You are on page 1of 8

REVIEW doi:10.

1038/nature17042

Antibacterial drug discovery


in the resistance era
Eric D. Brown1,2 & Gerard D. Wright1,2

The looming antibiotic-resistance crisis has penetrated the consciousness of clinicians, researchers, policymakers, poli-
ticians and the public at large. The evolution and widespread distribution of antibiotic-resistance elements in bacterial
pathogens has made diseases that were once easily treatable deadly again. Unfortunately, accompanying the rise in global
resistance is a failure in antibacterial drug discovery. Lessons from the history of antibiotic discovery and fresh under-
standing of antibiotic action and the cell biology of microorganisms have the potential to deliver twenty-first century
medicines that are able to control infection in the resistance era.

W
e are entering a period in infection control that has not been in pathogens is typical in the resistance era14 and is eroding our ability
experienced for more than three-quarters of a century. In to control infections with antibiotics. Pathogenic strains of bacteria
this era of ever-increasing resistance (Fig. 1), the manage- that are resistant to most, or all, available antibiotics are now isolated
ment of bacterial infection through the use of safe, cheap and plenti- routinely. We have entered the post-antibiotic age.
ful antibiotics can no longer be taken for granted. The serendipitous
discovery of penicillin in 1929 (ref. 1) enabled the effective control of Mechanisms and origins of antibiotics
infections caused by Gram-positive pathogens such as Staphylococcus Antibiotics perturb important biochemical processes, which results in
and Streptococcus, and the isolation of streptomycin in 1943 (ref. 2) the inhibition of cell growth and division and, in the case of bacteri-
facilitated the control of tuberculosis agent Mycobacterium tubercu- cidal agents, cell death15. The first antimicrobial agents were synthetic
losis — for the first time in history. These discoveries propelled the molecules that were discovered by screening libraries of chemicals, in
golden era of antibiotics, in which natural scaffolds and alternative particular dyes. This screening was superseded by the realization that
versions of these pioneering drugs were uncovered by mining the bacteria and fungi in the environment produce metabolites that could
specialized metabolism of bacteria and fungi or by the chemical modi- treat bacterial infections in humans with remarkable efficacy and mini-
fication of existing scaffolds (Table 1). The results were revolutionary. mal toxic side effects. The strategy adopted by Selman Waksman, a co-
Released from the hegemony of infection, clinicians began to trans- discoverer of streptomycin, involved screening soil-dwelling bacteria,
form medicine. Invasive surgeries became routine, immune-system- and in particular the spore-forming Actinomycetes, for the production
shattering chemotherapy was introduced to fight the ‘war’ on cancer, of metabolites that block the growth of pathogens — a process that was
organ transplantation extended lives and the radical replacement of later termed the Waksman platform16. The simplicity and effectiveness
deteriorating joints, diseased corneas and burnt skin improved quality of the platform ushered in the golden era of antibiotic discovery, the
of life for millions of people. In the resistance era, these breakthroughs period in which most of the microbial natural scaffolds that serve as our
— and the consequent improvements in our quality of life and life antibiotic arsenal were discovered (Fig. 1). The platform also imposed
expectancy — are at risk. measures of success that subsequent drug-discovery efforts would need
The microbial world has always had the molecular tools to drive to use, in particular use of the inhibition of cell growth in vitro, which
resistance. Surveys of microorganisms in the environment and in sam- is assessed on rich media, as the main way to determine the minimal
ples of ancient permafrost revealed that the antibiotic resistome — inhibitory concentration (MIC) of a compound.
the global collection of all microbial resistance genes — is genetically By the mid-1960s, new and effective antibiotic scaffolds were
diverse3,4, widespread across all environmental niches5–8 and pre-dates becoming harder to identify using the Waksman platform. Because
the modern antibiotic era by millennia9. The use of large quantities of these specialized metabolites are the product of microbial evolution
antibiotics to control infection in human and animal diseases and in within a specific environment and were not designed as drugs, most
agriculture has created unprecedented conditions for the mobiliza- had considerable pharmacological or toxicological drawbacks. And
tion of resistance elements in bacterial populations and their capture resistance to these early antibiotics, the result of the horizontal transfer
by previously antibiotic-sensitive pathogens. Over time, these condi- of resistance genes between bacteria or chromosomal mutation, was
tions have enabled the selection of evermore drug-resistant bacteria also becoming a problem. These issues spawned the era of medici-
through three main mechanisms: the sequential capture of a myriad nal chemistry, the next period of innovation in antibiotic discovery
of resistance genes, mostly through mobilization and horizontal trans- (Fig. 1). The development of antibiotics in this period was dominated
fer from environmental sources; the ‘freezing’ of polymorphisms in by cycles of innovation that focused largely on creating synthetic ver-
antibiotic target genes through secondary mutations that neutralize sions of the natural scaffolds of the golden era of discovery. These
the effects on fitness that drug-resistance mutations often impart10; derivatives led to outstanding improvements in the application of
and the upregulation of intrinsic mechanisms of resistance, such as antibiotics, which included lower doses, an expanded antimicrobial
efflux11,12 or antibiotic-inactivating enzymes13. Multidrug resistance spectrum against various pathogens, and the avoidance of resistance.
1
Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario L8S 4L8, Canada. 2Department of Biochemistry and Biomedical Sciences, McMaster
University, Hamilton, Ontario L8S 4L8, Canada.

3 3 6 | N AT U R E | VO L 5 2 9 | 2 1 JA N UA RY 2 0 1 6
© 2016 Macmillan Publishers Limited. All rights reserved
REVIEW INSIGHT

These newer drugs substantially changed the practice of medicine


and ushered in the ‘miracles’ of modern medicine that we now take 1925
for granted.
Most existing antibiotics are therefore derived from natural products
Golden era
and tend to target the bacterial cell wall, DNA or ribosomes. With a few • Natural products
exceptions, these compounds exert pleiotropic and complex effects on • Whole-cell screens
1950
the bacterial cell and often have more than one molecular target. The • High success

