13693780410001661482

Embed Size (px)

Citation preview

  • 7/30/2019 13693780410001661482

    1/5

    Biochemical characterization of terbinafine-resistant

    Trichophyton rubrum isolates

    BERTRAND FAVRE*%, MAHMOUD A. GHANNOUM$ & NEIL S. RYDER*

    *Novartis Research Institute, Vienna, Austria and$Center for Medical Mycology, Depar tment of Dermatology, Case Western

    Reserve University and University Hospitals of Cleveland, Cleveland, OH, USA

    We investigated the biochemical basis for resistance in six sequential clinical

    isolates ofTrichophyton rubrum , from the same patient, which exhibited high-level

    primary resistance to terbinafine. Cellular ergosterol biosynthesis was measured by

    incorporation of [14C]acetate, and microsomal squalene epoxidase was assayed by

    conversion of [3H]squalene to squalene epoxide and lanosterol. Direct comparison

    was made with a terbinafine-susceptible reference strain of T. rubrum in which

    squalene epoxidase was previously studied. Resistant isolates displayed normal

    cellular ergosterol biosynthesis, although slight accumulation of radiolabeled

    squalene suggested reduced squalene epoxidase activity. Ergosterol biosynthesis

    in the resistant isolates was only inhibited by terbinafine concentrations above 1 mg/ml (IC50 5 mg/ml). In the reference strain, ergosterol biosynthesis was eliminated by

    terbinafine at 0.03 mg/ml in accordance with historical data. There was no

    significant difference in sensitivity between the six resistant isolates. Squalene

    epoxidase from resistant strains was three orders of magnitude less sensitive than

    normal enzyme to terbinafine (IC50 of 30 mmol/l and 19 n mol/l respectively). The

    epoxidase in the resistant strains was also unresponsive to tolnaftate. Resistance to

    terbinafine in these T. rubrum isolates appears to be due to alterations in the

    squalene epoxidase gene or a factor essential for its activity.

    Keywords dermatophyte, resistance, Terbinafine, Trichophyton

    Introduction

    The allylamine terbinafine and the related compounds

    naftifine and butenafine are selective inhibitors of

    fungal squalene epoxidase (SE) [1]. This property is

    shared by the thiocarbamates tolnaftate and tolciclate

    [2], which are chemically distinct from the allylamines.

    Accumulation of squalene appears to be toxic to

    filamentous fungi, especially in dermatophytes [3].

    Despite the extensive use of terbinafine for the treat-

    ment of dermatophytosis and onychomycosis, the first

    clinically confirmed case of terbinafine resistance in

    dermatophytes was only recently reported [4]. Six

    sequential clinical isolates of Trichophyton rubrum

    originating from nail of a single patient who failed on

    therapy with oral terbinafine, were found to be resistant

    to terbinafine, with MIC ]/4 versus 5/0.001 mg/ml for

    normal strains. They were cross-resistant to the other

    SE inhibitors tested, butenafine, naftifine, tolnaftate

    and tolciclate, but normally susceptible to antifungals

    with a different mode of action, such as azoles and

    griseofulvin [4], as well as amorolfine and amphotericin

    B (N. S. Ryder, unpublished data, 2000). These results

    suggested a target-specific mechanism of resistance.

    The aim of the present study was to determine at the

    biochemical level whether resistance was indeed linked

    to abnormalities in the ergosterol biosynthesis pathway

    and SE enzyme.

    Correspondence: Neil S. Ryder, Infectious Diseases Biology, Novartis

    Institutes for BioMedical Research Inc., 100 Technology Square,

    Cambridge, MA 02139, USA. Tel.: '/1 617 871 3143; Fax: '/1 617

    871 7047; E-mail: [email protected]

    Received 8 September 2003; Accepted 16 December 2003

    % Present address: Laboratory of Dermatology, University Hospital

    CHUV, Lausanne, Switzerland.

    2004 ISHAM DOI: 10.1080/13693780410001661482

    Medical Mycology December 2004, 42, 525/529

  • 7/30/2019 13693780410001661482

    2/5

    Materials and methods

    Fungal isolates

    Six consecutive clinical isolates ofT. rubrum designated

    (Novartis Fungal Index) NFI5146 to NFI5151, includ-

    ing the baseline isolate NFI5146, were derived from

    nail material of a single patient with toenail onycho-mycosis sampled at intervals. Details of the clinical

    origin, culturing and mycological testing of these

    isolates have been provided previously [4]. For compar-

    ison, the reference strain T. rubrum NFI1895 was

    tested in parallel as this strain has been studied

    extensively in terms of cellular ergosterol biosynthesis

    and SE inhibition by terbinafine [5].

