Multidrug-resistant bacteria: what is the threat?

Similar documents
Appropriate antimicrobial therapy in HAP: What does this mean?

Mono- versus Bitherapy for Management of HAP/VAP in the ICU

Fighting MDR Pathogens in the ICU

Duke University Hospital Guideline for Empiric Inpatient Treatment of Cancer- Related Neutropenic Fever in Adult Patients

4/3/2017 CLINICAL PEARLS: UPDATES IN THE MANAGEMENT OF NOSOCOMIAL PNEUMONIA DISCLOSURE LEARNING OBJECTIVES

Update on Resistance and Epidemiology of Nosocomial Respiratory Pathogens in Asia. Po-Ren Hsueh. National Taiwan University Hospital

Antimicrobial stewardship in managing septic patients

Safe Patient Care Keeping our Residents Safe Use Standard Precautions for ALL Residents at ALL times

The International Collaborative Conference in Clinical Microbiology & Infectious Diseases

PRACTIC GUIDELINES for APPROPRIATE ANTIBIOTICS USE

Antibiotic Updates: Part II

Northwestern Medicine Central DuPage Hospital Antimicrobial Criteria Updated 11/16/16

Bacterial Resistance in Haematology-ECIL 4 Study Groups & Participants

Antimicrobial Cycling. Donald E Low University of Toronto

OPTIMIZATION OF PK/PD OF ANTIBIOTICS FOR RESISTANT GRAM-NEGATIVE ORGANISMS

Outline. Antimicrobial resistance. Antimicrobial resistance in gram negative bacilli. % susceptibility 7/11/2010

Dr. Shaiful Azam Sazzad. MD Student (Thesis Part) Critical Care Medicine Dhaka Medical College

Other Beta - lactam Antibiotics

UCSF guideline for management of suspected hospital-acquired or ventilatoracquired pneumonia in adult patients

Multi-drug resistant microorganisms

MICRONAUT MICRONAUT-S Detection of Resistance Mechanisms. Innovation with Integrity BMD MIC

Successful stewardship in hospital settings

SHC Clinical Pathway: HAP/VAP Flowchart

Infectious Disease Issues in the Intensive Care Unit

Learning Points. Raymond Blum, M.D. Antimicrobial resistance among gram-negative pathogens is increasing

Sepsis is the most common cause of death in

New Drugs for Bad Bugs- Statewide Antibiogram

PIPERACILLIN- TAZOBACTAM INJECTION - SUPPLY PROBLEMS

What does multiresistance actually mean? Yohei Doi, MD, PhD University of Pittsburgh

Recommendations for Implementation of Antimicrobial Stewardship Restrictive Interventions in Acute Hospitals in Ireland

The Inpatient Management of Febrile Neutropenia

Combination vs Monotherapy for Gram Negative Septic Shock

Taiwan Crit. Care Med.2009;10: %

Summary of unmet need guidance and statistical challenges

Multidrug-Resistant Organisms: How Do We Define them? How do We Stop Them?

Other β-lactamase Inhibitor (BLI) Combinations: Focus on VNRX-5133, WCK 5222 and ETX2514SUL

The β- Lactam Antibiotics. Munir Gharaibeh MD, PhD, MHPE School of Medicine, The University of Jordan November 2018

Witchcraft for Gram negatives

Konsequenzen für Bevölkerung und Gesundheitssysteme. Stephan Harbarth Infection Control Program

EARS Net Report, Quarter

Management of Hospital-acquired Pneumonia

Antibiotic Resistance. Antibiotic Resistance: A Growing Concern. Antibiotic resistance is not new 3/21/2011

Nosocomial Infections: What Are the Unmet Needs

Antimicrobial stewardship: Quick, don t just do something! Stand there!

Escherichia Coli: an Important Pathogen in Patients with Hematologic Malignancies

2012 ANTIBIOGRAM. Central Zone Former DTHR Sites. Department of Pathology and Laboratory Medicine

Antimicrobial Stewardship 101

These recommendations were approved for use by the Pharmaceutical and Therapeutics Committee, RCWMCH on 1 February 2017.

