Abstract

The increasing incidence of invasive fungal infections is the result of many factors, including an increasing number of patients with severe immunosuppression. Although new drugs have been introduced to combat this problem, the development of resistance to antifungal drugs has become increasingly apparent, especially in patients who require long-term treatment or who are receiving antifungal prophylaxis, and there is growing awareness of shifts of flora to more-resistant species. The frequency, interpretation, and, in particular, mechanism of resistance to current classes of antifungal agents, particularly the azoles (where resistance has climbed most prominently) are discussed in this review.

Incidence of Fungal Infections

Life expectancy has increased by at least 10 years since the first use of antimicrobial drugs for the treatment of microbial infections [1]. Interestingly, this success has also increased the number of fungal infections. The incidence of fungal infections varies widely according to the underlying condition. In solid organ—transplant recipients, the incidence ranges from 5% after renal transplantation to 35% in lung and heart transplant recipients and up to 40% after liver transplantation [2, 3]. Cancer chemotherapy and allogeneic bone marrow transplantation are associated with fungal disease, and up to 30% of patients with acute leukemia experience invasive fungal infections. Candidemia has increased markedly in hospitalized patients. Mucosal and systemic fungal infections are also common in patients with AIDS. Oral candidiasis (or colonization of the mouth, pharynx, and esophagus) was seen in up to 84% of patients with HIV infection [4, 5], and 10% of patients with AIDS in the pre-HAART era developed cryptococcal meningitis, one of the most prevalent AIDS-defining infections [6]. Histoplasmosis, coccidioidomycosis, and infections with Penicillium spp. play important roles in certain geographical regions in both immunocompetent and immunocompromised hosts.

Definition of Resistance

The term “resistance” is used to describe a relative insensitivity of a microbe to an antimicrobial drug as tested in vitro and compared with other isolates of the same species. In contrast, clinical failure describes failure of an appropriate therapy for a certain indication to result in a clinical response. The reason for clinical failure may be, for example, antifungal resistance, but other causes, such as an impaired immune function, poor bioavailability of the drug given, or an accelerated metabolism of the drug, are possible causes of treatment failure [7].

Primary resistance occurs in organisms never exposed in that host to the drug of interest. In contrast, secondary resistance, also defined as acquired resistance, arises only after exposure of the organism to the drug. Intrinsic resistance is defined as resistance of all or almost all isolates of one species to a certain drug—for example, the resistance of Candida krusei to fluconazole. In addition, the term “clinical resistance” can be used to describe failure of therapy or relapse of infection with an organism not associated with in vitro resistance—for example, changes in the host's immune system, such as neutropenia [7].

In vitro—in vivo correlations for susceptibility abound in mycology [8], which emphasizes the predictive value of standardized testing. The National Committee for Clinical Laboratory Standards has published a standardized method (M27-A) for antifungal susceptibility testing of yeast [9] and has proposed a standardized method for testing filamentous fungi (M38-P) [10]. However, the many causes of treatment success and failure in vivo unrelated to drug susceptibility must be kept in mind, as must the fact that test breakpoints are never intended to be absolute discriminators ("resistance" is intended to convey a high, but not absolute, probability of failure).

Antifungal Drugs

The appropriate antifungal therapy and selection of drug should be based on criteria such as immune status of the host, site of infection, characteristics of the infection (the fungal species and its susceptibility to different antifungal drugs), and pharmacokinetic characteristics of the drug (e.g., absorption, elimination, and toxicity). Only a limited number of antimycotic drugs are available for the treatment of systemic fungal infections. The mode of action of the most important antifungal drugs can be divided into 4 different classes [11], as follows: (1) polyene macrolides that lead to an alteration of membrane functions (amphotericin B [AmB] and its lipid formulations); (2) azole derivatives that inhibit the 14α lanosterol demethylase, a key enzyme in ergosterol biosynthesis (ketoconazole, fluconazole, itraconazole, and voriconazole); (3) DNA and RNA synthesis inhibitors (flucytosine); and (4) 1,3-β-glucan synthase inhibitors (echinocandins).

Polyenes

Activity and mechanism of action. AmB is active against a wide variety of fungi, including yeasts and molds such as Candida spp. and Aspergillus spp., Cryptococcus neoformans, Zygomycetes, dimorphic fungi, and some dematiaceous fungi. The target structure is plasma membrane ergosterol, where AmB forms a channel. Through this channel, the fungal cell leaks potassium ions, resulting in a disruption of the proton gradient [12]. It is hypothesized that a ring of 8–10 polyene molecules form aqueous pores within the membrane bilayer structure. In addition, AmB causes oxidative damage to plasma membranes [11, 13]. In higher concentrations, polyenes also inhibit chitin synthase, a cell wall synthetic enzyme localized in the membrane. The interaction between AmB and human cell membranes containing cholesterol results in toxic side effects of the drug. The most important side effect is an impairment of renal function due to decreased filtration in the glomeruli.

Frequency of resistance. Reports of AmB resistance are limited. Primary resistance to AmB exists in some isolates of Candida lusitaniae [14], C. lipolytica [15], and C. guillermondii [16, 17]. Trichosporon beigelii [18, 19], Pseudallescheria boydii [20], and Scopulariopsis spp. often show intrinsic resistance to AmB. Until now, invasive infections caused by these fungi have been relatively infrequent. Secondary resistance has occasionally been described in Candida spp. as well as in C. neoformans [16, 17, 21–25]. In vitro susceptibility to AmB of Aspergillus species has, in one study, been correlated with the clinical outcome of invasive aspergillosis [26]. Nolte et al. [27] reported the isolation of AmB-resistant C. albicans from patients with leukemia. Bryce et al. [28] observed that Candida isolates with MIC < 0.78 μg/mL led to a higher mortality among patients with candidemia. Powderly et al. [17] noted that there is a correlation between failure of AmB therapy and in vitro resistance in yeasts isolated from cancer patients with invasive fungal infection. Lack of activity of AmB is described in systemic murine fusarial infection [29] as well as in clinical isolates [30]. In a study investigating 22 isolates of Fusarium spp., Arikan et al. [31] noted that AmB MICs spanned a relatively narrow range. The MICs were higher for Fusarium isolates than for Aspergillus isolates, with slightly higher MICs for F. oxysporum than for F. solani.

Whether repeated treatment with AmB, or other factors such as treatment with immunosuppressive chemotherapy, is responsible for polyene resistance still needs to be clarified. Patients with cryptococcal meningitis often receive AmB over long time periods [32]. Casadevall et al. [33] tested the susceptibility of C. neoformans isolates from patients with recurrent meningitis to AmB and found no increased MIC values. Reasons for recurrence might have been poor penetration of the drug, inconsistent compliance with other drug regimens, or deterioration of the immune system of the host.

It has been hypothesized that concurrent or previous therapy with azoles might lead to AmB resistance, such as AmB-refractory aspergillosis after itraconazole therapy [34]. The mechanism might be altered sterol type or content in the membrane, the result of the azole therapy. Kelly et al. [35] observed cross-resistance to AmB in 2 fluconazole-resistant C. albicans strains isolated from patients with AIDS. The strains were accumulating 14α-methylfecosterol instead of 14α-methylergosta-8,24-dien-3β,6α-diol, causing the cross-resistance. The same group reported 2 AmB-resistant mutants of Ustilago maydis with cross-resistance to nystatin. An alteration of the ergosterol biosynthesis was precluded as the cause of polyene resistance in these isolates [36]. Resistance to nystatin and AmB of a C. albicans isolate has been also documented in an immunocompetent trauma patient [37].

For Aspergillus terreus, it was shown in a murine model of disseminated aspergillosis that the mortality rate and survival time were identical for mice treated with AmB and untreated mice. This demonstrates the high resistance of this strain to AmB [38]. Only a few reports exist about the prevalence of resistance to the lipid formulations of AmB. Results of in vitro testing with these formulations may be of limited relevance because the active moiety of the complex in vivo is the amphotericin, which is released from sites of tissue binding of the lipid-amphotericin complex. Moreover, even for that fraction of drug circulating as the complex, tissue lipases may release the active drug in vivo [39]. Treatment with AmB is often guided by toxicity rather than by therapeutic efficacy, whereas lipid formulations have reduced toxicity, thus allowing higher doses to be administered. In neutropenic mice infected with an Aspergillus fumigatus strain with reduced susceptibility to AmB, it was shown that high doses of liposomal AmB or amphotericin lipid complex could overcome reduced susceptibility to the standard preparation, AmB deoxycholate [40]. It is known that lipid formulation alters tissue distribution [41]. Selective increased uptake in certain tissues could produce improved results.

Mechanism of resistance. The cause of resistance to polyenes is a significant alteration of the lipid composition in the plasma membrane (e.g., a reduction in the ergosterol content due to a lack of the Δ(8,7) isomerase) [42, 43]. This circumstance leads to a lower affinity of AmB to the plasma membrane, probably the result of a lack of the binding site [44, 45]. However, for a highly resistant strain of A. terreus, it was shown that the major sterol in its plasma membrane was ergosterol, suggesting that resistance to AmB was not related to a modified sterol composition [38].

