Community-Associated Methicillin-Resistant Staphylococcus ...antimicrobe.org/Rybak-MRSA...

12
Community-Associated Methicillin-Resistant Staphylococcus aureus: A Review Michael J. Rybak, Pharm.D., and Kerry L. LaPlante, Pharm.D. Methicillin-resistant Staphylococcus aureus (MRSA) is a common bacterial pathogen responsible for a variety of infections in both children and adults. Treatment of infections caused by this organism is problematic due to its resistance to many drugs. Recent reports of community-associated MRSA (CA-MRSA) infections in patients with no known risk factors have serious public health implications. Therapeutic options for these infections are untested; therefore, the potential exists for high morbidity and mortality. Recently, clinical definitions have been established, and new molecular approaches have allowed investigators to distinguish CA-MRSA more easily from traditional nosocomial-derived MRSA strains. Identifying potential risk factors for CA-MRSA acquisition and fully characterizing the epidemiologic, clinical, and molecular properties of these strains are necessary to provide effective therapeutic guidelines. Keywords: community-associated methicillin-resistant Staphylococcus aureus, community-associated MRSA, CA-MRSA, SCCmec IV, staphylococcal cassette chromosome, Panton-Valentine leucocidin, MRSA, disk diffusion test, clindamycin, daptomycin, trimethoprim-sulfamethoxazole. (Pharmacotherapy 2005;25(1):74–85) OUTLINE Epidemiology Risk Factors Molecular Analysis of Community- and Health Care–Associated MRSA Virulence Factors and Toxins Outbreaks Treatment Options Nonantimicrobial Treatment Option Conclusion Methicillin-resistant Staphylococcus aureus (MRSA) is a formidable bacterial pathogen responsible for a variety of infections commonly seen in patients of all ages. 1–3 Acquisition of this organism is typically associated with particular settings (health care institutions, such as hospitals and long-term care facilities) and patient groups (patients with prolonged hospitalization, past antimicrobial use, indwelling catheters, decubitis ulcers, postoperative surgical wounds, and use of intravenous drugs or treatment with enteral feedings or dialysis). 4–7 Infections due to MRSA present a considerable dilemma to clinicians, since therapeutic options are limited and suboptimal dosing contributes to heightened mortality and increased length of hospital stay. 6, 8, 9 Although alteration of target penicillin-binding proteins is the primary mechanism of resistance to -lactam antibiotics, over the years MRSA strains have gained multiple mechanisms of resistance to several classes of antimicrobials, From the Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, and the School of Medicine, Wayne State University, Detroit, Michigan (Dr. Rybak); Detroit Receiving Hospital and University Health Center, Detroit, Michigan (Dr. Rybak); and the Department of Pharmacy Practice, University of Rhode Island, Kingston, Rhode Island (Dr. LaPlante). Address reprint requests to Michael J. Rybak, Pharm.D., Anti-Infective Research Laboratory, Pharmacy Practice – 4148, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, 259 Mack Avenue, Detroit, MI 48201; e-mail: [email protected]. Complete Table of Contents for Pharmacotherapy Subscription Information for Pharmacotherapy

Transcript of Community-Associated Methicillin-Resistant Staphylococcus ...antimicrobe.org/Rybak-MRSA...

Community-Associated Methicillin-ResistantStaphylococcus aureus: A Review

Michael J. Rybak, Pharm.D., and Kerry L. LaPlante, Pharm.D.

Methicillin-resistant Staphylococcus aureus (MRSA) is a common bacterialpathogen responsible for a variety of infections in both children and adults.Treatment of infections caused by this organism is problematic due to itsresistance to many drugs. Recent reports of community-associated MRSA(CA-MRSA) infections in patients with no known risk factors have seriouspublic health implications. Therapeutic options for these infections areuntested; therefore, the potential exists for high morbidity and mortality.Recently, clinical definitions have been established, and new molecularapproaches have allowed investigators to distinguish CA-MRSA more easilyfrom traditional nosocomial-derived MRSA strains. Identifying potential riskfactors for CA-MRSA acquisition and fully characterizing the epidemiologic,clinical, and molecular properties of these strains are necessary to provideeffective therapeutic guidelines.Keywords: community-associated methicillin-resistant Staphylococcus aureus,community-associated MRSA, CA-MRSA, SCCmec IV, staphylococcal cassettechromosome, Panton-Valentine leucocidin, MRSA, disk diffusion test,clindamycin, daptomycin, trimethoprim-sulfamethoxazole.(Pharmacotherapy 2005;25(1):74–85)

OUTLINE

EpidemiologyRisk FactorsMolecular Analysis of Community- and Health

Care–Associated MRSAVirulence Factors and ToxinsOutbreaksTreatment OptionsNonantimicrobial Treatment OptionConclusion

Methicillin-resistant Staphylococcus aureus(MRSA) is a formidable bacterial pathogenresponsible for a variety of infections commonlyseen in patients of all ages.1–3 Acquisition of thisorganism is typically associated with particularsettings (health care institutions, such ashospitals and long-term care facilities) andpatient groups (patients with prolongedhospitalization, past antimicrobial use, indwellingcatheters, decubitis ulcers, postoperative surgicalwounds, and use of intravenous drugs ortreatment with enteral feedings or dialysis).4–7

Infections due to MRSA present a considerabledilemma to clinicians, since therapeutic optionsare limited and suboptimal dosing contributes toheightened mortality and increased length ofhospital stay.6, 8, 9

Although alteration of target penicillin-bindingproteins is the primary mechanism of resistanceto b-lactam antibiotics, over the years MRSAstrains have gained multiple mechanisms ofresistance to several classes of antimicrobials,

From the Anti-Infective Research Laboratory, Departmentof Pharmacy Practice, Eugene Applebaum College ofPharmacy and Health Sciences, and the School of Medicine,Wayne State University, Detroit, Michigan (Dr. Rybak);Detroit Receiving Hospital and University Health Center,Detroit, Michigan (Dr. Rybak); and the Department ofPharmacy Practice, University of Rhode Island, Kingston,Rhode Island (Dr. LaPlante).

Address reprint requests to Michael J. Rybak, Pharm.D.,Anti-Infective Research Laboratory, Pharmacy Practice –4148, Eugene Applebaum College of Pharmacy and HealthSciences, Wayne State University, 259 Mack Avenue,Detroit, MI 48201; e-mail: [email protected].

Complete Table of Contents for Pharmacotherapy Subscription Information for Pharmacotherapy

COMMUNITY-ASSOCIATED MRSA Rybak and LaPlante

including macrolides, aminoglycosides, fluoro-quinolones, tetracyclines, and lincosamideantibiotics such as clindamycin. For the pastseveral decades, glycopeptide antibiotics, such asvancomycin, have been considered the onlyagents to which MRSAs have not developedresistance. Unfortunately, due to overuse ofglycopeptide antibiotics, MRSAs have emergedwith reduced susceptibility to these agents aswell.10, 11

In recent years, there have been several reportsof community-associated MRSA (CA-MRSA)infections throughout the world, includingseveral outbreaks in the United States.12–15 Mostof these outbreaks have been associated with asingle-clone strain. Transmission has occurredby close physical contact in situations involvingchildren in day-care centers, children and adultson Indian reservations, athletes, militarypersonnel, correctional facilities, and men havingsex with men.16–18 Of concern, these patients areotherwise healthy individuals with no knownrisk factors for MRSA acquisition.3, 7, 13, 15, 19–26

The prevalence of MRSA infections is increasingin the community.27–29 Recent investigations haverevealed several characteristics that differentiateCA-MRSA from health care–associated MRSA(HA-MRSA) strains. Community isolates tend tobe susceptible to a variety of non–b-lactamantibiotics, whereas HA-MRSAs are typicallyresistant to multiple antibiotics. Other differencesare that genotypes of community isolates are notthe same as those of health care–derived isolates,community strains harbor a novel methicillinresistance cassette gene element not identified todate among strains that are endemic to healthcare setting, and community isolates occur inpatients lacking typical risk factors for MRSA.1–5

Finally, community isolates are more likely thanhealth care–derived isolates to encode putativevirulence factors, such as Panton-Valentineleucocidin, a cytotoxin virulence factor that hasbeen associated with severe pneumonia inchildren and with skin and soft tissue infectionsin adults.

