<<

View metadata, citation and similar papers at core.ac.uk brought to you by CORE

provided by Elsevier - Publisher Connector

History of the development of derivatives J. A. Maertens

Department of Haematology, University Hospital Gasthuisberg, Leuven, Belgium

ABSTRACT Until the 1940s, relatively few agents were available for the treatment of systemic fungal infections. The development of the polyene represented a major advance in medical mycology. Although quickly became the mainstay of therapy for serious infections, its use was associated with infusion-related side-effects and dose-limiting nephrotoxicity. The continued search for new and less toxic antifungals led to the discovery of the several decades later. , the first available compound for the oral treatment of systemic fungal infections, was released in the early 1980s. For almost a decade, ketoconazole was regarded as the drug of choice in nonlife-threatening endemic mycoses. The introduction of the first-generation represented a second major advance in the treatment of fungal infections. Both fluconazole and displayed a broader spectrum of activity than the and had a markedly improved safety profile compared with amphotericin B and ketoconazole. Despite widespread use, however, these agents became subject to a number of clinically important limitations related to their suboptimal spectrum of activity, the development of resistance, the induction of hazardous drug–drug interactions, their less than optimal pharmacokinetic profile (itraconazole capsules), and toxicity. In order to overcome these limitations, several analogues have been developed. These so-called ‘second-generation’ triazoles, including , and , have greater potency and possess increased activity against resistant and emerging pathogens, in particular against Aspergillus spp. If the toxicity profile of these agents is comparable to or better than that of the first-generation triazoles and drug interactions remain manageable, then these compounds represent a true expansion of our antifungal arsenal. Keywords Antifungal, azole, overview

Clin Microbiol Infect 2004; 10 (Suppl. 1): 1–10

death due to bacterial sepsis, thereby setting the INTRODUCTION stage for fungal colonisation and putting patients Despite the implementation of several preventive at risk for subsequent mycotic infections. Medical measures and the use of antifungal chemopro- procedures have become more invasive and phylaxis, physicians have witnessed an increased aggressive; the accompanying disruption of pro- incidence of both mucosal and invasive fungal tective anatomical barriers as a result of indwell- infections during the past two decades [1–4]. This ing catheters, therapy-induced mucositis, viral increase is linked with progress in medical tech- infections, and graft-versus-host disease, or fol- nology and novel therapeutic options and lowing major abdominal surgery or associated appears to be multifactorial. The widespread use with extensive burns, allows fungi to reach of quinolone prophylaxis in neutropenic cancer normally sterile body sites [5]. In addition, the patients and the availability of broad-spectrum community of vulnerable patients is continuously antibacterial agents has virtually eliminated early expanding as a result of the spread of human immunodeficiency virus (HIV) infections, the Corresponding author and reprint requests: J. A. Maertens, increased use of (novel) immunosuppressive MD, Department of Haematology, University Hospital Gasth- drugs in autoimmune disorders and to prevent uisberg, Herestraat 49, 3000 Leuven, Belgium or treat rejection in the expanding area of trans- Tel: + 32 16 34 68 80 Fax: + 32 16 34 68 81 plant medicine, the popularity of dose-escalated, E-mail: [email protected] often myelo-ablative cytotoxic therapy, the

2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases 2 Clinical Microbiology and Infection, Volume 10 Supplement 1, 2004 improved survival rate in premature infants, and membranes, through inhibition of the fungal the availability of sophisticated life-saving med- cytochrome P450-dependent enzyme lanosterol ical techniques [5–7]. Unfortunately, the attribut- 14-a-demethylase. The resulting depletion of able mortality rate of (systemic) fungal infections and the concomitant accumulation of remains high [8,9]. This may partly be explained 14-a-methylated precursors interferes with the by the difficulty of diagnosing these infections at bulk function of ergosterol in fungal membranes an early stage of their development, because and alters both the fluidity of the membrane and definite proof often requires time-consuming the activity of several membrane-bound enzymes and labour-intensive approaches that cannot al- (e.g. chitin synthase). The net effect is an inhibi- ways be achieved in these severely ill patients. tion of fungal growth and replication. In addition, However, an additional explanation may be a number of secondary effects, such as inhibition found in shortcomings of the current antifungal of the morphogenetic transformation of yeasts to armamentarium. the mycelial form, decreased fungal adherence, Clearly, progress in the development of new and direct toxic effects on membrane phospho- antifungals has lagged behind antibacterial lipids, have been reported [14]. research, a fact that can be explained by at least Unfortunately, as a result of the nonselective two factors. First, before the HIV-era, the occur- nature of the therapeutic target, cross-inhibition rence of fungal infections was believed to be too of P450-dependent enzymes involved in mamma- low to warrant aggressive research by the phar- lian biosynthesis has been responsible for some maceutical industry. Second, the ‘apparent’ lack toxicity, although significantly lower and less of a highly selective fungal target, not present in severe with fluconazole, itraconazole and voric- other eukaryotic (including mammalian) cells, onazole than with the older compounds. The precluded the development of new agents. Until improved toxicity profile of the triazoles com- recently, the arsenal that was available for the pared to the imidazoles (especially endocrine- treatment of systemic fungal infections was lim- related side-effects) can be explained by their ited in number and consisted mainly of the greater affinity for fungal rather than mammalian polyene antibiotic amphotericin B, some azole P450-enzymes at therapeutic concentrations [15]. derivatives, the –thiocarbamates and 5-flucytosine. With the exception of 5-flucytosine, HISTORY OF AZOLES all other agents acted by interfering with the structural or functional integrity of the fungal Although the first report of antifungal activity of an plasma membrane, either by physical disruption azole compound, , was already des- or by blocking the biosynthesis of membrane cribed in 1944 by Woolley, it was not until after the sterols. The past decade, however, has witnessed introduction of topical in 1958 that an expansion of basic and clinical research in researchers became interested in the antifungal antifungal pharmacology and many companies activity of azole compounds [16]. In the late 1960s, have launched new compounds, including sev- three new topical compounds were introduced: eral new azole compounds and the candins [5]. , developed by Bayer Ag (Germany), and and , both developed by Janssen Pharmaceutica (Belgium) [17]. MECHANISM OF ACTION The in-vitro activity of clotrimazole against For a detailed discussion of the mechanism of dermatophytes, yeasts, and dimorphic as well as action, the reader is referred to original work by filamentous fungi, is well-established and com- Vanden Bossche et al. [ 10–12] and a recent review parable to that of amphotericin B for many article by White et al. [13]. Azole antifungals are pathogens [18]. However, unacceptable side- divided into the imidazoles (e.g. miconazole and effects following oral administration [19] and ketoconazole) and the triazoles (e.g. itraconazole, unpredictable pharmacokinetics as a result of fluconazole, voriconazole). The latter group has the induction of hepatic microsomal enzymes [20] three instead of two atoms in the azole have limited the use of clotrimazole to the topical ring. All of the azoles operate via a common mode treatment of dermatophytic infections and super- of action: they prevent the synthesis of ergosterol, ficial candida infections, including oral thrush the major sterol component of fungal plasma and vaginal candidiasis.

