137
Views
1
CrossRef citations to date
0
Altmetric
Review

Optimizing antimicrobial therapy in critically ill patients

, , , , &
Pages 261-271 | Published online: 20 Oct 2014

Abstract

Critically ill patients with infection in the intensive care unit (ICU) would certainly benefit from timely bacterial identification and effective antimicrobial treatment. Diagnostic techniques have clearly improved in the last years and allow earlier identification of bacterial strains in some cases, but these techniques are still quite expensive and not readily available in all institutions. Moreover, the ever increasing rates of resistance to antimicrobials, especially in Gram-negative pathogens, are threatening the outcome for such patients because of the lack of effective medical treatment; ICU physicians are therefore resorting to combination therapies to overcome resistance, with the direct consequence of promoting further resistance. A more appropriate use of available antimicrobials in the ICU should be pursued, and adjustments in doses and dosing through pharmacokinetics and pharmacodynamics have recently shown promising results in improving outcomes and reducing antimicrobial resistance. The aim of multidisciplinary antimicrobial stewardship programs is to improve antimicrobial prescription, and in this review we analyze the available experiences of such programs carried out in ICUs, with emphasis on results, challenges, and pitfalls. Any effective intervention aimed at improving antibiotic usage in ICUs must be brought about at the present time; otherwise, we will face the challenge of intractable infections in critically ill patients in the near future.

Early diagnosis of infection: new tools

Effective antimicrobial administration within the first hour of documented hypotension is associated with increased survival in patients with septic shock,Citation1 whereas inappropriate empirical antimicrobial therapy has been associated with a five-fold reduction in survival.Citation2 Rapid and accurate identification of bacterial species in blood cultures is therefore warranted to improve the management of these patients.Citation3

Bacterial identification is routinely based initially on simple tests like Gram staining, catalase and oxidase tests. Subsequent phenotypic tests complete the identification. Although some of these tests are performed within minutes, complete identification is routinely achieved within 24 to 48 hours, but it may take several days for fastidious organisms. Blood cultures, which are the gold standard for the diagnosis of bloodstream infections, still need 48 to 72 hours for a complete identification.

However, two newer methods may allow an earlier identification of pathogens in patients with severe sepsis: 1) the use of protein profiles obtained by matrix-assisted laser desorption ionization time-of-flight mass spectrometry (MALDI-TOF-MS) that allows rapid and accurate identification of bacteria as well as fungi directly from colonies; and 2) molecular biology tools that enable rapid bacterial identification using real-time polymerase chain reaction (RT-PCR).

In this review, we will not discuss biomarkers for the diagnosis of invasive infection, as they have been extensively investigated and relevant reviews have already been published.Citation4,Citation5

MALDI-TOF-MS

Mass spectrometry is an analytical technique that produces spectra of the masses of the atoms or molecules constituting a sample of material. The principle of mass spectrometry is to detect the mass:charge ratio of a bioanalyte, providing its own specific spectrum. This method is used to profile microorganism proteins from cell extracts and allows identification of bacteria, yeasts, and filamentous fungi. The procedure provides a unique mass spectral fingerprint of the microorganisms. In practice, bacterial cells are spread across the well of a conductive metallic plate called “target”. Each specimen is then covered with an appropriate “matrix”, which creates a mixture with the analyte molecules. The target is then placed in the MALDI-TOF-MS machine, and brief laser pulses hit the mixture. The small desorbed and ionized molecules are accelerated through an electrostatic field, and drift through a field-free tunnel until they reach the mass spectrometer detector. Molecules of different masses and charges will fly at different speeds (“time-of-flight”). The result is a spectral signature, with specific spikes. This signature is then searched for in a database for the identification of the microorganism.Citation6

This method needs a minimal amount of labor compared with conventional methods,Citation7 and different studies confirm the excellent results obtained by this technology.Citation8Citation10 The time necessary for the identification in blood is less than 5 minutes,Citation7,Citation11 and this technique can be extended to other biological fluids such as cerebrospinal fluidCitation12 or urine.Citation13

The most widespread application of MALDI-TOF-MS is bacterial identification from bacterial colonies, and one main interest is the identification of anaerobes and other fastidious organisms, which are poorly identified by current phenotypic methods. However, a major current limitation is failure to accurately identify Streptococcus pneumoniae. Erroneous identifications were also obtained for some strains of Stenotrophomonas maltophilia, Propionibacterium acnes, and Shigella spp.Citation9 Moreover, when the infection is due to several bacterial species, only the most abundant germ detected by Gram staining is identified by MALDI-TOF-MS.Citation11 The difficulty in identifying polymicrobial cultures by this method underscores the importance of continued reliance on Gram stain and subcultures for definitive identification.

MALDI-TOF-MS is a rapid and precise method for identification of bacteria, compared to conventional phenotypic techniques. It is expected to become a widely used technique in routine clinical laboratories for bacterial identification, replacing other phenotypic techniques.

RT-PCR

RT-PCR has been developed in order to rapidly detect pathogens.Citation14 After a first step of extraction and purification of deoxyribonucleic acid (DNA), this method can detect several target pathogens. This promising technology is obviously of interest in order to quickly identify the pathogen of patients with severe sepsis or septic shock. RT-PCR combines amplification and detection of amplified products in a unique reaction. It is based on amplification of the 16S or 23S ribosomal ribonucleic acid gene, which is present in all bacteria. A positive detection is recorded if the fluorescent signal emitted by internal hybridization probes reaches the threshold; subsequently, a melting curve analysis proceeds to identify the species.Citation14

This technique is the most promising for the routine diagnosis of bloodstream infections in clinical microbiology laboratories because it is based on amplification of the internal transcribed spacer. This non-coding region of the ribosomal DNA is localized among highly conserved genes, shows a high level of heterogeneity among bacterial genera and species, and allows a high level of identification using a limited pool of slightly degenerated primers.Citation15

The obvious advantage of RT-PCR in the intensive care unit (ICU) is to obtain a result in whole blood quicker than conventional blood culture. This technology could give valuable information to the clinician in order to adapt antimicrobial therapy rapidly in the ICU.

