Você está na página 1de 12

The n e w e ng l a n d j o u r na l of m e dic i n e

Original Article

Once-Daily Plazomicin for Complicated


Urinary Tract Infections
Florian M.E. Wagenlehner, M.D., Daniel J. Cloutier, Pharm.D.,
Allison S. Komirenko, Pharm.D., Deborah S. Cebrik, M.S., M.P.H.,
Kevin M. Krause, M.B.A., Tiffany R. Keepers, Ph.D., Lynn E. Connolly, M.D., Ph.D.,
Loren G. Miller, M.D., M.P.H., Ian Friedland, M.D., and Jamie P. Dwyer, M.D.,
for the EPIC Study Group*​​

A BS T R AC T

BACKGROUND
The increasing multidrug resistance among gram-negative uropathogens necessitates From the Justus Liebig University, Gies-
new treatments for serious infections. Plazomicin is an aminoglycoside with bactericidal sen, Germany (F.M.E.W.); Achaogen, South
San Francisco (D.J.C., A.S.K., D.S.C.,
activity against multidrug-resistant (including carbapenem-resistant) Enterobacteriaceae. K.M.K., T.R.K., L.E.C., I.F.), the David
METHODS ­Geffen School of Medicine, University of
California Los Angeles (UCLA), Los An-
We randomly assigned 609 patients with complicated urinary tract infections (UTIs), geles (L.G.M.), and Los Angeles Biomed-
including acute pyelonephritis, in a 1:1 ratio to receive intravenous plazomicin (15 mg ical Research Institute at Harbor–UCLA
per kilogram of body weight once daily) or meropenem (1 g every 8 hours), with op- Medical Center, Torrance (L.G.M.) — all
in California; and Vanderbilt University
tional oral step-down therapy after at least 4 days of intravenous therapy, for a total of Medical Center, Nashville (J.P.D.). Address
7 to 10 days of therapy. The primary objective was to show the noninferiority of plazo- reprint requests to Dr. Wagenlehner at the
micin to meropenem in the treatment of complicated UTIs, including acute pyelone- Clinic for Urology, Pediatric Urology and
Andrology, Justus Liebig University Giessen,
phritis, with a noninferiority margin of 15 percentage points. The primary end points Rudolf-Buchheim Str. 7, 35392 Giessen,
were composite cure (clinical cure and microbiologic eradication) at day 5 and at the Germany, or at ­f lorian​.­wagenlehner@​
test-of-cure visit (15 to 19 days after initiation of therapy) in the microbiologic modified ­chiru​.­med​.­uni-giessen​.­de.

intention-to-treat population. * A complete list of the principal investi-


gators in the EPIC Study is provided in
RESULTS the Supplementary Appendix, available
Plazomicin was noninferior to meropenem with respect to the primary efficacy end at NEJM.org.
points. At day 5, composite cure was observed in 88.0% of the patients (168 of 191 N Engl J Med 2019;380:729-40.
patients) in the plazomicin group and in 91.4% (180 of 197 patients) in the meropenem DOI: 10.1056/NEJMoa1801467
Copyright © 2019 Massachusetts Medical Society.
group (difference, –3.4 percentage points; 95% confidence interval [CI], –10.0 to 3.1).
At the test-of-cure visit, composite cure was observed in 81.7% (156 of 191 patients) and
70.1% (138 of 197 patients), respectively (difference, 11.6 percentage points; 95% CI, 2.7
to 20.3). At the test-of-cure visit, a higher percentage of patients in the plazomicin group
than in the meropenem group were found to have microbiologic eradication, including
eradication of Enterobacteriaceae that were not susceptible to aminoglycosides (78.8%
vs. 68.6%) and Enterobacteriaceae that produce extended-spectrum β-lactamases
(82.4% vs. 75.0%). At late follow-up (24 to 32 days after initiation of therapy), fewer
patients in the plazomicin group than in the meropenem group had microbiologic re-
currence (3.7% vs. 8.1%) or clinical relapse (1.6% vs. 7.1%). Increases in serum creati-
nine levels of 0.5 mg or more per deciliter (≥40 μmol per liter) above baseline occurred
in 7.0% of patients in the plazomicin group and in 4.0% in the meropenem group.
CONCLUSIONS
Once-daily plazomicin was noninferior to meropenem for the treatment of complicated
UTIs and acute pyelonephritis caused by Enterobacteriaceae, including multidrug-resis-
tant strains. (Funded by Achaogen and the Biomedical Advanced Research and Develop-
ment Authority; EPIC ClinicalTrials.gov number, NCT02486627.)

n engl j med 380;8 nejm.org  February 21, 2019 729


The New England Journal of Medicine
Downloaded from nejm.org at Midwestern University on February 20, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

U
rinary tract infections (UTIs) are Plazomicin is an aminoglycoside that is engi-
common bacterial infections that are as- neered to evade modification by aminoglycoside-
sociated with substantial morbidity, mor- modifying enzymes, and it maintains activity in
tality, and economic burden.1 Complicated UTIs the presence of most mechanisms that lead to
are UTIs in patients who have underlying condi- resistance in Enterobacteriaceae, including mu-
tions, such as anatomical abnormalities, or risk tations in sites targeted by fluoroquinolones and
factors, such as indwelling urinary catheters. the production of aminoglycoside-modifying en-
Acute pyelonephritis is an infection of the renal zymes, extended-spectrum β-lactamases, and car­
pelvis and kidney.2-4 In the period from 2006 bapenemases.12-17 A phase 3 trial has assessed
through 2009, a total of 10.8 million patients the efficacy and safety of plazomicin in patients
presented to U.S. emergency departments with a with serious infections due to carbapenem-resis-
primary diagnosis of UTI. Of these, 17% were tant Enterobacteriaceae.18 The current trial as-
admitted for further disease management, and sessed the efficacy and safety of plazomicin as
14% had pyelonephritis.5 compared with meropenem in patients with com-
Enterobacteriaceae are the most common plicated UTIs, including acute pyelonephritis.
causes of complicated UTIs, and multidrug re-
sistance within this family is a global concern.6 Me thods
Patients with multidrug-resistant UTIs are three
times as likely to receive inappropriate empirical Trial Design and Oversight
antibiotic therapy as patients who do not have The Evaluating Plazomicin in cUTI (EPIC) trial
multidrug-resistant infections. They also have was a multicenter, multinational, randomized,
longer hospital stays, incur higher hospital double-blind, phase 3 trial that was designed in
costs, and have a higher risk of septic shock and accordance with FDA guidelines4,19 and was con-
death.7,8 ducted from January through September 2016.
The previous mainstays of empirical therapy The primary objective of the trial was to show
for complicated UTIs, such as fluoroquinolones the noninferiority of plazomicin to meropenem
and cephalosporins, are not widely recommended in the treatment of complicated urinary tract
because of concerns about resistance.3 Carbapen- infections, including acute pyelonephritis. Patients
ems, which have been traditionally reserved for were enrolled at 68 sites in North America and
the treatment of multidrug-resistant infections, Europe. The institutional review board or ethics
are increasingly being used to treat complicated committee at each site approved the final proto-
UTIs.9 The incidence of multidrug resistance has col, and all patients or their representatives
continued to increase — and now includes car- provided written informed consent. An indepen-
bapenem resistance; therefore, alternative treat- dent data and safety monitoring committee re-
ment options are needed.10,11 viewed interim trial data. Full details of the trial
Aminoglycosides are an alternative treatment design are provided in the protocol and statisti-
for complicated UTIs that are caused by multidrug- cal analysis plan, available with the full text of
resistant organisms, such as extended-spectrum this article at NEJM.org.
β-lactamase–producing Enterobacteriaceae or car­ The trial was designed by Achaogen and the
bapenem-resistant Enterobacteriaceae.6 However, first author. Data were collected by Achaogen
bacterial strains that are resistant to cephalospo- and analyzed in collaboration with three of the
rins and carbapenems commonly produce amino- academic authors. A data confidentiality agree-
glycoside-modifying enzymes, which result in ment was in place between Achaogen and the
resistance to many aminoglycosides.12,13 U.S. sur- investigators. All the authors vouch for the ac-
veillance data from 2014–2015 show that ap- curacy and completeness of the data and for the
proximately 80% of the carbapenem-resistant fidelity of the trial to the protocol. The first
Enterobacteriaceae isolates tested positive for one draft of the manuscript was written by a medical
or more aminoglycoside-modifying enzyme genes. writer from a contract research organization
Amikacin, gentamicin, and tobramycin had re- who was paid by Achaogen, and all the authors
duced activity against these isolates, with only contributed to subsequent drafts, provided final
59.7% of these isolates susceptible to amikacin, approval of the manuscript, and made the deci-
49.4% to gentamicin, and 0% to tobramycin.13 sion to submit the manuscript for publication.