Rise of Resistance: From MRSA to CRE

Original Articles. K A M S W Gunarathne 1, M Akbar 2, K Karunarathne 3, JRS de Silva 4. Sri Lanka Journal of Child Health, 2011; 40(4):

GUIDE TO INFECTION CONTROL IN THE HOSPITAL. Antibiotic Resistance

Antimicrobial Susceptibility Patterns

Intrinsic, implied and default resistance

DR. MICHAEL A. BORG DIRECTOR OF INFECTION PREVENTION & CONTROL MATER DEI HOSPITAL - MALTA

National Clinical Guideline Centre Pneumonia Diagnosis and management of community- and hospital-acquired pneumonia in adults

RESISTANT PATHOGENS. John E. Mazuski, MD, PhD Professor of Surgery

CF WELL Pharmacology: Microbiology & Antibiotics

UCSF Medical Center Guidelines for Inpatient Management of Febrile Neutropenia

Breaking the Ring. β-lactamases and the Great Arms Race. Bryce M Kayhart, PharmD, BCPS PGY2 Pharmacotherapy Resident Mayo Clinic - Rochester

Bacterial infections complicating cirrhosis

What s next in the antibiotic pipeline?

Suggestions for appropriate agents to include in routine antimicrobial susceptibility testing

* gender factor (male=1, female=0.85)

ETX2514SUL (sulbactam/etx2514) for the treatment of Acinetobacter baumannii infections

Antibacterials. Recent data on linezolid and daptomycin

Initial Management of Infections in the Era of Enhanced Antimicrobial Resistance

Florida Health Care Association District 2 January 13, 2015 A.C. Burke, MA, CIC

Disclosures. Principles of Antimicrobial Therapy. Obtaining an Accurate Diagnosis Obtain specimens PRIOR to initiating antimicrobials

Summary of the latest data on antibiotic resistance in the European Union

Misericordia Community Hospital (MCH) Antimicrobial Stewardship Report. July December 2013 Second and Third Quarters 2014

2015 Antibiotic Susceptibility Report

48 th Annual Meeting. IDWeek and ICAAC: The Cliffs Notes Version. Skin and Soft Tissue Infections. Skin and Soft Tissue Infections.

Rational use of antibiotics

Int.J.Curr.Microbiol.App.Sci (2017) 6(3):

Resistant Gram-negative Bacteria

2016 Antibiotic Susceptibility Report

Mercy Medical Center Des Moines, Iowa Department of Pathology. Microbiology Department Antibiotic Susceptibility January December 2016

How is Ireland performing on antibiotic prescribing?

Concise Antibiogram Toolkit Background

Approach to pediatric Antibiotics

Mechanism of antibiotic resistance

Presenter: Ombeva Malande. Red Cross Children's Hospital Paed ID /University of Cape Town Friday 6 November 2015: Session:- Paediatric ID Update

High-Risk MDR clones news in treatment

Antimicrobial Stewardship Strategy: Antibiograms

Antimicrobial Pharmacodynamics

The role of carbapenems in the hospital

GENERAL NOTES: 2016 site of infection type of organism location of the patient

Considerations in antimicrobial prescribing Perspective: drug resistance

Changing trends in clinical characteristics and antibiotic susceptibility of Klebsiella pneumoniae bacteremia

ETX0282, a Novel Oral Agent Against Multidrug-Resistant Enterobacteriaceae

Collecting and Interpreting Stewardship Data: Breakout Session

General Approach to Infectious Diseases

Does Screening for MRSA Colonization Have A Role In Healthcare-Associated Infection Prevention Programs?

