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

Natural Product

Reports
View Article Online
VIEWPOINT View Journal | View Issue

Opportunities for natural products in 21st century


antibiotic discovery
Published on 01 June 2017. Downloaded by Cornell University Library on 12/07/2017 22:19:04.

Cite this: Nat. Prod. Rep., 2017, 34, 694

Gerard D. Wright

Natural products and their derivatives are mainstays of our antibiotic drugs, but they are increasingly in peril.
The combination of widespread multidrug resistance in once susceptible bacterial pathogens,
disenchantment with natural products as sources of new drugs, lack of success using synthetic
compounds and target-based discovery methods, along with shifting economic and regulatory issues,
conspire to move investment in research and development away from the antibiotics arena. The result is
a growing crisis in antibiotic drug discovery that threatens modern medicine. 21st century natural
product research is perfectly positioned to fill the antibiotic discovery gap and bring new drug
candidates to the clinic. Innovations in genomics and techniques to explore new sources of
antimicrobial chemical matter are revealing new chemistry. Increasing appreciation of the value of
narrow-spectrum drugs and re-examination of once discarded chemical scaffolds coupled with
synthetic biology methods to generate new compounds and improve yields offer new strategies to
revitalize once moribund natural product programs. The increasing awareness that the combination of
antibiotics with adjuvants, non-antibiotic compounds that overcome resistance and enhance drug
Received 17th March 2017
activity, can rescue older chemical scaffolds, and concepts such as blocking pathogen virulence present
orthogonal strategies to traditional antibiotics. In all these areas, natural products offer chemical matter,
DOI: 10.1039/c7np00019g
shaped by natural selection, that is privileged in this therapeutic area. Natural product research is poised
rsc.li/npr to regain prominence in delivering new drugs to solve the antibiotic crisis.

medical procedures including immune-depleting cancer


1. The antibiotics crisis chemotherapy, organ transplantation, heart surgery, and the
Microbially-derived natural products occupy a storied place in replacement of skeletal joints, are now commonplace because
antibiotic drug discovery and development. The rst wave of of the infection control provided by antibiotics. It is fair to say
antibiotics that emerging before the 1940s consisted of that together with anesthetics that control pain and enable
synthetic compounds such as the arsenical salvarsan and the radical surgical interventions, antibiotics are the foundation
sulfonamides. However, the bulk of the antimicrobials that drugs of modern medicine, forever changing what we expect
form the basis of our current antibiotic drug arsenal arose from from medical practice. Antibiotics have a massive impact on
studies of the secondary metabolites produced by microbes. both health and society. Yet we are in increasing danger of
Fleming’s discovery in 1928 of the bactericidal activity of the losing them.
fungal metabolite penicillin, followed by Dubos’s exploration of In his Nobel address in 1945, Fleming warned of the
antimicrobial peptides produced by bacilli and augmented by potential for the emergence of bacterial resistance to under-
Waksman’s mining of the compounds produced by soil acti- mine the efficacy of the newly discovered penicillin.2 Now, 70
nomycetes, ushered in the Golden Age of antibiotic discovery.1 years later, Fleming’s warning proved prescient: once manage-
The turning point in this history was the development of able infections are increasingly difficult to control due to the
penicillin into a ‘miracle drug’ during the Second World War. emergence of resistance. In a phenomenon not predicted by
The next three decades witnessed the discovery of the majority Fleming, we now know that bacteria can collect many genetic
of antibiotic scaffolds that are in clinical use today. elements that confer high-level resistance to several antibiotics
These natural product antibiotics irrevocably changed resulting in multidrug resistance (MDR) phenotypes.3 Para-
medicine. Feared infections that plagued humanity for doxically, despite a growing clinical need, the pharmaceutical
millennia were now controllable diseases. Once unthinkable industry, which was once at the forefront of infectious disease
research and development, has largely abandoned antibiotics.
Instead, efforts are channeled towards more tractable chronic
M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry diseases that have more favorable prospects for a return on
and Biomedical Sciences, DeGroote School of Medicine, McMaster University, 1280 investment, for example diabetes and cardiovascular disease.
Main Street West, Hamilton, ON L8N 4K1, Canada. E-mail: wrightge@mcmaster.ca

694 | Nat. Prod. Rep., 2017, 34, 694–701 This journal is © The Royal Society of Chemistry 2017
View Article Online