However, RT-PCR technologies have a number of limitations which restrict their applicability. The sensitivity of universal RT-PCR is lower than that of many species-specific RT-PCRs. A major issue is the restricted panel of pathogens. In a meta-analysis conducted by Chang et al, who enrolled 34 studies with 6,012 patients with suspected sepsis, the tool showed a positive post-test probability of 80% but a negative post-test probability of 5% including bacteremia and fungi.Citation16

Another issue is the work time required for RT-PCR in the real-life setting. Although results are obtained within 6 hours, the technique requires a level of expertise that is not usually available around the clock. The time to the final result in clinical settings may therefore be significantly longer.Citation17 At this time, the delay in real-life settings makes the clinical usefulness of the RT-PCR test for rapid diagnosis questionable. Last, PCR detects DNA rather than living microorganisms. A positive RT-PCR signal in the presence of a negative blood culture can be challenging, making the results difficult to interpret.Citation14

New technologies such as MALDI-TOF-MS and RT-PCR have appeared over the last 10 years. MALDI-TOF-MS is a reliable tool and has become a widely used technique in routine clinical laboratories. RT-PCR is a promising tool, considering the ability to detect and identify pathogens without any previous culture. Nevertheless, RT-PCR remains expensive, has a limited panel of pathogens for the moment, and should be used paired with conventional blood culture. Prospective studies are warranted in order to assess specifically the benefits and drawbacks of these tools in the clinical management of patients with infection in the ICU.

The global threat of multi-drug resistant Gram-negative pathogens

The ICU population is highly susceptible to colonization and infection by pathogens with reduced antimicrobial susceptibility or resistance. In addition, the host response to infection may be significantly impaired by acute illness, altered immune function or underlying co-morbidities. Given the high rates of infection-related morbidity and mortality in patients with septic shock, prompt antimicrobial therapy along with infection source control and supportive care are important determinants of the clinical outcome.

ICU physicians are challenged by the threats of multi-drug resistant (MDR) organisms or extensively or pan- drug resistant (XDR) strains, especially among Enterobacteriaceae, Pseudomonas aeruginosa, and Acinetobacter baumannii.Citation18Citation20 In some countries or geo graphical areas, the epidemic spread of such strains endangers the possibility of curing critically ill patients with infection.Citation21 As a matter of fact, the increasing prevalence of MDR organisms within ICUs has caused physicians to broaden the spectrum of antimicrobials used, at least for empirical therapy, with the direct consequence of promoting the emergence of new resistance patterns.Citation22 However, resistance mechanisms are not superimposable from a clinical point of view, and knowledge of them should prompt ICU physicians to streamline treatment as soon as possible, targeting the causative pathogen with an effective drug with the least selection pressure on the environment.

Surveillance of local epidemiology is obviously of paramount importance in ICUs to monitor resistance rates and adapt empirical treatment accordingly. The main mechanisms conferring resistance to β-lactams among Enterobacteriaceae are alteration of the penicillin-binding protein; increased active efflux; and reduced or absent expression of outer membrane receptor and β-lactamase enzymes. These enzymes have within their active site either a serine group (serine-β-lactamases) or metallic ions (Zn2+) (metallo-β-lactamases [MBLs]), and these active site inclusions are the most important mechanism of β-lactam resistance in Enterobacteriaceae.

Extended-spectrum β-lactamases

Extended-spectrum β-lactamases (ESBLs) are molecular class A enzymes, and are able to hydrolyze all the oxymino-cephalosporins (cefotaxime, ceftriaxone, cefuroxime, ceftazidime, and cefepime) and monobactams, but not carbapenems and cephamycins (cefoxitin and cefotetan). TEM, SHV, and CTX-M types, among others, belong to this class of hydrolyzing enzymes.Citation23Citation25

This kind of enzyme is inactivated by β-lactamase inhibitors such as clavulanic acid, tazobactam, or sulbactam (whilst kinds of high-level cephalosporinases [AmpC] are not). The major importance of ESBLs resides in their ability to efficiently spread among Enterobacteriaceae through different transmission mechanisms, with epidemic diffusion not only in nosocomial strains, but also in the community.Citation26

Strains harboring ESBLs are frequently resistant to several antimicrobial classes (fluoroquinolones and aminoglycosides). Amoxicillin/clavulanic acid, piperacillin/tazobactam, or carbapenems (mostly ertapenem) should be considered the treatment of choice according to the site of infection.Citation23

Cephalosporinases (AmpC)

AmpC cephalosporinases are enzymes hydrolyzing penicillins, cephalosporins, and cephamycins, whereas cefepime and cefpirome are resistant to hydrolysis. These enzymes may be chromosomal or transferable, constitutive or inducible.Citation27 One important risk is represented by the fact that empirical treatment with third-generation cephalosporins induces derepression of AmpC during therapy, especially in Enterobacter spp., Serratia spp., Citrobacter spp., Morganella morganii, Proteus vulgaris, and Providencia spp., and these strains may develop complete resistance to cephalosporins within 3–4 days of treatment as a consequence.Citation28 Overall, AmpC cephalosporinases are able to inactivate penicillins (except temocillin), third-generation cephalosporins and cephamycins; show variable activity on aztreonam; and are inhibited by cefepimeCitation29 and cefpirome, by β-lactamase inhibitors (clavulanic acid, sulbactam, and tazobactam) and carbapenems. It has recently been shown that cefepime should be considered a drug of choice against pathogens carrying cephalosporinases, in order to limit the use of carbapenems and avoid consequent selection pressure.Citation29 Clinical relevance relies on the possible selection of constitutive mutants during therapy and the possibility of cross-resistance with other antimicrobial classes.

Carbapenemases

Carbapenem-resistant Enterobacteriaceae are increasingly prevalent in many parts of the world.Citation30,Citation31 It should be remarked that resistance to carbapenems is not always associated with the presence of carbapenemases; resistance to carbapenems, indeed, may be driven by two main mechanisms: 1) membrane impermeability and 2) carbapenemases. Impermeability yields to decreased susceptibility to carbapenems because of lack of porins in the outer membrane, leading to low-level resistance to carbapenems and higher minimum inhibitory concentration (MIC) only to ertapenem. Normally, MDR to other antimicrobial classes is atypical. Enterobacter cloacae is the primary carrier of this phenotype.Citation32 True carbapenemases, on the other hand, may be either serine-β-lactamases or MBLs, showing low to high level of resistance that often translates also into MDR (aminoglycosides, fluoroquinolo-nes), and may be detected in various strains of Enterobacteriaceae; they usually show a true increased MIC to imipenem (and ertapenem as well).