β-lactam antibiotics, such as penicillin, covalently modify a number of


target enzymes known as penicillin-binding proteins (PBPs). Collec- Medicinal chemistry era
tively, these enzymes are responsible for the synthesis and remodelling • Synthetic tweaking
of the bacterial cell wall for growth and division. Antibiotics that inhibit 1975 • Whole-cell screens
• Broad spectrum
protein synthesis target the ribosome, which is encoded by multiple • High success
copies of ribosomal DNA, and those that block DNA synthesis act on
several topoisomerase enzymes. Importantly, this effect on multiple Resistance era
cellular targets limits the frequency of spontaneous resistance that can • Modern discovery
2000
arise from mutation in the target gene. Inhibition of the molecular • Target-based
• Broad spectrum
targets of antibiotics commonly results in complex downstream effects • Low success
that exceed those of simple enzyme inhibition. Mounting evidence sug-
gests that β-lactam antibiotics disorganize the activity of the bacterial Narrow-spectrum era
cell-wall synthesis machinery in a manner far more complicated than 2025 • Unconventional discovery
simple inhibition17. The ribosome is a macromolecular complex with • In vivo essential targets
• Combinatorial approaches
many enzymatic functions, regulatory sites and components. When it • Diagnostic development
is targeted by aminoglycosides, for example, the resulting synthesis of • Predicted success
aberrant proteins leads to pleiotropic and toxic effects on the bacterial
cell18,19. Systems-biology approaches suggest that reactive oxygen spe-
cies have been overlooked as contributors to cell death20. Although this Figure 1 | Models of antibiotic drug discovery and development.  Organized
hypothesis remains controversial, there is an increasing appreciation with respect to the prevalent models of each era, this timeline highlights the
that bacterial cell death is complex and probably requires the involve- history and future of antibiotic drug discovery. The golden era was initiated by
ment of several cellular pathways. Many natural-product antibiotics the discovery of sulfonamide and penicillin and is typified by the methods of
are the product of selection for these complex traits over millions of Selman Waksman and his contemporaries, who used whole-cell screening of
natural product extracts to find new antibiotic scaffolds. In subsequent years,
years of evolution. Therefore, it is perhaps unsurprising that modern
termed the medicinal-chemistry era, these scaffolds were modified chemically.
methods of drug discovery have yet to deliver compounds with efficacy Emphasis in the resistance era has fallen on target-based drug discovery to find
comparable to that of the first generation of natural antibiotics and broad-spectrum agents — a model that has failed to provide new antibiotics. In
their semisynthetic derivatives. the future, a focus on innovative methods and unconventional targets should
help to create narrow-spectrum agents and associated diagnostics.
Modern antibacterial drug discovery
Successes in the golden era were followed by a considerable hiatus generation of antibiotics that are insusceptible to existing mechanisms
in the identification of new scaffolds that began in the 1960s and of resistance28.
lasted until the early 1990s, when a renaissance in discovery efforts In the face of increasing resistance, the failure to yield new anti-
was precipitated by a fresh wave of resistance. This coincided with biotics has been the cause of much concern. Unsuccessful antibac-
the emergence of innovative drug-discovery approaches in all thera- terial programmes have been documented by industry experts29–31
peutic areas. These new methods were underpinned by remarkable and common themes are evident. Large-scale gene dispensability
advances in technology, such as breakthroughs in the manipulation studies generated numerous targets — 160 in one account29 — and
of recombinant DNA to produce desired proteins at high yields and in many were tested in in vitro screening campaigns. However, these
high-throughput synthesis to create large chemical libraries. Improve- high-throughput biochemical screens of large collections of synthetic
ments in facile protein-structure determination enabled rational chemicals often were unable to find promising compounds with the
drug design, and robotic liquid handling facilitated high-throughput necessary physical and chemical properties. When encouraging lead
screens of biochemical assays. Furthermore, a computing revolution compounds were found, these failed, largely because of difficulties in
made it possible to handle much larger data sets. When coupled with developing antibacterial drugs with a suitable product profile — for
emergent genomic technologies, these advances enabled a ‘genes-to- example, efficacy against a broad spectrum of pathogens or against
drugs’ model that has been applied with success to many therapeutic difficult-to-treat Gram-negative organisms. Gram-negative bacteria
areas21 and become the convention. Yet more than two decades into have proved especially daunting owing to a poor understanding of
the practice of modern antibacterial drug discovery, no new medicines drug penetration and efflux systems that are pervasive in this group
have been discovered through this approach. of pathogens. There were also concerns about the rapid evolution of
Antibacterial drug discovery has been especially influenced by resistance that is often evident in these single-target approaches. Aside
genomic data, technology and innovation. The first free-living organ- from its emphasis on target-based efforts, modern antibacterial drug
ism to be sequenced was the pathogen Haemophilus influenzae22 and, discovery has also employed large chemical screens of whole cells for
since then, thousands of bacterial genomes have been sequenced. growth inhibition. However, such screens produce huge numbers of
High-throughput techniques that can create precise deletions at the active compounds that are difficult to follow up because of a lack of
genome scale have been used to explore methodically the dispen- tools for prioritization. Indeed, there has been a resurgence in cell-
sability of each gene in both the model Gram-positive bacterium based approaches that includes innovative strategies for focusing on
Bacillus subtilis23 and in the model Gram-negative bacterium Escheri- promising leads that target different aspects of bacterial survival32.
chia coli24. Pathogens such as Staphylococcus aureus and Pseudomonas Ultimately, these approaches will focus on specific targets to facilitate
aeruginosa have also been the subject of systematic approaches to the lead-optimization strategies that are inherent to modern drug dis-
mutagenesis25,26, as is the fastidious pathogen M. tuberculosis27. The covery. Therefore, it will become increasingly important to under-
goal of these large-scale surveys has been to identify new targets for a stand the failures of target-based approaches.

2 1 JA N UA RY 2 0 1 6 | VO L 5 2 9 | N AT U R E | 3 3 7
© 2016 Macmillan Publishers Limited. All rights reserved
INSIGHT REVIEW

The essential-gene paradox from a person with cystic fibrosis39. Vitamin biosynthesis was found to be
With its focus on the optimization of lead compounds that target indi- an important requirement for bacterial viability — noteworthy because
vidual proteins using state-of-the-art tools of chemistry, biochemistry and searches for antibacterial chemicals have overwhelmingly emphasized
pharmacology, modern industrial drug discovery is an inherently reduc- rich media conditions.
tionist endeavour. Thus, an important premise is that the target is valid, In vivo essential genes represent a set of emerging targets that remain
and the weight of evidence to support a campaign against any given target untested in modern antibacterial drug discovery. However, they hold
can vary considerably. For example, although systematic surveys of gene great promise for expanding the pool of targets for new drugs. Inhibi-
dispensability in bacteria have illuminated a large number of potential tors of this class of targets often fail to show the standard phenotype of
targets, a substantial fraction of these genes encode proteins of unknown cell-growth inhibition, which is used to calculate the MIC. The MIC is
function. A clear understanding of function is commonly inherent in an important driver of compound optimization in preclinical antibi-
the process of target validation and — from a practical standpoint — it otic development and a gold standard for efficacy that is understood
is difficult to build a chemical screening assay for a protein of unknown by regulators, discoverers and practitioners alike. In the absence of a
function. We are coming to understand that dispensability phenotypes MIC, an assay that is amenable to high-throughput screening and down-
are contextual even for well-characterized protein targets, and that growth stream drug discovery needs to be built. One innovative solution involves
conditions and genomic context can greatly influence the dispensability screening in a non-conventional growth media. This approach was used
of the corresponding gene33. to target the glyoxylate shunt of P. aeruginosa because of its involve-
ment in pulmonary infection40. The screening workflow prioritized
In vivo essential genes compounds that were active in nutrient-limited media that contained
Approximately 7% of the E. coli genome, some 303 genes, has been acetate as the only source of carbon but inactive when glucose was the
shown to be essential for growth in rich media, which represents typi- only source, and led to the discovery of eight lead compounds. Another
cal Waksman-screen conditions24. Nevertheless, systematic studies of study built on a platform to discover small molecules that block growth
stress conditions such as nutrient deprivation and chemical perturba- solely under nutrient-limited conditions, and used it to target the bio-
tion have demonstrated that a further 258 genes are conditionally essen- synthesis of amino acids, nucleobases and vitamins41. After prioritizing
tial34,35. These conditionally essential genes largely encode enzymes that antibacterial compounds that were active only in the absence of nutri-
are important for the utilization of carbon sources or the biosynthesis ent supplements, the approach used systematic supplementation with
of the outer membrane as well as in the synthesis of amino acids, vita- individual and pools of metabolites to illuminate mechanisms of action.
mins and nucleobases. Yet these processes have long been ignored by Lead compounds that target glycine, folate and biotin synthesis in E. coli
antibacterial drug discovery. Indeed, comprehensive efforts to define were uncovered as a result. Research that targets the quorum-sensing
essential sets of bacterial genes have been performed only in vitro and, virulence pathway in the pathogen P. aeruginosa has generated promising
most extensively, using model microorganisms. Thus, the set of genes compounds that are active in mouse models of infection without per-
that is required for the viability of pathogens in the context of infection turbing growth in vitro42. Through an innovative screening platform, the
is less well understood. These in vivo essential genes have been assessed quorum-sensing system and expression of the toxic gene product SacB
in various pathogens using random transposon mutagenesis approaches were linked such that ‘hits’ in the screen led to cell viability. Another route
that, coincidentally, were invented while the first bacterial genomes were to the discovery of in vivo targets involves screening in host models of
being sequenced36. The methodology has revealed candidate roles for disease. For example, a high-content microscopic screen of macrophages
hundreds of genes in aspects of virulence or colonization, including the that were infected with M. tuberculosis identified a series of lead com-
bacterial stress response, nutrient biosynthesis, type III secretion and pounds that targeted the cytochrome bc1 complex in the bacterium43.
attachment functions that are mediated by pili, fimbriae or surface car- The essential-gene paradox reveals a fascinating interplay between
bohydrates37. Typically, these approaches assess large pools of mutants risk and the target set of genes that has consequences for industrial drug
and have been enabled by the introduction of massively parallel DNA discovery (Fig. 2). For instance, a pathogen might have a set of genes that
sequencing38. An intriguing application of this technology was an investi- is essential for growth on rich media and includes conventional targets
gation of the genomic requirements for growth of P. aeruginosa in sputum that are already under investigation. A greater innovation risk would be to

