Top Banner

of 13

Bacterial Resistance Origins, Epidemiology

Apr 07, 2018

Download

Documents

leila.hilout
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
  • 8/3/2019 Bacterial Resistance Origins, Epidemiology

    1/13

    Bacterial Resistance CID 2003:36 (Suppl 1) S11

    S U P P L E M E N T A R T I C L E

    Bacterial Resistance: Origins, Epidemiology,and Impact

    David M. Livermore

    Antibiotic Resistance Monitoring and Reference Laboratory, Central Public Health Laboratory, London, United Kingdom

    The basic mechanisms of antibacterial resistance are well known, but critical newaspectscontinue to be discovered.

    Recently discovered factors with major implications for the emergence, dissemination, and maintenance of

    resistance include multidrug efflux, hypermutability, integrons, and plasmid addiction. Some resistances are

    widespread and others local, with prevalence rates often worst in newly prosperous countries and in those

    specialist units where antibacterial use is heaviest. Multidrug-resistant epidemic strains are critical to the total

    accumulation of resistance (e.g., among Streptococcus pneumoniae, methicillin-resistant Staphylococcus aureus,

    Klebsiella pneumoniae), but it remains unclear why some bacterial lineages achieve epidemic spread whereas

    others that are equally resistant do not. The correlation between in vitro resistance and treatment failure is

    imperfect, but resistance undoubtedly increases mortality, morbidity, and costs in many settings. Recent concern

    has led to a plethora of governmental and agency reports advocating less antibacterial use, better antibacterial

    use, better infection control, and the development of new antibacterials. The evidence that better prescribing

    can reduce resistance rates is mixed, and although changes to hospital regimens may reduce one resistance

    problem, other opportunistic bacteria may fill the vacant niche. Overall, the best that can reasonably be anticipated

    is an improved balance between the accumulation of resistance and new antibacterial development.

    Most bacteria have multiple routes to resistance to any

    drug and, once resistant, can rapidly give rise to vast

    numbers of resistant progeny. Natural selection favors

    mechanisms that confer resistance with the least fitness

    cost and those strains that are least burdened by their

    resistance. Selection may also favor determinants that

    prevent their own counterselection and resistant strains

    with enhanced survival ability or virulence. To this ge-

    netic and biochemical potential must be added the wide

    variety of bacteria that cause opportunistic infections

    in vulnerable human patients and the factthat the num-

    bers of vulnerable patients grow steadily with advances

    in other fields of medicine. In short, the emergence ofresistance is profoundly unsurprising; what is remark-

    Reprints or correspondence: Dr. David M. Livermore, Antibiotic Resistance

    Monitoring and Reference Laboratory, Central Public Health Laboratory, 61

    Colindale Ave., London NW9 5HT, United Kingdom ([email protected]).

    Clinical Infectious Diseases 2003;36(Suppl 1):S1123

    2003 by the Infectious Diseases Society of America. All rights reserved.

    1058-4838/2003/3602S1-0004$15.00

    able is how long it has taken for the problem to become

    a source of public, as well as scientific, concern.

    Resistance can result from modification of an anti-

    bacterials target or from functional bypassing of that

    target, or it can be contingent on impermeability, efflux,

    or enzymatic inactivation. All members of a species may

    be resistant. Alternatively, resistance may arise in hitherto

    susceptible organisms via mutation or DNA transfer.The

    aim of this article is notto catalog individualmechanisms

    or their prevalencethat has been done elsewherebut

    to emphasize the continuing dynamism of resistance, its

    impact on therapy, and the difficultybut also the po-

    tentialfor combating the problem.

    SELECTION OF SPECIES WITH INHERENT

    RESISTANCE

    Antibacterial use disrupts the microbial ecology of the

    patient, unit, or population. Entire species may be se-

    lected. The increasing role of enterococci as opportunist

    pathogens in the past 20 years partly reflects increasing

  • 8/3/2019 Bacterial Resistance Origins, Epidemiology

    2/13

    S12 CID 2003:36 (Suppl 1) Livermore

    use of fluoroquinolones and cephalosporins, to which these

    organisms are inherently resistant [1, 2]. The increasing role

    of coagulase-negative staphylococci (especially) and a-hemo-

    lytic streptococci in hematology patients also may reflect an-

    timicrobial use: a-hemolytic streptococci are resistant to fluor-

    oquinolones, which are often used as prophylaxis in these

    patients, and coagulase-negative staphylococci frequently have

    acquired multidrug resistance [3]. A greater factor behind therise of coagulase-negative staphylococci is, however, the in-

    creased use of indwelling lines, which provide entry portals for

    the skin microflora. Among gram-negative pathogens, Acine-

    tobacter baumannii and Stenotrophomonas maltophilia are in-

    creasingly prevalent in many intensive care units (ICUs), with

    A. baumanniinotoriously associated with ventilator-associated

    pneumonias [4]. A. baumannii commonly has acquired resis-

    tance to all antibacterials except carbapenems, minocycline,

    sulbactam, and colistin, whereas S. maltophiliais often resistant

    to all antibacterials except co-trimoxazole (and perhaps ticar-

    cillin-clavulanate). Some publications associate an increasing

    incidence ofS. maltophiliainfections with carbapenem use [5],but others find a less specific relationship to the use of multiple

    antibacterials, including those (e.g., erythromycin) with pre-

    dominantly antigram-positive spectra [6, 7].

    RESISTANCE VIA MUTATION

    As DNA is replicated, uncorrected base substitutions occur ran-

    domly, at a frequency of109 to 1010 per gene [8]. In ad-

    dition, copying errors may lead to the partial or complete de-

    letion of individual genes [8]. As a result, the targets of

    antibacterials may be altered, drug-inactivation or efflux sys-tems may be up- or downregulated, and uptake pathways (po-

    rins and active transporters) may be lost or activated. Resistance

    genes or their repressors also can be activated or inactivated

    by the migration of insertion sequences. Approximately 3% of

    Bacteroides fragilis isolates have the carbapenemase gene ccrA

    (cfiA), but its enzyme product is expressed only if an insertion

    sequence has migrated upstream of this structural gene [9].

    Classical experiments showed that antibacterials cause the

    selection of preexisting variants, not the emergence of new

    mutants. This observation entirely agrees with the precepts of

    Darwinian evolution, but a twist is given by the observations

    that bacteria can become hypermutable through inactivation

    of the proofreading and DNA mismatchrepair systems that

    normally correct DNA copying errors [8]. Hypermutators have

    up to 200-fold higher mutation rates than normal cells and so

    are more likely to become resistant to a first antibacterial by

    mutation. Once selected by this first drug, they are then

    primed to develop resistance to further agents. To this extent,

    antibacterials may cause the emergence of variants with an

    increased propensity to develop further resistance. Hypermut-

    ability also may arise transiently through induction of the SOS

    system, a stress response that involves the expression and func-

    tion of alternative DNA polymerases with reduced copying fi-

    delity [8, 10]. The SOS system is induced, inter alia, by star-

    vation, and it is also notable that the spectrum of mutations

    seen in non- or slow-growing cells differs from those among

    logarithmic-phase organisms. Also relevant is the fact that

    DNA-damaging forms of reactive oxygen accumulate in non-growing cells [8, 11], perhaps acting as mutagens.

    The selection or transient induction of hypermutability may

    explain why variants with multiple mutations have emerged

    more rapidly than was predicted from laboratory studies. Non-

    clonal strains of fluoroquinolone-resistant Escherichia colihave

    emerged worldwide and have become highly prevalent, for ex-

    ample, in India [12], Spain [13], and China [14]. This is despite

    the fact that substantive fluoroquinolone resistance in Enter-

    obacteriaceae requires mutations to the genes for subunits of

    topoisomerases II and IV (gyrA and parC, respectively), to-

    gether with further mutations that upregulate efflux and reduce

    permeability, or both [1417]. Perhaps the emergence of these

    multiple mutants is favored by the quinolone-mediated in-

    duction of the SOS response, with its contingent hypermuta-

    bility [18]. This is speculation, but there is hard evidence of

    the role of hypermutators in the cystic fibrosis lung, where

    Oliver et al. [19] found that 36% of 30 chronically colonized

    patients carried hypermutable Pseudomonas aeruginosa. Hy-

    permutators were not found in 75 control (noncystic) patients

    with acute P. aeruginosa infections.

