Sie sind auf Seite 1von 8

Review TRENDS in Molecular Medicine Vol.13 No.

New strategies for combating


multidrug-resistant bacteria
Gerard D. Wright and Arlene D. Sutherland
Antimicrobial Research Centre, Department of Biochemistry and Biomedical Sciences, DeGroote School of Medicine,
McMaster University, 1200 Main St W, Hamilton, Ontario, L8N 3Z5, Canada

Antibiotic resistance is a problem that continues to does not produce new antibiotics for confronting the increas-
challenge the healthcare sector. In particular, multidrug ing problem of bacterial MDR, clinical options for treating
resistance is now common in familiar pathogens such as infections caused by these pathogens will be very limited.
Staphylococcus aureus and Mycobacterium tuberculosis, Combating MDR requires: (i) a detailed understanding
as well as emerging pathogens such as Acinetobacter of the molecular basis, evolution and dissemination of
baumannii. New antibiotics and new therapeutic strat- resistance; (ii) new chemicals with antibiotic properties
egies are needed to address this challenge. Advances in to fill the traditional antibiotic pipeline; and (iii) innovative
identifying new sources of antibiotic natural products and strategies that can extend the life of antibiotic molecules or
expanding antibiotic chemical diversity are providing that can uncover new approaches for controlling pathogen
chemical leads for new drugs. Inhibitors of resistance growth. Recent whole-genome sequence data for bacteria
mechanisms and microbial virulence are orthogonal strat- as well as exploration of the resistance burden in environ-
egies that are also generating new chemicals that can mental bacteria suggest resistance is much more wide-
extend the life of existing antibiotics. This new chemistry, spread than previously thought. Could MDR be the natural
coupled with a growing understanding of the mechan- state of microorganisms? If so, is resistance inevitable?
isms, origins and distribution of antibiotic resistance, Where will new antibiotics come from, and how can the
position us to tackle the challenges of antibiotic resist- emergence of resistance be countered? A renaissance in
ance in the 21st century. natural-product discovery, based on an expanding effort to
explore non-traditional sources of microbial biodiversity
The problem of antibiotic resistance and molecular tools that can exploit natural-product bio-
One of the foremost challenges in the management of synthesis, is revealing new chemical classes with the
infectious diseases is antimicrobial drug resistance. This potential to prime the antibiotic pipeline. Furthermore,
is an issue that impacts the treatment of all infections, non-traditional anti-infection targets, such as inhibitors of
whether they are caused by viruses, parasites, fungi or resistance mechanisms and microbial virulence, are emer-
bacteria. However, it is the manifestation of multidrug ging as alternative strategies. Recent progress in each of
resistance in bacteria over the past several decades that these areas will be discussed in this review.
is resulting in one of the most pressing clinical problems in
modern medicine: Where does drug resistance come from, Antibiotics and resistance
and how can we address it? The major mechanisms of antibiotic resistance include
Antibiotic resistance in bacteria is the product of innate enzymatic transformation, modification of the molecular
genetics and physiology, which are vertically transmitted target, sequestration of the drug, active efflux from the cell
through species, and the remarkable propensity of bacteria interior and, conversely, prevention of entry of the com-
to exchange genetic material horizontally across species and pound into the cell (reviewed in Refs [4–6]). Resistance
genera. This combinatorial genetic strategy has resulted in arises either passively as a result of pre-existing innate
the accumulation of multidrug-resistance (MDR) pheno- mechanisms or actively via the acquisition of new genetic
types in many species of bacteria. The Infectious Disease material by means of mobile genetic elements such as
Society of America has identified methicillin-resistant plasmids or transposons [7,8].
Staphylococcus aureus (MRSA), vancomycin-resistant Innate resistance includes physiological barriers such
Enterococcus faecium, extended-spectrum b-lactamase-pro- as the outer membrane of Gram-negative bacteria, the
ducing Enterobacteriaceae and MDR Acinetobacter bau- expression of intrinsic efflux pumps and natural mutations
mannii and Pseudomonas aeruginosa as bacterial in antibiotic targets. Some bacteria are replete with such
pathogens that are especially challenging for the manage- innate defense systems, for example, P. aeruginosa [9].
ment of infectious diseases [1]. This list will surely grow in This opportunistic pathogen is widely distributed in
the future; witness the recent emergence of extensively aquatic and soil environments and can express a stunning
drug-resistant tuberculosis (XDR-TB) [2] and MDR Clostri- variety of efficient efflux-pumps as well as antibiotic-inac-
dium difficile [3], for example. If the drug-discovery pipeline tivating enzymes. These have probably evolved as a means
for the organism’s survival in numerous environments,
Corresponding author: Wright, G.D. (wrightge@mcmaster.ca). each with its own antibacterial chemical challenges to
Available online 9 May 2007. overcome. When associated with infection, P. aeruginosa
www.sciencedirect.com 1471-4914/$ – see front matter ß 2007 Elsevier Ltd. All rights reserved. doi:10.1016/j.molmed.2007.04.004
Review TRENDS in Molecular Medicine Vol.13 No.6 261

