ArticlePDF AvailableLiterature Review

Beta-lactams and Beta-lactamase-inhibitors in current- or potential-clinical practice: A comprehensive update

Taylor & Francis
Critical Reviews in Microbiology
Authors:
  • Jazan University.

Abstract and Figures

The use of successive generations of beta-lactams has selected successive generations of beta-lactamases including CTX-M ESBLs, AmpC beta-lactamases, and KPC carbapenamases in Enterobacteriaceae. Moreover, this cephalosporin resistance, along with rising resistance to fluoroquinolones, is now driving the use of carbapenems and unfortunately the carbapenem resistance has emerged markedly, especially in Acinetobacter spp. due to OXA- and metallo-carbapenemases. The industry responded to the challenge of rising resistance and recently developed some novel beta-lactams such as ceftobiprole, ceftaroline etc. and many beta-lactam compounds, including beta-lactamase-inhibitors, such as BMS-247243, S-3578, RWJ-54428, CS-023, SMP-601, NXL 104, BAL 30376, LK 157, and so on are under trials. This review provides the comprehensive accounts of the developments in penicillins, cephalosporins, carbapenems, and beta-lactamase-inhibitors, and the insight about medicinal chemistry, mechanism(s) of action and resistance, potential strategies to overcome resistance due to beta-lactamases, and also the recent advancements in the development of newer beta-lactam compounds; some of which are still under trials and yet to be classified. This review will fill the gap since previously published reviews and will serve as a comprehensive update on the current topic.
Content may be subject to copyright.
PLEASE SCROLL DOWN FOR ARTICLE
This article was downloaded by:
[INFLIBNET India Order]
On:
20 June 2009
Access details:
Access Details: [subscription number 909277340]
Publisher
Informa Healthcare
Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: Mortimer House,
37-41 Mortimer Street, London W1T 3JH, UK
Critical Reviews in Microbiology
Publication details, including instructions for authors and subscription information:
http://www.informaworld.com/smpp/title~content=t713400901
Beta-lactams and Beta-lactamase-inhibitors in current- or potential-clinical
practice: A comprehensive update
M. Shahid a; F. Sobia a; A. Singh a; A. Malik a; H. M. Khan a; D. Jonas b; P. M. Hawkey cd
a Section of Antimicrobial Resistance Researches and Molecular Biology, Department of Microbiology,
Jawaharlal Nehru Medical College & Hospital, Aligarh Muslim University, Aligarh, Uttar Pradesh, India b
Department of Environmental Health Sciences, Freiburg University Medical Centre, Breisacher Straße,
Freiburg, Germany c Antimicrobial Agents Research Group, Institute of Biomedical Research, University of
Birmingham, Edgbaston, Birmingham, UK d West Midlands Health Protection Agency, Heart of England NHS
Foundation Trust, Bordesley Green East, Birmingham, UK
Online Publication Date: 01 May 2009
To cite this Article Shahid, M., Sobia, F., Singh, A., Malik, A., Khan, H. M., Jonas, D. and Hawkey, P. M.(2009)'Beta-lactams and Beta-
lactamase-inhibitors in current- or potential-clinical practice: A comprehensive update',Critical Reviews in Microbiology,35:2,81 — 108
To link to this Article: DOI: 10.1080/10408410902733979
URL: http://dx.doi.org/10.1080/10408410902733979
Full terms and conditions of use: http://www.informaworld.com/terms-and-conditions-of-access.pdf
This article may be used for research, teaching and private study purposes. Any substantial or
systematic reproduction, re-distribution, re-selling, loan or sub-licensing, systematic supply or
distribution in any form to anyone is expressly forbidden.
The publisher does not give any warranty express or implied or make any representation that the contents
will be complete or accurate or up to date. The accuracy of any instructions, formulae and drug doses
should be independently verified with primary sources. The publisher shall not be liable for any loss,
actions, claims, proceedings, demand or costs or damages whatsoever or howsoever caused arising directly
or indirectly in connection with or arising out of the use of this material.
Critical Reviews in Microbiology, 2009; 35(2): 81–108
R E VI EW A RTICLE
Beta-lactams and Beta-lactamase-inhibitors
in current- or potential- clinical practice:
A comprehensive update
M. Shahid1, F. Sobia1, A. Singh1, A. Malik1, H. M. Khan1, D. Jonas2, and P. M. Hawkey3,4
1Section of Antimicrobial Resistance Researches and Molecular Biology, Department of Microbiology, Jawaharlal
Nehru Medical College & Hospital, Aligarh Muslim University, Aligarh-202002, Uttar Pradesh, India, 2Department
of Environmental Health Sciences, Freiburg University Medical Centre, Breisacher Straße 115 BD-79106, Freiburg,
Germany, 3Antimicrobial Agents Research Group, Institute of Biomedical Research, University of Birmingham, Vincent
Drive, Edgbaston, Birmingham B15 2TT, UK, and 4West Midlands Health Protection Agency, Heart of England NHS
Foundation Trust, Bordesley Green East, Birmingham, B9 5SS, UK
Address for Correspondence: shahidsahar@yahoo.co.in; drmohdshahid123@yahoo.com
(Received 29 July 2008; revised 11 November 2008; accepted 08 January 2009)
Introduction
e history of B-lactam antibiotics began with Alexander
Fleming and his observation in 1928 that a strain of the
mould Penicillium produced a diusible antibacte-
rial agent, which he named penicillin (Fleming 1929).
However, the modern era of antimicrobial chemo-
therapy dates back to 1936, with the introduction of
sulfanilamide into clinical practice. Penicillin became
available in quantities sucient for clinical use in 1941,
and streptomycin, chloramphenicol, and tetracycline
were identied toward the end of or soon after World
ISSN 1040-841X print/ISSN 1549-7828 online © 2009 Informa UK Ltd
DOI: 10.1080/10408410902733979
Abstract
The use of successive generations of β-lactams has selected successive generations of β-lactamases includ-
ing CTX-M ESBLs, AmpC β-lactamases, and KPC carbapenamases in Enterobacteriaceae. Moreover, this
cephalosporin resistance, along with rising resistance to uoroquinolones, is now driving the use of carbap-
enems and unfortunately the carbapenem resistance has emerged markedly, especially in Acinetobacter
spp. due to OXA- and metallo-carbapenemases. The industry responded to the challenge of rising resist-
ance and recently developed some novel β-lactams such as ceftobiprole, ceftaroline etc. and many
β-lactam compounds, including β-lactamase-inhibitors, such as BMS-247243, S-3578, RWJ-54428, CS-023,
SMP-601, NXL 104, BAL 30376, LK 157, and so on are under trials. This review provides the comprehensive
accounts of the developments in penicillins, cephalosporins, carbapenems, and β-lactamase-inhibitors,
and the insight about medicinal chemistry, mechanism(s) of action and resistance, potential strategies to
overcome resistance due to β-lactamases, and also the recent advancements in the development of newer
β-lactam compounds; some of which are still under trials and yet to be classied. This review will ll the gap
since previously published reviews and will serve as a comprehensive update on the current topic.
Keywords: Newer- β-lactams; penicillins; cephalosporins; carbapenems; β-lactamase-inhibitors; resistance
Abbreviations: 3GCs: Third generation cephalosporins; 6-APA: 6-amino penicillanic acid; ABC:
Adenosine triphosphate binding cassette; CA-MRSA: Community-acquired methicillin-resistant S. aureus;
CFU: colony forming unit; ESBLs: Extended spectrum beta-lactamases; ISCRs: Insertion sequence common
regions; MATE: Multidrug and toxic compound extrusion; MBL: Metallo-beta-lactamases; MDR: Multi-drug
resistance; MFP: Membrane fusion protein; MFS: Major facilitator superfamily; MIC: Minimum inhibitory
concentration; MRCoNS: Methicillin-resistance coagulase negative Staphylococci; MRSA: Methicillin
resistant S. aureus; MRSE: Methicillin resistant S. epidermidis; MSSA: Methicillin susceptible S. aureus;
OMP: Outer membrane protein; PBPs:Penicillin binding proteins; PRSP: Penicillin-resistant S. pneumoniae;
RND: Resistance nodulation cell division; RTI: Reproductive tract infection; SMR: Small multidrug
resistance; UDP: Uridine diphosphate; UTI: Urinary tract infection; VRE: Vancomycin resistant Enterococci
http://www.informapharmascience.com/mcb
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
82 Shahid et al.
War II (Mandell and Petri 1996). Since then, numerous
classes of antimicrobial agents have been discovered
and even continue to be discovered in recent years with
some potent antimicrobials including newer B-lactams
and carbapenems. However, the pace of antimicrobial
drug development has drastically slowed during the last
decade, with only few newer agents, some of which are
really novel.
Since the introduction of antimicrobials into clinical
practice a little over 6 decades ago, the resistant bacterial
strains have arisen due to selective pressure of their use.
During this period, numerous classes of antibiotics were
developed and, in response, antibiotics resistance genes
continued to emerge to prove the Darwinian dictum of
the survival of the ttest. Taking as an example, in 1950s,
the problem due to resistant bacterial isolates, espe-
cially due to Staphylococcus aureus, was tried to solve by
the development of penicillinase-stable penicillins such
as methicillin and the more clinically useful derivatives
cloxacillin and ucloxacillin. However, it was disappoint-
ing that following a year of the introduction of methicillin,
the rst methicillin-resistant isolate and clinical failure
was discovered (Jevons 1961). Similarly, the rapid rise of
resistance to ampicillin in the early 1960s turned out to
be due to a plasmid mediated B-lactamase, one of the
rst described in Gram-negative bacteria, known as TEM
(Hawkey 2008). In turn, industry responded by develop-
ing more sophisticated B-lactam compounds resistant
to hydrolysis by TEM and SHV B-lactamases that were
popularly known as third-generation cephalosporins
(3GCs). e further selection of resistant mutants and
acquisition of novel antibiotic resistance genes led to the
appearance of extended-spectrum B-lactamases (ESBLs)
that now compromise the use of 3GCs for treatment of
serious infections caused by Gram-negative bacteria.
e industry further responded with the introduction
through the 1990s of carbapenems, which are extremely
stable to degradation by B-lactamases and which was
claimed to be the answer (Hawkey 2008). However, a
variety of B-lactamases capable of hydrolyzing carbap-
enems, including IMP, VIM, GIM, SPM, KPC, and OXA,
are seen increasingly in clinical Gram-negative bacterial
isolates (Walsh et al. 2005; Hawkey 2008). To combat this
threatening problem of drug resistances, researchers
are still trying to develop newer compounds including
newer cephalosporins and carbapenems. e purpose
of this review is, therefore, to provide comprehensive
accounts of the developments in penicillins, cepha-
losporins, carbapenems, and B-lactamase-inhibitors,
and also the recent advancements in the development
of newer B-lactams; some of which are still under trials
and yet to be classied.
e penicillins
e history of B-lactams began with Alexander Fleming
and his observation in 1928 that a strain of the mould
Penicillium produced a diusible antibacterial agent,
which he named penicillin (Fleming 1929). Attempts
to isolate and purify penicillin in the 1930s were largely
unsuccessful and by the end of the 1930s interest in pen-
icillin had almost disappeared. A decade later, penicil-
lin was developed as a semisynthetic therapeutic agent
(Chain et al. 1940). Subsequently in the 1940s and 1950s,
the deep fermentation procedure for the biosynthesis of
penicillin marked a crucial advance in the large-scale
production of this antibiotic (readers are advised to see
Rolinson 1998 for detailed description of the fermen-
tative strategies, and development of semisynthetic
penicillins). During this period, B-lactams comprised
only two compounds, namely penicillin G and peni-
cillin V, and the major extension of the B-lactam eld
only occurred in the early 1960s with the development
of the semisynthetic penicillin, quickly followed by the
development of semisynthetic cephalosporins and
other B-lactam antibiotics. Until the middle of the 1970s
virtually all the signicant developments in B-lactam
N
SCH3
CH3
O
O
OH
R
O
HH
BA
H
N
N
SCH3
CH3
O
O
OH
H2NH
CH
O
R
Penicillins
Amidase
Penicillinase
O
HC
S
C
CH3
HC COOHN
H
CH3
NH CH
C
OH
CHR
O
Penicilloic Acids
R฀6- Aminopenicillanic Acid
Figure 1. Penicillins and product of their enzymatic hydrolysis.
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
b-lactams in clinical practice 83
antibiotics had been achieved by adding dierent side
chains to the classical penam and cephem nucleus.
Chemistry of penicillins
e basic structure of penicillins consists of a thiazo-
lidine ring (B) connected to a B -lactam ring (A) {the
penicillin nucleus; 6-amino penicillanic acid (6-APA)}
and to which is attached a side chain (R) (see Fig. 1). e
penicillin nucleus (6-APA) is the chief structural require-
ment for biological activity and requires the presence of
an acid residue on the thiazolidine ring for binding to
the penicillin binding proteins. Chemical alterations of
this portion of the molecule cause loss of all signicant
antibacterial activity. e side chain determines many of
the antibacterial and pharmacological characteristics of
a particular type of penicillin (Mendell and Petri 1996).
Production of 6-APA by enzymatic hydrolysis
e penicillin nucleus, 6-APA (Fig. 2), was rst obtained
as a naturally occurring fermentation product (Batchelor
et al. 1959). However, the yields were always lower than
those of penicillin G or penicillin V obtained in fermen-
tations in which the appropriate precursor was used.
Subsequently, an alternative route to obtain 6-APA was
discovered by enzymatic removal of the side-chain of
the penicillin molecule (Rolinson et al. 1960). A process
for the manufacture of the 6-APA by deacylation was
rst developed in Beecham Research laboratories using
a deacylase obtained from Streptomyces lavendulae with
penicillin V as substrate. Subsequently the deacylase of
bacterial origin was discovered independently in a num-
ber of laboratories (Rolinson et al. 1960; Claridge et al.
1960; Huang et al. 1960; Kaufmann and Bauer 1960) and
6-APA was obtained by using this enzyme with peni-
cillin G as substrate. 6-APA is now produced in large
quantities with the aid of an amidase from Penicillium
chrysogenum (Rolinson et al. 1960).
Semisynthetic penicillins
e discovery of 6-APA, depleted of side chain pre-
cursors, led to the development of the semisynthetic
penicillins. Side chains can be added that alter the
susceptibility of the resultant compounds to inactivat-
ing enzymes, like B-lactamases, and that change the
antibacterial spectrum and pharmacological properties
of the drugs (Mandell and Petri 1996). e rst semi-
synthetic penicillin introduced in the clinical practice
was phenethicillin (a close analogue of Penicillin V).
is was followed by methicillin (Rolinson et al. 1960),
ampicillin (Rolinson and Stevens 1961), amoxycillin,
the isoxazolyl penicillins (Geraci et al. 1962) like oxa-
cillin, cloxacillin, dicloxacillin and ucloxacillin, the
B lactam active against Pseudomonas aeruginosa, like
carbenicillin (Rolinson and Sutherland 1967) and ticar-
cillin (Rolinson and Sutherland 1967) (for more detailed
description of historical aspects of B -lactam research,
the readers are encouraged to read Rolinson 1998).
e cephalosporins
e rst source of cephalosporins was Cephalosporium
acremonium and the culture ltrate in which the fungus
was grown was found to contain three distinct antibiotics,
namely, cephalosporin P, N, and C. e discovery of the
nucleus, 7-aminocephalosporanic acid (Fig. 3), made it
possible to introduce semisynthetic compounds, with anti-
bacterial activity greater than that of the parent substance,
by addition of side chains (for detailed historical review and
discussion of biochemistry, Abraham 1962; Flynn 1972).
Chemistry of cephalosporins
Cephalosporin C contains a side chain derived from
D-A-aminoadipic acid, which is condensed with a
dihydrothiazine B-lactam ring system (7-aminocepha-
losporanic acid). Cephalosporin C can be hydrolyzed
by acid to 7-aminocephalosporanic acid. Subsequent
modications had been achieved by addition of dier-
ent side chains to create a whole family of cephalosporin
antibiotics (Mandell and Petri 1996). e cephamycins
are similar to the cephalosporins, but have a methoxy
group at position 7 of the B-lactam ring of the 7-aminoc-
ephalosporanic acid nucleus (Fig. 2).
Classication and spectrum of activity of
cephalosporins
e classication by “generations”, as shown in Table 1 is
very useful, although it is somewhat arbitrary. is clas-
sication is based on several features of antimicrobial
N
SCH3
CH3
H
O
R1
O
OOH
H
N
Figure 2.6-APA Nucleus.
H
N
N
O
R1
O
S
R2
COOH
Figure 3. 7-ACA Nucleus.
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
84 Shahid et al.
activity (Karchmer 2000). e rst generation cepha-
losporins have good activity against Gram-positive
bacteria and relatively modest activity against Gram-
negative microorganisms. Most Gram-positive cocci
are susceptible with exception of enterococci, methi-
cillin-resistant S. aureus (MRSA) and S. epidermidis.
Most anaerobes of gastrointestinal tract, including oral
cavity, except Bacteroides fragilis are susceptible. e
activity against Moraxella catarrhalis,Escherichia coli,
Klebsiella pneumoniae, and Proteus mirabilis is good.
e second-generation cephalosporins have somewhat
increased activity against Gram-negative organisms, but
are less active than the third-generation agents. A sub-
set of second-generation agents (cefoxitin, cefotetan,
and cefmetazole) is also active against B. fragilis
group. ird-generation cephalosporins are generally
less active than rst-generation agents against Gram-
positive cocci, but they are much more active against the
Enterobacteriaceae, including B-lactamase–producing
strains. A subset of third generation agents (ceftazidime
and cefoperazone) is also active against Pseudomonas
aeruginosa but less active than other third-generation
agents against Gram-positive cocci (Donowitz and
Mandell 1988). Fourth-generation cephalosporins
have an extended spectrum of activity compared to
the third-generation and have increased stability from
hydrolysis by plasmid and chromosomally mediated B-lactamases (Mandell and Petri 1996). It must be noted
that none of the cephalosporins on the market has reli-
able activity against penicillin-resistant Streptococcus
pneumoniae, MRSA, methicillin-resistant S. epidermidis
(MRSE) and other coagulase-negative staphylococci,
enterococci, Listeria monocytogenes,Legionella pneu-
mophila,Legionella micdadei,Clostridium dicile,
Stenotrophomonas maltophilia, Campylobacter jejuni,
and Acinetobacter species.
Mechanism of action of penicillins and
cephalosporins
Penicillin acts by inhibiting the bacterial cell wall syn-
thesis i.e. the inhibition of the transpeptidation reaction
of the peptidoglycan synthesis. Numerous researchers
have provided information that allows understand-
ing of the basic phenomenon of mechanism (Ghuysen
1991; Du Bois et al. 1995; Bayles 2000). Peptidoglycan
is a heteropolymeric component of the cell wall that
provides rigid mechanical stability. e peptidoglycan is
composed of glycan chains, which are linear strands of
two alternating amino sugars (N-acetylglucosamine and
N-acetylmuramic acid) that are cross-linked by peptide
chains. e biosynthesis of the peptidoglycan involves
about 30 bacterial enzymes and may be considered in
Table 1. Classication of Cephalosporins.
First Generation Second Generation ird Generation Fourth Generation Yet to be classied
Cefacetrile* Cefonicid* Cefcapene Cefepime* Cefaclomezine
Cefadroxil* (Monocid) Cefdaloxime (Maxipime) Cefaloram
(Cefadroxyl; Duricef) Cefprozil*(Cefproxil; Cefdinir* (Omnicef) Cefclidine Cefaparole
Cefalexin*(Cephalexin; Cefzil) Cefditoren* Ceuprenam Cefcanel
Keex) Cefmandole* Cefetamet* Cefoselis Cefedroler
Cephaloglycin* Cefuroxime* (Zinnat, Cexime* (Suprax) Cefozopran Cefempidone
Cefalonium Zinacef, Ceftin, Cefmenoxime* Cefpirome* Cefetrizole
Cefaloridine* Biofuroksym) Cefodizime* Cefquinome Cevitril
Cefalotin* Ceforanide* Cefoperazone* (Cefobid) Cefmatilen
(Cephalothin; Cefuzonam Cefotaxime* (Claferan) Cefmepidium
Cefadryl) Cefotium* Cefpimizole Cefovecin
Cefapirin*(cephapirin; Cefacler* (cecler, Cefpodoxime* (Vantin) Cefoxazole
Cefadryl) Distaclor, Keor, Cefpiramide* Cefrotil
Cefatrizine* Raniclor) Cefteram Cefsumide
Cefazaur Ceftibuten* (Cedax) Ceftioxide
Cefazedone* With antianaerobic Ceftiofur Ceftobiprole
Cefazolin* activity Ceftiolene (Previously BAL 9141 &
Ro 63-9141)
(Cephazolin; Ancef, Cefoxitin Ceftizoxime* (Cezax) Cefuracetime
Kefzol) Cefmetazole* Cefsulodin* Ceftriaxone*
(Rocephin)
Cefradine* cefotetan Antipseudomonal
(Cephradine; Velosef) Ceftazidime* (Fortum,
Cefroxadine* fertaz)
Ceftezole* Cefoperazone
* Antibacterials for systemic use.
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
b-lactams in clinical practice 85
three stages (Mandell and Petri 1996). In the rst stage,
which takes place in cytoplasm, the product Uridine
diphosphate (UDP)-acetylmuramyl-pentapeptide accu-
mulates in the cells when subsequent synthetic stages
are inhibited. e last reaction in the synthesis of this
compound is the addition of a dipeptide, D-analyl-
D-alanine. Synthesis of the dipeptide involves prior
racemization of L-alanine and condensation catalyzed
by D-alanyl-D-alanine synthetase. During the second
stage reactions, UDP-acetylmuramyl-pentapeptide and
UDP-acetylglucosamine are linked, with the release
of uridine nucleotides, to form a long polymer. In the
third stage, the transpeptidation reaction occurs outside
the cell membrane, though the transpeptidase itself is
membrane bound. e terminal glycine residue of the
pentaglycine bridge is linked to the fourth residue of the
pentapeptide (D-alanine), releasing the fth residue
(also D-alanine). It is this last step in the peptidoglycan
synthesis that is inhibited by the B-lactam antibiotics.
Stereo models reveal that the conformation of penicillin
is very similar to that of D-analyl-D-alanine (Waxman
et al. 1980; Kelly et al. 1982). e transpeptidase is
probably acylated by penicillin, i.e., penicilloyl enzyme
is formed with cleavage of the −CO-N- bond of the
B-lactam ring (Mandell and petri 1996).
e targets for the action of penicillins and cepha-
losporins are collectively termed as penicillin-binding
proteins (PBPs) (Spratt 1980; Ghuysen 1991). e
higher molecular weight PBPs of E. coli (PBP 1A and 1B)
includes the transpeptidases responsible for synthesis
of peptidoglycan. Enzymes inhibited by B-lactams are
PBPs (these are transpeptidases and are involved in nal
transpeptidation step in synthesis of peptidoglycan).
Bacteria have multiple PBPs for eg. E.coli has seven,
each with a distinct role. PBP 1A and B are important in
cell elongation, PBP 2 maintain rod shape of cell wall,
PBP 3 forms septum between dividing cells. PBPs 4, 5,
and 6 are non-essential or redundant, since mutants
lacking any of them were viable. Individual PBPs have
dierent sensitivity with individual B-lactams, although
at clinical doses most drugs bind to more than one PBP
(Tomasz 1986). Inhibition of the transpeptidases causes
spheroplast formation and rapid lysis. However, inhibi-
tion of other PBPs may cause delayed lysis (PBP 2) or
the production of long, lamentous forms of bacterium
(PBP 3). Non-lytic killing by penicillin may involve
holin-like proteins in the bacterial membrane that col-
lapse the membrane potential (Bayles 2000).
Newer cephalosporins
RWJ-54428
RWJ-54428 is a parenteral cephalosporin (Fig. 4) with
a high level of activity against Gram-positive bacteria
including MRSA (Hecker et al. 2000; Chamberlandt et al.
