Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

REVIEW

Targeting virulence: a new paradigm for


antimicrobial therapy
Anne E Clatworthy, Emily Pierson & Deborah T Hung

Clinically significant antibiotic resistance has evolved against virtually every antibiotic deployed. Yet the development of new
classes of antibiotics has lagged far behind our growing need for such drugs. Rather than focusing on therapeutics that target in
vitro viability, much like conventional antibiotics, an alternative approach is to target functions essential for infection, such as
virulence factors required to cause host damage and disease. This approach has several potential advantages including expanding the
repertoire of bacterial targets, preserving the host endogenous microbiome, and exerting less selective pressure, which may result
in decreased resistance. We review new approaches to targeting virulence, discuss their advantages and disadvantages, and propose
that in addition to targeting virulence, new antimicrobial development strategies should be expanded to include targeting bacterial
gene functions that are essential for in vivo viability. We highlight both new advances in identifying these functions and prospects for
antimicrobial discovery targeting this unexploited area.

Since the introduction of penicillin, the deployment of any novel antibio- stand pathogenesis has begun to elucidate mechanisms that could be
tic has been followed by the evolution of clinically significant resistance targeted to clear the infection in lieu of targeting simply in vitro bacterial
to that antibiotic in as little as a few years (Fig. 1)1. It is clear that we are viability. Instead, targeting bacterial virulence or disrupting the interac-
in a race to develop new antimicrobials to supplement our dwindling tion between the host and the pathogen are attractive options that are
antibiotic arsenal for combating the growing emergence of antibiotic- increasingly being explored.
resistant strains. Currently, we are losing this race. The Infectious Disease The conventional concept of virulence is defined by the ability of a
Society of America estimates that 70% of hospital-acquired infections pathogen to cause disease; virulence determinants are defined as bacte-
in the United States are resistant to one or more antibiotics. Yet with rial factors (for example, toxins, cytolysins or proteases) or mechanisms
the exception of the recent development of the narrow-spectrum drugs that actively cause damage to host tissues. Thus, efforts to develop anti-
daptomycin and linezolid, there have been no new classes of clinically virulence therapies are geared at ‘disarming’ the pathogen by inhibiting
relevant antibiotics discovered in over 40 years. virulence factors that can cause direct harm to the host. In theory then,
Traditional antibiotics have been identified for their ability to kill compounds that target virulence create an in vivo scenario that is similar
bacteria (bacteriocidal) or inhibit growth (bacteriostatic). They act by to vaccination with a live, attenuated strain. The bacteria are eventually
inhibiting bacterial functions (such as cell wall synthesis, DNA replica- cleared by the host immune response with little to no impact on the
tion, RNA transcription and protein synthesis) that are essential for in normal human microbiota. A potential (though yet unproven) advantage
vitro, logarithmic growth (Fig. 2). Although it is clear that antibiotics of this approach is that new antimicrobials aimed at inhibiting virulence
aimed at cellular viability have historically been highly effective, these rather than growth may impose weaker selective pressure for the develop-
modes of action impose selective pressure that fosters the growth of ment of antibiotic resistance relative to current antibiotics.
antibiotic-resistant strains. Given the current gap between our ability to Here we review recent strategies that target various pathways related to
develop novel antibiotics and the real need for such drugs, the threat of virulence, including inhibiting toxin function, toxin delivery, regulation
a postantibiotic era is looming large on the horizon. Therefore, in addi- of virulence expression, and bacterial adhesion (summarized in Table 1
tion to compounds that act by targeting in vitro cell growth, we must and Figure 3). Though this review is not entirely comprehensive, we
consider developing antimicrobials that have novel modes of action. New highlight some of the major approaches being taken to target virulence.
approaches have been revealed by the tremendous effort over the past few At the end of this review, we explore an even broader framework for new
decades to understand how bacteria cause disease. This effort to under- antimicrobial development that includes not only the strategy of target-
ing virulence but also the strategy of targeting bacterial in vivo essential
Department of Molecular Biology and Center for Computational and Integrative gene functions—gene functions that are required for survival within the
Biology, Massachusetts General Hospital, 185 Cambridge St., Boston, host yet that are distinct from those required for in vitro viability. The
Massachusetts 02114, USA; Department of Microbiology and Molecular
Genetics, Harvard Medical School, 200 Longwood Ave., Boston, Massachusetts
targets of both of these therapeutic strategies (that is, virulence factors
02115, USA; and the Broad Institute of MIT and Harvard, 7 Cambridge Center, and in vivo essentials) are required for infection, yet the utility of these
Cambridge, Massachusetts 02142, USA. Correspondence should be addressed to strategies has only recently begun to be explored. Given the need for
D.T.H. (hung@molbio.mgh.harvard.edu). antibiotics with novel modes of action, consideration of these approaches
Published online 20 August 2007; doi:10.1038/nchembio.2007.24 is warranted.