Table 1 | Unique chemical classes of antibiotics discovered in the golden era


Chemical class Target Mode of action Examples

Sulfonamides* Folate synthesis Bacteriostatic Sulfanilamide


β-Lactams† Cell-wall synthesis Bacteriocidal Penicillins
Cephalosporins
Carbapenems
Aminoglycosides† Protein synthesis Bacteriocidal Spectinomycin
Kanamycin
Neomycin
Tetracyclines† Protein synthesis Bacteriostatic Tetracycline
Doxycycline
Chloramphenicols† Protein synthesis Bacteriostatic Chloramphenicol
Macrolides† Protein synthesis Bacteriostatic Erythromycin
Clarithromycin
Glycopeptides† Cell-wall synthesis Bacteriocidal Vancomycin
Teicoplanin
Oxazolidinones* Protein synthesis Bacteriostatic Linezolid
Ansamycins† RNA synthesis Bacteriocidal Rifamycin
Quinolones† DNA synthesis Bacteriocidal Ciprofloxacin
Streptogramins† Protein synthesis Bacteriocidal Pristinamycin
*Synthetic chemical †Natural product..

3 3 8 | N AT U R E | VO L 5 2 9 | 2 1 JA N UA RY 2 0 1 6
© 2016 Macmillan Publishers Limited. All rights reserved
REVIEW INSIGHT

place the focus on the discovery of targets that are required for growth on
nutrient-limited media. Such targets are in vivo essential genes but would In vivo infection
also have in vitro phenotypes that facilitate conventional approaches to
antibiotic drug development. And an even greater innovation risk would 2 In vitro growth on
nutrient-limited media
be to focus on genes that are uniquely essential for infection. Studies so
far suggest that infection genes could represent the largest set of targets. In vitro growth on
nutrient-rich media
However, the lack of in vitro phenotypes, such as growth inhibition, means
3
that it can be a challenge to enable established methods of drug discovery 1

for such targets.

Genetic-interaction networks
As modern drug discovery has come of age, we have witnessed a trans-

innovation risk
formation in our understanding of the cell that is changing our view

Relative
of the druggable genome. Studies of gene and protein interactions in
yeast44,45 and bacteria46,47 have revealed a network of abundant func-
tional interactions. The classic maps of biochemical metabolism and
cell signalling are being supplanted by a network model of the cell that
is characterized by a highly connected web of proteins and genes that 1 2 3
Target set
exhibits remarkable complexity and redundancy. In the model micro-
organism S. cerevisiae, in which genome-scale experiments to create a Figure 2 | Target gene sets and innovation risks for a bacterial pathogen. 
matrix of pairwise deletions for all 5,000 dispensable genes are nearly A Venn diagram to show a relative profile of target genes for a hypothetical
complete, around 200,000 pairs of genes were revealed to be synthetic bacterial pathogen. Three potential target sets can be imagined. Each has
lethal in vitro44. These double-deletion strains suffered a substantial a relative innovation risk within the context of modern industrial drug
deficiency in fitness compared with the corresponding single-deletion discovery, which is indicated on the graph. By far the biggest set can be found
in the genes that are needed for infection in a relevant animal model, but
strains, for which the network revealed about 40 interactions per gene. much validation remains to be done for these targets. The set that is required
This is in contrast with the approximately 1,000 essential genes that for growth on nutrient-limited media is well charted. However, not all of
have been mapped for this yeast. Early studies of synthetic lethality in the genes in this set will be required for in vivo infection. Antibiotic drug
the model bacterium E. coli revealed a similar density of gene interac- discovery under nutrient-rich conditions has been the convention for many
tions47 and predicted a dramatically expanded body of targets for drugs years, and the corresponding set of target genes is the smallest.
or combinations of drugs that inhibit interacting gene products. Indeed,
these realities make a compelling case for combinatorial approaches to resistance. Therefore, emerging knowledge of bacterial cell systems is
new antibiotics. challenging the modern genes-to-drugs approach through the idea that
The contextual nature of gene essentiality is perhaps best demon- target-validation measures must capture the implications for the network
strated by synthetic-viable gene interactions. Here, the double mutant as a whole. An important example of this is the growing appreciation for
is considerably more fit than expected with respect to the fitness of the the role of metabolic states and reactive oxygen species in the effective-
corresponding single mutants. An example is the interaction of toxin ness of existing antibiotics58,59. Indeed, the inherent complexity of cell
and antitoxin genes that have a role in bacterial stress responses48. The systems is a compelling argument for the use of whole-cell, phenotype-
gene that encodes the antitoxin has an essential phenotype, but becomes based screens over biochemical, target-focused efforts in the discovery
dispensable in a genetic background that lacks the toxin gene. Another, of antibacterial compounds. Once a new potential compound has been
more remarkable, example of synthetic viability can be found in the wall identified, chemical-genomic characterization can provide important
teichoic acid (WTA) biosynthetic pathway of Gram-positive bacteria. information and predictions about its mechanism and resistance profile.
Although early steps in the pathway are dispensable, genes that encode Compounds that are discovered and characterized in this way therefore
late-step enzymes have an essential phenotype49. Curiously, the deletion provide a ready probe for target validation and a greater understanding
of an early gene renders any of the late genes dispensable. The essential of the network that underpins the target.
phenotype of the late genes is thought to arise from the accumulation
of WTA biosynthetic intermediates that are linked to undecaprenyl Combinatorial discovery
moieties and the sequestration of undecaprenyl phosphate from its The effectiveness of combinations of antibiotics has been demonstrated
role as a lipid carrier in the truly essential process of peptidoglycan- in the clinic and is gaining favour as a design strategy. An effective
wall assembly50. The unique dispensability patterns of WTA genes have pairing is trimethoprim and sulfamethoxazole, which have a spectacu-
provided the basis for sophisticated platforms for the discovery of new larly synergistic impact on folate metabolism. Another combination
lead compounds that target early and late steps in the WTA pathway in combines amoxicillin, a β-lactam antibiotic, with clavulanic acid, an
S. aureus 51–53 as well as the related pathway of undecaprenyl-phosphate inhibitor of β-lactamase drug resistance enzymes. Clinicians have
synthesis54. Primary and secondary screens for antagonism that are ena- long combined antibiotics from different classes, such as β-lactams
bled by synthetic-viable phenotypes facilitate the rapid elimination of with aminoglycosides, to achieve synergy, to cover a broad spectrum
nuisance compounds that often affect whole-cell screening campaigns. of pathogens when the infectious agent is unknown or to suppress the
By taking advantage of such knowledge, innovative screens for new anti- emergence of resistance. In fact, combinations are the mainstay of treat-
biotics can be developed. ment for stubborn infections such as tuberculosis. With the exception
of β-lactam–β-lactamase inhibitor pairs, existing antimicrobial drug
Chemical genomics combinations are largely the product of clinical experimentation with
Further examples of the complexity of bacterial viability are provided established antibiotics. However, a new model of discovery is emerging
by studies that map enhancers or suppressors of antibiotic action on a that recognizes the network view of the cell and searches for efficacious
genome scale55–57. Such efforts have revealed that antibiotics have dense combinations of small molecules in a systematic manner.
and complicated networks of chemical and genetic interactions that seem Platforms that aim to discover active combinations have almost exclu-
to be designed to resist perturbation (Fig. 3). These networks offer a rich sively searched for adjuvants. This is because the systematic examination
vista of new targets for antibiotic adjuvants, which are nonantibiotic of combinations, even in relatively small chemical collections, leads to
compounds that enhance the activity of antibiotics or help to overcome prohibitively large screens. Typically, an existing antibiotic is tested for