2001). RWJ-54428 was also found to be the most active
agents against drug-resistant enterococci. e potency
of this new cephalosporin against MRSA is related to a
high anity for Penicillin binding protein 2a (PBP 2a)
(Dudley et al. 2003). RWJ-54428 also displayed excel-
lent anity for PBP 5 from Enterococcus hirae R40, with
a MIC50 of 0. 8mg/L. RWJ-54428 also showed stability to
hydrolysis by puried type A B-lactamase isolates from
S. aureus PC1. RWJ-54428 was 8-fold more active than
vancomycin and 32-fold more active than ampicillin
against vancomycin susceptible E. faecalis (Hecker et al.
2000). Vancomycin resistant E. faecalis remained sus-
ceptible to RWJ-54428, which was found to be 1000-fold
more active than vancomycin against these isolates. e
potent activities of RWJ-54428 against B-lactam resist-
ant Staphylococci and enterococci were correlated with
high anities for Staphylococcal PBP 2a and PBP 5 from
enterococci, both of them being major determinants of
resistance to B-lactam antibiotics. e enhanced bind-
ing of RWJ-54428 to MRSA PBP 2a is correlated to a
pharmacophore that is structurally distinct from that of
other reported anti- MRSA B-lactam (Hecker etal. 2003).
e pharmacophore of RWJ-54428 also provides bind-
ing anity to E.hirae PBP 5, which shares some com-
mon features with MRSA PBP 2a and E.faecium PBP 5
(Hartman and Tomasz. 1984; El Kharroubi et al. 1991).
Recent surveys of the susceptibilities of multidrug-resist-
ant Gram-positive bacteria to RWJ-54428 indicate that
this agent has the potential to be a useful agent against
Gram-positive bacteria. However, this compound cur-
rently does not appear to be in active development.
S-3578
S-3578 is a parental cephalosporin with a chemical for-
mula (6R,7R)-7-[(Z)-2-(5-amino-1,2,4-thiadiazol-3-yl)-
2-ethoxyiminoacetylamino]-3-{1-[3-(N-methylamino)
pr op yl]}-1H-imi dazo(4,5- b]pyrid ine-4 -i um-4- yl )
methyl-8-oxo-5-thia-1-azabicyclo[4.2.0] Oct-2-ene-2-
carboxylate monosulfate. It is an inner salt monosulfate
with an imidazopyridinium group in the side chain at
position 3 and displays broad and potent activities in vitro
against both MRSA and P. aeruginosa (Yoshizawaet al.
2002; Fujimura et al. 2003). is may be attributed to:
(1) S-3578 possesses better bactericidal activity than
H
N
N
S
S
S
NH2
Cl
S
N
H2N
N
OH
O
O
CO2H
N
CH3SO3H
Figure 4. RWJ-54428 (MC-02, 479).
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
86 Shahid et al.
vancomycin, (2) Levels of binding of S-3578 to serum
protein was 22% and that of vancomycin was 47%, hence
the levels of free drug are responsible for greater ecacy
(Tsuji et al. 2003). Anti-MRSA ecacy of S-3578 may also
be related to structural aspects, such as a new side chain
at position 3 rather than at position 7 on the cephem
nucleus. is compound was suggested as a promising
new cephalosporin for the treatment of polymicrobial
infections or superinfection caused by Gram-positive
and Gram-negative bacteria including MRSA and P. aer-
uginosa, however, similar to RWJ 54428, this compound
does not appear to be in active development.
Ceftobiprole (BAL 9141)
Ceftobiprole, formerly known as BAL 9141 and RO
63-9141, is a member of class of parenteral pyrro-
lidinone -3- ylidemethyl cephalosporin. BAL 9141 has
documented activity against methicillin-resistant (MR)
staphylococci (MIC90 2- 4 mg/L), Enterococcus faecalis
(MIC90 4 mg/L) and penicillin resistant pneumococci
(MIC90 2 mg/L), while preserving anti-Gram-negative
activity of third and fourth generation cephalosporin.
BAL 9141 exhibits in vivo cidal activity with a moderate
post-antibiotic effect (Davies et al. 2006). In addition
to inhibiting normal complement of PBPs in most spe-
cies, it also binds with high affinity to acquired PBP2a
(PBP2’) of MRSA strains, PBP2a of S. epidermidis and
PBP2x of penicillin resistant S.pneumoniae (Hebeisen
et al. 2001; Davies et al. 2006; Lovering et al. 2006)
while it does not bind to PBP5 of E.faecium. Signature
activity of ceftobiprole is its activity against MRSA,
due to combination of potent binding to PBP2a and
stability to Staphylococcal penicillinase (Hebiesen
et al. 2001). Ceftobiprole was also shown by Hebeisen
et al. (2001) to be measurably hydrolysed by TEM-3
and TEM-4 ESBLs. For AmpC producing organisms,
potent activities of ceftobiprole and cefepime were
not directly correlated with B-lactamase stability,
suggesting that ceftobiprole may possess other prop-
erties, such as rapid penetration into periplasm of
Gram-negative organisms. It was found to be refrac-
tory to hydrolysis by common Staphylococcal PC 1,
the classA TEM-1 and class C AmpC B-lactamase but
was labile to hydrolysis by class B and D and class A
ESBLs (Queenan et al. 2007).
In vitro activities, as reported in recent studies,
conrmed the potency and promising activity of BAL
9141 against MR-Staphylococci including multidrug-
resistant strains. MICs of ceftobiprole for MRSA ranged
from 0.12 to 4 mg/L, with MIC90 values of 2mg/L gen-
erally observed (Jones et al. 2002; Bogdanovich et al.
2005; Davies et al. 2006). Ceftobiprole is poorly hydro-
lysed by PC 1 B-lactamase of S. aureus (Hebiesen et al.
2001), resulting in ceftobiprole MIC90 value of 0. 5mg/L
for methicillin-sensitive Staphylococcus aureus (MSSA)
strains (Jones et al. 2002). It also showed excellent
activity against penicillin-resistant S. pneumoniae
(MIC90 0. 25 mg/L), vancomycin susceptible and resist-
ant E. faecalis (MIC90 4 mg/L), penicillin-resistant
viridans group Streptococci (MIC90 1 mg/L), H. inu-
enzae (MIC90 0.06–0.5mg/L) and M. catarrhalis (MIC90
0.5mg/L ) (Queenan et al. 2007). Ceftobiprole was also
found to be active against pathogenic Neisseria (MIC90
0.004–0. 006 mg/L) and Gram-positive anaerobes (MIC90
< 0. 25 mg/L).
BMS-247243
BMS-247243 is a B-lactam with improved anity
for PBP2a (a mecA gene product), the target enzyme
responsible for methicillin resistance in Staphylococci.
It is bactericidal for MRSA, killing the bacteria twice as
fast as vancomycin. BMS-247243 has cephalosporin
nucleus and unique features on C-7 and C-3 (Fung
Tomc et al. 2002). At C-7, dichloro group imparts
lipophilicity (necessary for good anti- MRSA activity).
At C-3, the morpholinium quaternary component is
important for potency and dimethyl group contribute
to improved solubility. As reported in recent study
(Fung Tomc et al. 2002), MIC90 of BMS-247243 for
methicillin-susceptible Staphylococci ranged from <
0.25 to 1 mg/L while MIC90 for B-lactamase produc-
ing S.aureus strains was four fold higher than for
B-lactamase non-producing strains. e anity of
BMS-247243 for PBP 2a was >100-fold better than that
of methicillin or cefotaxime. Hence it may prove to be
useful in the treatment of infections caused by multi-
drug resistant organisms, especially MRSA, S. epider-
midis, and S. haemolyticus.
Ceftaroline (PPI-0903)
Ceftaroline fosamil which was formerly known as PPI-
0903 and TAK-599 is an N-phosphano-type prodrug
cephalosporin and upon hydrolysis of phosphonate
group it is released in its active form in vivo (Iizawa et al.
2004; Sader et al. 2005; Sader et al. 2008). Ceftaroline
has potent activity against MRSA, and many other
Gram-positive organisms, as well as against Gram-
negative bacilli (Sader et al. 2005; Jacqueline et al. 2007;
Mushtaq et al. 2007; Sader et al. 2008). Staphylococcus
spp., including MRSA and MRCoNS were reported to
be susceptible to ceftaroline with an MIC90 ranging
between 0.25- 2mg/L (Iizawa et al. 2004; Sader et al.
2005). Ceftaroline has also been reported to posses
potent activity against vancomycin-resistant MRSA
strains, multidrug-resistant S. pneumoniae strains, and
Haemophilus inuenza (Kurazono et al. 2004; Sader
et al. 2006).
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
b-lactams in clinical practice 87
In a more recent study (Sader et al. 2008), ceftaroline
showed an MIC50 and MIC90 of 0.5 mg/L, and the highest
MIC was found as 1 mg/L against community acquired
methicillin-resistant S. aureus (CA-MRSA) strains. In
that study performed on 152 geographically diverse
strains, 97.4% strains had an MIC of 0.5 mg/L, and the
MICs ranged between 0.25 - 1 mg/L in the total study
population (Sanders et al. 2008). us, to conclude, the
preliminary results on this investigational anti-MRSA
cephalosporin demonstrated that it could be a potent
antimicrobial for the treatment of both community- and
hospital-acquired infections.
Mechanism of resistance
e signicantly high consumption of B-lactams
exerts considerable selection for resistance to these
antibiotics. e resistance may occur via one of these
mechanisms:
(a) modication of normal penicillin binding proteins
(PBPs)
(b) bypassing of the normal PBPs
(c) impermeability of outer membranes to drugs
(d) production of B-lactamases
(e) ability to pump out (eux) B-lactams
Eux system
Drug eux system pump out antibiotics from bacteria
in an energy dependent manner using transporters clas-
sied as follows:
- Major Facilitator Superfamily (MFS)
- Adenosine triphosphate binding cassette (ABC)
- Small Multidrug Resistance (SMR)
- Resistance Nodulation Cell Division (RND)
- Multidrug and toxic Compound Extrusion (MATE)
e transporters used could be single component
transport across the cytoplasmic membrane or multi-
component pumps-transport substrate in conjunction
with a periplasmic membrane fusion protein (MFP)
component and an outer membrane protein (OMP)
component across the entire cell envelope (Butaye et al.
2003). In Gram-negative bacteria, RND pumps are most
prominent because of their major role in the develop-
ment of acquired and intrinsic resistance to a number
of antibiotics and the transportation of biocides, dyes,
detergents and organic solvents out of cell. Genome of
E.coli has the AcrAB-Tol C System, P. aeruginosa has the
Mex AB-OprM, Mex CD-Opr J, Mex EF-OprN, Mex XY,
Mex JK, Mex GHI-Opm D and Mex VW Systems. While
other Gram-negative bacteria, such as B.cepacia com-
plex with emerging resistance in cystic brosis patients,
S. maltophilia and Neisseria gonorrhae may have Ceo
AB-Opc M, Amr AB-Opr A, Sme ABC, Sme DEF or Mtr
CDE Systems, respectively (Jo et al. 2003).
Modication of PBPs and bypassing are the most
important mechanisms of resistance in Gram-
positive cocci, but the hydrolysis of B-lactam rings by
B-lactamases is the prevalent mechanism in Gram-
negative bacterial species (Livermore 1998).
b-lactamases and resistance
e common form of resistance is either through lack
of drug penetration [outer membrane protein (OMP)
mutation and eux pumps], ESBL production including
hyperproduction of an AmpC type B-lactamase, and/
or carbapenem hydrolyzing B-lactamases. Increased
clinical use of B-lactams has selected for B-lactamase-
producing organisms. Sequencing of B-lactamases
allows them to be divided into four classes, A to D. Class
A, C, and D function by a serine ester hydrolysis mecha-
nism. Whereas class B B-lactamases function using
a zinc ion to attack B-lactam ring. Attempts to classify
B-lactamases began in the late 1960s and the rst scheme
to achieve wide acceptance was proposed by Richmond
and Sykes (1973). is scheme was based on whether
an enzyme hydrolyzed penicillin more or less rapidly
than cephaloridine, and whether its activity was inhib-
ited by cloxacillin and/or p-chloromercuribenzoate.
Subsequently, Bush proposed a major revision in 1989
and updated in 1995 (Bush et al. 1995). Another type of
B-lactamases called SHV-type ESBLs is found frequently
in clinical isolates than any other type of ESBL. SHV
refers to sulfhydryl variable, and this designation was
made because it was thought that the inhibition of SHV
activity by p-chloromercuribenzoate was substrate-
related and was variable according to the substrate used
for the assay. SHV-type ESBLs have been detected in a
wide range of Enterobacteriaceae and outbreaks of SHV-
producing Pseudomonas aeruginosa and Acinetobacter
spp. have also been reported. TEM-type ESBLs are deriv-
ative of TEM-1 and TEM-2. TEM-1 was reported in 1965
from an E. coli isolate from a patient in Athens, Greece,
named Temoniera (hence the designation TEM). TEM-1
is able to hydrolyze ampicillin at a greater rate than
carbenicillin, oxacillin, or cephalothin, and has negligi-
ble activity against extended-spectrum cephalosporins.
It is inhibited by clavulanic acid (readers are encouraged
to read Paterson and Bonomo 2005, for description of
TEM- and SHV-ESBLs in greater detail). In recent years,
CTX-M types ESBLs are expanding rapidly. e name
CTX reects the potent hydrolyzing activity of these
B-lactamases against cefotaxime. Organisms produc-
ing CTX-M type B-lactamases typically have cefotaxime
MICs in the resistant range (>64mg/L), while ceftazi-
dime MICs are usually in the apparently susceptible
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
88 Shahid et al.
range (2 to 8 mg/L). However, some CTX-M type ESBLs
may actually hydrolyze ceftazidime and confer resist-
ance to this cephalosporin (MICs as high as 256 mg/L)
(Baraniaket al. 2002; Poirel et al. 2002). CTX-M type
B-lactamases hydrolyze cefepime with high eciency,
and cefepime MICs are higher than observed in bacteria
producing other ESBL types. It appears that CTX-M type
B-lactamases are closely related to B-lactamases of kluy-
vera spp. ( Decousser et al. 2001; Di Conza et al. 2002;
Humeniuk et al. 2002; Poirel et al. 2002). For example, a
chromosomal B-lactamases gene of Kluyvera georgiana
encoded an ESBL, KLUG-1, which shares 99% amino
acid identity with CTX-M-8 (Poirel et al. 2002). CTX-M-
type B-lactamases have 40% or less identity with TEM
and SHV-type ESBLs.
Two types of carbapenem-hydrolyzing enzymes have
been described, (1) serine enzymes possessing a serine
moiety at the active site, (2) and metallo-B-lactamases
(MBLs), requiring divalent cations, usually zinc, as
metal cofactor for enzyme activity (Bush et al. 1995;
Bush 2001). e serine carbapenemases are invariably
derivative of class A or class B enzymes and usually
mediate carbapenem resistance in Enterobacteriaceae
or Acinetobacter spp. Carbapenemases characterized
from Enterobacteriaceae include Nmc A, Sme-1, Sme-2,
Sme-3, Imi-1, KPC-1, KPC-2, KPC-3, and GES-2 (Yang
et al. 1990; Nordmann et al. 1993; Rasmussen et al. 1994;
Queenan et al. 2000; Poirel et al. 2001; Yigit et al. 2001).
Despite the avidity of these enzymes for carbapenems,
they do not always mediate high-level resistance, and
clavulanate inhibits not all of them. All MBLs hydrolyze
imipenem, but their ability to achieve this varies consid-
erably and rate of hydrolyses may or may not correlate
with the bacterium’s level of resistance to carbapenem
(Massidda et al. 1991). e zinc ions, in MBLs, in turn
usually coordinate two water molecules necessary for
hydrolysis (Rasmussen and Bush 1997). e principal
zinc-binding motif is histidine-x-histidine-x-aspartic
acid (HXHXD) (Rasmussen and Bush 1997) and as they
are zinc dependent, their activities are inhibited by
EDTA. Unlike serine B-lactamases, MBLs possess a wide
plastic active site groove and accordingly can accom-
modate most B-lactam substrate, facilitating their very
broad spectrum of activity.
IMP-1, IMP-2, IMP-4, IMP-7, IMP-9, IMP-10, IMP-11,
IMP-16, and IMP-18 has been identied in P. aeruginosa
isolates while A. baumanii strains were also found to pos-
sess variety of IMP type MBLs including IMP-1, IMP-2,
IMP-4, IMP-5, IMP-6, and IMP-11. Other isolates in
which IMP MBLs were detected include Shigella exneri,
Serratia marcescens, Pseudomonas putida, Alcaligenes
spp., Enterobacter cloacae, Klebsiella oxytoca, K. pneu-
moniae, Proteus spp., and Providencia spp. etc. (Walsh
et al. 2005). IMP-3 may actually be the progenitor of
IMP-1 rather than being a variant of IMP-1 (Iyobe et al.
2000). Another dominant group of acquired MBLs is the
VIM type enzymes. VIM-1 (Varonese Imepenemase)
was described rst in Verona, Italy from a P. aeruginosa
isolates (Lauretti et al. 1999). Later VIM-1 has also been
detected in E. coli (Scoulica et al. 2004) and in several K.
pneumoniae isolates in Greece (Giakkoupi et al. 2003),
and very recently VIM-1 positive K. pneumoniae isolates
has been detected in France (P. Nordmann, unpublished
data) that was associated with the ESBL SHV-5. VIM-2
producing P. aeruginosa strains have also been reported
from other countries such as Japan, South Korea,
Portugal, Spain, Poland, Croatia, Chile, Venezuela,
Argentina, Belgium, and recently in the United States
(Prats et al. 2002; Yum et al. 2002; Mendes et al. 2004).
VIM-3, a novel variant of VIM series has been identied
inP. aeruginosa isolates in Taiwan (Yan et al. 2001). VIM-4
was reported from P. aeruginosa from Greece. VIM-5,
VIM-6 and VIM-7 have also been characterized. VIM-7
has been isolated from carbapenem-resistant P. aerugi-
nosa isolate from Houston, Texas (Toleman et al. 2004),
and shares 77% identity with VIM-1 and 74% with VIM-2
type B-lactamase. Although its sequence is very dierent
from that of other VIMs and probably arose from a dif-
ferent ancestral source (Toleman et al. 2004). Moreover,
new MBL-types, including SPM, GIM, SIM have also
been described in recent years (Walsh 2002; Castanheira
et al. 2004; Lee et al. 2005). SPM-1 (Sao Paulo MBL) was
isolated from P. aeruginosa isolate in 1997 (Walsh 2002),
showed highest identities with IMP-1 (35.5%), ImiS
(32.2%), CphA (32.1%), BCII (30.0%), and Ccr (27.0%)
(Walsh 2002). Sequence of SPM-1 diers signicantly
from that of IMP and VIM, not least due to presence of an
insertion of 24 amino acids just after the active site. is
insertion has been shown to be very exible and acts as a
“loop”, probably augmenting the binding and hydrolysis
of B-lactams (T. R. Walsh, unpublished data). Preferred
substrates of SPM are penicillins, ampicillin, piperacil-
lin, carbenicillin, azlocillin, and cephalothin (Livermore
1995; Koh et al. 2001; Docquier et al. 2003; Giakkoupi
etal. 2003). Like IMP-1 and VIM-1, SPM-1 does not hydro-
lyze clavulanate or aztreonam. In general, SPM-1 binds
cephalosporins more tightly than penicillins. GIM-1
(German Imipenemase), a novel class B B-lactamase,
was isolated from P. aeruginosa (Castanheira etal. 2004)
and displayed identity with IMP-1, IMP-4 and IMP-6
(43.1%, 43.1%, and 43.5%, respectively) while it showed
31.2% identity to VIM-7, 28.8% to VIM-1, VIM-4, and
VIM-5 and only 28.0% similarity with SPM-1. It possess
major consensus feature of MBL class B1 family, such as
principal zinc-binding motif (HXHXD) and possess two
zinc at its active site (Rasmussen and Bush 1997). GIM-1
demonstrates a hydrolytic prole similar to IMP-1 but is
a weaker enzyme.
AmpC B-lactamases, belonging to Ambler’s molec-
ular class C and group 1 of Bush-Jacoby-Medieros
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
b-lactams in clinical practice 89
functional classication (Bush et al. 1995), are a large
group of enzymes of broad substrate specicity. ey can
degrade penicillins and most cephalosporins (including
7-A-methoxy derivatives) and are poorly inhibited by
serine B-lactamase inactivators. AmpC B-lactamases
are usually not active or very poorly active on aztre-
onam, the zwitterionic oxyimino-cephalosporins (like
cefepime and cefpirome), and imipenem, but can
contribute to decreased susceptibility or even resist-
ance to these compounds especially when enzyme
overproduction is associated with permeability defects
or drug eux (Hanson 2003; Toleman et al. 2006). A
number of AmpC B-lactamase are encoded by chromo-
somal genes resident in some Gram-negative pathogens
(P. aeruginosa, Acinetobacter spp., and several members
of Enterobacteriaceae), while others are encoded by
genes associated with mobile DNA elements that can
be acquired by horizontal gene transfer (Hanson 2003).
ISCR1 is recognized as a very important element in the
mobilization of many dierent antibiotic resistance
genes, including plasmid-mediated AmpC B-lactamase
genes (Toleman et al. 2006).
Other important types of ESBLs are OXA-type ESBLs.
ese are named because of their oxacillin-hydrolyzing
abilities and they were characterized by hydrolysis
rates for cloxacillin and oxacillin being greater than
50% in comparison to that for benzyl penicillins (Bush
et al. 1995). OXA-1, which is most common OXA-type,
has been found in 1-10% of E. coli isolates (Hanson
2003). Most OXA-type-B-lactamases do not hydrolyze
the extended-spectrum cephalosporins to a signicant
degree and are not regarded as ESBLs however OXA-10
hydrolyze cefotaxime, ceftriaxone and aztreonam.
Other OXA-type ESBLs include OXA-11, -14, -16, -17,
-19, -15, -18, -28, -31, -32, -35 and -45 (Toleman et al.
2003). e oxacillinases that have been character-
ized from A. baumanii includes OXA-23 to OXA-27,
OXA-40, and OXA-48 (Livermore and Woodford 2006).
ese enzymes confer resistance to cefotaxime and
sometimes ceftazidime and aztreonam (Danel et al.
1998; Danel et al. 1999). e evolution of OXA-type
B-lactamases from parent enzymes with narrower
spectra has many parallels with evolution of SHV- and
TEM-type ESBLs.
Eorts to combat resistance due to
b- lactamases
As the resistance has spread, pharmaceutical chem-
ists have responded by developing compounds with
increased stability, or by protecting labile agents by
inhibiting the B-lactamases. Stability to B-lactamases
can be achieved by attaching to the B-lactam ring a
substituent that hinders access of the active-site serine
or that displaces water from preventing completion of
hydrolysis (Frere 1995).
Strategies to achieve these include
(a) attaching a bulky acyl group to the amino group of
6-aminopenicillanic acid, or 7-aminocephalospo-
ranic acid (see Fig. 5 for the acyl groups attached
in some of the representative B-lactams)
(b) replacing the hydrogen on carbon 6 (penicillins)
or 7 (cephalosporins) with an A-methoxy group
(Abraham 1987).
Although responded by the pharmaceutical chemists,
these new agents, in turn, have selected for new forms of
B-lactamase-mediated resistance. us, it appeared to be
as a cyclic process of drug development and the emergence
of resistance. Some of the representative B-lactamases and
their preferred substrate are shown in Table 2.
B-lactamase stability was more dicult to achieve in
B-lactams that have activity against Gram-negative spe-
cies than in those directed against Gram-positive because
Gram-negative organisms produce a wider variety of
B-lactamases than Gram-positive organisms (Tomasz
1994). Incorporating broad-spectrum stability proved
particularly dicult with penicillins, the only one with
such stability being temocillin, the 6-alpha-methoxy
analogue of ticarcillin. Benzylpenicillin and cephalothin
Figure 5. Side chain substitution to confer B-lactamase stability.
e gure above shows bulky acyl groups attached to amino group of
6-APA or 7-ACA of some of the representative B-lactam antibiotics.