Most carbapenemase producers are almost completely resistant to β-lactam antibiotics, except those with OXA-48 alone, which remain susceptible to several cephalosporins.Citation33 Serine-carbapenemases belong to A or D molecular class; class As are inhibited by clavulanic acid and tazobactam, and therefore remain clinically susceptible to amoxicillin/clavulanic acid or piperacillin/tazobactam. Both may be chromosomal or plasmidic, or even inducible, and ertapenem is used to screen their presence, as it is the most sensitive carbapenem to these enzymes.Citation34 MBLs may be either chromosomal or plasmidic and are resistant to β-lactamase inhibitors, third- and fourth-generation cephalosporins and display elevated MIC to carbapenems; however, MBLs remain susceptible to aztreonam: monobactams are therefore the first-line treatment in case of infection sustained by MBL-producers.Citation34 Evidence of resistance to aztreonam implies that ESBLs or AmpCs are also present in the same strain.Citation35 All the main features of carbapenemases have been recently reviewed by Patel and Bonomo.Citation36

Klebsiella pneumoniae carbapenemase-producing Enterobacteriaceae

Since the beginning of 2000, K. pneumoniae carbapenemase-producing (KPC) Enterobacteriaceae have been increasingly detected in several regions worldwide and in some of them, such as in Israel and in Greece, they have become endemic.Citation37Citation39 Moreover, bla-KPC genes are easily transferable and are often linked with various non-β-lactam resistance determinants, further compromising the therapeutic alternatives for clinically significant infections. Clinical reports have already documented that hospital infections due to KPC are commonly associated with increasing therapeutic failureCitation40 and mortality.Citation41,Citation42

KPC enzymes confer various levels of resistance to all β-lactams, including carbapenems. However, optimized carbapenem dosing has been shown to be effective in overcoming resistance to some extent.Citation43 Concomitant aminoglycoside resistance is extensive but variable, as is resistance for multiple classes. Susceptibility testing data suggest that treatment of infections caused by KPC requires the use of tigecycline or colistin as last-resort drugs, often associated with carbapenem, fosfomycin, or rifampin.Citation40,Citation44 The ever-changing scenario about KPC and potential XDR-resistant Gram-negative pathogens, such as A. baumannii and P. aeruginosa, as well as their potential treatment options with different drug cocktails, will not herein be reviewed, as detailed reviews have been recently published.Citation44Citation49

The treatment in ICUs of acute infection from MDR germs and XDR germs needs a good understanding and knowledge of the local ecology. The prudent use of antibiotics, mainly those used as last-resort treatment, like carbapenems, is of utmost importance in order to prevent increasing pressure that may lead to the emergence of highly resistant strains. Furthermore, appropriate antimicrobial therapy must consider the significant pathophysiological changes associated with critical illness that may alter the pharmacokinetics (PK) (eg, increased volume of distribution [Vd], augmented clearance [CL]) and therefore dosing in this patient population (see the “Principles and practice of β-lactam pharmacokinetics/pharmacodynamics in ICUs” section). The development of new antibiotics effective against drug-resistant bacteria remains important, but optimized use of available ones based on local surveillance data and specific pharmacologic characteristics may allow improving clinical outcome and lessening selection pressure. However, any intervention in ICUs aimed at improving antimicrobial prescription practices should not leave aside infection control and prevention procedures, whose usefulness has been clearly established in containing the spread of antimicrobial resistance in such a critical setting.Citation50

Principles and practice of β-lactam pharmacokinetics/pharmacodynamics in ICUs

Although selecting the appropriate antimicrobial in terms of spectrum of activity is certainly the mainstay of antimicrobial therapy in critical ill patients, the choice of correct dose and dosing is also very important in ensuring clinical cure and microbiological eradication. β-lactam antibiotics are among the first-line therapies for critically ill patients, because of their large antimicrobial spectrum and low toxicity. β-lactams are time-dependent antimicrobials whose activity is mainly related to the duration of time the free drug level exceeds the pathogen MIC (T > MIC). A T > MIC of 100% of the dosage interval should be a theoretical target for β-lactams.Citation51Citation53 For carbapenems, which have a longer post-antibiotic effect, a bactericidal effect is observed for a T > MIC of 40%. Further improvement in efficacy has been observed when concentrations four- to five-fold greater than the MIC are achieved for prolonged time periods during each dosing interval (100% T >4–5× MIC).Citation54Citation56

T > MIC is dependent on drug half-life and serum concentration, which in turn depends on the dose delivered and its Vd. β-lactams are hydrophilic drugs with a low Vd, a low intracellular penetration, and predominant renal CL.Citation51 In septic patients, Vd may be increased because of a capillary leak syndrome, hypoalbuminemia, and therapeutic procedures (fluid replacement, mechanical ventilation, extracorporeal circuits, surgical drains).Citation57,Citation58 Increased Vd reduces drug concentration, but might increase the half-life if the CL remains unchanged.Citation59 Hypoalbuminemia increases the unbound fraction of the drug and consequently its Vd and CL.Citation60,Citation61