is therefore very challenging to eradicate as a result of this through the SOS response [14]. Antibiotic resistance,
innate, combinatorial protection-system. therefore, is far reaching and highly adaptable, can emerge
By contrast, the often commensal organisms, such as rapidly and can progress through bacterial populations
Escherichia coli and S. aureus, that are commonly associ- vertically and horizontally with relative ease.
ated with infections in the community and hospitals
readily acquire antibiotic-resistance gene clusters through New antibiotics
resistance plasmids and other mobile genetic elements. One of the major problems facing the antibiotic-drug-
Such gene clusters frequently confer resistance to several discovery sector is the difficulty in identifying new
antibiotics at once, and as a result individual resistance drug-like compounds with suitable antibacterial activity.
genes can be maintained even without direct selection. Historically, the first successful antibacterial compounds
This fact can be an impediment to antibiotic-cycling strat- were synthetic in origin. These include salvarsan, which
egies for overcoming antibiotic resistance in health-care was identified at the turn of the last century for the
settings. Resistance gene cassettes are also associated with treatment of syphilis, and the sulfonamides that target
transposons and integrons [10]. The former enable the p-aminobenzoic acid biosynthesis. Despite the initial
mobilization of genes from plasmids into bacterial chromo- supremacy of synthetic antimicrobial drugs, it has been
somes and vice versa. Integrons, whether on the chromo- natural-product antibiotics, largely of bacterial origin, that
some or a plasmid, enable the continued accretion of have dominated as the preferred chemical scaffolds of
resistance genes resulting in an expansion of MDR [7,8,11]. antibacterial drugs since the middle of the last century.
Whether the antibiotic is of natural origin or synthetic, However, the frequency of identification of new antibacter-
resistance will eventually emerge (although perhaps not in ial compounds from this source has dropped since the
all bacterial species) and may already be circulating in the 1970s. Because many soil bacteria (the traditional source
environment. Evidence for this assertion comes from a of antibiotics in current clinical use) produce a similar
recent study that sampled spore-forming soil bacteria in range of antibiotics identifying new antibiotics from these
several different environments and found all of these sources becomes ever more difficult. Richard Baltz has
microbes to be multidrug resistant [12]. On average, each discussed this problem [15] and has noted that the anti-
strain of non-pathogenic bacteria was found to be resistant biotic daptomycin (approved in 2003 as Cubicin1) was only
to 7–8 antibiotics from a panel of 21 sampled. Resistance found in 2 of 107 soil actinomycetes. By contrast, the
was not restricted to antibiotics of natural origin, as may frequency of identification of a streptothricin producer is
have been predicted from such a screen. Rather, significant 2 in 10. This frequency range of 1067 for identifying
levels of resistance were encountered even to new synthetic producers of new antibiotic scaffolds is daunting. Never-
antibiotics, such as linezolid and ciprofloxacin, as well as to theless, promising new antibiotics such as platensimycin
semi-synthetic natural-antibiotic derivatives designed to [16] (Figure 1) continue to be identified from soil bacteria.
overcome known resistance mechanisms; one example of Despite the challenges of identifying new natural-pro-
such a drug is telithromycin (Ketek1). This work, along duct antibiotics, recent work has proven that there are
with the identification of antibiotic-resistance genes in untapped sources of antibiotic natural products. These
metagenomic screens of environmental bacteria [13], include microorganisms from new ecological niches,
shows that the collection of all resistance genes, the anti- natural products from plants, natural-product biosynthetic
biotic resistome [7], is genetically extensive and can clusters in known organisms and previously discovered
respond to a broad range of chemical diversity. compounds that are re-evaluated. Furthermore, synthetic
Over the past 40 years, it has been well established in antimicrobials must not be forgotten and will continue to
both the clinic and the farm that antibiotic use selects for have their place in the future of antibiotics.
the horizontal spread of acquired resistance elements on
mobile genetic elements. Resistance can also emerge as a New sources of antimicrobial chemicals
result of mutation in target genes and persist as a result of Identifying new ecological niches for antibiotic-producing
second-site mutations. Furthermore, the presence of anti- bacteria is providing access to new organisms and new
biotics can accelerate the development of innate resistance bioactive chemicals. The recent realization that there are

Figure 1. New natural-product antibiotics. Platensimycin (soil bacterium), abyssomicin C (marine bacterium), horminone (plant).

