You are on page 1of 18

Postgraduate Medicine

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/ipgm20

Treatment of urinary tract infections in the era of


antimicrobial resistance and new antimicrobial
agents

Mazen S. Bader, Mark Loeb, Daniela Leto & Annie A. Brooks

To cite this article: Mazen S. Bader, Mark Loeb, Daniela Leto & Annie A. Brooks (2020) Treatment
of urinary tract infections in the era of antimicrobial resistance and new antimicrobial agents,
Postgraduate Medicine, 132:3, 234-250, DOI: 10.1080/00325481.2019.1680052

To link to this article: https://doi.org/10.1080/00325481.2019.1680052

Published online: 24 Oct 2019.

Submit your article to this journal

Article views: 3309

View related articles

View Crossmark data

Citing articles: 38 View citing articles

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


https://www.tandfonline.com/action/journalInformation?journalCode=ipgm20
POSTGRADUATE MEDICINE
2020, VOL. 132, NO. 3, 234–250
https://doi.org/10.1080/00325481.2019.1680052

CLINICAL FOCUS: GASTROENTEROLOGY, HEPATOLOGY & NEPHROLOGY


REVIEW

Treatment of urinary tract infections in the era of antimicrobial resistance and new
antimicrobial agents
Mazen S. Badera, Mark Loebb, Daniela Letoc and Annie A. Brooksd
a
Staff Physician, Department of Medicine, Hamilton Health Sciences, Juravinski hospital and Cancer Centre, Hamilton, Ontario, Canada; bDepartments of
Pathology & Molecular Medicine and Clinical, Epidemiology & Biostatistics, McMaster University, Hamilton, Ontario, Canada; cDepartment of Medicine
and Pathology and Molecular Medicine, McMaster University, Hamilton Health Sciences, Juravinski Hospital and Cancer Centre, Hamilton, Ontario,
Canada; dDepartment 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 concern due Received 22 June 2019
to limited treatment options. Knowledge of the common uropathogens in addition to local susceptibility Accepted 10 October 2019
patterns is essential in determining appropriate empiric antibiotic therapy of UTIs. The recommended first-line KEYWORDS
empiric antibiotic therapy for acute uncomplicated bacterial cystitis in otherwise healthy adult nonpregnant Antibiotic resistance; cystitis;
females is a 5-day course of nitrofurantoin, a 3-g single dose of fosfomycin tromethamine, or a 5-day course of enterobacteriales; gram-
pivmecillinam. High rates of resistance for trimethoprim-sulfamethoxazole and ciprofloxacin preclude their negative bacteria;
use as empiric treatment of UTIs in several communities, particularly if patients who were recently exposed to pyelonephritis; urinary tract
them or in patients who are at risk of infections with extended-spectrum β-lactamases (ESBLs)-producing infections
Enterobacteriales. Second-line options include oral cephalosporins such as cephalexin or cefixime, fluoroqui-
nolones and β-lactams, such as amoxicillin-clavulanate. Current treatment options for UTIs due to AmpC- β -
lactamase-producing Enterobacteriales include nitrofurantoin, fosfomycin, pivmecillinam, fluoroquinolones,
cefepime, piperacillin–tazobactam and carbapenems. Treatment oral options for UTIs due to ESBLs-E coli
include nitrofurantoin, fosfomycin, pivmecillinam, amoxicillin-clavulanate, finafloxacin, and sitafloxacin while
pivmecillinam, fosfomycin, finafloxacin, and sitafloxacin are treatment oral options for ESBLs- Klebsiella
pneumoniae. Parenteral treatment options for UTIs due to ESBLs–producing Enterobacteriales include piper-
acillin-tazobactam (for ESBL-E coli only), carbapenems including meropenem/vaborbactam, imipenem/cilas-
tatin-relebactam, and sulopenem, ceftazidime-avibactam, ceftolozane-tazobactam, aminoglycosides
including plazomicin, cefiderocol, fosfomycin, sitafloxacin, and finafloxacin. Ceftazidime-avibactam, merope-
nem/vaborbactam, imipenem/cilastatin-relebactam, colistin, fosfomycin, aztreonam and ceftazidime-
avibactam, aztreonam and amoxicillin-clavulanate, aminoglycosides including plazomicin, cefiderocol, tigecy-
cline are treatment options for UTIs caused by carbapenem-resistant Enterobacteriales (CRE). Treatment
options for UTIs caused by multidrug resistant (MDR)-Pseudomonas spp. include fluoroquinolones, ceftazi-
dime, cefepime, piperacillin-tazobactam, carbapenems including imipenem-cilastatin/relebactam, merope-
nem, and fosfomycin, ceftolozane-tazobactam, ceftazidime-avibactam, aminoglycosides including plazomicin,
aztreonam and ceftazidime-avibactam, cefiderocol, and colistin. It is important to use the new antimicrobials
wisely for treatment of UTIs caused by MDR-organisms to avoid resistance development.

Introduction Management of UTIs requires a systematic approach to


confirm the presence of infection and its type (site and either
Urinary tract infections (UTIs) are one of the most common
complicated or uncomplicated), assess risk factors of infection
bacterial infections encountered by clinicians [1]. UTIs are classi-
with antibiotic-resistant organisms, and to select the optimal
fied according to the site of infection into upper (pyelonephritis)
dose, route and duration of the empiric antibiotic regimen
and lower UTIs (urethritis, cystitis, prostatitis) [2,3]. UTIs are
based on a local antibiogram [2,5].
further classified into complicated or uncomplicated infections
This review is an update of our previously published article
according to the presence of certain risk factors [2,3].
on this topic [6]. It will focus on the role of both old and new
Gram-negative bacteria, specifically Enterobacteriales, are com-
antimicrobial agents in the treatment of UTIs.
mon causes of UTIs [1]. UTIs due to antibiotic-resistant Gram-
negative bacilli are a challenge due to increasing prevalence and
limited oral treatment options. Examples of these organisms
Uncomplicated cystitis
include AmpC- β -lactamase- and extended-spectrum β-
lactamases (ESBLs)-producing Enterobacteriales, carbapenem- An uncomplicated lower UTI is usually defined as acute
resistant Enterobacteriales (CRE), multidrug-resistant (MDR) and cystitis occurring in a healthy, premenopausal, nonpregnant
Pseudomonas aeruginosa [4]. female with no known structural urological abnormalities

CONTACT Mazen S. Bader msbader1@gmail.com Department of Medicine, Juravinski Hospital and Cancer Centre, 711 Concession Street, Hamilton, Ontario L8V1C3
© 2019 Informa UK Limited, trading as Taylor & Francis Group
POSTGRADUATE MEDICINE 235

[2]. Uncomplicated UTIs are responsible for a large propor- a 3-g fosfomycin trometamol dose administered to 40 healthy
tion of all antibiotic prescriptions and pathogen resistance is women with normal renal function [21].
increasing worldwide which requires a responsible and wise In an open-label randomized trial of 513 women with
antibiotic prescription across all health-care professions uncomplicated lower UTIs, nitrofurantoin (100 mg thrice
[2,5,6]. Urine culture and antimicrobial susceptibility testing daily for 5 days) was found to be superior to fosfomycin (a
for patients with uncomplicated acute cystitis are not single 3-g dose) clinically (symptoms resolution at 2 weeks
recommended by current guidelines because of lack of 75% vs. 66%, P= 0.03 and at 4 weeks 70% vs. 58%, P= 0.004)
management impact [5,6]. Acute uncomplicated cystitis is and microbiologically (cure at 4 weeks 74% vs. 63%. P= 0.04),
most commonly due to Enterobacteriales, enterococci and even in UTIs due to E coli [22].
Staphylococcus saprophyticus. The most commonly isolated Therefore, until there is further data, nitrofurantoin should
Enterobacteriales spp are Escherichia coli, Klebsiella spp, and be used as first-line option for treatment of acute cystitis
Proteus spp., which are responsible for over 80% of uncom- except in case of resistance, allergy or kidney dysfunction
plicated UTIs [7,8]. (creatinine clearance <30 mL/min) then fosfomycin can be
The first-line empiric antibiotics for treating acute bacterial used as alternative while monitoring for clinical failure.
cystitis in otherwise healthy adult non-pregnant females that are Trimethoprim-sulfamethoxazole is no longer recommended
recommended by the Infectious Diseases Society of America as empiric treatment option for uncomplicated cystitis in sev-
(IDSA) includes 5 days of nitrofurantoin, a three-day course of eral geographical locations due to high resistance rates of
double-strength trimethoprim-sulfamethoxazole in settings urinary E. coli which in excess of 20% particularly in large
where the prevalence of trimethoprim-sulfamethoxazole resis- urban areas (Table 1). However, regional difference in antibio-
tance to E coli is <20%, or a 3 g single dose of fosfomycin tic resistance patterns should be considered [7,9,10,23].
trometamol. Fluoroquinolones and oral β-lactams (e.g., amoxicil- Trimethoprim-sulfamethoxazole can be used as empiric or
lin-clavulanate, cephalexin) are second-line therapies (Table 1) definitive treatment of uncomplicated cystitis in case of resis-
[2]. However, selection of empirical antimicrobial agent should tance and allergy to first-line antibiotics (nitrofurantoin and
be based on local or regional susceptibility data and it is recom- fosfomycin), UTIs due to Enterobacteriales other than E coli
mended to choose the most appropriate and narrowest effective (Enterobacter sp, klebsiella pneumoniae, proteus mirabilis, serra-
antibiotic to combat the rise in resistance due to inappropriate tia marcescens) and stenotrophomonas maltophila [8]. It should
use of broad-spectrum antibiotics [2,5]. be used with caution in elderly patients due to risk of acute
Nitrofurantoin, fosfomycin, and pivmecillinam are the most kidney injury and hyperkalemia [24].
active antimicrobial agents in vitro against E. coli, including Fluoroquinolones are not recommended for treatment of
multi-drug resistant (MDR) isolates such as ESBL- and AmpC- β uncomplicated cystitis due to increasing resistance rates, par-
-lactamase-producing E coli, isolated among outpatients with ticularly among E coli, and safety issues (Table 1) [25].
acute cystitis. Furthermore, the resistance rates to these anti- Resistance rates of urinary E. coli to fluoroquinolones are
biotics are relatively stable over time [7,9,10]. increasing over time and can reach up in excess of 20%
Meta-analysis of 27 controlled trials including 4807 patients [7,9,10,23]. However, fluoroquinolones can be used as empiric
found similar clinical cure rates, but nonsignificant lower or definitive treatment of uncomplicated cystitis in case of
microbiological efficacy, in patients with UTI treated with resistance and allergy to first-line antibiotics (nitrofurantoin
either nitrofurantoin or other antibiotics such as trimetho- and fosfomycin), kidney dysfunction (creatinine clearance
prim/sulfamethoxazole, ciprofloxacin and amoxicillin [11]. <30 mL/min), UTIs due to Enterobacteriales other than E coli
However, nitrofurantoin is favored over fluoroquinolones (Enterobacter sp, klebsiella pneumoniae, proteus mirabilis, pro-
because it is primarily used for treatment of UTIs, narrow videncia sp, serratia marcescens), pseudomonas aeruginosa, and
spectrum, and safety profile (Table 1) [12]. stenotrophomonas maltophila [8].
Fosfomycin tromethamine, a soluble salt of fosfomycin is Oral cephalosporins (cephalexin, cefpodoxime, ceftibuten,
approved as single-dose oral therapy for uncomplicated UTIs cefadroxil, cefixime) are the fourth most active antimicrobial
in women caused by E. coli and Enterococcus faecalis (Table 1) agents, after nitrofurantoin, fosfomycin, and pivmecillinam,
[2]. Fosfomycin is not approved for the treatment of UTIs due against E. coli (Table 1) [7,8,26–28]. However, cephalexin is
to Klebsiella spp. Although it is active against other antibiotic- among the broad-spectrum antibiotics that are associated with
resistant gram-negative organisms, data supporting its efficacy increased rates of drug-related adverse events and antibiotic-
for treatment of MDR-uropathogens or complicated urinary associated diarrhea [29]. For unclear reasons, elderly patients
tract infections (cUTIs) are limited and there is a concern of with UTIs who were prescribed cephalexin had greater risk of
increasing resistance with its widespread use [13–19]. hospitalization due to sepsis and death [30]. The outcome was
The interpretation of fosfomycin susceptibility varies world- not adjusted for the severity of infection and patients who
wide, with different sensitivity thresholds proposed by labora- received cephalexin might have more severe infection than
tory regulatory bodies and variation between susceptibility patients who received nitrofurantoin. However, oral cephalos-
testing methods. Fosfomycin susceptibility testing has low porins can be used as empiric or definitive treatment of uncom-
sensitivity for the detection of resistant isolates and is highly plicated cystitis in case of resistance and allergy to first-line
method dependent on low agreement rate for K. pneumoniae antibiotics (nitrofurantoin and fosfomycin), kidney dysfunction
[20]. Finally, Wijma et al. found a considerable inter-individual (creatinine clearance <30 mL/min), UTIs due to Enterobacteriales
pharmacokinetic variability in the urinary concentrations after other than E coli (Klebsiella pneumoniae, proteus mirabilis) [8].
236

Table 1. First- and second-line oral treatment options for uncomplicated cystitis.

