You are on page 1of 22

b-Lactams and b-Lactamase Inhibitors:

An Overview

Karen Bush1 and Patricia A. Bradford2


1
Molecular and Cellular Biochemistry, Indiana University, Bloomington, Indiana 47405
2
AstraZeneca Pharmaceuticals, Waltham, Massachusetts 02451
Correspondence: karbush@indiana.edu

b-Lactams are the most widely used class of antibiotics. Since the discovery of benzylpeni-
cillin in the 1920s, thousands of new penicillin derivatives and related b-lactam classes of
cephalosporins, cephamycins, monobactams, and carbapenems have been discovered.
Each new class of b-lactam has been developed either to increase the spectrum of activity
to include additional bacterial species or to address specific resistance mechanisms that
have arisen in the targeted bacterial population. Resistance to b-lactams is primarily
because of bacterially produced b-lactamase enzymes that hydrolyze the b-lactam ring,
thereby inactivating the drug. The newest effort to circumvent resistance is the development
of novel broad-spectrum b-lactamase inhibitors that work against many problematic b-lac-
tamases, including cephalosporinases and serine-based carbapenemases, which severely
www.perspectivesinmedicine.org

limit therapeutic options. This work provides a comprehensive overview of b-lactam anti-
biotics that are currently in use, as well as a look ahead to several new compounds that are
in the development pipeline.

hen Alexander Fleming was searching for a consortium of scientists from England and the
W an antistaphylococcal bacteriophage in
his laboratory in the 1920s, he deliberately left
United States were able to optimize the isolation
and identification of benzylpenicillin to assist in
plates out on the bench to capture airborne the treatment of Allied soldiers in World War II
agents that might also serve to kill staphylococci (Macfarlane 1979). These activities set the stage
(Fleming 1929). His success was greater than he for the launch of the most successful class of
must have hoped for. His initial publication on antibiotics in history.
benzylpenicillin described a substance that was b-Lactam antibiotics are currently the most
unstable in aqueous solution but that might used class of antibacterial agents in the in-
serve as an antiseptic or as a selective agent for fectious disease armamentarium. As shown in
isolation of Gram-negative bacteria that were Figure 1, b-lactams account for 65% of all
present in mixed cultures of staphylococci and prescriptions for injectable antibiotics in the
streptococci. As the potential utility of penicillin United States. Of the b-lactams, cephalosporins
G as a parenteral therapeutic agent became comprise nearly half of the prescriptions (Table
more obvious, Fleming, Abraham, Florey, and 1). The b-lactams are well tolerated, efficacious,

Editors: Lynn L. Silver and Karen Bush


Additional Perspectives on Antibiotics and Antibiotic Resistance available at www.perspectivesinmedicine.org
Copyright # 2016 Cold Spring Harbor Laboratory Press; all rights reserved; doi: 10.1101/cshperspect.a025247
Cite this article as Cold Spring Harb Perspect Med 2016;6:a025247

1
K. Bush and P.A. Bradford

Tetracyclines
Trimethoprim/sulfa
Macrolides/ketolides
Fluoroquinolones
Aminoglycosides
Glycopeptides
Polymyxins
All other antibacterials
β-Lactams

Figure 1. Proportion of prescriptions in the United States for injectable antibiotics by class for years 2004– 2014.
The percentage of standard units for each injectable antibiotic prescribed in the United States from 2004 to 2014
is shown as follows: b-lactams, 65.24%; glycopeptides, 9%; fluoroquinolones, 8%; macrolides/ketolides, 6%;
aminoglycosides, 5%; polymyxins, 1%; trimethoprim/sulfamethoxazole, 0.5%; tetracyclines (excluding tigecy-
cline), 0.4%; all other antibiotics (including daptomycin, linezolid, and tigecycline), 4.21%. (Data from the IMS
MDART Quarterly Database on file at AstraZeneca.)
www.perspectivesinmedicine.org

and widely prescribed. Their major toxicity is by b-lactams is perceived to be a major advan-
related to an allergic response in a small per- tage in the treatment of serious infections.
centage of patients who react to related side When these agents were threatened by the rapid
chain determinants; notably, these reactions emergence of b-lactamases, b-lactamase-stable
are most common with penicillins and cepha- agents were developed, as well as potent b-lac-
losporins with minimal reactivity caused by tamase inhibitors (BLIs). In this introductory
monobactams (Saxon et al. 1984; Moss et al. description of the b-lactams, the most com-
1991). The bactericidal mechanism of killing monly available b-lactams and BLIs will be pre-
sented, with a brief summary of their general
characteristics. Occasional agents have been in-
Table 1. Usage of parenteral b-lactams by class from cluded for their historical or scientific impor-
2004– 2104 in the United States tance. Note that resistance mechanisms will
Percentage of be discussed in detail in other articles in this
Class of b-lactam prescriptionsa collection.
Narrow spectrum penicillins 3.12
Broad spectrum penicillinsb 36.54
MECHANISM OF ACTION
Cephalosporins 47.49
Monobactams 1.66 b-Lactam antibiotics are bactericidal agents
Carbapenems 11.20 that interrupt bacterial cell-wall formation as
a
The percentage for each injectable antibiotic class a result of covalent binding to essential penicil-
prescribed in the United States from 2004 to 2014. (Data lin-binding proteins (PBPs), enzymes that are
from the IMS MDART Quarterly Database on file at
involved in the terminal steps of peptidoglycan
AstraZeneca.)
b
Broad-spectrum penicillins include the b-lactam/ cross-linking in both Gram-negative and Gram-
b-lactam-inhibitor combinations piperacillin-tazobactam, positive bacteria. Every bacterial species has its
ticarcillin-clavulanate, and ampicillin-sulbactam. own distinctive set of PBPs that can range from

2 Cite this article as Cold Spring Harb Perspect Med 2016;6:a025247


b-Lactams and b-Lactamase Inhibitors

three to eight enzymes per species (Georgopa- PENICILLINS


padakou and Liu 1980). The inhibition of bac-
terial peptidoglycan transpeptidation by penicil- Penicillin G (benzylpenicillin) was the first b-
lin was described mechanistically in a classical lactam to be used clinically, most frequently to
paper by Tipper and Strominger (1965), who treat streptococcal infections for which it had
noted a structural similarity of penicillin G to high potency (Rammelkamp and Keefer 1943;
the terminal D-Ala-D-Ala dipeptide of the na- Hirsh and Dowling 1946). Another naturally
scent peptidoglycan in the dividing bacterial occurring penicillin, penicillin V ( phenoxy-
cell. This mechanism is now known to involve methylpenicillin), in an oral formulation is still
binding of penicillin, or another b-lactam, to an used therapeutically and prophylactically for
active site serine found in all functional PBPs mild to moderate infections caused by suscep-
(Georgopapadakou et al. 1977). The resulting tible Streptococcus spp., including use in pedi-
inactive acyl enzyme may then slowly hydrolyze atric patients (Pottegard et al. 2015). However,
the antibiotic to form a microbiologically inac- the selection of penicillin-resistant penicillin-
tive entity (Frère and Joris 1985). In addition to ase-producing staphylococci in patients treated
these functionalities, recent work has shown with penicillin G led to decreased use of this
the binding of selected b-lactams, such as cef- agent, and prompted the search for more pen-
taroline, to an allosteric site in PBP2a from icillins with greater stability to the staphylococ-
Staphylococcus aureus, resulting in an increased cal b-lactamases (Kirby 1944, 1945; Medeiros
sensitization of the organism to the antibiotic 1984). A list of historically important and clin-
(Otero et al. 2013; Gonzales et al. 2015). ically useful penicillins is provided in Table 2.
PBPs may be divided into classes according Among the penicillinase-stable penicillins of
to molecular mass (Goffin and Ghuysen 1998; clinical significance are methicillin, oxacillin,
www.perspectivesinmedicine.org

Massova and Mobashery 1998), with low-mo- cloxacillin, and nafcillin, with the latter suggest-
lecular-mass PBPs serving mainly as mono- ed as the b-lactam of choice for skin infections,
functional D-Ala-D-Ala carboxypeptidases. catheter infections, and bacteremia caused by
High-molecular-mass PBPs have been divided methicillin-susceptible S. aureus (Bamberger
into two subclasses, one of which (class A) in- and Boyd 2005). All were used primarily for
cludes bifunctional enzymes with both a trans- staphylococcal infections until the emergence
peptidase and a transglycosylase domain, and of methicillin-resistant S. aureus (MRSA) in
the second of which (class B) encompasses D- 1979 –1980 (Hemmer et al. 1979; Saroglou et
Ala-D-Ala-dependent transpeptidases. At least al. 1980).
one PBP is deemed to be essential in each spe- Penicillins with improved activity against
cies, with a unique specificity for b-lactam Gram-negative pathogens included the orally
binding that varies among each species and bioavailable ampicillin and amoxicillin, both
each b-lactam class (Curtis et al. 1979; Georgo- of which were introduced in the 1970s. These
papadakou and Liu 1980). In Gram-negative agents were initially used for the treatment of
bacteria, essential PBPs include the high-molec- infections caused by Enterobacteriaceae and did
ular-weight PBPs 1a and 1b that are involved not effectively inhibit the growth of Pseudomo-
in cell lysis, PBP2, the inhibition of which re- nas aeruginosa, which became more of a con-
sults in a cessation of cell division and the for- cern during the late 1970s. Carbenicillin was
mation of spherical cells, and PBP3 for which the first antipseudomonal penicillin to be in-
inhibition arrests cell division, resulting in fila- troduced, but lacked stability to b-lactamase
mentation. Cell death may occur as a result of hydrolysis and was less potent than piperacillin
inhibiting one or more of these PBPs (Spratt or ticarcillin, later antipseudomonal penicil-
1977, 1983). The roles of PBPs in Gram-positive lins. These latter drugs were considered to be
bacteria and Mycobacterium tuberculosis are potent broad-spectrum penicillins that includ-
discussed in detail in Fisher and Mobashery ed penicillin-susceptible staphylococci, enteric
(2016). bacteria, anaerobes, and P. aeruginosa in their