Viewpoint Natural Product Reports

The result is a growing innovation gap that, if unsolved, will toxic antibiotic that can be used to treat severe MDR infections
return us to the pre-antibiotic era where the fruits of modern may never be approved or invested in because of the historical
medicine are out of reach for many. Indeed, as seen by recent efficacy and safety margin of drugs like penicillin. These issues
events where patients succumb to once treatable pathogens that bring to the fore the difficulty that clinicians face in that, for the
are now resistant to all available antibiotics, we are already in most part, the causative agent of the infection is not known. The
a grave situation.4–6 lack of molecular diagnostics that accurately and rapidly iden-
In many ways, the remarkable success of natural product tify the pathogen to guide clinicians and clinical trials is a major
antibiotics has contributed to the current problem. First, while confounding problem. Addressing the antibiotics crisis will
it is certain that MDR pathogens are on the rise all across the take a concerted effort across many aspects of society and areas
globe, in particular in hospitals and other health care facilities, of research.8 Natural products must continue to play a funda-
Published on 01 June 2017. Downloaded by Cornell University Library on 12/07/2017 22:19:04.

our legacy antibiotics oen remain quite useful. This is espe- mental role in solving this global problem.
cially true for community-acquired infections, though MDR is
on the rise here as well. These old drugs are cheap and non- 2. The impact of natural products in
toxic. This is good for patients, health care providers, and
funders. That said, complacent reliance on these drugs fails to
antibiotic discovery
anticipate the inevitable – that they are vulnerable to resistance Natural products of microbial origin are privileged in the sphere
and that we need replacements and new ways of dealing with of antibiotic development. The sulfa drugs, trimethoprim, u-
infection. oroquinolones, and oxazolidinones (Fig. 1) are examples of
Second, the efficacy and safety of our legacy antibiotics highly successful synthetic compounds in clinical antibiotic use
provide demanding benchmarks to be met. Antibiotics are and provide proof that these should not be abandoned in our
frequently given on the gram scale, orders of magnitude higher search for 21st century antibiotics. Nevertheless, the majority of
than other drugs. Toxicity vs. efficacy ratios are consequently antibiotic drugs are derived from microbial natural products.
tough to match for new compounds. This also brings into Decades of activity-guided purication of antibiotics from
question the difficulty of clinical trials for new antibiotics.7 microbial sources has identied an estimated 28 000
Placebo-controlled trials are unethical in the antibiotic realm compounds, approx. 200 of which have found direct use as
where we know that withholding a drug can lead to death. drugs (Fig. 2).9
Instead, antibiotics are measured against a best available Improvements by semi-synthesis on these scaffolds gener-
comparator. If the new drug is designed to overcome resistance, ated another 200–300 drugs, greatly exceeding the number of
yet the population of resistant bacteria causing disease in the synthetic antibiotics in clinical use (Fig. 3).
trial is low, statistical efficacy may not be met. Furthermore,
a drug that is designed for resistant bacteria, but yet has some 2.1. Why are natural product antibiotics privileged in this
manageable host toxicity, may never see sufficient patients in therapeutic space?
a trial (oen conducted at large Western hospitals) and the
therapeutic index may never be adequately addressed. A mildly These compounds are the products of millions of years of
evolution. They are fashioned by natural selection to interact
with cellular targets with high efficiency and selectivity and to
Dr Gerard (Gerry) Wright is the avoid resistance. Furthermore, as products of evolution, they
Michael G DeGroote Chair in intrinsically possess the physiochemical properties necessary
antiinfective research and the for penetrating bacterial cells, unlike many synthetic mole-
director of the Michael G. cules.10,11 Molecular selectivity is a hallmark of many natural
DeGroote Institute for Infectious product antibiotics, with relatively few broadly toxic
Disease Research at McMaster compounds, and those which are, such as the enediynes, have
University in Hamilton, Ontario. a tightly regulated biosynthesis and are molecularly neutralized
As an undergraduate student, he through co-expression of protective proteins.12
worked on the biosynthesis of the
siderophore aerobactin. He 2.2. If natural products are so successful as antibiotics, why
received his PhD in Chemistry at can’t we easily identify new ones?
the University of Waterloo Most natural product antibiotics were detected using ‘The
working on fungal P-450 enzymes Waksman Platform’.13 In this strategy, natural product extracts,
as targets for antifungal drugs. He was a postdoctoral fellow with principally derived from bacteria and fungi, are screened for
Christopher Walsh at Harvard Medical School (1991–1992) where bacterial cell killing effects in the laboratory using simple
he worked on the molecular mechanism of the newly emerging culture-based assays. This approach powered the Golden Age of
resistance to vancomycin in enterococci. In 1993 he joined the antibiotic discovery resulting in our current highly successful
Department of Biochemistry at McMaster University where for over drugs. Because of the difficulty in the clinic of accurately iden-
two decades he has explored the mechanisms of resistance and tify the disease-causing pathogen and the efficacy and safety
biosynthesis of a large number of antibiotics with the support and index of the rst successful natural product drugs, the criteria
collaboration of a merry gang of creative and dedicated colleagues. governing antibiotic discovery was strict. Candidate antibiotics

