Top Banner
See discussions, stats, and author profiles for this publication at: http://www.researchgate.net/publication/273469084 Therapeutic Options and Emerging Alternatives for Multidrug Resistant Staphylococcal Infections ARTICLE · MARCH 2015 DOI: 10.2174/1381612821666150310101851 · Source: PubMed DOWNLOADS 26 VIEWS 115 8 AUTHORS, INCLUDING: Cristian Bologa University of New Mexico 74 PUBLICATIONS 1,582 CITATIONS SEE PROFILE Stylianos Chatzipanagiotou National and Kapodistrian University of Athens 69 PUBLICATIONS 398 CITATIONS SEE PROFILE Michael Hamblin Massachusetts General Hospital 407 PUBLICATIONS 9,568 CITATIONS SEE PROFILE Available from: Cristian Bologa Retrieved on: 31 July 2015
16

Therapeutic options Staphylococcus

Dec 14, 2015

Download

Documents

A review for MRSA and therapeutic modalities
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: Therapeutic options Staphylococcus

Seediscussions,stats,andauthorprofilesforthispublicationat:http://www.researchgate.net/publication/273469084

TherapeuticOptionsandEmergingAlternativesforMultidrugResistantStaphylococcalInfections

ARTICLE·MARCH2015

DOI:10.2174/1381612821666150310101851·Source:PubMed

DOWNLOADS

26

VIEWS

115

8AUTHORS,INCLUDING:

CristianBologa

UniversityofNewMexico

74PUBLICATIONS1,582CITATIONS

SEEPROFILE

StylianosChatzipanagiotou

NationalandKapodistrianUniversityofAthens

69PUBLICATIONS398CITATIONS

SEEPROFILE

MichaelHamblin

MassachusettsGeneralHospital

407PUBLICATIONS9,568CITATIONS

SEEPROFILE

Availablefrom:CristianBologa

Retrievedon:31July2015

Page 2: Therapeutic options Staphylococcus

2058 Current Pharmaceutical Design, 2015, 21, 2058-2072

Therapeutic Options and Emerging Alternatives for Multidrug Resistant Staphylo-coccal Infections

Maria Magana1, Anastasios Ioannidis

1,2, Emmanouil Magiorkinis

3, Oleg Ursu

4,5, Cristian G. Bologa

4,5,

Stylianos Chatzipanagiotou2, Michael R. Hamblin

6,7,8 and George P. Tegos*

7,8,9

1Department of Nursing, Faculty of Human Movement and Quality of Life Sciences, University of Peloponnese,

Sparta, Greece; 2Department of Clinical Microbiology, Athens Medical School, Aeginition Hospital, Athens,

Greece; 3Department of Hygiene, Epidemiology and Medical Statistics, Medical School, University of Athens,

Athens-Goudi, Greece; 4Translational Informatics Division, Department of Internal Medicine, University of New

Mexico Health Sciences Center, Albuquerque, NM, USA; 5Center for Molecular Discovery, University of New

Mexico, Albuquerque, NM, USA; 6Harvard-MIT Division of Health Science and Technology, Cambridge, MA,

USA; 7Department of Dermatology, Harvard Medical School, Boston, MA, USA;

8Wellman Center for Pho-

tomedicine, Massachusetts General Hospital, Boston, MA, USA; 9Torrey Pines Institute for Molecular Studies

11350 SW Village Parkway, Port St. Lucie, FL 34987 USA

Abstract: Methicillin-resistant Staphylococcus aureus (MRSA) remains the single biggest challenge in infectious disease in the civilized world. Moreover, vancomycin resistance is also spreading, leading to fears of untreatable infections as were

common in ancient times. Molecular microbiology and bioinformatics have revealed many of the mechanisms involved in resistance de-velopment. Mobile genetic elements, up-regulated virulence factors and multi-drug efflux pumps have been implicated. A range of ap-

proved antibiotics from the glycopeptide, lipopeptide, pleuromutilin, macrolide, oxazolidinone, lincosamide, aminoglycoside, tetracy-cline, steptogramin, and cephalosporin classes has been employed to treat MRSA infections. The upcoming pipeline of drugs for MRSA

includes some new compounds from the above classes, together with fluoroquinolones, antibacterial peptide mimetics, aminomethylci-clines, porphyrins, peptide deformylase inhibitors, oxadiazoles, and diaminopyrimidines. A range of non-drug alternative approaches has

emerged for MRSA treatment. Bacteriophage-therapy including purified lysins has made a comeback after being discovered in the 1930s. Quorum-sensing inhibitors are under investigation. Small molecule inhibitors of multi-drug efflux pumps may potentiate existing antibi-

otics. The relative failure of staphylococcal vaccines is being revisited by efforts with multi-valent vaccines and improved adjuvants. Photodynamic therapy uses non-toxic photosensitizers and harmless visible light to produce reactive oxygen species that can non-

specifically destroy bacteria while preserving host cells. Preparation of nanoparticles can kill bacteria themselves, as well as improve the delivery of anti-bacterial drugs. Anti-MRSA drug discovery remains an exciting field with great promise for the future.

Keywords: MRSA, multidrug resistance, staphylococcal infections, drug discovery.

1. INTRODUCTION: THE THERAPEUTIC CHALLENGE

Antibiotics have been a valuable weapon against bacterial in-fections for over a half century. However, the overuse and misuse of antimicrobials have contributed to the occurrence of multi-antibiotic resistance, a collection of phenomena that constitute one of the leading threats to public health with steadily increasing rates. As a result, each year over 13 million deaths worldwide can be attributed to the emergence of new infections or to the re-emergence of previously well-controlled infectious diseases.

Methicillin-resistant Staphylococcus aureus (MRSA) is a causa-tive agent of high mortality, prolonged hospitalization and increas-ing healthcare costs. MRSA infections account for up to 50% of both nosocomial and community-associated staphylococcal infec-tions [1, 2]. The term "superbugs", often applied to generally resis-tant pathogens, was originally coined to describe MRSA. MRSA incidence rates as well as the epidemiologically-related trends have been monitored very closely during the last decade. Despite the downward trend in mortality in several European countries, MRSA still remains a globally significant public health threat [3, 4]. In addition to MRSA, VRSA (vancomycin resistant S. aureus) and VISA (vancomycin intermediate S. aureus) strains also pose an important threat to second-line treatments for MRSA. The first

*Address correspondence to this author at the Torrey Pines Institute for

Molecular Studies, Port St. Lucie, FL, USA; Wellman Center for Pho-

tomedicine, Massachusetts General Hospital, Boston MA, USA; E-mail: [email protected]

report of VRSA in Europe was published this year from Portugal [5]. Resistance to linezolid and daptomycin has also been docu-mented [6, 7].

Colonization with S. aureus occurs in the anterior nares but it can also occur in the throat, axilla, respiratory tract, skin and soft tissues, perineum and rectum [8-11]. The transmission of S. aureus occurs by “person to person” contact as well as via fomites and colonized surfaces [12-15]. Since S. aureus naturally colonizes the human body, bacteria found to be involved in wound infections usually come from the endogenous flora [16]. Bacterial penetration into tissue is permitted by invasive procedures used in patient care [17, 18]. Foreign body-related infections account for a notable oc-currence rate of MRSA infections in healthcare settings [19]. Medi-cal devices such as catheters or artificial prostheses serve as the major causative factor for the incidence of infections in the major organs of the body that have particularly poor prognosis.

The prevalence of MRSA infections as well as their lethality is frequently attributed to virulence factors that can be classified as both secreted products and as structural features of the bacteria with specific roles in the infection process. The generally accepted belief is that MRSA strains are generated by the insertion of a mobile genetic element called the “Staphylococcal Cassette Chromo-some mec” (SCCmec) through horizontal gene transfer. The mecA gene encodes the penicillin-binding protein 2A (PBP2A) that has a decreased affinity for �-lactam antibiotics. SCCmec is flanked by cassette chromosome recombinase genes (ccrA/ccrB or ccrC) that permit intra- and inter-species transmission of the genetic element.

Page 3: Therapeutic options Staphylococcus

New Treatments for Resistant Staphylococci Current Pharmaceutical Design, 2015, Vol. 21, No. 16 2059

Resistance to vancomycin is mediated by the acquisition of the VanA phenotype from Enterococcus sp. strains through the Tn1546 transposon carrying the vanA gene [20].

Community-associated MRSA (CA-MRSA) is caused by strains that are genetically distinguished from hospital-associated MRSA (HA-MRSA). CA-MRSAs are more virulent than typical HA-MRSA probably due to the fact that many of them have been found to carry genes for Panton-Valentine Leukocidin (PVL) a virulence factor associated with MRSA infections [1, 21-23]. In CA-MRSA, mecA is carried within SCCmec type IV, which is dis-tinct from the SCCmec types I, II, and III that are typically found in HA-MRSA [24-26]. In fact, CA-MRSA strains have now been found to display enhanced virulence causing severe infections and they appear to spread faster than HA-MRSA [27].

A plethora of disease states are caused by MRSA and most of them are quite challenging to treat. MRSA is found to be among the most frequently identified pathogens causing pneumonia, and is associated with increased morbidity and mortality rates [28, 29], accounting for 20%–40% of all hospital-acquired pneumonia (HAP) and ventilator-associated pneumonia (VAP) [30-32]. Skin and soft tissue infections (SSTIs), such as diabetic MRSA wound infections, lead to increased costs, prolonged healing time and poor prognosis [33, 34]. MRSA infections may also be associated with persistent or recurrent bacteremia especially in long-term hemo-dialysis patients with renal disease [35-38]. Persistent MRSA bac-teremia is associated with infective endocarditis eventually leading to heart failure and even death [39-42]. Bone infections constitute another difficult-to-treat clinical entity, with diabetes and peripheral vascular disease predisposing patients to MRSA osteomyelitis [43-45]. Last but not the least, toxic shock syndrome, a life-threatening condition gathered the public attention because of its association with the use of tampons colonized with S. aureus during use by the female population [46].

