime, ceftazidime, aztreonam,
and other expanded-spectrum cephalosporins, whereas imipenem is usually
resistant. These enzymes are encoded also in plasmids containing genes for
aminoglycoside resistance, trimethoprimsulfamethoxazole, and often fluoro-
quinolones. ESBL are often present in Escherichia coli or Klebsiella
pneumoniae but can be transferred to Proteus mirabilis, Citrobacter, Serratia,
and other AGNB [12]. Every E. coli and K. pneumoniae with reduced sus-
ceptibility to these drugs or to aztreonam should be considered at risk of
possessing these enzymes. Treating infections caused by ESBL-producing
strains is difficult due to the high risk of concomitant resistance to aminogly-
cosides, trimethoprimsulfamethoxazole, and fluoroquinolones. Carbapenems
are the agents of choice against ESBL-producing bacteria because they are
highly stable against b-lactamase hydrolysis [12].
Inactivation is by far the most important mechanism of acquired microbial
resistance toward aminoglycosides in clinical practice. Aminoglycosides may
become the substrate of several microbial enzymes that may phosphorylate,
adenylate, or acetylate specific hydroxyl or amino groups. Phosphorylation is the
main mechanism of inactivation for aminoglycosides, it is determined by either
Gram-positive or AGNB strains, and it results in complete inactivation of
aminoglycosides. Some aminoglycosides are resistant to phosphorylation due to
the presence of side chains, such as amikacin, or the absence of specific hydroxyl
groups, such as tobramycin and gentamicin. Aminoglycoside-inactivating enzymes
are often encoded in plasmids that are especially common in hospital environ-
ments. Pharmacological strategies to inhibit aminoglycoside-inactivating enzymes
are under evaluation.
may increase the release of endotoxins that trigger the septic response by
binding to specific receptors upon mononuclear phagocytes, endothelial cells,
and polymorphonuclear leucocytes [1417]. This effect has been studied for
several agents, including b-lactams, glycopeptides, aminoglycosides, and
fluoroquinolones.
Regarding b-lactams, different patterns of endotoxin release seem to be cor-
related with the inhibition of different PBPs. In particular, inhibition of PBP1a and
1b results in a massive bactericidal activity against AGNB associated with
bacterial wall molecule degradation. These changes lead to production of
spheroplasts that undergo immediate bacteriolysis. In AGNB, PBP2 inhibition
results in spheroid elements osmotically stable but unable to proliferate, and thus
increases bacterial mass. Conversely, inhibition of PBP3 located in bacterial septa
prevents separation of the proliferating bacteria, resulting in long filaments that
are vital syncytia resistant to lysis but able to produce and release endotoxins.
Carbapenems, ceftriaxone, cefepime, and b-lactams in combination with
b-lactamase inhibitors inhibit PBP2 and 3 in AGNB. Therefore, they have a rapid
bactericidal activity without increments of bacterial mass and are associated with
poor endotoxin release. Ceftazidime, ticarcillin, and cefoxitin inhibit PBP3 and, at
high concentrations, PBP1; they have a slower bactericidal activity associated with
a slight increase in bacterial mass and a moderate release of endotoxins. Pipera-
cillin, monobactams, cefuroxime, and cefotaxime inhibit mainly PBP3, resulting in
slow bactericidal activity associated with an increase in bacterial mass and
endotoxin release [17].
Glycopeptides induce bacteriolysis and release endotoxins, but the lipid part
of the endotoxin is inactivated by these antibiotics. Fluoroquinolones result in a
slightly greater endotoxin release than do carbapenems, as they cause DNA
damage associated with the inhibition of cellular division that produces bac-
terial filaments able to release endotoxins. Besides the role of antibiotic therapy
in endotoxin release, they may have also endotoxin-neutralizing properties that
may inhibit the effects of endotoxin [18]. Antibiotics with endotoxin-neutral-
izing properties are polycationic molecules that can link the polyanionic moi-
eties of lipopolysaccharides, including polymyxins, teicoplanin, and
aminoglycosides [14, 15]. In experimental studies, polymyxin B resulted in
reduced endotoxin shock, pyrogenicity, Shwartzman reaction, and endotoxin
lethality [15]. Teicoplanin shows an endotoxin-neutralizing property that results
in reduced levels of tumor necrosis factor (TNF)-a and interleukin (IL)-1 after
endotoxin stimulation.
Antibiotics inhibiting protein synthesis result in a lower release of endotoxins
than the agents acting on the cell wall. Aminoglycosides inhibit endotoxin syn-
thesis, and although they favor endotoxin release, they neutralize them chemically
by binding their anionic moieties. In this regard, variable effects have been
observed for different compounds of this class of antibiotics: tobramycin seems to
be the most effective in neutralizing endotoxins.
6 Systemic Antibiotics 77
The relationship between the microbes and host is not just a fight between two
contenders [19]. Ecological studies show symbiosis in the microbial communities
of the skin and mucous membranes. In these ecosystems, the resident microor-
ganisms play an important physiological role with mutualistic relationships,
protection from infections included [20, 21]. The normal microflora acts as a
barrier against carriage of potentially pathogenic microorganisms and against
overgrowth of opportunistic microorganisms (colonization resistance) [21, 22].
For this protective effect to occur, great stability of these ecosystems is funda-
mental [22]. Administration of antimicrobial agents interferes with the ecological
balance between host and normal microflora.
The impact of antibiotics on normal intestinal flora has been widely studied.
The gastrointestinal tract is a complex ecosystem that may be altered by
antibiotic administration, resulting clinically in diarrhea and fungal infections
that usually cease after treatment. Pseudomembranous colitis is an example of
the deleterious effects of antimicrobial therapy that disturbs the normal ecologic
balance of the bowel flora, leading to an abnormal proliferation of Clostridium
difficile. The effects of antibiotics on oropharyngeal and skin microflora may be
important as well. Antibiotic therapy may cause alterations in skin microbial
flora with a decline in the number of bacteriamostly anaerobic bacteria [23].
Moreover, exposure to antibiotics often results in selection of antibiotic-
resistant organisms that can transfer resistance to other microorganisms. By
using antimicrobial agents that do not alter the level of resistance of colonizing
microorganisms, the risk of emergence and spread of resistant strains is reduced
[20, 21].
Respect for the individuals ecology should be a prerequisite for every antibi-
otic policy, and the use of antibiotics with little or no impact on the normal flora
should always be encouraged. Antibiotics that are not active against the indigenous
bacteria in the mouth and intestine and are not excreted to a significant degree via
the intestine, saliva, or skin are therefore preferred [20, 21]. First generation
b-lactams, aminoglycosides, and polymyxins are favorable from an ecological
point of view.
Systemic antibiotics include drugs commonly used in the ICU, such as b-lactams;
drugs to be used under specific indications, such as aminoglycosides, glycopeptides,
fluoroquinolones, and macrolides; and drugs used only for multi-drug-resistant
microorganisms, including streptogramins, linezolid, colistin, advanced-generation
cephalosporins, glycylcycline, and daptomycin (Table 6.2).
6
Table 6.2 Main systemic antibiotic used in ICU and their spectrum of antimicrobial activity
6.11.1 b-Lactams
6.11.2 Penicillin G
6.11.7 Cephalosporins
Staphylococcus aureus. Its most common side effects are nausea and dysgeusia.
Ceftaroline is a novel, broad-spectrum, with antimicrobial activity against
MRSA, Streptococcus pneumonia and respiratory Gram-negative pathogens
such as Moraxella catarrhalis and H. influenzae. It is eliminated primarily by
renal excretion, with a T1/2 of approximately 3 h [29].
b-lactamase inhibitors bind to b-lactamases and inactivate them. They are mostly
active against plasmid-encoded b-lactamases, including the extended-spectrum
ESBL, but are inactive against type I chromosomal b-lactamases of AGNB.
b-lactamase inhibitors include clavulanic acid, sulbactam, and tazobactam [12].
Clavulanic acid has been combined with amoxicillin and ticarcillin with extension
of the antimicrobial activity to b-lactamase-producing strains of staphylococci,
H. influenzae, gonococci, and E. coli. Sulbactam is usually combined with ampi-
cillin; tazobactam has been combined with piperacillin. Although this combination
extends its spectrum of activity, it does not enhance that against P. aeruginosa, as
resistance is due to either chromosomal b-lactamases or decreased permeability of
piperacillin into the periplasmic space due to either porin protein OprD loss or
multidrug efflux system upregulation.
These drugs are used primarily to treat infections caused by AGNB [31].
6.11.12 Fluoroquinolones
6.11.13 Macrolides
6.11.16 Oxazolidinones
Linezolid is the first member (and at present the only available molecule) of a new
class of antibiotics known as oxazolidinones [44, 45].
These drugs are available in an injectable combination in a ratio of 70:30 [46, 47].
References
1. Gunderson BW, Ross GH, Ibrahim KH, Rotschafer JC (2001) What do we really know about
antibiotic pharmacodynamics? Pharmacotherapy 21:302s318s
2. Mehrota R, De Gaudio R, Palazzo M (2004) Antibiotic pharmacokinetic and
pharmacodynamic considerations in critical illness. Int Care Med 30:21452156
3. Kollef MH (2001) Optimizing antibiotic therapy in the intensive care unit setting. Crit Care
5:189195
4. Ambrose PG, Bhavnani SM, Rubino CM et al (2007) Pharmacokinetics-pharmacodynamics
of antimicrobial therapy: its not just for mice anymore. Clin Infect Dis 44:7986
5. Pea F, Viale P (2009) Bench-to-bedside review: appropriate antibiotic therapy in severe
sepsis and septic shockdoes the dose matter? Crit Care 13:214
6. Kumar A (2009) Optimizing antimicrobial therapy in sepsis and septic shock. Crit Care Clin
25:733751
7. Roberts JA, Lipman J (2009) Pharmacokinetic issues for antibiotics in the critically ill
patient. Crit Care Med 37:840851
8. Figueiredo Costa S (2008) Impact of antimicrobial resistance on the treatment and outcome
of patients with sepsis. Shock 30:2329
9. Lipman J, Boots R (2009) A new paradigm for treating infections: go hard and go home.
Crit Care Resusc 11:276281
10. Lodise TP Jr, Lomaestro B, Drusano GL (2007) Piperacillin-tazobactam for Pseudomonas
aeruginosa infection: clinical implications of an extended-infusion dosing strategy. Clin
Infect Dis 44(3):357363
11. Barbour A, Scaglione F, Derendorf H (2010) Class-dependent relevance of tissue distribution
in the interpretation of anti-infective pharmacokinetic/pharmacodynamic indices. Int J
Antimicrob Agents 35:431438
96 A. R. De Gaudio et al.
12. Drawz SM, Bonomo RA (2010) Three decades of beta-lactamase inhibitors. Clin Microbiol
Rev 23:160201
13. Novelli A, Mini E, Mazzei T (2004) Pharmacological interactions between antibiotics and
other drugs in the treatment of lower respiratory tract infections. Eur Respir Mon 28:126
14. Nagaoka I, Hirota S, Niyonsaba F et al (2001) Cathelicidin family of antibacterial peptides
CAP18 and CAP11 inhibit the expression of TNF-alpha by blocking the binding of LPS to
CD14(+) cells. J Immunol 167:33293338
15. Zhang L, Dhillon P, Yan H et al (2000) Interactions of bacterial cationic peptide antibiotics
with outer and cytoplasmic membranes of Pseudomonas aeruginosa. Antimicrob Agents
Chemother 44:33173321
16. Tsuzuki H, Tani T, Ueyama H, Kodama M (2001) Lipopolysaccharide: neutralization by
polymyxin B shuts down the signaling pathway of nuclear factor kappa B in peripheral blood
mononuclear cells, even during activation. J Surg Res 100:127134
17. Surez C, Gudiol F (2009) Beta-lactam antibiotics. Enferm Infecc Microbiol Clin
27(2):116129 Article in Spanish
18. Augusto LA, Decottignies P, Synguelakis M et al (2003) Histones: a novel class of
lipopolysaccharide-binding molecules. Biochemistry 42:39293938
19. Sullivan A, Edlund C, Nord CE (2001) Effect of antimicrobial agents on the ecological
balance of human microflora. Lancet Infect Dis 1:101114
20. Klein G (2003) Taxonomy, ecology and antibiotic resistance of enterococci from food and
the gastro-intestinal tract. Int J Food Microbiol 88:123131
21. Tannock GW (2001) Molecular assessment of intestinal microflora. Am J Clin Nutr 73(Suppl
2):410S414S
22. Dunne C (2001) Adaptation of bacteria to the intestinal niche: probiotics and gut disorder.
Inflamm Bowel Dis 7:136145
23. Brook I (2000) The effects of amoxicillin therapy on skin flora in infants. Pediatr Dermatol
17:360363
24. Antonelli M, Mercurio G, Di Nunno S, Recchioni G, Deangelis G (2001) De-escalation
antimicrobial chemotherapy in critically ill patients: pros and cons. J Chemother 1:218223
25. Dogan O, Glmez D, Haselik G (2010) Effect of new breakpoints proposed by Clinical and
Laboratory Standards Institute in 2008 for evaluating penicillin resistance of Streptococcus
pneumoniae in a Turkish University Hospital. Microb Drug Resist 16:3941
26. Feldman C (2004) Clinical relevance of antimicrobial resistance in the management of
pneumococcal community-acquired pneumonia. J Lab Clin Med 143:269283
27. Goosen H (2003) Susceptibility of multi-drug-resistant Pseudomonas aeruginosa in intensive
care units: results from the European MYSTIC study group. Clin Microbiol Infect 9:980983
28. Kahlmeter G (2008) Breakpoints for intravenously used cephalosporins in
EnterobacteriaceaeEUCAST and CLSI breakpoints. Clin Microbiol Infect 14:169174
29. Bazan JA, Martin SI, Kaye KM (2009) Newer beta-lactam antibiotics: doripenem,
ceftobiprole, ceftaroline, and cefepime. Infect Dis Clin North Am 23:983996
30. Baughman RP (2009) The use of carbapenems in the treatment of serious infections.
J Intensive Care Med 24:230241
31. Smith CA, Baker EN (2002) Aminoglycoside antibiotic resistance by enzymatic deactivation.
Curr Drug Targets Infect Disord 2:143160
32. Viale P, Pea F (2003) What is the role of fluoroquinolones in intensive care? J Chemother
15(Suppl 3):510
33. Blondeau JM (2004) Fluoroquinolones: mechanism of action, classification, and development
of resistance. Surv Ophthalmol 49(Suppl 2):S73S78
34. Labro MT (2004) Macrolide antibiotics: current and future uses. Expert Opin Pharmacother
5:541550
35. Ackermann G, Rodloff AC (2003) Drugs of the 21st century: telithromycin (HMR 3647)
the first ketolide. J Antimicrob Chemother 51:497511
36. Malabarba A, Ciabatti R (2001) Glycopeptide derivatives. Curr Med Chem 8:17591773
6 Systemic Antibiotics 97
37. Esposito S, Noviello S (2003) What is the role of glycopeptides in intensive care? J
Chemother 15(Suppl 3):1116
38. Parenti F, Schito GC, Courvalin P (2000) Teicoplanin chemistry and microbiology.
J Chemother 12(Suppl 5):514
39. Harding I, Sorgel F (2000) Comparative pharmacokinetics of teicoplanin and vancomycin.
J Chemother 12(Suppl 5):1520
40. Lundstrom TS, Sobel JD (2000) Antibiotics for Gram-positive bacterial infections.
Vancomycin, teicoplanin, quinupristin/dalfopristin, and linezolid. Infect Dis Clin North
Am 14:463474
41. Rocha JL, Kondo W, Baptista MI et al (2002) Uncommon vancomycin-induced side effects.
Braz J Infect Dis 6:196200
42. Beringer P (2001) The clinical use of colistin in patients with cystic fibrosis. Curr Opin Pulm
Med 7:434440
43. Tsubery H, Ofek I, Cohen S et al (2002) Modulation of the hydrophobic domain of
polymyxin B nonapeptide: effect on outer-membrane permeabilization and
lipopolysaccharide neutralization. Mol Pharmacol 62:10361042
44. Diekema DJ, Jones RN (2001) Oxazolidinone antibiotics. Lancet 358:19751982
45. Paradisi F, Corti G, Messeri D (2001) Antistaphylococcal (MSSA, MRSA, MSSE, MRSE)
antibiotics. Med Clin North Am 85:117
46. De Gaudio AR, Di Filippo A (2003) What is the role of streptogramins in intensive care? J
Chemother 15(Suppl 3):1721
47. Hershberger E, Donabedian S, Konstantinou K, Zervos MJ (2004) Quinupristin-dalfopristin
resistance in Gram-positive bacteria: mechanism of resistance and epidemiology. Clin Infect
Dis 38:9298
48. Devasahayam G, Scheld WM, Hoffman PS (2010) Newer antibacterial drugs for a new
century. Expert Opin Investig Drugs 19:215234
49. Zuckerman JM, Qamar F, Bono BR (2009) Macrolides, ketolides, and glycylcyclines:
azithromycin, clarithromycin, telithromycin, tigecycline. Infect Dis Clin North Am
23:9971026
50. Barton E, MacGowan A (2009) Future treatment options for Gram-positive infections
looking ahead. Clin Microbiol Infect 15:1725
51. Bouza E (2009) New therapeutic choices for infections caused by methicillin-resistant
Staphylococcus aureus. Clin Microbiol Infect 15:4452
Systemic Antifungals
7
C. J. Collins and Th. R. Rogers
7.1 Introduction
C. J. Collins (&)
Clinical Microbiology, Trinity College Dublin, Dublin,
Leinster, Republic of Ireland
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 99
DOI: 10.1007/978-88-470-1601-9_7, Springer-Verlag Italia 2012
100 C. J. Collins and Th. R. Rogers
7.2.1 Posaconazole
had mostly received amphotericin B, the response rates overall were superior for
posaconazole, irrespective of the Aspergillus spp. [6]. In two studies and a case
report including a review of the literature, posaconazole as salvage therapy for
zygomycosis was evaluated with encouraging results [79]. However, there are no
data yet supporting its use as first-line therapy for IA, and this may have to await
the development of an intravenously administered formulation. Posaconazole has
also been investigated as treatment for other mould infections in immunocom-
promised patients, with similarly encouraging results [2, 10].
So far, clinical experience of using posaconazole in ICU patients appears to be
restricted to small case report series [11]. Due to the lack of an intravenously
administered formulation, therapeutic drug monitoring may be required in patients
with invasive fungal disease. Studies to date are limited on its value, but one small
series in critical care patients has shown that serum concentrations [0.5 mg/1 are
more likely to achieve therapeutic success [12].
Caspofungin, anidulafungin and micafungin are now approved for treating inva-
sive Candida infections. In the 2009 IDSA guidelines for managing candidiasis,
echinocandins along with fluconazole received a grade A-I recommendation for
treating candidaemia in non-neutropenic patients, with the expert panel favouring
echinocandins in those with moderate to severe illness or with recent azole
exposure [13]. They also received a grade A-II recommendation for the initial
treatment of candidaemia in neutropenic patients and a grade B-III recommen-
dation for the treatment of C. glabrata. Although head-to-head studies among
the three approved echinocandins are limited [14], overall, there have been no
significant differences found in their clinical efficacy [15], although there are
pharmacokinetic and pharmacodynamic differences [16].
7.2.3 Caspofungin
Caspofungin (Cancidas) was reviewed in the last edition of this series [1]. It was
the first echinocandin to be approved for clinical use in Europe and is the first and
only member of this group to be licensed by the European Medicines Agency
(EMEA) and the FDA for treating patients with invasive Aspergillus infections
refractory to other therapy. It has also been licensed as empirical therapy for
presumed fungal infections in febrile, neutropenic patients and for treating inva-
sive candidiasis. It is also the only echinocandin with both an FDA and EMEA
license for therapeutic use in the paediatric population.
Standard dosing is a 70-mg loading dose followed by 50 mg daily. Dosing in
the paediatric population is based on the patients body surface area (BSA).
High-dose therapy with caspofungin was recently evaluated primarily for safety.
102 C. J. Collins and Th. R. Rogers
7.2.4 Micafungin
7.2.5 Anidulafungin
Until recently, the mechanism of action of antifungal drugs was based mostly on
activity on the fungal cell membrane, such as for amphotericin B and triazoles.
Therefore, the benefit of a combination of antifungals that both acted on the cell
membrane was debatable. However, the echinocandins act at a different site, i.e.,
the fungal cell wall, so arguments for combination therapy, such as an echino-
candin and a polyene or an echinocandin and an azole, make more theoretical
sense. Unfortunately, high-quality clinical data on combination antifungal therapy
is limited. Recommendations for their use are often based on in vitro data, animal
studies and limited clinical experience. The IDSA published guidelines on treating
various fungal infections and recommended combination therapy in some
instances [5, 13, 2933]. For Candida spp. infections, combination therapy is
recommended for treating some forms of invasive infection but not for
candidaemia. A combination of amphotericin B and flucytosine is recommended
by the IDSA as first-line therapy for central nervous system infections, endocar-
ditis, endophthalmitis, urinary fungal balls, infected pacemakers, intracardiac
7 Systemic Antifungals 105
devices and ventricular assist devices and as second-line therapy for pyelonephritis
[13]. Combinations such as amphotericin B and flucytosine, amphotericin B and
fluconazole, and fluconazole and flucytosine may be used for treating cryptococcal
infection [30].
For initial primary IA therapy, combination therapy is not routinely recom-
mended. However, addition of a second agent may be considered as part of salvage
therapy when a primary agent is failing [5]. For endemic mycoses, combination
therapy is again not routinely recommended. However, combination therapy with
amphotericin B and an azole has been used to treat coccidioidomycosis, especially
when infection is widespread or there is single-agent failure [32].
Consensus guidelines for treating rare mould infections, such as zygomycosis,
are not available and are much needed. Combination therapy has been used to treat
some of these infections, but there are very limited clinical data to support definite
strategies of management.
vitro data are available to set these breakpoints, there is still much work to be done
in setting interpretative breakpoints for all antifungal agents against all clinically
relevant fungi. For example, whereas there are standard guidelines with inter-
pretative breakpoints for several antifungal agents versus Candida spp. [35], there
are still no interpretative criteria for any antifungal agent versus Cryptococcus
neoformans. However, based on current in vitro and clinical data, the activities of
the antifungal agents currently in use are known for many of the common IFIs
(Table 7.1).
Since the last edition of this book [1], there have been further epidemiological
studies to determine what the predominant fungal infections are, and in particular,
the frequency of candidaemia/candidiasis is in intensive care practice [36].
Differences in the reported incidence of candidaemia between European countries
have been noted, although there is no clear explanation for this [37]. C. albicans
continues to account for about half of all documented cases, although improved
microbiological investigation has meant that other Candida spp. are being more
reliably identified. In a recent Italian study [38], the incidence of candidaemia and
distribution of the causative species were recorded for 19992007 in an 18-bed
mixed medical and surgical ICU. The incidence rate was 1.42 episodes/10,000
patient days/year. Overall, C. albicans accounted for 46% of isolates and other
Candida spp. 54%, mainly C. parapsilosis (22%) and C. glabrata (13%).
Data from the 20082009 SENTRY Antimicrobial Surveillance Program on
nosocomial Candida bloodstream infections showed that C. glabrata was the
second most commonly isolated Candida spp. after C. albicans and was associated
with the highest frequency of resistance to both fluconazole and other triazoles
(range 5.17.7%) and the echinocandins (range 3.25.1%) [39].
Several recent studies assessed the relationship between patient survival, time of
starting antifungal therapy and its appropriateness following candidaemia docu-
mentation. In a Canadian study, administering adequate empirical antifungal
therapydefined as giving an adequate dose of antifungal drug to which the
identified pathogen was shown to be susceptible in vitrowas associated with a
significant decrease in mortality rate [40]. When assessing the appropriateness
of empirical antibiotic therapy in septic patients admitted to the ICU,
Garnacho-Montero et al. [41] found that fungal infection was an independent
variable related to inappropriate therapy, which in turn was a predictor of
7
Table 7.1 Comparison of the major antifungal agents used to treat fungal infections in the intensive care unit
Class Polyenes
Generic name Amphotericin B Amphotericn B colloidal dispersion Liposomal amphotericin B Amphotericin B lipid complex
deoxycholate
(conventional)
Trade name(s) Fungizone Amphocil (Eur) AmBisome Abelcet
Fungilin Amphotec (US)
Systemic Antifungals
(continued)
Table 7.1 (continued)
108
Class Polyenes
Generic name Amphotericin B Amphotericn B colloidal dispersion Liposomal amphotericin B Amphotericin B lipid complex
deoxycholate
(conventional)
Trade name(s) Fungizone Amphocil (Eur) AmBisome Abelcet
Fungilin Amphotec (US)
Indication Fungizone: Amphocil: AmBisome: Abelcet:
(Please refer to specific Systemic fungal Severe systemic or deep mycoses 1. Severe systemic or deep 1. Severe invasive candidiasis
licensed indications and full infections where toxicity and renal failure mycoses where toxicity 2. Severe systemic fungal infections
prescribing information Fungilin: preclude use of conventional (especially nephrotoxicity) in patients not responding to
according to jurisdiction; Oral and perioral amphotericin precludes use of conventional amphotericin or to
adults only unless specified) fungal infections Amphotec conventional amphotericin other antifungal drugs or where
Invasive aspergillosis in patients not 2. Suspected or proven toxicity or renal impairment
responding to conventional infection in febrile precludes conventional
amphotericin or where toxicity or neutropenic patients amphotericin, including invasive
renal impairment precludes unresponsive to broad- aspergillosis, cryptococcal
conventional amphotericin spectrum antibacterials meningitis and disseminated
cryptococcosis in HIV patients
Routes of administration Fungizone: IV, neb IV, neb IV, neb
(labelled and unlabelled IV, neb, IT,
routes) bladder
irrigation
Fungilin: topical
PO dose NA NA NA NA
(Adult only)
(continued)
C. J. Collins and Th. R. Rogers
7
Table 7.1 (continued)
Class Polyenes
Generic name Amphotericin B Amphotericn B colloidal dispersion Liposomal amphotericin B Amphotericin B lipid complex
deoxycholate
(conventional)
Trade name(s) Fungizone Amphocil (Eur) AmBisome Abelcet
Fungilin Amphotec (US)
Systemic Antifungals
Class Azoles
Generic name Fluconazole Itraconazole Voriconazole Posaconazole
Trade name(s) Diflucan Trican Sporanox Vfend Noxafil
Main spectrum of activity (relative clinical and in vitro activity)
Candida spp.
C. albicans ++ ++ ++ ++
C. glabrata + + +
C. tropicalis ++ ++ ++ ++
C. parapsilosis ++ ++ ++ ++
C. krusei + + +
Cryptococcus neoformans + ++ ++
Trichosporon spp. ++ N N N
Aspergillus spp.
A. fumigatus ++ ++ ++
A. flavus ++ ++ ++
A. terreus ++ ++ ++
A. niger ++ ++ ++
A. nidulans ++ ++ ++
Zygomycetes +
Fusarium spp. +
S. prolificans N N N
S. apiospermum + N
(continued)
C. J. Collins and Th. R. Rogers
7
Table 7.1 (continued)
Class Azoles
licensed indications and full and candidal balanitis 2. Vulvovaginal candidiasis 2. Serious infections caused infections in:
prescribing information 2. Mucosal candidiasis 3. Fungal skin infections by S. apiospermum, - those receiving remission-
according to jurisdiction; 3. Fungal skin 4. Onychomycosis Fusarium spp., or invasive induction chemotherapy AML
adults only unless specified) infections 5. Histoplasmosis fluconazole-resistant or MDS expected to result in
4. Invasive candidal 6. Systemic aspergillosis, Candida spp. (including C. prolonged neutropenia and who
infections including candidiasis and cryptococcosis krusei) are at high risk of developing
candidaemia and including cryptococcal invasive fungal infections
disseminated meningitis where other antifungal - HSCT recipients who are
candidiasis drugs are inappropriate or undergoing high-dose
5. Cryptococcal ineffective immunosuppressive therapy for
infections including 7. Maintenance in AIDS patients GVHD and who are at high risk of
meningitis to prevent relapse of underlying developing invasive fungal
6. Prevention of fungal infection infections
relapse of 8. Prophylaxis in haematological 2. Severe oropharyngeal
cryptococcal malignancy or if undergoing candidiasis or if immuno-
meningitis in AIDS BMT compromised
patients 3. Salvage therapy for invasive
7. Prevention of fungal aspergillosis, fusariosis,
infections in immuno- chromoblastomycosis, mycetoma
compromised patients and coccidioidomycosis in patients
refractory to or intolerant of first-
line therapy with other antifungal
agents
FDA: not licensed for indication
number 3
111
(continued)
Table 7.1 (continued)
112
Class Azoles
(continued)
7
Table 7.1 (continued)
Class Azoles
(continued)
113
Table 7.1 (continued)
114
Class Echinocandins
Class Echinocandins
Main side effects Hypotension, peripheral oedema, tachycardia, fever, Fever, headache, hypokalaemia, Hypokalaemia,
chills, headache, rash, hypokalaemia, GI disturbance, hypomagnesaemia, GI GI disturbance,
abnormal LFTs, phlebitis, infusion reactions, anaemia disturbance, neutropenia, abnormal LFTs
thrombocytopenia
Drug interactions Cyclosporine, tacrolimus, rifampicin Few Few
Monitor for toxicity due to
sirolimus, nifedipine and
itraconazole if co-prescribed
S. Scedosporium, spp species, no activity, slight activity, + modest activity, ++ good activity, N no data, Eur Europe, US United States, EMEA European
Medicines Agency, FDA US Food and drug administration, BMT bone marrow transplantation, IV intravenous, neb nebulised, IT intrathecal, PO oral, NA not
applicable, OD once daily, BD twice daily, TDS three times daily, QDS four times daily, TDM therapeutic drug monitoring, GI gastrointestinal, LFTs liver
function tests, CYP3A4, Cytochrome P450 3A4 enzyme, CYP2C19, Cytochrome P450 2C19 enzyme.
a
Where revelant, the doses categorised numerically correspond to the numerically categorised indications in the Indication section
b
In immunocompromised patients with invasive mycoses
117
118 C. J. Collins and Th. R. Rogers
How best to utilise systemic antifungal agents in ICU patients is the subject of
ongoing debate [43]. The challenges faced by intensivists and microbiologists are
to identify which patients are most at risk for invasive candidiasis, to determine at
what point it is appropriate to start antifungal therapy in a critically ill patient and
which antifungal agent to use.
It is now accepted that antifungals can be prescribed in one of several ways.
Firstly, there is prophylaxis, a practice that is more established in neutropenic
haematological malignancy patients. There are less convincing supportive data in
the ICU population. In considering what a preventive strategy should include,
Pfaller and Diekema [35] remind practitioners of the need to educate staff on the
importance of hand washing, optimal central vascular catheter care and prudent
antibiotic prescribing to prevent nosocomial candidiasis. They also reviewed meta-
analyses of published antifungal prophylaxis trials. The incidence of invasive
candidiasis was reduced in all meta-analyses by between 50 and 80%; three of the
five studies showed that the mortality rate was reduced while there was no
apparent overall increase in antifungal drug resistance. Despite these encouraging
results, the authors [35] suggest that further controlled trials are needed to identify
subsets of patients who would benefit most from prophylaxis and also to
emphasise the need to assess the efficacy of echinocandins, as earlier studies
predominantly evaluated fluconazole.
In the study of Bassetti et al. [38], stopping the use of prophylactic fluconazole
in their ICU resulted in a significant reduction in candidaemia caused by non-
albicans Candida spp., and there was no change in the incidence of invasive
candidiasis. Although studies showing that antifungal prophylaxis in ICU patients
reduced the number of IFIs, the effect on mortality rate and the overall benefits of
this strategy are not established. For reasons such as cost, concern about antifungal
resistance and drug-related adverse events, antifungal prophylaxis should only be
considered for selected high-risk patients.
Antifungal prophylaxis has received a grade BI recommendation from the
IDSA for patients at highest risk ([10% risk of invasive candidiasis) in ICUs with
high rates of invasive candidiasis (compared with normal rates of 12%) [13].
Fluconazole 400 mg (6 mg/kg) daily is recommended for such patients in this
setting. However, accurately identifying patients at [10% risk needs further
refining, as yet there is no universally agreed-upon risk prediction algorithm that
7 Systemic Antifungals 119
drug costs and cost per quality adjusted life year (QALY). The authors concluded
that preferred use of the echinocandin was cost effective. Their study builds on
earlier analysis of the optimal choice of empirical therapy, which also favoured an
echinocandin [50]. In an accompanying commentary to the Zilberberg study [49],
Golan [51] suggests the study may have overestimated the benefits of micafungin
compared with fluconazole and suggest further studies should be undertaken in
this area.
For antifungal therapy of a documented case of candidaemia in the ICU, the
IDSA guidelines [13] recommend either fluconazole or an echinocandin (AI grade)
with some caveats: Liposomal amphotericin B (AmBisome) was found to be as
effective as micafungin in a randomised double-blind trial [18], suggesting that it
is an alternative option, although as with other amphotericin B formulations, a
higher rate of treatment-related adverse events might be expected.
References
1. Cooke FJ, Rogers T (2005) Systemic antifungals. In: van Saene HKF, Silvestri L, de la Cal
MA (eds) Infection control in the intensive care unit, 2nd edn. Springer, Berlin, pp 155170
2. Schiller DS, Fung HB (2007) Posaconazole: an extended-spectrum triazole antifungal agent.
Clin Ther 29:18621886
3. Ullmann AJ, Lipton JH, Vesole DH et al (2007) Posaconazole or fluconazole for prophylaxis
in severe graft-versus-host disease. N Engl J Med 356:335347
4. Cornely OA, Maertens J, Winston DJ et al (2007) Posaconazole vs. fluconazole or
itraconazole prophylaxis in patients with neutropenia. N Engl J Med 356:348359
5. Walsh TJ, Anaissie EJ, Denning DW et al (2008) Treatment of aspergillosis: clinical practice
guidelines of the Infectious Diseases Society of America. Clin Infect Dis 46:327360
6. Walsh TJ, Raad I, Patterson TF et al (2007) Treatment of invasive aspergillosis with
posaconazole in patients who are refractory to or intolerant of conventional therapy: an
externally controlled trial. Clin Infect Dis 44:212
7. Greenberg RN, Mullane K, van Burik JA et al (2006) Posaconazole as salvage therapy for
zygomycosis. Antimicrob Agents Chemother 50:126133
8. van Burik JA, Hare RS, Solomon HF et al (2006) Posaconazole is effective as salvage therapy
in zygomycosis: a retrospective summary of 91 cases. Clin Infect Dis 42:e61e65
9. Page RL 2nd, Schwiesow J, Hilts A (2007) Posaconazole as salvage therapy in a patient with
disseminated zygomycosis: case report and review of the literature. Pharmacotherapy
27:290298
10. Zoller E, Valente C, Klepser ME (2010) Development, clinical utility, and place in therapy of
posaconazole for prevention and treatment of invasive fungal infections. Drug Des Devel
Ther 4:299311
11. Strorzinger D, Lichtenstern C, Weigand MA et al (2011) Posaconazole as part of the
antifungal armamentarium in the intensive care unit-case reports from a surgical ICU.
Mycoses 54(suppl 1):4548
12. Shields RK, Clancy CJ, Vadnerkar A et al (2010) Posaconazole serum concentrations among
cardiothoracic transplant recipients: factors impacting levels and correlation with clinical
response. Antimicrob Agents Chemother 55(3):13081311
13. Pappas PG, Kauffman CA, Andes D et al (2009) Clinical practice guidelines for the
management of candidiasis: 2009 update by the Infectious Diseases Society of America. Clin
Infect Dis 48:503535
7 Systemic Antifungals 121
14. Pappas PG, Rotstein CM, Betts RF et al (2007) Micafungin versus caspofungin for treatment
of candidemia and other forms of invasive candidiasis. Clin Infect Dis 45:883893
15. Vazquez JA (2010) Invasive fungal infections in the intensive care unit. Semin Respir Crit
Care Med 31:7986
16. Wagner C, Graninger W, Presterl E et al (2006) The echinocandins: comparison of their
pharmacokinetics, pharmacodynamics and clinical applications. Pharmacology 78:161177
17. Betts RF, Nucci M, Talwar D et al (2009) A multicenter, double-blind trial of a high-dose
caspofungin treatment regimen versus a standard caspofungin treatment regimen for adult
patients with invasive candidiasis. Clin Infect Dis 48:16761684
18. Kuse ER, Chetchotisakd P, da Cunha CA et al (2007) Micafungin versus liposomal
amphotericin B for candidaemia and invasive candidosis: a phase III randomised double-
blind trial. Lancet 369:15191527
19. Pappas PG, Rotstein CM, Betts RF et al (2007) Micafungin versus caspofungin for treatment
of candidemia and other forms of invasive candidiasis. Clin Infect Dis 45:883893
20. Dupont BF, Lortholary O, Ostrosky-Zeichner L et al (2009) Treatment of candidemia and
invasive candidiasis in the intensive care unit: post hoc analysis of a randomized, controlled
trial comparing micafungin and liposomal amphotericin B. Crit Care 13:R159
21. Queiroz-Telles F, Berezin E, Leverger G et al (2008) Micafungin versus liposomal
amphotericin B for pediatric patients with invasive candidiasis. Pediatr Infect Dis 27:820826
22. de Wet N, Llanos-Cuentas A, Suleiman J et al (2004) A randomized, double-blind, parallel-
group, dose-response study of micafungin compared with fluconazole for the treatment of
esophageal candidiasis in HIV-positive patients. Clin Infect Dis 39:842849
23. de Wet NT, Bester AJ, Viljoen JJ et al (2005) A randomized, double blind, comparative trial
of micafungin (FK463) vs. fluconazole for the treatment of oesophageal candidiasis. Aliment
Pharmacol Ther 21:899907
24. van Burik JA, Ratanatharathorn V, Stepan DE et al (2004) Micafungin versus fluconazole for
prophylaxis against invasive fungal infections during neutropenia in patients undergoing
hematopoietic stem cell transplantation. Clin Infect Dis 39:14071416
25. Denning DW, Marr KA, Lau WM et al (2006) Micafungin (FK463), alone or in combination
with other systemic antifungal agents, for the treatment of acute invasive aspergillosis.
J Infect 53:337349
26. Perkhofer S, Lass-Florl C (2009) Anidulafungin and voriconazole in invasive fungal disease:
pharmacological data and their use in combination. Expert Opin Investig Drugs 18:13931404
27. Reboli AC, Rotstein C, Pappas PG et al (2007) Anidulafungin versus fluconazole for invasive
candidiasis. N Engl J Med 356:24722482
28. Krause DS, Simjee AE, van Rensburg C et al (2004) A randomized, double-blind trial of
anidulafungin versus fluconazole for the treatment of esophageal candidiasis. Clin Infect Dis
39:770775
29. Kauffman CA, Bustamante B, Chapman SW et al (2007) Clinical practice guidelines for the
management of sporotrichosis: 2007 update by the Infectious Diseases Society of America.
Clin Infect Dis 45:12551265
30. Perfect JR, Dismukes WE, Dromer F et al (2010) Clinical practice guidelines for the
management of cryptococcal disease: 2010 update by the Infectious Diseases Society of
America. Clin Infect Dis 50:291322
31. Chapman SW, Dismukes WE, Proia LA et al (2008) Clinical practice guidelines for the
management of blastomycosis: 2008 update by the Infectious Diseases Society of America.
Clin Infect Dis 46:18011812
32. Galgiani JN, Ampel NM, Blair JE et al (2005) Coccidioidomycosis. Clin Infect Dis
41:12171223
33. Wheat LJ, Freifeld AG, Kleiman MB et al (2007) Clinical practice guidelines for the
management of patients with histoplasmosis: 2007 update by the Infectious Diseases Society
of America. Clin Infect Dis 45:807825
122 C. J. Collins and Th. R. Rogers
34. Cuenca-Estrella M, Rodriguez-Tudela JL (2010) The current role of the reference procedures
by CLSI and EUCAST in the detection of resistance to antifungal agents in vitro. Expert Rev
Anti Infect Ther 8:267276
35. Pfaller MA, Diekema DJ (2007) Epidemiology of invasive candidiasis: a persistent public
health problem. Clin Microbiol Rev 20:133163
36. Kett DH, Azoulay E, Echeverria PM et al. for the Extended Prevalence of Infection in the
ICU Study (EPIC. II) Group of investigators (2010) Candida bloodstream infections in
intensive care units: analysis of the extended prevalence of infection in an intensive care unit
study. Crit Care Med 39(4):665-670
37. Lass-Florl C (2009) The changing face of epidemiology of invasive fungal disease in Europe.
Mycoses 52:197205
38. Bassetti M, Ansaldi F, Nicolini L et al (2009) Incidence of candidaemia and relationship with
fluconazole use in an intensive care unit. J Antimicrob Chemother 64:625629
39. Pfaller MA, Moet GJ, Messer SA et al (2010) Candida bloodstream infections: comparison of
species distribution and antifungal resistance in community onset and nosocomial isolates in
the SENTRY antimicrobial surveillance program (20082009). Antimicrob Agents
Chemother 55(2):561566
40. Parkins MD, Sabuda DM, Elsayed S et al (2007) Adequacy of empirical antifungal therapy
and effect on outcome among patients with invasive Candida species infections. J Antimicrob
Chemother 60:613618
41. Garnacho-Montero J, Garcia-Garmendia JL, Barrero-Almodovar A et al (2003) Impact of
adequate empirical antibiotic therapy on the outcome of patients admitted to the intensive
care unit with sepsis. Crit Care Med 31:27422751
42. Morrell M, Fraser VJ, Kollef MH (2005) Delaying the empiric treatment of Candida
bloodstream infection until positive blood culture results are obtained: a potential risk factor
for hospital mortality. Antimicrob Agents Chemother 49:36403645
43. Cruciani M, Serpelloni G (2008) Management of Candida infections in the adult intensive
care unit. Expert Opin Pharmacother 9:175191
44. Clerihew L, Austin N, McGuire W (2007) Prophylactic systemic antifungal agents to prevent
mortality and morbidity in very low birth weight infants. Cochrane Database Syst Rev
(4):CD003850
45. Guery BP, Arendrup MC, Auzinger G et al (2009) Management of invasive candidiasis and
candidemia in adult non-neutropenic intensive care unit patients. Part II: Treatment. Intensive
Care Med 35:206214
46. Troughton JA, Browne G, McAuley DF et al (2010) Prior colonisation with Candida species
fails to guide empirical therapy for candidaemia in critically ill adults. J Infect 61:403409
47. Spellberg BJ, Filler SG, Edwards JE Jr (2006) Current treatment strategies for disseminated
candidiasis. Clin Infect Dis 42:244251
48. Rex JH, Bennett JE, Sugar AM et al (1994) A randomized trial comparing fluconazole with
amphotericin B for the treatment of candidemia in patients without neutropenia. N Engl J
Med 331:13251330
49. Zilberberg MD, Kothan S, Shorr AF (2009) Cost-effectiveness of micafungin as an
alternative to fluconazole empiric treatment of suspected ICU-acquired candidemia among
patients with sepsis: a model simulation. Crit Care 13:R94
50. Golan Y, Wolf MP, Pauker SG et al (2005) Empirical anti-Candida therapy among selected
patients in the intensive care unit: a cost effectiveness analysis. Ann Intern Med 143:857869
51. Golan Y (2009) Empiric anti-Candida therapy for patients with sepsis in the ICU: how little
is too little? Crit Care 13:180
Enteral Antimicrobials
8
M. Sanchez Garca, M. Nieto Cabrera, M. A. Gonzalez Gallego
and F. Martnez Sagasti
8.1 Introduction
Healthy individuals may carry one or more of the six potentially pathogenic
microorganisms (PPM), considered either normal or community-acquired, in their
upper [1, 2] and lower [3] gastrointestinal tract flora. These PPMs are Streptococcus
pneumoniae, Haemophilus influenzae, Moraxella catarrhalis, Staphylococcus
aureus, Escherichia coli, and Candida albicans. The presence of the opportunistic or
abnormal aerobic gram-negative bacilli (AGNB), Klebsiella, Enterobacter, Proteus,
Morganella, Citrobacter, Serratia, Acinetobacter, and Pseudomonas species, and of
methicillin-resistant S. aureus (MRSA) in the oropharynx and gastrointestinal tract
of healthy individuals is uncommon (see Chap. 2), although extended-spectrum
beta-lactamases (ESBL) harboring Enterobacteriaceae, as well as MRSA, are
increasingly being detected at hospital admission [4]. The nine abnormal bacteria are
predominantly carried by patients with an underlying condition, either chronic or
acute [5]. Illness severity and antibiotic use [3, 5, 6] are the most important factors in
conversion of the normal to the abnormal carrier state. Carriage of abnormal flora
invariably leads to high concentrations, i.e., abnormal bacteria overgrowth, in the
throat and gut of the critically ill [7, 8]. Overgrowth is defined as C105 of abnormal
flora per milliliter of saliva and/or gram of feces [9]. In turn, intestinal overgrowth
with AGNB is associated with systemic immune-system suppression [10, 11] and has
also been shown to be an independent risk factor for endogenous infection, endo-
toxemia, emergence of resistance, transmission via hands of carers, and outbreaks.
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 123
DOI: 10.1007/978-88-470-1601-9_8, Springer-Verlag Italia 2012
124 M. Sanchez Garca et al.
8.2.2.1 Polymyxins
Polymyxins given enterally are nonabsorbable [33] and cover AGNB, including
ESBLs in general, as well as ESBLs conferring carbapenem resistance to
enterobacteria [34, 35]. These compounds have excellent activity against
P. aeruginosa and Acinetobacter spp., although not against Proteus, Morganella,
or Serratia spp. Polymyxins are selective in that they are not active against the
indigenousmainly anaerobicflora [36]. The mode of action is disruption of the
bacterial cell wall, making the bacterial cell permeable and leading to cell death.
This mechanism is independent of enzymatic systems [37], and acquired resistance
against polymyxins is uncommon [38]. Polymyxins are inactivated to a moderate
extent by proteins, fiber, food, cell debris, and salivary and fecal compounds, and
should therefore be given in a relatively high daily dose of 400 mg of polymyxin
E (300 mg of polymyxin B) [36]. Polymyxins should also be combined with
an aminoglycoside due to the gap in activity against Proteus, Morganella and
Serratia spp. The aminoglycoside should be active against P. aeruginosa because
polymyxins lose activity against this common intensive care unit (ICU) bacterium
126 M. Sanchez Garca et al.
8.2.2.2 Aminoglycosides
Aminoglycosides have several ideal features cited above for enterally adminis-
tered antimicrobials. They are active against a wide range of AGNB, including
P. aeruginosa, have a potent bactericidal activity similar to polymyxins, and
synergistic activity with polymyxins. Antipseudomonal aminoglycosides include
gentamicin, tobramycin, and amikacin. They are nonabsorbable, and the bacteri-
cidal activity is by inhibiting protein synthesis. Tobramycin is the least inactivated
by feces, followed by amikacin and gentamicin [4042]. According to an older
study [43] based on culture results, tobramycin is considered to be selective in
terms of leaving the indigenous flora undisturbed in doses \500 mg a day. A recent
investigation [8], ancillary to a randomized clinical trial [44], used more precise
molecular quantification methods and found reductions in the Faecalibacterium
prausnitzii group, which were attributed to enterally administered tobramycin.
Although an important component of intestinal microbiotaF. prausnitzii seems
to have several beneficial functions in the normal hostit is important to note that
the study is potentially biased because it was performed in a very small subgroup,
approximately 20%, of patients enrolled in one center and that, as recently shown
by the same group [45], other factors, including nutrition type and systemic
administration of antibiotics, may have been responsible for reductions in the
F. prausnitzii group count. The clinical relevance of this finding in critically ill
patients therefore remains to be elucidated. Blood levels of tobramycin and gen-
tamicin have been monitored during SDD [4648]. Aminoglycoside levels were
8 Enteral Antimicrobials 127
8.2.2.3 Polyenes
The two polyenes used as decontaminating agents are either amphotericin B or
nystatin. They are fungicidal and highly selective, as fungi are the only PPM
covered by polyenes. They bind to a sterol of the plasma membrane and alter the
membrane permeability of the fungal cell, which leads to leakage of essential
metabolites and finally to fungal cell lysis. Absorption of polyenes is minimal
[47, 48], and emergence of resistance to them amongst yeasts and fungi is highly
uncommon [58]. Fecal inactivation of polyenes is high, explaining the high daily
dose of 2 g of amphotericin B and of 8 9 106 U of nystatin required for decon-
tamination purposes [59, 60] (Table 8.1).
8.2.2.4 Glycopeptides
Of the two glycopeptides, vancomycin and teicoplanin, the most experience as
decontaminating agents has been gathered for enterally administered vancomycin
[19]. Vancomycin is active against MRSA but cannot be considered as a selective
decontaminating agent as it covers the vast majority of the anaerobic Clostridium
spp. Thus, SDD protocols do not routinely include enterally administered van-
comycin because of its negative impact on the gut ecology. Enterally administered
vancomycin is only recommended to eradicate MRSA carriage and overgrowth
and should always be given in combination with polymyxin/tobramycin/ampho-
tericin B (PTA) to offset the potential for AGNB and yeast overgrowth as a
consequence of using a nonselective decontaminating agent [61]. The mode of
action is bactericidal, as vancomycin is bound rapidly and irreversibly to cell
walls of sensitive bacteria, thereby inhibiting cell-wall synthesis. Vancomycin
128 M. Sanchez Garca et al.
absorption is rare [48]. Inactivation by proteins, fiber, food, and feces is substantial,
hence the high daily dose of 2 g (Table 8.1).
Sixty randomized controlled trials (RCTs) evaluating SDD were conducted [62]
between 1987 and 2010 (Chap. 13). The antibiotic combinations most frequently
used were polymyxin/tobramycin and polymyxin/gentamicin, in approximately
two-thirds and one-third of trials, respectively. Ten meta-analyses of RCTs on SDD
all invariably show a significant reduction in infection, and five meta-analyses report
a mortality rate reduction [63].
Surveillance cultures of the throat and rectum are an integral part of enterally
administered antimicrobial protocols [64]. Monitoring the carrier state in the
critically ill receiving antimicrobials enterally is essential, as only surveillance
cultures allow monitoring of compliance and efficacy. SDD is considered to be
effective only if surveillance samples show AGNB, MRSA, MSSA, and yeast
eradication. In addition, comparison of microorganisms cultured in surveillance
and diagnostic samples allows classification of ICU-acquired infections in
endogenous, i.e., SDD failure, and exogenous, i.e., hygiene failure [64]. Surveil-
lance samples were taken in most trials, predominantly twice-weekly throat and/or
rectal swab, although only a few provided classification of the type of infections
detected [65].
8.3.2 Yeasts
8.3.3 MRSA
Six RCTs included vancomycin enterally but none analyzed the impact on MRSA
carriage and infections [48, 6872]. A 4% vancomycin gel applied in the lower
cheeks was associated with a significantly reduced OR for oropharyngeal MRSA
carriage to 0.25 (0.090.69) in one RCT [73]. A beforeafter study performed in a
burn unit observed significant relative risk (RR) reductions [80% for acquisition
of MRSA carriage and infection [74] after the introduction of routine enterally
administered vancomycin, and a prospective nonrandomized study performed in a
medical/surgical unit revealed similar findings [75]. The recent detection of an
outbreak of linezolid-resistant but vancomycin-susceptible MRSA [76] implies a
potential application for prevention and control of this type of problem with
topical vancomycin.
Seven studies report fecal levels of one or more decontaminating agents poly-
myxin, tobramycin, gentamicin, amphotericin B, nystatin, and vancomycin [19, 43,
59, 60, 7779]. Compared with polymyxin, tobramycin was less inactivated by
fecal material. In one study, fecal specimens contained tobramycin levels of at least
100 mg/L feces following the daily intake of 300 mg of tobramycin [43]. In
another study, individuals taking 600 mg of tobramycin daily showed [500 mg/L
of fecal sample [77]. Polymyxin is moderately inactivated by mucosal cells, fiber
and feces, and hence the variation in fecal drug levels. Polymyxin was not detected
in one-third of individuals who took 600 mg of polymyxin daily [78]. One-third
had fecal levels [1,000 mg/L of feces, whereas the remaining individuals showed
polymyxin levels between 16 and 1,000 mg/L of feces. Tobramycin at a daily dose
of 320 added to 400 mg of polymyxin is the most commonly used combination for
eradicating AGNB carriage and overgrowth due to its synergism and relatively less
fecal inactivation [80]. Vancomycin inactivation by fecal material is high. In one
study, vancomycin po was given in doses of 2 g daily for 7 days, and the mean
concentration in 25 stool samples obtained during treatment was 3,100 400 lg/g
(range of 9058,760 lg/g) [26]. Fecal concentrations of polymyxin E and genta-
micin were measured in 38 stool samples obtained from 15 patients [79]. The levels
of both were \20 lg/ml of feces in ten stools. The remaining 28 samples showed
fecal polymyxin E levels of 94 174 mg/L (median 42 lg/ml, range 01,055 mg/L)
and gentamicin levels of 466 545 mg/L (median 196 mg/L, range 02,098 lg/ml).
Inactivation of polyenes, including amphotericin B and nystatin, by fecal material
is high. Daily doses of 2,000 mg of amphotericin B or 8 9 106 U of nystatin were
associated with fecal levels of 60 and 20 mg/L of feces, respectively [59, 60].
Most parenterally administered antimicrobials do not act upon gut flora.
However, fluoroquinolones including ciprofloxacin have been shown to possess
the pharmacokinetic characteristic of transintestinal secretion [81]. A substantial
amount of intravenously administrated ciprofloxacin is excreted via mucus rather
than via bile (15 vs. 1%), leading to high fecal ciprofloxacin concentrations. The
mean fecal level of ciprofloxacin was 108.7 mg/L of feces following parenteral
administration of a daily dose of 400 mg of ciprofloxacin [81].
130 M. Sanchez Garca et al.
The antifungal 5-flucytosine is a small molecule that also possesses the phar-
macokinetic property of transintestinal secretion, i.e., 10% of systematically
administered 5-flucytosine is excreted via mucus into the gut [82]. The good
penetration of flucytosine into most body tissues and fluids has been ascribed to its
high water solubility, low molecular weight, and low protein binding properties.
These features of ciprofloxacin and 5-flucytosine can be useful in critically ill
patients in whom rectal swabs remain positive for AGNB and yeasts following
1 week of polymyxin/tobramycin/amphotericin B treatment. Failure of the classic
PTA protocol may be due to AGNB and yeasts already translocated into the gut-
associated lymphoid tissue on admission and hence escape the intraluminal lethal
activity of nonabsorbable PTA. Three days of high intravenous doses of cipro-
floxacin or 5 flucytosine has been shown to assist effective SDD as measured by
surveillance samples negative for AGNB and yeasts in patients with inflamed gut.
Apart from monitoring efficacy and compliance of enterally administered
antimicrobials, surveillance samples, in particular rectal swabs, provide the unique
method of detecting antimicrobial resistance at an early stage, allowing prompt
treatment adjustment. In three RCTs [22, 70, 83], throat and rectal swabs were also
cultured on agar plates containing 2 mg/l of polymyxin, 4 mg/l of tobramycin, and
6 mg/l of vancomycin. For example, Proteus and Serratia spp. intrinsically
resistant to polymyxins may become resistant to tobramycin. There are reports that
describe the enteral use of amikacin [84] and paromomycin [29] replacing
tobramycin to eradicate tobramycin-resistant Proteus and Serratia, respectively.
Infection-control policies that include SDD have four fundamental features [64]:
1. enterally administered antimicrobials to decontaminate the gastrointestinal
tract; they are combined with an oropharyngeal decontamination procedure
using a paste or gel containing 2% PTA; the aim of administering oropha-
ryngeal and intestinal nonabsorbable antimicrobials is preventing secondary
endogenous infections;
2. antibiotics given parenterally immediately upon admission to control primary
endogenous infections;
3. hygiene to control exogenous infections;
4. surveillance samples to monitor the SDD protocol.
The two most complete meta-analyses demonstrate that enterally administered
polymyxins with tobramycin or gentamicin significantly reduce AGNB infections
[85, 86]. Enterally administered polyenes significantly reduced both the number of
patients with yeast infections and episodes of yeast infections in 42 of 54 RCTs
evaluating the efficacy of antifungal polyenes as part of SDD [67]. Lower-airway
8 Enteral Antimicrobials 131
8.4.2 Endotoxemia
AGNBs present as overgrowth in the gut are the major source of endotoxin in the
human body. Up to 10 mg of fecal endotoxin per gram of feces has been measured
in critically ill patients with AGNB gut overgrowth [87]. For example, gut
ischemia at the time of cardiac surgery and liver transplantation promotes trans-
mural migration or translocation of AGNB present in concentrations of C105/g of
feces [88]. Most translocating AGNB are killed by macrophages of gut-associated
lymphoid tissue, including the liver, which maintains bloodstream sterility.
However, the subsequent release of endotoxin may spill over into the bloodstream
and often lead to fluctuating levels of endotoxemia [89]. The enterally adminis-
tered combination of polymyxin/tobramycin has been shown to significantly
reduce fecal endotoxin load by a factor of 104 [90]. Five RCTs evaluated the
impact of SDD on endotoxemia: three during cardiopulmonary bypass surgery
[9193] and two in liver transplant patients [94, 95]. Two trials reported a sig-
nificant reduction following SDD [91, 92]; three trials failed to show a difference
[9395]. The cardiac patients received enterally administered polymyxin/tobra-
mycin 3 days preoperatively in the two positive studies. In the negative cardiac
study, tobramycin was replaced by neomycin, a poor antiendotoxin agent [96, 97].
In the two liver transplant studies, polymyxin/tobramycin was started 12 h
preoperatively and postoperatively only [98].
The most frequently used argument against the SDD strategy, and also a sincere
concern of some intensivists, is that the routine use of SDD may promote the
development of bacterial resistance or select resistant PPMs. This type of rea-
soning directly derives from the well-known fact that extensive systemic antibiotic
132 M. Sanchez Garca et al.
[105]. This beneficial effect has been known for many years [106] and explains
why the efficacy of SDD increases with higher prevalence of the maneuver, i.e.,
during routine use in all eligible patients or without a simultaneous placebo control
group during trials. After SDD discontinuation, significant increases in some
resistance markers were detected (ceftazidime), although not in others (cipro-
floxacin, tobramycin). Resistance levels remained stable over the next 12 months
without SDD [105]. The various sources of bias for the results of this are appro-
priately addressed by the authors in the discussion section of their manuscript.
Six RCTs conducted in ICUs in which MRSA was endemic at the time of the
study showed a trend toward higher MRSA colonization [65] or infection rates in
patients receiving SDD [107112]. MRSA, by design, is not covered by SDD
antimicrobials. Inevitably, SDD exerts selective pressure on this PPM. Hence,
proponents of SDD have always accepted this possibility and proposed an SDD
strategy consisting of surveillance cultures to detect MRSA overgrowth in carriers,
combined with vancomycin administered either oropharyngeally [73, 113] or
oropharyngeally and enterally to control MRSA overgrowth [74, 75]. When there
is a serious clinical MRSA problem, this approach can be used as a prophylactic
policy in all high-risk groups. These patients should receive PTA combined with
enterally administered vancomycin on admission, and throat and rectal swabs
should be taken during the entire ICU stay. With an incidence of less than one
event per week, SDD that includes vancomycin enterally can be commenced as
treatment if swabs prove positive [114].
The high density of S. aureus, both methicillin sensitive and resistant, in the
oropharynx and gut promotes skin carriage and hand and environmental con-
tamination. Quantitative studies from the early 1950s demonstrated that S. aureus
overgrowth in the nasal cavity leads to skin carriage of S. aureus in 44% of
individuals, but only in 16% if the level of contamination of nasal secretions was
\105 S. aureus [115]. Airborne dissemination was also a function of the number of
microorganisms present in the nose. However, similar research from the late 1950s
demonstrated that the weight of microorganisms released to the environment by
the fecal carrier greatly exceeded that of organisms released by the nasal carrier
[116, 117]. Twenty years later, the importance of fecal carriage of MRSA in
children was shown from air contamination studies during nappy changing, when
patients who were fecal carriers yielded the same type from the air [118]. More
recently, gut overgrowth of MRSA was shown to be associated with a significant
amount of MRSA dispersed from the perianal site into clothing and bedding and
hence into the environment [119]. Long-stay patients invariably have overgrowth
in their throat and gut of [109 potential pathogens per milliliter of saliva or gram
of feces [7]. Washing a patient or changing a nappy may lead to hand contami-
nation of health-care workers to levels of [106 PPM/cm2 of finger surface [120].
For hand hygiene to be effective, a disinfecting agent such as 0.5% chlorhexidine
134 M. Sanchez Garca et al.
in 70% alcohol is required, and the procedure must take at least 2 min. Under these
circumstances, contamination levels are lowered at most by 104 microorganisms,
still leaving up to 102 per cm2 of finger surface [121]. These quantitative data show
that the intervention of hand disinfection in a busy ICU with a few long-stay
patients can only ever hope to reduce transmission but never abolish it [122]. From
an SDD perspective, even under the hypothetical circumstances of completely
clearing hand contamination, hand hygiene could never exert an influence on the
major infection problem of primary endogenous infection with a magnitude of
between 60 and 85% of all infections (Chap. 5). The intervention of hand disin-
fection also fails to clear oropharyngeal and gastrointestinal carriage and/or
overgrowth of PPM present on arrival. However, high standards of hygiene,
including hand disinfection, are part of the SDD infection-control protocol. This
protocol aims to reduce the level of hand contamination below which transmission
occurs. It is possible to achieve these low levels, as the enterally administered
antimicrobials eradicate throat and gut carriage of PPM and substantially reduce
overall levels of PPM density on the patients skin. In this way, handwashing
becomes more effective in controlling transmission of PPM and subsequent
endogenous and exogenous infections.
8.4.5 Outbreaks
all patients at high risk was more effective in controlling endemicity compared
with when administered to confirmed carriers only. The main concerns about its
prophylactic use is vancomycin-resistant enterococci (VRE) [126]. Vancomycin
resistance among low-level pathogens such as enterococci is an endemic problem
in ICUs in the USA.
SDD, including enterally administered vancomycin, has been evaluated in five
European studies in mechanically ventilated patients [61, 73, 75, 113, 127]. None of the
studies report an increased infection rate due to VRE. They evaluated SDD including
vancomycin in ICUs without VRE history, and in one study VRE was imported into the
unit but no change in policy was required, as rapid and extensive spread did not occur
[75]. Recent literature shows that parenterally administered antibiotics that do not
respect the patients gut ecology rather than high doses of enterally administered
vancomycin promote the emergence of VRE in the gut [128, 129].
8.5 Conclusions
References
1. Aas JA, Paster BJ, Stokes LN et al (2005) Defining the normal bacterial flora of the oral
cavity. J Clin Microbiol 43(11):57215732
2. Preza D, Olsen I, Willumsen T et al (2009) Diversity and site-specificity of the oral
microflora in the elderly. Eur J Clin Microbiol Infect Dis 28(9):10331040
3. Raum E, Lietzau S, von Baum H et al (2008) Changes in Escherichia coli resistance
patterns during and after antibiotic therapy: a longitudinal study among outpatients in
Germany. Clin Microbiol Infect 14(1):4148
4. Friedmann R, Raveh D, Zartzer E et al (2009) Prospective evaluation of colonization with
extended-spectrum beta-lactamase (ESBL)-producing Enterobacteriaceae among patients at
hospital admission and of subsequent colonization with ESBL-producing Enterobacteriaceae
among patients during hospitalization. Infect Control Hosp Epidemiol 30(6):534542
5. Tacconelli E, De Angelis G, Cataldo MA et al (2009) Antibiotic usage and risk of
colonization and infection with antibiotic-resistant bacteria: a hospital population-based
study. Antimicrob Agents Chemother 53(10):42644269
136 M. Sanchez Garca et al.
25. Ketai LH, Rypka G (1993) The course of nosocomial oropharyngeal colonization in patients
recovering from acute respiratory failure. Chest 103(6):18371841
26. van Saene HK, 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(5):677690
27. Yao YM, Lu LR, Yu Y et al (1997) Influence of selective decontamination of the digestive
tract on cell-mediated immune function and bacteria/endotoxin translocation in thermally
injured rats. J Trauma 42(6):10731079
28. van der Waaij D (1992) History of recognition and measurement of colonization resistance
of the digestive tract as an introduction to selective gastrointestinal decontamination.
Epidemiol Infect 109(3):315326
29. Bodey GP (1981) Antibiotic prophylaxis in cancer patients: regimens of oral, nonabsorbable
antibiotics for prevention of infection during induction of remission. Rev Infect Dis
3(suppl):S259S268
30. van Saene HK, Stoutenbeek CP (1987) Selective decontamination. J Antimicrob Chemother
20(4):462465
31. Harris JC, Dupont HL, Hornick RB (1972) Fecal leukocytes in diarrheal illness. Ann Intern
Med 76(5):697703
32. Stoutenbeek CP (1989) Topical antibiotic regimen. In: van Saene HK, Stoutenbeek CP,
Lawin P, Ledingham IM (eds) Infection control by selective decontamination. Springer,
Heidelberg, pp 95101
33. Guyonnet J, Manco B, Baduel L et al (2010) Determination of a dosage regimen of colistin
by pharmacokinetic/pharmacodynamic integration and modeling for treatment of G.I.T.
disease in pigs. Res Vet Sci 88(2):307314
34. Nordmann P, Cuzon G, Naas T (2009) The real threat of Klebsiella pneumoniae
carbapenemase-producing bacteria. Lancet Infect Dis 9(4):228236
35. Kumarasamy KK, Toleman MA, Walsh TR et al (2010) Emergence of a new antibiotic
resistance mechanism in India, Pakistan, and the UK: a molecular, biological, and
epidemiological study. Lancet Infect Dis 10(9):597602
36. van Saene JJ, van Saene HK, Tarko-Smit NJ, Beukeveld GJ (1988) Enterobacteriaceae
suppression by three different oral doses of polymyxin E in human volunteers. Epidemiol
Infect 100(3):407417
37. Sogaard H (1982) The pharmacodynamics of polymyxin antibiotics with special reference
to drug resistance liability. J Vet Pharmacol Ther 5(4):219231
38. Hawser SP (2010) Susceptibility of Klebsiella pneumoniae clinical isolates from 2007 to
2009 to colistin and comparator antibiotics. Int J Antimicrob Agents 2010 36(4):383384
39. Danner RL, Joiner KA, Rubin M et al (1989) Purification, toxicity, and antiendotoxin
activity of polymyxin B nonapeptide. Antimicrob Agents Chemother 33(9):14281434
40. Veringa EM, van der Waaij D (1984) Biological inactivation by faeces of antimicrobial
drugs applicable in selective decontamination of the digestive tract. J Antimicrob
Chemother 14(6):605612
41. van Saene JJ, van Saene HK, Stoutenbeek CP, Lerk CF (1985) Influence of faeces on the
activity of antimicrobial agents used for decontamination of the alimentary canal. Scand J
Infect Dis 17(3):295300
42. Hazenberg MP, Pennock-Schroder AM, van de Merwe JP (1985) Binding to and
antibacterial effect of aztreonam, temocillin, gentamicin and tobramycin on human
faeces. J Hyg (Lond) 95(2):255263
43. Mulder JG, Wiersma WE, Welling GW, van der WD (1984) Low dose oral tobramycin
treatment for selective decontamination of the digestive tract: a study in human volunteers.
J Antimicrob Chemother 13(5):495504
44. de Smet AM, Kluytmans JA, Cooper BS et al (2009) Decontamination of the digestive tract
and oropharynx in ICU patients. N Engl J Med 360(1):2031
138 M. Sanchez Garca et al.
45. Benus RF, van der Werf TS, Welling GW et al (2010) Association between
Faecalibacterium prausnitzii and dietary fibre in colonic fermentation in healthy human
subjects. Br J Nutr 29:18
46. Cavaliere F, Sciarra M, Crociani E et al (1988) Serum levels of tobramycin during selective
decontamination of the gastrointestinal tract. Minerva Anestesiol 54(5):223226
47. Zobel G, Kuttnig M, Grubbauer HM et al (1991) Reduction of colonization and infection
rate during pediatric intensive care by selective decontamination of the digestive tract. Crit
Care Med 19(10):12421246
48. Gaussorgues P, Salord M, Sirodot M et al (1991) Efficacit de la dcontamination digestive
sur la survenue des bactrimies nosocomiales chez les patients sous ventilation
mchanique et recevant des betamimtiques. Ranimation Soins Intensifs Mdecin
dUrgence 7:169174
49. Mol M, van Kan HJ, Schultz MJ, De Jonge E (2008) Systemic tobramycin concentrations
during selective decontamination of the digestive tract in intensive care unit patients on
continuous venovenous hemofiltration. Intensive Care Med 34(5):903906
50. Smith CA, Baker EN (2002) Aminoglycoside antibiotic resistance by enzymatic
deactivation. Curr Drug Targets Infect Disord 2(2):143160
51. Rodriguez-Bano J, Navarro MD, Romero L et al (2006) Clinical and molecular
epidemiology of extended-spectrum beta-lactamase-producing Escherichia coli as a cause
of nosocomial infection or colonization: implications for control. Clin Infect Dis 42(1):
3745
52. Al Naiemi N, Heddema ER, Bart A et al (2006) Emergence of multidrug-resistant gram-
negative bacteria during selective decontamination of the digestive tract on an intensive care
unit. J Antimicrob Chemother 58(4):853856
53. Brun-Buisson C, Legrand P, Rauss A et al (1989) Intestinal decontamination for control of
nosocomial multiresistant gram-negative bacilli. Study of an outbreak in an intensive care
unit. Ann Intern Med 110(11):873881
54. Abecasis F, Kerr S, Sarginson RE et al (2007) Comment on: emergence of multidrug-
resistant gram-negative bacteria during selective decontamination of the digestive tract on
an intensive care unit. J Antimicrob Chemother 60(2):445
55. Hoyen CK, Pultz NJ, Paterson DL et al (2003) Effect of parenteral antibiotic administration
on establishment of intestinal colonization in mice by Klebsiella pneumoniae strains
producing extended-spectrum beta-lactamases. Antimicrob Agents Chemother
47(11):36103612
56. Martins IS, Pessoa-Silva CL, Nouer SA et al (2006) Endemic extended-spectrum beta-
lactamase-producing Klebsiella pneumoniae at an intensive care unit: risk factors for
colonization and infection. Microb Drug Resist 12(1):5058
57. Sjolin J, Goscinski G, Lundholm M et al (2000) Endotoxin release from Escherichia coli
after exposure to tobramycin: dose-dependency and reduction in cefuroxime-induced
endotoxin release. Clin Microbiol Infect 6(2):7481
58. Vanden Bossche H, Dromer F, Improvisi I et al (1998) Antifungal drug resistance in
pathogenic fungi. Med Mycol 36(Suppl 1):119128
59. Hofstra W, Vries-Hospers HG, van der WD (1979) Concentrations of nystatin in faeces after
oral administration of various doses of nystatin. Infection 7(4):166170
60. Hofstra W, de Vries-Hospers HG, van der Waaij D (1982) Concentrations of amphotericin
B in faeces and blood of healthy volunteers after the oral administration of various doses.
