You are on page 1of 14

Review

Multitarget ligands in
antibacterial research: progress
and opportunities
1. Introduction
Stephen P East† & Lynn L Silver
2. DNA replication †
Evotec (UK) Ltd., Oxfordshire, UK
3. Cell wall inhibitors
4. Miscellaneous multitarget Introduction: Resistance to current antibacterial therapies is an inevitability
ligands that represents a significant global health concern. Bacteria have the capacity
Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Sussex Library on 01/20/13

5. Future opportunities to render all current drug treatments ineffective, which places a demand on
the drug discovery community to constantly develop new antibacterial
6. Expert opinion
agents. Compounds that inhibit multiple biological targets, often referred
to as multitarget ligands, are an inviting prospect in antibacterial research
because, although they will not solve the issue of resistance, they might
help to delay the onset.
Areas covered: This review covers some of the recent progress in identifying
new ligands that deliberately interact with more than one essential biological
target in bacteria. The two principal areas covered are inhibitors of DNA
replication and cell wall biosynthesis.
Expert opinion: Antibacterial programs for the design of multitarget ligands
present an important opportunity for production of antibacterial agents.
For personal use only.

Their longevity, due to slow development of resistance, is comparable to


that seen with other successful agents -- but is much improved over single-
targeted agents for which resistance can appear in vitro overnight. The pre-
clinical development of these agents will have to overcome the standard
problems of antibacterial discovery. Such problems include optimization of
characteristics favoring cell entry and particularly the demonstration of selec-
tivity of inhibition of the desired multiple targets without inhibition of other
bacterial or any mammalian functions.

Keywords: antibacterials, bacterial topoisomerases, cell wall biosynthesis, multitarget inhibitors,


resistance, structure-based drug design

Expert Opin. Drug Discov. (2013) 8(2):143-156

1. Introduction

Research into new antibacterial drugs is of paramount importance because of the


innate ability of bacteria to develop resistance to all existing therapies [1]. Walsh
neatly summarized the scale of the problem in 2000 when he commented
‘development of resistance is not a matter of if but only a matter of when’ and in
doing so he highlighted the continuing need for the identification of new anti-
bacterial agents [2]. One aspect of antibacterial research that is of current interest
is concerned with multitargeted therapies or polypharmacology [3]. This strategy is
an inviting prospect in the search for new antibacterial agents because, although it
would not solve the issue of resistance long term, it is expected to delay the onset
of resistance as the bacteria would have to develop viable mutations on more than
one biological target before the drug or combination of drugs are rendered ineffec-
tive. Spontaneous resistance mutations on single targets generally emerge at frequen-
cies between 10-6 and 10-9 [1] and so a multitarget approach might mean that
resistance develops at significantly lower frequencies than this.

10.1517/17460441.2013.743991 © 2013 Informa UK, Ltd. ISSN 1746-0441, e-ISSN 1746-045X 143
All rights reserved: reproduction in whole or in part not permitted
S. P. East & L. L. Silver

inhibit multiple biological targets has largely been determined


Article highlights. retrospectively. Since the elucidation of the bacterial genome,
. Established strategies and new opportunities for advances in screening and improvements in protein structure
multitarget ligands as antibacterials with the potential determination, the rational design of multitarget inhibitors
to slow the onset of resistance in bacteria. of antibacterial enzymes have been possible. Screening techni-
. Impact of X-ray crystallography for lead identification
ques have identified new natural products (historically a fertile
through computationally led virtual screens but also
for optimization of novel multitarget ligands using hunting ground for antibiotics) that are multitarget ligands.
structure-based drug design techniques. The emergence of structural information through X-ray
. New preclinical antibacterials targeting the enzymes crystallography and NMR has facilitated structure-based
gyrase and topoisomerase IV enzymes that are involved design opportunities for high-throughput screening hits as
in DNA replication.
.
well as providing the necessary tools to conduct computation-
Recent early stage advances in the search for novel
ally led virtual screens. This review will discuss some of the
Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Sussex Library on 01/20/13

multitarget inhibitors of enzymes in the cell wall


biosynthetic pathway. ongoing research programs aimed at identifying multitarget
. Optimization of compound physicochemical properties ligands of essential bacterial enzymes. The focus is on chemi-
as well as potency on multiple enzyme targets are cal matter reported in recent publications and presentations
necessary to achieve good antibacterial activity.
(2010 -- 2012).
This box summarizes key points contained in the article.
2. DNA replication

Multitargeted chemotherapy [4,5] can be broadly divided Enzymes that function as part of the bacterial DNA synthesis
into two categories: i) combination therapies, that is, two or machinery play an essential role in bacterial cell survival [13].
more compounds that when taken together, either as a cock- Two enzymes that contribute to DNA replication are the
tail or a multicomponent drug, work synergistically but typi- bacterial type II topoisomerases: DNA gyrase and topoisom-
cally elicit their pharmacological response by acting on erase (topo) IV. These enzymes are responsible for controlling
For personal use only.

independent biological targets [6] and ii) single compound DNA topology at various stages of DNA replication. Both
therapies, also termed multiple ligands [4,7] or multitarget enzymes form heterotetrameric complexes, which in the case
ligands [8], that is, one compound that modulates the pharma- of DNA gyrase is an A2B2 complex and for topo IV is a
cological response of two or more relevant biological targets. complex consisting of two ParC and two ParE subunits. As
Multitarget ligands can be a product of two or more discrete a consequence of their similar structural homology, dual
molecules which, when separate, interact independently with inhibition of gyrase and topo IV has been enabled, albeit
quite different biological targets, but they have been joined serendipitously [14], and members of the quinolone (including
(via a linker) or fused together to form a single compound. fluoroquinolones) class of antibiotics have validated this
Indeed this approach has been used successfully in the search approach clinically for antibacterial infections [12]. The quino-
for new antibacterial agents and reviewed recently [9]. Alterna- lones inhibit the related GyrA and ParC subunits. The GyrB
tively, multitarget ligands can contain specific structural fea- and ParE subunits bind and process ATP and so the similarity
tures that are important in the formation of key binding in the ATP-binding site of these proteins provides an addi-
interactions with not just one but multiple biological targets tional opportunity for inhibition of two targets. Indeed dual
of interest. Morphy and Rankovic [4] have referred to these inhibitors of GyrB/ParE have been identified but currently
types of ligands as being ‘merged’. In the scope of antibacterial there are no approved GyrB/ParE drugs.
research such multitarget ligands have been identified, most
typically in cases where the biological targets are structurally 2.1 GyrA/ParC inhibitors
similar at the ligand-binding site and/or they have closely Despite a plethora of quinolone antibacterials in the market-
related substrates and it is these ligands that are the focus of place, emerging resistance and issues with toxicity have meant
this review. A possible advantage of these merged ligands that the search for alternative GyrA/ParC inhibitors continues
over the joined or fused ligands is that the resulting drugs unabated. Several compounds are progressing in the latter
might be expected to have a lower molecular weight and con- stages of clinical development and these have been covered
sequently might occupy more attractive chemical space in elsewhere [15] but there are others that are at a preclinical stage
terms of their physicochemical and DMPK properties [10]. or that have recently entered Phase I, and some of these
So the questions are: How have we identified multitarget developments will be covered here.
ligands in antibacterial research programs in the past and Pucci et al. [16] have detailed the in vitro and in vivo profile
how might we do it better in the future? For the former of Achillion’s advanced isothiazoloquinolone ACH-702
question, the leading classes of multitarget antibacterials are (Figure 1). This compound is active in Staphylococcus aureus
the b-lactams that inhibit penicillin-binding proteins (PBPs) topo IV decatenation assays (IC50 120 nM) and DNA gyrase
(e.g., carbapenems [11]) and the quinolones that inhibit supercoiling (IC50 680 nM) assays and shows superior
DNA topoisomerases [12], but the discovery that these drugs dual-targeting inhibition compared with benchmark