2. CLINICALLY AVAILABLE TREATMENTS

The use of penicillin was rapidly associated with the spread of resistant S. aureus strains in both the hospital and community set-tings soon after its introduction in the 1940s [1]. Methicillin was introduced in 1961 to treat penicillin-resistant S. aureus isolates, but methicillin-resistant strains soon made their appearance through the acquisition and expression of a chromosomally encoded gene for PBP2A. Hyper-virulent staphylococcal strains have evolved resis-tance against the most renowned antibiotic classes commonly used in clinical practice. The diversity in the different mechanisms of resistance that have evolved results from the interplay between microbial physiology, the nature of the genetic elements, as well as the landscape in which those resistance mechanisms occur (Table

1). Especially, mutations and genetic rearrangement carried out by mobile genetic elements are found among the most common resis-

tance mechanisms of Staphylococcus. The US Food and Drug Ad-ministration (FDA) (Table 2) has approved a number of antibacte-rial agents to treat MRSA infections.

Vancomycin has been the gold standard for the treatment of MRSA infections for decades and still remains the first-line option for the clinical treatment of multiple diseases caused by MRSA. Failure rates of vancomycin treatment of staphylococcal bactere-mia, endocarditis and pneumonia are now emerging, raising ques-tions about the supremacy of vancomycin in the management of MRSA infections [45, 54-56]. MRSA with reduced vancomycin susceptibility often leads to VISA or heterogeneous VISA (hVISA) with documented worse clinical outcomes [56]. Teicoplanin is a semi-synthetic anti-MRSA drug which has been known since the 1980s. It has similar effects to vancomycin, yet is more cost-effective and less toxic. Initially approved in Europe, teicoplanin was subsequently approved in many other countries, but is not yet approved by the FDA due to its unusual pharmacokinetic properties [57].

Linezolid is a bacteriostatic synthetic drug against MRSA that also exhibits in vitro efficacy against VRSA and VISA [58]. Pa-tients treated with Linezolid are more likely to have better out-comes than those treated with vancomycin for MRSA pneumonia and especially for MRSA VAP; according to a variety of clinical trials this is most likely due to its enhanced ability to penetrate tis-sues [59]. Despite the fact that linezolid is a well-established antibi-otic for the treatment of serious MRSA infections, the reported cases of linezolid-resistant S. aureus worldwide raise concerns about its possible overuse in clinical practice [60].

Daptomycin exhibits bactericidal activity and reduced toxicity and is often used to treat multi-resistant pathogens [61]. Daptomy-cin was first approved by the US FDA in 2003 for the treatment of MRSA SSTIs and in 2006 for the treatment of bacteremia and right-sided endocarditis. It is not recommended for the treatment of respi-ratory infections due to its inactivation by pulmonary surfactant [58]. Daptomycin-resistant S. aureus strains have been reported due to failures in treatment. These failures may be either attributed to unsuccessful therapy with daptomycin or to inappropriate surgical intervention [62].

Topically administered mupirocin is a naturally occurring anti-biotic, introduced into clinical practice in 1985, but only two years after its introduction there was the first report of the isolation of mupirocin-resistant S. aureus strains [63, 64].

Fusidic acid is a bacteriostatic drug that has been approved for topical use worldwide to treat bacterial skin infections since the 1960s, but is not yet approved by the US FDA. Fusidic acid still remains a valuable treatment option for skin infections with a high therapeutic potential and low resistance rates [61, 65].

Table 1. S. aureus resistance mechanisms to antibiotics according to their genetic base.

Resistance Mechanism Genetic Basis Target-Phenotype References

�-lactamase production Plasmid-encoded bla genes penicillins [47]

Acquired low-affinity PBP2A mecA gene in SCCmec cassettes �-lactams (MRSA) [48]

Modified peptidoglycan target vanA and vanB genes on transposons glycopeptides (VRSA) [49]

Altered cell wall physiology Chromosomal mutations glycopeptides [50]

Ribosomal target modification G2576T mutation linezolid [51]

Ribosomal methylation at A2503 residue Plasmid-encoded cfr genes linezolid [52]

Amino acid substitutions Chromosomal mutations in the genes gyrA, grlA and gyrB fluoroquinolones [53]

Page 4: Therapeutic options Staphylococcus

2060 Current Pharmaceutical Design, 2015, Vol. 21, No. 16 Magana et al.

Table 2. Current therapeutic options for MRSA infections.

Drug Structure Route Activity

Range

Reg.

Status Class Target

Medical

conditions

treated

Daptomycin PO Gram-

positive

FDA

(2003) Lipopeptide

cell

wall

SSTIs

bacteremia

endocardi-

tis

Vancomycin IV/PO broad

spectrum

FDA

(1964) Glycopeptide

cell

wall

bacteremia

endocardi-

tis osteo-

myelitis

septic

arthritis

meningitis

Teicoplanin IV/IM Gram-

positive - Glycopeptide

cell

wall SSTIs

Retapamulin TD Gram-

positive

FDA

(2007) Pleuromutilin

protein

synthe-

sis

SSTIs

Telavancin IV Gram-

positive

FDA

(2009)

Lipoglycopept

ide/vancomy-

cin derivative

cell

wall

SSTIs

HAP

Page 5: Therapeutic options Staphylococcus

New Treatments for Resistant Staphylococci Current Pharmaceutical Design, 2015, Vol. 21, No. 16 2061

(Table 2) Contd….

Drug Structure Route Activity

Range

Reg.

Status Class Target

Medical

conditions

treated

Telithromycin PO broad

spectrum

FDA

(2004) Macrolide

protein

synthe-

sis

CAP

Oritavancin IV Gram-

positive

FDA

(2014) Glycopeptide

cell

wall SSTIs

Dalbavancin IV broad

spectrum

FDA

(2014) Glycopeptide

cell

wall SSTIs

Tedizolid

phosphate IV/PO

Gram-

positive

FDA

(2014)

Oxazolidi-

none

protein

synthe-

sis

SSTIs

Page 6: Therapeutic options Staphylococcus

2062 Current Pharmaceutical Design, 2015, Vol. 21, No. 16 Magana et al.

(Table 2) Contd….

Drug Structure Route Activity

Range

Reg.

Status Class Target

Medical

conditions

treated

Linezolid IV/PO broad

spectrum

FDA

(2000)

Oxazolidi-

none

protein

synthe-

sis

pneumo-

nia

meningitis

SSTIs

Clindamycin IV/PO/

TD

broad

spectrum

FDA

(1970) Lincosamide

protein

synthe-

sis

SSTIs

Sepsis

Necrotiz-

ing infec-

tions

Gentamicin TD broad

spectrum

FDA

(1964)

Aminoglyco-

side

protein

synthe-

sis

SSTIs

Bacitracin TD/IM broad

spectrum

FDA

(1973) Polypeptide

cell

wall SSTIs

Fusidic acid TD Gram-

positive -

Natural prod-

uct

protein

synthe-

sis

SSTIs

Mupirocin TD broad

spectrum

FDA

(1987)

Natural prod-

uct

protein

synthe-

sis

SSTIs

Ceftaroline

fosamil IV

broad

spectrum

FDA

(2010) Cephalosporin

cell

wall

SSTIs

CAP

Page 7: Therapeutic options Staphylococcus

New Treatments for Resistant Staphylococci Current Pharmaceutical Design, 2015, Vol. 21, No. 16 2063

(Table 2) Contd….

Drug Structure Route Activity

Range

Reg.

Status Class Target

Medical

conditions

treated

Doxycycline PO broad

spectrum

FDA

(1967) Tetracycline

protein

synthe-

sis

SSTIs

Minocycline PO broad

spectrum

FDA

(1971) Tetracycline

protein

synthe-

sis

SSTIs

Quinopristin/

Dalfopristin IV

Gram-

positive

FDA

(1999) Streptogramin

protein

synthe-

sis

SSTIs

Tigecycline IV broad

spectrum

FDA

(2005) Tetracycline

protein

synthe-

sis

SSTIs

CAP

Compli-

cated

intra-

abdominal

infections

Nadifloxacin TD broad

spectrum

FDA

(2009)

Fluoroquinolo

ne

DNA

synthe-

sis

SSTIs

(IV: intravenous, PO: per os, TD: transdermal, IM: intramuscular, SSTIs: skin and soft tissue infections, HAP: hospital-acquired pneumonia, CAP: community-acquired pneumonia)

Bacitracin is a complex of cyclic peptide antibiotics produced by the Tracy-I strain of Bacillus subtilis. The zinc salt of bacitracin is combined with other topical antibiotics (polymyxin B and neo-mycin) to form an ointment, that is used for topical treatment of a variety of localized skin and eye infections [66].

Fifth-generation cephalosporins (ceftobiprole and ceftaroline fosamil) have demonstrated extensive activity against Gram-positive bacteria. Ceftaroline fosamil was approved by the FDA in 2010 and by the European Medical Agency (EMA) in 2012 for the treatment of MRSA SSTIs and community-acquired pneumonia (CAP), and is the only cephalosporin active against hVISA and VRSA [65, 67]. On the other hand, ceftobiprole was rejected by the

FDA in 2008 and by EMA in 2010 due to invalid data provided by the clinical studies [67]. Especially, the FDA and EMA Good Clinical Practice (GCP) inspections discovered that there was a scarcity in monitoring the studies and in the study conduct [68].

Clindamycin has a primarily bacteriostatic effect and is useful for the treatment of a number of infections [58]. Inducible macrol-ide-lincosamide-streptogramin resistance caused by erythromycin limits its broad use for the treatment of empiric MRSA infection [69, 70]. Doxycycline and minocycline are broad-spectrum tetracy-clines with efficient oral bioavailability and tissue penetration. They present an efficient alternative for the treatment of non-invasive MRSA infections that do not require intravenous (IV) therapy [71-

Page 8: Therapeutic options Staphylococcus

2064 Current Pharmaceutical Design, 2015, Vol. 21, No. 16 Magana et al.

73]. Minocycline does not exhibit a good safety profile and for this reason it was added to the FDA Adverse Event Reporting System (AERS) in 2009 [74]. Tigecycline exhibits bacteriostatic efficacy against MRSA. It is structurally similar to the tetracyclines but not efficient when used for the treatment of HAP or VAP since it has demonstrated low therapeutic effects [75].