2 1 JA N UA RY 2 0 1 6 | VO L 5 2 9 | N AT U R E | 3 3 9
© 2016 Macmillan Publishers Limited. All rights reserved
INSIGHT REVIEW

Figure 3 | A chemical–genomic interaction


Genetic lethal interaction
network.  The network map illustrates the dense
connectivity of enhancing interactions that
Folate metabolism
affect the activity of 39 antibiotics that target the
bacterium Escherichia coli. Grey balls represent
Cell-wall biogenesis genes and coloured ones are for antibiotics of
various mechanistic classes. Interactions are
DNA replication denoted by connecting lines. The map is derived
from data in ref. 35 and was prepared with the
Protein translation BioLayout Express3D network visualization and
analysis tool83.
RNA synthesis

growth inhibition at subinhibitory concentrations in combination with a of legacy drugs, and because the delivery and utility of such drugs are
chemical collection. A screen for synthetic compounds that augment the familiar to clinicians, barriers to entry into clinical practice might be
activity of the narrow-spectrum Gram-positive antibiotic novobiocin in minimized. However, the combination approach also presents challenges.
the Gram-negative bacterium E. coli identified four new compounds60. These include the need to match the pharmacology and efficacy of each
They were found to affect cell shape and membrane permeability, which component drug, which adds to the burden of drug discovery. Resistance
provided an explanation for the observed synergies. Another combination is also a concern — as it is for any antimicrobial therapy. The potential for
screen that used natural product antibiotics aimed to reverse carbapenem unexpected drug–drug interactions must also be examined thoroughly.
resistance in metallo-β-lactamase-positive Gram-negative pathogens61. Nevertheless, multicomponent therapies, which are well established in
It found that aspergillomarasmine A could block the notorious carbap- treatment of cancer and of HIV, tuberculosis and bacterial infections —
enemase NDM-1 and was efficacious in a mouse model of infection by with combinations of β-lactam antibiotics and β-lactamase inhibitors,
NDM-1-containing Klebsiella pneumoniae. in particular — offer an alternative to traditional antibiotic discovery in
Drugs that have already been approved have attracted considerable the resistance era.
interest as sources of bioactive chemical matter for combination stud-
ies62. This stems from a growing understanding that small molecules with What chemical matter should be the focus?
proven therapeutic activity are privileged chemicals that bind to biologi- The history of antibiotic discovery suggests that natural products, espe-
cal polymers and often have potential for alternative uses. Compounds cially those produced by microorganisms, represent privileged chemical
that are used in the clinic have a proven track record in patients and are matter for the discovery of antibiotics. Because they are the result of natu-
accompanied by a deep record of study that offers a substantial edge over ral selection, these compounds have been proved to be highly effective
chemicals for which little is known. Several discovery campaigns that and are the source of most of our antibiotic medicines. Yet they have many
used such libraries have yielded exciting combinations of drugs. For drawbacks. Natural-product antibiotics are often chemically complex
example, the antidiarrhoeal drug loperamide was shown to destabilize with challenging and intractable routes to synthesis in the laboratory that
the membrane potential of bacterial cells, which facilitated the uptake of make it difficult to prepare derivatives. The pharmacology of most first-
the antibiotic minocycline in Gram-negative bacteria63. And the antihy- generation antibiotics is also not ideal, which reflects the origins of these
pertensive drug ticlopidine was found to strengthen the action of β-lactam drugs as specialized microbial metabolites not medicines. Furthermore,
antibiotics against methicillin-resistant S. aureus (MRSA) by inhibiting because these ancient metabolites are important components of microbial
the synthesis of WTA53. chemical ecology, genetic elements that provide resistance against them
Combinatorial approaches are consistent with an increased under- have been retained by and are often widely dispersed in communities of
standing that the most successful antibiotics seem to interact with microorganisms in the environment64,65. Finally, although screens that use
more than one target30. Furthermore, such approaches are aligned the Waksman platform can readily detect antimicrobial metabolites from
with the network view of bacteria and their resistome, which resides libraries of environmental microbes, most such metabolites have already
within the cell to buffer against perturbation. Nevertheless, they have been identified and catalogued. More than 20,000 microbial natural
received relatively little attention from industry. Indeed, modern products have been described over the past several decades66. Waksman-
target-based discovery approaches seek overwhelmingly to achieve platform screens of typical libraries of natural products that are derived
exquisite target selectivity to minimize toxicity that might arise from from the actinomycetes group of bacteria, a favoured source of antibiot-
drug promiscuity. ics, tend to result in the identification of known compounds67. A further
The adjuvant approach offers advantages such as the potential to complication is that the collection of libraries of natural-product produc-
decrease the concentration of antibiotic that is needed to achieve a thera- ing microorganisms, the generation of compound extracts for screening
peutic effect, which should reduce the emergence of resistance and lessen and the purification and characterization of active compounds is very
issues of off-target toxicity. Combinations can extend the effective lifespan labour intensive and costly. Overall, this has resulted in the abandonment