LEARNING OBJECTIVES ANTIMICROBIAL USES AND ABUSES INFECTIOUS DISEASE SCARES

INCIDENCE OF BACTERIAL COLONISATION IN HOSPITALISED PATIENTS WITH DRUG-RESISTANT TUBERCULOSIS

Surveillance of Antimicrobial Resistance among Bacterial Pathogens Isolated from Hospitalized Patients at Chiang Mai University Hospital,

Le infezioni di cute e tessuti molli

Infection Control of Emerging Diseases

2017 Introduction to Infectious Diseases Clinical Seminar Saturday 30th September - Sunday 1st October 2017 Hotel Grand Chancellor Hobart, Tasmania

Defining Extended Spectrum b-lactamases: Implications of Minimum Inhibitory Concentration- Based Screening Versus Clavulanate Confirmation Testing

Transcription:

INFECTIOUS DISEASE COMPLICATIONS ENCOUNTERED BY THE PRACTICING HEMATOLOGIST Multidrug-resistant bacteria: what is the threat? Matteo Bassetti 1 and Elda Righi 1 1 Infectious Diseases Division, Santa Maria Misericordia University Hospital, Udine, Italy Despite big advances in antimicrobial therapies and infection strategies, the emergence of antibiotic resistance represents an emergency situation, especially in immunocompromised hosts. Specifically, infections due to multidrug resistant, gram-negative pathogens are responsible for high mortality rates and may leave few effective antimicrobial options. Furthermore, although new compounds are available for severe methicillin-resistant staphylococcal infections, there is a paucity of novel classes of antimicrobials to target resistant gram-negatives. A careful assessment of the clinical conditions and underlying comorbidities, along with knowledge about the previous history of colonization or infections due to multidrug-resistant bacteria, represent key points in approaching the hematological patient with signs of infection. A de-escalation therapy with initial use of wide-spectrum antimicrobials followed by a reassessment after 72 hours of treatment may represent a good option in severe infections if a resistant pathogen is suspected. Prompt empiric or targeted therapy using combination regimens (ie, antipseudomonal beta-lactam plus an aminoglycoside or a quinolone) with the addition of colistin, along with increased dosage and therapeutic drug monitoring, represent options for these life-threatening infections. Continuous epidemiological surveillance of local bacteremias is necessary, along with stringent enforcement of antibiotic stewardship programs in cancer patients. Introduction Reemergence of gram-negative infections and increased antimicrobial resistance due to overuse of antibiotics in cancer patients have changed the epidemiology of bacteremia in neutropenic patients in the past decade 1-4 Classically, gram-positive organisms have been the predominant bacterial pathogens in this setting. 5,6 Among gram-negatives, which are isolated in up to 80% of polymicrobial bacteremias, Escherichia coli, Klebsiella spp., and Pseudomonas aeruginosa remain the predominant species, although other Enterobacteriaceae, Stenotrophomonas maltophilia, and Acinetobacter spp. can also be isolated. 7-9 Of particular challenge are the infections due to methicillin-resistant Staphylococcus aureus (MRSA), representing up to 50% of all staphylococcal infections; vancomycin-resistant enterococci (VRE), representing nearly 30% in the United States; extended-spectrum beta-lactamase (ESBL) producing gram-negative bacteria; and other multidrug-resistant (MDR) organisms such as A. baumannii, Klebsiella pneumoniae carbapenem-resistant (KPC), P aeruginosa, and fluoroquinoloneresistant E coli 10-12 (Table 1). Although recent clinical practice guidelines recommend prophylaxis with ciprofloxacin or levofloxacin during profound neutropenia in acute leukemia and hematopoietic stem cell transplantation, their widespread use has been linked with a rising prevalence of MDR pathogens including gram-negatives, MRSA, and VRE. 3,4 The use of inappropriate antibiotic regimens and, consequently, the delay in starting appropriate treatment account is considered the main cause for poor outcomes in severe infections, especially among human stem cell transplantation recipients during the early posttransplant period. 13,14 The identification of risk factors associated with the development of MDR infection is important to guide the strategies that may decrease their adverse impact. 15 Furthermore, different factors can affect in diverse ways the development of infections in hematological patients, including the duration and the severity of granulocytopenia, the mechanical defence damage (such as the presence of mucositis, use of central venous catheters [CVCs], tumor infiltration), and the reactivation of previous infections due to immunosuppression. Therefore, the initial approach to hematological patients with a suspected drug-resistant infection includes the evaluation of underlying disease, the severity and the duration of the immunodeficiency, the presence of additional predisposing factors to infections, and the severity of clinical presentation. Bloodstream infections (BSIs, including CVC-related BSIs), pneumonia, and mucositis represent the majority of infectious sites. In this setting, prompt initiation of an appropriate empiric therapy, reassessment after the availability of susceptibility test, and the continuation of a targeted therapy taking into account the patient comorbidities, type and site of the infection, and pathogen resistances may contribute to a reduction of the mortality. Below we discuss the different approaches and the antimicrobial strategies for empiric and targeted therapies in hematological patients presenting with severe infections. How to treat resistant infections in hematological patients 1. Initial clinical assessment and risk factors for resistant infections In the selection of an antimicrobial therapy to approach a patient with febrile neutropenia, of key importance is a knowledge of the epidemiology and resistance patterns at each single institution. Also of paramount importance is the obtainment of a detailed medical history of the patient s colonization and the documentation of previous infections caused by resistant pathogens. A patient s age, comorbidities, duration of aplasia, and complications (eg, bleeding, dehydration, liver and kidney failure) should be assessed, along with the clinical parameters that are evocative of a severe infectious process (septic shock or hemodynamic instability, 428 American Society of Hematology

