Antibiotic Resistance and Persistence-Implications For Human Health and Treatment Perspectives
Antibiotic Resistance and Persistence-Implications For Human Health and Treatment Perspectives
Antibiotic Resistance and Persistence-Implications For Human Health and Treatment Perspectives
See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Review
Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
*Corresponding author. Tel. +41 44 255 25 41; E-mail: [email protected]
Glossary
AACs antibody–antibiotic conjugates MDK minimal duration of killing
ABDs antibacterial drones MDR multidrug resistant
AmpC b-lactamase MIC minimum inhibitory concentration
AMPs antimicrobial peptides MLST multilocus sequence type
AMR antimicrobial resistance MRSA methicillin-resistant S. aureus
ATP adenosine triphosphate NDM-1 New Delhi metallo-b-lactamase 1
blaZ b-lactamase nsSCs non-stable small colonies
C10 3-[4-(4-methoxyphenyl)piperazin-1-yl] piperidin-4-yl ParC DNA topoisomerase IV
biphenyl-4-carboxylate PBP2a penicillin binding protein 2a
CA community-acquired PJIs prosthetic joint infections
CCCP carbonyl cyanide m-chlorophenyl hydrazine pmf proton motive force
CDI C. difficile infections QRDR quinolone resistance determining region
CF cystic fibrosis REPTIS Replica Plating Tolerance Isolation System
CFUs colony-forming units RNA ribonucleic acid
CIP ciprofloxacin RND resistance nodulation division
CRISPR Clustered Regularly Interspaced Short Palindromic Repeats ROS reactive oxygen species
DNA deoxyribonucleic acid RpoB RNA polymerase beta-subunit
dTMP deoxythymidine monophosphate SaPIs staphylococcal pathogenicity islands
dUMP deoxyuridine monophosphate SCCmec staphylococcal cassette chromosome mec
ECDC European Centre of Disease Prevention and Control SCVs small colony variants
EMBO European Molecular Biology Organization TA toxin/ antitoxin
ESBLs extended-spectrum b-lactamases TCA tricarboxylic acid
ESKAPE E. faecium, S. aureus, K. pneumoniae, A. baumannii, P. TCS two-component systems
aeruginosa and Enterobacter species TD Tolerance Disk
EU European Union TMP-SMX trimethoprim-sulfamethoxazole
fabI/V enoyl-ACP reductase gene UN United Nations
GltX aminoacyl-tRNA synthetase UPEC uropathogenic E. coli
GyrA DNA gyrase USA United States of America
hip high persister UTIs urinary tract infections
HR heteroresistance UV ultraviolet
IMP imipenem VRE vancomycin-resistant Enterococci
KPC K. pneumoniae carbapenamases VRSA vancomycin-resistant S. aureus
MBC minimal bactericidal concentration WHO World Health Organization
mcr mobilized colistin resistance
inherited ability of microorganisms to grow at high antibiotic naturally susceptible to almost every antibiotic that has been devel-
concentrations (Brauner et al, 2016). It is usually quantified by oped, but it is well known for quickly acquiring antibiotic resistance
measuring the minimum inhibitory concentration (MIC) of a partic- by means of obtaining specific genetic modifications (mutations or
ular antibiotic wherein resistant bacteria are able to multiply and horizontal gene transfer), causing infections that come in epidemic
grow at concentrations of antibiotics, which are fatal to other strains waves (Chambers & Deleo, 2009).
of the same species. Antibiotic resistance comes in distinct flavours. Several mechanisms can lead to resistance and have been inves-
On the one hand, there is natural intrinsic resistance due to lack or tigated in detail. These molecular mechanisms are categorized in
presence of certain structures resulting in ineffectiveness of antibi- three main mechanistic groups of resistance: (i) reduction of intra-
otics. On the other hand, bacteria can acquire resistance via muta- cellular antibiotic concentrations, (ii) modification of the antibiotic
tions in chromosomal genes or via horizontal gene transfer of target and (iii) inactivation of the antibiotic (Blair et al, 2015).
chromosomes or plasmids that lead to antibiotic resistance. Intracellular concentrations of antibiotics can be kept low via
In Pseudomonas aeruginosa for example, antimicrobial decreased permeability of the bacterial cell membrane, e.g. by
substances such as triclosan, which inhibit fatty acid synthesis by reducing or mutating porins that would allow entry of antibiotics
targeting the enoyl-ACP reductase, are ineffective, as the bacteria into the cell. This has been well described for several Gram-negative
carry an insensitive homologue of fabI (enoyl-ACP reductase), fabV, pathogens such as carbapenem-resistant Enterobacter species,
that encodes an enzyme that is not inhibited by triclosan (Zhu et al, Escherichia coli and Klebsiella pneumoniae that display reduced
2010). In general, Gram-negative bacteria are less permeable than porin expression or mutated porins without expressing a carbapene-
Gram-positive and show an intrinsic resistance to many antibacte- mase (Wozniak et al, 2012; Baroud et al, 2013; Kong et al, 2018).
rial compounds such as the cell wall active glycopeptide vanco- Another way to decrease intracellular antibiotic concentrations is to
mycin (Zgurskaya et al, 2015). These large molecules are unable to increase the efflux of chemotherapeutics by using either substrate
cross the outer bacterial membrane of Gram-negative bacteria and specific or MDR efflux pumps. Although many bacteria carry multi-
thus cannot target the cell wall of Gram-negative bacteria. In ple genes that encode MDR efflux pumps on their chromosomes,
contrast, the Gram-positive pathobiont Staphylococcus aureus is some MDR efflux pump genes have been mobilized onto plasmids
9
15,000 4,000 10,000 8
2012 2014 2016 2018 2014 2016 2018 2012 2014 2016 2018
© EMBO
Figure 1. Invasive bacterial isolates and resistance development over time in the European Union (EU) and European Economic Area (EEA).
(A) Total number of invasive E. coli isolates tested and percentage with combined resistance to fluoroquinolones, 3rd-generation cephalosporins and aminoglycosides,
including 95% confidence intervals (95% CI, shaded area). (B) Total number of invasive E. faecium isolates tested and percentage with resistance to vancomycin,
including 95% CI. (C) Total number of invasive S. aureus isolates tested and percentage with resistance to methicillin (MRSA), including 95% CI. (D) Total number of
invasive K. pneumoniae isolates tested and percentage with combined resistance to fluoroquinolones, 3rd-generation cephalosporins and aminoglycosides, including 95%
CI. (E) Total number of invasive Acinetobacter spp., including most of the disease-causing species (A. baumannii, A. pittii and A. nosocomialis) and the generally less
pathogenic A. non-baumannii group, isolates tested and percentage with combined resistance to fluoroquinolones, aminoglycosides and carbapenems, including 95% CI.
(F) Total number of invasive P. aeruginosa isolates tested and percentage with combined resistance (resistance to three or more antimicrobial groups among piperacillin
—tazobactam, ceftazidime, fluoroquinolones, aminoglycosides and carbapenems), including 95% CI. (A-F) Data are derived from European Centre for Disease Prevention
and Control’s yearly Surveillance of antimicrobial resistance in Europe (reports 2013-2018 were used). ECDC collects data from invasive isolated reported to the
European Antimicrobial Resistance Surveillance Network (EARS-Net) by 30 EU and EEA (Iceland and Norway) countries.
that can be transferred between bacteria (Lv et al, 2020). Genes gyrase GyrA or DNA topoisomerase IV ParC can cause ciprofloxacin
coding for a novel tripartite resistance nodulation division (RND) resistance in S. aureus (Sreedharan et al, 1990; Hooper & Jacoby,
pump were found to be carried on a plasmid together with genes 2015). Therefore, this domain has been named the quinolone resis-
encoding for the antibiotic-targeting enzyme New Delhi metallo-b- tance determining region (QRDR) (Yoshida et al, 1990). Although
lactamase 1 (NDM-1) (Dolejska et al, 2013; Lv et al, 2020). This is most of clinical methicillin-resistant S. aureus (MRSA) isolates are
especially worrying as it shows that different resistance mechanisms susceptible to rifampicin (Moran et al, 2006), resistance develop-
can be carried together on one plasmid and are thereby transmissi- ment occurs rapidly if rifampicin is used as monotherapy. Point
ble between bacteria, highlighting the importance of MDR efflux mutations in the gene encoding for the b-subunit of the DNA-depen-
pumps as a major resistance mechanism. Many clinical isolates that dent RNA polymerase (RpoB) cause decreased affinity for rifampicin
overexpress drug efflux pumps, including P. aeruginosa and and quickly confer resistance (Maranan et al, 1997).
S. aureus, have been isolated from patients with systemic infections Finally, resistance can be acquired by directly modifying antibi-
(Pumbwe & Piddock, 2000; Kosmidis et al, 2012). otics. Antibiotics can be inactivated by the transfer of chemical
Spontaneous mutations or post-translational modifications of an groups to vulnerable sites thus inhibiting target binding and func-
antibiotic target molecule can lead to conformational changes that tion or directly destroyed by hydrolysis. Inactivation of antibiotics
result in ineffective target binding and attenuation of antibiotic by addition of a chemical group, such as acyl, nucleotidyl,
activity. Single amino acid changes in close proximity to the active phosphate and ribitoyl groups, resulting in steric hindrance can
site tyrosine of the bacterial type II topoisomerase enzymes, DNA cause antibiotic resistance. Especially, the large molecules of
aminoglycosides with many exposed hydroxyl and amide groups 1940s, the prognosis of patients rapidly improved. Penicillin became
are susceptible to such a inactivation mechanism. A genomic island the first choice of treatment for S. aureus infections, but extensive
found in Campylobacter coli isolated from chicken in China harbours use quickly led to the evolution of b-lactamases (blaZ)-producing
genes for six aminoglycoside-modifying enzymes and confers resis- strains. Already in 1948, more than 80% of clinical isolates were
tance to aminoglycosides, including gentamicin (Qin et al, 2012). resistant to penicillin G (Appelbaum, 2007; Brown & Ngeno, 2007;
The development of new derivatives of already known antibiotic Wu et al, 2010). b-lactamase, a primarily extracellular enzyme,
classes led to the evolution of a diverse range of enzymes that can hydrolyses the b-lactam ring, rendering the b-lactam inactive
degrade different antibiotics within the same class. For example, the (Lacey, 1984). Already one year after the introduction of semi-
early b-lactamases were active against the 1st-generation b-lactams synthetic penicillins in 1961, MRSA strains were isolated and there
and were followed by extended-spectrum b-lactamases (ESBLs) that is even evidence that S. aureus developed methicillin resistance
are able to hydrolyse extended-spectrum oxyimino cephalosporins even before its clinical use (Harkins et al, 2017; Turner et al, 2019).
such as cefuroxime, cefotaxime, cefmenoxime and ceftriaxone The widespread use of first-generation b-lactams, such as penicillin,
(Johnson & Woodford, 2013). Together with the increasing numbers prior to the introduction of methicillin, selected for strains carrying
of bacteria carrying ESBL genes, the use of carbapenems the mecA determinant that confers resistance to methicillin (Harkins
has increased in the last two decades leading to the emergence of et al, 2017). Nowadays, MRSA prevalence ranges from < 1 to more
carbapenemase-producing strains (Queenan & Bush, 2007). than 40% in Europe, with very low levels of invasive MRSA isolates
Carbapenemases can degrade a variety of b-lactams, including in Scandinavian countries such as Iceland (0.0%) and Norway
extended-spectrum cephalosporins, and many carbapenemase-genes (0.9%), low levels, e.g. in the Netherlands (1.2%), in Switzerland
are now carried on plasmids and have been found in Enterobacteri- (4.4%), in Germany (7.6%) and in France (12.1%) and up to 34%
aceae, K. pneumoniae, P. aeruginosa and Acinetobacter baumannii in Italy, 38% in Portugal and 43% in Romania (Hassoun et al, 2017;
(Tzouvelekis et al, 2012) (Fig 1A). FOPH, 2018; ECDC, 2019) (Fig 1C). Methicillin resistance is medi-
ated by the gene mecA that is spread via horizontal gene transfer of
ESKAPE pathogens the mobile genetic element named staphylococcal cassette chromo-
The acronym ESKAPE describes nosocomial pathogenic bacteria some mec (SCCmec) (Katayama et al, 2000). The mecA gene
with growing levels of multidrug resistance and virulence. They encodes for a penicillin binding protein (PBP2a) that is responsible
place a significant burden on patient health, healthcare systems and for crosslinking peptidoglycans during cell wall synthesis and the
economies. ESKAPE stands for the Gram-positive bacterial species resistance arises through the low affinity of PBP2a to b-lactams
Enterococcus faecium and S. aureus and the Gram-negative bacteria (Hartman & Tomasz, 1984) rendering them ineffective. One
K. pneumoniae, A. baumannii, P. aeruginosa and Enterobacter prominent pandemic and hypervirulent clonal lineage of commu-
species. They all commonly cause hospital-acquired and potentially nity-acquired (CA)-MRSA is the multilocus sequence type (MLST) 8-
life-threatening infections in critically ill and immunocompromised MRSA-IV USA300 that started spreading in the USA 20 years ago
patients and are characterized by high levels of multidrug resistance and is a significant health threat and economic factor nowadays
(Rice, 2010). The WHO listed all ESKAPE pathogens among the (Tenover & Goering, 2009).
twelve bacterial species against which new chemotherapeutics are Colonization with MRSA increases the MRSA infection risk, as
desperately needed (Tacconelli et al, 2018). 50–80% of isolated invasive strains were found to originate from
colonizing strains as determined by molecular typing methods
E. faecium including whole genome sequencing (Benoit et al, 2018; Thomsen
The Gram-positive commensal E. faecium has a low virulence but is et al, 2019). It is a dynamic process, with strains persisting for
difficult to eradicate from the hospital environment. Therefore and long periods of time and others evolving or being replaced by
because of its potential to cause nosocomial infections and another strain within the same host (Azarian et al, 2016). In
outbreaks, it is strongly feared in the healthcare system (Elsner addition, transmission of S. aureus commonly occurs via hospital
et al, 2000). In the last decades, the incidence of drug-resistant ente- coats, mobile phones and tablets, regularly used in clinics (Frey
rococcal infections increased, especially for vancomycin-resistant et al, 2019; Turner et al, 2019). In clinics, glycopeptide antibiotics
Enterococci (VRE) reaching 14.9% in 2017 in the European Union such as vancomycin and teicoplanin, are used nowadays as first-
(EU) (ECDC, 2018b) (Fig 1B) and 30% in the United States of Amer- line treatment for MRSA infections. Shortly after the initiation of
ica (USA) (CDC, 2019). Treatment of these nosocomial VRE vancomycin therapy in the 1980s, MRSA strains with reduced
infections is very difficult and sometimes impossible due to the sensitivity to teicoplanin appeared in Europe (Kaatz et al, 1990).
resistance. In 2002, the first vancomycin-resistant S. aureus (VRSA) was
detected in the USA (Chang et al, 2003) and eleven years later in
S. aureus Europe (Melo-Cristino et al, 2013). Effective alternatives to vanco-
The Gram-positive bacterium S. aureus is part of the normal skin mycin include daptomycin, a lipopeptide bactericidal antibiotic,
microbiota, found especially in the nose. Carriage rates are high, linezolid, tigecycline, trimethoprim–sulfamethoxazole (TMP-SMX)
with around 50% of the population intermittently or permanently or a 5th-generation b-lactam (e.g. ceftaroline or ceftobiprole).