Renal CL of antibiotics depends on renal function. In septic patients without significant organ dysfunction, there is often an increased renal perfusion (massive fluid infusions, use of vasopressor agents) and consequently increased creatinine CL (CLCR) and elimination of hydrophilic antibiotics. The incidence of augmented renal CL (ARC) is high and varies between 30% and 85% depending on the studied population and the cut-off used for its definition.Citation62Citation65 In septic and trauma patients, ARC defined as a CLCR ≥ 130 mL/min/1.73 m2 was observed in 57.7% of the patients with a higher prevalence in trauma (85.7%) than in septic patients (39.5%). Young (≤50 years of age) trauma patients, without significant organ dysfunction (modified Sequential Organ Failure Assessment score ≤4) appear to be at greater risk of ARC. ARC appears to be an important predictor of subtherapeutic β-lactam concentrations. In the study by Udy et al,Citation64 CLCR values ≥130 mL/min/1.73 m2 were associated with β-lactam trough concentrations less than MIC in 82% and less than 4× MIC in 72% of cases, and multivariate modeling confirmed CLCR as a significant covariate for predicting low trough concentrations. Carlier et alCitation66 also found that ARC was associated with a higher risk of not attaining PK/pharmacodynamics (PD) targets even when administering β-lactams through extended infusion. In the study by Casu et al,Citation67 the proportion of patients with insufficient β-lactam concentrations progressively increased with the increasing of CLCR, reaching >50% when CLCR exceeded 120 mL/min. If β-lactam PK is significantly correlated with CLCR, β-lactam PK changes are not predicted by CLCR changes, and dosing adjustment could not be reliably adapted to changes in renal function alone. Moreover, in the critically ill patient there is no readily available method to measure accurately the glomerular filtration rateCitation68 and the derived estimates of glomerular filtration (Modification of Diet in Renal Disease and Cockcroft–Gault formulae) significantly underestimate the measured CLCR in patients with ARC.Citation69 Renal replacement therapies (RRTs) are very efficient in removing hydrophilic antibiotics, especially those with low protein binding. The amount of antibiotic eliminated will depend on the type and dose of RRT delivered, blood flow rate, filter material, and surface area.Citation70Citation72 As the loading dose mainly depends on the Vd and is unaffected by RRT, an increase of the initial dose may be required in critically ill patients.Citation73 Trotman et alCitation74 formulated recommendations for antibiotic dosing in critically ill patients receiving continuous RRT (CRRT) with an ultrafiltration rate of 1 L/h or a dialysate flow rate of 1 L/h and no residual renal function.Citation75 In the study of Roberts et al,Citation76 30.6% of patients receiving CRRT (dialysis flow rate of 1,000 mL/h and ultrafiltration rate of 2,000 mL/h) achieved target concentrations, 19.4% required a dose increase, and 50% a dose decrease. With empirical dosing, Roberts et alCitation77 reported a significant variability in β-lactam trough concentrations in patients receiving CRRT, with no correlation with the efflux flow rate (25 or 40 mL/kg/h). The lower therapeutic target (100% T > MIC) was achieved in 100% of patients, but the higher target (100% T >4× MIC) was achieved only in 76% of patients for meropenem and 86% for piperacillin.

Antimicrobial target concentration attainment in the infected tissue is also an important determinant of clinical outcome. The plasma concentration of unbound antibiotic is predictive of interstitial tissue fluid concentration,Citation52 but in critically ill patients, distribution of antibiotics in tissue may be substantially impaired, and a discrepancy between plasma and interstitial fluid level may occur.Citation78 In septic shock patients, piperacillin concentrations in the interstitium of soft tissues (skeletal muscle, subcutaneous fat tissue), as evaluated by microdialysis, have been found five- to ten-fold lower than free plasma concentrations, and several-fold lower than in a control group of healthy volunteers.Citation79 The tissue penetration of cefpirome is also significantly impaired in septic patients compared with that in healthy subjects.Citation80

β-lactams should be more effective when delivered by continuous infusion after a loading dose to reach a steady state more rapidly, or by extended infusion if the drug is unstable once reconstituted at room temperature.Citation81,Citation82 A recent study confirms that continuous administration of β-lactams in severe sepsis produces higher plasma and interstitial fluid antibiotic concentration than intermittent administration, with significant improvement in clinical cure.Citation83 The available evidence from mainly nonrandomized studies suggests also that extended or continuous infusion of carbapenem or piperacillin/tazobactam is associated with lower mortality (relative risk, 0.59; 95% confidence interval, 0.41–0.83), and this difference in mortality was higher in patients with pneumonia (relative risk, 0.50; 95% confidence interval, 0.26–0.96).Citation84

As mentioned above, PK properties of β-lactams in ICU patients may be profoundly altered due to the dynamic and unpredictable pathophysiological changes that occur in severe sepsis.Citation85 Therapeutic drug monitoring (TDM) may be useful to improve β-lactam dosing, as any assumptions about drug concentrations are unreliable, and dose–effect relationships are rather unpredictable in this setting. Targeting a 100% T >4–5× MIC attainment, Roberts et alCitation76 reported that dose adjustment was required in 175 (74.2%) of the ICU patients, 50.4% requiring dose increase after the first TDM, and 23.7% required dose decrease. In 92 ICU patients, Aubert et alCitation86 reported that the serum ceftazidime concentration was <5× MIC of the targeted pathogen in <15.7% of patients, and with a target of 40±10 mg/L (P. aeruginosa breakpoint MIC of 8 mg/L), the serum level was insufficient in 36.9% and excessive in 27.2% of patients. These studies support both the need for adjusting dosing and the major role of TDM in tailoring antimicrobial therapy in critically ill patients whenever possible. However, the positive impact of β-lactam TDM on clinical outcome remains to be assessed in randomized controlled clinical trials.

Usefulness and pitfalls of antimicrobial stewardship programs in ICUs

Antimicrobial stewardship programs (ASPs) are multidisciplinary programs whose primary aim is to optimize antibiotic use (improve clinical outcomes; minimize the untoward effects of antimicrobial use, and selection of resistant pathogens; and reduce ICU length of stay and costs). As detailed in the antimicrobial stewardship guidelines from the Infectious Diseases Society of America,Citation87 ASPs usually include several strategies: educational programs, implementation of guidelines, prospective audit and feedback, antibiotics formulary restriction (preauthorization), computer-assisted decision and prescription, PK/PD optimization, de-escalation, shortened antibiotic treatment, prevention of patient-to-patient transfer of resistant microorganisms, and intravenous-to-oral conversion. We will briefly review the main available studies on ASPs in ICUs, with their strategy and endpoints, and discuss pitfalls.

Several studies have demonstrated that ASPs consistently reduce antimicrobial use. Global reduction in antimicrobial consumption ranged from 22% to 36%.Citation88Citation90 Ng et alCitation91 showed that antibiotic restriction could reduce consumption of restricted antibiotic (by 47.2%) but interestingly, it also decreases consumption of non-restricted antibiotics (by 7.9%). This is an important point, as reduction in antibiotic prescription is correlated to reduction in antimicrobial resistance. Accordingly, Carling et al,Citation88 in a 7-year study, evaluated the impact of an interventional multidisciplinary ASPs on vancomycin-resistant enterococci (VRE), methicillin-resistant Staphylococcus aureus, and Clostridium difficile through minimization of third-generation cephalosporin use. They showed a 22% decrease (P<0.0001) of intravenous broad-spectrum antibiotics use and a significant decrease in nosocomial infections caused by C. difficile (P=0.002) or resistant Enterobacteriaceae (P=0.02). However, prevalence of VRE and S. aureus did not change significantly. Another pitfall of antibiotic restriction is that resistance decrease is often transitory, even with multimodal ASPs, as described by Slain et al.Citation92 They indeed evaluated the impact of a multimodal ASP on P. aeruginosa resistance and showed a decrease between a pre-2004 and post-2007 ASP period concerning intravenous ciprofloxacin and ceftazidime use, correlated with a significant decrease in ciprofloxacin-resistant P. aeruginosa prevalence. Unfortunately, in this study, the ciprofloxacin resistant rate increased to 47.6% in 2010, and it seems difficult to maintain prolonged low bacterial resistance rate.

