Abstract

Staphylococcus aureus is a major human pathogen causing a vast array of infections with significant mortality. Its versatile physiology enables it to adapt to various environments. Specific physiological changes are thought to underlie the frequent failure of antimicrobial therapy despite susceptibility in standard microbiological assays. Bacteria capable of surviving high antibiotic concentrations despite having a genetically susceptible background are described as ‘antibiotic tolerant’. In this review, we put current knowledge on environmental triggers and molecular mechanisms of increased antibiotic survival of S. aureus into its clinical context. We discuss animal and clinical evidence of its significance and outline strategies to overcome infections with antibiotic-tolerant S. aureus.

Introduction

Staphylococcus aureus is one of the most versatile pathogens infecting humans. It harmlessly colonizes one-third of the human population in the nares or throat or on the skin,1 but can also lead to a vast array of potentially lethal invasive infections. S. aureus can infect virtually any tissue of the body such as skin, muscles, bones and joints, lungs, kidneys, heart or even the eyes or brain.2,3 Moreover, it readily grows on artificial surfaces of implanted foreign bodies such as orthopaedic devices, prosthetic heart valves or intravascular and urinary catheters. More than three decades ago, S. aureus was named the ‘persistent pathogen’4 due to its unusual recalcitrance towards antibiotic treatment. This has remained, to date, a major clinical challenge. S. aureus can persist for several days in the blood of up to one-third of patients despite correct antibiotic treatment;5–9 this is by far more common than with other bacteria.8,10 The associated mortality rate of 15%–50% leads to a calculated annual 2–10 deaths/100 000 population.11 Finally, current antibiotics have limited capacity to eradicate S. aureus in abscesses or in implant infections without the support of surgery.11–14 Physicians choose antibiotics according to a strain’s susceptibility measured as the MIC in the microbiology laboratory. When treatment nevertheless fails, this can have several clinical or pharmacological reasons (Table 1), many of which are preventable through optimized application of the antibiotics. However, evidence is emerging that the ability of S. aureus to become antibiotic tolerant represents a major microbiological reason for the high rate of treatment failure.

Table 1.

Reasons for treatment failure despite correct susceptibility-guided antibiotic therapy of the infecting S. aureus measured by the MIC

Delayed initiation of antibiotic treatment
Low serum concentration of antibiotic
 low oral bioavailability
 long interval between dosing
 augmented hepatic or renal clearance
 dosage not adjusted to body weight
 non-compliance of the patient
Insufficient antibiotic penetration to the site of infection
 infections of niches such as bone or cerebrospinal fluid
 intracellular infections
 infection of necrotic tissue
Physicochemical inactivation of antibiotics at the site of infection
 low pH
 interaction with debris
 interaction with biofilm matrix
Heteroresistant bacterial subpopulation
Bacterial tolerance to the applied antibiotic(s)
Delayed initiation of antibiotic treatment
Low serum concentration of antibiotic
 low oral bioavailability
 long interval between dosing
 augmented hepatic or renal clearance
 dosage not adjusted to body weight
 non-compliance of the patient
Insufficient antibiotic penetration to the site of infection
 infections of niches such as bone or cerebrospinal fluid
 intracellular infections
 infection of necrotic tissue
Physicochemical inactivation of antibiotics at the site of infection
 low pH
 interaction with debris
 interaction with biofilm matrix
Heteroresistant bacterial subpopulation
Bacterial tolerance to the applied antibiotic(s)
Table 1.

Reasons for treatment failure despite correct susceptibility-guided antibiotic therapy of the infecting S. aureus measured by the MIC

Delayed initiation of antibiotic treatment
Low serum concentration of antibiotic
 low oral bioavailability
 long interval between dosing
 augmented hepatic or renal clearance
 dosage not adjusted to body weight
 non-compliance of the patient
Insufficient antibiotic penetration to the site of infection
 infections of niches such as bone or cerebrospinal fluid
 intracellular infections
 infection of necrotic tissue
Physicochemical inactivation of antibiotics at the site of infection
 low pH
 interaction with debris
 interaction with biofilm matrix
Heteroresistant bacterial subpopulation
Bacterial tolerance to the applied antibiotic(s)
Delayed initiation of antibiotic treatment
Low serum concentration of antibiotic
 low oral bioavailability
 long interval between dosing
 augmented hepatic or renal clearance
 dosage not adjusted to body weight
 non-compliance of the patient
Insufficient antibiotic penetration to the site of infection
 infections of niches such as bone or cerebrospinal fluid
 intracellular infections
 infection of necrotic tissue
Physicochemical inactivation of antibiotics at the site of infection
 low pH
 interaction with debris
 interaction with biofilm matrix
Heteroresistant bacterial subpopulation
Bacterial tolerance to the applied antibiotic(s)

Earlier studies arbitrarily defined antibiotic tolerance as in vitro measurement of a ratio of MBC to MIC of ≥32. This definition, however, does not encompass the various populations and mechanisms of tolerance and its measurement is fraught with considerable variability.15,16 Therefore, more recently, leading experts have provided clarification about the terminology defining the effect of antibiotics on bacterial growth and survival (for details, see Balaban et al.17). Accordingly, tolerance describes the bacterial ability to survive usually lethal concentrations of a bactericidal antibiotic without change of the MIC.17,18 This can classically be visualized and measured in a slower killing rate in in vitro time–kill curves. However, so far, no new classification or measurable cut-offs have been introduced that may be applicable to clinical practice. From a population-level perspective, the term tolerance applies if an entire bacterial population shows increased survival whereas the term persistence is used if only a small fraction of a population (named persisters) exhibit a tolerant phenotype. Although the clinical relevance of this differentiation is not clear, we will use the term ‘increased antibiotic survival’19,20 to encompass both tolerance and persistence.

In this review, we put mechanisms and triggers of increased antibiotic survival into the context of various types of infection and life forms of S. aureus. By taking a broad clinical perspective, we extend the scope of previous reviews on tolerance and persistence in general17,18,21 as well as on S. aureus in particular.22,23 With the goal of translating basic research to clinical practice, we provide an overview of highly discussed as well as rather neglected studies of in vitro, in vivo and clinical aspects of antibiotic-tolerant S. aureus. We discuss approaches of how to overcome infections with antibiotic-tolerant S. aureus and identify future research questions and open gaps in knowledge.

Environmental triggers of increased antibiotic survival of S. aureus

Development of tolerant phenotypes can be interpreted as a survival strategy of a bacterial population in quickly changing and potentially toxic environments.24 Tolerant phenotypes can develop due to genetic traits,25–29 can evolve stochastically30,31 or can be triggered by specific environmental factors18 in both Gram-positive and Gram-negative bacteria. The majority of studies on increased antibiotic survival have focused on Gram-negative bacteria.32 Basic mechanisms of becoming tolerant are probably common for both Gram-positive and Gram-negative bacteria.20,21,33,34 However, not only do the identified molecular targets leading to tolerant phenotypes differ but S. aureus also stands out through the variety of evolved life forms causing infections associated with characteristics of increased antibiotic survival. The scientific pioneer Bigger had already noticed by 1944 that growth-inhibiting conditions induce increased antibiotic survival in S. aureus.35 Renewed research on the topic has now confirmed these observations and found several environmental factors such as low oxygen level,36 low pH37 or low amount of nutrients38 as responsible for triggering tolerant phenotypes (Figure 1).

Examples of environmental triggers shown to increase S. aureus antibiotic survival. The insets show examples of situations associated with these triggers and with increased antibiotic survival. This figure appears in colour in the online version of JAC and in black and white in the print version of JAC.
Figure 1.

Examples of environmental triggers shown to increase S. aureus antibiotic survival. The insets show examples of situations associated with these triggers and with increased antibiotic survival. This figure appears in colour in the online version of JAC and in black and white in the print version of JAC.

Most mechanistic in vitro studies have focused on the free-floating planktonic single-cell life form of S. aureus in liquid culture. High planktonic population density is typically strongly associated with development of increased antibiotic survival. This is due to the exhaustion of growth-sustaining factors and accumulation of metabolic waste products or bacterial signalling molecules.39 However, it is not known which role such tolerant planktonic forms of S. aureus play in the clinical context. Single cells or small aggregates of S. aureus dividing in the interstitial space are most likely responsible for several acute infections such as pneumonia or skin and soft tissue infections. Short antibiotic regimens generally cure such infections12 and recurrence is uncommon. Hence, even if tolerant planktonic subpopulations survive the exposure to antibiotic treatment in these acute infections, they presumably do not withstand killing by the immune system in the unprotected environment of the interstitial space.

S. aureus increases its survival in vivo through the formation of multicellular conglomerates. Indeed, aggregating into cell clusters seems to be an intrinsic growth pattern of S. aureus,41 a phenotype constantly observed since its first discovery in the 1880s and serving as inspiration for its microbiological name (Greek: σταφνλοσ meaning grape cluster).42 The tendency to cluster dramatically increases in the presence of fibrinogen/fibronectin, which is abundant in synovial fluid of joints (e.g. in septic arthritis) or in intravascular fibrin and platelet clots (e.g. in endocarditis).43 These cell clusters can survive antibiotic exposures that would usually be bactericidal even without attachment to a surface,43,44 requiring prolonged antibiotic treatment to cure the respective infections. Inside a tissue, staphylococcal cell clusters can evolve into abscesses by digesting and transforming the surrounding tissue into a protective capsule of fibrin and necrotic debris.45 Antibiotics in the absence of surgery generally fail to eradicate S. aureus in abscesses.46 This has been attributed to reduced antibiotic penetration and physicochemical or enzymatic inactivation.47 However, since several antibiotics can achieve adequate concentrations inside an abscess,47 tolerant S. aureus in the acidic and low-oxygen environment of an abscess may play an important role in their recalcitrance against antibiotics. Concordantly, tolerant subpopulations have been isolated from abscesses manifesting as non-stable small colony variants (SCVs) with extended lag periods before regrowth.48

S. aureus can further increase its survival by forming multicellular biofilms encased by a self-produced matrix on organic or inorganic surfaces.49,50 Prototypical examples of such biofilm infections include endocarditis or implant-related infections.51,52 These are clinically relevant due to their ability to survive more than 100–1000-fold higher antibiotic concentrations compared with planktonic S. aureus.53–56 In such cases, increased survival cannot be explained by lack of biofilm penetration since even large antibiotics such as vancomycin,57,58 daptomycin59 or telavancin60 easily penetrate S. aureus biofilms. Gradients of oxygen and nutrients inside staphylococcal biofilms lead to spatial patterns of diverse physiological states with different levels of cellular respiration, DNA replication and protein synthesis61 and therefore potentially diverse levels of increased survival. Non-growing, tolerant fractions of biofilm bacteria are thought to be responsible for the prolonged survival of biofilms during exposure to bactericidal antibiotics requiring extended treatment and responsible for relapses once treatment ends.62 The number of tolerant cells in a biofilm increases with age and cell density,63,64 supporting the clinical concept that after a certain time of biofilm maturation, prosthetic implants need to be surgically removed to achieve cure.65,66 Moreover, biofilm-derived detached cell clusters exhibit a higher level of antibiotic survival than purely single-cell planktonic bacteria67,68 and may play a major role in perpetuating and spreading an infection.

With its versatile physiology, S. aureus can also survive by invading professional and non-professional phagocytic cells.69–73 This may lead to treatment failure74 and even the spread of infection through haematogenous transport inside host cells.75 Intracellular S. aureus has been documented in skin and soft tissue, bone and upper airway infections72,76,77 and is probably involved in an even wider spectrum of chronic and recurrent infections.58,75,78 The intracellular compartment is a protected niche since it shields S. aureus from the immune system and from the high extracellular antibiotic drug concentrations.75,79,80 However, the specific metabolic state of intracellular S. aureus further reduces the lethality of cell-penetrating antibiotics.81,82,S. aureus from intracellular locations can manifest on the laboratory growth plate as an SCV due to its slowed or delayed growth and hence eponymous reduced colony size.83–85 Compared with normal phenotypes, SCVs not only show reduced metabolism but also reduced expression of regulator and virulence factors.84 SCVs from clinical samples often present as so-called ‘non-stable’ or ‘dynamic’ SCVs, since they rapidly revert to normal colony sizes and metabolism after reculture on rich media.86,87 SCVs have been found to be tolerant to aminoglycosides and cell-wall active antibiotics88,89 and are associated with recalcitrant and relapsing infections.90,91 Nutrient starvation, oxidative stress and low pH are characteristics of the encountered intracellular milieu and all induce the formation of SCVs.85,92,93 Concordantly, alkalinization of the phagolysosome by the antimalarial drug chloroquine reduced survival in antibiotic treatment of intracellular S. aureus.93 Finally, exposure to host defence mechanisms can stimulate SCVs and other tolerant phenotypes in S. aureus during an infection.85,94

Taken together, S. aureus has developed various life forms to persist in different niches of the human body and thereby encounters or produces environmental triggers of increased survival, rendering an antibiotic treatment ineffective.

In vitro evidence for mechanisms of increased antibiotic survival of S. aureus

Understanding the mechanisms of antibiotic survival is essential in order to improve therapeutic strategies against S. aureus infections. Reduced cellular metabolism leading to decreased growth rate or dormancy seems to be a shared feature of most tolerant bacteria.18,30,95,96 This may not just be a passive state but may involve active metabolic changes focusing on adaptation to a hostile environment.24,97–99 Various molecular targets have been found to change the level of antibiotic survival in S. aureus (Table 2). Individual targets may only induce survival to some antibiotics tested whereas other antibiotics may stay effective (Table 2). However, separate studies have found some conflicting results, which likely reflect the complexity of mechanisms and the sensitivity of its regulation. Results may differ greatly depending on the genetic background of the individual strains and on the experimental setup.17 The growth phase strongly influences the level of antibiotic survival since slow- or non-growing S. aureus from high-density stationary-phase cultures are considerably more tolerant to antibiotics than when in exponential growth.30,37,100–104 A low intracellular level of ATP may represent a mechanism of antibiotic survival for S. aureus30 as seen in other bacteria.105 The reduced cellular fuel in the form of ATP slows down or halts cellular processes targeted by antibiotics and thereby protects the bacterial cell from their bactericidal action. Nevertheless, low ATP levels seem not to be a prerequisite, since increased antibiotic survival has been found in S. aureus with normal ATP levels.106,107 Moreover, antibiotic survival may not always be associated with decreased growth according to a study that found a higher rate of cells tolerant to vancomycin in a more rapidly growing WT S. aureus compared with its mutant with defects in purine synthesis.108

Table 2.

