12
part of 10.2217/17460913.2.6.655 © 2007 Future Medicine Ltd ISSN 1746-0913 REVIEW Future Microbiol. (2007) 2(6), 655–666 655 Novel targets for antibiotics in Staphylococcus aureus Knut Ohlsen & Udo Lorenz Author for correspondence University of Würzburg, Institute for Molecular Infection Biology, Röntgenring 11, 97070 Würzburg, Germany Tel.: +49 931 312 155; Fax: +49 931 312 578; knut.ohlsen@mail. uni-wuerzburg.de Keywords: antibiotic resistance, Gram-positive pathogens, immunotherapy, MRSA, new targets, novel antibacterials, Staphylococcus aureus Multiple resistant staphylococci that cause significant morbidity and mortality are the leading cause of nosocomial infections. Meanwhile, methicillin-resistant Staphylococcus aureus (MRSA) also spreads in the community, where highly virulent strains infect children and young adults who have no predisposing risk factors. Although some treatment options remain, the search for new antibacterial targets and lead compounds is urgently required to ensure that staphylococcal infections can be effectively treated in the future. Promising targets for new antibacterials are gene products that are involved in essential cell functions. In addition to antibacterials, active and passive immunization strategies are being developed that target surface components of staphylococci such as cell wall-linked adhesins, teichoic acids and capsule or immunodominant antigens. In the past decade, a notable rise in the number of infections caused by antibiotic resistant Gram-positive pathogens such as methi- cillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant enterococci (VRE) and penicillin-resistant Streptococcus pneumoniae (PRSP) pose specific therapeutic challenges. Spe- cifically, MRSA is now the most common cause of hospital-acquired infections worldwide. The spectrum of diseases caused by S. aureus can range from mild superficial lesions to life-threat- ening infections including bacteremia, pneumo- nia and endocarditis. Newborns, patients with chronic bronchopulmonary disorders, those who have undergone surgical procedures, patients with diabetes mellitus, indwelling intravascular catheters and immunocompromised individuals, are especially susceptible to S. aureus. According to the National Nosocomial Infections Surveil- lance (NNIS) system, MRSA strains accounted for more than 50% of all S. aureus infections in intensive care units in the USA [1]. High MRSA rates have also been reported in Europe, increas- ing most rapidly in the UK (now up to 45%) [2]. This situation is further complicated by the recent spread of community acquired (CA)- MRSA [3]. Although almost all of the MRSA iso- lates are susceptible to vancomycin, and also to the newly introduced antibiotics linezolid and daptomycin, there is an urgent need to discover antibiotics targeting novel cellular functions not yet targeted by current antibacterial agents. This is especially important as S. aureus isolates with intermediate and high resistance to vanco- mycin (as well as to all other antibiotics) have now been reported [4–6]. There is substantial fear in the medical community that a ‘superbug’ will emerge that is resistant to all antibiotics. In this review, the recent development of new strategies to combat infections caused by S. aureus are summarized and trends for the future are discussed. Search for essential gene products The search of new targets for novel antibacterials represents a challenging task. In principle, each essential protein can serve as a target for anti- microbials. However, further criteria have to be met, including selectivity (functional and/or structural differences between human and bacte- rial target molecules), distribution (which micro- organisms are susceptible), ease of assay (can an assay for high-throughput screening be devel- oped?), intracellular concentration of the poten- tial target, mutational potential (development of resistance), bacteriocidal versus bacteriostatic action (in vivo efficiency) and drugability (likeli- hood of being able to modulate a target with a small-molecule drug), before a target is selected for lead development. Several attempts for defin- ing essential factors of bacteria have recently been published, including theoretical calculations on the minimal genome of bacteria and extensive mutational analysis [7–9]. For example, in Bacillus subtilis, 271 genes were predicted to be essential [10]. The vast majority of essential genes encode factors that are involved in synthesis of the cell envelope, information processing, the determina- tion of cell shape and division and generation of energy. In S. aureus various essential proteins have been defined by antisense-RNA interference and mutagenesis studies [11,12]. These studies generated lists of putative novel targets covering all known key pathways, for example translation,

Novel targets for antibiotics in Staphylococcus aureus

  • Upload
    udo

  • View
    214

  • Download
    1

Embed Size (px)

Citation preview

Page 1: Novel targets for antibiotics in Staphylococcus aureus

part of

10.2217/17460913.2.6.655 © 2007 Future Medicine Ltd ISSN 1746-0913

REVIEW

Future Microbiol. (2007) 2(6), 655–666 655

Novel targets for antibiotics in Staphylococcus aureusKnut Ohlsen† & Udo Lorenz†Author for correspondenceUniversity of Würzburg,Institute for Molecular Infection Biology,Röntgenring 11,97070 Würzburg, GermanyTel.: +49 931 312 155;Fax: +49 931 312 578;[email protected]

Keywords: antibiotic resistance, Gram-positive pathogens, immunotherapy, MRSA, new targets, novel antibacterials, Staphylococcus aureus

Multiple resistant staphylococci that cause significant morbidity and mortality are the leading cause of nosocomial infections. Meanwhile, methicillin-resistant Staphylococcus aureus (MRSA) also spreads in the community, where highly virulent strains infect children and young adults who have no predisposing risk factors. Although some treatment options remain, the search for new antibacterial targets and lead compounds is urgently required to ensure that staphylococcal infections can be effectively treated in the future. Promising targets for new antibacterials are gene products that are involved in essential cell functions. In addition to antibacterials, active and passive immunization strategies are being developed that target surface components of staphylococci such as cell wall-linked adhesins, teichoic acids and capsule or immunodominant antigens.

In the past decade, a notable rise in the numberof infections caused by antibiotic resistantGram-positive pathogens such as methi-cillin-resistant Staphylococcus aureus (MRSA),vancomycin-resistant enterococci (VRE) andpenicillin-resistant Streptococcus pneumoniae(PRSP) pose specific therapeutic challenges. Spe-cifically, MRSA is now the most common causeof hospital-acquired infections worldwide. Thespectrum of diseases caused by S. aureus canrange from mild superficial lesions to life-threat-ening infections including bacteremia, pneumo-nia and endocarditis. Newborns, patients withchronic bronchopulmonary disorders, those whohave undergone surgical procedures, patientswith diabetes mellitus, indwelling intravascularcatheters and immunocompromised individuals,are especially susceptible to S. aureus. Accordingto the National Nosocomial Infections Surveil-lance (NNIS) system, MRSA strains accountedfor more than 50% of all S. aureus infections inintensive care units in the USA [1]. High MRSArates have also been reported in Europe, increas-ing most rapidly in the UK (now up to 45%) [2].This situation is further complicated by therecent spread of community acquired (CA)-MRSA [3]. Although almost all of the MRSA iso-lates are susceptible to vancomycin, and also tothe newly introduced antibiotics linezolid anddaptomycin, there is an urgent need to discoverantibiotics targeting novel cellular functionsnot yet targeted by current antibacterial agents.This is especially important as S. aureus isolateswith intermediate and high resistance to vanco-mycin (as well as to all other antibiotics) havenow been reported [4–6]. There is substantial fearin the medical community that a ‘superbug’

will emerge that is resistant to all antibiotics. Inthis review, the recent development of newstrategies to combat infections caused byS. aureus are summarized and trends for thefuture are discussed.

