Sei sulla pagina 1di 42

CLINICAL MICROBIOLOGY REVIEWS, Jan. 2010, p. 160–201 Vol. 23, No.

1
0893-8512/10/$12.00 doi:10.1128/CMR.00037-09
Copyright © 2010, American Society for Microbiology. All Rights Reserved.

Three Decades of ␤-Lactamase Inhibitors


Sarah M. Drawz1 and Robert A. Bonomo2,3,4,5*
Departments of Pathology,1 Medicine,2 Pharmacology,3 and Molecular Biology and Microbiology,4
Case Western Reserve University School of Medicine, Cleveland, Ohio, and Research Service,
Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, Ohio5

INTRODUCTION .......................................................................................................................................................161
MECHANISM OF ACTION OF ␤-LACTAM ANTIBIOTICS .............................................................................161
RESISTANCE TO ␤-LACTAM ANTIBIOTICS .....................................................................................................161
␤-LACTAMASES ........................................................................................................................................................164
Classification ...........................................................................................................................................................164
Class A serine ␤-lactamases .............................................................................................................................164
Class A ESBLs ....................................................................................................................................................164
Class A serine carbapenemases ........................................................................................................................165
Class B metallo-␤-lactamases ...........................................................................................................................165
Class C serine cephalosporinases.....................................................................................................................166
Class D serine oxacillinases ..............................................................................................................................166
␤-Lactamase Hydrolytic Mechanisms..................................................................................................................166
Class A .................................................................................................................................................................166
Class C .................................................................................................................................................................168
Class D .................................................................................................................................................................168
Class B .................................................................................................................................................................168
CIRCUMVENTING ␤-LACTAMASES ....................................................................................................................169
␤-Lactamase Inhibitors: Mechanistic Considerations.......................................................................................169
␤-Lactamase Inhibitors in Clinical Practice ......................................................................................................169
Clavulanic acid, sulbactam, and tazobactam .......................................................................................169
Mechanism of inhibition....................................................................................................................................170
␤-Lactam–␤-lactamase inhibitor combinations: clinical use .......................................................................171
(i) Amoxicillin-clavulanate ............................................................................................................................171
(ii) Ticarcillin-clavulanate .............................................................................................................................172
(iii) Ampicillin-sulbactam..............................................................................................................................173
(iv) Piperacillin-tazobactam ..........................................................................................................................173
Inhibitory Activity of Carbapenems .....................................................................................................................174
INHIBITOR-RESISTANT CLASS A ␤-LACTAMASES........................................................................................176
Epidemiology and Detection of ␤-Lactamase Inhibitor Resistance.................................................................176
Substitutions in Class A Enzymes Conferring Inhibitor Resistance...............................................................178
Arginine 244.........................................................................................................................................................179
Methionine 69......................................................................................................................................................179
Asparagine 276 ....................................................................................................................................................179
Serine 130 ............................................................................................................................................................180
Arginine 275.........................................................................................................................................................180
Lysine 234 ............................................................................................................................................................180
THE PROMISE OF NOVEL ␤-LACTAMASE INHIBITORS .............................................................................180
Monobactam Derivatives .......................................................................................................................................180
ATMO Derivatives ..................................................................................................................................................182
Penems .....................................................................................................................................................................182
BRL 42715 and Syn 1012...................................................................................................................................182
BRL 42715 derivatives........................................................................................................................................183
Oxapenems...........................................................................................................................................................184
Tricyclic carbapenems (trinems) ......................................................................................................................184
1-␤-Methylcarbapenems.....................................................................................................................................184
Penicillin and Cephalosporin Sulfone Derivatives.............................................................................................184
C-2/C-3-substituted penicillin and cephalosporin sulfones ..........................................................................185
C-6-substituted penicillin sulfones ...................................................................................................................185

* Corresponding author. Mailing address: Infectious Diseases Sec-


tion, Louis Stokes Cleveland Department of Veterans Affairs Medical
Center, 10701 East Blvd., Cleveland, OH 44106. Phone: (216) 791-
3800, ext. 4399. Fax: (216) 231-3482. E-mail: robert.bonomo@med.va
.gov.

160
VOL. 23, 2010 ␤-LACTAMASE INHIBITORS 161

Non-␤-Lactam Inhibitors.......................................................................................................................................186
Boronic acid transition state analogs (TSAs).................................................................................................186
Phosphonates.......................................................................................................................................................187
NXL104.................................................................................................................................................................187
Hydroxamates......................................................................................................................................................187
Other non-␤-lactam inhibitors .........................................................................................................................188
INHIBITION OF METALLO-␤-LACTAMASES ...................................................................................................188
Thiol Derivatives .....................................................................................................................................................189
Pyridine Dicarboxylates .........................................................................................................................................189
Trifluoromethyl Ketones and Alcohols ................................................................................................................189
Carbapenem Analogs..............................................................................................................................................189
Tricyclic Natural Products ....................................................................................................................................189
Succinate Derivatives .............................................................................................................................................189
C-6-Mercaptomethyl Penicillinates ......................................................................................................................190
LESSONS LEARNED ................................................................................................................................................190
A PERSPECTIVE .......................................................................................................................................................191
ACKNOWLEDGMENTS ...........................................................................................................................................191
REFERENCES ............................................................................................................................................................191

INTRODUCTION the bacterial cell wall is essential to maintaining cell shape in a


hypertonic and hostile environment (249). Osmotic stability is
The development of antibiotics remains one of the most sig- preserved by a rigid cell wall comprised of alternating N-acetyl-
nificant advances in modern medicine (364). Antibiotics have muramic acid (NAM) and N-acetylglucosamine (NAG) units.
saved countless lives and continue to be a mainstay of therapy for These glycosidic units are linked by transglycosidases. A pen-
bacterial infections. The clinical success of the first ␤-lactam, tapeptide is attached to each NAM unit, and the cross-linking of
penicillin G (benzylpenicillin [Fig. 1, compound 1), prompted the two D-alanine–D-alanine NAM pentapeptides is catalyzed by
search for and development of additional derivatives. This quest PBPs, which act as transpeptidases (142, 377). This cross-linking
gave rise to the ␤-lactam antibiotics in clinical use today (penicil- of adjacent glycan strands confers the rigidity of the cell wall.
lins, narrow- and extended-spectrum cephalosporins, monobac- The ␤-lactam ring is sterically similar to the D-alanine–D-
tams, and carbapenems [Fig. 1, compounds 1 to 7) (14). The alanine of the NAM pentapeptide, and PBPs “mistakenly” use
common structural feature of these classes of antibiotics is the the ␤-lactam as a “building block” during cell wall synthesis
highly reactive four-membered ␤-lactam ring. (459). This results in acylation of the PBP, which renders the
Unfortunately, ␤-lactamase-mediated resistance to ␤-lactam enzyme unable to catalyze further transpeptidation reactions
antibiotics emerged as a significant clinical threat to these life- (125). As cell wall synthesis slows to a halt, constitutive pepti-
saving drugs. In response to this challenge, two strategies were doglycan autolysis continues. The breakdown of the murein
advanced to preserve the utility of ␤-lactam antibiotics: (i) dis- sacculus leads to cell wall compromise and increased perme-
cover or design ␤-lactam antibiotics that are able to evade bacte- ability. Thus, the ␤-lactam-mediated inhibition of transpepti-
rial enzymatic inactivation conferred by ␤-lactamases, or (ii) in- dation causes cell lysis, although the specific details of penicil-
hibit ␤-lactamases so the partner ␤-lactam can reach the penicillin lin’s bactericidal effects are still being unraveled (20).
binding proteins (PBPs), the target of ␤-lactam antibiotics.
In this review, we summarize 3 decades of investigation of
␤-lactamase inhibition. This perspective is framed by our back- RESISTANCE TO ␤-LACTAM ANTIBIOTICS
ground in clinical infectious diseases. First, we highlight the fun-
damental principles of ␤-lactamase enzymology. We then sum- There are four primary mechanisms by which bacteria can
marize the salient features of ␤-lactam–␤-lactamase inhibitor overcome ␤-lactam antibiotics (14).
combinations that are used in clinical practice. Next, we define the (i) Production of ␤-lactamase enzymes is the most common
problem of resistance to ␤-lactamase inhibitors by explaining the and important mechanism of resistance in Gram-negative bac-
important changes in class A ␤-lactamases that define this phe- teria and will be the focus of this review.
notype. With this background, we review the ␤-lactamase inhib- (ii) Changes in the active site of PBPs can lower the affinity
itors that have been developed to this point and discuss the novel for ␤-lactam antibiotics and subsequently increase resistance
␤-lactamase inhibitors that are hoped to extend the life span of to these agents, such as those seen in PBP2x of Streptococcus
our current ␤-lactams. We view these agents as extremely impor- pneumoniae (212). Through natural transformation and re-
tant to the future of ␤-lactam therapy: inhibitors not only can combination with DNA from other organisms, Neisseria spp.
preserve our current armamentarium but may also be used as and Streptococcus spp. have acquired highly resistant, low-
novel ␤-lactams are introduced into the clinic. Finally, we con- affinity PBPs (39, 313, 459). In a related manner, penicillin
clude with some “lessons learned.” resistance in Streptococcus sanguis, Streptococcus oralis, and
Streptococcus mitis developed from horizontal transfer of a
MECHANISM OF ACTION OF ␤-LACTAM ANTIBIOTICS PBP2b gene from Streptococcus pneumoniae (107, 348). Methicil-
lin resistance in Staphylococcus spp. is also a significant clinical
␤-Lactam antibiotics exhibit their bactericidal effects by in- challenge. While there are many reasons for this resistance, the
hibiting enzymes involved in cell wall synthesis. The integrity of ␤-lactam resistance phenotype is also conferred by acquisition of
162 DRAWZ AND BONOMO CLIN. MICROBIOL. REV.

FIG. 1. Chemical structures of compounds discussed in the text. Compounds 1 to 7, a representative penicillin (compound 1), an extended-
spectrum cephalosporin (compound 2), a monobactam (compound 3), and carbapenems (compounds 4 to 7). The numbering scheme for
penicillins, cephalosporins, and monobactams is shown. Compounds 8 to 10, ␤-lactamase inhibitors in clinical practice. Compounds 11 to 38,
investigational ␤-lactamase inhibitors: monobactam derivatives (compounds 11 to 14), a penicillin derivative (compound 15), penems (compounds
16 to 20), penam sulfones (compounds 21 to 24), a boronic acid transition state analog (compound 25), non-␤-lactams (compounds 26 to 28), and
metallo-␤-lactamase inhibitors (compounds 29 to 38).

the mecA gene which produces PBP2a (also denoted PBP2⬘) (76). (iii) Decreased expression of outer membrane proteins
PBP2a can assemble new cell wall in the presence of high con- (OMPs) is another mechanism of resistance. In order to access
centration of penicillins and cephalosporins. PBPs on the inner plasma membrane, ␤-lactams must either
VOL. 23, 2010 ␤-LACTAMASE INHIBITORS 163

FIG. 1—Continued.

diffuse through or directly traverse porin channels in the outer (iv) Efflux pumps, as part of either an acquired or intrinsic
membrane of Gram-negative bacterial cell walls. Some Entero- resistance phenotype, are capable of exporting a wide range of
bacteriaceae (e.g., Enterobacter spp., Klebsiella pneumoniae, and substrates from the periplasm to the surrounding environment
Escherichia coli) exhibit resistance to carbapenems based on (347). These pumps are an important determinant of multidrug
loss of these OMPs; the loss of OprD is associated with imi- resistance in many Gram-negative pathogens, particularly P.
penem resistance and reduced susceptibility to meropenem in aeruginosa and Acinetobacter spp. In P. aeruginosa, upregulation
the nonfermenter Pseudomonas aeruginosa (169, 187, 236, 286, of the MexA-MexB-OprD system, in combination with the or-
306, 395). Resistance to imipenem and meropenem has also ganism’s low outer membrane permeability, can contribute to
been associated with the loss of the CarO OMP in clinical decreased susceptibility to penicillins and cephalosporins, as well
isolates of multidrug-resistant Acinetobacter baumannii (278, as quinolones, tetracycline, and chloramphenicol (224–226, 373,
346). Point mutations or insertion sequences in porin-encoding 389). To illustrate, an increase in the carbenicillin MIC from 32
genes can produce proteins with decreased function and thus ␮g/ml to 1,028 ␮g/ml is associated with overproduction of this
lower permeability to ␤-lactams (106). Of note, the disruption efflux pump (12, 225). Additionally, an upregulated efflux pump
of porin proteins alone is not always sufficient for producing (e.g., AdeABC, an RND-type efflux pump, in A. baumannii) can
the resistance phenotype, and typically this mechanism is augment the carbapenem resistance conferred by a catalytically
found in combination with ␤-lactamase expression (106, 235). poor ␤-lactamase (e.g., OXA-23) (162, 333).
164 DRAWZ AND BONOMO CLIN. MICROBIOL. REV.

TABLE 1. ␤-Lactamase classification schemesa


Bush-Jacoby- Inhibited by
Ambler class Preferred substrates Representative enzyme(s)
Medeiros class clavulanate

A (serine penicillinases) 2a Penicillins ⫹ PC1 from S. aureus


2b Penicillins, narrow-spectrum ⫹ TEM-1, TEM-2, SHV-1
cephalosporins
2be Penicillins, narrow-spectrum and extended- ⫹ SHV-2 to SHV-6, TEM-3 to
spectrum cephalosporins TEM-26, CTX-Ms
2br Penicillins ⫺ TEM-30, SHV-72
2c Penicillins, carbenicillin ⫹ PSE-1
2e Extended-spectrum cephalosporins ⫹ FEC-1, CepA
2f Penicillins, cephalosporins, carbapenems ⫾ KPC-2, SME-1, NMC-A

B (metallo-␤-lactamases) 3 Most ␤-lactams, including carbapenems ⫺ IMP-1, VIM-1, CcrA, and BcII
(B1); CphA (B2); L1(B3)

C (cephalosporinases) 1 Cephalosporins ⫺ AmpC, CMY-2, ACT-1

D (oxacillinases) 2d Penicillins, cloxacillin ⫾ OXA-1, OXA-10

Not classified 4
a
Based on data from reference 57. An updated classification system by Bush and Jacoby is in press (56a) at the time of this writing.

␤-LACTAMASES Class A serine ␤-lactamases. In general, class A enzymes are


susceptible to the commercially available ␤-lactamase inhibi-
The first ␤-lactamase enzyme was identified in Bacillus (Esch- tors (clavulanate, tazobactam, and [less so] sulbactam), al-
erichia) coli before the clinical use of penicillin. In a sentinel paper though the K. pneumoniae carbapenemase KPC may be an
published nearly 70 years ago, E. P. Abraham and E. Chain important exception to this generalization (319a). The first
described the B. coli “penicillinase” (1). The enzyme was not plasmid-mediated ␤-lactamase was identified in E. coli in 1963
thought to be clinically relevant, since penicillin was targeted to (and reported in 1965), and was named “TEM” after the pa-
treat staphylococcal and streptococcal infections, and Abraham, tient from whom it was isolated (100). SHV, another common
Chain, and their colleagues were unable to isolate the enzyme ␤-lactamase found primarily in K. pneumoniae, was named
from these Gram-positive organisms (2, 55). It is sobering now to from the term “sulfhydryl reagent variable.” Early studies of
consider the ramifications of this observation. SHV-1 showed that p-chloromercuribenzoate inhibited the hy-
Four years later, Kirby successfully extracted these cell-free drolysis of cephaloridine but not that of benzylpenicillin (251).
“penicillin inactivators” from Staphylococcus aureus, which fore-
TEM and SHV are common ␤-lactamases detected in clinical
shadowed the emergence of a significant clinical problem (202).
isolates of E. coli and K. pneumoniae, pathogens responsible
The growing number of ␤-lactam antibiotics has since increased
for urinary tract, hospital-acquired respiratory tract, and
the selective pressure on bacteria, promoting the survival of or-
bloodstream infections (60, 145, 372). While SHV-1 and
ganisms with multiple ␤-lactamases (249, 256). Currently, more
TEM-1 share 68% sequence homology, the active site of
than 850 ␤-lactamases are identified (K. Bush, personal commu-
SHV-1 is approximately 0.7 to 1.2 Å wider than that of TEM-1,
nication). As authors, we speculate that the rapid replication rate,
which may have important structural implications, especially
recombination rates, and high mutation frequency permit bacte-
related to the substrate profiles of SHV variants (421).
ria to adapt to novel ␤-lactams by evolution of these ␤-lactamases
Although blaTEM and blaSHV may be found on plasmids,
(332).
other class A enzymes are encoded on the chromosome (e.g.,
blaPenA from Burkholderia pseudomallei) or on integrons (e.g.,
Classification blaGES-1 from K. pneumoniae and blaVEB-1 in P. aeruginosa and
A. baumannii) (57, 280).
Two major classification schemes exist for categorizing Class A ESBLs. The growing number of ␤-lactamases in E.
␤-lactamase enzymes: Ambler classes A through D, based on coli and K. pneumoniae, as well as the emergence of these
amino acid sequence homology, and Bush-Jacoby-Medeiros enzymes in other pathogens (e.g., Haemophilus influenzae and
groups 1 through 4, based on substrate and inhibitor profile Neisseria gonorrhoeae), led to the development of extended-
(Table 1) (10, 57). A “family portrait” reveals the structural spectrum cephalosporins with an oxyimino side chain, carbap-
similarity of class A, C, and D serine ␤-lactamases (Fig. 2). enems, cephamycins, and monobactams (71, 115, 188, 321).
Class B ␤-lactamases (“a class apart”) are metallo-␤-lactama- Upon the introduction of the penems and cephems in the early
ses (MBLs) (52). MBLs possess either a single Zn2⫹ ion or a 1980s, these new agents were effective against many ␤-lactam
pair of Zn2⫹ ions coordinated to His/Cys/Asp residues in the resistant bacteria. However, selective pressure quickly fostered
active site. In this review, the Ambler classification scheme will be the emergence of extended-spectrum ␤-lactamases (ESBLs),
used. which could hydrolyze many of the oxyimino-cephalosporins
VOL. 23, 2010 ␤-LACTAMASE INHIBITORS 165

FIG. 2. “Family portrait” of ␤-lactamase enzymes. (A) Class A SHV-1; (B) class B IMP-1; (C) class C E. coli AmpC; (D) class D OXA-1. For
classes A, C, and D, the active-site serine is shown in yellow; for class B, the two Zn2⫹ ions are shown. (Based on PDB entries 1SHV, 1DDK, 2BLS,
and 1M6K, respectively.)

(186, 188). VEB, and PER enzymes, and sometimes these enzymes do not
In a dramatic parallel to the observations of Abraham and belong to any defined family (280).
Chain, within 2 years of the introduction of the extended- Class A serine carbapenemases. Class A serine carbapen-
spectrum cephalosporins cefotaxime and ceftazidime, novel emases include the nonmetallocarbapenamase of class A
ESBLs in E. coli and K. pneumoniae were reported (204). (NMC-A), IMI, SME, and KPC. Members of this group of
Interestingly, these “new ␤-lactamases” harbored point muta- ␤-lactamases can hydrolyze carbapenems as well as cephalo-
tions in the parent blaTEM-1 and blaSHV-1 genes that led to sporins, penicillins, and aztreonam (356). These carbapenem-
single amino acid changes in the ␤-lactamases. Other ESBLs, hydrolyzing enzymes have been identified primarily in Entero-
such as CTX-M, arose by plasmid transfer from preexisting bacter cloacae, Serratia marcescens, and K. pneumoniae,
chromosomal ESBL genes from Kluyvera spp., which typically bacteria which often harbor multiple resistance determinants,
are nonpathogenic commensal organisms (14, 32). CTX-M narrowing the range of treatment options (290, 291, 359, 454,
ESBLs now represent important enzymes found in isolates 455). The bla gene for the former two organisms is typically found
from the community and are the most commonly isolated on the chromosome, while the K. pneumoniae carbapenemase
ESBLs in many parts of the world, particularly Europe (343). blaKPC gene is carried on plasmids containing Tn4401 (292).
ESBLs hydrolyze penicillins, narrow- and extended-spec- MICs of carbapenems in carbapenemase-expressing strains can
trum cephalosporins (including the anti-methicillin-resistant S. vary from moderately increased (2 to 4 ␮g/ml) to resistant (ⱖ 32
aureus [MRSA] cephalosporin ceftobiprole), and the mono- ␮g/ml) (116b, 356).
bactam aztreonam (11, 321, 357). In contrast, ESBLs cannot Class B metallo-␤-lactamases. Class B enzymes are Zn2⫹-
efficiently degrade cephamycins, carbapenems, and ␤-lacta- dependent ␤-lactamases that demonstrate a hydrolytic mech-
mase inhibitors. Since their initial description, more than 200 anism different from that of the serine ␤-lactamases of classes
different ESBLs have been identified, posing a significant risk A, C, and D (57). Organisms producing these enzymes usually
to public health and to hospitalized patients in intensive care exhibit resistance to penicillins, cephalosporins, carbapenems,
units, where infection with an ESBL may lead to significant and the clinically available ␤-lactamase inhibitors (432). Inter-
morbidity and mortality (up-to-date listings of verified ESBL estingly, the hydrolytic profile of MBLs does not typically in-
sequences are available on the website www.lahey.org/studies clude aztreonam. MBLs likely evolved separately from the
/webt.asp maintained by G. A. Jacoby and K. Bush) (321). The other Ambler classes, which have serine at their active site
majority of ESBLs are from the SHV, TEM, and CTX-M (249). The blaMBL genes are located on the chromosome, plas-
families; less frequently they are derived from BES, GES-1, mid, and integrons (133, 432). P. aeruginosa, K. pneumoniae,
166 DRAWZ AND BONOMO CLIN. MICROBIOL. REV.

and A. baumannii produce class B enzymes encoded by mobile ␤-Lactamase Hydrolytic Mechanisms
genetic elements (93, 175, 177). In contrast, Bacillus spp.,
Serine ␤-lactamases acylate ␤-lactam antibiotics, much like
Chryseobacterium spp., and Stenotrophomonas maltophilia pos-
PBPs, and then use strategically positioned water molecules to
sess chromosomally encoded MBLs, but the majority of these
hydrolyze the acylated ␤-lactam (265). In this manner, the
host pathogens are not frequently responsible for serious in-
␤-lactamase is regenerated and can inactivate additional ␤-lac-
fections (432). The role that these ␤-lactamases in Bacillus spp.
tam molecules. This enzymatic reaction may be represented by
and Chryseobacterium spp. will play in the clinical arena is still the following equation:
unknown.
Class C serine cephalosporinases. Class C AmpC ␤-lacta- k1 k2 k 3, H 2O
mases include CMY-2, P99, ACT-1, and DHA-1, which are E ⫹ S| ¡ E⫺S O
L; E:S O ¡E ⫹ P (1)
usually encoded by bla genes located on the bacterial chromo- k⫺1
some, although plasmid-borne AmpC enzymes are becoming
In this scheme, E is a ␤-lactamase, S is a ␤-lactam substrate,
more prevalent (340). Organisms expressing the AmpC ␤-lac-
E:S is the Michaelis complex, E ⫺ S is the acyl-enzyme, and P
tamase are typically resistant to penicillins, ␤-lactam–␤-lacta-
is the product devoid of antibacterial activity. The rate con-
mase inhibitor combinations, and cephalosporins, including
stants for each step are represented by k1, k⫺1, k2, and k3. k1
cefoxitin, cefotetan, ceftriaxone, and cefotaxime. AmpC en- and k⫺1 are the association and dissociation rate constants for
zymes poorly hydrolyze cefepime and are inhibited by cloxacil- the preacylation complex, respectively; k2 is the acylation rate
lin, oxacillin, and aztreonam (57). Members of the Enterobac- constant, and k3 is the deacylation rate constant. More com-
teriaceae family, such as Enterobacter spp. and Citrobacter spp., plicated branched reaction mechanisms are seen with some
are AmpC ␤-lactamase producers that resist inhibition by cla- ␤-lactamases (129). In order to be uniform, we now define
vulanate and sulbactam, although Klebsiella spp., Salmonella basic terms that are often used to describe the kinetic behavior
spp., and Proteus spp. normally do not harbor chromosomal of ␤-lactamases.
blaAmpC genes (19, 184). The Michaelis constant, Km, is defined as (129)
Production of chromosomal AmpCs in Gram-negative bac-
teria is at a low level (“repressed”) but can be “derepressed” by Km ⫽ k3Ks/共k2 ⫹ k3兲 (2)
induction with certain ␤-lactams, particularly cefoxitin (14, 24, where the kinetic constant, Ks, is (k⫺1 ⫹ k2)/k1. The turnover
148). The mechanism of this regulation has been the subject of number, kcat, is a composite rate constant that represents mul-
intense investigation (183). Of significant concern is the selec- tiple chemical steps and is defined as (92)
tion of mutant bacterial populations that are genetically dere-
pressed for AmpC production, which can cause a dramatic kcat ⫽ k2k3/共k2 ⫹ k3兲 (3)
increase in MICs during the course of ␤-lactam therapy (e.g.,
or
after 14 days of ceftazidime therapy, a strain of P. aeruginosa
with MICs increased from 1 to 32 ␮g/ml was selected) (184, Vmax ⫽ kcat关E兴 (4)
193, 233).
Class D serine oxacillinases. Class D ␤-lactamases were Km, expressed in terms of concentration, represents the rela-
initially categorized as “oxacillinases” because of their ability tive affinity of the ES encounter and the rate at which the ES
to hydrolyze oxacillin at a rate of at least 50% of that of is converted to P; a large Km value represents poor affinity
benzylpenicillin, in contrast to the relatively slow hydrolysis of (large Ks).
oxacillin by classes A and C (98). In bacteria, OXA ␤-lacta- Class A. In Fig. 3, we represent the reaction scheme of a
mases can also confer resistance to penicillins, cephalosporins, typical class A ␤-lactamase. In brief, (i) after formation of the
extended-spectrum cephalosporins (OXA-type ESBLs), and Henri-Michaelis complex, the active-site serine performs a nu-
cleophilic attack on the carbonyl of the ␤-lactam antibiotic that
carbapenems (OXA-type carbapenemases). Generally speak-
results in a high-energy tetrahedral acylation intermediate
ing, OXA enzymes are resistant to inhibition by clavulanate,
(‡1); (ii) this intermediate transitions into a lower-energy co-
sulbactam, and tazobactam, (with some exceptions; e.g.,
valent acyl-enzyme following protonation of the ␤-lactam ni-
OXA-2 and OXA-32 are inhibited by tazobactam but not sul-
trogen and cleavage of the C-N bond; (iii) next, an activated
bactam and clavulanate, and OXA-53 is inhibited by clavu-
water molecule attacks the covalent complex and leads to a
lanate) (98, 276, 279, 345). Interestingly, sodium chloride at high-energy tetrahedral deacylation intermediate (‡2); and (iv)
concentrations of ⬎50 to 75 mM inhibits some carbapenem- hydrolysis of the bond between the ␤-lactam carbonyl and the
hydrolyzing oxacillinases (e.g., OXA-25 and OXA-26) (6). oxygen of the nucleophilic serine regenerates the active en-
Site-directed mutagenesis studies suggest that susceptibility to zyme and releases the inactive ␤-lactam. Acylation and deacy-
inhibition by sodium chloride is related to the presence of a lation require the activation of the nucleophilic serine and
Tyr residue at position 144 (161, 279). Presumably, Tyr144 may hydrolytic water, respectively. An entire body of experimenta-
facilitate sodium chloride binding better than the Phe residue tion is devoted to examining the mechanistic roles of the indi-
found in resistant oxacillinases, although the molecular mech- vidual residues in the active sites of ␤-lactamases; here we
anism remains unexplained. Examples of OXA enzymes in- present only several leading insights to provide a foundation
clude those rapidly emerging in A. baumannii (e.g., OXA-23 for the discussion of inhibitory pathways.
and OXA-24/40) and constitutively expressed in P. aeruginosa For class A enzymes, a leading theory holds that Glu166 acts
(e.g., OXA-50) (141a, 346, 435). as the activating base of the hydrolytic water in deacylation
VOL. 23, 2010 ␤-LACTAMASE INHIBITORS 167

FIG. 3. Proposed reaction mechanism for a penicillin ␤-lactam substrate and a class A serine ␤-lactamase enzyme in which Glu166 participates
in activating a water molecule for both acylation and deacylation (additional evidence exists to suggest that this acylation scheme may exist in
competition with a mechanism where Lys73 acts as the general base to activate Ser70) (261). Dashed lines represent hydrogen bonds, and the
number labels are included to help the reader appreciate the general order of events, not as absolute and discrete steps. Following activation of
the hydroxyl group, Ser70 performs a nucleophilic attack on the carbonyl group of the ␤-lactam antibiotic, resulting in a high-energy acylation
intermediate. Protonation of the ␤-lactam nitrogen leads to cleavage of the C-N bond and formation of the covalent acyl-enzyme, which adopts
a lower energy state. Attack by a catalytic water leads to a high-energy deacylation intermediate, with subsequent hydrolysis of the bond between
the ␤-lactam carbonyl and the oxygen of Ser70. Deacylation regenerates the active enzyme and releases the inactive ␤-lactam.

(166, 392). In contrast, the acylation mechanism is less clear, clude four polar regions: (i) the active-site pocket Ser70-Xaa-
and several hypotheses and lines of evidence exist. The first Xaa-Lys73, (ii) the “⍀-loop” residues 163 to 178, (iii) Ser130-
notion proposes that Glu166, via a water molecule, deproto- Asp131-Asn132, and (iv) Lys234-Thr(Ser)235-Gly236. The
nates the Ser70 hydroxyl before addition to the ␤-lactam bond. roles of residues in the “SDN” loop comprised of Ser130-
This Glu166 general base theory is supported by quantum Asp131-Asn132 are related to maintaining the structure of the
mechanical/molecular mechanical (QM/MM) calculations and active site cavity, enzyme stability, and stabilization of the
the protonated status of Glu166 in the ultra-high-resolution enzyme transition state, respectively (182, 305). The functional
(0.85-Å) structure of TEM-1 in complex with an acylation explanation for the conservation of the Lys234-Thr(Ser)235-
transition state analog (TSA) (Research Collaboratory for Gly236 triad is less well understood. Removal of the hydroxyl
Structural Bioinformatics Protein Data Bank [PDB] entry group from the middle residue has little impact on penicillin
1M40) (163, 265). Surprisingly, substitutions at residue 166 do hydrolysis in TEM (but more on the catalytic efficiency of
not prevent acylation (139, 146, 207, 392). cephalosporins), and despite what may be expected from crys-
A second view of acylation maintains that Lys73 can serve as tal structure alignments, the study of modified ␤-lactams ar-
the general base deprotonating Ser70 (166, 261, 401). Recent gues against an interaction between the ␤-lactam carboxylate
QM/MM studies by Meroueh et al. demonstrate that the path- and the Thr(Ser) hydroxyl group (112, 181). In SHV, the
way involving Lys73 as the activating base is energetically fa- Thr235Ala substitution lowers MICs of both penicillins and
vorable and may exist in competition with the pathway where cephalosporins (174). Interestingly, the expression of the struc-
Glu166 serves as the activating residue (261). Additional ideas turally conserved SHV Thr235Ser variant has no effect on
on the mechanism of acylation have been proposed, including ␤-lactam MICs but does lead to an increased susceptibility to
both (i) a role for the C-3 or C-4 substrate carboxylate and the piperacillin-tazobactam combination (174).
Ser130 in activating Ser70 and (ii) a general acid pathway The most common mechanism for transformation of a
where protonation of the ␤-lactam nitrogen is the primary broad-spectrum ␤-lactamase to an ESBL is point mutations
event (13, 103). Further clarification of the acylation mecha- that result in amino acid sequence changes near the enzyme
nism may prove useful for the design of inhibitors that capi- active site, facilitating hydrolysis of oxyimino-cephalosporins
talize on the enzymes’ native machinery. (186, 341). However, the specific amino acid replacements and
Highly conserved structural motifs in class A enzymes in- resistance mechanisms vary between enzymes (124). In TEM,
168 DRAWZ AND BONOMO CLIN. MICROBIOL. REV.

changes at Ambler positions Arg164 (-His, -Ser), Gly238 (-Ser,


-Ala), and Glu240 (-Lys) result in variants that confer the
ESBL phenotype (40; www.lahey.org/studies/webt.asp). Sev-
eral crystallographic studies demonstrated that the “new”
TEM active site is expanded or remodeled, compared to
TEM-1, to accommodate the larger side chain of expanded-
spectrum cephalosporins (83, 206, 298, 304). The crystal struc-
ture of SHV-2 (Gly238Ser) at 0.91 Å resolution (PDB 1N9B),
FIG. 4. Schematic representation of the Zn2⫹-binding site of
compared to SHV-1, showed a displacement in the b3 ␤-strand dinuclear subclass B1 metallo-␤-lactamases such as B. cereus BcII.
containing residues 238 to 242 which created an expanded
␤-lactam binding site but preserved the positioning of the es-
sential catalytic residues (124, 298). Similarly, the PER-1
ESBL active site is expanded by a novel fold of the ⍀-loop and that deprotonates the catalytic water for hydrolysis (84). In-
the insertion of four residues after Lys240 (417). stead, deacylation may proceed by “substrate-activated cataly-
In contrast, the crystal structure of the acyl-enzyme inter- sis” whereby the nitrogen on the ␤-lactam acts as a base and
mediate of the ESBL Toho-1 in complex with cefotaxime did transiently accepts the proton from the hydrolytic water, while
not resemble the enlarged active sites seen with the ESBL the Tyr150 proton helps stabilize the water’s developing neg-
TEM and SHV ␤-lactamases. Rather, the Toho-1 enzyme fa- ative charge (51, 84). The crystallographic evidence and mu-
cilitates cephalosporin binding through movement of the tagenesis studies do not rule out a role of Lys67 in the coor-
⍀-loop toward the active site, interactions between conserved dinate base mechanism, and further studies exploring the role
residues Asn104 and Asp240 and the bulky side chain of cefo- of Lys67 are anticipated (84, 271).
taxime, and contacts with Ser237 that help position the ␤-lac- Class D. Our understanding of the hydrolytic mechanism of
tam carbonyl in the oxyanion hole (176, 382). The high-reso- class D ␤-lactamases is based on the careful study of OXA-10,
lution crystal structures of four additional CTX-M -13, and -1 (253, 310, 334, 379, 398, 399). This class of enzymes
␤-lactamases confirmed that the active sites of these enzymes is unique because of the direct role of carboxylation of the
were not enlarged compared to those of the narrow-spectrum active-site Lys70. The carbamic acid on Lys70 can ionize to
TEM-1 and SHV-1 (83, 85). Instead, the structural basis for yield a carbamate that hydrogen bonds with the nucleophilic
the extended substrate profiles in the CTX-M enzyme family Ser67 residue. In this manner, the carboxylated Lys70 may
appears to be specific amino acid (i.e., Ser237 and Asp104) serve as the general base by activating both Ser67 for acylation
interactions with the oxyimino-cephalosporins and increased and the hydrolytic water for deacylation (144, 252, 253).
mobility of the b3 ␤-strand (83). Overall, this increased mobil- Class B. In contrast to the serine ␤-lactamases described
ity and activity “costs the enzyme” thermal stability, a theme above, class B includes Zn2⫹-dependent enzymes that follow a
recapitulated by the evolution of many resistance phenotypes different hydrolytic mechanism. In general, MBLs use the -OH
(383). group from a water molecule that is coordinated by Zn2⫹ to
Point mutations are not the only mechanism for the ESBL hydrolyze the amide bond of a ␤-lactam. MBLs are divided
phenotype; other alterations in enzyme regulation produce into three classes based on their Zn2⫹ dependency, i.e.,
resistance, such as promoter sequence changes leading to in- whether they (i) are fully active with either one or two ions
creased enzyme expression or alterations in outer membrane (subclass B1, e.g., IMP-1, VIM-2, BcII, and CcrA), (ii) require
porins (293, 302). two ions (subclass B3, e.g., L1), or (iii) employ one ion and are
At this time, the biochemical basis for class A serine carbapen- inhibited by binding of an additional ion (subclass B2, e.g.,
emase activity is an area of active investigation. Current evidence CphA) (130, 164, 165, 325, 326). Crowder and colleagues com-
suggests that this property rests upon a remodeling of the active site. piled the recent structural and mechanistic data and summa-
Unlike class A ESBLs (with expanded or enhanced mobility of the rized the common features for the three subclasses of MBLs
b3 ␤-strand), the active sites of class A carbapenemases such as (96). MBLs utilizing two Zn2⫹ ions for hydrolysis, such as the
KPC-2, NMC-A, and SME-1 reveal that there are multiple alter- subclass B3 S. maltophilia L1, coordinate the ␤-lactam sub-
ations in the spatial positions of catalytic residues (200, 338, 388, 402). strate by the carboxylate and carbonyl groups, bridged by a
We are just beginning to understand how this remodeling allows hydroxide ion. After substrate binding, one of the Zn2⫹ ions, in
enhanced carbapenemase activity compared to that of other class A conjunction with enzyme residues, polarizes the ␤-lactam car-
enzymes. bonyl for attack by the -OH group, which is hydrogen bonded
Class C. The prevailing evidence regarding the class C hy- to deprotonated Asp120. Nucleophilic attack by the -OH
drolytic mechanism suggests that Tyr150 behaves as a general group creates a tetrahedral species which rapidly collapses into
base by increasing the nucleophilicity of Ser64 for acylation an intermediate in which the ␤-lactam nitrogen is anionic.
(299). Early studies of the deacylation mechanism indicated Protonation of the nitrogen leads to product formation. The
that Tyr150 also acts as the catalytic base, accepting a proton source of the proton is not certain, and it may come from
from the deacylating water (111, 299). For example, QM/MM Asp120 or a water molecule.
calculations suggested that Tyr150 interacts with Lys67 in a The B1 enzyme Bacillus cereus BcII is active in both its
conjugate base manner (136). However, the crystal structure of mononuclear and dinuclear forms, and in the resting state, the
the E. coli AmpC in complex with a deacylation transition state Zn2⫹-bound -OH group is hydrogen bonded to the
analog revealed that Tyr150 remains protonated throughout deprotonated Asp120 as well as several other active-site resi-
the reaction and therefore is unlikely to be the anionic base dues (Fig. 4) (96, 440). After attack by this -OH group, the
VOL. 23, 2010 ␤-LACTAMASE INHIBITORS 169