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
90 Shahid et al.
are labile to many B-lactamases but methicillin and
cefotaxime, with bulkier substituents, are more stable.
e development of B-lactamase-stable cephalosporins
has been more successful. An oxyimino-aminothiazolyl
7-acyl side chain makes cephalosporin stable to TEM-1
and SHV-1 enzymes. Such groups are incorporated in
most third-generation cephalosporins. A 7-A-methoxy
group (as in cefoxitin, cefmetazole, and latamoxef) con-
fers stability to TEM-1 and SHV-1 enzymes and to the
CepA chromosomal B-lactamase of Bacteroides fragilis
(Livermore 1998). Similarly, addition of an A-methoxy
group at position 6 (penicillins) or 7 (cephalosporins)
provides stability against many B-lactamases. Ticarcillin
and cephalothin, which lack this substituent, are labile
to many B-lactamases but their A-methoxy analogues
temocillin and cefoxitin are more stable.
During the mid 1980s, a threat to B-lactamase sta-
ble oxyimino aminothiazolyl cephalosporins became
apparent, with the emergence of extended-spectrum
B-lactamases (ESBLs). Most of the ESBLs are mutants of
the classic TEM-1, TEM-2 and SHV-1 types (Jacoby and
Medieros 1991; Du Bois et al. 1995). ese ESBLs have
one to four amino acid replacements compared with
their parent enzymes, and these changes suciently
remodel the active site to allow attack on aminothiazolyl
compounds (Livermore 1998).
Other B-lactam antibiotics with broader spectrum of
activity are described in the following section.
Carbapenems
Carbapenems (Imipenem and meropenem) are
B-lactams that contain a fused B-lactam ring and a
5-membered ring system that diers from the penicil-
lins in being unsaturated and containing a carbon atom
instead of the sulphur atom.
Chemistry of carbapenems
Imipenem (Fig. 6a) is derived from a compound
(thienamycin) produced by Streptomyces catt-
leya. ienamycin is unstable, but imipenem, the
Table 2. Classications of B-lactamases.
Structural
class(Ambler)
Functional
class (Bush
et al.**)
Richamond and
Sykes class#Preferred substrate
Inhibited by
Clavulanate;Aztreonam;EDTA Representative enzymes
Serine B-lactamases
A 2a NL Penicillin ++ - - Staphylococcal
penicillinase
2b II and III Penicillin, Cephalosporin ++ - TEM-1,TEM-2,SHV-1
2be III and IV Penicillin, Cephalosporin,
Aztreonam, Monobactam
++ - - TEM-3 to TEM-29,SHV-2
to SHV-7 K.oxytoca K1,
PER1
2br NL Penicillin - - - TEM-30 to TEM-41,TRC-1,
2c II and V Carbenicillin, Penicillin + - - PSE-1,PSE-3,PSE-4,BRO-
1,BRO-2
2e IC Penicillin, Carbenicillin
Aztreonam, Cephalosporin
++ - - Cephalosporinases
from P.vulgaris FEC-I,
Bacteroides Cep A
2f NL Penicillin, Aztreanem,
Imipenem
+ - - Carbapenemase Imi-1,
NMC-A, Sme-I
C 1 I, except Ic Cephalosporin, Penicillin - ++ - AmpC enzymes from
Gram-negative bacteria,
MIR-I.
D 2d V Oxacillin, Penicillin V - - Oxa-1 to Oxa-11,13-15
Aeromonas OXA-12,PSE-2
Undetermined* 4* NL Penicillin, Carbenicillin,
Oxacillin
- - - Penicillinase from
Burkholderia cepacia,,
SAR-2
Zinc
B-lactamasesB
3 NL Penicillin, Carbenicillin,
Oxacillin Cephalosporin,
Imipenem
- - ++ L1 from
Stenotrophomonas
maltophilia, IMP-I,
Aeromonas hydrophilia
A 2h, Aeromonas A2s,
B. fragilis Cer A, P.
aeruginosa pMS 350.
NL = Not listed* None of Bush’s group 4 enzymes has yet been sequenced. V = Varies within group. ** For a comprehensive listing of dierent
B-lactamase types, see Bush et al. 1995. # For detailed classication, see Richmond and Sykes 1973.
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
b-lactams in clinical practice 91
Figure 6. Newer carbapenems.
N
HH
C
O
COOH
SCH2CH2NHCH
H
HO
H3C
NH
(a) Imipenem
S
N
O
N
H
H
N
O
HO CH3
CO2Na
(b) Ertapenem
CO2Na
NSNH
SNH2
O
O
H
H
CH3
H
HH
H
HO
O
H2O
H
N
H3C
CO2H
(c) Doripenem (formerly S-4661)
R
S
N
H
H
O
R:
HO Me
NH2
NH
NH-Me
CONH2
NH2
NH2
J-114 ,871
CONH2
(d) trans-3,5 disubstrituted 1B-methyl carbapenems
J-114,870
J-111,225
J-111,347 (Prototype)
COOH
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
92 Shahid et al.
(e) 1B-methyl-2-(thiazol-2-ylthio) carbapenem
S
N
H
H
O
N
S
R
R:
Me
HO Me
COOH
OH
Me
NH
NH
H
N
SM-232724SM-197436 SM-232721
(f) CS-023.
N
S
O
H
NN
H
HO
CON
NHCO NH2
NH
CO2H
(g) Faropenem
N
SO
OHO
O
H
H
OH
H
(h) Tebipenem
NS
CH3
H
HH
O
COOH
N
N
S
H
OH
H3C
N
O
H
O
O
H
S
NN
N
O
OH
NH2
(i) ME-1036
(j) Sanfetrinem
N
O
H
H
OH
O
OCH3
OH
Figure 6. Continued.
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
b-lactams in clinical practice 93
N-formimidoyl derivative, is stable. Imipenem is mar-
keted in combination with cilastatin, a drug that inhibits
the degradation of imipenem by a renal tubular dehy-
dropeptidase. Meropenem is a dimethyl-carbamoyl-
pyrolidinyl derivative of thienamycin. It does not require
co-administration with cilastatin as it is not sensitive to
renal dehydropeptidase. Its in vitro activity and toxicity
is very much similar to imipenem.
Activity and stability to b-lactamases
Imipenem is active against a wide variety, including pen-
icillinase-producing strains, and Listeria. Some strains
of methicillin-resistant staphylococci are susceptible
but many strains are not. Activity is excellent against the
Enterobacteriaceae and most strains of Pseudomonas
and Acinetobacter are inhibited. Meropenem is active
against some imipenem-resistant P.aeruginosa but
less active against Gram-positive cocci. Imipenem and
meropenem have wider B-lactamase stability than
other B-lactams, being eectively resistant to all the
common plasmid encoded class A, C, and D enzymes,
including extended-spectrum TEM and SHV deriva-
tives (Heritage et al. 1999), PER-1 and AmpC enzymes
(Livermore 1998). is stability partly depends on the
presence of a simple trans-6-hydroxyethyl group; ana-
logues with acyl substituents or with 6-hydroxyethyl
in the cis conguration are less stable to B-lactamases
(Christensen 1981).
Unlike new cephalosporins, they are not prone to
attack by B-lactamases types that are widely prevalent,
so B-lactamase-mediated resistance has been slow to
emerge and spread. Nevertheless, carbapenems are
attacked by few B-lactamases, most notably the zinc
(molecular class B) enzymes (Livermore 1998). A seri-
ous concern is the emergence of plasmid-mediated class
B enzymes in Enterobacteriaceae and Pseudomonas.
Such enzymes, including IMP, VIM, and KPC, have been
found in Serratia marcescens,K. pneumoniae,P. aerugi-
nosa,Acinetobacter, and other bacterial species (Osano
et al. 1994; Livermore and Woodford 2006). is enzyme
confers resistance to all B-lactams except monobactams.
Carbapenemase activity also occurs in three closely
related class A B-lactamases, Sme-I (Yang et al. 1990),
Imi (Rasmussen et al. 1994), and NMC-A (Nordmann
et al. 1993), found in Enterobacter cloacae and Serratia
marcescens strains.
ere have been reports of multiple B-lactamase pro-
ductions in a single Gram-negative pathogen (Bradford
et al. 2004; Bratu etal. 2005). Isolates of Klebsiella spp.
producing KPC-2, SHV, and inhibitor-resistant TEM-30
B-lactamases have been reported as endemic in New York
city (Moland et al. 2007). Recently, Moland et al (2007)
reported a K. pneumoniae isolate from New York city that
produced up to eight dierent B-lactamases including
FOX-like plasmid-mediated AmpC B-lactamase, SHV-
1-like, SHV-12-like ESBL, a KPC-like carbapenemase,
OXA-like, TEM-1-like, TEM-30-like, and PSE-1-like
B-lactamase. Currently rising problem includes CTX-M
ESBLs, plasmid-mediated AmpC B-lactamases, and KPC
carbapenemases in Enterobacteriaceae, while OXA- and
metallo-carbapenemases are of growing importance
in Acinetobacter spp. Clones of K. pneumoniae and
E. cloacae with KPC enzymes have spread in multiple
hospitals around New York since 2003 (Livermore and
Woodford 2006).
Newer carbapenems
Carbapenem development continues to discover agents
with greater potency or improved pharmacokinetic
properties (Tsuji et al. 1998). Various newer carbapen-
ems have been discovered, some in recent years, and are
really novel.
Ertapenem
Ertapenem (formerly MK-0826 and L-749, 345) (Fig. 6b)
is a carbapenem that shares the broad spectrum of imi-
penem and meropenem against Enterobacteriaceae,
Gram-positive organisms, and anaerobes, but is less
active against non-fermenters. Ertapenem diers from
meropenem in its 2` substituents, where it carries a
meta-substituted benzoic acid group. Carbon 1 car-
ries a B-methyl group, protecting against hydrolysis
by renal dehydropeptidase I (Livermore et al. 2003).
Ertapenem has a trans hydroxyethyl on the 6-position
and this conguration is critical to B-lactamase stability
in carbapenems.
Like meropenem, ertapenem binds most strongly
to penicillin binding protein (PBP-2) of E.coli, as com-
pared to PBP-3, and has good anity also for PBP-1a
and 1b (Kohler et al. 1999). However in contrast, imi-
penem binds primarily to PBP-2, then 1a and 1b, and
has only weak anity for PBP-3 (Sumita et al. 1990).
Inactivation of PBP-1a and b achieves rapid bactericidal
action without the prior lamentation that occurs with
agents such as third-generation cephalosporins, which
binds primarily to PBP-3. In vitro activities, as reported
in recent studies, showed that MICs of ertapenem for
Enterobacteriaceae fall between 0.008 and 0. 12mg/L
(Livermore et al. 2003). ese values are similar to those
of meropenem, and eight to 16-fold below those of imi-
penem. Like imipenem and meropenem, ertapenem is
active against methicillin-susceptible S. aureus (MSSA)
but not against MRSA. However, ertapenem has only
marginal activity against important non-fermenters
(Livermore et al. 2001; Fuchs et al. 2001). Ertapenem
overcomes most resistance to other B-lactams in
Enterobacteriaceae, but unlike earlier carbapenems,
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
94 Shahid et al.
lacks signicant activity against non-fermenters, limit-
ing its empirical role in nosocomial infections.
Doripenem
Doripenem (Fig. 6c), which was formerly S-4661, is a novel,
broad-spectrum parenteral carbapenem with a chemical
formula (+)-(4R,5S,6S)-6-[(1R)-1-1hydroxyethyl]-4-me-
thyl-7-oxo-3[[3S,5S)-S-(sulfamoylaminomethyl)-pyr-
rolidin-3-yl]thio]-1-azabicyclo[3.2.0]hept-2-ene-2-
carboxylic acid monohydrate. is structure confers
B-lactamase stability and resistance to inactivation by
renal dehydropeptidases. Recent studies have indicated
that doripenem has a spectrum and potency against
Gram-positive cocci most similar to imipenem or ertap-
enem (Tsuji et al. 1998; Curran et al. 2001; Jones 2001;
PanKuch et al. 2002; Hoellman et al. 2003; Livermore
et al. 2003) and a Gram-negative activity like meropenem
but two- or four-fold superior to imipenem (Wiseman
et al. 1995).
Earlier reports on the microbiological/pharmacologi-
cal features of doripenem demonstrated:
1. high anity for PBP targets that are species-specic
(PBP3 in P.aeruginosa, PBPs 1, 2 and 4 in S. aureus,
PBP2 in E.coli (Hanaki et al. 1996)
2. bactericidal action (Inoue et al. 1996; Miwa et al.
1996; Nishino et al. 1996; Sasaki et al. 1994)
3. a post antibiotic eect of 1.8 h (in vitro) to 4.3 h
(in vivo) for P.aeruginosa (Nishino et al. 1996;
Totsuka et al. 1996)
4. inux in Gram-negative species by OprD channels
with eux sensitivity via the MexAB-OprM system
(Yamano et al. 1997)
5. pharmacokinetic parameters resembling mero-
penem (Nakashima et al. 1994)
6. low risk of convulsive side eects (Hori et al. 1997)
7. stability to wide variety of B-lactamases (Inoue et al.
1996; Sasaki et al. 1994)
8. high level stability to human recombinant dehy-
dropeptidase-I (Mori et al. 1996)
9. clinical success from human trials in Japan (Saito
et al. 1997; Arakawa et al. 1997)
Early in vitro development trials clearly placed dorip-
enem as a very broad-spectrum B-lactam, having a
compromised spectrum when used alone (Kobayashi
et al. 1997) against oxacillin-resistant staphylococci, E.
faecium, some Burkholderia spp. and Stenotrophomonas
maltophilia. is spectrum was similar to imipenem
and meropenem (Wiseman et al. 1995; Tsuji et al.
1998) but markedly superior to ertapenem (Jones 2001;
Pankuch et al. 2002; Curran et al. 2003; Hoellman et al.
2003; Livermore et al. 2003 ). In a recent clinical study,
doripenem exhibited potency and /or spectrum advan-
tages compared with both imipenem and meropenem
against the entire group of non-fermentative Gram-
negative bacilli, and for Enterobacteriaceae. is newer
carbapenem was four-to 32 fold more active than imi-
penem (MIC range, < 0.015–0.5mg/L).
trans-3,5-disubstituted Pyrrolidinylthio-1b-
methylcarbapenems
In the course of derivatisation, study of 1B-methyl
carbapenems, a novel trans-3,5 –disubstituted
Pyrrolidinylthio-1-B methyl carbapenem, J-111, 347,
was identied as a broad spectrum agent with activ-
ity against MRSA, as well as other Gram-positive and
Gram-negative organisms, including P.aeruginosa.
Subsequently, the 1B-methylcarbapenems, J -111, 225,
J-114, 870 and J-114, 871 (Fig. 6d) were synthesized from
the prototype J-111, 347 (Hashizume et al. 2000). e
stereochemistry of the side chains in these compounds
is novel. Known Pyrrolidinylthio-1B-carbapenems like
meropenem (Sunagawa et al. 1990), SM-17466 (Sumita
et al. 1995), BO-2727 (Nakagawa et al. 1993), MK-826,
and E1010 (formerly ER-35786) (Ohba et al. 1997) share a
cis-counterpart at the side chains. In a recent study, 90%
isolates of methicillin-resistant S. aureus (MRSA) and
methicillin-resistant coagulase-negative Staphylococci
(MRCoNS) were inhibited by J-111,347 (prototype), as
well as by, J-111,225, J-114,870, and J-114,871 at con-
centrations of 2,4,4, and 4mg/L (MIC-90) respectively
indicating that these agents were 32- to 64-fold more
potent than imipenem, which had MIC90 of 128 mg/L.
Although these drugs were less active in vitro than van-
comycin, which had MIC90s of 1 and 2 mg/L for MRSA
and MRCoNS respectively. However, these new carbap-
enems displayed better kinetics than vancomycin.
2- (iazol – 2 – ylthio) - 1b – methyl carbapenems
SM-197436, SM-232721, and SM-232724 are new
1B–methyl carbapenems with a unique 4- substituted
thiazol –2– ylthio moiety at the C-2 side chain (Fig. 6e).
ese newer carbapenems are subsequent modica-
tion of 2–(4-arylthiazol-2-ylthio)-1 B-methyl carbap-
enems. SM-17466 and its derivatives exhibited potent
anti-MRSA activity corresponding to a high anity for
PBP 2a. Although SM-17466 showed good ecacy in a
murine systemic infection model of MRSA, it did not
show sucient activity against E. faecium and thus
prompted for the search of new carbapenems (Sumita
et al. 1995). ese newer carbapenems listed above
are novel carbapenems having a ve or six member
cyclic amino group instead of the pyridium moiety of
SM-17466 and showed improved activity against E.fae-
cium. In recent studies (Ueda and Sunagawa 2003), the
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
b-lactams in clinical practice 95
novel carbapenems were found active against MRSA and
MRSE with MIC90 of < 4mg/L. e MICs of SM-197436,
SM-232721, and SM-232724 for Streptococci including
penicillin resistant S.pneumoniae strains, ranged from
< 0.063 to 0. 5mg/L. ese drugs were the most active
B-lactams tested against E.faecium, and the MIC90S
for ampicillin resistant E.faecium ranged between 8
and 16 mg/L, which were slightly higher than the value
for linezolid. However, time-kill assays revealed the
superior bactericidal activity of SM-232724 compared to
those of quinupristin / dalfopristin and linezolid against
E. faecium strain with a 4-log reduction in CFU at four
times the MIC after 24h of exposure to antibiotics. Among
the Gram-negative bacteria, these carbapenems were
highly active against Haemophilus inuenzae,Moraxella
catarrhalis,Bacteroides fragilis, and showed antibacte-
rial activity equivalent to that of imipenem for E.coli,
K.pneumoniae, and Proteus spp. ough require further
evaluation, these carbapenems seem to be promising
to treat nosocomial infections caused by Gram-positive
and Gram-negative bacteria including MRSA or VRE.
CS-023 (RO 4908463)
CS-023 (RO 4908463, formerly known as R-115685)
(Fig. 6f ) is a new parenteral 2- substituted 1 B-methyl
carbapenem that has unique guanidine–pyrrolidine side
chain and binds with high anity to PBP1, PBP2 and
PBP4 from S. aureus ATCC 6538P and PBP1a, PBP1b,
PBP2 and PBP3 from E. coli NIHJ (Spratt 1980). CS-023 is
more stable to hydrolysis by human renal dehydropepti-
dase-I than meropenem or imipenem and is bactericidal
against P.aeruginosa and MRSA and also has broad
spectrum of activity against Gram-positive and Gram-
negative bacteria, including MRSA, penicillin-resistant
Streptococcus pneumoniae, H.inuenzae, members
of Enterobacteriaceae, and P. aeruginosa (Jacoby and
Medieros 1991; Kuwamoto et al. 2003; omson and
Moland 2004; Koga et al. 2005).
In recent studies, it was noted that, overall, CS-023
exhibited activity comparable to that of imipenem
against most isolates of Gram-positive pathogens,
whereas it was similar to meropenem against Gram-
negative pathogens (omson and Moland 2004).
However, the CS-023 was signicantly more potent than
imipenem and meropenem against oxacillin-resistant
S. aureus at MIC of 4mg/L, compared with 32mg/L for
meropenem and > 32mg/L for imipenem. CS-023 was
highly potent against B-lactamase–producing or non-
fermenting Moraxella catarrhalis (MIC 0. 008mg/L),
Haemophillus inuenzae (MIC 0. 06 mg/L) and against
most isolates of Enterobacteriaceae, including E.coli,
Shigella,Salmonella, and Klebsiella (MIC 0. 06mg/L). It
was interesting to note that the CS-023 was 4- to 8-fold
more potent against the isolates of meropenem resistant
P.aeruginosa. e broad spectrum of in vitro activity of
CS-023, in recent studies, suggest potential for therapy
of a wide range of infections. However, further studies
are warranted to determine its detailed clinical ecacy.
CS-023 is predominantly distributed to the extracel-
lular uid in body after intravenous administration
(Davies and Morris 1993). is limited distribution
would be favorable property of an antimicrobial agent,
since most infecting microorganisms resides in extra
cellular space (Shentag 1990). A low protein binding
ratio would also be favorable property for CS-023, since
the protein unbound, free form of CS-023 in plasma is a
pharmacologically active fraction, which achieve rapid
equilibrium with drug in extracellular uid (Shibayama
et al. 2007).
Faropenem
Faropenem medoxil (formerly faropenem daloxate)
(Fig. 6g) is an oral prodrug that is rapidly cleaved, releas-
ing the microbiologically active faropenem following
absorption in plasma. Faropenem is active against the
primary respiratory pathogens, including non–penicillin
susceptible S.pneumoniae and M.catarrhalis (Critchley
et al. 2002). Faropenem is most active B-lactam against
S. pneumoniae with MIC90 of 0. 5mg/L and is four fold
active than amoxicillin clavulanate (MIC90, 2mg/L)
eight-fold more active than cefdinir and cefuroxime
(MIC90, 4 mg/L). Activity of faropenem was aected
by penicillin susceptibility status of isolates, with faro-
penem MIC90 increasing from 0.015mg/L for penicillin
susceptible isolates to 1mg/L for penicillin resistant
strains (Stone et al. 2007). Although faropenem exhib-
ited higher MIC against penicillin resistant S. pneumo-
niae it had activity equivalent to that of telithromycin
and levooxacin (MIC90, 1mg/L) and was more active
than all other B-lactam tested, including amoxicillin-
clavulanate, cefdinir and cefuroxime, with MIC90 of 2, 4,
and 8 mg/L .
Tebipenem
Tebipenem (TBM)-pivoxil (PI) is an oral carbapenem
agent with a 1-(1,3-thiazolin-2-yl) azetidin-3-yl thio
group at C-2 position (Fig. 6h). e agent (previously
reported to be LJC 11,036; Miyazaki et al. 2001) shows high
degree of stability to dehydropeptidase-I and absorption
of the active metabolite, which is converted by esterase,
into blood from intestine. e prominent feature of TBM
is its potent activity against the main causative microor-
ganisms, except metallo B-lactamases-producing path-
ogens and MRSA strains causing RTIs and UTIs (Hikida
et al. 1999; Miyazaki et al. 2001). TBM shows higher
anities for PBP1A and PBP1B and for PBP2X (Hikida
et al. 1999). Inhibition of cell wall synthesis towards long
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
96 Shahid et al.
axis and subsequent cell lysis was observed after treat-
ment with TBM. It showed excellent bactericidal activity
against penicillin-resistant Streptococcus pneumoniae
(PRSP) (Hikida et al. 1999; Miyazaki et al. 2001).
ME 1036 (CP5609)
ME 1036, formerly known as CP5609, is a parenteral
carbapenem with a 7- acylated imidazo [5,1-b] thiazole
-2-yl group directly attached to the carbapenem skel-
eton at C-2 position (Fig. 6i). It showed excellent activity
against MRSA and MRCoNS and is unique in that it also
showed potent activity against penicillin resistant S.
pneumoniae, E. faecalis, H. inuenzae, and members of
family Enterobacteriaceae (Kurazono etal. 2004). e ME
1036 MICs (ranging 0.5 to 4mg/L) for ceftriaxone non-
susceptible strains (ceftriaxone MICs >8 mg/L) were
higher than those for ceftriaxone susceptible strains
(ranging, 0.063 to 1 mg/L) (Kurazono et al. 2004). In
a more recent study, ME 1036 showed an MIC50 of
0. 12mg/L, an MIC90 of 0.25 mg/L, and a highest MIC
of only 0.5mg/L against community-acquired methi-
cillin-resistant S. aureus (CA-MRSA) (Sader et al. 2008).
ME 1036 may be slightly less stable than imipenem and
meropenem, in the presence of AmpC B-lactamases.
Like most other carbapenems, ME 1036 was found to
be inactive against E. faecium and also it had no activ-
ity against P.aeruginosa (ME 1036 might not reach
the targets due to low permeability or the presence
of eux pumps, B–lactamases, and other factors in P.
aeruginosa). ME 1036 inhibited the growth of MRSA
at one-fourth the MIC and this may occur because of
its binding to PBPs (except PBP 2a) as also observed in
case of imipenem.