NATURE CHEMICAL BIOLOGY VOLUME 3 NUMBER 9 SEPTEMBER 2007 541


REVIEW

Antibiotic deployment a search for small molecules that would pro-


tect the population from anthrax, Merck has
Tetracycline
identified a hydroxymate (LFI) that inhibits LF
Chloramphenicol Vancomycin
protease activity and promotes cellular survival
Streptomycin Ampicillin
in a macrophage cytotoxicity assay9. LFI (Fig.
Sulfonamides Erythromycin Cephalosporins Daptomycin
3b) binds the active site of Bacillus anthracis
Penicillin Methicillin Linezolid
LF and offers complete protection from spore
infection when administered to mice in combi-
1930 1935 1940 1945 1950 1955 1960 1965 1970 1975 1980 1985 1990 1995 2000 2005
nation with ciprofloxacin 66 h postinfection in
a model in which ciprofloxacin alone only offers
Sulfonamides Chloramphenicol Ampicillin Vancomycin Linezolid
50% protection from lethality9. Along similar
Penicillin Streptomycin Erythromycin lines, a mixture-based peptide library was used
Daptomycin
Tetracycline Methicillin to determine the optimal peptide substrate
Cephalosporins sequence for LF, which was then used to design
Antibiotic resistance observed peptide analogs that inhibit LF activity in vitro
and protect macrophages from LF-induced
Figure 1 Timeline of antibiotic deployment and the evolution of antibiotic resistance. The year each cytolysis10.
antibiotic was deployed is depicted above the timeline, and the year resistance to each antibiotic was Compounds that inhibit anthrax toxin are
observed is depicted below the timeline (with the caveat that the appearance of antibiotic resistance not limited to targeting LF or EF. PA is also
does not necessarily imply that a given antibiotic has lost all clinical utility). a potential target that could be inhibited in
multiple ways. Strategies include inhibition of
binding of PA to its host receptor, processing of
Inhibition of toxin function PA by host proteases, binding of processed PA to LF or EF, or transloca-
Many pathogenic bacteria cause damage to host tissues through the tion of LF or EF by physically blocking the PA heptamer pore8. Recent
release of toxins, which are proteins that act to perturb host cell functions work indicates that a phenylalanine clamp controls protein translocation
and may ultimately result in host cell death. Thus, an obvious approach through the heptameric PA pore and that this clamp can be targeted with
to inhibiting bacterial virulence is disruption of toxin function, which small molecules that block the pore11. Alternatively, one could interfere
can occur in a direct manner by inhibition of the toxin activity itself, or with toxin translocation by inhibiting formation of the pore itself by
in an indirect manner, by modulating the host response to the toxin. inhibiting endosome acidification12,13 or by inhibiting PA heptamer
In fact, direct inhibition is the basis for the historical use of antitoxins assembly. Cisplatin seems to prevent both LF and EF toxicity by inhibit-
(antibodies) against toxins such as diptheria, botulinus, tetanus and other ing heptamer assembly, and simultaneous administration of cisplatin
toxins. Recently, much effort has been focused on inhibiting the effects of with a lethal dose of anthrax lethal toxin has been shown to be protec-
the three proteins that comprise anthrax toxin: lethal factor (LF), edema tive in rodent models, though a delay in cisplatin administration was
factor (EF) and protective antigen (PA) (Fig. 3a). shown to be ineffective14. This result should be contrasted with the result
Though each toxin component alone is nontoxic, the pairing of either obtained in ciprofloxacin and LFI combination therapy, which works
LF or EF with PA results in characteristic toxin activity that is ultimately even in delayed administration. This difference underscores the concept
responsible for the pathology of the disease2. Much is already understood that the specific mechanism of virulence inhibition may determine the
about toxin entry into mammalian cells. First, individual PA monomers efficacy of a particular small molecule in targeting virulence at different
diffuse to the surface of mammalian cells where they are proteolytically stages of infection (see below).
cleaved by host proteases. Cleaved PA spontaneously oligomerizes into Rather than directly inhibiting toxin function, one could block the
heptamers, which can bind either LF or EF. PA-EF PA-LF complexes are downstream effects of the toxin by targeting host proteins. For example, in
then endocytosed and trafficked to the endosome, where a drop in the pH secretory diarrheas and cholera, Cl– secretion is central to intestinal fluid
triggers a conformational change in PA that converts it to a transmem- secretion (the disease pathology); thus inhibitors of Cl– channels may
brane pore. Ultimately LF and EF are translocated through PA’s pore into have therapeutic value in blocking these diarrheas. A screen for inhibi-
the host cytosol where they exert their respective toxic effects (Fig. 3a). tors of the cystic fibrosis transmembrane conductance regulator (CFTR)
EF is a calmodulin-dependent adenylate cyclase whose action is known protein15, a cAMP-activated Cl– channel that is defective in individuals
to result in prolonged increases in cyclic AMP levels, but it is otherwise with cystic fibrosis, found that compounds of the 2-thioxo-4-thiazolidi-
poorly understood. The exact mechanism by which LF, a Zn2+ protease, none class inhibit the CFTR protein. The most potent thiazolidinone
exerts its cytotoxic effect in vivo is also not clearly understood. Genetic compound was evaluated for its ability to inhibit mouse intestinal fluid
evidence suggests that LF-mediated cell death is dependent on suscep- secretion after oral administration of cholera toxin. A single intraperito-
tible alleles of the Nlrp1b gene, as well as the gene encoding caspase-1 in neal injection of the CFTR inhibitor decreased the level of fluid secretion
murine macrophages3. LF can also cleave the N termini of several mito- by more than 90% after administration of cholera toxin. Although it is
gen-activated protein (MAP) kinase kinases4,5. However, it is unclear how still unclear whether administration of this compound can positively
LF-mediated MAPKK cleavage is related to cytotoxicity, though small influence the resolution of infection with Vibrio cholerae, this finding
molecules that activate MAP kinase cascades have been shown to protect underscores the notion that new therapeutics aimed at targeting virulence
mouse macrophages from LF-induced cell death6. Apart from causing cell need not be absolutely restricted to pathogen proteins.
death, LF can also paralyze actin-based motility in neutrophils—an effect
that has been correlated with decreased levels of Hsp27 and p38 MAP Targeting bacterial toxin delivery
kinase phosphorylation in LT-treated, cultured neutrophils7. In addition to targeting bacterial toxin function, one could also inhibit
Regardless of the precise mechanism of activity of LF, it has certainly virulence by interfering with appropriate delivery of the toxin to its
been the focus of great attention as a target for new antimicrobials8. In site of action. This principle has been applied for several decades in the

542 VOLUME 3 NUMBER 9 SEPTEMBER 2007 NATURE CHEMICAL BIOLOGY


REVIEW

treatment of the antibiotic-associated diarrheal disease caused by


Clostridium difficile16. Cholestyramine simply binds the clostridial tox-
ins, preventing their delivery or accessibility to the intestinal epithelium
and thus blunting their toxic effects. Cholestyramine binds toxin B (a Inhibition of
cytotoxin) and likely toxin A (an enterotoxin) as well, though its use has Inhibition of DNA or RNA synthesis protein synthesis

been limited to relatively mild cases of the disease.


A more recent application of this principle is prevention of toxin (or
effector) delivery by inhibiting bacterial secretion systems. Many proteins
involved in bacterial secretion are specific to prokaryotes and are therefore
dTTP
rational candidates to target with novel antimicrobials. Recently, there has GTP
Bacterial cell
been interest in targeting the type III secretion system (T3SS) common DHP DHF
dTMP

to Yersinia spp., Pseudomonas aeruginosa, pathogenic Escherichia coli, THF dUMP Inhibition of
Shigella spp., Salmonella spp. and Chlamydia spp. The T3SS is a syringe- Inhibition of cell wall synthesis
folate synthesis
like apparatus that facilitates the injection of bacterial effectors from these Depolarization of
species directly into the host cytosol (Fig. 3a)17. Depending on the spe- membrane potential