    Most mutations affect only a single antibacterial class, but

    those affecting impermeability or efflux may have a pleiotropic

    effect. The potential for porin loss to affect multiple drug classesby restricting nonspecific permeability is self-evident, but the

    role of broad-spectrum efflux has come as a surprise. Its func-

    tion is best understood in P. aeruginosa, where mutation at

    mexR upregulates the mexA-mexB-oprMoperon and raises the

    MICs ofb-lactams (except imipenem), fluoroquinolones, tet-

    racyclines, chloramphenicol, macrolides, various disinfectants,

    detergents, and organic solvents [20]. MexAB-OprM also plays

    a roleperhaps coincidentallyin excretion of the quorum-

    sensing mediator, homoserine lactone. Even in typical P. aeru-

    ginosa isolates, MexAB-OprM is expressed to some degree and

    accounts for much of the resistance previously ascribed to im-

    permeability [21]. Isolates with further upregulation have ad-ditional resistance to substrate drugs (table 1). P. aeruginosa

    strains have 3 further well-characterized efflux systems, 2 of

    which (MexCD-Opr J and MexEF-Opr N) are normally re-

    pressed but may be activated by mutation (table 1). The ge-

    nomic sequence for P. aeruginosa suggests the presence of at

    least 5 further efflux systems, which await even preliminary

    characterization [20]. Multidrug efflux systems are also being

    reported in many other gram-negative bacteria, including En-

  • 8/3/2019 Bacterial Resistance Origins, Epidemiology

    3/13

    Bacterial Resistance CID 2003:36 (Suppl 1) S13

    Table 1. Drug efflux systems of Pseudomonas aeruginosa.

    System

    Regulatory

    gene

    Mutation causing

    upregulation Substrates

    MexAB-OprM mexR nalB (affects mexR) and nalC

    (lies outside mexR)

    b-Lac (not imipenem), Cm, Em, Fq, Nv, Tm, Su, Tc, ethidium bromide,

    acriflavine, SDS, aromatic hydrocarbons, irgasan, triclosan, homoserine

    lactone

    MexCD-OprJ nfxB nfxB b-Lac (not imipenem), Cm, Em, Fq, Nv, Tm, Tc, ethidium bromide, acriflavine,

    SDS, aromatic hydrocarbons, triclosanMexEF-OprN mexT nfxC Cm, Fq, Tm, aromatic hydrocarbons, triclosan

    MexXY-OprM mexZ Agl, b-lac (not carbenicillin, ceftazidime or imipenem), Cm, Em, Fq, Nv, Tc

    NOTE. Agl, aminoglycosides; b-lac, b-lactam; Cm, chloramphenicol; Em, erythromycin; Fq, fluoroquinolones; Nv, novobiocin; SDS, sodium dodecyl sulfate;

    Su, sulfonamides; Tc, tetracycline; Tm, trimethoprim. Data from [20, 21].

    terobacteriaceae as well as other nonfermenters [20, 22]. At a

    functional level, the role of broad-spectrum efflux pumps may

    be to remove amphipathic substances (i.e., those with hydro-

    philic and hydrophobic parts) from membranes, preventing

    disorganization. Such a cleaning role would account for their

    broad activity and wide distribution of these pumps, but theirsubstrate recognition sites await elucidation

    Efflux systems can be coregulated with porin expression. In

    P. aeruginosa, the nfxC mutation at mexTupregulates MexEF-

    OprNmediated efflux (table 1), and also reduces expression

    of OprD (D2 porin in older literature), thereby reducing

    permeability to carbapenems [23]. In E. coli (and, probably,

    other Enterobacteriaceae), mutation at marsimultaneously af-

    fects the expression of160 genes [24] reducing, inter alia, ex-

    pression of porin OmpF and upregulating AcrAB, which is

    implicated in the efflux of b-lactams, fluoroquinolones, chlo-

    ramphenicol, tetracycline, dyes, and detergents [25].

    If mutational resistance emerges at high frequency withoutdeleterious side effects, it can swiftly undermine an antibac-

    terials utility by allowing multifocal emergence of resistance

    wherever the drug is used. Several antimicrobial groups intro-

    duced in the 1980s and 1990s are prone to mutational resis-

    tance, at least with some species. Early optimism about fluoro-

    quinolones against staphylococci was dispelled by the discovery

    that resistance emerged by upregulation of NorA-mediated ef-

    flux [26]. Oxyimino-aminothiazolyl cephalosporins at first ap-

    peared almost universally active against Enterobacter, Citrobac-

    ter, and Serratiaspp., but it soon became apparent that activity

    depended on a failure to induce chromosomal AmpC b-lac-

    tamases [27]. Activity was lost against derepressed mutants,which hyperproduce AmpC b-lactamases independently of in-

    duction. Such mutants arise at frequencies of 105 to 107 and

    are selected in 20% of patients treated with cephalosporins

    for Enterobacterbacteremia, with this selection often leading to

    clinical failures [28]. Although imipenem evades this mecha-

    nism, imipenem resistance emerges readily in P. aeruginosa by

    loss of the OprD porin, which provides carbapenem-specific

    pores through the outer membrane [29, 30]. Most recently,

    there has been concern about linezolid resistance emerging in

    enterococci or, more rarely, in Staphylococcus aureus via mod-

    ification of the domain V of 23S rRNA, which contains the

    drugs binding site. Mutational resistance to linezolid is difficult

    to obtain in vitro, apparently because the bacteria have multiple

    copies of the encoding gene, with modification of more thanone copy required for resistance. Nevertheless, linezolid resis-

    tance can be selected in vivo, particularly in difficult-to-reach

    infections, those requiring prolonged therapy and if the drug

    is underdosed [31, 32]. It is not clear whether DNA recom-

    bination events follow the mutation of one gene copy or

    whether multiple gene copies undergo sequential mutations,

    perhaps by induction of a transient hypermutability (above).

    ACQUISITION OF RESISTANCE BY DNA

    TRANSFER

    DNA transfer among bacteria is critical to the disseminationof resistance and has recently been reviewed [33]. Transfer of

    DNA is most often via plasmids. These existed long before

    humans used antibacterials but did not then carry resistance

    determinants, or rarely did so [34]. Since the advent of the

    antibacterial era, plasmids have, however, proved to be the ideal

    vehicles for recruitment and dissemination of resistance genes.

    Within plasmids, resistance genes are often carried by trans-

    posons, which can shuttle determinants between more and less

    promiscuous plasmids, or into and out of the chromosome.

    Some transposons are directly transmissible between bacteria,

    particularly (but not exclusively) among gram-positive species.

    Resistance genes also may be transferred by lysogenic bacte-

    riophage. This latter mechanism seems likely with the mecA

    determinant staphylococci, which has never been located on

    mobile DNA but which has spread among a few S. aureus

    lineages, and among different coagulase-negative species [35].

    Integrons are natural recombination systems that facilitate

    the acquisition and expression of resistance determinants be-

    hind a single promoter. They are widely distributed among

    gram-negative bacteria, often occurring within plasmids and

  • 8/3/2019 Bacterial Resistance Origins, Epidemiology

    4/13

    S14 CID 2003:36 (Suppl 1) Livermore

    Table 2. Sources of b-lactamase resistance genes now found on transferableDNA.

    Gene(s) or product(s) Source

    SHV b-lactamases Klebsiella pneumoniae chromosome

    CTX-M2,4,5,6,7 and Toho-1 b-lactamases Kluyvera spp. chromosome

    CMY-2, 3, 4, 5, 6, 7, LAT-1, -2, -3, -4, BIL-1

    AmpC b-lactamases Citrobacter freundii chromosome

    ACC-1 AmpC b-lactamases Hafnia alvei chromosome

    DHA-1 and -2 AmpC b-lactamases Morganella morganii chromosome

    TEM, OXA,a

    PSE, staphylococcal

    penicillinase Unknown

    a

    Although source organisms for plasmid-mediated OXA enzymes have not been identified,

    some Aeromonasspp. have chromosomal enzymes belonging to this family.

    transposons, and are particularly important in the dissemina-

    tion of resistance genes to sulfonamides (sul1), and strepto-

    mycin (aadA3) [36]. Other genes often located in integrons

    include those for various OXA, PSE, VIM, and IMP b-lacta-

    mases and for many aminoglycoside-modifying enzymes [36,37]. In principle, integrons have a fearsome capacity for the

    recruitment, spread, and expression of resistance genes, and

    surveys show that they are widespread among gram-negative

    bacteria in countries as far apart as The Netherlands and Taiwan

    [38, 39]. Nevertheless, it is striking that the most successful b-

    lactamase genes (i.e., blaTEM

    derivatives) are carried directly by

    transposons, not within integrons, whereas the integron-asso-

    ciated OXA and PSE b-lactamases are considerably rarer [27].