Table 1. Main resistant organisms, mechanism of resistance, and drugs not to be used Resistant organism Mechanism of resistance Drugs not to be used ESBL -lactamases Penicillins, cephalosporins, monobactams KPC Carbapenemase Penicillins, cephalosporins, monobactams, standard-dose carbapenems NDM Metallo- -lactamase enzymes Penicillins, cephalosporins, monobactams, carbapenems sensory loss) and a localized infection (pneumonia, enteritis, catheter-related infection). Risk factors including known colonization with resistant bacteria, previous infections, or exposure to broad-spectrum antibiotics (ie, beta-lactams and quinolones), advanced underlying disease with severe clinical presentation, prolonged hospital stay, and catheter placement (ie, urinary catheters for ESBL-associated infections and CVCs for MRSA) should be taken into consideration during the initial approach to high-risk hematological patients (Table 2). 2. Choice of empiric therapy regimens A classic approach is represented by escalation therapy consisting of an initial empiric monotherapy using a beta-lactam (eg, ceftazidime, cefepime, or piperacillin-tazobactam) targeting Enterobacteriaceae and P aeruginosa infections without ESBL or carbapenemaseproducing gram-negative coverage. If clinical deterioration or isolation of a resistant pathogen occurs, then therapy is escalated using an antibiotic with a broader spectrum or a combination of antimicrobials. This approach may be inappropriate in the following instances: compromised patients with severe presentation; a hospital setting in which a high prevalence of ESBL-producing bacteria ( 20%) is reported; and patients with previous history of colonization or infection due to resistant pathogens (Table 3) or presenting with risk factors for resistances. In these cases, the escalation approach may lead to an initial inadequate antimicrobial treatment with a potentially negative impact on the outcome. In these cases, another approach is represented by de-escalation therapy starting from a very broad initial regimen possibly covering highly resistant pathogens and followed (after a reassessment at 48-72 hours) by a step-down therapy upon availability of susceptibility tests. Antimicrobials used in these regimens include monotherapy with a carbapenem or antimicrobial associations targeting gram-negative pathogens (eg, colistin with a beta-lactam or an aminoglycoside plus a beta-lactam) and gram-positive resistant bacteria, if specific risk factors are identified. Fluoroquinolones are recommended as a possible part of a combination therapy only in patients who were not Table 2. Risk factors for development of resistant infections in hematological patients 16-18 Colonization with a resistant bacteria (MRSA, VRE, ESBL, or carbapenemase-producing Enterobacteriaceae)* Previous infection due to resistant bacteria documented by susceptibility tests Previous exposure to broad-spectrum antibiotics (eg, third-generation cephalosporins, carbapenems) and/or antimicrobial prophylaxis with quinolones Advanced underlying disease with severe clinical presentation Prolonged hospital stay Urinary catheter and CVC placement, mechanical ventilation, or tracheostomy *See Table 3 for therapeutic suggestions. receiving fluoroquinolone-based prophylactic regimens. Early coverage of resistant gram-positives using a glycopeptide or newer agents (eg, linezolid or daptomycin) should be used if risk factors for gram-positive infections, such as CVC placement, mucositis, or soft tissue involvement are present. Although characterized by high efficacy as an empiric first-line treatment of febrile neutropenia, carbapenems should be avoided in uncomplicated patients lacking risk factors for resistant bacteria to preserve their activity for seriously ill patients. Nevertheless, centers with reported high rates of resistance due to ESBL production ( 20%) or carbapenemase-producing organisms should undergo a prompt infection control review. Details regarding the indications, the antimicrobial options for a de-escalation approach, and the suggested dosages are summarized in Tables 4 and 5. In general, a case-by-case adjustment of the initial regimen should be considered after 24 to 72 hours according to the clinical course and the microbiological results. After the start of an escalation therapy, there may be no availability of microbiological documentation. In this case, the antimicrobial therapy should be changed if the patient presents persistent fever in association with deteriorating clinical conditions. Furthermore, investigation of the infectious source through repeated microbiological cultures, imaging (CT scan, MRI), and nonculture tests (eg, galactomannan, beta-glucan) is warranted. The new empiric regimen should include resistant gram-negative pathogens and should be based on the documentation of previous infections, prior use of wide-spectrum antimicrobials, and patient risk factors. Combination therapies including colistin and/or carbapenems may represent valid options. In cases requiring a de-escalation approach without microbiological tests available, the antimicrobial regimen should be reassessed after 48 to 72 hours and changed as follows. In cases of clinical stability, a narrower-spectrum agent is preferred. To avoid renal toxicity and selection of resistances, aminoglycosides and quinolones should be discontinued. In cases of persistent fever or clinical deterioration, the presence of a resistant organism [eg, carbapenem-resistant P aeruginosa, KPC, New-Delhi metallo-beta-lactamase (NDM-1) E coli] or the occurrence of an infection due to MRSE, VRE, or a fungal infection should be suspected. Addition of colistin to a carbapenem monotherapy and anti-gram-positive agents may be performed, along with adequate imaging and nonculture tests to detect possible fungal infections. 3. Targeted therapy for severe infections in hematological patients If a bacterial infection is documented, the results of the in vitro susceptibility tests, including minimum inhibitory concentration (MIC) when possible, should be used to guide the appropriate antibiotic regimen. Nevertheless, depending on the clinical indication and the site of the infection, agents with the less broad spectrum should be used when the same in vitro susceptibility is detected for Hematology 2013 429