The Centers for Disease Control andPrevention (CDC) has established criteria todistinguish CA-MRSA from HA-MRSA isolates.30

According to these criteria, the diagnosis of CA-MRSA must be made in an outpatient setting orby culture showing MRSA within 48 hours afteradmission to the hospital. The patient must nothave experienced any of the following during theyear before infection: hospitalization; admissionto a nursing home, skilled nursing facility, or

hospice; dialysis; or surgery. In addition, thepatient must be without permanent indwellingcatheters or medical devices that pass throughthe skin into the body.

Epidemiology

In 2000, the CDC began working closely withfour states with a combined population of about12 million persons to study the epidemiology ofCA-MRSA infections. Information from thesestudies is helping the CDC understand the natureof the disease, the reasons why people getinfected, and the types of research needed to helpprevent these infections. These data are beingcollected in Connecticut, Minnesota, Georgia,and Maryland as part of CDC’s EmergingInfections Program. This program was expandedto six states in 2004.

In one of the more recent studies, a group ofinvestigators characterized the epidemiologic andmicrobiologic characteristics of both CA- andHA-MRSA in 1100 MRSA infections.31 Based onpulsed field gel electrophoresis (PFGE) andstaphylococcal exotoxin gene testing, theydetermined that 12% of the infecting strains wereCA-MRSA and 85% were HA-MRSA. Seventy-five percent of skin and soft tissue infectionswere caused by CA-MRSA, whereas 37% werecaused by HA-MRSA (odds ratio [OR] 4.25; 95%confidence interval [CI] 2.97–5.90). The CA-MRSA isolates typically possessed differentexotoxin gene profiles than the HA-MRSAisolates.

Earlier studies investigated the epidemiologyand clonality of CA-MRSA in Minnesota healthcare facilities.23 Ten health care facilitiescontributed data on 354 patients with CA-MRSAfrom 1996–1998. Patient records were examinedfor demographic data, and all infection typeswere recorded. All available isolates wereanalyzed with PFGE to determine if they were ofhospital or community origin. The median ageof patients was 16 years (range 1–78 yrs), and themost common infection type was skin and softtissue (84%). Examination of the isolates byPFGE indicated that most (86%) were distinctlydifferent from HA-MRSA organisms. In addition,as has been found in other evaluations of CA-MRSA strains, these pathogens tended to be moresusceptible to antimicrobial agents than HA-MRSA.

To estimate the prevalence of CA-MRSAinfections in Finland, a study evaluated MRSAculture–positive patients from a national hospital

75

PHARMACOTHERAPY Volume 25, Number 1, 2005

discharge register.24 The definition for CA-MRSAwas the lack of hospitalization for a minimum of2 years. Of the 526 patients identified withMRSA-positive status, 21% were determined tohave MRSA of community origin. Three MRSAstrains were identified as community strains onthe basis of phage typing, PFGE, and ribotyping.Of interest, none of the community-acquiredstrains were multidrug resistant, and all strainsdemonstrated a mec hypervariable region. Theauthors concluded that CA-MRSA may arise denovo, through acquisition of the mecA gene.

A hospital-based observational study comparednosocomial and community-acquired S. aureusbacteremias.22 The researchers classified 32% ofall bacteremias as hospital acquired, whereas18.5% were deemed community acquired.However, on further examination, all patientswith CA-MRSA were found to have regularcontact with health care settings, making theterm community-acquired misleading.

A meta-analysis applied to various types of CA-MRSA publications yielded several sets of keystatistics.32 In nine studies where researchersobtained culture samples before making riskassessments, the pooled MRSA colonization ratewas 2.1% (among 4825 patients). Anotherrevealing finding was that nearly one half ofpatients with CA-MRSA had one or more riskfactors for HA-MRSA, and among the remaining3525 patients the colonization rate of CA-MRSAstrains dropped to only 0.2%. Patients fromwhom samples were obtained in a health carefacility were 2.4 times more likely to carry MRSAthan community members cultured outside of ahealth care setting (95% CI 1.56–3.53). Finally,17.8% of household contacts of patientscolonized with HA-MRSA were found to alsocarry the index strain. To date, the trueincidence of CA-MRSA is unknown, since moststudies have characterized this organism in arelatively small group of patients over a short,fixed time interval.

Risk Factors

It is of vital importance to identify risk factorsfor CA-MRSA to enable clinicians to rapidlyrecognize and appropriately treat infectedpatients.23, 33 Several studies have demonstratedan increase in the prevalence of CA-MRSA; however,risk factors have not been fully characterized.3, 7, 15,

19–21, 23, 25, 34 Children and young adults haveserved as the primary patient source for asignificant number of these studies.23, 35 Other

than injection drug use, no other community-associated risk factors have been identified.7, 36

Unfortunately, great inconsistencies exist in thedefinitions of CA-MRSA in these investigations.Early studies did not discuss exclusion criteria,whereas other studies discussed exclusion criteriafor health care contact ranging from a few monthsto up to 1 year after MRSA colonization.15, 21, 23, 34, 37

Most report that routine contact with health carefacilities was a significant risk factor for CA-MRSA acquisition.

Molecular Analysis of Community- and HealthCare–Associated MRSA

Several investigations have explored themolecular aspects of CA-MRSA. One researchgroup investigated the potential for a sharedcommon origin of HA-MRSA and CA-MRSA.38

Twenty-three well characterized strains of CA-MRSA were compared with 12 non–multidrug-resistant, oxacillin-resistant S. aureus (NORSA)strains—strains that are frequently isolated inhospitals but are considered to be decedents ofCA-MRSA isolates—and with representativehospital isolates. Most but not all of the CA-MRSA strains were susceptible to a variety ofnon–b-lactams, as was generally the case withNORSA isolates. This indicated that CA-MRSAstrains can acquire resistance to non–b-lactamsthrough exposure. The CA-MRSA strains werealso found to have lower levels of resistance tooxacillin and imipenem-cilastatin (minimuminhibitory concentrations of 8–32 µg/ml), implyingthat CA-MRSA strains, unlike HA-MRSA strains,were not selected by exposure to the potent b-lactam agents typically used in hospital settings.It was also observed that doubling times weresignificantly shorter for CA-MRSA than for HA-MRSA. The authors speculated that this highgrowth rate may help CA-MRSA achievesuccessful colonization by enabling it tooutcompete other bacterial species that arenormally part of the commensal flora.

Application of multilocus sequence typing andevolutionary mathematical models to aninternational collection of 359 MRSA isolates hasrevealed that all MRSAs share a common geneticlineage and can be traced to a single MRSA clone.This strain, known as ST-250, appears to haveevolved from a methicillin-susceptible S. aureus(MSSA) isolate, which subsequently acquired themec gene from an unknown source. Minorvariations of this organism, such as ST-247; theIberian clone; and ST-5, 22, and 45, have evolved

76

COMMUNITY-ASSOCIATED MRSA Rybak and LaPlante

into some of the more common MRSA isolatesthat have been found around the world. Thediversity of MRSA strains is secondary tohorizontal transfer of genetic elements that insertinto the bacterial genome. Current MRSAisolates are either descendants of preexistingclones or have been created by horizontaltransfer of the mec determinant into successfulMSSA.39 The mecA gene in staphylococci isresponsible for b-lactam resistance. This geneencodes a penicillin-binding protein that has lowaffinity for b-lactam–type antibiotics. The mecAgene complex is carried on a specific integrativegenetic element known as the staphylococcalcassette chromosome (SCC). These mobilecassettes consist of the mec complex and thecassette recombinase genes, which are responsiblefor encoding integration and excision of theSCCmec element on the staphylococcal chromo-some (Figure 1).