3 4 0 | N AT U R E | VO L 5 2 9 | 2 1 JA N UA RY 2 0 1 6
© 2016 Macmillan Publishers Limited. All rights reserved
REVIEW INSIGHT

of microbial natural products by most large pharmaceutical companies. the ability of synthetic compounds to inhibit essential bacterial targets
However, libraries of synthetic chemicals can exceed several million by developing delivery systems that solve the cell-envelope penetration
compounds and, in principle, provide access to a vast spectrum of chemi- and efflux challenges. Conjugation to siderophores is an example of such
cal space. By definition, they are chemically tractable, which enables their a ‘Trojan horse’ approach that has been explored but not yet proved to be
scale up and relatively facile expansion by medicinal chemistry to explore clinically efficacious73. A further solution is to combine these compounds
structure–activity relationships, improve efficacy and optimize pharma- with molecules that enhance their transport or otherwise help to breach
cology. Indeed, one of the most successful classes of antibiotics over the the permeability barrier of the bacterial cell. In-depth study of the bacte-
past four decades has been the synthetic fluoroquinolones. Synthetic rial cell envelope and the development of rules to guide the synthesis of
libraries are the mainstay of most pharmaceutical companies and are the potential antibiotics should enhance the ability of such compounds to
source of many medicines that are crucial for human health, including penetrate cells and avoid efflux.
inhibitors of protein kinases, G-protein-coupled receptors and ion chan- The crisis in antibiotic discovery is prompting a return to the inves-
nels. Although these collections have been explored as sources of new tigation of natural products. Known chemical scaffolds that were aban-
antibiotics using modern drug-discovery platforms for more than three doned in the past are being revisited with some success. Daptomycin,
decades, they have yet to deliver new medicines — partly because they are for example, is now being used to treat serious infections caused by
sampling such a narrow area of chemical space. Driven by metrics such as Gram-positive pathogens. Eli Lilly and Company had discarded the
the Lipinski rule of five for orally available drugs68, the synthetic libraries compound in the 1980s owing to toxic adverse effects that were iden-
of most pharmaceutical companies tend to be dominated by compounds tified during clinical trials. But in the late 1990s, Cubist Pharmaceu-
that are optimized for human biology. But most antibiotics in clinical use ticals licensed the compound and were able to overcome the adverse
do not conform to these parameters69 because they are natural products or effects by altering the formulation and dosage. The US Food and Drug
their derivatives. The Achilles’ heel of synthetic compounds as new anti- Administration (FDA) approved the drug in 2003 (ref. 74), and by
biotic drugs seems to be their general inability to penetrate readily the cell 2014, annual sales of daptomycin had exceeded US$1 billion. Similarly,
envelope of bacteria, which comprises inner and outer membranes, porins the poorly absorbed antibiotic fidaxomicin, which was discovered in
and complex carbohydrate polymers, coupled with their susceptibility the 1970s at Hoechst Marion Roussel, saw no clinical use until infec-
to active efflux by membrane-associated pumps. Because they are prod- tions associated with the gut bacterium Clostridium difficile started to
ucts of evolution, antibiotic natural products have evolved to overcome rise around a decade ago75. Fidaxomicin’s shortcomings as a broad-
these challenges. The overall result is a stalemate in discovery: screens of spectrum, orally available antibiotic were revisited as strengths in an
natural-product libraries identify bioactive but known compounds, and agent that targets C. difficile by taking advantage of high concentra-
screens of synthetic libraries identify potent ligands of biochemical targets tions of the drug in faeces, and fidaxomicin was approved for use by
but with poor bioactivity. the FDA in 2011.
A solution to this impasse is the development of synthetic libraries Over the past 20 years, we have taken amazing strides in our under-
that capture the chemical diversity and physicochemical properties of standing of the mechanisms and genomics of microbial natural-product
natural products. These are being pursued for a number of therapeutic biosynthesis. Genome sequencing of microorganisms has revealed a
classes70, including antibiotics71,72. Another solution is to capitalize on spectrum of potential for specialized metabolite production that greatly

BOX 1
Hurdles and solutions for antibiotic discovery
If not for the emergence of resistance to existing antibiotics, these need to adapt. In fact, promising new strategies are already being
medicines might be thought of as ‘perfect’ therapeutics with an used in the search for combinations of compounds that will target
extraordinary record of success in the clinic. Thus, the bar is set high cell networks in ways that are reminiscent of the complex action of
for the next generation of these medicines. Here are four hurdles and existing antibiotics.
solutions to the antibiotic-discovery problem. ●●Physical and chemical properties of existing antibiotics are
not compatible with conventional collections of compounds and
●●Resistance has arisen partly because only a narrow selection medicinal-chemistry approaches. There is great need for research
of chemical compounds are available, and they have a limited into the antibiotic permeability barriers of bacteria, particularly of
range of mechanisms. Genomics-driven efforts to exploit new Gram-negative organisms, and the physical properties of chemicals
potential targets, however, have been unsuccessful. New targets that can overcome these barriers. Those who have a role in drug
and approaches to antibiotic drug discovery are fraught with risk. discovery must look critically at existing compound collections and
Thus, there is a substantial need for fundamental research to assess medicinal chemistry strategies within this context. Natural
mitigate them. Academic research on unconventional targets and sources of chemical compounds have been eschewed in past decades
discovery platforms has made much progress in past decades. in favour of synthetic chemicals that are better suited to conventional
However, most resources in new antibiotic drug discovery have been models of drug discovery. It is time to revisit natural products as an
invested conservatively in conventional targets and approaches. With alternative to synthetic collections and to embrace new technologies
growing potential for unconventional targets and new approaches that will help to overcome problems of compound rediscovery.
to antibiotic drug discovery, there is great opportunity for successful ●●The yardstick for early stage antibiotic discovery is the inhibition
efforts to industrialize these platforms. of cell growth and the determination of the MIC. The ease of
●●The complex mechanisms of action of existing antibiotics could identifying antibiotics through the Waksman platform, and its
be crucial to their success and elusive to modern drug-discovery success in the golden era, solidified a protocol that narrowly focused
platforms. The solution is to understand these mechanisms better. success on this simple laboratory test. The realization that numerous
Biology is complex and our understanding of the action of antibiotics targets, such as virulence factors and aspects of metabolism, are not
is incomplete. Research that uses both traditional and systems captured by the traditional MIC approach requires innovation in the
approaches will be required to expand our knowledge and, if this laboratory and an acceptance that twenty-first century antibiotics
hypothesis proves to be true, our approaches to drug discovery will might not have the same properties as existing drugs.