www.sciencedirect.com
262 Review TRENDS in Molecular Medicine Vol.13 No.6

bona fide marine actinomycetes and the discovery of the identification of the number and type of natural
laboratory growth conditions for their culture has resulted products that an organism is predicted to be able to
in the identification of several new groups of actinomy- biosynthesize. For example, it was well established that
cetes, including the Salinispora and Marinispora (revie- Bacillus subtilis, which has been studied for more than a
wed in Ref. [17]). These marine bacteria have proven to be century, produced several antibiotics, such as surfactin
sources of several new antitumor and antibiotic natural and fengycin. However, the complete genome sequence of
products. For example, abyssomicin C (Figure 1) is a novel B. subtilis 168 revealed a new 80 kb predicted natural-
inhibitor of bacterial p-aminobenzoic acid biosynthesis product biosynthetic cluster thought to be non-functional
[18,19]. [28]. This cluster has now been shown to produce the
Another source of new antibiotic-producing bacteria are polyketide-peptide hybrid compound bacillaene [29]. Sim-
bacterial symbionts of insects [20,21] and other arthropods ilarly, Streptomyces coelicolor, which has been studied for
[22]. These bacteria have evolved to exist in highly specific more than 50 years, was known to produce 5–6 natural
niches and as a result have their own specific antimicrobial products. Its genome sequence, however, revealed poten-
chemical diversity tuned to their complex interactions with tial gene clusters for 20 natural products [30]. Recently,
the host. These probably represent just a small portion of one of these was shown to produce a new siderophore,
the available antibiotic-producing biodiversity available coelochelin [31].
for exploration. Baltz has estimated that, even in the soil, The ability to predict not only the possibility of natural
only 2% of the antibiotics produced by Streptomyces have product biosynthesis from the presence of a biosynthetic
been identified [15]. gene cluster but also the chemical structure of the metab-
Plants [23] and the engineering of known biosynthetic olite has been improved dramatically by the refinement of
pathways to expand antibiotic chemical diversity [24,25] bioinformatics tools. The structure of coelochelin [31] is one
are additional sources of new antibiotics. Medicinal plants example where a mixture of bioinformatic prediction fol-
have been used for millennia in the treatment of infection. lowed by experimental analysis resulted in the structural
Efforts to isolate active agents in plants have led to the elucidation of a new natural product. Another example is a
identification of several antibacterial agents such as hor- novel cyclic lipopeptide from Pseudomonas syringae [32].
minone (reviewed in ref. [26]) (Figure 1). The group at Ecopia Biosciences has also pioneered a new
The recent ability to modify antibiotic chemical scaffolds informatics approach called DECIPHER1 to successfully
in combinatorial fashion via the manipulation of biosyn- mine bacterial genomes for natural-product biosynthesis
thetic genes has greatly expanded the chemical diversity of clusters and predict the structure of the metabolites
several classes of antibiotics, including the polyketides and [33,34].
cyclic peptides. This approach makes it possible to explore
chemical space on antibiotic scaffolds that in many cases Old antibiotics – new medicines
are out of reach for synthetic chemistry. So far no new It is instructive to recall that all of the three new classes of
antibiotics have been brought to market from plants or antibiotics brought to market in the past decade, Syner-
from biosynthetic-engineering studies, but these are early cid1 (streptogramin combination), linezolid (oxazolidi-
days for these sources of antimicrobial agents and there is none), and daptomycin (lipopeptide), were discovered
reason to be optimistic that they will contribute clinically- and then discounted as viable antibiotics. The streptogra-
useful agents in the future. mins, a combination of two natural-product antibiotics
Finally, we must not forget that chemists have the produced by the same species of bacteria, were first ident-
ability to synthesize de novo new classes of antibiotics. ified in the 1950s. They were used primarily as animal-feed
The first new class of antibiotics in clinical use in more additives until medicinal chemists at Rhone-Poulenc
than 25 years is the oxazolidinones, represented by line- improved the solubility of both components of the antibiotic
zolid (Zyvox1). Unlike most other classes of antibiotics, the to make them suitable for human therapy. The oxazolidi-
oxazolidinones do not have their origins in natural-product nones were first discovered in the mid-1980s by chemists at
chemistry. This group of compounds emerged from syn- Dupont, and daptomycin was identified at roughly the
thetic-chemistry laboratories first at DuPont and then at same time at Lilly. Both antibiotic classes were dropped
Pharmacia and Pfizer more than 15 years ago. The more as clinical candidates because of toxicity concerns. Sub-
structurally complex diversity-oriented synthetic-chemical sequent revisiting of these agents with new chemistry or,
libraries inspired by natural products might be tremen- in the case of daptomycin, new therapeutic dosing regi-
dous sources of new chemistry and provide the opportunity mens has rescued promising agents for clinical use. Other
to exhibit antimicrobial activity [27]. examples of the revisiting of ‘old’ antibiotic scaffolds are the
mannopeptimycins, which block lipid II [35], and ever-
Mining natural-product biosynthetic clusters ninomycin, which inhibits protein translation [36]. Neither
uncovered in bacterial genomes of these has emerged from early-stage clinical trials, but
One of the many surprises of the growing availability of they illustrate the point that historical antibiotic collec-
bacterial-genome sequences is the number of unknown tions are worthy of re-investigation. Since the first screens
natural-product biosynthetic gene clusters that have of soil organisms for antibiotic activity, several thousand
emerged even from very well-studied organisms. The antibiotics have been reported [37]. Are there blockbuster
genes encoding natural products (including antibiotics) drugs awaiting rediscovery in this historical literature?
produced by bacteria are generally clustered in a contig- The examples of daptomycin and the streptogramins
uous fashion along the genome. This greatly facilitates suggest there are.
www.sciencedirect.com
Review TRENDS in Molecular Medicine Vol.13 No.6 263

Certainly, there are leads for new antimicrobial several class A Ser-b-lactamases [40]. These resistance
chemicals and associated targets in the thousands of enzymes act by catalyzing a two-step hydrolytic reaction
known compounds with antimicrobial activity. Modern to inactivate the antibiotic. First, an active-site Ser residue
biochemical and genomic tools enable rapid identification forms a covalent bond with the antibiotic by cleaving the
of the mode of action of antibiotics. These tools, coupled b-lactam ring. This is followed by a second step where a
with powerful synthetic-chemical-library methods for molecule of water hydrolyzes the antibiotic-enzyme com-
rapidly sampling and expanding chemical diversity around plex and thus regenerates active b-lactamase and releases
an active scaffold, allow us to gain new and extensive the inactive antibiotic. Clavulanic acid inhibits b-lacta-
information on target-compound pairs in relatively short mases by forming a highly stable enzyme-Ser-antibiotic
periods of time. For example, a family of acyldepsipeptides complex in the first catalytic step, but it is unable to
produced by Streptomyces hawaiiensis and reported in undergo the second hydrolytic step to release the free
1985 has now been shown to be potent inhibitors of the enzyme, and this failure effectively inactivates the anti-
regulation of the ClpP protease, which leads to bacterial biotic-resistance enzyme. When certain drugs are pack-
cell death, and this finding has led to the identification of a aged with a b-lactam antibiotic such as amoxicillin,
new target-inhibitor pair that can be exploited in down- clavulanic acid has led to the development of potent drug
stream drug design [38]. combinations for clinical use; one such drug is Augmen-
tin1. Similarly, combinations of the b-lactamase inhibitors
New strategies for extending the life of antibiotics sulbactam and tazobactam with b-lactam antibiotics are in
Inhibitors of resistance enzymes clinical use.
The concept of utilizing cognate pairs of antibiotics and These combinations have been most effective against
inhibitors of antibiotic-resistance proteins is clinically pro- the Ambler class A [39] Ser-b-lactamases; however, resist-
ven in the b-lactamase field. There are hundreds of known ant isozymes and the increasing frequency of non-suscept-
b-lactamases, and these have been classified by Ambler ible class B, C and D enzymes is a growing problem. This
et al. [39] as serine (Ser) dependent (classes A, C and D) or has spurred efforts to synthesize and evaluate broad-spec-
metal dependent (class B). Clavulanic acid (Figure 2), trum inhibitors of b-lactamases. For example, Buynak and
produced by Streptomyces clavuligerus, is a b-lactam with colleagues have reported several potent inhibitors of class
weak antibacterial activity but that potently inhibits A, C and D b-lactamase inhibitors based on both penicillin