144 Expert Opin. Drug Discov. (2013) 8(2)


Multitarget ligands in antibacterial research: progress and opportunities

O O O O O O
F F
N OH N OH
NH
N N S N N
H2 N
H2N OCH3
H2N

ACH-702 ABT-719 GC-061

OCH3
O
N OCH3
OH H
S N

N
N O N
S S F N
OH NC
Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Sussex Library on 01/20/13

Viquidacin GSK299423

Figure 1. Quinolone-related dual GyrA/ParC inhibitors and other prototypical topoisomerase inhibitors.

fluoroquinolones moxifloxacin, gemifloxacin and ciprofloxa- fluoroquinolone-resistant organisms. For example, a represen-
cin. It also retains some activity in vitro against single- and tative compound in this series, GC-061 (Figure 1), is active in
double-mutated gyrases that show resistance to the same set S. aureus (MIC range < 0.015 -- 0.5 µg/ml), which includes
of control compounds. Broad-spectrum antibacterial activity strains that are ciprofloxacin- and levofloxacin-resistant (i.e.,
against clinical isolates was shown with ACH-702 having a MIC ‡ 16 µg/ml). In Gram-negative organisms, GC-061
better profile than levofloxacin on Gram-positives including has excellent activity against Haemophilus influenzae
activity on fluoroquinolone nonsusceptible organisms. On (MICs < 0.015 µg/ml) but also shows some activity against
For personal use only.

Gram-negative organisms, ACH-702 has a similar profile to strains of Escherichia coli (median MIC 16 µg/ml), Pseudomo-
ciprofloxacin. Time-kill analysis revealed that ACH-702 is nas aeruginosa (median MIC 8 µg/ml) and other organisms.
bactericidal and resistant mutations were observed at frequen- Confirmation of dual-targeting with GC-061 was demon-
cies of < 10-10 at 4  MIC in strains of S. aureus and this strated in gyrase and topo IV in vitro assays with
represents a ~ 2 log units improvement over ciprofloxacin. IC50s £ 1.42 and £ 2.33 µM reported. Spontaneous resis-
Positive data on ACH-702 in two Gram-positive models of tance frequencies for compounds in this series are reported
infection have been reported. A dose-dependent effect in a to be low (i.e., ~ 10-11 in E. coli and S. aureus at 4  MIC)
methicillin-resistance S. aureus (MRSA) mouse thigh infec- which is suggestive but not conclusive of dual inhibition in
tion model was observed, with 5 mg/kg leading to stasis in a cellular context. In vivo efficacy of GC-061 was achieved in
infection after 24 h, and a 2.5-log reduction in colony a mouse thigh MRSA infection model when dosed orally at
forming units (CFUs) was observed after the same timeframe 30 mg/kg with ~ 5-log reduction in bacterial tissue burden
following a dose of 20 mg/kg. On the Achillion website [17], being observed after 24 h. The same compound showed
ACH-702 is also reported to be active on DNA primase, a ~ 3-log units reduction in CFUs over the same time period
which is another enzyme involved in bacterial DNA replica- in a mouse E. coli thigh infection model. These data are at
tion. Although no data demonstrating activity of ACH-702 least as good as ciprofloxacin (20 mg/kg i.v. dose) in the
on this target are available, DNA primase and the topoisomer- same models. GC-061 was selective over human topoisomer-
ases have been shown to share a homologous catalytic site, the ases (IC50 Topo II > 35 µM; IC50 Topo I > 100 µM), and
TOPRIM domain, which may underlie activity against all there are no apparent in vitro toxicity issues (e.g., hERG
three proteins [18]. Detailed structure--activity relationship IC50 60 µM, inactive at the highest concentration tested in a
studies that led to the identification of ACH-702 have been HepG2 cell line, a micronucleus genotoxicity assay and
described by Kim et al. [19]. cytochrome p450 assays).
Evolva are progressing broad-spectrum bactericidal In addition to the quinolone and related classes of antibac-
2-pyridones (termed the EV-035 series) [20,21] that are isosteric terial agents, other novel bacterial topoisomerase II inhibitors
to the traditional quinolones and related in structure to that target the GyrA/ParC subunits have been described in the
ABT-719 (Figure 1), a compound that reached Phase II literature. A prototype compound was viquidacin (NXL101,
clinical trials [22]. In silico modeling has suggested that the Figure 1) [23], and this antibacterial agent advanced into
substitution pattern on the EV-035 series is crucial for Phase I studies before issues associated with QT prolongation
compounds to adopt an overlapping but different binding halted its progression in the clinic. Several research programs,
mode to the quinolones. This subtle change in the ligands including GSK [24,25], AstraZeneca [26-28] and Achillion [29],
interaction with DNA gyrase/topo IV is probably responsible have all reported on chemotypes related to viquidacin with a
for the 2-pyridones showing good antibacterial activity against focus on addressing the cardiovascular toxicity side effects by

Expert Opin. Drug Discov. (2013) 8(2) 145


S. P. East & L. L. Silver

OCH3
tuning logD and pKa and monitoring hERG inhibition

N
in vitro. These chemotypes are of significant interest because

S. aureus (n = 7) 0.5 -- 2 µg/ml


OH X-ray crystal studies on GSK299423 (Figure 1) [30,31] using
S. aureus gyrase revealed that it binds to a site on GyrA and

Topo IV (S. aureus) 69 µM


Gyrase (S. aureus) 0.7 µM
O

OH
bridges to the bound DNA substrate such that it intercalates
between base-pairs, thus preventing DNA cleavage. This
represents an alternative binding mode to the quinolones [32]
N

and supports the potential usefulness in terms of their activity


on fluoroquinolone-resistant organisms. Some representative
Achillion
S

examples and data on recent inhibitors are shown in Table 1.


Although IC50 data on both DNA gyrase and topoisomerase
Se

[29]
are not provided in all of the publications, the implication is
Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Sussex Library on 01/20/13

that these compounds are dual-targeting agents. For example,


Reck et al. [28] commented that there was a good correlation
between IC50s on E. coli topo IV activity and S. aureus
gyrase. Of the compounds shown in Table 1, the AstraZeneca
model at 40 mg/kg; 1 log unit drop in CFUs

compound is perhaps the furthest advanced as it was


Efficacy in mouse S. aureus thigh infection

reported to have moved into clinical trials. It should be noted


that IC50 data on both targets is not sufficient to prove
S. aureus (n = 5) 0.06 -- 0.125 µg/ml
CN

dual-targeting at the whole cell level and resistance studies


S. pneumoniae (n = 1) 0.13 µg/ml

P. aeruginosa (n = 1) > 8 µg/ml

(or other means) are necessary. Resistance studies supporting


N

dual-targeting, however, have been carried out in many of


O

Topo IV (E. coli) 0.048 µM

the referenced cases.