breakdown of the tetrahedral intermediate requires protona- ␤-Lactamase Inhibitors in Clinical Practice
tion of the ␤-lactam nitrogen; the source of this proton is under
Clavulanic acid, sulbactam, and tazobactam. Clavulanic
investigation. In the case of the CphA B2 MBL from Aeromo-
acid, the first ␤-lactamase inhibitor introduced into clinical
nas hydrophila, a second bound Zn2⫹ ion is inhibitory. The
medicine, was isolated from Streptomyces clavuligerus in the
proposed mechanism includes a water molecule activated by
1970s, more than 3 decades ago (360). Clavulanate (the salt
either His118 or Asp120, rather than a Zn2⫹-bound -OH
form of the acid in solution) showed little antimicrobial
group; the singular Zn2⫹ appears to help coordinate the ␤-lac-
tam nitrogen (132, 451). activity alone, but when combined with amoxicillin, clavu-
lanate significantly lowered the amoxicillin MICs against S.
aureus, K. pneumoniae, Proteus mirabilis, and E. coli (47).
CIRCUMVENTING ␤-LACTAMASES Sulbactam and tazobactam are penicillinate sulfones that
were later developed by the pharmaceutical industry as syn-
␤-Lactamase Inhibitors: Mechanistic Considerations thetic compounds in 1978 and 1980, respectively (117, 121).
All three ␤-lactamase inhibitor compounds share struc-
A successful strategy for combating ␤-lactamase-medi-
ated resistance is the use of agents designed to bind at the tural similarity with penicillin; are effective against many
active site, which are frequently ␤-lactams. This strategy can susceptible organisms expressing class A ␤-lactamases (in-
take two forms: (i) create substrates that reversibly and/or cluding CTX-M and the ESBL derivatives of TEM-1,
irreversibly bind the enzyme with high affinity but form TEM-2, and SHV-1); and are generally less effective against
unfavorable steric interactions as the acyl-enzyme or (ii) class B, C, and D ␤-lactamases (53, 60, 67, 341). The activity
develop mechanism-based or irreversible “suicide inhibi- of an inhibitor can be evaluated by the turnover number (tn)
tors” (53). Examples of the former are extended-spectrum (also equivalent to the partition ratio [kcat/kinact]), defined as
cephalosporins, monobactams, or carbapenems which form the number of inhibitor molecules that are hydrolyzed per
acyl-enzymes and adopt catalytically incompetent conforma- unit time before one enzyme molecule is irreversibly inac-
tions that are poorly hydrolyzed (Fig. 1, compounds 2, 3, and tivated (58). For example, S. aureus PC1 requires one clav-
4 to 7, respectively). For reversible inhibition, the reaction ulanate molecule to inactivate one ␤-lactamase enzyme,
can be described as was shown above in equation 1, where S while TEM-1 needs 160 clavulanate molecules, SHV-1 re-
represents the (very) slowly hydrolyzed substrate. An equi- quires 60, and B. cereus I requires more than 16,000 (53, 69,
librium constant, Ki, can be calculated from the pre-steady- 113, 123, 411). For comparison, sulbactam tns are 10,000 and
state rate constants, k⫺1/k1, and yields an estimate of affinity. 13,000 for TEM-1 and SHV-1, respectively (180, 410).
Irreversible “suicide inhibitors” can permanently inactivate The low KIs of the inhibitors for class A ␤-lactamases (nM to
the ␤-lactamase through secondary chemical reactions in the ␮M), the ability to occupy the active site “longer” than ␤-lactams
enzyme active site. Equation 5 represents a general mechanism (high acylation and low deacylation rates), and the failure to be
of irreversible inhibitors (I) leading to permanent enzyme in- hydrolyzed efficiently are integral to their efficacy (158). Clavu-
activation (E ⫺ I*): lanate, sulbactam, and tazobactam differ from ␤-lactam antibiot-
ics as they possess a leaving group at position C-1 of the five-
k1 k2 k3 membered ring (sulbactam and tazobactam are sulfones, while
E⫹I | ¡ E⫺I O
L; E:I O ¡ E ⫺ I* (5) clavulanate has an enol ether oxygen at this position). The better
k⫺1 leaving group allows for secondary ring opening and ␤-lactamase
enzyme modification. Compared to clavulanate, the unmodified
Examples of these inactivators are the commercially avail- sulfone in sulbactam is a relatively poor leaving group, a property
able class A inhibitors clavulanic acid, sulbactam, and reflected in the high partition ratios for this inhibitor (e.g., for
tazobactam (Fig. 1, compounds 8 to 10). As will be described TEM-1, sulbactam tn ⫽ 10,000 and clavulanate tn ⫽ 160) (179,
below, these types of inactivators often display additional 180). Tazobactam possesses a triazole group at the C-2 ␤-methyl
pathways to inhibition. Irreversible inhibitors can be char- position. This modification leads to tazobactam’s improved IC50s,
acterized by first-order rate constants for inhibition (kinact, partition ratios, and lowered MICs for representative class A and
the rate of inactivation achieved with an “infinite” concen- C ␤-lactamases (36, 58, 60).
tration of inactivator) and KI values (the concentration of The efficacy of the mechanism-based inhibitors can vary
inactivator which yields an inactivation rate that is half the within and between the classes of ␤-lactamases (Table 2).
value of kinact) (54, 92). While KI closely approximates the For class A, SHV-1 is more resistant to inactivation by
meaning of Km for enzyme substrates, depending on the sulbactam than TEM-1 but more susceptible to inactivation
individual rate constants comprising the reaction, the KI by clavulanate (328). Comparative studies of TEM- and
may or may not equal the equilibrium constant Ki (⫽ k⫺1/k1) SHV-derived enzymes, including ESBLs, found that the
determined under pre-steady-state conditions. IC50s for clavulanate were 60- and 580-fold lower than those
The 50% inhibitory concentration (IC50) measures the for sulbactam against TEM-1 and SHV-1, respectively (328).
amount of inhibitor required to decrease enzyme activity to In our opinion, the explanations for these differences in
50% of its uninhibited velocity. While an IC50 can reflect an inactivation chemistry are likely subtle, yet highly important,
inhibitor’s affinity or kcat/kinact ratio, these parameters are not differences in the enzyme active sites. For example, Thom-
always congruent; e.g., an inhibitor can have a very poor “af- son et al. compared the atomic structure models of TEM-1
finity” and acylate the enzyme slowly but still yield a low IC50 and SHV-1 and found that the distance between Val216 and
because of very low deacylation rates. Arg244, residues responsible for positioning of the water
170 DRAWZ AND BONOMO CLIN. MICROBIOL. REV.

TABLE 2. Kinetic properties of representative ␤-lactamasesa


Clavulanate Sulbactam Tazobactam
Ambler
␤-Lactamase IC50 KI IC50 KI IC50 Reference(s)
class KI (␮M) tn tn tn
(nM) (␮M) (nM) (␮M) (nM)

TEM-1 A 0.1 60 160 1.6 900 10,000 0.01 97 140 73, 180, 283
SHV-1 A 1 12 60 8.6 12,000 13,000 0.07 150 5 58, 153, 410, 411
SHV-5 A 20 1,800 80 137
PC1 A 30 1 80 27 1 47, 58, 69
CTX-M-2 A 200 2,100 20 18
CcrA B ⬎500,000 ⬎500,000 ⬎500,000 400,000 4,000 58
P99 C ⬎100,000 ⬎500,000 5,600 8.5 50 58
CMY-2 C 4,365 101 50 116a
OXA-1 D 1,800 4,700 380 1400 196, 328
OXA-2 D 1,400 0.1 140 10 211, 328
a
The definition of KI may not be uniform from laboratory to laboratory. See the cited references for the methods used in each determination.

molecule important in the inactivation mechanism of clavu- suggests that clavulanate, sulbactam, and tazobactam follow
lanate, was more than 2 Å greater in SHV-1 than in TEM-1 similar reaction pathways, beginning with formation of an acyl-
(411). This increased distance may be too great for coordi- enzyme species (48, 81, 307, 308). After acylation, opening of
nation of a water molecule, suggesting that the strategic the five-membered ring leads to formation of a transient imine
water is positioned elsewhere in SHV-1 and may be re- intermediate. This imine species is likely the common inter-
cruited into the active site with acylation of the substrate or mediate preceding the chemical conversions that lead to tran-
inhibitor. This variation underscores the notion that mech- sient enzyme inhibition (44, 179, 197). Raman spectroscopy of
anism-based inhibitors may undergo different inactivation SHV/inhibitor crystals show that the imine species then rear-
chemistry even in highly similar enzymes (410, 411). ranges to form enamine intermediates (197, 307). This en-
Mechanism of inhibition. Evidence from X-ray crystallog- amine intermediate, in either the trans or cis conformation,
raphy, UV difference spectra, isoelectric focusing, mass represents a second important intermediate in the inactivation
spectrometry (MS), and Raman microscopy suggests that mechanism (69, 81, 368). Depending on the properties of the
the inactivators of class A ␤-lactamases undergo complex enzyme and inhibitor, the reaction will ultimately proceed to
reaction schemes with multiple branch points after forma- deacylation or irreversible (prolonged) inactivation. In the case
tion of the acyl-enzyme (45, 78, 79, 157, 309, 396). As rep- of deacylation of the enamine intermediate, the acyl-enzyme
resented in equations 5 and 6, the acyl-enzyme intermediate undergoes decarboxylation and ester bond hydrolysis, regen-
can (i) undergo a reversible change that generates a tran- erating the active ␤-lactamase, albeit very slowly (260).
siently inhibited enzyme, a tautomer (E ⫺ T); (ii) lead to The duration of transient inhibition is determined, in part,
permanent inactivation as a covalent acyl-enzyme species by the stability of the intermediate species. Preceding acylation
(E ⫺ I*); or (iii) regenerate the active enzyme via hydrolysis of the inhibitor, a persistent noncovalent Michaelis complex
(E ⫹ P): may account for inhibition of enzyme activity (196). Following
formation of the acyl-enzyme, stabilization of the enamine
E⫺T
intermediate is a significant factor in prolonged enzyme inhi-
bition. Crystals of tazobactam in complex with the SHV
k4
/ k⫺4 Glu166Ala variant (PDB 1RCJ) showed that tazobactam
formed stoichiometric amounts of the trans-enamine (308).
The trans-enamine intermediate of tazobactam, as opposed to
k5
the trans-enamine intermediates of clavulanate and sulbactam,
E⫺I 3 E ⫹ P (6) may be stabilized by intra- and intermolecular interactions.
These interactions between the sulfone and triazolyl moieties
k3
2 of the tazobactam intermediate and the enzyme active site may
explain, in part, tazobactam’s potent in vitro and in vivo inhi-
bition of many serine ␤-lactamases (36, 60, 308, 312).
Data from Raman crystallography also support the hypoth-
E ⫺ I*
esis that compared to clavulanate and sulbactam, tazobactam
more readily forms the trans-enamine intermediate (197). In
The functional inhibition of the enzyme is determined by the this case, Raman spectroscopy allows for the identification of
relative rates (k3, k4, k⫺4, and k5) of each of these pathways reaction intermediates and calculation of their rates of decay
and in particular by the formation of the E ⫺ I* species (53). and accumulation by examining single crystals in solution.
Figure 5 shows a more detailed mechanism describing clav- Studies of SHV-1 in complex with each of the mechanism-
ulanate inhibition of PC1, TEM-1, or SHV-1 (78, 79, 81, 122, based inhibitors revealed that tazobactam forms a predomi-
179, 180). Kinetic and mass spectrometry analysis of inactiva- nant population of trans-enamine, as opposed to clavulanate
tion mechanisms, combined with crystallographic studies, and sulbactam, which form a mixture of trans-enamine and the
VOL. 23, 2010 ␤-LACTAMASE INHIBITORS 171

FIG. 5. Proposed mechanism of inhibition of class A ␤-lactamases by clavulanate, showing the different acyl-enzyme fragmentation products
(expressed in daltons), that have been experimentally observed. (Based on data from references 48, 108, 197, 411, and 453.)

more chemically labile cis-enamine and imine species (197). pact on susceptibility results, which may or may not reflect
Analysis of the inactivation reactions of PC1, TEM, and clinical efficacy. We present to the reader the following exam-
SHV ␤-lactamases using mass spectrometry has identified a ple. The ticarcillin-clavulanate preparation for parental admin-
series of intermediates beyond the acyl-enzyme, imine, and istration is 3.0 g-0.1 g, while ampicillin-sulbactam is at a 2.0
enamine (48, 81, 311, 396, 411). The terminal end products of g-1.0 g ratio. However, both combinations are tested at a 2:1
inhibition with these mechanism-based inhibitors suggest that ratio in vitro, and since clavulanate has higher “gram-for-gram”
a covalent modification of the active-site Ser70 persists potency than sulbactam, clavulanate may “test better” in MIC
through the inhibition process. Mass adducts (in daltons) cor- profiling. However, clinically ampicillin-sulbactam may retain
respond to different inactivated enzyme species; e.g., (i) Ser70- efficacy because of the relatively large amount of inhibitor
Ser130 cross-linked enzyme or propynyl enzyme (⌬ ⫹ 52 ⫾ 3), delivered relative to the ␤-lactam and relative to the amount of
(ii) aldehyde (⌬ ⫹ 70 ⫾ 3), (iii) hydrated aldehyde (⌬ ⫹ 88 ⫾ ␤-lactamase in the cell. This scenario illustrates an important
3), and (iv) decarboxylated trans-enamine (⌬ ⫹ 156 ⫾ 3) are issue in susceptibility testing and its correlate to clinical effi-
some of the products (Fig. 5). cacy.
␤-Lactam–␤-lactamase inhibitor combinations: clinical use. (i) Amoxicillin-clavulanate. Amoxicillin-clavulanate was the
Generally, the inhibitors do not inactivate PBPs, but notable first ␤-lactam–␤-lactamase inhibitor combination introduced
exceptions include (i) the intrinsic activities of sulbactam into clinical practice, in 1981 in the United Kingdom and in
against Bacteroides spp., Acinetobacter spp., and N. gonor- 1984 in the United States, and remains the only combination
rhoeae; (ii) clavulanate action against Haemophilus influenzae available for oral use. The activity of amoxicillin against sus-
and N. gonorrhoeae; and (iii) tazobactam inhibition of PBPs in ceptible bacteria that do not possess a ␤-lactamase (e. g.,
Borrelia burgdorferi (53, 167, 222, 264, 285, 423, 424, 447). As streptococci, enterococci, E. coli, and Listeria spp.) is not im-
these “antibacterial effects” are relatively weak, the inhibitors proved by the use of clavulanate. However, the addition of
are always combined with ␤-lactam antibiotics for clinical use. clavulanate significantly expands amoxicillin’s spectrum to in-
Currently, there are five ␤-lactam–␤-lactamase inhibitor for- clude penicillinase-producing S. aureus, H. influenzae, Morax-
mulations available. Amoxicillin-clavulanate, ticarcillin-clavu- ella catarrhalis, Bacteroides spp., N. gonorrhoeae, E. coli, Kleb-
lanate, ampicillin-sulbactam, and piperacillin-tazobactam are siella spp., and P. mirabilis (59, 387). The oral availability of
available in the United States. Cefoperazone-sulbactam is used amoxicillin-clavulanate makes it well suited for the outpatient
in several European countries, Japan, and India but is not clinic, and this ␤-lactam–␤-lactamase inhibitor combination
available in the United States, and therefore we will not in- exerted its greatest impact on the treatment of community-
clude this combination in this review. Table 3 summarizes acquired respiratory infections (59). Additionally, amoxicillin-
selected clinical features of these available combinations. clavulanate is sometimes used as an oral equivalent (“step-
Importantly, as in clinical usage, the ␤-lactamase inhibitors down therapy”) of ampicillin-sulbactam or ticarcillin-
are combined with ␤-lactams for in vitro susceptibility testing. clavulanate in the treatment of skin, soft tissue, abdominal, and
The composition of these formulations can have a major im- gynecological infections. Intravenous formulations of amoxicil-
172 DRAWZ AND BONOMO CLIN. MICROBIOL. REV.

TABLE 3. Selected clinical features of available ␤-lactam–␤-lactamase inhibitor combinations


␤-Lactam–␤- Route of
Proven clinical efficacy for
lactamase Trade name; administration, t1/2, h (% Mean peak serum Approved clinical
susceptible causative
inhibitor manufacturer formulations, and protein bound) levels (␮g/ml) indications
organism(s):
composition dosing intervalsa

Piperacillin- Zosyn; Wyeth i.v., 2.0 g/0.25 g q6h, Piperacillin and 1 h after 3.375-g dose: Appendicitis E. coli, Bacteroides spp.
tazobactam 3.0 g/0.375 g q6h, 4.0 tazobactam, piperacillin, 106; Skin and soft tissue S. aureus
g/0.5 g q6h 0.7–1.2 (30) tazobactam 10.7 infections, including
diabetic foot infections
Postpartum endometritis E. coli
or pelvic inflammatory
disease
Community-acquired H. influenzae
pneumonia
Hospital-acquired, S. aureus, A. baumannii,
ventilator-acquired H. influenzae, K.
pneumonia pneumoniae, P.
aeruginosab

Ampicillin- Unasyn; Pfizer i.v./i.m., 1.0 g/0.5 g q6h, Ampicillin, 1.0 After 15-min infusion Skin and skin structure S. aureus, E. coli,
sulbactam 2.0 g/1.0 g q6h (28); of 1-g/0.5-g dose: infections Klebsiella spp., P.
sulbactam, ampicillin, 40-71; mirabilis, B. fragilis,
1.0 (38) sulbactam, 21-40 Enterobacter spp., A.
calcoaceticus
Intra-abdominal E. coli, Klebsiella spp.,
infections Bacteroides spp.,
Enterobacter spp.
Gynecological infections E. coli, Bacteroides spp.

Amoxicillin- Augmentin; Pediatric oral Amoxicillin, 1.3 1 h after 250-mg/ Otitis media S. pneumoniae, H.
clavulanate GlaxoSmithKline suspension, 600 mg/ (18); 62.5-mg dose: influenzae, Moraxella
42.9 mg/5 ml q12hc clavulanate, amoxicillin, 6.9; catarrhalis
Chewable tablet and 1.0 (25) clavulanate, 1.6 Community-acquired H. influenzae, M.
oral suspension, 125 pneumonia or acute catarrhalis, H.
mg/31.25 mg q8h, bacterial sinusitis parainfluenzae, K.
200 mg/28.5 mg pneumoniae, methicillin-
q12h, 250 mg/62.5 susceptible S. aureus, S.
mg q8h, 400 mg/57.0 pneumoniae
mg q12h
Tablet, 250 mg/125 mg Sinusitis H. influenzae, M.
q8h, 500 mg/125 mg catarrhalis
q8-12h, 875 mg/125
mg q12h
Extended-release Skin and skin structure S. aureus, E. coli,
tablet, 1,000 mg/62.5 infections Klebsiella spp.
mg q12h Urinary tract infections E. coli, Klebsiella spp.,
Enterobacter spp.

Ticarcillin- Timentin; i.v., 3.0 g/0.1 g q4-6h Ticaracillin, 1.1 1 h after 3.1-g dose: Septicemia Klebsiella spp., E. coli, S.
clavulanate GlaxoSmithKline (45); ticarcillin, 131; aureus, P. aeruginosa
clavulanate, clavulanate, 1.8 Lower respiratory tract S. aureus, H. influenzae,
1.1 (25) infections Klebsiella spp.
Bone and joint infections S. aureus
Skin and skin structure S. aureus, Klebsiella spp.,
infections E. coli
Urinary tract infections E. coli, Klebsiella spp., P.
aeruginosa, Citrobacter
spp., Enterobacter
cloacae, Serratia
marcescens, S. aureus
Gynecologic infections Prevotella melaninogenicus,
Enterobacter spp., E.
coli, K. pneumoniae, S.
aureus, S. epidermidis
Intra-abdominal E. coli, K. pneumoniae, B.
infections fragilis
a
i.v., intravenous; i.m., intramuscular; q6h, every 6 h.
b
When treated with piperacillin-tazobactam, hospital-acquired and ventilator-acquired pneumonia caused by P. aeruginosa should be administered with an
aminoglycoside.
c
The dose of amoxicillin-clavulanate in the pedatric oral suspension is 45 mg/kg q12h based on information available in the material supplied by the manufacturer.

lin-clavulanate are available in Europe and have been studied increase activity against pathogens for which ticarcillin alone is
primarily for their efficacy against ESBL-producing E. coli bac- effective, such as non-␤-lactamase-producing Haemophilus
teremia (239, 370). spp., E. coli, Proteus spp., Enterobacter spp., Morganella spp.,
(ii) Ticarcillin-clavulanate. When introduced in 1985, ticar- Providencia spp., and P. aeruginosa. However, the combination
cillin-clavulanate was the first ␤-lactam–␤-lactamase inhibitor of ticarcillin and clavulanate does increase activity against
combination available for parenteral administration. Similar to ␤-lactamase-producing staphylococci, E. coli, H. influenzae,
the case for amoxicillin, the addition of clavulanate does not Klebsiella spp., Proteus spp., Pseudomonas spp., Providencia
VOL. 23, 2010 ␤-LACTAMASE INHIBITORS 173

spp., N. gonorrhoeae, Moraxella catarrhalis, and Bacteroides spp. genic abscesses, and diabetic foot infections. The early clinical
(59, 127, 319). Surprisingly, ticarcillin-clavulanate exhibits ac- success of this combination (and susceptibility of many patho-
tivity against the multidrug-resistant, non-lactose-fermenting gens to ampicillin-sulbactam and ticarcillin-clavulanate) estab-
S. maltophilia, and the use of this combination in addition to lished confidence in the role of ␤-lactam–␤-lactamase inhibitor
other antimicrobials (e.g., aztreonam or trimethoprim-sulfa- therapy. Unfortunately, the resistance to ampicillin-sulbactam
methoxazole) leads to synergistic killing (118, 275). This phe- among clinical isolates of E. coli is increasing (199).
nomenon may represent a very interesting opportunity to study Sulbactam is not well absorbed orally and must be admin-
the activity of ticarcillin-clavulanate against S. maltophilia. istered parenterally (126). However, the intravenous ampi-
Clavulanate induces the expression of chromosomally medi- cillin-sulbactam combination is well tolerated, with very few
ated AmpC ␤-lactamases in many Enterobacteriaceae and can reported side effects (7, 66).
antagonize the antibacterial effects of ticarcillin as a partner (iv) Piperacillin-tazobactam. Piperacillin in combination
␤-lactam (8, 203, 231, 237). This ticarcillin antagonism was with tazobactam became available in the United States in 1993.
observed in laboratory checkerboard studies with E. cloacae Piperacillin is a broad-spectrum penicillin that is bactericidal
and Morganella morganii strains (237). However, the impact of against many Gram-positive and Gram-negative aerobes and
␤-lactamase induction in the clinic is very hard to measure. anaerobes (140). As a single agent, piperacillin demonstrates
When testing or evaluating antibiotics in preclinical or clinical activity against P. aeruginosa, pneumococci, streptococci,
trials, ␤-lactamase induction may bear importance as an even- anaerobes, and Enterococcus faecalis, and this activity is re-
tual predictor of efficacy. In our opinion, the ability of ␤-lac- tained in combination with tazobactam (232). Clinicians must
tamase inhibitors to induce cephalosporinase production remember that the addition of tazobactam does not always
should be carefully examined and defined before clinical trials increase susceptibility of P. aeruginosa and other Gram-nega-
are performed. tive bacilli expressing AmpC ␤-lactamases (140). However,
In response to the potential for increased AmpC production tazobactam does extend piperacillin’s activity against most
due to ␤-lactam inducers (such as clavulanate, but including ␤-lactamase producing strains of Enterobacteriaceae, H. influ-
also cefoxitin, imipenem, and ceftazidime) as well as the rela- enzae, N. gonorrhoeae, and M. catarrhalis and has the potential
tively poor performance of amoxicillin-clavulanate and ticar- to lower MICs against these strains expressing ESBLs (59,
cillin-clavulanate against ESBL expressors, Livermore et al. 192). The in vitro inhibition of CMY-type ␤-lactamases by
and Nikaido et al. have proposed the combination of clavu- tazobactam is reported, but the clinical relevance of this phe-
lanate with a cephalosporin, such as cefepime or cefpirome, notype is still uncertain (34, 116a).
that is more stable against AmpC enzymes (239, 288). Several recent retrospective studies of clinical outcomes
Cefepime-clavulanate and cefpirome-clavulanate combina- from E. coli and K. pneumoniae ESBL-producing isolates
tions, with a constant concentration of 4 ␮g/ml clavulanate, argue for the efficacy of piperacillin-tazobactam in the treat-
were effective at ⱕ1 ␮g/ml against ESBL-producing Enterobac- ment of these infections (135, 336, 370, 420). Gavin et al.
teriaceae (239). The combination of cefepime and clavulanate showed that when in vitro testing revealed susceptibility to
enjoys a particular advantage due to the inability of class C piperacillin-tazobactam (ⱕ16/4 ␮g/ml), successful outcomes
enzymes to hydrolyze cefepime and the ability of clavulanate to were seen in 10 out of 11 non-urinary tract infections (135).
inhibit ESBLs. Currently, there are important obstacles to An examination of 43 bloodstream infections caused pri-
combining these agents (i.e., all three agents are at the end of marily by CTX-M ESBL-producing E. coli isolates revealed
patent protection, and the required trials of the formulations that piperacillin-tazobactam, amoxicillin-clavulanate (i.v.),
would be very expensive), but provided that the clinical re- or carbapenem treatment led to lower mortality than with
search can be performed, these combinations may be an at- cephalosporin or fluoroquinolone treatment (9% versus
tractive alternative to carbapenem treatment of ESBL-produc- 35%) (370). Another study, including infections caused by
ing organisms. We hasten to add that attention should be paid SHV- and TEM-type ESBL-producing K. pneumoniae, es-
to the pharmacokinetics and pharmacodynamics of these novel tablished that empirical therapy with ␤-lactam–␤-lactamase
combinations before randomized controlled trials are per- inhibitor combinations was validated by in vitro susceptibil-
formed, as it is not possible to “fix” a concentration of clavu- ity testing in 75% of cases, compared to 0% for oxymino-
lanate in vivo. cephalosporin treatment (420). Furthermore, initial treat-
(iii) Ampicillin-sulbactam. Ampicillin shows activity against ment with an adequate antibiotic was associated with lower
most streptococci, enterococci, Listeria spp., and strains of S. mortality rates (37%) than inadequate empirical therapy
aureus, H. influenzae, E. coli, P. mirabilis, Salmonella spp., and (67%).While this analysis did not stratify the outcome data
Shigella spp. that are devoid of ␤-lactamases (an important based on ESBL type, other authors have reported that
exception are ampicillin-resistant strains of H. influenzae that TEM-type ESBLs are more susceptible to piperacillin-ta-
do not contain a ␤-lactamase and often have substitutions in zobactam than are SHV-type ESBLs (185, 361).
PBP sequences) (194, 257). In combination with sulbactam, the Several observational studies have strongly suggested that
activity extends to ␤-lactamase-containing S. aureus, H. influ- the replacement of extended-spectrum cephalosporins with
enzae, M. catarrhalis, E. coli, Proteus spp., Klebsiella spp., and piperacillin-tazobactam can help lower the rates of infection
anaerobes (59, 362, 444). When the combination of ampicillin from Gram-negative ESBL producers and vancomycin-resis-
at 2.0 g and sulbactam at 1.0 g was marketed in 1987, this tant Enterococcus spp. (16, 43, 215, 216, 323, 355, 365). While
broad-spectrum activity made ampicillin-sulbactam ideal ther- the mechanism for decreasing ESBL-producing isolates is not
apy for polymicrobial infections such as abdominal and gyne- clear, it must be noted that the efficacy of ␤-lactam–␤-lacta-
cological surgical infections, aspiration pneumonia, odonto- mase inhibitor combinations may be reduced for organisms
174 DRAWZ AND BONOMO CLIN. MICROBIOL. REV.

producing multiple ESBLs, particularly if they also have an


AmpC ␤-lactamase (42, 321, 381). For this reason, we caution
against the use of piperacillin-tazobactam for the treatment of
ESBL-producing pathogens.
So, why do the in vitro susceptibility data and the clinically
cited data suggest that piperacillin-tazobactam can be used
FIG. 6. Imipenem tautomers hypothesized to form after acylation
in the treatment of ESBL producers? The reasons for this of carbapenems by serine ␤-lactamases. The deacylation rate of the
contradiction include that piperacillin, as a partner ␤-lactam ⌬1-pyrroline is significantly lower than that of the ⌬2-pyrroline, which
for ␤-lactamase inhibitors, is relatively resistant to hydroly- is thought to play a large role in the inhibitory activity of these com-
sis by certain plasmid-mediated ␤-lactamases (e.g., TEM) pounds (407, 458).
compared to amoxicillin, ampicillin, or ticarcillin. This sub-
strate stability may be due, in part, to piperacillin’s lower
affinity for and lower turnover (kcats) by these enzymes and In addition to their broad antibacterial spectrum, carbapenems
also piperacillin’s greater affinity for bacterial PBPs (234). are also effective inhibitors, or “slow substrates,” of most serine
We also propose that the pharmacokinetics and pharmaco- ␤-lactamases.
dynamics of the 3.375-g and 4.5-g formulations lead to ex- The stability of thienamycins to ␤-lactamase hydrolysis was
tended periods of time when free drug concentrations noted in early studies of imipenem, including low kcat values
(⬎100 ␮g/ml piperacillin in serum) are greater than the for the class A ␤-lactamase of B. cereus and a class C P.
MIC for ⱖ50% of the dosing interval for many nosocomial aeruginosa ␤-lactamase (150, 208, 270, 366). Charnas and
pathogens (242). It is likely that the high serum concentra- Knowles observed biphasic kinetics when RTEM was acylated
tions combined with the superior activity of piperacillin and by carbapenem compounds; an initial burst was followed by a
the relatively “better” inhibitory activity of tazobactam ver- slower steady-state reaction, strongly suggestive of a branched
sus clavulanate or sulbactam is enough to inhibit a majority reaction pathway (80, 114). Further work by Mobashery’s
of ␤-lactamases in a complex ␤-lactamase background (class group demonstrated that acylation of TEM-1 by imipenem
A and C ␤-lactamases) (328). However, this efficacy is not occurs readily and is followed by ⌬2- to ⌬1-tautomerization of
always clinically certain, and the physician must be aware the pyrroline double bond in the acyl-enzyme species (Fig. 6)
that the susceptibility pattern of an infecting Gram-negative (407). The rate of deacylation differs for the tautomers (the ⌬1
pathogen does not always reveal the number and complexity tautomer is approximately 10-fold slower than the ⌬2 tau-
of the ␤-lactamases present. Moreover, the formulation that tomer), and it was initially accepted that molecular rearrange-
is tested in the lab may not mirror what is administered to ments to the more stable (i.e., more slowly deacylated) species
patients. accounted for enzyme inhibition. Site-directed mutagenesis
studies of TEM-1 Arg244Ser showed that the guanidinium
group on Arg244 provides directly, or indirectly via a water
Inhibitory Activity of Carbapenems
molecule, the necessary proton for tautomerization to the ⌬1-
It is our opinion that consideration of carbapenems, al- pyrroline acyl-enzyme (407, 458). Incidentally, this water ap-
though they are not explicitly ␤-lactamase inhibitors, is es- pears to be the same molecule that serves as the proton donor
sential to a review of ␤-lactamase inhibition. These thien- necessary for clavulanate inhibition in class A enzymes (179).
amycin derivatives are among the “last line of defense” Elucidation of the crystal structures of several ␤-lactamases
against many multidrug-resistant pathogens. An increasing in complex with carbapenems has offered additional insight
body of evidence suggests that carbapenems are effective into the versatile inhibition mechanism of these compounds.
not just for their broad-spectrum antibacterial activity but Maveyraud et al. found that in the structure of TEM-1 com-
also for their multiple ␤-lactamase-inhibitory mechanisms. plexed with imipenem (PDB 1BT5), the ester carbonyl oxygen
The insight gleaned from the ability of carbapenems to of the acyl-enzyme was not located in the oxyanion hole (255).
inhibit serine ␤-lactamases may serve as a useful starting This approximately 180° rotation of the carbonyl was also seen
point for intelligent inhibitor design. Also, while we await in the crystal structure of acyl-enzyme species of imipenem
the development and release of novel ␤-lactamase inhibi- complexed via Ser64 with the AmpC ␤-lactamase (PDB 1LL5)
tors, it behooves us to wisely implement the ␤-lactams cur- (21). In contrast, the SHV-1/meropenem crystal demonstrated
rently in our armamentarium. With this in mind, a discus- two conformations of the intermediate acyl species, one where
sion of the inhibitory activity of carbapenems follows. the meropenem carbonyl oxygen of the acyl-enzyme is in the
Meropenem, imipenem, ertapenem, and doripenem (dorip- oxyanion hole and one where it is not (Fig. 7) (295). Proper
enem was released in the U.S. and European markets in 2007) position in this oxyanion hole, the binding pocket for the ␤-lac-
are often reserved for the most difficult-to-treat Gram-negative tam carbonyl, is important for both initial enzyme acylation
infections (Fig. 1, compounds 4 to 7) (320). Although resis- and hydrolysis of the ester bond (277, 425). A conformation
tance to carbapenems is rare in the clinic, several carbapenem- which places the ␤-lactam carbonyl outside of this electrophilic
hydrolyzing serine ␤-lactamases are identified, such as KPC-2, center may be delayed in hydrolysis.
NMC-A, SME-1, and IMI-1 (358, 455). These enzymes may be Computational models of TEM-1 with imipenem demon-
found in organisms that also produce additional ␤-lactamases, strated that the covalent adduct formed first after acylation did
conferring broad substrate profiles. For example, E. cloacae have its carbonyl in the oxyanion hole (255). However, this
isolates can produce IMI-1 or NMC-A, which hydrolyze car- conformation created a steric clash between the hydroxyethyl
bapenems, and also TEM and AmpC-type enzymes (359). group and residues 129 to 131, which forces the rearrangement
VOL. 23, 2010 ␤-LACTAMASE INHIBITORS 175

FIG. 7. Molecular representation of SHV-1/meropenem acyl-enzyme, based on PDB coordinates 2ZD8. The conformation with the carbonyl
of the acyl-serine bond in the oxyanion hole (formed by NH amides of Ser70 and Ala237) is colored teal, while the conformation with the carbonyl
flipped out is colored purple. Residues 244, 130, and 105, which demonstrate significant orientation changes compared to the apo SHV-1
␤-lactamase, are labeled. Glu166 and Asn132, important for making hydrogen bonds with the deacylation water (the approximate location is
represented as a red sphere) and C-6 hydroxyethyl substituent, respectively, are shown. The large R⬘ carbamoyl-pyrrolidinyl group of meropenem
was disordered and thus is only partially illustrated.

of the acyl-enzyme rotating the carbonyl outside the oxyanion cies with the carbonyl outside the oxyanion hole and the
hole. Thus, while producing a conformational change in the ⌬2-pyrroline tautomer to the acyl-enzyme with the carbonyl
enzyme, the C-6 hydroxyethyl substituent also facilitates rear- inside the electrophilic center (195). Further, this work sug-
rangement of the carbapenem to a more slowly deacylated gests that in crystal form the ⌬1- and ⌬2-tautomers are
form. Subsequent examination of the TEM Asn132Ala variant present in equal amounts but that ⌬1-pyrroline predomi-
demonstrated that replacement of the residue relieved the steric nates in solution. This ratio is consistent with the kinetic
encumbrance of the TEM-1/imipenem acyl-enzyme and allowed data implicating the ⌬1-tautomer as the primary inhibitory
the carbonyl to rotate back into the oxyanion hole (437). Further- species.
more, the TEM-1/imipenem structure indicated that hydrogen The recent structure of OXA-1, a class D monomeric ␤-lac-
bonds were formed between the hydroxyl group of the C-6 ␣-1R- tamase, inactivated by doripenem provided further support for
hydroxyethyl substituent on imipenem and both the Glu166-as- the role of carbapenems as inactivators of serine ␤-lactamases
sociated hydrolytic water and the side chain oxygen of Asn132 (PDB 3ISG) (379a). In this structure by Schneider and col-
(255). Similarly, the SHV-1/meropenem and class D OXA-13/ leagues, although the carbonyl oxygen is positioned in the
meropenem structures (PDB 2ZD8 and 1H8Y, respectively) re- oxyanion hole, water molecules are absent. The carbamate of
vealed that the putative deacylation water molecule has an addi- Lys70 forms a hydrogen bond to the C-6 hydroxyethyl group,
tional hydrogen-bonding interaction with the -OH group of and Lys212 and Thr213 form a salt bridge and hydrogen bond
meropenem’s C-6 ␣-1R-hydroxyethyl substituent (295, 334). This to doripenem. Furthermore, the bond angles of the acyl-en-
interaction weakens the nucleophilicity and/or changes the direc- zyme suggest the ⌬1-tautomer is formed.
tion of the lone pair of electrons of the water molecule and results In addition to the imipenem-induced conformation change
in poor turnover of the carbapenems. of class A and C ␤-lactamases, the (albeit slow) turnover of
As individual hydrogen atoms are difficult to resolve by carbapenems leads to a stable enzyme-product species that
X-ray crystallography, the structural data for ␤-lactamases occupies the ␤-lactamase active site for relatively longer than
and carbapenems have not been able to discriminate be- the enzyme-products of other ␤-lactams, such as penicillin
tween the ⌬1- and ⌬2-pyrroline tautomers. In contrast, Ra- (whose dissociation is limited by diffusion for class A enzymes)
man spectroscopy allows the monitoring of specific bond (149, 254). However, the deacylation rate was still lower than
character and compound reactivity. Recent studies of Ra- the dissociation rate for imipenem, suggesting that acyl-en-
man difference spectra in SHV-1 reacted with meropenem, zyme rearrangement accounts most significantly for inhibition.
ertapenem, and imipenem propose the correlation that the But notably, the presence of hydrolyzed inhibitor in the active
⌬1-pyrroline tautomer corresponds to the acyl-enzyme spe- site can contribute to enzyme inhibition.
176 DRAWZ AND BONOMO CLIN. MICROBIOL. REV.