Cite this article as Cold Spring Harb Perspect Med 2016;6:a025247 3


www.perspectivesinmedicine.org
4

K. Bush and P.A. Bradford


Table 2. Penicillins of current and historical utility

O R1
R2 C HN
S CH3
N CH3
O CO2H

Route of
Name R1 R2 administration Approval dateb,c Status
Benzylpenicillin ( penicillin G) —H IM or IV 1946 Approved worldwide
H2C

Phenoxymethylpenicillin —H Oral 1968 Approved worldwide


O CH2
( penicillin V)
Methicillin —H OCH3 IV 1960 No longer available; of historical interest
Cite this article as Cold Spring Harb Perspect Med 2016;6:a025247

OCH3
Oxacillin —H Oral, IV 1962 Widely available, but not in the United
Kingdom

N
O CH3
Cloxacillin —H Cl Oral, IV 1974 Widely available, but not in the United
Kingdom

N
O CH3
Ampicillin —H NH2 Oral, IV 1963 Widely available

Continued
www.perspectivesinmedicine.org

Table 2. Continued
Cite this article as Cold Spring Harb Perspect Med 2016;6:a025247

Route of
Name R1 R2 administration Approval dateb,c Status
Nafcillin —H IV 1970 Limited availability

Amoxicillin —H NH2 Oral, IV 1972 Widely available

HO
Carbenicillin —H H Oral 1972 Discontinued
C
O
HO
Ticarcillin —H H IV 1976 Limited availability
C
O
S HO
Piperacillin —H IV 1981 Widely available, primarily in
O combination with tazobactam
N
N N
H

b-Lactams and b-Lactamase Inhibitors


O
O
Temocillin —OCH3 H IV 1985 in Europe Limited availability (Europe)
C (Harvengt 1985)
O
S HO
Mecillinam IV 1978 Limited availability
N
N
S CH3
N CH3
O CO2H
IM, Intramuscular; IV, intravenous.
a
FDA approval unless otherwise noted.
5

b
Dates were updated from Medeiros (1997) (www.accessdata.fda.gov/scripts/cder/drugsatfda; www.drugs.com).
K. Bush and P.A. Bradford

spectrum of activity. They were used extensively CEPHALOSPORINS


to treat serious nosocomial infections, especial-
ly when combined with a b-lactamase inhibitor During the 1950s, the discovery of the naturally
(see below). occurring penicillinase-stable cephalosporin C
Two parenteral penicillins with unusual opened a new pathway to the development of
chemical structures, mecillinam and temocillin hundreds of novel cephalosporins (Newton and
(Table 2), were introduced to treat infections Abraham 1956; Abraham 1987) to treat infec-
caused by enteric bacteria before the global tions caused by the major penicillinase-pro-
emergence of extended-spectrum b-lactamases ducing pathogen of medical interest at that
(ESBLs) in the late 1980s. Mecillinam (also time, S. aureus. Dozens of cephalosporins were
known as amdinocillin), with a 6-b-amidino introduced into clinical practice (Abraham
side chain, is a narrow-spectrum b-lactam that 1987), either as parenteral or oral agents. The
binds exclusively to PBP2 in enteric bacteria molecules exhibited antibacterial activity with
(Curtis et al. 1979). Because of this specificity, MICs often 4 mg/mL against not only staph-
it shows synergy in vitro in combination with ylococci, but also Streptococcus pneumoniae and
other b-lactams that bind to PBPs 1a/1b and/ non-b-lactamase-producing enteric bacteria.
or PBP3 in Gram-negative bacteria (Hanberger The parenteral agents were generally eightfold
et al. 1991), thus decreasing the possibility that more potent than the oral agents that were used
a point mutation in a single PBP would lead in some cases to replace oral penicillins in pen-
to resistance (Hickman et al. 2014). Temocillin, icillin-allergic patients. The early cephalospo-
the 6-a-methoxypenicillin analog of ticarcillin, rins, for example, those in the cephalosporin
had greater stability than ticarcillin to hydrolysis I subclass (Bryskier et al. 1994) introduced
by serine b-lactamases, but lost antibacterial before 1980, were labile to hydrolysis by many
www.perspectivesinmedicine.org

activity against Gram-positive bacteria, anaero- b-lactamases that emerged following their in-
bic Gram-negative pathogens, and some enteric troduction into clinical practice, so that only a
bacteria that included the important pathogens few of the early molecules remain in use (see
Enterobacter spp. and Serratia marcescens (Mar- Table 3), primarily to treat mild to moderate
tinez-Beltran et al. 1985). Mecillinam and te- skin infections caused by methicillin-suscepti-
mocillin are currently enjoying a resurgence in ble S. aureus (MSSA) (Giordano et al. 2006).
interest owing to their stability to many ESBLs Cefazolin with high biliary concentrations is
(Livermore et al. 2006; Rodriguez-Villalobos still used for surgical prophylaxis and for treat-
et al. 2006), often resulting in greater than ment of abdominal infections (Sudo et al. 2014)
90% susceptibility when tested against many and is effective as empiric therapy in 80% of
contemporary ESBL-producing Enterobacter- Japanese children with their first upper urinary
iaceae (Giske 2015; Zykov et al. 2016). tract infection (Abe et al. 2016).
Because increasing numbers of b-lactamas- When the TEM-1 penicillinase began to ap-
es have compromised the use of penicillins as pear on transmissible plasmids in Neisseria gon-
single agents (Bush 2013), there is currently orrhoeae (Ashford et al. 1976) and Haemophilus
limited therapeutic use of the penicillins as influenzae (Gunn et al. 1974; Khan et al. 1974),
monotherapy. Ampicillin, amoxicillin, pipera- it was quickly recognized that the penicillins
cillin, and ticarcillin have continued to be use- and cephalosporins in medical use were becom-
ful, primarily as a result of their combination ing ineffective, not only in treating those TEM-
with an appropriate b-lactamase inhibitor (see 1-producing organisms, but also for the enteric
below). However, even ampicillin, amoxicillin, bacteria and P. aeruginosa that could all acquire
penicillin G, and penicillin V are still active this enzyme. Another surge of synthetic activ-
as monotherapy against Group A streptococci, ity in the pharmaceutical industry provided
and Treponema pallidum, two of the few bacte- both oral and parenteral cephalosporins with
rial species that do not produce b-lactamases stability to this common enzyme. These agents
(Schaar et al. 2014). tended to have decreased potency against the

6 Cite this article as Cold Spring Harb Perspect Med 2016;6:a025247


www.perspectivesinmedicine.org

Table 3. Cephalosporins of current clinical utility or of historical interest

HN R2 S
R3

O N
O R1

O OH
Route of
Name Subclassa R1 R2 R3 administration Approval dateb,c Status
Cephalexin Cephalosporin I H –H – CH3 Oral 1971 Limited availability
C
NH2
Cefaclor Cephalosporin I – Cl –H NH2 Oral 1979 Widely available

Cefixime Cephalosporin V –H OH Oral 1989 Widely available

N O
H2N N

S
Cefpodoxime Cephalosporin IV O –H N Oral 1992 Widely available
CH3
H2N N
S
O
CH3

Ceftibutin Cephalosporin III –H –H O Oral 1995 Widely available


N
H2N OH
S
Continued
www.perspectivesinmedicine.org

Table 3. Continued
Route of
Name Subclassa R1 R2 R3 administration Approval dateb,c Status
Cefdinir Cephalosporin V –H HO Oral 1997 Widely available
N
H2N N

S
Cefazolin Cephalosporin I S S –H N IV 1973 Widely available
N N
N N N

Cefuroxime Cephalosporin II O NH2 –H H 3C O Oral,d IV 1983 Widely available


N
O
O
Cefotaxime Cephalosporin III O CH3 –H CH3 IV 1981 Widely available
O
O N
H2 N N

S
Cefoperazone Cephalosporin III –H OH IV 1982 Widely available
S N
N
N N O
N
N N
H
O
O
Ceftriaxone Cephalosporin III S N O –H CH3 IV 1984 Widely available
O
N N
N O
H H2 N N

S
Continued
www.perspectivesinmedicine.org

Table 3. Continued
Route of
Name Subclassa R1 R2 R3 administration Approval dateb,c Status
Ceftazidime Cephalosporin III –H OH IV 1985 Widely available
N
+
H3C O
C CH3
O
N
H2N N

S
Cefepime Cephalosporin IV –H CH3 IV 1996 Widely available
+
N O
N
H2 N N

S
Ceftaroline Anti-MRSA Me –H OCH2CH3 IV 2010 Widely available
+ N
(fosamil) cephalosporin N
H
N N
H2 O3 P
S N
N

S S

Ceftobiprole Anti-MRSA NH –H OH IV 2013 (Europe) Limited availability


cephalosporin N N
H2N N
O
S N
Ceftolozane Antipseudomonal NH2 –H CH3 IV 2014 Limited availability
cephalosporin – H3C CO2H
HN
cephalosporin VI
HN N O
O H2 N N

NH2 S
N N
+ N
Me
Continued
www.perspectivesinmedicine.org

Table 3. Continued
Route of
Name Subclassa R1 R2 R3 administration Approval dateb,c Status
S-649266 Siderophore O Cl –H OH IV Not approved Phase 2
cephalosporin – + H3C O
N OH
cephalosporin V N C CH3
H O
OH N
H2N N

