Sei sulla pagina 1di 18

Postgraduate Medicine

ISSN: 0032-5481 (Print) 1941-9260 (Online) Journal homepage: http://www.tandfonline.com/loi/ipgm20

An update on the management of urinary tract


infections in the era of antimicrobial resistance

Mazen S Bader, Mark Loeb & Annie A Brooks

To cite this article: Mazen S Bader, Mark Loeb & Annie A Brooks (2016): An update on the
management of urinary tract infections in the era of antimicrobial resistance, Postgraduate
Medicine, DOI: 10.1080/00325481.2017.1246055

To link to this article: http://dx.doi.org/10.1080/00325481.2017.1246055

Accepted author version posted online: 07


Oct 2016.
Published online: 21 Oct 2016.

Submit your article to this journal

Article views: 62

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=ipgm20

Download by: [Cornell University Library] Date: 22 October 2016, At: 11:33
POSTGRADUATE MEDICINE, 2017
http://dx.doi.org/10.1080/00325481.2017.1246055

CLINICAL FEATURE
REVIEW

An update on the management of urinary tract infections in the era of antimicrobial


resistance
Mazen S Badera, Mark Loebb and Annie A Brooksc
a
Department of Medicine, Juravinski Hospital and Cancer Centre, Hamilton, Ontario, Canada; bDepartments of Pathology & Molecular Medicine and
Clinical Epidemiology & Biostatistics, McMaster University Michael G DeGroote School of Medicine, Hamilton, Ontario, Canada; cDepartment of
Pharmacy, Hamilton Health Sciences, Juravinski hospital and Cancer Centre, Hamilton, Ontario, Canada

ABSTRACT ARTICLE HISTORY


Urinary tract infections (UTIs) caused by antibiotic-resistant Gram-negative bacteria are a growing Received 10 July 2016
concern due to limited therapeutic options. Gram-negative bacteria, specifically Enterobacteriaceae, Accepted 5 October 2016
are common causes of both community-acquired and hospital acquired UTIs. These organisms can Published online
21 October 2016
acquire genes that encode for multiple antibiotic resistance mechanisms, including extended-spectrum-
lactamases (ESBLs), AmpC- β -lactamase, and carbapenemases. The assessment of suspected UTI KEYWORDS
includes identification of characteristic symptoms or signs, urinalysis, dipstick or microscopic tests, Antibiotic resistance; cystitis;
and urine culture if indicated. UTIs are categorized according to location (upper versus lower urinary Enterobacteriaceae;
tract) and severity (uncomplicated versus complicated). Increasing rates of antibiotic resistance neces- Gram-negative bacteria;
sitate judicious use of antibiotics through the application of antimicrobial stewardship principles. pyelonephritis; urinary tract
Knowledge of the common causative pathogens of UTIs including local susceptibility patterns are infections
essential in determining appropriate empiric therapy. The recommended first-line empiric therapies
for acute uncomplicated bacterial cystitis in otherwise healthy adult nonpregnant females is a 5-day
course of nitrofurantion or a 3-g single dose of fosfomycin tromethamine. Second-line options include
fluoroquinolones and β-lactams, such as amoxicillin-clavulanate. Current treatment options for UTIs due
to AmpC- β -lactamase-producing organisms include fosfomycin, nitrofurantion, fluoroquinolones,
cefepime, piperacillin–tazobactam and carbapenems. In addition, treatment options for UTIs due to
ESBLs–producing Enterobacteriaceae include nitrofurantion, fosfomycin, fluoroquinolones, cefoxitin,
piperacillin-tazobactam, carbapenems, ceftazidime-avibactam, ceftolozane-tazobactam, and aminogly-
cosides. Based on identification and susceptibility results, alternatives to carbapenems may be used to
treat mild-moderate UTIs caused by ESBL-producing Enterobacteriaceae. Ceftazidime-avibactam, colistin,
polymixin B, fosfomycin, aztreonam, aminoglycosides, and tigecycline are treatment options for UTIs
caused by carbapenem-resistant Enterobacteriaceae (CRE). Treatment options for UTIs caused by multi-
drug resistant (MDR)-Pseudomonas spp. include fluoroquinolones, ceftazidime, cefepime, piperacillin-
tazobactam, carbapenems, aminoglycosides, colistin, ceftazidime-avibactam, and ceftolozane-tazobac-
tam. The use of fluoroquinolones for empiric treatment of UTIs should be restricted due to increased
rates of resistance. Aminoglycosides, colistin, and tigecycline are considered alternatives in the setting
of MDR Gram-negative infections in patients with limited therapeutic options.

Introduction associated UTI (HAUTI), which include community-onset


health care-associated infections. Health care-acquired UTIs
Urinary tract infections (UTIs) are among the most common
represent 40% of all hospital-acquired UTIs [1].
bacterial infections managed by clinicians [1]. UTIs exert a
Gram-negative bacteria, specifically Enterobacteriaceae, are
considerable impact on economic and public health resources
common causes of both CAUTI and HAUTI [1]. Antibiotic resis-
and substantially affect the quality of life of those afflicted
tance among causative Gram-negative bacilli is increasing and
with recurrent infections [2]. It is estimated that 0.7% of
considered to be a challenge for clinicians since there are
ambulatory visits are attributed to UTIs, and each year 7
limited treatment options. Common examples of these organ-
million ambulatory care visits by females are due to UTIs
isms include AmpC β-lactamase- and extended-spectrum β-
[3,4]. UTIs are the fourth most common health care-associated
lactamases (ESBLs)-producing Enterobacteriaceae, carbape-
infection with a prevalence of 12.9%, and two-thirds are cathe-
nem-resistant Enterobacteriaceae (CRE), and multidrug-resis-
ter associated [5].
tant (MDR) Pseudomonas aeruginosa [7,8].
The clinical spectrums of UTIs include urethritis, cystitis,
Management of UTIs in the era of antimicrobial resistance
prostatitis, and pyelonephritis [6]. UTIs can also be stratified
requires a systematic approach to diagnose the type of infec-
as either community-associated UTI (CAUTI) or health care-
tion and to select the optimal dose, route, and duration of the

CONTACT Mazen S Bader msbader1@gmail.com Juravinski Hospital and Cancer Centre, Department of Medicine, 711 Concession Street, Hamilton, Ontario
L8V1C3, Canada
© 2016 Informa UK Limited, trading as Taylor & Francis Group
2 M. S. BADER ET AL.

antibiotic regimen. Unfortunately, standardized evidence-based Table 1. Factors associated with complicated urinary tract infections.
clinical management is often lacking in general practice [9]. Pregnancy
Diabetes mellitus
When approaching a patient with a suspected UTI, consid-
Renal failure
eration should be given to the following essential elements. Immunocompromised conditions
Kidney transplantation
Infection with antibiotic-resistant pathogens
Does the patient have a UTI? Hospital-acquired infection
Symptoms persisting ≥7 days prior to seeking medical care
Patients suspected of having UTIs should be initially evaluated Urinary tract obstruction (benign prostatic hypertrophy, stenosis, stone,
tumor, and hematoma)
through history taking and physical examination to assess
Indwelling urethral catheter, stent, nephrostomy tube, or urinary diversion
signs or symptoms consistent with symptomatic UTI and to Functional or anatomic abnormality of the urinary tract (e.g. neurogenic
guide the selection of adjunct diagnostic and therapeutic bladder)
Recent urinary tract instrumentation
strategies. Classic symptoms and signs of UTIs include fever,
acute dysuria, urinary urgency or frequency, hematuria, back
pain, suprapubic tenderness, or costovertebral angle pain or
tenderness. Laboratory tests confirming the diagnosis include with lower UTIs [6]. Urine culture is not indicated in patients
bacteriuria (≥100,000 CFU [colony forming unit]/ml) and with uncomplicated cystitis since it is not associated with
pyuria (≥10 WBC/hpf [white blood cells per high power improved outcomes such symptom scores [19]. However,
field]) [6,10,11]. Patients with uncomplicated cystitis usually urine cultures are indicated in patients with complicated UTIs
require dipstick urinalysis (UA) to support the diagnosis of (cUTIs), recurrent UTIs, acute pyelonephritis, or in those at risk
UTIs. Conversely, the combination of dysuria and frequency of infection with antibiotic-resistant organisms [6,10,11]. Blood
or urgency without vaginal discharge or irritation increases the cultures do not contribute to the improved management of
likelihood of acute uncomplicated cystitis to >90% in females. acute pyelonephritis without obtaining an appropriate urine
Therefore, these patients should be treated empirically with- sample [20].
out further testing [12].
Urinary dipstick, used to detect the presence of leukocyte Does the patient have complicated or uncomplicated
esterase and nitrite, exhibits variable test characteristics UTI?
depending on the population studied and clinical presenta-
tion. This test has a high negative predictive value (90–100%) UTIs are usually classified as either complicated or uncompli-
in various patient populations including the elderly. Therefore, cated infections, irrespective of the location and severity of
a negative urinary dipstick for leukocyte esterase and nitrite the infection [18]. An uncomplicated lower UTI is usually
suggests that further evaluation for UTI is unnecessary, parti- defined as acute cystitis occurring in a healthy, premenopau-
cularly if the pretest probability of UTIs is low [13]. However, sal, nonpregnant female with no known structural urological
despite the excellent negative predictive value, a positive abnormalities [6]. cUTIs are associated with an underlying
urinary dipstick or UA result is nonspecific with subsequent condition that increases the risk of recurrent infections or
low positive predictive values [13]. Asymptomatic pyuria and treatment failure due to functional or anatomic abnormalities
asymptomatic bacteriuria are highly prevalent (25–50%) in of the urinary tract (Table 1). cUTIs have a greater risk of
elderly patients, particularly in female residents of long-term morbidity and mortality when compared with uncomplicated
care facilities (LTCFs) and patients with indwelling urinary UTIs [18].
catheters [10,13,14]. Asymptomatic pyuria or bacteriuria in It is imperative to determine the severity of infection since
the setting of nonspecific symptoms (e.g. fatigue, malaise, uncomplicated UTIs usually require fewer diagnostic tests and
worsening urinary frequency or urgency, incontinence, dizzi- can be treated with narrow-spectrum antimicrobial agents for
ness, and confusion) is a trigger for the inappropriate use of shorter durations. On the other hand, all patients with cUTIs
diagnostic tests such as urine culture and inappropriate anti- require urine culture in order to identify the causative patho-
biotic use among elderly, commonly in residents of LTCFs gen due to the wide diversity of potential pathogens and risk
[15,16]. of antimicrobial resistance. Escherichia coli, Klebsiella pneumo-
Vaginitis, sexually transmitted diseases (STDs), and pelvic niae, Enterococcus faecalis, Enterobacter spp., Proteus mirabilis,
inflammatory disease can mimic symptoms of UTIs. In fact, a Serratia marcescens, and P. aeruginosa are common causative
recent study showed that approximately one-third of cases of pathogens in the setting of cUTIs with an increase in relative
STDs was misdiagnosed as UTIs [17]. frequency of non-E. coli pathogens. Recurrent infection, instru-
mentation, and repeat courses of antimicrobial therapy
increase the risk of antibiotic-resistant organisms causing
What type of UTI does the patient have? cUTIs. [6,20].
UTIs can be classified according to the specific site of infection Due to a low yield, routine blood cultures are not recom-
and are often differentiated into upper and lower UTIs. These mended for all patients with cUTIs but in pyelonephritis [20].
include urethritis, cystitis, prostatitis (lower UTIs), and pyelone-
phritis (upper UTIs) [6,18]. Clinical presentation helps differ-
What is the severity of UTI in this patient?
entiate between upper and lower UTIs. Fever in UTIs usually
represents the presence of tissue invasion and inflammation The clinical spectrum of UTIs can range from uncomplicated
such as pyelonephritis or prostatitis although it may occur cystitis to septic shock due to severe pyelonephritis. It is
POSTGRADUATE MEDICINE 3