This journal is © The Royal Society of Chemistry 2017 Nat. Prod. Rep., 2017, 34, 694–701 | 695
View Article Online

Natural Product Reports Viewpoint

The past – our legacy synthetic antibiotics. These scaffolds span the Golden Age of antibiotics with the sulfonamides such as sulfa-
Fig. 1
methoxazole discovered in the 1930s, and the oxazolidinone linezolid in the 1980s.
Published on 01 June 2017. Downloaded by Cornell University Library on 12/07/2017 22:19:04.

had to show broad-spectrum efficacy and the absence of human


toxicity. Compounds with narrow-spectrum activity, e.g. to one
genus or species, those with any toxic effects, or potential
pharmacological liabilities were discarded early in the discovery
process, oen never to be further studied.
Furthermore, most discovery efforts in the Golden Age
focused on mining the compounds produced by bacteria of the
order Actinomycetales, commonly derived from the soil. This
emphasis on broad-spectrum compounds from a single order of
bacteria eventually resulted in the frequent re-isolation of the
same compounds that are produced by many species and
genera. Removing producers of such compounds early in drug
discovery, a process termed dereplication, became a priority but
was resource consuming. The growing realization that new Fig. 3 The past – our legacy semi-synthetic antibiotics. These are the
antibiotic discovery from this traditional source resulted in result of successful semi-synthetic derivatives of natural products
diminishing returns caused a pivot away from natural products (changes shown in blue) improved efficacy, safety and expanded
towards synthetic compounds. This occurred at a historic point, spectrum.
the 1980s and early 1990s, when large-scale combinatorial
synthesis and genome-inuenced target-based drug discovery
emerged on the scene; subsequently, natural product divisions 3. Prospects for natural products in
in many pharmaceutical companies were scaled back or shut-
tered. Ironically, this occurred at a time when the genomics,
21st century antibiotic discovery
molecular biology, and biochemistry of natural products To address the antibiotics crisis, we must tackle the problem
became increasingly tractable and popular in academic using several parallel approaches. First, we need to identify new
research programs, revealing a wealth of opportunity for new antibiotic chemical scaffolds that evade the resistance mecha-
discovery strategies beyond the traditional Waksman Platform. nisms circulating in pathogens today. Second, we need to
preserve as best we can our existing drugs. Finally, we need to

Fig. 2 The past – our legacy natural product antibiotics. These drugs were discovered using the Waksman Platform,13 where microbial extracts
were screened in whole cell killing assays.

696 | Nat. Prod. Rep., 2017, 34, 694–701 This journal is © The Royal Society of Chemistry 2017
View Article Online

Viewpoint Natural Product Reports

explore non-traditional anti-infective strategies. Natural prod- new antimicrobial chemistry. Ironically, despite the success of
ucts can contribute to all these areas.14 b-lactams, which account for 60% of antibiotics in clinical
use, fungal sources of antibiotics have not been as extensively
3.1. New antibiotics pursued (the exception is fusidic acid, a topical antibiotic),
though, like actinomycetes, genome sequencing identies
The Actinomycetales are the source of the majority of our
dozens of natural product biosynthetic clusters per fungal
current antibiotic drugs. While the Waksman Platform
genome.19
frequently re-identies known compounds from extracts of
It is well accepted that our capacity to grow most bacterial
these bacteria, these can be readily identied.15 Genome
species in the lab is quite limited. We are therefore missing
sequencing reveals that each strain can produce 20–40
a signicant number of microbial genomes using the standard
Published on 01 June 2017. Downloaded by Cornell University Library on 12/07/2017 22:19:04.