Five SCCmec types have been identified for S.

aureus. The gene elements differ in size, compo-sition, and associated antimicrobial resistanceexpression (Table 1).40–42 The SCCmec types I, II,and III are found predominately in HA-MRSAisolates. These isolates carry a number ofinserted plasmids and transposable geneticelements downstream of the mecA complex. TheSCCmec types II and III are responsible for the

77

Figure 1. Structure of the staphylococcal cassette chromosome mec, with the recombinase genes complex upstream of the meccomplex. The mec complex contains the mecA gene responsible for b-lactam resistance in Staphylococcus aureus. IS1272 =insertion sequence–like element; ccrA and ccrB = cassette chromosome recombinase genes A and B that mobilize the mecelement; mecR1 = mec sensor transducer and repressor genes that regulate production of PBP-2A, which is responsible for b-lactam resistance; IS431 = integrated plasmid that encodes tetracycline resistance; and orfx = open reading frame in which themobile elements (staphylococcal cassette chromosome) are located. (Adapted in part from reference 37.)

Table 1. The Five Types of the mec Gene Complex40–42

SCCmec SizeType (kb) FeaturesI 34.3 Lacks other resistance genesII 53.0 Associated with multiple drug

resistanceIII 66.9 Associated with multiple drug

resistanceIV 20.9–24.3 Resistant to b-lactam antibioticsV 28.0 Lacks antibiotic resistance genes

other than mecASCC = staphylococcal cassette chromosome.

PHARMACOTHERAPY Volume 25, Number 1, 2005

multiple non–b-lactam antimicrobial resistanceoften expressed in these health care–relatedstrains. The SCCmec type IV is typically found inCA-MRSA strains and in NORSA isolates. ThisSCCmec type is smaller in size and lacks othermultidrug-resistance genes. Recently, a SCCmectype V was described. Similar to type IV, it issmall in size, does not contain antimicrobialresistance genes other than mecA, and is foundpredominately in CA-MRSA and NORSAstrains.40

Virulence Factors and Toxins

Staphylococcus aureus produces numerousunique toxins and virulence factors, such as thetoxic shock syndrome toxin (TSST-1), entero-toxins, and exotoxins, that can inflict severeclinical syndromes, such as septic shock,necrotizing pneumonia, and complicated skinand soft tissue infections.43 Highly diverse andunique virulence genes appear to be charac-teristics of CA-MRSA that clearly distinguishthem from typical HA-MRSA strains. Genomicstudies have shown that CA-MRSA strains carry arange of virulence genes that are distinct fromthose found in other S. aureus strains. A researchgroup compared 32 CA-MRSA strains withrespect to clonal relatedness and S. aureussuperantigen toxins.35 As expected, most isolates(81%) were susceptible to a wide variety ofnon–b-lactam antimicrobials. Thirty-one of the32 CA-MRSA isolates were highly related asdetermined by PFGE and superantigen testing,and were genetically unrelated to HA-MRSAstrains. The 31 related CA-MRSA isolatesproduced either staphylococcal enterotoxin B orenterotoxin C. No isolates produced TSST-1. Astudy of the MW2 CA-MRSA strain isolated fromNorth Dakota in 1998 identified 18 toxins thatwere not found in any of five comparativehospital-derived strains.44 The genes seh and seo,which encode for superantigen enterotoxins Hand O, were found in close proximity to theSCCmec complex. Enterotoxins H and O haveparticularly high binding affinities for the majorhistocompatibility complex type II molecules andwere reported only in this CA-MRSA isolate. Ofinterest, enterotoxin H is produced in dispropor-tionate amounts compared with other super-antigens and is involved in acute toxic shock–like syndromes.45

Another virulence factor specific to CA-MRSAstrains is the Panton-Valentine leucocidin (PVL)toxin. This is a bicomponent cytotoxin previously

reported to be produced by less than 5% of S.aureus isolates. The toxin is encoded by twogenes, lukS-PV and lukF-PV, which are carried ona bacteriophage that has incorporated itself intothe S. aureus chromosome. It is capable ofdestroying human leukocytes and inflictingsevere tissue damage. It has been associated withnecrotic skin lesions and severe necrotizingpneumonia in both children and adults.46, 47 Oneinvestigator characterized 14 CA-MRSA strainsrecovered from patients in France who had beenhealthy during the 18 months before becominginfected.33 Most patients had skin or soft tissueinfections, and two patients had necrotizingpneumonia. The PVL gene and lukE-lukD(leucocidin genes) were detected in all 14isolates. Earlier studies have shown that PVLgenes are rarely detected in MRSA isolatesassociated with hospital infections.48 Theauthors concluded that the combination of mecAand the PVL gene have created a superadaptableS. aureus strain capable of spreading rapidlythrough the community.35

A recent study investigated the geneticrelatedness of five community-acquired S. aureusisolates obtained from four consecutive pediatricpatients who presented with sepsis syndrome andsevere pneumonia over a 3-week period in2000.37 Two of the patients were infected withthree MSSA isolates, whereas the other two wereinfected with MRSA. The two MRSA strainscontained the SCCmec IV element that charac-terizes CA-MRSA isolates. Of interest, all fiveisolates contained the staphylococcal toxin genessea, seh, and seo and the PVL genes lukS-PV andlukF-PV. Analysis with PFGE revealed only onedifference among the strains: the two MRSAstrains, unlike the MSSA strains, contained twobands reflecting the presence of the SCCmec IVelement that distinguishes CA-MRSA from HA-MSSA. According to the researchers, the geneticrelatedness of these isolates suggests that CA-MRSA infections arose from MSSA isolates thatsuccessfully incorporated the SCCmec IVelement.

A small town in western Switzerland was thesite of a large outbreak of skin infections fromSeptember 1999–2000.43 Twenty-two studentsfrom a single third-grade classroom had 13episodes of skin infections that includedfuruncles, abscesses, and cellulitis. Two patientswere hospitalized, and the rest were treated withsystemic antibiotics, with or without surgicaldrainage. All cultures grew MSSA. One of threeclones isolated was positive for PVL. This clone

78

COMMUNITY-ASSOCIATED MRSA Rybak and LaPlante

was associated with nine persons, consisting ofclassmates, teachers, and family members whowere either nasal carriers or infected. Nasalcarriage was detected only after six students andthree relatives were noted to have relapsingepisodes of skin infections over a 13-monthperiod. The authors suggested that PVL-positiveS. aureus may easily spread between persons inclose contact, infecting otherwise healthychildren and adults. Because of their highvirulence capability, skin infections can rapidlyprogress to severe necrotizing pneumonia with ahigh mortality rate.