essential to determine the severity of the infection in order to ESBLs in Gram-negative bacteria have emerged as a major
guide the choice and route of empiric antibiotic therapy and global public health concern, and the rate is rapidly increasing.
determine whether hospital admission is warranted. Severe They are commonly encountered in nosocomial-acquired
infections usually require treatment with broad-spectrum infections or in outbreak settings but are also prevalent in
intravenous empiric antibiotic therapy. Risk factors for severe the community, particularly in LTCFs [36–40]. ESBLs are
UTIs include urinary tract obstruction, hydronephrosis, muco- reported in other Gram-negative species (e. g. Klebsiella oxy-
sal trauma from indwelling catheters or urologic surgery in the toca and Proteus species) in addition to K. pneumonia and E.
presence of bacteriuria, prolonged urologic surgery, infection coli. Genes that encode for ESBLs are usually found on large
with certain antibiotic-resistant organism, hospital-acquired plasmids accompanied by genetic determinants of resistance
infection, and liver cirrhosis [21–24]. against multiple classes of antibiotics, such as aminoglyco-
sides, sulfonamides, and fluoroquinolones [41].
Enterobacteriaceae producing CTX-M-type ESBLs (CTX-M-14
Does the patient have risk factors for UTI with
and CTX-M-15 enzymes) are linked to the dissemination of
antibiotic-resistant organisms?
‘high-risk clone’ sequence type (ST) 131. The H30 and H30Rx
Gram-negative bacteria, specifically Enterobacteriaceae, can subclones account for most ST131 isolates and for the associa-
acquire genes to encode for multiple antibiotic resistance tion of ST131 with fluoroquinolones resistance and ESBLs
mechanisms, such as AmpC β-lactamase, ESBLs, and carbape- production [36,42,43]. The prevalence of ESBLs among patho-
nemases [25]. Increasing resistance in causative Gram-negative gens causing UTIs varies according to region, the patient
bacilli complicates selection of empirical antimicrobial agent. population, and risk factors. ESBL production was detected in
Antimicrobial resistance is associated with inappropriate 8.5% and 8.8% of E. coli and K. pneumoniae in North America
empiric antibiotic therapy that can lead to poor clinical out- and in 17.6% and 38.9% in Europe, respectively [36]. Risk
comes including treatment failure, development of bactere- factors for UTI caused by ESBL-producing organisms include
mia, requirement for intravenous therapy, hospitalization, and hospital-acquired infection, LTCF residence, diabetes, recur-
extended length of hospital stay [26–28]. rent UTIs, and comorbidities [44]. Patients with infection due
Risk factors of UTIs due to antibiotic-resistant Gram-nega- to ESBL-producing organisms are more likely to receive inap-
tive organisms include age older than 60 years, prior UTI propriate antibiotics to experience longer hospital stays and to
history, cUTIs, presence of a urinary catheter, chronic medical present with severe sepsis and septic shock [24,27].
conditions, recent hospitalizations or antibiotic treatment, and Fosfomycin trometamol, nitrofurantoin, and pivmecillinam
recent travel [1,2,7,20]. are active against more than 90% of urinary isolates of ESBL-
E. coli is the most common organism isolated for CAUTI and producing organisms and are considered choices for the oral
among the most common causes of HAUTI [1,20,29,30]. There treatment of lower UTIs (Table 2) [45–49]. Treatment options
was a substantial increase in E. coli resistance to commonly for pyelonephritis caused by ESBL-producing organisms
used antibiotics for the treatment of UTIs such as ciprofloxacin include carbapenems, ceftazidime–avibactam, fosfomycin
(3.6–11.8%) and trimethoprim–sulfamethoxazole (TMP/SMX) (intravenous), cefoxitin, cefepime, temocillin, piperacillin–tazo-
(17.2–22.2%) among outpatient females with UTIs in the per- bactam, ceftolozane–tazobactam, and aminoglycosides
iod of 2003–2012 [29]. Furthermore, the resistance rate of E. [45,50–52].
coli to ampicillin and cefazolin is high, approximately 40% in Carbapenemases, such as K. pneumoniae carbapenemase
this population [29,31]. (KPC), Verona integron-encoded metallo-β-lactamase (VIM),
AmpC β-lactamase is encoded by genes that are commonly New Delhi metallo-β-lactamase (NDM), and oxacillin-hydrolyz-
chromosomally mediated in bacteria (such as Enterobacter ing (OXA)-48 types, are enzymes that neutralize carbapenems
cloacae) but can be mobilized by plasmids to other bacteria. in addition to penicillins and cephalosporins. KPC, most com-
AmpC β-lactamase-producing Enterobacteriaceae typically mon in the USA, and NDM-1 are the most clinically relevant
confer clinically relevant resistance to all penicillins, cephalos- enzymes in this class [53]. These enzymes are encountered in
porins, and cephamycins and are not effectively inhibited by Enterobacteriaceae species, particularly K. pneumonia and E.
clavulanate or tazobactam [32]. The exact prevalence of UTIs coli, and less frequently in other Gram-negative organisms
due to AmpC β-lactamase-producing Enterobacteriaceae is such as P. aeruginosa, Acinetobacter baumannii, and S. marces-
uncertain. Among the 323 non-ESBL-producing cens [54]. CRE are often resistant to all β-lactam agents and
Enterobacteriaceae identified in community-onset UTIs in frequently exhibit various resistance mechanisms such as
Taiwan, 50 isolates were phenotypically positive for AmpC. E. AmpCs and ESBLs including genes conferring resistance to
coli was the most commonly isolated AmpC-producing organ- multiple antimicrobial classes, further limiting treatment
ism (60%), followed by K. pneumoniae (8%), E. cloacae (6%), options [54]. Although CRE infections are relatively infrequent
and P. mirabilis (6%) [33]. Canadian studies demonstrated that worldwide, they are more common in the United States since
1.8–2.5% of urine isolates of E. coli were AmpC producers emerging [53]. In a recent study, the incidence of CRE was 2.93
[34,35], and risk factors include prior use of fluoroquinolones per 100,000 population in the US where most CRE cases were
and cephamycin [33]. isolated from a urinary source [55]. Rate varies from region to
Treatment options for UTIs due to AmpC β-lactamase-pro- region, and it is estimated to be 1.8–2.4% based on the US,
ducing Enterobacteriaceae include nitrofurantoin or fosfomycin European, and Latin American data as reported in the SENTRY
for cystitis and cefepime, piperacillin–tazobactam, fluoroqui- study [56]. Risk factors for infection with CRE include prior
nolones, and carbapenems for pyelonephritis [34,35].
4

Table 2. Treatment options for uncomplicated cystitis and pyelonephritis.


Dosing for
uncomplicated Dosing for uncomplicated
Anti-infective cystitis pyelonephritis Advantages Disadvantages Adverse effects Comments
Nitrofurantoin monohydrate 100 mg twice daily N/A Active against many common urinary No activity against S. marcescens, Nausea Not recommended for empiric
macrocrystals with food for pathogens such as Staphylococcus Morganella spp., P. mirabilis, P. Headache treatment for hospital-acquired
M. S. BADER ET AL.

5 days saprophyticus, Enterococci, E. coli, K. aeruginosa, and CRE Cough UTIs


pneumoniae, and AmpC β-lactamase Not recommended for CrCl of Dyspnea
and ESBL-producing Gram-negative <40 ml/min Rare side effects such as
organisms High failure rate for short-course acute allergic
High urine concentration therapy (3 days) pneumonitis, bone
Low rates of resistance Contraindicated for marrow suppression,
Low risk of Clostridium difficile pyelonephritis and hepatotoxicity
infection Hemolysis in patients with
Pregnancy category B glucose-6-phosphate
dehydrogenase
deficiency and
neuropathy in patients
with chronic kidney
disease
Trimethoprim– 160 mg/800 mg 160 mg/800 mg twice Active against S. saprophyticus, E. coli, No activity against Enterococci, P. Nausea Not recommended for empiric
sulfamethoxazole twice daily for daily for 7–14 days K. pneumoniae, and P. mirabilis aeruginosa, and CRE. Vomiting treatment of pyelonephritis
3 days (females) (definitive treatment) Definitive therapy for susceptible Increasing resistance to common Hypersensitivity reactions Avoid empiric use for cystitis if
OR for 7 days AmpC β-lactamase and ESBL- uropathogens such as E. coli such as fever, rash, resistance rate in the community
(males) producing Gram-negative organisms and AmpC β-lactamase and urticarial, and Stevens– is >20%
Short-course therapy ESBL-producing Gram- Johnson syndrome Avoid coadministration with
Indicated for both negative organisms Bone marrow suppression spironolactone, angiotensin-
uncomplicated cystitis and Not recommended in pregnancy Acute kidney injury converting enzyme inhibitors, or
uncomplicated pyelonephritis (avoid in first trimester and Hyperkalemia and angiotensin-receptor blockers due
High urine concentration last 6 weeks) hyponatremia to increased potential risk of
Low risk of C. difficile infection Potential drug–drug interactions: Hemolysis in patients with hyperkalemia and sudden death
warfarin, phenytoin, and glucose-6-phosphate
sulfonylureas dehydrogenase
Requires dose adjustment in deficiency
renal impairment
Trimethoprim 100 mg twice daily N/A Active against S. saprophyticus, E. coli, No activity against Enterococci, P. Nausea and vomiting Not recommended for empiric
or 200 mg once K. pneumoniae, and P. mirabilis aeruginosa, and CRE. Rash treatment of cystitis if resistance
daily for 3 days High urine concentration Increasing resistance to common Hyperkalemia and rate in the community is >20%
Low risk of C. difficile infection uropathogens such as E. coli, hyponatremia
AmpC β-lactamase, and ESBL- Bone marrow suppression
producing Gram-negative Megaloblastic anemia
organisms Methemoglobinemia
Not indicated for pyelonephritis Acute kidney injury
Drug–drug interaction: dapsone,
methotrexate, and phenytoin
Not recommended in first
trimester of pregnancy
Requires dose adjustment in
renal impairment
(Continued )
Table 2. (Continued).
Dosing for
uncomplicated Dosing for uncomplicated
Anti-infective cystitis pyelonephritis Advantages Disadvantages Adverse effects Comments
Fosfomycin tromethamine 3 g sachet as a N/A Active against many common urinary No activity against Morganella Diarrhea For cUTIs, administer 3 g once daily
single dose pathogens such as S. saprophyticus, spp., Acinetobacter spp., and P. Nausea for 3–5 days or every 48–72 h for
Enterococci, E. coli, K. pneumoniae, Aeruginosa Dyspepsia 3–5 doses (off-label use)
AmpC β-lactamase, and ESBL- Increasing resistance of ESBLs-K. Headache Not recommended for empiric
producing Gram-negative organisms pneumoniae and ESBLs 025b/ Vaginitis treatment of hospital-acquired
and CRE B2 E. coli strains Esophageal discomfort UTIs
Low rates of resistance Oral formulation contraindicated Transient elevation of liver
No cross-resistance with other for pyelonephritis due to low enzymes
antimicrobial classes plasma concentrations
High urine concentration Limited worldwide availability of
Good safety profile IV formulation
Improved compliance (single-dose Drug–drug interaction: antacids
therapy)
Oral formulation
Pregnancy category B
Low risk of C. difficile infection
Fluoroquinolones Ciprofloxacin: Ciprofloxacin 500 mg Active against S. saprophyticus, No activity against Enterococci Nausea For patients who cannot tolerate
250 mg twice twice daily for 7 days Enterobacteriaceae, and P. and CRE Vomiting oral therapy, give first dose IV
daily for 3 days (definitive treatment) aeruginosa Increasing resistance to common Diarrhea If local resistance for
(females) OR for OR 1 g extended- Definitive therapy for susceptible uropathogens such as E. coli, Headache fluoroquinolones exceeds 10%,
7 days (males) release daily for 7 days AmpC β-lactamase and ESBL- AmpC β-lactamase, ESBL- Drowsiness use other options for treatment of
OR (definitive treatment) producing Gram-negative organisms producing Gram-negative Insomnia pyelonephritis such as ceftriaxone
levofloxacin: OR Short-course therapy – lower cost, organisms, P. aeruginosa, and Dizziness or ampicillin–gentamycin
250 mg daily for levofloxacin 500 mg daily improved compliance, and less Acinetobacter spp. Photosensitivity/ combination
3 days (female) for 7 days (definitive adverse effects High risk of C. difficile infection phototoxicity Not recommended for empiric
OR for 7 days treatment) Short course for uncomplicated Pregnancy category C Vaginitis treatment of hospital-acquired
(male) OR pyelonephritis for definitive (contraindicated in pregnancy QTc prolongation UTIs
OR levofloxacin 750 mg daily treatment and breast-feeding) Seizures
norfloxacin for 5 days (definitive Indicated for both uncomplicated Drug–drug interactions: cations Tendinitis and tendon
400 mg twice treatment) cystitis and uncomplicated (antacids and vitamins), rupture
daily for 3 days pyelonephritis warfarin, and theophylline
(female) OR for High urine concentration (monitor)
7 days (male) Require dose adjustment in renal
impairment
Amoxicillin–clavulanate CrCl >30 ml/min CrCl >30 ml/min 875/ Active against S. saprophyticus, No activity against P. aeruginosa, Diarrhea Variation in the clavulanate
875/125 mg 125 mg twice daily Enterococci, E. coli, K. pneumoniae, Morganella morganii, Nausea concentration used by EUCAST
twice daily for 10–14 days and P. mirabilis Providencia stuartii, AmpC β- Vomiting and CLSI results in variable
5–7 days CrCl <30 ml/min 500 mg Definitive treatment for susceptible lactamase, and ESBL- Allergic reactions: rash, susceptibility testing results,
CrCl <30 ml/min twice daily for ESBL-producing Gram-negative producing Gram-negative urticarial, or particularly among ESBL-
500 mg twice 10–14 days organisms organisms and CRE anaphylactic reaction producing E. coli isolates
daily for Pregnancy category B Longer treatment courses Vaginitis Not recommended for empiric
5–7 days required Headaches treatment of acute pyelonephritis
High rates of relapse and failure and hospital-acquired UTIs
even when organism
susceptible
Low urine concentration
High risk of C. difficile infection
POSTGRADUATE MEDICINE

Requires dose adjustment in


renal impairment
(Continued )
5
6
M. S. BADER ET AL.