ASP could also impact not only on quantity but also on quality of antimicrobial prescription and justification of antimicrobial regimen choice. Katsios et alCitation93 evaluated the antimicrobial treatment of positive clinically relevant culture in a mixed ICU over 2 months before and after ASP implementation. In the post-ASP period, they showed a significant increase in the treatment of sterile site cultures (64% pre-ASP versus [vs] 83% post-ASP, P=0.01), and a reduction in the treatment of non-sterile site cultures (which may represent colonization or contamination) (71% pre-ASP vs 46% post-ASP, P=0.0002). They also showed that ASPs improved documentation of antimicrobial use in the medical record (26% pre-ASP vs 71% post-ASP, P<0.0001). Moreover, strategies employed in this study did not use formulary restriction, and did preserve prescriber autonomy.

Other authors showed that antimicrobial restriction (formulary restriction with prior authorization) not only decreased bacterial resistance, but also improved patient outcome (length of stay), especially considering ICU patients. Gentry et alCitation94 developed a stewardship program which used the core strategy of formulary restriction with prior authorization (combined with protocol development and one-on-one education of physicians). Comparing pre- and post-ASP periods, they showed a significantly decreased length of stay, down from 13.2±15.3 to 10.8±12.7 days (P,0.0001), especially in the subgroup of ICU patients (from 15.0±14.4 days to 12.8±16.7 days, P=0.0004). Readmission rate within 30 days and mortality were not significantly affected. However, in the study by Rahal et al,Citation95 even though the restriction of cephalosporin use was associated with significant decrease in the development of resistant Klebsiella spp., imipenem/cilastatin use increased 141% during the study period and was accompanied by a 69% increase in the incidence of imipenem-resistant P. aeruginosa, as confirmed also by Tam et al.Citation22 Moreover, antibiotic restriction strategies with preau-thorization require availability of personnel to approve the use of the antimicrobial, which could lead to delayed treatment administration in critically ill patients with potential unintended consequences.

Only one randomized study (open-label monocentric study) evaluated the impact of broad-spectrum antibiotic followed with de-escalation vs no de-escalation.Citation96 There was neither statistical difference in length of stay, nor in 14-and 28-day mortality between the two groups. Interestingly, Garnacho-Montero et alCitation97 prospectively evaluated the impact on in-hospital mortality and 90-day mortality of de-escalation therapy in patients admitted to the ICU with severe sepsis or septic shock. De-escalation was applied in 219 patients (34.9%). De-escalation therapy was also a protective factor for 90-day mortality, even after a strict adjustment for confounding variables including baseline characteristics and severity of illness on the day of culture results.

As a matter of fact, no randomized controlled trials or well-done observational studies have assessed the clinical impact of de-escalation strategy in critically ill patients with severe sepsis or septic shock until recently. Indeed, observational studies that assessed de-escalation in episodes of hospital-acquired severe sepsis show that this strategy was accomplished in only 50% of the cases,Citation98 even in microbiologically confirmed episodes (bacteremia) where de-escalation occurred in 39% to 81% of cases.Citation99 All the relevant studies concerning ASPs in the ICU are summarized in .

Table 1 Summary of main studies on ASPs in ICUs

To limit the spread of antimicrobial resistance, practitioners should be aware of prevention of patient-to-patient transfer of resistant microorganisms. Indeed, it has been shown that 31% of cases of imipenem-resistant P. aeruginosa acquisition among patients in medical and surgical ICUs were due to patient-to-patient transfer of organisms, whereas only 19% of the cases were thought to be due to acquisition from the endogenous flora.Citation100 The combination of a comprehensive infection control strategy and an effective ASP may lead to the prevention of emergence and transmission of resistant pathogens. Hand hygiene promotion, barrier precautions, and environmental decontamination should be the cornerstones of this strategy.

To summarize, ASPs should promote the optimal use of antimicrobial therapy, leading to the best clinical outcome for patients. The relative paucity of outcome data demonstrating the benefits of antimicrobial stewardship is likely due to its infancy: according to George and Morris,Citation101 ASPs today are where infection control programs were roughly 30 years ago. ASPs should be multidisciplinary, taking advantage of expertise from intensivists, infectious disease specialists, microbiologists, and pharmacists, and new tools, such as PK/PD-driven dosing, should be the next step of ASPs in the ICU.

Conclusion

Optimizing antimicrobial therapy in critically ill patients with suspected or proven infections remains a challenge. Joint efforts by different professionals should concur to this aim. Ever-improving diagnostic techniques must be paralleled by the consciousness that any antimicrobial prescription today will impact on further prescriptions tomorrow, and that the extraordinary progress of ICU medicine should not be frustrated by the impossibility of treating infections in critically ill patients.

Disclosure

The authors report no conflicts of interest in this work.