INHIBITOR-RESISTANT CLASS A ␤-LACTAMASES significant resistance to the ␤-lactamase inhibitors (32, 68).
Present studies indicate that the KPC-2 ␤-lactamase is not
Within several years of the introduction of clavulanate com- inactivated by clavulanate, sulbactam, and tazobactam, as in-
binations for clinical use, resistance to amoxicillin-clavulanate dicated by high MICs and turnover numbers of 2,500, 1,000,
and ticarcillin-clavulanate was observed in isolates of E. coli and 500, respectively (319a). The ability of KPC-2 to avoid
and K. pneumoniae (247, 248, 376, 446). In 1987, Martinez et inactivation by the currently available inhibitors may represent
al. reported resistance rates in E. coli in Madrid hospitals and a novel class of highly IR class A enzymes. The rapid emer-
estimated that of amoxicillin-resistant strains, 20 to 30% were gence of Enterobacteriaceae harboring these carbapenemases is
co-amoxicillin-clavulanate resistant (248). Investigations in the a significant public health concern (290).
late 1980s demonstrated that this phenotype may result from
production of ␤-lactamases not susceptible to the inhibitors
(e.g., AmpCs from Enterobacter spp. or P. aeruginosa or me- Epidemiology and Detection of ␤-Lactamase
tallo-␤-lactamases) or enzyme hyperproduction (68). Hyper- Inhibitor Resistance
production of a ␤-lactamase can be mediated by mutations in Resistance to ␤-lactam–␤-lactamase inhibitor combinations
the promoter region of the gene and/or high copy numbers of challenges the ability to successfully treat serious urinary tract,
plasmids carrying the bla gene; both scenarios were identified respiratory tract, and bloodstream infections (154, 219, 376).
for the TEM-1 enzyme (247, 248, 446, 448, 449). The prevalence of organisms expressing inhibitor-resistant en-
Additional resistance mechanisms were concurrently be- zymes varies throughout the world. In 1993, a French study of
ing examined in research laboratories. In 1989, Oliphant 2,972 E. coli isolates from urinary tract infections (UTIs) found
and Struhl studied clavulanate and sulbactam resistance in that 25% and 10% of hospital and community isolates, respec-
TEM-1 by introducing a series of random substitutions into tively, showed amoxicillin-clavulanate MICs of ⬎16/2 ␮g/ml
the DNA segment which coded for the enzyme’s active site (160). Characterization of these isolates, including MIC pro-
(300). Through mutagenesis of a 17-amino-acid segment of files (amoxicillin-clavulanate MIC90 of ⬎1,042 ␮g/ml and
the TEM-1 active site (Arg61 to Cys77) and functional se- cephalosporin MIC of ⬍32 ␮g/ml), isoelectric focusing, and
lection assays, they found that enzymes with an Ile, Leu, or DNA-DNA hybridization, suggested that 27.5% and 45% of
Val substitution at Met69 had increased resistance to ampi- hospital and community isolates, respectively, were TEM-1
cillin-clavulanate and ampicillin-sulbactam. This foreshad- derived and had substitutions at previously described amino acid
owed an important amino acid substitution in inhibitor- positions that confer the IRT phenotype. In this survey, 4.9% of
resistant (IR) TEM. E. coli isolates from UTIs were IRT producing. In 1998, a geri-
Three years later, Zafaralla et al. engineered by site-directed atric department of a French hospital reported an outbreak of
mutagenesis the Arg244Ser variant of TEM-1 to explore the amoxicillin-clavulanate-resistant isolates which all produced the
role of this key amino acid and its interaction with the peni- same IR TEM ␤-lactamase, TEM-30 (Arg244Ser) (141). In a
cillin C-3 carboxylate (457). This work advanced the earlier study published in 2000, Leflon-Guibout et al. determined the
hypothesis by Moews et al. that Arg244 played an important molecular mechanism of amoxicillin-clavulanate resistance (de-
role in the recognition of the conserved substrate C-3/C-4 carbox- fined by MICs of ⬎16/2 ␮g/ml) in E. coli isolates from three
ylate (179, 267). Further studies by Mobashery’s laboratory also French hospitals from 1996 to 1998 (218). The overall resistance
revealed that Arg244Ser was more resistant to inactivation by rate was 5%, and the majority of these resistant organisms were
clavulanate (179). Interestingly, the first IR class A ␤-lactamase from patients with respiratory tract infections. Production of IRTs
was isolated from a clinical isolate of E. coli in the same year. The was implicated in resistance in 30 to 41% of the isolates over the
enzyme was determined to be related to TEM-1 or TEM-2 and time period, which was only slightly less frequent than the hyper-
hence was designated inhibitor-resistant TEM (IRT-1) (29, 427). production of chromosomal AmpC enzymes. Two independent
Sequencing information revealed an Arg-to-Cys or -Ser substitu- Spanish studies reported production of IRTs in 5.4% and 9.5% of
tion at residue 244 (23, 427). amoxicillin-clavulanate-resistant E. coli isolates (266, 331).
Since that time in the early 1990s, the number of clinically The first IRT identified in the United States was not reported
identified IRTs has expanded to include more than 35 unique until 2004 in a 14-month survey of E. coli isolates from a north-
enzymes (Table 4) (www.lahey.org/studies/webt.asp). In addi- eastern tertiary care clinical microbiology laboratory (198). Kaye
tion to amoxicillin-clavulanate-resistant E. coli, IRTs have et al. found that 24% of the 283 isolates classified as ampicillin-
been identified in Klebsiella, Proteus, Shigella, and Citrobacter sulbactam resistant by disk diffusion and MIC testing were also
spp. (385). IR SHVs have also been isolated from K. pneu- resistant to amoxicillin-clavulanate as determined by disk diffu-
moniae, with a total of five IR SHVs presently described (Table sion zone diameters of ⱕ13 mm. Of the amoxicillin-clavulanate-
5) (109). Operationally, inhibitor resistance refers to resistance resistant isolates, 83% were from community-acquired infections.
to amoxicillin-clavulanate and may, but does not necessarily, Many of these 69 isolates were recovered from UTIs of outpa-
include resistance to sulbactam and tazobactam (410). More- tients, and two expressed the IR TEM-34 (Met 69Val) and one
over, we stress that this is a “relative resistance”; provided that the TEM-122 (Arg275Gln) enzyme. Incidentally, also reported in
sufficient clavulanate concentrations are used, these IR en- 2004 was a KPC-2-producing K. pneumoniae isolate from New
zymes can be inactivated. York City that contained IRT-2 (41).
Currently, reports describing non-TEM, non-SHV family IR Isolates producing IRTs are more frequently reported in
enzymes are becoming more common. The CTX-M ESBLs, Europe than in the United States. The explanation for this
which typically hydrolyze penicillins and extended-spectrum discrepancy is not clear and probably reflects a combination
cephalosporins (preferentially cefotaxime), have not yet shown of environmental, methodological, and clinical practice fac-
VOL. 23, 2010 ␤-LACTAMASE INHIBITORS 177

TABLE 4. Clinically identified inhibitor-resistant TEM (IRT) ␤-lactamasesa


Amino acid at positionb:
␤-Lactamase
21 39 69 104 127 130 165 182 221 238 244 261 265 275 276

TEM-1 Leu Gln Met Glu Ile Ser Trp Met Leu Gly Arg Val Thr Arg Asn
TEM-30 (IRT-2) Ser
TEM-31 (IRT-1) Cys
TEM-32 (IRT-3) Ile Thr
TEM-33 (IRT-5) Leu
TEM-34 (IRT-6) Val
TEM-35 (IRT-4) Leu Asp
TEM-36 (IRT-7) Val Asp
TEM-37 (IRT-8) Ile Asp
TEM-38 (IRT-9) Val Leu
TEM-39 (IRT-10) Leu Arg Asp
TEM-40 (IRT-11) Ile
TEM-44 (IRT-13) Lys Ser
TEM-45 (IRT-14) Leu Gln
TEM-51 (IRT-15) His
TEM-54 Leu
TEM-58 Ser Ile
TEM-59 (IRT-17) Lys Gly
TEM-65 (IRT-16) Lys Cys
TEM-67 Ile Lys Cys
TEM-73 (IRT-18) Phe Cys Met
TEM-74 (IRT-19) Phe Ser Met
TEM-76 (IRT-20) Gly
TEM-77 (IRT-21) Leu Ser
TEM-78 (IRT-22) Val Arg Asp
TEM-79 Gly
TEM-80 (IRT-24) Leu Val Asp
TEM-81 Leu Val
TEM-82 Val Gln
TEM-83 Leu Cys Gln
TEM-84 Asp
TEM-103 (IRT-28) Leu
TEM-122 Gln
TEM-145 Met His
TEM-159 Phe Ile
TEM-160 Lys Val Thr
a
Based on data from www.lahey.org/studies/webt.asp.
b
Unless otherwise indicated, the amino acid is the same as in the non-IR TEM-1.

tors. The phenomenon does not appear to be related to 198, 301). Currently, an international standard for the amount
large differences in the use of ␤-lactam–␤-lactamase inhib- of ␤-lactamase inhibitor that should be combined with the
itor combinations, as these agents are widely used in both partner ␤-lactam for detection of IR enzymes is not in use, and
Europe and the United States and likely produce similar different combinations can significantly alter the assigned results
selective pressures on bacteria (68, 159). (301, 409). For example, the use of the 2:1 ratio for amoxicillin-
The detection of IRTs presents a significant number of op- clavulanate appears to underestimate the presence of IR ␤-lac-
erational challenges. Disagreements between the results of tamases compared to employing the fixed concentration of 2
MIC testing and disk diffusion assays are often seen, especially ␮g/ml clavulanate (409). Further, organisms producing low levels
among isolates with intermediate resistance phenotypes (68, of IR enzymes may be undetectable at the 2:1 ratio (302). As a

TABLE 5. Clinically identified inhibitor-resistant SHV ␤-lactamasesa


Amino acid at positionb:
␤-Lactamase
35 69 130 140 146 187 192 193 234 238 240

SHV-1 Leu Met Ser Ala Ala Ala Lys Leu Lys Gly Glu
SHV-10 Gly Arg Asn Val Ser Lys
SHV-26 Thr
SHV-49 Ile
SHV-56 Gln Arg
SHV-72 Val Arg
a
Based on data from www.lahey.org/studies/webt.asp.
b
Unless otherwise indicated, the amino acid is the same as in the non-IR SHV-1.
178 DRAWZ AND BONOMO CLIN. MICROBIOL. REV.

TABLE 6. Clinically identified complex mutants of TEM (CMT) ␤-lactamasesa


Amino acid at positionb:
␤-Lactamase
39 69 104 130 164 237 238 240 244 265 275 276 284

TEM-1 Gln Met Glu Ser Arg Ala Gly Glu Arg Thr Arg Asn Ala
TEM-50 (CMT-1) Leu Lys Ser Asp
TEM-68 Ser Lys Met Leu
TEM-89 (CMT-3) Lys Lys Gly Ser
TEM-109 (CMT-5) Leu Lys His Cys
TEM-121 (CMT-4) Lys Lys Ser Thr Lys Ser
TEM-151 Val His Asp Gly
TEM-152 Val His Lys Asp
TEM-154 Leu Ser
TEM-158 (CMT-9) Leu Ser Asp
a
Based on data from www.lahey.org/studies/webt.asp.
b
Unless otherwise indicated, the amino acid is the same as in the non-IR TEM-1.

result of the methodological complications of identifying IR generated in the laboratory. IRTs, when derived from mutated
␤-lactamase producers, the actual IRT prevalence may be under- wild-type bla genes, typically display a reduction in ␤-lactam
estimated (68). Definitive identification requires specific enzyme catalytic efficiency (kcat/Km), mediated by lower kcat and in-
kinetic testing, determination of isoelectric points, and molecular creased Km values for penicillins compared to the wild-type
characterization (60, 72, 77). Routine susceptibility tests may not enzymes (35). IR ␤-lactamases have increased KIs and IC50s
accurately detect strains expressing IRTs. A direct correlation for clavulanate and sulbactam; smaller increases are observed
between the level of resistance (MIC) and the predicted clinical for tazobactam. In general, the MICs for organisms expressing
outcome is not yet known. IR enzymes are lowered for penicillins and increased for cla-
vulanate and sulbactam. MICs may remain in the susceptible
Substitutions in Class A Enzymes range for tazobactam when obtained with the combination of
Conferring Inhibitor Resistance piperacillin-tazobactam. This susceptibility is likely due to pip-
eracillin’s greater potency and rapid cell entry (see above) as
The amino acid substitutions associated with clavulanate
resistance are different from those that produce the ESBL well as the relative preservation of tazobactam’s KI (36). Of
phenotype, and thus IRTs have unique hydrolytic profiles com- significant interest is the recent description of the kinetic prop-
pared to ESBLs. For example, the E. coli isolates that contain erties of SHV-72 (Ile8Phe, Ala146Val, Lys234Arg) (258). In
IRTs may be more susceptible to cephalosporins, carbapen- contrast to most other IR enzymes, this unique IR SHV dem-
ems, and monobactams (35). However, among TEM and SHV onstrates increased catalytic efficiency for penicillins.
␤-lactamases, substitutions that confer an IR phenotype are Amino acid substitutions in TEM and SHV result in resis-
sometimes found in pairs or are combined with amino acid tance to clavulanate by two primary mechanisms: (i) alter-
changes that also increase the ability of the enzyme to hydro- ations in the geometry of shape of the oxyanion hole and (ii)
lyze oxyimino-cephalosporins. These variants are designated changes in the position of Ser130 or Arg244 (33, 73, 101, 120,
complex mutants of TEM (CMTs) and may foreshadow the 408, 411, 416). The individual residues that are most commonly
emergence of a new class of versatile ␤-lactamases with even substituted in IR TEM enzymes are Arg244, the nearby
broader hydrolytic and resistance profiles (Table 6) (369). Asn276 and Arg275, Met69, and the active-site Ser130 (www
Table 7 summarizes kinetic properties of representative .lahey.org/studies/webt.asp) (Fig. 8).
class A IR enzymes that have been identified clinically or

TABLE 7. Kinetic properties of representative class A inhibitor-resistant and wild-type enzymesa


Clavulanate Sulbactam Tazobactam
␤-Lactamase Reference(s)
⫺1 ⫺1
KI (␮M) kinact (s ) tn KI (␮M) kinact (s ) tn KI (␮M) kinact (s⫺1) tn

TEM-1 0.1 0.02 160 1.6 0.002 10,000 0.01 ⬎0.02 140 73, 180
TEM Met69Leu 1.65 0.002 2,800 22 0.0009 7,000 0.20 0.004 2,900 73
TEM Ser130Gly 105 0.003 1,600 220 0.03 1,900 95 0.04 630 408
TEM Arg244Ser 33 44 0.00001 7,800 179, 180
TEM Asn276Asp 15.6 0.01 250 378
TEM Met69Leu, Asn276Asp 27 49 0.6 386
SHV-1 1 0.04 60 8.6 0.056 13,000 0.07 0.1 5 153, 410, 411
SHV Arg244Ser 63 0.09 50 240 0.13 100 410, 411
SHV Ser130Gly 47 0.03 40 4.2 0.06 5 153
OHIO-1 0.4 0.01 80 17 0.001 40,000 0.30 0.01 400 229
OHIO Met69Ile 10 0.002 2,000 68 0.0004 200,000 18 0.0016 2,000 229
a
The definition of KI may not be uniform from laboratory to laboratory. See the cited references for the methods used in each determination.
VOL. 23, 2010 ␤-LACTAMASE INHIBITORS 179

and are related to slight differences in the inactivation mech-


anisms for these enzymes (179, 410, 411, 436, 457).
Methionine 69. Met69 is the most commonly substituted res-
idue in inhibitor-resistant enzyme isolates, appearing either as a
single substitution or in combination with additional amino acid
replacements in 15 of the 36 presently defined IRTs. Despite the
proximity to enzyme nucleophilic Ser70, the Met69 side chain is
oriented away from the catalytic serine and not part of the sub-
strate binding site (191). Residue 69 forms the back wall of the
oxyanion hole or “electrophilic center” that polarizes the ␤-lac-
tam for nucleophilic attack by Ser70 and, in the case of inhibitors,
possibly Ser130 as well (206). The residue is not highly conserved
in class A ␤-lactamases, and TEM can tolerate many substitutions
at the site and still retain activity against most substrates (10, 101).
The SHV Met69Phe, -Lys, and -Tyr variants have increased MICs
and hydrolytic activity for the extended-spectrum ceftazidime
(155). While the residue displays some flexibility in its role in
substrate binding, certain substitutions in TEM and SHV confer
FIG. 8. Molecular representation of TEM-1 active site, showing resistance to the inhibitors.
residues that are most frequently implicated in the development of The TEM variants, Met69Ile, -Val, and -Leu, each show an
inhibitor-resistant TEM enzymes. Based on PDB coordinates 1TEM.
approximately 10-fold increase in KI and 10-fold decrease in
kinact, leading to a 100-fold decrease in inhibitory efficiency
Arginine 244. To date, 12 IRT variants with substitutions at (kinact/KI) (73). Evidence from X-ray crystallography and mo-
residue 244 have been reported. The guanidinium group lecular dynamic studies has revealed that each substitution
(CH5N3⫹) of Arg244 contributes to substrate affinity through introduces subtle active-site changes that perturb inhibitor rec-
hydrogen bonding to the conserved ␤-lactam carboxylate ognition and enzyme catalysis (262, 438). Similarly, SHV
(436). Structural and computational studies of the mechanism Met69Ile, -Val, and -Leu show increased clavulanate MICs
of clavulanate resistance in TEM-1 reveal that a hydrogen on and IC50s (155). Reduced inhibitor acylation efficiency in SHV
the N␩2 atom of the guanidinium group of Arg244 and the was elucidated by observed changes in the oxyanion hole in the
carbonyl oxygen atom of the backbone of Val216 coordinate a crystal structures of SHV Met69Val/Glu166Ala in complex
water molecule (Fig. 9) (179, 411, 436, 457). When clavulanate with clavulanate, sulbactam, and tazobactam (PDB 2H0T,
is bound, this water molecule is the likely donor of a proton 2H0Y, and 2H10, respectively) (416).
that saturates the double bond at the C-2 position, facilitating Asparagine 276. Fourteen TEM variants with substitutions
the opening of the five-membered ring, formation of an inter- at Asn276 have been discovered; seven demonstrate an IR
mediate, and eventual ␤-lactamase inactivation (179). Loss of phenotype. Ambler position 276 remains unchanged in cur-
the guanidinium group of Arg244 displaces this water molecule rently identified SHV variants. Despite its relative distance (8
and may explain the inhibitor resistance and catalytic impair- to 10 Å) from the enzyme active site, the crystallographic
ment that is observed in the TEM Arg244 variants. Without structure of TEM Asn276Asp (PDB 1CK3) reported by
protonation of the double bond, elimination of oxygen at C-1 Swaren et al. confirmed the importance of the electrostatic
is thermodynamically unfavored, which may lead to increased environment surrounding residue 276 (400). In TEM, amino
inhibitor turnover (179). acid 276 is located on the enzyme’s C-terminal ␣-helix H11,
Interestingly, we have not found a clinical report of an SHV which lies behind the ␤-sheet including Arg244. The
enzyme that has the 244 substitution. However, mutagenesis of Asn276Asp substitution creates new interactions with Arg244
SHV at Arg244 revealed that the 244Leu and -Ser substitu- and significant displacement of ␣-helices H1, H10, and H11.
tions at this residue also confer resistance to amoxicillin-clav- The Asp at residue 276 is displaced only 0.75 Å compared to
ulanate (138, 411). The biochemical correlates of these clav- the wild-type Asn, but this movement results in a tighter at-
ulanate-resistant phenotypes are different for SHV and TEM traction between Arg244 and 276Asp.
This enhanced interaction has two major effects (378, 400).
First, there is an effective “neutralization” of the Arg244 con-
tribution. This decreased positive charge may lower the local
affinity for the C-3 carboxylate of clavulanate and penicillins
(400). Charge changes in the active site may have a more
profound consequence for the inhibitors, which are positioned
as small penicillin substrates and rely heavily on the interac-
tions with Arg244 for binding affinity (179, 378, 457). Second,
the TEM Asn276Asp variant’s increased clavulanate kcat val-
ues may be explained by the displacement of the water mole-
cule necessary for secondary ring opening of clavulanate, as
FIG. 9. Schematic representation of the proposed Michaelis discussed above for TEM Arg244 variants (378, 400).
preacylation complex of TEM-1 and clavulanate. Similar to the case for Arg244, IR SHV 276 variants have
180 DRAWZ AND BONOMO CLIN. MICROBIOL. REV.

not yet reached clinical attention (411). Molecular modeling of for inhibition is intact, the decreased accumulation of acyl-
SHV Asn276Asp showed that novel electrostatic interactions enzyme intermediates and inactivation products can lead to
are formed between Arg244N␩2 and both 276AspO␦1 and inhibitor resistance in vivo.
O␦2, reminiscent of the TEM Asn276Asp crystal structure Arginine 275. Similar to observations regarding Arg244 and
(108). Additional data from both kinetic studies and timed Asn276, the Arg275 variant has been identified only in TEM.
electrospray ionization mass spectrometry (ESI-MS) of clavu- Before description of the IR variants TEM-103 and TEM-122,
lanate-inhibited SHV-1 and SHV Asn276Asp suggest that the which possess only the Arg275Leu and -Gln changes, respec-
Asn276Asp phenotype is produced by decreased affinity for tively, substitution at 275 was seen only in combination with
the C-3 carboxylate of clavulanate and not by a change in the the Met69Val and -Leu substitutions (9, 198). Hence, the con-
reaction mechanism. These results are consistent with the no- tribution of Arg275 to the IR phenotype was not appreciated,
tion advanced by Thomson et al., suggesting that clavulanate and rigorous kinetic and structural studies of this variant have
inactivation in SHV may not rely on a water molecule coordi- not yet been completed. However, from crystal structures of
nated by Arg244 and Val216, as in TEM enzymes (191, 210, the wild-type enzymes and a better understanding of properties
411). These important differences in TEM and SHV mecha- at nearby Arg244 and Asn276, Chaibi et al. postulated that
nisms support a divergent evolution of these enzymes vis-a-vis substitutions at Arg275 (like those at Asn276) disrupt local
substrate and inhibitor profiles. electrostatic interactions and displace the water molecule that
Serine 130. Ser130 is a conserved amino acid among all class is key to inactivation by clavulanate (74).
A ␤-lactamases but is replaced by a Gly residue in two IRTs Lysine 234. The recent definition of two clinical IR SHV
(TEM-59 and -76) and one IR SHV enzyme (SHV-10). The ␤-lactamases with Lys234Arg substitutions draws attention to
multiple roles of this residue include anchoring ␤-lactams to this important active-site residue (110, 258). Previously, TEM
and stabilizing the active site through hydrogen bonding with or SHV variants obtained from the clinic had not been iden-
the C-3/C-4 carboxylate of the inhibitors and substrates and tified as having changes at this highly conserved class A resi-
facilitating proton transfer to the ␤-lactam nitrogen during due. Site-directed mutagenesis of Lys234 to Arg in TEM-1
acylation, leading to ␤-lactam ring opening (179, 180, 214, leads to a 10-fold-decreased affinity for penicillins, presumably
426). Ser130 also serves as a second nucleophile which attacks because of the hydrogen-bonding role of Lys234 to the C-3
the imine intermediate and becomes covalently cross-linked to carboxylate (220) (Fig. 3). However, for the clinically isolated
Ser70 in the terminal inactivation of the mechanism-based inhib- SHV-72 enzyme (Ile8Phe, Ala146Val, Lys234Arg), Km values
itors (209). Removing the cross-linking residue seems a clear way for penicillins were not significantly changed and IC50s for
to overcome inactivation, but inhibitors, and particularly tazobac- clavulanate increased 10-fold compared to those of SHV-1
tam, retain efficacy against the Ser130Gly enzyme. Based on these (258). Interestingly, the kcat values for the penicillins were
mechanistic roles, how does the Ser130Gly variant maintain hy- increased, up to 5.8-fold for ticarcillin. Molecular dynamics
drolytic activity and why does it confer resistance to inhibitors simulations show that Lys234Arg induces movement of the
(408)? In response to the first part of the question, evidence from Ser130 oxygen away from Ser70. Mendonca et al. proposed
X-ray crystallography of SHV and TEM Ser130Gly (PDB 1TDL that the mechanism of IR in SHV-72 may be due to movement
and 1YT4, respectively) confirmed that a relocated water mole- of Ser130 based on its role in both hydrolysis and terminal
cule, initially predicted by Helfand et al., compensates for the loss inactivation by cross-linking with Ser70 (258).
of the Ser130 hydroxyl group by donating a proton to the ␤-lac-
tam nitrogen (153, 157, 180, 408, 438). The decreased catalytic
efficiencies seen in the TEM and SHV variants are likely a result THE PROMISE OF NOVEL ␤-LACTAMASE INHIBITORS
of perturbations caused by the repositioned water molecule.
This section reviews compounds that have demonstrated
These perturbations vary somewhat between SHV and TEM
favorable inactivation properties against ␤-lactamases and in-
Ser130Gly (e.g., reorientation of Ser70 and Lys73 in SHV and
troduces newer compounds showing promise as “second-gen-
TEM Ser130Gly, respectively), but both involve changes in hy-
eration” inhibitors. Table 8 provides Ki (or KI) values and IC50s
drogen bonding networks in the active site, which are modified to
for representative inhibitors against the different ␤-lactamase
accommodate the new water molecule (397, 408).
classes to illustrate the challenge of achieving broad-spectrum
In regard to the second part of the question concerning
inhibition.
inhibitor resistance, the data on Ser130Gly variants argue that
cross-linking is not essential for enzyme inactivation. Despite
increased MICs for the inhibitors, the turnover numbers for Monobactam Derivatives
clavulanate and tazobactam by SHV Ser130Gly are equivalent
to those by the wild-type enzyme (153). This “paradox of in- Monocyclic, N-sulfonated ␤-lactams are a family of antibi-
hibitor resistance” illustrates that despite replacement of a key otic compounds produced by bacteria (178, 404, 405). Struc-
catalytic residue, SHV Ser130Gly is still capable of inactivation ture-activity studies of these monobactams led to the develop-
by the mechanism-based inhibitors (153, 157, 311, 397). The ment of the synthetic compound aztreonam, which interacted
resistant phenotype presents primarily in whole-cell assays, with the PBPs of a wide range of aerobic Gram-negative bac-
where bacteria are exposed to a limited inhibitor concentra- teria, including P. aeruginosa, and was introduced into clinical
tion. Structural perturbations created by the Ser130Gly substi- practice in 1984 (151, 168, 403). In addition, aztreonam (Fig. 1,
tution discourage formation of the preacylation complex (in- compound 3) resists hydrolysis by many plasmid-mediated
creased inhibitor KIs), which ultimately leads to fewer ␤-lactamases such as TEM-1 and -2, OXA-2, and SHV-1, be-
inactivation events (157). Thus, while the enzymatic machinery having as a very poor substrate with low affinity (e.g., Km of 2.9
VOL. 23, 2010 ␤-LACTAMASE INHIBITORS 181

TABLE 8. Kinetic properties of selected inhibitors against different ␤-lactamase Ambler classesa

Kinetic Value (␮M) for Ambler class:


Inhibitor (compound no. in Fig. 1) Reference(s)
parameter A B C D

Aztreonam (3) Km or Ki 200 ⬎1,000 0.0019 56, 217, 337


BAL30072 siderophore monobactam (14) IC50 1.7 ⬎100 0.09 134
ATMO-penicillin (15) IC50 0.90 418
BRL 42715 methylidene penem (16) IC50 0.008 0.008 0.004 87
BLI-489 methylidene penem IC50 0.009 0.062 294
Novel tri- and bicyclic methylidene IC50 0.0004–0.0031 0.014–0.260 0.0015–0.0048 0.012 25, 441
penems (including 17 and 18)
LK-157 tricyclic carbapenems (20) IC50 0.055 0.062 344
Ro 48-1220 penam sulfone (21) IC50 0.05–1.07 24–200 0.21–3.4 0.10–0.43 367, 422
LN-1-255 penam sulfone (23) KI or IC50 0.100 0.026 0.70 63, 322; Drawz et al.,
unpublished data
Boronic acid meta-carboxyphenyl Ki 3.9 0.001 272, 411
cephalosporin analog (25)
NXL104 (26) IC50 0.008 0.080 31
J-111,225 1-␤-methylcarbapenem (33) Ki 11.1 0.18 0.93 281
Phosphonates Ki 0.08–76 0.016–12.1 0.094 4, 244
C-6-mercaptomethyl penicillnate (38) IC50 6.8 0.10 10.5 61
a
The definition of KI may not be uniform from laboratory to laboratory. See the cited references for the methods used in each determination.

mM for TEM-2) (403). With class C enzymes, such as E. C-3 and C-4 limits this rotation and stabilizes the acyl-enzyme
cloacae P99, aztreonam forms a stable acyl-enzyme that is very against hydrolysis by AmpC enzymes (99, 171).
slowly hydrolyzed (low kcat) (128, 403). Aztreonam also exhib- Heinze-Krauss et al. synthesized and evaluated of a panel of
its very low Ki values for chromosomally encoded cephalospo- bridged monobactams that exhibited very favorable inhibition
rinases (e.g., 1.9 nM for P99) and serves as a transient inacti- of the class C C. freundii and P. aeruginosa ␤-lactamases (IC50s
vator (56, 128). In vivo, where periplasmic ␤-lactamase of as low as 10 nM) but lower affinities for the class A TEM-3
concentrations can be in the low mM range, clinically signifi- (IC50s of ⬎100 ␮M) (152). At sufficiently high concentrations,
cant catalytic efficiencies may be obtainable (128). class A ␤-lactamases were acylated rapidly, but the rate of
Further modification of the monobactam nucleus was em- deacylation was higher, leading to high hydrolysis rates and low
barked upon to capitalize on the inhibitory activity of aztreo- active-site occupancy. Comparison of the crystal structures of
nam. The 1,5-dihydroxy-4-pyridone monobactam Syn 2190 class A TEM-1, PC1, the ␤-lactamase from Bacillus lichenifor-
(Fig. 1, compound 11) possesses a novel C-3 side chain that mis 749/C, and the class C ␤-lactamase from C. freundii re-
may utilize the iron uptake pathway to enter Gram-negative vealed that in class A enzymes, hydrolysis occurs on the oppo-
bacteria. Syn 2190 displayed IC50s in the nM range for cepha- site side of the bridged monobactam ester by a water molecule
losporinases (e.g., 6 nM for E. cloacae P99) and synergy with activated by hydrogen bond networks not present in class C
ceftazidime and cefpirome in MIC testing of clinical strains, enzymes (152, 166, 191, 267, 299, 392). Rotation about the
including AmpC-derepressed variants of P. aeruginosa (15, C-3–C-4 bond is less critical to the inhibition mechanism in
289). In addition, Syn 2190 in combination with ceftazidime or class A enzymes, and restricting this rotation does less to sta-
cefpirome (at a 1:1 ratio) showed improved efficacy compared bilize the acyl-enzyme. For example, the bridged monobactam
to ceftazidime-tazobactam in murine models of systemic infec- Ro 48-1256 (Fig. 1, compound 12) (at 4 and 8 ␮g/ml) effec-
tions with cephalosporin-resistant P. aeruginosa. Syn 2190 has tively potentiated the activity of imipenem against both induc-
also shown utility for the challenging clinical task of identifying ible and derepressed class C-expressing P. aeruginosa isolates
production of plasmid-mediated AmpC ␤-lactamase in Kleb- (238). The activities of piperacillin and ceftazidime were also
siella spp. In combination with cefotetan, Syn 2190 achieved potentiated by the combination with Ro 48-1256 against the
100% specificity and 91% sensitivity for AmpC producers (28). strains expressing elevated quantities of AmpC enzyme. How-
However, Syn 2190 has lower affinity than tazobactam for class ever, Ro 48-1256 did not improve the activity of piperacillin,
A enzymes and could not restore susceptibility to cephalospo- ceftazidime, or carbapenems against organisms expressing
rins in bacteria expressing these enzymes. This limitation class A, B, or D ␤-lactamases.
against class A enzymes may partly explain the absence of Modification of the monobactam structure at the ␤-lactam
further development of this compound. nitrogen has had some success against class A ␤-lactamases
The design of bridged monobactam derivatives was guided (50, 171). Analysis of a series of N-sulfonyloxy-␤-lactam
by the crystal structure of the monobactam aztreonam in com- monobactam derivatives demonstrated rapid acylation of the
plex with the class C ␤-lactamase from Citrobacter freundii TEM-1 active site and resistance to deacylation, as evident
(PDB 1FR1) (152, 299). Studies of the structure demonstrated from turnover numbers ranging from 2 to 8 (50). Additional
that before deacylation, aztreonam had to rotate around the analysis of one of these derivatives (Fig. 1, compound 13)
C-3–C-4 bond to allow a hydrolytic water access to the ester revealed that following enzyme acylation in the class A car-
bond. This rearrangement requires breaking and reforming of bapenemase NMC-A, the tosylate group is eliminated and
the active-site hydrogen bond network and leads to rate-limit- gives rise to two acyl-enzyme species (273). These inhibited en-
ing deacylation (205). The introduction of a bridge between zyme species are trapped in local energy minima which yield low
182 DRAWZ AND BONOMO CLIN. MICROBIOL. REV.

deacylation rates. Based on molecular simulations, Mourey et al. groups onto conserved substrate skeletons, which will facilitate
postulated that the acyl-enzyme alternates between two different recognition by the enzyme (418).
conformations that move in and out of the oxyanion hole (273).
This motion requires breaking and reforming of hydrogen bonds
Penems
between the acyl-enzyme ester and carbonyl oxygen atoms and
the oxyanion hole backbone nitrogens and Ser130 oxygen atom. BRL 42715 and Syn 1012. Methylidene penems are potent
In both conformations, the water implicated in the deacylation inhibitors in which the inhibitory properties of the penem
reaction is positioned poorly for hydrolysis, offering a structural structure are enhanced by introduction of a double bond at C-6
explanation for the irreversible inhibition. and a sulfide at penem position 1 (260). These compounds
A series of monobactam derivatives was recently developed offer advantages over the traditional ␤-lactamase inhibitors,
to target pathogens harboring multiple ␤-lactamases. Some of including (i) a large R1 side chain that may aid in cell perme-
these monobactam analogues act as very effective bactericidal ability and contribute to active-site affinity and (ii) a novel
agents for difficult-to-treat Gram-negative pathogens, while inactivation mechanism. The parent compound from this
others are bridged monobactams, such as BAL29880, that are group, BRL 42715 C-6-(N1-methyl-1,2,3-triazolylmethyl-
inhibitors of AmpC enzymes (99, 102, 116a, 317). Additional ene)penem (Fig. 1, compound 16), is an effective inhibitor of
derivatives include monobactams bearing a siderophore side class A, C, and D ␤-lactamases (119, 250). IC50s of BRL 42715
chain which can enhance cell entry through bacterial iron up- were 10- to 100-fold lower than those of clavulanate, tazobac-
take systems (49). Of this siderophore type, BAL19764 and tam, and sulbactam for class A TEM-1 and SHV-1, class C P99,
BAL30072 (Fig. 1, compound 14) have potent antibacterial class D OXA-1, and the S. aureus ␤-lactamase NCTC 11561
activity against carbapenem-resistant P. aeruginosa, Acineto- (87). However, BRL 42715 was a substrate for class B metallo-
bacter spp., S. maltophilia, and S. marcescens as well as against ␤-lactamases from Aeromonas hydrophila and S. maltophilia
Gram-negative bacilli expressing VIM and IMP ␤-lactamases and for BcII (250). In susceptibility testing, BRL 42715 con-
(37, 102). Additionally, BAL19764 is stable to hydrolysis by centrations of 0.25 ␮g/ml or lower were able to restore amoxi-
MBLs (314). Further, these siderophore compounds are po- cillin MICs to ⬍16 ␮g/ml for TEM-1- and OXA-1-producing
tent enzyme inhibitors; BAL30072 has low ␮M IC50s for organisms (87). Further, amoxicillin-BRL 42715 combinations
TEM-3 (1.7 ␮M), and the AmpCs from C. freundii and P. were more effective than amoxicillin-clavulanate for E. coli, K.
aeruginosa (0.09 and 15 ␮M, respectively) (134). By combining pneumoniae, H. influenzae, S. aureus, and N. gonorrhoeae
BAL19764 or BAL30072 with clavulanate and a bridged strains producing plasmid-mediated ␤-lactamases (mostly
monobactam (all at a fixed concentration of 4 ␮g/ml), the TEM and SHV expressors) and for K. oxytoca, Proteus vulgaris,
MICs of the siderophore compounds were improved for a and P. mirabilis strains producing chromosomal ␤-lactamases.
range of Gram-negative bacteria (including P. aeruginosa) pro- Kinetic studies of BRL 42715 and NCTC 11561, TEM-1, and
ducing AmpCs as well as carbapenemases or overexpression of P99 showed rapid, irreversible inactivation with an inhibition
efflux systems (315, 316). However, the activity of carbapenems stoichiometry of 1:1, or turnover numbers of 0 (119).
is still superior to that of these new monobactams for many Initial studies to elucidate the BRL 42715 inactivation product
ESBL-producing isolates (e.g., K. pneumoniae CTX-M and using UV difference spectroscopy for both enzyme- and base-
SHV-5, E. coli, and E. cloacae) (38, 102, 173). The dual anti- catalyzed hydrolysis of the methylidene penem suggested the
bacterial/inhibitory action of these monobactam derivatives presence of a dihydrothiazepine rearrangement product (46, 250).
makes them important leads, and the further development of Accordingly, mass spectrometry studies demonstrated that the
the siderophore compound BAL30072 seems likely. methylidene penem inhibition pathway does not involve fragmen-
tation in the active site (119, 250, 406). When TEM-1, P99, and
the class A ␤-lactamase from B. cereus I were inactivated by BRL
ATMO Derivatives
42715, spectra showed a mass increases equivalent to the molec-
Certain cephems adopt catalytically incompetent conforma- ular masses of the inhibitor plus the enzyme (119). The crystal
tions in the active site, effectively “trapping” the enzyme. structure of the class C ␤-lactamase E. cloacae 908R in complex
Evidence from the crystal structure of an AmpC enzyme com- with BRL 42715 (PDB 1Y54) confirmed the formation of a stable
plexed with ceftazidime (PDB 1IEL) suggested that the pres- seven-membered thiazepine ring covalently attached to Ser64
ence of the 2-amino-4-thiazolyl methoxyimino (ATMO) R1 (263). Studies of both BRL 42715 and related penems support the
side group, which is common among extended-spectrum ceph- mechanism involving formation of an imine intermediate from
alosporins (e.g., cefotaxime and ceftazidime), destabilized the the acyl-enzyme, which then undergoes rearrangement, and even-
formation of the deacylation transition state (350). Synthesis of tually endo-trig cyclization to form the seven-membered thiaz-
inhibitors containing this ATMO side chain on a penicillin epine ring end product of inactivation for class A, C, and D
(Fig. 1, compound 15) and carbacephem backbone led to enzymes (Fig. 10) (250, 263, 294, 428).
AmpC IC50s of 900 nM and 80 nM, respectively (418). Subse- Crystallization of BLI-489, a related heterocyclic O C-6-substi-
quent examination of the crystal structure of the ATMO-pen- tuted penem with IC50s of 9.0 and 6.2 nM for SHV-1 and class C
icillin in complex with the AmpC (PDB 1LLB) showed that it GC1, respectively (see below), revealed the common seven-mem-
resembled the acyl-enzyme formed by ceftazidime. The bered thiazepine ring when complexed with the SHV-1 and GC1
ATMO groups were located in very similar positions, in both enzymes (PDB 1ONG and 1ONH, respectively) (294). Interest-
cases leading to steric hindrance between the deacylating water ingly, the orientation of the ring in SHV-1 and GC1 differed by
and the ␤-lactam ring nitrogen, accounting for inhibition. This 180° about the bond to the acylated serine. This rotameric pref-
design highlights the potential of incorporating destabilizing erence may be due to the relative “openness” of the class C
VOL. 23, 2010 ␤-LACTAMASE INHIBITORS 183