Table 2. (Continued).
Dosing for
uncomplicated Dosing for uncomplicated
Anti-infective cystitis pyelonephritis Advantages Disadvantages Adverse effects Comments
Ampicillin (IV) Only if unable to Ampicillin IV 1 to 2 g Active against S. saprophyticus, E. No activity against K. Diarrhea Not recommended for empiric
Amoxicillin tolerate oral every 6 h with faecalis, E. coli, and P. mirabilis pneumoniae, P. aeruginosa, M. Nausea treatment of either cystitis or
formulation gentamicin 5 mg/kg Pregnancy category B morganii (%), P. stuartii, AmpC Vomiting pyelonephritis
500 mg every 8 h every 24 h for β-lactamase, and ESBL- Allergic reactions: rash,
for 5–7 days 10–14 days producing Gram-negative urticarial, or
organisms and CRE anaphylactic reaction
Generally inferior to Vaginitis
fluoroquinolones Headaches
High rate of resistance among S.
saprophyticus
Low urine concentration
High risk of C. difficile infection
Requires dose adjustment in
renal impairment
Ceftriaxone N/A 1 to 2 g IV every 24 h for Active against E. coli, K. pneumoniae, P. No activity against Enterococci, P. Diarrhea
7–14 days mirabilis, Enterobacter, Serratia, aeruginosa, AmpC β- Thrombocytopenia,
Citrobacter, Morganella spp., and P. lactamase, and ESBL- thrombocytosis, and
stuartii producing Gram-negative leukopenia
For empiric use of pyelonephritis organisms and CRE Biliary pseudolithiasis
No dose adjustment required in renal Low urine concentration Risk of cross-reactivity in
dysfunction High risk of C. difficile infection patients with beta-
Administered once daily lactam allergy
Pregnancy category B
N/A: not applicable; ESBLs: extended-spectrum β-lactamases; CRE: carbapenem-resistant Enterobacteriaceae; CrCl: creatinine clearance; UTIs: urinary tract infections; cUTIs: complicated UTIs; IV: intravenous; EUCAST: the
European Committee on Antimicrobial Susceptibility Testing; CLSI: clinical and laboratory standards institute.
POSTGRADUATE MEDICINE 7

hospitalizations, indwelling devices, prolonged intensive care to meropenem-, piperacillin–tazobactam-, and ceftazidime-
unit (ICU) stay, surgery or invasive procedures, and prior car- resistant Pseudomonas spp. [63,69].
bapenems exposure [55,57–59]. Infections with CRE are asso-
ciated with higher mortality rates than those for infections
caused by carbapenem-susceptible organisms but not specifi- Where should the patient be treated?
cally in patients with UTIs [60,61]. The majority of uncomplicated UTIs including pyelonephritis
Limited treatment options for UTIs due to CRE include can be treated in the outpatient setting. However, patients
ceftazidime–avibactam, carbapenems, colistin, polymyxin B, with pyelonephritis or cUTIs who are moderately–severely ill
aztreonam, aminoglycosides, fosfomycin, and tigecycline are not responding to outpatient therapy and have concomi-
[62,63]. Despite in vitro resistance, high-dose meropenem tant unstable comorbid conditions and should be managed in
may be an option against infections caused by CRE, particu- the hospital setting [6,10].
larly OXA-48 carbapenemases and in settings where the mini-
mum inhibitory concentration (MIC) for the CRE pathogen is
≤8 mg/l [64]. Synergy with combination treatment for CRE has What are the indications for imaging?
been an area with emerging data although clear guidance is
limited. Imaging is useful in diagnosing the underlying structural
Combination therapy may be considered for severely ill causes of cUTIs including calculi, tumor, abscesses, and
patients [65]. Combination of ertapenem-containing double- infected cysts. Imaging at initial evaluation of UTIs is typically
carbapenem therapy was associated with 67% success in not necessary unless one suspects a concomitant emergent
treating UTIs due to carbapenem-resistant K. pneumo- condition requiring prompt intervention, such as an obstruc-
niae [66]. tive uropathy by a renal stone or tumor. Other indications for
P. aeruginosa also represents a major cause of UTIs and imaging include persistent fever for 72 h after appropriate
often exhibit decreased susceptibility to various antimicrobial antibiotic therapy, rapid recurrence after antibiotic treatment,
agents. MDR Pseudomonas spp.is defined as an isolate that is and in diabetics who have a higher incidence of complications
nonsusceptible to at least one agent in three antimicrobial (e.g. emphysematous pyelonephritis which may require
classes that have activity against Pseudomonas spp. [25]. It is immediate nephrectomy and obstruction from necrotic renal
usually associated with health-care infections where it is the papillae that are sloughed into the collecting system and
second most common isolated pathogens in catheter-asso- obstruct the ureter). Imaging with kidney and bladder ultra-
ciated UTIs (CAUTIs). MDR Pseudomonas spp. account for 14% sound or a non-contrast computed tomography scan of the
of cUTIs, and the resistance rates for fluoroquinolones, carba- abdomen and pelvis may be useful for these indications [70].
penem, piperacillin–tazobactam, and aminoglycoside were
34%, 21%, 17%, and 11%, respectively [67]. Among a total of
Treatment
7272 unique patient clinical urinary isolates collected from
patients with UTIs in 71 US medical centers in a period of The management of uncomplicated cystitis includes confirm-
2012–2014 as part of the International Network for Optimal ing the diagnosis with urinary dipstick or UA if required,
Resistance Monitoring program, 6% were due to Pseudomonas initiating narrow-spectrum antibiotics, and then assessing
spp. The resistance rate of Pseudomonas spp. to ceftazidime, response to empiric therapy (Table 2). The management of
levofloxacin, piperacillin–tazobactam, meropenem, and cefta- cUTIs and acute pyelonephritis in general includes obtaining
zidime–avibactam was 8.1%, 26.9%, 6.3%, 13.6%, and 2.3%, urine culture, initiating broad-spectrum antibiotic, and de-
respectively. The rates increase significantly once the isolate escalating the empiric antibiotic selection once susceptibility-
develops resistance to one of these antibiotics except for testing results are available (Table 2) [6,10].
ceftazidime–avibactam [63]. The Study for Monitoring Key considerations in the selection of empiric antibiotic
Antimicrobial Resistance Trends study which has monitored therapy for UTIs include the bacterial spectrum, local resis-
the susceptibilities of Gram-negative bacilli from inpatient UTIs tance patterns, comorbid conditions (e.g. renal impairment
since late 2009 in 24 sites in the USA showed that P. aerugi- and liver impairment), history of drug allergy, prior antibiotic
nosa accounted for 9.6% of the isolates. The susceptibility rate exposure, antibiotic safety profile, and urinary concentrations
of P. aeruginosa to cefepime, imipenem, ciprofloxacin, and of antibiotics [6,10]. The most frequently isolated pathogens
piperacillin–tazobactam was 67.6%, 68%, 52.5%, and 74%, from outpatients with UTIs include E. coli (64.9%), K. pneumo-
respectively [68]. niae (10.1%), P. mirabilis (5.0%), E. faecalis (4.1%), and P. aeru-
Treatment options of UTIs due to MDR-Pseudomonas spp. ginosa (2.7%) [29,71]. The most frequently isolated pathogens
include aminoglycosides (amikacin, gentamycin, and tobra- from inpatients with UTIs or cUTIs are E. coli (48.9%),
mycin), fluoroquinolones (ciprofloxacin and levofloxacin), Enterococcus spp. (18%), K. pneumoniae (14.5%), P. aeruginosa
antipseudomonal cephalosporins (ceftazidime and cefe- (9.6%), P. mirabilis (6.4%), E. cloacae (4.6%), K. oxytoca (2.5%),
pime), antipseudomonal penicillins (piperacillin–tazobactam), and others such as Citrobacter freundii, Citrobacter koseri,
carbapenems (meropenem, imipenem, and doripenem), Enterobacter aerogenes, M. morganii, and S. marcescens
colistin, aztreonam, ceftazidime–avibactam, and ceftolo- [20,68,71].
zane–tazobactam [63,67,68]. Ceftolozane–tazobactam and Assessment of a patient’s previous urinary pathogen and
ceftazidime–avibactam are therapeutic options for UTIs due susceptibility profile is essential in selecting appropriate
empiric antibiotic therapy. Prior microbiologic data improved
8 M. S. BADER ET AL.

the rate of accuracy of the empiric treatment against the for the treatment of cUTIs caused by ESBL-producing
pathogen for UTIs from 32% to 76% [72]. Enterobacteriaceae, particularly ESBLs-E. coli [86,87].
Understanding the local susceptibility patterns for common Although there is still a low rate of resistance of uropatho-
Gram-negative pathogens is important to guide empiric anti- gens to fosfomycin worldwide, there is a growing concern
biotic therapy for UTIs [6,10,73]. with resistance, particularly to ESBLs-K. pneumonia, hospital-
Clinicians should strive to streamline empiric antibiotic acquired strains, and certain strains of ESBLs-E. coli. The resis-
therapy once culture and susceptibility profiles are available. tance rate to fosfomycin was 19.9% among 204 MDR Gram-
Selective pressure exerted by broad-spectrum antibiotics plays negative uropathogens collected from January 2010 to June
a crucial role in the emergence of antibiotic-resistant bac- 2013 in the Boston area where 32% of ESBL-Klebsiella spp.
teria [74]. uropathogens were resistant to fosfomycin [88]. Fosfomycin
Without treatment, 25–42% of uncomplicated acute cystitis resistance varies according to ESBL type where CTX-M-15 and
cases in females will resolve spontaneously. In fact, in the O25b/phylogroup B2 isolates are associated with fosfomycin-
absence of effective treatment, only approximately 2% of resistant ESBL-E. coli [89].
uncomplicated acute cystitis will progress to pyelonephritis.
Thus, delaying antibiotic treatment while evaluating sympto-
matic lower UTIs generally does not lead to adverse outcomes Trimethoprim–sulfamethoxazole
[75–77].
Unfortunately, TMP/SMX is no longer recommended as first-
line empiric therapy for outpatients with uncomplicated cysti-
Nitrofurantoin tis, cUTIs, and acute pyelonephritis due to the high rate of
resistance in several communities [6,20]. E. coli resistance to
Nitrofurantoin remains a reliable first-line agent for the empiri-
TMP/SMX increased significantly from 17.2% to 22.2% among
cal treatment of acute uncomplicated cystitis [6,31,78]. E. coli
outpatient adult females in the period of 2003–2012 in the US
resistance to nitrofurantoin among outpatient adult females
[29]. In regions where TMP–SMX resistance is lower than 20%,
with UTIs is low (0.9%) and have remained unchanged in the
TMP–SMX is not a reliable empiric treatment option for UTIs in
period of 2003–2012 in the US [29]. There is a growing con-
patients with recent exposure to either TMP–SMX or ciproflox-
cern of increasing resistance of ESBL-producing organisms
acin, in patients with prior UTIs with ESBL-producing E. coli or
against nitrofurantoin, particularly in hospital-acquired infec-
in those who have traveled recently to an area with high rates
tions. In Canada, the rate of susceptibility of urinary isolates of
of TMP–SMX resistance [20,31,35]. TMP-SMX is not recom-
ESBLs-E. coli to nitrofurantoin has decreased from 91% to 83%
mended as empiric treatment option of UTIs caused by
in the period of 2007–2013 [35]. There is a high resistance rate
ESBL-E. coli or K. pneumonia and AmpC β-lactamase-producing
to nitrofurantoin (20–30%) among E. cloacae, E. aerogenes, K.
E. coli due to high levels of resistance, >66% and 35–40%,
pneumoniae, and Serratia spp. [29,71]. Risk factors of resistance
respectively [20,35,89].
to nitrofurantoin include male gender, hospital-acquired infec-
However, TMP–SMX can be used as definitive therapy if the
tion, resistant to other oral antibiotics including ciprofloxacin,
isolated organism is susceptible [6]. Other information is sum-
and ESBL-producing organisms [29,35,79]. Other relevant
marized in Table 2 [90,91].
information is summarized in Table 2 [6,80–82].