Search for essential gene productsThe search of new targets for novel antibacterialsrepresents a challenging task. In principle, eachessential protein can serve as a target for anti-microbials. However, further criteria have to bemet, including selectivity (functional and/orstructural differences between human and bacte-rial target molecules), distribution (which micro-organisms are susceptible), ease of assay (can anassay for high-throughput screening be devel-oped?), intracellular concentration of the poten-tial target, mutational potential (development ofresistance), bacteriocidal versus bacteriostaticaction (in vivo efficiency) and drugability (likeli-hood of being able to modulate a target with asmall-molecule drug), before a target is selectedfor lead development. Several attempts for defin-ing essential factors of bacteria have recently beenpublished, including theoretical calculations onthe minimal genome of bacteria and extensivemutational analysis [7–9]. For example, in Bacillussubtilis, 271 genes were predicted to be essential[10]. The vast majority of essential genes encodefactors that are involved in synthesis of the cellenvelope, information processing, the determina-tion of cell shape and division and generation ofenergy. In S. aureus various essential proteinshave been defined by antisense-RNA interferenceand mutagenesis studies [11,12]. These studiesgenerated lists of putative novel targets coveringall known key pathways, for example translation,

k.rowland
Text Box
For reprint orders, please contact: [email protected]
Page 2: Novel targets for antibiotics in Staphylococcus aureus

656

REVIEW – Ohlsen & Lorenz

Future Microbiol. (2007) 2(6) future science groupfuture science group

transcription, cell division and metabolism.Importantly, for approximately 30% of the puta-tive essential genes the function remains unclear.Within the group of proteins with no knownfunction some attractive novel targets have beenidentified. For example, in an attempt to evaluatethe potential of proteins of unknown functionfor target search, Zalacain et al. investigated 144open reading frames (ORFs) without significanthomologies to proteins with known functions inS. pneumoniae, 36 of which were essential inS. pneumoniae and 14 of these were also essentialin S. aureus and Haemophilus influenzae [13]. Inaddition to a number of previously validated orsuggested targets, in vivo essentiality in S. aureushas been proven for five ORFs: spoIIIJ2, trmU,ydiC, ydiE and yneS. Interestingly, by testing ofnonessential in vitro genes, in vivo, four out of 17genes were also essential, suggesting a broaderselection of bacterial targets [13]. Consequently,essentiality of a given gene product is not onlyspecies dependent but is also dependent on theenvironmental test conditions. For example, theavailability of specific macromolecules or nutri-ents has a pivotal impact on the growth of thebacteria. Thus, every novel target has to be vali-dated in vitro and also in vivo in different patho-gens under various test conditions. This isespecially of importance for the development ofbroad-spectrum antibiotics.

In S. aureus particularly, three systems havebeen applied for conditional gene expression tovalidate putative essential genes. The Pxyl–tetO-system contains the xylose promoter controlledby tetracycline or anhydrotetracycline, thePxyl–xylR system can be induced by xylose andrepressed by glucose, and the Pspac–lacI system,including the Pspac hybrid promoter, is controlledby the lac-operator inducible by isopropyl β-D-1-thiogalactopyranoside (IPTG) [14–16]. Systemsusing tetracycline as the inducer are commonlyapplied for in vivo gene expression as tetracycline,or its nontoxic derivative anhydrotetracycline, iseasily distributed in the body and also penetratesintracellularly [12,17]. Recently, an alternative con-ditional in vivo expression system has beendeveloped based on the Pspac-lacI-regulatablepromoter element (Figure 1) [14,18].

Novel targets for antibacterials in Staphylococcus aureusThe investigation of novel targets in new path-ways has been suggested to be especially promis-ing as it is regarded that antimicrobials targetingnew pathways will most likely exhibit no

cross-resistance to commonly used antimicrobialsand, therefore, it should take more time for resist-ant strains to arise. However, a recent study inves-tigating Salmonella metabolic pathways suggesteda shortage of new metabolic targets for broad-spectrum antibiotics. In a comprehensive in vitroand in vivo approach, 155 promising targets totreat Salmonella infections were identified, 64 ofwhich were also conserved in other importantpathogens such as S. aureus, Enterococcus faecalis,S. pneumoniae and H. influenzae. Almost all ofthese targets belong to pathways already inhibitedby current antibiotics (peptidoglycan biosynthe-sis, folate biosynthesis, isoprenoid biosynthesis,fatty acid biosynthesis and tRNA synthases) orpathways previously considered for antimicrobialdevelopment [19]. Consequently, novel targets inalready approved pathways are being investigated.However, the discovery of new drugs againstolder targets, based on new structural informa-tion, represents an additional promising strategythat will be discussed in the following sections.

Cell division (FtsZ)FtsZ is a tubulin-like GTPase that plays anessential role in bacterial cell division. Itshomologs are present in almost all eubacteriaand archaea. During cell division, FtsZ formspolymers in the presence of GTP, which recruitother proteins implicated in division to form thecell-division apparatus. Inhibition of FtsZpolymerization prevents cells from dividing,leading to cell death [20,21]. Recently, a com-pound named viriditoxin has been identifiedthat blocked FtsZ polymerization. Viriditoxinexhibited broad-spectrum antibacterial activityagainst clinically relevant Gram-positive patho-gens, including MRSA and VRE, withoutaffecting the viability of eukaryotic cells [21].

Deformylase (Pdf1) Peptide deformylase (PDF) is an essential bac-terial metalloenzyme which deformylates theN-formylmethionine of newly synthesizedpolypeptides. In prokaryotes, the amino groupof the methionyl moiety carried by the initiatortRNAfMet is N-formylated by formyltransferaseprior to its incorporation into a polypeptide.Consequently, N-formylmethionine is alwayspresent at the N-terminus of a nascent bacterialpolypeptide. To further proceed translation, theformyl group is hydrolyzed by peptide deformy-lase. PDF is essential for bacterial growth andrepresents an attractive novel target [22,23]. Thegene encoding PDF (def ) is present in all

Page 3: Novel targets for antibiotics in Staphylococcus aureus

657

Novel targets for antibiotics in Staphylococcus aureus – REVIEW

future science groupfuture science group www.futuremedicine.com

sequenced pathogenic bacterial genomes. Inter-estingly, a human mitochondrial peptidedeformylase has been reported recently thatmay provide a new anticancer target of actin-onin-based antibiotics [24]. The x-ray structureof PDF has been resolved, defining PDF as anew class of metalloproteases [25]. Severalhydroxamic acid derivatives have been reportedto be potent PDF inhibitors with in vitro anti-bacterial activity [26,27]. A number of relatedcompounds inhibited the growth of severalclinically relevant bacterial pathogens. Impor-tantly, one compound termed BB-83698 wasalso active in systemic models of S. aureus infec-tion in the mouse and is the first compound inthis novel class to enter clinical trials [28].

Teichoic acid synthesisTeichoic acid (TA) synthesis has been regarded asthe target for antimicrobials as its essentiality hasbeen postulated for B. subtilis and Listeria mono-cytogenes [29,30]. However, more recent work dem-onstrated that wall teichoic acids (WTAs) aredispensible both in B. subtilis and S. aureus. Para-doxically, the gene catalyzing the first step inWTA synthesis, tagO in B. subtilis and tarO inS. aureus, could be deleted to yield viable mutantslacking TA in the cell wall while later-actingenzymes of the pathway display an essential phe-notype [31,32]. Possibly, an accumulation of unde-caprenyl-linked precursors in the WTA-synthesispathway results in cessation of the bacterial cellwall synthesis due to depletion of the small

Figure 1. Use of in vivo imaging technology to assess essential gene function of ligA.

The time course of bioluminescence levels was monitored for seven consecutive days (from 2 h until day 7) after subdermal infection of the lower back area of mice with 1 x 108 colony forming units of Staphylococcus aureus Xen29-Pspac:ligA. Conditional expression of essential ligA depends (in this strain) on the presence of the inducer IPTG [14]. S. aureus infection has been established in mice that have received IPTG (upper row). In animals that had not received IPTG the bacteria were not able to cause infection (lower row). Signal intensity is indicated by a pseudocolor scale. IPTG: Isopropyl β-d-1-thiogalactopyranoside; p.i.: Post-infection.