Recently Hirai et al. (2007) investigated the bind-
ing anity of ME 1036 for various PBPs of E. faecalis
ATCC 29212, E. coli NIHJ JC-2 and H. inuenzae MSC
07274. As compared to other carbapenems, ME 1036
showed much stronger anity to PBPs especially
PBP3 and PBP4 of E. faecalis (IC50 of ME 1036 for PBP3
and PBP4 were 0.09 and 0. 03 mg/L while that of mero-
penem, a comparator drug, for same PBPs were 9.76
and 0. 20mg/L). Binding of ME 1036 to PBPs in turn
indicates its antibacterial activity as MICs of ME 1036
and meropenem for E. faecalis were 0.12 and 4. 0mg/L,
respectively. For H. inuenzae, binding of ME 1036 to
almost all PBPs, more precisely PBP3, was shown to be
much stronger than that of meropenem (IC5 0 of PBP3a
and PBP3b were 0.07 and 0. 06 mg/L, respectively
and that of meropenem for PBP3a and PBP3b were
0. 67 mg/L and 0.44 mg/L, respectively). Antibacterial
activity of ME 1036 can be compared with mero-
penem in terms of their MICs (MICs of ME 1036 and
meropenem were found to be 0. 12 mg/L and 1 mg/L,
respectively). For E. coli, a correlation was observed
between binding anity of ME 1036 for PBP2, PBP4,
PBP7/8, and its antibacterial activity. Hence, ME
1036 is supposed to be highly promising carbapenem
against E. faecalis, E. coli and H. inuenzae due to its
high anity for dierent PBPs.
SMP-601 (PZ-601)
PZ-601 is a novel broad-spectrum carbapenem. High
anity of PZ-601 (SMP-601) for penicillin-binding pro-
teins enhanced its activity against multidrug-resistant
Gram-positive organisms including MRSA, and it
retained the typical carbapenem-like spectrum against
Gram-negative bacteria including Pseudomonas spp.
Paterson et al. (2007) investigated the activity of PZ-601
against ESBL producers, which included (1) wild type
ESBL producers (K. pneumoniae and E. coli isolates
which were known by PCR/sequencing to harbor genes
coding for ESBL but which do not posses currently
known mechanisms of carbapenem resistance. ey
posses various combinations of CTX-M, SHV and TEM
type ESBLs), (2) carbapenem-resistant ESBL producers
(K. pneumoniae and E. coli isolates that were ESBL pro-
ducers as well as carbapenem resistant) and, (3) ESBL
producing E. cloacae. e MIC50 and MIC90 for PZ-601
were found to be 1 mg/L and 4 mg/L, respectively,
while carbapenem resistant-ESBL producers showed
elevated MICs (ranging from 4 mg/L to > 32 mg/L).
ESBL-producing E. cloacae seems to be less susceptible
to PZ-601. Hence PZ-601 was found to be more active
against ESBL-producing K. pneumoniae and E. coli
which are susceptible to currently used carbapenems.
Bouchillon et al. (2007) showed in vitro activity of PZ-601
against various Gram-positive, Gram-negative and
anaerobic microorganisms. MIC50 and MIC90, respec-
tively, were 0.25 mg/L and 1 mg/L (against staphylococci
including MRSA), 1mg/L and 2 mg/L (against E. faecalis
including VRE) and 0.06mg/L and 0.25 mg/L (against
S. pneumoniae including penicillin-resistant strains).
For E. coli and K. pneumoniae MIC50/MIC90 were found
to be 0.25/ 1mg/L and 1/4mg/L, respectively, for ESBL-
producers. PZ-601 also showed better activity against
E. cloacae (MIC50 4mg/L; MIC90 8mg/L) and peptostrep-
tococci (MIC50 < 0.015mg/L; MIC90 0.06mg/L).
In vitro antimicrobial activity of SMP-601 was also
shown by Kanazawa et al. (2007). ey showed that
SMP was broadly active against Gram-negative iso-
lates including BLNAR and CF-R-Proteus spp. MIC90
of E. coli, K. pneumoniae, M. morganii, Proteus spp.
(including cefepime-resistant strains), H. inuenzae
BLNAS (including BLNAR), M. catarrhalis, and N. gon-
orrhoeae were 0.5, 0.5, 2, 0.25, 0.06 (0.12 for BLNAR),
0.03, and 0. 12mg/L respectively. MIC90 of SMP against
S. marcescens (8 mg/L), P. aeruginosa ( 32 mg/L) and B.
cepacia (32 mg/L) were found to be higher than those
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
b-lactams in clinical practice 97
of meropenem and imipenem. MICs of SMP against
E. coli, K. pneumoniae, M. morganii, and Proteus spp.
were comparable or superior to those of imipenem,
whereas M. catarrhalis and N. gonorrhoeae were highly
susceptible to SMP. us SMP proves to be a promising
candidate of a broad-spectrum anti-MRSA carbapenem
agent for serious nosocomial infections.
SMP had potent activity against wide range of Gram-
positive and Gram-negative bacteria however it showed
relatively weak activity against some Gram-negative
organisms. Its activity can be improved by combining it
with dierent antibacterial agents. Fujimoto et al (2007)
showed in vitro activity of SMP-601 in combination
with other antimicrobial agents against Burkholderia
cepacia and Serratia marcescens. MIC range of SMP
was 0.15– 16 mg/L for B. cepacia and 0.25– 16 mg/L for
S. marcescens. Combination of SMP and meropenem or
ciprooxacin was found to be synergistic (in 4% of tested
strains) (Min FIC ≤ 0.5) or additive (0.5< to ≤1) (92% of
tested strains) against B. cepacia. For S. marcescens, the
combination of SMP and amikacin showed synergis-
tic (19% of tested strains) or additive eect. But it was
observed that there occurs antagonism (in 4% of total
strains of B. cepacia tested) in combination of SMP and
minocycline and hence it should not be recommended,
while other combinations could represent an eective
treatment of serious infections caused by Gram-negative
and multidrug-resistant Gram-positive pathogens.
Trinems
Trinems (previously tribactams) have a Carbapenem
related structure but with a Cyclohexane ring attached
across carbon 1 and 2 (Fig. 6j). Sanfetrinem (GV 104326),
which is a trinem B-lactam, can be administered orally
as a hexatil ester. Like imipenem, sanfetrinem was
found to be stable to TEM-1 and TEM-10 enzyme, and
like imipenem and cexime but unlike cefpodoxime,
N
SCH3
CH3
O
OOH
X
X=I or Br
(a) Halogenated penicillanic acid
(b) Olivanic acid
(f) Oxapenems
(g) AVE-1330
(c) Penems (BRL 42715) (d) Clavulanic acid
(e) Sulbactam and Tazobactam
N
O
S
N
H
O
OSO3H
CH3
COOH
CH3
N
N
N
CH3
N
S
O
COOH
H
CH2OH
N
O
OCOOH
H
H
N
S
O
OO
CH3
R
OOH R = CH3Sulbactam
CH2N
N
N
Tazobactam
N
O
O
H
R
1
R
2
R
3
COO
Oxapenem compound
AM-112
AM-113
AM-114
AM-115
R1
OH
OH
H
H
R2
H
H
OH
OH
R3
(CH3)2NH3
CH3
CH3
(CH3)2NH3
N
N
O
S O
O
O
OCONH2
Na
Figure 7. B-Lactamase inhibitors.
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
98 Shahid et al.
also retained activity against P. vulgaris and K. oxy-
toca that hyperproduced potent chromosomal class A
B-lactamase (Livermore 1998). Sme1, NMC-A and an
unnamed enzyme from Acinetobacter spp. increased
the Sanfetrinem MICs by up to 64-fold.
b-Lactamase Inhibitors
Mechanism of action: General
B-lactamases can be found either extracellularly
or within the periplasmic space; in general active
B-lactamases from Gram-positive bacteria are excreted
into the medium. B-lactamase activity in Gram-negative
organism is found primarily in the periplasmic space,
although some leakage of enzyme into the medium can
occur. B-lactamase production lead to resistance to com-
mon B-lactam antibiotics. Protection of B-lactam with an
inhibitor is used as an alternative strategy to overcome
resistance due to B-lactamases. Enzyme inhibitors can
be classied as reversible or irreversible. Reversible
inhibitors are those that bind to an enzyme in such a
manner that enzyme activity may be restored. Inhibitors
of B-lactamases that bind at or close to the active site are
often B-lactams. Although these molecules may act as
inhibitor, they also can be hydrolyzed as substrates. us,
many reversible inhibitors are really poor substrates that
are bound with high anity but are hydrolyzed at low
rates. Irreversible inhibitors may be more eective than
reversible inhibitors in that the eventual result is destruc-
tion of enzymatic activity. e preferred terminology for
inhibitor that renders their targets useless is “inactivator”.
A special subclass is the suicide inactivator, a molecule
that must bind initially at the enzyme active site, but
which is converted into an inactivator through catalytic
action of enzyme itself (Abeles and Maycock 1976).
Several classes of successive inhibitors were found in
the mid 1970s, including clavams, penicillanic acid sul-
phones, halogenated penicillanic acids (Fig. 7a), olivanic
acids (Fig. 7b), and various penems (Fig. 7c), and many
more are in experimental stages. Of these beta-lactamase
inhibitors, clavulanic acid, tazobactam, and sulbactam
are now established molecules that are used in clinic,
and other molecules are still under experimental stages
which include oxapenems, NXL 104 (AVE 1330A), Mono-
and bicyclic-bridged monobactams, BAL 30376, LK-157,
BLI-489, and so on.
Clavulanic acid
Clavulanic acid was the rst suicide inactivator of
B-lactamases (Brown et al. 1976) (Fig. 7d). is natural
product was isolated from Streptomyces clavuligerus on
the basis of its potent inhibitory activity against the broad-
spectrum B-lactamase from K. pneumoniae. Clavulanic
acid was quite eective in preventing the destruction of
substrates of enzymes that could eectively hydrolyze
penicillins. Inhibition of cephalosporinases from a variety
of sources was considerably weaker. e mechanism for
inactivation was studied in detail for the PC 1 (Cartwright
and Coulson 1979), TEM-2 (Charnas et al. 1978; Fisher
etal. 1978),Bacillus cereus I (Durkin and Viswanatha 1978),
K1, and Proteus mirabilis (Reading and Farmer 1981)
B-lactamases. Although interaction of clavulanic acid with
these B-lactamase showed enzymes inactivation, but the
individual mechanism varied somewhat among dierent
systems. Results from Knowles and coworkers provided
the rst evidence that clavulanic acid behaved like a sui-
cide inactivator (Charnas et al. 1978; Fisher et al. 1978).
After interacting with TEM-2 B-lactamases, clavulanic
acid forms acyl enzyme. Once acyl enzyme was formed,
there were several potential fates for this acyl enzyme, (1)
the acyl enzyme could yield a transiently inhibited form,
which is not permanently inactivated. is type of inhibi-
tion would be similar to reversible inhibition in that inhi-
bition can be reversed with addition of substrate and free
enzyme can eventually be recovered, (2) the acyl enzyme
can simply act as enzyme-substrate complex, with even-
tual hydrolyses of clavulanic acid, again resulting in the
release of free enzyme, (3) there occurs permanent loss
of enzymatic activity, a desirable property for a bacterial
enzyme inhibitor. Hence the extent to which inactivation
occurs depends upon the interrelationship among vari-
ous pathways.
Tazobactam
Tazobactam is a compound with chemical formula (2S,
3S,5R)-3-methyl-4,4,7-trioxo-3-(triazol-1-ylmethyl)-4-
$1^{6}-1-azabicyclo[3.2.0]heptane-2-carboxylic acid
(Fig. 7e) which inhibit action of bacterial B-lactamases.
It exhibits an almost 10-fold greater inhibitory activity
than clavulanic acid against CTX-M type B-lactamases
(Bush et al. 1993). Movement of Ser130 in the inhibi-
tor resistant TEM-30 (Arg244Ser), TEM-32 (Met69Ile-
Met182r), and TEM-34 (Met69Val) B-lactamases
emphasizes the strategic role of Ser130 (Wang et al.
2002). Kuzin et al. (1999) illustrated the importance
of Ser130 in their structural analysis of the SHV-1 and
tazobactam inactivated SHV-1 (Kuzin et al. 1999; Kuzin
et al. 2001). e crystal structure of the SHV-1- tazo-
bactam complex showed the formation of an acyclic
form of tazobactam attached to Ser70 (assigned to the
imine on the basis of dihedral angles) and a 5-atom
vinyl carboxylic acid fragment attached to Ser130-OH
in the inactive species. As in the case with clavulanic
acid inactivation of TEM-2, the amino acids Ser70
and Ser130 were again the tazobactam-modied
residues. Evidence for an aldehyde and hydrated
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
b-lactams in clinical practice 99
aldehyde attached to the Ser130 residue was also
detected. Hence the critical involvement of Ser130 in
the inactivation process, as rst emphasized by Imtiaz
et al. (1994), is central to the studies of the inhibition
of class A B-lactamases by clavulanic acid and tazo-
bactam. Tazobactam undergoes fragmentation while
still attached to the active site Ser70 in Ser130Gly
B-lactamase. After acylation of Ser130Gly B-lactamase
by tazobactam, inactivation proceeds independent of
any additional covalent interaction.
Sulbactam
Sulbactam was developed as potential B-lactamase
inhibitor with chemical formula (2R,5R)-3,3-dimethyl-
4,4,7-trioxo-4L6-thia-1-azabicyclo[3.2.0]heptane-2-
carboxylic acid (Fig.7e). It exhibits some of the same
properties as clavulanic acid. Cephalosporinases were
inhibited less eectively than were penicillinases or
broad-spectrum B-lactamases. However, the dierential
between the enzyme classes was not as great as observed
with clavulanic acid in that sulbactam was slightly more
active against cephalosporinases than clavulanic acid. In
addition to having similar inhibition prole, they exhib-
ited a time dependent mode of inhibition. Mechanism
of action of sulbactam was examined, it was apparent
that the same kind of inhibitory pathway was operative
for both clavulanic acid and sulbactam. In extensive
studies of sulbactam is the E. coli TEM-2 B-lactamase,
the sulfone was observed to form a transiently inhibited
complex with the enzyme; hydrolysis of sulbactam was
also observed before irreversible inactivation resulted.
Sulbactam is able to inhibit the most common forms of
B-lactamase but is not able to interact with AmpC cepha-
losporinase. us it confers little protection against bac-
teria such as P. aeruginosa,Citrobacter, Enterobacter, and
Serratia, which often express this gene. It does possess
some antibacterial activity when administered alone,
but it is too weak to have any clinical importance.
Oxapenem
Oxapenem a ve membered, oxygen-containing ring
fused to B-lactam ring with a double bond between C2
and C3, were found to be potent B-lactamase inhibitor
but have poor stability (Fig. 7f). Oxapenems were potent
B-lactamase inhibitors however their activity varied
within the group for eg. AM-113 and its stereoisomer
AM-114 proving to be most active compounds (Lambert
et al. 2003). MIC50 of these agents were up to 105 than
that of clavulanate against class C and class D enzymes.
Oxapenems reduced MICs for ceftazidime against
class C hyperproducing, TEM, and SHV derived ESBL
producing strains however these compounds failed to
reduce MICs of ceftazidime against derepressed Amp C
producing P. aeruginosa strains. AM-112 had anity for
PBPs of E. coli DCO, with PBP2 being inhibited by lowest
concentration of AM-112 tested (0. 1mg/L). It was found
to be eective at protecting ceftazidime and reduces
ceftazidime MICs by 16- and 2048-fold (Jamieson et al.
2003). Protection of ceftazidime with AM-112 was
maintained against E. cloaceae P99 and K. pneumoniae
SHV-5. e activity of the combination of ceftazidime
and AM-112 against ESBL producing E. coli strains was
similar to that of piperacillin and tazobactam. AM-114
and AM-115 were found to be most potent inhibitor of
class A enzymes, AM-113 and AM-114 of class C enzymes
while AM-112 and AM-113 were most active oxapenems
against class D enzymes. e orientation of hydroxyl
ethyl group at C-6 position and nature of C-2 side chain
appears to be responsible for extent of inhibitory activity
against dierent B-Lactamases.
NXL 104 (AVE 1330 A)
NXL 104, previously designated as AVE 1330 A, is a
bridged diazabicyclo [3.2.1]octanone (Fig. 7g) and is
B-lactamase inhibitor able to inhibit both class A and
CB-lactamase. Its combination with ceftazidime exhib-
ited broad-spectrum activity against Ambler class A-
and class C- producing Enterobacteriaceae members.
Two to ve AVE 1330 A molecules only were needed
to inactivate one molecule of TEM-1 or P99 enzyme
and is far less than reported for clavulanate (Bonnefoy
et al. 2004). Beta-lactam-based B-lactamase inhibitors
generally act as a substrate of enzymes, giving an unsta-
ble acyl enzyme intermediate, which is responsible for
transient inhibition. is acyl enzyme then undergoes
a rearrangement, leading to a more stable inhibition.
MICs of ceftazidime/AVE 1330 A for Enterobacteriaceae
were at least eight fold lower than those of ceftazidime
alone (Bonnefoy et al. 2004). All of E. coli, K. pneumo-
neae,Citrobacter, and Proteus mirabilis strains, includ-
ing ceftazidime-resistant isolates, were inhibited at
4- 8 mg/L. Only 2 mg/L were required to inhibit other
Proteus, Enterobacter, Salmonella, and Serratia.
Miossec et al. (2007) evaluated the in vitro ecacy of
combination of ceftazidime and NXL 104 against carbap-
enem resistant Enterobacteriaceae strains, either medi-
ated by class A or D carbapenemases, or by class A or C
enzymes along with membrane impermeability. NXL 104
was found eective in restoration of activity of ceftazi-
dime, cefotaxime or imipenem against class A carbapen-
emase-producers (IMI-1, NMC-A, GES-2, GES-3, GES-4,
KPC-2, and KPC-3) however, activity of ceftazidime/NXL
104 against KPC-2 or KPC-3 enzymes was noticeable with
MIC ≤ 0.015 to 0.5 mg/L as compared to 64 to >128mg/L
for ceftazidime alone. OXA-48 (class D carbapenemase)
was also inhibited eectively by NXL 104 (IC50 value was
0.2 µM) and ceftazidime activity was found to be fully
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
100 Shahid et al.
restored against these strains. OXA-23, OXA-40, and
OXA-58, which mostly occur in Acinetobacter spp., were
poorly inhibited by NXL 104. Mushtaq et al. (2007) dem-
onstrated the activity of NXL 104/cephalosporins against
CTX-M ESBL-producers as CTX-M B-lactamase are
increasingly becoming the most prevalent ESBLs world-
wide (though the predominant types vary with dierent
geographic areas, CTX-M-15 are frequently encountered
in Europe, CTX-M-2 in South America, and CTX-M-9/
CTX-M-14 in East Asia). MICs of cefotaxime/NXL 104
were 1mg/L for all members of Enterobacteriaceae with
128-8000-fold potentiation, irrespective of CTX-M type,
co-production of OXA-1 and/or ertapenem resistance.
MICs of ceftazidime/NXL 104 were found to be ≤ 1mg/L
for all ertapenem-susceptible CTX-M producers and
were 4 mg/L for ertapenem-resistant isolates. Hence,
ceftazidime/NXL 104 combinations could provide useful
alternative for treatment of Enterobacteriaceae strains
that are carbapenem-resistant. It can also be noted that
NXL 104/cephalosporin combinations successfully over-
come oxyimino-cephalosporin resistance mediated by
CTX-M B-lactamases even when they were co-expressed
with OXA-1 enzymes or impermeability. Within class
A, KPCs represent a new family having a potential for
wide dissemination and its production confers resist-
ance to all B-lactams including carbapenems. Stachyra
et al. (2007) evaluated activity of NXL 104 against KPC
B-lactamases and NXL 104 showed an extremely potent
inhibitory activity against KPC-2 B-lactamase, with
IC50 of 38nM. Clavulanic acid and tazobactam, the two
potent comparators, were found to less active with IC50
in µM range. Resistant isolates (KPC producers) showed
MICs > 128mg/L for ceftazidime and ceftriaxone and 16
to > 128mg/L for imipenem, while after adding NXL 104
MICs were <1mg/L each for ceftriaxone/NXL 104 and
imipenem/NXL 104, while MICs ranged between 0.25
to 8mg/L for ceftazidime/NXL 104. NXL 104 was found
to have remarkable activity against KPC-2 enzyme and
eciently restored antimicrobial activity of ceftazidime,
ceftriaxone and imipenem against KPC-2 and KPC-3
B-lactamase-producing members of Enterobacteriaceae.
Mono- and bicyclic-bridged monobactams
Monobactams, such as aztreonam and bridged mono-
bactams, are B-lactamase inhibitors and showed potent
inhibitory activities against AmpC B-lactamase. Gaucher
et al. (2007) synthesize three series of novel bridged
monobactams using parallel chemistry approaches
from benzyl (1S, 5R)-7-oxo-2,6-diazabicyclo[3.2.0]
heptane-2-carboxylate. e siderophore monobactam
BAL0019764 (which is stable to hydrolysis by class B met-
allo B-lactamases and many class D B-lactamases and
has enhanced uptake by many species) reacted with AZT
with AmpC and ESBL enzymes and resisted attack by
metallo-B-lactamases. BAL 0030072 had a stronger inter-
action with ESBLs but found to be a weaker inhibitor of
AmpC enzymes. e bridged monobactam B-lactamases
were potent inhibitors of class C B-lactamases but had no
interaction with ESBLs or metallo- B-lactamases. It has
been noted that by combining bridged monobactams
(BMBs) with either BAL 0019764 or BAL 0030072, elevated
MICs of compounds with strains of Enterobacteriaceae
and P. aeruginosa that hyperproduce AmpCs, can be
depressed. BMBs were able to inhibit the enzymes and
thus aord protection to antibiotics. ey can be used in
combination with monobactam to improve the activity
of monobactam against Enterobacteriaceae and P. aeru-
ginosa. Recent in vitro activities showed the ecacy of
these inhibitors. IC50 of BAL 30072 is 0.09 µM and that of
clavulanate is >100 µM against AmpC enzyme from C.
freundii, while IC50 of aztreonam was found to be 0.01 µM
for same AmpC enzyme (Gaucher et al. 2007).
BAL 30376
BAL 30376 is a novel B-lactamase inhibitor and is a com-
bination of BAL 0019764 (a siderophore monobactam
which is stable to hydrolysis by class B B-lactamases),
BAL 0029880 (a bridged monobactam which is class C
inhibitor), and clavulanic acid. All the three components
are used in the ratio of 1:1:1 by weight. In vitro activity of
BAL 30376 against Gram-negative bacilli was evaluated
by Bowker et al. (2007) and they observed that MIC90
of BAL 30376 for E. coli, K. pneumoniae, and E. cloacae
were 4, 1, and 4mg/L, while MIC50 were found to be 0.5,
0.5, and 1 mg/L for respective organisms. e strains
which had CMY-2, CTX-M, multiple combinations of
SHV, CTX-M, CMY, OXA, and TEM-type enzymes, BAL
30376 had an MIC50 of 1-4mg/L while meropenem, a
most potent comparator drug, had MIC50 of 0. 03 mg/L.
MICs of BAL 30376 for >90% of such strains was found
to be ≤ 8 mg/L and for meropenem all MICs were also
< 8 mg/L. Against IMP containing Gram-negative
bacilli, MIC50 of BAL 30376 was 0.5- 2mg/L and that of
meropenem was 2- ≥64 mg/L. MIC50 of this novel inhib-
itor was 1- 4 mg/L for VIM, GIM and SPM producing
strains. It was observed that BAL 30376 was most potent
against IMP-4, IMP-13 or IMP-16, VIM-1, VIM-2, VIM-4,
GIM-1, and SPM-1 producers. However, meropenem/
imipenem were found to be more potent as compared
N
O
ONa
O
OMe
H
Figure 8. LK-157.