Fosfomycin Fluoroquinolones
It is available in oral (fosfomycin tromethamine) and intrave- Fluoroquinolones (norfloxacin, ciprofloxacin, and levofloxacin)
nous (disodium fosfomycin) formulations, which are not are second-line options for uncomplicated cystitis and one of
widely available except in a few countries [83]. Fosfomycin is the treatment options for cUTIs and pyelonephritis when the
recommended as first-line definitive treatment for acute overall resistance rate is less than 10% (Table 2) [6,10].
uncomplicated cystitis in females as a single oral dose of Fluoroquinolone resistance among urinary pathogens is on
fosfomycin trometamol 3 g (Table 2) [6]. The susceptibility of the rise in both community and hospital settings. Resistance
uropathogens to fosfomycin has remained relatively stable of E. coli to ciprofloxacin among outpatient adult females from
over time [84]. About 99.4% of 868 urinary isolates of E. coli the US increased from 3.6% to 11.8% in the period of
obtained by clinical laboratories across Canada from 2010 to 2003–2012 [29]. There is a high resistance rate of E. coli
2013 were fosfomycin susceptible [35]. It is a valuable oral (34.5%) and P. aeruginosa (34%) to fluoroquinolones among
option for treatment of UTIs due to MDR pathogens such as hospitalized patients with UTIs [92,93]. Furthermore, these
ESBL-producing Enterobacteriaceae in both community and agents are not recommended for empiric treatment of UTIs
health-care settings [85]. It is the most active agent in hospi- due to ESBL-producing Enterobacteriaceae. Only 7% and 18%
talized patients or in patients presenting to the emergency of ESBL-E. coli and ESBL-K. pneumonia, respectively, are sus-
department with UTIs due to MDR pathogens where 94.5% of ceptible to ciprofloxacin among hospitalized patients with
organisms including ESBLs are susceptible [8]. In addition, 95% UTIs in North America [36,68]. Infection with ESBL-producing
and 97% of ESBLs- and AmpC-producing E. coli isolates, organism particularly CTX-M-15 strain, urinary catheterization,
respectively, obtained by clinical laboratories across Canada recent hospitalization, hospital-acquired infection, residents of
from 2007 to 2013 were susceptible [35]. Fosfomycin tro- LTCFs, elderly patients, male gender, previous UTI, previous
methamine is as effective as and less costly than carbapenems exposure to TMP–SMX, metronidazole, cephalosporins, and
POSTGRADUATE MEDICINE 9

fluoroquinolones are risk factors associated with increased ST131-like E. coli isolate (75%, 66.7%) than non-ST131-like
fluoroquinolones resistance [8,31,71,94–97]. isolate (93.8% versus 86.7%, respectively) [107].
REPRISE (Ceftazidime-Avibactam for the Treatment of
Infections Due to Ceftazidime Resistant Pathogens) is an inter-
national, randomized, open-label, Phase III trial that recruited
Cephalosporins
patients with cUTIs and complicated intra-abdominal infection
Ceftriaxone is a reasonable empiric treatment option for cUTIs due to ceftazidime-resistant Enterobacteriaceae and P. aerugi-
and acute pyelonephritis since the majority of urinary patho- nosa from hospitals across 16 countries worldwide. It showed
gens are susceptible (Table 2) [6,10]. For example, 89%, 92%, that the clinical cure at the test-of-cure visit, 7–10 days after
and 96% of E. coli, K. pneumoniae, and P. mirabilis isolated last infusion of study therapy, was similar between ceftazi-
from hospitalized patients with UTIs in North America were dime–avibactam and the best available therapy, carbapenem,
susceptible to ceftriaxone [36]. However, a high rate of resis- in patients with cUTIs [108].
tance to ceftriaxone has been reported among AmpC β-lacta- Ceftolozane–tazobactam is a novel cephalosporin com-
mase- and ESBL-producing organisms and in patients from bined with a β-lactamase inhibitor, and it has potent in vitro
LTCFs [68,92]. Therefore, ceftriaxone should be used with cau- activity against MDR-P. aeruginosa and other common Gram-
tion as empiric treatment of HAUTIs and should be avoided in negative pathogens, including most ESBL-producing
patients with pyelonephritis who are critically ill or at risk of Enterobacteriaceae spp. (Table 3) [69,109]. In Phase III, rando-
ESBLs, Pseudomonas spp., or other resistant organisms. mized, multicenter, double-blind studies, 1083 adult hospita-
Ceftazidime has similar antibacterial activity to ceftriaxone lized patients with either pyelonephritis, 82% of study
in addition to activity against Pseudomonas spp. (Table 3) and patients, or cUTIs were randomly to receive either ceftolo-
is considered a reasonable empiric treatment option for cUTIs zane–tazobactam 1.5 g every 8 h (n = 543) or levofloxacin
and acute pyelonephritis [36,63,68,71]. 750 mg daily (n = 540) for 7 days. Ceftolozane–tazobactam
Cefepime has similar antibacterial activity to ceftazidime demonstrated improved overall cure rate compared to high-
(Table 3). Cefepime is relatively stable to AmpC hydrolysis, dose levofloxacin (76.9% of 398 vs. 68.4% of 402, respectively;
and 96.6% AmpC β-lactamase-producing E. coli are susceptible 95% CI: 2.3–14.6) [110]. In a post hoc analysis, ceftolozane/
to cefepime; therefore, it is an option for treating cUTIs and tazobactam demonstrated significantly higher composite cure
pyelonephritis caused by these organisms [33,34,98,99]. A high rates than levofloxacin in UTIs due to levofloxacin-resistant
dose of cefepime (2 g every 12 h or 1 to 2 g every 8 h) is organisms (60.0% vs. 39.3%) and ESBL-producing uropatho-
recommended for organisms with MIC 4–8 μg/ml in order to gens (58.3% of patients vs. 34.9%). In patients with levoflox-
maximize the pharmacodynamics profile [100]. Up to 53% of acin-resistant P. aeruginosa, eradication was achieved in 100%
ESBL-E. coli and 58% ESBLs-K. pneumonia are susceptible to who received ceftolozane/tazobactam and 37.5% who
cefepime; however, this agent should not be used for the received levofloxacin (95% CI for the treatment difference,
treatment of ESBL infections, particularly in critically ill patients 22.1–86.3%) [111].
or when MICs >2 μg/ml [34]. Cefepime is rapidly hydrolyzed
by ESBLs such as CTX-M-10 and TEM-10 [101,102].
β-Lactam and β-lactamase inhibitor
The Food and Drug Administration (FDA) has approved the
ceftazidime–avibactam combination for treatment of cUTIs, Amoxicillin and ampicillin should be avoided as empiric treat-
including pyelonephritis, in patients with limited or no alter- ment for UTIs; however, these agents are effective as definitive
native treatment options (Table 3). Avibactam is a novel therapy if the pathogen proves susceptible (Table 2) [6]. The
bridged diazabicyclooctane, a semisynthetic, non-β-lactam, β- resistance rate of E. coli against ampicillin was 41% in out-
lactamase inhibitor that is active against Ambler class A (e.g. patient adult females with uncomplicated cystitis [29]. The
SHV, TEM, CTX-M, and KPC), class C (e.g. AmpC, FOX, CMY-2, percentage of resistance of E. coli, K. pneumonia, and P. mir-
and AAC-1), and some class D (e.g. OXA-48) serine β-lacta- abilis to amoxicillin–clavulanate was 3.9%, 3.1%, and 0.8%,
mases. Therefore, avibactam improves the antimicrobial activ- respectively [29]. Although amoxicillin–clavulanate is consid-
ity of ceftazidime against AmpC β-lactamases, ESBL-producing ered a second-line agent for UTIs, the increasing prevalence of
Enterobacteriaceae, ceftazidime-resistant E. cloacae, KPC-pro- E. coli resistance to TMP–SMX and fluoroquinolones may be
ducing K. pneumonia, and MDR-P. aeruginosa [39,63,103–105]. driving increased reliance on this agent for the management
A Phase II, prospective, randomized, double-blind trial that of cystitis (Table 2). In 2014, the European Committee on
included 135 patients with cUTIs (63% had pyelonephritis) Antimicrobial Susceptibility Testing introduced a urinary
caused by a Gram-negative pathogen (mainly E. coli, 93%) amoxicillin–clavulanate susceptible breakpoint of ≤32 µg/ml
demonstrated a favorable microbiological response in 70.4% for uncomplicated UTI, which may explain high failure rates of
(19/27) of patients treated with ceftazidime–avibactam in amoxicillin–clavulanate compared to ciprofloxacin for the
comparison with 71.4% (25/35) treated with imipenem (differ- treatment of acute uncomplicated cystitis [112,113].
ence of −1.1%, 95% confidence interval [CI]: −27.2% to 25%) Piperacillin–tazobactam is a broad-spectrum antibiotic that
[106]. In a subgroup analysis, the efficacy of ceftazidime–avi- has activity against Enterobacteriaceae and Pseudomonas spp.
bactam (72.7%, 93.8%) and imipenem (61.5%, 90.9%) was less [34,36,63,68,69,71] and is one of the empiric treatment options
for ESBL-producing Enterobacteriaceae than non-ESBL-produ- for moderate–severe cUTIs and HAUTIs (Table 3) [6,10].
cing Enterobacteriaceae. The clinical response for both ceftazi- The role of β-lactam/β-lactamase inhibitors (BLBLIs) (amox-
dime–avibactam and imipenem was lower in cUTIs due to icillin–clavulanic acid, piperacillin–tazobactam, ampicillin–
10

Table 3. Drugs for treatment of urinary tract infections (UTIs) caused by antibiotic-resistant Gram-negative organisms.
Anti-infective Dosing Advantages Disadvantages Adverse effects Comments
Ceftazidime 1 to 2 g IV every 8 h Active against E. coli, K. pneumoniae, P. mirabilis, Proteus Lacks activity against Enterococci and CRE Seizures Avoid in patients with history of
vulgaris, Enterobacter, Serratia, Citrobacter, Morganella spp., High resistance rate of AmpC β- Encephalopathy cephalosporin-associated
P. stuartii, and P. aeruginosa including some MDR lactamase and ESBL-producing Gram- Prolonged prothrombin hemolytic anemia
Pseudomonas strains negative organisms and A. baumannii time
High urine concentration Increasing resistance of P. aeruginosa Local infusion-related
Pregnancy category B Not indicated UTI due to S. saprophyticus effects
Risk of cross-reactivity with penicillins Nausea
M. S. BADER ET AL.

