You are on page 1of 13

PERSPECTIVES

for patients with septicaemia due to MDR


OPINION
Escherichia coli3. Data from the USA show
a similar pattern. The 2013 report from the
Antibiotic resistance breakers: can CDC highlighted carbapenem-resistant
Enterobacteriaceae (CREs) as an urgent
repurposed drugs fill the antibiotic threat4. In Asia, substantial resistance has
emerged in both India and China, with
discovery void? resistance levels reported in the range of
50–80%. This has caused increased use
of carbapenems, which were previously
David Brown reserved for extreme cases of infection in
the very sick, the immune-compromised or
Abstract | Concern over antibiotic resistance is growing, and new classes of as a last resort. Now, bacteria have adapted
antibiotics, particularly against Gram-negative bacteria, are needed. However, and selected for carbapenem-destroying
even if the scientific hurdles can be overcome, it could take decades for sufficient enzymes, known as carbapenemases, and
numbers of such antibiotics to become available. As an interim solution, few antibiotics remain effective against
these CREs. K. pneumoniae, E. coli,
antibiotic resistance could be ‘broken’ by co‑administering appropriate
P. aeruginosa and A. baumannii produce
non-antibiotic drugs with failing antibiotics. Several marketed drugs that do not metallo-β-lactamases such as K. pneumoniae
currently have antibacterial indications can either directly kill bacteria, reduce carbapenemase (KPC) and New Delhi
the antibiotic minimum inhibitory concentration when used in combination with metallo-β-lactamase (NDM) — enzymes
existing antibiotics and/or modulate host defence through effects on host innate that degrade numerous antibiotics containing
a β-lactam ring, such as penicillins,
immunity, in particular by altering inflammation and autophagy. This article
cephalosporins and carbapenems. Bacteria
discusses how such ‘antibiotic resistance breakers’ could contribute to reducing carrying the genes that encode these
the antibiotic resistance problem, and analyses a priority list of candidates for enzymes are becoming resistant to all
further investigation. available penicillins, cephalosporins and
β-lactamase inhibitors, including clavulanic
Resistance to current antibiotics is rapidly owing to their role in many infections in acid and avibactam (FIG. 1). These bacteria
increasing. In its 2014 report of global human organs (such as the lung and urinary are also resistant to virtually all other anti-
antimicrobial resistance, the World Health tract), the frequency of antibiotic resistance biotics, with the exception of colistin, an old
Organization (WHO) portrayed high levels amongst them and the lack of alternative (and somewhat toxic) polymixin class anti-
of antibiotic resistance in the bacteria that antibiotics1. Several of these pathogens biotic, although even colistin resistance has
cause common infections. A number of are Gram-negative bacteria, which are of now emerged in South Asia. Both KPC and
leading authorities have issued passionate particular concern as in these organisms NDM, as well as Verona integron-encoded
statements urging action, including the resistance of up to 50% against carbapenems, metallo-β-lactamase (VIM), have been
Director-General of the WHO, Margaret the current last line of defence, has been reported in Pseudomonas spp. In some areas
Chan; the Director of the Wellcome Trust, reported in some developing countries1. of the world, including the United States,
Jeremy Farrar; and the Director of the A few new antibiotics against Gram-positive Israel, Italy, Greece and China, the emer-
US Center for Disease Control (CDC), bacteria have become available in recent gence of bacteria that produce KPCs, which
Tom Frieden. The United Kingdom’s Chief years, but no totally new class of antibiotic render them resistant to carbapenems, is
Medical Officer, Sally Davies, warned that has been introduced for the treatment of becoming a serious threat.
the country could find itself back in the Gram-negative infections for more than A few derivatives of older antibiotic
nineteenth century in terms of its ability to 40 years. classes or combinations incorporating new
treat bacterial infections. The seriousness In South Asia, the Middle East and β-lactamase inhibitors such as avibactam
of the threat has been compared with the Mediterranean, modern medicine is and tazobactam offer some hope in the short
those of global warming and terrorism already under threat from these multidrug term. Two compounds that have reached
(see Further information). resistant (MDR) Gram-negative bacteria2 Phase III trials — eravacycline5, a next-
The so‑called ESKAPE pathogens (K. pneumoniae, A. baumannii, P. aeruginosa, generation tetracycline, and plazomicin6,
(Enterococcus faecium, Staphylococcus and Enterobacter spp.). European data span- a next-generation aminoglycoside —
aureus, Klebsiella pneumoniae, Acinetobacter ning 2005–2010 indicate growing resistance have activity against Gram-negative organ-
baumannii, Pseudomonas aeruginosa, and to cephalosporins, fluoroquinolones and isms. The recently approved ceftazidime–
Enterobacter spp.) are especially important aminoglycosides, and a 30% mortality rate avibactam7 combination is effective against

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | DECEMBER 2015 | 821

© 2015 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

Cell wall synthesis


antibiotic research and development.
• β-lactams • Carbapenems Folate synthesis One ARB could potentially revitalize
• Penicillins • Monobactams • Sulfonamides several antibiotics in a class11, and some ARBs
• Cephalosporins • Glycopeptides • Trimethoprim may even work across classes. Lethal bacterial
infections might be effectively treated with
Nucleic acid far fewer compounds than would be required
synthesis to replace existing antibiotics. Moreover, the
• DNA gyrase concept may help to extend the lifespan of
• Quinolones
future antibiotic classes. Here, after high­
50S
lighting the priority bacteria, key antibiotics
mRNA Peptide PABA DHF THF to be salvaged and the properties of ARBs, we
30S
discuss a list of proposed priority candidates
for further investigation and issues for their
RNA polymerase development.
• Rifamycins

LPS and/or Protein synthesis Repurposing to provide ARBs


TLR4 blockers 50S subunit 30S subunit Priority antibiotics and bacteria. ARBs
Cell membrane • Macrolides • Aminoglycosides
disrupters • Oxazolidinones • Tetracyclines should be sought to salvage one or more
• Polymyxins • Chloramphenicol key members of each mechanistic antibiotic
class, particularly those used against
Figure 1 | Sites of antibacterial action and mechanisms of resistance.  Antibiotics can be classi- Gram-negative bacteria. Thus, the antibiotics
fied by their mechanism of action. Resistance to one antibiotic within a class can confer resistance that most need ARBs are: cephalosporins
Nature Reviews | Drug Discovery
to others with the same target. Resistance arises by two main mechanisms: random mutations
and carbapenems (which disrupt cell wall
during DNA replication and transfer of DNA between bacteria, often as plasmids. The transferred
DNA can contain genes that confer resistance, and natural selection then favours the survival of the synthesis); polymyxins (which disrupt cell
resistant bacteria during antibiotic treatment of a patient. DHF, dihydrofolic acid; LPS, lipopoly­ membrane synthesis); fluoroquinolones
saccharide; PABA, para-aminobenzoic acid; THF, tetrahydrofolic acid; TLR4, Toll-like receptor 4. (which disrupt DNA synthesis); tetracyclines
and aminoglycosides (which disrupt protein
synthesis by inhibiting the 30S ribosomal
several MDR Gram-negative bacteria; hope of counteracting resistance to β-lactam subunit); and macrolides (which disrupt
the ceftolozane–tazobactam8 combination antibiotics in the near term, but further protein synthesis by inhibiting the 50S
(also recently approved) has good exploitation of β-lactamase inhibitors may ribosomal subunit).
anti-pseudomonal activity; and the be of limited use in the longer term, as there Acquired carbapenemases have been
aztreonam–avibactam combination7, which has been a 100‑fold increase in the number of highlighted as the greatest immediate threat
has completed Phase I trials, works in vitro known β-lactamases in the past 40 years11. to the effectiveness of the antibiotic arsenal2.
and in animal models against many organ- Surprisingly, the success of β-lactamase Carbapenemases are encoded by genes that
isms that produce metallo-β-lactamase. inhibitors has not led to substantial clinical are transferable between bacteria and confer
However, resistance to these com­binations and commercial exploitation of the concept resistance to many of the most heavily used
will probably soon arise and there of ARBs beyond this class. Attempts to antibiotics ­— carbapenems and β-lactam
are no totally new chemical classes of reduce resistance by blocking efflux pumps antibiotics. The carbapenems are the last
antibiotic on the horizon for the treatment on bacterial cells — which can diminish the good line of defence against MDR Gram-
of Gram-negative bacteria. effectiveness of antibiotics by lowering their negative bacteria, and the consensus is that
It is therefore crucial that ways of intracellular concentration — have been extending the useful lifespan of this class of
breaking resistance to current antibiotics are pursued for many years, so far without notable drugs is a top priority. As such, the intra­
found as soon as possible. One strategy to success. However, efforts continue and venous formulation of a broad-spectrum
achieve this goal is to co‑administer another deserve further attention12. Novel combina- ARB for use together with a carbapenem
drug with the failing antibiotic, which tions of existing classes of antibiotics could (or a cephalosporin) in intensive-care
restores sufficient antibacterial activity. also be investigated; for example, macrolides hospital settings should be top priority.
The use of such antibiotic resistance breakers may be able to synergize with β-lactams and ARBs that are effective against
(ARBs) to salvage antibiotics is exemplified by fluoroquinolones13–18. K. pneumoniae, E. coli, P. aeruginosa and
the long-standing, successful and widespread This article, however, focuses on the A. baumannii, the four Gram-negative
co‑administration of β-lactamase inhibitors, identification of broad-spectrum ARBs by organisms whose resistance to antibiotics
such as clavulanic acid, with β-lactam anti- repurposing marketed drugs and nutra- is of greatest concern (all of which produce
biotics, such as amoxicillin9,10. Resistance to ceuticals. ARBs selected from marketed carbapenemases and are thus resistant to
amoxicillin and to this combination of drugs drugs would be particularly useful as their many β-lactam antibiotics), are of utmost
has been slow to emerge. However, mutation development could be faster, cheaper and importance. A secondary priority is to
of the β-lactamase TEM1 — thought to be probably have a higher success rate than that target Gram-positive bacteria, especially
the greatest driver of resistance to this class for new molecules. This could be crucial, methicillin-resistant S. aureus (MRSA) and
of antibiotics — has now occurred owing to given the pressing need for strategies to Clostridium difficile (which causes C. difficile-
the selection of organisms with clavulanate- tackle antibiotic resistance, the long develop- associated disease (CDAD)), as these
insensitive β-lactamases. As noted above, ment timelines for new antibiotics and the organisms cause recurring problems that
several new β-lactamase inhibitors offer the challenging financial environment for new are associated with substantial death rates.