colonized (Lowy, 1998). Nevertheless, S. aureus is a leading cause Similarly, if a reduced daptomycin susceptibility is detected
of infective endocarditis, osteomyelitis, skin and soft tissue infec- together with a reduced vancomycin susceptibility, a combination
tions as well as bacteraemia. Before the antibiotic era, mortality or single use of the following is recommended; the streptogramin
rates for S. aureus bacteraemia were dramatically high (over 80%) antibiotics quinupristin–dalfopristin, TMP-SMX, linezolid or
(Skinner & Keefer, 1941). With the introduction of penicillin in the telavancin (Liu et al, 2011).
the phenomenon of susceptible bacteria surviving antibiotic chal- the production of the stringent response alarmone (p)ppGpp that
lenges. In the last 30 years, many mechanisms and characteristics inhibits DNA replication, and therefore, blockage of translation can
underlying bacterial persistence have been revealed. However, indirectly result in additional tolerance towards fluoroquinolones
research mainly focused on Gram-negative bacteria, e.g. E. coli, (Schreiber et al, 1995; Korch & Hill, 2006; Kaspy et al, 2013;
P. aeruginosa and S. enterica ser. Typhimurium, and little is known Wilmaerts et al, 2019). Next to hipA, hipB encodes an auto-
about the mechanisms and influence of bacterial persistence in repressor of hipBA transcription (Black et al, 1991; Black et al,
Gram-positive bacteria especially S. aureus and coagulase-negative 1994). HipB directly inhibits HipA, which suggested that hipBA
staphylococci (S. epidermidis), which are often associated with constitutes a toxin/ antitoxin (TA) locus (Korch et al, 2003). Today,
chronic and recurrent, biofilm-associated infections. The group of numerous publications support the model of TA loci cumulatively
Kim Lewis was the first to link bacterial persistence to biofilms in regulating antibiotic persistence in E. coli (Vazquez-Laslop et al,
P. aeruginosa (Brooun et al, 2000; Spoering & Lewis, 2001). Chronic 2006; Dorr et al, 2010; Page & Peti, 2016).
infections are associated with bacterial biofilms, and the newly
linked phenomenon of antibiotic persistence added a novel point of
view to recurring and difficult-to-treat infections and attracted the Toxin/ Antitoxin modules and persistence
interest of more and more scientists worldwide (Bjarnsholt, 2013;
Lebeaux et al, 2014). In 2018 at the European Molecular Biology TA modules are composed of a stable toxin (always a protein) that
Organization (EMBO) Workshop “Bacterial Persistence and Antimi- intoxicates the bacterial cell by inhibiting essential processes, e.g.
crobial Therapy”, 121 scientists studying antibiotic persistence and translation, transcription and DNA replication, and a labile antitoxin
tolerance came together in Ascona, Switzerland, and laid the foun- (either RNA or protein) that neutralizes the corresponding toxin
dations for a “Consensus Statement” on the definitions and guideli- (Harms et al, 2018). Up to now, there are six described classes of
nes for research on antibiotic tolerance (Balaban et al, 2019a, b). TA modules, defined by their neutralization mechanisms. Type I
Here, we follow the definitions published in the consensus state- antitoxins are antisense RNAs that inhibit the translation of the
ment (Balaban et al, 2019a). mRNA of their corresponding toxins, therefore inhibiting toxin
Antibiotic persistence or heterotolerance is defined as the ability production (Thisted et al, 1994). In the largest and best studied
of a bacterial subpopulation to survive high bactericidal drug group of TA modules, the type II TA systems, antitoxins are proteins
concentrations to which the bacteria are fully susceptible (no that directly bind their cognate toxins thereby neutralizing the toxin
change in MIC). One major characteristic of antibiotic persistence is (Pandey & Gerdes, 2005). Additionally, the antitoxin can be a toxin-
a biphasic killing, with a rapid killing of the susceptible population binding sRNA (type III) (Brantl & Jahn, 2015), which protects the
and a slower killing of the persister subpopulation. This phenom- toxin targets via binding to and directly inhibiting the toxin, instead
enon highlights the phenotypic heterogeneity in a bacterial culture of counteracting the toxins’ effect (type IV) (Masuda et al, 2012).
and that not all bacteria in a clonal population are killed at the same The antitoxin can also be an endoribonuclease that degrades the
rate. In contrast to antibiotic resistance and heteroresistance, where toxins’ mRNA (type V) (Wang et al, 2012) or a proteolytic adapter
a small subpopulation transiently displays a higher MIC, persistent of the toxin (type VI) (Aakre et al, 2013). Often, TA modules are
bacteria are not able to grow in the presence of antibiotics. Antibi- described as stress-responsive elements that are activated under
otic persistence and tolerance are often used interchangeably, and specific stress conditions (Ronneau & Helaine, 2019). As described
both describe an increased survival in the presence of an antibiotic before, the type II TA system toxin HipA7 can induce antibiotic
without a change in the MIC. However, persistence characterizes a persistence by phosphorylation of its target GltX in E. coli (Moyed &
bacterial subpopulation, whereas tolerance describes the ability of Bertrand, 1983). Therefore, TA modules were proposed to be perfect
an entire bacterial population to survive antibiotic exposure. Thus, candidates to trigger antibiotic persistence.
antibiotic persistence is a subcategory of tolerance, in which a small
subpopulation of persisters tolerates antibiotic exposure, reflected
by a biphasic killing curve. Environmental triggers of antibiotic persistence
PMF
Lex box containing Inhibition of
cellular functions T/A systems
ATP T/A systems
ATP tisAB/ istr1 • Transcription
symER • Translation
ATP hokE / sokE • DNA replication
yafN/ yafO •
… • Energy production
Drop in Accumulation of
ATP levels insoluble proteins
Accumulation of (p)ppGpp has been shown to upregulate or activate tisAB/istr1, symER, hokE/sokE and yafN/yafO (Pedersen & Gerdes,
TA systems during stringent response. For example, isoleucine star- 1999; Fernandez De Henestrosa et al, 2000; Vogel et al, 2004;
vation of E. coli increases (p)ppGpp levels and leads to an upregula- Kawano et al, 2007). The type I toxin TisB, a small 29 amino acid
tion of many type II TA modules, like MazEF, HicAB, RelBE and hydrophobic peptide, for example, binds to the bacterial cell
MqsRA as well as the type I toxin HokB (Fig. 2) (Traxler et al, 2008; membrane and disrupts the proton motive force (pmf), leading to a
Verstraeten et al, 2015; Shan et al, 2017; LeRoux et al, 2020). Muta- decrease in intracellular adenosine triphosphate (ATP) levels and a
tions that affect (p)ppGpp synthesis significantly reduce persister multidrug tolerant phenotype (Fig. 2) (Unoson & Wagner, 2008;
levels in both Gram-positive and Gram-negative bacteria, indicating Lewis, 2012). A drop in ATP levels has been associated with antibi-
the prominent role and connections of stringent response with otic tolerance in E. coli (Shan et al, 2017) and S. aureus (Conlon
bacterial stress response and antibiotic persistence (Amato et al, et al, 2016). In both studies, treatment of exponentially growing
2013; Amato et al, 2014). bacteria with arsenate, a drug known to inhibit ATP synthesis,
DNA damage is caused by many factors including oxidative resulted in elevated levels of antibiotic tolerance. Similarly, pretreat-
stress, UV light exposure and drugs and usually leads to the induc- ment with the metabolic poison carbonyl cyanide m-chlorophenyl
tion of the SOS response (Dorr et al, 2010; Baharoglu & Mazel, hydrazine (CCCP), an uncoupling agent that inhibits ATP synthesis,
2014). The SOS response not only activates DNA repair mechanism, resulted in increased persister levels (Kwan et al, 2013). Congruently
but also upregulates the Lex box containing TA systems such as with stringent response-mediated persistence, inhibition of
translation by pretreatment with the antibiotic tetracycline yielded 2013), recognition by immune cells (Leid et al, 2002; Jesaitis et al,
an increased fraction of persisters (Kwan et al, 2013). Recently, 2003; Vuong et al, 2004), killing by many antibiotics (Jolivet-
Conlon and co-workers showed that inactivation of the tricarboxylic Gougeon & Bonnaure-Mallet, 2014) and contains persister cells (Spo-
acid (TCA) cycle enzymes aconitase and succinate dehydrogenase ering & Lewis, 2001; Kamble Ekta, 2020). The increasing age of the
by reactive oxygen species (ROS) produced by human macrophages human population results in more tissue-related biofilm-associated
during infection, decreased ATP levels and increased persister levels infections such as osteomyelitis and endocarditis, but also in more
in S. aureus, further strengthening the link between reduced energy foreign body-associated biofilm infections due to the constantly
levels and antibiotic tolerance (Rowe et al, 2020). rising numbers of pacemaker and orthopaedic device implants.
Depletion or reduction of intracellular ATP levels could therefore Biofilm formation has been linked to persistent infections, especially
be a general mechanism of downregulating central bacterial to difficult-to-treat foreign body infections such as pacemaker-related
processes, such as transcription, translation and peptidoglycan infections and prosthetic joint infections (PJIs) (Jansen & Peters,
synthesis, thereby rendering antibiotics ineffective as their respec- 1993; Costerton et al, 1999; Dengler Haunreiter et al, 2019).
tive targets are not active (Fig. 2). PJIs are often caused by S. aureus or coagulase-negative staphy-
In addition, accumulation of insoluble endogenous proteins in lococci (e.g. S. epidermidis) and require prolonged antibiotic treat-
deep-stationary phase, starved E. coli, promoted by decreased levels ment and often surgical removal of the infected device (Tande &
of ATP, has been linked to dormancy depth and antibiotic tolerance Patel, 2014). The majority of PJIs occur in the first year after implan-
(Fig. 2) (Pu et al, 2019). Pu and co-workers showed that the tation and are typically caused by commensal microorganisms that
dormancy depth that correlates with antibiotic tolerance is linked to were inoculated during surgery (Del Pozo & Patel, 2009). Either
the formation of protein aggresomes. Resuscitation of these dormant physical contact or droplets lead to the contamination of the pros-
cells leads to degradation of protein aggregates and requires DnaK thesis surface and even very low inocula (< 102 CFUs for S. aureus)
and ClpB activity. The chaperone DnaK was previously identified to result in infection (Southwood et al, 1985). PJIs manifesting later
be involved in bacterial antibiotic persistence by analysing an E. coli than 2 years after surgery are typically caused by haematogenous
mutant library further strengthening the involvement of protein spread during bacteraemia. A study including 14,378 patients with a
aggregation in antibiotic persistence (Hansen et al, 2008). primary knee or hip replacement showed that 3.8% (542/14,378)
had at least one significant bacteremic episode and 8.3% (45/542)
developed a PJI during bacteraemia. The most common cause of PJI
Chronic and difficult-to-treat infections was S. aureus, with 21% PJIs resulting from bacteraemia (Honka-
nen et al, 2019). Many studies investigating PJI caused by S. aureus
There is a fundamental difference between the terms “persistent and other staphylococcal species found small colony variants
infection” and “antibiotic persistence”. Persistent infections are (SCVs) (Kahl et al, 2016). Proctor et al were the first to describe the
ongoing infections, which are not cleared by the host, whereas occurrence of SCVs in a patient suffering from MRSA bacteraemia
antibiotic persistence is used to describe a bacterial population that with a hip prosthesis infection (Proctor et al, 1995). However, SCVs
survives antibiotic exposure without being resistant (Balaban et al, are not only found in patients suffering from PJI, but there are many
2019a). Nevertheless, antibiotic persistence is thought to be studies reporting SCVs in other persistent biofilm-associated
involved in persistent infections, potentially impeding successful infections. Among these difficult-to-treat infections are pacemaker-
treatment (Fauvart et al, 2011). Persistent infections are also known related infections (Baddour et al, 1990; von Eiff et al, 1999),
as chronic infections and are often associated with antibiotic treat- endocarditis (Quie, 1969; Bhattacharyya et al, 2012), osteomyelitis
ment failure (Rhen et al, 2003; Fisher et al, 2017). These infections (Proctor et al, 1995), airway infections (Kahl et al, 1998; Besier
are not sufficiently cleared by the hosts’ immune system. Many dif- et al, 2007; Schneider et al, 2008) and skin and soft tissue infections
ferent factors, such as cell wall modification, capsule production, (Abele-Horn et al, 2000; Vulin et al, 2018).
antigenic mimicry and inhibition of effector proteins, contribute to
bacterial immune evasion (Hornef et al, 2002). Usually, the bacterial
cell wall is the first target of the hosts’ immune system and the first Colony size heterogeneity in persistent and difficult-
line of defence of the bacteria. For example, many Gram-negative to-treat infections
pathogens such as E. coli and S. enterica acetylate lipid A molecules
in their cell walls, which switches their negative surface charge to Even before the introduction of antibiotics and the first description
positive, repelling positively charged antimicrobial peptides (AMPs) of bacterial persistence, SCVs were reported for S. aureus and
from the host (Li et al, 2012). Others, like Borrelia spp. and Neisse- several other coagulase-negative staphylococci (Proctor et al, 2006).
ria gonorrhoea, vary their surface antigens during infection and thus Similar to bacterial persisters, SCVs have been associated with
evade the hosts’ humoral immune response (Nataro et al, 2000; recurrent and chronic infections (Proctor et al, 2006) and have been
Norris, 2006). Some pathogens such as Listeria monocytogenes and isolated from many different clinical specimens, e.g. abscesses and
Mycobacterium tuberculosis can trigger an anti-inflammatory soft tissue, bones and joints, the respiratory tract and
response by inducing the production of interleukin-10 that blood (Swingle, 1935; Goudie & Goudie, 1955; von Eiff et al, 1997a;
suppresses interferon-c production by macrophages, which in Kahl et al, 1998; Seifert et al, 1999; von Eiff et al, 1999; Rolauffs
general would inhibit bacterial growth (Redpath et al, 2001). et al, 2002).