secondary metabolites, many of which have not yet been char-


Waksman Platform approach, raising the question of what
acterized and some no doubt have antibiotic activity. Most of
chemical diversity we are failing to sample as a result. Lewis and
these are not readily produced under laboratory settings and are
Epstein’s laboratory have pioneered efforts such as the iChip to
therefore not tested in traditional assays of extracts. Efforts to
tame unculturable bacteria for growth in the lab that enables
awaken these so-called ‘cryptic’ or ‘silent’ biosynthetic gene
the mining of antibiotic ‘dark matter’.20 Importantly, with the
clusters have been pursued including altering the growth
isolation of teixobactin (Fig. 6), a new antibiotic scaffold, they
conditions, the overexpression of genetic regulators, the addi-
have demonstrated that such efforts can yield unique chem-
tion of chemical elicitors, and the capture of biosynthetic gene
istry.21 Another method is to forego cell culture and capture
clusters for expression in heterologous hosts. These efforts have
biosynthetic gene clusters from total metagenomic DNA. Using
resulted in several rare and new compounds being identied
such an approach, Brady and co-workers have identied several
(e.g. refs 16–18) (Fig. 4) offering encouragement that such
new antibiotic compounds including tetarimycin A (Fig. 6).22,23
strategies, especially if performed in high throughput as is
These strategies demonstrate concretely that the der-
occurring in companies such as Warp Drive Bio, can produce
eplication barrier can be overcome, and that new antibiotic
new antibiotic scaffolds.
scaffolds can be found in microbes. Plant-derived compounds
An alternative to deep mining of Actinomycetales genomes is
tend to be less selective and less sufficiently potent, conse-
the exploration of genera that have not been as extensively
quently making them less desirable as conventional antibiotic
explored for antibiotics in the past but, nevertheless, are known
drugs.
to produce antibiotic substances. These include proteobacteria
Furthermore, altering the screening strategies from the
such as myxobacteria, pseudomonads, burkholderias, among
traditional Waksman approach to include for example nutrient-
other Gram negative bacteria (Fig. 5). Firmicutes such as bacilli
limiting conditions,24 stress,25 target depletion to sensitize
were among the rst bacteria to be shown to produce antibi-
strains,26–28 genetic and chemical synthetic lethality29–32 and
otics, some such as gramicidin are still in use as topical agents
today, and these also are worthy of systematic exploration for

Fig. 4The present – antibiotics from cryptic biosynthetic gene clusters. Awakening of ‘cryptic’ or ‘silent’ biosynthetic clusters that are found in all
sequenced actinomycetes offers a route to new chemical matter. Examples shown here are taromycin,18 N-acetylmureidomycin,17 and
pulvomycin.16

This journal is © The Royal Society of Chemistry 2017 Nat. Prod. Rep., 2017, 34, 694–701 | 697
View Article Online

Natural Product Reports Viewpoint


Published on 01 June 2017. Downloaded by Cornell University Library on 12/07/2017 22:19:04.

Fig. 5 The present – antibiotics from Gram negative bacteria. Mupirocin is a successful anti-staphylococcal topical antibiotic in clinical use while
the other examples, enacyloxin IIa, myxopyronin and pantocin A have yet to be introduced into the clinic.

screens in whole organisms such as Caenorhabditis elegans33 Variations of known antibiotic scaffolds also have value.
offers new opportunities to identify antibiotic leads.34 Even small chemical changes can result in substantial effects on
efficacy and drug-like properties. Access to many scaffold vari-
ations provides valuable starting material for expanding the
3.2. ‘Old’ antibiotics chemical space around natural product antibiotics that can be
Of the 28 000 natural product antibiotics discovered over the further elaborated by semi-synthesis. Using resistance as an
past decades, >0.1% are in clinical use. Many of the other 99.9% initial screen is an easy way to enrich strain collection
will never be suitable as drugs because of the intractable issues surrounding distinct chemical scaffolds37 and to predict
of efficacy, toxicity, stability, etc. Nevertheless, some of these potential resistance mechanisms that may emerge in the
compounds no doubt are worthy of reinvestigation. The clinic.38
example of daptomycin, once discarded for issues of toxicity
and narrow spectrum but now a highly protable drug proves
the point.35 A major challenge to pursuing this hypothesis is 3.3. Rescuing legacy antibiotics
that there is no effective way to revisit these compounds in An orthogonal strategy to new antibiotic discovery is the iden-
a systematic fashion. Most are unavailable for purchase, and the tication of compounds that lack antibiotic activity but which
historical libraries of pharmaceutical companies are not readily enhance the activity of our existing antibiotics. Such antibiotic
available. An international effort to generate and distribute adjuvants39 can improve the efficacy of legacy antibiotics and
a library of microbially-derived natural products could have decrease the frequency of resistance, thereby preserving drugs
a signicant impact on antibiotic discovery. The efforts of CO- that are safe and well known to clinicians. Adjuvants are clas-
ADD (the Community for Open Antimicrobial Drug Discovery), sied into two types, those that act on bacteria directly (Type I),
which seeks to mine synthetic and natural product chemicals and those that enhance host antimicrobial properties (Type II).
generated by labs across the globe is a potential opportunity Type I adjuvants include inhibitors of acquired resistance (Type
here.36 Ia), and examples include inhibitors of b-lactamases that are in