cies, T3SS effectors have been implicated in perturbing a variety of host K+

cellular processes, including cytoskeletal dynamics, gene expression, cell


cycle progression and apoptotic cell death programs. K+ K+
Chemical screens for inhibitors of the T3SS in Yersinia pseudotuber- K+
culosis identified acylated hydrazones of different salicylaldehydes18,19.
One compound was found to directly target the Y. pseudotuberculosis Figure 2 Traditional targets of antibacterial compounds. Traditional
T3SS, thereby preventing effector molecule translocation and attenuat- antibiotics function by inhibiting DNA or RNA synthesis (for
ing the pathogen in a cell culture model of infection without interfering example, fluoroquinolones), inhibiting protein synthesis (for example,
with in vitro growth19. Compounds of this class have also been found to aminoglycosides), inhibiting cell wall synthesis (for example, β-lactams),
inhibit intracellular replication and infectivity, and translocation of the inhibiting folate synthesis (for example, sulfa drugs), or depolarizing
T3SS effectors IncG and IncA during early and middle stages of infec- membrane potential (daptomycin).
tion of Chlamydia trachomatis (Fig. 3c)20. They have also been shown to
interfere with the life cycle of Chlamydia pneumoniae21. Very recently,
acylated hydrazones of different salicyclaldehydes have also been shown expands until a critical threshold concentration is reached. At critical
to be effective at inhibiting both the secretion of T3SS effectors and the threshold concentrations (when the bacteria are ‘quorate’), quorum
invasion of Salmonella enterica serovar Typhimurium into cultured epi- sensing signaling molecules can influence the expression of a variety of
thelial cells22. However, compounds of this class were only able to sup- different genes, including the expression of genes involved in virulence.
press T3SS-dependent secretory and inflammatory responses in a bovine Teleologically, the bacteria may begin to express virulence factors only
intestinal loop model of S. enterica serovar Typhimurium infection if when they have reached a significantly large population size to cause an
the bacteria were first preincubated with the compounds. Simultaneous ‘effective’ infection. Thus, by interfering with quorum sensing pathways,
infection and administration of these compounds was ineffective in one could inhibit a bacterial population’s ability to monitor the number
preventing fluid accumulation and neutrophil influx into the infected of cells within that population and thereby interfere with quorum sensing
intestinal loops22. Thus, while it seems that acylated hydrazones of dif- activation of virulence factor expression.
ferent salicyaldehydes may effectively target the T3SS of many different In many Gram-negative bacteria, quorum sensing is mediated by acyl-
pathogens, more work will need to be done to evaluate whether this class homoserine lactone molecules (AHLs) that are synthesized and recog-
of compounds has broad-spectrum therapeutic value. nized by quorum sensing circuits composed of LuxI and LuxR homologs
(Fig. 3a)24. LuxI homologs synthesize AHL molecules from the common
Targeting the regulation of virulence expression metabolic intermediate S-adenosylmethionine (SAM) and an acyl-acyl
In addition to targeting toxin function or delivery, disrupting the regula- carrier protein. At critical threshold concentrations, AHL molecules
tory mechanisms that result in virulence expression is attractive because are recognized by their cognate transcriptional activator (LuxR homo-
it would prevent the formation of toxin. To some degree, the use of the logs), which in turn regulates the transcription of genes associated with
protein synthesis inhibitor clindamycin, in addition to a cell wall anti- virulence. Thus, one could inhibit AHL-mediated quorum sensing by
biotic such as penicillin, in the treatment of streptococcal toxic shock inhibiting the enzymes that synthesize quorum sensing molecules (LuxI
syndrome to prevent the production of toxin (a phenomenon known homologs). In vitro, the activity of the P. aeruginosa LuxI homolog RhlI
as the “Eagle effect”) is a clinical application of this principle23. Recent can be inhibited with analogs of SAM such as S-adenosylhomocysteine,
advances in our understanding of virulence regulation have identified sinefungin and S-adenosylcysteine25, resulting in the inability to synthe-
many different regulatory steps that could be targeted. size quorum sensing molecules. Though SAM is a common metabolic
Most efforts to inhibit the regulation of virulence factor expression intermediate, it is possible that improved analogs of SAM could be gener-
have focused on interfering with quorum sensing (Fig. 3a), a mode of ated that could interfere with LuxI-mediated AHL synthesis but not host
bacterial communication used by multiple bacterial species to regulate metabolic processes that rely on SAM.
processes such as bioluminescence, antibiotic synthesis, biofilm forma- Alternatively, one could inhibit quorum sensing by interfering with
tion and virulence factor expression as a function of population den- the concentration of the AHL signaling molecules through degradation.
sity24. Quorum sensing is a phenomenon whereby bacteria can sense For example, Gram-positive Bacillus species produce acylhomoserine
their population density by releasing diffusible signaling molecules that lactonase, an enzyme that hydrolyzes the lactone ring of AHLs, thereby
accumulate in the population and surrounding environment. These sig- rendering them unable to mediate signaling26,27. Tobacco plants engi-
naling molecules increase in concentration as the bacterial population neered to express AHL-lactonase show an enhanced resistance to Erwinia

NATURE CHEMICAL BIOLOGY VOLUME 3 NUMBER 9 SEPTEMBER 2007 543


REVIEW

Table 1 Targets of antivirulence inhibitors, their modes of action and their effects on infection in vivo
Effect on in vivo (or ex vivo)
Targets Inhibitors (examples) Modes of action in vitro infection References
Toxin function
B. anthracis lethal factor (LF) (2R)-2-[(4-fluoro-3-methylphenyl) Binds LF active site and inhibits Protects mice from spore infection 9
sulfonylamino]-N-hydroxy-2- LF protease activity when used in combination with
(tetrahydro-2H-pyran-4-yl) ciprofloxacin; provides complete
acetamide (LFI) protection in mice immunized with
LF and PA
Peptide analogs derived from Inhibit LF cleavage of MKKs Protect macrophages from 10
the optimal peptide substrate in vitro LF-induced cytolysis ex vivo
sequence
B. anthracis protective antigen (PA) Cisplatin Inhibits LF translocation into the Protective when administered 14
host cytosol simultaneously with anthrax lethal
toxin in murine models
Hexa-D-arginine Inhibits PA processing Delays lethal toxin-induced toxemia 62
in rodent models and cultured
macrophages
Toxin delivery
Type III secretion system (T3SS) Acylated hydrazones of different Prevent effector molecule Attenuate Y. pseudotuberculosis 18–22
salicylaldehydes (for example, translocation infection and inhibit intracellular
INP0400) replication of C. trachomatis and
C. pneumoniae ex vivo; preincubation
of compound with bacteria before
infection suppresses secretory and
inflammatory responses in a bovine
intestinal loop model of S. enterica
serovar Typhimurium infection
Regulators of virulence expression
V. cholerae ToxT Virstatin Prevents expression of the Protects infant mice from intestinal 44
toxin-coregulated pilus and colonization with V. cholerae
cholera toxin in V. cholerae
Quorum sensing: LuxR homologs Structural analogs of AHLs (for Accelerate turnover of LuxR Promote clearance of P. aeruginosa 30–32,
example, halogenated furanones) homologs; inhibit expression of from the lungs of mice in a 34–38
quorum sensing regulated genes; pulmonary infection model;
inhibit the production of increase the survival time of mice
carbapenem in E. carotovora and in a lethal P. aeruginosa lung
virulence factors in P. aeruginosa infection model
Quorum sensing: S. aureus AgrC Inhibitory autoinducing peptides Inhibits agr locus activation Administration of inhibitory AIP 41–43
(AIPs) to mice during S. aureus infection
inhibits abscess formation
Adhesion
Pilin chaperones (for example, PapD Pilicides: bicyclic 2-pyridones Inhibit pilus assembly Bicyclic 2-pyridones inhibit 47,48
in UPEC) and N-substituted amino acid adhesion of E. coli to bladder
derivatives carcinoma cells ex vivo