Another investigation described the clonaldistribution of PVL-carrying MRSA strains andtheir association with skin and soft tissueinfections in the San Francisco Bay area.49 A totalof 671 isolates collected from inpatients andoutpatients during 1997–2002 were evaluated forstrain relatedness and virulence factors.Approximately 70% of PVL-carrying MRSAstrains were isolated from jail inmates andpatients receiving surgical treatment inoutpatient settings specializing in skin and softtissue infections. Although as many as nineclonal types were identified, the vast majority(88.5%) of PVL-carrying MRSA belonged to onlytwo clonal groups. Of interest, these two clonalgroups also carried the SCCmec IV resistanceelement and were more likely to be associated(p<0.0001) with skin and soft tissue infectionsthan any other infection site. Overall, the datasuggest that most CA-MRSA strains probablyarose from successful MSSA strains thatincorporated a variety of specific virulencefactors, thus improving their ability to colonizeand cause infection.1

Outbreaks

Outbreaks of CA-MRSA were first described inthe early 1980s.7, 50, 51 In the late 1990s,increasing reports began to emerge.18, 24, 48, 52–60

Unfortunately, CA-MRSA is now a commoncommunity-based pathogen. It has demonstratedgreat geographic diversity, with outbreaksreported in the Unites States, Canada, Europe,Finland, Saudi Arabia, India, Asia, Australia, andNew Zealand.24, 25, 53, 54, 59, 61–67 The strainsinvolved in these outbreaks have in common themec type IV cassette but are genetically distinctfrom one another. Outbreaks of CA-MRSA aretypically characterized by clusters of skin andsoft tissue infections among persons who haveclose contact with one another. Most concerning

is that persons infected are otherwise healthyindividuals with no known risk factors forinfection by drug-resistant bacteria.

The first report of CA-MRSA came from a largeurban Michigan hospital in 1982.68 It describedcommunity-associated S. aureus infections in 24intravenous drug users and 16 nonusers. Allpersons infected were otherwise healthyindividuals with no risk factors for MRSAcolonization. On the basis of PFGE data,investigators proposed that MRSA infection arosein the community as well as in the hospital andhad the potential to disseminate in both settings.It has been subsequently noted that intravenousdrug users have frequent contact with health careinstitutions, and the strains they are colonizedwith may have originated in the hospital.

During the 1980s and through the mid 1990s,CA-MRSA infections were infrequent inpopulations other than intravenous drug users.However, in October 2001, the Mississippi StateDepartment of Health notified the CDC that 31prison inmates had acquired MRSA skin or softtissue infections.56 This number was unexpectedlyhigh for a non–health care setting. The nextyear, inmates in the Los Angeles County Jailbegan reporting a high frequency of spider bites.Further investigation revealed that these so-called “bites” were actually MRSA skininfections. Also that same year, the Los AngelesCounty Department of Health Services investigatedcases of invasive MRSA infection in two athleteson the same team who were hospitalized. Inaddition, outbreaks among men having sex withmen were also reported.56 In 2003, researchersdescribed an outbreak of MRSA skin infectionsamong a military beneficiary population.69 Anadditional study reported CA-MRSA infections inroommates sharing instruments used forplucking and trimming hair.70 These reports ofinvasive MRSA skin infections emphasized thepotential for rapid spread of the organism withinthe community among individuals who mayacquire it through close personal contact.56, 71, 72

Children are also vulnerable to CA-MRSA. Oneof the first major reports of invasive CA-MRSAinfections occurred in Minnesota and NorthDakota between 1997 and 1999. This outbreakwas associated with four pediatric deaths frominfections that progressed to pneumonia andsepsis syndrome.73

Humans are not the only hosts for CA-MRSA,which has raised concern as a possible emergingzoonotic and veterinary disease. A report in2003 discussed a common clonal mec type IV

79

PHARMACOTHERAPY Volume 25, Number 1, 2005

strain isolated from horses and their caretakers atan equine farm in Ontario. Horses are notknown to harbor S. aureus naturally. Based ontiming of isolation, subtyping, and evaluation ofanimal and human contact, combinations ofhorse-human, human-horse, or horse-horsetransmission were suspected.74

Few surveillance studies have characterizedhow this pathogen develops within thecommunity. One research group conducted S.aureus surveillance during an outbreak in a ruralAlaska village that is not connected to othervillages by roads.46 The researchers used PFGEand PVL production to identify the relatedness ofthe isolates. They discovered that patients livingin a community associated with an outbreak hadreceived more antibiotic courses during the past12 months then patients in a nonoutbreak setting(p=0.01). Also, individuals involved in theoutbreak were more likely to use a sauna that wasknown to be colonized by MRSA. The investi-gators determined that the likely cause for MRSAcolonization was the sauna’s plywood, which wassemiporous and had an irregular shape thatallowed for biofilm formation attachment andhence MRSA survival at high temperatures.

Due to the rise in CA-MRSA outbreaks, theCDC has published recommendations to preventthe spread of MRSA among persons living in thesame household.30 These recommendationsinclude covering infections that drain or producepus, washing hands frequently, avoiding thesharing of personal items (e.g., towels, washcloth,razor, clothing, or uniforms), washing soiledlinens and clothes with hot water, and dryingclothes in a hot dryer rather than air-drying.

Treatment Options

Although the epidemiology of CA-MRSA hasbeen widely explored, therapeutic managementof this infection has not been well studied and isnot well established. Infections caused by CA-MRSA fall into a broad spectrum, ranging fromuncomplicated skin and soft tissue infections,which can be treated in outpatient settings, tosevere sepsis and toxic shock syndrome, whichrequire hospitalization and aggressive treatment.Because CA-MRSA strains tend to be susceptibleto a wide variety of non–b-lactam antibiotics, itwould seem that several treatment options areavailable (Table 2). However, most potentialtreatments have not been tested clinically, andtheir efficacy is therefore unknown. Moreover,there is great uncertainty about development ofresistance. Researchers have expressed renewedinterest in the use of clindamycin, tetracyclines,and trimethoprim-sulfamethoxazole (TMP-SMX)in treating MRSA infections, as these drugsgenerally have activity against CA-MRSA (Table3).33, 40, 75 For severe infections requiringhospitalization together with intravenousantibiotics, vancomycin and newer agents, suchas linezolid, quinupristin-dalfopristin, anddaptomycin, can be feasible options.

Clindamycin remains a viable treatment optionfor CA-MRSA, as it demonstrates in vitrosusceptibility to most MRSA isolates. However,inducible resistance and treatment failure havebeen reported.76, 77 The disk diffusion (D-test)method can detect S. aureus isolates withinducible macrolide-lincosamide-streptogramin B(iMLSB) resistance.78 However, this is a cumber-

80

Table 2. Antimicrobial Agents with Potential for Use in Treating Methicillin-Resistant Staphylococcus aureus

Agent Traditional Regimens CommentsClindamycin 300–450 mg p.o. q6h Caution is warranted due to iMLSB resistance

600–900 mg i.v. q8h

Daptomycina 4 mg/kg i.v. q24h Approved for use in complicated skin and soft tissue infections

Doxycycline 100 mg i.v. or p.o. q12h Limited clinical data exists in treating MRSA infections

Linezolid 600 mg i.v. or p.o. q12h Approved for complicated skin and soft tissue infections, concernfor overuse and resistance due to oral availability

Quinupristin- 7.5 mg/kg i.v. q12h Caution is warranted due to iMLSB resistance and poor tolerabilitydalfopristin

TMP-SMXa 160 mg p.o. q12hb Caution is warranted in sulfa-allergic patients2.5 mg/kg i.v. q12hb

Vancomycina 1 g i.v. q12h Targeted concentrations are controversialiMLSB = inducible macrolide-lincosamide-streptogramin B; MRSA = methicillin-resistant Staphylococcus aureus; TMP-SMX = trimethoprim-sulfamethoxazole.aDosage should be adjusted for renal function.bDosage is based on the trimethoprim component.

