Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Dec 1.
Published in final edited form as: Transpl Infect Dis. 2017 Oct 25;19(6):10.1111/tid.12762. doi: 10.1111/tid.12762

Multidrug-resistant Enterobacteriaceae, Pseudomonas aeruginosa, and vancomycin-resistant enterococci: Three major threats to hematopoietic stem cell transplant recipients

Michael J Satlin 1,2, Thomas J Walsh 1,2,3
PMCID: PMC5745272  NIHMSID: NIHMS900313  PMID: 28815897

Abstract

Hematopoietic stem cell transplant (HSCT) recipients are uniquely threatened by the emergence of multidrug-resistant (MDR) bacteria because these patients rely on immediate active antimicrobial therapy to combat bacterial infections. This review describes the epidemiology and treatment considerations for three challenging MDR bacterial pathogens in HSCT recipients: MDR Enterobacteriaceae, including extended-spectrum β-lactamase (ESBL)-producing and carbapenem-resistant Enterobacteriaceae (CRE), Pseudomonas aeruginosa, and vancomycin-resistant Enterococcus (VRE). These bacteria are common causes of infection in this population and bacteremias caused by these organisms are associated with high mortality rates. Carbapenems remain the treatments of choice for serious infections due to ESBL-producing Enterobacteriaceae in HSCT recipients. Administration of β-lactam agents as an extended infusion is associated with improved outcomes in patients with severe infections caused by P. aeruginosa. Older agents used for the treatment of CRE and MDR P. aeruginosa infections, such as polymyxins and aminoglycosides, have major limitations. Newer agents, such as ceftazidime-avibactam and ceftolozane-tazobactam have great potential for the treatment of Klebsiella pneumoniae carbapemenase-producing CRE and MDR P. aeruginosa, respectively, but more pre-clinical and clinical data are needed to better evaluate their efficacy. Daptomycin dosages ≥ 8 mg/kg/day are recommended to treat VRE infections in this population, particularly in the setting of increasing daptomycin resistance. Strategies to prevent these infections include strict adherence to recommended infection control practices and multidisciplinary antimicrobial stewardship. Lastly, gastrointestinal screening to guide empirical therapy and the use of polymerase chain reaction-based rapid diagnostics may decrease the time to administration of appropriate therapy for these infections, thereby leading to improved outcomes.

Keywords: carbapenemases, Enterobacteriaceae, extended-spectrum β-lactamases, hematopoietic stem cell transplantation Pseudomonas aeruginosa, vancomycin-resistant enterococci

1 INTRODUCTION

Hematopoietic stem cell transplant (HSCT) recipients are at high risk of bacteremia early after transplantation because of two major insults to their innate immune system. First, these patients have prolonged neutropenia after receipt of their conditioning regimen and thus lack the first and most important phagocytes to combat bacterial infections.1,2 Second, their conditioning regimen leads to marked gastrointestinal (GI) mucositis, and thus the integrity of their mucosal barrier is damaged. These two key insults establish a high-risk setting for bacteremia caused by enteric organisms and for severe complications from these infections.

Before the 1960s, empirical antibacterial therapy was not routinely administered to febrile neutropenic patients. This strategy of delaying therapy until culture results were available was associated with poor outcomes in neutropenic patients with gram-negative bacteremia. For example, bacteremia caused by Pseudomonas aeruginosa carried a 60%–80% mortality rate in this era.3,4 These outcomes improved dramatically after a critical change in practice to administer antibacterial agents with in vitro activity against prominent gram-negative bacteria to neutropenic patients immediately for fever or at the first sign of infection.46

Current recommendations for the management of fever and neutropenia in oncology patients are to administer antimicrobial agents that are active against the three most common causes of gram-negative bacteremia in neutropenic patients: Escherichia coli, Klebsiella pneumoniae, and P. aeruginosa.79 Recommended agents include ceftazidime, cefepime, piperacillin-tazobactam, or a carbapenem as monotherapy, based on results from randomized clinical trials that were conducted over a decade ago.1014 However, over the last decade, multidrug-resistant (MDR) Enterobacteriaceae and P. aeruginosa have become increasingly prevalent among hospitalized patients.15,16 Furthermore, vancomycin-resistant Enterococcus has increasingly been reported as a common cause of bacteremia in HSCT recipients.17,18 This increase in resistance among the most common causes of bacteremia in neutropenic patients and HSCT recipients threatens the applicability of currently used algorithms.

The objective of this review is to delineate the current epidemiology and treatment considerations for three of the most challenging bacterial pathogens in HSCT recipients: MDR Enterobacteriaceae, including extended-spectrum β-lactamase (ESBL) and carbapenemase producers, P. aeruginosa, and vancomycin-resistant Enterococcus (VRE). We also discuss strategies to curb the threat posed by these pathogens, including an evaluation of preventative measures and strategies to minimize delays in administration of appropriate therapies for infected patients.

2 ESBL-PRODUCING ENTEROBACTERIACEAE (ESBL-E)

2.1 Epidemiology

ESBLs are enzymes that are capable of hydrolyzing penicillins, monobactams, and extended-spectrum cephalosporins.19 Multiple reports from oncology centers in North America, Europe, and Asia have identified ESBL prevalence rates of 17%–37% among bloodstream isolates of Enterobacteriaceae from patients with hematologic malignancies (Table 1 9,2026). Not only did these reports demonstrate a high prevalence of ESBL-E in this population, but many also reported higher mortality rates associated with ESBL-E bacteremia compared to bacteremia caused by non-ESBL-producing Enterobacteriaceae. For example, in a study of 14 Italian hematologic oncology centers, the 21-day mortality rate was 26% after bacteremia caused by to third-generation cephalosporin-resistant Enterobacteriaceae, most of which were likely ESBL-E, compared to 5% after bacteremia caused by third-generation cephalosporin-susceptible Enterobacteriaceae.9

TABLE 1.

Prevalence and characteristics of bacteremia due to ESBL-producing Enterobacteriaceae in patients with hematologic malignancies and HSCT recipients.

First author Reference Country Years Organisms All patients with hematologic malignancies: % of isolates that were ESBL producers HSCT recipients only: % of isolates that were ESBL producers Mortality of ESBL BSI vs non-ESBL BSI Mortality type
Mikulska (20) Italy 2004–2007 Escherichia coli
Klebsiella pneumoniae
HSCT only 41% (13/32) NR NR
Guidol (21) Spain 2006–2008 E. coli 17% (13/76) 9% (1/11) 38% vs. 7%1 30-day
Cornejo-Juarez (22) Mexico 2004–2009 E. coli 23% (205/670) NR 34% vs. 19%1 Overall
Kang (23) Korea 2008–2009 E. coli
K. pneumoniae
33% (37/156) NR 45% vs. 14%1 30-day
Ha (24) Korea 2010–2012 E. coli 32% (42/130) 44% (15/34) 14% vs. 10% 30-day
Kim (25) Korea 2007–2008 E. coli K. pneumoniae 26% (26/101) 38% (8/21) 15% vs. 5% 30-day
Trecarichi (9) Italy 2009–2012 Enterobacteriaceae 37% (98/265)2 NR 26% vs. 5%1 21-day
Yemişen (26) Turkey 2000–2011 Enterobacteriaceae HSCT only 21% (5/22) NR NR
1

P value <.05 for this comparison.

2

Third-generation cephalosporin resistance was used as a surrogate for ESBL production in this study.

HSCT, hematopoietic stem cell transplantation; ESBL, extended-spectrum beta-lacatamse; BSI, bloodstream infection; NR, not reported.

Furthermore, ESBL-E have expanded their host range into HSCT recipients. In a report from Korea, 44% of E. coli bacteremias in HSCT recipients were ESBL producers and having an HSCT was a risk factor for ESBL-E bacteremia.24 HSCT centers from Italy and Turkey reported that 18% and 23%, respectively, of E. coli bloodstream isolates and 86% and 22%, respectively, of K. pneumoniae bloodstream isolates were ESBL-producing strains.20,26 At our own HSCT center in New York City (NYC), 29 of 89 (33%) E. coli bloodstream isolates from 2011–2016 had a phenotypic profile suggestive of ESBL production (ceftriaxone resistant, meropenem susceptible).

2.2 Treatment

The ESBL-E test highly susceptible to carbapenems and these agents are the treatments of choice for serious ESBL-E infections in HSCT recipients.19 ESBL-E have variable susceptibility to other first-line recommended therapies for fever and neutropenia, with susceptibility rates to ceftazidime, cefepime, and piperacillin-tazobactam of 20%–30%, 60%–70% and 70%–95%, respectively.27,28 However, even when ESBL-E test susceptible to cefepime or piperacillin-tazobactam, concerns have been raised about their use for treating invasive ESBL-E infections. One concern with cefepime and piperacillin-tazobactam is the “inoculum effect”, whereby these agents have increases in their minimum inhibitory concentrations (MICs) against ESBL-E when higher inocula are used for testing.29,30 Thus, many ESBL-E that are reported as susceptible based on a standard inoculum would test resistant if a higher inoculum were used. This inoculum effect is typically not seen with carbapenems.

Clinical data characterizing the use of cefepime for the treatment of ESBL-E bacteremia substantiate these in vitro concerns. Three cohort studies comparing cefepime to carbapenems for the treatment of ESBL-E bacteremia demonstrated increased mortality with cefepime, including in multivariable and propensity-score matched analyses.3133 Thus, cefepime should be avoided for the treatment of ESBL-E infections in HSCT recipients.

The role of piperacillin-tazobactam for the treatment of ESBL-E infections is unclear, as no randomized clinical trials comparing piperacillin-tazobactam to carbapenems have been completed. Two large multicenter observational studies of patients with ESBL-E bacteremia found that outcomes of patients treated empirically or as targeted therapy with β-lactam/β-lactamase inhibitors were similar to those of patients treated with a carbapenem in multivariable analyses.34,35 However, a large single-center cohort study from the United States (U.S.) of patients with ESBL-E bacteremia demonstrated a two-fold increased risk of death in patients who received piperacillin-tazobactam empirically compared to those who received a carbapenem empirically, and this difference persisted in multivariable and propensity score-adjusted analyses.36 In the first two studies demonstrating the equivalence of piperacillin-tazobactam and carbapenems, two-thirds of the bacteremias originated from the urinary or biliary tracts. Piperacillin-tazobactam may be more effective in these settings because it achieves high urinary and biliary tract concentrations and biliary infections are often treated concurrently with biliary tract decompression.37 Furthermore, piperacillin-tazobactam was administered at dosages of 4.5 g every 6 hours or as a prolonged infusion in these studies. Conversely, in the study demonstrating the superiority of carbapenems, the source of the majority of bacteremias was central line-associated or intra-abdominal, and the most common dosage of piperacillin-tazobactam was 3.375 g every 6 hours. In addition, very few neutropenic patients or HSCT recipients were included in these studies. In summarizing results from these studies, it could be concluded that piperacillin-tazobactam, dosed at either 4.5 g every 6 hours or as a prolonged infusion, is appropriate for ESBL-E bacteremias that originate from the urinary or biliary tract in immunocompetent hosts, but for ESBL-E bacteremias from other sources, or bacteremias that occur in neutropenic patients or HSCT recipients, carbapenems remain the preferred treatment options.

Group 2 carbapenems (e.g., imipenem and meropenem) are typically utilized for the initial treatment of ESBL-E infections in HSCT recipients. Ertapenem, a group 1 carbapenem, has the advantage of once-daily dosing and has a narrower antimicrobial spectrum; unlike group 2 carbapenems, it does not have activity against P. aeruginosa and Acinetobacter baumannii.38 Unlike group 2 carbapenems, which are almost uniformly active in vitro against ESBL-E, up to 10% of ESBL-E will not test susceptible to ertapenem using current Clinical and Laboratory Standards Institute (CLSI) breakpoints.27,39 Provided that the organism is susceptible to ertapenem, outcomes of patients with ESBL-E infections treated with ertapenem are comparable to those of patients treated with group 2 carbapenems.40,41 Thus, HSCT recipients with infections caused by ESBL-E can potentially be transitioned from a group 2 carbapenem to ertapenem once clinically stable, provided that they have no other bacterial infections and that the organism tests susceptible in vitro.