Targets and mechanisms associated with increased antibiotic survival of S. aureus

Target associated with increased antibiotic survival and main known functionPresumable or associated mechanism(s) of increased antibiotic survivalChange of antibiotic survival shown for these antibioticsNo change of antibiotic survival found for these antibioticsComments
Increased antibiotic survival if amount or function of target decreased

  • agr

  • → global regulator of virulence factors

dysfunction of agr
  • decreases secreted phenol soluble modulins

  • increases detoxification of ROS by derepression of glutathione peroxidase BsaA

  • increases agr-repressed secreted factors interfering with extracellular antibiotics

  • oxacillin130

  • daptomycin130 (opposite effect)

  • strain-dependent effect

  • growth-phase-dependent effect

  • agr deficiency clinically associated with persistent infection

  • contradicting studies for daptomycin

  • ATP

    • → main cellular energy substrate

low level of ATP decreases activity of antibiotic targets and downstream bactericidal mechanism
  • ciprofloxacin30,31

  • oxacillin31

  • gentamicin31

not reported
  • may be a common trait of tolerant S. aureus; however, not prerequisite

  • GdpP

    • → membrane-located ci-d-AMP-phosphodiesterase

inactivation of GdpP
  • leads to increased intracellular second messenger ci-d-AMP 229

  • followed by more biofilm formation

  • ileS

  • → encodes isoleucine tRNA synthase

dysfunction of ileS
  • decreases protein synthesis through change of tRNA binding site

  • vancomycin28

not reported
  • murein hydrolases

    • → cleave components of cell wall

  • → mediators of autolysis

decreased activity of hydrolases
  • decreases antibiotic-induced autolysis

  • can be mediated by decreased activity of their regulators (e.g. cidABC and lrgAB)

  • may be mediated through vancomycin-induced inhibition of access to their targets

not reported
  • addition of glucose can increase activity of murein regulators CidABC/LrgA and decrease tolerance

  • pitA

    • → encodes inorganic phosphate transporter

point mutation in pitA leads to
  • intracellular accumulation of inorganic phosphate, which up-regulates the dlt operon

  • increased cell wall thickness

  • daptomycin ± ampicillin/gentamicin/vancomycin/ciprofloxacin/rifampicin127,128

  • ADEP4 ± rifampicin/daptomycin127,128

not reported
  • TCA cycle

  • → main metabolic pathway to transfer stored energy into ATP and NADH

inactivation by mutations of selected TCA enzymes
  • reduces membrane potential without change of intracellular ATP levels stochastic fluctuation of TCA cycle expression

  • produces tolerant subpopulation with low intracellular ATP levels

  • contradicting studies regarding ATP level after TCA-cycle inhibition

  • Dsp1

    • → hypothetical lipoprotein

  • Asp1

    • → alkaline shock protein

deletion mutation of dsp1 or asp1
  • increased cell wall thickness

  • reduced autolysis

not reported
Increased antibiotic survival if amount or function of target increased

  • ppGpp

    • → nucleotides mediating the stringent response

  • RelA

  • → enzyme producing ppGpp

ppGpp-activated stringent response
  • results in global physiological changes and inactivation of antibiotic targets

  • gentamicin30

  • ciprofloxacin30

  • induces SCVs and promotes intraphagocyte survival

  • contradicting studies for ciprofloxacin

  • Saturated fatty acid in membrane

  • → influences membrane fluidity

increased amount of saturated fatty acid
  • reduces oligomerization and pore formation of daptomycin

not reported
  • growth phase- and strain-dependent effect on tolerance

  • effect dependent on the distribution of fatty acids in the growth medium


Reduced antibiotic survival if amount or function of target decreased

  • Clp

    • → important protease involved in stress response and virulence

  • → degrades antitoxins

lack of Clp
  • increases levels of antitoxin

  • decreases bacterial response to antibiotic stress

  • cfu-dependent/growth phase-dependent effect

  • MazF/MazE

  • → toxin/antitoxin system

presumable mechanism not known
  • oxacillin30

  • vancomycin30

  • ciprofloxacin30

  • rifampicin30

  • gentamicin30

  • associated with growth inhibition and SCVs

  • contradicting studies for β-lactams and vancomycin

  • msaABCR

  • → regulator involved in virulence and resistance

presumable mechanism not knownplanktonic exponential232
  • rifampicin

  • gentamicin

  • vancomycin

  • daptomycin

planktonic exponential232
  • linezolid

  • large variability according to growth phase

planktonic stationary232
  • linezolid

  • gentamicin

planktonic stationary232
  • daptomycin

  • vancomycin

  • rifampicin

biofilm232
  • daptomycin

  • vancomycin

  • linezolid

biofilm232
  • gentamicin

  • rifampicin

  • superoxide dismutase

  • → enzyme detoxifying ROS

lack of superoxide dismutase
  • increases antibiotic-induced ROS damaging DNA deletion of purF leads to

not reported
  • purF

  • → encodes enzyme for purine biosynthesis

  • higher ATP levels allowing bactericidal action of antibiotic

not reported
  • faster growth rate of tolerant strain compared with less tolerant ΔpurF mutant

Target associated with increased antibiotic survival and main known functionPresumable or associated mechanism(s) of increased antibiotic survivalChange of antibiotic survival shown for these antibioticsNo change of antibiotic survival found for these antibioticsComments
Increased antibiotic survival if amount or function of target decreased

  • agr

  • → global regulator of virulence factors

dysfunction of agr
  • decreases secreted phenol soluble modulins

  • increases detoxification of ROS by derepression of glutathione peroxidase BsaA

  • increases agr-repressed secreted factors interfering with extracellular antibiotics

  • oxacillin130

  • daptomycin130 (opposite effect)

  • strain-dependent effect

  • growth-phase-dependent effect

  • agr deficiency clinically associated with persistent infection

  • contradicting studies for daptomycin

  • ATP

    • → main cellular energy substrate

low level of ATP decreases activity of antibiotic targets and downstream bactericidal mechanism
  • ciprofloxacin30,31

  • oxacillin31

  • gentamicin31

not reported
  • may be a common trait of tolerant S. aureus; however, not prerequisite

  • GdpP

    • → membrane-located ci-d-AMP-phosphodiesterase

inactivation of GdpP
  • leads to increased intracellular second messenger ci-d-AMP 229

  • followed by more biofilm formation

  • ileS

  • → encodes isoleucine tRNA synthase

dysfunction of ileS
  • decreases protein synthesis through change of tRNA binding site

  • vancomycin28

not reported
  • murein hydrolases

    • → cleave components of cell wall

  • → mediators of autolysis

decreased activity of hydrolases
  • decreases antibiotic-induced autolysis

  • can be mediated by decreased activity of their regulators (e.g. cidABC and lrgAB)

  • may be mediated through vancomycin-induced inhibition of access to their targets

not reported
  • addition of glucose can increase activity of murein regulators CidABC/LrgA and decrease tolerance

  • pitA

    • → encodes inorganic phosphate transporter

point mutation in pitA leads to
  • intracellular accumulation of inorganic phosphate, which up-regulates the dlt operon

  • increased cell wall thickness

  • daptomycin ± ampicillin/gentamicin/vancomycin/ciprofloxacin/rifampicin127,128

  • ADEP4 ± rifampicin/daptomycin127,128

not reported
  • TCA cycle

  • → main metabolic pathway to transfer stored energy into ATP and NADH

inactivation by mutations of selected TCA enzymes
  • reduces membrane potential without change of intracellular ATP levels stochastic fluctuation of TCA cycle expression

  • produces tolerant subpopulation with low intracellular ATP levels

  • contradicting studies regarding ATP level after TCA-cycle inhibition

  • Dsp1

    • → hypothetical lipoprotein

  • Asp1

    • → alkaline shock protein

deletion mutation of dsp1 or asp1
  • increased cell wall thickness

  • reduced autolysis

not reported
Increased antibiotic survival if amount or function of target increased

  • ppGpp

    • → nucleotides mediating the stringent response

  • RelA

  • → enzyme producing ppGpp

ppGpp-activated stringent response
  • results in global physiological changes and inactivation of antibiotic targets

  • gentamicin30

  • ciprofloxacin30

  • induces SCVs and promotes intraphagocyte survival

  • contradicting studies for ciprofloxacin

  • Saturated fatty acid in membrane

  • → influences membrane fluidity

increased amount of saturated fatty acid
  • reduces oligomerization and pore formation of daptomycin

not reported
  • growth phase- and strain-dependent effect on tolerance

  • effect dependent on the distribution of fatty acids in the growth medium


Reduced antibiotic survival if amount or function of target decreased

  • Clp

    • → important protease involved in stress response and virulence

  • → degrades antitoxins

lack of Clp
  • increases levels of antitoxin

  • decreases bacterial response to antibiotic stress

  • cfu-dependent/growth phase-dependent effect

  • MazF/MazE

  • → toxin/antitoxin system

presumable mechanism not known
  • oxacillin30

  • vancomycin30

  • ciprofloxacin30

  • rifampicin30

  • gentamicin30

  • associated with growth inhibition and SCVs

  • contradicting studies for β-lactams and vancomycin

  • msaABCR

  • → regulator involved in virulence and resistance

presumable mechanism not knownplanktonic exponential232
  • rifampicin

  • gentamicin

  • vancomycin

  • daptomycin

planktonic exponential232
  • linezolid

  • large variability according to growth phase

planktonic stationary232
  • linezolid

  • gentamicin

planktonic stationary232
  • daptomycin

  • vancomycin

  • rifampicin

biofilm232
  • daptomycin

  • vancomycin

  • linezolid

biofilm232
  • gentamicin

  • rifampicin

  • superoxide dismutase

  • → enzyme detoxifying ROS

lack of superoxide dismutase
  • increases antibiotic-induced ROS damaging DNA deletion of purF leads to

not reported
  • purF

  • → encodes enzyme for purine biosynthesis

  • higher ATP levels allowing bactericidal action of antibiotic

not reported
  • faster growth rate of tolerant strain compared with less tolerant ΔpurF mutant

ROS, reactive oxygen species.

Table 2.

Targets and mechanisms associated with increased antibiotic survival of S. aureus

Target associated with increased antibiotic survival and main known functionPresumable or associated mechanism(s) of increased antibiotic survivalChange of antibiotic survival shown for these antibioticsNo change of antibiotic survival found for these antibioticsComments
Increased antibiotic survival if amount or function of target decreased

  • agr

  • → global regulator of virulence factors

dysfunction of agr
  • decreases secreted phenol soluble modulins

  • increases detoxification of ROS by derepression of glutathione peroxidase BsaA

  • increases agr-repressed secreted factors interfering with extracellular antibiotics

  • oxacillin130

  • daptomycin130 (opposite effect)

  • strain-dependent effect

  • growth-phase-dependent effect

  • agr deficiency clinically associated with persistent infection

  • contradicting studies for daptomycin

  • ATP

    • → main cellular energy substrate

low level of ATP decreases activity of antibiotic targets and downstream bactericidal mechanism
  • ciprofloxacin30,31

  • oxacillin31

  • gentamicin31

not reported
  • may be a common trait of tolerant S. aureus; however, not prerequisite

  • GdpP

    • → membrane-located ci-d-AMP-phosphodiesterase

inactivation of GdpP
  • leads to increased intracellular second messenger ci-d-AMP 229

  • followed by more biofilm formation

  • ileS

  • → encodes isoleucine tRNA synthase

dysfunction of ileS
  • decreases protein synthesis through change of tRNA binding site

  • vancomycin28

not reported
  • murein hydrolases

    • → cleave components of cell wall

  • → mediators of autolysis

decreased activity of hydrolases
  • decreases antibiotic-induced autolysis

  • can be mediated by decreased activity of their regulators (e.g. cidABC and lrgAB)

  • may be mediated through vancomycin-induced inhibition of access to their targets

not reported
  • addition of glucose can increase activity of murein regulators CidABC/LrgA and decrease tolerance

  • pitA

    • → encodes inorganic phosphate transporter

point mutation in pitA leads to
  • intracellular accumulation of inorganic phosphate, which up-regulates the dlt operon

  • increased cell wall thickness

  • daptomycin ± ampicillin/gentamicin/vancomycin/ciprofloxacin/rifampicin127,128

  • ADEP4 ± rifampicin/daptomycin127,128

not reported
  • TCA cycle

  • → main metabolic pathway to transfer stored energy into ATP and NADH

inactivation by mutations of selected TCA enzymes
  • reduces membrane potential without change of intracellular ATP levels stochastic fluctuation of TCA cycle expression

  • produces tolerant subpopulation with low intracellular ATP levels

  • contradicting studies regarding ATP level after TCA-cycle inhibition

  • Dsp1

    • → hypothetical lipoprotein

  • Asp1

    • → alkaline shock protein

deletion mutation of dsp1 or asp1
  • increased cell wall thickness

  • reduced autolysis

not reported
Increased antibiotic survival if amount or function of target increased

  • ppGpp

    • → nucleotides mediating the stringent response

  • RelA

  • → enzyme producing ppGpp

ppGpp-activated stringent response
  • results in global physiological changes and inactivation of antibiotic targets

  • gentamicin30

  • ciprofloxacin30

  • induces SCVs and promotes intraphagocyte survival

  • contradicting studies for ciprofloxacin

  • Saturated fatty acid in membrane

  • → influences membrane fluidity

increased amount of saturated fatty acid
  • reduces oligomerization and pore formation of daptomycin

not reported
  • growth phase- and strain-dependent effect on tolerance

  • effect dependent on the distribution of fatty acids in the growth medium


Reduced antibiotic survival if amount or function of target decreased

  • Clp

    • → important protease involved in stress response and virulence

  • → degrades antitoxins

lack of Clp
  • increases levels of antitoxin

  • decreases bacterial response to antibiotic stress

  • cfu-dependent/growth phase-dependent effect

  • MazF/MazE

  • → toxin/antitoxin system

presumable mechanism not known
  • oxacillin30

  • vancomycin30

  • ciprofloxacin30

  • rifampicin30

  • gentamicin30

  • associated with growth inhibition and SCVs

  • contradicting studies for β-lactams and vancomycin

  • msaABCR

  • → regulator involved in virulence and resistance

presumable mechanism not knownplanktonic exponential232
  • rifampicin

  • gentamicin

  • vancomycin

  • daptomycin

planktonic exponential232
  • linezolid

  • large variability according to growth phase

planktonic stationary232
  • linezolid

  • gentamicin

planktonic stationary232
  • daptomycin

  • vancomycin

  • rifampicin

biofilm232
  • daptomycin

  • vancomycin

  • linezolid

biofilm232
  • gentamicin

  • rifampicin

  • superoxide dismutase

  • → enzyme detoxifying ROS

lack of superoxide dismutase
  • increases antibiotic-induced ROS damaging DNA deletion of purF leads to

not reported
  • purF

  • → encodes enzyme for purine biosynthesis

  • higher ATP levels allowing bactericidal action of antibiotic

not reported
  • faster growth rate of tolerant strain compared with less tolerant ΔpurF mutant