E. coli (n = 1) 4 µg/ml
N
F
For personal use only.

2.2 GyrB/ParE inhibitors


H
N

Natural product inhibitors of the ATPase activity of DNA


gyrase and topo IV, such as the aminocoumarin compound
N

novobiocin, have been well characterized biochemically, struc-


turally and pharmacologically; however, their clinical utility
O

[28]
O

AZ

stalled, owing to resistance and toxicity [33]. The cyclothiali-


dine class of natural product antibacterials have also been
widely studied as a result of their inhibition of bacterial
type II topoisomerases and synthetic analogs demonstrating
in vivo efficacy based on this chemical series have been
reported recently [34]. For example, compound 1 (Figure 2)
OCH3

was reported to have an ED50 of 3 mg/kg in a mouse S. aureus


N

Efficacy in rat S. pneumoniae lung


S. pneumoniae (n = 1) 0.06 µg/ml
Table 1. Recent bacterial topoisomerase II inhibitors.

septicemia model.
N

Gyrase (H. influenzae) 0.17 µM

H. influenzae (n = 1) 0.5 µg/ml

Phillips et al. [35] reported a new naturally occurring ATPase


OH

infection model at 50 mg/kg


S. aureus (n = 1) 0.03 µg/ml
F

inhibitor, kibdelomycin (Figure 2), which was isolated


[DNA replication assay]

from Kibdelosporangium sp. and shown to have activity on


N

Gram-positive pathogens (e.g., S. aureus MIC 0.5 -- 64 µg/ml;


Streptococcus pneumoniae 1 µg/ml) as well as H. influenzae
H
N

(MIC 2 µg/ml). The discovery of kibdelomycin followed an


N

examination of S. aureus fitness test profiles and comparisons


with known antibiotic profiles, where it exhibited a similar
GSK
O

[25]

mechanism of action to novobiocin. Macromolecular biosyn-


thesis assays confirmed that kibdelomycin inhibited DNA
replication by blocking the incorporation of 14C-thymidine;
however, at the MIC, synthesis of other macromolecules was
affected as well. Subsequently, dual inhibition of DNA gyrase
n: Number of isolates.

and topo IV was confirmed in catalytic ATPase in vitro


Organization

assays in E. coli (Gyr IC50 11 nM; Topo IV IC50 900 nM),


Comment
Structure

as well as gyrase supercoiling (IC50 9 nM) and topoisomerase


decatenation (IC50 500 nM) assays in S. aureus. Resistance
MIC
IC50

Ref.

frequencies were shown to be < 5  10-10 CFU/ml and little

146 Expert Opin. Drug Discov. (2013) 8(2)


Multitarget ligands in antibacterial research: progress and opportunities

O N Interest in non-natural product inhibitors of the ATPase


N
activity of DNA gyrase and topo IV has been stimulated by
OH S
the availability of structural information on these targets.
HN O
Following pioneering work by Boehm et al. [39] on
O
HN
fragment-based screening, Charifson et al. were responsible
H3CO
Br O
for demonstrating the utility of synthetic GyrB/ParE inhibi-
O tors in vivo with a series of benzimidazole ethyl ureas of which
1 compound 3 in Figure 3 has been characterized extensively [40].
O
These inhibitors have been shown to be antibacterial solely
Cl
O OCH3 via their inhibition of GyrB and topo IV ParE [41]. Based on
Cl published crystallographic data [42], the crucial pharmaco-
O
O N OH NH phoric features for binding of this scaffold to ParE are the
Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Sussex Library on 01/20/13

H
H2N O N donor NH groups on the ethyl ureas that interact with an
O O O O
Asp residue in the ATP pocket together with the N-1 of the
HO H H H OH
O benzimidazole that acts as an acceptor donor to a conserved
water molecule that also hydrogen bonds to the same Asp
Kibdelomycin residue. In a very recent patent disclosure [43], Vertex have
reported the latest generation dual-targeting benzimidazoles
OH with compound 4a (Figure 3) showing good enzymatic inhibi-
HO O
tion (S. aureus Gyr Ki 9 nM; S. aureus Topo IV Ki 12 nM)
OH and antibacterial activity particularly against Gram-positive
O
F strains (S. aureus MIC90 0.03 µg/ml (n = 67); S. pneumoniae
F OH O O
MIC90 0.015 µg/ml (n = 67); Enterococcus faecalis MIC90
O 0.06 µg/ml (n = 34)). The compound 4a is shown to be
For personal use only.

O
OH
O active in a mouse S. aureus kidney infection model and a
O OH
S. aureus neutropenic rat thigh model. The patent also descri-
2 bes the prodrug 4b which was presumably investigated due to
the low solubility of 4a and indeed 4b might be the preferred
delivery method for active ingredient 4a.
Figure 2. Naturally occurring/derived dual DNA gyrase and Recognizing the key inhibitor design features of the benz-
topo IV inhibitors. imidazole ethyl ureas, several other groups have published
research in which the benzimidazole scaffold has been inter-
or no cross-resistance with aminocoumarin-resistant bacterial changed with a number of other bicyclic and biaryl systems
strains were observed. The lack of kibdelomycin-resistant that have the potential to retain the critical H-bond acceptor
mutants found could be due to inhibition of other essential as well as the ethyl urea moiety [44]. The benzothiazoles pre-
functions above the MIC. sented by Biota are the most recent refinement of the ethyl
Hossion et al. described quercetin diacylglycosides as dual urea series [45]. Fine tuning of the substituents in the 5- and
DNA gyrase/topo IV inhibitors [36,37]. One of the most inter- 7-position of the benzothiazole scaffold provided compounds
esting analogs, compound 2 (Figure 2), was reported to be active such as example 5 in Figure 3 that has excellent activity on
in an E. coli gyrase supercoiling assay (IC50 0.78 µM) and Gram-positive pathogens (S. aureus MIC 0.06 µg/ml;
E. coli topo IV (IC50 9.2 µM) and S. aureus topo IV Streptococcus pyogenes MIC 0.06 µg/ml; E. faecalis MIC
(IC50 0.22 µM) decantenation assays. The compound is active 0.02 µg/ml) and a good pharmacokinetic profile with
on Gram-positive organisms (MICs 0.13 -- 0.5 µg/ml on > 65% oral bioavailability being achieved in rodents. Some
S. pneumoniae, methicillin-resistant S. aureus and vancomycin- Gram-negative activity has been observed (H. influenzae
resistant enterococci) but no activity on Gram-negative species MIC 0.5 -- 2 µg/ml); Moraxella catarrhalis (MIC
was observed (MICs > 128 µg/ml on P. aeruginosa and E. coli). < 0.125 µg/ml) suggesting that the compound class could be
Molecular docking studies postulated that the compounds suitable for the treatment of respiratory tract infections. The
bind to the ATP-binding site on GyrB, although no data in structures of the most advanced compounds have not been
ATPase assays were reported to confirm that this was the revealed but spontaneous resistance frequencies for lead com-
site of action of this inhibitor class. It should be noted that pounds in this class were reported as being < 10-9 in S. aureus
quercetin is a flavonoid that has been shown to inhibit a great and S. pneumoniae, which is consistent with a dual-
number of enzymes, eukaryotic, viral as well as bacterial [38] targeting mechanism of action. In vivo efficacy via oral
and it is unlikely that gyrase/topoisomerase inhibition is the administration with a compound from this series was demon-
sole or even major mechanism of action of quercetin in strated in models of S. aureus including a neutropenic thigh
Gram-positives. model. In a S. pneumoniae mouse model at a 30 mg/kg dose

Expert Opin. Drug Discov. (2013) 8(2) 147


S. P. East & L. L. Silver

RO
N N

N H N N H
N H N H
N N F N N
H O H
F O
N O

3 Vertex 4a R = H
4b R = P(=O)(ONa)2
HO2C

N N OCH3 N H
N N H
N N N
H
Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Sussex Library on 01/20/13

N H
S N N O
O N N
O
N HN
CF3
O
5 Biota 6 AstraZeneca

O–
N+
S
N H
N N
N

N
Cl
H
For personal use only.

H2N
7 Trius

Figure 3. Dual GyrB/ParE inhibitors.