carotovora infection27, which demonstrates that degradation of the AHL nated furanones inhibit production of exotoxins, repress the expression
signal is a viable approach to attenuating infection with E. carotovora. of quorum sensing regulated genes, and increase the susceptibility of
Mammalian sera also show strong AHL lactonase activity that is similar P. aeruginosa biofilms to tobramycin in vitro36,37. In vivo, administration
to the activity of paraoxonases28, which themselves have lactonase activi- of synthetic halogenated furanones promotes clearance of P. aeruginosa
ty against AHLs in vitro29. Thus, interference with quorum sensing by from the lungs of infected mice37,38 and increases the survival time of
increasing degradation of AHL molecules may be a natural phenome- mice in a lethal P. aeruginosa lung infection model38. However, AHL
non used by multiple species. Though it has not been exploited in anti- molecules themselves as well as other structural variant lactones are base-
microbial discovery, it represents an alternate mechanism for targeting labile and are substrates for mammalian paraoxonases28,29,39; they are
virulence. thus likely to be suboptimal for therapeutic use. Halogenated furanones
Finally, one could inhibit AHL-mediated quorum sensing by interfer- are also too reactive to be used therapeutically. For this reason, the recent
ing with the recognition of and subsequent transcriptional response to identification of a triphenyl compound that is a potent agonist of quorum
the signaling molecules by LuxR homologs. For example, a number of sensing in P. aeruginosa is promising in that this compound is structurally
structural analogs of AHLs have been shown to inhibit the expression of unrelated to AHL molecules and therefore may be useful as a scaffold for
quorum sensing regulated genes and virulence factor production as well developing future quorum sensing inhibitors that are more stable and
as biofilm formation in P. aeruginosa30–32. In particular, one class of struc- perhaps less reactive in vivo40.
tural analogs of AHLs, halogenated furanones, have been shown to have Quorum sensing is not a phenomenon that is unique to Gram-nega-
quorum sensing inhibitory properties33 and seem to function by accele- tive pathogens. Staphylococcus aureus strains make autoinducing peptides
rating the turnover of LuxR homologs34. In E. carotovora, halogenated (AIP) that function as quorum sensing molecules in these bacteria. In gene-
furanones have been shown to inhibit the production of carbapenem, ral, Gram-positive bacteria synthesize a precursor protein that is proteo-
which is regulated by the LuxR homolog CarR35. In P. aeruginosa, haloge- lytically cleaved to form the processed quorum sensing signaling peptide.

544 VOLUME 3 NUMBER 9 SEPTEMBER 2007 NATURE CHEMICAL BIOLOGY


REVIEW

a Host cell
Figure 3 Bacterial protein functions that can be targeted to inhibit virulence
and examples of virulence inhibitors. (a) Virulence inhibitors could target:
Endosome toxin function (for example, B. anthracis LF catalytic activity or translocation
Inhibition of through PA); toxin delivery, by inhibiting various bacterial systems such as
toxin function LF
type II or type III secretion (T3SS); virulence gene regulation (for example,
PA AHL-mediated quorum sensing circuitry (LuxI or LuxR homologs) or
Inhibition of transcriptional regulators that control virulence gene expression); or bacterial
bacterial adhesion LF adhesion to host cells (for example, inhibition of the formation of pili by
pilicides). (b–e) Examples of virulence factor inhibitors include: B. anthracis
lethal factor inhibitor9 (b), the T3SS inhibitor INP0400 (ref. 20) (c), the
quorum sensing inhibitor furanone C-30 (ref. 37) (d), and a pilicide (a
Inhibition of bicyclic 2-pyridone compound)48 (e).
toxin delivery

AHL T3SS
AHL Apart from quorum sensing, transcriptional regulators that coordinate
AHL the expression of genes involved in adhesion, toxin production and secre-
AHL
Type II secretion
tion are themselves potential targets for future antimicrobials. A recent
AHL LuxI example is the small molecule virstatin (Fig. 3d), which was identified
AHL
in a screen for small-molecule inhibitors of the V. cholerae cholera toxin
LuxR
promoter44. Virstatin inhibits the transcriptional regulator ToxT, thereby
preventing the expression of two critical virulence factors in V. cholerae:
Inhibition of virulence regulation the toxin-coregulated pilus, which is involved in attachment to the intes-
tinal epithelium, and cholera toxin. In vivo, administration of virstatin
Virulence genes protected infant mice from intestinal colonization with V. cholerae44. This
Bacterial cell work also demonstrated that late administration of virstatin (12 h after
infection) still results in a 3-log drop in the ability to recover bacteria from
the mouse intestine, which shows that inhibition of virulence expression,
even in established infection, can still have potential therapeutic value.
Another example of inhibition of virulence regulation and expression
b O c was found in a screen for inhibitors of the T3SS in enteropathogenic
HO
E. coli (EPEC)45. In this study, the imine adduct from the condensation
O of a halogenated salicylaldehyde and 3-aminoacetophenone seemed to
HN OH
O N decrease the transcription of T3SS effectors and components of the T3SS
S NH N
O H machinery without affecting growth or motility. Thus, toxin delivery can
O
Cl be inhibited by novel therapeutics targeting not only structural compo-
nents of the secretion apparatus but also transcriptional regulators of
F secretory systems.

Inhibition of adhesion
d e Equally important for establishing infection is adhesion of the bacte-
COOH rial cell to the host. The human body has evolved multiple lines of
defense to prevent the majority of bacteria from adhering to host tis-
O N O S
sues, including continual shedding of epithelial cell surfaces, mucus
N lining epithelial cell surfaces of the respiratory, intestinal and repro-
Li ductive tracts, and direct inhibition of adhesion of bacteria to host
N O O
O surfaces by antibody molecules that line these tracts and perfuse host
tissues. These multiple lines of defense erected by the body under-
score the importance of preventing bacterial adhesion to the host.
This peptide is then actively transported extracellularly and can then Moreover, because proteins involved in bacterial adhesion are specific
be recognized by a histidine-sensor kinase protein of a two-component to prokaryotes, like bacterial secretion systems, they are rational tar-
regulatory cascade. Upon recognition of the peptide autoinducer, the gets for novel antimicrobials.
two-component regulator activates a phosphorelay response, ultimately A potentially broad class of antimicrobials that target adhesion are
resulting in activation of a quorum sensing transcriptional regulator. called ‘pilicides’. Pilicides are aimed at inhibiting the formation of pili
Different S. aureus strains produce peptides whose sequences vary slightly or fimbriae (Fig. 3a), which are hair-like projections protruding from
from those produced by other strains. Thus, a given peptide sequence can bacterial cells that facilitate adhesion. Pili consist of repeating subunits
activate its own histidine kinase two-component regulatory cascade while of immunoglobulin-like domains wherein the N terminus of one sub-
antagonizing the histidine kinase two-component regulatory cascades in unit is donated to complete the immunoglobulin fold of its neighbor
other strains of S. aureus41. This has been exploited in a mouse model subunit; these structures are formed by donor-strand complementation
of S. aureus–mediated abscess formation, in which administration of via the chaperone usher pathway46. In this pathway, the chaperone pro-
inhibitory AIP to mice during the first 3 h of S. aureus infection inhibited tein binds pilin subunits by donating its edge β-strand to complete the
the pathology42,43. Clearly, such a therapeutic approach would require folding of the pilin subunit’s immunoglobulin fold. Chaperone–pilin
a small molecule that could inhibit the quorum sensing systems of all complexes then traffic to outer-membrane usher channels where the
S. aureus strains. pilin fiber is formed by donor strand exchange between the chaperone