References

  • KumarARobertsDWoodKEDuration of hypotension before initiation of effective antimicrobial therapy is the critical determinant of survival in human septic shockCrit Care Med20063461589159616625125
  • KumarAEllisPArabiYCooperative Antimicrobial Therapy of Septic Shock Database Research GroupInitiation of inappropriate antimicrobial therapy results in a fivefold reduction of survival in human septic shockChest200913651237124819696123
  • TrenholmeGMKaplanRLKarakusisPHClinical impact of rapid identification and susceptibility testing of bacterial blood culture isolatesJ Clin Microbiol1989276134213452473995
  • DupuyAMPhilippartFPéanYMaurice Rapin Institute Biomarkers GroupRole of biomarkers in the management of antibiotic therapy: an expert panel review: I – currently available biomarkers for clinical use in acute infectionsAnn Intensive Care2013312223837559
  • QuenotJPLuytCERocheNRole of biomarkers in the management of antibiotic therapy: an expert panel review II: clinical use of biomarkers for initiation or discontinuation of antibiotic therapyAnn Intensive Care2013312123830525
  • EmonetSShahHNCherkaouiASchrenzelJApplication and use of various mass spectrometry methods in clinical microbiologyClin Microbiol Infect201016111604161320969670
  • La ScolaBRaoultDDirect identification of bacteria in positive blood culture bottles by matrix-assisted laser desorption ionisation time-of-flight mass spectrometryPloS One2009411e804119946369
  • NevilleSALecordierAZiochosHUtility of matrix-assisted laser desorption ionization-time of flight mass spectrometry following introduction for routine laboratory bacterial identificationJ Clin Microbiol20114982980298421632894
  • SengPDrancourtMGourietFOngoing revolution in bacteriology: routine identification of bacteria by matrix-assisted laser desorption ionization time-of-flight mass spectrometryClin Infect Dis200949454355119583519
  • BlondiauxNGaillotOCourcolR-JMALDI-TOF mass spectrometry to identify clinical bacterial isolates: evaluation in a teaching hospital in LillePathol Biol (Paris)20105815557 French19892496
  • FerroniASuarezSBerettiJLReal-time identification of bacteria and Candida species in positive blood culture broths by matrix-assisted laser desorption ionization-time of flight mass spectrometryJ Clin Microbiol20104851542154820237092
  • Nyvang HartmeyerGKvistholm JensenABöcherSMass spectrometry: pneumococcal meningitis verified and Brucella species identified in less than half an hourScand J Infect Dis201042971671820429713
  • FerreiraLSánchez-JuanesFGonzález-AvilaMDirect identification of urinary tract pathogens from urine samples by matrix-assisted laser desorption ionization-time of flight mass spectrometryJ Clin Microbiol20104862110211520392910
  • ChangSSHsiehWHLiuTSMultiplex PCR system for rapid detection of pathogens in patients with presumed sepsis - a systemic review and meta-analysisPLoS One201385e6232323734173
  • ManciniNCarlettiSGhidoliNCicheroPBurioniRClementiMThe era of molecular and other non-culture-based methods in diagnosis of sepsisClin Microbiol Rev201023123525120065332
  • ChangSSHsiehWHLiuTSMultiplex PCR system for rapid detection of pathogens in patients with presumed sepsis – a systemic review and meta-analysisPloS One201385e6232323734173
  • DierkesCEhrensteinBSiebigSLindeHJReischlUSalzbergerBClinical impact of a commercially available multiplex PCR system for rapid detection of pathogens in patients with presumed sepsisBMC Infect Dis2009912619671147
  • HirschEBTamVHImpact of multidrug-resistant Pseudomonas aeruginosa infection on patient outcomesExpert Rev Pharmacoecon Outcomes Res201010444145120715920
  • ZarrilliRPournarasSGiannouliMTsakrisAGlobal evolution of multidrug-resistant Acinetobacter baumannii clonal lineagesInt J Antimicrob Agents2013411111923127486
  • FalagasMEMarakiSKarageorgopoulosDEKastorisACMavromanolakisESamonisGAntimicrobial susceptibility of multidrug-resistant (MDR) and extensively drug-resistant (XDR) Enterobacteriaceae isolates to fosfomycinInt J Antimicrob Agents201035324024320034765
  • KontopidouFGiamarellouHKaterelosPGroup for the Study of KPC-producing Klebsiella pneumoniae infections in intensive care unitsInfections caused by carbapenem-resistant Klebsiella pneumoniae among patients in intensive care units in Greece: a multi-centre study on clinical outcome and therapeutic optionsClin Microbiol Infect2014202O117O12323992130
  • TamVHHirschEBLascoTMGentryLOPalmerHRCorrelation of hospital carbapenem consumption and resistance trends in selected gram-negative bacteriaAnn Pharmacother2012467–81120112222764323
  • LivermoreDMDefining an extended-spectrum beta-lactamaseClin Microbiol Infect200814Suppl 131018154524
  • LivermoreDMCantonRGniadkowskiMCTX-M: changing the face of ESBLs in EuropeJ Antimicrob Chemother200759216517417158117
  • RossoliniGMD’AndreaMMMugnaioliCThe spread of CTX-M-type extended-spectrum beta-lactamasesClin Microbiol Infect200814Suppl 1334118154526
  • PatersonDLBonomoRAExtended-spectrum β-lactamases: a clinical updateClin Microbiol Rev200518465768616223952
  • BabicMHujerAMBonomoRAWhat’s new in antibiotic resistance? Focus on beta-lactamasesDrug Resist Updat20069314215616899402
  • HansonNDAmpC beta-lactamases: what do we need to know for the future?J Antimicrob Chemother20035212412775673
  • TammaPDGirdwoodSCTGopaulRThe use of cefepime for treating AmpC β-lactamase-producing EnterobacteriaceaeClin Infect Dis201357678178823759352
  • BrinkAJCoetzeeJCorcoranCEmergence of OXA-48 and OXA-181 carbapenemases among Enterobacteriaceae in South Africa and evidence of in vivo selection of colistin resistance as a consequence of selective decontamination of the gastrointestinal tractJ Clin Microbiol201351136937223152549
  • HirschEBChangKTLascoTMCaeiroJPTamVHEmergence of KPC-producing Klebsiella pneumoniae in TexasDiagn Microbiol Infect Dis201169223423521251574
  • CornagliaGAkovaMAmicosanteGESCMID Study Group for Antimicrobial Resistance Surveillance (ESGARS)Metallo-beta-lactamases as emerging resistance determinants in Gram-negative pathogens: open issuesInt J Antimicrob Agents200729438038817223319
  • Walther-RasmussenJHøibyNOXA-type carbapenemasesJ Antimicrob Chemother200657337338316446375
  • QueenanAMBushKCarbapenemases: the versatile beta-lactamasesClin Microbiol Rev200720344045817630334
  • Walther-RasmussenJHøibyNClass A carbapenemasesJ Antimicrob Chemother200760347048217595289