FIG. 10. Proposed reaction mechanisms for the inactivation of a serine ␤-lactamase by BRL 42715, showing formation of the seven-membered
thiazepine ring (A), and by LN-1-255, showing intermolecular capture by the pyridyl nitrogen leading to a bicyclic aromatic intermediate (B).
(Based on data from references 263, 294, 322, and 428.)

binding site at the bottom of the b3 ␤-sheet. Crystallization of a BRL 42715 derivatives. Derivatives of BRL 42715, including
tricyclic 6-methylidene derivative in complex with SHV-1 and tricyclic and bicyclic heterocyclic methylidene penems, have been
GC1 (PDB 1Q2P and 1Q2Q, respectively) also showed the com- the focus of a large body of recent structure-activity studies and
mon seven-membered thiazepine ring covalently attached to continue to show promising inhibition properties in investiga-
Ser70 and Ser64, respectively (429). The SHV-1/penem complex tional studies. The bicyclic heterocycles are synthesized as
was similar to what was observed with BLI-489, where the ring 6-methylidene penems attached to thiophene, imidazole, and pyr-
had R stereochemistry at the new C-7 moiety (294). However, in azole rings (428). Extension of the second heterocyclic ring yields
the GC1/penem complex, both the R and S chiral forms of the the tricyclic derivatives, which show improved stability in solution
intermediate were found. and increased lipophilicity in in vivo studies (429). As a group,
Taken together, these findings suggest that the 6-methylidene these compounds have proven to be potent inhibitors of class A,
penems’ acyl-enzyme stability is likely due to the low occupancy C, and D ␤-lactamases (25, 156, 335, 428, 441).
or disorder of the hydrolytic water molecule and, in part, to the A panel of one tricyclic and six bicyclic penem compounds
conjugation of the ester with the ring system. The nucleophilic displayed IC50s of 0.4 to 3.1 nM for TEM-1 (class A), 7.8 to 72
attack of the double bond at C-6 appears to be important to the nM for IMI-1 (class A), 1.5 to 4.8 nM for AmpC (class C), and
inhibition mechanism of the methylidene penems, and these 14 to 260 nM for a CcrA (class B) (441). Compared to tazobac-
novel inhibitors seem to share a similar inactivation mechanism tam IC50s, these penems were 100- to 56,000-fold more active
for different ␤-lactamase classes (429). against TEM-1 and the AmpC enzymes. Each of the seven
Most ␤-lactamases appear to follow this linear pathway to penems, at a constant concentration of 4 ␮g/ml, was combined
inactivation by the penem compounds, i.e., formation of an with piperacillin, which significantly lowered MICs in ESBL
acyl-enzyme and rearrangement to the stable dihydrothia- producers and restored susceptibility in piperacillin-resistant
zepine ring adduct (119, 250). However, some class A en- Enterobacter spp., Citrobacter spp., and Serratia spp. In a mouse
zymes, such as the ␤-lactamases from Staphylococcus albus model of infection caused by E. coli and Enterobacter aerogenes
(Staphylococcus epidermidis) and K. pneumoniae K1, exhibit class A (including ESBL) and C ␤-lactamase-producing organ-
more complex kinetics (46, 250). A branched pathway leads isms, piperacillin-penem combinations decreased the overall
to an acyl-enzyme hydrolysis product, which also rapidly 50% effective dose (ED50) of piperacillin from 1.6- to 8-fold
rearranges to the dihydrothiazepine species. (441). However, class B metallo-␤-lactamase producers were
While BRL 42715 and the structurally related investiga- not reported for susceptibility tests or the murine model, and
tional penem Syn 1012 showed promising in vitro kinetics they warrant further attention for clinical translation.
against a large spectrum of Gram-positive and Gram-negative Of the panel of tricyclic and bicyclic penem compounds
isolates, they were relatively unstable in human and mouse tested, the bicyclic BLI-489 was chosen as a lead compound to
plasma (342). Further, serum levels of BRL 42715 and Syn be combined with piperacillin (335). With 4 ␮g/ml of BLI-489,
1012 were undetectable 1 h after intravenous administration in 92% of non-piperacillin-susceptible clinical strains (including ex-
a rabbit model. Thus, the successful elements of BRL 42715 pressors of representative ␤-lactamases from all four classes) had
have been applied to additional drug development efforts di- piperacillin–BLI-489 MICs of ⱕ16 ␮g/ml, in contrast to 66% for
rected at improving in vivo stability (see below). piperacillin-tazobactam. Furthermore, the BLI-489 combination
184 DRAWZ AND BONOMO CLIN. MICROBIOL. REV.

demonstrated improved activity over piperacillin-tazobactam shows nM affinity for both the class A TEM-1 and class C E.
against ESBL- and AmpC-expressing strains, as well as against a cloacae 908R ␤-lactamases (431). The IC50s of clavulanate and
panel of staphylococcal and streptococcal isolates. Notably, BLI- LK-157 for TEM-1 were similar (0.030 ␮M and 0.055 ␮M,
489 did not improve the activity of piperacillin against the entero- respectively), but that of LK-157 was over 2000-fold better for
cocci and penicillin-intermediate and -resistant strains of S. pneu- E. cloacae AmpC (136.2 ␮M and 0.062 ␮M, respectively)
moniae (which may reflect that the piperacillin resistance (344). In combination with ampicillin, LK-157 (at 30 ␮g/ml)
mechanism is not ␤-lactamase mediated) (335). While these prac- restored susceptibility for the AmpC-overexpressing E. cloacae
tical studies help to bring closer the possible clinical introduction P99 strain in MIC testing (431). Against a large panel of
of these novel inhibitors, only a small number of the isolates were bacteria producing class A ESBLs (excepting KPC and
KPC or IRT producers. These KPC- and IRT-producing strains CTX-M) and class C ␤-lactamases, LK-157 in combination
also expressed up to three other ␤-lactamases, which complicates with cephalosporins showed improved potency compared to
the interpretation of activity against these enzymes. Thus, one clavulanate, tazobactam, and sulbactam (324). The lowest
cannot yet endorse the efficacy of this inhibitor combination for MICs (ranging from ⱕ0. 025 to 1.6 ␮g/ml) were achieved for
these relevant resistance threats. cefepime or cefpirome in combination with LK-157 at 4 ␮g/ml.
Two of these bicyclic investigational penems (Fig. 1, com- Despite these promising data with class A and C ␤-lactamase-
pounds 17 and 18), containing either a dihydropyrazolo[1,5- producing Gram-negative bacteria, LK-157 behaves as a sub-
c][1,3]thiazole or a dihydropyrazolo[5,1-c][1,4]thiazine moiety, strate with the carbapenem-hydrolyzing enzymes BcII and
are extremely effective inactivators of the class D OXA-1 ␤-lac- NMC-A (431). Further, while the inhibitor displays nM affinity
tamase, with nM affinity and low turnover values compared to for and rapidly inactivates the class D OXA-10 ␤-lactamase,
tazobactam (tn ⫽ 0 and 350, respectively) (25). Additionally, the acyl-enzyme is unstable and rapidly hydrolyzed (431).
these penems demonstrate efficacy with IR class A ␤-lactama- The crystal structure of LK-157 in complex with the E. clo-
ses. Against the SHV Arg244Ser variant, the penem inhibitors acae P99 (PDB 2Q9N), together with spectroscopic data for
showed significantly lower KI values than clavulanate and sul- reaction intermediates, suggests that after deacylation at the
bactam (penems 1 and 2, 1.0 to 4.4 ␮M; clavulanate and sul- active-site Ser64, the C-4 methoxy group is eliminated (344,
bactam, 63 and 240 ␮M, respectively) and extremely low turn- 431). Further, the catalytic water molecule presumed to be
over numbers (tn ⫽ 0, 50, and 100 for the penems, clavulanate, responsible for deacylation in class C enzymes is not observed
and sulbactam, respectively) (410). in the AmpC/LK-157 crystal structure. Rotation about the
Oxapenems. Oxapenems are derivatives of clavulanic acid, opened ␤-lactam ring after acylation likely leads to displace-
possessing an oxygen atom at penem position 1. Their synthesis ment of this water molecule and subsequent enzyme inhibition
was first described in the 1970s, and Cherry et al. introduced an (344). The C-10 ethyldiene group is probably not bulky enough
oxapenem that had improved in vitro activity over clavulanate to induce the conformational change seen with inhibition of
against the E. cloacae AmpC and staphylococcal ␤-lactamases class A enzymes by carbapenems, but resonance stabilization
(86). However, the compound was unstable and did not have of the acyl-enzyme through the acyl carbonyl and ethyldiene
activity in whole-cell assays. In 1993, Pfaendler et al. improved group may contribute to stability of the intermediate (255,
the stability of the compounds by adding bulky substituents at 344). Trinems are worthy of attention for the potential to serve
the C-2 position (339). The lead compound from this panel of as leads for additional structure-based inhibitor design, and
oxapenems, the zwitterionic AM-112 (Fig. 1, compound 19), Lek Pharmaceuticals continues to investigate LK-157. Devel-
has shown potent ␤-lactamase-inhibitory properties, particu- oped for parental use, the half-life (t1/2) of LK-157 in rats is
larly for class C and D enzymes (189, 190). Against class A similar to those of other ␤-lactams after intravenous adminis-
enzymes, IC50s were 10- to 20-fold higher for AM-112 com- tration (354). Additional studies have focused on developing
pared to clavulanate (190). However, the IC50s of AM-112 for an oral agent with improved permeability over LK-157, and
the class C enzymes from E. cloacae, S. marcescens, and P. while prodrug esters have good solubility, the intestinal per-
aeruginosa, as well as OXA-1 and OXA-5 from E. coli, were meability remains low.
1,000- to 100,000-fold lower than those of clavulanate. AM-112 1-␤-Methylcarbapenems. A panel of carbapenem deriva-
in combination with ceftazidime at 1:1 or 1:2 reduced ceftazi- tives bearing a methyl group at C-1 and various substituents at
dime MICs against class A ESBL producers and the class C C-2 were screened for inhibitory activity against the MBL
enzyme hyperproducers. For the class D enzyme producers IMP-1 (282). The study revealed a compound, J-110,441, that
and P. aeruginosa strains tested, AM-112 did not enhance the had potent inhibitory activity against several MBLs but also
activity of ceftazidime. The susceptibility results were similar low Ki values for class A TEM and class C E. cloacae enzymes
for AM-112’s protection of cefepime, ceftriaxone, and ce- (2.54 ␮M and 0.037 ␮M, respectively). Susceptibility testing of
foperazone (189). In a mouse sepsis model, ceftazidime pro- the AmpC-expressing E. cloacae demonstrated an MIC de-
tection was maintained against E. cloacae P99 and K. pneu- crease from 64 to 4 ␮g/ml with imipenem and J-110,411 at 4
moniae SHV-5 (190). The preclinical development of AM-112 ␮g/ml. Further work with these carbapenem derivatives has
has been led by Amura Ltd., and it warrants further examina- focused on their potential as MBL inhibitors and will be dis-
tion as a potential multi-class ␤-lactamase inhibitor. cussed below (see Inhibition of Metallo-␤-Lactamases).
Tricyclic carbapenems (trinems). Rational structure-based
design informed the synthesis of novel tricyclic carbapenems
Penicillin and Cephalosporin Sulfone Derivatives
built by the fusion of a cyclohexane ring onto a carbapenem
scaffold (90, 91). LK-157 (Fig. 1, compound 20) was derived by Following the success of sulbactam and tazobactam, medic-
introduction of a methoxy substituent at position C-4 and inal chemists have focused much effort on the development of
VOL. 23, 2010 ␤-LACTAMASE INHIBITORS 185

penicillin and cephalosporin sulfones with functionalities that ceftazidime and cefpirome against E. coli and K. pneumoniae
may improve inhibitor efficiency. One of the first series of expressing CTX-M, OXA, and CMY ␤-lactamases (63, 322).
investigational sulfones, the C-7 alkylidene cephems, had lead Meropenem MICs were lowered from 32 to 1 ␮g/ml with 4 ␮g/ml
compounds that were potent inhibitors of class C ␤-lactamases of LN-1-255 against S. marcescens possessing SME-1. The C-3
(65). Subsequent reports have explored the roles of C-2 and hydrophobic catechol moiety of this investigational compound
C-6 penam and C-3 cephem substituents. The findings for each appears to improve both its entry into cells via siderophore iron
will be discussed in the context of the derivative group. channels and its affinity for ␤-lactamase active sites (75).
C-2/C-3-substituted penicillin and cephalosporin sulfones. X-ray crystallography of LN-1-255 in complex with SHV-1
␤-Lactamase inhibitors with C-2 substitutions have shown ef- (PDB 3D4F) revealed that the inhibitor’s C-6 (heteroaryl)
ficacy against TEM- and SHV-type enzymes, including ESBLs alkylidene group plays an important role in the formation of a
(367, 422). The acrylonitrile derivative Ro 48-1220 (Fig. 1, planar bicyclic aromatic intermediate (322). The pyridyl nitro-
compound 21) achieves an IC50 for TEM-1 (0.08 ␮M) that is gen of the C-6 substituent acts as a base and promotes intramo-
comparable to that of clavulanate (0.10 ␮M), but the IC50 for lecular capture, leading to the formation of a bicyclic species
SHV-1 (0.27 ␮M) is slightly higher than that of clavulanate (Fig. 10B). The acyl-enzyme ester carbonyl of the intermediate
(0.05 ␮M) (422). Inhibition with Ro 48-1220 was as effective as is resonance stabilized by the conjugated ␲ system. Addition-
tazobactam for TEM-1 and SHV-1, with IC50s within 0.03 ␮M ally, the carbonyl group of the intermediate is positioned out-
for both enzymes. Most class C enzymes tested were inhibited side the oxyanion hole. The decreased deacylation rates of this
at lower concentrations of Ro 48-1220 than of tazobactam species are likely due to resonance stabilization and the loca-
(IC50 range approximately 2- to 30-fold lower) (367, 422). The tion of the carbonyl, at an increased distance from the hydro-
IC50s for X. maltophilia and Bacteroides fragilis class B enzymes lytic water and improperly positioned for nucleophilic attack
were 24 and 200 ␮M, in comparison to 4 and ⬎10 mM for (322).
tazobactam (367). Ro 48-1220 (at 4 ␮g/ml) performed well in By expanding the five-membered penicillin sulfone design to
susceptibility testing, protecting the activity of ceftriaxone and a six-membered cephalosporin sulfone, Buynak et al. devel-
ceftazidime against class C- and ESBL-producing organisms oped synthetic methodologies allowing modification of C-3
(422). Central to the potency of this sulfone inhibitor was the substituents (62, 64). The different functional groups intro-
low enzyme recovery rate compared to tazobactam, especially duced at C-3 had significant impact on whether the inhibitor
for class C ␤-lactamases from C. freundii and E. coli (367). UV was selective for either class A or class C ␤-lactamases, with
absorption spectroscopy and X-ray crystallography revealed some features, such as electronegativity, increasing inhibition
the identity of a linear enamine formed by elimination of the of both classes. DVR-II-41S (Fig. 1, compound 24) has both
SO2 group, ring opening, and rearrangement into a linear the C-7 (heteroaryl)alkylidene group of LN-1-255 and a C-3
conjugated system (367). This particularly stable intermediate vinyl substituent, similar to nitrocefin, an excellent substrate
presumably accounted for enhanced inhibitor potency. for nearly all classes of ␤-lactamases (62, 64). This cephalo-
The design of the C-2-substituted penicillin sulfone SA2-13 sporin sulfone demonstrated IC50s of 90 nM for class A TEM-1
(Fig. 1, compound 22) drew on the insight that the tazobactam and 10 nM for class C GC1. The crystal structure of GC1/
imine is the common intermediate, or “gatekeeper,” in both DVR-II-41S (PDB 1GA0) revealed a bicyclic aromatic system
permanent interaction and enzyme regeneration (58, 309, 453). reminiscent of the LN-1-255 intermediate (94). Additional sta-
If the more stable enamine, the trans-enamine, can be favored, bility results from the placement of the inhibitor’s anionic
then the reaction will be “trapped” in transient inhibition. In sulfinate group between Tyr105 and the acyl-enzyme bond,
fact, IR enzymes do exhibit decreased trans-enamine forma- likely blocking the approach of the deacylating water molecule.
tion, as in the case of the SHV Met69Val/Glu166Ala variant C-6-substituted penicillin sulfones. C-6-substituted penicil-
reacted with tazobactam and clavulanate (416). SA2-13 in- linates are nM to low ␮M inhibitors of serine ␤-lactamases,
cludes a negatively charged carboxyl group attached to a linker with the sulfone oxidation state at the penam thiazolidine
which is designed to stabilize the trans-enamine conformation sulfur showing improved affinities over the sulfide state (27, 61,
in close proximity to conserved active-site residues. Interest- 374, 375). The stereochemistry and specific functionality of the
ingly, the crystal structure of SA2-13 in complex with SHV-1 C-6 substituents can affect the specificity of inhibition. For
(PDB 2H5S) showed an additional salt bridge with Lys234 and example, ␤ isomer alkenyl derivatives, as opposed to ␣ isomer
hydrogen bonds with Ser130 and Thr235. Compared to SHV-1 derivatives, show improved IC50s for class B ␤-lactamases, and
inhibited by tazobactam, SA2-13 had a 10-fold-lower deacyla- derivatives with thiazole rings, particularly with fluorine,
tion rate, presumably mediated by the stabilization of the trans- amino, or hydroxyl groups, show potency for class A ␤-lacta-
enamine species (309). However, the dissociation constant for mases (374). Further, compounds with a mercaptomethyl
SA2-13 was 17-fold lower than that for tazobactam (0.1 versus group at C-6, as opposed to hydroxymethyl, are better inhibi-
1.7 ␮M, respectively), and there was 47% 24-hour recovery of tors of class B enzymes, while C-6-mercapto-penicillinates are
activity of the SA2-13/SHV-1 mixture versus complete inacti- relatively less active inhibitors of classes A, B, and C (61).
vation with tazobactam. The C-6-hydroxyakylpenicillinates have been employed as
The 6-alkylidene-2⬘-substituted penicillin sulfone LN-1-255 tools for studying the mechanism of ␤-lactamase hydrolysis and
(Fig. 1, compound 23), attains nM KIs for the class A SHV-1 and inhibition (143). The crystal structure of 6-␣-hydroxymeth-
ESBL SHV-2 and class D oxacillinase-, ESBL-, and even carbap- ylpenicillanate complexed to TEM-1 (PDB 1TEM) suggests
enemase-type OXA enzymes (63, 322) (S. M. Drawz et al., un- that while C-6-substituted penicillin sulfones are acylated in
published data). Moreover, LN-1-255 restores the activity of pip- the same way as ␤-lactam substrates, the hydroxymethyl group
eracillin against E. coli IR SHV enzyme producers and of at C-6 impedes the approach of the hydrolytic water and pro-
186 DRAWZ AND BONOMO CLIN. MICROBIOL. REV.

longs the lifetime of the acyl-enzyme species, effectively inhib-


iting the enzyme (254). However, the same compound acted as
a substrate for the class A NMC-A. Crystal structure determi-
nation of NMC-A complexed with 6-␣-[(1-hydroxy-1-methyl)
ethyl]penicillanate (PDB 1BUL) showed that the repositioning
of residue Asn132 in NMC-A, compared to TEM-1, enlarged
the substrate binding cavity and allowed Asn132 to make new
hydrogen bonds with the C-6 substituent (274). These interac-
tions permit approach by the hydrolytic water and facilitate
deacylation of the acyl-enzyme intermediate for the 6-␣-hy-
droxymethylpenicillanate and C-6-substituted carbapenems
such as imipenem in NMC-A. Upon introduction of a bulkier
C-6 hydroxypropyl group, deacylation was impeded, in a mech-
anism very similar to that for the inhibition of TEM-1 by
FIG. 11. Scheme illustrating the interactions of a serine ␤-lacta-
imipenem (407). mase with a cefotaxime substrate (A) compared to a cefotaxiime-like
Generalization of these and other findings leads to the con- boronic acid TSA (B). Notice that the reaction with the inhibitor is
clusion that when the hydrolytic water molecule approaches reversible.
the acyl-enzyme from the “␣ direction,” as with class A en-
zymes, ␣-hydroxyalkylpenicillinates are effective inhibitors.
Conversely, ␤-hydroxyalkylpenicillinates inhibit the “␤ ap- for the E. coli AmpC of 27 nM.
proach” in class C enzymes (143). A group at Wyeth Research In addition to their demonstrated potential as effective in-
found that a 6-␤-hydroxymethyl penicillin sulfone has the best hibitors, these compounds have been used to probe the impor-
activity against both class A and C enzymes, with IC50s of 8 nM tant interactions between the inhibitor and the enzyme active
for TEM-1 and 1.2 ␮M for AmpC (26). Presumably, the in- site, thereby informing the design of future TSAs. Using the
hibitor worked like tazobactam or sulbactam in class A en- individual crystal structures for nine TSAs and four ␤-lactams
zymes, but the specific crowding of the ␤-face of the ester complexed with the E. coli AmpC ␤-lactamase, computational
prevents approach of the hydrolytic water in class C ␤-lacta- analyses have generated consensus binding pockets in the
mases (51). The class D OXA-10 enzyme showed inhibition by AmpC active site (352). The functionalities recognized by cer-
both C-6 ␣- and ␤-hydroxyisopropylpenicillinates (252). While tain residues led to the development of new glycylboronic acid
only the ␤-isomer could be crystallized with OXA-10 (PDB TSAs, including a compound containing the R1 side chain of
1K54), this structure and computational models of the ␣-iso- cephalothin. One of these cephalothin analogs achieved a Ki of
mer suggest related, but different, mechanisms of inactivation 1 nM for the E. coli AmpC (Fig. 1, compound 25), a 300-fold
based on the C-6 stereochemistry of the inhibitor. However, improvement over the parent compound, which lacked the
both mechanisms involved displacing the hydrolytic water nec- meta-carboxylphenyl substituent (272). The substituent was de-
essary for deacylation. signed to resemble both the dihydrothiazine ring and the C-4
carboxylate of the cephalosporin nucleus. In the crystal struc-
ture of this cephalothin meta-carboxyphenyl analog complexed
Non-␤-Lactam Inhibitors with an AmpC (PDB 1MY8), the inhibitor carboxylate inter-
acted with Asn289, a residue not previously implicated in struc-
Boronic acid transition state analogs (TSAs). In the 1970s, tural or modeling studies as interacting with the cephalosporin’s
boronic acid compounds were described as forming reversible, C-4 carboxylate. Asn289 is not a highly conserved residue
dative covalent bonds with serine proteases and inhibiting among class C ␤-lactamases, but the low Ki is maintained when
these enzymes by assuming tetrahedral reaction intermediates different AmpCs with other residues at position 289 are tested
(30, 230). Kiener and Waley then applied the chemistry to the (e.g., Kis of 29 nM for E. cloacae P99 and 11 nM for A.
class A ␤-lactamase from B. cereus and showed that boronic baumannii (107a, 241, 371). The versatility of this inhibitor may
acid derivatives, as well as the phenyl and m-aminophenyl reflect an important flexibility in AmpC active sites that could
derivatives, were mM inhibitors of the enzyme (201). Further affect the recognition of both substrates and inhibitors.
study by other groups demonstrated that these compounds, High-affinity inhibitors will be clinically useful only if they
which lack the ␤-lactam motif, form adducts that resemble the can effectively permeate the bacterial cell membrane and re-
geometry of the tetrahedral transition state of the ␤-lactamase store susceptibility to partner ␤-lactams. The boronic acid de-
hydrolytic reaction (22, 82, 95) (Fig. 11). By modifying these rivatives enter both S. aureus producing class A ␤-lactamases
transition state analogs to contain the R1 side chains of natural and Enterobacteriaceae such as E. coli, C. freundii, E. cloacae,
substrates, affinities in the nM range for both class A TEM-1 and P. aeruginosa expressing class C ␤-lactamases, and they
and ESBL-type SHV and class C enzymes have been deter- effectively protect ampicillin and ceftazidime (70, 272, 349,
mined (70, 392, 394, 412). Weston et al. used the crystallo- 430, 436). Boronates have not yet been developed for clinical
graphic structure of E. coli AmpC ␤-lactamase in complex with use in combination with a ␤-lactam, in part because of con-
m-aminophenylboronic acid (PDB 3BLS) as a model for de- cerns about the toxicity of boron. The safety of boron-contain-
signing boronic acid-based inhibitors specifically for the class C ing compounds is currently being addressed by clinical studies
active site (425, 443). The compound showing the greatest sponsored by Anacor Pharmaceuticals. Anacor develops boron
affinity was benzo[b]thiophene-2-boronic acid, which had a Ki compounds for medical uses and has current trials for boron-
VOL. 23, 2010 ␤-LACTAMASE INHIBITORS 187

based topical treatments of fungal, bacterial, and anti-inflam- lin-tazobactam for class C enzyme-producing Enterobacteri-
matory diseases, as well as research stage data on a systemic aceae, including both ceftazidime-resistant and -susceptible
antibacterial agent. strains of C. freundii and E. cloacae. The NXL104 concen-
Phosphonates. Phosphonate monoester derivatives can tration of 4 ␮g/ml restored MICs into the susceptible range
acylate the active-site serines of class A, C, and D ␤-lactama- for a collection of clinical P. aeruginosa isolates (21% non-
ses, leading to effective inhibition (4, 223, 243, 244, 353). These susceptible to ceftazidime versus 6% nonsusceptible to
compounds exhibit branched kinetic pathways, in some cases ceftazidime-NXL104) (380). In in vivo murine studies, in-
reflecting inhibition by the products that are formed. Acyl cluding models of sepsis with CTX-M Enterobacteriaceae
phosphonates inhibit class C enzymes, while cyclic acyl phos- producers, pneumonia with AmpC and SHV-11 K. pneu-
phonates inhibit both class A and C ␤-lactamases. For diacyl moniae producers, and thigh infection and sepsis with
phosphonates, hydrophobic substituents decrease the inhibi- KPC-2, TEM-1, and SHV-11 K. pneumoniae producers, the
tor’s Ki value, and the acylation rates of these compounds ceftazidime-NXL104 combination has demonstrated prom-
follow the order class D ⬎ class C ⬎ class A (243, 244). ising therapeutic efficacy (221, 259, 442).
Majumdar et al. posit that this relative inhibitor efficiency NXL104 also enhanced the activity of cefotaxime in a study
reflects the general hydrophobicity of the enzyme active sites of Enterobacteriaceae expressing CTX-M, KPC, or OXA-48,
and that rational design of diacyl phosphate side chains could where 4 ␮g/ml of the inhibitor lowered MICs to ⱕ1 ␮g/ml. For
lead to nM inhibition of all serine ␤-lactamase classes. isolates with ertapenem resistance due to combinations of
Crystal structures with these phosphonic acids have also offered ␤-lactamases (ESBLs or AmpCs) and impermeability, the
important insight into the reaction mechanisms of class C en- MICs were still ⱕ2 ␮g/ml. While cefotaxime-NXL104 lowered
zymes (297). The ability of the E. cloacae GC1 ␤-lactamase to the MIC of one S. marcescens isolate expressing SME-1 from
hydrolyze extended-spectrum cephalosporins was elucidated by 0.5 to 0.12 ␮g/ml, MICs for K. pneumoniae and E. coli express-
trapping of the enzyme in complex with a phosphonate cefo- ing class B IMP and VIM metallo-␤-lactamases were not low-
taxime transition state analog (297). A three-residue insertion ered by either cephalosporin with NXL104.
into the ⍀-loop of the enzyme, compared to the C. freundii ␤-lac- Two recent studies examined more closely the ability of
tamase/transition state analog complex, allowed for a better fit of NXL104 to restore susceptibility to ␤-lactams for KPC en-
cefotaxime’s side chain in the enzyme active site and facilitated zyme-producing clinical isolates (116, 390). Stachyra et al.
the activation of the hydrolytic water molecule. The clinical po- demonstrated that NXL104 at 4 ␮g/ml restored susceptibility
tential of phosphonates has been limited by their poor stability in of six KPC-2 or -3 producers to piperacillin, ceftazidime, ceftri-
aqueous solution and susceptibility to phosphodiesterases. axone, and imipenem (390). Endimiani et al. tested NXL104 at
NXL104. NXL104 (also known as AVE1330A) (Fig. 1, various concentrations (1, 2, and 4 ␮g/ml) against a panel of 42
compound 26) is a bridged diazabicyclo[3.2.1]octanone non- K. pneumoniae isolates with ⱖ3 bla genes each and MIC90s of
␤-lactam inhibitor with very promising activity against class ⱖ128 ␮g/ml for all ␤-lactams tested (116). All of these K.
A, C, and D ␤-lactamases (31, 116, 240, 390). The majority pneumoniae strains were susceptible to NXL104 at 2 ␮g/ml
of the published work on NXL104 describes MIC testing, with cefotaxime, ceftazidime, cefepime, and aztreonam and at
although IC50s of 8, 38, and 80 nM have been reported for 4 ␮g/ml with piperacillin. Collectively, these data suggest that
class A TEM-1 and KPC-2 and class C P99 enzymes, respec- NXL104, in combination with several cephalosporins, aztreo-
tively (31, 390). A remarkable property of NXL104 is the nam, and even imipenem, may be an excellent candidate for
prolonged deacylation rate. The 50% enzyme recovery time restoring susceptibility to difficult-to-treat emerging carbapen-
point was 7 days for both TEM-1 and P99 (compared to 7 emase- and ESBL-producing organisms.
min for TEM-1 and clavulanate and 290 min for P99 and NXL104, manufactured by Novexel Inc., is currently in
tazobactam), suggesting the presence of a very stable and phase II clinical trials in combination with ceftazidime for
long-lived intermediate species (31). The formation of a treatment of complicated urinary tract and intra-abdominal
covalent complex has been supported by an ESI-MS study of infections (www.clinicaltrials.gov). Additionally, Forest Labo-
TEM-1 and NXL104 (391). Currently, the only reported ratories Inc. intends to begin phase I clinical trials with
crystal structure of the inhibitor is in complex with CTX-M- NXL104 in combination with the novel broad-spectrum ceph-
15, and it shows opening of the NXL104 ring upon acylation, alosporin ceftaroline (which is currently in phase III trials as
interaction with several conserved active-site residues, and monotherapy for complicated skin infections and community-
displacement of the putative deacylation water by approxi- acquired pneumonia). The results of clinical trials are awaited.
mately 1 Å (104). Hydroxamates. O-Aryloxycarbonyl hydroxamate is an irre-
With ceftazidime as a partner ␤-lactam in a 4:1 ratio versible inhibitor of the class C E. cloacae P99 ␤-lactamase that
(ceftazidime at 4 ␮g/ml and NXL104 at 1 ␮g/ml), NXL104 utilizes a mechanism of action which differs from those of the
lowered the MICs for a series of Enterobacteriaceae strains currently available inhibitors (Fig. 1, compound 27) (330, 450).
at least 8-fold compared to those of ceftazidime alone, to ⱕ4 The proposed inhibition mechanism involves rate-limiting
␮g/ml for E. coli, K. pneumoniae, Citrobacter diversus, and P. acylation of the hydroxamate. However, the inhibitor is stabi-
mirabilis strains and to 2 ␮g/ml for E. cloacae and Serratia lized not by the traditional oxyanion hole comprised of the
sp. strains (31). Against class A ␤-lactamase producers (pri- backbone nitrogens of Ser64 and Ser318 but rather by the side
marily TEM and SHV types), the ceftazidime-NXL104 com- chains of Tyr150 and Lys315 (330). As with clavulanate and
bination was comparable to ceftazidime-clavulanate but tazobactam, the acyl-enzyme proceeds to either hydrolysis or
superior to amoxicillin-clavulanate and piperacillin-tazobac- inactivation. The inactivation mechanism leads to aminolysis of
tam. Ceftazidime-NXL104 was more potent than piperacil- Lys315 and a covalent cross-linking of Ser64 and Lys315, as
188 DRAWZ AND BONOMO CLIN. MICROBIOL. REV.

carboxylate yielded a compound with a 26-fold-improved Ki (1


␮M) (413). In MIC testing, the inhibitor restored the suscep-
tibility to ceftazidime (at a 1:1 ratio) in the AmpC producers E.
cloacae and C. freundii (but not P. aeruginosa) and, in contrast
to clavulanate, did not upregulate ␤-lactamase expression in an
inducible strain of E. cloacae.
␤-Sultams are the sulfonyl analogs of ␤-lactams, and N-acyl
␤-sultams (Fig. 1, compound 29) inactivate the P99 enzyme (318,
419). ESI-MS studies suggest that the ␤-sultams sulfonylate the
serine residue to form a sulfonate ester. Elimination of the sul-
fonate anion leads to C-O bond fission and formation of a dehy-
droalanine at the previous Ser64 residue (419). These compounds
do not show inhibition of class B MBLs, and there are currently
no studies examining their activities in class A or D enzymes.
BLIP is a 165-amino-acid protein isolated from the same
organism that produces clavulanic acid, Streptomyces clavulig-
erus (105). Studies of BLIP’s inhibitory activity have revealed
affinities for class A enzymes that vary from pM to ␮M (the
enzymes with the highest affinities for BLIP are KPC-2 and the
P. vulgaris ␤-lactamases) (147, 393). Many of the individual
FIG. 12. Molecular representation of cross-linked active-site resi- class A residues which contribute to BLIP binding have been
dues Ser64 and Lys315 (shown in yellow) formed after aminolysis of an
O-aryloxycarbonyl hydroxamate inhibitor in E. cloacae P99 ␤-lacta-
identified through crystallography and mutagenesis (363, 456,
mase. (Based on PDB 2P9V and data from references 330 and 450.) 460). While the clinical viability of a peptide inhibitor based on
this protein is limited because of protein degradation in vivo,
the high-affinity interactions offer direction for the design of
seen in the crystal structure of the inhibited P99 (Fig. 12) (PDB novel ␤-lactamase inhibitors.
2P9V) (450). TEM-2 and OXA-1 ␤-lactamases were also in- The mixture of vanadate and catechol compounds in aque-
hibited by O-aryloxycarbonyl hydroxamates, and the MBL ous solution produces vanadate-catechol complexes which can
GIM-1 was inhibited by a “reverse” hydroxamate (hydroxy- inhibit class A TEM-2, class C P99, and class D OXA-1 ␤-lac-
lamino replacing hydroxylamine) conjugated to a cephalospo- tamases with apparent Kis of 62, 0.58, and 1.5 ␮M, respectively
rin nucleus (131, 330). Additional mechanistic studies of hy- (5). A crystal structure with the complex has not been attained,
droxamates and their derivatives, as well as in vitro activity
but molecular modeling suggests that the active-site serine is
data, will be of much interest.
bound to a penta- or hexa-coordinated vanadium. Vandate-
Other non-␤-lactam inhibitors. Work by Conrath et al. ex-
catechols represent another group of compounds that are use-
plored the ␤-lactamase-inhibitory potential of soluble frag-
ful as tools for novel inhibitory active-site interactions, but lack
ments derived from the variable region of heavy-chain drom-
edary antibodies (89). IC50s of 35 ␮M and 300 ␮M were clear clinical potential.
obtained for TEM-1 and the B. cereus MBL BcII, but the
development of this strategy would require innovative tech-
niques such as increasing the permeativity of these 15-kDa
INHIBITION OF METALLO-␤-LACTAMASES
polypeptides to outer membranes of Gram negative ␤-lacta-
mase producers. We note that designing and/or discovering inhibitors to
Inhibitors with a sulfonamide core and varied substituents MBLs presents special challenges. The diversity of class B
were developed by structure-based optimization (351, 413). enzyme subclasses (i.e., B1, B2, and B3) and the mechanistic
This non-␤-lactam design approach is intended to avoid both complexities regarding the role of one or two Zn2⫹ ions have
hydrolysis by ␤-lactamases and the upregulation of ␤-lactama-
impeded effective inhibitor design. In addition, indiscrimi-
ses induced by some ␤-lactam-based inhibitors. Starting from a
nately inhibiting or chelating metal ions can have untoward
map of binding “hot spots” developed from crystal structures
effects on natural or endogenous metalloenzymes. For exam-
of 13 different ligands in complex with the E. coli AmpC ␤-lac-
ple, similarities between the active-site architectures of MBLs
tamase, 200,000 commercially available small molecules were
docked into the active site of the enzyme by computer simu- and essential mammalian enzymes (e.g., human glyoxalase II)
lation (351, 352). Based on these results, lead compounds were complicate the design of an inhibitor that would be safe for
screened in vitro, and the molecule with the highest affinity (26 human use (97). As a class, the MBLs are resistant to all the
␮M) was crystallized in complex with the AmpC (PDB 1L2S) mechanism-based inhibitors of the serine enzymes (the inhib-
(Fig. 1, compound 28). The structure closely resembled the itory activity of the methylidene penems against class B ␤-lac-
prediction of the docking simulation, revealing the plasticity of tamases may be an important exception), and few investiga-
the enzyme active site to recognize functional groups different tional inhibitors demonstrate sub-␮M efficacy (335, 441). Here,
from ␤-lactams. This catalyzed the optimization of a novel we provide a brief survey of the more promising agents in
inhibitor, and replacement of the chloride substituent with a development.
VOL. 23, 2010 ␤-LACTAMASE INHIBITORS 189