Cefoxitin Cephamycin O NH2 –OCH3 IV 1978 Widely available


S
O
Moxalactam Oxacephem HO IV 1982e Limited availability

O
OCH3
HO O
HN
O N
N S
O N
N N
O OH
IM, Intramuscular; IV, intravenous.
a
Subclasses assigned according to CLSI (2016), Bryskier et al. (1994), or Bryskier and Belfiglio (1999).
b
FDA approved unless otherwise noted.
c
Dates were updated from Medeiros (1997) (www.accessdata.fda.gov/scripts/cder/drugsatfda; www.drugs.com; www.price-rx.com/lists/lantibiotics.shtml).
d
Oral when dosed as cefuroxime axetil.
e
Anonymous (1982).
b-Lactams and b-Lactamase Inhibitors

staphylococci, but gained antibacterial activity ished activity against staphylococci and entero-
against Gram-negative pathogens. Cefuroxime, cocci compared to earlier cephalosporins, but
dosed parenterally or orally as the axetil ester, have more potent activity against Gram-nega-
was the only member of the cephalosporin II tive organisms. Cefepime tends to have lower
class (Bryskier et al. 1994) with both oral and MICs against enteric bacteria than the other
systemic dosage forms, but its stability to b-lac- expanded-spectrum cephalosporins, attributed
tamase hydrolysis was diminished compared to to greater penetration through the OmpF outer-
later oral cephalosporins (Jacoby and Carreras membrane porin protein (Nikaido et al. 1990;
1990). As seen with cefuroxime, acceptable oral Bellido et al. 1991). Cefotaxime and ceftriaxone
bioavailability of cefpodoxime required esterifi- are often used to treat susceptible streptococcal
cation through addition of a proxetil group to infections; all can be used to treat serious infec-
attain sufficient absorption for efficacy (Brys- tions caused by enteric bacteria if the organisms
kier and Belfiglio 1999). Of the oral agents test susceptible. Notably, ceftazidime and cefe-
approved after 1983 in Table 3, cefdinir was pime have maintained their observed activity
generally more stable to hydrolysis, not only against P. aeruginosa, with recent susceptibility
to the original TEM enzyme, but also to the rates exceeding 80% (Sader et al. 2015). A lia-
AmpC cephalosporinases that are produced at bility of the expanded-spectrum cephalospo-
a basal level in many enteric bacteria and rins, however, began to emerge only a few years
P. aeruginosa (Payne and Amyes 1993; Labia after the introduction of cefotaxime, when the
and Morand 1994). ESBLs were identified with the ability to hydro-
Among the parenteral agents introduced in lyze all of the b-lactams, with the exception of
the 1980s were the cephamycin cefoxitin, and the carbapenems. These enzymes, in addition to
cephalosporins in the cephalosporin III and both serine and metallo-carbapenemases, have
www.perspectivesinmedicine.org

cephalosporin IV subclasses (Bryskier et al. severely compromised the activity of almost all
1994), which continue to serve as important penicillins and cephalosporins, necessitating
antibiotics for the treatment of serious infec- the development of combination therapy with
tions caused by Gram-negative pathogens. The other b-lactams, b-lactamase inhibitors, or an-
novel oxacephem moxalactam, or latamoxef, tibiotics from other classes.
which had similar antimicrobial activity to the Ceftolozane, recently approved in combi-
cephalosporin III/IV subclasses, has exquisite nation with tazobactam for the treatment of
stability to hydrolysis by b-lactamases (Sato complicated urinary tract infections and com-
et al. 2015), but was not a highly successful an- plicated intraabdominal infections, shows po-
tibiotic owing, in part, to a relatively high fre- tent antipseudomonal activity, and includes
quency of bleeding in patients treated with this activity against enteric bacteria that produce
drug (Brown et al. 1986). The cephamycin ce- some ESBLs (Zhanel et al. 2014), particularly
foxitin is notable for its characteristic 7-me- CTX-M-producing isolates (Estabrook et al.
thoxy side chain that confers stability to the 2014). Another recent addition to the cephalo-
TEM-type b-lactamases, including ESBLs. It sporin family is the siderophore-substituted
has useful antibacterial activity against MSSA cephalosporin S-649266 with a catechol in the
and enteric bacteria that do not produce high 3-position, thus allowing the molecule to enter
levels of AmpC cephalosporinases (Jacoby and the cells via an iron transport mechanism (Ko-
Han 1996). Cefotaxime, cefoperazone, ceftriax- hira et al. 2015). In addition to increased pen-
one, and ceftazidime, designated as subclass etrability, the cephalosporin is stable to hydro-
cephalosporin III, and cefepime in the cepha- lysis by many carbapenemases, resulting in
losporin IV subclass, are also known as expand- activity against many b-lactam-resistant enteric
ed-spectrum cephalosporins with increased bacteria (Kohira et al. 2015).
hydrolytic stability to the common penicillinas- In the mid-1990s, reports began to emerge
es, SHV-1 and TEM-1 b-lactamase (Martinez- describing cephalosporins with MICs ,4 mg/
Martinez et al. 1996). These agents have dimin- mL against MRSA (Hanaki et al. 1995) as a

Cite this article as Cold Spring Harb Perspect Med 2016;6:a025247 11


K. Bush and P.A. Bradford

result of targeted binding to PBP2a. PBP2a is an general, bind strongly to PBP2 in Gram-nega-
acquired low-affinity PBP responsible for the tive bacteria, but may also bind to PBP1a, 1b,
observed lack of antibacterial activity of most and 3, thus providing supplemental killing
b-lactams in MRSA isolates. Ceftobiprole (Ha- mechanisms that may serve to lessen the emer-
naki et al. 1995; Hebeisen et al. 2001) and cef- gence of resistance (Sumita and Fukasawa 1995;
taroline (Moisan et al. 2010), two cephalospo- Yang et al. 1995). Carbapenems are notable for
rins with IC50 values ,1 mg/mL for binding to their stability to most b-lactamases (Bonfiglio
the staphylococcal PBP2a, have been developed et al. 2002), with the exception of the emerging
for clinical use (Table 3). Ceftaroline is approx- carbapenemases found primarily in Gram-neg-
imately twofold to fourfold more potent than ative bacteria (Bush 2013). Because of the labil-
ceftobiprole in inhibiting staphylococcal and ity of imipenem to hydrolysis by the human
streptococcal growth (Karlowsky et al. 2011), renal dehydropeptidase (DHP) causing inacti-
but ceftobiprole is up to fourfold more potent vation of the drug (Kropp et al. 1982), it is dosed
against Enterococcus faecalis (Karlowsky et al. in combination with cilastatin, a DHP inhibitor
2011). Ceftobiprole generally has at least four- that also acts as a nephroprotectant (Kahan et
fold to eightfold lower MICs than ceftaroline al. 1983).
against enteric bacteria, P. aeruginosa, and Aci- Based on the potent broad-spectrum activ-
netobacter spp. (Pillar et al. 2008; Karlowsky ity of the early carbapenems, other related
et al. 2011). Neither cephalosporin is stable to agents, including meropenem, ertapenem, and
hydrolysis by ESBLs or carbapenemases (Pillar doripenem, have been developed for global use,
et al. 2008; Castanheira et al. 2012), although with generally the same group of organisms in-
the combination of ceftaroline with the b-lac- cluded in their activity spectrum (Baughman
tamase inhibitor avibactam overcomes many of 2009). All these carbapenems are more stable
www.perspectivesinmedicine.org

these issues (Mushtaq et al. 2010; Flamm et al. chemically than imipenem, thus allowing for a
2014) (see below). Both drugs are highly insol- longer shelf life for the formulated drug and the
uble and have been derivatized as prodrugs for potential for prolonged infusion times (Cie-
therapeutic use, as ceftaroline fosamil (Talbot lecka-Piontek et al. 2008; Prescott et al. 2011).
et al. 2007) and ceftobiprole medocaril (He- Like imipenem, they are stable to most b-lacta-
beisen et al. 2001), respectively. mases, other than the carbapenemases (Bush
2013). Following the introduction of imipen-
em, later carbapenems contained a 1b-methyl
CARBAPENEMS
group that conferred stability to the human
Thienamycin was identified in the mid-1970s DHP, thus negating the necessity for coadmin-
as a potent broad-spectrum antibiotic with istration of an inhibitor such as cilastatin (Zha-
the typical four-membered b-lactam structure nel et al. 2007). In terms of antibacterial activity,
fused to a novel five-membered ring in which meropenem is generally twofold to fourfold
carbon rather than sulfur was present at the more potent that imipenem against enteric bac-
1-position (Kahan et al. 1979). Because of its teria (Jorgensen et al. 1991), is similar in poten-
chemical instability, this carbapenem was never cy against P. aeruginosa, but may have twofold
developed as a therapeutic agent, but was stabi- to eightfold less antibacterial activity against
lized by adding the N-formimidoyl group to Gram-positive bacteria (Neu et al. 1989). In
the 2-position, resulting in imipenem (Table addition, meropenem and doripenem retain
4). Imipenem has been widely used for infec- greater activity against isolates of P. aeruginosa
tions caused by Gram-positive, Gram-negative, lacking the outer membrane porin protein
nonfermentative, and anaerobic bacteria based OprD than imipenem (Riera et al. 2011). Mer-
on its sustained high activity against these or- openem is the only carbapenem approved for
ganisms, particularly among non-carbapene- use in meningitis because of its excellent pene-
mase-producing enteric bacteria (Bradley et al. tration into the meninges (Dagan et al. 1994).
1999; Kiratisin et al. 2012). Carbapenems, in Doripenem, a carbapenem with somewhat

12 Cite this article as Cold Spring Harb Perspect Med 2016;6:a025247


b-Lactams and b-Lactamase Inhibitors

Table 4. Carbapenems of current clinical utility


OH
R1

R2
N
O
OH
O
Name R1 R2 Approval datea,b Status
Imipenem H H NH 1985 Widely available
N
H
S
Meropenem CH3 H O 1996 Widely available
N
N

S
Ertapenem CH3 CO2H 2001 Widely available

O
N
H
NH
S
Doripenem CH3 H O O 2007 Widely available
N
www.perspectivesinmedicine.org

N S NH2
H
S
Biapenem CH3 N 2001 (Japan) Available in Japan
N +
N

S
Tebipenemc CH3 2009 (Japan) Available in Japan
S
N
N

S
a
FDA approved unless otherwise noted.
b
Dates were updated from Medeiros (1997) (www.accessdata.fda.gov/scripts/cder/drugsatfda; www.drugs.com; adisin
sight.springer.com/drugs/800010812).
c
Formulated as the pivoxil ester.

higher chemical stability than imipenem or dosed most commonly two or three times a
meropenem (Prescott et al. 2011), follows the day. Although its antibacterial spectrum is sim-
antibacterial profile of meropenem, but is ilar to the other carbapenems against Entero-
slightly more potent against Gram-negative or- bacteriaceae, ertapenem differs from imipenem,
ganisms (Nordmann et al. 2011). Ertapenem, meropenem, and doripenem in that it has no
recognized for its long elimination half-life in useful activity against P. aeruginosa (Kohler et al.
humans because of its high protein binding 1999). Two carbapenems approved for use only
(95%) (Majumdar et al. 2002), may be effective- in Japan include biapenem, with an antimicro-
ly administered once daily (Kattan et al. 2008) bial spectrum similar to meropenem and dor-
in contrast to the other carbapenems that are ipenem (Neu et al. 1992; Papp-Wallace et al.