NATURE CHEMICAL BIOLOGY VOLUME 3 NUMBER 9 SEPTEMBER 2007 545


REVIEW

and the adjacent subunit in the growing pilin fiber. So far, efforts at population toward conventional antibiotics, they would not alter the
developing pilicides have focused on inhibiting the periplasmic chape- natural microbiome of the host, and they may work optimally to pre-
rone proteins of the chaperone usher pathway in uropathogenic E. coli vent infection, as evidenced by cases in which the drugs work best when
(UPEC). Bicyclic 2-pyridones (Fig. 3e) and N-substituted amino acid administered with the pathogen rather than in established infection. It is
derivatives have been shown to competitively inhibit binding of chaper- likely that the unique mechanism of action for each antivirulence thera-
ones to pilin subunits by surface plasmon resonance47. In vitro, bicyclic peutic will ultimately determine its utility as a prophylactic, mono- or
2-pyridones have also been shown to inhibit hemagglutination and combination therapeutic agent.
biofilm formation in laboratory and clinical E. coli strains, and ex vivo Because virulence factors are required at different times during infec-
they have been shown to inhibit adhesion of the bacteria to bladder tion, the definition of the time window in which a given inhibitor will
carcinoma cells by ~90%48. It has been suggested that pilicides may have be effective will likely be mechanism- and thus inhibitor-specific. For
broad-spectrum activity due to the conservation of both chaperone example, as described above, in V. cholerae infection, inhibition of the
structure and the chaperone-usher pathway49. transcriptional regulator ToxT, which results in the downstream inhibi-
tion of cholera toxin synthesis, has therapeutic efficacy in established
Virulence inhibitors as therapeutic candidates infant mouse colonization 12 h after infection44. On the other hand,
Novel therapeutics that target virulence rather than simply in vitro cell cisplatin inhibition of LF translocation into the host cytosol by blocking
growth would both supplement and add diversity to our current antimi- PA heptamer assembly is completely ineffective in mice if the inhibitor is
crobial armamentarium, with the caveat that this type of strategy may not administered after anthrax toxin inoculation14. In contrast, if the mecha-
be effective in immunocompromised individuals who lack the ability to nism of protection is due to inhibition of the proteolytic activity of LF,
clear the ‘disarmed’ pathogen. However, for the appropriate patient popu- delayed administration of LFI is effective (granted, in combination with
lation, a tantalizing advantage of targeting virulence is the potential of ciprofloxacin) in rescuing infected mice9. Thus, the therapeutic efficacy
this approach to impose weaker selective pressure that would be less likely of inhibiting virulence factors needs to be evaluated on an individualized,
to foster the growth of antibiotic resistance, in contrast to therapeutics mechanistic basis, and efforts will need to be invested in determining
targeted at straightforward viability. If the function of a given virulence the in vivo time window in which a protein involved in virulence may be
factor is unrelated to its viability within the host, there should be no in effectively targeted. This analysis would provide insight into the timing
vivo selection for the rare, resistant mutant to an inhibitor of that factor. of different virulence programs and their requirement throughout the
The difficulty is that it is unclear which virulence factors fit this criterion, course of infection.
and thus each individual case will need to be examined carefully. Though some virulence inhibitors such as pilicides and T3SS inhibitors
Even if an inhibitor targets a virulence factor required for in vivo via- have the potential to target a wider spectrum of bacteria, other virulence
bility, the circumstances under which resistance could arise against the inhibitors that disrupt mechanisms specific only to single pathogens or
specific inhibitor relative to conventional antibiotics are more limited, that target bacterial proteins that do not have structurally similar ortho-
because antivirulence inhibitors are often pathogen-specific and selec- logs in many clinically relevant bacterial species will be exquisitely nar-
tion must occur in vivo. Thus, the window of opportunity during which row-spectrum by definition. Although this new paradigm could be the
resistance could be selected for is more narrow, equaling the length of solution to the resistance crisis we currently face, it raises two problems
an individual infectious cycle. Additionally, use of antivirulence drugs that are not insurmountable but that need to be addressed. One problem
could also have an impact on the development of resistance to our cur- is diagnostic; the other is economic.
rent broad-spectrum antibiotics by offering an alternative therapy in The utility of therapeutic intervention with narrow-spectrum anti-
certain cases, thus reserving our current broad-spectrum agents to a more virulence antimicrobials will be very dependent on the clinician’s ability
limited set of cases in which they are most needed. Finally, because of to precisely diagnose the genus if not the species of bacterial infection in
their narrow spectrum and different mechanisms of action, therapeutics a patient in order to select the correct antimicrobial therapeutic agent.
that act to inhibit virulence rather than cell growth also have a signifi- Thus, antimicrobials that target virulence will need to be developed hand
cant advantage in helping to preserve normal and potentially beneficial in glove with new diagnostic tests that will allow the clinician real-time
members of the normal human microbiota. This type of strategy would diagnosis. Certainly these technological problems are challenging, but
likely have a significant impact on the morbidity associated with shifts they are currently the focus of great interest, with efforts intensified par-
in host normal flora after antibiotic treatment. ticularly in the area of biodefense. In addition to real-time identification
The exact therapeutic role of antimicrobials that target virulence is as of the culprit pathogen, a related issue is determining the susceptibility of
yet unclear. Antivirulence therapies have the potential to function effec- the culprit pathogen to virulence inhibitors. Definition of conventional
tively when used alone, when used in combination therapy with antibi- antibiotic susceptibilities has become a well-tuned science that includes
otics, or simply as a prophylactic treatment. It is premature to rule out Kirby-Bauer (disc diffusion) tests and broth dilution measurements. In
the use of antivirulence drugs simply because they are not bacteriocidal, contrast, determining susceptibilities to a virulence inhibitor cannot
as this would underestimate the role of host immunity. A long-standing use simple cell growth as a sensitivity indicator. Assays exist and can be
dogma suggests a preference for bacteriocidal over bacteriostatic anti- developed for determining susceptibility in vitro, such as the screening
biotics. However, there is actually little clinical evidence to suggest that assays used to identify these inhibitors or ones engineered to include
this dogma has significant bearing on the resolution of infection, with various gene reporter readouts. However, these assays too will need to
other variables such as tissue penetration and bioavailability potentially be individualized for each specific inhibitor.
having a greater impact. Thus, a new paradigm for antimicrobial therapy Perhaps the greatest challenge of changing to a new paradigm is not
redefines the goal to be simply the tipping of the balance in favor of the technological but economic. If compounds that target virulence are effec-
host, thus enabling it to control infection, rather than complete in vivo tive against a much more narrow range of bacteria, the economic incen-
killing of a pathogen by the drug itself. tive for a pharmaceutical company to develop that compound into a drug,
Antivirulence drugs could have a role in prophylactic treatment during with the accompanying diagnostic and susceptibility tools, is even lower
an epidemic, a bioterrorist threat, or in select populations (for example, than current economic incentives to develop wide-spectrum antibiotics.
travelers) for several reasons. They would not engender resistance in the If we are to avoid plummeting into a postantibiotic era in the near future,