Infection 10(4):223227
61. Silvestri L, Milanese M, Oblach L et al (2002) Enteral vancomycin to control methicillin-
resistant Staphylococcus aureus outbreak in mechanically ventilated patients. Am J Infect
Control 30(7):391399
62. Silvestri L, van Saene HK, Zandstra DF et al (2010) Impact of selective decontamination of
the digestive tract on multiple organ dysfunction syndrome: systematic review of
randomized controlled trials. Crit Care Med 38(5):13701376
8 Enteral Antimicrobials 139
63. Silvestri L, van Saene HK, Folla L, Milanese M (2010) Selective digestive decontamination
is superior to oropharyngeal chlorhexidine in preventing pneumonia and reducing mortality
in critically ill patients. J Bras Pneumol 36(2):270272
64. Baxby D, van Saene HK, Stoutenbeek CP, Zandstra DF (1996) Selective decontamination
of the digestive tract: 13 years on, what it is and what it is not. Intensive Care Med
22(7):699706
65. Snchez Garca M, Cambronero Galache JA, Lopez DJ et 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(3):908916
66. Silvestri L, van Saene HK, Casarin A et al (2008) Impact of selective decontamination of
the digestive tract on carriage and infection due to gram-negative and gram-positive
bacteria: a systematic review of randomised controlled trials. Anaesth Intensive Care
36(3):324338
67. Silvestri L, van Saene HK, Milanese M, Gregori D (2005) Impact of selective
decontamination of the digestive tract on fungal carriage and infection: systematic review
of randomized controlled trials. Intensive Care Med 31(7):898910
68. Bergmans DC, Bonten MJ, Gaillard CA et al (2001) Prevention of ventilator-associated
pneumonia by oral decontamination: a prospective, randomized, double-blind, placebo-
controlled study. Am J Respir Crit Care Med 164(3):382388
69. Korinek AM, Laisne MJ, Nicolas MH et al (1993) Selective decontamination of the
digestive tract in neurosurgical intensive care unit patients: a double-blind, randomized,
placebo-controlled study. Crit Care Med 21(10):14661473
70. Krueger WA, Lenhart FP, 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, double-
blind, placebo-controlled clinical trial. Am J Respir Crit Care Med 166(8):10291037
71. Pugin J, Auckenthaler R, Lew DP, Suter PM (1991) Oropharyngeal decontamination
decreases incidence of ventilator-associated pneumonia. A randomized, placebo-controlled,
double-blind clinical trial. JAMA 265(20):27042710
72. Schardey HM, Joosten U, Finke U et al (1997) The prevention of anastomotic leakage after
total gastrectomy with local decontamination. A prospective, randomized, double-blind,
placebo-controlled multicenter trial. Ann Surg 225(2):172180
73. Silvestri L, van Saene HK, Milanese M et al (2004) Prevention of MRSA pneumonia by oral
vancomycin decontamination: a randomised trial. Eur Respir J 23(6):921926
74. Cerda E, Abella A, de La Cal MA et al (2007) Enteral vancomycin controls methicillin-
resistant Staphylococcus aureus endemicity in an intensive care burn unit: a 9-year
prospective study. Ann Surg 245(3):397407
75. de la Cal MA, Cerda E, van Saene HK et al (2004) Effectiveness and safety of enteral
vancomycin to control endemicity of methicillin-resistant Staphylococcus aureus in a
medical/surgical intensive care unit. J Hosp Infect 56(3):175183
76. Sanchez Garcia M, De la Torre MA, Morales G et al (2010) Clinical outbreak of linezolid-
resistant Staphylococcus aureus in an intensive care unit. JAMA 303(22):22602264
77. Bodey GP (1980) Absorption of tobramycin after chronic oral administration. Curr Ther Res
28:394401
78. Gotoff SP, Lepper MH, Fiedler MA (1965) Treatment of salmonella carriers with colistin
sulfate. Am J Med Sci 249:399403
79. Misset B, Kitzis MD, Conscience G et al (1994) Mechanisms of failure to decontaminate the
gut with polymixin E, gentamicin and amphotericin B in patients in intensive care. Eur J
Clin Microbiol Infect Dis 13(2):165170
80. Stoutenbeek CP, van Saene HK (1990) Infection prevention in intensive care by selective
decontamination of the digestive tract. J Crit Care 5:137156
140 M. Sanchez Garca et al.
119. Brady LM, Thomson M, Palmer MA, Harkness JL (1990) Successful control of endemic
MRSA in a cardiothoracic surgical unit. Med J Aust 152(5):240245
120. Salzman TC, Clark JJ, Klemm L (1967) Hand contamination of personnel as a mechanism
of cross-infection in nosocomial infections with antibiotic-resistant Escherichia coli and
Klebsiella-Aerobacter. Antimicrobial.Agents Chemother 7:97100
121. Nystrom B (1983) Optimal design/personnel for control of intensive care unit infection.
Infect Control 4(5):388390
122. Crossley K, Landesman B, Zaske D (1979) An outbreak of infections caused by strains of
Staphylococcus aureus resistant to methicillin and aminoglycosides. II. Epidemiologic
studies. J Infect Dis 139(3):280287
123. Viviani M, van Saene HK, Dezzoni R et al (2005) Control of imported and acquired
methicillin-resistant Staphylococcus aureus (MRSA) in mechanically ventilated patients: a
dose-response study of enteral vancomycin to reduce absolute carriage and infection.
Anaesth Intensive Care 33(3):361372
124. Taylor ME, Oppenheim BA (1991) Selective decontamination of the gastrointestinal tract as
an infection control measure. J Hosp Infect 17(4):271278
125. Damjanovic V, Connolly CM, van Saene HK et al (1993) Selective decontamination with
nystatin for control of a Candida outbreak in a neonatal intensive care unit. J Hosp Infect
24(4):245259
126. Rice LB (2001) Emergence of vancomycin-resistant enterococci. Emerg Infect Dis
7(2):183187
127. Snchez M, Mir N, Cantn R et al (1997) Incidence of carriage and colonization pattern of
vancomycin-resistant enterococcus (VRE) in intubated patients (Pts) receiving topical
vancomycin (V) (abstract). 37th Interscience Conference on Antimicrobial Agents and
Chemotherapy (ICAAC)
128 Stiefel U, Paterson DL, Pultz NJ et al (2004) Effect of the increasing use of piperacillin/
tazobactam on the incidence of vancomycin-resistant enterococci in four academic medical
centers. Infect Control Hosp Epidemiol 25(5):380383
129. Salgado CD, Giannetta ET, Farr BM (2004) Failure to develop vancomycin-resistant
Enterococcus with oral vancomycin treatment of Clostridium difficile. Infect Control Hosp
Epidemiol 25(5):413417
Part III
Infection Control
Evidence-Based Infection Control
in the Intensive Care Unit 9
J. Hughes and R. P. Cooke
9.1 Introduction
J. Hughes (&)
Infection Prevention and Control, 5 Boroughs Partnership NHS
FoundationTrust/University of Chester Warrington,
Winwick, Warrington, UK
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 145
DOI: 10.1007/978-88-470-1601-9_9, Springer-Verlag Italia 2012
146 J. Hughes and R. P. Cooke
Category Application
IA Strongly recommended for implementation and then supported by
well-designed experimental, clinical or epidemiological studies
IB Strongly recommended for implementation and then supported by
certain experimental, clinical or epidemiological studies and a strong
theoretical rationale
IC Required for implementation, as mandated by federal and/or state
regulation or standard
II Suggested for implementation and then supported by suggestive
clinical or epidemiological studies or a theoretical rationale
No recommendation/ Practices for which insufficient evidence or no consensus regarding
unresolved issue efficacy exist
effective results, adherence to IPC practices is one of the best means of minimising
HCAIs [7].
Universally accepted standards for IPC include hand hygiene, use of personal
protective equipment (PPE), patient isolation, care of equipment and environ-
mental cleanliness [7, 8]. The role of invasive devices, antibiotics and surveil-
lance-related policies are discussed in Chaps. 10 and 11. Although there are a
number of studies on the benefits of such IPC measures, the evidence base is often
questionable or lacking [9]. However, several studies and reviews have tried to
address this issue [1012]. In 2010, the European Centre for Disease Prevention
and Control (ECDC) convened a group of experts to discuss patient safety and
infection prevention and control to formulate risk-based strategies and evidence-
based measures for effective HCAI reduction [13].
Table 9.2 Recommendations for hand hygiene (HCWs healthcare workers; classification of the
US Centre for Disease Control guidelines)
Recommendation Level of
evidence
When hands are visibly dirty or contaminated with proteinaceous material or are IA
visibly soiled with blood or other body fluids, wash hands with water and either
a nonantimicrobial or an antimicrobial soap
If hands are not visibly soiled, use an alcohol-based hand rub for routinely IC
decontaminating hands in all other clinical situations described
Decontaminate hands after contact with a patients intact skin (e.g. when taking IB
a pulse or blood pressure and lifting)
Decontaminate hands after contact with body fluids or excretions, mucous IA
membranes, nonintact skin and wound dressings if hands are not visibly soiled
Decontaminate hands after removing gloves IB
Before eating and after using a restroom, wash hands with water and either a IB
nonantimicrobial or antimicrobial soap
No recommendation can be made regarding the routine use of non-alcohol-based Unresolved
hand rubs for hand hygiene in healthcare settings issue
When decontaminating hands with an alcohol-based hand rub, apply product to IB
palm of one hand and rub hands together, covering all surfaces of hands and
fingers, until hands are dry
When washing hands with soap and water, wet hands first with water, apply an IB
amount of product recommended by the manufacturer to hands, and rub hands
together vigorously for at least 15 s, covering all surfaces of the hands and
fingers. Rinse hands with water and dry thoroughly with a disposable towel.
Use paper towel to turn off the faucet
Provide personnel with efficacious hand-hygiene products that have low irritancy IB
potential, particularly when these products are used many times per shift. This
recommendation applies to products used for hand antisepsis before and after
patient care in clinical areas and to products used for surgical hand antisepsis by
surgical personnel
Do not add soap to a partially empty soap dispenser. This practice of topping IA
up dispensers can lead to a bacterial contamination of the soap
Provide HCWs with hand lotions or creams to minimise the occurrence of IA
irritant contact dermatitis associated with hand antisepsis or handwashing
As part of a multidisciplinary programme to improve hand-hygiene adherence, IA
provide HCWs with a readily accessible alcohol-based hand-rub product
To improve hand-hygiene adherence among personnel who work in areas in IA
which high workloads and high intensity of patient care are anticipated, make an
alcohol-based hand rub available at the entrance to all patient rooms or at the
bedside, in other convenient locations and in individual pocket-sized containers
to be carried by HCWs
HCW healthcare workers
148 J. Hughes and R. P. Cooke
Table 9.3 Recommendations for protective clothing and care of equipment and environment
Recommendation Level of
evidence
Select protective equipment on the basis of a risk assessment of microorganism II
transmission
Gloves Wear gloves (clean, nonsterile gloves are adequate) when touching IB
blood, body fluids, secretions, excretions and contaminated items. Put on clean
sterile gloves just before touching mucous membranes and nonintact skin.
Change gloves between tasks and procedures on the same patient after contact
with material that may contain a high concentration of microorganisms. Remove
gloves promptly after use, before touching noncontaminated items and
environmental surfaces and before going to another patient, and decontaminate
hands immediately to avoid transfer of microorganisms to other patients or
environments
Face and eye protection Wear a mask or a face shield to protect mucous IB
membranes of the eyes, nose and mouth during procedures and patient-care
activities that are likely to generate splashes or sprays of blood, body fluids,
secretions or excretions
Gown Wear a gown (a clean, nonsterile gown is adequate) to protect skin and IB
prevent soiling of clothing during procedures and patient-care activities that are
likely to generate splashes or sprays of blood, body fluids, secretions or
excretions. Select a gown that is appropriate for the activity and amount of fluid
likely to be encountered. Remove a soiled gown as promptly as possible, and
wash hands to avoid transfer of microorganisms to other patients or
environments
Isolation Place in a private room a patient who contaminates the environment or IB
who does not (or cannot be expected to) assist in maintaining appropriate
hygiene or environmental control. If a private room is not available, consult with
infection-control professionals regarding patient placement or other alternatives
Patient-care equipment Handle used equipment soiled with blood, body fluids, IB
secretions or excretions in a manner that prevents skin and mucous-membrane
exposure, clothing contamination and microorganism transfer to other patients
and environments. Ensure that reusable equipment is not used for the care of
another patient until it has been cleaned and reprocessed appropriately. Ensure
that single-use items are discarded properly
Environmental control Ensure that the hospital has adequate procedures for the IB
routine care, cleaning and disinfecting of environmental surfaces, beds, bedrails,
bedside equipment and other frequently touched surfaces, and ensure that these
procedures are being followed. Handle, transport, and process used linen soiled
with blood, body fluids, secretions or excretions in a manner that prevents skin
and mucous-membrane exposure and clothing contamination and that avoids
microorganism transfer to other patients and environments
9 Evidence-Based Infection Control in the Intensive Care Unit 149
Hand washing is often referred to as the single most important means of preventing
HCAIs [11, 12, 15, 16]. The UK epic2: Guidelines for Preventing Healthcare
Associated Infections in NHS Hospitals in England suggest that effective hand
decontamination can significantly reduce infection rates in high-risk areas such as
the ICU [11]. However, there is no supporting level 1 evidence, (i.e. randomised
controlled trials), as ethical approval for such studies would be not possible. The
available evidence is based on expert consensus opinion and several observational
epidemiological studies [11, 12, 15]. WHO guidelines on hand hygiene identify
Five Moments for Hand Hygiene [12]. Hands must be decontaminated and dried
thoroughly immediately before each direct patient contact/care episode and after
any activity or contact that can result in hands becoming contaminated, such as
contact with patient-care equipment and their immediate environment.
Many guidelines and studies review a variety of differing products for hand
decontamination, with some studies suggesting that soap and water is as effective
as antimicrobial-based hand-washing products [11]. Alcohol-based hand rubs
(ABHRs) are highly popular in ICUs but alone are not effective in removing
physical dirt of soiling, with some studies suggesting that they are not deemed as
effective in removing spore-forming bacteria such as Clostridium difficile,
although other studies repute this [17]. Some studies have also shown that
once alcohol gel has evaporated, a residue is left that can be deposited in the
environment and which may facilitate growth of organisms such as Acinetobacter
species [18]. To combat these problems, newer gels with an aloe vera base and
containing copper-based biocidal formulations are being trialled that may also be
less irritating to HCWs hands [19].
Most studies fail to find compelling evidence for the general use of one hand
contaminant agent over another. However, all infer that HCWs acceptability and
effective hand hygiene techniques and adequate drying are the most essential
factors when selecting products to promote compliance with hand washing.
In Europe, there are a set of standards, referred to as European Norms (EN), which
are laboratory tests that any product needs to pass before it can be marketed.
Therefore, any product chosen should comply with EN standards [20].
Hand decontamination is often poorly performed, and several studies demon-
strate that compliance can be suboptimal [11, 12, 21], particularly by some phy-
sicians. The various reasons for this include availability of hand-decontamination
facilities, selection of harsh hand-care products, workload and HCW attitudes.
ICUs should always have enough easily accessible hand-wash basins, the rec-
ommended ratio being 1 to 1 per ICU bed in the UK. The increased availability of
ABHRs has also been found to improve compliance [12], as have regular hand-
decontamination training sessions/audits and feedback to all ICU staff [11, 12, 21].
Reinforcing the importance of physicians, in particular, as positive role models to
150 J. Hughes and R. P. Cooke
influence behaviour could also help, as they are often the main leads for ICU.
Hand-decontamination policies should also ensure that no jewellery, including
watches and rings (other than a plain wedding band) are worn, nails are kept short
and acrylic/false fingernails and nail polish are prohibited [11, 12]. Clinical staff
should wear short sleeves (or long sleeves rolled up to above the elbow) when
performing patient-care procedures. In the UK, this is referred to as the principle of
bare below the elbow [22]. However, although hand hygiene is the cornerstone
of IPC, it is nonetheless not a stand-alone procedure and should be part of a
multifaceted approach [23].
PPE includes the use of aprons, gowns, gloves, eye protection and face masks and
should be easily accessible to all HCWs. Employers have a duty to provide PPE
for staff who in turn have to wear PPE to comply with health and safety guidelines,
such as the Personal Protective Equipment Regulations (1992), which are com-
pulsory in some countries, e.g. the UK [24]. However, wearing PPE should always
be based on a patient- and task-risk assessment.
Gloves are one of the most effective barriers against microorganisms, although
studies have shown both lack of understanding and poor compliance with glove use
[11, 15, 25]. This includes gloves being worn when not required or worn for prolonged
periods with hands not being washed following glove removal. Gloves can also cause
adverse skin reactions and skin sensitivity [11, 25]. Sterile gloves should be worn for all
invasive procedures and contact with nonintact skin. Nonsterile gloves are suitable for
all other procedures. Gloves should also be single-use items and discarded immedi-
ately after each activity, followed by hand decontamination [25].
Disposable plastic aprons are just as effective as gowns in most situations.
However, where there is a possibility of contamination of the HCWs clothing with
blood or body fluids, it is recommended that disposable plastic aprons are worn.
Gowns are only required where there is a risk of gross contamination of splashing,
such as in major burn patients or severe trauma, or when dealing with biohazard
group 4 pathogens (e.g. Lassa fever) [11, 15, 26]. HCWs should also have access to
facial and eye protection. Personal respiratory protection is required when caring for
patients with serious infections, such as tuberculosis pandemic flu when aerosol-
generating procedures are performed [26, 27]. The choice of mask/respirator will
depend on the level and extent of protection required. Eye protection and visors
should be worn when there is a risk of body-fluid contamination to the face or eyes.
9.5 Isolation
High-risk items include invasive devices in contact with a break in the skin or with
mucous membranes, or those introduced into a sterile body area, such as surgical
instruments, catheters, prosthetic devices etc. These items require sterilisation, but
if that is not practically achievable, e.g. in the case of flexible endoscopes, then
high-level disinfection.
These include items in contact with intact mucous membranes or body fluids,
are contaminated with particularly virulent or readily transmissible organisms, or
items to be used on highly susceptible patients or sites, e.g. endoscopes and
respiratory equipment require disinfection.
These items are those in contact with normal or intact skin, e.g. thermometers,
stethoscopes, washbowls, toilets and bedding. Cleaning and drying is usually
sufficient, but disinfecting is required in the case of a patient with a known
infection risk, e.g. MRSA.
152 J. Hughes and R. P. Cooke
Minimal-risk items are items that do not come into contact with the patient, such
as floors and surfaces. Cleaning and drying is usually adequate unless in an
outbreak situation. Equipment used for invasive procedures should be decontam-
inated after being used on every patient, not only on those known to be infected.
Equipment such as stethoscopes and thermometers should be designated to
individual patients wherever possible. When dedicated equipment is not practical,
e.g. portable X-ray and ultrasound machines, staff should ensure that such
equipment is decontaminated between patients. Policies should ensure that all staff
receive training and are aware of their roles and responsibilities in relation to
decontamination. The policy should also ensure close liaison with procurement
officers and the IPC team when purchasing any new equipment.
Although the ICU patients surrounding is classed as low risk, a clean environment
provides the background for good standards of hygiene as well as maintaining
patient, staff and visitor confidence [1]. In the UK, the ICU environment is cate-
gorised as a high-risk area under the Standards of Environmental Cleanliness in
Hospitals Guidelines [36]. When considering the role of the environment in the
ICU, the design of the unit should ensure that there is adequate space between beds
to allow easy access for staff and equipment [37, 38]. Furniture and fixtures should
be kept to a minimum and be of materials that are easy to clean. A suitable area
should also be designated for safe storage of equipment. Dirty and clean
areas should be separate so that no mixing of equipment occurs, and clinical waste
should be stored where there is no risk of transporting waste through clean or
patient areas. Despite the pressure for ICU beds, ICU mangers should maintain
close liaison with domestic services staff to maintain standards.
As stated the IPC, manoeuvres referred to should be used in conjunction and are
rarely stand alone. Therefore, it is often a challenge to gauge exactly which
manoeuvres reviewed reduce HCAIs, and the evidence should be viewed in light
of this. However, to ensure compliance with IPC and monitor HCAIs, it is essential
that there are effective audit and surveillance systems in place to monitor HCAIs
[68, 39, 40].
The Saving Lives High Impact Interventions (HIIs), launched in the UK in
2005, are practical EBP tools to audit practice and compliance and help reduce
HCAI [68]. These are based on a care-bundle approach and have been imple-
mented by many ICUs in the UK. There are several available, e.g. care of invasive
devices such as peripheral, central venous and urinary catheters, reducing
9 Evidence-Based Infection Control in the Intensive Care Unit 153
surgical-site infections, reducing the risk of C. difficile and cleaning and decon-
tamination of the environment.
In relation to surveillance the rapid rise in the rate of bacterial antibiotic
resistance in ICU patients is well recognised [41]. Four key organism groups
warrant regular surveillance in the ICU setting, as highlighted by the National
Nosocomial Infections Surveillance (NNIS) system report in MDRPMs in US
ICUs [42]. These include MRSA, vancomycin-resistant Enterococcus faecium
(VRE), Pseudomonas aeruginosa resistant to carbapenems or fluoroquinolones
and Enterobacteriaceae resistant to third-generation cephalosporins [including
production of extended-spectrum beta-lactamases (ESBLs)]. ICU screening pro-
tocols for MRSA and VRE are well established through infection control;
guidelines for ESBL surveillance are lacking.
Many factors contribute to the high incidence of nosocomial infections in the ICU
and associated poor patient outcomes. Though most studies have come from
industrialised countries, the rates of infection in developing countries may be higher
[43]. This relates particularly to the use of invasive devices, the use of which should
be linked to an ICU audit programme. Such a programme should look at not only
infection rates but mortality rates, length of ICU stay and hospital costs.
Indwelling devices, such as central venous catheters (VCs), Foley catheters and
endotracheal tubes, bypass natural host defence mechanisms and predispose to
infection. The Surveillance and Control of Pathogens of Epidemiological Impor-
tance (SCOPE) database found that 51% of all catheter-associated blood-stream
infections (CR-BSIs) in 49 US hospitals occurred in the ICU. CR-BSI rates per
1,000 catheter days for a variety of vascular devices were listed in the report [44].
A recent systematic review indicated that CR-BSIs per 1,000 catheter days range
from 0.5 for peripheral VCs to 1.6 for long-term central VCs, 1.7 for arterial
catheters, 2.1 for peripherally inserted central VCs and 2.7 for short-term central
VCs [45]. EPIC guidelines recommend using antimicrobial-impregnated central
VC if short-term CR-BSI rates are high despite introducing a comprehensive
strategy to reduce rates [11].
The highest risk for nosocomial pneumonia is in patients on mechanical venti-
lation [i.e. ventilator-associated pneumonia (VAP)], which has been the most studied
form of nosocomial pneumonia. ICUs should therefore identify cases of VAP and
collect data on the number of ventilator-associated days on a routine basis.
Catheter-associated urinary tract infection (UTI) is overall the most common
nosocomial infection. In the ICU setting, auditing infection per 1,000 catheter days
will allow a rational approach for using urinary catheters impregnated with either
antimicrobial or antiseptic agents.
In this chapter, recommendations for EBP IPC practice and strategies have been
made following review of the literature. However, to ensure best practice, ICUs
should also be adequately staffed, as many studies demonstrate the detrimental effect
of poor staffing-to-patient ratios and increased workload [49, 50]. Good IPC practices
in the ICU rely on strong medical and nursing leadership plus good multidisciplinary
working practices. It also needs to be recognised that IPC in the ICU patient must be
practised slightly differently from the non-ICU patient. Hence, specific IPC policies
and procedures for the ICU should be drawn up. These should be underpinned by
audit and surveillance to demonstrate that they are working effectively, and they
should be reviewed by the multidisciplinary group on a regular basis.
References
1. Cooper M (2009) Prevention of infection in special wards and departmentscritical care
units. In: Fraise AP, Bradley C (eds) Ayliffes control of healthcare-associated infection, 5th
edn. Hodder Arnold, Kent
2. Fraise AP, Bradley C (eds) (2009) Ayliffes control of healthcare-associated infection, 5th
edn. Hodder Arnold, Kent UK
3. Department of Health (2006) Infection prevention and control in adult critical care reducing
the risk of infection through best practice. Department of Health, London
9 Evidence-Based Infection Control in the Intensive Care Unit 155
4. Department of Health (2009) The health and social care act 2008: code of practice for the
NHS on the prevention and control of health care associated infections and related guidance.
Department of Health, London
5. Care Quality Commission (2008) Registering with the Care Quality Commission in relation
to healthcare associated infection. Guidance for trust 2009/10. Care Quality Commission,
London
6. Department of Health (2006) Saving lives: a delivery programme to reduce healthcare
associated infection, including MRSA screening for methicillin-resistant Staphylococcus
aureus (MRSA) colonisation: a strategy for NHS trusts: a summary of best practice.
Department of Health, London
7. Department of Health (2006) Going further faster: implementing the saving lives delivery
programme sustainable change for cleaner safer care. Department of Health, London
8. Department of Health (2008) Clean safe care reducing infections and saving lives.
Department of Health, London
9. Ayliffe GJ (2000) Evidence-based practises in infection control. Br J Infect Control 1(4):59
10. NICE (2004) Guideline development methods: information for national collaborating centres
and guideline developers. National Institute for Health and Clinical Excellence (updated 2005)
11. Pratt RP, Pellowe CM, Wilson JA et al (2007) epic2: national evidence-based guidelines for
preventing healthcare-associated infections in hospitals in England. J Hosp Infect 65S:S1S64
12. World Health Organization (2009) Patient safety a world alliance for safer health care. WHO
guidelines on hand hygiene in health care. First global patient safety challenge clean care is
safer care. WHO, Geneva
13. European centre for disease prevention and control (2010) meeting report: expert consultation
on healthcare-associated infection prevention and control. ECDC Stockholm
14. Harbour R, Miller JA (2001) A new system for grading recommendations in evidence-based
guidelines. BMJ 323:334336
15. Siegel JD, Rhineheart E, Jackson M, Chiarello L (2007) Guideline for isolation precautions:
preventing transmission of infectious agents in healthcare settings. The Healthcare Infection
Control Practices Advisory Committee. Available at http://www.cdc.gov/ncidod/dhqp/pdf/
guidelines/isolation2007.pdf. Accessed Jun 2011
16. Humphreys H (2008) Can we do better in controlling and preventing methicillin-resistant
Staphylococcus aureus (MRSA) in the intensive care unit (ICU)? Eur J Clin Micro Infect Dis
27(6):409413
17. Boyce JM, Ligi C, Kohan C et al (2006) Lack of association between the increased incidence
of Clostridium difficile-associated disease and the increasing use of alcohol-based hand rubs.
Infect Control Hosp Epidemiol 27(5):479483
18. Edwards J, Patel G, Wareham DW (2007) Low concentrations of commercial alcohol hand
rubs facilitate the growth of and secretion of extracellular proteins by multidrug-resistant
strains of Acinetobacter baumannii. J Med Micro 56:15951599
19. Hall TJ, Wren MWD, Wareham DW et al (2010) Effect of the dried residues of two hand gels
on the survival of methicillinresistant Staphylococcus aureus and Acinetobacter
calcoaceticus-baumannii. J Inf Prev 11(3):7072
20. Fraise A, Bradley C (2007) Decontamination of equipment, the environment and the skin. In:
Fraise AP, Bradley C (eds) Ayliffes control of healthcare-associated infection, 5th edn.
Hodder Arnold, Kent
21. Pittet D, Simon A, Hugonnet S (2004) Hand hygiene among physicians: performance, beliefs,
and perceptions. Ann Intern Med 141:1118
22. Department of Health (2010) Uniform and workwear: guidance on uniform and workwear
policies for NHS employers. Department of Health, London
23. Silvestri L, Petros AJ, Sarginson RE et al (2005) Review: handwashing in the intensive care
unit: a big measure with modest effects. J Hosp Inf 59:172179
24. Health and Safety Executive (1992) The personal protective equipment at work regulations
guidance on regulations. The Stationery Office, London
156 J. Hughes and R. P. Cooke
25. Infection Control Nurses Association (2002) Protective clothing: principles and guidance.
Fitwise, Bathgate
26. Ho PL, Tang XP, Seto WH (2003) SARS: hospital infection control and admission strategies.
Respirology 8:S41S45
27. Humphreys H (2007) Control and prevention of healthcare-associated tuberculosis: the role
of respiratory isolation and personal respiratory protection. J Hosp Inf 66(1):15
28. Trautmann M, Pollitt A, Loh U et al (2007) Implementation of an intensified infection control
program to reduce MRSA transmission in a German tertiary care hospital. Am J Infect
Control 35(10):643649
29. Raineri E, Crema L, De Sivestri A et al (2007) Methicillin-resistant Staphylococcus aureus in
an intensive care unit: a 10 year analysis. J Hosp Infect 67(4):308315
30. Cepeda JA, Whitehouse T, Cooper B et al (2005) Isolation of patients in single rooms or
cohorts to reduce spread of MRSA in intensive-care units: prospective two-centre study.
Lancet 36(945):295304
31. Farr BM, Bellingan G (2004) Pro/con clinical debate: isolation precautions for all intensive
care unit patients with methicillin-resistant Staphylococcus aureus colonisation are essential.
Crit Care 8(3):153156
32. Groothuis J, Bauman J, Malinoski F, Eggleston M (2008) Strategies for prevention of RSV
nosocomial infection. J Perinatol 28(5):319323
33. Kappstein I, van der Muhlen K, Meschzan D et al (2009) Prevention of transmission of
methicillin-resistant Staphylococcus aureus (MRSA) infection: standard precautions instead
of isolation: a 6-year surveillance in a university hospital. Chirurg 80(1):4961
34. Kilpatrick C, Prieto J, Wigglesworth N (2008) Single room isolation to prevent transmission
of infection: development of a patient journey tool to support safe practice. Br J Inf Control
9(6):1925
35. King TA, Cooke RPD (2005) Cleaning, disinfection and sterilisation. In: Darcy AJ, Diba A
(eds) Wards anaesthetic equipment, 5th edn. Elsevier, London
36. National Patient Safety Agency (2007) The national specifications for cleanliness in the
NHS, London. NHS/NPSA. Available at www.npsa.nhs.uk/health/currentprojects/nutrition/
cleaning. Accessed Jun 2011
37. Dettenkofer M, Seegers S, Antes G et al (2004) Does the architecture of hospital facilities
influence infection rates? A systematic review. Infect Control Hosp Epidemiol 25(1):2125
38. Estates NHS (2005) Health building note 57: facilities for critical care. The Stationery Office,
London
39. McGingle KL, Gourlay ML, Buchanan IB (2008) The use of active surveillance cultures in
adult intensive care units to reduce methicillin-resistant Staphylococcus aureus-related
morbidity, mortality, and costs: a systematic review. Clin Infect Dis 46(11):17171725
40. Aragon D, Sole ML (2006) Implementing best practice strategies to prevent infection in the
ICU. Crit Care Clin N Am 18(4):441452
41. Ahern JW, Alston WK (2009) Use of longitudinal surveillance data to assess the effectiveness
of infection control in critical care. Infect Control Hosp Epidemiol 30(11):11091112
42. National Nosocomial Infections Surveillance (2000) NNIS system report, data summary from
January 1992April. Am J Infect Control 28:429
43. Depuydt PO, Blot SI, Benoit DD et al (2006) Antimicrobial resistance in nosocomial
bloodstream infection associated with pneumonia and the value of systematic surveillance
cultures in an adult intensive care unit. Crit Care Med 34(3):653659
44. Wisplinghoff H, Bischoff T, Tallent SM (2004) Nosocomial bloodstream a prospective
nationwide surveillance study. Clin Infect Dis 39:309317
45. Maki DG, Kluger DM, Crivich CJ (2006) The risk of bloodstream infection in adults with
different intravascular devices: a systematic review of 200 published prospective studies.
Mayo Clin Pro 23006:11591171
46. Cunningham R, Jenks B, Northwood J (2007) Effect of MRSA transmission of rapid PCR
testing of patients admitted to critical care. J Hosp Infect 65:2438
9 Evidence-Based Infection Control in the Intensive Care Unit 157
47. Silvestri L, van Saene HK, Casarin A (2008) Impact of selective decontamination of the
digestive tract on carriage and infection due to Gram-negative and Gram-positive bacteria: a
systematic review of randomised controlled trials. Anaesth Intensive Care 36:324338
48. Oostdijk EA, de Smet AM, Blok HE et al (2010) Ecological effects of selective
decontamination on resistant Gram-negative bacterial colonization. Am J Respir Crit Care
Med 181(5):452457
49. Stone PW, Mooney-Kane C, Larson EL (2007) Nurse working conditions and patient safety
outcomes. Med Care 45(6):571578
50. Hugonnet S, Chevrolet JC, Pittet D (2007) The effect of workload on infections in critically
ill patients. Crit Care Med 35(11):7681
Device Policies
10
A. R. De Gaudio, A. Casini and A. Di Filippo
10.1 Introduction
This chapter presents a summary of the latest guidelines for preventing infections
in intensive care units (ICUs), particularly regarding ventilation-associated
pneumonia (VAP), catheter-related bloodstream infections (CRBSI), and catheter-
associated urinary tract infections (UTI). Recent clinical trials were evaluated to
investigate the latest products, procedures, and treatments aimed at preventing
infections in ICUs. A summary table is provided, describing the most recent
acquisitions and their effectiveness (Table 10.1).
The 2004 guidelines for preventing VAP indicate the existence of fundamental
principles that possess level 1A evidence [1, 2]. These principles include educating
all workers about health epidemiology and VAP control procedures, instructing
them on how to effectively use various techniques, and continuous technique
updating through regular audits. The guidelines advise thorough cleaning of all
equipment (sterilization when possible, or high-level disinfection), with attention
to complete rinsing with sterile water, drying, and packaging. The ventilation
circuit should not be changed routinely on the basis of duration of use if used on a
single patient; rather, the circuit should be changed only when it is visibly dirty or
mechanically malfunctioning. When removing the condensation tube, healthcare
workers hands must be cleaned with soap and water or alcohol-based
A. Di Filippo (&)
Department of Critical Care Section of Anaesthesia,
c/o Careggi Teaching Hospital,
University of Florence, Firenze, Italy
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 159
DOI: 10.1007/978-88-470-1601-9_10, Springer-Verlag Italia 2012
160 A. R. De Gaudio et al.
Table 10.1 Recent advances in preventing intensive care patient infections and their
effectiveness assessed by large clinical trials
disinfectants. Nebulizers should only be used with sterile liquid and should be
placed with the most aseptic maneuver possible.
To prevent infection transmission from person to person, contaminated hands
should be frequently and thoroughly washed with water and soap (antimicrobial or
nonantimicrobial) or with an alcoholic antiseptic solution. This procedure must be
performed before and after any contact with intubated or tracheostomized
patients, as well as before and after contact with any respiratory device. It is also
recommended to wear gloves for handling respiratory secretions or contaminated
objects and to change gloves and decontaminate hands between contact with
different patients and between contact with a contaminated body site and the
respiratory tract.
To prevent inhalation of gastric content, it is good practice to remove all
devices, such as endotracheal, tracheostomy, or enteral feeding tubes when clinical
indications are absent. When the patients condition permits, orotracheal intuba-
tion should be preferred over nasotracheal intubation. Enteral tube placement
should be routinely controlled to prevent enteral nutrition-combined inhalation.
Postoperative pneumonia can be prevented by directing high-risk patients to
breathe deeply and ambulate as soon as possible. Indeed, this education would
benefit all postsurgery patients.
162 A. R. De Gaudio et al.
Recent clinical trials investigated the main methods for preventing VAP and
found that poor oral hygiene in patients undergoing mechanical ventilation is often
associated with secondary colonization of the respiratory tract, leading to sub-
sequent development of pneumonia. This observation implies that proper oral
hygiene should reduce VAP incidence; however, the existence of highly resistant
microorganisms makes the benefit of this strategy unclear.
Multiple strategies are used to decontaminate the oral cavity. For example,
Panchabhai et al. compared chlorhexidine (CHX) 0.2% with a control solution of
0.01% potassium permanganate in a prospective randomized trial [3]. Oral hygiene
was performed twice daily on 512 patients admitted to ICUs. Statistical analysis
indicated that the VAP incidence was not correlated with oral hygiene using CHX
or potassium permanganate. Mortality, seen as a secondary outcome, occurred at
rates of 34.8% for CHX-treated patients and 28.3% for the control group. How-
ever, the VAP incidence was lower (7.4%) 3 months after treatment compared
with 3 months before (21.7%), regardless of treatment type. Therefore, oral
hygiene seems to act as a protective factor against VAP development indepen-
dently of the pharmacological agent employed [3].
Koeman et al. conducted a randomized double-blind trial of 385 patients
divided into three groups: 130 placebo-treated patients, 127 treated with CHX 2%,
and 128 treated with CHX 2% ? colistin 2% [4]. The daily risk of VAP was
reduced in both treatment groups compared with placebo: 65% [hazard ratio
(HR) = 0.352; 95% confidence interval (CI), 0.1600.791; p = 0.012] for CHX
and 55% (HR = 0.454; 95% CI, 0.2240.925; p = 0.030) for CHX/COL. The
combination of colistin (COL) with CHX resulted in a significant reduction in
colonization by Gram-positive and Gram-negative endotracheal and oral micro-
organisms [4]. However, in a multicenter prospective double-blind trial con-
ducted in 228 patients undergoing mechanical ventilation for at least 5 days, oral
CHX gel decontamination was not associated with significant decreases in
VAP incidence, hospitalization duration, or mortality, nor was it effective against
multi-drug-resistant microorganisms (Pseudomonas aeruginosa, Acinetobacter,
Enterobacteriaceae) [5].
The use of povidone iodine for oral decontamination seems to be related to
reduced development of secondary infections. Regular use was associated with a
significant reduction in the incidence of VAP in a prospective randomized study
conducted on 98 patients with severe head trauma who underwent mechanical
ventilation for at least 48 h [6]. In this study, 36 patients were treated with
povidone iodine, 31 with saline, and the rest with simple secretion aspiration; VAP
incidence was 8, 39, and 42%, respectively. There were no statistically significant
differences in duration of hospitalization or intra-ICU mortality among the
groups [6].
Effective VAP reduction due to the use of electric toothbrushes is still con-
troversial. In a recent prospective trial conducted on 147 patients intubated for
more than 48 h, Pobo et al. demonstrated that the use of electric toothbrushes did
not significantly reduce VAP development compared with 0.12% CHX oral
hygiene [7].
10 Device Policies 163
Even circuit components used for ventilation and humidification of the gas
mixtures administered to patients can serve as a source of bacterial contamination,
prompting the onset of pneumonia. However, the likelihood of this occurrence is in
dispute. A study of an unselected population of 369 ICU patients who underwent
mechanical ventilation [48 h demonstrated that VAP onset was not related to the
device types used to humidify the gas mixture [18]. Similar results emerged from
another study conducted on 181 patients [19]. Moreover, bacterial filters do not
appear to be involved in reducing VAP incidence. Bacterial filters used in a group
of 230 patients undergoing mechanical ventilation [24 h did not reduce the
prevalence of respiratory infections associated with mechanical ventilation [20].
Tracheotomy is commonly performed in patients undergoing mechanical ven-
tilation over shorter and longer periods. An early tracheotomy may reduce the
duration of mechanical ventilation and the incidence of respiratory tract infections,
as well as improve patient comfort and reduce respiratory dead space. Evidence
also exists to the contrary. Early tracheotomy (within 8 days) in a small group
(60) of trauma patients was unable to reduce ventilation duration, pneumonia
frequency, or ICU hospitalization compared with a control group in which the
tracheotomy was performed C28 days after the acute event [21]. However, a
prospective study of 62 patients with isolated severe head trauma showed
that early tracheostomy shortened mechanical ventilation duration after VAP
onset [22].
The mode of mechanical ventilation also appears to play a role in preventing
VAP, but the use of positive end-expiratory pressure (PEEP) seems to be an issue.
PEEP effectiveness was recently studied in 131 mechanically ventilated patients
with chest radiographs and Horowitz indexes [250 [23]. The primary outcome
was intra-ICU mortality and secondary outcomes were VAP, acute respiratory
distress syndrome (ARDS), barotrauma, occurrence of atelectasis, and develop-
ment of hypoxemia. The application of prophylactic PEEP in ventilated hypox-
emic patients did not reduce the number of hypoxemia episodes or the VAP
incidence [23].
Placing the patient undergoing mechanical ventilation in the prone position
appears to be associated with reduced gastric content aspiration and VAP inci-
dence. However, there is a lack of data on large-scale clinical studies that validate
the effectiveness and feasibility of this strategy. For example, a multicenter pro-
spective study conducted on ICU patients on mechanical ventilation showed that
the prone position is not easily achieved and its effectiveness is therefore probably
negative [24]; these data were also confirmed in children [25].
The role of selective decontamination (SDD) of the digestive tract is contro-
versial with regards to VAP. De la Cal et al. employed a randomized double-blind
study of 107 patients with severe burns at high risk of inhalation to assess whether
SDD could reduce the incidence of infections, morbidity, and mortality in criti-
cally ill patients. Statistical analysis demonstrated that the treatment was able to
reduce mortality and incidence compared with a control group [26].
10 Device Policies 165
24 h of starting the infusion. If injections are performed, taps must first be dis-
infected, and all taps not in use should be covered. Contaminated fluids must not
be administered; it is therefore recommended that parenteral nutrition be prepared
aseptically in the pharmacy. It is also not recommended to use in-line filters for
preventing infections, as administering systemic antibiotic prophylaxis routinely
before insertion or during use of an intravascular catheter adequately prevents
colonization.
Devices with as few lines as possible should be selected for patient manage-
ment. Catheters impregnated with antimicrobial or antiseptic are best choices if
they remain in place [5 days and if the infection rate remains high in the ward
after using a global strategy. The risks and benefits of placing a CVC in the
recommended site must be weighed, comparing the reduction of infectious com-
plications and mechanical complications. Subclavian access is recommended for
reducing infection in nontunneled catheters, and using the jugular or femoral vein
is recommended regarding CVC dialysis (to avoid venous stasis). An episode of
fever should not lead to changing these devices, as a clinical evaluation will
determine whether to remove the catheter if infection is evidenced elsewhere or if
the cause is not infectious. Routine exchange by guidewire should not be used to
prevent infections; this technique should be reserved for cases of catheter mal-
function without signs of infection. Disposable peripheral arterial catheters should
be used with monitoring systems and should not substitute for routine peripheral
arterial catheters to prevent catheter infections.
Clinical trials have recently been used to assess various methods to prevent
CRBSI. For example, using particular types of medicated catheters appeared to
correlate with a reduced incidence of catheter-related infections. A randomized
study of 646 catheterizations compared the effectiveness of silverplatinum
carbon antimicrobial catheters with rifampicinminocycline medicated catheters.
The former served as effective antimicrobials, but the proportions of catheter-
related infections were extremely low in both groups (1.4% for silverplatinum
carbon catheters vs. 1.7% for medical catheters with rifampicinminocycline)
[29]. Rupp et al. initiated a multicenter randomized double-blind controlled study
of 780 patients to compare the incidence of infections due catheters medicated
with CHX and sulfadiazine versus standard catheters. The results suggested that
medicated catheters are highly tolerated by the patient, with less colonization by
pathogenic organisms at the time of removal. The main colonizing microorgan-
isms were coagulase-negative staphylococci and other Gram-positive microor-
ganisms. Noninfectious adverse events occurred in both groups with comparable
frequencies [30]. However, a recent prospective multicenter randomized con-
trolled study demonstrated that there was no effective reduction in the infection if
multilumen CVCs or catheters impregnated with silver were used. In both groups
of patients, the incidence of infections was high: 2.5% in the standard catheter
group versus 2.7% in the multilumen silver-impregnated catheter group [31].
Tunneling catheters are often used to prevent colonization and subsequent
infections related to long-standing catheters. Darouiche et al. showed that a group
of patients with long-stay antimicrobial-medicated catheters experienced a lower
10 Device Policies 167
Several studies provide useful information regarding UTI prevention [2, 37].
These studies argue that UTI prevention occurs in surgical patients by limiting the
use of bladder catheters to only those cases with real need. The necessity of
the bladder catheter should be assessed on the basis of clinical circumstances, and
the routine use of these devices is therefore not recommended. Indwelling urinary
catheters are recommended in long-term-care patients for managing incontinence.
Closed drainage systems are recommended after aseptically inserting the uri-
nary catheter; catheters should only be inserted under the appropriate indications
and should be kept in place only as long as necessary. Use and duration should be
minimized in all patients, especially in those at increased risk for UTI, such as
women, the elderly, and patients with compromised immune systems. Aseptic
insertion should be ensured; and hospital staff, patients, and family should be
educated in the correct insertion technique. It is necessary to attain a free
urine flow.
168 A. R. De Gaudio et al.
The risks and benefits of various catheterization approaches have also been
assessed. If an intermittent catheterization is preferable, this technique should be
performed at regular intervals to avoid bladder distension. When urinary catheter
placement is indicated in surgical patients, it must be removed as soon as possible
after surgery, preferably within 24 h, unless there are appropriate indications for
continuous use. If UTI rate is not reduced after applying a comprehensive strategy
(appropriate positioning and proper aseptic maintenance), using catheters impreg-
nated with an antimicrobial or antiseptic should be considered. After inserting the
catheter aseptically, maintaining a closed drainage system is recommended.
In the absence of specific clinical indications, the use of systemic antibiotics as
routine UTI prophylaxis is not recommended. For UTI prevention, it is not necessary
to clean the periurethral area when routine cleaning during daily hygiene is required.
Replacing catheters or drainage bags at regular intervals is also not recommended.
Catheter replacement should be based on clinical indications, such as infections,
obstructions, or when the closed system is compromised. Disposable sterile gel is
also recommended for catheter insertion. Medical staff and professionals who
manage catheters should continue to take refresher courses and further education for
insertion and removal of the maintenance device. Hand hygiene before catheter
insertion or any manipulation of the catheter site or devices is mandatory.
Several recent clinical trials have been performed regarding the prevention of
UTI. A randomized double-blind controlled study of 212 patients enrolled consec-
utively after traumatic events compared the UTI incidence in patients with silicone
urinary catheters with patients with catheters impregnated with nitrofurazone. There
were significantly fewer bacteriurias and fungurias associated with nitrofurazone-
impregnated bladders than with the silicone urinary catheters (9.1 vs. 24.7%). The
clinical significance of asymptomatic bacteria and fungi in urine was unclear, lim-
iting the study [38]. However, the use of hydrophilic catheters in a randomized
controlled trial for self-intermittent catheterization after spinal injury did not sig-
nificantly reduce the UTI incidence compared with standard catheters [39].
Finally, the efficacy of prophylactic antibiotic therapy before bladder catheter
removal was assessed. The prospective randomized study was conducted on 239
patients undergoing major abdominal surgery who were catheterized periopera-
tively; the usefulness and effectiveness of trimethoprimsulfamethoxazole in three
doses was evaluated. Urine cultures were taken before and 3 days after bladder
catheter removal. Trimethoprimsulfamethoxazole administration significantly
reduced the incidence of symptomatic UTI if administered before catheter removal
(4.9% of the analysis group vs. 21.6% in the control group, p \ 0.001) [40].
10.5 Conclusions
high-risk patients, endotracheal tubes with silver were all shown to be useful in
preventing VAP. Medicated catheters and CHX-based dressings were efficacious
against CRBSI. UTIs were shown to be prevented through the use of medical
catheters. All these procedures can be incorporated into departmental protocols for
preventing nosocomial infections in ICUs.
References
1. Centers for Disease Control and Prevention (2004) Guidelines for preventing health-care-
associated pneumonia, 2003: recommendations of CDC and the Healthcare Infection Control
Practices Advisory Committee. MMWR 53(RR-3)
2. Di Filippo A, De Gaudio AR (2003) Device-related infections in critically ill patients. Part II:
prevention of ventilator-associated pneumonia and urinary tract infections. J Chemother
15(6):536542
3. Panchabhai TS, Dangayach NS, Krishnan A et al (2009) Oropharyngeal cleansing with 0.2%
chlorhexidine for prevention of nosocomial pneumonia in critically ill patients: an open-label
randomized trial with 0.01% potassium permanganate as control. Chest 135(5):11501156
4. Koeman M, van der Ven AJ, Hak E et al (2006) Oral decontamination with chlorhexidine
reduces the incidence of ventilator-associated pneumonia. Am J Respir Crit Care Med
173(12):13481355
5. Fourrier F, Dubois D, Pronnier P et al (2005) Effect of gingival and dental plaque antiseptic
decontamination on nosocomial infections acquired in the intensive care unit: a double-blind
placebo-controlled multicenter study. Crit Care Med 33(8):17281735
6. Seguin P, Tanguy M, Laviolle B et al (2006) Effect of oropharyngeal decontamination by
povidone-iodine on ventilator-associated pneumonia in patients with head trauma. Crit Care
Med 34(5):15141519
7. Pobo A, Lisboa T, Rodriguez A, RASPALL Study Investigators et al (2009) A randomized
trial of dental brushing for preventing ventilator-associated pneumonia. Chest 136(2):
433439
8. Caruso P, Denari S, Ruiz SA et al (2009) Saline instillation before tracheal suctioning
decreases the incidence of ventilator-associated pneumonia. Crit Care Med 37(1):3238
9. Lorente L, Lecuona M, Martn MM et al (2005) Ventilator-associated pneumonia using a
closed versus an open tracheal suction system. Crit Care Med 33(1):115119
10. Bouza E, Prez MJ, Muoz P et al (2008) Continuous aspiration of subglottic secretions in
the prevention of ventilator-associated pneumonia in the postoperative period of major heart
surgery. Chest 134(5):938946
11. Poelaert J, Depuydt P, De Wolf A et al (2008) Polyurethane cuffed endotracheal tubes to
prevent early postoperative pneumonia after cardiac surgery: a pilot study. J Thorac
Cardiovasc Surg 135(4):771776
12. Lorente L, Lecuona M, Jimnez A et al (2007) Influence of an endotracheal tube with
polyurethane cuff and subglottic secretion drainage on pneumonia. Am J Respir Crit Care
Med 176(11):10791083
13. Valencia M, Ferrer M, Farre R et al (2007) Automatic control of tracheal tube cuff pressure in
ventilated patients in semirecumbent position: a randomized trial. Crit Care Med 35(6):
15431549
14. Young PJ, Pakeerathan S, Blunt MC et al (2006) A low-volume, low-pressure tracheal tube
cuff reduces pulmonary aspiration. Crit Care Med 34(3):632639
15. Sanjay PS, Miller SA, Corry PR et al (2006) The effect of gel lubrication on cuff leakage of
double lumen tubes during thoracic surgery. Anaesthesia 61(2):133137
170 A. R. De Gaudio et al.
16. Rello J, Kollef M, Diaz E et al (2006) Reduced burden of bacterial airway colonization with a
novel silver-coated endotracheal tube in a randomized multiple-center feasibility study. Crit
Care Med 34(11):27662772
17. Kollef MH, Afessa B, Anzueto A et al (2008) Silver-coated endotracheal tubes and incidence
of ventilator-associated pneumonia: the NASCENT randomized trial. JAMA 300(7):805813
18. Lacherade JC, Auburtin M, Cerf C et al (2005) Impact of humidification systems on
ventilator-associated pneumonia: a randomized multicenter trial. Am J Respir Crit Care Med
172(10):12761282
19. Boots RJ, George N, Faoagali JL et al (2006) Double-heater-wire circuits and heat-and-
moisture exchangers and the risk of ventilator-associated pneumonia. Crit Care Med
34(3):687693
20. Lorente L, Lecuona M, Mlaga J et al (2003) Bacterial filters in respiratory circuits: an
unnecessary cost? Crit Care Med 31(8):21262130
21. Barquist ES, Amortegui J, Hallal A et al (2006) Tracheostomy in ventilator dependent trauma
patients: a prospective, randomized intention-to-treat study. J Trauma 60(1):9197
22. Bouderka MA, Fakhir B, Bouaggad A et al (2004) Early tracheostomy versus prolonged
endotracheal intubation in severe head injury. J Trauma 57(2):251254
23. Manzano F, Fernndez-Mondjar E, Colmenero M et al (2008) Positive-end expiratory
pressure reduces incidence of ventilator-associated pneumonia in nonhypoxemic patients.
Crit Care Med 36(8):22252231
24. van Nieuwenhoven CA, Vandenbroucke-Grauls C, van Tiel FH et al (2006) Feasibility and
effects of the semirecumbent position to prevent ventilator-associated pneumonia:
a randomized study. Crit Care Med 34(2):396402
25. Aly H, Badawy M, El-Kholy A et al (2008) Randomized, controlled trial on tracheal
colonization of ventilated infants: can gravity prevent ventilator-associated pneumonia?
Pediatrics 122(4):770774
26. de La Cal MA, Cerd E, Garca-Hierro P et al (2005) Survival benefit in critically ill burned
patients receiving selective decontamination of the digestive tract: a randomized, placebo-
controlled, double-blind trial. Ann Surg 241(3):424430
27. Centers for Disease Control and Prevention (2002) Guidelines for the prevention of
intravascular catheter-related infections. MMWR 2002 51(RR-10)
28. De Gaudio AR, Di Filippo A (2003) Device-related infections in critically ill patients. Part I:
prevention of catheter-related bloodstream infections. I Chemother 15(5):419427
29. Fraenkel D, Rickard C, Thomas P et al (2006) A prospective, randomized trial of rifampicin-
minocycline-coated and silver-platinum-carbon-impregnated central venous catheters. Crit
Care Med 34(3):668675
30. Rupp ME, Lisco SJ, Lipsett PA et al (2005) Effect of a second-generation venous catheter
impregnated with chlorhexidine and silver sulfadiazine on central catheter-related infections:
a randomized, controlled trial. Ann Intern Med 143(8):570580
31. Kalfon P, de Vaumas C, Samba D et al (2007) Comparison of silver-impregnated with standard
multi-lumen central venous catheters in critically ill patients. Crit Care Med 35(4):10321039
32. Darouiche RO, Berger DH, Khardori N et al (2005) Comparison of antimicrobial impregnation
with tunneling of long-term central venous catheters: a randomized controlled trial. Ann Surg
242(2):193200
33. Parienti JJ, Thirion M, Mgarbane B et al (2008) Femoral vs jugular venous catheterization
and risk of nosocomial events in adults requiring acute renal replacement therapy:
a randomized controlled trial. JAMA 299(20):24132422
34. Mimoz O, Villeminey S, Ragot S et al (2007) Chlorhexidine-based antiseptic solution vs
alcohol-based povidone-iodine for central venous catheter care. Arch Intern Med 167(19):
20662072
35. Bleasdale SC, Trick WE, Gonzalez IM et al (2007) Effectiveness of chlorhexidine bathing to
reduce catheter-associated bloodstream infections in medical intensive care unit patients.
Arch Intern Med 167(19):20732079
10 Device Policies 171
36. Timsit JF, Schwebel C, Bouadma L et al (2009) Chlorhexidine-impregnated sponges and less
frequent dressing changes for prevention of catheter-related infections in critically ill adults:
a randomized controlled trial. JAMA 301(12):12311241
37. Healthcare Infection Control Practices Advisory Committee (2009) Guideline for prevention
of catheter-associated urinary tract infections. http://www.cdc.gov/ncidod/dhqp/pdf/
guidelines/CAUTI_Guideline2009final.pdf. Accessed June 2011
38. Stensballe J, Tvede M, Looms D et al (2007) Infection risk with nitrofurazone-impregnated
urinary catheters in trauma patients: a randomized trial. Ann Intern Med 147(5):285293
39. Cardenas DD, Hoffman JM (2009) Hydrophilic catheters versus noncoated catheters for
reducing the incidence of urinary tract infections: a randomized controlled trial. Arch Phys
Med Rehabil 90(10):16681671
40. Pfefferkorn U, Lea S, Moldenhauer J et al (2009) Antibiotic prophylaxis at urinary catheter
removal prevents urinary tract infections: a prospective randomized trial. Ann Surg
249(4):573575
Antibiotic Policies in the Intensive
Care Unit 11
H. K. F. van Saene, N. J. Reilly,
A. de Silvestre and F. Rios
11.1 Introduction
Every intensive care unit (ICU) should have well-structured guidelines on the use
of antimicrobial agents to guarantee that patients requiring intensive care receive
appropriate antimicrobials for a relevant period to prevent and treat infections.
These guidelines should meet the therapeutic needs of the consultants and allow
the intensivist, clinical microbiologist, and pharmacist to monitor efficacy, toxic-
ityincluding allergy and diarrheaand side effects, such as the emergence of
resistant strains and subsequent outbreaks of superinfections. Calculation of
infection rates is only feasible following implementation of an antibiotic policy.
Apart from audit and research, antimicrobial guidelines aid educational programs
and enable the clinical pharmacist to control drug expenditure.
The main feature of an antibiotic policy in the ICU is the use of a minimum of
well-established antimicrobial agents that are associated with a minimum of side
effects but also allow the control of the three patterns of ICU infections due to the
15 potentially pathogenic microorganisms (PPM) (Chaps. 3 and 5).
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 173
DOI: 10.1007/978-88-470-1601-9_11, Springer-Verlag Italia 2012
174 H. K. F. van Saene et al.
[42]. The aim is to achieve a tissue concentration at the time of the surgical trauma
in order to prevent wound infections. Patients who require intensive care due to an
acute trauma or worsening of the underlying disease invariably receive invasive
devices, including ventilation tube, urinary catheter, and intravascular lines. These
interventions are well-known risk factors for lower airway, bladder, and blood-
stream infections in the ICU patient whose immunoparalysis is at its nadir during
the first week of admission. This is the time during which primary endogenous
infections occur. Only the immediate parenteral administration of antimicrobials
can prevent this type of infection and early therapy of an already incubating
primary endogenous infection. If the primary endogenous infection is the indica-
tion for ICU admission, parenterally administered antimicrobials are required to
treat the established infection. The philosophy that preventing infection is always
better than cure dictates the immediate systemic administration of antibiotics to a
critically ill patient requiring mechanical ventilation. Hence, systemically
administered cefotaxime is an integral part of the concept of the prophylactic
protocol of selective decontamination of the digestive tract (SDD) [43].
The criteria for parenteral administration include flora friendliness and low
resistance potential (after excretion into throat and gut via saliva, bile, and mucus)
and anti-inflammation propensity. In addition, the pharmacokinetic properties
should include a high excretion in the target organs and, in particular, in bronchial
secretions. The ratio of the antimicrobial level in the target organ and the MBC
(Table 11.4) determines the efficacy against a particular microorganism (Chap. 7).
Protein binding should be minimal. Parenterally administered antimicrobials
require a good safety profile in terms of allergy, nephro- and ototoxicity, and
influence on hemostasis. Finally, the target PPM is a criterion of paramount
importance for choosing an antimicrobial to be administered parenterally. Primary
endogenous infections are caused by both normal, e.g., Streptococcus pneumoniae,
Haemophilus influenzae, and S. aureus, and abnormal potential pathogens,
including AGNB and MRSA. General health before ICU admission influences the
carrier state (Chap. 2). Previously, healthy individuals such as trauma, burn, acute
liver, and pancreatitis patients only carry normal potential pathogens, whereas
patients with chronic underlying diseases, including chronic obstructive pulmon-
ary disease, diabetes, and alcoholism, may carry AGNB and MRSA. It is obvious
that patients referred to the ICU from other hospitals or wards are highly likely to
be carriers of abnormal potential pathogens. Taking these criteria into consider-
ation, there are only a few antimicrobials suitable for prophylaxis. The first- and
second-generation b-lactams cover the normal potential pathogens but are less
effective against the abnormal AGNB. Cefotaxime, a third-generation cephalo-
sporin, has an adequate spectrum toward both normal and abnormal AGNB, with
the exception of P. aeruginosa and Acinetobacter spp. Ceftazidime adequately
covers AGNB but at the expense of S. aureus. Cefepime may be a suitable
alternative for ceftazidime. Three randomized SDD trials used the fluoroquino-
lones ciprofloxacin and ofloxacin as systemic prophylaxis despite inadequate cover
of S. pneumoniae [4446]. Most SDD studies employed cefotaxime as the
parenterally administered antimicrobial. Many patients admitted to a medical/
180 H. K. F. van Saene et al.
Desired endpoints
Efficacy: clinical endpoints
reduction in infectious morbidity (superinfection)
reduction in mortality following the immediate administration of empirical antimicrobial
Safety: microbiological endpoints (using surveillance samples)
impact on ecology: yeasts, Clostridium difficile (diarrhea)
antimicrobial resistance: supercarriage of aerobic Gram-negative bacilli, methicillin-resistant
Staphylococcus aureus, vancomycin-resistant enterococci
Costs
duration: an antimicrobial course of \1 week is as good as that of C2 weeks
is the newer, more expensive, antimicrobial superior than the older, less expensive one, in terms
of efficacy and safety?
References
1. Vollaard EJ, Clasener HAL (1994) Colonisation resistance. Antimicrob Agents Chemother
335:409414
2. Vlaspolder F, de Zeeuw G, Rozenberg-Arska M et al (1987) The influence of flucloxacillin
and amoxicillin with clavulanic acid on the aerobic flora of the alimentary tract. Infection
15:241244
3. Harbarth S, Liassine N, Dharan S et al (2000) Risk factors for persistent carriage of
methicillin-resistant Staphylococcus aureus. Clin Infect Dis 31:13801385
4. Shek FW, Stacey BSF, Rendell J et al (2000) The rise of Clostridium difficile: the effect of
length of stay, patient age and antibiotic use. J Hosp Infect 45:235237
5. Donskey CJ, Chowdry TK, Hecker MT et al (2000) Effect of antibiotic therapy on the density
of vancomycin-resistant enterococci in the stool of colonised patients. N Engl J Med
343:19251932
6. van Saene R, Fairclough S, Petros A (1998) Broad- and narrow-spectrum antibiotics: a
different approach. Clin Microbiol Infect 4:5657
7. Buck AC, Cooke EM (1969) The fate of ingested Pseudomonas aeruginosa in normal
persons. J Med Microbiol 2:521525
8. van Saene HKF, Stoutenbeek CP, Geitz JN et al (1988) Effect of amoxycillin on colonisation
resistance in human volunteers. Microb Ecol Health Dis 1:169177
11 Antibiotic Policies in the Intensive Care Unit 185
30. Dofferhoff AS, Nijland JH, de Vries-Hospers HG et al (1991) Effects of different types and
combinations of antimicrobial agents on endotoxin release from Gram-negative bacteria: an
in vitro and in vivo study. Scand J Infect Dis 23:745754
31. Jackson JJ, Kropp H (1992) Beta-lactam antibiotic-induced release of free endotoxin: in vitro
comparison of penicillin-binding protein (PBP) 2-specific imipenem and PBP 3-specific
ceftazidime. J Infect Dis 165:10331041
32. Prins JM, van Deventer SJH, Kuijper EJ, Speelman P (1994) Clinical relevance of antibiotic-
induced endotoxin release. Antimicrob Agents Chemother 38:12111218
33. Luchi M, Morrison DC, Opal S et al (2000) A comparative trial of imipenem versus
ceftazidime in the release of endotoxin and cytokine generation in patients with
Gram-negative urosepsis. J Endotoxin Res 6:2531
34. Prins JM, van Agtmael MA, Kuijper EJ et al (1995) Antibiotic-induced endotoxin release in
patients with Gram-negative urosepsis: a double-blind study comparing imipenem and
ceftazidime. J Infect Dis 172:888891
35. Holzheimer RG, Hirte JF, Reith B et al (1996) Different endotoxin release and IL-6 plasma
levels after antibiotic administration in surgical intensive care patients. J Endotoxin Res
3:261267
36. Crosby HA, Bion JF, Penn CW, Elliott TSJ (1994) Antibiotic-induced release of endotoxin
from bacteria in vitro. J Med Microbiol 40:2330
37. Artenstein AW, Cross AS (1989) Inhibition of endotoxin reactivity by aminoglycosides.
J Antimicrob Chemother 24:826828
38. Foca A, Matera G, Berlinghieri MC (1993) Inhibition of endotoxin-induced interleukin 8
release by teicoplanin in human whole blood. Eur J Clin Microbiol Infect Dis 12:940944
39. Siedlar M, Szczepanik A, Wieckiewicz J et al (1997) Vancomycin down-regulates
lipopolysaccharide-induced tumour necrosis factor alpha (TNF alpha) and TNF alpha
mRNA accumulation in human blood monocytes. Immunopharmacology 35:265271
40. Prentice HG, Hann IM, Herbrecht R et al (1997) A randomized comparison of liposomal
versus conventional amphotericin B for the treatment of pyrexia of unknown origin in
neutropenic patients. Br J Haematol 98:711718
41. Peters M, Petros A, Dixon G et al (1999) Acquired immuno-paralysis in paediatric intensive
care: prospective observational study. BMJ 319:609610
42. Kaiser AB (1986) Antimicrobial prophylaxis in surgery. N Engl J Med 315:11291138
43. Stoutenbeek CP (1989) The role of systemic antibiotic prophylaxis in infection prevention in
intensive care by SDD. Infection 17:418421
44. Lingnau W, Berger J, Javorsky F et al (1997) Selective intestinal decontamination in multiple
trauma patients: prospective, controlled trial. J Trauma 42:687694
45. Verwaest C, Verhaegen J, Ferdinande P et al (1997) Randomised, controlled trial of selective
digestive decontamination in 600 mechanically ventilated patients in a multi-disciplinary
intensive care unit. Crit Care Med 25:6371
46. Krueger WA, Lenhart FP, 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 patients. Am J Respir Crit Care Med 166:10291037
47. Baxby D, van Saene HKF, Stoutenbeek CP, Zandstra DF (1996) Selective decontamination of
the digestive tract: 13 years on, what it is and what it is not. Intensive Care Med 22:699706
48. Damjanovic V, Connolly CM, van Saene HKF et al (1993) Selective decontamination with
nystatin for control of a Candida outbreak in a neonatal intensive care unit. J Hosp Infect
24:245259
49. de la Cal MA, Cerda E, van Saene HKF et al (2004) Effectiveness and safety of enteral
vancomycin to control endemicity of methicillin-resistant Staphylococcus aureus in a
medical/surgical intensive care unit. J Hosp Infect 56:175183
50. Desai MH, Rutan RL, Heggers JP, Herndon DN (1992) Candida infection with and without
nystatin prophylaxis. Arch Surg 127:159162
11 Antibiotic Policies in the Intensive Care Unit 187
12.1 Introduction
Two recent sets of publications were taken into consideration when preparing our
analysis of infectious outbreaks in the intensive care unit (ICU). The first concerns
the emergence of severe acute respiratory syndrome (SARS) and avian flu in 2003,
and a spread across the world of a novel influenza caused by SwH1N1 in 2009. These
viral infections had a major impact on intensive care and are described in Chap. 20.
This chapter is dedicated to describing outbreaks caused by bacteria and fungi, with
references to secondary infections associated with flu and SARS [1, 2]. The second
publication concerns the International Study of the Prevalence and Outcomes of
Infection in Intensive Care Units published in December 2009 [3]. Although this is
a point-prevalence study, it provides information about the global epidemiology of
Infection in ICUs. Unfortunately, it could not give insight into outbreaks of infection
in ICUs, so we searched for specific publications describing such outbreaks.
In the second (2005) edition of this book, we analysed the usefulness of
molecular techniques in selected outbreaks [4]. The majority of outbreaks occurred
in the last decade of the twentieth century. However, reports were usually published
several years later. A similar pattern was observed when we analysed outbreaks
published in the first decade of the twenty-first century: the actual outbreaks
occurred a few years earlier. Indeed, the above-mentioned point-prevalence study
was conducted on 8 May 2007 but published in December 2009 [3]. Therefore, for
accuracy, this analysis indicates when outbreaks actually happened and when they
were subsequently published. Acinetobacter outbreaks were selected to illustrate
V. Damjanovic (&)
Institute of Ageing and Chronic Disease, University of Liverpool,
Liverpool, UK
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 189
DOI: 10.1007/978-88-470-1601-9_12, Springer-Verlag Italia 2012
190 V. Damjanovic et al.
this point (Fig. 12.1). In addition to the reported outbreaks, a number of publica-
tions considered many relevant aspects of infection and outbreaks in ICU. Some of
these are included in this chapter. We analysed 97 publications, the majority of
which met the definition of an outbreak in neonatal (NICU), paediatric (PICU) and
adult (AICU) ICUs and reported since 2000. The main objective of this analysis
was to find out whether there were any new features in the outbreaks of infection in
ICU at the beginning of the new century, including those influenced by new viruses.