3 CARBAPENEM-RESISTANT ENTEROBACTERIACEAE (CRE)

3.1 Epidemiology

Over the last decade, the prevalence has had an unprecedented increase in Enterobacteriaceae that are not only resistant to penicillins and cephalosporins, but also are resistant to carbapenems.42 In the U.S., CRE were initially reported in the NYC area,43,44 but have now been identified in almost every state and are considered an urgent antibiotic resistance threat by the Centers for Disease Control and Prevention (CDC).45,46 CRE have become most common in K. pneumoniae. For example, the percentage of carbapenem resistance among U.S. K. pneumoniae isolates causing central line-associated bloodstream infection (BSI) increased from <1% in 2000, to 8% in 2006–2007, to 12% in 2009–2010, to 13% in 2012–2013.47 CRE have also become endemic in many countries in South America, Europe, Africa, and Asia, and thus pose an international public health threat.48

This global emergence of CRE has been caused by the spread of plasmid-mediated enzymes that hydrolyze carbapenems (carbapenemases) and essentially all other β-lactamases.49 Furthermore, these enzymes are not inhibited by traditional β-lactamase inhibitors. Thus, CRE typically test resistant to all first-line agents for fever and neutropenia, such as ceftazidime, cefepime, piperacillin-tazobactam, and carbapenems.7 K. pneumoniae carbapenemase (KPC) is the predominant carbapenemase among Enterobacteriaceae in the U.S. and in many parts of the world,50 although other enzymes predominate in other regions (Table 246).

TABLE 2.

Characteristics of prominent carbapenemases among carbapenem-resistant Enterobacteriaceae (CRE).

Carbapenemase Ambler Molecular Class Requirement for Enzymatic Activity # of U.S. states reporting CRE with this enzyme1 Global Distribution Inhibited by Avibactam
KPC A Serine 49 U.S., Colombia, Brazil, Argentina, Italy, Greece, Israel, China Yes
NDM B Zinc 25 India, Pakistan, Bangladesh, United Kingdom No
VIM B Zinc 7 Greece, Italy, Spain No
IMP B Zinc 5 Japan, Taiwan No
OXA-48-types (OXA-48, OXA-181, OXA-232) D Serine 19 Turkey, Spain, Morocco, Algeria, Tunisia, India Yes
1

As of April 2016 (46).

KPC, Klebsiella pneumoniae carbapenemase; NDM, New Delhi metallo-β-lactamase; VIM, Verona integron-encoded metallo-β-lactamase.

Although the global threat of CRE has been well documented, the emergence of CRE in patients with hematologic malignancies and HSCT recipients has only recently been described.49,51 Publications reporting at least 10 HSCT recipients with CRE infection are outlined in Table 3.5256 Carbapenem-resistant K. pneumoniae (CRKP) has become a major cause of bacteremia in this patient population in Italy. In a prospective cohort study of 13 Italian hematologic oncology centers from 2010–2014, 161 (58%) of 278 episodes of K. pneumoniae bloodstream isolates were carbapenem-resistant.52 In a study of 52 Italian HSCT centers, the overall incidence of post-transplant CRKP infection from 2010–2013 was 0.4% after an autologous transplant and 2% after an allogeneic transplant.54 For comparison, the 12-month cumulative incidence of aspergillosis and candidiasis in HSCT recipients in the Transplant-Associated Infection Surveillance Network was 1.6% and 1.1%, respectively.57 CRE is also an established pathogen in hematologic oncology centers in NYC. Our center reported that CRE bacteremia occurred in 1.8% of HSCT recipients within 12 months after transplantation.51 In a study of BSIs in neutropenic patients with hematologic malignancies at two NYC oncology centers, 43 (2.2%) of 1992 BSI episodes were caused by CRE, CRE caused 4.7% of gram-negative bacteremias, and 18% of K. pneumoniae bacteremias were carbapenem-resistant.55

TABLE 3.

Carbapenem-resistant Enterobacteriaceae (CRE) infections in patients with hematologic malignancies and hematopoietic stem cell transplant (HSCT) recipients

First author Reference Location Patients HSCT recipients Infection type Organisms Mortality rate CRE bacteremia-related mortality rate
Trecarichi (52) Italy: 13 centers 161 26 Bacteremia Klebsiella pneumoniae 52% (21-day) NR
Tofas (53) Greece: 4 centers 50 NR Bacteremia K. pneumoniae 50% (14-day) 50%
Girmenia (54) Italy: 45 centers 112 112 Bacteremia (n=100)
Pneumonia (n=12)
K. pneumoniae 58% (90-day) 54%
Satlin (55) New York City, USA 43 18 Bacteremia K. pneumoniae (n=30)
Enterobacter cloacae (n=8)
Others (n=5)
53% (30-day) 51%
Hussein (56) Israel 28 18 Bacteremia K. pneumoniae NR NR

NR, not reported.

Not only are CRE emerging causes of bacteremia in HSCT recipients, but also CRE bacteremia is associated with high mortality rates in this population. In fact, both overall and CRE bacteremia-related mortality rates in patients with hematologic malignancies and HSCT recipients are consistently >50% (Table 3). Trecarichi et al.52 found that the 21-day mortality rate in patients with hematologic malignancies was 53% after CRKP bacteremia, compared to 15% after carbapenem-susceptible K. pneumoniae bacteremia, and carbapenem resistance was independently associated with mortality. A CRKP infection-related mortality rate of 64% was reported in allogeneic HSCT (allo-HSCT) recipients.54

A likely explanation for these high mortality rates is that CRE are resistant to recommended empirical therapies for fever and neutropenia in patients with cancer,7 and thus appropriate CRE-active therapy is delayed until the availability of organism identification and antimicrobial susceptibility testing (AST) results. In HSCT recipients with CRKP infection, CRKP-targeted empirical therapy has been associated with a nearly three-fold increase in survival in multivariable analysis.54 In our study, 86% of patients with CRE bacteremia did not receive empirical treatment with antimicrobial agents that had in vitro activity against their CRE bloodstream isolate and 21% died before AST results became available.55 Those who survived to availability of AST results had a median of 52 hours between blood culture collection and active therapy. Only one of six patients who received CRE-active empirical therapy died within 30 days, compared to 22 of 37 (59%) patients who did not receive CRE-active empirical therapy. These data suggest that CRE-active empirical therapy should be considered in HSCT recipients at high risk of CRE infection, such as a neutropenic patient with known CRE colonization or who is at a center where CRE are exceptionally common bloodstream pathogens.

3.2 Treatment

Treatment options for CRE infections are extremely limited, as in addition to being resistant to β-lactam agents, CRE are also typically resistant to fluoroquinolones and trimethoprim-sulfamethoxazole.49 Antimicrobial agents that have typically retained activity against CRE are outlined in Table 4.44,5871 Unfortunately, these options all have significant shortcomings. Out of necessity, polymyxins have been the mainstay of therapy for CRE infections. However, polymyxins have high rates of nephrotoxicity,58 they are historically less effective than β-lactam agents for the treatment of gram-negative bacteremia in oncology patients,72 and both chromosomally-mediated and plasmid-mediated polymyxin resistance is increasingly identified among CRE.73,74 Tigecycline is not bactericidal, achieves low bloodstream concentrations, and its use is associated with increased mortality in randomized clinical trials compared to other agents.63,64 Although an intravenous fosfomycin formulation is available in Europe, it is only available in the U.S. as a 3-g cachet that is mixed with water and ingested. Limited data suggest that oral fosfomycin may represent a treatment option for urinary tract infections caused by CRE, but this dosage is unlikely to achieve sufficient bloodstream concentrations to treat invasive CRE infections.66,75 Aminoglycosides are not consistently active against CRE and have the well-known limitations of nephrotoxicity and otovestibular toxicity.44,67,68 Like polymyxins, aminoglycosides have also been associated with worse outcomes than β-lactam agents when used as monotherapy to treat gram-negative bacteremia in oncology patients.72,76

TABLE 4.

Available antimicrobial agents with activity against CRE

Antimicrobial Agent (reference) Recommended Dosages (normal renal function) Limitations
Polymyxins (5862)
 Polymyxin B (administered as the active compound) 2.5 mg/kg loading dose, then 1.5 mg/kg every 12h1
  • Do not need to adjust dosage for renal insufficiency

  • Nephrotoxicity and neurotoxicity

  • Low concentrations in the urinary tract

 Colistin (administered as the prodrug colistin methanesulfonate) 5 mg/kg/day of CBA (up to 300 mg), in 2 divided doses2,3
  • Adjust dosage for renal insufficiency

Similar to polymyxin B, except:
  • Inefficient conversion to active compound

  • Active drug accumulates in the urinary tract

  • More nephrotoxic than polymyxin B

Tigecycline (6365) 100 mg loading dose, then 50 mg every 12h
  • Doses up to 100 mg every 12h have been evaluated in clinical trials and may be necessary for pneumonia

  • Use associated with increased mortality in randomized trials

  • Low bloodstream and urinary tract concentrations

  • Gastrointestinal side effects

Fosfomycin (66)
  • U.S.: oral formulation only: 3 g in 3–4 oz. of water (once, or every 2–3 d for 3 doses for urinary tract infections)

  • IV formulation: 8 g every 12h

  • IV formulation not available in the U.S

    • Used as adjunctive therapy for CRE infections where available

  • Low barrier to development of resistance

Aminoglycosides (44,67,68)4
 Gentamicin
  • Extended-interval: 5–7 mg/kg every 24h

  • Conventional: 2–3 mg/kg loading dose, followed by 1.5–2 mg/kg every 8 hours

  • Variable activity vs. CRE (40–60% of KPC producers in the U.S. are susceptible; most NDM producers are resistant)

  • Nephrotoxicity and otovestibular toxicity

  • Suboptimal clinical efficacy when used as monotherapy

 Amikacin
  • Extended-interval: 15 mg/kg every 24h

  • Conventional: 7.5 mg/kg every 12 hours

Similar to gentamicin, except:
  • Less nephrotoxicity and otovestibular toxicity

  • Usually less active vs. KPC producers

Ceftazidime-avibactam (6971) 2.5 g as a 2-hour infusion every 8h (2 g ceftazidime, 0.5 g avibactam) See Table 5 for limitations
1

1 mg polymyxin B = 10,000 units polymyxin B.

2

300 mg CBA is equal to approximately 9 millin international units (MIU).

3

A loading dose of 300 mg (9 MIU) should be considered in severe infections.

4

Tobramycin is rarely active against CRE.

CRE, carbapenem-resistant Enterocateriaceae; CBA, colistin base activity; IV, intravenous.

Given the limitations of the above agents, combination therapy is often employed for the treatment of CRE infections. No randomized clinical trials have been completed to definitively evaluate the merits of combination therapy. Some observational studies of CRE bacteremia (mostly KPC-producing K. pneumoniae) in the general population have demonstrated a mortality benefit when two or more drugs with in vitro activity against the organism are used,7779 whereas others have failed to detect a benefit to combination therapy.80,81 In a study of CRKP bacteremia specifically in neutropenic patients with hematologic malignancies, Tofas et al.53 found that use of more than one CRE-active agent was independently associated with decreased mortality.

Carbapenems are often included in treatment regimens for CRE infections even though CRE are characterized as carbapenem-resistant in vitro. High-dose, prolonged infusions of meropenem (2 g over 3 hours, every 8 hours) can maintain serum concentrations above the MIC for a sufficient portion of the dosing interval to achieve bacterial killing when MICs are 4–8 μg/mL (meropenem resistance is defined by the CLSI as an MIC ≥ 4 μg/mL).82 Clinical studies corroborate these models, demonstrating improved outcomes with the addition of high-dose, prolonged infusions of meropenem to an active agent for KPC-producing K. pneumoniae infections when the meropenem MIC is ≤ 8 μg/mL.77 Unfortunately, the majority of CRE bloodstream isolates have meropenem MICs ≥ 16 μg/mL, limiting the utility of this approach.77,80.