Table 3. Modification of the initial therapy due to colonization/previous infection with resistant bacteria Resistant bacteria Primary choice Secondary choice MRSA Daptomycin or linezolid Vancomycin VRE Daptomycin or linezolid Tigecycline ESBL Meropenem Ertapenem Carbapenemase-producing High-dose meropenem (6 g/d) tigecycline Tigecycline gentamicin colistin Enterobacteriaceae colistin MDR P aeruginosa Piperacillin-tazobactam amikacin Piperacillin-tazobactam colistin MDR Acinetobacter baumannii Colistin rifampin Tigecycline colistin Stenotrophomonas maltophilia Trimetoprim-sulfamethoxazole Quinolone or tigecycline more than one compound to prevent resistances. If MDR pathogens are detected, consultation with an infectious disease expert or clinical microbiologist is always recommended. Furthermore, because hematological patients often present specific pathophysiological conditions that may alter the pharmacokinetic behavior of antimicrobials, different administration schedule and antimicrobial dosing regimens from the standard ones may be required. Specifically, increased dose (eg, aminoglycosides, fluoroquinolones, daptomycin), prolonged infusion (eg, beta-lactams), and therapeutic drug monitoring are important to optimize the antimicrobial therapy. A summary of the suggested antimicrobial schedule doses for severe infections in hematological patients is provided in Table 4. Among MDR infections, particularly challenging are the infections due to difficult-to-treat MDR gram-negatives such as P aeruginosa and K pneumoniae (including KPC) because of the limited antimicrobial options available, along with reported high mortality rates. 19 In this context, the role of combination therapy has been advocated to provide broader coverage, prevent emergence of reduced susceptibility, and achieve bacterial synergy. 20-22 In pseudomonal infections, an initial combination therapy significantly reduced the likelihood of inappropriate therapy, although similar outcomes are reached using a targeted therapy once the susceptibility is documented. 23 Conversely, association of at least 2 active compounds (eg, colistin and high doses of meropenem, 6 g/d, with tigecycline or gentamycin) has been linked to increased survival rates in severe KPC-related infections. 21,24 Involvement of gram-positive pathogens is common in CVC-related infections and pneumonia. In these instances, antistaphylococcal drugs should include those with activities toward MRSA. Nevertheless, the widespread use of vancomycin has been related to the emergence of strains of MRSA with increased MIC to vancomycin ( 1 mg/l) causing severe infections. 25 Particularly, low eradication rates and increased mortality have been documented in MRSA-related BSIs and pneumonia when high MIC values to vancomycin were reported. 26-28 Daptomycin is highly active against MRSA BSIs and has shown to affect outcomes in cancer patients with BSIs, causing a faster resolution of symptoms compared with vancomycin. 29 In cases of MRSA nosocomial pneumonia, linezolid has exhibited a better performance than vancomycin with regard to clinical efficacy and pathogen eradication and can be a valid choice for an empiric choice, although limitations can be posed due to its BM suppression in hematological patients. 30 Conclusions MDR infections pose a real threat to immunocompromised patients, causing severe infections with poor outcomes. The increase in infections caused by gram-negative and gram-positive resistant bacteria is concerning for the success of empiric treatment of febrile neutropenia. Immediate evaluation, risk assessment, and treatment with empiric broad-spectrum antibiotics are warranted when a resistant infection is suspected in hematological patients with severe immune impairment. Reassessment after 48 to 72 hours from the beginning of an empiric treatment should always be done to avoid an unnecessary use of wide-spectrum antibiotics that are not required or to choose an appropriate targeted therapy in case of documented multiple resistances. Nevertheless, efforts to reduce the risk of febrile neutropenia should be based on appropriate antimicrobial stewardship and infection control policies. Disclosures Conflict-of-interest disclosure: M.B. has received research funding from MSD, Novartis, and Astellas; has consulted for Vifor, Trius, Bayer, MSD, Novartis, Astellas, Pfizer, Roche, and Basilea; has received honoraria from Gilead, Bayer, MSD, Novartis, Astellas, AstrazZeneca, and Pfizer; and has been affiliated with the speakers Table 4. Indications and antimicrobial options for de-escalation therapy in patients with complicated presentation (severely ill, septic shock) Situations/indications Known colonization or previous infection with ESBL-producing Enterobacteriaceae; gram-negatives resistant to narrowerspectrum beta-lactams; centers with a high prevalence ( 20%) of infections due to ESBL-producers Probable infection with resistant nonfermenter pathogens (P aeruginosa or Acinetobacter spp) based on local epidemiology, previous colonization or infection, carbapenem use during the month prior to infection MRSA or VRE colonization; probable CVC-related infection; skin or soft tissue infection (cellulitis) Antimicrobial options Meropenem or ertapenem; secondary option (not for bloodstream infections and pneumonia) tigecycline Piperacillin/tazobactam plus amikacin; colistin piperacillin/tazobactam or meropenem ( rifampicin) Addition of daptomycin, linezolid, or vancomycin* *Not to be used if VRE colonization is documented. 430 American Society of Hematology