COMMUNITY-ASSOCIATED MRSA Rybak and LaPlante

some procedure, and hospital laboratories havehad difficulty applying this method to each S.aureus isolate. The D-test is conducted witherythromycin-clindamycin disk pairs placed byhand on an agar dish streaked with the isolate inquestion. After incubation, zone diameters aremeasured, and significant ingrowth within a zoneup to the edge of the disk is considered constitutive(already present) resistance. In contrast, flatteningor blunting of the clindamycin zone (D shape)indicates inducible resistance (Figure 2).78

Inducible isolates (those with positive resultson the D-test) are a source of concern becausethey may have a heightened rate of mutations,which would enable them to develop constitutiveresistance to clindamycin during therapy. One

clinical laboratory screened over 150 S. aureusisolates for iMLSB resistance in erythromycin-resistant and clindamycin-susceptible clinical S.aureus isolates and found 56% of isolates to haveinducible resistance by the D-test.76 Littleinformation is available to characterize iMLSBresistance in CA-MRSA. However, a recentinvestigation examined 85 clinical MRSA isolatesfor iMLSB resistance.75 Isolates were evaluatedbased on CDC definition and molecular typing.In SCCmec type II isolates (HA-MRSA), 50% ofstrains harbored iMLSB resistance, whereas only17% of SCCmec type IV isolates (CA-MRSA) hadthis type of resistance pattern. The number ofCA-MRSA strains worldwide harboring thisinducible type of resistance is unknown. It istherefore not recommended to use clindamycinto treat MRSA infections unless the appropriateD-test for iMLSB resistance is conducted on thespecified isolate according to the guidelines ofthe National Committee for Clinical LaboratoryStandards.79, 80

Trimethoprim-sulfamethoxazole is anotherantibiotic with potential for treatment of CA-MRSA. However, limited clinical studies andpatient cases have found TMP-SMX to be usefulin treating MRSA infections. The reported rate ofTMP-SMX resistance in S. aureus is highlyvariable, but most CA-MRSA strains appear to besusceptible. The sulfa moiety of TMP-SMX,sulfamethoxazole, is bacteriostatic, whereas thetrimethoprim component blocks dihydrofolatereductase, thus inhibiting production ofmetabolically active folic acid. When both agentsare used together, TMP-SMX appears to produce

81

Table 3. Phenotypic Expression of SCCmec Types II, III, and IV Isolates

MIC Ranges (mg/L)mec IV mec II and III

(CA-MRSA) (HA-MRSA)Agent (n=55) (n=30)Daptomycin75 0.0625–1.0 0.0625–2.0Clindamycin75 0.06–0.125.0 0.125 to > 32.0Linezolid75 1.0–4.0 1.0–4.0Erythromycin38, 41, 75 0.5–32.0 > 32.0 to > 512.0Doxycycline 38, 75 0.125–8.0 0.25 to > 512.0Oxacillin38, 41, 75 4–64.0 32.0 to > 512.0Vancomycin75 1.0–4.0 2.0–8.0TMP-SMX75 0.06/1.2 to 1.0/20.0 0.06/1.5 to > 32.0/640.0Imipenem38, 41, 75 0.063–2.0 0.5–128.0Cefaclor75 64–128.0 > 128.0Ciprofloxacin75 0.5–64.0 2.0 to > 64.0MIC = minimum inhibitory concentration; CA-MRSA = community-associated methicillinStaphylococcus aureus; HA-MRSA = health care–associated methicillin Staphylococcusaureus; TMP-SMX = trimethoprim-sulfamethoxazole.

Figure 2. Double disk diffusion test (D-test) demonstratingerythromycin induction of clindamycin resistance. Bluntingof the clindamycin inhibition zone produces a D shape asindicated.

PHARMACOTHERAPY Volume 25, Number 1, 2005

a bactericidal effect against most isolates.One study investigated the use of TMP-SMX in

MRSA bacteremia in intravenous drug users.81

The investigators reported that TMP-SMX may beconsidered as an alternative to vancomycin inselected cases of MRSA infection; however,treatment failures were reported. More recentstudies report allergic reactions to sulfonamideantibiotics in approximately 10% of patients;therefore, caution is warranted in this patientpopulation.82 The development of resistance andthe therapeutic use of TMP-SMX in MRSA havenot been fully established. However, a smallretrospective study suggested that promptincision and drainage, followed by a 2–3-weekcourse of TMP-SMX in combination withrifampin was an effective treatment option forcutaneous infections caused by CA-MRSA.70

Although in vitro testing revealed susceptibilityto TMP-SMX and rifampin, infection recurred ina patient treated with TMP-SMX alone comparedwith the combination of TMP-SMX and rifampin.Resistance to rifampin develops rapidly when it isgiven as monotherapy. Therefore, rifampinshould be considered only when given incombination.

Minocycline is a tetracycline antibiotic that hasbeen used in the past for the treatment ofMRSA.83 Doxycycline, another tetracyclinecompound, has a similar susceptibility profile.Although these agents have been used clinically,limited published data are available on the use oftetracyclines in the treatment of MRSA; therefore,the efficacy of doxycycline and minocycline inthe treatment of MRSA have not been established.

Caution is warranted in using fluoroquinolonesto treat MRSA infections. Historically, overuse offluoroquinolones has been associated with MRSAselection.84, 85 Moreover, use of fluoroquinolonesfor treating MRSA infections is correlated withrapid acquisition of fluoroquinolone-resistantMRSA. The newer fluoroquinolones may have arole in the treatment of complicated skin and softtissue infections, especially in patients who areallergic to penicillin. More clinical studies areneeded before a formal recommendation can bemade.

Until the arrival of newer agents, mostclinicians would agree that vancomycin, aglycopeptide antibiotic, is the drug of choice fortreating serious MRSA infections. However,recent reports have described glycopeptide–intermediately susceptible S. aureus (GISA) andheteroresistant GISA. Vancomycin-resistant S.aureus (VRSA) has been identified in Michigan,

Pennsylvania, and New York.86, 87 Theseorganisms are often not responsive tovancomycin.79, 80

Although vancomycin has been proved as a safeand effective treatment option for MRSA,increasing reports document glycopeptideresistance. These reports have heightenedawareness of the need for newer and moreeffective agents. The relationship between CA-MRSA and heteroresistant GISA has not yet beendescribed.

Linezolid, which in 2000 became the firstoxazolidinone to be approved by the Food andDrug Administration (FDA), has good penetrationinto skin and soft tissue infections and isavailable in an oral formulation. Although it isconsidered a bacteriostatic agent, linezolid hasdemonstrated effectiveness in skin and soft tissueinfections, bacteremia, and pneumonia caused bygram-positive bacteria. Linezolid shows activityagainst MRSA, although S. aureus resistance hasbeen reported. Researchers have expressedconcern that linezolid is being overused.88

Quinupristin-dalfopristin is a streptogramincombination product with a gram-positivespectrum of activity that includes MRSA.Although the FDA has not approved quinu-pristin-dalfopristin for use in MRSA infections,the product is approved for use in complicatedskin and soft tissue infections. The poortolerability profile of quinupristin-dalfopristinlimits its use.89 Moreover, S. aureus maydemonstrate inducible or constitutive MLSB.Cross-resistance to macrolides, lincosamides, andstreptogramin B–type antibiotics by methylationof the ribosomal target is the most commonmechanism of S. aureus resistance to strepto-gramin combination products. If inducibleresistance is present, quinupristin remains activebecause it is not an inducer of the methylase.However, if constitutive, quinupristin is inactive,and quinupristin-dalfopristin becomesbacteriostatic rather than bactericidal. Thus, lossof activity may occur. This process has beendemonstrated both in vitro and in vivo.90, 91

Daptomycin is a novel lipopeptide antibioticapproved by the FDA in 2003 for treatment ofcomplicated skin and soft tissue infections,including those caused by MRSA. This drug hasreceived much interest because of its activityagainst multidrug-resistant gram-positivebacteria, especially MRSA.92 Daptomycin hasdemonstrated rapid bactericidal (99.9% kill)activity in several in vitro and animal pharmaco-dynamic studies.71, 93 Although considered a

82

COMMUNITY-ASSOCIATED MRSA Rybak and LaPlante

viable treatment option, its role in the treatmentof CA-MRSA has not yet been fully established.