546 VOLUME 3 NUMBER 9 SEPTEMBER 2007 NATURE CHEMICAL BIOLOGY


REVIEW

clearly we must attack not only the technological hurdles facing new Inhibitors of in vivo essential genes as therapeutics
antimicrobial development but also the economic hurdles. Inhibitors of in vivo essential gene functions can target (i) functions
required for viability in vitro (including conventional antibiotic tar-
Future approaches to new antimicrobial development gets), (ii) functions required for viability only in the host, (iii) functions
To supplement our dwindling antimicrobial arsenal, a broader range of required for virulence or (iv) some combination of the above. The advan-
targets than the set of in vitro essential genes that are typically targeted tages and disadvantages of a therapeutic will vary based on the functional
by conventional antibiotics must be considered. We propose that a new class or classes to which the target belongs. Advantages and disadvantages
paradigm for antimicrobial development should be based on targeting are manifested in the spectrum of activity against different pathogens,
gene functions that are required in vivo to cause disease. This strategy the window of time in which a drug is effective, and the level of selective
includes targeting genes that are essential for causing virulence as well pressure for resistance.
as those essential for in vivo viability. As the ability of pathogenic bac- Therapeutics that target gene functions essential for viability both in
teria to both cause damage and survive within the host is required for vivo and in vitro (that is, conventional antibiotics) often target proteins
causing disease, disrupting either of these processes could be exploited that are well conserved among species. Thus, this functional class has
therapeutically. the potential to be broad-spectrum, and the time window during which
In vitro and in vivo bacterial essential gene functions are distinct. The it should be effective is relatively large. The disadvantage, of course,
growth environment within a host is unlikely to be the same as the arti- is that this class of therapeutics can impose strong selective pressure
ficial ones induced in a laboratory, and therefore the genes required for that fosters the growth of antibiotic resistance. At the other end of
viability will likely also differ. This concept was illustrated in a trans- the extreme are therapeutics that target gene functions required for
poson site hybridization study conducted in Mycobacterium tuberculo- virulence. The advantages and disadvantages of this functional class
sis–infected mice. The study revealed that a different set of M. tuberculosis have been discussed above, and they include the imposition of less
genes, representing ~5% of the M. tuberculosis genome, were required selective pressure and a narrower window of efficacy and spectrum of
for in vivo survival than were required for in vitro growth50. Current activity. Therapeutics that target in vivo essential gene functions will
antibiotic discovery approaches have focused only on in vitro essentials, have advantages and disadvantages that fall between these two extremes.
and thus the realm of in vivo essentials that could be targeted has not yet They should exert some selective pressure, but only within the context
been sufficiently explored. of the host, and they may have a larger efficacy window and (poten-
One approach that has the capacity to identify new therapeutics that tially) a broader spectrum of activity compared to antivirulence drugs.
target either bacterial in vivo essential gene functions or the host itself is Therapeutics that fall into a combination of these functional classes will
chemical screening of whole-organism infection models. Whole organ- have even more complex profiles.
ism–based screening has the advantage of being able to identify small
molecules that are more likely to be permeable to the cell, effective at elici- Conclusion
ting the desired phenotype (such as attenuation of infection), unlikely This review highlights a number of novel strategies for developing new
to have gross toxic side effects, and have acceptable pharmacokinetic therapeutics against infection. There have been very few new classes of
profiles, at least in the model host, which may or may not translate from antibiotics discovered in the past 40 years, with efforts dwindling in large
host to host51. Finally, whole organism–based screening has the capac- pharmaceutical companies. The effort that has been invested has been
ity to define which proteins are, in fact, ‘drug-targetable’. Thus, whole- disappointing, resulting in a rather grim picture for the current state of
organism screening has the potential to leapfrog over some of the major antibiotic development. GlaxoSmithKline recently undertook a genomic,
hurdles commonly encountered after typical in vitro target-based screens. target and whole cell–based approach over a period of seven years to iden-
Practically, of course, in order to identify candidate novel antimicrobials tify compounds that inhibit genes thought to be essential for viability in
in a whole-organism infection model, the model host must be of a size a number of pathogens59. Disappointingly, only five leads were identified
suitable for chemical screening. The zebrafish (Danio rerio) has success- from a total of 70 high-throughput screens using the GSK compound col-
fully been used in high-throughput chemical screens52,53 and has also lection, which suggests that in vitro target-based screening of traditional
been used to model mycobacterial, streptococcal and Salmonella infec- compound libraries biased to follow Lipinski’s ‘rule of five’60 is an inef-
tions54–56. Thus, the marriage of chemical screening to zebrafish infection fective way to identify new antimicrobials. However, GlaxoSmithKline’s
models may be a new way of rapidly identifying compounds that target experience does suggest that future efforts at antimicrobial identification
in vivo essential gene functions in a vertebrate model host57. will need to include screening of libraries that sample a wider diversity
The utility of whole organism–based chemical screening for new anti- of chemical space with potentially differing physicochemical properties
microbial candidates has already been demonstrated using Caenorhabditis (possibly diversity oriented synthesis libraries61) and screening of natural
elegans as the model host. In this study, Moy and colleagues screened 6,000 products, which as yet have not been exhaustively mined for new antimi-
synthetic compounds and 1,136 natural product extracts for compounds crobials. In addition, the GlaxoSmithKline experience also suggests that
that promote nematode survival of Enterococcus fecalis infection58. They it is easier to find the target of a given lead compound than to engineer a
identified several compounds that rescue nematodes from the lethality of lead compound to have greater permeability; thus identification of new
infection at therapeutic concentrations that are well below the minimal antimicrobials is likely to be more fruitful in whole cell–based screens
inhibitory concentrations (MICs) of the compounds against E. fecalis. rather than in in vitro target-based screens. We suggest that this approach
Unless one posits that C. elegans is able to concentrate the compounds should be expanded to include whole-organism screening, which has the
above the therapeutic index, one must assume that these compounds are advantage that one can identify small molecules that target either the host
rescuing the nematodes either by targeting an E. fecalis in vivo essential or in vivo essential gene functions of the pathogen. Given the dearth of
gene function, inhibiting virulence, or targeting the nematode immune antimicrobials with novel modes of action against Gram-negative hospi-
defense. Though the targets of these compounds have not yet been identi- tal pathogens currently in phase 1 clinical trials, it is estimated that it may
fied, and though it is unclear as yet whether these compounds target the be 10 to 15 years before antimicrobials against this class of pathogen are
worm or the bacterium, this method highlights a novel approach for iden- available for therapeutic use59. We believe that this makes the argument
tifying compounds that target proteins essential for virulence in vivo. for pursuing therapeutics that target in vivo essential gene functions all