2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10 (Suppl. 1), 1–10 Maertens History of azole derivatives 3

Miconazole, a phenethyl synthesised • The use of ketoconazole was associated with in 1969, was the first azole available for parenteral several dose-related (gastrointestinal) side- administration (although not before 1978). Like effects [26]; in addition, ketoconazole could other azoles, it interferes with the biosynthesis of cause symptomatic, even fatal, drug-induced fungal ergosterol, but at high concentrations, hepatitis [32]. miconazole may also cause direct membrane • When given in doses exceeding 400 mg daily, damage that results in leakage of cell constituents. ketoconazole might reversibly inhibit the syn- The drug has a limited spectrum of activity thesis of testosterone and cortisol, resulting in a including dermatophytes, Candida species, dimor- variety of endocrine disturbances, including phic fungi, and Pseudallescheria boydii. The agent rare cases of adrenal insufficiency [33]. has proven to be an effective topical antifungal • A number of clinically important, often unpre- agent, but toxicity associated with the vehicle dictable, drug interactions (e.g. to cyclosporine) used for intravenous administration has limited have been reported [34]. its parenteral use [21], although it has been used Thus, the poor response rates and frequent successfully in the treatment of systemic candida recurrences of major fungal infections, as well as infections, pseudallescheriasis and some refract- the toxicity associated with ketoconazole therapy, ory cases of cryptococcal meningitis [22,23]. led to the search for a second chemical group of Miconazole has recently been withdrawn from azole derivatives, namely the triazoles. In general, the market. the triazoles demonstrate a broader spectrum of In 1981, the Food and Drug Administration antifungal activity and reduced toxicity when (FDA) approved the systemic use of ketoconaz- compared with the imidazole antifungals. Terc- ole, an imidazole derivative synthesised and onazole, the first marketed for human developed by Janssen Pharmaceutica [24]. For use, was active in the topical treatment of vaginal almost a decade it would be regarded as the candidiasis and dermatomycoses. standard and was the only available oral agent (Figure 1a), a broad-spectrum for the treatment of systemic fungal infections. triazole antifungal developed by Pfizer and Until the introduction of the triazoles, ketocon- approved for use in early 1990, covers many of azole was indicated as the drug of choice in the shortcomings of the imidazoles. In contrast to chronic mucocutaneous candidiasis [25] and as ketoconazole, fluconazole is highly water soluble an effective alternative to amphotericin B in less and can be given intravenously to seriously ill severe (nonimmunocompromised) cases of blas- patients. After oral administration, absorption is tomycosis [26], histoplasmosis [26], and paracoc- essentially complete ( 90% bioavailability) and cidioidomycosis [27]; in coccidioidomycosis, the not influenced by gastric pH [35]. In contrast relapse rate after discontinuation of the drug was to ketoconazole, fluconazole enters the cerebro- high [28]. Ketoconazole has not been adequately spinal fluid (CSF) extremely well, with CSF levels evaluated in deep-seated candida infections or of almost 80% of the corresponding serum levels cryptococcosis and was ineffective in aspergil- [36]. The serum half-life allows once-daily dos- losis and mucormycosis. ing, and, also in contrast to ketoconazole, renal Over the years, a number of clinically relevant clearance is the major route of elimination of shortcomings of this compound became evident: fluconazole, with 70–80% of unchanged drug • The absorption of orally administered ketocon- excreted in the urine [37]. Given this favourable azole showed considerable interindividual vari- pharmacokinetic profile (Table 1), fluconazole ation and was markedly influenced by gastric has been studied extensively in various clinical pH [29]. settings, both in prophylaxis and in therapy. • An intravenous formulation was not available. The drug is approved for the treatment of • The drug penetrated the blood–brain barrier oropharyngeal, oesophageal, vaginal, peritoneal poorly and could therefore not be recommen- and genito-urinary candida infections, dissem- ded for the treatment of fungal meningitis inated candidiasis (including chronic dissemin- [30,31]. ated candidiasis) and cryptococcal meningitis. • Ketoconazole was largely fungistatic and Fluconazole also has good activity against coc- proved to be less effective in immunocompro- cidioidomycosis and is a good alternative to keto- mised patients [17]. conazole in chronic mucocutaneous candidiasis.