Anti-infective Dosing Advantages Disadvantages Adverse effects Comments


Nitrofurantoin 100 mg twice daily with Active against common urinary Lacks activity against Serratia marcescens, Nausea First line for acute uncomplicated
monohydrate food for 5 days pathogens including Staphylococcus Morganella spp, Proteus mirabilis, Headache cystitis
macrocrystals saprophyticus, Enterococcus including Providencia spp, Pseudomonas Cough Not recommended for empiric
VRE, Escherichia coli, Klebsiella aeruginosa, Stenotrophomonas Dyspnea treatment of hospital-acquired
pneumonia, Citrobacter spp, and maltophila, Acinetobacter spp, and CRE Rare side effects include acute allergic UTIs or acute pyelonephritis
AmpC- β -lactamase and ESBLs- High rates of resistance to Klebsiella pneumonitis, bone marrow suppression,
M. S. BADER ET AL.

producing Gram-negative organisms pneumonia and Enterobacter spp, and hepatotoxicity


High urinary concentration Not recommended for CrCl<30ml/min Risk of hemolysis in patients with glucose-
Low rates of resistance High failure rate with short course 6-phosphate dehydrogenase deficiency,
Low risk of Clostridium difficile infection therapy (3 days) neuropathy in patients with chronic kidney
Pregnancy Category B Contraindicated for pyelonephritis due to disease
low plasma concentrations Alters methotrexate clearance
Fosfomycin 3 g sachet as a single dose Active against common urinary Lacks activity against Morganella spp, Diarrhea Single dose therapy is not
tromethamine. pathogens including Enterococci Acinetobacter spp., Stenotrophomonas Nausea, vomiting recommended in males
including VRE, Escherichia coli, maltophilia and Staphylococcus Dyspepsia Off label use: cUTIs dose: 3 g once
Klebsiella pneumonia, Proteus mirabilis, saprophyticus Headache daily for 3–5 days or every
Pseudomonas aeruginosa, Increasing resistance of ESBLs- Klebsiella Vaginitis 48–72 h for 3–5 doses
AmpC- β -lactamase and ESBLs- pneumonia and ESBLs 025b/B2 E. coli Esophageal discomfort Not recommended for empiric
producing Gram-negative organisms strains Transient elevation of liver enzymes treatment of hospital-acquired
and KPC- and MBLs-producing CRE High clinical failure with Pseudomonas UTIs
Low rates of resistance and no cross aeruginosa
resistance with other antimicrobial Oral formulation contraindicated for
classes pyelonephritis due to low plasma
High urine concentration concentrations
Good safety profile
Improved compliance
Low risk of Clostridium difficile infection
Pregnancy Category B
Pivmecillinam 400 mg twice daily for 5 Active against Escherichia coli including Poor activity against Rash and gastrointestinal upset, including
days ESBLs- producing strains, K pneumonia P. vulgaris and M. morganii nausea and vomiting
including ESBLs and carbapenemase- Vulvovaginal candidiasis
producing strains, K. oxytoca, Proteus Carnitine deficiency with long term or
mirabilis, Enterobacter spp, Citrobacter frequent use
spp Avoid concomitant use with valproate
products
Trimethoprim- 160 mg/800 mg twice Active against Staphylococcus Lacks activity against Enterococci spp, Nausea Not recommended for empiric
sulfamethoxazole daily for 3 days saprophyticus, Escherichia coli, Pseudomonas aeruginosa, and CRE Vomiting treatment of pyelonephritis
(females) and for 7 days Klebsiella pneumonia, Proteus mirabilis Increasing resistance to common Hypersensitivity reactions such as fever, rash, Not recommended as empiric use
(males) Short course therapy uropathogens such as Escherichia coli urticarial, and Stevens–Johnson syndrome for cystitis if E coli resistance
Indicated for both and AmpC- β -lactamase and ESBLs- Bone marrow suppression rate in the community is >20%
uncomplicated cystitis and producing Gram-negative organisms Acute kidney injury Avoid co-administration with
uncomplicated pyelonephritis Not recommended in pregnancy (avoid Hyperkalemia, hyponatremia spironolactone, angiotensin
High urine concentration in 1st trimester and last 6 weeks) Hemolysis in patients with glucose- converting – enzyme inhibitors
Low risk of Clostridium difficile infection Potential drug- drug interactions: 6-phosphate dehydrogenase deficiency or angiotensin-receptor blockers
warfarin, phenytoin and sulfonylureas due to increased potential risk
Requires dose adjustment in renal of hyperkalemia and sudden
impairment death

(Continued )
Table 1. (Continued).

Anti-infective Dosing Advantages Disadvantages Adverse effects Comments


Trimethoprim 100-200 mg twice daily for Active against Staphylococcus No activity against Enterococci, Nausea and vomiting Not recommended for empiric
3–5 days saprophyticus, Escherichia coli, Pseudomonas aeruginosa and CRE Rash treatment of cystitis if E coli
Klebsiella pneumonia, Proteus mirabilis Increasing resistance to common Hyperkalemia, hyponatremia resistance rate in the
High urine concentration uropathogens such as Escherichia coli, Bone marrow suppression community is >20%
Low risk of Clostridium difficile infection AmpC- β -lactamase and ESBLs- Megaloblastic anemia
producing Gram-negative organisms Methemoglobinemia
Not indicated for pyelonephritis Acute kidney injury
Drug-drug interaction: dapsone,
methotrexate phenytoin
Not recommended in first trimester of
pregnancy
Requires dose adjustment in renal
impairment
Cephalosporins Cephalexin Active against E. coli, Klebsiella spp, and Lacks activity against Enterococcus Headaches
250–500 mg orally every Proteus mirabilis species, AmpC- β -lactamase and Diarrhea
6–8 h for 7 days Indicated for acute prostatitis ESBLs-producing Gram-negative Nausea
Cefixime Pregnancy Category B organisms, Morganella spp, Vomiting
400 mg daily for 7 days Acinetobacter spp., Stenotrophomonas Allergic reactions: rash, urticarial, or
maltophilia and Pseudomonas anaphylactic reaction (<2% risk cross
Dose adjustment required in renal reactivity in patients with IgE mediated
dysfunction penicillin allergy)
Risk of Clostridium difficile infection Vaginitis
May result in elevated INR
Fluoroquinolones Ciprofloxacin: 250–500 mg Active against Staphylococcus Lacks activity against Enterococci and Nausea For patients who cannot tolerate
twice daily for 3 days saprophyticus, Enterobacterales spp CRE. Vomiting oral therapy give first dose IV
(females) OR for 7 days including E. coli, Klebsiella spp, and Increasing resistance to common Diarrhea If local resistance of E coli against
(males) OR Levofloxacin: Proteus mirabilis uropathogens including Escherichia Headache fluoroquinolones exceeds 10%,
250-500 mg daily for 3 , Pseudomonas aeruginosa coli, AmpC- β -lactamase, ESBLs- Drowsiness use other treatment options for
days (female) OR for 7 Definitive oral therapy for susceptible producing Gram-negative organisms, Insomnia pyelonephritis
days (male) OR AmpC- β -lactamase and ESBLs- Pseudomonas aeruginosa, and Dizziness Not recommended for empiric
Norfloxacin 400 mg producing Gram-negative organisms Acinetobacter spp. Photosensitivity/phototoxicity treatment of hospital-acquired
twice daily for 3 days Indicated for both uncomplicated cystitis High risk of Clostridium difficile infection Vaginitis UTIs
(female) OR for 7 days and uncomplicated pyelonephritis Pregnancy category C (Contraindicated in Nausea, vomiting, and diarrhea Norfloxacin has no known cardiac
(male) OR Ofloxacin Short course therapy – lower cost, and pregnancy and breastfeeding) QTc prolongation effects or interactions with
200-400 mg twice daily improved compliance Drug Drug interactions: cations (antacids Tendinitis and tendon rupture spironolactone and is not
Short course definitive therapy for and vitamins), warfarin, theophylline Central nervous system adverse events such associated with sudden death
uncomplicated pyelonephritis (monitor) as encephalopathy, delirium, and seizures
High urine concentration Require dose adjustment in renal
impairment
Penicillins Amoxicillin Active against, Enterococcus faecalis, Lacks activity against Staphylococcus Diarrhea Not recommended for empiric
500 mg every 8 h for 5–7 Escherichia coli, Proteus mirabilis saprophyticus, Klebsiella pneumonia, Nausea treatment of cystitis
days Pregnancy Category B Pseudomonas aeruginosa, Morganella Vomiting
morganii, Providencia stuartii, AmpC- β Allergic reactions: rash, urticarial, or
-lactamase and ESBLs-producing anaphylactic reaction
Gram-negative organisms and CRE. Vaginitis
Generally inferior to fluoroquinolones Headaches
Low urine concentration
High risk of Clostridium difficile infection
POSTGRADUATE MEDICINE

Requires dose adjustment in renal


impairment

(Continued )
237
238 M. S. BADER ET AL.

Pevmecillinam is oral synthetic penicillin marketed in sev-

VRE: vancomycin resistant Enterococci; N/A: not applicable; ESBLs: extended-spectrum β-lactamases; CRE: carbapenem-resistant Enterobacteriaceae; CrCl: creatinine clearance; UTIs: urinary tract infections; IV; intravenous; MBLs:
concentration used by EUCAST
eral European countries (Table 1) [9,31]. However, there is

susceptibility testing results,

pyelonephritis and hospital-


Not recommended for empiric
and CLSI results in variable

particularly among ESBLs-


a poor correlation between pivmecillinam MIC and clinical

Variation in the clavulanate

producing E. coli isolate


outcome and treatment failure in mecillinam – susceptible

Comments

treatment of acute
ESBLs-E. coli has been observed in some cases [32,33].
The prevalence of outpatient UTIs due to ESBLs-

acquired UTIs
Enterobacteriales is increasing [7–9]. Risk factors for UTIs due
to ESBLs-Enterobacterales include age, comorbid condition,
recent antibiotic exposure, recent hospitalization, and long-
term facility residency [34]. Oral treatment options for ESBLs-
E coli include nitrofurantoin, fosfomycin, pivmecillinam, amox-
icillin-clavulanate, and sitafloxacin. Treatment oral options for
ESBL- Klebsiella pneumoniae include pivmecillinam and fosfo-
Allergic reactions: rash, urticarial, or

mycin [5,6]. Unfortunately, the level of resistance of ESBLs


Adverse effects

Enterobacteriales to trimethoprim-sulfamethoxazole and fluor-


oquinolones is very high and they have very limited role in the
anaphylactic reaction

treatment of UTIs due to these organisms [7,35,36]. The role of


amoxicillin/clavulanic acid in the treatment of UTIs due to
ESBL-Enterobacteriales is very limited due to increasing rates
of resistance, limited evidence for its clinical efficacy, and
Headaches
Vomiting

Vaginitis
Diarrhea

Metallo-β-lactamases; EUCAST: The European Committee on Antimicrobial Susceptibility Testing; CLSI: clinical and laboratory standards institute.

increased rates of drug-related adverse events and antibiotic-


Nausea

associated diarrhea [6,7,29]. However, a combination aztreo-


nam, active on Metallo-β-Lactamase (MBLs) alone, with amox-
lactamase and ESBLs-producing Gram-

High risk of Clostridium difficile infection

icillin/clavulanic acid, inhibiting ESBLs if present, is an effective


aeruginosa, Morganella morganii,

therapeutic option to treat infections caused by MBLs-


Longer treatment courses required

including susceptible organisms

Requires dose adjustment in renal


Lacks activity versus Pseudomonas

Providencia stuartii, AmpC- β -

High rates of relapse and failure

producing Gram-negative bacteria [37].


negative organisms and CRE
Disadvantages

Low urine concentration

Complicated UTIs and acute pyelonephritis


Complicated UTIs are associated with an underlying condition
impairment

that increases the risk of recurrent infections or treatment


failure due to functional or anatomic abnormalities of the
urinary tract [5,6]. All patients with complicated UTIs and
acute pyelonephritis require urine culture and antimicrobial
susceptibility testing due to the wide diversity of potential
saprophyticus, Enterococci, Escherichia

pathogens, risk of antimicrobial resistance, and risk of compli-


coli, Klebsiella pneumonia, Proteus

Definitive treatment for susceptible


ESBLs-producing Gram-negative

cations [2,5]. Recurrent infection, instrumentation and repeat


Active against Staphylococcus

courses of antimicrobial therapy increase the risk of antibiotic-


Advantages

resistant organisms causing cUTIs [2,5,6].