    Similarly with sulfonamide resistance: sul2, which is not inte-

    gron-associated, is increasing in prevalence among E. coli in

    the UK, whereas sul1, which is integron-determined, is stable

    in prevalence, or declining [40]. It is notable also that thecomposition of integrons is more stable over time than might

    be expected [38]. In short, integrons are important, but their

    importance should not be overplayed relative to that of other

    vehicles of resistance.

    The dissemination of plasmids, transposons, and integrons

    among bacteria and species give rise to so-called gene epidem-

    ics. Plasmids encoding the TEM plasmid-mediated b-lacta-

    mases were first recognized in 1965 but have since spread

    varying with the country, unit, and speciesto 30%60% of

    clinical Enterobacteriaceae, to a few P. aeruginosa, and to any-

    where between 1%50% of Haemophilus influenzae and Neis-

    seria gonorrhoeae isolates [27]. Other determinants that have

    spread extremely widely include erm, sul1, sul2 strA, strB, aad3,

    tetA, and tetM. tetM has spread in both gram-positive and

    gram-negative organisms, but more generally, the genes of

    gram-positive and gram-negative species are distinct [41]. The

    factors that determine whether a mobile gene will spread widely

    are poorly understood. TEM-2 b-lactamase differs from TEM-

    1 by a single amino acid substitution, confers similar resistance,

    is coded by similarly promiscuous plasmids and transposons,

    and has been known for almost as long. Nevertheless, for no

    obvious reason, it is 10-fold less prevalent that TEM-1 b-lac-

    tamase in every country and species surveyed [27].

    Many of the resistance determinants now found on plasmids

    are believed to have originated in the chromosomes of otherspecies, although only a few of their source organisms have

    been identified definitively. The plasmid-mediated SHV b-lac-

    tamases are derived from the chromosomal b-lactamases of

    Klebsiella pneumoniae; the plasmid-borne AmpC enzymes

    emerging in Klebsiellaspp. and E. coliare chromosomal escapes

    from Citrobacter freundii, Hafnia alvei, Morganella morganii,

    and Enterobacter cloacae [42, 43], and several CTX-M cefotax-

    imases are chromosomal escapes from Kluyveraspp. [44] (table

    2). Some resistance determinants to nonb-lactam drugs seem-

    ingly originated in antibiotic-producing streptomycetes, which

    must protect themselves against their own products. Examples

    include several aminoglycoside-modifying enzymes and the ermdeterminants, which have products that methylate 23S rRNA

    so as to block binding of macrolides, lincosamides, and group

    B streptogramins [45]. Also, the genes that encode the d-ala-

    d-ala ligases are critical to glycopeptide resistance in enterococci

    (and now also S. aureus [46, 47]). Many plasmids and tran-

    sposons carry multiple resistance genes conferring resistance to

    different antibacterials, and selection for any one determinant

    may conserve the entire plasmid and its resistances. The fre-

    quent consequence is multidrug resistance, as illustratedin table

    3, which shows that klebsiellae with extended-spectrum b-lac-

    tamases (ESBL) were more often resistant to aminoglycosides

    and fluoroquinolones than those without ESBLs [48, 49]. ESBLs

    and aminoglycoside-modifying enzymes may be encoded by

    single plasmids, but the fluoroquinolone resistance, also seen

    in an excess of the ESBL producers, is chromosomal and

    independent.

    The problems of multiresistance are increasing and becoming

    more complex. When plasmid electrophoresis was developed

    in the 1970s, it was uncommon to see isolates with 123 plas-

    mids, and plasmid profiles were used to define strain epide-

  • 8/3/2019 Bacterial Resistance Origins, Epidemiology

    5/13

    Bacterial Resistance CID 2003:36 (Suppl 1) S15

    Table 3. Multidrug resistance among Klebsiellaspp. collected in 2 European surveysof isolates from intensive care units.

    Resistance

    1994 sur vey 19971998 sur vey

    ESBL

    positive

    ( )np 220

    ESBL

    negative

    ( )np 746

    ESBL

    positive

    ( )np 110

    ESBL

    negative

    ( )np 323

    Klebsiellae with the indicated ESBL

    phenotype 23 77 25 75

    Gentamicin resistance 76 8 72 10

    Amikacin resistance 52 3 61 4

    Ciprofloxacin resistant 31a

    2 31 7

    NOTE. Data are percentage of isolates. ESBL, extended-spectrum b-lactamase. Data from [42, 43].a

    Declined to 10% if members of a single widespread clone were discounted.

    miology. Nowadays, it is common to encounter Enterobacter-

    iaceae with 56 plasmids; moreover, plasmid profiles often vary

    greatly within the strains defined by PFGE [50]. These ob-

    servations (which complicate epidemiological analysis) reflect

    the gain and loss of plasmids by different representatives of the

    same strain and the gain and loss of genes among different

    representation of the same plasmid. Even when genes are not

    gained or lost, multiple copies of the same plasmid may carry

    different variants of a b-lactamase gene, reflecting mutation.

    Thus, for example, some members of a K. pneumoniae strain

    from Turkey had SHV-3 b-lactamase, whereas others had SHV-

    5 (table 4) [50]. More strikingly, 84 blaTEM

    and blaSHV

    copies

    were found among just 25 K. pneumoniae isolates collected at

    one hospital in Durban, South Africa [51].

    A few speciesNeisseria spp., Haemophilusspp., and a-he-

    molytic streptococcican absorb and incorporate DNA re-

    leased by dead cells of related organisms, allowing the gener-ation of mosaic genes [52]. This mechanism is absent from

    most other species, which restrict incoming DNA. Mosaic gene

    formation, acting in combination with mutation, is the basis

    of emerging penicillin resistance in pneumococci and of b-

    lactamaseindependent ampicillin or penicillin resistance in H.

    influenzae and Neisseria spp.; it is also implicated in sulfona-

    mide resistance in H. influenzae [53].

    EPIDEMIOLOGY OF RESISTANCE: LOCAL,

    NATIONAL, AND INTERNATIONAL

    At one level, the epidemiology of resistance is extremely local.

    Most outbreaks and clusters involve a few patients in a unit,

    and the prevalence of resistance is often highest in those units

    where the most vulnerable patients are congregated and where

    antibacterial use consequently is heaviest. Archibald et al. [54]

    found 2-fold higher rates of methicillin resistance among

    staphylococci, ceftazidime resistance among E. cloacae and P.

    aeruginosa, imipenem resistance among P. aeruginosa, and van-

    comycin resistance among enterococci in patients in ICUs than

    in patients in general wards or outpatients at the same hospitals.

    In virtually all European countries, the prevalence of methicillin-

    resistant S. aureus is higher in ICUs than in general wards [55].

    At another level, the epidemiology of resistance is national.

    In Europe, the common pattern is for resistance to increase in

    prevalence as one moves southward: it is lowest in Scandinavia

    and highest in the Mediterranean countries. In North America,

    resistance rates are mostly higher in the United States than in

    Canada. Some of the worst resistance rates are in the newly

    prosperous countries of East Asia and South America. A few

    examples: methicillin-resistant strains comprise 30%45% of

    all S. aureus from bacteremias in Spain, Portugal, Italy, France,

    and the United Kingdom and 10%15% in Germany and Aus-

    tria, but !1% in the Netherlands and Scandinavia (see, e.g.,

    European Animicrobial Resistance Surveillance System, http://

    www.earss.rivm.nl). In Korea, Japan, Taiwan, and Vietnam,

    70%80% of S. pneumoniae are resistant or intermediately re-sistant to penicillin, compared with 30%40% in France and

    Spain, 5%10% in the United Kingdom, and 1%2% in Scan-

    dinavia [5658]. As a final example, gentamicin resistance in

    E. coli remains considerably more frequent in most Southern

    European countries and the United States than in the United

    Kingdom, where it occurs in !3% of isolates [57].