Figure 2. Inhibitors of b-lactamases. Clavulanic acid, identified in 1976, is in wide clinical use in combination with b-lactam antibiotics and inhibits type A b-lactamases.
a–Vinylidene-b-lactams such as compound A and 3-substituted cephalosporins such as compound B are representative synthetic molecules that have expanded class
A, C, and D b-lactamase inhibition activity. The 6-methylidene penem compound C has broad-range anti-b-lactamase activity including activity against extended-spectrum
b–lactamases. Bulgecin A is a natural-product inhibitor of type B metallo-b-lactamases.

www.sciencedirect.com
264 Review TRENDS in Molecular Medicine Vol.13 No.6

and cephalosporin scaffolds (e.g. Figure 2, reviewed in Ref. shown to produce several inhibitors of antibiotic efflux
[41]), and Wyeth has reported on a series of 6-methylidene proteins; these include reserpine, one of the first pump
penems with activity against extended-spectrum b-lacta- inhibitors shown to potentiate antibiotics in Gram-positive
mases (Figure 2) [42]. bacteria (reviewed in Ref. [55]). The Lewis group has
The emergence of class B metallo-b-lactamases in the shown that in the case of the antibiotic berberine, species
clinic is a growing problem [43]. These enzymes use a of plants simultaneously produce 50 -methoxyhydnocarpin,
different catalytic mechanism than the Ser-b-lactamase an inhibitor of major-facilitator efflux proteins [56], pre-
to inactivate the antibiotic. Instead of a covalent enzyme- sumably to overcome the problem of antibiotic resistance.
antibiotic intermediate, an active-site Zn2+ ion (1–2 atoms Recently, this group has shown that chemically linking an
per active site) generates a reactive water molecule that is antibiotic and efflux-pump inhibitor makes it possible to
poised for cleavage of the b-lactam ring (reviewed in Ref. obtain successful hybrid compounds with activity against
[44]). b-Lactam-based inhibitors of the Ser-enzymes are in resistant bacteria [57].
fact substrates for these enzymes and therefore have no
inhibitory effect. As a result, new classes of small-molecule Novel antibiotic strategies
inhibitors specific to metallo-b-lactamases are being In addition to new antibiotic compounds and inhibitors of
sought. Recent studies have identified several such inhibi- resistance, new strategies to prolong the effectiveness of
tors that chelate the active site Zn2+; these include thiols the current arsenal of antibiotic drugs as well as agents
and tetrazoles (e.g. Refs. [45,46] and reviewed in Ref. [43]) that interfere with bacterial virulence and block infection
as well as the sulfated glycopeptide natural product bul- are being explored. For example, bacterial exposure to
gecin A [47] (Figure 2). certain classes of antibiotics has been demonstrated to
Inhibitors of aminoglycoside-resistance enzymes have actually accelerate the emergence of resistance by inducing
been described (reviewed in Ref. [48]). More recently, inhi- horizontal gene transfer [58] and mutation via the SOS
bition of an aminoglycoside kinase, APH(30 )-IIIa, by a response [14]. Preventing the SOS response, for example
rationally designed ankyrin-repeat protein that traps the by blocking the protease LexA [14] or by inhibiting RecA
enzyme in a non-productive complex has been reported [49]. [59], are novel strategies for preventing the emergence of
This ankyrin-repeat protein is unlikely to be clinically use- resistance during antibiotic chemotherapy and could be
ful. Nevertheless, this inhibition strategy, with a focus on used in combination with existing antibiotics.
non-active site-directed compounds, could be emulated with Another new strategy aimed at interfering with
small molecules. Bisubstrate inhibitors of aminoglycoside bacterial physiology is the blockade of intercellular com-
acetyltransferase, AAC(60 )-Ii, that reverse resistance in munication. Bacteria use quorum-sensing molecules such
bacterial culture have also been recently published [50]. as acyl homoserine lactones during infection to promote
One difficulty with targeting aminoglycoside resistance is both colonization and cell-type modulation (e.g. the for-
that many clinically resistant bacteria harbor more than one mation of biofilm [60]). Therefore, interfering with this
resistance enzyme, often with completely different inacti- process could be a target for novel antimicrobial agents
vation chemistry (kinase, acetyltransferase and adenylyl- that do not necessarily kill the cell but rather prevent or
transferase). This makes small-molecule targeting of attenuate infection by targeting virulence. A recent high-
aminoglycoside resistance a challenge. However, cationic throughput screen of 200 000 small molecules identified
peptides have shown in vitro ability to inhibit multiple cell-permeable inhibitors of the P. aeruginosa LasR-de-
enzymes, including kinases and acetyltransferases [51], pendent quorum-sensing system (Figure 3) that could
suggesting that these compounds could form the basis for serve as leads for new anti-virulence compounds [61].
an expanded search for a pan-aminoglycoside-resistance Other recent examples of new anti-virulence strategies
inhibitor. include inhibitors of Chlamydia trachomatis type III
secretion [62], ToxT-dependent gene expression in Vibrio
Inhibitors of antibiotic efflux cholerae [63] (Figure 3) and sortase-dependent protein
Gram-negative and Gram-positive bacteria use efflux modification of Gram-positive cell wall [64]. This approach
proteins to pump antibiotics from the periplasm or cytosol does not inhibit cell growth, and as a result, targeting
to the extracellular medium. These membrane-spanning virulence might be less prone to selection for resistance
proteins use energy, by performing ATP hydrolysis, by because the organisms are not in ‘adapt or die’ mode where
coupling efflux to ion transport or by harnessing the pro- the presence of an antibiotic can powerfully select for a
ton-motive force, to pump antibiotic out of the cell against a rare resistant mutant.
concentration gradient. There are five classes of antibiotic Another important virulence factor is the outer
efflux proteins, each with a distinctive topology, quatern- membrane of Gram-negative bacteria. This asymmetric
ary structure and primary sequence; these include the bilayer includes a lipopolysaccharide layer that has highly
ABC-transporter, major-facilitator, MATE, SMR and conserved and highly variable polymer components. Cells
RND families (reviewed in Refs [52,53]). The importance that are deficient in the synthesis of the conserved lipid
of these resistance proteins, in particular members of the and carbohydrate components of lipopolysaccharide are
RND and major-facilitator classes, in clinical antibiotic- either non-viable (e.g. LpxC) or avirulent (e.g. heptose
resistance has prompted the search for inhibitors of these biosynthesis). Furthermore, viable mutants impaired in
pumps [54]. heptose biosynthesis are hyper-susceptible to many anti-
Inhibitors of efflux pumps include synthetic and biotics [65]. This has resulted in the exploration of lipopo-
natural-product molecules. Plants in particular have been lysaccharide biosynthesis as a target for either novel
www.sciencedirect.com
Review TRENDS in Molecular Medicine Vol.13 No.6 265