2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10 (Suppl. 1), 1–10 4 Clinical Microbiology and Infection, Volume 10 Supplement 1, 2004

Fig. 1. Chemical structures of old and new triazoles.

Fluconazole has no clinically meaningful activity ‘non-albicans’ Candida species. In the SCOPE in infections caused by filamentous fungi. In study, 50% of isolates from bloodstream infec- addition, the drug is relatively safe (even at tions were non-albicans species, including C. daily doses up to 1600 mg) and does not inter- tropicalis, C. glabrata, C. krusei, C. parapsilosis and fere with the synthesis of testosterone or cortisol. C. lusitaniae [39]. Some species are considered less Also, fluconazole has fewer drug interactions virulent than C. albicans but are, on the other than does ketoconazole. The clinical experience hand, inherently less susceptible to fluconazole. with fluconazole is reviewed by Dr Pfaller in Recent data, however, have revealed important this issue. differences between patient groups at risk (e.g., The initial enthusiasm for fluconazole, how- oncology versus nononcology), between institu- ever, has been challenged by two recent develop- tions and between countries [9]. Although the ments: the evolving spectrum of fungal pathogens cause of this changing spectrum is multifactorial and the development of azole-resistance. and has not been evaluated systematically, the selective pressure resulting from the widespread use of fluconazole has most probably contributed The evolving spectrum of fungal pathogens to the higher proportion of non-albicans species In virtually every North American and European [40]. Nevertheless, links between spectral changes medical centre, hospital-acquired infections and prior use of fluconazole have not yet been caused by Candida species—both superficial and firmly proved; additional factors such as institu- deep-seated forms—have increased substantially tion-related differences in anti-infective protocols over the past two decades. According to the and treatment-specific factors may be equally National Nosocomial Infection Surveillance Pro- important for fungal colonisation and subsequent gram from the Centers for Disease Control and infection [41]. Prevention (CDC) and the Surveillance and Con- In addition, major observational and autopsy trol of Pathogens of Epidemiological Importance surveys, both in Europe and the USA, have (SCOPE) study, Candida species now account for shown that the incidence of invasive aspergillus 7–8% of all nosocomial bloodstream infections infections—a pathogen not covered by flucona- in the USA [38,39]. Although Candida albicans zole—has increased dramatically over the past remains the most commonly encountered patho- two decades [2,3]. genic yeast, a number of recent studies have Fungal infections have been important compli- demonstrated a shift towards infection with cations among patients with HIV from the begin-

2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10 (Suppl. 1), 1–10 Maertens History of azole derivatives 5

Table 1. Comparative data of avail- able triazole antifungal agents Parameter Fluconazole Itraconazole Voriconazole Formulation oral suspension oral solution tablets tablets capsules intravenous intravenous intravenous Oral bioavailability (%) > 90 55 > 85 Influence of food none › (capsules) fl fl (solution) Influence of › pH none fl (capsules none only) Tmax (h) 1–2 1.5–4.0 < 2 Steady-state plasma 10 1 2 concentration (mg ⁄ L) (200 mg bid) Protein binding (%) < 10 > 95 60 ⁄ Vd (L kg) 0.7–0.8 10.7 2 Principal route of elimination renal hepatic hepatic Elimination half-life 27–37 h 21–64 h 6–9 h Cl (mL ⁄ min.kg) 0.23 3.80 3 Unchanged drug in urine (%)80 <1 <5 Relative CSF levels (%) > 60–80 < 1 > 50 Important drug interactions + + + + + + + + Toxicity Nephrotoxicity – – – Hepatotoxicity + + + Relevant other toxicity – – visual Dose reduction in renal failure yes no ? Dialysable yes no ?

CSF ¼ cerebrospinal fluid; Cl ¼ clearance. ning of the pandemic. Population-based surveil- however, particularly as a result of the liberal lance studies, conducted by the CDC and others, use of fluconazole in immunosuppressed and have clearly reflected the impact of highly active critically ill patients, clear patterns of resistance antiretroviral therapy (HAART) on the incidence have emerged. However, the lack of an estab- and spectrum of these infections in HIV-positive lished definition of resistance remains problem- patients. The incidence of cryptococcal meningitis atic when analysing the scope of this problem. (from 10% to < 3%), mucosal candidiasis, asper- Clearly, the continuous or intermittent adminis- gillosis, and many other fungal infections (histo- tration of any antifungal agent may select for plasmosis, penicilliosis marneffei) has decreased overgrowth of intrinsically resistant or less sus- spectacularly with the use of protease inhibitors ceptible strains or species, as has been docu- [42,43]. mented in oncology patients receiving fluconazole Although Candida and Aspergillus species still prophylaxis. This phenomenon, however, was not represent the vast majority of fungal isolates reported in a very large study of female patients encountered in human pathology, a battery with, or at risk of, HIV infection. Unfortunately, of new species—both yeasts and filamentous resistance has too often been defined as ‘clinically fungi—is increasingly recognized as opportunis- resistant’, referring to a patient whose infection tic pathogens. Of particular concern is the fact that has progressed or persisted despite antifungal many of these so-called ‘emerging’ pathogens are therapy. Classical resistance refers to treatment not covered by fluconazole, including Trichospo- failure in association with high or rising mini- ron spp., Fusarium spp., Scedosporium prolificans, mum inhibitory concentrations (MICs) for the members of the Mucoraceae, and dematiaceous or same fungal strain while receiving therapy: a darkly pigmented fungi [44,45]. definition not used by many publications. Be- sides, key features such as pharmacodynamic parameters, fungal virulence factors, host factors The development of azole-resistance and differences in susceptibility testing methods Until the 1990s, acquired resistance to azole further impair the analysis of a possible link antifungals was uncommon. In recent years, between MIC and outcome [46]. Therefore, the