  • PatelGBonomoRA“Stormy waters ahead”: global emergence of carbapenemasesFront Microbiol201344823504089
  • NordmannPPoirelLThe difficult-to-control spread of carbapenemase producers in Enterobacteriaceae worldwideClin Microbiol Infect Epub6142014
  • Hidalgo-GrassCWarburgGTemperVKPC-9, a novel carbapenemase from clinical specimens in IsraelAntimicrob Agents Chemother201256116057605922964247
  • PournarasSProtonotariouEVoulgariEClonal spread of KPC-2 carbapenemase-producing Klebsiella pneumoniae strains in GreeceJ Antimicrob Chemother200964234835219525514
  • TumbarelloMVialePViscoliCPredictors of mortality in bloodstream infections caused by Klebsiella pneumoniae carbapenemase-producing K. pneumoniae: importance of combination therapyClin Infect Dis201255794395022752516
  • FalagasMETansarliGSKarageorgopoulosDEVardakasKZDeaths attributable to carbapenem-resistant Enterobacteriaceae infectionsEmerg Infect Dis20142071170117524959688
  • Papadimitriou-OlivgerisMMarangosMChristofidouMRisk factors for infection and predictors of mortality among patients with KPC-producing Klebsiella pneumoniae bloodstream infections in the intensive care unitScand J Infect Dis201446964264825017796
  • HoVPJenkinsSGAfanehCITurbendianHKNicolauDPBariePSUse of meropenem by continuous infusion to treat a patient with a Bla(kpc-2)-positive Klebsiella pneumoniae blood stream infectionSurg Infect (Larchmt)201112432532721859337
  • PetrosilloNGiannellaMLewisRVialePTreatment of carbapenem-resistant Klebsiella pneumoniae: the state of the artExpert Rev Anti Infect Ther201311215917723409822
  • Levy HaraGGouldIEndimianiADetection, treatment, and prevention of carbapenemase-producing Enterobacteriaceae: recommendations from an International Working GroupJ Chemother201325312914023783137
  • DjahmiNDunyach-RemyCPantelADekhilMSottoALavigneJPEpidemiology of carbapenemase-producing Enterobacteriaceae and Acinetobacter baumannii in Mediterranean countriesBioMed Res Int2014201430578424955354
  • StockIInfectious diseases caused by carbapenemase-producing Enterobacteriaceae – a particular challenge for antibacterial therapyMed Monatsschr Pharm2014375162172 German24908928
  • Munoz-PriceLSPoirelLBonomoRAClinical epidemiology of the global expansion of Klebsiella pneumoniae carbapenemasesLancet Infect Dis201313978579623969216
  • PaulMCarmeliYDurante-MangoniECombination therapy for carbapenem-resistant Gram-negative bacteriaJ Antimicrob Chemother20146992305230924872346
  • LandelleCMarimuthuKHarbarthSInfection control measures to decrease the burden of antimicrobial resistance in the critical care settingCurr Opin Crit Care201420549950625032821
  • TurnidgeJDThe pharmacodynamics of beta-lactamsClin Infect Dis199827110229675443
  • CraigWAPharmacokinetic/pharmacodynamic parameters: rationale for antibacterial dosing of mice and menClin Infect Dis19982611109455502
  • McKinnonPSPaladinoJASchentagJJEvaluation of area under the inhibitory curve (AUIC) and time above the minimum inhibitory concentration (T.MIC) as predictors of outcome for cefepime and ceftazidime in serious bacterial infectionsInt J Antimicrob Agents200831434535118313273
  • MoutonJWVinksAAContinuous infusion of beta-lactamsCurr Opin Crit Care200713559860617762242
  • MoutonJWden HollanderJGKilling of Pseudomonas aeruginosa during continuous and intermittent infusion of ceftazidime in an in vitro pharmacokinetic modelAntimicrob Agents Chemother19943859319368067772
  • RoosendaalRBakker-WoudenbergIAvan den Berghe-van RaffeMVink-van den BergJCMichelMFImpact of the duration of infection on the activity of ceftazidime, gentamicin and ciprofloxacin in Klebsiella pneumoniae pneumonia and septicemia in leukopenic ratsEur J Clin Microbiol Infect Dis19911012101910251839380
  • PeaFVialePFurlanutMAntimicrobial therapy in critically ill patientsClin Pharmacokinet200544101009103416176116
  • RobertsJAJoyntGMChoiGYSGomersallCDLipmanJHow to optimise antimicrobial prescriptions in the intensive care unit: principles of individualised dosing using pharmacokinetics and pharmacodynamicsInt J Antimicrob Agents201239318719222226651
  • MehrotraRDe GaudioRPalazzoMAntibiotic pharmacokinetic and pharmacodynamic considerations in critical illnessIntensive Care Med200430122145215615536528
  • RobertsJAPeaFLipmanJThe clinical relevance of plasma protein binding changesClin Pharmacokinet20135211823150213
  • UlldemolinsMRelloJThe relevance of drug volume of distribution in antibiotic dosingCurr Pharm Biotechnol201112121996200121554218
  • UdyAARobertsJABootsRJPatersonDLLipmanJAugmented renal clearance: implications for antibacterial dosing in the critically illClin Pharmacokinet201049111620000886
  • UdyAARobertsJAShorrAFBootsRJLipmanJAugmented renal clearance in septic and traumatized patients with normal plasma creatinine concentrations: identifying at-risk patientsCrit Care2013171R3523448570
  • UdyAAVargheseJMAltukroniMSubtherapeutic initial β-lactam concentrations in select critically ill patients: association between augmented renal clearance and low trough drug concentrationsChest20121421303922194591
  • CarlierMDe WaeleJJIdentifying patients at risk for augmented renal clearance in the ICU – limitations and challengesCrit Care201317213023672959
  • CarlierMCarretteSRobertsJAMeropenem and piperacillin/tazobactam prescribing in critically ill patients: does augmented renal clearance affect pharmacokinetic/pharmacodynamic target attainment when extended infusions are used?Crit Care2013173R8423642005
  • CasuGSHitesMJacobsFCan changes in renal function predict variations in β-lactam concentrations in septic patients?Int J Antimicrob Agents201342542242823993066
  • MolitorisBAMeasuring glomerular filtration rate in the intensive care unit: no substitutes pleaseCrit Care201317518124004539
  • BaptistaJPUdyAASousaEA comparison of estimates of glomerular filtration in critically ill patients with augmented renal clearanceCrit Care2011153R13921651804
  • EylerRFMuellerBAMedscapeAntibiotic dosing in critically ill patients with acute kidney injuryNat Rev Nephrol20117422623521343897
  • FissellWHAntimicrobial dosing in acute renal replacementAdv Chronic Kidney Dis2013201859323265600
  • JamalJAEconomouCJLipmanJRobertsJAImproving antibiotic dosing in special situations in the ICU: burns, renal replacement therapy and extracorporeal membrane oxygenationCurr Opin Crit Care201218546047122820155