We used the same framework as in the second edition of this book; however,
outbreaks were not presented separately per ICU type but according to caus-
ative organisms, in the following order: methicillin-resistant Staphylococcus
aureus (MRSA), vancomycin-resistant enterococci (VRE), aerobic Gram-
negative bacilli (AGNB), Pseudomonas spp., Acinetobacter spp. and fungi,
together with the selected features searched (Table 12.1). The number of ana-
lysed outbreaks is stated, but only selected outbreaks are shown and listed in
the references.
12 Outbreaks of Infection in the ICU 191
Causative Features
microorganisms
Emerging Methods used Pathogenesis Prevention Endpoint
threats and control
MRSA, VRE, New Surveillance Exogenous versus Hygiene, Morbidity
AGNB, antibiotic cultures, endogenous, anti-sepsis, and mortality
Pseudomonas resistance, molecular endemic infection SDD associated
spp., SARS, techniques, versus outbreak with outbreak
Acinetobacter H1N1 statistics
spp., fungi
SDD selective digestive decontamination
We retrieved reports on six outbreaks [510] published since 2000; five occurred
in AICUs and one in an animal ICU. Reports of two outbreaks were published
in 2002 and three in 2004, all occurring between 1997 and 2000. One report
published in 2007 did not report the actual time of the outbreak. These outbreaks
are summarised briefly according to their countries of origin. A paper from Italy
published in 2002 reported a unique experience of controlling a MRSA outbreak of
8 months duration in a medical/surgical AICU in 1998 using enterally adminis-
tered vancomycin in mechanically ventilated patients [5]. Another report from
Italy, published in 2004, described the identification of a variant of the Rome
clone of MRSA responsible for an outbreak in a cardiac surgery ICU, which
occurred in 1999 in a hospital in Rome. This strain had decreased sensitivity to
vancomycin and was resistant to many antibiotics [6]. A study from Germany
published in 2002 described the occurrence of MRSA in ICU in terms of endemic
and epidemic infections followed from January 1997 to June 2000.This study
involved 139 ICUs, 51 of which (37%) had MRSA infections. Outbreaks (three or
more MRSA infections within 3 months) were registered in 13 ICUs, clusters (two
MRSA infections within 3 months) in further 12 units and single events in 26 [7].
A publication from Spain showed that enterally administered vancomycin can
control endemic MRSA in ICUs without promoting VRE. This study was carried
out over a 49-month period from July 1996 to 2000 and published in 2004 [8].
In 2007, a report from Canada presented a recent outbreak of MRSA carriage in an
animal ICU. This finding appears important, as the strain responsible for the
animal outbreak was indistinguishable from a strain in humans commonly isolated
in Canada and the USA. Infection control measures, including active surveillance
of all animals in the ICU, were used to control the outbreak. As transmission of
MRSA within the unit occurred without infections and did not persist for a pro-
longed period of time, staff screening was surprisingly not initiated [9]. A paper
from China published in 2004 described an MRSA outbreak due to an increased
192 V. Damjanovic et al.
There have been ten outbreaks in AICUs published since 2000: eight were caused
by VRE, one was sensitive to vancomycin and one was sensitive to vancomycin
but resistant to linezolid. We selected seven reports and summarised them
according to the countries of origin and time of events and publishing.
A paper from Pakistan published in 2002 was the countrys first experience with
a vancomycin resistant Enterococcus faecium outbreak in the ICU and NICU. The
outbreak occurred in 2002, lasted 1 month and all but one isolate was of a single
clone [11]. All isolates were resistant to gentamicin, ampicillin and tetracycline
but sensitive to chloramphenicol. Six patients were colonised and four infected,
with positive blood cultures; two of each died before specific therapy could be
started (50% mortality rate). In 2005, a report from Italy described an outbreak of
VRE colonisation and infection in an ICU that lasted 16 months (20012002) [12].
Fifty-six patients were colonised by E. faecium, and E. faecalis was detected in
only two cases. Because of the low pathogenicity of VRE, the authors questioned
whether it was worthwhile to have a specific VRE surveillance programme. For
the 2004 Lowbury lecture, Pearman reported the Australian experience with VRE,
which he described as from disaster to ongoing control. This was the first
outbreak of VRE, which was caused by E. faecium in an ICU and hospital
wards and lasted 5 months in 2001. A vigilant VRE control programme
prevented the epidemic strain from becoming endemic in the hospital [13].
An outbreak due to glycopeptide-resistant enterococci (GRE) in an ICU with
simultaneous circulation of two different clones was reported from France in 2008.
The outbreak lasted several months in 2003 without infections, but the significant
colonisation caused organisational problems in the ICU [14]. An outbreak of
VRE in an ICU was reported from China in 2009. The outbreak was caused by
E. faecium and lasted 11 months (20062007). A detailed molecular analysis
showed that genetically unrelated isolates had transferred vancomycin resistance
by conjugation [15]. A paper from Korea reported an outbreak of VRE in a
neurological ICU. VRE was mainly isolated from urine specimens associated with
the presence of a Foley catheter. Of 52 patients colonised with VRE, only two had
active infection [16]. In 2009, a report from Spain presented an outbreak of lin-
ezolid-resistant E. faecalis in an ICU and reanimation unit [17]. This was the first
report of a clonal outbreak of linezolid-resistant E. faecalis in Spain. The strain
was sensitive to imipenem, vancomycin, teicoplanin and rifampicin. Most patients
were exposed to linezolid within a year (20052006). The use of linezolid began in
12 Outbreaks of Infection in the ICU 193
2002. The increase in its use continued until 2005 when a mutant was identified by
molecular analysis.
Fourteen reports on outbreaks were retrieved since 2000. Eight were caused
by Klebsiella pneumoniae, four by Serratia marcescens, one by Enterobacter
cloacae and one by simultaneous infection of E. cloacae and S. marcescens.
Three Klebsiella, three Serratia and the remaining two were selected for analysis.
We discuss Pseudomonas and Acinetobacter outbreaks separately.
An outbreak was reported from Germany in 2002 [22] in both the NICU and PICU,
lasted from September to November 1998 and involved 15 patients. Two epidemic
strains were associated with cross-infection in groups of five and ten patients,
respectively. Two epidemic clones were detected from the surfaces of an ICU room,
but an original source was not identified. The outbreak was stopped by routine
infection-control measures. A report from Malaysia in 2004 described an outbreak of
Serratia infections that lasted 10 days in an AICU [23]. The single outbreak strain
was found in insulin and sedative solutions administered to patients. An outbreak of
S. marcescens colonisation and infection in a neurological ICU that occurred from
May 2002 to March 2003 was reported from a Dutch university medical centre in
2006 [24]. The outbreak strain was traced to a healthcare worker (HCW) with long-
term carriage on the hands. The skin of the HCWs hands was psoriatic. The epidemic
ended after the colonised HCW went on leave, with subsequent eradication treatment.
A heterogeneous outbreak of E. cloacae and S. marcescens infections in a surgical
ICU was published by a group of authors from San Francisco, USA [25]. The outbreak
lasted from December 1997 through January 1998. Molecular techniques ruled out a
point source or significant cross-contamination as modes of transmission. The authors
concluded that patient-related factors, such as respiratory tract colonisation and
duration of central line placement might have played a role in this outbreak.
twice the national prevalence of 15% observed in ICUs. However, this high prev-
alence prompted the authors to conduct a prospective epidemiological study from
July 1997 to February 1998. We selected this study as a good example of activities
necessary to prevent a major outbreak. The authors described how systematic sur-
veillance was carried out (oropharyngeal and rectal swabs on admission and twice
weekly afterwards). This practice revealed that during the study period, the overall
incidence of P. aeruginosa carriage was 43%: 17% on admission and 26% acquired
in the ICU. In addition 16/191 (8%) patients developed the infection. The authors
also pointed out that intestinal carriage was a prerequisite for colonisation or
infection. Genotyping analysis of 81 isolates indicated that 70% belonged to geno-
type 1, 4% to genotype 2 and that remaining isolates were not genetically related. It
has also been shown that mechanical ventilation was associated with P. aeruginosa
carriage and ineffective antibiotics significantly increased the risk of colonisation
and infection in ICU. The authors concluded that not only do endogenous sources
account for the majority of colonisation or infection due to P. aeruginosa but that
exogenous sources may be involved in some instances. In an epidemic setting, the
authors stance was to reinforce standard barrier precautions. However, the main
message of this study is the necessity to adopt and pursue preventive measures.
In 2008, an outbreak of severe B. cepacia infections in an ICU was reported from
Spain [28]. The outbreak occurred over a period of 18 days in August 2006 when
B. cepacia were recovered from different clinical samples associated with bacter-
aemia in three cases, lower respiratory tract infection in one and urinary tract
infection in one. Samples of antiseptics, eau de cologne and moisturising milk
available on treatment carts were collected and cultured. B. cepacia was isolated not
only from three samples of the moisturising body milk that had been applied to the
patients but also from two new hermetically closed units. All strains recovered from
environmental and clinical samples belonged to the same clone. The cream was
withdrawn from all hospital units, and no new cases of B. cepacia developed. The
authors concluded that the presence of bacteria in cosmetic products, even within
accepted limits, may lead to severe life-threatening infections in severely ill patients.
died from sepsis despite treatment with a combination of meropenem and amikacin,
which were shown by laboratory tests to be synergistic. This high mortality rate (50%)
was explained by the authors as being due to persistent bacteraemia related to the
repeated infusions of contaminated solutions. Once aseptic preparation was carried out
in the hospital pharmacy, this outbreak was controlled, and further infusion-related
nosocomial bacteraemia was prevented. From the USA, a publication in 2001 reported
an outbreak of multiresistant Acinetobacter colonisation and infection in an ICU [30].
The strain was sensitive only to polymyxin. The outbreak lasted an entire year between
1996 and 1997 and involved 57 patients, 27 of whom were infected and 25 colonised.
The arrival of a colonised burn patient ([50% total body surface area) from an outside
hospital was responsible for the outbreak. Although on typing two strains were found,
the only identified primary source was the original burn patient. Ten deaths resulted
from infections (37% of infected patients). The authors claimed that this outbreak
served as a model of eradication of multi-drug-resistant organisms, as the
Acinetobacter was eliminated from all ICU patients by multidisciplinary measures that
included the following: cohort and contact isolation of all colonised and infected
patients; introduction of strict aseptic measures such as hand washing, barrier isolation,
equipment and room cleaning; sterilisation of ventilator equipment; and individual
dedication of medical equipment to each patient. A paper was published from Australia
in 2007 regarding carbapenem-resistant A. baumannii [31]. We selected this publi-
cation as an illustration of an extensive molecular analysis rather than for a critical
review of the outbreak, which occurred in an ICU between 1999 and 2000. Based on
their findings, the authors claim that antibiotic-resistant genes are readily exchanged
between co-circulating strains in epidemics of phenotypically indistinguishable
organisms. In conclusion, they recommend that epidemiological investigation of major
outbreaks should include whole-genome typing as well as analysis of potentially
transmissible genes and their vehicles. Finally, we found a paper in a journal from
Kuwait not found by our Internet research [32]. The authors reported three different
outbreaks of multi-drug-resistant A. baumannii infections involving 24 patients aged
1675 years that occurred in an ICU in the course of 1 year between 2006 and 2007.
The outbreak was polyclonal and successfully controlled with tigecycline, to which
two causative clones were sensitive. Three additional distinct clones were isolated
from the environment. Due to lack of appropriate surveillance cultures, no explanation
was offered for the origin of epidemic clones. Subsequently, in a letter to the editor, our
interpretation that microbial gut overgrowth increased spontaneous mutation,
which led to polyclonality and antibiotic resistance in the critically ill was accepted by
the authors [33, 34].
12.7 Discussion
Table 12.2 New and older trends at the beginning of the twenty-first century
of molecular techniques not only revealed a number of new genes responsible for
antibiotic resistance [18] but showed that genetically unrelated organisms readily
exchange antibiotic resistance genes [15, 31]. Yet further, a new trend is related to
the SDD concept. Two studies, one from Italy and one from Spain, reported the
use of enterally administered vancomycin to control and prevent, respectively,
MRSA outbreaks [5, 8]. This is further evidence that the principle of SDD can be
used with antimicrobials directed specifically to the causative organism. As early
as 1993 we reported how selective decontamination with nystatin successfully
controlled a Candida outbreak in an NICU [43].
Among older trends, surveillance cultures, or lack of them, are still prominent.
Even in 2009 there were authors responsible for infection control in hospitals and
ICUs who claimed that surveillance cultures of all patients with potential to
develop infection are difficult and very costly [44]. Some time ago (1994), we
expressed an alternative view in response to an identical attitude [45]. Needless to
say, lack of surveillance cultures not only delays the recognition of an outbreak
and its control but also precludes the understanding of the pathogenesis of
the majority of outbreaks. Surveillance cultures are also crucial for detecting
outbreaks of exogenous pathogenesis, i.e. without carriage. On the other hand, the
source of an exogenous outbreak is readily identified with molecular techniques.
Some of these outbreaks are striking, such as one from this analysis in which
Acinetobacter-contaminated parentally administered solutions were repeatedly
infused to patients, leading to a very high mortality rate of 50% [29].
In conclusion, new trends as well as old confirm what we indicated in the
previous edition of this book, which is that to control and prevent ICU outbreaks,
surveillance cultures and SDD should be integrated in routine infection-control
measures.
References
1. El-Masri MM, Williams KM, Fox-Wasylyshyn SM (2004) Severe acute respiratory
syndrome: another challenge for critical care nurses. AACN Clin Issues 15:150159
2. Burns SM (2009) H1N1 influenza is here. J Hosp Infect 73:200202
3. Vincent JL, Rello J, Marshall J et al (2009) International study of the prevalence and outcome
of infection in intensive care units. JAMA 302:23232329
4. Damjanovic V, Corbella X, van der Spoel JI, van Saene HKF (2005) Outbreaks of infection
in intensive care unitsusefulness of molecular techniques for outbreak analysis. In: van
Saene HKF, Silvestri L, de la Cal MA (eds) Infection control in the intensive care unit, 2nd
edn. Springer, Milan, pp 247296
5. Silvestri L, Milanese M, Oblach L et al (2002) Enteral vancomycin to control methicillin-
resistant Staphylococcus aureus outbreak in mechanically ventilated patients. Am J Infect
Control 30:391399
6. Cassone M, Campanile F, Pantosti A et al (2004) Identification of a variant Rome clone of
methicillin-resistant Staphylococcus aureus with decreased susceptibility to vancomycin,
responsible for an outbreak in an intensive care unit. Microb Drug Res 10:4349
7. Gastmeier P, Sohr D, Geffers C et al (2002) Occurrence of methicillin-resistant
Staphylococcus aureus infections in German intensive care units. Infection 4:198202
12 Outbreaks of Infection in the ICU 201
8. de la Cal MA, Cerda E, van Saene HKF et al (2004) Effectiveness and safety of enteral
vancomycin to control endemicity of methicillin-resistant Staphylococcus aureus in a
medical/surgical intensive care unit. J Hosp Infect 56:175183
9. Weese JS, Faires M, Rousseau J et al (2007) Cluster of methicillin-resistant Staphylococcus
aureus colonisation in a small animal intensive care unit. JAMA 231:13611364
10. Yap FH, Gomersall CD, Fung KS et al (2004) Increase in methicillin-resistant
Staphylococcus aureus acquisition rate and change in pathogen pattern associated with an
outbreak of severe acute respiratory syndrome. Clin Infect Dis 39:511516
11. Khan E, Sarwari A, Hasan R et al (2002) Emergence of vancomycin-resistant Enterococcus
faecium at a tertiary care hospital in Karachi, Pakistan. J Hosp Infect 52:292296
12. Peta M, Carretto E, Bdbarini D et al (2006) Outbreak of vancomycin-resistant Enterococcus
spp. on an Italian general intensive care unit. Clin Microbiol Infect 12:163169
13. Pearman JW (2006) 2004 Lowbury lecture: the Western Australian experience with
vancomycin-resistant enterococcifrom disaster to ongoing control. J Hosp Infect 63:1426
14. Delamare C, Lameloise V, Lozniewski A et al (2008) Glycopeptide-resistant Enterococcus
outbreak in an ICU with simultaneous circulation of two different clones. Pathol Biol 56:454460
15. Zhu X, Zeng B, Wang S et al (2009) Molecular characterization of outbreak-related strains of
vancomycin-resistant Enterococcus faecium from an intensive care unit in Beijing, China.
J Hosp Infect 72:147154
16. Se BY, Chun HJ, Yi HJ et al (2009) Incidence and risk factors of infections caused by
vancomycin-resistant Enterococcus colonization in neurosurgical intensive care unit patients.
J Korean Neurosurg Soc 46:123129
17. Gomez-Gil R, Romero-Gomez MP, Garcia-Arias A et al (2009) Nosocomial outbreak of
linezolid-resistant Enterococcus faecalis infection in a tertiary care hospital. Diag Microb
Infect Dis 65:175179
18. Brinas L, Lantero M, Zarazaga M et al (2004) Outbreak of SHV-5 beta-lactamase-producing
Klebsiella pneumoniae in a neonatalpediatric intensive care unit in Spain. Microb Drug Res
10:354358
19. vant Veen A, van der Zee A, Nelson J et al (2005) Outbreak of infection with a multiresistant
Klebsiella pneumoniae strain associated with contaminated roll boards in operating rooms.
J Clin Microbiol 43:49614967
20. Laurent C, Rodriguez-Villalobos H, Rost F et al (2008) Intensive care unit outbreak of
extended-spectrum beta-lactamase-producing Klebsiella pneumoniae controlled by cohorting
patients and reinforcing infection control measures. Infect Control Hosp Epidemiol 29:
517524
21. Manzur A, Tubau F, Pujol M et al (2007) Nosocomial outbreak due to extended-spectrum-
beta-lactamase-producing Enterobacter cloacae in a cardiothoracic intensive care unit. J Clin
Microbiol 45:23652369
22. Steppberger K, Walter S, Claros MC et al (2002) Nosocomial neonatal outbreak of Serratia
marcescensanalysis of pathogens by pulsed field gel electrophoresis and polymerase chain
reaction. Infection 30:277281
23. Alfizah H, Nordiah AJ, Rozaidi WS (2004) Using pulsed-field gel electrophoresis in the
molecular investigation of an outbreak of Serratia marcescens infection in an intensive care
unit. Singap Med J 45:214218
24. de Vries JJ, Bass WH, van der Ploeg K et al (2006) Outbreak of Serratia marcescens
colonization and infection traced to a healthcare worker with long-term carriage on the hands.
Infect Control Hosp Epidemiol 27:11531158
25. Dorsey G, Borneo HT, Sun SJ et al (2000) A heterogeneous outbreak of Enterobacter
cloacae and Serratia marcescens infections in a surgical intensive care unit. Infect Control
Hosp Epidemiol 21:465469
26. Bukholm G, Tannaes T, Kjelsberg AB et al (2002) An outbreak of multidrug-resistant
Pseudomonas aeruginosa associated with increased risk of patient death in an intensive care
unit. Infect Control Hosp Epidemiol 23:441446
202 V. Damjanovic et al.
13.1 Introduction
L. Silvestri (&)
Department of Emergency, Unit of Anesthesia and Intensive Care,
Presidio Ospedaliero di Gorizia, Gorizia, Italy
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 203
DOI: 10.1007/978-88-470-1601-9_13, Springer-Verlag Italia 2012
204 L. Silvestri et al.
To control the
efficacy of enteral
antimicrobials
Surveillance
To classify infections
cultures according to the carrier state
To identify a resistance
problem
Fig. 13.1 Stoutenbeeks tetralogy of SDD and type of infection prevented/controlled by each
component. SDD selective decontamination of the digestive tract; PPM potentially pathogenic
microorganism; ICU intensive care unit
13 Preventing Infection Using Selective Decontamination of the Digestive Tract 205
Table 13.1 The full four-component protocol of selective decontamination of the digestive tract
13.2 Efficacy
After 25 five years of clinical research, SDD has been assessed in 63 randomized
controlled trials (RCTs) [365] and ten meta-analyses of RCTs only (Table 13.2)
[6675].
13.2.1 Carriage
SDD significantly reduced oropharyngeal carriage by 87% [odds ratio (OR) 0.13,
95% confidence interval (CI) 0.070.2] and rectal carriage due to Gram-negative
PPMs by 85% (OR 0.15; 95% CI 0.070.31) [72]. Gram-positive carriage was also
reduced, but not significantly. Additionally, fungal carriage was significantly
reduced by 68% (OR 0.32, 95% CI 0.190.53) [70].
Table 13.2 Efficacy of selective decontamination of the digestive tract assessed in ten meta-
analyses of randomized controlled trials only
13.2.5 Mortality
Mortality rate was an outcome measure in eight of the ten meta-analyses [6669,
71, 7375]. There was a consistent survival benefit in all meta-analyses that
assessed the full four-component SDD protocol providing the sample size was
large enough [67, 68, 71, 73, 74]. The Italian meta-analysis, which assessed only
RCTs in which the full SDD protocol was used, showed a mortality rate reduction
of 29% (OR 0.71, 95% CI 0.610.82) [73]. This effect achieved a 42% mortality
rate reduction in studies where SDD eradicated the carrier state (OR 0.58, 95% CI
0.450.77) [73]. Eighteen patients need to be treated with the full SDD protocol to
prevent one death [73, 74]. The meta-analyses of Vandenbroucke-Grauls and
Vandenbroucke [66], Safdar et al. [69], and Silvestri et al. [75] showed an impact
on mortality rate that was not significant due to the small sample size. Two Dutch
RCTs with the primary endpoint of mortality have been published. In the first [17],
the randomization unit was the ICU and not the patient and included about 1,000
208 L. Silvestri et al.
patients. The risk of mortality was significantly reduced by 40% in the unit in
which SDD was administered to all patients (OR 0.6; 95% CI 0.40.8). The second
[19] is the largest study on SDD ever published and included about 6,000 patients.
The primary endpoint was mortality, whereas resistance was among the secondary
endpoints. The study compared SDD, selective oropharyngeal decontamination
(SOD), a modified SDD protocol without the gut component and the parenterally
administered antibiotic, and standard care. Both SDD and SOD significantly
reduced the odds of death compared with standard care [OR 0.83 (p = 0.02), and
0.86 (p = 0.045), respectively]. However, mortality rate reduction was higher,
albeit not significantly, in the SDD group than in the SOD group. These results
regarding SOD have been confirmed by a recent meta-analysis of the nine RCTs
using SOD and including 4,733 patients [76]. Although SOD has been shown to
significantly reduce the odds of pneumonia, the meta-analysis failed to demon-
strate any significant impact on survival (OR 0.93; 95% CI 0.811.07). Addi-
tionally, SOD has been shown to be associated with a 33% and SDD with a 45%
reduction in ICU-acquired Gram-negative bacteremia [77], explaining why SDD,
and not SOD, is associated with a significant mortality rate reduction.
13.2.6 Miscellaneous
13.3 Safety
The use of parenterally administered antibiotics has been shown to lead to the
emergence of antimicrobial resistance, which has not been shown in RCTs of
SDD [81]. This may be explained by the fact that the addition of enterally
administered antibiotics to the parenterally administered antibiotics may have
kept the systemic agents useful. An intriguing aspect of 25 years of clinical
research in SDD is the experience that the pre-1980s antibiotics, such as cefo-
taxime, are still active as long they are combined with successful eradication of
AGNB from the gut.
Resistance was the endpoint of three RCTs of SDD [13, 17, 19]. A Klebsiella
pneumoniaeproducing extended-spectrum beta-lactamase was endemic in a
French hospital [13]: carriage and infection rates were 19.6 and 9%, respectively.
Once enterally administered antimicrobials were added to those administered
parenterally, there was a significant reduction in both carriage and infection (19.6
vs. 1%; 9 vs. 0%). A Dutch single-center RCT of about 1,000 patients reported that
carriage of AGNB resistant to imipenem, ceftazidime, ciprofloxacin, tobramycin,
and polymyxins occurred in 16% of patients receiving parenterally and enterally
administered antimicrobials compared with 26% of control patients who received
antibiotics parenterally only, with a relative risk of 0.6 (95% CI 0.50.8) [17]. The
largest multicenter RCT to date is also from The Netherlands and comprised about
6,000 patients [19]. The proportion of patients with AGNB shown in rectal swabs
that were not susceptible to the marker antibiotics was lower with SDD than with
standard care or SOD. For example, carriage of multi-drug-resistant Pseudomonas
aeruginosa was 0.4% in SDD versus 0.8% in SOD and 1.3% in the group receiving
standard care (p \ 0.005). Moreover, the study authors reported in a separate
analysis of the same RCT results on bacteremia and lower respiratory tract col-
onization due to highly resistant microorganisms (HRMO), namely aerobic Gram-
negative bacilli [82]. Bacteremia due to HRMO was significantly reduced by SDD
compared with SOD (OR 0.37; 95% CI 0.160.85). Lower respiratory tract
colonization due to HRMO was less with SDD (OR 0.58; 95% CI 0.430.78) than
with SOD (OR 0.65; 95% CI 0.490.87) compared with standard care. Therefore,
SDD was superior to SOD and to standard care in preventing antimicrobial
resistance.
In an ecological study [83] conducted during the study periods of the Dutch
RCT [19], an increase in resistance after discontinuation of SOD and SDD was
observed, which seems to contradict the reduction in resistance. However, that
ecological analysis has an important limitation, i.e., the use of a point-prevalence
survey in which all patients in the unit (whether enrolled in the SDD or SOD trial)
were included. Moreover, the average prevalence of AGNB resistant to ceftazi-
dime, tobramycin, and ciprofloxacin in the respiratory tract was significantly lower
during SDD/SOD than the pre- and post-intervention periods, and AGNB resis-
tance to ciprofloxacin and tobramycin in rectal swabs was significantly reduced
during SDD compared with standard care/SOD [84, 85].
210 L. Silvestri et al.
The target microorganisms of SDD include PPMs belonging to the normal flora,
including S. pneumoniae and MSSA, as well as the opportunistic aerobic Gram-
negative bacilli, including Klebsiella, Acinetobacter, and Pseudomonas spp.
Methicillin-resistant S. aureus (MRSA), by design, is not covered by the original
SDD protocol, and hence, six randomized trials conducted in ICUs in which
MRSA was endemic at the time of the study showed a trend toward higher MRSA
infection rates in patients receiving SDD. These observations suggest that the
parenterally and enterally administered antimicrobials of the SDD protocol, i.e.,
cefotaxime, polymyxin, tobramycin, and amphotericin B, may select for and
promote MRSA. Under these circumstances, SDD requires the addition of oro-
pharyngeally and intestinally administered vancomycin. Two studies showed that
adding vancomycin to SDD is an effective and safe maneuver [86, 87].
SDD is not active against vancomycin-resistant enterococci (VRE). All SDD
randomized trials were undertaken in ICU and hospital settings without VRE
experience. SDD was evaluated in two observational studies undertaken in ICU
with a low VRE prevalence [87, 88]. In the Spanish study, VRE was imported into
the unit, but no change in policy was required, as extensive spread did not occur
[87]. In the American study, SDD was evaluated in a unit with a low incidence of
VRE, and the authors reported that SDD did not increase the incidence of VRE
carriage and infection [88].
13.4 Conclusions
SDD is the only evidence-based maneuver that prevents infection in the critically
ill. It significantly reduces lower respiratory tract infections, bloodstream
infections, multiple organ failure, mortality rates, and resistance if the full four-
component protocol is used. SOD only significantly reduces pneumonia but not
mortality rates. Moreover, the full SDD protocol significantly reduces intestinal
carriage of multi-drug-resistant aerobic Gram-negative microorganisms, thus
reducing the occurrence of ICU-acquired bacteremia.
References
1. Stoutenbeek CP, van Saene HKF, Miranda DR, Zandstra DF (1984) The effect of selective
decontamination of the digestive tract on colonization and infection rate in multiple trauma
patients. Intensive Care Med 10:185192
2. 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 29:677690
3. Abdel-Razek SM, Abdel-Khalek AH, Allam AM et al (2000) Impact of selective
gastrointestinal decontamination on mortality and morbidity in severely burned patients.
Ann Burns Fire Disasters 13:213215
4. Abele-Horn M, Dauber A, Bauernfeind A et al (1997) Decrease in nosocomial pneumonia in
ventilated patients by selective oropharyngeal decontamination (SOD). Intensive Care Med
23:18781895
13 Preventing Infection Using Selective Decontamination of the Digestive Tract 211
5. Aerdts SJA, van Dalen R, Clasener HAL et al (1991) Antibiotic prophylaxis of respiratory
tract infection in mechanically ventilated patients. A prospective, blinded, randomized trial of
the effect of a novel regimen. Chest 100:783791
6. Arnow PA, Caradang GC, Zabner R et al (1996) Randomized controlled trial of selective
decontamination for prevention of infections following liver transplantation. Clin Infect Dis
22:9971003
7. Barret JP, Jeschke MG, Herndon DN (2001) Selective decontamination of the digestive tract
on severely burned pediatric patients. Burns 27:439445
8. Bergmans DCJJ, Bonten MJM, Gaillard CA et al (2001) Prevention of ventilator-associated
pneumonia by oral decontamination. A prospective, randomized, double-blind, placebo-
controlled study. Am J Respir Crit Care Med 164:382388
9. 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 22:4049
10. Blair P, Rowlands BJ, Lowry K et al (1991) Selective decontamination of the digestive tract: a
stratified, randomized, prospective study in a mixed intensive care unit. Surgery 110:303310
11. Boland JP, Sadler DL, Stewart W et al (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]. 17th
congress of chemotherapy, Berlin, N0465
12. Bouter H, Schippers EF, Luelmo SAG et al (2002) No effect of preoperative selective gut
decontamination on endotoxemia and cytokine activation during cardiopulmonary bypass: a
randomized, placebo-controlled study. Crit Care Med 30:3843
13. Brun-Buisson C, Legrand P, Rauss A et al (1989) Intestinal decontamination for control of
nosocomial multiresistant gram-negative bacilli. Study of an outbreak in an intensive care
unit. Ann Intern Med 110:873881
14. Camus C, Bellisant E, Sebille V et al (2005) Prevention of acquired infections in
intubated patients with the combination of two decontamination regimens. Crit Care Med
33:307314
15. Cerra FB, Maddaus MA, Dunn DL et al (1992) Selective gut decontamination reduces
nosocomial infections and length of stay but not mortality or organ failure in surgical
intensive care unit patients. Arch Surg 127:163169
16. 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 117:545553
17. de Jonge E, 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: a
randomised controlled trial. Lancet 363:10111016
18. de la Cal MA, Cerd E, Garcia-Hierro P et al (2005) Survival benefit in critically ill burned
patients receiving selective decontamination of the digestive tract. A randomized, placebo-
controlled, double-blind trial. Ann Surg 241:424430
19. de Smet AMGA, Kluytmans JA, Cooper BS et al (2009) Decontamination of the digestive
tract and oropharynx in ICU patients. N Engl J Med 360:2031
20. Diepenhorst GMP, van Ruler O, Besselink MGH et al (2011) Influence of prophylactic
probiotics and selective decontamination on bacterial translocation in patients undergoing
pancreatic surgery. Shock 35:916
21. Farran L, Llop J, Sans M et al (2008) Efficacy of enteral decontamination in the prevention of
anastomotic dehiscence and pulmonary infection in esophagogastric surgery. Dis Esophagus
21:159164
22. Ferrer M, Torres A, Gonzalez J et al (1994) Utility of selective decontamination in
mechanically ventilated patients. Ann Intern Med 120:389395
23. Finch RG, Tomlinson P, Holliday M et al (1991) Selective decontamination of the digestive
tract (SDD) in the prevention of secondary sepsis in a medical/surgical intensive care unit
[abstract]. 17th congress of chemotherapy, Berlin, N0471
212 L. Silvestri et al.
24. Flaherty J, Nathan C, Kabins SA et al (1990) Pilot trial of selective decontamination for
prevention of bacterial infection in an intensive care unit. J Infect Dis 162:13931397
25. Gastinne H, Wolff M, Delatour F et al (1992) A controlled trial in intensive care units of
selective decontamination of the digestive tract with nonabsorbable antibiotics. N Engl J Med
326:594959
26. Gaussorgues Ph, Salord F, Sirodot M et al (1991) Efficacit de la dcontamination digestive
sur la survenue des bactrimies nosocomiales chez les patients sous ventilation mcanique et
recevant des btamimtiques. Ranimation Soins Intensive et Mdicine dUrgence 7:169174
27. Georges B, Mazerolles M, Decun J-F et al (1994) Decontamination digestive selective:
resultats dune etude chez le polytraumatise. Reanim Urgences 3:621627
28. Gosney M, Martin MV, Wright AE (2006) The role of selective decontamination of the
digestive tract in acute stroke. Age Aging 35:4247
29. Hammond JMJ, Potgieter PD, Saunders GL et al (1992) Double-blind study of selective
decontamination of the digestive tract in intensive care. Lancet 340:59
30. Hellinger WC, Yao JD, Alvarez S et al (2002) A randomized, prospective, double blinded
evaluation of selective bowel decontamination in liver transplantation. Transplantation
73:19041909
31. 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
Care 3:5258
32. Kerver AJH, Rommes JH, Mevissen-Verhage EAE et al (1988) Prevention of colonization
and infection in critically ill patients: a prospective randomized study. Crit Care Med
16:10871093
33. Korinek AM, Laisne MJ, Nicolas MH et al (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
34. 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, double-blind,
placebo-controlled clinical trial. Am J Respir Crit Care Med 166:10291037
35. 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 106:1519
36. Lingnau W, Berger J, Javorsky F et al (1997) Selective intestinal decontamination in multiple
trauma patients: prospective, controlled trial. J Trauma 42:687694
37. Luiten EJT, Hop WCJ, Lange JF et al (1995) Controlled clinical trial of selective
decontamination for the treatment of severe acute pancreatitis. Ann Surg 222:5765
38. Martinez-Pellus AE, Merino P, Bru M et al (1993) Can selective digestive decontamination
avoid the endotoxemia and cytokine activation promoted by cardiopulmonary bypass? Crit
Care Med 21:16841691
39. Martinez-Pellus AE, Merino P, Bru M et al (1997) Endogenous endotoxemia of intestinal
origin during cardiopulmonary bypass. Role of type of flow and protective effect of selective
digestive decontamination. Intensive Care Med 23:12511257
40. Oudhuis GJ, Bergmans DG, Dormans T et al (2011) Probiotics versus antibiotic
decontamination of the digestive tract: infection and mortality. Intensive Care Med
37:110117
41. Palomar M, Alvarez-Lerma F, Jord R et al (1997) Prevention of nosocomial infection in
mechanically ventilated patients: selective digestive decontamination versus sucralfate. Clin
Intensive Care 8:228235
42. Pneumatikos I, Koulouras V, Nathanail C et al (2002) Selective decontamination of
subglottic area in mechanically ventilated patients with multiple trauma. Intensive Care Med
28:432437
13 Preventing Infection Using Selective Decontamination of the Digestive Tract 213
79. Silvestri L, de la Cal MA, Taylor N et al (2010) Selective decontamination of the digestive
tract in burn patients: an evidence-based maneuver that reduces mortality. J Burn Care Res
31:372373
80. Silvestri L, van Saene HKF (2010) Selective digestive decontamination to prevent
pneumonia after esophageal surgery. Ann Thor Cardiovasc Surg 16(3):220221
81. Silvestri L, van Saene HKF (2006) Selective decontamination of the digestive tract does not
increase resistance in critically ill patient. Evidence from randomized controlled trials. Crit
Care Med 34:20272029
82. de Smet AM, Kluytmans J, Blok H et al (2010) Effects of selective digestive and selective
oropharyngeal decontamination on bacteraemia and respiratory tract colonization with highly
resistant micro-organisms. Clin Microbiol Infect 16(suppl 2):S98
83. Oodstijk EAN, de Smet AMGA, Blok HEM et al (2010) Ecological effects of selective
decontamination on resistant Gram-negative bacterial colonization. Am J Respir Crit Care
Med 181:452457
84. Zandstra DF, Petros AJ, Taylor N et al (2010) Withholding selective decontamination of the
digestive tract from critically ill patients must now surely be ethically questionable given the
vast evidence base. Crit Care 14:443
85. Petros AJ, Taylor N, V Damjanovic et al (2010) Worlds apart; proof that SDD works. Am J
Respir Crit Care Med 182:1564
86. Silvestri L, Milanese M, Oblach L et al (2002) Enteral vancomycin to control methicillin-
resistant Staphylococcus aureus outbreak in mechanically ventilated patients. Am J Infect
Control 30:391399
87. de la Cal MA, Cerd E, van Saene HKF et al (2004) Effectiveness and safety of enteral
vancomycin to control endemicity of methicillin-resistant Staphylococcus aureus in a
medical/surgical intensive care unit. J Hosp Infect 56:175183
88. Bhorade SM, Christensen J, Pohlman AS et al (1999) The incidence of and clinical variables
associated with vancomycin-resistant enterococcal colonization in mechanically ventilated
patients. Chest 115:10851091
Part IV
Infections on ICU
Lower Airway Infection
14
J. Almirall, A. Liapikou, M. Ferrer
and A. Torres
14.1 Definition
A. Torres (&)
Servei de Pneumologia i Allrgia Respiratria,
Hospital Clnic, Barcelona, Spain
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 219
DOI: 10.1007/978-88-470-1601-9_14, Springer-Verlag Italia 2012
220 J. Almirall et al.
14.2 Pathogenesis
14.3 Epidemiology
Nosocomial pneumonia accounts for 31% of all nosocomial infections, and a large
majority (83%) of patients who develop nosocomial pneumonia are mechanically
ventilated. The exact incidence of VAP is difficult to obtain due to overlapping
lower RTIs and the difficulty in diagnosing VAP correctly. The incidence of VAP
ranges from 9 to 67% of patients on MV. The rate of VAP, expressed as the total
number of episodes of VAP/1,000 ventilator days, ranges from 5 to 16 [6]. VAP
can increase the time on a ventilator by 10 days, length of ICU stay by 6 days, and
length of total hospital stay by 11 days.
Disease incidence depends greatly on the type of population studied, the
presence or absence of risk factors for colonization by multi-drug-resistant
pathogens, and the type and intensity of preventive strategies applied. Tracheal
intubation and MV are the main risk factors for VAP during the first week of
ventilation (risk assessed at approximately 3% per day in the first week of MV).
A one-day point-prevalence study conducted in 1,417 intensive care units (ICUs)
in Western Europe reported that VAP was the most common ICU-acquired
infection and MV was associated with a threefold increased risk of developing
pneumonia [7]. Studies conducted in several countries in the European Union
have shown varying incidence density ranging from approximately 925 cases/
1,000 ventilation days [6]. Epidemiological studies on a large United States
database with medical, surgical, and trauma patients have shown a VAP inci-
dence of 9.3%.
Hospital mortality rate of patients with VAP is significantly higher than that
of patients without VAP. Crude VAP mortality rates range between 20 and
50%, depending on comorbidities, illness severity, pathogens, and quality of
antibiotic treatment [1]. Ventilated ICU patients with VAP appear to have a
two- to tenfold higher risk of death compared with patients without pneumonia.
However, several patients with VAP die and not because of VAP. However,
mortality rates vary from one study to another, and the prognostic impact is
debated. It is well recognized that one-third to one-half of all VAP deaths are
directly attributable to the disease. Mortality rates are higher when VAP
associated with bacteremia, especially with P. aeruginosa or Acinetobacter
spp., medical rather than surgical illness, and treatment with ineffective anti-
biotic therapy [2].
VAP is associated with higher medical care costs. Patients who develop VAP
during a hospital stay remain longer in the ICU and the hospital, and the increased
level of care and need for additional invasive procedures drastically increases
healthcare costs. It has been reported that each case of VAP is associated with
additional hospital costs of $20000 to more than US $40000. Infection with MRSA
increases hospital costs by an additional $7731 per patient. These data emphasize
the need for prevention and better outcomes [8].
222 J. Almirall et al.
The high morbidity and mortality rates of VAP and the costs of the disease, both in
terms of treatment and increasing hospital length of stay, have led to efforts to
reach consensus in control measures and prevention. Many hospitals have devel-
oped and implemented evidence-based prevention protocols and educational
programs for physicians and nurses. These strategies have often improved quality
of care and reduced VAP incidence. When North American epidemiological data
from the 2008 National Healthcare Safety Network (NHSN) report are compared
with data from the 2003 National Nosocomial Infections Surveillance (NNIS),
pneumonia incidence densities are slightly lower overall, suggesting that new
preventive strategies applied in the meantime have had a positive effect [13].
224 J. Almirall et al.
Maintain semirecumbent
position
Supine positioning Maintain oral hygiene
Nasogastric tube Use continuous subglottic
suctioning
Large gastric volumes Avoid unplanned
extubations
Aspiration of
contaminated Patient/ventilator circuit Routinely drain circuit
condensate/aerosols into
lower airways
Fig. 14.1 Relationship between pathogenesis, risk factors, and preventive strategies for
ventilator-associated pneumonia (VAP)
226 J. Almirall et al.
There are no consistent data showing reduced VAP incidence [2] and better
outcome using either heat and moisture exchangers (HME) or heated humidifiers
(HH). Neither humidification strategy can be recommended as a pneumonia pre-
vention tool at this stage; however, inspiratory gases should be delivered at body
temperature or slightly below and at the highest relative humidity in order to
prevent heat and moisture loss from the airways and, more importantly, change in
rheologic properties of secretions and impairment of mucociliary clearance.
Enterally administered nutrition in supine patients is a risk factor for VAP devel-
opment through increased risk of aspiration of gastric contents. Residual volume
should be carefully monitored and, in the case of consistently large volumes, the use
of agents that increase gastrointestinal (GI) motility (e.g., metoclopramide). When
necessary, enterally administered nutrition should be withheld to reduce aspiration
risk. Enterally administered nutrition acidification and postpyloric tube placement
and nutrition suspension 8 h daily (intermittent nutrition) are strategies that should
reduce gastric colonization and risk of gastroesophageal reflux, although investi-
gators have reported inconsistent results [19]. However, the effectiveness of such
interventions awaits validation in clinical trials. Nevertheless, intubated patients
should be kept in a semirecumbent position (3045) to prevent aspiration, especially
when receiving enterally administered nutrition.
higher risk for GI bleeding during MV; hence, stress-ulcer prophylaxis is essential.
Antacids, histamine-2-receptor antagonists (H2 blockers), and proton-pump
inhibitors (PPI) are usually administered to prevent GI lesions. Sucralfate, an
alternative gastroprotective agent, does not change gastric acidity and prevents GI
bleeding, protecting gastric mucosa. Several randomized clinical trials and
meta-analyses investigated the rates of VAP using sucralfate versus agents that
alkalinize gastric juice (antacids, H2 blockers, PPI) with conflicting results. An
additional risk for GI bleeding using sucralfate has also been found [20]. Thus, the
use of sucralfate as VAP-preventive strategy should only be recommended in
patients with low risk of GI bleeding.
14.6.8 Probiotics
Probiotics are viable microorganisms that colonize the host GI tract by adhering
to the intestinal mucosa and compete with the adhesion of pathogens to epithelial
binding sites, thus creating an unfavorable local milieu for pathogen coloniza-
tion. Probiotic products have been shown to be of some benefit in the following
diseases: acute infectious diarrhea in children, necrotizing enterocolitis in very-
low-birth-weight infants, allergic atopic dermatitis prevention in children, and
prevention of relapses of ulcerative colitis. In critically ill patients, studies
demonstrate that oral administration of a probiotic Lactobacillus preparation
delayed respiratory tract colonization with P. aeruginosa and resulted in a
reduced rate of ventilator-associated pneumonia caused by P. aeruginosa. Also,
Morrow et al. [23] found that patients treated with Lactobacillus were signifi-
cantly less likely to develop microbiologically confirmed VAP compared with
patients treated with placebo (40.0 vs. 19.1%, p = 0.007). A meta-analysis of
five randomized controlled trials concluded that probiotic administration is
associated with lower incidence of VAP [24]. Future studies need to be designed
with standardization of the probiotic product and dosing (both daily dose and
therapy duration).
References
1. Chastre J, Fagon JY (2002) Ventilator-associated pneumonia. Am J Respir Crit Care Med
165:867903
2. Niederman MS, Craven DE, Bonten MJ (2005) American Thoracic Society and Infectious
Diseases Society of America (ATS/IDSA) Guidelines for the management of adults with
hospital-acquired, ventilator-associated, and healthcare-associated pneumonia. Am J Respir
Crit Care Med 171:388416
3. Garner JS, Jarvis WR, Emori TG et al (1998) CDC definitions for nosocomial infections
1998. Am J Infect Control 16:128140
4. Nseir S, Favory R, Jozefowicz E, VAT Study Group et al (2008) Antimicrobial treatment for
ventilator-associated tracheobronchitis: a randomized, controlled, multicenter study. Crit
Care 12:R62
5. Torres A, Ewig S, Lode H et al (2009) Defining, treating and preventing hospital acquired
pneumonia: European perspective. Intensive Care Med 35:929
6. National Nosocomial Infections Surveillance (NNIS) (2004) System Report, data summary
from January 1992 through June 2004, issued October 2004. Am J Infect Control 32:470485
7. Vincent JL, Bihari DJ, Suter PM et al (1995) The prevalence of nosocomial infection in
intensive care units in Europe. Results of the European prevalence of infection in intensive
care (EPIC) study. EPIC international advisory committee. JAMA 274:639644
8. Rello J, Ollendorf DA, Oster G et al (2002) Epidemiology and outcomes of ventilator-
associated pneumonia in a large US database. Chest 122:21152121
9. Fagon JY, Chastre J, Wolff M et al (2000) Invasive and noninvasive strategies for
management of suspected ventilator-associated pneumonia. Ann Intern Med 132:621630
10. Kollef MH, Morrow LE, Niederman MS et al (2006) Clinical characteristics and treatment
patterns among patients with ventilator-associated pneumonia. Chest 129:12101218
11. Agbaht K, Diaz E, Muoz E et al (2007) Bacteremia in patients with ventilator-associated
pneumonia is associated with increased mortality: a study comparing bacteremic vs
nonbacteremic ventilator-associated pneumonia. Crit Care Med 35:20642070
12. Kollef MH, Afeas B, Anzuelo A et al (2008) NASCENT investigation group silver-coated
endotracheal tubes and incidence of ventilator-associated pneumonia: the NASCENT
randomized trial. JAMA 300:805813
13. Rosenthal VD, Maki DG, Jamulitrat S, INICC members et al (2010) International nosocomial
infection control consortium (INICC) report, data summary for 20032008, issued June 2009.
Am J Infect Control 38(2):95104.e2
14. Resar R, Pronovost P, Haraden C et al (2005) Using a bundle approach to improve ventilator
care processes and reduce ventilator-associated pneumonia. Jt Comm J Qual Patient Saf
31:243248
15. Torres A, Gatell JM, Aznar E et al (1995) Re-intubation increases the risk of nosocomial
pneumonia in patients needing mechanical ventilation. Am J Respir Crit Care Med 152:
137141
232 J. Almirall et al.
15.1 Introduction
J. Valls (&)
Critical Care Center, Hospital Sabadell,
Sabadell, Barcelona, Spain
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 233
DOI: 10.1007/978-88-470-1601-9_15, Springer-Verlag Italia 2012
234 J. Valles and R. Ferrer
70
60
50
40
30
20
10
0
2000 2001 2002 2003 2004 2005 2006 2007 2008 2009
Fig. 15.1 Distribution of bloodstream infections (BSI) in the medicalsurgical intensive care
unit (ICU) of Hospital Sabadell (20002009) HAI-ICU, hospital-acquired BSI in ICU; HAI-non
ICU, hospital-acquired BSI in wards; CA, community-acquired BSI
15.2.1 Epidemiology
HA BSI in the ICU is defined in a patient with a clinically significant blood culture
positive for a bacterium or fungus and that is obtained more than 72 h after
admission or previously, if it is directly related to a invasive manipulation on
admission in the ICU (e.g., urinary catheterization or insertion of intravenous line)
[13]. Patients in the ICU not only have higher endemic rates of HAI than patients
in general wards, but the distribution of their infections also differs. The two most
important HAI in general wards are urinary tract and surgical wound infections,
whereas in the ICU, lower respiratory tract infections and BSI are the most
frequent [14]. This distribution is related to the widespread use of mechanical
ventilation and intravenous catheters. Data compiled through the national noso-
comial infections surveillance system (NNIS) of the centers for disease control and
prevention (CDC) in the USA revealed that bloodstream infections accounted for
almost 20% of HAI in ICU patients, 87% of which were associated with a central
line [15]. A recent nationwide surveillance study in 49 US hospitals (SCOPE)
reported that 51% of HA BSIs occurred in the ICU [16]. Studies conducted in
critically ill patients show that the incidence rate of nosocomial BSI in the ICU
ranges from 27 to 68 episodes per 1,000 admissions [1721] (Table 15.1),
depending on the type of ICU (surgical, medical, coronary care unit), severity of
patients illness, use of invasive devices, and the length of ICU stay. These
infection rates among ICU patients are as much as five to ten times higher than
those recorded for patients admitted to general wards.
15 Bloodstream Infection in the ICU Patient 235
Table 15.1 Rates of hospital-acquired bloodstream infections (BSIs) in the intensive care
unit (ICU)
Conditions that predispose an individual to BSI include not only the patients
underlying conditions but also therapeutic, microbial, and environmental factors.
Illnesses that have been associated with an increased BSI risk include hematologic
and nonhematologic malignancies, diabetes mellitus, renal failure requiring dial-
ysis, chronic hepatic failure, immune deficiency syndromes, and conditions
associated with the loss of normal skin barriers, such as serious burns and pressure
ulcers. In the ICU, therapeutic maneuvers associated with an increased risk of HA
BSI include procedures such as placement of intravascular and urinary catheters,
endoscopic procedures, and drainage of intra-abdominal infections. Several risk
factors have been associated with the acquisition of BSI by specific pathogens.
Coagulase-negative staphylococci are mainly associated with central venous line
infection and with the use of intravenously administered lipid emulsions. Candida
spp. infections are related to exposure to multiple antibiotics, hemodialysis,
isolation of Candida spp. from sites other than the blood, azotemia, and the use of
indwelling catheters [22]. In an analysis of risk factors for HA candidemia in our
ICU, we found that exposure to more than four antibiotics during the ICU stay
[odds ratio (OR) 4.10], parenterally administered nutrition (OR 3.37), previous
surgery (OR 2.60), and the presence of solid malignancy (OR 1.57) were the
variables that were independently associated with the development of Candida
spp. infection [23].
15.2.3 Microbiology
The spectrum of microorganisms that invade the bloodstream in patients with HAI
during their stay in the ICU has been evaluated in several studies. Although almost
any microorganism can produce BSI, staphylococci and Gram-negative bacilli
account for the vast majority of cases. However, among the staphylococci, coag-
ulase-negative staphylococci (CNS) have become a clinically significant agent of
BSIs in the ICU [17, 18, 24, 25]. The ascendance of this group of staphylococci has
increased the interpretative difficulties for clinicians, as a high number of CNS
236 J. Valles and R. Ferrer
Table 15.2 Microorganisms causing nosocomial bloodstream infection in adult intensive care
units
isolations represent contamination rather than true BSI. The increased importance
of CNS BSI seems to be related to the high incidence of multiple invasive devices
used in critically ill patients and to the multiple antimicrobial therapies used for
Gram-negative infections in ICU patients, which results in selection of Gram-
positive microorganisms. The change in the spectrum of organisms causing HA
BSIs in an adult ICU is confirmed by Edgeworth et al. [26], who analyzed the
evolution of HA BSIs over 25 years in the same ICU. Between 1971 and 1990, the
frequency of isolation of individual organisms changed little, with S. aureus,
P. aeruginosa, Escherichia coli, and Klebsiella pneumoniae predominating.
However, between 1991 and 1995, the number of BSIs doubled, largely due to the
increased isolation of CNS, Enterococcus spp., and intrinsically antibiotic-resistant
Gram-negative organisms, particularly P. aeruginosa and Candida spp.
The leading pathogens among cases of HA BSIs in the ICU are Gram-positive
microorganisms, representing nearly half of the organisms isolated [1719, 21, 27]
(Table 15.2). CNS, S. aureus, and enterococci are the most frequent Gram-positive
bacteria in all studies, and CNS is isolated in 2030% of all episodes of BSI.
15 Bloodstream Infection in the ICU Patient 237
Type of infection Rello [17] Pittet [19] Valls [18] Edgewort [26] Garrouste-Orgas
(%) (%) (%) (%) [21] (%)
Intravenous catheter 35 18 37.1 62 20.2
Respiratory tract 10 28 17.5 3 16.3
Intra-abdominal 9 NA 6.1 6.9 NA
Genitourinary tract 3.6 5.4 5.9 2.4 2.5
Surgical wound 8 8 2.4 3 9.9
Other 7 14.5 2.9 12.9
Unknown origin 27 20 28.1 22.4 32.7
Gram-negative bacilli are responsible for 3040% of BSI episodes, and the
remaining cases are mostly due to Candida spp. Polymicrobial episodes are
relatively common, representing about 10%. Anaerobic bacteria are isolated in
fewer than 5% of cases. Among Gram-positive BSIs, the incidence of pathogens is
similar in the different ICUs, with CNS being the most frequently isolated
organism and S. aureus the second commonest pathogen in all studies. Only the
incidence of strains with antibiotic resistance, such as methicillin-resistant
Staphylococcus aureus (MRSA) or vancomycin-resistant enterococci (VRE), dif-
fers substantially according to the characteristics of individual institutions and
depending on whether they become established as endemic nosocomial pathogens
in the ICU. On the other hand, the Gram-negative species isolated from HA BSIs
in ICUs of different institutions show marked variability. The relative contribution
of each Gram-negative species to the total number of isolates from blood varies
from hospital to hospital and over time. The antibiotic policy of the institution may
induce the appearance of highly drug-resistant microorganisms and the emergence
of endemic nosocomial pathogens, in particular, Pseudomonas spp, Acinetobacter
spp., and Enterobacteriaceae, with extended-spectrum beta-lactamase (ESBL).
15.2.4 Sources
The vast majority (70%) of nosocomial BSIs in the ICU are secondary bactere-
mias, including the BSIs related to intravascular catheter infection, and the
remaining 30% are bacteremias of unknown origin. Table 15.3 summarizes the
sources of nosocomial bacteremias in the ICU reported in several series [1719,
21, 26]. As shown, intravascular catheter-related infections and respiratory tract
infections are the leading sources of secondary episodes. The source of nosocomial
BSIs varies according to microorganism. Coagulase-negative staphylococci and S.
aureus commonly complicate intravenous-related infections, whereas Gram-
negative bacilli are the main etiology for secondary BSIs following respiratory
tract, intra-abdominal, and urinary tract infections. Among bacteremias of
unknown origin, most are caused by Gram-positive microorganisms, mainly CNS,
238 J. Valles and R. Ferrer
and may originate also in device-related infections not diagnosed at the time of
BSI development.
15.2.6 Prognosis
15.2.7 Prevention
by the CDC: hand washing, using full-barrier precautions during CVC insertion,
cleaning the skin with chlorhexidine, avoiding the femoral site if possible, and
removing unnecessary catheters. A checklist was used to ensure adherence to
infection-control practices. Three months after implementing the intervention, their
median rate of CR-BSI had decreased from 2.7/1,000 catheter days at baseline to 0/
1,000 catheter days (p \ 0.002), and their mean rate had decreased from 7.7/1,000
catheter days at baseline to 1.4/1,000 catheter days (p \ 0.002). This improvement
was maintained throughout the 18-month study period.
In 2007 in our ICU, we implemented a similar multiple-system intervention
applying evidence-based measures and reduced the incidence of catheter-relate
BSI from 6.7/1,000 catheter days to 2.4/1,000 catheter days (RR 0.36; 95% CI
0.160.80; p = 0.015), with a 20% reduction in the incidence of HA BSIs in the
ICU [38] (Fig. 15.1).
15.3.1 Epidemiology
15.3.2 Microbiology
15.3.3 Sources
The incidence of severe sepsis and septic shock in patients with CA BSIs is higher
than in HA episodes, in part because the severity of the systemic response is the
reason for ICU admission. In a multicenter French study, 74% of CA BSI episodes
presented severe sepsis or septic shock at admission [20]. In a multicenter Spanish
study carried out in 30 ICUs, the incidence of severe sepsis and septic shock was
75%. In that study, Gram-negative microorganisms and urinary tract and intra-
abdominal infections were associated more frequently with septic shock [39].
15.3.5 Prognosis
Patients admitted to the ICU with CA BSI present a crude mortality rate close to
40%, compared with 18% in patients admitted to general wards [12, 39, 40]. This
is due in part to the severity of systemic response (severe sepsis and septic shock)
and associated complications, which is the reason for admission [12, 39]. The
appropriateness of empiric antimicrobial treatment is the most important variable
influencing the outcome of these patients [12, 39]. The incidence of inappropriate
antibiotic treatment in CA BSIs ICU patients ranges between 15% and 20% in two
studies, and the mortality rate among patients with inappropriate empiric antibiotic
treatment is [70% [12, 39, 41].
242 J. Valles and R. Ferrer
15.4.1 Epidemiology
Patients residing in the community and who are receiving care at home, living in
personal-care and rehabilitation centers, receiving chronic dialysis, and receiving
chemotherapy in physicians offices may present BSIs, which have traditionally
been categorized as CA infections. However, the difference between community-
or HA BSIs has become less clear. Therefore, some investigators propose a new
classificationhealthcare-associated infectionsthat are distinct from CAI and
HAI [42, 43]. Health-care-associated BSI has been defined when a positive blood
culture is obtained from a patient at the time of or within 48 h of hospital
admission if the patient fulfilled any of the following criteria:
1. received intravenous therapy at home, received wound care or specialized
nursing care, or had self-administered intravenously administered medical
therapy;
2. attended a hospital hemodialysis clinic or received chemotherapy intravenously;
3. was hospitalized in an acute-care hospital for C2 days in the 90 days before the
BSI;
4. resided in a personal- or long-term-care facility [43].
According to this new classification, approximately 4050% of patients
admitted with BSI traditionally defined as CA should be classified as having
healthcare-associated BSI [42, 43]. In a large US database, healthcare-associated
BSI accounted for more than half of all BSIs. If patients with healthcare-associated
BSI had been included in the CA BSI category, according to the traditional
classification scheme, they would have accounted for approximately 60% of CA
BSI patients who needed hospitalization [44].
There are no studies available regarding the importance of healthcare-
associated BSI in ICU patients using this new classification. However, recent
studies suggest that healthcare-associated BSIs are less frequent than HA- and CA
BSI in critically ill patients. In a multicenter study carried out in three hospitals in
Spain, 1,157 BSI episodes were studied: 50% were CA, 26% HA, and 24%
healthcare-associated BSI [45]. In patients admitted to the ICU, 60% of BSIs were
HA, 30% CA, and 10% healthcare associated.
We conducted a multicenter study in 28 ICUs in Spain (unpublished data)
analyzing 1,590 BSI episodes and confirmed the low incidence of healthcare-
associated BSI in ICU patients compared with patients admitted to conventional
wards. The most frequent BSIs were HA (77%), CA (21%), and healthcare
associated (8%). Compared with patients with CA episodes, patients with
healthcare-associated BSI were older and more likely to have severe comorbidi-
ties, such as congestive heart failure, peripheral vascular disease, chronic renal
disease, and cancer. This high number of comorbidities and the patients basal
condition may be the reason for a lower ICU admission rate of these patients due
15 Bloodstream Infection in the ICU Patient 243
Table 15.5 Pathogens most frequently found in bloodstream infections through epidemiologic
type of infection in a Spanish multicenter study [45]
15.4.2 Microbiology
15.4.3 Sources
15.4.5 Prognosis
Friedman et al. [43] found a mortality rate at follow-up greater in patients with
healthcare-associated BSI (29% vs. 16%; p = 0.019) or HA infection (37% vs.
16%; p \ 0.001) than in patients with CA infection. Similar results were found by
Shorr et al. [44], where the mortality risk was significantly higher if BSI was
acquired in the hospital or associated with previous healthcare exposure. Consis-
tent with these reports, we found a significantly higher mortality rate at follow up
in groups with HA BSI (27.3%) and healthcare-associated BSI (27.5%) than in
those with CA BSI (10.4%) (p \ 0.001). Among patients with CA and healthcare-
associated BSIs, a multivariate analysis, adjusted for age and comorbidities,
showed healthcare-associated BSI (OR 2.4; 95% CI 1.53.7; p \ 0.001) as an
independent factor associated with mortality [45].
15.5 Treatment
BSIs are among the most serious infections causing severe sepsis or septic shock
acquired by hospitalized patients requiring intensive care. The mainstay of therapy
for patients with bacteremia remains antimicrobial therapy associated with a
optimal management of consequences of bacteremia, such as shock or metastatic
suppurative complications and surgical treatment, such as debridement or drainage
15 Bloodstream Infection in the ICU Patient 245
Patients with BSI who develop severe sepsis or septic shock require additional
treatment: early goal-directed therapy (EGDT) [48], corticosteroids for refractory
septic shock [49], recombinant human activated protein C or drotrecogin alfa
(activated) for multiorgan failure [50], and lung-protective ventilation strategies
[51] have all been associated with survival benefits. These and other therapeutic
advances led to the development of the surviving sepsis campaign (SSC) guide-
lines [52]. To improve care for patients with sepsis, the SSC and the Institute for
Healthcare Improvement recommend implementing two sepsis bundles: the
resuscitation bundle and the management bundle (Table 15.6):
1. The resuscitation bundle includes lactate determination, early cultures and
antibiotics, and EGDT. This bundle describes seven tasks that should begin
immediately and must be accomplished within the first 6 h of presentation of
severe sepsis or septic shock. Some items may not be completed if the clinical
conditions described in the bundle do not prevail in a particular case, but
clinicians must assess for these elements.
2. The management bundle includes optimization of glycemic control and respi-
ratory inspiratory plateau pressure and determination of the need for cortico-
steroids or drotrecogin alfa (activated). Efforts to accomplish these goals should
begin immediately, and these items must be completed within 24 h of pre-
sentation of severe sepsis or septic shock.
246 J. Valles and R. Ferrer
15.6 Summary
BSIs are among the most serious infections causing severe sepsis or septic shock in
hospitalized patients requiring intensive care. Nosocomial BSIs account for almost
20% of HAI in critically ill patients, [80% of which are associated with a central
line. These infection rates among ICU patients are as much as five to ten times
higher than those recorded for patients admitted to general wards. CA infections
represent an important reason for ICU admission: severe pneumonia and urinary
tract and intra-abdominal infections are the most frequent CA infections requiring
ICU admission, and approximately 20% of these patients will also present
15 Bloodstream Infection in the ICU Patient 247
bacteremia, which associated with a high incidence of severe sepsis and septic
shock. A new classification scheme for BSIshealthcare-acquired BSIshas been
proposed to distinguish between infections occurring among outpatients having
recurrent or recent contact with the healthcare system and patients with true
CA infections. According to this classification, approximately 4050% of patients
admitted to general hospital wards classified as having CA BSIs should be classified
as having healthcare-associated BSI. However, the rate of healthcare-acquired
BSI among critically ill patients in the ICU seems to be lower (\20% of
community-acquired infections) than among patients admitted to conventional
wards. BSIs in critically ill patients are associated with greater hospital mortality
rates. Clinical efforts should therefore be aimed at improving severe sepsis and
septic shock management, reducing the incidence of inadequate antimicrobial
treatment, and preventing BSI episodes associated with intravascular devices.
References
1. Wenzel RP (1990) Organization for infection control. In: Mandell GL, Douglas RG Jr,
Bennett JE (eds) Principles and practice of infectious diseases. Churchill Livingstone, New
York, pp 21762180
2. Weinstein RA (1991) Epidemiology and control of nosocomial infections in adult intensive
care units. Am J Med 91(Suppl 3B):179S184S
3. Wenzel RP, Thompson RL, Landry SM et al (1983) Hospital-acquired infections in intensive
care unit patients: an overview with emphasis on epidemics. Infect Control 4:371375
4. Maki DG (1989) Risk factors for nosocomial infection in intensive care: devices vs nature
and goals for the next decade. Arch Intern Med 149:3035
5. Donowitz LG, Wenzel RP, Hoyt JW (1982) High risk of hospital-acquired infection in the
ICU patient. Crit Care Med 10:355357
6. Brown RB, Hosmer D, Chen HC et al (1985) A comparison of infections in the different ICUs
within the same hospital. Crit Care Med 13:472476
7. Daschner F (1985) Nosocomial infections in intensive care units. Intensive Care Med
11:284287
8. Trilla A, Gatell JM, Mensa J et al (1991) Risk factors for nosocomial bacteremia in a large
Spanish teaching hospital: a case-control study. Infect Control Hosp Epidemiol 12:150156
9. Massanari RM, Hierholzer WJ Jr (1986) The intensive care unit. In: Bennett JV, Brachman
PS (eds) Hospital infections. Little, Brown and Company, Boston, pp 285298
10. Ponce de Len-Rosales S, Molinar-Ramos F, Domnguez-Cherit G et al (2000) Prevalence of
infections in intensive care units in Mexico: a multicenter study. Crit Care Med 28:13161321
11. Forgacs IC, Eykyn SJ, Bradley RD (1986) Serious infection in the intensive therapy unit: a
15-year study of bacteraemia. Q J Med 60:773779
12. Valls J, Ochagava A, Ru M et al (2000) Critically ill patients with community-acquired
bacteremia: characteristics and prognosis. Intensive Care Med 26(Suppl. 3):S222
13. Garner JS, Jarvis WR, Emori TG et al (1988) CDC definitions for nosocomial infections. Am
J Infect Control 16:128140
14. Trilla A (1994) Epidemiology of nosocomial infections in adult intensive care units. Intensive
Care Med 20:S1S4
15. Richards MJ, Edwards JR, Culver DH et al (1999) Nosocomial infections in medical
intensive care units in the United States. Crit Care Med 27:887892
16. Wisplinghoff H, Bischoff T, Tallent SM et al (2004) Nosocomial bloodstream infections in
US Hospitals: analysis of 24, 179 cases from a prospective nationwide surveillance study.
Clin Infect Dis 39:309317
248 J. Valles and R. Ferrer
16.1 Introduction
Many attempts have been made to classify peritonitis in general and secondary
peritonitis in particular, which include a large variety of different pathological
conditions ranging in severity from a local problem to a devastating disease.
A simplified version is reported in Table 16.1. This differentiates the relatively
G. Sganga (&)
Istituto di Clinica Chirurgica, Universit Cattolica del Sacro Cuore,
Rome, Italy
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 251
DOI: 10.1007/978-88-470-1601-9_16, Springer-Verlag Italia 2012
252 G. Sganga et al.
Primary peritonitis
Diffuse bacterial peritonitis in absence of GIT disruption
Spontaneous peritonitis in children
Spontaneous peritonitis in adults
Peritonitis in patients receiving continuous peritoneal dialysis
Tuberculous and other granulomatous peritonitis
Secondary peritonitis
Localized or diffuse peritonitis originating from a defect in GIT
Acute perforation peritonitis (GIT perforation, intestinal ischemia, pelviperitonitis, and other
forms)
Postoperative peritonitis (anastomotic leak, accidental perforation, and devascularization)
Posttraumatic peritonitis (after either blunt or penetrating abdominal trauma)
Tertiary peritonitis
Late peritonitis-like syndrome due to disturbance in the immune response
Peritonitis without evidence of pathogens
Peritonitis with fungi
Peritonitis with low-grade pathogenetic bacteria
GIT gastrointestinal tract
rare forms of primary peritonitis, which usually respond to medical treatment, and
tertiary peritonitis, which does not respond to any treatment, from the commonly
occurring secondary peritonitis that mandates surgical intervention and antibiotic
therapy [2]. IAIs include the following pathological conditions:
1. Infections of single organs (cholecystitis, appendicitis, diverticulitis, cholan-
gitis, pancreatitis, salpingitis, etc.), which may or may not be complicated by
peritonitis, even in the absence of perforation.
2. Peritonitis, classified as primary, secondary or tertiary.
3. Intra-abdominal abscesses, classified on the basis of their location and anatomic
configuration.
The term complicated IAI (C-IAI) is used to indicate infections that, originating
in an organ cavity, extend into the peritoneal space and form an abscess or peri-
tonitis. Resolution of this type of infection requires surgical treatment and per-
cutaneous drainage, as well as systemic antibiotic therapy [6]. They are divided
into two types: community c-IAIs, which can be mild or serious; and hospital c-
IAIs, which usually occur as postoperative infections. The nature of severe IAIs
makes it difficult to precisely define the disease, to assess its severity, and to
evaluate and compare therapeutic progress. Both the anatomical source of infec-
tion and, to a greater degree, the physiological compromise it inflicts, affects the
outcome of IAI.
16 Infections of Peritoneum, Mediastinum, Pleura, Wounds, and Urinary Tract 253
16.3 Microbiology
function and survival. IL-8 may have an especially important role in perpetuating
tissue inflammation. IL-6 and -10, which are perhaps counterregulatory, inhibit
the generation of TNF, augment the action of acute phase reactants and
immunoglobulins, and inhibit T-lymphocyte and macrophage function [11].
Cytokines are measurable in the systemic circulation and in peritoneal exudates,
and the magnitude of the phenomena is negatively correlated with outcome [2,
3]. Most peritoneal cytokines probably derive from macrophages exposed to
bacterial endotoxin [12]. Recent findings suggest that female sex hormones play
a critical role in maintaining the immune response after trauma hemorrhage by
suppressing the production of TNF-a and preventing increased mortality rates
from subsequent sepsis [12]. Other potential sources are direct translocation of
cytokines through the GIT barrier or production by traumatized tissues [13].
Timely therapeutic intervention is crucial to abort the ensuing, self-perpetuating,
SIRS, sequential MOF, and death [2, 1012].