2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10 (Suppl. 1), 1–10 6 Clinical Microbiology and Infection, Volume 10 Supplement 1, 2004

National Committee for Clinical Laboratory was approved by the Food and Drug Adminis- Standards has recently developed a standardized tration. Compared to ketoconazole, itraconazole and reproducible method for the in vitro suscep- was less toxic and showed a broad spectrum of tibility testing of yeasts to azoles (measuring true activity against Candida spp., Aspergillus spp., microbial resistance) [47]. Cryptococcus neoformans, , His- Apart from this, it is obvious from the literature toplasma capsulatum, Blastomyces dermatitidis, Para- that in-vitro resistance for C. albicans has emerged coccidioides brasiliensis, Sporothrix schenckii and rapidly during the pre-HAART era in HIV ⁄ some phaeohyphomycetes [50]. Since its intro- acquired immune deficiency syndrome (AIDS) duction, itraconazole has gradually replaced patients with oropharyngeal or oesophageal can- ketoconazole as the treatment of choice for non- didiasis [48]. In this setting, up to 20% of C. albicans meningeal, nonlife-threatening cases of histoplas- strains have become resistant to fluconazole, mosis, blastomycosis and paracoccidioidomycosis depending on the duration and total cumulative (amphotericin B for severe or meningeal cases). dose of fluconazole given, the frequency of expo- Itraconazole also has considerable spectral advan- sure and the patient’s CD4+ cell count. Fortu- tages over fluconazole with greater activity nately, at least in developed countries, the in aspergillosis and sporotrichosis [51]. However, introduction of HAART and the subsequent host fluconazole demonstrates a more favourable immune reconstitution has led to a marked reduc- pharmacological and toxicity profile. tion in the number of HIV-associated opportunis- Unlike fluconazole, itraconazole is highly lipid tic fungal infections; as such, azole-resistant soluble and when first introduced, itraconazole isolates from AIDS patients are now seldom was formulated in capsular form only. This encountered, although it remains to be seen formulation became widely used for the treat- whether this improvement will be maintained. In ment of onychomycosis, superficial fungal infec- addition, it is critical to recognize the concept of tions, endemic systemic infections, systemic dose dependency in azole-susceptibility when aspergillus infections, and to a much lesser extent, approaching the therapy of these infections [49]. systemic candida infections. The prophylactic For example, AIDS patients with oropharyngeal efficacy has been demonstrated in a number of candidiasis caused by strains with low MICs will trials in patients with neutropenia or with HIV typically respond to low doses of fluconazole. In infection [52]. However, because absorption of contrast, patients with ‘susceptible, dose-depend- itraconazole capsules can be erratic and low blood ent’ strains may still respond to fluconazole concentrations (< 500 ng ⁄ mL) have been asso- therapy provided that higher doses of 400– ciated with treatment failure, many clinicians 800 mg ⁄ day are given. In general, yeast in-vitro disapprove of its use in patients who were resistance remains manageable in HIV ⁄ AIDS suffering from therapy-induced mucositis or patients. who were receiving antacids [53]. A novel oral It is evident that the same phenomenon of azole formulation of itraconazole, containing the solu- resistance could also arise in deep-seated candida bilizing excipient hydroxypropyl-b-cyclodextrin, infections and candidaemia. However, evidence has therefore been developed (FDA approved in of emerging resistance in this setting remains 1997). When the capsules and oral solution are largely equivocal. Based on a number of large compared, the bioavailability of the solution is surveillance studies in developed countries, there approximately 60% greater than that of the is no evident trend towards an increased acquired capsules. This value was obtained in healthy resistance to fluconazole for bloodstream isolates volunteers [54], as well as in various patient of C. albicans [46]. Although an increased preval- groups, including neutropenic patients [55], HIV ence of C. krusei and C. glabrata bloodstream patients [56], and patients following autologous infections has been seen in oncology patients, and allogeneic transplantation [57]. bone marrow transplant recipients and patients in Recently, an intravenous formulation has been intensive care units, it remains debatable whether developed. In patients with haematological malig- this is (solely) the result of the introduction of nancy, patients with advanced HIV infection, or fluconazole or not. those in intensive care units, high and steady- In 1992, itraconazole (Figure 1b), a broad- state plasma concentrations can be achieved spectrum triazole from Janssen Pharmaceutica, within 2–3 days (as opposed to 1–2 weeks when