Cite this article as Cold Spring Harb Perspect Med 2016;6:a025247 13


K. Bush and P.A. Bradford

2011), and tebipenem, which lacks appreciable bic enteric bacteria and P. aeruginosa, with
antipseudomonal activity (Fujimoto et al. MICs against S. aureus, S. pneumoniae, and
2013) (Table 4). Tebipenem is notable for its E. faecalis 50 mg/mL (Sykes et al. 1982). It
dosing as the pivoxil ester, rendering it orally binds tightly to PBP3 in Gram-negative rods,
bioavailable for use in pediatric respiratory in- with weaker binding to PBP1a, leading to fila-
fections (Kato et al. 2010). Like the other carba- mentation followed by cell lysis (Sykes et al.
penems, they are stable to hydrolysis by most 1982). At the time that it was introduced into
serine b-lactamases, but can be hydrolyzed by clinical practice, aztreonam was stable to hydro-
both serine and metallo-carbapenemases. Bia- lysis by all of the common b-lactamases (Sykes
penem has been reported to have better hydro- et al. 1982); the emergence of ESBLs and the
lytic stability to metallo-b-lactamases (MBLs) serine carbapenemases has since rendered it
compared to imipenem or meropenem (Neu less effective against multidrug-resistant b-
et al. 1992; Inoue et al. 1995; Yang et al. 1995) lactamase-producing organisms (Wang et al.
with at least fourfold lower MICs than imipe- 2014). However, the monobactam nucleus is
nem when tested against organisms producing not a good substrate for hydrolysis by MBLs,
IMP, VIM, or NDM MBLs (Livermore and thus leading to a unique opportunity for this
Mushtaq 2013). monobactam to be used in combination thera-
py with a serine b-lactamase inhibitor to treat
infections caused by multi-b-lactamase-pro-
MONOCYCLIC b-LACTAMS
ducing bacteria (see below) (Wang et al. 2014).
Aztreonam, a monocyclic b-lactam with an BAL30072 is a novel monosulfactam with
N1-sulfonic acid substituent, originated as a de- an N1-O-sulfate group, an activity-enhancing
rivative from a novel antibiotic isolated from 3-dihydropyridone siderophore substituent,
www.perspectivesinmedicine.org

the New Jersey Pine Barrens (Cimarusti and and a 4-gem-dimethyl substitution on the aze-
Sykes 1983) (Table 5), and is the only mono- tidinone ring (Page et al. 2010) (Table 5). Its
bactam to gain regulatory approval for thera- spectrum of activity is similar to aztreonam,
peutic use. It has targeted activity against aero- but supplemented with activity against addi-

Table 5. Monocyclic b-lactams


R2 X

N
O R1

Name Subclass R1 R2 Approval date Status


a
Aztreonam Monobactam —SO3H O 1986 Widely available
X ¼ a-methyl N C C HN
H2N N
S
O
C CH3
H3C
CO2-
BAL30072 Monosulfactam —OSO3H O Not approved Phase 1
X ¼ gem-dimethyl OH

N
N O HO
H2N N H
N
S
O
a
U.S. approval date provided in Medeiros (1997).

14 Cite this article as Cold Spring Harb Perspect Med 2016;6:a025247


b-Lactams and b-Lactamase Inhibitors

tional nonfermentative bacteria. As a result of bination for nosocomial infections that include
the increased penetration of BAL30072 via iron P. aeruginosa as a causative pathogen (Neu
uptake mechanisms, it is more potent against 1990), and with amoxicillin as an orally bio-
some Gram-negative bacteria than other b-lac- available formulation for therapeutic use espe-
tams, with activity against Acinetobacter spp. cially in pediatric populations (Klein 2003). It is
and Burkholderia spp. eightfold to .256-fold also used in phenotypic testing to determine the
better than imipenem (Page et al. 2010). It is presence of ESBLs in Escherichia coli and Kleb-
susceptible to hydrolysis by ESBLs and many siella pneumoniae (Steward et al. 2001).
carbapenemases, and has shown synergistic Following the discovery of clavulanic acid,
activity in combination with b-lactamase in- medicinal chemists synthesized a number of
hibitors (Mushtaq et al. 2013) or meropenem penicillanic acid sulfones (Table 6) with b-lac-
(Hofer et al. 2013; Hornsey et al. 2013). Like tamase inhibitory activity (English et al. 1978;
aztreonam, it is stable to hydrolysis by MBLs; Fisher et al. 1981; Aronoff et al. 1984). Of these,
additionally, it was hydrolyzed 3000-fold less sulbactam (English et al. 1978) and tazobactam
efficiently by the KPC-2 serine carbapenemase (Aronoff et al. 1984) were successfully commer-
compared to aztreonam (Page et al. 2010). cialized. Both had a similar spectrum of activity
as clavulanic acid. Against class A b-lactamases,
sulbactam had less inhibitory activity than clav-
b-LACTAMASE INHIBITORS
ulanic acid or tazobactam based on IC50 values,
Attempts to identify inhibitors of common but both sulfones were better inhibitors of class
b-lactamases began in the mid-1970s, triggered C cephalosporinase b-lactamases (Bush et al.
by the appearance of the transferable TEM-1 1993). Each followed the same general inhibito-
penicillinase in Neisseria gonorrhoeae (Ashford ry/inactivation-mechanism as for clavulanic
www.perspectivesinmedicine.org

et al. 1976) and Haemophilus influenzae (Gunn acid (Easton and Knowles 1984; Bush et al.
et al. 1974; Khan et al. 1974). As the result of 1993). The number of hydrolytic events before
natural product screening, clavulanic acid with inactivation was at least 25-fold higher for sul-
a novel clavam structure (Table 6) was identified bactam than for clavulanic acid or tazobactam
as a broad spectrum inhibitor of the staphylo- for the TEM-2 b-lactamase (Bush et al. 1993;
coccal penicillinases and most of the recognized Easton and Knowles 1984). In contrast to clav-
plasmid-encoded penicillinases found in enter- ulanic acid, the sulfone inhibitors do not func-
ic bacteria (Reading and Cole 1977; Cole 1982), tion as inducers of chromosomally mediated
including the highly prevalent TEM and SHV AmpC b-lactamase (Weber and Sanders 1990).
enzymes (Simpson et al. 1980). The TEM b- Sulbactam has been combined with ampi-
lactamase was shown to be inactivated by this cillin for general global use (Neu 1990) and with
suicide inhibitor that initially acylates the active cefoperazone to provide additional synergistic
site serine with transient inhibition that in- activity against nonfermentative and anaerobic
cludes hydrolysis of the inhibitor before com- bacteria, primarily in Japan (Eliopoulos et al.
plete enzyme inactivation (Charnas et al. 1978; 1989). Tazobactam has been combined with
Charnas and Knowles 1981). The spectrum of piperacillin and, more recently, with cefopera-
the inhibitor is now recognized to include most zone and ceftolozane for nosocomial infections,
class A b-lactamases, including ESBLs (Steward including those caused by P. aeruginosa (Lister
et al. 2001) and, to a lesser extent, serine carba- 2000). In general, none of the inhibitors has
penemases (Nordmann and Poirel 2002; Yigit useful antibacterial activity as monotherapy, al-
et al. 2003). Clavulanic acid acts synergistically though there are several notable exceptions.
with penicillins and cephalosporins against b- Clavulanic acid alone has been reported to
lactamase-producing enteric bacteria to inhibit have an MIC as low as 1 mg/mL against N. gon-
sensitive b-lactamases, thus allowing the com- orrhoeae (Wise et al. 1978); sulbactam has mod-
panion b-lactam to kill the bacteria. It has been est activity against wild-type Acinetobacter spp.
combined with ticarcillin as a parenteral com- and Burkholderia cepacia, with MIC90 values

Cite this article as Cold Spring Harb Perspect Med 2016;6:a025247 15


K. Bush and P.A. Bradford

Table 6. b-lactamase inhibitors of current interest


Partner b- Approval
Name Structure Subclass lactam datea Status
Clavulanic O OH Clavam Amoxicillin 1984 Widely
acidb available
N
O
O OH
Sulbactamc O O Penicillanic Ampicillin 1986 Widely
S acid available
N sulfone
O
O OH
Tazobactam O O Penicillanic Piperacillin 1993 Widely
S N acid Ceftolozane 2014 available
N N
N sulfone Available in
O the United
O OH States and
Europe
Avibactamd O DBOe Ceftazidimed 2015 Widely
available
H2 N
N
N
O OSO3-
www.perspectivesinmedicine.org

Relebactam O DBO Imipenem Not Phase 3 in the


approved United States
HN
H2N+
N
N
O OSO3-
RG6080 O DBO Not selected Not Phase 1
O approved
H2N HN
N
N
O OSO3-
RPX7009 H Boronic acid Meropenem Not Phase 3 in the
S N approved United States
O
O B
HO O OH
a
Dates provided in Medeiros (1997) or www.accessdata.fda.gov/scripts/cder/drugsatfda.
b
Also initially combined with ticarcilllin to provide parenteral activity against Pseudomonas aeruginosa.
c
Also combined with cefoperazone outside the United States.
d
Also in development with ceftaroline or aztreonam.
e
Diazabicyclooctane.