Another cause for AmB resistance may be an altered content of β-1,3 glucans in the fungal cell wall. These components, which increase the stability of the cell wall, influence the access of large molecules such as AmB to the plasma membrane [46]. Seo et al. [47] showed that alteration of glucans led to AmB resistance in A. flavus.

Ergosterol Biosynthesis Inhibitors

Activity and mechanism of action. In the 1970s, a new class of antimycotic drugs was developed, the azoles. The first azole available for systemic use was clotrimazole. However, its use was limited largely because of inconsistent concentrations in the blood. Miconazole was the first effective azole for systemic infections, but its use was limited because it could only be given intravenously and it offered few advantages over amphotericin, aside from lessened toxicity [48].

Ketoconazole was the first azole to be orally available and showed consistent blood levels [49]. Susceptibility to ketoconazole was documented for the majority of Candida species, with the exception of C. glabrata [50, 51]. Ketoconazole was successfully used for the treatment of chronic mucocutaneous candidiasis. Ketoconazole also shows activity against Coccidioides, Histoplasma, Blastomyces, and dermatophytes. In addition to ketoconazole's rare hepatotoxicity, resistance was described, especially in patients with mucocutaneous candidiasis or AIDS and oropharyngeal and esophageal candidiasis [52–55].

Itraconazole, the first triazole used in humans, is available for oral (capsules and cyclodextrin solution) and intravenous administration. Cyclodextrin enhances the bioavailability of the drug [56], and the intravenous preparation offers the possibility of reliably high serum levels, which is especially a concern in patients with impaired absorption. Itraconazole shows broad in vitro activity against Candida spp., but also against Aspergillus spp. and dimorphic and dematiaceous fungi. Fluconazole, another triazole that can be administered orally and intravenously, is used for prophylaxis and treatment of oropharyngeal and esophageal candidiasis in patients with AIDS as well as in neutropenic patients [57, 58]. It could be demonstrated that after chemotherapy or bone marrow transplantation, the incidence of invasive candidiasis could be significantly reduced by using fluconazole prophylaxis [59–61]. In addition, fluconazole was used in nonneutropenic [62, 63] and surgical patients [64] with Candida colonization or invasive candidiasis. Fluconazole has only limited activity against molds and other filamentous fungi. Use of fluconazole in prophylaxis has resulted in a shift of host flora to resistant species and would be expected not to alter (and may even increase) the frequency of infection due to molds.

Voriconazole, structurally related to fluconazole, shows a spectrum similar to itraconazole. It is active against C. krusei, C. guillermondii, and C. lusitaniae [65]. Walsh et al. [66] recently published a randomized, international, multicenter trial comparing voriconazole with liposomal AmB for empirical antifungal treatment. They conclude that voriconazole is a suitable alternative to AmB preparations for empirical antifungal treatment in patients with neutropenia and persistent fever. In the same issue of the journal, other views, including that of the US Food and Drug Administration [67], as well as an editorial [68] concerning empirical antifungal therapy with voriconazole, were published. Voriconazole was approved in the United States for limited indications in May 2002. Posaconazole and ravuconazole, 2 additional triazoles, are currently under investigation. Posaconazole has in vitro and in vivo activity against Aspergillus [69], Cryptococcus [70], and Coccidioides [71] as well as Candida spp. [72]. Although penetration into CSF is low, there is activity against cerebral phaeohyphomycosis in animals. Ravuconazole (BMS 207147) shows in vitro activity against dimorphic fungi, such as Coccidioides, Histoplasma, and Blastomyces, as well as against Aspergillus, Fusarium, and fluconazole-sensitive and -resistant Candida species (e.g., C. krusei), and against the key dermatophytes [73].

Ergosterol is the most prevalent sterol in the fungal plasma membrane. Azoles act as ergosterol synthesis inhibitors by binding to lanosterol demethylase, a specific enzyme in ergosterol biosynthesis. The active binding site of lanosterol demethylase contains a heme domain [74, 75]. Azoles bind with a specific nitrogen atom in the azole ring nucleus to the iron atom of the heme domain, preventing the demethylation of lanosterol [76]. Azoles may also target lipids of the fungal plasma membranes [76] and may interact with the 3-ketosteroid reductase, an enzyme in methylsterol biosynthesis.

Frequency of resistance. Azole resistance is frequently described in patients with AIDS and mucosal candidiasis, oral candidiasis, or deep-seated candidiasis. Oropharyngeal colonization with Candida species was found in as many as 84% of patients infected with HIV, and symptomatic oral disease was found in up to 46% of these patients [4, 5]. Many of these patients require long-term treatment to suppress oropharyngeal and esophageal candidiasis. As a consequence, the number of patients with candidiasis refractory to azole treatment has increased markedly [77–84]. Resistance to azoles has been reported in 32% of symptomatic patients and in 14% of asymptomatic patients [4, 5, 85]. The prevalence correlates with the amount of CD4 cells, the fungal load, and the duration and doses of therapy [86]. Maenza et al. [86] reported that mean CD4 counts were lower (11 vs. 71 cells/mm3) and the duration of azole therapy was longer (419 vs. 118 days) in patients with fluconazole-resistant candidiasis. However, Vuffray et al. [87] showed that resistance can also occur in patients with high single doses of fluconazole. Recently, resistant C. neoformans was isolated from an immunocompetent patient without previous exposure to fluconazole [88]. Pelletier et al. [89] documented in HIV-infected children resistance of C. albicans to clotrimazole. They demonstrated a significant risk of cross-resistance to other azoles and showed that the resistance correlated with refractory mucosal candidiasis. Besides C. albicans, primary resistance has been also reported for C. dubliniensis, C. norvegensis, and C. inconspicua [90–92].

Candida species such as C. krusei are intrinsically resistant to fluconazole. They have been isolated in high frequency among HIV-positive patients [93], as well as cancer, bone marrow—transplant, and surgical patients [94–96]. There is controversy over the reason for the increasing incidence of intrinsically resistant non-albicans Candida species. The cause might be a selection of these species due to the widespread administration of fluconazole [95, 97]. Other causes might also be nosocomial transmission [98] or transmission between partners [99].

Mechanism of resistance. Different potential molecular mechanisms of azole resistance are possible. Some of these mechanisms are well known from antibacterial resistance. Modified target enzymes have been well characterized as a mechanism of resistance to penicillin in Streptococcus pneumoniae and Neisseria gonorrhoeae [100], efflux pumps have been implicated in resistance to chloramphenicol and tetracycline [101], and resistance to aminoglycosides has been associated with alterations in the plasma membrane [102]. Sometimes development of resistance to an azole drug leads to cross-resistance to other azoles, and sometimes the resistance is azole specific [103]; this will depend on the specificity of the resistance mechanism (e.g., the affinity of the target enzyme or the pump for a particular molecular structure). Rustad et al. [104] have recently reported a correlation of azole resistance in Candida albicans with homozygosity at the mating-type—like (MTL) locus, suggesting that key resistance genes may be regulated by this locus or in the vicinity of this locus.

Alteration of Drug Efflux

Alteration of drug efflux has been described as a recognized mechanism of resistance in bacteria (e.g., in S. pneumoniae resistance to macrolide antibiotics [105]). Furthermore, ATP-binding cassette transporters have been studied extensively and are known to cause drug resistance. These proteins span the plasma membrane several times and contain 2 ATP-binding cytoplasmic domains [106, 107]. In Candida, Candida drug resistance (CDR) genes have been associated with azole resistance. At least 5 different CDR genes (CDR1 to CDR5) have been described. CDR1 is a transporter protein in Candida and Cryptococcus, for which azoles (and other drugs—e.g., cycloheximide and chloramphenicol) are substrates [108, 109]. Its structure is related to the P-glycoprotein in humans, which is associated with resistance to chemotherapeutic drugs. Sanglard et al. [109] and Albertson et al. [110] compared the ability of C. albicans to accumulate fluconazole. In both studies, azole-resistant strains showed reduced intracellular concentrations of fluconazole. Northern blot testing showed as much as a 10-fold increase of CDR1 mRNA, indicating overexpression of CDR1. Parkinson et al. [111] described the accumulation of fluconazole in susceptible and resistant isolates of C. glabrata, where reduced ability of the resistant isolate to accumulate fluconazole was a consequence of energy-dependent drug efflux. Sanglard et al. [112] reported that C. albicans mutants that carry a CDR1 deletion showed hypersusceptibility to fluconazole, itraconazole, and ketoconazole as well as terbinafine and amorolfine. CDR2 overexpression was also shown to confer resistance to azoles. Strains with CDR1 and CDR2 double disruption were more hypersusceptible than strains with a single CDR1 disruption [113]. In addition, overexpression of MDR1 was observed in fluconazole-resistant C. albicans isolates [109, 114].