16.5 Diagnosis
Radiographic imaging is the definitive diagnostic tool for patients with suspected
IAI and can usually identify the problem prior to any planned intervention. White
abdomen usually indicates the presence of gas in the peritoneum, an intestinal
obstruction, or signs of intestinal ischemia. Studies with contrast medium using
hydrosoluble agents can reveal a fold break. Contrast fluid in a drain or fistula can
help delineate the anatomy of a complex infection and help verify the adequacy of
abscess drainage. US, and especially CT, scans play a principal role in the diagnosis
and therapeutic strategy. US has the advantage of being easily available, can be
carried out on the patient in bed, and repeated as often as necessary because of its
innocuous effect on the patient. It permits investigation of the entire abdominal
pelvic cavity, revealing even mild intraperitoneal effusion (\500 ml), detection of
the distribution of the peritoneal cavity or part of it (localized effusion), thus
assisting in hypothesizing about the nature of the effusion based on its possible
homogeneous aspects, and finally help guide fine-needle percutaneous aspiration
[20]. Abdominal CT is the reference standard for evaluating the abdomen in criti-
cally ill patients [21]. Most causes of secondary peritonitis can be diagnosed
promptly with CT using intravenously administered iodinated contrast fluid and
opacification of the digestive tube (using hydrosoluble contrast medium injected
orally or rectally) Although CT is an invaluable diagnostic instrument, it usually
involves moving a potentially unstable patient from one ward to another. Further-
more, iodinated contrast medium can worsen renal function. Renal insufficiency and
paralytic ileum are both contraindications for CT, so the risks must be taken into
account when deciding whether to carry out this diagnostic technique. One alter-
native in a very unstable patient is diagnostic peritoneal lavage, which consists of
injecting 1 l of normal saline solution or lactated ringers through a catheter into the
peritoneum and subsequently observe the characteristics of the fluid returning from
the lavage. This can evidence the presence of bacteria, leukocytes, bile, enteral
content, or blood in cases of acute intestinal ischemia [22]. Scintigraphy has a very
limited role in IAI diagnosis in critically ill patients because it is not specific enough
and cannot provide an accurate enough image to guide drainage [23, 24]. MRI has a
very high diagnostic accuracy in evaluating acute intra-abdominal abscess [22].
256 G. Sganga et al.
16.6 Management
Source control is defined as any and all physical measures necessary to eradicate
a focus of infection as well as influence factors that maintain infection and thus
promote microbial growth or impair host antimicrobial defenses [7, 25]. Primary
peritonitis is essentially a disease that is managed with antibiotics and not sur-
gery [26]. There is no reliable evidence that cefotaxime is the treatment of
choice for spontaneous bacterial peritonitis, although many authors have sug-
gested this. Furthermore, results indicate that 4 g/day cefotaxime may be as
effective as 8 g/day in terms of reducing mortality rates and symptoms resolu-
tion, and that treatment for 10 days is no more effective than treatment for
5 days. Goals for managing secondary peritonitis are summarized in Table 16.2.
Several studies have identified E. coli and B. fragilis as the main target
organisms for antibiotic therapy [2, 27]. The current practice of early empirical
antibiotic administration, targeted to these bacteria, is well established. However,
issues concerning drug choice and timing, the need for surgical cultures, and the
duration of postoperative administration are controversial. Despite several pub-
lished options, antibiotic therapy for secondary peritonitis is simple. The
emerging concepts suggest that less in terms of number of drugs and treatment
duration is better [2]. Furthermore, recent studies suggest that monotherapy with
a single broad-spectrum antibiotic that includes full activity against E. coli may
be equal or superior to polytherapy with multiple drug combinations [2730].
The surgical strategy depends on the source of the infection [31, 32], the degree
of peritoneal contamination, the patients clinical condition, and concomitant
disease. Moreover, early goal-directed therapy provides significant benefits with
respect to outcome in patients with severe sepsis and MOF [33]. Ideally, a severe
IAI should be cured with a single surgical procedure; unfortunately, infection
often persists or recurs. Traditionally, severe peritonitis has been treated by
performing a midline laparotomy to identify and eliminate the source of infec-
tion. In certain instances, complete control of the infective focus is not feasible
during the first operation [2]. Whereas eliminating the focus and reducing con-
tamination are accepted as conditions of successful treatment, surgical proce-
dures differ for treating residual infection. The following major approaches have
been developed: (1) continuous peritoneal lavage; (2) planned relaparotomy; and
(3) open treatment by laparostomy. Continuous peritoneal lavage takes the whole
concept of lavage to an extreme, with the hypothesis being that continual IA
irrigation will enhance removal of bacteria and their products and improve the
time to resolution [34]. Various forms of peritoneal lavage are routinely used to
manage patients with peritonitis. There is little evidence that supports this
approach in either the clinical and scientific literature; moreover, it has been
documented that lavage damages mesothelial cells, dilutes agents that are
involved in peritoneal defense, and may spread previously contained infection.
16 Infections of Peritoneum, Mediastinum, Pleura, Wounds, and Urinary Tract 257
Table 16.2 Principles for managing peritonitis and indications for staged abdominal repair
1. Supportive measures
Combat hypovolemia and shock and maintain adequate tissue oxygenation
Treat with antibiotics bacteria not eliminated by surgery
Support failing organ systems
Provide adequate nutrition
2. Operative treatment
Repair and/or control infection source
Evacuate bacterial inoculum, pus, and adjuvants
Treat abdominal compartment syndrome
Prevent or treat persistent and recurrent infection or verify both repair and purge
3. Staged abdominal repair
Critical patient condition due to hemodynamic instability, precluding definitive repair
Excessive peritoneal edema (abdominal compartment syndrome, pulmonary, cardiac, renal, or
hepatic dysfunction, decreased visceral perfusion) preventing abdominal closure without
tension
Intra-abdominal pressure [15 mmHg
Massive abdominal wall loss
Impossible to eliminate or to control the source of infection
Incomplete debridement of necrotic tissue
Uncertainty of viability of remaining bowel
Uncontrolled bleeding (the need for packing)
16.7.1 Pathophysiology
SAP is diagnosed if three or more of Ransons criteria are present, if the acute
physiology and chronic health evaluation (APACHE) II score is C8, or if one or
more of the following are present: shock, renal insufficiency, pulmonary insuffi-
ciency. Pancreatic glandular necrosis is usually associated with necrosis of
260 G. Sganga et al.
Fig. 16.2 Mechanisms of acute pancreatitis and resulting complications. Acute pancreatitis can
be triggered by several events, resulting in inflammation of the parenchyma. Ischemia of the
organ appears to transform mild edematous pancreatitis into severe necrotizing forms of the
disease. Pseudocysts can form if pancreatic juices and debris leak into the peripancreatic spaces.
Necrotic parenchyma becomes secondarily infected 4060% of the time, usually with Gram-
negative bacteria translocated from the GIT. Alternatively, the necrotic pancreas may release
toxic factors into the peripancreatic spaces, peritoneal cavity, or systemic circulation, leading to
local or systemic complications. Computed tomography scans shown a pancreatic pseudocyst,
and lack of enhancement representing necrosis of the head, body, and tail of the pancreas
(arrows)
peripancreatic fat and, by definition, represents SAP. The risk of infected necrosis
increases with the amount of glandular necrosis and the time from AP onset,
peaking at 3 weeks. Overall mortality rate in SAP is approximately 30%; as long
as necrotizing AP remains sterile, the mortality rate is approximately 10%, and it
at least triples if there is infected necrosis. In addition, patients with sterile necrosis
and high severity-of-illness scores accompanied by MOF, shock, or renal insuf-
ficiency have significantly higher mortality rates. Deaths occur in either of two
phases: early deaths (12 weeks after AP onset) are due to MOF caused by the
release of inflammatory mediators and cytokines; late deaths result from systemic
or local infection. Local infections, so-called secondary pancreatic infections, are
16 Infections of Peritoneum, Mediastinum, Pleura, Wounds, and Urinary Tract 261
16.7.3 Recognition
Acute phase ERC: diagnosis of biliary pancreatitis in severe cases with an uncertain
etiology
ES: treatment of severe cases of gallstones, biliary sludge, and other related
pathologies, such as ampullary carcinoma
Persisting ES: for bile duct stones in unresolving mild pancreatitis
pancreatitis ERP: disruption of main pancreatic duct or planning surgical intervention
Complications ERP: assessment of pseudocysts, ascites, and fistulas prior to surgery
Stent or NBC: pseudocysts, fistulas, and liquid necrosis in selected cases
ERC: endoscopic pseudocystogastrostomy, or endoscopic
pseudocystoduodenostomy in selected cases
Convalescent ERCP: establishment of diagnosis in idiopathic cases
phase ERC with bile duct cannulation: CCK-stimulated bile collection for crystal
analysis
ES: alternative to cholecystectomy in high risk cases
Recurrent ES: sphincter of Oddi dysfunction or stenosis
pancreatitis Stent: trial therapy for pancreas divisum
ERC endoscopic cholangiography; ERP endoscopic retrograde pancreatography; ES endoscopic
sphincterotomy; NBC nasobiliary catheter
well tolerated, had no adverse clinical effects, and resulted in significantly fewer
total and infectious complications [57, 58]. Acute phase-response scores and
disease-severity scores were significantly improved after EN. Studies of endo-
scopic retrograde cholangiopancreatography (ERCP) and biliary sphincterotomy
performed within 72 h of admission in patients with gallstone AP and choledo-
cholithiasis showed an improved outcome in the group of patients who presented
with clinically SAP. Improvement was attributed to the relief of pancreatic ductal
obstruction and from reduced biliary sepsis. In the presence of pancreatic ductal
disruption, a frequent occurrence in pancreatic necrosis, the introduction of
infectiontransforming sterile to infected pancreatic necrosisby incidental
pancreatography during ERCP may theoretically occur. Therefore, ERCP should
be reserved for patients in whom biliary obstruction is suspected (Table 16.4).
Timing and type of intervention for patients with pancreatic necrosis are contro-
versial. As the mortality rate from sterile necrosis is approximately 10% and
surgical intervention has not been shown to lower this figure, most investigators
recommend supportive medical therapy [45]. Conversely, infected necrosis is
considered uniformly fatal without intervention. Aggressive surgical pancreatic
debridement, so-called necrosectomy, remains the standard of care if drainage is
16 Infections of Peritoneum, Mediastinum, Pleura, Wounds, and Urinary Tract 263
There are three main types of surgical drainage after pancreatic debridement: con-
ventional drainage, open or semiopen procedures, and closed procedures with lesser
sac lavage [5971]. Conventional drainage involves necrosectomy with placement
of Penrose/sump drainage. Open or semiopen management involves necrosectomy
and either scheduled repeated laparotomies or open packing, which leaves the
abdominal wound exposed for frequent dressing changes. Closed management
involves necrosectomy with extensive intraoperative lavage of the pancreatic bed;
furthermore, the abdomen is closed over large-bore drains for continuous high-
volume postoperative lavage of the lesser sac. Results of these procedures are
reported in Table 16.5. The principal reason for failure of the conventional surgical
approach is the development of recurrent postoperative infection and sepsis. In an
extensive review [72] of [1,100 cases of severe pancreatic infections, 76% of
postoperative deaths were found to be attributable to persistent or recurrent infection
after surgical drainage. A closer examination of the reasons behind the demonstrated
failure of Penrose/sump drainage to provide adequate control of postoperative
infection unearths the fact that unscheduled re-explorations for recurrent sepsis are
264 G. Sganga et al.
necessary in one-third to one half of cases drained in this fashion [73, 74]. Leaving
the abdomen open eliminates the need for repeated laparotomy; packing may be
changed in ICU. Repeated debridement and manipulation of the abdominal viscera
with the open and semiopen techniques results in a high rate of postoperative local
complications (pancreatic fistulas, small- and large-bowel complications, bleeding
from pancreatic bed). Pancreatic or GIT fistulas occur in up to 40% of patients after
surgical necrosectomy and often require additional surgery for closure. The mor-
tality rate from debridement with the open or semiopen technique is approximately
15%. In patients who underwent lesser sac lavage, 1640% required reoperation for
persistent peripancreatic sepsis, and mortality rates varied from 5 to 50%, averaging
30% [72, 75].
Alternative methods for debriding pancreatic necrotic material have recently been
described [7678] but require considerable technical expertise, and the precise role
of these techniques in managing pancreatic necrosis will be better defined. One
study described the successful treatment of infected necrosis by aggressive irri-
gation and drainage through large-bore percutaneous catheters in 34 patients.
Pancreatic surgery was completely avoided in 47% of patients; in nine patients,
sepsis was controlled, and elective surgery was later performed to repair external
pancreatic fistulas related to catheter placement. Nine patients required immediate
surgery when percutaneous therapy failed, and four (12%) died [78]. Recently,
endoscopic drainage of sterile or infected pancreatic necrotic material has been
reported [79]. Complete necrosis resolution without the need for surgery was
achieved in 25 of 31 patients (81%). Surgical intervention was required more
commonly for acute complications of endoscopy than for drainage failure. Lap-
aroscopic approach to SAP treatment has been recently reported [77]. Advantages
are the possibility of exploring, irrigating, decompressing, and draining the pan-
creas, and performing postoperative lavage via the drainage tubes. Pathological
disease extent can be determined and appropriate treatment approaches thus
planned by laparoscopic exploration. The laparoscopic technique creates less
trauma in the early treatment of SAP but, at least theoretically, patients with early
severe multiorgan disease without retroperitoneal infection may become victims of
a later infection introduced surgically [80, 81].
This is often a mixed infection, with facultative and strict anaerobes acting
together. Streptococcus spp. are the most common facultative organisms (70% of
cases), whereas Bacteroides spp. are the most common strict anaerobes [91]. Other
organisms implicated include Pseudomonas aeruginosa and Fusobacterium,
Peptostreptococcus, and Staphylococcus spp. Mixed Gram-positive and Gram-
negative infections account for about 40% of cases. Isolated Gram-negative
infections rarely occur. Mediastinitis not associated with cardiac surgery is usually
due to anaerobic organisms. Mixed Gram-negative and Gram-positive infections
as well as Candida spp. are more common after esophageal perforation or head
and neck surgery [82, 92].
16.8.2 Diagnosis
16.8.3 Treatment
Treating mediastinitis requires correcting the inciting cause and aggressive sup-
portive therapies. Mediastinitis may result in airway compromise, and protecting
the airway is often vital. Appropriate, well-directed antibiotic therapy is crucial to
successful treatment. In postoperative mediastinitis, because up to 20% of
organisms cultured from infected sternotomy sites will be methicillin-resistant S.
aureus (MRSA), and because another 20% will be Gram-negative organisms,
antibiotic coverage must be very broad and deep to include Pseudomonas spp.
While awaiting results of stains and cultures, a regime of vancomycin, ceftazi-
dime, and a quinolone should provide adequate coverage. If septic shock is
present, some would substitute an aminoglycoside for the quinolone, although
close watch of drug levels is required to prevent renal damage. Therapy duration is
usually prolonged, ranging from weeks to months. Recently, it has suggested that
16 Infections of Peritoneum, Mediastinum, Pleura, Wounds, and Urinary Tract 267
PE characteristic Diseases
Transudative Congestive heart failure (most common transudative PE); hepatic cirrhosis
with and without ascites; nephrotic syndrome; peritoneal dialysis/continuous
ambulatory peritoneal dialysis; hypoalbuminemia (e.g., severe starvation);
glomerulonephritis; superior vena cava obstruction; urinothorax
Exudative Malignant disorders (metastatic disease to the pleura or lungs, primary lung
cancer, mesothelioma, Kaposi sarcoma, lymphoma, leukemia); infectious
diseases (bacterial, fungal, parasitic, and viral infections; infection with
atypical organisms such as Mycoplasma, Rickettsiae, Chlamydia,
Legionella); GIT diseases (pancreatic disease, Whipple disease, IA abscess,
esophageal perforation, abdominal surgery, diaphragmatic hernia, endoscopic
variceal sclerotherapy); collagen vascular diseases (rheumatoid arthritis,
systemic lupus erythematosus, drug-induced lupus syndrome, immunoblastic
lymphadenopathy, Sjgren syndrome, familial Mediterranean fever, Churg
Strauss syndrome, Wegener granulomatosis); benign asbestos effusion;
Meigs syndrome (benign solid ovarian neoplasm associated with ascites and
pleural effusion); drug-induced primary pleural disease (nitrofurantoin,
dantrolene, methysergide, bromocriptine, amiodarone, procarbazine,
methotrexate, ergonovine, ergotamine, oxprenolol, maleate, practolol,
minoxidil, bleomycin, interleukin-2, propylthiouracil, isotretinoin,
metronidazole, mitomycin); injury after cardiac surgery (Dressler syndrome);
uremic pleuritis; yellow nail syndrome; ruptured ectopic pregnancy;
electrical burns
Exudative/ Pulmonary embolism; hypothyroidism; pericardial disease (inflammatory or
transudative constrictive); atelectasis; trapped lung (usually a borderline exudate);
sarcoidosis (usually an exudate); amyloidosis
Miscellaneous Hemothorax; following coronary artery bypass graft surgery; after lung or
liver transplant; Milk of calcium pleural effusion; ARDS; systemic
cholesterol emboli; iatrogenic misplacement of lines or tubes into the
mediastinum or the pleural space; radiation pleuritis; necrotizing sarcoid
granulomatosis; ovarian hyperstimulation syndrome; postpartum pleural
effusion (immediate or delayed); rupture of a silicone bag mammary
prosthesis; rupture of a benign germ cell tumor into the pleural space (e.g.,
benign mediastinal teratoma; syphilis; echinococcosis
GIT gastrointestinal tract; IA intra-abdominal; ARDS acute respiratory distress syndrome
16.9.3 Diagnosis
This step is the most important in the PE evaluation. Common imaging studies
used to confirm PE are chest radiography, US, and CT scan [96101]. Chest
radiography is the primary diagnostic tool because of its availability, accuracy,
and low cost. It can be used to determine the cause of PE (enlarged cardiac
silhouette, underlying lung, parenchymal disease). The most common radiologic
appearance is blunting of the costophrenic angle and/or sulci. Upright postero-
anterior or anteroposterior radiographs may not show lateral costophrenic angle
16 Infections of Peritoneum, Mediastinum, Pleura, Wounds, and Urinary Tract 271
Characteristic Significance
Bloody Most likely an indication of malignancy in the absence of trauma;
can also indicate pulmonary embolism, infection, pancreatitis,
tuberculosis, mesothelioma, or spontaneous pneumothorax
Turbid Possible increased cellular or lipid content
Yellow or whitish, turbid Presence of chyle, cholesterol, or empyema
Brown Rupture of amoebic liver abscess into the pleural space (amebiasis
with a hepatopleural fistula)
Black Aspergillus involvement of pleura
Yellow green with debris Rheumatoid pleurisy
Highly viscous Malignant mesothelioma (due to increased levels of hyaluronic
acid), long-standing pyothorax
Putrid odor Anaerobic infection of pleural space
Ammonia odor Urinothorax
Purulent Empyema
Yellow and thick, with Effusions rich in cholesterol (longstanding chyliform effusion, e.g.,
metallic sheen tuberculous or rheumatoid pleuritis)
Table 16.8 Clinical significance of biochemical and cytological pleural fluid characteristics
Characteristic Significance
Amylase Can be elevated in acute pancreatitis, pancreatic pseudocyst,
esophageal rupture, malignancy, and ruptured ectopic pregnancy.
Pancreatic pseudocyst has the highest amylase levels
(frequently [100,000 UI). Determination of the amylase isoenzyme
level is useful in distinguishing PE caused by pancreatic disease from
effusions caused by esophageal rupture or nonpancreatic disease
Glucose A low PE glucose level is \60 mg/dl. Differential diagnosis includes
malignancy, rheumatoid pleurisy, complicated parapneumonic PE,
empyema, hemothorax, ChurgStrauss syndrome, and occasionally
lupus pleuritis
pH If \7.20, suggests empyema, complicated parapneumonic PE,
esophageal rupture, rheumatoid pleuritis, malignancy, hemothorax,
tuberculous pleuritis, lupus pleuritis, or urinothorax. Arterial pH
influences pleural fluid pH; therefore, acidemia must be ruled out
before any of the above causes are considered. With parapneumonic
PE, indications for tube thoracostomy include a pH \7.0, glucose
level \40 mg/dl, and positive finding with Gram stains or cultures.
To use the pH criteria for chest tube placement in systemic acidosis,
the PE pH should be at least 0.3 U less than the arterial pH. A low
pleural fluid pH almost always is associated with a high pCO2
LDH Indicator of the degree of pleural inflammation. The higher the value,
the more inflamed the pleural surface. High concentrations
([1,000 IU/l) occur with complicated parapneumonic PE
RBC and total WBC RBC counts [100,000 mm3 suggest trauma, malignancy, pulmonary
count embolism, injury after cardiac surgery, asbestos pleurisy, esophageal
rupture, pancreatitis, tuberculous pleurisy, and thoracic
endometriosis. The total PE leukocyte count is virtually never
diagnostic. Neutrophilic predominance indicates an acute
inflammatory process near the time of thoracentesis. Significant
eosinophilia occurs when the ratio of pleural fluid and total pleural
fluid counts is [10%; the most common cause is air or blood in the
pleural space. The differential diagnosis of pleural fluid eosinophilia
includes pneumothorax, hemothorax, pulmonary infarction, prior
thoracentesis, benign asbestos effusion, drug use, parasitic diseases,
fungal infections, and ChurgStrauss syndrome; in the absence of
these, eosinophilia with pneumonia and pleural effusion is a good
prognostic sign, because such effusions rarely become infected.
Significant basophilia (counts [10%) is distinctly uncommon;
however, if present, it suggests leukemic pleural infiltration
Lymphocyte count Lymphocytes indicate a long-standing chronic effusion. Pleural fluid
lymphocytosis ([50%) suggests malignant disease, particularly
lymphoma; however, other conditions (e.g., chronic rheumatoid
pleurisy, chronic fungal infection, yellow nail syndrome, chylothorax,
trapped lung, benign asbestos PE, sarcoidosis) must be considered.
The presence of an undiagnosed exudative effusion with
lymphocytosis is an indication for closed pleural biopsy
(continued)
16 Infections of Peritoneum, Mediastinum, Pleura, Wounds, and Urinary Tract 273
After the presence of PE is established, the cause should be identified. This step
can be critical because unnecessary invasive procedures cause morbidity and
mortality. When a decision is made to investigate the cause, thoracentesis is the
first-line invasive diagnostic procedure [100]. Thoracentesis also can be used as a
therapeutic modality. It is the least invasive procedure and it is relatively safe. For
stable and asymptomatic patients in whom PE most likely is caused by viral
pleurisy, manifestation of a systemic disease, thoracic or abdominal surgery, or
childbearing, thoracentesis may not be indicated, or it can be deferred. In this
situation, therapy for the specific cause should be initiated, and if no improvement
occurs after a few days, diagnostic thoracentesis should be performed. Thora-
centesis is also indicated in cases in which the specific cause of PE is unknown or
has never been investigated or when the thickness of the free pleural fluid level
is [10 mm on the lateral decubitus radiograph. In addition, thoracentesis is
indicated if the patient has respiratory compromise, hemodynamic instability, or
massive effusion with contralateral mediastinal shift. After thoracentesis, and
regardless of its success, chest radiography is recommended to rule out a sub-
sequent pneumothorax. Pneumothorax is the most common complication (inci-
dence 320% with unguided thoracentesis, 27% with US guidance) and is
operator dependent. Other complications include subcutaneous hematoma, infec-
tion of the pleural space or soft tissue overlying the thoracentesis site, pain at the
site, cough, chest pain, hemothorax, vasovagal reflex, reexpansion pulmonary
edema, hypovolemia, hypoxemia, splenic or hepatic laceration, hemoperitoneum,
and adverse reactions to local anaesthetics. Definite indications to tube thoracos-
tomy include empyema, hemothorax, large pneumothorax, and parapneumonic PE.
16.9.6 Treatment
Wound infection (WI) is the most important complication of surgical procedures and
continues to be a disconcerting source of mortality in surgical patients [10, 102
104]. Postoperative infections, also, prolong hospitalization and are important
causes of postoperative morbidity. Despite antisepsis, surgical site infections (SSI)
are the third most frequently reported nosocomial infection, accounting for 1416%
16 Infections of Peritoneum, Mediastinum, Pleura, Wounds, and Urinary Tract 275
Clean wounds Infection risk of about 15%. Prophylactic antibiotics are not indicated in
a clean operation if the patient has no host-risk factors. Factors suggesting
the need for prophylaxis are remote infection, diabetes, at least three
concomitant medical diagnoses. Additive risk factors are abdominal
operations and operations expected to last [2 h. Prostheses implants are
clean procedures, some of which requires antibiotic prophylaxis. Inguinal
hernia repair with biomaterials does not benefit from antibiotic
prophylaxis
Clean-contaminated B10% risk of infection. Clean-contaminated surgery usually requires
wounds prophylaxis
Contaminated A 1020% risk of infection. This type of surgery needs prophylaxis.
wounds Biliary, hepatobiliary, and pancreatic operations usually meet criteria of
clean-contaminated wounds definitions. In biliary-tract procedures,
prophylaxis is required only for cases at high risk of contamination: bile
obstruction, jaundice, stones in common duct, reoperation, and
cholecystitis. Prophylaxis is always required in hepatobiliary and
pancreatic surgery because these operations are long. In gastroduodenal
operations, the risk is low if gastric acidity is normal and bleeding, cancer,
gastric ulcer and obstruction are absent. Colorectal procedures are usually
contaminated cases. The unique goal of prophylaxis includes preoperative
reduction of bacterial concentration in feces. In association with
mechanical bowel preparation, prophylaxis is used in major elective
abdominal procedure (i.e., vascular graft) to prevent bacterial
translocation from the gut
Infected wounds Risk of infection of about 2050% meeting the following criteria: acute
bacterial inflammation, without pus; transection of clean tissue for the
purpose of surgical access to a collection of pus; traumatic wound with
retained ischemic tissues, foreign bodies, fecal contamination, or delayed
treatment. Appropriate treatment is to administer antibiotic therapy, not as
prophylaxis, because infection is already present
16.10.3 Treatment
The first important point about antibiotic prophylaxis is its timing of administra-
tion [104, 106]. Administration of antibiotics just before, during, and up to 3 h
after surgery effectively prevents WIs. Many studies demonstrate that prophylactic
antibiotics are most useful if given to patients before contamination occurs: the
most relevant protective effect was observed when antibiotic was given so that
good tissue levels were present at the time of the procedure and for the first 34 h
after surgical incision. A practical approach would then be to contemplate
administering a single preoperative dose, followed by an intraoperative dose if the
procedure lasts [3 h or twice the half-life of the antibiotic, and massive hemor-
rhage occurs during surgery [102]. Principles of the proper prophylaxis of WIs
include selecting bactericidal antibiotics effective against likely pathogens. Single-
agent prophylaxis is almost always effective in the majority of clinical situations,
provided that the half-life of the antibiotic is long enough to maintain adequate
tissue levels throughout the operation and that the given dose is equal to a full
therapeutic i.v. dose. Recommended antibiotics for prophylaxis of WI caused by
Gram-positive and Gram-negative aerobic bacteria are cefazolin (1 gm i.v./i.m.) or
vancomycin (1 g i.v.) in patients allergic to cephalosporins. First-generation
cephalosporins, such as cefazolin, are good choices because they are not expen-
sive, incur a low rate of allergic responses, and have a broad-spectrum of activity
against likely aerobic pathogens. Prophylaxis against both Gram-negative aerobes
and anaerobes includes clindamycin or metronidazole plus tobramycin, or a single
broad-spectrum agent such as cefoxitin or cefotetan, or sulbactam/ampicillin.
Gram-negative anaerobes (Bacteroides spp.) are of GIT origin and are synergistic
with Gram-negative aerobes in causing infections after GIT surgical procedures.
Even though the problem is still debated, combination of two antibiotics is in
general more powerful than a single broad-spectrum agent active against both
bacterial components [102, 106]. A second point concerns the cases for which
278 G. Sganga et al.
16.11.1 Pathophysiology
The urinary tract is normally sterile. Uncomplicated UTI involves the urinary
bladder in a host without underlying renal or neurological disease. The clinical
entity is termed cystitis and represents bladder mucosal invasion, most often by
enteric coliform bacteria (E. coli), which inhabit the periurethral vaginal introitus
and ascend into the bladder via the urethra. Sexual intercourse may promote this
migration, and cystitis is common in otherwise healthy young women [114]. Urine
is generally a good culture medium; factors unfavorable to bacterial growth
include a low pH (B5.5), a high concentration of urea, and the presence of organic
acids derived from a diet that includes fruits and protein. Frequent and complete
voiding has been associated with a reduced incidence of UTI [115119]. Nor-
mally, a thin film of urine remains in the bladder after emptying, and any bacteria
present are removed by the mucosal cell production of organic acids. If lower
urinary tract mechanisms fail, upper urinary tract or kidney involvement occurs,
which is termed pyelonephritis. Host defences at this level include local leukocyte
phagocytosis and renal production of antibodies that kill bacteria in the presence of
complement. Complicated UTI occurs in the setting of underlying structural,
medical, or neurological disease [115]. Patients with a neurogenic bladder or
bladder diverticulum, and postmenopausal women with bladder or uterine pro-
lapse, have an increased frequency of UTI due to incomplete bladder emptying.
16 Infections of Peritoneum, Mediastinum, Pleura, Wounds, and Urinary Tract 279
Classic symptoms of UTI in the adult are dysuria with accompanying urinary
urgency and frequency. A sensation of bladder fullness or lower abdominal dis-
comfort is often present. Bloody urine (hemorrhagic cystitis) is reported in as
many as 10% of cases of UTI in otherwise healthy women. Fevers, chills, and
malaise may be noted, though these are associated more frequently with pyelo-
nephritis. Most adult women with simple lower UTI have suprapubic tenderness
with no evidence of vaginitis, cervicitis, or pelvic tenderness. The patient with
pyelonephritis usually appears ill and, in addition to fever, sweating, and pros-
tration, is found to have flank tenderness in the majority of cases.
280 G. Sganga et al.
16.11.4 Diagnosis
16.11.5 Treatment
References
1. Bosscha K, van Vroonhoven JMV, van der Werken C (1999) Surgical management of
severe secondary peritonitis. Br J Surg 86:13711377
2. Sganga G, Brisinda G, Castagneto M (2001) Peritonitis: priorities and management
strategies. In: van Saene HKF, Sganga G, Silvestri L (eds) Infection in the critically ill: an
ongoing challenge. Springer, Berlin, pp 2333
3. Sganga G (2000) Sepsi addominali chirurgiche e insufficienza multiorgano (MOFS).
Edizioni Systems Comunicazioni, Milan
4. Bone RG, Balk RA, Cerra FB et al (1992) Definitions for sepsis and organ failure and
guidelines for the use of innovative therapies in sepsis. The ACCP/SCCM Consensus
Conference Committee. American College of Chest Physicians/Society of Critical Care
Medicine. Chest 101:16441655
5. Pieracci FM, Barie PS (2007) Intra-abdominal infections. Curr Opin Crit Care 13:440449
6. Solomkin JS, Mazuski JE, Baron EJ et al (2003) Guidelines for the selection of anti-
infective agents for complicated intra-abdominal infections. Clin Infect Dis 37:9971005
7. Brook I (2008) Microbiology and Management of Abdominal Infections. Dig Dis Sci
53:25852591
8. Krepel CJ, Gohr CM, Edmiston CE, Condon RE (1995) Surgical sepsis: constancy of
antibiotic susceptibility of causative organisms. Surgery 117:505509
9. Sganga G, Brisinda G, Castagneto M (2000) Nosocomial fungal infections in surgical
patients: risk factors and treatment. Minerva Anestesiol 66(Suppl 1):7177
10. Wheeler AP, Bernard GR (1999) Treating patients with severe sepsis. N Engl J Med
340:207214
11. Mannick JA, Rodrick ML, Lederer JA (2001) The immunologic response to injury. J Am
Coll Surg 193:237244
12. Knoferl MW, Angele MK, Diodato MD et al (2002) Female sex hormones regulate
macrophage function after trauma-hemorrhage and prevent increased death rate from
subsequent sepsis. Ann Surg 235:105112
16 Infections of Peritoneum, Mediastinum, Pleura, Wounds, and Urinary Tract 283
13. Sganga G, van Saene HKF, Brisinda G, Castagneto M (2001) Bacterial translocation. In:
van Saene HKF, Sganga G, Silvestri L (eds) Infection in the critically ill: an ongoing
challenge. Springer, Berlin, pp 3545
14. Gregor P, Prodger JD (1988) Mead Johnson Critical Care Symposium for the Practic
Surgeon. 4. Abdominal crisis in the intensive care unit. Can J Surg 31:331332
15. Gajic O, Errutia LE, Sewan H et al (2002) Acute abdomen in the medical intensive care
unit. Crit Care Med 30:11871190
16. Kollef MH, Allen BT (1994) Determinants of outcome for patients in the medical intensive
care unit requiring abdominal surgery: a prospective, single-center study. Chest
106:18221828
17. Velmahos GC, Kamel E, Berne TV et al (1999) Abdominal computed tomography for the
diagnosis of intra-abdominal sepsis in critically injured patients: fishing in murky waters.
Arch Surg 134:831836
18. Mokart D, Merlin M, Sannini A et al (2005) Procalcitonin, interleukin 6 and systemic
inflammatory response syndrome (SIRS): early markers of postoperative sepsis after major
surgery. Br J Anaesthesiol 94:767773
19. Wickel DJ, Cheadle WG, Mercer-Jones MA, Garrison RN (1997) Poor outcome from
peritonitis is caused by disease acuity and organ failure, not recurrent peritoneal infection.
Ann Surg 225:744756
20. Anbidge AE, Lynch D, Wison SR (2003) US of the peritoneum. Radiographics 23:663684
21. Go HL, Baarslag HJ, Vermeulen H et al (2005) A comparative study to validate the use of
ultrasonography and computed tomography in patients with post-operative intra-abdominal
sepsis. Eur J Radiol 54:383387
22. Whitehouse JS, Weigelt JA (2009) Diagnostic peritoneal lavage: a review of indications,
technique, and interpretation. Scandinavian J Trauma Resuscitation Emerg Med 17:13
23. Lin WY, Chao TH, Wang SJ (2002) Clinical features and gallium scan in the detection of
post-surgical infection in the elderly. Eur J Nucl Med 29:371375
24. Shih-Chuan T, Te-Hsin C, Lin WY, Shyh-Jen W (2001) Abdominal abscesses in patients
having surgery an application of Ga-67 scintigraphic and computed tomographic scanning.
Clin Nucl Med 26:761776
25. Marshall J, Maier RV, Jimenez M, Dellinger EP (2004) Source control in the management
of severe sepsis and septic shock: an evidence-based review. Crit Care Med 32(Suppl
11):S513S526
26. Soares-Weiser K, Paul M, Brezis M, Leibovici L (2002) Antibiotic treatment for
spontaneous bacterial peritonitis. BMJ 324:100102
27. Solomkin JS, Wilson SE, Christou N et al (2001) Results of a clinical trial of clinafloxacin
versus imipenem/cilastatin for intraabdominal infections. Ann Surg 233:7987
28. Mazuski JE (2007) Antimicrobial treatment for intra-abdominal infections. Expert Opin
Pharmacother 8(17):29332945
29. Laterre PF (2008) Progress in medical management of intra-abdominal infection. Curr Opin
Infect Dis 21(4):393398
30. Cohen J (2000) Combination antibiotic therapy for severe peritonitis. Lancet
356:15391540
31. Holzheimer RG, Dralle H (2001) Paradigm change in 30 years peritonitis treatment. A
review on source control. Eur J Med Res 6:161168
32. Schein M, Marshall J (2004) Source control for surgical infections. World J Surg
28(7):638645
33. Rivers E, Nguyen B, Havstad S et al (2001) Early goal-directed therapy in the treatment of
severe sepsis and septic shock. N Engl J Med 345:13681377
34. Platell C, Papadimitriou JM, Hall JC (2000) The influence of lavage on peritonitis. J Am
Coll Surg 191:672680
35. Sganga G, Brisinda G, Castagneto M (2002) Trauma operative procedures: timing of
surgery and priorities. In: Gullo A (ed) Critical care medicine. Springer, Berlin, pp 447467
284 G. Sganga et al.
36. Levison MA, Zeigler D (1991) Correlation of APACHE II score, drainage technique and
outcome in postoperative intra-abdominal abscess. Surg Gynecol Obstet 172:8994
37. Baril NB, Ralls PW, Wren SM et al (2000) Does an infected peripancreatic fluid collection
or abscess mandate operation? Ann Surg 231:361367
38. Brisinda G, Maria G, Ferrante A, Civello IM (1999) Evaluation of prognostic factors in
patients with acute pancreatitis. Hepatogastroenterology 46:19901997
39. Baron TH, Morgan DE (1999) Acute necrotizing pancreatitis. N Engl J Med 340:14121417
40. Beger HG, Rau B, Mayer J, Pralle U (1997) Natural course of acute pancreatitis. World J
Surg 21:130135
41. Bradley EL III (1993) A clinically based classification system for acute pancreatitis.
Summary of the international symposium on acute pancreatitis, Atlanta, GA, Sept. 1113,
1992. Arch Surg 128:586590
42. Marotta F, Geng TC, Wu CC, Barbi G (1996) Bacterial translocation in the course of acute
pancreatitis: beneficial role of nonabsorbable antibiotics and Lactinol enemas. Digestion
57:446452
43. Foitzik T, Fernandez-del Castillo C, Ferraro MJ et al (1995) Pathogenesis and prevention of
early pancreatic infection in experimental acute necrotizing pancreatitis. Ann Surg
222:179185
44. Isenmann R, Schwarz M, Rau B et al (2002) Characteristics of infection with Candida
species in patients with necrotizing pancreatitis. World J Surg 26(3):372376
45. Bradley EL III (1994) Surgical indications and techniques in necrotizing pancreatitis. In:
Bradley EL III (ed) Acute pancreatitis: diagnosis and therapy. Raven Press, New York,
pp 105117
46. Paye F, Rotman N, Radier C et al (1998) Percutaneous aspiration for bacteriological studies
in patients with necrotizing pancreatitis. Br J Surg 85:755759
47. Rau B, Pralle U, Mayer JM, Beger HG (1998) Role of ultrasonographically guided fine-
needle aspiration cytology in diagnosis of infected pancreatic necrosis. Br J Surg
85:179184
48. Mithofer K, Fernandez-del Castillo C, Ferraro MJ et al (1996) Antibiotic treatment improves
survival in experimental acute necrotizing pancreatitis. Gastroenterology 110:232240
49. Sainio V, Kemppainen E, Puolakkainen P et al (1995) Early antibiotic treatment in acute
necrotising pancreatitis. Lancet 346:663667
50. Luiten EJ, Hop WC, Lange JF, Bruining HA (1995) Controlled clinical trial of selective
decontamination for the treatment of severe acute pancreatitis. Ann Surg 222:5765
51. Luiten EJ, Hop WC, Lange JF, Bruining HA (1997) Differential prognosis of gram-negative
versus gram-positive infected and sterile pancreatic necrosis: results of a randomized trial in
patients with severe acute pancreatitis treated with adjuvant selective decontamination. Clin
Infect Dis 25:811816
52. Pederzoli P, Bassi C, Vesentini S, Campedelli A (1993) A randomized multicenter clinical
trial of antibiotic prophylaxis of septic complications in acute necrotizing pancreatitis with
imipenem. Surg Gynecol Obstet 176:480483
53. Ho HS, Frey CF (1997) The role of antibiotic prophylaxis in severe acute pancreatitis. Arch
Surg 132:487493
54. Bassi C, Falconi M, Talamini G et al (1998) Controlled clinical trial of pefloxacin versus
imipenem in severe acute pancreatitis. Gastroenterology 115:15131517
55. McClave SA, Snider H, Owens N, Sexton LK (1997) Clinical nutrition in pancreatitis. Dig
Dis Sci 42:20352044
56. Kalfarentzos F, Kehagias J, Mead N et al (1997) Enteral nutrition is superior to parenteral
nutrition in severe acute pancreatitis: results of a randomized prospective trial. Br J Surg
84:16651669
57. Windsor AC, Kanwar S, Li AG et al (1998) Compared with parenteral nutrition, enteral
feeding attenuates the acute phase response and improves disease severity in acute
pancreatitis. Gut 42:431435
16 Infections of Peritoneum, Mediastinum, Pleura, Wounds, and Urinary Tract 285
58. Bozzetti F, Braga M, Gianotti L et al (2001) Postoperative enteral versus parenteral nutrition
in malnourished patients with gastrointestinal cancer: a randomised multicentre trial. Lancet
358:14871492
59. Warshaw AL (2000) Pancreatic necrosis. To debride or not to debridethat is the question.
Ann Surg 232:627629
60. Buchler MW, Gloor B, Muller CA et al (2000) Acute necrotizing pancreatitis: treatment
strategy according to the status of infection. Ann Surg 232:619626
61. Farkas G, Marton J, Mandi Y, Szederkenyi E (1996) Surgical strategy and management of
infected pancreatic necrosis. Br J Surg 83:930933
62. Ashley SW, Perez A, Pierce EA et al (2001) Necrotizing pancreatitis. Contemporary
analysis of 99 consecutive cases. Ann Surg 234:572580
63. Gloor B, Muller CA, Worni M et al (2001) Pancreatic infection in severe pancreatitis. The
role of fungus and multiresistant organisms. Arch Surg 136:592596
64. Doglietto GB, Gui D, Pacelli F et al (1994) Open vs closed treatment of secondary
pancreatic infection. A review of 42 cases. Arch Surg 129:689693
65. Kriwanek S, Gschwantler M, Beckerhinn P et al (1999) Complications after surgery for
necrotising pancreatitis: risk factors and prognosis. Eur J Surg 165:952957
66. Dervenis C, Bassi C (2000) Evidence-based assessment of severity and management of
acute pancreatitis. Br J Surg 87:257258
67. Tsiotos GG, Luque-de Leon E, Sarr MG (1998) Long-term outcome of necrotizing
pancreatitis treated by necrosectomy. Br J Surg 85:16501653
68. del Castillo CF, Rattner DW, Makary MA et al (1998) Debridement and closed packing for
treatment of necrotizing pancreatitis. Ann Surg 228:676684
69. Farkas G (2000) Pancreatic head mass: how can we treat it? Acute pancreatitis: surgical
treatment. JOP J Pancreas 1(Suppl 3):138142
70. Mithofer K, Mueller PR, Warshaw AL (1997) Interventional and surgical treatment of
pancreatic abscess. World J Surg 21:162168
71. Widdison AL, Karanjia ND (1993) Pancreatic infection complicating acute pancreatitis. Br
J Surg 80:148154
72. Lumsden A, Bradley EL III (1990) Secondary pancreatic infections. Surg Gynecol Obstet
170:459467
73. Aranha GV, Prinz RA, Greenlee HB (1982) Pancreatic abscess: an unresolved surgical
problem. Am J Surg 144:534538
74. Warshaw AL, Jin G (1985) Improved survival in 45 patients with pancreatic abscess. Ann
Surg 202:408417
75. Brisinda G, Mazzari A, Crocco A et al (2011) Open pancreatic necrosectomy in the
multidisciplinary management of postinflammatory necrosis. Ann Surg 253(5):10491051
76. Ross Carter C, McKay CJ, Imrie CW (2000) Percutaneous necrosectomy and sinus tract
endoscopy in the management of infected pancreatic necrosis: an initial experience. Ann
Surg 232:175180
77. Kjossev KT, Losanoff JE (2001) Laparoscopic treatment of severe acute pancreatitis. Surg
Endosc 15:12391240
78. Freeny PC, Hauptmann E, Althaus SJ et al (1998) Percutaneous CT-guided catheter
drainage of infected acute necrotizing pancreatitis: techniques and results. AJR Am J
Roentgenol 170:969975
79. Baron TH, Morgan DE (1997) Organized pancreatic necrosis: definition, diagnosis, and
management. Gastroenterol Int 10:167178
80. Wada K, Takada T, Hirata K et al (2010) Treatment strategy for acute pancreatitis.
J Hepatobiliary Pancreat Sci 17:7986
81. Zhu JF, Fan XH, Zhang XH (2001) Laparoscopic treatment of severe acute pancreatitis.
Surg Endosc 15:146148
82. Clancy CJ, Nguyen MH, Morris AJ (1997) Candidal mediastinitis: an emerging clinical
entity. Clin Infect Dis 25:608613
286 G. Sganga et al.
83. Gamlin F, Caldicott LD, Shah MV (1994) Mediastinitis and sepsis syndrome following
intubation. Anaesthesia 49:883885
84. Isaacs L, Kotton B, Peralta MM Jr et al (1993) Fatal mediastinal abscess from upper
respiratory infection. Ear Nose Throat J 72:620622
85. Becker M, Zbaren P, Hermans R et al (1997) Necrotizing fasciitis of the head and neck: role
of CT in diagnosis and management. Radiology 202:471476
86. Brunelli A, Sabbatini A, Catalini G, Fianchini A (1996) Descending necrotizing
mediastinitis. Surgical drainage and tracheostomy. Arch Otolaryngol Head Neck Surg
122:13261329
87. Corsten MJ, Shamji FM, Odell PF et al (1997) Optimal treatment of descending necrotising
mediastinitis. Thorax 52:702708
88. Baldwin RT, Radovancevic B, Sweeney MS (1992) Bacterial mediastinitis after heart
transplantation. J Heart Lung Transpl 11:545549
89. El Oakley RM, Wright JE (1996) Postoperative mediastinitis: classification and
management. Ann Thorac Surg 61:10301036
90. Milano CA, Kesler K, Archibald N (1995) Mediastinitis after coronary artery bypass graft
surgery. Risk factors and long-term survival. Circulation 92:22452251
91. Brook I, Frazier EH (1996) Microbiology of mediastinitis. Arch Intern Med 156:333336
92. Shaffer HA Jr, Valenzuela G, Mittal RK (1992) Esophageal perforation. A reassessment of
the criteria for choosing medical or surgical therapy. Arch Intern Med 152:757761
93. Loop FD, Lytle BW, Cosgrove DM (1990) J Maxwell Chamberlain memorial paper. Sternal
wound complications after isolated coronary artery bypass grafting: early and late mortality,
morbidity, and cost of care. Ann Thorac Surg 49:179187
94. Gadek JE, DeMichele SJ, Karlstad MD (1999) Effect of enteral feeding with
eicosapentaenoic acid, gamma-linolenic acid, and antioxidants in patients with acute
respiratory distress syndrome. Enteral nutrition in ARDS study group. Crit Care Med
27:14091420
95. Weinzweig N, Yetman R (1995) Transposition of the greater omentum for recalcitrant
median sternotomy wound infections. Ann Plast Surg 34:471477
96. Andrews CO, Gora ML (1994) Pleural effusions: pathophysiology and management. Ann
Pharmacother 28:894903
97. Bartter T, Santarelli R, Akers SM (1994) The evaluation of pleural effusion. Chest
106:12091214
98. Fenton KN, Richardson JD (1995) Diagnosis and management of malignant pleural
effusions. Am J Surg 170:6974
99. Kennedy L, Sahn SA (1994) Noninvasive evaluation of the patient with a pleural effusion.
Chest Surg Clin N Am 4:451465
100. Light RW (1995) Pleural diseases, 3rd edn. Williams & Wilkins, New York
101. Sahn SA (1988) State of the art. The pleura. Am Rev Respir Dis 138:184234
102. Sganga G, Brisinda G, Castagneto M (2001) Practical aspects of antibiotic prophylaxis in
high-risk surgical patients. In: van Saene HKF, Sganga G, Silvestri L (eds) Infection in the
critically ill: an ongoing challenge. Springer, Berlin, pp 4758
103. Mangram AJ, Horan TC, Pearson ML et al (1996) The hospital infection control practices
advisory committee. Guideline for the prevention of surgical site infection. Infect Control
Hosp Epidemiol 20:247280
104. Sganga G, Cozza V (2009) Intra-abdominal infections: diagnostic and surgical strategies.
In: Gullo A, Besso J, Lumb PD, Williams GF (eds) Intensive and critical care medicine.
(WFSICCM) World federation of societies of intensive and critical care medicine. Springer,
Milan, pp 315324
105. Kurz A, Sessler DI, Lenhardt R (1996) Perioperative normothermia to reduce the incidende
of surgical-wound infection and shorten hospitalization. N Engl J Med 334:12091215
106. Classen DC, Evans RS, Pestotnik A (1992) The timing of prophylactic administration of
antibiotics and the risk of surgical-wound infection. N Engl J Med 326:281287
16 Infections of Peritoneum, Mediastinum, Pleura, Wounds, and Urinary Tract 287
107. Nathens AB, Marshall JC (1999) Selective decontamination of the digestive tract in surgical
patients. A systematic review of the evidence. Arch Surg 134:170176
108. Silvestri L, Mannucci F, van Saene HKF (2000) Selective decontamination of the digestive
tract: a life-saver. J Hosp Infect 45:185190
109. van Saene HKF, Silvestri L, de la Cal M (2000) Prevention of nosocomial infections in the
intensive care unit. Curr Opin Crit Care 6:323329
110. Greif R, Akca O, Horn EP et al (2000) Supplemental perioperative oxygen to reduce the
incidence of surgical-wound infection. N Engl J Med 342:161167
111. Jancel T, Dudas V (2002) Management of uncomplicated urinary tract infections. West J
Med 176:5155
112. Larcombe J (1999) Urinary tract infection in children. BMJ 319:11731175
113. Ellis AK, Verma S (2000) Quality of life in women with urinary tract infections: is benign
disease a misnomer? J Am Board Fam Pract 13:392397
114. Hooten TM, Scholes D, Stapleton AE (2000) A prospective study of asymptomatic
bacteriuria in sexually active young women. N Engl J Med 343:992997
115. Howes DS, Young WF (2000) Urinary tract infections. In: Tintinalli A (ed) Emergency
medicine. A comprehensive study guide. McGraw-Hill, New York, pp 625631
116. Leibovici L, Greenshtain S, Cohen O, Wysenbeek AJ (1992) Toward improved empiric
management of moderate to severe urinary tract infections. Arch Intern Med
152:24812486
117. Millar LK, Wing DA, Paul RH, Grimes DA (1995) Outpatient treatment of pyelonephritis in
pregnancy: a randomized controlled trial. Obstet Gynecol 86:560564
118. Safrin S, Siegel D, Black D (1988) Pyelonephritis in adult women: inpatient versus
outpatient therapy. Am J Med 85:793798
119. Stamm WE, Hooton TM (1993) Management of urinary tract infections in adults. N Engl J
Med 329:13281334
120. Foxman B, Barlow R, DArcy H et al (2000) Urinary tract infection: self-reported incidence
and associated costs. Ann Epidemiol 10:509515
121. Manges AR, Johnson JR, Foxman B et al (2001) Widespread distribution of urinary tract
infections caused by a multidrug-resistant Escherichia coli clonal group. N Engl J Med
345:10071013
122. Stark RP, Maki DG (1984) Bacteriuria in the catheterized patient. N Engl J Med
311:560564
123. Trautner BW, Darouiche RO (2004) Catheter-associated infections. Arch Intern Med
164:842850
124. Lo E, Lindsay N, Classen D et al (2008) Strategies to prevent catheter-associated urinary
tract infections in acute care hospitals. Infect Control Hosp Epidemiol 29:S41S50
Infection in the NICU and PICU
17
A. J. Petros, V. Damjanovic, A. Pigna and J. Farias
17.1 Introduction
Infections in neonates requiring intensive care are unique in each essential element
of the pathogenesis of infection, i.e. the potential pathogen and its source, the mode
of transmission and the susceptible host. The pathogen, e.g. hepatitis B virus, or
potential pathogen, e.g. Escherichia coli, are closely related to source and mode of
transmission. Many microorganisms are present in the maternal birth canal (the
source). They are most commonly Streptococcus agalactiae, E. coli, Herpes simplex
virus, Listeria monocytogenes and Candida albicans. One or more of these micro-
organisms can be vertically transmitted from the mother to the neonate. When this
type of infection occurs, it will always be present in the first week of the neonates
life (early onset). On the other hand, different microorganisms are acquired in
the NICU: in general, these are coagulase-negative staphylococci (CNS),
A. J. Petros (&)
PICU, Great Ormond Street Hospital, London, UK
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 289
DOI: 10.1007/978-88-470-1601-9_17, Springer-Verlag Italia 2012
290 A. J. Petros et al.
The overall infection rates in neonates on intensive care vary between 15 and 20%.
This is equal to rates reported for adult medical and surgical units and higher than
most paediatric units [10]. The main site of infection is the bloodstream, followed
by the lower airways. In a multicenter study of NIUCs in Oakland, New Haven,
CT, USA in 1994, Beck-Sague et al. [11] reported that nosocomial bloodstream
infection occurred at a rate of 5% when surveillance cultures were performed and
was actually half that reported in studies reporting the rate of all infections.
Bloodstream infections can account for 50% of all NICU infections. Lower airway
infections occur in approximately 3% of neonates during NICU stay [12]. The
main organisms are viruses, S. aureus and AGNB. The survival benefit of NICU
neonates has significantly increased over the last 25 years. In a 2-year study from
New York [19771978], the mortality rates for early-onset sepsis in neonates
\1,000 g was 53.4% and for late-onset sepsis 20.3% [13]. A 5-year study from
Oxford, UK (19821986) reports mortality rates of 28% and 4% for early- and
late-onset sepsis, respectively, in neonates [2]. Data from a 1-year study in a Dutch
NICU show a mortality rate \10% (1997) in 436 neonates of about 2,000 g [4].
children with a line in situ for a median of 6 days [16]. The causative organisms were
CNS, S. aureus, C. albicans and Klebsiella spp.
The magnitude of the problem can be assessed in different ways based upon carrier
state (Chap. 5). Endogenous must be distinguished from exogenous infection.
Endogenous infection is caused by potential pathogens previously carried by the
patient; if the potential pathogen was present on that patients admission, then the
infection due to this potential pathogen is called primary endogenous. This type of
infection tends to occur early, within the first week. If the infection is due to a
potential pathogen acquired in the unit, after the patient goes through the carriage
phase, then the infection is termed secondary endogenous. Infections caused by
microorganisms not carried by the patient at all are termed exogenous. Obviously,
surveillance cultures are indispensable for this classification [6, 9].
Some microorganisms cause more serious clinical disease than others. This dif-
ferential pathogenic effect can be used to develop a pathogenicity index for an
individual microorganism, in a specific organ system and in a particular homo-
geneous population for which surveillance cultures are useful [18]. The ratio
between the number of ICU patients infected by a particular microorganisms and
the number of patients simply carrying that organism in their throat and/or gut is
defined as the intrinsic pathogenicity index for a particular microorganism.
Indigenous flora, including anaerobes, will rarely cause infections in the lower
airways of patients requiring ventilation for more than 3 days despite being carried
in high concentrations. This is because they have intrinsic pathogenicity index
values of between 0.01 and 0.03. Low-level pathogens, such as viridans strepto-
cocci, enterococci and CNS are also carried in high concentrations in the
oropharynx by a substantial percentage of ICU patients and are unable to cause
lower airway infections. High-level pathogens such as S. pyogenes and Salmonella
spp. have an intrinsic pathogenicity index approaching 1 and diseases manifest in
virtually all oropharyngeal and gut carriers. The concept of carriage recognises
17 Infection in the NICU and PICU 293
Oropharynx Wound
N. mening
Line
Lung
S. aureus
S. pneum Skin
S. epi
S. epi
Candida
B Strep
E. coli
E. coli Colon
Bladder
Rectum
Fig. 17.1 Schematic representation of the digestive tract, illustrating that the throat and gut are
the major internal sources of potential pathogens causing endogenous infections of blood, lower
airways, bladder and wounds
17.5 Pathogenesis
study in 400 PICU children requiring ventilation demonstrated that 90% of all
lower airway infections were endogenous; 80% were primary endogenous, 10%
secondary endogenous and the remaining 10% exogenous [6].
Bloodstream infections occur due to translocation. Microorganisms in over-
growth of the terminal ileum ([105 microorganisms/ml) migrate into the blood-
stream [19]. This mechanism applies to S. agalactiae, S. aureus and C. spp.
Neonates and children staying longer that 1 week in the NICU or PICU, CNS and
AGNB cause septicaemia due to translocation [20].
Lower airway infections are caused by microorganisms carried in the oro-
pharynx, which then migrate into the lower airways. In a previous healthy child
S. pneumoniae, H. influenzae and S. aureus cause bacterial lower airway infec-
tions. AGNB and MRSA are causative organisms in children who require intensive
care [1 week.
Bladder infections are, in general, endogenous due to migrating faecal bacteria.
Wound infections of the head, neck and thorax are, in general, caused by oral
bacteria, whereas wound infections between the waist and knee are primarily
caused by gut bacteria.
Exogenous infections vary between 5% and 25% and are a particular problem in
patients with tracheostomies [21]. Children with wounds, particularly burns, are at
high risk of exogenous colonisation and infection [9]. Up to 16% of bloodstream
infections are of exogenous pathogenesis following contamination of an indwelling
intravascular device [19]. Gastrostomies can also be considered as a wound, and
recurrent exogenous colonisation/infection is not uncommon in children with such
devices [9]. To identify an exogenous infection, surveillance samples of throat and
rectum are indispensable. Blood cultures or lower airway secretions are positive for
a potential pathogen that is not present in throat and or rectal cultures.
Risk factor analysis in the pathogenesis of NICU and PICU infections invariably
includes low birth weight; administration of total parenteral nutrition; presence of
invasive and indwelling devices, including endotracheal tube and mechanical
ventilation; length of stay; and prior use of antibiotics [2, 22, 23]. All these factors
are reflected in illness severity and are difficult to modify to control infection. Risk
factor analysis cannot easily contribute to infection control.
17.6 Diagnosis
17.6.1 Infection
noninfectious conditions [2]. For instance, the clinical picture of respiratory dis-
tress in early onset sepsis may be identical to hyaline membrane disease.
Furthermore, the clinical diagnosis of local infection, such as meningitis, may not
differ from that of systemic sepsis without meningeal involvement. On the other
hand, infections in the PICU patient are more specific, and the following
description of local and general infection is related to paediatric patients.
17.6.2 Pneumonia
17.6.3 Tracheitis/Bronchitis
Infection of the urinary tract most often involves the bladder. The common fea-
tures of dysuria, suprapubic pain and urinary frequency and urgency are often not
assessable in PICU patients. Therefore, the diagnosis of cystitis is based upon
freshly obtained catheter urine containing C105 CFU/ml and C5 WBC/ml high
power light microscopy field.
17.7 Prevention
Besides the five infection-control interventions (Chap. 10), there is evidence for
the effectiveness of only two antibiotic manoeuvres that prevent infection in NICU
and PICU patients: surgical prophylaxis [25-27] and selective decontamination of
the digestive tract (SDD) (Table 17.1) [2833].
Table 17.1 Cardiac and general surgical prophylaxis and prevention protocol for selective
decontamination of the digestive tract (SDD)
The type of antimicrobial prescribed depends on the proposed surgery and the
associated contamination risk. Clean, sterile procedures do not need antibiotic
cover, whereas clean procedures with the likelihood of contamination need cover
with one antimicrobial, such as cefotaxime. If there is likely to be faecal con-
tamination, then an aminoglycoside such as gentamicin is also necessary to cover
AGNB and enterococci. Finally, if the surgical procedure is likely to be associated
with ischaemia and possible necrotic tissue, then metronidazole should be added to
the prophylactic regimen. Again, three doses as above will suffice.
SDD is a prophylactic intervention designed to prevent early and late infection and
is recommended in the critically ill child requiring [1 week of intensive care
(Chap. 14). Four prospective randomised controlled trials [2832] demonstrate a
significant reduction in infectious morbidity using SDD. As the overall mortality
rate in this population is approximately 10%, a reduction in that rate is harder to
demonstrate than in adults; a huge sample size would be necessary. However, in
adults, where the overall mortality rate is approximately 30%, this method dem-
onstrates a significant reduction of 40% [3335].
There is particular indication for SDD in the NICU, namely, for controlling an
infection outbreak. A decade ago, SDD with nystatin was used to control a
C. parapsilosis outbreak in the Mersey, UK, regional NICU: 76 of 106 neonates who
carried the outbreak strain received nystatin in the throat and gut during the
12-month open trial; six neonates developed fungaemia. Once the carriage rate fell
from 50 to 5%, no new cases of systemic Candida infection were observed. This was
the first report of SDD intervention to control an infection outbreak in an NICU [36].
Data was extracted from four randomized controlled trials (RCTs) of selective
digestive decontamination in the paediatric population (Table 17.2). The four
RCTs enrolled 335 patients. Pneumonia occurred in five of 170 (2.9%) of patients
17 Infection in the NICU and PICU 299
Table 17.2 Data extracted from four randomised controlled trials of selective decontamination
of the digestive tract in the paediatric population
Table 17.3 Meta-analysis of the impact of selective decontamination of the digestive tract on
secondary endpoints
Outcome RCTs No. of No. of patients with ORa (95 CI) p value I2 (%)
patients outcome
SDD C SDD C
Infection 3 54 55 10 24 0.34 (0.052.18) 0.25 4.7
Mortality 4 170 163 13 11 1.18 (0.502.76) 0.70 0
RCTs randomised controlled trials; OR odds ratio; CI confidence interval; SDD selective
decontamination of the digestive tract; C control
a
OR less than the unit favours treatment; OR more than the unit favours controls
who received SDD and in 16 of 163 (9.8%) in the control group. This was a
significant reduction in the incidence of pneumonia with SDD [odds ratio (OR)
0.31; 95% confidence interval (CI), 0.110.87; P = 0.027]. A meta-analysis was
performed on the impact of SDD on the secondary endpoints of pneumonia and
overall infection rates (Table 17.3). In the three eligible RCTs of a total of 109
children, infections of various origins were confirmed in ten of 54 (13%) on SDD
and in 24 of 55 (15.9%) in the control group. SDD had no impact on general
infection rates, with no significant difference between groups (OR 0.34; 95% CI
0.052.18; P = 0.25).
Subgroup analyses of type of SDD regimen, randomisation and blinding
revealed a significant impact on pneumonia and infection rates when the full
protocol of parenteral and enteral antimicrobials was used rather than solely
enterally administered antimicrobials. A significant impact on pneumonia and
overall infection was demonstrated when randomisation was adequate and in
unblinded studies. The subgroup analyses for mortality were consistent with
previous pooled results whether the intervention was parenteral/enteral or enteral,
whether the design was blinded or not and whether the randomisation process was
adequate or not. SDD made no significant impact on mortality rates.
300 A. J. Petros et al.
17.9 Treatment
cover AGNB and S. aureus. Late-onset infections are treated in the manner
described in Fig. 17.2.
When a child is admitted to the PICU with a severe infection, a decision must be
made as to which antimicrobial will be used. Antimicrobials are used in combi-
nation depending on the severity of the illness; our experience over 20 years led us
to choose cefotaxime and gentamicin. This choice is empirical due to the absence
of any knowledge regarding the causative microorganism, though reasonable
assumptions can be made depending of the childs presentation. For example, a
child with meningococcal disease requires cefotaxime only; metronidazole can be
added in case of presumed anaerobic involvement. When a presumptive identifi-
cation of the microorganism can be made, the physician can then tailor therapy.
Cefotaxime/gentamicin can be replaced by cephradine for an infection due to
S. pneumoniae, S. pyogenes and S. aureus. When P. aeruginosa is isolated, ceft-
azidime should replace cefotaxime and the gentamicin continued. Yeast infections
require liposomal amphotericin B in place of cefotaxime/gentamicin (Fig. 17.2).
The efficacy of the antimicrobial treatment can be monitored using C-reactive
protein (CRP) levels in addition to the clinical, radiographic and microbiological
variables. Providing the antimicrobials used are correct, the child will improve
within 3 days; in our experience, a short, 5-day course of intravenously admin-
istered antibiotics is as effective as a course of 2 weeks or more (Chap. 12). After
5 days, the child is monitored for signs of infection; when there are no signs of
infection, antibiotics are discontinued. Should there be no improvement after
5 days, a change in antibiotic regimen is necessary.
Metronidazole is given for 3 days only. The antifungal agent, liposomal
amphotericin B, is given for 3 weeks and may be discontinued once the CRP level
is normal. Systemic antimicrobials are combined with enterally administered SDD
agents to guarantee prevention of potential pathogens becoming resistant to the
systemic agents.
References
1. Damjanovic V, van Saene HKF (1998) Outbreaks of infection in a neonatal intensive care
unit. In: van Saene HKF, Silvestri L, de la Cal MA (eds) Infection control in the intensive
care unit. Springer, Milan, pp 237248
2. Isaacs D, Moxon ER (1991) Neonatal infections. Butterworth-Heinemann, Oxford
3. van Saene HKF, Leonard EM, Shears P (1989) Ecological impact of antibiotics in neonatal
units. Lancet II:509510
4. de Man P, Verhoeven BAN, Verburgh HA et al (2000) An antibiotic policy to prevent
emergence of resistant bacilli. Lancet 355:973978
5. Murphy N, Damjanovic V, Hart CA et al (1986) Infection and colonisation of neonates by
Hansenula anomala. Lancet I:291293
302 A. J. Petros et al.
6. Silvestri L, Sargison RE, Hughes J et al (2002) Most nosocomial pneumonias are not due to
nosocomial bacteria in ventilated patients: evaluation of the accuracy of the 48 h time cut-off
using carriage as the gold standard. Anaesth Intensive Care 30:275282
7. Petros AJ, OConnell M, Roberts C et al (2001) Systemic antibiotics fail to clear multidrug-
resistant Klebsiella from a paediatric ICU. Chest 119:862866
8. Goldmann DA, Leclair J, Macone A (1978) Bacterial colonization of neonates admitted to an
intensive care environment. J Pediatr 2:288293
9. Sarginson RE, Taylor N, Reilly N et al (2004) Infection in prolonged pediatric critical illness:
a prospective three year study based on knowledge of the carrier state. Crit Care Med
32(3):839847
10. Baltimore RS (1998) Neonatal nosocomial infections. Semin Perinatol 22:2532
11. Beck-Sague CM, Azimi P, Fonseca SN et al (1994) Bloodstream infections in neonatal
intensive care unit patients: results of a multicenter study. Pediatr Infect Dis J 13:11101116
12. Gaynes RP, Martone WJ, Culver DH et al (1991) Comparison of rates of nosocomial infections
in neonatal intensive care units in the United States. Am J Med 91(suppl 3B):192196
13. La Gamma EF, Drusin LM, Mackles AW et al (1983) Neonatal infections: an important
determinant of late NICU mortality in infants less than 1,000 g at birth. Am J Dis Child
137:838841
14. Richards MJ, Edwards JR, Culver DH, Gaynes RP, The National Nosocomial Infections
Surveillance System (1999) Nosocomial infection in pediatric intensive care units in the
United States. Pediatrics 103(4):e39
15. Gray J, Gossain S, Morris K (2001) Three-year survey of bacteraemia and fungemia in a
pediatric intensive care unit. Pediatr Infect Dis J 20:416421
16. Pierce CM, Wade A, Mok Q (2000) Heparin-bonded central venous lines reduce thrombotic
and infective complications in critically ill children. Intensive Care Med 26:967972
17. Patel JC, Mollitt DL, Pieper P, Tepas JJ III (2000) Nosocomial pneumonia in the pediatric
trauma patient: a single centers experience. Critical Care Med 28:35303533
18. Leonard EM, van Saene HKF, Stoutenbeek CP et al (1990) An intrinsic pathogenicity index
for micro-organisms causing infections in a neonatal surgical unit. Microb Ecol Health Dis
2:151157
19. van Saene HKF, Taylor N, Donnell SC et al (2003) Gut overgrowth with abnormal flora: the
missing link in parenteral nutrition-related sepsis in surgical neonates. Eur J Clin Nutrit
57:548553
20. Donnell SC, Taylor N, van Saene HKF et al (2002) Infection rates in surgical neonates and
infants receiving parenteral nutrition: a five year prospective study. J Hosp Infect 52:273280
21. Morar P, Singh V, Makura Z et al (2002) Differing pathways of lower airway colonization
and infection according to mode of ventilation (endotracheal vs. tracheostomy). Arch
Otolaryngol Head Neck Surg 128:10611066
22. Singh-Naz N, Sprague BM, Patel K et al (2000) Risk assessment and standardized
nosocomial infection rate in critically ill children. Crit Care Med 28:20692075
23. Mahieu LM, De Muynck AO, De Dooy JJ et al (2000) Prediction of nosocomial sepsis in
neonates by means of a computer-weighted bed side scoring system (NOSEP score). Crit
Care Med 28:20262033
24. ACourt CH, Garrard CS, Crook D et al (1993) Microbiological lung surveillance in
mechanically ventilated patients, using non-directed bronchial lavage and quantitative
culture. QJM 86:635638
25. Petros AJ, Marshall JC, van Saene HK (1995) Should morbidity replace mortality as an
endpoint for critical trials in intensive care? Lancet 345:369371
26. Kaiser AB (1986) Antimicrobial prophylaxis in surgery. New Engl J Med 315:11291138
27. Infection in Neurosurgery Working Party of the British Society for Antimicrobial
Chemotherapy (1994) Antimicrobial prophylaxis in neurosurgery and after head injury.