8 and 10 mg/mL, respectively (Jacoby and their modest activity against serine carbapene-
Sutton 1989; Fass et al. 1990), but does not re- mases does not translate into clinical suscepti-
tain activity against isolates with multiple resis- bility (Yigit et al. 2003; Woodford et al. 2004)
tance mechanisms (Dong et al. 2014). None of owing, at least in part, to the presence of mul-
these inhibitors is effective in inhibiting the tiple b-lactamases in the producing organisms
hydrolytic activity of MBLs (Bush 2015), and (Moland et al. 2007). Even the potent inhibitory

16 Cite this article as Cold Spring Harb Perspect Med 2016;6:a025247


b-Lactams and b-Lactamase Inhibitors

activity against individual ESBLs that is ob- (Hecker et al. 2015), it is being developed in
served with clavulanic acid and tazobactam is combination with meropenem to target patho-
not sufficient to protect their accompanying gens producing serine carbapenemases (La-
penicillins in the presence of multiple b-lacta- puebla et al. 2015).
mases (Jones-Dias et al. 2014).
Following a hiatus of approximately two
decades, a unique class of non-b-lactam b-lac- b-LACTAM RESISTANCE: CONCLUDING
tamase inhibitors emerged, based on a novel REMARKS
bridged diazabicyclooctane (DBO) structure
Resistance to the b-lactams continues to in-
(Table 6) (Coleman 2011). The first of these
crease, especially in Gram-negative organisms
inhibitors, avibactam, has a broader spectrum
(Vasoo et al. 2015), because of the widespread
of activity than clavulanic acid and the sulfone
therapeutic dependence on these efficacious
inhibitors. Not only are class A penicillinases,
and safe antibiotics (see Fig. 1). Major resistance
ESBLs, and serine carbapenemases potently in-
mechanisms will be expanded on in other
hibited, but class C cephalosporinases and some
articles in this collection. PBP acquisition or
class D oxacillinases are also effectively inhibited
mutation is the major b-lactam-resistance
(Ehmann et al. 2012, 2013). Unlike the previous
mechanism in Gram-positive bacteria (see Fish-
inactivators described above, avibactam is a
er and Mobashery 2016). The most prevalent
tight-binding, covalent, reversible inhibitor for
and most damaging resistance mechanisms
most enzymes, with the KPC-2 enzyme, a nota-
among Gram-negative pathogens are represent-
ble exception for which slow avibactam hydro-
ed by the b-lactamases (Babic et al. 2006; Liv-
lysis was observed (Ehmann et al. 2012). In
ermore 2012), both chromosomally encoded
addition, avibactam does not induce AmpC b-
www.perspectivesinmedicine.org

enzymes that may be produced at high levels


lactamases at clinically relevant concentrations
and transferable enzymes that travel on mobile
(Coleman 2011). Avibactam has been approved
elements among species (Bush 2013). When
for therapeutic use in combination with cefta-
these targeted mechanisms are combined with
zidime, and is under development for ceftaro-
decreased uptake or increased efflux of the b-
line – avibactam or aztreonam – avibactam com-
lactam, high-level resistance becomes a major
binations (Flamm et al. 2014; Biedenbach et al.
clinical problem (see Bonomo 2016). Perhaps
2015; Li et al. 2015). Other DBOs under devel-
the most encouraging prospect in counteracting
opment include RG6080 and relebactam (MK
resistance is the emergence of new classes of b-
7655), in combination with imipenem. The
lactamase inhibitors that will provide protec-
spectrum of relebactam shows a similar spec-
tion for some of the most valuable antibiotics
trum of activity to avibactam; however, it pro-
in clinical practice, at least for the present time.
vides less potentiation against important class D
b-lactamases such as OXA-48 (Livermore et al.
2013). RG6080 (formerly OP0565) is a DBO
REFERENCES
that has an inhibitory spectrum similar to the 
Reference is also in this collection.
other DBOs but has the additional benefit of
exhibiting some intrinsic antibacterial activity Abe Y, Wakabayashi H, Ogawa Y, Machida A, Endo M, Tamai
against enteric bacteria (Livermore et al. 2015). T, Sakurai S, Hibino S, Mikawa T, Watanabe Y, et al. 2016.
The boronic acid inhibitor RPX7009 (Table Validation of cefazolin as initial antibiotic for first upper
urinary tract infection in children. Global Ped Health 3:
6) represents another novel class of synthetic 1 –7.
non-b-lactam b-lactamase inhibitors (Hecker Abraham EP. 1987. Cephalosporins 1945– 1986. Drugs 34:
et al. 2015), although boronic acids have been 1 –14.
known for many years to be effective inhibitors Anonymous. 1982. FDA approves new antibiotic for surgical
infections, meningitis. Hospitals 56: 55–57.
of serine b-lactamases (Kiener and Waley 1978).
Aronoff SC, Jacobs MR, Johenning S, Yamabe S. 1984.
Despite the inhibitory activity of RPX7009 Comparative activities of the b-lactamase inhibitors
against many groups of serine b-lactamases YTR 830, sodium clavulanate, and sulbactam combined

Cite this article as Cold Spring Harb Perspect Med 2016;6:a025247 17


K. Bush and P.A. Bradford

with amoxicillin or ampicillin. Antimicrob Agents Che- Charnas RL, Fisher J, Knowles JR. 1978. Chemical studies on
mother 26: 580–582. the inactivation of Escherichia coli RTEM b-lactamase by
Ashford WA, Golash RG, Hemming VG. 1976. Penicillinase- clavulanic acid. Biochemistry 17: 2185– 2189.
producing Neisseria gonorrhoeae. Lancet 308: 657– 658. Cielecka-Piontek J, Zajac M, Jelinska A. 2008. A comparison
Babic M, Hujer AM, Bonomo RA. 2006. What’s new in of the stability of ertapenem and meropenem in pharma-
antibiotic resistance? Focus on b-lactamases. Drug Resist ceutical preparations in solid state. J Pharm Biomed Anal
Updat 9: 142– 156. 46: 52–57.
Bamberger DM, Boyd SE. 2005. Management of Staphy- Cimarusti CM, Sykes RB. 1983. Monobactams—Novel an-
lococcus aureus infections. Am Fam Physician 72: 2474– tibiotics. Chem Brit 19: 302– 303.
2481. CLSI. 2016. Performance standards for antimicrobial sus-
Baughman RP. 2009. The use of carbapenems in the ceptibility testing. In CLSI supplement M100S, 26th ed.
treatment of serious infections. J Intens Care Med 24: Clinical and Laboratory Standards Institute, Wayne, PA.
230–241. Cole M. 1982. Biochemistry and action of clavulanic acid.
Bellido F, Pechère J-C, Hancock RW. 1991. Novel method for Scott Med J 27: S10– S16.
measurement of outer membrane permeability to new Coleman K. 2011. Diazabicyclooctanes (DBOs): A potent
b-lactams in intact Enterobacter cloacae cells. Antimicrob new class of non-b-lactam b-lactamase inhibitors. Curr
Agents Chemother 35: 68– 72. Opin Microbiol 14: 550–555.
Biedenbach DJ, Kazmierczak K, Bouchillon SK, Sahm DF, Curtis N, Orr D, Ross GW, Boulton MG. 1979. Affinities of
Bradford PA. 2015. In vitro activity of aztreonam– avi- penicillins and cephalosporins for the penicillin-binding
bactam against a global collection of Gram-negative proteins of Escherichia coli K-12 and their antibacterial
pathogens from 2012 and 2013. Antimicrob Agents Che- activity. Antimicrob Agents Chemother 16: 533 –539.
mother 59: 4239– 4248. Dagan R, Velghe L, Rodda JL, Klugman KP. 1994. Penetra-
Bonfiglio G, Russo G, Nicoletti G. 2002. Recent develop- tion of meropenem into the cerebrospinal fluid of pa-
ments in carbapenems. Exp Opin Investig Drugs 11: tients with inflamed meninges. J Antimicrob Chemother
529–544. 34: 175– 179.
 Bonomo RA. 2016. b-Lactamases: A focus on current chal- Dong X, Chen F, Zhang Y, Liu H, Liu Y, Ma L. 2014. In vitro
lenges. Cold Spring Harb Perspect Med doi: 10.1101/ activities of rifampin, colistin, sulbactam and tigecycline
cshperspect.a025239. tested alone and in combination against extensively
www.perspectivesinmedicine.org