MDR1 (also known as BENr) was originally described to encode resistance to benomyl and methotrexate [115]. This transporter uses membrane potential instead of ATP as the energy source. Sanglard et al. [109] have demonstrated that if the Candida MDR1 gene was cloned into S. cerevisiae, overexpression of this gene potentially mediated resistance to azoles. Rhodamine 123, a fluorescent compound transported by MDR pumps, was used by Clark et al. [116] to search for the presence of these pumps in C. albicans and C. glabrata. They found evidence that in azole-resistant fungi, rhodamine 123 accumulated. In C. glabrata, a competition between rhodamine 123 and fluconazole for the transport out of the cell was demonstrated. Parkinson et al. [111] found an efflux competition between benomyl and fluconazole. Sanglard et al. [117] demonstrated that an increased expression of an ATP-binding cassette transporter gene plays a determinant role in the acquisition of resistance to azole antifungals in clinical isolates of C. glabrata. Recent studies suggest the resistance associated with brief exposures to high-dose azoles is transient (as opposed to stable resistance associated with long-term exposure to low-dose azoles) and associated with alterations in transcriptional regulation of CDR pump expression.

Alteration of the Target Enzyme

Overexpression of the target enzyme of the azoles, the lanosterol demethylase, was found to be a mechanism of resistance in C. glabrata [118]. This C. glabrata strain was resistant to fluconazole and itraconazole. An increased copy number of the lanosterol demethylase may result in increased ergosterol biosynthesis. However, in C. albicans, overexpression of the lanosterol demethylase probably accounts only for a minor effect on the development of resistance, because the expression demonstrated was only 3 times higher than in fluconazole-susceptible isolates [109].

As outlined above, azoles act as ergosterol synthesis inhibitors by binding to the lanosterol demethylase. The gene encoding this protein is ERG11, also known as ERG16, and the enzyme has been referred to as CYP51A1 in C. albicans. Alterations of this enzyme caused by defined point mutations in ERG11 have been described to be associated with azole resistance. White [119] described the presence of the amino acid substitution of lysine for arginine at position 467 (R467K). The substitution is located near the heme cofactor and thus causes structural and/or functional enzyme alterations. A substitution at amino acid 464 (G464S) in azole-resistant C. albicans isolates has been described [120]. Kelly et al. [121] confirmed that this mutation, resulting in changes in the heme-binding domain, causes fluconazole resistance through substantially reducing the inhibitory effect of fluconazole and described its association with perturbation of the heme environment. Similarly, this could be also demonstrated for the amino acid substitution T315A in C. albicans, where again the lanosterol demethylase activity was reduced 2-fold and the affinity to fluconazole was reduced [122], and for Y132H, a substitution that allows resistant mutants to produce ergosterol and retain fitness [123]. Additionally, in 5 clinical isolates of C. albicans, an amino acid substitution at position 105 from phenylalanine to leucine was described, potentially leading to reduced accessibility to the active binding site [120]. Fluconazole enters the active binding site through a channel and mutations therein affect the access of substrates and inhibitors. Marichal et al. [124] demonstrated that point mutations in this gene are clustered in 3 regions. Y132H was the only mutation found in the 2 N-terminal regions to be of importance for azole resistance, whereas in the C-terminal region, 3 mutations were associated with resistance.

Alteration of the ERG3 Genes

Altered sterol Δ(5,6) desaturase is another explanation for azole resistance. In azole-sensitive strains treated with azoles, 14-methyl-3,6-diol accumulates and leads to a fungistatic effect, whereas in sterol Δ(5,6) desaturase mutants (due to mutations in the gene ERG3), its precursor, 14-methylfecosterol, accumulates, which can support growth of the fungal cell. In S. cerevisiae, ∼20 fluconazole-resistant isolates were found to have altered sterol Δ(5,6) desaturase [125]. It has been suggested that ERG3 mutations alone can cause azole resistance in S. cerevisiae [126, 127].

In C. albicans, Kelly et al. [35, 128] demonstrated in 2 clinical isolates from patients with AIDS that resistance was caused by defective sterol Δ(5,6) desaturase, leading to an accumulation of 14-methylfecosterol. A consequence of this mechanism of resistance is that an absence of ergosterol causes cross-resistance to AmB. In addition, defective sterol Δ(5,6) desaturase has been described to be responsible for azole resistance in the plant pathogen U. maydis [129].

Alteration of the Drug Influx

The sterol composition of the plasma membrane can also affect the influx of drug into the cell. Alterations in the plasma membrane composition affect membrane fluidity and asymmetry, leading to a decreased uptake of drug [130]. It was demonstrated that altered phospholipid and fatty acid composition of the plasma membrane of C. albicans may lead to resistance to miconazole [131]. Hitchcock and Whittle [132] noted a larger lipid content and a lower polar-lipid-to-neutral-lipid ratio in an azole-resistant C. albicans isolate compared with wild-type strains, potentially causing a decreased permeability of the membrane and thus a reduced penetration of azoles. In a fluconazole-resistant C. albicans isolate, a decreased amount of ergosterol and a lower phosphatidylcholine : phosphatidylethanolamine ratio in the plasma membrane, which might be responsible for an altered uptake and thus for a reduced intracellular accumulation of fluconazole, was demonstrated [133]. In an itraconazole-resistant C. krusei isolate, it was indicated that reduced accumulation of drug accounts for resistance rather than drug efflux or modifications in the ergosterol [134].

Frequency of Mechanisms of Resistance

Perea et al. [135] studied matched sets of 20 fluconazole-susceptible and -resistant isolates from HIV-infected patients with oropharyngeal candidiasis, presumably treated with azoles. They found overexpression of efflux pumps in 85% of resistant isolates (the frequency of overexpresssion of CDR genes and the MDR1 gene was similar), amino acid substitutions in the target enzyme lanosterol demethylase in 58%–65%, and overexpression of the gene that encodes the enzyme in 35%–42%. Multiple mechanisms of resistance were found in 75% of resistant isolates. White et al. [136] addressed this question in a different way. They analyzed 38 random isolates, half resistant and half susceptible, and found overexpression of the CDR genes only in (some) resistant isolates and correlated with resistance. Neither MDR1, FLU1, or ERG11 overexpression nor amino acid point mutations correlated with resistance or were found frequently in resistant isolates. The latter study [136] suggests that putative resistance mechanisms found in resistant isolates, except for CDR-encoded pumps, may not be responsible for the resistance and that additional mechanisms of resistance remain to be uncovered. Both studies [135, 136] suggested CDR1 and CDR2 may be coregulated, and both reported additional data on cross-resistance to other azoles, in addition to that previously described [103].

Investigational Triazoles

In May 2002, voriconazole was approved in the United States for limited indications. Furthermore, to date, 2 new triazoles, posaconazole (Schering Plough) and ravuconazole (Bristol Myers Squibb), are being investigated. Both show potent activity against a variety of yeast and molds. Data on resistance against these azoles are still very limited. Pfaller et al. [137] reported that they were active against all species of Candida in children and in all cohorts of adult patients. For A. fumigatus, in 17 clinical isolates, most with elevated itraconazole MICs, susceptibility against itraconazole, posaconazole, ravuconazole, and voriconazole were tested. Posaconazole was the most active triazole against itraconazole-susceptible isolates. The authors suggested differences in activity and mechanisms of resistance for voriconazole and ravuconazole. They observed a complex pattern of cross-resistance and hypersusceptibility with these drugs [138]. Manavathu et al. [139] demonstrated that in voriconazole-resistant A. fumigatus strains, low-level cross-resistance exists between itraconazole, posaconazole, and voriconazole. The susceptible parent and the resistant strain accumulated similar amounts of voriconazole. Thus, they supposed that spontaneous mutants of A. fumigatus resistant to voriconazole could emerge and that the reduced susceptibility to voriconazole may not be due to reduced accumulation of the drug in the mycelia. Finally, for Scedosporium isolates, cross-resistance was observed among miconazole, itraconazole, and voriconazole. No resistance to posaconazole was reported [140].

5-Fluorocytosine

Activity and mode of action. Flucytosine (5-FC), a fluorinated pyrimidine, has been used to treat fungal infections since the 1960s. It has excellent penetration into body fluids. 5-FC is principally active against yeasts, including Candida spp. and C. neoformans. Today, 5-FC is usually administered in combination with other antifungal drugs. 5-FC is taken up into the fungal cell by the enzyme cytosine permease, then modified to 5-fluorouracil and converted to fluorouridine triphosphate, which in turn is incorporated into fungal RNA, causing disruption of protein synthesis [141]. 5-Fluorouracil also is converted to fluorodeoxyuridine monophosphate, which interferes with the enzyme thymidylate synthase. Inhibition of thymidylate synthase subsequently causes disruption of DNA synthesis [142].

Frequency of resistance. Primary resistance to 5-FC is not uncommon [143, 144]. 5-FC resistance also occurs in C. neoformans [12].

Mechanism of resistance. Two mechanisms of resistance have been described. First is a mutational decrease of activity of the cytosine permease or deaminase, leading to a decreased uptake or conversion of the drug. This mechanism is responsible for primary and intrinsic resistance [13]. Second is a loss of activity of uracil phosphoribosyltransferase, an enzyme responsible for conversion of 5-fluorouracil to 5-fluorouridylic acid [141, 145]. Whelan and Kerridge [146] described decreased activity of the uridine monophosphate pyrophosphorylase associated with resistance to 5-FC in C. albicans. In 29 clinical isolates of C. albicans with resistance to 5-FC, no cytosine permease—deficiency mutants were found. In contrast, for C. parapsilosis, a 5-FC—resistant strain was described that evolved while the patient received therapy and had >7% of the cytosine deaminase activity compared with the parent susceptible isolate [147].