Ceftazidime-avibactam was approved by the U.S. Food and Drug Administration (FDA) in 2015 and represents the first β-lactam/β-lactamase inhibitor with activity against KPC-producing CRE (Table 5 83,84). This agent has demonstrated comparable efficacy to carbapenems in phase 3 clinical trials of complicated urinary tract and intra-abdominal infections.6971 Given the excellent track record of β-lactam agents for the treatment of gram-negative infections in neutropenic patients and HSCT recipients, ceftazidime-avibactam is a promising treatment option for KPC-producing CRE infections in this patient population. However, almost all of the patients in clinical trials were infected with carbapenem-susceptible pathogens and few if any of these patients were neutropenic or a transplant recipient. Pre-clinical in vivo animal data and clinical data are urgently needed to assess the effectiveness of this promising compound for the treatment of CRE infections in these populations.

TABLE 5.

Comparison of two new cephalosporin/β-lactamase inhibitor combinations: ceftazidime-avibactam and ceftolozane-tazobactam (83)

Properties Ceftazidime-avibactam Ceftolozane-tazobactam
Spectrum of activity
 Enterobacteriaceae
  ESBL-producers Excellent activity Good activity against ESBL-producing Escherichia coli, but only modest activity against ESBL-producing Klebsiella pneumoniae
  CRE
  • Active against KPC-producers and other serine carbapenemase-producers

  • Not active vs. metallo-β-lactamase producers (e.g., NDM)

Not active
  AmpC-hyperproducers Excellent activity Modest activity
Pseudomonas aeruginosa Active in vitro against ~2/3 of strains that are resistant to CAZ, PTZ, and MER
Acinetobacter baumannii Poor activity
 Gram-negative anaerobes Limited activity vs. Bacteroides species
 MSSA Limited activity
U.S. FDA-approved indications Complicated urinary tract and intra-abdominal infections
Clinical data in HSCT recipients No
Dosing 2.5 g every 8 hours (2 g ceftazidime, 0.5 g avibactam) 1.5 g every 8 hours (1 g ceftolozane, 0.5 g avibactam)1
 Adjustment for renal insufficiency Yes
1

Ceftolozane-tazobactam is being evaluated in clinical trials as a 3 g every 8 hour dosage for nosocomial pneumonia (84).

CAZ, ceftazidime; CRE, carbapenem-resistant Enterobacteriaceae; ESBL, extended-spectrum β-lactamase; FDA, Food and Drug Administration; HSCT, hematopoietic stem cell transplant recipients; KPC, Klebsiella pneumoniae carbapenemase; MER, meropenem; MSSA, methicillin-susceptible Staphylococcus aureus; NDM, New Delhi metallo-β-lactamase; PTZ, piperacillin-tazobactam.

Despite its potent activity against ESBL-E and KPC-producing CRE, limitations of the antimicrobial spectrum of ceftazidime-avibactam should be noted. First, it is not active against metallo-β-lactamase-producing Enterobacteriaceae. This limitation is compounded by the fact that ceftazidime-avibactam is not currently on automated antimicrobial susceptibility platforms. Thus, AST for this agent requires additional manual testing and results are not available until at least 1 day after identification of a CRE. Knowledge of the most common carbapenem resistance mechanisms among Enterobacteriaceae in a given epidemiologic setting, therefore, becomes critical in selecting initial therapy once a CRE is identified. While ceftazidime-avibactam may be an appropriate choice for CRE in an area where KPC-producing CRE predominate, it would not be appropriate in an area where metallo-β-lactamase-producing Enterobacteriaceae are prevalent. A recent report from MD Anderson Cancer Center highlighted this point, finding that 7 of 10 oncology patients with CRE bacteremia had ceftazidime-avibactam-resistant isolates and six of these seven isolates produced New Delhi metallo-β-lactamase (NDM).85 Even in areas where KPC-producing CRE predominate, ceftazidime-avibactam AST is critical, as reports of ceftazidime-avibactam resistance among these organisms have already emerged.8688 It is also important to understand that, unlike most other β-lactam/β-lactamase inhibitors, ceftazidime-avibactam has only modest activity against streptococci and staphylococci and no activity against enterococci.83 Furthermore, it is not consistently active against Bacteroides or Clostridium species, and thus metronidazole was added when ceftazidime-avibactam was evaluated in the treatment of complicated intra-abdominal infections.69,83

4 PSEUDOMONAS AERUGINOSA

4.1 Epidemiology

Recommendations for empirical antibacterial therapy in neutropenic patients have largely focused on P. aeruginosa because of the high mortality rates observed in patients infected with this organism, who are not immediately treated with effective therapy.7,72 P. aeruginosa remains a common cause of gram-negative bacteremia in HSCT recipients, particularly in the pre-engraftment period, as recent studies demonstrate it is responsible for 9%–26% of gram-negative bacteremias in this setting.18,20,8991

Unfortunately, P. aeruginosa has a remarkable capacity to develop resistance, and many strains are now resistant to anti-pseudomonal β-lactam agents used for febrile neutropenia.92 For example, in a recent study of 66 P. aeruginosa bloodstream isolates from nine hematology wards in Italy, 55% were resistant to ceftazidime, 71% were resistant to meropenem, and 42% were resistant to piperacillin-tazobactam.9 This staggeringly high prevalence of resistance to anti-pseudomonal β-lactam agents is ominous, because inactive empirical therapy is consistently associated with increased mortality in neutropenic patients with gram-negative bacteremia.9,91,93 Not surprisingly, outcomes were poor in patients who were infected with MDR P. aeruginosa strains, who presumably did not receive active empirical therapy, as 42% of infected patients died within 21 days. Although these extremely high rates of resistance are likely not applicable to most U.S. HSCT centers, recent data suggest that β-lactam-resistance in P. aeruginosa is also emerging domestically. Of 279 P. aeruginosa central line-associated BSIs in patients on oncology units from 2009–2012 that were reported to the CDC’s National Healthcare Safety Network, 20% of isolates were carbapenem-resistant and 14% were resistant to anti-pseudomonal cephalosporins.94

4.2 Treatment

Given this substantial resistance to anti-pseudomonal β-lactam agents, invasive infections caused by P. aeruginosa in neutropenic patients should typically be treated with both an anti-pseudomonal β-lactam and an aminoglycoside or fluoroquinolone (if this agent is not being used prophylactically) while awaiting susceptibility data. If AST reveals that the organism is susceptible to an anti-pseudomonal β-lactam, the role of continuing combination therapy is less clear, as most observational studies have not identified a benefit to adding a second agent for P. aeruginosa bacteremia in oncology patients.10,12,76,95 However, the development of resistance while on therapy is a major problem, occurring in 10% of all P. aeruginosa infections and one-third of cases of P. aeruginosa pneumonia treated with carbapenems.96,97 Although in vitro models suggest that combination therapy with the addition of an aminoglycoside or fluoroquinolone can prevent the emergence of β-lactam resistance, 98,99 currently no clinical data confirm these findings.

The administration of β-lactam agents as an extended infusion (over hours instead of over 30 minutes) is a strategy has also been shown to decrease the emergence of resistance in P. aeruginosa in in vitro models.100 Furthermore, extended infusion strategies maintain the time that the concentration of the antibiotic is above the MIC for a longer portion of the dosing interval than standard infusions, and this parameter is correlated with increased bactericidal activity against P. aeruginosa in animal models.101 Although no randomized controlled trials have been conducted to definitively prove the benefit of this strategy for P. aeruginosa infections, two single-center observational studies demonstrated decreased mortality in critically ill patients with P. aeruginosa infection after switching from standard infusions of piperacillin-tazobactam and cefepime to extended infusions of these agents.101,102 These compelling in vitro and observational data provide rationale for using extended infusions of β-lactam agents for the treatment of invasive P. aeruginosa infections in HSCT recipients, provided that venous access is not an issue.

P. aeruginosa that is not susceptible to anti-pseudomonal β-lactam agents or fluoroquinolones has previously been treated with polymyxins or aminoglycosides as agents of last resort. Unfortunately, these antibiotics have high rates of toxicity and are less effective than β-lactam agents in oncology patients with P. aeruginosa bacteremia.72,76 Ceftolozane-tazobactam, an additional new cephalosporin/β-lactamase inhibitor, is a promising alternative for the treatment of these MDR P. aeruginosa infections (Table 5). Approximately two-thirds of P. aeruginosa isolates that are resistant to extended-spectrum cephalosporins, piperacillin-tazobactam, and meropenem test susceptible to this new agent.103 Murine models suggest that ceftolozane-tazobactam has potent in vivo bactericidal activity against MDR P. aeruginosa infections.104 However, clinical trials of this agent have only demonstrated efficacy for the treatment of complicated urinary tract and intra-abdominal infections caused by carbapenem-susceptible P. aeruginosa.105,106 Its efficacy has not been evaluated in the setting of BSIs, infections caused by carbapenem-resistant P. aeruginosa, or in neutropenic patients or HSCT recipients. Nevertheless, given the major limitations of polymyxin and aminoglycoside monotherapy, ceftolozane-tazobactam should be considered for HSCT recipients with infections caused by P. aeruginosa strains that are resistant to other anti-pseudomonal β-lactam agents.

Ceftazidime-avibactam represents an additional option for MDR P. aeruginosa infections. In addition to having in vitro activity against KPC-producing Enterobacteriaceae, it also has in vitro activity against MDR P. aeruginosa strains that is comparable to that of ceftolozane-tazobactam (Table 5).107,108 Preliminary in vivo murine models suggest that the addition of avibactam to ceftazidime enhances bacterial killing of P. aeruginosa infections compared to ceftazidime alone.109 As with ceftolozane-tazobactam, additional pre-clinical and clinical data are urgently needed to better understand the role of this new agent for MDR P. aeruginosa infections.

5 VRE

5.1 Epidemiology

VRE is increasingly recognized as a major pathogen in HSCT recipients. In fact, some centers have reported that VRE is the most common cause of bacteremia early after allogeneic transplantation.17,18 The reported cumulative incidence of post-transplant VRE bacteremia in allo-HSCT recipients at U.S. centers is 6%–16%.17,18,110,111 VRE bacteremia in this population typically occurs in the setting of broad-spectrum antimicrobial exposures during the neutropenic period, which leads to an absence of normal enteric bacteria and intestinal domination with VRE.18,112

VRE bacteremia after allogeneic transplantation has significant consequences. Although VRE is typically considered to be a relatively avirulent pathogen,113 it sometimes causes bacteremia and septic shock in neutropenic HSCT recipients.17,18,114 At our center, we found that VRE bacteremia led to septic shock and 7-day mortality at rates comparable to those observed after gram-negative bacteremia.18 Post-transplant VRE bacteremia has also been independently associated with increased mortality.110,111 However, whether this increased mortality is directly attributable to VRE infection or a marker for a complicated post-transplant course is unclear.