Figure 3. Small-molecule inhibitors of bacterial virulence. (a) Inhibitors of quorum sensing in P. aeruginosa [61]. (b) Virstatin is an inhibitor of ToxT-dependent gene
expression in V. cholerae [63], and INP0400 blocks C. trachomatis type III secretion [62].

antibacterials or antibiotic adjuvants, that is, compounds systems biology of antibiotic combinations is beginning to
that synergize with antibiotics. For example, inhibitors allow exploration of this concept in a rigorous fashion [72]
of LpxC have been shown to have narrow-spectrum and may yield new avenues for antibiotic therapy.
but potent antibacterial activity [66], and a recent high-
throughput screen of heptose biosynthesis identified the Conclusions
first small-molecule inhibitors of this pathway [67]. Sim- After a half century of clinical antibiotic use, resistance,
ilarly, in Gram-positive bacteria inhibiting the biosyn- and in particular MDR, has emerged as a formidable
thesis of cell-wall polymers teichoic and lipoteichoic acid problem with no signs of abating. In fact, MDR may be a
represent both novel antibacterial and antibiotic-sensitiz- natural state for many bacteria. Addressing this issue will
ing targets [68,69]. require the collaboration of many disciplines and inno-
The expanded concept of antibiotic adjuvants is worthy of vation in the identification of new therapeutics and com-
exploration. These are compounds that may not have sig- plimentary antimicrobial strategies.
nificant antibiotic activity themselves but that improve There are several challenges in identifying new
their biological activity when combined with antibiotics. antibiotics with the potential to make it through clinical
Inhibitors of resistance mechanisms and inhibitors of lipo- trials and into medical practice. Among these is the fact
polysaccharide biosynthesis, discussed above, are examples that discovering new chemically distinct antibiotics from
of such adjuvants. Synergistic combinations of antibiotics traditional sources (such as soil bacteria and existing
are another example that is well precedented in infectious- synthetic-chemical collections) is increasingly difficult.
disease medicine. Given the number of predicted natural- Sampling the biological and chemical diversity of atypical
product biosynthetic clusters in sequenced actinomycetes sources such as marine bacteria is beginning to identify
(20 in Streptomyces coelicolor and 30 in Streptomyces novel antibiotic scaffolds. Whether any of these will be
avermitilis), Challis and Hopwood hypothesized that suitable to be developed into new drugs remains to be seen.
natural-product synergy may be a normal state for anti- However, given the precedent of the previous 50 years, it
biotic-producing bacteria [70]. In other words, the unex- seems highly likely that this new chemistry will eventually
pected capacity for bacteria to produce multiple natural yield new drugs.
products may reflect an ecological advantage for co-pro- The focus of many antimicrobial drug-discovery
duction of secondary metabolites. Co-production of the programs has been, and continues to be, on broad-spec-
synergistic combination of chemically unrelated type A trum antibiotics that target as many bacteria as possible.
and type B streptogramin antibiotics are a proven example This reflects our woefully poor ability to rapidly identify
of genetically programmed synergy [71]. Analysis of the the causative agent of infection in most hospital and
www.sciencedirect.com
266 Review TRENDS in Molecular Medicine Vol.13 No.6