Bradley JS, Garau J, Lode H, Rolston KV, Wilson SE, Quinn drug-resistant Acinetobacter baumannii. J Antibiotics 67:
JP. 1999. Carbapenems in clinical practice: A guide to 677 –680.
their use in serious infection. Int J Antimicrob Agents Easton CJ, Knowles JR. 1984. Correlation of the effect of
11: 93– 100. b-lactamase inhibitors on the b-lactamase in growing
Brown RB, Klar J, Lemeshow S, Teres D, Pastides H, Sands cultures of Gram-negative bacteria with their effect on
M. 1986. Enhanced bleeding with cefoxitin or moxalac- the isolated b-lactamase. Antimicrob Agents Chemother
tam. Statistical analysis within a defined population of 26: 358– 363.
1493 patients. Archiv Intern Med 146: 2159– 2164. Ehmann DE, Jahic H, Ross PL, Gu RF, Hu J, Kern G, Walkup
Bryskier AB, Belfiglio SR. 1999. Cephalosporins: Oral. In GK, Fisher SL. 2012. Avibactam is a covalent, reversible,
Antimicrobial therapy and vaccines (ed. Yu VL, et al.), non-b-lactam b-lactamase inhibitor. Proc Natl Acad Sci
pp. 703 –747. Lippincott Williams & Wilkins, Philadel- 109: 11663–11668.
phia. Ehmann DE, Jahić H, Ross PL, Gu RF, Hu J, Durand-Réville
Bryskier A, Aszodi J, Chantot JF. 1994. Parenteral cephalo- TF, Lahiri S, Thresher J, Livchak S, Gao N, et al. 2013.
sporin classification. Exp Opin Investig Drugs 3: 145 –171. Kinetics of avibactam inhibition against class A, C, and D
Bush K. 2013. Proliferation and significance of clinically b-lactamases. J Biol Chem 288: 27960–27971.
relevant b-lactamases. Ann NY Acad Sci 1277: 84– 90. Eliopoulos GM, Klimm K, Ferraro MJ, Moellering RC Jr.
Bush K. 2015. A resurgence of b-lactamase inhibitor com- 1989. In vitro activity of cefoperazone–sulbactam com-
binations effective against multidrug-resistant Gram- binations against cefoperazone-resistant clinical bacterial
negative pathogens. Int J Antimicrob Agents 46: 483 –493. isolates. Eur J Clinl Microbiol Infect Dis 8: 624–626.
Bush K, Macalintal C, Rasmussen BA, Lee VJ, Yang Y. 1993. English AR, Retsema JA, Girard AE, Lynch JE, Barth WE.
Kinetic interactions of tazobactam with b-lactamases 1978. CP-45.899, a b-lactamase inhibitor that extends
from all major structural classes. Antimicrob Agents Che- the antibacterial spectrum of b-lactams: Initial bacterio-
mother 37: 851–858. logical characterization. Antimicrob Agents Chemother
Castanheira M, Sader HS, Farrell DJ, Mendes RE, Jones RN. 14: 414– 419.
2012. Activity of ceftaroline– avibactam tested against Estabrook M, Bussell B, Clugston SL, Bush K. 2014. In vitro
Gram-negative organism populations, including strains activity of ceftolozane-tazobactam as determined by
expressing one or more b-lactamases and methicillin- broth dilution and agar diffusion assays against recent
resistant Staphylococcus aureus carrying various staphy- U.S. Escherichia coli isolates from 2010 to 2011 carrying
lococcal cassette chromosome mec types. Antimicrob CTX-M-type extended-spectrum b-lactamases. J Clin
Agents Chemother 56: 4779–4785. Microbiol 52: 4049– 4052.
Charnas RL, Knowles JR. 1981. Inactivation of RTEM Fass RJ, Gregory WW, D’Amato RF, Matsen JM, Wright DN,
b-lactamase from Escherichia coli by clavulanic acid and Young LS. 1990. In vitro activities of cefoperazone and
9-deoxyclavulanic acid. Biochemistry 20: 3214–3219. sulbactam singly and in combination against cefopera-

18 Cite this article as Cold Spring Harb Perspect Med 2016;6:a025247


b-Lactams and b-Lactamase Inhibitors

zone-resistant members of the family Enterobacteriaceae Hanberger H, Nilsson LE, Svensson E, Maller R. 1991. Syn-
and nonfermenters. Antimicrob Agents Chemother 34: ergic post-antibiotic effect of mecillinam, in combina-
2256– 2259. tion with other b-lactam antibiotics in relation to mor-
 Fisher JF, Mobashery S. 2016. b-Lactam-resistance mecha- phology and initial killing. J Antimicrob Chemother 28:
nisms: Gram-positive bacteria and Mycobacterium tu- 523 –532.
berculosis. Cold Spring Harb Perspect Med doi: 10.1101/ Harvengt C. 1985. Drugs recently released in Belgium. Te-
cshperspect.a025221. mocillin. Acta Clin Belg 40: 398–399.
Fisher J, Charnas RL, Bradley SM, Knowles JR. 1981. Inac- Hebeisen P, Heinze-Krauss I, Angehrn P, Hohl P, Page MG,
tivation of the RTEM b-lactamase from Escherichia coli. Then RL. 2001. In vitro and in vivo properties of Ro 63–
Interaction of penam sulfones with enzyme. Biochemistry 9141, a novel broad-spectrum cephalosporin with activ-
20: 2726–2731. ity against methicillin-resistant staphylococci. Antimi-
Flamm RK, Farrell DJ, Sader HS, Jones RN. 2014. Antimi- crob Agents Chemother 45: 825–836.
crobial activity of ceftaroline combined with avibactam Hecker SJ, Reddy KR, Totrov M, Hirst GC, Lomovskaya O,
tested against bacterial organisms isolated from acute Griffith DC, King P, Tsivkovski R, Sun D, Sabet M, et al.
bacterial skin and skin structure infections in United 2015. Discovery of a cyclic boronic acid b-lactamase in-
States medical centers (2010–2012). Diagn Microbiol In- hibitor (RPX7009) with utility vs class A serine carbape-
fect Dis 78: 449– 456. nemases. J Med Chem 58: 3682– 3692.
Fleming A. 1929. On the antibacterial action of cultures of a Hemmer RJ, Vaudaux P, Waldvogel FA. 1979. Methicillin
penicillium, with special reference to their use in the potentiates the effect of gentamicin on methicillin-resis-
isolation of B. influenzae. Br J Exp Pathol 10: 226 –236. tant Staphylococcus aureus. Antimicrob Agents Chemother
Frère JM, Joris B. 1985. Penicillin-sensitive enzymes in 15: 34–41.
peptidoglycan biosynthesis. CRC Crit Rev Microbiol 11: Hickman RA, Hughes D, Cars T, Malmberg C, Cars O. 2014.
299–396. Cell-wall-inhibiting antibiotic combinations with activi-
Fujimoto K, Takemoto K, Hatano K, Nakai T, Terashita S, ty against multidrug-resistant Klebsiella pneumoniae and
Matsumoto M, Eriguchi Y, Eguchi K, Shimizudani T, Sato Escherichia coli. Clin Microbiol Infect 20: O267–O273.
K, et al. 2013. Novel carbapenem antibiotics for paren- Hirsh HL, Dowling HF. 1946. The treatment of Streptococcus
teral and oral applications: In vitro and in vivo activities viridans endocarditis with penicillin. Southern Med J 39:
of 2-aryl carbapenems and their pharmacokinetics in 55– 60.
laboratory animals. Antimicrob Agents Chemother 57:
www.perspectivesinmedicine.org

Hofer B, Dantier C, Gebhardt K, Desarbre E, Schmitt-Hoff-


697–707. mann A, Page MG. 2013. Combined effects of the side-
Georgopapadakou NH, Liu FY. 1980. Penicillin-binding rophore monosulfactam BAL30072 and carbapenems on
proteins in bacteria. Antimicrob Agents Chemother 18: multidrug-resistant Gram-negative bacilli. J Antimicrob
148–157. Chemother 68: 1120– 1129.
Georgopapadakou N, Hammarstrom S, Strominger JL. Hornsey M, Phee L, Stubbings W, Wareham DW. 2013. In
1977. Isolation of the penicillin-binding peptide from vitro activity of the novel monosulfactam BAL30072
D-alanine carboxypeptidase of Bacillus subtilis. Proc alone and in combination with meropenem versus a di-
Natl Acad Sci 74: 1009– 1012. verse collection of important Gram-negative pathogens.
Giordano PA, Elston D, Akinlade BK, Weber K, Notario GF, Int J Antimicrob Agents 42: 343–346.
Busman TA, Cifaldi M, Nilius AM. 2006. Cefdinir vs. Inoue K, Hamana Y, Mitsuhashi S. 1995. In vitro antibacte-
cephalexin for mild to moderate uncomplicated skin rial activity and b-lactamase stability of a new carbape-
and skin structure infections in adolescents and adults. nem, BO-2727. Antimicrob Agents Chemother 39: 2331–
Curr Med Res Opin 22: 2419–2428. 2336.
Giske CG. 2015. Contemporary resistance trends and mech- Jacoby GA, Carreras I. 1990. Activities of b-lactam antibi-
anisms for the old antibiotics colistin, temocillin, fosfo- otics against Escherichia coli strains producing extended-
mycin, mecillinam and nitrofurantoin. Clin Microbiol spectrum b-lactamases. Antimicrob Agents Chemother
Infect 21: 899–905. 34: 858– 862.
Goffin C, Ghuysen JM. 1998. Multimodular penicillin- Jacoby GA, Han P. 1996. Detection of extended-spectrum b-
binding proteins: An enigmatic family of orthologs and lactamases in clinical isolates of Klebsiella pneumoniae
paralogs. Microbiol Mol Biol Rev 62: 1079–1093. and Escherichia coli. J Clin Microbiol 34: 908–911.
Gonzales PR, Pesesky MW, Bouley R, Ballard A, Biddy BA, Jacoby GA, Sutton L. 1989. Pseudomonas cepacia suscepti-
Suckow MA, Wolter WR, Schroeder VA, Burnham CA, bility to sulbactam. Antimicrob Agents Chemother 33:
Mobashery S, et al. 2015. Synergistic, collaterally sensitive 583 –584.
b-lactam combinations suppress resistance in MRSA. Jones-Dias D, Manageiro V, Ferreira E, Louro D, Antibiotic
Nature Chem Biol 11: 855 –861. Resistance Surveillance Program in Portugal (ARSIP),
Gunn BA, Woodall JB, Jones JF, Thornsberry C. 1974. Letter: Canica M. 2014. Diversity of extended-spectrum and
Ampicillin-resistant Haemophilus influenzae. Lancet 2: plasmid-mediated AmpC b-lactamases in Enterobacter-
845. iaceae isolates from Portuguese health care facilities. J
Hanaki H, Akagi H, Masaru Y, Otani T, Hyodo A, Hiramatsu Microbiol 52: 496–503.
K. 1995. TOC-39, a novel parenteral broad-spectrum Jorgensen JH, Maher LA, Howell AW. 1991. Activity of mer-
cephalosporin with excellent activity against methicil- openem against antibiotic-resistant or infrequently en-
lin-resistant Staphylococcus aureus. Antimicrob Agents countered Gram-negative bacilli. Antimicrob Agents Che-
Chemother 39: 1120–1126. mother 35: 2410–2414.