5.2 Treatment

In addition to vancomycin resistance, VRE are typically resistant to all β-lactam agents, leaving few available treatment options.113 Daptomycin and linezolid are most commonly used for the treatment of invasive VRE infections, although only linezolid has U.S. FDA approval for this indication. No randomized clinical trials are comparing the efficacy of these two agents for the treatment of VRE bacteremia, and observational studies have provided conflicting results as to which agent is superior.115,116

Daptomycin is often preferred to linezolid for VRE bacteremia in HSCT recipients because it has bactericidal activity and a favorable safety profile. In contrast, linezolid is bacteriostatic, myelosuppressive, and associated with a slight delay in neutrophil recovery in oncology patients.117 Daptomycin is not an effective therapy for pneumonia because it is inactivated by alveolar surfactants.118 Furthermore, daptomycin resistance in VRE has been increasingly reported,119 particularly in patients with hematologic malignancies. One major cancer center reported that 15% of their VRE bloodstream isolates were daptomycin-non-susceptible.120 Moreover, a recent multicenter study of Enterococcus faecium bacteremia, which included many HSCT recipients and neutropenic patients, questioned whether the current CLSI susceptible breakpoint of ≤ 4 μg/mL for daptomycin is too high, implying an underreporting of daptomycin resistance.121 They found that patients with E. faecium bacteremia with daptomycin MICs of 3–4 μg/mL by Etest had higher rates of microbiologic failure compared to cases with MICs of ≤ 2 μg/mL, even after adjusting for co-variates. Although the U.S. FDA-approved dosage of daptomycin is 6 mg/kg/day, higher dosages are recommended for VRE bacteremia because daptomycin MICs are higher for enterococci than for other gram-positive bacteria. Recent data suggest that daptomycin dosages of 8–10 mg/kg/day are well tolerated and two observational studies have demonstrated decreased mortality after VRE bacteremia with dosages of ≥ 9 mg/kg and ≥ 10 mg/kg, respectively, compared to lower doses.122125 Some experts recommend using daptomycin in combination with β-lactam agents (e.g., ampicillin or ceftaroline) for VRE infections with elevated daptomycin MICs, as these combinations demonstrate marked in vitro synergy.126 Weekly monitoring of serum creatinine phosphokinase levels is recommended during daptomycin therapy, as almost 3% of patients receiving daptomycin in clinical trials developed elevated creatinine phosphokinase levels.127

Other agents with in vitro activity against VRE include quinupristin-dalfopristin, tigecycline, tedizolid, oritavancin, and telavancin.128 Quinupristin-dalfopristin is rarely used because of high rates of adverse effects that lead to treatment discontinuation in nearly 20% of patients.129 Tigecycline has major limitations as outlined in Table 4, and sparse clinical data support the use of these alternative agents for VRE bacteremia. Tedizolid is a new oxazolidinone that appears to be less myelosuppressive than linezolid,130 and thus is an attractive candidate for use in HSCT recipients. However, animal models have demonstrated that the efficacy of tedizolid is markedly reduced in the setting of neutropenia,130 thus making it a less attractive alternative for VRE infections in neutropenic patients. Oritavancin may be an option for linezolid- and daptomycin-non-susceptible VRE infections, but in vivo and clinical data are extremely limited.

6 PREVENTION OF MDR BACTERIAL INFECTIONS IN HSCT RECIPIENTS

6.1 Prevention of transmission of MDR pathogens

The limited antimicrobial armamentarium against MDR Enterobacteriaceae, P. aeruginosa, and VRE, and the poor outcomes associated with these infections in HSCT recipients, highlight the importance of preventing these infections. Infection prevention efforts should focus on minimizing the risk of transmission of these MDR pathogens among patients located on HSCT units. In 2009, guidelines representing a collaborative effort from several organizations were published that provide an excellent resource for recommendations for preventing infectious complications in HSCT recipients.131 Infection prevention strategies that are most relevant to preventing transmission of MDR bacteria are ensuring strict adherence to hand hygiene and environmental cleaning practices, following evidence-based guidelines for proper insertion and care of intravascular catheters,132 implementation of contact precautions for patients known to be colonized with MDR bacteria, and use of single-patient rooms, where feasible. Although the effectiveness of each of these strategies in preventing MDR bacterial infections in HSCT recipients is unclear, they appear to be effective when implemented together.133

6.2 Antimicrobial stewardship

Antimicrobial stewardship also plays an important role in preventing MDR bacterial infections in HSCT recipients. Broad-spectrum antimicrobial agents are consistently identified as risk factors for MDR gram-negative and VRE infections in oncology patients,18,21,55 and thus limiting unnecessary uses of these agents is an important goal. Although broad-spectrum antimicrobial therapy is often necessary in HSCT recipients, multidisciplinary stewardship teams that include transplant oncologists, infectious diseases physicians, microbiologists, and pharmacists should strive to implement the following recommend practices: de-escalation of broad-spectrum therapy in stable patients with a non-MDR pathogen, daily assessments for the need for continued antibacterial therapy, and optimization of antibacterial dosages.134 Furthermore, this multidisciplinary team should review their HSCT unit’s epidemiology and antibiogram on a regular basis and implement antimicrobial treatment algorithms for common scenarios, such as febrile neutropenia or sepsis.135

6.3 GI screening for implementation of contact precautions and targeted decolonization

Active surveillance, where HSCT recipients are screened for colonization with MDR gram-negative bacteria and VRE, can be utilized to implement multiple interventions. First, patients who are found to be colonized with an MDR pathogen can be placed on contact precautions, which may decrease the inpatient transmission of these organisms. Although this approach has led to reductions in CRE infection rates in certain settings,136 data are sparse to indicate that this intervention decreases the nosocomial transmission of ESBL-E, P. aeruginosa, or VRE. In fact, a recent article demonstrated that discontinuation of a program of active surveillance for VRE and contact isolation of colonized patients had no effect on the incidence of VRE infection on a hematologic oncology unit.137

Active surveillance can also be used to identify colonized patients who may be candidates for targeted decolonization strategies. Decolonization has primarily been evaluated using oral aminoglycosides and/or colistin to prevent ESBL-E and CRE infections in colonized patients. Although decolonization may reduce carriage of these organisms during therapy, this reduction is generally short-lived and can lead to resistance to the agents that are used.138140 Furthermore, it may not decrease the risk of infection in colonized HSCT recipients. In a study of 15 patients undergoing HSCT who were colonized with CRKP, 8 patients developed post-transplant CRKP infection despite receiving decolonization therapy with oral gentamicin.141 Given the unclear clinical benefit of decolonization strategies in HSCT recipients, as well as the risk of increasing resistance to the few remaining antimicrobial agents for MDR bacterial infections, we cannot recommend implementation of decolonization protocols at this time.

7 ADDITIONAL STRATEGIES TO IMPROVE OUTCOMES OF INFECTED PATIENTS

7.1 GI screening as a guide for empirical antibacterial therapy

The emergence of MDR Enterobacteriaceae, P. aeruginosa, and VRE as causes of bacteremia in HSCT recipients warrants consideration of strategies to identify patients who are at high risk of developing infections caused by these pathogens. Such patients could potentially have their initial empirical therapy modified to ensure coverage of MDR bacteria with which they are at high risk of being infected. Active surveillance to identify hematologic oncology patients who are colonized with MDR bacteria may identify a high-risk group of patients. Investigators from Germany and Mexico found that 7% and 22%, respectively, of patients with hematologic malignancies who were colonized with ESBL-E by cultures of stool or perianal swabs developed subsequent ESBL-E bacteremia during admission for chemotherapy.142,143 At our center, we found that 6 of 13 allo-HSCT recipients who were colonized with ESBL-E on admission for transplant developed ESBL-E bacteremia during neutropenia, compared to none of 118 patients who were not colonized (P<.001).144 A multicenter study of HSCT recipients in Italy also identified high rates of GI colonization to infection for CRKP. CRKP infection occurred after transplantation in 26% of colonized autologous HSCT recipients and 39% of colonized allo-HSCT recipients.54

These data suggest that screening for GI colonization with MDR Enterobacteriaceae may be a useful tool to identify HSCT recipients who are at high risk of developing subsequent infection caused by these organisms, and who may benefit from an adjustment of their empirical therapy to cover for these pathogens. However, prospective clinical trials are needed to evaluate the clinical impact of this strategy. Moreover, additional data are needed to assess whether screening should be performed on admission for transplant only or if it should be continued weekly during neutropenia. In addition, these screening strategies should only be considered in HSCT centers with a substantial incidence of infections caused by these organisms.

In contrast to MDR Enterobacteriaceae, assessment for GI colonization is not an effective tool in identifying patients at high risk for P. aeruginosa infection. In a study of 794 allo-HSCT recipients, the majority of patients who developed a P. aeruginosa infection did not have fecal colonization, suggesting a different source of infection.145

Screening HSCT recipients for GI colonization with VRE may also identify patients at high-risk for post-transplant VRE infection. Studies of allo-HSCT recipients demonstrate that 14%–19% of patients who are colonized with VRE prior to their transplant develop VRE bacteremia early after transplantation, compared to only a 4%–6% incidence in patients who are not colonized pre-transplant.17,110,146 Notably, however, a substantial proportion of VRE bacteremias occur in patients who are not colonized pre-transplant, but acquire VRE during their transplant admission.17 Despite the increased risk of VRE bacteremia in colonized patients undergoing allo-HSCT, it is unclear whether VRE-colonized patients should have their empirical therapy modified when they present with fever and neutropenia, particularly given reports of increasing resistance to VRE-active agents like daptomycin.118,119 Unlike gram-negative infections, no data show that delays in treatment of VRE bacteremia are associated with worse outcomes in this population. However, a recent study of hospital-onset VRE bacteremia in the general inpatient population demonstrated that delay in active therapy of >48 hours was associated with increased mortality.147 Thus, it is reasonable to initiate VRE-active therapy in colonized HSCT recipients who develop bacteremia with a Gram stain compatible with VRE (gram-positive cocci in pairs or short chains) or who present with severe sepsis.

7.2 Rapid identification of bloodstream pathogens

An additional strategy to improve the outcomes of HSCT recipients who are infected with MDR Enterobacteriaceae, P. aeruginosa, and VRE is to implement new technologies in the clinical microbiology laboratory to more rapidly identify these pathogens. This, in turn, could lead to shorter delays in the administration of effective antimicrobial therapy. Real-time multiplex polymerase chain reaction systems are now available that detect a variety of bacteria, yeasts, and important resistance determinants directly from positive blood culture bottles within 2 hours of culture positivity. Both systems, the FilmArray® Blood Culture Identification Panel (Biofire Diagnostics, bioMérieux, Salt Lake City, UT USA) and the Verigene® gram-positive and gram-negative Blood Culture Tests (Nanosphere, Northbrook, IL USA) detect enterococci, the most common Enterobacteriaceae, and P. aeruginosa, in addition to other prominent bloodstream pathogens.148150 It is important to note that both systems detect the vanA and vanB genes, which confer vancomycin resistance in enterococci, and blaKPC, the most common gene that confers carbapenem resistance in Enterobacteriaceae in the U.S. and many other countries. In addition, the Verigene system detects genes that encode other carbapenemases and the most common ESBL, CTX-M.

Implementation of one of these systems should decrease the time to identification of MDR Enterobacteriaceae, P. aeruginosa, and VRE from 24–72 hours after culture positivity to only 2 hours after culture positivity. This improved speed of detection should lead to more timely therapy for infections caused by these bacteria, which will hopefully improve outcomes. However, it should be noted that these assays do not provide information to predict antimicrobial susceptibilities of P. aeruginosa bloodstream isolates. Furthermore, no data have evaluated the impact of these tools on clinical outcomes in HSCT recipients. One study performed in the general inpatient population, where the microbiology laboratory randomized positive blood cultures to conventional processing or the use of the FilmArray® Blood Culture Identification Panel, found that use of the polymerase chain reaction assay led to a decrease in use of broad-spectrum antimicrobial agents, but no improvements in clinical outcomes.151 Studies evaluating the clinical impact of these technologies are urgently needed in HSCT recipients.

7.3 Management of infected central venous catheters (CVCs)

Bacteremia caused by MDR Enterobacteriaceae and VRE in HSCT recipients is most commonly from GI translocation during chemotherapy-induced neutropenia.7,112,144 However, CVCs are occasionally the sources of bacteremia caused by these organisms. P. aeruginosa is less likely to originate from the GI tract than VRE and MDR Enterobacteriaceae and is more likely to cause CVC-related bacteremias.7,145 If blood cultures are collected from a peripheral vein and a CVC simultaneously, then culture positivity that occurs 2 hours earlier in the CVC culture compared to the peripheral culture is suggestive that the catheter is the source of the infection.152 Catheter removal is imperative for all CVC-related infections that involve the tunnel track or port pocket site or in the setting of hemodynamic instability.153 In cases of CVC-related bacteremias in hemodynamically stable patients that do not involve a tunnel track or port pocket site, CVC retention can be considered, particularly in patients with refractory thrombocytopenia or lack of alternate intravenous access. If the CVC is retained, antimicrobial therapy should be administered through the infected catheter, potentially in combination with antibiotic lock therapy.7

8 CONCLUSIONS

In summary, MDR Enterobacteriaceae, P. aeruginosa, and VRE are becoming increasingly common bloodstream pathogens in HSCT recipients and outcomes of patients infected with these bacteria are worse than those of patients infected with more susceptible bacteria. Developing preclinical and clinical research data to guide the use of new antimicrobial agents for the treatment of these pathogens in the HSCT population is paramount. Identification of optimal strategies to prevent these infections is also an urgent need. Screening for GI colonization to guide empirical therapy and using rapid diagnostics may be approaches that lead to improved outcomes for infected HSCT recipients.