(intravenous), 100% survival was noted in 9 days after inocu- Gram-negative organisms was provided. Indeed Manchester
lation. In vitro toxicology assessment was good with no appar- et al. have reported in a separate publication [47] that the
ent issues identified and compounds were reported to be AstraZeneca experience of optimizing Gram-negative
progressing through in vivo safety experiments. antibacterial activity in new compounds classes is difficult.
Scaffolds that do not contain an ethyl urea group but have Efficient efflux systems together with more complex
been identified using structural information have also been permeability barriers in Gram-negatives compared with
described recently. Manchester et al. [46] assembled a library Gram-positives might be responsible for the apparent lack
of fragments previously observed in ATP-competitive kinase of data for the azaindoles.
inhibitor programs at AstraZeneca. From a virtual screening Trius have utilized a combination of structure-based virtual
exercise using a S. pneumoniae ParE structure, they identified screens and literature-led identification of new GyrB/ParE
an azaindole scaffold as satisfying the key molecular interac- dual inhibitor scaffolds. A key design feature in the Trius
tions with the Asp and conserved water residue in ParE. This program was to rationally design inhibitors of Gram-negative
scaffold was further elaborated to achieve low nanomolar pathogens as well as Gram-positive organisms. This was
potency on both S. pneumoniae ParE and S. aureus GyrB achieved by building in some degree of flexibility to the
with example 6 in Figure 3 being one of the most potent ana- compounds to compensate for the differences in the Gram-
logs described (ParE IC50 3 nM; GyrB IC50 2 nM; S. aureus positive and Gram-negative protein structures, as well as opti-
MIC 0.1 -- 0.2 µg/ml; S. pneumoniae MIC < 0.02 µg/ml). mizing the compounds for Gram-negative entry and lack of
Antibacterial activity did not correlate at all with enzymatic efflux. One of the advanced Trius compounds, 7 (Figure 3), is
activity but some relationship between logD and antibacte- from a series of pyrrolopyrimidines. This compound has
rial activity on Gram-positive organisms was observed. sub-nanomolar binding affinity on E. faecalis (Ki < 0.3 nM)
The hypothesis presented was that compounds with and Francisella tularensis GyrB (Ki < 0.3 nM) and single
higher logD show improved permeability properties (e.g., digit nanomolar activity on ParE (E. faecalis Ki 8.6 nM;
compound 6, logD = 2.91) to traverse the bacterial cell F. tularensis Ki 1.7 nM; E. coli Ki 2.5 nM) [48]. The in vitro
membrane. No activity for the azaindole compounds on antibacterial activity for compound 6 is good against

148 Expert Opin. Drug Discov. (2013) 8(2)


Multitarget ligands in antibacterial research: progress and opportunities

Gram-positive (S. aureus MIC 0.03 µg/ml; S. pneumoniae

MurC (E. coli) 26%@500 µM


MurD (E. coli) 50%@500 µM
NH
MIC 0.25 µg/ml) and Gram-negative pathogens (H. influenzae

O
MIC 2 µg/ml; E. coli 2 µg/ml; P. aeruginosa 4 µg/ml), and it

Cl
also has good coverage of biodefense pathogens. The com-
S

N
pound was reported to show activity in a S. aureus septicemia

NH
N

N mouse model with a 100% survival at 12.5 mg/kg dose. Interes-


tingly, by performing macromolecular synthesis experiments,
H
N

Trius observed that structurally related compounds in their

[60]
11

nd
pyrrolopyrimidine series did not necessarily show all the same
specific effect on DNA synthesis in bacterial cells. This
experiment illustrates (as noted in several places above) that
antibacterial activity is not always a consequence of targeted
MurC (E. coli) 52%@100 µM
Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Sussex Library on 01/20/13

inhibition in the cell and nonspecific effects might be at play.


MurE (S. aureus) 6 µM
NH

MurD (E. coli) 2 µM

Cell wall inhibitors


S

3.
MurF (E. coli) 2 µM
O

3.1 Mur ligases


No activity

Peptidoglycan is a crucial ingredient in bacterial cell walls of


OH

both Gram-positive and Gram-negative organisms. It


provides shape, integrity and the necessary protective barrier
[56]
HO

HO

10

to restrict permeability into and out of the bacterial cell.


Numerous antibacterial drugs, both naturally occurring and
synthetically derived, have been found to inhibit specific steps
NH

of peptidoglycan biosynthesis. For example, b-lactam antibi-


O
For personal use only.

otics inhibit transpeptidase enzymes that are responsible for


S

late-stage cross-linking of peptidoglycan. This class of drugs


thus compromises the rigidity of the cell wall leading to cell
lysis. Indeed, it was the identification of penicillin that
S. aureus (n = 2) 8 µg/ml
MurD (S. aureus) 6.4 µM

MurE (S. aureus) 17 µM

provided the basis for the current level of understanding of


MurD (E. coli) 8.2 µM

MurE (E. coli) 180 M


H
N

peptidoglycan assembly, a process that involves more than


E. faecalis 64 µg/ml

10 sequential enzymatic steps that are unique to bacteria.


Because peptidoglycan is essential for bacterial cell survival,
NH

its synthesis presents numerous opportunities for target-


CO2H

based inhibition and hence therapeutic solutions to bacterial


HO2C

infections [49].
[55]
9

The first six steps of peptidoglycan biosynthesis are


catalyzed by the enzymes MurA-F and result in the formation
of UDP-N-acetylmuramoyl (UDP-MurNAc)-pentapeptide.
Table 2. Multitargeting inhibitors of Mur ligases.

With the development of robust in vitro assays and high-


O

throughput screening, inhibitors of these enzymes have been


identified; however, with the exception of fosfomycin that
O
N

inhibits MurA, so far there are no other Mur inhibitors that


S

have advanced to the marketplace [50,51]. In terms of multitar-


S

MurE (S. aureus) 32 µM


MurD (E. coli) 270 µM

get therapies, the ligases MurC-F have been considered an


nd: No data available; n: Number of isolates.
O

attractive opportunity because they all catalyze the formation


CO2H

of a peptide bond and do so by binding ATP and the


UDP-MurNAc substrate. The emergence of structural
information on MurC-F suggested that the opportunity for
inhibitors that target all four enzymes might be limited
HO2C

[53]
nd

because, although the ATP-binding sites were well conserved,


8

it was revealed that UDP-MurNAc binds differently in


MurC/D compared with MurE/F [52] and so dual-targeting
Structure

rather than quad-targeting might be a more realistic outcome.


Cmpd #

Nevertheless, there have been some recent developments in


MIC
IC50

Ref.

the search for multitarget Mur ligase inhibitors and these

Expert Opin. Drug Discov. (2013) 8(2) 149


S. P. East & L. L. Silver

CO2H Br CO2H
O
NH NH
O S O OH
O O N
H
O
12 13 14

O NO2 O
OH OH
N B N B
H H
OH OH
F
Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Sussex Library on 01/20/13

15 16

Figure 4. Non-covalent PBP inhibitors.

have utilized the available X-ray structural data to perform based on the level of potency observed in the biochemical
pharmacophore-based virtual screens. assay, the compounds would not be expected to have a specific
Several publications reported on the use of MurD structures effect on bacterial cells.
for the virtual screen experiments. Perdih et al. [53] identified
benzene dicarboxylic acids that were postulated to act as gluta- 3.2 Penicillin-binding proteins
mic acid mimics in the catalytic site. The most interesting com- The b-lactam antibiotics were the first, and they are arguably
pound reported was compound 8 (Table 2), which shows the most widely, studied of all of the clinically prescribed
For personal use only.