Table 5. Recommendations of the dosage of the most common antimicrobials used for infections due to resistant pathogens Compound Amikacin Ceftazidime Cefepime Colistin Daptomycin Ertapenem Gentamicin Linezolid Meropenem Piperacillin/tazobactam Rifampin Tigecycline Trimetoprim-sulfamethoxazole Vancomycin c.i. indicates administered by continuous infusion. Administration schedule 1500-2000 mg/d every 24 h 6 g every 24 h c.i. 2 g every 8horc.i. 9 MU loading dose, 4.5 MU every 12 h 8-10 mg/kg/d every 24 h 1 g every 12 h 7 mg/kg/d every 24 h 600 mg every 12 h 1-2 g every 6-8 h in extended infusion (4 h) 4.5 g every 6 h c.i. 600-900 mg every 24 h 100-200 mg loading those, then 50-100 mg every 12 h 10 mg/kg/d divided q6h 1 g loading dose, then 2 g every 24 h c.i. bureau for Vifor, Gilead, Bayer, MSD, Novartis, Astellas, Astra- Zeneca, and Pfizer. E.R. declares no competing financial interests. Off-label drug use: None disclosed. Correspondence Matteo Bassetti, MD, PhD, Clinica Malattie Infettive, Azienda Ospedaliera Universitaria Santa Maria della Misericordia, Piazzale Santa Maria della Misericordia 15, 33100 Udine, Italy; Phone: 39-0432-559355; Fax: 39-0432-559360; e-mail: mattba@tin.it. References 1. Rolston KV. Challenges in the treatment of infections caused by gram-positive and gram-negative bacteria in patients with cancer and neutropenia. Clin Infect Dis. 2005;40:S246-S252. 2. Montassier E, Batard E, Gastinne T, et al. Recent changes in bacteremia in patients with cancer: a systematic review of epidemiology and antibiotic resistance. Eur J Clin Microbiol Infect Dis. 2013;32(7):841-850. 3. Bow EJ. Fluoroquinolones, antimicrobial resistance and neutropenic cancer patients. Curr Opin Infect Dis. 2011;24(6):545-553. 4. Gudiol C, Bodro M, Simonetti A, et al. Changing aetiology, clinical features, antimicrobial resistance, and outcomes of bloodstream infection in neutropenic cancer patients. Clin Microbiol Infect. 2013;19(5):474-479. 5. Wisplinghoff H, Seifert H, Wenzel RP, et al. Current trends in the epidemiology of nosocomial bloodstream infections in patients with hematological malignancies and solid neoplasms in hospitals in the United States. Clin Infect Dis. 2003;36(9): 1103-1110. 6. Yadegarynia D, Tarrand J, Raad I, et al. Current spectrum of bacterial infections in patients with cancer. Clin Infect Dis. 2003;37(8):1144-1145. 7. Rolston KV, Yadegarynia D, Kontoyiannis DP, et al. The spectrum of Gram-positive bloodstream infections in patients with hematologic malignancies, and the in vitro activity of various quinolones against Gram-positive bacteria isolated from cancer patients. Int J Infect Dis. 2006;10(3):223-230. 8. Rolston KV, Bodey GP, Safdar A: Polymicrobial infection in patients with cancer: an underappreciated and underreported entity. Clin Infect Dis. 2007;45(2):228-233. 9. Klastersky J, Ameye L, Maertens J, et al. Bacteraemia in febrile neutropenic cancer patients. Int J Antimicrob Agents. 