Pregnancy Category B

Management of cUTIs and acute pyelonephritis caused by


Enterobacteriales continues to pose a challenge for clinicians
due to the lack of viable traditional oral antibiotic options,
organisms
mirabilis

such as amoxicillin-clavulanate, cephalosporins, trimethoprim-


sulfamethoxazole, and fluoroquinolones [5,6]. As a result,
managing cUTIs and acute pyelonephritis often requires intra-
venous empiric broad-spectrum antibiotics [6]. However, once
twice daily for 5–7 days
125 mg twice daily for

CrCl<30 ml/min 500 mg

there is a response to initial therapy and the result antimicro-


CrCl > 30 ml/min 875/
Amoxicillin-clavulanate

bial susceptibility testing is available, a step down to oral


Dosing

antibiotic that the isolated organisms are susceptible is recom-


mended to complete the total duration of therapy even in
5–7 days

patients with bacteremia [38].

Cephalosporines
Table 1. (Continued).

Ceftriaxone is the first recommended empiric treatment of acute


Anti-infective

pyelonephritis in the absence of risk factors for resistance organ-


isms such as ESBLs-Enterobacteriales, Pseudomonas aeruginosa,
or Enterococci (Table 2) [2,5,39]. Risk factors of UTIs due to
extended-spectrum cephalosporin resistance (ESC-R) include
Table 2. Treatment options for acute pyelonephritis and complicated urinary tract infections.

Anti-infective Dosing Advantages Disadvantages Adverse effects Comments


Cephalosporins Ceftriaxone 1–2 g IV every 24 h Active against Escherichia coli, Klebsiella Lacks activity against Enterococci, Diarrhea First line empiric option for acute
pneumonia, Proteus mirabilis, Enterobacter Pseudomonas aeruginosa, AmpC- β - Thrombocytopenia, pyelonephritis in patients without risk
spp, Serratia, Citrobacter spp, and Morganella lactamase and ESBL-producing Gram- Thrombocytosis, factors for antibiotic-resistant
spp., and Providencia stuartii negative organisms and CRE Leukopenia organisms such as ESBLs producing
No dose adjustment required in renal Reduced activity versus Staphylococcus Biliary pseudolithiasis organisms or Pseudomonas aeruginosa
dysfunction saprophyticus Allergic reactions: rash,
Administered once daily Low urine concentration urticarial, or anaphylactic
Pregnancy category B Increased risk of Clostridium difficile reaction(<2% risk cross
infection reactivity in patients with
IgE mediated penicillin
allergy)
Ceftazidime 1–2 g IV every 8 h Active against Escherichia coli, Klebsiella Lacks activity against Enterococci, Seizures Indicated for the treatment of suspected
pneumonia, Proteus mirabilis, Proteus vulgaris, Staphylococcus saprophyticus Encephalopathy or proven cUTIs or acute pyelonephritis
Enterobacter, Serratia, Citrobacter, and and CRE Local infusion related due to Pseudomonas aeruginosa
Morganella spp., Providencia stuartii, High resistance rates in AmpC- β - effects
Pseudomonas aeruginosa including some lactamase and ESBLs-producing Gram- Nausea
MDR Pseudomonas strains negative organisms and Acinetobacter Vomiting
High urinary concentration baumannii Diarrhea
Pregnancy category B Increased resistance of Pseudomonas Allergic reactions: rash,
aeruginosa urticarial, or anaphylactic
Increased risk of Clostridium difficile reaction(<2% risk cross
infection reactivity in patients with
Requires dose adjustment in renal IgE mediated penicillin
impairment allergy)
Avoid in patients with
history of cephalosporin-
associated hemolytic
anemia
Cefepime 1–2 g IV every 8 h or 2 Active against many common urinary Lacks activity against Enterococci and CRE Encephalopathy associated Indicated for the treatment of suspected
g IV every 12 h pathogens including Staphylococcus High resistance rates in ESBLs-producing with higher doses in or proven cUTIs or acute pyelonephritis
saprophyticus, Escherichia coli, Klebsiella Gram-negative organisms and setting of renal due to AmpC- β -lactamase –
pneumonia, AmpC- β -lactamase – producing Acinetobacter baumannii dysfunction producing Gram-negative organisms
Gram-negative organisms and Pseudomonas Increased resistance of Pseudomonas Rash and Pseudomonas aeruginosa
aeruginosa aeruginosa Diarrhea
High urinary concentration Risk of cross reactivity with penicillins Nausea
Pregnancy category B High risk of Clostridium difficile infection Vomiting
Requires dose adjustment in renal Allergic reactions: rash,
impairment urticarial, or anaphylactic
reaction(<2% risk cross
reactivity in patients with
IgE mediated penicillin
allergy)
Avoid in patients with
history of cephalosporin-
associated hemolytic
anemia
POSTGRADUATE MEDICINE

(Continued )
239
240

Table 2. (Continued).

Anti-infective Dosing Advantages Disadvantages Adverse effects Comments


Ceftazidime-avibactam 2.5 g IV Activity comparable to ceftazidime with Lacks activity against Enterococci, Gastrointestinal such as Indicated for the treatment of suspected
every 8 h additional activity against AmpC- β - Staphylococcus saprophyticus, class nausea, vomiting, or proven cUTIs or acute pyelonephritis
M. S. BADER ET AL.

lactamase and ESBLs-producing Gram- B metallo- β -lactamases (eg, NDM, constipation, and due to CRE or MDR-Pseudomonas
negative organisms, CRE, and MDR- VIM, IMP, VEB, PER) and Acinetobacter elevated liver enzymes. aeruginosa
Pseudomonas aeruginosa baumannii, ceftazidime-resistant Cough
High urine concentration Stenotrophomonas Dizziness
Pregnancy category B Emergence of resistance during therapy Headache
particularly among KPC- K pneumonia Insomnia
Reduced efficacy in patients with Seizures
moderate renal impairment Myoclonus
Risk of cross reactivity with penicillins Encephalopathy
Requires dose adjustment in renal Avoid in patients with
impairment history of cephalosporin-
Risk of Clostridium difficile infection associated hemolytic
Limited data for the use in the elderly anemia
population
Ceftolozane-tazobactam 1.5 g IV Active against common urinary pathogens Lacks activity against Enterococci CRE, Nausea Indicated for the treatment of suspected
every 8 h including Escherichia coli, Klebsiella S. maltophilia, ESBLs-, KPC-, or MBLs- Diarrhea or proven cUTIs or acute pyelonephritis
pneumonia, AmpC- β -lactamase and ESBLs producing P. aeruginosa Headaches due to MDR- Pseudomonas aeruginosa
producing-Enterobacteriales, and MDR- Reduced activity against Staphylococci Pyrexia
Pseudomonas aeruginosa saprophyticus Avoid in patients with
High urine concentration Reduced activity against ESBLs- history of cephalosporin-
Pregnancy category B K pneumonia associated hemolytic
Variable activity against AmpC- producing anemia
Enterobacteriales
Reduced efficacy in patients with
moderate renal impairment
Risk of cross reactivity with penicillins
Requires dose adjustment in renal
impairment
Risk of Clostridium difficile infection
Piperacillin – 3.375–4.5 g IV every 6–8 h Active against common urinary pathogens such Lacks activity against most ESBLs- Rash Indicated for the treatment of suspected
tazobactam as Staphylococcus saprophyticus, Enterococci, producing Gram-negative organisms, Nausea or proven cUTIs or acute pyelonephritis
Escherichia coli, Klebsiella pneumonia, AmpC- CRE, and S. maltophilia. Diarrhea due to AmpC- β -lactamase –
β -lactamase- and ESBLs- producing Gram- Increasing resistance rates for Neutropenia producing Gram-negative organisms
negative organisms, and Pseudomonas Pseudomonas aeruginosa Hypokalemia and Pseudomonas aeruginosa
aeruginosa Requires dose adjustment in renal Prolonged prothrombin
High urine concentration impairment time (specifically in renal
Pregnancy category B Moderate risk of Clostridium difficile failure)
infection

(Continued )
Table 2. (Continued).

Anti-infective Dosing Advantages Disadvantages Adverse effects Comments


Carbapenems Ertapenem 1 g IV daily Active against common urinary pathogens such Lacks activity against E. faecium, Seizures Carbapenems are considered first line
Meropenem 500 mg every 6 h up as Staphylococcus saprophyticus, Enterococci, S. maltophilia, and CRE. Diarrhea treatment options for the treatment of
to 1 g IV every 8 h Escherichia coli, Klebsiella pneumonia, AmpC- Ertapenem has no activity against Nausea suspected or proven cUTIs or acute
Doripenem 500 mg IV every 8 h β -lactamase, ESBLs-producing Gram- Enterococci, Pseudomonas aeruginosa, Rash pyelonephritis due to ESBLs- producing
Imipenem/cilastatin 500 mg IV negative organisms, Pseudomonas and Acinetobacter baumannii Headache Gram-negative organisms
every 6 h aeruginosa, and Acinetobacter baumannii Increasing resistance rates of ESBLs- Hypokalemia Meropenem/vaborbactam is indicated for
High urinary concentration producing Gram-negative organisms Elevted liver enzymes the treatment of suspected or proven
Pregnancy category B (particularly K. pneumonia and Proteus cUTIs or acute pyelonephritis due to
(Meropenem, ertapenem, doripenem) mirabilis), Pseudomonas aeruginosa KPC-CRE
Low risk of cross reactivity for IgE- mediated , and Acinetobacter baumannii Meropenem is favored over imipenem in
hypersensitivity to penicillins Require dose adjustment in renal patients with a history of seizures,
impairment (all carbapenems) pregnancy or impaired renal function
Pregnancy category C for imipenem
Moderate risk of Clostridium difficile
infection
Meropenem-vaborbactam 4 Activity comparable to meropenem with Lacks activity against meropenem-
g every 8 h additional activity against KPC-producing resistant Acinetobacter baumannii,
CRE. Pseudomonas aeruginosa, or
Higher barrier for developing resistance in Stenotrophomonas maltophilia
comparison to ceftazidime–avibactam for Lacks activity against Ambler class B and
CRE D carbapenemases (MBLs including
NDM and OXA-like beta-lactamases,
respectively)
Aminoglycosides Gentamicin 5 mg/kg IV once daily Active against common urinary pathogens Lacks activity against Staphylococcus Nephrotoxicity Dosing based on ideal body weight
Tobramycin 5 mg/kg IV once daily including Escherichia coli, Klebsiella saprophyticus, Enterococci Vestibulocochlear toxicity Amikacin is preferred over gentamicin
Amikacin 15 mg/kg IV once daily pneumonia, AmpC- β -lactamase, ESBLs- Increasing resistance rates of ESBLs- Neurotoxicity when used for empiric treatment due
producing Enterobacteriales, MDR- producing Gram-negative organisms, Edema to antibiotic-resistant Gram-negative
Pseudomonas aeruginosa, CRE, and Pseudomonas aeruginosa, and organisms such ESBLs-producing
Acinetobacter baumannii Acinetobacter baumannii Enterobacteriales or MDR-
High urine concentration All aminoglycosides lack activity against Pseudomonas aeruginosa
Once daily dosing NDM-1-producing Enterobacteriales Combination therapy is recommended
Low risk of Clostridium difficile infection Require dose adjustment in renal (e.g. with ampicillin) when used as
impairment empiric treatment
Require Therapeutic Drug Monitoring Monotherapy is acceptable if used as
Pregnancy category D definitive therapy
Plazomicin has no activity against Plazomicin is indicated for the treatment
Enterococci, NDM-producing CRE, of cUTIs or acute pyelonephritis due to
High MIC Proteus mirabilis and indole- gram-negative organisms in patients
positive Proteeae, and morganella who have limited or no alternative
treatment options.
Plazomicin 15 mg/kg once daily Plazomicin has activity similar to other Dizziness, Somnolence,
aminoglycosides but displays additional Nausea
activity against aminoglycoside-resistant Transient hypotension or
Enterobacteriales containing aminoglycoside- hypertension
modifying enzymes, colistin-resistant Neuromuscular blockade
Enterobacteriales (including those expressing
the mcr-1 gene), gram positive bacteria such
POSTGRADUATE MEDICINE

Staphylococcus aureus including MRSA and


Staphylococcus saprophyticus.