NATURE CHEMICAL BIOLOGY VOLUME 3 NUMBER 9 SEPTEMBER 2007 547


REVIEW

the more compelling, as antibiotic resistance continues to evolve and the 29. Draganov, D.I. et al. Human paraoxonases (PON1, PON2, and PON3) are lactonases with
overlapping and distinct substrate specificities. J. Lipid Res. 46, 1239–1247 (2005).
need for new antimicrobials continues to grow. 30. Muh, U. et al. Novel Pseudomonas aeruginosa quorum-sensing inhibitors identified
in an ultra-high-throughput screen. Antimicrob. Agents Chemother. 50, 3674–3679
ACKNOWLEDGMENTS (2006).
We thank E.J. Rubin, J.E. Gomez and S.A. Stanley for helpful discussions and critical 31. Geske, G.D., Wezeman, R.J., Siegel, A.P. & Blackwell, H.E. Small molecule inhibitors of
reading of this manuscript and J.S.W. Lee for assistance in preparing figures. bacterial quorum sensing and biofilm formation. J. Am. Chem. Soc. 127, 12762–12763
(2005).
COMPETING INTERESTS STATEMENT 32. Smith, K.M., Bu, Y. & Suga, H. Induction and inhibition of Pseudomonas aeruginosa
The authors declare no competing financial interests. quorum sensing by synthtic autoinducer analogs. Chem. Biol. 10, 81–89 (2003).
33. Givskov, M. et al. Eukaryotic interference with homoserine lactone-mediated prokaryotic
Published online at http://www.nature.com/naturechemicalbiology signalling. J. Bacteriol. 178, 6618–6622 (1996).
34. Manefield, M. et al. Halogenated furanones inhibit quorum sensing through accelerated
Reprints and permissions information is available online at http://npg.nature.com/
LuxR turnover. Microbiology 148, 1119–1127 (2002).
reprintsandpermissions
35. Manefield, M., Welch, M., Givskov, M., Salmond, G.P. & Kjelleberg, S. Halogenated
furanones from the red alga, Delisea pulchra, inhibit carbapenem antibiotic synthesis
1. Palumbi, S.R. Humans as the world’s greatest evolutionary force. Science 293, 1786–
and exoenzyme virulence factor production in the phytopathogen Erwinia carotovora.
1790 (2001).
FEMS Microbiol. Lett. 205, 131–138 (2001).
2. Young, J.A. & Collier, R.J. Anthrax toxin: receptor-binding, internalization, pore formation,
36. Hentzer, M. et al. Inhibition of quorum sensing in Pseudomonas aeruginosa biofilm
and translocation. Annu. Rev. Biochem. 76, 243–265 (2007).
bacteria by a halogenated furanone compound. Microbiology 148, 87–102 (2002).
3. Boyden, E.D. & Dietrich, W.F. Nalp1b controls mouse macrophage susceptibility to
37. Hentzer, M. et al. Attenuation of Pseudomonas aeruginosa virulence by quorum sensing
anthrax lethal toxin. Nat. Genet. 38, 240–244 (2006).
inhibitors. EMBO J. 22, 3803–3815 (2003).
4. Duesbery, N.S. et al. Proteolytic inactivation of MAP-kinase-kinase by anthrax lethal
38. Wu, H. et al. Synthetic furanones inhibit quorum-sensing and enhance bacterial clear-
factor. Science 280, 734–737 (1998).
ance in Pseudomonas aeruginosa lung infection in mice. J. Antimicrob. Chemother. 53,
5. Vitale, G. et al. Anthrax lethal factor cleaves the N-terminus of MAPKKs and induces tyro-
1054–1061 (2004).
sine/threonine phosphorylation of MAPKs in cultured macrophages. Biochem. Biophys.
39. Yates, E.A. et al. N-acylhomoserine lactones undergo lactonolysis in a pH-, temperature-,
Res. Commun. 248, 706–711 (1998).
and acyl chain length-dependent manner during growth of Yersinia pseudotuberculosis
6. Panchal, R.G. et al. Chemical genetic screening identifies critical pathways in anthrax
and Pseudomonas aeruginosa. Infect. Immun. 70, 5635–5646 (2002).
lethal toxin-induced pathogenesis. Chem. Biol. 14, 245–255 (2007).
40. Muh, U. et al. A structurally unrelated mimic of a Pseudomonas aeruginosa acyl-homo-
7. During, R.L. et al. Anthrax lethal toxin paralyzes actin-based motility by blocking Hsp27
serine lactone quorum-sensing signal. Proc. Natl. Acad. Sci. USA 103, 16948–16952
phosphorylation. EMBO J. 26, 2240–2250 (2007).
(2006).
8. Rainey, G.J. & Young, J.A. Antitoxins: novel strategies to target agents of bioterrorism.
41. Lyon, G.J., Wright, J.S., Christopoulos, A., Novick, R.P. & Muir, T.W. Reversible and
Nat. Rev. Microbiol. 2, 721–726 (2004).
specific extracellular antagonism of receptor-histidine kinase signaling. J. Biol. Chem.
9. Shoop, W.L. et al. Anthrax lethal factor inhibition. Proc. Natl. Acad. Sci. USA 102,
277, 6247–6253 (2002).
7958–7963 (2005).
42. Mayville, P. et al. Structure-activity analysis of synthetic autoinducing thiolactone pep-
10. Turk, B.E. et al. The structural basis for substrate and inhibitor selectivity of the anthrax
tides from Staphylococcus aureus responsible for virulence. Proc. Natl. Acad. Sci. USA
lethal factor. Nat. Struct. Mol. Biol. 11, 60–66 (2004).
96, 1218–1223 (1999).
11. Krantz, B.A. et al. A phenylalanine clamp catalyzes protein translocation through the
anthrax toxin pore. Science 309, 777–781 (2005). 43. Wright, J.S. III, Jin, R. & Novick, R.P. Transient interference with staphylococcal quo-
12. Artenstein, A.W. et al. Chloroquine enhances survival in Bacillus anthracis intoxication. rum sensing blocks abscess formation. Proc. Natl. Acad. Sci. USA 102, 1691–1696
J. Infect. Dis. 190, 1655–1660 (2004). (2005).
13. Sanchez, A.M. et al. Amiodarone and bepridil inhibit anthrax toxin entry into host cells. 44. Hung, D.T., Shakhnovich, E.A., Pierson, E. & Mekalanos, J.J. Small-molecule inhibitor of
Antimicrob. Agents Chemother 51, 2403–2411 (2007). Vibrio cholerae virulence and intestinal colonization. Science 310, 670–674 (2005).