Overall, there appear to be several antimicrobialtherapeutic options for treating CA-MRSAinfections. However, as stated above, no definitivestudies have tested these options clinically. Sincea number of toxins are associated with CA-MRSA, research is needed to address theappropriate selection of antimicrobial agents withregard to the threat posed by toxin release.

Nonantimicrobial Options

For noncomplicated S. aureus skin and softtissue abscesses, incision and drainage therapy,without the use of antibiotics, is generally adequate.One retrospective chart review addressed 69children with skin and soft tissue abscessescaused by culture-proved CA-MRSA.27 Treatmentconsisted of drainage and wound packing. Allchildren received antibiotics; however, only 7%of patients were prescribed an antibiotic to whichtheir CA-MRSA isolate was susceptible (treat-ment began before culture results were known).The investigators addressed the status of eachinfection at 2–6 months after presentation. Theyconcluded that incision and drainage, withoutadjunctive antibiotic therapy, were effective forCA-MRSA skin and soft tissue abscesses with adiameter of less than 5 cm in immunocompetentchildren.

Conclusion

Evidence suggests that CA-MRSA—the neweststaphylococcal threat—will be increasinglyprevalent in the near future. Moreover, althoughmost strains are now susceptible to many non–b-lactam antibiotics, this may change due toexposure to multiple antimicrobials in varioussettings. Epidemiologic studies are needed tospecify the patient populations likely to harborthis pathogen. Alternative treatment options tob-lactams must be assessed under investigationaltrial conditions before specific treatmentguidelines can be recommended.

References1. Chambers HF. The changing epidemiology of Staphylococcus

aureus? Emerg Infect Dis 2001;7:178–82.2. Lowy FD. Staphylococcus aureus infections. N Engl J Med

1998;339:520–32.3. Frank AL, Marcinak JF, Mangat PD, Schreckenberger PC.

Increase in community-acquired methicillin-resistantStaphylococcus aureus in children. Clin Infect Dis 1999;29:935–6.

4. Lodise TP Jr, McKinnon PS, Rybak M. Prediction model toidentify patients with Staphylococcus aureus bacteremia at risk

for methicillin resistance. Infect Control Hosp Epidemiol2003;24:655–61.

5. Karchmer AW. Nosocomial bloodstream infections: organisms,risk factors, and implications. Clin Infect Dis 2000;31(suppl4):S139–43.

6. Graffunder EM, Venezia RA. Risk factors associated withnosocomial methicillin-resistant Staphylococcus aureus (MRSA)infection including previous use of antimicrobials. J AntimicrobChemother 2002;49:999–1005.

7. Levine DP, Cushing RD, Jui J, Brown WJ. Community-acquired methicillin-resistant Staphylococcus aureusendocarditis in the Detroit Medical Center. Ann Intern Med1982;97:330–8.

8. Soriano A, Martinez JA, Mensa J, et al. Pathogenic significanceof methicillin resistance for patients with Staphylococcus aureusbacteremia. Clin Infect Dis 2000;30:368–73.

9. Lodise TP, McKinnon PS, Swiderski L, Rybak MJ. Outcomesanalysis of delayed antibiotic treatment for hospital-acquiredStaphylococcus aureus bacteremia. Clin Infect Dis 2003;36:1418–23.

10. Howden BP, Ward PB, Charles PG, et al. Treatment outcomesfor serious infections caused by methicillin-resistantStaphylococcus aureus with reduced vancomycin susceptibility.Clin Infect Dis 2004;38:521–8.

11. Rybak MJ, Akins RL. Emergence of methicillin-resistantStaphylococcus aureus with intermediate glycopeptideresistance: clinical significance and treatment options. Drugs2001;61:1–7.

12. Centers for Disease Control and Prevention. Methicillin-resistant Staphylococcus aureus skin or soft tissue infections in astate prison—Mississippi, 2000. JAMA 2002;287:181–2.

13. Abudu L, Blair I, Fraise A, Cheng KK. Methicillin-resistantStaphylococcus aureus (MRSA): a community-based prevalencesurvey. Epidemiol Infect 2001;126:351–6.

14. Adhikari RP, Cook GM, Lamont I, Lang S, Heffernan H, SmithJM. Phenotypic and molecular characterization of communityoccurring, Western Samoan phage pattern methicillin-resistantStaphylococcus aureus. J Antimicrob Chemother 2002;50:825–31.

15. Moreno F, Crisp C, Jorgensen JH, Patterson JE. Methicillin-resistant Staphylococcus aureus as a community organism. ClinInfect Dis 1995;21:1308–12.

16. Centers for Disease Control and Prevention. Public healthdispatch: outbreaks of community-associated methicillin-resistant Staphylococcus aureus skin infections—Los AngelesCounty, California, 2002–2003. JAMA 2003;289:1377.

17. Centers for Disease Control and Prevention. Methicillin-resistant Staphylococcus aureus infections among competitivesports participants—Colorado, Indiana, Pennsylvania, and LosAngeles County, 2000–2003. MMWR Morb Mortal Wkly Rep2003;52:793–5.

18. Bansal S, Kashyap S, Pal LS, Goel A . Clinical andbacteriological profile of community acquired pneumonia inShimla, Himachal Pradesh. Indian J Chest Dis Allied Sci2004;46:17–22.

19. Cookson BD. Methicillin-resistant Staphylococcus aureus in thecommunity: new battlefronts, or are the battles lost? InfectControl Hosp Epidemiol 2000;21:398–403.

20. Groom AV, Wolsey DH, Naimi TS, et al. Community-acquiredmethicillin-resistant Staphylococcus aureus in a rural AmericanIndian community. JAMA 2001;286:1201–5.

21. Shopsin B, Mathema B, Martinez J, et al. Prevalence ofmethicillin-resistant and methicillin-susceptible Staphylococcusaureus in the community. J Infect Dis 2000;182:359–62.

22. Steinberg JP, Clark CC, Hackman BO. Nosocomial andcommunity-acquired Staphylococcus aureus bacteremias from1980 to 1993: impact of intravascular devices and methicillinresistance. Clin Infect Dis 1996;23:255–9.

23. Naimi TS, LeDell KH, Boxrud DJ, et al. Epidemiology andclonality of community-acquired methicillin-resistantStaphylococcus aureus in Minnesota, 1996–1998. Clin Infect Dis2001;33:990–6.

24. Salmenlinna S, Lyytikainen O, Vuopio-Varkila J. Community-acquired methicillin-resistant Staphylococcus aureus, Finland.

83

PHARMACOTHERAPY Volume 25, Number 1, 2005

Emerg Infect Dis 2002;8:602–7.25. Bukharie HA, Abdelhadi MS, Saeed IA, Rubaish AM, Larbi

EB. Emergence of methicillin-resistant Staphylococcus aureus asa community pathogen. Diagn Microbiol Infect Dis2001;40:1–4.

26. Morin CA, Hadler JL. Population-based incidence andcharacteristics of community-onset Staphylococcus aureusinfections with bacteremia in 4 metropolitan Connecticut areas,1998. J Infect Dis 2001;184:1029–34.

27. Lee MC, Rios AM, Aten MF, et al. Management and outcomeof children with skin and soft tissue abscesses caused bycommunity-acquired methicillin-resistant Staphylococcusaureus. Pediatr Infect Dis J 2004;23:123–7.