2 1 JA N UA RY 2 0 1 6 | VO L 5 2 9 | N AT U R E | 3 4 1
© 2016 Macmillan Publishers Limited. All rights reserved
INSIGHT REVIEW

exceeds the diversity of compounds that have been purified and char- microbiome and decrease the pressure to evolve resistance. Such narrow-
acterized over the past century. Automated prediction of the structure spectrum agents can be identified by mining the catalogue of discarded
and regulation of compounds from genome data is improving rapidly. antibiotics — as evidenced by daptomycin — and through systematic
The manipulation of products with techniques from synthetic biology is testing of combinations of compounds that selectively target genus- and
becoming more common, and offers an unprecedented opportunity for even species-specific pathways53,63. The switch to narrow-spectrum agents
tapping into the bioactive chemical space. Techniques such as metagen- should also expand the target base of antibiotics. Antibiotic discovery
omics and use of specialized growth chambers are enabling exploration of is considered to be target poor, with the drugs limited to targeting cell-
the microbial dark matter — microorganisms that have resisted easy cul- wall synthesis, membrane integrity, translation, transcription and DNA
tivation in the laboratory76 — as well as identification of new chemistry77 synthesis. This might simply reflect common targets that are available
and fresh leads for antibiotic drugs78. to broad-spectrum agents. Narrow-spectrum agents offer a much larger
Of the thousands of natural-product antibiotics that have been identi- vista of targets that are specific to bacteria and pathogenesis. For example,
fied over the past decades, only a select few have been seriously investi- many of the processes that enable in vivo growth and virulence are targets
gated as drugs. The examples that we describe in this Review suggest that for such antibiotics.
it would be fruitful to revisit some of these old scaffolds and to search for Because of time pressures and a lack of sensitive and reliable diagnostic
new ones using twenty-first century technology through the lens of the agents, clinicians most often favour broad-spectrum agents. Advances
clinical needs of the resistance era. Large-scale screens will be needed to in the accuracy of point-of-care diagnostics will be necessary to move
identify these candidate compounds. At present, no collection of micro- forward the field of antibiotic discovery. And the business case for narrow-
bial natural products adequately reflects the chemical diversity that has spectrum agents must address the consequences of lower sales volumes81.
already been reported in the literature, let alone that which is yet to be However, a spate of prizes for the development of point-of-care diagnos-
discovered. Indeed, many ‘old’ compounds are essentially lost to modern tics, such as the UK Longitude Prize that seeks to tackle global antibiotic
drug discovery because there is no supply of them available. A multi- resistance, together with stunning advances in genome sequencing and
national effort to develop a truly representative library of antimicrobial informatics tools that can identify pathogens and their resistance profiles82
natural products that are readily available to antibiotic researchers would are creating the conditions necessary to support the discovery of narrow-
be transformative. spectrum antibiotics and their implementation through improved diag-
nostics. The narrow-spectrum era requires innovation, new economic
The future models and collaboration across disciplines. Despite these challenges,
Ultimately, clinical need and the marketplace will dictate the product pro- the field of infectious-disease medicine can learn from other therapeutic
file of new antibiotics. The clinical need for such drugs is growing, and areas to move from broad-spectrum empirical therapy that is no longer
in some cases, such as multidrug-resistant Gram-negative pathogens, we effective in the resistance era to the personalized and strategic approach
are already experiencing a profound crisis. In the resistance era, we must of the emerging narrow-spectrum era. ■
accept and tackle this global challenge to continue to enjoy the benefits
Received 25 August; accepted 16 November 2015.
of antimicrobial agents and the improvements in quality of life and life
expectancy that they bring. Although considerable, the regulatory chal- 1. Fleming, A. On the antibacterial action of cultures of a penicillium, with special
lenges of developing new antibiotics are being slowly addressed to stream- reference to their use in the isolation of B. influenzae. Br. J. Exp. Pathol. 10,
226–236 (1929).
line approvals for drugs. For example, the FDA approved a combination 2. Comroe, J. H. Jr. Pay dirt: the story of streptomycin. Part I. From Waksman to
treatment that contains ceftazidime and avibactam on the basis of data Waksman. Am. Rev. Respir. Dis. 117, 773–781 (1978).
gathered in phase II clinical trials, therefore avoiding the need for phase 3. Wright, G. D. The antibiotic resistome: the nexus of chemical and genetic
diversity. Nature Rev. Microbiol. 5, 175–186 (2007).
III data. And the economic hurdles associated with drugs that cure rather 4. Perry, J. A., Westman, E. L. & Wright, G. D. The antibiotic resistome: what’s new?
than control diseases are being addressed by proposals such as the GAIN Curr. Opin. Microbiol. 21, 45–50 (2014).
(Generating Antibiotics Incentives Now) act in the United States, which 5. Forsberg, K. J. et al. Bacterial phylogeny structures soil resistomes across
habitats. Nature 509, 612–616 (2014).
seeks to extend market exclusivity for new antibiotics79, public–private 6. Nesme, J. & Simonet, P. The soil resistome: a critical review on antibiotic
partnerships such as the Innovative Medicines Initiative in Europe, and resistance origins, ecology and dissemination potential in telluric bacteria.
by incentives such as financial rewards for companies that successfully Environ. Microbiol. 17, 913–930 (2015).
launch new antimicrobial drugs80. 7. Finley, R. L. et al. The scourge of antibiotic resistance: the important role of the
environment. Clin. Infect. Dis. 57, 704–710 (2013).
The scientific challenges in identifying and developing new antibiotic 8. Forsberg, K. J. et al. The shared antibiotic resistome of soil bacteria and human
drugs remain substantial (Box 1). It is clear from the past two decades of pathogens. Science 337, 1107–1111 (2012).
effort that antibiotics that are highly effective, safe and broad spectrum 9. D’Costa, V. M. et al. Antibiotic resistance is ancient. Nature 477, 457–461 (2011).
This paper was the first to provide physical evidence of the ancient resistome,
are incredibly difficult to find. Consequently, we must ask whether our in 30,000-year-old permafrost.
current medical practices and ideas about infection are appropriate in 10. Andersson, D. I. & Hughes, D. Persistence of antibiotic resistance in bacterial
the resistance era. The availability and properties of the antibiotics of the populations. FEMS Microbiol. Rev. 35, 901–911 (2011).
11. Cox, G. & Wright, G. D. Intrinsic antibiotic resistance: mechanisms, origins,
golden and medicinal-chemistry eras radically changed the practice of challenges and solutions. Int. J. Med. Microbiol. 303, 287–292 (2013).
infection control and treatment. For example, the efficacy of antibiotics 12. Fajardo, A. et al. The neglected intrinsic resistome of bacterial pathogens. PLoS
enabled empirical therapy and prophylaxis. And broad-spectrum anti- ONE 3, e1619 (2008).
An introduction to the concept of the intrinsic resistome, which is a target for
biotics facilitated the practice of ‘best guess’ medicine and of overpre- antibiotic adjuvants.
scription. But we no longer have the luxury of empirical therapy, and 13. Abraham, E. P. & Chain, E. An enzyme from bacteria able to destroy penicillin.
infectious-disease medicine must adopt twenty-first century innova- Nature 146, 837 (1940).
14. Davies, J. & Davies, D. Origins and evolution of antibiotic resistance. Microbiol.
tions. Just as oncology has been transformed by fundamental research Mol. Biol. Rev. 74, 417–433 (2010).
and the realization that cancer encompasses many diseases that require 15. Fischbach, M. A. & Walsh, C. T. Antibiotics for emerging pathogens. Science 325,
individualized therapies, so too must infection research. This requires a 1089–1093 (2009).
much greater understanding of the specific microorganisms that cause 16. Lewis, K. Antibiotics: recover the lost art of drug discovery. Nature 485, 439–440
(2012).
disease, their biology and their interactions with the host. As oncology 17. Cho, H., Uehara, T. & Bernhardt, T. G. β-Lactam antibiotics induce a lethal
has done already, the treatment of infection should adopt the model of malfunctioning of the bacterial cell wall synthesis machinery. Cell 159,
personalized medicine, which would move the focus of the field from 1300–1311 (2014).
This paper provides evidence for the complexity of antibiotic-induced cell death.
broad-spectrum agents to narrow, disease-specific antibiotics. The result- 18. Wilson, D. N. Ribosome-targeting antibiotics and mechanisms of bacterial
ing narrow-spectrum era (Fig. 1) would have considerable impact on the resistance. Nature Rev. Microbiol. 12, 35–48 (2014).
next generation of antibiotics and generate fewer off-target effects on the 19. Demirci, H. et al. A structural basis for streptomycin-induced misreading of the