(Continued )
241
242

Table 2. (Continued).

Anti-infective Dosing Advantages Disadvantages Adverse effects Comments


Polymixins Polymixin B 15,000–25,000 Units/ Active against common urinary pathogens such Lacks activity against Staphylococcus Nephrotoxicity Dosing based on ideal body weight
M. S. BADER ET AL.

kg/day IV every q12 as Escherichia coli, Klebsiella pneumonia, saprophyticus, Enterococci, Proteus spp., Neurotoxicity Colistin (polymixin E) is preferred over
h (Maximum daily dose AmpC- β -lactamase, ESBLs-producing Gram- Providencia spp., Serratia marcescens, Neuromuscular blockade polymixin B due to higher urinary
2 million units) negative organisms, MDR-Pseudomonas Morganella morganii Respiratory arrest concentrations
Polymixin E (colistin) 2.5–5 mg aeruginosa, CRE, and Acinetobacter baumannii Colistin resistance is emerging in Colistin is indicated for the treatment of
CBA/kg/day in 2–4 divided Bladder irrigation considered in those who Acinetobacter baumannii and K. suspected or proven cUTIs or acute
doses cannot tolerate intravenous pneumonia, MDR- Pseudomonas pyelonephritis due to MDR- gram
Low resistance rates aeruginosa, and E.coli negative organisms in patients who
High urine concentration for polymixin E Decreased urine concentration for have limited or no alternative
Low risk of Clostridium difficile infection Polymixin B treatment options.
Narrow therapeutic window & requires
dose adjustment in renal impairment
Safety in pregnancy has not been
established for polymixin B
Polymixin E is Pregnancy Category C
Monobactams Aztreonam Active against common urinary pathogens such Lacks activity against Staphylococcus Neutropenia Indicated for the treatment of suspected
0.5–2 g IV every 6–8 h as Escherichia coli, Klebsiella pneumonia, MBLs saprophyticus, Enterococci, AmpC β - Increased serum or proven cUTIs or acute pyelonephritis
and OXA-48-like carbapenemases- producing lactamase, ESBLs and KPC-producing transaminases due to gram negative bacteria in
Enterobacteriales and Pseudomonas Enterobacteriales Rash patients with beta-lactam allergy as
aeruginosa. High level of resistance among Diarrhea cross reactivity rare
P aeruginosa Nausea
Vomiting
Fluoroquinolones **Refer to Table 1
Fosfomycin 4–6 g iv every 6–8 h Active against common urinary pathogens Lacks activity against Morganella spp, Headache
including Enterococci including VRE, Acinetobacter spp., S. maltophilia, NDM- Dizziness
Escherichia coli, Klebsiella pneumonia, Proteus producing CRE, Rash
mirabilis, Pseudomonas aeruginosa, and Staphylococcus saprophyticus. Diarrhea
AmpC- β -lactamase and ESBLs-producing Increasing resistance of ESBLs- Klebsiella Nausea
Gram-negative organisms and KPC- and pneumonia and ESBLs 025b/B2 E. coli Dyspepsia
MBLs-producing CRE strains Vaginitis
Low rates of resistance and no cross resistance High clinical failure with Pseudomonas Infusion related reaction,
with other antimicrobial classes aeruginosa Hypokalemia,
High urine concentration Sodium overload that might
Low risk of Clostridium difficile infection lead heart failure
Pregnancy Category B Hypertension
Elevated serum
aminotransferases
MDR: multi-drug resistant; ESBLs: extended-spectrum β-lactamases; CRE: carbapenem-resistant Enterobacteriaceae; MBLs: Metallo-β-lactamases; UTIs: urinary tract infections; IV; intravenous; CBA: colistin base activity; VRE:
vancomycin-resistant enterococci.
POSTGRADUATE MEDICINE 243

older age, nursing home residence, previous antimicrobial ther- and acute pyelonephritis [58]. Although randomized trial data
apy, trimethoprim-sulfamethoxazole exposure in the prior 6 have demonstrated high clinical cure rates with ceftolozane/
months, hospitalization, malignancy, diabetes mellitus, and tazobactam for the treatment of cUTIs caused by ESBLs-
recurrent UTIs [17,40]. Enterobacteriales, there was a high rate of resistance particu-
Although the resistance rate of E coli to oral cephalosporins is larly among ESBLs-Klebsiella pneumoniae [35,57,59,60].
<10% in several regions, it should not be used as empiric treat- Therefore, ceftolozane/tazobactam should be used an alterna-
ment of uncomplicated acute pyelonephritis due to lack of clinical tive, and not a primary choice, to carbapenems for the treat-
evidence [7,8]. However, oral cephalosporins can be used as a step ment of infections caused by ESBLs-Enterobacteriales.
down treatment from parenteral therapy if the isolated organisms Cefiderocol is a novel cephalosporin, siderophore antibio-
are susceptible or an empiric treatment of mild acute pyelone- tic, that forms a chelating complex with iron and is then
phritis when the resistance rate is very low [41–43]. actively transported into bacterial cells via iron transporters.
Avibactam, a non-β-lactam β-lactamases inhibitor (BLI), Cefiderocol is released in the periplasmic space and binds to
restores the activity of ceftazidime against Ambler class penicillin binding proteins (PBPs), primarily to PBP3, and inhi-
A (e.g. ESBL and KPC), class C (e.g. AmpC), and some class D β- bits peptidoglycan synthesis, causing cell death [61]. It has
lactamase–producing bacteria (OXA-48-like). It is approved for significant antimicrobial activity against Enterobacterales,
the treatment of cUTIs and acute pyelonephritis and for gram- Ambler class A, B, C and D β-lactamases, P. aeruginosa,
negative infections such as bacteremia with limited treatment A. baumannii, S. maltophilia and Burkholderia cepacia [62,63].
options (Table 2) [44]. In vitro studies of ceftazidime/avibac- A randomized, double-blind, phase II trial comparing cefi-
tam have demonstrated inhibition against >99.9% of derocol with imipenem/cilastatin for cUTIs due to gram-
Enterobacterales and 99.4% of P. aeruginosa isolates that negative organism, 25-29% had acute pyelonephritis, in hos-
were resistant to meropenem, ceftazidime and piperacillin/ pitalized patients demonstrated superiority of cefiderocol for
tazobactam and 100% of KPC and OXA-48 producers [45–48]. the composite clinical and microbiological outcome, primarily
Although clinical improvement can be seen in some patients due to higher microbiological eradication. The composite
infected with MBLs-producing pathogens, persistence of cau- microbiological and clinical responses at test of cure for cefi-
sative pathogen may occur and therefore ceftazidime- derocol and imipenem/cilastatin were 72.6% and 54.6%,
avibactam monotherapy should be avoided in treating infec- respectively, a difference of 18.58 (95% CI 8.23–28.92) [64]. It
tions due to MBLs-producing isolates [49]. However, is still not approved for treatment of cUTIs.
a combination aztreonam with ceftazidime-avibactam is an
effective therapeutic option to treat infections caused by
MBLs-producing Gram-negative bacteria [37].
Fluoroquinolones
Several Phase 2 and 3 trials showed that ceftazidime-
avibactam was non- inferior to carbapenems for the co- Fluoroquinolones are no longer recommended as an empiric
primary endpoints of clinical (symptom resolution) and com- treatment of acute pyelonephritis since the rate of fluoroqui-
bined clinical/microbiological but superior for the microbiolo- nolones-resistant E coli is in excess of 10% in several regions
gical endpoint for the treatment of cUTIs and acute and countries [7–9,23,65]. Furthermore, they cannot be used
pyelonephritis [50–52]. Serious adverse events, gastrointestinal as empiric treatment of cUTIs or acute pyelonephritis in
adverse events, and creatinine increase were significantly patients with risk factors for ESBLs-Enterobacteriales because
more common with ceftazidime/avibactam than carbapenems of high rates of resistance [7,35].
[53]. Furthermore, for unclear reasons the clinical cure rates of However, fluoroquinolones can be used as a step-down
ceftazidime-avibactam were lower than carbapenems for treatment from parenteral therapy if the isolated organisms
AmpC- β -lactamase-Enterobacteriales and pseudomonas aer- are susceptible [38,66].
uginosa. However, the number of cases is too small to allow Finafloxacin is an investigational intravenous and oral fluor-
for firm conclusions [49,54]. Finally, there is a concern of oquinolone that is highly excreted in the urine and has higher
resistance development to ceftazidime-avibactam during ther- potency at acidic pH in contrast to ciprofloxacin and levoflox-
apy particularly among K. pneumoniae bacteria harboring var- acin. It is effective against fluoroquinolones-resistant E coli and
iant KPC-3 enzymes [55]. ESBLs- Enterobacteriales (Table 1) [67,68].
Wagenlehner et al. reported at least a fourfold increase in MIC In a phase II trial, a total of 225 patients with cUTIs and or
of ceftazidime-avibactam at test of cure in 2% of isolates [50]. acute pyelonephritis were randomized 1.1:1 to treatment of
Ceftolozane/tazobactam is a combination of ceftolozane, finafloxacin 800 mg intravenous/oral daily for 5 days, finaflox-
a novel oxyimino – aminothiazolyl cephalosporin that resem- acin 800 mg intravenous/oral daily for 10 days, and ciproflox-
bles ceftazidime, and the β-lactamase inhibitor tazobactam. It acin (intravenous 400 mg and oral 500 mg twice daily) for 10
is approved for cUTIs and acute pyelonephritis (Table 2) [56]. It days. Both short and long course of finafloxacin resulted in
demonstrates remarkable stability against the numerous resis- comparable clinical and microbiological response rates as well
tance mechanisms employed by P. aeruginosa, including over- as composite response rates at the test of cure visit (TOC)
expression of AmpC, efflux pumps and loss of porin channels on day 17 and both showed higher treatment success rates
but not against ESBLs, KPC, or MBLs [57]. than ciprofloxacin. The treatment success rates (combined
In a randomized, double-blind, phase III trial, ceftolozane/ clinical and microbiological response) at the TOC visit on day
tazobactam was superior to high dose levofloxacin for com- 17 were 70.3% (95% CI, 57.6% to 81.1%) for the 5-days fina-
posite clinical and microbiological cure in patients with cUTIs floxacin group, 67.6% (95% CI, 55.2% to 78.5%) for the 10-days
244 M. S. BADER ET AL.