2007; 30(Suppl 1):S51-S59. 10. Satlin MJ, Calfee DP, Chen L, et al. Emergence of carbapenemresistant Enterobacteriaceae as causes of bloodstream infections in patients with hematologic malignancies. Leuk Lymphoma. 2013;54(4):799-806. 11. Quilty S, Kwok G, Hajkowicz K, et al. High incidence of methicillin-resistant Staphylococcus aureus sepsis and death in patients with febrile neutropenia at Royal Darwin Hospital. Intern Med J. 2009;39(8):557-559. 12. Bow EJ. There should be no ESKAPE for febrile neutropenic cancer patients: the dearth of effective antibacterial drugs threatens anticancer efficacy. J Antimicrob Chemother. 2013; 68(3):492-495. 13. Cosgrove SE. The relationship between antimicrobial resistance and patient outcomes: mortality, length of hospital stay, and health care costs. Clin Infect Dis. 2006;42:S82-89. 14. Ramphal R. Changes in the etiology of bacteremia in febrile neutropenic patients and the susceptibilities of the currently isolated pathogens. Clin Infect Dis. 2004;39:S25-S31. 15. Cordonnier C, Herbrecht R, Buzyn A, et al. Risk factors for Gram-negative bacterial infections in febrile neutropenia. Haematologica. 2005;90(8):1102-1109. 16. Sahin E, Ersoz G, Goksu M, et al. Risk factors for hospital infections with multidrug-resistant bacteria in patients with cancer (abstract P1450). Presented at the 19th European Congress of Clinical Microbiology and Infectious Diseases, Helsinki, Finland, May 16-19, 2009. 17. 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(2):333-341. 18. Kang CI, Chung DR, Ko KS, et al. Risk factors for infection and treatment outcome of extended-spectrum beta-lactamaseproducing Escherichia coli and Klebsiella pneumoniae bacteremia in patients with hematologic malignancy. Ann Hematol. 2012;91(1):115-121. 19. Aubron C, Poirel L, Fortineau N, et al. Nosocomial spread of Pseudomonas aeruginosa isolates expressing the metallo-betalactamase VIM-2 in a hematology unit of a French hospital. Microb Drug Resist. 2005;11(3):254-259. 20. Martinez JA, Cobos-Trigueros N, Soriano A, et al. Influence of empiric therapy with a beta-lactam alone or combined with an aminoglycoside on prognosis of bacteremia due to gramnegative microorganisms. Antimicrob Agents Chemother. 2010; 54(9):3590-3596. 21. Zarkotou O, Pournaras S, Tselioti P, et al. Predictors of mortality in patients with bloodstream infections caused by KPC-producing Klebsiella pneumoniae and impact of appropriate antimicrobial treatment. Clin Microbiol Infect. 2011;17(12): 1798-1803. 22. Heyland DK, Dodek P, Muscedere J, et al. Randomized trial of combination versus monotherapy for the empiric treatment of suspected ventilator-associated pneumonia. Crit Care Med. 2008;36(3):737-744. 23. Garnacho-Montero J, Sa-Borges M, Sole-Violan J, et al. Optimal management therapy for Pseudomonas aeruginosa ventilator-associated pneumonia: an observational, multicenter Hematology 2013 431