28. Cohen PR, Kurzrock R. Community-acquired methicillin-resistant Staphylococcus aureus skin infection: an emergingclinical problem. J Am Acad Dermatol 2004;50:277–80.

29. Buckingham SC, McDougal LK, Cathey LD, et al. Emergenceof community-associated methicillin-resistant Staphylococcusaureus at a Memphis, Tennessee children’s hospital. PediatrInfect Dis J 2004;23:619–24.

30. Centers for Disease Control and Prevention. Community-associated MRSA. Information from the U.S. Centers forDisease Control and Prevention, 2003. Available fromhttp://www.cdc.gov/ncidod/hip/Aresist/mrsa_comm_faq.htm.

31. Naimi TS, LeDell KH, Como-Sabetti K, et al. Comparison ofcommunity- and health care-associated methicillin-resistantStaphylococcus aureus infection. JAMA 2003;290:2976–84.

32. Salgado CD, Farr BM, Calfee DP. Community-acquiredmethicillin-resistant Staphylococcus aureus: a meta-analysis ofprevalence and risk factors. Clin Infect Dis 2003;36:131–9.

33. Dufour P, Gillet Y, Bes M, et al. Community-acquiredmethicillin-resistant Staphylococcus aureus infections in France:emergence of a single clone that produces Panton-Valentineleukocidin. Clin Infect Dis 2002;35:819–24.

34. Herold BC, Immergluck LC, Maranan MC, et al. Community-acquired methicillin-resistant Staphylococcus aureus in childrenwith no identified predisposing risk. JAMA 1998;279:593–8.

35. Fey PD, Said-Salim B, Rupp ME, et al. Comparative molecularanalysis of community- or hospital-acquired methicillin-resistant Staphylococcus aureus. Antimicrob Agents Chemother2003;47:196–203.

36. Saravolatz LD, Markowitz N, Arking L, Pohlod D, Fisher E.Methicillin-resistant Staphylococcus aureus. Epidemiologicobservations during a community-acquired outbreak. AnnIntern Med 1982;96:11–16.

37. Mongkolrattanothai K, Boyle S, Kahana MD, Daum RS. SevereStaphylococcus aureus infections caused by clonally relatedcommunity-acquired methicillin-susceptible and methicillin-resistant isolates. Clin Infect Dis 2003;37:1050–8.

38. Okuma K, Iwakawa K, Turnidge JD, et al. Dissemination ofnew methicillin-resistant Staphylococcus aureus clones in thecommunity. J Clin Microbiol 2002;40:4289–94.

39. Enright MC, Robinson DA, Randle G, Feil EJ, Grundmann H,Spratt BG. The evolutionary history of methicillin-resistantStaphylococcus aureus (MRSA). Proc Natl Acad Sci USA2002;99:7687–92.

40. Ito T, Ma XX, Takeuchi F, Okuma K, Yuzawa H, Hiramatsu K.Novel type V staphylococcal cassette chromosome mec drivenby a novel cassette chromosome recombinase, ccrC. AntimicrobAgents Chemother 2004;48:2637–51.

41. Ma XX, Ito T, Tiensasitorn C, et al. Novel type of staphylo-coccal cassette chromosome mec identified in community-acquired methicillin-resistant Staphylococcus aureus strains.Antimicrob Agents Chemother 2002;46:1147–52.

42. Daum RS, Ito T, Hiramatsu K, et al. A novel methicillin-resistance cassette in community-acquired methicillin-resistantStaphylococcus aureus isolates of diverse genetic backgrounds. JInfect Dis 2002;186:1344–7.

43. Boubaker K, Diebold P, Blanc DS, et al. Panton-Valentineleukocidin and staphyloccoccal skin infections inschoolchildren. Emerg Infect Dis 2004;10:121–4.

44. Baba T, Takeuchi F, Kuroda M, et al. Genome and virulencedeterminants of high virulence community-acquired MRSA.

Lancet 2002;359:1819–27.45. Ren K, Bannan JD, Pancholi V, et al. Characterization and

biological properties of a new staphylococcal exotoxin. J ExpMed 1994;180:1675–83.

46. Baggett HC, Hennessy TW, Rudolph K, et al. Community-onset methicillin-resistant Staphylococcus aureus associatedwith antibiotic use and the cytotoxin Panton-Valentineleukocidin during a furunculosis outbreak in rural Alaska. JInfect Dis 2004;189:1565–73.

47. Lina G, Piemont Y, Godail-Gamot F, et al. Involvement ofPanton-Valentine leukocidin-producing Staphylococcus aureusin primary skin infections and pneumonia. Clin Infect Dis1999;29:1128–32.

48. Vandenesch F, Naimi T, Enright MC, et al. Community-acquired methicillin-resistant Staphylococcus aureus carryingPanton-Valentine leukocidin genes: worldwide emergence.Emerg Infect Dis 2003;9:978–84.

49. Diep BA, Sensabaugh GF, Somboona NS, Carleton HA,Perdreau-Remington F. Widespread skin and soft-tissueinfections due to two methicillin-resistant Staphylococcusaureus strains harboring the genes for Panton-Valentineleucocidin. J Clin Microbiol 2004;42:2080–4.

50. Saravolatz LD, Markowitz N, Arking L, Pohlod D, Fisher E.Methicillin-resistant Staphylococcus aureus: epidemiologicobservations during a community-acquired outbreak. AnnIntern Med 1982;96:11–16.

51. Centers for Disease Control and Prevention. Community-acquired methicillin-resistant Staphylococcus aureusinfections—Michigan. MMWR Morb Mortal Wkly Rep1981;30:185–7.

52. Kayaba H, Kodama K, Tamura H, Fujiwara Y. The spread ofmethicillin-resistant Staphylococcus aureus in a ruralcommunity: will it become a common microorganismcolonizing among the general population? Surg Today1997;27:217–19.

53. Witte W, Cuny C, Strommenger B, Braulke C, Heuck D.Emergence of a new community acquired MRSA strain inGermany. Euro Surveill 2004;9:1–2.

54. Tambyah PA, Habib AG, Ng TM, Goh H, Kumarasinghe G.Community-acquired methicillin-resistant Staphylococcusaureus infection in Singapore is usually “healthcare associated.”Infect Control Hosp Epidemiol 2003;24:436–8.

55. Baggett HC, Hennessy TW, Leman R, et al. An outbreak ofcommunity-onset methicillin-resistant Staphylococcus aureusskin infections in southwestern Alaska. Infect Control HospEpidemiol 2003;24:397–402.

56. Centers for Disease Control and Prevention. Outbreaks ofcommunity-associated methicillin-resistant Staphylococcusaureus skin infections—Los Angeles County, California,2002–2003. MMWR Morb Mortal Wkly Rep 2003;52:88–9, 99.

57. Zanelli G, Sansoni A, Zanchi A, et al. Staphylococcus aureusnasal carriage in the community: a survey from central Italy.Epidemiol Infect 2002;129:417–20.

58. Sola C, Gribaudo G, Vindel A, Patrito L, Bocco JL .Identification of a novel methicillin-resistant Staphylococcusaureus epidemic clone in Cordoba, Argentina, involved innosocomial infections. J Clin Microbiol 2002;40:1427–35.

59. Saxena S, Singh K, Talwar V. Methicillin-resistant Staphylococcusaureus prevalence in community in the east Delhi area. Jpn JInfect Dis 2003;56:54–6.

60. Wagenvoort JH, Kepers-Rietrae M. Methicillin resistantStaphylococcus aureus (MRSA) as a community strain. EuroSurveill 1997;2:96–7.