    Antifungal drugs

    Terbinafine (batch 97045) was synthesized at Novartis

    (Basel, Switzerland). Tolnaftate was purchased from

    Sigma (St Louis, MO, USA, catalogue T-6638) andamorolfine was obtained from Hoffmann La Roche

    (Basel, Switzerland).

    Cellular ergosterol biosynthesis

    The method for measurement of ergosterol biosynth-

    esis inhibition was modified from a previously de-

    scribed procedure [6]. Inocula were prepared from

    stocks frozen at /808C. Cultures were grown in 500-

    ml conical flasks containing 150 ml Sabouraud 2%

    dextrose broth pH 6.5, inoculated at 3)/104 to 5)/105

    colony forming units (c.f.u.) per ml and incubated on a

    rotary shaker (LH Engineering, Stoke Poges, UK) at100 r.p.m. and 308C for 4 to 6 days until adequate

    fungal mycelium was obtained. Because of the slow

    growth rate of dermatophytes in culture, the organisms

    were still in the growth phase when harvested. Cultures

    were harvested by gravity filtration on a glass sinter

    (porosity 1), washed in K-Na-PO4 buffer, pH 6.5, and

    resuspended in incubation medium. Incubation med-

    ium consisted of yeast nitrogen base medium without

    (NH4)2SO4 and amino acids, containing 2% glucose

    (w/v) and 25 mmol/l K-Na-PO4 buffer pH 6.5. Cell

    suspensions were adjusted to a density equivalent to

    3/4 mg dry weight per ml.

    Incubations were performed in 50-ml conical glass

    flasks in a shaking water bath at 308C. Inhibitors were

    dissolved in DMSO and added at 100-fold final

    concentration. Controls received solvent only. Cell

    suspension (5 ml) was pre-incubated 10 min with the

    test compound and the assay initiated by addition of 25

    ml substrate mixture prepared from [U-14C]-acetate

    (Amersham Biosciences, Vienna, Austria, 1 mCi/5 ml,

    50/62 mCi/mmol), 1 mol/l Na-acetate and distilled

    water mixed in the ratio 2:1:7. After an incubation time

    of 2 h, assays were terminated and cells harvested by

    filtration on Whatman (Maidstone, UK) 2.5 cm GF/A

    glass fiber filter discs. The cell mats were transferred to

    glass tubes, to which was added 1.5 ml 30% (w/v) KOH

    in 90% (v/v) ethanol, 1.5 ml ethanol and 0.3 ml 2%(w/v) 1,2,3-benzenetriol (pyrogallol) in ethanol. Tubes

    were closed with screw caps and left overnight in the

    dark at room temperature, then incubated 30 min at

    808C. After cooling, 1.5 ml water was added and the

    non-saponifiable lipids were extracted twice with 2 ml

    petroleum ether (bp 40/608C). The extract was washed

    with 3 ml distilled water, evaporated to dryness and

    dissolved in cyclohexane. The non-saponifiable lipids

    were then separated by thin layer chromatography as

    described previously [6].

    The TLC sheets were analyzed by electronic auto-

    radiography using the Instant Imager (Canberra Pack-

    ard, Vienna, Austria, with software Packard Image forWindows 2.10). The Instant Imager provided both an

    autoradiographic image of the TLC sheet and a

    quantitative determination of the distribution of radio-

    activity. The non-specific radioactivity which accumu-

    lated at the origin of the plates was not included in the

    calculations. After automatic background subtraction,

    the radioactivity was calculated for each band (squa-

    lene, sterols etc.) as a percentage of the sum of the

    designated products. These data were transferred from

    the Imager results file to a template in Excel 97 for

    calculation of percent inhibition of ergosterol biosynth-

    esis at each drug concentration and the IC50 value.

    Each experiment was performed with duplicate incuba-

    tions, and repeated so that each final value was derived

    from four separate determinations.

    Squalene epoxidase assay

    Trichophyton rubrum microsomes (170 000 g pellet)

    were prepared from mycelium frozen in liquid nitrogen

    [5]. Microsomal SE activity was assayed by measure-

    ment of incorporation of [4,8,12,13,17,21-3H]squalene

    (American Radiochemicals, St Louis, MO, USA) into

    squalene epoxide and lanosterol exactly as previously

    described [5], except for the final protein concentration

    of 3 mg protein/ml (instead of 2 mg/ml) and the

    incubation time of 60 min (instead of 45 min).