Inhibitors of Glucan Synthesis

Activity and mode of action. The cell wall of fungi contains compounds that are not found elsewhere in nature. Some of these components may provide selective targets for antifungal drugs without target-associated toxicity in mammalian hosts. The fungal cell wall consists of a multilayer structure composed of glucan, chitin, mannan, and mannoprotein [148, 149].

Three antifungal compounds (caspofungin, FK-463, and VER-002) belonging to the class of echinocandins are currently available for clinical use or are under development [150]. All compounds inhibit the fungus-specific biosynthesis of 1,3 β-D-glucan [151]. They have potent in vitro activity against Candida spp. and some activity against Aspergillus, dimorphic molds, and Pneumocystis carinii [152]. As expected, isolates resistant to antifungal triazoles did not show cross-resistance to the 3 compounds [153].

Treatment of fungi with β-glucan synthase inhibitors causes noncompetitive inhibition of 1,3 β-glucan synthase with secondary effects on other constituents, such as an increase in the chitin content of the cell wall and a reduction in the ergosterol content of the fungal cell membrane [154].

Frequency of resistance. For C. dubliniensis, an organism closely related to C. albicans and often associated with oral candidiasis in HIV-infected patients, resistance to fluconazole has been described. It was shown that these resistant isolates were highly susceptible to an echinocandin [155]. Cuenca-Estrella et al. [156] compared the in vitro activity of an echinocandin to that of itraconazole and AmB against fluconazole-resistant isolates of Candida. They showed that the echinocandin was more potent against C. albicans, C. glabrata, C. krusei, and C. tropicalis but less potent in vitro against C. parapsilosis and C. guillermondii. Furthermore, it was shown that the relatively low efficacy of an echinocandin against C. neoformans may result from a reduced activity against the glucan synthase of C. neoformans [157].

Mechanism of resistance. Data on echinocandin resistance are limited and are based on laboratory-derived mutants of S. cerevisiae. In this fungus, the β-glucan synthase complex is encoded by 2 genes and regulated by a third. Kurtz and Douglas [158] and Kurtz [159] presumed that in S. cerevisiae, mutations in one of the genes, FKS1, cause resistance to echinocandins by alteration of β-glucan synthase. Echinocandins do not penetrate into the cytoplasm of the fungal cell. Thus, resistance mechanisms described for azoles, such as increased activity of efflux pumps or altered sterol composition of the plasma membrane, seem to be irrelevant. Kurtz and Douglas [158] and Kurtz [159] presumed that resistance to β-glucan synthase inhibitors may be very much alike in both organisms, S. cerevisiae and C. albicans.

Conclusions

Advances in medicine have led to more patients living longer. Commensurate with the growth in patients at risk, the number of patients with severe fungal infections has dramatically increased. Concern regarding the development of resistance to any of the few antifungal drugs available has developed. Although we are able to define certain mechanisms of drug resistance, continued efforts for a deeper understanding of the cellular and molecular mechanisms as well as the clinical components of antifungal resistance will be important [8]. In addition, new diagnostic tools for rapid, sensitive, and specific detection of fungi in clinical material, such as PCR-based techniques, are mandatory. Finally, our knowledge of drug-resistance mechanisms should maximize the utility of current drugs and assist in the development of new antifungal drugs and new treatment strategies.

References

1
Donowitz
GR
Mandell
GL
Beta-lactam antibiotics (1)
N Engl J Med
1988
, vol. 
318
 (pg. 
419
-
26
)
2
Kanj
SS
Welty
WK
Madden
J
, et al. 
Fungal infections in lung and heart-lung transplant recipients: report of 9 cases and review of the literature
Medicine (Baltimore)
1996
, vol. 
75
 (pg. 
142
-
56
)
3
Paya
CV
Fungal infections in solid-organ transplantation
Clin Infect Dis
1993
, vol. 
16
 (pg. 
677
-
88
)
4
Maenza
JR
Merz
WG
Romagnoli
MJ
Keruly
JC
Moore
RD
Gallant
JE
Infection due to fluconazole-resistant Candida in patients with AIDS: prevalence and microbiology
Clin Infect Dis
1997
, vol. 
24
 (pg. 
28
-
34
)
5
Martins
MD
Lozano-Chiu
M
Rex
JH
Point prevalence of oropharyngeal carriage of fluconazole-resistant Candida in human immunodeficiency virus—infected patients
Clin Infect Dis
1997
, vol. 
25
 (pg. 
843
-
6
)
6
Powderly
WG
Cryptococcal meningitis and AIDS
Clin Infect Dis
1993
, vol. 
17
 (pg. 
837
-
42
)
7
Rex
JH
Pfaller
MA
Galgiani
JN
, et al. 
Development of interpretive breakpoints for antifungal susceptibility testing: conceptual framework and analysis of in vitro—in vivo correlation data for fluconazole, itraconazole, and Candida infections. Subcommittee on Antifungal Susceptibility Testing of the National Committee for Clinical Laboratory Standards
Clin Infect Dis
1997
, vol. 
24
 (pg. 
235
-
47
)
8
Stevens
DA
Holmberg
K
Resistance to antifungal drugs: current status and clinical implications
Curr Opin Anti-Infect Invest Drugs
1999
, vol. 
1
 (pg. 
306
-
17
)
9
National Committee for Clinical Laboratory Standards
Reference method for broth dilution antifungal susceptibility testing of yeast. Standard M-27A
1997
Villanova, PA
National Committee for Clinical Laboratory Standards
10
National Committee for Clinical Laboratory Standards
Reference method for broth dilution antifungal susceptibility testing of conidium-forming filamentous fungi. Proposed standard M38-P
1998
Villanova, PA
National Committee for Clinical Laboratory Standards
11
Vanden Bossche
H
Willemsens
G
Marichal
P
Anti-Candida drugs—the biochemical basis for their activity
Crit Rev Microbiol
1987
, vol. 
15
 (pg. 
57
-
72
)
12
Vanden Bossche
H
Marichal
P
Odds
FC
Molecular mechanisms of drug resistance in fungi
Trends Microbiol
1994
, vol. 
2
 (pg. 
393
-
400
)
13
Vanden Bossche
H
Warnock
DW
Dupont
B
, et al. 
Mechanisms and clinical impact of antifungal drug resistance
J Med Vet Mycol
1994
, vol. 
32
 
1
(pg. 
189
-
202
)
14
Blinkhorn
RJ
Adelstein
D
Spagnuolo
PJ
Emergence of a new opportunistic pathogen, Candida lusitaniae
J Clin Microbiol
1989
, vol. 
27
 (pg. 
236
-
40
)
15
Walsh
TJ
Salkin
if
Dixon
DM
Hurd
NJ
Clinical, microbiological, and experimental animal studies of Candida lipolytica
J Clin Microbiol
1989
, vol. 
27
 (pg. 
927
-
31
)
16
Dick
JD
Rosengard
BR
Merz
WG
Stuart
RK
Hutchins
GM
Saral
R
Fatal disseminated candidiasis due to amphotericin-B—resistant Candida guilliermondii
Ann Intern Med
1985
, vol. 
102
 (pg. 
67
-
8
)
17
Powderly
WG
Kobayashi
GS
Herzig
GP
Medoff
G
Amphotericin B—resistant yeast infection in severely immunocompromised patients
Am J Med
1988
, vol. 
84
 (pg. 
826
-
32
)
18
Walsh
TJ
Newman
KR
Moody
M
Wharton
RC
Wade
JC
Trichosporonosis in patients with neoplastic disease
Medicine (Baltimore)
1986
, vol. 
65
 (pg. 
268
-
79
)
19
Walsh
TJ
Melcher
GP
Rinaldi
MG
, et al. 
Trichosporon beigelii, an emerging pathogen resistant to amphotericin B
J Clin Microbiol
1990
, vol. 
28
 (pg. 
1616
-
22
)
20
Lutwick
LI
Galgiani
JN
Johnson
RH
Stevens
DA
Visceral fungal infections due to Petriellidium boydii (Pseudallescheria boydii): in vitro drug sensitivity studies
Am J Med
1976
, vol. 
61
 (pg. 
632
-
40
)
21
Drutz
DJ
Lehrer
RI
Development of amphotericin B—resistant Candida tropicalis in a patient with defective leukocyte function
Am J Med Sci
1978
, vol. 
276
 (pg. 
77
-
92
)
22
Guinet
R
Chanas
J
Goullier
A
Bonnefoy
G
Ambroise
TP
Fatal septicemia due to amphotericin B—resistant Candida lusitaniae
J Clin Microbiol
1983
, vol. 
18
 (pg. 
443
-
4
)
23
Pappagianis
D
Collins
MS
Hector
R
Remington
J
Development of resistance to amphotericin B in Candida lusitaniae infecting a human
Antimicrob Agents Chemother
1979
, vol. 
16
 (pg. 
123
-
6
)
24
Safe
LM
Safe
SH
Subden
RE
Morris
DC
Sterol content and polyene antibiotic resistance in isolates of Candida krusei, Candida parakrusei, and Candida tropicalis
Can J Microbiol
1977
, vol. 
23
 (pg. 
398
-
401
)
25
Woods
RA
Bard
M
Jackson
IE
Drutz
DJ
Resistance to polyene antibiotics and correlated sterol changes in two isolates of Candida tropicalis from a patient with an amphotericin B—resistant funguria
J Infect Dis
1974
, vol. 
129
 (pg. 
53
-
8
)
26
Lass
FC
Kofler
G
Kropshofer
G
, et al. 
In-vitro testing of susceptibility to amphotericin B is a reliable predictor of clinical outcome in invasive aspergillosis
J Antimicrob Chemother
1998
, vol. 
42
 (pg. 
497
-
502
)
27
Nolte
FS
Parkinson
T
Falconer
DJ
, et al. 
Isolation and characterization of fluconazole- and amphotericin B—resistant Candida albicans from blood of two patients with leukemia
Antimicrob Agents Chemother
1997
, vol. 
41
 (pg. 
196
-
9
)
28
Bryce
EA
Roberts
FJ
Sekhon
AS
Coldman
AJ
Yeast in blood cultures: evaluation of factors influencing outcome
Diagn Microbiol Infect Dis
1992
, vol. 
15
 (pg. 
233
-
7
)
29
Anaissie
EJ
Hachem
R
Legrand
C
Legenne
P
Nelson
P
Bodey
GP
Lack of activity of amphotericin B in systemic murine fusarial infection
J Infect Dis
1992
, vol. 
165
 (pg. 
1155
-
7
)
30
Ellis
D
Amphotericin B: spectrum and resistance
J Antimicrob Chemother
2002
, vol. 
49
 