822 | DECEMBER 2015 | VOLUME 14 www.nature.com/reviews/drugdisc

© 2015 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

Properties and identification of potential Box 1 | Host-targeted drugs that induce autophagy may break antibiotic resistance
ARBs. There are several properties that
potential ARBs could possess. First, ARBs Autophagy eliminates unwanted constituents from cells, including pathogens, damaged
could have direct antibacterial activity, even organelles and aggregated proteins. During fasting or starvation, autophagy recycles cytoplasmic
if they are not used clinically as antibiotics. material to maintain cellular homeostasis. Over the past few years, an increased understanding of
Second, ARBs could increase the efficacy of the pathways of autophagy has led to recognition of its role in a broad range of disease processes,
including host defence against pathogens. There are several excellent reviews on the role of
antibiotics and/or combat antibiotic resist-
autophagy as a defence against microbial invasion125,126.
ance mechanisms. Third, ARBs could help
Bacteria that are degraded by intracellular autophagy include Group A Streptococcus spp.127,
to clear the infection by interacting with host Salmonella spp.128, Shigella spp.129, Listeria monocytogenes130 and Mycobacterium tuberculosis131–133.
targets to activate host defence mechanisms; However, some bacteria have evolved to subvert this process134–136.
for example, by blocking the pro-inflammatory The best characterized protein involved in autophagy is mammalian target of rapamycin
Toll-like receptors (TLRs) or promoting (mTOR). Autophagy is induced by direct inhibitors of mTOR or by inhibitors of pathways that
autophagy (BOX 1). Arguably the most activate mTOR — class I phosphoinositide 3‑kinases (PI3Ks) and receptor tyrosine kinases that
interesting potential ARBs are those that activate the AKT pathway — thereby repressing autophagy. Inhibitors of these enzymes may
display more than one of these properties. provide useful therapeutics by inducing autophagy, although none is marketed at the present
A literature review was conducted time. Similarly, modulators of 5′-AMP-activated protein kinase (AMPK)137, mitogen-activated
searching for potential non-antibiotic protein kinases (MAPKs; including extracellular signal-regulated kinases (ERKs)) and the WNT
candidate drugs or nutraceuticals that are signalling pathway138 may be useful to increase autophagy and promote bacterial clearance.
Drugs that inhibit some of these pathways are in development for the treatment of cancer and
not used as antibiotics but have one or more
might prove effective in treating some infectious diseases.
of these three ARB properties. Drug safety
Particularly intriguing is the recent observation that activation of autophagy specifically in the
and the ability to achieve a drug plasma gut leads to systemic effects139. Remarkably, activation of intestinal AMPK induces autophagy in
concentration (by intravenous or oral both the gut and the brain and slows systemic ageing. Activation of autophagy in the gut alone
routes) that is similar to published mini- could therefore be sufficient to aid the clearance of systemic infections. This possibility is very
mum inhibitory concentrations (MICs) for relevant in assessing the potential of the AMPK activators, some of which have relatively low
antibacterial action are also important, and bioavailability.
combinations are more often successful if It is currently unclear whether activators of autophagy would be sufficiently powerful to use
the combination partner attacks a molecu- without antibiotics, but they could be used as ARBs when co‑administered with antibiotics,
lar target that is different from that of the similarly to β-lactamase inhibitors. There are already autophagy-activating drugs in clinical use or
antibiotic. Therefore, these aspects were also under clinical investigation for other diseases. If modulation of autophagy does emerge as a useful
used for prioritization. Last, the priorities drug approach for the treatment or prevention of bacterial diseases, it is possible that useful
above were also considered with regard to medicines could be repurposed from other indications.
the type of infections.
For the drugs that were short-listed, a
written summary was prepared of relevant and moulds. Moreover, it has antibacterial iron permeases or transporters (FTR1, FTR2
mechanisms and in vitro and in vivo data, activity, although this has never been and FTH1), a copper permease (CCC2),
as well as any available clinical data. Then, exploited clinically. Ciclopirox kills a wide an iron reductase (CFL1) and a siderophore
through individual and group discussions range of bacteria including many Gram- transporter (SIT1)38. Addition of FeCl3 to
with global experts, the strengths and weak- negative and Gram-positive species36. ciclopirox-treated cells reversed the effect of
nesses of each drug were identified. Based Recently, it was reported that this drug the drug on gene regulation, indicating that
on these discussions, the priority drugs with has direct antibacterial activity against its antifungal activity may be at least partially
the strongest evidence supporting a potential several of the high-priority MDR Gram- caused by iron limitation38.
role in breaking resistance are presented in negative bacteria37. When tested against Other mechanistic studies have
TABLES 1,2 and grouped into three categories antibiotic resistant A. baumannii, E. coli and indicated that, in addition to the effects on
in the discussion below: potential ARBs for K. pneumoniae, ciclopirox inhibited bacterial iron, ciclopirox also downregulates nucleo-
Gram-negative bacteria, potential ARBs growth at concentrations of 5–15 μg per ml, tide binding proteins39 and inhibits mamma-
for Gram-positive bacteria and potential regardless of the antibiotic resistance status. lian target of rapamycin (mTOR) signalling,
ARBs for both classes. Some drugs such as The authors suggested that the compound thereby inducing autophagy in mammalian
aspirin19,20, diclofenac21–23, ibuprofen24–26, inhibited the synthesis of lipopolysaccharide cells40. It seems likely that ciclopirox would
ivermectin27,28, lauric acid or monolaurin29,30, (LPS) in the surface coat of Gram-negative also activate autophagy in immune cells
metformin31–33, and vitamin D3 (REFS 34,35) bacteria. This would be a particularly (BOX 1).
were excluded owing to a lack of compelling valuable mechanism, as the LPS coat protects The rapid development of ciclopirox
evidence, although future research Gram-negative bacteria from the entry of for use against bacterial infections either
could identify these drugs as potential many antibiotics. Inhibition of LPS synthesis alone or as an ARB could be aided by the
ARBs (TABLE 3). might render Gram-negative bacteria existing pharmacokinetic, metabolic,
susceptible to antibiotics that are normally toxicological and clinical data. Ciclopirox
Potential ARBs for Gram-negative bacteria reserved for Gram-positive organisms. is usually administered topically, however,
Ciclopirox. Ciclopirox has been used for Ciclopirox also chelates intracellular iron, owing to the interest in ciclopirox for treat-
several decades as a topical antifungal agent which probably results in the inhibition of ment of haematological malignancies such
without the emergence of resistance. It is a metal-dependent enzymes. In Candida  as multiple myeloma41,42, systemic dosing
broad-spectrum agent with activity against albicans, ciclopirox has also been reported of this drug has been investigated41. Data
most clinically relevant dermatophytes, yeasts, to alter the regulation of the genes encoding from animal studies and a single human

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | DECEMBER 2015 | 823

© 2015 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

Table 1 | Priority compounds for repurposing as ARBs


Compound Class Structure Potential mechanisms of action
For Gram-negative bacteria
Ciclopirox Antifungal: used without Ciclopirox inhibits the synthesis of the LPS coat
the development of of Gram-negative bacteria37, chelates iron and
resistance for several regulates the genes encoding iron permeases
decades N O or transporters (FTR1, FTR2 and FTH1), copper
OH
permease (CCC2), iron reductase (CFL1) and
siderophore transporter (SIT1). It may also
contribute to antimicrobial effects38, and it can
induce autophagy40
Loperamide Anti-motility: used for the Loperamide facilitates tetracycline uptake45.
(Imodium; treatment of diarrhoeal With cephalosporins, loperamide dissipated
Janssen diseases the electrical component (ΔΨ) of the proton
Pharmaceutica) OH
motive force (PMF). In this same assay,
N Cl cephalosporins selectively dissipated the
O N
transmembrane chemical component (ΔpH) of
the PMF. The elimination of both ΔΨ and ΔpH
completely abolishes PMF and explains the
observed synergy between loperamide and
cephalosporins45
For Gram-positive bacteria
Berberine A traditional medicine O Direct antibacterial action may be due, in
in Europe, Asia and the O part at least, to inhibition of Gram-positive
Americas. It is used for the bacterial sortase49. It binds to TLR4–MD2,
treatment of diarrhoea thereby antagonizing LPS signalling51.
caused by Giardia lamblia Berberine inhibits TLR4–NF-κB–MIP2
and the Gram-negative N+ signalling, thus decreasing neutrophil
O
bacteria Escherichia coli, infiltration53, downregulates the expression
Klebsiella spp. and Vibrio O of pro-inflammatory genes (such as those
cholerae encoding TNF, IL‑1β, IL‑6, MCP1, iNOS and
COX2 (REF. 54)) and activates AMPK, thus
inducing autophagy55
For both Gram-negative and Gram-positive bacteria
Curcumin A food flavouring, colouring O O Curcumin competes with the LPS of
and neutraceutical Gram-negative bacteria to block excessive
inflammatory responses and prevent
bacterial invasion. It inhibits TLR2 and
HO OH TLR4 signalling72,73, downregulates TLR
O O expression74,75, prevents the upregulation of
IL‑8 expression79 and induces autophagy by
inhibiting the AKT–mTOR pathway71
Epigallocatechin‑ EGCG is one of the most OH EGCG has a broad range of mechanisms,
3‑gallate (EGCG) abundant polyphenols in OH
including inhibition of DNA gyrase103,
green tea and is thought to blockade of TLR4 binding106 and signal
be responsible for most of HO O transmission, and inhibition of conjugative
the supposed therapeutic OH transfer of the R plasmid of E. coli107
benefits of green tea
consumption O
OH OH
O

OH
OH

(+)-Naltrexone These are selective opioid (+)-Naloxone (+)-Naltrexone Both compounds block TLR4–MD2
and (+)-Naloxone antagonists used to signalling119
counter the effects HO HO
of opioid overdose
O OH O OH

N N
O O

AMPK, 5ʹ-AMP-activated protein kinase; ARBs, antibiotic resistance breakers; COX2, cyclooxygenase 2; IL‑1β, interleukin‑1β; iNOS, inducible nitric oxide
synthase; LPS, lipopolysaccharide; MCP1, monocyte chemotactic protein 1; MIP2, macrophage inflammatory protein 2; mTOR, mammalian target of rapamycin;
NF-κB, nuclear factor κB; TLR4, Toll-like receptor 4; TNF, tumour necrosis factor.