730 n engl j med 380;8 nejm.org  February 21, 2019

The New England Journal of Medicine


Downloaded from nejm.org at Midwestern University on February 20, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
Plazomicin for Complicated Urinary Tr act Infections

Eligibility Criteria down therapy are provided in the Supplementary


Eligible patients were 18 years of age or older Appendix.
and had a creatinine clearance of more than 30 ml
per minute, pyuria, and clinical symptoms of a Analysis Populations, End Points,
complicated UTI or acute pyelonephritis that and Assessments
would require at least 4 days of intravenous The intention-to-treat population included all
therapy with an antibiotic agent. Complicated randomly assigned patients. The safety popula-
UTIs were indicated by at least two signs or tion and the modified intention-to-treat popula-
symptoms (chills, rigors, or fever [temperature tion included all patients in the intention-to-treat
>38°C]; dysuria or urinary frequency or urgency; population who received any amount of a trial
lower abdominal or pelvic pain; or nausea or drug. The microbiologic modified intention-to-
vomiting) and at least one complicating factor treat population included patients in the modi-
(a history of urinary retention [in male patients], fied intention-to-treat population who had at
current indwelling urinary catheter, obstructive least one qualifying baseline pathogen (≥105
uropathy, or any functional or anatomical abnor- colony-forming units [CFU] per milliliter) that
mality). Acute pyelonephritis was indicated by at was susceptible to both meropenem (minimum
least two of the following symptoms: chills, inhibitory concentration [MIC] of ≤1 μg per mil-
rigors, or fever; flank pain; tenderness in the liliter21) and plazomicin (MIC of ≤4 μg per milli-
costovertebral angle; and nausea or vomiting. liter). We also evaluated a population that in-
Details of eligibility criteria are provided in Ta- cluded all protocol-adherent patients in the
ble S1 in the Supplementary Appendix, available microbiologic modified intention-to-treat popu-
at NEJM.org. lation who had an available urine culture at day
5, at the test-of-cure visit, or both (microbiologic
Randomization and Treatment per-protocol population). Isolate and susceptibil-
Patients were randomly assigned in a 1:1 ratio to ity testing were conducted at a central laboratory
receive plazomicin or meropenem. Randomiza- (JMI Laboratories).
tion was performed by the site pharmacist or The primary efficacy end points were com-
designated staff member according to a prespeci- posite cure (clinical cure and microbiologic eradi-
fied randomization schedule. Block randomiza- cation) at day 5 and at the test-of-cure visit (15
tion was automated through an interactive Web- to 19 days after initiation of intravenous therapy
based or voice-response system and was stratified with the assigned trial drug) in the microbiologic
according to region and baseline diagnosis modified intention-to-treat population. Clinical
(complicated UTI or acute pyelonephritis). The cure was defined as a reduction in severity (at
sponsor, investigators, trial staff participating in day 5 and at the end of intravenous therapy) or
patient care or clinical evaluations, and patients complete resolution (at the test-of-cure visit) of all
were unaware of the group assignments. Desig- core symptoms with no new symptoms or as a
nated staff members who prepared the trial return to the patient’s status before development
drugs had knowledge of the group assignments. of the UTI, with no use of nontrial antibiotics
Patients were assigned to receive plazomicin for the current complicated UTI. Microbiologic
(15 mg per kilogram of body weight once daily) eradication was defined as a reduction in the
or meropenem (1 g every 8 hours), administered baseline uropathogen from 105 CFU or more per
intravenously, with the option for oral step-down milliliter to less than 104 CFU per milliliter.
therapy after a minimum of 4 days of intrave- Additional end points assessed in the micro-
nous therapy, for a total of 7 to 10 days of biologic modified intention-to-treat population
therapy. Levofloxacin (500 mg once daily) was were clinical cure and microbiologic eradication
the preferred oral agent, but other agents were at day 5, at the end of intravenous therapy
permitted if levofloxacin was contraindicated. (within 24 hours after the last dose of intrave-
Therapeutic drug monitoring was not imple- nous trial drug and before oral therapy), at the
mented. Doses of all trial drugs were adjusted test-of-cure visit, and during late follow-up (days
on the basis of creatinine clearance, which was 24 to 32); and microbiologic response at the test-
estimated with the use of the Cockcroft–Gault of-cure visit according to baseline pathogen. To
formula.20 Details on the criteria for oral step- evaluate consistency with the results in the over-

n engl j med 380;8 nejm.org  February 21, 2019 731


The New England Journal of Medicine
Downloaded from nejm.org at Midwestern University on February 20, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

all population, composite cure at the test-of-cure assessments are provided for descriptive pur-
visit was assessed in prespecified subgroups de- poses and have also not been corrected for mul-
fined according to geographic region (region 1 or tiple comparisons.
2), baseline diagnosis (complicated UTI or acute
pyelonephritis), presence of bacteremia, treatment R e sult s
received (intravenous only or intravenous plus
oral), presence of indwelling catheter (yes or no), Patients
and previous antibiotic use (yes or no) and in Of the 609 patients enrolled, 604 (99.2%) were
post hoc subgroups defined according to age included in the safety and modified intention-to-
(<65 or ≥65 years), renal function (creatinine treat populations and 388 (63.7%) were included
clearance ≤60 ml per minute or >60 ml per min- in the microbiologic modified intention-to-treat
ute), and sex. Definitions of efficacy end points population. The most common reason for exclu-
are provided in Table S2 in the Supplementary sion from the microbiologic modified intention-
Appendix. to-treat population was the absence of a qualify-
Safety analyses included assessments of ad- ing baseline uropathogen. Details regarding
verse events and laboratory values. Serum creati- patient disposition are provided in Figure S1 in
nine levels were assessed at baseline, daily during the Supplementary Appendix.
intravenous treatment, at the end of intravenous Baseline characteristics were balanced be-
treatment, at the test-of-cure visit, and at late tween the treatment groups (Table 1, and Table
follow-up. Decreased renal function was pre- S3 in the Supplementary Appendix). The mean
specified as an increase in the serum creatinine age was 59.4 years, and 72.2% of patients had
level of 0.5 mg or more per deciliter (≥40 μmol impaired renal function. There were more pa-
per liter) above baseline at any time during the tients with complicated UTIs than with acute
trial.22,23 pyelonephritis (58.2% vs. 41.8%), and 13.1% of
patients had an indwelling urinary catheter. The
Statistical Analysis mean duration of intravenous therapy was 5.5
We calculated that 394 patients would need to be days in each group; the mean duration of intrave-
enrolled for the trial to have at least 85% power nous plus oral therapy was 9.2 days in the plazo-
to show the noninferiority of plazomicin to micin group and 8.9 days in the meropenem
meropenem in the treatment of complicated UTIs group. Most patients (80.6% in the plazomicin
and acute pyelonephritis, with a noninferiority group and 76.6% in the meropenem group) re-
margin of 15 percentage points, at a one-sided ceived oral step-down therapy, which was primar-
significance level of 0.025 for each primary end ily levofloxacin. Of the 154 patients with a uro-
point, assuming that composite cure would oc- pathogen at baseline that was not susceptible to
cur in 64.0% of patients at day 5 and in 73.2% levofloxacin, 59 received oral levofloxacin (27
at the test-of-cure visit in both treatment groups. patients in the plazomicin group and 32 in the
Patients whose assessments did not result in meropenem group). A total of 60% of the patients
definitive findings were considered to have had who had a uropathogen that was not susceptible
treatment failure with respect to all efficacy end to levofloxacin received the maximum 7 days of
points assessed in the microbiologic modified intravenous therapy. Common alternative oral
intention-to-treat population. Two-sided 95% antibiotics were trimethoprim–sulfamethoxazole,
confidence intervals for the observed differ- amoxicillin–clavulanate, and cefixime.
ences in composite cure rates (plazomicin mi- The most common uropathogen was Escherichia
nus meropenem) at day 5 and at the test-of-cure coli, followed by Klebsiella pneumoniae. Of the 382
visit were calculated with the use of the New- patients with Enterobacteriaceae in the micro-
combe method with continuity correction. A biologic modified intention-to-treat population,
determination of noninferiority required that 107 patients (28.0%) had uropathogens with an
the lower limits of the 95% confidence inter- extended-spectrum β-lactamase phenotype, 115
vals at day 5 and at the test-of-cure visit be (30.1%) had multidrug-resistant uropathogens,
greater than −15 percentage points (details are and 101 (26.4%) had uropathogens that were not
provided in the Supplementary Appendix).19 No susceptible to other aminoglycosides (amikacin,
corrections for multiple comparisons were per- gentamicin, or tobramycin). MICs for plazomi-
formed. The confidence intervals for the other cin and meropenem among baseline uropatho-

732 n engl j med 380;8 nejm.org  February 21, 2019

The New England Journal of Medicine


Downloaded from nejm.org at Midwestern University on February 20, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
Plazomicin for Complicated Urinary Tr act Infections

Table 1. Characteristics of the Patients at Baseline (Microbiologic Modified Intention-to-Treat Population).*

Characteristic Plazomicin (N = 191) Meropenem (N = 197)


Age — yr 58.8±18.0 60.0±17.9
Male sex — no. (%) 84 (44.0) 99 (50.3)
Race — no. (%)†
White 189 (99.0) 197 (100.0)
Black 1 (0.5) 0
Other 1 (0.5) 0
Geographic region — no. (%)‡
Region 1 4 (2.1) 2 (1.0)
Region 2 187 (97.9) 195 (99.0)
Body-mass index§ 26.7±5.1 27.1±5.1
Infection type — no. (%)
Complicated urinary tract infection 107 (56.0) 119 (60.4)
Acute pyelonephritis 84 (44.0) 78 (39.6)
Calculated creatinine clearance — no./total no. (%)¶
>90 ml/min 57/188 (30.3) 45/194 (23.2)
>60–90 ml/min 70/188 (37.2) 75/194 (38.7)
>30–60 ml/min 61/188 (32.4) 71/194 (36.6)
≤30 ml/min 0/188 3/194 (1.5)
Missing data 3/191 (1.6) 3/197 (1.5)
Urosepsis — no. (%)‖ 39 (20.4) 34 (17.3)
Bacteremia — no. (%)** 25 (13.1) 23 (11.7)
Enterobacteriaceae uropathogens — no./total no. (%)†† 189/191 (99.0) 193/197 (98.0)
Not susceptible to levofloxacin 71/189 (37.6) 83/193 (43.0)
Not susceptible to at least one aminoglycoside 51/189 (27.0) 50/193 (25.9)
Not susceptible to amikacin 1/189 (0.5) 1/193 (0.5)
Not susceptible to gentamicin 37/189 (19.6) 41/193 (21.2)
Not susceptible to tobramycin 43/189 (22.8) 44/193 (22.8)
Not susceptible to any two aminoglycosides 29/189 (15.3) 35/193 (18.1)
Not susceptible to all three aminoglycosides 1/189 (0.5) 1/193 (0.5)
ESBL phenotype‡‡ 50/189 (26.5) 57/193 (29.5)
Not susceptible to carbapenem§§ 9/189 (4.8) 6/193 (3.1)
Multidrug resistant¶¶ 55/189 (29.1) 60/193 (31.1)

* Plus–minus values are means ±SD. The microbiologic modified intention-to-treat population included all patients
who underwent randomization, received at least one dose of the assigned trial drug, and had at least one qualifying
baseline pathogen that was susceptible to meropenem and that had a minimum inhibitory concentration (MIC) for
plazomicin of 4 μg per milliliter or less. Percentages may not total 100 because of rounding.
† Race was reported by the participants.
‡ Region 1 included the United States, Mexico, and Spain. Region 2 included Bulgaria, the Czech Republic, Estonia,
Georgia, Hungary, Latvia, Poland, Romania, Russia, Serbia, and Ukraine.
§ The body-mass index is the weight in kilograms divided by the square of the height in meters.
¶ Creatinine clearance was calculated with the use of the ideal body weight if the total body weight exceeded the ideal
body weight by 25% or more.24 Patients were eligible for inclusion in the trial if they had a serum creatinine level of
more than 30 ml per minute as calculated at the local laboratory (Table S1 in the Supplementary Appendix). However,
three patients in the meropenem group were subsequently confirmed to have a creatinine clearance at screening of
30 ml or less per minute as calculated at a central laboratory.
‖ Urosepsis was determined on the basis of criteria for systemic inflammatory response syndrome.25
** Baseline blood cultures were required in patients who had acute pyelonephritis or urosepsis or whose baseline urine
culture was obtained through a preexisting indwelling catheter.
†† Patients with more than one uropathogen in the same category were counted only once in that category.
Susceptibility was determined on the basis of breakpoints established by the Clinical and Laboratory Standards
Institute (see the Supplementary Appendix).21
‡‡ Extended-spectrum β-lactamase (ESBL) phenotype was defined as pathogens that had an MIC for ceftazidime, aztre-
onam, or ceftriaxone of at least 2 μg per milliliter, as determined at a central laboratory.
§§ The pathogens were not susceptible to imipenem or doripenem but were susceptible to meropenem.
¶¶ Multidrug resistance was defined as resistance to at least one antibiotic from at least three different classes.26

n engl j med 380;8 nejm.org  February 21, 2019 733


The New England Journal of Medicine
Downloaded from nejm.org at Midwestern University on February 20, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Table 2. Primary and Additional Efficacy End Points (Microbiologic Modified Intention-to-Treat Population).*

Plazomicin Meropenem Difference


Time of Assessment and End Point (N = 191) (N = 197) (95% CI)†
number (percent) percentage points
Day 5
Primary end point: composite cure at day 5 168 (88.0) 180 (91.4) −3.4 (−10.0 to 3.1)
Clinical cure 171 (89.5) 182 (92.4) −2.9 (−9.1 to 3.3)
Microbiologic eradication 188 (98.4) 193 (98.0) 0.5 (−3.1 to 4.1)
End of intravenous therapy
Composite cure 179 (93.7) 187 (94.9) −1.2 (−6.5 to 4.0)
Clinical cure 184 (96.3) 190 (96.4) −0.1 (−4.6 to 4.3)
Microbiologic eradication 186 (97.4) 192 (97.5) −0.1 (−4.1 to 3.9)
Test-of-cure visit
Primary end point: composite cure at 156 (81.7) 138 (70.1) 11.6 (2.7 to 20.3)
15 to 19 days after start of therapy
Clinical cure 170 (89.0) 178 (90.4) −1.4 (−7.9 to 5.2)
Microbiologic eradication 171 (89.5) 147 (74.6) 14.9 (7.0 to 22.7)
Late follow-up‡
Composite cure 147 (77.0) 119 (60.4) 16.6 (7.0 to 25.7)
Sustained clinical cure§ 169 (88.5) 168 (85.3) 3.2 (−4.0 to 10.3)
Sustained eradication¶ 161 (84.3) 128 (65.0) 19.3 (10.4 to 27.9)
Clinical relapse 3 (1.6) 14 (7.1) Not calculated
Microbiologic recurrence‖ 7 (3.7) 16 (8.1) Not calculated

* Composite cure was defined as clinical cure and microbiologic eradication. Clinical cure was defined as a reduction in
severity (at day 5 and at the end of intravenous therapy) or complete resolution (at the test-of-cure visit) of all core
symptoms with no new symptoms or as a return to the patient’s status before development of the urinary tract infec-
tion (UTI). Microbiologic eradication was defined as a reduction in the baseline uropathogen from 105 colony-forming
units (CFU) or more per milliliter to less than 104 CFU per milliliter.
† Confidence intervals were calculated with the use of the Newcombe method with continuity correction and were not
corrected for multiple comparisons.
‡ Late follow-up was conducted 24 to 32 days after initiation of intravenous therapy.
§ Patients were considered to have sustained clinical cure if they had not had clinical failure at the test-of-cure visit and if
they had complete resolution or a return to their status before development of the complicated UTI, with no new symp-
toms at the last follow-up visit.
¶ Patients were considered to have sustained eradication if a urine culture obtained at the last follow-up visit showed that
the baseline uropathogen or uropathogens had been reduced to less than 104 CFU per milliliter after showing eradica-
tion at the test-of-cure visit (or if no urine culture was obtained at the last follow-up visit and the patients met criteria
for sustained clinical cure).
‖ Patients were considered to have microbiologic recurrence if they had a urine culture obtained at any time after docu-
mented eradication at the test-of-cure visit, up to and including the last follow-up visit, that showed an increase in the
baseline pathogen or pathogens to 104 CFU or more per milliliter.

gens are provided in Table S4 in the Supplemen- –10.0 to 3.1). At the test-of-cure visit, composite
tary Appendix. cure was observed in 81.7% (156 of 191 patients)
and 70.1% (138 of 197 patients), respectively
End Points (difference, 11.6 percentage points; 95% CI, 2.7
Plazomicin was noninferior to meropenem with to 20.3). The lower limit of the 95% confidence
respect to the primary efficacy end points (Ta- interval for the between-group difference in the
ble 2). At day 5, composite cure was observed in rate of composite cure at the test-of-cure visit
88.0% of the patients (168 of 191 patients) in the exceeded zero. The directionality of the between-
plazomicin group and in 91.4% (180 of 197 pa- group difference in the rate of composite cure at
tients) in the meropenem group (difference, –3.4 the test-of-cure visits in subgroups was consis-
percentage points; 95% confidence interval [CI], tent with that in the overall population (Fig. 1).

734 n engl j med 380;8 nejm.org  February 21, 2019

The New England Journal of Medicine


Downloaded from nejm.org at Midwestern University on February 20, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
Plazomicin for Complicated Urinary Tr act Infections

Subgroup Meropenem Plazomicin Percentage-Point Difference (95% CI)


no. of patients with composite cure/no. of patients (%)
Overall 138/197 (70.1) 156/191 (81.7) 11.6 (2.7 to 20.3)
Diagnosis at baseline
Complicated UTI 82/119 (68.9) 84/107 (78.5) 9.6 (–2.6 to 21.3)
Acute pyelonephritis 56/78 (71.8) 72/84 (85.7) 13.9 (0.4 to 27.1)
Presence of bacteremia 13/23 (56.5) 18/25 (72.0) 15.5 (–13.7 to 41.9)
Treatment received
Intravenous only 28/46 (60.9) 29/37 (78.4) 17.5 (–4.3 to 36.6)
Intravenous plus oral 110/151 (72.8) 127/154 (82.5) 9.6 (–0.2 to 19.3)
Catheter
Present 15/29 (51.7) 18/29 (62.1) 10.3 (–16.6 to 35.5)
Absent 123/168 (73.2) 138/162 (85.2) 12.0 (2.8 to 20.9)
Age
<65 yr 68/95 (71.6) 90/101 (89.1) 17.5 (5.7 to 29.0)
≥65 yr 70/102 (68.6) 66/90 (73.3) 4.7 (–8.9 to 17.9)
Sex
Male 68/99 (68.7) 65/84 (77.4) 8.7 (–5.1 to 21.7)
Female 70/98 (71.4) 91/107 (85.0) 13.6 (1.6 to 25.4)
Creatinine clearance
≤60 ml/min 49/74 (66.2) 43/61 (70.5) 4.3 (–12.5 to 20.3)
>60 ml/min 87/120 (72.5) 112/127 (88.2) 15.7 (5.2 to 25.9)
−20 −15 −10 −5 0 5 10 15 20 25 30 35 40 45

Meropenem Better Plazomicin Better

Figure 1. Composite Cure at the Test-of-Cure Visit, According to Patient Subgroups in the Microbiologic Modified Intention-to-Treat Population.
The percentage-point difference is for plazomicin minus meropenem; some values may differ from the expected value because of
rounding. All subgroups were prespecified except for age, creatinine clearance, and sex. The 95% confidence intervals have not been
corrected for multiple comparisons. Post hoc analyses used the Gail–Simon test for qualitative interaction in all subgroups (qualitative
interaction, 0.0; P = 0.5). The vertical red line indicates the noninferiority margin of 15 percentage points. Results for patients according
to geographic region are not reported, since 6 of 388 (1.5%) patients were enrolled in region 1 (United States, Mexico, and Spain) and
382 of 388 (98.5%) patients were enrolled in region 2 (Bulgaria, the Czech Republic, Estonia, Georgia, Hungary, Latvia, Poland, Romania,
Russia, Serbia, and Ukraine). Results for patients according to previous antibiotic use (yes or no) are not reported because of the small
number of patients who received previous antibiotic therapy (2 patients in the plazomicin group and no patients in the meropenem
group). UTI denotes urinary tract infection.

The between-group difference favoring plazomi- tion, a higher percentage of patients in the plazo-
cin that was observed in the subgroup that re- micin group than in the meropenem group were
ceived only intravenous treatment was greater found to have microbiologic eradication of En-
than that observed in the subgroup that received terobacteriaceae that were not susceptible to other
intravenous plus oral treatment (percentage-point aminoglycosides (78.8% vs. 68.6%) (Table 3).
difference, 17.5 vs. 9.6), which suggests that oral At late follow-up, the rate of composite cure
therapy did not account for the between-group remained higher in the plazomicin group than
difference observed at the test-of-cure visit. in the meropenem group (Table 2). Clinical re-
Rates of microbiologic eradication were simi- lapse at late follow-up occurred in 1.6% of the
lar in the two treatment groups at day 5 and at patients in the plazomicin group and in 7.1% in
the end of intravenous therapy and were higher the meropenem group, and microbiologic recur-
in the plazomicin group than in the meropenem rence occurred in 3.7% and 8.1%, respectively.
group at the test-of-cure visit and at late follow- Most clinical relapses in the meropenem group
up (Table 2). Among patients with infections occurred in patients who had asymptomatic bac-
caused by extended-spectrum β-lactamase–pro- teriuria (microbiologic persistence and clinical
ducing Enterobacteriaceae, microbiologic eradi- cure) at the test-of-cure visit. Tables S5 and S6 in
cation at the test-of-cure visit was observed in the Supplementary Appendix show the outcomes
82.4% of the patients in the plazomicin group for the modified intention-to-treat population
and in 75.0% in the meropenem group. In addi- and the microbiologic per-protocol population.

n engl j med 380;8 nejm.org  February 21, 2019 735


The New England Journal of Medicine
Downloaded from nejm.org at Midwestern University on February 20, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Safety Discussion
The most frequent adverse events in the plazo-
micin group were diarrhea, hypertension, head- In the EPIC trial, plazomicin was noninferior to
ache, nausea, vomiting, and hypotension (Ta- meropenem in the treatment of patients with
ble 4). Adverse events associated with a decline complicated UTIs, including acute pyelonephri-
in renal function occurred in 11 of 303 patients tis, with higher rates of microbiologic eradica-
(3.6%) in the plazomicin group and in 4 of 301 tion and composite cure observed at the test-of-
patients (1.3%) in the meropenem group. Poten- cure visit in the plazomicin group than in the
tially ototoxic events were identified in 2 pa- meropenem group. The lower incidence of mi-
tients (1 in each group). Serious adverse events crobiologic recurrence and clinical relapse in
were reported in 1.7% of patients in each treat- the plazomicin group than in the meropenem
ment group (Table S8 in the Supplementary Ap- group at late follow-up suggests that the great-
pendix). er microbiologic eradication with plazomicin
During the trial, 21 of 300 patients (7.0%) in has additional clinical benefit for patients with
the plazomicin group and 12 of 297 patients complicated UTIs, including acute pyelone­
(4.0%) in the meropenem group had an increase phritis.
in serum creatinine level of 0.5 mg or more per Although the propensity for complicated UTIs
deciliter above the baseline level (Table 4, and to recur — necessitating frequent retreatment
Table S9 in the Supplementary Appendix). Of with antibiotics and increasing the potential for
these, the increase occurred after completion of the development of resistance — is well known,3
intravenous therapy in 10 of 300 patients (3.3%) comprehensive evaluations of the risk of recur-
in the plazomicin group and in 3 of 297 patients rence of complicated UTIs and relapse are lack-
(1.0%) in the meropenem group; of the 10 pa- ing. This trial assessed the sustained effects of
tients in the plazomicin group who had serum antibacterial therapy in patients with complicated
creatinine increases after completion of intrave- UTIs and showed that asymptomatic bacteriuria
nous therapy, 9 had had moderate renal impair- was more common after treatment with merope-
ment at baseline. In general, increases were less nem than after treatment with plazomicin and
than 1.0 mg per deciliter (<80 μmol per liter) was associated with subsequent clinical relapse.
from baseline and returned to less than 0.5 mg Asymptomatic bacteriuria is common among
per deciliter from the baseline value by the last patients with complicated UTIs after completion
follow-up. Among patients in the plazomicin of therapy,27 and treatment is generally not rec-
group, risk factors for an increase in serum cre- ommended for most patients because of the
atinine level of 0.5 mg or more per deciliter were perceived low risk of clinical consequences.3,28,29
moderate renal impairment (14 of 21 patients) However, findings from this trial suggest that
and receipt of plazomicin therapy for more than additional data should be generated to guide ap-
5 days (12 of 21 patients); 8 of 21 patients had proaches for managing asymptomatic bacteriuria
both risk factors. in patients at risk for recurrent complicated UTIs.
A total of 12 patients discontinued treatment Unlike recent phase 3 trials involving patients
because of adverse events (6 patients [2%] in with complicated UTIs,30-32 the EPIC trial excluded
each group) (Table S10 in the Supplementary Ap- patients from the microbiologic modified inten-
pendix). In both groups, these adverse events tion-to-treat population who had pathogens that
were most commonly associated with renal were resistant to the comparator; thus, the results
function; in accordance with the protocol, pa- were not biased toward plazomicin. Meropenem
tients with creatinine clearance of 30 ml or less was a reliably active comparator to evaluate the
per minute were to discontinue the trial drug. efficacy of plazomicin for multidrug-resistant
No deaths related to a trial drug were ob- pathogens. The higher observed rate of composite
served. One patient who received a single dose of cure at the test-of-cure visit in the plazomicin
plazomicin died on day 18 from metastatic uter- group, including in patients with important re-
ine cancer that had been diagnosed 48 hours sistant pathogens, such as extended-spectrum
after the patient underwent randomization. No β-lactamase–producing Enterobacteriaceae and
deaths occurred in the meropenem group. Enterobacteriaceae that are not susceptible to

736 n engl j med 380;8 nejm.org  February 21, 2019

The New England Journal of Medicine


Downloaded from nejm.org at Midwestern University on February 20, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
Plazomicin for Complicated Urinary Tr act Infections

Table 3. Microbiologic Eradication at the Test-of-Cure Visit According to Pathogen (Microbiologic Modified Intention-to-Treat Population).*

Baseline Uropathogen Plazomicin (N = 191) Meropenem (N = 197) Difference (95% CI)†

no. of uropathogens eradicated/


total no. of uropathogens percentage points
Enterobacteriaceae 177/198 (89.4) 157/208 (75.5) 13.9 (6.2 to 21.5)
Not susceptible to at least one aminoglycoside 41/52 (78.8) 35/51 (68.6) 10.2 (−8.1 to 27.8)
Not susceptible to amikacin 1/1 (100.0) 0/1
Not susceptible to gentamicin 27/37 (73.0) 31/42 (73.8) −0.8 (−21.8 to 19.7)
Not susceptible to tobramycin 36/44 (81.8) 32/45 (71.1) 10.7 (−8.6 to 29.0)
Not susceptible to any two aminoglycosides 22/29 (75.9) 28/36 (77.8) −1.9 (−24.8 to 19.8)
Not susceptible to all three aminoglycosides 1/1 (100.0) 0/1
Not susceptible to carbapenem‡ 7/9 (77.8) 5/6 (83.3) −5.6 (−46.3 to 44.7)
ESBL phenotype§ 42/51 (82.4) 45/60 (75.0) 7.4 (−9.6 to 23.1)
Multidrug resistant¶ 44/57 (77.2) 45/64 (70.3) 6.9 (−10.1 to 23.0)
Escherichia coli 120/128 (93.8) 106/142 (74.6) 19.1 (10.0 to 27.9)
Not susceptible to at least one aminoglycoside 20/23 (87.0) 16/26 (61.5) 25.4 (−2.4 to 48.3)
Not susceptible to amikacin 0/0 0/0
Not susceptible to gentamicin 10/12 (83.3) 14/19 (73.7) 9.6 (–26.6 to 38.3)
Not susceptible to tobramycin 16/18 (88.9) 15/24 (62.5) 26.4 (–4.4 to 50.0)
Not susceptible to any two aminoglycosides 6/7 (85.7) 13/17 (76.5) 9.2 (–37.2 to 39.2)
Not susceptible to all three aminoglycosides 0/0 0/0
Not susceptible to carbapenem‡ 0/0 0/0
ESBL phenotype§ 18/20 (90.0) 19/28 (67.9) 22.1 (−5.7 to 44.0)
Multidrug resistant¶ 19/23 (82.6) 21/33 (63.6) 19.0 (−8.0 to 40.8)
Klebsiella pneumoniae 27/33 (81.8) 32/43 (74.4) 7.4 (−13.9 to 26.5)
Not susceptible to at least one aminoglycoside 14/18 (77.8) 15/20 (75.0) 2.8 (−27.3 to 31.4)
Not susceptible to amikacin 1/1 (100.0) 0/1
Not susceptible to gentamicin 12/16 (75.0) 13/18 (72.2) 2.8 (−29.7 to 33.5)
Not susceptible to tobramycin 14/18 (77.8) 14/18 (77.8) 0.0 (−29.8 to 29.8)
Not susceptible to any two aminoglycosides 12/16 (75.0) 12/16 (75.0) 0.0 (−32.2 to 32.2)
Not susceptible to all three aminoglycosides 1/1 (100.0) 0/1
Not susceptible to carbapenem‡ 0/0 1/1 (100.0)
ESBL phenotype§ 15/20 (75.0) 20/26 (76.9) −1.9 (−29.7 to 24.1)
Multidrug resistant¶ 15/20 (75.0) 18/24 (75.0) 0 (−28.3 to 26.9)
Proteus mirabilis 9/11 (81.8) 4/7 (57.1) 24.7 (−21.4 to 64.5)
Enterobacter cloacae 13/16 (81.3) 3/3 (100.0) −18.8 (−46.3 to 51.6)

* Patients may have had more than one uropathogen at baseline.


† Confidence intervals have not been corrected for multiple comparisons. Confidence intervals were calculated with the use of the Newcombe
method with continuity correction and were calculated only if the total number of uropathogens across both treatment groups was 10 or
more. Susceptibility was determined on the basis of breakpoints established by the Clinical and Laboratory Standards Institute (see the
Supplementary Appendix).21
‡ The pathogens were not susceptible to imipenem or doripenem but were susceptible to meropenem.
§ ESBL phenotype was defined as pathogens that had an MIC for ceftazidime, aztreonam, or ceftriaxone of at least 2 μg per milliliter, as deter-
mined by a central laboratory.
¶ Multidrug resistance was defined as a resistance to at least one antibiotic from at least three different classes.26

n engl j med 380;8 nejm.org  February 21, 2019 737


The New England Journal of Medicine
Downloaded from nejm.org at Midwestern University on February 20, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
The n e w e ng l a n d j o u r na l of m e dic i n e

Table 4. Safety Analysis (Safety Population).*

Variable Plazomicin (N = 303) Meropenem (N = 301)


Patients with any adverse event — no. (%) 59 (19.5) 65 (21.6)
Adverse events reported in ≥1% of patients in the plazomicin group
— no. (%)
Diarrhea 7 (2.3) 5 (1.7)
Hypertension 7 (2.3) 7 (2.3)
Headache 4 (1.3) 9 (3.0)
Nausea 4 (1.3) 4 (1.3)
Vomiting 4 (1.3) 3 (1.0)
Hypotension 3 (1.0) 2 (0.7)
Adverse events related to renal function — no. (%)† 11 (3.6) 4 (1.3)
Adverse events related to cochlear or vestibular function — no. (%)‡ 1 (0.3) 1 (0.3)
Patients with any serious adverse event — no. (%)§ 5 (1.7) 5 (1.7)
Increase of ≥0.5 mg/dl in serum creatinine level — no./total no. (%)
Any time during the trial¶ 21/300 (7.0) 12/297 (4.0)
Onset during intravenous therapy‖ 11/300 (3.7) 9/297 (3.0)
Full recovery at end of intravenous therapy** 6/11 (54.5) 4/9 (44.4)
Full recovery at last follow-up visit** 9/11 (81.8) 9/9 (100.0)
Onset after end of intravenous therapy 10/300 (3.3) 3/297 (1.0)

* The safety population included all randomly assigned patients who received any amount of a trial drug. A summary
of patients who had uropathogens that developed resistance to the trial drug is provided in Table S7 in the Supple­
mentary Appendix. To convert the values for creatinine to micromoles per liter, multiply by 88.4.
† This category includes an increase in blood creatinine level, a decrease in creatinine clearance, acute kidney injury,
­renal failure, renal impairment, and chronic kidney disease; the events were identified in a blinded review and were
classified according to the preferred terms in the Medical Dictionary for Regulatory Activities (MedDRA), version 19.0.
‡ This category includes hypoacusis and tinnitus; the events were identified in a blinded review and were classified ac-
cording to the preferred terms in MedDRA.
§ Serious adverse events in the plazomicin group were acute kidney injury, metastatic neoplasm, pneumonia, urinary cal-
culus, and urosepsis; serious adverse events in the meropenem group were acute pyelonephritis, Clostridium difficile coli-
tis, orchitis, pancreatitis, pyrexia, septic shock, and urinary tract infection (see Table S8 in the Supplementary Appendix).
¶ Any time during the trial includes the period during intravenous therapy, after the end of intravenous therapy, or both.
‖ Onset during intravenous therapy was defined as on or after the start of the first dose of intravenous trial drug through
the scheduled visit at the end of intravenous therapy.
** Full recovery was defined as a serum creatinine level less than 0.5 mg per deciliter above the baseline value.

aminoglycosides, was therefore unrelated to me- is known to be associated with aminoglycoside


ropenem resistance. toxicity,33 and with the general observation that
This trial assessed possible nephrotoxicity chronic renal dysfunction is a predictor of acute
through analyses of adverse events and changes renal dysfunction.34 Conversely, among patients
in serum creatinine level as markers of de- with normal renal function (creatinine clearance
creased renal function. Because published re- >90 ml per minute), the incidence of nephrotox-
ports of clinical trials of other aminoglycosides icity was lower in the plazomicin group than in
do not systematically include analyses of serum the meropenem group. Renal function was de-
creatinine levels after completion of treatment, termined on the basis of ideal body weight, and
an important finding in this trial was that a plazomicin doses on the basis of adjusted body
small number of patients in the plazomicin weight, which may have prevented obese pa-
group had increases in serum creatinine levels tients from receiving a dose that was too high
approximately 1 week after completion of ther- and may have helped to minimize the risk of
apy. Risk factors for decreased renal function new-onset renal dysfunction.
were consistent with drug accumulation, which The proportion of patients who received oral

738 n engl j med 380;8 nejm.org  February 21, 2019

The New England Journal of Medicine


Downloaded from nejm.org at Midwestern University on February 20, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
Plazomicin for Complicated Urinary Tr act Infections

step-down therapy with levofloxacin despite objective of showing the noninferiority of plazo-
having a baseline pathogen that was not suscep- micin to meropenem in composite cure at day 5
tible to levofloxacin was similar in the two treat- and at the test-of-cure visit. These findings sup-
ment groups, which therefore minimized the port the use of once-daily plazomicin in adult
possible effect on the test-of-cure results. The patients with complicated UTIs or acute pyelone-
majority of these patients received the intrave- phritis, including infections caused by extended-
nous trial drug for the prespecified maximum spectrum β-lactamase–producing Enterobac­
duration. Subgroup analyses showed that com- teriaceae and Enterobacteriaceae that are not
posite cure rates were higher among patients susceptible to other aminoglycosides.
who received meropenem plus oral step-down Supported by Achaogen and by federal funds from the Bio-
therapy than among patients who received me- medical Advanced Research and Development Authority, U.S.
ropenem alone. The rates of composite cure Department of Health and Human Services Office of the Assistant
Secretary for Preparedness and Response, under contract no.
were more consistent in the subgroups of pa- HHSO100201000046C. Editorial support was funded by Achaogen.
tients who received plazomicin, which suggests Dr. Wagenlehner reports receiving advisory board fees and
that the higher rate of composite cure with participating in a study for Achaogen, Bionorica, OM Pharma/
Vifor Pharma, and Shionogi, receiving advisory board fees from
plazomicin than with meropenem observed at AstraZeneca, Janssen, Leo Pharma, MerLion, MSD, Pfizer, Rosen-
the test-of-cure visit was not confounded by a Pharma, VenatoRx, and GlaxoSmithKline, and participating in
switch to oral therapy. a study for Enteris BioPharma, Helperby Therapeutics, and
Deutsches Zentrum für Infektionsforschung (Giessen-Marburg-
A limitation of the trial is that patients from Langen); Dr. Cloutier and Mr. Krause, being employed by and
countries outside Europe and patients of non- holding stock in Achaogen; Dr. Komirenko, Dr. Keepers, Ms.
white race were underrepresented, a limitation Cebrik, and Dr. Connolly, being formerly employed by and hold-
ing stock in Achaogen; Dr. Miller, receiving grant support and
that is similar to that in previous studies of advisory board fees from Achaogen, advisory board fees from
complicated UTIs.31,32,35 However, this factor most Tetraphase, and grant support from Cepheid, Merck, Abbott,
likely did not affect the results, because the Gilead Sciences, Genentech, AtoxBio, and Paratek; Dr. Fried-
land, being formerly employed by, holding stock in, and receiv-
pharmacokinetics of plazomicin and meropenem ing consulting fees from Achaogen; and Dr. Dwyer, receiving
are not expected to be affected by ethnic group. consulting fees from Achaogen, ContraFect, Theravance, and
In addition, the analysis in the microbiologic Spero Therapeutics. No other potential conflict of interest rele-
vant to this article was reported.
modified intention-to-treat population excluded Disclosure forms provided by the authors are available with
patients with pathogens resistant to the trial the full text of this article at NEJM.org.
drugs, so geographic differences in resistance A data sharing statement provided by the authors is available
with the full text of this article at NEJM.org.
rates also would not have affected the results. We thank Kate Bradford for editorial support with an earlier
In conclusion, the EPIC trial met the primary version of the manuscript.

References
1. Foxman B. Urinary tract infection syn- infections in the era of antimicrobial re- super-challenge. N Engl J Med 2009;​360:​
dromes: occurrence, recurrence, bacteriol- sistance. Postgrad Med 2017;​129:​242-58. 439-43.
ogy, risk factors, and disease burden. In- 7. Lee YC, Hsiao CY, Hung MC, et al. 11. Meletis G. Carbapenem resistance:
fect Dis Clin North Am 2014;​28:​1-13. Bacteremic urinary tract infection caused overview of the problem and future per-
2. Johnson JR, Russo TA. Acute pyelone- by multidrug-resistant Enterobacteriaceae spectives. Ther Adv Infect Dis 2016;​3:​15-21.
phritis in adults. N Engl J Med 2018;​378:​ are associated with severe sepsis at ad- 12. Castanheira M, Woosley LN, Doyle
48-59. mission: implication for empirical therapy. TB, et al. Aminoglycoside-resistance genes
3. Bonkat G, Pickard R, Bartoletti R, et al. Medicine (Baltimore) 2016;​95(20):​e3694. among 2014-2015 US carbapenem-resis-
EAU guidelines on urological infections. 8. Zilberberg MD, Nathanson BH, Sul- tant Enterobacteriaceae isolates and activ-
Arnhem, the Netherlands:​European As- ham K, Fan W, Shorr AF. Carbapenem ity of plazomicin against characterized
sociation of Urology (https://uroweb​.org/​ resistance, inappropriate empiric treat- isolates. Presented at the American Soci-
guideline/​urological​-­infections/​#1). ment and outcomes among patients hos- ety of Microbiology (ASM) Microbe 2017,
4. Guidance for industry — complicated pitalized with Enterobacteriaceae urinary New Orleans, June 1–5, 2017 (poster).
urinary tract infections:​developing drugs tract infection, pneumonia and sepsis. 13. Castanheira M, Doyle T, Woosley LN,
for treatment. Silver Spring, MD:​Food BMC Infect Dis 2017;​17:​279. et al. Plazomicin activity against Euro-
and Drug Administration, February 2015. 9. Golan Y. Empiric therapy for hospital- pean Enterobacteriaceae isolates carrying
5. Sammon JD, Sharma P, Rahbar H, et al. acquired, Gram-negative complicated intra- aminoglycoside-modifying enzymes and
Predictors of admission in patients pre- abdominal infection and complicated uri- 16S rRNA methylases. Presented at the
senting to the emergency department nary tract infections: a systematic literature Annual European Congress of Clinical
with urinary tract infection. World J Urol review of current and emerging treatment Microbiology and Infectious Diseases,
2014;​32:​813-9. options. BMC Infect Dis 2015;​15:​313. Vienna, April 22–25, 2017 (poster).
6. Bader MS, Loeb M, Brooks AA. An up- 10. Arias CA, Murray BE. Antibiotic-resis- 14. Aggen JB, Armstrong ES, Goldblum
date on the management of urinary tract tant bugs in the 21st century — a clinical AA, et al. Synthesis and spectrum of the

n engl j med 380;8 nejm.org  February 21, 2019 739


The New England Journal of Medicine
Downloaded from nejm.org at Midwestern University on February 20, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.
Plazomicin for Complicated Urinary Tr act Infections

neoglycoside ACHN-490. Antimicrob tional supplement. Wayne, PA:​Clinical 30. Ellis-Grosse E, Eckburg P, Skarinsky
Agents Chemother 2010;​54:​4636-42. and Laboratory Standards Institute, 2015. D, et al. Phenotypic antibiotic resistance
15. Castanheira M, Davis AP, Mendes RE, 22. Waikar SS, Bonventre JV. Creatinine in ZEUS:​a multi-center, randomized,
Serio AW, Krause KM, Flamm RK. In vitro kinetics and the definition of acute kid- double-blind phase 2/3 study of ZTI-01
activity of plazomicin against gram-nega- ney injury. J Am Soc Nephrol 2009;​20:​ versus piperacillin-tazobactam (P-T) in the
tive and gram-positive isolates collected 672-9. treatment of patients with complicated
from U.S. hospitals and comparative 23. Chertow GM, Burdick E, Honour M, urinary tract infections (cUTI) including
activities of aminoglycosides against
­ Bonventre JV, Bates DW. Acute kidney in- acute pyelonephritis (AP). Presented at:​
carbapenem-resistant Enterobacteriaceae jury, mortality, length of stay, and costs IDWeek, San Diego, CA, October 4–8, 2017
and isolates carrying carbapenemase genes. in hospitalized patients. J Am Soc Nephrol (poster).
Antimicrob Agents Chemother 2018;​62(8):​ 2005;​16:​3365-70. 31. Kaye KS, Bhowmick T, Metallidis S,
e00313-18. 24. Devine BJ. Gentamicin therapy. Drug et al. Effect of meropenem-vaborbactam
16. Castanheira M, Doyle TB, Serio AW, Intell Clin Pharm 1974;​8:​5. vs piperacillin-tazobactam on clinical cure
et al. In vitro activity of plazomicin and 25. Bone RC. Toward an epidemiology and or improvement and microbial eradica-
comparator agents against urinary tract natural history of SIRS (systemic inflam- tion in complicated urinary tract infec-
infection isolates from the United States matory response syndrome). JAMA 1992;​ tion: the TANGO I randomized clinical
and Europe. Presented at the American 268:​3452-5. trial. JAMA 2018;​319:​788-99.
Society of Microbiology (ASM) Microbe 26. Magiorakos AP, Srinivasan A, Carey 32. Wagenlehner FM, Umeh O, Steenber-
2017, New Orleans, June 1–5, 2017 (poster). RB, et al. Multidrug-resistant, extensively gen J, Yuan G, Darouiche RO. Ceftolo-
17. Thwaites M, Hall D, Shinabarger D, drug-resistant and pandrug-resistant bac- zane-tazobactam compared with levo-
et al. Evaluation of the bactericidal activity teria: an international expert proposal for floxacin in the treatment of complicated
of plazomicin and comparators against interim standard definitions for acquired urinary-tract infections, including pyelo-
multidrug-resistant Enterobacteriaceae. resistance. Clin Microbiol Infect 2012;​18:​ nephritis: a randomised, double-blind,
Antimicrob Agents Chemother 2018;​62(8):​ 268-81. phase 3 trial (ASPECT-cUTI). Lancet 2015;​
e00236-18. 27. Spivak ES, Burk M, Zhang R, et al. 385:​1949-56.
18. McKinnell JA, Dwyer JP, Talbot GH, Management of bacteriuria in Veterans 33. Mingeot-Leclercq MP, Tulkens PM.
et al. Plazomicin for infections caused by Affairs hospitals. Clin Infect Dis 2017;​65:​ Aminoglycosides: nephrotoxicity. Anti-
carbapenem-resistant Enterobacteriaceae. 910-7. microb Agents Chemother 1999;​43:​1003-
N Engl J Med 2019;​380:791-3. 28. Hooton TM, Bradley SF, Cardenas DD, 12.
19. Guidance for industry:​antibacterial et al. Diagnosis, prevention, and treatment 34. Hsu CY, Ordoñez JD, Chertow GM,
therapies for patients with an unmet of catheter-associated urinary tract infec- Fan D, McCulloch CE, Go AS. The risk of
medical need for the treatment of serious tion in adults: 2009 international clinical acute renal failure in patients with chronic
bacterial diseases. Silver Spring, MD:​Food practice guidelines from the Infectious kidney disease. Kidney Int 2008;​74:​101-7.
and Drug Administration, August 2017 Diseases Society of America. Clin Infect 35. Wagenlehner FM, Sobel JD, Newell P,
(https://www​.fda​.gov/​downloads/​Drugs/​ Dis 2010;​50:​625-63. et al. Ceftazidime-avibactam versus dori­
Guidances/​UCM359184​.pdf). 29. Köves B, Cai T, Veeratterapillay R, et al. penem for the treatment of complicated
20. Cockcroft DW, Gault MH. Prediction Benefits and harms of treatment of asymp- urinary tract infections, including acute
of creatinine clearance from serum creati- tomatic bacteriuria: a systematic review pyelonephritis: RECAPTURE, a phase 3
nine. Nephron 1976;​16:​31-41. and meta-analysis by the European Asso- randomized trial program. Clin Infect Dis
21. Performance standards for antimicro- ciation of Urology Urological Infection 2016;​63:​754-62.
bial susceptibility testing:​25th informa- Guidelines Panel. Eur Urol 2017;​72:​865-8. Copyright © 2019 Massachusetts Medical Society.

740 n engl j med 380;8 nejm.org  February 21, 2019

The New England Journal of Medicine


Downloaded from nejm.org at Midwestern University on February 20, 2019. For personal use only. No other uses without permission.
Copyright © 2019 Massachusetts Medical Society. All rights reserved.

Você também pode gostar