finafloxacin group, and 57.4% (95% CI, 44.1% to 70.0%) for the to ESBLs-E. coli after the susceptibility results showed those
ciprofloxacin group [69]. isolates to have lower MICs and there is a clinical response to
the empiric Piperacillin-tazobactam therapy. Optimizing the
dosing of piperacillin-tazobactam to achieve the drug target
Carbapenems
attainment is necessary by a continuous infusion (4.5 g infused
Carbapenems are considered the antimicrobial agents of choice over 4 h every 6–8 h) [83,84].
for treatment of cUTIs and acute pyelonephritis, particularly The role of cefepime in the treatment of UTIs due to ESBLs-
severe infections, due to ESBLs-Enterobacteriales (Table 2). Enterobacteriales is fading because of the very limited evi-
Ertapenem is the preferred carbapenem to treat cUTIs and dence of its efficacy (Table 2) [85]. Several studies showed
acute pyelonephritis due to ESBLs- Enterobacteriales due to its high clinical failure rates with the use of cefepime to treat
efficacy, convenience for outpatient therapy, and narrower infections caused by ESBL-Enterobacterales [81,86–88].
spectrum comparing to other carbapenems (Table 2) [70,71]. Vaborbactam (formerly named RPX7009) is a cyclic boronic
Ertapenem remains highly active against Enterobacteriales acid β-lactamase inhibitor that has activity against Ambler
including ESBLs strains from UTIs. Resistance to ertapenem is class A (including KPC, CTX-M, SHV, TEM) and C enzymes
increasingly reported in parts of the world other than US/ (P99, MIR, FOX). Vaborbactam does not inhibit class D or
Canada particularly among Enterobacter cloacae, k pneumonia, class B carbapenemases. Meropenem–vaborbactam is four
ESBL- Enterobacteriales, and hospital-acquired infections times more potent than ceftazidime–avibactam against KPC-
[72,73]. producing isolates [89]. The FDA has approved a fixed-dose
The role of (β-lactam/β- lactamase inhibitor) BLBLIs (piper- combination of meropenem and vaborbactam for treatment
acillin-tazobactam) in the treatment of UTIs caused by ESBL- of adults with cUTIs and acute pyelonephritis (Table 2) [90].
Enterobacteriales is still controversial. Piperacillin/ tazobactam TANGO I is a multicenter, randomized, double-blinded, con-
still has retained good in vitro activity against ESBLs- trolled trial comparing meropenem–vaborbactam (2g/2g over 3
Enterobacteriales, particularly ESBLs- E coli, but its clinical effi- h every 8 h; n = 274) to piperacillin–tazobactam (4 g/0.5 g over 30
cacy remains controversial (Table 2) [74–80]. min every 8 h; n = 276) for the treatment of adults with cUTIs or
A small randomized study of hospitalized adult patients acute pyelonephritis (59%). For the primary end point (compo-
with healthcare-associated UTI due to ESBLs-E coli were ran- site clinical cure and microbiological eradication at end of intra-
domized to 10–14 days treatment with piperacillin- venous treatment), overall success occurred in 189 of 192 (98.4%)
tazobactam (33 patients), cefepime (6 patients), and ertape- with meropenem-vaborbactam vs 171 of 182 (94.0%) with piper-
nem (33 patients). The clinical and microbiological response to acillin-tazobactam (difference, 4.5%[95%CI, 0.7%to 9.1%]; P <
piperacillin-tazobactam and ertapenem treatment was 94% .001 for noninferiority, but superiority since the lower limit of
and 97%, respectively, but significantly lower for cefepime the 95% CI (0.7%) exceeded 0 (P = .01).) [91].
(33.3%) [81]. TANGO II was a phase III, open-label, multicenter, rando-
A retrospective study revealed that treatment failure, mized controlled trial designed to assess the effectiveness of
defined as a composite of in-hospital mortality, change of meropenem–vaborbactam versus best available therapy for
initial antibiotic regimen and microbiological eradication fail- documented or suspected CRE infections. Among 47 patients
ure, of patients with acute pyelonephritis caused by ESBLs- with CRE infections (34% had cUTIs and acute pyelonephritis),
E coli susceptible to piperacillin-tazobactam was not signifi- monotherapy with meropenem-vaborbactam was associated
cantly different for piperacillin-tazobactam and ertapenem with increased clinical cure at test of cure (59.4% vs. 26.7%,
(20.6 vs. 23.2%, respectively, 95% CI of difference 4.6–60.8, p = 0.02), decreased 28 days
P = 0.704) [76]. mortality (15.6% vs. 33.3%, 95% CI of difference −44.7 to 9.3,
In a noninferiority, parallel group, randomized clinical trial p = 0.2), and reduced nephrotoxicity compared with best-
of hospitalized adult patients with bloodstream infection available therapy (mono/combination therapy with polymix-
caused by ceftriaxone-nonsusceptible E coli (266) or ins, carbapenems, aminoglycosides, tigecycline; or ceftazi-
K pneumoniae (40) were randomly assigned 1:1 to intravenous dime-avibactam alone) [92].
piperacillin-tazobactam, 4.5 g every 6 h, (n = 188 participants) Among 25 patients harboring KPC-Enterobacteriales treated
or meropenem, 1 g every 8 h, (n = 191 participants) for with meropenem–vaborbactam, only one had a subsequent
a minimum of 4 days, up to a maximum of 14 days. isolate recovered with a ≥ fourfold increase in MIC (0.25–1 μg/
A urinary tract source for blood stream infection was 67% of mL) while two out four patients treated with ceftazidime –
meropenem and 55% in piperacillin-tazobactam group. avibactam had subsequent K. pneumoniae isolates recovered
Phenotypic ESBLs production was confirmed in 86.0% of iso- with a ≥ fourfold MIC increases to ceftazidime–avibactam and
lates (85.0%of E coli and 92.5% of K pneumoniae). All-cause one evolved into full resistance during therapy [93,94].
mortality at 30 days, primary outcome, occurred in 12.3% (23 Relebactam (formerly named MK-7655) is a diazabicyclooctanes
of 187) and 3.7% (7 of 191) patients who randomized to non β- lactam β-lactamase inhibitor that has activity similar to
piperacillin-tazobactam and meropenem, respectively (risk dif- vaborbactam in addition to improving the activity of imipenem
ference, 8.6% [one-sided 97.5% CI, -∞ to 14.5%]; P = .90 for against Pseudomonas aeruginosa. It is not active against imipenem-
noninferiority), demonstrating that piperacillin-tazobactam did resistant Enterobacteriales expressing IMPs, VIMs, NDM, or MBLs,
not meet criteria for noninferiority [82]. A. baumannii, or IMP- or VIM-producing P. aeruginosa [95].
It is reasonable to continue piperacillin-tazobactam for A randomized, double-blind, Phase 2 study comparing effi-
definitive monotherapy for cUTIs or acute pyelonephritis due cacy and safety of imipenem/cilastatin plus Relebactam (either
POSTGRADUATE MEDICINE 245

125 mg or 250 mg intravenous every 6 h) with imipenem/cilas- recently approved in Canada, despite being available for sev-
tatin alone in patients with cUTIs and acute pyelonephritis eral years in Europe. Fosfomycin has a broad spectrum of
showed non-inferiority of both imipenem/cilastatin + relebactam activity against a variety of clinically important MDR Gram-
doses to imipenem/cilastatin alone for both clinical (97.1%, negative pathogens such as ESBLs- and CRE-Enterobacteriales
98.7% and 98.8%, respectively) and microbiological response and Gram-positive pathogens such as vancomycin-resistant
(95.5%, 98.6% and 98.7%, respectively) [96]. enterococci (Table 2) [105].
Sulopenem is a new broad-spectrum carbapenem in devel- In a multicenter, phase II/III; randomized noninferiority
opment in both oral and intravenous formulations for the study of hospitalized adults with suspected or microbiologi-
treatment of uncomplicated and cUTIs. It has antimicrobial cally confirmed cUTIs and acute pyelonephritis (54%), 233 and
activity similar to other carbapenems except it has no activity 231 were treated with intravenous fosfomycin (6 g every 8 h)
against P. aeruginosa [97]. and piperacillin/tazobactam (4.5 g every 8 h), respectively.
Fosfomycin was not inferior to piperacillin/tazobactam in
the overall response (primary endpoint: clinical cure and
Aminoglycosides
microbiologic eradication at test of cure) (64.7% [119/184] vs.
Despite being not widely used because of concerns of toxicity, 54.5% [97/178], respectively, with a difference of 10.2% [95%
aminoglycosides are still an important therapeutic option for CI −0.4, 20.8]). The clinical and microbiological cure rates were
treatment of infections caused by MDR organisms for which similar between the groups: 90.8% (167/184) for fosfomycin
treatment options are limited (Table 2) [98]. However, most of versus 91.6% (163/178) for piperacillin/tazobactam and 66%
these MDR organisms carry plasmids that often harbor ami- (127/184) for fosfomycin and 57.3% (102/178) for piperacillin/
noglycoside-modifying enzymes (AMEs)-encoding genes able tazobactam, respectively [106].
to modify the clinically available aminoglycosides [99,100]. The major concerns of fosfomycin are the development of
Plazomicin is a new aminoglycoside derivative of sisomicin resistance during treatment and clinical failure with infections
that is resilient to the activity of all clinically relevant AMEs, the due to P aeruginosa particularly when used as monotherapy.
primary mechanism of aminoglycoside resistance [99–101]. It is Therefore, it should be combined with other antibiotics for the
approved by FDA for adults with cUTIs and acute pyelonephritis, treatment of infections due to MDR pathogens [107–110].
caused by certain Enterobacteriales in patients who have limited Eravacycline is a novel synthetic tigecycline analogue with
or no alternative treatment options (Table 2) [102]. broad-spectrum antibacterial activity against gram-negative
A multicenter, randomized, double-blind, phase 2 study organisms including ESBLs-Enterobacteriales, CRE, acinetobacter
comparing plazomicin with Levofloxacin showed that plazo- baumannii, and polymixin resistant strains and against Gram-
micin (10 or 15 mg/kg) given once daily for 5 days achieved positive pathogens, such as methicillin-susceptible and -resistant
microbiological eradication and clinical cure in 85% and 80% staphylococci, vancomycin-susceptible and -resistant enterococci.
of patients with cUTIs and acute pyelonephritis, respec- It has no activity against Pseudomonas aeruginosa [111].
tively [103]. A phase 3 study evaluating eravacycline (1.5 mg/kg daily)
In a multinational, randomized, double-blind, non- versus ertapenem (1 g daily) for a minimum of 5 days with an
inferiority phase III study, 609 patients with cUTIs, including oral stepdown for the treatment of cUTIs and acute pyelone-
acute pyelonephritis (40-44%), were randomized in to receive phritis (IGNITE2) did not demonstrate noninferiority. The co-
intravenous plazomicin (15 mg per kilogram of body weight primary endpoints of responder rates at test of cure were
once daily) or meropenem (1 g every 8 h) for a total of 7 to 10 68.5% and 74.9% for eravacycline and ertapenem, respectively
days of therapy [103]. Among the 382 patients with (−6.5% CI: −12.6%, −0.3%) [112]. Despite its broad antibacterial
Enterobacteriales in the microbiologic modified intention-to- activity and high urine concentration, it should not be used for
treat population, 28.0%, 30.1%, and 26.4% had uropathogens the treatment of cUTIs or acute pyelonephritis till further
with an ESBLs phenotype, multidrug-resistant uropathogens, studies demonstrating its efficacy.
and uropathogens that were not susceptible to other amino- Omadacycline is a semisynthetic tetracycline derivative that
glycosides, respectively. The primary end point (composite exhibits activity against Gram-positive and Gram-negative
clinical cure and microbiologic eradication) at test of cure for aerobes, anaerobes, and atypical bacteria. Its use in treatment
plazomicin vs. meropenem was 81.7% vs. 70.1% (difference of UTIs is in its preliminary stage [113].
11.6%, 95%CI 2.7–20.3%). Plazomicin achieved higher micro- Aztreonam is a β-lactam antibiotic and the only clinically
biological eradication than meropenem (89.5% vs. 74.6%, dif- available member of its monobactam class. Aztreonam remains
ference 14.9%, 95% CI 7.0–22.7%) including eradication of an option for treating infections due to MBLs) producing gram-
Enterobacteriales that were not susceptible to aminoglyco- negative organisms that test susceptible to this agent. However,
sides (78.8% vs. 68.6%) and ESBLs-Enterobacterales (82.4% vs. these carbapenemase-producing bacteria are often coupled with
75.0%). It was not associated with increased risk of nephro- additional resistance mechanisms, such as ESBLs and AmpC-type
toxicity or vestibular toxicity [104]. enzymes, which confer resistance to aztreonam (Table 2) [104].
Therefore, aztreonam should not be used as empiric monother-
apy for cUTIs or acute pyelonephritis due to high resistance and
Other antimicrobial agents
failure rate [114]. Combing aztreonam with other antibiotics
Fosfomycin in its intravenous formulation (fosfomycin diso- (ceftazidime-avibactam, amoxicillin-clavulanic acid) or β -lactam
dium) is being developed for treatment of cUTIs and acute inhibitors (avibactam, zidebactam, nacubactam, and WCK 5153)
pyelonephritis due to MDR-organisms in the United States, enhances activity against these resistant organisms [37,115–118].
246 M. S. BADER ET AL.