    Finally, the epidemiology of resistance is partly international,

    with some transferable determinants prevalent worldwide. The

    epidemiology is also international to the extent that some re-

    sistant strains spread between countries and continents. Multi-

    drug-resistant pneumococci of serotype 6B were imported from

    Spain into Iceland, apparently by nasopharyngeal carriage inthe children of returning holidaymakers [59]. These pneu-

    mococci then became established in child care centers in Ice-

    land, causing an increase in the penicillin resistance rate from

    1% in 1988 to 17% in 1993. Other penicillin-resistant pneu-

    mococci of serotype 23F have spread from Spain to the Far

    East, the Americas, and South Africa [60]. On a smaller scale,

    many of the few E. coli and Klebsiella spp. with plasmid-me-

    diated AmpC b-lactamases in the United Kingdom are epi-

  • 8/3/2019 Bacterial Resistance Origins, Epidemiology

    6/13

    S16 CID 2003:36 (Suppl 1) Livermore

    Table 4. Diverse b-lactamases and resistances of multiple representatives of a serotypeK62 Klebsiella pneumoniae strain collected at an intensive care unit in Istanbul, Turkey.

    Isolate

    MIC, mg/L

    b-LactamaseGentamicin Aztreonam Ceftazidime Ceftriaxone

    Piperacillin-

    tazobactam

    1951 8 512 1024 256 4 SHV-5

    1953 16 512 1024 256 4 SHV-3 TEM

    1954 8 8 32 0.5 1024 SHV-3 TEM

    1959 16 512 512 512 1024 SHV-5 TEM

    1964 16 512 512 256 512 SHV-3 TEM

    NOTE. Data from [50]. All 5 representatives had the same DNA profile as investigated by PFGE.

    demiologically linked to the Indian subcontinent, where there

    is evidence of local frequency in Punjab [61, 62]. Last, PER-1

    ESBL was first recorded from a P. aeruginosa isolate collected in

    France [63] and shortly afterward was found in numerous P.

    aeruginosa, Salmonella, and Acinetobacter spp. isolates from sev-

    eral cities in Turkey [64]. An inquiry revealed that the original

    source patient in France was a Turk, visiting for treatment [65].

    EPIDEMIC RESISTANT STRAINS

    Successful epidemic strains are critical to the accumulation of

    many resistances. Common vectors in hospitals are contact with

    staff members, contact with nonsterile devices, or procedures.

    Spread in the community is favored by those factors that have

    aided epidemics throughout history: crowding and travel. Many

    strains, resistant or otherwise, spread locally, but a few achieve

    a much wider distribution. The international spread of peni-

    cillin-resistant pneumococcal lineages was mentioned above,but further examples abound. In England and Wales, the pro-

    portion ofS. aureusbacteremias caused by methicillin-resistant

    S. aureus (MRSA) was steady at 1%3% from 1989 to 1993

    but increased rapidly afterward, reaching 42% by 2000 [57].

    The beginning of this increase coincided with the emergence

    of 2 new epidemic (E) strains, EMRSA 15 and 16, and recent

    analysis shows that these now account for 193% of all S. aureus

    bacteremias in England and Wales [66]. In France, a serotype

    K25 K. pneumoniae strain with SHV-4 b-lactamase and cross-

    resistance to amikacin and ciprofloxacin has disseminated

    widely, having been reported repeatedly since 1988, first around

    Paris and subsequently in hospitals from the Atlantic coast to

    the Mediterranean, with reports also from Ghent in Belgium

    [50, 67, 68]. A survey of ESBLs among 966 klebsiellae in 1994

    included 35 centers, only 5 of them in France, yet this single

    strain accounted for 52 of 220 ESBL producers collected [50].

    Also in France and Belgium, an Enterobacter aerogenes strain

    with TEM-24 b-lactamase and multiresistance to aminogly-

    cosides, quinolones, and, occasionally (via porin loss), carba-

    penems has become widely established [69]. Other examples

    where resistance has a major clonal element include Burkholderia

    cepacia from cystic fibrosis patients [70] and Salmonella typhi-

    murium, where major recent problems (now perhaps declining)

    have been associated with the intercontinental spread of multi-

    drug-resistant lineages of definitive type DT104 [71, 72].

    In the case of vancomycin-resistant Enterococcus faecium, re-

    cent studies by amplified DNA restriction fragment-length

    polymorphism suggest that epidemic strains from hospitals in

    Europe, the United States, and Australia, although differing

    from place to place, are more closely related to each other than

    to sporadic and agricultural isolates [73].

    Other strains with similar resistances to these epidemic or-

    ganisms are recorded, often in the same hospitals or patient

    groups, but these fail to spread extensively or fail to spread at

    all. The reason for epidemic success remain obscure, but po-

    tential factorsnot mutually exclusiveinclude the following:

    (1) increased adherence to host cells or prosthetic materials,

    (2) greater tolerance of desiccation, (3) elevated resistance todisinfectants, (4) faster growth rates, and (5) better adaptation

    to the fitness cost of resistance. Hard evidence for the role of

    any of these factors is scanty for many particular strains, and

    studies are confounded by the fact that researchers, having

    found one fact that may contribute to epidemic success, then

    concentrate on that factor in isolation. In the case of the suc-

    cessful serotype K25 K. pneumoniae strain from France and

    Belgium, one report suggests a plasmid-mediated fimbrial an-

    tigen that aids adherence to the gut mucosa [74].

    Local variation, national variation, and role of epidemic

    strains should be borne in mind when undertaking and as-

    sessing prevalence surveys. Grandiloquent statements about re-sistance rates in Europe or the Western Pacific abound in

    the literature but often are based on surveys with a very few

    centers per country, and with the extent of resistance differing

    greatly among the countries included within a region. The var-

    iation in resistance prevalence between Stockholm and Madrid,

    it should be noted, is vastly greater than between Boston and

    San Francisco. Inferences on the general distribution of resis-

    tance likewise should not be made on the basis of data for

  • 8/3/2019 Bacterial Resistance Origins, Epidemiology

    7/13

    Bacterial Resistance CID 2003:36 (Suppl 1) S17

    Figure 1. Complications among patients with peritonitis, in relation to whether initial empirical treatment was appropriate (group 1, white); waschanged from inappropriate to appropriate after isolation of resistant bacteria (group 2, gray); or was not changed despite the isolation of resistant

    bacteria (group 3, black). Data are replotted from Mosdell et al. [77]. For total complications and reoperation, there were significant differences

    ( , x2 test) between groups 1 and 3, and for abscess formation between groups 1 and 2 and between 1 and 3. Differences in wound infectionP! .01

    rates do not achieve statistical significance. Average lengths of stay were 10.9, 14.8, and 19.0 days for group 1, 2, and 3 patients, respectively, with

    a significant difference ( , paired t test) between groups 1 and 3.P! .01

    isolates from ICUs, where resistance rates are commonly higher

    than among isolates overall [54, 55]. A finalandbiggersource

    of bias, particularly for surveys of community isolates, is that

    microbiological investigations are more likely to be performed

    for recalcitrant infections, which may be recalcitrant because

    they are caused by resistant bacteria. Thus, MacGowan et al.

    [75] found that only 3% of patients presenting with respiratory

    symptoms to general practitioners in Bristol received micro-

    biological investigation and that the apparent prevalence of

    ampicillin resistance among H. influenzae isolates decreased

    from 22% to 11% when all the presenting patients had sputumculture performed.

    IMPACT OF RESISTANCE

    The consequences of resistance are harder to define than mi-

    crobiologists, health care managers, and politicians might wish.

    Some patients recover despite inadequate treatment, exactly as

    some recovered before antibacterials were available. The infec-

    tion of others failed to respond despite appropriate therapy. In

    compromised patients, it often remains debatable whether in-

    fection or an underlying disease was ultimately fatal. Perhaps

    the clearest link between in vitro resistance and in vivo re-

    sponses for penicillin and gonorrhea. Classical strains with

    MIC 0.06 mg/L respond to penicillin at low doses; those with

    chromosomally mediated resistance (MIC 0.252 mg/L) re-

    spond to high-dose penicillin; and those with b-lactamase can-

    not be cured by any achievable level of penicillin. However,

    gonorrhea is an uncomplicated infection mostly caught by

    those in otherwise good health! Elsewhere, relationships are less

    clear and pseudomonal infection in cystic fibrosis presents the

    opposite extreme. Drugs that are active in vitro consistently fail

    to eliminate infection, whereas some of those that entirely lack

    in vitro antipseudomonal activity (e.g., erythromycin) often

    ameliorate the symptoms of infection [76].