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
b-lactams in clinical practice 101
to BAL 30376 against recent ESBLs as well as CMY-2 and
CTX-M producers. BAL 30376 had superior activity than
aztreonam, ceftazidime, cefepime and piperacillin/
tazobactam. Page et al. (2007) demonstrated the in vitro
activity of BAL 30376 against various Gram-negative
bacteria and the MICs were observed in a range of
≤0.06 4 mg/L, including most carbapenem-resistant
strains. Higher MICs were observed for a few strains of
Acinetobacter spp., C. freundii, Enterobacter spp., and P.
aeruginosa. To test ecacy of this novel inhibitor, Hujer
et al. (2007) demonstrated its activity against ESBL
producing K. pneumoniae and MDR Acinetobacter
baumanii. ey have used BAL 30376-f (concentration
of BAL 19764 was varied while that of BAL 29880 and
clavulanic acid was xed at 4 mg/L and 2mg/L respec-
tively) and BAL 30376-v (concentration of BAL 19764,
BAL 29880 and clavulanic acid were varied in a xed
proportion of 5/3/1 respectively). ey observed that
based on MICs 8mg/L, BAL 30376-f and BAL 30376-v
demonstrated signicantly better activity (as compared
to imipenem) against Acinetobacter baumanii strains
possessing AmpC, blaADC and the carbapenemase
blaOXA-23 like genes. Similarly BAL 30376-f and BAL
30376-v were found to be more active against MDR K.
pneumoniae (especially CTX-M producing strains) as
compared to cefepime but slightly less active than imi-
penem. However, it has been observed that blaKPC bear-
ing strains of K. pneumoniae remain resistant to BAL
30376 but it oers promising activity against a wider
range of MDR strains bearing serine B-lactamases. It
also showed good activity against Enterobacteriaceae
and non-fermentive Gram-negative bacteria express-
ing AmpC B-lactamases, however it was found to be
compromised by certain carbapenemase when co-
expressed with other B-lactamases.
LK-157
LK-157 (Fig. 8) is a novel tricyclic carbapenem-inhibitor
of serine B-lactamases and is for parenteral use as an
IV agent. Recently, Kresken et al. (2007) investigated in
vitro activity of LK-157 to assess its potency. ey showed
that MIC90 of the novel inhibitor ranged between 16 and
> 128 mg/L for MRSA, penicillin-resistant S. pneumo-
niae, B. fragilis, P. aeruginosa, and Enterobacteriaceae
however, it was found to be ≤ 2mg/L for MSSA, S. pyo-
genes, penicillin-susceptible S. pneumoniae, and H.
inuenzae. When LK-157 was combined with cefotaxime
or cefepime, its spectrum of activity was found compara-
ble to that of tazobactam. LK-157 decreased the MICs of
aztreonam, ceftazidime, and cefuroxime for B. fragilis (8-
to ≥128-fold) and a wide range of B-lactamase-producing
Enterobacteriaceae members (up to ≥64-fold). It also
results in decreased MICs of cefuroxime for P. vulgaris
(≥32-fold) and those of aztreonam for MSSA (≥16-fold),
S. pyogenes (≥32-fold) and PSSP (≥64-fold) and those of
ceftazidime for MSSA (≥8-fold). It was noted that LK-157
did not aect (to a greater extent) the MICs of aztre-
onam, ceftazidime or cefuroxime against CTX-M pro-
ducing members of Enterobacteriaceae. LK-157 showed
potent in vitro activity against Gram-positive cocci and
also showed synergistic activity with B-lactams against
a wide range of Enterobacteriaceae that produce dier-
ent B-lactamases, and hence it can be a potent inhibitor
active against various pathogens including B-lactamase
producing several members of Enterobacteriaceae.
Prezelj et al. (2007) evaluated the activity of LK-157
against class A and C B-lactamase producing strains.
ey observed that IC50s of LK-157 (in nM range) were
superior to comparator drug, tazobactam, however its
activity in combination with cephalosporins against
TEM-, SHV- and BRO-type enzymes were found to be
comparable to cephalosporin/tazobactam combina-
tion. LK-157 was superior to tazobactam against AmpC-
producers and synergy was observed also against PC-type
B-lactamases. Prezelj et al. (2007) also demonstrated the
stability of 10-ethylidene trinems. As LK-157 is a very
low permeability drug, several drugs were designed
for per oral application. ey observed that among the
series of LK-157 analogues, LK-157E1 was most soluble
compound with low intestinal permeability. Pro-drug
ester of LK-157E1 exhibited improved solubility but per-
meability across rat jejunum was low. Hence LK-157 can
be a promising broad-spectrum tricyclic carbapenem
inhibitor of class A and C B-lactamase.
BLI-489
e novel penem B-lactamase inhibitor, BLI-489, has
shown activity against molecular class A or D enzymes,
including ESBLs as well as class C B-lactamases. Petersen
et al. (2007) demonstrated the in vitro activity of pipera-
cillin in combination with BLI-489 against B-lactamase-
producers. ey tested activity of combination of pip-
eracillin/BLI-489 in the ratio of 1:1, 2:1, 4:1, and 8:1 and a
constant concentration of either 2 or 4mg/L of BLI-489.
Piperacillin/tazobactam was used for comparison (tazo-
bactam was taken at a constant concentration of 4 mg/L).
Numbers of strains were found to be falsely reported
as susceptible or intermediate to piperacillin/BLI-489
N
N
O
S
CO2Na
O
Ph
O
O2
Figure 9. SA-1-204.
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
102 Shahid et al.
when tested at 1:1 and 2:1 ratio. e constant 2mg/L of
BLI-489 and 8:1 ratio had over-predict resistance. Some
strains that were class C or ESBL producers, classied
as non-susceptible (MICs ranging 32 to >256 mg/L) to
piperacillin/tazobactam, were found to be susceptible to
piperacillin/BLI-489 at a xed concentration of 4mg/L of
BLI-489 (MICs 1 to 64mg/L). Piperacillin/BLI-489 com-
bination showed potent in vitro activity against diverse
B-lactamase producers.
CP3242
It is a metallo- B-lactamase (MBL) inhibitor and com-
petitively inhibits both IMP-1 and VIM-2. Morinaka et al.
(2007) showed inhibitory mechanism of CP3242 along
with its in vitro activity. It specically lowered the MICs of
B-lactams against MBL-producing E. coli transformants.
It was noticed that combination ecacy of CP3242 with
carbapenems was not aected by existence of resistance
factors in P. aeruginosa (such as MexAB-OprM, and outer
membrane protein OprD-deciency). It signicantly low-
ered the MICs of biapenem in a concentration depend-
ent manner (2 to 32 mg/L) against MBL-producing P.
aeruginosa. MIC90 of biapenem was lowered from 512
to 16mg/L in the presence of 32mg/L of CP3242. MIC
lowering by CP3242 was also shown for IMP or VIM
producing E. coli, P. putida, S. marcescens, A. baumanii,
and K. pneumoniae. Hence, this novel competitive MBL
inhibitor can be eective against MBL-producers (Gram-
negative bacteria including P. aeruginosa) in combina-
tion with B-lactam antibiotics. It was also shown by Muto
et al. (2007) that the reducing eects of carbapenem on
a viable cell count of MBL-producing P. aeruginosa were
reduced in the presence of CP3242. us CP3242 co-
administered with carbapenem could be promising in
treatment of MBL-producing P. aeruginosa.
SA-1-204
It is a 6-alkylidiene penam sulphone B-lactamase
inhibitor (Fig. 9) that was found eective against SHV-
producing E. coli strains. Buynak et al. (2007) showed
ecacy of this B-lactamase inhibitor and found that
piperacillin/SA-1-204 was more potent than piperacil-
lin/tazobactam against strains bearing blaSHV-1 (MIC
1024/4 vs 64/ 4 mg/L) and blaSHV-5 (MIC 128/4 vs 16/4
mg/L). Both the inhibitors were found to be equally
eective against E. coli bearing SHV-2 (MIC 2-8/4mg/L)
and SHV-30 (MIC 1-2/4mg/L) ESBLs. Also piperacillin/
SA-1-204 was more eective against inhibitor-resistant
SHV-49 variant (128/4 vs 32/ 4mg/L). Alkylidene at the
6 position and 2’ B-substituent increases inhibitor e-
cacy. Hence, SA-1-204 can be considered as an eective
B-lactamase inhibitor against SHV-1, other ESBL vari-
ants of SHV, and SHV-49.
Synergistic activity of inhibitors
Although the B-lactamase-inhibitors act as eective
inactivators of isolated enzymes, a most important ques-
tion is whether these molecules can act to protect sus-
ceptible B-lactam antibiotic from hydrolysis in growing
cells. It is imperative that these inhibitors penetrate the
periplasm of Gram-negative organisms rapidly so as to
intercept the B-lactamase before all labile antibiotics
has been destroyed. Enzyme inhibition occur faster than
synthesis of new protein; otherwise, succeeding genera-
tions of cells will simply continue to elaborate additional
B-lactamase capable of hydrolyzing antibiotic. To evalu-
ate synergy between a susceptible B-lactam antibiotic
and a B-lactamase inhibitor MICs of antibiotic were
determined in the presence and absence of this xed
inhibitor concentration, and synergy was evaluated. In
other studies equal concentrations of inhibitor and anti-
biotic were varied stepwise by twofold dilution, and MICs
of combination were compared with those observed in
the absence of inhibitor. Synergy is dened as a fourfold
reduction in MIC for both components. As might be
predicted from the isolated enzyme studies, synergy in
B-lactamase-producing E. coli strains was much greater
with clavulanic acid than with sulbactam. is would be
expected, if one assumes that majority of these strains
produce plasmid mediated, TEM-type, broad-spectrum
B-lactamases. When a plasmid is present in a high copy
number, the elevated level of B-lactamase hydrolyze
sulbactam more quickly than the enzyme will become
inactivated because of reasonably high turnover number.
is was demonstrated in studies by Easton and Knowles,
who observed good synergy with both sulbactam and
clavulanic acid in a Proteus species that produced a
moderate level of TEM B- lactamase. Another factor that
will contribute to a dierential in activity is the ease with
which the inhibitor can penetrate the cell. As indicated
by Li et al. (1981), clavulanic acid may have less of a
penetration problem than sulbactam in P. aeruginosa,
Citrobacter, and Enterobacter strains.
Among the B-lactams, third generation cepha-
losporins, such as ceftazidime, cefotaxime, and ceftriax-
one are routinely used in our clinical setting, and resist-
ance to these drugs, due to B-lactamase production, is
rampant (Mahapatra et al. 2003). e combination of
cefoperazone/sulbactam shows marked degree of syn-
ergy against organisms that are resistant to cefoperazone
alone (Wexler and Finegold 1988). Broad-spectrum
B-lactams, such as imipenem, cefdinir, cefepime, and
cefpirome, and B-lactamase-inhibitor combination, such
as piperacillin/tazobactam, cefoperazone/sulbactam
and ticarcillin/clavulanate, have been introduced to
overcome the resistance. Piperacillin/tazobactam
exhibited greater in vitro activity only against E. coli and
P. vulgaris as compared to piperacillin/sulbactam. Both
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
b-lactams in clinical practice 103
combinations were found equally eective against other
Enterobacteriaceae and P. aeruginosa isolates. Sulbactam
may have higher intrinsic activity against Acinetobacter
isolates and thus may be considered as drug of choice
where Acinetobacter is a suspected pathogen.
e combination of piperacillin and tazobactam
is active against many piperacillin-resistant strains of
Staphylococci, Enterobacteriaceae and Bacteroides
species (Eliopoulos et al. 1989). However the two
piperacillin-B-lactam inhibitor combinations (pipera-
cillin/sulbactam and piperacillin/tazobactam) did not
display signicant dierences in antimicrobial suscepti-
bility of Gram-positive isolates (methicillin-susceptible
S. aureus and E. faecium) (Kuck et al. 1989).In vitro stud-
ies have demonstrated that antimicrobial activity does
not predominantly depend on equal proportions, but on
critical concentration of the inhibitor (Lister et al. 1997).
e combination of cefoperazone/sulbactam shows a
marked degree of synergy against organisms that are
resistant to cefoperazone alone, including Acinetobacter
species and Enterobacter species (Dias et al. 1986).
Piperacillin/tazobactam and cefoperazone/sulbactam
were usually much more active than ticarcillin/clavu-
lanate against strains producing elevated levels of non-
class I B-lactamase. However increased resistance to all
three combinations was associated with increased pro-
duction of certain B-lactamases. Ticarcillin/clavulanate
would be expected to be the most eective combina-
tion against TEM-1 producing strains. Yoshimura and
Nikiado (1985) reported that cefoperazone penetrate
the outer membrane of E. coli faster than piperacillin
and carbenicillin. If ticarcillin has penetration rate simi-
lar to that of carbenicillin, then greater potency of cef-
operazone/sulbactam could be consequence of faster
penetration by cefoperazone. is, however, does not
explain the greater potency of piperacillin/tazobactam
over ticarcillin/clavulanate.
e dierence between these combinations may be
due to greater anity of piperacillin for PBP3, the prin-
cipal target of both drugs. is is suggested by a 20-fold-
greater anity of ureidopenicillins-piperacillins, mezlo-
cillin and azlocillin- for PBP3 of E. coli compared with
carboxypenicillins-carbenicillin (omson et al. 1990).
e enhanced activity of ceftriaxone observed in the pres-
ence of B-lactamase inhibitor-sulbactam seems promis-
ing for the treatment of serious infections due to members
of Enterobacteriaceae producing an ESBL such as SHV-2
(Fantin et al. 1990). Recent In vitro studies indicated that
among available B-lactam/B-lactamase inhibitor combi-
nations, piperacillin/tazobactam has best activity against
nosocomial Gram-negative pathogens followed by cef-
operazone/sulbactam. However, very recently, ceftriax-
one-sulbactam was found even better than piperacillin-
tazobactam and the activity was demonstrated at par with
carbapenem, the imipenem (Shahid 2007a).
Conclusions
Resistance to third and fourth generation cephalosporins
has become a major concern worldwide. Even more
alarming is the emergence of carbapenem resistance;
the carbapenems are often considered to be a “drug
of choice” and increasingly used in empirical therapy.
Against this rising resistance the role of B-lactam-B-
lactamase inhibitor combinations needs to be consid-
ered. Sulbactam has been approved recently in many
countries to be combined with B-lactam antibiotics
including, recently, in India, and ceftriaxone-sulbactam
proved to be eective in vitro against diverse CTX-M-15-
producing E. coli strains. e third and fourth genera-
tion cephalosporins and combination of cephalosporins
(especially third generation) with new B-lactam inhibitor
can be used alternately to avoid selection pressure and
it can be an eective therapy to avoid drug resistance.
Since, B-lactams are often combined with aminogly-
cosides, in empirical therapy (Shahid 2007b), another
alternate approach to prevent selection pressure could
be periodical prescription rotation between B-lactam-
aminoglycoside combination therapy, B-lacatm-
B-lactamase-inhibitor, and the carbapenems.
Acknowledgements
e authors wish to thank Prof. Gopal Nath, Department
of Medical Microbiology, Banaras Hindu University,
Varanasi, and Prof. N. P. Singh, Department of
Microbiology, University College of Medical Sciences,
Delhi for critical suggestions during preparation of this
manuscript. M. Shahid is grateful to Department of
Science & Technology, Ministry of Science & Technology,
Government of India, for the award of Young Scientist
Project (SR/FT/L-111/2006), and to the University
Grants Commission, India, and the Association of
Commonwealth Universities and British Council, UK,
for awarding a Commonwealth Academic Fellowship
(INCF-108-05). e authors are also grateful to T.
Tripathi and M. Arif for graphical assistance especially
the preparation of chemical structures.
Declaration of interest: e authors report no conicts
of interest. e authors alone are responsible for the
content and writing of the paper.
References
Abeles, R H, & Maycock, A L 1976, Suicide enzyme inactivators Acct
Chem Res 9, pp 313–319
Abraham, E P 1962, e cephalosporins Pharmacol Rev 14,
pp 473–500
Abraham, E P 1987, Cephalosporins 1945–1986 Drugs 34 Suppl,:2,
pp 1–14
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
104 Shahid et al.
Arakawa, S, Kamidono, S, & Inamatsu T 1997, Clinical studies of
S–4661, new parenteral carbapenem antibiotic, in compli-
cated urinary tract infections 37th Interscience Conference on
Antimicrobial Agents and Chemotherapy American Society for
Microbiology, Washington, DC
Baraniak, A, Fiett, J, Hryniewicz, W, Nordman, P, & Gniadkowski, M
2002, Ceftazidime–hydrolyzing CTX–M–15 extended–spectrum–
beta–lactamase ESBL, in Poland J Antimicrob Chemother 50, pp
393–396
Batchelor, F R, Doyle, F P, Nayler, J H, & Rolinson, G N 1959, Synthesis
of penicillins: 6–aminopenicillanic acid in penicillin fermenta-
tions Nature 183, pp 257–258
Bayles, K W 2000, e bactericidal action of penicillin: new clues to
an unsolved mystery Trends Microbiol pp 81274–81278
Bogdanovich, T L, Ednie, M, Shapiro, S, & Appelbaum, P C 2005, Anti–
staphylococcal activity of ceftobiprole, a new broad–spectrum
cephalosporin Antimicrob Agents Chemother 49, pp 4210–4219
Bonnefoy, A, Hamelin, C D, Steier, V, Delachaume, C, Seys, C, Stachyra,
T, Fairley, M, Guitton, M, & Lampilas, M 2004, In vitro activ-
ity of AVE–1330A, an innovative broad– Spectrum non B–lactam
B–lactamases inhibitor J Antimicrob Chemother 54, pp 410–417
Bouchillon, S K, Hackel, M A, Johnson, J L, Poupard, J A, & Pace, J L
2007, e carbapenem PZ–601 SMP–601, has potent in vitro
Gram–positive, Gram–negative and anaerobic bacterial activ-
ity 47th Interscience Conference on Antimicrobial Agents and
Chemotherapy American Society for Microbiology, Washington,
DC
Bowker, K E, Noel, A R, & Macgowan, A P 2007, In vitro potency of
BAL30376 against Gram–Negative Bacilli GNB, 47th Interscience
Conference on Antimicrobial Agents and Chemotherapy
American Society for Microbiology, Washington, DC
Bradford, P A , Bratu, S, Urban, C, Visalli, M, Mariano, N, Landman, D,
Rahal, J J, Brooks, S, Cebular, S, & Quale, J 2004, Emergence of
carbapenem–resistant Klebsiella species possessing the class A
carbapenem–hydrolyzing KPC–2 and inhibitor–resistant TEM–30
beta–lactamases in New York city Clin Infect Dis 34, pp 55–60
Bratu, S, Landman, D, Haag, R, Recco, R, Eramo, A, Alam, M, &
Quale, J 2005, Rapid spread of carbapenem–resistant Klebsiella
pneumoniae in New York city: a new threat to our antibiotic
armamentarium Arch Int Med 165, pp 1430–1435
Brown, A G, Butterworth, D, Cole, M, Hanscomb, G, Hood, J D, Reading,
C, & Rolinson, G N 1976, Naturally occurring B–lactamase inhib-
itors with antibacterial activity J Antibiot 29, pp 668–669
Bush, K 2001, New B–lactamases in gram–negative bacteria: diversity
and impact on the selection of antimicrobial therapy Clin Infect
Dis 32, pp 1085–1089
Bush, K, Jacoby, G A, & Medeiros, A A 1995, A functional classica-
tion scheme for B–lactamases and its correlation with molecular
structure Antimicrob Agents Chemother 39, pp 1211–1233
Bush, K, Macalintal, C, Rasmussen, B A, Lee, V J, & Yang, Y 1993, Kinetic
interactions of tazobactam with beta–lactamases from all major
structural classes Antimicrob Agents Chemother 37, pp 851–858
Butaye, P, Cloeckaert, A, & Schwartz, S 2003, Mobile genes coding
for eux–mediated antimicrobial resistance in Gram–positive
and Gram–negative bacteria Int J Antimicrob Agents 22,
pp 205–210
Buynak, J D, Kalp, M, Bethel, C R, Carey, P R, & Bonomo, R A 2007,
SA–1–204, a novel 6–alkylidiene penam sulfone B–lactamase
inhibitor eective against Escherichia coli expressing SHV
B–lactamases 47th Interscience Conference on Antimicrobial
Agents and Chemotherapy American Society for Microbiology,
Washington, DC
Cartwright, S J, & Coulson, A F W 1979, A semisynthetic penicillinase
inactivator Nature London, 278, pp 577–609
Castanheira, M, Toleman, M A, Jones, R N, Schmidt, F J, & Walsh,
TR2004, Molecular characterization of a B–lactamase gene,
blaGIM–1, encoding a new subclass of metallo–B–lactamase
Antimicrob Agents Chemother 48, pp 4654–4661
Chain, E, Florey, H W, Gardner, A D, Heatly, N G, Jennings, M A, &
Orr–Ewing, J 1940, Penicillin as chemotherapeutic agent Lancetti
226–228
Chamberlandt, B J, Hoang, M, Dinh, C, Cotter, D, Bond, E, Gannon, C,
Park, C, Malouin, F, & Dudley, M N 2001, In vitro activities of
RWJ–54428 C–02, 479, against multi–resistant Gram–positive
bacteria Antimicrob Agents Chemother 45, pp 1422–1430
Charnas, R L, Fisher, J, & Knowles, R J 1978, Chemical studies on
the inactivation of E coli RTEM B–lactamase by clavulanic acid
Biochemistry 3, pp 341–382
Christensen, B G, Salton M R J, & Shockman, G D ed, 1981, Structure–
activity relationship in B–lactam antibiotics b–lactam Antibiotics:
Mode of action, New Developments, and future prospects pp 101–
102 Academic press, New York
Claridge, C A, Gourevitch, A, & Lein, J 1960, Bacterial penicillin ami-
dase Nature 187, pp 237–238
Critchley, I A, Karlowsky, J A , Draghi, D C, Jones, M E, ornsberry, C,
Murtt, K, & Sahm, D F 2002, Activities of Faropenem, an oral
B–lactam, against recent US isolate of S pneumoniae, H inuenzae
and M catarrhalis Antimicrob Agents Chemother 46, pp 550–55
Curran, M P, Simpson, D, Perry, C M 2003, Ertapenem, A review
of its use in the management of bacterial infections Drugs 63,
pp 1855–1878
Danel, F, Hall, L M, Duke, B, Gur, D, & Livermore, D M 1999, OXA–17,
a further extended–spectrum variant of OXA–10 beta–lactamase,
isolated from Pseudomonas aeruginosa Antimicrob Agents
Chemother 43, pp 1362–1366
Danel, F, Hall, L M, Gur, D, & Livermore, D M 1998, OXA–16, a fur-
ther extended–spectrum variant of OXA–10 beta–lactamase,
from two Pseudomonas aeruginosa isolates Antimicrob Agents
Chemother 42, pp 3117–3122
Davies, B, & Morris, T 1993, Physiological parameters in laboratory
animals and humans Pharm Res 10, pp 1093–1095
Davies, T A, Shang, W, & Bush, K 2006, Activities of ceftobiprole and
other B– lactams against Streptococcus pneumoniae clinical iso-