Cite this article as Cold Spring Harb Perspect Med 2016;6:a025247 19


K. Bush and P.A. Bradford

Kahan JS, Kahan FM, Goegelman R, Currie SA, Jackson M, against Gram-negative clinical isolates in New York
Stapley EO, Miller TW, Miller AK, Hendlin D, Mochales City. Antimicrob Agents Chemother 59: 4856– 4860.
S, et al. 1979. Thienamycin, a new b-lactam antibiotic. I: Li H, Estabrook M, Jacoby GA, Nichols WW, Testa RT, Bush
Discovery, taxonomy, isolation and physical properties. K. 2015. In vitro susceptibility of characterized b-lacta-
J Antibiotics 32: 1 –12. mase-producing strains tested with avibactam combina-
Kahan FM, Kropp H, Sundelof JG, Birnbaum J. 1983. Thie- tions. Antimicrob Agents Chemother 59: 1789–1793.
namycin: Development of imipenem–cilastatin. J Anti- Lister PD. 2000. b-lactamase inhibitor combinations with
microb Chemother 12: 1– 35. extended-spectrum penicillins: Factors influencing anti-
Karlowsky JA, Adam HJ, Decorby MR, Lagace-Wiens PR, bacterial activity against Enterobacteriaceae and Pseudo-
Hoban DJ, Zhanel GG. 2011. In vitro activity of ceftaro- monas aeruginosa. Pharmacotherapy 20: 213S–218S; dis-
line against Gram-positive and Gram-negative pathogens cussion 224S–228S.
isolated from patients in Canadian hospitals in 2009. Livermore DM. 2012. Current epidemiology and growing
Antimicrob Agents Chemother 55: 2837–2846. resistance of Gram-negative pathogens. Korean J Intern
Kato K, Shirasaka Y, Kuraoka E, Kikuchi A, Iguchi M, Suzuki Med 27: 128–142.
H, Shibasaki S, Kurosawa T, Tamai I. 2010. Intestinal Livermore DM, Mushtaq S. 2013. Activity of biapenem
absorption mechanism of tebipenem pivoxil, a novel (RPX2003) combined with the boronate b-lactamase in-
oral carbapenem: Involvement of human OATP family hibitor RPX7009 against carbapenem-resistant Entero-
in apical membrane transport. Mol Pharm 7: 1747– 1756. bacteriaceae. J Antimicrob Chemother 68: 1825– 1831.
Kattan JN, Villegas MV, Quinn JP. 2008. New developments Livermore DM, Hope R, Fagan EJ, Warner M, Woodford N,
in carbapenems. Clin Microbiol Infect 14: 1102–1111. Potz N. 2006. Activity of temocillin against prevalent
Khan W, Ross S, Rodriguez W, Controni G, Saz AK. 1974. ESBL- and AmpC-producing Enterobacteriaceae from
Haemophilus influenzae type B resistant to ampicillin: south-east England. J Antimicrob Chemother 57: 1012–
A report of two cases. JAMA 229: 298– 301. 1014.
Kiener PA, Waley SG. 1978. Reversible inhibitors of penicil- Livermore DM, Warner M, Mushtaq S. 2013. Activity of
linases. Biochemical J 169: 197– 204. MK-7655 combined with imipenem against Entero-
Kiratisin P, Chongthaleong A, Tan TY, Lagamayo E, Roberts bacteriaceae and Pseudomonas aeruginosa. J Antimicrob
S, Garcia J, Davies T. 2012. Comparative in vitro activity Chemother 68: 2286– 2290.
of carbapenems against major Gram-negative pathogens: Livermore DM, Mushtaq S, Warner M, Woodford N.
www.perspectivesinmedicine.org

Results of Asia-Pacific surveillance from the COMPACT 2015. Activity of OP0595/b-lactam combinations
II study. Int J Antimicrob Agents 39: 311– 316. against Gram-negative bacteria with extended-spectrum,
Kirby WM. 1944. Extraction of a highly potent penicillin AmpC and carbapenem-hydrolysing b-lactamases. J
inactivator from penicillin resistant staphylococci. Sci- Antimicrob Chemother 70: 3032–3041.
ence 99: 452– 453. Macfarlane G. 1979. Howard Florey, the making of a great
Kirby WM. 1945. Bacteriostatic and lytic actions of penicil- scientist. Oxford University Press, London.
lin on sensitive and resistant staphylococci. J Clin Invest Majumdar AK, Musson DG, Birk KL, Kitchen CJ, Holland
24: 165 –169. S, McCrea J, Mistry G, Hesney M, Xi L, Li SX, et al. 2002.
Klein JO. 2003. Amoxicillin/clavulanate for infections in Pharmacokinetics of ertapenem in healthy young volun-
infants and children: Past, present and future. Pediatr teers. Antimicrob Agents Chemother 46: 3506–3511.
Infect Dis J 22: S139– S148. Martinez-Beltran J, Loza E, Gomez-Alferez A, Romero-Vi-
Kohira N, West J, Ito A, Ito-Horiyama T, Nakamura R, Sato vas J, Bouza E. 1985. Temocillin. In vitro activity com-
T, Rittenhouse S, Tsuji M, Yamano Y. 2015. In vitro an- pared with other antibiotics. Drugs 29: 91– 97.
timicrobial activity of siderophore cephalosporin S- Martinez-Martinez L, Hernandez-Alles S, Alberti S, Tomas
649266 against Enterobacteriaceae clinical isolates in- JM, Benedi VJ, Jacoby GA. 1996. In vivo selection of
cluding carbapenem-resistant strains. Antimicrob Agents porin-deficient mutants of Klebsiella pneumoniae with
Chemother 60: 729– 734. increased resistance to cefoxitin and expanded-spec-
Kohler J, Dorso KL, Young K, Hammond GG, Rosen H, trum-cephalosporins. Antimicrob Agents Chemother 40:
Kropp H, Silver LL. 1999. In vitro activities of the 342 –348.
potent, broad-spectrum carbapenem MK-0826 (L-749, Massova I, Mobashery S. 1998. Kinship and diversification
345) against broad-spectrum b-lactamase and extended- of bacterial penicillin-binding proteins and b-lacta-
spectrum b-lactamase-producing Klebsiella pneumoniae mases. Antimicrob Agents Chemother 42: 1 –17.
and Escherichia coli clinical isolates. Antimicrob Agents Medeiros AA. 1984. b-lactamases. Br Med Bull 40: 18– 27.
Chemother 43: 1170–1176. Medeiros AA. 1997. Evolution and dissemination of b-lac-
Kropp H, Sundelof JG, Hadju R, Kahan FM. 1982. Metab- tamases accelerated by generations of b-lactam antibiot-
olism of thienamycin and related carbapenem antibiotics ics. Clin Infect Dis 24: S19– S45.
by the renal dipeptidase, dehydropeptidase-I. Antimicrob Moisan H, Pruneau M, Malouin F. 2010. Binding of ceftaro-
Agents Chemother 22: 62– 70. line to penicillin-binding proteins of Staphylococcus au-
Labia R, Morand A. 1994. Interaction of cefdinir with b- reus and Streptococcus pneumoniae. J Antimicrob Chemo-
lactamases. Drugs Exp Clin Res 20: 43–48. ther 65: 713 –716.
Lapuebla A, Abdallah M, Olafisoye O, Cortes C, Urban C, Moland ES, Hong SG, Thomson KS, Larone DH, Hanson
Quale J, Landman D. 2015. Activity of meropenem com- ND. 2007. A Klebsiella pneumoniae isolate producing at
bined with RPX7009, a novel b-lactamase inhibitor, least eight different b-lactamases including an AmpC

20 Cite this article as Cold Spring Harb Perspect Med 2016;6:a025247


b-Lactams and b-Lactamase Inhibitors

and KPC b-lactamase. Antimicrob Agents Chemother 51: Pottegard A, Broe A, Aabenhus R, Bjerrum L, Hallas J,
800–801. Damkier P. 2015. Use of antibiotics in children: A Danish
Moss RB, McClelland E, Williams RR, Hilman BC, Rubio T, nationwide drug utilization study. Pediatr Infect Dis J 34:
Adkinson NF. 1991. Evaluation of the immunologic e16–e22.
cross-reactivity of aztreonam in patients with cystic fibro- Prescott WA Jr, Gentile AE, Nagel JL, Pettit RS. 2011. Con-
sis who are allergic to penicillin and/or cephalosporin tinuous-infusion antipseudomonal b-lactam therapy in
antibiotics. Rev Infect Dis 13: S598–S607. patients with cystic fibrosis. P&T 36: 723– 763.
Mushtaq S, Warner M, Williams G, Critchley I, Livermore Rammelkamp CH, Keefer CS. 1943. Penicillin: Its antibac-
DM. 2010. Activity of chequerboard combinations of terial effect in whole blood and serum for the hemolytic
ceftaroline and NXL104 versus b-lactamase-producing Streptococcus and Staphylococcus aureus. J Clin Investig
Enterobacteriaceae. J Antimicrob Chemother 65: 1428– 22: 649– 657.
1432. Reading C, Cole M. 1977. Clavulanic acid: A b-lactamase
Mushtaq S, Woodford N, Hope R, Adkin R, Livermore DM. inhibitor from Streptomyces clavuligerus. Antimicrob
2013. Activity of BAL30072 alone or combined with Agents Chemother 11: 852– 857.
b-lactamase inhibitors or with meropenem against car- Riera E, Cabot G, Mulet X, Garcıa-Castillo M, del Campo R,
bapenem-resistant Enterobacteriaceae and non-fermen- Juan C, Canton R, Oliver A. 2011. Pseudomonas aerugi-
ters. J Antimicrob Chemother 68: 1601–1608. nosa carbapenem resistance mechanisms in Spain: Im-
Neu HC. 1990. b-lactamases, b-lactamase inhibitors, and pact on the activity of imipenem, meropenem and dor-
skin and skin-structure infections. J Am Acad Dermatol ipenem. J Antimicrob Chemother 66: 2022–2027.
22: 896 –904. Rodriguez-Villalobos H, Malaviolle V, Frankard J, de Men-
Neu HC, Novelli A, Chin NX. 1989. In vitro activity and donca R, Nonhoff C, Struelens MJ. 2006. In vitro activity
b-lactamase stability of a new carbapenem, SM-7338. of temocillin against extended spectrum b-lactamase-
Antimicrob Agents Chemother 33: 1009–1018. producing Escherichia coli. J Antimicrob Chemother 57:
771 –774.
Neu HC, Gu JW, Fang W, Chin NX. 1992. In vitro activity
and b-lactamase stability of LJC 10,627. Antimicrob Sader HS, Castanheira M, Mendes RE, Flamm RK, Farrell
Agents Chemother 36: 1418–1423. DJ, Jones RN. 2015. Ceftazidime-avibactam activity
against multidrug-resistant Pseudomonas aeruginosa iso-
Newton GGF, Abraham EP. 1956. Isolation of cephalosporin
lated in U.S. medical centers in 2012 and 2013. Antimi-
C, a penicillin-like antibiotic containing D-a amino-
www.perspectivesinmedicine.org

crob Agents Chemother 59: 3656– 3659.