Acknowledgments

Funding

This work was partially supported by the National Institute of Allergy and Infectious Diseases (NIAID) of the National Institutes of Health (K23 AI114994 to Dr. Satlin).

Footnotes

Author Contributions

Dr. Satlin and Dr. Walsh both contributed to the concept and design of this review and drafting and editing of the final article.

Potential conflicts of Interest

Dr. Satlin has received research grants from Allergan and Merck. Dr. Walsh has received research grants from Allergan, Medicines Company, and Merck and serves as an advisor to Allergan and Pfizer.

References

  • 1.Wingard JR, Hsu J, Hiemenz JW. Hematopoietic stem cell transplantation: An overview of infection risks and epidemiology. Infect Dis Clin North Am. 2010;24:257–272. doi: 10.1016/j.idc.2010.01.010. [DOI] [PubMed] [Google Scholar]
  • 2.Bodey GP, Buckley M, Sathe YS, Freireich EJ. Quantitative relationships between circulating leukocytes and infection in patients with acute leukemia. Ann Intern Med. 1966;64:328–340. doi: 10.7326/0003-4819-64-2-328. [DOI] [PubMed] [Google Scholar]
  • 3.Forkner CE, Jr, Frei E, 3rd, Edgcomb JH, Utz JP. Pseudomonas septicemia; observations on twenty-three cases. Am J Med. 1958;25:877–889. doi: 10.1016/0002-9343(58)90060-3. [DOI] [PubMed] [Google Scholar]
  • 4.Schimpff SC, Greene WH, Young VM, Wiernik PH. Pseudomonas septicemia: Incidence, epidemiology, prevention and therapy in patients with advanced cancer. Eur J Cancer. 1973;9:449–455. doi: 10.1016/0014-2964(73)90110-2. [DOI] [PubMed] [Google Scholar]
  • 5.Bodey GP, Rodriguez V, Luce JK. Carbenicillin therapy of gram-negative bacilli infections. Am J Med Sci. 1969;257:408–414. doi: 10.1097/00000441-196906000-00007. [DOI] [PubMed] [Google Scholar]
  • 6.Schimpff S, Satterlee W, Young VM, Serpick A. Empiric therapy with carbenicillin and gentamicin for febrile neutropenic patients with cancer and granulocytopenia. N Engl J Med. 1971;284:1061–1065. doi: 10.1056/NEJM197105132841904. [DOI] [PubMed] [Google Scholar]
  • 7.Freifeld AG, Bow EJ, Sepkowitz KA, et al. Clinical practice guideline for the use of antimicrobial agents in neutropenic patients with cancer: 2010 update by the Infectious Diseases Society of America. Clin Infect Dis. 2011;52:e56–93. doi: 10.1093/cid/cir073. [DOI] [PubMed] [Google Scholar]
  • 8.Bucaneve G, Micozzi A, Menichetti F, et al. Levofloxacin to prevent bacterial infection in patients with cancer and neutropenia. N Engl J Med. 2005;353:977–987. doi: 10.1056/NEJMoa044097. [DOI] [PubMed] [Google Scholar]
  • 9.Trecarichi EM, Pagano L, Candoni A, et al. Current epidemiology and antimicrobial resistance data for bacterial bloodstream infections in patients with hematologic malignancies: An Italian multicenter prospective survey. Clin Microbiol Infect. 2015;21:337–343. doi: 10.1016/j.cmi.2014.11.022. [DOI] [PubMed] [Google Scholar]
  • 10.Pizzo PA, Hathorn JW, Hiemenz J, et al. A randomized trial comparing ceftazidime alone with combination antibiotic therapy in cancer patients with fever and neutropenia. N Engl J Med. 1986;315:552–558. doi: 10.1056/NEJM198608283150905. [DOI] [PubMed] [Google Scholar]
  • 11.Cometta A, Calandra T, Gaya H, et al. Monotherapy with meropenem versus combination therapy with ceftazidime plus amikacin as empiric therapy for fever in granulocytopenic patients with cancer. The International Antimicrobial Therapy Cooperative Group of the European Organization for Research and Treatment of Cancer and the Gruppo Italiano Malattie Ematolgiche Maligne dell’Adulto Infection Program. Antimicrob Agents Chemother. 1996;40:1108–1015. doi: 10.1128/aac.40.5.1108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.De Pauw BE, Deresinski SC, Feld R, Lane-Allman EF, Donnelly JP. Ceftazidime compared with piperacillin and tobramycin for the empiric treatment of fever in neutropenic patients with cancer. A multicenter randomized trial. The Intercontinental Antimicrobial Study Group. Ann Intern Med. 1994;120:834–844. doi: 10.7326/0003-4819-120-10-199405150-00004. [DOI] [PubMed] [Google Scholar]
  • 13.Del Favero A, Menichetti F, Martino P, et al. A multicenter, double-blind, placebo-controlled trial comparing piperacillin-tazobactam with and without amikacin as empiric therapy for febrile neutropenia. Clin Infect Dis. 2001;33:1295–1301. doi: 10.1086/322646. [DOI] [PubMed] [Google Scholar]
  • 14.Raad II, Escalante C, Hachem RY, et al. Treatment of febrile neutropenic patients with cancer who require hospitalization: A prospective randomized study comparing imipenem and cefepime. Cancer. 2003;98:1039–1047. doi: 10.1002/cncr.11613. [DOI] [PubMed] [Google Scholar]
  • 15.Boucher HW, Talbot GH, Bradley JS, et al. Bad bugs, no drugs: No ESKAPE! An update from the Infectious Diseases Society of America. Clin Infect Dis. 2009;48:1–12. doi: 10.1086/595011. [DOI] [PubMed] [Google Scholar]
  • 16.Sievert DM, Ricks P, Edwards JR, et al. Antimicrobial-resistant pathogens associated with healthcare-associated infections: Summary of data reported to the National Healthcare Safety Network at the Centers for Disease Control and Prevention, 2009–2010. Infect Control Hosp Epidemiol. 2013;34:1–14. doi: 10.1086/668770. [DOI] [PubMed] [Google Scholar]
  • 17.Kamboj M, Chung D, Seo SK, et al. The changing epidemiology of vancomycin-resistant Enterococcus (VRE) bacteremia in allogeneic hematopoietic stem cell transplant (HSCT) recipients. Biol Blood Marrow Transplant. 2010;16:1576–1581. doi: 10.1016/j.bbmt.2010.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Satlin MJ, Soave R, Racanelli AC, et al. The emergence of vancomycin-resistant enterococcal bacteremia in hematopoietic stem cell transplant recipients. Leuk Lymphoma. 2014;55:2858–2865. doi: 10.3109/10428194.2014.896007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Paterson DL, Bonomo RA. Extended-spectrum beta-lactamases: A clinical update. Clin Microbiol Rev. 2005;18:657–686. doi: 10.1128/CMR.18.4.657-686.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Mikulska M, Del Bono V, Raiola AM, et al. Bloodstream infections in allogeneic hematopoietic stem cell transplant recipients: Reemergence of gram-negative rods and increasing antibiotic resistance. Biol Blood Marrow Transplant. 2009;15:47–53. doi: 10.1016/j.bbmt.2008.10.024. [DOI] [PubMed] [Google Scholar]
  • 21.Gudiol C, Calatayud L, Garcia-Vidal C, et al. Bacteraemia due to extended-spectrum beta-lactamase-producing Escherichia coli (ESBL-EC) in cancer patients: Clinical features, risk factors, molecular epidemiology and outcome. J Antimicrob Chemother. 2010;65:333–341. doi: 10.1093/jac/dkp411. [DOI] [PubMed] [Google Scholar]
  • 22.Cornejo-Juarez P, Perez-Jimenez C, Silva-Sanchez J, et al. Molecular analysis and risk factors for Escherichia coli producing extended-spectrum β-lactamase bloodstream infection in hematological malignancies. PLoS One. 2012;7:e35780. doi: 10.1371/journal.pone.0035780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kang CI, Chung DR, Ko KS, Peck KR, Song HJ Korean Network for Study of Infectious Diseases. Risk factors for infection and treatment outcome of extended-spectrum β-lactamase-producing Escherichia coli and Klebsiella pneumoniae bacteremia in patients with hematologic malignancy. Ann Hematol. 2012;91:115–121. doi: 10.1007/s00277-011-1247-7. [DOI] [PubMed] [Google Scholar]
  • 24.Ha YE, Kang CI, Cha MK, et al. Epidemiology and clinical outcomes of bloodstream infections caused by extended-spectrum β-lactamase-producing Escherichia coli in patients with cancer. Int J Antimicrob Agents. 2013;42:403–409. doi: 10.1016/j.ijantimicag.2013.07.018. [DOI] [PubMed] [Google Scholar]
  • 25.Kim SH, Kwon JC, Choi SM, et al. Escherichia coli and Klebsiella pneumoniae bacteremia in patients with neutropenic fever: Factors associated with extended-spectrum β-lactamase production and its impact on outcome. Ann Hematol. 2013;92:533–541. doi: 10.1007/s00277-012-1631-y. [DOI] [PubMed] [Google Scholar]
  • 26.Yemişen M, Balkan II, Salihoğlu A, et al. The changing epidemiology of bloodstream infections and resistance in hematopoietic stem cell transplantation recipients. Turk J Hematol. 2016;33:216–222. doi: 10.4274/tjh.2014.0378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Park YS, Adams-Haduch JM, Shutt KA, et al. Clinical and microbiologic characteristics of cephalosporin-resistant Escherichia coli at three centers in the United States. Antimicrob Agents Chemother. 2012;56:1870–186. doi: 10.1128/AAC.05650-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Castanheira M, Farrell SE, Krause KM, Jones RN, Sader HS. Contemporary diversity of β-lactamases among Enterobacteriaceae in the nine U.S. census regions and ceftazidime-avibactam activity tested against isolates producing the most prevalent β-lactamase groups. Antimicrob Agents Chemother. 2014;58:833–838. doi: 10.1128/AAC.01896-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Thomson KS, Moland ES. Cefepime, piperacillin-tazobactam, and the inoculum effect in tests with extended-spectrum beta-lactamase-producing Enterobacteriaceae. Antimicrob Agents Chemother. 2001;45:3548–3554. doi: 10.1128/AAC.45.12.3548-3554.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Harada Y, Morinaga Y, Kaku N, et al. In vitro and in vivo activities of piperacillin-tazobactam and meropenem at different inoculum sizes of ESBL-producing Klebsiella pneumoniae. Clin Microbiol Infect. 2014;20:831–839. doi: 10.1111/1469-0691.12677. [DOI] [PubMed] [Google Scholar]
  • 31.Lee NY, Lee CC, Huang WH, Tsui KC, Hsueh PR, Ko WC. Cefepime therapy for monomicrobial bacteremia caused by cefepime-susceptible extended-spectrum beta-lactamase-producing Enterobacteriaceae: MIC matters. Clin Infect Dis. 2013;56:488–495. doi: 10.1093/cid/cis916. [DOI] [PubMed] [Google Scholar]
  • 32.Chopra T, Marchaim D, Veltman J, et al. Impact of cefepime therapy on mortality among patients with bloodstream infections caused by extended-spectrum-β-lactamase-producing Klebsiella pneumoniae and Escherichia coli. Antimicrob Agents Chemother. 2012;56:3936–3942. doi: 10.1128/AAC.05419-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Wang R, Cosgrove SE, Tschudin-Sutter S, et al. Cefepime therapy for cefepime-susceptible extended-spectrum β-lactamase-producing Enterobacteriaceae bacteremia. Open Forum Infect Dis. 2016;20:ofw132. doi: 10.1093/ofid/ofw132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Rodriguez-Baño J, Navarro MD, Retamar P, et al. Extended-Spectrum Beta-Lactamases–Red Española de Investigación en Patología Infecciosa/Grupo de Estudio de Infección Hospitalaria Group. β-lactam/β-lactamase inhibitor combinations for the treatment of bacteremia due to extended-spectrum β-lactamase-producing Escherichia coli: A post hoc analysis of prospective cohorts. Clin Infect Dis. 2012;54:167–174. doi: 10.1093/cid/cir790. [DOI] [PubMed] [Google Scholar]