    Inhibitors were first dissolved in DMSO and then

    diluted 100-fold into the assay to achieve the desired

    final concentration. Experiments were performed in

    duplicate and repeated with each inhibitor twice. IC50values were calculated with the program Origin 6.1

    2004 ISHAM, Medical Mycology, 42, 525/529

    526 Favre et al.

  • 7/30/2019 13693780410001661482

    3/5

    (OriginLab Corporation, Northampton, MA, USA)

    using the curve fitting function.

    Results

    Reduced sensitivity of cellular ergosterol biosynthesis

    Analysis of the labeled non-saponifiable lipids in the

    absence of drug revealed that in the resistant isolate

    NFI5150 around 25% of the radioactivity was asso-

    ciated with squalene (Table 1). Similar results were

    observed with the other five resistant isolates (not

    shown). By contrast, in the reference strain NFI1895

    no significant radioactivity could be detected comigrat-

    ing with squalene or other ergosterol precursors (Table

    1), in agreement with earlier studies. This difference

    indicates that the flux of sterol precursors from

    [14C]acetate to ergosterol was not identical in the

    susceptible strain NFI1895 and the resistant isolates,

    and suggests the possibility of lower SE activity in thelatter. With the reference strain NFI1895, a terbinafine

    concentration of 0.004 mg/ml was sufficient to inhibit

    cellular ergosterol biosynthesis by 50%, while in the

    resistant isolates the IC50 was about 1000-fold higher

    (Table 2), confirming that inhibition of ergosterol

    biosynthesis underlies the antifungal activity of terbi-

    nafine in both susceptible and resistant isolates.

    Low sensitivity of squalene epoxidase

    A simple way of distinguishing target-based mechan-

    isms of resistance from the involvement of membrane

    transporters is by testing the sensitivity of the target tothe inhibitor in cell-free conditions, ruling out the

    action of transporters [7]. As the cellular studies

    revealed no apparent differences in sensitivity between

    the resistant isolates, the technically demanding studies

    with the SE enzyme were performed in only two of the

    resistant isolates. The specific activities of SE measured

    under the standard assay conditions were similar in the

    three isolates: 0.049/0.01 nmol/h/mg protein (9/SD,n0/5) for reference strain NFI1895, 0.049/0.01 nmol/h/

    mg protein (9/SD, n0/4) for NFI5146, and 0.059/0.01

    nmol/h/mg protein (9/SD, n0/4) for NFI5150. How-

    ever, a dramatic difference was found between the

    normal and resistant strains with respect to sensitivity

    Table 1 Comparison of radiolabeling patterns of ergosterol and intermediates in cells of Trichophyton rubrum NFI1895 (reference strain) and

    NFI5150 (resistant isolate) incubated with [14C]acetate and increasing concentrations of terbinafineaStrain Percent total counts measured in: (g/

    ml)

    Strain Terbinafine Ergosterol 4a-methyl-sterol Lanosterol Squalene

    NFI1895 0 929/1 29/0.5 49/1 29/0.3

    NFI1895 0.001 909/2 29/0.4 59/1 39/1

    NFI1895 0.003 479/28 29/1 49/2 479/31

    NFI1895 0.01 139/8 19/0.3 29/0.2 859/8NFI1895 0.03 59/2 19/0.2 19/0.2 949/3

    NFI5150 0 679/3 29/0.4 59/2 259/3

    NFI5150 0.01 679/3 39/0.4 69/2 249/4

    NFI5150 0.1 649/5 29/0.4 59/2 279/5

    NFI5150 1 589/6 39/0.4 69/2 339/6

    NFI5150 10 129/2 19/0.3 59/3 819/2

    aResults are given as mean9/standard deviation for four data points (two independent experiments each performed with duplicate incubations).

    Radiolabeling and analysis of lipids were performed as described in the Methods section.

    Table 2 Sensitivity of cellular ergosterol biosynthesis to terbinafine

    in reference and resistant strains

    Strain Visita Type Terbinafine IC50 (mg/ml)b

    NFI1895 / Reference 0.0049/0.002

    NFI5146 0 Resistant 5.59/0.9

    NFI5147 2 Resistant 4.19/0.8

    NFI5148 3 Resistant 5.09/0.4

    NFI5149 4 Resistant 4.29/1.2

    NFI5150 7 Resistant 5.09/1.2

    NFI5151 8 Resistant 5.49/2.5

    aClinical history: visit 0 was for screening and culture; therapy with

    terbinafine (250 mg once daily) lasting 24 weeks started at visit 1, and

    subsequent visits were at 6-week intervals [4]. bMean9/SD for four

    separate determinations.