High risk of C. difficile infection Vomiting


Requires dose adjustment in renal Diarrhea
impairment
Sodium content: 54 mg/g
Cefepime 1 to 2 g IV every 8 h or 2 g IV Active against many common urinary pathogens including S. It has no activity against Enterococci and Hypersensitivity For UTIs due to organisms with an
every 12 h saprophyticus, E. coli, K. pneumoniae, AmpC β-lactamase- CRE (except OXA-lactamases such as Encephalopathy MIC range of 4–8 μg/ml, a higher
producing Gram-negative organisms, and P. aeruginosa OXA-48, OXA-181) associated with doses may be required (2 g every
High urine concentration High resistance rate of ESBL-producing higher doses in 12 h or 1 to 2 g every 8 h)
Pregnancy category B Gram-negative organisms and A. setting of renal Avoid in patients with history of
baumannii dysfunction cephalosporin-associated
Increasing resistance of P. aeruginosa Rash hemolytic anemia
Risk of cross-reactivity with penicillins Diarrhea
High risk of C. difficile infection Nausea
Requires dose adjustment in renal Vomiting
impairment
Ceftazidime– 2.5 g IV every 8 h Activity similar to ceftazidime in addition to activity against Lacks activity against Enterococci, class B
Seizures Avoid in patients with history of
avibactam AmpC β-lactamase and ESBL-producing Gram-negative metallo-β-lactamases (e.g. NDM, VIM,
Myoclonia cephalosporin-associated
organisms, CRE, and MDR-P. aeruginosa IMP, VEB, and PER) and A. baumanniiNausea hemolytic anemia
Approved for cUTIs and pyelonephritis Risk of cross-reactivity with penicillins
Constipation
High urine concentration Requires dose adjustment in renal Vomiting
Pregnancy category B impairment Elevated liver enzymes
Risk of C. difficile infection Dizziness
Limited data for the use in the elderly
Headache
population Hypertension
Sodium content: 146 mg per 2.5 g Insomnia
Cough
Ceftolozane– 1.5 g IV every 8 h Active against common urinary pathogens such as E. coli, K. Lacks activity against Enterococci and CRE Nausea Avoid in patients with history of
tazobactam pneumoniae, AmpC β-lactamase and ESBL-producing Gram- Reduced efficacy in patients with Diarrhea cephalosporin-associated
negative organisms, and MDR-P. aeruginosa moderate renal impairment Headaches hemolytic anemia
Approved for treatment of cUTIs and pyelonephritis Risk of cross-reactivity with penicillins Pyrexia
High urine concentration Requires dose adjustment in renal
Pregnancy category B impairment
Risk of C. difficile infection
Piperacillin– 3.375 g IV every 6 h or 4.5 g Active against common urinary pathogens such as S. Lacks activity against most ESBL- Rash Not indicated for empiric treatment
tazobactam every 8 h saprophyticus, Enterococci, E. coli, K. pneumoniae, AmpC β- producing Gram-negative organisms Nausea of UTIs in patients at risk of ESBL-
lactamase producing Gram-negative organisms, ESBLs-E. and CRE Diarrhea producing Gram-negative
coli, and MDR-P. aeruginosa Increasing resistance rates for P. Neutropenia organisms
Indicated for both cUTIs and pyelonephritis aeruginosa Hypokalemia Continuous infusion may increase
High urine concentration Requires dose adjustment in renal Prolonged prothrombin activity through optimizing
Pregnancy category B impairment time (specifically in pharmacodynamics
Moderate risk of C. difficile infection renal failure)
Drug–drug interaction: reduces
elimination of methotrexate
Sodium 64 mg/g of piperacillin
(Continued )
Table 3. (Continued).
Anti-infective Dosing Advantages Disadvantages Adverse effects Comments
Carbapenems Ertapenem 1 g IV daily Active against common urinary pathogens such as S. Lacks activity against CRE and E. faecium Seizures Carbapenems are considered first-
Meropenem 500 mg every saprophyticus, Enterococci, E. coli, K. pneumoniae, AmpC β- Ertapenem has no activity against Diarrhea line options for serious systemic
6 h up to 1 g IV every 8 h lactamase, ESBL-producing Gram-negative organisms, P. Enterococci, P. aeruginosa, and A. Nausea infections caused by ESBL-
Doripenem 500 mg IV every aeruginosa, and A. baumannii baumannii Rash producing Gram-negative
8h Indicated for cUTIs and pyelonephritis Increasing resistance rates of ESBL- Headache organisms
Imipenem/cilastatin 500 mg High urinary concentration producing Gram-negative organisms Hypokalemia Combination therapy (dual
IV every 6 h Once-daily dosing for ertapenem (particularly K. pneumonia and P. Elevated liver enzymes carbapenem regimen) may exert
Low risk of C. difficile infection mirabilis), P. aeruginosa, effect against CRE
Pregnancy category B (meropenem, ertapenem, and and A. baumannii Continuous infusion may increase
doripenem) Require dose adjustment in renal activity through optimizing
impairment pharmacodynamics
Risk of cross-reactivity with penicillins
Pregnancy category C for imipenem
Aminoglycosides Gentamicin 5 mg/kg IV once Active against common urinary pathogens such as E. coli, K. Lacks activity against S. saprophyticus, Nephrotoxicity Dosing based on ideal body weight
daily pneumoniae, AmpC β-lactamase, ESBL-producing Gram- Enterococci Vestibulocochlear Monitor trough levels
Tobramycin 5 mg/kg IV once negative organisms, MDR-P. aeruginosa, CRE, and A. Increasing resistance rates of ESBL- toxicity Amikacin is preferred over
daily baumannii producing Gram-negative organisms, Neurotoxicity gentamicin when used for empiric
Amikacin 15 mg/kg IV once Indicated for cUTIs and pyelonephritis P. aeruginosa, Edema treatment due to antibiotic-
daily High urine concentration and A. baumannii resistant Gram-negative
Once-daily dosing Require dose adjustment in renal organisms such ESBL-producing
Low risk of C. difficile infection impairment Enterobacteriaceae or MDR-P.
Increased risk of nephrotoxicity with aeruginosa
concomitant nephrotoxins Combination therapy is
Pregnancy category D recommended (e.g. with
ampicillin) when used as empiric
treatment
Monotherapy is acceptable if used as
definitive therapy
Polymixins Polymixin B 15,000–25,000 Active against common urinary pathogens such as E. coli, K. Lacks activity against S. saprophyticus, Nephrotoxicity Dosing is based on ideal body
Units/kg/day IV every pneumoniae, AmpC β-lactamase, ESBL-producing Gram- Enterococci, Proteus spp., Providencia Neurotoxicity weight
q12 h (maximum daily negative organisms, MDR-P. aeruginosa, CRE, and A. spp., S. marcescens, and M. morganii Neuromuscular Colistin (polymixin E) is preferred
dose 2 million units) baumannii Colistin resistance is emerging in A. blockade over polymixin B because of its
Polymixin E (colistin) 2.5– Bladder irrigation may be an option for those who cannot baumannii and K. pneumonia, MDR-P. Respiratory arrest higher urine concentration
5 mg CBA/kg/day in two to tolerate intravenous aeruginosa, and E. coli
four divided doses Low resistance rates Increased failure rate with monotherapy,
High urine concentration for polymixin E particularly in patients with
Low risk of C. difficile infection bacteremia
Low urine concentration for polymixin B
These agents exhibit a narrow
therapeutic window.
Require dose adjustment in renal
impairment
Safety in pregnancy has not been
established for polymixin B
Polymixin E is pregnancy category C
MDR: multidrug resistant; ESBLs: extended-spectrum β-lactamases; CRE: carbapenem-resistant Enterobacteriaceae; IV: intravenous; CBA: colistin base activity; cUTIs: complicated UTIs; NDM: New Delhi metallo-β-lactamase; q 12
POSTGRADUATE MEDICINE

hours: every 12 hours; MIC: minimal inhibitory concentration.


11
12 M. S. BADER ET AL.

sulbactam, cefoperazone–sulbactam, and ticarcillin–clavulanic most active of the carbapenem agents, with 11.4% of isolates
acid) in the treatment of infections caused by ESBL producers being resistant to doripenem, in comparison with 21.9% and
is controversial [114–117]. In vitro activity of amoxicillin–clavu- 15.4% resistance for imipenem and meropenem, respectively.
lanate against ESBL-E. coli isolates obtained by clinical labora- The resistance rate of Pseudomonas isolates to carbapenems
tories across Canada from 2007 to 2013 was 33% susceptible was even higher in ICU patients (doripenem 14.8%, merope-
and 55% intermediate [35]. In North America, >80% of ESBL-E. nem 20.1%, and imipenem 29.1%) [121]. A study that collected
coli, almost 100% of ESBL-P. mirabilis, and 20–30% of ESBL- bacterial isolates from patients hospitalized with UTIs at 73
Klebsiella spp. were susceptible to piperacillin–tazobactam medical centers across all nine US census bureau regions
[36,37,63,68,71]. during 2012 showed that only 65% of ESBLs-K. pneumonia
The systematic review by Vardakas et al. and the post hoc were susceptible to meropenem [122]. Prior use of carbape-
analysis by Rodriguez-Bano showed that BLBLIs were non-inferior nems is associated with colonization and infection with carba-
to carbapenems as empiric or definitive treatment of bacteremia penem-resistant bacteria [57,58]. Therefore, carbapenems
due to ESBL-producing Enterobacteriaceae, particularly E. coli should be restricted to the aforementioned indication, and
[114,115]. A recent study showed the superiority of carbapenems when possible, carbapenems must be discontinued or de-
over piperacillin–tazobactam for the treatment of bacteremia due escalated [45,48,52]. Non-carbapenem treatment options can
to ESBL-producing Enterobacteriaceae (only 20% of the bacteremia be used in mild–moderate UTIs and as step-down therapy
were of urinary source) [116]. It should be noted that these organ- from carbapenems [123,124].
isms could confer resistance to piperacillin–tazobactam and amox-
icillin–clavulanic acid by AmpC β-lactamases, efflux pumps, loss of
Aminoglycosides
porin channels, and alterations in penicillin-binding proteins [116].
In summary [117], amoxicillin–clavulanate may be effective Aminoglycosides are almost exclusively renally excreted, achieve
as definitive treatment of lower UTIs due to susceptible ESBL-E. high urinary levels, and are approved by the FDA for the treat-
coli. Piperacillin–tazobactam may be used in the definitive treat- ment of UTIs (Table 3) [125]. The risk of nephrotoxicity and
ment of mild–moderate cUTIs and acute pyelonephritis due to vestibulocochlear toxicity and increasing antimicrobial resistance
ESBL-producing E. coli when the MIC is ≤16 mg/L, but prefer- limits the utility of aminoglycosides. Amikacin can be used for
ably ≤8 mg/L. A higher dose or prolonged or continuous infu- the empiric treatment of cUTIs and acute pyelonephritis due to
sion of piperacillin–tazobactam can be used to maximize antibiotic-resistant Gram-negative organisms including ESBL-
pharmacodynamic properties of the agent in these infections producing organisms, MDR-Pseudomonas spp., and
and is associated with decreased mortality [118]. Posttreatment Acinetobacter spp. Clinical and microbiologic success with ami-
urine culture may be indicated if these antibiotics are used to kacin was achieved in 97.2% and 94.1% of patients with cUTIs
treat UTIs due to susceptible ESBL-producing uropathogens. due to ESBL-producing bacteria, particularly E. coli [126]. A recent
BLBLIs should be avoided in severe UTIs and in areas where study reported that 98%, 100%, 94%, and 92% of urinary isolates
resistance to BLBLIs is common, especially where ESBL-produ- of P. aeruginosa, ceftazidime-non-susceptible P. aeruginosa, mer-
cing K. pneumoniae is prevalent. They should be avoided in openem-non-susceptible P. aeruginosa, and A. baumannii were
UTIs, particularly moderate–severe infection, caused by E. coli susceptible to amikacin, respectively, in hospitalized patients
ST131 due to the presence of OXA-1/30, which can confer with UTIs [122]. Amikacin may have a role in treatment of UTIs
resistance to both piperacillin and tazobactam, and K. pneumo- due to CRE since ceftazidime–avibactam has activity against
nia with the OXA-1/30 enzyme [119]. some class D (e.g. OXA-48) serine β-lactamases and no activity
versus class B metallo-β-lactamases [127].
To minimize the risk of nephrotoxicity, a single dose of amikacin
monotherapy or in combination therapy with other antibiotics (e.
Carbapenems
g. ampicillin) can be used as an empiric treatment option of cUTI or
Carbapenems (imipenem, meropenem, doripenem, and erta- acute pyelonephritis in patients at risk of infection with antibiotic-
penem) are broad-spectrum antibiotics that retain the greatest resistant organisms such as ESBL-producing organisms or MDR-
level of activity against ESBL-negative Enterobacteriaceae and pseudomonas spp. pending final results of culture and susceptibil-
ESBL-positive E. coli overall in both North America and Europe ity testing [128]. Gentamicin or tobramycin is not a viable empiric
(Table 3) [36,48,68,71,106]. Carbapenems are active against option due to high antibiotic-resistant Gram-negative organisms
most ESBL-producing strains and have been recommended such as ESBL-producing organisms and MDR-Pseudomonas spp.
as first-line empiric therapy. These agents are also options [48,79,122]. Similar to other antimicrobials, resistance to aminogly-
for targeted therapy in critically ill patients with severe infec- cosides is emerging in Gram-negative organisms and is commonly
tions when there is a risk or documentation of ESBL-producing encountered among hospital-acquired ESBL-E. coli, ESBL-K. pneu-
Gram-negative bacteria [106,114,116,120]. However, there is a moniae, and Acinetobacter spp. [36,68].
growing concern of reduced susceptibility of Pseudomonas
spp., K. pneumonia, particularly ESBL-producing strain, and P.
Polymixins
mirabilis to carbapenems [36,63]. The Carbapenem
Antimicrobials Pseudomonas Isolate Testing At regional Polymixins include polymixin B and polymixin E (colistin) given
Locations surveillance program which collected P. aeruginosa in the form of the sodium salt of colistin methanesulfonate,
clinical isolates, 11.4% were urinary, from 100 sites throughout which is also known as colistimethate (Table 3). The polymix-
the USA and Puerto Rico showed that doripenem was the ins are used as a last-resort treatment of MDR bacterial
POSTGRADUATE MEDICINE 13

infections caused by CRE, MDR-P. aeruginosa, and A. bauman- Fluoroquinolones are not recommended for empiric treat-
nii [121,122,127,129]. Among hospitalized patients with UTIs, ment of uncomplicated cystitis, cUTIs with risk of fluoroquino-
98%, 100%, 97%, and 100% of urinary isolates of P. aeruginosa, lones resistance, and hospital-acquired UTIs [6,10,93].
ceftazidime-non-susceptible P. aeruginosa, meropenem-non- Management of ESBL UTIs is challenging given the limited
susceptible P. aeruginosa, and A. baumannii were susceptible treatment options available outside the hospital setting.
to colistin, respectively [121,122]. Colistin is administered intra- Carbapenem-sparing strategy can be applied to treat mild–
venously to treat cUTIs and acute pyelonephritis. However, moderate UTIs due to ESBL-producing Enterobacteriaceae, parti-
colistin bladder irrigation (100,000 IU of colistin in 50 ml of cularly in case of E. coli, if the isolated organism is susceptible to
isotonic saline solution given through a single urinary catheter alternatives. However, alternatives to carbapenems should not
three times a day for 3–7 days) can be used to treat cystitis be used in the definitive treatment of severe infections caused
due to MDR-organisms such as Acinetobacter spp. in patients by these organisms or when the pathogen is not susceptible or
with kidney dysfunction who cannot be treated with systemic empiric treatment in patients at risk of infection by these organ-
colistin [130]. The rate of resistance to colistin remains low; isms [45,114–116]. Consultation with an infectious diseases spe-
however, with increased use, resistance is emerging in A. cialist and microbiologist for the management and treatment of
baumannii, K. pneumonia, MDR-Pseudomonas, and E. coli. The these infections including performing and interpretation of the
resistance to colistin occurs as a result of alterations in lipid A susceptibility testing is highly recommended.
as a consequence of chromosomal mutations and by mcr-1 Future options for antibiotic-resistant Gram-negative
gene that is carried on a plasmid and can transfer colistin organisms including plazomicin, eravacycline, carbavance
resistance [131,132]. Strategies to potentially decrease failure (meropenem/RPX7009), imipenem–cilistatin/relebactam, cef-
rate and resistance include optimizing dose, interval, and taroline–avibactam, and aztreonam–avibactam are currently
duration of therapy as well as combination therapy (for carba- in development phase [137–140].
penemase-producing K. pneumoniae strains, pathogen with an
MIC greater than 1 mg/l, or patients with creatinine clearance
Funding
greater than approximately 80 ml/min) [129].
This article was not funded.