Supplement_1
(pg. 
7
-
10
)
31
Arikan
S
Lozano-Chiu
M
Paetznick
V
Nangia
S
Rex
JH
Microdilution susceptibility testing of amphotericin B, itraconazole and voriconazole against clinical isolates of Aspergillus and Fusarium species
J Clin Microbiol
1999
, vol. 
37
 (pg. 
3946
-
51
)
32
Eng
RH
Bishburg
E
Smith
SM
Kapila
R
Cryptococcal infections in patients with acquired immune deficiency syndrome
Am J Med
1986
, vol. 
81
 (pg. 
19
-
23
)
33
Casadevall
A
Spitzer
ED
Webb
D
Rinaldi
MG
Susceptibilities of serial Cryptococcus neoformans isolates from patients with recurrent cryptococcal meningitis to amphotericin B and fluconazole
Antimicrob Agents Chemother
1993
, vol. 
37
 (pg. 
1383
-
6
)
34
Schaffner
A
Bohler
A
Amphotericin B refractory aspergillosis after itraconazole: evidence for significant antagonism
Mycoses
1993
, vol. 
36
 (pg. 
421
-
4
)
35
Kelly
SL
Lamb
DC
Kelly
DE
, et al. 
Resistance to fluconazole and cross-resistance to amphotericin B in Candida albicans from AIDS patients caused by defective sterol Δ 5,6-desaturation
FEBS Lett
1997
, vol. 
400
 (pg. 
80
-
2
)
36
Joseph
HT
Manning
N
Holoman
D
Kelly
S
Nonsterol related resistance in Ustilago maydis to the polyene antifungals, amphotericin B and nystatin
Phytochemistry
1996
, vol. 
42
 (pg. 
637
-
9
)
37
Conly
J
Rennie
R
Johnson
J
Farah
S
Hellman
L
Disseminated candidiasis due to amphotericin B—resistant Candida albicans
J Infect Dis
1992
, vol. 
165
 (pg. 
761
-
4
)
38
Dannaoui
E
Borel
E
Persat
F
Piens
MA
Picot
S
Amphotericin B resistance of Aspergillus terreus in a murine model of disseminated aspergillosis
J Med Microbiol
2000
, vol. 
49
 (pg. 
601
-
6
)
39
Swenson
CE
Perkins
WR
Roberts
P
, et al. 
In vitro and in vivo antifungal activity of amphotericin B lipid complex: are phospholipases important?
Antimicrob Agents Chemother
1998
, vol. 
42
 (pg. 
767
-
71
)
40
Denning
DW
Warn
P
Dose range evaluation of liposomal nystatin and comparisons with amphotericin B and amphotericin B lipid complex in temporarily neutropenic mice infected with an isolate of Aspergillus fumigatus with reduced susceptibility to amphotericin B
Antimicrob Agents Chemother
1999
, vol. 
43
 (pg. 
2592
-
9
)
41
Townsend
RW
Zutshi
A
Bekersky
I
Biodistribution of 4-[14C]cholesterol-ambisome following a single intravenous administration to rats
Drug Metab Dispos
2001
, vol. 
29
 (pg. 
681
-
5
)
42
Kelly
SL
Lamb
DC
Taylor
M
Corran
AJ
Baldwin
BC
Powderly
WG
Resistance to amphotericin B associated with defective sterol Δ 8→7 isomerase in a Cryptococcus neoformans strain from an AIDS patient
FEMS Microbiol Lett
1994
, vol. 
122
 (pg. 
39
-
42
)
43
Subden
RE
Safe
L
Morris
DC
Brown
RG
Safe
S
Eburicol, lichesterol, ergosterol, and obtusifoliol from polyene antibiotic-resistant mutants of Candida albicans
Can J Microbiol
1977
, vol. 
23
 (pg. 
751
-
4
)
44
Hitchcock
CA
Barrett-Bee
KJ
Russell
NJ
The lipid composition and permeability to azole of an azole- and polyene-resistant mutant of Candida albicans
J Med Vet Mycol
1987
, vol. 
25
 (pg. 
29
-
37
)
45
Joseph
HT
Hollomon
D
Loeffler
RS
Kelly
SL
Cross-resistance to polyene and azole drugs in Cryptococcus neoformans
Antimicrob Agents Chemother
1995
, vol. 
39
 (pg. 
1526
-
9
)
46
Gale
EF
Nature and development of phenotypic resistance to amphotericin B in Candida albicans
Adv Microb Physiol
1986
, vol. 
27
 (pg. 
277
-
320
)
47
Seo
K
Akiyoshi
H
Ohnishi
Y
Alteration of cell wall composition leads to amphotericin B resistance in Aspergillus flavus
Microbiol Immunol
1999
, vol. 
43
 (pg. 
1017
-
25
)
48
Stevens
DA
The role of miconazole in systemic fungal infections
Am Rev Respir Dis
1977
, vol. 
116
 (pg. 
801
-
6
)
49
Borelli
D
Bran
JL
Fuentes
J
, et al. 
Ketoconazole, an oral antifungal: laboratory and clinical assessment of imidazole drugs
Postgrad Med J
1979
, vol. 
55
 (pg. 
657
-
61
)
50
Ahearn
DG
McGlohn
MS
In vitro susceptibilities of sucrose-negative Candida tropicalis, Candida lusitaniae, and Candida norvegensis to amphotericin B, 5-fluorocytosine, miconazole, and ketoconazole
J Clin Microbiol
1984
, vol. 
19
 (pg. 
412
-
6
)
51
Pfaller
MA
Burmeister
L
Bartlett
MS
Rinaldi
MG
Multicenter evaluation of four methods of yeast inoculum preparation
J Clin Microbiol
1988
, vol. 
26
 (pg. 
1437
-
41
)
52
Horsburgh
CR
Jr
Kirkpatrick
CH
Long-term therapy of chronic mucocutaneous candidiasis with ketoconazole: experience with twenty-one patients
Am J Med
1983
, vol. 
74
 
1B
(pg. 
23
-
9
)
53
Rodriguez-Tudela
JL
Martinez-Suarez
JV
Dronda
F
Laguna
F
Chaves
F
Valencia
E
Correlation of in-vitro susceptibility test results with clinical response: a study of azole therapy in AIDS patients
J Antimicrob Chemother
1995
, vol. 
35
 (pg. 
793
-
804
)
54
Rosenblatt
HM
Byrne
W
Ament
ME
Graybill
J
Stiehm
ER
Successful treatment of chronic mucocutaneous candidiasis with ketoconazole
J Pediatr
1980
, vol. 
97
 (pg. 
657
-
60
)
55
Warnock
DW
Johnson
EM
Richardson
MD
Vickers
CF
Modified response to ketoconazole of Candida albicans from a treatment failure [letter]
Lancet
1983
, vol. 
1
 