  • 35.Ng TM, Khong WX, Harris PN, et al. Empiric piperacillin-tazobactam versus carbapenems in the treatment of bacteremia due to extended-spectrum beta-lactamase-producing Enterobacteriaceae. PLoS One. 2016;11:e0153696. doi: 10.1371/journal.pone.0153696. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Tamma PD, Han JH, Rock C, et al. Carbapenem therapy is associated with improved survival compared with piperacillin-tazobactam for patients with extended-spectrum β-lactamase bacteremia. Clin Infect Dis. 2015;60:1319–1325. doi: 10.1093/cid/civ003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Westphal JF, Brogard JM, Caro-Sampara F, et al. Assessment of biliary excretion of piperacillin-tazobactam in humans. Antimicrob Agents Chemother. 1997;41:1636–1640. doi: 10.1128/aac.41.8.1636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Gesser RM, McCarroll K, Teppler H, Woods GL. Efficacy of ertapenem in the treatment of serious infections caused by Enterobacteriaceae: Analysis of pooled clinical trial data. J Antimicrob Chemother. 2003;51(5):1253–1260. doi: 10.1093/jac/dkg237. [DOI] [PubMed] [Google Scholar]
  • 39.Lee NY, Lee CC, Huang WH, Tsui KC, Hsueh PR, Ko WC. Carbapenem therapy for bacteremia due to extended-spectrum-β-lactamase-producing Escherichia coli or Klebsiella pneumoniae: Implications of ertapenem susceptibility. Antimicrob Agents Chemother. 2012;56:2888–2893. doi: 10.1128/AAC.06301-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Collins VL, Marchaim D, Pogue JM, et al. Efficacy of ertapenem for treatment of bloodstream infections caused by extended-spectrum-β-lactamase-producing Enterobacteriaceae. Antimicrob Agents Chemother. 2012;56:2173–2177. doi: 10.1128/AAC.05913-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Gutiérrez-Gutiérrez B, Bonomo RA, Carmeli Y, et al. Ertapenem for the treatment of bloodstream infections due to ESBL-producing Enterobacteriaceae: A multinational pre-registered cohort study. J Antimicrob Chemother. 2016;71:1672–1680. doi: 10.1093/jac/dkv502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Centers for Disease Control and Prevention (CDC) Vital signs: Carbapenem-resistant Enterobacteriaceae. MMWR Morb Mortal Wkly Rep. 2013;62:165–170. [PMC free article] [PubMed] [Google Scholar]
  • 43.Bratu S, Landman D, Haag R, et al. Rapid spread of carbapenem-resistant Klebsiella pneumoniae in New York City: A new threat to our antibiotic armamentarium. Arch Intern Med. 2005;165:1430–1435. doi: 10.1001/archinte.165.12.1430. [DOI] [PubMed] [Google Scholar]
  • 44.Patel G, Huprikar S, Factor SH, Jenkins SG, Calfee DP. Outcomes of carbapenem-resistant Klebsiella pneumoniae infection and the impact of antimicrobial and adjunctive therapies. Infect Control Hosp Epidemiol. 2008;29:1099–1106. doi: 10.1086/592412. [DOI] [PubMed] [Google Scholar]
  • 45.Centers for Disease Control and Prevention (CDC) [Accessed 19 October 2016];Antibiotic Resistance Threats in the United States, 2013. http://www.cdc.gov/features/AntibioticResistanceThreats/index.html.
  • 46.Centers for Disease Control and Prevention (CDC) [Accessed 19 October 2016];Tracking CRE. http://www.cdc.gov/hai/organisms/cre/TrackingCRE.html.
  • 47.Weiner LM, Webb AK, Limbago B, et al. Antimicrobial-resistant pathogens associated with healthcare-associated infections: Summary of data reported to the National Healthcare Safety Network at the Centers for Disease Control and Prevention. Infect Control Hosp Epidemiol. 2016;37:1288–1301. doi: 10.1017/ice.2016.174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Pitout JD, Nordamnn P, Poirel L. Carbapenemase-producing Klebsiella pneumoniae, a key pathogen set for global nosocomial dominance. Antimicrob Agents Chemother. 2015;59:5873–5884. doi: 10.1128/AAC.01019-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Satlin MJ, Jenkins SG, Walsh TJ. The global challenge of carbapenem-resistant Enterobacteriaceae in transplant recipients and patients with hematologic malignancies. Clin Infect Dis. 2014;58:1274–1283. doi: 10.1093/cid/ciu052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Muñoz-Price LS, Poirel L, Bonomo RA, et al. Clinical epidemiology of the global expansion of Klebsiella pneumoniae carbapenemases. Lancet Infect Dis. 2013;13:785–796. doi: 10.1016/S1473-3099(13)70190-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Satlin MJ, Calfee DP, Chen L, et al. Emergence of carbapenem-resistant Enterobacteriaceae as causes of bloodstream infections in patients with hematologic malignancies. Leuk Lymphoma. 2013;54:799–806. doi: 10.3109/10428194.2012.723210. [DOI] [PubMed] [Google Scholar]
  • 52.Trecarichi EM, Pagano L, Martino B, et al. Bloodstream infections caused by Klebsiella pneumoniae in onco-hematological patients: Clinical impact of carbapenem resistance in a multicenter prospective study. Am J Hematol. 2016;91:1076–1081. doi: 10.1002/ajh.24489. [DOI] [PubMed] [Google Scholar]
  • 53.Tofas P, Skiada A, Angelopoulou M, et al. Carbapenemase-producing Klebsiella pneumoniae bloodstream infections in neutropenic patients with hematologic malignancies or aplastic anaemia: Analysis of 50 cases. Int J Antimicrob Agents. 2016;47:335–339. doi: 10.1016/j.ijantimicag.2016.01.011. [DOI] [PubMed] [Google Scholar]
  • 54.Girmenia C, Rossolini GM, Piciocchi A, et al. Infections by carbapenem-resistant Klebsiella pneumoniae in SCT recipients: A nationwide retrospective survey from Italy. Bone Marrow Transplant. 2015;50:282–288. doi: 10.1038/bmt.2014.231. [DOI] [PubMed] [Google Scholar]
  • 55.Satlin MJ, Cohen N, Ma KC, et al. Bacteremia due to carbapenem-resistant Enterobacteriaceae in neutropenic patients with hematologic malignancies. J Infect. 2016;73:336–345. doi: 10.1016/j.jinf.2016.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Hussein K, Raz-Pasteur A, Finkelstein R, et al. Impact of carbapenem resistance on the outcome of patients’ hospital-acquired bacteraemia caused by Klebsiella pneumoniae. J Hosp Infect. 2013;83:307–313. doi: 10.1016/j.jhin.2012.10.012. [DOI] [PubMed] [Google Scholar]
  • 57.Kontoyiannis DP, Marr KA, Park BJ, et al. Prospective surveillance for invasive fungal infections in hematopoietic stem cell transplant recipients, 2001–2006: Overview of the Transplant-Associated Infection Surveillance Network (TRANSNET) Database. Clin Infect Dis. 2010;50:1091–1100. doi: 10.1086/651263. [DOI] [PubMed] [Google Scholar]
  • 58.Rigatto MH, Oliveira MS, Perdigao-Neto LV, et al. Multicenter prospective cohort study of renal failure in patients treated with colistin versus polymyxin B. Antimicrob Agents Chemother. 2016;60:2443–2449. doi: 10.1128/AAC.02634-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Nation RL, Garonzik SM, Li J, et al. Updated US and European dose recommendations for intravenous colistin: How do they perform? Clin Infect Dis. 2016;62:552–558. doi: 10.1093/cid/civ964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Dalfino L, Puntillo F, Mosca A, et al. High-dose, extended-interval colistin administration in critically ill patients: Is this the right dosing strategy? A preliminary study. Clin Infect Dis. 2012;54:1720–1726. doi: 10.1093/cid/cis286. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Nation RL, Li J, Cars O, et al. Framework for optimization of the clinical use of colistin and polymyxin B:Tthe Prato polymyxin consensus. Lancet Infect Dis. 2015;15:225–234. doi: 10.1016/S1473-3099(14)70850-3. [DOI] [PubMed] [Google Scholar]
  • 62.Sandri AM, Landersdorfer CB, Jacob J, et al. Population pharmacokinetics of intravenous polymyxin B in critically ill patients: Implications for selection of dosage regimens. Clin Infect Dis. 2013;57:524–531. doi: 10.1093/cid/cit334. [DOI] [PubMed] [Google Scholar]
  • 63.Prasad P, Sun J, Danner RL, Natanson C. Excess deaths associated with tigecycline after approval based on noninferiority trials. Clin Infect Dis. 2012;54:1699–1709. doi: 10.1093/cid/cis270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.MacGowan AP. Tigecycline pharmacokinetic/pharmacodynamic update. J Antimicrob Chemother. 2008;62(Suppl 1):i11–16. doi: 10.1093/jac/dkn242. [DOI] [PubMed] [Google Scholar]
  • 65.Ramirez J, Dartois N, Gandjini H, Yan JL, Korth-Bradley J, McGovern PC. Randomized phase 2 trial to evaluate the clinical efficacy of two high-dosage tigecycline regimens versus imipenem-cilastatin for hospital-acquired pneumonia. Antimicrob Agents Chemother. 2013;57:1756–1762. doi: 10.1128/AAC.01232-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Falagas ME, Vouloumanou EK, Samonis G, Vardakas KZ. Fosfomycin. Clin Microbiol Rev. 2016;29:321–347. doi: 10.1128/CMR.00068-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Sanchez GV, Master RN, Clark RB, et al. Klebsiella pneumoniae antimicrobial drug resistance, United States, 1988–2010. Emerg Infect Dis. 2013;19:133–136. doi: 10.3201/eid1901.120310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Jain A, Hopkins KL, Turton J, et al. NDM carbapenemases in the United Kingdom: An analysis of the first 250 cases. J Antimicrob Chemother. 2014;69:1777–1784. doi: 10.1093/jac/dku084. [DOI] [PubMed] [Google Scholar]
  • 69.Mazuski JE, Gasink LB, Armstrong J, et al. Efficacy and safety of ceftazidime-avibactam plus metronidazole versus meropenem in the treatment of complicated intra-abdominal infection: Results from a randomized, controlled, double-blind, phase 3 program. Clin Infect Dis. 2016;62:1380–1389. doi: 10.1093/cid/ciw133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Wagenlehner FM, Sobel JD, Newell P, et al. Ceftazidime-avibactam versus doripenem for the treatment of complicated urinary tract infections, including acute pyelonephritis: RECAPTURE, a phase 3 randomized trial program. Clin Infect Dis. 2016;63:7547–7562. doi: 10.1093/cid/ciw378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Carmeli Y, Armstrong J, Laud PJ, et al. Ceftazidime-avibactam or best available therapy in patients with ceftazidime-resistant Enterobacteriaceae or Pseudomonas aeruginosa complicated urinary tract infections or intra-abdominal infections (REPRISE): A randomised, pathogen-directed, phase 3 study. Lancet Infect Dis. 2016;16:661–673. doi: 10.1016/S1473-3099(16)30004-4. [DOI] [PubMed] [Google Scholar]
  • 72.Bodey GP, Rodriguez V. Advances in the management of Pseudomonas aeruginosa infections in cancer patients. Eur J Cancer. 1973;9:435–441. doi: 10.1016/0014-2964(73)90108-4. [DOI] [PubMed] [Google Scholar]
  • 73.Monaco M, Giani T, Raffone A, et al. Colistin resistance superimposed to endemic carbapenem-resistant Klebsiella pneumoniae: A rapidly evolving problem in Italy, November 2013 to April 2014. Euro Surveill. 2014;19(42) doi: 10.2807/1560-7917.es2014.19.42.20939. [DOI] [PubMed] [Google Scholar]
  • 74.Liu YY, Wang Y, Walsh TR, et al. Emergence of plasmid-mediated colistin resistance mechanism MCR-1 in animals and human beings from China: A microbiological and molecular biological study. Lancet Infect Dis. 2016;16:161–168. doi: 10.1016/S1473-3099(15)00424-7. [DOI] [PubMed] [Google Scholar]
  • 75.Neuner EA, Sekeres J, Hall GS, van Duin D. Experience with fosfomycin for treatment of urinary tract infections due to multidrug-resistant organisms. Antimicrob Agents Chemother. 2012;56:5744–5748. doi: 10.1128/AAC.00402-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Bodey GP, Jadeja L, Elting L. Pseudomonas bacteremia. Retrospective analysis of 410 episodes. Arch Intern Med. 1985;145:1621–1629. doi: 10.1001/archinte.145.9.1621. [DOI] [PubMed] [Google Scholar]