Fig. 6 The present – antibiotics from microbial ‘dark matter’. Microbial genomes of rare, e.g. teixobactin from Eleftheria terrae,21 or uncultured
(metagenomes), e.g. tetarimycin A,23 offer routes to access new antibiotic chemical matter.

698 | Nat. Prod. Rep., 2017, 34, 694–701 This journal is © The Royal Society of Chemistry 2017
View Article Online

Viewpoint Natural Product Reports

clinical use today; Type Ib compounds overcome intrinsic antivirulence. Consequently, combinations with antibiotics
resistance mechanisms such as antibiotic entry and efflux. may offer the most logical way forward for many antibiotic
Natural products offer great opportunities here as well. The alternatives in the short- to medium-term. Compelling targets
rst inhibitor of a b-lactamase, clavulanic acid (Fig. 7), was include blocking the activities of bacterial toxins, secretion
isolated from a b-lactam antibiotic producer, Streptomyces systems, adhesins, impairing the acquisition of vital nutrients,
clavuligerus. It seems reasonable that, in addition to being and other molecular requirements for infection or biolm
sources of antibiotics, microbes are also sources of inhibitors of formation. Microbial natural products such as balomycin A1
resistance. Our recent isolations of an inhibitor of metallo-b- and brefeldin A (Fig. 8) block vesicle trafficking in eukaryotic
lactamases, aspergillomarasmine A, from a fungus40 and of cells and therefore have been identied as antivirulence
rifamycin efflux, A301A, from a bacterium41 are consistent with factors.44 But plants are rich in compounds with antivirulence
Published on 01 June 2017. Downloaded by Cornell University Library on 12/07/2017 22:19:04.

this hypothesis (Fig. 7). In the antibiotic adjuvant realm there is properties including hamamelitannin, which blocks Staphylo-
certainly opportunity for plant natural products, and indeed coccus aureus quorum sensing and sensitizes biolms to anti-
molecules such as berberine have already been shown to be biotics,45 phloretin which inhibits Clostridium difficile toxins,46
inhibitors of efflux for example.42 Natural products also can and avones and sapogenins that impede virulence in MRSA47
contribute to the discovery of Type II adjuvants as we have (Fig. 8). There is great opportunity here for identifying
shown with the macrophage-enhancing streptazolin (Fig. 7).43 compounds and mixtures from both plants and microbes that
attenuate microbial pathogenesis, and which can contribute to
21st century anti-infective medicines.
3.4. Alternatives to antibiotics
The unparalleled efficacy and potency of antibiotics have 3.5. Diversity through synthetic biology
resulted in the primacy of such compounds to control infection
Natural product biochemistry, molecular biology, and genomics
for over seven decades. Alternatives such as antivirulence
are now sufficiently advanced to allow the rational exploitation
compounds or biolm inhibitors have therefore not been put
of compound production in more directed studies with the aim
into clinical practice, despite being touted in academic circles.
of expanding chemical diversity. The rst efforts in this area,
One of the principal barriers to the success of such strategies in
pioneered almost 20 years ago,48 foresaw the opportunity but
the drug-discovery eld is the design of suitable clinical trials
were outside the skill sets and resources of most laboratories.
for efficacy and for disease prevention in the case of
The dropping costs of genome and metagenome sequencing,
coupled with advances in the increasingly accurate prediction

Fig. 7 The future – antibiotic adjuvants. Antibiotic adjuvants are non- Fig. 8 The future – antivirulence compounds. These compounds
antibiotic compounds that enhance the cell killing activities of anti- diminish the ability of pathogens to cause infection, but do not actually
biotics by blocking resistance or enhancing host defenses. kill the cells. These offer the ability to control infection before it starts.

This journal is © The Royal Society of Chemistry 2017 Nat. Prod. Rep., 2017, 34, 694–701 | 699
View Article Online

Natural Product Reports Viewpoint

of natural product structures from biosynthetic genes,49–52 are N. Morris, P. Mpangase, H. van der Meulen, S. V. Omar,
enabling the mining of heretofore rare or impossible-to-study T. S. Brown, A. Narechania, E. Shaskina, T. Kapwata,
natural product biosynthesis. The advent of improved cluster B. Kreiswirth and N. R. Gandhi, N. Engl. J. Med., 2017, 376,
capture methods53,54 together with optimized strategies for 243–253.
hosts of heterologous gene expression55,56 and the ability to 7 J. H. Rex, B. I. Eisenstein, J. Alder and M. Goldberger, Lancet
refactor entire genes, regulatory elements, and even clusters as Infect. Dis., 2013, 13, 269–275.
a result of the low-cost of gene synthesis18 is enabling the 8 J. O’Neill, Tackling Drug-Resistant Infections Globally: Final
movement of the eld rmly into the area of synthetic biology.57 Report and Recommendations, 2016 (https://amr-review.org/
This is an area where traditional natural product collection, sites/default/les/160518_Final%20paper_with%
chemistry, semi-synthesis, new cultivation strategies, and strain 20cover.pdf).
Published on 01 June 2017. Downloaded by Cornell University Library on 12/07/2017 22:19:04.

improvement can synergize to generate increased chemical 9 J. Bérdy, J. Antibiot., 2012, 65, 385–395.
diversity around scaffolds that can nd use in discovery efforts 10 R. O’Shea and H. E. Moser, J. Med. Chem., 2008, 51, 2871–
in the antibiotics eld. 2878.
11 L. L. Silver, Bioorg. Med. Chem., 2016, 24, 6379–6389.
4. Conclusions 12 S. Singh, M. H. Hager, C. Zhang, B. R. Griffith, M. S. Lee,
K. Hallenga, J. L. Markley and J. S. Thorson, ACS Chem.
The challenge of the antibiotics crisis needs to be met with Biol., 2006, 1, 451–460.
concerted efforts across many disciplines and areas of expertise. 13 K. Lewis, Nature, 2012, 485, 439–440.
Natural products led the way during the Golden Age of antibi- 14 T. A. Wencewicz, Bioorg. Med. Chem., 2016, 24, 6227–6252.
otic discovery and, despite a lull of about 30 years, there is 15 G. Cox, A. Sieron, A. M. King and G. De Pascale, Cell Chem.
considerable reason to believe that natural products still have Biol., 2017, 24, 98–109.
much to offer. Our ability to mine, extract, produce, charac- 16 N. L. McKenzie, M. Thaker, K. Koteva, D. W. Hughes,
terize, and manipulate natural products has never been greater. G. D. Wright and J. R. Nodwell, J. Antibiot., 2010, 63, 177–182.
The existing chemical diversity that has yet to be effectively 17 L. Jiang, L. Wang, J. Zhang, H. Liu, B. Hong, H. Tan and
explored in the antibiotic realm is staggeringly large. Further- G. Niu, Sci. Rep., 2015, 5, 14111.
more, these compounds are privileged starting points in the 18 K. Yamanaka, K. A. Reynolds, R. D. Kersten, K. S. Ryan,
antimicrobial eld that can now be enhanced using synthetic D. J. Gonzalez, V. Nizet, P. C. Dorrestein and B. S. Moore,
biology strategies. There is much to be excited about and Proc. Natl. Acad. Sci. U. S. A., 2014, 111, 1957–1962.
encouraged by. Whereas it is important to not be a Pollyanna, 19 T. A. J. van der Lee and M. H. Medema, Fungal Genet. Biol.,
since the barriers to moving new discoveries out of the lab and 2016, 89, 29–36.
into the clinic are fraught with peril and compounds can fail for 20 D. Nichols, N. Cahoon, E. M. Trakhtenberg, L. Pham,
many reasons, the eld of natural products is well positioned to A. Mehta, A. Belanger, T. Kanigan, K. Lewis and
once again lead in innovation for solving the antibiotic crisis in S. S. Epstein, Appl. Environ. Microbiol., 2010, 76, 2445–2450.
the Resistance Era. 21 L. L. Ling, T. Schneider, A. J. Peoples, A. L. Spoering,
I. Engels, B. P. Conlon, A. Mueller, T. F. Schäberle,
5. Acknowledgements D. E. Hughes, S. Epstein, M. Jones, L. Lazarides,
V. A. Steadman, D. R. Cohen, C. R. Felix, K. A. Fetterman,
I am grateful for funding from the Canadian Institutes of Health W. P. Millett, A. G. Nitti, A. M. Zullo, C. Chen and
Research, the Natural Sciences and Engineering Research K. Lewis, Nature, 2015, 517, 455–459.
Council, and by a Canada Research Chair in Antibiotic 22 A. Milshteyn, J. S. Schneider and S. F. Brady, Chem. Biol.,
Biochemistry. 2014, 21, 1211–1223.
23 D. Kallidas, H.-S. Kang and S. F. Brady, J. Am. Chem. Soc.,
6. References 2012, 134, 19552–19555.
24 S. Zlitni, L. F. Ferruccio and E. D. Brown, Nat. Chem. Biol.,
1 E. D. Brown and G. D. Wright, Nature, 2016, 529, 336–343. 2013, 9, 796–804.
2 A. Fleming, E. B. Chain and H. Florey, Sir Alexander Fleming- 25 J. M. Stokes, S. French, O. G. Ovchinnikova, C. Bouwman,
Nobel Lecture: Penicillin, Nobel Lectures, 1964. C. Whiteld and E. D. Brown, Cell Chem. Biol., 2016, 23,
3 J. Davies and D. Davies, Microbiol. Mol. Biol. Rev., 2010, 74, 267–277.
417–433. 26 J. Wang, S. M. Soisson, K. Young, W. Shoop, S. Kodali,
4 E. Bathoorn, C. Tsioutis, J. M. da Silva Voorham, A. Galgoci, R. Painter, G. Parthasarathy, Y. S. Tang,
E. V. Scoulica, E. Ioannidou, K. Zhou, J. W. Rossen, R. Cummings, S. Ha, K. Dorso, M. Motyl, H. Jayasuriya,
A. Gikas, A. W. Friedrich and H. Grundmann, J. Antimicrob. J. Ondeyka, K. Herath, C. Zhang, L. Hernandez, J. Allocco,
Chemother., 2016, 71, 1207–1212. Á. Basilio, J. R. Tormo, O. Genilloud, F. Vicente, F. Pelaez,
5 L. Chen, R. Todd, J. Kiehlbauch, M. Walters and A. Kallen, L. Colwell, S. H. Lee, B. Michael, T. Felcetto, C. Gill,
MMWR Morb. Mortal. Wkly. Rep., 2017, 66, 33. L. L. Silver, J. D. Hermes, K. Bartizal, J. Barrett,
6 N. S. Shah, S. C. Auld, J. C. M. Brust, B. Mathema, N. Ismail, D. Schmatz, J. W. Becker, D. Cully and S. B. Singh, Nature,
P. Moodley, K. Mlisana, S. Allana, A. Campbell, T. Mthiyane, 2006, 441, 358–361.

700 | Nat. Prod. Rep., 2017, 34, 694–701 This journal is © The Royal Society of Chemistry 2017
View Article Online

Viewpoint Natural Product Reports

27 B. Jiang, D. Xu, J. Allocco, C. Parish, J. Davison, K. Veillette, 42 K. Lewis and F. M. Ausubel, Nat. Biotechnol., 2006, 24, 1504–
S. Sillaots, W. Hu, R. Rodriguez-Suarez and S. Trosok, Chem. 1507.
Biol., 2008, 15, 363–374. 43 J. A. Perry, K. Koteva, C. P. Verschoor, W. Wang,
28 J. Ondeyka, G. Harris, D. Zink, Á. Basilio, F. Vicente, G. Bills, D. M. E. Bowdish and G. D. Wright, J. Antibiot., 2015, 68,
G. Platas, J. Collado, A. González, M. d. L. Cruz, J. Martin, 40–46.
J. N. Kahn, S. Galuska, R. Giacobbe, G. Abruzzo, E. Hickey, 44 M. Garland, S. Loscher and M. Bogyo, Chem. Rev., 2017, 117,
P. Liberator, B. Jiang, D. Xu, T. Roemer and S. B. Singh, J. 4422–4461.
Nat. Prod., 2009, 72, 136–141. 45 G. Brackman, K. Breyne, R. De Rycke, A. Vermote, F. Van
29 J. Campbell, A. K. Singh, J. P. Santa Maria, Y. Kim, S. Brown, Nieuwerburgh, E. Meyer, S. Van Calenbergh and T. Coenye,
J. G. Swoboda, E. Mylonakis, B. J. Wilkinson and S. Walker, Sci. Rep., 2016, 6, 20321.
Published on 01 June 2017. Downloaded by Cornell University Library on 12/07/2017 22:19:04.