Lancet 344:15471551
17 Infection in the NICU and PICU 303
28. Zobel G, Kuttnig GHM et al (1991) Reduction of colonization and infection rate during
pediatric intensive care by selective decontamination of the digestive tract. Crit Care Med
19:12421246
29. Smith SD, Jackson RJ, Hannakan CJ et al (1993) Selective decontamination in pediatric liver
transplants. Transplantation 55:13061309
30. Ruza F, Alvarado F, Herruzo R et al (1998) Prevention of nosocomial infection in a pediatric
intensive care unit (PICU) through the use of selective decontamination. Eur J Epidemiol
14:719727
31. Barret JP, Jeschke MG, Herndon DN (2001) Selective decontamination of the digestive tract
in severely burned pediatric patients. Burns 27:439445
32. Alder Hey Therapeutic Guideline Index.http://www.nppg.scot.nhs.uk/protocols/therapeutic_
guidelines_index.htm
33. DAmico R, Pifferi S, Leonetti C et al (1998) Effectiveness of antibiotic prophylaxis in
critically ill adult patients: systematic review of randomized controlled trials. BMJ 316:
12751285
34. Nathens AB, Marshall JC (1999) Selective decontamination of the digestive tract in surgical
patients: a systematic review of the evidence. Arch Surg 134:7076
35. De Jonge E, Schultz MJ, Spanjaard L et al (2002) Effects of selective decontamination of the
digestive tract on mortality and antibiotic resistance. Intensive Care Med 28(Suppl 1):S12
36. Damjanovic V, Connolly CM, van Saene HKF et al (1993) Selective decontamination with
nystatin for the control of a Candida outbreak in the neonatal intensive care unit. J Hosp
Infect 24:245259
Early Adequate Antibiotic Therapy
18
R. Reina and M. A. de la Cal
18.1 Introduction
M. A. de la Cal (&)
Department of Intensive Care Medicine,
Hospital Universitario de Getafe, Getafe, Spain
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 305
DOI: 10.1007/978-88-470-1601-9_18, Springer-Verlag Italia 2012
306 R. Reina and M. A. de la Cal
The rationale for appropriate antibiotic therapy is to increase the likelihood that the
infective pathogen will be susceptible in vivo to the prescribed antimicrobial. It
implies the in vitro susceptibility to the antibiotic, delay between symptoms and
antibiotic administration, dose, and pharmacodynamics (PD) and pharmacokinet-
ics (PK) of antimicrobials. According to these premises, prescription of the ade-
quate antibiotic must taken into account:
18 Early Adequate Antibiotic Therapy 307
18.4 De-escalation
Table 18.2 Antibiotics, their killing activities, and pharmacokinetic and pharmacodynamic
(PK/PD) parameters
18.6 Conclusions
Appropriate early antibiotic treatment for ICU patients with infection is associated
with strong reduction in mortality and morbidity rates. At present, the challenge is
to achieve a level of adequate empirical therapy C90%. Combined therapy is only
justified as empirical treatment when the infection is suspected to be caused by
highly resistant microorganisms or in the most severely ill patients to provide a
higher likelihood of adequate treatment. De-escalation of the initial antibiotic
therapy must be encouraged because is saves money and reduces costs. The role
of biomarkers in helping to discontinue antibiotic therapy is usually marginal.
The PK/PD approach to prescribing the correct dose and route of administration is
a potential field of outcome research in humans and should be included in the
differential diagnosis of treatment failure.
312 R. Reina and M. A. de la Cal
References
1. Davey PG, Marwick C (2008) Appropriate vs. inappropriate antimicrobial therapy. Clin
Microbiol Infect 14(Suppl 3):1521
2. Pea F, Viale P (2009) Bench-to-bedside review: appropriate antibiotic therapy in severe
sepsis and septic shockdoes the dose matter? Crit Care 13:214
3. Kollef MH, Sherman G, Ward S, Fraser VJ (1999) Inadequate antimicrobial treatment of
infections: a risk factor for hospital mortality among critically ill patients. Chest 115:462474
4. Harbarth S, Garbino J, Pugin J et al (2003) Inappropriate initial antimicrobial therapy and its
effect on survival in a clinical trial of immunomodulating therapy for severe sepsis. Am J
Med 115:529535
5. Kumar A, Roberts D, Wood KE et al (2006) Duration of hypotension before initiation of
effective antimicrobial therapy is the critical determinant of survival in human septic shock.
Crit Care Med 34:15891596
6. Garnacho-Montero J, Ortiz-Leyba C, Herrera-Melero I et al (2008) Mortality and morbidity
attributable to inadequate empirical antimicrobial therapy in patients admitted to the ICU
with sepsis: a matched cohort study. J Antimicrob Chemother 61:436441
7. Barochia AV, Cui X, Vitberg D et al (2010) Bundled care for septic shock: an analysis of
clinical trials. Crit Care Med 38:668678
8. Dellinger RP, Levy MM, Carlet JM et al for the International Surviving Sepsis Campaign
Guidelines Committee; American Association of Critical-Care Nurses; American College of
Chest Physicians; American College of Emergency Physicians; Canadian Critical Care Society;
European Society of Clinical Microbiology and Infectious Diseases; European Society of
Intensive Care Medicine; European Respiratory Society; International Sepsis Forum; Japanese
Association for Acute Medicine; Japanese Society of Intensive Care Medicine; Society of
Critical Care Medicine; Society of Hospital Medicine; Surgical Infection Society; World
Federation of Societies of Intensive and Critical Care Medicine (2008) Surviving sepsis
campaign: international guidelines for management of severe sepsis and septic shock: 2008. Crit
Care Med 36(1):296327. Erratum in: Crit Care Med 36:13941396
9. Rivers E, Nguyen B, Havstad S, Early Goal-Directed Therapy Collaborative Group et al
(2001) Early goal-directed therapy in the treatment of severe sepsis and septic shock. N Engl
J Med 345:13681377
10. Shapiro NI, Howell MD, Talmor D et al (2006) Implementation and outcomes of the Multiple
Urgent Sepsis Therapies (MUST) protocol. Crit Care Med 34:10251032
11. Micek ST, Roubinian N, Heuring T et al (2006) Beforeafter study of a standardized hospital
order set for the management of severe septic shock. Crit Care Med 34:27072713
12. Nguyen HB, Corbett SW, Steele R et al (2007) Implementation of a bundle of quality
indicators for the early management of severe sepsis and septic shock is associated with
decreased mortality. Crit Care Med 35:11051112
13. El Solh AA, Akinnusi ME, Alsawalha LN, Pineda LA (2008) Outcome of septic shock in
older adults after implementation of the sepsis bundle. J Am Geriatr Soc 56:272278
14. Cunha BA (2008) Sepsis and septic shock: selection of empiric antimicrobial therapy. Crit
Care Clin 24:313334
15. Abad CL, Kumar A, Safdar N (2011) Antimicrobial therapy of sepsis and septic shock
when are two drugs better than one? Crit Care Clin 27:e1e27
16. Heyland DK, Dodek P, Muscedere J, Canadian Critical Care Trials Group et al (2008)
Randomized trial of combination versus monotherapy for the empiric treatment of suspected
ventilator-associated pneumonia. Crit Care Med 36:737744
17. Garnacho-Montero J, Sa-Borges M, Sole-Violan J et al (2007) Optimal management therapy
for Pseudomonas aeruginosa ventilator-associated pneumonia: an observational, multicenter
study comparing monotherapy with combination antibiotic therapy. Crit Care Med 35:
18881895
18 Early Adequate Antibiotic Therapy 313
18. Safdar N, Handelsman J, Maki DG (2004) Does combination antimicrobial therapy reduce
mortality in Gram-negative bacteraemia? A meta-analysis. Lancet Infect Dis 4:519527
19. Paul M, Leibovici L (2005) Combination antibiotic therapy for Pseudomonas aeruginosa
bacteraemia. Lancet Infect Dis 5:192193
20. Paul M, Benuri-Silbiger I, Soares-Weiser K, Leibovici L (2004) b-lactam monotherapy
versus b-lactam-aminoglycoside combination therapy for sepsis in immunocompetent
patients: systematic review and meta-analysis of randomised trials. BMJ 328:668. Erratum
in BMJ 328:884
21. Kumar A, Safdar N, Kethireddy S, Chateau D (2010) A survival benefit of combination
antibiotic therapy for serious infections associated with sepsis and septic shock is contingent
only on the risk of death: a meta-analytic/meta-regression study. Crit Care Med 38:
16511664
22. Niederman MS (2006) De-escalation therapy in ventilator-associated pneumonia. Curr Opin
Crit Care 12:452457
23. Dellit TH, Owens RC, McGowan JE et al (2007) Infectious Diseases Society of America and
the Society for Healthcare Epidemiology of America guidelines for developing an
institutional program to enhance antimicrobial stewardship. Clin Infect Dis 44:159177
24. Alvarez-Lerma F, Alvarez B, Luque P et al (2006) ADANN study group. Empiric broad-
spectrum antibiotic therapy of nosocomial pneumonia in the intensive care unit: a prospective
observational study. Crit Care 10:R78
25. Kollef MH, Morrow LE, Niederman MS et al (2006) Clinical characteristics and treatment
patterns among patients with ventilator-associated pneumonia. Chest 129:12101218.
Erratum in Chest 130:138
26. Singh N, Rogers P, Atwood CW et al (2000) Short-course empiric antibiotic therapy for
patients with pulmonary infiltrates in the intensive care unit. A proposed solution for
indiscriminate antibiotic prescription. Am J Respir Crit Care Med 162:505511
27. Christ-Crain M, Jaccard-Stolz D, Bingisser R et al (2004) Effect of procalcitonin-guided
treatment on antibiotic use and outcome in lower respiratory tract infections: cluster-
randomised, single-blinded intervention trial. Lancet 363:600607
28. Christ-Crain M, Stolz D, Bingisser R et al (2006) Procalcitonin guidance of antibiotic therapy in
community-acquired pneumonia: a randomized trial. Am J Respir Crit Care Med 174:8493
29. Stolz D, Smyrnios N, Eggimann P et al (2009) Procalcitonin for reduced antibiotic exposure
in ventilator-associated pneumonia: a randomised study. Eur Respir J 34:13641375
30. Nobre V, Harbarth S, Graf JD et al (2008) Use of procalcitonin to shorten antibiotic treatment
duration in septic patients: a randomized trial. Am J Respir Crit Care Med 177(5):498505
31. Pea F, Viale P, Furlanut M (2005) Antimicrobial therapy in critically ill patients: a review of
pathophysiological conditions responsible for altered disposition and pharmacokinetic
variability. Clin Pharmacokinet 44:10091034
32. Roberts JA, Lipman J (2009) Dose adjustment and pharmacokinetics of antibiotics in severe
sepsis and septic shock. In: Rello J, Kollef M, Diaz E, Rodriguez A (eds) Infectious diseases
in critical care, 2nd edn. Springer-Verlag, Berlin-Heidelberg, pp 122146
33. Petrosillo N, Drapeau CM, Agrafiotis M, Falagas ME (2010) Some current issues in the
pharmacokinetics/pharmacodynamics of antimicrobials in intensive care. Minerva Anestesiol
76(7):509524
34. Kasiakou SK, Lawrence KR, Choulis N, Falagas ME (2005) Continuous versus intermittent
intravenous administration of antibacterials with time-dependent action: a systematic review
of pharmacokinetic and pharmacodynamic parameters. Drugs 65:24992511
35. Roberts JA, Webb S, Paterson D et al (2009) A systematic review on clinical benefits of
continuous administration of beta-lactam antibiotics. Crit Care Med 37:20712078
ICU Patients Following Transplantation
19
A. Martinez-Pellus and I. Cortes Puch
19.1 Introduction
Solid organ and bone marrow transplant recipients are at high risk of infection due to
long-term immunosuppressant therapy that is necessary to help prevent transplant
rejection. Infection prevention and control in this patient population requires a
multifactorial approach using general and pharmacologic measures. In this chapter, we
discuss selective decontamination of the digestive tract (SDD) (polymyxin, tobra-
mycin, and amphotericin B) as a prophylactic measure during patient admission to the
intensive care unit (ICU); the protective effect of fluconazole and topical antifungals
drugs; systematic use of trimethoprim/sulfamethoxazole to manage Pneumocystis
jirovecii; and ganciclovir as a valid strategy to prevent cytomegalovirus (CMV)
infection. Another measure considered is shortening postoperative ICU length
of stay by setting quick weaning protocols, using noninvasive mechanical ventilation,
initiating physiotherapy, and early catheter removal. Long-term maneuvers strongly
depend on patient adherence to a prophylactic regimen over long periods, and
educational programs and involving patients in their prognosis help achieve
this goal.
Solid organ transplantation is a widespread procedure that has become the therapy
of choice for patients with irreversible and progressive end-stage organ disease.
Potential severe postoperative complications (related to the inflammatory response
A. Martinez-Pellus (&)
Intensive Care Unit, University Hospital Virgen de la Arrixaca,
Murcia, Spain
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 315
DOI: 10.1007/978-88-470-1601-9_19, Springer-Verlag Italia 2012
316 A. Martinez-Pellus and I. Cortes Puch
due to ischemia/reperfusion injury of the graft) and the risk of acute organ
rejection require close postoperative patient monitoring. This surveillance usually
takes place in the intensive care unit (ICU). In the early post-transplant period,
patients will frequently require mechanical ventilation and invasive devices,
putting them at risk for a large variety of infections. Infections at this stage are
frequent [1] and have significant consequences on the prognosis, exceeding the
surgical complications as a cause of mortality [2]. The primary goal in the
transplant recipient is to avoid organ rejection. This requires achieving an adequate
state of immunosuppression, known as net state of immunosuppression [3], which
refers to all factors that contribute to the patients risk of infection. Its major
determinants are immunosuppressive treatment dose, regimen, and duration. This
state can also be affected by the presence of neutropenia, comorbidities, and
concomitant infections with immunomodulating viruses, particularly cytomega-
lovirus (CMV), Epstein-Barr virus (EBV), and hepatitis B and C viruses. When
some of these factors occur in a single patient, excessive immunosuppression takes
place, affecting humoral and cellular mechanisms and exposing the patient to
infection. On the other hand, the environmental exposure of the transplant reci-
pient to a large variety of potentially infectious agents (viruses, fungi, community-
acquired pathogens, endogenous flora, etc.) is continuous and takes place in the
community as well as in the hospital. Other pathogens that scarcely provoke
community-acquired infections in the immunocompetent host can lead to devas-
tating syndromes in the transplant recipient. This is the case with respiratory
viruses, fungi, and mycobacteria. Hence, the application of preventive strategies
for long periods is necessary. If infection takes places despite these preventive
measures, it will represent a serious problem, leading to hospitalization, complex
diagnostic procedures, and empirical broad-spectrum antibiotic treatment. Current
immunosuppressive regimens have been reached by relative consensus. They
always include steroids, together with cyclosporine or tacrolimus, and myco-
phenolate or azathioprine, in different combinations. There is evidence that links
all of these drugs with several opportunistic pathogens, such as P. jirovecii and
Aspergillus (steroids), reactivation and replication of latent viruses (cyclosporine
and tacrolimus), and CMV and bacterial infections (mycophenolate). The risk of
infection after transplantation changes over time, especially with modifications in
immunosuppression. The maximal immunosuppressive effect of these regimens
takes place after several months of treatment. Therefore, we can establish a pre-
dictable timeline of the specific infections after transplantation according to
the level of immunosuppression achieved (Table 19.1). Exceptions to this timeline
are so rare that they suggest either a massive environmental exposure or excessive
immunosuppression. This predictable pattern in which infections occur allows
the establishment of specific prophylactic measures against them. These pre-
ventive strategies must start before surgery and continue during prolonged periods
or even for life.
19 ICU Patients Following Transplantation 317
Table 19.1 Timing of infectious complications and preventive measures after transplantation
testing for CMV, EBV, Toxoplasma gondii, and syphilis. If the tuberculin skin test is
positive, the measures that should be taken are controversial due to the potential
hepatotoxicity of the tuberculostatics and their interferences with the pharmacoki-
netics of tacrolimus and cyclosporine. Periodic negative clinical and radiologic
evaluations should rule out the need for prophylaxis. Vaccination against tetanus,
diphtheria, mumps, influenza, pneumococcus, Haemophilus influenza type B, and
hepatitis B virus must be performed during the pretransplant period if they were not
previously completed [4]. Live vaccines are generally contraindicated after trans-
plantation, as they may trigger an infection or facilitate organ rejection. Varicella-
zoster vaccination should also be performed if the patient is seronegative, although
its effectiveness is doubted [5]. The importance of these measures lies in the fact that
all of these infections may be latent in the patient and undergo reactivation during
immunosuppression. The transplant recipient must not have a proven or suspected
infection by the time of surgery. Respiratory carriage of pathogens such as
Aspergillus or Burkholderia cepacia must be ruled out or eradicated before surgery.
In the case of Aspergillus, some studies demonstrate a significant reduction in the rate
of infections in lung or cardiac transplant recipients who received amphotericin B
aerosols as prophylaxis [6]. Nevertheless, the detection of this fungus in a respiratory
sample may only indicate colonization and, therefore, the interpretation of this
microbiological data is difficult. In an attempt to establish the predictive value of
Aspergillus isolates, a study revealed that cultures with more than two colonies or
more than one site of infection were predictive of significant infection and portended
a poor prognosis and development of invasive disease [7].
Surgical complexity and, particularly, the impact of cold ischemia times and
consequent graft reperfusion, as well as the risk of systemic complications, leads to
a close immediate postoperative management that usually takes place in the ICU.
The probability of ICU hospitalization is highest during the first month following
transplantation, when infectious complications are similar to those that occur in an
immunocompetent host undergoing surgery. They are comparable in site (surgical
wound infection, urinary tract infection, catheter-related bloodstream infections,
etc.) and etiology (typical nosocomial pathogens such as Staphylococcus aureus,
coagulase-negative staphylococcus, and Gram-negative bacilli, to which fungi are
progressively added). Prophylactic measures must be directed to these expected
microorganisms. However, we must be aware that patients with previous pro-
longed hospitalizations while waiting for transplantation may become colonized
with hospital-acquired antimicrobial-resistant organisms (Pseudomonas,
methicillin-resistant S. aureus (MRSA), Aspergillus, etc.).
patients. The results showed that SDD significantly reduced the incidence of
infection in transplant organ recipients [odds ratio (OR) 0.44; 95% confidence
interval (CI) 0.230.87]. Mortality rates were not significantly altered (OR 0.29;
95% CI 0.061.47). In a randomized placebo-controlled trial performed in a small
group of liver transplant patients, Zwaveling et al. [17] found a similar incidence
of infections in both groups but with a significant reduction in those caused by
aerobic Gram-negative bacilli (p \ 0.01) and Candida spp (p \ 0.05) in the SDD
group. In a recent meta-analysis [18] that reviewed four randomized trials com-
paring SDD versus placebo or no treatment at all, the overall incidence of infection
was similar despite the use of SDD (OR 0.88; CI 95%: 0.71.1). Nevertheless,
SDD was able to reduce the incidence of infections caused by aerobic Gram-
negative bacilli (OR 0.16; CI 95% 0.070.37) and the incidence of postoperative
pneumonias. SDD regimens should be adjusted, however, to the most prevalent
pathogens found in each ICU, bearing in mind their particular resistance profile.
The use of probiotics during the pretransplant period could represent an alter-
native to SDD. In a randomized trial, Rayes et al. [19] compared the administration
of a fiber-containing formula plus living Lactobacillus plantarum 299 versus SDD.
They observed a reduction in the incidence of bacterial infections from 48 to 13%,
with early enterally provided nutrition with the probiotics. These findings should
be validated in further studies, as this is the first study performed in this field, and
it is not powered high enough to draw solid conclusions.
recipient) received ganciclovir i.v. (5 mg/kg per day) for 510 days and then either
acyclovir po (400 mg t.i.d.) or ganciclovir po (1 g t.i.d.) for an additional
12 weeks. Treatment with ganciclovir po was associated with a significant
decrease in the incidence of CMV symptomatic disease when compared with the
acyclovir po group (32 vs. 50%; p \ 0.05) [30]. In another trial comparing the
different strategies of cytomegalovirus prophylaxis in liver transplant recipients,
ganciclovir po was found to be the most cost-effective strategy [31]. In a recent
systematic revision [32], prophylactic treatment with antivirals was found to be
associated with a significant decrease in CMV infection [relative risk (RR) 0.62;
95% CI 0.530.73; p \ 0.001) and disease (RR 0.51; 95% CI 0.410.64;
p \ 0.001) compared with placebo or no treatment. This revision failed to show
significant differences in the incidence of organ rejection or mortality rates.
Another meta-analysis [33] of 1,980 patients from 17 controlled trials raises
similar conclusions, showing a significant decrease in the incidence of CMV
disease (OR 0.20; 95% CI 0.130.31) and the rate of allograft rejection. CMV
infection is potentially severe but rarely requires ICU hospitalization. Regardless,
this disease should be considered with pneumonias presenting with an atypical
course, particularly in patients immunosuppressed with mycophenolate.
Beyond the sixth month after transplantation, the organ recipient achieves a stable
degree of immunosuppression, and infectious problems at this stage are similar to those
that take place in the immunocompetent host. However, patients with chronic rejec-
tion who need a strong and maintained state of immunosuppression, are vulnerable to
opportunistic infections (P. jirovecii, Listeria monocytogenes, Nocardia asteroides,
Aspergillus, Cryptococcus neoformans, etc.) [3]. This raises the need for prolonged
prophylaxis with cotrimoxazole and antifungals. Some of these prophylactic strategies
are already well defined. The use of daily single-dose cotrimoxazole, for example,
during the 412 months following transplantation, has nearly eradicated the incidence
of P. jirovecii and has decreased the rate of infections caused by L. monocytogenes,
N. asteroides, and T. gondii in high-risk patients such as cardiac transplant recipients.
In this late post-transplant period, the risk of tuberculosis (reactivation of a
latent infection) is 2070 times higher than in the general population. In a large
series of 4,634 solid organ transplant recipients, Garca-Goetz et al. [34] found an
incidence of 0.45%, but it can reach 6% in endemic areas [35]. Mortality rates of
these patients can be as high a 30%. According to this, patients with risk factors for
tuberculosis (latent tuberculosis, familiar exposure, or radiologic suspicion) should
receive prophylaxis with isoniazid at least for 912 months, starting early after
transplantation.
Table 19.2 Infectious episodes during intensive care unit (ICU) stay (transplanted patients vs
overall ICU)
last 7 years shows a global incidence of early infection of 17.5%, which is similar
to the incidence in other severely ill surgical patients and lower than in the rest of
the patients admitted with other diagnoses (29%). According to this analysis, there
was no difference in the site and incidence of infections between transplant
recipients and the rest of the patients, with exception of peritonitis, which was
more frequent in liver transplant recipients (1.8 vs. 0.8 per 1,000 days of stay)
(Table 19.2). According to etiology, the analysis showed no difference between
groups regarding prevailing bacterial infections over fungal and viral ones
(Table 19.3). A low incidence of multi-drug-resistant pathogens was found, with
the exception of Acinetobacter baumannii, which was more frequent in the
transplant recipients (9 vs. 4.3%; p \ 0.005). Aerobic Gram-negative bacilli were
more frequent in the group of nontransplant patients (50 vs. 44%; p \ 0.05),
whereas CMV infection rates were higher in transplant recipients (4 vs. 0%;
p \ 0.001). In a univariate analysis, the risk factors of infection were acute liver
failure or code 0 previous to transplantation, Acute Physiology and Chronic
Health Evaluation (APACHE) II score at admission, the need for reintervention for
any cause, early organ rejection, and pretransplant ICU length of stay. The rates of
days with mechanical ventilation and with central venous catheters were also
higher in patients with infection. In a regression analysis, the only risk factors
for infection found were code 0 as the cause of transplantation (OR 7.5, 95% CI
2.819.5), prolonged mechanical ventilation (OR 11.9, 95% CI 6.123.2), and
324 A. Martinez-Pellus and I. Cortes Puch
Table 19.3 Microbiology of infectious episodes in patients admitted to the intensive care unit
(ICU) after transplantation versus patients admitted for other reason (each infectious episode
could involve several microorganisms)
APACHE II score [16 at the admission (OR 2.19, 95% CI 1.14.1). Length of
ICU stay and mortality rate were highly conditioned by infection (Table 19.4).
Managing these infections should not be difficult, always bearing in mind the
characteristic pathogens of each center and their resistance profile.
Infection
In the rest of the transplant recipients, the most frequent infections during this late
period after transplantation are bloodstream and respiratory tract infections. The
latter can have an atypical clinical or radiologic pattern, frequently requiring
invasive diagnostic procedures and computed tomography (CT) [36]. Acute epi-
sodes with a radiologic pulmonary consolidation suggest bacterial infections
(including Legionella), whereas diffuse edema-like infiltrates usually correspond
to a viral processes. Subacute and chronic episodes are mainly caused by fungi,
Nocardia, tuberculosis, P. jiroveci, and viruses. According to our experience with
27 solid organ transplant recipients admitted to our ICU with severe sepsis, most
episodes had a bacterial etiology (five E. coli, three pneumococcus, three
Pseudomonas, one Corynebacterium, and one MRSA); only three cases of CMV
disease where diagnosed. Two patients had influenza H1N1 (during the 2009
pandemia), and in three cases of pneumonia, cultures yielded no results. Baseline
characteristics of the patients where similar to those of transplant recipients
admitted for any other cause (Table 19.5). The epidemiology of infections was
compared between patients with ICU-acquired infections in the immediate post-
transplant period and previously transplanted patients who were admitted to the
ICU with other diseases. This comparison showed no significant differences in the
site and number of infectious episodes between groups (Table 19.6).
During this late post-transplant period, the risk of reactivation of latent tuber-
culosis is still present. This situation is challenging as a result of drug interactions
between tuberculostatics and immunosuppressive agents. Rifampin, for example,
decreases tacrolimus and cyclosporine levels. This pharmacokinetic interaction
can lead to organ rejection. In these cases, treatment regimens should be based in a
combination of other tuberculostatics (isoniazid ? ethambutol ? pyrazinamide,
for example) and may include the possibility of using fluoroquinolones and
streptomycin if the course is torpid.
Unlike solid organ transplant patients, bone marrow transplant recipients are never
admitted to the ICU. Nevertheless, they require pretransplant treatment with high-
dose radiotherapy, chemotherapy, or both, in order to abolish the patients immune
system. This causes a severe neutropenia as well as damaging the integrity
of mucosal barriers, favoring bacterial translocation from the digestive tract.
Consequently, infections will be a frequent problem in these patients [37]. Bone
marrow recipients need at least 24 months to become immunocompetent. This
period may be even longer for allogenic transplants. This fact, as well as the wide
etiological spectrum (bacteria, viruses, and fungi) seen in these patients, hinders
the establishment of preventive strategies. Even though many infections of the
bone marrow recipient have an intestinal origin, prophylactic strategies do not
include SDD or systemic antibiotics in the asymptomatic patient due to the lack of
evidence of their effectiveness. Some studies in which fluoroquinolones were
administered as a prophylactic measure during the period of neutropenia showed a
19 ICU Patients Following Transplantation 327
Table 19.6 Infectious episodes during ICU stay (admission post-transplant vs. transplant, with
admission for other reason)
19.4 Conclusion
Solid-organ and bone marrow transplant recipients are at high risk for infection in
the short and medium term. This risk depends on the level of immunosuppression
necessary to avoid rejection. Preventive maneuvers aimed at minimizing this
problem have gradually increased, and most of them have proven to be effective.
The first goal in a global infection prevention plan is to minimize length of ICU
stay. This goal can be achieved by implementing quick weaning protocols,
19 ICU Patients Following Transplantation 329
removing central lines and drainages as soon as possible, and establishing inten-
sive physiotherapy programs. The fact that in an early phase the epidemiology of
infections is comparable in transplant recipients and other surgical patients
admitted to the ICU could justify the prophylactic use of SDD. This might be
particularly important while the patient is undergoing mechanical ventilation or
until a minimum level of immunosuppression can be achieved (cyclosporine
monotherapy, for example). The use of SDD could also be supported by the
emergence of fungal infections (mainly Candida spp.), favored by the use of
steroids in immunosuppressive protocols. In the liver transplant recipient, several
studies have demonstrated a protective effect of both fluconazole and topical
enterally administered nonabsorbable antifungals as part of SDD. The risk of
selecting non-albicans Candida spp. resistant to azoles, observed with the use of
fluconazole, is not likely to exist with topical use of amphotericin B. SDD has been
the subject of intense debate given the fact that it has no effect on post-transplant
mortality rates. However, this reason should not be enough to refuse treatment
with the potential benefit of avoiding infection. To resolve this controversial issue,
a study with a complex design and a large sample size is necessary. However, it is
unlikely that this kind of study will ever be performed.
In the medium- and long-term periods, the problems concerning prevention rely
on the patients willingness to maintain a long prophylactic regimen, together with
the risk of toxicity, interactions with immunosuppressive agents, and selection of
resistant pathogens. Reported rates of nonadherence to the medical recommen-
dations after transplantation are unacceptably high in some studies, reaching seven
to 36 cases of nonadherence to immunosuppressive treatment per 100 patients/year
[44]. Educational programs and greater patient participation in the prognosis are
useful tools to achieve this goal.
Regarding the profile of infections in transplant recipients, there are three
general time frames to consider: the first month (early postoperative period), the
second through the sixth month, and the late post-transplant period (beyond
the sixth month or first year). In the early postoperative period, infections are
similar in site and etiology to those occurring in general surgical patients.
The intermediate period (26 months after transplantation), is when the risk
of opportunistic infections (P. jirovecii and, less frequently, Listeria and Nocar-
dia), as well as reactivation of latent infections (tuberculosis, CMV, Toxoplasma)
become manifest. Therefore, these etiologies should be ruled out when a transplant
recipient is admitted with an infectious episode. Beyond the first year after
transplantation, if the level of immunosuppression is minimal, the recipient has the
same risk of infection as the general population, with the exception of the possi-
bility of a reactivation of latent tuberculosis. Although it is unusual that patients
with these infections of the late post-transplant period are admitted to the ICU,
they can raise a diagnostic challenge. A high index of suspicion is necessary to
choose the appropriate diagnostic procedures (serological tests, virus polymerase
chain reaction, biopsies, etc.) and to start directed treatment as soon as possible;
the patients prognosis will markedly depend on this.
330 A. Martinez-Pellus and I. Cortes Puch
References
1. Patel R, Paya CV (1997) Infections in solid-organ transplant recipients. Clin Microb Rev
10:86124
2. Torbenson M, Wang J, Nichols L et al (1998) Causes of death in autopsied liver
transplantation patients. Mod Pathol 11:3746
3. Fishman JA (2007) Infection in solid-organ transplant recipients. N Engl J Med 357:
26012614
4. Avery RK (2004) Prophylactic strategies before solid-organ transplantation. Curr Opin Infect
Dis 17:353356
5. White CJ (1997) Varicela-zoster virus vaccine. Clin Infect Dis 24:753
6. Reichenspurner H, Gamberg P, Nitschke M et al (1997) Significant reduction in the number
of fungal infections after lung, heart-lung, and heart transplantation using aerosolized
amphotericin B prophylaxis. Transpl Proc 29:627628
7. Brown RS, Lake JR, Katzman BA et al (1996) Incidence and significance of Aspergillus
cultures following liver and kidney transplantation. Transplantation 61:666669
8. Liberati A, DAmico R, Pifferi S, Brazzi L (2007) Antibiotic prophylaxis to reduce
respiratory tract infections and mortality in adults receiving intensive care. Cochrane Review.
Cochrane Library, Issue 3
9. Wiesner RH, Hermans PE, Rakela J et al (1988) Selective bowel decontamination to decrease
gram-negative aerobic bacterial and Candida colonization and prevent infection after
orthotopic liver transplantation. Transplantation 45:570574
10. Smith SD, Jackson RJ, Hannakan CJ et al (1993) Selective decontamination in pediatric liver
transplants. A randomized prospective study. Transplantation 55:13061309
11. 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 22:4049
12. van Saene JJM, Stoutenbeek CP, van Saene HKF et al (1996) Reduction of the intestinal
endotoxin pool by three different SDD regimens in human volunteers. J Endotoxin Res
3:337343
13. Emre S, Sebastian A, Chodoff L et al (1999) Selective decontamination of the digestive tract
helps prevent bacterial infections in the early postoperativ period after liver transplant. Mt
Sinai J Med 66:310313
14. van Enckevort PJ, Zwaveling JH, Bottema JT et al (2001) Cost effectiveness of selective
decontamination of the digestive tract in liver transplant patients. Pharmacoeconomics 19:523530
15. Garcia-San Vicente B, Canut A, Labora A et al (2010) Descontaminacin digestiva selectiva:
respercusin en la carga de trabajo y el coste de laboratorio de microbiologa y tendencias en
la resistencia bacteriana. Enferm Infecc Microbiol Clin 28:7581
16. Nathens AB, Marshall JC (1999) Selective decontamination of the digestive tract in surgical
patients: a systematic review of the evidence. Arch Surg 134:170176
17. Zwaveling JH, Maring JK, Klompmaker IJ et al (2002) Selective decontamination of the
digestive tract to prevent posoperative infection: a randomized placebo-controlled trial in
liver transplant patients. Crit Care Med 30:12041209
18. Sadfar 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 7:817827
19. Rayes N, Seehofer D, Hansen S et al (2002) Early enteral supply of lactobacillus and fiber
versus selective bowel decontamination: a controlled trial in liver transplant recipients.
Transplantation 74:123127
20. Mor E, Cohen J, Erez E et al (2001) Short intensive care unit stay reduces septic
complications and improves outcome after liver transplantation. Transplant Proc 33:
29392940
19 ICU Patients Following Transplantation 331
21. Antonelli M, Conti G, Bufi M et al (2000) Noninvasive ventilation for treatment of acute
respiratory failure in patients undergoing solid organ transplantation: a randomized trial.
JAMA 283:235241
22. Whiteman K, Nachtmann L, Kramer D et al (1995) Effects of continuous lateral rotation
therapy on pulmonary complications in liver transplant patients. Am J Crit Care 4:133139
23. Cole GT, Halawa AA, Anaissie EJ (1996) The role of the gastrointestinal tract in the
hematogenous candidiasis: from the laboratory to bedside. Clin Infect Dis 22(Suppl 2):
S73S88
24. Garrido RS, Aguado JM, Diaz-Pedroche C et al (2006) A review of critical periods for
opportunistic infection in the new transplantation era. Transplantation 82:14571462
25. Winston DJ, Pakrasi A, Busuttil RW (1999) Prophylactic fluconazole in liver transplant
recipients. A randomized, double-blind, placebo-controlled trial. Ann Int Med 131:729737
26. Fortun J, Martin-Davila P, Moreno S et al (2003) Prevention of invasive fungal infections in
liver transplant recipients: the role of prophylaxis with lipid formulations of amphotericin B
in high risk patients. J Antimicrob Chemother 52:813819
27. Biancofiore G, Bindi ML, Baldasarri R et al (2002) Antifungal prophylaxis in liver transplant
recipients: a randomized placebo-controlled study. Transpl Int 15:341347
28. Fernandez A, Amezquita Y, Fernandez-Tarrago E et al (2009) Prophylaxis and treatment of
cytomegalovirus infection postrenal transplantation in two Madrid units. Transpl Proc
41:24162418
29. Arthur SK, Pedersen RA, Kremers WK et al (2009) Delayed-onset primary cytomegalovirus
disease and the risk of allograph failure and mortality after kidney transplantation. Clin Infect
Dis 46:840846
30. Rubin RH, Kemmerly SA, Conti D et al (2000) Prevention of primary cytomegalovirus
disease in organ transplant recipients with oral ganciclovir or oral acyclovir prophylaxis.
Transpl Infect Dis 2:112117
31. Das A (2000) Cost-effectiveness of different strategies of cytomegalovirus prophylaxis in
orthotopic liver transplant recipients. Hepatology 31:311317
32. Couchoud C (2001) Cytomegalovirus prophylaxis with antiviral agents for solid organ
transplantation. Cochrane Database of Systematic Reviews, Issue 4
33. Kalil AC, Levitsky J, Lyden E et al (2005) Meta-analysis: the efficacy of strategies to prevent
organ disease by cytomegalovirus in solid organ transplant recipients. Ann Int Med 143:
870880
34. Garca-Goetz JF, Linares L, Benito N et al (2009) Tuberculosis in solid organ transplant
recipients at a tertiary hospital in the last 20 years in Barcelona, Spain. Transpl Proc 41:
22682270
35. Benito N, Sued O, Moreno A et al (2002) Diagnosis and treatment of latent tuberculosis
infection in liver transplant recipients in an endemic area. Transplantation 74:13811386
36. Torres A, Ewing S, Insausti J et al (2000) Etiology and microbial patterns of pulmonary
infiltrates in patients with orthotopic liver transplantation. Chest 117:502949
37. Kernan NA, Bartsch G, Ash RC et al (1993) Analysis of 462 transplantations from unrelated
donors facilitated by the National Marrow Donor Program. N Engl J Med 328(9):593
38. Cruciani M, Rampazzo R, Malena M et al (1996) Prophylaxis with fluoroquinolones for
bacterial infection in neutropenic patients: a meta-analysis. Clin Infect Dis 23:795
39. Marr KA, Siedel K, Slavin MA et al (2000) Prolonged fluconazole prophylaxis is associated
with persistent protection against candidiasis-related death in allogenic marrow transplant
recipients: long-term follow-up of a randomized, placebo-controlled trial. Blood 96:2055
40. Abi-Said D, Anaissie E, Uzun O et al (1997) The epidemiology of hematogenous candidiasis
caused by different Candida species. Clin Infect Dis 24:1122
41. Guiot HFL, Fibbe WE, Vant Wout JW (1996) Prevention of invasive candidiasis by
fluconazole in patients with malignant hematological disorders and a high grade of candida
colonisation. (Abstract LM33) 36th Interscience conference on antimicrobial agentes and
chemotherapy, New Orleans
332 A. Martinez-Pellus and I. Cortes Puch
42. Schmidt GM, Horak DA, Niland JC et al (1991) A randomized, controlled trial of
prophylactic ganciclovir for cytomegalovirus pulmonary infections in recipients of allogenic
bone marrow transplants: the City of Hope-Standford-Syntex CMV Study Group. N Engl J
Med 324:10051011
43. Hughes WT, Armstrong D, Bedey GP et al (1997) 1997 Guidelines for the use of
antimicrobials agents in neutropenic patients with unexplained fever. Clin Infect Dis
25(3):551
44. Dew A, Di Martni AF, De Vito A (2007) Rates and risk factors for nonadherence to the
medical regimen after adult solid organ transplantation. Transplantation 83:858873
Clinical Virology in NICU, PICU
and AICU 20
C. Y. W. Tong and S. Schelenz
20.1 Introduction
Viruses are significant causes of nosocomial infections, but their importance has
been underappreciated in the past. However, outbreak of severe acute respiratory
syndrome (SARS), avian and pandemic influenza with high morbidity and
mortality rates increased the awareness of intensivists regarding the devastating
effects of nosocomial spread of viral infections in intensive care units (ICU).
Advances in medicine have led to a large number of immunocompromised
patients susceptible to severe viral infections. Many such patients are cared for in
the ICU and in turn become an infectious hazard for other vulnerable patients.
Health care workers can acquire common viral infections from the community and
spread them to susceptible patients in the ICU. Patients in neonatal (NICU) and
paediatric (PICU) ICUs are most vulnerable because of the lack of prior immunity
against many viruses circulating in the community. Recent improvements in
diagnostic methods have enabled the rapid diagnosis and monitoring of many viral
infections. Rapid and accurate typing of viral strains using a molecular technique
can help identify the source of outbreaks. Also, specific postexposure prophylaxis
and treatment are now available for many important nosocomial viral infections.
In this chapter, we discuss some of the important viruses that could be associated
with nosocomial infections in the ICU, according to their usual route of trans-
mission (Table 20.1). Infection control measures recommended for preventing
these viral infections are listed in Table 20.2.
C. Y. W. Tong (&)
Infection, Guys and St Thomas NHS Foundation Trust
and Kings College London School of Medicine, London, UK
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 333
DOI: 10.1007/978-88-470-1601-9_20, Springer-Verlag Italia 2012
334 C. Y. W. Tong and S. Schelenz
Table 20.1 Mode of viral infection transmission in the intensive care unit
Virus Isolation or cohorting Hand Apron/ Gloves Masks/ Incubation Duration of infectivity
washing gown+ goggles
Influenza viruses 4(negative pressure) IB 4 4 4 4 IB 14 days Prodromal phase and 7 days after onset
Respiratory 4 II 4 IA 4 IB 4 IA 28 days 48 h before symptoms and 7 days from
syncytial viruses onset; longer in immunocompromised (up
(RSV) to 30 days)
Parainfluenza 4 4 4 4 24 days As long as symptoms last
viruses
Adenovirus 4 4 4 4 510 days As long as symptoms last
SARS coronavirus 4 (negative pressure) IA 4 4 4 4 (FFP3 46 days (max. Peak at day 10 of illness, no reported
or N95) reported transmission 10 days beyond resolution of
IB 14 days) fever
Clinical Virology in NICU, PICU and AICU
Varicella-zoster 4(negative pressure) IB 4 4 4 1021 days 2 days before first vesicle until all lesions
virus (chickenpox) are crusted
Rotavirus 4 4 4 4 23 days 2 days before symptoms and up to
47 days after onset of illness
Norovirus 4 IB 4 IA 4 IB 4 IB 1548 h Up to 48 h after becoming symptom free
Enterovirus/ 4 (young infants often 4 4 4 225 days 714 days from onset of illness;
parechovirus require care in SCBU or asymptomatic shedding common
NICU)
Hepatitis A virus 4 4 4 4 26 weeks Infectious 1 week before onset of illness,
infectivity declines rapidly after onset of
illness
Hepatitis B virus 4 4 23 months As long as patient is viremic
(continued)
335
Table 20.2 (continued)
336
Virus Isolation or cohorting Hand Apron/ Gloves Masks/ Incubation Duration of infectivity
washing gown+ goggles
Hepatitis C virus 4 4 23 months As long as patient is viremic
Human 4 4 36 weeks Indefinitely, though viremia can be
immunodeficiency controlled by therapy
virus (HIV)
Viral hemorrhagic 4 (high security 4 4 4 4 321 days High infectivity during illness
fever viruses isolation)
(VHF)
Cytomegalovirus 4 4 4 36 weeks Congenital infectionfrom birth.
(CMV) (congenital (congenital from primary Asymptomatic shedding common
CMV) CMV) infection.
Herpes simplex 4 4 Often due to Until lesions have healed
virus (HSV) reactivation
Varicella-zoster 4 4 4 Due to Until vesicles crusted over
virus (shingles) reactivation
Rabies virus 4 4 4 4 4 28 weeks or Duration of illness
longer
Categorisation of recommendations: IA strongly recommended for all hospitals and strongly supported by well-designed experimental or epidemiological
studies; IB strongly recommended for all hospitals and viewed as effective by experts in the field (these recommendations are based on strong rationale and
suggestive evidence, even though scientific studies may not have been performed). II suggested for implementation in many hospitals (these recommen-
dations may be supported by suggestive clinical or epidemiological studies, a strong theoretical rationale or definitive studies applicable to some but not all
hospitals
FFP3 or N95 high-filtration respirators, SCBU special care baby unit, NICU neonatal intensive care unit, 4 recommended and suggested for implementation
in most settings, SARS severe acute respiratory distress syndrome, HIV human immune deficiency virus, VHF viral haemorrhagic fevers, CMV
cytomegalovirus
C. Y. W. Tong and S. Schelenz
20 Clinical Virology in NICU, PICU and AICU 337
There were three influenza pandemics in the last century, of which the pandemic in
1918 due to the H1N1 virus was the most severe. The first pandemic of this
century occurred in 2009 [1] and was due to another H1N1 variant that emerged
through a quadruple reassortment of viral RNAs derived from human, avian,
Eurasian and North American swine influenza sources [2]. The presence of animal
influenza subtypes, particularly avian influenza viruses such as H5N1, is of con-
tinuous concern, as these could be the source of future pandemics. Though with
relatively high case-fatality rate, H5N1 avian influenza virus has so far only caused
a limited number of human infections in restricted geographical locations with
little evidence of human to human spread. However, the 2009 pandemic H1N1
virus proved to be a major burden for ICU staff [3].
Clinically, influenza infection is characterised by abrupt onset of fever, sore
throat, myalgia, cough, headache and malaise. Young children may develop croup,
pneumonia or middle ear infection. With seasonal influenza, complications are
often seen in the elderly, the immunocompromised and those with pre-existing
chronic heart or lung disease or diabetes. During the 2009 H1N1 pandemic,
children and young adults were more susceptible [4]. Overall fatality rate was
\0.5%, but as many as 931% of hospitalised patients needed ICU admission [5].
Severe disease and high mortality rates were seen in pregnant women, patients
with underlying medical pulmonary, cardiac, metabolic, neuromuscular illness and
severe obesity, and those in whom the diagnosis and admission was delayed [68].
Respiratory failures could be caused by viral pneumonia and acute respiratory
distress syndrome (ARDS). In addition, secondary bacterial infection with Strep-
tococcus pneumoniae or Staphylococcus aureus (often methicillin resistant) were
found in 2024% of ICU patients and 2638% of patients who died [3, 5, 9]. Fatal
cases were often complicated by multiorgan failure.
Influenza has a short incubation time of 14 days. The virus is transmitted via
droplets, and patients are infectious during the prodromal phase and up to 7 days
after symptom onset. Rapid antigen detection from respiratory secretions is
available, but this was found to be insensitive for the 2009 H1N1 pandemic virus
[10]. More sensitive and specific real-time polymerase chain reaction (PCR)
methods had to be used [11]. Due to the infection-control hazards of taking
nasopharyngeal aspirates or bronchoalveolar lavage, the use of throat and nasal
swabs were advocated. A complete respiratory diagnostic workup needed to be
performed to exclude other viral, bacterial and noninfectious causes. A single
negative influenza PCR result on an upper respiratory sample did not definitively
exclude the diagnosis [12]. In addition, other concurrent or secondary infections
had to be considered. Protocols needed to be in place to ensure satisfactory triage
of patients according to severity [13]. Early administration of specific neuramin-
idase inhibitors, such as oral doses of oseltamivir or inhalation zanamivir, seemed
to be beneficial [14]. In more refractory cases, the off-license use of intravenously
administered zanamivir or peramivir was tried. Extracorporeal membrane
oxygenation (ECMO) was found to be useful in very severe cases [12].
The risk of nosocomial transmission to other hospitalised patients and staff is
well documented. Infected patients should ideally be cared for in a single room
338 C. Y. W. Tong and S. Schelenz
the practice of enhanced seasonal infection control programs for RSV has been
shown to be effective [21]. The usefulness of wearing masks and goggles is less clear.
There is no safe and effective vaccine to prevent RSV infection. However,
immunoprophylaxis in the form of RSV immunoglobulin (RSV-IG) or humanised
monoclonal antibodies (palivizumab) is available as prophylaxis for some high-
risk patients to prevent serious RSV disease or to limit further nosocomial spread.
Both palivizumab and RSV-IG have been shown to decrease the incidence of RSV
hospitalisation and ICU admission, although there was no significant reduction in
the risk of mechanical ventilation or mortality rate. When given prophylaxis,
infants born\35 weeks gestational age and those with chronic lung and congenital
heart disease all had a significant reduction in the risk of RSV hospitalisation [22].
Treating RSV infection is mainly supportive, including oxygen, ventilation and
bronchodilatative drugs. Aerosolised ribavirin has often been used in severe cases,
with or without gamma globulin i.v. [23]. However, evidence for the clinical
efficacy of ribavirin in RSV infection remains inconclusive [24]. The use of
aerosolised ribavirin needs to be carefully controlled, as there are potential
teratogenic effects on pregnant staff and visitors. Others have tried a combination
of palivizumab i.v. with or without ribavirin [25].
Another paramyxovirus, known as human metapneumovirus (hMPV), shares a
similar spectrum of clinical illness as RSV. It is likely that general infection
control measures against RSV would also be effective against hMPV.
There are four types of human parainfluenza virus (PIV) types: PIV 14 (family
Paramyxoviridae). Infections with PIV1 and 2 are seasonal, with a peak in autumn
affecting mainly children between 6 months and 6 years of age. Clinically,
patients often present with croup or a febrile upper respiratory tract infection.
In contrast, PIV3 is endemic throughout the year and infects mostly young infants
in the first 6 month of life and up to 2 years of age. Clinically, there is no specific
presentation in PIV3, but bronchiolitis and pneumonia are not uncommon.
In immunocompromised adults, such as stem cell transplant recipients, PIV3 is
associated with a high mortality rate. Such patients often present with severe
pneumonia and many require admission to the ICU.
The diagnosis of PIV infection can be confirmed by immunofluorescence
antigen detection or NAAT [26]. Nosocomial transmission is often due to PIV3
and has been documented in neonatal care and adult haematology units [27].
Infection control precautions are the same as for RSV. Despite several uncon-
trolled case series of apparent successful use of intravenously, orally or aerosolised
administration of ribavirin to treat PIV infections, there is no clear evidence that
ribavirin with or without immunoglobulin alters mortality rates from PIV3
pneumonia or decreases the duration of viral shedding from the nasopharynx [28].
Nevertheless, there may be a role for pre-emptive early therapy with ribavirin to
prevent progression of upper airway infection to pneumonia.
340 C. Y. W. Tong and S. Schelenz
20.2.4 Adenovirus
20.3.1 Rotavirus
20.3.2 Norovirus
be in contact with live vaccine poliovirus shedding infants should ensure that they
are immunised.
Rigorous hand washing (Table 20.3) is the most important measure during an
outbreak. Cohort nursing, source isolation and screening are other measures fre-
quently used (Table 20.3). Clearance of the virus by the host is antibody-mediated
and many have advocated the use of normal human immunoglobulin (NHIG).
Table 20.4 Body fluids that may pose a risk for hepatitis B and C virus (HBV, HCV) and human
immunodeficiency virus (HIV) after significant exposure
Amniotic fluid
Breast milk
Cerebrospinal fluid
Exudate from burns or skin lesions
Pericardial fluid
Peritoneal fluid
Pleural fluid
Saliva after dental treatment
Synovial fluid
Unfixed tissues or organs
Any other fluid if visibly blood stained
Saliva, urine, vomitus or stool that are not blood stained are not considered as high risk for blood
borne viruses
HBV is the most infectious of the three common blood-borne viruses. The risk of
transmission depends on the viral load of the source patient. An HBV-infected
individual with hepatitis B e antigen (HBeAg) tends to have a high viral load
and is therefore more infectious than carriers without HBeAg. Estimate of infec-
tivity ranges from 2% (HBeAg absent) to 40% (HBeAg present). All health care
workers should be immunised against HBV. Exposed health care workers who are
susceptible (not immunised or vaccine nonresponders) should receive hepatitis B
immunoglobulin for postexposure prophylaxis. A booster dose of vaccine should
be given to those exposed individual who had previously been successfully
immunised.
Table 20.5 Example of points for sharps safety education (adapted from the Infection Control
Team of Guys and St. Thomas NHS Foundation Trust, London)
Table 20.6 Example of actions to be taken immediately after a blood/body fluid exposure
(adapted from the Infection Control Team of Guys and St. Thomas NHS Foundation Trust,
London)
12 weeks for HCV RNA testing and promptly referred for treatment if found
infected.
Table 20.7 Viruses responsible for viral haemorrhagic fevers with nosocomial concern in the
intensive care unit
Viral haemorrhagic fevers (VHFs) are severe and life-threatening diseases caused
by a range of viruses. They are either zoonotic or arthropod-borne infections and
are often endemic in certain parts of the world. They are often highly infectious
through close contact with infected blood and body fluid and therefore pose a
significant risk of hospital-acquired infection. As many patients with VHF
present with shock and require vigorous supportive treatment, it is a potential
problem in the ICU. The major viruses of nosocomial concern in this setting are
Marburg, Ebola, Rift Valley fever, Lassa and Crimean Congo haemorrhagic
fever (Table 20.7). The incubation period for these VHFs ranges from
321 days. Initial symptoms are often nonspecific but may eventually lead to
haemorrhage and shock. Any febrile patient who has returned from an endemic
area of one of the VHF agents or has a history of contact with cases suspected to
have VHF within 3 weeks should be considered as at risk. However, malaria
should always be excluded. A risk assessment needs to be performed, and any
patient known or strongly suspected to be suffering from VHF should be
admitted to a high-security infectious disease unit that is designed to manage
these patients. While awaiting transfer to a secure unit, such patients should be
placed in a negative-pressure room with strict source isolation. Specimens for
patient management should be processed in a high-security laboratory designated
for category 4 pathogens, and the aetiological agent established using PCR,
serology and virus culture. All areas and materials in contact with infected
patients should be autoclaved, incinerated or treated with hypochlorite
(10,000 ppm of available chlorine). If the patient dies, the body should be placed
in a sealable body bag sprayed or wiped with hypochlorite. Individuals who have
been in contact with a case of VHF should be put under surveillance for
3 weeks. The successful i.v. use of ribavirin has been reported in some cases of
VHFs (Lassa, Crimean Congo haemorrhagic fever and Hantaan). Apart from
yellow fever, no vaccines are available.
348 C. Y. W. Tong and S. Schelenz
Shingles or zoster is the result of the reactivation of latent VZV (family Herpes-
viridae) in the dorsal root or cranial nerve ganglia. The clinical presentation is a
painful vesicular eruption covering the affected dermatome. The clinical diagnosis
can be confirmed rapidly by immunofluorescence, electron microscopy or PCR of
the cellular material obtained from a vesicular scraping. The infection is usually
self-limiting but can be more severe in immunocompromised patients, in whom it
may present over multiple dermatomes or as a disseminated infection. The latter
cases should be managed as if they were chickenpox, and respiratory precautions
for infection control have to be enforced.
Patients or health care staff members with classic shingles are contagious from
the day the rash appears until the lesions are crusted over. There is some risk of
nosocomial transmission if the lesions are on exposed areas of the body or in
immunocompromised infected patients. Nonimmune (VZV-IgG negative) patients
or health care staff members with no history of chickenpox are susceptible if
they have close contact with shingles and should be managed as described for
chickenpox contact.
The herpes simplex virus (HSV) (family Herpesviridae) consist of two types:
HSV-1 and HSV-2. Clinically, they most commonly manifest with oral (mainly
HSV-1) or genital (mainly HSV-2) ulcerations/vesicles, and reactivation is com-
mon, particularly in the ICU. Other presentations include keratitis, encephalitis,
meningitis, herpetic whitlow or neonatal infection.
The diagnosis can be confirmed rapidly by immunofluorescence, electron
microscopy or PCR of vesicle/ulcer scrapings. In the immunocompromised
patient, HSV can cause life-threatening disseminated infection and, early treatment
with acyclovir i.v. is recommended. It has also been suggested that occult herpes
virus reactivation may increase the mortality risk of ICU patients [54].
As the infected lesions contain virus, there is an increased risk of nosocomial
transmission until the lesions have crusted over. Standard isolation precautions
should be in place to reduce transmission (Table 20.2). Patients with active lesions
should be nursed away from high-risk patients (i.e. immunocompromised, severe
eczema, burns, or neonates). As patients can be asymptomatic secretors, health
care workers should wear gloves when dealing with mucosal secretions (i.e. saliva)
to avoid infections such as herpetic whitlow. Infected staff should cover lesions if
possible and should not attend those at risk.
Neonatal herpes is usually transmitted from mother to the child at the time of
delivery and may not be noticed until the infant develops the disease. Universal
precautions, in particularly, hand washing, should always be in place to reduce
20 Clinical Virology in NICU, PICU and AICU 349
20.6 Summary
Viral infection can cause significant morbidity and mortality and has the potential
to result in cross infection, involving patients as well as health care workers. Good
infection-control practice is essential to prevent nosocomial infection. Intensivists
should be on the alert for important viruses causing infections according to age
group of patients and mode of transmission and should never be complacent. Good
liaison with the laboratory is essential for determining correct diagnostic tests and
timely report of results to help in patient management.
References
1. Fitzgerald DA (2009) Human swine influenza A. Paediatr Respir Rev 10:154158
2. Trifonov V, Khiabanian H, Rabadan R (2009) Geographic dependence, surveillance, and
origins of the 2009 influenza A (H1N1) virus. N Engl J Med 361:115119
3. Webb SA, Pettila V, Seppelt I et al (2009) Critical care services and 2009 H1N1 influenza in
Australia and New Zealand. N Engl J Med 361:19251934
4. Itoh Y, Shinya K, Kiso M et al (2009) In vitro and in vivo characterization of new swine-
origin H1N1 influenza viruses. Nature 460:10211025
5. Writing committee of the WHO consultation on clinical aspects of pandemic (H1N1) 2009
Influenza (2010) Clinical aspects of pandemic 2009 Influenza A (H1N1) virus infection.
N Engl J Med 362:17081719
6. Siston AM, Rasmussen SA, Honein MA et al (2010) Pandemic 2009 influenza A(H1N1)
virus illness among pregnant women in the United States. JAMA 303:15171525
350 C. Y. W. Tong and S. Schelenz
27. Maziarz RT, Sridharan P, Slater S et al (2010) Control of an outbreak of human parainfluenza
virus 3 in hematopoietic stem cell transplant recipients. Biol Blood Marrow Transpl
16:192198
28. Nichols WG, Corey L, Gooley T et al (2001) Parainfluenza virus infections after
hematopoietic stem cell transplantation: risk factors, response to antiviral therapy, and
effect on transplant outcome. Blood 98:573578
29. Lenaerts L, De Clercq E, Naesens L (2008) Clinical features and treatment of adenovirus
infections. Rev Med Virol 18:357374
30. Williams KM, Agwu AL, Dabb AA et al (2009) A clinical algorithm identifies high risk
pediatric oncology and bone marrow transplant patients likely to benefit from treatment of
adenoviral infection. J Pediatr Hematol Oncol 31:825831
31. Ksiazek TG, Erdman D, Goldsmith CS et al (2003) A novel coronavirus associated with
severe acute respiratory syndrome. N Engl J Med 348:19531966
32. Poutanen SM, Low DE, Henry B et al (2003) Identification of severe acute respiratory
syndrome in Canada. N Engl J Med 348:19952005
33. Lau EH, Hsiung CA, Cowling BJ et al (2010) A comparative epidemiologic analysis of
SARS in Hong Kong, Beijing and Taiwan. BMC Infect Dis 10:50
34. Wong SS, Yuen KY (2008) The management of coronavirus infections with particular
reference to SARS. J Antimicrob Chemother 62:437441
35. Cheng VC, Lau SK, Woo PC, Yuen KY (2007) Severe acute respiratory syndrome coronavirus
as an agent of emerging and reemerging infection. Clin Microbiol Rev 20:660694
36. Cameron JC, Allan G, Johnston F et al (2007) Severe complications of chickenpox in
hospitalised children in the UK and Ireland. Arch Dis Child 92:10621066
37. Aly NY, Al Obaid I, Al-Qulooshi N, Zahed Z (2007) Occupationally related outbreak of
chickenpox in an intensive care unit. Med Princ Pract 16:399401
38. Apisarnthanarak A, Kitphati R, Tawatsupha P et al (2007) Outbreak of varicella-zoster virus
infection among Thai healthcare workers. Infect Control Hosp Epidemiol 28:430434
39. Fruhwirth M, Heininger U, Ehlken B et al (2001) International variation in disease burden of
rotavirus gastroenteritis in children with community- and nosocomially acquired infection.
Pediatr Infect Dis J 20:784791
40. Verboon-Maciolek MA, Krediet TG, Gerards LJ et al (2005) Clinical and epidemiologic
characteristics of viral infections in a neonatal intensive care unit during a 12-year period.
Pediatr Infect Dis J 24:901904
41. Boyce JM, Pittet D (2002) Guideline for hand hygiene in health-care settings.
Recommendations of the healthcare infection control practices Advisory Committee and
the HIPAC/SHEA/APIC/IDSA Hand Hygiene Task Force. Am J Infect Control 30:S1S46
42. Cunliffe NA, Both JA, Lowe SJ et al (2010) Healthcare-associated viral gastroenteritis among
children in a large pediatric hospital, United Kingdom. Emerg Infect Dis 16:5562
43. Armbrust S, Kramer A, Olbertz D et al (2009) Norovirus infections in preterm infants: wide
variety of clinical courses. BMC Res Notes 2:96
44. Nix WA, Maher K, Pallansch MA, Oberste MS (2010) Parechovirus typing in clinical
specimens by nested or semi-nested PCR coupled with sequencing. J Clin Virol 48:202207
45. Harvala H, Robertson I, Chieochansin T et al (2009) Specific association of human
parechovirus type 3 with sepsis and fever in young infants, as identified by direct typing of
cerebrospinal fluid samples. J Infect Dis 199:17531760
46. Takami T, Sonodat S, Houjyo H et al (2000) Diagnosis of horizontal enterovirus infections in
neonates by nested PCR and direct sequence analysis. J Hosp Infect 45:283287
47. Gupta S, Fernandez D, Siddiqui A et al (2010) Extensive white matter abnormalities associated
with neonatal Parechovirus (HPeV) infection. Eur J Paediatr Neurol 14(6):531534
48. Benschop KS, Schinkel J, Minnaar RP et al (2006) Human parechovirus infections in Dutch
children and the association between serotype and disease severity. Clin Infect Dis 42:204210
49. Hanna JN, Loewenthal MR, Negel P, Wenck DJ (1996) An outbreak of hepatitis A in an
intensive care unit. Anaesth Intensive Care 24:440444
352 C. Y. W. Tong and S. Schelenz
50. Victor JC, Monto AS, Surdina TY et al (2007) Hepatitis A vaccine versus immune globulin
for postexposure prophylaxis. N Engl J Med 357:16851694
51. Evans B, Duggan W, Baker J et al (2001) Exposure of healthcare workers in England, Wales,
and Northern Ireland to bloodborne viruses between July 1997 and June 2000: analysis of
surveillance data. BMJ 322:397398
52. Maheshwari A, Thuluvath PJ (2010) Management of acute hepatitis C. Clin Liver Dis
14:169176
53. Cardo DM, Culver DH, Ciesielski CA et al (1997) A case-control study of HIV
seroconversion in health care workers after percutaneous exposure. Centers for Disease
Control and Prevention Needlestick Surveillance Group. N Engl J Med 337:14851490
54. Cook CH, Martin LC, Yenchar JK et al (2003) Occult herpes family viral infections
are endemic in critically ill surgical patients. Crit Care Med 31:19231929
AIDS Patients in the ICU
21
F. E. Arancibia and M. A. Aguayo
21.1 Introduction
In the early 1980s in the United States, the first medical reports described
outbreaks of Kaposis sarcoma and Pneumocystis carinii (now P. jiroveci)
pneumonia in homosexual men [1, 2]. These reports rate respiratory infections by
opportunistic germs and rare tumors affecting healthy young men. High mortality
rates were observed that were most likely caused by acute respiratory failure
(ARF). Additionally, each report had abnormal ratios of lymphocyte subgroups.
Subsequent reports described an escalating frequency of unusual infections and
tumors, suggesting a profound state of immune suppression in many homosexual
men, injection-drug users, sexual partners of infected persons, hemophiliacs who
had received blood transfusions, and children.
In early 1983, virologists at the Pasteur Institute first isolated the human immu-
nodeficiency virus (HIV) [3], a retrovirus that infects cells of the immune system
(subgroup of CD4+ T cells), destroying or impairing their function. The most
advanced stage of HIV infection was called AIDS. In the early years, the ICU
survival rate of patients with AIDS was low [4]. Based on the belief that ICU care of
patients with AIDS was futile, clinical, ethical, and economic issues were raised
regarding the benefits and burdens of the critical care of these patients. However, the
use of P. carinii pneumonia (PCP) prophylaxis, antiretroviral therapy, and cortico-
steroids for PCP has changed outcomes. Several studies have shown improved
survival rates and costs of HIV-infected patients admitted to the ICU [58].
F. E. Arancibia (&)
Unidad de Cuidados Intensivos,
Instituto Nacional Trax, Santiago, Chile
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 353
DOI: 10.1007/978-88-470-1601-9_21, Springer-Verlag Italia 2012
354 F. E. Arancibia and M. A. Aguayo
In the United States and Europe, from 4 to 11% of HIV patients hospitalized
require admission to the ICU. All studies show that respiratory failure has
remained the most common indication for an ICU admission [5, 8, 11, 12].
Other frequent indications are sepsis and neurological compromise (Table 21.1).
Occasionally, patients with severe gastrointestinal symptoms, cardiomyopathies
and/or complications such as lactic acidosis and pancreatitis may require intensive
care. Finally, these patients may have clinical or surgical conditions requiring ICU
admission unrelated to their HIV/AIDS. HIV-infected patients admitted to the ICU
following trauma, elective surgery, or gastrointestinal bleeding may have as good a
prognosis as patients without HIV infection [9].
However, opportunistic illness rates declined precipitously after the intro-
duction of highly active antiretroviral therapy (HAART) and stabilized at low
levels in the subsequent years. Buchacz et al. [13] reported that during
19942007, rates of opportunistic infections (per 1,000 person-years) decreased
from 89 to 13.3 and rates of opportunistic malignancies were from 23.4 to 3.0
(Fig. 21.1). Due to the dramatic improvement in the prognosis of HIV-infected
patients, our notion of HIV as a disease has transformed from a rapidly fatal
diagnosis to a treatable chronic condition.
The etiologies of respiratory failure are diverse and include: (1) PCP, initially
responsible for a high burden of disease and low survival rates; later studies
demonstrated improved survival, even among PCP patients; (2) bacterial pneu-
monia is more frequent in patients with HIV; (3) cytomegalovirus (CMV) is a
minor cause. Also, patients with other associated infections, such as tuberculosis
(TB) and fungal infection; or associated malignancy, such as Kaposis sarcoma
and non-Hodgkins lymphoma; or immune reconstitution syndrome (Fig. 21.2),
may develop respiratory failure during disease evolution [9, 14]. The patients with
ARF due to P. jiroveci and other opportunistic agents can be treated with
21 AIDS Patients in the ICU 355
Table 21.1 General characteristics, intensive care unit admission diagnosis, mechanical venti-
lation, and mortality rates of HIV-infected patients
Fig. 21.1 Incidence of AIDS-defining opportunistic infections and malignancies, the HIV
infected patient study, 19942007 [13]
noninvasive ventilation (NIV). The use of NIV improves gas exchange and avoids
intubation and mechanical ventilation. Patients who failed with the use of NIV or
present severe ARF still require intubation and mechanical ventilation.
356 F. E. Arancibia and M. A. Aguayo
Fig. 21.2 CD4+ cell count ranges for selected HIV-related respiratory illnesses. CMV
Cytomegalovirus; MAC Mycobacterium avium complex [18, 21, 23, 28, 31, 37, 38, 43, 47,
4951, 54, 57]
The global mortality ratios in hospitalized patients infected with HIV requiring
ICU admission went from 23 to 67% [48, 11, 12], most frequently due to
respiratory failure and septic shock. The death of many HIV-infected patients has
been linked directly to late diagnosis and initiation of appropriate antiviral therapy.
Several clinical factors are related to the outcome of AIDS patients in the ICU.
Predictors of increased mortality risk include the need for mechanical ventilation,
and disease severity [high Acute Physiology and Chronic Health Evaluation
(APACHE) II score or simplified acute physiological score (SAPS), modified
multisystem organ failure (MSOF) score, CD4 count, pneumothorax, presence of
cardiovascular instability, and low levels of serum albumin] [14, 15]. Mortality
rates of HIV-infected patients and the need for mechanical ventilation early in the
epidemicprior to 1988reached 82% [16]. However, the use of protective
ventilation (lower tidal volume ventilation) is associated with reduced mortality
rates in HIV-infected patients with acute lung injury and respiratory failure [17].
PCP remains the most prevalent opportunistic infection in patients infected with
the HIV and is often the AIDS-defining illness, occurring most frequently when
the T-helper cell count (CD4+) is \200 cells/ll. First identified as a protozoan
nearly 100 years ago and reclassified as a fungus in 1988, named P. carinii but
renamed P. jiroveci, has a unique tropism for the lung, where it exists primarily as
21 AIDS Patients in the ICU 357
Fig. 21.3 Chest radiograph of a 62-year-old woman who is unaware of her HIV status. Diffuse
bilateral reticular pattern with foci of consolidation in the left lower lobe. Pneumocystis jiroveci
was detected in bronchoalveolar fluid and transbronchial biopsy
an alveolar pathogen without invading the host. In rare cases, pneumocystis dis-
seminates in the setting of severe underlying immunosuppression or overwhelming
infection [14, 18].
At the beginning of the AIDS epidemic, around 75% of patients developed at
least one episode of PCP, with even higher mortality rates of 3585% in patients
requiring admission to an ICU. With the use of prophylaxis, the incidence of PCP
decreased from 47 to 25% [19]. Prophylaxis against P. jiroveci pneumonia can be
safely discontinued in patients with HIV infection who have had a positive
response to HAART (indicated by a CD4 count [200 cells/ll) with minimal risk
of recurrent P. jiroveci pneumonia [20].
Common symptoms of PCP include the subtle onset of progressive dyspnea,
nonproductive cough, and low-grade fever. Physical examination typically reveals
tachypnea, tachycardia, and normal findings on lung auscultation. It is typically
subacute with a clinical course of days or weeks. Acute dyspnea with pleuritic
chest pain may indicate the development of a pneumothorax [14, 18, 21].
Typical radiographic features of PCP are diffuse bilateral interstitial infiltrates
involving the entire lung or the lower lung fields [18, 22] (Fig. 21.3). Less com-
mon findings include localized infiltrates, upper-lobe infiltrates, solitary or mul-
tiple nodules, and pneumatoceles. Pleural effusions and thoracic lymphadenopathy
are rare. Approximately 6% of patients can develop spontaneous pneumothoraces
during the course of their illness. High-resolution computed tomography, which
is more sensitive than chest radiography, may reveal extensive ground-glass
358 F. E. Arancibia and M. A. Aguayo
Fig. 21.4 Transbronchial biopsy specimen stained with Grocotts of an adult woman showing
typical pneumocystis cyst forms of 56 lm (courtesy of Manuel Meneses M.D)
Early in the HIV epidemic, researchers noted that bacterial pneumonia (BP) was a
common cause of morbidity. BP is an important cause of morbidity and mortality
in patients with HIV infection and is at least five times more frequent in HIV-
infected patients compared with healthy individuals. In the precombination ART
era, the HIV Infection Study reported the incidence of BP ranged was 3.97.3
episodes per 100 person-years. Since the introduction of ART, a reduction in the
risk for BP has been observed [21, 31, 32]. BP is still among the most common
causes of respiratory failure resulting in ICU admission [32] and might be the first
manifestation of underlying HIV infection. BP can occur at any stage during HIV
disease and at any CD4+ T-cell count, but it is substantially more frequent among
those with \200 CD4+ T-cell counts. Other risk factors include drug use intra-
venously, previous bacterial infection or PCP, smoking, a low socioeconomic
status, alcohol abuse, comorbidities (including cardiovascular disease, renal dis-
ease, and hepatic cirrhosis), and malnutrition [3133].