Thiol Derivatives that the tetrazole portion of the molecule interacts directly
with the active-site Zn2⫹ ions (414). IC50 and Ki values in
Due to catalytic dependence on Zn2⫹ ions, the metalloen-
the low ␮M range were observed, and the incorporation of
zymes are inhibited by thiols or chelating agents that have an
substituents on the phenyl groups and their interactions with
affinity for the hydrophobic pocket of the enzyme active site
specific residues were essential to the strong binding of the
and bind to, or interfere with, the bonding network between
biphenyl tetrazoles (414). These inhibitors do appear to
the hydrolytic water and the Zn2⫹ ions (170, 227, 312). Thiol
enter cells, as the addition of imipenem or penicillin G to
derivatives, such as thiomandelic acids, typically show low-␮M
biphenyl tetrazole compounds increased inhibition zones for
Ki inhibition of B1 (e.g., BcII, IMP-1, and VIM-2) and B3 (e.g.,
an imipenem-resistant B. fragilis isolate (414).
L1) subclasses but over 100-fold-higher Ki values for the sub-
class B2 CphA MBL (268). Structural analysis of thiol-based
inhibitors complexed with the three MBL subclasses suggests Carbapenem Analogs
that the thiol group chelates the Zn2⫹ ions for subclasses B1
and B3 (227). For example, the crystal structure of IMP-1 from 1-␤-Methylcarbapenems with cyclic C-2 substituents, such as
P. aeruginosa bound to a thiol inhibitor (Fig. 1, compound 30) J-110,441, achieve sub-␮M Kis for the IMP-1, CcrA, LI, and
(PDB 1DD6) showed interactions with active-site residues, BcII MBLs as well as representative class A and C enzymes
allowing the thiol of the inhibitor to bridge the two catalytic (282). For several IMP-1-producing S. marcescens and P.
Zn2⫹ molecules and displace the bridging water important for aeruginosa isolates, imipenem MICs were lowered to 4 ␮g/ml
hydrolysis (88). A similar inhibition mechanism was resolved by the addition of J-110,441. The further development of
from the crystal structure of the B1 VIM-2 complexed to a J-111,225 included a trans-3,5-disubstituted pyrrolidinylthio
mercaptocarboxylate compound (452). However, this binding substituent at the C-2 position (Fig. 1, compound 33) (281).
mode was not observed in the crystal structure of the thiolate J-111,225 maintained inhibitory activity against IMP-1 (Ki of
D-captopril bound to the subclass B2 CphA from A. hydrophila 0.18 ␮M) and synergy with imipenem but had moderately
(227). Instead, the inhibitor’s main interactions with the en- improved MICs as a single agent against IMP-1-expressing S.
zyme were via the thiolate carboxylate group (227). marcescens and P. aeruginosa (MIC range of 4 to 32 ␮g/ml).
Mass spectrometric data suggest that mercaptoacetic esters However, J-111,225 acted as a substrate for other chromo-
may act as mechanism-based inhibitors for BcII by generating somally encoded class B MBLs, including CcrA, L1, and BcII.
mercaptoacetic acid in situ and forming a disulfide linkage with
a cysteine residue in the enzyme active site (327). However, as
Tricyclic Natural Products
with other thiols, the IC50s of these mercaptoacetic esters
varied from low ␮M to mM across MBL subclasses. Natural product screening for inhibitors to BcII led to the
discovery of extracts from Chaetomium funicola which demon-
strated IC50s ranging from 389 to 0.3 ␮M for BcII, P. aerugi-
Pyridine Dicarboxylates
nosa IMP-1, and B. fragilis CfiA MBLs (329). The “lead com-
Recent work demonstrates that pyridine carboxylates, in pound” of the three tricyclic compounds, SB238569 (Fig. 1,
particular 2-picolinic and pyridine-2,4-dicarboxylic acids (Fig. compound 34), also showed low Ki values for the same en-
1, compound 31), are competitive inhibitors of CphA (a sub- zymes (79, 17, and 3.4 ␮M, respectively). In combination with
class B2 MBL), with Ki values of 5.7 and 4.5 ␮M, respectively meropenem, 8 ␮g/ml of SB238569 restored susceptibility to B.
(170). The pyridine carboxylate derivatives showed little inhib- fragilis CfiA. The crystal structure of CfiA and SB236050 (PDB
itory activity for B1 and B3 MBLs. In the crystal structure of 1KR3) identified important polar (Lys184, Asn194, and
CphA/pyridine-2,4-dicarboxylic acid (PDB 2GKL), the cata- His162) and ring-stacking (Trp49) interactions which likely
lytic Zn2⫹ ion is coordinated by the nitrogen and one of the contribute to the inhibitory activity. Comparison of this struc-
carboxylate oxygens of the inhibitor (170). The Zn2⫹ ion is also ture with that of IMP-1/mercaptocarboxylate (PDB 1DD6)
engaged in electrostatic interactions with residues Asp120, suggests that the binding site created by the loop preceding
Cys221, and His263. Hydrophobic interactions with Asn233, Lys184 is shorter in IMP-1 and may leave a larger portion of
Trp87, and Val67 help to further stabilize the inhibitor in the the compound exposed to solvent (88). The decreased inhibi-
active site. The larger and more flexible active sites of subclass tory activity of the tricyclic inhibitor for S. maltophilia L1 may
B1 and B3 MBLs may not permit comparable stabilizing con- also be explained by active-site variations, including differences
tacts with the pyridine carboxylates, offering an explanation for in the positions of the Lys184 and Asn194 equivalents.
the B2 specificity.

Succinate Derivatives
Trifluoromethyl Ketones and Alcohols
The IMP-1 enzyme is encoded by a transferable blaIMP-1
Reactive trifluoromethyl ketones and alcohols, conju- gene and represents an increasing resistance threat, and thus
gated to a penicillin-like phenyl side chain, effectively inhibit the development of inhibitors for this target is a priority. Suc-
MBLs from B. cereus, P. aeruginosa, S. maltophilia, and A. cinic acid compounds have been screened as IMP-1 inhibitors,
hydrophila in vitro, presumably by binding the active-site and several derivatives have restored meropenem susceptibility
Zn2⫹ ion (433, 434). The crystal structure of a biphenyl of IMP-1-expressing E. coli (269, 415). Compound 35 (Fig. 1)
tetrazole inhibitor, L-159,061 (Fig. 1, compound 32), in belongs to a series of 2,3-(S,S)-disubstituted succinic acids
complex with the B. fragilis enzyme (PDB 1A8T) indicates which inhibit IMP-1 with nM IC50s (415). Crystal structure
190 DRAWZ AND BONOMO CLIN. MICROBIOL. REV.

analysis revealed that the inhibitors’ carboxylate groups inter- resemble the natural cephalosporin substrates (272). A guide-
act with both Zn2⫹ ions and active-site residues while displac- line for inhibitor design may be “penicillin analogs for penicil-
ing the bridging water (PDB 1JJE and 1JJT) (415). Disubsti- linases, and cephalosporin analogs for cephalosporinases.”
tuted succinic acids have also been described as inhibitors of This principle also ensures that the inhibition mechanism fol-
the L1 MBL and display binding modes similar to those in lows the substrate mechanism—save for hydrolysis. While
IMP-1 (284, 303). ␤-lactamases continue to develop resistance to all developed
IMP-1 is also inhibited by N-arylsulfonyl hydrazone com- ␤-lactams, designing inhibitors that follow the hydrolytic mech-
pounds, with IC50s as low as 1.6 ␮M (Fig. 1, compound 36) anism may help to thwart the emergence of resistant isolates,
(384). Structure-activity relationship studies revealed that as the enzyme must find novel ways to preserve function but
bulky aromatic substituents on both sides of the sulfonyl hy- evade inhibition. We hasten to add that this approach may not
drazone backbone improved the activity of these inhibitors. be practical in all cases.
Liénard et al. demonstrated the ability of dynamic combina- (iii) Stabilizing interactions in the active site. Stabilizing or
torial mass spectrometry (DCMS) to identify potent BcII in- prolonging the acyl-enzyme intermediate is central to success-
hibitor leads (228). A meso-2,3-dimer-captosuccinic acid was ful inactivation by the currently available class A inhibitors (81,
bound to the MBL active site via one thiol group, while the 396). Promising novel inhibitors, such as the methylidene pen-
second thiol group was linked in a disulfide bond to a thiol- ems (e.g., BRL 42715) and LN-1-255 are designed to capitalize
containing compound from a screening library. ESI-MS re- on and enhance this stabilization. One approach is to prolong
vealed which compounds bound the enzyme, and subsequent the acylation step (k2). Alternatively, acyl-enzyme intermedi-
structure optimization led to the development of a mercapto- ates can be stabilized by multiple interactions in the active site,
carboxylate that inhibits BcII with a Ki of 740 nM (Fig. 1, as observed for SHV-1 with tazobactam and SA2-13 (308, 309).
compound 37). The inhibition of NMC-A by the monobactam derivative 13
(Fig. 1) is enhanced by the incorporation of chemical compo-
nents that confer structural flexibility to the inhibitor, allowing
C-6-Mercaptomethyl Penicillinates
the enzyme-substrate species to adopt multiple conformations
Penicillin derivatives with C-6-mercaptomethyl substituents and trap the enzyme in energy minima unfavorable for hydro-
(Fig. 1, compound 38) show promising kinetics against class A, lysis (273, 308, 445). Another viable approach is to design
B, and C ␤-lactamases (IC50s of 6.8, 0.10, and 10.5 ␮M, re- inhibitors that induce conformational changes in the enzyme
spectively, against representative enzymes) (61). These peni- or displace key water molecules important for hydrolysis, such
cillinates, in combination with piperacillin, lowered MICs as seen with the large seven-membered intermediate formed
against MBL-producing strains of P. aeruginosa. Building in- from the methylidene penems (60, 294, 429).
hibitors on the backbone of the natural substrate for all ␤-lac- (iv) Reaction chemistry that slows deacylation and favors
tamases, the penicillin nucleus, may help bridge the gap be- inactivation over inhibitor hydrolysis. Features of an inhibitor
tween these diverse enzyme classes. which slow deacylation and drive toward terminal inactivation,
such as acyl-enzyme stabilization (see above) or the presence
of a good leaving group at C-1, contribute to effective inhibi-
LESSONS LEARNED tion. Inhibitors that can delay deacylation for longer than 15 to
We dedicate the penultimate section of this review to sum- 20 min (koff rate of ⱕ0.001 s⫺1) may outlast the bacterial
marizing the “lessons learned” from our consideration of the generation time, allowing the partner ␤-lactam time to disrupt
␤-lactamase inhibitors. From the perspective of clinicians, we the cell wall synthesis necessary for cell division (396, 410). For
propose that several important features must be regarded in example, the bridged monobactams delay deacylation by lim-
order to optimize the development of ␤-lactamase inactivators. iting the C-3–C-4 rotation necessary for approach of the hy-
(i) High affinity for the active site of the target ␤-lactamase. drolytic water in class C enzymes (152, 238). Additionally,
It is axiomatic that in order for an inhibitor to work efficiently, relatively minor changes introduced by the acylation of mero-
it must exhibit high affinity for its target. This principle is penem by SHV-1, including minor displacements to active-site
emphasized by the development of clavulanate resistance in residues (overall root mean square deviation [RMSD] of 0.29
TEM-1 and SHV-1 ␤-lactamases mediated by substitution of Å) and a restructured hydrogen bonding network, lead to per-
amino acid residues 69, 130, 244, and 276, leading to increased sistent complex formation (295).
inhibitor KIs and/or IC50s (73, 153, 378, 411). Particularly (v) Rapid cell penetration. ␤-Lactamases are typically found
promising are those inhibitors that maintain high affinities in the periplasmic space of Gram-negative bacteria, and the
across different groups of ␤-lactamases, such as the C-2-sub- rapidity with which inhibitors can access their target is a pre-
stituted sulfone LN-1-255, the methylidene penems, and phos- requisite for successful inhibition. Certain ␤-lactams are zwit-
phonates which achieve nM inhibition of class A, C, and D (4, terionic and rapidly penetrate the bacterial outer membrane
25, 63, 245, 322, 441) (Drawz et al., unpublished data). (e.g., cefepime). The same principle extends to the inhibitors;
(ii) Mimicking the “natural substrate.” The available inhib- e.g., the enhanced cell entry of LN-1-255 and the novel
itors are effective primarily against class A serine ␤-lactamases, monobactams BAL19764 and BAL30072 is likely due to up-
and the structures of class A ␤-lactamase inhibitors are similar take through siderophore channels (75, 316). Attention must
to those of penicillin, bearing a fused four- or five-membered also be paid to designing “permeable drugs” that are not hin-
ring system. Similarly, data on transition state analogs suggest dered as they pass through porin channels or quickly returned
that class C ␤-lactamases have high affinity for inhibitors that into the medium by multidrug efflux pumps, an important
VOL. 23, 2010 ␤-LACTAMASE INHIBITORS 191

resistance determinant independent of ␤-lactamases (see of the key structural features of these diverse proteins, keeping
above) (287). in mind that mechanism-based inhibitors may engage in
(vi) Low propensity to induce ␤-lactamase production. Suc- unique reaction chemistry with ␤-lactamases. Compounds with
cessful inhibitors need to avoid inducing expression of the the chemical features of the “ideal inhibitor,” such as en-
enzymes that they are designed to inhibit, particularly among hanced permeability through the cell membrane, affinity for
AmpC ␤-lactamases. Examples of this approach include the the active site of the ␤-lactamase, formation of intermediates
non-␤-lactam inhibitors with sulfonamide cores designed to that “trap” the enzyme, displacement of critical water mole-
evade hydrolysis as well as to avoid induction of AmpC enzyme cules, and prolonged time to enzyme recovery, many of which
expression (413). are illustrated by the investigational complexes highlighted
(vii) Identification of measureable biological correlates of herein, may have a significant impact on the development of
␤-lactamase inhibition in the cell. Regardless of how attractive “second-generation” inhibitors that target resistant ␤-lactamases.
an inhibitor’s laboratory characteristics may be (e.g., nM Ki We also acknowledge the magnitude of this challenge. In 30
values, low IC50s, high kinacts, or low MICs), a successful in- years of targeted research on ␤-lactamase inactivation, new in-
hibitor must prove its worth in the clinical setting. In vivo hibitors have been only slowly introduced into clinical trials.
efficacy requires an integration of many complex features, in- There have been many starts, but few agents cross the finish line.
cluding those listed above, partnership with an appropriate This is a testament to the versatility and complexity of ␤-lactama-
␤-lactam, sufficient serum and periplasm concentrations, and ses and the incredible evolutionary ability of the organisms har-
factors less well understood (e.g., serum protein binding and boring them. Is the perfect ␤-lactamase inhibitor an unattainable
individual clearance rates). Many microbiological properties goal? Perhaps. Yet, we anticipate with excitement the achieve-
(time kill, mutant prevention concentration, and synergy) are ments in the next 3 decades and suspect that better chemistries
important parameters that need to be considered. There is no and combinations will ultimately be found.
certain consensus on how best to identify and measure these
attributes in the early research on candidate inhibitors. Previ-
ACKNOWLEDGMENTS
ous studies with ␤-lactam antibiotics have demonstrated that
MICs may be poorly correlated with the sensitivity of PBPs for This work was supported in part by the Department of Veterans
inactivation (213). Finding reliable biological correlates of in- Affairs Merit Review Program and National Institutes of Health grant
1RO1 A1063517-01. R.A.B. is also supported by the Veterans Inte-
hibition is a substantial and relevant challenge, and our current grated Service Network (VISN) 10 Geriatric Research, Education, and
methodological outcome measurements may benefit from re- Clinical Center (GRECC). S.M.D. was supported in part by NIH grant
examination in this vein. T32 GM07250 and the Case Medical Scientist Training Program.
(viii) In the meantime, use what we have. While scientists We thank the four anonymous reviewers and A. Endimiani and
M. G. P. Page for careful reading and suggestions regarding this
and physicians continue struggling to stay ahead, or at least review.
abreast, of the abilities of ␤-lactamases, we must utilize intel-
ligently and productively the agents that are currently avail-
REFERENCES
able. For example, we can commit to applying the research that
1. Abraham, E. P., and E. Chain. 1940. An enzyme from bacteria able to
demonstrates the potential of novel combinations of ␤-lactams destroy penicillin. Nature 146:837.
and ␤-lactamase inhibitors, such as clavulanate with cefepime, 2. Abraham, E. P., E. Chain, C. M. Fletcher, H. W. Florey, A. D. Gardner,
cefpirome, or meropenem, to expand and enhance inhibitors’ N. G. Heatley, and M. A. Jennings. 1941. Further observations on penicillin.
Lancet ii:177.
abilities to protect partner ␤-lactams (172, 239). Several sig- 3. Adachi, H., T. Ohta, and H. Matsuzawa. 1991. Site-directed mutants, at
nificant hurdles stand in the way of these novel combinations, position 166, of RTEM-1 ␤-lactamase that form a stable acyl-enzyme in-
termediate with penicillin. J. Biol. Chem. 266:3186–3191.
such as corporate relationships affecting the funding of the 4. Adediran, S. A., M. Nukaga, S. Baurin, J. M. Frere, and R. F. Pratt. 2005.
trials necessary to demonstrate clinical efficacy, as well as op- Inhibition of class D ␤-lactamases by acyl phosphates and phosphonates.
timization of the pharmacokinetics, pharmacodynamics, and Antimicrob. Agents Chemother. 49:4410–4412.
5. Adediran, S. A., and R. F. Pratt. 2008. Inhibition of serine ␤-lactamases by
safety of the components. The ␤-lactamase-inhibitory activity vanadate-catechol complexes. Biochemistry 47:9467–9474.
of carbapenems should not be overlooked in the face of in- 6. Afzal-Shah, M., N. Woodford, and D. M. Livermore. 2001. Characterization
creasing ␤-lactam resistance. Carbapenems, for the time being, of OXA-25, OXA-26, and OXA-27, molecular class D ␤-lactamases asso-
ciated with carbapenem resistance in clinical isolates of Acinetobacter bau-
remain effective against many difficult-to-treat infections and mannii. Antimicrob. Agents Chemother. 45:583–588.
teach us that the distinction between an inhibitor and a slow 7. Akova, M. 2008. Sulbactam-containing ␤-lactamase inhibitor combinations.
Clin. Microbiol. Infect. 14(Suppl. 1):185–188.
substrate may be blurred. 8. Akova, M., Y. Yang, and D. M. Livermore. 1990. Interactions of tazobactam
and clavulanate with inducibly- and constitutively-expressed Class I ␤-lac-
tamases. J. Antimicrob. Chemother. 25:199–208.
9. Alonso, R., G. Gerbaud, M. Galimand, and P. Courvalin. 2002. TEM-103/
A PERSPECTIVE IRT-28 ␤-lactamase, a new TEM variant produced by Escherichia coli
BM4511. Antimicrob. Agents Chemother. 46:3627–3629.
From our vantage point, the main challenge in ␤-lactamase 10. Ambler, R. P., A. F. Coulson, J. M. Frere, J. M. Ghuysen, B. Joris, M.
Forsman, R. C. Levesque, G. Tiraby, and S. G. Waley. 1991. A standard
inhibitor development is discovering novel inhibitors with ac- numbering scheme for the class A ␤-lactamases. Biochem. J. 276:269–270.
tivity against a broad spectrum of inhibitor-susceptible and 11. Anderson, S. D., and J. G. Gums. 2008. Ceftobiprole: an extended-spectrum
anti-methicillin-resistant Staphylococcus aureus cephalosporin. Ann. Phar-
-resistant enzymes from multiple classes (17, 410). New inhib- macother. 42:806–816.
itors must also target carbapenemases (serine and metallo-), as 12. Angus, B. L., A. M. Carey, D. A. Caron, A. M. Kropinski, and R. E.
carbapenems are still the most potent ␤-lactams available for Hancock. 1982. Outer membrane permeability in Pseudomonas aeruginosa:
comparison of a wild-type with an antibiotic-supersusceptible mutant. An-
clinical use. Future advances in inhibitor design against versa- timicrob. Agents Chemother. 21:299–309.
tile ␤-lactamases must incorporate strategies that address each 13. Atanasov, B. P., D. Mustafi, and M. W. Makinen. 2000. Protonation of the
192 DRAWZ AND BONOMO CLIN. MICROBIOL. REV.

␤-lactam nitrogen is the trigger event in the catalytic action of class A a new siderophore monobactam BAL 30072 against ESBL-producing En-
␤-lactamases. Proc. Natl. Acad. Sci. U. S. A. 97:3160–3165. terobacteriacae and clinical isolates of Enterobacter cloacae, abstr. P1112.
14. Babic, M., A. M. Hujer, and R. A. Bonomo. 2006. What’s new in antibiotic Abstr. 19th Eur. Congr. Clin. Microbiol. Infect. Dis., Helsinki, Finland.
resistance? Focus on ␤-lactamases. Drug Resist. Updat. 9:142–156. 39. Bowler, L. D., Q. Y. Zhang, J. Y. Riou, and B. G. Spratt. 1994. Interspecies
15. Babini, G. S., and D. M. Livermore. 2000. Effect of conalbumin on the recombination between the penA genes of Neisseria meningitidis and commen-
activity of Syn 2190, a 1,5 dihydroxy-4-pyridon monobactam inhibitor of sal Neisseria species during the emergence of penicillin resistance in N. men-
AmpC ␤-lactamases. J. Antimicrob. Chemother. 45:105–109. ingitidis: natural events and laboratory simulation. J. Bacteriol. 176:333–337.
16. Bantar, C., E. Vesco, C. Heft, F. Salamone, M. Krayeski, H. Gomez, M. A. 40. Bradford, P. A. 2001. Extended-spectrum ␤-lactamases in the 21st century:
Coassolo, A. Fiorillo, D. Franco, C. Arango, F. Duret, and M. E. Oliva. characterization, epidemiology, and detection of this important resistance
2004. Replacement of broad-spectrum cephalosporins by piperacillin- threat. Clin. Microbiol. Rev. 14:933–951.
tazobactam: impact on sustained high rates of bacterial resistance. Antimi- 41. Bradford, P. A., S. Bratu, C. Urban, M. Visalli, N. Mariano, D. Landman,
crob. Agents Chemother. 48:392–395. J. J. Rahal, S. Brooks, S. Cebular, and J. Quale. 2004. Emergence of
17. Bassetti, M., E. Righi, and C. Viscoli. 2008. Novel ␤-lactam antibiotics and carbapenem-resistant Klebsiella species possessing the class A carbapenem-
inhibitor combinations. Expert Opin. Invest. Drugs 17:285–296. hydrolyzing KPC-2 and inhibitor-resistant TEM-30 b-lactamases in New
18. Bauernfeind, A., J. M. Casellas, M. Goldberg, M. Holley, R. Jungwirth, P. York City. Clin. Infect. Dis. 39:55–60.
Mangold, T. Rohnisch, S. Schweighart, and R. Wilhelm. 1992. A new 42. Bradford, P. A., C. E. Cherubin, V. Idemyor, B. A. Rasmussen, and K.
plasmidic cefotaximase from patients infected with Salmonella typhi- Bush. 1994. Multiply resistant Klebsiella pneumoniae strains from two Chi-
murium. Infection 20:158–163. cago hospitals: identification of the extended-spectrum TEM-12 and
19. Bauvois, C., and J. Wouters. 2007. Crystal structures of class C ␤-lactama- TEM-10 ceftazidime-hydrolyzing ␤-lactamases in a single isolate. Antimi-
ses: mechanistic implications and perspectives in drug design, p. 145–161. In crob. Agents Chemother. 38:761–766.
R. A. Bonomo and M. E. Tolmasky (ed.), Enzyme-mediated resistance to 43. Bradley, S. J., A. L. Wilson, M. C. Allen, H. A. Sher, A. H. Goldstone, and
antibiotics: mechanisms, dissemination, and prospects for inhibition. ASM G. M. Scott. 1999. The control of hyperendemic glycopeptide-resistant
Press, Washington, DC. Enterococcus spp. on a haematology unit by changing antibiotic usage. J.
20. Bayles, K. W. 2000. The bactericidal action of penicillin: new clues to an Antimicrob. Chemother. 43:261–266.
unsolved mystery. Trends Microbiol. 8:274–278. 44. Brenner, D. G., and J. R. Knowles. 1981. Penicillanic acid sulfone: an
21. Beadle, B. M., and B. K. Shoichet. 2002. Structural basis for imipenem unexpected isotope effect in the interaction of 6 ␣- and 6 ␤-monodeuterio
inhibition of class C ␤-lactamases. Antimicrob. Agents Chemother. 46: and of 6,6-dideuterio derivatives with RTEM ␤-lactamase from Escherichia
3978–3980. coli. Biochemistry 20:3680–3687.
22. Beesley, T., N. Gascoyne, V. Knott-Hunziker, S. Petursson, S. G. Waley, B. 45. Brenner, D. G., and J. R. Knowles. 1984. Penicillanic acid sulfone: nature of
Jaurin, and T. Grundstrom. 1983. The inhibition of class C ␤-lactamases by irreversible inactivation of RTEM ␤-lactamase from Escherichia coli. Bio-
boronic acids. Biochem. J. 209:229–233. chemistry 23:5833–5839.
23. Belaaouaj, A., C. Lapoumeroulie, M. M. Canica, G. Vedel, P. Nevot, R. 46. Broom, N. J. P., T. H. Farmer, N. F. Osborne, and J. W. Tyler. 1992. Studies
Krishnamoorthy, and G. Paul. 1994. Nucleotide sequences of the genes on the mechanism of action of (5R)-(Z)-6-(1-methyl-1,2,3-triazol-4-ylmeth-
coding for the TEM-like ␤-lactamases IRT-1 and IRT-2 (formerly called ylene) penem-3-carboxylic acid (BRL 42715), a potent inhibitor of bacterial
TRI-1 and TRI-2). FEMS Microbiol. Lett. 120:75–80. ␤-lactamase. J. Chem. Soc. Chem. Commun. 1992:1663–1664.
24. Bennett, P. M., and I. Chopra. 1993. Molecular basis of ␤-lactamase induc- 47. Brown, A. G. 1986. Clavulanic acid, a novel ␤-lactamase inhibitor—a case
tion in bacteria. Antimicrob. Agents Chemother. 37:153–158. study in drug discovery and development. Drug Des. Deliv. 1:1–21.
25. Bethel, C. R., A. M. Distler, M. W. Ruszczycky, M. P. Carey, P. R. Carey, 48. Brown, R. P., R. T. Aplin, and C. J. Schofield. 1996. Inhibition of TEM-2
A. M. Hujer, M. Taracila, M. S. Helfand, J. M. Thomson, M. Kalp, V. E. ␤-lactamase from Escherichia coli by clavulanic acid: observation of inter-
Anderson, D. A. Leonard, K. M. Hujer, T. Abe, A. M. Venkatesan, T. S. mediates by electrospray ionization mass spectrometry. Biochemistry 35:
Mansour, and R. A. Bonomo. 2008. Inhibition of OXA-1 ␤-lactamase by 12421–12432.
penems. Antimicrob. Agents Chemother. 52:3135–3143. 49. Budzikiewicz, H. 2001. Siderophore-antibiotic conjugates used as Trojan
26. Bitha, P., Z. Li, G. D. Francisco, B. A. Rasmussen, and Y. I. Lin. 1999. horses against Pseudomonas aeruginosa. Curr. Top. Med. Chem. 1:73–82.
6-(1-Hydroxyalkyl)penam sulfone derivatives as inhibitors of class A and 50. Bulychev, A., J. Bellettini, M. O’Brien, P. J. Crocker, J. P. Samama, M. J.
class C ␤-lactamases. I. Bioorg. Med. Chem. Lett. 9:991–996. Miller, and S. Mobashery. 2000. N-Sulfonyloxy-␤-lactam inhibitors for
27. Bitha, P., Z. Li, G. D. Francisco, Y. Yang, P. J. Petersen, E. Lenoy, and Y. I. ␤-lactamases. Tetrahedron 56:5719–5728.
Lin. 1999. 6-(1-Hydroxyalkyl)penam sulfone derivatives as inhibitors of 51. Bulychev, A., I. Massova, K. Miyashita, and S. Mobashery. 1997. Nuances
class A and class C ␤-lactamases. II. Bioorg. Med. Chem. Lett. 9:997–1002. of mechanisms and their implications for evolution of the versatile ␤-lac-
28. Black, J. A., K. S. Thomson, J. D. Buynak, and J. D. Pitout. 2005. Evalu- tamase activity: from biosynthetic enzymes to drug resistance factors.
ation of ␤-lactamase inhibitors in disk tests for detection of plasmid-medi- J. Am. Chem. Soc. 119:7619–7625.
ated AmpC ␤-lactamases in well-characterized clinical strains of Klebsiella 52. Bush, K. 1998. Metallo-␤-lactamases: a class apart. Clin. Infect. Dis.
spp. J. Clin. Microbiol. 43:4168–4171. 27(Suppl. 1):S48–S53.
29. Blazquez, J., M. R. Baquero, R. Canton, I. Alos, and F. Baquero. 1993. 53. Bush, K. 1988. ␤-Lactamase inhibitors from laboratory to clinic. Clin. Mi-
Characterization of a new TEM-type ␤-lactamase resistant to clavulanate, crobiol. Rev. 1:109–123.
sulbactam, and tazobactam in a clinical isolate of Escherichia coli. Antimi- 54. Bush, K. 1986. Evaluation of enzyme inhibition data in screening for new
crob. Agents Chemother. 37:2059–2063. drugs. Drugs Exp. Clin. Res. 12:565–576.
30. Bone, R., A. B. Shenvi, C. A. Kettner, and D. A. Agard. 1987. Serine 55. Bush, K., and P. A. Bradford. 2007. ␤-Lactamases: historical perspectives,
protease mechanism: structure of an inhibitory complex of alpha-lytic pro- p. 67–79. In R. A. Bonomo and M. E. Tolmasky (ed.), Enzyme-mediated
tease and a tightly bound peptide boronic acid. Biochemistry 26:7609–7614. resistance to antibiotics: mechanisms, dissemination, and prospects for in-
31. Bonnefoy, A., C. Dupuis-Hamelin, V. Steier, C. Delachaume, C. Seys, T. hibition. ASM Press, Washington, DC.
Stachyra, M. Fairley, M. Guitton, and M. Lampilas. 2004. In vitro activity 56. Bush, K., J. S. Freudenberger, and R. B. Sykes. 1982. Interaction of
of AVE1330A, an innovative broad-spectrum non-␤-lactam ␤-lactamase azthreonam and related monobactams with ␤-lactamases from gram-nega-
inhibitor. J. Antimicrob. Chemother. 54:410–417. tive bacteria. Antimicrob. Agents Chemother. 22:414–420.
32. Bonnet, R. 2004. Growing group of extended-spectrum ␤-lactamases: the 56a.Bush, K., and G. A. Jacoby. 2009. An updated functional clssification of
CTX-M enzymes. Antimicrob. Agents Chemother. 48:1–14. ␤-lactamases. Antimicrob. Agents Chemother. [Epub ahead of print.] doi:
33. Bonomo, R. A., C. Currie-McCumber, and D. M. Shlaes. 1992. OHIO-1 10.1128/AAC.01009-09.
␤-lactamase resistant to mechanism-based inactivators. FEMS Microbiol. 57. Bush, K., G. A. Jacoby, and A. A. Medeiros. 1995. A functional classification
Lett. 71:79–82. scheme for ␤-lactamases and its correlation with molecular structure. An-
34. Bonomo, R. A., J. Liu, Y. Chen, L. Ng, A. M. Hujer, and V. E. Anderson. timicrob. Agents Chemother. 39:1211–1233.
2001. Inactivation of CMY-2 ␤-lactamase by tazobactam: initial mass spec- 58. Bush, K., C. Macalintal, B. A. Rasmussen, V. J. Lee, and Y. Yang. 1993.
troscopic characterization. Biochim. Biophys. Acta 1547:196–205. Kinetic interactions of tazobactam with ␤-lactamases from all major struc-
35. Bonomo, R. A., and L. B. Rice. 1999. Inhibitor resistant class A ␤-lactama- tural classes. Antimicrob. Agents Chemother. 37:851–858.
ses. Front. Biosci. 4:e34–41. 59. Bush, L. M., and C. C. Johnson. 2000. Ureidopenicillins and ␤-lactam/␤-
36. Bonomo, R. A., S. A. Rudin, and D. M. Shlaes. 1997. Tazobactam is a potent lactam inhibitor combinations. Infect. Dis. Clin. North Am. 14:409–433.
inactivator of selected inhibitor-resistant class A ␤-lactamases. FEMS Mi- 60. Buynak, J. D. 2006. Understanding the longevity of the ␤-lactam antibiotics
crobiol. Lett. 148:59–62. and of antibiotic/␤-lactamase inhibitor combinations. Biochem. Pharmacol.
37. Bowker, K. E., A. R. Noel, and A. P. MacGowan. 2007. A monobactam plus 71:930–940.
double ␤-lactamase inhibitor combination (BAL 30376) designed to over- 61. Buynak, J. D., H. Chen, L. Vogeti, V. R. Gadhachanda, C. A. Buchanan, T.
come multiple resistance in gram negative bacilli: proof of concept in an in Palzkill, R. W. Shaw, J. Spencer, and T. R. Walsh. 2004. Penicillin-derived
vitro pharmacokinetic model, abstr. F1-314. Abstr. 47th Intersci. Conf. inhibitors that simultaneously target both metallo- and serine-␤-lactamases.
Antimicrob. Agents Chemother., Chicago, IL. Bioorg. Med. Chem. Lett. 14:1299–1304.
38. Bowker, K. E., T. R. Walsh, and A. P. MacGowan. 2009. In vitro activity of 62. Buynak, J. D., V. R. Doppalapudi, and G. Adam. 2000. The synthesis and
VOL. 23, 2010 ␤-LACTAMASE INHIBITORS 193