3 4 2 | N AT U R E | VO L 5 2 9 | 2 1 JA N UA RY 2 0 1 6
© 2016 Macmillan Publishers Limited. All rights reserved
REVIEW INSIGHT

genetic code. Nature Commun. 4, 1355 (2013). biogenesis uncovers an inhibitor of undecaprenyl diphosphate synthase. Proc.
20. Dwyer, D. J., Collins, J. J. & Walker, G. C. Unraveling the physiological complexities Natl Acad. Sci. USA 112, 11048–11053 (2015).
of antibiotic lethality. Annu. Rev. Pharmacol. Toxicol. 55, 313–332 (2015). 55. Liu, A. et al. Antibiotic sensitivity profiles determined with an Escherichia coli
21. Macarron, R. et al. Impact of high-throughput screening in biomedical research. gene knockout collection: generating an antibiotic bar code. Antimicrob. Agents
Nature Rev. Drug Discov. 10, 188–195 (2011). Chemother. 54, 1393–1403 (2010).
22. Fleischmann, R. D. et al. Whole-genome random sequencing and assembly of 56. Tan, C. M. et al. Restoring methicillin-resistant Staphylococcus aureus
Haemophilus influenzae Rd. Science 269, 496–512 (1995). susceptibility to β-lactam antibiotics. Sci. Transl. Med. 4, 126ra35 (2012).
23. Kobayashi, K. et al. Essential Bacillus subtilis genes. Proc. Natl Acad. Sci. USA 100, 57. Pathania, R. et al. Chemical genomics in Escherichia coli identifies an inhibitor of
4678–4683 (2003). bacterial lipoprotein targeting. Nature Chem. Biol. 5, 849–856 (2009).
24. Baba, T. et al. Construction of Escherichia coli K-12 in-frame, single-gene 58. Brynildsen, M. P., Winkler, J. A., Spina, C. S., Macdonald, I. C. & Collins, J. J.
knockout mutants: the Keio collection. Mol. Syst. Biol. 2, 2006.0008 (2006). Potentiating antibacterial activity by predictably enhancing endogenous
25. Forsyth, R. A. et al. A genome-wide strategy for the identification of essential microbial ROS production. Nature Biotechnol. 31, 160–165 (2013).
genes in Staphylococcus aureus. Mol. Microbiol. 43, 1387–1400 (2002). 59. Dwyer, D. J. et al. Antibiotics induce redox-related physiological alterations as
26. Jacobs, M. A. et al. Comprehensive transposon mutant library of Pseudomonas part of their lethality. Proc. Natl Acad. Sci. USA 111, E2100–E2109 (2014).
aeruginosa. Proc. Natl Acad. Sci. USA 100, 14339–14344 (2003). 60. Taylor, P. L., Rossi, L., De Pascale, G. & Wright, G. D. A forward chemical screen
27. Jain, P. et al. Specialized transduction designed for precise high-throughput identifies antibiotic adjuvants in Escherichia coli. ACS Chem. Biol. 7, 1547–1555
unmarked deletions in Mycobacterium tuberculosis. mBio 5, e01245-14 (2012).
(2014). 61. King, A. M. et al. Aspergillomarasmine A overcomes metallo-β-lactamase
28. Brown, E. D. & Wright, G. D. New targets and screening approaches in antibiotic resistance. Nature 510, 503–506 (2014).
antimicrobial drug discovery. Chem. Rev. 105, 759–774 (2005). 62. Borisy, A. A. et al. Systematic discovery of multicomponent therapeutics. Proc.
29. Payne, D. J., Gwynn, M. N., Holmes, D. J. & Pompliano, D. L. Drugs for bad bugs: Natl Acad. Sci. USA 100, 7977–7982 (2003).
confronting the challenges of antibacterial discovery. Nature Rev. Drug Discov. 6, 63. Ejim, L. et al. Combinations of antibiotics and nonantibiotic drugs enhance
29–40 (2007). antimicrobial efficacy. Nature Chem. Biol. 7, 348–350 (2011).
30. Silver, L. L. Challenges of antibacterial discovery. Clin. Microbiol. Rev. 24, 71–109 The first demonstration of a systematic screen for antibiotic adjuvants.
(2011). 64. D’Costa, V. M., McGrann, K. M., Hughes, D. W. & Wright, G. D. Sampling the
31. Tommasi, R., Brown, D. G., Walkup, G. K., Manchester, J. I. & Miller, A. A. antibiotic resistome. Science 311, 374–377 (2006).
ESKAPEing the labyrinth of antibacterial discovery. Nature Rev. Drug Discov. 14, 65. Bhullar, K. et al. Antibiotic resistance is prevalent in an isolated cave microbiome.
529–542 (2015). PLoS ONE 7, e34953 (2012).
An analysis of the challenges and failures of antibiotic discovery in ‘big pharma’. 66. Bérdy, J. Bioactive microbial metabolites. J. Antibiot. 58, 1–26 (2005).
32. Farha, M. A. & Brown, E. D. Unconventional screening approaches for antibiotic 67. Baltz, R. H. Marcel Faber Roundtable: is our antibiotic pipeline unproductive
discovery. Ann. NY Acad. Sci. 1354, 54–66 (2015). because of starvation, constipation or lack of inspiration? J. Ind. Microbiol.
33. D’Elia, M. A., Pereira, M. P. & Brown, E. D. Are essential genes really essential? Biotechnol. 33, 507–513 (2006).
Trends Microbiol. 17, 433–438 (2009). 68. Lipinski, C. A., Lombardo, F., Dominy, B. W. & Feeney, P. J. Experimental and
34. Joyce, A. R. et al. Experimental and computational assessment of conditionally computational approaches to estimate solubility and permeability in drug
essential genes in Escherichia coli. J. Bacteriol. 188, 8259–8271 (2006). discovery and development settings. Adv. Drug Deliv. Rev. 46, 3–26 (2001).
35. Nichols, R. J. et al. Phenotypic landscape of a bacterial cell. Cell 144, 143–156 69. O’Shea, R. & Moser, H. E. Physicochemical properties of antibacterial
(2011). compounds: implications for drug discovery. J. Med. Chem. 51, 2871–2878
36. Hensel, M., Shea, J. E., Gleeson, C., Jones, M. D. & Dalton, E. Simultaneous (2008).
identification of bacterial virulence genes by negative selection. Science 269, 70. Kim, J., Kim, H. & Park, S. B. Privileged structures: efficient chemical ‘navigators’
400–403 (1995). toward unexplored biologically relevant chemical spaces. J. Am. Chem. Soc. 136,
37. Autret, N. & Charbit, A. Lessons from signature-tagged mutagenesis on the 14629–14638 (2014).
infectious mechanisms of pathogenic bacteria. FEMS Microbiol. Rev. 29, 71. Galloway, W. R., Bender, A., Welch, M. & Spring, D. R. The discovery of