  • 77.Tumbarello M, Trecarichi EM, De Rosa FG, et al. Infections caused by KPC-producing Klebsiella pneumoniae: Difference in therapy and mortality in a multicenter study. J Antimicrob Chemother. 2015;70:2133–2143. doi: 10.1093/jac/dkv086. [DOI] [PubMed] [Google Scholar]
  • 78.Daikos GL, Tsaousi S, Tzouvelekis LS, et al. Carbapenemase-producing Klebsiella pneumoniae bloodstream infections: Lowering mortality by antibiotic combination schemes and the role of carbapenems. Antimicrob Agents Chemother. 2014;58:2322–2328. doi: 10.1128/AAC.02166-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Falcone M, Russo A, Iacovelli A, et al. Predictors of outcome in ICU patients with septic shock caused by Klebsiella pneumoniae carbapenemase-producing K. pneumoniae. Clin Microbiol Infect. 2016;22:444–450. doi: 10.1016/j.cmi.2016.01.016. [DOI] [PubMed] [Google Scholar]
  • 80.Gomez-Simmonds A, Nelson B, Eiras DP, et al. Combination regimens for treatment of carbapenem-resistant Klebsiella pneumoniae bloodstream infections. Antimicrob Agents Chemother. 2016;60:3601–3607. doi: 10.1128/AAC.03007-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.de Oliveira MS, de Assis DB, Freire MP, et al. Treatment of KPC-producing Enterobacteriaceae: Suboptimal efficacy of polymyxins. Clin Microbiol Infect. 2015;179:e1–7. doi: 10.1016/j.cmi.2014.07.010. [DOI] [PubMed] [Google Scholar]
  • 82.Jaruratanasirikul S, Thengyai S, Wongpoowarak W, et al. Population pharmacokinetics and Monte Carlo dosing simulations of meropenem during the early phase of severe sepsis and septic shock in critically ill patients in intensive care units. Antimicrob Agents Chemother. 2015;59:2995–3001. doi: 10.1128/AAC.04166-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.van Duin D, Bonomo RA. Ceftazidime/Avibactam and Ceftolozane/Tazobactam: Second-generation β-lactam/β-lactamase inhibitor combinations. Clin Infect Dis. 2016;63:234–241. doi: 10.1093/cid/ciw243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Xiao AJ, Miller BW, Huntington JA, Nicolau DP. Ceftolozane/tazobactam pharmacokinetic/pharmacodynamic-derived dose justification for phase 3 studies in patients with nosocomial pneumonia. J Clin Pharmacol. 2016;56:56–66. doi: 10.1002/jcph.566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Aitken SL, Tarrand JJ, Deshpande LM, et al. High rates of nonsusceptibility to ceftazidime-avibactam and identification of New Delhi metallo-β-lactamase production in Enterobacteriaceae bloodstream infections at a major cancer center. Clin Infect Dis. 2016:63954–958. doi: 10.1093/cid/ciw398. [DOI] [PubMed] [Google Scholar]
  • 86.Humphries RM, Yang S, Hemarajata P, et al. First report of ceftazidime-avibactam resistance in a KPC-producing Klebsiella pneumoniae isolate. Antimicrob Agents Chemother. 2015;59:6605–6607. doi: 10.1128/AAC.01165-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Shields RK, Potoski BA, Haidar G, et al. Clinical outcomes, drug toxicity, and emergence of ceftazidime-avibactam resistance among patients treated for carbapenem-resistant Enterobacteriaceae infections. Clin Infect Dis. 2016;63:1615–1618. doi: 10.1093/cid/ciw636. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Citron DM, Tyrrell KL, Merriam V, Goldstein EJ. In vitro activity of ceftazidime-NXL104 against 396 strains of beta-lactamase-producing anaerobes. Antimicrob Agents Chemother. 2011;55:3616–3620. doi: 10.1128/AAC.01682-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Seo SK, Xiao K, Huang YT, et al. Impact of peri-transplant vancomycin and fluoroquinolone administration on rates of bacteremia in allogeneic hematopoietic stem cell transplant (HSCT) recipients: A 12-year single institution study. J Infect. 2014;69:341–351. doi: 10.1016/j.jinf.2014.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Kikuchi M, Akahoshi Y, Nakano H, et al. Risk factors for pre- and post-engraftment bloodstream infections after allogeneic hematopoietic stem cell transplantation. Transpl Infect Dis. 2015;17:56–65. doi: 10.1111/tid.12345. [DOI] [PubMed] [Google Scholar]
  • 91.Stoma I, Karpov I, Milanovich N, Uss A, Iskrov I. Risk factors for mortality in patients with bloodstream infections during the pre-engraftment period after hematopoietic stem cell transplantation. Blood Res. 2016;51:102–106. doi: 10.5045/br.2016.51.2.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Livermore DM. Multiple mechanisms of antimicrobial resistance in Pseudomonas aeruginosa: Our worst nightmare? Clin Infect Dis. 2002;34:634–640. doi: 10.1086/338782. [DOI] [PubMed] [Google Scholar]
  • 93.Mikulska M, Del Bono V, Bruzzi P, et al. Mortality after bloodstream infections in allogeneic hematopoietic stem cell transplant (HSCT) recipients. Infection. 2012;40:271–278. doi: 10.1007/s15010-011-0229-y. [DOI] [PubMed] [Google Scholar]
  • 94.See I, Freifeld AG, Magill SS. Causative organisms and associated antimicrobial resistance in healthcare-associated, central line-associated bloodstream infections from oncology settings, 2009–2012. Clin Infect Dis. 2016;62:1203–1209. doi: 10.1093/cid/ciw113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Chatzinikolaou I, Abi-Said D, Bodey GP, Rolston KV, Tarrand JJ, Samonis G. Recent experience with Pseudomonas aeruginosa bacteremia in patients with cancer: Retrospective analysis of 245 episodes. Arch Intern Med. 2000;160:501–509. doi: 10.1001/archinte.160.4.501. [DOI] [PubMed] [Google Scholar]
  • 96.Zanetti G, Bally F, Greub G, et al. Cefepime versus imipenem-cilastatin for treatment of nosocomial pneumonia: A multicenter, evaluator-blind, prospective, randomized study. Antimicrob Agents Chemother. 2003;47:3442–3447. doi: 10.1128/AAC.47.11.3442-3447.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Carmeli Y, Troillet N, Eliopoulos GM, Samore MH. Emergence of antibiotic-resistant Pseudomonas aeruginosa: Comparison of risks associated with different antipseudomonal agents. Antimicrob Agents Chemother. 1999;43:1379–1382. doi: 10.1128/aac.43.6.1379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Louie A, Grasso C, Bahniuk N, et al. The combination of meropenem and levofloxacin is synergistic with respect to Pseudomonas aeruginosa kill rate and resistance suppression. Antimicrob Agents Chemother. 2010;54:2646–2654. doi: 10.1128/AAC.00065-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Drusano GL, Bonomo RA, Bahniuk N, et al. Resistance emergence mechanism and mechanism of resistance suppression by tobramycin for cefepime for Pseudomonas aeruginosa. Antimicrob Agents Chemother. 2012;56:231–242. doi: 10.1128/AAC.05252-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Louie A, Bied A, Fregeau C, et al. Impact of different carbapenems and regimens of administration on resistance emergence for three isogenic Pseudomonas aeruginosa strains with differing mechanisms of resistance. Antimicrob Agents Chemother. 2010;54:2638–2645. doi: 10.1128/AAC.01721-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Lodise TP, Jr, Lomaestro B, Drusano GL. Piperacillin-tazobactam for Pseudomonas aeruginosa infection: Clinical implications of an extended-infusion dosing strategy. Clin Infect Dis. 2007;44:357–363. doi: 10.1086/510590. [DOI] [PubMed] [Google Scholar]
  • 102.Bauer KA, West JE, O’Brien JM, Goff DA. Extended-infusion cefepime reduces mortality in patients with Pseudomonas aeruginosa infections. Antimicrob Agents Chemother. 2013;57:2907–2912. doi: 10.1128/AAC.02365-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Farrell DJ, Flamm RK, Sader HS, Jones RN. Antimicrobial activity of ceftolozane-tazobactam tested against Enterobacteriaceae and Pseudomonas aeruginosa with various resistance patterns isolated in U.S. hospitals (2011–2012) Antimicrob Agents Chemother. 2013;57:6305–6310. doi: 10.1128/AAC.01802-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Bulik CC, Tessier PR, Keel RA, Sutherland CA, Nicolau DP. In vivo comparison of CXA-101 (FR264205) with and without tazobactam versus piperacillin-tazobactam using human simulated exposures against phenotypically diverse gram-negative organisms. Antimicrob Agents Chemother. 2012;56:544–549. doi: 10.1128/AAC.01752-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Wagenlehner FM, Umeh O, Steenbergen J, Yuan G, Darouiche RO. Ceftolozane-tazobactam compared with levofloxacin in the treatment of complicated urinary tract infections, including pyelonephritis: A randomised, double-blind, phase 3 trial (ASPECT-cUTI) Lancet. 2015;385:1949–1956. doi: 10.1016/S0140-6736(14)62220-0. [DOI] [PubMed] [Google Scholar]
  • 106.Solomkin J, Hershberger E, Miller B, et al. Ceftolozane/Tazobactam plus metronidazole for complicated intra-abdominal infection in an era of multidrug resistance: Results from a randomized, double-blind, phase 3 trial (ASPECT-cIAI) Clin Infect Dis. 2015;60:1462–1471. doi: 10.1093/cid/civ097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Buehrle DJ, Shields RK, Chen L, et al. Evaluation of the in vitro activity of ceftazidime-avibactam and ceftolozane-tazobactam against meropenem-resistant Pseudomonas aeruginosa isolates. Antimicrob Agents Chemother. 2016;60:3227–3231. doi: 10.1128/AAC.02969-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Huband MD, Castanheira M, Flamm RK, Farrell DJ, Jones RN, Sader HS. In vitro activity of ceftazidime-avibactam against contemporary Pseudomonas aeruginosa isolates from U.S. medical centers by census region, 2014. Antimicrob Agents Chemother. 2016;60:2537–2541. doi: 10.1128/AAC.03056-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Berkhout J, Melchers MJ, van Mil AC, et al. Pharmacodynamics of ceftazidime and avibactam in neutropenic mice with thigh or lung infection. Antimicrob Agents Chemother. 2015;60:368–375. doi: 10.1128/AAC.01269-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Vydra J, Shanley RM, George I, et al. Enterococcal bacteremia is associated with increased risk of mortality in recipients of allogeneic hematopoietic stem cell transplantation. Clin Infect Dis. 2012;55:764–770. doi: 10.1093/cid/cis550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Tavadze M, Rybicki L, Mossad S, et al. Risk factors for vancomycin-resistant enterococcus bacteremia and its influence on survival after allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant. 2014;49:1310–1316. doi: 10.1038/bmt.2014.150. [DOI] [PubMed] [Google Scholar]
  • 112.Taur Y, Xavier JB, Lipuma L, et al. Intestinal domination and the risk of bacteremia in patients undergoing allogeneic hematopoietic stem cell transplantation. Clin Infect Dis. 2012;55:905–914. doi: 10.1093/cid/cis580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Arias CA, Murray BE. The rise of the Enterococcus: Beyond vancomycin resistance. Nat Rev Microbiol. 2012;10:266–278. doi: 10.1038/nrmicro2761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Avery R, Kalaycio M, Pohlman B, et al. Early vancomycin-resistant enterococcus (VRE) bacteremia after allogeneic bone marrow transplantation is associated with a rapidly deteriorating clinical course. Bone Marrow Transplant. 2005;35:497–499. doi: 10.1038/sj.bmt.1704821. [DOI] [PubMed] [Google Scholar]