evaluation of 3-substituted-7-(alkylidene)cephalosporin sulfones as ␤-lac- mase from Pseudomonas aeruginosa and its complex with a mercaptocar-
tamase inhibitors. Bioorg. Med. Chem. Lett. 10:853–857. boxylate inhibitor: binding determinants of a potent, broad-spectrum inhib-
63. Buynak, J. D., A. S. Rao, V. R. Doppalapudi, G. Adam, P. J. Petersen, and itor. Biochemistry 39:4288–4298.
S. D. Nidamarthy. 1999. The synthesis and evaluation of 6-alkylidene-2⬘␤- 89. Conrath, K. E., M. Lauwereys, M. Galleni, A. Matagne, J. M. Frere, J.
substituted penam sulfones as ␤-lactamase inhibitors. Bioorg. Med. Chem. Kinne, L. Wyns, and S. Muyldermans. 2001. ␤-Lactamase inhibitors de-
Lett. 9:1997–2002. rived from single-domain antibody fragments elicited in the camelidae.
64. Buynak, J. D., L. Vogeti, V. R. Doppalapudi, G. M. Solomon, and H. Chen. Antimicrob. Agents Chemother. 45:2807–2812.
2002. Cephalosporin-derived inhibitors of ␤-lactamase. 4. The C3 substitu- 90. Copar, A., T. Prevec, B. Anzic, T. Mesar, L. Selic, M. Vilar, and T. Solmajer.
ent. Bioorg. Med. Chem. Lett. 12:1663–1666. 2002. Design, synthesis and bioactivity evaluation of tribactam ␤-lactamase
65. Buynak, J. D., K. Wu, B. Bachmann, D. Khasnis, L. Hua, H. K. Nguyen, inhibitors. Bioorg. Med. Chem. Lett. 12:971–975.
and C. L. Carver. 1995. Synthesis and biological activity of 7-alkylidene- 91. Copar, A., T. Solmajer, B. Anzie, T. Kuzman, T. Mesar, and D. Kocjan. December
cephems. J. Med. Chem. 38:1022–1034. 2002. Ethylidene derivatives of tricyclic carbapenems. U.S. patent 6,489,318.
66. Campoli-Richards, D. M., and R. N. Brogden. 1987. Sulbactam/ampicillin. 92. Copeland, R. A. 2005. Evaluation of enzyme inhibitors in drug discovery.
A review of its antibacterial activity, pharmacokinetic properties, and ther- John Wiley & Sons, Inc., New York, NY.
apeutic use. Drugs 33:577–609. 93. Corvec, S., L. Poirel, E. Espaze, C. Giraudeau, H. Drugeon, and P. Nord-
67. Canton, R., and T. M. Coque. 2006. The CTX-M ␤-lactamase pandemic. mann. 2008. Long-term evolution of a nosocomial outbreak of Pseudomo-
Curr. Opin. Microbiol. 9:466–475. nas aeruginosa producing VIM-2 metallo-enzyme. J. Hosp. Infect. 68:73–82.
68. Canton, R., M. I. Morosini, O. Martin, S. de la Maza, and E. G. de la 94. Crichlow, G. V., M. Nukaga, V. R. Doppalapudi, J. D. Buynak, and J. R.
Pedrosa. 2008. IRT and CMT ␤-lactamases and inhibitor resistance. Clin. Knox. 2001. Inhibition of class C ␤-lactamases: structure of a reaction
Microbiol. Infect. 14(Suppl. 1):53–62. intermediate with a cephem sulfone. Biochemistry 40:6233–6239.
69. Cartwright, S. J., and A. F. Coulson. 1979. A semi-synthetic penicillinase 95. Crompton, I. E., B. K. Cuthbert, G. Lowe, and S. G. Waley. 1988. ␤-lacta-
inactivator. Nature 278:360–361. mase inhibitors. The inhibition of serine ␤-lactamases by specific boronic
70. Caselli, E., R. A. Powers, L. C. Blasczcak, C. Y. Wu, F. Prati, and B. K. acids. Biochem. J. 251:453–459.
Shoichet. 2001. Energetic, structural, and antimicrobial analyses of ␤-lac- 96. Crowder, M. W., J. Spencer, and A. J. Vila. 2006. Metallo-␤-lactamases:
tam side chain recognition by ␤-lactamases. Chem. Biol. 8:17–31. novel weaponry for antibiotic resistance in bacteria. Acc. Chem. Res. 39:
71. Centers for Disease Control. 1982. Global distribution of penicillinase- 721–728.
producing Neisseria gonorrhoeae (PPNG). MMWR Morb. Mortal. Wkly. 97. Daiyasu, H., K. Osaka, Y. Ishino, and H. Toh. 2001. Expansion of the zinc
Rep. 31:1–3. metallo-hydrolase family of the ␤-lactamase fold. FEBS Lett. 503:1–6.
72. Chaibi, E. B., S. Farzaneh, A. Morand, J. Peduzzi, M. Barthelemy, D. Sirot,
98. Danel, F., M. G. Page, and D. M. Livermore. 2007. Class D ␤-lactamases, p.
and R. Labia. 1996. Problems encountered in the characterization of IRT
163–194. In R. A. Bonomo and M. E. Tolmasky (ed.), Enzyme-mediated
␤-lactamase-producing clinical Escherichia coli isolates intermediate-resis-
resistance to antibiotics: mechanisms, dissemination, and prospects for in-
tant to cephalothin. J. Antimicrob. Chemother. 37:190–191.
hibition. ASM Press, Washington, DC.
73. Chaibi, E. B., J. Peduzzi, S. Farzaneh, M. Barthelemy, D. Sirot, and R.
99. Danel, F., and M. G. P. Page. 2007. Inhibition of AmpC ␤-lactamase by
Labia. 1998. Clinical inhibitor-resistant mutants of the ␤-lactamase TEM-1
monobactams, abstr. F-126. Abstr. 47th Intersci. Conf. Antimicrob. Agents
at amino-acid position 69. Kinetic analysis and molecular modelling. Bio-
Chemother., Chicago, IL.
chim. Biophys. Acta 1382:38–46.
100. Datta, N., and P. Kontomichalou. 1965. Penicillinase synthesis controlled
74. Chaibi, E. B., D. Sirot, G. Paul, and R. Labia. 1999. Inhibitor-resistant
by infectious R factors in Enterobacteriaceae. Nature 208:239–241.
TEM ␤-lactamases: phenotypic, genetic and biochemical characteristics. J.
Antimicrob. Chemother. 43:447–458. 101. Delaire, M., R. Labia, J. P. Samama, and J. M. Masson. 1992. Site-directed
75. Chakraborty, R., E. Storey, and D. van der Helm. 2007. Molecular mech- mutagenesis at the active site of Escherichia coli TEM-1 ␤-lactamase. Sui-
anism of ferricsiderophore passage through the outer membrane receptor cide inhibitor-resistant mutants reveal the role of arginine 244 and methio-
proteins of Escherichia coli. Biometals 20:263–274. nine 69 in catalysis. J. Biol. Chem. 267:20600–20606.
76. Chambers, H. F. 1999. Penicillin-binding protein-mediated resistance in 102. Desarbre, E., A. DeFontage, M. G. P. Page, F. Richalet, and P. Roussel.
pneumococci and staphylococci. J. Infect. Dis. 179(Suppl. 2):S353–S359. 2007. Antimicrobial activity of novel monobactams, abstr. F1-351. Abstr.
77. Chardon, H., S. Farzaneh, R. Labia, V. Jarlier, M. H. Nicolas, G. Paul, C. 47th Intersci. Conf. Antimicrob. Agents Chemother., Chicago, IL.
Poyart, D. Sirot, and J. Sirot. 1995. Analysis of ␤-lactamases produced by 103. Diaz, N., T. L. Sordo, K. M. Merz Jr., and D. Suarez. 2003. Insights into the
cephalothin-susceptible Escherichia coli clinical isolates resistant to co- acylation mechanism of class A ␤-lactamases from molecular dynamics
amoxiclav and ticarcillin-clavulanic acid. The CERIB Study Group. J. An- simulations of the TEM-1 enzyme complexed with benzylpenicillin. J. Am.
timicrob. Chemother. 36:267–269. Chem. Soc. 125:672–684.
78. Charnas, R. L., J. Fisher, and J. R. Knowles. 1978. Chemical studies on the 104. Docquier, J., T. Stachyra, M. Benvenuti, G. M. Rossolini, S. Mangani,
inactivation of Escherichia coli RTEM ␤-lactamase by clavulanic acid. Bio- M. C. Pechereau, J. M. Bruneau, M. Claudon, F. Barbosa, C. Miossec, and
chemistry 17:2185–2189. M. T. Black. 2009. High resolution crystal structure of CTX-M-15 in com-
79. Charnas, R. L., and J. R. Knowles. 1981. Inactivation of RTEM ␤-lacta- plex with the new ␤-lactamase inhibitor NXL104, abstr. C1-1098. Abstr.
mase from Escherichia coli by clavulanic acid and 9-deoxyclavulanic acid. 49th Intersci. Conf. Antimicrob. Agents Chemother., San Francisco, CA.
Biochemistry 20:3214–3219. 105. Doran, J. L., B. K. Leskiw, S. Aippersbach, and S. E. Jensen. 1990. Isolation
80. Charnas, R. L., and J. R. Knowles. 1981. Inhibition of the RTEM ␤-lacta- and characterization of a ␤-lactamase-inhibitory protein from Streptomyces
mase from Escherichia coli. Interaction of enzyme with derivatives of clavuligerus and cloning and analysis of the corresponding gene. J. Bacte-
olivanic acid. Biochemistry. 20:2732–2737. riol. 172:4909–4918.
81. Chen, C. C., and O. Herzberg. 1992. Inhibition of ␤-lactamase by clavu- 106. Doumith, M., M. J. Ellington, D. M. Livermore, and N. Woodford. 2009.
lanate. Trapped intermediates in cryocrystallographic studies. J. Mol. Biol. Molecular mechanisms disrupting porin expression in ertapenem-resistant
224:1103–1113. Klebsiella and Enterobacter spp. clinical isolates from the UK. J. Antimicrob.
82. Chen, C. C., J. Rahil, R. F. Pratt, and O. Herzberg. 1993. Structure of a Chemother. 63:659–667.
phosphonate-inhibited ␤-lactamase. An analog of the tetrahedral transition 107. Dowson, C. G., A. Hutchison, N. Woodford, A. P. Johnson, R. C. George,
state/intermediate of ␤-lactam hydrolysis. J. Mol. Biol. 234:165–178. and B. G. Spratt. 1990. Penicillin-resistant viridans streptococci have ob-
83. Chen, Y., J. Delmas, J. Sirot, B. Shoichet, and R. Bonnet. 2005. Atomic tained altered penicillin-binding protein genes from penicillin-resistant
resolution structures of CTX-M ␤-lactamases: extended spectrum activities strains of Streptococcus pneumoniae. Proc. Natl. Acad. Sci. U. S. A. 87:
from increased mobility and decreased stability. J. Mol. Biol. 348:349–362. 5858–5862.
84. Chen, Y., G. Minasov, T. A. Roth, F. Prati, and B. K. Shoichet. 2006. The 107a.Drawz, S. M., M. Babic, C. R. Bethel, M. Taracila, A. M. Distler, C. Ori, E.
deacylation mechanism of AmpC ␤-lactamase at ultrahigh resolution. Caselli, F. Prati, and R. A. Bonomo. Inhibition of the class C ␤-lactamase
J. Am. Chem. Soc. 128:2970–2976. from Acinetobacter spp.: insights into effective inhibitor design. Biochemis-
85. Chen, Y., B. Shoichet, and R. Bonnet. 2005. Structure, function, and inhi- try. [Epub ahead of print.] doi:10.1021/bi9015988.
bition along the reaction coordinate of CTX-M ␤-lactamases. J. Am. Chem. 108. Drawz, S. M., C. R. Bethel, K. M. Hujer, K. N. Hurless, A. M. Distler, E.
Soc. 127:5423–5434. Caselli, F. Prati, and R. A. Bonomo. 2009. The role of a second-shell
86. Cherry, P. C., C. E. Newall, and N. S. Weston. 1978. Preparation of the residue in modifying substrate and inhibitor interactions in the SHV ␤-lac-
7-oxo-4-oxa-1-azabicyclo[3.2.0]hept-2-ene system and the reversible cleav- tamase: a study of Ambler position Asn276. Biochemistry. 48:4557–4566.
age of its oxazoline ring. J. Chem. Soc. Chem. Commun. 1978:469–470. 109. Dubois, V., L. Poirel, C. Arpin, L. Coulange, C. Bebear, P. Nordmann, and
87. Coleman, K., D. R. Griffin, J. W. Page, and P. A. Upshon. 1989. In vitro C. Quentin. 2004. SHV-49, a novel inhibitor-resistant ␤-lactamase in a
evaluation of BRL 42715, a novel ␤-lactamase inhibitor. Antimicrob. clinical isolate of Klebsiella pneumoniae. Antimicrob. Agents Chemother.
Agents Chemother. 33:1580–1587. 48:4466–4469.
88. Concha, N. O., C. A. Janson, P. Rowling, S. Pearson, C. A. Cheever, B. P. 110. Dubois, V., L. Poirel, F. Demarthe, C. Arpin, L. Coulange, L. A. Minarini,
Clarke, C. Lewis, M. Galleni, J. M. Frere, D. J. Payne, J. H. Bateson, and M. C. Bezian, P. Nordmann, and C. Quentin. 2008. Molecular and bio-
S. S. Abdel-Meguid. 2000. Crystal structure of the IMP-1 metallo-␤-lacta- chemical characterization of SHV-56, a novel inhibitor-resistant ␤-lacta-
194 DRAWZ AND BONOMO CLIN. MICROBIOL. REV.

mase from Klebsiella pneumoniae. Antimicrob. Agents Chemother. 2005. A metallo-␤-lactamase enzyme in action: crystal structures of the
52:3792–3794 monozinc carbapenemase CphA and its complex with biapenem. J. Mol.
111. Dubus, A., P. Ledent, J. Lamotte-Brasseur, and J. M. Frere. 1996. The roles Biol. 345:785–795.
of residues Tyr150, Glu272, and His314 in class C ␤-lactamases. Proteins 133. Garau, G., I. Garcia-Saez, C. Bebrone, C. Anne, P. Mercuri, M. Galleni, J. M.
25:473–485. Frere, and O. Dideberg. 2004. Update of the standard numbering scheme for
112. Dubus, A., J. M. Wilkin, X. Raquet, S. Normark, and J. M. Frere. 1994. class B ␤-lactamases. Antimicrob. Agents Chemother. 48:2347–2349.
Catalytic mechanism of active-site serine ␤-lactamases: role of the conserved 134. Gaucher, B., C. Dantier, E. Desarbre, and M. G. P. Page. 2007. Novel mono-
hydroxy group of the Lys-Thr(Ser)-Gly triad. Biochem. J. 30:485–494. and bicyclic monobactams as potent inhibitors of class C ␤-lactamases, abstr.
113. Durkin, J. P., and T. Viswanatha. 1978. Clavulanic acid inhibition of ␤-lac- F1-310. Abstr. 47th Intersci. Conf. Antimicrob. Agents Chemother., Chicago,
tamase I from Bacillus cereus 569/H. J. Antibiot. (Tokyo) 31:1162–1169. IL.
114. Easton, C. J., and J. R. Knowles. 1982. Inhibition of the RTEM ␤-lactamase 135. Gavin, P. J., M. T. Suseno, R. B. Thomson, Jr., J. M. Gaydos, C. L. Pierson,
from Escherichia coli. Interaction of the enzyme with derivatives of olivanic D. C. Halstead, J. Aslanzadeh, S. Brecher, C. Rotstein, S. E. Brossette, and
acid. Biochemistry 21:2857–2862. L. R. Peterson. 2006. Clinical correlation of the CLSI susceptibility break-
115. Elwell, L. P., J. De Graaff, D. Seibert, and S. Falkow. 1975. Plasmid-linked point for piperacillin-tazobactam against extended-spectrum-beta-lacta-
ampicillin resistance in Haempohilus influenza type b. Infect. Immun. 12: mase-producing Escherichia coli and Klebsiella species. Antimicrob. Agents
404–410. Chemother. 50:2244–2247.
116. Endimiani, A., Y. Choudhary, and R. A. Bonomo. 2009. In vitro activity of 136. Gherman, B. F., S. D. Goldberg, V. W. Cornish, and R. A. Friesner. 2004.
NXL104 in combination with ␤-lactams against Klebsiella pneumoniae iso- Mixed quantum mechanical/molecular mechanical (QM/MM) study of the
lates producing KPC carbapenemases. Antimicrob. Agents Chemother. deacylation reaction in a penicillin binding protein (PBP) versus in a class
53:3599–3601. C ␤-lactamase. J. Am. Chem. Soc. 126:7652–7664.
116a.Endimiani, A., Y. Doi, C. R. Bethel, M. Taracila, J. M. Adams-Haduch, 137. Giakkoupi, P., E. Tzelepi, N. J. Legakis, and L. S. Tzouvelekis. 1999.
A. O’Keefe, A. M. Hujer, D. L. Paterson, M. J. Skalweit, M. G. Page, Aspartic acid for asparagine substitution at position 276 reduces suscepti-
S. M. Drawz, and R. A. Bonomo. 2009. Enhancing resistance to cephalo- bility to mechanism-based inhibitors in SHV-1 and SHV-5 ␤-lactamases. J.
sporins in class C ␤-lactamases: impact of Gly116Glu in CMY-2. Biochem- Antimicrob. Chemother. 43:23–29.
istry. [Epub ahead of print.] doi:10.1021/bi9015549. 138. Giakkoupi, P., E. Tzelepi, N. J. Legakis, and L. S. Tzouvelekis. 1998.
116b.Endimiani, A., A. M. Hujer, F. Perez, C. R. Bethel, K. M. Hujer, J. Kroeger, Substitution of Arg-244 by Cys or Ser in SHV-1 and SHV-5 ␤-lactamases
M. Oethinger, D. L. Paterson, M. D. Adams, M. R. Jacobs, D. J. Diekema, confers resistance to mechanism-based inhibitors and reduces catalytic ef-
G. S. Hall, S. G. Jenkins, L. B. Rice, F. C. Tenover, and R. A. Bonomo. 2009. ficiency of the enzymes. FEMS Microbiol. Lett. 160:49–54.
Characterization of blaKPC-containing Klebsiella pneumoniae isolates de- 139. Gibson, R. M., H. Christensen, and S. G. Waley. 1990. Site-directed mu-
tected in different institutions in the eastern USA. J. Antimicrob. Che- tagenesis of ␤-lactamase I. Single and double mutants of Glu-166 and
mother. 63:427–437. Lys-73. Biochem. J. 272:613–619.
117. English, A. R., J. A. Retsema, A. E. Girard, J. E. Lynch, and W. E. Barth. 140. Gin, A., L. Dilay, J. A. Karlowsky, A. Walkty, E. Rubinstein, and G. G.
1978. CP-45,899, a ␤-lactamase inhibitor that extends the antibacterial Zhanel. 2007. Piperacillin-tazobactam: a ␤-lactam/␤-lactamase inhibitor
spectrum of ␤-lactams: initial bacteriological characterization. Antimicrob. combination. Expert Rev. Anti-Infect. Ther. 5:365–383.
Agents Chemother. 14:414–419. 141. Girlich, D., A. Karim, L. Poirel, M. H. Cavin, C. Verny, and P. Nordmann.
118. Falagas, M. E., P. E. Valkimadi, Y. T. Huang, D. K. Matthaiou, and P. R. 2000. Molecular epidemiology of an outbreak due to IRT-2 ␤-lactamase-
Hsueh. 2008. Therapeutic options for Stenotrophomonas maltophilia infec- producing strains of Klebsiella pneumoniae in a geriatric department. J.
tions beyond co-trimoxazole: a systematic review. J. Antimicrob. Che- Antimicrob. Chemother. 45:467–473.
mother. 62:889–894. 141a.Girlich, D., T. Naas, and P. Nordmann. 2004. Biochemical characterization
119. Farmer, T. H., J. W. Page, D. J. Payne, and D. J. Knowles. 1994. Kinetic and of the naturally occurring oxacillinase OXA-50 of Pseudomonas aeruginosa.
physical studies of ␤-lactamase inhibition by a novel penem, BRL 42715. Antimicrob. Agents Chemother. 48:2043–2048.
Biochem. J. 303:825–830. 142. Goffin, C., and J. M. Ghuysen. 1998. Multimodular penicillin-binding pro-
120. Farzaneh, S., E. B. Chaibi, J. Peduzzi, M. Barthelemy, R. Labia, J. teins: an enigmatic family of orthologs and paralogs. Microbiol. Mol. Biol.
Blazquez, and F. Baquero. 1996. Implication of Ile-69 and Thr-182 residues Rev. 62:1079–1093.
in kinetic characteristics of IRT-3 (TEM-32) ␤-lactamase. Antimicrob. 143. Golemi, D., L. Maveyraud, A. Ishiwata, S. Tranier, K. Miyashita, T.
Agents Chemother. 40:2434–2436. Nagase, I. Massova, L. Mourey, J. P. Samama, and S. Mobashery. 2000.
121. Fisher, J., J. G. Belasco, R. L. Charnas, S. Khosla, and J. R. Knowles. 1980. 6-(Hydroxyalkyl)penicillinates as probes for mechanisms of ␤-lactamases. J.
␤-Lactamase inactivation by mechanism-based reagents. Philos. Trans. R. Antibiot. (Tokyo) 53:1022–1027.
Soc. Lond. B Biol. Sci. 289:309–319. 144. Golemi, D., L. Maveyraud, S. Vakulenko, J. P. Samama, and S. Mobashery.
122. Fisher, J., R. L. Charnas, S. M. Bradley, and J. R. Knowles. 1981. Inacti- 2001. Critical involvement of a carbamylated lysine in catalytic function of
vation of the RTEM ␤-lactamase from Escherichia coli. Interaction of class D ␤-lactamases. Proc. Natl. Acad. Sci. U. S. A. 98:14280–14285.
penam sulfones with enzyme. Biochemistry. 20:2726–2731. 145. Gorbach, S. L. 1994. Antibiotic treatment of anaerobic infections. Clin.
123. Fisher, J., R. L. Charnas, and J. R. Knowles. 1978. Kinetic studies on the Infect. Dis. 18(Suppl. 4):S305–310.
inactivation of Escherichia coli RTEM ␤-lactamase by clavulanic acid. Bio- 146. Guillaume, G., M. Vanhove, J. Lamotte-Brasseur, P. Ledent, M. Jamin, B.
chemistry 17:2180–2184. Joris, and J. M. Frere. 1997. Site-directed mutagenesis of glutamate 166 in
124. Fisher, J. F., S. O. Meroueh, and S. Mobashery. 2005. Bacterial resistance two ␤-lactamases. Kinetic and molecular modeling studies. J. Biol. Chem.
to ␤-lactam antibiotics: compelling opportunism, compelling opportunity. 272:5438–5444.
Chem. Rev. 105:395–424. 147. Hanes, M. S., K. M. Jude, J. M. Berger, R. A. Bonomo, and T. M. Handel.
125. Fisher, J. F., and S. Mobashery. 2009. Three decades of the class A ␤-lac- 2009. Structural and biochemical characterization of the interaction be-
tamase acyl-enzyme. Curr. Protein Pept. Sci. 10:401–407. tween KPC-2 ␤-lactamase and ␤-lactamase inhibitor protein. Biochemistry
126. Foulds, G., W. E. Barth, J. R. Bianchine, A. R. English, D. Girard, S. L. 48:9185–9193.
Hayes, M. O’Brien, and P. Somani. 1980. Pharmacokinetics of CP-45.899 148. Hanson, N. D., and C. C. Sanders. 1999. Regulation of inducible AmpC
and pro-drug CP-47.904 in animals and humans, p. 353–356. In J. D. Nelson ␤-lactamase expression among Enterobacteriaceae. Curr. Pharm. Des.
and C. Grassi (ed.), Current chemotherapy and infectious disease. Amer- 5:881–894.
ican Society for Microbiology, Washington DC. 149. Hardy, L. W., and J. F. Kirsch. 1984. Diffusion-limited component of reactions
127. Fuchs, P. C., A. L. Barry, C. Thornsberry, and R. N. Jones. 1984. In vitro catalyzed by Bacillus cereus ␤-lactamase I. Biochemistry 23:1275–1282.
activity of ticarcillin plus clavulanic acid against 632 clinical isolates. Anti- 150. Hashizume, T., A. Yamaguchi, T. Hirata, and T. Sawai. 1984. Kinetic
microb. Agents Chemother. 25:392–394. studies on the inhibition of Proteus vulgaris ␤-lactamase by imipenem.
128. Galleni, M., G. Amicosante, and J. M. Frere. 1988. A survey of the kinetic Antimicrob. Agents Chemother. 25:149–151.
parameters of class C ␤-lactamases. Cephalosporins and other ␤-lactam 151. Hedstrom, S. A. 1984. Treatment with ␤-lactam antibiotics in skin and soft
compounds. Biochem. J. 255:123–129. tissue infections. Scand. J. Infect. Dis. Suppl. 42:135–142.
129. Galleni, M., and J. M. Frere. 2007. Kinetics of ␤-lactamases and pencillin- 152. Heinze-Krauss, I., P. Angehrn, R. L. Charnas, K. Gubernator, E. M. Gut-
binding proteins, p. 195–213. In R. A. Bonomo and M. E. Tolmasky (ed.), knecht, C. Hubschwerlen, M. Kania, C. Oefner, M. G. Page, S. Sogabe, J. L.
Enzyme-mediated resistance to antibiotics: mechanisms, dissemination, Specklin, and F. Winkler. 1998. Structure-based design of ␤-lactamase
and prospects for inhibition. ASM Press, Washington, DC. inhibitors. 1. Synthesis and evaluation of bridged monobactams. J. Med.
130. Galleni, M., J. Lamotte-Brasseur, G. M. Rossolini, J. Spencer, O. Dideberg, Chem. 41:3961–3971.
and J. M. Frere. 2001. Standard numbering scheme for class B ␤-lactama- 153. Helfand, M. S., C. R. Bethel, A. M. Hujer, K. M. Hujer, V. E. Anderson, and
ses. Antimicrob. Agents Chemother. 45:660–663. R. A. Bonomo. 2003. Understanding resistance to ␤-lactams and ␤-lacta-
131. Ganta, S. R., S. Perumal, S. R. Pagadala, O. Samuelsen, J. Spencer, R. F. mase inhibitors in the SHV ␤-lactamase: lessons from the mutagenesis of
Pratt, and J. D. Buynak. 2009. Approaches to the simultaneous inactivation of SER-130. J. Biol. Chem. 278:52724–52729.
metallo- and serine-␤-lactamases. Bioorg. Med. Chem. Lett. 19:1618–1622. 154. Helfand, M. S., and R. A. Bonomo. 2005. Current challenges in antimicro-
132. Garau, G., C. Bebrone, C. Anne, M. Galleni, J. M. Frere, and O. Dideberg. bial chemotherapy: the impact of extended-spectrum ␤-lactamases and
VOL. 23, 2010 ␤-LACTAMASE INHIBITORS 195

metallo-␤-lactamases on the treatment of resistant Gram-negative patho- resistance genes in multidrug-resistant Acinetobacter sp. isolates from mil-
gens. Curr. Opin. Pharmacol. 5:452–458. itary and civilian patients treated at the Walter Reed Army Medical Center.
155. Helfand, M. S., A. M. Hujer, F. D. Sonnichsen, and R. A. Bonomo. 2002. Antimicrob. Agents Chemother. 50:4114–4123.
Unexpected advanced generation cephalosporinase activity of the M69F 176. Ibuka, A. S., Y. Ishii, M. Galleni, M. Ishiguro, K. Yamaguchi, J. M. Frere,
variant of SHV ␤-lactamase. J. Biol. Chem. 277:47719–47723. H. Matsuzawa, and H. Sakai. 2003. Crystal structure of extended-spectrum
156. Helfand, M. S., M. A. Taracila, L. Borgianni, J. Docquier, A. M. Venkate- ␤-lactamase Toho-1: insights into the molecular mechanism for catalytic
san, and T. S. Mansour. 2007. Inhibition of class A, B and C ␤-lactamases reaction and substrate specificity expansion. Biochemistry 42:10634–10643.
by bicyclic 6-methylidene penems via Michaelis complexes and cyclic enam- 177. Ikonomidis, A., D. Tokatlidou, I. Kristo, D. Sofianou, A. Tsakris, P.
ines, abstr. C1-94. Abstr. 47th Intersci. Conf. Antimicrob. Agents Che- Mantzana, S. Pournaras, and A. N. Maniatis. 2005. Outbreaks in distinct
mother., Chicago, IL. regions due to a single Klebsiella pneumoniae clone carrying a bla VIM-1
157. Helfand, M. S., M. A. Taracila, M. A. Totir, R. A. Bonomo, J. D. Buynak, metallo-␤-lactamase gene. J. Clin. Microbiol. 43:5344–5347.
F. van den Akker, and P. R. Carey. 2007. Raman crystallographic studies of 178. Imada, A., K. Kitano, K. Kintaka, M. Muroi, and M. Asai. 1981. Sulfazecin
the intermediates formed by Ser130Gly SHV, a ␤-lactamase that confers and isosulfazecin, novel ␤-lactam antibiotics of bacterial origin. Nature
resistance to clinical inhibitors. Biochemistry 46:8689–8699. 289:590–591.
158. Helfand, M. S., M. A. Totir, M. P. Carey, A. M. Hujer, R. A. Bonomo, and 179. Imtiaz, U., E. M. Billings, J. R. Knox, E. K. Manavathu, S. A. Lerner, and
P. R. Carey. 2003. Following the reactions of mechanism-based inhibitors S. Mobashery. 1993. Inactivation of class A ␤-lactamases by clavulanic acid:
with ␤-lactamase by Raman crystallography. Biochemistry 42:13386–13392. the role of Arginine 244 in a proposed noncerted sequence of events. J. Am.
159. Henquell, C., C. Chanal, D. Sirot, R. Labia, and J. Sirot. 1995. Molecular Chem. Soc. 115:4435–4442.
characterization of nine different types of mutants among 107 inhibitor- 180. Imtiaz, U., E. M. Billings, J. R. Knox, and S. Mobashery. 1994. A structure-
resistant TEM ␤-lactamases from clinical isolates of Escherichia coli. An- based analysis of the inhibition of class A ␤-lactamases by sulbactam.
timicrob. Agents Chemother. 39:427–430. Biochemistry 33:5728–5738.
160. Henquell, C., D. Sirot, C. Chanal, C. De Champs, P. Chatron, B. Lafeuille, 181. Imtiaz, U., E. K. Manavathu, S. A. Lerner, and S. Mobashery. 1993. Critical
P. Texier, J. Sirot, and R. Cluzel. 1994. Frequency of inhibitor-resistant hydrogen bonding by serine 235 for cephalosporinase activity of TEM-1
TEM ␤-lactamases in Escherichia coli isolates from urinary tract infections b-lactamase. Antimicrob. Agents Chemother. 37:2438–2442.
in France. J. Antimicrob. Chemother. 34:707–714. 182. Jacob, F., B. Joris, S. Lepage, J. Dusart, and J. M. Frere. 1990. Role of the
161. Heritier, C., L. Poirel, D. Aubert, and P. Nordmann. 2003. Genetic and conserved amino acids of the ’SDN⬘ loop (Ser130, Asp131 and Asn132) in
functional analysis of the chromosome-encoded carbapenem-hydrolyzing a class A ␤-lactamase studied by site-directed mutagenesis. Biochem. J.
oxacillinase OXA-40 of Acinetobacter baumannii. Antimicrob. Agents Che- 271:399–406.
mother. 47:268–273. 183. Jacobs, C., J. M. Frere, and S. Normark. 1997. Cytosolic intermediates for
162. Heritier, C., L. Poirel, T. Lambert, and P. Nordmann. 2005. Contribution cell wall biosynthesis and degradation control inducible ␤-lactam resistance
of acquired carbapenem-hydrolyzing oxacillinases to carbapenem resis- in gram-negative bacteria. Cell 88:823–832.
tance in Acinetobacter baumannii. Antimicrob. Agents Chemother. 184. Jacoby, G. A. 2009. AmpC ␤-lactamases. Clin. Microbiol. Rev. 22:161–182.
49:3198–3202. 185. Jacoby, G. A., and I. Carreras. 1990. Activities of ␤-lactam antibiotics
163. Hermann, J. C., C. Hensen, L. Ridder, A. J. Mulholland, and H. D. Holtje. against Escherichia coli strains producing extended-spectrum ␤-lactamases.
2005. Mechanisms of antibiotic resistance: QM/MM modeling of the acy- Antimicrob. Agents Chemother. 34:858–862.
lation reaction of a class A ␤-lactamase with benzylpenicillin. J. Am. Chem. 186. Jacoby, G. A., and A. A. Medeiros. 1991. More extended-spectrum ␤-lac-
Soc. 127:4454–4465. tamases. Antimicrob. Agents Chemother. 35:1697–1704.
164. Hernandez Valladares, M., A. Felici, G. Weber, H. W. Adolph, M. Zeppe- 187. Jacoby, G. A., D. M. Mills, and N. Chow. 2004. Role of ␤-lactamases and
zauer, G. M. Rossolini, G. Amicosante, J. M. Frere, and M. Galleni. 1997. porins in resistance to ertapenem and other ␤-lactams in Klebsiella pneu-
Zn(II) dependence of the Aeromonas hydrophila AE036 metallo-␤-lacta- moniae. Antimicrob. Agents Chemother. 48:3203–3206.
mase activity and stability. Biochemistry 36:11534–11541. 188. Jacoby, G. A., and L. S. Munoz-Price. 2005. The new ␤-lactamases. N Engl.
165. Hernandez Valladares, M., M. Kiefer, U. Heinz, R. P. Soto, W. Meyer- J. Med. 352:380–391.
Klaucke, H. F. Nolting, M. Zeppezauer, M. Galleni, J. M. Frere, G. M. 189. Jamieson, C. E., P. A. Lambert, and I. N. Simpson. 2003. In vitro activities
Rossolini, G. Amicosante, and H. W. Adolph. 2000. Kinetic and spectro- of novel oxapenems, alone and in combination with ceftazidime, against
scopic characterization of native and metal-substituted ␤-lactamase from gram-positive and gram-negative organisms. Antimicrob. Agents Che-
Aeromonas hydrophila AE036. FEBS Lett. 467:221–225. mother. 47:2615–2618.
166. Herzberg, O., and J. Moult. 1987. Bacterial resistance to ␤-lactam antibi- 190. Jamieson, C. E., P. A. Lambert, and I. N. Simpson. 2003. In vitro and in vivo
otics: crystal structure of ␤-lactamase from Staphylococcus aureus PC1 at 2.5 activities of AM-112, a novel oxapenem. Antimicrob. Agents Chemother.
A resolution. Science 236:694–701. 47:1652–1657.
167. Higgins, P. G., H. Wisplinghoff, D. Stefanik, and H. Seifert. 2004. In vitro 191. Jelsch, C., L. Mourey, J. M. Masson, and J. P. Samama. 1993. Crystal
activities of the ␤-lactamase inhibitors clavulanic acid, sulbactam, and structure of Escherichia coli TEM1 ␤-lactamase at 1.8 A resolution. Pro-
tazobactam alone or in combination with ␤-lactams against epidemiologi- teins 16:364–383.
cally characterized multidrug-resistant Acinetobacter baumannii strains. An- 192. Jones, R. N., M. A. Pfaller, P. C. Fuchs, K. Aldridge, S. D. Allen, and E. H.
timicrob. Agents Chemother. 48:1586–1592. Gerlach. 1989. Piperacillin/tazobactam (YTR 830) combination. Comparative
168. Hollingsworth, J. A., J. F. Donohoe, R. Hone, and P. J. Keelan. 1984. antimicrobial activity against 5889 recent aerobic clinical isolates and 60 Bac-
Clinical experience with aztreonam in urinary and respiratory tract infec- teroides fragilis group strains. Diagn. Microbiol. Infect. Dis. 12:489–494.
tions. Curr. Med. Res. Opin. 9:316–322. 193. Juan, C., O. Gutierrez, A. Oliver, J. I. Ayestaran, N. Borrell, and J. L.
169. Hopkins, J. M., and K. J. Towner. 1990. Enhanced resistance to cefotaxime Perez. 2005. Contribution of clonal dissemination and selection of mutants
and imipenem associated with outer membrane protein alterations in En- during therapy to Pseudomonas aeruginosa antimicrobial resistance in an
terobacter aerogenes. J. Antimicrob. Chemother. 25:49–55. intensive care unit setting. Clin. Microbiol. Infect. 11:887–892.
170. Horsfall, L. E., G. Garau, B. M. Lienard, O. Dideberg, C. J. Schofield, J. M. 194. Kaczmarek, F. S., T. D. Gootz, F. Dib-Hajj, W. Shang, S. Hallowell, and M.
Frere, and M. Galleni. 2007. Competitive inhibitors of the CphA metallo- Cronan. 2004. Genetic and molecular characterization of ␤-lactamase-neg-
␤-lactamase from Aeromonas hydrophila. Antimicrob. Agents Chemother. ative ampicillin-resistant Haemophilus influenzae with unusually high resis-
51:2136–2142. tance to ampicillin. Antimicrob. Agents Chemother. 48:1630–1639.
171. Hubschwerlen, C., P. Angehrn, K. Gubernator, M. G. Page, and J. L. 195. Kalp, M., and P. R. Carey. 2008. Carbapenems and SHV-1 ␤-lactamase
Specklin. 1998. Structure-based design of ␤-lactamase inhibitors. 2. Syn- form different acyl-enzyme populations in crystals and solution. Biochem-
thesis and evaluation of bridged sulfactams and oxamazins. J. Med. Chem. istry 47:11830–11837.
41:3972–3975. 196. Kalp, M., A. Sheri, J. D. Buynak, C. R. Bethel, R. A. Bonomo, and P. R.
172. Hugonnet, J. E., L. W. Tremblay, H. I. Boshoff, C. E. Barry III, and J. S. Carey. 2007. Efficient inhibition of class A and class D ␤-lactamases by
Blanchard. 2009. Meropenem-clavulanate is effective against extensively Michaelis complexes. J. Biol. Chem. 282:21588–21591.
drug-resistant Mycobacterium tuberculosis. Science 323:1215–1218. 197. Kalp, M., M. A. Totir, J. D. Buynak, and P. R. Carey. 2009. Different
173. Hujer, A. M., E. Desarbre, A. Endimiani, D. L. Paterson, M. G. P. Page, and intermediate populations formed by tazobactam, sulbactam, and clavu-
R. A. Bonomo. 2007. Activity of BAL30376 against multidrug resistant lanate reacting with SHV-1 ␤-lactamases: Raman crystallographic evidence.
(MDR) Klebsiella and Acinetobacter spp., abstr. F1-333a. Abstr. 47th Inter- J. Am. Chem. Soc. 131:2338–2347.
sci. Conf. Antimicrob. Agents Chemother., Chicago, IL. 198. Kaye, K. S., H. S. Gold, M. J. Schwaber, L. Venkataraman, Y. Qi, P. C. De
174. Hujer, A. M., K. M. Hujer, and R. A. Bonomo. 2001. Mutagenesis of amino acid Girolami, M. H. Samore, G. Anderson, J. K. Rasheed, and F. C. Tenover.
residues in the SHV-1 ␤-lactamase: the premier role of Gly238Ser in penicillin 2004. Variety of ␤-lactamases produced by amoxicillin-clavulanate-resistant
and cephalosporin resistance. Biochim. Biophys. Acta 1547:37–50. Escherichia coli isolated in the northeastern United States. Antimicrob.
175. Hujer, K. M., A. M. Hujer, E. A. Hulten, S. Bajaksouzian, J. M. Adams, Agents Chemother. 48:1520–1525.
C. J. Donskey, D. J. Ecker, C. Massire, M. W. Eshoo, R. Sampath, J. M. 199. Kaye, K. S., A. D. Harris, H. Gold, and Y. Carmeli. 2000. Risk factors for
Thomson, P. N. Rather, D. W. Craft, J. T. Fishbain, A. J. Ewell, M. R. recovery of ampicillin-sulbactam-resistant Escherichia coli in hospitalized
Jacobs, D. L. Paterson, and R. A. Bonomo. 2006. Analysis of antibiotic patients. Antimicrob. Agents Chemother. 44:1004–1009.
196 DRAWZ AND BONOMO CLIN. MICROBIOL. REV.