Temocillin is a 6-α-methoxy derivative of ticarcillin that Declaration of interest


shows in vitro activity against AmpC-, ESBLs-, and KPC- The contents of the paper and the opinions expressed within are those of
producing Enterobacteriales with minimal risk of Clostridium the authors, and it was the decision of the authors to submit the manu-
difficile infection [119]. It can be used for treatment UTIs as script for publication.
carbapenem-sparing antibiotic. Current recommended regi- None of the authors has any conflict of interest.
mens are 2 g q12h or 2 g q8h depending on infection severity Peer reviewers on this manuscript have no relevant financial or other
relationships to disclose.
and strain susceptibility [118]. However, there is very limited
data on its clinical efficacy in the treatment of UTIs and debate
on its susceptibility breakpoints [120].
References
1. Tandogdu Z, Wagenlehner FM. Global epidemiology of urinary
Conclusion tract infections. Curr Opin Infect Dis. 2016;29(1):73–79.
2. Gupta K, Hooton TM, Naber KG, et al. Infectious Diseases Society of
There are recently new antimicrobial agents (ceftolozane/ America; European Society for Microbiology and Infectious
tazobactam, ceftazidime/avibactam, meropenem/vaborbac- Diseases. International clinical practice guidelines for the treatment
tam, plazomicin) that are approved for the treatment of of acute uncomplicated cystitis and pyelonephritis in women:
A 2010 update by the Infectious Diseases Society of America and
cUTIs and acute pyelonephritis due to MDR-organisms. It is the European Society for Microbiology and Infectious Diseases. Clin
crucial to use these new antimicrobials only as last-resort Infect Dis. 2011;52(5):e103–20.
treatments when UTIs caused by MDR-organisms that resis- 3. Johansen TE, Botto H, Cek M, et al. Critical review of current definitions
tant to old antimicrobial options are confirmed or strongly of urinary tract infections and proposal of an EAU/ESIU classification
system. Int J Antimicrob Agents. 2011;38(Suppl.):64–70.
suspected to safe-guard these antibiotics from resistance
4. Sader HS, Castanheira M, Duncan LR, et al. Antimicrobial suscept-
development. It is also strongly recommended to use old ibility of enterobacteriaceae and pseudomonas aeruginosa isolates
existing antimicrobial agents wisely in the treatment of UTIs from united states medical centers stratified by infection type:
according to local antimicrobial susceptibility information, results from the international network for optimal resistance mon-
clinical settings, and patient’s risk factor for antimicrobial itoring (INFORM) surveillance program, 2015-2016. Diagn Microbiol
Infect Dis. 2018;92(1):69–74.
resistance. Nitrofurantoin is the first recommended empiric
5. European Association of Urology (EAU). Guidelines on urological
antibiotic therapy for uncomplicated cystitis due to its high infections. 2017. Available from: http://uroweb.org/guideline/urolo
activity against E. coli, safety profile, and limited impact on gical-infections/.
the intestinal microbiota [2,5]. Oral cephalosporins can be 6. Bader MS, Loeb M, Brooks AA. An update on the management of
used as empiric treatment of uncomplicated cystitis, if urinary tract infections in the era of antimicrobial resistance.
neither nitrofurantoin nor fosfomycin can be used, and as Postgrad Med. 2017;129(2):242–258.
7. Karlowsky JA, Legace-Wiens P, Adam H, et al. In vitro susceptibility
a step down treatment from parenteral therapy of acute of urinary isolates of escherichia coli to first- and second-line
pyelonephritis if the isolated organisms are susceptible empirically prescribed oral antimicrobial agents: CANWARD surveil-
[7,41,42]. lance study results for Canadian outpatients from 2007 to 2016.
Fluoroquinolones and trimethoprim-sulfamethoxazole are Int J Antimicrob Agents. 2019. pii: S0924-8579(19)30106-2s.
8. Rank EL, Lodise T, Avery L, et al. Antimicrobial susceptibility trends
not recommended for empiric treatment of uncomplicated
observed in urinary pathogens obtained from New York state.
cystitis, cUTIs with risk of fluoroquinolones resistance or Open Forum Infect Dis. 2018;5(11):1–6.
ESBLs- Enterobacteriales, acute pyelonephritis, and hospital- 9. Ny S, Edquist P, Dumpis U, et al. Antimicrobial resistance of
acquired UTIs [7,9,23,25]. The prevalence of UTIs due to Escherichia coli isolates from outpatient urinary tract infections in
ESBLs Enterobacteriales even in outpatient settings is women in six European countries including Russia. J Glob
Antimicrob Resist. 2018;17:25–34.
increasing [7–9]. Carbapenems remain the antibiotic of
10. Morrill HJ, Morton JB, Caffrey AR, et al. Antimicrobial Resistance of
choice with proven efficacy for patients with cUTIs and Escherichia coli Urinary Isolates in the Veterans Affairs Health Care
acute pyelonephritis, particularly severe infections, due to System. Antimicrob Agents Chemother. 2017;61(5):e02236–16.
ESBL-Enterobacteirales [70,82]. Piperacillin-tazobactam might 11. Huttner A, Verhaegh EM, Harbarth S, et al. Nitrofurantoin revisited:
have a role in the treatment of mild-moderate UTIs caused a systematic review and meta-analysis of controlled trials.
J Antimicrob Chemother. 2015;70(9):2456–2464.
by susceptible ESBL-E coli or when the MIC is low while
12. Ahmed H, Farewell D, Francis NA, et al. Risk of adverse outcomes
cefepime should not be used for this indication [80]. following urinary tract infection in older people with renal impair-
Although ceftolozane/tazobactam and ceftazidime/avibac- ment: retrospective cohort study using linked health record data.
tam provide carbapenem-sparing options for the treatment PloS Med. 2018;15(9):e1002652.
of ESBLs infections, they should be reserved for the treat- 13. Mezzatesta ML, La Rosa G, Maugeri G, et al. In vitro activity of
fosfomycin trometamol and other oral antibiotics against
ment of cUTIs and acute pyelonephritis due to MDR-
multidrug-resistant uropathogens. Int J Antimicrob Agents.
P aeruginosa (both) and CRE (ceftazidime/avibactam only) 2017;49(6):763–766.
[44,56]. Meropenem–vaborbactam is the preferred therapy 14. Giancola SE, Mahoney MV, Hogan MD, et al. Assessment of fosfo-
for cUTIs and acute pyelonephritis due to KPC-producing mycin for complicated or multidrug-resistant urinary tract infec-
Enterobacteriales because of efficacy and potential barrier tions: patient characteristics and outcomes. Chemotherapy.
2017;62(2):100–104.
for resistance development [89,90,92]. Consultation with an
15. Cai T, Cocci A, Verze P, et al. The use of oral fosfomycin-trometamol
infectious diseases specialist, microbiologist, or antimicrobial in patients with catheter-associated urinary tract infections (CAUTI):
stewardship team for the management and treatment of new indications for an old antibiotic?. J Chemother. 2018;30
UTIs due to resistant organisms is highly recommended. (5):290–295.
POSTGRADUATE MEDICINE 247

16. Matthews PC, Barrett LK, Warren S, et al. Oral fosfomycin for treat- 35. Bouxom H, Fournier D, Bouiller K, et al. Which non-carbapenem
ment of urinary tract infection: a retrospective cohort study. BMC antibiotics are active against extended-spectrum β-lactamase-
Infect Dis. 2016;16(1):556. producing Enterobacteriaceae?. Int J Antimicrob Agents. 2018;52
17. Quek WM, Teng CB, Tan YZ, et al. Outcomes of Fosfomycin Use in (1):100–103.
Ceftriaxone-Resistant Enterobacteriaceae Urinary Tract Infection in 36. Veve MP, Wagner JL, Kenney RM, et al. Comparison of fosfomycin
the Elderly. Int J Antimicrob Agents. 2019;53(2):195–196. to ertapenem for outpatient or step-down therapy of extended-
18. Senol T, Tasbakan M, Pullukcu H, et al. Carbapenem versus fosfo- spectrum β-lactamase urinary tract infections. Int J Antimicrob
mycin tromethanol in the treatment of extended-spectrum Agents. 2016;48(1):56–60.
beta-lactamase-producing Escherichia coli- related complicated 37. Emeraud C, Escaut L, Boucly A, et al. Aztreonam plus clavulanate,
lower urinary tract infection. J Chemother. 2010;22:355–357. tazobactam, or avibactam for treatment of infections caused by
19. Peretz A, Naamneh B, Tkhawkho L, et al. High Rates of Fosfomycin metallo-β-lactamase-producing gram-negative bacteria.
Resistance in Gram-Negative Urinary Isolates from Israel. Microb Antimicrob Agents Chemother. 2019;63(5):e00010–19.
Drug Resist. 2019;25(3):408–412. 38. Tamma PD, Conley AT, Cosgrove SE, et al. Antibacterial resistance
20. van Den Bijllaardt W, Schijffelen MJ, Bosboom RW, et al. leadership group. Association of 30-day mortality with oral
Susceptibility of ESBL Escherichia coli and Klebsiella pneumoniae step-down vs continued intravenous therapy in patients hospita-
to fosfomycin in the Netherlands and comparison of several testing lized with enterobacteriaceae bacteremia. JAMA Intern Med.
methods including E test, MIC test strip, Vitek2, Phoenix and disc 2019;179(3):316–323.
diffusion. J Antimicrob Chemother. 2018;73(9):2380–2387. 39. Wang SS, Ratliff PD, Judd WR. Retrospective review of ceftriaxone
21. Wijma RX, Koch BCP, van Gelder T, et al. High interindividual versus levofloxacin for treatment of E. coli urinary tract infections.
variabilityin urinary fosfomycin concentrations in healthy female Int J Clin Pharm. 2018;40(1):143–149.
volunteers. Clin Microbiol Infect. 2018;24:528–532. 40. Chua KYL, Stewardson AJ. Individual and community predictors of
22. Huttner A, Kowalczyk A, Turjeman A, et al. Effect of 5-day nitrofur- urinary ceftriaxone-resistant Escherichia coli isolates, Victoria,
antoin vs single-dose fosfomycin on clinical resolution of uncom- Australia. Antimicrob Resist Infect Control. 2019;8:36.
plicated lower urinary tract infection in women: A randomized 41. Moustafa F, Nguyen G, Mathevon T, et al. Evaluation of the efficacy
clinical trial. JAMA. 2018;319(17):1781–1789. and tolerance of a short 7 day third-generation cephalosporin
23. Delisle G, Quach C, Domingo MC, et al. Escherichia coli antimicro- treatment in the management of acute pyelonephritis in young
bial susceptibility profile and cumulative antibiogram to guide women in the emergency department. J Antimicrob Chemother.
empirical treatment of uncomplicated urinary tract infections in 2016;71(6):1660–1664.
women in the province of Québec, 2010-15. J Antimicrob 42. Lojanapiwat B, Nimitvilai S, Bamroongya M, et al. Oral sitafloxacin
Chemother. 2016;71(12):3562–3567. vs intravenous ceftriaxone followed by oral cefdinir for acute pye-
24. Crellin E, Mansfield KE, Leyrat C, et al. Trimethoprim use for urinary lonephritis and complicated urinary tract infection: a randomized
tract infection and risk of adverse outcomes in older patients: controlled trial. Infect Drug Resist. 2019;12:173–181.
cohort study. BMJ. 2018;360:341. 43. National Institute for Health Care and Excellence (NICE).
25. FDA drug safety communication. US Food and Drug Pyelonephritis (acute): antimicrobial prescribing. Available from:
Administration. Available from: https://www.fda.gov/downloads/ https://www.nice.org.uk/guidance/ng111/chapter/
Drugs/DrugSafety/UCM612834.pdf. Recommendations#choice-of-antibiotic.
26. Kavatha D, Giamarellou H, Alexiou Z, et al. Cefpodoxime-proxetil 44. Shirley M. Ceftazidime-avibactam: a review in the treatment of
versus trimethoprim-sulfamethoxazole for short-term therapy of serious gram-negative bacterial infections. Drugs. 2018;78
uncomplicated acute cystitis in women. Antimicrob Agents (6):675–692.
Chemother. 2003;47:897–900. 45. Stone GG, Bradford PA, Yates K, et al. In vitro activity of ceftazi-
27. López IA, Montes JC, Álvarez MJ, et al. Cephalothin is not a reliable dime/avibactam against urinary isolates from patients in a Phase 3
surrogate marker for oral cephalosporins in susceptibility testing of clinical trial programme for the treatment of complicated urinary
Enterobacteriaceae causing urinary tract infection. Diagn Microbiol tract infections. J Antimicrob Chemother. 2017;72(5):1396–1399.
Infect Dis. 2016;86:412–416. 46. Sader HS, Castanheira M, Flamm RK, et al. Antimicrobial activities of
28. Bunnell KL, Wenzler E, Harrington AT, et al. Impact of Clinical and ceftazidime-avibactam and comparator agents against
Laboratory Standards Institute breakpoint changes on susceptibil- gram-negative organisms isolated from patients with urinary tract
ity rates of cephalosporins in uncomplicated urinary tract infections infections in U.S. medical centers, 2012 to 2014. Antimicrob Agents
caused by Enterobacteriaceae. Diagn Microbiol Infect Dis. 2018;90 Chemother. 2016;60(7):4355–4360.
(4):335–336. 47. Garcia-Castillo M, Garcia-Fernandez S, Gomez-Gil R, et al. Activity of
29. Shehab N, Lovegrove MC, Geller AI, et al. US emergency depart- ceftazidime-avibactam against carbapenemase-producing
ment visits for outpatient adverse drug events, 2013–2014. JAMA. Enterobacteriaceae from urine specimens obtained during the
2016;316:2115–2125. infection-carbapenem resistance evaluation surveillance trial
30. Ahmed H, Farewell D, Francis NA, et al. Choice of empirical anti- (iCREST) in Spain. Int J Antimicrob Agents. 2018;51(3):511–515.
biotic therapy and adverse outcomes in older adults with sus- 48. Castanheira M, Mendes RE, Sader HS. Low frequency of
pected urinary tract infection: cohort study. Open Forum Infect ceftazidime-avibactam resistance among enterobacteriaceae iso-
Dis. 2019;6:3. lates carrying blakpc collected in U.S. hospitals from 2012 to
31. Fuchs F, Hamprecht A. Results from a Prospective In Vitro Study on 2015. Antimicrob Agents Chemother. 2017;61(3):e02369–16.
the Mecillinam (Amdinocillin) Susceptibility of Enterobacterales. 49. Mendes RE, Castanheira M, Woosley LN, et al. Characterization of β-
Antimicrob Agents Chemother. 2019 Mar 27;63(4):e02402–18. lactamase content of ceftazidime-resistant pathogens recovered
32. Monsen TJ, Holm SE, Ferry BM, et al. Mecillinam resistance and during the pathogen-directed Phase 3 trial (REPRISE) for ceftazi-
outcome of evmecillinam treatment in uncomplicated lower urin- dime-avibactam: correlation of efficacy against β-lactamase produ-
ary tract infection in women. APMIS. 2014;122:317–323. cers. Antimicrob Agents Chemother. 2019;63:e02655–18.
33. Soraas A, Sundsfjord A, Jorgensen SB, et al. High rate of per oral 50. Wagenlehner FM, Sobel JD, Newell P, et al. Ceftazidime-avibactam
mecillinam treatment failure in community-acquired urinary tract versus doripenem for the treatment of complicated urinary tract
infections caused by ESBL-producing Escherichia coli. PloS One. infections, including acute pyelonephritis: RECAPTURE, a Phase 3
2014;9:e85889. randomized trial program. Clin Infect Dis. 2016;63:754–762.
34. Malcolm W, Fletcher E, Kavanagh K, et al. Risk factors for resistance 51. Carmeli Y, Armstrong J, Laud PJ, et al. Ceftazidime-avibactam or
and MDR in community urine isolates: population-level analysis best available therapy in patients with ceftazidime-resistant
using the NHS Scotland Infection Intelligence Platform. Enterobacteriaceae and Pseudomonas aeruginosa complicated
J Antimicrob Chemother. 2018;73(1):223–230. urinary tract infections or complicated intra-abdominal infections
248 M. S. BADER ET AL.