    Most correlations fall between these extremes. Analysis by

    Mosdell et al. [77] showed that the incidence of complications,

    including reoperation, abscess formation, and wound infection,

    increased 2-fold if empirical therapy for intra-abdominal sep-

    sis failed to cover all the pathogens subsequently isolated and

    that the length of hospital stay was likewise extended (figure

    1). The incidence of complications rose further if inadequateempirical regimen was not modified when resistant pathogens

    were isolated. In recent analyses, Kollef and Ward [78] and

    Ibrahim et al. [79] found 2-fold higher mortality among ICU

    patients and those with ventilator-associated pneumonia when

    the pathogens proved resistant to the antibacterials used em-

    pirically (figure 2). Many of the failures in these series reflected

    infection by P. aeruginosa or MRSA, which were not well cov-

    ered by the empirical regimens routinely used. Risk factors for

    isolation of these pathogens and so for poor outcomes included

    previous use of antibacterials and previous hospitalization.Such

    factors, as well as the likely pathogens and their likely local

    resistance patterns, should always be taken into account whendesigning empirical regimens for hospital units. It should be

    added that the science of pharmacodynamics is allowing more

    precise modeling of the probability of cure of a given pathogen

    in a given infection, underscoring the fact that for many in-

    fection types, there is a strong relationship between in vitro

    susceptibility and outcome. These aspects are addressed else-

    where in this supplement by Drusano [80].

    Increased morbidity and mortality are the most dramatic

  • 8/3/2019 Bacterial Resistance Origins, Epidemiology

    8/13

    S18 CID 2003:36 (Suppl 1) Livermore

    Figure 2. Mortality among critically ill patients in intensive care ( ) in relation to whether empirical therapy was appropriate (gray) ornp 655inappropriate (black) in relation to the resistances of the bacteria subsequently isolated. Reproduced with permission from Ibrahim et al. [79].

    consequence of resistance. Other effects are more insidious.

    Physicians and surgeons are forced to use previously reserved

    agents as first-line therapy. These may be inherently less potent

    or more toxic that classical regimens: vancomycin is increas-

    ingly used as a first-line antistaphylococcal (and for prophy-laxis) but is less convenient to administer safely and less bac-

    tericidal than the semisynthetic antistaphylococcal penicillins,

    which themselves are 100-fold less active than benzylpenicillin

    against fully susceptible staphylococci. Previously reserved

    agentsnow used earliermay be undermined by resistance.

    The accumulation of cephalosporin-resistant bacteria is driving

    the earlier clinical use of carbapenems and is a reasonable jus-

    tification for the development of oral and long half-life car-

    bapenems. Nevertheless, the selection pressure that mass use

    of these will cause is disturbing, coming precisely when the

    number of reported carbapenemases is growing sharply [81].

    Finally, resistance adds cost: treatment failures extend thelength of hospital stay or demand repeated physician visits;

    hospital beds are blocked to new patients, and productive time

    is lost. If new or hitherto reserved antibacterials are needed as

    therapy, these are usually more expensive than previous regi-

    mens. These costs seem unlikely to decline in the future, es-

    pecially with the growing demand of regulators and the new

    costs of genomics-based drug discovery. A new antibacterial

    already costs $0.5 billion to develop; this sum and the fi-

    nancing costs (for the $0.5 billion is spent before income is

    generated) must be recouped in the 1012 years of patent life

    remaining after the compound is launched.

    RESPONSES TO RESISTANCE

    Concern about resistance increased in the late 1990s. Since then,

    many governmental and agency reports have been published,

    adding to those of professional societies [8284]. These reports

    vary in emphasis, especially as regards the agricultural use of

    antibacterials, but all advise (1) less use of antibacterials, (2)

    more appropriate choices of antibacterials and regimens, (3)

    prevention of cross-infection, and (4) development of new an-

    tibacterials. Measuring the effect of these charges demands bet-

    ter surveillance of resistance prevalence and of prescribing. Op-

    timists hold that it may be possible to reverse resistance trends,and pessimists hold that it may only be possible to slow the

    accumulation of resistance sufficiently to keep one step ahead

    of bacterial evolution.

    EFFECTS OF REDUCED PRESCRIBING

    In a few cases, reductions in prescribing at a national level have

    been followed by a reduced prevalence of resistance. In one

    example of success, the prevalence of penicillin-resistant pneu-

    mococci in Iceland was reduced from 19.3% (1993) to 14%

    (19982000) after a 12.9% reduction in drug use [85]. In Fin-

    land, a national advisory to reduce prescribing of macrolideswas followed by a decline in use from 3 doses/1000 population

    per month in 1988 to 1.1 doses/1000 population per month in

    1994 and by a decrease in the prevalence of erythromycin-

    resistant Streptococcus pyogenes from 19% in 1993 to 8.5% in

    1996 [86]. By 1998, however, macrolide use had increased to

    2.1 doses/1000 population per month, and the resistance rate

    among S. pyogenes was back to 18% (Bacterial Resistance to

    Antimicrobial Agents in Finland FINRES 1999; http://www

    .mmm.fi/el/julk/finres99en.htm). In both these cases, the re-

    sistances displaced were associated with clonal strainstwo

    widely disseminated S. pyogenes lineages in Finland [87]and,

    in Iceland, the serotype 6B S. pneumoniae strain mentioned

    earlier. Displacement seems more difficult when resistances are

    multiple and linked and when they have disseminated among

    different strains. Resistance to streptomycin and chloramphen-

    icol remains frequent in gram-negative bacteria, although these

    drugs have fallen into virtual disuse in humans, and are com-

    promised by mechanisms that do not directly affect any anti-

    bacterial that remains in extensive human use [88]. As a further

  • 8/3/2019 Bacterial Resistance Origins, Epidemiology

    9/13

  • 8/3/2019 Bacterial Resistance Origins, Epidemiology

    10/13

    S20 CID 2003:36 (Suppl 1) Livermore

    to imipenem was followed by the emergence of imipenem-

    resistant Acinetobacterand P. aeruginosa as well as by a decline

    in cephalosporin-resistant klebsiellae [96, 100]. Among op-

    portunistic infections in the seriously ill, the cynic can argue

    that resistance is like a balloon: squeeze it on one side, and it

    bulges on the other.

    It is salutary to emphasize how much remains unknown.

    Does drug cycling have positive effects, or does it lead to theaccumulation of multidrug-resistant strains [101]? At what

    prevalence of resistance should empirical therapy be changed

    in different types of infection? To what extent does combination

    therapy militate against resistance (except in the case of tu-

    berculosis, where its value is beyond dispute)? What are the

    relative selectivities of different antibacterials, allowing that a

    recent Finnish study found a correlation between macrolide use

    and resistance in S. pneumoniaebut not between penicillin use

    and resistance [102]? Is it in any way desirable to encourage

    all community physicians to use the same therapies in the same

    indications, or is it wiser to make the selection pressure more

    diffuse [103]? What is the ideal duration of therapy, allowingthat underdosing may fail to eliminate the least susceptible

    members of the original population and that excessive duration

    may exacerbate disruption of the normal flora? Are drugs with

    unlinked resistances (e.g., fosfomycin, nitrofurantoin, rifampin,

    and fucidin) to be favored as unlikely to select multiresistance

    plasmids or avoided because they may select hypermutable

    strains primed to develop further resistances? The new res-

    piratory quinolones exemplify a further conundrum. As a result

    of greater antipneumococcal activity, they are less likely to than

    ciprofloxacin to select first-step quinolone-resistant mutants of

    S. pneumoniae; however, they are less active than ciprofloxacin

    against Enterobacteriaceae and may be selective for resistance

    in the gut microflora [104], which is gratuitously exposed.

    SOCIAL CONTEXT

    The scientific answers to these questions on antibacterial use

    are uncertain, and to complicate matters, the whole problem

    of resistance is intertwined with moral, social, political, and

    commercial issues. Concern about resistance is used as am-

    munition for other agendas, most obviously including mar-

    keting by the pharmaceutical industry and cost containment

    within managed or socialized health care. To some extent, the

    individual patient gains when powerful antibacterials are used

    early, but the resistance risks for society are raised. In reality,

    matters are complex. Failed treatments with old and simple

    drugs may lead to more severe disease or to the spread of

    infection, resulting in a demand for further therapy, along with

    its contingent selection pressure. Hungary, before 1989, had a

    restricted list of antibacterials for community prescription, yet

    achieved one of the worlds highest prevalence rates for peni-

    cillin-resistant pneumococci [105]. Moreover, the argument as-

    sumes a vacuum in which no new drugs are developed. This

    assumption, made in many reports on resistance, is already

    untrue for gram-positive pathogens [106].