200. Ke, W., C. R. Bethel, J. M. Thomson, R. A. Bonomo, and F. van den Akker. a contributing factor to ␤-lactam resistance. Antimicrob. Agents Che-
2007. Crystal structure of KPC-2: insights into carbapenemase activity in mother. 38:1742–1752.
class A ␤-lactamases. Biochemistry 46:5732–5740. 225. Li, X. Z., H. Nikaido, and K. Poole. 1995. Role of MexA-MexB-OprM in
201. Kiener, P. A., and S. G. Waley. 1978. Reversible inhibitors of penicillinases. antibiotic efflux in Pseudomonas aeruginosa. Antimicrob. Agents Che-
Biochem. J. 169:197–204. mother. 39:1948–1953.
202. Kirby, W. M. 1944. Extraction of a highly potent penicillin inactivator from 226. Li, X. Z., L. Zhang, and K. Poole. 2000. Interplay between the MexA-
penicillin resistant staphylococci. Science 99:452–453. MexB-OprM multidrug efflux system and the outer membrane barrier in
203. Kitzis, M. D., B. Ferre, A. Coutrot, J. F. Acar, and L. Gutmann. 1989. In the multiple antibiotic resistance of Pseudomonas aeruginosa. J. Antimi-
vitro activity of combinations of ␤-lactam antibiotics with ␤-lactamase in- crob. Chemother. 45:433–436.
hibitors against cephalosporinase-producing bacteria. Eur. J. Clin. Micro- 227. Lienard, B. M., G. Garau, L. Horsfall, A. I. Karsisiotis, C. Damblon, P.
biol. Infect. Dis. 8:783–788. Lassaux, C. Papamicael, G. C. Roberts, M. Galleni, O. Dideberg, J. M. Frere,
204. Knothe, H., P. Shah, V. Krcmery, M. Antal, and S. Mitsuhashi. 1983. and C. J. Schofield. 2008. Structural basis for the broad-spectrum inhibition of
Transferable resistance to cefotaxime, cefoxitin, cefamandole and cefuro- metallo-␤-lactamases by thiols. Org. Biomol. Chem. 6:2282–2294.
xime in clinical isolates of Klebsiella pneumoniae and Serratia marcescens. 228. Lienard, B. M., R. Huting, P. Lassaux, M. Galleni, J. M. Frere, and C. J.
Infection 11:315–317. Schofield. 2008. Dynamic combinatorial mass spectrometry leads to metallo-
205. Knott-Hunziker, V., S. Petursson, S. G. Waley, B. Jaurin, and T. Grund- ␤-lactamase inhibitors. J. Med. Chem. 51:684–688.
strom. 1982. The acyl-enzyme mechanism of ␤-lactamase action. The evi- 229. Lin, S., M. Thomas, S. Mark, V. Anderson, and R. A. Bonomo. 1999. OHIO-1
dence for class C ␤-lactamases. Biochem. J. 207:315–322. ␤-lactamase mutants: the Arg244Ser mutant and resistance to ␤-lactams and
206. Knox, J. R. 1995. Extended-spectrum and inhibitor-resistant TEM-type ␤-lactamase inhibitors. Biochim. Biophys. Acta 1432:125–136.
␤-lactamases: mutations, specificity, and three-dimensional structure. An- 230. Lindquist, R. N., and C. Terry. 1974. Inhibition of subtilisin by boronic
timicrob. Agents Chemother. 39:2593–2601. acids, potential analogs of tetrahedral reaction intermediates. Arch. Bio-
207. Knox, J. R., P. C. Moews, W. A. Escobar, and A. L. Fink. 1993. A catalyt- chem. Biophys. 160:135–144.
ically-impaired class A ␤-lactamase: 2 A crystal structure and kinetics of the 231. Lister, P. D., V. M. Gardner, and C. C. Sanders. 1999. Clavulanate induces
Bacillus licheniformis E166A mutant. Protein Eng. 6:11–18. expression of the Pseudomonas aeruginosa AmpC cephalosporinase at phys-
208. Kropp, H., L. Gerckens, J. G. Sundelof, and F. M. Kahan. 1985. Antibac- iologically relevant concentrations and antagonizes the antibacterial activity
terial activity of imipenem: the first thienamycin antibiotic. Rev. Infect. Dis. of ticarcillin. Antimicrob. Agents Chemother. 43:882–889.
7(Suppl. 3):S389–S410. 232. Livermore, D. M. 1998. ␤-lactamase-mediated resistance and opportunities
209. Kuzin, A. P., M. Nukaga, Y. Nukaga, A. Hujer, R. A. Bonomo, and J. R. for its control. J. Antimicrob. Chemother. 41(Suppl. D):25–41.
Knox. 2001. Inhibition of the SHV-1 ␤-lactamase by sulfones: crystallo- 233. Livermore, D. M. 1987. Clinical significance of ␤-lactamase induction and
graphic observation of two reaction intermediates with tazobactam. Bio- stable derepression in gram-negative rods. Eur. J. Clin. Microbiol. 6:439–445.
chemistry 40:1861–1866. 234. Livermore, D. M. 1993. Determinants of the activity of ␤-lactamase inhib-
210. Kuzin, A. P., M. Nukaga, Y. Nukaga, A. M. Hujer, R. A. Bonomo, and J. R. itor combinations. J. Antimicrob. Chemother. 31(Suppl. A):9–21.
Knox. 1999. Structure of the SHV-1 ␤-lactamase. Biochemistry 38:5720–5727. 235. Livermore, D. M. 1992. Interplay of impermeability and chromosomal
211. Labia, R., A. Morand, and J. Peduzzi. 1986. Timentin and ␤-lactamases. J. ␤-lactamase activity in imipenem-resistant Pseudomonas aeruginosa. Anti-
Antimicrob. Chemother. 17(Suppl. C):17–26. microb. Agents Chemother. 36:2046–2048.
212. Laible, G., B. G. Spratt, and R. Hakenbeck. 1991. Interspecies recombina- 236. Livermore, D. M. 2001. Of Pseudomonas, porins, pumps and carbapenems.
tional events during the evolution of altered PBP 2x genes in penicillin- J. Antimicrob. Chemother. 47:247–250.
resistant clinical isolates of Streptococcus pneumoniae. Mol. Microbiol.
237. Livermore, D. M., M. Akova, P. J. Wu, and Y. J. Yang. 1989. Clavulanate
5:1993–2002.
and ␤-lactamase induction. J. Antimicrob. Chemother. 24(Suppl. B):23–33.
213. Lakaye, B., A. Dubus, S. Lepage, S. Groslambert, and J. M. Frere. 1999.
238. Livermore, D. M., and H. Y. Chen. 1997. Potentiation of ␤-lactams against
When drug inactivation renders the target irrelevant to antibiotic resis-
Pseudomonas aeruginosa strains by Ro 48–1256, a bridged monobactam
tance: a case story with ␤-lactams. Mol. Microbiol. 31:89–101.
inhibitor of AmpC ␤-lactamases. J. Antimicrob. Chemother. 40:335–343.
214. Lamotte-Brasseur, J., G. Dive, O. Dideberg, P. Charlier, J. M. Frere, and
239. Livermore, D. M., R. Hope, S. Mushtaq, and M. Warner. 2008. Orthodox
J. M. Ghuysen. 1991. Mechanism of acyl transfer by the class A serine
and unorthodox clavulanate combinations against extended-spectrum
␤-lactamase of Streptomyces albus G. Biochem. J. 279:213–221.
␤-lactamase producers. Clin. Microbiol. Infect. 14(Suppl. 1):189–193.
215. Lan, C. K., P. R. Hsueh, W. W. Wong, C. P. Fung, Y. T. Lau, J. Y. Yeung, G. T.
240. Livermore, D. M., S. Mushtaq, M. Warner, C. Miossec, and N. Woodford.
Young, and C. C. Su. 2003. Association of antibiotic utilization measures and
2008. NXL104 combinations versus Enterobacteriaceae with CTX-M ex-
reduced incidence of infections with extended-spectrum ␤-lactamase-produc-
tended-spectrum ␤-lactamases and carbapenemases. J. Antimicrob. Che-
ing organisms. J. Microbiol. Immunol. Infect. 36:182–186.
mother. 62:1053–1056.
216. Landman, D., M. Chockalingam, and J. M. Quale. 1999. Reduction in the
incidence of methicillin-resistant Staphylococcus aureus and ceftazidime- 241. Lobkovsky, E., E. M. Billings, P. C. Moews, J. Rahil, R. F. Pratt, and J. R.
resistant Klebsiella pneumoniae following changes in a hospital antibiotic Knox. 1994. Crystallographic structure of a phosphonate derivative of the
formulary. Clin. Infect. Dis. 28:1062–1066. Enterobacter cloacae P99 cephalosporinase: mechanistic interpretation of a
217. Laraki, N., N. Franceschini, G. M. Rossolini, P. Santucci, C. Meunier, E. de ␤-lactamase transition-state analog. Biochemistry 33:6762–6772.
Pauw, G. Amicosante, J. M. Frere, and M. Galleni. 1999. Biochemical 242. Lodise, T. P., Jr., B. Lomaestro, K. A. Rodvold, L. H. Danziger, and G. L.
characterization of the Pseudomonas aeruginosa 101/1477 metallo-␤-lacta- Drusano. 2004. Pharmacodynamic profiling of piperacillin in the presence of
mase IMP-1 produced by Escherichia coli. Antimicrob. Agents Chemother. tazobactam in patients through the use of population pharmacokinetic models
43:902–906. and Monte Carlo simulation. Antimicrob. Agents Chemother. 48:4718–4724.
218. Leflon-Guibout, V., V. Speldooren, B. Heym, and M. Nicolas-Chanoine. 243. Majumdar, S., S. A. Adediran, M. Nukaga, and R. F. Pratt. 2005. Inhibition of
2000. Epidemiological survey of amoxicillin-clavulanate resistance and cor- class D ␤-lactamases by diaroyl phosphates. Biochemistry 44:16121–16129.
responding molecular mechanisms in Escherichia coli isolates in France: 244. Majumdar, S., and R. F. Pratt. 2009. Inhibition of class A and C ␤-lacta-
new genetic features of blaTEM genes. Antimicrob. Agents Chemother. mases by diaroyl phosphates. Biochemistry 48:8285–8292.
44:2709–2714. 245. Majumdar, S., and R. F. Pratt. 2009. Intramolecular cooperativity in the
219. Leflon-Guibout, V., G. Ternat, B. Heym, and M. H. Nicolas-Chanoine. 2002. reaction of diacyl phosphates with serine ␤-lactamases. Biochemistry 48:
Exposure to co-amoxiclav as a risk factor for co-amoxiclav-resistant Escherichia 8293–8298.
coli urinary tract infection. J. Antimicrob. Chemother. 49:367–371. 246. Reference deleted.
220. Lenfant, F., R. Labia, and J. M. Masson. 1991. Replacement of lysine 234 247. Martinez, J. L., E. Cercenado, M. Rodriguez-Creixems, M. F. Vincente-
affects transition state stabilization in the active site of ␤-lactamase TEM1. Perez, A. Delgado-Iribarren, and F. Baquero. 1987. Resistance to ␤-lactam/
J. Biol. Chem. 266:17187–17194. clavulanate. Lancet ii:1473.
221. Levasseur, P., A. Girard, L. Lavallade, C. Miossec, D. M. Shlaes, 248. Martinez, J. L., M. F. Vicente, A. Delgado-Iribarren, J. C. Perez-Diaz, and
M. T. Black, J. L. Pace, and K. Coleman. 2009. Efficacy of ceftazidime/ F. Baquero. 1989. Small plasmids are involved in amoxicillin-clavulanate
NXL104 combination in murine septicaemia caused by CTX-M-producing resistance in Escherichia coli. Antimicrob. Agents Chemother. 33:595.
Enterobacteriaceae species, abstr. A1-005. Abstr. 49th Intersci. Conf. Anti- 249. Massova, I., and S. Mobashery. 1998. Kinship and diversification of bacte-
microb. Agents Chemother., San Francisco, CA. rial penicillin-binding proteins and ␤-lactamases. Antimicrob. Agents Che-
222. Levin, A. S. 2002. Multiresistant Acinetobacter infections: a role for sulbac- mother. 42:1–17.
tam combinations in overcoming an emerging worldwide problem. Clin. 250. Matagne, A., P. Ledent, D. Monnaie, A. Felici, M. Jamin, X. Raquet, M.
Microbiol. Infect. 8:144–153. Galleni, D. Klein, I. Francois, and J. M. Frere. 1995. Kinetic study of
223. Li, N., and R. F. Pratt. 1998. Inhibition of serine ␤-lactamases by acyl interaction between BRL 42715, ␤-lactamases, and D-alanyl-D-alanine pep-
phosph(on)ates: a new source of inert acyl [and phosphyl] enzymes. tidases. Antimicrob. Agents Chemother. 39:227–231.
J. Chem. Soc. 120:4264–4268. 251. Matthew, M., R. W. Hedges, and J. T. Smith. 1979. Types of ␤-lactamase
224. Li, X. Z., D. Ma, D. M. Livermore, and H. Nikaido. 1994. Role of efflux determined by plasmids in gram-negative bacteria. J. Bacteriol. 138:657–662.
pump(s) in intrinsic resistance of Pseudomonas aeruginosa: active efflux as 252. Maveyraud, L., D. Golemi-Kotra, A. Ishiwata, O. Meroueh, S. Mobashery, and
VOL. 23, 2010 ␤-LACTAMASE INHIBITORS 197

J. P. Samama. 2002. High-resolution X-ray structure of an acyl-enzyme species 275. Muder, R. R. 2007. Optimizing therapy for Stenotrophomonas maltophilia.
for the class D OXA-10 ␤-lactamase. J. Am. Chem. Soc. 124:2461–2465. Semin. Respir. Crit. Care Med. 28:672–677.
253. Maveyraud, L., D. Golemi, L. P. Kotra, S. Tranier, S. Vakulenko, S. Mo- 276. Mulvey, M. R., D. A. Boyd, L. Baker, O. Mykytczuk, E. M. Reis, M. D.
bashery, and J. P. Samama. 2000. Insights into class D ␤-lactamases are Asensi, D. P. Rodrigues, and L. K. Ng. 2004. Characterization of a Salmo-
revealed by the crystal structure of the OXA10 enzyme from Pseudomonas nella enterica serovar Agona strain harbouring a class 1 integron containing
aeruginosa. Structure 8:1289–1298. novel OXA-type ␤-lactamase (blaOXA-53) and 6⬘-N-aminoglycoside acetyl-
254. Maveyraud, L., I. Massova, C. Birck, K. Miyashita, J. P. Samama, and S. transferase genes [aac(6⬘)-I30]. J. Antimicrob. Chemother. 54:354–359.
Mobashery. 1996. Crystal structure of 6 ␣-(hydroxymethyl)penicillanate 277. Murphy, B. P., and R. F. Pratt. 1988. Evidence for an oxyanion hole in
complexed to the TEM-1 ␤-lactamase from Escherichia coli: evidence on serine ␤-lactamases and DD-peptidases. Biochem. J. 256:669–672.
the mechanism of action of a novel inhibitor designed by a computer-aided 278. Mussi, M. A., A. S. Limansky, and A. M. Viale. 2005. Acquisition of
process. J. Am. Chem. Soc. 118:7435–7440. resistance to carbapenems in multidrug-resistant clinical strains of Acineto-
255. Maveyraud, L., L. Mourey, L. P. Kotra, J.-D. Pedelacq, V. Guillet, and S. bacter baumannii: natural insertional inactivation of a gene encoding a
Mobashery. 1998. Structural basis for clinical longevity of carbapenem member of a novel family of ␤-barrel outer membrane proteins. Antimi-
antibiotics in the face of challenge by the common class A ␤-lactamases crob. Agents Chemother. 49:1432–1440.
from the antibiotic-resistant bacteria. J. Am. Chem. Soc. 120:9748–9752. 279. Naas, T., and P. Nordmann. 1999. OXA-type ␤-lactamases. Curr. Pharm.
256. Medeiros, A. A. 1997. Evolution and dissemination of ␤-lactamases accel- Des. 5:865–879.
erated by generations of ␤-lactam antibiotics. Clin. Infect. Dis. 24(Suppl. 280. Naas, T., L. Poirel, and P. Nordmann. 2008. Minor extended-spectrum
1):S19–S45. ␤-lactamases. Clin. Microbiol. Infect. 14(Suppl. 1):42–52.
257. Mendelman, P. M., D. O. Chaffin, and G. Kalaitzoglou. 1990. Penicillin- 281. Nagano, R., Y. Adachi, T. Hashizume, and H. Morishima. 2000. In vitro
binding proteins and ampicillin resistance in Haemophilus influenzae. J. antibacterial activity and mechanism of action of J-111,225, a novel 1␤-
Antimicrob. Chemother. 25:525–534. methylcarbapenem, against transferable IMP-1 metallo-␤-lactamase pro-
258. Mendonca, N., V. Manageiro, F. Robin, M. J. Salgado, E. Ferreira, M. ducers. J. Antimicrob. Chemother. 45:271–276.
Canica, and R. Bonnet. 2008. The Lys234Arg substitution in the enzyme 282. Nagano, R., Y. Adachi, H. Imamura, K. Yamada, T. Hashizume, and H.
SHV-72 is a determinant for resistance to clavulanic acid inhibition. Anti- Morishima. 1999. Carbapenem derivatives as potential inhibitors of various
microb. Agents Chemother. 52:1806–1811. ␤-lactamases, including class B metallo-␤-lactamases. Antimicrob. Agents
259. Merdjan, H., A. Girard, C. Miossec, M. Robertson, and P. Levasseur. 2009. Chemother. 43:2497–2503.
Pharmacokinetics and efficacy of ceftazidime / NXL104 combination in a 283. Naumovski, L., J. P. Quinn, D. Miyashiro, M. Patel, K. Bush, S. B. Singer,
murine pneumonia model caused by an AmpC-producing Klebsiella pneu- D. Graves, T. Palzkill, and A. M. Arvin. 1992. Outbreak of ceftazidime
moniae, abstr. A1-006. Abstr. 49th Intersci. Conf. Antimicrob. Agents Che- resistance due to a novel extended-spectrum ␤-lactamase in isolates from
mother., San Francisco, CA. cancer patients. Antimicrob. Agents Chemother. 36:1991–1996.
260. Meroueh, S., J. Cha, and S. Mobashery. 2007. Inhibition of class A ␤-lac- 284. Nauton, L., R. Kahn, G. Garau, J. F. Hernandez, and O. Dideberg. 2008.
tamases, p. 101–114. In R. A. Bonomo and M. E. Tolmasky (ed.), Enzyme- Structural insights into the design of inhibitors for the L1 metallo-␤-lacta-
mediated resistance to antibiotics: mechanisms, dissemination, and pros- mase from Stenotrophomonas maltophilia. J. Mol. Biol. 375:257–269.
pects for inhibition. ASM Press, Washingtion, DC. 285. Neu, H. C., and K. P. Fu. 1978. Clavulanic acid, a novel inhibitor of
261. Meroueh, S. O., J. F. Fisher, H. B. Schlegel, and S. Mobashery. 2005. Ab ␤-lactamases. Antimicrob. Agents Chemother. 14:650–655.
initio QM/MM study of class A ␤-lactamase acylation: dual participation of 286. Nikaido, H. 1994. Prevention of drug access to bacterial targets: permeabil-
Glu166 and Lys73 in a concerted base promotion of Ser70. J. Am. Chem. ity barriers and active efflux. Science 264:382–388.
Soc. 127:15397–15407. 287. Nikaido, H. 1998. The role of outer membrane and efflux pumps in the
262. Meroueh, S. O., P. Roblin, D. Golemi, L. Maveyraud, S. B. Vakulenko, Y. resistance of gram-negative bacteria. Can we improve drug access? Drug
Zhang, J. P. Samama, and S. Mobashery. 2002. Molecular dynamics at the Resist. Updat. 1:93–98.
root of expansion of function in the M69L inhibitor-resistant TEM ␤-lac- 288. Nikaido, H., W. Liu, and E. Y. Rosenberg. 1990. Outer membrane perme-
tamase from Escherichia coli. J. Am. Chem. Soc. 124:9422–9430. ability and ␤-lactamase stability of dipolar ionic cephalosporins containing
263. Michaux, C., P. Charlier, J. M. Frere, and J. Wouters. 2005. Crystal structure methoxyimino substituents. Antimicrob. Agents Chemother. 34:337–342.
of BRL 42715, C6-(N1-methyl-1,2,3-triazolylmethylene)penem, in complex 289. Nishida, K., C. Kunugita, T. Uji, F. Higashitani, A. Hyodo, N. Unemi, S. N.
with Enterobacter cloacae 908R ␤-lactamase: evidence for a stereoselective Maiti, O. A. Phillips, P. Spevak, K. P. Atchison, S. M. Salama, H. Atwal,
mechanism from docking studies. J. Am. Chem. Soc. 127:3262–3263. and R. G. Micetich. 1999. In vitro and in vivo activities of Syn2190, a novel
264. Miller, J. M., C. N. Baker, and C. Thornsberry. 1978. Inhibition of ␤-lac- ␤-lactamase inhibitor. Antimicrob. Agents Chemother. 43:1895–1900.
tamase in Neisseria gonorrhoeae by sodium clavulanate. Antimicrob. Agents 290. Nordmann, P., G. Cuzon, and T. Naas. 2009. The real threat of Klebsiella
Chemother. 14:794–796. pneumoniae carbapenemase-producing bacteria. Lancet Infect. Dis. 9:228–236.
265. Minasov, G., X. Wang, and B. K. Shoichet. 2002. An ultrahigh resolution 291. Nordmann, P., S. Mariotte, T. Naas, R. Labia, and M. H. Nicolas. 1993.
structure of TEM-1 ␤-lactamase suggests a role for Glu166 as the general Biochemical properties of a carbapenem-hydrolyzing ␤-lactamase from En-
base in acylation. J. Am. Chem. Soc. 124:5333–5340. terobacter cloacae and cloning of the gene into Escherichia coli. Antimicrob.
266. Miro, E., F. Navarro, B. Mirelis, M. Sabate, A. Rivera, P. Coll, and G. Agents Chemother. 37:939–946.
Prats. 2002. Prevalence of clinical isolates of Escherichia coli producing 292. Nordmann, P., and L. Poirel. 2002. Emerging carbapenemases in Gram-
inhibitor-resistant beta-lactamases at a University Hospital in Barcelona, negative aerobes. Clin. Microbiol. Infect. 8:321–331.
Spain, over a 3-year period. Antimicrob. Agents Chemother. 46:3991–3994. 293. Nuesch-Inderbinen, M. T., H. Hachler, and F. H. Kayser. 1995. New system
267. Moews, P. C., J. R. Knox, O. Dideberg, P. Charlier, and J. M. Frere. 1990. based on site-directed mutagenesis for highly accurate comparison of re-
␤-lactamase of Bacillus licheniformis 749/C at 2 A resolution. Proteins sistance levels conferred by SHV ␤-lactamases. Antimicrob. Agents Che-
7:156–171. mother. 39:1726–1730.
268. Mollard, C., C. Moali, C. Papamicael, C. Damblon, S. Vessilier, G. Ami- 294. Nukaga, M., T. Abe, A. M. Venkatesan, T. S. Mansour, R. A. Bonomo, and
cosante, C. J. Schofield, M. Galleni, J. M. Frere, and G. C. Roberts. 2001. J. R. Knox. 2003. Inhibition of class A and class C ␤-lactamases by penems:
Thiomandelic acid, a broad spectrum inhibitor of zinc ␤-lactamases: kinetic crystallographic structures of a novel 1,4-thiazepine intermediate. Biochem-
and spectroscopic studies. J. Biol. Chem. 276:45015–45023. istry 42:13152–13159.
269. Moloughney, J. G., J. D. Thomas, and J. H. Toney. 2005. Novel IMP-1 295. Nukaga, M., C. R. Bethel, J. M. Thomson, A. M. Hujer, A. Distler, V. E.
metallo-␤-lactamase inhibitors can reverse meropenem resistance in Esch- Anderson, J. R. Knox, and R. A. Bonomo. 2008. Inhibition of class A
erichia coli expressing IMP-1. FEMS Microbiol. Lett. 243:65–71. ␤-lactamases by carbapenems: crystallographic observation of two confor-
270. Monks, J., and S. G. Waley. 1988. Imipenem as substrate and inhibitor of mations of meropenem in SHV-1. J. Am. Chem. Soc. 130:12656–12662.
␤-lactamases. Biochem. J. 253:323–328. 296. Reference deleted.
271. Monnaie, D., A. Dubus, and J. M. Frere. 1994. The role of lysine-67 in a 297. Nukaga, M., S. Kumar, K. Nukaga, R. F. Pratt, and J. R. Knox. 2004.
class C ␤-lactamase is mainly electrostatic. Biochem. J. 302:1–4. Hydrolysis of third-generation cephalosporins by class C ␤-lactamases.
272. Morandi, F., E. Caselli, S. Morandi, P. J. Focia, J. Blazquez, B. K. Shoichet, Structures of a transition state analog of cefotaxime in wild-type and ex-
and F. Prati. 2003. Nanomolar inhibitors of AmpC ␤-lactamase. J. Am. tended spectrum enzymes. J. Biol. Chem. 279:9344–9352.
Chem. Soc. 125:685–695. 298. Nukaga, M., K. Mayama, A. M. Hujer, R. A. Bonomo, and J. R. Knox. 2003.
273. Mourey, L., L. P. Kotra, J. Bellettini, A. Bulychev, M. O’Brien, M. J. Miller, Ultrahigh resolution structure of a class A ␤-lactamase: on the mechanism
S. Mobashery, and J. P. Samama. 1999. Inhibition of the broad spectrum and specificity of the extended-spectrum SHV-2 enzyme. J. Mol. Biol.
nonmetallocarbapenamase of class A (NMC-A) ␤-lactamase from Entero- 328:289–301.
bacter cloacae by monocyclic ␤-lactams. J. Biol. Chem. 274:25260–25265. 299. Oefner, C., A. D’Arcy, J. J. Daly, K. Gubernator, R. L. Charnas, I. Heinze,
274. Mourey, L., K. Miyashita, P. Swaren, A. Bulychev, J. P. Samama, and S. C. Hubschwerlen, and F. K. Winkler. 1990. Refined crystal structure of
Mobashery. 1998. Inhibition of the NMC-A ␤-lactamase by a penicillanic ␤-lactamase from Citrobacter freundii indicates a mechanism for ␤-lactam
acid derivative and the structural bases for the increase in substrate profile hydrolysis. Nature 343:284–288.
of this antibiotic resistance enzyme. J. Am. Chem. Soc. 120:9382–9383. 300. Oliphant, A. R., and K. Struhl. 1989. An efficient method for generating
198 DRAWZ AND BONOMO CLIN. MICROBIOL. REV.

proteins with altered enzymatic properties: application to ␤-lactamase. multiple-drug resistance in Klebsiella pneumoniae. Infect. Control Hosp.
Proc. Natl. Acad. Sci. U. S. A. 86:9094–9098. Epidemiol. 21:455–458.
301. Oliver, A., M. Perez-Vazquez, M. Martinez-Ferrer, F. Baquero, L. De 324. Paukner, S., L. Hesse, A. Prezelj, T. Solmajer, and U. Urleb. 2009. In vitro
Rafael, and R. Canton. 1999. Ampicillin-sulbactam and amoxicillin-clavu- activity of LK-157, a novel tricyclic carbapenem as broad-spectrum ␤-lac-
lanate susceptibility testing of Escherichia coli isolates with different beta- tamase inhibitor. Antimicrob. Agents Chemother. 53:505–511.
lactam resistance phenotypes. Antimicrob. Agents Chemother. 43:862–867. 325. Paul-Soto, R., R. Bauer, J. M. Frere, M. Galleni, W. Meyer-Klaucke, H.
302. Oliver, A., L. M. Weigel, J. K. Rasheed, J. E. McGowan Juniorperiod, Jr., Nolting, G. M. Rossolini, D. de Seny, M. Hernandez-Valladares, M. Zeppe-
P. Raney, and F. C. Tenover. 2002. Mechanisms of decreased susceptibility zauer, and H. W. Adolph. 1999. Mono- and binuclear Zn2⫹-␤-lactamase.
to cefpodoxime in Escherichia coli. Antimicrob. Agents Chemother. 46: Role of the conserved cysteine in the catalytic mechanism. J. Biol. Chem.
3829–3836. 274:13242–13249.
303. Olsen, L., S. Jost, H. W. Adolph, I. Pettersson, L. Hemmingsen, and F. S. 326. Paul-Soto, R., M. Hernandez-Valladares, M. Galleni, R. Bauer, M. Zeppe-
Jorgensen. 2006. New leads of metallo-␤-lactamase inhibitors from struc- zauer, J. M. Frere, and H. W. Adolph. 1998. Mono- and binuclear Zn-␤-
ture-based pharmacophore design. Bioorg. Med. Chem. 14:2627–2635. lactamase from Bacteroides fragilis: catalytic and structural roles of the zinc
304. Orencia, M. C., J. S. Yoon, J. E. Ness, W. P. Stemmer, and R. C. Stevens. ions. FEBS Lett. 438:137–140.
2001. Predicting the emergence of antibiotic resistance by directed evolu- 327. Payne, D. J., J. H. Bateson, B. C. Gasson, D. Proctor, T. Khushi, T. H.
tion and structural analysis. Nat. Struct. Biol. 8:238–242. Farmer, D. A. Tolson, D. Bell, P. W. Skett, A. C. Marshall, R. Reid, L.
305. Osuna, J., H. Viadiu, A. L. Fink, and X. Soberon. 1995. Substitution of Asp Ghosez, Y. Combret, and J. Marchand-Brynaert. 1997. Inhibition of me-
for Asn at position 132 in the active site of TEM ␤-lactamase. Activity tallo-␤-lactamases by a series of mercaptoacetic acid thiol ester derivatives.
toward different substrates and effects of neighboring residues. J. Biol. Antimicrob. Agents Chemother. 41:135–140.
Chem. 270:775–780. 328. Payne, D. J., R. Cramp, D. J. Winstanley, and D. J. Knowles. 1994. Compar-
306. Oteo, J., A. Delgado-Iribarren, D. Vega, V. Bautista, M. C. Rodriguez, M. ative activities of clavulanic acid, sulbactam, and tazobactam against clinically
Velasco, J. M. Saavedra, M. Perez-Vazquez, S. Garcia-Cobos, L. Martinez- important ␤-lactamases. Antimicrob. Agents Chemother. 38:767–772.
Martinez, and J. Campos. 2008. Emergence of imipenem resistance in clinical 329. Payne, D. J., J. A. Hueso-Rodriguez, H. Boyd, N. O. Concha, C. A. Janson,
Escherichia coli during therapy. Int. J. Antimicrob. Agents 32:534–537. M. Gilpin, J. H. Bateson, C. Cheever, N. L. Niconovich, S. Pearson, S.
307. Padayatti, P. S., M. S. Helfand, M. A. Totir, M. P. Carey, P. R. Carey, R. A. Rittenhouse, D. Tew, E. Diez, P. Perez, J. De La Fuente, M. Rees, and A.
Bonomo, and F. van den Akker. 2005. High resolution crystal structures of Rivera-Sagredo. 2002. Identification of a series of tricyclic natural products
the trans-enamine intermediates formed by sulbactam and clavulanic acid as potent broad-spectrum inhibitors of metallo-␤-lactamases. Antimicrob.
and E166A SHV-1 ␤-lactamase. J. Biol. Chem. 280:34900–34907. Agents Chemother. 46:1880–1886.
308. Padayatti, P. S., M. S. Helfand, M. A. Totir, M. P. Carey, A. M. Hujer, P. R. 330. Pelto, R. B., and R. F. Pratt. 2008. Kinetics and mechanism of inhibition of
Carey, R. A. Bonomo, and F. van den Akker. 2004. Tazobactam forms a a serine ␤-lactamase by O-aryloxycarbonyl hydroxamates. Biochemistry 47:
stoichiometric trans-enamine intermediate in the E166A variant of SHV-1 12037–12046.
␤-lactamase: 1.63 A crystal structure. Biochemistry 43:843–848. 331. Perez-Moreno, M. O., M. Perez-Moreno, M. Carulla, C. Rubio, A. M. Jardi,
309. Padayatti, P. S., A. Sheri, M. A. Totir, M. S. Helfand, M. P. Carey, V. E. and J. Zaragoza. 2004. Mechanisms of reduced susceptibility to amoxycil-
Anderson, P. R. Carey, C. R. Bethel, R. A. Bonomo, J. D. Buynak, and F. van lin-clavulanic acid in Escherichia coli strains from the health region of
den Akker. 2006. Rational design of a ␤-lactamase inhibitor achieved via Tortosa (Catalonia, Spain). Clin. Microbiol. Infect. 10:234–241.
stabilization of the trans-enamine intermediate: 1.28 A crystal structure of wt 332. Perez, F., A. Endimiani, K. M. Hujer, and R. A. Bonomo. 2007. The
SHV-1 complex with a penam sulfone. J. Am. Chem. Soc. 128:13235–13242. continuing challenge of ESBLs. Curr. Opin. Pharmacol. 7:459–469.
310. Paetzel, M., F. Danel, L. de Castro, S. C. Mosimann, M. G. Page, and N. C. 333. Perez, F., A. M. Hujer, K. M. Hujer, B. K. Decker, P. N. Rather, and R. A.
Strynadka. 2000. Crystal structure of the class D ␤-lactamase OXA-10. Nat. Bonomo. 2007. Global challenge of multidrug-resistant Acinetobacter bau-
Struct. Biol. 7:918–925. mannii. Antimicrob. Agents Chemother. 51:3471–3484.
311. Pagan-Rodriguez, D., X. Zhou, R. Simmons, C. R. Bethel, A. M. Hujer, 334. Pernot, L., F. Frenois, T. Rybkine, G. L’Hermite, S. Petrella, J. Delettre, V.
M. S. Helfand, Z. Jin, B. Guo, V. E. Anderson, L. M. Ng, and R. A. Bonomo. Jarlier, E. Collatz, and W. Sougakoff. 2001. Crystal structures of the class D
2004. Tazobactam inactivation of SHV-1 and the inhibitor-resistant ␤-lactamase OXA-13 in the native form and in complex with meropenem.
Ser1303Gly SHV-1 ␤-lactamase: insights into the mechanism of inhibition. J. Mol. Biol. 310:859–874.
J. Biol. Chem. 279:19494–19501. 335. Petersen, P. J., C. H. Jones, A. M. Venkatesan, T. S. Mansour, S. J. Projan,
312. Page, M. G. 2000. ␤-Lactamase inhibitors. Drug Resist. Updat. 3:109–125. and P. A. Bradford. 2009. Establishment of in vitro susceptibility testing
313. Page, M. G. 2007. Resistance mediated by penicllin-binding proteins, p. methodologies and comparative activities of piperacillin in combination
81–99. In R. A. Bonomo and M. E. Tolmasky (ed.), Enzyme-mediated with the penem ␤-lactamase inhibitor BLI-489. Antimicrob. Agents Che-
resistance to antibiotics: mechanisms, dissemination, and prospects for In- mother. 53:370–384.
hibition. ASM Press, Washington, DC. 336. Peterson, L. R. 2008. Antibiotic policy and prescribing strategies for therapy
314. Page, M. G., and J. Heim. 2009. New molecules from old classes: revisiting of extended-spectrum ␤-lactamase-producing Enterobacteriaceae: the role
the development of ␤-lactams. Drugs 12:561–565. of piperacillin-tazobactam. Clin. Microbiol. Infect. 14(Suppl. 1):181–184.
315. Page, M. G., and J. Heim. 2009. Prospects for the next anti-Pseudomonas 337. Petit, A., L. Maveyraud, F. Lenfant, J. P. Samama, R. Labia, and J. M.
drug. Curr. Opin. Pharmacol. 9:558–565. Masson. 1995. Multiple substitutions at position 104 of ␤-lactamase
316. Page, M. G. P., E. Desarbre, C. Geier, and B. Hofer. 2007. Activity of TEM-1: assessing the role of this residue in substrate specificity. Biochem.
monobactam ␤-lactamase inhibitor combinations against ␤-lactam-resistant J. 305:33–40.
gram-negative bacteria, abstr. F1-311. Abstr. 47th Intersci. Conf. Antimi- 338. Petrella, S., N. Ziental-Gelus, C. Mayer, M. Renard, V. Jarlier, and W.
crob. Agents Chemother., Chicago, IL. Sougakoff. 2008. Genetic and structural insights into the dissemination
317. Page, M. G. P., C. Mueller, B. Hofer, and E. Desarbre. 2009. Bactericidal potential of the extremely broad-spectrum class A ␤-lactamase KPC-2
effect of the novel siderophore monobactam BAL30072, abstr. P1111. Ab- identified in an Escherichia coli strain and an Enterobacter cloacae strain
str. 19th Eur. Congr. Clin. Microbiol. Infect. Dis., Helsinki, Finland. isolated from the same patient in France. Antimicrob. Agents Che-
318. Page, M. I., P. S. Hinchliffe, J. M. Wood, L. P. Harding, and A. P. Laws. mother. 52:3725–3736.
2003. Novel mechanism of inhibiting ␤-lactamases by sulfonylation using 339. Pfaendler, H. R., F. Weisner, and K. Metzger. 1993. Synthesis and antibac-
␤-sultams. Bioorg. Med. Chem. Lett. 13:4489–4492. terial activity of (1⬘R, 5R, 6R)-2-tert-butyl-6-(1⬘-hydroxyethyl)oxapenem-3-
319. Paisley, J. W., and J. A. Washington, 2nd. 1978. Combined activitiy of carboxylic acid. Bioorg. Med. Chem. Lett. 3:2211–2218.
clavulanic acid and ticarcillin against ticarcillin-resistant, gram-negative ba- 340. Philippon, A., G. Arlet, and G. A. Jacoby. 2002. Plasmid-determined
cilli. Antimicrob. Agents Chemother. 14:224–227. AmpC-type ␤-lactamases. Antimicrob. Agents Chemother. 46:1–11.
319a.Papp-Wallace, K. M., C. R. Bethel, A. Distler, C. Kasuboski, M. Taracila, 341. Philippon, A., R. Labia, and G. Jacoby. 1989. Extended-spectrum ␤-lacta-
and R. A. Bonomo. 2009. Inhibitor resistance in the KPC-2 ␤-lactamase: a mases. Antimicrob. Agents Chemother. 33:1131–1136.
pre-eminent property of this class A ␤-lactamase. Antimicrob. Agents Che- 342. Phillips, O. A., D. P. Czajkowski, P. Spevak, M. P. Singh, C. Hanehara-
mother. [Epub ahead of print.] doi:10.1128/AAC.00693-09. Kunugita, A. Hyodo, R. G. Micetich, and S. N. Maiti. 1997. SYN-1012: a new
320. Paterson, D. L. 2006. Resistance in gram-negative bacteria: Enterobacteri- ␤-lactamase inhibitor of penem skeleton. J. Antibiot. (Tokyo) 50:350–356.
aceae. Am. J. Infect. Control 34:S20–S28. (Discussion, S64–S73.) 343. Pitout, J. D., and K. B. Laupland. 2008. Extended-spectrum ␤-lactamase-
321. Paterson, D. L., and R. A. Bonomo. 2005. Extended-spectrum ␤-lactamases: producing Enterobacteriaceae: an emerging public-health concern. Lancet
a clinical update. Clin. Microbiol. Rev. 18:657–686. Infect. Dis. 8:159–166.
322. Pattanaik, P., C. R. Bethel, A. M. Hujer, K. M. Hujer, A. M. Distler, M. 344. Plantan, I., L. Selic, T. Mesar, P. S. Anderluh, M. Oblak, A. Prezelj, L.
Taracila, V. E. Anderson, T. R. Fritsche, R. N. Jones, S. R. Pagadala, F. van Hesse, M. Andrejasic, M. Vilar, D. Turk, A. Kocijan, T. Prevec, G. Vilfan,
den Akker, J. D. Buynak, and R. A. Bonomo. 2009. Strategic design of an D. Kocjan, A. Copar, U. Urleb, and T. Solmajer. 2007. 4-Substituted
effective ␤-lactamase inhibitor: LN-1-255, a 6-alkylidene-2⬘-substituted trinems as broad spectrum ␤-lactamase inhibitors: structure-based design,
penicillin sulfone. J. Biol. Chem. 284:945–953. synthesis, and biological activity. J. Med. Chem. 50:4113–4121.
323. Patterson, J. E., T. C. Hardin, C. A. Kelly, R. C. Garcia, and J. H. Jor- 345. Poirel, L., and P. Nordmann. 2002. Acquired carbapenem-hydrolyzing ␤-lac-
gensen. 2000. Association of antibiotic utilization measures and control of tamases and their genetic support. Curr. Pharm. Biotechnol. 3:117–127.
VOL. 23, 2010 ␤-LACTAMASE INHIBITORS 199