The microbiologic cause of community-acquired BP identified most frequently
in HIV-infected patients, are Streptococcus pneumoniae and Haemophilus species
[21]. S. pneumoniae is the most common causative agent and is frequently asso-
ciated with bacteremic disease [31, 33]. The rate of pneumococcal bacteremia is
higher in patients with than without HIV infection [33]. Patients with HIV
infection are at increased risk for infection with penicillin and cotrimoxazole-
resistant S. pneumoniae, and identifying this microorganism could lead to changes
in patient management [21, 23, 33].
H. influenzae, both the encapsulated and nonencapsulated types, is also com-
mon. Pseudomonas aeruginosa pneumonia in some studies has been reported as a
common pulmonary complication [33], especially in patients with low leukocyte
360 F. E. Arancibia and M. A. Aguayo
and CD4+ T-cell counts and ill enough to require ICU admission. Also, there is a
growing number of literature reports about the occurrence of pneumonia due to
Staphylococcus aureus, especially oxacillin-resistant strains, is this population.
Atypical pathogens (Mycoplasma, Chlamydia, and Legionella) seem not to play a
significant role in HIV-infected patients. Rare causes of pneumonia presenting
with cavitation are Rhodococcus equi and Nocardia asteroides [21, 23, 33].
The clinical and radiographic presentation of BP does not differ substantially
for HIV-infected compared with HIV-uninfected patients. Compared with
P. jiroveci pneumonia and other opportunistic infections of lung, the onset of
fever and other symptoms is more abrupt and the patients is more likely to
experience a productive cough and pleuritic chest pain. In contrast, patients with
low CD4 cell count, who are at an increased risk of BP, often present an atypical
clinical picture with milder symptoms and signs, especially when liver cirrhosis
is also present. The white blood cell count is usually elevated in persons with
BP, and a left shift also might be present. Radiographic features typically include
unilateral, focal, segmental, or lobar consolidation. Also, HIV-infected persons
might present with multifocal or multilobar involvement and with parapneu-
monic effusions [3133]. The American Thoracic Society (ATS) severity criteria
developed to assess community-acquired pneumonia (CAP) in patients not
infected with HIV have been found to be valid also for HIV-infected patients
with bacterial CAP [33].
Prompt and accurate diagnosis is essential, because the outcome of HIV-
associated BP appears to be reasonably good with appropriate treatment. Usu-
ally, collection of specimens for microbiologic studies should be performed
before the initiation of antibiotic therapy. An etiologic diagnosis is obtained in
an average of 35% of cases with standard culture methods. In such conditions,
urinary antigen test for S. pneumoniae identification may help in reaching a rapid
and etiologic diagnosis [31]. However, antibiotic therapy should be administered
promptly, without waiting for the results of diagnostic testing. Guidelines for
managing CAP in persons without HIV infection also apply to HIV-infected
persons [34]. Persons with severe pneumonia who require intensive care should
be treated with an IV beta-lactam plus either azithromycin intravenously or an
IV respiratory fluoroquinolone. If risk factors for P. aeruginosa or S. aureus
infection are present, empiric therapy to cover these pathogens should be con-
templated [28].
In all patients presenting with antimicrobial treatment failure, a regular
microbial reinvestigation is mandatory in order to find potentially life-threatening
etiologies. Given the increased incidence of Mycobacterium tuberculosis in HIV-
infected persons, the diagnosis of TB should always be suspected in those with
pneumonia. Also, noninfectious causes with pulmonary dysfunction should be
considered [21, 28, 31].
BP mortality rate is high, and some studies may reach 30%. Factors associated
with increased mortality in HIV patients with BP include the presence of septic
shock, radiologic progression of infiltrates, and CD4 counts \100 cells/ll.
21 AIDS Patients in the ICU 361
CMV is the most frequent viral pneumonia seen in persons with HIV infection.
Although CMV is often detected in BAL fluid, documented CMV pneumonia is
rare and occurs only in severely immunosuppressed patients with CD4 cell counts
\50/ll [38]. Some studies suggest that CMV in BAL fluid reflects bronchopul-
monary replication of the virus. Although the majority of patients with CMV
pneumonia have additional forms of pulmonary pathology, CMV is the only
causative agent frequently identified in patients with severe pulmonary disease.
Due to the high coinfection rate with P. jiroveci, in cases of PCP treatment failure
and severe immunosuppression, the main differential diagnosis must be established
with CMV. Some authors believe that it represents a preterminal phenomenon in
advanced AIDS [21].
Criteria for establishing that CMV is the cause of pneumonitis and pulmonary
dysfunction have been difficult to establish. Clinical features are nonproductive
cough, fever, progressive dyspnea, hypoxemia, and diffuse interstitial infiltrates
[23]. Respiratory symptoms are typically present for 24 weeks. Physical exam-
ination of the chest may be normal or may reveal crackles or evidence of pleural
effusion. The chest radiographic findings of CMV pneumonia vary and include
reticular or ground-glass opacities, alveolar infiltrates, and nodules or nodular
opacities. Pleural effusions may be seen as well. The latter finding may be helpful
in distinguishing CMV pneumonia from P. jiroveci, in which pleural effusions are
rare. Persons suspected of having CMV pneumonia should undergo a careful
dilated retinal examination by an experienced ophthalmologist. Definitive diag-
nosis of CMV pneumonia requires demonstration of cytopathic inclusions and
widespread specific cytopathic changes in the lungs. Confirming the diagnosis is
often not easy due to the typically extremely serious condition, making it difficult
to perform a lung biopsy. Autopsy studies revealed that patients with AIDS and
CMV pneumonia were successfully diagnosed antemortem in only 1324% of
cases. New techniques using in situ DNA hybridization or monoclonal antibodies
to detect the virus may improve the diagnostic yield of less invasive procedures,
such as bronchoalveolar lavage [39].
When suspected CMV pulmonary disease occurs, therapy must be initiated
immediately. Ganciclovir and foscarnet or cidofovir have been used to treat CMV
pneumonia [28], although few data establish that such therapy affects outcome.
Ganciclovir appears to be less effective against pulmonary infections than against
retinitis or gastrointestinal disease, with response rates of 5060%. Despite the
monolithic use of ganciclovir for CMV-related illness, reports of CMV-resistant
strains have been mostly limited to long-term usage in patients with HIV infection.
21 AIDS Patients in the ICU 363
21.6 Mycobacteriosis
21.6.1 Tuberculosis
Fig. 21.5 A 27-year-old man with advanced HIV infection and highly active antiretroviral
treatment. Chest radiographic (a) and high-resolution computed tomography scans (b) showing
peribronchial thickening, nodularity, and septal lines. Two months later (c), progression of lung
involvement to multiple nodular infiltrates, confluent lesions, and acute respiratory failure; video
thoracoscopic biopsy showed Kaposis sarcoma
21.7 Mycoses
21.7.1 Cryptococcosis
such infections is now 2025% of that seen in the mid-1990s [47]. However,
fungal opportunistic infections remain significant causes of morbidity and mor-
tality in persons with HIV in developing countries. After TB and P. jiroveci
pneumonia, cryptococcosis was the third most common opportunistic infection
reported in Thailand [48].
Cryptococcus neoformans is the most common fungal pulmonary infection in
patients with AIDS and usually coexists with cryptococcal meningitis. Also,
cryptococcal pneumonia may be underdiagnosed and not recognized until dis-
semination. The majority of cases are observed in patients who have CD4 counts
\100 cells/ll. When pulmonary infection is present, symptoms and signs include
cough, fever, and dyspnea in association with an abnormal chest radiograph [28].
The radiographic manifestation includes a diffuse reticular or reticulonodular
pattern that resembles PCP, lobar or segmental consolidation, or multiple nodules
that have a propensity to cavitate. Disseminated disease can occur and manifest as
a miliary pattern that may be associated with lymphadenopathy or pleural effusion.
ARF occurring as a complication of cryptococcosis (including pulmonary
infection) was initially thought to be uncommon, with only a handful of case
reports. Visnegarwala et al. [49] documented ARF as occurring in 29 of 210 cases
of AIDS-associated cryptococcosis (13.8%). The clinical presentation was iden-
tical to that of PCP. Independent predictors of ARF were black race, LDH level
C500 IU/L, presence of interstitial infiltrates, and cutaneous lesions. ARF with
cryptococcosis in AIDS patients is associated with disseminated disease and high
mortality rates.
Diagnosis frequently is not defined before death. Serum cryptococcal antigen
testing is a sensitive and rapid screening method in diagnosing cryptococcosis in
HIV-infected patients. Also, routine blood cultures are useful [50]. The recom-
mended initial standard treatment is amphotericin B deoxycholate combined with
flucytosine or fluconazole [28].
21.7.2 Aspergillosis
Fig. 21.6 Open lung biopsy specimen shows proliferation of capillaries in a background of
spindle-shaped tumor cells in the lung interstitium, which are characteristic histological features
of Kaposis sarcoma (hematoxylin and eosin) (courtesy of Manuel Meneses M.D)
biopsies of the bronchus or lung parenchyma have a high risk of hemorrhage and
reveal crush artifacts difficult to distinguish from Kaposis sarcoma. On cytology,
there is no diagnostic feature. Thus, tissue must be obtained on either open lung
biopsy or video-assisted thoracoscopy (Fig. 21.6), or a presumptive diagnosis must
be made when Kaposis sarcoma in seen in the tracheobronchial tree and bron-
choalveolar lavage reveals no other likely pathogens. Often, there is an associated
bloody pleural effusion when thoracentesis is performed [9, 56].
Pulmonary Kaposis sarcoma can respond well to chemotherapy [57]. HAART
and opportunistic infection prophylaxis has contributed to the success rates of
management strategies.
Lymphoma continues to be a cause of pulmonary disease. Although primary
central nervous system (CNS) lymphomas have greatly diminished in frequency
among patients treated with HAART; primary B-cell lymphomas elsewhere con-
tinue to occur. Patchy pulmonary infiltrates have been well described. Biopsy or
cytology is needed to establish a diagnosis. Combination chemotherapy for HIV-
associated lymphoma has become impressively more successful when HAART is
continued with opportunistic infection prophylaxis. Stem cell transplantation has
also been used successfully.
As patients are now living longer, and experience with large patient populations
has increased, other pulmonary neoplastic processes have been recognized that
clinicians should be aware of. Primary effusion cell lymphoma can present in the
pleural, pericardial, or abdominal cavities as effusions. This HHV-8 and EBV-
associated tumor is diagnosed by cytology in many cases. It is not clear how
effective chemotherapy is for this tumor.
21 AIDS Patients in the ICU 369
During the first few months of HAART, immune reconstitution may be compli-
cated by clinical events in which either previously subclinical infections are found
or preexisting partially treated opportunistic infections deteriorate. This condition,
termed immune reconstitution inflammatory syndrome (IRIS), is thought to be
caused by improvement in the hosts immune response to pathogens [58]. The
inflammatory response may be such that the patient develops ARF and requires
ICU. Abdool Karim et al. [45] reported that the incidence of IRIS was 9.5%.
However, the study found the incidence was higher in the integrated-therapy group
(anti-TB and ART) than in the sequential-therapy group: 12.4 versus 3.8%,
respectively. The term IRIS is most commonly used for mycobacterial infections
(TB and disseminated MAC disease) but is also used for other opportunistic
infections, including P. jirovecii pneumonia, toxoplasmosis, hepatitis B and C
viruses, CMV, varicellazoster virus, cryptococcal infection, and histoplasmosis
[28]. The syndrome is manifested as paradoxical worsening of the underlying
respiratory disease and occurs days to months after HAART initiation. However,
IRIS usually develops within the 48 weeks following HAART initiation and is
caused by an exuberant inflammatory response to pneumocystis or mycobacterial
antigens. On the basis of current knowledge, it is tempting to hypothesize that the
immunological basis of IRIS is a HAART-induced rapid clonal expansion and
redistribution of M. tuberculosis-specific memory T cells, which drives a dereg-
ulated immune activation [59] and a cytokine storm [60]. Antigen load could be
responsible for the overvigorous inflammatory response of a recovering immune
system.
Diagnosing IRIS requires excluding other causes of respiratory decompensa-
tion. Clinical presentations are transient worsening or appearance of new symp-
toms and signs, such as fever, increasing chest radiographic infiltrate, peripheral
and mediastinal lymphadenopathy, or changes in radiographic manifestations.
Studies of TB-associated IRIS indicate that this complication is rarely fatal and
that severe episodes can be successfully managed with corticosteroids [45].
Patients with severe cases are able to continue ART.
References
1. Centers for Disease Control and Prevention (1981) Pneumocystis pneumoniaLos Angeles.
MMWR Morb Mortal Wkly Rep 30:250252
2. Centers for Disease Control and Prevention (1981) Kaposis sarcoma and Pneumocystis
pneumonia among homosexual menNew York City and California. MMWR Morb Mortal
Wkly Rep 30:305308
3. Barr-Sinoussi F, Chermann JC, Rey F et al (1983) Isolation of a T-lymphotropic retrovirus
from a patient at risk for acquired immune deficiency syndrome (AIDS). Science 220:
868871
4. Schein RMH, Fischl MA, Pitchenik AE et al (1986) ICU survival of patients with acquired
immunodeficiency syndrome. Crit Care Med 14:10261027
370 F. E. Arancibia and M. A. Aguayo
27. Acute Respiratory Distress Syndrome Network (2000) Ventilation with lower tidal volumes
as compared with traditional tidal volumes for acute lung injury and the acute respiratory
distress syndrome. N Engl J Med 342:13011308
28. Kaplan JE, Benson C, Holmes KH (2009) Guidelines for prevention and treatment of
opportunistic infections in HIV-infected adults and adolescents: recommendations from
CDC, the National Institutes of Health, and the HIV medicine association of the infectious
diseases society of America. MMWR Recomm Rep 58(RR-4):1207
29. Bozzette SA, Sattler FR, Chiu J (1990) A controlled trial of early adjunctive treatment with
corticosteroids for Pneumocystis carinii pneumonia in the acquired immunodeficiency
syndrome. N Engl J Med 323(21):14511457
30. Morris A, Wachter RM, Luce J et al (2003) Improved survival with highly active antiretroviral
therapy in HIV-infected patients with severe Pneumocystis carinii pneumonia. AIDS 17:7380
31. Madeddu G, Fiori ML, Mura MS (2010) Bacterial community-acquired pneumonia in HIV-
infected patients. Curr Opin Pulm Med 16:201207
32. Wolff AJ, ODonnell AE (2001) Pulmonary manifestations of HIV infection in the era of
highly active antiretroviral therapy. Chest 120:18881893
33. Cordero E, Pachn J, Rivero A et al (2000) Community-acquired bacterial pneumonia in
human immunodeficiency virus-infected patients validation of severity criteria. Am J Respir
Crit Care Med 162:20632068
34. Mandell LA, Wunderink RG, Anzueto A et al (2007) Infectious diseases society of
American/American thoracic society consensus guidelines on the management of
community-acquired pneumonia in adults. Clin Infect Dis 44(Suppl 2):S27S72
35. Tumbarello M, Tacconelli E, de Gaetano Donati K et al (2001) Nosocomial bacterial
pneumonia in human immunodeficiency virus infected subjects: incidence, risk factors and
outcome. Eur Respir J 17:636640
36. Petrosillo N, Nicastri E, Viale P (2005) Nosocomial pulmonary infections in HIV-positive
patients. Curr Opin Pulm Med 11:231235
37. Afessa B, Morales I, Weaver B (2001) Bacteremia in hospitalized patients with human
immunodeficiency virus: a prospective, cohort study. BMC Infect Dis 1:13
38. Millar AB, Patou G, Miller RF et al (1990) Cytomegalovirus in the lungs of patients with
AIDS. Respiratory pathogen or passenger. Am Rev Respir Dis 141(6):14741477
39. Drew WL (2007) Laboratory diagnosis of cytomegalovirus infection and disease in
immunocompromised patients. Curr Opin Infect Dis 20:408411
40. Cohen JI (2000) EpsteinBarr virus infection. N Engl J Med 343:481488
41. Writing Committee of the WHO Consultation on Clinical Aspects of Pandemic (H1N1) 2009
Influenza (2010) Clinical aspects of pandemic 2009 influenza A (H1N1) virus infection.
N Engl J Med 362:17081719
42. Griffith DE, Aksamit T, Brown-Elliott BA et al (2007) An official ATS/IDSA statement:
diagnosis, treatment, and prevention of nontuberculous mycobacterial diseases. Am J Respir
Crit Care Med 175:367416
43. Small PM, Fujiwara PI (2001) Management of tuberculosis in the United States. N Engl J
Med 345:189200
44. Harries A, Maher D, Graham S (2004) TB/HIV: a clinical manual, 2nd edn. Publications of
the World Health Organization, Geneva
45. Abdool Karim SS, Naidoo K, Grobler A et al (2010) Timing of initiation of antiretroviral
drugs during tuberculosis therapy. N Engl J Med 362:697706
46. Wallace JM, Hannah JB (1988) Mycobacterium avium complex infection in patients with the
acquired immunodeficiency syndrome. A clinicopathologic study. Chest 93:926932
47. Clark TA, Hajjeh RA (2002) Recent trends in the epidemiology of invasive mycoses. Curr
Opin Infect Dis 15:569574
48. Chariyalertsak S, Supparatpinyo K, Sirisanthana T et al (2002) A controlled trial of
itraconazole as primary prophylaxis for systemic fungal infections in patients with advanced
human immunodeficiency virus infection in Thailand. Clin Infect Dis 34:277284
372 F. E. Arancibia and M. A. Aguayo
49. Visnegarwala F, Graviss EA, Lacke CE (1998) Acute respiratory failure associated with
cryptococcosis in patients with AIDS: analysis of predictive factors. Clin Infect Dis 27:
12311237
50. Feldman CH (2003) Cryptococcal pneumonia. Clin Pulm Med 10:6771
51. Segal BH, Walsh TJ (2006) Current approaches to diagnosis and treatment of invasive
aspergillosis. Am J Respir Crit Care Med 173:707717
52. Holding KJ, Dworkin MS, Wan PC et al (2000) Aspergillosis among people infected with
human immunodeficiency virus: incidence and survival. Adult and adolescent spectrum of
HIV disease project. Clin Infect Dis 31:12531257
53. Zmeili OS, Soubani AO (2007) Pulmonary aspergillosis: a clinical update. Q J Med 100:
317334
54. Moore PS, Chang Y (1995) Detection of herpesvirus-like DNA sequences in Kaposis
sarcoma in patients with and those without HIV infection. N Engl J Med 332:11811185
55. Antman K, Chang Y (2000) Kaposis sarcoma. N Engl J Med 342:10271038
56. Aboulaphia DM (2000) The epidemiologic, pathologic, and clinical features of AIDS-
associated pulmonary Kaposis sarcoma. Chest 117:11281145
57. Martin-Carbonero L, Barrios A, Saballs P et al (2004) Pegylated liposomal doxorubicin plus
highly active antiretroviral therapy versus highly active antiretroviral therapy alone in HIV
patients with Kaposis sarcoma. AIDS 18:17371740
58. Hirsch HH, Kaufmann G, Sendi P et al (2004) Immune reconstitution in HIV infected
patients. Clin Infect Dis 38:11591166
59. Lawn SD, Bekker LG, Miller RF (2005) Immune reconstitution disease associated with
mycobacterial infections in HIV-infected individuals receiving antiretrovirals. Lancet Infect
Dis 5:361373
60. Bourgarit A, Carcelain G, Martinez V et al (2006) Explosion of tuberculin-specific Th1-
responses induces immune restoration syndrome in tuberculosis and HIV co-infected
patients. AIDS 20:F1F7
Therapy of Infection in the ICU
22
J. H. Rommes, N. Taylor and L. Silvestri
22.1 Introduction
J. H. Rommes (&)
Gelre Ziekenhuizen Apeldoorn, Intensive Care, Apeldoorn, The Netherlands
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 373
DOI: 10.1007/978-88-470-1601-9_22, Springer-Verlag Italia 2012
374 J. H. Rommes et al.
Box 22.1 Six basic principles for treating infections in ICU patients
Box 22.2 Initial and empirical therapy of infections in critically ill patients
Previously healthy
Pneumonia Cefotaxime ? erythromycin
Intra-abdominal infection Gentamicin/cefotaxime/metronidazole/amphotericin B
Urosepsis Gentamicin/cefotaxime
Sepsis of unknown origin Gentamicin/cefotaxime
Meningitis Cefotaxime/ampicillin
Cholangitis Gentamicin/ceftriaxone/metronidazole
Underlying disease/transferred from another ward or ICU
Pneumonia Gentamicin/cefotaxime ? erythromycin
Intra-abdominal infection Gentamicin/cefotaxime/metronidazole/amphotericin B
Urosepsis Gentamicin/cefotaxime
Sepsis of unknown origin Gentamicin/cefotaxime
Meningitis Gentamicin/cefotaxime/ampicillin
Cholangitis Gentamicin/ceftriaxone/metronidazole
and superinfections. In endogenous infections, the throat and gut of the criti-
cally ill are the internal sources of potential pathogens, whereas in exogenous
infections, the source of potentially pathogenic microorganisms (PPM) is
external, i.e., outside the patient. All patients expected to require C2 days of
mechanical ventilation, immediately receive SDD (Chapter XX). Oropharyn-
geal, gastrointestinal, and vaginal carriage can be abolished by topical
administration of the nonabsorbable polymyxin E, tobramycin, and amphoter-
icin B (PTA), with the aim of eradicating the internal sources. If a patient has a
tracheostomy, a paste containing PTA is applied around the tracheostoma [9].
The administration of these nonabsorbable topical antimicrobials is continued
until the patient is weaned from the ventilator and extubated. Identifying and
eradicating an external source is often more difficult and requires close coop-
eration between intensivists, nurses, and the infection control team.
4. Deliver topical antimicrobials to achieve high antibiotic concentrations on the
site of infection. Topical application of antimicrobials is safe and contributes to
a more rapid killing of PPMs, resulting in cultures of colonized/infected sites
becoming sterile earlier. For example, to increase the antimicrobial activity in
the lower airway secretions, topical therapy using aerosolized antibiotics should
be considered [10]. Pastes with PTA can be applied topically to tracheostomies,
gastrostomies, and pressure sores. These antimicrobial agents mixed with a
translucent aquaform gel can also be applied in thin layers over fine mesh gauze
to cover, for example, grafted burn wounds [11].
5. Removal or replacement of invasive devices. Endotracheal tubes, intravascular
lines, and urinary catheters are readily contaminated with microorganisms.
376 J. H. Rommes et al.
The majority of patients are admitted to the ICU due to or with an infection.
Immediate and adequate therapy based on the six SDD principles should be given
to prevent mortality. Details of treatment for the most frequently encountered
infections on admission are now described (Box 22.3).
There are two types of lower airway infections: tracheobronchitis and pneumonia.
Tracheobronchitis is an infection of the trachea and/or bronchi with localized
(purulent secretion) and generalized clinical signs (fever, leukocytosis, increased
C-reactive protein (CRP)). Chest X-ray does not show infiltrates. The tracheal
aspirate yields [3+ or [105 CFU/ml of a PPM in the presence of [2+ leukocytes.
The clinical setting in which this type infection most frequently is encountered is
in chronic obstructive pulmonary disease (COPD) patients with acute on chronic
respiratory failure or a patient with neuromuscular weakness who develops
respiratory failure due to retention of secretions and atelectasis followed by
infection. Pneumonia is an infection of the pulmonary tissue. The clinical diag-
nosis is based on the presence of fever, leukocytosis, and increased CRP and a new
or progressive pulmonary infiltrate on chest X-ray. Tracheal aspirate is macro-
scopically purulent and contains [3+ or [105 CFU/ml of a PPM and [2+
leukocytes. Early administration of appropriate antibiotic reduces mortality in
patients with a lower respiratory tract infection [15]. Immediately after obtaining
diagnostic blood samples, systemic antibiotics should be started. In a previously
healthy patient, cefotaxime in combination with a macrolide, e.g., erythromycin,
should provide adequate cover of normal and atypical PPMs. Patients with chronic
underlying disease and those transferred from the ward or another ICU carry both
normal and abnormal PPMs and hence require combination therapy with an
aminoglycoside and cefotaxime. Erythromycin is added in case of CAP. Cultures
will identify the next day normal or abnormal PPMs so that if necessary, anti-
microbial therapy can be adjusted. Infections caused by normal and abnormal
22 Therapy of Infection in the ICU 377
PPMs can be treated with monotherapy, such as cefotaxime for 5 days. If ESBL
producing PPMs, Serratia or Morganella spp are isolated cefotaxim should be
replaced by meropenem [16]. If Pseudomonas spp. are isolated from throat and/or
tracheal aspirate, cefotaxime has to be discontinued and combination therapy with
gentamicin (3 days) and ceftazidime should be prescribed. To increase the anti-
microbial activity in the lower airway secretions, topical therapy using aerosolized
antibiotics should be applied (Box 22.4). Topical application of antimicrobials by
nebulization is safe and contributes to a more rapid killing of PPMs, resulting in
cultures of the tracheal aspirate becoming sterile earlier [10]. The doses of the
different aerosolized antimicrobials are shown in Box 22.4. Tracheal aspirate is
obtained daily until cultures are sterile. To prevent recolonization of the lower
airways, the tracheal tube should be replaced after 3 days. Gentamicin should be
guided by therapeutic drug monitoring (TDM) and generally can be discontinued
after 3 days, to prevent toxicity. Systemic antibiotics and nebulized antimicrobials
are discontinued when cultures are sterile; usually within 5 days.
378 J. H. Rommes et al.
restores digestive tract function. Blind loops are decontaminated with SDD
suspension containing 50 mg polymyxin E, 40 mg of tobramycin, and 500 mg of
amphotericin B administered via catheters placed in the stoma. In case of rectal
overgrowth, i.e., [105 CFU/ml of PPM, SDD enemas or suppositories are
administered twice daily until surveillance cultures are free from PPM.
4. During laparotomy and repeat laparotomy after obtaining cultures, the abdominal
cavity is extensively rinsed with a disinfecting agent, 2% Taurolin [19].
5. All potentially contaminated devices may act as a source of infection and
should be replaced or, if possible, removed.
6. Treatment is evaluated by ongoing surveillance cultures of throat, stomas and
rectum.
Clinical signs of a wound infection are purulent discharge, redness, swelling, tender-
ness, and local warmth. The clinical diagnosis is confirmed by isolating C3+ or C105
microorganisms and C2+ leukocytes in the purulent discharge [20]. Systemic
antimicrobial therapy is seldom indicated, unless symptoms of sepsis, septicemia,
or septic shock occur. Local treatment, drainage, debridement, and removal of
plastic devices are essential and generally sufficient. Following local treatment, the
wounds are rinsed twice daily with a disinfectant, 2% Taurolin, for 3 days.
Aquaform gel mixed with 2% PTA and/or vancomycin can be applied to colo-
nized/infected wounds [11].
Appropriate use of the SDD strategy reduces severe infections of the lower air-
ways and bloodstream by 72 and 37%, respectively [21, 22]. However, although
properly decontaminated, some patients may develop signs of inflammation, and
the diagnostic process in such a patient is a challenge for the intensive care team.
A systematic approach is required (Box 22.5).
If a patient develops symptoms of inflammation such as fever, increased CRP,
and leucocytosis, the first step is a thorough clinical reevaluation: Has the tracheal
aspirate changed in quantity or quality? Is there deterioration of the pulmonary
function? Is the urine cloudy, indicating the presence of leucocytes? How long ago
was the central venous catheter inserted and under what conditions? Are there
signs of an intra-abdominal problem? Is there a new cardiac murmur? In surgical
patients: How are the wounds? Are there signs of an intra-abdominal problem?
The next step is to reevaluate the results of the surveillance cultures. If cultures of
throat, vagina, and gut are free of PPM, an endogenous infection is very unlikely.
Systemic antibiotics should not be prescribed unless vital functions deteriorate
progressively and the intensivist is convinced that the patient is suffering from
sepsis. Diagnostic samples of lower airways, blood, urine, and wound fluid should
be obtained. During decontamination, low-level pathogens such as enterococci and
coagulase-negative staphylococci (CNS) may be found in the tracheal aspirate.
The clinical impact of isolation of these low-level pathogens in the tracheal
aspirate is nil; neither enterococci nor CNS cause lower airway infection.
Colonization by Candida spp. of the lower respiratory tract on admission is
frequently observed and may persist despite decontamination. Fortunately, Candida
pneumonia is extremely rare and occurs only following hematogenous spread.
Changes on X-ray of the thorax require bronchoscopy followed by culture (bacteria,
fungi), stains (Gram/Ziehl-Neelsen), and cytology. Cytology of the bronchoalveolar
lavage (BAL) should include stains aimed at viral-inclusion bodies suggesting
viral infection. Viral pneumonia due to reactivation of herpes or cytomegalovirus
(CMV) may occur in critically ill patients. The value of polymerase chain
reaction (PCR) on BAL fluid aimed at a virus is unknown but probably limited.
In abdominal surgery patients, an abdominal CT scan may provide the diagnosis.
A rather frequent cause of inflammation in a properly decontaminated intensive
care patient is an endogenous bloodstream infection with low-level pathogens,
i.e. CNS catheter-related bloodstream infection, particularly if the catheter
is [7 days in situ [23]. Infections with low-level pathogens are associated with
fever and an increase of CRP but do not cause sepsis syndrome or septic shock, as
these microorganisms lack endotoxin. Diagnosing catheter-related bloodstream
infection is based on one of the following [24]: (1) at least 2 positive blood
cultures before catheter removal and persistently negative cultures after removal;
(2) isolation of the same microorganism from 2 of the following 3 sites: blood
drawn via the suspected line, blood drawn from a peripheral vein, or from the
382 J. H. Rommes et al.
catheter tip; (3) quantitative blood cultures drawn via the central venous line and a
peripheral vein reveal the same microorganism in a ratio [5:1. However the
clinical relevance of this expensive microbiological exercise is limited, as the only
effective treatment of a catheter-related bloodstream infection is removal of the
contaminated line. If the diagnosis of catheter-related infection is correct, clinical
signs of infection, particularly temperature, will normalize within 24 h. The pre-
dominant microorganisms involved in this type of infection are the low-level
pathogens CNS and enterococci. Systemic treatment with antimicrobials is seldom
indicated, even if PPMs such as aerobic Gram-negative bacilli and yeasts are
involved. However an S. aureus catheter-related bloodstream infection carries the
risk of metastatic abscesses and hence requires treatment with a first-generation
cephalosporin for 5 days [25].
More complex is the patient with a prosthetic heart valve who develops a
catheter-related bloodstream infection. If the signs of infection (fever, increased
CRP, leukocytosis) do not resolve within 24 h of removal of the contaminated
catheter, an aggressive approach is indicated. Combination therapy with vanco-
mycin and gentamicin should be commenced. If ultrasonography reveals vegeta-
tions, antibiotic treatment should be continued for 3 weeks. If after 3 weeks
treatment clinical evaluation reveals signs of persistent infection, surgical
replacement of the prosthetic heart valve is indicated [26].
22 Therapy of Infection in the ICU 383
The classic SDD protocol comprising parenteral cefotaxime and enteral PTA is not
designed to control MRSA infections. Control requires the addition of enterally
administered vancomycin [2830]. The policy of surveillance cultures of the throat
and rectum combined with enterally administered vancomycin in the at-risk
patient is analogous to the way in which aerobic Gram-negative bacillary and
fungal carriage is managed by the enterally administered antimicrobials PTA.
It should be remembered that the principal aim of enterally administered
nonabsorbable antimicrobials is the eradication of carriage of potential pathogen
384 J. H. Rommes et al.
22.5 Corticosteroids
22.6 Conclusion
The old, dogmatic microbiology has been replaced by the sound, evidence-based
SDD approach for treating and controlling infection in ICU patients. Recom-
mendations in this chapter, particularly regarding the use of antimicrobial drugs,
differ completely from those generated during the numerous consensus meetings
and require a renewed mindset from the reader. The reader should keep in mind
that evidence showing treatment of infections in critically ill patients using the
SDD approach and a limited number of old antimicrobials has reached a grade 1A
recommendation for reducing morbidity and mortality rates and preventing the
emergence of resistance.
References
1. Zandstra DF, van Saene HKF (2011) Selective decontamination of the digestive tract as
infection prevention in the critically ill. A level 1 evidence-based strategy. Minerva
Anaesthesiologica 77:212219
2. Garnacho-Montero J, Garcia-Garmendia JL, Barrero-Almodovar A et al (2003) Impact of
adequate empirical antibiotic therapy on the outcome of patients admitted to the intensive
care unit with sepsis. Crit Care Med 31:27422751
3. Ibraham EH, Sherman G, Ward S et al (2000) The influence of inadequate antimicrobial
treatment of bloodstream infections on patient outcomes in the ICU setting. Chest 118:146155
4. Martin-Loeches I, Lisboa T, Rodriguez A et al (2010) Combination antibiotic therapy with
macrolides improves survival in intubated patients with community-acquired pneumonia.
Intensive Care Med 36:612620
5. Montravers P, Dupont H, Gauzit R et al (2006) Candida as a risk factor for mortality in
peritonitis. Crit Care Med 34:646652
6. Viviani M, van Saene HKF, Pisa F et al (2010) The role of admission surveillance cultures in
the patients requiring prolonged mechanical ventilation in the intensive care unit. Anaesth
Intensive Care 38:325335
7. Kumar A, Safdar N, Kethireddy S et al (2010) A survival benefit of combination antibiotic
therapy for serious infections associated with sepsis and septic shock is contingent only on
the risk of death: a meta-analytic/meta-regression study. Crit Care Med 38:16511664
8. Chastre J, Wolff M, Fagon JY et al (2003) Comparison of 8 versus 15 days of antibiotic
therapy for ventilator associated pneumonia in adults. A randomised trial. JAMA 290:
25882598
9. Morar P, Makura Z, Jones AS et al (2000) Topical antibiotics on tracheostoma prevents
exogenous colonization and infection of lower airways in children. Chest 117:513518
10. Palmer LB (2009) Aerosolized antibiotics in critically ill ventilated patients. Curr Opinion
Crit Care 115:413418
22 Therapy of Infection in the ICU 387
11. Desai MH, Rutan RL, Heggers JP et al (1992) Candida Infection with and without nystatin
prophylaxis. Arch Surg 127:159162
12. Brown EM (1997) Empirical antimicrobial therapy of mechanically ventilated patients with
nosocomial pneumonia. J Antimicrob Chemother 40:463468
13. Dennesen PJW, van der Ven AJAM, Kessels AGH et al (2001) Resolution of infectious
parameters after antimicrobial therapy in patients with ventilator-associated pneumonia. Am
J Respir Crit Care Med 163:13711375
14. van der Spoel JI, Oudemans-van Straaten HM, Stoutenbeek CP et al (2001) Neostigmine
resolves critical illness-related colonic ileus in intensive care patients with multiple
organ failurea prospective, double-blind, placebo-controlled trial. Intensive Care Med 27:
822827
15. Alvarez-Lerma F, Group ICU-Acquired Pneumonia (1996) Modification of empiric antibiotic
treatment in patients with pneumonia acquired in the intensive care unit. Intensive Care Med
22:387394
16. Abecasis F, Sarginson RE, Kerr S, Taylor N, van Saene HK (2011) Is selective digestive
decontamination useful in controlling aerobic gram-negative bacilli producing extended
spectrum beta-lactamases? Microb Drug Resist 17:1723
17. Marshall JC, Innes M (2003) Intensive care unit management of intra-abdominal infection.
Crit Care Med 31:22282237
18. Lamme B, Boermeester MA, Reitsma JB et al (2002) Meta-analysis of relaparatomy for
secondary peritonitis. Br J Surg 89:15161524
19. Gormans SP, McCafferty DF, Woolfson AD (1987) Reduced adherence of micro-organisms
to human mucosal epithelial cells following treatment with taurolin, a novel antimicrobial
agent. J Appl Bacteriol 62:315320
20. Weber JM, Sheridan RL, Pasternack ME et al (1997) Nosocomial infections in pediatric
patients with burns. Am J Infect Control 25:195201
21. Liberati A, DAmico R, Pifferi S et al (2009) Antibiotic prophylaxis to reduce respiratory
tract infections and mortality in adults receiving intensive care. Cochrane Database Syst Rev
CD 000022
22. Silvestri L, van Saene HKF, Milanese M et al (2007) Selective decontamination of the
digestive tract reduces bacterial bloodstream infections and mortality in critically ill patients.
Systematic review of randomised, controlled trials. J Hosp Infect 65:187203
23. Elliott TSJ, Faroqui MH, Armstrong RF et al (1994) Guidelines for good practice in central
venous catheterisation. J Hosp Infect 28:163176
24. Kurkchubasche AG, Smith MD, Rowe MI (1992) Catheter-sepsis in short bowel syndrome.
Arch Surg 127:2125
25. Naber CK, Baddour LM, Giamarellos-Bourboulis EJ et al (2009) Clinical consensus
conference: survey on Gram-positive bloodstream infections with a focus on Staphylococcus
aureus. Clin Infect Dis 48:S260S270
26. Brush JL (1998) Infective endocarditis in critical care. In: Cunha BA (ed) Infectious diseases
in critical care medicine. Dekker, New York, pp 387434
27. Grayson ML (ed) (2010) Kucers The use of antibiotics, 6th edn. Hodder Arnold, London
28. Silvestri L, Milanese M, Oblach L et al (2002) Enteral vancomycin to control methicillin-
resistant Staphylococcus aureus outbreak in mechanically ventilated patients. Am J Infect
Control 30:391399
29. de la Cal MA, Cerda E, van Saene HKF et al (2004) Effectiveness and safety of enteral
vancomycin to control endemicity of methicillin-resistant Staphylococcus aureus in a
medical/surgical intensive care unit. J Hosp Infect 56:175183
30. Silvestri L, van Saene HKF, Milanese M et al (2004) Prevention of MRSA pneumonia by oral
vancomycin decontamination: a randomised trial. Eur Respir J 23:921926
31. Coello R, Jimenez J, Garcia M et al (1994) Prospective study of infection, colonization and
carriage of methicillin-resistant Staphylococcus aureus in an outbreak affecting 990 patients.
Eur J Clin Microbiol Infect Dis 13:7481
388 J. H. Rommes et al.
32. Steinberg JP, Clark CC, Hackman BO (1996) Nosocomial and community-acquired
staphylococcus bacteremias from 1980 to 1993: impact of intravascular devices and
methicillin-resistance. Clin Infect Dis 23:255259
33. Chang FY, Singh N, Gayowski T et al (1998) Staphylococcus aureus nasal colonisation in
patients with cirrhosis: prospective assessment of association with infection. Infect Control
Hosp Epidemiol 19:328332
34. de Man P, Verhoeven BA, Verbrugh HA et al (2000) An antibiotic policy to prevent
emergence of resistant bacilli. Lancet 355:973978
35. Hoffman SL, Punjabi NH, Kumala S et al (1984) Reduction of mortality in chloramphenicol-
treated severe typhoid fever by high-dose dexamethasone. N Engl J Med 310:8288
36. Sprung CL, Caralis PV, Marcial EH et al (1984) The effects of high-dose corticosteroids in
patients with septic shock. New Engl J Med 311:11371143
Part V
Special Topics
The Gut in the Critically Ill: Central
Organ in Abnormal Microbiological 23
Carriage, Infections, Systemic
Inflammation, Microcirculatory
Failure, and MODS
23.1 Introduction
For many years, the gut has been proposed to play a central role in the patho-
genesis of infections, multiple organ failure, and other diseases frequently
encountered in the critically ill. Also, it is recognized that therapeutic interventions
in the critically ill, such as stress ulcer prophylaxis, systemic antibiotics and,
vasoconstrictors, can cause inadvertent adverse effects. For example, physiological
balances in microbial colonization and microcirculation are impaired, thereby
contributing to increased susceptibility to bacterial overgrowth (i.e., abnormal
carriage), infections, sepsis, intestinal barrier dysfunction and, ultimately, multiple
organ dysfunction syndrome (MODS) [14].
The gut consists of different components: microcirculation, mucosa, immune
system, enteric nervous system, commensal microflora, andduring critical
illnessacquired microorganisms. All these components interact with each other
during critical illness, and all elements may become disturbed as a consequence of
disease and also its treatment. This may result in alterations in crosstalk between
the different elements and yield both beneficial and pathologic responses. The
latter responses may induce MODS. Clark and Coopersmith propose that the
intestinal epithelium, the intestinal immune system, and the intestines endogenous
bacteria all play vital roles in driving MODS [5]. The complex crosstalk between
these three interrelated parts of the gastrointestinal tract cumulatively makes the
gut a motor of critical illness. The importance of the close relation between
D. F. Zandstra (&)
Department of Intensive Care, Onze Lieve Vrouwe Gasthuis,
Amsterdam, The Netherlands
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 391
DOI: 10.1007/978-88-470-1601-9_23, Springer-Verlag Italia 2012
392 D. F. Zandstra et al.
23.2 Microcirculation
Hippocrates described the clinical triad of cold extremities, fever, and thirst, which
implied a bad prognosis. He was probably describing the situation of hypodynamic
septic shock. In the first century A.D, Celsus described the classic signs of
inflammation: rubor, tumor, calor, dolor. Two centuries later, Galen added
the functio laesa of the affected part as the fifth symptom. These signs can be
recognized in systemic inflammatory response syndrome (SIRS), which frequently
occurs in patients after trauma, sepsis, and major surgery. Rubor is the result of
increased blood flow caused by vasodilatation and clinically measured as hypo-
tension. Tumor develops as the consequence of the generalized edema associated
with increased capillary leak. Calor can be recognized systemically as fever
caused by substances such as interleukins and other inflammatory mediators that
alter the set point of the thermoregulatory center. Dolor can be seen as generalized
pain, and functio laesa as the onset of multiple organ dysfunctions.
The description of multiple organ failure is a rather recent event in the history of
medicine. This syndrome was first reported by Tilney et al. in 1973 [6]. They
described the onset of sequential organ failure in patients after surgery for ruptured
abdominal aneurysm and in whom acute renal failure had developed. Sequential
failure of circulation, respiration, liver, and intestines occurred despite intensive
therapy. All patients died. All showed gastric mucosal lesions on postmortem
investigation, and most patients showed renal tubular necrosis. These findings
indicated impaired perfusion of the stomach and kidneys. Postmortem examina-
tions revealed Gram-negative microorganisms in lung slices of all patients. The
relationship between shock, impaired gut perfusion, and the ultimate failure of
organs was evident. However, the role of Gram-negative infections in the patho-
genesis of the syndrome became evident much later [7]. Moreover, the relationship
between sepsis and (gut) mucosal ischemia became apparent. Le Gall et al.
reported in 1976 extensive gastric mucosal erosions seen on endoscopy in patients
with sepsis, whereas patients without sepsis showed normal mucosal surfaces in
the stomach [8].
The acid hypothesis as the major factor in the pathogenesis of gut mucosal
stress ulceration held sway for many years. Impaired microcirculation, as a con-
sequence of sepsis and shock, was subsequently proposed as a major step in the
pathogenesis of gastric mucosal ulceration and bleeding. In a group of high-risk
critically ill patients on prolonged mechanical ventilation, a clinical strategy of
23 The Gut in the Critically Ill 393
such as pneumonia and Clostridium infections, has been reported [2022]. The use
of PPIs should be carefully considered in the critically ill, as inadvertent effects
may cause harm.
present within the lumen and enter the lymph system, activating the GALT
immune response.
A relationship between BT and microcirculation injury has been reported in an
experimental setting. Lymph deviated from entering the systemic circulation
prevented the onset of mesenteric microcirculation injury; in non-lymph-deviated
animals, important and long-lasting injury to the mesenteric microcirculation was
observed [50, 53]. It is hypothesized by these authors that the lymphatic route of
BT up to the mesenteric lymph nodes (MLN) is the relevant route for the induction
of immune response, whereas the portal hematologic route might be the major
route for bacteria dissemination from the intestines into systemic organs [49].
These events also occur under clinical conditions of low cardiac output, such as
chronic heart failure. Sandek et al. reviewed the impact of chronic heart failure on
gut functions [54, 55]. Chronic heart failure (CHF) results in increased sympa-
thetic tone, hormonal derangements, anabolic/catabolic imbalance, endothelial
dysfunction, and systemic low-grade inflammation affecting various organ sys-
tems. Proinflammatory cytokines appear to play important roles in that context.
There is increasing evidence that the gut has a pathophysiological role for both
chronic inflammation and malnutrition in CHF. Indeed, disturbed intestinal
microcirculation and barrier function in CHF seem to trigger cytokine generation,
thereby contributing to further impairment in cardiac function. On the other hand,
myocardial dysfunction can induce microcirculatory injuries, leading to disruption
in the intestinal barrier. This amplifies the inflammatory response. The increased
number of adherent bacteria on the intestinal mucosa seen in patients with CHF
and elevated systemic levels of antilipopolysaccharide IgA emphasizes this fact.
Therefore, the gut is an interesting target for therapeutic interventions in patients
with CHF, in many of whom attempts to eliminate Gram-negative bacteria and
endotoxins from the gut by using nonabsorbable antibiotics (SDD) improved
vascular reactivity and peripheral circulation. Reducing the intestinal endotoxin
pool in the gastrointestinal tract by SDD led to decreased monocyte CD14
expression and intracellular cytokine production in patients with severe CHF.
The improved peripheral endothelial function could be a marker of the anti-
inflammatory effect of SDD [56].
23.7 Probiotics
The clinical value of these observations is still debatable [58]. However, these
observations emphasize the important interaction between intestinal bacterial
content and immune responses.
Probiotic treatment to improve clinical outcome in critically ill patients with
pancreatitis resulted in increased mortality rates [59]. The beneficial role of pro-
biotics in the critically ill to prevent infections is still under debate [60, 61].
Eliminating aerobic Gram-negative bacilli (AGNB) from the intestinal canal using
SDD with nonabsorbable antibiotics has been shown to reduce the severity and
incidence of MODS [7, 62].
23.8 Conclusion
References
1. Deitch EA (1990) Bacterial translocation of the gut flora. J Trauma 30:S184S189
2. MacFie J, OBoyle C, Mitchell CJ et al (1999) Gut origin of sepsis: a prospective study
investigating associations between bacterial translocation, gastric micro flora, and septic
morbidity. Gut 45:223228
3. Berg RD (1992) Bacterial translocation from the gastrointestinal tract. J Med 23:217244
4. Marston A, Bulkley GB, Fiddian Green RC (eds) (1989) Splanchnic ischaemia and multiple
organ failure. Edward Arnold, London
5. Clark JA, Coopersmith CM (2007) Intestinal crosstalk: a new paradigm for understanding the
gut as the motor of critical illness. Shock 28:384393
6. Tilney NL, Bailey GL, Morgan AP (1973) Sequential system failure after rupture of
abdominal aortic aneurysms: an unsolved problem in the postoperative care. Ann Surg
178:117122
7. Stoutenbeek ChP, van Saene HKF, Zandstra DF (1996) Prevention of multiple organ failure
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 approaches, vols 1, 2. Pabst Science, Berlin, pp 10551066
8. Le Gall JR, Mignon FC, Rapin M et al (1976) Acute gastroduodenal lesions related to severe
sepsis. Surg Gynecol Obstet 142:377380
9. Zandstra DF, Stoutenbeek CP (1994) The virtual absence of stress-ulceration related bleeding
in ICU patients receiving prolonged mechanical ventilation without any prophylaxis.
A prospective cohort study. Intensive Care Med 20:335340
10. Zandstra DF, van der Voort PH (2004) A more appropriate critical appraisal of the available
evidence? Crit Care Med 32:21662167
398 D. F. Zandstra et al.
11. Zandstra DF, van der Voort PH (2004) Comment on surviving sepsis campaign guidelines for
the management of severe sepsis and septic shock by dellinger et al. Intensive Care Med 30:1984
12. Van Spreuwel-Verheijen M, Bosman RJ, Oudemans-Van Straaten HM et al (2006) Is the
surviving sepsis campaign for stress ulcer prophylaxis justified? Intensive Care Med
32(Suppl 1):S23
13. Silvestri L, van Saene HK, Milanese M et al (2007) Selective decontamination of the
digestive tract reduces bacterial bloodstream infection and mortality in critically ill patients.
Systematic review of randomized, controlled trials. J Hosp Infect 65:187203
14. de Smet AM, Kluytmans JA, Cooper BS et al (2009) Decontamination of the digestive tract
and oropharynx in ICU patients. N Engl J Med 360:2031
15. Herbert MK, Holzer P (2008) Standardized concept for the treatment of gastrointestinal
dysmotility in critically ill patientscurrent status and future options. Clin Nutr 27:2541
16. van der Spoel JI, Oudemans-van Straaten HM, Kuiper MA et al (2007) Laxation of critically
ill patients with lactulose or polyethylene glycol: a two-centre randomized, double-blind,
placebo-controlled trial. Crit Care Med 35:27262731
17. van der Spoel JI, Oudemans-van Straaten HM, Stoutenbeek CP et al (2001) Neostigmine
resolves critical illness-related colonic ileus in intensive care patients with multiple organ
failurea prospective, double-blind, placebo-controlled trial. Intensive Care Med 27:822827
18. Vesper BJ, Jawdi A, Altman KW et al (2009) The effect of proton pump inhibitors on the
human microbiota. Curr Drug Metab 10:8489
19. Louis P, Flint H (2009) Diversity, metabolism and microbial ecology of butyrate-producing
bacteria from the human large intestine. FEMS Microbiol Lett 294:18
20. Dalton BR, Lye-Maccannell T, Henderson EA et al (2009) Proton pump inhibitors increase
significantly the risk of clostridium difficile infection in a low-endemicity, non-outbreak
hospital setting. Aliment Pharmacol Ther 29(6):626634
21. Sultan N, Nazareno J, Gregor J (2008) Association between proton pump inhibitors and
respiratory infections: a systematic review and meta-analysis of clinical trials. Can J
Gastroenterol 22:761766
22. Vakil N (2009) Acid inhibition and infections outside the gastrointestinal tract. Am J
Gastroenterol 104(Suppl 2):S17S20
23. Spronk PE, Ince C, Gardien MJ et al (2002) Nitroglycerin in septic shock after intravascular
volume resuscitation. Lancet 360(9343):13951396
24. Spronk PE, Zandstra DF, Ince C (2004) Bench to bedside review: Sepsis is a disease of the
microcirculation. Crit Care 8:462468
25. Sakr Y, Dubois MJ, De Backer D et al (2004) Persistent microcirculatory alterations are
associated with organ failure and death in patients with septic shock. Crit Care Med 32:
18251831
26. De Backer D, Creteur J, Preiser JC et al (2002) Microvascular blood flow is altered in patients
with sepsis. Am J Respir Crit Care Med 166:98104
27. Boerma EC, van der Voort PH, Spronk PE et al (2007) Relationship between sublingual and
intestinal microcirculatory perfusion in patients with abdominal sepsis. Crit Care Med
35:10551060
28. Boerma EC, Kuiper MA, Kingma WP et al (2008) Disparity between skin perfusion and
sublingual microcirculatory alterations in severe sepsis and septic shock: a prospective
observational study. Intensive Care Med 34:12941298
29. Lush CW, Kvietys PR (2000) Micro vascular dysfunction in sepsis. Microcirculation 7:83101
30. Dietzman RH, Manax WG, Lillehei RC (1967) Shock: mechanisms and therapy. Can
Anaesth Soc J 14:276286
31. Weil MH, Shubin H, Carlson R (1975) The treatment of circulatory shock. Use of
sympathomimetic and related vasoactive agents. JAMA 231:12801286
32. Dubin A, Oposo M, Casabella CA et al (2009) Increasing arterial blood pressure with
norepinephrine does not improve microcirculatory blood flow: a prospective study. Crit Care
13:r92
23 The Gut in the Critically Ill 399
33. Breslow MJ, Miller CF, Parker SD et al (1987) Effect of vasopressors on organ blood flow
during endotoxin shock in pigs. Am J Physiol 252:H291H300
34. Johannes T, Mik EG, Klingel K et al (2009) Low-dose dexamethasone-supplemented fluid
resuscitation reverses endotoxin-induced acute renal failure and prevents cortical
microvascular hypoxia. Shock 31(5):521528
35. Bersten AD, Hersch M, Cheung H et al (1992) The effects of various sympathomimetics on
the regional circulations in hyperdynamic sepsis. Surgery 112:549561
36. Priebe HJ, Noldge GF, Ambruster K et al (1995) Differential effects of dobutamine,
dopamine and noradrenaline on splanchnic hemodynamics and oxygenation in the pig. Acta
Anesthesiol Scand 39:10881096
37. Marsunaga T, Fujisaki Y, Yamamoto K et al (1985) Norepinephrine in cochlear
microcirculation of guinea pigs. Am J Otolaryngol 6:226230
38. Ikaruga H, Taka T, Nakajima S et al (1999) Norepinephrine, but not epinephrine enhances
platelet reactivity and coagulation after exercise in humans. J Appl Physiol 86:133138
39. Dietrich GV, Heesen M, Boldt J et al (1996) Platelet function and adrenoceptors during and
after induced hypotension using nitroprusside. Anesthesiology 85:13341340
40. Muellner M, Urbanek B, Havel C et al (2004) Vasopressors for shock (Cochrane review).
Cochrane Library, issue 4. Wiley, Chichester
41. Indrambarya T, Boyd JH, Wang Y et al (2009) Low-dose vasopressin infusion results in
increased mortality and cardiac dysfunction following ischemia-reperfusion injury in mice.
Crit Care 13:98
42. Communal C, Singh K, Pimentell DR et al (1998) Norepinephrine stimulates apoptosis in
adult rat ventricular myocytes by activation of the B-adrenergic pathway. Circulation
98:13291334
43. Dincer HE, Gangopadhyay N, Wang R et al (2001) Norepinephrine induces alveolar
epithelial apoptosis mediated by alfa-, beta-, and angiotensin receptor activation. Am J
Physiol Lung Cell Mol Physiol 281:L624L630
44. Minneci PC, Deans KJ, Banks SM et al (2004) Different effects of epinephrine,
norepinephrine, and vasopressin on survival in a canine model of septic shock. Am J
Physiol Heart Circ Physiol 287:H2545H2554
45. Lima A, Jansen TC, van Bommel J et al (2008) The prognostic value of the subjective
assessment of peripheral perfusion in critically ill patients. Crit Care Med 34:12941298
46. Rivers E, Nguyen B, Havstad S et al (2001) Early goal-directed therapy collaborative group.
Early goal-directed therapy in the treatment of severe sepsis and septic shock. N Engl J Med
345:13681377
47. Rivers EP, Coba V, Whitmill M (2008) Early goal-directed therapy in severe sepsis and
septic shock: a contemporary review of the literature. Curr Opin Anaesthesiol 21:128140
48. Cheroutre H, Madakamutil L (2004) Acquired and natural memory T cells join forces at the
mucosal front line. Nat Rev Immunol 4:290300
49. Koh IHJ, Liberatore AMA, Menchaca-Diaz JL et al (2006) Bacterial translocation,
microcirculation injury and sepsis. Endocr Metab Immune Disord Drug Targets 6:143150
50. Koh IH, Menchaca-Diaz JL, Farsky SH (2002) Injuries to the mesenteric microcirculation
due to bacterial translocation. Transplant Proc 34:10031004
51. Rath HC (2003) The role of endogenous bacterial flora: bystander or the necessary
prerequisite? Eur J Gastroenterol Hepatol 15:615620
52. Takebayashi K, Hokari R, Kurihara C et al (2009) Oral tolerance induced by enterobacteria
altered the process of lymphocyte recruitment to intestinal microvessels: roles of endothelial
cell adhesion molecules, TGF-beta and negative regulators of TLR signaling.
Microcirculation 16:251264
53. Ruiz-Silva M, Silva RM (2006) Can bacterial translocation be a beneficial event? Transplant
Proc 38:18361837
54. Sandek A, Anker SD, von Haehling S (2009) The gut and intestinal bacteria in chronic heart
failure. Curr Drug Metab 10:2228
400 D. F. Zandstra et al.
55. Sandek A, Rauchhaus M, Anker SD et al (2008) The emerging role of the gut in chronic heart
failure. Curr Opin Clin Nutr Metab Care 11:632639
56. Conraads VM, Jorens PG, De Clerck LS et al (2004) Selective intestinal decontamination in
advanced chronic heart failure: a pilot trial. Eur J Heart Fail 6:483491
57. Spth G, Hirner A (1998) Microbial translocation and impairment of mucosal immunity
induced by an elemental diet in rats is prevented by selective decontamination of the
digestive tract. Eur J Surg 164:223228
58. Alberda C, Gramlich L, Meddings J et al (2007) Effects of probiotic therapy in critically ill
patients: a randomized, double-blind, placebo-controlled trial. Am J Clin Nutr 85:816823
59. Besselink MG, van Santvoort HC, Buskens E et al (2008) Probiotic prophylaxis in predicted
severe acute pancreatitis: a randomised, double-blind, placebo-controlled trial. Lancet
371:651659
60. van Silvestri L, van Saene HK, Gregori D et al (2010) Probiotics to prevent ventilator-
associated pneumonia: no robust evidence from randomized controlled trials. Crit Care Med
38:16161617
61. Morrow LE (2009) Probiotics in the intensive care unit. Curr Opin Crit Care 15:144148
62. Silvestri L, van Saene HKF, Zandstra DF et al (2010) Impact of selective decontamination of
the digestive tract on multiple organ dysfunction syndrome: systematic review of randomized
controlled trials. Crit Care Med 38:18
Nonantibiotic Measures to Control
Ventilator-Associated Pneumonia 24
A. Gullo, A. Paratore and C. M. Celestre
24.1 Introduction
A. Gullo (&)
Department of Anesthesia and Intensive Care, School of Medicine,
University Hospital Catania, Catania, Italy
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 401
DOI: 10.1007/978-88-470-1601-9_24, Springer-Verlag Italia 2012
402 A. Gullo et al.
Strategy Recommendation
Conventional infection control Handwashing and use of protective gowns and gloves and
measures chlorhexidine oral rinse
Strategies related to the Respiratory airway care; design of endotracheal tubes;
artificial airway continuous subglottic aspiration
Strategies related to mechanical Maintenance of ventilator equipment; heat and moisture
ventilation exchangers; sedation adjustment; noninvasive mechanical
ventilation
Strategies related to the Stress-ulcer prophylaxis; gastric overdistension: nasogastric
gastrointestinal tract tubes, enteral nutrition
Strategies related to patient Semirecumbent position; rotational bed therapy
placement
measures, related to correct care of the artificial airway, and strategies related to
the position of intubated patients, the maintenance of mechanical ventilators and
equipment, and the gastrointestinal tract (Table 24.1). The Canadian Critical Care
trials groups report comprises three classes of evidence: (1) recommended
strategies are based on strong rationale and suggestive evidence, (2) strategies
may be supported by suggestive clinical or epidemiologic studies, and (3) no
recommendations are given for practices for which insufficient evidence or con-
sensus regarding efficacy exists. The report asks three questions related to HAP
prevention of HAP: What is not controversial? What is still controversial? What
should be investigated? [6].
Table 24.2 shows the preventive measures for VAP with insufficient evidence or
consensus regarding efficacy.
Preventive strategies
Infrastructure
Multidisciplinary team Programs developed by team consensus are more
effective. Input by critical care staff and respiratory
therapists is crucial
Target staff education Staff education/awareness programs reduce VAP.
Such programs are adaptable to local needs and are
cost effective
Adequate staffing Critical for maintaining patient safety and
adherence to protocols. Particularly important in
critical care units; current nursing shortages exist
Patient
Do not routinely change the breathing Not controversial
circuit more frequently than every week
Humidification system: heat and moisture Still controversial
exchangers versus heated humidification
Handwashing and protective gowns and Recommended
gloves
Chlorhexidine oral rinse Should be considered
Stress-ulcer prophylaxis Still controversial, should be investigated
Avoid gastric overdistension Recommended
Semirecumbent body position and head of Recommended
bed elevation to 3045
Postural changes by rotating beds Should be considered
Enteral nutrition Should be investigated
Avoid deep sedation Still controversial, should be investigated
Closed-system suction catheter versus open- Still controversial, should be investigated
system catheter
Orotracheal instead of nasotracheal Not controversial
intubation
Cuff-pressure optimization Not controversial
Subglottic secretion drainage Recommended
Noninvasive mechanical ventilation Still controversial, should be investigated
Early tracheostomy Still controversial, should be investigated
organisms (AROs), and hands or gloves of hospital personnel are potential res-
ervoirs for spread [7].
24.2.1.2 Handwashing
Infection control programs, such as hand disinfection; handwashing; and use of
protective gowns and gloves, aprons, and masks, to avoid contact with patient
secretions have repeatedly demonstrated efficacy in reducing infection rates. Hand
washing is widely accepted as the cornerstone of infection control in the ICU.
Literature reports show that handwashing and using protective gowns and gloves
during patient contact do not significantly reduce the rate of acquired nosocomial
infections, especially when handling respiratory secretions or during patient con-
tact when the patient carries an antibiotic-resistant pathogen. However, poor staff
compliance is not the only reason for this failure. Although handwashing alone
reduces transmission, it does not eliminate it, as transmission is dependent on the
bacterial load on health care workers hands [8]. The lack of easily reachable
appropriate physical facilities (sinks, bathrooms) has led many institutions to
alcohol-based gels, and clinical data indicate that rates of all nosocomial infection
may be significantly reduced by their use. A randomized clinical trial of ICUs is
required to support handwashing as the cornerstone of infection control.
24.2.1.3 Modulating Bacterial Colonization
Colonization of the oropharynx with pathogenic organisms is an important risk
factor leading to subsequent HAP/VAP. Host-related factors reported in the
literature that predispose to oropharyngeal colonization include renal dysfunction,
diabetes, coma, shock, advanced age, underlying lung disease, and thoracic or
upper abdominal surgery [9]. Oral care has been recommended in several studies,
and adequate daily oral hygiene using topical antiseptic agents yielded mixed
results.
Topical oral application of antiseptics such as chlorhexidine, an antiseptic
solution for controlling dental plaque, or povidoneiodine to the oral mucosa to
prevent VAP was studied in randomized controlled trials (RCTs) with conflicting
results [10]. Several RCTs examined the influence of chlorhexidine in preventing
nosocomial lower respiratory tract infection. Bacteria accumulated in dental pla-
que have been implicated as VAP pathogens when aspirated to lower airways.
Preventive oral washes with chlorhexidine therefore seem reasonable in selected
high-risk patients given its easy administration and reasonable cost. The prophy-
lactic use of chlorhexidine prevention strategies is still controversial and is sug-
gested in selected risk patients [11, 12]. The majority of meta-analyses concluded
that oral antiseptic rinses seem to be effective in reducing VAP. However, RCTs
and meta-analyses should be interpreted with caution, as it seems that these
antiseptics may be effective for preventing lower respiratory tract infection only in
patients who receive mechanical ventilation no longer than 48 h [13]; their use did
not significantly reduce mortality rates. It seems that chemical decontamination
with chlorhexidine as a solitary intervention may be insufficient to significantly
decrease the risk of pneumonia and that thorough mechanical cleaning is still
24 Nonantibiotic Measures to Control Ventilator-Associated Pneumonia 405
24.2.1.4 Probiotics
Previous reviews showed no benefit of probiotic administration in critically ill
patients, but they did not focus on VAP. Probiotics normally function as colonizers
and contribute to the overall health of their hosts by multiple mechanisms,
including immune and antibacterial effects. Enteral administration of probiotics
may modify the gastrointestinal environment in a manner that preferentially favors
growth of minimally virulent species. No adverse events related to probiotic
administration were identified [14]. There is no clinical evidence to support the use
of probiotics to restore normal human flora in critically ill patients and reduce
HAP rates [15]. Literature reports suggest that probiotics (e.g., Lactobacillus
rhamnosus GG) are safe and recommended in a select, high-risk ICU populations,
but administration is not associated with lower incidence of VAP [16, 17].
24.5 Conclusion
References
1. Restrepo MI, Anzueto A, Arroliga AC et al (2010) Economic burden of ventilator-associated
pneumonia based on total resource utilization. Infect Control Hosp Epidemiol 31:509515
2. Diaz E, Lorente L, Valles J, Rello J (2010) Mechanical ventilation associated pneumonia.
Med Intensiva 34(5):318324
408 A. Gullo et al.
3. Torres A, Ewig S, Lode H, Carlet J, European HAP Working Group (2009) Defining, treating
and preventing hospital acquired pneumonia: European perspective. Intensive Care Med
35:929
4. Omrane R, Eid J, Perreault MM et al (2007) Impact of a protocol for prevention of ventilator-
associated pneumonia. Ann Pharmacother 41:13901396
5. Lorente L, Blot S, Rello J (2010) New issue and controversies in the prevention of ventilator-
associated pneumonia. Am J Respir Crit Care Med 182(7):870876
6. Muscedere J, Dodek P, Keenan S et al, VAP Guidelines Committee and the Canadian Critical
Care Trials Group (2008) Comprehensive evidence-based clinical practice guidelines for
ventilator-associated pneumonia: prevention. J Crit Care 23:126137
7. Rotstein C, Evans G, Born A et al (2008) Clinical practice guidelines for hospital-acquired
pneumonia and ventilator-associated pneumonia in adults. Can J Infect Dis Med Microbiol
19(1):1953
8. Silvestri L, Petros AJ, Sarginson RE et al (2005) Handwashing in the intensive care unit: a
big measure with modest effects. J Hosp Infect 59(3):172179
9. Feider LL, Mitchell P, Bridges E (2010) Oral care practices for orally intubated critically ill
adults. Am J Crit Care 19:175183
10. Panchabhai TS, Dangayach NS, Krishnan A, Karnad DR (2009) Effect of oropharyngeal
cleansing with 0.2% chlorhexidine in critically ill patients: an open label randomized
controlled trial. Chest 135:11501156
11. Chan EY, Ruest A, Meade MO, Cook DJ (2007) Oral decontamination for prevention of
pneumonia in mechanically ventilated adults: systematic review and meta-analysis. BMJ
334(7599):889
12. Tantipong H, Morkchareonpong C, Jaiyindee S, Thamlikitkul V (2009) Randomized
controlled trial and meta-analysis of oral decontamination with 2% chlorhexidine solution for
the prevention of ventilator-associated pneumonia. Infect Control Hosp Epidemiol 30:
101102
13. Kola A, Gastmaier P (2007) Efficacy of oral chlorhexidine in preventing lower respiratory
tract infections. Meta-analysis of randomized controlled trials. J Hosp Infect 66:207216
14. Morrow LE, Kollef MH, Casale TB (2010) Probiotic prophylaxis of ventilator-associated
pneumonia: a blinded, randomized, controlled trial. Am J Respir Crit Care Med 182(8):1058
1064
15. Isakow W, Morrow LE, Kollef MH (2007) Probiotics for preventing and treating nosocomial
infections: review of current evidence and recommendations. Chest 132:286294
16. Silvestri L, van Saene HK, Gregori D et al (2010) Probiotics to prevent ventilator-associated
pneumonia: no robust evidence from randomized controlled trials. Crit Care Med 38(7):
16161617
17. Siempos II, Ntaidou TK, Falagas ME (2010) Impact of the administration of probiotics on the
incidence of ventilator-associated pneumonia: a meta-analysis of randomized controlled
trials. Crit Care Med 38(3):954962
18. Dezfulian C, Shojania K, Collard HR et al (2005) Subglottic secretion drainage for
preventing ventilator associated pneumonia: a meta-analysis. Am J Med 118:1118
19. Silvestri L, Milanese M, Piacente N et al (2008) Impact of subglottic secretion drainage on
ventilator-associated pneumonia and mortality. Systematic review of randomized controlled
trials. In: Proceedings of the 21st anesthesia and ICU symposium Alpe Adria, pp 2629
20. Lorente L, Lecuona M, Jimnez A et al (2007) Influence of an endotracheal tube with
polyurethane cuff and subglottic secretion drainage on pneumonia. Am J Respir Crit Care
Med 176:10791083
21. Overend TJ, Anderson CM, Brooks D et al (2009) Updating the evidence-base for suctioning
adult patients: a systematic review. Can Respir J 16(3):e6e17
22. Van Saene HKF, Zandstra DF, Petros AJ et al (2009) Infections in ICU: an ongoing
challenge. In: Gullo A, Besso J, Lumb PD, Williams GF (eds) Intensive and critical care
medicine. Springer, Milan, pp 261272
24 Nonantibiotic Measures to Control Ventilator-Associated Pneumonia 409
23. Niederman MS (2010) Fighting vampires and ventilator-associated pneumonia: is silver the
magic bullet? Chest 135:10071009
24. Kollef MH, Afessa B, Anzueto A et al, NASCENT Investigation Group (2008) Silver-coated
endotracheal tubes and incidence of ventilator-associated pneumonia: the NASCENT
randomized trial. JAMA 300:805813
25. Silvestri L, van Saene HKF, de la Cal MA, de Gaudio AR (2009) Carriage classification of
pneumonia rather than time improves survival. Chest 136(4):11881189
26. Westwell S (2008) Implementing a ventilator care bundle in an adult intensive care unit. Nurs
Crit Care 13:203207
27. Alexiou VG, Ierodiakonou V, Dimopoulos G, Falagas ME (2009) Impact of patient position
on the incidence of ventilator-associated pneumonia: a meta-analysis of randomized
controlled trials. J Crit Care 24(4):515522
28. Silvestri L, Gregori D, van Saene HK et al (2010) Semirecumbent position to prevent
ventilator-associated pneumonia is not evidence based. J Crit Care 25(1):152153
29. Niel-Weise BS, van den Broek PJ (2009) Semi-recumbent position or not? In: Dutch working
party for infection prevention (WIP). http://www.wip.nl/systrev.asp?nr=12
30. Rosenthal VD, Maki DG, Jamulitrat S et al, for the INICC Members (2010) International
Nosocomial Infection Control Consortium (INICC) report, data summary for 20032008,
issued June 2009. Am J Infect Control 38:95104
31. Efrati S, Deutsch I, Antonelli M et al (2010) Ventilator-associated pneumonia: current status
and future recommendations. J Clin Monit Comput 24(2):161168
32. van Saene HKF, Silvestri L, de la Cal MA, Baines PB (2009) The emperors new clothes; the
fairy tale continues. J Crit Care 24:149152
Impact of Nutritional Route
on Infections: Parenteral Versus 25
Enteral
25.1 Introduction
The significance of nutrition in the intensive care unit (ICU) cannot be overstated
[1]. Malnutrition is a marker of poor outcomes and is correlated with longer
hospital stays, nutrition-related complications during and after hospitalization, and
other adverse outcomes. There is a clear association between malnutrition and
postoperative complications. Nutritional status also worsens during hospitalization
in surgical patients and during critical illness; malnutrition rates were higher at
discharge than at admission. Among seriously ill patients, malnutrition is associ-
ated with increased infectious morbidity and prolonged hospital stay. Critical
illness is typically associated with a catabolic stress state in which patients com-
monly demonstrate a systemic inflammatory response. This response is coupled
with complications of increased infectious morbidity, multiorgan dysfunction,
prolonged hospitalization, and disproportionate mortality rates.