61. Lin JC, Yeh KM, Peng MY, Chang FY. Community-acquiredmethicillin-resistant Staphylococcus aureus bacteremia inTaiwan: risk factors for acquisition, clinical features andoutcome. J Microbiol Immunol Infect 2004;37:24–8.

62. Liassine N, Auckenthaler R, Descombes MC, Bes M,Vandenesch F, Etienne J. Community-acquired methicillin-resistant Staphylococcus aureus isolated in Switzerland containsthe Panton-Valentine leukocidin or exfoliative toxin genes. JClin Microbiol 2004;42:825–8.

63. Fang YH, Hsueh PR, Hu JJ, et al. Community-acquired

84

COMMUNITY-ASSOCIATED MRSA Rybak and LaPlante

methicillin-resistant Staphylococcus aureus in children innorthern Taiwan. J Microbiol Immunol Infect 2004;37:29–34.

64. Dietrich DW, Auld DB, Mermel LA. Community-acquiredmethicillin-resistant Staphylococcus aureus in southern NewEngland children. Pediatrics 2004;113:e347–52.

65. Schlesinger Y, Yahalom S, Raveh D, et al. Methicillin-resistantStaphylococcus aureus nasal colonization in children inJerusalem: community vs chronic care institutions. Isr MedAssoc J 2003;5:847–51.

66. Hiramatsu K, Okuma K, Ma XX, Yamamoto M, Hori S, KapiM . New trends in Staphylococcus aureus infections:glycopeptide resistance in hospital and methicillin resistance inthe community. Curr Opin Infect Dis 2002;15:407–13.

67. Li JT, Li Y, Wang J. Surveillance on gram-positive bacteriaisolated from patients with hospital acquired infections orcommunity acquired infections [in Chinese]. Zhonghua Yi XueZa Zhi 2003;83:365–74.

68. Centers for Disease Control and Prevention. Methicillin-resistant Staphylococcus aureus skin or soft tissue infections in astate prison—Mississippi, 2000. MMWR Morb Mortal WklyRep 2001;50:919–22.

69. Baum SE, Morris JT, Dooley DP, Watson R. Methicillin-resistant Staphylococcus aureus in an adult military beneficiarypopulation lacking risk factors: susceptibility to orally availableagents. Mil Med 2003;168:126–30.

70. Iyer S, Jones DH. Community-acquired methicillin-resistantStaphylococcus aureus skin infection: a retrospective analysis ofclinical presentation and treatment of a local outbreak. J AmAcad Dermatol 2004;50:854–8.

71. Cha R, Grucz JR Jr, Rybak MJ. Daptomycin dose-effectrelationship against resistant gram-positive organisms.Antimicrob Agents Chemother 2003;47:1598–603.

72. Centers for Disease Control and Prevention. Public healthdispatch: outbreaks of community-associated methicillin-resistant Staphylococcus aureus skin infections—Los AngelesCounty, California, 2002–2003. Can Commun Dis Rep2003;29:110–12.

73. Centers for Disease Control and Prevention. Four pediatricdeaths from community-acquired methicillin-resistantStaphylococcus aureus—Minnesota and North Dakota,1997–1999. JAMA 1999;282:1123–5.

74. Willey BM, Archambault M, Kreeiswirth BN, et al. Methicillinresistant infection in horses and horse personnel (2000–2002).Presented at the 43rd interscience conference on antimicrobialagents and chemotherapy, Chicago. IL, September 14–17, 2003.

75. LaPlante KL, Ponomareva A, Rybak, MJ . Comparingphenotypic expressions of community and hospital associatedmethicillin resistant Staphylococcus aureus on the basis ofSCCmec type. Presented at the annual meeting of the AmericanCollege of Clinical Pharmacy, Dallas, TX, October 24–27, 2004.

76. Siberry GK, Tekle T, Carroll K, Dick J. Failure of clindamycintreatment of methicillin-resistant Staphylococcus aureusexpressing inducible clindamycin resistance in vitro. Clin InfectDis 2003;37:1257–60.

77. Frank AL, Marcinak JF, Mangat PD, et al. Clindamycintreatment of methicillin-resistant Staphylococcus aureusinfections in children. Pediatr Infect Dis J 2002;21:530–4.

78. Fiebelkorn KR, Crawford SA, McElmeel ML, Jorgensen JH.

Practical disk diffusion method for detection of inducibleclindamycin resistance in Staphylococcus aureus and coagulase-negative staphylococci. J Clin Microbiol 2003;41:4740–4.

79. National Committee for Clinical Laboratory Standards.Supplemental tables M100-S13. Wayne, PA: NationalCommittee for Clinical Laboratory Standards, 2003.

80. National Committee for Clinical Laboratory Standards.Approved standard M7-A6; methods for dilution antimicrobialsusceptibility tests for bacteria that grow aerobically. Wayne,PA: National Committee for Clinical Laboratory Standards,2003.

81. Markowitz N, Quinn EL, Saravolatz LD. Trimethoprim-sulfamethoxazole compared with vancomycin for the treatmentof Staphylococcus aureus infection. Ann Intern Med1992;117:390–8.

82. Strom BL, Schinnar R, Apter AJ, et al. Absence of cross-reactivity between sulfonamide antibiotics and sulfonamidenonantibiotics. N Engl J Med 2003;349:1628–35.

83. Nicolau DP, Freeman CD, Nightingale CH, Coe CJ,Quintiliani R. Minocycline versus vancomycin for treatment ofexperimental endocarditis caused by oxacillin-resistantStaphylococcus aureus. Antimicrob Agents Chemother1994;38:1515–18.

84. Drinka PJ, Gauerke C, Le D . Antimicrobial use andmethicillin-resistant Staphylococcus aureus in a large nursinghome. J Am Med Dir Assoc 2004;5:256–8.

85. Blumberg HM, Rimland D, Carroll DJ, Terry P, WachsmuthIK . Rapid development of ciprofloxacin resistance inmethicillin-susceptible and -resistant Staphylococcus aureus. JInfect Dis 1991;163:1279–85.

86. Tenover FC, Weigel LM, Appelbaum PC, et al. Vancomycin-resistant Staphylococcus aureus isolate from a patient inPennsylvania. Antimicrob Agents Chemother 2004;48:275–80.

87. Appelbaum PC, Bozdogan B. Vancomycin resistance inStaphylococcus aureus. Clin Lab Med 2004;24:381–402.

88. Pillai SK, Sakoulas G, Wennersten C, et al . Linezolidresistance in Staphylococcus aureus: characterization andstability of resistant phenotype. J Infect Dis 2002;186:1603–7.

89. Rubinstein E, Prokocimer P, Talbot GH. Safety and tolerabilityof quinupristin/dalfopristin: administration guidelines. JAntimicrob Chemother 1999;44(suppl A):37–46.

90. Leclercq R. Mechanisms of resistance to macrolides andlincosamides: nature of the resistance elements and theirclinical implications. Clin Infect Dis 2002;34:482–92.

91. Leclercq R, Courvalin P. Bacterial resistance to macrolide,lincosamide, and streptogramin antibiotics by targetmodification. Antimicrob Agents Chemother 1991;35:1267–72.

92. Barry AL, Fuchs PC, Brown SD . In vitro activities ofdaptomycin against 2789 clinical isolates from 11 NorthAmerican medical centers. Antimicrob Agents Chemother2001;45:1919–22.

93. Akins RL, Rybak MJ. Bactericidal activities of two daptomycinregimens against clinical strains of glycopeptide intermediate-resistant Staphylococcus aureus, vancomycin-resistantEnterococcus faecium, and methicillin-resistant Staphylococcusaureus isolates in an in vitro pharmacodynamic model withsimulated endocardial vegetations. Antimicrob AgentsChemother 2001;45:454–9.

85