346. Poirel, L., and P. Nordmann. 2006. Carbapenem resistance in Acineto- 372. Roy, C., C. Segura, M. Tirado, R. Reig, M. Hermida, D. Teruel, and A. Foz.
bacter baumannii: mechanisms and epidemiology. Clin. Microbiol. In- 1985. Frequency of plasmid-determined ␤-lactamases in 680 consecutively
fect. 12:826–836. isolated strains of Enterobacteriaceae. Eur. J. Clin. Microbiol. 4:146–147.
347. Poole, K. 2004. Efflux-mediated multiresistance in Gram-negative bacteria. 373. Saito, K., H. Yoneyama, and T. Nakae. 1999. nalB-type mutations causing
Clin. Microbiol. Infect. 10:12–26. the overexpression of the MexAB-OprM efflux pump are located in the
348. Potgieter, E., and L. J. Chalkley. 1995. Relatedness among penicillin-bind- mexR gene of the Pseudomonas aeruginosa chromosome. FEMS Microbiol.
ing protein 2b genes of Streptococcus mitis, Streptococcus oralis, and Strep- Lett. 179:67–72.
tococcus pneumoniae. Microb. Drug Resist. 1:35–42. 374. Sandanayaka, V. P., G. B. Feigelson, A. S. Prashad, Y. Yang, and P. J.
349. Powers, R. A., J. Blazquez, G. S. Weston, M. I. Morosini, F. Baquero, and Petersen. 2001. Allyl and propargyl substituted penam sulfones as versatile
B. K. Shoichet. 1999. The complexed structure and antimicrobial activity of intermediates toward the syntheses of new ␤-lactamase inhibitors. Bioorg.
a non-␤-lactam inhibitor of AmpC ␤-lactamase. Protein Sci. 8:2330–2337. Med. Chem. Lett. 11:997–1000.
350. Powers, R. A., E. Caselli, P. J. Focia, F. Prati, and B. K. Shoichet. 2001. 375. Sandanayaka, V. P., and Y. Yang. 2000. Dipolar cycloaddition of novel
Structures of ceftazidime and its transition-state analogue in complex with 6-(nitrileoxidomethyl) penam sulfone: an efficient route to a new class of
AmpC ␤-lactamase: implications for resistance mutations and inhibitor ␤-lactamase inhibitors. Org. Lett. 2:3087–3090.
design. Biochemistry 40:9207–9214. 376. Sanders, C. C., J. P. Iaconis, G. P. Bodey, and G. Samonis. 1988. Resistance
351. Powers, R. A., F. Morandi, and B. K. Shoichet. 2002. Structure-based to ticarcillin-potassium clavulanate among clinical isolates of the family
discovery of a novel, noncovalent inhibitor of AmpC ␤-lactamase. Structure Enterobacteriaceae: role of PSE-1 ␤-lactamase and high levels of TEM-1
10:1013–1023. and SHV-1 and problems with false susceptibility in disk diffusion tests.
352. Powers, R. A., and B. K. Shoichet. 2002. Structure-based approach for binding Antimicrob. Agents Chemother. 32:1365–1369.
site identification on AmpC ␤-lactamase. J. Med. Chem. 45:3222–3234. 377. Sauvage, E., F. Kerff, M. Terrak, J. A. Ayala, and P. Charlier. 2008. The
353. Pratt, R. F. 1989. Inhibition of a class C ␤-lactamase by a specific phos- penicillin-binding proteins: structure and role in peptidoglycan biosynthe-
phonate monoester. Science 246:917–919. sis. FEMS Microbiol. Rev. 32:234–258.
354. Prezelj, A., A. Plantan, G. Vilfan, P. Stefanic Anderluh, L. Selic, P. Iglicar, 378. Saves, I., O. Burlet-Schiltz, P. Swaren, F. Lefevre, J. M. Masson, J. C.
V. Car, S. Andrensek, I. Legen, P. Smrdel, M. Cerne, U. Urleb, I. Locatelli, Prome, and J. P. Samama. 1995. The asparagine to aspartic acid substitu-
A. Mrhar, and N. Kovacic. 2007. Stability of 10-ethylidene trinems, PK of tion at position 276 of TEM-35 and TEM-36 is involved in the ␤-lactamase
LK-157 and design of pro-drug esters, abstr. F1-0882. Abstr. 47th Intersci. resistance to clavulanic acid. J. Biol. Chem. 270:18240–18245.
Conf. Antimicrob. Agents Chemother., Chicago, IL. 379. Schneider, K. D., C. R. Bethel, A. M. Distler, A. M. Hujer, R. A. Bonomo,
355. Quale, J., D. Landman, G. Saurina, E. Atwood, V. DiTore, and K. Patel. 1996. and D. A. Leonard. 2009. Mutation of the active site carboxy-lysine (K70)
Manipulation of a hospital antimicrobial formulary to control an outbreak of of OXA-1 ␤-lactamase results in a deacylation-deficient enzyme. Biochem-
vancomycin-resistant enterococci. Clin. Infect. Dis. 23:1020–1025. istry 48:6136–6145.
356. Queenan, A. M., and K. Bush. 2007. Carbapenemases: the versatile ␤-lac- 379a.Schneider, K. D., M. E. Karpen, R. A. Bonomo, D. A. Leonard, and R. A.
tamases. Clin. Microbiol. Rev. 20:440–458. Powers. 2009. The 1.4 Å crystal structure of the class D ␤-lactamase OXA-1
357. Queenan, A. M., W. Shang, M. Kania, M. G. Page, and K. Bush. 2007. complexed with doripenem. Biochemistry 48:11840–11847.
Interactions of ceftobiprole with ␤-lactamases from molecular classes A to 380. Sham, D., C. Pillar, N. Brown, V. Alluru, C. Miossec, and P. Levasseur.
D. Antimicrob. Agents Chemother. 51:3089–3095. 2009. Activity of ceftazidime/NXL104 and select comparators against geo-
358. Rasmussen, B. A., and K. Bush. 1997. Carbapenem-hydrolyzing ␤-lactama- graphically diverse clinical isolates of Pseudomonas aeruginosa, abstr.
ses. Antimicrob. Agents Chemother. 41:223–232. E-194. Abstr. 49th Intersci. Conf. Antimicrob. Agents Chemother., San
359. Rasmussen, B. A., K. Bush, D. Keeney, Y. Yang, R. Hare, C. O’Gara, and Francisco, CA.
A. A. Medeiros. 1996. Characterization of IMI-1 ␤-lactamase, a class A 381. Shen, D., P. Winokur, and R. N. Jones. 2001. Characterization of extended
carbapenem-hydrolyzing enzyme from Enterobacter cloacae. Antimicrob. spectrum ␤-lactamase-producing Klebsiella pneumoniae from Beijing,
Agents Chemother. 40:2080–2086. China. Int. J. Antimicrob. Agents 18:185–188.
360. Reading, C., and M. Cole. 1977. Clavulanic acid: a ␤-lactamase-inhibiting 382. Shimamura, T., A. Ibuka, S. Fushinobu, T. Wakagi, M. Ishiguro, Y. Ishii,
␤-lactam from Streptomyces clavuligerus. Antimicrob. Agents Chemother. and H. Matsuzawa. 2002. Acyl-intermediate structures of the extended-
11:852–857. spectrum class A ␤-lactamase, Toho-1, in complex with cefotaxime, ceph-
361. Rebuck, J. A., K. M. Olsen, P. D. Fey, K. L. Bergman, and M. E. Rupp. alothin, and benzylpenicillin. J. Biol. Chem. 277:46601–46608.
2000. In vitro activities of parenteral ␤-lactam antimicrobials against 383. Shoichet, B. K., W. A. Baase, R. Kuroki, and B. W. Matthews. 1995. A
TEM-10-, TEM-26- and SHV-5-derived extended-spectrum ␤-lactama- relationship between protein stability and protein function. Proc. Natl.
ses expressed in an isogenic Escherichia coli host. J. Antimicrob. Che- Acad. Sci. U. S. A. 92:452–456.
mother. 46:461–464. 384. Siemann, S., D. P. Evanoff, L. Marrone, A. J. Clarke, T. Viswanatha, and
362. Retsema, J. A., A. R. English, A. Girard, J. E. Lynch, M. Anderson, L. G. I. Dmitrienko. 2002. N-Arylsulfonyl hydrazones as inhibitors of IMP-1
Brennan, C. Cimochowski, J. Faiella, W. Norcia, and P. Sawyer. 1986. metallo-␤-lactamase. Antimicrob. Agents Chemother. 46:2450–2457.
Sulbactam/ampicillin: in vitro spectrum, potency, and activity in models of 385. Sirot, D., C. Chanal, R. Bonnet, C. De Champs, and L. Bret. 2001. Inhib-
acute infection. Rev. Infect. Dis. 8(Suppl. 5):S528–S534. itor-resistant TEM-33 ␤-lactamase in a Shigella sonnei isolate. Antimicrob.
363. Reynolds, K. A., J. M. Thomson, K. D. Corbett, C. R. Bethel, J. M. Berger, Agents Chemother. 45:2179–2180.
J. F. Kirsch, R. A. Bonomo, and T. M. Handel. 2006. Structural and com- 386. Sirot, D., C. Recule, E. B. Chaibi, L. Bret, J. Croize, C. Chanal-Claris, R.
putational characterization of the SHV-1 ␤-lactamase-␤-lactamase inhibi- Labia, and J. Sirot. 1997. A complex mutant of TEM-1 ␤-lactamase with
tor protein interface. J. Biol. Chem. 281:26745–26753. mutations encountered in both IRT-4 and extended-spectrum TEM-15,
364. Rice, L. B. 2008. Federal funding for the study of antimicrobial resistance produced by an Escherichia coli clinical isolate. Antimicrob. Agents Che-
in nosocomial pathogens: no ESKAPE. J. Infect. Dis. 197:1079–1081. mother. 41:1322–1325.
365. Rice, L. B., E. C. Eckstein, J. DeVente, and D. M. Shlaes. 1996. Ceftazidime- 387. Slocombe, B., A. S. Beale, R. J. Boon, K. E. Griffin, P. J. Masters, R.
resistant Klebsiella pneumoniae isolates recovered at the Cleveland Depart- Sutherland, and A. R. White. 1984. Antibacterial activity in vitro and in vivo
ment of Veterans Affairs Medical Center. Clin. Infect. Dis. 23:118–124. of amoxicillin in the presence of clavulanic acid, p. 29–49. In Progress and
366. Richmond, M. 1981. The semi-synthetic thienamycin derivative MK0787 perspectives on ␤-lactamase inhibition: a review of Augmentin. Custom
and its properties with respect to a range of ␤-lactamases from clinically Communications: Postgraduate Medicine, New York, NY.
relevant bacterial species. J. Antimicrob. Chemother. 7:279–285. 388. Sougakoff, W., G. L’Hermite, L. Pernot, T. Naas, V. Guillet, P. Nordmann,
367. Richter, H. G., P. Angehrn, C. Hubschwerlen, M. Kania, M. G. Page, J. L. V. Jarlier, and J. Delettre. 2002. Structure of the imipenem-hydrolyzing
Specklin, and F. K. Winkler. 1996. Design, synthesis, and evaluation of 2 class A ␤-lactamase SME-1 from Serratia marcescens. Acta Crystallogr. D
␤-alkenyl penam sulfone acids as inhibitors of ␤-lactamases. J. Med. Chem. Biol. Crystallogr. 58:267–274.
39:3712–3722. 389. Srikumar, R., C. J. Paul, and K. Poole. 2000. Influence of mutations in the
368. Rizwi, I., A. K. Tan, A. L. Fink, and R. Virden. 1989. Clavulanate inactiva- mexR repressor gene on expression of the MexA-MexB-OprM multidrug
tion of Staphylococcus aureus ␤-lactamase. Biochem. J. 258:205–209. efflux system of Pseudomonas aeruginosa. J. Bacteriol. 182:1410–1414.
369. Robin, F., J. Delmas, C. Chanal, D. Sirot, J. Sirot, and R. Bonnet. 2005. 390. Stachyra, T., P. Levasseur, M. C. Pechereau, A. M. Girard, M. Claudon, C.
TEM-109 (CMT-5), a natural complex mutant of TEM-1 ␤-lactamase com- Miossec, and M. T. Black. 2009. In vitro activity of the ␤-lactamase inhibitor
bining the amino acid substitutions of TEM-6 and TEM-33 (IRT-5). Anti- NXL104 against KPC-2 carbapenemase and Enterobacteriaceae expressing
microb. Agents Chemother. 49:4443–4447. KPC carbapenemases. J. Antimicrob. Chemother. 64:326–329.
370. Rodriguez-Bano, J., M. D. Navarro, L. Romero, M. A. Muniain, M. de 391. Stachyra, T., M. C. Pechereau, J. M. Bruneau, C. Miossec, J. M. Frere, and
Cueto, M. J. Rios, J. R. Hernandez, and A. Pascual. 2006. Bacteremia due M. T. Black. 2009. The nature of inhibition of TEM-1 ␤-lactamase by the
to extended-spectrum ␤-lactamase-producing Escherichia coli in the non-␤-lactam inhibitor NXL104, abstr. C1-1374. Abstr. 49th Intersci. Conf.
CTX-M era: a new clinical challenge. Clin. Infect. Dis. 43:1407–1414. Antimicrob. Agents Chemother., San Francisco. CA.
371. Roth, T. A., G. Minasov, S. Morandi, F. Prati, and B. K. Shoichet. 2003. 392. Strynadka, N. C., H. Adachi, S. E. Jensen, K. Johns, A. Sielecki, C. Betzel, K.
Thermodynamic cycle analysis and inhibitor design against ␤-lactamase. Sutoh, and M. N. James. 1992. Molecular structure of the acyl-enzyme inter-
Biochemistry 42:14483–14491. mediate in ␤-lactam hydrolysis at 1.7 A resolution. Nature 359:700–705.
200 DRAWZ AND BONOMO CLIN. MICROBIOL. REV.

393. Strynadka, N. C., S. E. Jensen, K. Johns, H. Blanchard, M. Page, A. G. P. Rouen, S. H. Olson, M. L. Hammond, M. L. Greenlee, and Y. D. Gao.
Matagne, J. M. Frere, and M. N. James. 1994. Structural and kinetic 2001. Succinic acids as potent inhibitors of plasmid-borne IMP-1 metallo-
characterization of a ␤-lactamase-inhibitor protein. Nature 368:657–660. ␤-lactamase. J. Biol. Chem. 276:31913–31918.
394. Strynadka, N. C., R. Martin, S. E. Jensen, M. Gold, and J. B. Jones. 1996. 416. Totir, M. A., P. S. Padayatti, M. S. Helfand, M. P. Carey, R. A. Bonomo,
Structure-based design of a potent transition state analogue for TEM-1 P. R. Carey, and F. van den Akker. 2006. Effect of the inhibitor-resistant
␤-lactamase. Nat. Struct. Biol. 3:688–695. M69V substitution on the structures and populations of trans-enamine
395. Studemeister, A. E., and J. P. Quinn. 1988. Selective imipenem resistance ␤-lactamase intermediates. Biochemistry 45:11895–11904.
in Pseudomonas aeruginosa associated with diminished outer membrane 417. Tranier, S., A. T. Bouthors, L. Maveyraud, V. Guillet, W. Sougakoff, and
permeability. Antimicrob. Agents Chemother. 32:1267–1268. J. P. Samama. 2000. The high resolution crystal structure for class A
396. Sulton, D., D. Pagan-Rodriguez, X. Zhou, Y. Liu, A. M. Hujer, C. R. Bethel, ␤-lactamase PER-1 reveals the bases for its increase in breadth of activity.
M. S. Helfand, J. M. Thomson, V. E. Anderson, J. D. Buynak, L. M. Ng, and J. Biol. Chem. 275:28075–28082.
R. A. Bonomo. 2005. Clavulanic acid inactivation of SHV-1 and the inhib- 418. Trehan, I., F. Morandi, L. C. Blaszczak, and B. K. Shoichet. 2002. Using
itor-resistant S130G SHV-1 ␤-lactamase. Insights into the mechanism of steric hindrance to design new inhibitors of class C ␤-lactamases. Chem.
inhibition. J. Biol. Chem. 280:35528–35536. Biol. 9:971–980.
397. Sun, T., C. R. Bethel, R. A. Bonomo, and J. R. Knox. 2004. Inhibitor- 419. Tsang, W. Y., N. Ahmed, P. S. Hinchliffe, J. M. Wood, L. P. Harding, A. P.
resistant class A ␤-lactamases: consequences of the Ser130-to-Gly mutation Laws, and M. I. Page. 2005. Different transition-state structures for the
seen in Apo and tazobactam structures of the SHV-1 variant. Biochemistry reactions of ␤-lactams and analogous ␤-sultams with serine ␤-lactamases.
43:14111–14117. J. Am. Chem. Soc. 127:17556–17564.
398. Sun, T., M. Nukaga, K. Mayama, E. H. Braswell, and J. R. Knox. 2003. 420. Tumbarello, M., T. Spanu, M. Sanguinetti, R. Citton, E. Montuori, F.
Comparison of ␤-lactamases of classes A and D: 1.5-A crystallographic Leone, G. Fadda, and R. Cauda. 2006. Bloodstream infections caused by
structure of the class D OXA-1 oxacillinase. Protein Sci. 12:82–91. extended-spectrum-␤-lactamase-producing Klebsiella pneumoniae: risk fac-
399. Sun, T., M. Nukaga, K. Mayama, G. V. Crichlow, A. P. Kuzin, and J. R. tors, molecular epidemiology, and clinical outcome. Antimicrob. Agents
Knox. 2001. Crystallization and preliminary X-ray study of OXA-1, a class Chemother. 50:498–504.
D ␤-lactamase. Acta Crystallogr. D Biol. Crystallogr. 57:1912–1914. 421. Tzouvelekis, L. S., and R. A. Bonomo. 1999. SHV-type ␤-lactamases. Curr.
400. Swaren, P., D. Golemi, S. Cabantous, A. Bulychev, L. Maveyraud, S. Mo- Pharm. Des. 5:847–864.
bashery, and J. P. Samama. 1999. X-ray structure of the Asn276Asp variant 422. Tzouvelekis, L. S., M. Gazouli, E. E. Prinarakis, E. Tzelepi, and N. J.
of the Escherichia coli TEM-1 ␤-lactamase: direct observation of electro- Legakis. 1997. Comparative evaluation of the inhibitory activities of the
static modulation in resistance to inactivation by clavulanic acid. Biochem- novel penicillanic acid sulfone Ro 48-1220 against ␤-lactamases that belong
istry 38:9570–9576. to groups 1, 2b, and 2be. Antimicrob. Agents Chemother. 41:475–477.
401. Swaren, P., L. Maveyraud, V. Guillet, J. M. Masson, L. Mourey, and J. P. 423. Urban, C., E. Go, N. Mariano, B. J. Berger, I. Avraham, D. Rubin, and J. J.
Samama. 1995. Electrostatic analysis of TEM1 ␤-lactamase: effect of sub- Rahal. 1993. Effect of sulbactam on infections caused by imipenem-resistant
strate binding, steep potential gradients and consequences of site-directed Acinetobacter calcoaceticus biotype anitratus. J. Infect. Dis. 167:448–451.
mutations. Structure 3:603–613. 424. Urban, C., J. J. Rahal, and B. Luft. 1991. Effect of a ␤-lactamase inhibitor,
402. Swaren, P., L. Maveyraud, X. Raquet, S. Cabantous, C. Duez, J. D. Pede- tazobactam, on growth and penicillin-binding proteins of Borrelia burgdor-
lacq, S. Mariotte-Boyer, L. Mourey, R. Labia, M. H. Nicolas-Chanoine, P. feri. FEMS Microbiol. Lett. 66:113–116.
Nordmann, J. M. Frere, and J. P. Samama. 1998. X-ray analysis of the 425. Usher, K. C., L. C. Blaszczak, G. S. Weston, B. K. Shoichet, and S. J. Rem-
NMC-A ␤-lactamase at 1.64-A resolution, a class A carbapenemase with ington. 1998. Three-dimensional structure of AmpC ␤-lactamase from Esche-
broad substrate specificity. J. Biol. Chem. 273:26714–26721. richia coli bound to a transition-state analogue: possible implications for the
403. Sykes, R. B., D. P. Bonner, K. Bush, and N. H. Georgopapadakou. 1982. oxyanion hypothesis and for inhibitor design. Biochemistry 37:16082–16092.
Azthreonam (SQ 26,776), a synthetic monobactam specifically active against 426. Vakulenko, S. B., B. Geryk, L. P. Kotra, S. Mobashery, and S. A. Lerner.
aerobic gram-negative bacteria. Antimicrob. Agents Chemother. 21:85–92. 1998. Selection and characterization of ␤-lactam–␤-lactamase inactivator-
404. Sykes, R. B., D. P. Bonner, K. Bush, N. H. Georgopapadakou, and J. S. resistant mutants following PCR mutagenesis of the TEM-1 ␤-lactamase
Wells. 1981. Monobactams–monocyclic ␤-lactam antibiotics produced by gene. Antimicrob. Agents Chemother. 42:1542–1548.
bacteria. J. Antimicrob. Chemother. 8(Suppl. E):1–16. 427. Vedel, G., A. Belaaouaj, L. Gilly, R. Labia, A. Philippon, P. Nevot, and G.
405. Sykes, R. B., C. M. Cimarusti, D. P. Bonner, K. Bush, D. M. Floyd, N. H. Paul. 1992. Clinical isolates of Escherichia coli producing TRI ␤-lactamases:
Georgopapadakou, W. M. Koster, W. C. Liu, W. L. Parker, P. A. Principe, novel TEM-enzymes conferring resistance to ␤-lactamase inhibitors. J. An-
M. L. Rathnum, W. A. Slusarchyk, W. H. Trejo, and J. S. Wells. 1981. timicrob. Chemother. 30:449–462.
Monocyclic ␤-lactam antibiotics produced by bacteria. Nature 291:489–491. 428. Venkatesan, A. M., A. Agarwal, T. Abe, H. Ushirogochi, I. Yamamura,
406. Tabei, K., X. Feng, A. M. Venkatesan, T. Abe, U. Hideki, T. S. Mansour, M. Ado, T. Tsuyoshi, O. Dos Santos, Y. Gu, F. W. Sum, Z. Li, G.
and M. M. Siegel. 2004. Mechanism of inactivation of ␤-lactamases by novel Francisco, Y. I. Lin, P. J. Petersen, Y. Yang, T. Kumagai, W. J. Weiss,
6-methylidene penems elucidated using electrospray ionization mass spec- D. M. Shlaes, J. R. Knox, and T. S. Mansour. 2006. Structure-activity
trometry. J. Med. Chem. 47:3674–3688. relationship of 6-methylidene penems bearing 6,5 bicyclic heterocycles
407. Taibi, P., and S. Mobashery. 1995. Mechanism of turnover of imipenem by as broad-spectrum ␤-lactamase inhibitors: evidence for 1,4-thiazepine
the TEM ␤-lactamase revisited. J. Am. Chem. Soc. 117:7600–7605. intermediates with C7 R stereochemistry by computational methods.
408. Thomas, V. L., D. Golemi-Kotra, C. Kim, S. B. Vakulenko, S. Mobashery, J. Med. Chem. 49:4623–4637.
and B. K. Shoichet. 2005. Structural consequences of the inhibitor-resistant 429. Venkatesan, A. M., Y. Gu, O. Dos Santos, T. Abe, A. Agarwal, Y. Yang, P. J.
Ser130Gly substitution in TEM ␤-lactamase. Biochemistry 44:9330–9338. Petersen, W. J. Weiss, T. S. Mansour, M. Nukaga, A. M. Hujer, R. A.
409. Thomson, C. J., R. S. Miles, and S. G. Amyes. 1995. Susceptibility testing Bonomo, and J. R. Knox. 2004. Structure-activity relationship of 6-meth-
with clavulanic acid: fixed concentration versus fixed ratio. Antimicrob. ylidene penems bearing tricyclic heterocycles as broad-spectrum ␤-lacta-
Agents Chemother. 39:2591–2592. mase inhibitors: crystallographic structures show unexpected binding of
410. Thomson, J. M., A. M. Distler, and R. A. Bonomo. 2007. Overcoming 1,4-thiazepine intermediates. J. Med. Chem. 47:6556–6568.
resistance to ␤-lactamase inhibitors: comparing sulbactam to novel inhibi- 430. Venturelli, A., D. Tondi, L. Cancian, F. Morandi, G. Cannazza, B. Segatore,
tors against clavulanate resistant SHV enzymes with substitutions at Am- F. Prati, G. Amicosante, B. K. Shoichet, and M. P. Costi. 2007. Optimizing
bler position 244. Biochemistry 46:11361–11368. cell permeation of an antibiotic resistance inhibitor for improved efficacy.
411. Thomson, J. M., A. M. Distler, F. Prati, and R. A. Bonomo. 2006. Probing J. Med. Chem. 50:5644–5654.
active site chemistry in SHV ␤-lactamase variants at Ambler position 244. 431. Vilar, M., M. Galleni, T. Solmajer, B. Turk, J. M. Frere, and A. Matagne.
Understanding unique properties of inhibitor resistance. J. Biol. Chem. 2001. Kinetic study of two novel enantiomeric tricyclic ␤-lactams which
281:26734–26744. efficiently inactivate class C ␤-lactamases. Antimicrob. Agents Chemother.
412. Thomson, J. M., F. Prati, C. R. Bethel, and R. A. Bonomo. 2007. Use of 45:2215–2223.
novel boronic acid transition state inhibitors to probe substrate affinity in 432. Walsh, T. R., M. A. Toleman, L. Poirel, and P. Nordmann. 2005. Me-
SHV-type extended-spectrum ␤-lactamases. Antimicrob. Agents Che- tallo-␤-lactamases: the quiet before the storm? Clin. Microbiol. Rev.
mother. 51:1577–1579. 18:306–325.
413. Tondi, D., F. Morandi, R. Bonnet, M. P. Costi, and B. K. Shoichet. 2005. 433. Walter, M. W., R. M. Adlington, J. E. Baldwin, and C. J. Schofield. 1997.
Structure-based optimization of a non-␤-lactam lead results in inhibitors Synthesis of metallo-␤-lactamase inhibitors. Tetrahedron 53:7275–7290.
that do not up-regulate ␤-lactamase expression in cell culture. J. Am. 434. Walter, M. W., A. Felici, M. Galleni, R. P. Soto, R. M. Adlington, J. E.
Chem. Soc. 127:4632–4639. Baldwin, J. M. Frere, M. Gololobox, and C. J. Schofield. 1996. Trifluoro-
414. Toney, J. H., P. M. Fitzgerald, N. Grover-Sharma, S. H. Olson, W. J. May, methyl alcohol and ketone inhibitors of metallo-␤-lactamases. Bioorg. Med.
J. G. Sundelof, D. E. Vanderwall, K. A. Cleary, S. K. Grant, J. K. Wu, J. W. Chem. Lett. 6:2455–2458.
Kozarich, D. L. Pompliano, and G. G. Hammond. 1998. Antibiotic sensiti- 435. Walther-Rasmussen, J., and N. Hoiby. 2006. OXA-type carbapenemases. J.
zation using biphenyl tetrazoles as potent inhibitors of Bacteroides fragilis Antimicrob. Chemother. 57:373–383.
metallo-␤-lactamase. Chem. Biol. 5:185–196. 436. Wang, X., G. Minasov, J. Blazquez, E. Caselli, F. Prati, and B. K. Shoichet. 2003.
415. Toney, J. H., G. G. Hammond, P. M. Fitzgerald, N. Sharma, J. M. Balkovec, Recognition and resistance in TEM ␤-lactamase. Biochemistry 42:8434–8444.
VOL. 23, 2010 ␤-LACTAMASE INHIBITORS 201

437. Wang, X., G. Minasov, and B. K. Shoichet. 2002. Noncovalent interaction 450. Wyrembak, P. N., K. Babaoglu, R. B. Pelto, B. K. Shoichet, and R. F. Pratt.
energies in covalent complexes: TEM-1 ␤-lactamase and ␤-lactams. Pro- 2007. O-Aryloxycarbonyl hydroxamates: new ␤-lactamase inhibitors that
teins 47:86–96. cross-link the active site. J. Am. Chem. Soc. 129:9548–9549.
438. Wang, X., G. Minasov, and B. K. Shoichet. 2002. The structural bases of 451. Xu, D., D. Xie, and H. Guo. 2006. Catalytic mechanism of class B2 metallo-
antibiotic resistance in the clinically derived mutant ␤-lactamases TEM-30, ␤-lactamase. J. Biol. Chem. 281:8740–8747.
TEM-32, and TEM-34. J. Biol. Chem. 277:32149–32156. 452. Yamaguchi, Y., W. Jin, K. Matsunaga, S. Ikemizu, Y. Yamagata, J.
439. Wang, Z., W. Fast, and S. J. Benkovic. 1999. On the mechanism of the metallo- Wachino, N. Shibata, Y. Arakawa, and H. Kurosaki. 2007. Crystallographic
␤-lactamase from Bacteroides fragilis. Biochemistry 38:10013–10023. investigation of the inhibition mode of a VIM-2 metallo-␤-lactamase from
440. Wang, Z., W. Fast, A. M. Valentine, and S. J. Benkovic. 1999. Metallo-␤- Pseudomonas aeruginosa by a mercaptocarboxylate inhibitor. J. Med. Chem.
lactamase: structure and mechanism. Curr. Opin. Chem. Biol. 3:614–622.
50:6647–6653.
441. Weiss, W. J., P. J. Petersen, T. M. Murphy, L. Tardio, Y. Yang, P. A.
Bradford, A. M. Venkatesan, T. Abe, T. Isoda, A. Mihira, H. Ushirogochi, 453. Yang, Y., K. Janota, K. Tabei, N. Huang, M. M. Siegel, Y. I. Lin, B. A. Rasmussen,
T. Takasake, S. Projan, J. O’Connell, and T. S. Mansour. 2004. In vitro and and D. M. Shlaes. 2000. Mechanism of inhibition of the class A ␤-lactamases PC1
in vivo activities of novel 6-methylidene penems as ␤-lactamase inhibitors. and TEM-1 by tazobactam. Observation of reaction products by electrospray
Antimicrob. Agents Chemother. 48:4589–4596. ionization mass spectrometry. J. Biol. Chem. 275:26674–26682.
442. Weiss, W. J., M. E. Pulse, A. Endimiani, K. M. Hujer, A. M. Hujer, and 454. Yang, Y. J., P. J. Wu, and D. M. Livermore. 1990. Biochemical characterization
R. A. Bonomo. 2009. Efficacy of NXL104 in combination with ceftazidime in of a ␤-lactamase that hydrolyzes penems and carbapenems from two Serratia
murine infection models, abstr. B-1339. Abstr. 49th Intersci. Conf. Antimi- marcescens isolates. Antimicrob. Agents Chemother. 34:755–758.
crob. Agents Chemother., San Francisco, CA. 455. Yigit, H., A. M. Queenan, J. K. Rasheed, J. W. Biddle, A. Domenech-
443. Weston, G. S., J. Blazquez, F. Baquero, and B. K. Shoichet. 1998. Structure- Sanchez, S. Alberti, K. Bush, and F. C. Tenover. 2003. Carbapenem-
based enhancement of boronic acid-based inhibitors of AmpC ␤-lactamase. resistant strain of Klebsiella oxytoca harboring carbapenem-hydrolyzing
J. Med. Chem. 41:4577–4586. ␤-lactamase KPC-2. Antimicrob. Agents Chemother. 47:3881–3889.
444. Wexler, H. M., B. Harris, W. T. Carter, and S. M. Finegold. 1985. In vitro 456. Yuan, J., W. Huang, D. C. Chow, and T. Palzkill. 2009. Fine mapping of the
efficacy of sulbactam combined with ampicillin against anaerobic bacteria. sequence requirements for binding of ␤-lactamase inhibitory protein
Antimicrob. Agents Chemother. 27:876–878. (BLIP) to TEM-1 ␤-lactamase using a genetic screen for BLIP function. J.
445. Wilkinson, A. S., S. Ward, M. Kania, M. G. Page, and C. W. Wharton. Mol. Biol. 389:401–412.
1999. Multiple conformations of the acylenzyme formed in the hydroly- 457. Zafaralla, G., E. K. Manavathu, S. A. Lerner, and S. Mobashery. 1992.
sis of methicillin by Citrobacter freundii ␤-lactamase: a time-resolved Elucidation of the role of Arginine-244 in the turnover processes of class A
FTIR spectroscopic study. Biochemistry 38:3851–3856.
␤-lactamases. Biochemistry 31:3847–3852.
446. Williams, H., A. King, K. Shannon, and I. Phillips. 1988. Amoxycillin/
clavulanate resistant Escherichia coli. Lancet i:304–305. 458. Zafaralla, G., and S. Mobashery. 1992. Facilitation of the ⌬2 to ⌬1 pyrroline
447. Williams, J. D. 1997. ␤-Lactamase inhibition and in vitro activity of sulbac- tautomerization of carbapenem antibiotics by the highly conserved argi-
tam and sulbactam/cefoperazone. Clin. Infect. Dis. 24:494–497. nine-244 of class A ␤-lactamases during the course of turnover. J. Chem.
448. Wu, P. J., K. Shannon, and I. Phillips. 1994. Effect of hyperproduction of Soc. 114:1505–1506.
TEM-1 ␤-lactamase on in vitro susceptibility of Escherichia coli to ␤-lactam 459. Zapun, A., C. Contreras-Martel, and T. Vernet. 2008. Penicillin-binding
antibiotics. Antimicrob. Agents Chemother. 38:494–498. proteins and ␤-lactam resistance. FEMS Microbiol. Rev. 32:361–385.
449. Wu, P. J., K. Shannon, and I. Phillips. 1995. Mechanisms of hyperproduc- 460. Zhang, Z., and T. Palzkill. 2004. Dissecting the protein-protein interface
tion of TEM-1 ␤-lactamase by clinical isolates of Escherichia coli. J. Anti- between ␤-lactamase inhibitory protein and class A ␤-lactamases. J. Biol.
microb. Chemother. 36:927–939. Chem. 279:42860–42866.

Sarah M. Drawz received her bachelor’s de- Robert A. Bonomo received his M.D. from
gree from Amherst College magna cum the Case Western Reserve University
laude. She is currently an M.D./Ph.D. stu- School of Medicine. He trained in Inter-
dent at Case Western Reserve University nal Medicine and Infectious Diseases at
School of Medicine in the Department of University Hospitals of Cleveland and re-
Pathology. Her Ph.D. dissertation work fo- ceived a Certificate of Added Qualifica-
cuses on the inhibition of ␤-lactamase en- tion in Geriatrics. Dr. Bonomo served as
zymes. She plans to return to her medical section chief of the infectious disease di-
studies in the coming year. vision at the Cleveland Veterans Affairs
Medical Center before becoming Director
of the Veterans Integrated Service Net-
work 10 Geriatric Research, Education, and Clinical Center
(GRECC). He is a Professor of Medicine at Case Western Reserve
University School of Medicine and also holds appointments in the
Departments of Pharmacology and Molecular Biology and Micro-
biology. Dr. Bonomo’s research focuses on microbial resistance to
antibiotics, especially ␤-lactams.

Potrebbero piacerti anche