Tigecycline
Declaration of interest
Tigecycline is a glycylcycline antibiotic and a derivative of
The authors have no relevant affiliations or financial involvement with any
minocycline [133]. Tigecycline is not usually recommended organization or entity with a financial interest in or financial conflict with
for the treatment of UTIs due to low peak serum concentra- the subject matter or materials discussed in the manuscript. This includes
tions, limited excretion into urine, and subsequent develop- employment, consultancies, honoraria, stock ownership or options, expert
ment of resistance [134]. In the absence of effective and safe testimony, grants or patents received or pending, or royalties.
antibiotic options for UTIs, high-dose tigecycline (e.g. loading
dose of 200–400 mg followed by 100–200 mg daily) can be
References
considered an alternate option to treat infections due to anti-
biotic-resistant organisms and minimize the risk of resistance 1. Tandogdu Z, Wagenlehner FME. Global epidemiology of urinary
[135]. Tigecycline should not be used as monotherapy for the tract infections. Curr Opin Infect Dis. 2016;29(1):73–79.
treatment of moderate-to-severe UTIs, particularly in the pre- 2. Foxman B. Urinary tract infection syndromes: occurrence, recur-
rence, bacteriology, risk factors, and disease burden. Infect Dis
sence of bacteremia and should be combined with other Clin North Am. 2014;28:1–13.
active antibiotic such as carbapenems, colistin, or aminoglyco- 3. Schappert SM, Rechtsteiner EA. Ambulatory medical care utilization
sides [65]. Tigecycline use was associated with increased all- estimates for 2007. Vital Health Stat. 2011;13:1–38.
cause mortality in patients with severe infections [136]. 4. Foxman B. The epidemiology of urinary tract infection. Nat Rev
Urol. 2010;7:653–660.
5. Magill SS, Edwards JR, Bamberg W, et al. Multistate point-preva-
lence survey of healthcare-associated infections. N Engl J Med.
Conclusion 2014;370:1198–1208.
6. Gupta K, Hooton TM, Naber KG, et al. Infectious Diseases Society of
The assessment of suspected UTI involves identifying charac- America; European Society for Microbiology and Infectious
teristic signs or symptoms associated with symptomatic UTI Diseases. International clinical practice guidelines for the treatment
and appropriate interpretation of three potential tests: bed- of acute uncomplicated cystitis and pyelonephritis in women: a
side urinary dipstick, UA, and urine culture [11]. Obtaining a 2010 update by the Infectious Diseases Society of America and the
European Society for Microbiology and Infectious Diseases. Clin
UA in the absence of consistent initial history and physical
Infect Dis. 2011;52(5):e103–20.
examination may lead to inappropriate use of diagnostic tests 7. Linhares I, Raposo T, Rodrigues A, et al. Frequency and antimicro-
such as urine culture and inappropriate antibiotic therapy for bial resistance patterns of bacteria implicated in community urin-
asymptomatic bacteriuria [15–17]. ary tract infections: a ten-year surveillance study (2000-2009). BMC
Consideration of the limited options for oral treatment of Infect Dis. 2013;13:19.
8. Hirsch EB, Zucchi PC, Chen A, et al. Susceptibility of multidrug-
ESBLs-Enterobacteriaceae infections, it may be prudent to
resistant Gram-negative urine isolates to oral antibiotics.
reserve fosfomycin for management of culture-confirmed Antimicrob Agents Chemother. 2016;60(5):3138–3140.
MDR infections rather than using it as a first-line agent for 9. Ironmonger D, Edeghere O, Gossain S, et al. Use of antimicrobial
empirical therapy of common UTIs [8,19,78,84,89]. resistance information and prescribing guidance for management
14 M. S. BADER ET AL.

of urinary tract infections: survey of general practitioners in the 28. Spoorenberg V, Hulscher ME, Akkermans RP, et al. Appropriate
West Midlands. BMC Infect Dis. 2016;16(1):226. antibiotic use for patients with urinary tract infections reduces
10. Hooton TM, Bradley SF, Cardenas DD, et al. Diagnosis, prevention, length of hospital stay. Clin Infect Dis. 2014;58:164–169.
and treatment of catheter-associated urinary tract infection in 29. Sanchez GV, Babiker A, Master RN, et al. Antibiotic resistance
adults: 2009 International Clinical Practice Guidelines from the among urinary isolates from female outpatients in the United
Infectious Diseases Society of America. Clin Infect Dis. 2010;50 States in 2003 and 2012. Antimicrob Agents Chemother.
(5):625–663. 2016;60:2680–2683.
11. Baron EJ, Miller JM, Weinstein MP, et al. A guide to utilization of the 30. Cek M, Tandogdu Z, Wagenlehner F, et al. Healthcare-associated
microbiology laboratory for diagnosis of infectious diseases: 2013 urinary tract infections in hospitalized urological patients-a global
recommendations by the Infectious Diseases Society of America perspective: results from the GPIU studies 2003–2010. World J Urol.
(IDSA) and the American Society for Microbiology (ASM)(a). Clin 2014;32:1587–1594.
Infect Dis. 2013;57(4):e22–e121. 31. Sanchez GV, Baird AM, Karlowsky JA, et al. Nitrofurantoin retains anti-
12. Bent S, Nallamothu BK, Simel DL, et al. Does this woman have an microbial activity against multidrug-resistant urinary Escherichia coli
acute uncomplicated urinary tract infection? JAMA. 2002;287 from US outpatients. J Antimicrob Chemother. 2014;69:3259–3262.
(20):2701–2710. 32. Jacoby GA. AmpC beta-lactamases. Clin Microbiol Rev.
13. Devillé WL, Yzermans JC, Van Duijn NP, et al. The urine dipstick test 2009;22:161–182.
useful to rule out infections. A Meta-Analysis of the Accuracy. BMC 33. Lee CH, Lee YT, Kung CH, et al. Risk factors of community-onset
Urol. 2004;4:4. urinary tract infections caused by plasmid-mediated AmpC β-lacta-
14. Ducharme J, Neilson S, Ginn JL. Can urine cultures and reagent test mase-producing Enterobacteriaceae. J Microbiol Immunol Infect.
strips be used to diagnose urinary tract infection in elderly emer- 2015;48(3):269–275.
gency department patients without focal urinary symptoms? Cjem. 34. Denisuik AJ, Lagacé-Wiens PR, Pitout JD, et al. Molecular epidemiol-
2007;9(2):87–92. ogy of extended-spectrum b-lactamase-, AmpC b-lactamase- and
15. Yin P, Kiss A, Leis JA. Urinalysis orders among patients admitted to carbapenemase-producing Escherichia coli and Klebsiella pneumoniae
the general medicine service. JAMA Intern Med. 2015;175 isolated from Canadian hospitals over a 5 year period: CANWARD
(10):1711–1713. 2007–11. J Antimicrob Chemother. 2013;68(Suppl 1):i57–i65.
16. Daley P, Penney C, Wakeham S, et al. Urinary tract infection diag- 35. Karlowsky JA, Denisuik AJ, Lagacé-Wiens PR, et al. In vitro activity
nosis and response to therapy in long-term care: a prospective of fosfomycin against Escherichia coli isolated from patients with
observational study. Can J Infect Dis Med Microbiol. 2015;26 urinary tract infections in Canada as part of the CANWARD sur-
(3):133–136. veillance study. Antimicrob Agents Chemother. 2014;58(2):1252–
17. Tomas ME, Getman D, Donskey CJ, et al. Overdiagnosis of urinary 1256.
tract infection and underdiagnosis of sexually transmitted infection 36. Hoban DJ, Lascols C, Nicolle LE, et al. Antimicrobial susceptibility of
in adult women presenting to an emergency department. J Clin Enterobacteriaceae, including molecular characterization of
Microbiol. 2015;53:2686–2692. extended-spectrum β-lactamase-producing species, in urinary
18. Johansen TE, Botto H, Cek M, et al. Critical review of current tract isolates from hospitalized patients in North America and
definitions of urinary tract infections and proposal of an EAU/ESIU Europe: results from the SMART study 2009–2010. Diagn
classification system. Int J Antimicrob Agents. 2011;38(Suppl.):64– Microbiol Infect Dis. 2012;74:62–67.
70. 37. Sorlozano A, Jimenez-Pacheco A, de Dios Luna Del Castillo J, et al.
19. Little P, Moore MV, Turner S, et al. Effectiveness of five different Evolution of the resistance to antibiotics of bacteria involved in
approaches in management of urinary tract infection: randomised urinary tract infections: a 7-year surveillance study. Am J Infect
controlled trial. Bmj. 2010;340:c199. Control. 2014;42(10):1033–1038.
20. Peterson J, Kaul S, Khashab M, et al. Identification and pretherapy 38. Meier S, Weber R, Zbinden R, et al. Extended-spectrum beta- lacta-
susceptibility of pathogens in patients with complicated urinary mase-producing Gram-negative pathogens in community-acquired
tract infection or acute pyelonephritis enrolled in a clinical study in urinary tract infections: an increasing challenge for antimicrobial
the United States from November 2004 through April 2006. Clin therapy. Infection. 2011;39:333–340.
Ther. 2007;29(10):2215–2221. 39. Castanheira M, Mendes RE, Jones RN, et al. Changing of the fre-
21. Mariappan P, Smith G, Moussa SA, et al. One week of ciprofloxacin quencies of β-Lactamase genes among Enterobacteriaceae in US
before percutaneous nephrolithotomy significantly reduces upper hospitals (2012-2014): activity of ceftazidime-avibactam tested
tract infection and urosepsis: a prospective controlled study. BJU against β-lactamase-producing isolates. Antimicrob Agents
Int. 2006;98:1075–1079. Chemother. 2016;60(8):4770–4777.
22. Lee JH, Lee YM, Cho JH. Risk factors of septic shock in bacteremic 40. Thaden JT, Fowler VG, Sexton DJ, et al. Increasing incidence of
acute pyelonephritis patients admitted to an ER. J Infect extended-spectrum β-lactamase-producing Escherichia coli in com-
Chemother. 2012;18:130–133. munity hospitals throughout the Southeastern United States. Infect
23. Laupland KB, Bagshaw SM, Gregson DB, et al. Intensive care unit Control Hosp Epidemiol. 2016;37(1):49–54.
acquired urinary tract infections in a regional critical care system. 41. Lee JH, Bae IK, Lee SH. New definitions of extended-spectrum β-
Crit Care. 2005;9:R60–5. lactamase conferring worldwide emerging antibiotic resistance.
24. Marschall J, Zhang L, Foxman B, et al. Both host and pathogen Med Res Rev. 2012;32:216–232.
factors predispose to Escherichia coli urinary source bacteremia in 42. Johnson JR, Urban C, Weissman SJ, et al. Molecular epidemiological
hospitalized patients. Clin Infect Dis. 2012;54:1692–1698. analysis of Escherichia coli sequence type ST131 (O25:H4)and
25. Magiorakos AP, Srinivasan A, Carey RB, et al. Multidrug-resistant, blaCTX-M-15 among extended-spectrum-b-lactamase-producing E.
extensively drug-resistant and pandrug-resistant bacteria: an inter- coli from the United States, 2000 to 2009. Antimicrob Agents
national expert proposal for interim standard definitions for Chemother. 2012;56:2364–2370.
acquired resistance. Clin Microbiol Infect. 2011;18:268–281. 43. Peirano G, Richardson D, Nigrin J, et al. High prevalence of ST131
26. Lee SS, Kim Y, Chung DR. Impact of discordant empirical therapy isolates producing CTX- M-15 and CTX-M-14 among extended
on outcome of community-acquired bacteremic acute pyelone- spectrum- β-lactamase-producing Escherichia coli isolates from
phritis. J Infect. 2011;62:159–164. Canada. Antimicrob Agents Chemother. 2010;54:1327–1330.
27. Yang YS, Ku CH, Lin JC, et al. Impact of extended-spectrum β- 44. Briongos-Figuero LS, Gómez-Traveso T, Bachiller-Luque P, et al.
lactamase-producing Escherichia coli and Klebsiella pneumoniae on Epidemiology, risk factors and comorbidity for urinary tract infec-
the outcome of community-onset bacteremic urinary tract infec- tions caused by extended-spectrum beta-lactamase (ESBL)-produ-
tions. J Microbiol Immunol Infect. 2010;43:194–199. cing enterobacteria. Int J Clin Pract. 2012;66(9):891–896.
POSTGRADUATE MEDICINE 15