703–717 (2005). antibacterial agents using diversity-oriented synthesis. Chem. Commun. 2009,
38. van Opijnen, T. & Camilli, A. Transposon insertion sequencing: a new tool for 2446–2462 (2009).
systems-level analysis of microorganisms. Nature Rev. Microbiol. 11, 435–442 72. Rachakonda, V., Alla, M., Kotipalli, S. S. & Ummanni, R. Design, diversity-oriented
(2013). synthesis and structure activity relationship studies of quinolinyl heterocycles as
39. Turner, K. H., Wessel, A. K., Palmer, G. C., Murray, J. L. & Whiteley, M. Essential antimycobacterial agents. Eur. J. Med. Chem. 70, 536–547 (2013).
genome of Pseudomonas aeruginosa in cystic fibrosis sputum. Proc. Natl Acad. 73. Han, S., Zaniewski, R. P. & Marr, E. S. Structural basis for effectiveness of
Sci. USA 112, 4110–4115 (2015). siderophore-conjugated monocarbams against clinically relevant strains of
40. Fahnoe, K. C. et al. Non-traditional antibacterial screening approaches for Pseudomonas aeruginosa. Proc. Natl Acad. Sci. USA 107, 22002–22007 (2010).
the identification of novel inhibitors of the glyoxylate shunt in Gram-negative 74. Eisenstein, B. I., Oleson, F. B. & Baltz, R. H. Daptomycin: from the mountain to the
pathogens. PLoS ONE 7, e51732 (2012). clinic, with essential help from Francis Tally, MD. Clin. Infect. Dis. 50 (suppl. 1),
41. Zlitni, S., Ferruccio, L. F. & Brown, E. D. Metabolic suppression identifies new S10–S15 (2010).
antibacterial inhibitors under nutrient limitation. Nature Chem. Biol. 9, 796–804 75. Gerber, M. & Ackermann, G. OPT-80, a macrocyclic antimicrobial agent for the
(2013). treatment of Clostridium difficile infections: a review. Expert Opin. Investig. Drugs
This paper describes a systematic screening approach for identifying 17, 547–553 (2008).
antibacterial antimetabolites. 76. Lok, C. Mining the microbial dark matter. Nature 522, 270–273 (2015).
42. Starkey, M. et al. Identification of anti-virulence compounds that disrupt 77. Wilson, M. C. et al. An environmental bacterial taxon with a large and distinct
quorum-sensing regulated acute and persistent pathogenicity. PLoS Pathog. 10, metabolic repertoire. Nature 506, 58–62 (2014).
e1004321 (2014). 78. Ling, L. L. et al. A new antibiotic kills pathogens without detectable resistance.
43. Pethe, K. et al. Discovery of Q203, a potent clinical candidate for the treatment of Nature 517, 455–459 (2015).
tuberculosis. Nature Med. 19, 1157–1160 (2013). 79. Brown, E. D. Is the GAIN Act a turning point in new antibiotic discovery? Can. J.
44. Costanzo, M. et al. The genetic landscape of a cell. Science 327, 425–431 Microbiol. 59, 153–156 (2013).
(2010). 80. Spellberg, B., Bartlett, J., Wunderink, R. & Gilbert, D. N. Novel approaches are
45. Ho, Y. et al. Systematic identification of protein complexes in Saccharomyces needed to develop tomorrow’s antibacterial therapies. Am. J. Respir. Crit. Care
cerevisiae by mass spectrometry. Nature 415, 180–183 (2002). Med. 191, 135–140 (2015).
46. Butland, G. et al. Interaction network containing conserved and essential protein 81. Spellberg, B. The future of antibiotics. Crit. Care 18, 228 (2014).
complexes in Escherichia coli. Nature 433, 531–537 (2005). 82. McArthur, A. G. & Wright, G. D. Bioinformatics of antimicrobial resistance in the
47. Babu, M. et al. Genetic interaction maps in Escherichia coli reveal functional age of molecular epidemiology. Curr. Opin. Microbiol. 27, 45–50 (2015).
crosstalk among cell envelope biogenesis pathways. PLoS Genet. 7, e1002377 83. Theocharidis, A., van Dongen, S., Enright, A. J. & Freeman, T. C. Network
(2011). visualization and analysis of gene expression data using BioLayout Express3D.
48. Gerdes, K., Christensen, S. K. & Løbner-Olesen, A. Prokaryotic toxin–antitoxin Nature Protoc. 4, 1535–1550 (2009).
stress response loci. Nature Rev. Microbiol. 3, 371–382 (2005).
49. D’Elia, M. A. et al. Lesions in teichoic acid biosynthesis in Staphylococcus Acknowledgements The authors thank M. Farha for stimulating discussion with
aureus lead to a lethal gain of function in the otherwise dispensable pathway. regards to the manuscript and S. French for creating Fig. 3. This work was supported
J. Bacteriol. 188, 4183–4189 (2006). by Discovery grants from the Natural Sciences and Engineering Research Council
50. D’Elia, M. A. et al. Probing teichoic acid genetics with bioactive molecules reveals of Canada to E.D.B. (RGPIN 04384-2014) and G.D.W. (RGPIN 237480), by salary
new interactions among diverse processes in bacterial cell wall biogenesis. awards from the Canada Research Chairs Program to both E.D.B. and G.D.W., and by
Chem. Biol. 16, 548–556 (2009). grants from the Canadian Institutes of Health Research to E.D.B. (MOP-81330 and
51. Swoboda, J. G. et al. Discovery of a small molecule that blocks wall teichoic acid MOP-15496) and G.D.W. (MT-13536).
biosynthesis in Staphylococcus aureus. ACS Chem. Biol. 4, 875–883 (2009).
52. Wang, H. et al. Discovery of wall teichoic acid inhibitors as potential anti-MRSA Author Information Reprints and permissions information is available at
β-lactam combination agents. Chem. Biol. 20, 272–284 (2013). www.nature.com/reprints. The authors declare no competing interests. Readers are
53. Farha, M. A. et al. Inhibition of WTA synthesis blocks the cooperative action of welcome to comment on the online version of this paper at go.nature.com/srti9i.
PBPs and sensitizes MRSA to β-lactams. ACS Chem. Biol. 8, 226–233 (2013). Correspondence should be addressed to E.D.B. (ebrown@mcmaster.ca) or G.D.W.
54. Farha, M. A. et al. Antagonism screen for inhibitors of bacterial cell wall (wrightge@mcmaster.ca).

2 1 JA N UA RY 2 0 1 6 | VO L 5 2 9 | N AT U R E | 3 4 3
© 2016 Macmillan Publishers Limited. All rights reserved

You might also like