lates from the united status with dened substitutions in pen-
icillin–binding proteins PBP1a, PBP2b and PBP2x Antimicrob
Agents Chemother 50, pp 2530–2532
Decousser, J W, Poirel, L, Nordman, P 2001, Characterization
of a chromosomally encoded extended–spectrum class A
beta–lactamase from Kluyvera cryocrescens Antimicrob Agents
Chemother 45, pp 3595–3598
Di Conza, J, Ayala, J A, Power, P, Mollerach, M, & Gutkind, G 2002,
Novel class 1 integron InS21, carrying blaCTX–M–2 in Salmonella
enterica serovar infantis Antimicrob Agents Chemother 46,
pp 2257–2261
Dias, M B S, Jacobus, N V, & Tally, F P 1986, In vitro activity of cef-
operazone–sulbactam against Bacteroides species J Antimicrob
Chemother 18, pp 467–471
Docquier, J D, Riccio, M L, Mugnaioli, C, Luzzaro, F, Endimiani, A,
Toniolo, A, Amicosante, G, & Rossolini, G M 2003, IMP–12, a new
plasmid encoded metallo–B–lactamase from a Pseudomonas
putida clinical isolate Antimicrob Agents Chemother 47,
pp 1522–1528
Donowitz, G R, & Mandell, G L 1988, Beta–Lactam antibiotics N Engl
J Med 318, pp 419–426
Du Bois, S K, Marriott, M S, & Amyes, S G B 1995, TEM–and SHV
derived extended spectrum B–lactamases: relationship between
selection, structure and function J Antimicrob Chemother 35,
pp 7–22
Dudley, M N, Malouin, F, Blais, J, Chamberland, S, Hoang, M, Park, C,
Chan, C, Mathias, K, Hakem, S, Dupree, K, Liu, E, & Nguyen T
2003, RWJ– 54428 MC–02,479,, a new cephalosporin with high
anity for penicillin–binding proteins, including PBP 2a, and
stability to staphylococcal Beta–lactamases Antimicrob Agents
Chemother 47, pp 658–664
Durkin, J P, & Viswanatha, T 1978, Clavulanic acid inhibition
of B–lactamase I from Bacillus cereus 596/H J Antibiot 31,
pp 1162–1169
El Kharroubi, A, Jacques, P, Piras, G, Van Beeumen, J, Coyette, J, &
Ghuysen, J M 1991, e Enterococcus hirae R–40 penicillin–
binding protein 5 and the methicillin–resistant Staphylococcus
aureus penicillin binding protein 2’ are similar J Biochem 280,
pp 463–469
Eliopoulos, G M, Klimm, K, Ferraro, M J, Jacoby, G A, & Moellering,
R C 1989, Comparative in vitro activity of piperacillin com-
bined with B–lactamase inhibitor tazobactam YTR 830, Diagn
Microbiol Infect Dis 12, pp 482–488
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
b-lactams in clinical practice 105
Fantin, B, Pangon, B, Potel, G, Caron, F, Vallee, E, Vallois, J M,
Mohler, J, Brure, A , Phillipon, A, & Carbon, C 1990, Activity of
sulbactam in combination with ceftriaxone in vitro and in vivo
experimental endocarditis caused by E coli producing SHV–2
like B–lactamase Antimicrob Agents Chemother 34, pp 581–586
Fisher, J, Charnas, R L, & Knowles, J R 1978, Kinetic studies on the
inactivation of E coli RTEM B–lactamase by clavulanic acid
Biochemistry 17, pp 2180–2184
Fleming, A 1929, On the antibacterial action of cultures of Penicillium
with special reference to their use in the isolation of B inuenzae
British J Exp pathol 10, pp 226–239
Flynn, E H 1972, Cephalosporins and penicillins: chemistry and Biology
Academic press, Inc, New York
Frere, J M 1995, Beta lactamases and bacterial resistance to antibiot-
ics J Mol Microbiol 16, pp 385–395
Fuchs, P C, Barry, A L, & Brown, S D 2001, In vitro activities of ertap-
enem MK–0826, against clinical bacterial isolates from 11 North
American Medical Centres Antimicrob Agents Chemother 45,
pp 1915–1918
Fujimoto, K, & Kanazawa, K 2007, In vitro activity of SMP–
601SMP;PZ–601, in combination with other antimicrobial
agents against Burkholderia cepacia Bc, and Serratia marc-
escens Sm, 47th Interscience Conference on Antimicrobial
Agents and Chemotherapy American Society for Microbiology,
Washington, DC
Fujimura, T, Yamano, Y, Yoshida, I, Shimada, J, & Kuwahara, S 2003,
In vitro activity of S– 3578, a new broad spectrum Cephalosporin
active against methicillin–resistant Staphylococci Antimicrob
Agents Chemother 47, pp 923–931
Fung–Tomc, J C, Clark, J, Minassian, B, Pucci, M, Tsai, Y H,
Gradelski, E, Lamb, L, Medina, I, Huczko, E, Kolek, B,
Chaniewski, S, Ferraro, C, Washo, T, & Bonner, D P 2002, In vitro
and in vivo activities of a novel Cephalosporin, BMS–247243,
against Methicillin–resistant and –susceptible Staphylococci
Antimicrob Agents Chemother 46, pp 971–976
Gaucher, B, Dantier, C, Desarbre, E, & Page, M G P 2007, Novel mono–
and bicyclic monobactams as potent inhibitors of class C
B–lactamases 47th Interscience Conference on Antimicrobial
Agents and Chemotherapy American Society for Microbiology,
Washington, DC
Geraci, J E, Homeister, R T, Nichols, D R, & Needham, G M 1962,
Preliminary clinical and laboratory experiences with a new
oral penicillin: 5–methyl–3–phenyl–4–isoxazolyl penicillin
Proceedings of sta Meetings 181, pp 739–744
Ghuysen, J M 1991, Serine beta–lactamases and penicillin–binding
proteins Annual Rev Microbiol 45, pp 37–67
Giakkoupi, P, Xanthaki, A, Kanelopoulou, M, Vlahaki, A, Miriagou, V,
Kontou, S, Papafraggas, E, Malamou–Lada, H, Tzouvelekis, L S,
Legakis, N J, & Vatopoulos, A C 2003, VIM–1 metallo–B
lactamase–producing Klebsiella pneumoniae strains in Greek
hospitals J Clin Microbiol 41, pp 3893–3896
Hanaki, H, Konodo, N, & Inaba, Y 1996, In vitro activity of S–4661,
a new 1 b–methyl carbapenem, against Gram–positive and
Gram–negative bacterial isolates 36th Interscience Conference
on Antimicrobial Agents and Chemotherapy American Society
for Microbiology, Washington, DC
Hanson, N D 2003, AmpC B–lactamases: what do we need to know for
the future J Antimicrob Chemother 52, pp 2–4
Hartman, B J, & Tomasz, A 1984, Low anity penicillin binding pro-
tein associated with beta–lactam resistance in Staphylococcus
aureus J Bacteriol 158, pp 513–516
Hashizume, T, Nagano, R, Shibata, K, Adachi, Y, Imamura, H, &
Morishima, H 2000, In vitro activities of Novel trans–3,5–disub-
stituted pyrrolidinylthio–IB–methyl Carbapenems with potent
activities against methicillin–resistant Staphylococcus aureus
and Pseudomonas aeruginosa Antimicrob Agents chemother 44,
pp 489–495
Hawkey, P M 2008, Molecular epidemiology of clinically signi-
cant antibiotic resistance genes British J Pharmacol 153, pp
406S–413S
Hebeisen, P, Heinze–Krauss, I, Angehrn, P, Hohl, P, Page, M G, &
en, R L 2001, In vitro and in vivo properties of RO 63–9141,
a novel broad– spectrum Cephalosporin with activity against
methicillin–resistant Staphylococci Antimicrob Agents
Chemother 45, pp 825–836
Hecker, S J, Glinka, T W, Cho, A, Zhang, Z J, Price, M E, Chamberland,S,
Grith, D, & Lee, V J 2000, Discovery of RWJ–54428 MC–02,479,
A new cephalosporin active against resistant Gram–positive
bacteria J Antibiot 53, pp 1272–1281
Heritage, J, M’ Zali F H, Gascoyne–Binzi, D, & Hawkey, P M 1999,
Evolution and spread of SHV extended–spectrum beta–
lactamases in Gram–negative bacteria J Antimicrob Chemother
44, pp 309–318
Hikida, M, Itahashi, K, Igarashi, A, Shiba, T, & Kitamura, M 1999, In vitro
antibacterial activity of LJC–11,036, an active metabolite of L–084,
a new oral Carbapenem antibiotic with potent anti–pneumococcal
activity Antimicrob Agents Chemother 43, pp2010–2016
Hirai, Y, Osaki, Y, Morinaka, A, Yamada, K, Yoshida, T, &
Yonezawa, M 2007, Potent antibacterial activity of ME1036
against E faecalis, E coli, and H inuenzae due to its high an-
ity to PBPs 47th Interscience Conference on Antimicrobial
Agents and Chemotherapy American Society for Microbiology,
Washington, DC
Hoellman, D B, Kelly, L M, & Credito, K 2003, In vitro anti–anaero-
bic activity of ertapenem MK–0826,, a long acting carbapenem,
tested against selected resistant strains J Antimicrob Chemother
46, pp 220–224
Hori, S, Sato, J, & Kawamura, M 1997, S– 4661, a new carbapenem,
has weak convulsant activity A comparative study on convulsant
activity of carbapenems and cephalosporins 37th Interscience
Conference on Antimicrobial Agents and Chemotherapy
American society for Microbiology, Washington, DC
Huang, H T, English, A R, Seto, T A, Shull, G M, & Sobin, B A 1960,
Enzymatic hydrolysis of the side chain of penicillins J American
chemical society 82, pp 3790–3791
Hujer, A M, Desabre, E, Endimiani, A, Paterson, D L, Page, M G P, &
Bonomo, R A 2007, Activity of BAL30376 against Multidrug
Resistant MDR, Klebsiella and Acinetobacter spp 47th Interscience
Conference on Antimicrobial Agents and Chemotherapy
American Society for Microbiology, Washington, DC
Humeniuk, C, Alert, G, Gautier, V, Grimont, P, Labia, R, & Phillipon, A
2002, Beta–lactamases of Kluyvera ascorbata, probable progeni-
tor of some plasmid encoded CTX–M types Antimicrob Agents
Chemother 46, pp 3045–3049
Iizawa, Y, Nagai, J, Ishikawa, T, Hashiguchi, S, Nakao, M, Miyake,
A, & Okonogi, K 2004, In vitro antimicrobial activity of
T–91825, a novel anti–MRSA cephalosporin, and in vivo anti–
MRSA activity of its prodrug, TAK–599 J Infect Chemother 10,
pp 146–156
Imtiaz, U, Billings, E M, Knox, J R, & Mobashery, S 1994, A structure–
based analysis of the inhibition of class A P–lactamases by sul-
bactam Biochemistry 33, pp 5728–5738
Inoue, M, & Mitsuhashi, S 1996, Antibacterial activity of new carbap-
enem S–4661 and stability to beta–lactamase 36th Interscience
Conference on Antimicrobial Agents and Chemotherapy
American Society for Microbiology, Washington, DC
Iyobe, S, Kusadokoro, H, Ozaki, J, Matsumura, N, Minami, S, Haruta, S,
Sawai, T, & Hara, K O 2000, Amino acid substitutions in a variant
of IMP–1 metallo–B–lactamase Antimicrob Agents Chemother 44,
pp 2023–2027
Jacoby, G A, & Medieros, A A 1991, More extended Spectrum
B–lactamases J Antimicrob Chemother 35, pp 1697–1704
Jacqueline, C, Caillon, J, Le Mabecque, V, Miegeville, A F, Hamel, A,
Bugnon, D, Ge, J Y, & Potel, G 2007, In vivo ecacy of ceftaroline
PPI–0903,, a new broad–spectrum cephalosporin, against methi-
cillin–resitant and vancomycin–intermediate Staphylococcus
aureus: comparison with linezolid and vancomycin in a rab-
bit endocarditis model Antimicrob Agents Chemother 51,
pp 3397–3400
Jamieson, C E, Lambert, P A, & Simpson, I N 2003, In vitro and in
vivo activities of AM–112, a novel oxapenem Antimicrob Agents
Chemother 47, pp 1652–1657
Jevons, M P 1961, ‘Celbenin’–resistant Staphylococci Br Med J 1,
pp 124–125
Jo, J T H, Brinkman, F S L, & Hancock, R E W 2003, Aminoglycoside
eux in Pseudomonas aeruginosa: involvement of novel
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
106 Shahid et al.
outer membrane proteins Antimicrob Agents Chemother 47,
pp 1101–1111
Jones, R N 2001, In vitro evaluation of ertapenem MK–0826,, a long
acting carbapenem, tested against selected resistant strains J
Antimicrob Chemother 13, pp 363–376
Jones, R N, Deshpande, L M, Mutnik, A H, & Biedenbach, D J 2002, In
vitro evaluation of BAL 9141, a novel parenteral cephalosporin
active against oxacillin–resistant Staphylococci J Antimicrob
Chemother 50, pp 915–932
Kanazawa, K, Takemoto, K, Fujimoto, K, & Eguchi, K 2007, In vitro
antimicrobial activity of SMP–601SMP; PZ–601,, a novel anti
MRSA carbapenem, against resent Gram–Negative GN, clinical
isolates in Japan 47th Interscience Conference on Antimicrobial
Agents and Chemotherapy American Society for Microbiology,
Washington, DC
Karchmer, A W Mandell, G L , Douglas, R, & Bennetts, J E eds, 2000,
Cephalosporins Principles and practice of Infectious diseases
pp 274–291 Churchill Livingstone, Inc, Philadelphia
Kaufmann, W, & Bauer, K 1960, Enymatische spaltung und resyn-
these von Penicillin Naturwissenchaften 47, pp 474–475
Kelly, J A , Moews, P C, Knox, J R, Frere, J M, Ghuysen, J M 1982,
Penicillin target enzyme and the antibiotic binding site Science
218, pp 479–481
Kobayashi, Y, Kizaki, M, & Mutou, A 1997, Synergy with S–4661 and
Vancomycin or teicoplanin against imipenem–resistant MRSA
identied by the PCR method 37th Interscience Conference on
Antimicrobial Agents and Chemotherapy American Society for
Microbiology, Washington, DC
Koga, T, Abe, T, Inoue, H, Takenouchi, T, Kitayama, A, Yoshida, T,
Masuda, N, Sugihara, C, Kakuto, M, Nakagawa, M, Shibaya, T,
Matsushita, Y, Hirota, T, Ohya, S, Utsui, Y, Fukuoka, T, &
Kuwahara, S 2005, In vitro and in vivo antibacterial activities of
CS–023 RO4908463,, a novel parenteral Carbapenem Antimicrob
Agents Chemother 49, pp 3239–3250
Koh, T H, Sng, L H, Babini, G S, Woodford, N, Livermore, D M,& Hall,L M
Carbapenem–resistant Klebsiella pneumoniae in Singapore pro-
ducing IMP–1 B–lactamase and lacking an outer membrane pro-
tein Antimicrob Agents Chemother 45, pp 1939–1940
Kohler, J, Dorso, K L, & Young, K 1999, In vitro activities of the
potent, broad–spectrum carbapenem MK–0826 L–749,345,
against broad–spectrum B–lactamase– and extended–spectrum
B–lactamase– producing Klebsiella pneumoniae and E coli clini-
cal isolates Antimicrob Agents Chemother 43, pp 1170–1176
Kresken, M, Brauers, J, & Prezelj, A 2007, In vitro activity of LK–157, a
novel tricyclic carbapenem B–lactamase inhibitor 47th Interscience
Conference on Antimicrobial Agents and Chemotherapy American
Society for Microbiology, Washington, DC
Kuck, N K, Jacobus, N V, Petersen, P J, Weiss, W J, & Testa, R T 1989,
Comparative in vitro and in vivo activities of piperacillin combined
with the B–lactamase inhibitors tazobactam, clavulanic acid, and
sulbactam Antimicrob Agents Chemother 33, pp 1964–1969
Kurazono, M, Ida, T, Yamada, K, Hirai, Y, Maruyama, T, Shitara, E,
& Yonezawa, M 2004, In vitro activities of ME1036 CP5609,, a
novel parenteral Carbapenem, against Methicillin–resistant
Staphylococci Antimicrob Agents Chemother 48, pp 2831–2837
Kurazono, M, Ida, T, Yamada, K, Hirai, Y, Maruyama, T, Shitara, E, &
Yonezawa, M 2004, In vitro activities of ME1036 CP5609,, a novel
parenteral carbapenem, against methicillin–resistant staphylo-
cocci Antimicrob Agents Chemother 48, pp 2831–2837
Kuwamoto, I, Shimoji, Y, Kanno, O, Kojima, K, Ishkawa, K,
Matsuyama, E, Ashida, Y, Shibayama, T, Fukuoka, T, & Ohya,
S 2003, Synthesis and structure activity relationships of novel
parenteral Carbapenem, CS–023 R–115685, and related com-
pound containing an amidine moiety J Antibiot 56, pp 565–579
Kuzin, A P, Nukaga, M, Nukaga, Y, Hujer, A M, Bonomo, R A, & Knox, J R
1999, Structure of the SHV–1 beta–lactamase Biochemistry 38,
pp 5720–5727
Kuzin, A P, Nukaga, M, Nukaga, Y, Hujer, A, Bonomo, R A, & Knox, J R
2001, Inhibition of the SHV–1 beta–Lactamase by Sulfones:
Crystallographic Observation of Two Reaction Intermediates
with Tazobactam Biochemistry 40, pp 1861–1866
Lambert, P A, Jamieson, C E, & Simpsom, J N 2003, In vitro activities
of novel oxapenem, alone and in combination with ceftazidime,
against Gram–positive and Gram–negative organism Antimicrob
Agents Chemother 47, pp 2615–2618
Lauretti, L, Riccio, M L, Mazzariol, A, Cornaglia, G, Amicosante, G,
Fontana, R, & Rossolini, G M 1999, Cloning and characteriza-
tion of blaVIM, a new integron–borne metallo–B–lactamase
gene from a Pseudomonas aeruginosa clinical isolate Antimicrob
Agents Chemother 43, pp 1584–1590
Lee, K, Yum, J H, Yong, D, Lee, H M, Kim, H D, Docquier, J D,
Rossolini, G M, & Chong, Y 2005, Novel acquired metallo–
beta–lactamase gene, blaSIM–1,, in a class 1 integron from
Acinetobacter baumannii clinical isolates from Korea Antimicrob
Agents Chemother 49, pp 4485–4491
Li, J T, Moosdeen, F, & Marriner, J M 1981, Augmentin Amoxycillin
and clavulanic acid, therapy in complicated infections due to
B–lactamase producing bacteria J Antimicrob Chemother 7,
pp 229–236
Lister, P D, Prevan, A M, & Sanders, C C 1997, Importance of
B–lactamase inhibitor pharmacokinetics and the pharmacody-
anamics of inhibitor–drug combinations: studies with piperacil-
lin–tazobactam and piperacillin–sulbactam Antimicrob Agents
Chemother 41, pp 721–727
Livermore, D M 1995, ß–lactamases in laboratory and clinical resist-
ance Clin Microbiol Rev 8, pp 557–584
Livermore, D M 1998, B–lactamase–mediated resistance and opportun-
ities for its control J Antimicrob Chemother 41, suppl D, pp 25–41
Livermore, D M, & Woodford, N 2006, e B–lactamase threat in
Enterobacteriaceae, Pseudomonas, and Acinetobacter Trends
Microbiol 14, pp 413–420
Livermore, D M, Sefton A M, & Scott, G M 2003, Properties and poten-
tial of ertapenem J Antimicrob Chemother 52, pp 331–44
Livermore, D M, Carter, M W, & Bagel, S 2001, In vitro activities of ertap-
enem MK–0826, against recent clinical bacteria collected in Europe
and Australia Antimicrob Agents Chemother 45, pp1860–1867
Lovering, A, Danel, F, Page, M G P, & Strynadka, N I 2006, Mechanism
of action of ceftobiprole: structural basis for anti–MRSA activity
16th European Congress of Clinical Microbiology and Infectious
Diseases Nice, France
Mahapatra, A, Samal, B, & Pattnaik, D 2003, Antimicrobial suscep-
tibility pattern of clinical isolates of non–fermentive bacteria
Indian J Pathol Microbiol 46 3, pp 526–527
Mandell, G L, Petri, W A Hardman, J G, Limbird, L E, Molino,
P B, Ruddon, R W, & Gilman, A G eds, 1996, Antimicrobial
agents: Penicillins, Cephalosporins, and other B–lactam anti-
biotics e pharmacological basis of therapeutics pp 1073–1101
McGraw Hill
Massidda, O, Rossolini, G M, & Satta, G 1991, e Aeromonas
hydrophila cphA gene: molecular heterogeneity among class B
metallo–B–lactamases J Bacteriol 173, pp 4611–4617
Mendes, R E, Castanheira, M, Garcia, P, Guzman, M, Toleman, M A,
Walsh, T R, & Jones, R N 2004, First isolation of blaVIM–2 in Latin
America: Report from the SENTRY Antimicrobial Surveillance
Program Antimicrob Agents Chemother 48, pp 1433–1434
Miossec, C, Poirel, L, Livermore, D, Stachyra, T, Nordmann, P,
Black, M, & Levasseur, P 2007, In vitro activity of the new
B–lactamase inhibitor NXL104: restoration of ceftazidime CAZ,
ecacy against carbapenem–resistant Enterobacteriaceae
strains 47th Interscience Conference on Antimicrobial Agents
and Chemotherapy American Society for Microbiology,
Washington, DC
Miwa, H, Matsuda, H, & Shimada, J 1996, Eect of S–4661, a new
Carbapenem antibiotic on endotoxin release from Pseudomonas
aeruginosa 36th Interscience Conference on Antimicrobial
Agents Chemotherapy American Society for Microbiology,
Washington, DC
Miyazaki, S, Hosoyama, T, Furuya, N, Ishii, Y, Matsumoto, T, Ohno, A,
Tateda, K, & Yamaguchi, K 2001, In vitro and in vivo antibacte-
rial activities of L–084, a novel oral Carbapenem, against causa-
tive organism of respiratory tract infection Antimicrob Agents
Chemother 45, pp 203–07
Moland, E S, Hong, S G, omson, K S, Larone, D H, & Hanson, N D
2007, Klebsiella pneumoniae isolate producing at least eight
dierent B–lactamases, including AmpC and KPC B–lactamases
Antimicrob Agents Chemother 2007; 51, pp 800–801
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
b-lactams in clinical practice 107
Mori, M, Hikida, M, & Nishihara, T 1996, Comparative stability of car-
bapenem and penem antibiotics to human recombinant dehy-
dropeptidase–1 J Antimicrob Chemother 37, pp 1034–1036
Morinaka, A, Osaki, Y, Fukushima, T, Mikuniya, T, Yoshida, T, &
Yonezawa, M 2007, CP3242, a novel metallo–B–lactamase inhibi-
tor: inhibitory mechanism and in vitro activity 47th Interscience
Conference on Antimicrobial Agents and Chemotherapy
American Society for Microbiology, Washington, DC
Mushtaq, S, Warner, M, Ge, Y, Kaniga, K, & Livermore, D M 2007, In
vitro activity of ceftaroline PPI–0903M, T–91825, against bac-
teria with dened resistance mechanisms and phenotypes
J Antimicrob Chemother 60, pp 300–311
Mushtaq, S, Warner, M, Miossec, C, Woodford, N, & Livermore, D 2007,
NXL104/cephalosporin combinations vs Enterobacteriaceae
with CTX–M ESBLs 47th Interscience Conference on
Antimicrobial Agents and Chemotherapy American Society for
Microbiology, Washington, DC
Muto, Y, Matsumoto, K, Morinaka, A, Baba, N, Takayama, Y, Osaki, Y,
Kato, Y, Sugano, T, Yamada, K, Shimizu, A, Mikuniya, T, Takata,
H, Yoshida, T, & Yonezawa, M 2007, CP3242, a novel metallo–
B–lactamase inhibitor : Pharmacokinetics and in vivo pharma-
codynamic activities against metallo–B–lactamase–producing
P aeruginosa in a murine thigh infection model 47th Interscience
Conference on Antimicrobial Agents and Chemotherapy
American Society for Microbiology, Washington, DC
Nakagawa, S, Hashizume, T, Matsuda, K, Sanada, M, Okamoto, O,
Fukatsu, H, & Tanaka, N 1993, In vitro activity of a new
carbapenem antibiotic, BO–2727, with potent anti–pseudomo-
nal activity Antimicrob Agents Chemother 37, pp 2756–2759
Nakashima, M, Kato, T, & Kimura, Y 1994, S–4661, a new carbapenem:
IV pharmacokinetics in healthy volunteers 34th Interscience
Conference on Antimicrobial Agents and Chemotherapy
American society for microbiology, Washington, DC
Nishino, T, Otsuki, M, & Izawa, M 1996, In vitro and in vivo antibacterial
activity of S–4661, a new Carbapenem antibiotic 36th Interscience
Conference on Antimicrobial Agents and Chemotherapy American
Society for Microbiology, Washington, DC
Nordmann, P, Mariotte, S, Naas, T, Labia, R, & Nicholas, M H
1993, Biochemical properties of a carbapenem–hydrolyzing
B–lactamase from Enterobacter cloacae and cloning of the
gene into Escherichia coli Antimicrob Agents Chemother 37, pp
939–946
Ohba, F, Nakamura–Kamijo, M, Watanabe, N, & Katsu, K 1997, In
vitro and in vivo activity of ER–35786, a new anti–pseudomonal
carbapenem Antimicrob Agents Chemother 41, pp 298–307
Osano, E, Arakawa, Y, Wacharotayankun, R, Ohta, M, Horii, T, & Ito, H
1994, Molecular characterization of an enterobacterial metallo–
B–lactamase found in a clinical isolate of serratia marcescens
that shows imipenem resistance Antimicrob Agents Chemother
38, pp 71–78
Page, M G P, Desarbre, E, Geier, C, & Hofer, B 2007, B–lactamase
inhibitor combinations against B–lactam–resistant Gram–
negative bacteria 47th Interscience Conference on Antimicrobial
Agents and Chemotherapy American Society for Microbiology,
Washington, DC
PanKuch, G A, Davis, T A, & Jacobs, M R 2002, Anti–pneumococcal
activity of ertapenem MK–0826, compared to those of other
agents Antimicrob Agents Chemother 46, pp 42–46
Paterson, D L, & Bonomo, R A 2005, Extended–spectrum B–lactamases:
a clinical update Clin Microbiol Rev 18, pp 657–686
Paterson, D L, Adams, J, & Endimiani, A 2007, In vitro activity of
PZ–601, a novel carbapenem, against extended–spectrum beta–
lactamase producing organisms 47th Interscience Conference
on Antimicrobial Agents and Chemotherapy American Society
for Microbiology, Washington, DC
Petersen, P J, Jones, C H, & Bradford, P A 2007, In vitro activity of pip-
eracillin in combination with the penem B–lactamase inhibitor
BLI–489 against a predictor panel of characterized B–lactamase
producing organisms 47th Interscience Conference on
Antimicrobial Agents and Chemotherapy American Society for
Microbiology, Washington, DC
Poirel, L, Kampfer, P, & P Nordman 2002, Chromosome–encoded
Ambler class A beta–lactamase of Kluyvera georgiana, a probable
progenitor of a subgroup of CTX–M extended–spectrum beta–
lactamases Antimicrob Agents Chemother 46, pp 4038–4040
Poirel, L, Weldhagen, G F, Naas, T, De Champs, C, Dove, M G, &
Nordmann, P 2001, GES–2, a class A B–lactamase from
Pseudomonas aeruginosa with increased hydrolysis of imipenem
Antimicrob Agents Chemother 45, pp 2598–2603
Prats, G, Miro, E, Mirelis, B, Poirel, L, Bellais, S, & Nordmann, P 2002,
First isolation of a carbapenem–hydrolyzing B–lactamase in
Pseudomonas aeruginosa in Spain Antimicrob Agents Chemother
46, pp 932–933
Prezelj, A, Plantan, A, Vilfan, G, Stefanic Anderluh, P, Selic, L,
Iglicar, P, Car, V, Andrensek, S, Legen, I, Smrdel, P, Cerne, M,
Urleb, U, Locatelli, I, Mrhar, A, & Kovacic, N, 2007, Stability
of 10–ethylidene trinems, PK of LK–157 and design of pro-
drug esters 47th Interscience Conference on Antimicrobial
Agents and Chemotherapy American Society for Microbiology,
Washington, DC
Prezelj, A, Plantan, I, Vilfan, G, Stefanic Anderluh, P, Selic, L,
Iglicar, P, Iglicar, P, Car, V, Oblak, M, Solmajer, T, Copar, A ,
Urleb, U, Andrejasic, M, Turk, D, Paukner, S, & Hesse, L, 2007,
10– ethylidene trinems as broad–spectrum B–lactamase inhibi-
tors 47th Interscience Conference on Antimicrobial Agents and
Chemotherapy American Society for Microbiology, Washington,
DC
Queenan, A M, Shang, W, Kania, M, Page, M G P, & Bush, K 2007,
Interactions of Ceftobiprole with B–lactamases from molecular
classes A to D Antimicrob Agents Chemother 51, pp 3089–3095
Queenan, A M, Torres–Viera, C, Gold, H S, Carmeli, Y, Eliopoulos, GM,
Moellering, R C, Quinn, J P, Hindler, J, Medeiros, A A, & Bush, K
2000, SME–type carbapenem–hydrolyzing class A B–lactamases
from geographically diverse Serratia marcescens strains
Antimicrob Agents Chemother 44, pp 3035–3039
Rasmussen, B A, & Bush, K 1997, Carbapenem–hydrolyzing–B
lactamases Antimicrob Agents Chemother 41, pp 223–232
Rasmussen, B A, Keeney, D, Yang, Y, O’ Gara, C, Bush, K, &
Medeiros, A A 1994, Cloning, Sequencing and biochemical char-
acterization of a novel carbapenem–hydrolyzing B–lactamase
from Enterobacter cloacae 1994 34th Interscience Conference on
Antimicrobial Agents and Chemotherapy American Society for
Microbiology, Washington, DC
Reading, C, & Farmer, T 1981, e inhibition of B–lactamases from
Gram–negative bacteria by clavulanic acid Biochem J 199, pp
779–787
Richmond, M H, & Sykes, R B 1973, e B–lactamases of Gram–
negative bacteria and their possible physiological role Adv
Microb Physiol 9, pp 31–88
Rolinson, G N, & Stevens, S 1961, Microbiological studies on a new
broad spectrum penicillin ‘Penbritin’ British Med J 191–196
Rolinson, G N, & Sutherland, R 1967, Carbenicillin, a new semi–
synthetic penicillin active against Pseudomonas aeruginosa
Antimicrob Agents Chemother 7, pp 609–613
Rolinson, G N, Batchelor, F R, Butterworth, D, Cameronwood, J, Cole, M,
& Eustace, G C 1960, Formation of 6–aminopenicillanic acid from
pencillin by enzymatic hydrolysis Nature 187, pp 236–237
Rolinson, G N, Stevens, S, Batchelor, F R, Cameron–wood, J C, &
Chain, E B 1960, Bacteriological studies on a new penicillin
BRL– 1241 Lancettii, pp 564–567
Rolison, G N 1998, Forty years of B–lactam research 41, pp 589–603
Sader, H S, Fritsche, T R, & Jones, R N 2008, Antimicrobial activity of
ceftaroline and ME1036 tested against clinical strains of com-
munity–acquired methicillin–resistant Staphylococcus aureus
Antimicrob Agents Chemother 52, pp 1153–1155
Sader, H S, Fritsche, T R, Kaniga, K, Ge, Y, & Jones, R N 2005,
Antimicrobial activity and spectrum of PPI–0903M T–91825, a
novel cephalosporin, tested against worldwide collection of
clinical strains Antimicrob Agents Chemother 49, pp 3501–3512
Sader, H, Moet, G J, Fritsche, T R, & Jones, R N 2006, Evaluation of the
bactericidal activity of the novel cephalosporin ceftaroline PPI–
0903M, compared to ceftriaxone against Streptococcus pneumo-
niae 46th Interscience Conference on Antimicrobial Agents and
Chemotherapy San Francisco, CA
Saito, A, Inamatsu, T, & Shimada, J 1997, Clinical studies of S–4661,
new parenteral carbapenem antibiotic, in chronic respiratory
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
108 Shahid et al.
tract infections 37th Interscience Conference on Antimicrobial
Agents and Chemotherapy American Society for Microbiology,
Washington, DC
Sasaki, S, Murakami, K, & Nishitani, Y 1994, S– 4661, a new carbap-
enem, In vitro antibacterial activity 34th Interscience Conference
on Antimicrobial Agents and Chemotherapy American society
for Microbiology Washington, DC
Scoulica, E V, Neonakis, I K, Gikas, A I, & Tselentis, Y J 2004, Spread
of blaVIM–1–producing E coli in a university hospital in Greece
Genetic analysis of the integron carrying the blaVIM–1 metallo–
B–lactamase gene Diagn Microbiol Infect Dis 8, pp 167–172
Shahid, M 2007a, In vitro ecacy of Ceftriaxone/sulbactam against
Escherichia coli isolates producing CTX–M–15 Extended spec-
trum beta–lactamases J Antimicrob Chemother 60, pp 187–188
Shahid, M 2007b, Aminoglycosidic aminocyclitol antibiotics– a won-
der, but toxic drugs: developments and clinical implications
Anti–infective Agents Med Chem 6, pp 107–117
Shentag, J J 1990, e signicance of the relationship between
tissue:serum ratios, tissue concentrations and location of micro-
organisms Res Clin Forums 12, pp 23–27
Shibayama, T, Matsushita, Y, Kawai, K, Hirota, T, Ikeda, T, &
Kuwahara, S 2007, Pharmacokinetics and disposition of
CS–023RO4908463, a novel parenteral Carbapenem in animals
Antimicrob Agents Chemother 51, pp 257–263
Spratt, B G 1980, Biochemical and genetical approaches to the mech-
anism of action of penicillin Philos Trans R Soc Lond B Biol Sci
289, pp 273–283
Stachyra, T, Pechereau, M C, Petrella, S, Ziental–Gelus, N,
Sougako, W, Claudon, M, Black, M, Levasseur, P, Girard, AM, &
Miossec, C 2007, Activity of the new B–lactamase inhibitor
NXL104 against KPC B–lactamases 47th Interscience Conference
on Antimicrobial Agents and Chemotherapy American Society
for Microbiology, Washington, DC
Stone, K C, Dagan, R, Arguedas, A, Leibovitz, E, Wang, E, Echols, RM,
Janjic, N, & Critchley, I A 2007, Activity of Faropenem against
middle ear uid pathogens from children with acute otitis
media in Costa Rica and Israel Antimicrob Agents Chemother 51,
pp 2230–2235
Sumita, Y, Fukasawa, M, & Okuda, T 1990, Comparison of two car-
bapenems, SM–7338 and imipenem: anities for penicillin–
binding proteins and morphological changes J Antibiot Tokyo,
43, pp 314–320
Sumita, Y, Nouda, N, Kanazawa, K, & Fukasawa, M 1995, Antimicrobial
activity of SM–17466, a novel carbapenem antibiotic with potent
activity against methicillin–resistant Staphylococcus aureus
Antimicrob Agents chemother 39, pp 910–916
Sunagawa, M, Matsumura, H, Inoue, T, Fukasawa, M, & Kato, M 1990,
A novel carbapenem antibiotic, SM–7338: Structure–activity
relationships J Antibiot 43, pp 519–532
omson, K S, & Moland, E S 2004, CS–023 R–115685,, a novel carbap-
enem with enhanced in vitro activity against oxacillin– resistant
Staphylococci and P aeruginosa J Antimicrob Chemother 54, pp
557–562
omson, K S, Weber, D A, Sanders, C C, & Sanders, W E 1990, Beta–
lactamase production in members of family Enterobacteriaceae
and resistance to B–lactam–enzyme inhibitor combinations
Antimicrob Agents Chemother 34, pp 622–627
Toleman, M A, Bennett, P M, Walsh, T R 2006, ISCR elements: novel
gene capturing systems of the 21st century? Microbiol Mol Biol
Rev 70, pp 296–316
Toleman, M A, Rolston, K, Jones, R N, Walsh, T R 2003, Molecular and
biochemical characterization of OXA–45, an extended–spectrum
class 2d’ beta–lactamase in Pseudomonas aeruginosaAntimicrob
Agents Chemother 47, pp 2859–2863
Toleman, M A, Rolston, K, Jones, R N, & Walsh, T R 2004, blaVIM–7,
an evolutionarily distinct metallo–B–lactamase gene in a
Pseudomonas aeruginosa isolate from the United States
Antimicrob Agents Chemother 48, pp 329–332
Tomasz, A 1986, Penicillin–binding protiens and the antibacterial
eectiveness of B–lactam antibiotics Rev Infect Dis 8, Suppl3,
pp 260S–278S
Tomasz, A 1994, Multiple–antibiotic–resistant pathogenic bacteria A
report on the Rockefeller University workshop N Engl J Med 330,
pp 1247–1251
Totsuka, K, Shiseki, M, & Vchiyama, T 1996, In vitro postantibi-
otic eect and in vivo antimicrobial activity of novel carbap-
enem, S–4661 36th Interscience Conference on Antimicrobial
Agents and Chemotherapy American Society for Microbiology
Washington, DC
Tsuji, M, Ishii, Y, & Ohno, A 1998, In vitro and in vivo antibacte-
rial activities of S–4661, a new carbapenem Antimicrob Agents
Chemother 42, pp 184–187
Tsuji, M, Takema, M, Miwa, H, Shimada, J, & Kuwahara, S 2003, In
vivo antibacterial activity of S–3578, a new broad spectrum
Cephalosporin: Methicillin–resistant S aureus and P aeruginosa
experimental infection models Antimicrob Agents Chemother 47,
pp 2507–2512
Ueda, Y, & Sunagawa, M 2003, In vitro and in vivo activities of novel
2–thiazol–2–ylthio,– 1 B– methylcarbapenems with potent activ-
ities against multi–resistant Gram–positive bacteria Antimicrob
Agents chemother 47, pp 2471–2480
Walsh, T R 2002, Molecular characterization of SPM–1, a novel met-
allo– B–lactamase isolated in Latin America: report from the
SENTRY antimicrobial surveillance programme J Antimicrob
Chemother 50, pp 673–679
Walsh, T R, Toleman, M A, Poirel, L, & Nordman, P 2005, Metallo–
beta–lactamases: the quiet before the storm? Clin Microbiol Rev
18, pp 306–325
Wang, X, Minasov, G, & Shoichet, B K 2002, e structural bases of anti-
biotic resistance in the clinically derived mutant beta–lactamases
TEM–30, TEM–32, and TEM–34 J Biol Chem 77, pp32149–32156
Waxman, D J, Yocum, R R, & Strominger, J L 1980, Penicillins and
cephalosporins are active site directed acylating agents:
Evidence in support of the substrate analogue hypothesis Philos
Trans R– Soc Lond B Biol Sci 289, pp 257–271
Wexler, H M, & Finegold, S M 1988, In vitro activity of cefoperazone
plus sulbactam compared with that of other antimicrobial
agents against anaerobic bacteria Antimicrob Agents Chemother
323, pp 403–406
Wiseman, L R, Wagsta, A J, & Brogden, R N 1995, Meropenem A
review of its antibacterial activity, Pharmacokinetic properties
and clinical ecacy Drugs 50, pp 73–101
Yamano, Y, Nishikawa, T, & Komatsu, Y 1997, Eects of alterations
in porins and eux pumps of Pseudomonas aeruginosa on the
in vitro anti pseudomonal activity of S– 4661 37th Interscience
Conference on Antimicrobial Agents and Chemotherapy
American Society for Microbiology, Washington, DC
Yan, J J, Hsueh, P R, Ko, W C, Luh, K T, Tsai, S H, Wu, H M, & Wu, J J
2001, Metallo–B–lactamases in clinical Pseudomonas isolates in
Taiwan and identication of VIM–3, a novel variant of the VIM–2
enzyme Antimicrob Agents Chemother 45, pp 2224–2228
Yang, Y J, Wu, P–J, & Livermore, D M 1990, Biochemical characteri-
zation of a B–lactamase that hydrolyzes penems and carbap-
enems from two Serratia marcescens isolates Antimicrob Agents
Chemother 34, pp 755–758
Yigit, H, Queenan, A M, Anderson, G J, Domenech–Sanchez, A,
Biddle, J W, Steward, C D, Alberti, S, Bush, K, & Tenover F C
2001, Novel carbapenem–hydrolyzing B–lactamase, KPC–1,
from a carbapenem–resistant strain of Klebsiella pneumoniae
Antimicrob Agents Chemother 45, pp 1151–1161
Yoshimura, F, & Nikiado, H 1985, Diusion of B–lactam antibiotics
through the porin channels of E coli K–12 Antimicrob Agents
Chemother 27, pp 84–92
Yoshizawa, H, Itani, H, Ishikura, K, Irie, T, Yokoo, K, Kubota, T,
Minami, K, Iwaki, T, Miwa, H, & Nishitani, Y 2002, S–3578, a new
broad spectrum parenteral Cephalosporin exhibiting potent
activity against both methicillin–resistant Staphylococcus aureus
and Pseudomonas aeruginosa Synthesis and structure–activity
relationships J Antibiot Tokyo, 55, pp 975–992
Yum, J H, Yong, D, Lee, K, Kim, H S, & Chong, Y 2002, A new integron
carrying VIM–2 metallo–B–lactamase gene cassette in a Serratia
marcescens isolate Diagn Microbiol Infect Dis 42, pp 217–219
Downloaded By: [INFLIBNET India Order] At: 08:16 20 June 2009
... The antimicrobial susceptibility profile showed that most of the pathogens were resistant to TET, ampicillin, lincomycin, and penicillin, which include some of the most widely used antibiotics in agriculture [40]. MDR bacteria use multiple mechanisms to overcome the action of antimicrobials, such as active efflux or enzymatic degradation of the antimicrobial, mutation of an antimicrobial target site, and reduced cell wall permeability [41][42][43]. In Salmonella, the most common efflux pump system, belonging to the RND family is the AcrAB-TolC, responsible for resistance to various antibiotics [44]. ...
... We screened various combinations of antibiotics and effector molecules based on results from bacterial profiling for AMR and virulence characteristics. Synergism observed in the ampicillin-tazobactam combination could be ascribed to the action of tazobactam on the beta-lactamase enzyme, which was highly prevalent in both isolates [42]. Similarly, the synergistic interaction of the ampicillin-cinnamon EO combination could be attributed to membrane disruption by the EO, allowing easy passage of ampicillin into the cells. ...
Article
Full-text available
The emergence of multidrug-resistant (MDR) bacterial pathogens in farm animals and their zoonotic spread is a concern to both animal agriculture and public health. Apart from antimicrobial resistance (AMR), bacterial pathogens from the genera of Salmonella and Staphylococcus take refuge inside host cells, thereby demanding intervention strategies that can eliminate intracellular MDR pathogens. In this study, seven clinical isolates of Salmonella and Staphylococcus from swine farms were characterized for antibiotic (n = 24) resistance, resistance mechanisms, and virulence characteristics. All isolates showed resistance to one or more antibiotics and S. enterica ser. Typhimurium isolate had the highest resistance to the panel of antibiotics tested. Major resistance mechanisms identified were efflux pump and beta-lactamase enzyme activities. Staphylococcus isolates showed complete hemolysis and strong biofilm formation, while Salmonella isolates caused partial hemolysis, but showed no or weak biofilm formation. MDR isolates of S. aureus M12 and S. enterica ser. Typhimurium bacteria were subsequently tested against combinations of antibiotics and potentiating adjuvants for improved antibacterial efficacy using a checkerboard assay, and their fractional inhibitory concentration index (FICI) was calculated. A combination of chitosan and silica nanoparticles containing tetracycline (TET) and efflux pump inhibitor chlorpromazine (CPZ), respectively, was characterized for physicochemical properties and effectiveness against MDR Salmonella enterica ser. Typhimurium isolate. This combination of nano-encapsulated drugs improved the antibacterial efficacy by inhibiting AMR mechanisms (efflux activity, beta-lactamase enzyme activity, and hydrogen sulfide (H2S) production) and reducing intracellular pathogen load by 83.02 ± 14.35%. In conclusion, this study sheds light on the promising applicability of nanoparticle-enabled combination therapy to combat multidrug-resistant pathogens encountered in animal agriculture. Keywords: multi-drug resistance; combination therapy; nanoparticle; antimicrobial resistance; Staphylococcus; Salmonella; animal agriculture; pathogen.
... In 1928 Alexander Fleming's discovery of diffusible antimicrobial from mold pencillium known as opencillin lead to the evolution of β-lactam (Shahid et al., 2009). Antibiotics of β-lactam account for sixty percent of global consumption of antibiotic by mass and are considered to be the highly potent therapeutic compounds (Öztürk et al., 2015). ...
Chapter
Full-text available
With the increased prevalence of gram-negative bacterial infections, for which there are some effective therapies, the significance of hydrolyzing β-lactamase enzymes is also rising in certain significant clinical issues. Since the arrival of antibiotics, antibiotics of β-lactams are regarded as the highly effective antimicrobial agent. The primary pathway responsible for acquired β-lactam resistance in Gram-negative organisms is β-lactamase synthesis. Four different classes, i.e., A, B, C, and D, of β-lactamases have been explained. Enzymes of class B can also be called metallo-β-lactamases (MBLs) because they need a metal cofactor (for example, zinc in its natural form) to function. A, C, and D classes comprise of serine component as an activated center which catalyze the β-lactam ring hydrolysis via acyl-intermediate of serine. This chapter constitutes the knowledge of class A to D and brief about some of the β-lactamase inhibitors with their action mechanism.
... The changes corroborate the mode of action of meropenem, as it interferes with cell wall synthesis by blocking the activity of the penicillin-binding protein. 27,45,52 In Gram-negative bacteria, antibiotics belonging to cell wall synthesis inhibitors like cephalosporins and β-lactams also cause DNA damage. 46,53 Hence, the Raman spectral changes of meropenem-treated cells are similar to that of ciprofloxacin-treated cells. ...
Article
There has been a steep rise in the emergence of antibiotic-resistant bacteria in the past few years. A timely diagnosis can help in initiating appropriate antibiotic therapy. However, conventional techniques for diagnosing antibiotic resistance are time-consuming and labor-intensive. Therefore, we investigated the potential of Raman spectroscopy as a rapid surveillance technology for tracking the emergence of antibiotic resistance. In this study, we used Raman spectroscopy to differentiate clinical isolates of antibiotic-resistant and -sensitive bacteria of Escherichia coli, Acinetobacter baumannii, and Enterobacter species. The spectra were collected with or without exposure to various antibiotics (ciprofloxacin, gentamicin, meropenem, and nitrofurantoin), each having a distinct mechanism of action. Ciprofloxacin- and meropenem-treated sensitive strains showed a decrease in the intensity of Raman bands associated with DNA (667, 724, 785, 1378, 1480, and 1575 cm-1) and proteins (640 and 1662 cm-1), coupled with an increase in the intensity of lipid bands (891, 960, and 1445 cm-1). Gentamicin- and nitrofurantoin-treated sensitive strains showed an increase in the intensity of nucleic acid bands (668, 724, 780, 810, 1378, 1480, and 1575 cm-1) while a decrease in the intensity of protein bands (640, 1003, 1606, and 1662 cm-1) and the lipid band (1445 cm-1). The Raman spectral changes observed in the antibiotic-resistant strains were opposite to that of antibiotic-sensitive strains. The Raman spectral data correlated well with the antimicrobial susceptibility test results. The Raman spectral dataset was used for partial least-squares (PLS) analysis to validate the biomarkers obtained from the univariate analysis. Overall, this study showcases the potential of Raman spectroscopy for detecting antibiotic-resistant and -sensitive bacteria.
... The common enzymes were β-lactamase,include the penicillinases,cephalosporinases, oxacillinases, carbapenemases and extended spectrum β-lactamases , that hydrolyzes and resistance to cephalosporins, penicillins, and carbapenems (Bonomo and Szabo., 2006). The total number of the ESBLs now exceeds 200, the general principle of all the ESBL-detection methods are that the activity of the extended-spectrum of cephalosporins against the ESBL-producing organisms will be promote by presence the clavulanic acid (Shahid et al.,2009). Due to the increase in the spread of Acinetobacter baumannii resistant to β-lactams antibiotics especially the cephalosporins in the world and their emergence in our local hospitals, the this study was aimed to determine the ability of bacteria to produce Extended spectrum β-lactamase (ESBLs) by phenotypic methods. ...
Article
Full-text available
The aim of present study is to isolate and identify of Acinetobacter baumannii from burns infect as well as study their production for Extended spectrum β-lactamases (ESBLs).This study included 205 burn patients at Al-Hussien Teaching Hospital and other burn care units in Al-Nasiriyah city. The specimens were collected during period from August 2018 to February 2019. It were taken by burn swabs from pus of the burned area and then inoculated on blood agar and MacConkey agar. The isolates were diagnosed by microscopic examination, biochemical tests , API 20 E and Vitek-2 system. It was found that 20 isolates of A.baumannii and 2 isolates of A. loffii. Where identified from this study. The study showed 18 (90%) out of 20 isolates of A. baumannii were appeared resistance to cefotaxime and ceftazidime(screening test of ESBLs). Seven (35%) of isolates were producing ESBLs by confirmatory test(Modified double-disk synergy test (MDDST)) , and 13(65%) of isolates were negative results to production of the ESBLs.