2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10 (Suppl. 1), 1–10 Maertens History of azole derivatives 7 using capsules) using intravenous itraconazole, anisms mediate cross-resistance amongst the 400 mg ⁄ day for 2 days followed by 200 mg ⁄ day azoles [62]. for 5 days. Subsequent administration of itracon- Given these shortcomings, the characteristics of azole oral solution or capsules, 400 mg daily, will the ideal azole can easily be deduced: the agent maintain these high plasma levels [58]. Thus, the should be available in oral and intravenous availability of new formulations has introduced dosage forms, demonstrate a broad spectrum of more flexibility in the use of itraconazole. How- activity, covering both yeast as well as classic and ever, when compared with fluconazole, clinical emerging filamentous fungi, be fungicidal, dis- experience remains limited, especially with the play a good pharmacokinetic profile with min- intravenous formulation. imal drug interactions, be stable to resistance and Although the availability over the past decade be cost-effective. Whether the ‘second-generation’ of both fluconazole and itraconazole represents a triazole derivatives that are currently under clin- major advance in the management of systemic ical development or that have recently been fungal infections, these triazole antifungal drugs launched on the market will eliminate or reduce have some important limitations. these shortcomings remains to be seen. Several azole–drug interactions may result in Voriconazole (Figure 1c), structurally related to hazardous and unpredictable toxicity, especially fluconazole, was developed by Pfizer Pharmaceu- in patients receiving chemotherapy (e.g. vincris- ticals as part of a programme designed to enhance tine), transplant recipients (e.g. cyclosporine A, the potency and spectrum of activity of flucona- tacrolimus) and AIDS patients (e.g. indinavir, zole [63]. Voriconazole displays wide-spectrum ritonavir). The potential for drug interactions was in-vitro activity against fungi from all clinically greater with ketoconazole, but similar interactions important pathogenic groups such as Candida have been described with the triazoles. One type spp., Aspergillus spp., C. neoformans, dimorphic of azole–drug interaction may result in decreased fungi, dermatophytes, and some of the emerging plasma concentrations of the azole, either as a mould pathogens including Fusarium spp., Peni- result of decreased absorption or increased meta- cillium, Scedosporium, Acremonium and Trichospo- bolism of the azole. A second type of interaction ron. Members of the zygomycetes still appear to may result in increased toxicity of the coadmin- be resistant. Compared to reference triazoles, istered drug via interference with the cytochrome voriconazole is several-fold more active than P450 systems that are involved in the metabolism fluconazole and itraconazole against Candida of many drugs [59]. spp. However, C. albicans isolates with decreased Their antifungal spectrum remains suboptimal, susceptibility to fluconazole and itraconazole also especially when considering the growing diversity demonstrate significantly higher MICs for voric- of offending species. The activity of fluconazole is onazole, and isolates (Candida as well as Aspergil- limited to dermatophytes, C. neoformans, C. albicans lus) that are highly resistant to both fluconazole and the dimorphic fungi. Although itraconazole and itraconazole show apparent cross-resistance displays a broader spectrum of activity, including to voriconazole. The drug is orally and parenteral- Aspergillus spp. and some yeast strains that are ly active but exhibits complex pharmacokinetics. intrinsically resistant to fluconazole, neither com- Interestingly, animal studies have revealed good pound has documented activity against some of penetration into the CSF and central nervous the emerging pathogens, such as Fusarium spp., system. The promising in-vitro activity has been Scedosporium spp. and the Zygomycetes. confirmed in a range of infections in immuno- Clinical resistance associated with microbiolo- suppressed animal models where voriconazole gical resistance has been reported, both for fluc- proved to be more effective than amphotericin B, onazole (mainly C. albicans) and itraconazole fluconazole and itraconazole. Data from phase II (including Aspergillus spp.) [60]. Meanwhile, sev- and III clinical trials indicate that voriconazole is a eral mechanisms of azole-resistance have been promising agent for the treatment of oropharyn- identified, including enhanced efflux of the azole geal candidiasis in AIDS patients, oesophageal by up-regulation of multidrug efflux pumps, candidiasis, and acute and chronic invasive alterations in the cellular target of azoles (Erg11p), aspergillosis, including cerebral aspergillosis. A and modification of the ERG11 gene at the number of cases have reported activity in unusual molecular level [61]. Of note is that these mech- mould infections, such as scedosporiosis and