    Concerns about resistance have led to the banning of most

    agricultural growth promoters in Europe. Such concerns are

    also used to support wider objections to intensive farming andto the genetic modification of crop plants (where unexpressed

    resistance genes remain within the cloning vectors). Less is said

    on the other aspects of modern life that potentially exacerbate

    resistance: large hospitals; the concentration of the very young

    and very old in socialized care; and increasing travel. Action

    on these would be socially and politically impossible, even if

    they are more pertinent to the sum total of resistance than the

    use (recently banned in the European Union) of zinc bacitracin

    as a agricultural growth promoter! Perhaps the contradictions

    are best exemplified by the World Health Organization, which

    argues on the one hand for effective, affordable antibacterials

    for all the worlds population and on the other hand expresses

    concern about the worldwide accumulation of resistance

    (World Health Organization Report on Infectious Diseases

    2000, http://w ww.who.int/infectious-disease-report/2000/

    index.html). Both concerns are ethical, humane, and

    honorablebut counterpoised.

    CONCLUSIONS

    Antibacterial resistance is complex and dynamic. Although the

    major genetic and biochemical mechanisms have long beenrecognized, new factors continue to be discovered, including

    integrons, multidrug efflux, hypermutability, and plasmid ad-

    diction. Within many individual isolates, the complexity of

    resistance is increasing, with multiple determinants carried, and

    with genes being gained, amplified, and lost. Many international

    resistance problems reflect the spread of a few multidrug-resis-

    tant strains, but the reasons underlying the success of particular

    lineages remain almost universally obscure. Resistance is a sig-

    nificant cause of excess morbidity, mortality, and cost. Nu-

    merous reports have emphasized the need for less and better

    use of antibacterials, improved infection control, and the de-

    velopment of new agents. However, reductions in antibacterialuse do not always lead to reduced resistance, perhaps because

    bacteria are now well adapted to the carriage of resistance. It

    is probably naive to anticipate reaching a grand control over

    resistance, and attempts should center on management rather

    than elimination, with the objective of slowing the development

    of new resistance while continuing to develop new agents at a

    sufficient rate to keep ahead of the bacteria.

  • 8/3/2019 Bacterial Resistance Origins, Epidemiology

    11/13

    Bacterial Resistance CID 2003:36 (Suppl 1) S21

    References

    1. Woodford N. Glycopeptide-resistant enterococci: a decade of expe-

    rience. J Med Microbiol 1998; 47:84962.

    2. French GL. Enterococci and vancomycin resistance. Clin Infect Dis

    1998; 27(Suppl 1):S7583.

    3. Oppenheim BA. The changing pattern of infection in neutropenic

    patients. J Antimicrob Chemother 1998; 41:711.

    4. Gomez J, Simarro E, Banos V, et al. Six-year prospective study of risk

    and prognostic factors in patients with nosocomial sepsis caused by

    Acinetobacter baumannii. Eur J Clin Microbiol Infect Dis 1999;18:

    35861.

    5. Elting LS, Khardori N, Bodey GP, et al. Nosocomial infection caused

    by Xanthomonas maltophilia: a case-control study of predisposing

    factors. Infect Control Hosp Epidemiol 1990; 11:1348.

    6. VanCouwenberghe CJ, Farver TB, Cohen SH. Risk factors associated

    with isolation ofStenotrophomonas (Xanthomonas) maltophiliainclin-

    ical specimens. Infect Control Hosp Epidemiol 1997; 18:31621.

    7. Carmeli Y, Samore MH. Comparison of treatment with imipenem vs.

    ceftazidime as a predisposing factor for nosocomial acquisition of

    Stenotrophomonas maltophilia: a historical cohort study. Clin Infect

    Dis 1997; 24:11314.

    8. Bridges BA. Hypermutation in bacteria and other cellular systems.

    Philos Trans R Soc Lond B Biol Sci 2001; 356:2939.

    9. Podglajen I, Breuil J, Collatz E. Insertion of a novel DNA sequence,

    1S1186, upstream of the silent carbapenemase gene cfiA, promotesexpression of carbapenem resistance in clinical isolates of Bacteroides

    fragilis. Mol Microbiol 1994; 12:10514.

    10. Friedberg EC, Walker GC, Siede W. DNA repair and mutagenesis.

    Washington, DC: ASM Press, 1995.

    11. Rebeck GW, Samson L. Increased spontaneous mutation and alkyl-

    ation sensitivity of Escherichia coli strains lacking the ogt O6-meth-

    ylguanine DNA repair methyltransferase. J Bacteriol 1991;173:

    206876.

    12. Nema S, Premchandani P, Asolkar MV, et al. Emerging bacterial drug

    resistance in hospital practice. Indian J Med Sci 1997; 51:27580.

    13. Garau J, Xercavins M, Rodriguez-Carballeira M, et al. Emergence and

    dissemination of quinolone-resistant Escherichia coli in the commu-

    nity. Antimicrob Agents Chemother 1999; 43:273641.

    14. Wang H, Dzink-Fox JL, Chen M, Levy SB. Genetic characterization

    of highly fluoroquinolone-resistant clinical Escherichia coli strainsfrom China: role of acrR mutations. Antimicrob Agents Chemother

    2001; 45:151521.

    15. Everett MJ, Jin YF, Ricci V, Piddock LJ. Contributions of individual

    mechanisms to fluoroquinolone resistance in 36 Escherichia colistrains

    isolated from humans and animals. Antimicrob Agents Chemother

    1996; 40:23806.

    16. Hooper DC. Emerging mechanisms of fluoroquinolone resistance.

    Emerg Infect Dis 2001; 7:33741.

    17. Oethinger M, Kern WV, Jellen-Ritter AS, et al. Ineffectiveness of to-

    poisomerase mutations in mediating clinically significant fluoroquin-

    olone resistance in Escherichia coli in the absence of the AcrAB efflux

    pump. Antimicrob Agents Chemother 2000; 44:103.

    18. Ysern P, Clerch B, Castano M, et al. Induction of SOS genes in

    Escherichia coliand mutagenesis in Salmonella typhimurium by fluor-

    oquinolones. Mutagenesis 1990; 5:636.

    19. Oliver A, Canton R, Campo P, et al. High frequency of hypermutable

    Pseudomonas aeruginosa in cystic fibrosis lung infection. Science

    2000; 288:12514.

    20. Poole K. Multidrug efflux pumps and antimicrobial resistance in Pseu-

    domonas aeruginosaand related organisms. J Mol Microbiol Biotech-

    nol 2001; 3:25564.

    21. Li XZ, Livermore DM, Nikaido H. Role of efflux pump(s) in intrinsic

    resistance of Pseudomonas aeruginosa: resistance to tetracycline, chlo-

    ramphenicol, and norfloxacin. Antimicrob Agents Chemother 1994;

    38:173241.

    22. Lewis K. In search of natural substrates and inhibitorsof MDRpumps.

    J Mol Microbiol Biotechnol 2001; 3:24754.

    23. Kohler T, Epp SF, Curty LK, et al. Characterization of MexT, the

    regulator of the MexE-MexF-OprN multidrug efflux system of Pseu-

    domonas aeruginosa. J Bacteriol 1999; 181:63005.

    24. Barbosa TM, Levy SB. Differential expression of over 60 chromosomal

    genes in Escherichia coliby constitutive expression of MarA.J Bacteriol

    2000; 182:346774.

    25. Cohen SP, McMurry LM, Hooper DC, et al. Cross-resistance to fluor-

    oquinolones in multiple-antibioticresistant (Mar) Escherichia colise-

    lected by tetracycline or chloramphenicol: decreased drug accumu-

    lation associated with membrane changes in addition to OmpF

    reduction. Antimicrob Agents Chemother 1989; 33:131825.

    26. Piddock LJ. Mechanisms of fluoroquinolone resistance: an update

    19941998. Drugs 1999; 58(Suppl 2):118.

    27. Livermore DM. b-Lactamases in laboratory and clinical resistance.

    Clin Microbiol Rev 1995; 8:55784.

    28. Chow JW, Fine MJ, Shlaes DM, et al. Enterobacterbacteremia: clinical

    features and emergence of antibiotic resistance during therapy. Ann

    Intern Med 1991; 115:58590.

    29. Carmeli Y, Troillet N, Eliopoulos GM, et al. Emergence of antibiotic-

    resistant Pseudomonas aeruginosa: comparison of risks associated with

    different antipseudomonal agents. Antimicrob Agents Chemother

    1999; 43:137982.

    30. Quinn JP, Studemeister AE, DiVincenzo CA, et al. Resistance to im-

    ipenem in Pseudomonas aeruginosa: clinical experience and biochem-ical mechanisms. Rev Infect Dis 1988; 10:8928.