40247i l M d l O ADO200705021A 4113334A i l M d l

2h p.i. d3 p.i. d5 p.i. d7 p.i. d1 p.i.

600

500

400

300

200

100

x103

ImageMin = -34018Max = 5.2636e+06p/sec/cm2/sr

600

500

400

300

200

100

x103

O200 043013A 0 0 0316 010ADO200 0 0 14A

Xen29-Pspac::ligA+ IPTG

Xen29-Pspac::ligA- IPTG

Color barMin = 30,000Max = 6e+05

ImageMin = -33533Max = 44672p/sec/cm2/sr

O200 0 0 08A

Color barMin = 30,000Max = 6e+05

Page 4: Novel targets for antibiotics in Staphylococcus aureus

658

REVIEW – Ohlsen & Lorenz

Future Microbiol. (2007) 2(6) future science groupfuture science group

bactoprenyl-phosphate pool. Results from theauthors’ laboratory support an essential pheno-type of one of the downstream genes. By use of aconditional mutant, in vitro essentiality ofSA0245 (tarF ) has been demonstrated [Unpublished

Data]. Moreover, TagO has been demonstrated toplay a critical role in nasal colonization, which is animportant risk factor for nosocomial infections [33].Furthermore, alanylation of TAs undertaken byfactors encoded by the dlt operon play an essentialrole in resistance to cationic antimicrobialpeptides (CAMPs) of the host innate immunesystem [34]. More work has to be done to definethe exact function of TAs for cell viability andvirulence of Gram-positive pathogens.

Other essential targetsA number of components have been suggestedto be attractive targets in S. aureus based onin vitro essentiality data (Table 1), such as thetwo-component systems YycFG and YhcSR,type I signal peptidase (SPase) SpsB, the ABCtransporter, a glycoprotease Gcp, thioredoxinTrxA, thioredoxin reductase TrxB, DNApolymerase III DnaE, DNA ligase LigA andribonuclease P [35–42]. For most of these targetsessentiality has been investigated only in vitroand in vivo validation is still lacking. Neverthe-less, for some of these targets (e.g., SpsB),inhibitors have been discovered that arecurrently under investigation [43].

Table 1. Promising targets for novel antibacterials against Staphylococcus aureus.

Pathway/target Function Ref.

Fatty acid synthesis

FabI Enoyl-acyl carrier protein reductase [44]

FabF/H β-ketoacyl-(acyl carrier protein) synthase I/II [46]

DNA replication

GyrA DNA-gyrase [57,58]

ParE Topoisomerase IV

Protein modification

Pdf Peptide deformylase [23,28]

Protein elongation

tRNA synthetases Protein biosynthesis [54]

Peptidoglycan synthesis

PBP2 Peptidoglycan glycosyltransferases [49]

MurB UDP-N-acetylglucosamine-enolpyruvyl reductase [50]

FmhB Pentaglycine interpeptide biosynthesis [16]

FemAB Pentaglycine interpeptide biosynthesis [84]

Ddl D-alanine:D-alanine ligase [85]

Regulation

YycG/YycF (VicRK) Autolysis [42]

YhcSR Unknown [40]

Gcp Glycoprotease, autolysis [41,86]

Protein secretion

SpsB Signal peptidase [43]

Cell division

FtsZ GTPase [21]

Teichoic acid biosynthesis

TarB, TarD, TarF, TarIJ, TarH Teichoic acid polymer formation [32]

Stress response

TrxA Thioredoxin [36]

TrxB Thioredoxin reductase [36]

ClpP Proteolytic component of Clp complex [87]

LigA DNA-ligase [39,88]

Page 5: Novel targets for antibiotics in Staphylococcus aureus

659

Novel targets for antibiotics in Staphylococcus aureus – REVIEW

future science groupfuture science group www.futuremedicine.com

Old targets: new drugsRenewed interest is directed towards the discoveryof new compounds against old targets or previouslytargeted pathways. For example, new structuraldata on target proteins will help to define novelinhibitors based on computer-aided modeling.

Type II fatty acid-synthesis pathwayType II fatty acid synthesis is essential for bacte-rial cell viability. In bacteria, FabH catalyzes thefirst condensation reaction using acetyl-CoA andmalonyl-acetyl carrier protein (ACP) to produceacetoacetyl-ACP. This product is reduced byFabG, dehydrated by FabA/Z and then reducedby FabI to generate butyryl-ACP. In the next stepof this cycle, FabF synthesizes β-ketoacyl-ACP.Several inhibitors of this pathway are knownincluding the antiseptic triclosan targeting FabI,isoniazid, which also targets this enzyme inMycobacterium tuberculosis and cerulenin, whichis a selective FabF/B inhibitor. Novel com-pounds have been isolated including platensi-mycin acting on FabF/B or platencin (acting onFabF/B and FabH), or naphthyridin acrylamidederivatives targeting FabI [44–46]. The main prob-lem with FabI inhibitors is the lack of broad-spectrum activity as, for example, S. pneumoniaepossesses FabK as enoyl-ACP reductase [47]. Nev-ertheless, potent FabI inhibitors may be usedagainst S. aureus and other staphylococci. Therecently described FabF and FabF/FabH inhibi-tors platensimycin and platensin, isolated fromStreptomyces platensis, are promising candidatesfor novel antibacterial drugs as these compoundsdemonstrated broad-spectrum Gram-positiveactivity and, importantly, have demonstratedin vivo efficacy [45,46].

Cell wall synthesisNovel, effective compounds against targets of thecell wall machinery are excellent candidates fornew antibacterials. Several potential targets of cellwall-synthesis pathways have been postulated,including penicillin-binding proteins (PBPs),UDP-N-acetylglucosamine-enolpyruvyl reductase(MurB), UDP-N-acetylglucosamine-enolpyruvyltransferase (MurA), UDP-N-acetyl muramylL-alanine ligase (MurC), N-acetylglucosamine-1-Pacetyl transferase (GlmU) and D-alanine:D-alanineligase (Ddl) or, specifically for S. aureus, enzymeswhich are involved in formation of the penta-glycine cross-bridge such as FemAB or FemX[16,48–51]. Inhibitor design should be facilitated bythe recent resolution of 3D structures of, forexample, MurB and PBP2 [49].

There are several compounds in preclinicaland clinical development targeting lipid II,which include glycopeptides (dalbavancin, orita-vancin and telavancin), lantibiotics (e.g. nisinand mersacidin), mannopeptimycins and ramo-planin [52]. Lipid II is a membrane-anchored cell-wall precursor that is essential for bacterial cell-wall biosynthesis. The effectiveness of inhibitinglipid II-derived pathways as an antibacterialstrategy is highlighted by the fact that this mole-cule is the target for at least four different classesof antibiotics, including the clinically importantglycopeptide antibiotic vancomycin. The spec-trum of activity of novel lipid II inhibitorsencompasses mostly Gram-positive pathogensincluding problematic resistant types such asMRSA, VRSA, VRE and pneumococci. Struc-tural modifications of peptide antibiotics alsoincreased activity against some Gram-negativebacteria such as Helicobacter pylori, Campylo-bacter jejuni, Neisseria spp., Salmonella spp.,Pseudomonas aeruginosa and Shigella spp.