2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10 (Suppl. 1), 1–10 8 Clinical Microbiology and Infection, Volume 10 Supplement 1, 2004 fusariosis. However, voriconazole is not yet the treatment of systemic fungal infections, was ideal azole, because the agent is not devoid of the released in the early 1980s. For almost a decade, classical azole–drug interactions and class-related ketoconazole was regarded as the drug of choice side-effects (including severe cases of hepatic in nonlife-threatening endemic mycoses. The dysfunction [64]). Furthermore, dose-related tran- introduction of intravenous and oral fluconazole sient visual disturbances (without morphological in 1990 and oral itraconazole in 1992 represented correlates) have been reported in up to 10% of a second major advance in the treatment of patients receiving this agent. Hence, additional fungal infections. Both first-generation triazoles work regarding (visual) safety, drug–drug inter- displayed a broader spectrum of antifungal actions, metabolism and exposure (given the activity than the imidazoles and had a markedly genetic polymorphism of CYP2C19), and emer- improved safety profile compared with ampho- gence of (cross)-resistance is needed. The current tericin B and ketoconazole. Fluconazole was status of voriconazole is reviewed by Dr Donnelly used frequently for prophylaxis and treatment and Dr De Pauw in this issue. of candidal and cryptococcal infections, especi- Posaconazole (Figure 1d), a hydroxylated ana- ally in the pre-HAART era, whereas itraconazole, logue of itraconazole developed by Schering- despite its erratic absorption, became the drug of Plough Research Institute, possesses potent choice for the treatment of less severe forms of broad-spectrum activity against opportunistic histoplasmosis and blastomycosis, and an attract- and endemic fungal pathogens, including zygo- ive alternative to amphotericin B for the treat- mycetes and some of the dematiaceous moulds. ment of select cases of invasive aspergillosis. In vitro, the drug is highly active against Asperg- Although expanded uses have been suggested illus spp. and at least eightfold more potent than for both agents—in prophylaxis as well as in fluconazole against Candida spp. Similar to voric- therapy—they also became subject to a number onazole, posaconazole was more effective than of clinically important limitations, including amphotericin B, fluconazole and itraconazole in suboptimal spectrum of activity, development animal studies [65]. Ravuconazole (Figure 1e), of resistance, induction of hazardous drug–drug another derivative of fluconazole developed by interactions, less than optimal pharmacokinetic Bristol-Myers Squibb, represents the third second- profile (itraconazole capsules), and toxicity. To generation triazole. Ravuconazole has a broader overcome these limitations, several structural antifungal spectrum than fluconazole and itrac- analogues have been developed and tested in onazole, particularly against strains of C. krusei various stages of clinical development. Three of and C. neoformans. In vitro, ravuconazole is not these so-called ‘second-generation’ triazoles, active against Fusarium spp. and Pseudallescheria including voriconazole, posaconazole and ravuc- boydii [65]. Both posaconazole and ravuconazole onazole, appear to have greater potency and are currently undergoing phase II and III clinical possess increased activity against resistant and trials. emerging pathogens. All three agents are active following oral administration and have demon- strated promising antifungal activity in vitro and CONCLUSION in animal models. These second-generation triaz- Until the 1940s, relatively few agents were oles seem particularly promising for the treat- available for the treatment of systemic fungal ment of Aspergillus infections and for unusual infections. The development of the polyene (but emerging) opportunistic infections that are antifungals, first and later amphotericin otherwise covered only by amphotericin B or B, represented a major advance in medical not covered at all. However, further clinical mycology. Although amphotericin B quickly investigation is warranted to identify the role of became the mainstay of therapy for serious these agents in the future treatment of systemic infections, its use was associated with a number fungal infections. If the toxicity profile of these of infusion-related side-effects and dose-limiting agents is comparable to or better than that of the nephrotoxicity. The continued search for new first-generation triazoles and drug interactions and less toxic antifungals led to the discovery of remain manageable, then these compounds rep- the azoles several decades later. Ketoconazole, resent a true expansion of our antifungal the first available compound for the oral arsenal.

2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10 (Suppl. 1), 1–10 Maertens History of azole derivatives 9