in vitro activity on both MurD and MurE. Tomašić et al. [54] antibacterial agents. They can also be considered as the arche-
identified glutamic acid containing MurD inhibitors and these typal multitarget inhibitors because they covalently modify
compounds have subsequently evolved into dual MurD/E members of a family of enzymes known as the PBPs. PBPs
inhibitors [55] with compound 9 (Table 2), which bears some have a variety of functions in the latter stages of bacterial
structural resemblance to compound 8, showing inhibition of cell wall biosynthesis including transpeptidase activity (as
ligase activity on E. coli and S. aureus MurD and MurE. The mentioned above). Although b-lactam antibiotics have been
antibacterial activity of compound 9 on S. aureus (including very successful, one of the resistance mechanisms that have
an MRSA strain) and E. faecalis was modest and no activity emerged to this class of compounds is as a consequence of their
was seen on E. coli or P. aeruginosa up to 128 µg/ml. inherent reactivity. Bacteria express a family of enzymes termed
A common feature of compounds 8 and 9 is the rhodanine b-lactamases that inactivate b-lactams via a similar mechanism
scaffold and Tomašić et al. [56] have also published on a set of to the way that b-lactams inhibit PBPs. An established
5-benzylidenethiazolidin-4-ones with the trihydroxy deriva- method to overcome this path of resistance is to co-administer
tive, compound 10 (Table 2) showing activity on all four Mur a b-lactam antibiotic with a b-lactamase inhibitor; however,
ligases although the compound does not display antibacterial this combination approach can complicate the clinical develop-
activity on Gram-positive or Gram-negative strains up to ment. Recently, there has been much interest in developing
128 µg/ml. Rhodanines and other five-membered multihetero- reversible PBP inhibitors that do not contain the b-lactam func-
cyclic scaffolds have been the subject of much debate in terms tionality and hence may not be as susceptible to b-lactamase
of whether they are privileged structural elements or simply inactivation (although they might still bind to b-lactamases as
promiscuous hitters [57-59]. In the case of the rhodanine well as PBPs).
compound 9, the published ligand-protein X-ray structure [55] Turk et al. [61] have reported some biaryl sulfonamides and
is compelling evidence for a specific interaction on MurD. biaryl amides as noncovalent binders to PBPs. Compounds
In what seems to be a complementary virtual screen to such as those shown in compounds 12 and 13 (Figure 4) were
those described previously, Tomašić et al. [60] scrutinized the identified from a limited compound screen and show weak
X-rays structures of available E. coli and H. influenzae inhibition (IC50s ~ 100 -- 400 µM) of PBP2a (S. aureus),
MurC-F enzymes focusing on the residues in the ATP- PBP2x (S. pneumoniae) and PBP5fm (E. faecium). Some
binding sites. They concluded that the binding sites were antibacterial activity against Gram-positive organisms was
similar and subsequently performed a virtual screen using also reported (MICs 1 -- 256 µg/ml), although it was not
E. coli MurD. A selection of ligands was tested and weak determined whether the cellular inhibition observed was a
dual inhibitors of MurC and MurD were identified with consequence of PBP inhibition. The compounds were inactive
compound 11 (Table 2) being the best compound of this (MICs > 512 µg/ml) on Gram-negative organisms. Shilabin
initial set. No antibacterial activity was reported, although, et al. [62] have reported a series of 4-quinolones as pan-PBP

150 Expert Opin. Drug Discov. (2013) 8(2)


Multitarget ligands in antibacterial research: progress and opportunities

Table 3. Miscellaneous multitargeting antibacterials.

Pathway Example compound Molecular targets Ref.

Fatty acid biosynthesis OH FabF and FabH [69]


O O

HO2C N
H H
OH

DNA replication H3CO DNA polymerases III: PolC and PolE [70]
O
OCH3
Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Sussex Library on 01/20/13

N N N N OCH3
H H

OCH3
Folate biosynthesis OH Dihydropteroate synthase (DHPS) [71]
N NO2 and dihydrofolate reductase (DHFR)

H2N N NH

inhibitors. Compound 14 (Figure 4) has Kis of 5 -- 220 µM observed. Despite these differences in the structural informa-
against PBP 1a/1b, PBP2, PBP3, PBP4 and PBP5/6 from tion it is reasonable to assume that this compound is also a
E. coli. No antibacterial activity up to 1 mM on a strain of reversible inhibitor.
E. coli was observed.
For personal use only.

Boronic acids have been investigated as PBP inhibitors. 3.3 Lipid A biosynthesis
Here the concept is that the boronic acid forms a tetrahedral Gram-negative bacteria have a layer of lipopolysaccharide
intermediate with a critical serine residue in the active site (LPS) on their outer membrane that functions as a protective
of the PBPs. This reaction is similar to what happens with coating to the cell. A constitutive ingredient of LPS is
b-lactam antibiotics and PBPs (and indeed b-lactamases) Lipid A which acts as the anchor for LPS to the cell mem-
but, in the case of the boronic acid inhibitors, the reaction is brane but it also functions as an endotoxin, mobilizing the
reversible because the boron-oxygen bond that is formed can innate immune system in humans and leading to sepsis.
be readily hydrolyzed. Structural information on how boronic Lipid A is crucial for the survival of Gram-negative bacteria
acids bind to PBPs is beginning to emerge and this should so an inhibitor of the one or more steps in the synthesis of
facilitate improved reversible inhibitors in the future. Lipid A could provide a new therapy. The first three steps
A series of N-alkyl boronates were reported by Woon in Lipid A biosynthesis are catalyzed by the enzymes LpxA,
et al. [63] and Contrerase-Martel et al. [64]. Compound 15 LpxC and LpxD. LpxC has garnered much attention from
(Figure 4) is one of the most interesting compounds in the drug discovery community [66] with ACHN-975 recently
this series and it was active on PBP1b (IC50 20 µM) and being the first LpxC inhibitor to enter human trials [67].
also shown to have activity on PBP2, PBP2a, PBP3, Jenkins and Dotson [68] have suggested that LpxA and
PBP4 together with sub-micromolar inhibition of the PBP LpxD might offer an opportunity for a dual-targeted
R39 (IC50 0.27 µM). Antibacterial activity on S. aureus therapy. These two enzymes are both acyltransferases
(MIC 32 µg/ml including MRSA strains) and other Gram- and have been shown to be inhibited by the dodecapeptide
positive organisms was demonstrated and X-ray structural RJPXD33 with Kd values measured as 22 µM (E. coli
studies suggested that compounds of this type can adopt LpxA) and 6.5 µM (E. coli LpxD), respectively. Clearly the
two different binding modes. In a more recent paper, development of a long peptide chain as a novel antibacterial
Zervosen et al. [65] have described related glycine boronic is likely to be challenging; however, improving the under-
acids, for example, compound 16 (Figure 4) with activity on standing of the minimum pharmacophoric features of the
S. pneumoniae PBP1b (IC50 26 µM), S. pneumoniae PBP2x peptide required for binding might present possibilities for
(IC50 138 µM) and Actinomadura R39 (IC50 0.6 µM). small molecule inhibition.
Curiously, X-ray data on compound 16 crystallized with
R39 revealed that the tetrahedral intermediate forms by inter- 4. Miscellaneous multitarget ligands
actions with two serine residues and a lysine residue in the
active site. This has been described as a tricovalent interaction; Other multitarget ligands have been reported in the literature
however, the ligand-protein complex of compound 16 with and some of the salient examples are included in Table 3
PBP1b revealed that only a monocovalent interaction is [69-71]. As there is little new relevant information that

Expert Opin. Drug Discov. (2013) 8(2) 151


S. P. East & L. L. Silver

merits inclusion in this review, the readers are directed to ligands have been identified for biological targets unrelated in
other articles that contain the key references and a more protein sequence or structure. The idea here is to find addi-
comprehensive summary of the work shown in Table 3 [1,3]. tional biological targets for ligands already known to inhibit
one enzyme of interest.
5. Future opportunities
6. Expert opinion
Multitarget ligands need to modulate the pharmacological
response of multiple enzymes. Looking at this challenge Multitarget ligands represent an approach to discovery of
simplistically, an ideal ligand would inhibit the enzymes of novel antibacterials that should yield drugs having a low
interest to a similar degree (perhaps within an order of magni- probability of selecting rapidly for high-level target-based
tude [4]), as determined by in vitro biochemical experiments. resistance. While all antibacterials will eventually fall to resis-
In reality, the situation is much more complex and there tance mechanisms, it is highly likely that antibacterial com-
Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Sussex Library on 01/20/13

will be many factors that ultimately determine what the pri- pounds targeting single enzymes will immediately select for
mary target is in vivo and these are not easy to predict. For pre-existing resistance mutations that arise at a measurable
example, ligand structure and enzyme-binding site structure rate per generation. Indeed, a standard method of identifying
have both been reported to influence the primary target of the mechanism of action of a novel compound is to select for
dual GyrB/ParE inhibitors [41] in different bacterial strains. such target-site mutations. Whether the rate of resistance seen
What is clear though is that true, multitarget ligands can in the laboratory will translate into therapeutic failure
reduce in vitro spontaneous resistance frequencies (selected depends upon (among other things) that rate, the bacterial
at high levels of compound) to frequencies below those for burden during the infection, the fitness and virulence of the
single mutations; sequential mutations in more than one resistant mutants and the possibility of reaching high enough
essential target would be necessary in order for the bacteria levels of compound at the site of infection to inhibit the
to develop high-level resistance to a multitargeted drug. If resistant organisms. While there are single-enzyme targeted
the inhibitor is well balanced, then the likelihood of single agents in clinical use, they are usually dosed as combinations
For personal use only.