tRNA synthetasestRNA synthetases meet many of the criteria forantibacterial targets. These enzymes are essen-tial for growth of all pathogenic species and cat-alyze a limiting step in a vital bacterial function.Several aminoacyl-tRNA synthetases have beenvalidated as potential drug targets, and leadcompounds with potent inhibitory activitieshave been generated to block, for example, Ile-,Phe-, Met-, Tyr-, Pro- and Trp-tRNA syn-thetase [35,53,54]. Although theoretically all 20amino acid-tRNA-synthetases represent attrac-tive targets and a large number of promisinglead compounds have been identified, only afew agents have moved forward into clinicaldevelopment [35,55]. Mupirocin is the onlyexample of an inhibitor of amino acid-tRNA-synthetases in clinical use. The drug is apotent inhibitor of isoleucyl-tRNA synthetaseand has a specific importance for control ofMRSA [56]. This topical antibiotic, mostly usedas a cream to eradicate MRSA from the nose, ishighly active against S. aureus and Streptococcuspyogenes, it also active against H. influenzae,Neisseria spp. and Bordetella pertussis, but isrelatively inactive against anaerobic strep-tococci, enterococci and most Gram-negativebacteria [54]. Phenyl-thiazolylurea-sulfona-mides were identified as a class of inhibitors ofphenylalanyl (Phe)-tRNA synthetase by high-throughput screening and chemical variation ofthe lead structures. The compounds inhibit

Page 6: Novel targets for antibiotics in Staphylococcus aureus

660

REVIEW – Ohlsen & Lorenz

Future Microbiol. (2007) 2(6) future science groupfuture science group

Phe-RS of E. coli, H. influenzae, S. pneumoniaeand S. aureus, with 50% inhibitory concentra-tions in the nanomolar range [53]. Further stud-ies are needed to develop active compounds thatalso act systematically against S. aureus andother pathogens.

Bacterial DNA gyrase & topoisomerase Bacterial DNA gyrase and topoisomerase IV(topo IV) are highly conserved type II topo-isomerases that play essential roles in promotingDNA replication and transcription. These mole-cules are attractive targets for antibacterial drugdiscovery. The fluoroquinolones bind to theGyrA subunit of gyrase and/or the ParC subunitof topo IV blocking essential functions. Theessentiality and evolutionary conservation ofgyrase and topo IV in bacteria suggests broad-spectrum antibacterial activity to the fluoro-quinolones. A recent advance was obtained witha structure-guided drug design approach to opti-mize a novel series of aminobenzimidazoles thatinhibit the essential ATPase activities of bacterialDNA gyrase and topoisomerase IV [57,58]. Thesestudies led to the development of compoundswith potent activities against a variety of bacterialpathogens, for example, S. aureus, H. influenzae,S. pneumoniae, E. coli and E. faecalis.

Immunotherapy: an alternative strategy to antibiotics for the treatment of infections by MRSAThe need of new therapeutic options to treatinfections due to MRSA strains has released anumber of initiatives for the development ofactive or passive immunotherapy approaches [59].There are some clearly defined at-risk populationsfor staphylococcal infections that would benefitfrom immunotherapeutics, including dialysispatients, patients with ventriculoperitonealshunts, patients at risk of infective endocarditis

and residents of nursing homes. Furthermore,increasing incidence of MRSA infections in chil-dren with no predisposing risk factors, as well asin young adults led to the discussion of whetherthere is a specific need for a general immunizationcampaign. In addition, passive immunoprophy-laxis using either polyclonal or monoclonal anti-bodies could aid immunocompromised patients,premature infants, mechanical ventilated persons,and patients who carry foreign medical devicessuch as catheters and implants to generate apotent immune response [60]. Consequently, allintensive care unit patients may benefit frompassive immunization.

The identification of effective target structuresfor antibody-based therapy is a prerequisite forthe development of immunotherapeutics. Prom-ising antigens can be selected from so-calledmicrobial surface components recognizing adhe-sive matrix molecules (MSCRAMMs). Theseproteins are located on the surface of S. aureusand play a prominent role in the process of adhe-sion to specific sites on human tissues orimplanted medical devices [61]. Antibodies recog-nizing MSCRAMMs promoted an enhancedimmune clearance and inhibited the adherenceof S. aureus bacteria to tissues in several animalmodels [62]. Several clinical trials evaluatingin vivo efficiency of immunotherapy in humanrisk populations have been published, however,most of these studies failed (Table 2). For example,a hyperimmune IgG preparation to clumpingfactor A (ClfA) called Veronate® (Inhibitex, GA,USA) has been developed by collecting sera frompatients with high titers to ClfA [63]. PositivePhase II data had indicated the potential for pro-tecting low birth weight infants from staphylo-coccal infections, but in a following Phase IIIstudy, Veronate failed to show efficiency [64,65].In another Phase III clinical trial that showed nosignificant protection, hemodialysis patients

Table 2. Antibody-based therapies against Staphylococcus aureus in clinical studies.

Drug Preparation Company Target Phase

Aurograb® Antibody fragment NeuTec Pharma (Novartis subsidiary)

ABC transporter III

Altastaph® Polyclonal antibody Nabi Capsular polysaccharide II

StaphVax® Polyclonal antibody Nabi Capsular polysaccharide III

Veronate® Polyclonal antibody Inhibitex Clumping factor A III

Aurexis® (tefibazumab) Monoclonal antibody Inhibitex Clumping factor A II

PagibaximabBSYX-A110

Humanized monoclonal antibody Biosynexus and GlaxoSmithKline

Lipoteichoic acid II

V710 Vaccine Intercell and Merck Conserved protein I

Page 7: Novel targets for antibiotics in Staphylococcus aureus

661

Novel targets for antibiotics in Staphylococcus aureus – REVIEW

future science groupfuture science group www.futuremedicine.com

were vaccinated with a polysaccharide conjugatevaccine containing capsule polysaccharides type 5and type 8 coupled to pseudomonas exotoxin Atoxoid (StaphVAX®) [66]. Two recent studies sug-gest that in addition to type 5 and 8 capsulepolysaccharides, the newly described type 336, apolyribitol phosphate N-acetylglucosamine,should be included into a capsular polysaccha-ride-protein conjugate vaccine as it would extendthe coverage substantially. Such a vaccine wouldtarget almost all MSSA and MRSA strains cur-rently circulating in Germany and France [67,68].Furthermore, tefibazumab (Aurexis®) a human-ized monoclonal antibody targeting clumpingfactor A, is in development as an adjunctive ther-apy for serious S. aureus infections. After aPhase I trial in 2006, additional trials provingefficacy are pending [69–71]. Another candidatefor immunotherapy that could possibly provideprotection against S. aureus is an immunodomi-nant ABC transporter [72]. NeuTec Pharma(Manchester, UK) has developed geneticallyrecombinant antibodies (Aurograb®) against thisstructure, which has intrinsic activity inS. aureus, synergistic activity with vancomycinand a broad spectrum of activity to differentstrains of S. aureus, including the currently epi-demic strains of MRSA [73]. The Phase II studywith Aurograb was completed in 2003 and PhaseIII trials have commenced. Moreover, a chimeric

antibody targeting lipoteichoic acid named Pag-ibaximab has entered Phase II studies to preventstaphylococci sepsis in the very low birth weightneonate [73]. Furthermore, recent clinical Phase Istudies were completed with a vaccine developedto target a conserved protein antigen [201].

Several other experimental vaccine candidatesthat demonstrated some efficiency in preclinicalstudies are in development. These vaccines targetmostly surface components such as the polysac-charide intercellular adhesin, alternatively desig-nated poly N-acetyl-β-1,6-glucosamine, ironsurface determinant A and B, fibrinogen-bind-ing proteins ClfA and ClfB, and serine-aspartate(SD) repeat-containing proteins SdrD, SdrE andSdrG (Table 3) [74–79]. Promising results in animalmodels justify ongoing clinical validation. How-ever, the studies also revealed that due to thegreat variety of virulence factors produced byS. aureus, a single immunologic target might notbe sufficient for complete protection. It is likelythat a cocktail containing a combination of sev-eral Staphylococcal virulence factors, as demon-strated recently by using a combination of IsdA,IsdB, SdrD and SdrE antigens, might be the bestapproach for a successful vaccine [78]. Further-more, completely unknown at present is thepotential of the so-called secretable expandedrepertoire adhesive molecules (SERAMs), suchas the extracellular adhesive protein, the

Table 3. Targets of Staphylococcus aureus for implementing antibody-based therapy.