Quinupristin-dalfopristin is used for the treatment of life-threatening infections but it is rarely applied in clinical practice due to its poor safety profile and the availability of better treatment options [58].

Dalbavancin and oritavancin are semi-synthetic compounds with high efficacy against bacterial skin infections. Dalbavancin belongs to the first-line antibiotics for the treatment of diabetic foot infections compared to vancomycin, daptomycin and linezolid [76]. Oritavancin is a newly approved lipoglycopeptide with antibacterial activity [77]. Oritavancin exhibits bactericidal activity against an array of antibiotic resistant isolates of S. aureus including methicil-lin-susceptible S. aureus (MSSA), VRSA, VISA, daptomycin non-susceptible S. aureus and MRSA in planktonic and biofilm species [77]. Both dalbavancin and oritavancin are active against MRSA and have a long half-life thus requiring a shorter administration regimen which allows them to be a promising outpatient option [78]. Telavancin is another recently-approved antibiotic that exhib-its concentration-dependent bactericidal activity against MRSA. It is as active as vancomycin and other antistaphylococcal penicillins in the treatment of SSTIs and hospital-acquired pneumonia [79].

Nadifloxacin is a topical fluoroquinolone that acts by inhibiting DNA gyrase and it has been shown to be effective against a broad-spectrum of bacteria including MRSA and coagulase-negative staphylococci. It is approved for use in the treatment of acne vul-garis and other skin infections in Japan where it has been used for decades representing good safety and efficacy profiles [80].

3. THE PRECLINICAL PIPELINE

The increased mortality rates as well as the variety of estab-lished resistance mechanisms constitute a therapeutic challenge, and underline the urgent need for more effective alternatives for MRSA treatment. There is a wealth of preclinical leads against MRSA stemming from validated targets and a rapidly growing pipeline of molecules arising from preliminary investigations based on the re-positioning of underexplored or newly discovered targets and approaches. Table 3 summarizes lead molecules that represent advances in the preclinical pipeline for staphylococcal infections. Antimicrobial peptides (AMPs) and natural derivatives exhibit strong antistaphylococcal activity with unique and rather unex-plored characteristics that could make them potential therapeutic agents [81, 82].

Two lipopeptide molecules have a unique mode of action com-pared with any other approved lipopeptides. MX-2401 is a semi-synthetic analog of the naturally occurring lipopeptide, amphomy-cin [83]. MX-2401, unlike daptomycin, is not affected in vitro by the presence of lung surfactant and has shown to be active in vivo in a bronchial-alveolar pneumonia mouse model [84]. Tripropeptin

C (TPPC) exhibits high efficacy against antibiotic-resistant strains such as MRSA and VRE [85]. Drugs targeting small molecular substrates usually delay the emergence of resistant bacteria. TPPC inhibits enzymatic reactions through binding to substrates repre-senting a promising novel class of antibiotics [86].

Although the utility of tetracyclines has been reduced since the onset of bacterial resistance, two newly introduced agents of this class, eravacycline and omadacycline exhibit good oral bioavail-ability and activity which is not impaired by the lung surfactant [67]. Plazomicin is a next-generation aminoglycoside that has demonstrated in vitro synergistic activity with daptomycin or cefto-biprole against MSRA, VISA, hVISA and VRSA [87]. Fluoroqui-nolones are of particular interest as many antibiotics from this class

are currently in clinical trials. Delafloxacin eradicates quinolone-resistant or susceptible MRSA as it becomes highly potent at re-duced pH of tissue following inflammation [88]. JNJ-Q2 appears to be among the most promising new members of this class with low potential resistance and increased potency compared to other marketed fluoroquinoles [89].

Iclaprim is a broad-spectrum drug that is in Phase III clinical trials and was found to be effective against MRSA exhibiting high cure rates along with a well tolerated toxicity profile [90]. The EMA accepted for review a marketing authorization application for iclaprim in 2008 but, the Anti-infective Drugs Advisory Committee of the FDA requested additional clinical efficacy data [91].

4. EMERGING NON-ANTIBIOTIC APPROACHES

A number of preclinical studies have been conducted utilizing natural viral predators of bacteria, bacteriophages. The use of bacte-riophage (phage) therapy has been tested for the elimination of multidrug resistant infections. Special emphasis has been placed on a class of highly evolved molecules, the bacteriophage lytic en-zymes (lysins) that digest the bacterial cell wall. Small quantities of purified recombinant lysin were found to cause rapid and many log-fold lyses of specific Gram-positive bacteria [92]. After replicating within the bacterial host, the bacteriophage must exit the bacterial cells to disseminate. The lytic enzymes have been refined by evolu-tion to serve this purpose [93, 94]. Lysins target one of the four major peptidoglycan bonds, thus weakening the cell wall. They can function either as an endo-beta-N-acetylglucosaminidase or an N-acetylmuramidase with activities targeted against the sugar moiety of the bacterial wall, as an endopeptidase acting on the peptide moiety, or as an N-acetylmuramoyl-L-alanine amidase that hydro-lyzes the amide bond connecting the glycan strand and peptide moieties [93]. Although lysins have been well known since 1940s, the rapid industrial advancement of antibiotics shifted the focus away from them.

Emerging infections have triggered a thorough reexamination of phage therapy in Russia [95]. Phage-based therapies were devel-oped in the 1930s in Eastern Europe (Russia and Poland) to treat a vast array of pathogenic microbes [96]. The first FDA approval of phage therapy was granted in 2006, for a cocktail of six individu-ally purified phages to serve as a therapeutic alternative for meat and poultry products contaminated with Listeria monocytogenes. This was the first time FDA approved the use of a phage prepara-tion as a food additive [92]. Moreover, whole phage derived lysins have been evaluated for their potential therapeutic effect. Phage lysins have been used to control a wide range of pathogens in vitro such as Group A Streptococci, Streptococcus pneumoniae, Bacillus anthracis, Enterococcus faecalis and S. aureus [97-101]. Lysin PlyC is potent in killing logarithmically grown Streptococcus pyo-genes at nanomolar concentrations [102].

The bacteriolytic activity of lysins has been effective in control-ling multidrug resistant Gram-positive bacteria in animal studies, with a selective effect on mucosal surfaces and in blood, infections often without a demonstrated immune response [103] and most importantly with no evidence of lysin inactivation by blood con-stituents. Lysin Cpl-1 (a muramidase that binds to choline) for ex-ample, exhibited in vivo activity in a penicillin-resistant pneumo-coccal bacteremia mouse model [93, 104].

The key advantages of phages over antibiotics have been sum-marized including specificity for the pathogen without documented effects on the normal flora nor the microbiome, low possibilities for resistance development, and the unique ability to eradicate coloniz-ing pathogens on mucosal surfaces.

Quorum sensing, secretion systems, as well as cell adhesion pathways have been extensively validated as potential targets but with very few preclinical studies available [104-106]. Bacterial cell

Page 9: Therapeutic options Staphylococcus

New Treatments for Resistant Staphylococci Current Pharmaceutical Design, 2015, Vol. 21, No. 16 2065

Table 3. Promising compounds currently in clinical trials.

Name Structures Class Target/ MOA Reg. Status

Brilacidin

(PMX-30063)

1224095-98-0

Drug belonging to

the ‘defensin-

mimetics’ class of

antibiotics

cell membrane Phase II com-

pleted

TD-1792

(Theravance)

1393900-12-3

Chimeric molecule

from the linking of

vancomycin to a

cephalosporin

cell membrane Phase II

XF-73

718638-68-7 Porphyrin cell membrane Phase II

Radezolid

(RX1741)

869884-78-6

Oxazolidinone protein synthesis Phase II

Omadacycline

(PTK-0796)

389139-89-3

Aminomethylci-

cline protein synthesis Phase III

Eravacycline

(TP-434)

1207283-85-9

Aminomethylci-

cline protein synthesis Phase III

Plazomicin

(ACHN-490)

1154757-24-0

Aminoglycoside protein synthesis Phase III

GSK1322322

1152107-25-9

Peptide deformy-

lase inhibitor protein synthesis Phase II

Page 10: Therapeutic options Staphylococcus

2066 Current Pharmaceutical Design, 2015, Vol. 21, No. 16 Magana et al.

(Table 3) Contd….

Name Structures Class Target/ MOA Reg. Status

BB-83698

325795-25-3 Oxadiazole

Peptide de-

formylase in-

hibitors

Phase I

Delafloxacin

(RX3341)

189279-58-1

Fluoroquinolone DNA gyrase

inhibitor Phase III

Avarofloxacin

JNJ-Q2

878592-87-1

Fluoroquinolone DNA gyrase

inhibitor

Phase III/Fast

track

Iclaprim

192314-93-5 Diaminopyrimidine DHFR inhibitor withdrawn

surfaces are of utmost importance for bacterial integrity and viabil-ity. The pathways and the enzymes that catalyze wall teichoic acid (TarO, TarA) and lipoteichoic acid (LtaA, LtaS) synthesis have been identified and exploited as plausible discovery targets [107].

Advances in the field of host-directed therapeutic strategies could eventually lead to the elimination of infections caused by MRSA. The innate immune system constitutes a complex defense mechanism against various microbial threats and from this perspec-tive it has become a promising target for the development of thera-peutics to control such infections [108]. Antistaphylococcal vac-

cines and immunoglobulins have been reported to be effective in vitro; although, both active and passive immunizations have failed to show efficient results in human studies [105]. The complex pathogenic mechanisms of S. aureus represent the major challenge leading to the failure of passive immunization strategies [109]. Monovalent vaccines failed to express sufficient efficacy in humans due to the partial protection they offered. To date, the vaccines that have been so far tested in humans targeted only single antigens, and ultimately led to inefficient antibody and cytokine production. Ide-ally, an antistaphylococcal vaccine should protect against the wide range of S. aureus toxins and immune evasion factors. Antibody-mediated protection is a significant factor that is clearly affected by the complex staphylococcal evasion factors that hamper the im-mune response of the host. Components of the host’s immune sys-tem may play a significant role in the battle against resistance. Pep-tides, complement fragments as well as cells of humoral immunity

and their derivatives could very well be in practice in near future always considering the complexity inherent to host-pathogen inter-actions [108, 110]. Peptides and synthetic analogs may be useful adjuvants for an increase in antibody titers and their efficacy lead-ing to the activation of both cell-mediated (opsonophagocytosis) and also humoral immunity. The design of proper adjuvant formu-lations through a more efficient combination of different antigens, the elucidation of various staphylococcal factors that inhibit the host immune response and the identification of the particular vul-nerabilities of the pathogen could ultimately lead to the develop-ment of improved multivalent antistaphylococcal vaccines [111].

5. NEW APPROACHES

Staphyloccocal drug efflux systems are potentially attractive targets, but they have never been systematically explored, partially due to the fact that their role in the physiology of the pathogen is far more complicated than just a central resistance mechanism to an-timicrobials. Efflux systems are membrane transporter proteins with classes identified as effluxing tetracyclines and fluoroquinolones in S. aureus [112, 113]. Multi-antibiotic resistance is attributed to the chromosomally encoded Major Facilitator Superfamily (MFS) in-cluding NorA, NorB, NorC, MdeA, the mepRAB, Multidrug And Toxic compound Extrusion (MATE) family, and the SepA of the SMR (small multidrug resistance) Family [114, 115]. Plasmid en-coded systems such as the QacA, QacB and Tet(K) that function as tetracycline-divalent metal complex/H

+ antiporters are contributing

Page 11: Therapeutic options Staphylococcus

New Treatments for Resistant Staphylococci Current Pharmaceutical Design, 2015, Vol. 21, No. 16 2067

in antibiotic efflux [116, 117]. These systems have a broad and overlapping substrate specificity including quinolones, tetracy-clines, monovalent and divalent antimicrobial cations, as well as plant secondary metabolites [118]. Efflux inhibition is considered a viable strategy for enhancing antibiotic efficacy. The discovery of small molecules that block efflux systems is a rapidly expanding field. The efforts to discover these small molecules have yielded a number of natural and synthetic staphylococcal efflux modulators [119, 120]. Although the concept of enhancing the utility of antibi-otics by employing efflux inhibitors appears appealing, it remains challenging in terms of clinical implementation [121, 122].

Photodynamic therapy (PDT) is a light-based technology platform, which uses harmless visible light in combination with non-toxic photosensitizers (PS) to control infections. Historically, PDT has had a prominent role in the cure of many serious diseases like cancer, producing reactive oxygen species (ROS) that non-specifically kill cells. This mechanism is the reason PDT is cur-rently being investigated as an alternative therapeutic approach for localized infections as resistance to ROS is unlikely to develop [123, 124]. Most often, the molecules tested as antimicrobial PSs are organic, aromatic dyes with a high degree of electron delocali-zation (porphyrins, halogenated xanthenes, chlorins, bacteriochlo-rins, phthalocyanines, perylenequinones, cationic fullerenes and psoralens) [125, 126]. The molecules should be designed to have affinity to bacterial cells and not to host cells, and methods to de-liver light to all parts of the body via fiberoptics have now been designed. Phenothiazinium dyes (methylene blue (MB), toluidine blue (TBO)) are the only clinically approved antimicrobial PS [127]. Phenothiazenium PSs are physicochemically similar to the natural product, the antibacterial alkaloid berberine, a well-characterized substrate of the NorA efflux system in S. aureus and other Gram-positive bacteria [118, 128]. It has been demonstrated that phenothiazinium PSs are substrates of microbial efflux systems and small molecule efflux pump inhibitors can enhance their photo-toxic effect in a range of pathogens including S. aureus [129]. This synergistic discovery platform along with the therapeutic efficacy of visible light, without the addition of exogenous PS, are under investigation for preclinical development in localized staphylococ-cal infections [126].

With the advent of the nanotechnology revolution, antistaphylo-coccal nanotechnology-based therapeutic applications are being intensively explored with an emphasis on the discovery of novel antimicrobial nano-structures or employing nanoparticles for deliv-ery purposes [130-132]. Nanotechnology by definition refers to the design, development and application of materials in the nano-scale range (< 100nm). The physicochemical properties of nanoparticles (small size and high volume-to-surface ratio) allow them to gain access to a variety of biologic structures and systems. Nanosized materials are quite amenable to manipulation of their size, shape, and chemical characteristics. This feature offers multiple opportuni-ties to engineer nanoparticles as vehicles for therapeutic or diagnos-tic agents in medical applications, including targeted drug delivery, gene therapy and cell labeling [133-136]. Nanoparticles have been deployed in a variety of localized staphylococcal infections and are under investigation for delivering antibiotics, new experimental therapeutics with promising in vitro MRSA efficacy profiles [137-141].

6. BIOINFORMATICS AS A TOOL FOR DRUG DISCOV-ERY IN S. AUREUS

Microbiology research in the post-genomic era has inevitably involved bioinformatics (genomics and proteomics) as a tool to investigate the physiology of microbes, micro and macro-evolution, dynamics of mutations and adaptation within different host popula-tions. As sequencing techniques (ST) mature, fuller microbial ge-nomes will become available and the need for the development of powerful bioinformatics tools for the analysis of the growing data is

imperative. As of November 2014, a total of 4,193 entries (full or draft genomes) regarding S. aureus were available at the GenBank database [142]. Applying MLST (Multi Locus Sequence Typing), a total of 2,832 ST sequences have already been identified for S. aureus which are grouped in multiple clonal complexes (CC), un-derlying the extreme genetic diversity for this pathogen [143]. The study of MRSA genomics in silico offers valuable insights into the development of novel therapeutic interventions.

One research direction involves the study of transmission and molecular tracing of various strains –molecular epidemiology of MRSA strains- within human or animal populations or between human and animals. Transmission of such strains from animals to humans may be associated with novel virulence genes and homo-logues [144, 145]. Therefore, the study of MRSA molecular epide-miology has a direct impact on the design of novel drugs since it offers an insight into the diversity of existing therapeutic targets. Studies pertaining to the genetic sequence of specific MRSA strains (particularly USA300) have given insight into the origin, the trans-mission route and networks of this clone in the US delineating its diversity and spread [146-148]. Other studies have pointed out the existence of MRSA strains within animals whereas extensive re-search has been conducted using other STs and CC in different geographic areas [149-162]. Shepheard et al. analyzing strains from human and animals showed a total of 15 historical switching events and at least two S. aureus CC (CC25 and 29) which have arisen from animal-associated ancestors [163].

Another intervention concerns the identification of novel viru-lence genes in MRSA. Phylogenetics have been applied in the iden-tification of novel virulence genes and toxins as well the exchange of genetic material between different bacterial species leading to the development of novel phenotypes in MRSA [145, 164, 165]. Study-ing the diversity of known virulence factors as well as the identifi-cation of conserved regions may provide a pipeline of potential novel drug targets [166-172]. Likewise, the study of the interaction between MRSA and hosts (animal or human) could help in pharma-ceutical design [173-175]. A study designed to identify novel tar-gets for the development of drugs specific to MRSA, employed a genome-wide scan for balancing selection in S. aureus strains iden-tifying ninety nine candidate genes at sixty two different genomic loci [176]. An alternative approach for identification of novel therapeutic targets involves subtractive genome analysis. A recent study on MRSA ST398 and MRSA 252 revealed six and twenty-one possible therapeutic targets, respectively, for future drug devel-opment [177].

Overall, the employment of bioinformatics may give insight into the evolution and molecular epidemiology of MRSA, as well as the identification of novel therapeutic targets for drug discovery.

7. SYNOPSIS AND PERSPECTIVES

The treatment of resistant MRSA strains is complicated both in systemic and Acute Bacterial Skin and Skin Structure Infections (ABSSSI). The clinical need for alternative therapeutic interven-tions targeting multi-antibiotic resistant pathogens remains high as: (a) the evolutionary adaptability of staphylococcal strains to antibi-otic pressure has been proven to be a successful survival strategy for the pathogen [178]; (b) the potent current major systemic anti-biotics (linezolid, daptomycin, vancomycin) and topical mupirocin have all been found to have at least some (higher or lower) inci-dence of resistance or non-susceptibility [178]; (c) the use of con-ventional broad-spectrum antibiotics is a risk factor for elevated resistance in healthcare facilities [179]; (d) the recent FDA ap-proval of oxazolidinones for ABSSSI [180] as well as the com-pounds in the preclinical development pipeline are promising but may provide only a temporary answer; (e) the economic burden of treatment persists [181].

The empirical use of antibiotics in patients with S. aureus infec-tions should be limited in clinical settings. Monitoring of patient

Page 12: Therapeutic options Staphylococcus

2068 Current Pharmaceutical Design, 2015, Vol. 21, No. 16 Magana et al.

compliance and susceptibility testing of the isolates to antibiotics used in current therapeutics should be taken into consideration in order to guide treatment.

CONFLICT OF INTEREST

This article content has no conflict of interest.

ACKNOWLEDGEMENTS

George P Tegos was supported by the US NIH grant 5U54MH084690-02 (CDP1) and by the US DTRA contract HDTRA1-13-C-0005 and Michael R Hamblin was supported by US NIH grant R01AI050875.

REFERENCES

[1] Chambers HF. The changing epidemiology of Staphylococcus

aureus? Emerg Infect Dis 2001; 7: 178-82. [2] Bassetti M, Righi E. Multidrug-resistant bacteria: what is the

threat? Hematology Am Soc Hematol Educ Program 2013; 2013: 428-32.

[3] ECDC, editor. Annual Epidemiological Report 2013. Reporting on 2011 surveillance data and 2012 epidemic intelligence data.

Stockholm: ECDC; 2013. [4] WHO, editor. Antimicrobial resistance: global report on

surveillance 2014. [5] Friaes A, Resina C, Manuel V, et al. Epidemiological survey of the

first case of vancomycin-resistant Staphylococcus aureus infection in Europe. Epidemiol Infect 2014; 1-4.

[6] Locke JB, Zuill DE, Scharn CR, et al. Linezolid-Resistant Staphylococcus aureus Strain 1128105, the First Known Clinical

Isolate Possessing the cfr Multidrug Resistance Gene. Antimicrob Agents Chemother 2014; 58: 6592-8.

[7] Cavalcante FS, Ferreira Dde C, Chamon RC, et al. Daptomycin and methicillin-resistant Staphylococcus aureus isolated from a

catheter-related bloodstream infection: a case report. BMC Res Notes 2014; 7: 759.

[8] Kluytmans J, van Belkum A, Verbrugh H. Nasal carriage of Staphylococcus aureus: epidemiology, underlying mechanisms, and

associated risks. Clin Microbiol Rev 1997; 10: 505-20. [9] Eveillard M, de Lassence A, Lancien E, et al. Evaluation of a

strategy of screening multiple anatomical sites for methicillin-resistant Staphylococcus aureus at admission to a teaching hospital.

Infect Control Hosp Epidemiol 2006; 27: 181-4. [10] Mertz D, Frei R, Jaussi B, et al. Throat swabs are necessary to

reliably detect carriers of Staphylococcus aureus. Clin Infect Dis 2007; 45: 475-7.

[11] Peters PJ, Brooks JT, Limbago B, et al. Methicillin-resistant Staphylococcus aureus colonization in HIV-infected outpatients is

common and detection is enhanced by groin culture. Epidemiol Infect 2011; 139: 998-1008.

[12] Scott E, Duty S, Callahan M. A pilot study to isolate Staphylococcus aureus and methicillin-resistant S aureus from

environmental surfaces in the home. Am J Infect Control 2008; 36: 458-60.

[13] Miller LG, Diep BA. Clinical practice: colonization, fomites, and virulence: rethinking the pathogenesis of community-associated

methicillin-resistant Staphylococcus aureus infection. Clin Infect Dis 2008; 46: 752-60.

[14] Champion A, Goodwin TA, Brolinson P, et al. Prevalence and characterization of methicillin-resistant. Ann Clin Microbiol

Antimicrob 2014; 13: 33. [15] Uemura E, Kakinohana S, Higa N, Toma C, Nakasone N.

Comparative chracterization of Staphylococcus aureus isolates from throats and noses of healthy volunteers. Jpn J Infect Dis 2004;

57: 21-4. [16] Golubchik T, Batty EM, Miller RR, et al. Within-host evolution of

Staphylococcus aureus during asymptomatic carriage. PLoS One 2013; 8: e61319.

[17] Jacobs A. Hospital-Acquired Methicillin-Resistant Staphylococcus aureus: Status and Trends. Radiol Technol 2014; 85: 623-48.

[18] Barkin JA, Miki RA, Mahmood Z, Landy DC, Owens P. Prevalence of methicillin resistant Staphylococcus aureus in upper

extremity soft tissue infections at Jackson Memorial Hospital, Miami-Dade County, Florida. Iowa Orthop J 2009; 29: 67-73.

[19] von Eiff C, Jansen B, Kohnen W, Becker K. Infections associated

with medical devices: pathogenesis, management and prophylaxis. Drugs 2005; 65: 179-214.

[20] Weigel LM, Clewell DB, Gill SR, et al. Genetic analysis of a high-level vancomycin-resistant isolate of Staphylococcus aureus.

Science 2003; 302: 1569-71. [21] Baba T, Takeuchi F, Kuroda M, et al. Genome and virulence

determinants of high virulence community-acquired MRSA. Lancet 2002; 359: 1819-27.

[22] Naimi TS, LeDell KH, Como-Sabetti K, et al. Comparison of community- and health care-associated methicillin-resistant

Staphylococcus aureus infection. JAMA 2003; 290: 2976-84. [23] Diep BA, Sensabaugh GF, Somboonna N, Carleton HA, Perdreau-

Remington F. Widespread skin and soft-tissue infections due to two methicillin-resistant Staphylococcus aureus strains harboring the

genes for Panton-Valentine leucocidin. J Clin Microbiol 2004; 42: 2080-4.

[24] Wu S, Piscitelli C, de Lencastre H, Tomasz A. Tracking the evolutionary origin of the methicillin resistance gene: cloning and

sequencing of a homologue of mecA from a methicillin susceptible strain of Staphylococcus sciuri. Microb Drug Resist 1996; 2: 435-

41. [25] Hiramatsu K, Cui L, Kuroda M, Ito T. The emergence and

evolution of methicillin-resistant Staphylococcus aureus. Trends Microbiol 2001; 9: 486-93.

[26] Wisplinghoff H, Rosato AE, Enright MC, et al. Related clones containing SCCmec type IV predominate among clinically

significant Staphylococcus epidermidis isolates. Antimicrob Agents Chemother 2003; 47: 3574-9.

[27] Rice LB. The clinical consequences of antimicrobial resistance. Curr Opin Microbiol 2009; 12: 476-81.

[28] Shorr AF, Tabak YP, Gupta V, et al. Morbidity and cost burden of methicillin-resistant Staphylococcus aureus in early onset

ventilator-associated pneumonia. Crit Care 2006; 10: R97. [29] Shorr AF, Haque N, Taneja C, et al. Clinical and economic

outcomes for patients with health care-associated Staphylococcus aureus pneumonia. J Clin Microbiol 2010; 48: 3258-62.

[30] Rubinstein E, Kollef MH, Nathwani D. Pneumonia caused by methicillin-resistant Staphylococcus aureus. Clin Infect Dis 2008;

46 (Suppl 5): S378-85. [31] Hidron AI, Low CE, Honig EG, Blumberg HM. Emergence of

community-acquired meticillin-resistant Staphylococcus aureus strain USA300 as a cause of necrotising community-onset

pneumonia. Lancet Infect Dis 2009; 9: 384-92. [32] Gillet Y, Vanhems P, Lina G, et al. Factors predicting mortality in

necrotizing community-acquired pneumonia caused by Staphylococcus aureus containing Panton-Valentine leukocidin.

Clin Infect Dis 2007; 45: 315-21. [33] Goldstein EJ, Citron DM, Nesbit CA. Diabetic foot infections.

Bacteriology and activity of 10 oral antimicrobial agents against bacteria isolated from consecutive cases. Diabetes Care 1996; 19:

638-41. [34] Tentolouris N, Jude EB, Smirnof I, Knowles EA, Boulton AJ.

Methicillin-resistant Staphylococcus aureus: an increasing problem in a diabetic foot clinic. Diabet Med 1999; 16: 767-71.

[35] Kaech C, Elzi L, Sendi P, et al. Course and outcome of Staphylococcus aureus bacteraemia: a retrospective analysis of 308

episodes in a Swiss tertiary-care centre. Clin Microbiol Infect 2006; 12: 345-52.

[36] Kern WV. Management of Staphylococcus aureus bacteremia and endocarditis: progresses and challenges. Curr Opin Infect Dis 2010;

23: 346-58. [37] Nori US, Manoharan A, Thornby JI, et al. Mortality risk factors in

chronic haemodialysis patients with infective endocarditis. Nephrol Dial Transplant 2006; 21: 2184-90.

[38] Johnson LB, Jose J, Yousif F, Pawlak J, Saravolatz LD. Prevalence of colonization with community-associated methicillin-resistant

Staphylococcus aureus among end-stage renal disease patients and healthcare workers. Infect Control Hosp Epidemiol 2009; 30: 4-8.

[39] Fowler VG, Jr., Miro JM, Hoen B, et al. Staphylococcus aureus endocarditis: a consequence of medical progress. JAMA 2005; 293:

3012-21. [40] Hawkins C, Huang J, Jin N, et al. Persistent Staphylococcus aureus

bacteremia: an analysis of risk factors and outcomes. Arch Intern Med 2007; 167: 1861-7.

Page 13: Therapeutic options Staphylococcus

New Treatments for Resistant Staphylococci Current Pharmaceutical Design, 2015, Vol. 21, No. 16 2069

[41] Baddour LM, Wilson WR, Bayer AS, et al. Infective endocarditis:

diagnosis, antimicrobial therapy, and management of complications: a statement for healthcare professionals from the

Committee on Rheumatic Fever, Endocarditis, and Kawasaki Disease, Council on Cardiovascular Disease in the Young, and the

Councils on Clinical Cardiology, Stroke, and Cardiovascular Surgery and Anesthesia, American Heart Association: endorsed by

the Infectious Diseases Society of America. Circulation 2005; 111: e394-434.

[42] Cooke FJ, Gkrania-Klotsas E, Howard JC, et al. Clinical, molecular and epidemiological description of a cluster of community-

associated methicillin-resistant Staphylococcus aureus isolates from injecting drug users with bacteraemia. Clin Microbiol Infect

2010; 16: 921-6. [43] Valour F, Bouaziz A, Karsenty J, et al. Determinants of

methicillin-susceptible Staphylococcus aureus native bone and joint infection treatment failure: a retrospective cohort study. BMC

Infect Dis 2014; 14: 443. [44] Joel J, Graham SM, Peckham-Cooper A, et al. Clinical results of

linezolid in arthroplasty and trauma MRSA related infections. World J Orthop 2014; 5: 151-7.

[45] Tice AD, Hoaglund PA, Shoultz DA. Risk factors and treatment outcomes in osteomyelitis. J Antimicrob Chemother 2003; 51:

1261-8. [46] Shalaby T, Anandappa S, Pocock NJ, Keough A, Turner A. Lesson

of the month 2: toxic shock syndrome. Clin Med 2014; 14: 316-8. [47] Zapun A, Contreras-Martel C, Vernet T. Penicillin-binding proteins

and beta-lactam resistance. FEMS Microbiol Rev 2008; 32: 361-85. [48] Banerjee R, Gretes M, Basuino L, Strynadka N, Chambers HF. In

vitro selection and characterization of ceftobiprole-resistant methicillin-resistant Staphylococcus aureus. Antimicrob Agents

Chemother 2008; 52: 2089-96. [49] Foucault ML, Courvalin P, Grillot-Courvalin C. Fitness cost of

VanA-type vancomycin resistance in methicillin-resistant Staphy-lococcus aureus. Antimicrob Agents Chemother 2009; 53: 2354-9.

[50] Howden BP, Davies JK, Johnson PD, Stinear TP, Grayson ML. Reduced vancomycin susceptibility in Staphylococcus aureus,

including vancomycin-intermediate and heterogeneous vanco-mycin-intermediate strains: resistance mechanisms, laboratory

detection, and clinical implications. Clin Microbiol Rev 2010; 23: 99-139.

[51] Besier S, Ludwig A, Zander J, Brade V, Wichelhaus TA. Linezolid resistance in Staphylococcus aureus: gene dosage effect, stability,

fitness costs, and cross-resistances. Antimicrob Agents Chemother 2008; 52: 1570-2.

[52] Morales G, Picazo JJ, Baos E, et al. Resistance to linezolid is mediated by the cfr gene in the first report of an outbreak of

linezolid-resistant Staphylococcus aureus. Clin Infect Dis 2010; 50: 821-5.

[53] Kaatz GW, Seo SM. Mechanisms of fluoroquinolone resistance in genetically related strains of Staphylococcus aureus. Antimicrob

Agents Chemother 1997; 41: 2733-7. [54] Chang FY, Peacock JE, Jr., Musher DM, et al. Staphylococcus

aureus bacteremia: recurrence and the impact of antibiotic treatment in a prospective multicenter study. Medicine (Baltimore)

2003; 82: 333-9. [55] Kim SH, Kim KH, Kim HB, et al. Outcome of vancomycin

treatment in patients with methicillin-susceptible Staphylococcus aureus bacteremia. Antimicrob Agents Chemother 2008; 52: 192-7.

[56] Barber KE, Ireland CE, Bukavyn N, Rybak MJ. Observation of "seesaw effect" with vancomycin, teicoplanin, daptomycin and

ceftaroline in 150 unique MRSA strains. Infect Dis Ther 2014; 3: 35-43.

[57] Pea F, Brollo L, Viale P, Pavan F, Furlanut M. Teicoplanin therapeutic drug monitoring in critically ill patients: a retrospective

study emphasizing the importance of a loading dose. J Antimicrob Chemother 2003; 51: 971-5.

[58] 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.

[59] Peyrani P, Wiemken TL, Kelley R, et al. Higher clinical success in patients with ventilator-associated pneumonia due to methicillin-

resistant Staphylococcus aureus treated with linezolid compared with vancomycin: results from the IMPACT-HAP study. Crit Care

2014; 18: R118.

[60] Gu B, Kelesidis T, Tsiodras S, Hindler J, Humphries RM. The

emerging problem of linezolid-resistant Staphylococcus. J Antimicrob Chemother 2013; 68: 4-11.

[61] Kurosu M, Siricilla S, Mitachi K. Advances in MRSA drug discovery: where are we and where do we need to be? Expert Opin

Drug Discov 2013; 8: 1095-116. [62] Humphries RM, Pollett S, Sakoulas G. A current perspective on

daptomycin for the clinical microbiologist. Clin Microbiol Rev 2013; 26: 759-80.

[63] Caffrey AR, Quilliam BJ, LaPlante KL. Risk factors associated with mupirocin resistance in meticillin-resistant Staphylococcus

aureus. J Hosp Infect 2010; 76: 206-10. [64] Simor AE, Stuart TL, Louie L, et al. Mupirocin-resistant,

methicillin-resistant Staphylococcus aureus strains in Canadian hospitals. Antimicrob Agents Chemother 2007; 51: 3880-6.

[65] Castanheira M, Watters AA, Bell JM, Turnidge JD, Jones RN. Fusidic acid resistance rates and prevalence of resistance

mechanisms among Staphylococcus spp. isolated in North America and Australia, 2007-2008. Antimicrob Agents Chemother 2010; 54:

3614-7. [66] 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.

[67] Bassetti M, Merelli M, Temperoni C, Astilean A. New antibiotics for bad bugs: where are we? Ann Clin Microbiol Antimicrob 2013;

12: 22. [68] Basilea, Pharmaceutica, Ltd. Basilea awarded USD 130 million by

arbitral tribunal 2010 Available from: www.basilea.com. [69] Yilmaz G, Aydin K, Iskender S, Caylan R, Koksal I. Detection and

prevalence of inducible clindamycin resistance in staphylococci. J Med Microbiol 2007; 56: 342-5.

[70] Siberry GK, Tekle T, Carroll K, Dick J. Failure of clindamycin treatment of methicillin-resistant Staphylococcus aureus expressing

inducible clindamycin resistance in vitro. Clin Infect Dis 2003; 37: 1257-60.

[71] van Hal SJ, Lodise TP, Paterson DL. The clinical significance of vancomycin minimum inhibitory concentration in Staphylococcus

aureus infections: a systematic review and meta-analysis. Clin Infect Dis 2012; 54: 755-71.

[72] Ruhe JJ, Menon A. Tetracyclines as an oral treatment option for patients with community onset skin and soft tissue infections

caused by methicillin-resistant Staphylococcus aureus. Antimicrob Agents Chemother 2007; 51: 3298-303.

[73] Ruhe JJ, Monson T, Bradsher RW, Menon A. Use of long-acting tetracyclines for methicillin-resistant Staphylococcus aureus

infections: case series and review of the literature. Clin Infect Dis 2005; 40: 1429-34.

[74] FDA [homepage on the Internet]. Potential Signals of Serious Risks/New Safety Information Identified from the Adverse Event

Reporting System (AERS) between January - March 2009. [updated 2014 Oct 07; cited 2011 Mar ]; Available from: http:

//www.fda.gov/Drugs/GuidanceComplianceRegulatoryInformation/Surveillance/AdverseDrugEffects/ucm185260.htm.

[75] FDA [homepage on the Internet]. FDA Drug Safety Commu-nication: Increased risk of death with Tygacil (tigecycline)

compared to other antibiotics used to treat similar infections. [updated 2014 Aug 14; cited 2010 Jan 01]; Available from: http:

//www.fda.gov/Drugs/DrugSafety/ucm224370.htm. [76] Goldstein EJ, Citron DM, Warren YA, et al. In vitro activities of

dalbavancin and 12 other agents against 329 aerobic and anaerobic gram-positive isolates recovered from diabetic foot infections.

Antimicrob Agents Chemother 2006; 50: 2875-9. [77] Belley A, Neesham-Grenon E, McKay G, et al. Oritavancin kills

stationary-phase and biofilm Staphylococcus aureus cells in vitro. Antimicrob Agents Chemother 2009; 53: 918-25.

[78] Tice A. Oritavancin: a new opportunity for outpatient therapy of serious infections. Clin Infect Dis 2012; 54 (Suppl 3): 239-43.

[79] Rubinstein E, Lalani T, Corey GR, et al. Telavancin versus vancomycin for hospital-acquired pneumonia due to gram-positive

pathogens. Clin Infect Dis 2011; 52: 31-40. [80] Narayanan V, Motlekar S, Kadhe G, Bhagat S. Efficacy and Safety

of Nadifloxacin for Bacterial Skin Infections: Results from Clinical and Post-Marketing Studies. Dermatol Ther (Heidelb) 2014; 4:

233-48. [81] Reddy KV, Yedery RD, Aranha C. Antimicrobial peptides:

premises and promises. Int J Antimicrob Agents 2004; 24: 536-47.

Page 14: Therapeutic options Staphylococcus

2070 Current Pharmaceutical Design, 2015, Vol. 21, No. 16 Magana et al.

[82] Hoskin DW, Ramamoorthy A. Studies on anticancer activities of

antimicrobial peptides. Biochim Biophys Acta 2008; 1778: 357-75. [83] Rubinchik E, Schneider T, Elliott M, et al. Mechanism of action

and limited cross-resistance of new lipopeptide MX-2401. Antimicrob Agents Chemother 2011; 55: 2743-54.

[84] Dugourd D, Yang H, Elliott M, et al. Antimicrobial properties of MX-2401, an expanded-spectrum lipopeptide active in the presence

of lung surfactant. Antimicrob Agents Chemother 2011; 55: 3720-8.

[85] Hashizume H, Sawa R, Harada S, et al. Tripropeptin C blocks the lipid cycle of cell wall biosynthesis by complex formation with

undecaprenyl pyrophosphate. Antimicrob Agents Chemother 2011; 55: 3821-8.

[86] Koyama N, Inokoshi J, Tomoda H. Anti-infectious agents against MRSA. Molecules 2012; 18: 204-24.

[87] Zhanel GG, Lawson CD, Zelenitsky S, et al. Comparison of the next-generation aminoglycoside plazomicin to gentamicin,

tobramycin and amikacin. Expert Rev Anti Infect Ther 2012; 10: 459-73.

[88] Remy JM, Tow-Keogh CA, McConnell TS, Dalton JM, Devito JA. Activity of delafloxacin against methicillin-resistant

Staphylococcus aureus: resistance selection and characterization. J Antimicrob Chemother 2012; 67: 2814-20.

[89] Biedenbach DJ, Farrell DJ, Flamm RK, et al. Activity of JNJ-Q2, a new fluoroquinolone, tested against contemporary pathogens

isolated from patients with community-acquired bacterial pneumonia. Int J Antimicrob Agents 2012; 39: 321-5.

[90] Morgan A, Cofer C, Stevens DL. Iclaprim: a novel dihydrofolate reductase inhibitor for skin and soft tissue infections. Future

Microbiol 2009; 4: 131-44. [91] Annual report 2008. Switzerland: Arpida Ltd.; 2009. Available

from: http: //www.evolva.com/sites/default/files/annual-reports/arpida-2008-e_1.pdf.

[92] Fischetti VA, Nelson D, Schuch R. Reinventing phage therapy: are the parts greater than the sum? Nat Biotechnol 2006; 24: 1508-11.

[93] Fischetti V. Bacteriophage lytic enzymes: novel anti-infectives. Trends Microbiol 2005; 13: 491-6.

[94] Loeffler J, Djurkovic S, Fischetti VA. Phage lytic enzyme Cpl-1 as a novel antimicrobial for pneumococcal bacteremia. Infect Immun

2003; 71: 6199-204. [95] Thiel K. Old dogma, new tricks--21st Century phage therapy. Nat

Biotechnol 2004; 22: 31-6. [96] Sulakvelidze A, Alavidze A, Morris JG, Jr. Bacteriophage therapy.

Antimicrob Agents Chemother 2001; 45: 649-59. [97] Nelson D, Loomis L, Fischetti VA. Prevention and elimination of

upper respiratory colonization of mice by group A streptocci by using a bacteriophage lytic enzyme. Proc Natl Acad Sci USA 2001;

98: 4107-12. [98] Loeffler J, Fischetti VA. Synergistic lethal effect of a combination

of phage lytic enzymes with different activities on penicillin-sensitive and –resistant Streptococcus pneumoniae strains.

Antimicrobial Agents Chemother 2003; 47: 375-7. [99] Schuch R, Nelson D, Fischetti VA. A bacteriolytic agent that

detects and kills Bacillus anthracis. Nature 2002; 418: 884-9. [100] Yoong P, Schuch R, Nelson D, Fischetti VA. Identification of a

Broadly Active Phage Lytic Enzyme with Lethal Activity against Antibiotic-Resistant Enterococcus faecalis and Enterococcus

faecium. J Bacteriol 2004; 186: 4808-12. [101] O’Flaherty S, Coffey A, Meaney W, Fitzgerald GF, Ross RP. The

recombinant Phage Lysin LysK Has a Broad Spectrum of Lytic Activity against Clinically Relevant Staphylococci, Including

Methicillin-Resistant Staphylococcus aureus. J Bacteriol 2005; 187: 7161-4.

[102] Nelson D, Schuch R, Chahales P, Zhu S, Fischetti VA. PlyC: A multimeric bacteriophage lysin. Proc Natl Acad Sci USA 2006;

103: 10765-70. [103] Cheng Q, Nelson D, Zhu S, Fischetti VA. Removal of group B

streptococci colonizing the vagina and oropharynx of mice with a bacteriophage lytic enzyme. Antimicrob Agents Chemother 2005;

49: 111-7. [104] Gilmer DB, Schmitz JE, Euler CW, Fischetti VA. Novel

bacteriophage lysin with broad lytic activity protects against mixed infection by Streptococcus pyogenes and methicillin-resistant

Staphylococcus aureus. Antimicrob Agents Chemother 2013; 57: 2743-50.

[105] Bagnoli F, Bertholet S, Grandi G. Inferring reasons for the failure

of Staphylococcus aureus vaccines in clinical trials. Front Cell Infect Microbiol 2012; 2: 16.

[106] Therien AG, Huber JL, Wilson KE, et al. Broadening the spectrum of beta-lactam antibiotics through inhibition of signal peptidase

type I. Antimicrob Agents Chemother 2012; 56: 4662-70. [107] Falconer SB, Brown ED. New screens and targets in antibacterial

drug discovery. Curr Opin Microbiol 2009; 12: 497-504. [108] Dean M, Rzhetsky A, Allikmets R. The human ATP-binding

cassette (ABC) transporter superfamily. Genome Res 2001; 11: 1156-66.

[109] Kim HK, Thammavongsa V, Schneewind O, Missiakas D. Recurrent infections and immune evasion strategies of

Staphylococcus aureus. Curr Opin Microbiol 2012; 15: 92-9. [110] Fletcher JI, Haber M, Henderson MJ, Norris MD. ABC transporters

in cancer: more than just drug efflux pumps. Nat Rev Cancer 2010; 10: 147-56.

[111] Reed SG, Orr MT, Fox CB. Key roles of adjuvants in modern vaccines. Nat Med 2013; 19: 1597-608.

[112] Piddock L. Clinically relevant chromosomally encoded multidrug resistance efflux pumps in bacteria. Clin Microbiol Rev 2006; 19:

382-402. [113] Piddock L. Multidrug-resistance efflux pumps - not just for

resistance. Nat Rev Microbiol 2006; 20: 629-366. [114] McAleese F, Petersen P, Ruzin A, et al. A novel MATE family

efflux pump contributes to the reduced susceptibility of laboratory-derived Staphylococcus aureus mutants to tigecycline. Antimicrob

Agents Chemother 2005; 49: 1865-71. [115] Narui K, Noguchi N, Wakasugi K, Sasatsu M. Cloning and

characterization of a novel chromosomal drug efflux gene in Staphylococcus aureus. Biol Pharm Bull 2002; 25: 1533-6.

[116] Xu Z, O'Rourke BA, Skurray RA, Brown MH. Role of transmembrane segment 10 in efflux mediated by the

staphylococcal multidrug transport protein QacA. J Biol Chem 2006; 281: 792-9.

[117] Jin J, Guffanti AA, Bechhofer DH, Krulwich TA. Tet(L) and tet(K) tetracycline-divalent metal/H+ antiporters: characterization of

multiple catalytic modes and a mutagenesis approach to differences in their efflux substrate and coupling ion preferences. J Bacteriol

2002; 184: 4722-32. [118] Tegos G, Stermitz FR, Lomovskaya O, Lewis K. Multidrug pump

inhibitors uncover remarkable activity of plant antimicrobials. Antimicrob Agents Chemother 2002; 46: 3133-41.

[119] Tegos G. Substrates and Inhibitors of microbial efflux pumps; Redifine the Role of Plant Antimicrobilas. In: Mahendra Rai CMC,

editor. Naturally occurring bioactive compounds: a new and safe alternative for control of pests and microbial diseases. Cambridge

Cambridge University Press 2006. [120] Fiamegos Y, Kastritis PL, Exarchou V, et al. 2011 Antimicrobial

and efflux pump inhibitory activity of caffeoylquinic acids from Artemisia absinthium against Gram-positive pathogenic bacteria

Plos One 2011; 6: e18127. [121] Tegos G, Haynes M, Strouse JJ, et al. Microbial Efflux Inhibition;

tactics & strategies. Current Pharmaceutical Design 2011; 17: 1291-302.

[122] Kourtesi C, Ball AR, Huang YY, et al. Microbial Efflux Systems and Inhibitors: Approaches to Drug Discovery and the Challenge of

Clinical Implementation. Open Microbiol J 2013; 7: 34-52. [123] Mitton D, Ackroyd R. A brief overview of photodynamic therapy

in Europe. Photodiagnosis Photodyn Ther 2008; 5: 103-11. [124] St. Denis T, Dai T, Izikson A, et al. All you need is light

Antimicrobial photoinactivation as an evolving and emerging discovery strategy against infectious disease Virulence 2011; 2: 1-

12. [125] Tegos GP, Hamblin MR. Phenothiazinium antimicrobial

photosensitizers are substrates of bacterial multidrug resistance pumps. Antimicrob Agents Chemother 2006; 50: 196-203.

[126] Sharma SK, Dai T, Kharkwal GB, et al. Drug discovery of antimicrobial photosensitizers using animal models. Curr Pharm

Des 2011; 17: 1303-19. [127] Sharma SK, Dai T, Hamblin MR. Antimicrobial Photosensitizers:

Harnessing the Power of Light to Treat Infections In: Tegos G, E M, editors. Antimicrobial Drug Discovery: Emerging Strategies:

CAB International 2012. pp. 313-8. [128] Stermitz F, Lorenz P, Tawara JN, Zenewicz LA, Lewis K. Synergy

in a medicinal plant: antimicrobial action of berberine potentiated

Page 15: Therapeutic options Staphylococcus

New Treatments for Resistant Staphylococci Current Pharmaceutical Design, 2015, Vol. 21, No. 16 2071

by 5'-methoxyhydnocarpin, a multidrug pump inhibitor. Proc Natl

Acad Sci USA 2000; 97: 1433-7. [129] Tegos G, Masago K, Aziz F, Higginbotham A, Stermitz FR,

Hamblin MR. Inhibitors of bacterial multidrug efflux pumps potentiate antimicrobial photoinactivation. Antimicrob Agents

Chemother 2008; 52: 3202-9. [130] Hansen S, Maynard A, Baun A, Joel A, Tickner JA. Late lessons

from early warnings for nanotechnology. Nature Nanotechnology 2008; 3: 444-7.

[131] Mazzola L. Commercializing nanotechnology. Nature Biotechnology 2003; 21: 1137-43.

[132] Han G, Martinez LR, Mihu MR, Friedman AJ, Friedman JM, Nosanchuk JD. Nitric oxide releasing nanoparticles are therapeutic

for Staphylococcus aureus abscesses in a murine model of infection. PLoS One 2009; 4: : e7804.

[133] Kim B, Rutka JT, WChan WCW. Nanomedicine. New England Journal of Medicine 2010; 363: 2434-43.

[134] Griffiths G, Nyström B, Sable SB, Khuller GK. Nanobead-based interventions for the treatment and prevention of tuberculosis. Nat

Rev Microbiol 2010; 8: 827-34. [135] Sekhon BS KS. Inorganic nanomedicine--part 1. Nanomedicine

2010; 6: 516-22. [136] Kell A, Stewart G, Ryan S, et al. Vancomycin-modified

nanoparticles for efficient targeting and preconcentration of Gram-positive and Gram-negative bacteria. ACS Nano 2008; 2: 1777-88.

[137] Taccone FS, Vincent JL, Denis O, Jacobs F. Should we abandon vancomycin for treatment of methicillin-resistant Staphylococcus

aureus pneumonia? Still questions to answer. Clin Infect Dis 2012; 55: 161-3; author reply 3-5.

[138] Corey GR, Wilcox MH, Talbot GH, et al. CANVAS 1: the first Phase III, randomized, double-blind study evaluating ceftaroline

fosamil for the treatment of patients with complicated skin and skin structure infections. J Antimicrob Chemother 2010; 65 (Suppl 4):

iv41-51. [139] Wilcox MH, Corey GR, Talbot GH, et al. CANVAS 2: the second

Phase III, randomized, double-blind study evaluating ceftaroline fosamil for the treatment of patients with complicated skin and skin

structure infections. J Antimicrob Chemother 2010; 65 (Suppl 4): iv53-iv65.

[140] File TMJ, Low DE, Eckburg PB, et al. FOCUS 1: a randomized, double-blinded, multicentre, Phase III trial of the efficacy and

safety of ceftaroline fosamil versus ceftriaxone in community-acquired pneumonia. J Antimicrob Chemother 2011; 66 (Suppl 3):

iii19-32. [141] Low DE, File TMJ, Eckburg PB, et al. FOCUS 2: a randomized,

double-blinded, multicentre, Phase III trial of the efficacy and safety of ceftaroline fosamil versus ceftriaxone in community-

acquired pneumonia. J Antimicrob Chemother 2011; 66 (Suppl 3): iii33-44.

[142] Genome [database on the Internet] [cited 2013 Dec 02]. Available from: http: //www.ncbi.nlm.nih.gov/genome/.

[143] Multi Locus Sequence Typing, Staphylococcus aureus. [cited 2014 Nov 14]; Available from: http: //saureus.mlst.net/sql/sthtml.asp.

[144] Harrison EM, Paterson GK, Holden MT, et al. Whole genome sequencing identifies zoonotic transmission of MRSA isolates with

the novel mecA homologue mecC. EMBO Mol Med 2013; 5: 509-15.

[145] Wendlandt S, Li B, Lozano C, et al. Identification of the novel spectinomycin resistance gene spw in methicillin-resistant and

methicillin-susceptible Staphylococcus aureus of human and animal origin. J Antimicrob Chemother 2013; 68: 1679-80.

[146] Uhlemann AC, Dordel J, Knox JR, et al. Molecular tracing of the emergence, diversification, and transmission of S. aureus sequence

type 8 in a New York community. Proc Natl Acad Sci USA 2014; 111: 6738-43.

[147] Strommenger B, Bartels MD, Kurt K, et al. Evolution of methicillin-resistant Staphylococcus aureus towards increasing

resistance. J Antimicrob Chemother 2014; 69: 616-22. [148] Uhlemann AC, Kennedy AD, Martens C, et al. Toward an

understanding of the evolution of Staphylococcus aureus strain USA300 during colonization in community households. Genome

Biol Evol 2012; 4: 1275-85. [149] Abdelbary MM, Wittenberg A, Cuny C, et al. Phylogenetic

analysis of Staphylococcus aureus CC398 reveals a sub-lineage epidemiologically associated with infections in horses. PLoS One

2014; 9: e88083.

[150] Himsworth CG, Miller RR, Montoya V, et al. Carriage of

methicillin-resistant Staphylococcus aureus by wild urban Norway rats (Rattus norvegicus). PLoS One 2014; 9: e87983.

[151] Garcia-Graells C, van Cleef BA, Larsen J, et al. Dynamic of livestock-associated methicillin-resistant Staphylococcus aureus

CC398 in pig farm households: a pilot study. PLoS One 2013; 8: e65512.

[152] Mitra SD, Velu D, Bhuvana M, et al. Staphylococcus aureus spa type t267, clonal ancestor of bovine subclinical mastitis in India. J

Appl Microbiol 2013; 114: 1604-15. [153] Vincze S, Stamm I, Monecke S, et al. Molecular analysis of human

and canine Staphylococcus aureus strains reveals distinct extended-host-spectrum genotypes independent of their methicillin

resistance. Appl Environ Microbiol 2013; 79: 655-62. [154] Wan MT, Lauderdale TL, Chou CC. Characteristics and virulence

factors of livestock associated ST9 methicillin-resistant Staphylococcus aureus with a novel recombinant staphylocoagulase

type. Vet Microbiol 2013; 162: 779-84. [155] Jamrozy DM, Fielder MD, Butaye P, Coldham NG. Comparative

genotypic and phenotypic characterisation of methicillin-resistant Staphylococcus aureus ST398 isolated from animals and humans.

PLoS One 2012; 7: e40458. [156] Lo YP, Wan MT, Chen MM, et al. Molecular characterization and

clonal genetic diversity of methicillin-resistant Staphylococcus aureus of pig origin in Taiwan. Comp Immunol Microbiol Infect

Dis 2012; 35: 513-21. [157] Camoez M, Sierra JM, Pujol M, et al. Prevalence and molecular

characterization of methicillin-resistant Staphylococcus aureus ST398 resistant to tetracycline at a Spanish hospital over 12 years.

PLoS One 2013; 8: e72828. [158] Prosperi M, Veras N, Azarian T, et al. Molecular epidemiology of

community-associated methicillin-resistant Staphylococcus aureus in the genomic era: a cross-sectional study. Sci Rep 2013; 3: 1902.

[159] Stegger M, Liu CM, Larsen J, et al. Rapid differentiation between livestock-associated and livestock-independent Staphylococcus

aureus CC398 clades. PLoS One 2013; 8: e79645. [160] Holden MT, Hsu LY, Kurt K, et al. A genomic portrait of the

emergence, evolution, and global spread of a methicillin-resistant Staphylococcus aureus pandemic. Genome Res 2013; 23: 653-64.

[161] Tristan A, Rasigade JP, Ruizendaal E, et al. Rise of CC398 lineage of Staphylococcus aureus among Infective endocarditis isolates

revealed by two consecutive population-based studies in France. PLoS One 2012; 7: e51172.

[162] Coombs GW, Goering RV, Chua KY, et al. The molecular epidemiology of the highly virulent ST93 Australian community

Staphylococcus aureus strain. PLoS One 2012; 7: e43037. [163] Shepheard MA, Fleming VM, Connor TR, et al. Historical

zoonoses and other changes in host tropism of Staphylococcus aureus, identified by phylogenetic analysis of a population dataset.

PLoS One 2013; 8: e62369. [164] Portillo BC, Moreno JE, Yomayusa N, et al. Molecular

epidemiology and characterization of virulence genes of community-acquired and hospital-acquired methicillin-resistant

Staphylococcus aureus isolates in Colombia. Int J Infect Dis 2013; 17: e744-9.

[165] Sahin F, Karasartova D, Ozsan TM, et al. Identification of methicillin-resistant Staphylococcus aureus carrying an exfoliative

toxin A gene encoding phage isolated from a hospitalized patient in Turkey. Can J Microbiol 2013; 59: 260-5.

[166] Ibarra JA, Perez-Rueda E, Carroll RK, Shaw LN. Global analysis of transcriptional regulators in Staphylococcus aureus. BMC

Genomics 2013; 14: 126. [167] Kahlon AK, Darokar MP, Sharma A. Probing the evolutionary

conserved regions within functional site of drug-resistant target proteins of Staphylococcus aureus: In silico phylogenetic motif

profiling approach. Indian J Biochem Biophys 2012; 49: 442-50. [168] Okumura K, Shimomura Y, Murayama SY, et al. Evolutionary

paths of streptococcal and staphylococcal superantigens. BMC Genomics 2012; 13: 404.

[169] Kos VN, Desjardins CA, Griggs A, et al. Comparative genomics of vancomycin-resistant Staphylococcus aureus strains and their

positions within the clade most commonly associated with Methicillin-resistant S. aureus hospital-acquired infection in the

United States. MBio 2012; 3. [170] Isobe H, Takano T, Nishiyama A, et al. Evolution and virulence of

Panton-Valentine leukocidin-positive ST30 methicillin-resistant

Page 16: Therapeutic options Staphylococcus

2072 Current Pharmaceutical Design, 2015, Vol. 21, No. 16 Magana et al.

Staphylococcus aureus in the past 30 years in Japan. Biomed Res

2012; 33: 97-109. [171] Wardyn SE, Forshey BM, Smith TC. High prevalence of Panton-

Valentine leukocidin among methicillin-sensitive Staphylococcus aureus colonization isolates in rural Iowa. Microb Drug Resist

2012; 18: 427-33. [172] Albrecht T, Raue S, Rosenstein R, Nieselt K, Gotz F. Phylogeny of

the staphylococcal major autolysin and its use in genus and species typing. J Bacteriol 2012; 194: 2630-6.

[173] Guo FB, Wei W, Wang XL, et al. Co-evolution of genomic islands and their bacterial hosts revealed through phylogenetic analyses of

17 groups of homologous genomic islands. Genet Mol Res 2012; 11: 3735-43.

[174] Li J, Wang L, Ip M, et al. Molecular and clinical characteristics of clonal complex 59 methicillin-resistant Staphylococcus aureus

infections in Mainland China. PLoS One 2013; 8: e70602. [175] Price JR, Didelot X, Crook DW, Llewelyn MJ, Paul J. Whole

genome sequencing in the prevention and control of Staphylococcus aureus infection. J Hosp Infect 2013; 83: 14-21.

[176] Thomas JC, Godfrey PA, Feldgarden M, Robinson DA. Candidate

targets of balancing selection in the genome of Staphylococcus aureus. Mol Biol Evol 2012; 29: 1175-86.

[177] Uddin R, Saeed K. Identification and characterization of potential drug targets by subtractive genome analyses of methicillin resistant

Staphylococcus aureus. Comput Biol Chem 2014; 48: 55-63. [178] Hede K. Antibiotic resistance: An infectious arms race. Nature

2014; 509: S2-3. [179] Couderc C, Jolivet S, Thiébaut AC, et al. Fluoroquinolone Use Is a

Risk Factor for Methicillin-Resistant Staphylococcus aureus Acquisition in Long-term Care Facilities: A Nested Case-Case-

Control Study. Clin Infect Dis 2014; pii: ciu236: [Epub ahead of print].

[180] Boucher H, Wilcox M, Talbot GH, Puttagunta S, Das AF, Dunne MW. Once-weekly dalbavancin versus daily conventional therapy

for skin infection. N Engl J Med 2014; 370: 2169-79. [181] Stephens J, Gao X, Patel DA, Verheggen BG, Shelbaya A, Haider

S. Economic burden of inpatient and outpatient antibiotic treatment for methicillin-resistant Staphylococcus aureus complicated skin

and soft-tissue infections: a comparison of linezolid, vancomycin, and daptomycin. Clinicoecon Outcomes Res 2013; 16: 447-57.

Received: January 21, 2015 Accepted: March 5, 2015