REFERENCES efficacy in systemic fungal infections. Drugs 1980; 19: 7–30. 1. Dixon DM, McNeil MM, Cohen ML, Gellin BG, 22. Bennett JE. Miconazole in cryptococcosis and systemic La Montagne JR. Fungal infections: a growing threat. candidiasis: a word of caution. Ann Intern Med 1981; 94: Public Health Rep 1996; 111: 226–35. 708–9. 2. Groll AH, Shah PM, Menzel C et al. Trends in the post- 23. Lutwick L, Rytel MW, Yanez JP et al. Deep infections mortem epidemiology of invasive fungal infections at a from Petriellidium boydii treated with miconazole. JAm University hospital. J Infect 1996; 33: 23–32. Med Assoc 1979; 241: 271–2. 3. Marr KA, Carter RA, Crippa F, Wald A, Corey L. 24. Heeres J, Backx LJ, Mostmans JH, Van Cutsem J. Epidemiology and outcome of mould infections in Antimycotic imidazoles. Synthesis and antifungal activity hematopoietic stem cell transplant recipients. Clin Infect of ketoconazole, a new potent orally active broad-spec- Dis 2002; 34: 909–17. trum antifungal agent. J Med Chem 1979; 22: 1003–5. 4. Fridkin SK, Jarvis WR. Epidemiology of nosocomial 25. Drouhet E, Dupont B. Chronic mucocutaneous candi- fungal infections. Clin Microbiol Rev 1996; 9: 499–511. dosis and other superficial and systemic mycoses suc- 5. Maertens J, Boogaerts M. Fungal cell wall inhibitors: cessfully treated with ketoconazole. Rev Infect Dis 1980; emphasis on clinical aspects. Curr Pharmaceut Des 2000; 6: 2: 606–19. 225–39. 26. National Institute of Allergy and Infectious Diseases 6. Minamoto GY, Rosenberg AS. Fungal infections in Mycoses Study Group. Treatment of blastomycosis and patients with acquired immunodeficiency syndrome. Med histoplasmosis with ketoconazole. Results of a prospect- Clin North Am 1997; 81: 381–409. ive randomized clinical trial. Ann Intern Med 1985; 103: 7. Paya CV. Fungal infections in solid-organ transplanta- 861–72. tion. Clin Infect Dis 1993; 16: 677–88. 27. Restrepo A, Gomez I, Cano LE et al. Treatment of para- 8. Patterson TF, Kirkpatrick WR, White M et al. Invasive coccidioidomycosis with ketoconazole: a three-year aspergillosis: disease spectrum, treatment practices, and experience. Am J Med 1983; 74 (Suppl. 1B): 48–52. outcomes. I3 Aspergillus Study Group Med (Baltimore) 2000; 28. Galgiani JN, Stevens DA, Graybill JR, Dismukes WE, 79: 250–60. Cloud GA, (NIAID Mycoses Study Group). Ketoconazole 9. Viscoli C, Girmenia C, Marinus A et al. Candidemia in therapy of progressive coccidioidomycosis: comparison cancer patients: a prospective, multi-center surveillance of 400- and 800-mg doses and observations at higher study by the Invasive Fungal Infection Group (IFIG) of doses. Am J Med 1988; 84: 603–10. the European Organization for research and Treatment of 29. Van Der Meer JWM, Keuning JJ, Scheijgrond HW et al. Cancer (EORTC). Clin Infect Dis 1999; 28: 1071–9. The influence of gastric acidity on the bioavailability 10. Vanden Bossche H, Koymans L. Review article: cyto- of ketoconazole (letter). J Antimicrob Chemother 1980; 6: chrome P450 in fungi. Mycoses 1998; 41(Suppl. 1): 32–8. 552–4. 11. Vanden Bossche H, Bellens D, Cools W et al. Cyto- 30. Brass C, Galgiani JN, Blaschke TF et al. Disposition of chrome P-450: target for itraconazole. Drug Dev Res 1986; ketoconazole, an oral antifungal, in humans. Antimicrob 8: 287–98. Agents Chemother 1982; 21: 151–8. 12. Vanden Bossche H, Heeres J, Backx LJ et al. Discovery, 31. Perfect JR, Durack DT, Hamilton JD, Gallis HA. Failure of chemistry, mode of action, and selectivity of itraconazole. ketoconazole in cryptococcal meningitis. JAMA 1982; 247: In: Rippon, JW, Fromtling, RA, eds. Cutaneous Antifungal 3349–51. Agents. New York-Basel-Hong Kong: Marcel Dekker Inc., 32. Lewis JH, Zimmerman HJ, Benson GD, Ishak KG. Hep- 1993; 263–83. atic injury associated with ketoconazole therapy. Analy- 13. White TC, Marr KA, Bowden RA. Clinical, cellular, and sis of 33 cases. Gastroenterology 1984; 86: 503–13. molecular factors that contribute to antifungal drug 33. Pont A, Williams PL, Loose DS et al. Ketoconazole blocks resistance. Clin Microbiol Rev 1998; 11: 382–402. adrenal steroid synthesis. Ann Intern Med 1982; 97: 370–2. 14. Como JA, Dismukes WE. Oral azole drugs as systemic 34. Dismukes WE. Introduction to antifungal drugs. Clin antifungal therapy. N Engl J Med 1994; 330: 263–71. Infect Dis 2000; 30: 653–7. 15. Sheehan DJ, Hitchcock CA, Sibley CM. Current and 35. Brammer KW, Farrow PR, Paulkner JK. Pharmacokinetics emerging azole antifungal agents. Clin Microbiol Rev 1999; and tissue penetration of fluconazole in humans. Rev 12: 40–79. Infect Dis 1990; 12 (Suppl. 3): 318–26. 16. Woolley DW. Some biological effects produced by benz- 36. Arndt CAS, Walsh TJ, McCully CL et al. Fluconazole imidazole and their reversal by purines. J Biol Chem 1944; penetration into cerebrospinal fluid: implications for 152: 225–32. treating fungal infections of the central nervous system. 17. Fromtling R. Overview of medically important antifungal J Infect Dis 1988; 157: 178–80. azole derivatives. Clin Microbiol Rev 1988; 1: 187–217. 37. Humphrey MJ, Jevons S, Tarbit MH. Pharmacokinetic 18. Shadomy S. In vitro antifungal activity of clotrimazole evaluation of UK-49,858, a metabolically stable triazole (Bay b 5097). Infect Immun 1971; 4: 143–8. antifungal drug, in animals and humans. Antimicrob 19. Tettenborn D. Toxicity of clotrimazole. Postgrad Med J Agents Chemother 1985; 28: 648–53. 1974; 50 (Suppl. 1): 17–20. 38. Banerjee SN, Emori TG, Culver DH et al. Secular trends in 20. Burgess MA, Bodey GP. Clotrimazole (Bay b 5097): nosocomial primary bloodstream infections in the United in vitro and clinical pharmacological studies. Antimicrob States, 1980–89. National Nosocomial Infections Surveil- Agents Chemother 1972; 2: 423–6. lance System. Am J Med 1991; 91: 86–9. 21. Heel RC, Brogden RN, Parke GE, Speight TM, Avery GS. 39. Jones RN, Marshall SA, Pfaller MA et al. Nosocomial Miconazole: a preliminary review of its therapeutic enterococcal blood stream infections in the SCOPE

2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10 (Suppl. 1), 1–10 10 Clinical Microbiology and Infection, Volume 10 Supplement 1, 2004