Function Ref.

Targets with efficacy in animal models

Clumping factor A (ClfA) Adhesion [75,77]

Capsule polysaccharides type 5 and 8 Attachment to surfaces and immune evasion [89,90]

Iron-responsive surface determinant A Adhesion [78,91]

Iron-responsive surface determinant B Adhesion [76,78]

Poly-N-acetyl glucosamine Biofilm production [79,92]

Collagen-binding adhesin Adhesion [74]

Fibronectin-binding protein Adhesion [74]

Fibronectin-binding proteins Adhesion [93–95]

Serine-aspartate repeat-containing proteins SdrD, SdrE

Adhesion [78]

Targets with theoretical efficacy

Exfoliative toxin A + B Esterase/protease activity [96]

Toxic shock syndrome toxin Superantigenic activity [97]

Bone sialoprotein-binding protein Adhesion [98]

α + γ-hemolysin Cell lysis [99]

Panton–Valentine leukocidin toxin Polymorphonuclear lysis [100]

Peptidoglycan Various [101]

Immunodominant antigen A and B Lytic transglycosylase and adhesion [102]

Page 8: Novel targets for antibiotics in Staphylococcus aureus

662

REVIEW – Ohlsen & Lorenz

Future Microbiol. (2007) 2(6) future science groupfuture science group

extracellular matrix binding protein, the coagu-lase or the extracellular fibrinogen binding pro-tein, as targets for vaccine development. Theseproteins bind to host matrix proteins and arereceiving progressively more attention becausemany of them possess immunmodulatory activi-ties [80]. Despite recent progress, more experi-mental work is necessary to developimmunotherapeutics that can protect humans atrisk of serious S. aureus infections.

Conclusion & future perspectiveThe spread of antibiotic-resistant bacteria in hos-pitals and communities raises serious concerns.However, only a handful of new antibacterialagents have been approved for therapy since1998, and only two of these, linezolid and dap-tomycin, have novel mechanisms of action.Although there is a clear need for new anti-bacterials, research and development expendi-tures for antibacterials spent by the largestpharmaceutical companies have continued todecrease [81,82]. Since 1995, when the first com-plete bacterial genome was published, scientists’and physicians’ enthusiasm has mounted and anew era for antibacterial drug discovery has beenheralded. Meanwhile, more than 500 bacterial

genomes have been sequenced and the completegenome sequences of 12 S. aureus strains areavailable in public databases. However, only ahandful of targets have been selected by highthroughput screens (HTSs) using genome-com-parision approaches to identify novel lead com-pounds. Surprisingly, the success of HTScampaigns run by several pharmaceutical com-panies was very poor. For example, GlaxoSmith-Kline ran 70 HTS campaigns during 1995 to2001 and only five leads were delivered. Like-wise, other companies failed to identify suitablecandidates. Therefore, such programmes werestopped and the focus of antibacterial researchshifted to optimization of known antibacterialmolecules, screening of small natural productlibraries and rational design initiatives [35]. Theenthusiasm of the genome era has passed, lead-ing to the realization that only a very limitednumber of proteins have the potential to be anovel target for drug development and immuno-therapy [19,83]. Further basic research is necessaryto understand the physiology and virulence ofS. aureus and other pathogens that will form thebasis for discovery of novel targets for antimicro-bial drugs and protective immunotherapeutics inthe future.

Executive summary

Methicillin-resistant Staphylococcus aureus infections

• Staphylococcus aureus is a leading cause of nosocomial and community acquired infections.• Resistance development has become a global health problem.• New resistance types such as vancomycin-resistant S. aureus have recently developed.• New pathotypes such as community-acquired methicillin-resistant S. aureus expressing Panton–Valentine leucotoxin have evolved.

Novel targets against S. aureus

• Identification of new targets addressing novel pathways is urgently needed.• In vivo validation of targets is essential to assess the role of targets for bacterial viability in vivo.• Novel targets with broad-spectrum activity are difficult to define.• New structural information may result in the design of new inhibitory molecules.• Search for novel inhibitors of old targets or pathways should be considered.

Immunotherapy

• Passive or active immunization strategies could help to protect persons at high risk.• Human antibodies in clinical trials mostly failed or provided only dissatisfying results.• Several immunotherapeutics targeting surface components with efficacy in animal models are under development.

Conclusion

• Novel targets against S. aureus have to be discovered. • The number of new targets is limited, so few antibiotics have been approved in the past 10 years. • Protection observed in experimental infections suggests that a staphylococcal immunotherapy is achievable.• More preclinical, epidemiological and clinical studies are required before an optimal approach can be developed.

Future perspective

• Exploitation of functions of proteins with unknown function will help to identify novel targets.• Structure-guided drug design will lead to the development of novel antibacterials.• Antigen combinations for active immunization and antibody cocktails for passive immunotherapy should protect persons at risk.

Page 9: Novel targets for antibiotics in Staphylococcus aureus

663

Novel targets for antibiotics in Staphylococcus aureus – REVIEW

future science groupfuture science group www.futuremedicine.com

AcknowledgementsWe thank U Wallner and T Schäfer for technical assistanceand support in animal experiments.

Financial & competing interests disclosure The work of Knut Ohlsen was supported by a grant of theBavarian Science Foundation (FORINGEN), the Deutsche

Forschungsgemeinschaft (SFB630, TR34), and the EUframework 6 project ‘StaphDynamics’. The authors have noother relevant affiliations or financial involvement with anyorganization or entity with a financial interest in or finan-cial conflict with the subject matter or materials discussed inthe manuscript apart from those disclosed. No writingassistance was utilized in the production of this manuscript.

BibliographyPapers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers.1. National Nosocomial Infections

Surveillance (NNIS) System Report, data summary from January 1992 through June 2004, issued October 2004. Am. J. Infect. Control 32, 470–485 (2004).

2. Tiemersma EW, Bronzwaer SL, Lyytikainen O et al.: Methicillin-resistant Staphylococcus aureus in Europe, 1999–2002. Emerg. Infect. Dis. 10, 1627–1634 (2004).

3. Moran GJ, Krishnadasan A, Gorwitz RJ et al.: Methicillin-resistant S. aureus infections among patients in the emergency department. N. Engl. J. Med. 355, 666–674 (2006).

4. Tsiodras S, Gold HS, Sakoulas G et al.: Linezolid resistance in a clinical isolate of Staphylococcus aureus. Lancet 358, 207–208 (2001).

5. Weigel LM, Clewell DB, Gill SR et al.: Genetic analysis of a high-level vancomycin-resistant isolate of Staphylococcus aureus. Science 302, 569–1571 (2003).

• High-level vancomycin-resistant Staphylococcus aureus developed by transfer of vanA cluster from Enterococcus faecalis.

6. Tenover FC, Weigel LM, Appelbaum PC et al.: Vancomycin-resistant Staphylococcus aureus isolate from a patient in Pennsylvania. Antimicrob. Agents Chemother. 48, 275–280 (2004).

7. Glass JI, Assad-Garcia N, Alperovich N et al.: Essential genes of a minimal bacterium. Proc. Natl Acad. Sci. USA 103, 425–430 (2006).

8. Salama NR, Shepherd B, Falkow S: Global transposon mutagenesis and essential gene analysis of Helicobacter pylori. J. Bacteriol. 186, 7926–7935 (2004).

9. Song JH, Ko KS, Lee JY et al.: Identification of essential genes in Streptococcus pneumoniae by allelic replacement mutagenesis. Mol. Cells 19, 365–374 (2005).

10. Kobayashi K, Ehrlich SD, Albertini A et al.: Essential Bacillus subtilis genes. Proc. Natl Acad. Sci. USA 100, 4678–4683 (2003).

• Complete evaluation of essential genes in the Gram-positive model organism Bacillus subtilis.

11. Forsyth RA, Haselbeck RJ, Ohlsen KL et al.: A genome-wide strategy for the identification of essential genes in Staphylococcus aureus. Mol. Microbiol. 43, 1387–1400 (2002).

12. Ji Y, Zhang B, Van SF et al.: Identification of critical Staphylococcal genes using conditional phenotypes generated by antisense RNA. Science, 293, 2266–2269 (2001).

13. Zalacain M, Biswas S, Ingraham KA et al.: A global approach to identify novel broad-spectrum antibacterialtargets among proteins of unknown function. J. Mol. Microbiol. Biotechnol. 6, 109–126 (2003).

14. Jana M, Luong TT, Komatsuzawa H et al.: A method for demonstrating gene essentiality in Staphylococcus aureus. Plasmid 44, 100–104 (2000).

15. Zhang L, Fan F, Palmer LM et al.: Regulated gene expression in Staphylococcus aureus for identifying conditional lethal phenotypes and antibiotic mode of action. Gene 255, 297–305 (2000).

16. Rohrer S, Ehlert K, Tschierske M, Labischinski H, Berger-Bachi B: The essential Staphylococcus aureus gene fmhB is involved in the first step of peptidoglycan pentaglycine interpeptide formation. Proc. Natl Acad. Sci. USA , 96, 9351–9356 (1999).

17. Ji Y, Marra A, Rosenberg M, Woodnutt G: Regulated antisense RNA eliminates α-toxin virulence in Staphylococcus aureus infection. J. Bacteriol. 181, 6585–6590 (2001).

18. Streker K, Freiberg C, Brötz-Oesterhelt H et al.: In vitro and in vivo characterization of novel antibiotics targets from Staphylococcus aureus. Antimicrob. Agents Chemother. (2007) (In press).

19. Becker D, Selbach M, Rollenhagen C et al.: Robust Salmonella metabolism limits possibilities for new antimicrobials. Nature 440, 303–307 (2006).

•• Comprehensive analysis of metabolic pathways in Salmonella to define new antimicrobial targets.

20. Wang J, Galgoci A, Kodali S et al.: Discovery of a small molecule thatinhibits cell division by blocking FtsZ, a novel therapeutic target of antibiotics. J. Biol. Chem. 278, 44424–44428 (2003).

21. Margalit DN, Romberg L, Mets RB et al.: Targeting cell division: small-molecule inhibitors of FtsZ GTPase perturb cytokinetic ring assembly and induce bacterial lethality. Proc. Natl Acad. Sci. USA 101, 11821–11826 (2004).

22. Yuan Z, White RJ: The evolution of peptide deformylase as a target: contribution of biochemistry, genetics and genomics. Biochem. Pharmacol. 71, 1042–1047 (2006).

23. Chen D, Yuan Z: Therapeutic potential of peptide deformylase inhibitors. Expert Opin. Investig. Drugs 14, 1107–1116 (2005).

24. Lee MD, She Y, Soskis MJ et al.: Human mitochondrial peptide deformylase, a new anticancer target of actinonin-based antibiotics. J. Clin. Invest. 114, 1107–1116 (2004).

25. Yoon HJ, Kim HL, Lee SK et al.: Crystal structure of peptide deformylase from Staphylococcus aureus in complex with actinonin, a naturally occurring antibacterial agent. Proteins 57, 639–642 (2004).

26. Hackbarth CJ, Chen DZ, Lewis JG et al.: N-alkyl urea hydroxamic acids as a new class of peptide deformylase inhibitors with antibacterial activity. Antimicrob. Agents Chemother. 46, 2752–2764 (2002).

27. Chen D, Hackbarth C, Ni ZJ et al.: Peptide deformylase inhibitors as antibacterial agents: identification of VRC3375, a proline-3-alkylsuccinyl hydroxamate derivative, by using an integrated combinatorial and medicinal chemistry approach. Antimicrob. Agents Chemother. 48, 250–261 (2004).

28. Ramanathan-Girish S, McColm J, Clements JM et al.: Pharmacokinetics in animals and humans of a first-in-class peptide deformylase inhibitor. Antimicrob. Agents Chemother. 48, 4835–4842 (2004).

29. Dubail I, Bigot A, Lazarevic V et al.: Identification of an essential gene of Listeria monocytogenes involved in teichoic acid biogenesis. J. Bacteriol. 188, 6580–6591 (2006).

Page 10: Novel targets for antibiotics in Staphylococcus aureus

664

REVIEW – Ohlsen & Lorenz

Future Microbiol. (2007) 2(6) future science groupfuture science group

30. Bhavsar AP, Erdman LK, Schertzer JW, Brown ED: Teichoic acid is anessential polymer in Bacillus subtilisthat is functionally distinct from teichuronic acid. J. Bacteriol. 186, 7865–7873 (2004).

31. D’Elia MA, Millar KE, Beveridge TJ, Brown ED: Wall teichoic acid polymers are dispensable for cell viability in Bacillus subtilis. J. Bacteriol. 188, 8313–8316 (2006).

32. D’Elia MA, Pereira MP, Chung YS et al.: Lesions in teichoic acid biosynthesis in Staphylococcus aureus lead to a lethal gain of function in the otherwise dispensable pathway. J. Bacteriol. 188, 4183–4189 (2006).

33. Weidenmaier C, Kokai-Kun JF, Kristian SA et al.: Role of teichoic acids in Staphylococcus aureus nasal colonization, a major risk factor in nosocomial infections. Nat. Med. 10, 243–245 (2004).

34. Weidenmaier C, Peschel A, Kempf VA, Lucindo N, Yeaman MR, Bayer AS: DltABCD- and MprF-mediated cell envelope modifications of Staphylococcus aureus confer resistance to platelet microbicidal proteins and contribute to virulence in a rabbit endocarditis model. Infect. Immun. 73, 8033–8038 (2005).

35. Payne DJ, Gwynn MN, Holmes DJ, Pompliano DL: Drugs for bad bugs: confronting the challenges of antibacterial discovery. Nat. Rev. Drug Discov. 6, 29–40 (2007).

36. Uziel O, Borovok I, Schreiber R, Cohen G, Aharonowitz Y: Transcriptional regulation of the Staphylococcus aureus thioredoxin and thioredoxin reductase genes in response to oxygen and disulfide stress. J. Bacteriol. 186, 326–334 (2004).

37. Cregg KM, Wilding I, Black MT: Molecular cloning and expression of the spsB gene encoding an essential type Isignal peptidase from Staphylococcus aureus. J. Bacteriol. 178, 5712–5718(1996).

38. Garmory HS, Titball RW: ATP-binding cassette transporters are targets for the development of antibacterial vaccines and therapies. Infect. Immun. 72, 6757–6763 (2004).

39. Kaczmarek FS, Zaniewski RP, Gootz TD et al.: Cloning and functional characterization of an NAD+-dependent DNA ligase from Staphylococcus aureus. J. Bacteriol. 183, 3016–3024 (2001).

40. Sun J, Zheng L, Landwehr C, Yang J, Ji Y: Identification of a novel essential two-component signal transduction system, YhcSR, in Staphylococcus aureus. J. Bacteriol. 187, 7876–7880 (2005).

41. Zheng L, Yang J, Landwehr C, Fan F, Ji Y: Identification of an essential glycoprotease in Staphylococcus aureus. FEMS Microbiol. Lett. 245, 279–285 (2005).

42. Dubrac S, Msadek T: Identification ofgenes controlled by the essential YycG/YycF two-component system of Staphylococcus aureus. J. Bacteriol. 186, 1175–1181 (2004).

43. Bruton G, Huxley A, O’Hanlon PJ et al.: Lipopeptide substrates for SpsB, the Staphylococcus aureus type I signal peptidase: design, conformation and conversion toα-ketoamide inhibitors. Eur. J. Med. Chem. 38, 351–356 (2003).

44. Payne DJ, Miller WH, Berry V et al.: Discovery of a novel and potent class of FabI-directed antibacterial agents. Antimicrob. Agents Chemother. 46, 3118–3124 (2002).

45. Wang J, Soisson SM, Young K et al.: Platensimycin is a selective FabF inhibitor with potent antibiotic properties. Nature 441, 358–361 (2006).

• Description of a new class of fatty acid synthesis inhibitor.

46. Wang J, Kodali S, Lee SH et al.: Discovery of platencin, a dual FabF and FabH inhibitor with in vivo antibiotic properties. Proc. Natl Acad. Sci. USA 104, 7612–7616 (2007).

47. Moir DT: Identification of inhibitors of bacterial enoyl-acyl carrier protein reductase. Curr. Drug Targets Infect. Disord. 5, 297–305 (2005).

48. Ton-That H, Labischinski H, Berger-Bachi B, Schneewind O: Anchor structure of staphylococcal surface proteins. III. Role of the FemA, FemB, and FemX factors in anchoring surface proteins to the bacterial cell wall. J. Biol. Chem. 273, 29143–29149 (1998).

49. Lovering AL, de Castro LH, Lim D, Strynadka NC: Structural insight into the transglycosylation step of bacterial cell-wall biosynthesis. Science 315, 1402–1405 (2007).

•• First 3D structure of a bifunctional transglycosylase-transpeptidase, which should aid development of new inhibitors of the transglycosylase domain.

50. Harris MS, Herberg JT, Cialdella JI et al.: Crystallization and preliminary x-ray analysis of UDP-N-acetylenolpyruvylglucosamine reductase (MurB) from Staphylococcus aureus. Acta Crystallogr. D. Biol. Crystallogr. 57, 1032–1035 (2001).

51. Koch AL: Bacterial wall as target for attack: past, present, and future research. Clin. Microbiol. Rev. 16, 673–687 (2003).

52. Breukink E, de Kruijff B: Lipid II as a target for antibiotics. Nat. Rev. Drug Discov. 5, 321–332 (2006).

53. Beyer D, Kroll HP, Endermann Ret al.: New class of bacterial phenylalanyl-tRNA synthetase inhibitors with high potency and broad-spectrum activity. Antimicrob. Agents Chemother. 2004, 48, 525–532 (2004).

54. Hurdle JG, O’Neill AJ, Chopra I: Prospects for aminoacyl-tRNA synthetase inhibitors as new antimicrobial agents. Antimicrob. Agents Chemother. 49, 4821–4833 (2005).

55. Ochsner UA, Sun X, Jarvis T, Critchley I, Janjic N: Aminoacyl-tRNA synthetases: essential and still promising targets for new anti-infective agents. Expert Opin. Investig. Drugs 16, 573–593 (2007).

56. Henkel T, Finlay J: Emergence of resistance during mupirocin treatment: is it a problem in clinical practice? J. Chemother. 11, 331–337 (1999).

57. Grossman TH, Bartels DJ, Mullin S et al.: Dual targeting of GyrB and ParE by a novel aminobenzimidazole class of antibacterial compounds. Antimicrob. Agents Chemother. 51, 657–666 (2007).

58. Mani N, Gross CH, Parsons JD et al.: In vitro characterization of the antibacterial spectrum of novel bacterialtype II topoisomerase inhibitors of the aminobenzimidazole class. Antimicrob.Agents Chemother. 50, 1228–1237 (2006).

59. Projan SJ, Nesin M, Dunman PM: Staphylococcal vaccines and immunotherapy: to dream the impossible dream? Curr. Opin. Pharmacol. 6, 473–479 (2006).

• Excellent review on current status of staphylococcal vaccines and immunotherapeutical approaches.

60. Götz F: Staphylococci in colonization and disease: prospective targets for drugs and vaccines. Curr. Opin. Microbiol. 7, 477–487 (2004).

61. Foster TJ, Höök M: Surface protein adhesins of Staphylococcus aureus. Trends Microbiol. 6, 484–488 (1998).

62. Rivas JM, Speziale P, Patti JM, Höök M: MSCRAMM-targeted vaccines and immunotherapy for staphylococcal infection. Curr. Opin. Drug Discov. Devel. 7, 223–227 (2004).

Page 11: Novel targets for antibiotics in Staphylococcus aureus

665

Novel targets for antibiotics in Staphylococcus aureus – REVIEW

future science groupfuture science group www.futuremedicine.com

63. Vernachio JH, Bayer AS, Ames B et al.: Human immunoglobulin G recognizing fibrinogen-binding surface proteins is protective against both Staphylococcus aureus and Staphylococcus epidermidis infections in vivo. Antimicrob. Agents Chemother. 50, 511–518 (2006).

64. Bloom BT: INH-A21: a donor-selected staphylococcal human immune globulin for the prevention of late-onset neonatal staphylococcal infection. Expert Opin. Investig. Drugs 15, 703–707 (2006).

65. Kaufman D: Veronate (Inhibitex®). Curr. Opin. Investig. Drugs 7, 172–179 (2006).

66. Fattom AI, Horwith G, Fuller S, Propst M, Naso R: Development of StaphVAX®, a polysaccharide conjugate vaccine against S. aureus infection: from the lab bench to Phase III clinical trials. Vaccine 22, 880–887 (2004).

67. Verdier I, Durand G, Bes M et al.: Identification of the capsular polysaccharides in Staphylococcus aureus clinical isolates by PCR and agglutination tests. J. Clin. Microbiol. 45, 725–729 (2007).

68. von Eiff C, Taylor KL, Mellmann A et al.: Distribution of capsular and surface polysaccharide serotypes of Staphylococcus aureus. Diagn. Microbiol. Infect. Dis. 58, 297–302(2007).

69. Weems JJ Jr, Steinberg JP, Filler S et al.: Phase II, randomized, double-blind, multicenter study comparing the safety and pharmacokinetics of tefibazumab to placebo for treatment of Staphylococcus aureus bacteremia. Antimicrob. Agents Chemother. 50, 2751–2755 (2006).

70. Reilley S, Wenzel E, Reynolds L, Bennett B, Patti JM, Hetherington S: Open-label, dose escalation study of the safety and pharmacokinetic profile of tefibazumab in healthy volunteers. Antimicrob. Agents Chemother. 49, 959–962 (2005).

71. Patti JM: A humanized monoclonal antibody targeting Staphylococcusaureus. Vaccine 22(Suppl. 1), S39–S43 (2004).

72. Burnie JP, Matthews RC, Carter T et al.: Identification of an immunodominant ABC transporter in methicillin-resistant Staphylococcus aureus infections. Infect. Immun. 68, 3200–3209 (2000).

73. Baker M: Anti-infective antibodies: finding the path forward. Nat. Biotechnol. 24, 1491–1493 (2006).

74. Zhou H, Xiong ZY, Li HP, Zheng YL, Jiang YQ: An immunogenicity study of a newly fusion protein Cna-FnBP vaccinated against Staphylococcus aureus infections in a mice model. Vaccine 24, 4830–4837 (2006).

75. Schaffer AC, Solinga RM, Cocchiaro J et al.: Immunization with Staphylococcus aureus clumping factor B, a major determinant in nasal carriage, reduces nasal colonization in a murine model. Infect. Immun. 74, 2145–2153 (2006).

76. Kuklin NA, Clark DJ, Secore S et al.: A novel Staphylococcus aureus vaccine: iron surface determinant B induces rapid antibody responses in rhesus macaques and specific increased survival in a murine S. aureus sepsis model. Infect. Immun. 74, 2215–2223 (2006).

77. Gaudreau MC, Lacasse P, Talbot BG: Protective immune responses to a multi-gene DNA vaccine against Staphylococcus aureus. Vaccine 25, 814–824 (2007).

78. Stranger-Jones YK, Bae T, Schneewind O: Vaccine assembly from surface proteins of Staphylococcus aureus. Proc. Natl Acad. Sci. USA 103, 16942–16947 (2007).

• Development of a vaccine based on four surface proteins of S. aureus showing complete protection in mice.

79. Maira-Litran T, Kropec A, Goldmann DA, Pier GB: Comparative opsonic and protective activities of Staphylococcus aureus conjugate vaccines containing native or deacetylated staphylococcal poly-N-acetyl-β-(1–6)-glucosamine. Infect. Immun. 73, 6752–6762 (2005).

80. Chavakis T, Wiechmann K, Preissner KT Herrmann M: Staphylococcus aureus interactions with the endothelium:the role of bacterial ‘secretable expanded repertoire adhesive molecules’ (SERAM)in disturbing host defense systems. Thromb. Haemost. 94, 278–285 (2005).

81. Spellberg B, Powers JH, Brass EP, Miller LG, Edwards JE Jr: Trends in antimicrobial drug development: implications for the future. Clin. Infect. Dis. 38, 1279–1286 (2004).

82. Projan SJ: Why is big Pharma gettingout of antibacterial drug discovery? Curr. Opin. Microbiol. 6, 427–430 (2003).

83. Norrby SR, Nord CE, Finch R: Lack of development of new antimicrobial drugs: a potential serious threat to public health. Lancet Infect. Dis. 5, 115–119 (2005).

84. Kopp U, Roos M, Wecke J, Labischinski H: Staphylococcal peptidoglycan interpeptide bridge biosynthesis: a novel antistaphylococcal target? Microb. Drug Resist. 2, 29–41 (1996).

85. Liu S, Chang JS, Herberg JT et al.: Allosteric inhibition of Staphylococcus aureus D-alanine:D-alanine ligase revealed by crystallographic studies. Proc. Natl Acad. Sci. USA 103, 15178–15183 (2006).

86. Zheng L, Yu C, Bayles K, Lasa I, Ji Y: Conditional mutation of an essential putative glycoprotease eliminates autolysis in Staphylococcus aureus. J. Bacteriol. 189, 2734–2742 (2007).

87. Brötz-Oesterhelt H, Beyer D,Kroll HP et al.: Dysregulation of bacterial proteolytic machinery by a new class of antibiotics. Nat. Med. 11, 1082–1087 (2005).

88. Brötz-Oesterhelt H, Knezevic I, Bartel S et al.: Specific and potent inhibition of NAD+-dependent DNAligase by pyridochromanones. J. Biol. Chem. 278, 39435–39442 (2003).

89. Lee JC, Park JS, Shepherd SE, Carey V, Fattom A: Protective efficacy of antibodies to the Staphylococcus aureus type 5 capsular polysaccharide in a modified model of endocarditis in rats. Infect. Immun. 65, 4146–4151 (1997).

90. O’Riordan K, Lee JC: Staphylococcus aureus capsular polysaccharides. Clin. Microbiol. Rev. 17, 218–234 (2004).

91. Clarke SR, Brummell KJ, Horsburgh MJ et al.: Identification of in vivo-expressed antigens of Staphylococcus aureus and their use in vaccinations for protection against nasal carriage. J. Infect. Dis. 193, 1098–1108 (2006).

92. Kelly-Quintos C, Cavacini LA, Posner MR, Goldmann D, Pier GB: Characterization of the opsonic and protective activity against Staphylococcus aureus of fully human monoclonal antibodies specific for the bacterial surface polysaccharide poly-N-acetylglucosamine. Infect. Immun. 74, 2742–2750 (2006).

93. Rennermalm A, Li YH, Bohaufs L et al.: Antibodies against a truncated Staphylococcus aureus fibronectin-binding protein protect against dissemination of infection in the rat. Vaccine 19, 3376–3383 (2001).

94. Shkreta L, Talbot BG, Diarra MS, Lacasse P: Immune responses to a DNA/protein vaccination strategy against Staphylococcus aureus induced mastitis in dairy cows. Vaccine 23, 114–126 (2004).

Page 12: Novel targets for antibiotics in Staphylococcus aureus

666

REVIEW – Ohlsen & Lorenz

Future Microbiol. (2007) 2(6) future science groupfuture science group

95. Menzies BE: The role of fibronectin binding proteins in the pathogenesis of Staphylococcus aureus infections. Curr. Opin Infect. Dis. 16, 225–229 (2003).

96. Ladhani S: Understanding the mechanism of action of the exfoliative toxins of Staphylococcus aureus. FEMS Immunol. Med. Microbiol. 39, 181–189 (2003).

97. McCormick JK, Yarwood JM, Schlievert PM: Toxic shock syndrome and bacterial superantigens: an update. Annu. Rev. Microbiol. 55, 77–104 (2001).

98. Ferry T, Perpoint T, Vandenesch F, Etienne J: Virulence determinants in Staphylococcus aureus and their involvement in clinical syndromes. Curr. Infect. Dis. Rep. 7, 420–428 (2005).

• Excellent review discussing therole of virulence factors of S. aureus in diseases.

99. Nilsson IM, Hartford O, Foster T, Tarkowski A: α-toxin and γ-toxin jointly promote Staphylococcus aureus virulence in murine septic arthritis. Infect. Immun. 67, 1045–1049 (1999).

100. Gillet Y, Issartel B, Vanhems P et al.: Association between Staphylococcus aureus strains carrying gene for Panton–Valentine leukocidin and highly lethal necrotising pneumonia in young immunocompetent patients. Lancet 359, 753–759 (2002).

101. De Kimpe SJ, Kengatharan M, Thiemermann C, Vane JR: The cell wall components peptidoglycan and lipoteichoic acid from Staphylococcus aureus act in synergy to cause shock and multiple organ failure. Proc. Natl Acad. Sci. USA 92, 10359–10363 (1995).

102. Lorenz U, Ohlsen K, Karch H, Hecker M, Thiede A, Hacker J: Human antibody response during sepsis against targets expressed by methicillin resistant Staphylococcus aureus. FEMS Immunol. Med. Microbiol. 29, 145–153 (2000).

Website201. Staphylococcus aureus vaccine development

on track– safe and immunogenic in Phase I clinical Trials. Intercell AG. www.intercell.com/images/content/binaries/952c2eba-0622–42e3-a7f0–5b831e83bb3b.pdf

Affiliations• Knut Ohlsen

University of Würzburg, Institute for Molecular Infection Biology, Röntgenring 11,97070 Würzburg, GermanyTel.: +49 931 312 155;Fax: +49 931 312 578;[email protected]

• Udo LorenzUniversity of Würzburg, Centre for Operative Medicine, Department of Surgery I,Oberdürrbacher Str. 6, 97080 Würzburg, GermanyTel.: +49 931 2013 8513;Fax: +49 931 2013 8609;[email protected]