  • 115.Balli EP, Venetis CA, Miyakis S. Systematic review and meta-analysis of linezolid versus daptomycin for treatment of vancomycin-resistant enterococcal bacteremia. Antimicrob Agents Chemother. 2014;58:734–739. doi: 10.1128/AAC.01289-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Britt NS, Potter EM, Patel N, Steed ME. Comparison of the effectiveness and safety of linezolid and daptomycin in vancomycin-resistant enterococcal bloodstream infection: A national cohort study of Veterans Affairs patients. Clin Infect Dis. 2015;61:871–878. doi: 10.1093/cid/civ444. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Jaksic B, Martinelli G, Perez-Oteyza J, et al. Efficacy and safety of linezolid compared with vancomycin in a randomized, double-blind study of febrile neutropenic patients with cancer. Clin Infect Dis. 2006;42:597–607. doi: 10.1086/500139. [DOI] [PubMed] [Google Scholar]
  • 118.Silverman JA, Motrin LI, Vanpraagh AD, Li T, Alder J. Inhibition of daptomycin by pulmonary surfactant: In vitro modeling and clinical impact. J Infect Dis. 2005;191:2149. doi: 10.1086/430352. [DOI] [PubMed] [Google Scholar]
  • 119.Kelesidis T, Humphries R, Uslan DZ, Pegues DA. Daptomycin nonsusceptible enterococci: An emerging challenge for clinicians. Clin Infect Dis. 2011;52:228–324. doi: 10.1093/cid/ciq113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Kamboj M, Cohen N, Gilhuley K, et al. Emergence of daptomycin-resistant VRE: Experience of a single institution. Infect Control Hosp Epidemiol. 2011;32:391–394. doi: 10.1086/659152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Shukla BS, Shelburne S, Reyes K, et al. Influence of minimum inhibitory concentration in clinical outcomes of Enterococcus faecium bacteremia treated with daptomycin: Is it time to change the breakpoint? Clin Infect Dis. 2016;62:1514–1520. doi: 10.1093/cid/ciw173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Casapao AM, Kullar R, Davis SL, et al. Multicenter study of high-dose daptomycin for treatment of enterococcal infections. Antimicrob Agents Chemother. 2013;57:4190–4196. doi: 10.1128/AAC.00526-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Durante-Mangoni E, Andini R, Parrella A, et al. Safety of treatment with high-dose daptomycin in 102 patients with infective endocarditis. Int J Antimicrob Agents. 2016;48:61–68. doi: 10.1016/j.ijantimicag.2016.04.022. [DOI] [PubMed] [Google Scholar]
  • 124.Chuang YC, Lin HY, Chen PY, Lin CY, Wang JT, Chang SC. Daptomycin versus linezolid for the treatment of vancomycin-resistant enterococcal bacteremia: Implications of daptomycin dose. Clin Microbiol Infect. 2016;22:890e1–890e7. doi: 10.1016/j.cmi.2016.07.018. [DOI] [PubMed] [Google Scholar]
  • 125.Britt NS, Potter EM, Patel N, Steed ME. Comparative effectiveness and safety of standard-, medium-, and high-dose daptomycin strategies for the treatment of vancomycin-resistant enterococcal bacteremia among Veterans Affairs patients. Clin Infect Dis. 2017;64:605–613. doi: 10.1093/cid/ciw815. [DOI] [PubMed] [Google Scholar]
  • 126.Yim J, Smith JR, Rybak MJ. Role of combination antimicrobial therapy for vancomycin-resistant Enterococcus faecium infections: Review of the current evidence. Pharmacotherapy. 2017;37:579–592. doi: 10.1002/phar.1922. [DOI] [PubMed] [Google Scholar]
  • 127.Papadopoulos S, Ball AM, Liewer SE, Martin CA, Winstead PS, Murphy BS. Rhabdomyolysis during therapy with daptomycin. Clin Infect Dis. 2006;42:e108. doi: 10.1086/504379. [DOI] [PubMed] [Google Scholar]
  • 128.Crotty MP, Krekel T, Burnham CA, Ritchie DJ. New gram-positive agents: The next generation of oxazolidinones and lipoglycopeptides. J Clin Microbiol. 2016;54:2225–2232. doi: 10.1128/JCM.03395-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Lamb HM, Figgitt DP, Faulds D. Quinupristin/dalfopristin: A review of its use in the management of serious gram-positive infections. Drugs. 1999;58:1061. doi: 10.2165/00003495-199958060-00008. [DOI] [PubMed] [Google Scholar]
  • 130.Lodise TP, Fang E, Minassian SL, Prokocimer PG. Platelet profile in patients with acute bacterial skin and skin structure infections receiving tedizolid or linezolid: Findings from the Phase 3 ESTABLISH clinical trials. Antimicrob Agents Chemother. 2014;58:7198–1204. doi: 10.1128/AAC.03509-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Tomblyn M, Chiller T, Einsele H, et al. Guidelines for preventing infectious complications among hematopoietic cell transplantation recipients: A global perspective. Biol Blood Marrow Transplant. 2009;15:1143–1238. doi: 10.1016/j.bbmt.2009.06.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.O’Grady NP, Alexander M, Burns LA, et al. Guidelines for the prevention of intravascular catheter-related infections. Clin Infect Dis. 2011;52:e162–193. doi: 10.1093/cid/cir257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Tacconelli E, Cataldo MA, Dancer SJ, et al. ESCMID guidelines for the management of the infection control measures to reduce transmission of multidrug-resistant gram-negative bacteria in hospitalized patients. Clin Microbiol Infect. 2014;20(Suppl 1):1–55. doi: 10.1111/1469-0691.12427. [DOI] [PubMed] [Google Scholar]
  • 134.Ruhnke M, Arnold R, Gastmeier P. Infection control issues in patients with hematological malignancies in the era of multidrug-resistant bacteria. Lancet Oncol. 2014;15:e609–616. doi: 10.1016/S1470-2045(14)70344-4. [DOI] [PubMed] [Google Scholar]
  • 135.Gyssens IC, Kern WV, Livermore DM, et al. The role of antibiotic stewardship in limiting antibacterial resistance among hematology patients. Haematologica. 2013;98:1821–1825. doi: 10.3324/haematol.2013.091769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Kochar S, Sheard T, Sharma R, et al. Success of an infection control program to reduce the spread of carbapenem-resistant Klebsiella pneumoniae. Infect Control Hosp Epidemiol. 2009;30:447–452. doi: 10.1086/596734. [DOI] [PubMed] [Google Scholar]
  • 137.Almyroudis NG, Osawa R, Samonis G, et al. Discontinuation of systemic surveillance and contact precautions for vancomycin-resistant enterococcus (VRE) and its impact on the incidence of VRE faecium bacteremia in patients with hematologic malignancies. Infect Control Hosp Epidemiol. 2016;37:398–403. doi: 10.1017/ice.2015.310. [DOI] [PubMed] [Google Scholar]
  • 138.Bar-Yoseph H, Hussein K, Braun E, Paul M. Natural history and decolonization strategies for ESBL/carbapenem-resistant Enterobacteriaceae carriage: Systematic review and meta-analysis. J Antimicrob Chemother. 2016;71:2729–239. doi: 10.1093/jac/dkw221. [DOI] [PubMed] [Google Scholar]
  • 139.Halaby T, Al Naiemi N, Kluytmans J, van der Palen J, Vandenbroucke-Grauls CM. Emergence of colistin resistance in Enterobacteriaceae after the introduction of selective digestive tract decontamination in an intensive care unit. Antimicrob Agents Chemother. 2013;57:3224–3229. doi: 10.1128/AAC.02634-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Oren I, Sprecher H, Finkelstein R, et al. Eradication of carbapenem-resistant Enterobacteriaceae gastrointestinal colonization with nonabsorbale oral antibiotic treatment: A prospective controlled trial. Am J Infect Control. 2013;41:1167–1172. doi: 10.1016/j.ajic.2013.04.018. [DOI] [PubMed] [Google Scholar]
  • 141.Zuckerman T, Benyamini N, Sprecher H, et al. SCT in patients with carbapenem-resistant Klebsiella pneumoniae: A single center experience with oral gentamicin for eradication of the carrier state. Bone Marrow Transplant. 2011;46:1226–1230. doi: 10.1038/bmt.2010.279. [DOI] [PubMed] [Google Scholar]
  • 142.Vehreschild MJ, Hamprecht A, Peterson L, et al. A multicenter cohort study on colonization and infection with ESBL-producing Enterobacteriaceae in high-risk patients with hematologic malignancies. J Antimicrob Chemother. 2014;69:3387–3392. doi: 10.1093/jac/dku305. [DOI] [PubMed] [Google Scholar]
  • 143.Cornejo-Juarez P, Suarez-Cuenca JA, Volkow-Fernandez P, et al. Fecal ESBL Escherichia coli carriage as a risk factor for bacteremia in patients with hematological malignancies. Support Care Cancer. 2016;24:253–259. doi: 10.1007/s00520-015-2772-z. [DOI] [PubMed] [Google Scholar]
  • 144.Satlin M, Chavda K, Jenkins S, et al. Abstract #626, IDWeek. San Diego, CA: 2015. Colonization with extended-spectrum beta-lactamase-producing Enterobacteriaceae and subsequent risk of bacteremia in neutropenic patients undergoing hematopoietic stem cell transplantation. [Google Scholar]
  • 145.Nesher L, Rolston KV, Shah DP, et al. Fecal colonization and infection with Pseudomonas aeruginosa in recipients of allogeneic hematopoietic stem cell transplantation. Transpl Infect Dis. 2015;17:33–38. doi: 10.1111/tid.12323. [DOI] [PubMed] [Google Scholar]
  • 146.Li L, Soave R, Small C, et al. Abstract #2322, IDWeek 2016. New Orleans, LA: 2016. Colonization with vancomycin-resistant enterococci and subsequent risk of bacteremia in hematopoietic stem cell transplant recipients. [Google Scholar]
  • 147.Zasowski EJ, Claeys KC, Lagnf AM, Davis SL, Rybak MJ. Time is of the essence: The impact of delayed antibiotic therapy on patient outcomes in hospital-onset enterococcal bloodstream infections. Clin Infect Dis. 2016;62:1242–1250. doi: 10.1093/cid/ciw110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Blaschke AJ, Heyrend C, Byington CL, et al. Rapid identification of pathogens from positive blood cultures by multiplex polymerase chain reaction using the FilmArray system. Diagn Microbiol Infect Dis. 2012;74:349–355. doi: 10.1016/j.diagmicrobio.2012.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Samuel LP, Tibbetts RJ, Agotesku A, Fey M, Hensley R, Meier FA. Evaluation of a microarray-based assay for rapid identification of gram-positive organisms and resistance markers in positive blood cultures. J Clin Microbiol. 2013;51:1188–1192. doi: 10.1128/JCM.02982-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Tojo M, Fujita T, Ainoda Y, et al. Evaluation of an automated rapid diagnostic assay for detection of gram-negative bacteria and their drug-resistance genes in positive blood cultures. PLoS One. 2014;9:e94064. doi: 10.1371/journal.pone.0094064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Banerjee R, Teng CB, Cunningham SA, et al. Randomized trial of rapid multiplex polymerase chain reaction-based blood culture identification and susceptibility testing. Clin Infect Dis. 2015;61:1071–1080. doi: 10.1093/cid/civ447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Raad I, Hanna HA, Alakech B, Chatzinikolaou I, Johnson MM, Tarrand J. Differential time to positivity: A useful method for diagnosis catheter-related bloodstream infections. Ann Intern Med. 2004;140:18–25. doi: 10.7326/0003-4819-140-1-200401060-00007. [DOI] [PubMed] [Google Scholar]
  • 153.Mermel LA, Allon M, Bouza E, et al. Clinical practice guidelines for the diagnosis and management of intravascular catheter-related infection: 2009 Update by the Infectious Diseases Society of America. Clin Infect Dis. 2009;49:1–45. doi: 10.1086/599376. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES