Download as docx, pdf, or txt
Download as docx, pdf, or txt
You are on page 1of 13

J Antimicrob Chemother

doi:10.1093/jac/dkaa183

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkaa183/5842233


Oral cephalosporin and b-lactamase inhibitor combinations for ESBL-
producing Enterobacteriaceae urinary tract infections
1,2 1,3 1,4 5,6
Adam G. Stewart , Patrick N. A. Harris , Andrew Henderson , Mark A. Schembri and David
1,2
L. Paterson *
1
Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane and Women’s Hospital
Campus, Brisbane, Australia; 2Department of Infectious Diseases, Royal Brisbane and Women’s Hospital, Brisbane,
Queensland, Australia; 3Department of Microbiology, Pathology Queensland, Royal Brisbane and Women’s Hospital, Brisbane,
Queensland, Australia; 4Infection Management Services, Princess Alexandra Hospital, Brisbane, Queensland, Australia; 5School
of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia; 6Australian Infectious
Diseases Research Centre, The University of Queensland, Brisbane, Queensland, Australia

*Corresponding author. E-mail: d.paterson1@uq.edu.au

ESBL-producing Enterobacteriaceae as uropathogens have given rise to a sizeable amount of global


morbidity. Community and hospital surveillance studies continue to report increasing proportions of these
organisms as causes of urinary tract infection (UTI). Due to limited treatment options and the presence of
cross-resistance amongst oral antibiotics of different classes, patients often require IV therapy, thereby
increasing healthcare costs and reducing the effectiveness of delivering healthcare. Oral cephalosporin
antibiotics are well known for their ability to achieve high urinary concentrations, in addition to achieving clinical
success for treatment of un-complicated UTI with a drug-susceptible pathogen. Novel cephalosporin/b-
lactamase inhibitor combinations have been developed and demonstrate good in vitro activity against ESBL-
producing isolates. A pooled analysis of in vitro activity of existing oral cephalosporin/clavulanate combinations
in ESBL-producing Enterobacteriaceae has shown MIC50s of 0.5–1, 0.125–1 and 0.25 mg/L for cefpodoxime,
ceftibuten and cefixime, respectively. A novel cyclic boronic acid b-lactamase inhibitor, QPX7728, was able to
produce MIC50 values of 0.5 and 0.06 mg/ L when paired with cefpodoxime and ceftibuten, respectively. Other
novel combinations, cefpodoxime/ETX0282 and ceftibuten/VNRX7145, have also demonstrated excellent
activity against ESBL producers. Clinical trials are now awaited.

Introduction not exhibit cross-class resistance with other classes of antibiotics

by Uppsala Universitetsbibliotek user on 23 May 2020


and their usage increased significantly in the late 2000s, associ-ated
Antimicrobial resistance continues to be a major threat to the de- with the reported rise in ESBL-producing bacteria. As such, they
livery of effective healthcare worldwide. MDR Gram-negative infec- have now become first-line agents for uncomplicated UTI in several
tions are now commonly seen in a variety of infections and 6
international guidelines. This now requires monitoring due to an
1
contribute substantially to this global public health problem. In the observed decline in efficacy due to their inability to adequate-ly treat
USA, of the 140 000 healthcare-associated Enterobacteriaceae complicated UTI and the slow development of resistance owing to
infections per year, 26 000 (18.6%) are MDR (predominantly due to 7
their increased use. In addition, fosfomycin activity against
2
EBSL-producing organisms), resulting in close to 1700 deaths. Klebsiella spp. has recently been challenged in an in vitro dynamic
Likewise, a point prevalence survey of US hospitals in 2017 8
bladder model. The older extended-spectrum penicillins,
revealed over 290 000 non-duplicate ESBL-producing pivmecillinam (also known as pivoxil amdinocillin) and temocillin,
3
Enterobacteriaceae isolates. Gram-negative uropathogens caus- have also demonstrated good in vitro and in vivo activity against
5,9–12
ing urinary tract infection (UTI) are a major contributor to global EBSL producers causing UTI. Pivmecillinam has significant
4 antibacterial potency, is stable to b-lactamase hydrolysis and shows
antibiotic use and resistance. Treatment options for ESBL-
producing uropathogens are often limited compared with non-ESBL- synergy when paired with b-lactamase inhibitors against ESBL-
13,14
producing pathogens. IV antimicrobial therapy is frequently producing Enterobacteriaceae. A recent cohort study showed
unavoidable as coexisting resistance to other oral agents (e.g. fluo- that oral pivmecillinam 400 mg three times daily yielded equivalent
roquinolones and sulphonamides) is often seen among ESBL pro- clinical outcomes in UTI caused by ESBL producers when
ducers. Nitrofurantoin and fosfomycin have provided some benefit in 10
compared with alternative oral antibiotics. More robust data from a
5
this patient group in the setting of uncomplicated UTI. They do randomized controlled trial are necessary in order for

VC The Author(s) 2020. Published by Oxford University Press on behalf of the British Society for Antimicrobial Chemotherapy. All rights reserved.
For permissions, please email: journals.permissions@oup.com.
1 of 10
Review
this strategy to be adopted worldwide. Recently, new compounds cefpodoxime, cefprozil, cefuroxime, cefalexin and loracarbef) and
pairing orally bioavailable cephalosporin antibiotics with novel b- where tested as non-susceptible to cefazolin, laboratories may go
lactamase inhibitors have been developed. This promises the po- on to test cefdinir, cefpodoxime and cefuroxime due to resistance
tential to improve oral treatment options and reduce healthcare over-calling by cefazolin.24 costs in patients with UTIs due to ESBL-
producing organisms.

Downloaded
First-generation cephalosporins

from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkaa183/5842233 by Uppsala Universitetsbibliotek user on 23 May 2020


Epidemiology of ESBL-producing organisms Cefalexin has activity against most Enterobacteriaceae causing UTIs;
causing UTI however, this activity is significantly reduced against ESBL producers. In
one study of non-ESBL-producing uropathogens, overall susceptibility
ESBLs render penicillins (except mecillinam and temocillin), first-, was 85% (Table 2). However, susceptibility of 981 ESBL-producing E.
second- and third-generation cephalosporins and monobactams coli and 439 ESBL-producing K. pneumoniae urinary isolates was only
5,15
ineffective via hydrolysis of the b-lactam ring. Uropathogens 6.32% and 0.91%, respectively, in one large study. 25 Cefadroxil, another
harbouring ESBLs, for example Escherichia coli ST131, are often first-generation cephalosporin, revealed average potency overall and a
re-sistant to other commonly used antibiotics, for example fluoroqui-
16,17 broad MIC50 range of 4 to >32 mg/L in a pooled analysis of 164
nolones via point mutations in the gyrA and parC genes. The
presence of ESBL genes among Enterobacteriaceae isolates has isolates.26,27
been progressively increasing in both hospital and community set-
tings, representing 15% of all Klebsiella spp. isolates and 12% of E. Second-generation cephalosporins
18
coli captured among 79 US hospitals. Considerable global Second-generation oral cephalosporins such as cefaclor exhibit
variability exists between countries, however, with Europe exhibiting greater activity against WT Gram-negative bacilli than does
6.8%–38.7% of E. coli and 4.5%–77.7% of Klebsiella pneumoniae cefa-lexin. Although still displaying a broad range of
19
as resistant to third-generation cephalosporins. Similarly, the susceptibility results—the majority reported against EUCAST
incidence of community-acquired ESBL-producing breakpoints—cefa-clor (47.5%–90.5%), cefuroxime (50.5%–
Enterobacteriaceae as a cause of UTI has increased worldwide, 86.4%) and cefprozil (90%–93.2%) were active against the
20
with some areas reporting rates of over 40% of all UTIs. Case– majority of uropathogens (Table 2). As with first-generation
control studies have identified numerous classical risk factors for cephalosporins, however, these antibiotics have poor
acquisition of ESBL-producing Enterobacteriaceae, which include susceptibility profiles against ESBL producers.28
recent antibiotic use (in particular fluoroquinolones), hospital
21
admission and surgery. The urinary tract is the most common site
of infection caused by ESBL producers and results in a sizeable
Third-generation cephalosporins
22
economic and public health burden. The hospitalization cost of an Oral third-generation cephalosporins perform better still, with MICs
ESBL-producing E. coli UTI compared with a non-ESBL-producing among Enterobacteriaceae consistently below EUCAST/CLSI
23 breakpoints for UTI. They demonstrate good in vitro efficacy against
E. coli was estimated to be e4980 versus e2612. uropathogenic Enterobacteriaceae and also exhibit some resistance
The most common globally disseminated ESBL genes found to hydrolysis by common ESBLs (Table 3). Cefpodoxime is a
amongst uropathogenic Enterobacteriaceae are those of the cephalosporin with activity against Gram-negative bacteria and
CTX-M group.4 In the modern era of increasing rates of MDR shows stability to many b-lactamases.
29,30
It exists as a pro-drug,
Enterobacteriaceae among urinary tract pathogens, oral agents cefpodoxime proxetil, that undergoes de-esterification in the
are likely to be less active compared with the time when many intestinal wall before active cefpodoxime (typically 50%) is released
31
guidelines were written. Unless novel oral antimicrobials are into the circulation. Optimal absorption requires low gastric pH and
devel-oped, the requirement for IV therapy will contribute a mean peak serum concentration of 2.18 g/L can be expected 3 h
significantly to the burden of care administered in hospitals and 32
post administration of a 200 mg dose. A dose of 100 mg 12 hourly
will no doubt drive a large increase in healthcare costs. 33
has been recommended for uncomplicated UTI. Similarly,
ceftibuten is another third-generation cephalo-sporin with stability to
Oral cephalosporins (without b-lactamase 34
plasmid-mediated b-lactamases. Ceftibuten resists hydrolysis by
inhibitors) and their activity against commonly occurring narrow-spectrum b-lactamases (e.g. OXA-1,
35
TEM-1 and SHV-1 types). However, it is readily hydrolysed by
Enterobacteriaceae 36
SHV-4/5 produced by some Klebsiella isolates. Ceftibuten is
Orally bioavailable cephalosporins are represented in first-, se-cond- absorbed (75%–90%) in its active form from the gastrointestinal
and third-generation classes. All have some level of in vitro activity tract, achieving peak plasma anti-biotic levels of 9.9 mg/L and 17
against non-ESBL-producing Enterobacteriaceae com-monly 37
mg/L after single oral doses of 200 mg and 400 mg, respectively.
isolated from urine. EUCAST has specified clinical break-points for Drug clearance is split be-tween renal (56%) and faecal (39%)
uncomplicated UTI against Enterobacteriaceae (Table 1) for excretion. Typical doses are 400 mg daily for respiratory tract
38
cefadroxil, cefuroxime, cefalexin, cefixime, cefpodoxime and infection; however, no dosing recommendations exist for UTI.
ceftibuten (susceptible if MICs are 16 mg/L, 8 mg/L, 16 mg/L, 1
mg/L, 1 mg/L and 1 mg/L respectively). However, CLSI has Among 301 US Enterobacteriaceae isolates, cefpodoxime dem-
recommended utilizing the cefazolin breakpoint for onstrated a higher susceptibility rate than cefuroxime (94% versus
Enterobacteriaceae due to uncomplicated UTI (susceptible 16 mg/L) 83%).39 Cefpodoxime and ceftibuten also performed well against 155
as a surrogate for oral agents (cefaclor, cefdinir, ESBL-negative MDR E. coli and K. pneumoniae urinary isolates,

2 of 10
Review JAC C8
5
>>0
e, 11.
f 665
p8
o7
ttt
d,
ooo
Table 1. Oral cephalosporin EUCAST and CLSI breakpoints for UTI o
x9
8
i 3>>>
EUCAST susceptible breakpoint (mg/L)
a
CLSI susceptible breakpoint
a,b
(mg/L) m86661.
Cephalosporin
e6444
1
First generation 2
,
cefadroxil 16 NA
cefalexin 16 NA C 81
Second generation e5, 6
f
cefuroxime 8 4
t
9t
cefaclor NA 8 i8
o
cefprozil NA 8 b,
u1
loracarbef NA 8
t902>
Third generation e94–6N6.
cefpodoxime 1 2 n484A
C 7
cefixime 1 1 e,
3

cefdinir NA 1 f
ceftibuten 1 8 p7
o5
cefetamet NA 4 d–7
o7
x,
NA, data not available. i
a
Uncomplicated UTI only. m 8

b e1
Cefazolin may be used as a surrogate (susceptible: 16 mg/L). /
c0
0
l .
.
Table 2. Pooled WT MIC data of oral cephalosporins against Enterobacteriaceae for which UTI breakpoints exist a 50
v6
u
t
l t
Total no. of isolates tested o
a0o8.
Oral cephalosporin Reference(s) across all studies MIC50 (mg/L) MIC90 (mg/L) Range (mg/L) Susceptibility (%) n1.
94, 95
a15>>
t94–33
Cefalexin 126 4–8 8–32 1 to >256 54.5–85
e14122
26, 27 C
Cefadroxil 164 8–16 4 to >32 4 to >32 — e
f
94, 95 t
Cefprozil 126 2 4–8 0.5 to >16 90–93.2 i
b
42, 95, 96
u
Cefaclor 27, 42, 94, 95 1432 2–16 4 to >32 0.5 to >128 47.5–90.5 t
Cefuroxime 837 2–8 4 to >32 0.25 to >32 50.5–86.4 e
n
94, 95 /
Cefdinir 126 0.12–0.5 0.5–1 0.12 to >4 90–95.2 c
l
42, 97
a
Cefixime 42, 95, 97 969 0.06–0.5 0.5 to >8 0.06 to >16 58.2–81.3 v
Cefpodoxime 1011 0.12–1 0.5 to >8 0.06 to >16 53.5–95.2 u00
l ..
27, 42 a11
Ceftibuten 711 0.12–0.25 0.5 to >16 0.01 to >16 66.3–91.3 n22
a55
t ––N
e411A
C 720
00
8
e,7...6.
f 6270
Table 3. In vitro activity of oral cephalosporins with or without b-lactamase inhibitors against ESBL-producing Enterobacteriaceae
i7559
x9 –
Oral cephalosporin ± Total no. of isolates tested MIC50 MIC90 Range Susceptibility i, 2
m4
b-lactamase inhibitor Reference(s) across all studies (mg/L) (mg/L) (mg/L) (%)
e8
1
/
Cefixime 78, 81 214 6 >64 0.5 to >64 7–8.6 c
84, 85

Cefpodoxime/QPX7728 NA 0.5 4 NA NA
84, 85

Ceftibuten/QPX7728 87, 100 NA 0.06 1 NA NA


Cefpodoxime/ETX0282 88, 89, 99, 101 937 0.015–0.5 0.03–1 0.12–2 NA
Ceftibuten/VNRX7145 884 0.06 to <1 0.12–1 NA 96.9–100

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkaa183/5842233 by Uppsala Universitetsbibliotek user on 23 May 2020


NA, data not available.
3 of 10
Review

32
showing 94% susceptibility each for cefpodoxime and 100% each subjects. Urinary excretion is 80% of the absorbed oral dose (ap-
40
for ceftibuten. Among 1190 Enterobacteriaceae isolates causing proximately 50% of total administered dose). After 8 days of 400 mg
twice-daily oral dosing of cefpodoxime proxetil, 146 mg (35.6%) and

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkaa183/5842233


UTI in Europe, ceftibuten and cefpodoxime had susceptibility rates
of 94% and 92%, respectively, with ceftibuten demonstrating in vitro 122 mg (30.6%) had been excreted in the urine in 12 h in young and
41 52
efficacy similar to that of ceftriaxone. Furthermore, 567 elderly healthy adults, respectively. Ceftibuten demonstrates a
community-acquired UTI Enterobacteriaceae isolates from fractional excretion of unchanged drug in the urine, with 56%–70%
Argentina, Mexico, Venezuela, Russia and the Philippines showed over 24 h. After an oral dose of 200 mg of ceftibuten, it can achieve
53
ceftibuten in vitro potency, being able to outperform other oral a maximum urinary concentration of ap-proximately 800 mg/L.
42
cephalosporins. The ceftibuten susceptibility rate was 77% by Cefixime undergoes less urinary clear-ance. After administering 200
54
EUCAST, with MIC50/90 of 0.25/16 mg/L and a range of 0.06 to mg, 53.3 mg (27%) can be recovered from the urine after 24 h. In
>16 mg/L. Cefixime, cefpodoxime, cefuroxime and cefaclor had another study, adminis-tering 200 mg and 400 mg of cefixime
42 55
in vitro susceptibilities of 67%, 65%, 65% and 58%, respectively. yielded urinary recovery of 20% and 16% of the dose. Maximum
Ceftibuten alone was tested in vitro against CTX-M-producing E. coli concentrations achieved in the urine were 107 mg/L and 164 mg/L
isolates and showed a disparity in susceptibility between for a 200 mg and 400 mg dose, respectively. Cefdinir has an even
CTX-M group 1 and group 9, with MIC 50/90 values of 8/32 and 1/1 lower fractional elimin-ation of unchanged drug in the urine (13%–
mg/L, respectively, and susceptibility rates of 9.8% and 94.5%, 56
23%). In one study, the mean percentage of the dose recovered
respectively.12 unchanged in the urine following 300 mg and 600 mg doses was
57
18.4% and 11.6%, respectively, over 24 h. Although third-
Antibiotic concentrations in the urinary tract generation cephalospor-ins achieve lower urinary concentrations
when compared with first-generation cephalosporins, both
First-generation cephalosporins cefpodoxime and ceftibuten appear to be superior to cefixime and
cefdinir.
Oral cephalosporins have been shown to have favourable pharma-
cokinetics and to achieve high concentrations of active drug in the
urine. Cefalexin is excreted unchanged in the urine, with 70%– Oral clavulanate
100% of the dose found in the urine 6–8 h after a dose and concen-
Clavulanate is usually paired with amoxicillin and has been used
trations many times greater than the MIC for a typical urinary
43 in the treatment of UTI. Clavulanate is also excreted in the urine
pathogen (urine cefalexin concentration 500–1000 mg/L). in its active unchanged form, although far less than many oral
Although increasing resistance rates to cefalexin have been noted, cephalo-sporins. After administration of 125 mg of clavulanic
these high urinary concentrations may allow it to retain effective- acid, cumula-tive excretion at 2, 4 and 6 h was 14%, 25.7% and
44
ness for uncomplicated UTIs. Similarly, cefadroxil is excreted in 27.8%, respectively.58 Other studies have documented similar
urine (via glomerular filtration and tubular secretion), with 93% of the values of between 18% and 38% of total excretion. Clavulanate
dose passing through the urine in the first 24 h, achieving con-
26
reaches a maximum urinary concentration of 40 mg/L, 4–6 h
centrations between 400 and 2400 mg/L. Its urinary excretion rate after a 125 mg oral dose. Approximately half of the drug is
45
is more prolonged when compared with cefalexin. metabolized elsewhere in the body.59

Second-generation cephalosporins Clinical trials of orally administered

by Uppsala Universitetsbibliotek user on 23 May 2020


Cefaclor achieves slightly lower urinary antibiotic concentrations cephalosporins (without b-lactamase
overall (50–1000 mg/L), with 70% of active drug passing through the
46
urine in the first 6 h. It also has other major non-renal routes of
inhibitors) for UTI
47
elimination, such as the biliary tract. Cefprozil shows slightly Numerous clinical trials have been conducted examining the use
poorer urinary concentrations, excreting around 61% of the of oral cephalosporins in the treatment of UTI. The majority of
administered dose and achieving urinary concentrations of 175 mg/L patients enrolled in these trials had uncomplicated infection with
48
and 658 mg/L at 4 h after a 250 mg and 1 g dose, re-spectively. urinary pathogens without ESBL production.
Cefuroxime axetil (oral formulation) has its parent compound,
cefuroxime, eliminated unchanged in the urine (42%– 57% in 24 h
First-generation cephalosporins
after a single oral 250 mg dose) and achieves high concentrations in
49
the renal parenchyma. It achieves peak con-centrations of 400 Oral cefalexin has been utilized as a comparator arm in many
50 therapeutic trials for uncomplicated UTI. In 109 patients receiving
mg/L at 4 h after a 500 mg oral dose.
oral cefalexin 250 mg four times a day for acute uncomplicated UTI,
93.6% and 80.6% were able to achieve clinical cure and micro-
Third-generation cephalosporins 60
biological cure, respectively. In a trial comparing cefalexin 500 mg
Third-generation or extended-spectrum oral cephalosporins also with ampicillin 500 mg, each four times a day, 30/31 (96.8%)
penetrate the kidneys and urinary tract well. Cefpodoxime clear- patients who received cefalexin achieved clinical cure and 20/31
ance occurs primarily through the renal route by both glomerular (64.5%) had a sterile urine sample collected 3 weeks after
filtration and tubular secretion.32 Over various approved doses 61
commencement of therapy. Furthermore, in another double-blind
(100–400 mg), 29%–33% of administered cefpodoxime was clinical trial, of 115 patients with complicated UTI receiving oral
excreted unchanged in urine in a 12 h period. 51 This increased cefalexin 500 mg four times a day, clinical cure was reported in only
to 42.8% over 24 h after a 100 mg dose was given to healthy 75 (65.2%) patients.
62
Cefadroxil 1 g twice daily was

4 of 10
Review JAC
compared with norfloxacin 400 mg twice daily in the treatment of Oral cephalosporin and clavulanic acid combinations
acute pyelonephritis in a multicentre clinical trial that enrolled 197 Clavulanic acid is a highly effective inhibitor of ESBLs in vitro and is

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkaa183/5842233 by Uppsala Universitetsbibliotek user on 23 May 2020


63
patients. Although cefadroxil produced an inferior microbiologic-al typically combined with amoxicillin in both oral and IV formula-
cure rate (65% versus 98%), the clinical response rate did not dif-fer 73
tions. Compared with other b-lactam/b-lactamase (BLBLI) com-
between groups. With regard to clinical cure, 7 days of oral binations, amoxicillin is less stable to ESBLs even in the presence of
cefadroxil 1 g daily performed just as well as amoxicillin 375 mg clavulanate. Oxyimino-cephalosporins are weaker substrates than
64 73
three times a day in women with uncomplicated UTI. amoxicillin for ESBLs and have a higher affinity for PBPs. A fixed-
dose combination of cefuroxime/clavulanic acid has been approved
and introduced in some countries, although currently this is not
Second-generation cephalosporins 74
approved by the US FDA. Many oral cephalosporin anti-biotics
The second-generation cephalosporins cefprozil and cefaclor have have been tested in vitro, in combination with clavulanic acid,
been compared against each other in a number of clinical trials for against ESBL-producing Enterobacteriaceae (Table 3). Among 380
uncomplicated UTI. In one study of cefprozil 500 mg daily and ESBL-producing E. coli and 226 Klebsiella spp. isolates, cef-
cefaclor 250 mg three times daily, clinical response was 87% ver- podoxime combined with clavulanate produced MIC50/90 values of
65 73
sus 84% and microbiological cure was 83% versus 85%. Among 0.5/2 and 0.5/1 mg/L, respectively. Twenty-eight of 37 (75.6%)
108 college women with acute UTI, the clinical response was 94% ESBL-producing E. coli from India had MIC values less than the
versus 94% and microbiological cure was 93% versus 94%, re- cefpodoxime-susceptible breakpoint when combined with clavu-
66 75
spectively. Oral cefuroxime axetil 125 mg twice daily was com- lanate. In another in vitro study, 101 ESBL-producing
pared with enoxacin (a fluoroquinolone) in a small clinical trial Enterobacteriaceae were tested against cefdinir and cefpodoxime,
involving 33 patients with recurrent UTI and achieved clinical re- in combination with amoxicillin/clavulanate at fixed concentra-tions
76
sponse and microbiological cure rates of 71.4% and 71.4%, com- of 8 mg/L and 4 mg/L respectively. Utilizing CLSI cefdinir and
pared with 92.3% and 85.7% in the enoxacin group. In a large
67 cefpodoxime breakpoints prior to 2011, susceptibility amongst CTX-
clinical trial involving 672 women with uncomplicated UTI, treated M producers was reported at 90.9% and 89.3%, respectively, whilst
producers of SHV-type ESBLs were reported as susceptible for 60%
with cefuroxime axetil 125 mg twice daily or 250 mg twice daily,
68 and 58.8%, possibly due to differences in substrate affinity for CTX-
628/672 (93.4%) achieved clinical cure. M enzymes and SHV ESBL enzymes towards cefdinir and
cefpodoxime (Table 4). In addition, against 303 CTX-M-producing E.
coli, cefpodoxime/clavulanate demonstrated potent activity
Third-generation cephalosporins (MIC50/90 0.5/0.5 mg/L) but, not surprisingly, was less active
Cefdinir 100 mg twice daily was compared with cefaclor 250 mg against those hyperproducing AmpC (MIC50/90 4/4 mg/L).77
three times a day, both for 5 days in uncomplicated UTI, in a large Cefixime plus amoxicillin/clavulanate tested against 64 ESBL-
69
multicentre trial involving 661 participants. Cefdinir achieved a producing E. coli isolates produced MIC50/90 values of 0.25/75 mg/L
clinical response and microbiological cure rate of 85.9% and 91.3%, (range 0.09–24 mg/L).78 Cefixime/clavulanate also performed well in
respectively, with cefaclor achieving similar results (80.5% and 93%, vitro against non-AmpC ESBL producers in 62 E. coli and K. pneu-
respectively). With regard to extended-spectrum oral moniae isolates.79 Both of these studies, however, demonstrated
cephalosporins, cefpodoxime 100 mg twice daily did not meet the synergy using non-standardized methods not recommended by
criteria for non-inferiority when compared with oral ciprofloxacin in a either EUCAST or CLSI, thereby limiting the value of their interpret-
70
randomized trial in acute uncomplicated cystitis. Patients who ation. Cefdinir plus a fixed concentration of amoxicillin/clavulanate
(8/4 mg/L) was tested against CTX-M (46 isolates) and SHV/TEM
received oral cefpodoxime proxetil 100 mg twice daily for 3 days
achieved an overall clinical cure rate at 30 day follow-up in the ITT (11 isolates) producers, which revealed in vitro MIC50/90 values of
population of 82% (123/150) compared with 93% (139/ 0.25/2 mg/L (89.1% cefdinir susceptible) and 0.06/8 mg/L (81.8%
70 cefdinir susceptible), respectively.80
150) in those receiving oral ciprofloxacin 250 mg twice daily. The
The addition of amoxicillin/clavulanate to oral cephalosporins
authors of that study speculate that the lower clinical response
was shown to have high rates of synergy when tested using disc
seen with b-lactam therapy may be due to their poorer activity in
70
approximation and disc replacement methods amongst 150 ESBL-
eradicating uropathogens from the vaginal flora. Cefpodoxime 100 producing E. coli isolates predominantly isolated from urinary sour-
mg twice daily was also compared with trimethoprim/sulfa- 81
ces. The frequency of synergy using this method with cefixime and
methoxazole in acute uncomplicated cystitis in a small open-label
cefpodoxime was 84% and 75.5%, respectively. Moreover, 86.3%
multicentre trial, which showed that 3 days of cefpodoxime was
and 74.1% of isolates were in the susceptible range when cefixime
as effective as trimethoprim/sulfamethoxazole.71 Another small and cefpodoxime were added to amoxicillin/clavulanate and
randomized clinical trial compared ceftibuten 200 mg twice daily interpreted using CLSI disc diffusion breakpoints. Twenty patients
and cefixime 200 mg twice daily in the treatment of complicated with confirmed ESBL-producing E. coli UTI, 85% of whom had
UTI and showed similar clinical and microbiological outcomes demonstrable positive in vitro synergy, received cefixime and
be-tween the two drugs.72 Thus, it is unclear from clinical trial amoxicillin/clavulanate, with a 90% clinical cure rate observed. A
data which oral third-generation cephalosporin is superior with case series of 10 patients treated with ceftibuten plus amoxicillin/
regard to clinical and microbiological cure. It should also be clavulanate for ESBL-producing E. coli and K. pneumoniae UTI
pointed out that all of these studies were performed in scenarios 82
showed all patients achieving clinical cure. Overall, the addition of
where ESBL-producing strains were rarely encountered. clavulanic acid to a third-generation cephalosporin was able to

5 of 10
Review
Table 4. Typical MICs (mg/L) of orally administered third-generation cephalosporins for selected b-lactamase-producing Enterobacteriaceae

MIC (mg/L)

MIC reduction

Downloaded from https://academic.oup.com/jac/advance-article-


Organism and b-lactamase References cefdinir cefixime ceftibuten cefpodoxime with clavulanate?

102, 103

E. coli ! AmpC >64 >64 >256 16 to >64 no


12, 36, 82, 103

E. coli ! CTX-M-1 8 4–16 1–8 1–64 yes


12, 82

E. coli ! CTX-M-9 1 0.5–1 yes


30, 102–106

E. coli ! TEM-1 0.5 0.25–0.5 0.12–1 0.5–1 yes


103, 105, 106

E. coli ! TEM-2 0.12 0.06–0.12 0.12 yes


30, 105, 106

E. coli ! OXA-1 0.5 0.1–0.25 0.25 0.5 yes


102, 105, 106

E. coli ! OXA-2 0.25 0.25–0.5 0.5 0.5 yes


36

K. pneumoniae ! CTX-M-1 NA 8 0.25 32 yes


36

K. pneumoniae ! AmpC NA >64 >64 >64 no


30

K. pneumoniae ! SHV-1 NA NA NA 0.25 yes


102, 105, 106

K. pneumoniae ! K14 30, 102, 105, 106 4 0.25 0.06 8 yes


K. pneumoniae ! hyperexpressed K1 30, 35, 102, 106, 107 4 0.25 0.06 0.25–8 yes
Enterobacter ! basal AmpC 0.12 to >128 4 to >32 0.5 to >32 2 to >128 no

NA, data not available.


MICs determined by BMD lie within the susceptible distribution
significantly reduce the MIC50/90 against organisms producing
range for cephalosporins alone.
N
spe-cific ESBLs (CTX-M, TEM or SHV-1) with little activity
o
against AmpC producers (Table 4).
v
e
Limitations of in vitro oral cephalosporin/b-
l
lactamase inhibitor susceptibility data
interpretation b
In the current literature, cephalosporin/b-lactamase inhibitor -
susceptibility testing (as with other BLBLI combinations) appears to
l
be frequently performed by non-standardized methods and exhibits
great heterogeneity in methodology. Methods to deter-mine a
antimicrobial susceptibility and synergy in oral cephalo-sporin/b- c
lactamase inhibitor combinations have included disc diffusion, broth t
microdilution (BMD), Etest, disc approximation and disc a
73,75–77
replacement. In addition, different concentra-tions of b- m
lactamase inhibitors are often used amongst testing strategies. With
regard to clavulanic acid, either fixed concentra-tions of 2 mg/L or 4
a
mg/L have been used, or ratios of 2:1 (cepha-losporin:clavulanate) s
have been reported. Despite having standardized and accepted e
clinical breakpoints for both EUCAST and CLSI for many oral
cephalosporins, when assessing them in combination with i
clavulanate, interpretation can be problematic as the two differ with
regard to methodology (fixed 2 mg/L for EUCAST and 2:1 ratio for
n
83
CLSI). Many reported studies used fixed combinations of 4 mg/L,
h
not complying with either EUCAST or CLSI. The spectrum of testing i
performed highlights a need for further research towards a unified b
approach. Ultimately, BMD and disc diffusion MIC distributions are i
required to determine ap-propriate breakpoints but, similarly to other
t
BLBLIs currently available, it would seem prudent to assess activity
for cephalo-sporin/clavulanate combinations based on whether the
o
rs in combination with ceftibuten or

abstract/doi/10.1093/jac/dkaa183/5842233 by Uppsala Universitetsbibliotek user on 23 May 2020


cefpodoxime
There has been a recent drive to develop new oral antimicrobials
that are active against ESBL-producing uropathogens. QPX7728 is
a next-generation cyclic boronic acid b-lactamase inhibitor that is
orally bioavailable and inhibits all clinically important b-lacta-
84
mases. It has been shown to restore the activity of cefpodoxime
and ceftibuten on ESBL producers, with MIC 50/90 values of 0.5/4 and
85
0.06/1 mg/L, respectively. ETX0282 is an oral prodrug that
is hydrolysed in vivo to release ETX1317, another novel b-lacta-
mase inhibitor that is orally bioavailable and inhibits Class A and C
86
b-lactamases. When paired with cefpodoxime it was able to sig-
nificantly reduce bacterial titres in kidney, bladder and urine in a
86
murine ESBL-producing E. coli UTI model. ETX1317 was able to
restore the activity of cefpodoxime in 1875 global
Enterobacteriaceae UTI isolates, with MIC50/90 values of 0.06/ 0.12
87
mg/L. It demonstrated similar potency against AmpC pro-ducers.
VNRX7145 is another b-lactamase inhibitor that undergoes
biotransformation to the active VNRX5236 with potent activity
against Class A, B and C enzymes. Among 100 isolates of
Enterobacteriaceae, ceftibuten/VNRX5236 maintained an MIC 90 of 1
mg/L across all enzyme subgroups tested (ESBL, KPC, AmpC and
88
OXA-48). Moreover, VNRX5236 was able to reduce the MIC
values of ceftibuten for a similar profile of susceptible isolates com-
pared with ceftazidime/avibactam and meropenem against ESBL
88
producers. Of 50 ESBL-producing isolates tested, MIC50/90 values
were 0.06/0.12 mg/L, with 98% susceptible by EUCAST criteria for
89
ceftibuten. Ceftibuten/VNRX5236 has also demonstrated in vivo
efficacy in a neutropenic thigh infection model against serine b-
lactamase-producing Enterobacteriaceae, achieving a mean re-
90
duction in bacterial burden of #0.2 log10 cfu/thigh. Ceftibuten/
clavulanate is another novel BLBLI combination that also has po-
tential utility in the management of UTIs caused by ESBL pro-
ducers. It exhibited promising activity in four ESBL-producing

6 of 10
Review JAC
isolates.91 It has also shown favourable in vivo activity against 3 Gupta V, Ye G, Olesky M et al. National prevalence estimates for
ESBL-producing Enterobacteriaceae in murine thigh model resistant Enterobacteriaceae and Acinetobacter species in hospitalized
patients in the United States. Int J Infect Dis 2019; 85: 203–11.
planned.92

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkaa183/5842233 by Uppsala Universitetsbibliotek user on 23 May 2020


studies and human clinical trials are
Pharmacodynamic studies have been helpful in designing 4 Zowawi HM, Harris PN, Roberts MJ et al. The emerging threat of multidrug-

potential dosage regimens for this combination molecule.91 resistant Gram-negative bacteria in urology. Nat Rev Urol 2015; 12: 570–84.

A Phase 1 clinical trial to establish the safety and 5 Giske CG. Contemporary resistance trends and mechanisms for the
pharmacokin-etics of ETX0282 is registered and underway. 93 old antibiotics colistin, temocillin, fosfomycin, mecillinam and
nitrofurantoin. Clin Microbiol Infect 2015; 21: 899–905.
Clinical trials involv-ing these other new agents are expected to
be registered in the near future. 6 Huttner A, Verhaegh EM, Harbarth S et al. Nitrofurantoin revisited: a
sys-tematic review and meta-analysis of controlled trials. J Antimicrob
Chemother 2015; 70: 2456–64.
Conclusions 7 Hoang P, Salbu RL. Updated nitrofurantoin recommendations in the elder-
The increase in frequency of ESBL-producing ly: a closer look at the evidence. Consult Pharm 2016; 31: 381–4.
Enterobacteriaceae as causes of UTI worldwide is a concern. 8 Abbott IJ, Meletiadis J, Belghanch I et al. Fosfomycin efficacy and emer-
More alarming still is the limited number of oral antibiotic options gence of resistance among Enterobacteriaceae in an in vitro dynamic bladder
currently available to clinicians to treat these infections. Recent infection model. J Antimicrob Chemother 2018; 73: 709–19.
enthusiasm and drug development directed towards 9 Dewar S, Reed LC, Koerner RJ. Emerging clinical role of
cephalosporin/b-lactamase inhibitor combinations with good oral pivmecillinam in the treatment of urinary tract infection in the context of
bioavailability has pro-vided some much-needed optimism. multidrug-resistant bac-teria. J Antimicrob Chemother 2014; 69: 303–8.
Novel oral b-lactamase in-hibitor compounds QPX7728, 10 Bollestad M, Grude N, Solhaug S et al. Clinical and bacteriological
ETX0282 and VNRX5236, when paired with the oral third- efficacy of pivmecillinam treatment for uncomplicated urinary tract infections
generation cephalosporins ceftibuten and cefpodoxime were caused by ESBL-producing Escherichia coli: a prospective, multicentre,
able to demonstrate good in vitro activity against MDR Gram- observational cohort study. J Antimicrob Chemother 2018; 73: 2503–9.
negative uropathogens. However, analysis of their potency is 11 Jansaker F, Frimodt-Moller N, Sjogren I et al. Clinical and bacteriological
significantly limited by non-standardized testing methods and effects of pivmecillinam for ESBL-producing Escherichia coli or Klebsiella pneu-
limited consensus regarding application of clinical breakpoints. moniae in urinary tract infections. J Antimicrob Chemother 2014; 69: 769–72.

Moreover, the combination of two established com-pounds, 12 Tarnberg M, Ostholm-Balkhed A, Monstein HJ et al. In vitro activity
ceftibuten and clavulanate, has also demonstrated encouraging of b-lactam antibiotics against CTX-M-producing Escherichia coli. Eur J
Clin Microbiol Infect Dis 2011; 30: 981–7.
efficacy. A clinical trial assessing the safety and effi-cacy of
cefpodoxime/ETX0282 is currently underway. Well-designed 13 Sougakoff W, Jarlier V. Comparative potency of mecillinam and other b-
studies assessing the pharmacokinetic/pharmacody-namic lactam antibiotics against Escherichia coli strains producing different b-lacta-
mases. J Antimicrob Chemother 2000; 46 Suppl 1: 9–65.
profile of both new and old antimicrobials used to treat ESBL-
producing UTI continue to be important. Orally adminis-tered 14 Wootton M, Walsh TR, Macfarlane L et al. Activity of mecillinam
carbapenems (e.g. tebipenem) are being developed and it against Escherichia coli resistant to third-generation cephalosporins. J
Antimicrob Chemother 2010; 65: 79–81.
remains to be seen how their efficacy compares with orally
administered BLBLI combinations. Given the rising incidence of 15 Paterson DL, Bonomo RA. Extended-spectrum b-lactamases: a
clinical update. Clin Microbiol Rev 2005; 18: 657–86.
carbapenem-resistant organisms worldwide, the search to find a
suitable ‘carbapenem-sparing’ agent has become a top priority. 16 Petty NK, Ben Zakour NL, Stanton-Cook M et al. Global
dissemination of a multidrug resistant Escherichia coli clone. Proc Natl
Acad Sci U S A 2014; 111: 5694–9.
Transparency declarations 17 Ben Zakour NL, Alsheikh-Hussain AS, Ashcroft MM et al. Sequential
P.N.A.H. has received funding from Pfizer, Merck Sharpe & Dohme acqui-sition of virulence and fluoroquinolone resistance has shaped the
(MSD), and Shionogi. D.L.P. has received funding from AstraZeneca, evolution of Escherichia coli ST131. MBio 2016; 7: e00347–16.
Leo Pharmaceuticals, Bayer, GlaxoSmithKline (GSK), Cubist, Venatorx 18 McDanel J, Schweizer M, Crabb V et al. Incidence of extended-
and Accelerate; reports board membership from Entasis, Qpex, Merck, spectrum b-lactamase (ESBL)-producing Escherichia coli and Klebsiella
Shionogi, Achaogen, AstraZeneca, Leo Pharmaceuticals, Bayer, GSK, infections in the United States: a systematic literature review. Infect
Cubist, Venatorx, and Accelerate; reports grants/grants pending from Control Hosp Epidemiol 2017; 38: 1209–15.
Shionogi and Merck; and has received payment for lectures including
service on speaker’s bureaus from Pfizer, outside the submitted work.
19 ECDC. Data from the ECDC Surveillance Atlas—Antimicrobial
Resistance. 2020. https://www.ecdc.europa.eu/en/antimicrobial-
All other authors: none to declare.
resistance/surveillance-and-disease-data/data-ecdc.
20 Castillo-Tokumori F, Irey-Salgado C, Malaga G. Worrisome high
References frequency of extended-spectrum b-lactamase-producing Escherichia coli
in community-acquired urinary tract infections: a case-control study. Int J
1 Stewart A, Wright H, Hajkowicz K. The rise and rise of antimicrobial Infect Dis 2017; 55: 16–9.
resist-ance in Gram-negative bacteria. Microbiol Aust 2019; 40: 62–5.
21 Osthoff M, McGuinness SL, Wagen AZ et al. Urinary tract infections
2 Exner M, Bhattacharya S, Christiansen B et al. Antibiotic resistance:
due to extended-spectrum b-lactamase-producing Gram-negative
what is so special about multidrug-resistant Gram-negative bacteria?
bacteria: identi-fication of risk factors and outcome predictors in an
GMS Hyg Infect Control 2017; 12: Doc05.
Australian tertiary referral hospital. Int J Infect Dis 2015; 34: 79–83.

7 of 10
Review

22 Hertz FB, Schonning K, Rasmussen SC et al. Epidemiological factors with community-acquired urinary tract infections in three European coun-
asso-ciated with ESBL- and non ESBL-producing E. coli causing urinary tract tries. Clin Microbiol Infect 2016; 22: 63.e1–5.
infec-tion in general practice. Infect Dis (Lond) 2016; 48: 241–5.

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkaa183/5842233 by Uppsala Universitetsbibliotek user on 23 May 2020


42 Biedenbach DJ, Badal RE, Huang MY et al. Erratum to: In vitro
23 Esteve-Palau E, Solande G, Sanchez F et al. Clinical and economic activity of oral antimicrobial agents against pathogens associated with
impact of urinary tract infections caused by ESBL-producing Escherichia coli community-acquired upper respiratory tract and urinary tract infections: a
requiring hospitalization: a matched cohort study. J Infect 2015; 71: 667–74. five country surveillance study. Infect Dis Ther 2016; 5: 405.

24 EUCAST. Clinical breakpoints and dosing of antibiotics. 2019. 43 Griffith RS. The pharmacology of cephalexin. Postgrad Med J 1983;
http://www. eucast.org/clinical_breakpoints/. 59 Suppl 5: 16–27.

25 Toner L, Papa N, Aliyu SH et al. Extended-spectrum b-lactamase- 44 Warren JW, Abrutyn E, Hebel JR et al. Guidelines for antimicrobial
produc-ing Enterobacteriaceae in hospital urinary tract infections: incidence treat-ment of uncomplicated acute bacterial cystitis and acute
and anti-biotic susceptibility profile over 9 years. World J Urol 2016; 34: pyelonephritis in women. Clin Infect Dis 1999; 29: 745–58.
1031–7. 45 Pfeffer M, Jackson A, Ximenes J et al. Comparative human oral
26 Hartstein AI, Patrick KE, Jones SR et al. Comparison of clinical pharmacology of cefadroxil, cephalexin, and cephradine.
pharmacological and antimicrobial properties of cefadroxil and Antimicrob Agents Chemother 1977; 11: 331–8.
cephalexin. Antimicrob Agents Chemother 1977; 12: 93–7. 46 Santoro J, Agarwal BN, Martinelli R et al. Pharmacology of cefaclor
27 Bragman SG, Casewell MW. The in-vitro activity of ceftibuten in nor-mal volunteers and patients with renal failure. Antimicrob Agents
against 475 clinical isolates of Gram-negative bacilli, compared with Chemother 1978; 13: 951–4.
cefuroxime and cefadroxil. J Antimicrob Chemother 1990; 25: 221–6. 47 Levison ME, Santoro J, Agarwal BN. In vitro activity and
28 Qiao LD, Chen S, Yang Y et al. Characteristics of urinary tract infection pharmacokinetics of cefaclor in normal volunteers and patients with
pathogens and their in vitro susceptibility to antimicrobial agents in China: renal failure. Postgrad Med J 1979; 55 Suppl 4: 12–6.
data from a multicenter study. BMJ Open 2013; 3: e004152. 48 Barbhaiya RH, Shukla UA, Gleason CR et al. Comparison of
29 Borin MT, Hughes GS, Spillers CR et al. Pharmacokinetics of cefprozil and cefaclor pharmacokinetics and tissue penetration.
cefpodoxime in plasma and skin blister fluid following oral dosing of Antimicrob Agents Chemother 1990; 34: 1204–9.
cefpodoxime proxetil. Antimicrob Agents Chemother 1990; 34: 1094–9. 49 Perry CM, Brogden RN. Cefuroxime axetil: a review of its antibacterial activity,
30 Wise R, Andrews JM, Ashby JP et al. The in-vitro activity of pharmacokinetic properties and therapeutic efficacy. Drugs 1996; 52: 125–58.
cefpodoxime: a comparison with other oral cephalosporins. J Antimicrob 50 Klepser ME, Marangos MN, Patel KB et al. Clinical pharmacokinetics
Chemother 1990; 25: 541–50. of newer cephalosporins. Clin Pharmacokinet 1995; 28: 361–84.
31 Sader HS, Jones RN, Washington JA et al. In vitro activity of
cefpodoxime compared with other oral cephalosporins tested against
51 US FDA. Cefpodoxime Product Information. 2013. https://www.access
5556 recent clinical isolates from five medical centers. Diagn Microbiol data.fda.gov/drugsatfda_docs/label/2013/050674s015,050675s018lbl.pdf.
Infect Dis 1993; 17: 143–50. 52 Borin MT, Hughes GS, Patel RK et al. Pharmacokinetic and tolerance
stud-ies of cefpodoxime after single- and multiple-dose oral administration of
32 Tremblay D, Dupront A, Ho C et al. Pharmacokinetics of
cefpodoxime in young and elderly volunteers after single doses. J cef-podoxime proxetil. J Clin Pharmacol 1991; 31: 1137–45.
Antimicrob Chemother 1990; 26 Suppl E: 21–8. 53 Novelli A, Rosi E. Pharmacological properties of oral antibiotics for
33 Portier H, Chavanet P, Waldner-Combernoux A et al. Five versus the treatment of uncomplicated urinary tract infections. J Chemother
ten days treatment of streptococcal pharyngotonsillitis: a randomized 2017; 29 Suppl 1: 10–8.
controlled trial comparing cefpodoxime proxetil and phenoxymethyl 54 Westphal JF, Jehl F, Adloff M et al. Role of intrahepatic protein
penicillin. Scand J Infect Dis 1994; 26: 59–66. binding in the hepatobiliary extraction profile of cefixime in humans. Clin
34 Wise R, Andrews JM, Ashby JP et al. Ceftibuten–in-vitro activity Pharmacol Ther 1993; 54: 476–84.
against re-spiratory pathogens, b-lactamase stability and mechanism of 55 Nakashima M, Uematsu T, Takiguchi Y et al. Phase I study of
action. J Antimicrob Chemother 1990; 26: 209–13. cefixime, a new oral cephalosporin. J Clin Pharmacol 1987; 27: 425–31.
35 Jones RN, Barry AL. Ceftibuten (7432-S, SCH 39720): comparative 56 Guay DR. Cefdinir: an expanded-spectrum oral cephalosporin. Ann
anti-microbial activity against 4735 clinical isolates, b-lactamase stability Pharmacother 2000; 34: 1469–77.
and broth microdilution quality control guidelines. Eur J Clin Microbiol
Infect Dis 1988; 7: 802–7. 57 US FDA. Cefdinir Product Information. 1997. https://www.accessdata.fda.
gov/drugsatfda_docs/label/1999/50739S2LBL.PDF.
36 Bauernfeind A. Ceftibuten and bactericidal kinetics. Comparative in
vitro activity against Enterobacteriaceae producing extended spectrum 58 Ferslew KE, Daigneault EA, Aten EM et al. Pharmacokinetics and
b-lacta-mases. Diagn Microbiol Infect Dis 1991; 14: 89–92. urinary excretion of clavulanic acid after oral administration of amoxicillin
and potas-sium clavulanate. J Clin Pharmacol 1984; 24: 452–6.
37 Radwanski E, Teal M, Affrime M et al. Multiple-dose pharmacokinetics of
ceftibuten in healthy adults and geriatric volunteers. Am J Ther 1994; 1: 42–8. 59 Staniforth DH, Jackson D, Horton R et al. Parenteral Augmentin:
pharma-cokinetics. Int J Clin Pharmacol Ther Toxicol 1984; 22: 430–4.
38 Stein GE, Christensen S, Mummaw N. Treatment of acute uncomplicated
urinary tract infection with ceftibuten. Infection 1991; 19: 124–6. 60 Menday AP. Comparison of pivmecillinam and cephalexin in acute
uncomplicated urinary tract infection. Int J Antimicrob Agents 2000; 13:
39 Bookstaver DA, Bland CM, Woodberry MW et al. Correlation of
183–7.
cefpodox-ime susceptibility with cephalothin and cefuroxime for urinary
tract isolates. J Med Microbiol 2014; 63: 218–21. 61 Davies JA, Strangeways JE, Mitchell RG et al. Comparative double-
40 Chen YT, Ahmad Murad K, Ng LS et al. In vitro efficacy of six alternative blind trial of cephalexin and ampicillin in treatment of urinary infections.
antibiotics against multidrug resistant Escherichia coli and Klebsiella pneumo-niae Br Med J 1971; 3: 215–7.
from urinary tract infections. Ann Acad Med Singapore 2016; 45: 245–50. 62 Kamidono S, Harada M, Ishigami J et al. Comparative double-blind
41 Kresken M, Korber-Irrgang B, Biedenbach DJ et al. Comparative in vitro study of cefroxadine and cephalexin in the treatment of complicated
ac-tivity of oral antimicrobial agents against Enterobacteriaceae from patients urinary tract infection. Jpn J Antibiot 1983; 36: 2571–94.

8 of 10
Review JAC
63 Sandberg T, Englund G, Lincoln K et al. Randomised double-blind 81 Al-Tamimi M, Abu-Raideh J, Albalawi H et al. Effective oral
study of norfloxacin and cefadroxil in the treatment of acute combination treatment for extended-spectrum b-lactamase-producing
pyelonephritis. Eur J Clin Microbiol Infect Dis 1990; 9: 317–23. Escherichia coli. Microb Drug Resist 2019; 25: 1132–41.

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkaa183/5842233 by Uppsala Universitetsbibliotek user on 23 May 2020


64 Sandberg T, Henning C, Iwarson S et al. Cefadroxil once daily for 82 Cohen Stuart J, Leverstein-Van Hall M, Kortmann W et al. Ceftibuten
three or seven days versus amoxycillin for seven days in uncomplicated plus amoxicillin-clavulanic acid for oral treatment of urinary tract infections
urinary tract infections in women. Scand J Infect Dis 1985; 17: 83–7. with ESBL producing E. coli and K. pneumoniae: a retrospective
65 Iravani A, Doyle CA, Durham SJ et al. Cefprozil versus cefaclor in observational case-series. Eur J Clin Microbiol Infect Dis 2018; 37: 2021–5.
the treat-ment of acute and uncomplicated urinary tract infections. 83 Delgado-Valverde M, Valiente-Mendez A, Torres E et al. MIC of
Cefprozil Multicenter Study Group. Clin Ther 1992; 14: 314–26. amoxicil-lin/clavulanate according to CLSI and EUCAST: discrepancies
66 Iravani A. Comparison of cefprozil and cefaclor for treatment of and clinical im-pact in patients with bloodstream infections due to
acute urinary tract infections in women. Antimicrob Agents Chemother Enterobacteriaceae. J Antimicrob Chemother 2017; 72: 1478–87.
1991; 35: 1940–2. 84 Lomovskaya OP. QPX7728 Ultra Broad Spectrum Beta Lactamase
67 Brumfitt W, Hamilton-Miller JM, Walker S. Enoxacin relieves Inhibitor for IV and Oral Combination Therapy. ASM Microbe, San
symptoms of recurrent urinary infections more rapidly than cefuroxime Francisco, USA, 2019.
axetil. Antimicrob Agents Chemother 1993; 37: 1558–9. 85 Griffith DC. Qpex Biopharma Product Pipeline. ASM Microbe, San
68 Bulpitt D, Potter CE, Jaderberg M. A large scale, general practice Francisco, USA, 2019.
based in-vestigation into the clinical efficacy and tolerability of 86 Weiss W. Efficacy of Cefpodoxime Proxetil and ETX0282 in a
cefuroxime axetil in women with uncomplicated urinary tract infection. Murine UTI Model with Escherichia coli and Klebsiella pneumoniae.
Curr Med Res Opin 1991; 12: 318–24. European Congress of Clinical Microbiology and Infectious Diseases,
69 Leigh AP, Nemeth MA, Keyserling CH et al. Cefdinir versus cefaclor Amsterdam, The Netherlands, 2019. Abstract P1991.
in the treatment of uncomplicated urinary tract infection. Clin Ther 2000; 87 Mcleod S. The Novel b-Lactamase Inhibitor ETX1317 Effectively Restores the
22: 818–25. Activity of Cefpodoxime against Recent Global Enterobacteriaceae Isolates from
70 Hooton TM, Roberts PL, Stapleton AE. Cefpodoxime vs ciprofloxacin Urinary Tract Infections. European Congress of Clinical Microbiology and Infectious
for short-course treatment of acute uncomplicated cystitis: a randomized Diseases, Amsterdam, The Netherlands, 2019. Abstract P1184.
trial. JAMA 2012; 307: 583–9. 88 Hamrick J. Selection of Ceftibuten as the Partner Antibiotic for the
71 Kavatha D, Giamarellou H, Alexiou Z et al. Cefpodoxime-proxetil versus Oral b-Lactamase Inhibitor VNRX-7145. European Congress of Clinical
trimethoprim-sulfamethoxazole for short-term therapy of uncomplicated acute Microbiology and Infectious Diseases, Amsterdam, The Netherlands,
cystitis in women. Antimicrob Agents Chemother 2003; 47: 897–900. 2019. Abstract P1181.
72 Ho MW, Wang FD, Fung CP et al. Comparative study of ceftibuten 89 Mendes RE. In Vitro Activity of the Orally Available Ceftibuten/VNRX-
and cef-ixime in the treatment of complicated urinary tract infections. J 7145 Combination against a Challenge Set of Enterobacteriaceae Pathogens
Microbiol Immunol Infect 2001; 34: 185–9. Carrying Molecularly Characterised b-Lactamase Genes. European Congress
73 Livermore DM, Hope R, Mushtaq S et al. Orthodox and unorthodox of Clinical Microbiology and Infectious Diseases, Amsterdam, The
clavu-lanate combinations against extended-spectrum b-lactamase Netherlands, 2019. Abstract P1180.
producers. Clin Microbiol Infect 2008; 14 Suppl 1: 189–93. 90 Avery L. In Vivo Pharmacodynamics of VNRX-7145 in the Neutropenic Murine
74 Gupta H, Gupta R, Rai S et al. Is empirical use of the antibiotic Thigh Infection Model When Administered in Combination with Humanized
combination of cefuroxime and clavulanic acid rational? J Glob Exposures of Twice Daily Ceftibuten (CTB) against Serine b-Lactamase-Producing
Antimicrob Resist 2019; 16: 150–1. Enterobacteriaceae (SBL-EB). IDWeek, Washington, DC, USA, 2019.

75 Pal RB, Pal P, Jain S et al. In vitro study to compare sensitivity of 91 Grupper M, Stainton SM, Nicolau DP et al. In vitro pharmacodynamics of
amoxicil-lin!clavulanic acid and cefpodoxime!clavulanic acid among b- a novel ceftibuten-clavulanate combination antibiotic against
lactamase positive clinical isolates of Gram-positive and Gram-negative Enterobacteriaceae. Antimicrob Agents Chemother 2019; 63: e00144–19.
pathogens. J Indian Med Assoc 2008; 106: 545–8. 92 Abdelraouf K, Stainton SM, Nicolau DP. In vivo pharmacodynamic
76 Campbell JD, Lewis JS 2nd, McElmeel ML et al. Detection of favorable profile of ceftibuten-clavulanate combination against extended-spectrum-
oral cephalosporin-clavulanate interactions by in vitro disk approximation b-lacta-mase-producing Enterobacteriaceae in the murine thigh infection
suscep-tibility testing of extended-spectrum-b-lactamase-producing members model. Antimicrob Agents Chemother 2019; 63: e00145–19.
of the Enterobacteriaceae. J Clin Microbiol 2012; 50: 1023–6. 93 ClinicalTrials.gov. A Study to Evaluate the Safety, Tolerability, and
77 Mischnik A, Baumert P, Hamprecht A et al. Susceptibility to Pharmacokinetics (PK, the Measure of How the Human Body Processes
cephalosporin combinations and aztreonam/avibactam among third- a Substance) of ETX0282 When Administered Orally to Healthy
generation cephalo-sporin-resistant Enterobacteriaceae recovered on Participants. https://clinicaltrials.gov/ct2/show/NCT03491748?
hospital admission. Int J Antimicrob Agents 2017; 49: 239–42. cond=ETX0282&rank=1.
78 Bingen E, Bidet P, Birgy A et al. In vitro interaction between cefixime and 94 Jones RN, Sader HS. Update on the cefdinir spectrum and potency
amoxicillin-clavulanate against extended-spectrum-b-lactamase-producing against pathogens isolated from uncomplicated skin and soft tissue
Escherichia coli causing urinary tract infection. J Clin Microbiol 2012; 50: 2540–1. infec-tions in North America: are we evaluating the orally administered
79 Rawat D, Hasan AS, Capoor MR et al. In vitro evaluation of a new cephalo-sporins correctly? Diagn Microbiol Infect Dis 2006; 55: 351–6.
cefixime-clavulanic acid combination for gram-negative bacteria. 95 Sader HS, Streit JM, Fritsche TR et al. Potency and spectrum
Southeast Asian J Trop Med Public Health 2009; 40: 131–9. reevaluation of cefdinir tested against pathogens causing skin and soft
80 Prakash V, Lewis JS 2nd, Herrera ML et al. Oral and parenteral tissue infections: a sample of North American isolates. Diagn Microbiol
therapeutic options for outpatient urinary infections caused by Infect Dis 2004; 49: 283–7.
Enterobacteriaceae pro-ducing CTX-M extended-spectrum b- 96 Preston DA, Turik M. Cefaclor: a contemporary look at susceptibility of
lactamases. Antimicrob Agents Chemother 2009; 53: 1278–80. key pathogens from around the globe. J Chemother 1998; 10: 195–202.

9 of 10
Review

97 Knapp CC, Sierra-Madero J, Washington JA. Antibacterial activities 103 Jacoby GA, Carreras I. Activities of b-lactam antibiotics against
of cef-podoxime, cefixime, and ceftriaxone. Antimicrob Agents Escherichia coli strains producing extended-spectrum b-lactamases.
Chemother 1988; 32: 1896–8. Antimicrob Agents Chemother 1990; 34: 858–62.

Downloaded from https://academic.oup.com/jac/advance-article-abstract/doi/10.1093/jac/dkaa183/5842233 by Uppsala Universitetsbibliotek user on 23 May 2020


98 Nakamura T, Komatsu M, Yamasaki K et al. Susceptibility of various oral 104 Bedenic B, Vranes J, Suto S et al. Bactericidal activity of oral b-
antibacterial agents against extended spectrum b-lactamase producing Escherichia lactam antibiotics in plasma and urine versus isogenic Escherichia coli
coli and Klebsiella pneumoniae. J Infect Chemother 2014; 20: 48–51. strains produc-ing broad- and extended-spectrum b-lactamases. Int J
Antimicrob Agents 2005; 25: 479–87.
99 Hackel M. Impact of Variations in Susceptibility Testing Parameters
on the in Vitro Activity of Ceftibuten in Combination with VNRX-7145. 105 Briggs BM, Jones RN, Erwin ME et al. In vitro activity evaluations
ASM Microbe, San Francisco, USA, 2019. of cefdi-nir (FK482, CI-983, and PD134393): a novel orally administered
100 Durand-Reville TF, Guler S, Comita-Prevoir J et al. ETX2514 is a broad- cephalo-sporin. Diagn Microbiol Infect Dis 1991; 14: 425–34.
spectrum b-lactamase inhibitor for the treatment of drug-resistant Gram-nega-tive 106 Jones RN. Antimicrobial activity and spectrum of ceftibuten (7432-
bacteria including Acinetobacter baumannii. Nat Microbiol 2017; 2: 17104. S, SCH 39720)—a review of United States and Canadian results. Diagn
Microbiol Infect Dis 1991; 14: 37–43.
101 John KJ. Rescue of Ceftibuten Activity by the Oral b-Lactamase
Inhibitor VNRX-7145 against Enterobacteriaceae Expressing Class A, C 107 Neu HC, Saha G, Chin NX. Comparative in vitro activity and b-
and/or D b-Lactamases ASM Microbe, San Francisco, USA, 2019. lactamase stability of FK482, a new oral cephalosporin. Antimicrob
102 Jones RN, Barry AL. Antimicrobial activity and disk diffusion susceptibility Agents Chemother 1989; 33: 1795–800.
testing of U-76,253A (R-3746), the active metabolite of the new cephalosporin
ester, U-76,252 (CS-807). Antimicrob Agents Chemother 1988; 32: 443–9.

10 of 10

You might also like