(REPRISE): a randomised, pathogen-directed, phase 3 study. Lancet patients with pyelonephritis, United States (1). Emerg Infect Dis
Infect Dis. 2016;16:661–673. 2016;22(9):1594–1603.
52. Vazquez JA, González Patzán LD, Stricklin D, et al. Efficacy and 66. Malaisri C, Phuphuakrat A, Wibulpolprasert A, et al. A randomized
safety of ceftazidime-avibactam versus imipenem-cilastatin in the controlled trial of sitafloxacin vs. ertapenem as a switch therapy
treatment of complicated urinary tract infections, including acute after treatment for acute pyelonephritis caused by extended-
pyelonephritis, in hospitalized adults: results of a prospective, spectrum β-lactamase-producing Escherichia coli: A pilot study.
investigator-blinded, randomized study. Curr Med Res Opin. J Infect Chemother. 2017;23(8):556–562.
2012;28(12):1921–1931. 67. Vente A, Bentley C, Lückermann M, et al. Early clinical assessment
53. Sternbach N, Leibovici Weissman Y, Avni T, et al. Efficacy and safety of the antimicrobial activity of finafloxacin compared to ciproflox-
of ceftazidime/avibactam: a systematic review and meta-analysis. acin in subsets of microbiologically characterized isolates.
J Antimicrob Chemother. 2018;73(8):2021–2029. Antimicrob Agents Chemother. 2018;62(4):e02325–17.
54. Mendes RE, Castanheira M, Woosley LN, et al. Molecular β- 68. Dalhoff A, Schubert S, Vente A. Pharmacodynamics of finafloxacin,
lactamase characterization of Gram-negative pathogens recovered ciprofloxacin, and levofloxacin in serum and urine against TEM-
from patients enrolled in the ceftazidime-avibactam phase 3 trials and SHV-type extended-spectrum-β-lactamase-producing entero-
(RECAPTURE 1 and 2) for complicated urinary tract infections: bacteriaceae isolates from patients with urinary tract infections.
efficacies analysed against susceptible and resistant subsets. Antimicrob Agents Chemother. 2017;61(5):e02446–16.
Int J Antimicrob Agents. 2018;52(2):287–292. 69. Wagenlehner F, Nowicki M, Bentley C, et al. Explorative randomized
55. Shields RK, Nguyen MH, Chen L, et al. Pneumonia and renal repla- phase ii clinical study of the efficacy and safety of finafloxacin
cement therapy are risk factors for ceftazidime-avibactam treat- versus ciprofloxacin for treatment of complicated urinary tract
ment failures and resistance among patients with infections. Antimicrob Agents Chemother. 2018;62(4):e02317.
carbapenem-resistant enterobacteriaceae infections. Antimicrob 70. Rattanaumpawan P, Werarak P, Jitmuang A, et al. Efficacy and
Agents Chemother. 2018;62(5):e02497–17. safety of de-escalation therapy to ertapenem for treatment of
56. Ceftolozane/Tazobactam (Zerbaxa)–a new intravenous antibiotic. infections caused by extended-spectrum-β-lactamase-producing
Med Lett Drugs Ther. 2015;57(1463):31–33. Enterobacteriaceae: an open-label randomized controlled trial.
57. Pfaller MA, Bassetti M, Duncan LR, et al. Ceftolozane/tazobactam BMC Infect Dis. 2017;17(1):183.
activity against drug-resistant Enterobacteriaceae and 71. Ramasubramanian V, Murlidharan P, Nambi S, et al. Efficacy and
Pseudomonas aeruginosa causing urinary tract and intraabdom- cost comparison of ertapenem as outpatient parenteral antimicro-
inal infections in Europe: report from an antimicrobial surveil- bial therapy in acute pyelonephritis due to extended- spectrum
lance programme (2012-15). J Antimicrob Chemother. beta-lactamase-producing Enterobacteriaceae. Indian J Nephrol.
2017;72:1386–1395. 2018;28:.351–357.
58. Wagenlehner FM, Umeh O, Steenbergen J, et al. Ceftolozane- 72. Lob SH, Hackel MA, Hoban DJ, et al. Activity of Ertapenem against
tazobactam compared with levofloxacin in the treatment of com- Enterobacteriaceae in seven global regions-SMART 2012-2016. Eur
plicated urinary-tract infections, including pyelonephritis: J Clin Microbiol Infect Dis. 2018;37(8):1481–1489.
a randomised, double-blind, phase 3 trial (ASPECT-cUTI. Lancet. 73. Rad MA, Zhong LH, Llorin RM, et al. Ertapenem in outpatient
2015;385:1949–1956. parenteral antimicrobial therapy for complicated urinary tract
59. Popejoy MW, Paterson DL, Cloutier D, et al. Efficacy of ceftolozane/ infections. J Chemother. 2017;29(1):25–29.
tazobactam against urinary tract and intra-abdominal infections 74. Tamma PD, Han JH, Rock C, et al. Carbapenem therapy is asso-
caused by ESBL-producing Escherichia coli and Klebsiella pneumo- ciated with improved survival compared with
niae: a pooled analysis of Phase 3 clinical trials. J Antimicrob piperacillin-tazobactam for patients with extended-spectrum
Chemother. 2017;72(1):268–272. beta-lactamase bacteremia. Clin Infect Dis. 2015;60:1319–1325.
60. SUPERIOR Study Group, García-Fernández S, García-Castillo M, 75. Rodriguez-Bano J, Navarro MD, Retamar P, et al. beta-Lactam/beta-
Bou G, et al. Activity of ceftolozane/tazobactam against lactam inhibitor combinations for the treatment of bacteremia due
Pseudomonas aeruginosa and Enterobacterales isolates recovered to extended-spectrum beta-lactamase-producing Escherichia coli:
from intensive care unit patients in Spain: the SUPERIOR multi- a post hoc analysis of prospective cohorts. Clin Infect Dis.
centre study. Int J Antimicrob Agents 2019;53(5):682–688. 2012;54:167–174.
61. Zhanel GG, Golden AR, Zelenitsky S, et al. Cefiderocol: 76. Ng TM, Khong WX, Harris PN, et al. Empiric piperacillin-tazobactam
A siderophore cephalosporin with activity against versus carbapenems in the treatment of bacteraemia due to
carbapenem-resistant and multidrug-resistant gram-negative extended-spectrum beta-lactamase-producing Enterobacteriaceae.
bacilli. Drugs. 2019;79(3):271–289. PLoS One. 2016;11:e0153696.
62. Jacobs MR, Abdelhamed AM, Good CE, et al. ARGONAUT-I: activity 77. Yoon YK, Kim JH, Sohn JW, et al. Role of piperacillin/tazobactam as
of Cefiderocol (S-649266), a Siderophore cephalosporin, against a carbapenem-sparing antibiotic for treatment of acute pyelone-
gram-negative bacteria, including carbapenem-resistant nonfer- phritis due to extended-spectrum beta-lactamase-producing
menters and enterobacteriaceae with defined extended-spectrum Escherichia coli. Int J Antimicrob Agents. 2017;49:410–415.
β-lactamases and carbapenemases. Antimicrob Agents Chemother. 78. Park SH, Choi SM, Chang YK, et al. The efficacy of non-carbapenem
2018;63(1):e01801–18. antibiotics for the treatment of community-onset acute pyelone-
63. Hsueh SC, Lee YJ, Huang YT, et al. In vitro activities of cefiderocol, phritis due to extended-spectrum beta-lactamase-producing
ceftolozane/ tazobactam,ceftazidime/avibactam and other com- Escherichia coli. J Antimicrob Chemother. 2014;69:2848–2856.
parative drugs against imipenem-resistant Pseudomonas aerugi- 79. Sfeir M, Askin G, Christos P. Beta-lactam/beta-lactamase inhibitors
nosa and Acinetobacter baumannii, and Stenotrophomonas versus carbapenem for bloodstream infections due to extended
maltophilia, all associated with bloodstream infections in Taiwan. spectrum beta- lactamase producing Enterobacteriaceae: systema-
J Antimicrob Chemother. 2019;74(2):380–386. tic review and meta-analysis. Int J Antimicrob Agents. 2018;52
64. Portsmouth S, van Veenhuyzen D, Echols R, et al. Cefiderocol versus (5):554–570.
imipenem-cilastatin for the treatment of complicated urinary tract 80. Son SK, Lee NR, Ko J-H, et al. Clinical effectiveness of carbapenems
infections caused by Gram-negative uropathogens: a phase 2, versus alternative antibiotics for treating ESBL-producing
randomised, double-blind, non-inferiority trial. Lancet Infect Dis. Enterobacteriaceae bacteraemia: a systematic review and
2018;18(12):1319–1328. meta-analysis. J Antimicrob Chemother. 2018;73:2631–2642.
65. EMERGENCY ID Net Study Group, Talan DA, Takhar SS, 81. Seo YB, Lee J, Kim YK, et al. Randomized controlled trial of
Krishnadasan A, et al. Fluoroquinolone-resistant and extended- piperacillin-tazobactam, cefepime and ertapenem for the treat-
spectrum β-lactamase-producing escherichia coli infections in ment of urinary tract infection caused by extended-spectrum
POSTGRADUATE MEDICINE 249