45. Fournier D, Chirouze C, Leroy J, et al. Alternatives to carbapenems United States medical centers (2012-2014). Antimicrob Agents
in ESBL-producing Escherichia coli infections. Med Mal Infect. Chemother. 2016;60:4355–4360.
2013;43(2):62–66. 64. Ghazi IM, Crandon JL, Lesho EP, et al. Efficacy of humanized high-
46. Asbakan MI, Pullukcu H, Sipahi OR, et al. Nitrofurantion in the dose meropenem, cefepime, and levofloxacin against
treatment of extended-spectrum b-lactamase-producing Enterobacteriaceae isolates producing Verona integron-encoded
Escherichia coli-related lower urinary tract infection. Int J metallo-β-lactamase (VIM) in a murine thigh infection model.
Antimicrob Agents. 2012;40:554–556. Antimicrob Agents Chemother. 2015;59:7145–7147.
47. Titelman E, Iversen A, Kalin M, et al. Efficacy of pivmecillinam for 65. Falagas ME, Lourida P, Poulikakos P, et al. Antibiotic treatment of
treatment of lower urinary tract infection caused by extended- infections due to carbapenem-resistant Enterobacteriaceae: sys-
spectrum b-lactamase-producing Escherichia coli and Klebsiella tematic evaluation of the available evidence. Antimicrob Agents
pneumoniae. Microb Drug Resist. 2011;18:189–192. Chemother. 2014;58(2):654–663.
48. Auer S, Wojna A, Hell M. Oral treatment options for ambulatory 66. Cprek JB, Gallagher JC. Ertapenem-containing double-carbapenem
patients with urinary tract infections caused by extended-spec- therapy for treatment of infections caused by carbapenem-resis-
trum-beta-lactamase-producing Escherichia coli. Antimicrob tant Klebsiella pneumonia. Antimicrob Agents Chemother.
Agents Chemother. 2010;54(9):4006–4008. 2016;60:669–673.
49. Falagas ME, Vouloumanou EK, Togias AG, et al. Fosfomycin versus 67. Sievert DM, Ricks P, Edwards JR, et al. Antimicrobial-resistant
other antibiotics for the treatment of cystitis: a meta-analysis of pathogens associated with healthcare-associated infections sum-
randomized controlled trials. J Antimicrob Chemother. 2010;65 mary of data reported to the National Healthcare Safety Network at
(9):1862–1877. the Centers for Disease Control and Prevention, 2009–2010. Infect
50. Livermore DM, Warner M, Mushtaq S, et al. What remains against Control Hosp Epidemiol. 2013;34:1–14.
carbapenem-resistant Enterobacteriaceae? Evaluation of chloram- 68. Bouchillon SK, Badal RE, Hoban DJ, et al. Antimicrobial susceptibil-
phenicol, ciprofloxacin, colistin, fosfomycin, minocycline, nitrofur- ity of inpatient urinary tract isolates of Gram-negative bacilli in the
antoin, temocillin and tigecycline. Int J Antimicrob Agents. United States: results from the Study for Monitoring Antimicrobial
2011;37:415–419. Resistance Trends (SMART) program: 2009-2011. Clinical
51. Michalopoulos A, Virtzili S, Rafailidis P, et al. Intravenous fosfomycin Therapeutics. 2013;35(6):872–877.
for the treatment of nosocomial infections caused by carbapenem- 69. Deanna J. Buehrle, Ryan K. Shields, et al. Evaluation of the in vitro
resistant Klebsiella pneumoniae in critically ill patients: a prospec- activity of ceftazidime-avibactam and ceftolozane-tazobactam
tive evaluation. Clin Microbiol Infect. 2010;16:184–186. against meropenem-resistant Pseudomonas aeruginosa isolates.
52. Pilmis B, Delory T, Groh M, et al. Extended-spectrum beta-lacta- Antimicrob. Agents Chemother. 2016;60:3227–3231.
mase-producing Enterobacteriaceae (ESBL-PE) infections: are carba- 70. Yu M, Robinson K, Siegel C, et al. Complicated genitourinary tract
penem alternatives achievable in daily practice? Int J Infect Dis. infections and mimics. Curr Probl Diagn Radiol. 2016 Feb;11. [Epub
2015;39:62–67. ahead of print].
53. Jacob JT, Klein E, Laxminarayan R, et al. Vital signs: carbapenem- 71. Karlowsky JA, Lagacé-Wiens PR, Simner PJ, et al. Antimicrobial
resistant Enterobacteriaceae. MMWR Morb Mortal Wkly Rep. resistance in urinary tract pathogens in Canada from 2007 to
2013;62:165–170. 2009: CANWARD surveillance study. Antimicrob Agents
54. Cantón R, Akóva M, Carmeli Y, et al. Rapid evolution and spread of Chemother. 2011;55(7):3169–3175.
carbapenemases among Enterobacteriaceae in Europe. Clin 72. Linsenmeyer K, Strymish J, Gupta K. Two simple rules for improving
Microbiol Infect. 2012;18(5):413–431. the accuracy of empiric treatment of multidrug-resistant urinary
55. Guh AY, Bulens SN, Mu Y, et al. Epidemiology of carbapenem- tract infections. Antimicrob Agents Chemother. 2015;59:7593–7596.
resistant Enterobacteriaceae in 7 US communities, 2012-2013. 73. Koningstein M, van der Bij AK, De Kraker ME, et al.
JAMA. 2015;314(14):1479–1487. Recommendations for the empirical treatment of complicated urin-
56. Castanheira M, Mendes RE, Woosley LN, et al. Trends in carbape- ary tract infections using surveillance data on antimicrobial resis-
nemase-producing Escherichia coli and Klebsiella spp. from Europe tance in The Netherlands. PLoS One. 2014;9:e86634.
and the Americas: report from the SENTRY antimicrobial surveil- 74. Barlam TF, Cosgrove SE, Abbo LM, et al. Implementing an antibiotic
lance programme (2007–09). J Antimicrob Chemother. stewardship program: guidelines by the Infectious Diseases Society
2011;66:1409–1411. of America and the Society for Healthcare Epidemiology of
57. McLaughlin M, Advincula MR, Malczynski M, et al. Correlations of America. Clin Infect Dis. 2016;62(10):e51–77.
antibiotic use and carbapenem resistance in Enterobacteriaceae. 75. Knottnerus BJ, Geerlings SE, Moll Van Charante EP, et al. Women
Antimicrob Agents Chemother. 2013;57:5131–5133. with symptoms of uncomplicated urinary tract infection are often
58. Chang H-J, Hsu P-C, Yang -C-C, et al. Risk factors and outcomes of willing to delay antibiotic treatment. BMC Fam Pract. 2013;14:71.
carbapenem-nonsusceptible Escherichia coli bacteremia: a matched 76. Christiaens TC, De Meyere M, Verschraegen G, et al. Randomised
case-control study. J Microbiol Immunol Infect. 2011;44:125–130. controlled trial of nitrofurantoin versus placebo in the treatment of
59. Shilo S, Assous MV, Lachish T, et al. Risk factors for bacteriuria with uncomplicated urinary tract infection in adult women. Br J Gen
carbapenem-resistant Klebsiella pneumoniae and its impact on mor- Pract. 2002;52(482):729–734.
tality: a case-control study. Infection. 2013;41(2):503–509. 77. Ferry SA, Holm SE, Stenlund H, et al. Clinical and bacteriological
60. Hauck C, Cober E, Richter SS, et al. Spectrum of excess mortality outcome of different doses and duration of pivmecillinam com-
due to carbapenem-resistant Klebsiella pneumoniae infections. Clin pared with placebo therapy of uncomplicated lower urinary tract
Microbiol Infect. 2016;22(6):513–519. infection in women: the LUTIW project. Scand J Prim Health Care.
61. Esterly JS, Wagner J, McLaughlin MM, et al. Evaluation of clinical 2007;25(1):49–57.
outcomes in patients with bloodstream infections due to Gram- 78. Huttner A, Verhaegh EM, Harbarth S, et al. Nitrofurantoin revisited:
negative bacteria according to carbapenem MIC stratification. a systematic review and meta-analysis of controlled trials. J
Antimicrob Agents Chemother. 2012;56:4885–4890. Antimicrob Chemother. 2015;70(9):2456–2464.
62. Tambyah PA, Hara GL, Daikos GL, et al. Treatment of extensively 79. Liu H-Y, Lin H-C, Lin Y-C, et al. Antimicrobial susceptibilities of urinary
drug-resistant Gram-negative infections in critically ill patients: out- extended-spectrum beta-lactamase- producing Escherichia coli and
come of a consensus meeting at the 13th Asia-Pacific Congress of Klebsiella pneumoniae to fosfomycin and nitrofurantoin in a teaching
Clinical Microbiology and Infection, October 2012. J Global hospital in Taiwan. J Microbiol Immunol Infect. 2011;44:364–368.
Antimicrob Res. 2013;1:117–122. 80. Geerts AF, Eppenga WL, Heerdink R, et al. Ineffectiveness and
63. Sader HS, Castanheira M, Flamm RK, et al. Antimicrobial activity of adverse events of nitrofurantoin in women with urinary tract infec-
ceftazidime-avibactam and comparator agents tested against tion and renal impairment in primary care. Eur J Clin Pharmacol.
Gram-negative organisms isolated from urinary tract infections in 2013;69:1701–1707.
16 M. S. BADER ET AL.