Nosocomial infection in critically ill patients is associated with higher mor-
bidity and mortality rates, prolonged ICU and hospital stay, and consequent higher
health care cost [2]. Providing nutritional support has become a standard of care
for critically ill patients. A post hoc analysis suggested preoperative administration
as the most important period. Preoperative supplementation is as effective as
perioperative supplementation in improving outcome [3]. Early nutritional
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 411
DOI: 10.1007/978-88-470-1601-9_25, Springer-Verlag Italia 2012
412 A. Gullo et al.
support, defined as initiation within the first 2448 h of ICU care, is recommended
by clinical practice guidelines as the first-line nutritional therapy in the ICU.
Nutrition administered enterally (EN) and parenterally (PN) should be initiated
if the caloric goal will be difficult to attain. The most important goal is to con-
tinuously supply the enteric mucosa with useful immunonutrients such as gluta-
mine and fiber to preserve the barrier effect, the mucus layer, and immunological
status of the mucosa, with consequent reduced infection rates [4].
Critically ill patients are at high risk for nosocomial infections, which can lead to
organ dysfunction and death. Thus, the benefits and risks of nutritional therapies in
preventing and managing infectious diseases are highly relevant [5, 6]. A major
methodological problem is related to the term critically ill, as it does not refer to
homogenous populations. The prevalence of malnutrition among critically ill
patients, especially those with a protracted clinical course, has remained largely
unchanged over several years and has implications on hospital length of stay, illness
course, and morbidity rates. The profound and stereotypic metabolic response to
critical illness and failure of carers to provide optimal nutritional support therapy
during a patients ICU stay are the principal factors contributing to malnutrition.
Critically ill patients should not be allowed to remain in a state of unopposed
starvation, because this increases morbidity and mortality, particularly in the
setting of multi-organ-system failure [7]. Immunosuppression occurs as a result of
malnutrition, and protein-energy malnutrition has been cited as the major cause of
immunodeficiency worldwide. Critical illness results in derangements of all
components of the acute immune response, which is organized and executed by
innate immunity influenced by the neuroendocrine system. This response starts
with sensing danger by pattern-recognition receptors on immunocompetent cells
and endothelium [8]. The sensed danger signals, through specific signalling
pathways, activate nuclear transcription factor kappa-B and other transcription
factors and gene regulatory systems, which up-regulate proinflammatory mediator
expression. Plasma cascades are also activated, which together with proinflam-
matory mediators further stimulate inflammatory biomarker production. The acute
inflammatory response underlies the pathophysiological mechanisms involved in
the development of multiorgan dysfunction syndrome (MODS). The inflammatory
mediators directly affect organ function mediating the production of nitric oxide
(NO), leading to mitochondrial anergy and cytopathic hypoxia, a condition of
cellular inability to use oxygen.
Understanding the mechanisms of acute immune responses in critical illness is
necessary for the development of therapeutic strategies, understanding molecular
and biological effects of nutrients in maintaining homeostasis has made expo-
nential advances. Perioperative immune modulation using specialized enteral diets
containing specific immunonutrients may improve postoperative outcomes [9].
Some studies investigated the role of nutrition as a modifier of the immune
25 Impact of Nutritional Route on Infections 413
response in specific clinical settings, especially the use of preoperative oral sup-
plementation with immunonutrients in comparison with standard nutrition in
surgical and critically ill patients [10]. Several specific nutrients have been shown
in laboratory and clinical studies to influence nutritional, immunological, and
inflammatory parameters. Immunonutrients are defined as nutrients that provide
specific benefits to the immune system and include glutamine, arginine, long-chain
n-3 polyunsaturated fatty acids, and nucleotides, either alone or in combination.
Usually provided in combination, these nutrients, when added to a standard enteral
formula, seem to improve outcomes by reducing infection rates.
The influence of malnutrition on immunity is complex. Studies investigated the
effects of immunonutrition on morbidity and mortality rates in critical ill patients,
but results are conflicting in terms of study design, population heterogeneity,
treatment timing, and suboptimal delivery of nutrients. In selected patient groups,
the immunonutrition, can be efficacious to reduce infection and mortality rates,
and hospital length of stay. However whether these immunonutrients are benefi-
cial, or should even be used, in critically ill patients remains controversial [11].
Immunonutrition formulae are indicated in specific subgroups of critically ill
patients (e.g. patients with trauma, mild sepsis, surgical patients); this conclusion
is supported by meta-analyses and recent guidelines.
Many tools are used to assess patients nutritional status. Traditional nutrition
assessment tools (albumin, prealbumin, and anthropometry) are not valid in critical
care. Most nutritional assessment techniques are based on their ability to predict
25 Impact of Nutritional Route on Infections 415
Nutritional support had three main objectives: to preserve lean body mass, to
maintain immune function, and to avert metabolic complications. Nutrition ther-
apy therefore specifically aims at attenuating the metabolic response to stress,
preventing oxidative cellular injury, and favorably modulating the immune
response. Because EN and PN carry both risks and benefits, in the average patient
in the ICU who has no contraindications, the choice of route for nutritional support
may be influenced by several factors.
Enteral tube feeding was first employed in the 1600s and was made popular in
the medical profession by the famous British surgeon John Hunter at the end of the
eighteenth century. The indication for early EN is supported by guidelines
published by the European Society for Clinical Nutrition and Metabolism and
American and Canadian guidelines, which recommend starting administration
within the first 2448 h of admission to the ICU [25]. Short-term access is usually
achieved using nasogastric (NG) or nasojejunal (NJ) tubes; percutaneous endo-
scopic gastrotomy (PEG) or jejunostomy should be considered if feeding is
planned for longer than 1 month. Early EN is recommended for critically ill
patients, with special formulas indicated in specific patient subgroups. Early EN
enhances immunocompetence, reduced clinical infection rates, and maintained gut
structure and function by preserving gut structure/function integrity, balancing
intestinal microflora, maintaining effective local and systemic immunocompe-
tence, and potentially attenuating catabolic stress responses in patients after sur-
gery. There is strong evidence that early enteral feeding prevents infections in a
variety of traumatic and surgical illnesses [26]. There is, however, little support for
similar early feeding in medical illnesses.
Recommendations are to initiate EN as soon as possible whenever the GIT is
functioning. The disadvantage of enteral support is that insufficient energy and
protein coverage can occur. Evidence shows that EN can result in underfeeding
and that nutritional goals are frequently reached only after 1 week. Several
observational studies in long-term ICU patients note that cumulative energy deficit
is related to increased infectious morbidity (infection rate, wound healing,
mechanical ventilation, length of stay, duration of recovery), and costs [27].
Morbidity and mortality rates seem to be linked to such an energy deficit, which
often occurs during the first week of stay. Supplemental PN combined with EN can
be considered to cover energy and protein targets when EN alone fails to achieve
the caloric goal [28]. EN is believed be safer and less expensive than PN. How-
ever, total enteral feeding (TEN) is associated with complications such as diarrhea,
abdominal distention and cramps, and contamination and infection of an enteral
feeding system. In fact EN provides an ideal environment for the development of
bacteria, and the ICU team thus plays a vital role in implementing and maintaining
appropriate standards of care and minimizing risks of bacterial contamination.
Again, if EN is insufficient or fails, PN should be instituted, respecting the often
reduced demand for exogenous substrates in critically ill patients [29].
25 Impact of Nutritional Route on Infections 417
Table 25.1 European Society for Clinical Nutrition and Metabolism (ESPEN) guidelines
modified. Enteral nutrition (EN): intensive care
Recommendations
Indications for EN All patients who are not expected to be on a full oral diet within
3 days should receive EN
Application of early EN The ESPEN committee recommends that hemodynamically stable
critically ill patients who have a functioning gastrointestinal tract
should be fed early (\24 h) using an appropriate amount of feed.
Exogenous energy supply:
During the acute and initial phase of critical illness: in excess of
2025 kcal/kg body weight/day may be associated with a less
favorable outcome
During the anabolic recovery phase, the aim should be to provide
2530 kcal/kg body weight/day
Patient with severe undernutrition should receive EN up 2530 total
Kcal/kg body weight/day. If these target values are not reached,
supplementary parenteral nutrition (PN) should be given. Consider
i.v. administration of metoclopramide or erythromycin in patients
with intolerance to enteral feeding (e.g., with high gastric residuals)
Route Use EN in patients who can be fed via the enteral route
There is no significant difference in the efficacy of jejunal versus
gastric feeding in critically ill patients
Use supplemental PN in patients who cannot be fed sufficiently via the
enteral route
safe to use EN instead of PN. The beneficial effects of EN when compared with PN
are well documented in numerous prospective randomized controlled trials
involving a variety of patient populations in critical illness, including trauma,
burns, head injury, major surgery, and acute pancreatitis. Whereas few studies
show a differential effect on mortality rates, the most consistent outcome effect
from EN is reduced infectious morbidity. When TPN is recommended, the risk of
infectious complications, especially infection related to central venous catheters, is
often thought greater than potential benefits. However, because early provision of
nutritional support improves outcomes in critically ill patients, avoiding or
delaying TPN administration is potentially harmful (Table 25.1).
When selecting the appropriate enteral formulation for the critically ill patient,
the clinician must first decide if the patient is a candidate for a specialty immu-
nomodulating formulation, which are those supplemented with agents such as
arginine, glutamine, nucleic acid, Omega-3 fatty acids, and antioxidants, and
which should be used for the appropriate patient population (major elective sur-
gery, trauma, burns, head and neck cancer, critically ill patients on mechanical
ventilation) [36]. Results strengthen the indication for a special formula in acute
respiratory distress syndrome (ARDS) and acute lung injury [37]. Administration
of probiotic, a combination of antioxidant vitamins, and trace minerals in specific
critically ill patient populations is still being debated (Table 25.2).
25 Impact of Nutritional Route on Infections 419
Table 25.2 European Society for Clinical Nutrition and Metabolism (ESPEN) guidelines
modified. Special enteral nutrition (EN) formulations for the appropriate patient population
EN via tube feeding is the preferred method of feeding the critically ill patient,
particularly on those who develop a severe inflammatory response, (i.e. patients
who have failure of at least one organ during ICU stay) and an important means of
counteracting the catabolic state induced by severe diseases. Evidence from pri-
marily low-quality trials shows that EN reduces infections, septic complications,
and length of hospital stay, resulting in a better outcome compared with total PN,
but not does affect noninfectious complication or hospital mortality rates. That
combined nutritional support provides additional benefit on overall outcomes has
yet to be proven in further studies, including outcomes of physical and cognitive
functioning, quality of life, cost-effectiveness, and cost utility. Meta-analyses of
ICU studies showed that PN is not related to a greater mortality rate and may even
be associated with improved survival [38].
25.6 Conclusion
response to critical illness includes early EN using the enteral route, which is seen as a
proactive therapeutic strategy that may reduce disease severity, diminish compli-
cations, decrease length of ICU stay, and favorably impact patient outcome.
Guidelines and studies confirm that EN versus PN, early EN initiation, enteral and
parenteral glutamine administration, and intensive insulin therapy are all associated
with reduced infectious morbidity in critically ill patients [40, 41]. EN compared
with PN results in an important decrease in the incidence of infectious complications
in the critically ill and may be less costly, and thus should be the first choice for
nutritional support in the critically ill. EN reduces infections, septic complications
and length of hospital stay, resulting in a better outcome compared with total PN, but
it not does affect noninfectious complication or hospital mortality rates.
Guidelines for using TPN while avoiding catheter-related infection may markedly
improve its outcome. Appropriately training personnel to care for central venous
catheters is imperative and is an effective method of reducing devastating compli-
cations of infection [42]. Catheters impregnated with antiseptics and coated with
antibiotics are now available. Conceivably, as our understanding of interventions
such as central venous catheters and TPN continues to improve, these catheters can
be used in situations in which the risk of catheter-related infection is high, such as
TPN administration. Evidence-based critical care nutrition clinical practice guide-
lines recommend EN over PN; time to initiate EN; use of formulas enriched with fish
oils; glutamine supplementation; glycemic control; arginine-enriched formulas;
motility agents; timing of supplemental PN when appropriate; delivery of hypoca-
loric PN when appropriate; and adoption of a feeding protocol [43, 44].
Nutritional assessment of the critically ill patient is crucial, as the deterioration
of nutritional status is a key factor in surgical and critically ill patient outcomes.
Perioperative nutrition with specialized enteral diets improves outcome when
compared with standard formulas. One meta-analysis suggests that antioxidant
supplementation is associated with no improvement in infectious complications
but is associated with increased survival rates. New prospects may be possible in
the fight against surgical infections by adding probiotics to EN in order to improve
the microenvironment of the colon. Many unanswered questions remain, however,
the last but not the least of which is the advocated proactive posture for metabolic
support in the ICU [45, 46].
References
1. Cahill NE, Dhaliwal R, Day AG, Jiang X, Heyland DK (2010) Nutrition therapy in the
critical care setting: what is best achievable practice? An international multicenter
observational study. Crit Care Med 38(2):395401
2. Farber MS, Moses J, Korn M (2005) Reducing costs and patient morbidity in the enterally fed
intensive care unit patient. JPEN J Parenter Enteral Nutr 29:S62S69
3. Martindale RG, Maerz LL (2006) Management of perioperative nutrition support. Curr Opin
Crit Care 12:290294
4. Gianotti L (2006) Nutrition and infections. Surg Infect (Larchmt) 7(Suppl 2):S29S32
25 Impact of Nutritional Route on Infections 421
5. Elia M, Engfer M, Green C, Silk DB (2010) Letter to the editor on the new guidelines for
adult critically ill patients. JPEN J Parenter Enteral Nutr 34(1):105
6. Taylor B, Krenitsky J (2010) Nutrition in the intensive care unit: year in review 20082009.
JPEN J Parenter Enteral Nutr 34(1):2131
7. Saka B, Kaya O, Ozturk GB et al (2010) Malnutrition in the elderly and its relationship with
other geriatric syndromes. Clin Nutr74 29(6):745748
8. Marshall JC, Charbonney E, Gonzalez PD (2008) The immune system in critical illness. Clin
Chest Med 29(4):605616
9. Mizock BA (2010) Immunonutrition and critical illness: an update. Nutrition 26(78):701707
10. Helminem H, Raitanen M, Kellosalo J (2007) Immunonutrition in elective gastrointestinal
surgery patients. Scand J Surg 96:4650
11. Calder PC (2007) Immunonutrition in surgical and critically ill patients. Br J Nutr 98 (Suppl 1):
S1339
12. 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(2):212220
13. Dhaliwal R, Heyland DK (2005) Nutrition and infection in the intensive care unit: what does
the evidence show? Curr Opin Crit Care 11(5):461467
14. Kang W, Kudsk KA (2007) Is there evidence that the gut contributes to mucosal immunity in
humans? JPEN J Parenter Enteral Nutr 31(3):246258
15. Silvestri L, van Saene HK, Milanese M, Gregori D (2005) Impact of selective
decontamination of the digestive tract on fungal carriage and infection: systematic review
of randomized controlled trials. Intensive Care Med 31(7):898910
16. Silvestri L, Zandstra DF, van Saene HK, Petros AJ et al (2008) Antifungal prophylaxis in
critically ill patients. Crit Care 12(3):420
17. Silvestri L, van Saene HK, Milanese M et al (2007) Selective decontamination of the
digestive tract reduces bacterial bloodstream infection and mortality in critically ill patients.
Systematic review of randomized, controlled trials. J Hosp Infect 65(3):187203
18. Silvestri L, van Saene HK, Weir I, Gullo A (2009) Survival benefit of the full selective
digestive decontamination regimen. J Crit Care 24(3):474.e7474.e14
19. Doig GS, Heighes PT, Simpson F, Sweetman EA (2010) Early enteral nutrition reduces
mortality in trauma patients requiring intensive care: a meta-analysis of randomised
controlled trials. Injury 42(1):5056
20. Hanna N, Bialowas C, Fernandez C (2010) Septicemia secondary to ileus in trauma patients:
a human model for bacterial translocation. South Med J 103(5):461463
21. Ziegler TR (2009) Parenteral nutrition in the critically ill patient. N Engl J Med
361(11):10881097; comment: N Engl J Med 362(1):83; author reply 8384
22. Kuzu MA, Terzioglu H, Gen V et al (2006) Preoperative nutritional risk assessment in
predicting postoperative outcome in patients undergoing major surgery. World J Surg
30(3):378390
23. Paillaud E, Herbaud S, Caillet P et al (2005) Relations between undernutrition and
nosocomial infections in elderly patients. Age Ageing 34(6):619625
24. Lohsiriwat V, Lohsiriwat D, Boonnuch W et al (2008) Pre-operative hypoalbuminemia is a
major risk factor for postoperative complications following rectal cancer surgery. World J
Gastroenterol 14(8):12481251
25. Kreymann KG (2008) Early nutrition support in critical care: a European perspective. Curr
Opin Clin Nutr Metab Care 11(2):156159
26. Artinian V, Krayem H, DiGiovine B (2006) Effects of early enteral feeding on the outcome of
critically ill mechanically ventilated medical patients. Chest 129:960967
27. Singer P, Pichard C, Heidegger CP, Wernerman J (2010) Considering energy deficit in the
intensive care unit. Curr Opin Clin Nutr Metab Care 13(2):170176
28. Wernerman J (2008) Paradigm of early parenteral nutrition support in combination with
insufficient enteral nutrition. Curr Opin Clin Nutr Metab Care 11(2):160163
422 A. Gullo et al.
26.1 Introduction
Traditionally, the critically ill patient is considered at risk for the development of
stress-ulcer-related bleeding (SURB) from the intestinal canal. Back-diffusion of
H+ is considered the most important mechanism in the etiology of SURB [1].
In the intensive care unit (ICU), routine administration of specific prophylaxis using
antacids, histamine-2 (H2) receptor antagonists, and cytoprotective agents has been
practiced for the past 40 years. Several meta-analyses have shown a reduction from
15 to 5% in SURB after administration of antacids and H2 receptor antagonists
[25]. Despite this reduced incidence, overt SURB contributes to both morbidity
and mortality [6]. The risk of death is increased only when the bleeding occurs
for [4 weeks after ICU admission, which suggests a different pathophysiology
for early- and late-onset bleeding [7]. Early bleeding is associated with acute
hemodynamic disturbances, such as shock and incomplete resuscitation, whereas
late bleeding is due to sepsis and multiple organ dysfunction syndrome (MODS).
The reported incidence of SURB in adult patients who do not receive prophylaxis
has also fallen, from 60% in 1978 to 0.65% in 1994 [8]. Since 1994, the incidence
of SURB has remained constant (15%), irrespective of whether the patient
D. F. Zandstra (&)
Department of Intensive Care Medicine,
Onze Lieve Vrouwe Gasthuis, Amsterdam,
The Netherlands
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 423
DOI: 10.1007/978-88-470-1601-9_26, Springer-Verlag Italia 2012
424 D. F. Zandstra et al.
received prophylaxis or not [8]. Reduction in mortality rates has never been
demonstrated using a prophylactic regimen [9, 10].
Time-sequence studies in animal experiments have shown that vascular injury due to
various substances is the rate-limiting step in the pathogenesis of mucosal injury.
Vascular injury of the superficial mucosal capillaries may lead to reduced or even
absent blood flow, with subsequent edema and congestion of the mucosal layer.
Whereas the blood supply remains intact, the self-restoring capacity of the mucosal
layer is enormous. When the vascular injury is minimal or absent, the lesions of the
epithelial surface are covered by migrating cuboidal cells within 60 min [11].
26.3.3 Ischemia
Mucosal ischemia plays a key role in SURB pathogenesis [16, 17] and occurs
during shock, sepsis, and endotoxemia. The fundus and corpus of the stomach are
particularly sensitive to ischemia. Mucosal cells do not have the ability to store
26 Gut Mucosal Protection in the Critically Ill Patient 425
26.3.4 Sepsis
The link between SURB and infection becomes increasingly clear in clinical
practice. The incidence of SURB was 20% in patients with ineffectively treated
pneumonias, whereas it was \10% in adequately treated patients [24]. There-
fore, pneumonia can be considered a major factor in SURB pathogenesis.
Ventilator-associated pneumonia in the critically ill patient can effectively be
prevented by selective decontamination of the digestive tract (SDD) [25]. The
potential role of sepsis in the pathogenesis of coagulation disorders that impact
on SURB should be emphasized [10]. Experimental studies that focused on the
role of sepsis in SURB pathogenesis are scarce. However, clinical experience
shows that sepsis is the most important risk factor of SURB [7]. Various
mechanisms are involved: (1) release of vasoactive substances, including
serotonin, histamine, adrenaline, and noradrenaline, promotes vasoconstriction
following endotoxin release; (2) hemodynamic changes may cause hypotension
during the early phase of sepsis and may lead to a redistribution of blood flow
between and within organs [26]. During sepsis, a substantial deficit in nutrient
flow to the mucosa has been observed despite fluid resuscitation [27]. Increased
arteriovenous shunting may cause tissue hypoxemia irrespective of increased
blood flow [28].
Many clinical conditions are associated with increased intestinal permeability and
may lead to endotoxemia, which is related to the degree of permeability [21].
Increased permeability can be prevented by vasodilators and adequate
426 D. F. Zandstra et al.
H. pylori infection is the most important factor in gastric and duodenal ulceration
pathogenesis in the non-ICU patient. In this population, the prevalence of H. pylori
infection is about 30%. Its potential role in SURB pathogenesis in critically ill
patients was investigated in detail in our unit [33, 3537]. The following endpoints
were studied:
1. a new method for detecting H. pylori in mechanically ventilated patients using
a urea breath test;
2. the incidence of H. pylori infection in patients requiring acute admission to the
ICU using the urea breath test and serology;
3. the impact of parenterally and enterally administered antibiotics in SDD on
H. pylori eradication;
4. the incidence and risk factors of mucosal damage by direct endoscopy at ICU
admission;
5. H. pylori infection as an ICU occupational hazard.
Serological tests are unreliable for identifying H. pylori infections ventilated
ICU patients; the laser-assisted ratio analyzer (LARA) 13C-urea breath test
(Alimenterics, NJ, USA) is more reliable [34]. The incidence of H. pylori infec-
tions in acute patients admitted to the ICU is 40% [33]. A relationship was found
between the degree of gastric mucosal lesions on admission and the presence of
active infection [35]. H. pylori infection of the stomach causes extensive micro-
vascular leucocyte adhesion and migration into the tissue parenchyma, and sig-
nificant inflammatory cell infiltration is found [36]. Antimicrobials may be
effective in treating H. pylori infection of the gastric mucosa, e.g., enterally
administered nonabsorbable antibiotics [37]. SDD reduces SURB, in part by
eliminating H. pylori. Transmission of H. pylori from infected patients to nursing
staff was shown in our study [38].
26 Gut Mucosal Protection in the Critically Ill Patient 427
26.3.7 Feeding
Enteral feeding can cause infectious morbidity [39]. Delay in initiating enteral
feeding promotes mucosal vasoconstriction due to reduced prostaglandin synthe-
sis. Stressful conditions combined with food deprivation increases the incidence of
ulceration. Enteral feeding protects the mucosa by: (1) neutralizing acid, (2)
stimulating perfusion, and (3) providing intraluminal substrate as fuel for the
colonic mucosal cell. Three studies show that enteral feeding has a beneficial effect
on the incidence of stress ulceration [4042]. As the critically ill patient is
deprived of adequate swallowing abilities, impaired salivary flow into the stomach
results in extreme low gastric levels of nitric oxide (NO). Nitrate-rich saliva
enhances bactericidal effects of gastric juices [43, 44]. Dietary nitrate increases
gastric mucosal blood flow and mucosal bactericidal defence [45]. Future studies
are needed to demonstrate whether NO donors applied topically or systemically
prevent SURB.
Circulation
Prevent microcirculatory stasis by aggressive correction of hypovolemia and of low cardiac
output
Prevent arteriovenous shunting using vasodilators
Preven corpuscular endothelial adhesion using steroids
Prevent infection
Prevent serious infections, including pneumonia and septicemia, by selective decontamination
of the digestive tract and immunonutrition
Intestinal contents
Remove fecal endotoxins using selective decontamination of the digestive tract
Prevent stasis of intestinal contents using enema and neostigmine
Enteral feeding to ensure mucosal energy supply
Control gastritis due to Helicobacter pylori
Eliminate H. pylori using enterally and parenterally administered antimicrobials in selective
decontamination of the digestive tract protocol
Provide nitric oxide donors enterally or systemically
Consider side effects of proton-pump inhibitors on gut ecology
0.6% in a 7-year cohort of critically ill ICU patients needing mechanical venti-
lation [48 h [35, 48]. These data, in combination with the lack of efficacy of
specific prophylaxis in most recent studies, support the concept that SURB pre-
vention is not only a matter of intragastric-acid control. The proposed guidelines in
the Surviving Sepsis Campaign for preventing stress ulceration therefore can be
challenged [50, 51]. Also, the potential role of NO donors via saliva into the
stomach or systemically administered contributes to improved gastric mucosal
microcirculation and increased intragastric microbial killing of potentially path-
ogenic microorganisms [43]. This challenges the perceived pivotal role of gastric
acid in the pathogenesis of SURB in the critically ill. Enhancement of endogenous
cytoprotective mechanisms by NO donors as a strategy for SURB prevention,
however, needs further evaluation.
Increased susceptibility of potentially the hazardous side effect of increased
transmucosal gastric leak caused by of proton-pump inhibitors (PPIs) and the effects
on the human (protective) microbiota may further contribute to a reconsideration of
PPIs being used to prevent SURB [52, 53]. PPIs impair intestinal colonization
resistance and increase susceptibility for Clostridium difficile infection [53].
26.6 Conclusions
During the past 40 years, clinical strategies controlling SURB have shifted from
interventions to neutralize intragastric acid (antacids) and inhibit acid synthesis
(H2 receptor antagonists, PPIs) toward maneuvers aimed at maintaining and
improving microcirculatory perfusion (aggressive circulatory support, including
vasodilators). The use of anti-inflammatory drugs to prevent microcirculatory
sludging is important to maintain adequate microcirculation. Infections are a
pivotal pathogenetic step in SURB and MODS development [58]. These infec-
tions, however, can be prevented by SDD (25). This therapy also suppresses
H. pylori gastritis, which plays a role in SURB pathogenesis. Implementing SDD
guarantees a consistently low incidence (\1%) of SURB in patients requiring a
minimum of 2 days of mechanical ventilation. Dietary nitrate is considered
important to maintain gastric mucosal blood flow and mucosal defence, including
antimicrobial defence. A better understanding of the side effects of PPIs and H2
antagonists should lead to a more restrictive use of these classes of drugs in the
critically ill, as overuse is acknowledged [59, 60].
References
1. Skillman JJ, Gould SA, Chung W et al (1970) The gastric mucosal barrier: clinical and
experimental studies in critically ill and normal man, and in the rabbit. Ann Surg 172:
564582
2. Cook DJ, Witt LJ, Cook RJ (1991) Stress ulcer prophylaxis in the critically ill: a meta-
analysis. Am J Med 91:519527
3. Shuman RB, Schuster DP, Zuckerman GR (1987) Prophylactic therapy for stress bleeding: a
reappraisal. Ann Intern Med 106:562567
4. Lacroix J, Infante-Rivard C, Jecinek M et al (1989) Prophylaxis of upper gastrointestinal
bleeding in intensive care units. Crit Care Med 17:862869
5. Tryba M (1991) Der Einfluss praeventiver Massnahmen auf Morbiditaet und Mortalitaet von
Intensivpatienten. Anaesthesiol Intensivmed Notfallmed Schmerzther (Suppl)1:4253
6. Cook DJ, Griffith LE, Walter S et al (2001) Canadian Critical Care Trials Group. The
attributable mortality and length of intensive care unit stay of clinically important
gastrointestinal bleeding in critically ill patients. Crit Care 5:368375
7. Zandstra DF, Stoutenbeek CP, Oudemans-van Straaten HM (1989) Pathogenesis of stress
ulcer bleeding. In: van Saene HKF et al (eds) Update in intensive and emergency medicine.
Springer, Berlin, pp 166172
8. Zandstra DF (1995) Stress ulceration in the critically ill. No longer a problem? Proceedings
SMART. Springer, Milan, pp 3032
9. Tryba M (1991) Stress bleeding prophylaxis 1990a meta-analysis. Clin J Gastroenterol
13(Suppl 2):4455
430 D. F. Zandstra et al.
10. Cook DJ, Fuller HD, Guyatt GH et al (1994) Risk factors for gastrointestinal bleeding in
critically ill patients. N Engl J Med 330:377381
11. Lacey ER, Ito S (1984) Rapid epithelial restitution of the rat gastric mucosa after ethanol
injury. Lab Invest 51:573583
12. Messori A, Trippoli S, Vaiani M et al (2000) Bleeding and pneumonia in intensive care
patients given ranitidine and sucralfate for prevention of stress ulcer: metaanalysis of
randomized controlled trials. BMJ 321:11031106
13. OKeefe GE, Gentilello LM, Maier RV (1998) Incidence of infectious complications
associated with the use of histamine2-receptor antagonists in critically ill trauma patients.
Ann Surg 227:120112
14. Mezey E, Palkovits M (1992) Localisation of targets for anti-ulcer drugs in cells of the
immune system. Science 258:16621665
15. Stannard VA, Hutchinson A, Morris DL et al (1988) Gastric exocrine failure in critically ill
patients: incidence and associated features. BMJ 296:155156
16. Fiddian-Green RG, McCough E, Pittenger G et al (1983) Predictive value of intramural pH
and other risk factors for massive bleeding from stress ulceration. Gastroenterology 85:
613620
17. Hottenrott C, Seufert RM, Becker H (1978) The role of ischaemia in the pathogenesis of
stress induced gastric lesions in piglets. Surg Gynecol Obstet 146:217220
18. Menguy R, Desbaillets L, Masters YF (1974) Mechanisms of stress: influence of
hypovolaemic shock on energy metabolism in the gastric mucosa. Gastroenterology 66:4655
19. Christenson JT, Schmuziger M, Maurice J et al (1994) Gastrointestinal complications after
coronary artery bypass grafting. J Thorac Cardiovasc Surg 108:899906
20. Love R, Choe E, Lipton H et al (1995) Positive end-expiratory pressure decreased mesenteric
blood flow despite normalization of cardiac output. J Trauma 39:195199
21. Oudemans-van Straaten HM, Jansen PG, Velthuis H et al (1996) Endotoxaemia and
postoperative hypermetabolism in coronary artery bypass surgery: the role of ketanserin. Br J
Anaesth 77:473479
22. Bhattacharjee M, Bhattacharjee S, Gupta A et al (2002) Critical role of an endogenous gastric
peroxidase in controlling oxidative damage in H. pylori-mediated and nonmediated gastric
ulcer. Free Radic Biol Med 32:731743
23. Biswas K, Bandyopadhyay U, Chattopadhyay I et al (2003) A novel antioxidant and
antiapoptotic role of omeprazole to block gastric ulcer through scavenging of hydroxyl
radical. J Biol Chem 278:10991001
24. Alvarez Lerma F, ICU Acquired Infection Group (1996) Modification of empiric antibiotic
treatment in patients with acquired pneumonia in the ICU. Intensive Care Med 22:387394
25. Liberati A, DAmico R, Pifferi S et al. (2009) Antibiotic prophylaxis to reduce respiratory
tract infections and mortality in adults receiving intensive care. Cochrane Database Syst Rev
4:CD000022
26. Lang CH, Bagby GJ, Ferguson JL et al (1984) Cardiac output and redistribution of organ
blood flow in hypermetabolic sepsis. Am J Physiol 246:331
27. Kreimeier U, Yang ZH, Messmer K (1988) The role of fluid replacement in acute endotoxin
shock. In: Kox W, Bihari D (eds) Shock and the adult respiratory distress syndrome.
Springer, Berlin
28. Bowen JC, LeDoux JC, Harkin GV (1979) Evidence for pathophysiologic arteriovenous
shunting in the pathogenesis of acute gastric mucosal ulceration. Adv Shock Res 1:3542
29. McCafferty DM, Granger DN, Wallace JL (1995) Indomethacin induced gastric injury and
leucocyte adherence in arthritic versus healthy rats. Gastroenterology 109:11731180
30. Cush JJ, Rothlein R, Lindley HB et al (1993) Increased levels of circulating intercellular
adhesion molecules in the sera of patients with rheumatoid arthritis. Arthritis Rheum
36:10981102
31. Low J, Grabow D, Sommers C et al (1995) Cytoprotective effects of CI-959 in the rat gastric
mucosa; modulation of leukocyte adhesion. Gastroenterology 109:12241223
26 Gut Mucosal Protection in the Critically Ill Patient 431
27.1 Introduction
N. J. Reilly (&)
Pharmacy Department, Alder Hey Childrens NHS Foundation Trust,
Liverpool, Merseyside, UK
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 433
DOI: 10.1007/978-88-470-1601-9_27, Springer-Verlag Italia 2012
434 N. J. Reilly et al.
Table 27.1 Potentially pathogenic microorganisms (PPMs) causing infection in intensive care
unit (ICU) patients
Previously healthy host (normal PPM) Host with severe underlying disease (abnormal PPM)
1. Streptococcus pneumoniae 7. Klebsiella spp.
2. Haemophilus influenzae 8. Proteus spp.
3. Moraxella catarrhalis 9. Morganella spp.
4. Escherichia coli 10. Enterobacter spp.
5. Staphylococcus aureus 11. Citrobacter spp.
6. Candida albicans 12. Serratia spp.
13. Acinetobacter spp.
14. Pseudomonas spp.
15. Methicillin-resistant Staphylococcus aureus (MRSA)
SDD aims to convert the abnormal carrier state into normal carriage using nonab-
sorbable antimicrobials. A paste or gel is applied into the inside of the lower cheeks
to prevent or eradicate pre-existing oral carriage of abnormal PPMs, i.e., to decon-
taminate the oropharynx. Suspensions are administered via the nasogastric tube
to decontaminate the stomach and gut and control abnormal carriage of aerobic
Gram-negative bacilli (AGNB), particularly Pseudomonas aeruginosa. Nonab-
sorbable antimicrobials are given throughout ICU treatment to control morbidity and
mortality associated with late secondary endogenous infection, which is caused by
abnormal bacteria not present in admission flora but acquired during ICU treatment.
It generally occurs after 1 week in the ICU and accounts for 30% of infections.
Topical antimicrobials are also used to control exogenous infections, e.g., by
applying paste to a tracheostomy site to control lower airway infections. Grafted
burn wounds may also be targeted by mixing topical antimicrobial agents with a
translucent hydrogel (Aquaform) and applying it in thin layers over fine mesh
gauze to the wound site [26]. Exogenous infection is caused by abnormal potential
pathogens never carried in the patients digestive tract but introduced directly into
the patient. Exogenous infection may occur at any time during ICU treatment and
accounts for 15% of infections.
The ideal SDD regimen should use antimicrobials that are nontoxic, inexpen-
sive, palatable and microbiologically active in the presence of faeces, saliva or
antacids. The most widely used SDD regimen is that of the Groningen group [2],
who devised a protocol using polymyxin E, tobramycin and amphotericin B
27 Selective Decontamination of the Digestive Tract 435
(PTA), which is applied as an oral paste and suspension to treat both the throat and
gut, respectively. In the UK, polymyxin E is known as colistin.
The combination of polymyxin E and tobramycin is synergistic against Proteus
and Pseudomonas spp. It is the most potent antipseudomonal combination asso-
ciated with an effective clearance of Pseudomonas from the gut. Emergence of
resistance to polymyxin is rare. Although there are bacteria-producing tobramycin-
inactivating enzymes, polymyxin is thought to protect tobramycin from being
destroyed by these enzymes [27]. Tobramycin is the preferred aminoglycoside
because it is intrinsically most active against Pseudomonas and is minimally
inactivated by saliva and faeces. It also has useful activity against Staphylococcus
aureus [27]. Both agents absorb endotoxins released by AGNB in the gut. This
feature is important because endotoxin can be absorbed from the gut of seriously
ill patients, producing fever, release of inflammatory mediators and shock. In order
to control yeast overgrowth, amphotericin B is also included in the regime. It is
intrinsically the most potent antifungal, but there is a high rate of inactivation in
the gut requiring the use of high doses [27]. By design, the PTA regimen is
inactive against indigenous flora, such as streptococci viridans, enterococci,
coagulase-negative staphylococci (CNS) and anaerobes [27], each of which is
necessary for normal physiological gut function. Modifications to the PTA regime
may need to be made. For example, SDD was not designed to cover methicillin-
resistant Staphylococcus aureus (MRSA), as MRSA was not a significant problem
in the early 1980s. Therefore, in cases of MRSA endemicity, enterally adminis-
tered vancomycin should be added to the PTA regime [28, 29]. In the case of
endemicity of AGNB producing extended-spectrum beta lactamases (ESBL)
resistant to tobramycin, tobramycin may have to be replaced by paromomycin, and
if Serratia endemicity is present, both polymyxin and tobramycin should be
replaced by paromomycin [29].
27.3 Indications
1. The antimicrobials are given orally or through the nasogastric tube. When the
patient requires gastric suction, the nasogastric tube is clamped and the suction
is discontinued for 1 h.
2. When the normal anatomy of the gastrointestinal tract is disrupted (gastro- or
intestinal-fistulae or colostomy), each (blind) loop should be separately treated
with approximately one half of the oral PTA dose in an adequate volume [30].
Table 27.2 Selective decontamination of the digestive tract (SDD): doses [30]
use medicines that have been appropriately researched and subjected to the
scrutiny of the medicines licensing process. One other problem with extempora-
neous production in the UK is that pharmacy departments are limited in the
quantities of products that they can prepare, unless they have a manufacturers
licence (specials) issued by the medicines and healthcare products regulatory
agency (MHRA).
Because SDD medication is not commercially available or supported by the
usual manufacturers marketing activity, scientific background to the formulations
(assay, rheology) is limited. To date, little work has been undertaken to develop
assays for PTA ingredients when combined in mixtures or formulations for local
and oral application. Assays should indicate activity of the constituent antimi-
crobials, and therefore, a microbiological assay is preferred to techniques such as
high performance liquid chromatography. Suitable microorganisms must be
selected for their resistance to the other PTA components, lack of reversion to
sensitivity must be demonstrated and diffusion from the gel to agar plate must be
matched to that from standard antimicrobial solutions.
438 N. J. Reilly et al.
27.6.1 Oropharynx
27.6.1.1 Paste
Paste has advantages in that it is easy to produce, has good adhesion to the mucosa,
has a prolonged release of medicament, is sugar free, is stable and has a well-
proven formula [33] (Table 27.3). Although paste effectively eliminates AGNB, it
has several drawbacks. It has an unpleasant taste and appearance, can cause
considerable drying of the oral mucosa and can be difficult to remove, occasionally
causing trauma to the mucosa. Because of this, it has poor acceptability with
patients, staff and relatives. We restrict use of the paste to applications around
tracheostomy and gastrostomy sites, where the adherence and barrier properties of
the paste are particularly useful.
27 Selective Decontamination of the Digestive Tract 439
Table 27.3 Formula for selective decontamination of the digestive tract (SDD) paste [33]
Ingredients g
Amphotericin B powder Ph. Eur 2 (adjusted for potency)
Tobramycin sulphate USP 2
Colistin sulphate Ph. Eur 2
Liquid paraffin Ph. Eur 10
Orabase paste (ConvaTec) to 100
Shelf life 1 month; store at room temperature
SDD paste is prepared by mixing each of the antimicrobial powders with 10% w/w liquid paraffin
and gradually incorporating Orabase . Vigorous mixing causes the Orabase to crack
Ph. Eur = European Pharmacopoeia
USP = United States Pharmacopoeia
Table 27.4 Formula selective decontamination of the digestive tract (SDD): pastilles [33]
Ingredients
Gelatine Ph. Eur 500 g
Glycerol Ph. Eur 700 g
Sucrose Ph. Eur 100 g
Sodium benzoate Ph. Eur 4g
Distilled water 600 ml
Lemon oil Ph. Eur 2 ml
Blackcurrant powder 10 g
Amphotericin B powder Ph. Eur 15.88 g (adjusted for potency)
Colistin Sulphate Ph. Eur 12.95 g
Tobramycin sulphate USP 12.95 g
To prepare SDD pastilles, soak the gelatine and water and heat to melt. Add most of the glycerol
and the other ingredients, except the antibiotics, and mix well. Heat for 30 min and then add the
antibiotics, wetting the amphotericin with glycerine
Shelf life 6 months; store at room temperature
A 1.5 g pastille contains 12 mg amphotericin and 10 mg tobramycin and colistin
Ph. Eur = European Pharmacopoeia
USP = United States Pharmacopoeia
27.6.1.2 Pastille
The advantages of the pastille are that it can be flavoured easily, it has good release
characteristics, it is easy to use and it is acceptable to the conscious patient
(Table 27.4). Studies in cancer patients have demonstrated that SDD pastilles are
effective in eradicating the carriage of AGNB and yeasts, reducing the incidence of
radiation mucositis and yeast stomatitis [34]. The pastille, however, has limited,
use as it cannot be used in comatose patients, it has high sugar content and
therefore cannot be used in diabetics and it is unsuitable for young children. It is
440 N. J. Reilly et al.
Table 27.5 Formula for selective decontamination of the digestive tract (SDD): lozenge [36]
Ingredients mg
Antibiotic mixture
Amphotericin B powder Ph. Eur 10
Colistin Sulphate Ph. Eur 2
Tobramycin sulphate USP 1.6
Basic mixture
Citric acid 40
Calcium diphosphate 150
Saccharine 795
Shelf life 3 months; store at room temperature
To prepare SDD lozenges, two powder mixtures are prepared. After sieving of the powders, the
total mixture is mixed in a Turbula mixer (90 rpm) for 15 min. The total powder mixture is then
moistened with 25 ml water, and thereafter, 25 ml sodium carboxymethylcellulose (low vis-
cosity) is added. Further mixing then takes place for 10 min, after which the moistened powder is
dried for a minimum of 4 h at 40C. The dried mixture is then mashed through a 0.75-mm sieve,
and the resulting granulate is sieved further through a 0.4 mm sieve to eliminate the fine powder.
Prior to the final tableting stage, the granulate is mixed with 0.5% magnesium stearate and 2.5%
talc in the Turbula mixer for 2 min
Ph. Eur = European Pharmacopoeia
USP = United States Pharmacopoeia
27.6.1.3 Lozenge
The advantages and disadvantages of the lozenge are very similar to that of the
pastille (Table 27.5). In cancer patients, eradication of AGNB and yeasts by SDD
lozenges has been shown by Spijkervet et al. [35] to take up to 3 weeks, therefore
comparing poorly with eradication rates of 34 days that have been achieved in
ICU patients with paste. One explanation for these differing eradication rates
would be that patients in the ICU are usually unconscious, permitting proper
application of sticky paste, whereas patients with head and neck cancer suck their
lozenges four times daily and eat normal, unsterilised food. Poor compliance
within this group of patients, a lower standard of personal hygiene and an altered
oropharyngeal anatomy may also contribute to the longer eradication times.
Lozenges, when sucked, will take approximately 15 min to dissolve in vivo.
Hence, they do not achieve the same length of contact time with the buccal mucosa
as the paste or gel, and this is therefore another factor contributing to poorer
eradication rates. These results would suggest a need for new formulations to be
developed to allow a more protracted and hence more effective delivery of the
antimicrobials to the buccal mucosa of ambulant patients [36].
27 Selective Decontamination of the Digestive Tract 441
Table 27.6 Formula for selective decontamination of the digestive tract (SDD): 2% gel [33]
Ingredients
Sodium carboxymethylcellulose Ph. Eur 10 g
Glycerol Ph. Eur 60 ml
Nipasept 600 mg
Concentrated peppermint water BP 10 ml
Distilled water 200 ml
Amphotericin B powder Ph. Eur 6 g (adjusted for potency)
Colistin sulphate Ph. Eur 6g
Tobramycin sulphate USP 6g
Shelf life one month; store at 28C
A gel base is prepared from sodium carboxymethylcellulose, propylene glycol or glycerol and
Nipasept solution. Peppermint water is added for flavour; 2% by weight each of amphotericin
B, colistin sulphate and tobramycin sulphate are stirred into the cold gel base, and the resulting
SDD gel is packed into aluminium tubes using a syringe and tube to aid filling. Colistin
sulphomethate sodium has been used in the gel in some centres, where a stringy texture has
been noted when using the sulphate. This method involves using the commercial powder for
injection (Colomycin injection, manufactured by Pharmax)
Ph. Eur = European Pharmacopoeia
USP = United States Pharmacopoeia
BP = British Pharmacopoeia
27.6.1.4 Gel
Gel, as with paste, is sugar free, but it is an improvement on paste in that it is more
palatable, much easier to remove and does not dry the oropharyngeal mucosa [33]
(Table 27.6). The efficacy of this formulation appears to be equal to that of paste,
but patient acceptability with the gel is higher and therefore compliance is better.
The gel presents problems in that it is difficult to produce and at present its long-
term stability is unknown.
Decontamination of the gut is not difficult. Most ICU patients have gut stasis, there
is good contact time between antimicrobials and organisms and it can be dem-
onstrated by surveillance culture that decontamination of the oesophagus, stomach
and small intestine occurs within 3 days. However, to clear PPMs from the large
intestine, there must be functioning gut motility. Due to this controlling factor,
decontamination of the colon and rectum may be longer and may take up to
7 days. A formulation for use in the gastrointestinal tract should ideally release the
antimicrobials in the terminal ileum and provide high concentrations of these
antimicrobials in the colon and rectum. The product should also be easy to use,
acceptable to patients and have good pharmaceutical stability [30]. To date, only
oral suspensions and solutions have been used (Tables 27.6, 27.7, 27.8 and 27.9).
442 N. J. Reilly et al.
Ingredients
Amphotericin B Ph. Eur 500 g (adjusted for potency)
Sodium Citrate Ph. Eur 25 g
Sodium carboxymethylcellulose Ph. Eur 12.5 g
VEEGUM K 25 g
Citric acid monohydrate Ph. Eur 5.95 g to adjust pH to 5.5
Saccharin powder Ph. Eur 470 mg
Lycasin 750 ml
Nipasept 6.5 g
Concentrated peppermint water BP 125 ml
Distilled water to 5330 g
Shelf life 6 months; store at 28C
A suspension is prepared with sodium carboxymethylcellulose as suspending and thickening
agent, distilled water, and Lycasin as the sweetener. VEEGUM K (hydrated magnesium
aluminium silicate) is then added as the anticaking agent. Amphotericin B powder is added
gradually to this mixture, stirring after each addition. Nipasept used as a preservative is
dissolved in the concentrated peppermint water and then added to the suspension. Peppermint
water is added to mask the metallic taste of amphotericin. Finally, saccharin powder is added to
improve palatability and citric acid monohydrate to adjust the pH of the suspension to 5.5. The
remaining water is then added to make up to the final weight
Ph. Eur = European Pharmacopoeia
BP = British Pharmacopoeia
Although Crome [31], in 1988, suggested that research was progressing into the
development of colon-positioned release tablets/capsules for use in conscious
patients, this research does not appear to have lead to the availability or wide-
spread use of these preparations [37]. We used coated colonic colistin capsules
only once [32] in conscious, immunocompromised patients who had functioning
guts. The aim of coated colonic preparations is that they should allow release of
the capsule contents at a pH of approximately 77.2, resulting in disintegration in
the ascending colon. The resultant local delivery of antibacterial agents into the
colon is thought to achieve faecal flora suppression and that, by bypassing the
oesophagus and stomach, gastrointestinal side effects such as nausea and vomiting
should be reduced.
The widespread use of the orally administered solutions and suspensions in
PICU therefore continues. The advantages of these products is that they are stable
[33], easy to produce and can be given via a nasogastric tube and are therefore
suitable to give to an unconscious patient [31]. Problems with poor taste, however,
particularly of colistin, may decrease compliance in conscious patients. Diarrhoea
reportedly may be a problem with SDD treatment [5], but a recent publication [38]
observed that, in fact, patients receiving SDD had more days with normal stools
than days with diarrhoea.
27 Selective Decontamination of the Digestive Tract 443
Table 27.10 Costs of selective decontamination of the digestive tract (SDD) hospital prepara-
tions (June 2010)
Formal costbenefit analyses of SDD in ICU patients have not been performed.
In theory, successful prevention of infection may make ICU more cost effective in
that reduced infection rates secondary to SDD may lead to a shorter patient ICU
stay and lower ICU, parenterally administered antibiotic usage and microbiology
laboratory costs. It has not been firmly established whether these potential savings
offset the additional costs that the SDD regimen incurs through the use of non-
absorbable antimicrobials, systemic antimicrobials and additional microbiological
cultures [43].
The costs of drugs used in the PTA regimen using products prepared by hospital
manufacturing units are listed in Table 27.10, and the costs of commercially
available products are listed in Table 27.11 [44]. The cost of treating a patient
[12 years of age using hospital or commercial products is also shown, although a
complete daily cost comparison cannot be made due to the lack of commercial
products.
It should be noted that the costs of hospital-made products have escalated
dramatically over the past few years, perhaps because of further research and
development costs in the production of more stable products, lack of competition
from commercial products in driving prices down and problems affecting the
446 N. J. Reilly et al.
Table 27.11 Costs of commercially available preparations (British National Formulary March
2010) [44]
Ingredient Supplier
Sodium carboxymethylcellulose Ph. Eur; sodium benzoate Ph. Eur, Fagron UK
tobramycin sulphate powder USP, amphotericin B powder Ph. Eur;
Tel/fax: +44-845-6522525
glycerol Ph. Eur; citric acid monohydrate Ph. Eur; sodium citrate
Ph. Eur; liquid paraffin Ph. Eur; peppermint concentrate BP www.fagron.co.uk
Colistin sulphate powder Ph. Eur. Duchefa Farma
Haarlem, The Netherlands.
Tel: +31-235-319093
Nipasept Clariant UK Ltd
Calverley Lane, Horsforth,
Leeds, UK.
LS18 4RP
Tel: +44-113-2584646
Orange syrup BP J.M. Loveridge Ltd
Southbrook Rd,
Southampton, UK.
SO15 1BH
Tel: +44-170-3228411
Fax:+44-170-3639836
One factor that may make it difficult for a hospital pharmacy to begin manu-
facturing preparations for SDD regimes is sourcing the raw ingredients necessary
to make the products. Table 27.12 therefore provides useful information to
overcome this problem.
27.8 Conclusion
Although application of the SDD concept to intensive care medicine has been
proven to reduce ICU-related morbidity and mortality rates, and despite a publi-
cation validating SDD as an evidence-based medicine manoeuvre [25], the SDD
approach is still not widely used in ICUs. Proposed reasons for this may be:
1. SDD is contrary to the traditional concept that prophylaxis creates resistance.
2. A primacy of opinion over evidence.
3. Opinion leaders control the medical media.
4. SDD formulations are not marketed by the pharmaceutical industry.
5. There is little physicianpharmaceutical industry interaction to stimulate
industry interest in manufacturing SDD products.
In a climate with a lack of commercial products, the necessary extemporaneous
production of SDD formulations must be undertaken by a pharmacy department
that is able to commit to the additional workload that this entails. This means that,
at present, the formulation and supply role of the hospital pharmacist is vital in
order to facilitate application of the SDD concept to clinical practice.
References
1. Silvestri L, de la Cal MA, van Saene HKF (2009) Selective decontamination of the digestive
tract [SDD]. Twenty five years of European experience. In: Gullo A, Besso J, Lumb PD,
Williams GF (eds) Intensive care and critical care medicine. Springer, Milan, pp 273283
2. Stoutenbeek CP, Van Saene HKF, Miranda DR, Zandstra DF (1984) The effect of selective
decontamination of the digestive tract on colonization and infection rate in multiple trauma
patients. Intensive Care Med 10:185192
3. Silvestri L, Mannucci F, van Saene HK (2000) Selective decontamination of the digestive
tract: a life saver. J Hosp Infect 45:185190
4. Abdel-Razek SM, Abdel-Khalek AH, Allam AM et al (2000) Impact of selective
gastrointestinal decontamination on mortality and morbidity in severely burned patients.
Ann Burns Fire Disasters 13:213216
5. Barret JP, Jeschke MG, Herndon DN (2001) Selective decontamination of the digestive tract
in severely burned paediatric patients. Burns 27:439445
6. Bergmans DC, Bonten MJ, Gaillard CA et al (2001) Prevention of ventilator-associated
pneumonia by oral decontamination: a prospective, randomised, double- blind, placebo-
controlled study. Am J Respir Crit Care Med 164:382388
7. Bouter H, Schippers EF, Luelmo SA et al (2002) No effect of preoperative selective gut
decontamination on endotoxemia and cytokine activation during cardiopulmonary bypass: a
randomised, placebo-controlled study. Crit Care Med 30:3843
8. Camus C, Bellissant E, Sebille V et al (2005) Prevention of acquired infections in intubated
patients with the combination of two decontamination regimens. Crit Care Med 33:307314
448 N. J. Reilly et al.
27. Sanchez M, Pizer BP, Alcock SR (2005) Enteral antimicrobials. In: van Saene HKF, Silvestri
L, de la Cal MA (eds) Infection control in the intensive care unit, 2nd edn edn. Springer,
Milan, pp 171187
28. Silvestri L, Milanese M, Oblach L et al (2002) Enteral vancomycin to control methicillin-
resistant Staphylococcus aureus outbreak in mechanically ventilated patients. Am J Infect
Control 30:391399
29. van Saene HKF, Petros AJ, Sarginson RE et al (2009) Is selective decontamination of the
digestive tract a solution to the anti-microbial problem in the UK? JICS 10(2):8689
30. Guidelines for the use of selective digestive decontamination (SDD) (1999, updated 2010).
Alder Hey Childrens NHS Foundation Trust, Liverpool, UK
31. Crome D (1989) Pharmaceutical technology in selective decontamination. In: van Saene
HKF, Stoutenbeek CP, Lawin P, McA Ledingham I (eds) Infection control by selective
decontamination. Springer, Berlin, pp 109112
32. Data on file. Alder Hey Childrens NHS Foundation Trust, Liverpool, UK
33. Data on file. Western Infirmary, Glasgow, UK
34. Symonds RP, Mcillroy P, Khorrami J et al (1996) The reduction of radiation mucositis by
selective decontamination antibiotic pastilles: a placebo-controlled double-blind trial. Br J
Cancer 74:312317
35. Spijkervet FKL, van Saene HKF, van Saene JJM et al (1991) Effect of selective elimination
of the oral flora on mucositis in irradiated head and neck cancer patients. J Surg Oncol
46:167173
36. Data on file. Organon, Oss, The Netherlands
37. van Saene JJM (1990) Colonic delivery of polymyxin E and four quinolones for flora
suppression. PhD thesis, University of Groningen. PAL, Amsterdam
38. van der Spoel JI, Schultz MJ, van der Voort PHJ, de Jonge E (2006) Influence of severity
of illness, medication and selective decontamination on defecation. Intensive Care Med 32:
875880
39. Smit MJ, van der Spoel JI, de Smet AMGA et al (2007) Accumulation of oral antibiotics as
an adverse effect of selective decontamination of the digestive tract: a series of three cases.
Intensive Care Med 33: 20252026
40. Jew RK, Owen D, Kaufman D, Balmer D (1997) Osmolality of commonly used medications
and formulas in the neonatal intensive care unit. Nutr Clin Practice 12:158163
41. van Saene HKF, Stoutenbeek CP, Faber-Nijholt R, van Saene JJM (1992) Selective
decontamination of the digestive tract contributes to the control of disseminated intravascular
coagulation in severe liver impairment. J Pediatr Gastroenterol Nutr 14(4):436442
42. Personal Communication (2010) Stockport pharmaceuticals, Stepping Hill Hospital,
Manchester, UK
43. Bonten MJ, Kullberg BJ, Van Dalen R et al (2000) Selective digestive decontamination in
patients in intensive care. The Dutch Working Group on Antibiotic Policy. J Antimicrob
Chemother 46(3):351362
44. British National Formulary (BNF) No 59 (2010) British Medical Association and the Royal
Pharmaceutical Society of Great Britain, March 2010
Antimicrobial Resistance
28
N. Taylor, I. Cortes Puch, L. Silvestri, D. F. Zandstra
and H. K. F. van Saene
28.1 Introduction
28.2 Definition
N. Taylor (&)
Institute of Ageing and Chronic Disease, University of Liverpool,
Liverpool, Merseyside, UK
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 451
DOI: 10.1007/978-88-470-1601-9_28, Springer-Verlag Italia 2012
452 N. Taylor et al.
SDD is based on the observation that critical illness changes body flora by two
methods: qualitatively and quantitatively. Carriage of normal flora is eroded and
then replaced by abnormal flora [2], and the density of growth shifts from low to
high grade (gut overgrowth) [3]
1. Normal flora consists of six potential pathogens that are commonly found in
the oropharynx and/or gut of healthy people. Streptococcus pneumoniae,
Haemophilus influenzae, Moraxella catarrhalis, which are found in the oro-
pharynx only; Escherichia coli in the gut; and Staphylococcus aureus and
Candida albicans in both the oropharynx and gut.
2. Abnormal flora consists of nine abnormal potential pathogens. Eight aerobic
Gram-negative bacilli (AGNB) and methicillin-resistant S. aureus (MRSA),
found in both the oropharynx and gut. The eight AGNB are Klebsiella,
Enterobacter, Citrobacter, Proteus, Morganella, Serratia, Acinetobacter, and
Pseudomonas spp.
Critical illness changes the normal carrier state into the abnormal carrier state,
which is the persistent presence of abnormal flora in the patients oropharynx and/
or gut [2, 4].
Infection, when classified according to the carrier state, can be categorised into
three different groups [7]: primary endogenous, secondary endogenous and
exogenous. All require a different prophylactic manoeuvre (Table 28.1).
1. Primary endogenous infection is the most frequent type, causing approximately
55% of infections in the intensive care unit (ICU). It occurs when the causative
microorganism is present in the patients admission flora in overgrowth concen-
trations, generally within 1 week of admission. The component of SDD that
prevents primary endogenous infections is the short course of parenterally
administered antibiotics given when the patient is admitted and for the next 4 days.
2. Secondary endogenous infection occurs when the causative microorganism is
not present in the patients admission flora but the abnormal flora is acquired
28 Antimicrobial Resistance 453
Table 28.1 Carriage classification of severe infections of lower airways and bloodstream
during treatment in the ICU, and the patient subsequently develops the carrier
state in overgrowth concentrations, which precedes the infection. Secondary
endogenous infection occurs after a week of ICU treatment at a frequency of
30%. The manoeuvre to impact this type of infection is enterally administered
nonabsorbable antimicrobials and high standards of hygiene.
3. Exogenous infection may occur at anytime during ICU treatment and develops
when the abnormal flora is introduced directly into the patient, bypassing the
carrier state. Approximately 15% of all ICU infections are exogenous. The
manoeuvre designed to combat this infection is hygiene and topical application
of antimicrobials. Enteral administration of antimicrobials also contributes to
exogenous infection control by reducing the reservoir of potential pathogens in
the unit that may be introduced into a patient
Surveillance cultures, the fourth component of SDD, are essential for three
reasons:
1. They are required to distinguish primary endogenous from secondary endog-
enous and exogenous infections.
2. They allow ongoing monitoring of eradication of gut overgrowth during SDD.
3. They are the most sensitive sampling methods for detecting antimicrobial
resistance.
The reason for SDD efficacy requires the understanding of two points
1. Acknowledgement of carriage classification [8]: In order to accept the concept
of SDD, it must be understood that carriage occurs. Primary carriage is when
the patient is admitted to the ICU with flora in the digestive tract; secondary
454 N. Taylor et al.
Table 28.2 Four components of selective decontamination of the digestive tract (SDD)
carriage when the patient acquires flora in the digestive tract during treatment in
the ICU.
2. The antimicrobials selected for SDD were selected for specific reasons and
invariably clear high-grade carriage (Table 28.2):
parenteral cefotaxime clears carriage of normal bacteria; it is active against
normal and against some abnormal pathogens and, moreover, has a good
safety profile;
enterally administered polyenes clear normal fungal carriage;
enterally administered polymyxin/tobramycin with or without vancomycin
clears carriage of abnormal bacteria AGNB and MRSA; these antimicrobials
are nonabsorbable and manage to achieve high intraluminal concentrations.
SDD antimicrobials are required in high concentrations against prevailing
microorganisms (Table 28.3). These concentrations are deemed more important
than sparing the colonisation resistance [9]. Colonisation resistance is the mech-
anism whereby indigenous flora are a barrier against the abnormal flora acquired,
e.g. from food, and then carried in the digestive tract.
Gut overgrowth [10] harms the critically ill in four main ways:
1. Infection. There is a quantitative relationship between surveillance and diag-
nostic samples. As soon as there is overgrowth in surveillance samples, the
28 Antimicrobial Resistance 455
Table 28.3 Effective concentrations against prevailing microorganisms are more important than
sparing the colonisation-resistant flora
28.6 Efficacy
SDD is one of the most investigated clinical interventions in critically ill patients.
Its efficacy in preventing infection-related morbidity and mortality has been
assessed in 11 meta-analyses [1828] covering 65 randomised controlled trials
(RCTs) [2993] (Table 28.4). Of the 11 meta-analyses, lower airway infection was
the endpoint in six [1820, 24, 26, 28]. All meta-analyses invariably demonstrated
a significant reduction in lower airway infections (odds ratio (OR) 0.28, 95%
confidence interval (CI) 0.200.38). Bloodstream infection was the endpoint in
three meta-analyses [2224] and was significantly reduced (OR 0.63, 95% CI
0.460.87). When assessing bloodstream infection, AGNB septicaemias were
significantly reduced, Gram-positive septicaemias were increased but not sig-
nificantly and fungaemia was reduced but not significantly due to the low incidence
in the control group (Table 28.4). Multiple organ dysfunction syndrome (MODS)
456
Table 28.4 Efficacy of selective decontamination of the digestive tract (SDD): 65 randomised controlled trials (RCTs) and 11 meta-analyses
Author No. RCTs Sample size Lower airway Bloodstream Multiple organ Mortality rate
infection infection dysfunction syndrome OR (95% CI)
OR (95% CI) OR (95% CI) OR (95% CI)
Vandenbroucke-Grauls [18] 6 491 0.12, 0.080.19 NR 0.92, 0.451.84
DAmico [19] 33 5727 0.35, 0.290.41 NR 0.80, 0.690.93
Safdar [20] 4 259 NR NR 0.82, 0.222.45
Liberati [21] 36 6922 0.35, 0.290.41 NR 0.78, 0.680.89
Silvestri [22] yeasts 42 6075 NR 0.89, 0.164.95 NR
Silvestri [23] 51 8065 NR 0.63, 0.460.87 0.74, 0.610.91
Silvestri [24] 54 9473
Gram 0.07, 0.040.13 0.36, 0.220.60 NR
Gram+ 0.52, 0.340.78 1.03, 0.751.41 NR
Silvestri [25] 21 4902 NR NR 0.71, 0.610.82
Liberati [26] 36 6914 0.28, 0.200.38 NR 0.75, 0.650.87
Silvestri [27] 7 1270 NR NR 0.50, 0.340.74 0.82, 0.511.32
Silvestri [28] 12 2252 0.54, 0.420.69 NR NR
OR odds ratio, CI confidence interval, NR not reported
N. Taylor et al.
28 Antimicrobial Resistance 457
was the endpoint in one meta-analysis [27]; the relative reduction of 50% was
significant. Mortality was the endpoint in nine meta-analyses [1821, 23, 2528].
SDD consistently reduced mortality rates as long as the sample size was large
enough; the sample size was too small in three meta-analyses [18, 20, 27].
28.7 Safety
SDD safety relies on the long-term level of resistance not emerging against the
SDD antimicrobials [94]. The concept of exposing vast numbers of critically ill
patients to broad-spectrum multiple drug cocktails runs counter to existing theo-
retical models (and dogma), as it is related to the genesis and promotion of
antimicrobial resistance in pathogens acquired in the ICU [95]. The dynamics of
resistance are driven by three mechanisms:
1. Import. The patient comes into the ICU with resistant microorganisms in
overgrowth concentrations in the gut [3].
2. Transmission. 33% of patients admitted as normal carriers to a mixed ICU
developed abnormal carriage of multi-drug-resistant K. pneumoniae and/or
A. baumannii, the two abnormal AGNB endemic in the ICU at the time of the
1-year prospective observational study [96]. A higher Severity of Illness score
on admission was a significant risk factor: the Simplified Acute Physiologically
Score (SAPS) was 13 4.6 in carriers versus 11.3 5 in noncarriers
(p = 0.0006).
3. De novo development. Gut overgrowth defined as C105 potential pathogens per
gram of faeces has been identified as a risk factor for de novo resistance
development [14, 97]. The gut of the critically ill patient with microbial
overgrowth is the ideal site for de novo development of new clones following
increased spontaneous mutation, termed hypermutation. In hypermutation,
microbial populations start mutating vigorously at random, presumably as an
adaptive mechanism that may cause a mutant to arise that would enable them to
overcome the unfavourable surroundings, resulting in polyclonality. A high
proportion of patients who require long-term ICU treatment receive parenter-
ally administered antimicrobials, which are invariably excreted via the bile into
the gut. Although low and fluctuating, the antibiotic levels will kill the sensitive
clones, promoting the emergence of clones resistant to the antibiotics.
Each mechanism is responsible for a third of antimicrobial resistance in the
ICU. The common denominator of all three mechanisms is gut overgrowth.
There are four categories in which antimicrobial resistance is a problem in the
ICU:
1. AGNB
(a) Sensitive to decontaminating agents polymyxin/tobramycin. de Jonge et al.
conducted a prospective open-label RCT in which 934 critically ill adult
patients were randomly assigned on admission to either a medical/surgical
ICU using routine SDD or a similar ICU in the same hospital that did not
458 N. Taylor et al.
use SDD [43]. Study participants were patients with expected duration of
mechanical ventilation of at least 48 h and/or ICU stay [3 days. Surveil-
lance cultures from the throat and rectum were obtained at ICU admission
and at discharge, weekly during ICU treatment and for the first week
postdischarge. The in-hospital mortality rate was significantly lower for
SDD recipients than for control patients (24 vs. 31%; p = 0.02). Carriage
of AGNB resistant to polymyxin E, tobramycin, ceftazidime, ciprofloxacin
and imipenem was significantly reduced in SDD patients compared with
controls (16 vs. 26%; p = 0.001). Similar results were observed by de Smet
et al. in their cluster RCT [45]. Monthly point-prevalence surveys for
carriage of multi-drug-resistant AGNB were obtained. The proportion of
rectal swabs with resistant AGNB was lower for SDD compared with
standard-care patients.
(b) Resistant to decontaminating agents polymyxin/tobramycin:
there is only one potential pathogen intrinsically resistant to polymyxin/
tobramycinSerratia species. In case of Serratia endemicity, polymyxin/
tobramycin must be replaced by paromomycin [98, 99].
extended-spectrum beta-lactamase (ESBL) producing AGNB are often resistant
to tobramycin but always sensitive to polymyxin [100]. In case of ESBL pro-
ducing AGNB endemicity, tobramycin needs to be replaced by another amino-
glycoside, e.g. neomycin, paromomycin [39, 93, 99].
2. MRSA
(a) Sensitive to glycopeptides. Practically all MRSA strains are sensitive to
glycopeptides such as vancomycin and teicoplanin. All RCTs [34, 52, 59,
60, 69, 80] and prospective studies [6, 101, 102] assessing the efficacy of
enterally administered vancomycin on the abnormal MRSA carrier state
demonstrated its efficacy. Therefore, it is recommended to add vancomycin
to PTA in cases of MRSA endemicity.