ACS Chem. Biol., 2011, 6, 106–116. 46 J. Tam, G. L. Beilhartz, A. Auger, P. Gupta, A. G. Therien and
30 M. A. Labroli, J. P. Caldwell, C. Yang, S. H. Lee, H. Wang, R. A. Melnyk, Chem. Biol., 2015, 22, 175–185.
S. Koseoglu, P. Mann, S.-W. Yang, J. Xiao, C. G. Garlisi, 47 A. Muhs, J. T. Lyles, C. P. Parlet, K. Nelson, J. S. Kavanaugh,
C. Tan, T. Roemer and J. Su, Bioorg. Med. Chem. Lett., 2016, A. R. Horswill and C. L. Quave, Sci. Rep., 2017, 7, 42275.
26, 3999–4002. 48 R. McDaniel, A. Thamchaipenet, C. Gustafsson, H. Fu,
31 M. A. Farha, A. Leung, E. W. Sewell, M. A. D’Elia, S. E. Allison, M. Betlach and G. Ashley, Proc. Natl. Acad. Sci. U. S. A.,
L. Ejim, P. M. Pereira, M. G. Pinho, G. D. Wright and 1999, 96, 1846–1851.
E. D. Brown, ACS Chem. Biol., 2013, 8, 226–233. 49 M. H. Medema and M. A. Fischbach, Nat. Chem. Biol., 2015,
32 L. Ejim, M. A. Farha, S. B. Falconer, J. Wildenhain, 11, 639–648.
B. K. Coombes, M. Tyers, E. D. Brown and G. D. Wright, 50 N. Ziemert, M. Alanjary and T. Weber, Nat. Prod. Rep., 2016,
Nat. Chem. Biol., 2011, 7, 348–350. 33, 988–1005.
33 T. I. Moy, A. R. Ball, Z. Anklesaria, G. Casadei, K. Lewis and 51 J. I. Tietz, C. J. Schwalen, P. S. Patel, T. Maxson, P. M. Blair,
F. M. Ausubel, Proc. Natl. Acad. Sci. U. S. A., 2006, 103, 10414– H.-C. Tai, U. I. Zakai and D. A. Mitchell, Nat. Chem. Biol.,
10419. 2017, 13, 470–478.
34 M. A. Farha and E. D. Brown, Ann. N. Y. Acad. Sci., 2015, 1354, 52 C. A. Dejong, G. M. Chen, H. Li, C. W. Johnston,
54–66. M. R. Edwards, P. N. Rees, M. A. Skinnider,
35 R. H. Baltz, V. Miao and S. K. Wrigley, Nat. Prod. Rep., 2005, A. L. H. Webster and N. A. Magarvey, Nat. Chem. Biol.,
22, 717–741. 2016, 12, 1007–1014.
36 M. A. Cooper, Nat. Rev. Drug Discovery, 2015, 14, 587–588. 53 X. Tang, J. Li, N. Millán-Aguiñaga, J. J. Zhang, E. C. O’Neill,
37 M. N. Thaker, W. Wang, P. Spanogiannopoulos, J. A. Ugalde, P. R. Jensen, S. M. Mantovani and
N. Waglechner, A. M. King, R. Medina and G. D. Wright, B. S. Moore, ACS Chem. Biol., 2015, 10, 2841–2849.
Nat. Biotechnol., 2013, 31, 922–927. 54 M. Xu, Y. Wang, Z. Zhao, G. Gao, S.-X. Huang, Q. Kang, X. He,
38 T. S. Cros, A. J. Gasparrini and G. Dantas, Nat. Rev. S. Lin, X. Pang, Z. Deng and M. Tao, Appl. Environ. Microbiol.,
Microbiol., 2017, 1–13. 2016, 82, 5795–5805.
39 G. D. Wright, Trends Microbiol., 2016, 24, 862–871. 55 J. P. Gomez-Escribano and M. J. Bibb, Microb. Biotechnol.,
40 A. M. King, S. A. Reid-Yu, W. Wang, D. T. King, G. De Pascale, 2010, 4, 207–215.
N. C. Strynadka, T. R. Walsh, B. K. Coombes and 56 K. Ochi, J. Antibiot., 2016, 70, 25–40.
G. D. Wright, Nature, 2014, 510, 503–506. 57 M. N. Thaker and G. D. Wright, ACS Synth. Biol., 2015, 4,
41 G. Cox, K. Koteva and G. D. Wright, J. Antimicrob. Chemother., 195–206.
2014, 69, 1844–1855.

This journal is © The Royal Society of Chemistry 2017 Nat. Prod. Rep., 2017, 34, 694–701 | 701

You might also like