... Currently, the majority of existing natural antibiotics are used to produce key intermediates of semisynthetic penicillins and cephalosporins, and only a small fraction of these drugs are directly applied in clinical treatment (Parmar et al., 2000). Compared with natural antibiotics, semisynthetic penicillins and cephalosporins have greater advantages, including higher clinical efficacy, lower toxicity, and a broader spectrum of bactericidal activity (Shahid et al., 2009). ...
Article
Full-text available
β-Lactam antibiotics are widely used anti-infection drugs that are traditionally synthesized via a chemical process. In recent years, with the growing demand for green alternatives, scientists have turned to enzymatic synthesis. Penicillin G acylase (PGA) is the second most commercially used enzyme worldwide with both hydrolytic and synthetic activities toward antibiotics, which has been used to manufacture the key antibiotic nucleus on an industrial level. However, the large-scale application of PGA-catalyzed antibiotics biosynthesis is still in the experimental stage because of some key limitations, such as low substrate concentration, unsatisfactory yield, and lack of superior biocatalysts. This paper systematically reviews the strategies adopted to improve the biosynthesis of β-lactam antibiotics by adjusting the enzymatic property and manipulating the reaction system in recent 20 years, including mining of enzymes, protein engineering, solvent engineering, in situ product removal, and one-pot reaction cascade. These advances will provide important guidelines for the future use of enzymatic synthesis in the industrial production of β-lactam antibiotics.
Article
Full-text available
Halo-cycloetherification of lactam-tethered alkenols enables the construction of oxygen-heterocycles that are fused to nitrogen heterocycles via intramolecular halonium-induced nucleophilic addition. Specifically, tetrahydropyrans (THPs) that are fused to a nitrogen heterocycle constitute the core of several bioactive molecules, including tachykinin receptor antagonists and alpha-1 adrenergic antagonists. Although the literature is replete with successful examples of the halo-cycloetherification of simple mono- or disubstituted primary alkenols, methods for the modular, efficient, regioselective, and stereocontrolled intramolecular haloetherification of sterically encumbered trisubstituted tertiary alkenols are rare. Here, we describe a simple intramolecular bromoetherification strategy that meets these benchmarks and proceeds with exclusive 6-endo regioselectivity. The transformation employs mild and water-tolerant conditions, which bodes well for late-stage diversification. The hindered ethers contain four contiguous stereocenters as well as one halogen-bearing tetrasubstituted stereocenter.
Article
Full-text available
The devastating impact of Tuberculosis (TB) has been a menace to mankind for decades. The World Health Organization (WHO) End TB Strategy aims to reduce TB mortality up to 95% and 90% of overall TB cases worldwide, by 2035. This incessant urge will be achieved with a breakthrough in either a new TB vaccine or novel drugs with higher efficacy. However, the development of novel drugs is a laborious process involving a timeline of almost 20-30 years with huge expenditure; on the other hand, repurposing previously approved drugs is a viable technique for overcoming current bottlenecks in the identification of new anti-TB agents. The present comprehensive review discusses the progress of almost all the repurposed drugs that have been identified to the present day (∼100) and are in the development or clinical testing phase against TB. We have also emphasized the efficacy of repurposed drugs in combination with already available frontline anti-TB medications along with the scope of future investigations. This study would provide the researchers a detailed overview of nearly all identified anti-TB repurposed drugs and may assist them in selecting the lead compounds for further in vivo/clinical research.
Article
Full-text available
We characterized five carbapenemase-producing Enterobacterales (CPE) isolates from two health care institutions in Lima, Peru. The isolates were identified as Klebsiella pneumoniae (n = 3), Citrobacter portucalensis (n = 1), and Escherichia coli (n = 1). All were identified as blaOXA-48-like gene carriers using conventional PCR. Whole-genome sequencing found the presence of the blaOXA-181 gene as the only carbapenemase gene in all isolates. Genes associated with resistance to aminoglycosides, quinolones, amphenicols, fosfomycins, macrolides, tetracyclines, sulfonamides, and trimethoprim were also found. The plasmid incompatibility group IncX3 was identified in all genomes in a truncated Tn6361 transposon flanked by ΔIS26 insertion sequences. The qnrS1 gene was also found downstream of blaOXA-181, conferring fluoroquinolone resistance to all isolates. CPE isolates harboring blaOXA-like genes are an increasing public health problem in health care settings worldwide. The IncX3 plasmid is involved in the worldwide dissemination of blaOXA-181, and its presence in these CPE isolates suggests the wide dissemination of blaOXA-181 in Peru. IMPORTANCE Reports of carbapenemase-producing Enterobacterales (CPE) isolates are increasing worldwide. Accurate detection of the β-lactamase OXA-181 (a variant of OXA-48) is important to initiate therapy and preventive measures in the clinic. OXA-181 has been described in CPE isolates in many countries, often associated with nosocomial outbreaks. However, the circulation of this carbapenemase has yet to be reported in Peru. Here, we report the detection of five multidrug-resistant CPE clinical isolates harboring blaOXA-181 in the IncX3-type plasmid, a potential driver of dissemination in Peru.
Article
Full-text available
Halolactonization of alkenoic acids enables the construction of oxygen-heterocycles via intramolecular halonium-induced nucleophilic addition. Although the literature is currently inundated with halolactonizations of 5-aryl-4(E)-pentenoic acids that predictably afford the 6-endo cyclization adducts, methods that reliably alter the innate regioselectivity bias to instead deliver the thermodynamically less favored 5-exo cyclization products are relatively rare. Here, we attempt to bridge this gap and have found mild conditions for contra-thermodynamic halolactonization of lactam-tethered 5-aryl-4(E)-pentenoic acids that lead to the formation of trans-fused lactam-γ-lactones. The natural proclivity for these 5-aryl-4(E)-pentenoic acids to undergo 6-endo cyclization is overridden and 5-exo-trig cyclization predominates. The success of the approach hinges on the use of N,N-dimethylformamide (DMF) as the solvent and N-methylmorpholine oxide as the catalyst. The lactam-lactone products are synthesized in high diastereoselectivity, modularity, and chemoselectivity. Notably, most of the bicycles contain one benzylic quaternary stereocenter as well as an α-alkoxy quaternary stereocenter.
Article
Full-text available
Methicillin resistance in Staphylococcus aureus has been associated with alterations in the penicillin-binding proteins (PBPs). An intriguing property of all methicillin-resistant staphylococci is the dependence of resistance on the pH value of the growth medium. Growth of such bacteria at pH 5.2 completely suppressed the expression of methicillin resistance. We have examined the PBP patterns of methicillin-resistant staphylococci grown at pH 7.0. We detected a high-molecular-weight PBP (PBP-2a; approximate size, 78,000 daltons) that was only present in the resistant bacteria but not in the isogenic sensitive strain. In cultures grown at pH 5.2, the extra PBP was not detectable.
Article
beta-Lactamases are the commonest single cause of bacterial resistance to beta-lactam antibiotics. Numerous chromosomal and plasmid-mediated types are known and may be classified by their sequences or phenotypic properties. The ability of a beta-lactamase to cause resistance varies with its activity, quantity, and cellular location and, for gram-negative organisms, the permeability of the producer strain. beta-Lactamases sometimes cause obvious resistance to substrate drugs in routine tests; often, however, these enzymes reduce susceptibility without causing resistance at current, pharmacologically chosen breakpoints. This review considers the ability of the prevalent beta-lactamases to cause resistance to widely used beta-lactams, whether resistance is accurately reflected in routine tests, and the extent to which the antibiogram for an organism can be used to predict the type of beta-lactamase that it produces.
Article
The Staphylococcus aureus lrgAB operon was recently shown to inhibit extracellular murein hydrolase activity and increase tolerance to penicillin. Further characterization of this operon could provide novel insight into the dynamics of S. aureus cell wall metabolism and the mechanism of penicillin-induced lethality.
Article
This article has no abstract; the first 100 words appear below. IT has been said that curing all cancers would add 2 years to the life expectancy of Americans but that the introduction of antibiotics added 10 years.¹ The agent most responsible for this remarkable effect is penicillin, the prototypic beta-lactam antibiotic. The beta-lactam antibiotics include penicillins, cephalosporins, monobactams, and carbapenems. All have a beta-lactam ring, which is essential for antibacterial activity. In general, these agents have very favorable ratios of therapeutic to toxic effects, and the newer agents have enhanced antibacterial spectrums or favorable pharmacokinetic characteristics. Mechanism of Action Beta-lactam antibiotics are bactericidal because they inhibit bacterial cell-wall synthesis. Their . . . Source Information From the Infectious Diseases Division, Department of Internal Medicine, University of Virginia Medical Center, Box 385, Charlottesville, VA 22908, where reprint requests should be addressed to Dr. Donowitz.
Article
The gene encoding the metallo-β-lactamase SPM-1 was cloned from a genomic library of Pseudomonas aeruginosa strain 48-1997A. The insert carrying spm-1 possessed a GC content of 47%, indicating that it is of non-Pseudomonas origin. Upstream of spm-1 there is a small open reading frame (ORF), which is homologous to the LysR family of proteins (69% identity to the LysR protein from Salmonella enterica serovar Typhimurium). Downstream of spm-1 there is the start of an ORF, the product of which shows close homology with the GroEL-type proteins from Xanthomonas campestris. No transmissible element could be identified upstream or down- stream of spm-1. The spm-1 gene is carried on a plasmid that can transform both Escherichia coli and P. aeruginosa to ceftazidime resistance. SPM-1 contains the classic metallo-β-lactamase zinc-binding motif HXHXD and shows the highest identity (35.5%) to IMP-1. SPM-1 is a distinctly different metallo-β-lactamase from VIM and IMP and, accordingly, represents a new subfamily of mobile metallo-β-lactamases. The predicted molecular weight of the protein was 27 515 Da, significantly higher than that of IMP (25 041 Da) or VIM (25 322 Da). SPM-1 possesses a unique loop of 23 residues that accounts for the higher molecular mass.
Article
Abstract Ertapenem, a carbapenem antibacterial, has in vitro activity against many Gramnegative (including Enterobacteriaceae) and Gram-positive aerobic and anaerobic bacteria that are commonly associated with various infections. Once-daily parenteral (intravenous or intramuscular) ertapenem 1g was as effective as comparator antimicrobial agents (piperacillin/tazobactam or ceftriax-one ± metronidazole) in patients with bacterial infections in randomised, double-blind, multicentre clinical trials. Response rates with ertapenem were 84% and 87% (combined microbiological and clinical) in patients with complicated intra-abdominal infections (CIAI), 82% (clinical) in patients with complicated skin and skin structure infections (CSSSI), 86% and 92% (microbiological) in patients with complicated urinary tract infections (CUTI), 92% (clinical) in patients with community-acquired pneumonia (CAP) associated with typical pathogens and 94% (clinical) in patients with acute pelvic infection. Respective response rates were statistically equivalent to those with comparators (81–94%). The efficacy of ertapenem was equivalent to that of piperacillin/tazobactam in patients infected with Enterobacteriaceae or anaerobes and to ceftriaxone in patients infected with Enterobacteriaceae. Ertapenem was generally well tolerated by patients with bacterial infections, with most adverse events being mild to moderate in severity. The most common ertapenem-associated adverse events were diarrhoea, infused vein complication, nausea, headache, vaginitis in females, phlebitis and/or thrombophlebitis and vomiting. Conclusion: Ertapenem is a broad-spectrum parenteral antibiotic with activity against many Gram-negative (including Enterobacteriaceae) and Gram-positive aerobic and anaerobic bacteria and is suitable for once-daily administration. Ertapenem has a role in the treatment of CAP associated with typical respiratory pathogens and is of particular value in the treatment of polymicrobial infections (such as CIAI, CSSSI, CUTI and acute pelvic infections), especially where Enterobacteriaceae and anaerobic bacteria are involved. Antibacterial Activity Ertapenem is active against many Enterobacteriaceae (including extended-spectrum and AmpC β-lactamase-producing isolates), common Gram-negative respiratory tract pathogens (including Moraxella catarrhalis and Haemophilus influenzae), but has negligible activity against nonfermenting aerobes (e.g. Pseudomonas aeruginosa and Acinetobacter baumanni). Ertapenem is active against most Gram-positive bacteria (including penicillin-susceptible and -resistant Streptococcus pneumoniae and methicillin/oxacillin-susceptible Staphylococcus aureus), but not Enterococcus spp. (E. faecalis and E. faecium) and methicillin/oxacillin-resistant S. aureus. Ertapenem is active against a wide range of clinical isolates of Gram-negative (Bacteroides spp., Prevotella spp., and Porphyromonas spp., including β-lactamase-producing strains) and Gram-positive (e.g. Peptostreptococcus spp. and Clostridium spp.) anaerobes. However, ertapenem has limited activity against Lactobacillus spp. and varying activity against Bilophila wadsworthia. Ertapenem shows a positive post-antibiotic effect (PAE) against Gram-positive isolates (S. pneumoniae, S. aureus), but limited (E. cloacae), none (H. influenzae) or negative (Escherichia coli) PAE against Gram-negative isolates. Ertapenem is bactericidal against many common pathogens; the antibacterial activity of ertapenem is time-dependent. Ertapenem is stable to most of the β-lactamases produced by Gram-negative bacteria; however, ertapenem is susceptible to the metallo-β-lactamases produced by some Enterobacteriaceae. Alterations in penicillin binding protein account for the resistance of methicillin/oxacillin-resistant Staphylococcus spp. and Enterococcus spp. to ertapenem. Reduced permeability as a result of porin deficiency may have contributed to resistance in a K. pneumoniae isolate. Pharmacokinetic Properties After administration of a single dose of ertapenem 1g, mean maximum total plasma concentrations were 164.6 mg/L at the end of a 30-minute intravenous (IV) infusion and 70.6 mg/L after a single intramuscular (IM) injection; mean area under the plasma concentration time curve (AUC) were 597.4 (IV) and 524.9 (IM) mg · h/L. Following IM administration of ertapenem 1 g/day, maximum drug plasma concentration was reached after 2 hours. Plasma protein binding is approximately 95% for plasma concentrations <100 mg/L and approximately 82–85% for concentrations of ≈300 mg/L. Mean volume of distribution at steady state is approximately 8L. Ertapenem exhibits good skin-blister fluid penetration. After multiple doses of ertapenem 1 g/day, the mean ratio of AUC for ertapenem in blister fluid to AUC in plasma was 0.61. The major metabolite of ertapenem is a ring-opened derivative formed by hydrolysis of the β-lactam ring. Ertapenem is primarily eliminated by the kidneys. The mean plasma elimination half-life (t1/2) of a single dose of IV or IM ertapenem 1g was ≈4 hours. The mean apparent plasma clearance of a single 30-minute infusion of ertapenem 1g was 1.7 L/h (28 mL/min). The AUC and t1/2 of ertapenem in patients with renal impairment were increased compared with those of healthy volunteers. For patients with impaired renal function given a single 1g dose of ertapenem, t1/2 values were 4–14 hours and AUC values increased by 7–192% depending on the extent of renal impairment. Therapeutic Efficacy In patients with complicated intra-abdominal infections (CIAI), complicated skin and skin structure infections (CSSSI), complicated urinary tract infection (CUTI), community-acquired pneumonia (CAP) and acute pelvic infections, parenteral ertapenem 1 g/day was as effective as parenteral administration of comparator antimicrobial agents (piperacillin/tazobactam 3.375g every 6 hours, ceftriaxone 1 g/day, ceftriaxone 2 g/day plus metronidazole 500mg every 8 hours) in randomised, double-blind, multicentre clinical trials. The duration of treatment (parenteral plus optional oral therapy, if permitted) was generally 3–14 days for CIAI, 7–14 days for CSSSI, 10–14 days for CUTI and CAP and 2–12 days for acute pelvic infections. In microbiologically evaluable patients with CIAI, 84% and 87% of ertapenem, 81% of piperacillin/tazobactam and 85% of ceftriaxone plus metronidazole recipients achieved favourable combined clinical and microbiological responses at the test of cure (TOC) visit (28–42 days after completion of therapy); statistical equivalence between ertapenem and the comparator treatment was established. In patients with CSSSI, treatment with ertapenem or piperacillin/tazobactam resulted in statistically equivalent clinical response (82% and 84% ) and microbiological response (both 83%) rates at the time of TOC (10–21 days after completion of therapy) in a well designed trial. Clinical response rates were similar in patients with different types or severities of infection. In patients with CUTI, the microbiological response rates were statistically equivalent in parenteral ertapenem (86% and 92%) and parenteral ceftriaxone (85% and 93%) recipients in a two well designed study at TOC assessment (5–9 days after completion of therapy). Clinical and microbiological response rates were high (>90%) and statistically equivalent in ertapenem and ceftriaxone recipients in two well designed trials in patients with CAP at TOC (7–14 days after completion of therapy). S. pneumoniae, M. catarrhalis, H. influenzae and S. aureus were the most commonly isolated pathogens. In microbiologically evaluable patients infected with S. pneumoniae (the most common pathogen), similar clinical and microbiological response rates occurred with ertapenem for both penicillin-susceptible and penicillin-nonsusceptible infections. In women with acute pelvic infections (obstetric and gynaecological postoperative infections), clinical responses rates at TOC (14–28 days after completion of therapy) were 94% with ertapenem and 92% with piperacillin/tazobactam in clinically evaluable patients in a well designed study. Statistical equivalence between the groups was established. In a combined subgroup analysis of 1167 patients with infections caused by Enterobacteriaceae from seven randomised, double-blind trials, ertapenem was as effective as piperacillin/tazobactam (CIAI, CSSSI and acute pelvic infection) or ceftriaxone (CUTI and CAP). Similarly, in a combined subgroup analysis of 296 evaluable patients with infections involving anaerobic bacteria from three randomised, double-blind trials (CIAI, CSSSI and acute pelvic infections), ertapenem was as effective as piperacillin/tazobactam (overall response rates 89.3% vs 85.9%). Tolerability Most adverse events associated with parenteral ertapenem 1 g/day in patients with bacterial infections were mild to moderate in severity. In a pooled analysis of clinical trials, discontinuation due to adverse experiences occurred in 1.3% of ertapenem recipients. The most common ertapenem-associated adverse events during therapy plus the 14-day follow-up period were diarrhoea (5%), infused vein complication (5%) and nausea (3%). The incidence of adverse experiences was similar in recipients of ertapenem and its comparator (piperacillin/tazobactam or ceftriaxone). scizure was reported in 0.5% of ertapenem recipients, 0.3% in piperacillin/tazobactam recipients and in none of the ceftriaxone recipients. Drug-related laboratory adverse events included increases in alanine transferase (5%), aspartate transferase (5%), serum alkaline phosphatase (4%) and platelet count (3%). Dosage and Administration In the US, ertapenem is indicated for the treatment of adult patients with CIAI, CSSSI, CAP, CUTI and acute pelvic infections (including postpartum endomyometritis, septic abortion and post surgical gynaecological infections) caused by susceptible strains of causative organisms. In Europe, ertapenem is indicated for CIAI, CAP and acute gynaecological infections, caused by susceptible strains of causative organisms. Ertapenem 1g is administered once daily by intravenous infusion (over 30 minutes) for up to 14 days, or by intramuscular injection for up to 7 days (in patients for which intramuscular therapy is appropriate). When clinically indicated, a switch to an appropriate oral antibacterial agent may be implemented if clinical improvement has occurred. Patients with advanced renal impairment and end-stage renal insufficiency should receive reduced ertapenem dosages. No dosage adjustment recommendations have been made based on age or gender or for patients with impaired hepatic function.
Article
Community-acquired and nosocomial infections caused by multidrug-resistant Gram-positive pathogens continue to increase in prevalence and have become a serious problem in many parts of the world. BAL9141 is a member of the class of parenteral pyrrolidinone-3-ylidenemethyl cephalosporins, and has a broad spectrum of activity. In the current study, BAL9141 was tested against a large number (n = 2263) of recent isolates from various international surveillance programmes including 1097 Gram-positive strains. Susceptibility to (S) and activity of (mg/L) to BAL9141, based on proposed breakpoints (MIC 50 /MIC 90 /% S) were as follows: methicillin- susceptible Staphylococcus aureus (0.5/0.5/100%), methicillin-resistant S. aureus (MRSA) (1/2/100%), methicillin-susceptible coagulase-negative staphylococci (CoNS) (0.12/0.25/100%), methicillin-resistant CoNS (MR-CoNS) (1/2/100%), Streptococcus pneumoniae (≤0.015/0.25/ 100%), viridans group streptococci (0.03/0.5/99%), β-haemolytic streptococci (≤0.015/≤0.015/ 100%), Enterococcus faecalis (0.5/16/90%), Enterococcus faecium (>32/>32/22%), Haemophilus influenzae (0.06/0.06/100%), Moraxella catarrhalis (0.06/0.5/100%), Neisseria gonorrhoeae (0.03/0.06/100%) and Neisseria meningitidis (≤0.002/0.004/100%). BAL9141 susceptibility at ≤4 mg/L (100% S) surpassed that of ceftriaxone (CRO; 1% S) and quinupristin/dalfopristin (Q-D; 92% S) against MRSA and MR-CoNS (CRO 0.9% S; Q-D 94% S). All S. pneumoniae were inhibited by BAL9141 at ≤1 mg/L compared with CRO (90% S) and levofloxacin (LVX; 98% S). Susceptibility rates for viridans group streptococci to BAL9141 (>98%) were also higher than to CRO (86%) and LVX (96%). BAL9141 demonstrated excellent activity against most species of wild-type enteric bacilli, with ≥95% of isolates being susceptible; however, only modest activity was observed for BAL9141 against non-fermentative Gram-negative species and ESBL-producing Escherichia coli or Klebsiella pneumoniae. BAL9141 demonstrated excellent activity against many tested pathogens displaying various resistance phenotypes, and should be particularly valuable in the treatment of MRSA as well as for drug-resistant streptococci, while maintaining a spectrum resembling a 'third-generation' cephalosporin against other clinically important species.
Article
In 1945, after penicillin had been introduced into medicine, an antibiotic-producing species of Cephalosporium was isolated from a sewage outfall in Sardinia. Four years later in Oxford, this organism was found to produce several antibiotics, one of which was a penicillin with a new side-chain, penicillin N. During a chemical study in 1953, this penicillin was shown to be contaminated with a second substance, cephalosporin C, which contained a β-lactam ring but was resistant to hydrolysis by a penicillinase (β-lactamase). At that time, penicillinase-producing Staphylococci were causing a serious problem in hospitals. The isolation of the nucleus of cephalosporin C (7-ACA) enabled pharmaceutical manufacturers to produce many thousands of cephalosporins, some of which have been effective in the treatment of serious infections by a number of Gram-positive and Gram-negative bacteria. The cephalosporins, like the newer penicillins, have a very low toxicity and have greatly extended the range of chemotherapy. New, sensitive screening methods have revealed further families of clinically useful substances that contain a reactive β-lactam ring. Genetic engineering has now begun to throw light on the nature of the enzymes that are involved in the biosynthesis of penicillins and cephalosporins, and x-ray crystallography may soon provide detailed 3-dimensional pictures of some of the bacterial enzymes with which the active β -lactam ring reacts. Rational approaches to the production and design of new and potentially useful compounds may then be within sight.
Article
Two species resulting from the reaction of the SHV-1 class A beta -lactamase with the sulfone inhibitor tazobactam have been trapped at 100 K and mapped by X-ray crystallography at 2.0 Angstrom resolution. An acyclic form of tazobactam is covalently bonded to the catalytic Ser70 side chain, and a second species, a five-atom vinyl carboxylic acid fragment of tazobactam, is bonded to Ser130. It is proposed that the electron density map of the crystal is a composite picture of two complexes, each with only a single bound species. It is estimated that the two complexes exist in the crystal in approximately equal populations. Results are discussed in relation to the mechanism-based inhibition of class A beta -lactamases by the similar inhibitors sulbactam and clavulanic acid.