14. Moayeri, M., Wiggins, J.F., Lindeman, R.E. & Leppla, S.H. Cisplatin inhibition of anthrax 45. Gauthier, A. et al. Transcriptional inhibitor of virulence factors in enteropathogenic
lethal toxin. Antimicrob. Agents Chemother. 50, 2658–2665 (2006). Escherichia coli. Antimicrob. Agents Chemother. 49, 4101–4109 (2005).
15. Ma, T. et al. Thiazolidinone CFTR inhibitor identified by high-throughput screening 46. Sauer, F.G. et al. Chaperone-assisted pilus assembly and bacterial attachment. Curr.
blocks cholera toxin-induced intestinal fluid secretion. J. Clin. Invest. 110, 1651–1658 Opin. Struct. Biol. 10, 548–556 (2000).
(2002). 47. Svensson, A. et al. Design and evaluation of pilicides: potential novel antibacterial agents
16. King, C.Y. & Barriere, S.L. Analysis of the in vitro interaction between vancomycin and directed against uropathogenic Escherichia coli. ChemBioChem 2, 915–918 (2001).
cholestyramine. Antimicrob. Agents Chemother. 19, 326–327 (1981). 48. Pinkner, J.S. et al. Rationally designed small compounds inhibit pilus biogenesis in
17. Galan, J.E. & Wolf-Watz, H. Protein delivery into eukaryotic cells by type III secretion uropathogenic bacteria. Proc. Natl. Acad. Sci. USA 103, 17897–17902 (2006).
machines. Nature 444, 567–573 (2006). 49. Lee, Y.M., Almqvist, F. & Hultgren, S.J. Targeting virulence for antimicrobial chemo-
18. Kauppi, A.M., Nordfelth, R., Uvell, H., Wolf-Watz, H. & Elofsson, M. Targeting bacte- therapy. Curr. Opin. Pharmacol. 3, 513–519 (2003).
rial virulence: inhibitors of type III secretion in Yersinia. Chem. Biol. 10, 241–249 50. Sassetti, C.M. & Rubin, E.J. Genetic requirements for mycobacterial survival during
(2003). infection. Proc. Natl. Acad. Sci. USA 100, 12989–12994 (2003).
19. Nordfelth, R., Kauppi, A.M., Norberg, H.A., Wolf-Watz, H. & Elofsson, M. Small-mol- 51. MacRae, C.A. & Peterson, R.T. Zebrafish-based small molecule discovery. Chem. Biol.
ecule inhibitors specifically targeting type III secretion. Infect. Immun. 73, 3104–3114 10, 901–908 (2003).
(2005). 52. Peterson, R.T. et al. Chemical suppression of a genetic mutation in a zebrafish model of
20. Muschiol, S. et al. A small-molecule inhibitor of type III secretion inhibits different aortic coarctation. Nat. Biotechnol. 22, 595–599 (2004).
stages of the infectious cycle of Chlamydia trachomatis. Proc. Natl. Acad. Sci. USA 103, 53. Stern, H.M. et al. Small molecules that delay S phase suppress a zebrafish bmyb mutant.
14566–14571 (2006). Nat. Chem. Biol. 1, 366–370 (2005).
21. Bailey, L. et al. Small molecule inhibitors of type III secretion in Yersinia block the 54. Davis, J.M. et al. Real-time visualization of Mycobacterium-macrophage interactions lead-
Chlamydia pneumoniae infection cycle. FEBS Lett. 581, 587–595 (2007). ing to initiation of granuloma formation in zebrafish embryos. Immunity 17, 693–702
22. Hudson, D.L. et al. Inhibition of type III secretion in Salmonella enterica serovar (2002).
Typhimurium by small-molecule inhibitors. Antimicrob. Agents Chemother 51, 2631– 55. Neely, M.N., Pfeifer, J.D. & Caparon, M. Streptococcus-zebrafish model of bacterial
2635 (2007). pathogenesis. Infect. Immun. 70, 3904–3914 (2002).
23. Stevens, D.L., Gibbons, A.E., Bergstrom, R. & Winn, V. The Eagle effect revisited: efficacy 56. van der Sar, A.M. et al. Zebrafish embryos as a model host for the real time analysis of
of clindamycin, erythromycin, and penicillin in the treatment of streptococcal myositis. Salmonella typhimurium infections. Cell. Microbiol. 5, 601–611 (2003).
J. Infect. Dis. 158, 23–28 (1988). 57. Mukhopadhyay, A. & Peterson, R.T. Fishing for new antimicrobials. Curr. Opin. Chem.
24. Miller, M.B. & Bassler, B.L. Quorum sensing in bacteria. Annu. Rev. Microbiol. 55, Biol. 10, 327–333 (2006).
165–199 (2001). 58. Moy, T.I. et al. Identification of novel antimicrobials using a live-animal infection model.
25. Parsek, M.R., Val, D.L., Hanzelka, B.L., Cronan, J.E. Jr. & Greenberg, E.P. Acyl homo- Proc. Natl. Acad. Sci. USA 103, 10414–10419 (2006).
serine-lactone quorum-sensing signal generation. Proc. Natl. Acad. Sci. USA 96, 4360– 59. Payne, D.J., Gwynn, M.N., Holmes, D.J. & Pompliano, D.L. Drugs for bad bugs: confront-
4365 (1999). ing the challenges of antibacterial discovery. Nat. Rev. Drug Discov. 6, 29–40 (2007).
26. Dong, Y.H., Xu, J.L., Li, X.Z. & Zhang, L.H. AiiA, an enzyme that inactivates the acylhomo- 60. Lipinski, C.A., Lombardo, F., Dominy, B.W. & Feeney, P.J. Experimental and computa-
serine lactone quorum-sensing signal and attenuates the virulence of Erwinia carotovora. tional approaches to estimate solubility and permeability in drug discovery and develop-
Proc. Natl. Acad. Sci. USA 97, 3526–3531 (2000). ment settings. Adv. Drug Deliv. Rev. 46, 3–26 (2001).
27. Dong, Y.H. et al. Quenching quorum-sensing-dependent bacterial infection by an N-acyl 61. Schreiber, S.L. Target-oriented and diversity-oriented organic synthesis in drug discovery.
homoserine lactonase. Nature 411, 813–817 (2001). Science 287, 1964–1969 (2000).
28. Yang, F. et al. Quorum quenching enzyme activity is widely conserved in the sera of 62. Sarac, M.S., Peinado, J.R., Leppla, S.H. & Lindberg, I. Protection against anthrax toxemia
mammalian species. FEBS Lett. 579, 3713–3717 (2005). by hexa-D-arginine in vitro and in vivo. Infect. Immun. 72, 602–605 (2004).

548 VOLUME 3 NUMBER 9 SEPTEMBER 2007 NATURE CHEMICAL BIOLOGY

You might also like