community settings as well as the fact that in many cases ‘Golden era’ from 1945–1960, when the majority of
infection may be the result of more than one microbe. This antibiotic scaffolds in current use were identified. The
‘carpet bombing’ strategy of infection control has surely new technologies and innovative strategies discussed
contributed to the problem of antibiotic resistance, as exem- above have the potential to answer the challenge of multi-
plified by the rise in highly virulent infections caused by drug-resistant bacteria in the coming years.
drug-resistant C. difficile [73]. Leveraging of improved diag-
nostic technology and microbial-genome sequencing has the Acknowledgements
potential to be applied in the rapid identification of the This work is supported by the Canadian Institutes of Health Research,
the Natural Sciences and Engineering Research Council of Canada and a
pathogens involved in infection. As a result, there is a Canada Research Chair in Antibiotic Biochemistry.
growing opportunity to explore organism-specific anti-
biotics. This will require new paradigms in infection control, References
regulatory approval of new diagnostics and, importantly, 1 Talbot, G.H. et al. (2006) Bad bugs need drugs: an update on the
confidence among physicians that the strategy will work. development pipeline from the Antimicrobial Availability Task
However, the benefits of preventing the eradication of Force of the Infectious Diseases Society of America. Clin. Infect. Dis.
42, 657–668
beneficial flora as collateral damage and the broad selective
2 Dorman, S.E. and Chaisson, R.E. (2007) From magic bullets back to the
pressure associated with antibiotic resistance merit rese- Magic Mountain: the rise of extensively drug-resistant tuberculosis.
arch efforts in this area. Nat. Med. 13, 295–298
Antibiotic adjuvants, including novel compound 3 Sebaihia, M. et al. (2006) The multidrug-resistant human pathogen
combinations and inhibitors of resistance mechanisms, Clostridium difficile has a highly mobile, mosaic genome. Nat. Genet.
38, 779–786
as well as targeting of microbial virulence, are approaches
4 Wright, G.D. (2005) Bacterial resistance to antibiotics: enzymatic
that can extend the life of successful antibiotic drugs. degradation and modification. Adv. Drug Deliv. Rev. 57, 1451–1470
Because these antibiotics have known pharmacology and 5 Walsh, C.T. (2003) Antibiotics: Actions, Origins, Resistance, ASM Press
sometimes decades of clinical history, the time to market 6 Levy, S.B. and Marshall, B. (2004) Antibacterial resistance worldwide:
may be relatively short. The problem of multiple methods causes, challenges and responses. Nat. Med. 10, S122–S129
7 Wright, G.D. (2007) The antibiotic resistome: the nexus of chemical and
of drug resistance is significant; however, the success of the genetic diversity. Nat. Rev. Microbiol. 5, 175–186
b-lactamase inhibitors demonstrates that there is a mar- 8 Summers, A.O. (2006) Genetic linkage and horizontal gene transfer,
ket for well-designed antibiotic adjuvants. The regulatory the roots of the antibiotic multi-resistance problem. Anim. Biotechnol.
and pharmacological challenges in this approach are not 17, 125–135
trivial (see Box 1): Compound combinations must have 9 Lee, D.G. et al. (2006) Genomic analysis reveals that Pseudomonas
aeruginosa virulence is combinatorial. Genome Biol. 7, R90
similar pharmacological profiles, and designing successful 10 Roy, P.H. (1999) Horizontal transfer of genes in bacteria. Microbiol.
clinical trials for drug combinations or anti-virulence com- Today 26, 168–170
pounds and the massive expense of these are major hurdles 11 Fluit, A.C. and Schmitz, F.J. (2004) Resistance integrons and super-
to be overcome. integrons. Clin. Microbiol. Infect. 10, 272–288
12 D’Costa, V.M. et al. (2006) Sampling the antibiotic resistome. Science
Despite these challenges, in the 21st Century we are in a
311, 374–377
better position to address the problem of resistance than 13 Riesenfeld, C.S. et al. (2004) Uncultured soil bacteria are a reservoir of
ever before. Exploration of new biological diversity is new antibiotic resistance genes. Environ. Microbiol. 6, 981–989
revealing new antimicrobial chemical diversity, our un- 14 Cirz, R.T. et al. (2005) Inhibition of mutation and combating the
derstanding of the molecular mechanisms of resistance evolution of antibiotic resistance. PLoS Biol. 3, e176
15 Baltz, R.H. (2005) Antibiotic discovery from actinomycetes: will a
and its evolution is improving and the genomics revolution
renaisssance follow the decline and fall? SIM News 55, 186–196
has resulted not only in a remarkable and growing number 16 Wang, J. et al. (2006) Platensimycin is a selective FabF inhibitor with
of whole-genome sequences, but also in the stunning ability potent antibiotic properties. Nature 441, 358–361
to generate atomic-resolution structures of a myriad of 17 Fenical, W. and Jensen, P.R. (2006) Developing a new resource for drug
antibiotic targets, from the ribosome to cell-wall synthetic discovery: marine actinomycete bacteria. Nat. Chem. Biol. 2, 666–673
18 Riedlinger, J. et al. (2004) Abyssomicins, inhibitors of the para-
machinery. In many ways we are entering into what should aminobenzoic acid pathway produced by the marine Verrucosispora
be a new ‘Golden era’ of antibiotics that builds on the first strain AB-18-032. J. Antibiot. (Tokyo) 57, 271–279
19 Bister, B. et al. (2004) Abyssomicin C-A polycyclic antibiotic from a
marine Verrucosispora strain as an inhibitor of the p-aminobenzoic
Box 1. Outstanding questions acid/tetrahydrofolate biosynthesis pathway. Angew. Chem. Int. Ed.
Engl. 43, 2574–2576
1. What are the best natural-product and synthetic-chemical
20 Kaltenpoth, M. et al. (2005) Symbiotic bacteria protect wasp larvae
sources of new antibiotic chemical scaffolds? How can we enrich
from fungal infestation. Curr. Biol. 15, 475–479
compound collections and natural-product extracts with anti-
21 Currie, C.R. et al. (1999) Fungus-growing ants use antibiotic-producing
microbial compounds?
bacteria to control garden parasites. Nature 398, 701–704
2. Can we identify new highly effective inhibitors of antibiotic-
22 Gebhardt, K. et al. (2002) Screening for biologically active metabolites
resistance mechanisms with the potential to reverse resistance
with endosymbiotic bacilli isolated from arthropods. FEMS Microbiol.
during infection?
Lett. 217, 199–205
3. Can novel drug combinations extend the life of ‘old’ antibiotics?
23 Lewis, K. and Ausubel, F.M. (2006) Prospects for plant-derived
4. Are inhibitors of microbial virulence good candidates for new
antibacterials. Nat. Biotechnol. 24, 1504–1507
prophylactic drugs?
24 Lamb, S.S. and Wright, G.D. (2005) Accessorizing natural products:
5. Can rapid diagnostics coupled with organism-specific antibiotics
adding to nature’s toolbox. Proc. Natl. Acad. Sci. U. S. A. 102, 519–520
help to reduce antibiotic resistance in microbial populations and
25 Baltz, R.H. (2006) Molecular engineering approaches to peptide,
off-target killing of beneficial microbial species?
polyketide and other antibiotics. Nat. Biotechnol. 24, 1533–1540
6. Will the regulatory community support new antibiotics ap-
26 Gibbons, S. (2004) Anti-staphylococcal plant natural products. Nat.
proaches such as drug combinations and anti-virulence drugs?
Prod. Rep. 21, 263–277