8325
(pg. 
642
-
3
)
56
Stevens
DA
Itraconazole in cyclodextrin solution
Pharmacotherapy
1999
, vol. 
19
 (pg. 
603
-
11
)
57
Como
JA
Dismukes
WE
Oral azole drugs as systemic antifungal therapy
N Engl J Med
1994
, vol. 
330
 (pg. 
263
-
72
)
58
Gallant
JE
Moore
RD
Chaisson
RE
Prophylaxis for opportunistic infections in patients with HIV infection
Ann Intern Med
1994
, vol. 
120
 (pg. 
932
-
44
)
59
Goodman
JL
Winston
DJ
Greenfield
RA
, et al. 
A controlled trial of fluconazole to prevent fungal infections in patients undergoing bone marrow transplantation
N Engl J Med
1992
, vol. 
326
 (pg. 
845
-
51
)
60
Slavin
MA
Osborne
B
Adams
R
, et al. 
Efficacy and safety of fluconazole prophylaxis for fungal infections after marrow transplantation—a prospective, randomized, double-blind study
J Infect Dis
1995
, vol. 
171
 (pg. 
1545
-
52
)
61
Walsh
TJ
Lee
JW
Prevention of invasive fungal infections in patients with neoplastic disease
Clin Infect Dis
1993
, vol. 
17
 
2
(pg. 
468
-
80
)
62
Rex
JH
Bennett
JE
Sugar
AM
, et al. 
A randomized trial comparing fluconazole with amphotericin B for the treatment of candidemia in patients without neutropenia. Candidemia Study Group and the National Institute of Allergy and Infectious Diseases Mycoses Study Group
N Engl J Med
1994
, vol. 
331
 (pg. 
1325
-
30
)
63
Solomkin
JS
Timing of treatment for nonneutropenic patients colonized with Candida
Am J Surg
1996
, vol. 
172
 
6A
(pg. 
44
-
8
)
64
Pittet
D
Monod
M
Suter
PM
Frenk
E
Auckenthaler
R
Candida colonization and subsequent infections in critically ill surgical patients
Ann Surg
1994
, vol. 
220
 (pg. 
751
-
8
)
65
Clancy
CJ
Fothergill
AW
Rinaldi
MG
Nguyen
MH
SCH 56592 and voriconazole against emerging yeast: in vitro efficacy compared with fluconazole and itraconazole [abstract]
Program and abstracts of the 39th Interscience Conference on Antimicrobial Agents and Chemotherapy (San Francisco)
1999
Washington, DC
American Society for Microbiology
pg. 
572
 
66
Walsh
TJ
Pappas
P
Winston
DJ
, et al. 
Voriconazole compared with liposomal amphotericin B for empirical antifungal therapy in patients with neutropenia and persistent fever
N Engl J Med
2002
, vol. 
346
 (pg. 
225
-
34
)
67
Powers
JH
Dixon
CA
Goldberger
MJ
Voriconazole versus liposomal amphotericin B in patients with neutropenia and persistent fever
N Engl J Med
2002
, vol. 
346
 (pg. 
289
-
90
)
68
Marr
KA
Empirical antifungal therapy—new options, new tradeoffs
N Engl J Med
2002
, vol. 
346
 (pg. 
278
-
80
)
69
Petraitiene
R
Petraitis
V
Groll
A
, et al. 
Efficacy of SCH 56592 in treatment and prevention of invasive pulmonary aspergillosis in persistently neutropenic rabbits [abstract]
Program and abstracts of the 39th Interscience Conference on Antimicrobial Agents and Chemotherapy (San Francisco)
1999
Washington, DC
American Society for Microbiology
pg. 
581
 
70
Yildiran
ST
Saracli
MA
Fothergill
AW
Rinaldi
MG
In vitro susceptibilities of environmental Cryptococcus neoformans isolates to six antifungal agents including SCH 56592 and voriconazole [abstract]
Program and abstracts of the 39th Interscience Conference on Antimicrobial Agents and Chemotherapy (San Francisco)
1999
Washington, DC
American Society for Microbiology
pg. 
572
 
71
Catanzaro
A
Cloud
G
Stevens
DA
, et al. 
Safety and tolerance of posaconazole (SCH 56592) in patients with nonmeningeal disseminated coccidioidomycosis [abstract]
Program and abstracts of the 40th Interscience Conference on Antimicrobial Agents and Chemotherapy (Toronto)
2000
Washington, DC
American Society for Clinical Microbiology
pg. 
382
 
72
Vazquez
JA
Northland
R
Miller
S
, et al. 
Posaconazole compared to fluconazole for oral candidiasis in HIV positive patients [abstract]
Program and Abstracts of the 40th Interscience Conference on Antimicrobial Agents and Chemotherapy (Toronto)
2000
Washington, DC
American Society for Clinical Microbiology
pg. 
372
 
73
Olsen
SJ
Mummaneni
V
Rolan
P
, et al. 
Ravuconazole single ascending oral dose study in healthy subjects [abstract]
Program and Abstracts of the 40th Interscience Conference on Antimicrobial Agents and Chemotherapy (Toronto)
2000
Washington, DC
American Society for Clinical Microbiology
pg. 
22
 