step resistance is decreased. (as in therapy of Mycobacterium tuberculosis) or used topically


To search for new multitarget inhibitors in the future, the or in indications where high concentrations can be achieved
most obvious opportunities are to identify enzymes that (such as urinary tract infections).
have either identical substrates (e.g., ATP in the case of As discussed here, DNA gyrase/topo IV and the PBPs are
GyrB and ParE) or structurally related substrates (e.g., the multienzyme targets of clinically validated drugs and
UDP-MurNAc in the case of Mur C-F enzymes) where the they represent new options going forward with novel chemi-
homology of the binding site is expected to be high. Protein cal classes: for example, dual inhibitors of GyrB and ParE as
sequence homology is usually a reliable guide for similar pro- well as reversible inhibitors of PBPs. Table 3 notes three
teins, but similar binding sites can be formed by the same spa- other inhibitors of different dual enzyme targets. Another
tial arrangement of similar residues in otherwise dissimilar dual targeting opportunity, specifically for Gram-negatives,
sequences. Once the proteins of interest have been identified, is the two early transacylases of Lipid A synthesis, LpxA
then X-ray crystallography data on these related targets can and LpxD. Recently a small peptide, RJPXD33, has been
then potentially be collected to be sure that the three- shown to bind to both enzymes (albeit to LpxD with greater
dimensional substrate binding sites of different enzymes are affinity than to LpxA), to be competitive with substrate
truly similar. As soon as this is confirmed, then screening for binding to LpxD and to exhibit antibacterial activity
methods in the form of HTS or virtual screens can be imple- (when produced intracellularly) that is abrogated by over-
mented to identify new multitarget ligands. Some of the expression of LpxD [68]. Thus with further study, this
examples presented in this review illustrates that structure- type of probe peptide may be useful in designing small mole-
led approaches like this have been successful in the recent cule inhibitors with the ability to enter the Gram-negative
past and it seems reasonable that there are opportunities to cytoplasm (see below).
exploit this further in the future. For example there are other The recognition that polypharmacology can have positive
bacterial ATPases that could be explored to identify new implications has been especially apparent in the area of kinase
classes inhibitors as suggested by Škedelj et al. [72]. inhibitors in cancer chemotherapy, with the caveat that such
Where X-ray crystal structures are not available, the chal- multitargeting must be balanced, demonstrating selectivity
lenge to identify new multitargeting opportunities requires without promiscuity [75]. In bacteria, it has been recognized
a more indirect approach. One strategy is to construct homol- that the successful systemic monotherapeutic antibacterials
ogy models based on enzymes with related sequence and are multitargeted, at least at the genetic level (in that targets
substrate. An alternative strategy to the protein-centric such as cell wall precursors, ribosomal RNA, membranes,
approaches described is to focus on known ligand similarity. DNA are products of multiple genes or structures produced
In silico tools have been utilized by Keiser et al. [73] and by multiple genes), and it has been proposed that this
Durrant et al. [74] where secondary biological targets to known multitargeting prevents rapid resistance selection [3]. But the

152 Expert Opin. Drug Discov. (2013) 8(2)


Multitarget ligands in antibacterial research: progress and opportunities

same caveats as for kinases would apply in the search for novel For Gram-negatives, the double membrane with its orthogo-
antibacterial multitarget ligands. nal sieving properties presents a formidable barrier for which
Indeed, an important aspect of antibacterial discovery in no set of chemical rules for its passage have been formulated.
general, also applicable in the case of multitarget ligands, is Thus, although target structure can guide rational design of
that the mechanism of action at the whole-cell level of inhibitors, the physicochemical properties of the compound
enzyme inhibitors cannot be inferred from their activity on enabling cell entry must be considered for antibacterial
the isolated enzymes or, in the case of putative dual-targeting activity -- preferably during the optimization process. For
agents, from the inability to obtain mutants. Independent example, it is highly likely that inability to find membrane
proof of mechanism is needed [1]. With specific pharmaco- permeant compounds has stymied the discovery, despite
phoric features in ligands that target multiple enzymes, there much effort, of validated antibacterial inhibitors of the Mur
is a finite likelihood that other targets will be inhibited as ligases, mentioned above as potential multitarget enzymes,
well. Such extra activities might have implications for toxic- since their common binding sites are for UDP-MurNAc
Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Sussex Library on 01/20/13

ity. The possibility of nonselectivity is illustrated by the find- and ATP substrates, and may thus require nonpromiscuous
ings, noted above, with series of GyrB/ParE compounds in permeant diphosphate isosteres.
studies by Trius [48]. Even small changes in structure that Thus, antibacterial programs for the design of multitarget
had little effect on topoisomerase inhibition led to altera- ligands present an important opportunity for production of
tions in whole cell activity which proved to be due to inhibi- antibacterial agents with longevity due to slow development
tion of macromolecular synthesis in addition to DNA. For of resistance comparable to that seen with other successful
the GyrB/ParE inhibitors kibdelomycin [35] and querce- agents, such as the fluoroquinolones -- but much improved
tin [38], the available data does show inhibition of the topoi- over single-targeted agents for which resistance can appear
somerases but does not exclude (and even demonstrates) overnight (at least in the laboratory).
inhibition of other cellular processes at concentration near
the MIC. With the rhodanine inhibitors of Mur ligases, Acknowledgments
mentioned above, while there is crystallographic data
For personal use only.

supporting specific enzyme interaction, the selectivity (anti- The authors would like to thank R Law, J Heim, J Manchester,
bacterial activity being due solely to inhibition of the ligases) J Finn, J Palmer and D Haydon for their assistance preparing
remains unproven [56,57]. this manuscript.
A major obstacle to target-based antibacterial discovery is
the necessity for enzyme inhibitors to reach their target Declaration of interest
site [1]. This is not as critical for activity against extracytoplas-
mic targets of Gram-positives (such as b-lactams and SP East is an employee of Evotec (UK) Ltd, while LL Silver is
glycopeptides), but is important for periplasmic targets in an independent consultant in antibacterial discovery at LL
Gram-negatives and cytoplasmic targets of all pathogens. Silver Consulting, LLC.