www.sciencedirect.com
Review TRENDS in Molecular Medicine Vol.13 No.6 267

27 Thomas, G.L. et al. (2006) Enriching chemical space with diversity- 51 Boehr, D.D. et al. (2003) Broad-spectrum peptide inhibitors of
oriented synthesis. Curr. Opin. Drug Discov. Devel. 9, 700–712 aminoglycoside antibiotic resistance enzymes. Chem. Biol. 10, 189–196
28 Kunst, F. et al. (1997) The complete genome sequence of the gram- 52 Piddock, L.J. (2006) Clinically relevant chromosomally encoded
positive bacterium Bacillus subtilis. Nature 390, 249–256 multidrug resistance efflux pumps in bacteria. Clin. Microbiol. Rev.
29 Straight, P.D. et al. (2007) A singular enzymatic megacomplex from 19, 382–402
Bacillus subtilis. Proc. Natl. Acad. Sci. U. S. A. 104, 305–310 53 Poole, K. (2005) Efflux-mediated antimicrobial resistance. J.
30 Bentley, S.D. et al. (2002) Complete genome sequence of the model Antimicrob. Chemother. 56, 20–51
actinomycete Streptomyces coelicolor A3(2). Nature 417, 141–147 54 Lomovskaya, O. and Bostian, K.A. (2006) Practical applications and
31 Lautru, S. et al. (2005) Discovery of a new peptide natural product by feasibility of efflux pump inhibitors in the clinic–a vision for applied
Streptomyces coelicolor genome mining. Nat. Chem. Biol. 1, 265–269 use. Biochem. Pharmacol. 71, 910–918
32 de Bruijn, I. et al. (2007) Genome-bases discovery, structure prediction 55 Stavri, M. et al. (2006) Bacterial efflux pump inhibitors from natural
and functional analysis of cyclic lipopeptide antibiotics in sources. J Antimicrob Chemother
Pseudomonas species. Mol. Microbiol. 63, 417–428 56 Stermitz, F.R. et al. (2000) Synergy in a medicinal plant: antimicrobial
33 Zazopoulos, E. et al. (2003) A genomics-guided approach for discovering action of berberine potentiated by 50 -methoxyhydnocarpin, a multidrug
and expressing cryptic metabolic pathways. Nat. Biotechnol. 21, 187– pump inhibitor. Proc. Natl. Acad. Sci. U. S. A. 97, 1433–1437
190 57 Ball, A.R. et al. (2006) Conjugating berberine to a multidrug efflux
34 McAlpine, J.B. et al. (2005) Microbial genomics as a guide to drug pump inhibitor creates an effective antimicrobial. ACS Chem Biol 1,
discovery and structural elucidation: ECO-02301, a novel antifungal 594–600
agent, as an example. J. Nat. Prod. 68, 493–496 58 Hastings, P.J. et al. (2004) Antibiotic-induced lateral transfer of
35 He, H. (2005) Mannopeptimycins, a novel class of glycopeptide antibiotic resistance. Trends Microbiol. 12, 401–404
antibiotics active against gram-positive bacteria. Appl. Microbiol. 59 Lee, A.M. et al. (2005) A molecular target for suppression of the
Biotechnol. 67, 444–452 evolution of antibiotic resistance: inhibition of the Escherichia coli
36 Boucher, H.W. et al. (2001) In vivo activity of evernimicin (SCH 27899) RecA protein by N(6)-(1-naphthyl)-ADP. J. Med. Chem. 48, 5408–5411
against methicillin-resistant Staphylococcus aureus in experimental 60 Gonzalez, J.E. and Keshavan, N.D. (2006) Messing with bacterial
infective endocarditis. Antimicrob. Agents Chemother. 45, 208–211 quorum sensing. Microbiol. Mol. Biol. Rev. 70, 859–875
37 Berdy, J. (2005) Bioactive microbial metabolites. J. Antibiot. (Tokyo) 61 Muh, U. et al. (2006) Novel Pseudomonas aeruginosa quorum-sensing
58, 1–26 inhibitors identified in an ultra-high-throughput screen. Antimicrob.
38 Brotz-Oesterhelt, H. et al. (2005) Dysregulation of bacterial proteolytic Agents Chemother. 50, 3674–3679
machinery by a new class of antibiotics. Nat. Med. 11, 1082–1087 62 Muschiol, S. et al. (2006) A small-molecule inhibitor of type III secretion
39 Ambler, R.P. et al. (1991) A standard numbering scheme for the class A inhibits different stages of the infectious cycle of Chlamydia