adipic acid. Biochem J 62: 651 –658.
Saroglou G, Cromer M, Bisno AL. 1980. Methicillin-resis-
Nikaido H, Liu W, Rosenberg EY. 1990. Outer membrane tant Staphylococcus aureus: Interstate spread of nosoco-
permeability and b-lactamase stability of dipolar ionic mial infections with emergence of gentamicin–methicil-
cephalosporins containing methoxyimino substituents. lin resistant strains. Infect Control 1: 81–89.
Antimicrob Agents Chemother 34: 337– 342.
Sato T, Hara T, Horiyama T, Kanazawa S, Yamaguchi T, Maki
Nordmann P, Poirel L. 2002. Emerging carbapenemases in H. 2015. Mechanism of resistance and antibacterial sus-
Gram-negative aerobes. Clin Microbiol Infect 8: 321 –331. ceptibility in extended-spectrum b-lactamase phenotype
Nordmann P, Picazo J, Mutters R, Korten V, Quintana A, Klebsiella pneumoniae and Klebsiella oxytoca isolated be-
Laeuffer J, Seak J, Flamm R, Morrissey I. 2011. Compar- tween 2000 and 2010 in Japan. J Med Microbiol 64: 538–
ative activity of carbapenem testing: The COMPACT 543.
study. J Antimicrob Chemother 66: 1070– 1078. Saxon A, Hassner A, Swabb EA, Wheeler B, Adkinson NF Jr.
Otero LH, Rojas-Altuve A, Llarrull LI, Carrasco-Lopez C, 1984. Lack of cross-reactivity between aztreonam, a
Kumarasiri M, Lastochkin E, Fishovitz J, Dawley M, He- monobactam antibiotic, and penicillin in penicillin-al-
sek D, Lee M, et al. 2013. How allosteric control of Staph- lergic subjects. J Infect Dis 149: 16– 22.
ylococcus aureus penicillin binding protein 2a enables Schaar V, Uddback I, Nordstrom T, Riesbeck K. 2014. Group
methicillin resistance and physiological function. Proc A streptococci are protected from amoxicillin-mediated
Natl Acad Sci 110: 16808–16813. killing by vesicles containing b-lactamase derived from
Page MGP, Dantier C, Desarbre E. 2010. In vitro properties Haemophilus influenzae. J Antimicrob Chemother 69:
of BAL30072, a novel siderophore sulfactam with activity 117 –120.
against multiresistant Gram-negative bacilli. Antimicrob Simpson IN, Harper PB, O’Callaghan CH. 1980. Principal
Agents Chemother 54: 2291–2302. b-lactamases responsible for resistance to b-lactam anti-
Papp-Wallace KM, Endimiani A, Taracila MA, Bonomo RA. biotics in urinary tract infections. Antimicrob Agents Che-
2011. Carbapenems: Past, present, and future. Antimicrob mother 17: 929 –936.
Agents Chemother 55: 4943–4960. Spratt BG. 1977. Properties of the penicillin-binding pro-
Payne DJ, Amyes SG. 1993. Stability of cefdinir (C1-983, teins of Escherichia coli K12. Eur J Biochem 72: 341– 352.
FK482) to extended-spectrum plasmid-mediated b-lac- Spratt BG. 1983. Penicillin-binding proteins and the future
tamases. J Med Microbiol 38: 114 –117. of b-lactam antibiotics. J Gen Microbiol 129: 1247–1260.
Pillar CM, Aranza MK, Shah D, Sahm DF. 2008. In vitro Steward CD, Rasheed JK, Hubert SK, Biddle JW, Raney PM,
activity profile of ceftobiprole, an anti-MRSA cephalo- Anderson GJ, Williams PP, Brittain KL, Oliver A, Mc-
sporin, against recent Gram-positive and Gram-negative Gowan JE Jr, et al. 2001. Characterization of clinical iso-
isolates of European origin. J Antimicrob Chemother 61: lates of Klebsiella pneumoniae from 19 laboratories using
595–602. the National Committee for Clinical Laboratory Stan-

Cite this article as Cold Spring Harb Perspect Med 2016;6:a025247 21


K. Bush and P.A. Bradford

dards extended-spectrum b-lactamase detection meth- Wise R, Andrews J, Bedford K. 1978. In vitro study of clav-
ods. J Clin Microbiol 39: 2864– 2872. ulanic acid in combination with penicillin, amoxycillin,
Sudo T, Murakami Y, Uemura K, Hashimoto Y, Kondo N, and carbenicillin. Antimicrob Agents Chemother 13: 389–
Nakagawa N, Ohge H, Sueda T. 2014. Perioperative anti- 393.
biotics covering bile contamination prevent abdominal Woodford N, Tierno PM Jr, Young K, Tysall L, Palepou MF,
infectious complications after pancreatoduodenectomy Ward E, Painter RE, Suber DF, Shungu D, Silver LL,
in patients with preoperative biliary drainage. World et al. 2004. Outbreak of Klebsiella pneumoniae producing
J Surg 38: 2952– 2959. a new carbapenem-hydrolyzing class A b-lactamase,
Sumita Y, Fukasawa M. 1995. Potent activity of meropenem KPC-3, in a New York Medical Center. Antimicrob Agents
against Escherichia coli arising from its simultaneous Chemother 48: 4793– 4799.
binding to penicillin-binding proteins 2 and 3. J Antimi- Yang Y, Bhachech N, Bush K. 1995. Biochemical comparison
crob Chemother 36: 53–64.
of imipenem, meropenem and biapenem: Permeability,
Sykes RB, Bonner DP, Bush K, Georgopapadakou NH. 1982. binding to penicillin-binding proteins, and stability to
Azthreonam (SQ 26,776), a synthetic monobactam spe- hydrolysis by b-lactamases. J Antimicrob Chemother 35:
cifically active against aerobic Gram-negative bacteria. 75– 84.
Antimicrob Agents Chemother 21: 85–92.
Yigit H, Queenan AM, Rasheed JK, Biddle JW, Domenech-
Talbot GH, Thye D, Das A, Ge Y. 2007. Phase 2 study of
Sanchez A, Alberti S, Bush K, Tenover FC. 2003. Carba-
ceftaroline versus standard therapy in treatment of com-
penem-resistant strain of Klebsiella oxytoca harboring
plicated skin and skin structure infections. Antimicrob
carbapenem-hydrolyzing b-lactamase KPC-2. Antimi-
Agents Chemother 51: 3612–3616.
crob Agents Chemother 47: 3881– 3889.
Tipper DJ, Strominger JL. 1965. Mechanism of action of
penicillins: A proposal based on their structural similar- Zhanel GG, Wiebe R, Dilay L, Thomson K, Rubinstein E,
ity to acyl-D-alanyl-D-alanine. Proc Natl Acad Sci 54: Hoban DJ, Noreddin AM, Karlowsky JA. 2007. Compar-
1133– 1141. ative review of the carbapenems. Drugs 67: 1027– 1052.
Vasoo S, Barreto JN, Tosh PK. 2015. Emerging issues in Zhanel GG, Chung P, Adam H, Zelenitsky S, Denisuik A,
Gram-negative bacterial resistance: An update for the Schweizer F, Lagace-Wiens PR, Rubinstein E, Gin AS,
practicing clinician. Mayo Clin Proc 90: 395 –403. Walkty A, et al. 2014. Ceftolozane/tazobactam: A novel
Wang X, Zhang F, Zhao C, Wang Z, Nichols WW, Testa R, Li cephalosporin/b-lactamase inhibitor combination with
activity against multidrug-resistant Gram-negative ba-
www.perspectivesinmedicine.org

H, Chen H, He W, Wang Q, et al. 2014. In vitro activities


of ceftazidime– avibactam and aztreonam–avibactam cilli. Drugs 74: 31– 51.
against 372 Gram-negative bacilli collected in 2011 and Zykov IN, Sundsfjord A, Smabrekke L, Samuelsen O. 2016.
2012 from 11 teaching hospitals in China. Antimicrob The antimicrobial activity of mecillinam, nitrofurantoin,
Agents Chemother 58: 1774–1778. temocillin and fosfomycin and comparative analysis of
Weber DA, Sanders CC. 1990. Diverse potential of b-lacta- resistance patterns in a nationwide collection of ESBL-
mase inhibitors to induce class I enzymes. Antimicrob producing Escherichia coli in Norway 2010– 2011. Infect
Agents Chemother 34: 156 –158. Dis (Lond) 48: 99–107.

22 Cite this article as Cold Spring Harb Perspect Med 2016;6:a025247

You might also like