Expert Opin. Drug Discov. (2013) 8(2) 153


S. P. East & L. L. Silver

Bibliography
Papers of special note have been highlighted as 10. Morphy R, Rankovic Z. The 23. Black MT, Stachyra T, Platel D, et al.
either of interest () or of considerable interest physicochemical challenges of designing Mechanism of action of the antibiotic
() to readers. multiple ligands. J Med Chem NXL101, a novel nonfluoroquinolone
2006;49:4961-70 inhibitor of bacterial type II
1. Silver LL. Challenges of antibacterial
11. Papp-Wallace KM, Endimiani A, topoisomerases.
discovery. Clin Microbiol Rev
Taracila MA, Bonomo RA. Antimicrob Agents Chemother
2011;24:71-109
Carbapenems: past, present, and future. 2008;52:3339-49
2. Walsh C. Molecular mechanisms that
Antimicrob Agents Chemother 24. Miles TJ, Barfoot C, Brooks G, et al.
confer antibacterial drug resistance.
2011;55:4943-60 Novel cyclohexyl-amides as potent
Nature 2000;406:775-81
12. Drlica K, Hiasa H, Kerns R, et al. antibacterials targeting bacterial type
3. Silver LL. Polypharmacology as an IIA topoisomerases. Bioorg Med
Quinolones: action and resistance
emerging trend in antibacterial discovery Chem Lett 2011;21:7483-8
Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Sussex Library on 01/20/13

updated. Curr Top Med Chem


in Polypharmacology in drug discovery.
2009;9:981-98 25. Miles TJ, Axten JM, Barfoot C, et al.
John Wiley & Sons, Inc., Hoboken,
13. Sanyal G, Doig P. Bacterial Novel amino-piperidines as potent
New Jersey; 2012. p. 167-202
DNA replication enzymes as targets for antibacterials targeting bacterial type
4. Morphy R, Rankovic Z. Designed IIA topoisomerases. Bioorg Med
antibacterial drug discovery. Expert Opin
multiple ligands. An emerging drug Chem Lett 2011;21:7489-95
Drug Discov 2012;7:327-39
discovery paradigm. J Med Chem
14. Collin F, Karkare S, Maxwell A. 26. Geng B, Comita-Prevoir J,
2005;48:6523-43
Exploiting bacterial DNA gyrase as a Eyermann CJ, et al. Exploring
.. Key perspective article on strategies
drug target: current state and Left-hand-side substitutions in the
employed in multitargeted drug
perspectives. Appl Microbiol Biotechnol benzoxazinone series of
discovery programs, classification of
2011;92:479-97 4-amino-piperidine bacterial type IIa
designed multiple ligands and
topoisomerase inhibitors. Bioorg Med
historical example of compounds 15. Butler MS, Cooper MA. Antibiotics in
Chem Lett 2011;21:5432-5
interacting with multiple the clinical pipeline in 2011. J Antibiot
For personal use only.

biological targets. 2011;64:413-25 27. Reck F, Alm R, Brassil P, et al. Novel


N-linked aminopiperidine inhibitors of
5. Metz JT, Hajduk PJ. Rational 16. Pucci MJ, Podos SD, Thanassi JA, et al.
bacterial topoisomerase type II:
approaches to targeted In vitro and in vivo profiles of
broad-spectrum antibacterial agents with
polypharmacology: creating and ACH-702, an isothiazoloquinolone,
reduced hERG activity. J Med Chem
navigating protein-ligand interaction against bacterial pathogens.
2011;54:7834-47
networks. Curr Opin Chem Biol Antimicrob Agents Chemother
2010;14:498-504 2011;55:2860-71 28. Reck F, Alm RA, Brassil P, et al. Novel
N-linked aminopiperidine inhibitors of
6. Fischbach MA. Combination therapies 17. http://www.achillion.com/ACH_702
bacterial topoisomerase type II with
for combating antimicrobial resistance. 18. Aravind L, Leipe DD, Koonin EV. reduced pKa: antibacterial agents with an
Curr Opin Microbiol 2011;14:519-23 Toprim -- a conserved catalytic domain improved safety profile. J Med Chem
. A compact review article describing in type IA and II topoisomerases, 2012;55:6916-33
different approaches for combination DnaG-type primases, OLD family
therapies with antibacterial drugs. 29. Wiles JA, Phadke AS, Bradbury BJ, et al.
nucleases and RecR proteins.
Selenophene-containing inhibitors of type
7. Morphy R, Kay C, Rankovic Z. From Nucleic Acids Res 1998;26:4205-13
IIA bacterial topoisomerases.
magic bullets to designed multiple 19. Kim HY, Wiles JA, Wang Q, et al. J Med Chem 2011;54:3418-25
ligands. Drug Discov Today Exploration of the activity of
2004;9:641-51 30. Bax BD, Chan PF, Eggleston DS, et al.
7-pyrrolidino-8-
Type IIA topoisomerase inhibition by a
8. Morphy R, Rankovic Z. Design of methoxyisothiazoloquinolones against
new class of antibacterial agents. Nature
multitarget ligands in Lead generation methicillin-resistant Staphylococcus
2010;466:935-40
approaches in drug discovery. aureus (MRSA). J Med Chem . X-ray evidence illustrating the binding
John Wiley & Sons, Inc., Hoboken, 2011;54:3268-82
mode of the non-quinolone bacterial
New Jersey; 2010. p. 141-64 20. Heim J. Safe drugs for bad bugs: topoisomerase II inhibitor
9. Pokrovskaya V, Baasov T. Dual-acting EV-035, a new topoisomerase inhibitor. GSK299423 with DNA gyrase, thus
hybrid antibiotics: a promising strategy Available from: http://www.evolva.com/ rationalizing the activity of this
to combat bacterial resistance. Exp Opin pharmaceuticals/ev-035 compound on fluoquinolone resistant
Drug Discov 2010;5:883-902 21. Evolva SA. 2-Pyridone antimicrobial bacterial strains.
. Comprehensive account of compositions. WO2012047487; 2012 31. Widdowson K, Hennessy A. Advances in
dual-targeting ligands that are a
22. Li Q, Mitscher LA, Shen LL. The structure-based drug design of novel
combination of two discrete
2-pyridone antibacterial agents: bacterial bacterial topoisomerase inhibitors.
antibacterial compounds
topoisomerase inhibitors. Med Res Rev Future Med Chem 2010;2:1619-22
joined together.
2000;20:231-93

154 Expert Opin. Drug Discov. (2013) 8(2)


Multitarget ligands in antibacterial research: progress and opportunities

32. Wohlkonig A, Chan PF, Fosberry AP, structure-activity relationships. a rich source of drug targets.
et al. Structural basis of quinolone J Med Chem 2008;51:5243-63 Crit Rev Biotechnol 2011;31:295-336
inhibition of type IIA topoisomerases . Medicinal chemistry summary of the 51. Silver LL. Does the cell wall of bacteria
and target-mediated resistance. benzimidazole ureas including a remain a viable source of targets for
Nat Struct Mol Biol 2010;17:1152-3 hypothesis on the importance of the novel antibiotics? Biochem Pharmacol
33. Oblak M, Kotnik M, Solmajer T. conformation of the small molecule for 2006;71:996-1005
Discovery and development of ATPase dual GyrB/ParE inhibition and in vivo
52. Smith CA. Structure, function and
inhibitors of DNA gyrase as antibacterial efficacy data.
dynamics in the mur family of bacterial
agents. Curr Med Chem 41. Grossman TH, Bartels DJ, Mullin S, cell wall ligases. J Mol Biol
2007;14:2033-47 et al. Dual targeting of GyrB and ParE 2006;362:640-55
34. Angehrn P, Goetschi E, Gmuender H, by a novel aminobenzimidazole class of
53. Perdih A, Kovač A, Wolber G, et al.
et al. A new DNA gyrase inhibitor antibacterial compounds.
Discovery of novel benzene
subclass of the cyclothialidine family Antimicrob Agents Chemother
Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Sussex Library on 01/20/13