  • BoumanCSAntimicrobial dosing strategies in critically ill patients with acute kidney injury and high-dose continuous veno-venous hemofiltrationCurr Opin Crit Care200814665465919023912
  • TrotmanRLWilliamsonJCShoemakerDMSalzerWLAntibiotic dosing in critically ill adult patients receiving continuous renal replacement therapyClin Infect Dis20054181159116616163635
  • SeylerLCottonFTacconeFSRecommended β-lactam regimens are inadequate in septic patients treated with continuous renal replacement therapyCrit Care2011153R13721649882
  • RobertsJAUlldemolinsMRobertsMSTherapeutic drug monitoring of beta-lactams in critically ill patients: proof of conceptInt J Antimicrob Agents201036433233920685085
  • RobertsDMRobertsJARobertsMSRENAL Replacement Therapy Study InvestigatorsVariability of antibiotic concentrations in critically ill patients receiving continuous renal replacement therapy: a multicentre pharmacokinetic studyCrit Care Med20124051523152822511133
  • MüllerMdela PeñaADerendorfHIssues in pharmacokinetics and pharmacodynamics of anti-infective agents: distribution in tissueAntimicrob Agents Chemother20044851441145315105091
  • JoukhadarCFrossardMMayerBXImpaired target site penetration of beta-lactams may account for therapeutic failure in patients with septic shockCrit Care Med200129238539111246321
  • SauermannRDelle-KarthGMarsikCPharmacokinetics and pharmacodynamics of cefpirome in subcutaneous adipose tissue of septic patientsAntimicrob Agents Chemother200549265065515673747
  • Van HerendaelBJeurissenATulkensPMContinuous infusion of antibiotics in the critically ill: the new holy grail for beta-lactams and vancomycin?Ann Intensive Care2012212222747633
  • Abdul-AzizMHDulhuntyJMBellomoRLipmanJRobertsJAContinuous beta-lactam infusion in critically ill patients: the clinical evidenceAnn Intensive Care2012213722898246
  • DulhuntyJMRobertsJADavisJSContinuous infusion of beta-lactam antibiotics in severe sepsis: a multicenter double-blind, randomized controlled trialClin Infect Dis201356223624423074313
  • FalagasMETansarliGSIkawaKVardakasKZClinical outcomes with extended or continuous versus short-term intravenous infusion of carbapenems and piperacillin/tazobactam: a systematic review and meta-analysisClin Infect Dis201356227228223074314
  • Gonçalves-PereiraJPóvoaPAntibiotics in critically ill patients: a systematic review of the pharmacokinetics of β-lactamsCrit Care2011155R20621914174
  • AubertGCarricajoACoudrotMGuyomarc’hSAuboyerCZeniFProspective determination of serum ceftazidime concentrations in intensive care unitsTher Drug Monit201032451751920571462
  • DellitTHOwensRCMcGowanJEInfectious Diseases Society of America and the Society for Healthcare Epidemiology of America guidelines for developing an institutional program to enhance antimicrobial stewardshipClin Infect Dis200744215917717173212
  • CarlingPFungTKillionATerrinNBarzaMFavorable impact of a multidisciplinary antibiotic management program conducted during 7 yearsInfect Control Hosp Epidemiol200324969970614510254
  • LaRoccoAJrConcurrent antibiotic review programs – a role for infectious diseases specialists at small community hospitalsClin Infect Dis200337574274312942418
  • RüttimannSKeckBHartmeierCMaetzelABucherHCLong-term antibiotic cost savings from a comprehensive intervention program in a medical department of a university-affiliated teaching hospitalClin Infect Dis200438334835614727204
  • NgCKWuTCChanWMClinical and economic impact of an antibiotics stewardship programme in a regional hospital in Hong KongQual Saf Health Care200817538739218842981
  • SlainDSarwariARPetrosKOImpact of a multimodal antimicrobial stewardship program on Pseudomonas aeruginosa susceptibility and antimicrobial use in the intensive care unit settingCrit Care Res Pract2011201141642621687626
  • KatsiosCMBurryLNelsonSAn antimicrobial stewardship program improves antimicrobial treatment by culture site and the quality of antimicrobial prescribing in critically ill patientsCrit Care201216R21623127353
  • GentryCAGreenfieldRASlaterLNWackMHuyckeMMOutcomes of an antimicrobial control program in a teaching hospitalAm J Health Syst Pharm200057326827410674779
  • RahalJJUrbanCHornDClass restriction of cephalosporin use to control total cephalosporin resistance in nosocomial KlebsiellaJAMA199828014123312379786372
  • KimJWChungJChoiSHEarly use of imipenem/cilastatin and vancomycin followed by de-escalation versus conventional antimicrobials without de-escalation for patients with hospital-acquired pneumonia in a medical ICU: a randomized clinical trialCrit Care2012161R2822336530
  • Garnacho-MonteroJGutiérrez-PizarrayaAEscoresca-OrtegaADe-escalation of empirical therapy is associated with lower mortality in patients with severe sepsis and septic shockIntensive Care Med2014401324024026297
  • Gomes SilvaBNAndrioloRBAtallahANSalomãoRDe-escalation of antimicrobial treatment for adults with sepsis, severe sepsis or septic shock [review]Cochrane Database Syst Rev201012CD00793421154391
  • ShimeNSatakeSFujitaNDe-escalation of antimicrobials in the treatment of bacteraemia due to antibiotic-sensitive pathogens in immunocompetent patientsInfection201139431932521509424
  • JohnsonJKSmithGLeeMSThe role of patient-to-patient transmission in the acquisition of imipenem-resistant Pseudomonas aeruginosa colonization in the intensive care unitJ Infect Dis2009200690090519673646
  • GeorgePMorrisAMPro/con debate: should antimicrobial stewardship programs be adopted universally in the intensive care unit?Crit Care201014120520236505
  • RimawiRHMazerMASirajDSGoochMCookPPImpact of regular collaboration between infectious diseases and critical care practitioners on antimicrobial utilization and patient outcomeCrit Care Med20134192099210723873275
  • JainRKralovicSMEvansMEVeterans Affairs initiative to prevent methicillin-resistant Staphylococcus aureus infectionsN Engl J Med2011364151419143021488764
  • HuskinsWCHuckabeeCMO’GradyNPSTAR*ICU Trial InvestigatorsIntervention to reduce transmission of resistant bacteria in intensive careN Engl J Med2011364151407141821488763
  • HuangAMNewtonDKunapuliAImpact of rapid organism identification via matrix-assisted laser desorption/ionization time-of-flight combined with antimicrobial stewardship team intervention in adult patients with bacteremia and candidemiaClin Infect Dis20135791237124523899684