    Table 3 Sensitivity of microsomal SE activity from reference and

    resistant strains to terbinafine, tolnaftate and amorolfineStrain IC50(mg/ml)a

    Strain Terbinafine Tolnaftate Amorolfine

    NFI1895 0.006 0.05 9

    NFI5146 11 /33 19

    NFI5150 9 /33 15

    aMean of IC50 values from two separate determinations, each of

    which did not vary by more than 30% from the mean value.

    2004 ISHAM, Medical Mycology, 42, 525/529

    Terbinafine-resistant T. rubrum 527

  • 7/30/2019 13693780410001661482

    4/5

    of SE to terbinafine. Microsomal SE activity from

    resistant isolates NFI5146 and NFI5150 was 1000-fold

    less sensitive to terbinafine than that from the reference

    strain NFI1895 (Table 3). A similar difference was

    observed with tolnaftate, in agreement with the cross-

    resistance of the isolates to thiocarbamates [4]. On the

    other hand, amorolfine, which also weakly inhibits T.

    rubrum SE [5], showed roughly similar activity against

    microsomal SE from reference and resistant strains

    (Table 3).

    Discussion

    At present, very little is known concerning drug-

    resistance mechanisms in dermatophytes. The recent

    discovery of the first terbinafine-resistant dermato-

    phyte associated with clinical failure [4] raised the

    important issue of the molecular mechanism of resis-

    tance to this widely used drug. Several mechanisms of

    resistance to another class of ergosterol biosynthesisinhibitors, the azoles, have been established in Candida

    species [8]. These include modification of the target

    enzyme, upregulation of the ergosterol biosynthesis

    pathway and, (most commonly) induction of drug

    efflux pumps leading to resistance to multiple drugs,

    including terbinafine in some cases [9]. We previously

    showed that the terbinafine-resistant T. rubrum isolates

    were fully cross-resistant to several classes of SE

    inhibitors, but displayed normal susceptibility to azoles

    and other antifungals [4]. This suggested that resistance

    was associated with changes in the target enzyme rather

    than with overexpression of efflux pumps. The data

    from the present study support this hypothesis. Bothcellular ergosterol biosynthesis and cell-free SE activity

    from resistant isolates displayed reduced sensitivity to

    terbinafine by several orders of magnitude. The resis-

    tant cells possessed a functioning ergosterol biosynth-

    esis pathway, but with an unusually high level of

    radiolabeled squalene in the control cells, which would

    be consistent with a reduced activity of squalene SE

    although alternative explanations are possible. Direct

    enzyme assays confirmed the existence of a functional

    SE in the resistant isolates and its abnormally low

    sensitivity to terbinafine. The specific activity of the

    resistant microsomal enzyme was similar to that from

    reference strain NFI1895, thus contradicting the evi-

    dence for reduced SE activity in intact cells. However,

    in vitro enzyme assays containing excess concentrations

    of cofactors and substrates may not detect differences

    in activity within living cells where these factors may be

    limiting. Interestingly, the resistant SE was also un-

    responsive to tolnaftate suggesting that allylamines and

    thiocarbamates may share a common binding site. In

    contrast, the sensitivity of the terbinafine-resistant SE

    to amorolfine was not very different from that of a

    normal enzyme suggesting that amorolfine has a

    mechanism of inhibition distinct from that of terbina-

    fine and tolnaftate. Amorolfine is a weak inhibitor ofT.

    rubrum SE [5], while the principal antifungal targets of

    the drug are sterol-D14-reductase and sterol-D7-D8-

    isomerase [10]. In the original report of the resistant

    isolates [4], there was an apparent further decrease in

    susceptibility (detectable only by the broth macro-

    dilution assay) over the course of terbinafine treatment.

    Since the sensitivity of ergosterol biosynthesis remained

    essentially unchanged over the sequence of isolates (see

    Table 2), this subtle change must have been due to a

    different, as yet unidentified, mechanism.