Another way in which bacteria evade host recognition is by form- SCVs are characterized by their small colony size, reduced viru-
ing biofilms. Secreted polysaccharides and extracellular DNA form a lence and slow growth. Most studies were done on genetically deter-
protective matrix that blocks complement-killing (Domenech et al, mined stable SCVs that display a small colony size because of
genetic mutations (e.g. in hemB (von Eiff et al, 1997b), hemH et al, 2019a; Balaban et al, 2004; Van den Bergh et al, 2017). Many
(Schaaff et al, 2003), menD (Bates et al, 2003) or ctaA (Clements factors have been reported to trigger antibiotic persistence in bacte-
et al, 1999)), which either lead to an altered electron transport or ria, including nutrient limitation (Gutierrez et al, 2017), high cell
defect thymidine metabolism (Proctor et al, 2006; Proctor, 2019) density (Vega et al, 2012), oxidative stress (Wu et al, 2012; Rowe
(Fig. 3A). Defective hemin or menadione synthesis causes a reduced et al, 2020), an acidic environment (Lewis, 2007; Vulin et al, 2018),
electron transport and therefore decreased ATP levels. Low energy immune factors (Manina et al, 2015) and exposure to immune cells
levels and reduced membrane potential lead to a slower growth rate (Helaine et al, 2014).
of the bacteria and finally to smaller colonies. This defect can be Phenotypic heterogeneity is a complex quality that cannot be
overcome by supplementing with hemin or menadione leading to detected when analysing a single bacterial cell at a certain time
normal growing colonies. The second group of stable SCVs caused point. It needs the comparison to multiple other bacterial cells in a
by a thymidine biosynthesis defect has been isolated mainly from population or at least multiple observations of a single bacterium
cystic fibrosis (CF) patients that received a TMP-SMX treatment (Gil- over time (Ackermann, 2015). In case of bacterial growth analysis
ligan et al, 1987; Kahl et al, 1998). Kahl and co-workers showed that and determination of lag phase, Balaban and co-workers developed
a thymidine-auxotrophic clinical S. aureus could be complemented a method for E. coli to monitor the growth of many bacterial colo-
with thyA that encodes for a thymidylate synthase, enabling synthe- nies and lag times in parallel, the so-called ScanLag method (Levin-
sis of dTMP from dUMP (Chatterjee et al, 2008; Kriegeskorte et al, Reisman et al, 2010; Levin-Reisman et al, 2014). ScanLag generates
2014). a two-dimensional distribution of the lag time and of the growth
However, most small colonies recovered in vitro, directly from time of colonies on agar plates. We recently modified the ScanLag
patient material in clinical microbiology laboratories or from murine setup for the analysis of S. aureus lag time distributions after direct
infection models revert to a normal colony size upon subcultivation sampling from infection sites and in vitro stress exposure, named
(Tuchscherr et al, 2011; Vulin et al, 2018). Consequently, these ColTapp (Vulin et al, 2018; B€ ar et al, 2020). Automated agar plate
small colonies are named non-stable small colonies (nsSCs) imaging as well as single-cell microscopy revealed that the small
(Fig 3A). Usually, these small colonies occur at a very low colonies are the result of a prolonged bacterial lag phase and can be
frequency in a bacterial population, but increase after stress expo- linked to an increased antibiotic persistence in S. aureus (Fig 3B).
sure, e.g. acidic pH (Leimer et al, 2016; Vulin et al, 2018), exposure Therefore, these small colonies reflect a subpopulation of persis-
to an intracellular milieu in the host (Coffey & De Duve, 1968; Vesga ters and might serve as a read out for antibiotic persistence in clini-
et al, 1996; Tranchemontagne et al, 2016) or reactive oxygen species cal laboratories.
(Painter et al, 2015; Loss et al, 2019). NsSCs share many characteris-
tics with bacterial persisters: (i) they both occur at a low frequency
in a bacterial population; (ii) they display a revertible phenotype; Antimicrobial heteroresistance
and (iii) they show an increased tolerance towards antibiotics. A
clear and meaningful definition of these small colonies is still miss- Another bacterial bet hedging strategy to survive antibiotic treat-
ing. Usually, nsSCs are defined as colonies with an area 5 or 10 ment is heteroresistance (HR). In contrast to persistence, HR
times smaller than the area of the most common colony type (Proc- describes the ability of a subpopulation of susceptible bacteria to
tor et al, 2006). Instead of using a binary classification in “small” grow despite antibiotic pressure. The exact mechanisms underlying
and “normal” colonies, more and more focus is put on phenotypic HR are still unclear, but there is evidence that unstable amplification
heterogeneity and colony size distributions that give a detailed over- of antibiotic resistance genes is one way bacteria become heterore-
view on all recovered colony phenotypes. sistant (Hjort et al, 2016; Anderson et al, 2018; Nicoloff et al, 2019).
Phenotypic heterogeneity can be beneficial for the survival of a In order to survive antibiotic challenges and to grow even in pres-
bacterial population (Ackermann, 2015). Bacterial bet hedging via ence of antibiotics, the heteroresistant phenotype must be trans-
the formation of a subpopulation of persister cells in a clonal popu- ferred to daughter cells to ensure growth. Antibiotic resistance gene
lation of bacteria might be a great advantage in clinical infection amplification as the reason for HR can explain how this is achieved,
settings. While the actively growing part of a clonal bacterial popu- at least for genetic HR. It is still not clear though how phenotypic
lation is mostly responsible for host colonization and the successful HR that is not based on any genetic traits, is propagated in a popula-
establishment of an infection, the persister cell subpopulation tion under antibiotic treatment. One possible explanation for the
ensures the survival of the genotype under changing environments, maintenance of the heteroresistant phenotype by bacteria could be
e.g. under antibiotic therapy (Claudi et al, 2014). Nevertheless, epigenetic inheritance. There are several studies describing epige-
formation of these persisters is costly as they do not contribute to netic trait propagation (inheritance) of phenotypic variations over
the growth of the bacterial population but serve as a back-up plan generations (Veening et al, 2008; Ni et al, 2012; Ram & Goulian,
in case of emergencies. Persister cells form in two ways, either 2013). This heteroresistant phenotype is not maintained indefinitely
spontaneously or triggered by environmental factors (Balaban et al, but is lost as soon as the antibiotic pressure is removed, resulting in
2019a). The spontaneous formation of a small group of dormant a population consisting of antibiotic-susceptible bacteria and a small
bacterial cells in an overall growing population was previously fraction of heteroresistant bacteria creating the initial phenotypic
described as type II-persistence (Balaban et al, 2004). This sponta- heterogeneity.
neous persistence leads to a constant fraction of persisters in a The clinical relevance of heteroresistance and effect on treatment
population (usually between 0.001% and 1%), ensuring bacterial outcome is still under debate. Vancomycin HR in S. aureus has been
survival in the presence of antibiotics, but seems to be less common suggested to cause vancomycin treatment failure (Hiramatsu et al,
than triggered persistence (previously type I-persistence) (Balaban 1997; Moore et al, 2003), but there is also evidence that vancomycin
treatment is still effective for treating vancomycin heteroresistant vancomycin and may be another reason for vancomycin treatment
strains, depending on the antibiotic concentrations used (Khatib failure. Another study describes colistin HR in Enterobacter cloacae,
et al, 2011). Vancomycin heteroresistance might not be the only where a subpopulation (1–10% of the entire population) displayed
explanation for vancomycin treatment failure. In addition, intracel- 1,000-fold increased resistance to colistin (Napier et al, 2014). A
lularly residing S. aureus (Dziewanowska et al, 1999; Jevon et al, murine infection model demonstrated that colistin therapy failed to
1999; Fraunholz & Sinha, 2012; Lehar et al, 2015) are protected from successfully treat an infection with a heteroresistant strain of E.
A Genetically determined
Control Stress-exposed auxotrophic mutants
Exponential growth e.g. • pH • Abscess e.g. • ΔmenD
• ROS • Temperature • ΔhemB
• Intracellular
millieu
Larger colonies the of same size Non-stable small colonies (nsSCs) Small colony variants (SCVs)
of colonies
of colonies
Proportion
Proportion
Proportion
50 50 50
of colonies
of colonies
Proportion
Proportion
Proportion
75 75 75
50 50 50
25 25 25
B
Single cell
Lag
level
Exponential
growth
Colony radius
Colony radius
Colony radius
Slow
Delayed growth
growth rate
© EMBO
Figure 3.
◀ Figure 3. Characterization of colony radius heterogeneity and single-cell growth dynamics of bacteria.
(A) Stress-exposed S. aureus and genetically determined auxotrophic mutants show small colony phenotypes. Top: The stress-exposed bacteria display colony radius
heterogeneity on agar plates by forming non-stable small colonies (nsSCs), whereas the auxotrophic mutants grow as genetically determined small colony variants
(SCVs). Middle: The colony radius histograms for stress-triggered bacteria show a broad distribution towards smaller colony sizes compared to exponentially growing
bacteria (most colonies have the same size) and SCVs (most colonies are small and the same size). Bottom: Translated into colony appearance times, the exponentially
growing bacteria almost all appear at the same time early after plating, whereas the stress-exposed bacteria show a broad lag time distribution with some colonies
appearing early and others later. In contrast, SCVs all appear later after a longer incubation time, but almost all at the same time. (B) The schemes show bacterial
growth dynamics on a single-cell level as well as different scenarios explaining the colony size at a certain time point. Short arrows indicate a fast division rate of wild
type bacteria (yellow), whereas longer arrows represent a slower growth rate for SCV forming mutants (green). Colonies can be small because of a late onset of growth
(lag time) or because of a slower growth rate.
cloacae, whereas infection with a fully susceptible strain could be lag and by slow growth (Ishida et al, 1982; Keren et al, 2004; Lewis,
treated with colistin and the mice survived, highlighting the impor- 2019).
tance of HR in infection settings and the adverse effect on treatment Balaban and co-workers used the standard disk diffusion assay
outcome (Band et al, 2016). as a basis to develop a semi-quantitative test for tolerance levels,
the so-called Tolerance Disk (TD) test (Gefen et al, 2017) (Fig 4B).
The TD test uses the diffusion and degradation of antibiotics in the
Detection of antibiotic tolerance and persistence in the agar after removal of the antibiotic-containing disc and enables
clinical setting the regrowth of tolerant and persistent bacteria that survived in the
inhibition zone after addition of fresh glucose. This test allowed
In the clinical microbiology laboratory, the options to test for antibi- detection of high levels of tolerance in clinical isolates of E. coli and,
otic persistence and tolerance are limited and typically focus on the most importantly, identified antibiotics that were able to kill these
detection of antibiotic resistance. Additionally, prevalence and pres- surviving bacteria. Therefore, the TD test might help tailoring the
ence of bacterial persisters and their clinical relevance in infection treatment regimens to treat persistent and recurrent infections.
per se are not well understood. One reason for this very limited test- However, this test cannot detect triggered antibiotic tolerance/per-
ing for tolerance in clinical microbiology laboratories is that there is sistence as this requires exposure to certain stressors like acidic pH
no quick and accurate testing protocol available so far. For resis- or nutrient limitation, as found in the host. Moreover, the standard
tance testing, the Kirby Bauer disc diffusion assay (Bauer et al, antibiotic discs used in clinical microbiology laboratories contain
1966) and the E-test (Joyce et al, 1992) are used to measure very high concentrations of antibiotics leading to high residual drug
increased MIC levels. These tests cannot be used for tolerance test- concentrations in the agar inhibiting regrowth of bacteria. Ideally,
ing, as antibiotic tolerance is defined as the ability of a bacterial discs should contain antibiotic concentrations high enough to create
population to survive antibiotic exposure without any changes in a large enough inhibition zone but low enough to fall below the
the MIC, meaning without resistance. MIC after 18 h. All these problematics make it difficult to standard-
Antibiotic tolerance can be determined by performing killing ize the TD test for clinical use.
assays in form of time–kill curves that are characterized by a Another way to test for antibiotic tolerance is to use the
bimodal killing in the case of antibiotic persistence, but this is “Replica Plating Tolerance Isolation System” (REPTIS) developed
very labour-intensive and shows high variability, which makes by Hiramatsu and co-workers to successfully identify and select
many repetitions necessary and undermines feasibility in routine ciprofloxacin (CIP)-tolerant mutants in S. aureus (Matsuo et al,
clinical microbiology laboratories (Fig 4A). In addition, the levels 2019) (Fig 4C). REPTIS does not require adjusting the antibiotic
of antibiotic persistence observed and evaluated in laboratories concentrations, overcoming this limitation of the TD test. Shortly,
under in vitro conditions might not correspond to the levels of instead of replacing the antibiotic disc by a glucose disc, this
persistence occurring in the patient from whom the strain was method uses a sterile silk cloth to transfer colony-forming units
isolated considering the complexities that contribute to this unsta- (CFUs) onto a fresh plate (replica plate) where surviving bacteria
ble phenomenon. Therefore, measurements of antibiotic persis- can regrow, and antibiotic tolerance is determined by the number
tence under laboratory conditions are only approximations of of growing bacteria in the former inhibition zone. Again, this test
bacterial phenotypes of infections in vivo. Yet, they provide impor- cannot detect triggered antibiotic persistence but has the potential
tant additional information that might help physicians in their to be adapted for an automated use in diagnostic microbiology
therapeutic decision-making. laboratories.
Many studies used the minimal bactericidal concentration (MBC) As mentioned above, the ScanLag method (Levin-Reisman
of an antibiotic necessary to kill 99.9% of the bacterial population et al, 2010) and ColTapp (B€ ar et al, 2020) can be used to detect
in 24 h to test for antibiotic tolerance (Handwerger & Tomasz, and measure colony growth heterogeneity and derive bacterial
1985). Although it was already known that tolerance is defined by a colonies’ lag times, which serve as a proxy for antibiotic toler-
decreased rate of killing, for clinical reasons a strain is considered ance (Fig 4D). To further validate and confirm the results from
tolerant, when the MBC is at least 32 times higher than the MIC. An the colony analysis at the single-cell level, single bacterial cell
MBC/MIC ratio of > 32 is defined as tolerance (Handwerger & microscopy can be used. However, these methods need specific
Tomasz, 1985; Jones, 2006; Gonzalez et al, 2013). However, the setups and equipment, laboratory space and are quite labour-
MBC/MIC ratio is not well suited to determine and measure antibi- intensive, which limits their usage in routine clinical microbiology
otic tolerance, and it especially fails to detect tolerance conferred by laboratories. For specific clinical cases of persistent and difficult-
0.01
Persistent
Susceptible
Prolonged
lag
MDK 99
© EMBO
Sterile velvet
on a block Concentration [mg/L]
to-treat infections, where physicians suspect persister cell forma- MDK99 can be evaluated by a statistical analysis of measurements
tion, these methods could be used to improve and enable individ- performed manually or automated using a robotic system. This
ual patient-tailored treatments, but a broad standard testing might makes it interesting for standard diagnostic testing. Additionally,
not be suitable. the MDK99 evaluation has the inherent advantage of containing
Balaban and colleagues introduced a metric and an automated duration, an essential feature of persisters surviving stress over a
experimental framework for measuring antibiotic tolerance and period of time, is therefore superior to the MBC/MIC ratio evalua-
persistence, by determining the minimal duration of killing 99% tion, which is poorly linked to antibiotic tolerance (Keren et al,
of the population (MDK99) (Brauner et al, 2017) (Fig 4E). The 2004).
Future treatment regimens and novel antibiotics and exhibits bactericidal activity against a wide range
therapeutic approaches of pathogens including M. tuberculosis, carbapenem-resistant
Enterobacteriaceae, C. difficile and pan-resistant A. baumannii.
Despite the increase in antimicrobial resistance and the growing Interestingly, halicin disrupts the flow of protons across the bacte-
awareness of antibiotic persistence as a clinically relevant issue, the rial cell membrane (Fig 5A), a novel killing method compared to
development of new antimicrobial substances is declining. The conventional antibiotics. Additionally, this study identified eight
reasons for this are complex but are associated with the current other antibacterial substances that are structurally different from
focus of the pharmaceutical industry on medication for non- conventional antibiotics and highlights the opportunities and
infectious chronic diseases such as cancer, metabolic and cardiovas- impact machine learning can have on antibiotic discovery and
cular disorders and a lack of confidence in the profitability of new prediction of properties of potential drugs while decreasing the
antibiotics. However, there are novel strategies and substances in cost of screening efforts (Stokes et al, 2020). However, these
basic research and preclinical studies that might offer treatment results will have to be carefully evaluated and tested in animal
options in the near future (Fig 5). models including pharmacokinetic and toxicology studies before
moving to clinical trials.
Novel antimicrobials against a broad-spectrum of Based on the structure of a natural antibacterial peptide, Nicolas
multidrug-resistant bacteria and colleagues generated a new family of peptidomimetics, the
The recently discovered chimeric peptidomimetic antibiotics possess cyclic hepta-pseudopeptides that show antibacterial activity against
a broad-spectrum antimicrobial activity against Gram-negative Gram-positive and Gram-negative multiresistant pathogens while
bacteria, including all Gram-negative ESKAPE pathogens (Luther having limited potential to lead to resistance development (Nicolas
et al, 2019). Luther and colleagues showed that the bactericidal et al, 2019) (Fig 5A). They identified two peptide analogues that
activity of these chimeric antibiotics involves binding to both were active against MRSA and P. aeruginosa in severe sepsis and
lipopolysaccharide and the main component of the b-barrel folding skin infection murine models and exhibited low nephrotoxicity.
complex, BamA, that is required for the folding and insertion of b- Despite these promising results, their efficacy in deep-seated infec-
barrel proteins into the outer membrane of Gram-negative bacteria, tions is still to be shown. Both compounds are considered potential
explaining the specific targeting of Gram-negative bacteria while candidates for drug development; however, extensive pharmacoki-
leaving eukaryotic cell membranes intact (Fig 5A). Yet, the exact netic and toxicology studies will be necessary before starting phase I
mechanism driving the killing of Gram-negative bacteria remains an clinical trials (Nicolas et al, 2019).
open question. The leading candidate POL7306 underwent preclini- Another approach taken towards the effort of clearing antibiotic-
cal toxicology studies. Before moving into clinical trials, the focus is resistant Gram-positive and Gram-negative bacteria is the use of
currently on a new formulation of POL7306 as well as optimizing bacteriophages (Fig 5A), viruses that infect and lyse bacteria
peptide designs aiming to identify further candidates to broaden (Twort, 1915; Salmond & Fineran, 2015). Due to the success of
their therapeutic margins (Luther et al, 2019). antibiotics, bacteriophage therapy was abandoned in Western coun-
Generally, it is difficult to find new substances against Gram- tries, but is now being revived, because of increasing MDR bacteria
negative bacteria as they evolved an outer membrane that protects and studies show that bacteriophage therapy successfully treats
them from unwanted compounds (Li & Nikaido, 2004; Payne et al, urinary tract infections (UTIs) (Leitner et al, 2017) and burn wound
2007). Thus, Lewis and co-workers reasoned that compounds infections by P. aeruginosa (Jault et al, 2019). Nevertheless, there
against Gram-negative bacteria would be present in microorganisms are several potential disadvantages of phage therapy: (i) not all
that need to compete with Gram-negative bacteria. They screened phages are suitable for use as therapeutics. Temperate or toxin-
isolates of Photorhabdus, symbionts of entomopathogenic nematode carrying phages and those with poor killing potential against target
microbiomes, for antimicrobial substances against Gram-negative bacteria should be avoided. (ii) The narrow host spectrum, wherein
bacteria and isolated a new compound named darobactin (Imai different species or even different strains from the same species,
et al, 2019). In general, antimicrobial drugs target bacterial might need specific phages for eliciting efficient killing. Although
enzymes, but darobactin has been shown to stabilize the closed broadly acting phage cocktails are normally more selective in their
conformation of the chaperone BamA, inhibiting insertion of its spectrum of activity than conventional narrow-spectrum antibiotics,
substrates into the membrane (Imai et al, 2019) (Fig 5A). This it might be challenging to produce or at least provide all the needed
newly discovered antimicrobial drug might provide novel treatment phage cocktails. It might be even difficult to find the exact phage
options for infections with multidrug-resistant Gram-negative patho- needed for therapy. (iii) Phages are protein-based, biological agents
gens and shows that there are diverse molecules against pathogens that can actively replicate, evolve and interact with the hosts’
already available in nature. immune system. Especially, Western countries are not familiar with
Using a machine learning approach, Collins and co-workers phages and there is still hesitancy to use such biological entities in
discovered completely new kinds of antibiotics, without using any humans (Loc-Carrillo & Abedon, 2011).
previous human assumption (Stokes et al, 2020). Screening a
pool of more than 100 million molecules led to the discovery of Novel antimicrobials against a narrow-spectrum of multidrug-
c-Jun N-terminal kinase inhibitor SU3327 (De et al, 2009), a resistant bacteria
preclinical nitrothiazole under investigation as a treatment for Because of the discussed disadvantages of phage therapy, research
diabetes, exhibiting a strong antimicrobial activity. Halicin, has focused more specifically on phage-encoded peptidoglycan
renamed after HAL, the intelligent computer in the film “A Space hydrolases, the endolysins, which lyse bacterial cells (Haddad
Odyssey” from 2001, is structurally divergent from conventional Kashani et al, 2018) (Fig 5A). One of the biggest advantages of
BamA
Lysis
CRISPR-Cas9
Bactericidal cargo or
Virulence-blocking cargo
Membrane permeability
H+ Leakage H+
H+ Cyclic H+
Halicin ΔpH hepta-pseudopeptides ΔpH Lugdunin + LL-37 or + DCD-1(L)
PMF Antibody
H+
Growth stimulation
Stringent Translation
response Antimicrobial-susceptible
Transcription state
Translation blockade
Mistranslation
Non-specific and lethal
(p)ppGpp protease activity
ClpP
ATP
ATP
(p)ppGpp analogue Mitomycin C ADEP4 Retinoid derivatives
e.g. relacin • CD437
© EMBO
• CD1530
•…
Figure 5.
◀ Figure 5. New approaches to target multidrug-resistant bacteria and bacterial persister cells.
(A) Scheme showing novel strategies to target multidrug-resistant Gram-negative and Gram-positive bacteria. New approaches to target multiresistant bacteria include
darobactin, chimeric peptidomimetic antibiotics, antibacterial drones (ABD), bacteriophages, endolysins, halicin and cyclic hepta-pseudopeptides and lugdunin. (B)
Scheme of persister-targeting approaches. Different strategies have been suggested to specifically target bacterial persister cells, e.g. addition of cell wall targeting
substances like endolysins, NT-61, XF-73 or NCK10, simple sugars, bacterial opsonization by antibody–antibiotic conjugates (AACs), 3-[4-(4-methoxyphenyl)piperazin-1-
yl] piperidin-4-yl biphenyl-4-carboxylate (C10), cis-2-decenoic acid, (p)ppGpp analogues like relacin, mitomycin C, acyldeptipeptide (ADEP4) or retinoid acid derivatives
(e.g. CD437 and CD1530).
endolysins is their species specificity, avoiding selective pressure on Rather than directly targeting the pathogen, an alternative
commensal bacteria (Schmelcher et al, 2012). Another advantage is approach is to attenuate their pathogenicity by targeting specific
the low probability of bacteria evolving resistances against endoly- virulence factors involved in the infection process. The blocking of
sins, as phages and host bacteria coevolved and endolysins target virulence factors also helps avoiding resistance development by the
highly conserved structures in the bacterial cell wall (Borysowski bacteria, as less antibiotics would have to be used (Keyser et al,
et al, 2006). However, to overcome certain limitations, such as 2008). An important strategy is to hinder bacterial dissemination
immunogenicity, short serum half-lives, low penetration into and tissue invasion by blocking bacterial attachment to host cells
eukaryotic cells to kill intracellular bacteria and toxicity, many and translocation into host tissue. Mannosides have been shown to
research groups focus on engineering improved variants that might inhibit the FimH component of type I fimbriae of uropathogenic
offer novel treatment options in the future (Schmelcher et al, 2012; E. coli (UPEC) thereby significantly decreasing colonization of UPEC
Haddad Kashani et al, 2018; Ro €hrig et al, 2020). in a murine infection model (Klein et al, 2010).
Independent of discovering new antibiotic substances and phages, The synthetic antibody ScFv-Fc KP3 blocks the type 3 fimbrial
a new method using CRISPR-Cas9, was recently developed to kill or subunit in K. pneumonia and has been shown to confer protection
block multidrug-resistant S. aureus (Ram et al, 2018). In this study, in murine pneumonia models (Wang et al, 2016). By neutralizing
they replaced the staphylococcal pathogenicity islands’ (SaPIs) toxin toxin B from C. difficile, the monoclonal antibody bezlotoxumab
genes with antibacterial cargos to generate antibacterial drones prevents recurrent C. difficile infections in combination with antibi-
(ABDs) that target S. aureus in the host (Fig 5A). By constructing otics and has already been approved for the use in humans
ABDs with either a CRISPR–Cas9 bactericidal or a CRISPR–dCas9 (Mullard, 2016). Similarly, the monoclonal anti-a-toxin antibody,
virulence-blocking module, bacteria are either killed or disarmed, MEDI4893, has been shown to decrease the S. aureus burden and
leading to the abrogation of the infection. This was also shown in a increase wound healing in diabetic foot ulcers and phase II clinical
murine abscess model, where mice were rescued by treatment with studies have just been concluded (ClinicalTrials.gov, 2019). Despite
these ABDs (Ram et al, 2018). However, the ABD approach comes promising novel strategies and molecules targeting pathogens and
with some potential obstacles, e.g. simultaneous packaging of host their virulence factors, further research and alternative approaches
genes, induction of or recombination with resident SaPIs or recombi- are needed to ensure effective treatment regimens against multi-
nation with plasmids carrying virulence or resistance genes. Addi- drug-resistant bacteria in the future.
tionally, resistance to the ABDs can occur (Ram et al, 2018).
Taking a different approach, Krismer, Peschel and co-workers Antipersister strategies
identified the potent antimicrobial compound lugdunin from The recognition of the phenomenon of antibiotic persistence and its
S. lugdunensis (Zipperer et al, 2016). Rather than treating an already clinical importance in chronic and relapsing infections including its
existing infection, they suggest using lugdunin or lugdunin-produc- connection to resistance development has led to both basic and
ing commensal staphylococci to prevent S. aureus colonization and application-oriented research. Today, there are three main strategies
invasive infections in high-risk patients such as immunocompro- for treatment and prevention of infections associated with bacterial
mised or before elective surgeries. The macrocyclic thiazolidine persisters: (i) directly targeting persister cells; (ii) inhibiting forma-
peptide antibiotic lugdunin is active against a wide range of Gram- tion of bacterial persisters; and (iii) resuscitating persisters and
positive pathogens including MRSA and VRE with a high barrier to sensitizing them to conventional antibiotic therapy.
resistance by mutation (Zipperer et al, 2016) (Fig 5A). It exhibits
immunomodulatory activity by inducing the expression of the host Directly targeting persister cells
AMP LL-37 and the pro-inflammatory chemokines CXCL8 and MIP-2 As mentioned above, persisters are often characterized by slow
in human keratinocytes, which leads to the recruitment of mono- growth or dormancy rendering antibiotic targets inactive. One of the
cytes and neutrophils potentially contributing to effective bacterial obvious targets for directly attacking persisters without the require-
eradication (Bitschar et al, 2019). Moreover, lugdunin acts synergis- ment of active targets is the bacterial membrane and cell wall.
tically with the human AMPs LL-37 and DCD-1(L) in killing Phage-derived endolysins and other synthetic lysins degrade the
S. aureus. The antimicrobial activity against S. aureus is strongly peptidoglycans in the cell wall independent of the presence of
associated with the disruption of the membrane potential without metabolically active and growing bacteria, targeting both growing
lysing the bacterial cells, demonstrating that the mode of action is and non-growing populations (Gutierrez et al, 2014; Rodrıguez-
translocation of protons (Schilling et al, 2019). Lugdunin could be €hrig et al, 2020) (Fig 5B).
Rubio et al, 2016; Ro
used either directly as a topically applied drug to inhibit growth of Synthetic retinoid acid derivatives, specifically CD437 and
S. aureus or indirectly applied with a safe probiotic strain such as an CD1530, have been shown to penetrate and disrupt the lipid bilayer
avirulent S. lugdunensis or an exclusively commensal species of the cell membrane of MRSA thereby killing growing and
containing the lug genes (Zipperer et al, 2016). non-growing bacteria (Fig 5B). Additionally, CD437 alone or in
made, we still do not fully understand how bacteria form persisters. Azarian T, Daum RS, Petty LA, Steinbeck JL, Yin Z, Nolan D, Boyle-Vavra S,
Clearly, the interplay of multiple mechanisms and the resulting Hanage WP, Salemi M, David MZ (2016) Intrahost evolution of methicillin-
phenotypic heterogeneity is beneficial for the survival of a bacterial resistant Staphylococcus aureus USA300 among individuals with
population during infection. Recent advances show novel therapeu- reoccurring skin and soft-tissue infections. J Infect Dis 214: 895 – 905
tic strategies aiming to eradicate such persisters, desperately needed Baddour LM, Barker LP, Christensen GD, Parisi JT, Simpson WA (1990)
to shorten treatment duration and thus reducing morbidity remark- Phenotypic variation of Staphylococcus epidermidis in infection of
ably. However, further research and focus on modulating the host transvenous endocardial pacemaker electrodes. J Clin Microbiol 28:
environment is needed to successfully tackle antibiotic resistance 676 – 679
and persistence. Novel technologies including the development of Baharoglu Z, Mazel D (2014) SOS, the formidable strategy of bacteria against
cutting-edge techniques such as single-cell analysis, microfluidics, aggressions. FEMS Microbiol Rev 38: 1126 – 1145
dual-RNAseq and bacterial sorting as well as bacterial epigenetics Balaban NQ, Helaine S, Lewis K, Ackermann M, Aldridge B, Andersson DI,
are being used and will be used more and more to track, enrich and Brynildsen MP, Bumann D, Camilli A, Collins JJ et al (2019a) Definitions
analyse rare, non-growing bacteria that are able to survive antibiotic and guidelines for research on antibiotic persistence. Nat Rev Microbiol 17:
therapy without being resistant. 441 – 448
Balaban NQ, Helaine S, Lewis K, Ackermann M, Aldridge B, Andersson DI,
Acknowledgements Brynildsen MP, Bumann D, Camilli A, Collins JJ et al (2019b) Publisher
We thank the members of the Zinkernagel laboratory for feedbacks and proof- correction: definitions and guidelines for research on antibiotic
reading of the manuscript, especially Judith Bergada Pijuan for her help with persistence. Nat Rev Microbiol 17: 460
the presentation of the resistance data. The authors apologise to colleagues in Balaban NQ, Merrin J, Chait R, Kowalik L, Leibler S (2004) Bacterial
the field for being unable to cite all relevant studies and thank the reviewers persistence as a phenotypic switch. Science 305: 1622 – 1625
for their helpful comments. This work was funded by the SNSF project grant Band VI, Crispell EK, Napier BA, Herrera CM, Tharp GK, Vavikolanu K, Pohl J,
31003A_176252 (to A.S.Z); by the Swedish Society for Medical Research (SSMF) Read TD, Bosinger SE, Trent MS et al (2016) Antibiotic failure mediated by
foundation grant P17-0179 (to S.M.S) and the Promedica Foundation 1449/M a resistant subpopulation in Enterobacter cloacae. Nat Microbiol 1: 16053
(to S.D.B). B€
ar J, Boumasmoud M, Kouyos RD, Zinkernagel AS, Vulin C (2020) Efficient
microbial colony growth dynamics quantification with ColTapp, an
Conflict of interest automated image analysis application. Sci Rep 10: 16084
The authors declare that they have no conflict of interest. Baroud M, Dandache I, Araj GF, Wakim R, Kanj S, Kanafani Z, Khairallah M,
Sabra A, Shehab M, Dbaibo G et al (2013) Underlying mechanisms of
carbapenem resistance in extended-spectrum beta-lactamase-producing
References Klebsiella pneumoniae and Escherichia coli isolates at a tertiary care centre
in Lebanon: role of OXA-48 and NDM-1 carbapenemases. Int J Antimicrob
Aakre CD, Phung TN, Huang D, Laub MT (2013) A bacterial toxin inhibits DNA Agents 41: 75 – 79
replication elongation through a direct interaction with the beta sliding Bates DM, von Eiff C, McNamara PJ, Peters G, Yeaman MR, Bayer AS, Proctor
clamp. Mol Cell 52: 617 – 628 RA (2003) Staphylococcus aureus menD and hemB mutants are as infective
Abele-Horn M, Schupfner B, Emmerling P, Waldner H, Goring H (2000) as the parent strains, but the menadione biosynthetic mutant persists
Persistent wound infection after herniotomy associated with small-colony within the kidney. J Infect Dis 187: 1654 – 1661
variants of Staphylococcus aureus. Infection 28: 53 – 54 Bauer AW, Kirby WM, Sherris JC, Turck M (1966) Antibiotic susceptibility
Ackermann M (2015) A functional perspective on phenotypic heterogeneity in testing by a standardized single disk method. Am J Clin Pathol 45:
microorganisms. Nat Rev Microbiol 13: 497 – 508 493 – 496
Adedeji WA (2016) The treasure called antibiotics. Ann Ib Postgrad Med 14: Benoit JB, Frank DN, Bessesen MT (2018) Genomic evolution of
56 – 57 Staphylococcus aureus isolates colonizing the nares and progressing to
Allison KR, Brynildsen MP, Collins JJ (2011) Metabolite-enabled eradication of bacteremia. PLoS One 13: e0195860
bacterial persisters by aminoglycosides. Nature 473: 216 – 220 Berglund B (2019) Acquired resistance to colistin via chromosomal and
Amato S, Fazen C, Henry T, Mok W, Orman M, Sandvik E, Volzing K, plasmid-mediated mechanisms in Klebsiella pneumoniae. Infect Microb Dis 1
Brynildsen M (2014) The role of metabolism in bacterial persistence. Front Bernier SP, Lebeaux D, DeFrancesco AS, Valomon A, Soubigou G, Coppee JY,
Microbiol 5: 70 Ghigo JM, Beloin C (2013) Starvation, together with the SOS response,
Amato SM, Orman MA, Brynildsen MP (2013) Metabolic control of persister mediates high biofilm-specific tolerance to the fluoroquinolone ofloxacin.
formation in Escherichia coli. Mol Cell 50: 475 – 487 PLoS Genet 9: e1003144
Aminov RI (2010) A brief history of the antibiotic era: lessons learned and Besier S, Smaczny C, von Mallinckrodt C, Krahl A, Ackermann H, Brade V,
challenges for the future. Front Microbiol 1: 134 Wichelhaus TA (2007) Prevalence and clinical significance of
Anderson SE, Sherman EX, Weiss DS, Rather PN (2018) Aminoglycoside Staphylococcus aureus small-colony variants in cystic fibrosis lung disease.
heteroresistance in Acinetobacter baumannii AB5075. mSphere 3: e00271-18 J Clin Microbiol 45: 168 – 172
Appelbaum PC (2007) Microbiology of antibiotic resistance in Staphylococcus Bhattacharyya S, Roy S, Mukhopadhyay P, Rit K, Dey J, Ganguly U, Ray R
aureus. Clin Infect Dis 45(Suppl 3): S165 – 170 (2012) Small Colony variants of Staphylococcus aureus isolated from a
Aslam B, Wang W, Arshad MI, Khurshid M, Muzammil S, Rasool MH, patient with infective endocarditis: a case report and review of the
Nisar MA, Alvi RF, Aslam MA, Qamar MU et al (2018) Antibiotic literature. Iran J Microbiol 4: 98 – 99
resistance: a rundown of a global crisis. Infect Drug Resist 11: Bigger J (1944) Treatment of staphylococcal infections with penicillin by
1645 – 1658 intermittent sterilisation. Lancet 244: 497 – 500
Bitschar K, Sauer B, Focken J, Dehmer H, Moos S, Konnerth M, Schilling NA, Coffey JW, De Duve C (1968) Digestive activity of lysosomes. I. The digestion
Grond S, Kalbacher H, Kurschus FC et al (2019) Lugdunin amplifies innate of proteins by extracts of rat liver lysosomes. J Biol Chem 243: 3255 – 3263
immune responses in the skin in synergy with host- and microbiota- Conlon BP, Nakayasu ES, Fleck LE, LaFleur MD, Isabella VM, Coleman K,
derived factors. Nat Commun 10: 2730 Leonard SN, Smith RD, Adkins JN, Lewis K (2013) Activated ClpP kills
Bjarnsholt T (2013) The role of bacterial biofilms in chronic infections. APMIS persisters and eradicates a chronic biofilm infection. Nature 503: 365 – 370
121: 1 – 58 Conlon BP, Rowe SE, Gandt AB, Nuxoll AS, Donegan NP, Zalis EA, Clair G,
Black DS, Irwin B, Moyed HS (1994) Autoregulation of hip, an operon that Adkins JN, Cheung AL, Lewis K (2016) Persister formation in Staphylococcus
affects lethality due to inhibition of peptidoglycan or DNA synthesis. J aureus is associated with ATP depletion. Nat Microbiol 1: 16051
Bacteriol 176: 4081 – 4091 Costerton JW, Stewart PS, Greenberg EP (1999) Bacterial biofilms: a common
Black DS, Kelly AJ, Mardis MJ, Moyed HS (1991) Structure and organization of cause of persistent infections. Science 284: 1318 – 1322
hip, an operon that affects lethality due to inhibition of peptidoglycan or Davidson DJ, Spratt D, Liddle AD (2019) Implant materials and prosthetic joint
DNA synthesis. J Bacteriol 173: 5732 – 5739 infection: the battle with the biofilm. EFORT Open Rev 4: 633 – 639
Blair JM, Webber MA, Baylay AJ, Ogbolu DO, Piddock LJ (2015) Molecular Davies DG, Marques CNH (2009) A fatty acid messenger is responsible for
mechanisms of antibiotic resistance. Nat Rev Microbiol 13: 42 – 51 inducing dispersion in microbial biofilms. J Bacteriol 191: 1393 – 1403
Boekhoud IM, Hornung BVH, Sevilla E, Harmanus C, Bos-Sanders IMJG, De Surya K, Stebbins JL, Chen LH, Riel-Mehan M, Machleidt T, Dahl R, Yuan
Terveer EM, Bolea R, Corver J, Kuijper EJ, Smits WK (2020) Plasmid- H, Emdadi A, Barile E, Chen V et al (2009) Design, synthesis, and
mediated metronidazole resistance in Clostridioides difficile. Nat Commun structure-activity relationship of substrate competitive, selective, and
11: 598 in vivo active triazole and thiadiazole inhibitors of the c-Jun N-terminal
Borysowski J, Weber-Daz browska B, Go rski A (2006) Bacteriophage endolysins kinase. J Med Chem 52: 1943 – 1952
as a novel class of antibacterial agents. Exp Biol Med 231: 366 – 377 Del Pozo JL, Patel R (2009) Clinical practice. Infection associated with
Boucher HW, Talbot GH, Bradley JS, Edwards JE, Gilbert D, Rice LB, Scheld prosthetic joints. N Engl J Med 361: 787 – 794
M, Spellberg B, Bartlett J (2009) Bad bugs, no drugs: no ESKAPE! an Dengler Haunreiter V, Boumasmoud M, Haffner N, Wipfli D, Leimer N,
update from the Infectious Diseases Society of America. Clin Infect Dis Rachmuhl C, Kuhnert D, Achermann Y, Zbinden R, Benussi S et al (2019)
48: 1 – 12 In-host evolution of Staphylococcus epidermidis in a pacemaker-associated
Brantl S, Jahn N (2015) sRNAs in bacterial type I and type III toxin-antitoxin endocarditis resulting in increased antibiotic tolerance. Nat Commun 10:
systems. FEMS Microbiol Rev 39: 413 – 427 1149
Brauner A, Fridman O, Gefen O, Balaban NQ (2016) Distinguishing between Dingsdag SA, Hunter N (2018) Metronidazole: an update on metabolism,
resistance, tolerance and persistence to antibiotic treatment. Nat Rev structure-cytotoxicity and resistance mechanisms. J Antimicrob Chemother
Microbiol 14: 320 – 330 73: 265 – 279
Brauner A, Shoresh N, Fridman O, Balaban NQ (2017) An experimental Dolejska M, Villa L, Poirel L, Nordmann P, Carattoli A (2013) Complete
framework for quantifying bacterial tolerance. Biophys J 112: 2664 – 2671 sequencing of an IncHI1 plasmid encoding the carbapenemase NDM-1,
Brooun A, Liu S, Lewis K (2000) A dose-response study of antibiotic resistance the ArmA 16S RNA methylase and a resistance-nodulation-cell
in Pseudomonas aeruginosa biofilms. Antimicrob Agents Chemother 44: division/multidrug efflux pump. J Antimicrob Chemother 68: 34 – 39
640 – 646 Domenech M, Ramos-Sevillano E, Garcia E, Moscoso M, Yuste J (2013) Biofilm
Brown PD, Ngeno C (2007) Antimicrobial resistance in clinical isolates of formation avoids complement immunity and phagocytosis of
Staphylococcus aureus from hospital and community sources in southern Streptococcus pneumoniae. Infect Immun 81: 2606 – 2615
Jamaica. Int J Infect Dis 11: 220 – 225 Dorr T, Vulic M, Lewis K (2010) Ciprofloxacin causes persister formation by
CDC (2019). Antibiotic resistance threats in the united states, 2019, in: inducing the TisB toxin in Escherichia coli. PLoS Biol 8: e1000317
Atlanta G.U.S.D.o.H.a.H.S. (Ed.). CDC. Dziewanowska K, Patti JM, Deobald CF, Bayles KW, Trumble WR, Bohach GA
Chambers HF, Deleo FR (2009) Waves of resistance: Staphylococcus aureus in (1999) Fibronectin binding protein and host cell tyrosine kinase are
the antibiotic era. Nat Rev Microbiol 7: 629 – 641 required for internalization of Staphylococcus aureus by epithelial cells.
Chang S, Sievert DM, Hageman JC, Boulton ML, Tenover FC, Downes FP, Shah Infect Immun 67: 4673 – 4678
S, Rudrik JT, Pupp GR, Brown WJ et al (2003) Infection with vancomycin- ECDC (2018a) Healthcare-associated infections: Clostridium difficile infections.
resistant Staphylococcus aureus containing the vanA resistance gene. N In: ECDC. Annual epidemiological report for 2016. Stockholm: ECDC
Engl J Med 348: 1342 – 1347 ECDC (2018b) Surveillance of antimicrobial resistance in Europe 2017.
Chatterjee I, Kriegeskorte A, Fischer A, Deiwick S, Theimann N, Proctor RA, ECDC (2019) Surveillance of antimicrobial resistance in Europe 2018,
Peters G, Herrmann M, Kahl BC (2008) In vivo mutations of thymidylate Stockholm: European Centre for Disease Prevention and Control
synthase (encoded by thyA) are responsible for thymidine dependency in Elsner HA, Sobottka I, Mack D, Claussen M, Laufs R, Wirth R (2000) Virulence
clinical small-colony variants of Staphylococcus aureus. J Bacteriol 190: factors of Enterococcus faecalis and Enterococcus faecium blood culture
834 – 842 isolates. Eur J Clin Microbiol Infect Dis 19: 39 – 42
€te P, Chirkova A, Personnic N, Zankl J, Schu
Claudi B, Spro €rmann N, Schmidt A, Fauvart M, De Groote VN, Michiels J (2011) Role of persister cells in chronic
Bumann D (2014) Phenotypic variation of salmonella in host tissues infections: clinical relevance and perspectives on anti-persister therapies. J
delays eradication by antimicrobial chemotherapy. Cell 158: 722 – 733 Med Microbiol 60: 699 – 709
Clements MO, Watson SP, Poole RK, Foster SJ (1999) CtaA of Staphylococcus Fernandez De Henestrosa AR, Ogi T, Aoyagi S, Chafin D, Hayes JJ, Ohmori H,
aureus is required for starvation survival, recovery, and cytochrome Woodgate R (2000) Identification of additional genes belonging to the
biosynthesis. J Bacteriol 181: 501 – 507 LexA regulon in Escherichia coli. Mol Microbiol 35: 1560 – 1572
ClinicalTrials.gov (2019) Study of the efficacy and safety of MEDI4893 Fisher RA, Gollan B, Helaine S (2017) Persistent bacterial infections and
(SAATELLITE). https://clinicaltrials.gov/ct2/show/study/NCT02296320 persister cells. Nat Rev Microbiol 15: 453 – 464
Fleming A (1929) On antibacterial action of culture of Penicillium, with Hansen S, Vulic M, Min J, Yen TJ, Schumacher MA, Brennan RG, Lewis K
special reference to their use in isolation of B. influenzae. Br J Exp Pathol (2012) Regulation of the Escherichia coli HipBA toxin-antitoxin system by
10: 226 – 236 proteolysis. PLoS One 7: e39185
FOPH (2018) Swiss Antibiotic Resistance Report 2018. Usage of Antibiotics Harkins CP, Pichon B, Doumith M, Parkhill J, Westh H, Tomasz A, de
and Occurrence of Antibiotic Resistance in Bacteria from Humans and Lencastre H, Bentley SD, Kearns AM, Holden MTG (2017) Methicillin-
Animals in Switzerland. resistant Staphylococcus aureus emerged long before the introduction of
Foster TJ (2017) Antibiotic resistance in Staphylococcus aureus. Current status methicillin into clinical practice. Genome Biol 18: 130
and future prospects. FEMS Microbiol Rev 41: 430 – 449 Harms A, Brodersen DE, Mitarai N, Gerdes K (2018) Toxins, targets, and
Fraunholz M, Sinha B (2012) Intracellular Staphylococcus aureus: live-in and triggers: an overview of toxin-antitoxin biology. Mol Cell 70: 768 – 784
let die. Front Cell Infect Microbiol 2: 43 Hartman BJ, Tomasz A (1984) Low-affinity penicillin-binding protein
Frey PM, Marti GR, Droz S, Derochevonarx M, Suter-Riniker F, Aujesky D, associated with beta-lactam resistance in Staphylococcus aureus. J
Brugger SD (2019) Bacterial colonization of handheld devices in a tertiary Bacteriol 158: 513 – 516
care setting: a hygiene intervention study. Antimicrobial Resist Infect Control Hassoun A, Linden PK, Friedman B (2017) Incidence, prevalence, and
8: 97 management of MRSA bacteremia across patient populations-a review of
Fukuoka T, Ohya S, Narita T, Katsuta M, Iijima M, Masuda N, Yasuda H, Trias recent developments in MRSA management and treatment. Crit Care 21: 211
J, Nikaido H (1993) Activity of the carbapenem panipenem and role of the Helaine S, Cheverton AM, Watson KG, Faure LM, Matthews SA, Holden DW
OprD (D2) protein in its diffusion through the Pseudomonas aeruginosa (2014) Internalization of Salmonella by macrophages induces formation of
outer membrane. Antimicrob Agents Chemother 37: 322 – 327 nonreplicating persisters. Science 343: 204 – 208
Gefen O, Chekol B, Strahilevitz J, Balaban NQ (2017) TDtest: easy detection of Hiramatsu K, Aritaka N, Hanaki H, Kawasaki S, Hosoda Y, Hori S, Fukuchi Y,
bacterial tolerance and persistence in clinical isolates by a modified disk- Kobayashi I (1997) Dissemination in Japanese hospitals of strains of
diffusion assay. Sci Rep 7: 41284 Staphylococcus aureus heterogeneously resistant to vancomycin. Lancet
Germain E, Castro-Roa D, Zenkin N, Gerdes K (2013) Molecular mechanism of 350: 1670 – 1673
bacterial persistence by HipA. Mol Cell 52: 248 – 254 Hjort K, Nicoloff H, Andersson DI (2016) Unstable tandem gene amplification
Ghosh C, Manjunath GB, Konai MM, Uppu DS, Hoque J, Paramanandham K, generates heteroresistance (variation in resistance within a population) to
Shome BR, Haldar J (2015) Aryl-Alkyl-lysines: agents that kill planktonic colistin in Salmonella enterica. Mol Microbiol 102: 274 – 289
cells, persister cells, biofilms of MRSA and protect mice from skin- Hobby GL, Meyer K, Chaffee E (1942) Observations on the mechanism of
infection. PLoS One 10: e0144094 action of penicillin. Proc Soc Exp Biol Med 50: 281 – 285
Gilligan PH, Gage PA, Welch DF, Muszynski MJ, Wait KR (1987) Prevalence of Hong Y-K, Lee J-Y, Ko KS (2018) Colistin resistance in Enterobacter spp.
thymidine-dependent Staphylococcus aureus in patients with cystic isolates in Korea. Journal of Microbiology 56: 435 – 440
fibrosis. J Clin Microbiol 25: 1258 – 1261 Honkanen M, J€
amsen E, Karppelin M, Huttunen R, Eskelinen A, Syrj€
anen J
Gollan B, Grabe G, Michaux C, Helaine S (2019) Bacterial persisters and (2019) Periprosthetic Joint infections as a consequence of bacteremia.
infection: past, present, and progressing. Annu Rev Microbiol 73: 359 – 385 Open Forum Infecti Dis 6
Gonzalez N, Sevillano D, Alou L, Cafini F, Gimenez MJ, Gomez-Lus ML, Prieto Hooper DC, Jacoby GA (2015) Mechanisms of drug resistance: quinolone
J, Aguilar L (2013) Influence of the MBC/MIC ratio on the antibacterial resistance. Ann N Y Acad Sci 1354: 12 – 31
activity of vancomycin versus linezolid against methicillin-resistant Hornef MW, Wick MJ, Rhen M, Normark S (2002) Bacterial strategies for
Staphylococcus aureus isolates in a pharmacodynamic model simulating overcoming host innate and adaptive immune responses. Nat Immunol 3:
serum and soft tissue interstitial fluid concentrations reported in diabetic 1033 – 1040
patients. J Antimicrob Chemother 68: 2291 – 2295 Hu Y, Shamaei-Tousi A, Liu Y, Coates A (2010) A new approach for the
Goudie JG, Goudie RB (1955) Recurrent infections by a stable dwarf-colony discovery of antibiotics by targeting non-multiplying bacteria: a novel
variant of Staphylococcus aureus. J Clin Pathol 8: 284 – 287 topical antibiotic for staphylococcal infections. PLoS One 5: e11818
Gutierrez A, Jain S, Bhargava P, Hamblin M, Lobritz MA, Collins JJ (2017) Huh K, Kang CI, Kim J, Cho SY, Ha YE, Joo EJ, Chung DR, Lee NY, Peck KR,
Understanding and sensitizing density-dependent persistence to Song JH (2014) Risk factors and treatment outcomes of bloodstream
quinolone antibiotics. Mol Cell 68: 1147 – 1154.e3 infection caused by extended-spectrum cephalosporin-resistant
Gutierrez D, Ruas-Madiedo P, Martinez B, Rodriguez A, Garcia P (2014) Enterobacter species in adults with cancer. Diagn Microbiol Infect Dis 78:
Effective removal of staphylococcal biofilms by the endolysin LysH5. PLoS 172 – 177
One 9: e107307 Imai Y, Meyer KJ, Iinishi A, Favre-Godal Q, Green R, Manuse S, Caboni M, Mori
Haddad Kashani H, Schmelcher M, Sabzalipoor H, Seyed Hosseini E, Moniri R M, Niles S, Ghiglieri M et al (2019) A new antibiotic selectively kills Gram-
(2018) Recombinant endolysins as potential therapeutics against negative pathogens. Nature 576: 459 – 464
antibiotic-resistant Staphylococcus aureus: current status of research and Irving SE, Corrigan RM (2018) Triggering the stringent response: signals
novel delivery strategies. Clin Microbiol Rev 31: e00071 – e117 responsible for activating (p)ppGpp synthesis in bacteria. Microbiology 164:
Hameed F, Khan MA, Muhammad H, Sarwar T, Bilal H, Rehman TU (2019) 268 – 276
Plasmid-mediated mcr-1 gene in Acinetobacter baumannii and Pseudomonas Ishida K, Guze PA, Kalmanson GM, Albrandt K, Guze LB (1982) Variables in
aeruginosa: first report from Pakistan. Rev Soc Bras Med Trop 52: e20190237 demonstrating methicillin tolerance in Staphylococcus aureus strains.
Handwerger S, Tomasz A (1985) Antibiotic tolerance among clinical isolates of Antimicrob Agents Chemother 21: 688 – 690
bacteria. Rev Infect Dis 7: 368 – 386 Jansen B, Peters G (1993) Foreign body associated infection. J Antimicrobial
Hansen S, Lewis K, Vulic M (2008) Role of global regulators and nucleotide Chemother 32(suppl A): 69 – 75
metabolism in antibiotic tolerance in Escherichia coli. Antimicrob Agents Jault P, Leclerc T, Jennes S, Pirnay JP, Que YA, Resch G, Rousseau AF, Ravat F,
Chemother 52: 2718 – 2726 Carsin H, Le Floch R et al (2019) Efficacy and tolerability of a cocktail of
bacteriophages to treat burn wounds infected by Pseudomonas aeruginosa Kim JS, Heo P, Yang TJ, Lee KS, Cho DH, Kim BT, Suh JH, Lim HJ, Shin D, Kim
(PhagoBurn): a randomised, controlled, double-blind phase 1/2 trial. Lancet SK et al (2011) Selective killing of bacterial persisters by a single chemical
Infect Dis 19: 35 – 45 compound without affecting normal antibiotic-sensitive cells. Antimicrob
Jesaitis AJ, Franklin MJ, Berglund D, Sasaki M, Lord CI, Bleazard JB, Duffy JE, Agents Chemother 55: 5380 – 5383
Beyenal H, Lewandowski Z (2003) Compromised host defense on Kim W, Zhu W, Hendricks GL, Van Tyne D, Steele AD, Keohane CE, Fricke N,
Pseudomonas aeruginosa biofilms: characterization of neutrophil and Conery AL, Shen S, Pan W et al (2018) A new class of synthetic retinoid
biofilm interactions. J Immunol 171: 4329 – 4339 antibiotics effective against bacterial persisters. Nature 556: 103 – 107
Jevon M, Guo C, Ma B, Mordan N, Nair SP, Harris M, Henderson B, Bentley G, Kim W, Zou G, Hari TPA, Wilt IK, Zhu W, Galle N, Faizi HA, Hendricks GL, Tori
Meghji S (1999) Mechanisms of internalization of Staphylococcus aureus K, Pan W et al (2019) A selective membrane-targeting repurposed
by cultured human osteoblasts. Infect Immun 67: 2677 – 2681 antibiotic with activity against persistent methicillin-resistant
Johnson AP, Woodford N (2013) Global spread of antibiotic resistance: the Staphylococcus aureus. Proc Natl Acad Sci USA 116: 16529 – 16534
example of New Delhi metallo-beta-lactamase (NDM)-mediated €thi
Klein T, Abgottspon D, Wittwer M, Rabbani S, Herold J, Jiang X, Kleeb S, Lu
carbapenem resistance. J Med Microbiol 62: 499 – 513 C, Scharenberg M, Bezençon J et al (2010) FimH antagonists for the oral
Jolivet-Gougeon A, Bonnaure-Mallet M (2014) Biofilms as a mechanism of treatment of urinary tract infections: from design and synthesis to in vitro
bacterial resistance. Drug Discov Today Technol 11: 49 – 56 and in vivo evaluation. J Med Chem 53: 8627 – 8641
Jones RN (2006) Microbiological features of vancomycin in the 21st century: Kong H-K, Pan Q, Lo W-U, Liu X, Law COK, Chan T-f, Ho P-L, Lau TC-K (2018)
minimum inhibitory concentration creep, bactericidal/static activity, and Fine-tuning carbapenem resistance by reducing porin permeability of
applied breakpoints to predict clinical outcomes or detect resistant bacteria activated in the selection process of conjugation. Sci Rep 8: 15248
strains. Clin Infect Dis 42(Suppl 1): S13 – 24 Korch SB, Henderson TA, Hill TM (2003) Characterization of the hipA7 allele
Jorge P, Magalhaes AP, Grainha T, Alves D, Sousa AM, Lopes SP, Pereira of Escherichia coli and evidence that high persistence is governed by (p)
MO (2019) Antimicrobial resistance three ways: healthcare crisis, ppGpp synthesis. Mol Microbiol 50: 1199 – 1213
major concepts and the relevance of biofilms. FEMS Microbiol Ecol Korch SB, Hill TM (2006) Ectopic overexpression of wild-type and mutant
95: fiz115 hipA genes in Escherichia coli: effects on macromolecular synthesis and
Joyce LF, Downes J, Stockman K, Andrew JH (1992) Comparison of five persister formation. J Bacteriol 188: 3826 – 3836
methods, including the PDM Epsilometer test (E test), for antimicrobial Kosmidis C, Schindler BD, Jacinto PL, Patel D, Bains K, Seo SM, Kaatz GW
susceptibility testing of Pseudomonas aeruginosa. J Clin Microbiol 30: (2012) Expression of multidrug resistance efflux pump genes in clinical
2709 – 2713 and environmental isolates of Staphylococcus aureus. Int J Antimicrob
Kaatz GW, Seo SM, Dorman NJ, Lerner SA (1990) Emergence of teicoplanin Agents 40: 204 – 209
resistance during therapy of Staphylococcus aureus endocarditis. J Infect Kriegeskorte A, Block D, Drescher M, Windmu€ller N, Mellmann A, Baum C,
Dis 162: 103 – 108 Neumann C, Lore NI, Bragonzi A, Liebau E et al (2014) Inactivation of thyA
Kahl B, Herrmann M, Everding AS, Koch HG, Becker K, Harms E, Proctor RA, in Staphylococcus aureus attenuates virulence and has a strong impact on
Peters G (1998) Persistent infection with small colony variant strains of metabolism and virulence gene expression. mBio 5: e01447 – e11414
Staphylococcus aureus in patients with cystic fibrosis. J Infect Dis 177: Kumarasamy KK, Toleman MA, Walsh TR, Bagaria J, Butt F, Balakrishnan R,
1023 – 1029 Chaudhary U, Doumith M, Giske CG, Irfan S et al (2010) Emergence of a
Kahl BC, Becker K, Loffler B (2016) Clinical significance and pathogenesis of new antibiotic resistance mechanism in India, Pakistan, and the UK: a
staphylococcal small colony variants in persistent infections. Clin Microbiol molecular, biological, and epidemiological study. Lancet Infect Dis 10:
Rev 29: 401 – 427 597 – 602
Kamble Ekta PK (2020) Antibiotic tolerance in biofilm and stationary-phase Kwan BW, Chowdhury N, Wood TK (2015) Combatting bacterial infections by
planktonic cells of Staphylococcus aureus. Microb Drug Resist https://doi. killing persister cells with mitomycin C. Environ Microbiol 17: 4406 – 4414
org/10.1089/mdr.2019.0425 Kwan BW, Valenta JA, Benedik MJ, Wood TK (2013) Arrested protein synthesis
Kaspy I, Rotem E, Weiss N, Ronin I, Balaban NQ, Glaser G (2013) HipA- increases persister-like cell formation. Antimicrob Agents Chemother 57:
mediated antibiotic persistence via phosphorylation of the glutamyl-tRNA- 1468 – 1473
synthetase. Nat Commun 4: 3001 Lacey RW (1984) Mechanisms of resistance to beta-lactam antibiotics in
Katayama Y, Ito T, Hiramatsu K (2000) A new class of genetic element, Staphylococcus aureus. Scand J Infect Dis Suppl 42: 64 – 71
staphylococcus cassette chromosome mec, encodes methicillin resistance Lebeaux D, Ghigo JM, Beloin C (2014) Biofilm-related infections: bridging the
in Staphylococcus aureus. Antimicrob Agents Chemother 44: 1549 – 1555 gap between clinical management and fundamental aspects of
Kawano M, Aravind L, Storz G (2007) An antisense RNA controls synthesis of recalcitrance toward antibiotics. Microbiol Mol Biol Rev 78: 510 – 543
an SOS-induced toxin evolved from an antitoxin. Mol Microbiol 64: Lehar SM, Pillow T, Xu M, Staben L, Kajihara KK, Vandlen R, DePalatis L, Raab
738 – 754 H, Hazenbos WL, Morisaki JH et al (2015) Novel antibody-antibiotic
Keren I, Kaldalu N, Spoering A, Wang Y, Lewis K (2004) Persister cells and conjugate eliminates intracellular S. aureus. Nature 527: 323 – 328
tolerance to antimicrobials. FEMS Microbiol Lett 230: 13 – 18 Leid JG, Shirtliff ME, Costerton JW, Stoodley P (2002) Human leukocytes
Keyser P, Elofsson M, Rosell S, Wolf-Watz H (2008) Virulence blockers as adhere to, penetrate, and respond to Staphylococcus aureus biofilms. Infect
alternatives to antibiotics: type III secretion inhibitors against Gram- Immun 70: 6339 – 6345
negative bacteria. J Internal Med 264: 17 – 29 Leimer N, Rachmuhl C, Palheiros Marques M, Bahlmann AS, Furrer A,
Khatib R, Jose J, Musta A, Sharma M, Fakih MG, Johnson LB, Riederer K, Eichenseher F, Seidl K, Matt U, Loessner MJ, Schuepbach RA et al (2016)
Shemes S (2011) Relevance of vancomycin-intermediate susceptibility and Nonstable Staphylococcus aureus small-colony variants are induced by low
heteroresistance in methicillin-resistant Staphylococcus aureus pH and sensitized to antimicrobial therapy by phagolysosomal
bacteraemia. J Antimicrob Chemother 66: 1594 – 1599 alkalinization. J Infect Dis 213: 305 – 313
Leitner L, Sybesma W, Chanishvili N, Goderdzishvili M, Chkhotua A, Maranan MC, Moreira B, Boyle-Vavra S, Daum RS (1997) Antimicrobial
Ujmajuridze A, Schneider MP, Sartori A, Mehnert U, Bachmann LM et al resistance in staphylococci. Epidemiology, molecular mechanisms, and
(2017) Bacteriophages for treating urinary tract infections in patients clinical relevance. Infect Dis Clin North Am 11: 813 – 849
undergoing transurethral resection of the prostate: a randomized, Marques CNH, Morozov A, Planzos P, Zelaya HM (2014) The fatty acid
placebo-controlled, double-blind clinical trial. BMC Urol 17: 90 signaling molecule cis-2-decenoic acid increases metabolic activity and
LeRoux M, Culviner PH, Liu YJ, Littlehale ML, Laub MT (2020) Stress can reverts persister cells to an antimicrobial-susceptible state. Appl Environ
induce transcription of toxin-antitoxin systems without activating toxin. Microbiol 80: 6976 – 6991
Mol Cell 79: 280 – 292.e288 Masuda H, Tan Q, Awano N, Wu KP, Inouye M (2012) YeeU enhances the
Leung V, Levesque CM (2012) A stress-inducible quorum-sensing peptide bundling of cytoskeletal polymers of MreB and FtsZ, antagonizing the
mediates the formation of persister cells with noninherited multidrug CbtA (YeeV) toxicity in Escherichia coli. Mol Microbiol 84: 979 – 989
tolerance. J Bacteriol 194: 2265 – 2274 Matsuo M, Hiramatsu M, Singh M, Sasaki T, Hishinuma T, Yamamoto N,
Levin-Reisman I, Fridman O, Balaban NQ (2014) ScanLag: high-throughput Morimoto Y, Kirikae T, Hiramatsu K (2019) Genetic and transcriptomic
quantification of colony growth and lag time. J Vis Exp: 51456 analyses of ciprofloxacin-tolerant Staphylococcus aureus isolated by the
Levin-Reisman I, Gefen O, Fridman O, Ronin I, Shwa D, Sheftel H, Balaban NQ replica plating tolerance isolation system (REPTIS). Antimicrob Agents
(2010) Automated imaging with ScanLag reveals previously undetectable Chemother 63: e02019-18
bacterial growth phenotypes. Nat Methods 7: 737 – 739 Melo-Cristino J, Resina C, Manuel V, Lito L, Ramirez M (2013) First case of
Levin-Reisman I, Ronin I, Gefen O, Braniss I, Shoresh N, Balaban NQ (2017) infection with vancomycin-resistant Staphylococcus aureus in Europe.
Antibiotic tolerance facilitates the evolution of resistance. Science 355: Lancet 382: 205
826 – 830 Moker N, Dean CR, Tao J (2010) Pseudomonas aeruginosa increases formation
Lewis K (2007) Persister cells, dormancy and infectious disease. Nat Rev of multidrug-tolerant persister cells in response to quorum-sensing
Microbiol 5: 48 – 56 signaling molecules. J Bacteriol 192: 1946 – 1955
Lewis K (2012) Persister cells: molecular mechanisms related to antibiotic Montefour K, Frieden J, Hurst S, Helmich C, Headley D, Martin M, Boyle DA
tolerance. Handb Exp Pharmacol 121–133 (2008) Acinetobacter baumannii: an emerging multidrug-resistant
Lewis K (2019) Persister cells and infectious disease. Springer: Springer pathogen in critical care. Crit Care Nurse 28: 15 – 25; quiz 26
Li XZ, Nikaido H (2004) Efflux-mediated drug resistance in bacteria. Drugs 64: Moore MR, Perdreau-Remington F, Chambers HF (2003) Vancomycin
159 – 204 treatment failure associated with heterogeneous vancomycin-intermediate
Li Y, Powell DA, Shaffer SA, Rasko DA, Pelletier MR, Leszyk JD, Scott AJ, Staphylococcus aureus in a patient with endocarditis and in the rabbit
Masoudi A, Goodlett DR, Wang X et al (2012) LPS remodeling is an model of endocarditis. Antimicrob Agents Chemother 47: 1262 – 1266
evolved survival strategy for bacteria. Proc Natl Acad Sci USA 109: Moran GJ, Krishnadasan A, Gorwitz RJ, Fosheim GE, McDougal LK, Carey RB,
8716 – 8721 Talan DA (2006) Methicillin-resistant S. aureus infections among patients
Liu C, Bayer A, Cosgrove SE, Daum RS, Fridkin SK, Gorwitz RJ, Kaplan SL, in the emergency department. N Engl J Med 355: 666 – 674
Karchmer AW, Levine DP, Murray BE et al (2011) Clinical practice Moyed HS, Bertrand KP (1983) hipA, a newly recognized gene of Escherichia
guidelines by the infectious diseases society of america for the treatment coli K-12 that affects frequency of persistence after inhibition of murein
of methicillin-resistant Staphylococcus aureus infections in adults and synthesis. J Bacteriol 155: 768 – 775
children. Clin Infect Dis 52: e18 – 55 Mullard A (2016) FDA approves antitoxin antibody. Nat Rev Drug Discov 15: 811
Loc-Carrillo C, Abedon ST (2011) Pros and cons of phage therapy. Napier BA, Band V, Burd EM, Weiss DS (2014) Colistin heteroresistance in
Bacteriophage 1: 111 – 114 Enterobacter cloacae is associated with cross-resistance to the host
Lopez-Jacome E, Franco-Cendejas R, Quezada H, Morales-Espinosa R, Castillo- antimicrobial lysozyme. Antimicrob Agents Chemother 58: 5594 – 5597
Juarez I, Gonzalez-Pedrajo B, Fernandez-Presas AM, Tovar-Garcia A, Nataro JP, Blaser MJ, Cunningham-Rundles S (2000) Persistent bacterial
Angarita-Zapata V, Licona-Limon P et al (2019) The race between drug infections. American Society of Microbiology
introduction and appearance of microbial resistance. Current balance and Nguyen D, Joshi-Datar A, Lepine F, Bauerle E, Olakanmi O, Beer K, McKay G,
alternative approaches. Curr Opin Pharmacol 48: 48 – 56 Siehnel R, Schafhauser J, Wang Y et al (2011) Active starvation responses
Loss G, Simoes PM, Valour F, Cortes MF, Gonzaga L, Bergot M, Trouillet- mediate antibiotic tolerance in biofilms and nutrient-limited bacteria.
Assant S, Josse J, Diot A, Ricci E et al (2019) Staphylococcus aureus small Science 334: 982 – 986
colony variants (SCVs): news from a chronic prosthetic joint infection. Ni M, Decrulle AL, Fontaine F, Demarez A, Taddei F, Lindner AB (2012) Pre-
Front Cell Infect Microbiol 9: 363 disposition and epigenetics govern variation in bacterial survival upon
Lowy FD (1998) Staphylococcus aureus infections. N Engl J Med 339: stress. PLoS Genet 8: e1003148
520 – 532 Nicolas I, Bordeau V, Bondon A, Baudy-Floc’h M, Felden B (2019) Novel
Luther A, Urfer M, Zahn M, Muller M, Wang SY, Mondal M, Vitale A, antibiotics effective against gram-positive and -negative multi-resistant
Hartmann JB, Sharpe T, Monte FL et al (2019) Chimeric peptidomimetic bacteria with limited resistance. PLoS Biol 17: e3000337
antibiotics against Gram-negative bacteria. Nature 576: 452 – 458 Nicoloff H, Hjort K, Levin BR, Andersson DI (2019) The high prevalence of
Lv L, Wan M, Wang C, Gao X, Yang Q, Partridge SR, Wang Y, Zong Z, Doi Y, antibiotic heteroresistance in pathogenic bacteria is mainly caused by
Shen J et al (2020) Emergence of a plasmid-encoded resistance- gene amplification. Nat Microbiol 4: 504 – 514
nodulation-division efflux pump conferring resistance to multiple drugs, Norris SJ (2006) Antigenic variation with a twist–the Borrelia story. Mol
including tigecycline, in Klebsiella pneumoniae. mBio 11: e02930-19 Microbiol 60: 1319 – 1322
Manina G, Dhar N, McKinney JD (2015) Stress and host immunity amplify O’Neill J (2014) Antimicrobial resistance: tackling a crisis for the health
Mycobacterium tuberculosis phenotypic heterogeneity and induce and wealth of nations. https://amr-revieworg/sites/default/files/AMR%
nongrowing metabolically active forms. Cell Host Microbe 17: 32 – 46 20Review%20Paper%20-%20Tackling%20a%20crisis%20for%20the%20hea
lth%20and%20wealth%20of%20nations_1pdf (last accessed online Ram S, Goulian M (2013) The architecture of a prototypical bacterial
10122019) signaling circuit enables a single point mutation to confer novel network
Ooi N, Miller K, Randall C, Rhys-Williams W, Love W, Chopra I (2010) XF-70 properties. PLoS Genet 9: e1003706
and XF-73, novel antibacterial agents active against slow-growing and Rather IA, Kim BC, Bajpai VK, Park YH (2017) Self-medication and antibiotic
non-dividing cultures of Staphylococcus aureus including biofilms. J resistance: crisis, current challenges, and prevention. Saudi J Biol Sci 24:
Antimicrob Chemother 65: 72 – 78 808 – 812
Page R, Peti W (2016) Toxin-antitoxin systems in bacterial growth arrest and Redpath S, Ghazal P, Gascoigne NR (2001) Hijacking and exploitation of IL-10
persistence. Nat Chem Biol 12: 208 – 214 by intracellular pathogens. Trends Microbiol 9: 86 – 92
Painter KL, Strange E, Parkhill J, Bamford KB, Armstrong-James D, Edwards Reyes J, Aguilar AC, Caicedo A (2019) Carbapenem-resistant klebsiella
AM (2015) Staphylococcus aureus adapts to oxidative stress by producing pneumoniae: microbiology key points for clinical practice. Int J Gen Med
H2O2-resistant small-colony variants via the SOS. Response. 83: 12: 437 – 446
1830 – 1844 €m S, Normark SJ (2003) The basis of
Rhen M, Eriksson S, Clements M, Bergstro
Pandey DP, Gerdes K (2005) Toxin-antitoxin loci are highly abundant in free- persistent bacterial infections. Trends Microbiol 11: 80 – 86
living but lost from host-associated prokaryotes. Nucleic Acids Res 33: Rice LB (2010) Progress and challenges in implementing the research on
966 – 976 ESKAPE pathogens. Infect Control Hosp Epidemiol 31(Suppl 1): S7 – 10
Payne DJ, Gwynn MN, Holmes DJ, Pompliano DL (2007) Drugs for bad bugs: Rodrıguez-Rubio L, Chang W-L, Gutierrez D, Lavigne R, Martınez B, Rodrıguez
confronting the challenges of antibacterial discovery. Nat Rev Drug Discov A, Govers SK, Aertsen A, Hirl C, Biebl M et al (2016) ‘Artilysation’ of
6: 29 – 40 endolysin kSa2lys strongly improves its enzymatic and antibacterial
Pedersen K, Gerdes K (1999) Multiple hok genes on the chromosome of activity against streptococci. Sci Rep 6: 35382
Escherichia coli. Mol Microbiol 32: 1090 – 1102 €hrig C, Huemer M, Lorge D, Luterbacher S, Phothaworn P, Schefer C, Sobieraj
Ro
Pedersen MG, Olesen HV, Jensen-Fangel S, Nørskov-Lauritsen N, Wang M AM, Zinsli LV, Mairpady Shambat S, Leimer N et al (2020) Targeting hidden
(2018) Colistin resistance in Pseudomonas aeruginosa and pathogens: cell-penetrating enzybiotics eradicate intracellular drug-resistant
Achromobacter spp. cultured from Danish cystic fibrosis patients is not Staphylococcus aureus. mBio 11: e00209 – 00220
related to plasmid-mediated expression of mcr-1. J Cyst Fibros 17: Rolauffs B, Bernhardt TM, von Eiff C, Hart ML, Bettin D (2002) Osteopetrosis,
e22 – e23 femoral fracture, and chronic osteomyelitis caused by Staphylococcus
Pendleton JN, Gorman SP, Gilmore BF (2013) Clinical relevance of the ESKAPE aureus small colony variants (SCV) treated by girdlestone resection–6-year
pathogens. Expert Rev Anti Infect Ther 11: 297 – 308 follow-up. Arch Orthop Trauma Surg 122: 547 – 550
Pournaras S, Koumaki V, Spanakis N, Gennimata V, Tsakris A (2016) Current Ronneau S, Hallez R (2019) Make and break the alarmone: regulation of (p)
perspectives on tigecycline resistance in Enterobacteriaceae: susceptibility ppGpp synthetase/hydrolase enzymes in bacteria. FEMS Microbiol Rev 43:
testing issues and mechanisms of resistance. Int J Antimicrob Agents 48: 389 – 400
11 – 18 Ronneau S, Helaine S (2019) Clarifying the link between toxin-antitoxin
Proctor R (2019) Respiration and small colony variants of Staphylococcus modules and bacterial persistence. J Mol Biol 431: 3462 – 3471
aureus. Microbiol Spectr 7. https://doi.org/10.1128/microbiolspec.GPP3- Rowe SE, Wagner NJ, Li L, Beam JE, Wilkinson AD, Radlinski LC, Zhang Q,
0069-2019 Miao EA, Conlon BP (2020) Reactive oxygen species induce antibiotic
Proctor RA, van Langevelde P, Kristjansson M, Maslow JN, Arbeit RD (1995) tolerance during systemic Staphylococcus aureus infection. Nat Microbiol 5:
Persistent and relapsing infections associated with small-colony variants 282 – 290
of Staphylococcus aureus. Clin Infect Dis 20: 95 – 102 Salmond GP, Fineran PC (2015) A century of the phage: past, present and
Proctor RA, von Eiff C, Kahl BC, Becker K, McNamara P, Herrmann M, Peters future. Nat Rev Microbiol 13: 777 – 786
G (2006) Small colony variants: a pathogenic form of bacteria that Schaaff F, Bierbaum G, Baumert N, Bartmann P, Sahl HG (2003) Mutations
facilitates persistent and recurrent infections. Nat Rev Microbiol 4: are involved in emergence of aminoglycoside-induced small colony
295 – 305 variants of Staphylococcus aureus. Int J Med Microbiol 293: 427 – 435
Pu Y, Li Y, Jin X, Tian T, Ma Q, Zhao Z, Lin SY, Chen Z, Li B, Yao G et al (2019) Schilling NA, Berscheid A, Schumacher J, Saur JS, Konnerth MC, Wirtz SN,
ATP-dependent dynamic protein aggregation regulates bacterial dormancy Beltran-Belena JM, Zipperer A, Krismer B, Peschel A et al (2019) Synthetic
depth critical for antibiotic tolerance. Mol Cell 73: 143 – 156.e4 lugdunin analogues reveal essential structural motifs for antimicrobial
Pumbwe L, Piddock LJ (2000) Two efflux systems expressed simultaneously in action and proton translocation capability. Angew Chem Int Ed Engl 58:
multidrug-resistant Pseudomonas aeruginosa. Antimicrob Agents Chemother 9234 – 9238
44: 2861 – 2864 Schmelcher M, Donovan DM, Loessner MJ (2012) Bacteriophage endolysins as
Qin S, Wang Y, Zhang Q, Chen X, Shen Z, Deng F, Wu C, Shen J (2012) novel antimicrobials. Future Microbiol 7: 1147 – 1171
Identification of a novel genomic island conferring resistance to multiple Schneider M, Muhlemann K, Droz S, Couzinet S, Casaulta C, Zimmerli S
aminoglycoside antibiotics in Campylobacter coli. Antimicrob Agents (2008) Clinical characteristics associated with isolation of small-colony
Chemother 56: 5332 – 5339 variants of Staphylococcus aureus and Pseudomonas aeruginosa from
Queenan AM, Bush K (2007) Carbapenemases: the versatile beta-lactamases. respiratory secretions of patients with cystic fibrosis. J Clin Microbiol 46:
Clin Microbiol Rev 20: 440 – 458, table of contents. 1832 – 1834
Quie PG (1969) Microcolonies (G-variants) of Staphylococcus aureus. Yale J Biol Schreiber G, Ron EZ, Glaser G (1995) ppGpp-mediated regulation of DNA
Med 41: 394 – 403 replication and cell division in Escherichia coli. Curr Microbiol 30: 27 – 32
Ram G, Ross HF, Novick RP, Rodriguez-Pagan I, Jiang D (2018) Conversion of Schumacher MA, Piro KM, Xu W, Hansen S, Lewis K, Brennan RG (2009)
staphylococcal pathogenicity islands to CRISPR-carrying antibacterial Molecular mechanisms of HipA-mediated multidrug tolerance and its
agents that cure infections in mice. Nat Biotechnol 36: 971 – 976 neutralization by HipB. Science 323: 396 – 401
Schwaber MJ, Graham CS, Sands BE, Gold HS, Carmeli Y (2003) Treatment Tuchscherr L, Medina E, Hussain M, Volker W, Heitmann V, Niemann S,
with a broad-spectrum cephalosporin versus piperacillin-tazobactam and Holzinger D, Roth J, Proctor RA, Becker K et al (2011) Staphylococcus
the risk for isolation of broad-spectrum cephalosporin-resistant aureus phenotype switching: an effective bacterial strategy to escape host
Enterobacter species. Antimicrob Agents Chemother 47: 1882 – 1886 immune response and establish a chronic infection. EMBO Mol Med 3:
Seifert H, von Eiff C, Fatkenheuer G (1999) Fatal case due to methicillin- 129 – 141
resistant Staphylococcus aureus small colony variants in an AIDS patient. Turner NA, Sharma-Kuinkel BK, Maskarinec SA, Eichenberger EM, Shah PP,
Emerg Infect Dis 5: 450 – 453 Carugati M, Holland TL, Fowler Jr VG (2019) Methicillin-resistant
Shan Y, Brown Gandt A, Rowe SE, Deisinger JP, Conlon BP, Lewis K (2017) Staphylococcus aureus: an overview of basic and clinical research. Nat Rev
ATP-dependent persister formation in Escherichia coli. mBio 8: e02267-16 Microbiol 17: 203 – 218
Skinner D, Keefer CS (1941) significance of bacteremia caused by Turnidge J, Christiansen K (2005) Antibiotic use and resistance-proving the
Staphylococcus aureus: a study of one hundred and twenty-two cases and obvious. Lancet 365: 548 – 549
a review of the literature concerned with experimental infection in Twort FW (1915) An investigation on the nature of ultra-microscopic viruses.
animals. Arch Intern Med 68: 851 – 875 Lancet 186: 1241 – 1243
Southwood RT, Rice JL, McDonald PJ, Hakendorf PH, Rozenbilds MA (1985) Tzouvelekis LS, Markogiannakis A, Psichogiou M, Tassios PT, Daikos GL
Infection in experimental hip arthroplasties. J Bone Joint Surg Br 67: (2012) Carbapenemases in Klebsiella pneumoniae and other
229 – 231 Enterobacteriaceae: an evolving crisis of global dimensions. Clin Microbiol
Spellberg B, Bartlett JG, Gilbert DN (2013) The future of antibiotics and Rev 25: 682 – 707
resistance. N Engl J Med 368: 299 – 302 Unoson C, Wagner EG (2008) A small SOS-induced toxin is targeted against
Spoering AL, Lewis K (2001) Biofilms and planktonic cells of Pseudomonas the inner membrane in Escherichia coli. Mol Microbiol 70: 258 – 270
aeruginosa have similar resistance to killing by antimicrobials. J Bacteriol Van den Bergh B, Fauvart M, Michiels J (2017) Formation, physiology, ecology,
183: 6746 – 6751 evolution and clinical importance of bacterial persisters. FEMS Microbiol
Sreedharan S, Oram M, Jensen B, Peterson LR, Fisher LM (1990) DNA gyrase Rev 41: 219 – 251
gyrA mutations in ciprofloxacin-resistant strains of Staphylococcus aureus: Vazquez-Laslop N, Lee H, Neyfakh AA (2006) Increased persistence in
close similarity with quinolone resistance mutations in Escherichia coli. J Escherichia coli caused by controlled expression of toxins or other
Bacteriol 172: 7260 – 7262 unrelated proteins. J Bacteriol 188: 3494 – 3497
Stokes JM, Yang K, Swanson K, Jin W, Cubillos-Ruiz A, Donghia NM, MacNair Veening JW, Stewart EJ, Berngruber TW, Taddei F, Kuipers OP, Hamoen LW
CR, French S, Carfrae LA, Bloom-Ackerman Z et al (2020) A Deep Learning (2008) Bet-hedging and epigenetic inheritance in bacterial cell
Approach to Antibiotic Discovery. Cell 180(688–702): e613 development. Proc Natl Acad Sci USA 105: 4393 – 4398
Swingle EL (1935) Studies on small colony variants of Staphylococcus aureus. Vega NM, Allison KR, Khalil AS, Collins JJ (2012) Signaling-mediated bacterial
J Bacteriol 29: 467 – 489 persister formation. Nat Chem Biol 8: 431 – 433
Tacconelli E, Carrara E, Savoldi A, Harbarth S, Mendelson M, Monnet DL, Ventola CL (2015) The antibiotic resistance crisis: part 1: causes and threats.
Pulcini C, Kahlmeter G, Kluytmans J, Carmeli Y et al (2018) Discovery, P T 40: 277 – 283
research, and development of new antibiotics: the WHO priority list of Verstraeten N, Knapen WJ, Kint CI, Liebens V, Van den Bergh B, Dewachter L,
antibiotic-resistant bacteria and tuberculosis. Lancet Infect Dis 18: Michiels JE, Fu Q, David CC, Fierro AC et al (2015) Obg and membrane
318 – 327 depolarization are part of a microbial bet-hedging strategy that leads to
Tande AJ, Patel R (2014) Prosthetic joint infection. Clin Microbiol Rev 27: antibiotic tolerance. Mol Cell 59: 9 – 21
302 – 345 Vesga O, Groeschel MC, Otten MF, Brar DW, Vann JM, Proctor RA (1996)
Tanwar J, Das S, Fatima Z, Hameed S (2014) Multidrug resistance: an Staphylococcus aureus small colony variants are induced by the
emerging crisis. Interdiscip Perspect Infect Dis 2014: 541340 endothelial cell intracellular milieu. J Infect Dis 173: 739 – 742
Tenover FC, Goering RV (2009) Methicillin-resistant Staphylococcus aureus Vila J, Marti S, Sanchez-Cespedes J (2007) Porins, efflux pumps and multidrug
strain USA300: origin and epidemiology. J Antimicrob Chemother 64: resistance in Acinetobacter baumannii. J Antimicrob Chemother 59:
441 – 446 1210 – 1215
Thisted T, Sorensen NS, Wagner EG, Gerdes K (1994) Mechanism of post- Vogel J, Argaman L, Wagner EG, Altuvia S (2004) The small RNA IstR inhibits
segregational killing: Sok antisense RNA interacts with Hok mRNA via its synthesis of an SOS-induced toxic peptide. Curr Biol 14: 2271 – 2276
5’-end single-stranded leader and competes with the 3’-end of Hok mRNA von Eiff C, Bettin D, Proctor RA, Rolauffs B, Lindner N, Winkelmann W, Peters
for binding to the mok translational initiation region. EMBO J 13: G (1997a) Recovery of small colony variants of Staphylococcus aureus
1960 – 1968 following gentamicin bead placement for osteomyelitis. Clin Infect Dis 25:
Thomsen IP, Kadari P, Soper NR, Riddell S, Kiska D, Creech CB, Shaw J 1250 – 1251
(2019) Molecular epidemiology of invasive Staphylococcus aureus von Eiff C, Heilmann C, Proctor RA, Woltz C, Peters G, Gotz F (1997b) A site-
infections and concordance with colonization isolates. J Pediatr 210: directed Staphylococcus aureus hemB mutant is a small-colony variant
173 – 177 which persists intracellularly. J Bacteriol 179: 4706 – 4712
Tranchemontagne ZR, Camire RB, O’Donnell VJ, Baugh J, Burkholder KM von Eiff C, Vaudaux P, Kahl BC, Lew D, Emler S, Schmidt A, Peters G, Proctor
(2016) Staphylococcus aureus strain USA300 perturbs acquisition of RA (1999) Bloodstream infections caused by small-colony variants of
lysosomal enzymes and requires phagosomal acidification for survival coagulase-negative staphylococci following pacemaker implantation. Clin
inside macrophages. Infect Immun 84: 241 – 253 Infect Dis 29: 932 – 934
Traxler MF, Summers SM, Nguyen HT, Zacharia VM, Hightower GA, Smith JT, Vulin C, Leimer N, Huemer M, Ackermann M, Zinkernagel AS (2018) Prolonged
Conway T (2008) The global, ppGpp-mediated stringent response to amino bacterial lag time results in small colony variants that represent a sub-
acid starvation in Escherichia coli. Mol Microbiol 68: 1128 – 1148 population of persisters. Nat Commun 9: 4074
Vuong C, Voyich JM, Fischer ER, Braughton KR, Whitney AR, DeLeo FR, Otto M Wozniak A, Villagra NA, Undabarrena A, Gallardo N, Keller N, Moraga M,
(2004) Polysaccharide intercellular adhesin (PIA) protects Staphylococcus Roman JC, Mora GC, Garcia P (2012) Porin alterations present in non-
epidermidis against major components of the human innate immune carbapenemase-producing Enterobacteriaceae with high and
system. Cell Microbiol 6: 269 – 275 intermediate levels of carbapenem resistance in Chile. J Med Microbiol
Wang Q, Chang C-s, Pennini M, Pelletier M, Rajan S, Zha J, Chen Y, Cvitkovic 61: 1270 – 1279
R, Sadowska A, Heidbrink Thompson J et al (2016) Target-agnostic Wu D, Wang Q, Yang Y, Geng W, Wang Q, Yu S, Yao K, Yuan L, Shen X (2010)
identification of functional monoclonal antibodies against Klebsiella Epidemiology and molecular characteristics of community-associated
pneumoniae multimeric MrkA fimbrial subunit. J Infect Dis 213: 1800 – 1808 methicillin-resistant and methicillin-susceptible Staphylococcus aureus
Wang X, Lord DM, Cheng HY, Osbourne DO, Hong SH, Sanchez-Torres V, from skin/soft tissue infections in a children’s hospital in Beijing, China.
Quiroga C, Zheng K, Herrmann T, Peti W et al (2012) A new type V toxin- Diagn Microbiol Infect Dis 67: 1 – 8
antitoxin system where mRNA for toxin GhoT is cleaved by antitoxin Wu Y, Vulic M, Keren I, Lewis K (2012) Role of oxidative stress in persister
GhoS. Nat Chem Biol 8: 855 – 861 tolerance. Antimicrob Agents Chemother 56: 4922 – 4926
Wexselblatt E, Oppenheimer-Shaanan Y, Kaspy I, London N, Schueler-Furman Yong D, Toleman MA, Giske CG, Cho HS, Sundman K, Lee K, Walsh TR (2009)
O, Yavin E, Glaser G, Katzhendler J, Ben-Yehuda S (2012) Relacin, a novel Characterization of a new metallo-beta-lactamase gene, bla(NDM-1), and
antibacterial agent targeting the Stringent Response. PLoS Pathog 8: a novel erythromycin esterase gene carried on a unique genetic structure
e1002925 in Klebsiella pneumoniae sequence type 14 from India. Antimicrob Agents
WHO (2017) Prioritization of pathogens to guide discovery, research and Chemother 53: 5046 – 5054
development of new antibiotics for drug-resistant bacterial infections, Yoshida H, Bogaki M, Nakamura M, Nakamura S (1990) Quinolone resistance-
including tuberculosis. World Health Organization determining region in the DNA gyrase gyrA gene of Escherichia coli.
Wilmaerts D, Windels EM, Verstraeten N, Michiels J (2019) General Antimicrob Agents Chemother 34: 1271 – 1272
mechanisms leading to persister formation and awakening. Trends Genet Zaman SB, Hussain MA, Nye R, Mehta V, Mamun KT, Hossain N (2017) A
35: 401 – 411 review on antibiotic resistance: alarm bells are ringing. Cureus 9: e1403
Windels EM, Michiels JE, Fauvart M, Wenseleers T, Van den Bergh B, Michiels Zgurskaya HI, Lopez CA, Gnanakaran S (2015) Permeability barrier of gram-
J (2019) Bacterial persistence promotes the evolution of antibiotic negative cell envelopes and approaches to bypass it. ACS Infect Dis 1:
resistance by increasing survival and mutation rates. ISME J 13: 512 – 522
1239 – 1251 Zhu L, Lin J, Ma J, Cronan JE, Wang H (2010) Triclosan resistance of
Wolfson JS, Hooper DC, McHugh GL, Bozza MA, Swartz MN (1990) Mutants of Pseudomonas aeruginosa PAO1 is due to FabV, a triclosan-resistant enoyl-
Escherichia coli K-12 exhibiting reduced killing by both quinolone and acyl carrier protein reductase. Antimicrob Agents Chemother 54: 689 – 698
beta-lactam antimicrobial agents. Antimicrob Agents Chemother 34: Zipperer A, Konnerth MC, Laux C, Berscheid A, Janek D, Weidenmaier C,
1938 – 1943 Burian M, Schilling NA, Slavetinsky C, Marschal M et al (2016) Human
WorldEconomicForum (2019) The Global Risks Report 2019. http://www3.wef commensals producing a novel antibiotic impair pathogen colonization.
orum.org/docs/WEF_Global_Risks_Report_2019.pdf Nature 535: 511 – 516