74
Hitchcock
CA
Cytochrome P-450—dependent 14 α-sterol demethylase of Candida albicans and its interaction with azole antifungals
Biochem Soc Trans
1991
, vol. 
19
 (pg. 
782
-
7
)
75
Vanden Bossche
H
Prasad
R
Ergosterol biosynthesis inhibitors
Candida albicans
1991
Berlin
Springer Verlag
(pg. 
239
-
57
)
76
Joseph-Horne
T
Hollomon
DW
Molecular mechanisms of azole resistance in fungi
FEMS Microbiol Lett
1997
, vol. 
149
 (pg. 
141
-
9
)
77
Dupont
B
Bennett
JE
Hay
RJ
Peterson
PK
Antifungal therapy in AIDS patients
New strategies in fungal diseases
1992
Edinburgh
Churchill Livingstone
(pg. 
290
-
300
)
78
Fox
R
Neal
KR
Leen
CL
Ellis
ME
Mandal
BK
Fluconazole resistant Candida in AIDS
J Infect
1991
, vol. 
22
 (pg. 
201
-
4
)
79
Heinic
GS
Stevens
DA
Greenspan
D
, et al. 
Fluconazole-resistant Candida in AIDS patients: report of two cases
Oral Surg Oral Med Oral Pathol
1993
, vol. 
76
 (pg. 
711
-
5
)
80
Kitchen
VS
Savage
M
Harris
JR
Candida albicans resistance in AIDS [letter]
J Infect
1991
, vol. 
22
 (pg. 
204
-
5
)
81
Ng
TT
Denning
DW
Fluconazole resistance in Candida in patients with AIDS—a therapeutic approach
J Infect
1993
, vol. 
26
 (pg. 
117
-
25
)
82
Sanguineti
A
Carmichael
JK
Campbell
K
Fluconazole-resistant Candida albicans after long-term suppressive therapy
Arch Intern Med
1993
, vol. 
153
 (pg. 
1122
-
4
)
83
Smith
D
Boag
F
Midgley
J
Gazzard
B
Fluconazole resistant Candida in AIDS [letter]
J Infect
1991
, vol. 
23
 (pg. 
345
-
6
)
84
Willocks
L
Leen
CL
Brettle
RP
Urquhart
D
Russell
TB
Milne
LJ
Fluconazole resistance in AIDS patients
J Antimicrob Chemother
1991
, vol. 
28
 (pg. 
937
-
9
)
85
Revankar
SG
Kirkpatrick
WR
McAtee
RK
, et al. 
Detection and significance of fluconazole resistance in oropharyngeal candidiasis in human immunodeficiency virus—infected patients
J Infect Dis
1996
, vol. 
174
 (pg. 
821
-
7
)
86
Maenza
JR
Keruly
JC
Moore
RD
Chaisson
RE
Merz
WG
Gallant
JE
Risk factors for fluconazole-resistant candidiasis in human immunodeficiency virus—infected patients
J Infect Dis
1996
, vol. 
173
 (pg. 
219
-
25
)
87
Vuffray
A
Durussel
C
Boerlin
P
, et al. 
Oropharyngeal candidiasis resistant to single-dose therapy with fluconazole in HIV-infected patients [letter]
AIDS
1994
, vol. 
8
 (pg. 
708
-
9
)
88
Orni
WR
Izkhakov
E
Siegman
IY
Bash
E
Polacheck
I
Giladi
M
Fluconazole-resistant Cryptococcus neoformans isolated from an immunocompetent patient without prior exposure to fluconazole
Clin Infect Dis
1999
, vol. 
29
 (pg. 
1592
-
3
)
89
Pelletier
R
Peter
J
Antin
C
Gonzalez
C
Wood
L
Walsh
TJ
Emergence of resistance of Candida albicans to clotrimazole in human immunodeficiency virus—infected children: in vitro and clinical correlations
J Clin Microbiol
2000
, vol. 
38
 (pg. 
1563
-
8
)
90
Baily
GG
Moore
CB
Essayag
SM
de Wit
S
Burnie
JP
Denning
DW
Candida inconspicua, a fluconazole-resistant pathogen in patients infected with human immunodeficiency virus
Clin Infect Dis
1997
, vol. 
25
 (pg. 
161
-
3
)
91
Moran
GP
Sullivan
DJ
Henman
MC
, et al. 
Antifungal drug susceptibilities of oral Candida dubliniensis isolates from human immunodeficiency virus (HIV)-infected and non-HIV—infected subjects and generation of stable fluconazole-resistant derivatives in vitro
Antimicrob Agents Chemother
1997
, vol. 
41
 (pg. 
617
-
23
)
92
Sandven
P
Nilsen
K
Digranes
A
Tjade
T
Lassen
J
Candida norvegensis: a fluconazole-resistant species
Antimicrob Agents Chemother
1997
, vol. 
41
 (pg. 
1375
-
6
)
93
Johnson
EM
Davey
KG
Szekely
A
Warnock
DW
Itraconazole susceptibilities of fluconazole susceptible and resistant isolates of five Candida species
J Antimicrob Chemother
1995
, vol. 
36
 (pg. 
787
-
93
)
94
Gleason
TG
May
AK
Caparelli
D
Farr
BM
Sawyer
RG
Emerging evidence of selection of fluconazole-tolerant fungi in surgical intensive care units
Arch Surg
1997
, vol. 
132
 (pg. 
1197
-
201
)
95
Wingard
JR
Merz
WG
Rinaldi
MG
Johnson
TR
Karp
JE
Saral
R
Increase in Candida krusei infection among patients with bone marrow transplantation and neutropenia treated prophylactically with fluconazole
N Engl J Med
1991
, vol. 
325
 (pg. 
1274
-
7
)
96
Wingard
JR
Merz
WG
Rinaldi
MG
Miller
CB
Karp
JE
Saral
R
Association of Torulopsis glabrata infections with fluconazole prophylaxis in neutropenic bone marrow transplant patients
Antimicrob Agents Chemother
1993
, vol. 
37
 (pg. 
1847
-
9
)
97
Price
MF
LaRocco
MT
Gentry
LO
Fluconazole susceptibilities of Candida species and distribution of species recovered from blood cultures over a 5-year period
Antimicrob Agents Chemother
1994
, vol. 
38
 (pg. 
1422
-
7
)
98
Merz
WG
Karp
JE
Schron
D
Saral
R
Increased incidence of fungemia caused by Candida krusei
J Clin Microbiol
1986
, vol. 
24
 (pg. 
581
-
4
)
99
Dromer
F
Improvisi
L
Dupont
B
, et al. 
Oral transmission of Candida albicans between partners in HIV-infected couples could contribute to dissemination of fluconazole-resistant isolates
AIDS
1997
, vol. 
11
 (pg. 
1095
-
101
)
100
Dowson
CG
Coffey
TJ
Spratt
BG
Origin and molecular epidemiology of penicillin-binding-protein—mediated resistance to beta-lactam antibiotics
Trends Microbiol
1994
, vol. 
2
 (pg. 
361
-
6
)
101
Cohen
SP
Hachler
H
Levy
SB
Genetic and functional analysis of the multiple antibiotic resistance (mar) locus in Escherichia coli
J Bacteriol
1993
, vol. 
175
 (pg. 
1484
-
92
)
102
Leclercq
R
Dutka
MS
Brisson
NA
, et al. 
Resistance of enterococci to aminoglycosides and glycopeptides
Clin Infect Dis
1992
, vol. 
15
 (pg. 
495
-
501
)
103
Stevens
DA
Stevens
JA
Cross-resistance phenotypes of fluconazole-resistant Candida species: results with 655 clinical isolates with different methods
Diagn Microbiol Infect Dis
1996
, vol. 
26
 (pg. 
145
-
8
)
104
Rustad
TR
Stevens
DA
Pfaller
MA
White
TC
Homozygosity at the Candida albicans MTL loci associated with azole resistance
Microbiology
2002
, vol. 
148
 (pg. 
1061
-
72
)
105
Sutcliffe
J
Tait-Kamradt
A
Wondrack
L
Streptococcus pneumoniae and Streptococcus pyogenes resistant to macrolides but sensitive to clindamycin: a common resistance pattern mediated by an efflux system
Antimicrob Agents Chemother
1996
, vol. 
40
 (pg. 
1817
-
24
)
106
Higgins
CF
ABC transporters: from microorganisms to man
Annu Rev Cell Biol
1992
, vol. 
8
 (pg. 
67
-
113
)
107
Jenkinson
HF
Ins and outs of antimicrobial resistance: era of the drug pumps
J Dent Res
1996
, vol. 
75
 (pg. 
736
-
42
)
108
Prasad
R
De Wergifosse
P
Goffeau
A
Balzi
E
Molecular cloning and characterization of a novel gene of Candida albicans, CDR1, conferring multiple resistance to drugs and antifungals
Curr Genet
1995
, vol. 
27
 (pg. 
320
-
9
)
109
Sanglard
D
Kuchler
K
Ischer
F
Pagani
JL
Monod
M
Bille
J
Mechanisms of resistance to azole antifungal agents in Candida albicans isolates from AIDS patients involve specific multidrug transporters
Antimicrob Agents Chemother
1995
, vol. 
39
 (pg. 
2378
-
86
)
110
Albertson
GD
Niimi
M
Cannon
RD
Jenkinson
HF
Multiple efflux mechanisms are involved in Candida albicans fluconazole resistance
Antimicrob Agents Chemother
1996
, vol. 
40
 (pg. 
2835
-
41
)
111
Parkinson
T
Falconer
DJ
Hitchcock
CA
Fluconazole resistance due to energy-dependent drug efflux in Candida glabrata
Antimicrob Agents Chemother
1995
, vol. 
39
 (pg. 
1696
-
9
)
112
Sanglard
D
Ischer
F
Monod
M
Bille
J
Susceptibilities of Candida albicans multidrug transporter mutants to various antifungal agents and other metabolic inhibitors
Antimicrob Agents Chemother
1996
, vol. 
40
 (pg. 
2300
-
5
)
113
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 transporter gene
Microbiology
1997
, vol. 
143
 
2
(pg. 
405
-
16
)
114
White
TC
Increased mRNA levels of ERG16, CDR, and MDR1 correlate with increases in azole resistance in Candida albicans isolates from a patient infected with human immunodeficiency virus
Antimicrob Agents Chemother
1997
, vol. 
41
 (pg. 
1482
-
7
)
115
Fling
ME
Kopf
J
Tamarkin
A
Gorman
JA
Smith
HA
Koltin
Y
Analysis of a Candida albicans gene that encodes a novel mechanism for resistance to benomyl and methotrexate
Mol Gen Genet
1991
, vol. 
227
 (pg. 
318
-
29
)
116
Clark
FS
Parkinson
T
Hitchcock
CA
Gow
NA
Correlation between rhodamine 123 accumulation and azole sensitivity in Candida species: possible role for drug efflux in drug resistance
Antimicrob Agents Chemother
1996
, vol. 
40
 (pg. 
419
-
25
)
117
Sanglard
D
Ischer
F
Calabrese
D
Majcherczyk
PA
Bille
J
The ATP binding cassette transporter gene CgCDR1 from Candida glabrata is involved in the resistance of clinical isolates to azole antifungal agents
Antimicrob Agents Chemother
1999
, vol. 
43
 (pg. 
2753
-
76
)
118
Vanden Bossche
H
Marichal
P
Odds
FC
Le Jeune
L
Coene
MC
Characterization of an azole-resistant Candida glabrata isolate
Antimicrob Agents Chemother
1992
, vol. 
36
 (pg. 
2602
-
10
)
119
White
TC
The presence of an R467K amino acid substitution and loss of allelic variation correlate with an azole-resistant lanosterol 14α demethylase in Candida albicans
Antimicrob Agents Chemother
1997
, vol. 
41
 (pg. 
1488
-
94
)
120
Loffler
J
Kelly
SL
Hebart
H
Schumacher
U
Lass
FC
Einsele
H
Molecular analysis of cyp51 from fluconazole-resistant Candida albicans strains
FEMS Microbiol Lett
1997
, vol. 
151
 (pg. 
263
-
8
)
121
Kelly
SL
Lamb
DC
Loeffler
J
Einsele
H
Kelly
DE
The G464S amino acid substitution in Candida albicans sterol 14α-demethylase causes fluconazole resistance in the clinic through reduced affinity
Biochem Biophys Res Commun
1999
, vol. 
262
 (pg. 
174
-
9
)
122
Lamb
DC
Kelly
DE
Schunck
WH
, et al. 
The mutation T315A in Candida albicans sterol 14α-demethylase causes reduced enzyme activity and fluconazole resistance through reduced affinity
J Biol Chem
1997
, vol. 
272
 (pg. 
5682
-
8
)
123
Kelly
SL
Lamb
DC
Kelly
DE
Y132H substitution in Candida albicans sterol 14α-demethylase confers fluconazole resistance by preventing binding to haem
FEMS Microbiol Lett
1999
, vol. 
180
 (pg. 
171
-
5
)
124
Marichal
P
Koymans
L
Willemsens
S
, et al. 
Contribution of mutations in the cytochrome P450 14α-demethylase (Erg11p, Cyp51p) to azole resistance in Candida albicans
Microbiology
1999
, vol. 
145
 