1,3-dicarboxylic acid inhibitors of


based on a bicyclic dilactam-lactone 2007;51:657-66
bacterial MurD and MurE ligases by
scaffold. Synthesis and antibacterial 42. Wei Y, Letiran A. PDB accession code: structure-based virtual screening
properties. J Med Chem 3FV5 approach. Bioorg Med Chem Lett
2011;54:2207-24 43. Vertex pharmaceticals incorporated. 2009;19:2668-73
35. Phillips JW, Goetz MA, Smith SK, et al. Pyrimidine gyrase and topoisomerase IV 54. Tomašić T, Zidar N, Rupnik V, et al.
Discovery of kibdelomycin, a potent new inhibitors. WO2012097269; 2012 Synthesis and biological evaluation of
class of bacterial type II topoisomerase 44. East SP, Czaplewski LG, Haydon DJ. new glutamic acid-based inhibitors of
inhibitor by chemical-genetic profiling in Ethyl urea inhibitors of the bacterial MurD ligase. Bioorg Med Chem Lett
Staphylococcus aureus. Chem Biol type II topoisomerase DNA gyrase 2009;19:153-7
2011;18:955-65 (GyrB) and topoisomerase IV (ParE) in 55. Tomašić T, Šink R, Zidar N, et al. Dual
36. Hossion AM, Otsuka N, Kandahary RK, designing multi-target drugs. inhibitor of MurD and MurE ligases
et al. Design, synthesis, and biological RSC Publishing, Cambridge; from Escherichia coli and Staphylococcus
evaluation of a novel series of quercetin 2012. p. 335-52
For personal use only.

aureus. ACS Med Chem Lett


diacylglucosides as potent 45. Palmer J. Dual-targeting 2012;3:626-30
anti-MRSA and anti-VRE agents. DNA supercoiling inhibitors for the 56. Tomašić T, Zidar N, Kovač A, et al.
Bioorg Med Chem Lett treatment of bacterial infections. 5-Benzylidenethiazolidin-4-ones as
2010;20:5349-52 Presented at the challenges of multitarget inhibitors of bacterial Mur
37. Hossion AM, Zamami Y, antibacterial drug development; ligases. Chem Med Chem 2010;5:286-95
Kandahary RK, et al. Quercetin San Diego; 2012
57. Tomašić T, Peterlin Mašič L. Rhodanine
diacylglycoside analogues showing dual 46. Manchester JI, Dussault DD, Rose JA, as a scaffold in drug discovery: a critical
inhibition of DNA gyrase and et al. Discovery of a novel azaindole class review of its biological activities and
topoisomerase IV as novel antibacterial of antibacterial agents targeting the mechanisms of target modulation.
agents. J Med Chem 2011;54:3686-703 ATPase domains of DNA gyrase and Expert Opin Drug Discov
38. Cushnie TP, Lamb AJ. Antimicrobial topoisomerase IV. Bioorg Med 2012;7:549-60
activity of flavonoids. Int J Chem Lett 2012;22:5150-6
58. Mendgen T, Steuer C, Klein CD.
Antimicrob Agents 2005;26:343-56 47. Manchester JI, Buurman ET, Privileged scaffolds or promiscuous
39. Boehm HJ, Boehringer M, Bur D, et al. Bisacchi GS, McLaughlin RE. Molecular binders: a comparative study on
Novel inhibitors of DNA gyrase: 3D determinants of AcrB-mediated bacterial rhodanines and related heterocycles in
structure based biased needle screening, efflux implications for drug discovery. medicinal chemistry. J Med Chem
hit validation by biophysical methods, J Med Chem 2012;55:2532-7 2012;55:743-53
and 3D guided optimization. 48. Finn J. The discovery of potent dual 59. Baell JB, Holloway GA. New
A promising alternative to random targeting pyrrolopyrimidine inhibitors of substructure filters for removal of pan
screening. J Med Chem bacterial DNA gyrase B and assay interference compounds (PAINS)
2000;43:2664-74 topoisomerase IV with broad spectrum
.
from screening libraries and for their
Important fragment-based screening antibacterial activity. Presented at the exclusion in bioassays. J Med Chem
paper in the identification of novel challenges of antibacterial drug 2010;53:2719-40
ligands modulating the effect of an development; San Diego; 2012
essential bacterial enzyme. 60. Tomašić T, Kovač A, Klebe G, et al.
49. Bugg TD, Braddick D, Dowson CG, Virtual screening for potential inhibitors
40. Charifson PS, Grillot AL, Grossman TH, Roper DI. Bacterial cell wall assembly: of bacterial MurC and MurD ligases.
et al. Novel dual-targeting benzimidazole still an attractive antibacterial target. J Mol Model 2012;18:1063-72
urea inhibitors of DNA gyrase and Trends Biotechnol 2011;29:167-73
topoisomerase IV possessing potent 61. Turk S, Verlaine O, Gerards T, et al.
50. Gautam A, Vyas R, Tewari R. New noncovalent inhibitors of
antibacterial activity: intelligent design
Peptidoglycan biosynthesis machinery: penicillin-binding proteins from
and evolution through the judicious use
of structure-guided design and

Expert Opin. Drug Discov. (2013) 8(2) 155


S. P. East & L. L. Silver

penicillin-resistant bacteria. PLoS One agents. Curr Med Chem 72. Škedelj V, Tomašić T, Mašič LP,
2011;6:e19418 2012;19:2038-50 Zega A. ATP-binding site of bacterial
62. Shilabin AG, Dzhekieva L, Misra P, 67. Achaogen announces all objectives met in enzymes as a target for antibacterial drug
et al. 4-Quinolones as noncovalent Phase 2 plazomicin complicated urinary design. J Med Chem 2011;54:915-29
Inhibitors of high molecular mass tract infections study and start of 73. Keiser MJ, Roth BL, Armbruster BN,
penicillin-binding proteins. ACS Med first-in-human study with ACHN-975. et al. Relating protein pharmacology by
Chem Lett 2012;3:592-5 Available from: http://www.achaogen. ligand chemistry. Nat Biotechnol
63. Woon ECY, Zervosen A, Sauvage E, com/news/148 2007;25:197-206
et al. Structure guided development of 68. Jenkins RJ, Dotson GD. Dual targeting 74. Durrant JD, Amaro RE, Xie L, et al.
potent reversibly binding penicillin antibacterial peptide inhibitor of early A multidimensional strategy to detect
binding protein inhibitors. ACS Med lipid A biosynthesis. ACS Chem Biol polypharmacological targets in the
Chem Lett 2011;2:219-23 2012;7:1170-7 absence of structural and sequence
homology. PLoS Comput Biol
Expert Opin. Drug Discov. Downloaded from informahealthcare.com by University of Sussex Library on 01/20/13

64. Contreras-Martel C, Amoroso A, 69. Wang J, Kodali S, Lee SH. Discovery of


Woon EC, et al. Structure-guided design platencin, a dual FabF and FabH 2010;6:e1000648
of cell wall biosynthesis inhibitors that inhibitor with in vivo antibiotic 75. Morphy R. Selectively nonselective kinase
overcome b-lactam resistance in properties. Proc Natl Acad Sci USA inhibition: striking the right balance.
Staphylococcus aureus (MRSA). 2007;104:7612-16 J Med Chem 2010;53:1413-37
ACS Chem Biol 2011;6:943-51 70. Ali A, Taylor GE. Development of
65. Zervosen A, Bouillez A, Herman A, et al. DNA polymerase IIIC inhibitors for the Affiliation
Synthesis and evaluation of boronic acids treatment of Gram-positive bacterial Stephen P East†1 & Lynn L Silver2

as inhibitors of penicillin binding infections. Expert Opin Ther Patents Author for correspondence
1
proteins of classes A, B and C. 2005;15:947-53 Evotec (UK) Ltd., 114 Milton Park,
Bioorg Med Chem 2012;20:3915-24 Abingdon, Oxfordshire OX14 4SA, UK
71. Bennett BC, Xu H, Simmerman RF,
66. Zhang J, Zhang L, Li X, Xu W. UDP-3- et al. Crystal structure of the anthrax E-mail: stephen.east@evotec.com
2
O-(R-3-hydroxymyristoyl)-N- drug target, Bacillus anthracis LL Silver Consulting, LLC,
955 S. Springfield Ave.,
For personal use only.

acetylglucosamine deacetylase (LpxC) dihydrofolate reductase. J Med Chem


inhibitors: a new class of antibacterial 2007;50:4374-81 Unit C403 Springfield,
NJ 07081, USA

156 Expert Opin. Drug Discov. (2013) 8(2)

You might also like