Target associated with increased antibiotic survival and main known functionPresumable or associated mechanism(s) of increased antibiotic survivalChange of antibiotic survival shown for these antibioticsNo change of antibiotic survival found for these antibioticsComments
Increased antibiotic survival if amount or function of target decreased

  • agr

  • → global regulator of virulence factors

dysfunction of agr
  • decreases secreted phenol soluble modulins

  • increases detoxification of ROS by derepression of glutathione peroxidase BsaA

  • increases agr-repressed secreted factors interfering with extracellular antibiotics

  • oxacillin130

  • daptomycin130 (opposite effect)

  • strain-dependent effect

  • growth-phase-dependent effect

  • agr deficiency clinically associated with persistent infection

  • contradicting studies for daptomycin

  • ATP

    • → main cellular energy substrate

low level of ATP decreases activity of antibiotic targets and downstream bactericidal mechanism
  • ciprofloxacin30,31

  • oxacillin31

  • gentamicin31

not reported
  • may be a common trait of tolerant S. aureus; however, not prerequisite

  • GdpP

    • → membrane-located ci-d-AMP-phosphodiesterase

inactivation of GdpP
  • leads to increased intracellular second messenger ci-d-AMP 229

  • followed by more biofilm formation

  • ileS

  • → encodes isoleucine tRNA synthase

dysfunction of ileS
  • decreases protein synthesis through change of tRNA binding site

  • vancomycin28

not reported
  • murein hydrolases

    • → cleave components of cell wall

  • → mediators of autolysis

decreased activity of hydrolases
  • decreases antibiotic-induced autolysis

  • can be mediated by decreased activity of their regulators (e.g. cidABC and lrgAB)

  • may be mediated through vancomycin-induced inhibition of access to their targets

not reported
  • addition of glucose can increase activity of murein regulators CidABC/LrgA and decrease tolerance

  • pitA

    • → encodes inorganic phosphate transporter

point mutation in pitA leads to
  • intracellular accumulation of inorganic phosphate, which up-regulates the dlt operon

  • increased cell wall thickness

  • daptomycin ± ampicillin/gentamicin/vancomycin/ciprofloxacin/rifampicin127,128

  • ADEP4 ± rifampicin/daptomycin127,128

not reported
  • TCA cycle

  • → main metabolic pathway to transfer stored energy into ATP and NADH

inactivation by mutations of selected TCA enzymes
  • reduces membrane potential without change of intracellular ATP levels stochastic fluctuation of TCA cycle expression

  • produces tolerant subpopulation with low intracellular ATP levels

  • contradicting studies regarding ATP level after TCA-cycle inhibition

  • Dsp1

    • → hypothetical lipoprotein

  • Asp1

    • → alkaline shock protein

deletion mutation of dsp1 or asp1
  • increased cell wall thickness

  • reduced autolysis

not reported
Increased antibiotic survival if amount or function of target increased

  • ppGpp

    • → nucleotides mediating the stringent response

  • RelA

  • → enzyme producing ppGpp

ppGpp-activated stringent response
  • results in global physiological changes and inactivation of antibiotic targets

  • gentamicin30

  • ciprofloxacin30

  • induces SCVs and promotes intraphagocyte survival

  • contradicting studies for ciprofloxacin

  • Saturated fatty acid in membrane

  • → influences membrane fluidity

increased amount of saturated fatty acid
  • reduces oligomerization and pore formation of daptomycin

not reported
  • growth phase- and strain-dependent effect on tolerance

  • effect dependent on the distribution of fatty acids in the growth medium


Reduced antibiotic survival if amount or function of target decreased

  • Clp

    • → important protease involved in stress response and virulence

  • → degrades antitoxins

lack of Clp
  • increases levels of antitoxin

  • decreases bacterial response to antibiotic stress

  • cfu-dependent/growth phase-dependent effect

  • MazF/MazE

  • → toxin/antitoxin system

presumable mechanism not known
  • oxacillin30

  • vancomycin30

  • ciprofloxacin30

  • rifampicin30

  • gentamicin30

  • associated with growth inhibition and SCVs

  • contradicting studies for β-lactams and vancomycin

  • msaABCR

  • → regulator involved in virulence and resistance

presumable mechanism not knownplanktonic exponential232
  • rifampicin

  • gentamicin

  • vancomycin

  • daptomycin

planktonic exponential232
  • linezolid

  • large variability according to growth phase

planktonic stationary232
  • linezolid

  • gentamicin

planktonic stationary232
  • daptomycin

  • vancomycin

  • rifampicin

biofilm232
  • daptomycin

  • vancomycin

  • linezolid

biofilm232
  • gentamicin

  • rifampicin

  • superoxide dismutase

  • → enzyme detoxifying ROS

lack of superoxide dismutase
  • increases antibiotic-induced ROS damaging DNA deletion of purF leads to

not reported
  • purF

  • → encodes enzyme for purine biosynthesis

  • higher ATP levels allowing bactericidal action of antibiotic

not reported
  • faster growth rate of tolerant strain compared with less tolerant ΔpurF mutant

ROS, reactive oxygen species.

Deletion of the Clp proteolytic system [involved in degradation of several antitoxins in S. aureus toxin/antitoxin systems109 and in regulation of the stress response110,111 as well as the tricarboxylic acid (TCA) cycle112] was associated with increased rates of tolerant S. aureus.113 Similarly, deletion mutants of the S. aureus toxin/antitoxin system mazF/mazE exhibited decreased antibiotic survival after exposure to β-lactam antibiotics or vancomycin in planktonic as well as biofilm growth.114,115 It should be cautioned that this effect on antibiotic survival could not be confirmed after simultaneously knocking out three major toxin–antitoxin systems including MazF/MazE.30 Nevertheless, dysregulation of the MazF/MazE system can phenotypically lead to growth inhibition116 and SCVs85 as an indirect clue to its association with antibiotic survival in S. aureus. Further, several studies found increased antibiotic survival of S. aureus associated with activation of the guanosine tetraphosphate ppGpp-dependent stringent response and vice versa.117–119 This may not be true for all strains or other experimental setups according to one conflicting study.30 Nonetheless, parallels to phenotypes of tolerant life forms again exist since higher levels of ppGpp have been associated with SCVs120 and intraphagosomal survival.121 More than 40 years ago, the lack of autolysis was found to lead to increased survival with β-lactam antibiotics in S. aureus.122 More recent studies were able to associate deficiencies in CidABC and LrgA, both regulators of extracellular murein hydrolases, to increased survival through lack of autolysis.123,124 Concordantly, WGS associated mutation in the murein hydrolase atl with in vitro-generated phenotypes tolerant to oxacillin.125 Addition of glucose increased the activity of CidABC and LrgAB, thereby simultaneously decreasing the level of antibiotic survival.126

Other studies have specifically investigated mechanisms of increased survival of S. aureus with daptomycin, a membrane-targeting bactericidal antibiotic known for its growth-independent killing capacity.101 Changes in the membrane-lipid composition in the stationary phase increased antibiotic survival to daptomycin treatment. This observation could be influenced by the lipids available in the growth medium.104 Similarly, up-regulation of the dlt operon, encoding proteins responsible for d-alanylation of teichoic acids in the Gram-positive cell wall, increased daptomycin-specific antibiotic survival.127,128 However, changes in available glucose can diminish the rate of daptomycin-tolerant S. aureus.129 Recently, a genome-wide analysis identified several new genomic regions associated with daptomycin-tolerant S. aureus, again a strong hint for the plasticity and versatility of mechanisms of antibiotic survival in S. aureus.29

Studies have found a higher level of antibiotic survival in mutants with defects in the quorum sensing accessory gene regulator (agr).107,130 Dysfunctional agr may promote tolerant phenotypes through increased biofilm formation131 or through excreted factors interfering extracellularly with antibiotics such as gentamicin130 or daptomycin.132 Such increased antibiotic survival is supported by clinical data linking agr mutants to persistent infections.133–136

Antibiotics themselves may trigger antibiotic survival by ‘toxifying’ the environment of S. aureus. Indeed, evidence suggests that antibiotics can not only induce biofilm137 and SCV85,138 formation, but also trigger other mechanisms of antibiotic survival.139,140 In the case of S. aureus, repeated in vitro exposure to antibiotics can lead to increased antibiotic survival, as shown for β-lactams,25,118,141 aminoglycosides,142 vancomycin118 or daptomycin.127 For vancomycin, the proposed mechanisms include the induction of the ppGpp-dependent stringent response118 and direct blocking of the access of murein hydrolases to their cell-wall substrates.134,143 Moreover, pre-treatment with an antibiotic can result in increased survival not only for the applied antibiotic but also for other antibiotic classes.144–146 This cross-effect can vary greatly depending on which antibiotics are chosen97,103,147,148 and even which sequence of antibiotics is selected.144 It stretches from full cross-effect (as shown for tobramycin followed by ciprofloxacin) to no cross-effect (as shown for tobramycin and daptomycin).144 Finally, the widely used topical antimicrobial triclosan can increase antibiotic survival up to 10 000-fold, a mechanism again requiring ppGpp synthesis.149

Taken together, multiple mechanisms may mediate increased antibiotic survival of S. aureus. It remains to be seen whether they act in parallel or as redundant or upstream/downstream pathways. Nevertheless, the selective effect on individual antibiotics is a strong clue that different mechanisms of antibiotic survival are present. Clearly, a better understanding of the mechanisms of cellular death mediated by individual antibiotics should help decipher the various mechanisms underlying antibiotic survival.

In vivo experimental evidence for increased antibiotic survival of S. aureus

Although knowledge about in vitro phenomena of increased antibiotic survival is extensive, how this translates in vivo has not been ascertained. Moreover, there is a paucity of in vivo data on the outcome of infections with tolerant S. aureus since most available studies were performed more than 20 years ago. An early animal study by Chuard et al.150 gave proof of the impact of the in vivo environment on antibiotic survival. Measuring the S. aureus MIC and MBC of various antibiotics before and after a 6 week untreated foreign-body infection revealed such a drastic rise of the MBC that even the highest antibiotic concentrations could no longer kill the recovered S. aureus, notably without changes to the MIC. Various other animal studies have shown both the failure of antibiotics to eradicate in vivo infections, possibly due to tolerant phenotypes,55,151,152 and the increased in vivo survival of S. aureus already determined in vitro to be tolerant.108,153,154 However, not all studies found a correlation of in vitro with in vivo antibiotic survival.155,156 In summary, in vivo studies show the relevance of the host environment on antibiotic survival but also reveal the difficulty of translating in vitro measurements into in vivo differences of outcome.

Evidence of the clinical impact of increased antibiotic survival of S. aureus

Although bedside experience and the growing in vitro evidence argue for a major role of antibiotic-tolerant phenotypes of S. aureus in the clinical setting, the number of studies analysing antibiotic survival of S. aureus from patients is limited. The awareness of the clinical relevance of the antibiotic killing capacity, however, is not new and earlier studies have analysed the correlation between in vitro antibiotic survival and clinical outcome (Table 3). The MBC/MIC ratio, the most commonly used clinical measurement of tolerance, is fraught with considerable variability.15,16 Nevertheless, up to now it has been the best available marker of its clinical relevance. Most studies focus on vancomycin and describe a rate of tolerant S. aureus from 6%–43% in clinical strains;157–164 however, up to 63% oxacillin-tolerant S. aureus have been reported in patients with endocarditis.165 Single case reports describe treatment failures with tolerant strains.166–168 Larger series present a wide spectrum of clinical outcomes associated with ‘in vitro tolerant’ S. aureus. It ranges from no clinical difference165 to longer duration of fever,169 longer duration of bacteraemia170,171 and increased treatment failure in composite endpoints172–175 to an increased mortality rate.176–178 The relevance and clinical impact of increased antibiotic survival may depend on the site of infection: one study showed, for example, a worse outcome with tolerant strains only in bacteraemic patients with endocarditis but not in non-endocarditis patients.177 Most studies focus on increased survival with vancomycin, which may be a marker for treatment failure even when no vancomycin is used in the treatment.175 Remarkably, Miyazaki et al.178 were able to demonstrate a correlation of the level of bacterial glycopeptide survival (measured as reduction of cfu in kill curves) with the probability of survival of patients with MRSA bacteraemia. In conclusion, several studies over the last few decades found a correlation between increased antibiotic survival and clinical failure, but the differences in patient population and the lack of standardized methods of measuring antibiotic survival make a comparison between them difficult. Hence, there is an urgent clinical need to reliably measure and understand the dynamics and rate of increased antibiotic survival of S. aureus in patients.179

Table 3.

Clinical studies analysing the outcome of patients infected with tolerant S. aureus

YearStudy typeIncluded patientsNo. of patientsTolerance measurementAntibiotic testedOutcome/observation if tolerantRef
1977case reportpneumonia1MBC vs MICpenicillin, methicillinSeveral recurrences with tolerant S. aureus163
1978case reportendocarditis/ bacteraemia2MBC vs MICvancomycinTreatment failure under vancomycin monotherapy168
1979retrospective case seriesendocarditis and soft tissue abscess20MIC and MBCvariousHigher mortality if receiving non-bactericidal antibiotics according to MBC176
1980prospective cohortendocarditis and SAB104MBC/MIC ≥16various
  • Prolonged fever, more complications, more ICU treatment and higher mortality endocarditis

  • No clinical difference if SAB without endocarditis

177
1982retrospective cohortSAB20MBC/MIC ≥32vancomycinMore treatment failure172
1982case reportpneumonia1MBC/MIC ≥32oxacillinInitial treatment failure with oxacillin164
1986prospective cohortSAB84MBC/MIC ≥32nafcillinLonger fever with tolerant strains (but all cured)169
1987prospective cohortosteomyelitis27serum bactericidal titre (trough <1:2)variousMore therapeutic failure if trough bactericidal titre <1:2173
2004prospective cohortSAB30killing (<4.71, 4. 71–6.26, >6.27 log cfu/72 h)vancomycinLower clinical success rate if decreased killing174
2006retrospective cohortSAB3MBC/MIC ≥32vancomycinPersistent bacteraemia >72 h167
2007prospective cohortSAB34killing (>/<2.5 log cfu/24 h)vancomycinLonger duration of bacteraemia (<6.5 days vs >10.5 days)168
2011retrospective cohortendocarditis62MBC/MIC ≥32
  • oxacillin

  • vancomycin

  • teicoplanin

  • No difference in clinical outcome between oxacillin tolerant/non-tolerant

  • Non-significant worse outcome in vancomycin-tolerant strains

165
2011retrospective cohortSAB66killing (log cfu/72 h)vancomycinQuantitative correlation between probability of survival and amount of killing178
2017retrospective cohortSAB225MBC/MIC ≥32vancomycinMore clinical failure with vancomycin tolerance175
YearStudy typeIncluded patientsNo. of patientsTolerance measurementAntibiotic testedOutcome/observation if tolerantRef
1977case reportpneumonia1MBC vs MICpenicillin, methicillinSeveral recurrences with tolerant S. aureus163
1978case reportendocarditis/ bacteraemia2MBC vs MICvancomycinTreatment failure under vancomycin monotherapy168
1979retrospective case seriesendocarditis and soft tissue abscess20MIC and MBCvariousHigher mortality if receiving non-bactericidal antibiotics according to MBC176
1980prospective cohortendocarditis and SAB104MBC/MIC ≥16various
  • Prolonged fever, more complications, more ICU treatment and higher mortality endocarditis

  • No clinical difference if SAB without endocarditis

177
1982retrospective cohortSAB20MBC/MIC ≥32vancomycinMore treatment failure172
1982case reportpneumonia1MBC/MIC ≥32oxacillinInitial treatment failure with oxacillin164
1986prospective cohortSAB84MBC/MIC ≥32nafcillinLonger fever with tolerant strains (but all cured)169
1987prospective cohortosteomyelitis27serum bactericidal titre (trough <1:2)variousMore therapeutic failure if trough bactericidal titre <1:2173
2004prospective cohortSAB30killing (<4.71, 4. 71–6.26, >6.27 log cfu/72 h)vancomycinLower clinical success rate if decreased killing174
2006retrospective cohortSAB3MBC/MIC ≥32vancomycinPersistent bacteraemia >72 h167
2007prospective cohortSAB34killing (>/<2.5 log cfu/24 h)vancomycinLonger duration of bacteraemia (<6.5 days vs >10.5 days)168
2011retrospective cohortendocarditis62MBC/MIC ≥32
  • oxacillin

  • vancomycin

  • teicoplanin

  • No difference in clinical outcome between oxacillin tolerant/non-tolerant

  • Non-significant worse outcome in vancomycin-tolerant strains

165
2011retrospective cohortSAB66killing (log cfu/72 h)vancomycinQuantitative correlation between probability of survival and amount of killing178
2017retrospective cohortSAB225MBC/MIC ≥32vancomycinMore clinical failure with vancomycin tolerance175

SAB, S. aureus bacteraemia.

Table 3.

Clinical studies analysing the outcome of patients infected with tolerant S. aureus

YearStudy typeIncluded patientsNo. of patientsTolerance measurementAntibiotic testedOutcome/observation if tolerantRef
1977case reportpneumonia1MBC vs MICpenicillin, methicillinSeveral recurrences with tolerant S. aureus163
1978case reportendocarditis/ bacteraemia2MBC vs MICvancomycinTreatment failure under vancomycin monotherapy168
1979retrospective case seriesendocarditis and soft tissue abscess20MIC and MBCvariousHigher mortality if receiving non-bactericidal antibiotics according to MBC176
1980prospective cohortendocarditis and SAB104MBC/MIC ≥16various
  • Prolonged fever, more complications, more ICU treatment and higher mortality endocarditis

  • No clinical difference if SAB without endocarditis

177
1982retrospective cohortSAB20MBC/MIC ≥32vancomycinMore treatment failure172
1982case reportpneumonia1MBC/MIC ≥32oxacillinInitial treatment failure with oxacillin164
1986prospective cohortSAB84MBC/MIC ≥32nafcillinLonger fever with tolerant strains (but all cured)169
1987prospective cohortosteomyelitis27serum bactericidal titre (trough <1:2)variousMore therapeutic failure if trough bactericidal titre <1:2173
2004prospective cohortSAB30killing (<4.71, 4. 71–6.26, >6.27 log cfu/72 h)vancomycinLower clinical success rate if decreased killing174
2006retrospective cohortSAB3MBC/MIC ≥32vancomycinPersistent bacteraemia >72 h167
2007prospective cohortSAB34killing (>/<2.5 log cfu/24 h)vancomycinLonger duration of bacteraemia (<6.5 days vs >10.5 days)168
2011retrospective cohortendocarditis62MBC/MIC ≥32
  • oxacillin

  • vancomycin

  • teicoplanin

  • No difference in clinical outcome between oxacillin tolerant/non-tolerant

  • Non-significant worse outcome in vancomycin-tolerant strains

165
2011retrospective cohortSAB66killing (log cfu/72 h)vancomycinQuantitative correlation between probability of survival and amount of killing178
2017retrospective cohortSAB225MBC/MIC ≥32vancomycinMore clinical failure with vancomycin tolerance175
YearStudy typeIncluded patientsNo. of patientsTolerance measurementAntibiotic testedOutcome/observation if tolerantRef
1977case reportpneumonia1MBC vs MICpenicillin, methicillinSeveral recurrences with tolerant S. aureus163
1978case reportendocarditis/ bacteraemia2MBC vs MICvancomycinTreatment failure under vancomycin monotherapy168
1979retrospective case seriesendocarditis and soft tissue abscess20MIC and MBCvariousHigher mortality if receiving non-bactericidal antibiotics according to MBC176
1980prospective cohortendocarditis and SAB104MBC/MIC ≥16various
  • Prolonged fever, more complications, more ICU treatment and higher mortality endocarditis

  • No clinical difference if SAB without endocarditis

177
1982retrospective cohortSAB20MBC/MIC ≥32vancomycinMore treatment failure172
1982case reportpneumonia1MBC/MIC ≥32oxacillinInitial treatment failure with oxacillin164
1986prospective cohortSAB84MBC/MIC ≥32nafcillinLonger fever with tolerant strains (but all cured)169
1987prospective cohortosteomyelitis27serum bactericidal titre (trough <1:2)variousMore therapeutic failure if trough bactericidal titre <1:2173
2004prospective cohortSAB30killing (<4.71, 4. 71–6.26, >6.27 log cfu/72 h)vancomycinLower clinical success rate if decreased killing174
2006retrospective cohortSAB3MBC/MIC ≥32vancomycinPersistent bacteraemia >72 h167
2007prospective cohortSAB34killing (>/<2.5 log cfu/24 h)vancomycinLonger duration of bacteraemia (<6.5 days vs >10.5 days)168
2011retrospective cohortendocarditis62MBC/MIC ≥32
  • oxacillin

  • vancomycin

  • teicoplanin

  • No difference in clinical outcome between oxacillin tolerant/non-tolerant

  • Non-significant worse outcome in vancomycin-tolerant strains

165
2011retrospective cohortSAB66killing (log cfu/72 h)vancomycinQuantitative correlation between probability of survival and amount of killing178
2017retrospective cohortSAB225MBC/MIC ≥32vancomycinMore clinical failure with vancomycin tolerance175

SAB, S. aureus bacteraemia.

Although expert consensus on the definition of tolerance and persistence in general has been achieved,17 there is so far no validated and reliable tool to quantify and most of all classify tolerance or persistence for clinical application and decision-making. An optimal test for measuring tolerance or persistence would need to be highly standardized, reproducible within and between laboratories and strains, rapid, not too labour-intensive and correlated with clinical outcome. First promising steps in this direction have been taken with proposed new metrics and methodological approaches.117,180–184 For example, the metric MDK99 (minimal duration necessary for killing 99% of a bacterial population) could be used to quantify tolerance.17,180 However, standardization of pre-test growth conditions, starting inoculum, test media and conditions as well as the applicable antibiotic concentrations still need to be established and clinically relevant cut-offs determined. Moreover, the antibiotic concentration needs to reach saturation for killing in the MDK99 assay, which does not work for all antibiotics.180,185 Finally, the proposed setup to measure MDK99 probably needs a robotic system to fit into the workflow and manpower of a routine diagnostic microbiology laboratory.180

Approaches to treating antibiotic-tolerant S. aureus

With the increasing awareness of increased antibiotic survival, research has turned its focus on new ways of treating and eradicating tolerant S. aureus. Three main approaches have been explored so far to combat tolerant S. aureus (Figure 2): (i) disrupting growth- or metabolism-independent targets of increased antibiotic survival with anti-tolerance compounds; (ii) improving the activity of existing antibiotics (by changing methods and duration of administration); and (iii) resensitizing tolerant cells to existing antibiotics.186

Published strategies for the possible treatment of tolerant S. aureus infections.
Figure 2.

Published strategies for the possible treatment of tolerant S. aureus infections.

Strategies targeting mechanisms of increased antibiotic survival have produced compounds capable of killing tolerant S. aureus, the majority of which target the cell membrane, an optimal target in metabolically quiescent cells. Retinoids, a new class of antibiotics, disrupt the lipid bilayer of tolerant S. aureus.187 Various membrane-active small molecules, such as NH125,188–190 XF-73,191 HT-61192 or NCK10193 have shown good activity against tolerant S. aureus. A broad clinical application may be limited by specific properties such as induction of significant haemolysis through NH125 or low bioavailability of XF-73 and HT-61 due to strong hydrophobicity. Nevertheless, XF-73 has successfully completed Phase I/II tests and HT-61 has passed Phase II trials and is currently undergoing Phase III trials.186 Engineered cationic amphipathic peptides,194 bacteriophage-derived or synthetic lysins,195–197 the potassium ionophore boromycin,198 surface-attached polyethylenimine199 or even the anthelminthic biothionol200 can kill tolerant S. aureus through cell wall/membrane interaction.

Another promising new synthetic antibiotic is ADEP4, which has the unique feature of not blocking but activating its target. It binds intracellularly to the protease ClpP resulting in an ATP-independent uncontrolled self-digestion and death of the cell.55,201,202 However, this substance must be used in combination with other antibiotics as its use is fraught with rapid development of escape mutants.55 Similarly, a clinical drug-library screen identified the quinolone tosufloxacin as highly active against tolerant S. aureus—even more active than ADEP4 in one in vitro study.203 Novel compounds such as SPI009,204 halogenated indoles,205 phenazine-inspired small molecules206 or chelators of iron and zinc207 have an effect on tolerant S. aureus cells. Even the anticancer drugs mitomycin and cisplatin demonstrate bactericidal action on tolerant S. aureus through crosslinking DNA.208,209 However, their clinical application in S. aureus infections seems limited due to their inherently unfavourable toxicity.

Enhancing existing antibiotics may be the most straightforward way of tackling tolerant S. aureus. For example, combinations of antibiotics may achieve a synergistic effect on tolerant S. aureus as shown in vitro for daptomycin and tobramycin.103,144 However, that study applied very high doses of 100× MIC, which is not achievable systemically in patients due to toxicity issues, especially considering protein binding of 92%. Localized administration could, in theory, circumvent systemic toxicity in implant-associated infection.210 Prolonging the duration of antibiotic treatment represents another approach. Concordantly, in vitro long-term antibiotic exposures of up to 24 days led to complete eradication of tolerant S. aureus populations.103 Recently, a mathematical model successfully predicted the duration of treatment for eradication of infections with tolerant bacteria.211 These results foster the discussion of treatment duration in patients and may also explain why short-course antibiotic treatment (<14 days) is associated with more relapse even in uncomplicated S. aureus bacteraemia.212 As early as 1944, Bigger suggested an intermittent antibiotic treatment to let the dormant tolerant subpopulation awaken in antibiotic-free intervals, rendering them amenable to the next antibiotic killing.35 The potential efficacy of this pulsed treatment was recently validated in vitro.213 However, whether this effect can be applied clinically to improve the outcome in treatment of tolerant infections remains to be proven. Another way of improving the available antibiotics is to synthetically fuse them to cleavable moieties with cell-penetrating properties79,214–217 or to change the carrier formulation (Figure 2).218–220 This either increases the penetration into S. aureus per se, as shown for the fusion antibiotic pentobra, or ameliorates the penetration into mammalian cells to access the intracellular S. aureus population.

Finally, resensitizing tolerant S. aureus to existing antibiotics has become a focus of research. Changing bacterial metabolism by varying the composition of environmental metabolites can influence antibiotic killing of tolerant S. aureus. As an example, addition of fructose to biofilm cultures enabled growth-independent killing through aminoglycosides;97 addition of glucose increased the effect of daptomycin,129 vancomycin and rifampicin,126 or of quinolones when combined with oxygen.38 Mechanistically, this has been linked to metabolite-enabled generation of proton-motive force,97 activation of cell respiration38 or increased transcription of murein hydrolase.126 It is worth noting that potentiation is generally antibiotic and metabolite specific,97 supportive of the notion that various tolerance mechanisms are at play. Changing the environment is not limited to metabolic stimuli, as pH modification by addition of the basic amino acid l-arginine or exposing tolerant S. aureus to alkaline conditions increased the killing effect of aminoglycosides.221,222 Further, interfering with chemical signalling by using supernatant from S. aureus medium223,224 or fatty acid signalling molecules225 succeeded in resuscitating dormant S. aureus. These results of sensitizing by ‘environmental reprogramming’ are intriguing; however, the complex nutritional and metabolic interactions in an infected tissue could make it challenging to specifically trigger the desired response for synergistic therapeutic reasons. Finally, the effect of existing antibiotics on tolerant S. aureus may be augmented by adding phenol-soluble modulins,226 bacteriophages227 or rhamnolipids228 to the treatment.

Open questions and conclusions

Our knowledge of the determinants and significance of antibiotic-tolerant S. aureus in patients remains patchy at best. Does the same mechanism of increased antibiotic survival account for a late relapse after stopping antibiotic treatment62 and continuous infection as found in persistent bacteraemia?7 Do the various microenvironments in the infected milieu determine antibiotic survival more than the cellular stochasticity that is thought to underlie the persister phenotype? How relevant is increased antibiotic survival in non-bloodstream infections such as in implant infections? What role does the antibiotic selected have on development of tolerant phenotypes during treatment? What is the therapeutic role of so-called bacteriostatic antibiotics in infections with tolerant S. aureus? What influence does the immune system exert on S. aureus to become tolerant? Can we identify bacterial or host markers for tolerant infections? How long can tolerant S. aureus stay in the metabolically quiescent state in the human body? Do they revert stochastically or only by induction through environmental triggers? Do tolerant subpopulations actually harbour mutations, which rapidly revert back upon subculturing? Would it be an advantage to regularly screen all patients for tolerant S. aureus or only in those with treatment failure? Could we adapt treatment duration to the level of antibiotic survival or should we aim to individualize an effective combination therapy? To answer these and other questions, a reliable and reproducible tool of measuring antibiotic survival is critical.

In conclusion, S. aureus may display increased survival to antibiotic treatment through a plethora of inducers and mechanisms, reflecting its physiological versatility and infectious context. Increased antibiotic survival should be considered as a potential factor for the unacceptably high rate of treatment failure. New insights and promising approaches are appearing on the horizon.

However, clinical studies are needed to better understand the dynamics, mechanisms and relevance of tolerant S. aureus at the site of infection and to step ahead in the fight against one of mankind’s enduring foes.

Funding

R. K. was supported by the research funds of the University of Basel.

Transparency declarations

None to declare.

References

1

Wertheim
HF
,
Melles
DC
,
Vos
MC
et al.
The role of nasal carriage in Staphylococcus aureus infections
.
Lancet Infect Dis
2005
;
5
:
751
62
.

2

Tong
SY
,
Davis
JS
,
Eichenberger
E
et al.
Staphylococcus aureus infections: epidemiology, pathophysiology, clinical manifestations, and management
.
Clin Microbiol Rev
2015
;
28
:
603
61
.

3

Freidlin
J
,
Acharya
N
,
Lietman
TM
et al.
Spectrum of eye disease caused by methicillin-resistant Staphylococcus aureus
.
Am J Ophthalmol
2007
;
144
:
313
5
.

4

Sheagren
JN.
Staphylococcus aureus. The persistent pathogen (second of two parts)
.
N Engl J Med
1984
;
310
:
1437
42
.

5

Fowler
VG
,
Olsen
MK
,
Corey
GR
et al.
Clinical identifiers of complicated Staphylococcus aureus bacteremia
.
Arch Intern Med
2003
;
163
:
2066
72
.

6

Chang
FY
,
MacDonald
BB
,
Peacock
JE
Jr
et al.
A prospective multicenter study of Staphylococcus aureus bacteremia: incidence of endocarditis, risk factors for mortality, and clinical impact of methicillin resistance
.
Medicine
2003
;
82
:
322
32
.

7

Chong
YP
,
Park
SJ
,
Kim
HS
et al.
Persistent Staphylococcus aureus bacteremia: a prospective analysis of risk factors, outcomes, and microbiologic and genotypic characteristics of isolates
.
Medicine
2013
;
92
:
98
108
.

8

Canzoneri
CN
,
Akhavan
BJ
,
Tosur
Z
et al.
Follow-up blood cultures in Gram-negative bacteremia: are they needed?
Clin Infect Dis
2017
;
65
:
1776
9
.

9

Welsh
KJ
,
Skrobarcek
KA
,
Abbott
AN
et al.
Predictors of relapse of methicillin-resistant Staphylococcus aureus bacteremia after treatment with vancomycin
.
J Clin Microbiol
2011
;
49
:
3669
72
.

10

Wiggers
JB
,
Xiong
W
,
Daneman
N.
Sending repeat cultures: is there a role in the management of bacteremic episodes? (SCRIBE study)
.
BMC Infect Dis
2016
;
16
:
286.

11

van Hal
SJ
,
Jensen
SO
,
Vaska
VL
et al.
Predictors of mortality in Staphylococcus aureus bacteremia
.
Clin Microbiol Rev
2012
;
25
:
362
86
.

12

Liu
C
,
Bayer
A
,
Cosgrove
SE
et al.
Clinical practice guidelines by the Infectious Diseases Society of America for the treatment of methicillin-resistant Staphylococcus aureus infections in adults and children
.
Clin Infect Dis
2011
;
53
:
319
.

13

Singer
AJ
,
Talan
DA.
Management of skin abscesses in the era of methicillin-resistant Staphylococcus aureus
.
N Engl J Med
2014
;
370
:
1039
47
.

14

Ascione
T
,
Pagliano
P
,
Mariconda
M
et al.
Factors related to outcome of early and delayed prosthetic joint infections
.
J Infection
2015
;
70
:
30
6
.

15

Ishida
K
,
Guze
PA
,
Kalmanson
GM
et al.
Variables in demonstrating methicillin tolerance in Staphylococcus aureus strains
.
Antimicrob Agents Chemother
1982
;
21
:
688
90
.

16

Sherris
JC.
Problems in in vitro determination of antibiotic tolerance in clinical isolates
.
Antimicrob Agents Chemother
1986
;
30
:
633
7
.

17

Balaban
NQ
,
Helaine
S
,
Lewis
K
et al.
Definitions and guidelines for research on antibiotic persistence
.
Nat Rev Microbiol
2019
;
17
:
441
8
.

18

Brauner
A
,
Fridman
O
,
Gefen
O
et al.
Distinguishing between resistance, tolerance and persistence to antibiotic treatment
.
Nat Rev Microbiol
2016
;
14
:
320
30
.

19

Hurdle
JG
,
O’Neill
AJ
,
Chopra
I
et al.
Targeting bacterial membrane function: an underexploited mechanism for treating persistent infections
.
Nat Rev Microbiol
2011
;
9
:
62
75
.

20

O’Neill
A
, Bacterial phenotypes refractory to antibiotic-mediated killing: mechanisms and mitigation. In:
Miller
A
, ed.
Emerging Trends in Antibacterial Discovery: Answering the Call to Arms
.
Caister Academic Press
,
2011
.

21

Gollan
B
,
Grabe
G
,
Michaux
C
et al.
Bacterial persisters and infection: past, present, and progressing
.
Annu Rev Microbiol
2019
;
73
:
359
85
.

22

Conlon
BP.
Staphylococcus aureus chronic and relapsing infections: evidence of a role for persister cells: an investigation of persister cells, their formation and their role in S. aureus disease
.
BioEssays
2014
;
36
:
991
6
.

23

Meylan
S
,
Andrews
IW
,
Collins
JJ.
Targeting antibiotic tolerance, pathogen by pathogen
.
Cell
2018
;
172
:
1228
38
.

24

Radzikowski
JL
,
Schramke
H
,
Heinemann
M.
Bacterial persistence from a system-level perspective
.
Curr Opin Biotechnol
2017
;
46
:
98
105
.

25

Tomasz
A
,
de Vegvar
ML.
Construction of a penicillin-tolerant laboratory mutant of Staphylococcus aureus
.
Eur J Clin Microbiol
1986
;
5
:
710
3
.

26

Yee
R
,
Cui
P
,
Shi
W
et al.
Genetic screen reveals the role of purine metabolism in Staphylococcus aureus persistence to rifampicin
.
Antibiotics
2015
;
4
:
627
42
.

27

Wang
W
,
Chen
J
,
Chen
G
et al.
Transposon mutagenesis identifies novel genes associated with Staphylococcus aureus persister formation
.
Frontiers Microbiol
2015
;
6
:
1437
.

28

Singh
M
,
Matsuo
M
,
Sasaki
T
et al.
In vitro tolerance of drug-naive Staphylococcus aureus strain FDA209P to vancomycin
.
Antimicrob Agents Chemother
2017
;
61
:
e01154
-
16
.

29

Berti
AD
,
Shukla
N
,
Rottier
AD
et al.
Daptomycin selects for genetic and phenotypic adaptations leading to antibiotic tolerance in MRSA
.
J Antimicrob Chemother
2018
;
73
:
2030
3
.

30

Conlon
BP
,
Rowe
SE
,
Gandt
AB
et al.
Persister formation in Staphylococcus aureus is associated with ATP depletion
.
Nat Microbiol
2016
;
1
:
16051.

31

Zalis
EA
,
Nuxoll
AS
,
Manuse
S
et al.
Stochastic variation in expression of the tricarboxylic acid cycle produces persister cells
.
mBio
2019
;
10
: doi:10.1128/mBio.01930-19.

32

Lewis
K.
Persister cells
.
Annu Rev Microbiol
2010
;
64
:
357
72
.

33

Stokes
JM
,
Lopatkin
AJ
,
Lobritz
MA
et al.
Bacterial metabolism and antibiotic efficacy
.
Cell Metabol
2019
;
30
:
251
9
.

34

Crabbe
A
,
Jensen
PO
,
Bjarnsholt
T
et al.
Antimicrobial tolerance and metabolic adaptations in microbial biofilms
.
Trends Microbiol
2019
;
27
:
850
63
.

35

Bigger
JW.
Treatment of staphylococcal infections with penicillin - by intermittent sterilisation
.
Lancet
1944
;
2
:
497
500
.

36

Pabst
B
,
Pitts
B
,
Lauchnor
E
et al.
Gel-entrapped Staphylococcus aureus bacteria as models of biofilm infection exhibit growth in dense aggregates, oxygen limitation, antibiotic tolerance, and heterogeneous gene expression
.
Antimicrob Agents Chemother
2016
;
60
:
6294
301
.

37

Lamp
KC
,
Rybak
MJ
,
Bailey
EM
et al.
In vitro pharmacodynamic effects of concentration, pH, and growth phase on serum bactericidal activities of daptomycin and vancomycin
.
Antimicrob Agents Chemother
1992
;
36
:
2709
14
.

38

Gutierrez
A
,
Jain
S
,
Bhargava
P
et al.
Understanding and sensitizing density-dependent persistence to quinolone antibiotics
.
Molecular Cell
2017
;
68
:
1147
54.e3
.

39

Fux
CA
,
Costerton
JW
,
Stewart
PS
et al.
Survival strategies of infectious biofilms
.
Trends Microbiol
2005
;
13
:
34
40
.

40

Liu
C
,
Bayer
A
,
Cosgrove
SE
et al.
Clinical practice guidelines by the Infectious Diseases Society of America for the treatment of methicillin-resistant Staphylococcus aureus infections in adults and children
.
Clin Infect Dis
2011
;
52
:
e18
55
.

41

Haaber
J
,
Cohn
MT
,
Frees
D
et al.
Planktonic aggregates of Staphylococcus aureus protect against common antibiotics
.
PLoS One
2012
;
7
:
e41075.

42

Newsom
SW.
Ogston’s coccus
.
J Hosp Infection
2008
;
70
:
369
72
.

43

Dastgheyb
SS
,
Hammoud
S
,
Ketonis
C
et al.
Staphylococcal persistence due to biofilm formation in synovial fluid containing prophylactic cefazolin
.
Antimicrob Agents Chemother
2015
;
59
:
2122
8
.

44

Dastgheyb
S
,
Parvizi
J
,
Shapiro
IM
et al.
Effect of biofilms on recalcitrance of staphylococcal joint infection to antibiotic treatment
.
J Infect Dis
2015
;
211
:
641
50
.

45

Cheng
AG
,
DeDent
AC
,
Schneewind
O
et al.
A play in four acts: Staphylococcus aureus abscess formation
.
Trends Microbiol
2011
;
19
:
225
32
.

46

Patel
AR
,
Alton
TB
,
Bransford
RJ
et al.
Spinal epidural abscesses: risk factors, medical versus surgical management, a retrospective review of 128 cases
.
Spine
2014
;
14
:
326
30
.

47

Wagner
C
,
Sauermann
R
,
Joukhadar
C.
Principles of antibiotic penetration into abscess fluid
.
Pharmacology
2006
;
78
:
1
10
.

48

Vulin
C
,
Leimer
N
,
Huemer
M
et al.
Prolonged bacterial lag time results in small colony variants that represent a sub-population of persisters
.
Nature Commun
2018
;
9
:
4074
.

49

Hanke
ML
,
Kielian
T.
Deciphering mechanisms of staphylococcal biofilm evasion of host immunity
.
Frontiers Cell Infection Microbiol
2012
;
2
:
62
.

50

Zapotoczna
M
,
O’Neill
E
,
O’Gara
JP.
Untangling the diverse and redundant mechanisms of Staphylococcus aureus biofilm formation
.
PLoS Pathog
2016
;
12
:
e1005671.

51

Holland
TL
,
Baddour
LM
,
Bayer
AS
et al.
Infective endocarditis
.
Nat Rev Dis Primers
2016
;
2
:
16059.

52

Darouiche
RO.
Treatment of infections associated with surgical implants
.
N Engl J Med
2004
;
350
:
1422
9
.

53

Costerton
JW
,
Cheng
KJ
,
Geesey
GG
et al.
Bacterial biofilms in nature and disease
.
Annu Rev Microbiol
1987
;
41
:
435
64
.

54

Molina-Manso
D
,
del Prado
G
,
Ortiz-Perez
A
et al.
In vitro susceptibility to antibiotics of staphylococci in biofilms isolated from orthopaedic infections
.
Int J Antimicrob Agents
2013
;
41
:
521
3
.

55

Conlon
BP
,
Nakayasu
ES
,
Fleck
LE
et al.
Activated ClpP kills persisters and eradicates a chronic biofilm infection
.
Nature
2013
;
503
:
365
70
.

56

Okuda
K
,
Zendo
T
,
Sugimoto
S
et al.
Effects of bacteriocins on methicillin-resistant Staphylococcus aureus biofilm
.
Antimicrob Agents Chemother
2013
;
57
:
5572
9
.

57

Darouiche
RO
,
Dhir
A
,
Miller
AJ
et al.
Vancomycin penetration into biofilm covering infected prostheses and effect on bacteria
.
J Infect Dis
1994
;
170
:
720
3
.

58

Jefferson
KK
,
Goldmann
DA
,
Pier
GB.
Use of confocal microscopy to analyze the rate of vancomycin penetration through Staphylococcus aureus biofilms
.
Antimicrob Agents Chemother
2005
;
49
:
2467
73
.

59

Stewart
PS
,
Davison
WM
,
Steenbergen
JN.
Daptomycin rapidly penetrates a Staphylococcus epidermidis biofilm
.
Antimicrob Agents Chemother
2009
;
53
:
3505
7
.

60

Kirker
KR
,
Fisher
ST
,
James
GA.
Potency and penetration of telavancin in staphylococcal biofilms
.
Int J Antimicrob Agents
2015
;
46
:
451
5
.

61

Rani
SA
,
Pitts
B
,
Beyenal
H
et al.
Spatial patterns of DNA replication, protein synthesis, and oxygen concentration within bacterial biofilms reveal diverse physiological states
.
J Bacteriol
2007
;
189
:
4223
33
.

62

Lewis
K.
Riddle of biofilm resistance
.
Antimicrob Agents Chemother
2001
;
45
:
999
1007
.

63

Stewart
PS.
Antimicrobial tolerance in biofilms
.
Microbiol Spectrum
2015
;
3
: MB-0010-2014.

64

Wolcott
RD
,
Rumbaugh
KP
,
James
G
et al.
Biofilm maturity studies indicate sharp debridement opens a time-dependent therapeutic window
.
J Wound Care
2010
;
19
:
320
8
.

65

Zimmerli
W
,
Trampuz
A
,
Ochsner
PE.
Prosthetic-joint infections
.
N Engl J Med
2004
;
351
:
1645
54
.

66

Barberan
J
,
Aguilar
L
,
Carroquino
G
et al.
Conservative treatment of staphylococcal prosthetic joint infections in elderly patients
.
Am J Med
2006
;
119
:
993.e7
10
.

67

Boles
BR
,
Horswill
AR.
Agr-mediated dispersal of Staphylococcus aureus biofilms
.
PLoS Pathog
2008
;
4
:
e1000052.

68

Fux
CA
,
Wilson
S
,
Stoodley
P.
Detachment characteristics and oxacillin resistance of Staphyloccocus aureus biofilm emboli in an in vitro catheter infection model
.
J Bacteriol
2004
;
186
:
4486
91
.

69

Kubica
M
,
Guzik
K
,
Koziel
J
et al.
A potential new pathway for Staphylococcus aureus dissemination: the silent survival of S. aureus phagocytosed by human monocyte-derived macrophages
.
PLoS One
2008
;
3
:
e1409.

70

Garzoni
C
,
Kelley
WL.
Staphylococcus aureus: new evidence for intracellular persistence
.
Trends Microbiol
2009
;
17
:
59
65
.

71

Fraunholz
M
,
Sinha
B.
Intracellular Staphylococcus aureus: live-in and let die
.
Front Cell Infect Microbiol
2012
;
2
:
43
.

72

Yang
D
,
Wijenayaka
AR
,
Solomon
LB
et al.
Novel insights into Staphylococcus aureus deep bone infections: the involvement of osteocytes
.
mBio
2018
;
9
:
e00415-18
.

73

Boudjemaa
R
,
Steenkeste
K
,
Jacqueline
C
et al.
Live intramacrophagic Staphylococcus aureus as a potential cause of antibiotic therapy failure: observations in an in vivo mouse model of prosthetic vascular material infections
.
J Antimicrob Chemother
2018
;
73
:
2418
21
.

74

Zautner
AE
,
Krause
M
,
Stropahl
G
et al.
Intracellular persisting Staphylococcus aureus is the major pathogen in recurrent tonsillitis
.
PLoS One
2010
;
5
:
e9452.

75

Thwaites
GE
,
Gant
V.
Are bloodstream leukocytes Trojan Horses for the metastasis of Staphylococcus aureus?
Nat Rev Microbiol
2011
;
9
:
215
22
.

76

Abu-Humaidan
AH
,
Elven
M
,
Sonesson
A
et al.
Persistent intracellular Staphylococcus aureus in keratinocytes lead to activation of the complement system with subsequent reduction in the intracellular bacterial load
.
Front Immunol
2018
;
9
:
396
.

77

Clement
S
,
Vaudaux
P
,
Francois
P
et al.
Evidence of an intracellular reservoir in the nasal mucosa of patients with recurrent Staphylococcus aureus rhinosinusitis
.
J Infect Dis
2005
;
192
:
1023
8
.

78

Tan
X
,
Coureuil
M
,
Ramond
E
et al.
Chronic Staphylococcus aureus lung infection correlates with proteogenomic and metabolic adaptations leading to an increased intracellular persistence
.
Clin Infect Dis
2019
;
69
:
1937
45
.

79

Lehar
SM
,
Pillow
T
,
Xu
M
et al.
Novel antibody-antibiotic conjugate eliminates intracellular S. aureus
.
Nature
2015
;
527
:
323
8
.

80

Rigaill
J
,
Morgene
MF
,
Gavid
M
et al.
Intracellular activity of antimicrobial compounds used for Staphylococcus aureus nasal decolonization
.
J Antimicrob Chemother
2018
;
73
:
3044
8
.

81

Sandberg
A
,
Jensen
KS
,
Baudoux
P
et al.
Intra- and extracellular activities of dicloxacillin against Staphylococcus aureus in vivo and in vitro
.
Antimicrob Agents Chemother
2010
;
54
:
2391
400
.

82

Krut
O
,
Sommer
H
,
Kronke
M.
Antibiotic-induced persistence of cytotoxic Staphylococcus aureus in non-phagocytic cells
.
J Antimicrob Chemother
2004
;
53
:
167
73
.

83

Tuchscherr
L
,
Heitmann
V
,
Hussain
M
et al.
Staphylococcus aureus small-colony variants are adapted phenotypes for intracellular persistence
.
J Infect Dis
2010
;
202
:
1031
40
.

84

Loffler
B
,
Tuchscherr
L
,
Niemann
S
et al.
Staphylococcus aureus persistence in non-professional phagocytes
.
Int J Med Microbiol
2014
;
304
:
170
6
.

85

Proctor
RA
,
Kriegeskorte
A
,
Kahl
BC
et al.
Staphylococcus aureus small colony variants (SCVs): a road map for the metabolic pathways involved in persistent infections
.
Frontiers Cell Infection Microbiol
2014
;
4
:
99
.

86

Tuchscherr
L
,
Medina
E
,
Hussain
M
et al.
Staphylococcus aureus phenotype switching: an effective bacterial strategy to escape host immune response and establish a chronic infection
.
EMBO Mol Med
2011
;
3
:
129
41
.

87

Guerillot
R
,
Kostoulias
X
,
Donovan
L
et al.
Unstable chromosome rearrangements in Staphylococcus aureus cause phenotype switching associated with persistent infections
.
Proc Natl Acad Sci USA
2019
;
116
:
20135
40
.

88

Chuard
C
,
Vaudaux
PE
,
Proctor
RA
et al.
Decreased susceptibility to antibiotic killing of a stable small colony variant of Staphylococcus aureus in fluid phase and on fibronectin-coated surfaces
.
J Antimicrob Chemother
1997
;
39
:
603
8
.

89

Garcia
LG
,
Lemaire
S
,
Kahl
BC
et al.
Antibiotic activity against small-colony variants of Staphylococcus aureus: review of in vitro, animal and clinical data
.
J Antimicrob Chemother
2013
;
68
:
1455
64
.

90

Proctor
RA
,
van Langevelde
P
,
Kristjansson
M
et al.
Persistent and relapsing infections associated with small-colony variants of Staphylococcus aureus
.
Clin Infect Dis
1995
;
20
:
95
102
.

91

Proctor
RA
,
von Eiff
C
,
Kahl
BC
et al.
Small colony variants: a pathogenic form of bacteria that facilitates persistent and recurrent infections
.
Nat Rev Microbiol
2006
;
4
:
295
305
.

92

Painter
KL
,
Strange
E
,
Parkhill
J
et al.
Staphylococcus aureus adapts to oxidative stress by producing H2O2-resistant small-colony variants via the SOS response
.
Infect Immun
2015
;
83
:
1830
44
.

93

Leimer
N
,
Rachmuhl
C
,
Palheiros Marques
M
et al.
Nonstable Staphylococcus aureus small-colony variants are induced by low pH and sensitized to antimicrobial therapy by phagolysosomal alkalinization
.
J Infect Dis
2016
;
213
:
305
13
.

94

Cardile
AP
,
Sanchez
CJ
Jr
,
Samberg
ME
et al.
Human plasma enhances the expression of staphylococcal microbial surface components recognizing adhesive matrix molecules promoting biofilm formation and increases antimicrobial tolerance in vitro
.
BMC Res Notes
2014
;
7
:
457
.

95

Balaban
NQ
,
Merrin
J
,
Chait
R
et al.
Bacterial persistence as a phenotypic switch
.
Science
2004
;
305
:
1622
5
.

96

Fridman
O
,
Goldberg
A
,
Ronin
I
et al.
Optimization of lag time underlies antibiotic tolerance in evolved bacterial populations
.
Nature
2014
;
513
:
418
21
.

97

Allison
KR
,
Brynildsen
MP
,
Collins
JJ.
Metabolite-enabled eradication of bacterial persisters by aminoglycosides
.
Nature
2011
;
473
:
216
20
.

98

Lechner
S
,
Prax
M
,
Lange
B
et al.
Metabolic and transcriptional activities of Staphylococcus aureus challenged with high-doses of daptomycin
.
Int J Med Microbiol
2014
;
304
:
931
40
.

99

Stapels
DAC
,
Hill
PWS
,
Westermann
AJ
et al.
Salmonella persisters undermine host immune defenses during antibiotic treatment
.
Science
2018
;
362
:
1156
60
.

100

Keren
I
,
Kaldalu
N
,
Spoering
A
et al.
Persister cells and tolerance to antimicrobials
.
FEMS Microbiol Lett
2004
;
230
:
13
8
.

101

Mascio
CT
,
Alder
JD
,
Silverman
JA.
Bactericidal action of daptomycin against stationary-phase and nondividing Staphylococcus aureus cells
.
Antimicrob Agents Chemother
2007
;
51
:
4255
60
.

102

Garrigos
C
,
Murillo
O
,
Euba
G
et al.
Efficacy of usual and high doses of daptomycin in combination with rifampin versus alternative therapies in experimental foreign-body infection by methicillin-resistant Staphylococcus aureus
.
Antimicrob Agents Chemother
2010
;
54
:
5251
6
.

103

Lechner
S
,
Lewis
K
,
Bertram
R.
Staphylococcus aureus persisters tolerant to bactericidal antibiotics
.
J Mol Microbiol Biotechnol
2012
;
22
:
235
44
.

104

Boudjemaa
R
,
Cabriel
C
,
Dubois-Brissonnet
F
et al.
Impact of bacterial membrane fatty acid composition on the failure of daptomycin to kill Staphylococcus aureus
.
Antimicrob Agents Chemother
2018
;
62
: doi:10.1128/AAC.00023-18.

105

Shan
Y
,
Brown Gandt
A
,
Rowe
SE
et al.
ATP-dependent persister formation in Escherichia coli
.
mBio
2017
;
8
:
e02267-16
.

106

Wang
Y
,
Bojer
MS
,
George
SE
et al.
Inactivation of TCA cycle enhances Staphylococcus aureus persister cell formation in stationary phase
.
Sci Rep
2018
;
8
:
10849.

107

Xu
T
,
Wang
XY
,
Cui
P
et al.
The Agr quorum sensing system represses persister formation through regulation of phenol soluble modulins in Staphylococcus aureus
.
Frontiers Microbiol
2017
;
8
:
2189
.

108

Li
L
,
Abdelhady
W
,
Donegan
NP
et al.
Role of purine biosynthesis in persistent methicillin-resistant Staphylococcus aureus (MRSA) infection
.
J Infect Dis
2018
;
8
:
e02267-16
.

109

Donegan
NP
,
Thompson
ET
,
Fu
Z
et al.
Proteolytic regulation of toxin-antitoxin systems by ClpPC in Staphylococcus aureus
.
J Bacteriol
2010
;
192
:
1416
22
.

110

Michel
A
,
Agerer
F
,
Hauck
CR
et al.
Global regulatory impact of ClpP protease of Staphylococcus aureus on regulons involved in virulence, oxidative stress response, autolysis, and DNA repair
.
J Bacteriol
2006
;
188
:
5783
96
.

111

Frees
D
,
Gerth
U
,
Ingmer
H.
Clp chaperones and proteases are central in stress survival, virulence and antibiotic resistance of Staphylococcus aureus
.
Int J Med Microbiol
2014
;
304
:
142
9
.

112

Chatterjee
I
,
Becker
P
,
Grundmeier
M
et al.
Staphylococcus aureus ClpC is required for stress resistance, aconitase activity, growth recovery, and death
.
J Bacteriol
2005
;
187
:
4488
96
.

113

Springer
MT
,
Singh
VK
,
Cheung
AL
et al.
Effect of clpP and clpC deletion on persister cell number in Staphylococcus aureus
.
J Med Microbiol
2016
;
65
:
848
57
.

114

Schuster
CF
,
Mechler
L
,
Nolle
N
et al.
The MazEF toxin-antitoxin system alters the β-lactam susceptibility of Staphylococcus aureus
.
PLoS One
2015
;
10
:
e0126118.

115

Ma
D
,
Mandell
JB
,
Donegan
NP
et al.
The toxin-antitoxin MazEF drives Staphylococcus aureus biofilm formation, antibiotic tolerance, and chronic infection
.
mBio
2019
;
10
: doi:10.1128/mBio.01658-19.

116

Fu
Z
,
Tamber
S
,
Memmi
G
et al.
Overexpression of MazFsa in Staphylococcus aureus induces bacteriostasis by selectively targeting mRNAs for cleavage
.
J Bacteriol
2009
;
191
:
2051
9
.

117

Matsuo
M
,
Hiramatsu
M
,
Singh
M
et al.
Genetic and transcriptomic analyses of ciprofloxacin-tolerant Staphylococcus aureus isolated by the replica plating tolerance isolation system (REPTIS)
.
Antimicrob Agents Chemother
2019
;
63
:
e02019-18.

118

Geiger
T
,
Kastle
B
,
Gratani
FL
et al.
Two small (p)ppGpp synthases in Staphylococcus aureus mediate tolerance against cell envelope stress conditions
.
J Bacteriol
2014
;
196
:
894
902
.

119

Corrigan
RM
,
Bellows
LE
,
Wood
A
et al.
ppGpp negatively impacts ribosome assembly affecting growth and antimicrobial tolerance in Gram-positive bacteria
.
Proc Natl Acad Sci USA
2016
;
113
:
E1710
9
.

120

Gao
W
,
Chua
K
,
Davies
JK
et al.
Two novel point mutations in clinical Staphylococcus aureus reduce linezolid susceptibility and switch on the stringent response to promote persistent infection
.
PLoS Pathog
2010
;
6
:
e1000944.

121

Geiger
T
,
Francois
P
,
Liebeke
M
et al.
The stringent response of Staphylococcus aureus and its impact on survival after phagocytosis through the induction of intracellular PSMs expression
.
PLoS Pathog
2012
;
8
:
e1003016.

122

Best
GK
,
Best
NH
,
Koval
AV.
Evidence for participation of autolysins in bactericidal action of oxacillin on Staphylococcus aureus
.
Antimicrob Agents Chemother
1974
;
6
:
825
30
.

123

Groicher
KH
,
Firek
BA
,
Fujimoto
DF
et al.
The Staphylococcus aureus lrgAB operon modulates murein hydrolase activity and penicillin tolerance
.
J Bacteriol
2000
;
182
:
1794
801
.

124

Rice
KC
,
Firek
BA
,
Nelson
JB
et al.
The Staphylococcus aureus cidAB operon: evaluation of its role in regulation of murein hydrolase activity and penicillin tolerance
.
J Bacteriol
2003
;
185
:
2635
43
.

125

Chung
M
,
Borges
V
,
Gomes
JP
et al.
Phenotypic signatures and genetic determinants of oxacillin tolerance in a laboratory mutant of Staphylococcus aureus
.
PLoS One
2018
;
13
:
e0199707.

126

Rice
KC
,
Nelson
JB
,
Patton
TG
et al.
Acetic acid induces expression of the Staphylococcus aureus cidABC and lrgAB murein hydrolase regulator operons
.
J Bacteriol
2005
;
187
:
813
21
.

127

Mechler
L
,
Herbig
A
,
Paprotka
K
et al.
A novel point mutation promotes growth phase-dependent daptomycin tolerance in Staphylococcus aureus
.
Antimicrob Agents Chemother
2015
;
59
:
5366
76
.

128

Mechler
L
,
Bonetti
EJ
,
Reichert
S
et al.
Daptomycin tolerance in the Staphylococcus aureus pitA6 mutant is due to upregulation of the dlt operon
.
Antimicrob Agents Chemother
2016
;
60
:
2684
91
.

129

Prax
M
,
Mechler
L
,
Weidenmaier
C
et al.
Glucose augments killing efficiency of daptomycin challenged Staphylococcus aureus persisters
.
PLoS One
2016
;
11
:
e0150907.

130

Kumar
K
,
Chen
J
,
Drlica
K
et al.
Tuning of the lethal response to multiple stressors with a single-site mutation during clinical infection by Staphylococcus aureus
.
mBio
2017
;
8
:
e01476-17
.

131

Dastgheyb
SS
,
Villaruz
AE
,
Le
KY
et al.
Role of phenol-soluble modulins in formation of Staphylococcus aureus biofilms in synovial fluid
.
Infect Immun
2015
;
83
:
2966
75
.

132

Pader
V
,
Hakim
S
,
Painter
KL
et al.
Staphylococcus aureus inactivates daptomycin by releasing membrane phospholipids
.
Nat Microbiol
2016
;
2
:
16194.

133

Fowler
VG
Jr,
Sakoulas
G
,
McIntyre
LM
et al.
Persistent bacteremia due to methicillin-resistant Staphylococcus aureus infection is associated with agr dysfunction and low-level in vitro resistance to thrombin-induced platelet microbicidal protein
.
J Infect Dis
2004
;
190
:
1140
9
.

134

Sakoulas
G
,
Gold
HS
,
Cohen
RA
et al.
Effects of prolonged vancomycin administration on methicillin-resistant Staphylococcus aureus (MRSA) in a patient with recurrent bacteraemia
.
J Antimicrob Chemother
2006
;
57
:
699
704
.

135

Painter
KL
,
Krishna
A
,
Wigneshweraraj
S
et al.
What role does the quorum-sensing accessory gene regulator system play during Staphylococcus aureus bacteremia?
Trends Microbiol
2014
;
22
:
676
85
.

136

Suligoy
CM
,
Lattar
SM
,
Noto Llana
M
et al.
Mutation of agr is associated with the adaptation of Staphylococcus aureus to the host during chronic osteomyelitis
.
Front Cell Infect Microbiol
2018
;
8
:
18
.

137

Kaplan
JB
,
Izano
EA
,
Gopal
P
et al.
Low levels of β-lactam antibiotics induce extracellular DNA release and biofilm formation in Staphylococcus aureus
.
mBio
2012
;
3
:
e00198
-
12
.

138

Vestergaard
M
,
Paulander
W
,
Ingmer
H.
Activation of the SOS response increases the frequency of small colony variants
.
BMC Res Notes
2015
;
8
:
749.

139

Harms
A
,
Maisonneuve
E
,
Gerdes
K.
Mechanisms of bacterial persistence during stress and antibiotic exposure
.
Science
2016
;
354
: doi:10.1126/science.aaf4268/.

140

Van den Bergh
B
,
Michiels
JE
,
Wenseleers
T
et al.
Frequency of antibiotic application drives rapid evolutionary adaptation of Escherichia coli persistence
.
Nat Microbiol
2016
;
1
:
16020.

141

Voorn
GP
,
Thompson
J
,
Goessens
WH
et al.
In vitro development and stability of tolerance to cloxacillin and vancomycin in Staphylococcus aureus
.
Eur J Clin Microbiol Infect Dis
1994
;
13
:
741
6
.

142

Michiels
JE
,
Van den Bergh
B
,
Verstraeten
N
et al.
In vitro emergence of high persistence upon periodic aminoglycoside challenge in the ESKAPE pathogens
.
Antimicrob Agents Chemother
2016
;
60
:
4630
7
.

143

Sieradzki
K
,
Tomasz
A.
Inhibition of the autolytic system by vancomycin causes mimicry of vancomycin-intermediate Staphylococcus aureus-type resistance, cell concentration dependence of the MIC, and antibiotic tolerance in vancomycin-susceptible S. aureus
.
Antimicrob Agents Chemother
2006
;
50
:
527
33
.

144

Lechner
S
,
Patra
P
,
Klumpp
S
et al.
Interplay between population dynamics and drug tolerance of Staphylococcus aureus persister cells
.
J Mol Microbiol Biotechnol
2012
;
22
:
381
91
.

145

Haaber
J
,
Friberg
C
,
McCreary
M
et al.
Reversible antibiotic tolerance induced in Staphylococcus aureus by concurrent drug exposure
.
mBio
2015
;
6
:
e02268-14
.

146

Lobritz
MA
,
Belenky
P
,
Porter
CB
et al.
Antibiotic efficacy is linked to bacterial cellular respiration
.
Proc Natl Acad Sci USA
2015
;
112
:
8173
80
.

147

Sabath
LD
,
Wheeler
N
,
Laverdiere
M
et al.
A new type of penicillin resistance of Staphylococcus aureus
.
Lancet
1977
;
1
:
443
7
.

148

Henry-Stanley
MJ
,
Hess
DJ
,
Wells
CL.
Aminoglycoside inhibition of Staphylococcus aureus biofilm formation is nutrient dependent
.
J Med Microbiol
2014
;
63
:
861
9
.

149

Westfall
C
,
Flores-Mireles
AL
,
Robinson
JI
et al.
The widely used antimicrobial triclosan induces high levels of antibiotic tolerance in vitro and reduces antibiotic efficacy up to 100-fold in vivo
.
Antimicrob Agents Chemother
2019
;
63
:
e02312
-
18
.

150

Chuard
C
,
Lucet
JC
,
Rohner
P
et al.
Resistance of Staphylococcus aureus recovered from infected foreign body in vivo to killing by antimicrobials
.
J Infect Dis
1991
;
163
:
1369
73
.

151

Tuomanen
E
,
Durack
DT
,
Tomasz
A.
Antibiotic tolerance among clinical isolates of bacteria
.
Antimicrob Agents Chemother
1986
;
30
:
521
7
.

152

Vaudaux
P.
Phenotypic antibiotic tolerance of Staphylococcus aureus in implant-related infections: relationship with in vitro colonization of artificial surfaces
.
Drug Resist Updat
1998
;
1
:
352
7
.

153

Goessens
WH
,
Fontijne
P
,
Michel
MF.
Responses of tolerant and nontolerant Staphylococcus aureus strains to methicillin treatment in an experimental infection in mice
.
Antimicrob Agents Chemother
1984
;
26
:
829
32
.

154

Voorn
GP
,
Thompson
J
,
Goessens
WH
et al.
Role of tolerance in cloxacillin treatment of experimental Staphylococcus aureus endocarditis
.
J Infect Dis
1991
;
163
:
640
3
.

155

Goldman
PL
,
Petersdorf
RG.
Significance of methicillin tolerance in experimental staphylococcal endocarditis
.
Antimicrob Agents Chemother
1979
;
15
:
802
6
.

156

Guze
PA
,
Kalmanson
GM
,
Guze
LB.
The role of antibiotic tolerance in the response to treatment of pyelonephritis due to Staphylococcus aureus in rats
.
J Infect Dis
1982
;
145
:
169
73
.

157

Rose
WE
,
Fallon
M
,
Moran
JJ
et al.
Vancomycin tolerance in methicillin-resistant Staphylococcus aureus: influence of vancomycin, daptomycin, and telavancin on differential resistance gene expression
.
Antimicrob Agents Chemother
2012
;
56
:
4422
7
.

158

Traczewski
MM
,
Katz
BD
,
Steenbergen
JN
et al.
Inhibitory and bactericidal activities of daptomycin, vancomycin, and teicoplanin against methicillin-resistant Staphylococcus aureus isolates collected from 1985 to 2007
.
Antimicrob Agents Chemother
2009
;
53
:
1735
8
.

159

Appleman
MD
,
Citron
DM.
Efficacy of vancomycin and daptomycin against Staphylococcus aureus isolates collected over 29 years
.
Diagn Microbiol Infect Dis
2010
;
66
:
441
4
.

160

Cazares-Dominguez
V
,
Cruz-Cordova
A
,
Ochoa
SA
et al.
Vancomycin tolerant, methicillin-resistant Staphylococcus aureus reveals the effects of vancomycin on cell wall thickening
.
PLoS One
2015
;
10
:
e0118791.

161

Sader
HS
,
Jones
RN
,
Rossi
KL
et al.
Occurrence of vancomycin-tolerant and heterogeneous vancomycin-intermediate strains (hVISA) among Staphylococcus aureus causing bloodstream infections in nine USA hospitals
.
J Antimicrob Chemother
2009
;
64
:
1024
8
.

162

Jones
RN.
Microbiological features of vancomycin in the 21st century: minimum inhibitory concentration creep, bactericidal/static activity, and applied breakpoints to predict clinical outcomes or detect resistant strains
.
Clin Infect Dis
2006
;
42 Suppl 1
:
S13
24
.

163

Perry
JD
,
Jones
AL
,
Gould
FK.
Glycopeptide tolerance in bacteria causing endocarditis
.
J Antimicrob Chemother
1999
;
44
:
121
4
.

164

May
J
,
Shannon
K
,
King
A
et al.
Glycopeptide tolerance in Staphylococcus aureus
.
J Antimicrob Chemother
1998
;
42
:
189
97
.

165

Pasticci
MB
,
Moretti
A
,
Stagni
G
et al.
Bactericidal activity of oxacillin and glycopeptides against Staphylococcus aureus in patients with endocarditis: looking for a relationship between tolerance and outcome
.
Ann Clin Microbiol Antimicrob
2011
;
10
:
26.

166

Haldane
EV
,
Affias
S.
Penicillin-tolerant Staphylococcus aureus
.
Lancet
1977
;
2
:
39.

167

Svenungsson
B
,
Kalin
M
,
Lindgren
LG.
Therapeutic failure in pneumonia caused by a tolerant strain of Staphylococcus aureus
.
Scand J Infect Dis
1982
;
14
:
309
11
.

168

Faville
RJ
Jr,
Zaske
DE
,
Kaplan
EL
et al.
Staphylococcus aureus endocarditis. Combined therapy with vancomycin and rifampin
.
JAMA
1978
;
240
:
1963
5
.

169

Rahal
JJ
Jr,
Chan
YK
,
Johnson
G.
Relationship of staphylococcal tolerance, teichoic acid antibody, and serum bactericidal activity to therapeutic outcome in Staphylococcus aureus bacteremia
.
Am J Med
1986
;
81
:
43
52
.

170

Safdar
A
,
Rolston
KV.
Vancomycin tolerance, a potential mechanism for refractory gram-positive bacteremia observational study in patients with cancer
.
Cancer
2006
;
106
:
1815
20
.

171

Moise
PA
,
Sakoulas
G
,
Forrest
A
et al.
Vancomycin in vitro bactericidal activity and its relationship to efficacy in clearance of methicillin-resistant Staphylococcus aureus bacteremia
.
Antimicrob Agents Chemother
2007
;
51
:
2582
6
.

172

Sorrell
TC
,
Packham
DR
,
Shanker
S
et al.
Vancomycin therapy for methicillin-resistant Staphylococcus aureus
.
Ann Intern Med
1982
;
97
:
344
50
.

173

Weinstein
MP
,
Stratton
CW
,
Hawley
HB
et al.
Multicenter collaborative evaluation of a standardized serum bactericidal test as a predictor of therapeutic efficacy in acute and chronic osteomyelitis
.
Am J Med
1987
;
83
:
218
22
.

174

Sakoulas
G
,
Moise-Broder
PA
,
Schentag
J
et al.
Relationship of MIC and bactericidal activity to efficacy of vancomycin for treatment of methicillin-resistant Staphylococcus aureus bacteremia
.
J Clin Microbiol
2004
;
42
:
2398
402
.

175

Britt
NS
,
Patel
N
,
Shireman
TI
et al.
Relationship between vancomycin tolerance and clinical outcomes in Staphylococcus aureus bacteraemia
.
J Antimicrob Chemother
2017
;
72
:
535
42
.

176

Denny
AE
,
Peterson
LR
,
Gerding
DN
et al.
Serious staphylococcal infections with strains tolerant to bactericidal antibiotics
.
Arch Intern Med
1979
;
139
:
1026
31
.

177

Rajashekaraiah
KR
,
Rice
T
,
Rao
VS
et al.
Clinical significance of tolerant strains of Staphylococcus aureus in patients with endocarditis
.
Ann Intern Med
1980
;
93
:
796
801
.

178

Miyazaki
M
,
Takata
T
,
Yoshimura
H
et al.
Vancomycin bactericidal activity as a predictor of 30-day mortality in patients with methicillin-resistant Staphylococcus aureus bacteremia
.
Antimicrob Agents Chemother
2011
;
55
:
1819
20
.

179

Van den Bergh
B
,
Michiels
JE
,
Fauvart
M
et al.
Should we develop screens for multi-drug antibiotic tolerance?
Expert Rev Anti Infect Ther
2016
;
14
:
613
6
.

180

Brauner
A
,
Shoresh
N
,
Fridman
O
et al.
An experimental framework for quantifying bacterial tolerance
.
Biophysical J
2017
;
112
:
2664
71
.

181

Levin-Reisman
I
,
Gefen
O
,
Fridman
O
et al.
Automated imaging with ScanLag reveals previously undetectable bacterial growth phenotypes
.
Nat Methods
2010
;
7
:
737
9
.

182

Kotkova
H
,
Cabrnochova
M
,
Licha
I
et al.
Evaluation of TD test for analysis of persistence or tolerance in clinical isolates of Staphylococcus aureus
.
J Microbiol Methods
2019
;
167
:
105705
.

183

Peterson
LR
,
Denny
AE
,
Gerding
DN
et al.
Determination of tolerance to antibiotic bactericidal activity on Kirby-Bauer susceptibility plates
.
Am J Clin Pathol
1980
;
74
:
645
50
.

184

Gefen
O
,
Chekol
B
,
Strahilevitz
J
et al.
TDtest: easy detection of bacterial tolerance and persistence in clinical isolates by a modified disk-diffusion assay
.
Sci Rep
2017
;
7
:
41284.

185

Abel Zur Wiesch
P
,
Abel
S
,
Gkotzis
S
et al.
Classic reaction kinetics can explain complex patterns of antibiotic action
.
Sci Transl Med
2015
;
7
:
287ra73.

186

Defraine
V
,
Fauvart
M
,
Michiels
J.
Fighting bacterial persistence: current and emerging anti-persister strategies and therapeutics
.
Drug Resist Updat
2018
;
38
:
12
26
.

187

Kim
W
,
Zhu
W
,
Hendricks
GL
et al.
A new class of synthetic retinoid antibiotics effective against bacterial persisters
.
Nature
2018
;
556
:
103
7
.

188

Kim
W
,
Conery
AL
,
Rajamuthiah
R
et al.
Identification of an antimicrobial agent effective against methicillin-resistant Staphylococcus aureus persisters using a fluorescence-based screening strategy
.
PLoS One
2015
;
10
:
e0127640.

189

Kim
W
,
Fricke
N
,
Conery
AL
et al.
NH125 kills methicillin-resistant Staphylococcus aureus persisters by lipid bilayer disruption
.
Future Med Chem
2016
;
8
:
257
69
.

190

Abouelhassan
Y
,
Basak
A
,
Yousaf
H
et al.
Identification of N-arylated NH125 analogues as rapid eradicating agents against MRSA persister cells and potent biofilm killers of Gram-positive pathogens
.
Chembiochem
2017
;
18
:
352
7
.

191

Ooi
N
,
Miller
K
,
Randall
C
et al.
XF-70 and XF-73, novel antibacterial agents active against slow-growing and non-dividing cultures of Staphylococcus aureus including biofilms
.
J Antimicrob Chemother
2010
;
65
:
72
8
.

192

Hu
Y
,
Shamaei-Tousi
A
,
Liu
Y
et al.
A new approach for the discovery of antibiotics by targeting non-multiplying bacteria: a novel topical antibiotic for staphylococcal infections
.
PLoS One
2010
;
5
:
e11818.

193

Ghosh
C
,
Manjunath
GB
,
Konai
MM
et al.
Aryl-alkyl-lysines: agents that kill planktonic cells, persister cells, biofilms of MRSA and protect mice from skin-infection
.
PLoS One
2015
;
10
:
e0144094.

194

Mandell
JB
,
Deslouches
B
,
Montelaro
RC
et al.
Elimination of antibiotic resistant surgical implant biofilms using an engineered cationic amphipathic peptide WLBU2
.
Sci Rep
2017
;
7
:
18098.

195

Gutierrez
D
,
Ruas-Madiedo
P
,
Martinez
B
et al.
Effective removal of staphylococcal biofilms by the endolysin LysH5
.
PLoS One
2014
;
9
:
e107307.

196

Schuch
R
,
Khan
BK
,
Raz
A
et al.
Bacteriophage lysin CF-301, a potent antistaphylococcal biofilm agent
.
Antimicrob Agents Chemother
2017
;
61
: doi:10.1128/AAC.02666-16.

197

de Breij
A
,
Riool
M
,
Cordfunke
RA
et al.
The antimicrobial peptide SAAP-148 combats drug-resistant bacteria and biofilms
.
Sci Transl Med
2018
;
10
: doi:10.1126/scitranslmed.aan4044.

198

Moreira
W
,
Aziz
DB
,
Dick
T.
Boromycin kills mycobacterial persisters without detectable resistance
.
Front Microbiol
2016
;
7
:
199
.

199

Milovic
NM
,
Wang
J
,
Lewis
K
et al.
Immobilized N-alkylated polyethylenimine avidly kills bacteria by rupturing cell membranes with no resistance developed
.
Biotechnol Bioeng
2005
;
90
:
715
22
.

200

Kim
W
,
Zou
G
,
Hari
TPA
et al.
A selective membrane-targeting repurposed antibiotic with activity against persistent methicillin-resistant Staphylococcus aureus
.
Proc Natl Acad Sci USA
2019
;
116
:
16529
34
.

201

Brotz-Oesterhelt
H
,
Beyer
D
,
Kroll
HP
et al.
Dysregulation of bacterial proteolytic machinery by a new class of antibiotics
.
Nat Med
2005
;
11
:
1082
7
.

202

Kirstein
J
,
Hoffmann
A
,
Lilie
H
et al.
The antibiotic ADEP reprogrammes ClpP, switching it from a regulated to an uncontrolled protease
.
EMBO Mol Med
2009
;
1
:
37
49
.

203

Niu
H
,
Cui
P
,
Yee
R
et al.
A clinical drug library screen identifies tosufloxacin as being highly active against Staphylococcus aureus persisters
.
Antibiotics
2015
;
4
:
329
36
.

204

Defraine
V
,
Verstraete
L
,
Van Bambeke
F
et al.
Antibacterial activity of 1-[(2,4-dichlorophenethyl)amino]-3-phenoxypropan-2-ol against antibiotic-resistant strains of diverse bacterial pathogens, biofilms and in pre-clinical infection models
.
Front Microbiol
2017
;
8
:
2585
.

205

Lee
JH
,
Kim
YG
,
Gwon
G
et al.
Halogenated indoles eradicate bacterial persister cells and biofilms
.
AMB Expr
2016
;
6
:
123.

206

Garrison
AT
,
Abouelhassan
Y
,
Kallifidas
D
et al.
Halogenated phenazines that potently eradicate biofilms, MRSA persister cells in non-biofilm cultures, and Mycobacterium tuberculosis
.
Angew Chem Int Ed
2015
;
54
:
14819
23
.

207

Abouelhassan
Y
,
Yang
Q
,
Yousaf
H
et al.
Nitroxoline: a broad-spectrum biofilm-eradicating agent against pathogenic bacteria
.
Int J Antimicrob Agents
2017
;
49
:
247
51
.

208

Kwan
BW
,
Chowdhury
N
,
Wood
TK.
Combatting bacterial infections by killing persister cells with mitomycin C
.
Environ Microbiol
2015
;
17
:
4406
14
.

209

Chowdhury
N
,
Wood
TL
,
Martinez-Vazquez
M
et al.
DNA-crosslinker cisplatin eradicates bacterial persister cells
.
Biotechnol Bioeng
2016
;
113
:
1984
92
.

210

Wahlig
H
,
Dingeldein
E
,
Bergmann
R
et al.
The release of gentamicin from polymethylmethacrylate beads. An experimental and pharmacokinetic study
.
J Bone Joint Surg
1978
;
60-B
:
270
5
.

211

Martinecz
A
,
Abel Zur Wiesch
P
.
Estimating treatment prolongation for persistent infections
.
Pathog Dis
2018
;
76
: doi:10.1093/femspd/fty065.

212

Chong
YP
,
Moon
SM
,
Bang
KM
et al.
Treatment duration for uncomplicated Staphylococcus aureus bacteremia to prevent relapse: analysis of a prospective observational cohort study
.
Antimicrob Agents Chemother
2013
;
57
:
1150
6
.

213

Cogan
NG
,
Rath
H
,
Kommerein
N
et al.
Theoretical and experimental evidence for eliminating persister bacteria by manipulating killing timing
.
FEMS Microbiol Lett
2016
;
363
: doi:10.1093/femsle/fnw264.

214

Schmidt
NW
,
Deshayes
S
,
Hawker
S
et al.
Engineering persister-specific antibiotics with synergistic antimicrobial functions
.
ACS Nano
2014
;
8
:
8786
93
.

215

Brezden
A
,
Mohamed
MF
,
Nepal
M
et al.
Dual targeting of intracellular pathogenic bacteria with a cleavable conjugate of kanamycin and an antibacterial cell-penetrating peptide
.
J Am Chem Soc
2016
;
138
:
10945
9
.

216

Mohamed
MF
,
Brezden
A
,
Mohammad
H
et al.
Targeting biofilms and persisters of ESKAPE pathogens with P14KanS, a kanamycin peptide conjugate
.
Biochim Biophys Acta
2017
;
1861
:
848
59
.

217

Wang
Z
,
Kong
L
,
Liu
Y
et al.
A phage lysin fused to a cell-penetrating peptide kills intracellular methicillin-resistant Staphylococcus aureus in keratinocytes and has potential as a treatment for skin infections in mice
.
Appl Environ Microbiol
2018
;
84
: doi:10.1128/AEM.00380-18.

218

Surewaard
BG
,
Deniset
JF
,
Zemp
FJ
et al.
Identification and treatment of the Staphylococcus aureus reservoir in vivo
.
J Exp Med
2016
;
213
:
1141
51
.

219

Kamaruzzaman
NF
,
Kendall
S
,
Good
L.
Targeting the hard to reach: challenges and novel strategies in the treatment of intracellular bacterial infections
.
Br J Pharmacol
2017
;
174
:
2225
36
.

220

Antonoplis
A
,
Zang
X
,
Huttner
MA
et al.
A dual-function antibiotic-transporter conjugate exhibits superior activity in sterilizing MRSA biofilms and killing persister cells
.
J Am Chem Soc
2018
;
140
:
16140
51
.

221

Lebeaux
D
,
Chauhan
A
,
Letoffe
S
et al.
pH-mediated potentiation of aminoglycosides kills bacterial persisters and eradicates in vivo biofilms
.
J Infect Dis
2014
;
210
:
1357
66
.

222

Jiafeng
L
,
Fu
X
,
Chang
Z.
Hypoionic shock treatment enables aminoglycosides antibiotics to eradicate bacterial persisters
.
Sci Rep
2015
;
5
:
14247.

223

Pascoe
B
,
Dams
L
,
Wilkinson
TS
et al.
Dormant cells of Staphylococcus aureus are resuscitated by spent culture supernatant
.
PLoS One
2014
;
9
:
e85998.

224

Pasquaroli
S
,
Zandri
G
,
Vignaroli
C
et al.
Antibiotic pressure can induce the viable but non-culturable state in Staphylococcus aureus growing in biofilms
.
J Antimicrob Chemother
2013
;
68
:
1812
7
.

225

Kalita
S
,
Kandimalla
R
,
Bhowal
AC
et al.
Functionalization of β-lactam antibiotic on lysozyme capped gold nanoclusters retrogress MRSA and its persisters following awakening
.
Sci Rep
2018
;
8
:
5778.

226

Bojer
MS
,
Lindemose
S
,
Vestergaard
M
et al.
Quorum sensing-regulated phenol-soluble modulins limit persister cell populations in Staphylococcus aureus
.
Front Microbiol
2018
;
9
:
255
.

227

Kumaran
D
,
Taha
M
,
Yi
Q
et al.
Does treatment order matter? Investigating the ability of bacteriophage to augment antibiotic activity against Staphylococcus aureus biofilms
.
Front Microbiol
2018
;
9
:
127
.

228

Radlinski
LC
,
Rowe
SE
,
Brzozowski
R
et al.
Chemical induction of aminoglycoside uptake overcomes antibiotic tolerance and resistance in Staphylococcus aureus
.
Cell Chem Biol
2019
;
26
:
1355
64.e4
.

229

Corrigan
RM
,
Abbott
JC
,
Burhenne
H
et al.
c-di-AMP is a new second messenger in Staphylococcus aureus with a role in controlling cell size and envelope stress
.
PLoS Pathog
2011
;
7
:
e1002217.

230

Griffiths
JM
,
O’Neill
AJ.
Loss of function of the GdpP protein leads to joint β-lactam/glycopeptide tolerance in Staphylococcus aureus
.
Antimicrob Agents Chemother
2012
;
56
:
579
81
.

231

Barros
EM
,
Martin
MJ
,
Selleck
EM
et al.
Daptomycin resistance and tolerance due to loss of function in Staphylococcus aureus dsp1 and asp23
.
Antimicrob Agents Chemother
2019
;
63
: doi:10.1128/AAC.01542-18.

232

Sahukhal
GS
,
Pandey
S
,
Elasri
MO.
msaABCR operon is involved in persister cell formation in Staphylococcus aureus
.
BMC Microbiol
2017
;
17
:
218.

233

Ladjouzi
R
,
Bizzini
A
,
Lebreton
F
et al.
Analysis of the tolerance of pathogenic enterococci and Staphylococcus aureus to cell wall active antibiotics
.
J Antimicrob Chemother
2013
;
68
:
2083
91
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)