824 | DECEMBER 2015 | VOLUME 14 www.nature.com/reviews/drugdisc

© 2015 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

oral dosing study suggest that drug safety is Table 2 | ARBs for each main antibiotic class
satisfactory for human trials of ciclopirox
for the treatment of cancers41. On dosing Drug (target) ARBs for Gram-negative ARBs for Gram-positive
bacteria bacteria
radiolabelled ciclopirox to humans, 96%
of the label was recovered from urine; Carbapenems, • Ciclopirox • Curcumin
cephalosporins and • Loperamide (intravenous) • EGCG
however, a specific plasma or serum assay penicillins (cell wall • Macrolides • Berberine
for ciclopirox olamine was not used in synthesis) • EGCG
this study43. The authors stated that the • Naloxone, naltrexone and
pharmacological drug levels of ciclopirox curcumin (for gut pathogens and
required for anti-tumour activity are LPS-driven endotoxic shock)
achievable even though its half-life is short, Polymyxins (cell membrane) Loperamide NA
but require dosing several times a day. Aminoglycosides None identified NA
They noted, however, that the drug seemed (protein synthesis)
to be well absorbed after oral administration. Fluoroquinolones None identified Curcumin
As a result of this analysis, this group spon- (nucleic acid synthesis)
sored a Phase I study 44, which demonstrated Tetracyclines Loperamide Curcumin
that oral ciclopirox olamine achieved (protein synthesis)
plasma levels of 50 ng per ml after a single
Glycopeptides NA Naloxone, naltrexone and
oral dose of 20–40 mg per m2 and had (cell wall synthesis) curcumin (with vancomycin
biological activity in patients with advanced or metronidazole for
haematological malignancies. Dose-limiting the treatment of CDAD)
gastrointestinal toxicities were observed Macrolides NA None identified
in patients receiving the highest oral dose (protein synthesis)
administered four times a day, but not at ‘Gram-positive antibiotics’ Loperamide NA
lower doses or at a less frequent dosing ARBs, antibiotic resistance breakers; CDAD, Clostridium difficile-associated diarrhoea; EGCG, epigallocatechin‑3
schedule. Dosing regimens against bacteria gallate; LPS, lipopolysaccharide; NA, not applicable.
are likely to be shorter than those for the
treatment of cancer, and gastrointestinal
toxicity is less likely if the drug is adminis- tetracycline antibiotics against Gram-negative nutraceuticals for similar effects against a
tered intravenously as an ARB against life- pathogens (but not Gram-positive patho- wide range of Gram-positive antibiotics.
threatening bacteria in a hospital setting, gens) and was active in combination with the One concern is that repurposing Gram-
where the need for ARBs is greatest. broad-spectrum tetracycline-class antibiotic positive antibiotics for Gram-negative
Ciclopirox has at least two of the three minocycline in a mouse model of salmonel- pathogens could promote resistance to these
properties suggested for an ARB as it is losis. In addition to tetracyclines, loperamide agents by transfer of resistance determinants,
directly antibacterial and it induces the host increased the efficacy of cephalosporins but in view of the greater threat of
defence response by causing autophagy. (but not other cell-wall-directed antibiotics) Gram-negative organisms this may
However, data are required on whether or and the outer-membrane-permeating anti­ be a risk worth taking.
not this drug is effective in combination with biotic polymyxin B in vitro. The authors Loperamide is not orally bioavailable,
antibiotics. Moreover, its activity against a concluded that it is improbable that the but it could be used orally as an ARB to treat
broad range of bacteria needs investigation. synergy observed in vivo was the result gut infections, such as diarrhoeal diseases,
The appropriate clinical dose can then be of the antiperistaltic activity of loperamide, and intravenously for other infections if it is
calculated, and safety studies will be required as the potentiation was observed at proven to be safe. Although loperamide is
to assess the therapeutic index, particularly con­centrations of minocycline that do not an opioid, it has no opiate-like effects when
for intravenous dosing. Concomitant activity impair bacterial growth even upon prolonged administered orally or intravenously. It does
against C. albicans is a bonus, because exposure. not cross the blood–brain barrier because
Candida spp. are a leading cause of Loperamide has also been shown to it is subject to efflux by P‑glycoprotein;
catheter-associated infections, which sensitize Gram-negative bacteria to ‘Gram- therefore, intravenous loperamide could not
have high mortality rates. positive antibiotics’ (REF. 46). In the presence be used with inhibitors of P‑glycoprotein.
of loperamide, the aminocoumarin anti-
Loperamide. This μ‑opioid receptor agonist biotic novobiocin inhibited the growth of Potential ARBs for Gram-positive bacteria
has long been used as an anti-motility agent E. coli. Loperamide may alter the cell shape Berberine. The plant-derived isoquinoline
in the treatment of diarrhoeal diseases. and small-molecule permeability of E. coli, alkaloid berberine has a long history of use
Ejim et al.45 recently showed that loperamide, similar to the mechanism through which for the treatment of several conditions.
which has no antibacterial activity per se, colistin boosts the effectiveness of vancomy- In particular, it has been used in traditional
acts synergistically with several classes of cin and rifampin47. The authors suggested medicine in Europe, Asia and the Americas
antibiotic. They screened off-patent non- that the altered cell shape may cause dysreg- to treat diarrhoea caused by Giardia lamblia
antimicrobial drugs as a source of molecules ulation of the influx and efflux machinery of as well as the Gram-negative bacteria E. coli,
that might synergize with antibiotics at Gram-negative bacteria and thereby enable Klebsiella spp. and Vibrio cholerae. It has
sub-MIC concentrations. Loperamide, the accumulation of otherwise-excluded broad-spectrum direct antibacterial activity
at a concentration of 16 μg per ml or greater, antibiotics. This concept could be further against staphylococcal, streptococcal
increased the antibacterial efficacy of eight exploited by screening current drugs and and enterococcal species, including MDR

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | DECEMBER 2015 | 825

© 2015 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

strains of Mycobacterium tuberculosis and kill Streptococcus pneumoniae and S. aureus When tested alone in vitro against
MRSA. In vitro, berberine is about ten- may be particularly relevant as these are clinical isolates of MRSA48, berberine
times more potent against Gram-positive among the most common bacterial causes showed moderate activity against all strains
bacteria than Gram-negative. Its ability to of pneumonia. tested, with MICs of 32–128 μg per ml.

Table 3 | Potential drugs excluded as high priority, based on the current lack of data
Candidate drug Current approved Summary of evidence Additional information required
for repurposing indication or
common usage
Aspirin Pain, inflammation • In vitro: reduces resistance to aminoglycosides in Confirmation of activity alone and in
and anti-platelet Klebsiella pneumoniae and enhances the susceptibility synergy with antibiotics against
of Helicobacter pylori to antibacterials140; however, it antibiotic-resistant MRSA. A safety
induces resistance to many other antibiotic classes141. assessment is needed for intravenous
• In vivo: reduces the incidence of Staphylococcus aureus dosing
in a rabbit model of aortic valve endocarditis142, is
additive or synergistic if combined with vancomycin in a
rabbit model of endocarditis caused by S. aureus143 and
reduced the prevalence of nasal S. aureus19and S. aureus
bacteremia20 in patients receiving haemodialysis
Diclofenac Pain and • In vitro: directly antibacterial against antibiotic- The MIC90 needs to be improved — it
inflammation sensitive and antibiotic-resistant clinical isolates of is typically 100 μg per ml, which is two
S. aureus, Listeria monocytogenes, Escherichia coli and orders of magnitude above the Cmax
Mycobacterium spp., including anti-plasmid activity21,22. exposure achieved with a 50 mg oral
Synergistic in combination with streptomycin against dose in humans. Direct or synergistic
E. coli and Mycobacterium spp., and with gentamicin antibacterial actions against drug-
against Listeria spp.144; blocked both cAMP-activated resistant Gram-negative bacteria should
and Ca2+-activated chloride secretion in intestinal be investigated at safe doses
epithelial cells infected with Vibrio cholerae23
• In vivo: Effective in mice in treating infections of
V. cholerae23, Salmonella spp.145, Listeria spp.144,
and Mycobacterium tuberculosis146
Ibuprofen Pain and • In vivo: mice infected with M. tuberculosis that were The in vitro and in vivo activity against
inflammation treated with ibuprofen lived longer than control the major drug-resistant Gram-negative
animals24,25. Oleocanthal, the active constituent of bacteria, alone and with antibiotics,
olive oil, affects the same receptor as ibuprofen and should be explored. A safety assessment
has antibacterial activity26 for intravenous dosing is also required
Ivermectin Clinical and • In vitro: inhibits growth of Chlamydia trachomatis in The effects of ivermectin in combination
veterinary epithelial cells27 with antibiotics should be investigated
nematode infections • In vivo: improved survival in mice subjected to ‘endotoxic against drug-resistant Gram-negative
shock’ with a lethal dose of LPS28, decreased levels of bacteria. Safety for intravenous dosing
inflammatory cytokines28 and activated autophagy147 should also be assessed
Lauric acid (active Neutraceutical • In vitro: inhibits the synthesis of most staphylococcal An exploration of in vitro and in vivo
metabolite is (coconut oil) toxins and other exoproteins29. Blocks induction, activity against the major drug-resistant
monolaurin) but not constitutive synthesis, of β-lactamase30 Gram-negative bacteria, alone and
with antibiotics, is necessary. A safety
assessment is also required for both oral
and intravenous dosing
Metformin Anti-diabetic • In vivo: enhances phagocytosis by macrophages For intestinal infections, the synergy with
in a mouse model of E. coli lung infection148. AMPK antibiotics against bacteria such as
activation with metformin increased the survival V. cholerae and E. coli needs to be assessed,
rate in mice challenged with LPS in an endotoxemia which may be possible at the current
model32, reduced cholera-toxin-mediated increases approved dose levels of metformin. For
in intestinal chloride secretion33 and decreased systemic infections, the plasma levels
disease severity in mice and humans infected with required for systemic effects to break
M. tuberculosis149,150 antibiotic resistance should be defined,
and their safety determined. It must be
determined whether AMPK activation in
the gut is sufficient for systemic synergy
with antibiotics139
Vitamin D Calcium absorption • In vitro: inhibits mycobacterial entry and survival Studies on IL‑32 and vitamin D3 should
and bone health, within macrophages through the induction of be extended to other bacteria. The levels
and tuberculosis151 autophagy34,152. IL‑32 stimulates the immune system of vitamin D3 required in vivo must be
to kill M. tuberculosis, but only in the presence of determined and the safety of the levels
sufficient vitamin D3 levels35 required for both oral and intravenous
dosing assessed
AMPK, 5ʹ-AMP-activated protein kinase; cAMP, cyclic AMP; Cmax, maximum concentration; IL‑32, interleukin‑32; LPS, lipopolysaccharide; MIC90, the minimum
concentration that inhibits 90% of bacterial isolates; MRSA, methicillin-resistant S. aureus.

826 | DECEMBER 2015 | VOLUME 14 www.nature.com/reviews/drugdisc

© 2015 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

Ninety per cent inhibition of MRSA growth Gram-negative bacteria-induced diarrhoeal concentration of 128 μg per ml, which is
was obtained with a concentration of 64 μg diseases despite its lower in vitro activity obtainable in the colon through ingestion
per ml or less of berberine. These effects against Gram-negative bacteria than against of food or by dosage in capsules66. In clini-
may be due, at least in part, to inhibition of Gram-positive species. cal practice, ingestion of 4 g per day would
the Gram-positive bacterial sortase enzyme, During infection, berberine drives achieve this concentration in the gut66. In
an important anti-virulence target 49. In the suppression of pro-inflammatory responses regions where curcumin is a regular dietary
Gram-negative E. coli, berberine targets through activation of 5ʹ-AMP-activated ingredient it is typically consumed at 2–4 g
assembly of the cell division protein FtsZ50. protein kinase (AMPK) in macrophages54, a per day.
It inhibits the assembly kinetics of the Z‑ring property that could also lead to antibacterial Curcumin also synergizes with antibiotics.
and perturbs cytokinesis. It also destabilizes activity via autophagy. Mechanistic studies In combination studies with cefaclor,
FtsZ protofilaments and inhibits the FtsZ have shown that berberine downregulates cefodizime, or cefotaxime, concentrations of
GTPase activity. Berberine binds to expression of proinflammatory genes such as 0.1–1.0 μg per ml reduced the MIC values by
FtsZ with high affinity (dissociation tumour necrosis factor (TNF), interleukin-1β three- to eightfold against diarrhoea-causing
constant (KD) = 0.023 μM) and thus halts (IL1B), IL6, monocyte chemotactic protein 1 bacteria, such as E. coli and V. cholerae,
the first stage in bacterial cell division. (MCP1; also known as CCL2), inducible as well as against another Gram-negative
In vivo, berberine protected mice nitric oxide synthase (iNOS; also known species, P. aeruginosa, and the Gram-positive
challenged with Salmonella enterica subsp. as NOS2), and cyclooxygenase 2 (COX2; S. aureus 67. Against MRSA, curcumin
enterica serovar Typhimurium: 50% of the also known as PTGS2) (REF. 54). potentiated the antimicrobial action of
mice that did not receive berberine treatment Metformin, a commonly used antidiabetic cefixime, cefotaxime, vancomycin, tetra­
died by the end of the eighth day after infec- agent, also activates AMPK, and so cycline, oxacillin, ampicillin, ciprofloxacin
tion, whereas, with doses of 10, 20, 30 and berberine has been studied as an antidiabetic and norfloxacin68,69.
40 mg per kg of berberine, 60, 60, 70 and agent 55,56. In patients with type 2 diabetes, Numerous potential mechanisms of
90% of the infected mice survived to berberine has been reported to have an action have been reported, including inhi-
the eighth day, respectively 51. efficacy equivalent to that of metformin57. bition of sortase70. Curcumin also induces
Encouragingly, another study reported Berberine has also been investigated as a autophagy by inhibiting the AKT–mTOR
that bacteria are poor at generating resist- chemotherapeutic agent, and so its potential pathway 71. Its chemical structure (a poly­
ance to berberine52. The MICs of bacterial for cell toxicity would need to be accounted phenol with the ability to bind to many
cultures (E. coli, S. aureus, Bacillus subtilis, for if it were to be used in any human studies proteins through ionic and hydrogen bonds)
Proteus vulgaris, S. Typhimurium and as an ARB. The bioavailability of berberine may explain its promiscuous activity (TABLE 1).
P. aeruginosa) did not increase over 200 is reportedly less than 5% owing to poor In host-defence studies, curcumin blocks
generations despite treatment with berberine absorption and rapid clearance. Berberine the binding of the LPSs from Gram-negative
at a concentration of 50% of its MIC. seems to be subject to P-glycoprotein- bacteria to MD2 in the TLR4–MD2
There is also evidence that berberine mediated efflux from the intestine and liver. complex 72,73 and downregulates expression
could be an ARB48: berberine markedly Absorption has been enhanced with sodium of intestinal TLR2, TLR4 and TLR9
lowered the MICs of ampicillin and oxacillin caprate, a medium chain fatty acid found in (REFS 74,75). It also decreases the production
against MRSA; an additive effect was found milk fat and coconut oil. However, bioavaila- of TNF, IL‑1, IL‑2, IL‑6, IL‑8 and IL‑12,
between berberine and ampicillin; and a syn- bility issues do not seem to have been limiting MCP1 and migration and invasion-inhibitory
ergistic effect was found between berberine in the human studies reported above. protein76–80. The ability of curcumin to block
and oxacillin48. In the presence of 1–50 μg the interaction between MD2 and bacterial
per ml berberine, levels of MRSA adhesion Potential ARBs for both bacterial classes proteins could also explain its efficacy
and intracellular invasion were notably Curcumin. Curcumin is a constituent of the in treating Gram-positive infections.
decreased compared with the vehicle-treated popular spice turmeric, which has been used C. difficile is a Gram-positive species with
control group. These results suggest that for centuries in both cooking and traditional no LPS coat, but its surface layer proteins
berberine may have direct antimicrobial medicine across the Indian subcontinent. are recognized by the MD2 component of
activity, the potential to restore the effective- It is currently being investigated for efficacy the TLR4–MD2 complex in monocytes and
ness of β-lactam antibiotics against MRSA, against a number of diseases, including epithelial cells, stimulating NF‑kB activation
and the ability to inhibit MRSA adhesion and cancer, and against mechanisms of ageing 58,59. and causing apoptotic intestinal epithelial
intracellular invasion. Berberine may also Curcumin has direct antibiotic activity cell detachment 81,82.
have ARB activity by increasing the host at concentrations of 125–1,000 μg per ml Through the inhibition of NF-κB83,
defence response. It protects against against a broad range of bacteria, including curcumin prevents the upregulation of IL‑8
LPS-induced intestinal injury in mice by some Gram-negative species (including expression in response to infection84.
inhibiting the TLR4–NF-κB–MIP2 (Toll-like E. coli, P. aeruginosa, V. cholerae, S. aureus IL‑8 levels are elevated in patients with severe
receptor 4–nuclear factor κB–macrophage and B. subtilis)60. Although curcumin C. difficile colitis85,86; in these patients, disease
inflammatory protein 2 (also known as had some antibacterial effects against severity correlates with increased levels of
CXCL2)) pathway in ileal cells and decreasing Helicobacter pylori infections in vitro61 IL‑8, IL‑6 and eotaxin, and IL‑8 expression
neutrophil infiltration53. Berberine can also and in animal studies62, human studies correlates with treatment failure after
act as an LPS antagonist by binding to have produced mixed results63–65. metronidazole and vancomycin therapy 87,88.
TLR4–MD2 (also known as LY96) complexes There is also evidence supporting use of Curcumin may be an effective ARB for
and blocking LPS–TLR4 signalling in murine curcumin against the Gram-positive organ- patients with C. difficile infections by modu-
macrophage-like cells (RAW 264.7)51. This ism C. difficile. In vitro, curcumin inhibited lating their gut cytokine response, especially
may explain its reported effectiveness against the growth of 21 strains of C. difficile at a in those patients with relapsing infection.

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | DECEMBER 2015 | 827

© 2015 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

Well-controlled oral intervention trials of its supposed therapeutic benefits. EGCG of EGCG given orally to septic mice
studying the use of curcumin to treat has been extensively studied in many disease (4 mg per kg, which is 10 mM) were
C. difficile infections are lacking. In one trial, areas and written about in several thousand comparable to those achievable in humans
the drug, in the form of turmeric, was dosed scientific publications, with most published after ingestion of a few cups of green tea
by enema89. In this trial, the turmeric enemas over the past two decades. The anti-infective (1 mM). Importantly, delayed and repeated
were as effective as vancomycin enemas for effect of green tea was first reported more administration of EGCG beginning 24 hours
treating C. difficile colitis — the infection than 100 years ago by Major J. G. McNaught, after onset of sepsis substantially rescued
was eradicated in 76% and 83% of patients, an army surgeon who showed that green mice from lethal sepsis, supporting a
respectively, compared with 66% in the tea killed the Gram-negative organisms that therapeutic potential for EGCG in the
placebo group. At 21 days post-treatment, lead to typhoid fever (Salmonella enterica clinical management of sepsis.
clinical severity was reduced by 60% in the subsp. enterica serovar Typhi) and brucellosis The mechanisms by which EGCG exerts
vancomycin and turmeric groups, compared (Brucella melitensis)94. its effects on bacteria seem to be very broad;
to a reduction of 38% in the placebo group. Two recent comprehensive reviews95,96 this is probably due to its chemical structure,
Recurrence developed in 10% of patients have detailed mild antibiotic activity of which contains phenolic groups capable
treated with vancomycin, 9% of those in the EGCG alone in vitro and substantial synergy of making ionic and hydrogen bonds with
turmeric group and 29% of patients who of EGCG with a broad range of antibiotics to multiple proteins. EGCG binds to the
received the placebo. treat both Gram-positive and Gram-negative peptidoglycans of the bacterial cell wall and
Some authors have reported that organisms, particularly if antibiotic resistance inhibits penicillinase activity, protecting
curcumin has poor bioavailability 90, which is is present. EGCG has positive synergistic penicillins from inactivation106. It also alters
due to a combination of adverse properties: effects, although the occasional adverse effect the cell wall of S. aureus 107. It has been sug-
poor aqueous solubility, poor absorption and on resistance in vitro has been reported101. gested that the ability of EGCG to reverse
rapid conjugative clearance. However, neuro­ EGCG can sensitize MRSA to all methicillin resistance is mediated by inhibi-
scientists have reported therapeutic levels types of β-lactam antibiotics, including tion of the synthesis of the penicillin-binding
in the brain following oral dosing of either benzylpenicillin, oxacillin, methicillin, protein 2a (PBP2a) as well as inhibition of
curcumin or preparations with enhanced ampicillin, carbapenems and cephalexin97–100. β-lactamase secretion97. In addition, EGCG
bioavailability 91. Brain levels reached 3 μM The fractional inhibitory concentration inhibits DNA gyrase108, dihydrofolate reduc-
for curcumin and 6 μM for tetrahydro- (FIC) indices of β-lactams tested against 25 tase102 and specific reductases (FabG and
curcumin92 and positive effects have been isolates of MRSA ranged from 0.126 to 0.625 FabI) in bacterial type II fatty acid synthesis109.
reported in animal models of Alzheimer when used in combination with EGCG at EGCG also blocked H. pylori binding to
disease91,93. Similar efficacy was observed 6.25, 12.5 or 25 μg per ml. When used in TLR4 on gastric epithelial cells110, inhibited
after intraperitoneal dosing in a model of combination with three carbapenems that conjugative transfer of the R plasmid in
cerebral ischaemia in rats73, suggesting that do not usually show strong activity against E. coli 111 — which could lead to decreased
bioavailability may not be limiting. MRSA, EGCG showed additive and syner- sharing of antimicrobial genes between
Further exploration of the ability of gistic effects, bringing potency to within a bacteria — and inhibited the activity of the
curcumin to synergize with antibiotics60 and useful range: the MICs of imipenem in the streptococcal efflux pump Tet(K), which
potentially reduce antibiotic resistance in presence of EGCG at 3.125, 6.25, 12.5 and is involved in resistance to tetracycline112.
bacterial pathogens of the gastrointestinal 25 μg per ml were restored to the susceptibility However, the effect of EGCG on a range
tract is warranted. In view of the diversity breakpoint (<4 μg per ml) for 8, 38, 46 and of bacterial efflux pumps needs further
of opinions on bioavailability, it would seem 75% of the MRSA isolates, respectively, thus definition. Additionally, EGCG activates
wise to focus additional oral investigations rendering these bacteria ‘susceptible’ (REF. 101). host defence and, therefore, it may be effec-
on those bacteria that cause diarrhoeal EGCG is able to break the resistance of many tive at lower plasma concentrations than
diseases and/or gain entry through the gut. bacteria to β-lactams and carbapenems, and would be expected by simple extrapolation
These can over-stimulate TLR4 in particular, it also increased the efficacy of inhibitors of from in vitro data. EGCG also increases
causing a ‘cytokine storm’ and excessive protein or nucleic acid synthesis102. However, autophagy 113,114, possibly through activation
inflammation that aids their entry into EGCG may have adverse effects when of AMPK115.
sterile gut wall tissues and the bloodstream. combined with glycopeptide antibiotics Human trials of EGCG in combination
The ability of curcumin to dampen down (vancomycin or teicoplanin)101. Importantly, with antibiotics are required. EGCG has rela-
this excessive inflammatory response may EGCG seems to have no adverse effects on tively poor bioavailability in animals and it
lead to preventative or treatment options for commensal bacteria103. is unclear whether plasma levels in humans
gut-invading Gram-negative bacteria, such EGCG may also be useful in treating would be sufficiently high to exert a syner-
as Salmonella spp., Shigella spp., and E. coli, Gram-negative infections. In vitro, EGCG gistic effect with antibiotics116. Prodrugs of
as well as the Gram-positive C. difficile. killed MDR and extended-spectrum EGCG could improve bioavailability 117,118.
Intravenous doses of curcumin may be effec- β-lactamase (ESBL)-producing strains of However, EGCG might be most useful as an
tive against systemic infections in which the E. coli that were isolated from urinary tract ARB in topical infections, such as MRSA,
cytokine storm has devastating effects, such infections104. In vivo, green tea and EGCG and possibly as an oral treatment for
as Gram-negative bacteria-mediated sepsis. dose-dependently abrogated endotoxin- gastrointestinal infections. In addition,
induced high mobility group protein B1 EGCG could be useful as an intravenous
Epigallocatechin‑3‑gallate (EGCG). (HMGB1) release from murine macrophage- agent combined with carbapenems or other
Epigallocatechin‑3‑gallate (EGCG) is one like RAW 264.7 cells and dose-dependently antibiotics against diseases caused by systemic
of the most abundant polyphenols in green protected mice against lethal endotoxemia MRSA infection, such as pneumonia,
tea and is thought to be responsible for most and sepsis105. The authors noted that doses septicemia and urinary tract infections.

828 | DECEMBER 2015 | VOLUME 14 www.nature.com/reviews/drugdisc

© 2015 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

(+)-naloxone and (+)-naltrexone. Box 2 | Regulatory pathways for repurposed drugs


The opioid antagonist (−)-naloxone and the
non-opioid (+)-naloxone inhibit TLR4 sig- In the United States, the US Food and Drug Administration (FDA) has in place a regulatory
nalling and block the MD2–TLR4‑mediated pathway, 505(b)(2), that applies to a new use or new formulation of an approved drug. It allows
inflammatory side effect of opioids. the applicant to use the existing safety, pharmacology and toxicology data for regulatory
purposes, provided that the doses and exposure used are the same or lower. The application can
They may bind preferentially to the LPS
refer to published literature, product labels and product monographs. Parallel regulations apply
binding pocket of MD2 rather than to to investigational new drug filings.
TLR4 (REF. 119). (+)-naloxone has no known Europe and other regions have similar regulations. The intellectual property could not be
off-target effects120, however, its half-life protected with a composition of matter claim, but in many countries a ‘method of use’ patent
is short, whereas the structurally related could be filed for the discovery of a new indication for an old drug that is novel, unexpected and of
(+)-naltrexone has a half-life of 4–6 hours value to humanity. Together with a use claim, the development of a new formulation, possibly
in humans121. incorporating a different dose or route of administration, can further support market exclusivity.
These (+)-isomers are devoid of opioid- Another possibility is to use repurposed drugs off-label. The properties of a repurposed drug
like activity, and theoretically they could be could be publicized through scientific literature and conferences. If the appropriate formulation
used to treat some bacterial infections, as is the same, the drug could be used without formal regulatory approval for the secondary use.
However, although a physician may prescribe a drug for a use other than the approved
blocking TLR4­–MD2 in the gut could
indication, drug companies have been sanctioned severely for marketing products along these
prevent bacterial LPS from triggering a lines. In addition, some payers, under certain conditions, restrict the reimbursement of products
cytokine storm driven by IL‑6 and IL‑8 and that are used off-label; the prescribing physician also incurs a greater element of product
thereby prevent invasion by gut pathogens. liability. Regulatory approval avoids these problems and may also be preferable as it safeguards
These or other opioid antagonists could standards of quality.
be useful if co‑administered orally with
anti­biotics to treat E. coli or Shigella spp.
intestinal infections or to prevent CDAD that EDTA could be used as an ARB124. known pharmacological, pharmacokinetic
in the elderly. They could also be useful if Future research aimed at identifying and safety profiles is possibly the best-case
administered intravenously for the treatment ARBs for these classes could be highly scenario for low-risk drug development.
of LPS-driven systemic endotoxic shock valuable. The data on the use of these molecules as
because of their potential to block the In the reviewed literature, most of the ARBs come from many different laboratories
release of pro-inflammatory cytokines. potential ARBs discussed above are shown using diverse methodologies and often give
to be effective as directly antibacterial and/or a range of potencies for each molecule.
Conclusions and next steps additive to antibiotics at concentrations The data need to be confirmed against the
Several potential ARBs are available for of 25 μg per ml or lower, which is the level current most lethal strains to enable calcula-
β-lactam antibiotics (carbapenems, cephalo­ required for interest by consortia such as tion of the plasma concentrations of ARBs
sporins and penicillins), which is the most the Innovative Medicines Initiative (IMI)’s that will be required. It will then be neces-
important class of antibiotic for the treat- ENABLE project, a European Union initia- sary to conduct safety assessments to
ment of antibiotic-resistant Gram-negative tive that supports new drug development determine a therapeutic index for each ARB.
bacteria. Several of these ARBs disrupt the of antibacterials against Gram-negative The agents considered in this Perspective are
bacterial cell wall, which contains poly- organisms. Ciclopirox, loperamide, cur- all ingested by humans today, so they already
anionic LPS and is stabilized by the cross- cumin, EGCG and berberine are potential have a lengthy safety record; however, their
bridging of divalent cations122. Drugs that ARBs with encouraging data at 25 μg per safety does need to be confirmed for the
target these divalent cations destabilize the ml. Most of the drugs considered in this particular doses, combinations and routes of
membrane, increase its permeability and Perspective have direct antibiotic activity: administration that would be used to treat
allow access of molecules that were partially sometimes they exhibit additive effects bacterial infections.
or fully excluded. Indeed, several polycation with antibiotics, and sometimes they Additionally, there are regulatory
antibiotics — for example, polymyxins, synergize with co‑administered antibiotics considerations that need to be addressed
aminoglycosides and dibasic macrolides at 0.50–50 μg per ml. For the treatment of (BOX 2). Will regulatory authorities require
such as azithromycin — act through this systemic infections, the maximum dose three‑way clinical trials, comparing each
mechanism123 and these are ARBs them- of drug approved for use in humans must, drug individually with the combination?
selves when co‑administered with β-lactam at minimum, achieve this level of exposure Do we have the time to perform such perfect
antibiotics. In addition to potentially at the maximum concentration (Cmax). clinical trials? The Ebola epidemic in West
salvaging our best Gram-negative anti- For topical (including gastrointestinal) Africa has shown that society is now
biotics, this approach may also make infections, Cmax may not be relevant because amenable to fast-track development of new
‘Gram-positive antibiotics’ useful against high local concentrations of both drug drugs when a global emergency dictates it.
Gram-negative bacteria. Further careful and antibiotic will be present, with a much Profit margins for combinations of
screening of polycation molecules in the higher probability of achieving therapeutic known drugs may be low even if they are
drug pharmacopeia may identify new concentrations at the relevant site. The first lifesavers. Pharmaceutical or biotechnology
ARBs of this type. application of ARBs to reach the clinic companies are unlikely to invest in this
No compelling ARBs were identified for would almost certainly be in topical and approach, although those that currently sell
two classes of antibiotics that are particularly gastrointestinal infections, although the antibiotics may be able to preserve sales and
useful, the fluoroquinolones and the amino- most urgent clinical need is for intravenous reach new patents by adding an ARB in a
glycosides, although a report of successful agents against Gram-negative bacteria. new combination. In particular, as no ARBs
use of EDTA with gentamycin may indicate The combination of two known drugs with have been identified for fluoroquinolones

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | DECEMBER 2015 | 829

© 2015 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

and aminoglycosides, a screening campaign 17. Molinari, G. et al. Inhibition of Pseudomonas 40. Zhou, H. et al. Ciclopirox induces autophagy through
aeruginosa virulence factors by subinhibitory reactive oxygen species-mediated activation of JNK
to find ARBs for these antibiotics could be concentrations of azithromycin and other macrolide signaling pathway. Oncotarget 5, 10140–10150
commercially viable if supported by ‘method antibiotics. J. Antimicrob. Chemother. 31, 681–688 (2014).
(1993). 41. Weir, S. J. et al. The repositioning of the anti-fungal
of use’ patents (BOX 2). However, in general, 18. Nguyen, T. et al. Potential role of macrolide agent ciclopirox olamine as a novel therapeutic agent
unless a new economic model is developed antibiotics in the management of cystic fibrosis lung for the treatment of haematologic malignancy.
disease. Curr. Opin. Pulmonary Med. 8, 521–528 J. Clin. Pharm. Ther. 36, 128–134 (2011).
for antibiotics, the development of ARBs (2002). 42. Eberhard, Y. et al. Chelation of intracellular iron with
will have to be pursued by government, 19. Karabay, O. et al. A new effect of acetylsalicylic acid? the antifungal agent ciclopirox olamine induces cell
Significantly lower prevalence of nasal carriage of death in leukemia and myeloma cells. Blood 114,
public sector or philanthropic agencies, Staphylococcus aureus among patients receiving 3064–3073 (2009).
or combinations of these. orally administered acetylsalicylic acid. Infect. Control 43. Kellner, H. M. et al. Pharmacokinetics and
Hosp. Epidemiol. 27, 318–319 (2006). biotransformation of the antimycotic drug
20. Sedlacek, M. et al. Aspirin treatment is associated ciclopiroxolamine in animals and man after topical and
David Brown is at Alchemy Biomedical Consulting, with a significantly decreased risk of Staphylococcus systemic administration. Arzneimittelforschung 31,
St Johns Innovation Centre, Cowley Road, aureus bacteremia in hemodialysis patients with 1337–1353 (in German) (1981).
Cambridge CB4 0WS, UK. tunneled catheters. Am. J. Kidney Dis. 49, 401–408 44. Minden, M. D. et al. Oral ciclopirox olamine displays
e-mail: davidbrown1000@btinternet.com (2007). biological activity in a phase I study in patients with
21. Mazumdar, K. et al. Diclofenac in the management of advanced hematologic malignancies. Am. J. Hematol.
doi:10.1038/nrd4675 E. coli urinary tract infections. In Vivo 20, 613–619 89, 363–368 (2014).
Published online 23 October 2015; (2006). 45. Ejim, L. et al. Combinations of antibiotics and
corrected online 29 January 2016 22. Mazumdar, K. et al. The anti-inflammatory nonantibiotic drugs enhance antimicrobial efficacy.
non-antibiotic helper compound diclofenac: Nat. Chem. Biol. 7, 348–350 (2011).
an antibacterial drug target. Eur. J. Clin. Microbiol. 46. Taylor. P. L. et al. A forward chemical screen identifies
1. Rice, L. B. Federal funding for the study of Infect. Dis. 28, 881–891 (2009). antibiotic adjuvants in Escherichia coli. ACS Chem.
antimicrobial resistance in nosocomial pathogens: 23. Pongkorpsakol, P. et al. Inhibition of cAMP-activated Biol. 7, 1547–1555 (2012).
no ESKAPE. J. Infect. Dis. 197, 1079–1081 (2008). intestinal chloride secretion by diclofenac: cellular 47. Tascini, C. et al. Synergistic activity of colistin
2. Woodford, N., Wareham, D. W., Guerra, B. & Teale, C. mechanism and potential application in cholera. plus rifampin against colistin-resistant
Carbapenemase-producing Enterobacteriaceae and PLoS Negl. Trop. Dis. 8, e3119 (2014). KPC-producing Klebsiella pneumoniae.
non-Enterobacteriaceae from animals and the 24. Vilaplana, C. et al. Ibuprofen therapy resulted in Antimicrob. Agents Chemother. 57, 3990–3993
environment: an emerging public health risk of our significantly decreased tissue bacillary loads and (2013).
own making? J. Antimicrob. Chemother. 69, 287–291 increased survival in a new murine experimental 48. Yu, H.‑H. et al. Antimicrobial activity of berberine
(2014). model of active tuberculosis. J. Infect. Dis. 208, alone and in combination with ampicillin or oxacillin
3. Davis, S. C. Infections and the rise of antimicrobial 199–202 (2013). against methicillin-resistant Staphylococcus aureus.
resistance. UK Government [online], https://www.gov. 25. Eisen, D. P. et al. Low-dose aspirin and ibuprofen J. Med. Food 8, 454–461 (2005).
uk/government/uploads/system/uploads/attachment_ sterilizing effects on Mycobacterium tuberculosis 49. Kim, S.‑H. et al. Inhibition of the bacterial surface
data/file/138331/CMO_Annual_Report_ suggest safe new adjuvant therapies for tuberculosis. protein anchoring transpeptidase sortase by
Volume_2_2011.pdf (2015) J. Infect. Diseases 208, 1925–1927 (2013). isoquinoline alkaloids. Biosci. Biotechnol. Biochem.
4. Centers for Disease Contol and Prevention. 26. Cicerale, S., Lucas, L. J. & Keast, R. S. Antimicrobial, 68, 421–424 (2004).
Antibiotic resistance threats in the United States, antioxidant and anti-inflammatory phenolic activities 50. Domadia, P. N. Berberine targets assembly of
2013. CDC [online], http://www.cdc.gov/ in extra virgin olive oil. Curr. Opin. Biotechnol. 23, Escherichia coli cell division protein FtsZ. Biochemistry
drugresistance/pdf/ar-threats-2013-508.pdf (2013). 129–135 (2012). 47, 3225–3234 (2008).
5. Bassetti, M. & Righi, E. Eravacycline for the treatment 27. Pettengill, M. et al. Ivermectin inhibits growth of 51. Chu, M. et al. Role of berberine in anti-bacterial as a
of intra-abdominal infections. Expert Opin. Investigat. Chlamydia trachomatis in epithelial cells. PLoS ONE high-affinity LPS antagonist binding to TLR4/MD‑2
Drugs 23, 1575–1584 (2014). 7, e48456 (2012). receptor. BMC Complement. Altern. Med. 14, 89
6. Walkty, A. et al. In vitro activity of plazomicin against 28. Zhang, X. et al. Ivermectin inhibits LPS-induced (2014).
5015 Gram-negative and Gram-positive clinical isolates production of inflammatory cytokines and improves 52. Jin, J. L. et al. Antibacterial mechanisms of berberine
obtained from patients in Canadian hospitals as part of LPS-induced survival in mice. Inflamm. Res. 57, and reasons for little resistance of bacteria. Chinese
the CANWARD study, 2011–2012. Antimicrob. Agents 524–529 (2008). Herbal Med. 3, 27–35 (2010).
Chemother. 58, 2554–2563 (2014). 29. Schlievert, P. M. et al. Effect of glycerol monolaurate 53. Li, H.‑M. et al. Berberine protects against
7. Zhanel, G. G. et al. Ceftazidime–avibactam: a novel on bacterial growth and toxin production. Antimicrob. lipopolysaccharide-induced intestinal injury
cephalosporin/β-lactamase inhibitor combination Agents Chemother. 36, 626–631 (1992). in mice via α 2 adrenoceptor-independent
Drugs.73, 159–177 (2013). 30. Projan, S. J. et al. Glycerol monolaurate inhibits the mechanisms. Acta Pharmacol. Sin. 32, 1364–1372
8. Zhanel, G. G. et al. Ceftolozane/tazobactam: a novel production of β-lactamase, toxic shock syndrome (2011).
cephalosporin/β-lactamase inhibitor combination with toxin‑1, and other staphylococcal exoproteins by 54. Jeong, H. W. et al. Berberine suppresses
activity against multidrug-resistant gram-negative interfering with signal transduction. J. Bacteriol. proinflammatory responses through AMPK activation
bacilli. Drugs.74, 31–51(2014). 176, 4204–4209 (1994). in macrophages. Am. J. Physiol. Endocrinol. Metab.
9. White, A. R. et al. Augmentin (amoxicillin/clavulanate) 31. Zhao, X. et al. Activation of AMPK attenuates 296, 955–964 (2009).
in the treatment of community-acquired respiratory neutrophil proinflammatory activity and decreases the 55. Zhang, M. & Chen, L. Berberine in type 2 diabetes
tract infection: a review of the continuing development severity of acute lung injury. Am. J. Physiol. Lung Cell. therapy: a new perspective for an old antidiarrheal
of an innovative antimicrobial agent. J. Antimicrob. Mol. Physiol. 295, 497–504 (2008). drug? Acta Pharmaceutica Sinica B 2, 379–386
Chemother. 53 (Suppl. 1), i3–i20 (2004). 32. Tsoyi, K. et al. Metformin inhibits HMGB1 release in (2012).
10. Prabhudesai, P. P. et al. The efficacy and safety of LPS-treated RAW 264.7 cells and increases survival 56. Zhang, H. et al. Berberine lowers blood glucose in
amoxicillin-clavulanic acid 1000/125mg twice daily rate of endotoxaemic mice. Br. J. Pharmacol. 162, type 2 diabetes mellitus patients through increasing
extended release (XR) tablet for the treatment of 1498–1508 (2010). insulin receptor expression. Metabolism 59,
bacterial community-acquired pneumonia in adults. 33. Rogers, A. C. et al. Activation of AMPK inhibits 285–292 (2009).
J. Indian Med. Assoc. 109, 124–127 (2011). cholera toxin stimulated chloride secretion in human 57. Yin, J., Xing, H. & Ye, J. Efficacy of berberine in
11. Coates, A. & Hu, Y. in Novel Antimicrobial Agents and murine intestine. PLoS ONE 8, e69050 (2013). patients with type 2 diabetes mellitus. Metabolism
and Strategies Ch. 2 (eds Phoenix, D. A., Harris, F. 34. Yuk, J. M. et al. Vitamin D3 induces autophagy in 57, 712–717 (2008).
& Dennison, S. R.) (Wiley, 2014). human monocytes/ macrophages via cathelicidin. 58. Fürst, R. & Zündorf, I. Plant-derived
12. Blair, J. M., Richmond, G. E. & Piddock, L. J. Cell Host Microbe 6, 231–234 (2009). anti-inflammatory compounds: hopes and
Multidrug efflux pumps in Gram-negative bacteria and 35. Montoya, D. et al. IL‑32 is a molecular marker disappointments regarding the translation of
their role in antibiotic resistance. Future Microbiol. 9, of a host defense network in human tuberculosis. preclinical knowledge into clinical progress.
1165–1177 (2014). Sci. Transl. Med. 20, 250 (2014). Mediators Inflamm. 2014, 146832 (2014).
13. Amsden, G. W. Anti-inflammatory effects of macrolides 36. Dittmar, W. et al. Microbiological laboratory studies 59. Gupta, S. C., Patchva, S. & Aggarwal, B. B.
— an under-appreciated benefit in the treatment of with ciclopiroxolamine. Drug Res. 31, 1317–1322 Therapeutic roles of curcumin: lessons learned from
community-acquired respiratory tract infections and (1981). clinical trials. AAPS J. 15, 195–218 (2013).
chronic inflammatory pulmonary conditions? 37. Carlson-Banning, K. M. et al. Toward repurposing 60. Moghadamtousi, S. Z. et al. A review on antibacterial,
J. Antimicrob. Chemother. 55, 10–21 (2005). Ciclopirox as an antibiotic against drug-resistant antiviral, and antifungal activity of curcumin.
14. Kudoh, S. et al. Improvement of survival in patients Acinetobacter baumannii, Escherichia coli, and Biomed. Res. Int. 186864 (2014).
with diffuse panbronchiolitis treated with low dose Klebsiella pneumoniae. PLoS ONE 8, e69646 61. Mahady, G. B. et al. Turmeric (Curcuma longa) and
erythromycin. Amer. J. Resp. Crit. Care Med. 157, (2013). curcumin inhibit the growth of Helicobacter pylori,
1829–1832 (1998). 38. Niewerth, M. et al. Ciclopirox olamine treatment a group 1 carcinogen. Anticancer Res. 22 4179–4181
15. Kudoh, S. et al. Clinical effects of low-dose affects the expression pattern of Candida albicans (2002).
long-term erythromycin chemotherapy on diffuse genes encoding virulence factors, iron metabolism 62. De, R. et al. Antimicrobial activity of curcumin against
panbronchiolitis. Nihon Kyobu Shikkan Gakkai Zasshi proteins, and drug resistance factors. Antimicrob. Helicobacter pylori isolates from India and during
25, 632–642 (in Japanese) (1987). Agents Chemother. 47, 1805–1817 (2003). infections in mice. Antimicrob. Agents Chemother.
16. Tateda, K. et al. Azithromycin inhibits quorum sensing 39. Dihazi, G. H. et al. Impact of the antiproliferative 53, 1592–1597 (2009).
in Pseudomonas aeruginosa. Antimicrob. Agents agent ciclopirox olamine treatment on stem cells 63. Aljamal, A. Effect of turmeric in peptic ulcer and H
Chemother. 45, 1930–1933 (2001). proteome. World J. Stem Cells 5, 9–25 (2013). pylori. Plant Sci. Res. 3, 25–28 (2011).

830 | DECEMBER 2015 | VOLUME 14 www.nature.com/reviews/drugdisc

© 2015 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

64. Di Mario, F. et al. A curcumin-based 1‑week triple 86. Jafari, N. V. et al. Clostridium difficile modulates 109. Zhang, Y. M. & Rock, C. O. Evaluation of
therapy for eradication of Helicobacter pylori host innate immunity via toxin-independent and epigallocatechin gallate and related plant polyphenols
infection: something to learn from failure? dependent mechanism(s). PLoS ONE 8, e69846 as inhibitors of the FabG and FabI reductases of
Helicobacter 12, 238–243 (2007). (2013). bacterial type II fatty-acid synthesis. J. Biol. Chem.
65. Koosirirat, C. et al. Investigation of the 87. Rao, K. et al. The systemic inflammatory response to 279, 30994–31001 (2004).
antiinflammatory effect of Curcuma longa in Clostridium difficile infection. PLoS ONE 9, e92578 110. Lee, K. M. et al. Protective mechanism of
Helicobacter pylori-infected patients. (2014). epigallocatechin‑3‑gallate against Helicobacter
Int. Immunopharmacol. 10, 815–818 (2010). 88. Feghaly, R. et al. Markers of intestinal inflammation, pylori-induced gastric epithelial cytotoxicity via
66. Patel, R. & Yang, N. Inhibiting hospital associated not bacterial burden, correlate with clinical outcomes the blockage of TLR‑4 signaling. Helicobacter 9,
infection of toxigenic Clostridium difficile using natural in Clostridium difficile infection. Clin. Infect. Dis. 56, 632–642 (2004).
spice-turmeric (curcumin). Amer. J. Gastroenterol. 1713–1721 (2013). 111. Zhao, W. H. et al. Inhibition by epigallocatechin
105, S122–S122 (2010). 89. Basu, P. P. et al. Turmeric enema: a novel therapy for gallate (EGCG) of conjugative R plasmid transfer in
67. Sasidharan, N. K. et al. In vitro synergistic effect of C. difficile colitis (CDAD): A randomized, double Escherichia coli. J. Infect. Chemother. 7, 195–197
curcumin in combination with third generation blinded, placebo controlled prospective clinical trial. (2001).
cephalosporins against bacteria associated with Internat. J. Infectious Diseases 15 (Suppl. 15), S39 112. Sudano Roccaro, A. et al. Epigallocatechin-gallate
infectious diarrhea. Biomed. Res. Int. 2014, 561456 (2011). enhances the activity of tetracycline in staphylococci
(2014). 90. Sharma, R. A. et al. Pharmacodynamic and by inhibiting its efflux from bacterial cells. Antimicrob.
68. Moghaddam, K. M. et al. The combination effect pharmacokinetic study of oral curcuma extract in Agents Chemother. 48, 1968–1973 (2004).
of curcumin with different antibiotics against patients with colorectal cancer. Clin. Cancer Res. 113. Li, W. et al. EGCG stimulates autophagy and reduces
Staphylococcus aureus. Int. J. Green Pharm. 3, 7, 1894–1900 (2001). cytoplasmic HMGB1 levels in endotoxin-stimulated
141–143 (2009). 91. Lim, G. P. et al. The curry spice curcumin reduces macrophages. Biochem. Pharmacol. 81, 1152–1163
69. Mun, S. H. et al. Synergistic antibacterial effect of oxidative damage and amyloid pathology in an (2011).
curcumin against methicillin-resistant Staphylococcus Alzheimer transgenic mouse. J. Neurosci. 21, 114. Kim, H. S. et al. Epigallocatechin gallate (EGCG)
aureus. Phytotherapy Research 19, 599–604 8370–8377 (2001). stimulates autophagy in vascular endothelial cells:
(2013). 92. Begum, A. N. et al. Curcumin structure function, a potential role for reducing lipid accumulation.
70. Park, B. S. et al. Curcuma longa L. constituents inhibit bioavailability, and efficacy in models of J. Biol. Chem. 288, 22693–22705 (2013).
sortase A and Staphylococcus aureus cell adhesion neuroinflammation and Alzheimer’s disease. 115. Zhou, J. et al. Epigallocatechin‑3‑gallate (EGCG), a
to fibronectin. J. Agr. Food Chem. 53, 9005–9009 J. Pharmacol. Exp. Ther. 326, 196–208 (2008). green tea polyphenol, stimulates hepatic autophagy
(2005). 93. Yang, F. et al. Curcumin inhibits formation of amyloid-β and lipid clearance. PLoS ONE 9, e87161 (2014).
71. Aoki, H. et al. Evidence that curcumin suppresses oligomers and fibrils, binds plaques, and reduces 116. Ullmann, U. et al. A single ascending dose study
the growth of malignant gliomas in vitro and in vivo amyloid in vivo. J. Biol. Chem. 280, 5892–5901 of epigallocatechin gallate in healthy volunteers.
through induction of autophagy: role of Akt and (2005). J. Int. Med. Res. 31, 88–101 (2003).
extracellular signal-regulated kinase signaling 94. McNaught, J. On the action of cold or lukewarm tea 117. Lambert, J. D. et al. Peracetylation as a means of
pathways. Mol. Pharmacol. 72, 29–39 (2007). on Bacillus typhosus. J. R. Army Med. Corps 7, enhancing in vitro bioactivity and bioavailability of
72. Gradisar, H. et al. MD‑2 as the target of curcumin in 372–373 (1906). epigallocatechin‑3‑gallate. Drug Metab. Dispos.
the inhibition of response to LPS. J. Leukocyte Biol. 95. Steinmann, J. et al. Anti-infective properties of 34, 2111–2116 (2006).
82, 968–974 (2007). epigallocatechin‑3‑gallate (EGCG), a component of 118. Matsumoto, Y. et al. Antibacterial and antifungal
73. Tu, X.‑K. et al. Curcumin inhibits TLR2/4‑NF‑κB green tea. Br. J. Pharmacol. 168, 1059–1073 activities of new acylated derivatives of
signaling pathway and attenuates brain damage (2013). epigallocatechin gallate. Front. Microbiol. 3,
in permanent focal cerebral ischemia in rats. 96. Wolska, K. I., Grzes´, K. & Kurek, A. Synergy between 53 (2012).
Inflammation 37, 1544–1551 (2014). novel antimicrobials and conventional antibiotics 119. Hutchinson, M. R. et al. Evidence that opioids
74. Shuto, T. et al. Curcumin decreases toll-like receptor‑2 or bacteriocins. Pol. J. Microbiol. 61, 95–104 may have toll like receptor 4 and MD‑2 effects.
gene expression and function in human monocytes (2012). Brain Behav. Immun. 24, 83–95 (2010).
and neutrophils. Biochem. Biophys. Res. Commun. 97. Yam, T. S., Hamilton-Miller, J. M. & Shah S. 120. Hutchinson, M. R. et al. Opioid activation of toll-like
398, 647–652 (2010). The effect of a component of tea (Camellia sinensis) receptor 4 contributes to drug reinforcement.
75. Tu, C.‑T. et al. Curcumin attenuates concanavalin on methicillin resistance, PBP2ʹ synthesis, and J. Neurosci. 32, 11187–11200 (2012).
A‑induced liver injury in mice by inhibition of Toll-like β-lactamase production in Staphylococcus aureus. 121. Dawson, A. in Medical Toxicology 3rd edn
receptor (TLR) 2, TLR4 and TLR9 expression. J. Antimicrob. Chemother. 42, 211–216 (1998). (ed. Dart, R.) 228–230 (Lippincott, Williams and
Intnl Immunopharmacol. 12, 151–157 (2012). 98. Stapleton, P. D. et al. Modulation of β‑lactam Wilkins, 2004).
76. Chan, M. M. Inhibition of tumor necrosis factor by resistance in Staphylococcus aureus by catechins and 122. Clifton, L. A. et al. Effect of divalent cation removal
curcumin, a phytochemical. Biochem. Pharmacol. gallates. Int. J. Antimicrob. Agents 23, 462–467 on the structure of Gram-negative bacterial outer
49, 1551–1556 (1995). (2004). membrane models. Langmuir 31, 404 − 412 (2015).
77. Chainani‑Wu, N. Safety and anti-inflammatory 99. Zhao, W. et al. Mechanism of synergy between 123. Gill, E. E., Franco, O. L. & Hancock, R. E.
activity of curcumin: a component of tumeric epigallocatechin gallate and β-lactams against Antibiotic adjuvants: diverse strategies for controlling
(Curcuma longa). J. Altern. Compl. Med. 9, 161–168 methicillin-resistant Staphylococcus aureus. drug-resistant pathogens. Chem. Biol. Drug Des.
(2003). Antimicrob. Agents Chemother. 45, 1737–1742 85, 56–78 (2015).
78. Bengmark, S. Curcumin, an atoxic antioxidant and (2001). 124. Chauhan, A. et al. Full and broad-spectrum in vivo
natural NFκB, cyclooxygenase‑2, lipooxygenase, 100. Hu, Z.‑Q. et al. Epigallocatechin gallate synergy with eradication of catheter-associated biofilms using
and inducible nitric oxide synthase inhibitor: a shield ampicillin/sulbactam against 28 clinical isolates of gentamicin-EDTA antibiotic lock therapy. Antimicrob.
against acute and chronic diseases. J. Parenteral methicillin-resistant Staphylococcus aureus. Agents Chemother. 56, 6310–6318 (2012).
Enteral Nutr. 30, 45–51 (2006). J. Antimicrob. Chemother. 48, 361–364 (2001). 125. Deretic, V. Autophagy in immunity and cell-
79. Jain, S. K. et al. Curcumin supplementation lowers 101. Hu, Z.‑Q. et al. Epigallocatechin gallate synergistically autonomous defense against intracellular microbes.
TNF-α, IL‑6, IL‑8, and MCP‑1 secretion in high enhances the activity of carbapenems against Immunol. Rev. 240, 92–104 (2011).
glucose-treated cultured monocytes and blood levels methicillin-resistant Staphylococcus aureus. 126. Campoy, E. & Colombo, M. I. Autophagy in intracellular
of TNF-α, IL‑6, MCP‑1, glucose, and glycosylated Antimicrob. Agents Chemother. 46, 558–560 bacterial infection. Biochim. Biophys. Acta 1793,
hemoglobin in diabetic rats. Antioxid. Redox Signal. (2002). 1465–1477 (2009).
11, 241–249 (2009). 102. Navarro-Martinez, M. D. et al. Antifolate activity of 127. Nakagawa, I. et al. Autophagy defends cells against
80. Hansen, E. et al. A versatile high throughput epigallocatechin gallate against Stenotrophomonas invading group A Streptococcus. Science 306,
screening system for the simultaneous identification maltophilia. Antimicrob. Agents Chemother. 49, 1037–1040 (2004).
of anti-inflammatory and neuroprotective 2914–2920 (2005). 128. Birmingham, C. L. et al. Autophagy controls
compounds. J. Alzheimer’s Disease 19, 451–464 103. Lee, H. C. et al. Effect of tea phenolics and their salmonella infection in response to damage to the
(2010). aromatic fecal bacterial metabolites on intestinal salmonella-containing vacuole. J. Biol. Chem. 281,
81. Ryan, A. et al. A role for TLR4 in Clostridium difficile microbiota. Res. Microbiol. 157, 876–884 (2006). 11374–11383 (2006).
infection and the recognition of surface layer proteins. 104. Reygaert, W. & Jusufi, I. Green tea as an effective 129. Ogawa, M. et al. Escape of intracellular Shigella from
PLoS Pathog. 7, e1002076 (2011). antimicrobial for urinary tract infections caused by autophagy. Science 307, 727–731 (2005).
82. Pothoulakis, C. Effects of Clostridium difficile toxins Escherichia coli. Front. Microbiol. 4, 162 (2013). 130. Yano, T. et al. Autophagic control of Listeria through
on epithelial cell barrier. Ann. NY Acad. Sci. 915, 105. Li, W. et al. A major ingredient of green tea rescues intracellular innate immune recognition in drosophila.
347–356 (2000). mice from lethal sepsis partly by inhibiting HMGB1. Nat. Immunol. 9, 908–916 (2008).
83. Sintara, K. et al. Curcumin suppresses gastric NF‑κB PLoS ONE 2, e1153 (2007). 131. Gutierrez, M. G. et al. Autophagy is a defense
activation and macromolecular leakage in 106. Zhao, W.‑H. et al. Inhibition of penicillinase by mechanism inhibiting BCG and Mycobacterium
Helicobacter pylori-infected rats. World epigallocatechin gallate resulting in restoration of tuberculosis survival in infected macrophages.
J. Gastroenterol. 16, 4039– 4046 (2010). antibacterial activity of penicillin against penicillinase- Cell 119, 753–766 (2004).
84. Brennan, P. & O’Neill, L. A. Inhibition of nuclear factor producing Staphylococcus aureus. Antimicrob. Agents 132. Amano, A., Nakagawa, I. & Yoshimori, T. Autophagy
κB by direct modification in whole cells — mechanism Chemother. 46, 2266–2268 (2002). in innate immunity against intracellular bacteria.
of action of nordihydroguaiaritic acid, curcumin and 107. Stapleton, P. D. et al. The β-lactam-resistance modifier J. Biochem. 140, 161–166 (2006).
thiol modifiers. Biochem. Pharmacol. 55, 965–973 (–)-epicatechin gallate alters the architecture of the 133. Vergne, I. et al. Autophagy in immune defense against
(1998). cell wall of Staphylococcus aureus. Microbiology Mycobacterium tuberculosis. Autophagy 2, 175–178
85. Steiner, T. S. et al. Faecal lactoferrin, interleukin 1b, 153, 2093–2103 (2007). (2006).
and interleukin‑8 are elevated in patients with severe 108. Grandišar, H. et al. Green tea catechins inhibit 134. Mostowy, S. Autophagy and bacterial clearance:
Clostridium difficile colitis. Clin. Diagn. Lab. Immunol. bacterial DNA gyrase by interaction with its ATP a not so clear picture. Cell. Microbiol. 15, 395–402
4, 719–722 (1997). binding site. J. Med. Chem. 50, 264–271 (2007). (2013).

NATURE REVIEWS | DRUG DISCOVERY VOLUME 14 | DECEMBER 2015 | 831

© 2015 Macmillan Publishers Limited. All rights reserved


PERSPECTIVES

135. Kuballa, P. et al. Autophagy and the immune system. 144. Dutta, N. K. et al. The anti-inflammatory drug 151. Salahuddin, N. et al. Vitamin D accelerates clinical
Annu. Rev. Immunol. 30, 611–646 (2012). diclofenac retains anti-listerial activity in vivo. recovery from tuberculosis: results of the SUCCINCT
136. Levine, B., Mizushima, N. & Virgin, H. W. Lett. Appl. Microbiol. 47, 106–111 (2008). Study [Supplementary Cholecalciferol in recovery from
Autophagy in immunity and inflammation. 145. Dutta, N. K. et al. Potential management of tuberculosis]. A randomized, placebo-controlled clinical
Nature 469, 323–335 (2011). resistant microbial infections with a novel non- trial of vitamin D supplementation in patients with
137. Poels, J. et al. Expanding roles for AMP-activated antibiotic: the anti-inflammatory drug diclofenac pulmonary tuberculosis. BMC Infect. Dis. 13, 22 (2013).
protein kinase in neuronal survival and autophagy. sodium. Int. J. Antimicrob. Agents 30, 242–249 152. Anand, P. K. & Kaul, D. Vitamin D3‑dependent
Bioessays 31, 944–952 (2009). (2007). pathway regulates TACO gene transcription. Biochem.
138. Inoki, K. et al. TSC2 integrates Wnt and energy signals 146. Dutta, N. K. et al. Activity of diclofenac used Biophys. Res. Commun. 310, 876–877 (2003).
via a coordinated phosphorylation by AMPK and alone and in combination with streptomycin
GSK3 to regulate cell growth. Cell 126, 955–968 against Mycobacterium tuberculosis in mice. Acknowledgements
(2006). Int. J. Antimicrob. Agents 30, 336–340 The author thanks the following for expert discussions on the
139. Ulgherait, M. et al. AMPK modulates tissue and (2007). drugs reviewed: A. Coates (clinical antibiotic resistance and
organismal aging in a non-cell-autonomous manner. 147. Zhang, X. et al. Inhibitory effects of ivermectin ARB concept); S. Shaunak (clinical antibiotic resistance, TLRs
Cell Rep. 8, 1767–1780 (2014). on nitric oxide and prostaglandin E2 production and innate immune system); N. Ktistakis (autophagy);
140. Wang, W. H. et al. Aspirin inhibits the growth of in LPS-stimulated RAW 264.7 macrophages. D. Cavalla (drug repurposing); and members of the Science and
Helicobacter pylori and enhances its susceptibility to Int. Immunopharmacol. 9, 354–359 (2009). Technology Advisory Committee of Antibiotics Research UK.
antimicrobial agents. Gut 52, 490–495 (2003). 148. Bae, H.‑B. et al. AMP-activated protein kinase
141. Price, C. T. et al. The effects of salicylate on bacteria. enhances the phagocytic ability of macrophages Competing interests statement
Internat. J. Biochem. Cell Biol. 32, 1029–1043 (2000). and neutrophils. FASEB J. 25, 4358–4368 The author declares no competing interests.
142. Nicolau, D. P. et al. Influence of aspirin on (2011).
development and treatment of experimental 149. Singhal, A. et al. Metformin as adjunct FURTHER INFORMATION
Staphylococcus aureus endocarditis. Antimicrob. antituberculosis therapy. Sci. Transl. Med. 6, WHO — antimicrobial resistance: global report on
Agents Chemother. 39, 1748–1751 (1995). 263ra159 (2014). surveillance 2014: http://www.who.int/drugresistance/
143. Nicolau, D. P. et al. Reduction of bacterial titers by 150. Maeurer, M. & Zumla, A. The host battles documents/surveillancereport/en/
low-dose aspirin in experimental aortic valve drug-resistant tuberculosis. Sci. Transl. Med.
ALL LINKS ARE ACTIVE IN THE ONLINE PDF
endocarditis. Infect. Immun. 61, 1593–1595 (1993). 6, 263fs47 (2014).

832 | DECEMBER 2015 | VOLUME 14 www.nature.com/reviews/drugdisc

© 2015 Macmillan Publishers Limited. All rights reserved


ERRATUM

Antibiotic resistance breakers: can repurposed drugs fill the


antibiotic discovery void?
David Brown
Nature Reviews Drug Discovery 14, 821–832 (2015)
The article contained incorrect information regarding the susceptible bacteria and clinical trial phase of the
aztreonam–avibactam combination, and incorrect nomenclature for Salmonella enterica subsp. enterica serovar Typhimurium.
These errors have been corrected in the online version of the article.

© 2016 Macmillan Publishers Limited. All rights reserved

You might also like