(b) Resistant to glycopeptides. MRSA is now endemic in many hospitals
throughout the world. Several clusters of glycopeptide-resistant MRSA
have been reported. A 39-case outbreak, defined as involving any patient
carrying or infected by a strain of MRSA with reduced or intermediate
susceptibility to glycopeptides, was recorded in a hospital in Paris, France,
from October 1998 to March 1999 [103]. Another outbreak from Lyon,
France, included 15 patients [104]. MRSA has been acknowledged as a gut
rather than a nasal bacterium [6, 101, 102]. Patients requiring long-term
ICU treatment invariably develop gut overgrowth of MRSA when it is
endemic. MRSA gut overgrowth precedes endogenous MRSA infection,
whichin generalis treated with glycopeptides. The combination of low
antimicrobial concentrations in the faeces following biliary excretion and
overgrowth leads to increased spontaneous mutation, polyclonality and
antimicrobial resistance against glycopeptides [14].
3. Azole-resistant Candida spp.
Practically all critically ill patients who require long-term ICU treatment have
fungal overgrowth [105]. Parenteral azoles administered either prophylactically
28 Antimicrobial Resistance 459
or therapeutically fail to clear fungal overgrowth, because the faecal levels are
not fungicidal and/or fluctuating following biliary excretion [106]. In contrast,
fungal overgrowth promotes increased spontaneous mutation, polyclonality and
antifungal resistance [107, 108].
4. Vancomycin-resistant enterococci (VRE)
Enterococci are normal residents of the large bowel but not the oropharynx.
Enterococci are organisms of low-level pathogenicity present in the gut of
healthy people at concentrations between 1036 enterococci/gram of faeces.
Enterococcus faecalis and E. faecium are the two most common enterococci
associated with infection: E. faecalis is carried by 80% of healthy individuals at
concentrations between 1057 CFU/g of faeces; E. faecium is carried by 30% of
healthy individuals at lower concentrations. Enterococci are resistant to many
antimicrobials but in general sensitive to glycopeptides (vancomycin, teicopl-
anin) and linezolid. A VRE is defined as an Enterococcus with a minimum
inhibitory concentration (MIC) of C16 mg of vancomycin per litre [109].
(a) Linezolid sensitive VRE. Carriage of VRE even sensitive to linezolid is
abnormal [110]. As far as we are aware, there are no trials attempting to
eradicate VRE carriage. Theoretically, it is not impossible to clear abnor-
mal VRE carriage using vancomycin enterally: 2 g is associated with faecal
vancomycin levels varying between 3,000 and 24,000 mg per litre, with an
MIC around 16 mg/L [111].
Five RCTs of SDD involving 5,229 patients (2,631 SDD, 2,598 controls) reported
data on resistance [39, 43, 45, 50, 61]. There were 74 (2.8%) patients with resistant
microorganisms in the SDD group and 124 (4.8%) in controls, indicating a 44%
reduction in the odds of resistance by SDD (OR 0.56, 95% CI 0.410.76;
p \ 0.001) (Fig. 28.1). Heterogeneity was not shown (v2 = 2.58888, p = 0.63;
I2 = 0).
Fig. 28.1 Meta-analysis of five randomised controlled trials (RCTs) of selective decontamina-
tion of the digestive tract (SDD) with relevant data on resistance Odds ratio\1 favours SDD; [1
favours controls. Results presented with the fixed-effect model, as heterogeneity was not
demonstrated T+, test; T, control Brun-Buisson [39], de Smet [45], Flaherty [50], Laggner [61],
de Jonge [43]
However, the sensitivity analysis showed that after excluding the de Jonge trial
[43], the reduction in resistance was not significant (OR 0.75, 95% CI 0.351.61;
p = 0.46; heterogeneity not significant).
In recent years, RCTs have shown that five different manoeuvres reduce mortality
rates:
ventilation with low tidal volumes for acute lung injury and respiratory distress
syndrome [112];
recombinant human activated protein C for severe sepsis [113];
intensive insulin therapy [114];
low doses of steroids in patients with septic shock [115];
SDD [43, 45, 60].
Table 28.5 reports the levels of evidence obtained using the Grading of
Recommendations Assessment, Development and Evaluation (GRADE) system
[116], which classifies the quality of evidence as high (A), moderate (B), low (C)
or very low (D). RCTs may be downgraded due to limitations in implementation,
inconsistency or imprecision of results, indirectness of the evidence and possible
reporting bias [116]. An example of this is tight glucose control (A down to C): the
28 Antimicrobial Resistance 461
Table 28.5 Intensive care unit (ICU) interventions that reduce mortality rates
success of the original Belgian RCT [114] in reducing mortality rates has not to
date been reproduced [117121]. The GRADE system classifies recommendations
as strong (1) or weak (2). The grade of strong or weak is considered of greater
clinical importance than a difference in letter level of quality of evidence. A strong
recommendation in favour of an intervention reflects that the desirable effects of
adherence to a recommendation (beneficial health outcomes, less burden on staff
and patients and cost savings) will clearly outweigh the undesirable effects (harms,
more burdens, greater costs).
All RCTs and meta-analyses of SDD that assessed the full four-component
protocol consistently demonstrated a significant survival benefit, providing the
sample size was large enough. Mortality data show an intriguing observation that
trial design determines the magnitude of the survival benefit [122]. The relative
reduction in the OR for mortality was 41% when all patients received the full
SDD protocol [43], 29% when half the patients received SDD [122] and 17%
when one third of the population was treated with SDD [45, 123]. In the trial of
the unit-wide application of SDD [43], SDD virtually eliminated transmission of
potential pathogens via the hands of carers and hence exogenous infection in
decontaminated patients. Survival benefit is diluted by mixing decontaminated
and nondecontaminated patients in the same unit. This is the case in the RCT
design, wherein patients receiving and not receiving SDD are treated within the
same unit [45, 122]. Decontaminated patients protect control patients from
transmission, acquisition, carriage, and subsequent infection, whereas patients
receiving SDD remain at risk of acquiring potential pathogens and subsequent
exogenous infections, resulting in a reduction in the true effect of SDD. The
most recent multicentre RCT with a 17% relative reductionalbeit statistically
significantclearly emphasises the issue of diluting the SDD effect by
increasing the number of nondecontaminated patients treated in the same unit
with patients receiving SDD [45, 122].
462 N. Taylor et al.
Although the cost-effectiveness of SDD has not yet been formally calculated, the
daily costs of 612 euros [45, 70] can hardly be an issue for an ICU intervention
that reduces pneumonia, septicaemia and mortality rates by 72, 37 and 29%,
respectively (Table 28.4).
References
1. WFSICCM congress Firenze 2009 Gullo A (2009) Intensive and Critical Care Medicine.
Springer, Milan, pp 273283
2. Johanson WG, Pierce AK, Sanford JP (1969) Changing pharyngeal bacterial flora of
hospitalized patients. Emergence of Gram-negative bacilli. N Engl J Med 281:11371140
3. Viviani M, van Saene HK, Pisa F et al (2010) The role of admission surveillance cultures in
patients requiring prolonged mechanical ventilation in the intensive care unit. Anaesth
Intensive Care 38:325335
4. Mobbs KJ, van Saene HK, Sunderland D, Davies PD (1999) Oropharyngeal Gram-negative
bacillary carriage: a survey of 120 healthy individuals. Chest 115:15701575
5. van Saene HKF, Damjanovic V, Murray AE, de la Cal MA (1996) How to classify infections in
intensive care unitsthe carrier state, a criterion whose time has come? J Hosp Infect 33:112
6. de la Cal MA, Cerd E, van Saene HKF et al (2004) Effectiveness and safety of enteral
vancomycin to control endemicity of methicillin-resistant Staphylococcus aureus in a
medical/surgical intensive care unit. J Hosp Infect 56:175183
7. Stoutenbeek CP, van Saene HKF, Liberati A (1994) Prevention of respiratory tract infections
in intensive care by selective decontamination of the digestive tract. In: Niederman MS,
Sarosi GA, Glassroth J (eds) Respiratory infections, 1st edn. A scientific basis for
management, Philidelphia, pp 579594
8. Silvestri L, van Saene HK, de la Cal MA, De Gaudio AR (2009) Carriage classification of
pneumonia rather than time improves survival. Chest 136:11881189
9. Vollaard EJ (1991) The concept of colonization resistance. PhD thesis, Benda BV,
Nijmegen, The Netherlands
10. Husebye E (1995) Gastrointestinal motility disorders and bacterial overgrowth. J Intern Med
237:419427
11. Van Uffelen R, van Saene HK, Fidler V, Lwenberg A (1984) Oropharyngeal flora as a
source of bacteria colonizing the lower airways in patients on artificial ventilation. Intensive
Care Med 10:233237
12. Deitch EA, Xu DZ, Qi L, Berg RD (1991) Bacterial translocation from the gut impairs
systemic immunity. Surgery 104:269276
13. Baue AE (1993) The role of the gut in the development of multiple organ dysfunction in
cardiothoracic patients. Ann Thorac Surg 55:822829
14. van Saene HKF, Taylor N, Damjanovic V, Sarginson RE (2008) Microbial gut overgrowth
guarantees increased spontaneous mutation leading to polyclonality and antibiotic resistance
in the critically ill. Curr Drug Targets 9:419421
15. Horton JW, Maass DL, White J, Minei JP (2007) Reducing susceptibility to bacteremia after
experimental burn injury: a role for selective decontamination of the digestive tract. J Appl
Physiol 102:22072216
16. Conraads VM, Jorens PG, De Clerck LS (2004) Selective intestinal decontamination in
advanced chronic heart failure: a pilot trial. Eur J Heart Fail 6:483491
17. van Saene HKF, Stoutenbeek CP, Zandstra DF (1988) Cefotaxime combined with selective
decontamination in long term intensive care patients. Virtual absence of emergence of
rsistance. Drugs 35(Suppl 2):2934
28 Antimicrobial Resistance 463
37. Boland JP, Sadler DL, Stewart WA et al. (1991)Reduction of nosocomial respiratory tract
infections in multiple trauma patient requiring mechanical ventilation by selective
parenteral and enteral antisepsis regimen (SPEAR) in the intensive care unit. 17th
International congress of chemotherapy, Berlin, Abstract 0465
38. Bouter H, Schippers EF, Luelmo SA et al (2002) No effect of preoperative selective gut
decontamination on endotoxemia and cytokine activation during cardiopulmonary bypass: a
randomized, placebo-controlled study. Crit Care Med 30:3843
39. Brun-Buisson C, Legrand P, Rauss A et al (1989) Intestinal decontamination for control of
nosocomial multiresistant Gram-negative bacilli. Study of an outbreak in an intensive care
unit. Ann Intern Med 110:873881
40. Camus C, Bellissant E, Sebille V et al (2005) Prevention of acquired infections in
intubated patients with the combination of two decontamination regimens. Crit Care Med
33:307314
41. Cerra FB, Maddaus MA, Dunn DL et al (1992) Selective gut decontamination reduces
nosocomial infections and length of stay but not mortality or organ failure in surgical
intensive care unit patients. Arch Surg 127:163169
42. Cockerill FR 3rd, Muller SR, Anhalt JP et al (1992) Prevention of infection in critically ill
patients by selective decontamination of the digestive tract. Ann Intern Med 117:545553
43. de Jonge E, Schultz MJ, Spanjaard L et al (2003) Effects of selective decontamination of
digestive tract on mortality and acquisition of resistant bacteria in intensive care: a
randomised controlled trial. Lancet 362:10111016
44. de la Cal MA, Cerd E, Garca-Hierro P et al (2005) Survival benefit in critically ill burned
patients receiving selective decontamination of the digestive tract: a randomized, placebo-
controlled, double-blind trial. Ann Surg 241:424430
45. de Smet AM, Kluytmans JA, Cooper BS et al (2009) Decontamination of the digestive tract
and oropharynx in ICU patients. N Engl J Med 360:2031
46. Diepenhorst GM, van Ruter O, Besselink MG, van Santvoort HC, Wijnandts PR, Renooij
W, Gouma DJ, Goosen HG, Boermeester MA (2011) Influence of prophylactic probiotics
and selective decontamination on bacterial translocation in patients undergoing pancreatic
surgery: a randomized controlled trial. Shock 35: 916
47. Farran L, Llop J, Sans M et al (2008) Efficacy of enteral decontamination in the prevention
of anastomotic dehiscence and pulmonary infection in esophagogastric surgery. Dis
Esophagus 21:159164
48. Ferrer M, Torres A, Gonzlez J et al (1994) Utility of selective digestive decontamination in
mechanically ventilated patients. Ann Intern Med 120:389395
49. Finch RG, Tomlinson P, Holiday M, Sole K, Stack C, Rocker G. (1991) Selective
decontamination of the digestive tract (SDD) in the prevention of secondary sepsis in a
medical/surgical intensive care unit. 17th International congress of chemotherapy, Berlin,
Abstract 0474
50. Flaherty J, Nathan C, Kabins SA, Weinstein RA (1990) Pilot trial of selective
decontamination for prevention of bacterial infection in an intensive care unit. J Infect
Dis 162:13931397
51. Gastinne H, Wolff M, Delatour F, Faurisson F, Chevret S (1992) A controlled trial in
intensive care units of selective decontamination of the digestive tract with nonabsorbable
antibiotics. The French Study Group on Selective Decontamination of the Digestive Tract.
N Eng J Med 326:594599
52. Gaussorgues PH, Salord F, Sirodot M, Tigaud S, Cagin S, Gerard M, Robert D (1991)
Efficacite de la decontamination digestive sur la survenue des bacteriemies nosocomiales
chez les patients sous ventilation mecanique et recevant des betamimetiques. Rean Soins
Intens Med Urg 7:169174
53. Georges B, Mazerolles M, Decun JF, Rouge P, Pomies S, Cougot P, Andrieu P, Virenque
CH (1994) Dcontamination Digestive Slective Rsultats Dune tude Chez Le
Polytraumatis. Rean Urg 3:621627
28 Antimicrobial Resistance 465
54. Gosney M, Martin MV, Wright AE (2006) The role of selective decontamination of the
digestive tract in acute stroke. Age Ageing 35:4247
55. Hammond JM, Potgieter PD, Saunders GL, Forder AA (1992) Double-blind study of
selective decontamination of the digestive tract in intensive care. Lancet 340:59
56. Hellinger WC, Yao JD, Alvarez S, Blair JE, Cawley JJ, Paya CV et al (2007) A randomized,
prospective, double-blinded evaluation of selective bowel decontamination in liver
transplantation. Transplantation 73:19041909
57. 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 Care 3:5258
58. Kerver AJ, Rommes JH, Mevissen-Verhage EA, Hulstaert PF, Vos A, Verhoef J et al (1988)
Prevention of colonization and infection in critically ill patients: a prospective randomized
study. Crit Care Med 16:10871093
59. Korinek AM, Laisne MJ, Nicolas MH, Raskine L, Deroin V, Sanson-Lepors MJ et al (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
60. Krueger WA, Lenhart FP, Neeser G, Ruckdeschel G, Schreckhase H, Eissner HJ 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, double-blind, placebo-controlled clinical trial. Am J Respir Crit Care
Med 166:10291037
61. Laggner AN, Tryba M, Georgopoulos A et al (1994) Oropharyngeal decontamination with
gentamicin for long-term ventilated patients on stress ulcer prophylaxis with sucralfate?
Wien Klin Wochenschr 106:1519
62. Lingnau W, Berger J, Javorsky F, Lejeune P, Mutz N, Benzer H et al (1997) Selective
intestinal decontamination in multiple trauma patients: prospective, controlled trial.
J Trauma 42:687694
63. Luiten EJ, Hop WC, Lange JF, Bruining HA (1995) Controlled clinical trial of selective
decontamination for the treatment of severe acute pancreatitis. Ann Surg 222:5765
64. Martinez-Pells AE, Merino P, Bru M, Conejero R, Seller G, Muoz C et al (1992) Can
selective digestive decontamination avoid the endotoxemia and cytokine activation
promoted by cardiopulmonary bypass? Crit Care Med 21:16841691
65. Martinez-Pells AE, Merino P, Bru M, Canovas J, Seller G, Sapia J et al (1997)
Endogenous endotoxemia of intestinal origin during cardiopulmonary bypass. Role of type
of flow and protective effect of selective digestive decontamination. Intensive Care Med
23:12511257
66. Oudhuis GJ, Bergmans DC, Dormans T, Zwaveling JH, Kessels A, Prins MH, Stobberingh
EE, Verbon A (2011) Probiotics versus antibiotic decontamination of the digestive tract:
infection and mortality. Intensive Care Med 37:110-117
67. Palomar M, Alvarez-Lerma F, Jorda R, Bermejo B (1997) Catalan study group of
nosocomial pneumonia prevention. Prevention of nosocomial infection in mechanically
ventilated patients: selective digestive decontamination versus sucralfate. Clin Intensive
Care 8:228235
68. Pneumatikos I, Koulouras V, Nathanail C, Geo D, Nakos G (2002) Selective
decontamination of subglottic area in mechanically ventilated patients with multiple
trauma. Intensive Care Med 28:432437
69. Pugin J, Auckenthaler R, Lew DP, Suter PM (1991) Oropharyngeal decontamination
decreases incidence of ventilator-associated pneumonia. A randomized, placebo-controlled,
double-blind clinical trial. JAMA 265:27042710
70. Quinio B, Albanse J, Bues-Charbit M, Viviand X, Martin C (1996) Selective
decontamination of the digestive tract in multiple trauma patients. A prospective double-
blind, randomized, placebo-controlled study. Chest 109:765772
466 N. Taylor et al.
71. Rayes N, Seehofer D, Hansen S et al (2002) Early enteral supply of Lactobacillus and fiber
versus selective bowel decontamination: a controlled trial in liver transplant recipients.
Transplantation 74:123128
72. Rios F, Maskin B, Saenex Valiente A, Galante A, Cazes Camarero P, Aguilar L et al (2005)
Prevention of ventilator associated pneumonia (VAP) by oral decontamination (OD).
Prospective, randomized, double-blind, placebo-controlled study. American Thoracic
Society International Conference, San Diego, USA, C95; poster 608
73. Rocha LA, Martin MJ, Pita S, Paz J, Seco C, Margusina L et al (1992) Prevention of
nosocomial infection in critically ill patients by selective decontamination of the digestive
tract. A randomized, double blind, placebo-controlled study. Intensive Care Med
18:398404
74. Rodrguez-Roldn JM, Altuna-Cuesta A, Lpez A, Carrillo A, Garcia J, Len J et al (1990)
Prevention of nosocomial lung infection in ventilated patients: use of an antimicrobial
pharyngeal nonabsorbable paste. Crit Care Med 18:12391242
75. 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 17:196201
76. Rolando N, Wade JJ, Stangou A, Gimson AE, Wendon J, Philpott-Howard J et al (1996)
Prospective study comparing the efficacy of prophylactic parenteral antimicrobials, with or
without enteral decontamination, in patients with acute liver failure. Liver Transpl Surg
2:813
77. Roos D, Dijksman LM, Oudemans-van Straaten HM, de Wit LT, Gouma DJ, Gerhards MF
(2011) Randomized clinical trial of perioperative selective decontamination of the digestive
tract versus placebo in elective gastrointestinal surgery. Br J Surg [Epub ahead of print]
78. Ruza F, Alvarado F, Herruzo R, Delagado MA, Garcia S, Dorao P et al (1998) Prevention of
nosocomial infection in a pediatric intensive care unit (PICU) through the use of selective
digestive decontamination. Eur J Epidemiol 14:719727
79. Snchez Garca M, Cambronero Galache JA, Lpez Diaz J, Cerd Cerd E, Rubio Blasco J,
Gmez Aguinaga MA et 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 158:908916
80. Schardey HM, Joosten U, Finke U Staubach KH, Schauer R, Heiss A et al (1997) The
prevention of anastomotic leakage after total gastrectomy with local decontamination. A
prospective, randomized, double-blind, placebo-controlled multicenter trial. Ann Surg
225:172180
81. Smith SD, Jackson RJ, Hannakan CJ, Wadowsky RM, Tzakis AG, Rowe MI (1993)
Selective decontamination in pediatric liver transplants. A randomized prospective study.
Transplantation 55:13061309
82. Stoutenbeek CP, van Saene HKF, Zandstra DF (1996) Prevention of multiple organ
system failure by selective decontamination of the digestive tract in multiple trauma
patients. Faist E, Baue AE, Schildberg FW (Eds.) In: The immune Consequence of
Trauma, Shock and SepsisMechanisms and Therapeutic Approaches. Pabst Science,
Lengerich, pp 10551066
83. Stoutenbeek CP, van Saene HK, Little RA, Whitehead A for the Working Group on
Selective Decontamination of the Digestive Tract (2007) The effect of selective
decontamination of the digestive tract on mortality in multiple trauma patients: a
multicenter randomized controlled trial. Intensive Care Med 33:261270
84. Tetteroo GW, Wagenvoort JH, Castelein A, Tilanus HW, Ince C, Bruining HA (1990)
Selective decontamination to reduce Gram-negative colonisation and infections after
oesophageal resection. Lancet 335:704707
85. 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 15:424431
28 Antimicrobial Resistance 467
29.1 Introduction
J. C. Marshall (&)
Department of Surgery and Interdepartmental Division of Critical Care,
General Hospital and University of Toronto, Toronto, Canada
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 469
DOI: 10.1007/978-88-470-1601-9_29, Springer-Verlag Italia 2012
470 J. C. Marshall and K. A. M. Marshall
Table 29.1 Types of economic analyses (adapted from Second American Thoracic Society
Workshop on Outcomes Research [12])
Fagon et al. [28] evaluated the impact of VAP on ICU outcome. They demonstrated
a crude mortality rate of 52.4% for 328 patients developing pneumonia, signifi-
cantly higher than the 22.4% ICU mortality experienced by the 1,650 patients who
did not develop pneumonia. The APACHE II score, number of failing organs,
presence of pneumonia, development of nosocomial bacteremia, presence of sig-
nificant underlying disease, and admission from another ICU were all indepen-
dently associated with adverse outcome by logistic regression analysis. Similarly, a
French casecontrol study suggested that VAP was responsible for a twofold
increase in mortality and a 5-day prolongation of ICU stay [29]. A Canadian study
found that pneumonia was associated with an increased length of stay and a trend
toward an increased mortality rate [9]. In contrast, a Spanish study of 1,000 con-
secutive ICU admissions found that the development of VAP increased the length
of stay, but not the mortality rate, of ventilated ICU patients [30]. This conclusion
was also reached by a French matched casecontrol study, which showed rates of
VAP to be comparable in patients who died while in the ICU compared with those
who survived, when matched on the basis of a panel of risk factors for adverse
outcome [31]. A study of pneumonia complicating the course of acute respiratory
distress syndrome reported that the development of VAP increases the duration of
476 J. C. Marshall and K. A. M. Marshall
mechanical ventilation but does not adversely impact on patient survival [32].
Pooled data from a systematic review suggest that the development of VAP results
in an increased ICU length of stay of 4 days and an attributable mortality rate of
2030% [33].
There is little available evidence regarding the attributable morbidity and mortality
rates of other nosocomial infections in critically ill patients. Urinary tract infec-
tions, although relatively common, are generally thought to be of only modest
clinical significance. For example, an Argentinian study reported that catheter-
related infections, the most common ICU-acquired infection (comprising 32% of
all infections), carry an attributable mortality of 25% and are associated with an
excess length of stay of 11 days. VAP (25% of all nosocomial infections) was
associated with a 35% attributable mortality and a prolongation of ICU stay of
10 days. In contrast, urinary tract infections (23% of infections) had a 5%
attributable mortality and an increased length of ICU stay of 5 days [34].
Several authors have attempted to generate estimates of the costs associated with
infection in critically ill patients. Angus et al. [40] used administrative data to
generate an estimate that there are approximately 750,000 new cases of severe
sepsis (sepsis in association with organ dysfunction) in the United States each year
and that the total costs attributable to these are US $16.7 billion annually. However,
these estimates include cases of community-acquired sepsis and sepsis developing in
patients who are not in an ICU, and they fail to include the costs of episodes of
nosocomial ICU-acquired infection that do not meet the criteria for severe
sepsis. Brun-Buisson et al. [41] found that the costs of sepsis in association with
478 J. C. Marshall and K. A. M. Marshall
Fig. 29.1 The relationship between cost and clinical effectiveness. Increasing costs are
represented on the y axis, with increasing clinical benefit on the x axis. Examples shown are
approximations of the incremental cost associated with increased (or reduced) clinical benefit
ICU-acquired infection were three times higher than those incurred when sepsis was
present at the time of ICU admission. The total costs were approximately 40,000
for each patient developing sepsis in the ICU. Nosocomial infection complicating
community-acquired sepsis increased costs by 2.5 times. These observations mirror
those of a British study that found a fivefold increase in costs when patients devel-
oping sepsis after the second day of their ICU stay were compared with patients
admitted with a diagnosis of sepsis [42]. Thus, preventing nosocomial infection in the
ICU has the potential to significantly impact on the costs of ICU care.
Pittet et al. [43] reported that nosocomial bloodstream infections complicate the
course of 3% of patients admitted to an ICU and prolong both the ICU and hospital
stay, generating costs of approximately US $40,000 per survivor. Similar estimates
have been derived by others. Digiovine et al. [23], for example, found that
although nosocomial bloodstream infection in the ICU did not increase mortality
rates, it was associated with increased direct costs of US $34,508 per episode,
whereas Dimick et al. (unpublished data) suggested that catheter-related blood-
stream infection in the ICU results in increased total hospital costs of US $56,167
per case. Two analyses concluded that the use of antibiotic-coated catheters is cost
effective in preventing nosocomial bloodstream infections [44, 45]. However, the
conclusion is highly dependent on the estimate of the efficacy of such catheters,
29 ICU-Acquired Infection: Mortality, Morbidity, and Costs 479
and intrinsic limitations in the design of the studies evaluating them limit the
estimate of their benefits [46].
Warren et al. [47] estimated the attributable cost of an episode of VAP in the United
States to be approximately US $12,000. Because a variety of prophylactic strategies
have been shown to be effective in preventing VAP [48], and because these are
generally relatively inexpensive to institute, VAP prevention is readily demonstrable
to be cost effective. Zack et al. [49], for example, showed that instituting a com-
prehensive preventive program resulted in a 57.6% reduction in VAP rate and in cost
savings of as much as US $4 million/year. Other strategies, such as minimizing
intubation through the use of noninvasive positive-pressure ventilation [50] and
reducing the frequency of ventilator circuit changes [51] are also cost effective.
Our focus has been on infectious complications that are amenable to prevention, in
no small part because there are few proven effective therapies for infection in the
ICU. Thus, although it is widely believed that specific antimicrobial therapy,
adequate surgical source control, and the spectrum of supportive measures that
comprise ICU care will improve clinical outcome, the attributable effect of any of
480 J. C. Marshall and K. A. M. Marshall
these in the patient with infection is unknown, and therefore cost-utility analyses
are impossible.
The approval of activated protein C for treating patients with severe sepsis has
provided the first opportunity for cost-utility analyses in critically ill infected
patients. Treatment with activated protein C has been shown in a cohort of patients
with severe sepsis to reduce mortality rates by 6.1% but at a cost of approximately
US $7,000 per course of therapy. Two independent analyses of the cost-
effectiveness of activated protein C show a favorable profile when it is used in the
sickest patients. Manns et al. [38] calculated the cost per life-year gained to be US
$27,936 for all patients in the cohort, whereas Angus et al. [58] suggested that the
cost-utility of activated protein C is US $48,800 per quality-adjusted life year.
29.7 Conclusions
References
1. National Nosocomial Infections Surveillance System (NNIS) (2002) System report, data
summary from January 1992 to June 1992, issued August 2002. Am J Infect Control 30:458475
2. Richards MJ, Edwards JR, Culver DH et al (1999) Nosocomial infections in medical
intensive care units in the United States. National nosocomial infections surveillance system.
Crit Care Med 27:887892
3. Marshall JC, Sweeney D (1990) Microbial infection and the septic response in critical
surgical illness. Sepsis, not infection, determines outcome. Arch Surg 125:1723
29 ICU-Acquired Infection: Mortality, Morbidity, and Costs 481
27. Blot SI, Vandewoude KH, Hoste EA, Colardyn FA (2002) Outcome and attributable
mortality in critically ill patients with bacteremia involving methicillin-susceptible and
methicillin-resistant Staphylococcus aureus. Arch Intern Med 162:22292235
28. Fagon J-Y, Chastre J, Vuagnat A et al (1996) Nosocomial pneumonia and mortality among
patients in intensive care units. JAMA 275:866869
29. Bercault N, Boulain T (2001) Mortality rate attributable to ventilator-associated nosocomial
pneumonia in an adult intensive care unit: a prospective case-control study. Crit Care Med
29:23032309
30. Rello J, Quintana E, Ausina V et al (1991) Incidence, etiology, and outcome of nosocomial
pneumonia in mechanically ventilated patients. Chest 100:439444
31. Bregeon F, Ciais V, Carret V et al (2001) Is ventilator-associated pneumonia an independent
risk factor for death? Anesthesiology 94:554560
32. Markowicz P, Wolff M, Djedaini K et al (2000) Multicenter prospective study of ventilator-
associated pneumonia during acute respiratory distress syndrome. Am J Respir Crit Care Med
161:19421948
33. Cook D (2000) Ventilator associated pneumonia: perspectives on the burden of illness.
Intensive Care Med 26(1):S31S37
34. Rosenthal VD, Guzman S, Orellano PW (2003) Nosocomial infections in medical-surgical
intensive care units in Argentina: attributable mortality and length of stay. Am J Infect
Control 31:291295
35. Singh N, Rogers P, Atwood CW et al (2000) Short-course empiric antibiotic therapy for
patients with pulmonary infiltrates in the intensive care unit. A proposed solution for
indiscriminate antibiotic prescription. Am J Respir Crit Care Med 162:505511
36. Fagon J-Y, Chastre J, Wolff M et al (2000) Invasive and noninvasive strategies for
management of suspected ventilator-associated pneumonia. A randomized trial. Ann Intern
Med 132:621630
37. Hebert PC, Wells G, Blajchman MA et al (1999) A multicentre randomized controlled
clinical trial of transfusion requirements in critical care. N Engl J Med 340:409417
38. Manns BJ, Lee H, Doig CJ et al (2002) An economic evaluation of activated protein C
treatment for severe sepsis. N Engl J Med 347:9931000
39. Takala J, Ruokonen E, Webster NR et al (1999) Increased mortality associated with growth
hormone treatment in critically ill adults. N Engl J Med 341:785792
40. Angus DC, Linde-Zwirble WT, Lidicker J et al (2001) Epidemiology of severe sepsis in the
United States: analysis of incidence, outcome, and associated costs of care. Crit Care Med
29:13031310
41. Brun-Buisson C, Roudot-Thoaval F, Girou E et al (2003) The costs of septic syndromes
in the intensive care unit and influence of hospital-acquired sepsis. Intensive Care Med
29:14641471
42. Edbrooke DL, Hibbert CL, Kingsley JM et al (1999) The patient-related costs of care for
sepsis patients in a United Kingdom adult general intensive care unit. Crit Care Med
27:17601767
43. Pittet D, Tarara D, Wenzel RP (1994) Nosocomial bloodstream infection in critically ill
patients. Excess length of stay, extra costs, and attributable mortality. JAMA 271:15981601
44. Shorr AF, Humphreys CW, Helman DL (2003) New choices for central venous catheters:
potential financial implications. Chest 124:275284
45. Marciante KD, Veenstra DL, Lipsky BA, Sainst S (2003) Which antimicrobial impregnated
central venous catheter should we use? modeling the costs and outcomes of antimicrobial
catheter use. Am J Infect Control 31:18
46. McConnell SA, Gubbins PO, Anaissie EJ (2003) Do antimicrobial-impregnated central
venous catheters prevent catheter-related bloodstream infection? Clin Infect Dis 37:
6572
29 ICU-Acquired Infection: Mortality, Morbidity, and Costs 483
47. Warren DK, Shukla SJ, Olsen MA et al (2003) Outcome and attributable cost of ventilator-
associated pneumonia among intensive care unit patients in a suburban medical center. Crit
Care Med 31:13121317
48. Kollef MH (1999) The prevention of ventilator-associated pneumonia. N Engl J Med
340:627634
49. Zack JE, Garrison T, Trovillion E et al (2002) Effect of an education program aimed at
reducing the occurrence of ventilator-associated pneumonia. Crit Care Med 30:24072412
50. Sinuff T, Cook DJ (2003) Health technology assessment in the ICU: noninvasive positive
pressure ventilation for acute respiratory failure. J Crit Care 18:5967
51. Kotilainen HR, Keroack MA (1997) Cost analysis and clinical impact of weekly ventilator
circuit changes in patients in intensive care unit. Am J Infect Control 25:117120
52. Niederman MS (2001) Impact of antibiotic resistance on clinical outcomes and the cost of
care. Crit Care Med 29(Suppl):N114N120
53. 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 282:17451751
54. Cosgrove SE, Kaye KS, Eliopoulos GM, Carmeli Y (2002) Health and economic outcomes of
the emergence of third-generation cephalosporin resistance in Enterobacter species. Arch
Intern Med 162:185190
55. Carmeli Y, Eliopoulos G, Mozaffari E, Samore M (2002) Health and economic outcomes of
vancomycin-resistant enterococci. Arch Intern Med 162:22232228
56. Pelz RK, Lipsett PA, Swoboda SM et al (2002) Vancomycin-sensitive and vancomycin-
resistant enterococcal infections in the ICU: attributable costs and outcomes. Intensive Care
Med 28:692697
57. Bantar C, Sartori B, Vesco E et al (2003) A hospital-wide intervention program to optimize
the quality of antibiotic use: impact on prescribing practice, antibitoic consumption, cost
savings, and bacterial resistance. Clin Infect Dis 37:180186
58. Angus DC, Linde-Zwirble WT, Clermont G et al (2003) Cost-effectiveness of drotrecogin
alfa (activated) in the treatment of severe sepsis. Crit Care Med 31:111
Evidence-Based Medicine in ICU
30
A. J. Petros, K. G. Lowry, H. K. F. van Saene and J. C. Marshall
30.1 Introduction
Evidence-based medicine (EBM) was extolled by David Sackett, who described two
processes: one for assessing the quality of a therapy on a scale of 14, and a second
for making recommendations for using that therapy on a scale of AD. However,
more recently, a newer method of grading the quality of evidence and strength of
recommendation for a new therapy has been developed. The Grading of Recom-
mendations Assessment, Development and Evaluation (GRADE) Working Group
reported its suggestions in 2004, with further refinement in 2008 [1, 2]. The use of a
structured approach to collect, analyse and summarise all the relevant evidence
allows the production of grades of recommendations. GRADE is being increasingly
used as the structure on which to develop guidelines [3] and is used widely by the
World Health Organization, the American College of Physicians, the American
Thoracic Society, the Cochrane Collaboration and many other organisations, with
up to 25 groups demonstrating GRADEs success as a methodology [4].
GRADE guides assessment of the quality of evidence for a particular treatment or
therapy in one of four levelshigh (A), moderate (B), low (C) and very low (D).
Study design, quality, consistency and directness are all assessed. The factors
influencing the decision on quality are described in Table 30.1. Evidence from
randomised controlled trials (RCTs) contributes to high-quality evidence, but
confidence in that evidence may be decreased for several reasons: study limita-
tions; inconsistent results; indirectness of evidence; imprecision; reporting bias [1]
(Table 30.1). Observational studies, such as cohort and case-control studies, start
with a low-quality rating; grading upwards may be possible if, for example,
A. J. Petros (&)
PICU, Great Ormond Street Hospital, London, UK
e-mail: [email protected]
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 485
DOI: 10.1007/978-88-470-1601-9_30, Springer-Verlag Italia 2012
486 A. J. Petros et al.
Table 30.1 Descriptions of levels and quality used in the GRADE system
the size of the treatment effect is very large, or if there is a strong causal rela-
tionship. GRADE also makes recommendations from strong (1) to weak (2).
The former is where the intervention clearly outweighs its undesirable effects and
the latter where the trade-off between desirable and undesirable effects is less
clear. Making recommendations for a specific therapy involves a balance between
benefits and harms and inevitably involves placing a relative value on each
outcome, though it is difficult to judge how much weight to give to different
outcomes [1]. In making a recommendation, four main factors should be consid-
ered [1] (Table 30.2):
trade-offsthese should consider the estimated size of the effect for the main
outcomes, confidence limits around those estimates, and relative values placed
on each outcome;
quality of evidence;
30 Evidence-Based Medicine in ICU 487
Recommendations
Net benefits The intervention clearly does more good than harm
Trade-offs There are important trade-offs between benefits and harms
Uncertain trade-offs It is not clear whether the intervention does more good than harm
No net benefits The intervention clearly does not do more good than harm
translation of evidence into specific practice, allowing for factors that could
qualify the expected effect, such as proximity to a hospital or availability of
necessary expertise;
uncertainty about the baseline risk for the population of interest.
The strong or weak grading is felt to be of greater clinical important than
classifying the quality of the intervention.
Using GRADE provides a framework for structured assessment and can help
ensure that appropriate judgments are made about a new therapy or manoeuvre.
We screened intensive care unit (ICU) literature using these GRADE rules for
manoeuvres that may impact on infectious morbidity and mortality rates and
classified the most common manoeuvres according to levels of evidence and
grades of recommendations (Table 30.3).
It has never been shown in a RCT that hand hygiene prevents pneumonia and
reduces mortality rates in ventilated patients. The efficacy of hand hygiene on the
incidence of infection was studied in eight nonrandomised studies [512]
(Table 30.4); however, the incidence of pneumonia was not presented. The only
study demonstrating an impact on mortality due to hand hygiene was the cohort
study of Semmelweis in 1861 in postpartum women for reducing mortality due to
puerperal sepsis, with a decrease from 11 to 3% [13]. There are no data available
on the effect of isolation, protective clothing, equipment care and environment on
pneumonia and mortality rates in ventilated patients.
These five traditional infection control measures target microorganism trans-
mission via carriers hands. Although they are important, the impact should not be
overestimated. An optimal infection-control policy can only reduce infections
due to microorganisms acquired in the ICU, i.e. secondary endogenous and
exogenous infections. They fail to influence primary endogenous infections due to
488 A. J. Petros et al.
Table 30.3 Analysis of the literature, grading of evidence and recommendations for controlling
morbidity and mortality rates due to infection in ventilated patients in the intensive care unit
Table 30.4 Studies into the effect of hand hygiene on the incidence of nosocomial infections, including pneumonia
patients
Stasis of saliva contaminated with potential pathogens above the cuff on the
endotracheal tube increase the risk of aspiration pneumonia. Removing and pre-
venting this salivary stasis using continuous aspiration via a specially designed
endotracheal tube is thought to prevent pneumonia. The intervention of subglottic
secretion drainage (SSD) was evaluated in ten RCTs [2231] performed in a mixed
ICU population requiring ventilation for [72 h, and the fourth study in cardiac
surgery patients. Results were not consistent. Two studies showed a significant
reduction in pneumonia; the other two failed to show any impact on pneumonia
during ventilation. There was no difference in mortality rates between test and
control groups in any of these studies. Although the specially designed tubes and
suction equipment are expensive, this technique has been suggested to be cost
effective on theoretical grounds. There were no harmful side effects associated
with this manoeuvre in any of the studies. Bo et al. found that the presence of
subglottic secretion may be an origin of the pathogenetic organisms of VAP [27].
The morbidity rate of VAP in mechanically ventilated patients can be reduced by
SSD. Liu et al. confirmed that migration of the dominant bacteria of the subglottic
secretion was one of the important factors for ventilator-associated lower airway
30 Evidence-Based Medicine in ICU 493
There are 16 RCTs that report varying degrees of success with oropharyngeal
decontamination using antiseptics [3449]. However, the outcome of six meta-
analyses revealed that antiseptic usage has no benefit in reducing pneumonia or
mortality rates [5055] (Table 30.5). In 1,202 patients, Pineda et al. [50] reported
that use of oral decontamination with chlorhexidine did not result in significant
reduction in the incidence of nosocomial pneumonia in patients who received
mechanical ventilation, and it did not alter the mortality rate. Chlebicki and Safdar
[51] demonstrated no mortality benefit with chlorhexidine, though in seven small
RCTs there was a reduction in VAP, which was most marked in cardiac surgery
patients. Neither antiseptic nor antibiotic oral decontamination reduced mortality,
duration of mechanical ventilation or ICU stay in a meta-analysis of 11 studies by
Chan et al. [52]. Kola and Gastmeier [53] found in seven RCTs a reduction in RR
of lower respiratory tract infections in patients receiving chlorhexidine [RR
(random): 0.58]. However, these results only applied to patients ventilated for up
to 48 h. From 10 studiesbut not all RCTsCarvajal et al. [55] report a reduction
in the risk of VAP with chlorhexidine (OR 0.56, 95% CI 0.440.73). However, no
reduction in mortality rates, length of mechanical ventilation or ICU length of stay
was seen.
494
Table 30.6 Randomised controlled trials into the effect of nonantibiotic interventions on the general infection and mortality rates in ventilated patients
Manoeuvre Author Year Study Design No. Infection Rate Mortality Evidence
Immunonutrition Beale [56] 1999 Meta-analysis of 12 studies 1,482 RR 0.67 RR 0.05 2A
(0.500.89) (0.781.41)
P = 0.006 P = 0.76
Heyland [57] 2001 Meta-analysis of 22 studies 2,419 RR 0.66 RR 1.1 2A
(0.540.80) (0.931.31)
Steroids Cronin [58] 1995 Metaanalysis of 9 RCTs 1,232 No difference RR 1.13 2A
(0.991.29)
Lefering [59] 1995 Meta-analysis of 10 RCTs 1,329 No difference Difference in mortality 2A
0.2% (9.2 to 8.8)
Bollaert [60] 1998 RCT 41 No difference Difference in mortality 2A
31% (161)
Briegel [61] 1999 RCT 40 No difference No difference 2A
Annane [62] 2002 RCT 300 No difference Significant reduction 2A
RCT randomised controlled trial, RR relative risk (95% confidence intervals)
A. J. Petros et al.
30 Evidence-Based Medicine in ICU 495
30.2.5 Immunomodulation
30.2.5.2 Steroids
High doses of steroids given to septic patients are thought to be beneficial for three
reasons [5862] (Table 30.6): steroids effectively suppress generalised inflam-
mation due to microorganisms and their toxins; they have been shown to signif-
icantly reduce septic shock and early mortality within 72 h; they have been shown
to significantly reduce mortality rates due to particularly severe invasive infection,
including meningitis, typhoid and Pneumocystis carinii pneumonia (PCP). The
major perceived side effects of high-dose steroids are the associated immune
suppression and subsequent risk of superinfections. Indeed, the two meta-analyses
show a trend towards increased mortality rates from secondary infection in patients
receiving steroids. A systematic review by Annane et al. [63] examining the
benefits and risks of steroids in sepsis reviewed 17 RCTs encompassing 2,138
patients, and three quasi-RCTs of 246 patients. Sub group analysis of prolonged
low-dose corticosteroid therapy suggests a beneficial effect on short-term mortality
rates [63]. The Corticosteroid Treatment and Intensive Insulin Therapy for Septic
Shock in Adults (COIITSS) study [64] demonstrated that intensive insulin therapy
together with hydrocortisone for septic shock did not improve in-hospital mortality
rates. The addition of oral fludrocortisone did not result in a statistically significant
improvement in in-hospital mortality [64].
The next logical step would be to combine steroids with SDD, whereby the
perceived harmful effects of steroids could be abolished. In that way, the early
survival benefit from steroids can be preserved, while keeping the patient free from
secondary infections using SDD. The time has come to perform a randomised trial
of SDD and steroids versus SDD only, with the endpoint as mortality rate.
496 A. J. Petros et al.
30.2.5.3 Immunoglobulins
Polyclonal intravenously administered immunoglobulins significantly reduce
mortality rates and can be used as an extra treatment option for sepsis and septic
shock [65]. Overall mortality rates were reduced in patients who received poly-
clonal immunoglobulin i.v. (492; RR 0.64; 95% CI 0.510.80). For the two high-
quality trials on polyclonal immunoglobulin i.v., the RR for overall mortality was
0.30, but the CI was wide (0.090.99; n = 91). However, all trials were small, and
the totality of the evidence is insufficient to support a robust conclusion of benefit.
Adjunctive therapy with monoclonal immunoglobulins i.v. remains experimental.
conflicting results and tight glucose control can cause dangerous hypoglycaemia,
data underlying this recommendation should be critically evaluated [71].
An RCT by Vlasselaers et al. [72] of intensive insulin therapy to achieve age-
adjusted normal fasting concentrations showed improved short-term outcome of
patients in a paediatric ICU (PICU). However, the Neonatal Insulin Replacement
Therapy in Europe (NIRTURE) study of tight glucose control in neonates and
infants did not conclusively demonstrate the value of insulin therapy in preterm
infants [73].
The practice of tight glucose control is accompanied by an increased incidence
of hypoglycaemia. Hermanides et al. [74] demonstrated that hypoglycaemia
increased the rate of death to 40:1,000 in those who experienced hypoglycaemia
and 17:1,000 for those who were not hypoglycaemic.
498 A. J. Petros et al.
To control the
efficacy of PTA
The causative hospital PPM is
not carried in the patients
Hygiene Exogenous digestive tract and is introduced
directly into the sterile organ
Surveillance
To classify infections according
cultures
to the carrier state
To identify a resistance
problem
Fig. 30.1 The full four component protocol of SDD, that aims to control the three different types
of infection that occur on ICU
SDD is based on the observation that critical illness changes body flora, promoting
a shift: (1) from normal (Streptococcus pneumoniae in the throat and Escherichia
coli in the gut) towards abnormal [aerobic Gram-negative bacilli (AGNB) and
methicillin-resistant Staphylococcus aureus (MRSA) in throat and gut] carriage
(Table 30.7); (2) from low- to high-grade carriage (gut overgrowth) of both normal
and abnormal flora. Parenterally administered cefotaxime controls gut overgrowth
due to normal bacteria; enterally administered polyenes control gut overgrowth
30 Evidence-Based Medicine in ICU 499
Table 30.8 Carriage classification of severe infections of lower airways and blood
Author No. of Sample Lower airway Bloodstream Multiple organ dysfunction Mortality
RCTs size infection infection syndrome
OR (95% CI) OR (95% CI) OR (95% CI) OR (95% CI)
Vandenbroucke-Grauls 6 491 0.12, 0.080.19 NR 0.92, 0.45
[76] 1.84
DAmico [77] 33 5,727 0.35, 0.290.41 NR 0.80, 0.69
0.93
Safdar [78] 4 259 NR NR 0.82, 0.22
2.45
Liberati [79] 36 6,922 0.35, 0.290.41 NR 0.78, 0.68
0.89
Silvestri [80] yeasts 42 6,075 NR 0.89, 0.164.95 NR
Silvestri [81] 51 8,065 NR 0.63, 0.460.87 0.74, 0.61
0.91
Silvestri [82] 54 9,473 0.07, 0.040.13 0.36, 0.220.60 NR
G- 0.52, 0.340.78 1.03, 0.751.41 NR
G+
Silvestri [83] 21 4,902 NR NR 0.71, 0.61
0.82
Liberati [84] 36 6,914 0.28, 0.200.38 NR 0.75, 0.65
0.87
Silvestri [85] 7 1,270 NR NR 0.50, 0.340.74 0.82, 0.51
1.32
Silvestri [86] 12 2,252 0.54, 0.420.69 NR NR
A. J. Petros et al.
30 Evidence-Based Medicine in ICU 501
infection (OR 0.28, 95% CI 0.200.38). Bloodstream infection was the endpoint
in three meta-analyses [8082] and was significantly reduced (OR 0.63, 95% CI
0.460.87). When assessing bloodstream infection, AGNB septicaemias were
significantly reduced; Gram-positive ones were increased but not significantly
due to the low incidence in the control group (Table 30.9). Multiple organ
dysfunction syndrome (MODS) was the endpoint in one of the most recent
meta-analyses [85], in which the relative reduction of 50% was significant.
Mortality was the endpoint in eight meta-analyses [7679, 81, 8385]. SDD
consistently reduced mortality rates as long as the sample size was large
enough; the sample size was too small in three meta-analyses [76, 78, 85].
References
1. Atkins D, Best D, Briss PA et al, for the GRADE working group (2004) Grading quality of
evidence and strength of recommendations. BMJ 328:14901495
2. Schnemann HJ, Oxman AD, Brozek J et al, for the GRADE working group (2008) Grading
quality of evidence and strength of recommendations for diagnostic tests and strategies. BMJ
336:11061110
3. Jaeschke R, Guyatt GH, Dellinger P et al, for the GRADE working group (2008) Use of
GRADE grid to reach decisions on clinical practice guidelines when consensus is elusive.
BMJ 337:a744
4. Guyatt GH, Oxman AD, Vist GE et al, for the GRADE working group (2008) GRADE: an
emerging consensus on rating quality of evidence and strength of recommendations.
336:924926
5. Casewell M, Philips I (1977) Hands as route of transmission for Klebsiella species. BMJ
2:13151317
6. Massanari RM, Hierholzer J (1984) A cross-over comparison of antiseptic soaps on
nosocomial infection rates in the intensive care units. Am J Infect Control 12:247248
7. Maki DG (1989) The use of antiseptics for handwashing by medical personnel. J Chemother
1(Suppl 1):311
8. Simmons B, Bryant J, Neiman K et al (1990) The role of handwashing in prevention of
endemic intensive care unit infections. Infect Control Hosp Epidemiol 11:589594
9. Doebbeling RN, Stanley G, Sheetz CT et al (1992) Comparative efficacy of alternative
handwashing agents in reducing nosocomial infections in intensive care units. New Engl J
Med 327:8893
10. Webster J, Faogali JL, Cartwright D (1994) Elimination of methicillin-resistant Staphylococcus
aureus from a neonatal intensive care unit after handwashing with tricloson. J Paediatr Child
Health 30:5964
11. Koss WG, Khalili TM, Lemus JF et al (2001) Nosocomial pneumonia is not prevented by
protective contact isolation in the surgical intensive care unit. Am Surg 67:11401144
12. Slota M, Green M, Farley A et al (2001) The role of gown and glove isolation and strict
handwashing in the reduction of nosocomial infection in children with solid organ
transplantation. Crit Care Med 29:405412
13. Silvestri L, Petros AJ, Sarginson RE et al (2005) Handwashing in the intensive care unit: a
big measure with modest effects. J Hosp Infect 59:172179
14. Choi SC, Nelson LD (1992) Kinetic therapy in critically ill patients: combined results based
on meta-analysis. J Crit Care 7:5762
15. Summer WR, Curry P, Haponik EF et al (1989) Continuous mechanical turning of intensive care
unit patients shortens length of stay in some diagnostic-related groups. J Crit Care 4:4553
502 A. J. Petros et al.
16. Traver GA, Tyler ML, Hudson LD et al (1995) Continuous oscillation: outcome in critically
ill patients. J Crit Care 10:97103
17. Craven DE, Steger KA (1996) Nosocomial pneumonia in mechanically ventilated patients:
epidemiology and prevention in 1996. Semin Respir Infect 11:3253
18. Drakulovic MB, Torres A, Bauer TT et al (1999) Supine body position as a risk factor for
nosocomial pneumonia in mechanically ventilated patients: a randomised trial. Lancet
354:18511858
19. van Nieuwenhoven CA, van Tiel FH, Vandenbroucke-Grauls C et al (2002) The effect of
semi-recumbent position on development of ventilator-associated pneumonia (VAP).
Intensive Care Med 27(Suppl 2):S285, Abstract 585
20. Keeley L (2007) Reducing the risk of ventilator-acquired pneumonia through head of bed
elevation. Nurs Crit Care 12:287294
21. Silvestri L, Gregori D, van Saene HKF, Belli R (2010) Semirecumbent position to prevent
ventilator-associated pneumonia is not evidence based. J Critical Care 25:152153
22. Mahul P, Auboyer C, Jaspe R et al (1992) Prevention of nosocomial pneumonia in intubated
patients: respective role of mechanical subglottic secretions drainage and stress ulcer
prophylaxis. Intensive Care Med 18:2025
23. Valles J, Artigas A, Rello J et al (1995) Continuous aspiration of subglottic secretions in
preventing ventilator-associated pneumonia. Ann Intern Med 122:179186
24. Kollef MH, Skubas NJ, Sundt TM (1999) A randomised clinical trial of continuous aspiration
of subglottic secretions in cardiac surgery patients. Chest 116:13391346
25. Smulders K, van der Hoeven H, Weers-Pothoff I et al (2002) A randomised clinical trial of
intermittent subglottic secretion drainage in patients receiving mechanical ventilation. Chest
121:858862
26. Metz C, Linde HJ, Gobel L et al (1998) Influence of intermittent subglottic lavage on
subglottic colonization and ventilator associated pneumonia. Clin Intensive Care 9:2024
27. Bo H, He L, Qu J et al (2000) Influence of the subglottic secretion drainage on the morbidity
of ventilator associated pneumonia in mechanically ventilated patients. Zhonghua Jie He He
Hu Xi Za Zhi 23:472474
28. Liu SH, Yan XX, Cao SQ et al (2006) The effect of subglottic secretion drainage on
prevention of ventilator-associated lower airway infection. Zhonghua Jie He He Hu Xi Za Zhi
29:1922
29. Lorente L, Lecuona M, Jimenez A et al (2007) Influence of an endotracheal tube with
polyurethane cuff and subglottic secretion drainage on pneumonia. Am J Respir Crit Care
Med 176:10791083
30. Zheng RQ, Lin H, Shao J et al (2008) A clinical study of subglottic secretion drainage for
prevention of ventilator associated pneumonia. Zhongguo Wie Zhong Bing Ji Jiu Yi Xue
20:338340
31. Bouza E, Perez MJ, Munoz P et al (2008) Continuous aspiration of subglottic secretions
(CASS) in the prevention of ventilator-associated pneumonia in the postoperative period of
major heart surgery. Chest 134:938946
32. Dezfulian C, Shojania K, Collard HR et al (2005) Subglottic secretion drainage for
preventing ventilator associated pneumonia: a meta-analysis. Am J Med 118:1118
33. Silvestri L, Milanese M, van Saene HKF et al (2008) Impact of subglottic secretion drainage
on ventilator-associated pneumonia and mortality: systematic review of randomized
controlled trials. In: Proceedings of the 21st anesthesia and ICU symposium Alpe Adria.
Udine, 56 Sept 2008, pp 2629
34. De Riso AJII, Ladowski JS, Dillon TA et al (1996) Chlorhexidine gluconate 0.12% oral rinse
reduces the incidence of total nosocomial respiratory infection and nonprophylactic systemic
antibiotic use in patients undergoing heart surgery. Chest 109:15561561
35. Fourrier F, Cau-Pottier E, Boutigny H et al (2000) Effects of dental plaque on antiseptic
decontamination on bacterial colonisation and nosocomial infections in critically ill patients.
Intensive Care Med 26:12391247
30 Evidence-Based Medicine in ICU 503
56. Beale RJ, Bryg DJ, Bihari DJ (1999) Immunonutrition in the critically ill: a systematic review
of clinical outcome. Crit Care Med 27:27992805
57. Heyland DK, Novak F, Drover JW et al (2001) Should immunonutrition become routine in
critically ill patients? a systematic review of the evidence. JAMA 286:944953
58. Cronin L, Cook DJ, Carlet J et al (1995) Corticosteroid treatment for sepsis: a critical
appraisal and meta-analysis of the literature. Crit Care Med 23:14301439
59. Lefering R, Neugebauer EAM (1995) Steroid controversy in sepsis and septic shock: a meta-
analysis. Crit Care Med 23:12941303
60. Bollaert PE, Charpentier C, Levy B et al (1998) Reversal of late septic shock with
supraphysiologic doses of hydrocortisone. Crit Care Med 26:645650
61. Briegel J, Forst H, Haller M et al (1999) Stress doses of hydrocortisone reverse hyperdynamic
septic shock: a prospective, randomised, double-blind, single-center study. Crit Care Med
27:723732
62. Annane D, Sebille V, Charpentier C et al (2002) Effect of treatment with low doses of
hydrocortisone and fludrocortisone on mortality in patients with septic shock. JAMA
288:862871
63. Annane D, Bellissant E, Bollaert PE et al (2009) Corticosteroids in the treatment of severe
sepsis and septic shock in adults: a systematic review. JAMA 301:23622375
64. Annane D, Cariou A, Maxime V et al, for the COIITSS study investigators (2010) Corticosteroid
treatment and intensive insulin therapy for septic shock in adults: a randomized controlled trial.
JAMA 303:341348
65. Alejandria MM, Lansang MA, Dans LF, Mantaring JBV (2000) Intravenous immunoglobulin
for treating sepsis and septic shock (Cochrane review). In: The Cochrane library, issue 3.
Update Software, Oxford
66. Bernard GR, Vincent JL, Laterre PF et al (2001) Efficacy and safety of recombinant human
activated protein C for severe sepsis. New Engl J Med 344:699709
67. Marshall J (2000) Clinical trials of mediator-directed therapy in sepsis: what have we
learned? Intensive Care Med 26:575583
68. Zeni F, Freeman B, Natanson C (1997) Anti-inflammatory therapies to treat sepsis and septic
shock: a reassessment. Crit Care Med 25:10951100
69. van den Berghe G, Wouters P, Weekers F et al (2001) Intensive insulin therapy in the
critically ill patients. N Engl J Med 345:13591367
70. Van den Berghe G, Wilmer A, Hermans G (2006) Intensive insulin therapy in the medical
ICU. N Engl J Med 354:449461
71. Wiener RS, Wiener DC, Larson RJ (2008) Benefits and risks of tight glucose control in
critically ill adults: a meta-analysis. JAMA 300:933944
72. Vlasselaers D, Milants I, Desmet L et al (2009) Intensive insulin therapy for patients in
paediatric intensive care: a prospective, randomised controlled study. Lancet 373:547556
73. Beardsall K, Vanhaesebrouck S, Ogilvy-Stuart AL et al (2007) A randomised controlled trial
of early insulin therapy in very low birth weight infants, NIRTURE (neonatal insulin
replacement therapy in Europe). BMC Pediatr 7:29
74. Hermanides J, Bosman RJ, Vriesendorp TM (2010) Hypoglycemia is associated with
intensive care unit mortality. Crit Care Med 38:14301434
75. Stoutenbeek CP, van Saene HKF, Miranda DR et al (1984) The effect of selective
decontamination of the digestive tract on colonization and infection rate in multiple trauma
patients. Intensive Care Med 10:185192
76. Vandenbroucke-Grauls CMJ, Vandenbroucke JP (1991) Effect of selective decontamination
of the digestive tract on respiratory tract infections and mortality in the intensive care unit.
Lancet 338:859862
77. DAmico R, Pifferi S, Leonetti C et al, on behalf of the study investigators (1998)
Effectiveness of antibiotic prophylaxis in critically ill adult patients: systematic review of
randomised controlled trials. BMJ 316:12751285
30 Evidence-Based Medicine in ICU 505
78. Safdar N, Said A, Lucey MR (2004) The role of selective decontamination for reducing
infection in patients undergoing liver transplantation: a systematic review and meta-analysis.
Liver Transpl 10:817827
79. Liberati A, DAmico R, Pifferi S et al (2004) Antibiotic prophylaxis to reduce respiratory
tract infections and mortality in adults receiving intensive care (Cochrane review). In: The
Cochrane library, issue 1. Wiley, Chichester
80. Silvestri L, van Saene HKF, Milanese M, Gregori D (2005) Impact of selective
decontamination of the digestive tract on fungal carriage and infection: systematic review
of randomized controlled trials. Intensive Care Med 31:898910
81. Silvestri L, van Saene HKF, Milanese M et al (2007) Selective decontamination of the
digestive tract reduces bloodstream infections and mortality in critically ill patients: a
systematic review of randomized controlled trials. J Hosp Infect 65:187203
82. Silvestri L, van Saene HKF, Casarin AL et al (2008) Impact of selective decontamination of
the digestive tract on carriage and infection due to Gram-negative and Gram-positive
bacteria: systematic review of randomized controlled trials. Anaesths Intens Care 36:324338
83. Silvestri L, van Saene HKF, Weir I, Gullo A (2009) Survival benefit of the full selective
digestive decontamination regimen. J Crit Care 24:474.e7474.e14
84. Liberati A, DAmico R, Pifferi S (2009) Antibiotic prophylaxis to reduce respiratory tract
infections and mortality in adults receiving intensive care. Cochrane Database Syst Rev
4:CD000022
85. Silvestri L, van Saene HKF, Zandstra DF et al (2010) Selective decontamination of the
digestive tract reduces multiple organ failure and mortality in critically ill patients:
systematic review of randomized controlled trials. Crit Care Med 38:13701376
86. Silvestri L, van Saene HKF, Zandstra DF (2010) Selective digestive decontamination reduces
ventilator-associated tracheobronchitis. Respir Med 104:19531955
Index
H. K. F. van Saene et al. (eds.), Infection Control in the Intensive Care Unit, 507
DOI: 10.1007/978-88-470-1601-9, Springer-Verlag Italia 2012
508 Index
Diagnostic samples, 6, 16, 17, 43, 5153, 57, Exogenous, 3, 59, 11, 21, 23, 25, 29, 42, 43,
121, 186, 354, 357, 362, 366, 432 4547, 53, 5557, 59, 121, 123, 125,
Drainage, 11, 157159, 209, 220, 229, 236, 127, 170, 173, 176, 179, 183, 186, 188,
239, 241, 242, 246248, 251, 266, 191193, 205, 211, 275, 277, 356, 363,
268270, 310, 359361, 364, 382, 364, 367, 382, 385, 393, 394, 396, 411,
384387, 464, 466, 468, 469, 478 412, 429, 430, 438, 463, 474, 475
Extended infusion, 65
E
Echinocandins, 92, 94, 97, 99, 107111, F
114, 348 Faecaloral transmission
Efficacy, 9, 24, 64, 6668, 81, 89, 9496, Formulations, 113, 140, 302, 312, 397, 411,
111, 121, 123, 126, 134, 137, 415, 416, 418, 425
150, 151, 158, 162, 168170, Four-quadrant method, 53, 54
172, 173, 186, 191196, 199,
201, 209, 214, 215, 234, 245,
281, 284, 288, 292, 302, 312, G
320, 355, 357, 381, 383, 387, Ganciclovir, 296, 298, 303, 309, 312, 313,
396, 406, 411, 414, 419, 426, 343, 344
430, 432, 433, 435, 441, 443, Glutamine, 391, 396398, 400
445, 450, 455, 463, 474, 476, Grade, 50, 51, 55, 93, 94, 97, 111, 113, 180,
477, 479, 480 212, 236, 246, 312, 338, 367, 375,
Empirical antibiotic treatment, 230, 288 429431, 437, 438, 461464, 472, 474,
Empirical, 4, 5, 65, 72, 81, 92, 94, 99, 108, 477
112, 113, 115, 172174, 230, 240, 251, Gut overgrowth, 8, 13, 23, 26, 50, 51, 53, 55,
283, 284, 288291, 293, 294, 297, 306, 58, 117, 124, 126, 129, 184, 187, 190,
309, 354356, 359, 361, 367, 368 285, 413, 428431, 434, 435, 439, 444,
Endogenous, 3, 57, 9, 11, 17, 21, 29, 39, 474, 475
4247, 51, 53, 5557, 116, 118, 121,
123, 124, 127, 133, 168170, 173, 179,
186, 191193, 200, 205, 211, 275277, H
297, 300, 356, 362, 365, 370, 378, 385, Haart, 335, 336, 338, 340, 343,
406, 408, 411413, 429, 430, 435, 442, 346351
463, 472, 474, 475 Hand hygiene, 55, 126, 127, 137, 138, 140,
Endotracheal tube, 21, 23, 72, 136, 144, 141, 146, 155, 158, 191, 319, 323, 324,
150, 151, 153, 159, 160, 205, 332, 366, 380, 463, 465
210, 211, 214, 216, 277, 356, Hand washing, 6, 11, 111, 140, 225,
364, 366, 381, 384, 385, 387, 316, 317, 324, 325, 329,
388, 457, 468, 469, 478 365, 366, 463
Enteral feeding, 151, 208, 212, 270, 359, 386, Helicobacter pylori, 402, 404,
392, 394, 396, 399, 400, 402, 405407, 406, 409
457, 471 High-level pathogen, 6, 191, 275
Enteral nutrition, 18, 19, 23, 26, 129, 151, 156, HIV infection, 327, 334, 335, 338346, 348,
209, 210, 217, 245, 268270, 277, 285, 351353
375, 381, 382, 385, 395397, 399, 400, Hospital, 4, 6, 7, 13, 18, 19, 27, 29, 3134, 36,
409, 471 37, 3942, 47, 48, 57, 58, 69, 72, 83,
Eradication, 9, 24, 66, 117, 119, 121, 123, 90, 111, 115, 116, 125, 128, 129, 134,
171, 182, 184, 190, 197, 300, 136, 137, 139, 140, 143, 144, 146, 147,
324, 326, 359, 364, 365, 404, 149, 152, 154, 157, 164, 168, 172, 174,
418, 430 176, 179185, 188190, 197, 198, 204,
Evidence-based practice, 137 206, 208, 215, 216, 218220, 222225,
Exogenous infection, 57, 9, 42, 4547, 53, 227229, 231234, 236, 237, 258260,
5557, 123, 127, 173, 191, 193, 275, 263, 265, 270272, 276, 280, 287, 288,
277, 364, 385, 412, 430, 438, 463, 475 290, 291, 294, 296, 297, 299, 303, 312,
510 Index
317324, 328, 331, 332, 335, 337, 342, 253, 266268, 270, 279, 355, 356, 359,
343, 352, 355, 361, 377, 380, 383, 387, 362, 368, 372, 393
389393, 395, 397, 398, 411, 414, 416, Intrinsic pathogenicity index, 32, 48, 275,
418, 423427, 434, 435, 439, 444447, 285, 473
452, 455, 456, 458461, 463, 471, 474 Isolation, 6, 11, 12, 17, 28, 30, 31, 54, 56, 57,
112, 137, 139, 141, 142, 146, 147, 184,
220, 221, 233, 279, 287, 316, 317, 319,
I 323325, 328330, 346, 348, 350, 362,
IA abscess, 235, 253, 258 365, 463, 464, 477
Immunonutrients, 390, 391
Immunonutrition, 391, 399, 406, 409, 470,
471, 480 K
Immunosuppression, 8, 18, 19, 33, 42, 44, 51, Kaposis sarcoma, 334, 335, 345,
55, 56, 129, 260, 289, 297, 299, 303, 348350, 353
306, 309, 310, 322, 338, 342, 343, 346,
364, 390, 432
Import, 1, 2, 4, 18, 2022, 24, 25, 33, 37, 39, L
4245, 53, 55, 58, 63, 6769, 71, 77, Leadership, 145
85, 92, 98, 111, 116119, 125, 126, Lowest resistance potential, 163
128, 135, 140, 155, 168, 172, 179, 181, Low-level pathogen, 44, 128, 191, 241, 275,
184, 186, 193, 197, 198, 204, 205, 208, 362, 436
212, 215, 219, 221, 224227, 231, 234,
238, 245, 251, 254, 258, 260, 261, 264,
266, 273, 274, 278, 285, 299, 310, 314, M
319, 321, 323, 325, 330, 340, 342, 344, Macconkey agar, 54
354, 355, 364, 366, 370, 375, 376, Mediastinitis, 248251, 269, 270
382384, 386, 389392, 395, 397, 398, Meningitis, 4, 63, 68, 101, 104, 278, 329, 347,
401407, 413, 431, 432, 434, 438, 444, 355, 356, 361, 471
447, 448, 451, 453, 462, 463, 468, 472 MIC, 2, 13, 15, 18, 2045, 4773, 7585, 87,
Indigenous flora, 7, 17, 20, 21, 31, 32, 34, 118, 89102, 104, 106136, 138142,
119, 163, 164, 275, 366, 413, 144147, 151158, 160, 162176, 179,
431, 473 180, 182195, 197, 203, 205, 207, 211,
Infection control, 1, 7, 15, 24, 27, 39, 47, 48, 213216, 218226, 228230, 232238,
50, 57, 59, 61, 92, 113, 116, 128, 130, 240, 241, 243245, 249, 251254,
135, 136, 138, 140, 142, 144, 146149, 256267, 270, 272295, 299301, 303,
159, 161, 162, 177, 179, 180, 183, 305, 307, 309, 311323, 328335,
187189, 191, 204, 216, 218, 232, 235, 337342, 346, 348, 351, 352, 354380,
270, 272, 277, 284, 287, 296, 314, 316, 383387, 389, 391, 392, 394396, 398,
319, 320, 323, 327, 329, 331334, 354, 401, 403409, 411419, 423, 426436,
356, 370, 380, 381, 383, 388, 389, 401, 438440, 442445, 449460, 463, 464,
405, 411, 426428, 430, 446, 456, 468, 471, 473, 475481
461, 463 Morbidity, 7, 28, 72, 115, 118, 123, 128,
Infection, 129, 31, 3353, 5563, 65, 66, 69, 133, 147, 154, 172, 173, 179,
71, 72, 7578, 8083, 86, 89, 9092, 198, 208, 209, 218, 224, 242,
94101, 104, 108, 112116, 118, 250, 252, 257, 258, 263, 270,
121152, 154163, 165, 166, 168481 274, 281, 285, 288, 289, 293,
Inflammation, 2, 5, 6, 8, 12, 1618, 24, 28, 42, 294, 314, 321, 330, 340, 342,
51, 52, 55, 129, 165, 166, 168, 169, 347, 367, 376, 380, 384, 389391,
238, 241, 244, 256, 259, 277, 279, 339, 394396, 398, 401, 405, 412,
340, 344, 359, 362364, 366, 367, 370, 425, 428, 432, 440, 446448,
371, 375, 392, 402, 403, 405, 413, 432 450, 454, 456458, 460, 463465, 468,
Inoculum effect, 62, 63 472, 478
Intra-abdominal, 6, 89, 96, 97, 220, 222, 223, Mortality, 7, 13, 18, 28, 3335, 37, 40, 47, 53,
225, 231, 235, 236, 238, 239, 241, 242, 58, 60, 65, 72, 96, 99, 111, 112, 115,
511 Index