    The six resistant isolates, including one taken at

    baseline before start of therapy, were obtained sequen-

    tially from the same patient and displayed identical

    random amplified polymorphic DNA (RAPD) profiles

    [4], indicating that they represent a single genotypicstrain. Several lines of evidence point to a stable genetic

    alteration, probably in the SE gene, as the primary

    cause of terbinafine resistance in this case. These

    include (i) the drastic reduction in enzyme sensitivity,

    (ii) the rare occurrence of resistance, and (iii) the stable

    nature of the resistance in cultures taken over 24 weeks

    of therapy and after subculturing in the absence of the

    drug. Previous studies have shown that clinical failure

    of terbinafine was not associated with development of

    in-vitro resistance during therapy [11]. However, it

    appears that strains expressing primary resistance to

    the drug occur at a low frequency in the T. rubrum

    population. Occasional instances of high terbinafineMIC values have also been reported, but without

    clinical information [12]. Fungal SE genes have been

    cloned and sequenced from the yeasts Saccharomyces

    cerevisiae [13] and Candida albicans [14]. The amino

    acid substitution Leu0/Phe251, apparently associated

    with a terbinafine-resistant phenotype, was identified in

    the S. cerevisiae gene [14], and a second mutation

    leading to the substitution Pro0/Ala430 was recently

    reported to confer partial loss of sensitivity to inhibi-

    tion by terbinafine [15]. Investigations are in progress

    to determine whether amino acid substitution(s) are

    indeed fully responsible for terbinafine resistance in T.

    rubrum, and whether other mechanisms of resistance

    can also be detected.

    Acknowledgements

    We thank Wolfgang Mlineritsch for performing the

    cellular sterol biosynthesis assay.

    2004 ISHAM, Medical Mycology, 42, 525/529

    528 Favre et al.

  • 7/30/2019 13693780410001661482

    5/5

    References

    1 Ryder NS. Terbinafine: mode of action and properties of the

    squalene epoxidase inhibition. Br J Dermatol 1992; 126(Suppl.

    39): 2/7.

    2 Ryder NS, Frank I, Dupont MC. Ergosterol biosynthesis inhibi-

    tion by the thiocarbamate antifungal agents tolnaftate and

    tolciclate. Antimicrob Agents Chemother 1986; 29: 858/860.

    3 Ryder NS, Mieth H. Allylamine antifungal drugs. Curr Top Med

    Mycol 1992; 4: 158/188.

    4 Mukherjee PK, Leidich SD, Isham I, Leitner I, Ryder NS,

    Ghannoum MA. Clinical Trichophyton rubrum strain exhibiting

    primary resistance to terbinafine. Antimicrob Agents Chemother

    2003; 47: 82/86.

    5 Favre B, Ryder NS. Characterization of squalene epoxidase

    activity from the dermatophyte Trichophyton rubrum and its

    inhibition by terbinafine and other antimycotic agents. Antimicrob

    Agents Chemother 1996; 40: 443/447.

    6 Ryder NS. Specific inhibition of fungal sterol biosynthesis by SF

    86-327, a new allylamine antimycotic agent. Antimicrob Agents

    Chemother 1985; 27: 252/256.

    7 Favre B, Didmon M, Ryder NS. Multiple amino acid substitu-

    tions in lanosterol 14a-demethylase contribute to azole resistance

    in Candida albicans. Microbiology 1999; 145: 2715/2725.

    8 Balkis MM, Leidich SD, Mukherjee PK, Ghannoum MA.

    Mechanisms of fungal resistance: an overview. Drugs 2002; 62:

    1025/1040.

    9 Sanglard D, Ischer F, Monod M, Bille J. Cloning of Candida

    albicans genes conferring resistance to azole antifungal

    agents:characterization of CDR2, a new multidrug ABC trans-

    porter gene. Microbiology 1997; 143: 405/416.

    10 Polak A. Preclinical data and mode of action of amorolfine.

    Dermatology 1992; 184(Suppl. 1): 3/7.11 Bradley MC, Leidich S, Isham N, Elewski BE, Ghannoum MA.

    Antifungal susceptibilities and genetic relatedness of serial

    Trichophyton rubrum isolates from patients with onychomycosis

    of the toenail. Mycoses 1999; 42(Suppl. 2): 105/110.

    12 Fernandez-Torres B, Carrillo AJ, Martn E, et al. In vitro

    activities of 10 antifungal drugs against 508 dermatophyte strains.

    Antimicrob Agents Chemother 2001; 45: 2524/2528.

    13 Jandrositz A, Turnowsky F, Hogenauer G. The gene encoding

    squalene epoxidase from Saccharomyces cerevisiae : cloning and

    characterization. Gene 1991; 107: 155/160.

    14 Favre B, Ryder NS. Cloning and expression of squalene epoxidase

    from the pathogenic yeast Candida albicans. Gene 1997; 189: 119/

    126.

    15 Klobucnikova V, Kohut P, Leber R, et al. Terbinafine resistance in

    a pleiotropic yeast mutant is caused by a single point mutation inthe ERG1 gene. Biochem Biophys Res Comm 2003; 309: 666/671.

    2004 ISHAM, Medical Mycology, 42, 525/529

    Terbinafine-resistant T. rubrum 529