Program: antimicrobial resistance, species occurrence, 51. Terrell CL. Antifungal agents. Part II. The azoles. Mayo molecular testing results, and laboratory testing accur- Clin Proc 1999; 74: 78–100. ancy. Diagn Microbiol Infect Dis 1997; 29: 95. 52. Boogaerts M, Maertens J. Clinical experience with itrac- 40. Abi-Said D, Anaissie E, Uzun O et al. The epidemiology onazole in systemic fungal infections. Drugs 2001; 61 of hematogenous candidiasis caused by different Candida (Suppl. 1): 39–47. species. Clin Infect Dis 1997; 24: 1122–8. 53. Glasmacher A, Hahn C, Leutner C et al. Breakthrough 41. Wingard JR. Importance of Candida species other than invasive fungal infections in neutropenic patients after C. albicans as pathogens in oncology patients. Clin Infect prophylaxis with itraconazole. Mycoses 1999; 42: 443–51. Dis 1995; 20: 115–25. 54. Barone JA, Moskovitz BL, Guarnieri J et al. Enhanced 42. Powderly WG, Landay A, Lederman M et al. Recovery of bioavailability of itraconazole in hydroxypropyl-b-cyclo- the immune system with antiretroviral therapy: the end dextrin solution versus capsules in healthy volunteers. of opportunism? JAMA 1998; 280: 72–7. Antimicrob Agents Chemother 1998; 42: 1862–5. 43. Palella FJ, Delaney KM, Moorman AC et al. Declining 55. Prentice AG, Warnock DW, Johnson SA et al. Multiple morbidity and mortality among patients with advanced dose pharmacokinetics of an oral solution of itraconazole human immunodeficiency virus infection. HIV Out- in patients receiving chemotherapy for acute myeloid patient Study Investigation. N Engl J Med 1998; 338: 853– leukaemia. J Antimicrob Chemother 1995; 36: 657–63. 60. 56. Cartledge JD, Midgely J, Gazzard BG. Itraconazole solu- 44. Vartivarian SE, Anaissie EJ, Bodey GP. Emerging fungal tion: higher serum drug concentrations and better clinical pathogens in immunocompromised patients. Classifica- response rates than capsule formulation in acquired tion, diagnosis and management. Clin Infect Dis 1993; 17 immunodeficiency syndrome patients with candidosis. (Suppl. 2): 487–91. J Clin Pathol 1997; 50: 477–80. 45. Perfect JR, Schell WA. The new fungal opportunists are 57. Michallet M, Persat F, Kranzhofer N et al. Pharmacoki- coming. Clin Infect Dis 1996; 22 (Suppl. 2): 112–18. netics of itraconazole oral solution in allogeneic bone 46a. Balkis MM, Leidich SD, Mukherjee PK, Ghannoum MA. marrow transplant recipients receiving total body irra- Mechanisms of fungal resistance: an overview. Drugs diation. Bone Marrow Transplant 1998; 21: 1239–43. 2002; 62: 1025–40. 58. Boogaerts M, Maertens J, Van Der Geest R et al. Phar- 46. Sanglard D, Odds FC. Resistance of Candida species to macokinetics and safety of a 7-day administration of antifungal agents: molecular and clinical consequences. intravenous itraconazole followed by a 14-day admin- Lancet Infect Dis 2002; 2: 73–85. istration of itraconazole oral solution in patients with 47a. Sobel JD, Ohmit SE, Schuman P et al. The evolution of hematologic malignancy. Antimicrob Agents Chemother Candida species and fluconazole susceptibility among oral 2001; 45: 981–5. and vaginal isolates recovered from human immunode- 59. Albengres E, Louet H, Tillement JP. Drug interactions of ficiency virus seropositive and at-risk HIV-seronegative systemic antifungal agents. Drug Safety 1998; 18: 83–97. women. J Infect Dis 2001; 183: 286–93. 60. Denning DW, Venkateswarlu K, Oakley L et al. Itracon- 47. National Committee for Clinical Laboratory Standards. azole resistance in Aspergillus fumigatus. Antimicrob Agents Reference method for broth dilution antifungal suscepti- Chemother 1997; 41: 1364–8. bility testing of yeasts; approved standards. Wayne, PA: 61. Ghannoum MA, Rice LB. Antifungal agents: mode of National Committee for Clinical Laboratory Standards, action, mechanisms of resistance, and correlation of these 1997. mechanisms with bacterial resistance. Clin Microbiol Rev 48. Rex JH, Rinaldi MG, Pfaller MA. Resistance of Candida 1999; 12: 501–17. species to fluconazole. Antimicrob Agents Chemother 1995; 62. Mosquera J, Denning DW. Azole cross-resistance in 39: 1. Aspergillus fumigatus. Antimicrob Agents Chemother 2002; 49. Rex JH, Pfaller MA, Galgiani JN et al. Development of 46: 556–7. interpretive breakpoints for antifungal susceptibility 63. Sabo JA, Abdel-Rahman SM. Voriconazole: a new triazole testing – conceptual framework and analysis of in vitro antifungal. Ann Pharmacother 2000; 34: 1032–43. in vivo correlation data for fluconazole, itraconazole, and 64. Potoski BA, Brown J. The safety of voriconazole (corres- Candida infections. Clin Infect Dis 1997; 24: 235–47. pondence). Clin Infect Dis 2002; 35: 1373–5. 50. Espinel-Ingroff A, Shadomy S, Gebhart RJ. In vitro studies 65. Chiou CC, Groll AH, Walsh TJ. New drugs and novel with R 51211 (itraconazole). Antimicrob Agents Chemother targets for treatment of invasive fungal infections in 1984; 26: 5–9. patients with cancer. The Oncologist 2000; 5: 120–35.

2004 Copyright by the European Society of Clinical Microbiology and Infectious Diseases, CMI, 10 (Suppl. 1), 1–10