beta-lactamase-producing Escherichia coli. BMC Infect Dis. 2017;17 97. Karlowsky JA, Adam HJ, Baxter MR, et al. In vitro activity of sulo-
(1):404. penem, an oral penem, against urinary isolates of escherichia coli.
82. Harris PNA, Tambyah PA, Lye DC, et al. MERINO trial investigators Antimicrob Agents Chemother. 2018 Dec 21;63(1):e01832–18.
and the Australasian society for infectious disease clinical research 98. Goodlet KJ, Benhalima FZ, Nailor MD. A systematic review of
network (ASID-CRN) . effect of piperacillin-tazobactam vs merope- single-dose aminoglycoside therapy for urinary tract infection: is
nem on 30-day mortality for patients with e coli or klebsiella it time to resurrect an old strategy? Antimicrob Agents Chemother.
pneumoniae bloodstream infection and ceftriaxone resistance: 2018;63(1):e02165–18.
a randomized clinical trial. JAMA. 2018;320(10):984–994. 99. Castanheira M, Deshpande LM, Woosley LN, et al. Activity of plazomi-
83. Falagas ME, Tansarli GS, Ikawa K, et al. Clinical outcomes with cin compared with other aminoglycosides against isolates from
extended or continuous versus short-term intravenous infusion of European and adjacent countries, including Enterobacteriaceae mole-
carbapenems and piperacillin/tazobactam: a systematic review and cularly characterized for aminoglycoside-modifying enzymes and
meta-analysis. Clin Infect Dis. 2013;56:272–282. other resistance mechanisms. J Antimicrob Chemother. 2018;73
84. Guet-Revillet H, Tomini E, Emirian A, et al. Piperacillin/tazobac- (12):3346–3354.
tam as an alternative antibiotic therapy to carbapenems in the 100. Castanheira M, Davis AP, Serio AW, et al. In vitro activity of
treatment of urinary tract infections due to extended-spectrum Plazomicin against Enterobacteriaceae isolates carrying genes
β-lactamase-producing Enterobacteriaceae: an in silico pharma- encoding aminoglycoside-modifying enzymes most common in
cokinetic study. Int J Antimicrob Agents. 2017;49(1):62–66. US Census divisions. Diagn Microbiol Infect Dis. 2019;94(1):73–77.
85. Kim SA, Altshuler J, Paris D, et al. Cefepime versus carbapenems for the 101. Walkty A, Karlowsky JA, Baxter MR, et al. In vitro activity of plazo-
treatment of urinary tract infections caused by extended-spectrum β- micin against gram-negative and gram-positive bacterial patho-
lactamase-producing enterobacteriaceae. Int J Antimicrob Agents. gens isolated from patients in Canadian hospitals from 2013 to
2018;51(1):155–158. 2017 as part of the CANWARD surveillance study. Antimicrob
86. Lee NY, Lee CC, Huang WH, et al. Cefepime therapy for monomi- Agents Chemother. 2018 Dec 21;63(1):e02068–18.
crobial bacteremia caused by cefepime-susceptible extended- 102. Plazomicin (Zemdri) - a new aminoglycoside antibiotic. Med Lett
spectrum β-lactamase-producing Enterobacteriaceae: MIC matters. Drugs Ther. 2018;60(1559):180–182.
Clin Infect Dis. 2013;56:488–495. 103. Connolly LE, Riddle V, Cebrik D, et al. A multicenter, randomized,
87. Wang R, Cosgrove SE, Tschudin-Sutter S, et al. Cefepime therapy for double-blind, phase 2 study of the efficacy and safety of plazomi-
cefepime-susceptible extended-spectrum β-lactamase– producing cin compared with levofloxacin in the treatment of complicated
Enterobacteriaceae bacteremia. Open Forum Infect Dis. urinary tract infection and acute pyelonephritis. Antimicrob Agents
2016;17:404. Chemother. 2018;62(4):e01989–17.
88. Chopra T, Marchaim D, Veltman J, et al. Impact of cefepime therapy on 104. Wagenlehner FME, Cloutier DJ, Komirenko AS, et al. EPIC study
mortality among patients with bloodstream infections caused by group. once-daily plazomicin for complicated urinary tract
extended-spectrum β-lactamase-producing Klebsiella pneumoniae infections. N Engl J Med. 2019;380(8):729–740.
and Escherichia coli. Antimicrob Agents Chemother. 105. Burgos RM, Rodvold KA. ZTI-01 (fosfomycin for injection) in the
2012;56:3936–3942. treatment of hospitalized patients with complicated urinary tract
89. Hackel MA, Lomovskaya O, Dudley MN, et al. Activity of infections. Future Microbiol. 2019;14:461–475.
meropenem-vaborbactam against clinical isolates of KPC-positive 106. Kaye KS, Rice LB, Dane A, et al. Fosfomycin for injection (ZTI-01) vs
Enterobacteriaceae. Antimicrob Agents Chemother. 2017;21(62): piperacillin-tazobactam (PIP-TAZ) for the treatment of complicated
e01904–17. urinary tract infection (cUTI) including acute pyelonephritis (AP):
90. Meropenem/vaborbactam (Vabomere) for complicated urinary ZEUS, A phase 2/3 randomized trial. Clin Infect Dis. 2019. [cited 6
tract infection. Med Lett Drugs Ther. 2018;60(1549):103–105. Mar 2019].
91. Kaye KS, Bhowmick T, Metallidis S, et al. Effect of meropenem- 107. Dimopoulos G, Koulenti D, Parker SL, et al. Intravenous fosfomycin
vaborbactam vs piperacillin-tazobactam on clinical cure or for the treatment of multidrug-resistant pathogens: what is the
improvement and microbial eradication in complicated urinary evidence on dosing regimens? Expert Rev Anti Infect Ther.
tract infection: the TANGO I randomized clinical trial. JAMA. 2019;17(3):201–210.
2018;319(8):788–799. 108. Aghamali M, Sedighi M, Zahedi Bialvaei A, et al. Fosfomycin:
92. Wunderink RG, Giamarellos-Bourboulis EJ, Rahav G, et al. Effect and mechanisms and the increasing prevalence of resistance. J Med
safety of meropenem-vaborbactam versus best-available therapy in Microbiol. 2019;68(1):11–25.
patients with carbapenem-resistant enterobacteriaceae infections: 109. Albiero J, Mazucheli J, Barros JPDR, et al. Pharmacodynamic attain-
the TANGO II randomized clinical trial. Infect Dis Ther. 2018;7 ment of the synergism of meropenem and fosfomycin combination
(4):439–455. against Pseudomonas aeruginosa producing metallo-β-lactamase.
93. Lomovskaya O, Castanheira M, Vazquez J, et al. Assessment of MIC Antimicrob Agents Chemother. 2019;63:e00126–19.
increases with meropenem-vaborbactam and ceftazidime- 110. Florent A, Chichmanian RM, Cua E, et al. Adverse events associated
avibactam in Tango II (a phase 3 study of the treatment of CRE with intravenous fosfomycin. Int J Antimicrob Agents. 2011;37:82–83.
infections. San Diego, CA: Infectious diseases week, Infectious 111. Zhanel GG, Baxter MR, Adam HJ, et al. In vitro activity of eravacycline
Disease Society of America; 2017. against 2213 Gram-negative and 2424 Gram-positive bacterial patho-
94. Sun D, Rubio-Aparicio D, Nelson K, et al. Meropenem- gens isolated in Canadian hospital laboratories: CANWARD surveil-
vaborbactam resistance selection, resistance prevention, and lance study 2014-2015. Diagn Microbiol Infect Dis. 2018;91(1):55–62.
molecular mechanisms in mutants of KPC-producing Klebsiella 112. Tetraphase announces top-line results from IGNITE3 phase 3 clin-
pneumoniae. Antimicrob Agents Chemother. 2017;61(12): ical trial of eravacycline in complicated urinary tract infections
e01694–17. (cUTI). tetraphase pharmaceuticals. Available from: https://ir.
95. Karlowsky JA, Lob SH, Kazmierczak KM, et al. In vitro activity of tphase.com/news-releases/news-release-details/tetraphase-
imipenem/relebactam against Gram-negative ESKAPE patho- announces-top-line-results-ignite3-phase-3-clinical.
gens isolated in 17 European countries: 2015 SMART surveil- 113. Overcash JS, Bhiwandi P, Garrity-Ryan L, et al. Pharmacokinetics,
lance programme. J Antimicrob Chemother. 2018;73 safety, and clinical outcomes of omadacycline in women with
(7):1872–1879. cystitis: results from a phase 1b study. Antimicrob Agents
96. Sims M, Mariyanovski V, McLeroth P, et al. Prospective, randomized, Chemother. 2019;63(5):e02083–18.
double-blind, Phase 2 dose-ranging study comparing efficacy and 114. Hogan M, Bridgeman MB, Min GH, et al. Effectiveness of empiric
safety of imipenem/cilastatin plus relebactam with imipenem/cilas- aztreonam compared to other beta-lactams for treatment of
tatin alone in patients with complicated urinary tract infections. Pseudomonas aeruginosa infections. Infect Drug Resist.
J Antimicrob Chemother. 2017;72(9):2616–2626. 2018;11:1975–1981.
250 M. S. BADER ET AL.

115. Moya B, Barcelo IM, Cabot G, et al. In vitro and in vivo activities of carrying enterobacteriaceae isolated as part of the international
β-lactams in combination with the novel β-lactam enhancers zide- network for optimal resistance monitoring (INFORM) global surveil-
bactam and WCK 5153 against multidrug-resistant metallo-β- lance program from 2012 to 2015. Antimicrob Agents Chemother.
lactamase-producing klebsiella pneumoniae. Antimicrob Agents 2018;62(12):e00592–18.
Chemother. 2019;63(5):e00128–19. 119. Alexandre K. Re v́ eillon-Istin M, Fabre R, et al. Temocillin against
116. Mushtaq S, Vickers A, Woodford N, et al. Activity of nacubactam Enterobacteriaceae isolates from community-acquired urinary tract
(RG6080/OP0595) combinations against MBL-producing infections: low rate of resistance and good accuracy of routine
Enterobacteriaceae. J Antimicrob Chemother. 2019;74(4):953–960. susceptibility testing methods. J Antimicrob Chemother.
117. Zhang W, Guo Y, Li J, et al. In vitro and in vivo bactericidal activity 2018;73:1848–1853.
of ceftazidime-avibactam against Carbapenemase-producing 120. Balakrishnan I, Awad-El-Kariem FM, Aali A, et al. Temocillin use in
Klebsiella pneumoniae. Antimicrob Resist Infect Control. England: clinical and microbiological efficacies in infections caused
2018;7:142. by extended-spectrum and/or derepressed AmpC b-lactamase-
118. Kazmierczak KM, Bradford PA, Stone GG, et al. In vitro activity of producing Enterobacteriaceae. J Antimicrob Chemother.
ceftazidime-avibactam and aztreonam-avibactam against OXA-48- 2011;66:2628–2631.

You might also like