beta-lactamases. Biochem. J. 276, 269–270 trachomatis. Proc. Natl. Acad. Sci. U. S. A. 103, 14566–14571
40 Neu, H.C. and Fu, K.P. (1978) Clavulanic acid, a novel inhibitor of beta- 63 Hung, D.T. et al. (2005) Small-molecule inhibitor of Vibrio cholerae
lactamases. Antimicrob. Agents Chemother. 14, 650–655 virulence and intestinal colonization. Science 310, 670–674
41 Buynak, J.D. (2004) The discovery and development of modified 64 Frankel, B.A. et al. (2004) Vinyl sulfones: inhibitors of SrtA, a
penicillin- and cephalosporin-derived beta-lactamase inhibitors. transpeptidase required for cell wall protein anchoring and
Curr. Med. Chem. 11, 1951–1964 virulence in Staphylococcus aureus. J. Am. Chem. Soc. 126, 3404–3405
42 Weiss, W.J. et al. (2004) In vitro and in vivo activities of novel 6- 65 Valvano, M.A. et al. (2002) Novel pathways for biosynthesis of
methylidene penems as beta-lactamase inhibitors. Antimicrob. Agents nucleotide-activated glycero-manno-heptose precursors of bacterial
Chemother. 48, 4589–4596 glycoproteins and cell surface polysaccharides. Microbiology 148,
43 Walsh, T.R. et al. (2005) Metallo-beta-lactamases: the quiet before the 1979–1989
storm? Clin. Microbiol. Rev. 18, 306–325 66 Onishi, H.R. et al. (1996) Antibacterial agents that inhibit lipid A
44 Crowder, M.W. et al. (2006) Metallo-beta-lactamases: novel weaponry biosynthesis. Science 274, 980–982
for antibiotic resistance in bacteria. Acc. Chem. Res. 39, 721–728 67 De Leon, G.P. et al. (2006) An in vitro screen of bacterial
45 Toney, J.H. et al. (1998) Antibiotic sensitization using biphenyl lipopolysaccharide biosynthetic enzymes identifies an inhibitor of
tetrazoles as potent inhibitors of Bacteroides fragilis metallo-beta- ADP-heptose biosynthesis. Chem. Biol. 13, 437–441
lactamase. Chem. Biol. 5, 185–196 68 May, J.J. et al. (2005) Inhibition of the D-alanine:D-alanyl carrier protein
46 Concha, N.O. et al. (2000) Crystal structure of the IMP-1 metallo beta- ligase from Bacillus subtilis increases the bacterium’s susceptibility to
lactamase from Pseudomonas aeruginosa and its complex with a antibiotics that target the cell wall. FEBS J. 272, 2993–3003
mercaptocarboxylate inhibitor: binding determinants of a potent, 69 Brown, E.D. and Wright, G.D. (2005) New targets and screening
broad-spectrum inhibitor. Biochemistry 39, 4288–4298 approaches in antimicrobial drug discovery. Chem. Rev. 105, 759–774
47 Simm, A.M. et al. (2005) Bulgecin A: a novel inhibitor of binuclear 70 Challis, G.L. and Hopwood, D.A. (2003) Synergy and contingency as
metallo-beta-lactamases. Biochem. J. 387, 585–590 driving forces for the evolution of multiple secondary metabolite
48 Wright, G.D. (2000) Resisting resistance: new chemical strategies for production by Streptomyces species. Proc. Natl. Acad. Sci. U. S. A.
battling superbugs. Chem. Biol. 7, R127–R132 100 (Suppl 2), 14555–14561
49 Kohl, A. et al. (2005) Allosteric inhibition of aminoglycoside 71 Cocito, C. (1979) Antibiotics of the virginiamycin family, inhibitors
phosphotransferase by a designed ankyrin repeat protein. Structure which contain synergistic components. Microbiol. Rev. 43, 145–192
13, 1131–1141 72 Yeh, P. et al. (2006) Functional classification of drugs by properties of
50 Gao, F. et al. (2006) Synthesis and structure-activity relationships of their pairwise interactions. Nat. Genet. 38, 489–494
truncated bisubstrate inhibitors of aminoglycoside 60 -N-acetyltran- 73 Bartlett, J.G. (2006) Narrative review: the new epidemic of Clostridium
sferases. J. Med. Chem. 49, 5273–5281 difficile-associated enteric disease. Ann. Intern. Med. 145, 758–764

www.sciencedirect.com

Das könnte Ihnen auch gefallen