81. Oplinger M, Andrews CO. Nitrofurantoin contraindication in infected by cefepime-susceptible dose-dependent isolates.
patients with a creatinine clearance below 60mL/min: looking for Antimicrob Agents Chemother. 2015;59:7558–7563.
the evidence. Ann Pharmacother. 2013;47:106–111. 101. Lee NY, Lee CC, Huang WH, et al. Cefepime therapy for monomi-
82. Singh N, Gandhi S, McArthur E, et al. Kidney function and the use of crobial bacteremia caused by cefepime-susceptible extended-spec-
nitrofurantoin to treat urinary tract infections in older women. trum beta-lactamase-producing Enterobacteriaceae: MIC matters.
Cmaj. 2015;187(9):648–656. Clin Infect Dis. 2013;56:488–495.
83. Falagas ME, Vouloumanou EK, Samonis G, et al. Fosfomycin. Clin 102. Chopra T, Marchaim D, Veltman J, et al. Impact of cefepime therapy
Microbiol Rev. 2016;29(2):321–347. on mortality among patients with bloodstream infections caused
84. Vardakas KZ, Legakis NJ, Triarides N, et al. Susceptibility of con- by extended-spectrum-beta-lactamase-producing Klebsiella pneu-
temporary isolates to fosfomycin: a systematic review of the litera- moniae and Escherichia coli. Antimicrob Agents Chemother.
ture. Int J Antimicrob Agents. 2016;47(4):269–285. 2012;56:3936–3942.
85. Sastry S, Clarke LG, Alrowais H, et al. Clinical appraisal of fosfomycin 103. Zhanel GG, Lawson CD, Adam H, et al. Ceftazidime-avibactam: a
in the era of antimicrobial resistance. Antimicrob Agents novel cephalosporin/β-lactamase inhibitor combination. Drugs.
Chemother. 2015;59(12):7355–7361. 2013;73:159–177.
86. Senol S, Tasbakan M, Pullukcu H, et al. Carbapenem versus fosfo- 104. De Jonge BL, Karlowsky JA, Kazmierczak KM, et al. In vitro suscept-
mycin tromethanol in the treatment of extended- spectrum beta- ibility to ceftazidime-avibactam of carbapenem-nonsusceptible
lactamase-producing Escherichia coli-related complicated lower Enterobacteriaceae isolates collected during the INFORM Global
urinary tract infection. J Chemother. 2010;22:355–357. Surveillance Study (2012 to 2014). Antimicrob Agents Chemother.
87. Veve MP, Wagner JL, Kenney RM, et al. Comparison of fosfomycin 2016;60(5):3163–3169.
to ertapenem for outpatient or step-down therapy of extended- 105. Sader HS, Castanheira M, Mendes RE, et al. Ceftazidime-avibactam
spectrum β-lactamase urinary tract infections. Int J Antimicrob activity against multidrug-resistant Pseudomonas aeruginosa iso-
Agents. 2016;48(1):56–60. lated in U.S. medical centers in 2012 and 2013. Antimicrob
88. Linsenmeyer K, Strymish J, Weir S, et al. Activity of fosfomycin Agents Chemother. 2015;59(6):3656–3659.
against extended-spectrum-β-lactamase-producing uropathogens 106. Vazquez JA, González Patzán LD, Stricklin D, et al. Efficacy and
in patients in the community and hospitalized patients. safety of ceftazidime-avibactam versus imipenem-cilastatin in the
Antimicrob Agents Chemother. 2015;60(2):1134–1136. treatment of complicated urinary tract infections, including acute
89. Oteo J, Bautista V, Lara N, et al. Parallel increase in community use pyelonephritis, in hospitalized adults: results of a prospective,
of fosfomycin and resistance to fosfomycin in extended-spectrum investigator-blinded, randomized study. Curr Med Res Opin.
beta-lactamase (ESBL)-producing Escherichia coli. J Antimicrob 2012;28:1921–1931.
Chemother. 2010;65(11):2459–2463. 107. Mendes RE, Castanheira M, Gasink L, et al. β-Lactamase character-
90. Antoniou T, Hollands S, Macdonald EM, et al. Trimethoprim–sulfa- ization of Gram-negative pathogens recovered from patients
methoxazole and risk of sudden death among patients taking enrolled in the phase 2 trials for ceftazidime-avibactam: clinical
spironolactone. CMAJ. 2015;187:E138–43. efficacies analyzed against subsets of molecularly characterized
91. Fralick M, Macdonald EM, Gomes T, et al. Co-trimoxazole and isolates. Antimicrob Agents Chemother. 2015;60(3):1328–1335.
sudden death in patients receiving inhibitors of renin-angiotensin 108. Carmeli Y, Armstrong J, Laud PJ, et al. Ceftazidime-avibactam or
system: population based study. Bmj. 2014;349:6196. best available therapy in patients with ceftazidime-resistant
92. Bidell MR, Palchak M, Mohr J, et al. Fluoroquinolone and third- Enterobacteriaceae and Pseudomonas aeruginosa complicated urin-
generation-cephalosporin resistance among hospitalized patients ary tract infections or complicated intra-abdominal infections
with urinary tract infections due to Escherichia coli: do rates vary by (REPRISE): a randomised, pathogen-directed, phase 3 study.
hospital characteristics and geographic region? Antimicrob Agents Lancet Infect Dis. 2016;16:661–673.
Chemother. 2016;60:3170–3173. 109. Ceftolozane/tazobactam (Zerbaxa) – a new intravenous antibiotic.
93. Fasugba O, Gardner A, Mitchell BG, et al. Ciprofloxacin resistance in Med Lett Drugs Ther. 2015;57:31–33.
community- and hospital-acquired Escherichia coli urinary tract 110. Wagenlehner FM, Umeh O, Steenbergen J, et al. Ceftolozane-tazo-
infections: a systematic review and meta-analysis of observational bactam compared with levofloxacin in the treatment of compli-
studies. BMC Infect Dis. 2015 Nov 25;15:545. cated urinary-tract infections, including pyelonephritis: a
94. Van Der Starre WE, Van Nieuwkoop C, Paltansing S, et al. Risk randomised, double-blind, phase 3 trial (ASPECT-cUTI). Lancet.
factors for fluoroquinolone-resistant Escherichia coli in adults with 2015;385(9981):1949–1956.
community-onset febrile urinary tract infection. J Antimicrob 111. Huntington JA, Sakoulas G, Umeh O, et al. Efficacy of ceftolozane/
Chemother. 2011;66(3):650–656. tazobactam versus levofloxacin in the treatment of complicated
95. Rattanaumpawan P, Tolomeo P, Bilker WB, et al. Risk factors for urinary tract infections (cUTIs) caused by levofloxacin-resistant
fluoroquinolone resistance in Gram-negative bacilli causing health- pathogens: results from the ASPECT-cUTI trial. J Antimicrob
care-acquired urinary tract infections. J Hosp Infect. 2010;76:324– Chemother. 2016);71(7):2014–2021.
327. 112. European Committee on Antimicrobial Susceptibility Testing.
96. Rooney PJ, O’Leary MC, Loughrey AC, et al. Nursing homes as a Breakpoint tables for interpretation of MICs and zone diameters,
reservoir of extended-spectrum β-lactamase (ESBL)-producing version 6. [cited 2016 Jun 2]. Available from: www.eucast.org.
ciprofloxacin-resistant Escherichia coli. J Antimicrob Chemother. 113. Hooton TM, Scholes D, Gupta K, et al. Amoxicillin-clavulanate vs
2009;64:635–641. ciprofloxacin for the treatment of uncomplicated cystitis in women:
97. O’Brien KA, Zhang J, Mauldin PD, et al. Impact of a stewardship- a randomized trial. JAMA. 2005;293(8):949–955.
initiated restriction on empirical use of ciprofloxacin on nonsus- 114. Vardakas KZ, Tansarli GS, Rafailidis PI, et al. Carbapenems versus
ceptibility of Escherichia coli urinary isolates to ciprofloxacin. alternative antibiotics for the treatment of bacteraemia due to
Pharmacotherapy. 2015;35(5):464–469. Enterobacteriaceae producing extended-spectrum b-lactamases: a
98. Tamma PD, Girdwood SC, Gopaul R, et al. The use of cefepime for systematic review and meta-analysis. J Antimicrob Chemother.
treating AmpC beta-lactamase-producing Enterobacteriaceae. Clin 2012;67:2793–2803.
Infect Dis. 2013;57:781–788. 115. Rodriguez-Bano J, Navarro MD, Retamar P, et al. b-Lactam/b-lactam
99. Blanchette LM, Kuti JL, Nicolau DP, et al. Clinical comparison of inhibitor combinations for the treatment of bacteremia due to
ertapenem and cefepime for treatment of infections caused by extended-spectrum b-lactamase-producing Escherichia coli: a post
AmpC beta-lactamase-producing Enterobacteriaceae. Scand J hoc analysis of prospective cohorts. Clin Infect Dis. 2012;54:167–
Infect Dis. 2014;46(11):803–808. 174.
100. Lee NY, Lee CC, Li CW, et al. Cefepime therapy for monomicrobial 116. Tamma PD, Han JH, Rock C, et al. Carbapenem therapy is asso-
Enterobacter cloacae bacteremia: unfavorable outcomes in patients ciated with improved survival compared with piperacillin-
POSTGRADUATE MEDICINE 17

tazobactam for patients with extended-spectrum β-lactamase bac- tertiary hospital in Spain in the setting of a prolonged, hospital-
teremia. Clin Infect Dis. 2015;60(9):1319–1325. wide outbreak. J Antimicrob Chemother. 2013;68(1):89–96.
117. Kang CI, Park SY, Chung DR, et al. Piperacillin-tazobactam as an 128. Cobussen M, De Kort JM, Dennert RM, et al. No increased risk of
initial empirical therapy of bacteremia caused by extended-spec- acute kidney injury after a single dose of gentamicin in patients
trum b-lactamase-producing Escherichia coli and Klebsiella pneumo- with sepsis. Infect Dis (Lond). 2015;15:1–7.
niae. J Infect. 2012;64:533–534. 129. Nation RL, Li J, Cars O, et al. Framework for optimisation of the
118. Bliziotis IA, Samonis G, Vardakas KZ, et al. Clinical outcomes with clinical use of colistin and polymyxin B: the Prato polymyxin con-
extended or continuous versus short-term intravenous infusion of sensus. Lancet Infect Dis. 2015;15(2):225–234.
carbapenems and piperacillin/tazobactam: a systematic review and 130. Giua R, Pedone C, Cortese L, et al. Colistin bladder instillation, an
meta-analysis. Clin Infect Dis. 2005 Jul 15;41(2):149–158. alternative way of treating multi-resistant Acinetobacter urinary
119. Rodriguez-Bano J, Mingorance J, Fernandez- Romero N, et al. tract infection: a case series and review of literature. Infection.
Virulence profiles of bacteremic extended-spectrum beta-lacta- 2014;42(1):199–202.
mase-producing Escherichia coli: association with epidemiological 131. Qureshi ZA, Hittle LE, O’Hara JA, et al. Colistin-resistant acinetobac-
and clinical features. PloS One. 2012;7:e44238. ter baumannii: beyond carbapenem resistance. Clin Infect Dis.
120. Trad MA, Zhong LH, Llorin RM, et al. Ertapenem in outpatient 2015;60(9):1295–1303.
parenteral antimicrobial therapy for complicated urinary tract 132. Liu YY, Wang Y, Walsh TR, et al. Emergence of plasmid-mediated
infections. J Chemother. 2016 May 30;1–5. [Epub ahead of print]. colistin resistance mechanism MCR-1 in animals and human beings
121. Morrow BJ, Pillar CM, Deane J, et al. Activities of carbapenem and in China: a microbiological and molecular biological study. Lancet
comparator agents against contemporary US Pseudomonas aerugi- Infect Dis. 2016;16(2):161–168.
nosa isolates from the CAPITAL surveillance program. Diagn 133. Pankey GA. Tigecycline. J Antimicrob Chemother. 2005;56:470–
Microbiol Infect Dis. 2013;75(4):412–416. 480.
122. Flamm RK, Farrell DJ, Sader HS, et al. Ceftazidime/avibactam activ- 134. van Duin D, Cober ED, Richter SS, et al. Tigecycline therapy for
ity tested against Gram-negative bacteria isolated from blood- carbapenem-resistant K. pneumoniae (CRKP) bacteriuria leads to
stream, pneumonia, intra-abdominal and urinary tract infections tigecycline resistance. Clin Microbiol Infect. 2014;20(12):O1117–20.
in US medical centres (2012). J Antimicrob Chemother. 135. Brust K, Evans A, Plammons R. Tigecycline in treatment of multi-
2014;69:1589–1598. drug resistant Gram-negative bacillus urinary tract infections: a
123. Asakura T, Ikeda M, Nakamura A, et al. Efficacy of empirical therapy systematic review. J Antimicrob Chemother. 2014;69:2606–2610.
with non-carbapenems for urinary tract infections with extended- 136. Prasad P, Sun J, Danner RL, et al. Excess deaths associated with
spectrum beta-lactamase-producing Enterobacteriaceae. Int J Infect tigecycline after approval based on noninferiority trials. Clin Infect
Dis. 2014;29:91–95. Dis. 2012;54:1699–1709.
124. Matsumura Y, Yamamoto M, Nagao M, et al. Cefmetazole and 137. Connolly L, Serio A, Riddle V, et al. Baseline pathogens and patient
flomoxef for the treatment of extended-spectrum β-lactamase-pro- outcomes in a phase 2 study comparing plazomicin (ACHN-490) to
ducing Escherichia coli bacteremia: a multicenter retrospective levofloxacin in complicated urinary tract infection (cUTI) including
study. Antimicrob Agents Chemother. 2015;59(9):5107–5113. acute pyelonephritis (AP) presentation O200. 25th European
125. Vidal L, Gafter-Gvili A, Borok S, et al. Efficacy and safety of amino- Congress of Clinical Microbiology and Infectious Diseases
glycoside monotherapy: systematic review and meta-analysis of (ECCMID); 25-28 April 2015; Copehhagen.
randomized controlled trials. J Antimicrob Chemother. 2007;60 138. Grossman TH, Murphy TM, Slee AM, et al. Eravacycline (TP-434) is
(2):247–257. efficacious in animal models of infection. Antimicrob Agents
126. Ipekci T, Seyman D, Berk H, et al. Clinical and bacteriological Chemother. 2015;59(5):2567–2571.
efficacy of amikacin in the treatment of lower urinary tract infec- 139. Livermore DM, Mushtaq S. Activity of biapenem (RPX2003) com-
tion caused by extended-spectrum beta-lactamase-producing bined with the boronate β-lactamase inhibitor RPX7009 against
Escherichia coli or Klebsiella pneumonia. J Infect Chemother. carbapenem-resistant Enterobacteriaceae. J Antimicrob
2014;20:762–767. Chemother. 2013;68(8):1825–1831.
127. Paño-Pardo JR, Ruiz-Carrascoso G, Navarro-San Francisco C, et al. 140. Shlaes DM. New β-lactam-β-lactamase inhibitor combinations in
Infections caused by OXA-48-producing Klebsiella pneumoniae in a clinical development. Ann N Y Acad Sci. 2013;1277:105–114.

Potrebbero piacerti anche