    31. Prystowsky J, Siddiqui F, Chosay J, et al. Resistance to linezolid: char-

    acterization of mutations in rRNA and comparison of their occur-

    rences in vancomycin-resistant enterococci. Antimicrob Agents Che-

    mother 2001; 45:21546.

    32. Tsiodras S, Gold HS, Sakoulas G, et al. Linezolid resistance in a clinical

    isolate of Staphylococcus aureus. Lancet 2001; 358:2078.

    33. Rice LB. Bacterial monopolists: the bundling and dissemination of

    antimicrobial resistance genes in gram-positive bacteria. Clin Infect

    Dis 2000; 31:7629.

    34. Hughes VM, Datta N. Conjugative plasmids in bacteria of the pre-

    antibiotic era. Nature 1983; 302:7256.

    35. Livermore DM. Antibiotic resistance in staphylococci. Int J Antimi-

    crob Agents 2000; 16(Suppl 1):S310.

    36. Hall RM. Mobile gene cassettes and integrons: moving antibiotic re-sistance genes in gram-negative bacteria. Ciba Found Symp 1997;

    207:192202.

    37. Hall RM, Collis CM, Kim MJ, et al. Mobile gene cassettes and in-

    tegrons in evolution. Ann N Y Acad Sci 1999; 870:6880.

    38. Martinez-Freijo P, Fluit AC, Schmitz FJ, et al. Many class I integrons

    comprise distinct stable structures occurring in different species of

    Enterobacteriaceae isolated from widespread geographic regions in

    Europe. Antimicrob Agents Chemother 1999; 43:6869.

    39. Chang CY, Chang LL, Chang YH, et al. Characterisation of drug

    resistance gene cassettes associated with class 1 integrons in clinical

    isolates of Escherichia coli from Taiwan, ROC. J Med Microbiol

    2000; 49:1097102.

    40. Enne VI, Livermore DM, Stephens P, et al. Persistence of sulphon-

    amide resistance in Escherichia coli in the UK despite national pre-

    scribing restriction. Lancet 2001; 357:13258.

    41. Courvalin P. Transfer of antibiotic resistance genes between gram-

    positive and gram-negative bacteria. Antimicrob Agents Chemother

    1994; 38:144751.

    42. Bauernfeind A, Chong Y, Lee K. Plasmid-encoded AmpC b-lacta-

    mases: how far have we gone 10 years after the discovery? Yonsei Med

    J 1998; 39:5205.

    43. Philippon A, Arlet G, Jacoby GA. Plasmid-determined AmpC-type

    b-lactamases. Antimicrob Agents Chemother 2002; 46:111.

    44. Oliver A, Perez-Diaz JC, Coque TM, et al. Nucleotide sequence and

    characterization of a novel cefotaxime-hydrolyzingb-lactamase(CTX-

  • 8/3/2019 Bacterial Resistance Origins, Epidemiology

    12/13

    S22 CID 2003:36 (Suppl 1) Livermore

    M-10) isolated in Spain. Antimicrob Agents Chemother 2001;45:

    61620.

    45. Trieu-Cuot P, Arthur M, Courvalin P. Origin, evolution and dissem-

    ination of antibiotic resistance genes. Microbiol Sci 1987; 4:2636.

    46. Sievert DM, Boulton ML, Stoltman G, et al. Staphylococcus aureus

    resistant to vancomycinUnited States, 2002. MMWR Morb Mortal

    Wkly Rep 2002; 51:5657.

    47. Marshall CG, Broadhead G, Leskiw BK, et al.d-ala-d-ala ligases from

    glycopeptide antibiotic-producing organisms are highly homologous

    to the enterococcal vancomycin-resistance ligases VanA and VanB.Proc Natl Acad Sci USA 1997; 94:64803.

    48. Livermore DM, Yuan M. Antibiotic resistance and production of ex-

    tended-spectrum b-lactamases amongst Klebsiellaspp. from intensive

    care units in Europe. J Antimicrob Chemother 1996; 38:40924.

    49. Babini GS, Livermore DM. Antimicrobial resistance amongst Kleb-

    siella spp. collected from intensive care units in Southern and Western

    Europe in 19971998. J Antimicrob Chemother 2000; 45:1839.

    50. Yuan M, Aucken H, Hall LM, et al. Epidemiological typing of kleb-

    siellae with extended-spectrum b-lactamases from European intensive

    care units. J Antimicrob Chemother 1998; 41:52739.

    51. Essack SY, Hall LM, Pillay DG, et al. Complexity and diversity of

    Klebsiella pneumoniae stains with extended-spectrum b-lactamases in

    1994 and 1996 at a teaching hospital in Durban, South Africa. An-

    timicrob Agents Chemother 2001; 45:8895.

    52. Spratt BG. Resistance to antibiotics mediated by target alterations.

    Science 1994; 264:38893.

    53. Enne VI, Livermore DM, Hall LMC. Sulfonamide resistance in Hae-

    mophilus influenzaemediated bysul2or an insertion in chromosomal

    folP [abstract 2275]. In: Program and Abstracts of the 40th Intersci-

    ence Conference on Antimicrobial Agents and Chemotherapy (To-

    ronto). Washington, DC: American Society for Microbiology, 2000.

    54. Archibald L, Phillips L, Monnet D, et al. Antimicrobial resistance in

    isolates from inpatients and outpatients in the United States: increas-

    ing importance of the intensive care unit. Clin Infect Dis 1997;24:

    2115.

    55. Voss A, Milatovic D, Wallrauch-Schwarz C, et al. Methicillin-resistant

    Staphylococcus aureus in Europe. Eur J Clin Microbiol Infect Dis

    1994; 13:505.

    56. Song JH, Lee NY, Ichiyama S, et al. Spread of drug-resistant Strep-

    tococcus pneumoniae in Asian countries: Asian Network for Surveil-

    lance of Resistant Pathogens (ANSORP) Study. Clin Infect Dis

    1999; 28:120611.

    57. Reacher MH, Shah A, Livermore DM, et al. Bacteraemia and antibiotic

    resistance of its pathogens reported in England and Wales between

    1990 and 1998: trend analysis. BMJ 2000; 320:2136.

    58. Baquero F. Pneumococcal resistance to beta-lactam antibiotics: a

    global geographic overview. Microb Drug Resist 1995; 1:11520.

    59. Kristinsson KG. Epidemiology of penicillin resistant pneumococci in

    Iceland. Microb Drug Resist 1995; 1:1215.

    60. Munoz R, Coffey TJ, Daniels M, et al. Intercontinental spread of a

    multiresistant clone of serotype 23F Streptococcus pneumoniae. J Infect

    Dis 1991; 164:3026.

    61. MZali FH, Heritage J, Gascoyne-Binzi DM, et al. Transcontinental

    importation into the UK of Escherichia coli expressing a plasmid-

    mediated AmpC-type b-lactamase exposed during an outbreak of

    SHV-5 extended-spectrum b-lactamase in a Leeds hospital. J Anti-

    microb Chemother 1997; 40:82331.

    62. Child JA. A study of b-lactamase mediated antibiotic resistance in

    clinical isolates originating from Punjab, India [thesis]. London: Uni-

    versity of London, 2001.

    63. Nordmann P, Ronco E, Naas T, et al. Characterization of a novel

    extended-spectrum b-lactamase from Pseudomonas aeruginosa. An-

    timicrob Agents Chemother 1993; 37:9629.

    64. Vahaboglu H, Ozturk R, Aygun G, et al. Widespread detection of

    PER-1type extended-spectrum b-lactamases among nosocomial Aci-

    netobacter and Pseudomonas aeruginosa isolates in Turkey: a nation-

    wide multicenter study. Antimicrob Agents Chemother 1997;41:

    22659.

    65. Danel F, Hall LM, Gur D, et al. Transferable production of PER-1b-

    lactamase in Pseudomonas aeruginosa. J Antimicrob Chemother

    1995; 35:28194.

    66. Johnson AP, Aucken H, Cavendish S, et al. Dominance of EMRSA-

    15 and -16 among MRSA causing nosocomial bacteraemia in the UK:

    analysis of isolates from the European Antimicrobial Resistance Sur-

    veillance System (EARSS). J Antimicrob Chemother 2001; 48:144.

    67. Bure A, Legrand P, Arlet G, et al. Dissemination in five French hos-

    pitals of Klebsiella pneumoniaeserotype K25 harbouring a new trans-

    ferable enzymatic resistance to third generation cephalosporins and

    aztreonam. Eur J Clin Microbiol Infect Dis 1988; 7:7802.