study comparing monotherapy with combination antibiotic therapy. Crit Care Med. 2007;35(8):1888-1895. 24. Tumbarello M, Viale P, Viscoli C, et al. Predictors of mortality in bloodstream infections caused by Klebsiella pneumoniae carbapenemase-producing K. pneumoniae: importance of combination therapy. Clin Infect Dis. 2012;55(7):943-950. 25. Dhand A, Sakoulas G. Reduced vancomycin susceptibility among clinical Staphylococcus aureus isolates ( the MIC Creep ): implications for therapy. F1000 Med Rep. 2012;4:4. 26. Haque NZ, Zuniga LC, Peyrani P, et al. Relationship of vancomycin minimum inhibitory concentration to mortality in patients with methicillin-resistant Staphylococcus aureus hospital-acquired, ventilator-associated, or health-care-associated pneumonia. Chest. 2010;138(6):1356-1362. 27. Bassetti M, Mesini A, Molinari MP, et al. Vancomycin susceptibility of methicillin-resistant Staphylococcus aureus (MRSA) bacteraemia isolates in an Italian hospital. Int J Antimicrob Agents. 2011;38(5):453-454. 28. Moise PA, Sakoulas G, Forrest A, et al. Vancomycin in vitro bactericidal activity and its relationship to efficacy in clearance of methicillin-resistant Staphylococcus aureus bacteremia. Antimicrob Agents Chemother. 2007;51(7):2582-2586. 29. Chaftari AM, Hachem R, Mulanovich V, et al. Efficacy and safety of daptomycin in the treatment of Gram-positive catheterrelated bloodstream infections in cancer patients. Int J Antimicrob Agents. 2010;36(2):182-186. 30. Wunderink RG, Niederman MS, Kollef MH, et al. Linezolid in methicillin-resistant Staphylococcus aureus nosocomial pneumonia: a randomized, controlled study. Clin Infect Dis. 2012; 54(5):621-629. 432 American Society of Hematology