10
(pg. 
2701
-
13
)
125
Kelly
SL
Kelly
DE
Maresca
B
Kobayashi
GS
Yamaguchi
H
Molecular studies on azole sensitivity in fungi
Molecular biology and its application to medical mycology
1993
Berlin
Springer Verlag
(pg. 
199
-
213
)
126
Arthington
BA
Bennett
LG
Skatrud
PL
, et al. 
Cloning, disruption and sequence of the gene encoding yeast C-5 sterol desaturase
Gene
1991
, vol. 
102
 (pg. 
39
-
44
)
127
Kelly
SL
Lamb
DC
Corran
AJ
Baldwin
BC
Kelly
DE
Mode of action and resistance to azole antifungals associated with the formation of 14 α-methylergosta-8,24(28)-dien-3 β,6 α-diol
Biochem Biophys Res Commun
1995
, vol. 
207
 (pg. 
910
-
5
)
128
Kelly
SL
Lamb
DC
Kelly
DE
Loeffler
J
Einsele
H
Resistance to fluconazole and amphotericin in Candida albicans from AIDS patients [letter]
Lancet
1996
, vol. 
348
 
9040
(pg. 
1523
-
4
)
129
Joseph-Horne
T
Manning
NJ
Hollomon
D
Kelly
SL
Defective sterol Δ 5 (6) desaturase as a cause of azole resistance in Ustilago maydis
FEMS Microbiol Lett
1995
, vol. 
127
 (pg. 
29
-
34
)
130
Parks
LW
Casey
WM
Prasad
R
Ghannoum
MA
Fungal sterols
Lipids of pathogenic fungi
1996
Boca Raton, FL
CRC Press
(pg. 
63
-
82
)
131
Mago
N
Khuller
GK
Influence of lipid composition on the sensitivity of Candida albicans to antifungal agents
Indian J Biochem Biophys
1989
, vol. 
26
 (pg. 
30
-
3
)
132
Hitchcock
CA
Whittle
PT
Rippon
JW
Fromtling
RA
Chemistry and mode of action of fluconazole
Cutaneous antifungal agents: selected compounds in clinical practice and development
1993
New York
Marcel Dekker
(pg. 
183
-
97
)
133
Loffler
J
Einsele
H
Hebart
H
Schumacher
U
Hrastnik
C
Daum
G
Phospholipid and sterol analysis of plasma membranes of azole-resistant Candida albicans strains
FEMS Microbiol Lett
2000
, vol. 
185
 (pg. 
59
-
63
)
134
Venkateswarlu
K
Denning
DW
Manning
NJ
Kelly
SL
Reduced accumulation of drug in Candida krusei accounts for itraconazole resistance
Antimicrob Agents Chemother
1996
, vol. 
40
 (pg. 
2443
-
6
)
135
Perea
S
Lopez-Ribot
JL
Kirkpatrick
WR
, et al. 
Prevalence of molecular mechanisms of resistance to azole antifungal agents in Candida albicans strains displaying high-level fluconazole resistance isolated from human immunodeficiency virus—infected patients
Antimicrob Agents Chemother
2001
, vol. 
45
 (pg. 
2676
-
84
)
136
White
TC
Holleman
S
Dy
F
Mirels
LF
Stevens
DA
Analysis of resistance mechanisms in clinical isolates of Candida albicans
Antimicrob Agents Chemother
2002
, vol. 
46
 (pg. 
1704
-
13
)
137
Pfaller
MA
Diekema
DJ
Jones
RN
Messer
SA
Holly
RJ
Trends in antifungal susceptibility of Candida species isolated from pediatric and adult patients with bloodstream infections: SENTRY Antimicrobial Surveillance Program, 1997 to 2000
J Clin Microbiol
2002
, vol. 
40
 (pg. 
852
-
60
)
138
Mosquera
J
Denning
DW
Azole cross-resistance in Aspergillus fumigatus
Antimicrob Agents Chemother
2002
, vol. 
46
 (pg. 
556
-
7
)
139
Manavathu
EK
Abraham
OC
Chandrasekar
PH
Isolation and in vitro susceptibility to amphotericin B, itraconazole and posaconazole of voriconazole-resistant laboratory isolates of Aspergillus fumigatus
Clin Microbiol Infect
2001
, vol. 
7
 (pg. 
130
-
7
)
140
Meletiadis
J
Meis
JF
Mouton
JW
Rodriquez-Tudela
JL
Donnelly
JP
Verweij
PE
In vitro activities of new and conventional antifungal agents against clinical Scedosporium isolates
Antimicrob Agents Chemother
2002
, vol. 
46
 (pg. 
62
-
8
)
141
Polak
A
Scholer
HJ
Mode of action of 5-fluorocytosine and mechanisms of resistance
Chemotherapy
1975
, vol. 
21
 (pg. 
113
-
30
)
142
Diasio
RB
Bennett
JE
Myers
CE
Mode of action of 5-fluorocytosine
Biochem Pharmacol
1978
, vol. 
27
 (pg. 
703
-
7
)
143
Stiller
RL
Bennett
JE
Scholer
HJ
Wall
M
Polak
A
Stevens
DA
Susceptibility to 5-fluorocytosine and prevalence of serotype in 402 Candida albicans isolates from the United States
Antimicrob Agents Chemother
1982
, vol. 
22
 (pg. 
482
-
7
)
144
Defever
KS
Whelan
WL
Rogers
AL
Beneke
ES
Veselenak
JM
Soll
DR
Candida albicans resistance to 5-fluorocytosine: frequency of partially resistant strains among clinical isolates
Antimicrob Agents Chemother
1982
, vol. 
22
 (pg. 
810
-
5
)
145
Normark
S
Schonebeck
J
In vitro studies of 5-fluorocytosine resistance in Candida albicans and Torulopsis glabrata
Antimicrob Agents Chemother
1973
, vol. 
2
 (pg. 
114
-
21
)
146
Whelan
WL
Kerridge
D
Decreased activity of UMP pyrophosphorylase associated with resistance to 5-fluorocytosine in Candida albicans
Antimicrob Agents Chemother
1984
, vol. 
26
 (pg. 
570
-
4
)
147
Hoeprich
PD
Ingraham
JL
Kleker
E
Winship
MJ
Development of resistance to 5-fluorocytosine in Candida parapsilosis during therapy
J Infect Dis
1974
, vol. 
130
 (pg. 
112
-
8
)
148
Poulain
D
Tronchin
G
Dubremetz
JF
Biguet
J
Ultrastructure of the cell wall of Candida albicans blastospores: study of its constitutive layers by the use of a cytochemical technique revealing polysaccharides
Ann Microbiol (Paris)
1978
, vol. 
129
 (pg. 
141
-
53
)
149
Sullivan
PA
Yin
CY
Molloy
C
Templeton
MD
Shepherd
MG
An analysis of the metabolism and cell wall composition of Candida albicans during germ-tube formation
Can J Microbiol
1983
, vol. 
29
 (pg. 
1514
-
25
)
150
Hector
RF
Compounds active against cell walls of medically important fungi
Clin Microbiol Rev
1993
, vol. 
6
 (pg. 
1
-
21
)
151
Denning
DW
Echinocandins and pneumocandins—a new antifungal class with a novel mode of action
J Antimicrob Chemother
1997
, vol. 
40
 (pg. 
611
-
4
)
152
Current
WL
Boylan
CJ
Rabb
PP
Anti-Pneumocystis activity of LY 303366 and other echinocandin B analogues [abstract]
Program and abstracts of the 33rd Interscience Conference on Antimicrobial Agents and Chemotherapy (New Orleans)
1993
Washington, DC
American Society for Microbiology
pg. 
186
 
153
Vazquez
JA
Lynch
M
Boikov
D
Sobel
JD
In vitro activity of a new pneumocandin antifungal, L-743,872, against azole-susceptible and -resistant Candida species
Antimicrob Agents Chemother
1997
, vol. 
41
 (pg. 
1612
-
4
)
154
Pfaller
M
Riley
J
Koerner
T
Effects of cilofungin (LY121019) on carbohydrate and sterol composition of Candida albicans
Eur J Clin Microbiol Infect Dis
1989
, vol. 
8
 (pg. 
1067
-
70
)
155
Quindos
G
Carrillo-Munoz
AJ
Arevalo
MP
, et al. 
In vitro susceptibility of Candida dubliniensis to current and new antifungal agents
Chemotherapy
2000
, vol. 
46
 (pg. 
395
-
401
)
156
Cuenca-Estrella
M
Mellado
E
Diaz-Guerra
TM
Monzon
A
Rodriguez-Tudela
JL
Susceptibility of fluconazole-resistant clinical isolates of Candida species to echinocandin LY303366, itraconazole and amphotericin B
J Antimicrob Chemother
2000
, vol. 
46
 (pg. 
475
-
7
)
157
Feldmesser
M
Kress
Y
Mednick
A
Casadevall
A
The effect of the echinocandin analogue caspofungin on cell wall glucan synthesis by Cryptococcus neoformans
J Infect Dis
2000
, vol. 
182
 (pg. 
1791
-
5
)
158
Kurtz
MB
Douglas
CM
Lipopeptide inhibitors of fungal glucan synthase
J Med Vet Mycol
1997
, vol. 
35
 (pg. 
79
-
86
)
159
Kurtz
MB
New antifungal drugs: a vision for the future
1997
ASM News
(pg. 
31
-
9
)

Comments

0 Comments
Submit a comment
You have entered an invalid code
Thank you for submitting a comment on this article. Your comment will be reviewed and published at the journal's discretion. Please check for further notifications by email.