    68. Arlet G, Rouveau M, Casin I, et al. Molecular epidemiology ofKleb-

    siella pneumoniaestrains that produce SHV-4 b-lactamase and which

    were isolated in 14 French hospitals. J Clin Microbiol1994; 32:25538.

    69. Bosi C, Davin-Regli A, Bornet C, et al. Most Enterobacter aerogenes

    strains in France belong to a prevalent clone. J Clin Microbiol1999;37:

    21659.

    70. Pitt TL, Kaufmann ME, Patel PS, et al. Type characterisation and

    antibiotic susceptibility ofBurkholderia (Pseudomonas) cepaciaisolates

    from patients with cystic fibrosis in the United Kingdom and the

    Republic of Ireland. J Med Microbiol 1996; 44:20310.

    71. Casin I, Breuil J, Brisabois A, et al. Multidrug-resistant human and

    animal Salmonella typhimurium isolates in France belong predomi-

    nantly to a DT104 clone with the chromosome- and integron-encodedb-lactamase PSE-1. J Infect Dis 1999; 179:117382.

    72. Threlfall EJ. Antimicrobial drug resistance in Salmonella: problems

    and perspectives in food- and water-borne infections. FEMSMicrobiol

    Rev 2002; 26:1418.

    73. Willems RJ, Homan W, Top J, et al. Variant esp gene as a marker of

    a distinct genetic lineage of vancomycin-resistant Enterococcus faecium

    spreading in hospitals. Lancet 2001; 357:8535.

    74. Di Martino P, Livrelli V, Sirot D, et al. A new fimbrial antigen harbored

    by CAZ-5/SHV-4producing Klebsiella pneumoniae strains involved

    in nosocomial infections. Infect Immun 1996; 64:226673.

    75. MacGowan AP, Bowker KE, Bennett PM, et al. Surveillance of an-

    timicrobial resistance. Lancet 1998; 352:1783.

    76. Doring G, Conway SP, Heijerman HG, et al. Antibiotic therapy against

    Pseudomonas aeruginosa in cystic fibrosis: a European consensus. Eur

    Respir J 2000; 16:74967.77. Mosdell DM, Morris DM, Voltura A, et al. Antibiotic treatment for

    surgical peritonitis. Ann Surg 1991; 214:5439.

    78. Kollef MH, Ward S. The influence of mini-BAL cultures on patient

    outcomes: implications for the antibiotic management of ventilator-

    associated pneumonia. Chest 1998; 113:41220.

    79. Ibrahim EH, Sherman G, Ward S, et al. The influence of inadequate

    antimicrobial treatment of bloodstream infections on patient out-

    comes in the ICU setting. Chest 2000; 118:14655.

    80. Drusano GL. Prevention of resistance: a goal for dose selection for

    antimicrobial agents. Clin Infect Dis 2002; 35:in press.

    81. Livermore DM, Woodford N. Carbapenemases: a problem in waiting?

    Curr Opin Microbiol 2000; 3:48995.

    82. Nash JM. The antibiotics crisis: were in the midst of an escalating

    arms race with the microbe, and we may be losing. Time 2001; 157:

    902.

    83. Interagency Task Force on Antibiotic Resistance. A Public health ac-

    tion plan to combat antimicrobial resistance. Atlanta, GA: Centers

    for Diseases Control and Prevention, 2001.

    84. Shlaes DM, Gerding DN, John JF, et al. Society for Healthcare Epi-

    demiology of America and Infectious Diseases Society of America

    joint committee on the prevention of antimicrobial resistance guide-

    lines for the prevention of antimicrobial resistance in hospitals. Clin

    Infect Dis 1997; 25:58499.

    85. Austin DJ, Anderson RM. Studies of antibiotic resistance within the

    patient, hospitals and the community using simple mathematical

    models. Philos Trans R Soc Lond B Biol Sci 1999; 354:72138.

  • 8/3/2019 Bacterial Resistance Origins, Epidemiology

    13/13

    Bacterial Resistance CID 2003:36 (Suppl 1) S23

    86. Seppala H, Klaukka T, Vuopio-Varkila J, et al. The effect of changes

    in the consumption of macrolide antibiotics on erythromycin resis-

    tance in group A streptococci in Finland. Finnish Study Group for

    Antimicrobial Resistance. N Engl J Med 1997; 337:4416.

    87. Kataja J, Huovinen P, Muotiala A, et al. Clonal spread of group A

    streptococcus with the new type of erythromycin resistance. Finnish

    Study Group for Antimicrobial Resistance. J Infect Dis 1998;177:

    7869.

    88. Chiew YF, Yeo SF, Hall LM, et al. Can susceptibility to an antimicrobial

    be restored by halting its use? The case of streptomycin versus En-terobacteriaceae. J Antimicrob Chemother 1998; 41:24751.

    89. Bouma JE, Lenski RE. Evolution of a bacteria/plasmid association.

    Nature 1988; 335:3512.

    90. Lenski RE. The cost of antibiotic resistancefrom the perspective of

    a bacterium. Ciba Found Symp 1997; 207:13140.

    91. Rawlings DE. Proteic toxin-antitoxin, bacterial plasmid addiction sys-

    tems and their evolution with special reference to the pas system of

    pTF-FC2. FEMS Microbiol Lett 1999; 176:26977.

    92. King JW, White MC, Todd JR, et al. Alterations in the microbial flora

    and in the incidence of bacteremia at a university hospital after adop-

    tion of amikacin as the sole formulary aminoglycoside. Clin Infect

    Dis 1992; 14:90815.

    93. Betts RF, Valenti WM, Chapman SW, et al. Five-year surveillance of

    aminoglycoside usage in a university hospital. Ann Intern Med

    1984; 100:21922.94. Bradley SJ, Wilson AL, Allen MC, et al. The control of hyperendemic

    glycopeptide-resistant Enterococcus spp. on a haematology unit by

    changing antibiotic usage. J Antimicrob Chemother 1999; 43:2616.

    95. Levison M, Evangelista A, Hayburn J, et al. The effects of formulary

    conversion from ceftazidime to cefepime on in vitro susceptibility of

    Enterobacter cloacae at 7 medical centers [abstract 92]. In: Program

    and Abstracts of the 40th Interscience Conference on Antimicrobial

    Agents and Chemotherapy (Toronto). Washington, DC: American

    Society for Microbiology, 2000.

    96. Rahal JJ, Urban C, Horn D, et al. Class restriction of cephalosporin

    use to control total cephalosporin resistance in nosocomial Klebsiella.

    JAMA 1998; 280:12337.

    97. Rice LB. Successful interventions for gram-negative resistance to ex-

    tended-spectrum b-lactam antibiotics. Pharmacotherapy 1999;19:

    120S8S.

    98. Meyer KS, Urban C, Eagan JA, et al. Nosocomial outbreakofKlebsiella

    infection resistant to late-generation cephalosporins. Ann Intern Med

    1993; 119:3538.

    99. Friedland IR, Funk E, Khoosal M, et al. Increased resistance to am-

    ikacin in a neonatal unit following intensive amikacin usage. Antim-

    icrob Agents Chemother 1992; 36:1596600.

    100. Urban C, Go E, Mariano N, et al. Effect of sulbactam on infections

    caused by imipenem-resistant Acinetobacter calcoaceticus biotype an-

    itratus. J Infect Dis 1993; 167:44851.

    101. John JF. Antibiotic cycling: is it ready for prime time? Infect Control

    Hosp Epidemiol 2000; 21:911.

    102. Pihlajamaki M, Kotilainen P, Kaurila T, et al. Macrolide-resistant

    Streptococcus pneumoniaeand use of antimicrobial agents. Clin Infect

    Dis 2001; 33:4838.

    103. Millar MR, Russell E, Karcher AM, et al. Prescribing quality is not

    synonymous with cost minimization for antibiotics. J Antimicrob

    Chemother 2001; 47:3678.

    104. Kern WV, Oethinger M, Jellen-Ritter, et al. Non-target gene mutationsin the development of fluoroquinolone resistance in Escherichia coli.

    Antimicrob Agents Chemother 2000; 44:81420.

    105. Marton A, Meszner Z. Epidemiological studies on drug resistance in

    Streptococcus pneumoniaein Hungary: an update for the 1990s. Microb

    Drug Resist 1999; 5:2015.

    106. Livermore DM. Quinupristin/dalfopristin and linezolid: where, when,

    which and whether to use? J Antimicrob Chemother 2000; 46:34750.